1
|
Gerhalter T, Marty B, Gast LV, Roemer F, Baudin PY, Trollmann R, Uder M, Carlier PG, Nagel AM. Longitudinal Follow-Up of Patients With Duchenne Muscular Dystrophy Using Quantitative 23Na and 1H MRI. J Cachexia Sarcopenia Muscle 2025; 16:e13812. [PMID: 40254293 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 Erlangen, FAU, Erlangen, Germany
| | - Benjamin Marty
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France
| | - Lena V Gast
- Institute of Radiology University Hospital Erlangen, FAU, Erlangen, Germany
| | - Frank Roemer
- Institute of Radiology University Hospital Erlangen, FAU, Erlangen, Germany
| | - Pierre-Yves Baudin
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France
| | - Regina Trollmann
- Department of Pediatrics, Division Neuropediatrics, FAU, Erlangen, Germany
| | - Michael Uder
- Institute of Radiology University Hospital Erlangen, FAU, Erlangen, Germany
| | - Pierre G Carlier
- Saint-Luc University Hospital, Brussels, Belgium
- Erasme University Hospital, Brussels, Belgium
- Liège State University, Liège, Belgium
| | - Armin M Nagel
- Institute of Radiology University Hospital Erlangen, FAU, Erlangen, Germany
- Division of Medical Physics in Radiology, DKFZ, Heidelberg, Germany
| |
Collapse
|
2
|
Sherlock SP, McCrady A, Palmer J, Aghamolaey H, Ahlgren A, Widholm P, Dahlqvist Leinhard O, Karlsson M. Relationship Between Quantitative Magnetic Resonance Imaging Measures and Functional Changes in Patients With Duchenne Muscular Dystrophy. Muscle Nerve 2025; 71:343-352. [PMID: 39713935 PMCID: PMC11799397 DOI: 10.1002/mus.28321] [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: 12/20/2023] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION/AIMS Improved methodologies to monitor the progression of Duchenne muscular dystrophy (DMD) are needed, especially in the context of clinical trials. We report changes in muscle magnetic resonance imaging (MRI) parameters in participants with DMD, including changes in lean muscle volume (LMV), muscle fat fraction (MFF), and muscle fat infiltration (MFI) and their relationship to changes in functional parameters. METHODS MRI data were obtained as part of a clinical study (NCT02310763) of domagrozumab, an antibody-targeting myostatin that negatively regulates skeletal muscle mass. This post hoc analysis evaluated participants with Dixon MRI data and corresponding functional data at baseline and weeks 49 and 97. Images were analyzed to evaluate changes in adductors, hamstrings, and quadriceps. RESULTS There was a positive correlation between increases in LMV and function. LMV changes in adductors (R = 0.51) and quadriceps (R = 0.54) showed a stronger correlation with function than LMV changes in hamstrings (R = 0.30). There was a negative correlation between MFF and MFI, respectively, and function in adductors (R = -0.57, R = -0.42), quadriceps (R = -0.59, R = -0.50), and hamstrings (R = -0.53, R = -048). Participants with preserved North Star Ambulatory Assessment scores had high total LMV (LMVtot) and low total MFI (MFItot). Low ratios of LMVtot to MFItot, or participants with small LMVtot and high MFItot, appeared to have a rapid decline in function and loss of ambulation. DISCUSSION These findings support the use of MRI biomarkers as potential surrogate endpoints in clinical trials of patients with DMD. TRIAL REGISTRATION ClinicalTrials.gov identifiers: NCT02310763.
Collapse
Affiliation(s)
| | - Allison McCrady
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | | | - Haleh Aghamolaey
- Department of StatisticsQuanticate Clinical Research OrganizationOntarioCanada
| | | | | | | | | |
Collapse
|
3
|
Caldas de Almeida Araujo E, Barthélémy I, Fromes Y, Baudin P, Blot S, Reyngoudt H, Marty B. Comprehensive quantitative magnetic resonance imaging assessment of skeletal muscle pathophysiology in golden retriever muscular dystrophy: Insights from multicomponent water T2 and extracellular volume fraction. NMR IN BIOMEDICINE 2025; 38:e5278. [PMID: 39434514 PMCID: PMC11602680 DOI: 10.1002/nbm.5278] [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: 06/26/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
Quantitative MRI and MRS have become important tools for the assessment and management of patients with neuromuscular disorders (NMDs). Despite significant progress, there is a need for new objective measures with improved specificity to the underlying pathophysiological alteration. This would enhance our ability to characterize disease evolution and improve therapeutic development. In this study, qMRI methods that are commonly used in clinical studies involving NMDs, like water T2 (T2H2O) and T1 and fat-fraction (FF) mapping, were employed to evaluate disease activity and progression in the skeletal muscle of golden retriever muscular dystrophy (GRMD) dogs. Additionally, extracellular volume (ECV) fraction and single-voxel bicomponent water T2 relaxometry were included as potential markers of specific histopathological changes within the tissue. Apart from FF, which was not significantly different between GRMD and control dogs and showed no trend with age, T2H2O, T1, ECV, and the relative fraction of the long-T2 component, A2, were significantly elevated in GRMD dogs across all age ranges. Moreover, longitudinal assessment starting at 2 months of age revealed significant decreases in T2H2O, T1, ECV, A2, and the T2 of the shorter-T2 component, T21, in both control and GRMD dogs during their first year of life. Notably, insights from ECV and bicomponent water T2 indicate that (I) the elevated T2H2O and T1 values observed in dystrophic muscle are primarily driven by an expansion of the extracellular space, likely driven by the edematous component of inflammatory responses to tissue injury and (II) the significant decrease of T2H2O and T1 with age in control and GRMD dogs reflects primarily the progressive increase in fiber diameter and protein content during tissue development. Our study underscores the potential of multicomponent water T2 relaxometry and ECV to provide valuable insights into muscle pathology in NMDs.
Collapse
Affiliation(s)
| | - Inès Barthélémy
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France; EnvA, IMRBMaisons‐AlfortFrance
| | - Yves Fromes
- NMR Laboratory, Neuromuscular Investigation CenterInstitute of MyologyParisFrance
| | - Pierre‐Yves Baudin
- NMR Laboratory, Neuromuscular Investigation CenterInstitute of MyologyParisFrance
| | - Stéphane Blot
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France; EnvA, IMRBMaisons‐AlfortFrance
| | - Harmen Reyngoudt
- NMR Laboratory, Neuromuscular Investigation CenterInstitute of MyologyParisFrance
| | - Benjamin Marty
- NMR Laboratory, Neuromuscular Investigation CenterInstitute of MyologyParisFrance
| |
Collapse
|
4
|
Yoon DY, Daniels MJ, Willcocks RJ, Triplett WT, Morales JF, Walter GA, Rooney WD, Vandenborne K, Kim S. Five multivariate Duchenne muscular dystrophy progression models bridging six-minute walk distance and MRI relaxometry of leg muscles. J Pharmacokinet Pharmacodyn 2024; 51:671-683. [PMID: 38609673 PMCID: PMC11470134 DOI: 10.1007/s10928-024-09910-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/15/2024] [Indexed: 04/14/2024]
Abstract
The study aimed to provide quantitative information on the utilization of MRI transverse relaxation time constant (MRI-T2) of leg muscles in DMD clinical trials by developing multivariate disease progression models of Duchenne muscular dystrophy (DMD) using 6-min walk distance (6MWD) and MRI-T2. Clinical data were collected from the prospective and longitudinal ImagingNMD study. Disease progression models were developed by a nonlinear mixed-effect modeling approach. Univariate models of 6MWD and MRI-T2 of five muscles were developed separately. Age at assessment was the time metric. Multivariate models were developed by estimating the correlation of 6MWD and MRI-T2 model variables. Full model estimation approach for covariate analysis and five-fold cross validation were conducted. Simulations were performed to compare the models and predict the covariate effects on the trajectories of 6MWD and MRI-T2. Sigmoid Imax and Emax models best captured the profiles of 6MWD and MRI-T2 over age. Steroid use, baseline 6MWD, and baseline MRI-T2 were significant covariates. The median age at which 6MWD is half of its maximum decrease in the five models was similar, while the median age at which MRI-T2 is half of its maximum increase varied depending on the type of muscle. The models connecting 6MWD and MRI-T2 successfully quantified how individual characteristics alter disease trajectories. The models demonstrate a plausible correlation between 6MWD and MRI-T2, supporting the use of MRI-T2. The developed models will guide drug developers in using the MRI-T2 to most efficient use in DMD clinical trials.
Collapse
Affiliation(s)
- Deok Yong Yoon
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Michael J Daniels
- Department of Statistics, University of Florida, Gainesville, FL, USA
| | | | - William T Triplett
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Juan Francisco Morales
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Glenn A Walter
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - William D Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - Sarah Kim
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA.
| |
Collapse
|
5
|
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
|
6
|
Peng F, Tang D, Qing W, Chen W, Li S, Guo Y, Luo G, Zhao H. Utilization of Multi-Parametric Quantitative Magnetic Resonance Imaging in the Early Diagnosis of Duchenne Muscular Dystrophy. J Magn Reson Imaging 2024; 60:1402-1413. [PMID: 38095338 DOI: 10.1002/jmri.29182] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND It is challenging to diagnose suspected Duchenne muscular dystrophy (DMD) patients in the very early stage of the disease. More evidence is needed to demonstrate the potential of quantitative MRI (qMRI) in precisely identifying patients before substantial physical decline occurs. PURPOSE To assess the early diagnostic performance of multi-parametric qMRI for DMD patients, and the ability to identify DMD patients with mild functional decline. STUDY TYPE Prospective. SUBJECTS One hundred and forty DMD subjects (9.0 ± 2.2 years old), 24 male healthy controls (HCs) (9.2 ± 2.5 years old). FIELD STRENGTH/SEQUENCE 3.0 T/3-point Dixon, T1-mapping, and T2-mapping. ASSESSMENT qMRI measurements (fat fraction [FF], T1, and T2) of 11 thigh muscles (rectus femoris [RF], vastus lateralis [VL], vastus intermedius, vastus medialis, gracilis, sartorius, adductor longus, adductor magnus [AM], semitendinosus, semimembranosus, biceps femoris long head [BFLH]) on the right side were conducted. NorthStar ambulatory assessment (NSAA) score used to evaluate the function of DMD patients and divided them into three subgroups: mild (76-100 score), moderate (51-75 score), and severe (0-50 score) functional decline. STATISTICAL TESTS Independent t-test, ANOVA analysis, and receiver operating characteristic (ROC) curves. A P-value <0.05 was considered statistically significant. RESULTS Compared with HCs, FF and T2 were significantly higher in the group of all DMD patients, while T1 was significantly lower. The combination of T1 and T2 in RF, VL, AM, and BFLH achieved excellent area under curve (AUCs) (0.967-0.992) in differentiating five DMD patients without abnormal fat infiltration from HCs. Overall, T2 reached higher AUCs than FF and T1 in distinguishing DMD with mild functional decline from HCs, whereas FF achieved higher AUCs than T1 and T2 in distinguishing three DMD subgroups with functional decline. DATA CONCLUSION Multi-parametric qMRI demonstrate effective diagnostic capabilities for DMD patients in the early stage of the disease, and can identify patients with mild physical decline. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Fei Peng
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Medical Imaging center, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Deqiu Tang
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Weipeng Qing
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Wei Chen
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Shuhao Li
- Department of Medical Imaging center, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yingkun Guo
- Department of Radiology, Key Laboratory of Obstetric and Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Guanghua Luo
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Heng Zhao
- Department of Radiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
7
|
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
|
8
|
Dabaj I, Ducatez F, Marret S, Bekri S, Tebani A. Neuromuscular disorders in the omics era. Clin Chim Acta 2024; 553:117691. [PMID: 38081447 DOI: 10.1016/j.cca.2023.117691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023]
Abstract
Neuromuscular disorders encompass a spectrum of conditions characterized by primary lesions within the peripheral nervous system, which include the anterior horn cell, peripheral nerve, neuromuscular junction, and muscle. In pediatrics, most of these disorders are linked to genetic causes. Despite the considerable progress, the diagnosis of these disorders remains a challenging due to wide clinical presentation, disease heterogeneity and rarity. It is noteworthy that certain neuromuscular disorders, once deemed untreatable, can now be effectively managed through novel therapies. Biomarkers emerge as indispensable tools, serving as objective measures that not only refine diagnostic accuracy but also provide guidance for therapeutic decision-making and the ongoing monitoring of long-term outcomes. Herein a comprehensive review of biomarkers in neuromuscular disorders is provided. We highlight the role of omics-based technologies that further characterize neuromuscular pathophysiology as well as identify potential therapeutic targets to guide treatment strategies.
Collapse
Affiliation(s)
- Ivana Dabaj
- Normandie Univ, UNIROUEN, INSERM U1245, Nord/Est/Ile de France Neuromuscular Reference Center CHU Rouen, Department of Neonatalogy, Pediatric Intensive Care, and Neuropediatrics, F-76000 Rouen, France.
| | - Franklin Ducatez
- Normandie Univ, UNIROUEN, INSERM U1245, Nord/Est/Ile de France Neuromuscular Reference Center CHU Rouen, Department of Neonatalogy, Pediatric Intensive Care, and Neuropediatrics, F-76000 Rouen, France
| | - Stéphane Marret
- Normandie Univ, UNIROUEN, INSERM U1245, Nord/Est/Ile de France Neuromuscular Reference Center CHU Rouen, Department of Neonatalogy, Pediatric Intensive Care, and Neuropediatrics, F-76000 Rouen, France
| | - Soumeya Bekri
- Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, Department of Metabolic Biochemistry, F-76000 Rouen, France
| | - Abdellah Tebani
- Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, Department of Metabolic Biochemistry, F-76000 Rouen, France
| |
Collapse
|
9
|
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
|
10
|
Otto LA, Froeling M, van Eijk RP, Wadman RI, Cuppen I, van der Woude DR, Bartels B, Asselman FL, Hendrikse J, van der Pol WL. Monitoring Nusinersen Treatment Effects in Children with Spinal Muscular Atrophy with Quantitative Muscle MRI. J Neuromuscul Dis 2024; 11:91-101. [PMID: 38073395 PMCID: PMC10789331 DOI: 10.3233/jnd-221671] [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: 10/16/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Spinal muscular atrophy (SMA) is caused by deficiency of survival motor neuron (SMN) protein. Intrathecal nusinersen treatment increases SMN protein in motor neurons and has been shown to improve motor function in symptomatic children with SMA. OBJECTIVE We used quantitative MRI to gain insight in microstructure and fat content of muscle during treatment and to explore its use as biomarker for treatment effect. METHODS We used a quantitative MRI protocol before start of treatment and following the 4th and 6th injection of nusinersen in 8 children with SMA type 2 and 3 during the first year of treatment. The MR protocol allowed DIXON, T2 mapping and diffusion tensor imaging acquisitions. We also assessed muscle strength and motor function scores. RESULTS Fat fraction of all thigh muscles with the exception of the m. adductor longus increased in all patients during treatment (+3.2%, p = 0.02). WaterT2 showed no significant changes over time (-0.7 ms, p = 0.3). DTI parameters MD and AD demonstrate a significant decrease in the hamstrings towards values observed in healthy muscle. CONCLUSIONS Thigh muscles of children with SMA treated with nusinersen showed ongoing fatty infiltration and possible normalization of thigh muscle microstructure during the first year of nusinersen treatment. Quantitative muscle MRI shows potential as biomarker for the effects of SMA treatment strategies.
Collapse
Affiliation(s)
- Louise A.M. Otto
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - M. Froeling
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ruben P.A. van Eijk
- Biostatistics & Research Support, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Renske I. Wadman
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Inge Cuppen
- Department of Neurology and Child Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Danny R. van der Woude
- Department of Child Development and Exercise Center, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Bart Bartels
- Department of Child Development and Exercise Center, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Fay-Lynn Asselman
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jeroen Hendrikse
- Department of Radiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - W. Ludo van der Pol
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
11
|
McDonald C, Camino E, Escandon R, Finkel RS, Fischer R, Flanigan K, Furlong P, Juhasz R, Martin AS, Villa C, Sweeney HL. Draft Guidance for Industry Duchenne Muscular Dystrophy, Becker Muscular Dystrophy, and Related Dystrophinopathies - Developing Potential Treatments for the Entire Spectrum of Disease. J Neuromuscul Dis 2024; 11:499-523. [PMID: 38363616 DOI: 10.3233/jnd-230219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Background Duchenne muscular dystrophy (DMD) and related dystrophinopathies are neuromuscular conditions with great unmet medical needs that require the development of effective medical treatments. Objective To aid sponsors in clinical development of drugs and therapeutic biological products for treating DMD across the disease spectrum by integrating advancements, patient registries, natural history studies, and more into a comprehensive guidance. Methods This guidance emerged from collaboration between the FDA, the Duchenne community, and industry stakeholders. It entailed a structured approach, involving multiple committees and boards. From its inception in 2014, the guidance underwent revisions incorporating insights from gene therapy studies, cardiac function research, and innovative clinical trial designs. Results The guidance provides a deeper understanding of DMD and its variants, focusing on patient engagement, diagnostic criteria, natural history, biomarkers, and clinical trials. It underscores patient-focused drug development, the significance of dystrophin as a biomarker, and the pivotal role of magnetic resonance imaging in assessing disease progression. Additionally, the guidance addresses cardiomyopathy's prominence in DMD and the burgeoning field of gene therapy. Conclusions The updated guidance offers a comprehensive understanding of DMD, emphasizing patient-centric approaches, innovative trial designs, and the importance of biomarkers. The focus on cardiomyopathy and gene therapy signifies the evolving realm of DMD research. It acts as a crucial roadmap for sponsors, potentially leading to improved treatments for DMD.
Collapse
Affiliation(s)
| | - Eric Camino
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Rafael Escandon
- DGBI Consulting, LLC, Bainbridge Island, Washington, DC, USA
| | | | - Ryan Fischer
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Kevin Flanigan
- Center for Experimental Neurotherapeutics, Department of Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Pat Furlong
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Rose Juhasz
- Nationwide Children's Hospital, Columbus, OH, USA
| | - Ann S Martin
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Chet Villa
- Trinity Health Michigan, Grand Rapids, MI, USA
| | - H Lee Sweeney
- Cincinnati Children's Hospital Medical Center within the UC Department of Pediatrics, Cincinnati, OH, USA
| |
Collapse
|
12
|
Hiyoshi T, Zhao F, Baba R, Hirakawa T, Kuboki R, Suzuki K, Tomimatsu Y, O'Donnell P, Han S, Zach N, Nakashima M. Electrical impedance myography detects dystrophin-related muscle changes in mdx mice. Skelet Muscle 2023; 13:19. [PMID: 37980539 PMCID: PMC10657153 DOI: 10.1186/s13395-023-00331-1] [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: 08/29/2023] [Accepted: 10/27/2023] [Indexed: 11/20/2023] Open
Abstract
BACKGROUND The lack of functional dystrophin protein in Duchenne muscular dystrophy (DMD) causes chronic skeletal muscle inflammation and degeneration. Therefore, the restoration of functional dystrophin levels is a fundamental approach for DMD therapy. Electrical impedance myography (EIM) is an emerging tool that provides noninvasive monitoring of muscle conditions and has been suggested as a treatment response biomarker in diverse indications. Although magnetic resonance imaging (MRI) of skeletal muscles has become a standard measurement in clinical trials for DMD, EIM offers distinct advantages, such as portability, user-friendliness, and reduced cost, allowing for remote monitoring of disease progression or response to therapy. To investigate the potential of EIM as a biomarker for DMD, we compared longitudinal EIM data with MRI/histopathological data from an X-linked muscular dystrophy (mdx) mouse model of DMD. In addition, we investigated whether EIM could detect dystrophin-related changes in muscles using antisense-mediated exon skipping in mdx mice. METHODS The MRI data for muscle T2, the magnetic resonance spectroscopy (MRS) data for fat fraction, and three EIM parameters with histopathology were longitudinally obtained from the hindlimb muscles of wild-type (WT) and mdx mice. In the EIM study, a cell-penetrating peptide (Pip9b2) conjugated antisense phosphorodiamidate morpholino oligomer (PPMO), designed to induce exon-skipping and restore functional dystrophin production, was administered intravenously to mdx mice. RESULTS MRI imaging in mdx mice showed higher T2 intensity at 6 weeks of age in hindlimb muscles compared to WT mice, which decreased at ≥ 9 weeks of age. In contrast, EIM reactance began to decline at 12 weeks of age, with peak reduction at 18 weeks of age in mdx mice. This decline was associated with myofiber atrophy and connective tissue infiltration in the skeletal muscles. Repeated dosing of PPMO (10 mg/kg, 4 times every 2 weeks) in mdx mice led to an increase in muscular dystrophin protein and reversed the decrease in EIM reactance. CONCLUSIONS These findings suggest that muscle T2 MRI is sensitive to the early inflammatory response associated with dystrophin deficiency, whereas EIM provides a valuable biomarker for the noninvasive monitoring of subsequent changes in skeletal muscle composition. Furthermore, EIM reactance has the potential to monitor dystrophin-deficient muscle abnormalities and their recovery in response to antisense-mediated exon skipping.
Collapse
Affiliation(s)
- Tetsuaki Hiyoshi
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Fuqiang Zhao
- Center of Excellence for Imaging, Preclinical and Translational Sciences, Takeda Development Center Americas, Inc., 95 Hayden Avenue, Lexington, MA, 02141, USA
| | - Rina Baba
- Muscular Disease and Neuropathy Unit, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Takeshi Hirakawa
- Muscular Disease and Neuropathy Unit, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Ryosuke Kuboki
- Muscular Disease and Neuropathy Unit, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Kazunori Suzuki
- Muscular Disease and Neuropathy Unit, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Yoshiro Tomimatsu
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Patricio O'Donnell
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Development Center Americas, Inc., 95 Hayden Avenue, Lexington, MA, 02141, USA
| | - Steve Han
- Neuroscience Therapeutic Area Unit, Takeda Development Center Americas, Inc., 95 Hayden Avenue, Lexington, MA, 02141, USA
| | - Neta Zach
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Development Center Americas, Inc., 95 Hayden Avenue, Lexington, MA, 02141, USA
| | - Masato Nakashima
- Neuroscience Translational Medicine, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chome, Fujisawa, Kanagawa, 251-8555, Japan.
| |
Collapse
|
13
|
Reyngoudt H, Baudin PY, Carlier PG, Lopez Kolkovsky AL, de Almeida Araujo EC, Marty B. New Insights into the Spread of MRS-Based Water T2 Values Observed in Highly Fatty Replaced Muscles. J Magn Reson Imaging 2023; 58:1557-1568. [PMID: 36877200 DOI: 10.1002/jmri.28669] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 03/07/2023] Open
Abstract
BACKGROUND The reference standard for assessing water T2 (T2,H2O ) at high fat fraction (FF) is 1 H MRS. T2,H2O (T2,H2O,MRS ) dependence on FF (FFMRS ) has recently been demonstrated in muscle at high FF (i.e. ≥60%). PURPOSE To investigate the relationship between T2,H2O,MRS and FFMRS in the thigh/leg muscles of patients with neuromuscular diseases and to compare with quantitative MRI. STUDY TYPE Retrospective case-control study. POPULATION A total of 151 patients with neuromuscular disorders (mean age ± standard deviation = 52.5 ± 22.6 years, 54% male), 44 healthy volunteers (26.5 ± 13.0 years, 57% male). FIELD STRENGTH/SEQUENCE A 3-T; single-voxel stimulated echo acquisition mode (STEAM) MRS, multispin echo (MSE) imaging (for T2 mapping, T2,H2O,MRI ), three-point Dixon imaging (for FFMRI andR 2 * mapping). ASSESSMENT Mono-exponential and bi-exponential models were fitted to water T2 decay curves to extract T2,H2O,MRS and FFMRS . Water resonance full-width-at-half-maximum (FWHM) and B0 spread (∆B0 ) values were calculated. T2,H2O,MRI (mean), FFMRI (mean, kurtosis, and skewness), andR 2 * (mean) values were estimated in the MRS voxel. STATISTICAL TESTS Mann-Whitney U tests, Kruskal-Wallis tests. A P-value <0.05 was considered statistically significant. RESULTS Normal T2,H2O,MRS threshold was defined as the 90th percentile in healthy controls: 30.3 msec. T2,H2O,MRS was significantly higher in all patients with FFMRS < 60% compared to healthy controls. We discovered two subgroups in patients with FFMRS ≥ 60%: one with T2,H2O,MRS ≥ 30.3 msec and one with T2,H2O,MRS < 30.3 msec including abnormally low T2,H2O,MRS . The latter subgroup had significantly higher water resonance FWHM, ∆B0 , FFMRI kurtosis, and skewness values but nonsignificantly differentR 2 * (P = 1.00) and long T2,H2O,MRS component and its fraction (P > 0.11) based on the bi-exponential analysis. DATA CONCLUSION The findings suggest that the cause for (abnormally) T2,H2O,MRS at high FFMRS is biophysical, due to differences in susceptibility between muscle and fat (increased FWHM and ∆B0 ), rather than pathophysiological such as compartmentation changes, which would be reflected by the bi-exponential analysis. EVIDENCE LEVEL 3 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Harmen Reyngoudt
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France
| | - Pierre-Yves Baudin
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France
| | - Pierre G Carlier
- Université Paris Saclay, CEA, Service Hospitalier Frédéric Joliot, Orsay, France
| | | | | | - Benjamin Marty
- NMR Laboratory, Neuromuscular Investigation Center, Institute of Myology, Paris, France
| |
Collapse
|
14
|
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
|
15
|
Engelke K, Chaudry O, Gast L, Eldib MAB, Wang L, Laredo JD, Schett G, Nagel AM. Magnetic resonance imaging techniques for the quantitative analysis of skeletal muscle: State of the art. J Orthop Translat 2023; 42:57-72. [PMID: 37654433 PMCID: PMC10465967 DOI: 10.1016/j.jot.2023.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/04/2023] [Accepted: 07/19/2023] [Indexed: 09/02/2023] Open
Abstract
Background Magnetic resonance imaging (MRI) is the dominant 3D imaging modality to quantify muscle properties in skeletal muscle disorders, in inherited and acquired muscle diseases, and in sarcopenia, in cachexia and frailty. Methods This review covers T1 weighted and Dixon sequences, introduces T2 mapping, diffusion tensor imaging (DTI) and non-proton MRI. Technical concepts, strengths, limitations and translational aspects of these techniques are discussed in detail. Examples of clinical applications are outlined. For comparison 31P-and 13C-MR Spectroscopy are also addressed. Results MRI technology provides a rich toolset to assess muscle deterioration. In addition to classical measures such as muscle atrophy using T1 weighted imaging and fat infiltration using Dixon sequences, parameters characterizing inflammation from T2 maps, tissue sodium using non-proton MRI techniques or concentration or fiber architecture using diffusion tensor imaging may be useful for an even earlier diagnosis of the impairment of muscle quality. Conclusion Quantitative MRI provides new options for muscle research and clinical applications. Current limitations that also impair its more widespread use in clinical trials are lack of standardization, ambiguity of image segmentation and analysis approaches, a multitude of outcome parameters without a clear strategy which ones to use and the lack of normal data.
Collapse
Affiliation(s)
- Klaus Engelke
- Department of Medicine III, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
- Institute of Medical Physics (IMP), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Henkestr. 91, 91052, Erlangen, Germany
- Clario Inc, Germany
| | - Oliver Chaudry
- Department of Medicine III, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
| | - Lena Gast
- Institute of Radiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Maximiliansplatz 3, 91054, Erlangen, Germany
| | | | - Ling Wang
- Department of Radiology, Beijing Jishuitan Hospital, Beijing, China
| | - Jean-Denis Laredo
- Service d’Imagerie Médicale, Institut Mutualiste Montsouris & B3OA, UMR CNRS 7052, Inserm U1271 Université de Paris-Cité, Paris, France
| | - Georg Schett
- Department of Medicine III, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
| | - Armin M. Nagel
- Institute of Radiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), University Hospital Erlangen, Maximiliansplatz 3, 91054, Erlangen, Germany
- Division of Medical Physics in Radiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| |
Collapse
|
16
|
Monte JR, Hooijmans MT, Froeling M, Oudeman J, Tol JL, Strijkers GJ, Nederveen AJ, Maas M. Diffusion tensor imaging and quantitative T2 mapping to monitor muscle recovery following hamstring injury. NMR IN BIOMEDICINE 2023; 36:e4902. [PMID: 36630472 DOI: 10.1002/nbm.4902] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 01/07/2023] [Accepted: 01/09/2023] [Indexed: 06/15/2023]
Abstract
MRI examinations are accurate for diagnosing sports-related acute hamstring injuries. However, sensitive imaging methods for assessing recovery of these injuries are lacking. Diffusion tensor imaging (DTI) and quantitative T2 (qT2) mapping have both shown promise for assessing recovery of muscle micro trauma and exercise effects. The purpose of this study was to explore the potential of DTI and qT2 mapping for monitoring the muscle recovery processes after acute hamstring injury. In this prospective study, athletes with an acute hamstring injury underwent a 3-T MRI examination of the injured and contralateral hamstrings including DTI and qT2 measurements at three time points: (1) within 1 week after sustaining the injury, (2) 2 weeks after time point 1, and (3) return to play (RTP). A linear mixed model was used for time-effect analysis and paired t-tests for the detection of differences between injured and uninjured muscles. Forty-one athletes (age 27.8 ± 7 years; two females and 39 males) were included. Mean RTP time was 50 (range 12-169) days. A significant time effect was found for mean diffusivity, radial diffusivity, and the second and third eigenvalues (p ≤ 0.001) in the injured muscles. Fractional anisotropy (p = 0.40), first eigenvalue (p = 0.02), and qT2 (p = 0.61) showed no significant time effect. All DTI indices, except for fractional anisotropy, were significantly elevated compared with control muscles right after the injury (p < 0.001). Values normalized during the recovery period, with no significant differences between control and injured muscles at RTP (p values ranged from 0.08 to 0.51). Mean qT2 relaxation times in injured muscles were not significantly elevated compared with control muscles at any time point (p > 0.04). In conclusion, DTI can be used to monitor recovery after an acute hamstring injury. Future work should explore the potential of DTI indices to predict RTP and recovery times in athletes after an acute strain injury.
Collapse
Affiliation(s)
- Jithsa R Monte
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Melissa T Hooijmans
- Department of Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Martijn Froeling
- Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jos Oudeman
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Johannes L Tol
- Department of Orthopaedic Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
- Academic Center for Evidence Based Sports Medicine (ACES), Amsterdam, the Netherlands
- Amsterdam Collaboration for Health and Safety in Sports (ACHSS), AMC/VUmc IOC Research Center, Amsterdam, the Netherlands
| | - Gustav J Strijkers
- Department of Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Aart J Nederveen
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| | - Mario Maas
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
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
|
20
|
Kan HE. Effects of muscle damage on 31 P-MRS indices of energetic status and sarcolemma integrity in young mdx mice. NMR IN BIOMEDICINE 2022; 35:e4688. [PMID: 35060211 DOI: 10.1002/nbm.4688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 06/14/2023]
Abstract
The article of Lopez et al describes the use of a multi-parametric MR approach to study muscle T2 relaxation times and 31 P-MRS indices of energetics and sarcolemma integrity in a mouse model of DMD, the mdx mouse. Muscular dystrophies have a multi-factorial disease cascade, and there are several MR methods used to assess these. Aspects that reflect disease progression are outlined on the left, while features that are more related to disease activity are outlined on the right.
Collapse
Affiliation(s)
- Hermien E Kan
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Duchenne Center, The Netherlands
| |
Collapse
|
21
|
Kaslow JA, Sokolow AG, Donnelly T, Buchowski MS, Damon BM, Markham LW, Burnette WB, Soslow J. Leveraging Cardiac Magnetic Resonance Imaging to Assess Skeletal Muscle Progression in Duchenne Muscular Dystrophy. Neuromuscul Disord 2022; 32:390-398. [PMID: 35300894 PMCID: PMC9117482 DOI: 10.1016/j.nmd.2022.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 01/11/2022] [Accepted: 01/21/2022] [Indexed: 10/19/2022]
Abstract
Duchenne muscular dystrophy (DMD) is characterized by muscle deterioration and progressive weakness. As a result, patients with DMD have significant cardiopulmonary morbidity and mortality that worsens with age and loss of ambulation. Since most validated muscle assessments require ambulation, new functional measures of DMD progression are needed. Despite several evaluation methods available for monitoring disease progression, the relationship between these measures is unknown. We sought to assess the correlation between imaging metrics obtained from cardiac magnetic resonance imaging (CMR) and functional assessments including quantitative muscle testing (QMT), spirometry, and accelerometry. Forty-nine patients with DMD were enrolled and underwent CMR, accelerometry and QMT at baseline, 1-year and 2-year clinic visits with temporally associated pulmonary function testing obtained from the medical record. Imaging of the upper extremity musculature (triceps and biceps) demonstrated the most robust correlations with accelerometry (p<0.03), QMT (p<0.02) and spirometry (p<0.01). T1-mapping of serratus anterior muscle showed a similar, but slightly weaker relationship with accelerometry and QMT. T2-mapping of serratus anterior demonstrated weak indirect correlation with aspects of accelerometry. These images are either routinely obtained in standard CMR or can be added to a protocol and may allow for a more comprehensive assessment of a patient's disease progression.
Collapse
|
22
|
Buckon CE, Sienko SE, Fowler EG, Bagley AM, Staudt LA, Sison-Williamson M, Heberer KR, McDonald CM, Sussman MD. A Longitudinal Study of Quantitative Muscle Strength and Functional Motor Ability in Ambulatory Boys with Duchenne Muscular Dystrophy. J Neuromuscul Dis 2021; 9:321-334. [PMID: 34924398 DOI: 10.3233/jnd-210704] [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 Duchenne muscular dystrophy (DMD) is an X-linked recessive genetic disorder, that is characterized by progressive muscle degeneration and loss of ambulation between 7-13 years of age. Novel pharmacological agents targeting the genetic defects and disease mechanisms are becoming available; however, corticosteroid (CS) therapy remains the standard of care. OBJECTIVE The purpose of this longitudinal study was to elucidate the effect of CS therapy on the rate of muscle strength and gross motor skill decline in boys with DMD and assess the sensitivity of selected outcome measures. METHODS Eighty-four ambulatory boys with DMD (49-180 months), 70 on CS, 14 corticosteroid naïve (NCS), participated in this 8-year multi-site study. Outcomes included; isokinetic dynamometry, the Standing (STD) and Walking/Running/jumping (WRJ) dimensions of the Gross Motor Function Measure (GMFM), and Timed Function Tests (TFTs). Nonlinear mixed modeling procedures determined the rate of change with age and the influence of steroids. RESULTS Despite CS therapy the rate of decline in strength with age was significant in all muscle groups assessed. CS therapy significantly slowed decline in knee extensor strength, as the NCS group declined at 3x the rate of the CS group. Concurrently, WRJ skills declined in the NCS group at twice the rate of the CS group. 4-stair climb and 10 meter walk/run performance was superior in the boys on CS therapy. CONCLUSION CS therapy slowed the rate of muscle strength decline and afforded longer retention of select gross motor skills in boys on CS compared to boys who were NCS. Isokinetic dynamometry, Walk/Run/Jump skills, and select TFTs may prove informative in assessing the efficacy of new therapeutics in ambulatory boys with DMD.
Collapse
Affiliation(s)
| | | | - Eileen G Fowler
- Department of Orthopaedics, University of California, Los Angeles, CA, California
| | - Anita M Bagley
- Shriners Hospitals for Children, Northern California, CA, California
| | - Loretta A Staudt
- Department of Orthopaedics, University of California, Los Angeles, CA, California
| | | | - Kent R Heberer
- Department of Orthopaedics, University of California, Los Angeles, CA, California
| | - Craig M McDonald
- Department of Physical Medicine, University of California Davis Medical Center, Sacramento, CA, California
| | | |
Collapse
|
23
|
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
|
24
|
Wang L, Xu M, Liu D, Liang Y, Feng P, Li H, Zhu Y, He R, Lin J, Zhang H, Liao Z, Zhang C. Serum creatinine as a biomarker for dystrophinopathy: a cross-sectional and longitudinal study. BMC Neurol 2021; 21:372. [PMID: 34563158 PMCID: PMC8464115 DOI: 10.1186/s12883-021-02382-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 08/28/2021] [Indexed: 12/03/2022] Open
Abstract
Background Dystrophinopathy, a common neuromuscular disorder, includes Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD). Many researches are currently ongoing to develop curative approaches, which results in an urgent need for biomarkers of disease progression and treatment response. This study investigated whether the serum creatinine (SCRN) level can be used as a biomarker of disease progression in dystrophinopathy. Methods We enrolled 377 male patients with dystrophinopathy and 520 male non-dystrophinopathy controls in a cross-sectional study. From this cohort, 113 follow-up patients were enrolled in a longitudinal study. Patients’ demographic information, motor function, muscle fatty infiltration, and muscle dystrophin levels were evaluated. We investigated correlations between these parameters and SCRN levels, and determined changes in SCRN levels with maturation and with motor function changes. Results Our results showed SCRN levels correlated with motor function (FDR < 0.001) and timed test results (FDR between < 0.001–0.012), as well as with muscle fatty infiltration (FDR < 0.001) and dystrophin levels (FDR = 0.015 and 0.001). SCRN levels increased with maturation in control individuals; it slowly increased with maturation in patients with BMD but decreased generally with maturation in patients with DMD. The longitudinal study further demonstrated that SCRN levels were associated with motor function. Conclusions These findings indicated that the SCRN level is a promising biomarker for assessing disease progression in dystrophinopathy and could be used as a potential outcome measure in clinical trials. Supplementary Information The online version contains supplementary material available at 10.1186/s12883-021-02382-7.
Collapse
Affiliation(s)
- Liang Wang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Min Xu
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou Medical University, No. 250 Changgang East Road, Guangzhou, 510260, China
| | - Dawei Liu
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Yingyin Liang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Pinning Feng
- Department of Laboratory, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Huan Li
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Yuling Zhu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Ruojie He
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Jinfu Lin
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Huili Zhang
- Department of Neurology, Guangzhou First People's Hospital, No. 1 Panfu Road, Guangzhou, 510180, China
| | - Ziyu Liao
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Cheng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou, 510080, China.
| |
Collapse
|
25
|
Batra A, Lott DJ, Willcocks R, Forbes SC, Triplett W, Dastgir J, Yun P, Reghan Foley A, Bönnemann CG, Vandenborne K, Walter GA. Lower Extremity Muscle Involvement in the Intermediate and Bethlem Myopathy Forms of COL6-Related Dystrophy and Duchenne Muscular Dystrophy: A Cross-Sectional Study. J Neuromuscul Dis 2021; 7:407-417. [PMID: 32538860 DOI: 10.3233/jnd-190457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Collagen VI-related dystrophies (COL6-RDs) and Duchenne muscular dystrophy (DMD) cause progressive muscle weakness and disability. COL6-RDs are caused by mutations in the COL6 genes (COL6A1, COL6A2 and COL6A3) encoding the extracellular matrix protein collagen VI, and DMD is caused by mutations in the DMD gene encoding the cytoplasmic protein dystrophin. Both COL6-RDs and DMD are characterized by infiltration of the muscles by fatty and fibrotic tissue. This study examined the effect of disease pathology on skeletal muscles in lower extremity muscles of COL6-RDs using timed functional tests, strength measures and qualitative/ quantitative magnetic resonance imaging/spectroscopy measures (MRI/MRS) in comparison to unaffected (control) individuals. Patients with COL6-RD were also compared to age and gender matched patients with DMD.Patients with COL6-RD presented with a typical pattern of fatty infiltration of the muscle giving rise to an apparent halo effect around the muscle, while patients with DMD had evidence of fatty infiltration throughout the muscle areas imaged. Quantitatively, fat fraction, and transverse relaxation time (T2) were elevated in both COL6-RD and DMD patients compared to unaffected (control) individuals. Patients with COL6-RD had widespread muscle atrophy, likely contributing to weakness. In contrast, patients with DMD revealed force deficits even in muscle groups with increased contractile areas.
Collapse
Affiliation(s)
- Abhinandan Batra
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Donovan J Lott
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Rebecca Willcocks
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Sean C Forbes
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - William Triplett
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Jahannaz Dastgir
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Pomi Yun
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - A Reghan Foley
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Glenn A Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
26
|
Marty B, Reyngoudt H, Boisserie JM, Le Louër J, C A Araujo E, Fromes Y, Carlier PG. Water-Fat Separation in MR Fingerprinting for Quantitative Monitoring of the Skeletal Muscle in Neuromuscular Disorders. Radiology 2021; 300:652-660. [PMID: 34254855 DOI: 10.1148/radiol.2021204028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Background Quantitative MRI is increasingly proposed in clinical trials related to neuromuscular disorders (NMDs). Purpose To investigate the potential of an MR fingerprinting sequence for water and fat fraction (FF) quantification (MRF T1-FF) for providing markers of fatty replacement and disease activity in patients with NMDs and to establish the sensitivity of water T1 as a marker of disease activity compared with water T2 mapping. Materials and Methods Data acquired between March 2018 and March 2020 from the legs of patients with NMDs were retrospectively analyzed. The MRI examination comprised fat-suppressed T2-weighted imaging, mapping of the FF measured with the three-point Dixon technique (FFDixon), water T2 mapping, and MRF T1-FF, from which the FF measured with MRF T1-FF (FFMRF) and water T1 were derived. Data from the legs of healthy volunteers were prospectively acquired between January and July 2020 to derive abnormality thresholds for FF, water T2, and water T1 values. Kruskal-Wallis tests and receiver operating characteristic curve analysis were performed, and linear models were used. Results A total of 73 patients (mean age ± standard deviation, 47 years ± 12; 45 women) and 15 healthy volunteers (mean age, 33 years ± 8; three women) were evaluated. A linear correlation was observed between FFMRF and FFDixon (R2 = 0.97, P < .001). Water T1 values were higher in muscles with high signal intensity at fat-suppressed T2-weighted imaging than in muscles with low signal intensity (mean value, 1281 msec [95% CI: 1165, 1604] vs 1198 msec [95% CI: 1099, 1312], respectively; P < .001), and a correlation was found between water T1 and water T2 distribution metrics (R2 = 0.66 and 0.79 for the median and 90th percentile values, respectively; P < .001). Water T1 classified the patients' muscles as abnormal based on quantitative water T2, with high sensitivity (93%; 68 of 73 patients) and specificity (80%; 53 of 73 patients) (area under the receiver operating characteristic curve, 0.92 [95% CI: 0.83, 0.97]; P < .001). Conclusion Water-fat separation in MR fingerprinting is robust for deriving quantitative imaging markers of intramuscular fatty replacement and disease activity in patients with neuromuscular disorders. © RSNA, 2021 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Benjamin Marty
- From the Nuclear Magnetic Resonance Laboratory, Neuromuscular Investigation Center, Institute of Myology, Bâtiment Babinski, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Blvd Vincent Auriol, 75651 Paris Cedex 13, France; and Nuclear Magnetic Resonance Laboratory, CEA, DRF, IBFJ, Molecular Imaging Research Center, Paris, France
| | - Harmen Reyngoudt
- From the Nuclear Magnetic Resonance Laboratory, Neuromuscular Investigation Center, Institute of Myology, Bâtiment Babinski, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Blvd Vincent Auriol, 75651 Paris Cedex 13, France; and Nuclear Magnetic Resonance Laboratory, CEA, DRF, IBFJ, Molecular Imaging Research Center, Paris, France
| | - Jean-Marc Boisserie
- From the Nuclear Magnetic Resonance Laboratory, Neuromuscular Investigation Center, Institute of Myology, Bâtiment Babinski, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Blvd Vincent Auriol, 75651 Paris Cedex 13, France; and Nuclear Magnetic Resonance Laboratory, CEA, DRF, IBFJ, Molecular Imaging Research Center, Paris, France
| | - Julien Le Louër
- From the Nuclear Magnetic Resonance Laboratory, Neuromuscular Investigation Center, Institute of Myology, Bâtiment Babinski, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Blvd Vincent Auriol, 75651 Paris Cedex 13, France; and Nuclear Magnetic Resonance Laboratory, CEA, DRF, IBFJ, Molecular Imaging Research Center, Paris, France
| | - Ericky C A Araujo
- From the Nuclear Magnetic Resonance Laboratory, Neuromuscular Investigation Center, Institute of Myology, Bâtiment Babinski, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Blvd Vincent Auriol, 75651 Paris Cedex 13, France; and Nuclear Magnetic Resonance Laboratory, CEA, DRF, IBFJ, Molecular Imaging Research Center, Paris, France
| | - Yves Fromes
- From the Nuclear Magnetic Resonance Laboratory, Neuromuscular Investigation Center, Institute of Myology, Bâtiment Babinski, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Blvd Vincent Auriol, 75651 Paris Cedex 13, France; and Nuclear Magnetic Resonance Laboratory, CEA, DRF, IBFJ, Molecular Imaging Research Center, Paris, France
| | - Pierre G Carlier
- From the Nuclear Magnetic Resonance Laboratory, Neuromuscular Investigation Center, Institute of Myology, Bâtiment Babinski, Groupe Hospitalier Pitié-Salpêtrière, 47-83 Blvd Vincent Auriol, 75651 Paris Cedex 13, France; and Nuclear Magnetic Resonance Laboratory, CEA, DRF, IBFJ, Molecular Imaging Research Center, Paris, France
| |
Collapse
|
27
|
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
|
28
|
Sherlock SP, Zhang Y, Binks M, Marraffino S. Quantitative muscle MRI biomarkers in Duchenne muscular dystrophy: cross-sectional correlations with age and functional tests. Biomark Med 2021; 15:761-773. [PMID: 34155911 PMCID: PMC8253163 DOI: 10.2217/bmm-2020-0801] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/23/2021] [Indexed: 01/07/2023] Open
Abstract
Aim: Using baseline data from a clinical trial of domagrozumab in Duchenne muscular dystrophy, we evaluated the correlation between functional measures and quantitative MRI assessments of thigh muscle. Patients & methods: Analysis included timed functional tests, knee extension/strength and North Star Ambulatory Assessment. Patients (n = 120) underwent examinations of one thigh, with MRI sequences to enable measurements of muscle volume (MV), MV index, mean T2 relaxation time via T2-mapping and fat fraction. Results: MV was moderately correlated with strength assessments. MV index, fat fraction and T2-mapping measures had moderate correlations (r ∼ 0.5) to all functional tests, North Star Ambulatory Assessment and age. Conclusion: The moderate correlation between functional tests, age and baseline MRI measures supports MRI as a biomarker in Duchenne muscular dystrophy clinical trials. Trial registration: ClinicalTrials.gov, NCT02310763; registered 4 November 2014.
Collapse
Affiliation(s)
| | - Yao Zhang
- Pfizer Inc, Cambridge, MA 02139, USA
| | | | | |
Collapse
|
29
|
Naarding KJ, Keene KR, Sardjoe Mishre ASD, Veeger TTJ, van de Velde NM, Prins AJ, Burakiewicz J, Verschuuren JJGM, van der Holst M, Niks EH, Kan HE. Preserved thenar muscles in non-ambulant Duchenne muscular dystrophy patients. J Cachexia Sarcopenia Muscle 2021; 12:694-703. [PMID: 33963807 PMCID: PMC8200430 DOI: 10.1002/jcsm.12711] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/10/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Clinical trials in Duchenne muscular dystrophy (DMD) focus primarily on ambulant patients. Results cannot be extrapolated to later disease stages due to a decline in targeted muscle tissue. In non-ambulant DMD patients, hand function is relatively preserved and crucial for daily-life activities. We used quantitative MRI (qMRI) to establish whether the thenar muscles could be valuable to monitor treatment effects in non-ambulant DMD patients. METHODS Seventeen non-ambulant DMD patients (range 10.2-24.1 years) and 13 healthy controls (range 9.5-25.4 years) underwent qMRI of the right hand at 3 T at baseline. Thenar fat fraction (FF), total volume (TV), and contractile volume (CV) were determined using 4-point Dixon, and T2water was determined using multiecho spin-echo. Clinical assessments at baseline (n = 17) and 12 months (n = 13) included pinch strength (kg), performance of the upper limb (PUL) 2.0, DMD upper limb patient reported outcome measure (PROM), and playing a video game for 10 min using a game controller. Group differences and correlations were assessed with non-parametric tests. RESULTS Total volume was lower in patients compared with healthy controls (6.9 cm3 , 5.3-9.0 cm3 vs. 13.0 cm3 , 7.6-15.8 cm3 , P = 0.010). CV was also lower in patients (6.3 cm3 , 4.6-8.3 cm3 vs. 11.9 cm3 , 6.9-14.6 cm3 , P = 0.010). FF was slightly elevated (9.7%, 7.3-11.4% vs. 7.7%, 6.6-8.4%, P = 0.043), while T2water was higher (31.5 ms, 30.0-32.6 ms vs. 28.1 ms, 27.8-29.4 ms, P < 0.001). Pinch strength and PUL decreased over 12 months (2.857 kg, 2.137-4.010 to 2.243 kg, 1.930-3.339 kg, and 29 points, 20-36 to 23 points, 17-30, both P < 0.001), while PROM did not (49 points, 36-57 to 44 points, 30-54, P = 0.041). All patients were able to play for 10 min at baseline or follow-up, but some did not comply with the study procedures regarding this endpoint. Pinch strength correlated with TV and CV in patients (rho = 0.72 and rho = 0.68) and controls (both rho = 0.89). PUL correlated with TV, CV, and T2water (rho = 0.57, rho = 0.51, and rho = -0.59). CONCLUSIONS Low thenar FF, increased T2water , correlation of muscle size with strength and function, and the decrease in strength and function over 1 year indicate that the thenar muscles are a valuable and quantifiable target for therapy in later stages of DMD. Further studies are needed to relate these data to the loss of a clinically meaningful milestone.
Collapse
Affiliation(s)
- Karin J Naarding
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Kevin R Keene
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands
| | | | - Thom T J Veeger
- C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands
| | - Nienke M van de Velde
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Arina J Prins
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Jedrzej Burakiewicz
- C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands
| | - Jan J G M Verschuuren
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Menno van der Holst
- Duchenne Center, Leiden, Netherlands.,Department of Orthopedics, Rehabilitation and Physiotherapy, Leiden University Medical Center, Leiden, Netherlands
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Hermien E Kan
- Duchenne Center, Leiden, Netherlands.,C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands
| |
Collapse
|
30
|
The increasing role of muscle MRI to monitor changes over time in untreated and treated muscle diseases. Curr Opin Neurol 2021; 33:611-620. [PMID: 32796278 DOI: 10.1097/wco.0000000000000851] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW This review aims to discuss the recent results of studies published applying quantitative MRI sequences to large cohorts of patients with neuromuscular diseases. RECENT FINDINGS Quantitative MRI sequences are now available to identify and quantify changes in muscle water and fat content. These two components have been associated with acute and chronic injuries, respectively. Studies show that the increase in muscle water is not only reversible if therapies are applied successfully but can also predict fat replacement in neurodegenerative diseases. Muscle fat fraction correlates with muscle function tests and increases gradually over time in parallel with the functional decline of patients with neuromuscular diseases. There are new spectrometry-based sequences to quantify other components, such as glycogen, electrolytes or the pH of the muscle fibre, extending the applicability of MRI to the study of several processes in neuromuscular diseases. SUMMARY The latest results obtained from the study of long cohorts of patients with various neuromuscular diseases open the door to the use of this technology in clinical trials, which would make it possible to obtain a new measure for assessing the effectiveness of new treatments. The challenge is currently the popularization of these studies and their application to the monitoring of patients in the daily clinic.
Collapse
|
31
|
Finkel RS, Finanger E, Vandenborne K, Sweeney HL, Tennekoon G, Shieh PB, Willcocks R, Walter G, Rooney WD, Forbes SC, Triplett WT, Yum SW, Mancini M, MacDougall J, Fretzen A, Bista P, Nichols A, Donovan JM. Disease-modifying effects of edasalonexent, an NF-κB inhibitor, in young boys with Duchenne muscular dystrophy: Results of the MoveDMD phase 2 and open label extension trial. Neuromuscul Disord 2021; 31:385-396. [PMID: 33678513 DOI: 10.1016/j.nmd.2021.02.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/12/2020] [Accepted: 02/01/2021] [Indexed: 12/18/2022]
Abstract
Chronic activation of NF-κB is a key driver of muscle degeneration and suppression of muscle regeneration in Duchenne muscular dystrophy. Edasalonexent (CAT-1004) is an orally-administered novel small molecule that covalently links two bioactive compounds (salicylic acid and docosahexaenoic acid) that inhibit NF-κB. This placebo-controlled, proof-of-concept phase 2 study with open-label extension in boys ≥4-<8 years old with any dystrophin mutation examined the effect of edasalonexent (67 or 100 mg/kg/day) compared to placebo or off-treatment control. Endpoints were safety/tolerability, change from baseline in MRI T2 relaxation time of lower leg muscles and functional assessment, as well as pharmacodynamics and biomarkers. Treatment was well-tolerated and the majority of adverse events were mild, and most commonly of the gastrointestinal system (primarily diarrhea). There were no serious adverse events in the edasalonexent groups. Edasalonexent 100 mg/kg was associated with slowing of disease progression and preservation of muscle function compared to an off-treatment control period, with decrease in levels of NF-κB-regulated genes and improvements in biomarkers of muscle health and inflammation. These results support investigating edasalonexent in future trials and have informed the design of the edasalonexent phase 3 clinical trial in boys with Duchenne.
Collapse
Affiliation(s)
- Richard S Finkel
- St. Jude Children's Research Hospital, Memphis, TN and Nemours Children's Hospital, Orlando, FL, United States.
| | - Erika Finanger
- Oregon Health & Science University, Portland, OR, United States
| | | | - H Lee Sweeney
- University of Florida Health, Gainesville, FL, United States
| | - Gihan Tennekoon
- The Children's Hospital of Philadelphia, and the University of Pennsylvania, Philadelphia, PA, United States
| | - Perry B Shieh
- University of California, Los Angeles, Los Angeles, CA, United States
| | | | - Glenn Walter
- University of Florida Health, Gainesville, FL, United States
| | | | - Sean C Forbes
- University of Florida Health, Gainesville, FL, United States
| | | | - Sabrina W Yum
- The Children's Hospital of Philadelphia, and the University of Pennsylvania, Philadelphia, PA, United States
| | - Maria Mancini
- Catabasis Pharmaceuticals, Inc., Boston, MA, United States
| | | | | | - Pradeep Bista
- Catabasis Pharmaceuticals, Inc., Boston, MA, United States
| | - Andrew Nichols
- Catabasis Pharmaceuticals, Inc., Boston, MA, United States
| | | |
Collapse
|
32
|
Güttsches AK, Rehmann R, Schreiner A, Rohm M, Forsting J, Froeling M, Tegenthoff M, Vorgerd M, Schlaffke L. Quantitative Muscle-MRI Correlates with Histopathology in Skeletal Muscle Biopsies. J Neuromuscul Dis 2021; 8:669-678. [PMID: 33814461 DOI: 10.3233/jnd-210641] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Skeletal muscle biopsy is one of the gold standards in the diagnostic workup of muscle disorders. By histopathologic analysis, characteristic features like inflammatory cellular infiltrations, fat and collagen replacement of muscle tissue or structural defects of the myofibers can be detected. In the past years, novel quantitative MRI (qMRI) techniques have been developed to quantify tissue parameters, thus providing a non-invasive diagnostic tool in several myopathies. OBJECTIVE This proof-of-principle study was performed to validate the qMRI-techniques to skeletal muscle biopsy results. METHODS Ten patients who underwent skeletal muscle biopsy for diagnostic purposes were examined by qMRI. Fat fraction, water T2-time and diffusion parameters were measured in the muscle from which the biopsy was taken. The proportion of fat tissue, the severity of degenerative and inflammatory parameters and the amount of type 1- and type 2- muscle fibers were determined in all biopsy samples. The qMRI-data were then correlated to the histopathological findings. RESULTS The amount of fat tissue in skeletal muscle biopsy correlated significantly with the fat fraction derived from the Dixon sequence. The water T2-time, a parameter for tissue edema, correlated with the amount of vacuolar changes of myofibers and endomysial macrophages in the histopathologic analysis. No significant correlations were found for diffusion parameters. CONCLUSION In this proof-of-principle study, qMRI techniques were related to characteristic histopathologic features in neuromuscular disorders. The study provides the basis for further development of qMRI methods in the follow-up of patients with neuromuscular disorders, especially in the context of emerging treatment strategies.
Collapse
Affiliation(s)
- Anne-Katrin Güttsches
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Robert Rehmann
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Anja Schreiner
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Marlena Rohm
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Johannes Forsting
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Martijn Froeling
- Department of Radiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Martin Tegenthoff
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Matthias Vorgerd
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Lara Schlaffke
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
33
|
Otto LA, Froeling M, van Eijk RP, Asselman F, Wadman R, Cuppen I, Hendrikse J, van der Pol W. Quantification of disease progression in spinal muscular atrophy with muscle MRI-a pilot study. NMR IN BIOMEDICINE 2021; 34:e4473. [PMID: 33480130 PMCID: PMC7988555 DOI: 10.1002/nbm.4473] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/30/2020] [Indexed: 05/02/2023]
Abstract
OBJECTIVES Quantitative MRI (qMRI) of muscles is a promising tool to measure disease progression or to assess therapeutic effects in neuromuscular diseases. Longitudinal imaging studies are needed to show sensitivity of qMRI in detecting disease progression in spinal muscular atrophy (SMA). In this pilot study we therefore studied one-year changes in quantitative MR parameters in relation to clinical scores. METHODS We repeated quantitative 3 T MR analysis of thigh muscles and clinical testing one year after baseline in 10 treatment-naïve patients with SMA, 5 with Type 2 (21.6 ± 7.0 years) and 5 with Type 3 (33.4 ± 11.9 years). MR protocol consisted of Dixon, T2 mapping and diffusion tensor imaging (DTI). The temporal relation of parameters was examined with a mixed model. RESULTS We detected a significant increase in fat fraction (baseline, 38.2% SE 0.6; follow-up, 39.5% SE 0.6; +1.3%, p = 0.001) in all muscles. Muscles with moderate to high fat infiltration at baseline show a larger increase over time (+1.6%, p < 0.001). We did not find any changes in DTI parameters except for low fat-infiltration muscles (m. adductor longus and m. biceps femoris (short head)). The T2 of muscles decreased from 28.2 ms to 28.0 ms (p = 0.07). Muscle strength and motor function scores were not significantly different between follow-up and baseline. CONCLUSION Longitudinal imaging data show slow disease progression in skeletal muscle of the thigh of (young-) adult patients with SMA despite stable strength and motor function scores. Quantitative muscle imaging demonstrates potential as a biomarker for disease activity and monitoring of therapy response.
Collapse
Affiliation(s)
- Louise A.M. Otto
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Martijn Froeling
- Department of RadiologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Ruben P.A. van Eijk
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
- Biostatistics & Research Support, Julius Center for Health Sciences and Primary CareUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Fay‐Lynn Asselman
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Renske Wadman
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Inge Cuppen
- Department of Neurology and Child Neurology, UMC Utrecht Brain CenterUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Jeroen Hendrikse
- Department of RadiologyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - W‐Ludo van der Pol
- Department of Neurology, UMC Utrecht Brain CenterUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
34
|
Merlini L, Cecconi I, Parmeggiani A, Cordelli DM, Dormi A. Quadriceps muscle strength in Duchenne muscular dystrophy and effect of corticosteroid treatment. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2021; 39:200-206. [PMID: 33458575 PMCID: PMC7783426 DOI: 10.36185/2532-1900-023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 11/12/2022]
Abstract
Objectives In Duchenne muscular dystrophy, quadriceps weakness is recognized as a key factor in gait deterioration. The objective of this work was three-fold: first, to document the strength of the quadriceps in corticosteroid-naïve DMD boys; second, to measure the effect of corticosteroids on quadriceps strength; and third, to evaluate the correlation between baseline quadriceps strength and the age when starting corticosteroids with the loss of ambulation. Methods Quadriceps muscle strength using hand-held dynamometry was measured in 12 ambulant DMD boys who had never taken corticosteroids and during corticosteroid treatment until the loss of ambulation. Results Baseline quadriceps muscle strength at 6 years of age was 28% that of normal children of the same age; it decreased to 15% at 8 years and to 6% at 10 years. The increase in quadriceps muscle strength obtained after 1 year of corticosteroid treatment had a strong direct correlation with the baseline strength (R = 0.96). With corticosteroid treatment, the age of ambulation loss showed a very strong direct relationship (R = 0.92) with baseline quadriceps muscle strength but only a very weak inverse relationship (R = -0.73) with the age of starting treatment. Age of loss of ambulation was 10.3 ± 0.5 vs 19.1 ± 4.7 (P < 0.05) in children with baseline quadriceps muscle strength less than or greater than 40 N, respectively. Conclusions Corticosteroid-naïve DMD boys have a quantifiable severe progressive quadriceps weakness. This long-term study, for the first time, shows that both of the positive effects obtained with CS treatment, i.e. increasing quadriceps strength and delaying the loss of ambulation, have a strong and direct correlation with baseline quadriceps muscle strength. As such, hand-held dynamometry may be a useful tool in the routine physical examination and during clinical trial assessment.
Collapse
Affiliation(s)
- Luciano Merlini
- Department of Biomedical and Neuromotor Sciences DIBINEM, University of Bologna, Italy
| | - Ilaria Cecconi
- Child Neurology and Psychiatry Unit, S. Orsola Hospital, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Italy
| | - Antonia Parmeggiani
- Child Neurology and Psychiatry Unit, S. Orsola Hospital, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Italy
| | - Duccio Maria Cordelli
- Child Neurology and Psychiatry Unit, S. Orsola Hospital, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Italy
| | - Ada Dormi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Italy
| |
Collapse
|
35
|
Alic L, Griffin JF, Eresen A, Kornegay JN, Ji JX. Using MRI to quantify skeletal muscle pathology in Duchenne muscular dystrophy: A systematic mapping review. Muscle Nerve 2021; 64:8-22. [PMID: 33269474 PMCID: PMC8247996 DOI: 10.1002/mus.27133] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 12/11/2022]
Abstract
There is a great demand for accurate non‐invasive measures to better define the natural history of disease progression or treatment outcome in Duchenne muscular dystrophy (DMD) and to facilitate the inclusion of a large range of participants in DMD clinical trials. This review aims to investigate which MRI sequences and analysis methods have been used and to identify future needs. Medline, Embase, Scopus, Web of Science, Inspec, and Compendex databases were searched up to 2 November 2019, using keywords “magnetic resonance imaging” and “Duchenne muscular dystrophy.” The review showed the trend of using T1w and T2w MRI images for semi‐qualitative inspection of structural alterations of DMD muscle using a diversity of grading scales, with increasing use of T2map, Dixon, and MR spectroscopy (MRS). High‐field (>3T) MRI dominated the studies with animal models. The quantitative MRI techniques have allowed a more precise estimation of local or generalized disease severity. Longitudinal studies assessing the effect of an intervention have also become more prominent, in both clinical and animal model subjects. Quality assessment of the included longitudinal studies was performed using the Newcastle‐Ottawa Quality Assessment Scale adapted to comprise bias in selection, comparability, exposure, and outcome. Additional large clinical trials are needed to consolidate research using MRI as a biomarker in DMD and to validate findings against established gold standards. This future work should use a multiparametric and quantitative MRI acquisition protocol, assess the repeatability of measurements, and correlate findings to histologic parameters.
Collapse
Affiliation(s)
- Lejla Alic
- Department of Electrical & Computer Engineering, Texas A&M University, Doha, Qatar.,Magnetic Detection and Imaging group, Technical Medical Centre, University of Twente, The Netherlands
| | - John F Griffin
- College of Vet. Med. & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Aydin Eresen
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Department of Electrical & Computer Engineering, Texas A&M University, College Station, Texas, USA
| | - Joe N Kornegay
- College of Vet. Med. & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Jim X Ji
- Department of Electrical & Computer Engineering, Texas A&M University, Doha, Qatar.,Department of Electrical & Computer Engineering, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
36
|
Willcocks RJ, Forbes SC, Walter GA, Sweeney L, Rodino-Klapac LR, Mendell JR, Vandenborne K. Assessment of rAAVrh.74.MHCK7.micro-dystrophin Gene Therapy Using Magnetic Resonance Imaging in Children With Duchenne Muscular Dystrophy. JAMA Netw Open 2021; 4:e2031851. [PMID: 33394000 PMCID: PMC7783546 DOI: 10.1001/jamanetworkopen.2020.31851] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
This case-control study uses magnetic resonance imaging and spectroscopy to evaluate the association between treatment with recombinant adeno-associated virus serotype rh74 (rAAVrh74) and muscle quality in children with Duchenne muscular dystrophy.
Collapse
Affiliation(s)
| | - Sean C. Forbes
- Department of Physical Therapy, University of Florida, Gainesville
| | - Glenn A. Walter
- Department of Physical Therapy, University of Florida, Gainesville
| | - Lee Sweeney
- Department of Physical Therapy, University of Florida, Gainesville
| | | | - Jerry R. Mendell
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio
| | | |
Collapse
|
37
|
Quattrocelli M, Zelikovich AS, Salamone IM, Fischer JA, McNally EM. Mechanisms and Clinical Applications of Glucocorticoid Steroids in Muscular Dystrophy. J Neuromuscul Dis 2021; 8:39-52. [PMID: 33104035 PMCID: PMC7902991 DOI: 10.3233/jnd-200556] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glucocorticoid steroids are widely used as immunomodulatory agents in acute and chronic conditions. Glucocorticoid steroids such as prednisone and deflazacort are recommended for treating Duchenne Muscular Dystrophy where their use prolongs ambulation and life expectancy. Despite this benefit, glucocorticoid use in Duchenne Muscular Dystrophy is also associated with significant adverse consequences including adrenal suppression, growth impairment, poor bone health and metabolic syndrome. For other forms of muscular dystrophy like the limb girdle dystrophies, glucocorticoids are not typically used. Here we review the experimental evidence supporting multiple mechanisms of glucocorticoid action in dystrophic muscle including their role in dampening inflammation and myofiber injury. We also discuss alternative dosing strategies as well as novel steroid agents that are in development and testing, with the goal to reduce adverse consequences of prolonged glucocorticoid exposure while maximizing beneficial outcomes.
Collapse
Affiliation(s)
- Mattia Quattrocelli
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Molecular Cardiovascular Biology Division, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Aaron S Zelikovich
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Isabella M Salamone
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Julie A Fischer
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
38
|
Lott DJ, Taivassalo T, Cooke KD, Park H, Moslemi Z, Batra A, Forbes SC, Byrne BJ, Walter GA, Vandenborne K. Safety, feasibility, and efficacy of strengthening exercise in Duchenne muscular dystrophy. Muscle Nerve 2020; 63:320-326. [PMID: 33295018 DOI: 10.1002/mus.27137] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 12/02/2020] [Accepted: 12/06/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND This two-part study explored the safety, feasibility, and efficacy of a mild-moderate resistance isometric leg exercise program in ambulatory boys with Duchenne muscular dystrophy (DMD). METHODS First, we used a dose escalation paradigm with varying intensity and frequency of leg isometric exercise to determine the dose response and safety in 10 boys. Second, we examined safety and feasibility of a 12-wk in-home, remotely supervised, mild-moderate intensity strengthening program in eight boys. Safety measures included T2 MRI, creatine kinase levels, and pain. Peak strength and function (time to ascend/descend four stairs) were also measured. RESULTS Dose-escalation revealed no signs of muscle damage. Seven of the eight boys completed the 12-wk in-home program with a compliance of 84.9%, no signs of muscle damage, and improvements in strength (knee extensors P < .01; knee flexors P < .05) and function (descending steps P < .05). CONCLUSIONS An in-home, mild-moderate intensity leg exercise program is safe with potential to positively impact both strength and function in ambulatory boys with DMD.
Collapse
Affiliation(s)
- Donovan J Lott
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Tanja Taivassalo
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| | - Korey D Cooke
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Hyunjun Park
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Zahra Moslemi
- Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Abhinandan Batra
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Sean C Forbes
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Barry J Byrne
- Department of Pediatrics, University of Florida, Gainesville, Florida, 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
|
39
|
Lott DJ, Taivassalo T, Senesac CR, Willcocks RJ, Harrington AM, Zilke K, Cunkle H, Powers C, Finanger EL, Rooney WD, Tennekoon GI, Vandenborne K. Walking activity in a large cohort of boys with Duchenne muscular dystrophy. Muscle Nerve 2020; 63:192-198. [PMID: 33188573 DOI: 10.1002/mus.27119] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 01/30/2023]
Abstract
INTRODUCTION In this study we explored walking activity in a large cohort of boys with Duchenne muscular dystrophy (DMD). METHODS Step activity (monitored for 7 days), functional ability, and strength were quantified in ambulatory boys (5-12.9 years of age) with DMD and unaffected boys. Ambulatory status was determined 2 years later. RESULTS Two to 5 days of activity monitoring predicted weekly step activity (adjusted R2 = 0.80-0.95). Age comparisons revealed significant declines for step activity with increasing age, and relationships were found between step activity with both function and strength (P < .01). Our regression model predicted 36.5% of the variance in step activity. Those who were still ambulatory after 2 years demonstrated baseline step activity nearly double that of those who were no longer walking 2 years later (P < .01). DISCUSSION Step activity for DMD is related to and predictive of functional declines, which may be useful for clinical trials.
Collapse
Affiliation(s)
- Donovan J Lott
- Department of Physical Therapy, University of Florida, College of Public Health & Health Professions, Gainesville, Florida, USA
| | - Tanja Taivassalo
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| | - Claudia R Senesac
- Department of Physical Therapy, University of Florida, College of Public Health & Health Professions, Gainesville, Florida, USA
| | - Rebecca J Willcocks
- Department of Physical Therapy, University of Florida, College of Public Health & Health Professions, Gainesville, Florida, USA
| | - Ann M Harrington
- Department of Physical Therapy, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kirsten Zilke
- Oregon Health & Science University, Shriners Hospitals for Children, Portland, Oregon, USA
| | - Hilary Cunkle
- Department of Physical Therapy, University of Florida, College of Public Health & Health Professions, Gainesville, Florida, USA
| | - Catherine Powers
- Department of Physical Therapy, University of Florida, College of Public Health & Health Professions, Gainesville, Florida, USA
| | - Erika L Finanger
- Department of Pediatrics, Oregon Health and Science University, Portland, Oregon, USA
| | - William D Rooney
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, Oregon, USA
| | - Gihan I Tennekoon
- Department of Physical Therapy, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, College of Public Health & Health Professions, Gainesville, Florida, USA
| |
Collapse
|
40
|
Hooijmans MT, Froeling M, Koeks Z, Verschuuren JJ, Webb A, Niks EH, Kan HE. Multi-parametric MR in Becker muscular dystrophy patients. NMR IN BIOMEDICINE 2020; 33:e4385. [PMID: 32754921 PMCID: PMC7687231 DOI: 10.1002/nbm.4385] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 05/14/2023]
Abstract
Quantitative MRI and MRS of muscle are increasingly being used to measure individual pathophysiological processes in Becker muscular dystrophy (BMD). In particular, muscle fat fraction was shown to be highly associated with functional tests in BMD. However, the muscle strength per unit of contractile cross-sectional area is lower in patients with BMD compared with healthy controls. This suggests that the quality of the non-fat-replaced (NFR) muscle tissue is lower than in healthy controls. Consequently, a measure that reflects changes in muscle tissue itself is needed. Here, we explore the potential of water T2 relaxation times, diffusion parameters and phosphorus metabolic indices as early disease markers in patients with BMD. For this purpose, we examined these measures in fat-replaced (FR) and NFR lower leg muscles in patients with BMD and compared these values with those in healthy controls. Quantitative proton MRI (three-point Dixon, multi-spin-echo and diffusion-weighted spin-echo echo planar imaging) and 2D chemical shift imaging 31 P MRS data were acquired in 24 patients with BMD (age 18.8-66.2 years) and 13 healthy controls (age 21.3-63.6 years). Muscle fat fractions, phosphorus metabolic indices, and averages and standard deviations (SDs) of the water T2 relaxation times and diffusion tensor imaging (DTI) parameters were assessed in six individual leg muscles. Phosphodiester levels were increased in the NFR and FR tibialis anterior, FR peroneus and FR gastrocnemius lateralis muscles. No clear pattern was visible for the other metabolic indices. Increased T2 SD was found in the majority of FR muscles compared with NFR and healthy control muscles. No differences in average water T2 relaxation times or DTI indices were found between groups. Overall, our results indicate that primarily muscles that are further along in the disease process showed increases in T2 heterogeneity and changes in some metabolic indices. No clear differences were found for the DTI indices between groups.
Collapse
Affiliation(s)
- Melissa T. Hooijmans
- C.J. Gorter Center, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
- Department of Biomedical Engineering & PhysicsAmsterdam University Medical CentersAmsterdamThe Netherlands
| | - Martijn Froeling
- Department of RadiologyUtrecht University Medical CenterUtrechtThe Netherlands
| | - Zaida Koeks
- Department of NeurologyLeiden University Medical CenterLeidenThe Netherlands
| | - Jan J.G.M. Verschuuren
- Department of NeurologyLeiden University Medical CenterLeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| | - Andrew Webb
- C.J. Gorter Center, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Erik H. Niks
- Department of NeurologyLeiden University Medical CenterLeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| | - Hermien E. Kan
- C.J. Gorter Center, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| |
Collapse
|
41
|
Gerhalter T, Marty B, Gast LV, Porzelt K, Heiss R, Uder M, Schwab S, Carlier PG, Nagel AM, Türk M. Quantitative 1H and 23Na muscle MRI in Facioscapulohumeral muscular dystrophy patients. J Neurol 2020; 268:1076-1087. [PMID: 33047224 PMCID: PMC7914168 DOI: 10.1007/s00415-020-10254-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022]
Abstract
Objective Our aim was to assess the role of quantitative 1H and 23Na MRI methods in providing imaging biomarkers of disease activity and severity in patients with Facioscapulohumeral muscular dystrophy (FSHD). Methods We imaged the lower leg muscles of 19 FSHD patients and 12 controls with a multimodal MRI protocol to obtain STIR-T2w images, fat fraction (FF), water T2 (wT2), water T1 (wT1), tissue sodium concentration (TSC), and intracellular-weighted sodium signal (inversion recovery (IR) and triple quantum filter (TQF) sequence). In addition, the FSHD patients underwent muscle strength testing. Results Imaging biomarkers related with water mobility (wT1 and wT2) and ion homeostasis (TSC, IR, TQF) were increased in muscles of FSHD patients. Muscle groups with FF > 10% had higher wT2, wT1, TSC, IR, and TQF values than muscles with FF < 10%. Muscles with FF < 10% resembled muscles of healthy controls for these MRI disease activity measures. However, wT1 was increased in few muscles without fat replacement. Furthermore, few STIR-negative muscles (n = 11/76) exhibited increased wT1, TSC, IR or TQF. Increased wT1 as well as 23Na signals were also present in muscles with normal wT2. Muscle strength was related to the mean FF and all imaging biomarkers of tibialis anterior except wT2 were correlated with dorsal flexion. Conclusion The newly evaluated imaging biomarkers related with water mobility (wT1) and ion homeostasis (TSC, IR, TQF) showed different patterns compared to the established markers like FF in muscles of FSHD patients. These quantitative biomarkers could thus contain valuable complementary information for the early characterization of disease progression. Electronic supplementary material The online version of this article (10.1007/s00415-020-10254-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Teresa Gerhalter
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Maximiliansplatz 3, 91054, Erlangen, Germany.
| | - Benjamin Marty
- NMR Laboratory, Institute of Myology, Paris, France
- NMR Laboratory, CEA/DRF, IBFJ/MIRCen, Paris, France
| | - Lena V Gast
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Maximiliansplatz 3, 91054, Erlangen, Germany
- Institute of Medical Physics, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Katharina Porzelt
- Department of Neurology, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Rafael Heiss
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Maximiliansplatz 3, 91054, Erlangen, Germany
| | - Michael Uder
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Maximiliansplatz 3, 91054, Erlangen, Germany
| | - Stefan Schwab
- Department of Neurology, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Pierre G Carlier
- NMR Laboratory, Institute of Myology, Paris, France
- NMR Laboratory, CEA/DRF, IBFJ/MIRCen, Paris, France
| | - Armin M Nagel
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Maximiliansplatz 3, 91054, Erlangen, Germany
- Division of Medical Physics in Radiology, German Cancer Research Centre, Heidelberg, Germany
- Institute of Medical Physics, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Matthias Türk
- Department of Neurology, Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| |
Collapse
|
42
|
Dahlqvist JR, Widholm P, Leinhard OD, Vissing J. MRI in Neuromuscular Diseases: An Emerging Diagnostic Tool and Biomarker for Prognosis and Efficacy. Ann Neurol 2020; 88:669-681. [PMID: 32495452 DOI: 10.1002/ana.25804] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 05/05/2020] [Accepted: 05/25/2020] [Indexed: 12/12/2022]
Abstract
There is an unmet need to identify biomarkers sensitive to change in rare, slowly progressive neuromuscular diseases. Quantitative magnetic resonance imaging (MRI) of muscle may offer this opportunity, as it is noninvasive and can be carried out almost independent of patient cooperation and disease severity. Muscle fat content correlates with muscle function in neuromuscular diseases, and changes in fat content precede changes in function, which suggests that muscle MRI is a strong biomarker candidate to predict prognosis and treatment efficacy. In this paper, we review the evidence suggesting that muscle MRI may be an important biomarker for diagnosis and to monitor change in disease severity. ANN NEUROL 2020;88:669-681.
Collapse
Affiliation(s)
- Julia R Dahlqvist
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| | - Per Widholm
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
- AMRA Medical AB, Linköping, Sweden
| | - Olof Dahlqvist Leinhard
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
- AMRA Medical AB, Linköping, Sweden
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
43
|
Heskamp L, Okkersen K, van Nimwegen M, Ploegmakers MJ, Bassez G, Deux JF, van Engelen BG, Heerschap A. Quantitative Muscle MRI Depicts Increased Muscle Mass after a Behavioral Change in Myotonic Dystrophy Type 1. Radiology 2020; 297:132-142. [PMID: 32808888 DOI: 10.1148/radiol.2020192518] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background Patients with myotonic dystrophy type 1 (DM1) increased their physical activity and exercise capacity following a behavioral intervention. However, it is unknown what is altered in muscles of patients with DM1 as a result of this intervention. The increased exercise capacity suggests that decelerated fat infiltration or increased muscle cross-sectional area (CSA) could be involved. Purpose To assess the effect of this activity-stimulating behavioral intervention on the lower extremity muscles of patients with DM1 with longitudinal quantitative muscle MRI. Materials and Methods In this prospective trial, participants with DM1 were randomized to a behavioral intervention (n = 14) or continued regular care (standard care; n = 13); no age-matched pairing was performed. Participants underwent MRI of the lower extremities at baseline and 10-month follow-up (January 2015 to March 2016). Fat fraction (FF), muscle CSA, and muscle water T2 (T2water) as markers for fat infiltration, muscle mass, and alteration in tissue water distribution (edema), respectively, were assessed with a chemical shift-encoded Dixon sequence and multiecho spin-echo sequence. Longitudinal within-group and between-group changes were assessed with paired-samples t tests and multivariable regression models. Results A total of 27 patients with DM1 (15 men) were evaluated. Patient age was comparable between groups (intervention, 45 years ± 13 [standard deviation]; standard care, 5 years ± 12; P = .96). Muscle CSA increased 5.9 cm2 ± 7.8 in the intervention group during the 10-month follow-up (P = .03) and decreased 3.6 cm2 ± 7.2 in the standard care group (P = .13). After 10 months, the mean difference between the groups was 9.5 cm2 (P = .01). This effect was stronger in muscles with baseline FF below the mean ± standard deviation of unaffected volunteers (-0.4 cm2 ± 0.15; P < .001). FF increased 0.9% ± 1.0 in the intervention group (P = .02) and 1.2% ± 1.2 for standard care (P = .02), with no between-group difference (P = .56). T2water did not change significantly in either group (intervention, P = .08; standard care, P = .88). Conclusion A behavioral intervention targeting physical activity increased lower extremity muscle cross-sectional area in patients with myotonic dystrophy, preferentially in healthy-appearing muscle. © RSNA, 2020 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Linda Heskamp
- From the Department of Radiology and Nuclear Medicine, Radboud Institute for Molecular Life Sciences (L.H., M.J.P., A.H.), and Department of Neurology, Donders Institute for Brain, Cognition and Behaviour (K.O., M.v.N., B.G.v.E.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Neuromuscular Reference Center, Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital, Paris, France (G.B.); and Department of Radiology, Henri Mondor University Hospital, Paris, France (J.F.D.)
| | - Kees Okkersen
- From the Department of Radiology and Nuclear Medicine, Radboud Institute for Molecular Life Sciences (L.H., M.J.P., A.H.), and Department of Neurology, Donders Institute for Brain, Cognition and Behaviour (K.O., M.v.N., B.G.v.E.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Neuromuscular Reference Center, Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital, Paris, France (G.B.); and Department of Radiology, Henri Mondor University Hospital, Paris, France (J.F.D.)
| | - Marlies van Nimwegen
- From the Department of Radiology and Nuclear Medicine, Radboud Institute for Molecular Life Sciences (L.H., M.J.P., A.H.), and Department of Neurology, Donders Institute for Brain, Cognition and Behaviour (K.O., M.v.N., B.G.v.E.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Neuromuscular Reference Center, Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital, Paris, France (G.B.); and Department of Radiology, Henri Mondor University Hospital, Paris, France (J.F.D.)
| | - Marieke J Ploegmakers
- From the Department of Radiology and Nuclear Medicine, Radboud Institute for Molecular Life Sciences (L.H., M.J.P., A.H.), and Department of Neurology, Donders Institute for Brain, Cognition and Behaviour (K.O., M.v.N., B.G.v.E.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Neuromuscular Reference Center, Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital, Paris, France (G.B.); and Department of Radiology, Henri Mondor University Hospital, Paris, France (J.F.D.)
| | - Guillaume Bassez
- From the Department of Radiology and Nuclear Medicine, Radboud Institute for Molecular Life Sciences (L.H., M.J.P., A.H.), and Department of Neurology, Donders Institute for Brain, Cognition and Behaviour (K.O., M.v.N., B.G.v.E.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Neuromuscular Reference Center, Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital, Paris, France (G.B.); and Department of Radiology, Henri Mondor University Hospital, Paris, France (J.F.D.)
| | - Jean-Francois Deux
- From the Department of Radiology and Nuclear Medicine, Radboud Institute for Molecular Life Sciences (L.H., M.J.P., A.H.), and Department of Neurology, Donders Institute for Brain, Cognition and Behaviour (K.O., M.v.N., B.G.v.E.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Neuromuscular Reference Center, Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital, Paris, France (G.B.); and Department of Radiology, Henri Mondor University Hospital, Paris, France (J.F.D.)
| | - Baziel G van Engelen
- From the Department of Radiology and Nuclear Medicine, Radboud Institute for Molecular Life Sciences (L.H., M.J.P., A.H.), and Department of Neurology, Donders Institute for Brain, Cognition and Behaviour (K.O., M.v.N., B.G.v.E.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Neuromuscular Reference Center, Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital, Paris, France (G.B.); and Department of Radiology, Henri Mondor University Hospital, Paris, France (J.F.D.)
| | - Arend Heerschap
- From the Department of Radiology and Nuclear Medicine, Radboud Institute for Molecular Life Sciences (L.H., M.J.P., A.H.), and Department of Neurology, Donders Institute for Brain, Cognition and Behaviour (K.O., M.v.N., B.G.v.E.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Neuromuscular Reference Center, Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital, Paris, France (G.B.); and Department of Radiology, Henri Mondor University Hospital, Paris, France (J.F.D.)
| | -
- From the Department of Radiology and Nuclear Medicine, Radboud Institute for Molecular Life Sciences (L.H., M.J.P., A.H.), and Department of Neurology, Donders Institute for Brain, Cognition and Behaviour (K.O., M.v.N., B.G.v.E.), Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands; Neuromuscular Reference Center, Sorbonne University, INSERM UMRS 974, AP-HP, Pitié-Salpêtrière Hospital, Paris, France (G.B.); and Department of Radiology, Henri Mondor University Hospital, Paris, France (J.F.D.)
| |
Collapse
|
44
|
Araujo ECA, Marty B, Carlier PG, Baudin P, Reyngoudt H. Multiexponential Analysis of the Water
T2
‐Relaxation in the Skeletal Muscle Provides Distinct Markers of Disease Activity Between Inflammatory and Dystrophic Myopathies. J Magn Reson Imaging 2020; 53:181-189. [DOI: 10.1002/jmri.27300] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 11/06/2022] Open
Affiliation(s)
- Ericky C. A. Araujo
- NMR laboratory, Neuromuscular Investigation Center Institute of Myology Paris France
- CEA, DRF, IBFJ, MIRCen Paris France
| | - Benjamin Marty
- NMR laboratory, Neuromuscular Investigation Center Institute of Myology Paris France
- CEA, DRF, IBFJ, MIRCen Paris France
| | - Pierre G. Carlier
- NMR laboratory, Neuromuscular Investigation Center Institute of Myology Paris France
- CEA, DRF, IBFJ, MIRCen Paris France
| | | | - Harmen Reyngoudt
- NMR laboratory, Neuromuscular Investigation Center Institute of Myology Paris France
- CEA, DRF, IBFJ, MIRCen Paris France
| |
Collapse
|
45
|
Muscle MRI in two SMA patients on nusinersen treatment: A two years follow-up. J Neurol Sci 2020; 417:117067. [PMID: 32745721 PMCID: PMC7388822 DOI: 10.1016/j.jns.2020.117067] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/20/2020] [Accepted: 07/27/2020] [Indexed: 12/21/2022]
Abstract
Introduction The effects of nusinersen in adults with SMA rely on neuromotor function scales and qualitative assessments. There are limited clinical or imaging data on muscle changes over time. Methods Two adult SMA patients underwent clinical assessments including measures of upper and lower limb function with Revised Upper Limb Module (RULM) and Hammersmith Function Motor Scale Expanded (HFMSE); both patients were also studied with whole-body muscle MRI (T1-weighted and Diffusion Tensor Imaging/DTI sequences), at baseline and after 10 and 24 months from the beginning of treatment with nusinersen. Results After two years of treatment, HFMSE and RULM scores were stable in both patients. DTI sequences revealed an increased number, length and organization of muscle fiber tracks, and Fractional Anisotropy (FA) values showed a significant reduction after 10 and 24 months from baseline, in their corresponding maps. Discussion Muscle DTI imaging seems to play an interesting role to monitor treatment effects over time in adult SMA patients. Nusinersen treatment has created great expectations in older SMA patients having long-lasting muscular atrophy. DTI is a very sensitive technique to identify small changes in muscle architecture. DTI shows that nusinersen treatment may have a positive effect on size, length and organization of fiber tracts.
Collapse
|
46
|
Previtali SC, Gidaro T, Díaz-Manera J, Zambon A, Carnesecchi S, Roux-Lombard P, Spitali P, Signorelli M, Szigyarto CAK, Johansson C, Gray J, Labolle D, Porte Thomé F, Pitchforth J, Domingos J, Muntoni F. Rimeporide as a first- in-class NHE-1 inhibitor: Results of a phase Ib trial in young patients with Duchenne Muscular Dystrophy. Pharmacol Res 2020; 159:104999. [PMID: 32535224 PMCID: PMC7482441 DOI: 10.1016/j.phrs.2020.104999] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 12/23/2022]
Abstract
Rimeporide, a first-in-class sodium/proton exchanger Type 1 inhibitor (NHE-1 inhibitor) is repositioned by EspeRare for patients with Duchenne Muscular Dystrophy (DMD). Historically, NHE-1 inhibitors were developed for cardiac therapeutic interventions. There is considerable overlap in the pathophysiological mechanisms in Congestive Heart Failure (CHF) and in cardiomyopathy in DMD, therefore NHE-1 inhibition could be a promising pharmacological approach to the cardiac dysfunctions observed in DMD. Extensive preclinical data was collected in various animal models including dystrophin-deficient (mdx) mice to characterise Rimeporide’s anti-fibrotic and anti-inflammatory properties and there is evidence that NHE-1 inhibitors could play a significant role in modifying DMD cardiac and also skeletal pathologies, as the NHE-1 isoform is ubiquitous. We report here the first study with Rimeporide in DMD patients. This 4-week treatment, open label phase Ib, multiple oral ascending dose study, enrolled 20 ambulant boys with DMD (6–11 years), with outcomes including safety, pharmacokinetic (PK) and pharmacodynamic (PD) biomarkers. Rimeporide was safe and well-tolerated at all doses. PK evaluations showed that Rimeporide was well absorbed orally reaching pharmacological concentrations from the lowest dose, with exposure increasing linearly with dose and with no evidence of accumulation upon repeated dosing. Exploratory PD biomarkers showed positive effect upon a 4-week treatment, supporting its therapeutic potential in patients with DMD, primarily as a cardioprotective treatment, and provide rationale for further efficacy studies.
Collapse
Affiliation(s)
- Stefano C Previtali
- IRCCS San Raffaele Scientific Institute, Department of Neurology and INSPE, Milan, Italy
| | - Teresa Gidaro
- Institute of Myology, Hopital Trousseau, I- Motion, Paris, France
| | - Jordi Díaz-Manera
- Hospital de la Santa Creu i Sant Pau de Barcelona Servei de Neurologia, Barcelona, Spain; Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Spain
| | - Alberto Zambon
- IRCCS San Raffaele Scientific Institute, Department of Neurology and INSPE, Milan, Italy
| | | | - Pascale Roux-Lombard
- Geneva University Hospital (HUG), Immunology and Allergology Department, Geneva, Switzerland
| | | | | | | | - Camilla Johansson
- Science for Life Laboratory, Department of Protein Science, Division of Systems Biology, Solna, Sweden
| | | | | | | | - Jacqueline Pitchforth
- UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital Dubowitz Neuromuscular Centre, London, UK
| | - Joana Domingos
- UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital Dubowitz Neuromuscular Centre, London, UK
| | - Francesco Muntoni
- UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital Dubowitz Neuromuscular Centre, London, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, Great Ormond Street Hospital Trust, University College London, London, UK
| |
Collapse
|
47
|
Forbes SC, Arora H, Willcocks RJ, Triplett WT, Rooney WD, Barnard AM, Alabasi U, Wang DJ, Lott DJ, Senesac CR, Harrington AT, Finanger EL, Tennekoon GI, Brandsema J, Daniels MJ, Sweeney HL, Walter GA, Vandenborne K. Upper and Lower Extremities in Duchenne Muscular Dystrophy Evaluated with Quantitative MRI and Proton MR Spectroscopy in a Multicenter Cohort. Radiology 2020; 295:616-625. [PMID: 32286193 PMCID: PMC7263287 DOI: 10.1148/radiol.2020192210] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 02/05/2020] [Accepted: 02/13/2020] [Indexed: 12/18/2022]
Abstract
Background Upper extremity MRI and proton MR spectroscopy are increasingly considered to be outcome measures in Duchenne muscular dystrophy (DMD) clinical trials. Purpose To demonstrate the feasibility of acquiring upper extremity MRI and proton (1H) MR spectroscopy measures of T2 and fat fraction in a large, multicenter cohort (ImagingDMD) of ambulatory and nonambulatory individuals with DMD; compare upper and lower extremity muscles by using MRI and 1H MR spectroscopy; and correlate upper extremity MRI and 1H MR spectroscopy measures to function. Materials and Methods In this prospective cross-sectional study, MRI and 1H MR spectroscopy and functional assessment data were acquired from participants with DMD and unaffected control participants at three centers (from January 28, 2016, to April 24, 2018). T2 maps of the shoulder, upper arm, forearm, thigh, and calf were generated from a spin-echo sequence (repetition time msec/echo time msec, 3000/20-320). Fat fraction maps were generated from chemical shift-encoded imaging (eight echo times). Fat fraction and 1H2O T2 in the deltoid and biceps brachii were measured from single-voxel 1H MR spectroscopy (9000/11-243). Groups were compared by using Mann-Whitney test, and relationships between MRI and 1H MR spectroscopy and arm function were assessed by using Spearman correlation. Results This study evaluated 119 male participants with DMD (mean age, 12 years ± 3 [standard deviation]) and 38 unaffected male control participants (mean age, 12 years ± 3). Deltoid and biceps brachii muscles were different in participants with DMD versus control participants in all age groups by using quantitative T2 MRI (P < .001) and 1H MR spectroscopy fat fraction (P < .05). The deltoid, biceps brachii, and triceps brachii were affected to the same extent (P > .05) as the soleus and medial gastrocnemius. Negative correlations were observed between arm function and MRI (T2: range among muscles, ρ = -0.53 to -0.73 [P < .01]; fat fraction, ρ = -0.49 to -0.70 [P < .01]) and 1H MR spectroscopy fat fraction (ρ = -0.64 to -0.71; P < .01). Conclusion This multicenter study demonstrated early and progressive involvement of upper extremity muscles in Duchenne muscular dystrophy (DMD) and showed the feasibility of MRI and 1H MR spectroscopy to track disease progression over a wide range of ages in participants with DMD. © RSNA, 2020 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Sean C. Forbes
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Harneet Arora
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Rebecca J. Willcocks
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - William T. Triplett
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - William D. Rooney
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Alison M. Barnard
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Umar Alabasi
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Dah-Jyuu Wang
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Donovan J. Lott
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Claudia R. Senesac
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Ann T. Harrington
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Erika L. Finanger
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Gihan I. Tennekoon
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - John Brandsema
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Michael J. Daniels
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - H. Lee Sweeney
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Glenn A. Walter
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| | - Krista Vandenborne
- From the Department of Physical Therapy (S.C.F., H.A., R.J.W., W.T.T., A.M.B., U.A., D.J.L. C.R.S., K.V.), Department of Statistics (M.J.D.), Department of Pharmacology and Therapeutics (H.L.S.), and Department of Physiology and Functional Genomics (G.A.W.), University of Florida, Box 100154, UFHSC, Gainesville, FL 32610; Advanced Imaging Research Center, Oregon Health and Science University, Portland, Ore (W.D.R., E.L.F.); The Children’s Hospital of Philadelphia, Philadelphia, Pa (D.J.W., A.T.H., G.I.T., J.B.); and Department of Neurology, Shriners Hospital for Children, Portland, Ore (E.L.F.)
| |
Collapse
|
48
|
Rooney WD, Berlow YA, Triplett WT, Forbes SC, Willcocks RJ, Wang DJ, Arpan I, Arora H, Senesac C, Lott DJ, Tennekoon G, Finkel R, Russman BS, Finanger EL, Chakraborty S, O'Brien E, Moloney B, Barnard A, Sweeney HL, Daniels MJ, Walter GA, Vandenborne K. Modeling disease trajectory in Duchenne muscular dystrophy. Neurology 2020; 94:e1622-e1633. [PMID: 32184340 DOI: 10.1212/wnl.0000000000009244] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 10/17/2019] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To quantify disease progression in individuals with Duchenne muscular dystrophy (DMD) using magnetic resonance biomarkers of leg muscles. METHODS MRI and magnetic resonance spectroscopy (MRS) biomarkers were acquired from 104 participants with DMD and 51 healthy controls using a prospective observational study design with patients with DMD followed up yearly for up to 6 years. Fat fractions (FFs) in vastus lateralis and soleus muscles were determined with 1H MRS. MRI quantitative T2 (qT2) values were measured for 3 muscles of the upper leg and 5 muscles of the lower leg. Longitudinal changes in biomarkers were modeled with a cumulative distribution function using a nonlinear mixed-effects approach. RESULTS MRS FF and MRI qT2 increased with DMD disease duration, with the progression time constants differing markedly between individuals and across muscles. The average age at half-maximal muscle involvement (μ) occurred 4.8 years earlier in vastus lateralis than soleus, and these measures were strongly associated with loss-of-ambulation age. Corticosteroid treatment was found to delay μ by 2.5 years on average across muscles, although there were marked differences between muscles with more slowly progressing muscles showing larger delay. CONCLUSIONS MRS FF and MRI qT2 provide sensitive noninvasive measures of DMD progression. Modeling changes in these biomarkers across multiple muscles can be used to detect and monitor the therapeutic effects of corticosteroids on disease progression and to provide prognostic information on functional outcomes. This modeling approach provides a method to transform these MRI biomarkers into well-understood metrics, allowing concise summaries of DMD disease progression at individual and population levels. CLINICALTRIALSGOV IDENTIFIER NCT01484678.
Collapse
Affiliation(s)
- William D Rooney
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR.
| | - Yosef A Berlow
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - William T Triplett
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Sean C Forbes
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Rebecca J Willcocks
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Dah-Jyuu Wang
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Ishu Arpan
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Harneet Arora
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Claudia Senesac
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Donovan J Lott
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Gihan Tennekoon
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Richard Finkel
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Barry S Russman
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Erika L Finanger
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Saptarshi Chakraborty
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Elliott O'Brien
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Brendan Moloney
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Alison Barnard
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - H Lee Sweeney
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Michael J Daniels
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Glenn A Walter
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| | - Krista Vandenborne
- From the Advanced Imaging Research Center (W.D.R., Y.A.B., I.A., E.O., B.M.), Department of Neurology (W.D.R., I.A., B.S.R., E.L.F.), Department of Biomedical Engineering (W.D.R.), Department of Behavioral Neuroscience (W.D.R., Y.A.B.), and Department of Pediatrics (B.S.R., E.L.F.), Oregon Health & Science University, Portland; Departments of Physical Therapy (W.T.T., S.C.F., R.J.W., H.A., C.S., D.J.L., K.V.), Statistics (S.C., M.J.D.), Physiology and Functional Genomics (A.B., G.A.W.), and Pharmacology & Therapeutics (H.L.S.), University of Florida, Gainesville; Department of Radiology (D.-J.W.) and Division of Neurology (G.T.), Children's Hospital of Philadelphia, PA; Department of Pediatrics (R.F.), Nemours Children's Hospital, Orlando, FL; and Shriners Hospital (B.S.R., E.L.F.), Portland, OR
| |
Collapse
|
49
|
Barnard AM, Willcocks RJ, Triplett WT, Forbes SC, Daniels MJ, Chakraborty S, Lott DJ, Senesac CR, Finanger EL, Harrington AT, Tennekoon G, Arora H, Wang DJ, Sweeney HL, Rooney WD, Walter GA, Vandenborne K. MR biomarkers predict clinical function in Duchenne muscular dystrophy. Neurology 2020; 94:e897-e909. [PMID: 32024675 PMCID: PMC7238941 DOI: 10.1212/wnl.0000000000009012] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 08/29/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE To investigate the potential of lower extremity magnetic resonance (MR) biomarkers to serve as endpoints in clinical trials of therapeutics for Duchenne muscular dystrophy (DMD) by characterizing the longitudinal progression of MR biomarkers over 48 months and assessing their relationship to changes in ambulatory clinical function. METHODS One hundred sixty participants with DMD were enrolled in this longitudinal, natural history study and underwent MR data acquisition of the lower extremity muscles to determine muscle fat fraction (FF) and MRI T2 biomarkers of disease progression. In addition, 4 tests of ambulatory function were performed. Participants returned for follow-up data collection at 12, 24, 36, and 48 months. RESULTS Longitudinal analysis of the MR biomarkers revealed that vastus lateralis FF, vastus lateralis MRI T2, and biceps femoris long head MRI T2 biomarkers were the fastest progressing biomarkers over time in this primarily ambulatory cohort. Biomarker values tended to demonstrate a nonlinear, sigmoidal trajectory over time. The lower extremity biomarkers predicted functional performance 12 and 24 months later, and the magnitude of change in an MR biomarker over time was related to the magnitude of change in function. Vastus lateralis FF, soleus FF, vastus lateralis MRI T2, and biceps femoris long head MRI T2 were the strongest predictors of future loss of function, including loss of ambulation. CONCLUSIONS This study supports the strong relationship between lower extremity MR biomarkers and measures of clinical function, as well as the ability of MR biomarkers, particularly those from proximal muscles, to predict future ambulatory function and important clinical milestones. CLINICALTRIALSGOV IDENTIFIER NCT01484678.
Collapse
Affiliation(s)
- Alison M Barnard
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Rebecca J Willcocks
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - William T Triplett
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Sean C Forbes
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Michael J Daniels
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Saptarshi Chakraborty
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Donovan J Lott
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Claudia R Senesac
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Erika L Finanger
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Ann T Harrington
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Gihan Tennekoon
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Harneet Arora
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Dah-Jyuu Wang
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - H Lee Sweeney
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - William D Rooney
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Glenn A Walter
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA
| | - Krista Vandenborne
- From the Departments of Physical Therapy (A.M.B., R.J.W., W.T.T., S.C.F., D.J.L., C.R.S., H.A., K.V.), Statistics (M.J.D., S.C.), Pharmacology and Therapeutics (H.L.S.), and Physiology and Functional Genomics (G.A.W.), University of Florida, Gainesville; Departments of Pediatrics and Neurology (E.L.F., G.T., D.-J.W.) and Advanced Imaging Research Center (W.D.R.), Oregon Health & Science University, Portland; and Children's Hospital of Philadelphia (A.T.H.), PA.
| |
Collapse
|
50
|
Maggi L, Moscatelli M, Frangiamore R, Mazzi F, Verri M, De Luca A, Pasanisi MB, Baranello G, Tramacere I, Chiapparini L, Bruzzone MG, Mantegazza R, Aquino D. Quantitative Muscle MRI Protocol as Possible Biomarker in Becker Muscular Dystrophy. Clin Neuroradiol 2020; 31:257-266. [PMID: 31974637 DOI: 10.1007/s00062-019-00875-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/30/2019] [Indexed: 12/16/2022]
Abstract
PURPOSE Aim of this study is to compare Quantitative Magnetic Resonance Imaging (qMRI) measures between Becker Muscular Dystrophy (BMD) and Healthy Subjects (HS) and to correlate these parameters with clinical scores. METHODS Ten BMD patients (mean age ±standard deviation: 38.7 ± 15.0 years) and ten age-matched HS, were investigated through magnetic resonance imaging (MRI) at thigh and calf levels, including: 1) a standard axial T1-weighted sequence; 2) a volumetric T2-weighted sequence; 3) a multiecho spin-echo sequence; 4) a 2-point Dixon sequence; 5) a Diffusion Tensor Imaging (DTI) sequence. RESULTS Mean Fat Fraction (FF), T2-relaxation time and Fractional Anisotropy (FA) DTI at thigh and calf levels were significantly higher in BMD patients than in HS (p-values < 0.01). FF at thigh and calf levels significantly correlated with North Star Ambulatory Assessment (NSAA) score (p-values < 0.01) and6 Minutes Walking Test (6MWT) (p-values < 0.01), whereas only calf muscle FF was significantly associated with time to get up from floor (p-value = 0.01). T2 significantly correlated with NSAA score (p-value < 0.01), 6MWT (p-value = 0.02) and time to get up from floor (p-value < 0.01) only at calf level. Among DTI values, only FA in thigh and calf muscles significantly correlated with NSAA score, 6MWT and 10-m walk (all p-values < 0.05); only FA in calf muscles significantly correlated with time to get up from floor (p = 0.01). CONCLUSIONS Muscle FF, T2-relaxometry and DTI, seem to be a promising biomarker to assess BMD disease severity, although further studies are needed to evaluate changes over the time.
Collapse
Affiliation(s)
- Lorenzo Maggi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy. .,Neurology IV-Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico "Carlo Besta", Via Celoria 11, 20133, Milan, Italy.
| | - Marco Moscatelli
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Rita Frangiamore
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Federica Mazzi
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Mattia Verri
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Alberto De Luca
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maria Barbara Pasanisi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giovanni Baranello
- Developmental Neurology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Irene Tramacere
- Department of Research and Clinical Development, Scientific Directorate, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Luisa Chiapparini
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Maria Grazia Bruzzone
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Renato Mantegazza
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Domenico Aquino
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| |
Collapse
|