1
|
Maden Ç, Karabulut DG, Yiğit S. Validity and Reliability of the 6-min Pegboard and Ring Test in Patients With Duchenne Muscular Dystrophy. Muscle Nerve 2025; 71:200-207. [PMID: 39660725 DOI: 10.1002/mus.28314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/12/2024]
Abstract
INTRODUCTION/AIMS Tests for assessing upper extremity (UE) functional capacity in patients with Duchenne muscular dystrophy (DMD) are limited. This study aimed to evaluate the validity and reliability of the 6-min pegboard and ring test (6PBRT) as a practical tool for this purpose. METHODS Children with DMD (n = 22) were evaluated using the 6PBRT for UE functional capacity, the Quick Disabilities of the Arm, Shoulder, and Hand (Q-DASH) for functionality, the Pediatric Quality of Life Inventory (PedsQL) for quality of life, and a dynamometer for handgrip strength and UE muscle strength. RESULTS The 6PBRT showed excellent test-retest reliability, with an intraclass correlation coefficient (ICC) of 0.978 (95% confidence interval, 0.946-0.984). A very strong positive correlation was observed between the test and retest 6PBRT mean scores (r = 0.981). The mean 6PBRT score exhibited moderate-to-strong correlations with handgrip strength (r = 0.653, r = 0.646, right/left, respectively), muscle strength (shoulder flexors [r = 0.793, r = 0.797, right/left, respectively], shoulder abductors (r = 0.763, r = 0.743, right/left, respectively), elbow flexors [r = 0.743, r = 0.755, right/left, respectively]), mean Q-DASH score (r = -0.555), and mean PedsQL score (r = 0.611). DISCUSSION The 6PBRT appears to be a valid and reliable measure for assessing upper extremity functional capacity in patients with DMD. This test is suitable for patients who are able to lift both hands above their heads. TRIAL REGISTRATION NCT06174025.
Collapse
Affiliation(s)
- Çağtay Maden
- Faculty of Health Sciences, Department of Physiotherapy and Rehabilitation, Gaziantep Islam Science and Technology University, Gaziantep, Turkey
| | - Demet Gözaçan Karabulut
- Faculty of Health Sciences, Department of Physiotherapy and Rehabilitation, Gaziantep Islam Science and Technology University, Gaziantep, Turkey
| | - Sedat Yiğit
- Faculty of Health Sciences, Department of Physiotherapy and Rehabilitation, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
2
|
Hooijmans MT, Schlaffke L, Bolsterlee B, Schlaeger S, Marty B, Mazzoli V. Compositional and Functional MRI of Skeletal Muscle: A Review. J Magn Reson Imaging 2024; 60:860-877. [PMID: 37929681 PMCID: PMC11070452 DOI: 10.1002/jmri.29091] [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: 08/18/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
Due to its exceptional sensitivity to soft tissues, MRI has been extensively utilized to assess anatomical muscle parameters such as muscle volume and cross-sectional area. Quantitative Magnetic Resonance Imaging (qMRI) adds to the capabilities of MRI, by providing information on muscle composition such as fat content, water content, microstructure, hypertrophy, atrophy, as well as muscle architecture. In addition to compositional changes, qMRI can also be used to assess function for example by measuring muscle quality or through characterization of muscle deformation during passive lengthening/shortening and active contractions. The overall aim of this review is to provide an updated overview of qMRI techniques that can quantitatively evaluate muscle structure and composition, provide insights into the underlying biological basis of the qMRI signal, and illustrate how qMRI biomarkers of muscle health relate to function in healthy and diseased/injured muscles. While some applications still require systematic clinical validation, qMRI is now established as a comprehensive technique, that can be used to characterize a wide variety of structural and compositional changes in healthy and diseased skeletal muscle. Taken together, multiparametric muscle MRI holds great potential in the diagnosis and monitoring of muscle conditions in research and clinical applications. EVIDENCE LEVEL: 5 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Melissa T Hooijmans
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Lara Schlaffke
- Department of Neurology BG-University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Bart Bolsterlee
- Neuroscience Research Australia (NeuRA), Sydney, New South Wales, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Sarah Schlaeger
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Radiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Benjamin Marty
- Institute of Myology, Neuromuscular Investigation Center, NMR Laboratory, Paris, France
| | - Valentina Mazzoli
- Department of Radiology, Stanford University, Stanford, California, USA
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, NYU Langone Medical Center, New York, New York, USA
| |
Collapse
|
3
|
Woodcock IR, Tachas G, Desem N, Houweling PJ, Kean M, Emmanuel J, Kennedy R, Carroll K, de Valle K, Adams J, Lamandé SR, Coles C, Tiong C, Burton M, Villano D, Button P, Hogrel JY, Catling-Seyffer S, Ryan MM, Delatycki MB, Yiu EM. A phase 2 open-label study of the safety and efficacy of weekly dosing of ATL1102 in patients with non-ambulatory Duchenne muscular dystrophy and pharmacology in mdx mice. PLoS One 2024; 19:e0294847. [PMID: 38271438 PMCID: PMC10810432 DOI: 10.1371/journal.pone.0294847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/19/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND ATL1102 is a 2'MOE gapmer antisense oligonucleotide to the CD49d alpha subunit of VLA-4, inhibiting expression of CD49d on lymphocytes, reducing survival, activation and migration to sites of inflammation. Children with DMD have dystrophin deficient muscles susceptible to contraction induced injury, which triggers the immune system, exacerbating muscle damage. CD49d is a biomarker of disease severity in DMD, with increased numbers of high CD49d expressing T cells correlating with more severe and progressive weakess, despite corticosteroid treatment. METHODS This Phase 2 open label study assessed the safety, efficacy and pharmacokinetic profile of ATL1102 administered as 25 mg weekly by subcutaneous injection for 24 weeks in 9 non-ambulatory boys with DMD aged 10-18 years. The main objective was to assess safety and tolerability of ATL1102. Secondary objectives included the effect of ATL1102 on lymphocyte numbers in the blood, functional changes in upper limb function as assessed by Performance of Upper Limb test (PUL 2.0) and upper limb strength using MyoGrip and MyoPinch compared to baseline. RESULTS Eight out of nine participants were on a stable dose of corticosteroids. ATL1102 was generally safe and well tolerated. No serious adverse events were reported. There were no participant withdrawals from the study. The most commonly reported adverse events were injection site erythema and skin discoloration. There was no statistically significant change in lymphocyte count from baseline to week 8, 12 or 24 of dosing however, the CD3+CD49d+ T lymphocytes were statistically significantly higher at week 28 compared to week 24, four weeks past the last dose (mean change 0.40x109/L 95%CI 0.05, 0.74; p = 0.030). Functional muscle strength, as measured by the PUL2.0, EK2 and Myoset grip and pinch measures, and MRI fat fraction of the forearm muscles were stable throughout the trial period. CONCLUSION ATL1102, a novel antisense drug being developed for the treatment of inflammation that exacerbates muscle fibre damage in DMD, appears to be safe and well tolerated in non-ambulant boys with DMD. The apparent stabilisation observed on multiple muscle disease progression parameters assessed over the study duration support the continued development of ATL1102 for the treatment of DMD. TRIAL REGISTRATION Clinical Trial Registration. Australian New Zealand Clinical Trials Registry Number: ACTRN12618000970246.
Collapse
Affiliation(s)
- Ian R. Woodcock
- Department of Neurology, The Royal Children’s Hospital, Melbourne, Australia
- The Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | | | - Nuket Desem
- Antisense Therapeutics Ltd, Melbourne, Australia
| | - Peter J. Houweling
- The Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Michael Kean
- Department of Medical Imaging, The Royal Children’s Hospital, Melbourne, Australia
| | - Jaiman Emmanuel
- Department of Medical Imaging, The Royal Children’s Hospital, Melbourne, Australia
| | - Rachel Kennedy
- Department of Neurology, The Royal Children’s Hospital, Melbourne, Australia
- The Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Physiotherapy, University of Melbourne, Melbourne, Australia
| | - Kate Carroll
- Department of Neurology, The Royal Children’s Hospital, Melbourne, Australia
- The Murdoch Children’s Research Institute, Melbourne, Australia
| | - Katy de Valle
- Department of Neurology, The Royal Children’s Hospital, Melbourne, Australia
- The Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Physiotherapy, University of Melbourne, Melbourne, Australia
| | - Justine Adams
- The Murdoch Children’s Research Institute, Melbourne, Australia
| | - Shireen R. Lamandé
- The Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Chantal Coles
- The Murdoch Children’s Research Institute, Melbourne, Australia
| | - Chrystal Tiong
- The Murdoch Children’s Research Institute, Melbourne, Australia
| | - Matthew Burton
- The Murdoch Children’s Research Institute, Melbourne, Australia
| | - Daniella Villano
- Department of Neurology, The Royal Children’s Hospital, Melbourne, Australia
| | | | | | - Sarah Catling-Seyffer
- Department of Neurology, The Royal Children’s Hospital, Melbourne, Australia
- The Murdoch Children’s Research Institute, Melbourne, Australia
| | - Monique M. Ryan
- Department of Neurology, The Royal Children’s Hospital, Melbourne, Australia
- The Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Martin B. Delatycki
- Victorian Clinical Genetics Service, Melbourne, Australia
- Murdoch Children’s Research Institute, Bruce Lefroy Centre for Genetic Health Research, Melbourne, Australia
| | - Eppie M. Yiu
- Department of Neurology, The Royal Children’s Hospital, Melbourne, Australia
- The Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| |
Collapse
|
4
|
Decostre V, De Antonio M, Servais L, Hogrel JY. Relationship Between Hand Strength and Function in Duchenne Muscular Dystrophy and Spinal Muscular Atrophy: Implications for Clinical Trials. J Neuromuscul Dis 2024; 11:777-790. [PMID: 38788084 PMCID: PMC11307056 DOI: 10.3233/jnd-230182] [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] [Accepted: 04/16/2024] [Indexed: 05/26/2024]
Abstract
Background Measurement of muscle strength and motor function is recommended in clinical trials of neuromuscular diseases, but the loss of hand strength at which motor function is impacted is not documented. Objectives To establish the relationship between hand strength and function, and to determine the strength threshold that differentiates normal and abnormal hand function in individuals with Duchenne Muscular Dystrophy (DMD) or Spinal Muscular Atrophy (SMA). Methods Maximal handgrip and key pinch strength were measured with the MyoGrip and MyoPinch dynamometers, respectively. Hand function was assessed using the MoviPlate, the Motor Function Measure items for distal upper limb (MFM-D3-UL) and the Cochin Hand Function Scale (CHFS). Results Data from 168 participants (91 DMD and 77 SMA, age 6-31 years) were analyzed. Relationships between strength and function were significant (P < 0.001). Hand function was generally preserved when strength was above the strength threshold determined by Receiver-Operating Characteristic (ROC) analysis: For MFM-D3-UL, the calculated handgrip strength thresholds were 41 and 13% of the predicted strength for a healthy subject (% pred) and the key pinch strength thresholds were 42 and 26% pred for DMD and SMA, respectively. For the MoviPlate, handgrip strength thresholds were 11 and 8% pred and key pinch strength thresholds were 21 and 11% pred for DMD and SMA, respectively. For participants with sub-threshold strength, hand function scores decreased with decreasing strength. At equal % pred strength, individuals with SMA had better functional scores than those with DMD. Conclusions Hand function is strength-dependent for most motor tasks. It declines only when strength falls below a disease-specific threshold. Therefore, therapies capable of maintaining strength above this threshold should preserve hand function.
Collapse
Affiliation(s)
| | - Marie De Antonio
- Institut de Myologie, GH Pitié-Salpêtrière, Paris, France
- Present address: Direction de la Recherche Clinique et de l’Innovation, Centre hospitalier universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Laurent Servais
- Institut de Myologie, GH Pitié-Salpêtrière, Paris, France
- Present address: Department of Paediatrics, MDUK Oxford Neuromuscular Centre & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
- Present address: Department of Pediatrics, Division of Child Neurology, Centre de Référence des Maladies Neuromusculaires, University Hospital Liège and University of Liège, Liège, Belgium
| | | |
Collapse
|
5
|
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
|
6
|
Lin CW, Shieh JY, Tsui PH, Chen CL, Lu CH, Hung YH, Lee HY, Weng WC, Gau SSF. Acoustic radiation force impulse shear wave elastography quantifies upper limb muscle in patients with Duchenne muscular dystrophy. ULTRASONICS SONOCHEMISTRY 2023; 101:106661. [PMID: 37924615 PMCID: PMC10641721 DOI: 10.1016/j.ultsonch.2023.106661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/09/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023]
Abstract
We investigated whether the upper limb muscle stiffness quantified by the acoustic radiation force impulse shear wave elastography (ARFI/SWE) is a potential biomarker for age-related muscle alteration and functional decline in patients with Duchenne muscular dystrophy (DMD). 37 patients with DMD and 30 typically developing controls (TDC) were grouped by age (3-8, 9-11, and 12-18 years). ARFI/SWE measured the biceps and deltoid muscle's shear wave velocities (SWVs). Performance of Upper Limb Module (PUL 1.2 module) assessed muscle function in DMD patients. Mann Whitney test compared muscle SWVs between DMD and TDC, stratified by three age groups. We used analysis of variance with Bonferroni correction to compare muscle SWVs between DMD and TDC and correlated muscle SWVs with PUL results in the DMD group. Results showed that the SWVs of biceps differentiated DMD patients from TDC across age groups. Younger DMD patients (3-8 years) exhibited higher SWVs (p = 0.013), but older DMD patients (12-18 years) showed lower SWVS (p = 0.028) than same-aged TDC. DMD patients had decreasing biceps SWVs with age (p < 0.001), with no such age effect in TDC. The SWVs of deltoid and biceps positively correlated with PUL scores (r = 0.527 ∼ 0.897, P < 0.05) and negatively correlated with PUL timed measures (r = -0.425 ∼ -0.542, P < 0.05) in DMD patients. Our findings suggest that ARFI/SWE quantifying the SWVs in upper limb muscle could be a potential biomarker to differentiate DMD from TDC across ages and that DMD patients showed age-related muscle alteration and limb functional decline.
Collapse
Affiliation(s)
- Chia-Wei Lin
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, and College of Medicine, National Taiwan University, No. 7 Chung-Shan South Road, Taipei 10002, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, No.1, Chang-Te St., Taipei 10048, Taipei, Taiwan
| | - Jeng-Yi Shieh
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, and College of Medicine, National Taiwan University, No. 7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Po-Hsiang Tsui
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan Dist, Tao-Yuan City 33302, Taiwan
| | - Chia-Ling Chen
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, and College of Medicine, National Taiwan University, No. 7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Chun-Hao Lu
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan Dist, Tao-Yuan City 33302, Taiwan
| | - Yi-Hsuan Hung
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, and College of Medicine, National Taiwan University, No. 7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Hsiao-Yuan Lee
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital Hsin-Chu Branch, No. 2, Sec. 1, Shengyi Rd., Zhubei City, Hsinchu County 302, Taiwan
| | - Wen-Chin Weng
- Department of Pediatrics, National Taiwan University Hospital, and College of Medicine, National Taiwan University, No. 7, Chung-Shan South Road, Taipei, Taiwan 10002, Taiwan; Department of Pediatric Neurology, National Taiwan University Children's Hospital, No. 7, Chung-Shan South Road, Taipei, Taiwan 10002, Taiwan.
| | - Susan Shur-Fen Gau
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, No.1, Chang-Te St., Taipei 10048, Taipei, Taiwan; Department of Psychiatry, National Taiwan University Hospital, and College of Medicine, No. 7 Chung-Shan South Road, Taipei 10002, Taiwan; Graduate Institute of Brain and Mind Sciences, National Taiwan University, No.1 Jen Ai road section 1, Taipei 100 Taiwan.
| |
Collapse
|
7
|
Wood CL, Hollingsworth KG, Bokaie E, Hughes E, Muni-Lofra R, Mayhew A, Mitchell RT, Guglieri M, McElvaney J, Cheetham TD, Straub V. Is ongoing testosterone required after pubertal induction in Duchenne muscular dystrophy? Endocr Connect 2023; 12:EC-23-0245. [PMID: 37768006 PMCID: PMC10620460 DOI: 10.1530/ec-23-0245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/28/2023] [Indexed: 09/29/2023]
Abstract
Glucocorticoids (GC) reduce inflammation and preserve muscle function in boys with Duchenne muscular dystrophy (DMD) but cause pubertal delay. Pubertal induction with testosterone is recommended but longer-term outcome is unknown. OBJECTIVE To assess hypothalamic-pituitary-gonadal axis, muscle volume and function 5 years after pubertal induction. METHODS A prospective observational follow-up of a clinical study was conducted. 15 GC-treated males with DMD were treated with incremental testosterone for 2 years (end of regimen +2y) then evaluated at +2.5y and +5y (final follow-up~ 3 years after last injection). Data collected included testicular volume (TV), gonadotrophin, testosterone, inhibin B, muscle function and limb muscle MRI. RESULTS Participants were 18.7 years (SD 1.6) at final follow-up and had been on GC for 11.2 years (SD 2.2). Testosterone levels were similar at +2.5y (8.6nmol/l (SD 3.4) and 5y (11.0 nmol/l (SD 6.1). TV increased from 2.8 mls (SD 0.9) at +2y to 7.1 mls (SD 1.8) then 10.6 mls (SD 3.5) at +2.5y and +5.0y(p<0.001). Inhibin B levels increased from 55.6 pg/ml (SD 47.0) at baseline to 158.2 pg/ml (SD 87.6), p=0.004 at 5y but remained lower than reference values (mean 305 pg/ml). Muscle contractile bulk decreased. INTERPRETATION Pubertal induction with testosterone in DMD is associated with HPG axis activation and ongoing increases in Inhibin B, TV and testosterone concentrations. Some patients have normal levels which is promising regarding future fertility. Given the beneficial impact of testosterone on bone health, muscle and wellbeing, monitoring testosterone levels in this population and supplementation of sub-optimal levels is important.
Collapse
Affiliation(s)
- Claire L Wood
- Department of Paediatric Endocrinology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Kieren G Hollingsworth
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Edrina Bokaie
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Eric Hughes
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Robert Muni-Lofra
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Anna Mayhew
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The University of Edinburgh, Queens Medical Research Institute, Edinburgh, UK
| | - Michela Guglieri
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Joseph McElvaney
- Department of Paediatric Endocrinology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Timothy D Cheetham
- Department of Paediatric Endocrinology, Royal Victoria Infirmary, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Volker Straub
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
8
|
Wang F, Fang S, Li J, Yuan L, Hou B, Zhu J, Jiao Y, Liu Z, Qian M, Santini F, Wang Q, Chen L, Feng F. Correlation analysis of quantitative MRI measurements of thigh muscles with histopathology in patients with idiopathic inflammatory myopathy. Eur Radiol Exp 2023; 7:51. [PMID: 37589922 PMCID: PMC10435435 DOI: 10.1186/s41747-023-00350-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 04/29/2023] [Indexed: 08/18/2023] Open
Abstract
OBJECTIVES To validate the correlation between histopathological findings and quantitative magnetic resonance imaging (qMRI) fat fraction (FF) and water T2 mapping in patients with idiopathic inflammatory myopathy (IIM). METHODS The study included 13 patients with histopathologically confirmed IIM who underwent dedicated thigh qMRI scanning within 1 month before open muscle biopsy. For the biopsied muscles, FF derived from the iterative decomposition of water and fat with echo asymmetry and least-squares estimation quantitation (IDEAL-IQ) and T2 time from T2 mapping with chemical shift selective fat saturation were measured using a machine learning software. Individual histochemical and immunohistochemical slides were evaluated using a 5-point Likert score. Inter-reader agreement and the correlation between qMRI markers and histopathological scores were analyzed. RESULTS Readers showed good to perfect agreement in qMRI measurements and most histopathological scores. FF of the biopsied muscles was positively correlated with the amount of fat in histopathological slides (p = 0.031). Prolonged T2 time was associated with the degree of variation in myofiber size, inflammatory cell infiltration, and amount of connective tissues (p ≤ 0.008 for all). CONCLUSIONS Using the machine learning-based muscle segmentation method, a positive correlation was confirmed between qMRI biomarkers and histopathological findings of patients with IIM. This finding provides a basis for using qMRI as a non-invasive tool in the diagnostic workflow of IIM. RELEVANCE STATEMENT By using ML-based muscle segmentation, a correlation between qMRI biomarkers and histopathology was found in patients with IIM: qMRI is a potential non-invasive tool in this clinical setting. KEY POINTS • Quantitative magnetic resonance imaging measurements using machine learning-based muscle segmentation have good consistency and reproductivity. • Fat fraction of idiopathic inflammatory myopathy (IIM) correlated with the amount of fat at histopathology. • Prolonged T2 time was associated with muscle inflammation in IIM.
Collapse
Affiliation(s)
- Fengdan Wang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Shiyuan Fang
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia Li
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ling Yuan
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Hou
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jinxia Zhu
- MR Collaboration, Siemens Healthineers Ltd., Beijing, China
| | - Yang Jiao
- Department of General Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi Liu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Qian
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Francesco Santini
- Department of Research and Analytic Services, University Hospital Basel, Basel, Switzerland.
- Radiological Physics, University Hospital Basel, Basel, Switzerland.
- Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland.
| | - Qian Wang
- Department of Rheumatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Chen
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feng Feng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
Pane M, Coratti G, Brogna C, Bovis F, D'Amico A, Pegoraro E, Bello L, Sansone V, Albamonte E, Ferraroli E, Mazzone ES, Fanelli L, Messina S, Catteruccia M, Cicala G, Ricci M, Frosini S, De Luca G, Rolle E, De Sanctis R, Forcina N, Norcia G, Passamano L, Gardani A, Pini A, Monaco G, D'Angelo MG, Capasso A, Leone D, Zanin R, Vita GL, Panicucci C, Bruno C, Mongini T, Ricci F, Berardinelli A, Battini R, Masson R, Baranello G, Dosi C, Bertini E, Politano L, Mercuri E. Longitudinal Analysis of PUL 2.0 Domains in Ambulant and Non-Ambulant Duchenne Muscular Dystrophy Patients: How do they Change in Relation to Functional Ability? J Neuromuscul Dis 2023:JND221556. [PMID: 37066919 DOI: 10.3233/jnd-221556] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
BACKGROUND The performance of upper limb 2.0 (PUL) is widely used to assess upper limb function in DMD patients. The aim of the study was to assess 24 month PUL changes in a large cohort of DMD patients and to establish whether domains changes occur more frequently in specific functional subgroups. METHODS The PUL was performed in 311 patients who had at least one pair of assessments at 24 months, for a total of 808 paired assessments. Ambulant patients were subdivided according to the ability to walk: >350, 250-350, ≤250 meters. Non ambulant patients were subdivided according to the time since they lost ambulation: <1, 1-2, 2-5 or >5 years. RESULTS At 12 months, the mean PUL 2.0 change on all the paired assessments was -1.30 (-1.51--1.05) for the total score, -0.5 (-0.66--0.39) for the shoulder domain, -0.6 (-0.74--0.5) for the elbow domain and -0.1 (-0.20--0.06) for the distal domain.At 24 months, the mean PUL 2.0 change on all the paired assessments was -2.9 (-3.29--2.60) for the total score, -1.30 (-1.47--1.09) for the shoulder domain, -1.30 (-1.45--1.11) for the elbow domain and -0.4 (-1.48--1.29) for the distal domain.Changes at 12 and 24 months were statistically significant between subgroups with different functional abilities for the total score and each domain (p < 0.001). CONCLUSION There were different patterns of changes among the functional subgroups in the individual domains. The time of transition, including the year before and after loss of ambulation, show the peak of negative changes in PUL total scores that reflect not only loss of shoulder but also of elbow activities. These results suggest that patterns of changes should be considered at the time of designing clinical trials.
Collapse
Affiliation(s)
- Marika Pane
- Pediatric Neurology, Università Cattolica delSacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Giorgia Coratti
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Claudia Brogna
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Francesca Bovis
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
| | - Adele D'Amico
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children'sHospital, Rome, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Luca Bello
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Valeria Sansone
- The NEMO Clinical Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, Milan, Italy
| | - Emilio Albamonte
- The NEMO Clinical Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, Milan, Italy
| | | | | | - Lavinia Fanelli
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Sonia Messina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Michela Catteruccia
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children'sHospital, Rome, Italy
| | - Gianpaolo Cicala
- Pediatric Neurology, Università Cattolica delSacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Martina Ricci
- Pediatric Neurology, Università Cattolica delSacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Silvia Frosini
- Department of Developmental Neuroscience, IRCCS Stella Maris, Pisa, Italy
| | - Giacomo De Luca
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children'sHospital, Rome, Italy
| | - Enrica Rolle
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Torino, Turin, Italy
| | - Roberto De Sanctis
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Nicola Forcina
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Giulia Norcia
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Luigia Passamano
- Department of Experimental Medicine, Cardiomiology and Medical Genetics, Second University of Naples, Naples, Italy
| | - Alice Gardani
- Child and Adolescence NeurologicalUnit, National Neurological Institute Casimiro MondinoFoundation, IRCCS, Pavia, Italy
| | - Antonella Pini
- Child Neurologyand Psychiatry Unit, IRCCS Institute of Neurological Sciences, Bellaria Hospital, Bologna, Italy
| | - Giulia Monaco
- Child Neurologyand Psychiatry Unit, IRCCS Institute of Neurological Sciences, Bellaria Hospital, Bologna, Italy
| | | | - Anna Capasso
- Pediatric Neurology, Università Cattolica delSacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Daniela Leone
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome
| | - Riccardo Zanin
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Gian Luca Vita
- Unit of Neurology, IRCCS Centro Neurolesi Bonino-Pulejo - P.O. Piemonte, Messina, Italy
| | - Chiara Panicucci
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, and Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and ChildHealth-DINOGMI, University of Genova, Genova, Italy
| | - Claudio Bruno
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, and Department of Neuroscience, Rehabilitation, Ophtalmology, Genetics, Maternal and ChildHealth-DINOGMI, University of Genova, Genova, Italy
| | - Tiziana Mongini
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Torino, Turin, Italy
| | - Federica Ricci
- Neuromuscular Center, AOU Città della Salute e della Scienza, University of Torino, Turin, Italy
| | - Angela Berardinelli
- Child and Adolescence NeurologicalUnit, National Neurological Institute Casimiro MondinoFoundation, IRCCS, Pavia, Italy
| | - Roberta Battini
- Department of Developmental Neuroscience, IRCCS Stella Maris, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Riccardo Masson
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giovanni Baranello
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Claudia Dosi
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children'sHospital, Rome, Italy
| | - Luisa Politano
- Cardiomyology and Medical Genetics Unit, Università degli Studi della CampaniaLuigi Vanvitelli Scuola di Medicina e Chirurgia, Napoli, Italy
| | - Eugenio Mercuri
- Pediatric Neurology, Università Cattolica delSacro Cuore, Rome, Italy
| |
Collapse
|
10
|
Peng F, Xu H, Song Y, Xu K, Li S, Cai X, Guo Y, Gong L. Longitudinal study of multi-parameter quantitative magnetic resonance imaging in Duchenne muscular dystrophy: hyperresponsiveness of gluteus maximus and detection of subclinical disease progression in functionally stable patients. J Neurol 2023; 270:1439-1451. [PMID: 36385201 DOI: 10.1007/s00415-022-11470-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To describe the disease progression of Duchenne muscular dystrophy (DMD) in the pelvic and thigh muscles over 1-year using multiple-parameter quantitative magnetic resonance imaging (qMRI), and to determine the most responsive muscle and predict subclinical disease progression in functionally stable patients. METHODS Fifty-four DMD patients (mean age 8.9 ± 2.5, range 5-15 years) completed baseline and 1-year follow-up qMRI examinations/biomarkers [3-point Dixon/fat fraction (FF); T1 mapping/T1; T2 mapping/T2]. Meanwhile, clinical assessments [NorthStar ambulatory assessment (NSAA) score] and timed function tests were performed in DMD patients. Twenty-four healthy male controls (range 5-15 years) accomplished baseline qMRI examinations. Group differences were compared using the Wilcoxon test. The standardized response mean (SRM) was taken as the responsiveness to the disease progression index. RESULTS FF, T1, and T2 in all DMD age subgroups changed significantly over 1-year (P < 0.05). Even in functionally stable patients (NSAA score increased, unchanged, or decreased by 1-point) over 1-year, significant increases in FF and T2 and decreases in T1 were observed in gluteus maximus (GMa), gluteus medius, vastus lateralis, and adductor magnus (P < 0.05). Overall, the SRM of FF, T1, and T2 was all the highest in GMa, which were 1.25, - 0.92, and 0.93, respectively. CONCLUSIONS qMRI biomarkers are responsive to disease progression and can also detect subclinical disease progression in functionally stable DMD patients over 1-year. GMa is the most responsive to disease progression of all the muscles analyzed. TRIAL REGISTRATION Chinese Clinical Trial Registry ( http://www.chictr.org.cn/index.aspx ) ChiCTR1800018340, 09/12/2018, prospectively registered.
Collapse
Affiliation(s)
- Fei Peng
- Department of Medical Imaging Center, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi Province, China
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Huayan Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Yu Song
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Ke Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China
| | - Shuhao Li
- Department of Medical Imaging Center, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi Province, China
| | - Xiaotang Cai
- Department of Pediatrics Neurology, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China.
| | - Yingkun Guo
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, Sichuan Province, China.
| | - Lianggeng Gong
- Department of Medical Imaging Center, The Second Affiliated Hospital of Nanchang University, Minde Road No. 1, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
11
|
Huang M, Chen T, Wang Y, Zhou C, Cao J, Lu X. Responsiveness and Minimal Clinically Important Difference of the Chinese Version of the Motor Function Measure-32 in Children and Adolescents with Duchenne Muscular Dystrophy. Dev Neurorehabil 2022; 25:370-377. [PMID: 34964685 DOI: 10.1080/17518423.2021.2020352] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
PURPOSE This study aimed to examine the responsiveness and minimal clinically important difference of the Motor Function Measure 32-Chinese version (MFM 32-CN) in children and adolescents with Duchenne muscular dystrophy (DMD). METHODS This observational, prospective cohort study assessed changes in motor function over 12 months using the MFM 32-CN in 108 pediatric patients with DMD. RESULTS Moderate to high internal responsiveness was found for MFM total, D1, and D2 scores (SRM = 0.46-0.83). Sufficient external responsiveness was observed in all MFM scores, as determined by a good correlation with the Patient Global Impression of Change (PGIC) ratings (r = 0.54-0.72, p < .01). The estimated MCID values derived from different anchor-based methods ranged from 4.7 to 6.0, with a median of 5.0 points (%). CONCLUSION The MFM 32-CN demonstrates sufficient internal and external responsiveness as a measure of motor function in children and adolescents with DMD. The present study established the MCID of MFM-32 in pediatric patients with DMD.
Collapse
Affiliation(s)
| | - Turong Chen
- Shenzhen Children's Hospital, Shenzhen, China
| | - Yujuan Wang
- Shenzhen Children's Hospital, Shenzhen, China
| | | | - Jianguo Cao
- Shenzhen Children's Hospital, Shenzhen, China
| | - Xinguo Lu
- Shenzhen Children's Hospital, Shenzhen, China
| |
Collapse
|
12
|
de Souza MA, Martinez EZ, da Silva Lizzi EA, Cezarani A, de Queiroz Davoli GB, Bená MI, da Rosa Sobreira CF, Mattiello-Sverzut AC. Alternative instrument for the evaluation of handgrip strength in Duchenne muscular dystrophy. BMC Pediatr 2022; 22:334. [PMID: 35689212 PMCID: PMC9185969 DOI: 10.1186/s12887-022-03388-x] [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: 04/29/2021] [Accepted: 05/09/2022] [Indexed: 11/10/2022] Open
Abstract
Background The commonly used dynamometers can be ineffective in evaluating handgrip in patients with Duchenne muscular dystrophy (DMD), especially children with generalized muscle weakness. The aim of this study was to analyze whether the modified sphygmomanometer is an effective instrument for handgrip strength evaluation in patients with DMD, during different stages of the disease. Method The handgrip strength of 33 patients was evaluated by the Jamar dynamometer and the modified sphygmomanometer. Motor function was evaluated by the Motor Function Measurement (MFM) scale. Four evaluations, with a six-month interval between each, were performed: Evaluation 1 (N = 33), Evaluation 2 (N = 24), Evaluation 3 (N = 15), and Evaluation 4 (N = 8). A linear regression model with mixed effects was used for the longitudinal data and descriptive analysis of strength for all four evaluations. Result The first evaluation data presented very high correlations between the dynamometer and the modified sphygmomanometer (r = 0.977; p < 0.001). The longitudinal analysis showed a significant difference between Evaluation 1 and the other handgrip strength evaluations obtained using the dynamometer (p < 0.05) but not the modified sphygmomanometer (p > 0.05). Null values were obtained only when using the dynamometer device. Conclusion The modified sphygmomanometer seems to be more suitable than the dynamometer for measuring handgrip strength in all stages of DMD.
Collapse
Affiliation(s)
- Mariana Angélica de Souza
- Department of Health Science, Ribeirao Preto Medical School, University of São Paulo, Av. Bandeirantes, 3900, Campus, Ribeirão Preto, SP, 14049-900, Brazil
| | - Edson Zangiacomi Martinez
- Department of Social Medicine, Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Ananda Cezarani
- Department of Health Science, Ribeirao Preto Medical School, University of São Paulo, Av. Bandeirantes, 3900, Campus, Ribeirão Preto, SP, 14049-900, Brazil
| | - Gabriela Barroso de Queiroz Davoli
- Department of Health Science, Ribeirao Preto Medical School, University of São Paulo, Av. Bandeirantes, 3900, Campus, Ribeirão Preto, SP, 14049-900, Brazil
| | - Marjory Irineu Bená
- Department of Neurosciences of the Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | | | - Ana Claudia Mattiello-Sverzut
- Department of Health Science, Ribeirao Preto Medical School, University of São Paulo, Av. Bandeirantes, 3900, Campus, Ribeirão Preto, SP, 14049-900, Brazil.
| |
Collapse
|
13
|
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: 0.3] [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
|
14
|
Mensch A, Nägel S, Zierz S, Kraya T, Stoevesandt D. Bildgebung der Muskulatur bei Neuromuskulären Erkrankungen
– von der Initialdiagnostik bis zur Verlaufsbeurteilung. KLIN NEUROPHYSIOL 2022. [DOI: 10.1055/a-1738-5356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
ZusammenfassungDie bildgebende Diagnostik hat sich zu einem integralen Element der Betreuung von
PatientInnen mit neuromuskulären Erkrankungen entwickelt. Als
wesentliches Diagnostikum ist hierbei die Magnetresonanztomografie als breit
verfügbares und vergleichsweise standardisiertes Untersuchungsverfahren
etabliert, wobei die Sonografie der Muskulatur bei hinreichend erfahrenem
Untersucher ebenfalls geeignet ist, wertvolle diagnostische Informationen zu
liefern. Das CT hingegen spielt eine untergeordnete Rolle und sollte nur bei
Kontraindikationen für eine MRT in Erwägung gezogen werden.
Zunächst wurde die Bildgebung bei Muskelerkrankungen primär in
der Initialdiagnostik unter vielfältigen Fragestellungen eingesetzt. Das
Aufkommen innovativer Therapiekonzepte bei verschiedenen neuromuskulären
Erkrankungen machen neben einer möglichst frühzeitigen
Diagnosestellung insbesondere auch eine multimodale Verlaufsbeurteilung zur
Evaluation des Therapieansprechens notwendig. Auch hier wird die Bildgebung der
Muskulatur als objektiver Parameter des Therapieerfolges intensiv diskutiert und
in Forschung wie Praxis zunehmend verwendet.
Collapse
Affiliation(s)
- Alexander Mensch
- Universitätsklinik und Poliklinik für Neurologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
| | - Steffen Nägel
- Universitätsklinik und Poliklinik für Neurologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
| | - Stephan Zierz
- Universitätsklinik und Poliklinik für Neurologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
| | - Torsten Kraya
- Universitätsklinik und Poliklinik für Neurologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
- Klinik für Neurologie, Klinikum St. Georg,
Leipzig
| | - Dietrich Stoevesandt
- Universitätsklinik und Poliklinik für Radiologie,
Martin-Luther-Universität Halle-Wittenberg und
Universitätsklinikum Halle, Halle (Saale)
| |
Collapse
|
15
|
Lopez C, Batra A, Moslemi Z, Rennick A, Guice K, Zeng H, Walter GA, Forbes SC. Effects of muscle damage on 31 phosphorus magnetic resonance spectroscopy indices of energetic status and sarcolemma integrity in young mdx mice. NMR IN BIOMEDICINE 2022; 35:e4659. [PMID: 34841594 PMCID: PMC9804208 DOI: 10.1002/nbm.4659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 10/09/2021] [Accepted: 11/09/2021] [Indexed: 06/13/2023]
Abstract
31 Phosphorus magnetic resonance spectroscopy (31 P-MRS) has been shown to detect altered energetic status (e.g. the ratio of inorganic phosphate to phosphocreatine: Pi/PCr), intracellular acid-base status, and free intracellular magnesium ([Mg2+ ]) in dystrophic muscle compared with unaffected muscle; however, the causes of these differences are not well understood. The purposes of this study were to examine 31 P-MRS indices of energetic status and sarcolemma integrity in young mdx mice compared with wild-type and to evaluate the effects of downhill running to induce muscle damage on 31 P-MRS indices in dystrophic muscle. In vivo 31 P-MRS spectra were acquired from the posterior hindlimb muscles in young (4-10 weeks of age) mdx (C57BL/10ScSn-DMDmdx) and wild-type (C57BL/10ScSnJ) mice using an 11.1-T MR system. The flux of phosphate from PCr to ATP was estimated by 31 P-MRS saturation transfer experiments. Relative concentrations of high-energy phosphates were measured, and intracellular pH and [Mg2+ ] were calculated. 1 H2 O-T2 was measured using single-voxel 1 H-MRS from the gastrocnemius and soleus using a 4.7-T MR system. Downhill treadmill running was performed in a subset of mice. Young mdx mice were characterized by elevated 1 H2 O-T2 (p < 0.01), Pi/PCr (p = 0.02), PCr to ATP flux (p = 0.04) and histological inflammatory markers (p < 0.05) and reduced (p < 0.01) [Mg2+ ] compared with wild-type. Furthermore, 24 h after downhill running, an increase (p = 0.02) in Pi/PCr was observed in mdx and wild-type mice compared with baseline, and a decrease (p < 0.001) in [Mg2+ ] and a lower (p = 0.048) intracellular [H+ ] in damaged muscle regions of mdx mice were observed, consistent with impaired sarcolemma integrity. Overall, our findings demonstrate that 31 P-MRS markers of energetic status and sarcolemma integrity are altered in young mdx compared with wild-type mice, and these indices are exacerbated following downhill running.
Collapse
Affiliation(s)
- Christopher Lopez
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Abhinandan Batra
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Zahra Moslemi
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Andrew Rennick
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Kimberly Guice
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Huadong Zeng
- Advanced Magnetic Resonance Imaging and Spectroscopy Facility, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Glenn A. Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| | - Sean C. Forbes
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
16
|
Szabo SM, Gooch KL, Mickle AT, Salhany RM, Connolly AM. The impact of genotype on outcomes in individuals with Duchenne muscular dystrophy: A systematic review. Muscle Nerve 2021; 65:266-277. [PMID: 34878187 DOI: 10.1002/mus.27463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 11/07/2022]
Abstract
Duchenne muscular dystrophy (DMD) is associated with progressive muscle weakness, loss of ambulation (LOA), and early mortality. In this review we have synthesized published data on the clinical course of DMD by genotype. Using a systematic search implemented in Medline and Embase, 53 articles were identified that describe the clinical course of DMD, with pathogenic variants categorizable by exon skip or stop-codon readthrough amenability and outcomes presented by age. Outcomes described included those related to ambulatory, cardiac, pulmonary, or cognitive function. Estimates of the mean (95% confidence interval) age at LOA ranged from 9.1 (8.7-9.6) years among 90 patients amenable to skipping exon 53 to 11.5 (9.5-13.5) years among three patients amenable to skipping exon 8. Although function worsened with age, the impact of genotype was less clear for other outcomes (eg, forced vital capacity and left ventricular ejection fraction). Understanding the distribution of pathogenic variants is important for studies in DMD, as this research suggests major differences in the natural history of disease. In addition, specific details of the use of key medications, including corticosteroids, antisense oligonucleotides, and cardiac medications, should be reported.
Collapse
Affiliation(s)
- Shelagh M Szabo
- Broadstreet Heath Economics & Outcomes Research, Vancouver, British Columbia, Canada
| | | | - Alexis T Mickle
- Broadstreet Heath Economics & Outcomes Research, Vancouver, British Columbia, Canada
| | | | - Anne M Connolly
- Division of Neurology, Nationwide Children's Hospital, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
17
|
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: 1.0] [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
|
18
|
Markati T, De Waele L, Schara-Schmidt U, Servais L. Lessons Learned from Discontinued Clinical Developments in Duchenne Muscular Dystrophy. Front Pharmacol 2021; 12:735912. [PMID: 34790118 PMCID: PMC8591262 DOI: 10.3389/fphar.2021.735912] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 10/12/2021] [Indexed: 02/04/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked condition caused by a deficiency of functional dystrophin protein. Patients experience progressive muscle weakness, cardiomyopathy and have a decreased life expectancy. Standards of care, including treatment with steroids, and multidisciplinary approaches have extended the life expectancy and improved the quality of life of patients. In the last 30 years, several compounds have been assessed in preclinical and clinical studies for their ability to restore functional dystrophin levels or to modify pathways involved in DMD pathophysiology. However, there is still an unmet need with regards to a disease-modifying treatment for DMD and the attrition rate between early-phase and late-phase clinical development remains high. Currently, there are 40 compounds in clinical development for DMD, including gene therapy and antisense oligonucleotides for exon skipping. Only five of them have received conditional approval in one jurisdiction subject to further proof of efficacy. In this review, we present data of another 16 compounds that failed to complete clinical development, despite positive results in early phases of development in some cases. We examine the reasons for the high attrition rate and we suggest solutions to avoid similar mistakes in the future.
Collapse
Affiliation(s)
- Theodora Markati
- MDUK Oxford Neuromuscular Center, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Liesbeth De Waele
- KU Leuven Department of Development and Regeneration, Leuven, Belgium
- Department of Paediatric Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Urlike Schara-Schmidt
- Department of Pediatric Neurology, Center for Neuromuscular Diseases, Center for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| | - Laurent Servais
- MDUK Oxford Neuromuscular Center, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Division of Child Neurology, Reference Center for Neuromuscular Disease, Centre Hospitalier Régional de Références des Maladies Neuromusculaires, Department of Pediatrics, University Hospital Liège, Liège, Belgium
| |
Collapse
|
19
|
Davoli GBDQ, Cardoso J, Silva GC, Moreira RDFC, Mattiello-Sverzut AC. Instruments to assess upper-limb function in children and adolescents with neuromuscular diseases: a systematic review. Dev Med Child Neurol 2021; 63:1030-1037. [PMID: 33834485 DOI: 10.1111/dmcn.14887] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/06/2021] [Indexed: 11/29/2022]
Abstract
AIM To synthesize clinical and scientific evidence regarding the instruments available to assess upper-limb function in paediatric patients with neuromuscular disease (NMD). METHOD This systematic review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses and COnsensus-based Standards for the selection of health Measurement INstruments (COSMIN) guidelines (Prospective Registry of Systematic Reviews no. CRD42020140343). Two independent reviewers searched the PubMed/MEDLINE, LILACS, Embase, and Scopus databases. Inclusion criteria were cross-sectional or longitudinal studies or randomized controlled trials that used scales or questionnaires to assess upper-limb function in paediatric patients with NMDs. The COSMIN Risk of Bias checklist and criteria for good measurement properties were applied to assess the methodological quality of the instruments. RESULTS In total, 34 articles and 12 instruments were included. The Brooke Upper Extremity (n=16) and Performance of Upper Limb (PUL) (n=12) instruments were the most used tools. The PUL and Duchenne muscular dystrophy (DMD) Upper Limb patient-reported outcome measures (PROMs) tested more measurement properties and provided higher methodological quality scores for patients with DMD. Likewise, the Revised Upper Limb Module (RULM) was the most suitable instrument for patients with spinal muscular atrophy. No instrument has been devised to assess upper-limb function in patients with Charcot-Marie-Tooth disease and no other disease-specific instruments were found. INTERPRETATION The PUL, DMD Upper Limb PROM, and RULM are the most suitable instruments to assess upper-limb function in the two most prevalent paediatric NMDs. The identified gaps and methodological flaws of the available instruments indicate a need to develop high-quality instruments to assess other types of paediatric NMDs. What this paper adds The most suitable observer-rater instrument to assess upper-limb function in Duchenne muscular dystrophy (DMD) is the Performance of Upper Limb. The most suitable observer-rater instrument to assess upper-limb function in spinal muscular atrophy is the Revised Upper Limb Module. The DMD Upper Limb patient-reported outcome measure is recommended to assess the upper-limb performance of patients with DMD. Literature gaps and methodological flaws indicate the need to develop high-quality instruments to assess other types of paediatric neuromuscular disease.
Collapse
Affiliation(s)
| | - Juliana Cardoso
- Department of Health Science, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Giovanna Constantin Silva
- Department of Health Science, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | | |
Collapse
|
20
|
Lilien C, Reyngoudt H, Seferian AM, Gidaro T, Annoussamy M, Chê V, Decostre V, Ledoux I, Le Louër J, Guemas E, Muntoni F, Hogrel JY, Carlier PG, Servais L. Upper limb disease evolution in exon 53 skipping eligible patients with Duchenne muscular dystrophy. Ann Clin Transl Neurol 2021; 8:1938-1950. [PMID: 34453498 PMCID: PMC8528463 DOI: 10.1002/acn3.51417] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/24/2021] [Accepted: 06/07/2021] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE To understand the natural disease upper limb progression over 3 years of ambulatory and non-ambulatory patients with Duchenne muscular dystrophy (DMD) using functional assessments and quantitative magnetic resonance imaging (MRI) and to exploratively identify prognostic factors. METHODS Forty boys with DMD (22 non-ambulatory and 18 ambulatory) with deletions in dystrophin that make them eligible for exon 53-skipping therapy were included. Clinical assessments, including Brooke score, motor function measure (MFM), hand grip and key pinch strength, and upper limb distal coordination and endurance (MoviPlate), were performed every 6 months and quantitative MRI of fat fraction (FF) and lean muscle cross sectional area (flexor and extensor muscles) were performed yearly. RESULTS In the whole population, there were strong nonlinear correlations between outcome measures. In non-ambulatory patients, annual changes over the course of 3 years were detected with high sensitivity standard response mean (|SRM| ≥0.8) for quantitative MRI-based FF, hand grip and key pinch, and MFM. Boys who presented with a FF<20% and a grip strength >27% were able to bring a glass to their mouth and retained this ability in the following 3 years. Ambulatory patients with grip strength >35% of predicted value and FF <10% retained ambulation 3 years later. INTERPRETATION We demonstrate that continuous decline in upper limb strength, function, and MRI measured muscle structure can be reliably measured in ambulatory and non-ambulatory boys with DMD with high SRM and strong correlations between outcomes. Our results suggest that a combination of grip strength and FF can be used to predict important motor milestones.
Collapse
Affiliation(s)
- Charlotte Lilien
- Institut de Myologie, Paris, France.,Department of Paediatrics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, United Kingdom
| | - Harmen Reyngoudt
- Institut de Myologie, Paris, France.,CEA/DRF/IBFJ/MIRCen, Paris, France
| | | | | | | | | | | | | | - Julien Le Louër
- Institut de Myologie, Paris, France.,CEA/DRF/IBFJ/MIRCen, Paris, France
| | | | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London, United Kingdom.,National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom
| | | | - Pierre Georges Carlier
- Institut de Myologie, Paris, France.,Université Paris-Saclay, CEA, DRF, Service Hospitalier Frederic Joliot, Orsay, France
| | - Laurent Servais
- Institut de Myologie, Paris, France.,Department of Paediatrics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, United Kingdom.,Division of Child Neurology Reference Center for Neuromuscular Disease, Centre Hospitalier Régional de Références des Maladies Neuromusculaires, Department of Paediatrics, University Hospital Liège & University of La Citadelle, Liège, Belgium
| | | |
Collapse
|
21
|
Marty B, Reyngoudt H, Boisserie JM, Le Louër J, C A Araujo E, Fromes Y, Carlier PG. Water-Fat Separation in MR Fingerprinting for Quantitative Monitoring of the Skeletal Muscle in Neuromuscular Disorders. Radiology 2021; 300:652-660. [PMID: 34254855 DOI: 10.1148/radiol.2021204028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Background Quantitative MRI is increasingly proposed in clinical trials related to neuromuscular disorders (NMDs). Purpose To investigate the potential of an MR fingerprinting sequence for water and fat fraction (FF) quantification (MRF T1-FF) for providing markers of fatty replacement and disease activity in patients with NMDs and to establish the sensitivity of water T1 as a marker of disease activity compared with water T2 mapping. Materials and Methods Data acquired between March 2018 and March 2020 from the legs of patients with NMDs were retrospectively analyzed. The MRI examination comprised fat-suppressed T2-weighted imaging, mapping of the FF measured with the three-point Dixon technique (FFDixon), water T2 mapping, and MRF T1-FF, from which the FF measured with MRF T1-FF (FFMRF) and water T1 were derived. Data from the legs of healthy volunteers were prospectively acquired between January and July 2020 to derive abnormality thresholds for FF, water T2, and water T1 values. Kruskal-Wallis tests and receiver operating characteristic curve analysis were performed, and linear models were used. Results A total of 73 patients (mean age ± standard deviation, 47 years ± 12; 45 women) and 15 healthy volunteers (mean age, 33 years ± 8; three women) were evaluated. A linear correlation was observed between FFMRF and FFDixon (R2 = 0.97, P < .001). Water T1 values were higher in muscles with high signal intensity at fat-suppressed T2-weighted imaging than in muscles with low signal intensity (mean value, 1281 msec [95% CI: 1165, 1604] vs 1198 msec [95% CI: 1099, 1312], respectively; P < .001), and a correlation was found between water T1 and water T2 distribution metrics (R2 = 0.66 and 0.79 for the median and 90th percentile values, respectively; P < .001). Water T1 classified the patients' muscles as abnormal based on quantitative water T2, with high sensitivity (93%; 68 of 73 patients) and specificity (80%; 53 of 73 patients) (area under the receiver operating characteristic curve, 0.92 [95% CI: 0.83, 0.97]; P < .001). Conclusion Water-fat separation in MR fingerprinting is robust for deriving quantitative imaging markers of intramuscular fatty replacement and disease activity in patients with neuromuscular disorders. © RSNA, 2021 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Benjamin Marty
- From the Nuclear Magnetic Resonance Laboratory, Neuromuscular Investigation Center, Institute of Myology, Bâtiment Babinski, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Blvd Vincent Auriol, 75651 Paris Cedex 13, France; and Nuclear Magnetic Resonance Laboratory, CEA, DRF, IBFJ, Molecular Imaging Research Center, Paris, France
| | - Harmen Reyngoudt
- From the Nuclear Magnetic Resonance Laboratory, Neuromuscular Investigation Center, Institute of Myology, Bâtiment Babinski, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Blvd Vincent Auriol, 75651 Paris Cedex 13, France; and Nuclear Magnetic Resonance Laboratory, CEA, DRF, IBFJ, Molecular Imaging Research Center, Paris, France
| | - Jean-Marc Boisserie
- From the Nuclear Magnetic Resonance Laboratory, Neuromuscular Investigation Center, Institute of Myology, Bâtiment Babinski, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Blvd Vincent Auriol, 75651 Paris Cedex 13, France; and Nuclear Magnetic Resonance Laboratory, CEA, DRF, IBFJ, Molecular Imaging Research Center, Paris, France
| | - Julien Le Louër
- From the Nuclear Magnetic Resonance Laboratory, Neuromuscular Investigation Center, Institute of Myology, Bâtiment Babinski, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Blvd Vincent Auriol, 75651 Paris Cedex 13, France; and Nuclear Magnetic Resonance Laboratory, CEA, DRF, IBFJ, Molecular Imaging Research Center, Paris, France
| | - Ericky C A Araujo
- From the Nuclear Magnetic Resonance Laboratory, Neuromuscular Investigation Center, Institute of Myology, Bâtiment Babinski, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Blvd Vincent Auriol, 75651 Paris Cedex 13, France; and Nuclear Magnetic Resonance Laboratory, CEA, DRF, IBFJ, Molecular Imaging Research Center, Paris, France
| | - Yves Fromes
- From the Nuclear Magnetic Resonance Laboratory, Neuromuscular Investigation Center, Institute of Myology, Bâtiment Babinski, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Blvd Vincent Auriol, 75651 Paris Cedex 13, France; and Nuclear Magnetic Resonance Laboratory, CEA, DRF, IBFJ, Molecular Imaging Research Center, Paris, France
| | - Pierre G Carlier
- From the Nuclear Magnetic Resonance Laboratory, Neuromuscular Investigation Center, Institute of Myology, Bâtiment Babinski, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Blvd Vincent Auriol, 75651 Paris Cedex 13, France; and Nuclear Magnetic Resonance Laboratory, CEA, DRF, IBFJ, Molecular Imaging Research Center, Paris, France
| |
Collapse
|
22
|
Naarding KJ, Keene KR, Sardjoe Mishre ASD, Veeger TTJ, van de Velde NM, Prins AJ, Burakiewicz J, Verschuuren JJGM, van der Holst M, Niks EH, Kan HE. Preserved thenar muscles in non-ambulant Duchenne muscular dystrophy patients. J Cachexia Sarcopenia Muscle 2021; 12:694-703. [PMID: 33963807 PMCID: PMC8200430 DOI: 10.1002/jcsm.12711] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/10/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Clinical trials in Duchenne muscular dystrophy (DMD) focus primarily on ambulant patients. Results cannot be extrapolated to later disease stages due to a decline in targeted muscle tissue. In non-ambulant DMD patients, hand function is relatively preserved and crucial for daily-life activities. We used quantitative MRI (qMRI) to establish whether the thenar muscles could be valuable to monitor treatment effects in non-ambulant DMD patients. METHODS Seventeen non-ambulant DMD patients (range 10.2-24.1 years) and 13 healthy controls (range 9.5-25.4 years) underwent qMRI of the right hand at 3 T at baseline. Thenar fat fraction (FF), total volume (TV), and contractile volume (CV) were determined using 4-point Dixon, and T2water was determined using multiecho spin-echo. Clinical assessments at baseline (n = 17) and 12 months (n = 13) included pinch strength (kg), performance of the upper limb (PUL) 2.0, DMD upper limb patient reported outcome measure (PROM), and playing a video game for 10 min using a game controller. Group differences and correlations were assessed with non-parametric tests. RESULTS Total volume was lower in patients compared with healthy controls (6.9 cm3 , 5.3-9.0 cm3 vs. 13.0 cm3 , 7.6-15.8 cm3 , P = 0.010). CV was also lower in patients (6.3 cm3 , 4.6-8.3 cm3 vs. 11.9 cm3 , 6.9-14.6 cm3 , P = 0.010). FF was slightly elevated (9.7%, 7.3-11.4% vs. 7.7%, 6.6-8.4%, P = 0.043), while T2water was higher (31.5 ms, 30.0-32.6 ms vs. 28.1 ms, 27.8-29.4 ms, P < 0.001). Pinch strength and PUL decreased over 12 months (2.857 kg, 2.137-4.010 to 2.243 kg, 1.930-3.339 kg, and 29 points, 20-36 to 23 points, 17-30, both P < 0.001), while PROM did not (49 points, 36-57 to 44 points, 30-54, P = 0.041). All patients were able to play for 10 min at baseline or follow-up, but some did not comply with the study procedures regarding this endpoint. Pinch strength correlated with TV and CV in patients (rho = 0.72 and rho = 0.68) and controls (both rho = 0.89). PUL correlated with TV, CV, and T2water (rho = 0.57, rho = 0.51, and rho = -0.59). CONCLUSIONS Low thenar FF, increased T2water , correlation of muscle size with strength and function, and the decrease in strength and function over 1 year indicate that the thenar muscles are a valuable and quantifiable target for therapy in later stages of DMD. Further studies are needed to relate these data to the loss of a clinically meaningful milestone.
Collapse
Affiliation(s)
- Karin J Naarding
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Kevin R Keene
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands
| | | | - Thom T J Veeger
- C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands
| | - Nienke M van de Velde
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Arina J Prins
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Jedrzej Burakiewicz
- C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands
| | - Jan J G M Verschuuren
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Menno van der Holst
- Duchenne Center, Leiden, Netherlands.,Department of Orthopedics, Rehabilitation and Physiotherapy, Leiden University Medical Center, Leiden, Netherlands
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Hermien E Kan
- Duchenne Center, Leiden, Netherlands.,C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands
| |
Collapse
|
23
|
The increasing role of muscle MRI to monitor changes over time in untreated and treated muscle diseases. Curr Opin Neurol 2021; 33:611-620. [PMID: 32796278 DOI: 10.1097/wco.0000000000000851] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW This review aims to discuss the recent results of studies published applying quantitative MRI sequences to large cohorts of patients with neuromuscular diseases. RECENT FINDINGS Quantitative MRI sequences are now available to identify and quantify changes in muscle water and fat content. These two components have been associated with acute and chronic injuries, respectively. Studies show that the increase in muscle water is not only reversible if therapies are applied successfully but can also predict fat replacement in neurodegenerative diseases. Muscle fat fraction correlates with muscle function tests and increases gradually over time in parallel with the functional decline of patients with neuromuscular diseases. There are new spectrometry-based sequences to quantify other components, such as glycogen, electrolytes or the pH of the muscle fibre, extending the applicability of MRI to the study of several processes in neuromuscular diseases. SUMMARY The latest results obtained from the study of long cohorts of patients with various neuromuscular diseases open the door to the use of this technology in clinical trials, which would make it possible to obtain a new measure for assessing the effectiveness of new treatments. The challenge is currently the popularization of these studies and their application to the monitoring of patients in the daily clinic.
Collapse
|
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: 2.3] [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
|
Annoussamy M, Seferian AM, Daron A, Péréon Y, Cances C, Vuillerot C, De Waele L, Laugel V, Schara U, Gidaro T, Lilien C, Hogrel JY, Carlier P, Fournier E, Lowes L, Gorni K, Ly-Le Moal M, Hellbach N, Seabrook T, Czech C, Hermosilla R, Servais L. Natural history of Type 2 and 3 spinal muscular atrophy: 2-year NatHis-SMA study. Ann Clin Transl Neurol 2020; 8:359-373. [PMID: 33369268 PMCID: PMC7886049 DOI: 10.1002/acn3.51281] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 12/19/2022] Open
Abstract
Objective To characterize the natural history of spinal muscular atrophy (SMA) over 24 months using innovative measures such as wearable devices, and to provide evidence for the sensitivity of these measures to determine their suitability as endpoints in clinical trials. Methods Patients with Type 2 and 3 SMA (N = 81) with varied functional abilities (sitters, nonsitters, nonambulant, and ambulant) who were not receiving disease‐modifying treatment were assessed over 24 months: motor function (Motor Function Measure [MFM]), upper limb strength (MyoGrip, MyoPinch), upper limb activity (ActiMyo®), quantitative magnetic resonance imaging (fat fraction [FFT2] mapping and contractile cross‐sectional area [C‐CSA]), pulmonary function (forced vital capacity [FVC], peak cough flow, maximum expiratory pressure, maximum inspiratory pressure, and sniff nasal inspiratory pressure), and survival of motor neuron (SMN) protein levels. Results MFM32 scores declined significantly over 24 months, but not 12 months. Changes in upper limb activity could be detected over 6 months and continued to decrease significantly over 12 months, but not 24 months. Upper limb strength decreased significantly over 12 and 24 months. FVC declined significantly over 12 months, but not 24 months. FFT2 increased over 12 and 24 months, although not with statistical significance. A significant increase in C‐CSA was observed at 12 but not 24 months. Blood SMN protein levels were stable over 12 and 24 months. Interpretation These data demonstrate that the MFM32, MyoGrip, MyoPinch, and ActiMyo® enable the detection of a significant decline in patients with Type 2 and 3 SMA over 12 or 24 months.
Collapse
Affiliation(s)
- Mélanie Annoussamy
- Institute of Myology, GH Pitié Salpêtrière, Paris, France.,Sysnav, Vernon, France
| | | | - Aurore Daron
- Centre de Référence des Maladies Neuromusculaires, CHU de Liège, Liege, Belgium
| | - Yann Péréon
- Centre de Référence Maladies Neuromusculaires Atlantique-Occitanie-Caraïbes, Hôpital Hôtel-Dieu, Nantes, France
| | - Claude Cances
- Centre de Référence des Maladies, Neuromusculaires, Hôpital des Enfants, Toulouse, France.,Unité de Neurologie Pédiatrique, Hôpital des Enfants, Toulouse, France
| | - Carole Vuillerot
- Service de rééducation pédiatrique infantile L'Escale, Hôpital Mère Enfant, CHU-Lyon, Bron, France.,Neuromyogen Institute, CNRS, UMR 5310 INSERM U1217, Université de Lyon, Lyon, France
| | - Liesbeth De Waele
- Department of Pediatric Neurology, University Hospitals Leuven, Leuven, Belgium.,Department of Development and Regeneration, KU Leuven Kulak Kortrijk, Kortrijk, Belgium
| | - Vincent Laugel
- Neuropédiatrie, INSERM CIC 1434, CHU Strasbourg Hautepierre, Strasbourg, France
| | - Ulrike Schara
- Paediatric neurology and Neuromuscular Center, University of Essen, Essen, Germany
| | - Teresa Gidaro
- Institute of Myology, GH Pitié Salpêtrière, Paris, France
| | - Charlotte Lilien
- Institute of Myology, GH Pitié Salpêtrière, Paris, France.,Department of Paediatrics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK
| | | | - Pierre Carlier
- Institute of Myology, GH Pitié Salpêtrière, Paris, France
| | | | - Linda Lowes
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Ksenija Gorni
- PDMA Neuroscience and Rare Disease, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | | | - Nicole Hellbach
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Timothy Seabrook
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Christian Czech
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland.,Rare Disease Research Unit, Pfizer, Nice, France
| | - Ricardo Hermosilla
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - Laurent Servais
- Institute of Myology, GH Pitié Salpêtrière, Paris, France.,Department of Paediatrics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK.,Division of Child Neurology, Centre de Références des Maladies Neuromusculaires, Department of Pediatrics, University Hospital Liège & University of Liège, Liège, Belgium
| | | |
Collapse
|
26
|
Hooijmans MT, Froeling M, Koeks Z, Verschuuren JJ, Webb A, Niks EH, Kan HE. Multi-parametric MR in Becker muscular dystrophy patients. NMR IN BIOMEDICINE 2020; 33:e4385. [PMID: 32754921 PMCID: PMC7687231 DOI: 10.1002/nbm.4385] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 05/14/2023]
Abstract
Quantitative MRI and MRS of muscle are increasingly being used to measure individual pathophysiological processes in Becker muscular dystrophy (BMD). In particular, muscle fat fraction was shown to be highly associated with functional tests in BMD. However, the muscle strength per unit of contractile cross-sectional area is lower in patients with BMD compared with healthy controls. This suggests that the quality of the non-fat-replaced (NFR) muscle tissue is lower than in healthy controls. Consequently, a measure that reflects changes in muscle tissue itself is needed. Here, we explore the potential of water T2 relaxation times, diffusion parameters and phosphorus metabolic indices as early disease markers in patients with BMD. For this purpose, we examined these measures in fat-replaced (FR) and NFR lower leg muscles in patients with BMD and compared these values with those in healthy controls. Quantitative proton MRI (three-point Dixon, multi-spin-echo and diffusion-weighted spin-echo echo planar imaging) and 2D chemical shift imaging 31 P MRS data were acquired in 24 patients with BMD (age 18.8-66.2 years) and 13 healthy controls (age 21.3-63.6 years). Muscle fat fractions, phosphorus metabolic indices, and averages and standard deviations (SDs) of the water T2 relaxation times and diffusion tensor imaging (DTI) parameters were assessed in six individual leg muscles. Phosphodiester levels were increased in the NFR and FR tibialis anterior, FR peroneus and FR gastrocnemius lateralis muscles. No clear pattern was visible for the other metabolic indices. Increased T2 SD was found in the majority of FR muscles compared with NFR and healthy control muscles. No differences in average water T2 relaxation times or DTI indices were found between groups. Overall, our results indicate that primarily muscles that are further along in the disease process showed increases in T2 heterogeneity and changes in some metabolic indices. No clear differences were found for the DTI indices between groups.
Collapse
Affiliation(s)
- Melissa T. Hooijmans
- C.J. Gorter Center, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
- Department of Biomedical Engineering & PhysicsAmsterdam University Medical CentersAmsterdamThe Netherlands
| | - Martijn Froeling
- Department of RadiologyUtrecht University Medical CenterUtrechtThe Netherlands
| | - Zaida Koeks
- Department of NeurologyLeiden University Medical CenterLeidenThe Netherlands
| | - Jan J.G.M. Verschuuren
- Department of NeurologyLeiden University Medical CenterLeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| | - Andrew Webb
- C.J. Gorter Center, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Erik H. Niks
- Department of NeurologyLeiden University Medical CenterLeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| | - Hermien E. Kan
- C.J. Gorter Center, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| |
Collapse
|
27
|
Dahlqvist JR, Widholm P, Leinhard OD, Vissing J. MRI in Neuromuscular Diseases: An Emerging Diagnostic Tool and Biomarker for Prognosis and Efficacy. Ann Neurol 2020; 88:669-681. [PMID: 32495452 DOI: 10.1002/ana.25804] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 05/05/2020] [Accepted: 05/25/2020] [Indexed: 12/12/2022]
Abstract
There is an unmet need to identify biomarkers sensitive to change in rare, slowly progressive neuromuscular diseases. Quantitative magnetic resonance imaging (MRI) of muscle may offer this opportunity, as it is noninvasive and can be carried out almost independent of patient cooperation and disease severity. Muscle fat content correlates with muscle function in neuromuscular diseases, and changes in fat content precede changes in function, which suggests that muscle MRI is a strong biomarker candidate to predict prognosis and treatment efficacy. In this paper, we review the evidence suggesting that muscle MRI may be an important biomarker for diagnosis and to monitor change in disease severity. ANN NEUROL 2020;88:669-681.
Collapse
Affiliation(s)
- Julia R Dahlqvist
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | - Per Widholm
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
- AMRA Medical AB, Linköping, Sweden
| | - Olof Dahlqvist Leinhard
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
- AMRA Medical AB, Linköping, Sweden
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
28
|
Neuhaus SB, Wallgren-Pettersson C, Bönnemann CG, Schara U, Servais L. 250th ENMC International Workshop: Clinical trial readiness in nemaline myopathy 6-8 September 2019, Hoofdorp, the Netherlands. Neuromuscul Disord 2020; 30:866-875. [PMID: 32919842 DOI: 10.1016/j.nmd.2020.08.356] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Sarah B Neuhaus
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, U.S.A
| | - Carina Wallgren-Pettersson
- The Folkhälsan Institute of Genetics and the Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, 20892, U.S.A
| | - Ulrike Schara
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders in Children and Adolecents, University Hospital Essen, University of Duisburg-Essen, Germany
| | - Laurent Servais
- Division of Child Neurology, Centre de Références des Maladies Neuromusculaires, Department of Pediatrics, University Hospital Liège & University of Liège, Liège, Belgium; MDUK Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford, United Kingdom.
| | | |
Collapse
|
29
|
Marty B, Lopez Kolkovsky AL, Araujo ECA, Reyngoudt H. Quantitative Skeletal Muscle Imaging Using 3D MR Fingerprinting With Water and Fat Separation. J Magn Reson Imaging 2020; 53:1529-1538. [PMID: 32996670 DOI: 10.1002/jmri.27381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Quantitative muscle MRI is a robust tool to monitor intramuscular fatty replacement and disease activity in patients with neuromuscular disorders (NMDs). PURPOSE To implement a 3D sequence for quantifying simultaneously fat fraction (FF) and water T1 (T1,H2O ) in the skeletal muscle, evaluate regular undersampling in the partition-encoding direction, and compare it to a recently proposed 2D MR fingerprinting sequence with water and fat separation (MRF T1 -FF). STUDY TYPE Prospective. PHANTOM/SUBJECTS Seventeen-vial phantom at different FF and T1,H2O , 11 healthy volunteers, and 6 subjects with different NMDs. FIELD STRENGTH/SEQUENCE 3T/3D MRF T1 -FF, 2D MRF T1 -FF, STEAM MRS ASSESSMENT: FF and T1,H2O measured with the 2D and 3D sequences were compared in the phantom and in vivo at different undersampling factors (US). Data were acquired in healthy subjects before and after plantar dorsiflexions and at rest in patients. STATISTICAL TESTS Linear correlations, Bland-Altman analysis, two-way repeated measures analysis of variance (ANOVA), Student's t-test. RESULTS Up to a US factor of 3, the undersampled acquisitions were in good agreement with the fully sampled sequence (R2 ≥ 0.98, T1,H2O bias ≤10 msec, FF bias ≤4 × 10-4 ) both in phantom and in vivo. The 2D and 3D MRF T1 -FF sequences provided comparable T1,H2O and FF values (R2 ≥ 0.95, absolute T1,H2O bias ≤35 msec, and absolute FF bias ≤0.003). The plantar dorsiflexion induced a significant increase of T1,H2O in the tibialis anterior and extensor digitorum (relative increase of +10.8 ± 1.7% and + 7.7 ± 1.4%, respectively, P < 0.05), that was accompanied by a significant reduction of FF in the tibialis anterior (relative decrease of -16.3 ± 4.0%, P < 0.05). Some subjects with NMDs presented increased and heterogeneous T1,H2O and FF values throughout the leg. DATA CONCLUSION Quantitative 3D T1,H2O and FF maps covering the entire leg were obtained within acquisition times compatible with clinical research (4 minutes 20 seconds) and a 1 × 1 × 5 mm3 spatial resolution. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Benjamin Marty
- Neuromuscular Investigation Center, NMR Laboratory, Institute of Myology, Paris, France.,CEA, DRF, IBFJ, MIRCen, NMR Laboratory, Paris, France
| | - Alfredo L Lopez Kolkovsky
- Neuromuscular Investigation Center, NMR Laboratory, Institute of Myology, Paris, France.,CEA, DRF, IBFJ, MIRCen, NMR Laboratory, Paris, France
| | - Ericky C A Araujo
- Neuromuscular Investigation Center, NMR Laboratory, Institute of Myology, Paris, France.,CEA, DRF, IBFJ, MIRCen, NMR Laboratory, Paris, France
| | - Harmen Reyngoudt
- Neuromuscular Investigation Center, NMR Laboratory, Institute of Myology, Paris, France.,CEA, DRF, IBFJ, MIRCen, NMR Laboratory, Paris, France
| |
Collapse
|
30
|
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: 0.8] [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
|
31
|
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: 28] [Impact Index Per Article: 5.6] [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
|
32
|
Reyngoudt H, Marty B, Caldas de Almeida Araújo E, Baudin PY, Le Louër J, Boisserie JM, Béhin A, Servais L, Gidaro T, Carlier PG. Relationship between markers of disease activity and progression in skeletal muscle of GNE myopathy patients using quantitative nuclear magnetic resonance imaging and 31P nuclear magnetic resonance spectroscopy. Quant Imaging Med Surg 2020; 10:1450-1464. [PMID: 32676364 DOI: 10.21037/qims-20-39] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Quantitative nuclear magnetic resonance imaging (NMRI) is an objective and precise outcome measure for evaluating disease progression in neuromuscular disorders. We aimed to investigate predictive 'disease activity' NMR indices, including water T2 and 31P NMR spectroscopy (NMRS), and its relation to NMR markers of 'disease progression', such as the changes in fat fraction (ΔFat%) and contractile cross-sectional area (ΔcCSA), in GNE myopathy (GNEM) patients. Methods NMR was performed on a 3T clinical scanner, at baseline and at a 1-year interval, in 10 GNEM patients and 29 age-matched controls. Dixon-based fat-water imaging and water T2 mapping were acquired in legs and thighs, and in the dominant forearm. 31P NMRS was performed at the level of quadriceps and hamstring. Water T2 and 31P NMRS indices were determined for all muscle groups and visits. Correlations were performed with 'disease progression' indices ΔFat%, ΔcCSA and the muscle fat transformation rate (Rmuscle_transf). Results In quadriceps, known to be relatively preserved in GNEM, water T2 at baseline was significantly higher compared to controls, and correlated strongly with the one-year evolution of Fat% and cCSA and Rmuscle_transf. Various 31P NMRS indices showed significant differences in quadriceps and hamstring compared to controls and correlations existed between these indices and ΔFat%, ΔcCSA and Rmuscle_transf. Conclusions This study demonstrates that disease activity indices such as water T2 and 31P NMRS may predict disease progression in skeletal muscles of GNEM patients, and suggests that these measures may be considered to be valuable surrogate endpoints in the assessment of GNEM disease progression.
Collapse
Affiliation(s)
- Harmen Reyngoudt
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA, DRF, IBFJ, MIRCen, Paris, France
| | - Benjamin Marty
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA, DRF, IBFJ, MIRCen, Paris, France
| | - Ericky Caldas de Almeida Araújo
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA, DRF, IBFJ, MIRCen, Paris, France
| | - Pierre-Yves Baudin
- Consultants for Research in Imaging and Spectroscopy (C.R.I.S.), Tournai, Belgium
| | - Julien Le Louër
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA, DRF, IBFJ, MIRCen, Paris, France
| | - Jean-Marc Boisserie
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA, DRF, IBFJ, MIRCen, Paris, France
| | - Anthony Béhin
- Neuromuscular Reference Center, Institute of Myology, Pitié-Salpêtrière Hospital (AP-HP), Paris, France
| | - Laurent Servais
- Institute of Myology, Pitié-Salpêtrière Hospital (AP-HP), Paris, France.,I-Motion-Pediatric Clinical Trials Department, Trousseau Hospital (AP-HP), Paris, France.,Centre de référence des maladies Neuromusculaires, CHU, University of Liège, Liège, Belgium.,MDUK Oxford Neuromuscular Center, Department of Pediatrics, University of Oxford, Oxford, UK
| | - Teresa Gidaro
- Institute of Myology, Pitié-Salpêtrière Hospital (AP-HP), Paris, France.,I-Motion-Pediatric Clinical Trials Department, Trousseau Hospital (AP-HP), Paris, France
| | - Pierre G Carlier
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA, DRF, IBFJ, MIRCen, Paris, France
| |
Collapse
|
33
|
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: 4.8] [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
|
34
|
Dahlqvist JR, Salim R, Thomsen C, Vissing J. A quantitative method to assess muscle edema using short TI inversion recovery MRI. Sci Rep 2020; 10:7246. [PMID: 32350361 PMCID: PMC7190715 DOI: 10.1038/s41598-020-64287-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 04/07/2020] [Indexed: 12/16/2022] Open
Abstract
Muscle inflammation is an important component of disease pathophysiology in several muscular dystrophies. Hyperintensities on MRI sequences with short TI inversion recovery (STIR) reflect edema, or inflammation (STIR+). Conventionally, STIR evaluation has been done by visual inspection. In this study, we developed a quantitative STIR method, and tested its ability to identify STIR+ lesions in healthy controls and patients with Facioscapulohumeral muscular dystrophy and compared the results with visual STIR evaluation and quantitative T2 relaxation time mapping. The method was based on pixel-by-pixel histograms of the distribution of signal intensities from muscles. Signal intensities from healthy control muscles were averaged and used to define an upper reference limit. Muscles with >2.5% pixels above the limit were defined as being STIR+. The new method showed agreement with T2 relaxation time mapping in 95% of muscles. The visual STIR method only showed agreement with the quantitative STIR method and T2 relaxation time mapping in 88 and 84%, respectively. STIR sequences are available on most MR scanners and the post-processing used in the new quantitative method can be performed using free software. We therefore believe that the new method can play an important role in identifying STIR+ lesions in patients with neuromuscular diseases.
Collapse
Affiliation(s)
- Julia R Dahlqvist
- Copenhagen Neuromuscular Center, Section 3342, Department of Neurology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Ruth Salim
- Copenhagen Neuromuscular Center, Section 3342, Department of Neurology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Carsten Thomsen
- Department of Radiology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - John Vissing
- Copenhagen Neuromuscular Center, Section 3342, Department of Neurology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| |
Collapse
|
35
|
Hogrel JY, Decostre V, Ledoux I, de Antonio M, Niks EH, de Groot I, Straub V, Muntoni F, Ricotti V, Voit T, Seferian A, Gidaro T, Servais L. Normalized grip strength is a sensitive outcome measure through all stages of Duchenne muscular dystrophy. J Neurol 2020; 267:2022-2028. [PMID: 32206900 DOI: 10.1007/s00415-020-09800-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 01/20/2023]
Abstract
OBJECTIVE The main aim was to explore the changes in hand-grip strength in patients with Duchenne muscular dystrophy (DMD) aged 5-29 years. Secondary aims were to test the effect of mutation, ambulatory status and glucocorticoid use on grip strength and its changes over time and to compute the number of subjects needed for a clinical trial to stabilize grip strength. METHODS The analysis was performed on data collected during five international natural history studies on a cohort of DMD patients. Two hundred and two patients with genetically proven DMD were pooled from five different natural history studies. Excepting 13 patients with only one visit, the mean duration of follow-up was 2.2 ± 1.6 years. A total of 977 measurement points were collected. Grip strength was measured on the dominant side with a high precision dynamometer. The analysis was performed using absolute values and normalized values expressed in percentage of predicted values for age. RESULTS For absolute values, grip strength typically increased in ambulatory boys and decreased in non-ambulatory patients. However, when normalized, grip strength was already reduced at age 5 years and thereafter continued to fall away from normal values. The weaker the patients, the less strength they are prone to lose over again. INTERPRETATION Grip strength constitutes a sensitive and continuous outcome measure that can be used across all stages of DMD. Its measurement is easy to standardized, can be used in ambulatory and non-ambulatory patients and does not present any floor or ceiling effect. It is thus attractive as an outcome measure in therapeutic trials.
Collapse
Affiliation(s)
- Jean-Yves Hogrel
- Institute of Myology, GH Pitié-Salpêtrière, 75651, Paris Cedex 13, France.
| | - Valérie Decostre
- Institute of Myology, GH Pitié-Salpêtrière, 75651, Paris Cedex 13, France
| | - Isabelle Ledoux
- Institute of Myology, GH Pitié-Salpêtrière, 75651, Paris Cedex 13, France
| | - Marie de Antonio
- Institute of Myology, GH Pitié-Salpêtrière, 75651, Paris Cedex 13, France
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Imelda de Groot
- Department of Rehabilitation, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Newcastle Upon Tyne, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Valeria Ricotti
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Thomas Voit
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Andreea Seferian
- Institute of Myology, GH Pitié-Salpêtrière, 75651, Paris Cedex 13, France
| | - Teresa Gidaro
- Institute of Myology, GH Pitié-Salpêtrière, 75651, Paris Cedex 13, France
| | - Laurent Servais
- Centre de Référence Des Maladies Neuromusculaires, CHU de Liège, Liège, Belgium.,Department of Paediatrics, MDUK Neuromuscular Center, University of Oxford, Oxford, UK
| |
Collapse
|
36
|
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: 65] [Impact Index Per Article: 13.0] [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
|
37
|
Maggi L, Moscatelli M, Frangiamore R, Mazzi F, Verri M, De Luca A, Pasanisi MB, Baranello G, Tramacere I, Chiapparini L, Bruzzone MG, Mantegazza R, Aquino D. Quantitative Muscle MRI Protocol as Possible Biomarker in Becker Muscular Dystrophy. Clin Neuroradiol 2020; 31:257-266. [PMID: 31974637 DOI: 10.1007/s00062-019-00875-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/30/2019] [Indexed: 12/16/2022]
Abstract
PURPOSE Aim of this study is to compare Quantitative Magnetic Resonance Imaging (qMRI) measures between Becker Muscular Dystrophy (BMD) and Healthy Subjects (HS) and to correlate these parameters with clinical scores. METHODS Ten BMD patients (mean age ±standard deviation: 38.7 ± 15.0 years) and ten age-matched HS, were investigated through magnetic resonance imaging (MRI) at thigh and calf levels, including: 1) a standard axial T1-weighted sequence; 2) a volumetric T2-weighted sequence; 3) a multiecho spin-echo sequence; 4) a 2-point Dixon sequence; 5) a Diffusion Tensor Imaging (DTI) sequence. RESULTS Mean Fat Fraction (FF), T2-relaxation time and Fractional Anisotropy (FA) DTI at thigh and calf levels were significantly higher in BMD patients than in HS (p-values < 0.01). FF at thigh and calf levels significantly correlated with North Star Ambulatory Assessment (NSAA) score (p-values < 0.01) and6 Minutes Walking Test (6MWT) (p-values < 0.01), whereas only calf muscle FF was significantly associated with time to get up from floor (p-value = 0.01). T2 significantly correlated with NSAA score (p-value < 0.01), 6MWT (p-value = 0.02) and time to get up from floor (p-value < 0.01) only at calf level. Among DTI values, only FA in thigh and calf muscles significantly correlated with NSAA score, 6MWT and 10-m walk (all p-values < 0.05); only FA in calf muscles significantly correlated with time to get up from floor (p = 0.01). CONCLUSIONS Muscle FF, T2-relaxometry and DTI, seem to be a promising biomarker to assess BMD disease severity, although further studies are needed to evaluate changes over the time.
Collapse
Affiliation(s)
- Lorenzo Maggi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy. .,Neurology IV-Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Via Celoria 11, 20133, Milan, Italy.
| | - Marco Moscatelli
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Rita Frangiamore
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Federica Mazzi
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Mattia Verri
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Alberto De Luca
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maria Barbara Pasanisi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giovanni Baranello
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Irene Tramacere
- Department of Research and Clinical Development, Scientific Directorate, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Luisa Chiapparini
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Maria Grazia Bruzzone
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Renato Mantegazza
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Domenico Aquino
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|
38
|
Naarding KJ, Reyngoudt H, van Zwet EW, Hooijmans MT, Tian C, Rybalsky I, Shellenbarger KC, Le Louër J, Wong BL, Carlier PG, Kan HE, Niks EH. MRI vastus lateralis fat fraction predicts loss of ambulation in Duchenne muscular dystrophy. Neurology 2020; 94:e1386-e1394. [PMID: 31937624 PMCID: PMC7274919 DOI: 10.1212/wnl.0000000000008939] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 10/08/2019] [Indexed: 02/04/2023] Open
Abstract
Objective We studied the potential of quantitative MRI (qMRI) as a surrogate endpoint in Duchenne muscular dystrophy by assessing the additive predictive value of vastus lateralis (VL) fat fraction (FF) to age on loss of ambulation (LoA). Methods VL FFs were determined on longitudinal Dixon MRI scans from 2 natural history studies in Leiden University Medical Center (LUMC) and Cincinnati Children's Hospital Medical Center (CCHMC). CCHMC included ambulant patients, while LUMC included a mixed ambulant and nonambulant population. We fitted longitudinal VL FF values to a sigmoidal curve using a mixed model with random slope to predict individual trajectories. The additive value of VL FF over age to predict LoA was calculated from a Cox model, yielding a hazard ratio. Results Eighty-nine MRIs of 19 LUMC and 15 CCHMC patients were included. At similar age, 6-minute walking test distances were smaller and VL FFs were correspondingly higher in LUMC compared to CCHMC patients. Hazard ratio of a percent-point increase in VL FF for the time to LoA was 1.15 for LUMC (95% confidence interval [CI] 1.05–1.26; p = 0.003) and 0.96 for CCHMC (95% CI 0.84–1.10; p = 0.569). Conclusions The hazard ratio of 1.15 corresponds to a 4.11-fold increase of the instantaneous risk of LoA in patients with a 10% higher VL FF at any age. Although results should be confirmed in a larger cohort with prospective determination of the clinical endpoint, this added predictive value of VL FF to age on LoA supports the use of qMRI FF as an endpoint or stratification tool in clinical trials.
Collapse
Affiliation(s)
- Karin J Naarding
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH.
| | - Harmen Reyngoudt
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Erik W van Zwet
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Melissa T Hooijmans
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Cuixia Tian
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Irina Rybalsky
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Karen C Shellenbarger
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Julien Le Louër
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Brenda L Wong
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Pierre G Carlier
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Hermien E Kan
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| | - Erik H Niks
- From the Department of Neurology (K.J.N., E.H.N.), Department of Biostatistics (E.W.v.Z), and C.J. Gorter Center for High Field MRI (M.T.H., H.E.K.), Department of Radiology, Leiden University Medical Center, Zuid-Holland; Duchenne Center Netherlands (K.J.N., H.E.K., E.H.N.); AIM and CEA NMR Laboratory (H.R., J.L.L., P.G.C.), Neuromuscular Investigation Center, Institute of Myology, Paris, France; and Department of Neurology (C.T., I.R., K.C.S., B.L.W.), Cincinnati Children's Hospital Medical Center, OH
| |
Collapse
|
39
|
Strijkers GJ, Araujo EC, Azzabou N, Bendahan D, Blamire A, Burakiewicz J, Carlier PG, Damon B, Deligianni X, Froeling M, Heerschap A, Hollingsworth KG, Hooijmans MT, Karampinos DC, Loudos G, Madelin G, Marty B, Nagel AM, Nederveen AJ, Nelissen JL, Santini F, Scheidegger O, Schick F, Sinclair C, Sinkus R, de Sousa PL, Straub V, Walter G, Kan HE. Exploration of New Contrasts, Targets, and MR Imaging and Spectroscopy Techniques for Neuromuscular Disease - A Workshop Report of Working Group 3 of the Biomedicine and Molecular Biosciences COST Action BM1304 MYO-MRI. J Neuromuscul Dis 2020; 6:1-30. [PMID: 30714967 PMCID: PMC6398566 DOI: 10.3233/jnd-180333] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neuromuscular diseases are characterized by progressive muscle degeneration and muscle weakness resulting in functional disabilities. While each of these diseases is individually rare, they are common as a group, and a large majority lacks effective treatment with fully market approved drugs. Magnetic resonance imaging and spectroscopy techniques (MRI and MRS) are showing increasing promise as an outcome measure in clinical trials for these diseases. In 2013, the European Union funded the COST (co-operation in science and technology) action BM1304 called MYO-MRI (www.myo-mri.eu), with the overall aim to advance novel MRI and MRS techniques for both diagnosis and quantitative monitoring of neuromuscular diseases through sharing of expertise and data, joint development of protocols, opportunities for young researchers and creation of an online atlas of muscle MRI and MRS. In this report, the topics that were discussed in the framework of working group 3, which had the objective to: Explore new contrasts, new targets and new imaging techniques for NMD are described. The report is written by the scientists who attended the meetings and presented their data. An overview is given on the different contrasts that MRI can generate and their application, clinical needs and desired readouts, and emerging methods.
Collapse
Affiliation(s)
| | - Ericky C.A. Araujo
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology & NMR Laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | - Noura Azzabou
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology & NMR Laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | | | - Andrew Blamire
- Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, UK
| | - Jedrek Burakiewicz
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Pierre G. Carlier
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology & NMR Laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | - Bruce Damon
- Vanderbilt University Medical Center, Nashville, USA
| | - Xeni Deligianni
- Department of Radiology, Division of Radiological Physics, University Hospital Basel, Basel, Switzerland & Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | | | - Arend Heerschap
- Radboud University Medical Center, Nijmegen, the Netherlands
| | | | | | | | | | | | - Benjamin Marty
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology & NMR Laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | - Armin M. Nagel
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany & Division of Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | - Francesco Santini
- Department of Radiology, Division of Radiological Physics, University Hospital Basel, Basel, Switzerland & Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Olivier Scheidegger
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Fritz Schick
- University of Tübingen, Section on Experimental Radiology, Tübingen, Germany
| | | | | | | | - Volker Straub
- Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, UK
| | | | - Hermien E. Kan
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
40
|
Chrzanowski SM, Darras BT, Rutkove SB. The Value of Imaging and Composition-Based Biomarkers in Duchenne Muscular Dystrophy Clinical Trials. Neurotherapeutics 2020; 17:142-152. [PMID: 31879850 PMCID: PMC7007477 DOI: 10.1007/s13311-019-00825-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
As the drug development pipeline for Duchenne muscular dystrophy (DMD) rapidly advances, clinical trial outcomes need to be optimized. Effective assessment of disease burden, natural history progression, and response to therapy in clinical trials for Duchenne muscular dystrophy are critical factors for clinical trial success. By choosing optimal biomarkers to better assess therapeutic efficacy, study costs and sample size requirements can be reduced. Currently, functional measures continue to serve as the primary outcome for the majority of DMD clinical trials. Quantitative measures of muscle health, including magnetic resonance imaging and spectroscopy, electrical impedance myography, and ultrasound, sensitively identify diseased muscle, disease progression, and response to a therapeutic intervention. Furthermore, such non-invasive techniques have the potential to identify disease pathology prior to onset of clinical symptoms. Despite robust supportive evidence, non-invasive quantitative techniques are still not frequently utilized in clinical trials for Duchenne muscular dystrophy. Non-invasive quantitative techniques have demonstrated the ability to quantify disease progression and potential response to therapeutic intervention, and should be used as a supplement to current standard functional measures. Such methods have the potential to significantly accelerate the development and approval of therapies for DMD.
Collapse
Affiliation(s)
- Stephen M Chrzanowski
- Department of Medicine, Boston Children's Hospital, 300 Longwood Ave., Boston, MA, 02115, USA.
| | - Basil T Darras
- Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Seward B Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
41
|
Ropars J, Gravot F, Ben Salem D, Rousseau F, Brochard S, Pons C. Muscle MRI: A biomarker of disease severity in Duchenne muscular dystrophy? A systematic review. Neurology 2019; 94:117-133. [PMID: 31892637 DOI: 10.1212/wnl.0000000000008811] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/29/2019] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE To assess the evidence of a relationship between muscle MRI and disease severity in Duchenne muscular dystrophy (DMD). METHODS We conducted a systematic review of studies that analyzed correlations between MRI measurements and motor function in patients with DMD. PubMed, Cochrane, Scopus, and Web of Science were searched using relevant keywords and inclusion/exclusion criteria (January 1, 1990-January 31, 2019). We evaluated article quality using the Joanna Briggs Institute scale. Information regarding the samples included, muscles evaluated, MRI protocols and motor function tests used was collected from each article. Correlations between MRI measurements and motor function were reported exhaustively. RESULTS Seventeen of 1,629 studies identified were included. Most patients included were ambulant with a mean age of 8.9 years. Most studies evaluated lower limb muscles. Moderate to excellent correlations were found between MRI measurements and motor function. The strongest correlations were found for quantitative MRI measurements such as fat fraction or mean T2. Correlations were stronger for lower leg muscles such as soleus. One longitudinal study reported that changes in soleus mean T2 were highly correlated with changes in motor function. CONCLUSION The findings of this systematic review showed that MRI measurements can be used as biomarkers of disease severity in ambulant patients with DMD. Guidelines are proposed to help clinicians choose the most appropriate MRI measurements and muscles to evaluate. Studies exploring upper limb muscles, other stages of the disease, and sensitivity of measurements to change are needed.
Collapse
Affiliation(s)
- Juliette Ropars
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France.
| | - France Gravot
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| | - Douraied Ben Salem
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| | - François Rousseau
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| | - Sylvain Brochard
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| | - Christelle Pons
- From the Department of Pediatrics (J.R., F.G.), CHU Brest, Brest, France; Neuromuscular Center (J.R., S.B., C.P), Brest, France; Laboratoire du Traitement de l'Information Médicale (J.R., D.B.S., F.R, S.B., C.P.), LaTIM INSERM UMR1101, Brest, France; Department of Radiology (D.B.S.), CHU Brest, Brest, France; Institut Mines Télécom Atlantiques (F.R), Brest, France; and Department of Pediatric Physical and Medical Rehabilitation (S.B., C.P.), Fondation ILDYS, Brest, France
| |
Collapse
|
42
|
Gidaro T, Reyngoudt H, Le Louër J, Behin A, Toumi F, Villeret M, Araujo ECA, Baudin PY, Marty B, Annoussamy M, Hogrel JY, Carlier PG, Servais L. Quantitative nuclear magnetic resonance imaging detects subclinical changes over 1 year in skeletal muscle of GNE myopathy. J Neurol 2019; 267:228-238. [PMID: 31616990 DOI: 10.1007/s00415-019-09569-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/06/2019] [Accepted: 10/09/2019] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND OBJECTIVE To identify the most responsive and sensitive clinical outcome measures in GNE myopathy. METHODS ClinBio-GNE is a natural history study in GNE myopathy. Patients were assessed prospectively by clinical, functional and quantitative nuclear magnetic resonance imaging (qNMRI) evaluations. Strength and functional tests included Myogrip, Myopinch, MoviPlate and Brooke assessments for upper limb and the 6-min walk distance for lower limb. qNMRI was performed for determining the degree of fatty infiltration and trophicity in leg, thigh, forearm and hand skeletal muscles. Ten GNE myopathy patients were included. Three patients were non-ambulant. Age and gender-matched healthy subjects were used as controls. RESULTS Fatty infiltration and contractile cross-sectional area changed inversely and significantly in lower distal limbs and in proximal lower and distal upper limbs over 1 year. qNMRI indices and functional assessment results were strongly correlated. CONCLUSIONS Even in a limited number of patients, qNMRI could detect a significant change over a 1-year period in GNE myopathy, which suggests that qNMRI could constitute a surrogate endpoint in this slowly progressive disease. Quantitative NMRI outcome measures can monitor intramuscular fat accumulation with high responsiveness. Longer follow-up should improve our understanding of GNE myopathy evolution and also lead to the identification of non-invasive outcome measures with the highest discriminant power for upcoming clinical trials.
Collapse
Affiliation(s)
- Teresa Gidaro
- I-Motion-Pediatric Clinical Trials Department, Hôpital Armand Trousseau, Bâtiment Lemariey-Porte 20 * 2ème étage, 26 Avenue du Dr Arnold Netter, 75012, Paris, France.
| | - Harmen Reyngoudt
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA, DRF, IBFJ, MIRCen, Paris, France
| | - Julien Le Louër
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA, DRF, IBFJ, MIRCen, Paris, France
| | - Anthony Behin
- Neuromuscular Reference Center, Institute of Myology, Pitié-Salpêtrière Hospital (AP-HP), Paris, France
| | - Ferial Toumi
- I-Motion-Pediatric Clinical Trials Department, Hôpital Armand Trousseau, Bâtiment Lemariey-Porte 20 * 2ème étage, 26 Avenue du Dr Arnold Netter, 75012, Paris, France
| | - Melanie Villeret
- I-Motion-Pediatric Clinical Trials Department, Hôpital Armand Trousseau, Bâtiment Lemariey-Porte 20 * 2ème étage, 26 Avenue du Dr Arnold Netter, 75012, Paris, France
| | - Ericky C A Araujo
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA, DRF, IBFJ, MIRCen, Paris, France
| | - Pierre-Yves Baudin
- Consultants for Research in Imaging and Spectroscopy (C.R.I.S.), Tournai, Belgium
| | - Benjamin Marty
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA, DRF, IBFJ, MIRCen, Paris, France
| | - Melanie Annoussamy
- I-Motion-Pediatric Clinical Trials Department, Hôpital Armand Trousseau, Bâtiment Lemariey-Porte 20 * 2ème étage, 26 Avenue du Dr Arnold Netter, 75012, Paris, France
| | - Jean-Yves Hogrel
- Neuromuscular Physiology Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France
| | - Pierre G Carlier
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France.,NMR Laboratory, CEA, DRF, IBFJ, MIRCen, Paris, France
| | - Laurent Servais
- I-Motion-Pediatric Clinical Trials Department, Hôpital Armand Trousseau, Bâtiment Lemariey-Porte 20 * 2ème étage, 26 Avenue du Dr Arnold Netter, 75012, Paris, France.,Centre de référence Des Maladies Neuromusculaires, CHU de Liège, Liège, Belgium
| |
Collapse
|
43
|
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: 31] [Impact Index Per Article: 5.2] [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
|
44
|
Reyngoudt H, Lopez Kolkovsky AL, Carlier PG. Free intramuscular Mg 2+ concentration calculated using both 31 P and 1 H NMRS-based pH in the skeletal muscle of Duchenne muscular dystrophy patients. NMR IN BIOMEDICINE 2019; 32:e4115. [PMID: 31184793 DOI: 10.1002/nbm.4115] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 04/03/2019] [Accepted: 04/23/2019] [Indexed: 06/09/2023]
Abstract
Early studies have demonstrated that (total) magnesium was decreased in skeletal muscle of Duchenne muscular dystrophy (DMD) patients. Free intramuscular Mg2+ can be derived from 31 P NMRS measurements. The value of free intramuscular magnesium concentration ([Mg2+ ]) is highly dependent on precise knowledge of intracellular pH, which is abnormally alkaline in dystrophic muscle, possibly due to an expanded interstitial space, potentially causing an underestimation of [Mg2+ ]. We have recently shown that intracellular pH can be derived using 1 H NMRS of carnosine. Our aim was to determine whether 31 P NMRS-based [Mg2+ ] is, in fact, abnormally low in DMD patients, taking advantage of the 1 H NMRS-based pH. A comparative analysis was, therefore, made between [Mg2+ ] values calculated with both 1 H and 31 P NMRS-based approaches to determine pH in 25 DMD patients, on a 3-T clinical NMR scanner. [Mg2+ ] was also assessed with 31 P NMRS only in (forearm or leg) skeletal muscle of 60 DMD patients and 63 age-matched controls. Additionally, phosphodiester levels as well as quantitative NMRI indices including water T2 , fat fraction, contractile cross-sectional area and one-year changes were evaluated. The main finding was that the significant difference in [Mg2+ ] between DMD patients and controls was preserved even when the intracellular pH determined with 1 H NMRS was similar in both groups. Consequently, we observed that [Mg2+ ] is significantly lower in DMD patients compared with controls in the larger database where only 31 P NMRS data were obtained. Significant yet weak correlations existed between [Mg2+ ] and PDE, water T2 and fat fraction. We concluded that low [Mg2+ ] is an actual finding in DMD, whether intracellular pH is normal or alkaline, and that it is a likely consequence of membrane leakiness. The response of Mg2+ to therapeutic treatment remains to be investigated in neuromuscular disorders. Free [Mg2+ ] determination with 31 P NMRS is highly dependent on a precise knowledge of intracellular pH. The pH of Duchenne muscular dystrophy (DMD) patients, as determined by 31 P NMRS, is abnormally alkaline. We have recently shown that intracellular pH could be determined using 1 H NMRS of carnosine, and that intracellular pH was alkaline in a proportion of, but not all, DMD patients with a 31 P NMRS-based alkaline pH. Taking advantage of this 1 H NMRS-based intracellular pH, we found that free intramuscular [Mg2+ ] is in fact abnormally low in DMD patients.
Collapse
Affiliation(s)
- Harmen Reyngoudt
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France
- NMR Laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | - Alfredo L Lopez Kolkovsky
- 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
|
45
|
Murphy AP, Morrow J, Dahlqvist JR, Stojkovic T, Willis TA, Sinclair CDJ, Wastling S, Yousry T, Hanna MS, James MK, Mayhew A, Eagle M, Lee LE, Hogrel J, Carlier PG, Thornton JS, Vissing J, Hollingsworth KG, Straub V. Natural history of limb girdle muscular dystrophy R9 over 6 years: searching for trial endpoints. Ann Clin Transl Neurol 2019; 6:1033-1045. [PMID: 31211167 PMCID: PMC6562036 DOI: 10.1002/acn3.774] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 03/08/2019] [Indexed: 01/21/2023] Open
Abstract
OBJECTIVE Limb girdle muscular dystrophy type R9 (LGMD R9) is an autosomal recessive muscle disease for which there is currently no causative treatment. The development of putative therapies requires sensitive outcome measures for clinical trials in this slowly progressing condition. This study extends functional assessments and MRI muscle fat fraction measurements in an LGMD R9 cohort across 6 years. METHODS Twenty-three participants with LGMD R9, previously assessed over a 1-year period, were re-enrolled at 6 years. Standardized functional assessments were performed including: myometry, timed tests, and spirometry testing. Quantitative MRI was used to measure fat fraction in lower limb skeletal muscle groups. RESULTS At 6 years, all 14 muscle groups assessed demonstrated significant increases in fat fraction, compared to eight groups in the 1-year follow-up study. In direct contrast to the 1-year follow-up, the 6-min walk test, 10-m walk or run, timed up and go, stair ascend, stair descend and chair rise demonstrated significant decline. Among the functional tests, only FVC significantly declined over both the 1- and 6-year studies. INTERPRETATION These results further support fat fraction measurements as a primary outcome measure alongside functional assessments. The most appropriate individual muscles are the vastus lateralis, gracilis, sartorius, and gastrocnemii. Using composite groups of lower leg muscles, thigh muscles, or triceps surae, yielded high standardized response means (SRMs). Over 6 years, quantitative fat fraction assessment demonstrated higher SRM values than seen in functional tests suggesting greater responsiveness to disease progression.
Collapse
Affiliation(s)
- Alexander P. Murphy
- The John Walton Muscular Dystrophy Research CentreInstitute of Genetic MedicineNewcastle UniversityNewcastle Hospitals NHS Foundation TrustCentral ParkwayNewcastle Upon TyneUnited KingdomNE1 4EP
| | - Jasper Morrow
- Department of Molecular NeurosciencesMRC Centre for Neuromuscular DiseasesUCL Institute of NeurologyLondonUnited Kingdom
| | - Julia R. Dahlqvist
- Department of NeurologyCopenhagen Neuromuscular CenterRigshospitaletUniversity of CopenhagenBlegdamsvej 92100CopenhagenDenmark
| | - Tanya Stojkovic
- Institute of MyologyAP6HP, G‐H Pitié‐Salpêtrière47‐83 boulevard de l'hôpital75651Paris Cedex 13France
| | - Tracey A. Willis
- The Robert Jones and Agnes Hunt Orthopaedic HospitalOswestryShropshireUnited Kingdom
| | - Christopher D. J. Sinclair
- Department of Molecular NeurosciencesMRC Centre for Neuromuscular DiseasesUCL Institute of NeurologyLondonUnited Kingdom
| | - Stephen Wastling
- Department of Molecular NeurosciencesMRC Centre for Neuromuscular DiseasesUCL Institute of NeurologyLondonUnited Kingdom
| | - Tarek Yousry
- Department of Molecular NeurosciencesMRC Centre for Neuromuscular DiseasesUCL Institute of NeurologyLondonUnited Kingdom
| | - Michael S. Hanna
- Department of Molecular NeurosciencesMRC Centre for Neuromuscular DiseasesUCL Institute of NeurologyLondonUnited Kingdom
| | - Meredith K. James
- The John Walton Muscular Dystrophy Research CentreInstitute of Genetic MedicineNewcastle UniversityNewcastle Hospitals NHS Foundation TrustCentral ParkwayNewcastle Upon TyneUnited KingdomNE1 4EP
| | - Anna Mayhew
- The John Walton Muscular Dystrophy Research CentreInstitute of Genetic MedicineNewcastle UniversityNewcastle Hospitals NHS Foundation TrustCentral ParkwayNewcastle Upon TyneUnited KingdomNE1 4EP
| | - Michelle Eagle
- The John Walton Muscular Dystrophy Research CentreInstitute of Genetic MedicineNewcastle UniversityNewcastle Hospitals NHS Foundation TrustCentral ParkwayNewcastle Upon TyneUnited KingdomNE1 4EP
| | - Laurence E. Lee
- Department of Molecular NeurosciencesMRC Centre for Neuromuscular DiseasesUCL Institute of NeurologyLondonUnited Kingdom
| | - Jean‐Yves Hogrel
- Institute of MyologyNeuromuscular Investigation CenterPitié‐Salpêtrière HospitalParisFrance
| | - Pierre G. Carlier
- Institute of MyologyNeuromuscular Investigation CenterPitié‐Salpêtrière HospitalParisFrance
| | - John S. Thornton
- Department of Molecular NeurosciencesMRC Centre for Neuromuscular DiseasesUCL Institute of NeurologyLondonUnited Kingdom
| | - John Vissing
- Department of NeurologyCopenhagen Neuromuscular CenterRigshospitaletUniversity of CopenhagenBlegdamsvej 92100CopenhagenDenmark
| | - Kieren G. Hollingsworth
- Newcastle Magnetic Resonance CentreInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneUnited Kingdom
| | - Volker Straub
- The John Walton Muscular Dystrophy Research CentreInstitute of Genetic MedicineNewcastle UniversityNewcastle Hospitals NHS Foundation TrustCentral ParkwayNewcastle Upon TyneUnited KingdomNE1 4EP
| |
Collapse
|
46
|
Janssen MM, Harlaar J, Koopman B, de Groot IJ. Unraveling upper extremity performance in Duchenne muscular dystrophy: A biophysical model. Neuromuscul Disord 2019; 29:368-375. [DOI: 10.1016/j.nmd.2019.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/26/2019] [Accepted: 03/10/2019] [Indexed: 12/14/2022]
|
47
|
Ricotti V, Selby V, Ridout D, Domingos J, Decostre V, Mayhew A, Eagle M, Butler J, Guglieri M, Van der Holst M, Jansen M, Verschuuren JJGM, de Groot IJM, Niks EH, Servais L, Straub V, Voit T, Hogrel JY, Muntoni F. Respiratory and upper limb function as outcome measures in ambulant and non-ambulant subjects with Duchenne muscular dystrophy: A prospective multicentre study. Neuromuscul Disord 2019; 29:261-268. [PMID: 30852071 DOI: 10.1016/j.nmd.2019.02.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/05/2019] [Accepted: 02/08/2019] [Indexed: 11/19/2022]
Abstract
The field of translational research in Duchenne muscular dystrophy (DMD) has been transformed in the last decade by a number of therapeutic targets, mostly studied in ambulant patients. A paucity of studies focus on measures that capture the non-ambulant stage of the disease, and the transition between the ambulant and non-ambulant phase. In this prospective natural history study, we report the results of a comprehensive assessment of respiratory, upper limb function and upper limb muscle strength in a group of 89 DMD boys followed in 3 European countries, 81 receiving corticosteroids, spanning a wide age range (5-18 years) and functional abilities, from ambulant (n = 60) to non-ambulant (n = 29). Respiratory decline could be detected in the early ambulatory phase using Peak Expiratory Flow percentage predicted (PEF%), despite glucocorticoid use (mean annual decline: 4.08, 95% CI [-7.44,-0.72], p = 0.02 in ambulant; 4.81, 95% CI [-6.79,-2.82], p < 0.001 in non-ambulant). FVC% captured disease progression in non-ambulant DMD subjects, with an annual loss of 5.47% (95% CI [-6.48,-4.45], p < 0.001). Upper limb function measured with the Performance of Upper Limb (PUL 1.2) showed an annual loss of 4.13 points (95% CI [-4.79,3.47], p < 0.001) in the non-ambulant cohort. Measures of upper limb strength (MyoGrip and MyoPinch) showed a continuous decline independent of the ambulatory status, when reported as percentage predicted (grip force -5.51%, 95% CI [-6.54,-4.48], p < 0.001 in ambulant and a slower decline -2.86%; 95% CI -3.29,-2.43, p < 0.001, in non-ambulant; pinch force: -2.66%, 95% CI [-3.82,-1.51], p < 0.001 in ambulant and -2.23%, 95% CI [-2.92,-1.53], p < 0.001 in non-ambulant). Furthermore, we also explored the novel concept of a composite endpoint by combining respiratory, upper limb function and force domains: we were able to identify clear clinical progression in patients in whom an isolated measurement of only one of these domains failed to appreciate the yearly change. Our study contributes to the field of natural history of DMD, linking the ambulant and non-ambulant phases of the disease, and suggests that composite scores should be explored further.
Collapse
Affiliation(s)
- V Ricotti
- NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, Great Ormond Street Hospital Trust, University College London, London, UK; Solid Biosciences, London, UK.
| | - V Selby
- NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, Great Ormond Street Hospital Trust, University College London, London, UK; Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, Great Ormond Street Hospital Trust, London, UK
| | - D Ridout
- NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, Great Ormond Street Hospital Trust, University College London, London, UK; Population, Policy and Practice Program, UCL Great Ormond Street Institute of Child Health, London, UK
| | - J Domingos
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, Great Ormond Street Hospital Trust, London, UK
| | - V Decostre
- Groupe Hospitalier Pitié Salpêtrière, Institut de Myologie, Paris, France
| | - A Mayhew
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle, UK
| | - M Eagle
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle, UK
| | - J Butler
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, Great Ormond Street Hospital Trust, London, UK
| | - M Guglieri
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle, UK
| | | | - M Jansen
- Department of Rehabilitation, Donders Centre of Neuroscience, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | | | - I J M de Groot
- Department of Rehabilitation, Donders Centre of Neuroscience, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - E H Niks
- Leiden University Medical Centre, Leiden, The Netherlands
| | - L Servais
- Groupe Hospitalier Pitié Salpêtrière, Institut de Myologie, Paris, France
| | - V Straub
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle, UK
| | - T Voit
- NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, Great Ormond Street Hospital Trust, University College London, London, UK; Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, Great Ormond Street Hospital Trust, London, UK
| | - J Y Hogrel
- Groupe Hospitalier Pitié Salpêtrière, Institut de Myologie, Paris, France
| | - F Muntoni
- NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, Great Ormond Street Hospital Trust, University College London, London, UK; Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, Great Ormond Street Hospital Trust, London, UK.
| |
Collapse
|
48
|
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: 37] [Impact Index Per Article: 6.2] [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
|
49
|
Gerhalter T, Gast LV, Marty B, Martin J, Trollmann R, Schüssler S, Roemer F, Laun FB, Uder M, Schröder R, Carlier PG, Nagel AM. 23 Na MRI depicts early changes in ion homeostasis in skeletal muscle tissue of patients with duchenne muscular dystrophy. J Magn Reson Imaging 2019; 50:1103-1113. [PMID: 30719784 DOI: 10.1002/jmri.26681] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/24/2019] [Accepted: 01/24/2019] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a hereditary neuromuscular disease leading to progressive muscle wasting. Since there is a need for MRI variables that serve as early sensitive indicators of response to treatment, several quantitative MRI methods have been suggested for disease monitoring. PURPOSE To evaluate the potential of sodium (23 Na) and proton (1 H) MRI methods to assess early pathological changes in skeletal muscle of DMD. STUDY TYPE Prospective clinical study. POPULATION 23 Na and 1 H MRI of the right leg were performed in 13 patients with DMD (age 7.8 ± 2.4) and 14 healthy boys (age 9.5 ± 2.2). FIELD STRENGTH/SEQUENCE 3 T including a multiecho-spin-echo sequence, diffusion-weighted sequences, 1 H spectroscopy, 3-pt Dixon, and 23 Na ultrashort echo time sequences. ASSESSMENT We obtained water T2 maps, fat fraction (FF), pH, and diffusion properties of the skeletal muscle tissue. Moreover, total tissue sodium concentration (TSC) was calculated from the 23 Na sequence. Intracellular-weighted 23 Na signal (ICwS) was derived from 23 Na inversion-recovery imaging. STATISTICAL TESTS Results from DMD patients and controls were compared using Wilcoxon rank-sum tests and repeated analysis of variance (ANOVA). Spearman-rank correlations and area under the curve (AUC) were calculated to assess the performance of the different MRI methods to distinguish dystrophic from healthy muscle tissue. RESULTS FF, water T2 , and pH were higher in DMD patients (0.07 ± 0.03, 39.4 ± 0.8 msec, 7.06 ± 0.03, all P < 0.05) than in controls (0.02 ± 0.01, 36.0 ± 0.4 msec, 7.03 ± 0.02). No difference was observed in diffusion properties. TSC (26.0 ± 1.3 mM, P < 0.05) and ICwS (0.69 ± 0.05 a.u., P < 0.05) were elevated in DMD (controls: 16.5 ± 1.3 mM and 0.47 ± 0.04 a.u.). The ICwS was frequently abnormal in DMD even when water T2 , FF, and pH were in the normal range. 23 Na MRI showed higher AUC values in comparison to the 1 H methods. DATA CONCLUSION Sodium anomalies were regularly observed in patients with DMD compared with controls, and were present even in absence of fatty degenerative changes and water T2 increases. LEVEL OF EVIDENCE 1 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2019;50:1103-1113.
Collapse
Affiliation(s)
- Teresa Gerhalter
- NMR Laboratory, Institute of Myology, Paris, France.,NMR laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France.,Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lena V Gast
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Benjamin Marty
- NMR Laboratory, Institute of Myology, Paris, France.,NMR laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | - Jan Martin
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Regina Trollmann
- Department of Pediatrics, Division Neuropediatrics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Stephanie Schüssler
- Department of Pediatrics, Division Neuropediatrics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Frank Roemer
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Frederik B Laun
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Michael Uder
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Rolf Schröder
- Department of Neuropathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Pierre G Carlier
- NMR Laboratory, Institute of Myology, Paris, France.,NMR laboratory, CEA/DRF/IBFJ/MIRCen, Paris, France
| | - Armin M Nagel
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.,Division of Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institute of Medical Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
50
|
Brogna C, Cristiano L, Tartaglione T, Verdolotti T, Fanelli L, Ficociello L, Tasca G, Battini R, Coratti G, Forcina N, De Santis R, Norcia G, Carnicella S, Colosimo C, Carlier P, Pane M, Mercuri E. Functional levels and MRI patterns of muscle involvement in upper limbs in Duchenne muscular dystrophy. PLoS One 2018; 13:e0199222. [PMID: 29924868 PMCID: PMC6010282 DOI: 10.1371/journal.pone.0199222] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 02/18/2018] [Indexed: 11/19/2022] Open
Abstract
The aim of the study was to evaluate the spectrum of upper limb functional activities and imaging finding in a cohort of patients affected by Duchenne muscular dystrophy. Thirty-one patients of age between 5 and 29 years were included in the study (17 ambulant and 14 non-ambulant). They were all assessed using the Performance of Upper Limb (PUL) test and muscle MRI of shoulder, arm and forearm in order to establish if the functional scores obtained at shoulder, mid and distal level related to specific patterns of involvement in each upper limb segment on muscle MRI. At shoulder level, latissimus dorsi, serratus anterior, infraspinatus and subscapularis were always involved, even in patients with full functional scores at shoulder level. Diffuse and severe involvement of all muscles was found in the patients with a PUL shoulder functional score of ≤ 5. At arm level biceps brachii, brachialis and triceps were generally concordantly involved or spared. Some degree of involvement could already be detected in patients with reduced scores on the PUL mid domain. They were generally severely involved in patients with functional scores less than 6 at mid-level. At distal level supinator and pronator muscles were often involved, followed by brachioradialis and, less frequently, by the muscles of the flexor compartment. The extensor muscles were generally completely spared. A diffuse and severe involvement was found only in patients who had very low scores (8 or below) on the PUL distal domain. The integrated use of functional scales and imaging allowed to establish patterns of involvement at each level, and the functional scores that were more frequently associated with diffuse and severe involvement.
Collapse
Affiliation(s)
- Claudia Brogna
- Pediatric Neurology and Nemo Clinical Centre, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
| | - Lara Cristiano
- Pediatric Neurology and Nemo Clinical Centre, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
| | - Tommaso Tartaglione
- Department of Radiology, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
| | - Tommaso Verdolotti
- Department of Radiology, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
| | - Lavinia Fanelli
- Pediatric Neurology and Nemo Clinical Centre, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
| | - Luana Ficociello
- Department of Radiology, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
| | - Giorgio Tasca
- Institute of Neurology, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
| | - Roberta Battini
- Pediatric Neurology and Nemo Clinical Centre, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
- IRCSS Stella Maris, Pisa, Italy
| | - Giorgia Coratti
- Pediatric Neurology and Nemo Clinical Centre, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
| | - Nicola Forcina
- Pediatric Neurology and Nemo Clinical Centre, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
| | - Roberto De Santis
- Pediatric Neurology and Nemo Clinical Centre, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
| | - Giulia Norcia
- Pediatric Neurology and Nemo Clinical Centre, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
| | - Sara Carnicella
- Pediatric Neurology and Nemo Clinical Centre, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
| | - Cesare Colosimo
- Department of Radiology, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
| | - Pierre Carlier
- AIM and CEA NMR laboratory, Institute of Myology, Paris, France
| | - Marika Pane
- Pediatric Neurology and Nemo Clinical Centre, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
| | - Eugenio Mercuri
- Pediatric Neurology and Nemo Clinical Centre, Fondazione Policlinico Universitario "A. Gemelli IRCSS", Università Cattolica del sacro Cuore, Rome, Italy
- * E-mail:
| |
Collapse
|