151
|
Michael E, Sofou K, Wahlgren L, Kroksmark AK, Tulinius M. Long term treatment with ataluren-the Swedish experience. BMC Musculoskelet Disord 2021; 22:837. [PMID: 34592975 PMCID: PMC8485550 DOI: 10.1186/s12891-021-04700-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 09/09/2021] [Indexed: 11/26/2022] Open
Abstract
Introduction
Ataluren is a relatively new treatment for male patients with Duchenne muscular dystrophy (DMD) due to a premature stop codon. Long-term longitudinal data as well as efficacy data on non-ambulant patients are still lacking. Here we present the results from a long-term follow-up study of all DMD patients treated with ataluren and followed at the Queen Silvia Children’s Hospital in Gothenburg, Sweden, with focus on the evolution of patients’ upper motor and respiratory function over time. Methods This is a retrospective longitudinal case-series study of all male DMD patients treated with ataluren and followed at the Queen Silvia Children’s Hospital in Gothenburg, Sweden, since 2008. Results Our eleven patients had a median exposure to ataluren of 2312 days which is almost a fourfold higher than previous studies. Loss of ambulation occurred at a median age of 13.2 years. Patients who lost ambulation prior to 13.2 years of age had received ataluren for 5 years, whereas patients who continued to be ambulatory after 13.2 years of age had received ataluren for 6.5 years until loss of ambulation or last follow-up if still ambulatory. Four of six non ambulatory patients had Performance of the Upper Limb scores above the expected mean values over time. All but one patient maintained a pulmonary decline above the expected over time. All ambulatory patients increased in their predicted forced vital capacity (FVC) with 2.8 to 8.2% annually. Following loss of ambulation, 5 of 6 patients declined in predicted FVC (%), with annual rate of decline varying from 1.8 to 21.1%. The treatment was safe and well tolerated throughout the follow-up period. Conclusions This is the first study to present long-term cumulative treatment outcomes over a median period of 6.3 years on ataluren treatment. Our results indicate a delay in loss of ambulation, as well as a slower decline in FVC and upper limb motor function even after loss of ambulation. We suggest that treatment with ataluren should be initiated as soon as the diagnosis is confirmed, closely monitored and, in case of sustainable benefit, continued even after loss of ambulation.
Collapse
Affiliation(s)
- Eva Michael
- Department of Paediatrics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden. .,Department of Paediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Kalliopi Sofou
- Department of Paediatrics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Paediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lisa Wahlgren
- Department of Paediatrics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Paediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna-Karin Kroksmark
- Department of Paediatrics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Paediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Már Tulinius
- Department of Paediatrics, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Paediatrics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
152
|
Ohlendieck K, Swandulla D. Complexity of skeletal muscle degeneration: multi-systems pathophysiology and organ crosstalk in dystrophinopathy. Pflugers Arch 2021; 473:1813-1839. [PMID: 34553265 PMCID: PMC8599371 DOI: 10.1007/s00424-021-02623-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy is a highly progressive muscle wasting disorder due to primary abnormalities in one of the largest genes in the human genome, the DMD gene, which encodes various tissue-specific isoforms of the protein dystrophin. Although dystrophinopathies are classified as primary neuromuscular disorders, the body-wide abnormalities that are associated with this disorder and the occurrence of organ crosstalk suggest that a multi-systems pathophysiological view should be taken for a better overall understanding of the complex aetiology of X-linked muscular dystrophy. This article reviews the molecular and cellular effects of deficiency in dystrophin isoforms in relation to voluntary striated muscles, the cardio-respiratory system, the kidney, the liver, the gastrointestinal tract, the nervous system and the immune system. Based on the establishment of comprehensive biomarker signatures of X-linked muscular dystrophy using large-scale screening of both patient specimens and genetic animal models, this article also discusses the potential usefulness of novel disease markers for more inclusive approaches to differential diagnosis, prognosis and therapy monitoring that also take into account multi-systems aspects of dystrophinopathy. Current therapeutic approaches to combat muscular dystrophy are summarised.
Collapse
Affiliation(s)
- Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Co. Kildare, Maynooth, W23F2H6, Ireland.
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Co. Kildare, Maynooth, W23F2H6, Ireland.
| | - Dieter Swandulla
- Institute of Physiology, University of Bonn, 53115, Bonn, Germany.
| |
Collapse
|
153
|
Saclier M, Ben Larbi S, My Ly H, Moulin E, Mounier R, Chazaud B, Juban G. Interplay between myofibers and pro-inflammatory macrophages controls muscle damage in mdx mice. J Cell Sci 2021; 134:272022. [PMID: 34471933 DOI: 10.1242/jcs.258429] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/30/2021] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy is a genetic muscle disease characterized by chronic inflammation and fibrosis mediated by a pro-fibrotic macrophage population expressing pro-inflammatory markers. Our aim was to characterize cellular events leading to the alteration of macrophage properties and to modulate macrophage inflammatory status using the gaseous mediator hydrogen sulfide (H2S). Using co-culture experiments, we first showed that myofibers derived from mdx mice strongly skewed the polarization of resting macrophages towards a pro-inflammatory phenotype. Treatment of mdx mice with NaHS, an H2S donor, reduced the number of pro-inflammatory macrophages in skeletal muscle, which was associated with a decreased number of nuclei per fiber, as well as reduced myofiber branching and fibrosis. Finally, we established the metabolic sensor AMP-activated protein kinase (AMPK) as a critical NaHS target in muscle macrophages. These results identify an interplay between myofibers and macrophages where dystrophic myofibers contribute to the maintenance of a highly inflammatory environment sustaining a pro-inflammatory macrophage status, which in turn favors myofiber damage, myofiber branching and establishment of fibrosis. Our results also highlight the use of H2S donors as a potential therapeutic strategy to improve the dystrophic muscle phenotype by dampening chronic inflammation. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Marielle Saclier
- Department of Biosciences, University of Milan, 20133 Milan, Italy
| | - Sabrina Ben Larbi
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, 69008 Lyon, France
| | - Ha My Ly
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, 69008 Lyon, France
| | - Eugénie Moulin
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, 69008 Lyon, France
| | - Rémi Mounier
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, 69008 Lyon, France
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, 69008 Lyon, France
| | - Gaëtan Juban
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U1217, Université de Lyon, 69008 Lyon, France
| |
Collapse
|
154
|
Donaldson A, Guntrum D, Ciafaloni E, Statland J. Achieving Life Milestones in Duchenne/Becker Muscular Dystrophy: A Retrospective Analysis. Neurol Clin Pract 2021; 11:311-317. [PMID: 34484931 DOI: 10.1212/cpj.0000000000000970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 07/30/2020] [Indexed: 01/03/2023]
Abstract
Objective To understand the milestones achieved in the transition from childhood to adulthood for patients with Duchenne and Becker muscular dystrophies (DMD/BMD). Methods We performed a retrospective chart review on patients aged 15 years or older with a clinical diagnosis of DMD/BMD who received care from January 1, 2008, to January 1, 2018 at the University of Kansas Medical Center and the University of Rochester Medical Center. Participants were identified using local Muscular Dystrophy Asssociation-funded clinic lists, neuromuscular research databases, and electronic medical record review. Data were abstracted using a uniform template on education, employment, community resources, relationships, and end-of-life discussions and is presented as mean, median, or frequency with associated 95% confidence interval (CI). Results A total of 109 patients were identified: patients ranged in age from 15 to 56 years with a median of 24, and covered a 5-state region and Ontario, Canada. Seventy-eight percent of patients had DMD and were, on average, 8.5 years younger than patients with BMD. Over half (56.9%, 95% CI 47.6-66.2) were high school graduates or beyond. Sixteen percent did not have their highest level of education documented. Only 20.0% had an occupation (95% CI 12.7-27.7), most frequently in education and administrative support (34%). The majority were still living with parents (80.7%, 95% CI 73.3-88.1). A minority reported having end-of-life discussions (17.4%, 95% CI 10.3-24.6). Conclusions Psychosocial elements reflecting the transition to adulthood are inconsistently reported in clinical documentation. A prospective study will further elucidate this transition.
Collapse
Affiliation(s)
- Andrew Donaldson
- University of Kansas Medical Center (AD, JS); and University of Rochester Medical Center (DG, EC), NY
| | - Debra Guntrum
- University of Kansas Medical Center (AD, JS); and University of Rochester Medical Center (DG, EC), NY
| | - Emma Ciafaloni
- University of Kansas Medical Center (AD, JS); and University of Rochester Medical Center (DG, EC), NY
| | - Jeffrey Statland
- University of Kansas Medical Center (AD, JS); and University of Rochester Medical Center (DG, EC), NY
| |
Collapse
|
155
|
Dang UJ, Ziemba M, Clemens PR, Hathout Y, Conklin LS, Hoffman EP. Serum biomarkers associated with baseline clinical severity in young steroid-naïve Duchenne muscular dystrophy boys. Hum Mol Genet 2021; 29:2481-2495. [PMID: 32592467 PMCID: PMC7471506 DOI: 10.1093/hmg/ddaa132] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/28/2020] [Accepted: 06/20/2020] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by loss of dystrophin in muscle, and while all patients share the primary gene and biochemical defect, there is considerable patient–patient variability in clinical symptoms. We sought to develop multivariate models of serum protein biomarkers that explained observed variation, using functional outcome measures as proxies for severity. Serum samples from 39 steroid-naïve DMD boys 4 to <7 years enrolled into a clinical trial of vamorolone were studied (NCT02760264). Four assessments of gross motor function were carried out for each participant over a 6-week interval, and their mean was used as response for biomarker models. Weighted correlation network analysis was used for unsupervised clustering of 1305 proteins quantified using SOMAscan® aptamer profiling to define highly representative and connected proteins. Multivariate models of biomarkers were obtained for time to stand performance (strength phenotype; 17 proteins) and 6 min walk performance (endurance phenotype; 17 proteins) including some shared proteins. Identified proteins were tested with associations of mRNA expression with histological severity of muscle from dystrophinopathy patients (n = 28) and normal controls (n = 6). Strong associations predictive of both clinical and histological severity were found for ERBB4 (reductions in both blood and muscle with increasing severity), SOD1 (reductions in muscle and increases in blood with increasing severity) and CNTF (decreased levels in blood and muscle with increasing severity). We show that performance of DMD boys was effectively modeled with serum proteins, proximal strength associated with growth and remodeling pathways and muscle endurance centered on TGFβ and fibrosis pathways in muscle.
Collapse
Affiliation(s)
- Utkarsh J Dang
- Department of Health Outcomes and Administrative Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University-SUNY, Binghamton, NY 13902, USA
| | - Michael Ziemba
- Department of Biomedical Engineering, Watson School of Engineering, Binghamton University-SUNY, Binghamton, NY 13902, USA
| | - Paula R Clemens
- Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Veteran Affairs Medical Center, Pittsburgh, PA 15213, USA
| | - Yetrib Hathout
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University-SUNY, Binghamton, NY 13902, USA
| | | | | | - Eric P Hoffman
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University-SUNY, Binghamton, NY 13902, USA.,ReveraGen BioPharma, Rockville, MD 20850, USA
| |
Collapse
|
156
|
Houwen-van Opstal SLS, Heutinck L, Jansen M, Krom YD, Cup EHC, Hendriksen JGM, Willemsen MAAP, Verschuuren JJGM, Niks EH, de Groot IJM. Occurrence of symptoms in different stages of Duchenne muscular dystrophy and their impact on social participation. Muscle Nerve 2021; 64:701-709. [PMID: 34453345 PMCID: PMC9292483 DOI: 10.1002/mus.27406] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 08/19/2021] [Accepted: 08/21/2021] [Indexed: 01/08/2023]
Abstract
Introduction/Aims As life expectancy improves for patients with Duchenne muscular dystrophy (DMD), new symptoms are likely to arise. This aims of this study are: (1) to explore the prevalence of a broad variety of symptoms in the various stages of DMD (with and without steroid use); (2) to explore the prevalence of common secondary diagnoses; and (3) to evaluate the social participation level of patients with DMD older than 16 y of age; and to explore correlations between social participation and symptoms. Methods A cross‐sectional self‐report questionnaire, including questions on functional level and health status, as well as a standardized participation scale was distributed among Dutch patients with DMD. Results Eighty‐four male patients with a mean age of 22.0 (SD = 10.0) y were enrolled. The most prevalent and limiting symptoms were difficulty coughing (58%), coldness of hands (57%), contractures (51%), stiffness (49%), fatigue (40%), myalgia (38%), and low speech volume (33%). Prevalent secondary diagnoses included cardiac disease (14%), neurobehavioral diagnosis (13%), low blood pressure (13%), and arthrosis (5%). Social participation correlated negatively with coldness of hands (r = − .29; P < .03), decreased intelligibility (r = − .40; P < .003), and chewing problems (r = − .33; P < .02). Discussion The prevalence of a broad spectrum of symptoms and secondary diagnoses is high in patients with DMD, and some of these symptoms are correlated with social participation. Growing awareness of new symptoms and secondary diagnoses among patients, caregivers, and professionals can enhance their recognition, possibly facilitating prevention and early treatment.
Collapse
Affiliation(s)
- Saskia L S Houwen-van Opstal
- Amalia Children's Hospital, Nijmegen, The Netherlands.,Radboud University, Nijmegen, The Netherlands.,Donders Centre for Neuroscience, Nijmegen, The Netherlands.,Duchenne Centre Netherlands, Nijmegen and Leiden, The Netherlands
| | - Lotte Heutinck
- Radboud University, Nijmegen, The Netherlands.,Duchenne Centre Netherlands, Nijmegen and Leiden, The Netherlands
| | | | - Yvonne D Krom
- Duchenne Centre Netherlands, Nijmegen and Leiden, The Netherlands.,Leiden University Medical Center, Leiden, The Netherlands
| | | | - Jos G M Hendriksen
- Duchenne Centre Netherlands, Nijmegen and Leiden, The Netherlands.,Kempenhaeghe Center for Neurological Learning Disabilities, Heeze, The Netherlands
| | - Michel A A P Willemsen
- Amalia Children's Hospital, Nijmegen, The Netherlands.,Radboud University, Nijmegen, The Netherlands
| | - Jan J G M Verschuuren
- Duchenne Centre Netherlands, Nijmegen and Leiden, The Netherlands.,Leiden University Medical Center, Leiden, The Netherlands
| | - Erik H Niks
- Duchenne Centre Netherlands, Nijmegen and Leiden, The Netherlands.,Leiden University Medical Center, Leiden, The Netherlands
| | - Imelda J M de Groot
- Amalia Children's Hospital, Nijmegen, The Netherlands.,Radboud University, Nijmegen, The Netherlands.,Duchenne Centre Netherlands, Nijmegen and Leiden, The Netherlands
| |
Collapse
|
157
|
Lai X, Chen J. C-X-C motif chemokine ligand 12: a potential therapeutic target in Duchenne muscular dystrophy. Bioengineered 2021; 12:5428-5439. [PMID: 34424816 PMCID: PMC8806931 DOI: 10.1080/21655979.2021.1967029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused by a mutant dystrophin protein. DMD patients undergo gradual progressive paralysis until death. Chronic glucocorticoid therapy remains one of the main treatments for DMD, despite the significant side effects. However, its mechanisms of action remain largely unknown. We used bioinformatics tools to identify pathogenic genes involved in DMD and glucocorticoid target genes. Two gene expression profiles containing data from DMD patients and healthy controls (GSE38417 and GSE109178) were downloaded for further analysis. Differentially expressed genes (DEGs) between DMD patients and controls were identified using GEO2R, and glucocorticoid target genes were predicted from the Pharmacogenetics and Pharmacogenomics Knowledge Base. Surprisingly, only one gene, CXCL12 (C-X-C motif chemokine ligand 12), was both a glucocorticoid target and a DEG. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, Gene Ontology term enrichment analysis, and gene set enrichment analysis were performed. A protein-protein interaction network was constructed and hub genes identified using the Search Tool for the Retrieval of Interacting Genes (STRING) database and Cytoscape. Enriched pathways involving the DEGs, including CXCL12, were associated with the immune response and inflammation. Levels of CXCL12 and its receptor CXCR4 (C-X-C motif chemokine receptor 4) were increased in X-linked muscular dystrophy (mdx) mice (DMD models) but became significantly reduced after prednisone treatment. Metformin also reduced the expression of CXCL12 and CXCR4 in mdx mice. In conclusion, the CXCL12-CXCR4 pathway may be a potential target for DMD therapy.
Collapse
Affiliation(s)
- Xinsheng Lai
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| | - Jie Chen
- School of Life Science, Nanchang University, Nanchang, Jiangxi, China.,Laboratory of Synaptic Development and Plasticity, Institute of Life Science, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
158
|
Mitelman O, Abdel-Hamid HZ, Byrne BJ, Connolly AM, Heydemann P, Proud C, Shieh PB, Wagner KR, Dugar A, Santra S, Signorovitch J, Goemans N, McDonald CM, Mercuri E, Mendell JR. A Combined Prospective and Retrospective Comparison of Long-Term Functional Outcomes Suggests Delayed Loss of Ambulation and Pulmonary Decline with Long-Term Eteplirsen Treatment. J Neuromuscul Dis 2021; 9:39-52. [PMID: 34420980 PMCID: PMC8842766 DOI: 10.3233/jnd-210665] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background: Studies 4658-201/202 (201/202) evaluated treatment effects of eteplirsen over 4 years in patients with Duchenne muscular dystrophy and confirmed exon-51 amenable genetic mutations. Chart review Study 4658-405 (405) further followed these patients while receiving eteplirsen during usual clinical care. Objective: To compare long-term clinical outcomes of eteplirsen-treated patients from Studies 201/202/405 with those of external controls. Methods: Median total follow-up time was approximately 6 years of eteplirsen treatment. Outcomes included loss of ambulation (LOA) and percent-predicted forced vital capacity (FVC%p). Time to LOA was compared between eteplirsen-treated patients and standard of care (SOC) external controls and was measured from eteplirsen initiation in 201/202 or, in the SOC group, from the first study visit. Comparisons were conducted using univariate Kaplan-Meier analyses and log-rank tests, and multivariate Cox proportional hazards models with regression adjustment for baseline characteristics. Annual change in FVC%p was compared between eteplirsen-treated patients and natural history study patients using linear mixed models with repeated measures. Results: Data were included from all 12 patients in Studies 201/202 and the 10 patients with available data from 405. Median age at LOA was 15.16 years. Eteplirsen-treated patients experienced a statistically significant longer median time to LOA by 2.09 years (5.09 vs. 3.00 years, p < 0.01) and significantly attenuated rates of pulmonary decline vs. natural history patients (FVC%p change: –3.3 vs. –6.0 percentage points annually, p < 0.0001). Conclusions: Study 405 highlights the functional benefits of eteplirsen on ambulatory and pulmonary function outcomes up to 7 years of follow-up in comparison to external controls.
Collapse
Affiliation(s)
| | | | | | - Anne M Connolly
- Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA
| | | | - Crystal Proud
- Children's Hospital of The King's Daughters, Norfolk, VA, USA
| | - Perry B Shieh
- University of California Los Angeles, Los Angeles, CA, USA
| | - Kathryn R Wagner
- Kennedy Krieger Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | | | | | | | - Craig M McDonald
- University of California Davis Health System, Sacramento, CA, USA
| | | | | | | | - Jerry R Mendell
- Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
159
|
Houwen-van Opstal SLS, van den Elzen YMEM, Jansen M, Willemsen MAAP, Cup EHC, De Groot IJM. Facilitators and Barriers to Wearing Hand Orthoses by Adults with Duchenne Muscular Dystrophy: A Mixed Methods Study Design. J Neuromuscul Dis 2021; 7:467-475. [PMID: 32568104 PMCID: PMC7592678 DOI: 10.3233/jnd-200506] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Background: To retard shortening of finger flexors in patients with Duchenne muscular dystrophy (DMD), hand orthoses are prescribed. However, many patients do not wear the orthoses regularly. To optimize orthotic interventions, we need insight into the factors influencing compliance. Objective: To evaluate the compliance regarding hand orthoses in an adult DMD population and to explore experiences and perceptions of DMD patients wearing orthoses, and of their caregivers. Methods: Mixed methods observational study, combining quantitative and qualitative data from medical charts combined with qualitative semi-structured interviews using a constant comparative method and a short validated questionnaire (D-QUEST). Results: 65 medical charts were analyzed. 48 patients were assessed as needing hand orthoses, of whom 37.5 % were compliant. Qualitative data analyses revealed (1) motivation: preservation of hand function; (2) barriers: discomfort and impediments; (3) facilitators: good fit and personalized wearing schedule; (4) fitting process: satisfactory, but patients do not readily seek help when barriers appear. Conclusions: Patients are motivated to wear hand orthoses, but often discontinue use because of orthosis-and disease-specific barriers. The identification of these barriers leads to practical and feasible recommendations concerning the orthoses and the fitting process, such as less rigid material, preservation of some function while wearing the orthoses, and fixed evaluation points. The findings were confirmed by the D-QUEST.
Collapse
Affiliation(s)
- S L S Houwen-van Opstal
- Department of Rehabilitation, Radboud University Medical Center, Nijmegen, The Netherlands.,Amalia Children Hospital, Radboud University Medical Center, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Y M E M van den Elzen
- Department of Rehabilitation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M Jansen
- Department of Rehabilitation, Radboud University Medical Center, Nijmegen, The Netherlands.,Amalia Children Hospital, Radboud University Medical Center, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M A A P Willemsen
- Amalia Children Hospital, Radboud University Medical Center, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| | - E H C Cup
- Department of Rehabilitation, Radboud University Medical Center, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| | - I J M De Groot
- Department of Rehabilitation, Radboud University Medical Center, Nijmegen, The Netherlands.,Amalia Children Hospital, Radboud University Medical Center, Nijmegen, The Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
160
|
Licandro SA, Crippa L, Pomarico R, Perego R, Fossati G, Leoni F, Steinkühler C. The pan HDAC inhibitor Givinostat improves muscle function and histological parameters in two Duchenne muscular dystrophy murine models expressing different haplotypes of the LTBP4 gene. Skelet Muscle 2021; 11:19. [PMID: 34294164 PMCID: PMC8296708 DOI: 10.1186/s13395-021-00273-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/27/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND In the search of genetic determinants of Duchenne muscular dystrophy (DMD) severity, LTBP4, a member of the latent TGF-β binding protein family, emerged as an important predictor of functional outcome trajectories in mice and humans. Nonsynonymous single-nucleotide polymorphisms in LTBP4 gene associate with prolonged ambulation in DMD patients, whereas an in-frame insertion polymorphism in the mouse LTBP4 locus modulates disease severity in mice by altering proteolytic stability of the Ltbp4 protein and release of transforming growth factor-β (TGF-β). Givinostat, a pan-histone deacetylase inhibitor currently in phase III clinical trials for DMD treatment, significantly reduces fibrosis in muscle tissue and promotes the increase of the cross-sectional area (CSA) of muscles in mdx mice. In this study, we investigated the activity of Givinostat in mdx and in D2.B10 mice, two mouse models expressing different Ltbp4 variants and developing mild or more severe disease as a function of Ltbp4 polymorphism. METHODS Givinostat and steroids were administrated for 15 weeks in both DMD murine models and their efficacy was evaluated by grip strength and run to exhaustion functional tests. Histological examinations of skeletal muscles were also performed to assess the percentage of fibrotic area and CSA increase. RESULTS Givinostat treatment increased maximal normalized strength to levels that were comparable to those of healthy mice in both DMD models. The effect of Givinostat in both grip strength and exhaustion tests was dose-dependent in both strains, and in D2.B10 mice, Givinostat outperformed steroids at its highest dose. The in vivo treatment with Givinostat was effective in improving muscle morphology in both mdx and D2.B10 mice by reducing fibrosis. CONCLUSION Our study provides evidence that Givinostat has a significant effect in ameliorating both muscle function and histological parameters in mdx and D2.B10 murine models suggesting a potential benefit also for patients with a poor prognosis LTBP4 genotype.
Collapse
Affiliation(s)
| | - Luca Crippa
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | | | | | | | - Flavio Leoni
- Preclinical Development, Italfarmaco S.p.A., Milan, Italy
| | | |
Collapse
|
161
|
Stücker R, Stücker S, Mladenov K. [Spinal deformity in Duchenne muscular dystrophy]. DER ORTHOPADE 2021; 50:638-642. [PMID: 34279679 DOI: 10.1007/s00132-021-04127-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 11/26/2022]
Abstract
Boys with Duchenne muscular dystrophy develop scoliosis in 95% of cases. Conservative management to avoid progression of the deformity is not successful. However, treatment with corticosteroids may avoid the onset of scoliosis in some patients. Early surgical treatment and spine fusion is usually recommended before severe cardiopulmonary symptoms have developed. With modern surgical techniques, the sitting ability and quality of life can be preserved with a moderate complication rate. An interdisciplinary approach is mandatory to achieve these goals.
Collapse
Affiliation(s)
- Ralf Stücker
- Kinderorthopädische Abteilung, Altonaer Kinderkrankenhaus, Bleickenallee 38, 22763, Hamburg, Deutschland.
- Klinik für Orthopädie, Universitätsklinik Hamburg-Eppendorf, Hamburg, Deutschland.
| | - Sebastian Stücker
- Kinderorthopädische Abteilung, Altonaer Kinderkrankenhaus, Bleickenallee 38, 22763, Hamburg, Deutschland
- Klinik für Orthopädie, Universitätsklinik Hamburg-Eppendorf, Hamburg, Deutschland
| | - Kiril Mladenov
- Kinderorthopädische Abteilung, Altonaer Kinderkrankenhaus, Bleickenallee 38, 22763, Hamburg, Deutschland
- Klinik für Orthopädie, Universitätsklinik Hamburg-Eppendorf, Hamburg, Deutschland
| |
Collapse
|
162
|
Thangarajh M, Bello L, Gordish-Dressman H. Longitudinal motor function in proximal versus distal DMD pathogenic variants. Muscle Nerve 2021; 64:467-473. [PMID: 34255858 DOI: 10.1002/mus.27371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 07/02/2021] [Accepted: 07/09/2021] [Indexed: 11/08/2022]
Abstract
INTRODUCTION/AIMS There is considerable heterogenicity in clinical outcomes in Duchenne muscular dystrophy (DMD). The aim of this study was to assess whether dystrophin gene (DMD) pathogenic variant location influences upper or lower extremity motor function outcomes in a large prospective cohort. METHODS We used longitudinal timed and quantitative motor function measurements obtained from 154 boys with DMD over a 10-y period by the Cooperative International Neuromuscular Research Group Duchenne Natural History Study (CINRG-DNHS) to understand how the trajectories of motor function differ based on proximal versus distal DMD pathogenic variants. Proximal variants were defined as located proximal to 5' DMD intron 44, and distal variants as those including nucleotides 3' DMD including intron 44. Distal DMD variants are predicted to alter the expression of short dystrophin isoforms (Dp140, Dp116, and Dp71). We compared various upper extremity and lower extremity motor function measures in these two groups, after adjusting for total lifetime corticosteroid use. RESULTS The time to loss-of-ambulation and timed motor function measurements of both upper and lower limbs over a 10-y period were comparable between boys with proximal (n = 53) and distal (n = 101) DMD pathogenic variants. Age had a significant effect on several motor function outcomes. Boys younger than 7 y of age (n = 49) showed gain in function whereas boys 7 y and older (n = 71) declined, regardless of dystrophin pathogenic variant location. DISCUSSION The longitudinal decline in upper and lower motor function is independent of proximal versus distal location of DMD pathogenic variants.
Collapse
Affiliation(s)
- Mathula Thangarajh
- Department of Neurology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Luca Bello
- Department of Neurosciences DNS, University of Padova, Padova, Italy
| | - Heather Gordish-Dressman
- Center for Genetic Medicine, Children's Research Institute, Children's National Health System, Washington, District of Columbia, USA
| | | |
Collapse
|
163
|
The Interplay of Mitophagy and Inflammation in Duchenne Muscular Dystrophy. Life (Basel) 2021; 11:life11070648. [PMID: 34357020 PMCID: PMC8307817 DOI: 10.3390/life11070648] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked neuromuscular disease caused by a pathogenic disruption of the DYSTROPHIN gene that results in non-functional dystrophin protein. DMD patients experience loss of ambulation, cardiac arrhythmia, metabolic syndrome, and respiratory failure. At the molecular level, the lack of dystrophin in the muscle results in myofiber death, fibrotic infiltration, and mitochondrial dysfunction. There is no cure for DMD, although dystrophin-replacement gene therapies and exon-skipping approaches are being pursued in clinical trials. Mitochondrial dysfunction is one of the first cellular changes seen in DMD myofibers, occurring prior to muscle disease onset and progresses with disease severity. This is seen by reduced mitochondrial function, abnormal mitochondrial morphology and impaired mitophagy (degradation of damaged mitochondria). Dysfunctional mitochondria release high levels of reactive oxygen species (ROS), which can activate pro-inflammatory pathways such as IL-1β and IL-6. Impaired mitophagy in DMD results in increased inflammation and further aggravates disease pathology, evidenced by increased muscle damage and increased fibrosis. This review will focus on the critical interplay between mitophagy and inflammation in Duchenne muscular dystrophy as a pathological mechanism, as well as describe both candidate and established therapeutic targets that regulate these pathways.
Collapse
|
164
|
Florczyk-Soluch U, Polak K, Dulak J. The multifaceted view of heart problem in Duchenne muscular dystrophy. Cell Mol Life Sci 2021; 78:5447-5468. [PMID: 34091693 PMCID: PMC8257522 DOI: 10.1007/s00018-021-03862-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/29/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022]
Abstract
Dystrophin is a large protein serving as local scaffolding repetitively bridging cytoskeleton and the outside of striated muscle cell. As such dystrophin is a critical brick primarily in dystrophin-associated protein complex (DAGC) and in a larger submembranous unit, costamere. Accordingly, the lack of functional dystrophin laying at the root of Duchenne muscular dystrophy (DMD) drives sarcolemma instability. From this point on, the cascade inevitably leading to the death of myocyte begins. In cardiomyocytes, intracellular calcium overload and related mitochondrial-mediated cell death mainly contribute to myocardial dysfunction and dilation while other protein dysregulation and/or mislocalization may affect electrical conduction system and favor arrhythmogenesis. Although clinically DMD manifests as progressive muscle weakness and skeletal muscle symptoms define characteristic of DMD, it is the heart problem the biggest challenge that most often develop in the form of dilated cardiomyopathy (DCM). Current standards of treatment and recent progress in respiratory care, introduced in most settings in the 1990s, have improved quality of life and median life expectancy to 4th decade of patient's age. At the same time, cardiac causes of death related to DMD increases. Despite preventive and palliative cardiac treatments available, the prognoses remain poor. Direct therapeutic targeting of dystrophin deficiency is critical, however, hindered by the large size of the dystrophin cDNA and/or stochastic, often extensive genetic changes in DMD gene. The correlation between cardiac involvement and mutations affecting specific dystrophin isoforms, may provide a mutation-specific cardiac management and novel therapeutic approaches for patients with CM. Nonetheless, the successful cardiac treatment poses a big challenge and may require combined therapy to combat dystrophin deficiency and its after-effects (critical in DMD pathogenesis). This review locates the multifaceted heart problem in the course of DMD, balancing the insights into basic science, translational efforts and clinical manifestation of dystrophic heart disease.
Collapse
Affiliation(s)
- Urszula Florczyk-Soluch
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| | - Katarzyna Polak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
165
|
Missiuna PC, Sarraj M, Bosakhar B, Thornley P, Donnellan J, Kishta W, Darby P, Maizlin ZV. Vertebroplasty for Treatment of Steroid-Induced Osteoporotic Vertebral Compression Fractures in an Adolescent With Duchenne Muscular Dystrophy. HSS J 2021; 17:223-226. [PMID: 34421434 PMCID: PMC8361581 DOI: 10.1177/1556331621990648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 12/03/2022]
Affiliation(s)
- Paul C. Missiuna
- Department of Surgery, Division of Orthopedic Surgery, McMaster University Medical Centre, Hamilton, ON, Canada,Paul C. Missiuna, MD, FRCS (C), Department of Surgery, Division of Orthopedic Surgery, McMaster University Medical Centre, Hamilton, ON L8N 3Z5, Canada.
| | - Mohamed Sarraj
- Department of Surgery, Division of Orthopedic Surgery, McMaster University Medical Centre, Hamilton, ON, Canada
| | - Batool Bosakhar
- Department of Surgery, Division of Orthopedic Surgery, McMaster University Medical Centre, Hamilton, ON, Canada
| | - Patrick Thornley
- Department of Surgery, Division of Orthopedic Surgery, McMaster University Medical Centre, Hamilton, ON, Canada
| | - John Donnellan
- Department of Radiology, McMaster University Medical Centre, Hamilton, ON, Canada
| | - Waleed Kishta
- Department of Surgery, Division of Orthopedic Surgery, McMaster University Medical Centre, Hamilton, ON, Canada
| | - Peter Darby
- Department of Anesthesia, McMaster University Medical Centre, Hamilton, ON, Canada
| | - Zeev V. Maizlin
- Department of Radiology, McMaster University Medical Centre, Hamilton, ON, Canada
| |
Collapse
|
166
|
Contreras O, Rossi FMV, Theret M. Origins, potency, and heterogeneity of skeletal muscle fibro-adipogenic progenitors-time for new definitions. Skelet Muscle 2021; 11:16. [PMID: 34210364 PMCID: PMC8247239 DOI: 10.1186/s13395-021-00265-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
Striated muscle is a highly plastic and regenerative organ that regulates body movement, temperature, and metabolism-all the functions needed for an individual's health and well-being. The muscle connective tissue's main components are the extracellular matrix and its resident stromal cells, which continuously reshape it in embryonic development, homeostasis, and regeneration. Fibro-adipogenic progenitors are enigmatic and transformative muscle-resident interstitial cells with mesenchymal stem/stromal cell properties. They act as cellular sentinels and physiological hubs for adult muscle homeostasis and regeneration by shaping the microenvironment by secreting a complex cocktail of extracellular matrix components, diffusible cytokines, ligands, and immune-modulatory factors. Fibro-adipogenic progenitors are the lineage precursors of specialized cells, including activated fibroblasts, adipocytes, and osteogenic cells after injury. Here, we discuss current research gaps, potential druggable developments, and outstanding questions about fibro-adipogenic progenitor origins, potency, and heterogeneity. Finally, we took advantage of recent advances in single-cell technologies combined with lineage tracing to unify the diversity of stromal fibro-adipogenic progenitors. Thus, this compelling review provides new cellular and molecular insights in comprehending the origins, definitions, markers, fate, and plasticity of murine and human fibro-adipogenic progenitors in muscle development, homeostasis, regeneration, and repair.
Collapse
Affiliation(s)
- Osvaldo Contreras
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia.
- St. Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Kensington, 2052, Australia.
- Departamento de Biología Celular y Molecular and Center for Aging and Regeneration (CARE-ChileUC), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.
| | - Fabio M V Rossi
- Biomedical Research Centre, Department of Medical Genetics and School of Biomedical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Marine Theret
- Biomedical Research Centre, Department of Medical Genetics and School of Biomedical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
167
|
Abstract
Duchenne muscular dystrophy (DMD) is a devastating, rare disease. While clinically described in the 19th century, the genetic foundation of DMD was not discovered until more than 100 years later. This genetic understanding opened the door to the development of genetic treatments for DMD. Over the course of the last 30 years, the research that supports this development has moved into the realm of clinical trials and regulatory drug approvals. Exon skipping to therapeutically restore the frame of an out-of-frame dystrophin mutation has taken center stage in drug development for DMD. The research reviewed here focuses on the clinical development of exon skipping for the treatment of DMD. In addition to the generation of clinical treatments that are being used for patient care, this research sets the stage for future therapeutic development with a focus on increasing efficacy while providing safety and addressing the multi-systemic aspects of DMD.
Collapse
Affiliation(s)
- Shin'ichi Takeda
- Honorary Director General, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Japan
| | - Paula R Clemens
- Professor and Vice Chair of VA Affairs, Department of Neurology, University of Pittsburgh School of Medicine, Division Chief, Neurology, Medical Service Line, VA Pittsburgh Healthcare System, Pittsburgh, PA USA
| | - Eric P Hoffman
- Professor, Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University - State University of New York, Binghamton, NY USA
| |
Collapse
|
168
|
Naarding KJ, Keene KR, Sardjoe Mishre ASD, Veeger TTJ, van de Velde NM, Prins AJ, Burakiewicz J, Verschuuren JJGM, van der Holst M, Niks EH, Kan HE. Preserved thenar muscles in non-ambulant Duchenne muscular dystrophy patients. J Cachexia Sarcopenia Muscle 2021; 12:694-703. [PMID: 33963807 PMCID: PMC8200430 DOI: 10.1002/jcsm.12711] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/10/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Clinical trials in Duchenne muscular dystrophy (DMD) focus primarily on ambulant patients. Results cannot be extrapolated to later disease stages due to a decline in targeted muscle tissue. In non-ambulant DMD patients, hand function is relatively preserved and crucial for daily-life activities. We used quantitative MRI (qMRI) to establish whether the thenar muscles could be valuable to monitor treatment effects in non-ambulant DMD patients. METHODS Seventeen non-ambulant DMD patients (range 10.2-24.1 years) and 13 healthy controls (range 9.5-25.4 years) underwent qMRI of the right hand at 3 T at baseline. Thenar fat fraction (FF), total volume (TV), and contractile volume (CV) were determined using 4-point Dixon, and T2water was determined using multiecho spin-echo. Clinical assessments at baseline (n = 17) and 12 months (n = 13) included pinch strength (kg), performance of the upper limb (PUL) 2.0, DMD upper limb patient reported outcome measure (PROM), and playing a video game for 10 min using a game controller. Group differences and correlations were assessed with non-parametric tests. RESULTS Total volume was lower in patients compared with healthy controls (6.9 cm3 , 5.3-9.0 cm3 vs. 13.0 cm3 , 7.6-15.8 cm3 , P = 0.010). CV was also lower in patients (6.3 cm3 , 4.6-8.3 cm3 vs. 11.9 cm3 , 6.9-14.6 cm3 , P = 0.010). FF was slightly elevated (9.7%, 7.3-11.4% vs. 7.7%, 6.6-8.4%, P = 0.043), while T2water was higher (31.5 ms, 30.0-32.6 ms vs. 28.1 ms, 27.8-29.4 ms, P < 0.001). Pinch strength and PUL decreased over 12 months (2.857 kg, 2.137-4.010 to 2.243 kg, 1.930-3.339 kg, and 29 points, 20-36 to 23 points, 17-30, both P < 0.001), while PROM did not (49 points, 36-57 to 44 points, 30-54, P = 0.041). All patients were able to play for 10 min at baseline or follow-up, but some did not comply with the study procedures regarding this endpoint. Pinch strength correlated with TV and CV in patients (rho = 0.72 and rho = 0.68) and controls (both rho = 0.89). PUL correlated with TV, CV, and T2water (rho = 0.57, rho = 0.51, and rho = -0.59). CONCLUSIONS Low thenar FF, increased T2water , correlation of muscle size with strength and function, and the decrease in strength and function over 1 year indicate that the thenar muscles are a valuable and quantifiable target for therapy in later stages of DMD. Further studies are needed to relate these data to the loss of a clinically meaningful milestone.
Collapse
Affiliation(s)
- Karin J Naarding
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Kevin R Keene
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands
| | | | - Thom T J Veeger
- C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands
| | - Nienke M van de Velde
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Arina J Prins
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Jedrzej Burakiewicz
- C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands
| | - Jan J G M Verschuuren
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Menno van der Holst
- Duchenne Center, Leiden, Netherlands.,Department of Orthopedics, Rehabilitation and Physiotherapy, Leiden University Medical Center, Leiden, Netherlands
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Duchenne Center, Leiden, Netherlands
| | - Hermien E Kan
- Duchenne Center, Leiden, Netherlands.,C.J. Gorter Center for High Field MRI, Department of Radiology, LUMC, Leiden, Netherlands
| |
Collapse
|
169
|
Early ambulatory Duchenne muscular dystrophy: First symptomatic stage of ambulation - A case report. INTERNATIONAL JOURNAL OF SURGERY OPEN 2021. [DOI: 10.1016/j.ijso.2021.100349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
170
|
Szabo SM, Salhany RM, Deighton A, Harwood M, Mah J, Gooch KL. The clinical course of Duchenne muscular dystrophy in the corticosteroid treatment era: a systematic literature review. Orphanet J Rare Dis 2021; 16:237. [PMID: 34022943 PMCID: PMC8141220 DOI: 10.1186/s13023-021-01862-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 05/10/2021] [Indexed: 12/19/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is a severe rare progressive inherited neuromuscular disorder, leading to loss of ambulation (LOA) and premature mortality. The standard of care for patients with DMD has been treatment with corticosteroids for the past decade; however a synthesis of contemporary data describing the clinical course of DMD is lacking. The objective was to summarize age at key clinical milestones (loss of ambulation, scoliosis, ventilation, cardiomyopathy, and mortality) in the corticosteroid-treatment-era. Methods A systematic review was conducted using MEDLINE and EMBASE. The percentage experiencing key clinical milestones, and the mean or median age at those milestones, was synthesized from studies from North American populations, published between 2007 and 2018. Results From 5637 abstracts, 29 studies were included. Estimates of the percentage experiencing key clinical milestones, and age at those milestones, showed heterogeneity. Up to 30% of patients lost ambulation by age 10 years, and up to 90% by 15 years of age. The mean age at scoliosis onset was approximately 14 years. Ventilatory support began from 15 to 18 years, and up to half of patients required ventilation by 20 years of age. Registry-based estimates suggest that 70% had evidence of cardiomyopathy by 15 years and almost all by 20 years of age. Finally, mortality rates up to 16% by age 20 years were reported; among those surviving to adulthood mortality was up to 60% by age 30 years. Conclusions Contemporary natural history studies from North America report that LOA on average occurs in the early teens, need for ventilation and cardiomyopathy in the late teens, and death in the third or fourth decade of life. Variability in rates may be due to differences in study design, treatment with corticosteroids or other disease-modifying agents, variations in clinical practices, and dystrophin mutations. Despite challenges in synthesizing estimates, these findings help characterize disease progression among contemporary North American DMD patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-01862-w.
Collapse
Affiliation(s)
- Shelagh M Szabo
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, V6A 1A4, Canada.
| | - Renna M Salhany
- Sarepta Therapeutics, 215 First St, Cambridge, MA, 02142, USA
| | - Alison Deighton
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, V6A 1A4, Canada
| | - Meagan Harwood
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, V6A 1A4, Canada
| | - Jean Mah
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | |
Collapse
|
171
|
Xiao T, Wu B, Cao Y, Liu R, Cheng G, Wang L, Zhuang D, Zhao Z, Wang H, Zhou W. Genetic identification of pathogenic variations of the DMD gene: a retrospective study from 10,481 neonatal patients based on next-generation sequencing data. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:766. [PMID: 34268379 PMCID: PMC8246177 DOI: 10.21037/atm-20-7102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 02/07/2021] [Indexed: 12/20/2022]
Abstract
Background An elevated level of creatine kinase (CK) is usually the primary screening marker for Duchenne muscular dystrophy (DMD)/Becker muscular dystrophy (BMD). This study investigated the clinical application of next-generation sequencing (NGS) in newborns with a possible diagnosis of DMD/BMD in the neonatal intensive care unit (NICU). Methods NGS data from the NICU between June 1, 2016, and June 30, 2020, were reanalyzed by an in-house pipeline. Other methods confirmed the genetic findings, and clinical follow-up was performed until August 1, 2020. Results Of the 10,481 newborns, 19 (0.18%, 19/10,481) cases with pathogenic variations of the DMD gene were identified, including 13 (68.4%, 13/19) deletions, 4 (21.1%, 4/19) duplications, and 2 (10.5%, 2/19) nonsense mutations. Eight of the cases were diagnosed with DMD. Therapeutic strategies were modified for these patients. Six cases were diagnosed with BMD. Five patients except for 1 deceased patient were further followed-up, and clinical management was adjusted based on the clinical symptoms. The remaining 5 cases were indeterminate for DMD and BMD. Genetic counseling and further follow-up were performed or suggested. Conclusions Our study showed that DMD/BMD could be diagnosed earlier in the neonatal stage before the typical clinical symptoms appear. Early diagnosis may provide an opportunity for guiding the care and treatment of patients. However, ethical issues need to be kept in mind in the process of genetic counseling.
Collapse
Affiliation(s)
- Tiantian Xiao
- Department of Neonates, Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China.,Department of Neonatology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Bingbing Wu
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Yun Cao
- Department of Neonates, Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Renchao Liu
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Guoqiang Cheng
- Department of Neonates, Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Laishuan Wang
- Department of Pediatrics, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Deyi Zhuang
- Department of Pediatrics, Xiamen Children's Hospital, Xiamen, China
| | - Zhengyan Zhao
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huijun Wang
- Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Wenhao Zhou
- Department of Neonates, Key Laboratory of Neonatal Diseases, Ministry of Health, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China.,Center for Molecular Medicine, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| |
Collapse
|
172
|
Zhang S, Qin D, Wu L, Li M, Song L, Wei C, Lu C, Zhang X, Hong S, Ma M, Wu S. Genotype characterization and delayed loss of ambulation by glucocorticoids in a large cohort of patients with Duchenne muscular dystrophy. Orphanet J Rare Dis 2021; 16:188. [PMID: 33910603 PMCID: PMC8082961 DOI: 10.1186/s13023-021-01837-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/20/2021] [Indexed: 02/05/2023] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is the most common genetic muscle disease in human. We aimed to describe the genotype distribution in a large cohort of Chinese DMD patients and their delayed loss of ambulation by glucocorticoid (GC) treatments. This is to facilitate protocol designs and outcome measures for the emerging DMD clinical trials. Results A total of 1163 patients with DMD were recruited and genotyped. Genotype variations were categorized as large deletions, large duplications, and small mutations. Large deletions were further analyzed for those amenable to exon-skipping therapies. Participants aged 5 years or older were grouped into GC-treated and GC-naïve groups. Clinical progression among different genotypes and their responses to GC treatments were measured by age at loss of ambulation (LOA). Among the mutation genotypes, large deletions, large duplications, and small mutations accounted for 68.79%, 7.14%, and 24.07%, respectively. The mean age at diagnosis was 4.59 years; the median ages at LOA for the GC-naïve, prednisone/prednisolone-treated, and deflazacort-treated groups were 10.23, 12.02, and 13.95 years, respectively. The “deletion amenable to skipping exon 44” subgroup and the nonsense-mutation subgroup had older ages at LOA than the “other deletions” subgroup. Subgroups were further analyzed by both genotypes and GC status. All genotypes showed significant beneficial responses to GC treatment. Deletions amenable to skipping exon 44 showed a lower hazard ratio (0.155). The mean age at death was 18.57 years in this DMD group. Conclusion Genotype variation influences clinical progression in certain DMD groups. Beneficial responses to GC treatment were observed among all DMD genotypes. Compared with other genotypes, deletions amenable to skipping exon 44 had a lower hazard ratio, which may indicate a stronger protective effect of GC treatments on this subgroup. These data are valuable for designing future clinical trials, as clinical outcomes may be influenced by the genotypes.
Collapse
Affiliation(s)
- Shu Zhang
- Department of Neurology, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.,Department of Neurology, Third Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Dongdong Qin
- Department of Physiology, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan Province, China
| | - Liwen Wu
- Department of Neurology, Hunan Children's Hospital, Changsha, 410008, Hunan Province, China
| | - Man Li
- Department of Neurology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi Province, China
| | - Lifang Song
- Department of Pediatric Neurology, Henan Children's Hospital, Zhengzhou, 450018, Henan Province, China
| | - Cuijie Wei
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Chunling Lu
- Department of Muscle Atrophy, Affiliated Yiling Hospital of Hebei Medical University, Shijiazhuang, 050091, Hebei Province, China
| | - Xiaoli Zhang
- Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Siqi Hong
- Department of Pediatrics, Chongqing Medical University Affiliated Children's Hospital, Chongqing, 400042, China
| | - Mingming Ma
- Department of Neurology, Affiliated People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan Province, China
| | - Shiwen Wu
- Department of Neurology, First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China. .,Department of Neurology, Third Medical Center of Chinese PLA General Hospital, Beijing, 100039, China. .,Department of Neurology, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | | |
Collapse
|
173
|
Lin D, Klein A, Cella D, Beutler A, Fang F, Magestro M, Cremer P, LeWinter MM, Luis SA, Abbate A, Ertel A, Litcher-Kelly L, Klooster B, Paolini JF. Health-related quality of life in patients with recurrent pericarditis: results from a phase 2 study of rilonacept. BMC Cardiovasc Disord 2021; 21:201. [PMID: 33882846 PMCID: PMC8061027 DOI: 10.1186/s12872-021-02008-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Impact of recurrent pericarditis (RP) on patient health-related quality of life (HRQoL) was evaluated through qualitative patient interviews and as an exploratory endpoint in a Phase 2 trial evaluating the efficacy and safety of rilonacept (IL-1α/IL-1β cytokine trap) to treat RP. METHODS Qualitative interviews were conducted with ten adults with RP to understand symptoms and HRQoL impacts, and the 10-item Patient-Reported Outcomes Measurement Information System Global Health (PROMIS GH) v1.2 was evaluated to determine questionnaire coverage of patient experience. The Phase 2 trial enrolled participants with active symptomatic RP (A-RP, n = 16) and corticosteroid-dependent participants with no active recurrence at baseline (CSD-RP, n = 9). All participants received rilonacept weekly during a 6-week base treatment period (TP) plus an optional 18-week extension period (EP). Tapering of concomitant medications, including corticosteroids (CS), was permitted during EP. HRQoL was assessed using the PROMIS GH, and patient-reported pain and blood levels of c-reactive protein (CRP) were collected at Baseline and follow-up periods. A secondary, descriptive analysis of the Phase 2 trial efficacy results was completed using HRQoL measures to characterize both the impact of RP and the treatment effect of rilonacept. RESULTS Information from qualitative interviews demonstrated that PROMIS GH concepts are relevant to adults with RP. From the Phase 2 trial, both participant groups showed impacted HRQoL at Baseline (mean PROMIS Global Physical Health [GPH] and Global Mental Health [GMH], were lower than population norm average). In A-RP, GPH/MPH improved by end of base TP and were sustained through EP (similar trends were observed for pain and CRP). Similarly, in CSD-RP, GPH/MPH improved by end of TP and further improved during EP, during CS tapering or discontinuation, without disease recurrence (low pain scores and CRP levels continued during the TP and EP). CONCLUSION This is the first study demonstrating impaired HRQoL in RP. Rilonacept treatment was associated with HRQoL improvements using PROMIS GH scores. Maintained/improved HRQoL during tapering/withdrawal of CS without recurrence suggests that rilonacept may provide an alternative to CS. TRIAL REGISTRATION ClinicalTrials.Gov; NCT03980522; 5 June 2019, retrospectively registered; https://clinicaltrials.gov/ct2/show/NCT03980522 .
Collapse
Affiliation(s)
- David Lin
- Abbott Northwestern's Heart Hospital, Minneapolis Heart Institute, 800 East 28th Street, 2nd Floor, Minneapolis, MN, 55407, USA.
| | | | | | - Anna Beutler
- Kiniksa Pharmaceuticals Corp., 100 Hayden Avenue, Lexington, MA, 02421, USA
| | - Fang Fang
- Kiniksa Pharmaceuticals Corp., 100 Hayden Avenue, Lexington, MA, 02421, USA
| | - Matt Magestro
- Kiniksa Pharmaceuticals Corp., 100 Hayden Avenue, Lexington, MA, 02421, USA
| | | | | | | | | | - Andrew Ertel
- Medstar Heart and Vascular Institute, Washington, DC, USA
| | | | | | - John F Paolini
- Kiniksa Pharmaceuticals Corp., 100 Hayden Avenue, Lexington, MA, 02421, USA
| |
Collapse
|
174
|
Levine H, Prais D, Aharoni S, Nevo Y, Katz J, Rahmani E, Goldberg L, Scheuerman O. COVID-19 in advanced Duchenne/Becker muscular dystrophy patients. Neuromuscul Disord 2021; 31:607-611. [PMID: 34053847 PMCID: PMC8021445 DOI: 10.1016/j.nmd.2021.03.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 12/24/2022]
Abstract
Duchenne muscular dystrophy (DMD) is the most common childhood muscular dystrophy. As a result of progressive muscle weakness, pulmonary function decreases during the second decade of life and lung disease contributes significantly to morbidity and mortality in these patients. Corticosteroids are the current standard of care for patients with DMD, despite known adverse effects such as obesity and immunosuppression. Over the past year (2020), the novel coronavirus (COVID-19/SARS-CoV2) outbreak has caused a global pandemic. Restrictive lung disease due to low lung volumes, chronic immunosuppressive treatment with corticosteroids, and obesity are potential risk factors that may contribute to a more severe course of the disease. Out of 116 Duchenne/Becker muscular dystrophy patients treated in our tertiary neuromuscular center, six patients with DMD and one with advanced Becker muscular dystrophy were found to be positive for COVID-19 infection. Two of the DMD patients were admitted for hospitalization, of whom one was dependent on daily nocturnal non-invasive ventilation. All patients recovered without complications despite obesity, steroid treatment and severe restrictive lung disease.
Collapse
Affiliation(s)
- Hagit Levine
- Pulmonary Institute, Schneider Children's Medical Center, Petah-Tikva 49100, Israel; Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel.
| | - Dario Prais
- Pulmonary Institute, Schneider Children's Medical Center, Petah-Tikva 49100, Israel; Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel
| | - Sharon Aharoni
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel; Neurology Institute, Schneider Children's Medical Center, Petah-Tikva, Israel
| | - Yoram Nevo
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel; Neurology Institute, Schneider Children's Medical Center, Petah-Tikva, Israel
| | - Julia Katz
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel
| | - Eyal Rahmani
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel; Department of Pediatrics B, Schneider Children's Medical Center, Petah-Tikva, Israel
| | - Lotem Goldberg
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel; Department of Pediatrics B, Schneider Children's Medical Center, Petah-Tikva, Israel
| | - Oded Scheuerman
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel; Department of Pediatrics B, Schneider Children's Medical Center, Petah-Tikva, Israel; Pediatric Infectious Disease Unit, Schneider Children's Medical Center
| |
Collapse
|
175
|
Cid-Díaz T, Leal-López S, Fernández-Barreiro F, González-Sánchez J, Santos-Zas I, Andrade-Bulos LJ, Rodríguez-Fuentes ME, Mosteiro CS, Mouly V, Casabiell X, Relova JL, Pazos Y, Camiña JP. Obestatin signalling counteracts glucocorticoid-induced skeletal muscle atrophy via NEDD4/KLF15 axis. J Cachexia Sarcopenia Muscle 2021; 12:493-505. [PMID: 33687156 PMCID: PMC8061369 DOI: 10.1002/jcsm.12677] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 12/16/2020] [Accepted: 12/30/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND A therapeutic approach for the treatment of glucocorticoid-induced skeletal muscle atrophy should be based on the knowledge of the molecular mechanisms determining the unbalance between anabolic and catabolic processes and how to re-establish this balance. Here, we investigated whether the obestatin/GPR39 system, an autocrine signalling system acting on myogenesis and with anabolic effects on the skeletal muscle, could protect against chronic glucocorticoid-induced muscle atrophy. METHODS In this study, we used an in vivo model of muscle atrophy induced by the synthetic glucocorticoid dexamethasone to examine the liaison molecules that define the interaction between the glucocorticoid receptor and the obestatin/GPR39 systems. The findings were extended to in vitro effects on human atrophy using human KM155C25 myotubes. RESULTS KLF15 and FoxO transcription factors were identified as direct targets of obestatin signalling in the control of proteostasis in skeletal muscle. The KLF15-triggered gene expression program, including atrogenes and FoxOs, was regulated via KLF15 ubiquitination by the E3 ubiquitin ligase NEDD4. Additionally, a specific pattern of FoxO post-translational modification, including FoxO4 phosphorylation by Akt pathway, was critical in the regulation of the ubiquitin-proteasome system. The functional cooperativity between Akt and NEDD4 in the regulation of FoxO and KLF15 provides integrated cues to counteract muscle proteostasis and re-establish protein synthesis. CONCLUSIONS The effective control of FoxO activity in response to glucocorticoid is critical to counteract muscle-related pathologies. These results highlight the potential of the obestatin/GPR39 system to fine-tune the effects of glucocorticoids on skeletal muscle wasting.
Collapse
Affiliation(s)
- Tania Cid-Díaz
- Laboratorio de Endocrinología Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Trav. Choupana s/n, Santiago de Compostela, Spain
| | - Saúl Leal-López
- Laboratorio de Patología Digestiva, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Trav. Choupana s/n, Santiago de Compostela, Spain
| | - Fátima Fernández-Barreiro
- Laboratorio de Endocrinología Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Trav. Choupana s/n, Santiago de Compostela, Spain
| | - Jessica González-Sánchez
- Laboratorio de Endocrinología Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Trav. Choupana s/n, Santiago de Compostela, Spain
| | - Icía Santos-Zas
- Paris Cardiovascular Research Center-PARCC, Université Paris Descartes, Sorbonne Paris Cité, INSERM UMRS 970, Paris, France
| | - Luis J Andrade-Bulos
- Laboratorio de Endocrinología Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Trav. Choupana s/n, Santiago de Compostela, Spain
| | - Manuel E Rodríguez-Fuentes
- Laboratorio de Endocrinología Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Trav. Choupana s/n, Santiago de Compostela, Spain
| | - Carlos S Mosteiro
- Laboratorio de Endocrinología Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Trav. Choupana s/n, Santiago de Compostela, Spain
| | - Vincent Mouly
- Center of Research in Myology, Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS 974, Paris, France
| | - Xesús Casabiell
- Departamento de Fisiología, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Jose Luis Relova
- Laboratorio de Endocrinología Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Trav. Choupana s/n, Santiago de Compostela, Spain.,Departamento de Fisiología, Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Yolanda Pazos
- Laboratorio de Patología Digestiva, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Trav. Choupana s/n, Santiago de Compostela, Spain
| | - Jesus P Camiña
- Laboratorio de Endocrinología Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Trav. Choupana s/n, Santiago de Compostela, Spain
| |
Collapse
|
176
|
Clemens PR, Rao VK, Connolly AM, Harper AD, Mah JK, Smith EC, McDonald CM, Zaidman CM, Morgenroth LP, Osaki H, Satou Y, Yamashita T, Hoffman EP. Safety, Tolerability, and Efficacy of Viltolarsen in Boys With Duchenne Muscular Dystrophy Amenable to Exon 53 Skipping: A Phase 2 Randomized Clinical Trial. JAMA Neurol 2021; 77:982-991. [PMID: 32453377 PMCID: PMC7251505 DOI: 10.1001/jamaneurol.2020.1264] [Citation(s) in RCA: 150] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Question What are the safety, tolerability, and efficacy of viltolarsen in boys with Duchenne muscular dystrophy (DMD) amenable to exon 53 skipping? Findings Results of this 4-week randomized clinical trial for safety followed by a 20-week open-label treatment period in 16 patients with DMD indicated significant drug-induced dystrophin production in both viltolarsen groups (40 mg/kg per week and 80 mg/kg week) after 20 to 24 weeks of treatment. Timed function tests provided supportive evidence of treatment-related clinical improvement, and viltolarsen was well tolerated. Meaning Viltolarsen may provide a new therapeutic option for patients with DMD amenable to exon 53 skipping. Importance An unmet need remains for safe and efficacious treatments for Duchenne muscular dystrophy (DMD). To date, there are limited agents available that address the underlying cause of the disease. Objective To evaluate the safety, tolerability, and efficacy of viltolarsen, a novel antisense oligonucleotide, in participants with DMD amenable to exon 53 skipping. Design, Setting, and Participants This phase 2 study was a 4-week randomized clinical trial for safety followed by a 20-week open-label treatment period of patients aged 4 to 9 years with DMD amenable to exon 53 skipping. To enroll 16 participants, with 8 participants in each of the 2 dose cohorts, 17 participants were screened. Study enrollment occurred between December 16, 2016, and August 17, 2017, at sites in the US and Canada. Data were collected from December 2016 to February 2018, and data were analyzed from April 2018 to May 2019. Interventions Participants received 40 mg/kg (low dose) or 80 mg/kg (high dose) of viltolarsen administered by weekly intravenous infusion. Main Outcomes and Measures Primary outcomes of the trial included safety, tolerability, and de novo dystrophin protein production measured by Western blot in participants’ biceps muscles. Secondary outcomes included additional assessments of dystrophin mRNA and protein production as well as clinical muscle strength and function. Results Of the 16 included boys with DMD, 15 (94%) were white, and the mean (SD) age was 7.4 (1.8) years. After 20 to 24 weeks of treatment, significant drug-induced dystrophin production was seen in both viltolarsen dose cohorts (40 mg/kg per week: mean [range] 5.7% [3.2-10.3] of normal; 80 mg/kg per week: mean [range] 5.9% [1.1-14.4] of normal). Viltolarsen was well tolerated; no treatment-emergent adverse events required dose reduction, interruption, or discontinuation of the study drug. No serious adverse events or deaths occurred during the study. Compared with 65 age-matched and treatment-matched natural history controls, all 16 participants treated with viltolarsen showed significant improvements in timed function tests from baseline, including time to stand from supine (viltolarsen: −0.19 s; control: 0.66 s), time to run/walk 10 m (viltolarsen: 0.23 m/s; control: −0.04 m/s), and 6-minute walk test (viltolarsen: 28.9 m; control: −65.3 m) at the week 25 visit. Conclusions and Relevance Systemic treatment of participants with DMD with viltolarsen induced de novo dystrophin production, and clinical improvement of timed function tests was observed. Trial Registration ClinicalTrials.gov Identifier: NCT02740972
Collapse
Affiliation(s)
- Paula R Clemens
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,Department of Veterans Affairs Medical Center, Pittsburgh, Pennsylvania
| | - Vamshi K Rao
- Division of Neurology, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Anne M Connolly
- Division of Neurology, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus
| | - Amy D Harper
- Children's Hospital of Richmond at Virginia Commonwealth University, Richmond
| | - Jean K Mah
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - Edward C Smith
- Division of Pediatric Neurology, Duke University Medical Center, Durham, North Carolina
| | - Craig M McDonald
- Department of Physical Medicine and Rehabilitation, Department of Pediatrics, UC Davis Health, University of California, Davis, Sacramento
| | - Craig M Zaidman
- Department of Neurology, Washington University at St Louis, St Louis, Missouri
| | - Lauren P Morgenroth
- Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | | | | | - Eric P Hoffman
- AGADA BioSciences, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Pharmaceutical Sciences, State University of New York at Binghamton
| | | |
Collapse
|
177
|
Wittekind SG, Villa CR. Cardiac medication management in Duchenne muscular dystrophy. Pediatr Pulmonol 2021; 56:747-752. [PMID: 33647187 DOI: 10.1002/ppul.25175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/24/2020] [Accepted: 10/30/2020] [Indexed: 12/21/2022]
Abstract
There have been significant improvements in the skeletal muscle and respiratory care for patients with Duchenne muscular dystrophy (DMD) over the last two decades. This has resulted in longer expected survival as many patients will live into their 20s and 30s. This timeline has resulted in a greater proportion of patients experiencing heart failure and cardiac-related mortality. Herein, we describe the current indications for medical therapy for patients with DMD.
Collapse
Affiliation(s)
- Samuel G Wittekind
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Chet R Villa
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
178
|
Mackenzie SJ, Nicolau S, Connolly AM, Mendell JR. Therapeutic Approaches for Duchenne Muscular Dystrophy: Old and New. Semin Pediatr Neurol 2021; 37:100877. [PMID: 33892842 DOI: 10.1016/j.spen.2021.100877] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/20/2020] [Accepted: 11/21/2020] [Indexed: 12/13/2022]
Abstract
Duchenne muscular dystrophy (DMD) is marked by pathogenic variants in the DMD gene, leading to reduced or absent dystrophin translation, muscle fiber destruction, loss of ambulation, cardiomyopathy, respiratory failure, and eventually death. Disease progression is slowed with use of prednisone or other corticosteroid agents. Gene replacement therapy, which is one of the focus points of this review, has emerged as the most promising potential treatment for DMD, though alternative RNA-based strategies have been employed for patients with specific pathogenic variants. While challenges remain, many of these novel therapeutic approaches hold promise for treating this devastating disease.
Collapse
Affiliation(s)
- Samuel J Mackenzie
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH; Department of Pediatrics and Neurology; The Ohio State University, Columbus, OH.
| | - Stefan Nicolau
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH
| | - Anne M Connolly
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH; Department of Pediatrics and Neurology; The Ohio State University, Columbus, OH
| | - Jerry R Mendell
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH; Department of Pediatrics and Neurology; The Ohio State University, Columbus, OH
| |
Collapse
|
179
|
Politano L. Read-through approach for stop mutations in Duchenne muscular dystrophy. An update. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2021; 40:43-50. [PMID: 33870095 PMCID: PMC8033424 DOI: 10.36185/2532-1900-041] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 01/12/2023]
Abstract
Dystrophinopathies are allelic conditions caused by deletions, duplications and point-mutations in the DMD gene, located on the X chromosome (Xp21.2). Mutations that prematurely interrupt the dystrophin protein synthesis lead to the most severe clinical form, Duchenne muscular Dystrophy, characterized by early involvement of muscle strength. There is no known cure for dystrophinopathies. In DMD, treatment with corticosteroids have changed the natural history and the progression of the disease, prolonging ambulation, and slowing the onset of respiratory and cardiac involvement and scoliosis by several years. In the last few years, new perspectives and options are deriving from the discovery of pharmacological approaches able to restore normal, full-length dystrophin and potentially reverse the course of the disease. Read-through (RT) of nonsense mutations, thanks to its ability to bypass the premature stop codon and to act on virtually any region of the dystrophin gene, independently of the location in which the mutation resides, is one of these promising approaches. This non-systematic review shows the different steps that, passing from yeast to humans, have made it possible to use this innovative successful approach to treat serious diseases such as Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Luisa Politano
- Cardiomiology and Medical Genetics, "Luigi Vanvitelli" University, Naples, Italy
| |
Collapse
|
180
|
Powell PA, Carlton J, Rowen D, Chandler F, Guglieri M, Brazier JE. Development of a New Quality of Life Measure for Duchenne Muscular Dystrophy Using Mixed Methods: The DMD-QoL. Neurology 2021; 96:e2438-e2450. [PMID: 33785551 PMCID: PMC8166440 DOI: 10.1212/wnl.0000000000011896] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/12/2021] [Indexed: 11/29/2022] Open
Abstract
Objective Based on concerns about existing patient-reported outcome measures (PROMs) for assessing quality of life (QoL) in Duchenne muscular dystrophy (DMD), we describe the mixed methods development of a new QoL PROM for use in boys and men with DMD: the DMD-QoL. Methods The DMD-QoL was developed in 3 stages. First, draft items were generated from 18 semistructured qualitative interviews with boys and men with DMD, analyzed using framework analysis. Second, cognitive debriefing interviews with patients (n = 10), clinicians (n = 8), and patients' parents (n = 10) were undertaken, and a reduced item set was selected and refined. Third, psychometric data on the draft items from a cross-sectional online survey (n = 102) and stakeholder input from patients and patients' parents were used to produce the final questionnaire. Patient and public involvement and engagement was embedded throughout the process. Results From an initial draft of 47 items, a revised set of 27 items was produced at stage 2, and this set was further refined at stage 3 to generate the DMD-QoL, a 14-item QoL PROM. The DMD-QoL is designed for use from 7 years of age by proxy report and from 10 years of age by self-report or proxy report. The final measure showed good psychometric properties. Conclusion The DMD-QoL is a new 14-item QoL PROM for boys and men with DMD, with demonstrable content and face validity.
Collapse
Affiliation(s)
- Philip A Powell
- From the School of Health and Related Research (P.A.P., J.C., D.R., J.E.B.), University of Sheffield; Duchenne UK (F.C.), London; and John Walton Dystrophy Research Centre (M.G.), Institute of Genetic Medicine, University of Newcastle and Newcastle Hospitals NHS Foundation Trust, UK.
| | - Jill Carlton
- From the School of Health and Related Research (P.A.P., J.C., D.R., J.E.B.), University of Sheffield; Duchenne UK (F.C.), London; and John Walton Dystrophy Research Centre (M.G.), Institute of Genetic Medicine, University of Newcastle and Newcastle Hospitals NHS Foundation Trust, UK
| | - Donna Rowen
- From the School of Health and Related Research (P.A.P., J.C., D.R., J.E.B.), University of Sheffield; Duchenne UK (F.C.), London; and John Walton Dystrophy Research Centre (M.G.), Institute of Genetic Medicine, University of Newcastle and Newcastle Hospitals NHS Foundation Trust, UK
| | - Fleur Chandler
- From the School of Health and Related Research (P.A.P., J.C., D.R., J.E.B.), University of Sheffield; Duchenne UK (F.C.), London; and John Walton Dystrophy Research Centre (M.G.), Institute of Genetic Medicine, University of Newcastle and Newcastle Hospitals NHS Foundation Trust, UK
| | - Michela Guglieri
- From the School of Health and Related Research (P.A.P., J.C., D.R., J.E.B.), University of Sheffield; Duchenne UK (F.C.), London; and John Walton Dystrophy Research Centre (M.G.), Institute of Genetic Medicine, University of Newcastle and Newcastle Hospitals NHS Foundation Trust, UK
| | - John E Brazier
- From the School of Health and Related Research (P.A.P., J.C., D.R., J.E.B.), University of Sheffield; Duchenne UK (F.C.), London; and John Walton Dystrophy Research Centre (M.G.), Institute of Genetic Medicine, University of Newcastle and Newcastle Hospitals NHS Foundation Trust, UK
| |
Collapse
|
181
|
Liu X, Chai Y, Liu G, Su W, Guo Q, Lv X, Gao P, Yu B, Ferbeyre G, Cao X, Wan M. Osteoclasts protect bone blood vessels against senescence through the angiogenin/plexin-B2 axis. Nat Commun 2021; 12:1832. [PMID: 33758201 PMCID: PMC7987975 DOI: 10.1038/s41467-021-22131-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 02/27/2021] [Indexed: 01/31/2023] Open
Abstract
Synthetic glucocorticoids (GCs), one of the most effective treatments for chronic inflammatory and autoimmune conditions in children, have adverse effects on the growing skeleton. GCs inhibit angiogenesis in growing bone, but the underlying mechanisms remain unclear. Here, we show that GC treatment in young mice induces vascular endothelial cell senescence in metaphysis of long bone, and that inhibition of endothelial cell senescence improves GC-impaired bone angiogenesis with coupled osteogenesis. We identify angiogenin (ANG), a ribonuclease with pro-angiogenic activity, secreted by osteoclasts as a key factor for protecting the neighboring vascular cells against senescence. ANG maintains the proliferative activity of endothelial cells through plexin-B2 (PLXNB2)-mediated transcription of ribosomal RNA (rRNA). GC treatment inhibits ANG production by suppressing osteoclast formation in metaphysis, resulting in impaired endothelial cell rRNA transcription and subsequent cellular senescence. These findings reveal the role of metaphyseal blood vessel senescence in mediating the action of GCs on growing skeleton and establish the ANG/PLXNB2 axis as a molecular basis for the osteoclast-vascular interplay in skeletal angiogenesis.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Bone Development/drug effects
- Cell Proliferation/drug effects
- Cellular Senescence/drug effects
- Cellular Senescence/genetics
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Glucocorticoids/pharmacology
- Human Umbilical Vein Endothelial Cells
- Humans
- Immunohistochemistry
- In Situ Hybridization, Fluorescence
- Methylprednisolone/pharmacology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Neovascularization, Pathologic
- Neovascularization, Physiologic/drug effects
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Osteoclasts/drug effects
- Osteoclasts/enzymology
- Osteoclasts/metabolism
- Osteogenesis/drug effects
- RNA, Ribosomal/biosynthesis
- RNA, Small Interfering
- Recombinant Proteins
- Ribonuclease, Pancreatic/genetics
- Ribonuclease, Pancreatic/metabolism
- Ribonuclease, Pancreatic/pharmacology
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Tomography Scanners, X-Ray Computed
Collapse
Affiliation(s)
- Xiaonan Liu
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yu Chai
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guanqiao Liu
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Weiping Su
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qiaoyue Guo
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiao Lv
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peisong Gao
- Johns Hopkins Asthma & Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bin Yu
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Gerardo Ferbeyre
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC, Canada
| | - Xu Cao
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mei Wan
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
182
|
Zhang T, Kong X. Recent advances of glucocorticoids in the treatment of Duchenne muscular dystrophy (Review). Exp Ther Med 2021; 21:447. [PMID: 33777191 PMCID: PMC7967797 DOI: 10.3892/etm.2021.9875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common degenerative neuromuscular disease. The incidence of DMD in live births is 1/3,600-1/6,000. Although glucocorticoid-dependent medication is the mainstay treatment option for DMD, a standard treatment regimen has yet to be determined. The present review discusses the literature on the timing, methods and courses of glucocorticoid treatment for DMD. The review highlights the importance of the immediate commencement of glucocorticoid treatment following the diagnosis of DMD, with weekend-only administration being advantageous. Adherence to long-term single-glucocorticoid therapy can delay the loss of ambulation ability, and the side effects of the treatment are controllable. However, the standard medication for patients of different ages and stages of disease development, and the use of combination therapy require further investigation.
Collapse
Affiliation(s)
- Tianyuan Zhang
- Center of Prenatal Diagnosis, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xiangdong Kong
- Center of Prenatal Diagnosis, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
183
|
Andreana I, Repellin M, Carton F, Kryza D, Briançon S, Chazaud B, Mounier R, Arpicco S, Malatesta M, Stella B, Lollo G. Nanomedicine for Gene Delivery and Drug Repurposing in the Treatment of Muscular Dystrophies. Pharmaceutics 2021; 13:278. [PMID: 33669654 PMCID: PMC7922331 DOI: 10.3390/pharmaceutics13020278] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/07/2021] [Accepted: 02/14/2021] [Indexed: 12/11/2022] Open
Abstract
Muscular Dystrophies (MDs) are a group of rare inherited genetic muscular pathologies encompassing a variety of clinical phenotypes, gene mutations and mechanisms of disease. MDs undergo progressive skeletal muscle degeneration causing severe health problems that lead to poor life quality, disability and premature death. There are no available therapies to counteract the causes of these diseases and conventional treatments are administered only to mitigate symptoms. Recent understanding on the pathogenetic mechanisms allowed the development of novel therapeutic strategies based on gene therapy, genome editing CRISPR/Cas9 and drug repurposing approaches. Despite the therapeutic potential of these treatments, once the actives are administered, their instability, susceptibility to degradation and toxicity limit their applications. In this frame, the design of delivery strategies based on nanomedicines holds great promise for MD treatments. This review focuses on nanomedicine approaches able to encapsulate therapeutic agents such as small chemical molecules and oligonucleotides to target the most common MDs such as Duchenne Muscular Dystrophy and the Myotonic Dystrophies. The challenge related to in vitro and in vivo testing of nanosystems in appropriate animal models is also addressed. Finally, the most promising nanomedicine-based strategies are highlighted and a critical view in future developments of nanomedicine for neuromuscular diseases is provided.
Collapse
Affiliation(s)
- Ilaria Andreana
- Laboratoire d’Automatique, de Génie des Procédés et de Génie Pharmaceutique, Université Claude Bernard Lyon 1, CNRS UMR 5007, 43 bd 11 Novembre 1918, 69622 Villeurbanne, France; (I.A.); (M.R.); (D.K.); (S.B.)
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy;
| | - Mathieu Repellin
- Laboratoire d’Automatique, de Génie des Procédés et de Génie Pharmaceutique, Université Claude Bernard Lyon 1, CNRS UMR 5007, 43 bd 11 Novembre 1918, 69622 Villeurbanne, France; (I.A.); (M.R.); (D.K.); (S.B.)
- Department of Neurosciences, Biomedicine and Movement Sciences, Anatomy and Histology Section, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (F.C.); (M.M.)
| | - Flavia Carton
- Department of Neurosciences, Biomedicine and Movement Sciences, Anatomy and Histology Section, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (F.C.); (M.M.)
- Department of Health Sciences, University of Eastern Piedmont, Via Solaroli 17, 28100 Novara, Italy
| | - David Kryza
- Laboratoire d’Automatique, de Génie des Procédés et de Génie Pharmaceutique, Université Claude Bernard Lyon 1, CNRS UMR 5007, 43 bd 11 Novembre 1918, 69622 Villeurbanne, France; (I.A.); (M.R.); (D.K.); (S.B.)
- Hospices Civils de Lyon, 69437 Lyon, France
| | - Stéphanie Briançon
- Laboratoire d’Automatique, de Génie des Procédés et de Génie Pharmaceutique, Université Claude Bernard Lyon 1, CNRS UMR 5007, 43 bd 11 Novembre 1918, 69622 Villeurbanne, France; (I.A.); (M.R.); (D.K.); (S.B.)
| | - Bénédicte Chazaud
- Institut NeuroMyoGène, University of Lyon, INSERM U1217, CNRS UMR 5310, 8 Avenue Rockefeller, 69008 Lyon, France; (B.C.); (R.M.)
| | - Rémi Mounier
- Institut NeuroMyoGène, University of Lyon, INSERM U1217, CNRS UMR 5310, 8 Avenue Rockefeller, 69008 Lyon, France; (B.C.); (R.M.)
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy;
| | - Manuela Malatesta
- Department of Neurosciences, Biomedicine and Movement Sciences, Anatomy and Histology Section, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (F.C.); (M.M.)
| | - Barbara Stella
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Torino, Italy;
| | - Giovanna Lollo
- Laboratoire d’Automatique, de Génie des Procédés et de Génie Pharmaceutique, Université Claude Bernard Lyon 1, CNRS UMR 5007, 43 bd 11 Novembre 1918, 69622 Villeurbanne, France; (I.A.); (M.R.); (D.K.); (S.B.)
| |
Collapse
|
184
|
Abstract
Duchenne muscular dystrophy is a severe, progressive, muscle-wasting disease that leads to difficulties with movement and, eventually, to the need for assisted ventilation and premature death. The disease is caused by mutations in DMD (encoding dystrophin) that abolish the production of dystrophin in muscle. Muscles without dystrophin are more sensitive to damage, resulting in progressive loss of muscle tissue and function, in addition to cardiomyopathy. Recent studies have greatly deepened our understanding of the primary and secondary pathogenetic mechanisms. Guidelines for the multidisciplinary care for Duchenne muscular dystrophy that address obtaining a genetic diagnosis and managing the various aspects of the disease have been established. In addition, a number of therapies that aim to restore the missing dystrophin protein or address secondary pathology have received regulatory approval and many others are in clinical development.
Collapse
Affiliation(s)
- Dongsheng Duan
- Department of Molecular Microbiology and Immunology and Department of Neurology, School of Medicine; Department of Biomedical Sciences, College of Veterinary Medicine; Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO, USA
| | - Nathalie Goemans
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | | | - Eugenio Mercuri
- Centro Clinico Nemo, Policlinico Gemelli, Rome, Italy
- Peadiatric Neurology, Catholic University, Rome, Italy
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
185
|
Biressi S, Filareto A, Rando TA. Stem cell therapy for muscular dystrophies. J Clin Invest 2021; 130:5652-5664. [PMID: 32946430 DOI: 10.1172/jci142031] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Muscular dystrophies are a heterogeneous group of genetic diseases, characterized by progressive degeneration of skeletal and cardiac muscle. Despite the intense investigation of different therapeutic options, a definitive treatment has not been developed for this debilitating class of pathologies. Cell-based therapies in muscular dystrophies have been pursued experimentally for the last three decades. Several cell types with different characteristics and tissues of origin, including myogenic stem and progenitor cells, stromal cells, and pluripotent stem cells, have been investigated over the years and have recently entered in the clinical arena with mixed results. In this Review, we do a roundup of the past attempts and describe the updated status of cell-based therapies aimed at counteracting the skeletal and cardiac myopathy present in dystrophic patients. We present current challenges, summarize recent progress, and make recommendations for future research and clinical trials.
Collapse
Affiliation(s)
- Stefano Biressi
- Department of Cellular, Computational and Integrative Biology (CIBIO) and.,Dulbecco Telethon Institute, University of Trento, Povo, Italy
| | - Antonio Filareto
- Department of Research Beyond Borders, Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Conneticut, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences and.,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, California, USA.,Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
186
|
Zheng WB, Dai Y, Hu J, Zhao DC, Wang O, Jiang Y, Xia WB, Xing XP, Li M. Effects of Bisphosphonates on Osteoporosis Induced by Duchenne Muscular Dystrophy: A Prospective Study. Endocr Pract 2021; 26:1477-1485. [PMID: 33471740 DOI: 10.4158/ep-2020-0073] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 07/24/2020] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Duchenne muscular dystrophy (DMD) is a severe X-linked progressive neuromuscular disease that brings a significantly increased risk of osteoporosis and bone fractures. We prospectively evaluated the effects of oral and intravenous bisphosphonates on the bones of children with DMD. METHODS This study included a total of 52 children with DMD. They were divided into zoledronic acid (ZOL), alendronate (ALN), and control groups according to bone mineral density (BMD) and history of fragility fractures. For 2 years, all patients took calcium, vitamin D, and calcitriol. Meanwhile, 17 patients received infusions of ZOL, and 18 patients received ALN. BMD, serum levels of alkaline phosphatase (ALP) and the cross-linked C-telopeptide of type I collagen (β-CTX) were evaluated. RESULTS After 24 months of treatment, the percentage changes in lumbar spine BMD were 23.2 ± 9.7% and 23.6 ± 8.8% in the ZOL and ALN groups (all P<.01 vs. baseline). The increases did not differ between the ZOL and ALN groups, but were significantly larger than those of the control group (P<.01). Serum β-CTX and ALP levels, respectively, were decreased by 44.4 ± 18.0% and 31.9 ± 26.7% in the ZOL group and by 36.0 ± 20.3% and 25.8 ± 14.4% in the ALN group (all P<.01 vs. baseline). CONCLUSION Zoledronic acid and alendronate had similar protective effects to increase bone mineral density and reduce bone resorption in children with DMD, which were superior to treatment of calcium, vitamin D, and calcitriol. ABBREVIATIONS 25OHD = 25 hydroxyvitamin D; ALN = alendro-nate; ALP = alkaline phosphatase; ALT = alanine aminotransferase; BMD = bone mineral density; BP = bisphosphonate; Ca = calcium; β-CTX = cross-linked C-telopeptide of type I collagen; DMD = Duchenne muscular dystrophy; FN = femoral neck; GC = glucocorticoid; LS = lumbar spine; ZOL = zoledronic acid.
Collapse
Affiliation(s)
- Wen-Bin Zheng
- From the (1)Department of Endocrinology, National Health Commission Key Laboratory of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China, and the
| | - Yi Dai
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Hu
- From the (1)Department of Endocrinology, National Health Commission Key Laboratory of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China, and the
| | - Di-Chen Zhao
- From the (1)Department of Endocrinology, National Health Commission Key Laboratory of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China, and the
| | - Ou Wang
- From the (1)Department of Endocrinology, National Health Commission Key Laboratory of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China, and the
| | - Yan Jiang
- From the (1)Department of Endocrinology, National Health Commission Key Laboratory of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China, and the
| | - Wei-Bo Xia
- From the (1)Department of Endocrinology, National Health Commission Key Laboratory of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China, and the
| | - Xiao-Ping Xing
- From the (1)Department of Endocrinology, National Health Commission Key Laboratory of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China, and the
| | - Mei Li
- From the (1)Department of Endocrinology, National Health Commission Key Laboratory of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China, and the.
| |
Collapse
|
187
|
Merlini L, Cecconi I, Parmeggiani A, Cordelli DM, Dormi A. Quadriceps muscle strength in Duchenne muscular dystrophy and effect of corticosteroid treatment. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2021; 39:200-206. [PMID: 33458575 PMCID: PMC7783426 DOI: 10.36185/2532-1900-023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 11/12/2022]
Abstract
Objectives In Duchenne muscular dystrophy, quadriceps weakness is recognized as a key factor in gait deterioration. The objective of this work was three-fold: first, to document the strength of the quadriceps in corticosteroid-naïve DMD boys; second, to measure the effect of corticosteroids on quadriceps strength; and third, to evaluate the correlation between baseline quadriceps strength and the age when starting corticosteroids with the loss of ambulation. Methods Quadriceps muscle strength using hand-held dynamometry was measured in 12 ambulant DMD boys who had never taken corticosteroids and during corticosteroid treatment until the loss of ambulation. Results Baseline quadriceps muscle strength at 6 years of age was 28% that of normal children of the same age; it decreased to 15% at 8 years and to 6% at 10 years. The increase in quadriceps muscle strength obtained after 1 year of corticosteroid treatment had a strong direct correlation with the baseline strength (R = 0.96). With corticosteroid treatment, the age of ambulation loss showed a very strong direct relationship (R = 0.92) with baseline quadriceps muscle strength but only a very weak inverse relationship (R = -0.73) with the age of starting treatment. Age of loss of ambulation was 10.3 ± 0.5 vs 19.1 ± 4.7 (P < 0.05) in children with baseline quadriceps muscle strength less than or greater than 40 N, respectively. Conclusions Corticosteroid-naïve DMD boys have a quantifiable severe progressive quadriceps weakness. This long-term study, for the first time, shows that both of the positive effects obtained with CS treatment, i.e. increasing quadriceps strength and delaying the loss of ambulation, have a strong and direct correlation with baseline quadriceps muscle strength. As such, hand-held dynamometry may be a useful tool in the routine physical examination and during clinical trial assessment.
Collapse
Affiliation(s)
- Luciano Merlini
- Department of Biomedical and Neuromotor Sciences DIBINEM, University of Bologna, Italy
| | - Ilaria Cecconi
- Child Neurology and Psychiatry Unit, S. Orsola Hospital, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Italy
| | - Antonia Parmeggiani
- Child Neurology and Psychiatry Unit, S. Orsola Hospital, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Italy
| | - Duccio Maria Cordelli
- Child Neurology and Psychiatry Unit, S. Orsola Hospital, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Italy
| | - Ada Dormi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Italy
| |
Collapse
|
188
|
Agboola F, Lin GA, Fluetsch N, Walton SM, Rind DM, Pearson SD. The Effectiveness and Value of Deflazacort and Exon-Skipping Therapies for the Management of Duchenne Muscular Dystrophy. J Manag Care Spec Pharm 2021; 26:361-366. [PMID: 32223597 PMCID: PMC10391264 DOI: 10.18553/jmcp.2020.26.4.361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
DISCLOSURES Funding for this summary was contributed by Arnold Ventures, Commonwealth Fund, California Health Care Foundation, National Institute for Health Care Management (NIHCM), New England States Consortium Systems Organization, Blue Cross Blue Shield of Massachusetts, Harvard Pilgrim Health Care, Kaiser Foundation Health Plan, and Partners HealthCare to the Institute for Clinical and Economic Review (ICER), an independent organization that evaluates the evidence on the value of health care interventions. ICER's annual policy summit is supported by dues from Aetna, America's Health Insurance Plans, Anthem, Allergan, Alnylam, AstraZeneca, Biogen, Blue Shield of CA, Cambia Health Services, CVS, Editas, Express Scripts, Genentech/Roche, GlaxoSmithKline, Harvard Pilgrim, Health Care Service Corporation, Health Partners, Johnson & Johnson (Janssen), Kaiser Permanente, LEO Pharma, Mallinckrodt, Merck, Novartis, National Pharmaceutical Council, Premera, Prime Therapeutics, Regeneron, Sanofi, Spark Therapeutics, and United Healthcare. Agboola, Fluetsch, Rind, and Pearson are employed by ICER. Lin reports support from ICER during work on this economic model and grants from Mount Zion Health Fund, National Institutes of Health (National Cancer Institute and National Heart, Lung, and Blood Institute), and the Tobacco-Related Diseases Research Program, unrelated to this work. Walton reports support from ICER for work on this economic model and unrelated consulting fees from Baxter.
Collapse
Affiliation(s)
- Foluso Agboola
- Institute for Clinical and Economic Review, Boston, Massachusetts
| | - Grace A Lin
- Department of Medicine and Philip R Lee Institute for Health Policy Studies, University of California, San Francisco
| | - Noemi Fluetsch
- Institute for Clinical and Economic Review, Boston, Massachusetts
| | - Surrey M Walton
- Center for Pharmacoepidemiology and Pharmacoeconomic Research, University of Illinois at Chicago
| | - David M Rind
- Institute for Clinical and Economic Review, Boston, Massachusetts
| | - Steven D Pearson
- Institute for Clinical and Economic Review, Boston, Massachusetts
| |
Collapse
|
189
|
Doorenweerd N, de Rover M, Marini-Bettolo C, Hollingsworth KG, Niks EH, Hendriksen JGM, Kan HE, Straub V. Resting-state functional MRI shows altered default-mode network functional connectivity in Duchenne muscular dystrophy patients. Brain Imaging Behav 2021; 15:2297-2307. [PMID: 33389442 PMCID: PMC8500880 DOI: 10.1007/s11682-020-00422-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 07/28/2020] [Accepted: 11/24/2020] [Indexed: 11/29/2022]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive neuromuscular disorder caused by absence of dystrophin protein. Dystrophin is expressed in muscle, but also in the brain. Difficulties with attention/inhibition, working memory and information processing are well described in DMD patients but their origin is poorly understood. The default mode network (DMN) is one of the networks involved in these processes. Therefore we aimed to assess DMN connectivity in DMD patients compared to matched controls, to better understand the cognitive profile in DMD. T1-weighted and resting state functional MRI scans were acquired from 33 DMD and 24 male age-matched controls at two clinical sites. Scans were analysed using FMRIB Software Library (FSL). Differences in the DMN were assessed using FSL RANDOMISE, with age as covariate and threshold-free cluster enhancement including multiple comparison correction. Post-hoc analyses were performed on the visual network, executive control network and fronto-parietal network with the same methods. In DMD patients, the level of connectivity was higher in areas within the control DMN (hyperconnectivity) and significant connectivity was found in areas outside the control DMN. No hypoconnectivity was found and no differences in the visual network, executive control network and fronto-parietal network. We showed differences both within and in areas outside the DMN in DMD. The specificity of our findings to the DMN can help provide a better understanding of the attention/inhibition, working memory and information processing difficulties in DMD.
Collapse
Affiliation(s)
- Nathalie Doorenweerd
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK. .,C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, C-03-Q, P.O. Box 9600, 2300, RC, Leiden, The Netherlands.
| | - Mischa de Rover
- Department of Anesthesiology, Leiden University Medical Center, Leiden, The Netherlands.,Clinical Psychology Unit, Institute of Psychology, Leiden University, Leiden, The Netherlands
| | - Chiara Marini-Bettolo
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Kieren G Hollingsworth
- Newcastle Magnetic Resonance Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Erik H Niks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.,
| | - Jos G M Hendriksen
- .,Department of Neurological Learning Disabilities, Kempenhaeghe Epilepsy Center, Heeze, The Netherlands.,Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Hermien E Kan
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, C-03-Q, P.O. Box 9600, 2300, RC, Leiden, The Netherlands.,
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| |
Collapse
|
190
|
Alghamdi F, Al-Tawari A, Alrohaif H, Alshuaibi W, Mansour H, Aartsma-Rus A, Mégarbané A. Case Report: The Genetic Diagnosis of Duchenne Muscular Dystrophy in the Middle East. Front Pediatr 2021; 9:716424. [PMID: 34595143 PMCID: PMC8476401 DOI: 10.3389/fped.2021.716424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/17/2021] [Indexed: 11/28/2022] Open
Abstract
The timely and accurate genetic diagnosis of Duchenne muscular dystrophy (DMD) enables prompt initiation of disease management and genetic counseling and optimal patient care. Despite the existence of best practice guidelines for the diagnosis of DMD, implementation of these recommendations in different parts of the world is challenging. Here, we present 4 unique case studies which illustrate the different diagnostic pathways of patients with DMD in Middle Eastern countries and highlight region-specific challenges to achieving timely and accurate genetic diagnosis of DMD. A lack of disease awareness and consequential failure to recognize the signs and symptoms of DMD significantly contributed to the delayed diagnoses of these patients. Additional challenges included limited available funding for genetic testing and a lack of local specialist and genetic testing centers, causing patients and their families to travel vast distances for appointments in some countries. Earlier and more accurate genetic diagnosis of DMD in this region would allow patients to benefit from effective disease management, leading to improvements in health-related quality of life.
Collapse
Affiliation(s)
- Fouad Alghamdi
- Neuroscience Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Asmaa Al-Tawari
- Pediatric Neurology Unit, Pediatric Department, Al-Sabah Hospital, Kuwait City, Kuwait
| | - Hadil Alrohaif
- Kuwait Medical Genetics Centre, Al-Sabah Hospital, Kuwait City, Kuwait
| | - Walaa Alshuaibi
- College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Medical Genetics Division, Pediatrics Department, King Saud University, Riyadh, Saudi Arabia
| | - Hicham Mansour
- Pediatric Department, Saint George Hospital, Balamand University, Beirut, Lebanon
| | | | - André Mégarbané
- Department of Human Genetics, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| |
Collapse
|
191
|
Duong T, Canbek J, Birkmeier M, Nelson L, Siener C, Fernandez-Fernandez A, Henricson E, McDonald CM, Gordish-Dressman H. The Minimal Clinical Important Difference (MCID) in Annual Rate of Change of Timed Function Tests in Boys with DMD. J Neuromuscul Dis 2021; 8:939-948. [PMID: 34151852 PMCID: PMC8673528 DOI: 10.3233/jnd-210646] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a rare x-linked recessive genetic disorder affecting 1 in every 5000-10000 [1, 2]. This disease leads to a variable but progressive sequential pattern of muscle weakness that eventually causes loss of important functional milestones such as the ability to walk. With promising drugs in development to ameliorate the effects of muscle weakness, these treatments must be associated with a clinically meaningful functional change. OBJECTIVE The objective of this analysis is to determine both distribution, minimal detectable change (MDC), and anchor-based, minimal clinically important difference, (MCID) of 12 month change values in standardized time function tests (TFT) used to monitor disease progression in DMD. METHOD This is a retrospective analysis of prospectively collected data from a multi-center prospective natural history study with the Cooperative International Neuromuscular Research Group (CINRG). This study calculated MDC and MCID values for 3 commonly used timed function tests typically used to monitor disease progression; supine to stand (STS), 10 meter walk/run (10MWT), and 4 stair climb (4SC). MDC used standard error of measurement (SEM) while MCID measurements used the Vignos scale as an anchor to determine clinical change in functional status. RESULTS All 3 TFT were significantly important clinical endpoints to detect MDC and MCID changes. MDC and MCID 12 month changes were significant in 10MWT (-0.138, -0.212), Supine to Stand (-0.026, -0.023) and 4 stair climb (-0.034, -0.035) with an effect size greater or close to 0.2. CONCLUSION The 3 TFT are clinically meaningful endpoints used to establish change in DMD. MCID values were higher than MDC values indicating that an anchor-based approach using Vignos as a clinically meaningful loss of lower extremity abilities is appropriate to assess change in boys with DMD.
Collapse
Affiliation(s)
- Tina Duong
- Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer Canbek
- Physical Therapy Department, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Marisa Birkmeier
- Department of Health, Human Function, and Rehabilitation Sciences, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Leslie Nelson
- Department of Physical Therapy, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Catherine Siener
- Department of Neurology, Washington University, St. Louis, MO, USA
| | | | - Erik Henricson
- University of California Davis Health, Department of Physical Medicine and Rehabilitation, Sacramento CA, USA
| | - Craig M. McDonald
- University of California Davis Health, Department of Physical Medicine and Rehabilitation, Sacramento CA, USA
| | | | | |
Collapse
|
192
|
Quattrocelli M, Zelikovich AS, Salamone IM, Fischer JA, McNally EM. Mechanisms and Clinical Applications of Glucocorticoid Steroids in Muscular Dystrophy. J Neuromuscul Dis 2021; 8:39-52. [PMID: 33104035 PMCID: PMC7902991 DOI: 10.3233/jnd-200556] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Glucocorticoid steroids are widely used as immunomodulatory agents in acute and chronic conditions. Glucocorticoid steroids such as prednisone and deflazacort are recommended for treating Duchenne Muscular Dystrophy where their use prolongs ambulation and life expectancy. Despite this benefit, glucocorticoid use in Duchenne Muscular Dystrophy is also associated with significant adverse consequences including adrenal suppression, growth impairment, poor bone health and metabolic syndrome. For other forms of muscular dystrophy like the limb girdle dystrophies, glucocorticoids are not typically used. Here we review the experimental evidence supporting multiple mechanisms of glucocorticoid action in dystrophic muscle including their role in dampening inflammation and myofiber injury. We also discuss alternative dosing strategies as well as novel steroid agents that are in development and testing, with the goal to reduce adverse consequences of prolonged glucocorticoid exposure while maximizing beneficial outcomes.
Collapse
Affiliation(s)
- Mattia Quattrocelli
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Molecular Cardiovascular Biology Division, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Aaron S Zelikovich
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Isabella M Salamone
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Julie A Fischer
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
193
|
Novak JS, Spathis R, Dang UJ, Fiorillo AA, Hindupur R, Tully CB, Mázala DA, Canessa E, Brown KJ, Partridge TA, Hathout Y, Nagaraju K. Interrogation of Dystrophin and Dystroglycan Complex Protein Turnover After Exon Skipping Therapy. J Neuromuscul Dis 2021; 8:S383-S402. [PMID: 34569969 PMCID: PMC8673539 DOI: 10.3233/jnd-210696] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Recently, the Food and Drug Administration granted accelerated approvals for four exon skipping therapies -Eteplirsen, Golodirsen, Viltolarsen, and Casimersen -for Duchenne Muscular Dystrophy (DMD). However, these treatments have only demonstrated variable and largely sub-therapeutic levels of restored dystrophin protein in DMD patients, limiting their clinical impact. To better understand variable protein expression and the behavior of truncated dystrophin protein in vivo, we assessed turnover dynamics of restored dystrophin and dystrophin glycoprotein complex (DGC) proteins in mdx mice after exon skipping therapy, compared to those dynamics in wild type mice, using a targeted, highly-reproducible and sensitive, in vivo stable isotope labeling mass spectrometry approach in multiple muscle tissues. Through statistical modeling, we found that restored dystrophin protein exhibited altered stability and slower turnover in treated mdx muscle compared with that in wild type muscle (∼44 d vs. ∼24 d, respectively). Assessment of mRNA transcript stability (quantitative real-time PCR, droplet digital PCR) and dystrophin protein expression (capillary gel electrophoresis, immunofluorescence) support our dystrophin protein turnover measurements and modeling. Further, we assessed pathology-induced muscle fiber turnover through bromodeoxyuridine (BrdU) labeling to model dystrophin and DGC protein turnover in the context of persistent fiber degeneration. Our findings reveal sequestration of restored dystrophin protein after exon skipping therapy in mdx muscle leading to a significant extension of its half-life compared to the dynamics of full-length dystrophin in normal muscle. In contrast, DGC proteins show constant turnover attributable to myofiber degeneration and dysregulation of the extracellular matrix (ECM) in dystrophic muscle. Based on our results, we demonstrate the use of targeted mass spectrometry to evaluate the suitability and functionality of restored dystrophin isoforms in the context of disease and propose its use to optimize alternative gene correction strategies in development for DMD.
Collapse
Affiliation(s)
- James S. Novak
- Center for Genetic Medicine Research, Children’sResearch Institute, Children’s National Hospital, Washington, DC, USA
- Department of Genomics and PrecisionMedicine, The George Washington University School of Medicine and Health Sciences, Washington DC, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington DC, USA
- Correspondence to: James Novak, 111 Michigan Avenue NW, Washington, DC, 20010-2916 USA. Tel.: +1 202 476 6135; E-mail: . and Kanneboyina Nagaraju, PO Box 6000, Binghamton, NY, 13902-6000 USA. Tel.: +1 607 777 5814; E-mail:
| | - Rita Spathis
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY USA
| | - Utkarsh J. Dang
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY USA
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Alyson A. Fiorillo
- Center for Genetic Medicine Research, Children’sResearch Institute, Children’s National Hospital, Washington, DC, USA
- Department of Genomics and PrecisionMedicine, The George Washington University School of Medicine and Health Sciences, Washington DC, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington DC, USA
| | - Ravi Hindupur
- Center for Genetic Medicine Research, Children’sResearch Institute, Children’s National Hospital, Washington, DC, USA
| | - Christopher B. Tully
- Center for Genetic Medicine Research, Children’sResearch Institute, Children’s National Hospital, Washington, DC, USA
| | - Davi A.G. Mázala
- Center for Genetic Medicine Research, Children’sResearch Institute, Children’s National Hospital, Washington, DC, USA
- Department of Kinesiology, College of Health Professionals, Towson University, Towson, MD, USA
| | - Emily Canessa
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY USA
| | | | - Terence A. Partridge
- Center for Genetic Medicine Research, Children’sResearch Institute, Children’s National Hospital, Washington, DC, USA
| | - Yetrib Hathout
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY USA
| | - Kanneboyina Nagaraju
- Department of Genomics and PrecisionMedicine, The George Washington University School of Medicine and Health Sciences, Washington DC, USA
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY USA
- Correspondence to: James Novak, 111 Michigan Avenue NW, Washington, DC, 20010-2916 USA. Tel.: +1 202 476 6135; E-mail: . and Kanneboyina Nagaraju, PO Box 6000, Binghamton, NY, 13902-6000 USA. Tel.: +1 607 777 5814; E-mail:
| |
Collapse
|
194
|
Bronisz-Budzyńska I, Kozakowska M, Podkalicka P, Kachamakova-Trojanowska N, Łoboda A, Dulak J. The role of Nrf2 in acute and chronic muscle injury. Skelet Muscle 2020; 10:35. [PMID: 33287890 PMCID: PMC7722332 DOI: 10.1186/s13395-020-00255-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022] Open
Abstract
The nuclear factor erythroid 2-related factor 2 (Nrf2) is considered as a master cytoprotective factor regulating the expression of genes encoding anti-oxidant, anti-inflammatory, and detoxifying proteins. The role of Nrf2 in the pathophysiology of skeletal muscles has been evaluated in different experimental models, however, due to inconsistent data, we aimed to investigate how Nrf2 transcriptional deficiency (Nrf2tKO) affects muscle functions both in an acute and chronic injury. The acute muscle damage was induced in mice of two genotypes-WT and Nrf2tKO mice by cardiotoxin (CTX) injection. To investigate the role of Nrf2 in chronic muscle pathology, mdx mice that share genetic, biochemical, and histopathological features with Duchenne muscular dystrophy (DMD) were crossed with mice lacking transcriptionally active Nrf2 and double knockouts (mdx/Nrf2tKO) were generated. To worsen the dystrophic phenotype, the analysis of disease pathology was also performed in aggravated conditions, by applying a long-term treadmill test. We have observed slightly increased muscle damage in Nrf2tKO mice after CTX injection. Nevertheless, transcriptional ablation of Nrf2 in mdx mice did not significantly aggravate the most deleterious, pathological hallmarks of DMD related to degeneration, inflammation, fibrotic scar formation, angiogenesis, and the number and proliferation of satellite cells in non-exercised conditions. On the other hand, upon chronic exercises, the degeneration and inflammatory infiltration of the gastrocnemius muscle, but not the diaphragm, turned to be increased in Nrf2tKOmdx in comparison to mdx mice. In conclusion, the lack of transcriptionally active Nrf2 influences moderately muscle pathology in acute CTX-induced muscle injury and chronic DMD mouse model, without affecting muscle functionality. Hence, in general, we demonstrated that the deficiency of Nrf2 transcriptional activity has no profound impact on muscle pathology in various models of muscle injury.
Collapse
Affiliation(s)
- Iwona Bronisz-Budzyńska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Magdalena Kozakowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Paulina Podkalicka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | | | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Kraków, Poland
| |
Collapse
|
195
|
Kawamura K, Fukumura S, Nikaido K, Tachi N, Kozuka N, Seino T, Hatakeyama K, Mori M, Ito YM, Takami A, Hinotsu S, Kuno A, Kawasaki Y, Horio Y, Tsutsumi H. Resveratrol improves motor function in patients with muscular dystrophies: an open-label, single-arm, phase IIa study. Sci Rep 2020; 10:20585. [PMID: 33239684 PMCID: PMC7688653 DOI: 10.1038/s41598-020-77197-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
Muscular dystrophies (MDs) are inherited disorders characterized by progressive muscle weakness. Previously, we have shown that resveratrol (3,5,4′-trihydroxy-trans-stilbene), an antioxidant and an activator of the protein deacetylase SIRT1, decreases muscular and cardiac oxidative damage and improves pathophysiological conditions in animal MD models. To determine whether resveratrol provides therapeutic benefits to patients with MDs, an open-label, single-arm, phase IIa trial of resveratrol was conducted in 11 patients with Duchenne, Becker or Fukuyama MD. The daily dose of resveratrol was 500 mg/day, which was increased every 8 weeks to 1000 and then 1500 mg/day. Primary outcomes were motor function, evaluated by a motor function measure (MFM) scale, muscular strength, monitored with quantitative muscle testing (QMT), and serum creatine kinase (CK) levels. Adverse effects and tolerability were evaluated as secondary outcomes. Despite the advanced medical conditions of the patients, the mean MFM scores increased significantly from 34.6 to 38.4 after 24 weeks of medication. A twofold increase was found in the mean QMT scores of scapula elevation and shoulder abduction. Mean CK levels decreased considerably by 34%. Diarrhoea and abdominal pain was noted in six and three patients, respectively. Resveratrol may provide some benefit to MD patients.
Collapse
Affiliation(s)
- Kentaro Kawamura
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan
| | - Shinobu Fukumura
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan
| | - Koki Nikaido
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan
| | - Nobutada Tachi
- Faculty of Health Science, Hokkaido Chitose College of Rehabilitation, Chitose, 066-0055, Japan
| | - Naoki Kozuka
- Department of Physical Therapy, Sapporo Medical University School of Health Sciences, Sapporo, 060-8556, Japan
| | - Tsugumi Seino
- Department of Physical Therapy, Sapporo Medical University School of Health Sciences, Sapporo, 060-8556, Japan
| | - Kingya Hatakeyama
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan
| | - Mitsuru Mori
- Faculty of Health Science, Hokkaido Chitose College of Rehabilitation, Chitose, 066-0055, Japan
| | - Yoichi M Ito
- Biostatistics Division, Clinical Research and Medical Innovation Center, Hokkaido University Hospital, Sapporo, 060-8648, Japan
| | - Akiyoshi Takami
- Division of Hematology, Department of Internal Medicine, Aichi Medical University School of Medicine, Nagakute, 480-1195, Japan
| | - Shiro Hinotsu
- Department of Biostatistics, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Atsushi Kuno
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Yukihiko Kawasaki
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan
| | - Yoshiyuki Horio
- Department of Pharmacology, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan.
| | - Hiroyuki Tsutsumi
- Department of Pediatrics, Sapporo Medical University School of Medicine, Sapporo, 060-8543, Japan.
| |
Collapse
|
196
|
de Feraudy Y, Ben Yaou R, Wahbi K, Stalens C, Stantzou A, Laugel V, Desguerre I, Servais L, Leturcq F, Amthor H. Very Low Residual Dystrophin Quantity Is Associated with Milder Dystrophinopathy. Ann Neurol 2020; 89:280-292. [PMID: 33159473 PMCID: PMC7894170 DOI: 10.1002/ana.25951] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/16/2022]
Abstract
Objective This study was undertaken to determine whether a low residual quantity of dystrophin protein is associated with delayed clinical milestones in patients with DMD mutations. Methods We performed a retrospective multicentric cohort study by using molecular and clinical data from patients with DMD mutations registered in the Universal Mutation Database–DMD France database. Patients with intronic, splice site, or nonsense DMD mutations, with available muscle biopsy Western blot data, were included irrespective of whether they presented with severe Duchenne muscular dystrophy (DMD) or milder Becker muscular dystrophy (BMD). Patients were separated into 3 groups based on dystrophin protein levels. Clinical outcomes were ages at appearance of first symptoms; loss of ambulation; fall in vital capacity and left ventricular ejection fraction; interventions such as spinal fusion, tracheostomy, and noninvasive ventilation; and death. Results Of 3,880 patients with DMD mutations, 90 with mutations of interest were included. Forty‐two patients expressed no dystrophin (group A), and 31 of 42 (74%) developed DMD. Thirty‐four patients had dystrophin quantities < 5% (group B), and 21 of 34 (61%) developed BMD. Fourteen patients had dystrophin quantities ≥ 5% (group C), and all but 4 who lost ambulation beyond 24 years of age were ambulant. Dystrophin quantities of <5%, as low as <0.5%, were associated with milder phenotype for most of the evaluated clinical outcomes, including age at loss of ambulation (p < 0.001). Interpretation Very low residual dystrophin protein quantity can cause a shift in disease phenotype from DMD toward BMD. ANN NEUROL 2021;89:280–292
Collapse
Affiliation(s)
- Yvan de Feraudy
- Paris-Saclay University, UVSQ, Inserm, END-ICAP, Versailles, France.,Neuromuscular Reference Center, Pediatric Department, Raymond Poincaré Hospital, Garches, France
| | - Rabah Ben Yaou
- Neuromuscular Reference Center, Myology Institute, Pitié-Salpêtrière Hospital, Paris, France.,Center of Research in Myology, Sorbonne University, Inserm UMRS 974, Myology Institute, Pitié-Salpêtrière Hospital, Paris, France
| | - Karim Wahbi
- Cardiology Department, APHP, Cochin Hospital, FILNEMUS, Paris-Descartes, Sorbonne Paris Cité University, Paris, France
| | - Caroline Stalens
- Biostatistic, Medical Affairs Direction, AFM-Théléthon, Evry, France
| | - Amalia Stantzou
- Paris-Saclay University, UVSQ, Inserm, END-ICAP, Versailles, France
| | - Vincent Laugel
- Neuromuscular Reference Center, Pediatric Department, Hautepierre Hospital, Strasbourg, France
| | - Isabelle Desguerre
- Neuromuscular Reference Center, Pediatric Department, Necker-Enfants Malades Hospital, Paris, France
| | | | - Laurent Servais
- Department of Pediatrics, Neuromuscular Disease Reference Center, Division of Child Neurology, Faculty of Medicine, University of Liège, Liège, Belgium.,MDUK Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford, UK
| | - France Leturcq
- Laboratory for Biochemistry and Molecular Genetics, Cochin Hospital, Paris, France
| | - Helge Amthor
- Paris-Saclay University, UVSQ, Inserm, END-ICAP, Versailles, France.,Neuromuscular Reference Center, Pediatric Department, Raymond Poincaré Hospital, Garches, France
| |
Collapse
|
197
|
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
|
198
|
Dubinin MV, Talanov EY, Tenkov KS, Starinets VS, Belosludtseva NV, Belosludtsev KN. The Effect of Deflazacort Treatment on the Functioning of Skeletal Muscle Mitochondria in Duchenne Muscular Dystrophy. Int J Mol Sci 2020; 21:E8763. [PMID: 33228255 PMCID: PMC7699511 DOI: 10.3390/ijms21228763] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/17/2020] [Accepted: 11/17/2020] [Indexed: 01/10/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe hereditary disease caused by a lack of dystrophin, a protein essential for myocyte integrity. Mitochondrial dysfunction is reportedly responsible for DMD. This study examines the effect of glucocorticoid deflazacort on the functioning of the skeletal-muscle mitochondria of dystrophin-deficient mdx mice and WT animals. Deflazacort administration was found to improve mitochondrial respiration of mdx mice due to an increase in the level of ETC complexes (complexes III and IV and ATP synthase), which may contribute to the normalization of ATP levels in the skeletal muscle of mdx animals. Deflazacort treatment improved the rate of Ca2+ uniport in the skeletal muscle mitochondria of mdx mice, presumably by affecting the subunit composition of the calcium uniporter of organelles. At the same time, deflazacort was found to reduce the resistance of skeletal mitochondria to MPT pore opening, which may be associated with a change in the level of ANT2 and CypD. In this case, deflazacort also affected the mitochondria of WT mice. The paper discusses the mechanisms underlying the effect of deflazacort on the functioning of mitochondria and contributing to the improvement of the muscular function of mdx mice.
Collapse
MESH Headings
- Adenine Nucleotide Translocator 2/genetics
- Adenine Nucleotide Translocator 2/metabolism
- Adenosine Triphosphate/biosynthesis
- Animals
- Calcium/metabolism
- Calcium Channels/genetics
- Calcium Channels/metabolism
- Peptidyl-Prolyl Isomerase F/genetics
- Peptidyl-Prolyl Isomerase F/metabolism
- Electron Transport Complex III/genetics
- Electron Transport Complex III/metabolism
- Electron Transport Complex IV/genetics
- Electron Transport Complex IV/metabolism
- Gene Expression Regulation/drug effects
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Mitochondria, Muscle/drug effects
- Mitochondria, Muscle/genetics
- Mitochondria, Muscle/metabolism
- Mitochondrial Proton-Translocating ATPases/genetics
- Mitochondrial Proton-Translocating ATPases/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Pregnenediones/pharmacology
Collapse
Affiliation(s)
- Mikhail V. Dubinin
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia; (K.S.T.); (V.S.S.); (K.N.B.)
| | - Eugeny Yu. Talanov
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.Y.T.); (N.V.B.)
| | - Kirill S. Tenkov
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia; (K.S.T.); (V.S.S.); (K.N.B.)
| | - Vlada S. Starinets
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia; (K.S.T.); (V.S.S.); (K.N.B.)
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.Y.T.); (N.V.B.)
| | - Natalia V. Belosludtseva
- Laboratory of Mitochondrial Transport, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia; (E.Y.T.); (N.V.B.)
| | - Konstantin N. Belosludtsev
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia; (K.S.T.); (V.S.S.); (K.N.B.)
- Biophotonics Center, Prokhorov General Physics Institute, Russian Academy of Sciences, Vavilov st. 38, 119991 Moscow, Russia
| |
Collapse
|
199
|
Haber G, Conway KM, Paramsothy P, Roy A, Rogers H, Ling X, Kozauer N, Street N, Romitti PA, Fox DJ, Phan HC, Matthews D, Ciafaloni E, Oleszek J, James KA, Galindo M, Whitehead N, Johnson N, Butterfield RJ, Pandya S, Venkatesh S, Bhattaram VA. Association of genetic mutations and loss of ambulation in childhood-onset dystrophinopathy. Muscle Nerve 2020; 63:181-191. [PMID: 33150975 DOI: 10.1002/mus.27113] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 10/28/2020] [Accepted: 11/01/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND Quantifying associations between genetic mutations and loss of ambulation (LoA) among males diagnosed with childhood-onset dystrophinopathy is important for understanding variation in disease progression and may be useful in clinical trial design. METHODS Genetic and clinical data from the Muscular Dystrophy Surveillance, Tracking, and Research Network for 358 males born and diagnosed from 1982 to 2011 were analyzed. LoA was defined as the age at which independent ambulation ceased. Genetic mutations were defined by overall type (deletion/duplication/point mutation) and among deletions, those amenable to exon-skipping therapy (exons 8, 20, 44-46, 51-53) and another group. Cox proportional hazards regression modeling was used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). RESULTS Mutation type did not predict time to LoA. Controlling for corticosteroids, Exons 8 (HR = 0.22; 95% CI = 0.08, 0.63) and 44 (HR = 0.30; 95% CI = 0.12, 0.78) were associated with delayed LoA compared to other exon deletions. CONCLUSIONS Delayed LoA in males with mutations amenable to exon-skipping therapy is consistent with previous studies. These findings suggest that clinical trials including exon 8 and 44 skippable males should consider mutation information prior to randomization.
Collapse
Affiliation(s)
- Gregory Haber
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Kristin M Conway
- Department of Epidemiology, University of Iowa, Iowa City, Iowa, USA
| | - Pangaja Paramsothy
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Anindya Roy
- Department of Mathematics and Statistics, University of Maryland, Baltimore County, Baltimore, Maryland, USA
| | - Hobart Rogers
- Center for Drug Evaluation and Research, Food & Drug Administration, Silver Spring, Maryland, USA
| | - Xiang Ling
- Center for Drug Evaluation and Research, Food & Drug Administration, Silver Spring, Maryland, USA
| | - Nicholas Kozauer
- Center for Drug Evaluation and Research, Food & Drug Administration, Silver Spring, Maryland, USA
| | - Natalie Street
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Paul A Romitti
- Department of Epidemiology, University of Iowa, Iowa City, Iowa, USA
| | - Deborah J Fox
- Bureau of Environmental and Occupational Epidemiology, New York State Department of Health, Albany, New York, USA
| | - Han C Phan
- Department of Pediatrics, Division of Neurology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Dennis Matthews
- Department of Physical Medicine and Rehabilitation, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Emma Ciafaloni
- Department of Neurology, University of Rochester, Rochester, New York, USA
| | - Joyce Oleszek
- Department of Physical Medicine and Rehabilitation, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Katherine A James
- School of Public Health, University of Colorado, Boulder, Colorado, USA
| | - Maureen Galindo
- Department of Pediatrics, University of Arizona, Tucson, Arizona, USA
| | - Nedra Whitehead
- Research Triangle Institute International, Research Triangle Park, North Carolina, USA
| | - Nicholas Johnson
- Department of Neurology, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Russell J Butterfield
- Department of Pediatrics and Neurology, University of Utah, Salt Lake City, Utah, USA
| | - Shree Pandya
- Department of Neurology, University of Rochester, Rochester, New York, USA
| | - Swamy Venkatesh
- Department of Neurology, University of South Carolina, Columbia, South Carolina, USA
| | - Venkatesh Atul Bhattaram
- Center for Drug Evaluation and Research, Food & Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
200
|
Barnard AM, Riehl SL, Willcocks RJ, Walter GA, Angell AM, Vandenborne K. Characterizing Enrollment in Observational Studies of Duchenne Muscular Dystrophy by Race and Ethnicity. J Neuromuscul Dis 2020; 7:167-173. [PMID: 31929119 DOI: 10.3233/jnd-190447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Observational research benefits from inclusion of diverse cohorts. To characterize racial and ethnic diversity in observational and natural history research studies of Duchenne muscular dystrophy (DMD), highly cited and influential observational studies were identified. Fourteen United States-based articles were included. All studies cited >70% White participants with the majority having few racial minority participants. Enrollment of Black/African American individuals was particularly limited (<5% in all but one study), and Hispanic/Latino participants ranged from 3.3- 26.5% of cohorts. These results suggest a need for effective strategies to recruit, enroll, and retain racially and ethnically diverse populations into observational research in DMD.
Collapse
Affiliation(s)
- Alison M Barnard
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| | - Samuel L Riehl
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | | | - Glenn A Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| | - Amber M Angell
- Department of Occupational Therapy, University of Florida, Gainesville, FL, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| |
Collapse
|