1
|
Sun Z, White Z, Theret M, Bernatchez P. Apolipoprotein E knockout, but not cholesteryl ester transfer protein (CETP)-associated high-density lipoprotein cholesterol (HDL-C) lowering, exacerbates muscle wasting in dysferlin-null mice. Lipids Health Dis 2024; 23:247. [PMID: 39138561 PMCID: PMC11321019 DOI: 10.1186/s12944-024-02227-5] [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: 05/11/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Dysferlin-deficient limb-girdle muscular dystrophy type 2B (Dysf) mice are notorious for their mild phenotype. Raising plasma total cholesterol (CHOL) via apolipoprotein E (ApoE) knockout (KO) drastically exacerbates muscle wasting in Dysf mice. However, dysferlinopathic patients have abnormally reduced plasma high-density lipoprotein cholesterol (HDL-C) levels. The current study aimed to determine whether HDL-C lowering can exacerbate the mild phenotype of dysferlin-null mice. METHODS Human cholesteryl ester transfer protein (CETP), a plasma lipid transfer protein not found in mice that reduces HDL-C, and/or its optimal adapter protein human apolipoprotein B (ApoB), were overexpressed in Dysf mice. Mice received a 2% cholesterol diet from 2 months of age and characterized through ambulatory and hanging functional tests, plasma analyses, and muscle histology. RESULTS CETP/ApoB expression in Dysf mice caused reduced HDL-C (54.5%) and elevated ratio of CHOL/HDL-C (181.3%) compared to control Dysf mice in plasma, but without raising CHOL. Compared to the severe muscle pathology found in high CHOL Dysf/ApoE double knockout mice, Dysf/CETP/ApoB mice did not show significant changes in ambulation, hanging capacity, increases in damaged area, collagen deposition, or decreases in cross-sectional area and healthy myofibre coverage. CONCLUSIONS CETP/ApoB over-expression in Dysf mice decreases HDL-C without increasing CHOL or exacerbating muscle pathology. High CHOL or nonHDL-C caused by ApoE KO, rather than low HDL-C, likely lead to rodent muscular dystrophy phenotype humanization.
Collapse
Affiliation(s)
- Zeren Sun
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Zoe White
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Marine Theret
- School of Biomedical Engineering, Department of Medical Genetics, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada.
| |
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
|
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
|
4
|
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
|
5
|
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
|
6
|
Khattri RB, Batra A, Matheny M, Hart C, Henley-Beasley SC, Hammers D, Zeng H, White Z, Ryan TE, Barton E, Pascal B, Walter GA. Magnetic resonance quantification of skeletal muscle lipid infiltration in a humanized mouse model of Duchenne muscular dystrophy. NMR IN BIOMEDICINE 2023; 36:e4869. [PMID: 36331178 PMCID: PMC10308798 DOI: 10.1002/nbm.4869] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 06/16/2023]
Abstract
Rodent models of Duchenne muscular dystrophy (DMD) often do not recapitulate the severity of muscle wasting and resultant fibro-fatty infiltration observed in DMD patients. Having recently documented severe muscle wasting and fatty deposition in two preclinical models of muscular dystrophy (Dysferlin-null and mdx mice) through apolipoprotein E (ApoE) gene deletion without and with cholesterol-, triglyceride-rich Western diet supplementation, we sought to determine whether magnetic resonance imaging and spectroscopy (MRI and MRS, respectively) could be used to detect, characterize, and compare lipid deposition in mdx-ApoE knockout with mdx mice in a diet-dependent manner. MRI revealed that both mdx and mdx-ApoE mice exhibited elevated proton relaxation time constants (T2 ) in their lower hindlimbs irrespective of diet, indicating both chronic muscle damage and fatty tissue deposition. The mdx-ApoE mice on a Western diet (mdx-ApoEW ) presented with greatest fatty tissue infiltration in the posterior compartment of the hindlimb compared with other groups, as detected by MRI/MRS. High-resolution magic angle spinning confirmed elevated lipid deposition in the posterior compartments of mdx-ApoEW mice in vivo and ex vivo, respectively. In conclusion, the mdx-ApoEW model recapitulates some of the extreme fatty tissue deposition observed clinically in DMD muscle but typically absent in mdx mice. This preclinical model will help facilitate the development of new imaging modalities directly relevant to the image contrast generated in DMD, and help to refine MR-based biomarkers and their relationship to tissue structure and disease progression.
Collapse
Affiliation(s)
- Ram B. Khattri
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Abhinandan Batra
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| | - Michael Matheny
- Department of Pharmacology & Therapeutics, University of Florida, Gainesville, FL, USA
| | - Cora Hart
- Department of Pharmacology & Therapeutics, University of Florida, Gainesville, FL, USA
| | | | - David Hammers
- Department of Pharmacology & Therapeutics, University of Florida, Gainesville, FL, USA
| | - Huadong Zeng
- Advanced Magnetic Resonance Imaging and Spectroscopy Facility, University of Florida, Gainesville, FL, USA
| | - Zoe White
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, Canada
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
- Center of Exercise Science, University of Florida, Gainesville, FL, United States
| | - Elisabeth Barton
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA
| | - Bernatchez Pascal
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, Canada
| | - Glenn A. Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
7
|
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
|
8
|
Sherlock SP, Palmer J, Wagner KR, Abdel-Hamid HZ, Bertini E, Tian C, Mah JK, Kostera-Pruszczyk A, Muntoni F, Guglieri M, Brandsema JF, Mercuri E, Butterfield RJ, McDonald CM, Charnas L, Marraffino S. Quantitative magnetic resonance imaging measures as biomarkers of disease progression in boys with Duchenne muscular dystrophy: a phase 2 trial of domagrozumab. J Neurol 2022; 269:4421-4435. [PMID: 35396602 PMCID: PMC9294028 DOI: 10.1007/s00415-022-11084-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 01/14/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a progressive, neuromuscular disorder caused by mutations in the DMD gene that results in a lack of functional dystrophin protein. Herein, we report the use of quantitative magnetic resonance imaging (MRI) measures as biomarkers in the context of a multicenter phase 2, randomized, placebo-controlled clinical trial evaluating the myostatin inhibitor domagrozumab in ambulatory boys with DMD (n = 120 aged 6 to < 16 years). MRI scans of the thigh to measure muscle volume, muscle volume index (MVI), fat fraction, and T2 relaxation time were obtained at baseline and at weeks 17, 33, 49, and 97 as per protocol. These quantitative MRI measurements appeared to be sensitive and objective biomarkers for evaluating disease progression, with significant changes observed in muscle volume, MVI, and T2 mapping measures over time. To further explore the utility of quantitative MRI measures as biomarkers to inform longer term functional changes in this cohort, a regression analysis was performed and demonstrated that muscle volume, MVI, T2 mapping measures, and fat fraction assessment were significantly correlated with longer term changes in four-stair climb times and North Star Ambulatory Assessment functional scores. Finally, less favorable baseline measures of MVI, fat fraction of the muscle bundle, and fat fraction of lean muscle were significant risk factors for loss of ambulation over a 2-year monitoring period. These analyses suggest that MRI can be a valuable tool for use in clinical trials and may help inform future functional changes in DMD.Trial registration: ClinicalTrials.gov identifier, NCT02310763; registered December 2014.
Collapse
Affiliation(s)
| | | | - Kathryn R Wagner
- Kennedy Krieger Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hoda Z Abdel-Hamid
- Division of Child Neurology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Enrico Bertini
- Unit of Neuromuscular Disease, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Cuixia Tian
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Jean K Mah
- Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Michela Guglieri
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle, UK
| | | | - Eugenio Mercuri
- Pediatric Neurology, Catholic University, Rome, Italy
- Centro Nemo, Fondazione Policlinico Gemelli IRCCS, Rome, Italy
| | | | | | | | | |
Collapse
|
9
|
Hooijmans MT, Habets LE, van den Berg‐Faay SAM, Froeling M, Asselman F, Strijkers GJ, Jeneson JAL, Bartels B, Nederveen AJ, van der Pol WL. Multi-parametric quantitative magnetic resonance imaging of the upper arm muscles of patients with spinal muscular atrophy. NMR IN BIOMEDICINE 2022; 35:e4696. [PMID: 35052014 PMCID: PMC9286498 DOI: 10.1002/nbm.4696] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/24/2021] [Accepted: 01/17/2022] [Indexed: 06/09/2023]
Abstract
Quantitative magnetic resonance imaging (qMRI) is frequently used to map the disease state and disease progression in the lower extremity muscles of patients with spinal muscular atrophy (SMA). This is in stark contrast to the almost complete lack of data on the upper extremity muscles, which are essential for carrying out daily activities. The aim of this study was therefore to assess the disease state in the upper arm muscles of patients with SMA in comparison with healthy controls by quantitative assessment of fat fraction, diffusion indices, and water T2 relaxation times, and to relate these measures to muscle force. We evaluated 13 patients with SMA and 15 healthy controls with a 3-T MRI protocol consisting of DIXON, diffusion tensor imaging, and T2 sequences. qMRI measures were compared between groups and related to muscle force measured with quantitative myometry. Fat fraction was significantly increased in all upper arm muscles of the patients with SMA compared with healthy controls and correlated negatively with muscle force. Additionally, fat fraction was heterogeneously distributed within the triceps brachii (TB) and brachialis muscle, but not in the biceps brachii muscle. Diffusion indices and water T2 relaxation times were similar between patients with SMA and healthy controls, but we did find a slightly reduced mean diffusivity (MD), λ1, and λ3 in the TB of patients with SMA. Furthermore, MD was positively correlated with muscle force in the TB of patients with SMA. The variation in fat fraction further substantiates the selective vulnerability of muscles. The reduced diffusion tensor imaging indices, along with the positive correlation of MD with muscle force, point to myofiber atrophy. Our results show the feasibility of qMRI to map the disease state in the upper arm muscles of patients with SMA. Longitudinal data in a larger cohort are needed to further explore qMRI to map disease progression and to capture the possible effects of therapeutic interventions.
Collapse
Affiliation(s)
- Melissa T. Hooijmans
- Department of Radiology and Nuclear Medicine, Amsterdam Movement SciencesAmsterdam University Medical CenterAmsterdamThe Netherlands
| | - Laura E. Habets
- Center for Child Development, Exercise and Physical Literacy, Wilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Sandra A. M. van den Berg‐Faay
- Department of Radiology and Nuclear Medicine, Amsterdam Movement SciencesAmsterdam University Medical CenterAmsterdamThe Netherlands
| | - Martijn Froeling
- Department of RadiologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Fay‐Lynn Asselman
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Gustav J. Strijkers
- Department of Biomedical Engineering and Physics, Amsterdam Movement SciencesAmsterdam University Medical CenterAmsterdamThe Netherlands
| | - Jeroen A. L. Jeneson
- Center for Child Development, Exercise and Physical Literacy, Wilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Bart Bartels
- Center for Child Development, Exercise and Physical Literacy, Wilhelmina Children's HospitalUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Aart J. Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam Movement SciencesAmsterdam University Medical CenterAmsterdamThe Netherlands
| | - W. Ludo van der Pol
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
10
|
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
|
11
|
Yu HK, Liu X, Pan M, Chen JW, Liu C, Wu Y, Li ZB, Wang HY. Performance of Passive Muscle Stiffness in Diagnosis and Assessment of Disease Progression in Duchenne Muscular Dystrophy. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:414-421. [PMID: 34893358 DOI: 10.1016/j.ultrasmedbio.2021.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/07/2021] [Accepted: 09/02/2021] [Indexed: 06/14/2023]
Abstract
The aim of this study was to evaluate the performance of passive muscle stiffness in diagnosing and assessing disease progression in Duchenne muscular dystrophy (DMD). Boys with DMD and age-matched controls were recruited. Shear wave elastography (SWE) videos were collected by performing dynamic stretching of the gastrocnemius medius (GM). At ankle angles from plantar flexion (PF) 30° to dorsiflexion (DF) 20°, the shear modulus of the GM was measured for each 10° of ankle movement. Shear modulus at each ankle angle was compared between the DMD and control group. Correlation between passive muscle stiffness and motor function grading was also analyzed. A total of 26 patients with DMD and 20 healthy boys were enrolled. At multiple stretch levels, passive muscle stiffness of the GM was significantly higher in patients with DMD than in those in the control group (all p values <0.05). The shear modulus of GM at an ankle angle of DF 10° had the largest area under the receiver operating characteristic curve in differentiating DMD patients from normal subjects (AUC = 0.902, 95% confidence interval: 0.814-0.990). Motor function grading was a significant determinant of passive muscle stiffness at an ankle angle of DF 10° (B = 21.409, t = 3.372, p = 0.003). Passive muscle stiffness may potentially serve as a useful non-invasive tool to monitor disease progression in DMD patients.
Collapse
Affiliation(s)
- Hong-Kui Yu
- Department of Ultrasonography, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiao Liu
- Department of Ultrasonography, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Fu-tian), Shenzhen, Guangdong, China
| | - Min Pan
- Department of Ultrasonography, Shenzhen Hospital of Guangzhou University of Chinese Medicine (Fu-tian), Shenzhen, Guangdong, China
| | - Jin-Wei Chen
- Department of Ultrasonography, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chen Liu
- Department of Ultrasonography, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yu Wu
- Department of Radiology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhi-Bin Li
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Hong-Ying Wang
- Department of Ultrasonography, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
12
|
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
|
13
|
Locher N, Wagner B, Balsiger F, Scheidegger O. Quantitative water T2 relaxometry in the early detection of neuromuscular diseases: a retrospective biopsy-controlled analysis. Eur Radiol 2022; 32:7910-7917. [PMID: 35596779 PMCID: PMC9668929 DOI: 10.1007/s00330-022-08862-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 03/15/2022] [Accepted: 05/01/2022] [Indexed: 01/03/2023]
Abstract
OBJECTIVES To assess quantitative water T2 relaxometry for the early detection of neuromuscular diseases (NMDs) in comparison to standard qualitative MR imaging in a clinical setting. METHODS This retrospective study included 83 patients with suspected NMD who underwent multiparametric MRI at 3 T with a subsequent muscle biopsy between 2015 and 2019. Qualitative T1-weighted and T2-TIRM images were graded by two neuroradiologists to be either pathological or normal. Mean and median water T2 relaxation times (water T2) were obtained from manually drawn volumes of interests in biopsied muscle from multi-echo sequence. Histopathologic pattern of corresponding muscle biopsies was used as a reference. RESULTS In 34 patients, the T1-weighted images showed clear pathological alternations indicating late-stage fatty infiltration in NMDs. In the remaining 49 patients without late-stage changes, T2-TIRM grading achieved a sensitivity of 56.4%, and mean and median water T2 a sensitivity of 87.2% and 97.4% to detect early-stage NMDs. Receiver operating characteristic (ROC) analysis revealed an area under the curve (AUC) of 0.682, 0.715, and 0.803 for T2-TIRM, mean water T2, and median water T2, respectively. Median water T2 ranged between 36 and 42 ms depending on histopathologic pattern. CONCLUSIONS Quantitative water T2 relaxometry had a significantly higher sensitivity in detecting muscle abnormalities than subjective grading of T2-TIRM, prior to late-stage fatty infiltration signal alternations in T1-weighted images. Normal-appearing T2-TIRM does not rule out early-stage NMDs. Our findings suggest considering water T2 relaxometry complementary to T2-TIRM for early detection of NMDs in clinical diagnostic routine. KEY POINTS • Quantitative water T2 relaxometry is more sensitive than subjective assessment of fat-suppressed T2-weighted images for the early detection of neuromuscular diseases, prior to late-stage fatty infiltration signal alternations in T1-weighted images. • Normal-appearing muscles in fat-suppressed T2-weighted images do not rule out early-stage neuromuscular diseases. • Quantitative water T2 relaxometry should be considered complementary to subjectively rated fat-suppressed T2-weighted images in clinical practice.
Collapse
Affiliation(s)
- Noah Locher
- Centre for Neuromuscular Diseases, Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Benedikt Wagner
- Support Center for Advanced Neuroimaging (SCAN), Institute for Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Fabian Balsiger
- Centre for Neuromuscular Diseases, Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Support Center for Advanced Neuroimaging (SCAN), Institute for Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Olivier Scheidegger
- Centre for Neuromuscular Diseases, Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Support Center for Advanced Neuroimaging (SCAN), Institute for Diagnostic and Interventional Neuroradiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Universitätsklinik für Neurologie, Inselspital, Freiburgstrasse, CH-3010, Bern, Switzerland.
| |
Collapse
|
14
|
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
|
15
|
Petković Ramadža D, Kuhtić I, Žarković K, Lochmüller H, Čavka M, Kovač I, Barić I, Prutki M. Case Report: Advanced Skeletal Muscle Imaging in S-Adenosylhomocysteine Hydrolase Deficiency and Further Insight Into Muscle Pathology. Front Pediatr 2022; 10:847445. [PMID: 35463910 PMCID: PMC9026168 DOI: 10.3389/fped.2022.847445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
INTRODUCTION S-Adenosylhomocysteine hydrolase deficiency (SAHHD) is a rare inherited multisystemic disease with muscle involvement as one of the most prominent and poorly understood features. To get better insight into muscle involvement, skeletal muscles were analyzed by magnetic resonance imaging (MRI) and MR spectroscopy (MRS) in three brothers with SAHHD in the different age group. METHOD The study was based on analysis of MRI and MRS of skeletal muscles of the lower and the proximal muscle groups of the upper extremities in three SAHHD patients. RESULTS Three siblings presented in early infancy with similar signs and symptoms, including motor developmental delay. All manifested myopathy, more pronounced in the lower extremities and the proximal skeletal muscle groups, and permanently elevated creatine kinase. At the time of MRI and MRS study, the brothers were at the age of 13, 11, and 8 years, respectively. MRI revealed lipid infiltration, and the MRS curve showed an elevated muscle lipid fraction (higher peak of lipid), which increased with age, and was more prominent in the proximal skeletal muscles of the lower extremities. These results were consistent with muscle biopsy findings in two of them, while the third patient had no specific pathological changes in the examined muscle tissue. CONCLUSIONS These findings demonstrate that an accessible and non-invasive method of MRI and MRS is useful for an insight into the extent of muscle involvement, monitoring disease progression, and response to treatment in SAHHD.
Collapse
Affiliation(s)
- Danijela Petković Ramadža
- Department of Pediatrics, University Hospital Centre Zagreb, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ivana Kuhtić
- Department of Radiology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Kamelija Žarković
- School of Medicine, University of Zagreb, Zagreb, Croatia.,Department of Pathology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Hanns Lochmüller
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada.,Division of Neurology, Department of Medicine, The Ottawa Hospital, Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Mislav Čavka
- School of Medicine, University of Zagreb, Zagreb, Croatia.,Department of Radiology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Ida Kovač
- Department of Rehabilitation and Orthopaedic Devices, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Ivo Barić
- Department of Pediatrics, University Hospital Centre Zagreb, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Maja Prutki
- School of Medicine, University of Zagreb, Zagreb, Croatia.,Department of Radiology, University Hospital Centre Zagreb, Zagreb, Croatia
| |
Collapse
|
16
|
Brogna C, Cristiano L, Verdolotti T, Norcia G, Ficociello L, Ruiz R, Coratti G, Fanelli L, Forcina N, Petracca G, Chieppa F, Tartaglione T, Colosimo C, Pane M, Mercuri E. Longitudinal Motor Functional Outcomes and Magnetic Resonance Imaging Patterns of Muscle Involvement in Upper Limbs in Duchenne Muscular Dystrophy. Medicina (B Aires) 2021; 57:medicina57111267. [PMID: 34833484 PMCID: PMC8624281 DOI: 10.3390/medicina57111267] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 11/25/2022] Open
Abstract
Background and Objectives: The aim of this study was to evaluate longitudinal changes using both upper limb muscle Magnetic Resonance Imaging (MRI) at shoulder, arm and forearm levels and Performance of upper limb (PUL) in ambulant and non-ambulant Duchenne Muscular Dystrophy (DMD) patients. We also wished to define whether baseline muscle MRI could help to predict functional changes after one year. Materials and Methods: Twenty-seven patients had both baseline and 12month muscle MRI and PUL assessments one year later. Results: Ten were ambulant (age range 5–16 years), and 17 non ambulant (age range 10–30 years). Increased abnormalities equal or more than 1.5 point on muscle MRI at follow up were found on all domains: at shoulder level 12/27 patients (44%), at arm level 4/27 (15%) and at forearm level 6/27 (22%). Lower follow up PUL score were found in 8/27 patients (30%) at shoulder level, in 9/27 patients (33%) at mid-level whereas no functional changes were found at distal level. There was no constant association between baseline MRI scores and follow up PUL scores at arm and forearm levels but at shoulder level patients with moderate impairment on the baseline MRI scores between 16 and 34 had the highest risk of decreased function on PUL over a year. Conclusions: Our results confirmed that the integrated use of functional scales and imaging can help to monitor functional and MRI changes over time.
Collapse
Affiliation(s)
- Claudia Brogna
- Pediatric Neurology Unit, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy;
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (G.N.); (G.C.); (L.F.); (N.F.); (M.P.)
| | - Lara Cristiano
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.C.); (R.R.); (G.P.); (F.C.)
| | - Tommaso Verdolotti
- Institute of Radiology, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (T.V.); (L.F.); (C.C.)
| | - Giulia Norcia
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (G.N.); (G.C.); (L.F.); (N.F.); (M.P.)
| | - Luana Ficociello
- Institute of Radiology, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (T.V.); (L.F.); (C.C.)
| | - Roberta Ruiz
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.C.); (R.R.); (G.P.); (F.C.)
| | - Giorgia Coratti
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (G.N.); (G.C.); (L.F.); (N.F.); (M.P.)
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.C.); (R.R.); (G.P.); (F.C.)
| | - Lavinia Fanelli
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (G.N.); (G.C.); (L.F.); (N.F.); (M.P.)
| | - Nicola Forcina
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (G.N.); (G.C.); (L.F.); (N.F.); (M.P.)
| | - Giorgia Petracca
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.C.); (R.R.); (G.P.); (F.C.)
| | - Fabrizia Chieppa
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.C.); (R.R.); (G.P.); (F.C.)
| | - Tommaso Tartaglione
- Department of Radiology, Istituto Dermatologico Italiano, IRCCS, 00167 Rome, Italy;
| | - Cesare Colosimo
- Institute of Radiology, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (T.V.); (L.F.); (C.C.)
- Institute of Radiology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marika Pane
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (G.N.); (G.C.); (L.F.); (N.F.); (M.P.)
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.C.); (R.R.); (G.P.); (F.C.)
| | - Eugenio Mercuri
- Pediatric Neurology Unit, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy;
- Nemo Clinical Centre, Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, 00168 Rome, Italy; (G.N.); (G.C.); (L.F.); (N.F.); (M.P.)
- Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (L.C.); (R.R.); (G.P.); (F.C.)
- Correspondence: ; Tel.: +39-06-30155340; Fax: +39-06-30154363
| |
Collapse
|
17
|
Muzic SI, Paoletti M, Solazzo F, Belatti E, Vitale R, Bergsland N, Bastianello S, Pichiecchio A. Reproducibility of manual segmentation in muscle imaging. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2021; 40:116-123. [PMID: 34632293 PMCID: PMC8489167 DOI: 10.36185/2532-1900-052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/12/2021] [Indexed: 11/23/2022]
Abstract
Purpose To assess the reproducibility of a manual muscle MRI segmentation method that follows a specific set of recommendations developed in our center. Materials and methods Nine healthy volunteers underwent a muscle MRI examination that included a TSE T2 sequence of the thighs. Muscle segmentation was performed by three operators: an expert operator (OP1) with 3 years of experience and two radiology residents (OP2 and 3) who were both given basic segmentation instructions, whereas only OP2 underwent additional supervised training from OP1. Intra- and inter-operator Dice similarity coefficient (DSC) was calculated. Results OP1 showed the highest average intra-operator DSC values (0.885), whereas OP2 had higher average DSC (0.856) compared to OP3 (0.818). The highest inter-operator agreement was observed between Operators 1 and 2 (0.814) and the lowest between OP2 and OP3 (0.702). Confidence interval (CI) analysis showed that the most experienced operator also had the least variability in drawing the ROIs, whereas OP2 showed both higher intra-operator reproducibility compared to OP3 and higher inter-operator agreement with OP1. The muscles that showed the least reproducibility were the semimembranosus and the short head of the biceps femoris. Discussion Following specific recommendations such as these ones derived from our single-center experience leads to an overall high reproducibility of manual muscle segmentation and is helpful in improving both intra-operator and inter-operator reproducibility in less experienced operators.
Collapse
Affiliation(s)
| | - Matteo Paoletti
- Department of Neuroradiology, IRCCS Mondino Foundation, Pavia, Italy
| | | | | | | | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.,Don Carlo Gnocchi ONLUS Foundation IRCCS, Milan, Italy
| | - Stefano Bastianello
- University of Pavia, Pavia, Italy.,Department of Neuroradiology, IRCCS Mondino Foundation, Pavia, Italy
| | - Anna Pichiecchio
- University of Pavia, Pavia, Italy.,Department of Neuroradiology, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
18
|
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
|
19
|
Finkel RS, McDonald CM, Lee Sweeney H, Finanger E, Neil Knierbein E, Wagner KR, Mathews KD, Marks W, Statland J, Nance J, McMillan HJ, McCullagh G, Tian C, Ryan MM, O'Rourke D, Müller-Felber W, Tulinius M, Bryan Burnette W, Nguyen CT, Vijayakumar K, Johannsen J, Phan HC, Eagle M, MacDougall J, Mancini M, Donovan JM. A Randomized, Double-Blind, Placebo-Controlled, Global Phase 3 Study of Edasalonexent in Pediatric Patients with Duchenne Muscular Dystrophy: Results of the PolarisDMD Trial. J Neuromuscul Dis 2021; 8:769-784. [PMID: 34120912 PMCID: PMC8543277 DOI: 10.3233/jnd-210689] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Background: Edasalonexent (CAT-1004) is an orally-administered novel small molecule drug designed to inhibit NF-κB and potentially reduce inflammation and fibrosis to improve muscle function and thereby slow disease progression and muscle decline in Duchenne muscular dystrophy (DMD). Objective: This international, randomized 2 : 1, placebo-controlled, phase 3 study in patients ≥4 – < 8 years old with DMD due to any dystrophin mutation examined the effect of edasalonexent (100 mg/kg/day) compared to placebo over 52 weeks. Methods: Endpoints were changes in the North Star Ambulatory Assessment (NSAA; primary) and timed function tests (TFTs; secondary). Assessment of health-related function used the Pediatric Outcomes Data Collection tool (PODCI). Results: One hundred thirty one patients received edasalonexent (n = 88) and placebo (n = 43). At week 52, differences between edasalonexent and placebo for NSAA total score and TFTs were not statistically significant, although there were consistently less functional declines in the edasalonexent group. A pre-specified analysis by age demonstrated that younger patients (≤6.0 years) showed more robust and statistically significant differences between edasalonexent and placebo for some assessments. Treatment was well-tolerated and the majority of adverse events were mild, and most commonly involved the gastrointestinal system (primarily diarrhea). Conclusions: Edasalonexent was generally well-tolerated with a manageable safety profile at the dose of 100 mg/kg/day. Although edasalonexent did not achieve statistical significance for improvement in primary and secondary functional endpoints for assessment of DMD, subgroup analysis suggested that edasalonexent may slow disease progression if initiated before 6 years of age. (NCT03703882)
Collapse
Affiliation(s)
- Richard S Finkel
- St. Jude Children's Research Hospital, Memphis, TN and Nemours Children's Hospital, Orlando, FL
| | | | - H Lee Sweeney
- University of Florida College of Medicine, Gainesville, FL
| | | | | | - Kathryn R Wagner
- Kennedy Krieger Institute, The Johns Hopkins School of Medicine, Baltimore, MD
| | | | | | | | | | | | | | - Cuixia Tian
- Cincinnati Children's Hospital & University of Cincinnati, Cincinnati, OH
| | | | | | | | - Mar Tulinius
- Queen Silvia Children's Hospital, Gothenburg, Sweden
| | | | | | | | | | - Han C Phan
- Rare Disease Research, LLC, Atlanta GA, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
20
|
Quantitative Muscle MRI in Patients with Neuromuscular Diseases-Association of Muscle Proton Density Fat Fraction with Semi-Quantitative Grading of Fatty Infiltration and Muscle Strength at the Thigh Region. Diagnostics (Basel) 2021; 11:diagnostics11061056. [PMID: 34201303 PMCID: PMC8230029 DOI: 10.3390/diagnostics11061056] [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: 04/16/2021] [Revised: 06/05/2021] [Accepted: 06/06/2021] [Indexed: 11/17/2022] Open
Abstract
(1) Background and Purpose: The skeletal muscles of patients suffering from neuromuscular diseases (NMD) are affected by atrophy, hypertrophy, fatty infiltration, and edematous changes. Magnetic resonance imaging (MRI) is an important tool for diagnosis and monitoring. Concerning fatty infiltration, T1-weighted or T2-weighted DIXON turbo spin echo (TSE) sequences enable a qualitative assessment of muscle involvement. To achieve higher comparability, semi-quantitative grading scales, such as the four-point Mercuri scale, are commonly applied. However, the evaluation remains investigator-dependent. Therefore, effort is being invested to develop quantitative MRI techniques for determination of imaging markers such as the proton density fat fraction (PDFF). The present work aims to assess the diagnostic value of PDFF in correlation to Mercuri grading and clinically determined muscle strength in patients with myotonic dystrophy type 2 (DM2), limb girdle muscular dystrophy type 2A (LGMD2A), and adult Pompe disease. (2) Methods: T2-weighted two-dimensional (2D) DIXON TSE and chemical shift encoding-based water-fat MRI were acquired in 13 patients (DM2: n = 5; LGMD2A: n = 5; Pompe disease: n = 3). Nine different thigh muscles were rated in all patients according to the Mercuri grading and segmented to extract PDFF values. Muscle strength was assessed according to the British Medical Research Council (BMRC) scale. For correlation analyses between Mercuri grading, muscle strength, and PDFF, the Spearman correlation coefficient (rs) was computed. (3) Results: Mean PDFF values ranged from 7% to 37% in adults with Pompe disease and DM2 and up to 79% in LGMD2A patients. In all three groups, a strong correlation of the Mercuri grading and PDFF values was observed for almost all muscles (rs > 0.70, p < 0.05). PDFF values correlated significantly to muscle strength for muscle groups responsible for knee flexion (rs = -0.80, p < 0.01). (4) Conclusion: In the small, investigated patient cohort, PDFF offers similar diagnostic precision as the clinically established Mercuri grading. Based on these preliminary data, PDFF could be further considered as an MRI-based biomarker in the assessment of fatty infiltration of muscle tissue in NMD. Further studies with larger patient cohorts are needed to advance PDFF as an MRI-based biomarker in NMD, with advantages such as its greater dynamic range, enabling the assessment of subtler changes, the amplified objectivity, and the potential of direct correlation to muscle function for selected muscles.
Collapse
|
21
|
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
|
22
|
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
|
23
|
Carraro U, Yablonka-Reuveni Z. Translational research on Myology and Mobility Medicine: 2021 semi-virtual PDM3 from Thermae of Euganean Hills, May 26 - 29, 2021. Eur J Transl Myol 2021; 31:9743. [PMID: 33733717 PMCID: PMC8056169 DOI: 10.4081/ejtm.2021.9743] [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: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 02/08/2023] Open
Abstract
On 19-21 November 2020, the meeting of the 30 years of the Padova Muscle Days was virtually held while the SARS-CoV-2 epidemic was hitting the world after a seemingly quiet summer. During the 2020-2021 winter, the epidemic is still active, despite the start of vaccinations. The organizers hope to hold the 2021 Padua Days on Myology and Mobility Medicine in a semi-virtual form (2021 S-V PDM3) from May 26 to May 29 at the Thermae of Euganean Hills, Padova, Italy. Here the program and the Collection of Abstracts are presented. Despite numerous world problems, the number of submitted/selected presentations (lectures and oral presentations) has increased, prompting the organizers to extend the program to four dense days.
Collapse
Affiliation(s)
- Ugo Carraro
- Department of Biomedical Sciences of the University of Padova, Italy; CIR-Myo - Myology Centre, University of Padova, Italy; A-C Mioni-Carraro Foundation for Translational Myology, Padova.
| | - Zipora Yablonka-Reuveni
- Department of Biological Structure, University of Washington School of Medicine, Seattle, WA.
| |
Collapse
|
24
|
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
|
25
|
Warman-Chardon J, Diaz-Manera J, Tasca G, Straub V. 247th ENMC International Workshop: Muscle magnetic resonance imaging - Implementing muscle MRI as a diagnostic tool for rare genetic myopathy cohorts. Hoofddorp, The Netherlands, September 2019. Neuromuscul Disord 2020; 30:938-947. [PMID: 33004285 DOI: 10.1016/j.nmd.2020.08.360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 08/19/2020] [Indexed: 12/28/2022]
Affiliation(s)
- Jodi Warman-Chardon
- Jodi Warman Chardon, Neurology/Genetics, The Ottawa Hospital/Research Institute, Canada; Children's Hospital of Eastern Ontario/Research Institute, Canada
| | - Jordi Diaz-Manera
- Neuromuscular Disorders Unit, Neurology department, Hospital Universitari de la Santa Creu i Sant Pau, Spain; Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Barcelona, Spain; John Walton Muscular Dystrophy Research Center, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, UK
| | - Giorgio Tasca
- Unità Operativa Complessa di Neurologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Volker Straub
- John Walton Muscular Dystrophy Research Center, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, UK.
| | | |
Collapse
|
26
|
Senesac CR, Barnard AM, Lott DJ, Nair KS, Harrington AT, Willcocks RJ, Zilke KL, Rooney WD, Walter GA, Vandenborne K. Magnetic Resonance Imaging Studies in Duchenne Muscular Dystrophy: Linking Findings to the Physical Therapy Clinic. Phys Ther 2020; 100:2035-2048. [PMID: 32737968 PMCID: PMC7596892 DOI: 10.1093/ptj/pzaa140] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/30/2020] [Indexed: 12/31/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a muscle degenerative disorder that manifests in early childhood and results in progressive muscle weakness. Physical therapists have long been an important component of the multidisciplinary team caring for people with DMD, providing expertise in areas of disease assessment, contracture management, assistive device prescription, and exercise prescription. Over the last decade, magnetic resonance imaging of muscles in people with DMD has led to an improved understanding of the muscle pathology underlying the clinical manifestations of DMD. Findings from magnetic resonance imaging (MRI) studies in DMD, paired with the clinical expertise of physical therapists, can help guide research that leads to improved physical therapist care for this unique patient population. The 2 main goals of this perspective article are to (1) summarize muscle pathology and disease progression findings from qualitative and quantitative muscle MRI studies in DMD and (2) link MRI findings of muscle pathology to the clinical manifestations observed by physical therapists with discussion of any potential implications of MRI findings on physical therapy management.
Collapse
Affiliation(s)
| | | | | | - Kavya S Nair
- Department of Physical Therapy, University of Florida
| | - Ann T Harrington
- Center for Rehabilitation, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania; and Department of Physical Therapy, Arcadia University, Glenside, Pennsylvania
| | | | - Kirsten L Zilke
- Oregon Health & Science University, Shriners Hospitals for Children, Portland, Oregon
| | - William D Rooney
- Advanced Imaging Research Center, Oregon Health & Science University
| | - Glenn A Walter
- Department of Physiology and Functional Genomics, University of Florida
| | | |
Collapse
|
27
|
Dahlqvist JR, Poulsen NS, Østergaard ST, Fornander F, de Stricker Borch J, Danielsen ER, Thomsen C, Vissing J. Evaluation of inflammatory lesions over 2 years in facioscapulohumeral muscular dystrophy. Neurology 2020; 95:e1211-e1221. [PMID: 32611642 DOI: 10.1212/wnl.0000000000010155] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 03/05/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE We followed up patients with facioscapulohumeral muscular dystrophy (FSHD) with sequential examinations over 2 years to investigate whether inflammatory lesions always precede fat replacement, if inflammation can be resolved without muscle degeneration, and if inflammatory lesions in muscle are always followed by fat replacement. METHODS In this longitudinal study of 10 sequential MRI assessments over 2.5 years, we included 10 patients with FSHD. We used MRI with short TI inversion recovery to identify regions of interest (ROIs) with hyperintensities indicating muscle inflammation. Muscle T2 relaxation time mapping was used as a quantitative marker of muscle inflammation. Dixon sequences quantified muscle fat replacement. Ten healthy controls were examined with a magnetic resonance scan once for determination of normal values of T2 relaxation time. RESULTS We identified 68 ROIs with T2 elevation in the patients with FSHD. New ROIs with T2 elevation arising during the study had muscle fat content of 6.4% to 33.0% (n = 8) and 47.0% to 78.0% lesions that resolved (n = 6). ROIs with T2 elevation had a higher increase in muscle fat content from visits 1 to 10 (7.9 ± 7.9%) compared to ROIs with normal muscle T2 relaxation times (1.7 ± 2.6%; p < 0.0001). Severe T2 elevations were always followed by an accelerated replacement of muscle by fat. CONCLUSIONS Our results suggest that muscle inflammation starts in mildly affected muscles in FSHD, is related to a faster muscle degradation, and continues until the muscles are completely fat replaced. CLINICALTRIALSGOV IDENTIFIER NCT02159612.
Collapse
Affiliation(s)
- Julia R Dahlqvist
- From the Copenhagen Neuromuscular Center (J.R.D., N.S.P., S.T.Ø, F.F., J.d.S.B., J.V.), Section 3342, Department of Neurology, and Department of Radiology (C.T.), Rigshospitalet, Copenhagen University; and Department of Radiology (E.R.D., C.T.), Zealand University Hospital, Roskilde, Denmark.
| | - Nanna S Poulsen
- From the Copenhagen Neuromuscular Center (J.R.D., N.S.P., S.T.Ø, F.F., J.d.S.B., J.V.), Section 3342, Department of Neurology, and Department of Radiology (C.T.), Rigshospitalet, Copenhagen University; and Department of Radiology (E.R.D., C.T.), Zealand University Hospital, Roskilde, Denmark
| | - Sofie T Østergaard
- From the Copenhagen Neuromuscular Center (J.R.D., N.S.P., S.T.Ø, F.F., J.d.S.B., J.V.), Section 3342, Department of Neurology, and Department of Radiology (C.T.), Rigshospitalet, Copenhagen University; and Department of Radiology (E.R.D., C.T.), Zealand University Hospital, Roskilde, Denmark
| | - Freja Fornander
- From the Copenhagen Neuromuscular Center (J.R.D., N.S.P., S.T.Ø, F.F., J.d.S.B., J.V.), Section 3342, Department of Neurology, and Department of Radiology (C.T.), Rigshospitalet, Copenhagen University; and Department of Radiology (E.R.D., C.T.), Zealand University Hospital, Roskilde, Denmark
| | - Josefine de Stricker Borch
- From the Copenhagen Neuromuscular Center (J.R.D., N.S.P., S.T.Ø, F.F., J.d.S.B., J.V.), Section 3342, Department of Neurology, and Department of Radiology (C.T.), Rigshospitalet, Copenhagen University; and Department of Radiology (E.R.D., C.T.), Zealand University Hospital, Roskilde, Denmark
| | - Else R Danielsen
- From the Copenhagen Neuromuscular Center (J.R.D., N.S.P., S.T.Ø, F.F., J.d.S.B., J.V.), Section 3342, Department of Neurology, and Department of Radiology (C.T.), Rigshospitalet, Copenhagen University; and Department of Radiology (E.R.D., C.T.), Zealand University Hospital, Roskilde, Denmark
| | - Carsten Thomsen
- From the Copenhagen Neuromuscular Center (J.R.D., N.S.P., S.T.Ø, F.F., J.d.S.B., J.V.), Section 3342, Department of Neurology, and Department of Radiology (C.T.), Rigshospitalet, Copenhagen University; and Department of Radiology (E.R.D., C.T.), Zealand University Hospital, Roskilde, Denmark
| | - John Vissing
- From the Copenhagen Neuromuscular Center (J.R.D., N.S.P., S.T.Ø, F.F., J.d.S.B., J.V.), Section 3342, Department of Neurology, and Department of Radiology (C.T.), Rigshospitalet, Copenhagen University; and Department of Radiology (E.R.D., C.T.), Zealand University Hospital, Roskilde, Denmark
| |
Collapse
|
28
|
Dieckmeyer M, Zoffl F, Grundl L, Inhuber S, Schlaeger S, Burian E, Zimmer C, Kirschke JS, Karampinos DC, Baum T, Sollmann N. Association of quadriceps muscle, gluteal muscle, and femoral bone marrow composition using chemical shift encoding-based water-fat MRI: a preliminary study in healthy young volunteers. Eur Radiol Exp 2020; 4:35. [PMID: 32518982 PMCID: PMC7283400 DOI: 10.1186/s41747-020-00162-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/17/2020] [Indexed: 12/04/2022] Open
Abstract
Background We investigated the composition of the gluteal (gluteus maximus, medius, and minimus) and quadriceps (rectus femoris, vastus lateralis, medialis, and intermedius) muscle groups and its associations with femoral bone marrow using chemical shift encoding-based water-fat magnetic resonance imaging (CSE-MRI) to improve our understanding of muscle-bone interaction. Methods Thirty healthy volunteers (15 males, aged 30.5 ± 4.9 years [mean ± standard deviation]; 15 females, aged 29.9 ± 7.1 years) were recruited. A six-echo three-dimensional spoiled gradient-echo sequence was used for 3-T CSE-MRI at the thigh and hip region. The proton density fat fraction (PDFF) of the gluteal and quadriceps muscle groups as well as of the femoral head, neck, and greater trochanter bone marrow were extracted and averaged over both sides. Results PDFF values of all analysed bone marrow compartments were significantly higher in men than in women (p ≤ 0.047). PDFF values of the analysed muscles showed no significant difference between men and women (p ≥ 0.707). After adjusting for age and body mass index, moderate significant correlations of PDFF values were observed between the gluteal and quadriceps muscle groups (r = 0.670) and between femoral subregions (from r = 0.613 to r = 0.655). Regarding muscle-bone interactions, only the PDFF of the quadriceps muscle and greater trochanter bone marrow showed a significant correlation (r = 0.375). Conclusions The composition of the muscle and bone marrow compartments at the thigh and hip region in young, healthy subjects seems to be quite distinct, without evidence for a strong muscle-bone interaction.
Collapse
Affiliation(s)
- Michael Dieckmeyer
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Florian Zoffl
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Lioba Grundl
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Stephanie Inhuber
- Department of Sport and Health Sciences, Technische Universität München, Georg-Brauchle-Ring 60/62, 80992 Munich, Germany
| | - Sarah Schlaeger
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.,Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | - Egon Burian
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Claus Zimmer
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Jan S Kirschke
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany.,TUM-Neuroimaging Center, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Dimitrios C Karampinos
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | - Thomas Baum
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany
| | - Nico Sollmann
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675, Munich, Germany. .,TUM-Neuroimaging Center, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
| |
Collapse
|
29
|
Forbes SC, Arora H, Willcocks RJ, Triplett WT, Rooney WD, Barnard AM, Alabasi U, Wang DJ, Lott DJ, Senesac CR, Harrington AT, Finanger EL, Tennekoon GI, Brandsema J, Daniels MJ, Sweeney HL, Walter GA, Vandenborne K. Upper and Lower Extremities in Duchenne Muscular Dystrophy Evaluated with Quantitative MRI and Proton MR Spectroscopy in a Multicenter Cohort. Radiology 2020; 295:616-625. [PMID: 32286193 PMCID: PMC7263287 DOI: 10.1148/radiol.2020192210] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 02/05/2020] [Accepted: 02/13/2020] [Indexed: 12/18/2022]
Abstract
Background Upper extremity MRI and proton MR spectroscopy are increasingly considered to be outcome measures in Duchenne muscular dystrophy (DMD) clinical trials. Purpose To demonstrate the feasibility of acquiring upper extremity MRI and proton (1H) MR spectroscopy measures of T2 and fat fraction in a large, multicenter cohort (ImagingDMD) of ambulatory and nonambulatory individuals with DMD; compare upper and lower extremity muscles by using MRI and 1H MR spectroscopy; and correlate upper extremity MRI and 1H MR spectroscopy measures to function. Materials and Methods In this prospective cross-sectional study, MRI and 1H MR spectroscopy and functional assessment data were acquired from participants with DMD and unaffected control participants at three centers (from January 28, 2016, to April 24, 2018). T2 maps of the shoulder, upper arm, forearm, thigh, and calf were generated from a spin-echo sequence (repetition time msec/echo time msec, 3000/20-320). Fat fraction maps were generated from chemical shift-encoded imaging (eight echo times). Fat fraction and 1H2O T2 in the deltoid and biceps brachii were measured from single-voxel 1H MR spectroscopy (9000/11-243). Groups were compared by using Mann-Whitney test, and relationships between MRI and 1H MR spectroscopy and arm function were assessed by using Spearman correlation. Results This study evaluated 119 male participants with DMD (mean age, 12 years ± 3 [standard deviation]) and 38 unaffected male control participants (mean age, 12 years ± 3). Deltoid and biceps brachii muscles were different in participants with DMD versus control participants in all age groups by using quantitative T2 MRI (P < .001) and 1H MR spectroscopy fat fraction (P < .05). The deltoid, biceps brachii, and triceps brachii were affected to the same extent (P > .05) as the soleus and medial gastrocnemius. Negative correlations were observed between arm function and MRI (T2: range among muscles, ρ = -0.53 to -0.73 [P < .01]; fat fraction, ρ = -0.49 to -0.70 [P < .01]) and 1H MR spectroscopy fat fraction (ρ = -0.64 to -0.71; P < .01). Conclusion This multicenter study demonstrated early and progressive involvement of upper extremity muscles in Duchenne muscular dystrophy (DMD) and showed the feasibility of MRI and 1H MR spectroscopy to track disease progression over a wide range of ages in participants with DMD. © RSNA, 2020 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Sean C. Forbes
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Harneet Arora
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Rebecca J. Willcocks
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - William T. Triplett
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - William D. Rooney
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Alison M. Barnard
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Umar Alabasi
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Dah-Jyuu Wang
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Donovan J. Lott
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Claudia R. Senesac
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Ann T. Harrington
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Erika L. Finanger
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Gihan I. Tennekoon
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - John Brandsema
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Michael J. Daniels
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - H. Lee Sweeney
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Glenn A. Walter
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Krista Vandenborne
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| |
Collapse
|
30
|
Judge SM, Deyhle MR, Neyroud D, Nosacka RL, D'Lugos AC, Cameron ME, Vohra RS, Smuder AJ, Roberts BM, Callaway CS, Underwood PW, Chrzanowski SM, Batra A, Murphy ME, Heaven JD, Walter GA, Trevino JG, Judge AR. MEF2c-Dependent Downregulation of Myocilin Mediates Cancer-Induced Muscle Wasting and Associates with Cachexia in Patients with Cancer. Cancer Res 2020; 80:1861-1874. [PMID: 32132110 DOI: 10.1158/0008-5472.can-19-1558] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 11/27/2019] [Accepted: 02/24/2020] [Indexed: 12/27/2022]
Abstract
Skeletal muscle wasting is a devastating consequence of cancer that contributes to increased complications and poor survival, but is not well understood at the molecular level. Herein, we investigated the role of Myocilin (Myoc), a skeletal muscle hypertrophy-promoting protein that we showed is downregulated in multiple mouse models of cancer cachexia. Loss of Myoc alone was sufficient to induce phenotypes identified in mouse models of cancer cachexia, including muscle fiber atrophy, sarcolemmal fragility, and impaired muscle regeneration. By 18 months of age, mice deficient in Myoc showed significant skeletal muscle remodeling, characterized by increased fat and collagen deposition compared with wild-type mice, thus also supporting Myoc as a regulator of muscle quality. In cancer cachexia models, maintaining skeletal muscle expression of Myoc significantly attenuated muscle loss, while mice lacking Myoc showed enhanced muscle wasting. Furthermore, we identified the myocyte enhancer factor 2 C (MEF2C) transcription factor as a key upstream activator of Myoc whose gain of function significantly deterred cancer-induced muscle wasting and dysfunction in a preclinical model of pancreatic ductal adenocarcinoma (PDAC). Finally, compared with noncancer control patients, MYOC was significantly reduced in skeletal muscle of patients with PDAC defined as cachectic and correlated with MEF2c. These data therefore identify disruptions in MEF2c-dependent transcription of Myoc as a novel mechanism of cancer-associated muscle wasting that is similarly disrupted in muscle of patients with cachectic cancer. SIGNIFICANCE: This work identifies a novel transcriptional mechanism that mediates skeletal muscle wasting in murine models of cancer cachexia that is disrupted in skeletal muscle of patients with cancer exhibiting cachexia.
Collapse
Affiliation(s)
- Sarah M Judge
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, Florida.
| | - Michael R Deyhle
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, Florida
| | - Daria Neyroud
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, Florida
| | - Rachel L Nosacka
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, Florida
| | - Andrew C D'Lugos
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, Florida
| | - Miles E Cameron
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, Florida.,Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Ravneet S Vohra
- Department of Physiology, College of Medicine, University of Florida Health Science Center, Gainesville, Florida
| | - Ashley J Smuder
- Department of Health and Human Performance, University of Florida, Gainesville, Florida
| | - Brandon M Roberts
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, Florida
| | - Chandler S Callaway
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, Florida
| | - Patrick W Underwood
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Stephen M Chrzanowski
- Department of Physiology, College of Medicine, University of Florida Health Science Center, Gainesville, Florida
| | - Abhinandan Batra
- Department of Physiology, College of Medicine, University of Florida Health Science Center, Gainesville, Florida
| | - Meghan E Murphy
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, Florida
| | - Jonathan D Heaven
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, Florida
| | - Glenn A Walter
- Department of Physiology, College of Medicine, University of Florida Health Science Center, Gainesville, Florida
| | - Jose G Trevino
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Andrew R Judge
- Department of Physical Therapy, University of Florida Health Science Center, Gainesville, Florida.
| |
Collapse
|
31
|
Barnard AM, Willcocks RJ, Triplett WT, Forbes SC, Daniels MJ, Chakraborty S, Lott DJ, Senesac CR, Finanger EL, Harrington AT, Tennekoon G, Arora H, Wang DJ, Sweeney HL, Rooney WD, Walter GA, Vandenborne K. MR biomarkers predict clinical function in Duchenne muscular dystrophy. Neurology 2020; 94:e897-e909. [PMID: 32024675 PMCID: PMC7238941 DOI: 10.1212/wnl.0000000000009012] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 08/29/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE To investigate the potential of lower extremity magnetic resonance (MR) biomarkers to serve as endpoints in clinical trials of therapeutics for Duchenne muscular dystrophy (DMD) by characterizing the longitudinal progression of MR biomarkers over 48 months and assessing their relationship to changes in ambulatory clinical function. METHODS One hundred sixty participants with DMD were enrolled in this longitudinal, natural history study and underwent MR data acquisition of the lower extremity muscles to determine muscle fat fraction (FF) and MRI T2 biomarkers of disease progression. In addition, 4 tests of ambulatory function were performed. Participants returned for follow-up data collection at 12, 24, 36, and 48 months. RESULTS Longitudinal analysis of the MR biomarkers revealed that vastus lateralis FF, vastus lateralis MRI T2, and biceps femoris long head MRI T2 biomarkers were the fastest progressing biomarkers over time in this primarily ambulatory cohort. Biomarker values tended to demonstrate a nonlinear, sigmoidal trajectory over time. The lower extremity biomarkers predicted functional performance 12 and 24 months later, and the magnitude of change in an MR biomarker over time was related to the magnitude of change in function. Vastus lateralis FF, soleus FF, vastus lateralis MRI T2, and biceps femoris long head MRI T2 were the strongest predictors of future loss of function, including loss of ambulation. CONCLUSIONS This study supports the strong relationship between lower extremity MR biomarkers and measures of clinical function, as well as the ability of MR biomarkers, particularly those from proximal muscles, to predict future ambulatory function and important clinical milestones. CLINICALTRIALSGOV IDENTIFIER NCT01484678.
Collapse
Affiliation(s)
- Alison M Barnard
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Rebecca J Willcocks
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - William T Triplett
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Sean C Forbes
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Michael J Daniels
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Saptarshi Chakraborty
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Donovan J Lott
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Claudia R Senesac
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Erika L Finanger
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Ann T Harrington
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Gihan Tennekoon
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Harneet Arora
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Dah-Jyuu Wang
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - H Lee Sweeney
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - William D Rooney
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Glenn A Walter
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Krista Vandenborne
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA.
| |
Collapse
|
32
|
Koolstra K, Webb AG, Veeger TTJ, Kan HE, Koken P, Börnert P. Water-fat separation in spiral magnetic resonance fingerprinting for high temporal resolution tissue relaxation time quantification in muscle. Magn Reson Med 2020; 84:646-662. [PMID: 31898834 PMCID: PMC7217066 DOI: 10.1002/mrm.28143] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 12/16/2022]
Abstract
Purpose To minimize the known biases introduced by fat in rapid T1 and T2 quantification in muscle using a single‐run magnetic resonance fingerprinting (MRF) water–fat separation sequence. Methods The single‐run MRF acquisition uses an alternating in‐phase/out‐of‐phase TE pattern to achieve water–fat separation based on a 2‐point DIXON method. Conjugate phase reconstruction and fat deblurring were applied to correct for B0 inhomogeneities and chemical shift blurring. Water and fat signals were matched to the on‐resonance MRF dictionary. The method was first tested in a multicompartment phantom. To test whether the approach is capable of measuring small in vivo dynamic changes in relaxation times, experiments were run in 9 healthy volunteers; parameter values were compared with and without water–fat separation during muscle recovery after plantar flexion exercise. Results Phantom results show the robustness of the water–fat resolving MRF approach to undersampling. Parameter maps in volunteers show a significant (P < .01) increase in T1 (105 ± 94 ms) and decrease in T2 (14 ± 6 ms) when using water–fat‐separated MRF, suggesting improved parameter quantification by reducing the well‐known biases introduced by fat. Exercise results showed smooth T1 and T2 recovery curves. Conclusion Water–fat separation using conjugate phase reconstruction is possible within a single‐run MRF scan. This technique can be used to rapidly map relaxation times in studies requiring dynamic scanning, in which the presence of fat is problematic.
Collapse
Affiliation(s)
- Kirsten Koolstra
- C.J. Gorter Center for High Field MRI, Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Andrew G Webb
- C.J. Gorter Center for High Field MRI, Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Thom T J Veeger
- C.J. Gorter Center for High Field MRI, Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Hermien E Kan
- C.J. Gorter Center for High Field MRI, Radiology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Peter Börnert
- C.J. Gorter Center for High Field MRI, Radiology, Leiden University Medical Center, Leiden, Netherlands.,Philips Research, Hamburg, Germany
| |
Collapse
|
33
|
Finanger E, Vandenborne K, Finkel RS, Lee Sweeney H, Tennekoon G, Yum S, Mancini M, Bista P, Nichols A, Liu H, Fretzen A, Donovan JM. Phase 1 Study of Edasalonexent (CAT-1004), an Oral NF-κB Inhibitor, in Pediatric Patients with Duchenne Muscular Dystrophy. J Neuromuscul Dis 2020; 6:43-54. [PMID: 30452422 PMCID: PMC6398836 DOI: 10.3233/jnd-180341] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background: Edasalonexent is an orally administered small molecule designed to inhibit NF-κB, which is activated from infancy in Duchenne muscular dystrophy and is central to causing muscle damage and preventing muscle regeneration. Objective: Evaluate the safety, tolerability, pharmacokinetics and exploratory pharmacodynamics of three doses of edasalonexent in ambulatory males ≥4 to <8 years of age with genetically confirmed Duchenne muscular dystrophy. Methods: This was a 1-week, open-label, multiple-dose study with 3 sequential ascending doses (33, 67 and 100 mg/kg/day) of edasalonexent administered under different dietary conditions to 17 males with a mean age of 5.5 years. Results: All doses of edasalonexent were well tolerated, with no serious adverse events, no drug discontinuations and no dose reductions. The majority of adverse events were mild, and the most common adverse events were gastrointestinal (primarily diarrhea). Edasalonexent was rapidly absorbed with peak levels observed 2–6 hours after dosing and exposures appeared to increase nearly proportionally to dose for the 2 lower and all 3 doses under low-fat and high-fat meal conditions, respectively. Only minor plasma accumulation of edasalonexent was observed with 7 days of dosing. After treatment with edasalonexent for 7 days, levels of NF-κB-regulated genes and serum proteins were decreased. Conclusions: This first report of edasalonexent oral administration for one week in male pediatric patients with Duchenne muscular dystrophy showed that treatment was well tolerated and inhibited NF-kB pathways.
Collapse
Affiliation(s)
- Erika Finanger
- Oregon Health Sciences University Pediatrics, Portland, OR, USA
| | | | - Richard S Finkel
- Nemours Children's Hospital, Division of Pediatric Neurology, Orlando, FL, USA
| | - H Lee Sweeney
- University of Florida Health Myology Institute, Gainesville, FL, USA
| | - Gihan Tennekoon
- Children's Hospital of Philadelphia Pediatric Neurology, Philadelphia, PA, USA
| | - Sabrina Yum
- Children's Hospital of Philadelphia Pediatric Neurology, Philadelphia, PA, USA
| | | | | | | | - Hanlan Liu
- Catabasis Pharmaceuticals, Inc., Cambridge, MA, USA
| | | | | |
Collapse
|
34
|
Chrzanowski SM, Darras BT, Rutkove SB. The Value of Imaging and Composition-Based Biomarkers in Duchenne Muscular Dystrophy Clinical Trials. Neurotherapeutics 2020; 17:142-152. [PMID: 31879850 PMCID: PMC7007477 DOI: 10.1007/s13311-019-00825-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
As the drug development pipeline for Duchenne muscular dystrophy (DMD) rapidly advances, clinical trial outcomes need to be optimized. Effective assessment of disease burden, natural history progression, and response to therapy in clinical trials for Duchenne muscular dystrophy are critical factors for clinical trial success. By choosing optimal biomarkers to better assess therapeutic efficacy, study costs and sample size requirements can be reduced. Currently, functional measures continue to serve as the primary outcome for the majority of DMD clinical trials. Quantitative measures of muscle health, including magnetic resonance imaging and spectroscopy, electrical impedance myography, and ultrasound, sensitively identify diseased muscle, disease progression, and response to a therapeutic intervention. Furthermore, such non-invasive techniques have the potential to identify disease pathology prior to onset of clinical symptoms. Despite robust supportive evidence, non-invasive quantitative techniques are still not frequently utilized in clinical trials for Duchenne muscular dystrophy. Non-invasive quantitative techniques have demonstrated the ability to quantify disease progression and potential response to therapeutic intervention, and should be used as a supplement to current standard functional measures. Such methods have the potential to significantly accelerate the development and approval of therapies for DMD.
Collapse
Affiliation(s)
- Stephen M Chrzanowski
- Department of Medicine, Boston Children's Hospital, 300 Longwood Ave., Boston, MA, 02115, USA.
| | - Basil T Darras
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Seward B Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
35
|
Marty B, Carlier PG. MR fingerprinting for water T1 and fat fraction quantification in fat infiltrated skeletal muscles. Magn Reson Med 2019; 83:621-634. [PMID: 31502715 DOI: 10.1002/mrm.27960] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 07/11/2019] [Accepted: 07/31/2019] [Indexed: 12/28/2022]
Abstract
PURPOSE To develop a fast MR fingerprinting (MRF) sequence for simultaneous estimation of water T1 (T1H2O ) and fat fraction (FF) in fat infiltrated skeletal muscles. METHODS The MRF sequence for T1H2O and FF quantification (MRF T1-FF) comprises a 1400 radial spokes echo train, following nonselective inversion, with varying echo and repetition time, as well as prescribed flip angle. Undersampled frames were reconstructed at different acquisition time-points by nonuniform Fourier transform, and a bi-component model based on Bloch simulations applied to adjust the signal evolution and extract T1H2O and FF. The sequence was validated on a multi-vial phantom, in three healthy volunteers and five patients with neuromuscular diseases. We evaluated the agreement between MRF T1-FF parameters and reference values and confounding effects due to B0 and B1 inhomogeneities. RESULTS In phantom, T1H2O and FF were highly correlated with references values measured with multi-inversion time inversion recovery-stimulated echo acquisition mode and Dixon, respectively (R2 > 0.99). In vivo, T1H2O and FF determined by the MRF T1-FF sequence were also correlated with reference values (R2 = 0.98 and 0.97, respectively). The precision on T1H2O was better than 5% for muscles where FF was less than 0.4. Both T1H2O and FF values were not confounded by B0 nor B1 inhomogeneities. CONCLUSION The MRF T1-FF sequence derived T1H2O and FF values in voxels containing a mixture of water and fat protons. This method can be used to comprehend and characterize the effects of tissue water compartmentation and distribution on muscle T1 values in patients affected by chronic fat infiltration.
Collapse
Affiliation(s)
- Benjamin Marty
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA, DRF, IBFJ, MIRCen, Paris, France
| | - Pierre G Carlier
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA, DRF, IBFJ, MIRCen, Paris, France
| |
Collapse
|
36
|
Muntoni F, Domingos J, Manzur AY, Mayhew A, Guglieri M, Sajeev G, Signorovitch J, Ward SJ. Categorising trajectories and individual item changes of the North Star Ambulatory Assessment in patients with Duchenne muscular dystrophy. PLoS One 2019; 14:e0221097. [PMID: 31479456 PMCID: PMC6719875 DOI: 10.1371/journal.pone.0221097] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 07/30/2019] [Indexed: 01/16/2023] Open
Abstract
Functional variability among boys with Duchenne muscular dystrophy (DMD) is well recognised and complicates interpretation of clinical studies. We hypothesised that boys with DMD could be clustered into groups sharing similar trajectories of ambulatory function over time, as measured by the North Star Ambulatory Assessment (NSAA) total score. We also explored associations with other variables such as age, functional abilities, and genotype. Using the NorthStar Clinical Network database, 395 patients with >1 NSAA assessment were identified. We utilised latent class trajectory analysis of longitudinal NSAA scores, which produced evidence for at least four clusters of boys sharing similar trajectories versus age in decreasing order of clinical severity: 25% of the boys were in cluster 1 (NSAA falling to ≤ 5 at age ~10y), 35% were in cluster 2 (NSAA ≤ 5 ~12y), 21% in were cluster 3 (NSAA≤ 5 ~14y), and 19% in cluster 4 (NSAA > 5 up to 15y). Mean ages at diagnosis of DMD were similar across clusters (4.2, 3.9, 4.3, and 4.8y, respectively). However, at the first NSAA assessment, a significant (p<0.05) association was observed between earlier declining clusters and younger age, worse NSAA, slower rise from supine, slower 10 metre walk/run times, and younger age of steroid initiation. In order to assess the probability of observing complete loss of function for individual NSAA items, we examined the proportion of patients who shifted from a score of 1 or 2 at baseline to a score of 0. We also assessed the probability of gain of function using the inverse assessment and stratified the probability of deterioration, improvement-or static behavior-by age ranges and using baseline functional status. Using this tool, our study provides a comprehensive assessment of the NSAA in a large population of patients with DMD and, for the first time, describes discrete clusters of disease progression; this will be invaluable for future DMD clinical trial design and interpretation of findings.
Collapse
Affiliation(s)
- Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, United Kingdom
- National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- * E-mail:
| | - Joana Domingos
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, United Kingdom
| | - Adnan Y. Manzur
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, United Kingdom
| | - Anna Mayhew
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle, United Kingdom
| | - Michela Guglieri
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle, United Kingdom
| | | | - Gautam Sajeev
- Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - James Signorovitch
- Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Susan J. Ward
- Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
| |
Collapse
|
37
|
Schlaeger S, Weidlich D, Klupp E, Montagnese F, Deschauer M, Schoser B, Bublitz S, Ruschke S, Zimmer C, Rummeny EJ, Kirschke JS, Karampinos DC. Decreased water T 2 in fatty infiltrated skeletal muscles of patients with neuromuscular diseases. NMR IN BIOMEDICINE 2019; 32:e4111. [PMID: 31180167 DOI: 10.1002/nbm.4111] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 03/07/2019] [Accepted: 03/17/2019] [Indexed: 05/22/2023]
Abstract
Quantitative imaging techniques are emerging in the field of magnetic resonance imaging of neuromuscular diseases (NMD). T2 of water (T2w ) is considered an important imaging marker to assess acute and chronic alterations of the muscle fibers, being generally interpreted as an indicator for "disease activity" in the muscle tissue. To validate the accuracy and robustness of quantitative imaging methods, 1 H magnetic resonance spectroscopy (MRS) can be used as a gold standard. The purpose of the present work was to investigate T2w of remaining muscle tissue in regions of higher proton density fat fraction (PDFF) in 40 patients with defined NMD using multi-TE single-voxel 1 H MRS. Patients underwent MR measurements on a 3 T system to perform a multi-TE single-voxel stimulated echo acquisition method (STEAM) MRS (TE = 11/15/20/25(/35) ms) in regions of healthy, edematous and fatty thigh muscle tissue. Muscle regions for MRS were selected based on T2 -weighted water and fat images of a two-echo 2D Dixon TSE. MRS results were confined to regions with qualitatively defined remaining muscle tissue without edema and high fat content, based on visual grading of the imaging data. The results showed decreased T2w values with increasing PDFF with R2 = 0.45 (p < 10-3 ) (linear fit) and with R2 = 0.51 (exponential fit). The observed dependence of T2w on PDFF should be considered when using T2w as a marker in NMD imaging and when performing single-voxel MRS for T2w in regions enclosing edematous, nonedematous and fatty infiltrated muscle tissue.
Collapse
Affiliation(s)
- Sarah Schlaeger
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional of Neuroradiology, Technical University of Munich, Munich, Germany
| | - Dominik Weidlich
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany
| | - Elisabeth Klupp
- Department of Diagnostic and Interventional of Neuroradiology, Technical University of Munich, Munich, Germany
| | - Federica Montagnese
- Friedrich-Baur-Institut, Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
| | - Marcus Deschauer
- Department of Neurology, Technical University of Munich, Munich, Germany
| | - Benedikt Schoser
- Friedrich-Baur-Institut, Department of Neurology, Ludwig-Maximilians-University, Munich, Germany
| | - Sarah Bublitz
- Department of Neurology, Technical University of Munich, Munich, Germany
| | - Stefan Ruschke
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany
| | - Claus Zimmer
- Department of Diagnostic and Interventional of Neuroradiology, Technical University of Munich, Munich, Germany
| | - Ernst J Rummeny
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany
| | - Jan S Kirschke
- Department of Diagnostic and Interventional of Neuroradiology, Technical University of Munich, Munich, Germany
| | - Dimitrios C Karampinos
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany
| |
Collapse
|
38
|
Effectiveness of High-Speed T2-Corrected Multiecho MR Spectroscopic Method for Quantifying Thigh Muscle Fat Content in Boys With Duchenne Muscular Dystrophy. AJR Am J Roentgenol 2019; 212:1354-1360. [PMID: 30860898 DOI: 10.2214/ajr.18.20354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE. The purpose of this study was to investigate the potential of high-speed T2-corrected multiecho (HISTO) MR spectroscopy (MRS) for rapidly quantifying the fat content of thigh muscles in children with Duchenne muscular dystrophy (DMD). SUBJECTS AND METHODS. This study prospectively enrolled 58 boys with DMD (mean age, 7.5 years; range, 4-11 years) and 30 age-matched healthy boys (mean age, 7.2 years; range, 4-11 years) at one institution over a 1-year period. T1- and T2-weighted, multiecho Dixon, and HISTO sequences were performed on the right adductor magnus and vastus lateralis muscles. The fat fractions of these muscles were acquired from HISTO and multiecho Dixon images. An experienced radiologist graded the degree of fat infiltration of the adductor magnus and vastus lateralis muscles on axial T1-weighted images. The Bland-Altman method was used to assess the consistency and repeatability of the HISTO sequence. Pearson linear correlation analysis was used to determine the correlation coefficient relating HISTO fat fraction to multiecho Dixon fat fraction values. Spearman rank correlation analysis was used to assess the relation between the HISTO fat fraction values and T1-weighted image fat infiltration grades. The independent t test was used to compare the HISTO fat fraction values of the boys with DMD with those of the healthy control subjects. RESULTS. Bland-Altman analysis showed that 95.5% of the HISTO fat fraction values of the adductor magnus were within the 95% CI. HISTO fat fraction and multiecho Dixon fat fraction values of the adductor magnus and vastus lateralis muscles were highly positively correlated (adductor magnus, r = 0.983; vastus lateralis, r = 0.967; p < 0.0001). HISTO fat fraction values were also highly positively correlated with the grades of fat infiltration on T1-weighted images (adductor magnus, r = 0.911; vastus lateralis, r = 0.937; p < 0.0001). The HISTO fat fraction of the adductor magnus muscle was 33.3% ± 22.6% and of the vastus lateralis muscle was 25.6% ± 20.3% in patients with DMD. The corresponding values were 2.9% ± 2.1% and 2.3% ± 1.9% in the control group. The differences were statistically significant (p < 0.0001). CONCLUSION. The HISTO sequence is a rapid and feasible noninvasive MRS technique for quantifying the fat infiltration of thigh muscles in children with known or suspected DMD. It is useful for diagnosis and for assessment of disease activity and prognosis.
Collapse
|
39
|
Heskamp L, van Nimwegen M, Ploegmakers MJ, Bassez G, Deux JF, Cumming SA, Monckton DG, van Engelen BGM, Heerschap A. Lower extremity muscle pathology in myotonic dystrophy type 1 assessed by quantitative MRI. Neurology 2019; 92:e2803-e2814. [PMID: 31118244 PMCID: PMC6598795 DOI: 10.1212/wnl.0000000000007648] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 02/07/2019] [Indexed: 01/08/2023] Open
Abstract
Objective To determine the value of quantitative MRI in providing imaging biomarkers for disease in 20 different upper and lower leg muscles of patients with myotonic dystrophy type 1 (DM1). Methods We acquired images covering these muscles in 33 genetically and clinically well-characterized patients with DM1 and 10 unaffected controls. MRIs were recorded with a Dixon method to determine muscle fat fraction, muscle volume, and contractile muscle volume, and a multi-echo spin-echo sequence was used to determine T2 water relaxation time (T2water), reflecting putative edema. Results Muscles in patients with DM1 had higher fat fractions than muscles of controls (15.6 ± 11.1% vs 3.7 ± 1.5%). In addition, patients had smaller muscle volumes (902 ± 232 vs 1,097 ± 251 cm3), smaller contractile muscle volumes (779 ± 247 vs 1,054 ± 246 cm3), and increased T2water (33.4 ± 1.0 vs 31.9 ± 0.6 milliseconds), indicating atrophy and edema, respectively. Lower leg muscles were affected most frequently, especially the gastrocnemius medialis and soleus. Distribution of fat content per muscle indicated gradual fat infiltration in DM1. Between-patient variation in fat fraction was explained by age (≈45%), and another ≈14% was explained by estimated progenitor CTG repeat length (r2 = 0.485) and somatic instability (r2 = 0.590). Fat fraction correlated with the 6-minute walk test (r = −0.553) and muscular impairment rating scale (r = 0.537) and revealed subclinical muscle involvement. Conclusion This cross-sectional quantitative MRI study of 20 different lower extremity muscles in patients with DM1 revealed abnormal values for muscle fat fraction, volume, and T2water, which therefore may serve as objective biomarkers to assess disease state of skeletal muscles in these patients. ClinicalTrials.gov identifier NCT02118779.
Collapse
Affiliation(s)
- Linda Heskamp
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK.
| | - Marlies van Nimwegen
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Marieke J Ploegmakers
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Guillaume Bassez
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Jean-Francois Deux
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Sarah A Cumming
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Darren G Monckton
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Baziel G M van Engelen
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Arend Heerschap
- From the Department of Radiology and Nuclear Medicine (L.H., M.J.P., A.H.) and Department of Neurology (M.v.N., B.G.M.v.E.), Radboud University Medical Center, Nijmegen, the Netherlands; Neuromuscular Reference Center (G.B.), Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital; Department of Radiology (J.-F.D.), Henri Mondor University Hospital, Paris, France; and Institute of Molecular, Cell and Systems Biology (S.A.C., D.G.M.), College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| |
Collapse
|
40
|
Marty B, Carlier PG. Physiological and pathological skeletal muscle T1 changes quantified using a fast inversion-recovery radial NMR imaging sequence. Sci Rep 2019; 9:6852. [PMID: 31048765 PMCID: PMC6497638 DOI: 10.1038/s41598-019-43398-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 04/24/2019] [Indexed: 12/18/2022] Open
Abstract
We investigated the response of skeletal muscle global T1 under different physiological and pathological conditions using an inversion-recovery radial T1 mapping sequence. Thirty five healthy volunteers, seven patients with Becker muscular dystrophy (BMD) and seven patients with sporadic inclusion body myositis (IBM) were investigated in order to evaluate the effects of gender, age, muscle group, exercise and pathological processes on global T1 values. In addition, the intramuscular fat content was measured using 3-point Dixon and the global T2 and water T2 (T2H2O) were determined with a multi-spin-echo sequence. In the muscles of healthy volunteers, there was no impact of age on global T1. However, we measured a significant effect of sex and muscle group. After exercise, a significant 7.7% increase of global T1 was measured in the recruited muscles, and global T1 variations were highly correlated to T2H2O variations (R = 0.91). In pathologies, global T1 values were reduced in fat infiltrated muscles. When fat fraction was taken into account, global T1 values were higher in IBM patients compared to BMD. Global T1 variations are a sensitive indicator of tissue changes in skeletal muscle related to several physiological and pathological events.
Collapse
Affiliation(s)
- Benjamin Marty
- Institute of Myology, Neuromuscular Investigation Center, NMR Laboratory, Paris, France. .,CEA, DRF, IBFJ, MIRCen, NMR Laboratory, Paris, France.
| | - Pierre G Carlier
- Institute of Myology, Neuromuscular Investigation Center, NMR Laboratory, Paris, France.,CEA, DRF, IBFJ, MIRCen, NMR Laboratory, Paris, France
| |
Collapse
|
41
|
Lee-McMullen B, Chrzanowski SM, Vohra R, Forbes S, Vandenborne K, Edison AS, Walter GA. Age-dependent changes in metabolite profile and lipid saturation in dystrophic mice. NMR IN BIOMEDICINE 2019; 32:e4075. [PMID: 30848538 PMCID: PMC6777843 DOI: 10.1002/nbm.4075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 11/20/2018] [Accepted: 12/30/2018] [Indexed: 06/09/2023]
Abstract
Duchenne Muscular Dystrophy (DMD) is a fatal X-linked genetic disorder. In DMD, the absence of the dystrophin protein causes decreased sarcolemmal integrity resulting in progressive replacement of muscle with fibrofatty tissue. The effects of lacking dystrophin on muscle and systemic metabolism are still unclear. Therefore, to determine the impact of the absence of dystrophin on metabolism, we investigated the metabolic and lipid profile at two different, well-defined stages of muscle damage and stabilization in mdx mice. We measured NMR-detectable metabolite and lipid profiles in the serum and muscles of mdx mice at 6 and 24 weeks of age. Metabolites were determined in muscle in vivo using 1 H MRI/MRS, in isolated muscles using 1 H-HR-MAS NMR, and in serum using high resolution 1 H/13 C NMR. Dystrophic mice were found to have a unique lipid saturation profile compared with control mice, revealing an age-related metabolic change. In the 6-week-old mdx mice, serum lipids were increased and the degree of lipid saturation changed between 6 and 24 weeks. The serum taurine-creatine ratio increased over the life span of mdx, but not in control mice. Furthermore, the saturation index of lipids increased in the serum but decreased in the tissue over time. Finally, we demonstrated associations between MRI-T2 , a strong indicator of inflammation/edema, with tissue and serum lipid profiles. These results indicate the complex temporal changes of metabolites in the tissue and serum during repetitive bouts of muscle damage and regeneration that occur in dystrophic muscle.
Collapse
Affiliation(s)
- Brittany Lee-McMullen
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
- Department of Biochemistry and Molecular Biology, Southeast Center for Integrated Metabolomics, University of Florida, Gainesville, FL, USA
| | | | - Ravneet Vohra
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Sean Forbes
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Arthur S. Edison
- Department of Biochemistry and Molecular Biology, Southeast Center for Integrated Metabolomics, University of Florida, Gainesville, FL, USA
- Current address: Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Glenn A. Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
- Department of Biochemistry and Molecular Biology, Southeast Center for Integrated Metabolomics, University of Florida, Gainesville, FL, USA
| |
Collapse
|
42
|
Burian E, Rohrmeier A, Schlaeger S, Dieckmeyer M, Diefenbach MN, Syväri J, Klupp E, Weidlich D, Zimmer C, Rummeny EJ, Karampinos DC, Kirschke JS, Baum T. Lumbar muscle and vertebral bodies segmentation of chemical shift encoding-based water-fat MRI: the reference database MyoSegmenTUM spine. BMC Musculoskelet Disord 2019; 20:152. [PMID: 30961552 PMCID: PMC6454744 DOI: 10.1186/s12891-019-2528-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 03/24/2019] [Indexed: 12/17/2022] Open
Abstract
Background Magnetic resonance imaging (MRI) is the modality of choice for diagnosing and monitoring muscular tissue pathologies and bone marrow alterations in the context of lower back pain, neuromuscular diseases and osteoporosis. Chemical shift encoding-based water-fat MRI allows for reliable determination of proton density fat fraction (PDFF) of the muscle and bone marrow. Prior to quantitative data extraction, segmentation of the examined structures is needed. Performed manually, the segmentation process is time consuming and therefore limiting the clinical applicability. Thus, the development of automated segmentation algorithms is an ongoing research focus. Construction and content This database provides ground truth data which may help to develop and test automatic lumbar muscle and vertebra segmentation algorithms. Lumbar muscle groups and vertebral bodies (L1 to L5) were manually segmented in chemical shift encoding-based water-fat MRI and made publically available in the database MyoSegmenTUM. The database consists of water, fat and PDFF images with corresponding segmentation masks for lumbar muscle groups (right/left erector spinae and psoas muscles, respectively) and lumbar vertebral bodies 1–5 of 54 healthy Caucasian subjects. The database is freely accessible online at https://osf.io/3j54b/?view_only=f5089274d4a449cda2fef1d2df0ecc56. Conclusion A development and testing of segmentation algorithms based on this database may allow the use of quantitative MRI in clinical routine.
Collapse
Affiliation(s)
- Egon Burian
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
| | - Alexander Rohrmeier
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Sarah Schlaeger
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Michael Dieckmeyer
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.,Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Maximilian N Diefenbach
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Jan Syväri
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Elisabeth Klupp
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Dominik Weidlich
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Claus Zimmer
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Ernst J Rummeny
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Dimitrios C Karampinos
- Department of Diagnostic and Interventional Radiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Jan S Kirschke
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Thomas Baum
- Department of Diagnostic and Interventional Neuroradiology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
43
|
Batra A, Vohra RS, Chrzanowski SM, Hammers DW, Lott DJ, Vandenborne K, Walter GA, Forbes SC. Effects of PDE5 inhibition on dystrophic muscle following an acute bout of downhill running and endurance training. J Appl Physiol (1985) 2019; 126:1737-1745. [PMID: 30946638 DOI: 10.1152/japplphysiol.00664.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Lack of sarcolemma-localized neuronal nitric oxide synthase mu (nNOSμ) contributes to muscle damage and fatigue in dystrophic muscle. In this study, we examined the effects of compensating for lack of nNOSμ with a phosphodiesterase type 5 (PDE5) inhibitor in mdx mice following downhill running and endurance training. Dystrophic mice (mdx) were treated with sildenafil citrate and compared with untreated mdx and wild-type mice after an acute bout of downhill running and during a progressive low-intensity treadmill running program (5 days/wk, 4 wk). Magnetic resonance imaging (MRI) and spectroscopy (MRS) transverse relaxation time constant (T2) of hindlimb and forelimb muscles were measured as a marker of muscle damage after downhill running and throughout training. The MRI blood oxygenation level dependence (BOLD) response and 31phosphorus MRS (31P-MRS) data were acquired after stimulated muscle contractions. After downhill running, the increase in T2 was attenuated (P < 0.05) in treated mdx and wild-type mice compared with untreated mdx. During training, resting T2 values did not change in wild-type and mdx mice from baseline values; however, the running distance completed during training was greater (P < 0.05) in treated mdx (>90% of target distance) and wild-type (100%) than untreated mdx (60%). The post-contractile BOLD response was greater (P < 0.05) in treated mdx that trained than untreated mdx, with no differences in muscle oxidative capacity, as measured by 31P-MRS. Our findings indicate that PDE5 inhibition reduces muscle damage after a single bout of downhill running and improves performance during endurance training in dystrophic mice, possibly because of enhanced microvascular function. NEW & NOTEWORTHY This study examined the combined effects of PDE5 inhibition and exercise in dystrophic muscle using high-resolution magnetic resonance imaging and spectroscopy. Our findings demonstrated that sildenafil citrate reduces muscle damage after a single bout of downhill running, improves endurance-training performance, and enhances microvascular function in dystrophic muscle. Collectively, the results support the combination of exercise and PDE5 inhibition as a therapeutic approach in muscular dystrophies lacking nNOSμ.
Collapse
Affiliation(s)
- Abhinandan Batra
- Department of Physical Therapy, University of Florida , Gainesville, Florida
| | - Ravneet S Vohra
- Department of Physical Therapy, University of Florida , Gainesville, Florida
| | - Steve M Chrzanowski
- Department of Physiology and Therapeutics, University of Florida , Gainesville, Florida
| | - David W Hammers
- Department of Pharmacology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Donovan J Lott
- Department of Physical Therapy, University of Florida , Gainesville, Florida
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida , Gainesville, Florida
| | - Glenn A Walter
- Department of Physiology and Therapeutics, University of Florida , Gainesville, Florida
| | - Sean C Forbes
- Department of Physical Therapy, University of Florida , Gainesville, Florida
| |
Collapse
|
44
|
Yin L, Xie ZY, Xu HY, Zheng SS, Wang ZX, Xiao JX, Yuan Y. T2 Mapping and Fat Quantification of Thigh Muscles in Children with Duchenne Muscular Dystrophy. Curr Med Sci 2019; 39:138-145. [PMID: 30868504 DOI: 10.1007/s11596-019-2012-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/12/2018] [Indexed: 11/24/2022]
Abstract
Quantitative magnetic resonance image (MRI) in individual muscles may be useful for monitoring disease progression in Duchenne muscular dystrophy (DMD). The purpose of this study was to measure T2 relaxation time of thigh muscles in children with DMD and healthy boys, and to correlate the T2 relaxation time of muscles with the fat fraction (FF) at quantitative magnetic resonance and results of clinical assessment. Thirty-two boys with DMD and 18 healthy boys were evaluated with T2 mapping and three-point Dixon MRI. Age, body mass index (BMI), muscle strength assessment, timed functional tests (time to walk or run 10 metres, rise from the floor and ascend four stairs), and the North Star Ambulatory Assessment (NSAA) were evaluated. Spearman's correlation was used to assess the relationships between FF and clinical assessments and T2 relaxation time. The mean T2 relaxation time of thigh muscles in DMD was significantly longer than that in the control group (P<0.05), except for the gracilis (P=0.952). The gracilis, sartorius and adductor longus were relatively spared by fatty infiltration in DMD patients. The T2 relaxation time was correlated significantly with the mean FF in all muscles. Age, BMI, total muscle strength score, timed functional tests and NSAA were significantly correlated with the overall mean T2 relaxation time. T2 mapping may prove clinically useful in monitoring muscle changes as a result of the disease process and in predicting the outcome of DMD patients.
Collapse
Affiliation(s)
- Liang Yin
- Department of Radiology, Peking University First Hospital, Beijing, 100034, China
| | - Zhi-Ying Xie
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China
| | - Hai-Yan Xu
- Department of Radiology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Sui-Sheng Zheng
- Department of Radiology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhao-Xia Wang
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China
| | - Jiang-Xi Xiao
- Department of Radiology, Peking University First Hospital, Beijing, 100034, China.
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
45
|
Paoletti M, Pichiecchio A, Cotti Piccinelli S, Tasca G, Berardinelli AL, Padovani A, Filosto M. Advances in Quantitative Imaging of Genetic and Acquired Myopathies: Clinical Applications and Perspectives. Front Neurol 2019; 10:78. [PMID: 30804884 PMCID: PMC6378279 DOI: 10.3389/fneur.2019.00078] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 01/21/2019] [Indexed: 12/11/2022] Open
Abstract
In the last years, magnetic resonance imaging (MRI) has become fundamental for the diagnosis and monitoring of myopathies given its ability to show the severity and distribution of pathology, to identify specific patterns of damage distribution and to properly interpret a number of genetic variants. The advances in MR techniques and post-processing software solutions have greatly expanded the potential to assess pathological changes in muscle diseases, and more specifically of myopathies; a number of features can be studied and quantified, ranging from composition, architecture, mechanical properties, perfusion, and function, leading to what is known as quantitative MRI (qMRI). Such techniques can effectively provide a variety of information beyond what can be seen and assessed by conventional MR imaging; their development and application in clinical practice can play an important role in the diagnostic process and in assessing disease course and treatment response. In this review, we briefly discuss the current role of muscle MRI in diagnosing muscle diseases and describe in detail the potential and perspectives of the application of advanced qMRI techniques in this field.
Collapse
Affiliation(s)
- Matteo Paoletti
- Neuroradiology Department, IRCCS Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Anna Pichiecchio
- Neuroradiology Department, IRCCS Mondino Foundation, Pavia, Italy.,Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Stefano Cotti Piccinelli
- Unit of Neurology, Center for Neuromuscular Diseases, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Giorgio Tasca
- Neurology Department, Dipartimento di Scienze dell'Invecchiamento, Neurologiche, Ortopediche e della Testa-Collo, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Alessandro Padovani
- Unit of Neurology, Center for Neuromuscular Diseases, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Massimiliano Filosto
- Unit of Neurology, Center for Neuromuscular Diseases, ASST Spedali Civili and University of Brescia, Brescia, Italy
| |
Collapse
|
46
|
Park JS, Vohra R, Klussmann T, Bengtsson NE, Chamberlain JS, Lee D. Non-invasive tracking of disease progression in young dystrophic muscles using multi-parametric MRI at 14T. PLoS One 2018; 13:e0206323. [PMID: 30365532 PMCID: PMC6203357 DOI: 10.1371/journal.pone.0206323] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 10/10/2018] [Indexed: 12/02/2022] Open
Abstract
In this study, multi-parametric magnetic resonance imaging (MRI) was conducted to monitor skeletal muscle changes in dystrophic (mdx4cv) and age-matched control (C57BL/6J) mice starting at 3 weeks of age. The objective of this study was to evaluate and characterize changes in muscle tissue characteristics of hind limbs in young, dystrophic mice using MRI. Mdx4cv (n = 25) and age-matched C57BL/6J (n = 5) were imaged at 3, 5, 7, 9, and 11 weeks of age. Multiple MR measurements were taken from the tibialis anterior, gastrocnemius, and soleus muscles. There were significant differences between dystrophic and control groups for all three muscle types when comparing transverse relaxation times (T2) in lower hind limb muscles. Additionally, fractional anisotropy, radial diffusivity, and eigenvalue analysis of diffusion tensor imaging also demonstrated significant differences between groups. Longitudinal relaxation times (T1) displayed no significant differences between groups. The earliest time points in the magnetization transfer ratio measurements displayed a significant difference. Histological analysis revealed significant differences in the tibialis anterior and gastrocnemius muscles between groups with the mdx mice displaying greater variability in muscle fiber size in later time points. The multi-parametric MRI approach offers a promising alternative for future development of a noninvasive avenue for tracking both disease progression and treatment response.
Collapse
Affiliation(s)
- Joshua S. Park
- Department of Radiology, University of Washington, Seattle, WA, United States of America
| | - Ravneet Vohra
- Department of Radiology, University of Washington, Seattle, WA, United States of America
| | - Thomas Klussmann
- Department of Radiology, University of Washington, Seattle, WA, United States of America
| | - Niclas E. Bengtsson
- Department of Neurology, University of Washington, Seattle, WA, United States of America
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Washington, Seattle, WA, United States of America
| | - Jeffrey S. Chamberlain
- Department of Neurology, University of Washington, Seattle, WA, United States of America
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Washington, Seattle, WA, United States of America
- Department of Biochemistry, University of Washington, Seattle, WA, United States of America
- Department of Medicine, University of Washington, Seattle, WA, United States of America
| | - Donghoon Lee
- Department of Radiology, University of Washington, Seattle, WA, United States of America
- * E-mail:
| |
Collapse
|
47
|
T2-Weighted Dixon Turbo Spin Echo for Accelerated Simultaneous Grading of Whole-Body Skeletal Muscle Fat Infiltration and Edema in Patients With Neuromuscular Diseases. J Comput Assist Tomogr 2018; 42:574-579. [PMID: 29613984 DOI: 10.1097/rct.0000000000000723] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The assessment of fatty infiltration and edema in the musculature of patients with neuromuscular diseases (NMDs) typically requires the separate performance of T1-weighted and fat-suppressed T2-weighted sequences. T2-weighted Dixon turbo spin echo (TSE) enables the generation of T2-weighted fat- and water-separated images, which can be used to assess both pathologies simultaneously. The present study examines the diagnostic performance of T2-weighted Dixon TSE compared with the standard sequences in 10 patients with NMDs and 10 healthy subjects. METHODS Whole-body magnetic resonance imaging was performed including T1-weighted Dixon fast field echo, T2-weighted short-tau inversion recovery, and T2-weighted Dixon TSE. Fatty infiltration and intramuscular edema were rated by 2 radiologists using visual semiquantitative rating scales. To assess intermethod and interrater agreement, weighted Cohen's κ coefficients were calculated. RESULTS The ratings of fatty infiltration showed high intermethod and high interrater agreement (T1-weighted Dixon fast field echo vs T2-weighted Dixon TSE fat image). The evaluation of edematous changes showed high intermethod and good interrater agreement (T2-weighted short-tau inversion recovery vs T2-weighted Dixon TSE water image). CONCLUSIONS T2-weighted Dixon TSE imaging is an alternative for accelerated simultaneous grading of whole-body skeletal muscle fat infiltration and edema in patients with NMDs.
Collapse
|
48
|
Thigh muscle segmentation of chemical shift encoding-based water-fat magnetic resonance images: The reference database MyoSegmenTUM. PLoS One 2018; 13:e0198200. [PMID: 29879128 PMCID: PMC5991744 DOI: 10.1371/journal.pone.0198200] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 05/15/2018] [Indexed: 01/10/2023] Open
Abstract
Magnetic resonance imaging (MRI) can non-invasively assess muscle anatomy, exercise effects and pathologies with different underlying causes such as neuromuscular diseases (NMD). Quantitative MRI including fat fraction mapping using chemical shift encoding-based water-fat MRI has emerged for reliable determination of muscle volume and fat composition. The data analysis of water-fat images requires segmentation of the different muscles which has been mainly performed manually in the past and is a very time consuming process, currently limiting the clinical applicability. An automatization of the segmentation process would lead to a more time-efficient analysis. In the present work, the manually segmented thigh magnetic resonance imaging database MyoSegmenTUM is presented. It hosts water-fat MR images of both thighs of 15 healthy subjects and 4 patients with NMD with a voxel size of 3.2x2x4 mm3 with the corresponding segmentation masks for four functional muscle groups: quadriceps femoris, sartorius, gracilis, hamstrings. The database is freely accessible online at https://osf.io/svwa7/?view_only=c2c980c17b3a40fca35d088a3cdd83e2. The database is mainly meant as ground truth which can be used as training and test dataset for automatic muscle segmentation algorithms. The segmentation allows extraction of muscle cross sectional area (CSA) and volume. Proton density fat fraction (PDFF) of the defined muscle groups from the corresponding images and quadriceps muscle strength measurements/neurological muscle strength rating can be used for benchmarking purposes.
Collapse
|
49
|
Monitoring skeletal muscle chronic fatty degenerations with fast T1-mapping. Eur Radiol 2018; 28:4662-4668. [PMID: 29713767 DOI: 10.1007/s00330-018-5433-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/05/2018] [Accepted: 03/16/2018] [Indexed: 10/17/2022]
Abstract
OBJECTIVES To develop a fast, high-resolution T1-mapping sequence dedicated to skeletal muscle imaging, and to evaluate the potential of T1 as a robust and sensitive biomarker for the monitoring of chronic fatty degenerations in a dystrophic disease. METHODS The magnetic resonance imaging sequence consisted of the acquisition of a 1,000-radial-spokes FLASH echo-train following magnetisation inversion, resulting in 10s scan time per slice. Temporal image series were reconstructed using compressed sensing and T1 maps were computed using Bloch simulations. Ten healthy volunteers and 30 patients suffering from Becker muscular dystrophy (BMD) participated in this prospective study, in order to evaluate the repeatability, the precision and the sensitivity of the proposed approach. Intramuscular fat fraction (FF) was also measured using a standard three-point Dixon method. The protocol was approved by a local ethics committee. RESULTS The mean T1 evaluated in the thighs muscles of healthy volunteers was 1,199 ± 45 ms, with a coefficient of reproducibility of 2.3%. Mean T1 values were statistically decreased in the thighs of BMD patients and were linearly correlated with intramuscular FF (R = -0.98). CONCLUSIONS T1-mapping is a good candidate for fast, sensitive and quantitative monitoring of fatty infiltrations in neuromuscular disorders. KEY POINTS • A T1 mapping sequence dedicated to skeletal muscle imaging was implemented. • The acquisition time was 10 s per slice. • Muscle T1 values were significantly decreased in dystrophic muscles compared to healthy muscles. • T1 values correlated with intramuscular fat fraction measured by three-point Dixon. • T1 represents an alternative biomarker for monitoring fatty infiltrations in neuromuscular disorders.
Collapse
|
50
|
Barnard AM, Willcocks RJ, Finanger EL, Daniels MJ, Triplett WT, Rooney WD, Lott DJ, Forbes SC, Wang DJ, Senesac CR, Harrington AT, Finkel RS, Russman BS, Byrne BJ, Tennekoon GI, Walter GA, Sweeney HL, Vandenborne K. Skeletal muscle magnetic resonance biomarkers correlate with function and sentinel events in Duchenne muscular dystrophy. PLoS One 2018; 13:e0194283. [PMID: 29554116 PMCID: PMC5858773 DOI: 10.1371/journal.pone.0194283] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/28/2018] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To provide evidence for quantitative magnetic resonance (qMR) biomarkers in Duchenne muscular dystrophy by investigating the relationship between qMR measures of lower extremity muscle pathology and functional endpoints in a large ambulatory cohort using a multicenter study design. METHODS MR spectroscopy and quantitative imaging were implemented to measure intramuscular fat fraction and the transverse magnetization relaxation time constant (T2) in lower extremity muscles of 136 participants with Duchenne muscular dystrophy. Measures were collected at 554 visits over 48 months at one of three imaging sites. Fat fraction was measured in the soleus and vastus lateralis using MR spectroscopy, while T2 was assessed using MRI in eight lower extremity muscles. Ambulatory function was measured using the 10m walk/run, climb four stairs, supine to stand, and six minute walk tests. RESULTS Significant correlations were found between all qMR and functional measures. Vastus lateralis qMR measures correlated most strongly to functional endpoints (|ρ| = 0.68-0.78), although measures in other rapidly progressing muscles including the biceps femoris (|ρ| = 0.63-0.73) and peroneals (|ρ| = 0.59-0.72) also showed strong correlations. Quantitative MR biomarkers were excellent indicators of loss of functional ability and correlated with qualitative measures of function. A VL FF of 0.40 was an approximate lower threshold of muscle pathology associated with loss of ambulation. DISCUSSION Lower extremity qMR biomarkers have a robust relationship to clinically meaningful measures of ambulatory function in Duchenne muscular dystrophy. These results provide strong supporting evidence for qMR biomarkers and set the stage for their potential use as surrogate outcomes in clinical trials.
Collapse
Affiliation(s)
- Alison M. Barnard
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Rebecca J. Willcocks
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Erika L. Finanger
- Departments of Pediatrics and Neurology, Oregon Health & Science University, Portland, OR, United States of America
| | - Michael J. Daniels
- Department of Statistics, University of Florida, Gainesville, FL, United States of America
| | - William T. Triplett
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - William D. Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, United States of America
| | - Donovan J. Lott
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Sean C. Forbes
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Dah-Jyuu Wang
- Department of Radiology, Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Claudia R. Senesac
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Ann T. Harrington
- The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | | | - Barry S. Russman
- Departments of Pediatrics and Neurology, Oregon Health & Science University, Portland, OR, United States of America
| | - Barry J. Byrne
- Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center, University of Florida, Gainesville, FL, United States of America
| | - Gihan I. Tennekoon
- The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Glenn A. Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States of America
| | - H. Lee Sweeney
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States of America
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
- * E-mail:
| |
Collapse
|