1
|
Applebaum V, Baker E, Kim T, Stimpson G, Challenor P, Wedgwood KCA, Anderson M, Bamsey I, Baranello G, Manzur A, Muntoni F, Tsaneva-Atanasova K. Fully personalized modelling of Duchenne Muscular Dystrophy ambulation. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2025; 383:20240218. [PMID: 40172561 DOI: 10.1098/rsta.2024.0218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/20/2024] [Accepted: 01/14/2025] [Indexed: 04/04/2025]
Abstract
Duchenne Muscular Dystrophy is a progressive neuromuscular disorder characterized by the gradual weakening and deterioration of muscles, leading to loss of ambulation in affected individuals. This decline in mobility can be effectively assessed using the North Star Ambulatory Assessment (NSAA) scores, along with measures such as the 10-m walk time and the time taken to rise from the floor. We propose a dynamic linear model to predict the trajectories of these clinical outcomes, with a primary focus on NSAA scores. Our model aims to assist clinicians in forecasting the progression of the disease, thereby enabling more informed and personalized treatment plans for their patients. We also evaluate the effectiveness of our models in generating synthetic NSAA score datasets. We assess the performance of our modelling approach and compare the results with those of a previous study. We show that the most robust model demonstrates narrower prediction intervals and improved quantile coverage, indicating superior predictive accuracy and reliability.This article is part of the theme issue 'Uncertainty quantification for healthcare and biological systems (Part 2)'.
Collapse
Affiliation(s)
- Victor Applebaum
- Department of Mathematics and Statistics and EPSRC Hub for Quantitative Modelling in Healthcare, University of Exeter, Exeter, UK
| | - Evan Baker
- Department of Mathematics and Statistics and EPSRC Hub for Quantitative Modelling in Healthcare, University of Exeter, Exeter, UK
| | - Thomas Kim
- Certus Technology Associates Ltd, Exeter, UK
| | - Georgia Stimpson
- Dubowitz Neuromuscular Centre, NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, Great Ormond Street Hospital Trust, London, UK
| | - Peter Challenor
- Department of Mathematics and Statistics and EPSRC Hub for Quantitative Modelling in Healthcare, University of Exeter, Exeter, UK
| | - Kyle Carlton Abesser Wedgwood
- Department of Mathematics and Statistics and EPSRC Hub for Quantitative Modelling in Healthcare, University of Exeter, Exeter, UK
| | | | - Ian Bamsey
- Certus Technology Associates Ltd, Exeter, UK
| | - Giovanni Baranello
- Dubowitz Neuromuscular Centre, NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, Great Ormond Street Hospital Trust, London, UK
| | - Adnan Manzur
- Dubowitz Neuromuscular Centre, NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, Great Ormond Street Hospital Trust, London, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, Great Ormond Street Hospital Trust, London, UK
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics and Statistics and EPSRC Hub for Quantitative Modelling in Healthcare, University of Exeter, Exeter, UK
| |
Collapse
|
2
|
Gerhalter T, Marty B, Gast L, Roemer F, Baudin P, Trollmann R, Uder M, Carlier P, Nagel A. Longitudinal Follow-Up of Patients With Duchenne Muscular Dystrophy Using Quantitative 23Na and 1H MRI. J Cachexia Sarcopenia Muscle 2025; 16:e13812. [PMID: 40254293 PMCID: PMC12009636 DOI: 10.1002/jcsm.13812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 02/25/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Quantitative muscle MRI commonly evaluates disease activity and muscle wasting in Duchenne muscular dystrophy (DMD). Disturbances in ion homeostasis contribute to DMD pathophysiology, but their relationships with disease progression is unclear. 23Na MRI may provide insights into the disease course and treatment response. This longitudinal study assessed whether sodium levels are elevated in DMD patients regardless of fat fraction (FF) and whether baseline sodium levels influence FF changes over time. Additionally, we quantified the effect of slice selection on measured sodium values. METHODS Thirteen DMD boys (age 7.8 ± 2.4 years) underwent MRI of lower leg muscles at 3T at three visits, spaced 6 months apart. We assessed FF for disease progression and water T2, pH, apparent tissue sodium concentration (aTSC), and intracellular-weighted 23Na signal (ICwS) for disease activity. Fourteen healthy boys (age 9.5 ± 1.7 years) underwent the same MRI protocol once. Linear regression and mixed-effect modelling were used to examine sodium level increases and their impact on FF changes. RESULTS In DMD, muscles with FF < 10% exhibited significantly elevated aTSC (24.8 ± 4.6 mM vs. 14.5 ± 2.1 mM in controls, p < 0.001) and higher ICwS (23.6 ± 2.5 a.u. vs. 14.1 ± 2.1 a.u., p < 0.001). At Visit 1, FF values showed a significant negative association with aTSC (β = -17.30, p = 0.016) and ICwS (β = -21.02, p < 0.001). The first mixed-effect model, which assessed aTSC alone, showed no significant effect on FF progression but indicated a weak trend (p = 0.098). The second, more comprehensive model-incorporating also ICwS and water T2-revealed that FF changes were positively associated with aTSC (p = 0.0023) and negatively associated with ICwS and wT2 (p < 0.001 and p = 0.025, respectively), with ICwS showing a significant interaction with time (p = 0.0033). Varying slice positioning and slice number demonstrated minimal impact on aTSC and ICwS, with low CV (2%-4%) in the mid-belly region. CONCLUSIONS The study demonstrates significant MRI-based changes related to dystrophic alterations in DMD. We identified early alterations in sodium homeostasis, independent of FF. Our findings suggest that the relationship between sodium levels and FF progression is complex and may not be fully explained by total sodium measurements alone. Given the small sample size, further validation in larger cohorts is needed. Combined 1H and 23Na-MRI may offer deeper insights into how metabolic and ionic changes interact with FF progression and overall disease activity.
Collapse
Affiliation(s)
- Teresa Gerhalter
- Institute of Radiology University Hospital ErlangenFAUErlangenGermany
| | - Benjamin Marty
- NMR LaboratoryNeuromuscular Investigation Center, Institute of MyologyParisFrance
| | - Lena V. Gast
- Institute of Radiology University Hospital ErlangenFAUErlangenGermany
| | - Frank Roemer
- Institute of Radiology University Hospital ErlangenFAUErlangenGermany
| | - Pierre‐Yves Baudin
- NMR LaboratoryNeuromuscular Investigation Center, Institute of MyologyParisFrance
| | - Regina Trollmann
- Department of Pediatrics, Division NeuropediatricsFAUErlangenGermany
| | - Michael Uder
- Institute of Radiology University Hospital ErlangenFAUErlangenGermany
| | - Pierre G. Carlier
- Saint‐Luc University HospitalBrusselsBelgium
- Erasme University HospitalBrusselsBelgium
- Liège State UniversityLiègeBelgium
| | - Armin M. Nagel
- Institute of Radiology University Hospital ErlangenFAUErlangenGermany
- Division of Medical Physics in RadiologyDKFZHeidelbergGermany
| |
Collapse
|
3
|
Greiner E, Villa CR. "An Ounce of Prevention…". Muscle Nerve 2025. [PMID: 40091848 DOI: 10.1002/mus.28395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Affiliation(s)
- Eleanor Greiner
- Heart Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Chet R Villa
- Heart Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
4
|
Tamaroff J, Joy N, Damon B, Markham LW, Donnelly T, Su K, Buchowski MS, Crum K, Slaughter JC, Xu M, Burnette WB, Soslow J. Physical Activity Correlates With Skeletal Muscle MRI Findings in Individuals With Duchenne Muscular Dystrophy. Muscle Nerve 2025; 71:353-359. [PMID: 39719383 PMCID: PMC11799400 DOI: 10.1002/mus.28323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 12/04/2024] [Accepted: 12/08/2024] [Indexed: 12/26/2024]
Abstract
INTRODUCTION/AIMS Skeletal muscle magnetic resonance imaging (MRI) is a validated noninvasive tool to assess Duchenne muscular dystrophy (DMD) progression. There is interest in finding DMD biomarkers that decrease the burden of clinical trial participation, such as wearable devices. Our aim was to evaluate the relationship between activity, via accelerometry, and skeletal muscle MRI, particularly T2 mapping. METHODS DMD children and young adults completed skeletal muscle MRI and were asked to wear an accelerometer on the dominant wrist for 7 days. MRI data included fat-suppressed transverse relaxation time (T2) mapping of the calves and longitudinal relaxation time (T1) mapping. Activity was assessed as vector magnitudes (VMs) and fraction of time (FOT) in activity groups (sedentary 1 or 2, low 1 or 2, moderate-to-vigorous physical activity (MVPA)). RESULTS Participants (n = 22; median age 11.4 years, 41% ambulatory) wore the accelerometer for a median of 7 days. Longer T2 in multiple lower extremity muscles was negatively correlated with VMs per minute (tibialis posterior Spearman's rho = -0.68, p < 0.001), even when accounting for age, ambulatory status, or glucocorticoid use. Longer T2 of the tibialis posterior was positively correlated with FOT in sedentary 1 (rho = 0.49, p = 0.02) and negatively correlated with FOT in higher activity levels (low 1 (rho = -0.58, p = 0.004), low 2 (rho = -0.67, p = 0.002), MVPA (rho = -0.7, p < 0.001)). DISCUSSION In individuals with DMD, longer T2 on skeletal muscle MRI of the calves moderately correlated with lower activity levels indicating the potential use of home accelerometry as a future clinical trial biomarker of skeletal muscle health and progression in DMD.
Collapse
Affiliation(s)
- Jaclyn Tamaroff
- Division of Pediatric Endocrinology and DiabetesVanderbilt University Medical CenterNashvilleTennesseeUS
| | - Nicholas Joy
- Division of Pediatric CardiologyVanderbilt University Medical CenterNashvilleTennesseeUS
| | - Bruce Damon
- Vanderbilt University Institute of Imaging Science and the Department of Radiology and Radiological SciencesVanderbilt University Medical CenterNashvilleTennesseeUS
- Departments of Biomedical Engineering and Molecular Physiology and BiophysicsVanderbilt UniversityNashvilleTennesseeUS
- Carle Clinical Imaging Research ProgramStephens Family Clinical Research Institute, Carle HealthUrbanaIllinoisUS
| | - Larry W. Markham
- Division of Pediatric CardiologyRiley Hospital for Children and Indiana University School of MedicineIndianapolisIndianaUS
| | - Thomas Donnelly
- Division of Pediatric CardiologyVanderbilt University Medical CenterNashvilleTennesseeUS
| | - Karry Su
- Division of Pediatric CardiologyVanderbilt University Medical CenterNashvilleTennesseeUS
| | - Maciej S. Buchowski
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUS
| | - Kimberly Crum
- Division of Pediatric CardiologyVanderbilt University Medical CenterNashvilleTennesseeUS
| | - James C. Slaughter
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUS
| | - Meng Xu
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUS
| | - W. Bryan Burnette
- Division of Pediatric NeurologyVanderbilt University Medical CenterNashvilleTennesseeUS
| | - Jonathan Soslow
- Division of Pediatric CardiologyVanderbilt University Medical CenterNashvilleTennesseeUS
| |
Collapse
|
5
|
Vincenten SCC, Teeselink S, Mul K, Heskamp L, Kan HE, Heerschap A, Cameron D, Tasca G, Leung DG, Voermans NC, van Engelen BGM, van Alfen N. Muscle imaging in facioscapulohumeral muscular dystrophy research: A scoping review and expert recommendations. Neuromuscul Disord 2025; 47:105274. [PMID: 39884029 DOI: 10.1016/j.nmd.2025.105274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 12/17/2024] [Accepted: 01/03/2025] [Indexed: 02/01/2025]
Abstract
Clinical trial readiness is an important topic in the field of facioscapulohumeral muscular dystrophy (FSHD). As FSHD is a slowly progressive and clinically heterogeneous disease, imaging biomarkers have been proposed to complement clinical outcome measures. Muscle magnetic resonance imaging (MRI), ultrasound and dual energy X-ray absorptiometry (DEXA) have been used to measure disease severity, activity and progression. We conducted a scoping review of the literature on these imaging modalities to assess gaps in knowledge and subsequently collaborated with a panel of neuromuscular imaging experts to generate recommendations on the road ahead. We systematically searched PubMed, EMBASE and Cochrane Library databases. Three-hundred and twenty-eight studies were screened and one hundred and five studies were included. MRI indices related to intramuscular fat content, STIR positivity and T2water are used as diagnostic as well as prognostic and monitoring biomarkers. Ultrasound echogenicity can be used as a diagnostic and potentially as a prognostic and monitoring biomarker. DEXA lean muscle mass may be used as an additional monitoring biomarker. Each imaging modality has its own benefits but also challenges. Based on our expert opinions, we propose a roadmap to address these challenges, ensuring the optimal use of each modality in multi-center clinical trials in FSHD.
Collapse
Affiliation(s)
- Sanne C C Vincenten
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Sjan Teeselink
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Karlien Mul
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Linda Heskamp
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hermien E Kan
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands & Duchenne Center Netherlands, The Netherlands
| | - Arend Heerschap
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Donnie Cameron
- Department of Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Giorgio Tasca
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle upon Tyne, United Kingdom
| | - Doris G Leung
- Center for Genetic Muscle Disorders, Kennedy Krieger Institute, 1741 Ashland Ave., Baltimore, MD, 21205, USA
| | - Nicol C Voermans
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Baziel G M van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nens van Alfen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
6
|
Güttsches A, Forsting J, Kneifel M, Rehmann R, De Lorenzo A, Enax‐Krumova E, Froeling M, Vorgerd M, Schlaffke L. Pre- and post-skeletal muscle biopsy quantitative magnetic resonance imaging reveals correlations with histopathological findings. Eur J Neurol 2024; 31:e16479. [PMID: 39283047 PMCID: PMC11555129 DOI: 10.1111/ene.16479] [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: 03/29/2024] [Revised: 07/31/2024] [Accepted: 08/28/2024] [Indexed: 11/13/2024]
Abstract
BACKGROUND AND PURPOSE Quantitative muscle magnetic resonance imaging (MRI) is a promising non-invasive method in the diagnostic workup as well as follow-up of neuromuscular disorders. The aim of this study was to correlate quantitative MRI (qMRI) parameters to histopathological changes in skeletal muscle tissue and thus to verify the data from our pilot study. METHODS Twenty-six patients (eight females, 46.4 ± 15.1 years) were examined within 72 h before and within 24 h after a skeletal muscle biopsy using quantitative muscle MRI. Post-biopsy MRI was employed to pinpoint the exact localization of the biopsy. qMRI parameters including fat fraction, water T2 relaxation time and diffusion metrics including fractional anisotropy, mean diffusivity, axial diffusivity and radial diffusivity were extracted from the localization of the biopsy and correlated with histopathological findings. Additionally, three different segmentation masks were applied to the qMRI dataset, to evaluate whether the whole muscle represents the exact biopsy location. RESULTS Fat fraction and water T2 relaxation time in qMRI correlated significantly with the fat fraction in the muscle biopsy and histopathological inflammatory markers. Fractional anisotropy correlated with the quantity of type 2 fibres, whilst mean diffusivity correlated with p62. No differences were found using different segmentation masks in qMRI. CONCLUSIONS In this follow-up study, the results from our previous study were verified regarding the correlation of qMRI parameters with histopathological features in muscle biopsies, indicating that qMRI serves as a suitable non-invasive method in the follow-up of patients with neuromuscular disorders. If post-biopsy MRI is not available, whole muscle volume can be used for histopathological correlations.
Collapse
Affiliation(s)
- Anne‐Katrin Güttsches
- Department of NeurologyBG‐University Hospital Bergmannsheil, Ruhr‐University BochumBochumGermany
- Department of Neurology, Heimer Institute for Muscle ResearchBG‐University Hospital BergmannsheilBochumGermany
| | - Johannes Forsting
- Department of NeurologyBG‐University Hospital Bergmannsheil, Ruhr‐University BochumBochumGermany
| | - Moritz Kneifel
- Department of NeurologyBG‐University Hospital Bergmannsheil, Ruhr‐University BochumBochumGermany
- Department of Neurology, Heimer Institute for Muscle ResearchBG‐University Hospital BergmannsheilBochumGermany
| | - Robert Rehmann
- Department of NeurologyBG‐University Hospital Bergmannsheil, Ruhr‐University BochumBochumGermany
| | - Alice De Lorenzo
- Department of NeurologyBG‐University Hospital Bergmannsheil, Ruhr‐University BochumBochumGermany
| | - Elena Enax‐Krumova
- Department of NeurologyBG‐University Hospital Bergmannsheil, Ruhr‐University BochumBochumGermany
| | - Martijn Froeling
- Department of RadiologyUniversity Medical Centre UtrechtUtrechtNetherlands
| | - Matthias Vorgerd
- Department of NeurologyBG‐University Hospital Bergmannsheil, Ruhr‐University BochumBochumGermany
- Department of Neurology, Heimer Institute for Muscle ResearchBG‐University Hospital BergmannsheilBochumGermany
| | - Lara Schlaffke
- Department of NeurologyBG‐University Hospital Bergmannsheil, Ruhr‐University BochumBochumGermany
| |
Collapse
|
7
|
Yoon DY, Daniels MJ, Willcocks RJ, Triplett WT, Morales JF, Walter GA, Rooney WD, Vandenborne K, Kim S. Five multivariate Duchenne muscular dystrophy progression models bridging six-minute walk distance and MRI relaxometry of leg muscles. J Pharmacokinet Pharmacodyn 2024; 51:671-683. [PMID: 38609673 PMCID: PMC11470134 DOI: 10.1007/s10928-024-09910-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/15/2024] [Indexed: 04/14/2024]
Abstract
The study aimed to provide quantitative information on the utilization of MRI transverse relaxation time constant (MRI-T2) of leg muscles in DMD clinical trials by developing multivariate disease progression models of Duchenne muscular dystrophy (DMD) using 6-min walk distance (6MWD) and MRI-T2. Clinical data were collected from the prospective and longitudinal ImagingNMD study. Disease progression models were developed by a nonlinear mixed-effect modeling approach. Univariate models of 6MWD and MRI-T2 of five muscles were developed separately. Age at assessment was the time metric. Multivariate models were developed by estimating the correlation of 6MWD and MRI-T2 model variables. Full model estimation approach for covariate analysis and five-fold cross validation were conducted. Simulations were performed to compare the models and predict the covariate effects on the trajectories of 6MWD and MRI-T2. Sigmoid Imax and Emax models best captured the profiles of 6MWD and MRI-T2 over age. Steroid use, baseline 6MWD, and baseline MRI-T2 were significant covariates. The median age at which 6MWD is half of its maximum decrease in the five models was similar, while the median age at which MRI-T2 is half of its maximum increase varied depending on the type of muscle. The models connecting 6MWD and MRI-T2 successfully quantified how individual characteristics alter disease trajectories. The models demonstrate a plausible correlation between 6MWD and MRI-T2, supporting the use of MRI-T2. The developed models will guide drug developers in using the MRI-T2 to most efficient use in DMD clinical trials.
Collapse
Affiliation(s)
- Deok Yong Yoon
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Michael J Daniels
- Department of Statistics, University of Florida, Gainesville, FL, USA
| | | | - William T Triplett
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Juan Francisco Morales
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Glenn A Walter
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - William D Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Sarah Kim
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA.
| |
Collapse
|
8
|
Willis AB, Zelikovich AS, Sufit R, Ajroud-Driss S, Vandenborne K, Demonbreun AR, Batra A, Walter GA, McNally EM. Serum protein and imaging biomarkers after intermittent steroid treatment in muscular dystrophy. Sci Rep 2024; 14:28745. [PMID: 39567576 PMCID: PMC11579281 DOI: 10.1038/s41598-024-79024-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024] Open
Abstract
Weekly Steroids in Muscular Dystrophy (WSiMD) was a pilot study to evaluate once weekly prednisone in patients with Limb Girdle and Becker muscular dystrophy (LGMD and BMD, respectively). At study endpoint, there were trends towards increased lean mass, reduced fat mass, reduced creatine kinase and improved motor function. The investigation was motivated by studies in mouse muscular dystrophy models in which once weekly glucocorticoid exposure enhanced muscle strength and reduced fibrosis. WSiMD participants provided blood samples for aptamer serum profiling at baseline and after 6 months of weekly steroids. A subset completed magnetic resonance (MR) evaluation of muscle at study onset and endpoint. At baseline compared to age and sex-matched healthy controls, the aggregate serum protein profile in the WSiMD cohort was dominated by muscle proteins, reflecting leak of muscle proteins into serum. Disease status produced more proteins differentially present in serum compared to steroid-treatment effect. Nonetheless, a response to prednisone was discernable in the WSiMD cohort, even at this low dose. Glucocorticoids decreased muscle proteins and increased certain immune process- and matrix-associated proteins. Muscle MR fat fraction showed trends with functional status. The prednisone-responsive markers could be used in larger trial of prednisone efficacy.
Collapse
Affiliation(s)
- Alexander B Willis
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, 303 E Superior SQ 5-516, Chicago, IL, 60611, USA
| | - Aaron S Zelikovich
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, 303 E Superior SQ 5-516, Chicago, IL, 60611, USA
| | - Robert Sufit
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Senda Ajroud-Driss
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, 303 E Superior SQ 5-516, Chicago, IL, 60611, USA
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Abhinandan Batra
- Department of Physical Therapy, University of Louisiana at Monroe, Monroe, LA, USA
| | - Glenn A Walter
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, 303 E Superior SQ 5-516, Chicago, IL, 60611, USA.
| |
Collapse
|
9
|
Jenkins BM, Dixon LD, Kokesh KJ, Zingariello CD, Vandenborne K, Walter GA, Barnard AM. Skeletal muscle symptoms and quantitative MRI in females with dystrophinopathy. Muscle Nerve 2024; 70:988-999. [PMID: 39221574 PMCID: PMC11493146 DOI: 10.1002/mus.28235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION/AIMS The dystrophinopathies primarily affect males; however, female carriers of pathogenic dystrophin variants can develop skeletal muscle symptoms. This study aimed to evaluate muscle involvement and symptoms in females with dystrophinopathy using quantitative magnetic resonance imaging (MRI), functional assessments, and patient-reported outcomes. METHODS Controls and females with dystrophinopathy with muscle symptoms of pain, weakness, fatigue, or excessive tightness were enrolled in this cross-sectional study. Participants underwent lower extremity MRI to quantify muscle inflammation, replacement by fat, and disease asymmetry. Cardiac MRI, functional ability, muscle symptoms, and serum creatine kinase levels were also evaluated. RESULTS Six pediatric females with dystrophinopathy (mean age: 11.7 years), 11 adult females with dystrophinopathy (mean age: 41.3 years), and seven controls enrolled. The mean fat fraction was increased in females with dystrophinopathy compared to controls in the soleus (0.11 vs. 0.03, p = .0272) and vastus lateralis (0.16 vs. 0.03, p = .004). Magnetic resonance spectroscopy water T2, indicative of muscle inflammation, was elevated in the soleus and/or vastus lateralis in 11 of 17 individuals. North Star Ambulatory Assessment score was lower in the dystrophinopathy group compared to controls (29 vs. 34 points, p = .0428). From cardiac MRI, left ventricle T1 relaxation times were elevated in females with dystrophinopathy compared to controls (1311 ± 55 vs. 1263 ± 25 ms, p < .05), but ejection fraction and circumferential strain did not differ. DISCUSSION Symptomatic females with dystrophinopathy quantitatively demonstrate muscle replacement by fat and inflammation, along with impairments in functional ability and cardiac function. Additional research is needed to evaluate how symptoms and muscle involvement change longitudinally.
Collapse
Affiliation(s)
| | | | - Kevin J Kokesh
- Department of Pediatrics, Division of Pulmonology; University of Florida
| | - Carla D Zingariello
- Department of Pediatrics, Division of Pediatric Neurology; University of Florida
| | | | - Glenn A Walter
- Department of Physiology and Aging; University of Florida
| | | |
Collapse
|
10
|
Huang Y, Chen T, Hu Y, Li Z. Muscular MRI and magnetic resonance neurography in spinal muscular atrophy. Clin Radiol 2024; 79:673-680. [PMID: 38945793 DOI: 10.1016/j.crad.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/08/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024]
Abstract
Spinal muscular atrophy (SMA) is an autosomal recessive genetic disease caused by the degeneration of the α-motor neurons in the anterior horn of the spinal cord. SMA is clinically characterized by progressive and symmetrical muscle weakness and muscle atrophy and ends up with systemic multisystem abnormalities. Quantitative MRI (qMRI) has the advantages of non-invasiveness, objective sensitivity, and high reproducibility, and has important clinical value in evaluating the severity of neuromuscular diseases and monitoring the efficacy of treatment. This article summarizes the clinical use of muscular MRI and magnetic resonance neurography in assessing the progress of SMA.
Collapse
Affiliation(s)
- Y Huang
- Department of Radiology, Shenzhen Childrens Hospital, Shantou University Medical College Affiliated Shenzhen Childrens Hospital, Shenzhen, China
| | - T Chen
- Department of Radiology, Shenzhen Childrens Hospital, Shantou University Medical College Affiliated Shenzhen Childrens Hospital, Shenzhen, China; Department of Radiology, Shenzhen Children's Hospital, China Medical University, Shenzhen, China
| | - Y Hu
- Department of Radiology, Shenzhen Childrens Hospital, Shantou University Medical College Affiliated Shenzhen Childrens Hospital, Shenzhen, China; Department of Radiology, Shenzhen Children's Hospital, China Medical University, Shenzhen, China
| | - Z Li
- Department of Radiology, Shenzhen Childrens Hospital, Shantou University Medical College Affiliated Shenzhen Childrens Hospital, Shenzhen, China.
| |
Collapse
|
11
|
Norris AM, Fierman KE, Campbell J, Pitale R, Shahraj M, Kopinke D. Studying intramuscular fat deposition and muscle regeneration: insights from a comparative analysis of mouse strains, injury models, and sex differences. Skelet Muscle 2024; 14:12. [PMID: 38812056 PMCID: PMC11134715 DOI: 10.1186/s13395-024-00344-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/13/2024] [Indexed: 05/31/2024] Open
Abstract
Intramuscular fat (IMAT) infiltration, pathological adipose tissue that accumulates between muscle fibers, is a shared hallmark in a diverse set of diseases including muscular dystrophies and diabetes, spinal cord and rotator cuff injuries, as well as sarcopenia. While the mouse has been an invaluable preclinical model to study skeletal muscle diseases, they are also resistant to IMAT formation. To better understand this pathological feature, an adequate pre-clinical model that recapitulates human disease is necessary. To address this gap, we conducted a comprehensive in-depth comparison between three widely used mouse strains: C57BL/6J, 129S1/SvlmJ and CD1. We evaluated the impact of strain, sex and injury type on IMAT formation, myofiber regeneration and fibrosis. We confirm and extend previous findings that a Glycerol (GLY) injury causes significantly more IMAT and fibrosis compared to Cardiotoxin (CTX). Additionally, females form more IMAT than males after a GLY injury, independent of strain. Of all strains, C57BL/6J mice, both females and males, are the most resistant to IMAT formation. In regard to injury-induced fibrosis, we found that the 129S strain formed the least amount of scar tissue. Surprisingly, C57BL/6J of both sexes demonstrated complete myofiber regeneration, while both CD1 and 129S1/SvlmJ strains still displayed smaller myofibers 21 days post injury. In addition, our data indicate that myofiber regeneration is negatively correlated with IMAT and fibrosis. Combined, our results demonstrate that careful consideration and exploration are needed to determine which injury type, mouse model/strain and sex to utilize as preclinical model especially for modeling IMAT formation.
Collapse
Affiliation(s)
- Alessandra M Norris
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Kiara E Fierman
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Jillian Campbell
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Rhea Pitale
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Muhammad Shahraj
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA
| | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, Myology Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
12
|
Greiner E, Breaux A, Kasten J, Seo J, Ollberding NJ, Spar D, Ryan TD, Lang SM, Tian C, Sawnani H, Villa CR. Cardiac atrial pathology in Duchenne muscular dystrophy. Muscle Nerve 2024; 69:572-579. [PMID: 38426616 DOI: 10.1002/mus.28072] [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: 06/30/2023] [Revised: 02/17/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
INTRODUCTION/AIMS Duchenne muscular dystrophy (DMD) is characterized by fibrofatty replacement of muscle. This has been documented in the ventricular myocardium of DMD patients, but there is limited description of atrial involvement. The purpose of this study is to examine the arrhythmia and ectopy burden in patients with DMD and non-DMD dilated cardiomyopathy (DCM) and to characterize the cardiac histopathologic changes in DMD patients across the disease spectrum. METHODS This was a retrospective analysis of age-matched patients with DMD and non-DMD DCM who received a Holter monitor and cardiac imaging within 100 days of each other between 2010 and 2020. Twenty-four-hour Holter monitors were classified based on the most recent left ventricular ejection fraction at the time of monitoring. Cardiac histopathologic specimens from whole-heart examinations at the time of autopsy from three DMD patients and one DCM patient were reviewed. RESULTS A total of 367 patients with 1299 Holter monitor recordings were included over the study period, with 94% representing DMD patients and 6% non-DMD DCM. Patients with DMD had more atrial ectopy across the cardiac function spectrum (p < 0.05). There was no difference in ventricular ectopy. Four DMD patients developed symptomatic atrial arrhythmias. Autopsy specimens from DMD patients demonstrated fibrofatty infiltration of both atrial and ventricular myocardium. DISCUSSION The atrial myocardium in patients with DMD is unique. Autopsy specimens reveal fibofatty replacement of the atrial myocardium, which may be a nidus for both ectopy and arrhythmias in DMD patients.
Collapse
Affiliation(s)
- Eleanor Greiner
- Heart Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Andrea Breaux
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jennifer Kasten
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - JangDong Seo
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Nicholas J Ollberding
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - David Spar
- Heart Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Thomas D Ryan
- Heart Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Sean M Lang
- Heart Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Cuixia Tian
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Pediatric Neurology, Cincinnati Children's Hospital Medical Center & University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Hemant Sawnani
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Division of Pediatric Pulmonology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Chet R Villa
- Heart Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
13
|
Mercuri E, Vilchez JJ, Boespflug-Tanguy O, Zaidman CM, Mah JK, Goemans N, Müller-Felber W, Niks EH, Schara-Schmidt U, Bertini E, Comi GP, Mathews KD, Servais L, Vandenborne K, Johannsen J, Messina S, Spinty S, McAdam L, Selby K, Byrne B, Laverty CG, Carroll K, Zardi G, Cazzaniga S, Coceani N, Bettica P, McDonald CM. Safety and efficacy of givinostat in boys with Duchenne muscular dystrophy (EPIDYS): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Neurol 2024; 23:393-403. [PMID: 38508835 DOI: 10.1016/s1474-4422(24)00036-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Duchenne muscular dystrophy, the most common childhood muscular dystrophy, is caused by dystrophin deficiency. Preclinical and phase 2 study data have suggested that givinostat, a histone deacetylase inhibitor, might help to counteract the effects of this deficiency. We aimed to evaluate the safety and efficacy of givinostat in the treatment of Duchenne muscular dystrophy. METHODS This multicentre, randomised, double-blind, placebo-controlled, phase 3 trial was done at 41 tertiary care sites in 11 countries. Eligible participants were ambulant, male, and aged at least 6 years, had a genetically confirmed diagnosis of Duchenne muscular dystrophy, completed two four-stair climb assessments with a mean of 8 s or less (≤1 s variance), had a time-to-rise of at least 3 s but less than 10 s, and had received systemic corticosteroids for at least 6 months. Participating boys were randomly assigned (2:1, allocated according to a list generated by the interactive response technology provider) to receive either oral givinostat or matching placebo twice a day for 72 weeks, stratified by concomitant steroid use. Boys, investigators, and site and sponsor staff were masked to treatment assignment. The dose was flexible, based on weight, and was reduced if not tolerated. Boys were divided into two groups on the basis of their baseline vastus lateralis fat fraction (VLFF; measured by magnetic resonance spectroscopy): group A comprised boys with a VLFF of more than 5% but no more than 30%, whereas group B comprised boys with a VLFF of 5% or less, or more than 30%. The primary endpoint compared the effects of givinostat and placebo on the change in results of the four-stair climb assessment between baseline and 72 weeks, in the intention-to-treat, group A population. Safety was assessed in all randomly assigned boys who received at least one dose of study drug. When the first 50 boys in group A completed 12 months of treatment, an interim futility assessment was conducted, after which the sample size was adapted using masked data from the four-stair climb assessments. Furthermore, the starting dose of givinostat was reduced following a protocol amendment. This trial is registered with ClinicalTrials.gov, NCT02851797, and is complete. FINDINGS Between June 6, 2017, and Feb 22, 2022, 359 boys were assessed for eligibility. Of these, 179 were enrolled into the study (median age 9·8 years [IQR 8·1-11·0]), all of whom were randomly assigned (118 to receive givinostat and 61 to receive placebo); 170 (95%) boys completed the study. Of the 179 boys enrolled, 120 (67%) were in group A (81 givinostat and 39 placebo); of these, 114 (95%) completed the study. For participants in group A, comparing the results of the four-stair climb assessment at 72 weeks and baseline, the geometric least squares mean ratio was 1·27 (95% CI 1·17-1·37) for boys receiving givinostat and 1·48 (1·32-1·66) for those receiving placebo (ratio 0·86, 95% CI 0·745-0·989; p=0·035). The most common adverse events in the givinostat group were diarrhoea (43 [36%] of 118 boys vs 11 [18%] of 61 receiving placebo) and vomiting (34 [29%] vs 8 [13%]); no treatment-related deaths occurred. INTERPRETATION Among ambulant boys with Duchenne muscular dystrophy, results of the four-stair climb assessment worsened in both groups over the study period; however, the decline was significantly smaller with givinostat than with placebo. The dose of givinostat was reduced after an interim safety analysis, but no new safety signals were reported. An ongoing extension study is evaluating the long-term safety and efficacy of givinostat in patients with Duchenne muscular dystrophy. FUNDING Italfarmaco.
Collapse
Affiliation(s)
- Eugenio Mercuri
- Pediatric Neurology, Department of Woman and Child Health and Public Health, Child Health Area, Universita Cattolica del Sacro Cuore, Rome, Italy; Centro Clinico Nemo Fondazione Policlinico Gemelli IRCCS, Rome, Italy.
| | - Juan J Vilchez
- Servicio de Neurología, Neuromuscular Unit, CIBERER, EURO-RN-NMD, Hospital Universitario y Politécnico La Fe Valencia, Valencia, Spain
| | - Odile Boespflug-Tanguy
- I-Motion, Institut de Myologie, Hôpital Armand-Trousseau, APHP, Sorbonne Université, Paris, France; Université Paris Cité UMR INSERM 1141, Hôpital Robert Debré, Paris, France
| | | | - Jean K Mah
- Division of Pediatric Neurology, Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Nathalie Goemans
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Wolfgang Müller-Felber
- LMU Munich, University Hospital, Hauner Children's Hospital, Pediatric Neurology and Developmental Medicine, Munich, Germany
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands; Duchenne Center Netherlands, Netherlands
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology, Children's University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Enrico Bertini
- Research Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Giacomo P Comi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Milan, Italy; Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Katherine D Mathews
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Laurent Servais
- MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; Neuromuscular Reference Center, Department of Paediatrics, University and University Hospital of Liege, Belgium
| | - Krista Vandenborne
- ImagingDMD, University of Florida, Gainesville, FL, USA; Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Jessika Johannsen
- University Medical Center Hamburg-Eppendorf, Department of Pediatrics, Hamburg, Germany
| | - Sonia Messina
- Department of Clinical and Experimental Medicine, Unit of Neurodegenerative Diseases, AOU Policlinico G Martino, University of Mesina, Messina, Italy
| | - Stefan Spinty
- Department of Paediatric Neurology, Alder Hey Children's Hospital NHS Trust, Liverpool, UK
| | - Laura McAdam
- Holland Bloorview Kids Rehabilitation Hospital, Bloorview Research Institute, Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Kathryn Selby
- The University of British Columbia, Children's and Women's Health Centre, Vancouver, BC, Canada
| | - Barry Byrne
- Child Health Research Institute, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Chamindra G Laverty
- Department of Neuroscience, University of California, San Diego, San Diego, CA, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Willcocks RJ, Barnard AM, Daniels MJ, Forbes SC, Triplett WT, Brandsema JF, Finanger EL, Rooney WD, Kim S, Wang D, Lott DJ, Senesac CR, Walter GA, Sweeney HL, Vandenborne K. Clinical importance of changes in magnetic resonance biomarkers for Duchenne muscular dystrophy. Ann Clin Transl Neurol 2024; 11:67-78. [PMID: 37932907 PMCID: PMC10791017 DOI: 10.1002/acn3.51933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 10/03/2023] [Accepted: 10/08/2023] [Indexed: 11/08/2023] Open
Abstract
OBJECTIVE Magnetic resonance (MR) measures of muscle quality are highly sensitive to disease progression and predictive of meaningful functional milestones in Duchenne muscular dystrophy (DMD). This investigation aimed to establish the reproducibility, responsiveness to disease progression, and minimum clinically important difference (MCID) for multiple MR biomarkers at different disease stages in DMD using a large natural history dataset. METHODS Longitudinal MR imaging and spectroscopy outcomes and ambulatory function were measured in 180 individuals with DMD at three sites, including repeated measurements on two separate days (within 1 week) in 111 participants. These data were used to calculate day-to-day reproducibility, responsiveness (standardized response mean, SRM), minimum detectable change, and MCID. A survey of experts was also performed. RESULTS MR spectroscopy fat fraction (FF), as well as MR imaging transverse relaxation time (MRI-T2 ), measures performed in multiple leg muscles, and had high reproducibility (Pearson's R > 0.95). Responsiveness to disease progression varied by disease stage across muscles. The average FF from upper and lower leg muscles was highly responsive (SRM > 0.9) in both ambulatory and nonambulatory individuals. MCID estimated from the distribution of scores, by anchoring to function, and via expert opinion was between 0.01 and 0.05 for FF and between 0.8 and 3.7 ms for MRI-T2 . INTERPRETATION MR measures of FF and MRI T2 are reliable and highly responsive to disease progression. The MCID for MR measures is less than or equal to the typical annualized change. These results confirm the suitability of these measures for use in DMD and potentially other muscular dystrophies.
Collapse
Affiliation(s)
- Rebecca J. Willcocks
- Department of Physical Therapy, College of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
| | - Alison M. Barnard
- Department of Physical Therapy, College of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
| | | | - Sean C. Forbes
- Department of Physical Therapy, College of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
| | - William T. Triplett
- Department of Physical Therapy, College of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
| | - John F. Brandsema
- Division of NeurologyThe Children's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Erika L. Finanger
- Department of Pediatrics and NeurologyOregon Health & Science UniversityPortlandOregonUSA
| | - William D. Rooney
- Advanced Imaging Research CenterOregon Health & Science UniversityPortlandOregonUSA
| | - Sarah Kim
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Dah‐Jyuu Wang
- Department of RadiologyChildren's Hospital of PhiladelphiaPhiladelphiaPennsylvaniaUSA
| | - Donovan J. Lott
- Department of Physical Therapy, College of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
| | - Claudia R. Senesac
- Department of Physical Therapy, College of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
| | - Glenn A. Walter
- Department of Physiology and Functional Genomics, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - H. Lee Sweeney
- Department of Pharmacology and Therapeutics, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Krista Vandenborne
- Department of Physical Therapy, College of Public Health and Health ProfessionsUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
15
|
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
|
16
|
Landfeldt E, Alemán A, Abner S, Zhang R, Werner C, Tomazos I, Ferizovic N, Lochmüller H, Kirschner J. Predictors of Loss of Ambulation in Duchenne Muscular Dystrophy: A Systematic Review and Meta-Analysis. J Neuromuscul Dis 2024; 11:579-612. [PMID: 38669554 PMCID: PMC11091649 DOI: 10.3233/jnd-230220] [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: 03/20/2024] [Indexed: 04/28/2024]
Abstract
Objective The objective of this study was to describe predictors of loss of ambulation in Duchenne muscular dystrophy (DMD). Methods This systematic review and meta-analysis included searches of MEDLINE ALL, Embase, and the Cochrane Database of Systematic Reviews from January 1, 2000, to December 31, 2022, for predictors of loss of ambulation in DMD. Search terms included "Duchenne muscular dystrophy" as a Medical Subject Heading or free text term, in combination with variations of the term "predictor". Risk of bias was assessed using the Newcastle-Ottawa Scale. We performed meta-analysis pooling of hazard ratios of the effects of glucocorticoids (vs. no glucocorticoid therapy) by fitting a common-effect inverse-variance model. Results The bibliographic searches resulted in the inclusion of 45 studies of children and adults with DMD from 17 countries across Europe, Asia, and North America. Glucocorticoid therapy was associated with delayed loss of ambulation (overall meta-analysis HR deflazacort/prednisone/prednisolone: 0.44 [95% CI: 0.40-0.48]) (n = 25 studies). Earlier onset of first signs or symptoms, earlier loss of developmental milestones, lower baseline 6MWT (i.e.,<350 vs. ≥350 metres and <330 vs. ≥330 metres), and lower baseline NSAA were associated with earlier loss of ambulation (n = 5 studies). Deletion of exons 3-7, proximal mutations (upstream intron 44), single exon 45 deletions, and mutations amenable of skipping exon 8, exon 44, and exon 53, were associated with prolonged ambulation; distal mutations (intron 44 and downstream), deletion of exons 49-50, and mutations amenable of skipping exon 45, and exon 51 were associated with earlier loss of ambulation (n = 13 studies). Specific single-nucleotide polymorphisms in CD40 gene rs1883832, LTBP4 gene rs10880, SPP1 gene rs2835709 and rs11730582, and TCTEX1D1 gene rs1060575 (n = 7 studies), as well as race/ethnicity and level of family/patient deprivation (n = 3 studies), were associated with loss of ambulation. Treatment with ataluren (n = 2 studies) and eteplirsen (n = 3 studies) were associated with prolonged ambulation. Magnetic resonance biomarkers (MRI and MRS) were identified as significant predictors of loss of ambulation (n = 6 studies). In total, 33% of studies exhibited some risk of bias. Conclusion Our synthesis of predictors of loss of ambulation in DMD contributes to the understanding the natural history of disease and informs the design of new trials of novel therapies targeting this heavily burdened patient population.
Collapse
Affiliation(s)
| | - A. Alemán
- Department of Pediatrics, Division of Neurology, Children’s Hospital of Eastern Ontario, Research Institute, University of Ottawa, Ottawa, ON, Canada
- Department of Medicine, Division of Neurology, The Ottawa Hospital, Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | | | - R. Zhang
- PTC Therapeutics Sweden AB, Askim, Sweden
| | - C. Werner
- PTC Therapeutics Germany GmbH, Frankfurt, Germany
| | - I. Tomazos
- PTC Therapeutics Inc, South Plainfield, NJ, USA
| | | | - H. Lochmüller
- Department of Pediatrics, Division of Neurology, Children’s Hospital of Eastern Ontario, Research Institute, University of Ottawa, Ottawa, ON, Canada
- Department of Neuropediatrics and Muscle Disorders, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany
- Department of Medicine, Division of Neurology, The Ottawa Hospital, Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | - J. Kirschner
- Department of Neuropediatrics and Muscle Disorders, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| |
Collapse
|
17
|
Song Y, Xu K, Xu HY, Guo YK, Xu R, Fu H, Yuan WF, Zhou ZQ, Xu T, Chen XJ, Wang YL, Fu C, Zhou H, Cai XT, Li XS. Longitudinal changes in magnetic resonance imaging biomarkers of the gluteal muscle groups and functional ability in Duchenne muscular dystrophy: a 12-month cohort study. Pediatr Radiol 2023; 53:2672-2682. [PMID: 37889296 PMCID: PMC10697878 DOI: 10.1007/s00247-023-05791-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Quantitative magnetic resonance imaging (MRI) is considered an objective biomarker of Duchenne muscular dystrophy (DMD), but the longitudinal progression of MRI biomarkers in gluteal muscle groups and their predictive value for future motor function have not been described. OBJECTIVE To explore MRI biomarkers of the gluteal muscle groups as predictors of motor function decline in DMD by characterizing the progression over 12 months. MATERIALS AND METHODS A total of 112 participants with DMD were enrolled and underwent MRI examination of the gluteal muscles to determine fat fraction and longitudinal relaxation time (T1). Investigations were based on gluteal muscle groups including flexors, extensors, adductors, and abductors. The North Star Ambulatory Assessment and timed functional tests were performed. All participants returned for follow-up at an average of 12 months and were divided into two subgroups (functional stability/decline groups) based on changes in timed functional tests. Univariable and multivariable logistic regression methods were used to explore the risk factors associated with future motor function decline. RESULTS For the functional decline group, all T1 values decreased, while fat fraction values increased significantly over 12 months (P<0.05). For the functional stability group, only the fat fraction of the flexors and abductors increased significantly over 12 months (P<0.05). The baseline T1 value was positively correlated with North Star Ambulatory Assessment and negatively correlated with timed functional tests at the 12-month follow-up (P<0.001), while the baseline fat fraction value was negatively correlated with North Star Ambulatory Assessment and positively correlated with timed functional tests at the 12-month follow-up (P<0.001). Multivariate regression showed that increased fat fraction of the abductors was associated with future motor function decline (model 1: odds ratio [OR]=1.104, 95% confidence interval [CI]: 1.026~1.187, P=0.008; model 2: OR=1.085, 95% CI: 1.013~1.161, P=0.019), with an area under the curve of 0.874. CONCLUSION Fat fraction of the abductors is a powerful predictor of future motor functional decline in DMD patients at 12 months, underscoring the importance of focusing early on this parameter in patients with DMD.
Collapse
Affiliation(s)
- Yu Song
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ke Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hua-Yan Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying-Kun Guo
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Rong Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hang Fu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei-Feng Yuan
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Zi-Qi Zhou
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xi-Jian Chen
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Yi-Lei Wang
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuan Fu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hui Zhou
- Department of Rehabilitation Medicine, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiao-Tang Cai
- Department of Rehabilitation Medicine, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xue-Sheng Li
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
18
|
Huang XC, Huang YL, Guo YT, Li SY, Gao C, Chen JX, Ma JY, He B. An experimental study for quantitative assessment of fatty infiltration and blood flow perfusion in quadriceps muscle of rats using IDEAL-IQ and BOLD-MRI for early diagnosis of sarcopenia. Exp Gerontol 2023; 183:112322. [PMID: 37929293 DOI: 10.1016/j.exger.2023.112322] [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: 04/20/2023] [Revised: 10/15/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Severe sarcopenia may result in severe disability. Early diagnosis is currently the key to enhancing the treatment of sarcopenia, and there is an urgent need for a highly sensitive and dependable tool to evaluate the course of early sarcopenia in clinical practice. This study aims to investigate longitudinally the early diagnosability of magnetic resonance imaging (MRI)-based fat infiltration and blood flow perfusion technology in sarcopenia progression. METHODS 48 Sprague-Dawley rats were randomly assigned into six groups that were based on different periods of dexamethasone (DEX) injection (0, 2, 4, 6, 8, 10 days). Multimodal MRI was scanned to assess muscle mass. Grip strength and swimming exhaustion time of rats were measured to assess muscle strength and function. Immunofluorescence staining for CD31 was employed to assess skeletal muscle capillary formation, and western blot was used to detect vascular endothelial growth factor-A (VEGF-A) and muscle ring finger-1 (MuRF-1) protein expression. Subsequently, we analyzed the correlation between imaging and histopathologic parameters. A receiver operating characteristic (ROC) analysis was conducted to assess the effectiveness of quantitative MRI parameters for discriminating diagnosis in both pre- and post-modeling of DEX-induced sarcopenic rats. RESULTS Significant differences were found in PDFF, R2* and T2 values on day 2 of DEX-induction compared to the control group, occurring prior to the MRI-CSA values and limb grip strength on day 6 of induction and swimming exhaustion time on day 8 of induction. There is a strong correlation between MRI-CSA with HE-CSA values (r = 0.67; p < 0.001), oil red O (ORO) area with PDFF (r = 0.67; p < 0.001), microvascular density (MVD) (r = -0.79; p < 0.001) and VEGF-A (r = -0.73; p < 0.001) with R2*, MuRF-1 with MRI-CSA (r = -0.82; p < 0.001). The AUC of PDFF, R2*, and T2 values used for modeling evaluation are 0.81, 0.93, and 0.98, respectively. CONCLUSION Imaging parameters PDFF, R2*, and T2 can be used to sensitively evaluate early pathological changes in sarcopenia. The successful construction of a sarcopenia rat model can be assessed when PDFF exceeds 1.25, R2* exceeds 53.85, and T2 exceeds 33.88.
Collapse
Affiliation(s)
- Xin-Chen Huang
- Department of Radiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yi-Long Huang
- Department of Radiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yi-Tong Guo
- Department of Radiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Si-Yu Li
- Department of Physiology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Chao Gao
- Department of Radiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jia-Xin Chen
- Department of Radiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ji-Yao Ma
- Department of Radiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bo He
- Department of Radiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
19
|
Kim S, Willcocks RJ, Daniels MJ, Morales JF, Yoon DY, Triplett WT, Barnard AM, Conrado DJ, Aggarwal V, Belfiore‐Oshan R, Martinez TN, Walter GA, Rooney WD, Vandenborne K. Multivariate modeling of magnetic resonance biomarkers and clinical outcome measures for Duchenne muscular dystrophy clinical trials. CPT Pharmacometrics Syst Pharmacol 2023; 12:1437-1449. [PMID: 37534782 PMCID: PMC10583249 DOI: 10.1002/psp4.13021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/08/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023] Open
Abstract
Although regulatory agencies encourage inclusion of imaging biomarkers in clinical trials for Duchenne muscular dystrophy (DMD), industry receives minimal guidance on how to use these biomarkers most beneficially in trials. This study aims to identify the optimal use of muscle fat fraction biomarkers in DMD clinical trials through a quantitative disease-drug-trial modeling and simulation approach. We simultaneously developed two multivariate models quantifying the longitudinal associations between 6-minute walk distance (6MWD) and fat fraction measures from vastus lateralis and soleus muscles. We leveraged the longitudinal individual-level data collected for 10 years through the ImagingDMD study. Age of the individuals at assessment was chosen as the time metric. After the longitudinal dynamic of each measure was modeled separately, the selected univariate models were combined using correlation parameters. Covariates, including baseline scores of the measures and steroid use, were assessed using the full model approach. The nonlinear mixed-effects modeling was performed in Monolix. The final models showed reasonable precision of the parameter estimates. Simulation-based diagnostics and fivefold cross-validation further showed the model's adequacy. The multivariate models will guide drug developers on using fat fraction assessment most efficiently using available data, including the widely used 6MWD. The models will provide valuable information about how individual characteristics alter disease trajectories. We will extend the multivariate models to incorporate trial design parameters and hypothetical drug effects to inform better clinical trial designs through simulation, which will facilitate the design of clinical trials that are both more inclusive and more conclusive using fat fraction biomarkers.
Collapse
Affiliation(s)
- Sarah Kim
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of PharmacyUniversity of FloridaOrlandoFloridaUSA
| | | | | | - Juan Francisco Morales
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of PharmacyUniversity of FloridaOrlandoFloridaUSA
| | - Deok Yong Yoon
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of PharmacyUniversity of FloridaOrlandoFloridaUSA
| | | | - Alison M. Barnard
- Department of Physical TherapyUniversity of FloridaGainesvilleFloridaUSA
| | | | | | | | | | - Glenn A. Walter
- Department of Physiology and AgingUniversity of FloridaGainesvilleFloridaUSA
| | - William D. Rooney
- Advanced Imaging Research CenterOregon Health & Science UniversityPortlandOregonUSA
| | - Krista Vandenborne
- Department of Physical TherapyUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
20
|
Mitra A, Ahmed MA, Krishna R, Sun K, Gibbons FD, Campagne O, Rayad N, Roman YM, Albusaysi S, Burian M, Younis IR. Model-Informed Approaches and Innovative Clinical Trial Design for Adeno-Associated Viral Vector-Based Gene Therapy Product Development: A White Paper. Clin Pharmacol Ther 2023; 114:515-529. [PMID: 37313953 DOI: 10.1002/cpt.2972] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/02/2023] [Indexed: 06/15/2023]
Abstract
The promise of viral vector-based gene therapy (GT) as a transformative paradigm for treating severely debilitating and life-threatening diseases is slowly coming to fruition with the recent approval of several drug products. However, they have a unique mechanism of action often necessitating a tortuous clinical development plan. Expertise in such complex therapeutic modality is still fairly limited in this emerging class of adeno-associated virus (AAV) vector-based gene therapies. Because of the irreversible mode of action and incomplete understanding of genotype-phenotype relationship and disease progression in rare diseases careful considerations should be given to GT product's benefit-risk profile. In particular, special attention needs to be paid to safe dose selection, reliable dose exposure response (including clinically relevant endpoints), or creative approaches in study design targeting small patient populations during clinical development. We believe that quantitative tools encompassed within model-informed drug development (MIDD) framework fits quite well in the development of such novel therapies, as they enable us to benefit from the totality of data approach in order to support dose selection as well as optimize clinical trial designs, end point selection, and patient enrichment. In this thought leadership paper, we provide our collective experiences, identify challenges, and suggest areas of improvement in applications of modeling and innovative trial design in development of AAV-based GT products and reflect on the challenges and opportunities for incorporating MIDD tools and more in rational development of these products.
Collapse
Affiliation(s)
- Amitava Mitra
- Clinical Pharmacology, Kura Oncology, Boston, Massachusetts, USA
| | - Mariam A Ahmed
- Quantitative Clinical Pharmacology, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Rajesh Krishna
- Integrated Drug Development, Certara USA, Inc., Princeton, New Jersey, USA
| | - Kefeng Sun
- Quantitative Clinical Pharmacology, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Francis D Gibbons
- Quantitative Solutions, Preclinical and Translational Sciences, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Olivia Campagne
- Quantitative Clinical Pharmacology, Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Noha Rayad
- Clinical Pharmacology, Modeling and Simulation, Parexel International (MA) Corporation, Mississauga, Ontario, Canada
| | - Youssef M Roman
- Department of Pharmacotherapy and Outcomes Science, Virginia Commonwealth University School of Pharmacy, Richmond, Virginia, USA
| | - Salwa Albusaysi
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Maria Burian
- Translational Medicine Neuroscience and Gene Therapy, UCB Biopharma SRL, Braine-l'Alleud, Belgium
| | - Islam R Younis
- Clinical Pharmacology Sciences, Gilead Science, Inc, Foster City, California, USA
| |
Collapse
|
21
|
Martín-Noguerol T, Barousse R, Wessell DE, Rossi I, Luna A. Clinical applications of skeletal muscle diffusion tensor imaging. Skeletal Radiol 2023; 52:1639-1649. [PMID: 37083977 DOI: 10.1007/s00256-023-04350-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023]
Abstract
Diffusion tensor imaging (DTI) may allow the determination of new threshold values, based on water anisotropy, to differentiate between healthy muscle and various pathological processes. Additionally, it may quantify treatment monitoring or training effects. Most current studies have evaluated the potential of DTI of skeletal muscle to assess sports-related injuries or therapy, and training monitoring. Another critical area of application of this technique is the characterization and monitoring of primary and secondary myopathies. In this manuscript, we review the application of DTI in the evaluation of skeletal muscle in these and other novel clinical scenarios, with emphasis on the use of quantitative imaging-derived biomarkers. Finally, the main limitations of the introduction of DTI in the clinical setting and potential areas of future use are discussed.
Collapse
Affiliation(s)
| | | | | | | | - Antonio Luna
- MRI Unit, Radiology Department, HT Médica, Jaén, Spain
| |
Collapse
|
22
|
Jiang N, Xv Y, Sun X, Feng L, Wang YB, Jiang XL. Study on self-management of real-time and individualized support in stroke patients based on resilience: a protocol for a randomized controlled trial. Trials 2023; 24:493. [PMID: 37537646 PMCID: PMC10401848 DOI: 10.1186/s13063-023-07475-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/24/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND The transitional period from hospital to home is vital for stroke patients, but it poses serious challenges. Good self-management ability can optimize disease outcomes. However, stroke patients in China have a low level of self-management ability during the transitional period, and a lack of effective support may be the reason. With the rapid development of technology, using wearable monitors to achieve real-time and individualized support may be the key to solving this problem. This study uses a randomized controlled trial design to assess the efficacy of using wearable technology to realize real-time and individualized self-management support in stroke patients' self-management behavior during the transitional period following discharge from hospital. METHODS This parallel-group randomized controlled trial will be conducted in two hospitals and patients' homes. A total of 183 adult stroke patients will be enrolled in the study and randomly assigned to three groups in a 1:1:1 ratio. The smartwatch intervention group (n = 61) will receive Real-time and Individualized Self-management Support (RISS) program + routine care, the wristband group (n = 61) will wear a fitness tracker (self-monitoring) + routine care, and the control group (n = 61) will receive routine stroke care. The intervention will last for 6 months. The primary outcomes are neurological function status, self-management behavior, quality of life, biochemical indicators, recurrence rate, and unplanned readmission rate. Secondary outcomes are resilience, patient activation, psychological status, and caregiver assessments. The analysis is intention-to-treat. The intervention effect will be evaluated at baseline (T0), 2 months after discharge (T1), 3 months after discharge (T2), and 6 months after discharge (T3). DISCUSSION The cloud platform designed in this study not only has the function of real-time recording but also can push timely solutions when patients have abnormal conditions, as well as early warnings or alarms. This study could also potentially help patients develop good self-management habits through resilience theory, wearable devices, and individualized problem-solution library of self-management which can lay the foundation for long-term maintenance and continuous improvement of good self-management behavior in the future. TRIAL REGISTRATION The ethics approval has been granted by the Ethics Committee of West China Hospital, Sichuan University (2022-941). All patients will be informed of the study details and sign a written informed consent form before enrollment. The research results will be reported in conferences and peer-reviewed publications. The trial registration number is ChiCTR2300070384 . Registered on 11 April 2023.
Collapse
Affiliation(s)
- N Jiang
- West China School of Nursing/West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Y Xv
- Department of Surgical Anesthesiology, Tai'an City Central Hospital, Tai'an, China
| | - X Sun
- Department of Neurology, Tianjin Third Central Hospital, Tianjin, China
| | - L Feng
- Department of Neurology/West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China
| | - Y B Wang
- Tai'an Tax Bureau, State Administration of Taxation, Tai'an, China
| | - X L Jiang
- West China School of Nursing/West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
23
|
Mohassel P, Yun P, Syeda S, Batra A, Bradley AJ, Donkervoort S, Monges S, Cohen JS, Leung DG, Munell F, Ortez C, Sánchez‐Montáñez A, Karachunski P, Brandsema J, Medne L, Chaudhry V, Tasca G, Foley AR, Udd B, Arai AE, Walter GA, Bönnemann CG. A comprehensive study of skeletal muscle imaging in FHL1-related reducing body myopathy. Ann Clin Transl Neurol 2023; 10:1442-1455. [PMID: 37483011 PMCID: PMC10424657 DOI: 10.1002/acn3.51834] [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: 02/07/2023] [Revised: 05/09/2023] [Accepted: 05/31/2023] [Indexed: 07/25/2023] Open
Abstract
OBJECTIVE FHL1-related reducing body myopathy is an ultra-rare, X-linked dominant myopathy. In this cross-sectional study, we characterize skeletal muscle ultrasound, muscle MRI, and cardiac MRI findings in FHL1-related reducing body myopathy patients. METHODS Seventeen patients (11 male, mean age 35.4, range 12-76 years) from nine independent families with FHL1-related reducing body myopathy underwent clinical evaluation, muscle ultrasound (n = 11/17), and lower extremity muscle MRI (n = 14/17), including Dixon MRI (n = 6/17). Muscle ultrasound echogenicity was graded using a modified Heckmatt scale. T1 and STIR axial images of the lower extremity muscles were evaluated for pattern and distribution of abnormalities. Quantitative analysis of intramuscular fat fraction was performed using the Dixon MRI images. Cardiac studies included electrocardiogram (n = 15/17), echocardiogram (n = 17/17), and cardiac MRI (n = 6/17). Cardiac muscle function, T1 maps, T2-weighted black blood images, and late gadolinium enhancement patterns were analyzed. RESULTS Muscle ultrasound showed a distinct pattern of increased echointensity in skeletal muscles with a nonuniform, multifocal, and "geographical" distribution, selectively involving the deeper fascicles of muscles such as biceps and tibialis anterior. Lower extremity muscle MRI showed relative sparing of gluteus maximus, rectus femoris, gracilis, and lateral gastrocnemius muscles and an asymmetric and multifocal, "geographical" pattern of T1 hyperintensity within affected muscles. Cardiac studies revealed mild and nonspecific abnormalities on electrocardiogram and echocardiogram with unremarkable cardiac MRI studies. INTERPRETATION Skeletal muscle ultrasound and muscle MRI reflect the multifocal aggregate formation in muscle in FHL1-related reducing body myopathy and are practical and informative tools that can aid in diagnosis and monitoring of disease progression.
Collapse
Affiliation(s)
- Payam Mohassel
- Neurogenetics BranchNational Institute of Neurological Disorders and StrokeBethesdaMDUSA
- Present address:
Department of NeurologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Pomi Yun
- Neurogenetics BranchNational Institute of Neurological Disorders and StrokeBethesdaMDUSA
| | - Safoora Syeda
- Neurogenetics BranchNational Institute of Neurological Disorders and StrokeBethesdaMDUSA
| | - Abhinandan Batra
- Department of Physical TherapyUniversity of FloridaGainesvilleFLUSA
| | | | - Sandra Donkervoort
- Neurogenetics BranchNational Institute of Neurological Disorders and StrokeBethesdaMDUSA
| | - Soledad Monges
- Servicio de NeurologíaHospital de Pediatría J.P. GarrahanBuenos AiresArgentina
| | - Julie S. Cohen
- Department of NeurologyKennedy Krieger Institute, Johns Hopkins University School of MedicineBaltimoreMDUSA
| | - Doris G. Leung
- Department of NeurologyKennedy Krieger Institute, Johns Hopkins University School of MedicineBaltimoreMDUSA
| | - Francina Munell
- Pediatric NeurologyVall d'Hebron University HospitalBarcelonaSpain
| | - Carlos Ortez
- Department of Pediatric Neurology, Neuromuscular UnitHospital Sant Joan de Déu and Institut de Recerca Sant Joan de DéuBarcelonaSpain
| | - Angel Sánchez‐Montáñez
- Pediatric NeuroradiologyHospital Universitari Vall d'Hebron, Vall d'Hebron, Autonomous University of BarcelonaBarcelonaSpain
| | | | - John Brandsema
- Division of NeurologyChildren's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Livija Medne
- Division of NeurologyChildren's Hospital of PhiladelphiaPhiladelphiaPAUSA
| | - Vinay Chaudhry
- Department of NeurologyUniversity of North CarolinaChapel HillNCUSA
| | - Giorgio Tasca
- Unità Operativa Complessa di NeurologiaFondazione Policlinico Universitario A. Gemelli IRCCSRomeItaly
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastleUpon TyneUK
| | - A. Reghan Foley
- Neurogenetics BranchNational Institute of Neurological Disorders and StrokeBethesdaMDUSA
| | - Bjarne Udd
- Folkhalsan Research Center, Department of Medical GeneticsUniversity of HelsinkiHelsinkiFinland
| | - Andrew E. Arai
- Advanced Cardiovascular Imaging LaboratoryNHLBI, NIHBethesdaMDUSA
| | - Glenn A. Walter
- Department of Physiology and Functional GenomicsUniversity of FloridaGainesvilleFLUSA
| | - Carsten G. Bönnemann
- Neurogenetics BranchNational Institute of Neurological Disorders and StrokeBethesdaMDUSA
| |
Collapse
|
24
|
Nieves-Rodriguez S, Barthélémy F, Woods JD, Douine ED, Wang RT, Scripture-Adams DD, Chesmore KN, Galasso F, Miceli MC, Nelson SF. Transcriptomic analysis of paired healthy human skeletal muscles to identify modulators of disease severity in DMD. Front Genet 2023; 14:1216066. [PMID: 37576554 PMCID: PMC10415210 DOI: 10.3389/fgene.2023.1216066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 07/04/2023] [Indexed: 08/15/2023] Open
Abstract
Muscle damage and fibro-fatty replacement of skeletal muscles is a main pathologic feature of Duchenne muscular dystrophy (DMD) with more proximal muscles affected earlier and more distal affected later in the disease course, suggesting that different skeletal muscle groups possess distinctive characteristics that influence their susceptibility to disease. To explore transcriptomic factors driving differential gene expression and modulating DMD skeletal muscle severity, we characterized the transcriptome of vastus lateralis (VL), a more proximal and susceptible muscle, relative to tibialis anterior (TA), a more distal and protected muscle, in 15 healthy individuals using bulk RNA sequencing to identify gene expression differences that may mediate their relative susceptibility to damage with loss of dystrophin. Matching single nuclei RNA sequencing data was generated for 3 of the healthy individuals, to infer cell composition in the bulk RNA sequencing dataset and to improve mapping of differentially expressed genes to their cell source of expression. A total of 3,410 differentially expressed genes were identified and mapped to cell type using single nuclei RNA sequencing of muscle, including long non-coding RNAs and protein coding genes. There was an enrichment of genes involved in calcium release from the sarcoplasmic reticulum, particularly in the myofibers and these myofiber genes were higher in the VL. There was an enrichment of genes in "Collagen-Containing Extracellular Matrix" expressed by fibroblasts, endothelial, smooth muscle and pericytes, with most genes higher in the TA, as well as genes in "Regulation Of Apoptotic Process" expressed across all cell types. Previously reported genetic modifiers were also enriched within the differentially expressed genes. We also identify 6 genes with differential isoform usage between the VL and TA. Lastly, we integrate our findings with DMD RNA sequencing data from the TA, and identify "Collagen-Containing Extracellular Matrix" and "Negative Regulation Of Apoptotic Process" as differentially expressed between DMD compared to healthy. Collectively, these findings propose novel candidate mechanisms that may mediate differential muscle susceptibility in muscular dystrophies and provide new insight into potential therapeutic targets.
Collapse
Affiliation(s)
- Shirley Nieves-Rodriguez
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, United States
| | - Florian Barthélémy
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, United States
- Department of Microbiology, David Geffen School of Medicine and College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jeremy D. Woods
- Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Emilie D. Douine
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, United States
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Richard T. Wang
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, United States
| | - Deirdre D. Scripture-Adams
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, United States
- Department of Microbiology, David Geffen School of Medicine and College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA, United States
| | - Kevin N. Chesmore
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, United States
| | - Francesca Galasso
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - M. Carrie Miceli
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, United States
- Department of Microbiology, David Geffen School of Medicine and College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA, United States
| | - Stanley F. Nelson
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Duchenne Muscular Dystrophy at UCLA, Los Angeles, CA, United States
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
25
|
McCormack NM, Nguyen NY, Tully CB, Oliver T, Fiorillo AA, Heier CR. Vamorolone improves Becker muscular dystrophy and increases dystrophin protein in bmx model mice. iScience 2023; 26:107161. [PMID: 37534133 PMCID: PMC10391915 DOI: 10.1016/j.isci.2023.107161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/15/2023] [Accepted: 06/13/2023] [Indexed: 08/04/2023] Open
Abstract
There is no approved therapy for Becker muscular dystrophy (BMD), a genetic muscle disease caused by in-frame dystrophin deletions. We previously developed the dissociative corticosteroid vamorolone for treatment of the allelic, dystrophin-null disease Duchenne muscular dystrophy. We hypothesize vamorolone can treat BMD by safely reducing inflammatory signaling in muscle and through a novel mechanism of increasing dystrophin protein via suppression of dystrophin-targeting miRNAs. Here, we test this in the bmx mouse model of BMD. Daily oral treatment with vamorolone or prednisolone improves bmx grip strength and hang time phenotypes. Both drugs reduce myofiber size and decrease the percentage of centrally nucleated fibers. Vamorolone shows improved safety versus prednisolone by avoiding or reducing key side effects to behavior and growth. Intriguingly, vamorolone increases dystrophin protein in both heart and skeletal muscle. These data indicate that vamorolone, nearing approval for Duchenne, shows efficacy in bmx mice and therefore warrants clinical investigation in BMD.
Collapse
Affiliation(s)
- Nikki M. McCormack
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Nhu Y. Nguyen
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Christopher B. Tully
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Trinitee Oliver
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
- Department of Biology, Howard University, Washington, DC, USA
| | - Alyson A. Fiorillo
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC, USA
| | - Christopher R. Heier
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC, USA
| |
Collapse
|
26
|
Song Y, Xu HY, Xu K, Guo YK, Xie LJ, Peng F, Xu R, Fu H, Yuan WF, Zhou ZQ, Cheng BC, Fu C, Zhou H, Cai XT, Li XS. Clinical utilisation of multimodal quantitative magnetic resonance imaging in investigating muscular damage in Duchenne muscular dystrophy: a study on the association between gluteal muscle groups and motor function. Pediatr Radiol 2023; 53:1648-1658. [PMID: 36892624 PMCID: PMC10359373 DOI: 10.1007/s00247-023-05632-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/03/2023] [Accepted: 02/13/2023] [Indexed: 03/10/2023]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a neuromuscular disease characterised by progressive muscular weakness and atrophy. Currently, studies on DMD muscle function mostly focus on individual muscles; little is known regarding the effect of gluteal muscle group damage on motor function. OBJECTIVE To explore potential imaging biomarkers of hip and pelvic muscle groups for measuring muscular fat replacement and inflammatory oedema in DMD with multimodal quantitative magnetic resonance imaging (MRI). MATERIALS AND METHODS One hundred fifty-nine DMD boys and 32 healthy male controls were prospectively included. All subjects underwent MRI examination of the hip and pelvic muscles with T1 mapping, T2 mapping and Dixon sequences. Quantitatively measured parameters included longitudinal relaxation time (T1), transverse relaxation time (T2) and fat fraction. Investigations were all based on hip and pelvic muscle groups covering flexors, extensors, adductors and abductors. The North Star Ambulatory Assessment and stair climbing tests were used to measure motor function in DMD. RESULTS T1 of the extensors (r = 0.720, P < 0.01), flexors (r = 0.558, P < 0.01) and abductors (r = 0.697, P < 0.001) were positively correlated with the North Star Ambulatory Assessment score. In contrast, T2 of the adductors (r = -0.711, P < 0.01) and fat fraction of the extensors (r = -0.753, P < 0.01) were negatively correlated with the North Star Ambulatory Assessment score. Among them, T1 of the abductors (b = 0.013, t = 2.052, P = 0.042), T2 of the adductors (b = -0.234, t = -2.554, P = 0.012) and fat fraction of the extensors (b = -0.637, t = - 4.096, P < 0.001) significantly affected the North Star Ambulatory Assessment score. Moreover, T1 of the abductors was highly predictive for identifying motor dysfunction in DMD, with an area under the curve of 0.925. CONCLUSION Magnetic resonance biomarkers of hip and pelvic muscle groups (particularly T1 values of the abductor muscles) have the potential to be used as independent risk factors for motor dysfunction in DMD.
Collapse
Affiliation(s)
- Yu Song
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hua-Yan Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ke Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying-Kun Guo
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Lin-Jun Xie
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Fei Peng
- Department of Radiology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Rong Xu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hang Fu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei-Feng Yuan
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Zi-Qi Zhou
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Bo-Chao Cheng
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuan Fu
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hui Zhou
- Department of Rehabilitation, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xiao-Tang Cai
- Department of Rehabilitation, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xue-Sheng Li
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
27
|
Bello L, Hoffman EP, Pegoraro E. Is it time for genetic modifiers to predict prognosis in Duchenne muscular dystrophy? Nat Rev Neurol 2023; 19:410-423. [PMID: 37308617 DOI: 10.1038/s41582-023-00823-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/14/2023]
Abstract
Patients with Duchenne muscular dystrophy (DMD) show clinically relevant phenotypic variability, despite sharing the same primary biochemical defect (dystrophin deficiency). Factors contributing to this clinical variability include allelic heterogeneity (specific DMD mutations), genetic modifiers (trans-acting genetic polymorphisms) and variations in clinical care. Recently, a series of genetic modifiers have been identified, mostly involving genes and/or proteins that regulate inflammation and fibrosis - processes increasingly recognized as being causally linked with physical disability. This article reviews genetic modifier studies in DMD to date and discusses the effect of genetic modifiers on predicting disease trajectories (prognosis), clinical trial design and interpretation (inclusion of genotype-stratified subgroup analyses) and therapeutic approaches. The genetic modifiers identified to date underscore the importance of progressive fibrosis, downstream of dystrophin deficiency, in driving the disease process. As such, genetic modifiers have shown the importance of therapies aimed at slowing this fibrotic process and might point to key drug targets.
Collapse
Affiliation(s)
- Luca Bello
- Department of Neurosciences (DNS), University of Padova, Padova, Italy
| | - Eric P Hoffman
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University (State University of New York), Binghamton, NY, USA
| | - Elena Pegoraro
- Department of Neurosciences (DNS), University of Padova, Padova, Italy.
| |
Collapse
|
28
|
Zygmunt AM, Wong BL, Horn PS, Lambert J, Bange JE, Rybalsky I, Chouteau W, Tian C. A longitudinal study of creatine kinase and creatinine levels in Duchenne muscular dystrophy. Muscle Nerve 2023; 67:138-145. [PMID: 36444146 DOI: 10.1002/mus.27760] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 11/11/2022] [Accepted: 11/21/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION/AIMS Management of Duchenne muscular dystrophy (DMD) has entered an era featuring novel treatments. Trackable noninvasive biomarkers could improve disease progression monitoring and drug effect detection. Our aim in this study was to measure changes in selected noninvasive biomarkers and assess their relationship to age and motor function. METHODS We retrospectively studied 555 patients with DMD who had at least 12 months of treatment of glucocorticoids and were not enrolled in trials of potential disease-modifying therapies. We extracted biomarker data of serum creatine kinase (CK), serum creatinine (Cr), urine Cr, and urine Cr/urine osmolality (osm), as well as functional data for age at loss of ambulation and Functional Motor Scale (FMS) values from patients' clinical records. Data were analyzed using linear mixed-model analyses. RESULTS CK, serum Cr, urine Cr, and urine Cr/urine osm all decreased with declining motor function. CK consistently decreased and FMS score consistently worsened with age without clear inflection points. There was an increased odds ratio for LOA with lower values of CK, serum Cr, urine Cr, and urine Cr/urine osm, most notably for urine Cr. DISCUSSION Although individual biomarker values are challenging to directly apply clinically, our study has demonstrated that trends over time may complement functional measures in the assessment of individuals with DMD. Future studies could elucidate predictive utility of these biomarkers in assessing motor function changes in DMD.
Collapse
Affiliation(s)
- Alexander M Zygmunt
- Division of Neurology, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Brenda L Wong
- Department of Pediatrics, University of Massachusetts Chan Medical School, Worcester, Massachusetts
| | - Paul S Horn
- Division of Neurology, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joshua Lambert
- Department of Psychology, University of Alabama, Tuscaloosa, Alabama
| | - Jean E Bange
- Division of Neurology, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Irina Rybalsky
- Division of Neurology, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Wendy Chouteau
- Division of Neurology, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Cuixia Tian
- Division of Neurology, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
29
|
Comi GP, Niks EH, Vandenborne K, Cinnante CM, Kan HE, Willcocks RJ, Velardo D, Magri F, Ripolone M, van Benthem JJ, van de Velde NM, Nava S, Ambrosoli L, Cazzaniga S, Bettica PU. Givinostat for Becker muscular dystrophy: A randomized, placebo-controlled, double-blind study. Front Neurol 2023; 14:1095121. [PMID: 36793492 PMCID: PMC9923355 DOI: 10.3389/fneur.2023.1095121] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/09/2023] [Indexed: 02/03/2023] Open
Abstract
Objective No treatments are approved for Becker muscular dystrophy (BMD). This study investigated the efficacy and safety of givinostat, a histone deacetylase pan-inhibitor, in adults with BMD. Methods Males aged 18-65 years with a diagnosis of BMD confirmed by genetic testing were randomized 2:1 to 12 months treatment with givinostat or placebo. The primary objective was to demonstrate statistical superiority of givinostat over placebo for mean change from baseline in total fibrosis after 12 months. Secondary efficacy endpoints included other histological parameters, magnetic resonance imaging and spectroscopy (MRI and MRS) measures, and functional evaluations. Results Of 51 patients enrolled, 44 completed treatment. At baseline, there was greater disease involvement in the placebo group than givinostat, based on total fibrosis (mean 30.8 vs. 22.8%) and functional endpoints. Mean total fibrosis did not change from baseline in either group, and the two groups did not differ at Month 12 (least squares mean [LSM] difference 1.04%; p = 0.8282). Secondary histology parameters, MRS, and functional evaluations were consistent with the primary. MRI fat fraction in whole thigh and quadriceps did not change from baseline in the givinostat group, but values increased with placebo, with LSM givinostat-placebo differences at Month 12 of -1.35% (p = 0.0149) and -1.96% (p = 0.0022), respectively. Adverse events, most mild or moderate, were reported by 88.2% and 52.9% patients receiving givinostat and placebo. Conclusion The study failed to achieve the primary endpoint. However, there was a potential signal from the MRI assessments suggesting givinostat could prevent (or slow down) BMD disease progression.
Collapse
Affiliation(s)
- Giacomo P. Comi
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy,Department of Pathophysiology and Transplantation, Dino Ferrari Center, University of Milan, Milan, Italy,*Correspondence: Giacomo P. Comi ✉
| | - Erik H. Niks
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands,Duchenne Center Netherlands, Netherlands
| | | | | | - Hermien E. Kan
- Duchenne Center Netherlands, Netherlands,Department of Radiology, C.J. Gorter MRI Center, Leiden University Medical Center, Leiden, Netherlands
| | | | - Daniele Velardo
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesca Magri
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Michela Ripolone
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Jules J. van Benthem
- Department of Orthopedics, Rehabilitation and Physiotherapy, Leiden University Medical Center, Leiden, Netherlands
| | - Nienke M. van de Velde
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands,Duchenne Center Netherlands, Netherlands
| | - Simone Nava
- Radiology Department, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | | | | | | |
Collapse
|
30
|
Vegezzi E, Cortese A, Bergsland N, Mussinelli R, Paoletti M, Solazzo F, Currò R, Ascagni L, Callegari I, Quartesan I, Lozza A, Deligianni X, Santini F, Marchioni E, Cosentino G, Alfonsi E, Tassorelli C, Bastianello S, Merlini G, Palladini G, Obici L, Pichiecchio A. Muscle quantitative MRI as a novel biomarker in hereditary transthyretin amyloidosis with polyneuropathy: a cross-sectional study. J Neurol 2023; 270:328-339. [PMID: 36064814 DOI: 10.1007/s00415-022-11336-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND The development of reproducible and sensitive outcome measures has been challenging in hereditary transthyretin (ATTRv) amyloidosis. Recently, quantification of intramuscular fat by magnetic resonance imaging (MRI) has proven as a sensitive marker in patients with other genetic neuropathies. The aim of this study was to investigate the role of muscle quantitative MRI (qMRI) as an outcome measure in ATTRv. METHODS Calf- and thigh-centered multi-echo T2-weighted spin-echo and gradient-echo sequences were obtained in patients with ATTRv amyloidosis with polyneuropathy (n = 24) and healthy controls (n = 12). Water T2 (wT2) and fat fraction (FF) were calculated. Neurological assessment was performed in all ATTRv subjects. Quantitative MRI parameters were correlated with clinical and neurophysiological measures of disease severity. RESULTS Quantitative imaging revealed significantly higher FF in lower limb muscles in patients with ATTRv amyloidosis compared to controls. In addition, wT2 was significantly higher in ATTRv patients. There was prominent involvement of the posterior compartment of the thighs. Noticeably, FF and wT2 did not exhibit a length-dependent pattern in ATTRv patients. MRI biomarkers correlated with previously validated clinical outcome measures, Polyneuropathy Disability scoring system, Neuropathy Impairment Score (NIS) and NIS-lower limb, and neurophysiological parameters of axonal damage regardless of age, sex, treatment and TTR mutation. CONCLUSIONS Muscle qMRI revealed significant difference between ATTRv and healthy controls. MRI biomarkers showed high correlation with clinical and neurophysiological measures of disease severity making qMRI as a promising tool to be further investigated in longitudinal studies to assess its role at monitoring onset, progression, and therapy efficacy for future clinical trials on this treatable condition.
Collapse
Affiliation(s)
- Elisa Vegezzi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Neuroncology and Neuroinflammation Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Andrea Cortese
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy. .,Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK.
| | - Niels Bergsland
- Department of Neurology, Buffalo Neuroimaging Analysis Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.,IRCCS Fondazione Don Carlo Gnocchi ONLUS, Milan, Italy
| | - Roberta Mussinelli
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Matteo Paoletti
- Neuroradiology Department, Advanced Imaging and Radiomics Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Francesca Solazzo
- Specialization School in Occupational Medicine, University of Pavia, Pavia, Italy
| | - Riccardo Currò
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Lucia Ascagni
- Neuroscience Department, Meyer Children's University Hospital, University of Florence, Florence, Italy
| | - Ilaria Callegari
- Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, 4031, Basel, Switzerland
| | - Ilaria Quartesan
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Alessandro Lozza
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Xeni Deligianni
- Division of Radiological Physics, Department of Radiology, University Hospital Basel, Basel, Switzerland.,Department of Biomedical Engineering, Basel Muscle MRI Group, University of Basel, Allschwil, Switzerland
| | - Francesco Santini
- Division of Radiological Physics, Department of Radiology, University Hospital Basel, Basel, Switzerland.,Department of Biomedical Engineering, Basel Muscle MRI Group, University of Basel, Allschwil, Switzerland
| | - Enrico Marchioni
- Neuroncology and Neuroinflammation Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Giuseppe Cosentino
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Translational Neurophysiology Research Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Enrico Alfonsi
- Translational Neurophysiology Research Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Cristina Tassorelli
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Headache Science and Neurorehabilitation Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Stefano Bastianello
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Neuroradiology Department, Advanced Imaging and Radiomics Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Giampaolo Merlini
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy.,Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Giovanni Palladini
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy.,Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Laura Obici
- Amyloidosis Research and Treatment Center, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Anna Pichiecchio
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy.,Neuroradiology Department, Advanced Imaging and Radiomics Center, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
31
|
Liu J, Barrett JS, Leonardi ET, Lee L, Roychoudhury S, Chen Y, Trifillis P. Natural History and Real-World Data in Rare Diseases: Applications, Limitations, and Future Perspectives. J Clin Pharmacol 2022; 62 Suppl 2:S38-S55. [PMID: 36461748 PMCID: PMC10107901 DOI: 10.1002/jcph.2134] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/28/2022] [Indexed: 12/04/2022]
Abstract
Rare diseases represent a highly heterogeneous group of disorders with high phenotypic and genotypic diversity within individual conditions. Due to the small numbers of people affected, there are unique challenges in understanding rare diseases and drug development for these conditions, including patient identification and recruitment, trial design, and costs. Natural history data and real-world data (RWD) play significant roles in defining and characterizing disease progression, final patient populations, novel biomarkers, genetic relationships, and treatment effects. This review provides an introduction to rare diseases, natural history data, RWD, and real-world evidence, the respective sources and applications of these data in several rare diseases. Considerations for data quality and limitations when using natural history and RWD are also elaborated. Opportunities are highlighted for cross-sector collaboration, standardized and high-quality data collection using new technologies, and more comprehensive evidence generation using quantitative approaches such as disease progression modeling, artificial intelligence, and machine learning. Advanced statistical approaches to integrate natural history data and RWD to further disease understanding and guide more efficient clinical study design and data analysis in drug development in rare diseases are also discussed.
Collapse
Affiliation(s)
- Jing Liu
- Pfizer, Inc., Groton, Connecticut, USA
| | - Jeffrey S Barrett
- Critical Path Institute, Rare Disease Cures Accelerator Data Analytics Platform, Tucson, Arizona, USA
| | | | - Lucy Lee
- PTC Therapeutics, Inc., South Plainfield, New Jersey, USA
| | | | - Yong Chen
- Pfizer, Inc., Groton, Connecticut, USA
| | | |
Collapse
|
32
|
Moore U, Caldas de Almeida Araújo E, Reyngoudt H, Gordish‐Dressman H, Smith FE, Wilson I, James M, Mayhew A, Rufibach L, Day JW, Jones KJ, Bharucha‐Goebel DX, Salort‐Campana E, Pestronk A, Walter MC, Paradas C, Stojkovic T, Mori‐Yoshimura M, Bravver E, Pegoraro E, Mendell JR, Bushby K, Blamire AM, Straub V, Carlier PG, Diaz‐Manera J. Water T2 could predict functional decline in patients with dysferlinopathy. J Cachexia Sarcopenia Muscle 2022; 13:2888-2897. [PMID: 36058852 PMCID: PMC9745487 DOI: 10.1002/jcsm.13063] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/18/2022] [Accepted: 07/04/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Water T2 (T2H2O ) mapping is increasingly being used in muscular dystrophies to assess active muscle damage. It has been suggested as a surrogate outcome measure for clinical trials. Here, we investigated the prognostic utility of T2H2O to identify changes in muscle function over time in limb girdle muscular dystrophies. METHODS Patients with genetically confirmed dysferlinopathy were assessed as part of the Jain Foundation Clinical Outcomes Study in dysferlinopathy. The cohort included 18 patients from two sites, both equipped with 3-tesla magnetic resonance imaging (MRI) systems from the same vendor. T2H2O value was defined as higher or lower than the median in each muscle bilaterally. The degree of deterioration on four functional tests over 3 years was assessed in a linear model against covariates of high or low T2H2O at baseline, age, disease duration, and baseline function. RESULTS A higher T2H2O at baseline significantly correlated with a greater decline on functional tests in 21 out of 35 muscles and was never associated with slower decline. Higher baseline T2H2O in adductor magnus, vastus intermedius, vastus lateralis, and vastus medialis were the most sensitive, being associated bilaterally with greater decline in multiple timed tests. Patients with a higher than median baseline T2H2O (>40.6 ms) in the right vastus medialis deteriorated 11 points more on the North Star Ambulatory Assessment for Dysferlinopathy and lost an additional 86 m on the 6-min walk than those with a lower T2H2O (<40.6 ms). Optimum sensitivity and specificity thresholds for predicting decline were 39.0 ms in adductor magnus and vastus intermedius, 40.0 ms in vastus medialis, and 40.5 ms in vastus lateralis from different sites equipped with different MRI systems. CONCLUSIONS In dysferlinopathy, T2H2O did not correlate with current functional ability. However, T2H2O at baseline was higher in patients who worsened more rapidly on functional tests. This suggests that inter-patient differences in functional decline over time may be, in part, explained by different severities of the active muscle damage, assessed by T2H2O measure at baseline. Significant challenges remain in standardizing T2H2O values across sites to allow determining globally applicable thresholds. The results from the present work are encouraging and suggest that T2H2O could be used to improve prognostication, patient selection, and disease modelling for clinical trials.
Collapse
Affiliation(s)
- Ursula Moore
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research InstituteNewcastle University and Newcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Ericky Caldas de Almeida Araújo
- NMR Laboratory, Neuromuscular Investigation CenterInstitute of MyologyParisFrance
- NMR LaboratoryCEA/DRF/IBFJ/MIRCenParisFrance
| | - Harmen Reyngoudt
- NMR Laboratory, Neuromuscular Investigation CenterInstitute of MyologyParisFrance
- NMR LaboratoryCEA/DRF/IBFJ/MIRCenParisFrance
| | - Heather Gordish‐Dressman
- Center for Translational Science, Division of Biostatistics and Study MethodologyChildren's National Health SystemWashingtonDCUSA
- Pediatrics, Epidemiology and BiostatisticsGeorge Washington UniversityWashingtonDCUSA
| | - Fiona E. Smith
- Magnetic Resonance Centre, Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Ian Wilson
- Magnetic Resonance Centre, Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Meredith James
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research InstituteNewcastle University and Newcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Anna Mayhew
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research InstituteNewcastle University and Newcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | | | - John W. Day
- Department of Neurology and Neurological SciencesStanford University School of MedicineStanfordCAUSA
| | - Kristi J. Jones
- The Children's Hospital at Westmead and The University of SydneySydneyNSWAustralia
| | - Diana X. Bharucha‐Goebel
- Department of NeurologyChildren's National Health SystemWashingtonDCUSA
- National Institutes of Health (NINDS)BethesdaMDUSA
| | | | - Alan Pestronk
- Department of NeurologyWashington University School of MedicineSt. LouisMOUSA
| | - Maggie C. Walter
- Friedrich‐Baur‐Institute, Department of NeurologyLudwig‐Maximilians‐University of MunichMunichGermany
| | - Carmen Paradas
- Neuromuscular Unit, Department of NeurologyHospital U. Virgen del Rocío/Instituto de Biomedicina de SevillaSevillaSpain
| | - Tanya Stojkovic
- Centre de référence des maladies neuromusculairesInstitut de Myologie, AP‐HP, Sorbonne Université, Hôpital Pitié‐SalpêtrièreParisFrance
| | - Madoka Mori‐Yoshimura
- Department of NeurologyNational Center Hospital, National Center of Neurology and PsychiatryTokyoJapan
| | - Elena Bravver
- Neuroscience InstituteCarolinas Neuromuscular/ALS‐MDA Center, Carolinas HealthCare SystemCharlotteNCUSA
| | - Elena Pegoraro
- Department of NeuroscienceUniversity of PadovaPaduaItaly
| | - Jerry R. Mendell
- The Abigail Wexner Research Institute at Nationwide Children's HospitalColumbusOHUSA
| | | | - Kate Bushby
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research InstituteNewcastle University and Newcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Andrew M. Blamire
- Magnetic Resonance Centre, Translational and Clinical Research InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research InstituteNewcastle University and Newcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Pierre G. Carlier
- Université Paris‐Saclay, CEA, DRF, Service Hospitalier Frederic JoliotOrsayFrance
| | - Jordi Diaz‐Manera
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research InstituteNewcastle University and Newcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
- Neuromuscular Disorders Unit, Neurology DepartmentHospital de la Santa Creu i Sant PauBarcelonaSpain
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER)MadridSpain
| |
Collapse
|
33
|
Barnard AM, Hammers DW, Triplett WT, Kim S, Forbes SC, Willcocks RJ, Daniels MJ, Senesac CR, Lott DJ, Arpan I, Rooney WD, Wang RT, Nelson SF, Sweeney HL, Vandenborne K, Walter GA. Evaluating Genetic Modifiers of Duchenne Muscular Dystrophy Disease Progression Using Modeling and MRI. Neurology 2022; 99:e2406-e2416. [PMID: 36240102 PMCID: PMC9687406 DOI: 10.1212/wnl.0000000000201163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/11/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Duchenne muscular dystrophy (DMD) is a progressive muscle degenerative disorder with a well-characterized disease phenotype but considerable interindividual heterogeneity that is not well understood. The aim of this study was to evaluate the effects of dystrophin variations and genetic modifiers of DMD on rate and age of muscle replacement by fat. METHODS One hundred seventy-five corticosteroid treated participants from the ImagingDMD natural history study underwent repeated magnetic resonance spectroscopy (MRS) of the vastus lateralis (VL) and soleus (SOL) to determine muscle fat fraction (FF). MRS was performed annually in most instances; however, some individuals had additional visits at 3 or 6 monthss intervals. FF changes over time were modeled using nonlinear mixed effects to estimate disease trajectories based on the age that the VL or SOL reached half-maximum change in FF (mu) and the time required for FF change (sigma). Computed mu and sigma values were evaluated for dystrophin variations that have demonstrated the ability to lead to a mild phenotype as well as compared between different genetic polymorphism groups. RESULTS Participants with dystrophin gene deletions amenable to exon 8 skipping (n = 4) had minimal increases in SOL FF and had an increase in VL mu value by 4.4 years compared with a reference cohort (p = 0.039). Participants with nonsense variations within exons that may produce milder phenotypes (n = 11) also had minimal increases in SOL and VL FFs. No differences in estimated FF trajectories were seen for individuals amenable to exon 44 skipping (n = 10). Modeling of the SPP1, LTBP4, and thrombospondin-1 (THBS1) genetic modifiers did not result in significant differences in muscle FF trajectories between genotype groups (p > 0.05); however, trends were noted for the polymorphisms associated with long-range regulation of LTBP4 and THBS1 that deserve further follow-up. DISCUSSION The results of this study link the historically mild phenotypes seen in individuals amenable to exon 8 skipping and with certain nonsense variations with alterations in trajectories of lower extremity muscle replacement by fat.
Collapse
Affiliation(s)
- Alison M Barnard
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - David W Hammers
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - William T Triplett
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Sarah Kim
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Sean C Forbes
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Rebecca J Willcocks
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Michael J Daniels
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Claudia R Senesac
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Donovan J Lott
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Ishu Arpan
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - William D Rooney
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Richard T Wang
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Stanley F Nelson
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - H Lee Sweeney
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Krista Vandenborne
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville
| | - Glenn A Walter
- From the Department of Physical Therapy (A.M.B., W.T.T., S.C.F., R.J.W., C.R.S., D.J.L., K.V.) Pharmacology and Therapeutics (D.W.H., H.L.S.), University of Florida, Gainesville; Center for Pharmacometrics and Systems Pharmacology (S.K.), Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando; Department of Statistics (M.J.D.), University of Florida, Gainesville; Department of Neurology (I.A.), Oregon Health & Science University, Portland; Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; Department of Human Genetics (R.T.W., S.F.N.), University of California Los Angeles, CA; and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville.
| |
Collapse
|
34
|
Samani A, Hightower RM, Reid AL, English KG, Lopez MA, Doyle JS, Conklin MJ, Schneider DA, Bamman MM, Widrick JJ, Crossman DK, Xie M, Jee D, Lai EC, Alexander MS. miR-486 is essential for muscle function and suppresses a dystrophic transcriptome. Life Sci Alliance 2022; 5:e202101215. [PMID: 35512829 PMCID: PMC9087951 DOI: 10.26508/lsa.202101215] [Citation(s) in RCA: 8] [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: 08/25/2021] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 02/02/2023] Open
Abstract
miR-486 is a muscle-enriched microRNA, or "myomiR," that has reduced expression correlated with Duchenne muscular dystrophy (DMD). To determine the function of miR-486 in normal and dystrophin-deficient muscles and elucidate miR-486 target transcripts in skeletal muscle, we characterized mir-486 knockout mice (mir-486 KO). mir-486 KO mice developed disrupted myofiber architecture, decreased myofiber size, decreased locomotor activity, increased cardiac fibrosis, and metabolic defects were exacerbated in mir-486 KO:mdx 5cv (DKO) mice. To identify direct in vivo miR-486 muscle target transcripts, we integrated RNA sequencing and chimeric miRNA eCLIP sequencing to identify key transcripts and pathways that contribute towards mir-486 KO and dystrophic disease pathologies. These targets included known and novel muscle metabolic and dystrophic structural remodeling factors of muscle and skeletal muscle contractile transcript targets. Together, our studies identify miR-486 as essential for normal muscle function, a driver of pathological remodeling in dystrophin-deficient muscle, a useful biomarker for dystrophic disease progression, and highlight the use of multiple omic platforms to identify in vivo microRNA target transcripts.
Collapse
Affiliation(s)
- Adrienne Samani
- Department of Pediatrics, Division of Neurology at Children's of Alabama and the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rylie M Hightower
- Department of Pediatrics, Division of Neurology at Children's of Alabama and the University of Alabama at Birmingham, Birmingham, AL, USA
- University of Alabama at Birmingham Center for Exercise Medicine (UCEM), Birmingham, AL, USA
| | - Andrea L Reid
- Department of Pediatrics, Division of Neurology at Children's of Alabama and the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Katherine G English
- Department of Pediatrics, Division of Neurology at Children's of Alabama and the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael A Lopez
- Department of Pediatrics, Division of Neurology at Children's of Alabama and the University of Alabama at Birmingham, Birmingham, AL, USA
- University of Alabama at Birmingham Center for Exercise Medicine (UCEM), Birmingham, AL, USA
| | - J Scott Doyle
- Department of Orthopedic Surgery, at the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael J Conklin
- Department of Orthopedic Surgery, at the University of Alabama at Birmingham, Birmingham, AL, USA
| | - David A Schneider
- Department of Biochemistry and Molecular Genetics at the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Marcas M Bamman
- University of Alabama at Birmingham Center for Exercise Medicine (UCEM), Birmingham, AL, USA
| | - Jeffrey J Widrick
- Division of Genetics and Genomics at Boston Children's Hospital, Boston, MA, USA
| | - David K Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Min Xie
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, School of Medicine, Birmingham, AL, USA
| | - David Jee
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
- Weill Graduate School of Medical Sciences, Cornell University, New York, NY, USA
| | - Eric C Lai
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
- Weill Graduate School of Medical Sciences, Cornell University, New York, NY, USA
| | - Matthew S Alexander
- Department of Pediatrics, Division of Neurology at Children's of Alabama and the University of Alabama at Birmingham, Birmingham, AL, USA
- University of Alabama at Birmingham Center for Exercise Medicine (UCEM), Birmingham, AL, USA
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
- UAB Civitan International Research Center (CIRC), at the University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
35
|
Veeger TTJ, van de Velde NM, Keene KR, Niks EH, Hooijmans MT, Webb AG, de Groot JH, Kan HE. Baseline fat fraction is a strong predictor of disease progression in Becker muscular dystrophy. NMR IN BIOMEDICINE 2022; 35:e4691. [PMID: 35032073 PMCID: PMC9286612 DOI: 10.1002/nbm.4691] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/24/2021] [Accepted: 01/10/2022] [Indexed: 06/14/2023]
Abstract
In Becker muscular dystrophy (BMD), muscle weakness progresses relatively slowly, with a highly variable rate among patients. This complicates clinical trials, as clinically relevant changes are difficult to capture within the typical duration of a trial. Therefore, predictors for disease progression are needed. We assessed if temporal increase of fat fraction (FF) in BMD follows a sigmoidal trajectory and whether fat fraction at baseline (FFbase) could therefore predict FF increase after 2 years (ΔFF). Thereafter, for two different MR-based parameters, we tested the additional predictive value to FFbase. We used 3-T Dixon data from the upper and lower leg, and multiecho spin-echo MRI and 7-T 31 P MRS datasets from the lower leg, acquired in 24 BMD patients (age: 41.4 [SD 12.8] years). We assessed the pattern of increase in FF using mixed-effects modelling. Subsequently, we tested if indicators of muscle damage like standard deviation in water T2 (stdT2 ) and the phosphodiester (PDE) over ATP ratio at baseline had additional value to FFbase for predicting ∆FF. The association between FFbase and ΔFF was described by the derivative of a sigmoid function and resulted in a peak ΔFF around 0.45 FFbase (fourth-order polynomial term: t = 3.7, p < .001). StdT2 and PDE/ATP were not significantly associated with ∆FF if FFbase was included in the model. The relationship between FFbase and ∆FF suggests a sigmoidal trajectory of the increase in FF over time in BMD, similar to that described for Duchenne muscular dystrophy. Our results can be used to identify muscles (or patients) that are in the fast progressing stage of the disease, thereby facilitating the conduct of clinical trials.
Collapse
Affiliation(s)
- Thom T. J. Veeger
- C. J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical Center (LUMC)LeidenThe Netherlands
| | - Nienke M. van de Velde
- Department of Neurology, Leiden University Medical Center (LUMC)LeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| | - Kevin R. Keene
- Department of Neurology, Leiden University Medical Center (LUMC)LeidenThe Netherlands
| | - Erik H. Niks
- Department of Neurology, Leiden University Medical Center (LUMC)LeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| | - Melissa T. Hooijmans
- Department of Radiology & Nuclear MedicineAmsterdam University Medical CentersAmsterdamThe Netherlands
| | - Andrew G. Webb
- C. J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical Center (LUMC)LeidenThe Netherlands
| | - Jurriaan H. de Groot
- Department of Rehabilitation Medicine, Leiden University Medical Center (LUMC)LeidenThe Netherlands
| | - Hermien E. Kan
- C. J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical Center (LUMC)LeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| |
Collapse
|
36
|
Suslov V, Suslova G, Lytaev S. MRI Assessment of Motor Capabilities in Patients with Duchenne Muscular Dystrophy According to the Motor Function Measure Scale. Tomography 2022; 8:948-960. [PMID: 35448710 PMCID: PMC9025497 DOI: 10.3390/tomography8020076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 11/16/2022] Open
Abstract
The research was aimed on the study of motor capabilities on the Motor Function Measure (MFM) scale in ambulant and non-ambulant patients with Duchenne muscular dystrophy, and to conduct a correlation analysis between the results of the MFM scale and Magnetic Resonance Imaging (MRI) data. A total of 46 boys who had genetically confirmed Duchenne muscular dystrophy (age from 2.1 to 16.7 years) and were in clinical rehabilitation were investigated. An assessment was performed according to the Motor Function Measure scale (subsections D1, D2, D3, and the total score), an MRI obtaining T1-VI of the muscles of the pelvic girdle was conducted, and the thighs and lower legs were further assessed in terms of the severity of fibrous-fat degeneration according to the Mercuri scale. In ambulant patients, the ability to stand up and move (D1) was 74.4%, axial and proximal motor functions (D2)—97.6%, distal motor functions (D3)—96.2%, and total score was 87.9%. In non-ambulant patients, the ability to stand up and move (D1) was 1.7%, axial and proximal motor functions (D2)—47%, distal motor functions (D3)—67.5%, and the total score—33.1%. A high inverse correlation (r = −0.7, p < 0.05) of the MRI data of the pelvic girdle and thighs with tasks D1, as well as a noticeable inverse correlation with tasks D2 (r = −0.6, p < 0.05) of the scale MFM, were revealed in the ambulant group of patients. In the non-ambulant group of patients, the MRI data of the lower legs muscles were characterized by a high inverse correlation (r = −0.7, p < 0.05) with tasks D3 and a noticeable inverse correlation (r = −0.6, p < 0.05) with tasks D1 of the MFM scale. Conclusion: The Motor Function Measure scale allows effective assessment of the motor capabilities of patients with Duchenne muscular dystrophy at different stages of the disease, which is confirmed by visualization of fibro-fatty muscle replacement.
Collapse
Affiliation(s)
- Vasily Suslov
- Department of Rehabilitation, Saint Petersburg State Pediatric Medical University, 194100 Saint Petersburg, Russia;
- Correspondence: ; Tel.: +7-911-2297049
| | - Galina Suslova
- Department of Rehabilitation, Saint Petersburg State Pediatric Medical University, 194100 Saint Petersburg, Russia;
| | - Sergey Lytaev
- Department of Normal Physiology, Saint Petersburg State Pediatric Medical University, 194100 Saint Petersburg, Russia;
| |
Collapse
|
37
|
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
|
38
|
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
|
39
|
Barnard AM, Lott DJ, Batra A, Triplett WT, Willcocks RJ, Forbes SC, Rooney WD, Daniels MJ, Smith BK, Vandenborne K, Walter GA. Characterizing Expiratory Respiratory Muscle Degeneration in Duchenne Muscular Dystrophy Using MRI. Chest 2022; 161:753-763. [PMID: 34536384 PMCID: PMC9160975 DOI: 10.1016/j.chest.2021.08.078] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022] Open
Abstract
BACKGROUND Expiratory muscle weakness and impaired airway clearance are early signs of respiratory dysfunction in Duchenne muscular dystrophy (DMD), a degenerative muscle disorder in which muscle cells are damaged and replaced by fibrofatty tissue. Little is known about expiratory muscle pathology and its relationship to cough and airway clearance capacity; however, the level of muscle replacement by fat can be estimated using MRI and expressed as a fat fraction (FF). RESEARCH QUESTION How does abdominal expiratory muscle fatty infiltration change over time in DMD and relate to clinical expiratory function? STUDY DESIGN AND METHODS Individuals with DMD underwent longitudinal MRI of the abdomen to determine FF in the internal oblique, external oblique, and rectus abdominis expiratory muscles. FF data were used to estimate a model of expiratory muscle degeneration by using nonlinear mixed effects and a cumulative distribution function. FVC, maximal inspiratory and expiratory pressures, and peak cough flow were collected as clinical correlates to MRI. RESULTS Forty individuals with DMD (aged 6-18 years at baseline) participated in up to five visits over 36 months. Modeling estimated the internal oblique progresses most quickly and reached 50% replacement by fat at a mean patient age of 13.0 years (external oblique, 14.0 years; rectus abdominis, 16.2 years). Corticosteroid-untreated individuals (n = 4) reached 50% muscle replacement by fat 3 to 4 years prior to treated individuals. Individuals with mild clinical dystrophic phenotypes (n = 3) reached 50% muscle replacement by fat 4 to 5 years later than corticosteroid-treated individuals. Internal and external oblique FFs near 50% were associated with maximal expiratory pressures < 60 cm H2O and peak cough flows < 270 L/min. INTERPRETATION These data improve understanding of the early phase of respiratory compromise in DMD, which typically presents as airway clearance dysfunction prior to the onset of hypoventilation, and links expiratory muscle fatty infiltration to pulmonary function measures.
Collapse
Affiliation(s)
- Alison M Barnard
- Department of Physical Therapy, University of Florida, Gainesville, FL
| | - Donovan J Lott
- Department of Physical Therapy, University of Florida, Gainesville, FL
| | - Abhinandan Batra
- Department of Physical Therapy, University of Florida, Gainesville, FL
| | | | | | - Sean C Forbes
- Department of Physical Therapy, University of Florida, Gainesville, FL
| | - William D Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR
| | | | - Barbara K Smith
- Department of Physical Therapy, University of Florida, Gainesville, FL
| | | | - Glenn A Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL.
| |
Collapse
|
40
|
Rebecca JW, Alison MB, Ryan JW, Claudia RS, Donovan JL, Ann TH, Kirsten LZ, Sean CF, William DR, Dah-Jyuu W, Erika LF, Gihan IT, Michael JD, William TT, Glenn AW, Krista V. Development of Contractures in DMD in Relation to MRI-Determined Muscle Quality and Ambulatory Function. J Neuromuscul Dis 2022; 9:289-302. [PMID: 35124659 DOI: 10.3233/jnd-210731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Joint contractures are common in boys and men with Duchenne muscular dystrophy (DMD), and management of contractures is an important part of care. The optimal methods to prevent and treat contractures are controversial, and the natural history of contracture development is understudied in glucocorticoid treated individuals at joints beyond the ankle. OBJECTIVE To describe the development of contractures over time in a large cohort of individuals with DMD in relation to ambulatory ability, functional performance, and muscle quality measured using magnetic resonance imaging (MRI) and spectroscopy (MRS). METHODS In this longitudinal study, range of motion (ROM) was measured annually at the hip, knee, and ankle, and at the elbow, forearm, and wrist at a subset of visits. Ambulatory function (10 meter walk/run and 6 minute walk test) and MR-determined muscle quality (transverse relaxation time (T2) and fat fraction) were measured at each visit. RESULTS In 178 boys with DMD, contracture prevalence and severity increased with age. Among ambulatory participants, more severe contractures (defined as greater loss of ROM) were significantly associated with worse ambulatory function, and across all participants, more severe contractures significantly associated with higher MRI T2 or MRS FF (ρ: 0.40-0.61 in the lower extremity; 0.20-0.47 in the upper extremity). Agonist/antagonist differences in MRI T2 were not strong predictors of ROM. CONCLUSIONS Contracture severity increases with disease progression (increasing age and muscle involvement and decreasing functional ability), but is only moderately predicted by muscle fatty infiltration and MRI T2, suggesting that other changes in the muscle, tendon, or joint contribute meaningfully to contracture formation in DMD.
Collapse
Affiliation(s)
| | | | - J Wortman Ryan
- Tulane University School of Medicine, New Orleans, LA, USA
| | | | | | - T Harrington Ann
- Children's Hospital of Philadelphia, Philadelphia, PA, USA.,Arcadia University, Glennside, PA, USA
| | - L Zilke Kirsten
- Shriners Hospitals for Children -Portland, OR, USA.,Oregon Health and Science University, Portland, OR, USA
| | | | | | - Wang Dah-Jyuu
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Nair KS, Lott DJ, Forbes SC, Barnard AM, Willcocks RJ, Senesac CR, Daniels MJ, Harrington AT, Tennekoon GI, Zilke K, Finanger EL, Finkel RS, Rooney WD, Walter GA, Vandenborne K. Step Activity Monitoring in Boys with Duchenne Muscular Dystrophy and its Correlation with Magnetic Resonance Measures and Functional Performance. J Neuromuscul Dis 2022; 9:423-436. [PMID: 35466946 PMCID: PMC9257666 DOI: 10.3233/jnd-210746] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Muscles of boys with Duchenne muscular dystrophy (DMD) are progressively replaced by fatty fibrous tissues, and weakness leads to loss of ambulation (LoA). Step activity (SA) monitoring is a quantitative measure of real-world ambulatory function. The relationship between quality of muscle health and SA is unknown in DMD. OBJECTIVE To determine SA in steroid treated boys with DMD across various age groups, and to evaluate the association of SA with quality of muscle health and ambulatory function. METHODS Quality of muscle health was measured by magnetic resonance (MR) imaging transverse magnetization relaxation time constant (MRI-T2) and MR spectroscopy fat fraction (MRS-FF). SA was assessed via accelerometry, and functional abilities were assessed through clinical walking tests. Correlations between SA, MR, and functional measures were determined. A threshold value of SA was determined to predict the future LoA. RESULTS The greatest reduction in SA was observed in the 9- < 11years age group. SA correlated with all functional and MR measures.10m walk/run test had the highest correlation with SA. An increase in muscle MRI-T2 and MRS-FF was associated with a decline in SA. Two years prior to LoA, SA in boys with DMD was 32% lower than age matched boys with DMD who maintained ambulation for more than two-year period. SA monitoring can predict subsequent LoA in Duchenne, as a daily step count of 3200 at baseline was associated with LoA over the next two-years. CONCLUSION SA monitoring is a feasible and accessible tool to measure functional capacity in the real-world environment.
Collapse
Affiliation(s)
- Kavya S. Nair
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Donovan J. Lott
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Sean C. Forbes
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Alison M. Barnard
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Rebecca J. Willcocks
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Claudia R. Senesac
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Michael J. Daniels
- Department of Statistics, University of Florida, Gainesville, Florida, USA
| | - Ann T. Harrington
- Center for Rehabilitation, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Gihan I. Tennekoon
- Department of Neurology and Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kirsten Zilke
- Department of Pediatrics and Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | - Erika L. Finanger
- Department of Pediatrics and Neurology, Oregon Health & Science University, Portland, Oregon, USA
| | - Richard S. Finkel
- Center for Experimental Neurotherapeutics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - William D. Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Glenn A. Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
42
|
Comi GP, Niks EH, Cinnante CM, Kan HE, Vandenborne K, Willcocks RJ, Velardo D, Ripolone M, van Benthem JJ, van de Velde NM, Nava S, Ambrosoli L, Cazzaniga S, Bettica PU. Characterization of patients with Becker muscular dystrophy by histology, magnetic resonance imaging, function, and strength assessments. Muscle Nerve 2021; 65:326-333. [PMID: 34918368 PMCID: PMC9302983 DOI: 10.1002/mus.27475] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 12/07/2021] [Accepted: 12/11/2021] [Indexed: 12/21/2022]
Abstract
Introduction/Aims Becker muscular dystrophy (BMD) is characterized by variable disease severity and progression, prompting the identification of biomarkers for clinical trials. We used data from an ongoing phase II study to provide a comprehensive characterization of a cohort of patients with BMD, and to assess correlations between histological and magnetic resonance imaging (MRI) markers with muscle function and strength. Methods Eligible patients were ambulatory males with BMD, aged 18 to 65 years (200 to 450 meters on 6‐minute walk test). The following data were obtained: function test results, strength, fat‐fraction quantification using chemical shift‐encoded MRI (whole thigh and quadriceps), and fibrosis and muscle fiber area (MFA) of the brachial biceps. Results Of 70 patients screened, 51 entered the study. There was substantial heterogeneity between patients in muscle morphology (histology and MRI), with high fat replacement. Total fibrosis correlated significantly and mostly moderately with all functional endpoints, including both upper arm strength assessments (left and right elbow flexion rho −.574 and −.588, respectively [both P < .0001]), as did MRI fat fraction (whole thigh and quadriceps), for example, with four‐stair‐climb velocity −.554 and −.550, respectively (both P < .0001). Total fibrosis correlated significantly and moderately with both MRI fat fraction assessments (.500 [P = .0003] and .423 [.0024], respectively). Discussion In this BMD cohort, micro‐ and macroscopic morphological muscle parameters correlated moderately with each other and with functional parameters, potentially supporting the use of MRI fat fraction and histology as surrogate outcome measures in patients with BMD, although additional research is required to validate this.
Collapse
Affiliation(s)
- Giacomo P Comi
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.,Duchenne Center Netherlands, The Netherlands
| | - Claudia M Cinnante
- Neuroradiology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Hermien E Kan
- Duchenne Center Netherlands, The Netherlands.,C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Krista Vandenborne
- ImagingDMD and Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Rebecca J Willcocks
- ImagingDMD and Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Daniele Velardo
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Dino Ferrari Center, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Michela Ripolone
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Jules J van Benthem
- Department of Orthopedics, Rehabilitation and Physiotherapy, Leiden University Medical Center, Leiden, The Netherlands
| | - Nienke M van de Velde
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.,Duchenne Center Netherlands, The Netherlands
| | - Simone Nava
- Neuroradiology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | | | | |
Collapse
|
43
|
Gómez-Andrés D, Oulhissane A, Quijano-Roy S. Two decades of advances in muscle imaging in children: from pattern recognition of muscle diseases to quantification and machine learning approaches. Neuromuscul Disord 2021; 31:1038-1050. [PMID: 34736625 DOI: 10.1016/j.nmd.2021.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 07/30/2021] [Accepted: 08/01/2021] [Indexed: 11/29/2022]
Abstract
Muscle imaging has progressively gained popularity in the neuromuscular field. Together with detailed clinical examination and muscle biopsy, it has become one of the main tools for deep phenotyping and orientation of etiological diagnosis. Even in the current era of powerful new generation sequencing, muscle MRI has arisen as a tool for prioritization of certain genetic entities, supporting the pathogenicity of variants of unknown significance and facilitating diagnosis in cases with an initially inconclusive genetic study. Although the utility of muscle imaging is increasingly clear, it has not reached its full potential in clinical practice. Pattern recognition is known for a number of diseases and will certainly be enhanced by the use of machine learning approaches. For instance, MRI heatmap representations might be confronted with molecular results by obtaining a probabilistic diagnosis based in each disease "MRI fingerprints". Muscle ultrasound as a screening tool and quantified techniques such as Dixon MRI seem still underdeveloped. In this paper, we aim to appraise the advances in recent years in pediatric muscle imaging and try to define areas of uncertainty and potential advances that might become standardized to be widely used in the future.
Collapse
Affiliation(s)
- David Gómez-Andrés
- Pediatric Neurology, Vall d'Hebron Institut de Recerca (VHIR), Hospital Universitari Vall d'Hebron, ERN-RND - EURO-NMD, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain; European Network for Reference Centers on Neuromuscular Disorders (Euro-NMD ERN)
| | - Amal Oulhissane
- Université Paris-Saclay, APHP, Neuromuscular Unit, Pediatric Neurology and ICU Department, Raymond Poincaré Hospital, 92390 Garches, France
| | - Susana Quijano-Roy
- Université Paris-Saclay, APHP, Neuromuscular Unit, Pediatric Neurology and ICU Department, Raymond Poincaré Hospital, 92390 Garches, France; UMR 1179, Laboratoire handicap neuromusculaire: physiopathologie biothérapie pharmacologie appliquées (END-ICAP), UFR Simone Veil, Montigny Le Bretonneux, France; French Network of Neuromuscular Reference Centers (FILNEMUS), France.
| |
Collapse
|
44
|
Veeger TTJ, van Zwet EW, al Mohamad D, Naarding KJ, van de Velde NM, Hooijmans MT, Webb AG, Niks EH, de Groot JH, Kan HE. Muscle architecture is associated with muscle fat replacement in Duchenne and Becker muscular dystrophies. Muscle Nerve 2021; 64:576-584. [PMID: 34383334 PMCID: PMC9290788 DOI: 10.1002/mus.27399] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 08/02/2021] [Accepted: 08/07/2021] [Indexed: 12/21/2022]
Abstract
INTRODUCTION/AIMS Duchenne and Becker muscular dystrophies (DMD and BMD, respectively) are characterized by fat replacement of different skeletal muscles in a specific temporal order. Given the structural role of dystrophin in skeletal muscle mechanics, muscle architecture could be important in the progressive pathophysiology of muscle degeneration. Therefore, the aim of this study was to assess the role of muscle architecture in the progression of fat replacement in DMD and BMD. METHODS We assessed the association between literature-based leg muscle architectural characteristics and muscle fat fraction from 22 DMD and 24 BMD patients. Dixon-based magnetic resonance imaging estimates of fat fractions at baseline and 12 (only DMD) and 24 months were related to fiber length and physiological cross-sectional area (PCSA) using age-controlled linear mixed modeling. RESULTS DMD and BMD muscles with long fibers and BMD muscles with large PCSAs were associated with increased fat fraction. The effect of fiber length was stronger in muscles with larger PCSA. DISCUSSION Muscle architecture may explain the pathophysiology of muscle degeneration in dystrophinopathies, in which proximal muscles with a larger mass (fiber length × PCSA) are more susceptible, confirming the clinical observation of a temporal proximal-to-distal progression. These results give more insight into the mechanical role in the pathophysiology of muscular dystrophies. Ultimately, this new information can be used to help support the selection of current and the development of future therapies.
Collapse
Affiliation(s)
- Thom T. J. Veeger
- C.J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Erik W. van Zwet
- Department of BiostatisticsLeiden University Medical CenterLeidenThe Netherlands
| | - Diaa al Mohamad
- Department of BiostatisticsLeiden University Medical CenterLeidenThe Netherlands
| | - Karin J. Naarding
- Department of NeurologyLeiden University Medical CenterLeidenThe Netherlands
| | | | - Melissa T. Hooijmans
- Department of Radiology & Nuclear MedicineAmsterdam University Medical Centers, Location AMCAmsterdamThe Netherlands
| | - Andrew G. Webb
- C.J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Erik H. Niks
- Department of NeurologyLeiden University Medical CenterLeidenThe Netherlands
| | - Jurriaan H. de Groot
- Department of Rehabilitation MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Hermien E. Kan
- C.J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
45
|
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
|
46
|
Duong T, Canbek J, Fernandez-Fernandez A, Henricson E, Birkmeier M, Siener C, Rocha CT, McDonald C, Gordish-Dressman H. Knee Strength and Ankle Range of Motion Impacts on Timed Function Tests in Duchenne Muscular Dystrophy: In the Era of Glucocorticoids. J Neuromuscul Dis 2021; 9:147-159. [PMID: 34719507 DOI: 10.3233/jnd-210724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Duchenne Muscular Dystrophy (DMD) is a neuromuscular disorder that presents in childhood and is characterized by slowly progressive proximal weakness and lower extremity contractures that limit ambulatory ability [1, 2]. Contractures develop in the ankles, knees, and hips due to muscle imbalances, fibrotic changes, loss of strength, and static positioning [2, 5]. Currently, standards of care guidelines emphasize the importance of maintaining good musculoskeletal alignment through stretching, bracing, and glucocorticoid (GC) therapy to preserve strength and function. METHODS This is a retrospective analysis of prospectively collected data through the CINRG Duchenne Natural history study (DNHS). The objectives of this analysis are to understand the progression of ankle contractures for individuals with DMD and to investigate the relationship between progressive lower limb contractures, knee strength, and Timed Function Tests.A collection of TFTs including supine to stand (STS), 10 meter walk test (10MWT), and timed stair climbing (4SC) have been used to monitor disease progression and are predictive of loss of ambulation in these patients [4]. Multiple factors contribute to loss of ambulation, including progressive loss of strength and contracture development that leads to changing biomechanical demands for ambulation. A better understanding of the changes in strength and range of motion (ROM) that contribute to loss of function is important in a more individualized rehabilitation management plan. In this longitudinal study, we measured strength using quantitative muscle testing (QMT) with the CINRG Quantitative Measurement System (CQMS)), ROM was measuresed with a goniometer and TFTs were measured using a standard stopwatch and methodology. RESULTS We enrolled 440 participants; mean baseline age was 8.9 (2.1, 28.0) years with 1321 observations used for analysis. GC use was stratified based on duration on drug with 18.7%at < 6 months or naïve; 4.3%<1 year; 58.0%1 < 10 years; and 19.3%between 10-25 years of GC use. Ankle ROM was better for those on GC compared to GC naive but did not significantly influence long-term progression rates. QMT, ROM, age and GCs contribute to speed of TFTs. Knee extension (KE) strength and Dorsiflexion (DF) ROM are significant predictors of speed for all TFTs (p < 0.001). Of the variables used in this analysis, KE strength is the primary predictor of walking speed, estimating that every pound increase in KE results in a 0.042 m/s improvement in 10MWT, and a smaller similar increase of 0.009 m/s with every degree of ankle DF ROM. CONCLUSION GC use provides an improvement in strength and ROM but does not affect rate of change. Knee strength has a greater influence on speed of TFTs than DF ROM, although both are statistically significant predictors of speed. Results show that retaining knee strength [1, 2], along with joint flexibility, may be important factors in the ability to perform walking, climbing and supine to stand activities.
Collapse
Affiliation(s)
- Tina Duong
- Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Rehabilitation, Stanford Healthcare, Stanford, CA, USA
| | - Jennifer Canbek
- Physical Therapy Department, Nova Southeastern University, Fort Lauderdale, FL, USA
| | | | - Erik Henricson
- University of California, Davis, Department of Neurology, Sacramento, CA USA
| | - Marisa Birkmeier
- Department of Health, Human Function, and Rehabilitation Sciences, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Catherine Siener
- Department of Neurology, Washington University, St. Louis, MO, USA
| | - Carolina Tesi Rocha
- Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA
| | - Craig McDonald
- University of California, Davis, Department of Neurology, Sacramento, CA USA
| | | | | |
Collapse
|
47
|
Naarding KJ, van der Holst M, van Zwet EW, van de Velde NM, de Groot IJM, Verschuuren JJGM, Kan HE, Niks EH. Association of Elbow Flexor MRI Fat Fraction With Loss of Hand-to-Mouth Movement in Patients With Duchenne Muscular Dystrophy. Neurology 2021; 97:e1737-e1742. [PMID: 34493619 PMCID: PMC8605612 DOI: 10.1212/wnl.0000000000012724] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 08/10/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES To study the potential of quantitative MRI (qMRI) fat fraction (FF) as a biomarker in nonambulant patients with Duchenne muscular dystrophy (DMD), we assessed the additive predictive value of elbow flexor FF to age at loss of hand-to-mouth movement. METHODS Nonambulant patients with DMD (age ≥8 years) were included. Four-point Dixon MRI scans of the right upper arm were performed at baseline and at the 12-, 18-, or 24-month follow-up. Elbow flexor FFs were determined from 5 central slices. Loss of hand-to-mouth movement was determined at study visits and by phone calls every 4 months. FFs were fitted to a sigmoidal curve by use of a mixed model with random slope to predict individual trajectories. The added predictive value of elbow flexor FF to age at loss of hand-to-mouth movement was calculated from a Cox model with the predicted FF as a time-varying covariate, yielding a hazard ratio. RESULTS Forty-eight MRIs of 20 patients with DMD were included. The hazard ratio of a percent-point increase in elbow flexor FF for the time to loss of hand-to-mouth movement was 1.12 (95% confidence interval 1.04-1.21; p = 0.002). This corresponded to a 3.13-fold increase in the instantaneous risk of loss of hand-to-mouth movement in patients with a 10-percent points higher elbow flexor FF at any age. DISCUSSION In this prospective study, elbow flexor FF predicted loss of hand-to-mouth movement independently of age. qMRI-measured elbow flexor FF can be used as a surrogate endpoint or stratification tool for clinical trials in nonambulant patients with DMD. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that qMRI FF of elbow flexor muscles in patients with DMD predicts loss of hand-to-mouth movement independently of age.
Collapse
Affiliation(s)
- Karin J Naarding
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Menno van der Holst
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Erik W van Zwet
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nienke M van de Velde
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Imelda J M de Groot
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jan J G M Verschuuren
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hermien E Kan
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Erik H Niks
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
48
|
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
|
49
|
Alonso-Jiménez A, Nuñez-Peralta C, Montesinos P, Alonso-Pérez J, García C, Montiel E, Belmonte I, Pedrosa I, Segovia S, Llauger J, Díaz-Manera J. Different Approaches to Analyze Muscle Fat Replacement With Dixon MRI in Pompe Disease. Front Neurol 2021; 12:675781. [PMID: 34305788 PMCID: PMC8298190 DOI: 10.3389/fneur.2021.675781] [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] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
Quantitative MRI is an increasingly used method to monitor disease progression in muscular disorders due to its ability to measure changes in muscle fat content (reported as fat fraction) over a short period. Being able to objectively measure such changes is crucial for the development of new treatments in clinical trials. However, the analysis of the images involved continues to be a daunting task because of the time needed. Whether a more specific analysis selecting individual muscles or a global one analyzing the whole thigh or compartments could be a suitable alternative has only been marginally studied. In our study we compare three methods of analysis of 2-point-dixon images in a cohort of 34 patients with late onset Pompe disease followed over a period of one year. We measured fat fraction on MRIs obtained at baseline and at year 1, and we calculated the increment of fat fraction. We correlated the results obtained with the results of muscle function tests to investigate whether the three methods of analysis were equivalent or not. We observed significant differences between the three methods in the estimation of the fat fraction at both baseline and year 1, but no difference was found in the increment in fat fraction between baseline and year 1. When we correlated the fat fraction obtained with each method and the muscle function tests, we found a significant correlation with most tests in all three methods, although in most comparisons the highest correlation coefficient was found with the analysis of individual muscles. We conclude that the fastest strategy of analysis assessing compartments or the whole thigh could be reliable for certain cohorts of patients where the variable to study is the fat increment. In other sorts of studies, an individual muscle approach seems the most reliable technique.
Collapse
Affiliation(s)
- Alicia Alonso-Jiménez
- Neuromuscular Disorders Unit, Neurology Department, Departament de Medicina, Hospital de la Santa Creu I Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Neuromuscular Reference Center, Neurology Department, University Hospital of Antwerp, Edegem, Belgium
| | - Claudia Nuñez-Peralta
- Radiology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Jorge Alonso-Pérez
- Neuromuscular Disorders Unit, Neurology Department, Departament de Medicina, Hospital de la Santa Creu I Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Biomedical Network Research Centre on Rare Diseases (CIBERER), Barcelona, Spain
| | - Carme García
- Rehabilitation and Physiotherapy Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Elena Montiel
- Rehabilitation and Physiotherapy Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Izaskun Belmonte
- Rehabilitation and Physiotherapy Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Irene Pedrosa
- Rehabilitation and Physiotherapy Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sonia Segovia
- Neuromuscular Disorders Unit, Neurology Department, Departament de Medicina, Hospital de la Santa Creu I Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Biomedical Network Research Centre on Rare Diseases (CIBERER), Barcelona, Spain
| | - Jaume Llauger
- Radiology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jordi Díaz-Manera
- Neuromuscular Disorders Unit, Neurology Department, Departament de Medicina, Hospital de la Santa Creu I Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Biomedical Network Research Centre on Rare Diseases (CIBERER), Barcelona, Spain.,John Walton Muscular Dystrophy Research Centre, Newcastle University, International Centre for Life, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
50
|
Lopez C, Taivassalo T, Berru MG, Saavedra A, Rasmussen HC, Batra A, Arora H, Roetzheim AM, Walter GA, Vandenborne K, Forbes SC. Postcontractile blood oxygenation level-dependent (BOLD) response in Duchenne muscular dystrophy. J Appl Physiol (1985) 2021; 131:83-94. [PMID: 34013753 PMCID: PMC8325615 DOI: 10.1152/japplphysiol.00634.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 04/28/2021] [Accepted: 05/13/2021] [Indexed: 11/22/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by a progressive replacement of muscle by fat and fibrous tissue, muscle weakness, and loss of functional abilities. Impaired vasodilatory and blood flow responses to muscle activation have also been observed in DMD and associated with mislocalization of neuronal nitric oxide synthase mu (nNOSμ) from the sarcolemma. The objective of this study was to determine whether the postcontractile blood oxygen level-dependent (BOLD) MRI response is impaired in DMD and correlated with established markers of disease severity in DMD, including MRI muscle fat fraction (FF) and clinical functional measures. Young boys with DMD (n = 16, 5-14 yr) and unaffected controls (n = 16, 5-14 yr) were evaluated using postcontractile BOLD, FF, and functional assessments. The BOLD response was measured following five brief (2 s) maximal voluntary dorsiflexion contractions, each separated by 1 min of rest. FFs from the anterior compartment lower leg muscles were quantified via chemical shift-encoded imaging. Functional abilities were assessed using the 10 m walk/run and the 6-min walk distance (6MWD). The peak BOLD responses in the tibialis anterior and extensor digitorum longus were reduced (P < 0.001) in DMD compared with controls. Furthermore, the anterior compartment peak BOLD response correlated with function (6MWD ρ = 0.87, P < 0.0001; 10 m walk/run time ρ = -0.78, P < 0.001) and FF (ρ = -0.52, P = 0.05). The reduced postcontractile BOLD response in DMD may reflect impaired microvascular function. The relationship observed between the postcontractile peak BOLD response and functional measures and FF suggests that the BOLD response is altered with disease severity in DMD.NEW & NOTEWORTHY This study examined the postcontractile blood oxygen level-dependent (BOLD) response in boys with Duchenne muscular dystrophy (DMD) and unaffected controls, and correlated this measure to markers of disease severity. Our findings indicate that the postcontractile BOLD response is impaired in DMD after brief muscle contractions, is correlated to disease severity, and may be valuable to implement in future studies to evaluate treatments targeting microvascular function in DMD.
Collapse
Affiliation(s)
- Christopher Lopez
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Tanja Taivassalo
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Maria G Berru
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Andres Saavedra
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Hannah C Rasmussen
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Abhinandan Batra
- Department of Physical Therapy, University of Florida, Gainesville, Florida
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Harneet Arora
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Alex M Roetzheim
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Glenn A Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| | - Sean C Forbes
- Department of Physical Therapy, University of Florida, Gainesville, Florida
| |
Collapse
|