1
|
Krishna S, Quindry JC, Valentine RJ, Selsby JT. The Interaction of Duchenne Muscular Dystrophy and Insulin Resistance. Exerc Sport Sci Rev 2024; 52:31-38. [PMID: 38126403 DOI: 10.1249/jes.0000000000000328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Duchenne muscular dystrophy (DMD), caused by deficiency of functional dystrophin protein, is a fatal, progressive muscle disease that frequently includes metabolic dysregulation. Herein, we explore the physiologic consequences of dystrophin deficiency within the context of obesity and insulin resistance. We hypothesized that dystrophin deficiency increases the frequency of insulin resistance, and insulin resistance potentiates muscle pathology caused by dystrophin deficiency.
Collapse
Affiliation(s)
- Swathy Krishna
- Departments of Animal Science and Kinesiology, Iowa State University, Ames, IA
| | - John C Quindry
- School of Integrative Physiology and Athletic Training, University of Montana, Missoula, MT
| | - Rudy J Valentine
- Departments of Animal Science and Kinesiology, Iowa State University, Ames, IA
| | - Joshua T Selsby
- Departments of Animal Science and Kinesiology, Iowa State University, Ames, IA
| |
Collapse
|
2
|
Nitahara-Kasahara Y, Nakayama S, Kimura K, Yamaguchi S, Kakiuchi Y, Nito C, Hayashi M, Nakaishi T, Ueda Y, Okada T. Immunomodulatory amnion-derived mesenchymal stromal cells preserve muscle function in a mouse model of Duchenne muscular dystrophy. Stem Cell Res Ther 2023; 14:108. [PMID: 37106393 PMCID: PMC10142496 DOI: 10.1186/s13287-023-03337-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is an incurable genetic disease characterized by degeneration and necrosis of myofibers, chronic inflammation, and progressive muscle weakness resulting in premature mortality. Immunosuppressive multipotent mesenchymal stromal cell (MSC) therapy could be an option for DMD patients. We focused on amnion-derived mesenchymal stromal cells (AMSCs), a clinically viable cell source owing to their unique characteristics, such as non-invasive isolation, mitotic stability, ethical acceptability, and minimal risk of immune reaction and cancer. We aimed to identify novel immunomodulatory effects of AMSCs on macrophage polarization and their transplantation strategies for the functional recovery of skeletal and cardiac muscles. METHODS We used flow cytometry to analyze the expression of anti-inflammatory M2 macrophage markers on peripheral blood mononuclear cells (PBMCs) co-cultured with human AMSCs (hAMSCs). hAMSCs were intravenously injected into DMD model mice (mdx mice) to assess the safety and efficacy of therapeutic interventions. hAMSC-treated and untreated mdx mice were monitored using blood tests, histological examinations, spontaneous wheel-running activities, grip strength, and echocardiography. RESULTS hAMSCs induced M2 macrophage polarization in PBMCs via prostaglandin E2 production. After repeated systemic hAMSC injections, mdx mice exhibited a transient downregulation of serum creatin kinase. Limited mononuclear cell infiltration and a decreased number of centrally nucleated fibers were indicative of regenerated myofibers following degeneration, suggesting an improved histological appearance of the skeletal muscle of hAMSC-treated mdx mice. Upregulated M2 macrophages and altered cytokine/chemokine expressions were observed in the muscles of hAMSC-treated mdx mice. During long-term experiments, a significant decrease in the grip strength in control mdx mice significantly improved in the hAMSC-treated mdx mice. hAMSC-treated mdx mice maintained running activity and enhanced daily running distance. Notably, the treated mice could run longer distances per minute, indicating high running endurance. Left ventricular function in DMD mice improved in hAMSC-treated mdx mice. CONCLUSIONS Early systemic hAMSC administration in mdx mice ameliorated progressive phenotypes, including pathological inflammation and motor dysfunction, resulting in the long-term improvement of skeletal and cardiac muscle function. The therapeutic effects might be associated with the immunosuppressive properties of hAMSCs via M2 macrophage polarization. This treatment strategy could provide therapeutic benefits to DMD patients.
Collapse
Affiliation(s)
- Yuko Nitahara-Kasahara
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-Ku, Tokyo, 108-8639, Japan.
| | - Soya Nakayama
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| | - Koichi Kimura
- Department of Laboratory Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sho Yamaguchi
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| | - Yuko Kakiuchi
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-Ku, Tokyo, 108-8639, Japan
| | - Chikako Nito
- Laboratory for Clinical Research, Collaborative Research Center, Nippon Medical School, Tokyo, Japan
| | - Masahiro Hayashi
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| | - Tomoyuki Nakaishi
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| | - Yasuyoshi Ueda
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| | - Takashi Okada
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-Ku, Tokyo, 108-8639, Japan.
| |
Collapse
|
3
|
Younger DS. On the path to evidence-based therapy in neuromuscular disorders. HANDBOOK OF CLINICAL NEUROLOGY 2023; 195:315-358. [PMID: 37562877 DOI: 10.1016/b978-0-323-98818-6.00007-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Neuromuscular disorders encompass a diverse group of acquired and genetic diseases characterized by loss of motor functionality. Although cure is the goal, many therapeutic strategies have been envisioned and are being studied in randomized clinical trials and entered clinical practice. As in all scientific endeavors, the successful clinical translation depends on the quality and translatability of preclinical findings and on the predictive value and feasibility of the clinical models. This chapter focuses on five exemplary diseases: childhood spinal muscular atrophy (SMA), Charcot-Marie-Tooth (CMT) disorders, chronic inflammatory demyelinating polyradiculoneuropathy (CIDP), acquired autoimmune myasthenia gravis (MG), and Duchenne muscular dystrophy (DMD), to illustrate the progress made on the path to evidenced-based therapy.
Collapse
Affiliation(s)
- David S Younger
- Department of Clinical Medicine and Neuroscience, CUNY School of Medicine, New York, NY, United States; Department of Medicine, Section of Internal Medicine and Neurology, White Plains Hospital, White Plains, NY, United States.
| |
Collapse
|
4
|
Younger DS. Childhood muscular dystrophies. HANDBOOK OF CLINICAL NEUROLOGY 2023; 195:461-496. [PMID: 37562882 DOI: 10.1016/b978-0-323-98818-6.00024-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Infancy- and childhood-onset muscular dystrophies are associated with a characteristic distribution and progression of motor dysfunction. The underlying causes of progressive childhood muscular dystrophies are heterogeneous involving diverse genetic pathways and genes that encode proteins of the plasma membrane, extracellular matrix, sarcomere, and nuclear membrane components. The prototypical clinicopathological features in an affected child may be adequate to fully distinguish it from other likely diagnoses based on four common features: (1) weakness and wasting of pelvic-femoral and scapular muscles with involvement of heart muscle; (2) elevation of serum muscle enzymes in particular serum creatine kinase; (3) necrosis and regeneration of myofibers; and (4) molecular neurogenetic assessment particularly utilizing next-generation sequencing of the genome of the likeliest candidates genes in an index case or family proband. A number of different animal models of therapeutic strategies have been developed for gene transfer therapy, but so far these techniques have not yet entered clinical practice. Treatment remains for the most part symptomatic with the goal of ameliorating locomotor and cardiorespiratory manifestations of the disease.
Collapse
Affiliation(s)
- David S Younger
- Department of Clinical Medicine and Neuroscience, CUNY School of Medicine, New York, NY, United States; Department of Medicine, Section of Internal Medicine and Neurology, White Plains Hospital, White Plains, NY, United States.
| |
Collapse
|
5
|
Abstract
Viltolarsen is a phosphorodiamidate morpholino antisense oligonucleotide (PMO) designed to skip exon 53 of the DMD gene for the treatment of Duchenne muscular dystrophy (DMD), one of the most common lethal genetic disorders characterized by progressive degeneration of skeletal muscles and cardiomyopathy. It was developed by Nippon Shinyaku in collaboration with the National Center of Neurology and Psychiatry (NCNP) in Japan based on the preclinical studies conducted in the DMD dog model at the NCNP. After showing hopeful results in pre-clinical trials and several clinical trials across North America and Japan, it received US Food and Drug Administration (FDA) approval for DMD in 2020. Viltolarsen restores the reading frame of the DMD gene by skipping exon 53 and produces a truncated but functional form of dystrophin. It can treat approximately 8-10% of the DMD patient population. This paper aims to summarize the development of viltolarsen from preclinical trials to clinical trials to, finally, FDA approval, and discusses the challenges that come with fighting DMD using antisense therapy.
Collapse
Affiliation(s)
- Rohini Roy Roshmi
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
- The Friends of Garrett Cumming Research & Muscular Dystrophy Canada, HM Toupin Neurological Science Research Chair, Edmonton, Canada.
| |
Collapse
|
6
|
Abstract
PURPOSE OF REVIEW This article reviews the history, epidemiology, genetics, clinical presentation, multidisciplinary management, and established and emerging therapies for the dystrophinopathies. RECENT FINDINGS The multidisciplinary care of individuals with dystrophinopathies continues to improve in many ways, including early surveillance and implementation of respiratory, cardiac, and orthopedic health management. The era of genetic therapeutics has altered the treatment landscape in neuromuscular disorders, including the dystrophinopathies. SUMMARY The dystrophinopathies are a spectrum of X-linked genetic disorders characterized by childhood-onset progressive weakness and variable cardiac and cognitive involvement. Corticosteroids are the mainstay of therapy to slow disease progression. Additional strategies for disease amelioration and dystrophin restoration, including gene replacement therapy, are under investigation.
Collapse
|
7
|
Ritter P, Nübler S, Buttgereit A, Smith LR, Mühlberg A, Bauer J, Michael M, Kreiß L, Haug M, Barton E, Friedrich O. Myofibrillar Lattice Remodeling Is a Structural Cytoskeletal Predictor of Diaphragm Muscle Weakness in a Fibrotic mdx ( mdx Cmah-/-) Model. Int J Mol Sci 2022; 23:ijms231810841. [PMID: 36142754 PMCID: PMC9500669 DOI: 10.3390/ijms231810841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a degenerative genetic myopathy characterized by complete absence of dystrophin. Although the mdx mouse lacks dystrophin, its phenotype is milder compared to DMD patients. The incorporation of a null mutation in the Cmah gene led to a more DMD-like phenotype (i.e., more fibrosis). Although fibrosis is thought to be the major determinant of ‘structural weakness’, intracellular remodeling of myofibrillar geometry was shown to be a major cellular determinant thereof. To dissect the respective contribution to muscle weakness, we assessed biomechanics and extra- and intracellular architecture of whole muscle and single fibers from extensor digitorum longus (EDL) and diaphragm. Despite increased collagen contents in both muscles, passive stiffness in mdx Cmah−/− diaphragm was similar to wt mice (EDL muscles were twice as stiff). Isometric twitch and tetanic stresses were 50% reduced in mdx Cmah−/− diaphragm (15% in EDL). Myofibrillar architecture was severely compromised in mdx Cmah−/− single fibers of both muscle types, but more pronounced in diaphragm. Our results show that the mdx Cmah−/− genotype reproduces DMD-like fibrosis but is not associated with changes in passive visco-elastic muscle stiffness. Furthermore, detriments in active isometric force are compatible with the pronounced myofibrillar disarray of the dystrophic background.
Collapse
Affiliation(s)
- Paul Ritter
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
- Correspondence:
| | - Stefanie Nübler
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Andreas Buttgereit
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Lucas R. Smith
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, CA 95618, USA
| | - Alexander Mühlberg
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Julian Bauer
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Mena Michael
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Lucas Kreiß
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Michael Haug
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Elisabeth Barton
- College of Health & Human Performance, University of Florida, Gainesville, FL 32611, USA
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
- School of Medical Sciences, University of New South Wales, Wallace Wurth Building, 18 High Str., Sydney, NSW 2052, Australia
| |
Collapse
|
8
|
Butterfield RJ, Krikov S, Conway KM, Johnson N, Matthews D, Phan H, Cai B, Paramsothy P, Thomas S, Feldkamp ML. Evaluation of effects of continued corticosteroid treatment on cardiac and pulmonary function in non-ambulatory males with Duchenne muscular dystrophy from MD STARnet. Muscle Nerve 2022; 66:15-23. [PMID: 34994466 PMCID: PMC9197945 DOI: 10.1002/mus.27490] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 11/10/2022]
Abstract
INTRODUCTION/AIMS Corticosteroids have been shown to improve muscle strength and delay loss of ambulation (LOA) in Duchenne muscular dystrophy (DMD) and are considered standard of care despite significant side-effects. The objective of this study is to evaluate whether corticosteroid treatment after LOA is beneficial for cardiac or pulmonary functions among boys with DMD. METHODS We used the Muscular Dystrophy Surveillance, Tracking, and Research Network (MD STARnet) to characterize associations between corticosteroid use and onset of abnormal left ventricular (LV) function or abnormal percent predicted forced vital capacity (ppFVC) among 398 non-ambulatory boys with DMD. Kaplan-Meier curve estimation was used to compare time to onset by corticosteroid use groups; Cox proportional hazards modeling was used to estimate hazard ratios (HRs) and corresponding 95% confidence intervals. RESULTS We found no differences in time to onset of abnormal LV function by corticosteroid use groups. We observed a longer time from LOA to first abnormal ppFVC in boys that were treated with corticosteroid ≥1 y beyond LOA compared with those with no corticosteroid use or those who stopped corticosteroid use within 1 y of LOA. DISCUSSION Our findings show no association of corticosteroid use beyond LOA with the onset of abnormal LV function, but a significant association with a delay in onset of abnormal ppFVC. Prospective studies of corticosteroid use in boys with DMD who have lost ambulation may identify benefits and can better elucidate risks, allowing for more effective counseling of patients on continuing treatment after LOA.
Collapse
Affiliation(s)
- Russell J Butterfield
- Department of Neurology, University of Utah, Albany, NY, USA,Department of Pediatrics, University of Utah, Albany, NY, USA
| | - Sergey Krikov
- Department of Pediatrics, University of Utah, Albany, NY, USA
| | | | - Nicholas Johnson
- Department of Neurology, Virginia Commonwealth University, Albany, NY, USA
| | | | - Han Phan
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Albany, NY, USA
| | - Bo Cai
- University of South Carolina, Albany, NY, USA
| | - Pangaja Paramsothy
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Albany, NY, USA
| | - Shiny Thomas
- New York State Department of Health, Albany, NY, USA
| | | |
Collapse
|
9
|
Guglieri M, Bushby K, McDermott MP, Hart KA, Tawil R, Martens WB, Herr BE, McColl E, Speed C, Wilkinson J, Kirschner J, King WM, Eagle M, Brown MW, Willis T, Griggs RC. Effect of Different Corticosteroid Dosing Regimens on Clinical Outcomes in Boys With Duchenne Muscular Dystrophy: A Randomized Clinical Trial. JAMA 2022; 327:1456-1468. [PMID: 35381069 PMCID: PMC8984930 DOI: 10.1001/jama.2022.4315] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
IMPORTANCE Corticosteroids improve strength and function in boys with Duchenne muscular dystrophy. However, there is uncertainty regarding the optimum regimen and dosage. OBJECTIVE To compare efficacy and adverse effects of the 3 most frequently prescribed corticosteroid regimens in boys with Duchenne muscular dystrophy. DESIGN, SETTING, AND PARTICIPANTS Double-blind, parallel-group randomized clinical trial including 196 boys aged 4 to 7 years with Duchenne muscular dystrophy who had not previously been treated with corticosteroids; enrollment occurred between January 30, 2013, and September 17, 2016, at 32 clinic sites in 5 countries. The boys were assessed for 3 years (last participant visit on October 16, 2019). INTERVENTIONS Participants were randomized to daily prednisone (0.75 mg/kg) (n = 65), daily deflazacort (0.90 mg/kg) (n = 65), or intermittent prednisone (0.75 mg/kg for 10 days on and then 10 days off) (n = 66). MAIN OUTCOMES AND MEASURES The global primary outcome comprised 3 end points: rise from the floor velocity (in rise/seconds), forced vital capacity (in liters), and participant or parent global satisfaction with treatment measured by the Treatment Satisfaction Questionnaire for Medication (TSQM; score range, 0 to 100), each averaged across all study visits after baseline. Pairwise group comparisons used a Bonferroni-adjusted significance level of .017. RESULTS Among the 196 boys randomized (mean age, 5.8 years [SD, 1.0 years]), 164 (84%) completed the trial. Both daily prednisone and daily deflazacort were more effective than intermittent prednisone for the primary outcome (P < .001 for daily prednisone vs intermittent prednisone using a global test; P = .017 for daily deflazacort vs intermittent prednisone using a global test) and the daily regimens did not differ significantly (P = .38 for daily prednisone vs daily deflazacort using a global test). The between-group differences were principally attributable to rise from the floor velocity (0.06 rise/s [98.3% CI, 0.03 to 0.08 rise/s] for daily prednisone vs intermittent prednisone [P = .003]; 0.06 rise/s [98.3% CI, 0.03 to 0.09 rise/s] for daily deflazacort vs intermittent prednisone [P = .017]; and -0.004 rise/s [98.3% CI, -0.03 to 0.02 rise/s] for daily prednisone vs daily deflazacort [P = .75]). The pairwise comparisons for forced vital capacity and TSQM global satisfaction subscale score were not statistically significant. The most common adverse events were abnormal behavior (22 [34%] in the daily prednisone group, 25 [38%] in the daily deflazacort group, and 24 [36%] in the intermittent prednisone group), upper respiratory tract infection (24 [37%], 19 [29%], and 24 [36%], respectively), and vomiting (19 [29%], 17 [26%], and 15 [23%]). CONCLUSIONS AND RELEVANCE Among patients with Duchenne muscular dystrophy, treatment with daily prednisone or daily deflazacort, compared with intermittent prednisone alternating 10 days on and 10 days off, resulted in significant improvement over 3 years in a composite outcome comprising measures of motor function, pulmonary function, and satisfaction with treatment; there was no significant difference between the 2 daily corticosteroid regimens. The findings support the use of a daily corticosteroid regimen over the intermittent prednisone regimen tested in this study as initial treatment for boys with Duchenne muscular dystrophy. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01603407.
Collapse
Affiliation(s)
- Michela Guglieri
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, England
| | - Kate Bushby
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, England
| | - Michael P. McDermott
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York
| | - Kimberly A. Hart
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
| | - Rabi Tawil
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
| | - William B. Martens
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
| | - Barbara E. Herr
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
| | | | - Chris Speed
- Newcastle University, Newcastle upon Tyne, England
- NIHR Clinical Research Network North East and North Cumbria, Newcastle upon Tyne, England
| | | | - Janbernd Kirschner
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, University Hospital Freiburg, Freiburg, Germany
- Department of Neuropediatrics, University Hospital Bonn, Bonn, Germany
| | | | - Michelle Eagle
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, England
| | - Mary W. Brown
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
| | - Tracey Willis
- Robert Jones and Agnes Hunt Orthopaedic Hospital, NHS Foundation Trust, Oswestry, England
| | - Robert C. Griggs
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
| | | |
Collapse
|
10
|
Conway KM, Gedlinske A, Mathews KD, Perlman S, Johnson N, Butterfield R, Hung M, Bounsanga J, Matthews D, Oleszek J, Romitti PA. A population-based study of scoliosis among males diagnosed with a dystrophinopathy identified by the Muscular Dystrophy Surveillance, Tracking, and Research Network (MD STARnet). Muscle Nerve 2022; 65:193-202. [PMID: 34787322 PMCID: PMC8752499 DOI: 10.1002/mus.27464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 02/03/2023]
Abstract
INTRODUCTION/AIMS Scoliosis is a common comorbidity among individuals diagnosed with a dystrophinopathy. We examined associations between clinical predictors and scoliosis in childhood-onset dystrophinopathy. METHODS The progression and treatment of scoliosis were obtained from data collected by the US population-based Muscular Dystrophy Surveillance, Tracking, and Research Network. Associations between loss of independent ambulation (LoA) and corticosteroid use and scoliosis outcomes (ages at or exceeding Cobb angle thresholds [10°, 20°, 30°]; surgery) were estimated using Kaplan-Meier curve estimation and extended Cox regression modeling. RESULTS We analyzed curvature data for 513 of 1054 individuals ascertained. Overall, approximately one-half had at least one radiograph and one-quarter had a curvature of at least 20°. The average maximum curvature was 25.0° (SD = 21.5°) among all individuals and 42.8° (SD = 18.8°) among those recommended for surgery. Higher adjusted hazards ratio of curvature (aHR(curvature) [95% confidence interval]) were found among individuals with LoA compared to those without LoA (aHR(10) = 6.2 [4.4, 8.7], aHR(20) = 15.3 [7.4, 31.7], aHR(30) = 31.6 [7.7, 128.9]), among individuals who did not use corticosteroids compared to those who did (aHR(10) = 1.2 [0.9, 1.7], aHR(20) = 1.8 [1.1, 2.7], aHR(30) = 2.3 [1.3, 4.0]), and among non-ambulatory individuals who used corticosteroids after LoA compared to those who did not (aHR(10) = 1.8 [1.2, 2.8], aHR(20) = 1.6 [1.0, 2.6], aHR(30) = 3.6 [1.6, 7.9]). Scoliosis surgery among individuals with LoA who did not use corticosteroids was more than double compared to those who used (aHR = 2.3 [1.3, 4.2]). DISCUSSION Our retrospective observational study suggests corticosteroids may delay spinal curvature progression and need for scoliosis surgery. Continuing corticosteroids after LoA also showed potential benefits of delaying curvature progression, additional studies are needed to confirm this finding or address the magnitude of benefit.
Collapse
Affiliation(s)
- Kristin M Conway
- Department of Epidemiology, The University of Iowa, Iowa City, Iowa, USA
| | - Amber Gedlinske
- Department of Internal Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Katherine D Mathews
- Departments of Pediatrics and Neurology, The University of Iowa, Iowa City, Iowa, USA
| | - Seth Perlman
- Department of Neurology, Seattle Children's Hospital, Seattle, Washington, USA
| | - Nicholas Johnson
- Department of Neurology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Russell Butterfield
- Departments of Pediatrics and Neurology, University of Utah, Salt Lake City, Utah, USA
| | - Man Hung
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, Utah, USA
| | | | - Dennis Matthews
- Department of Physical Medicine and Rehabilitation, University of Colorado and Children's Hospital, Denver, Colorado, USA
| | - Joyce Oleszek
- Department of Physical Medicine and Rehabilitation, University of Colorado and Children's Hospital, Denver, Colorado, USA
| | - Paul A Romitti
- Department of Epidemiology, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
11
|
Rocha CT, Escolar DM. Treatment and Management of Muscular Dystrophies. Neuromuscul Disord 2022. [DOI: 10.1016/b978-0-323-71317-7.00020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
12
|
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
|
13
|
Galati A, Brown ES, Bove R, Vaidya A, Gelfand J. Glucocorticoids for therapeutic immunosuppression: Clinical pearls for the practicing neurologist. J Neurol Sci 2021; 430:120004. [PMID: 34598056 DOI: 10.1016/j.jns.2021.120004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
Given widespread use of glucocorticoid therapy in neurologic disease, understanding glucocorticoid pharmacology and risk is paramount for the practicing neurologist. While dosing and tapering regimens vary depending on the neurological disease and indication being treated, there are important general principles of glucocorticoid prescribing and monitoring that can guide clinical decision-making. Glucocorticoid-related toxicities can occur across multiple organ systems, including hypertension; dyslipidemia; weight gain; hyperglycemia; osteoporosis and avascular necrosis; myopathy; gastrointestinal bleeding; infection; and neuropsychiatric effects with sleep, mood disturbance and cognition. This narrative review provides a practical framework for safe and responsible prescribing of this therapeutic class of medications, including appreciation of immunosuppressive consequences, risk mitigation strategies, dosing and tapering, and recognition of adrenal insufficiency and glucocorticoid withdrawal.
Collapse
Affiliation(s)
- Alexandra Galati
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, USA.
| | - E Sherwood Brown
- Department of Psychiatry, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Riley Bove
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, USA
| | - Anand Vaidya
- Center for Adrenal Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jeffrey Gelfand
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, USA.
| |
Collapse
|
14
|
The Immune System in Duchenne Muscular Dystrophy Pathogenesis. Biomedicines 2021; 9:biomedicines9101447. [PMID: 34680564 PMCID: PMC8533196 DOI: 10.3390/biomedicines9101447] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/28/2021] [Accepted: 10/04/2021] [Indexed: 12/24/2022] Open
Abstract
Growing evidence demonstrates the crosstalk between the immune system and the skeletal muscle in inflammatory muscle diseases and dystrophic conditions such as Duchenne Muscular Dystrophy (DMD), as well as during normal muscle regeneration. The rising of inflammation and the consequent activation of the immune system are hallmarks of DMD: several efforts identified the immune cells that invade skeletal muscle as CD4+ and CD8+ T cells, Tregs, macrophages, eosinophils and natural killer T cells. The severity of muscle injury and inflammation dictates the impairment of muscle regeneration and the successive replacement of myofibers with connective and adipose tissue. Since immune system activation was traditionally considered as a consequence of muscular wasting, we recently demonstrated a defect in central tolerance caused by thymus alteration and the presence of autoreactive T-lymphocytes in DMD. Although the study of innate and adaptive immune responses and their complex relationship in DMD attracted the interest of many researchers in the last years, the results are so far barely exhaustive and sometimes contradictory. In this review, we describe the most recent improvements in the knowledge of immune system involvement in DMD pathogenesis, leading to new opportunities from a clinical point-of-view.
Collapse
|
15
|
Kochar GS, Sondhi V, Kabra SK, Yadav SL, Dwivedi SN, Gulati S. Intermittent versus daily regimen of prednisolone in ambulatory boys with Duchenne muscular dystrophy: A randomized, open-label trial. Muscle Nerve 2021; 65:60-66. [PMID: 34617309 DOI: 10.1002/mus.27428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 12/13/2022]
Abstract
INTRODUCTION/AIMS Corticosteroids prolong ambulation and improve muscle power among boys with Duchenne muscular dystrophy (DMD). However, the optimal steroid regimen remains unclear. Hence, this study was undertaken to compare the efficacy of daily- versus intermittent-steroid regimens in ambulatory boys with DMD. METHODS In this single-center, open-label randomized trial, 72 children were randomized to receive either daily prednisolone (0.75 mg/kg/day) or intermittent prednisolone (0.75 mg/kg/day, for first 10 days of every month). The primary outcome measure was the difference in average score on manual muscle testing (MMT) at baseline and after 6 mo of steroids. A difference of >0.2 was hypothesized to be significant. Secondary outcomes included changes in timed functions, muscular dystrophy-specific functional-rating scale score, peak torque, average power, and pulmonary function. RESULTS In the intention-to-treat analysis, the mean (SD) change in MMT scores was 0.17 (0.15) and 0.08 (0.10) for the daily and intermittent steroid groups, respectively. The mean difference between the two interventions was 0.10 (95% confidence interval [CI] = 0.04-0.16; P = .003), which although significant was less than the predefined value of 0.2. Statistically significantly improvements were observed with daily-steroid regimen in the Gowers time (P = .01), nine-metre walk test (P = .02) and average power (P = .02) as compared to intermittent-steroid regimen. A total of 19/32 (52.8%) children in the daily-steroid group and 8/29 (27%) children in the intermittent-steroid group experienced some form of adverse effect (P = .02). DISCUSSION Over a short-term period, the intermittent-steroid regimen was non-inferior to the daily-steroid regime in preserving muscle strength among children with DMD. However, better improvement of functional measures was observed with daily-steroid administration. The frequency of individual side effects was similar between the two groups.
Collapse
Affiliation(s)
| | - Vishal Sondhi
- Department of Pediatrics, Armed Forces Medical College, Pune, India
| | - Sushil K Kabra
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Shiv L Yadav
- Department of Physical Medicine and Rehabilitation, All India Institute of Medical Sciences, New Delhi, India
| | - Sada N Dwivedi
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi, India
| | - Sheffali Gulati
- Center of Excellence & Advanced Research on Childhood Neurodevelopmental Disorders, Child Neurology Division, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
16
|
Gallagher GW, Nowacek D, Gutgsell O, Callaghan BC. Comparison of the United Kingdom and United States approaches to approval of new neuromuscular therapies. Muscle Nerve 2021; 64:641-650. [PMID: 34448221 DOI: 10.1002/mus.27380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/14/2021] [Accepted: 07/18/2021] [Indexed: 01/08/2023]
Abstract
Many novel therapies are now available for rare neuromuscular conditions that were previously untreatable. Hereditary transthyretin amyloidosis and spinal muscular atrophy are two examples of diseases with new medications that have transformed our field. The United States and the United Kingdom have taken disparate approaches to the approval and coverage of medications, despite both providing incentives to develop therapies targeting rare diseases. The US requires less evidence for approval when compared with medications for common diseases and does not have a mechanism to ensure or even encourage cost-effectiveness. The Institute of Clinical and Economic Review provides in-depth cost-effectiveness analyses in the US, but does not have the authority to negotiate drug costs. In contrast, the UK has maintained a similar scientific threshold for approval of all therapies, while requiring negotiation with National Institute for Health and Care Excellence to ensure that medications are cost-effective for rare diseases. These differences have led to approval of medications for rare diseases in the US that have less evidence than required for common diseases. Importantly, these medications have not been approved in the UK. Even when medications meet traditional scientific thresholds, they uniformly arrive with high list prices in the US, whereas they are available at cost-effective prices in the UK. The main downsides to the UK approach are that cost-effective medications are often available months later than in the US, and some medications remain unavailable.
Collapse
|
17
|
Brennan CM, Emerson CP, Owens J, Christoforou N. p38 MAPKs - roles in skeletal muscle physiology, disease mechanisms, and as potential therapeutic targets. JCI Insight 2021; 6:e149915. [PMID: 34156029 PMCID: PMC8262482 DOI: 10.1172/jci.insight.149915] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
p38 MAPKs play a central role in orchestrating the cellular response to stress and inflammation and in the regulation of myogenesis. Potent inhibitors of p38 MAPKs have been pursued as potential therapies for several disease indications due to their antiinflammatory properties, although none have been approved to date. Here, we provide a brief overview of p38 MAPKs, including their role in regulating myogenesis and their association with disease progression. Finally, we discuss targeting p38 MAPKs as a therapeutic approach for treating facioscapulohumeral muscular dystrophy and other muscular dystrophies by addressing multiple pathological mechanisms in skeletal muscle.
Collapse
Affiliation(s)
| | - Charles P Emerson
- Wellstone Muscular Dystrophy Program, Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jane Owens
- Rare Disease Research Unit, Pfizer Inc., Cambridge, Massachusetts, USA
| | | |
Collapse
|
18
|
Riviello JJ, Erklauer J. Evidence-Based Protocols in Child Neurology. Neurol Clin 2021; 39:883-895. [PMID: 34215392 DOI: 10.1016/j.ncl.2021.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Medical care has become more complex as the scientific method has expanded medical knowledge. Medicine is also now practiced across different medical systems of varying complexity, and creating standard treatment guidelines is one way of establishing uniform treatment across these systems. The creation of guidelines ensures the delivery of quality medical care and improved patient outcomes. Evidence-based medicine is the application of scientific research to produce these treatment guidelines. This article shall focus on the current treatment guidelines used for inpatient pediatric neurology.
Collapse
Affiliation(s)
- James J Riviello
- Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, 6701 Fannin Street, Suite 1250, Houston, TX 77030, USA.
| | - Jennifer Erklauer
- Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, 6701 Fannin Street, Suite 1250, Houston, TX 77030, USA; Section of Pediatric Critical Care Medicine, Baylor College of Medicine, Texas Children's Hospital, 6701 Fannin Street, Suite 1250, Houston, TX 77030, USA
| |
Collapse
|
19
|
Nutraceutical Screening in a Zebrafish Model of Muscular Dystrophy: Gingerol as a Possible Food Aid. Nutrients 2021; 13:nu13030998. [PMID: 33808773 PMCID: PMC8003371 DOI: 10.3390/nu13030998] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/04/2021] [Accepted: 03/16/2021] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), caused by mutations in the dystrophin gene, is an inherited neuromuscular disorder that causes loss of muscle mass and motor skills. In the era of genomic medicine, there is still no known cure for DMD. In clinical practice, there is a growing awareness of the possible importance of nutrition in neuromuscular diseases. This is mostly the result of patients’ or caregivers’ empirical reports of how active substances derived from food have led to improved muscle strength and, thus, better quality of life. In this report, we investigate several nutraceutical principles in the sapje strain of zebrafish, a validated model of DMD, in order to identify possible natural products that, if supplemented in the diet, might improve the quality of life of DMD patients. Gingerol, a constituent of fresh ginger, statistically increased the locomotion of mutant larvae and upregulated the expression of heme oxygenase 1, a target gene for therapy aimed at improving dystrophic symptoms. Although three other compounds showed a partial positive effect on locomotor and muscle structure phenotypes, our nutraceutical screening study lent preliminary support to the efficacy and safety only of gingerol. Gingerol could easily be proposed as a dietary supplement in DMD.
Collapse
|
20
|
Nitahara-Kasahara Y, Kuraoka M, Guillermo PH, Hayashita-Kinoh H, Maruoka Y, Nakamura-Takahasi A, Kimura K, Takeda S, Okada T. Dental pulp stem cells can improve muscle dysfunction in animal models of Duchenne muscular dystrophy. Stem Cell Res Ther 2021; 12:78. [PMID: 33494794 PMCID: PMC7831244 DOI: 10.1186/s13287-020-02099-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/13/2020] [Indexed: 12/11/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is an inherited progressive disorder that causes skeletal and cardiac muscle deterioration with chronic inflammation. Dental pulp stem cells (DPSCs) are attractive candidates for cell-based strategies for DMD because of their immunosuppressive properties. Therefore, we hypothesized that systemic treatment with DPSCs might show therapeutic benefits as an anti-inflammatory therapy. Methods To investigate the potential benefits of DPSC transplantation for DMD, we examined disease progression in a DMD animal model, mdx mice, by comparing them with different systemic treatment conditions. The DPSC-treated model, a canine X-linked muscular dystrophy model in Japan (CXMDJ), which has a severe phenotype similar to that of DMD patients, also underwent comprehensive analysis, including histopathological findings, muscle function, and locomotor activity. Results We demonstrated a therapeutic strategy for long-term functional recovery in DMD using repeated DPSC administration. DPSC-treated mdx mice and CXMDJ showed no serious adverse events. MRI findings and muscle histology suggested that DPSC treatment downregulated severe inflammation in DMD muscles and demonstrated a milder phenotype after DPSC treatment. DPSC-treated models showed increased recovery in grip-hand strength and improved tetanic force and home cage activity. Interestingly, maintenance of long-term running capability and stabilized cardiac function was also observed in 1-year-old DPSC-treated CXMDJ. Conclusions We developed a novel strategy for the safe and effective transplantation of DPSCs for DMD recovery, which included repeated systemic injection to regulate inflammation at a young age. This is the first report on the efficacy of a systemic DPSC treatment, from which we can propose that DPSCs may play an important role in delaying the DMD disease phenotype.
Collapse
Affiliation(s)
- Yuko Nitahara-Kasahara
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan. .,Division of Cell and Gene Therapy, Nippon Medical School, Bunkyo-city, Tokyo, Japan. .,Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.
| | - Mutsuki Kuraoka
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Laboratory of Experimental Animal Science, Nippon Veterinary and Life Science University, Musashino, Tokyo, Japan
| | - Posadas Herrera Guillermo
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan.,Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, Institute of Medical Science, The University of Tokyo, Minato-city, Tokyo, Japan
| | - Hiromi Hayashita-Kinoh
- Division of Cell and Gene Therapy, Nippon Medical School, Bunkyo-city, Tokyo, Japan.,Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, Institute of Medical Science, The University of Tokyo, Minato-city, Tokyo, Japan
| | - Yasunobu Maruoka
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | | | - Koichi Kimura
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Department of General Medicine, The Institute of Medical Science, The University of Tokyo, Minato-city, Tokyo, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Takashi Okada
- Division of Cell and Gene Therapy, Nippon Medical School, Bunkyo-city, Tokyo, Japan. .,Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan. .,Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, Institute of Medical Science, The University of Tokyo, Minato-city, Tokyo, Japan.
| |
Collapse
|
21
|
LoMauro A, Gandossini S, Russo A, Velardo D, Comi GP, Turconi AC, Bresolin N, Aliverti A, D'Angelo MG. A Multidisciplinary Evaluation of Patients with DMD in An Italian Tertiary Care Center. J Neuromuscul Dis 2020; 8:235-249. [PMID: 33361606 DOI: 10.3233/jnd-190417] [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
With more widespread prolonged survival, Duchenne muscular dystrophy patients progressively experience multisystem complications. We retrospectively reviewed the charts of 132 Duchenne patients (112 alive/20 dead, age 3.5÷32.3 years) with the aims: 1) to provide a comprehensive description of the clinical status considering different aspects of the disease; 2) to propose a new scoring tool able to consider and pool together heterogeneous different functional. Five functions were analyzed: cardiac, respiratory, nutritional, ambulation and scoliosis. For each function, different items were considered and classified according to clinical severity (as indicated by international guidelines) and an incremental scoring was assigned. In addition, a global score incorporating all functions was defined. The scoring system confirmed that despite the significant protective role of steroids, all functions deteriorated with age. The severity of the global score became significantly higher since the age of 13 years. The severity of cardiac, respiratory and nutritional dysfunction was higher since 18 years. Deceased patients were characterized by significantly worse cardiac function, absence of steroid therapy and later use of respiratory assistive devices. The index proposed in this pilot study is a promising tool able to aggregate and correlate heterogeneous functions. It could become either an individual prognostic indicator of decline or a global score to evaluate changes in clinical trials therefore allowing multicenter studies, optimizing the management of both the primary and the secondary complications of the disease and understanding their relative impact.
Collapse
Affiliation(s)
- A LoMauro
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano, Italy
| | - S Gandossini
- Scientific Institute IRCCS E. Medea, Neuromuscular Unit, Bosisio Parini (Lecco) Italy
| | - A Russo
- Scientific Institute IRCCS E. Medea, Neuromuscular Unit, Bosisio Parini (Lecco) Italy
| | - D Velardo
- Scientific Institute IRCCS E. Medea, Neuromuscular Unit, Bosisio Parini (Lecco) Italy
| | - G P Comi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan
| | - A C Turconi
- Scientific Institute, IRCCS E. Medea- via don Luigi Monza, Bosisio P (Lecco) Italy
| | - N Bresolin
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan
| | - A Aliverti
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano, Italy
| | - M G D'Angelo
- Scientific Institute IRCCS E. Medea, Neuromuscular Unit, Bosisio Parini (Lecco) Italy
| |
Collapse
|
22
|
Flotats-Bastardas M, Hahn A. New Therapeutics Options for Pediatric Neuromuscular Disorders. Front Pediatr 2020; 8:583877. [PMID: 33330280 PMCID: PMC7719776 DOI: 10.3389/fped.2020.583877] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/22/2020] [Indexed: 12/14/2022] Open
Abstract
Neuromuscular disorders (NMDs) of Childhood onset are a genetically heterogeneous group of diseases affecting the anterior horn cell, the peripheral nerve, the neuromuscular junction, or the muscle. For many decades, treatment of NMDs has been exclusively symptomatic. But this has changed fundamentally in recent years due to the development of new drugs attempting either to ameliorate secondary pathophysiologic consequences or to modify the underlying genetic defect itself. While the effects on the course of disease are still modest in some NMDs (e.g., Duchenne muscular dystrophy), new therapies have substantially prolonged life expectancy and improved motor function in others (e.g., spinal muscular atrophy and infantile onset Pompe disease). This review summarizes recently approved medicaments and provides an outlook for new therapies that are on the horizon in this field.
Collapse
Affiliation(s)
| | - Andreas Hahn
- Department of Child Neurology, University of Giessen, Giessen, Germany
| |
Collapse
|
23
|
Takeuchi F, Nakamura H, Yonemoto N, Komaki H, Rosales RL, Kornberg AJ, Bretag AH, Dejthevaporn C, Goh KJ, Jong YJ, Kim DS, Khadilkar SV, Shen D, Wong KT, Chai J, Chan SHS, Khan S, Ohnmar O, Nishino I, Takeda S, Nonaka I. Clinical practice with steroid therapy for Duchenne muscular dystrophy: An expert survey in Asia and Oceania. Brain Dev 2020; 42:277-288. [PMID: 31980267 DOI: 10.1016/j.braindev.2019.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 01/12/2023]
Abstract
BACKGROUND Several studies on clinical practice for Duchenne muscular dystrophy (DMD) have been conducted in Western countries. However, there have been only a few similar studies in Asia and Oceania. Here, we investigate the steroid therapy-related clinical practice for DMD among the local experts. In 2015, we conducted a DMD expert survey in Asia and Oceania to acquire information regarding patients with DMD and to assess current clinical practice with the cooperation of Asian and Oceanian Myology Centre, a neuromuscular disease research network. RESULTS We obtained survey responses from 87 out of 148 clinicians (62%) from 13 countries and regions. In China, 1385 DMD patients were followed-up by 5 respondent neurologists, and 84% were between 0 and 9 years of age (15% were 10-19 years, 1% > 19 years). While in Japan, 1032 patients were followed-up by 20 clinicians, and the age distribution was similar between the 3 groups (27% were 0-9 years, 35% were 10-19 years, 38% were >19 years). Most respondent clinicians (91%) were aware of DMD standard of care recommendations. Daily prednisolone/prednisone administration was used most frequently at initiation (N = 45, 64%). Inconsistent opinion on steroid therapy after loss of ambulation and medication for bone protection was observed. CONCLUSIONS Rare disease research infrastructures have been underdeveloped in many of Asian and Oceanian countries. In this situation, our results show the snapshots of current medical situation and clinical practice in DMD. For further epidemiological studies, expansion of DMD registries is necessary.
Collapse
Affiliation(s)
- Fumi Takeuchi
- Department of Clinical Research Support, Translational Medical Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8551, Japan
| | - Harumasa Nakamura
- Department of Clinical Research Support, Translational Medical Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8551, Japan.
| | - Naohiro Yonemoto
- Department of Biostatistics, Kyoto University School of Public Health, Yoshidakonoe, Sakyo, Kyoto, Kyoto 606-8501, Japan
| | - Hirofumi Komaki
- Department of Clinical Research Support, Translational Medical Center, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8551, Japan; Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8551, Japan
| | - Raymond L Rosales
- The Department of Neurology & Psychiatry and The NeuroScience Institute, University of Santo Tomas Hospital, España Blvd, Sampaloc, Manila, 1008 Metro Manila, Philippines
| | - Andrew J Kornberg
- Children's Neuroscience Centre, The Royal Children's Hospital Melbourne, 50 Flemington Road, Parkville, Victoria 3052, Melbourne, Australia
| | - Allan H Bretag
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Charungthai Dejthevaporn
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Khean Jin Goh
- Division of Neurology, Department of Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yuh-Jyh Jong
- Departments of Pediatrics and Laboratory Medicine, Kaohsiung Medical University Hospital/Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Taiwan 100, Shih-Chuan 1st Rd, Kaohsiung 80708, Taiwan; College of Biological Science and Technology, National Chiao Tung University, 75 Po-Ai St, Hsinchu 30068, Taiwan
| | - Dae-Seong Kim
- Department of Neurology, Pusan National University Yangsan Hospital, 20 Kumo-ro, Yangsan, Gyeongnam, Republic of Korea
| | - Satish V Khadilkar
- Department of Neurology, Bombay Hospital Institute of Medical Sciences, 12, Marine Lines, Mumbai, Maharashtra 40020, India
| | - Dingguo Shen
- Fudan University, 220 Handan Rd, WuJiaoChang, Yangpu District, Shanghai 200433, China
| | - Kum Thong Wong
- Department of Pathology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Josiah Chai
- Department of Neurology, National Neuroscience Institute, 11 Jalan Tan Tock Seng, Singapore 308433, Singapore
| | - Sophelia Hoi-Shan Chan
- Division of Paediatric Neurology, Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Sara Khan
- Department of Neurology, Aga Khan University Hospital, Stadium Rd, Karachi, Karachi City, Sindh 74800, Pakistan
| | - Ohnmar Ohnmar
- Department of Medical Neurology, University of Medicine 1, No. 245, Myoma Kyaung Street, Lanmadaw Township, Yangon, Myanmar
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neurosciences, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8551, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8551, Japan
| | - Ikuya Nonaka
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8551, Japan; Department of Neuromuscular Research, National Institute of Neurosciences, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8551, Japan
| |
Collapse
|
24
|
Consensus on the diagnosis, treatment and follow-up of patients with Duchenne muscular dystrophy. NEUROLOGÍA (ENGLISH EDITION) 2019. [DOI: 10.1016/j.nrleng.2018.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
25
|
McMillan HJ. Intermittent glucocorticoid regimes for younger boys with duchenne muscular dystrophy: Balancing efficacy with side effects. Muscle Nerve 2019; 59:638-639. [PMID: 30993732 DOI: 10.1002/mus.26490] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/15/2019] [Accepted: 04/16/2019] [Indexed: 11/08/2022]
Affiliation(s)
- Hugh J McMillan
- Division of Neurology, Children's Hospital of Eastern Ontario, University of Ottawa, 401 Smyth Road, Ottawa, Ontario K1H 8L1, Canada
| |
Collapse
|
26
|
Yin L, Xie ZY, Xu HY, Zheng SS, Wang ZX, Xiao JX, Yuan Y. T2 Mapping and Fat Quantification of Thigh Muscles in Children with Duchenne Muscular Dystrophy. Curr Med Sci 2019; 39:138-145. [PMID: 30868504 DOI: 10.1007/s11596-019-2012-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/12/2018] [Indexed: 11/24/2022]
Abstract
Quantitative magnetic resonance image (MRI) in individual muscles may be useful for monitoring disease progression in Duchenne muscular dystrophy (DMD). The purpose of this study was to measure T2 relaxation time of thigh muscles in children with DMD and healthy boys, and to correlate the T2 relaxation time of muscles with the fat fraction (FF) at quantitative magnetic resonance and results of clinical assessment. Thirty-two boys with DMD and 18 healthy boys were evaluated with T2 mapping and three-point Dixon MRI. Age, body mass index (BMI), muscle strength assessment, timed functional tests (time to walk or run 10 metres, rise from the floor and ascend four stairs), and the North Star Ambulatory Assessment (NSAA) were evaluated. Spearman's correlation was used to assess the relationships between FF and clinical assessments and T2 relaxation time. The mean T2 relaxation time of thigh muscles in DMD was significantly longer than that in the control group (P<0.05), except for the gracilis (P=0.952). The gracilis, sartorius and adductor longus were relatively spared by fatty infiltration in DMD patients. The T2 relaxation time was correlated significantly with the mean FF in all muscles. Age, BMI, total muscle strength score, timed functional tests and NSAA were significantly correlated with the overall mean T2 relaxation time. T2 mapping may prove clinically useful in monitoring muscle changes as a result of the disease process and in predicting the outcome of DMD patients.
Collapse
Affiliation(s)
- Liang Yin
- Department of Radiology, Peking University First Hospital, Beijing, 100034, China
| | - Zhi-Ying Xie
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China
| | - Hai-Yan Xu
- Department of Radiology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Sui-Sheng Zheng
- Department of Radiology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhao-Xia Wang
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China
| | - Jiang-Xi Xiao
- Department of Radiology, Peking University First Hospital, Beijing, 100034, China.
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
27
|
Conway KM, Eichinger K, Trout C, Romitti PA, Mathews KD, Pandya SK. Needs management in families affected by childhood-onset dystrophinopathies. SAGE Open Med 2019; 7:2050312119834470. [PMID: 30854202 PMCID: PMC6399767 DOI: 10.1177/2050312119834470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/05/2019] [Indexed: 12/19/2022] Open
Abstract
Purpose To collect information about the needs of families affected by childhood-onset dystrophinopathies residing in the United States. Methods Individuals with an eligible dystrophinopathy were identified by the Muscular Dystrophy Surveillance, Tracking, and Research network. Between September 2008 and December 2012, 272 caregivers completed a 48-item survey about needs related to information, healthcare services, psychosocial issues, finances, caregiver demographics, and the individual's functioning. Results Overall, at least 80% of the survey items were identified as needs for more than one-half of caregivers. Among the needs identified, physical health and access to information were currently managed for most caregivers. Items identified as needed but managed less consistently were funding for needs not covered by insurance and psychosocial support. Conclusions Healthcare providers, public health practitioners, and policymakers should be aware of the many needs reported by caregivers, and focus on addressing gaps in provision of needed financial and psychosocial services.
Collapse
Affiliation(s)
- Kristin M Conway
- Department of Epidemiology, The University of Iowa, Iowa City, IA, USA
| | - Katy Eichinger
- Department of Neurology, University of Rochester, Rochester, NY, USA
| | - Christina Trout
- Department of Pediatrics, The University of Iowa Stead Family Children's Hospital, Iowa City, IA, USA
| | - Paul A Romitti
- Department of Epidemiology, The University of Iowa, Iowa City, IA, USA
| | - Katherine D Mathews
- Department of Neurology, School of Medicine, The University of Iowa, Iowa City, IA, USA
| | - Shree K Pandya
- Department of Neurology, University of Rochester, Rochester, NY, USA
| | | |
Collapse
|
28
|
Connolly AM, Zaidman CM, Golumbek PT, Cradock MM, Flanigan KM, Kuntz NL, Finkel RS, McDonald CM, Iannaccone ST, Anand P, Siener CA, Florence JM, Lowes LP, Alfano LN, Johnson LB, Nicorici A, Nelson LL, Mendell JR. Twice‐weekly glucocorticosteroids in infants and young boys with Duchenne muscular dystrophy. Muscle Nerve 2019; 59:650-657. [DOI: 10.1002/mus.26441] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Anne M. Connolly
- Department of NeurologyWashington University School of Medicine in Saint Louis St Louis Missouri USA 63110
- Department of PediatricsWashington University School of Medicine in Saint Louis St Louis Missouri USA
| | - Craig M. Zaidman
- Department of NeurologyWashington University School of Medicine in Saint Louis St Louis Missouri USA 63110
- Department of PediatricsWashington University School of Medicine in Saint Louis St Louis Missouri USA
| | - Paul T. Golumbek
- Department of NeurologyWashington University School of Medicine in Saint Louis St Louis Missouri USA 63110
- Department of PediatricsWashington University School of Medicine in Saint Louis St Louis Missouri USA
| | - Mary M. Cradock
- Department of PediatricsWashington University School of Medicine in Saint Louis St Louis Missouri USA
| | - Kevin M. Flanigan
- Department of Pediatrics, Nationwide Children's HospitalOhio State University Columbus Ohio USA
| | - Nancy L. Kuntz
- Department of NeurologyNorthwestern University Feinberg School of Medicine Chicago Illinois USA
| | - Richard S. Finkel
- Department of PediatricsNemours Children's Hospital Orlando Florida USA
| | - Craig M. McDonald
- Department of Physical Medicine and RehabilitationUniversity of California, Davis Medical Center Sacramento California USA
| | - Susan T. Iannaccone
- Department of PediatricsUniversity of Texas Southwestern Medical Center Dallas Texas USA
| | - Pallavi Anand
- Department of NeurologyWashington University School of Medicine in Saint Louis St Louis Missouri USA 63110
| | - Catherine A. Siener
- Department of NeurologyWashington University School of Medicine in Saint Louis St Louis Missouri USA 63110
| | - Julaine M. Florence
- Department of NeurologyWashington University School of Medicine in Saint Louis St Louis Missouri USA 63110
| | - Linda P. Lowes
- Department of Pediatrics, Nationwide Children's HospitalOhio State University Columbus Ohio USA
| | - Lindsay N. Alfano
- Department of Pediatrics, Nationwide Children's HospitalOhio State University Columbus Ohio USA
| | - Linda B. Johnson
- Department of Physical Medicine and RehabilitationUniversity of California, Davis Medical Center Sacramento California USA
| | - Alina Nicorici
- Department of Physical Medicine and RehabilitationUniversity of California, Davis Medical Center Sacramento California USA
| | - Leslie L. Nelson
- Department of PediatricsUniversity of Texas Southwestern Medical Center Dallas Texas USA
| | - Jerry R. Mendell
- Department of Pediatrics, Nationwide Children's HospitalOhio State University Columbus Ohio USA
| | | |
Collapse
|
29
|
Houweling PJ, Papadimitriou ID, Seto JT, Pérez LM, Coso JD, North KN, Lucia A, Eynon N. Is evolutionary loss our gain? The role of
ACTN3
p.Arg577Ter (R577X) genotype in athletic performance, ageing, and disease. Hum Mutat 2018; 39:1774-1787. [DOI: 10.1002/humu.23663] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 09/20/2018] [Accepted: 09/26/2018] [Indexed: 01/13/2023]
Affiliation(s)
- Peter J. Houweling
- Murdoch Children's Research Institute Melbourne, Victoria Australia
- Department of Paediatrics University of Melbourne The Royal Children's Hospital Melbourne, Victoria Australia
| | | | - Jane T. Seto
- Murdoch Children's Research Institute Melbourne, Victoria Australia
- Department of Paediatrics University of Melbourne The Royal Children's Hospital Melbourne, Victoria Australia
| | - Laura M. Pérez
- Universidad Europea de Madrid (Faculty of Sport Sciences) Madrid Spain
- Instituto de Investigación Hospital 12 de Octubre Madrid Spain
| | - Juan Del Coso
- Exercise Physiology Laboratory Camilo José Cela University Madrid Spain
| | - Kathryn N. North
- Murdoch Children's Research Institute Melbourne, Victoria Australia
- Department of Paediatrics University of Melbourne The Royal Children's Hospital Melbourne, Victoria Australia
| | - Alejandro Lucia
- Universidad Europea de Madrid (Faculty of Sport Sciences) Madrid Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable Madrid Spain
| | - Nir Eynon
- Institute for Health and Sport (iHeS) Victoria University Victoria Australia
| |
Collapse
|
30
|
Lamb MM, Cai B, Royer J, Pandya S, Soim A, Valdez R, DiGuiseppi C, James K, Whitehead N, Peay H, Venkatesh SY, Matthews D. The effect of steroid treatment on weight in nonambulatory males with Duchenne muscular dystrophy. Am J Med Genet A 2018; 176:2350-2358. [PMID: 30256515 DOI: 10.1002/ajmg.a.40517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/06/2018] [Accepted: 07/23/2018] [Indexed: 11/07/2022]
Abstract
To describe the long-term effect of steroid treatment on weight in nonambulatory males with Duchenne Muscular Dystrophy (DMD), we identified 392 males age 7-29 years with 4,512 weights collected after ambulation loss (176 steroid-naïve and 216 treated with steroids ≥6 months) from the Muscular Dystrophy Surveillance, Tracking, and Research Network (MD STARnet). Comparisons were made between the weight growth curves for steroid-naïve males with DMD, steroid-treated males with DMD, and the US pediatric male population. Using linear mixed-effects models adjusted for race/ethnicity and birth year, we evaluated the association between weight-for-age and steroid treatment characteristics (age at initiation, dosing interval, cumulative duration, cumulative dose, type). The weight growth curves for steroid-naïve and steroid-treated nonambulatory males with DMD were wider than the US pediatric male growth curves. Mean weight-for-age z scores were lower in both steroid-naïve (mean = -1.3) and steroid-treated (mean = -0.02) nonambulatory males with DMD, compared to the US pediatric male population. Longer treatment duration and greater cumulative dose were significantly associated with lower mean weight-for-age z scores. Providers should consider the effect of steroid treatment on weight when making postambulation treatment decisions for males with DMD.
Collapse
Affiliation(s)
- Molly M Lamb
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado
| | - Bo Cai
- Department of Epidemiology and Biostatistics, Arnold School of Public Health, Columbia, South Carolina
| | - Julie Royer
- South Carolina Revenue and Fiscal Affairs Office, Columbia, South Carolina
| | - Shree Pandya
- Department of Neurology, University of Rochester, Rochester, New York
| | - Aida Soim
- New York State Department of Health, Empire State Plaza, Albany, New York
| | - Rodolfo Valdez
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Carolyn DiGuiseppi
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado
| | - Katherine James
- Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado
| | - Nedra Whitehead
- Department of Social, Statistical, and Environmental Sciences, RTI International, Raleigh-Durham, Durham, North Carolina
| | - Holly Peay
- Department of Social, Statistical, and Environmental Sciences, RTI International, Raleigh-Durham, Durham, North Carolina
| | - Swamy Y Venkatesh
- Department of Neurology, University of South Carolina School of Medicine, Columbia, South Carolina
| | - Dennis Matthews
- Department of Pediatric Rehabilitation Medicine, Children's Hospital Colorado, Aurora, Colorado
| | | |
Collapse
|
31
|
Matsuo M, Shirakawa T, Awano H, Nishio H. Receiver operating curve analyses of urinary titin of healthy 3-y-old children may be a noninvasive screening method for Duchenne muscular dystrophy. Clin Chim Acta 2018; 486:110-114. [PMID: 30053403 DOI: 10.1016/j.cca.2018.07.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/21/2018] [Accepted: 07/23/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a progressive, fatal muscle wasting disease. Early detection of DMD by mass screening may enable the early treatment of these patients. We have reported that urinary titin concentration, an indicator of severe muscle wasting, is a diagnostic biomarker for DMD. METHODS Urinary titin concentrations were measured in healthy 3-y-old children and, by comparison with concentrations in 4 DMD patients, and validated as a screening biomarker for DMD. Urine samples were obtained from 100 healthy Japanese children, 52 boys and 48 girls, and their urinary titin concentrations measured by ELISA. RESULTS The mean ± SD urinary titin concentration was 1.5 ± 2.5 nmol/l, and the mean urinary titin concentration normalized to creatinine was 2.2 ± 4.1 pmol/mg creatinine, with no differences between boys and girls. Histograms and box-and-whisker plots showed that almost all titin and normalized titin concentrations were in narrow ranges, with one outlier in common. Receiver operating characteristic curve analysis showed that titin and normalized-titin concentrations from healthy 3-y-olds were completely separate from those of 3-y-old DMD patients. CONCLUSIONS These findings indicate that urinary titin may be an excellent non-invasive biomarker to screen for DMD.
Collapse
Affiliation(s)
- Masafumi Matsuo
- Research Center for Locomotion Biology, Kobe Gakuin University, Kobe, Japan; KNC Department of Nucleic Acid Drug Discovery, Faculty of Rehabilitation, Kobe Gakuin University, Kobe, Japan.
| | - Taku Shirakawa
- Research Center for Locomotion Biology, Kobe Gakuin University, Kobe, Japan; KNC Department of Nucleic Acid Drug Discovery, Faculty of Rehabilitation, Kobe Gakuin University, Kobe, Japan.
| | - Hiroyuki Awano
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Hisahide Nishio
- Research Center for Locomotion Biology, Kobe Gakuin University, Kobe, Japan; Department of Occupational Therapy, Faculty of Rehabilitation, Kobe Gakuin University, Kobe, Japan.
| |
Collapse
|
32
|
Andrews JG, Conway K, Westfield C, Trout C, Meaney FJ, Mathews K, Ciafaloni E, Cunniff C, Fox DJ, Matthews D, Pandya S. Implementation of Duchenne Muscular Dystrophy Care Considerations. Pediatrics 2018; 142:peds.2017-4006. [PMID: 29925575 DOI: 10.1542/peds.2017-4006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/17/2018] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is an X-linked disorder characterized by progressive muscle weakness and multisystem involvement. Recent advances in management of individuals with DMD have prolonged survival. Lack of standardized care spurred an international collaboration to develop consensus-based care considerations for diagnosis and management. In this study, we evaluate adherence to considerations at selected sites. METHODS We collaborated with the Muscular Dystrophy Surveillance, Tracking, and Research Network. Our sample included males with DMD and Becker muscular dystrophy <21 years as of December 31, 2010, with 1 health care encounter on or after January 1, 2012. We collected data from medical records on encounters occurring January 1, 2012, through December 31, 2014. Adherence was determined when frequency of visits or assessments were at or above recommendations for selected care considerations. RESULTS Our analytic sample included 299 individuals, 7% of whom (20/299) were classified as childhood-onset Becker muscular dystrophy. Adherence for neuromuscular and respiratory clinician visits was 65% for the cohort; neuromuscular assessments and corticosteroid side effect monitoring measures ranged from 16% to 68%. Adherence was 83% for forced vital capacity and ≤58% for other respiratory diagnostics. Cardiologist assessments and echocardiograms were found for at least 84%. Transition planning for education or health care was documented for 31% of eligible males. CONCLUSIONS Medical records data were used to identify areas in which practice aligns with the care considerations. However, there remains inconsistency across domains and insufficiency in critical areas. More research is needed to explain this variability and identify reliable methods to measure outcomes.
Collapse
Affiliation(s)
| | | | | | | | - F John Meaney
- Department of Pediatrics, University of Arizona, Tucson, Arizona
| | | | - Emma Ciafaloni
- Department of Neurology, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Christopher Cunniff
- Department of Pediatrics, Weill Cornell Medical College, New York City, New York; and
| | - Deborah J Fox
- Bureau of Environmental and Occupational Epidemiology
| | - Dennis Matthews
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of Colorado, Aurora, Colorado
| | - Shree Pandya
- Department of Neurology, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| |
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the current and emerging therapies for Duchenne muscular dystrophy (DMD). RECENT FINDINGS Coinciding with new standardized care guidelines, there are a growing number of therapeutic options to treat males with DMD. Treatment of the underlying pathobiology, such as micro-dystrophin gene replacement, exon skipping, stop codon read-through agents, and utrophin modulators showed variable success in animal and human studies. Symptomatic therapies to target muscle ischemia, enhance muscle regeneration, prevent muscle fibrosis, inhibit myostatin, and reduce inflammation are also under investigation. DMD is a complex, heterogeneous degenerative disease. The pharmacological and technological achievements made in recent years, plus timely supportive interventions will likely lead to an improved quality of life for many individuals with DMD.
Collapse
Affiliation(s)
- Megan Crone
- Division of Neurology, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada. .,Alberta Children's Hospital, 2888 Shaganappi Trail NW, Calgary, Alberta, T3B 6A8, Canada.
| | - Jean K Mah
- Division of Neurology, Department of Pediatrics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
34
|
Cataract development associated with long-term glucocorticoid therapy in Duchenne muscular dystrophy patients. J AAPOS 2018; 22:192-196. [PMID: 29733899 DOI: 10.1016/j.jaapos.2018.01.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/12/2018] [Accepted: 01/26/2018] [Indexed: 11/22/2022]
Abstract
PURPOSE To evaluate the development of cataracts or elevated intraocular pressure (IOP) in patients with Duchenne muscular dystrophy (DMD) on long-term glucocorticoid (GC) treatment. METHODS The medical records of DMD patients evaluated from 2010 to 2015 at a single center were reviewed retrospectively. The main outcome measures were prevalence of cataracts and elevated IOP, age of first detection of cataract, time from initial steroid use to first detection of cataract, and relative risk of cataract development for deflazacort versus prednisone treatment. RESULTS Of 596 DMD patients, 514 underwent GC therapy; all but one was male. The racial distribution was 82.1% white, 1.0% African American, 5.0% Hispanic, 2.9% Asian, and 8.0% more than one race or "other." The prevalence of cataracts was 22.4% in patients on GC therapy. The mean age at which cataract formation was first documented was 12.9 ± 4.1 years (IQR, 9.6-14.6). The mean time from initial steroid use to the first detection of cataract was 6.5 ± 3.6 years (IQR, 4.0-8.6). The odds of cataract development were 2.4-fold higher for patients on deflazacort compared with prednisone (95% CI, 1.3-4.5; P = 0.004). Only 7 patients (1.4%) underwent cataract surgery, at a mean age of 16.9 years (range, 10.7-24.6 years); all were on deflazacort. Among patients with available intraocular pressure measurements, elevated IOP occurred in only 1 patient (1.1%), who was on deflazacort. CONCLUSIONS In patients undergoing GC therapy for DMD, the rate of cataract formation was slow and well tolerated, with a higher risk among deflazacort patients. The percentage of patients requiring cataract extraction or with elevated IOP was very small. These findings suggest that a schedule of annual eye examinations is appropriate.
Collapse
|
35
|
Hendriksen RGF, Lionarons JM, Hendriksen JGM, Vles JSH, McAdam LC, Biggar WD. Development of a New Self-Reporting Instrument Measuring Benefits and Side Effects of Corticosteroids in Duchenne Muscular Dystrophy: Report from a Pilot Study. J Neuromuscul Dis 2018; 4:217-236. [PMID: 28800336 DOI: 10.3233/jnd-170223] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND There is no cure for Duchenne Muscular Dystrophy (DMD); treatment is symptomatic and corticosteroids slow the progression. Side effects of corticosteroids - especially the physical effects - have been described, however patients' and caregivers perception on chronic corticosteroid treatment and their side effects is less well known, in particular with regards to cognition, behaviour, and emotional functioning. OBJECTIVE The primary aim of this pilot study was to (i) construct a self-report questionnaire to assess the perceived benefits and side effects of corticosteroids for patients with DMD and their parents. Furthermore we aimed to (ii) investigate the psychometric qualities of this questionnaire, (iii) whether there was a difference between parents' and patient's perceptions, and finally (iv) to what extent reported side effects may alter over time. METHODS A 23-item questionnaire (SIDECORT: side effect of corticosteroids) was constructed to assess the perception of these benefits and side effects in a systematic manner. RESULTS In total, 86 patients (aged 5 - 28 years) and 125 of their parents completed the questionnaire. Internal consistency was good. Using factor analyses on the side effect items as reported by parents, two underlying factors were found, with the first factor describing cognitive, behavioural and emotional functioning, and the second factor describing physical functioning. The potential benefits of corticosteroids were highly rated among both parents and patients, although parents rated the importance of the benefits higher than their sons (p = 0.002). Similarly, parents rated the severity of the side effects generally higher than their sons (p = 0.011), especially with regards to the physical side effects (p = 0.014). Based on the parent's perception, the neurodevelopmental side effects generally appeared to decline the longer corticosteroids were used. CONCLUSIONS To our knowledge, this is the first explicit study on perceived cognitive-, behavioural-, and emotional side effects and the allocation of benefits to corticosteroids in DMD. On the basis of our research we suggest a short form questionnaire, which proves to be reliable and valid for research- and clinical practice. This questionnaire could provide useful insights for the care of boys and men with DMD.
Collapse
Affiliation(s)
- Ruben G F Hendriksen
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands.,School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Judith M Lionarons
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands.,School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Jos G M Hendriksen
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands.,Center for Neurological Learning Disabilities, Kempenhaeghe, Heeze, The Netherlands
| | - Johan S H Vles
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands.,School for Mental Health & Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Laura C McAdam
- Holland Bloorview Kids Rehabilitation Hospital, Bloorview Research Institute, Toronto, Canada.,Department of Pediatrics, University of Toronto, Toronto, Canada
| | - W Douglas Biggar
- Holland Bloorview Kids Rehabilitation Hospital, Bloorview Research Institute, Toronto, Canada.,Department of Pediatrics, University of Toronto, Toronto, Canada.,Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
36
|
Nascimento Osorio A, Medina Cantillo J, Camacho Salas A, Madruga Garrido M, Vilchez Padilla JJ. Consensus on the diagnosis, treatment and follow-up of patients with Duchenne muscular dystrophy. Neurologia 2018. [PMID: 29526319 DOI: 10.1016/j.nrl.2018.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is the most common myopathy in children, with a worldwide prevalence of approximately 0.5 cases per 10,000 male births. It is characterised by a progressive muscular weakness manifesting in early childhood, with the subsequent appearance of musculoskeletal, respiratory, and cardiac complications, causing disability, dependence, and premature death. Currently, DMD is mainly managed with multidisciplinary symptomatic treatment, with favourable results in terms of the progression of the disease. It is therefore crucial to establish clear, up-to-date guidelines enabling early detection, appropriate treatment, and monitoring of possible complications. DEVELOPMENT We performed a literature search of the main biomedical databases for articles published in the last 10years in order to obtain an overview of the issues addressed by current guidelines and to identify relevant issues for which no consensus has yet been established. The degree of evidence and level of recommendation of the information obtained were classified and ordered according to the criteria of the American Academy of Neurology. CONCLUSIONS DMD management should be multidisciplinary and adapted to the patient's profile and the stage of clinical progression. In addition to corticotherapy, treatment targeting gastrointestinal, respiratory, cardiac, and orthopaedic problems, as well as physiotherapy, should be provided with a view to improving patients' quality of life. Genetic studies play a key role in the management of the disease, both in detecting cases and potential carriers and in characterising the mutation involved and developing new therapies.
Collapse
Affiliation(s)
- A Nascimento Osorio
- Unidad de Patología Neuromuscular, Servicio de Neurología, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, España
| | - J Medina Cantillo
- Servicio de Medicina Física y Rehabilitación, Hospital Sant Joan de Déu Esplugues de Llobregat, Barcelona, España
| | - A Camacho Salas
- Sección de Neurología Infantil, Servicio de Neurología, Hospital Universitario 12 de Octubre, Madrid, España
| | - M Madruga Garrido
- Sección de Neurología Pediátrica, Hospital Universitario Virgen del Rocío, Sevilla, España
| | - J J Vilchez Padilla
- Servicio de Neurología, Hospital Universitario y Politécnico de La Fe, Valencia, España; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) y Departamento de Medicina, Universidad de Valencia, Valencia, España.
| |
Collapse
|
37
|
Thayer S, Bell C, McDonald CM. The Direct Cost of Managing a Rare Disease: Assessing Medical and Pharmacy Costs Associated with Duchenne Muscular Dystrophy in the United States. J Manag Care Spec Pharm 2018; 23:633-641. [PMID: 28530521 PMCID: PMC10398014 DOI: 10.18553/jmcp.2017.23.6.633] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND A Duchenne muscular dystrophy (DMD) cohort was identified using a claims-based algorithm to estimate health care utilization and costs for commercially insured DMD patients in the United States. Previous analyses have used broad diagnosis codes that include a range of muscular dystrophy types as a proxy to estimate the burden of DMD. OBJECTIVE To estimate DMD-associated resource utilization and costs in a sample of patients identified via a claims-based algorithm using diagnosis codes, pharmacy prescriptions, and procedure codes unique to DMD management based on DMD clinical milestones. METHODS DMD patients were selected from a commercially insured claims database (2000-2009). Patients with claims suggestive of a non-DMD diagnosis or who were aged 30 years or older were excluded. Each DMD patient was matched by age, gender, and region to controls without DMD in a 1:10 ratio (DMD patients n = 75; controls n = 750). All-cause health care resource utilization, including emergency department, inpatient, outpatient, and physician office visits, and all-cause health care costs were examined over a minimum 1-year period. Costs were computed as total health-plan and patient-paid amounts of adjudicated medical claims (in annualized U.S. dollars). RESULTS The average age of the DMD cohort was 13 years. Patients in the DMD cohort had a 10-fold increase in health care costs compared with controls ($23,005 vs. $2,277, P < 0.001). Health care costs were significantly higher for the DMD cohort across age strata and, in particular, for DMD patients aged 14-29 years ($40,132 vs. $2,746, P < 0.001). CONCLUSIONS In the United States, resource use and medical costs of DMD are substantial and increase with age. DISCLOSURES Funding for this study (GHO-10-4441) was provided by GlaxoSmithKline (GSK). Optum was contracted by GSK to conduct the study. Thayer was an employee of Optum Health Economics and Outcomes Research at the time of this study and was not compensated for her participation as an author of this manuscript. Bell is an employee and shareholder of GSK. McDonald has been a consultant for GSK, Sarepta, PTC Therapeutics, Biomarin, and Catabasis on clinical trials regarding Duchenne muscular dystrophy clinical trial design, endpoint selection, and data analysis; Mitobridge for drug development; and Eli Lilly as part of a steering committee for clinical trials. Study concept and design were contributed primarily by Bell, along with Thayer and McDonald. Thayer collected the data, and data interpretation was performed by Thayer and Bell, along with McDonald. The manuscript was written by Thayer and Bell, along with McDonald, and revised by all the authors.
Collapse
Affiliation(s)
| | - Christopher Bell
- 2 US Value, Evidence and Outcomes, GlaxoSmithKline, Research Triangle Park, North Carolina
| | - Craig M McDonald
- 3 Department of Physical Medicine and Rehabilitation and MDA Neuromuscular Disease Clinics, University of California Davis Health System, Sacramento
| |
Collapse
|
38
|
Krawczyk P, Jędrzejewska B, Cysewski P, Janek T. Synthesis, photophysical and biological properties of a new oxazolone fluorescent probe for bioimaging: an experimental and theoretical study. Org Biomol Chem 2018; 15:8952-8966. [PMID: 29043360 DOI: 10.1039/c7ob02439h] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In this study, a new oxazolone derivative 4-{N,N-bis[2-phenyl-4-benzylidene-1,3-oxazol-5(4H)-one]amino}benzaldehyde (PB3) was synthesized and investigated as a fluorescent dye. The spectroscopic properties in different solvents were thoroughly studied. The experimental data were supported by quantum-chemical calculations using density functional theory. Measurements and theoretical calculations showed that the PB3 dye is characterized by non-monotonic solvatochromism, a strongly polar charge transfer excited state, a large Stokes' shift, a high fluorescence quantum yield and a high fluorescence lifetime. Bioconjugate complexes (PB3-concanavalin A) were studied by circular dichroism (CD) spectroscopy. The results showed that the secondary structure of concanavalin A was not significantly influenced by the PB3-fluorophore. Conventional fluorescence microscopy imaging of Candida albicans cells, incubated with the PB3-concanavalin A, was demonstrated. The results from cytochemistry experiments demonstrate that the PB3 dye has valuable advantages compared to the other long-wavelength dyes in typical fluorescence-based cell labeling applications. In vitro tolerance was evaluated by the MTT method in the human colon adenocarcinoma cell line HT29. The PB3 and bioconjugate complexes (PB3-concanavalin A), in the range of concentrations tested, were not considerably toxic. The AutoDock simulations showed LYS46 as the most likely active site for covalent bond formation during PB3-concanavalin A conjugation. In addition, theoretical studies have shown that PB3 is characterized by good bioavailability and absorption/transmission across the cell membrane. This molecule will not bioaccumulate in living organisms and should be excreted in urine without interacting with other drugs. This work provided promising results for the red fluorescent probe (PB3) as a valuable alternative to commercial probes designed for cellular labeling in biological and biomedical research.
Collapse
Affiliation(s)
- Przemysław Krawczyk
- Nicolaus Copernicus University, Collegium Medicum, Faculty of Pharmacy, Chair and Department of Physical Chemistry, Kurpińskiego 5, 85-950 Bydgoszcz, Poland.
| | | | | | | |
Collapse
|
39
|
LoMauro A, Romei M, Gandossini S, Pascuzzo R, Vantini S, D'Angelo MG, Aliverti A. Evolution of respiratory function in Duchenne muscular dystrophy from childhood to adulthood. Eur Respir J 2018; 51:51/2/1701418. [PMID: 29437939 DOI: 10.1183/13993003.01418-2017] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/21/2017] [Indexed: 11/05/2022]
Abstract
In Duchenne muscular dystrophy (DMD), it is still to be determined if specific timepoints can be identified during the natural evolution of respiratory dysfunction from childhood to adulthood and if scoliosis, steroid therapy and nocturnal noninvasive mechanical ventilation (NIMV) have any effect on it.In a 7-year retrospective study performed on 115 DMD patients (6-24 years), evaluated once or twice per year, with 574 visits in total, evolution mean curves of spirometry, lung volumes, spontaneous breathing and thoraco-abdominal pattern (measured by optoelectronic plethysmography) parameters were obtained by nonlinear regression model analysis.While predicted values of forced vital capacity, forced expiratory volume in 1 s, and peak expiratory flow decline continuously since childhood, during spontaneous breathing the following parameters become significantly different than normal in sequence: abdominal contribution to tidal volume (lower after 14.8 years), tidal volume (lower after 17.2 years), minute ventilation (lower after 18.1 years) and respiratory rate (higher after 22.1 years). Restrictive lung pattern and diaphragmatic impairment are exacerbated by scoliosis severity, slowed by steroids treatment and significantly affected by NIMV.Spirometry, lung volumes, breathing pattern and thoraco-abdominal contributions show different evolution curves over time. Specific timepoints of respiratory impairment are identified during disease progression. These should be considered when defining outcome measures in clinical trials and treatment strategies in DMD.
Collapse
Affiliation(s)
- Antonella LoMauro
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano, Italy
| | - Marianna Romei
- Istituto di Ricerca e Cura a Carattere Scientifico "E. Medea", Bosisio Parini, Italy
| | - Sandra Gandossini
- Istituto di Ricerca e Cura a Carattere Scientifico "E. Medea", Bosisio Parini, Italy
| | | | - Simone Vantini
- MOX-Department of Mathematics, Politecnico di Milano, Milano, Italy
| | - Maria Grazia D'Angelo
- Istituto di Ricerca e Cura a Carattere Scientifico "E. Medea", Bosisio Parini, Italy
| | - Andrea Aliverti
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano, Italy
| |
Collapse
|
40
|
Schreiber A, Brochard S, Rippert P, Fontaine-Carbonnel S, Payan C, Poirot I, Hamroun D, Vuillerot C. Corticosteroids in Duchenne muscular dystrophy: impact on the motor function measure sensitivity to change and implications for clinical trials. Dev Med Child Neurol 2018; 60:185-191. [PMID: 28990163 DOI: 10.1111/dmcn.13590] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/24/2017] [Indexed: 12/01/2022]
Abstract
AIM To monitor the evolution of the motor function of ambulatory patients with Duchenne muscular dystrophy (DMD) treated by corticosteroids for 2 years in comparison with untreated patients. METHOD This observational, multicentre cohort study explores the evolution of the motor function measure (MFM) over a 24-month period for 29 ambulant corticosteroids-treated and 45 ambulant untreated patients with DMD. RESULTS Significant differences were found between mean MFM scores in corticosteroids-treated and untreated groups for domain 1 of the MFM (standing position and transfers; D1), domain 2 of the MFM (axial and proximal motor function; D2), and domain 3 of the MFM (distal motor function; D3). Subscores were between 0 months and 6 months, and 0 months and 24 months. For the D1 subscore specifically, there was a significant increase in the corticosteroids-treated group (mean±standard deviation [SD] slope of change=12.6±15.5%/y), while a decrease was observed in the untreated group (-17.8±17.7%/y) between 0 months and 6 months (p<0.001). Sensitivity to change as assessed by standardized response means was high between 12 months and 24 months for D1 of both corticosteroids-treated and untreated groups (1.0 and 1.2 respectively), and low for D2 and D3 of both treated and untreated groups. INTERPRETATION Patients with DMD treated by corticosteroids present a different course of the disease as assessed by MFM, confirming the sensitivity to change of the MFM in this population. WHAT THIS PAPER ADDS Corticosteroids have a quantitative impact on muscle strength 6 to 24 months after starting treatment. Motor function measure is a valid outcome measure in Duchenne muscular dystrophy patients under corticosteroid treatment.
Collapse
Affiliation(s)
- Audrey Schreiber
- Service de Médecine Physique et de Réadaptation, CHRU de Brest, Hôpital Morvan, Brest, France
| | - Sylvain Brochard
- Service de Médecine Physique et de Réadaptation, CHRU de Brest, Hôpital Morvan, Brest, France
| | - Pascal Rippert
- Pôle Information Médicale, Évaluation, Recherche, Hospices Civils de Lyon, Lyon, France.,Service de Médecine Physique et de Réadaptation Pédiatrique, Hospices Civils de Lyon, Hôpital Femme-Mère-Enfant, L'Escale, Bron, France
| | - Stephanie Fontaine-Carbonnel
- Service de Médecine Physique et de Réadaptation Pédiatrique, Hospices Civils de Lyon, Hôpital Femme-Mère-Enfant, L'Escale, Bron, France
| | - Christine Payan
- Service de Pharmacologie Clinique (AP-HP), IHU-AICM, Assistance Publique-Hôpitaux de Paris, Hôpital Pitié Salpêtrière, Paris, France
| | - Isabelle Poirot
- Service de Médecine Physique et de Réadaptation Pédiatrique, Hospices Civils de Lyon, Hôpital Femme-Mère-Enfant, L'Escale, Bron, France
| | - Dalil Hamroun
- Direction de la Recherche et de l'Innovation, CHRU de Montpellier, Hôpital Arnaud de Villeneuve, Montpellier, France
| | - Carole Vuillerot
- Service de Médecine Physique et de Réadaptation Pédiatrique, Hospices Civils de Lyon, Hôpital Femme-Mère-Enfant, L'Escale, Bron, France.,CNRS UMR 5558, Laboratoire de Biométrie et Biologie Evolutive, Equipe Biostatistique Santé, Pierre-Bénite, France.,Université Lyon I, Lyon, France
| | | |
Collapse
|
41
|
Mu X, Tang Y, Takayama K, Chen W, Lu A, Wang B, Weiss K, Huard J. RhoA/ROCK inhibition improves the beneficial effects of glucocorticoid treatment in dystrophic muscle: implications for stem cell depletion. Hum Mol Genet 2018; 26:2813-2824. [PMID: 28549178 DOI: 10.1093/hmg/ddx117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/10/2017] [Indexed: 12/31/2022] Open
Abstract
Glucocorticoid treatment represents a standard palliative treatment for Duchenne muscular dystrophy (DMD) patients, but various adverse effects have limited this treatment. In an effort to understand the mechanism(s) by which glucocorticoids impart their effects on the dystrophic muscle, and potentially reduce the adverse effects, we have studied the effect of prednisolone treatment in dystrophin/utrophin double knockout (dKO) mice, which exhibit a severe dystrophic phenotype due to rapid muscle stem cell depletion. Our results indicate that muscle stem cell depletion in dKO muscle is related to upregulation of mTOR, and that prednisolone treatment reduces the expression of mTOR and other pro-inflammatory mediators, consequently slowing down muscle stem cell depletion. However, prednisolone treatment was unable to improve the myogenesis of stem cells and reduce fibrosis in dKO muscle. We then studied whether glucocorticoid treatment can be improved by co-administration of an inhibitor of RhoA/ROCK signaling, which can be activated by glucocorticoids and was found in our previous work to be over-activated in dystrophic muscle. Our results indicate that the combination of RhoA/ROCK inhibition and glucocorticoid treatment in dystrophic muscle have a synergistic effect in alleviating the dystrophic phenotype. Taken together, our study not only shed light on the mechanism by which glucocorticoid imparts its beneficial effect on dystrophic muscle, but also revealed the synergistic effect of RhoA/ROCK inhibition and glucocorticoid treatment, which could lead to the development of more efficient therapeutic approaches for treating DMD patients.
Collapse
Affiliation(s)
- Xiaodong Mu
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Ying Tang
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Koji Takayama
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Wanqun Chen
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Department of Biochemistry and Molecular Biology, Jinan University, Guangdong, China
| | - Aiping Lu
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Bing Wang
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Kurt Weiss
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX 77054, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| |
Collapse
|
42
|
Ross LF, Clarke AJ. A Historical and Current Review of Newborn Screening for Neuromuscular Disorders From Around the World: Lessons for the United States. Pediatr Neurol 2017; 77:12-22. [PMID: 29079012 DOI: 10.1016/j.pediatrneurol.2017.08.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 07/31/2017] [Accepted: 08/20/2017] [Indexed: 01/16/2023]
Abstract
BACKGROUND We aimed to review the history of newborn screening for three neuromuscular disorders (Duchenne muscular dystrophy, Pompe disease, and spinal muscular atrophy [SMA]) to determine best practices. METHODS The history of newborn screening for Duchenne muscular dystrophy began in 1975 with the measurement of creatinine kinase on newborn male blood spots from two Midwestern hospitals in the United States. Over the next 40 years, ten programs were implemented around the globe although none currently remain. The first experimental pilot program for Pompe disease began in 2005 in Taiwan. In 2013, Missouri was the first US state to implement Pompe newborn screening before its inclusion in the Recommended Uniform Screening Panel (RUSP) in 2015 by the Advisory Committee on Heritable Disorders in Newborns and Children (United States). In 2008, SMA was reviewed and rejected for inclusion in the RUSP because no treatment existed. With the approval of nusinersen in late 2016, spinal muscular atrophy is being reconsidered for the RUSP. RESULTS A condition should meet public health screening criteria to be included in the RUSP. Duchenne muscular dystrophy, Pompe, and SMA challenge traditional screening criteria: Duchenne muscular dystrophy does not present in infancy and lacks effective treatment; Pompe and SMA may not present until adulthood; and safety and efficacy of long-term intrathecal treatment for SMA is unknown. Potential reproductive benefit and improved research recruitment do not justify a public health screening program. CONCLUSIONS This review provides lessons that could benefit US public health departments as they consider expanding screening to include neuromuscular disorders like Duchenne muscular dystrophy, Pompe, and SMA.
Collapse
Affiliation(s)
- Lainie Friedman Ross
- Clinical Ethics, Departments of Pediatrics, Medicine, Surgery and the College, MacLean Center for Clinical Medical Ethics, University of Chicago, Chicago Illinois.
| | - Angus John Clarke
- Clinical Genetics, Institute of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
43
|
Feingold B, Mahle WT, Auerbach S, Clemens P, Domenighetti AA, Jefferies JL, Judge DP, Lal AK, Markham LW, Parks WJ, Tsuda T, Wang PJ, Yoo SJ. Management of Cardiac Involvement Associated With Neuromuscular Diseases: A Scientific Statement From the American Heart Association. Circulation 2017; 136:e200-e231. [DOI: 10.1161/cir.0000000000000526] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
44
|
Finder J, Mayer OH, Sheehan D, Sawnani H, Abresch RT, Benditt J, Birnkrant DJ, Duong T, Henricson E, Kinnett K, McDonald CM, Connolly AM. Pulmonary Endpoints in Duchenne Muscular Dystrophy. A Workshop Summary. Am J Respir Crit Care Med 2017. [DOI: 10.1164/rccm.201703-0507ws] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Jonathan Finder
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Oscar Henry Mayer
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Daniel Sheehan
- Department of Pediatrics, University at Buffalo, The State University of New York, Buffalo, New York
| | - Hemant Sawnani
- Division of Pulmonology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | | | - Joshua Benditt
- Division of Pulmonary and Critical Care Medicine, University of Washington School of Medicine, Seattle, Washington
| | - David J. Birnkrant
- MetroHealth Medical Center, Cleveland, Ohio
- Case Western Reserve University, Cleveland, Ohio
| | - Tina Duong
- Department of Neurology, Stanford University, Stanford, California
| | | | - Kathi Kinnett
- Parent Project Muscular Dystrophy, Hackensack, New Jersey; and
| | - Craig M. McDonald
- Department of Physical Medicine and Rehabilitation and
- Department of Pediatrics, University of California Davis, Sacramento, California
| | - Anne M. Connolly
- Department of Neurology, Washington University School of Medicine, Saint Louis, Missouri
| |
Collapse
|
45
|
Mâncio RD, Hermes TDA, Macedo AB, Mizobuti DS, Rupcic IF, Minatel E. Dystrophic phenotype improvement in the diaphragm muscle of mdx mice by diacerhein. PLoS One 2017; 12:e0182449. [PMID: 28787441 PMCID: PMC5546703 DOI: 10.1371/journal.pone.0182449] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/18/2017] [Indexed: 12/14/2022] Open
Abstract
Chronic inflammation and oxidative stress are striking features of Duchenne muscular dystrophy disease. Diacerhein is an anthraquinone, which exhibits anti-inflammatory and antioxidant properties. Based on their actions, the present study evaluated the effects of diacerhein against myonecrosis, oxidative stress and inflammatory response in the diaphragm muscle of mdx mice and compared these results to current treatment widely used in DMD patients, with a main focus on the impact of prednisone. The results demonstrated that diacerhein treatment prevented muscle damage indicated by a decrease in the IgG uptake by muscle fibers, lower CK levels in serum, reduction of fibers with central nuclei with a concomitant increase in fibers with peripheral nuclei. It also had an effect on the inflammatory process, decreasing the inflammatory area, macrophage staining and TNF-α and IL-1β content. Regarding oxidative stress, diacerhein treatment was effective in reducing the ROS and lipid peroxidation in the diaphragm muscle from mdx mice. Compared to prednisone treatment, our findings demonstrated that diacerhein treatment improved the dystrophic phenotype in the diaphragm muscle of mdx mice similar to that of glucocorticoid therapy. In this respect, this work suggests that diacerhein has a potential use as an alternative drug in dystrophinopathy treatment and recommends that its anti-inflammatory and antioxidants properties in the dystrophic muscle should be better understood.
Collapse
Affiliation(s)
- Rafael Dias Mâncio
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Túlio de Almeida Hermes
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Aline Barbosa Macedo
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Daniela Sayuri Mizobuti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Ian Feller Rupcic
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Elaine Minatel
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
46
|
Montes J, Young SD, Mazzone E, Main M. Workshop report. Neuromuscul Disord 2017; 27:S0960-8966(17)30577-1. [PMID: 28917631 DOI: 10.1016/j.nmd.2017.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 07/04/2017] [Accepted: 07/06/2017] [Indexed: 11/21/2022]
Affiliation(s)
- Jacqueline Montes
- Departments of Rehabilitation and Regenerative Medicine and Neurology, Columbia University Medical Center, New York, NY, USA.
| | - Sally Dunaway Young
- Departments of Rehabilitation and Regenerative Medicine and Neurology, Columbia University Medical Center, New York, NY, USA
| | - Elena Mazzone
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | - Marion Main
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
47
|
Newborn screening for Duchenne muscular dystrophy in China: follow-up diagnosis and subsequent treatment. World J Pediatr 2017; 13:197-201. [PMID: 28466241 DOI: 10.1007/s12519-017-0036-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/06/2016] [Indexed: 01/15/2023]
Abstract
BACKGROUND Newborn screening for Duchenne muscular dystrophy (DMD) is currently being initiated in Zhejiang Province, China and is under consideration in other countries, including the United States. As China begins to implement DMD newborn screening (DMD-NBS), there is ongoing discussion regarding the steps forward for follow up care of positively identified patients as well as false positive and false negative results. DATA SOURCES Relevant papers related to DMD-NBS, and NBS in China were reviewed in PubMed. RESULTS The current state of DMD-NBS is discussed, along with the steps needed to effectively screen infants for this disease in China, recommendations for establishment of follow up care in patients with positive and negative screens, and measurement of patient outcomes. CONCLUSIONS Zhejiang Province, China is ready to implement DMD-NBS. Future challenges that exist for this program, and other countries, include the ability to track patients, assist with access to care, and ensure adequate follow-up care according to evidence-based guidelines. In addition, China's large rural population, lack of specialty providers, and difficulty in educating patients regarding the benefits of treatment create challenges that will need to be addressed.
Collapse
|
48
|
Ward LM, Kinnett K, Bonewald L. Proceedings of a Parent Project Muscular Dystrophy Bone Health Workshop: Morbidity due to osteoporosis in DMD: The Path Forward May 12-13, 2016, Bethesda, Maryland, USA. Neuromuscul Disord 2017; 28:64-76. [PMID: 28756052 DOI: 10.1016/j.nmd.2017.05.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/04/2017] [Accepted: 05/11/2017] [Indexed: 10/19/2022]
Affiliation(s)
- Leanne M Ward
- Division of Endocrinology and Metabolism, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada.
| | - Kathi Kinnett
- Parent Project Muscular Dystrophy, Middletown, OH, USA
| | - Lynda Bonewald
- Indiana Center for Musculoskeletal Health, Departments of Anatomy and Cell Biology and Orthopaedic Surgery, Indiana University, Indianapolis, IN, USA
| | | |
Collapse
|
49
|
Guglieri M, Bushby K, McDermott MP, Hart KA, Tawil R, Martens WB, Herr BE, McColl E, Wilkinson J, Kirschner J, King WM, Eagle M, Brown MW, Willis T, Hirtz D, Shieh PB, Straub V, Childs AM, Ciafaloni E, Butterfield RJ, Horrocks I, Spinty S, Flanigan KM, Kuntz NL, Baranello G, Roper H, Morrison L, Mah JK, Manzur AY, McDonald CM, Schara U, von der Hagen M, Barohn RJ, Campbell C, Darras BT, Finkel RS, Vita G, Hughes I, Mongini T, Pegoraro E, Wicklund M, Wilichowski E, Bryan Burnette W, Howard JF, McMillan HJ, Thangarajh M, Griggs RC. Developing standardized corticosteroid treatment for Duchenne muscular dystrophy. Contemp Clin Trials 2017; 58:34-39. [PMID: 28450193 DOI: 10.1016/j.cct.2017.04.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 03/27/2017] [Accepted: 04/24/2017] [Indexed: 10/19/2022]
Abstract
Despite corticosteroids being the only treatment documented to improve strength and function in boys with Duchenne muscular dystrophy (DMD) corticosteroid prescription is inconsistent and in some countries, corticosteroids are not prescribed. We are conducting a clinical trial that (1) compares the 3 most frequently prescribed corticosteroid regimes; (2) standardizes treatment of DMD complications; and (3) standardizes prevention of corticosteroid side effects. Investigators at 38 sites in 5 countries plan to recruit 300 boys aged 4-7 who are randomly assigned to one of three regimens: daily prednisone; daily deflazacort; or intermittent prednisone (10days on/10days off). Boys are followed for a minimum of 3years to assess the relative effectiveness and adverse event profiles of the different regimens. The primary outcome is a 3-dimensional variable consisting of log-transformed time to rise from the floor, forced vital capacity, and subject/parent satisfaction with treatment, each averaged over all post-baseline visits. The study protocol includes evidence- and consensus-based treatment of DMD complications and of corticosteroid side effects. This study seeks to establish a standard corticosteroid regimen for DMD. Since all new interventions for DMD are being developed as add-on therapies to corticosteroids, defining the optimum regimen is of importance for all new treatments.
Collapse
Affiliation(s)
- Michela Guglieri
- John Walton Muscular Dystrophy Research Centre, Newcastle University, United Kingdom.
| | - Kate Bushby
- John Walton Muscular Dystrophy Research Centre, Newcastle University, United Kingdom
| | | | | | - Rabi Tawil
- University of Rochester Medical Center, United States
| | | | | | | | | | | | - Wendy M King
- University of Rochester Medical Center, United States
| | - Michele Eagle
- John Walton Muscular Dystrophy Research Centre, Newcastle University, United Kingdom
| | - Mary W Brown
- University of Rochester Medical Center, United States
| | - Tracey Willis
- The Robert Jones and Agnes Hunt Orthopaedic Hospital, NHS Foundation Trust, Oswestry, United Kingdom
| | | | | | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Newcastle University, United Kingdom
| | | | | | | | - Iain Horrocks
- Greater Glasgow and Clyde NHS Yorkhill Hospital, United Kingdom
| | | | | | - Nancy L Kuntz
- Ann and Robert H. Lurie Children's Hospital, United States
| | | | - Helen Roper
- Birmingham Heartlands Hospital, United Kingdom
| | | | | | | | | | | | | | | | - Craig Campbell
- Children's Hospital London Health Sciences Centre, Canada
| | | | | | - Giuseppe Vita
- University of Messina AOU Policlinico Gaetano Martino, Italy
| | - Imelda Hughes
- Royal Manchester Children's Hospital, United Kingdom
| | | | | | | | | | | | - James F Howard
- University of North Carolina School of Medicine, United States
| | | | | | | |
Collapse
|
50
|
Wong BL, Rybalsky I, Shellenbarger KC, Tian C, McMahon MA, Rutter MM, Sawnani H, Jefferies JL. Long-Term Outcome of Interdisciplinary Management of Patients with Duchenne Muscular Dystrophy Receiving Daily Glucocorticoid Treatment. J Pediatr 2017; 182:296-303.e1. [PMID: 28043681 DOI: 10.1016/j.jpeds.2016.11.078] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 10/12/2016] [Accepted: 11/29/2016] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To evaluate clinical outcomes and steroid side effects in a cohort of patients with Duchenne muscular dystrophy (DMD) treated with long-term daily glucocorticoid therapy. Although daily glucocorticoid therapy has been shown to extend ambulatory function in DMD, less frequent dosing is often used because of side effect concerns. STUDY DESIGN Retrospective study of 97 patients with DMD aged 10 to <16 years treated with daily glucocorticoid (89% on deflazacort) for a mean of 8.5 years. Outcome measures were motor, pulmonary, and cardiac function, and scoliosis. Side effects were growth failure and weight gain, facial fullness, blood pressure, bone health, cataracts, gastrointestinal symptoms, behavior, hypertrichosis, and need for medication interventions. RESULTS For 13- to 16-year-old patients, 40% could rise from the floor and 50% could perform the 30-foot run test. Forced vital capacity for the entire cohort was well preserved. Thirteen percent of younger (10- to <13-year-old) and 21% of older patients had findings of left ventricle systolic dysfunction. Six percent (all aged 16 years) developed scoliosis (Cobb angle >20 degrees). Eighty-six percent had normal weight velocities; 30% had no increased facial fullness; 72% had short stature; and 19% had asymptomatic cataracts. Asymptomatic spine compression deformities were noted in 76% and long bone fractures in 30%. One patient stopped glucocorticoid because of behavioral concerns. CONCLUSIONS With evidence for improved outcomes and manageable side effects, we recommend use of daily glucocorticoid therapy for patients with DMD with anticipatory management of side effects and a coordinated interdisciplinary care approach.
Collapse
Affiliation(s)
- Brenda L Wong
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.
| | - Irina Rybalsky
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Karen C Shellenbarger
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Cuixia Tian
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Mary A McMahon
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Physical Medicine and Rehabilitation, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Meilan M Rutter
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Hemant Sawnani
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - John L Jefferies
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|