1
|
K J, Santiago R. Quantitative Structure-Property Relationship Modeling with the Prediction of Physicochemical Properties of Some Novel Duchenne Muscular Dystrophy Drugs. ACS OMEGA 2025; 10:3640-3651. [PMID: 39926532 PMCID: PMC11800030 DOI: 10.1021/acsomega.4c08572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/25/2024] [Accepted: 12/18/2024] [Indexed: 02/11/2025]
Abstract
Duchenne muscular dystrophy is a critical, progressively worsening, and ultimately deadly illness characterized by the deterioration of skeletal muscles, respiratory failure, and heart disease. The pharmaceutical industries are persistently innovating drug design processes to address the rise of infections and effectively treat emerging syndromes or genetically based disorders with the help of quantitative structure-property relationship models. These models are mathematical tools that correlate molecular structures with their physicochemical properties through structural characteristics. Different models can be generated based on the various structural features of the compounds, and topological indices are one such significant structural feature generated from the molecular graph and are key tools used in these models. This study focuses on creating quantitative structure-property relationship models using degree-based topological indices, which are highly effective in quantitative structure-property relationship analysis to explore the diverse physicochemical properties of Duchenne muscular dystrophy drugs with the prediction of properties of a recently approved drug givinostat. Furthermore, the drug discovery and development activities can be accelerated using the developed models to forecast the possible productiveness of novel Duchenne muscular dystrophy treatment drugs.
Collapse
Affiliation(s)
- Jyothish K
- Department of Mathematics,
School of Advanced Sciences, Vellore Institute
of Technology, Vellore 632014, India
| | - Roy Santiago
- Department of Mathematics,
School of Advanced Sciences, Vellore Institute
of Technology, Vellore 632014, India
| |
Collapse
|
2
|
Matesanz SE, Edelson JB, Iacobellis KA, Mejia E, Brandsema JF, Wittlieb-Weber CA, Okunowo O, Griffis H, Lin KY. Subspecialty Health Care Utilization in Pediatric Patients With Muscular Dystrophy in the United States. Neurol Clin Pract 2024; 14:e200312. [PMID: 38855715 PMCID: PMC11160481 DOI: 10.1212/cpj.0000000000200312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/25/2024] [Indexed: 06/11/2024]
Abstract
Background and Objectives Standards of care exist to optimize outcomes in Duchenne and Becker muscular dystrophy (DBMD), caused by alterations in the DMD gene; however, there are limited data regarding health care access in these patients. This study aims to characterize outpatient subspecialty care utilization in pediatric patients with DBMD. Methods This retrospective cohort study used administrative claims data from IBM MarketScan Medicaid and Commercial Claims and Encounters Research Databases (2013-2018). Male patients 1-18 years with an ICD-9/10 diagnosis code for hereditary progressive muscular dystrophy between January 1, 2013, and December 31, 2017, were included. Participants were stratified into 3 age cohorts: 1-6 years, 7-12 years, and 13-18 years. The primary outcome was rate of annual neurology visits. Secondary outcomes included annual follow-up rates in other subspecialties and proportion of days covered (PDC) by corticosteroids. Results A total of 1,386 patients met inclusion-347 (25.0%) age 1-6 years, 502 (36.2%) age 7-12 years, and 537 (38.7%) age 13-18 years. Heart failure, respiratory failure, and technology dependence increased with age (p for all<0.05). The rate of neurology visits per person-year was 0.36 and did not differ by age. Corticosteroid use was low; 30% of person-years (1452/4829) had a PDC ≥20%. Medicaid insurance was independently associated with a lower likelihood of annual neurology follow-up (OR 0.23; 95% CI 0.18-0.28). Discussion The rate of annual neurology follow-up and corticosteroid use in patients with DBMD is low. Medicaid insurance status was independently associated with a decreased likelihood of neurology follow-up, while age was not, suggesting that factors other than disease severity influence neurology care access. Identifying barriers to regular follow-up is critical in improving outcomes for patients with DBMD.
Collapse
Affiliation(s)
- Susan E Matesanz
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - Jonathan B Edelson
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - Katherine A Iacobellis
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - Erika Mejia
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - John F Brandsema
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - Carol A Wittlieb-Weber
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - Oluwatimilehin Okunowo
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - Heather Griffis
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - Kimberly Y Lin
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| |
Collapse
|
3
|
Contesse MG, Hodges J, Staunton H, Lowes LP, Elmankabadi B, Elder SJ, Ormazabal MG, Dalle Pazze L, Leffler MG. Home-based video assessment of ease of movement for patients with Duchenne: Interviews with physical therapists to select movement tasks. PHYSIOTHERAPY RESEARCH INTERNATIONAL 2023; 28:e1993. [PMID: 36718112 DOI: 10.1002/pri.1993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 11/01/2022] [Accepted: 12/31/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND AND PURPOSE Patients with Duchenne muscular dystrophy (DMD) change their movement patterns to compensate for muscle weakness. The Duchenne Video Assessment (DVA) measures ease of movement through evaluation of compensatory movements. The DVA directs caregivers to video record patients performing home-based movement tasks using a mobile application, and DVA-certified physical therapists evaluate the videos using scorecards with prespecified compensatory movement criteria. Two qualitative interview studies were conducted to select movement tasks for the DVA that are relevant to patients with DMD and able to reflect changes in function. METHODS Qualitative interviews with eligible physical therapists were conducted remotely. Physical therapists were asked to spontaneously suggest movement tasks prior to evaluating specific movement tasks selected a priori. Analysts conducted a content analysis to investigate whether movement tasks selected a priori were confirmed as relevant to the population of interest and able to show changes in function. RESULTS The studies included five physical therapists to select tasks for ambulatory patients with DMD and six for non-ambulatory patients. For an ambulatory population, all five experts confirmed Climb Five Stairs, Run, Stand Up from Sitting, Sit Up from Supine, and Jump Forward, and four (80%) confirmed Walk as relevant and able to show functional changes. For a non-ambulatory population, all six experts confirmed Eat 10 Bites, Roll Over in Bed, Shift Weight in Bed, Take T-Shirt Off, Put T-Shirt On, Put Arms on Armrest, and Reach Across Table to Grab Cell Phone, and five (83%) confirmed Raise Hands Above Head as relevant and able to show functional changes. DISCUSSION Physical therapists confirmed the DVA movement tasks as relevant to patients with DMD and able to reflect changes in function. The use of the DVA in clinical trials provides an opportunity to collect data not seen in clinic and reduce travel burden for families.
Collapse
|
4
|
Inherited myopathies in the Middle East and North Africa. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101674] [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]
|
5
|
Hildyard JC, Riddell DO, Harron RC, Rawson F, Foster EM, Massey C, Taylor-Brown F, Wells DJ, Piercy RJ. The skeletal muscle phenotype of the DE50-MD dog model of Duchenne muscular dystrophy. Wellcome Open Res 2022; 7:238. [PMID: 36865375 PMCID: PMC9971692 DOI: 10.12688/wellcomeopenres.18251.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Animal models of Duchenne muscular dystrophy (DMD) are essential to study disease progression and assess efficacy of therapeutic intervention, however dystrophic mice fail to display a clinically relevant phenotype, limiting translational utility. Dystrophin-deficient dogs exhibit disease similar to humans, making them increasingly important for late-stage preclinical evaluation of candidate therapeutics. The DE50-MD canine model of DMD carries a mutation within a human 'hotspot' region of the dystrophin gene, amenable to exon-skipping and gene editing strategies. As part of a large natural history study of disease progression, we have characterised the DE50-MD skeletal muscle phenotype to identify parameters that could serve as efficacy biomarkers in future preclinical trials. Methods: Vastus lateralis muscles were biopsied from a large cohort of DE50-MD dogs and healthy male littermates at 3-monthly intervals (3-18 months) for longitudinal analysis, with multiple muscles collected post-mortem to evaluate body-wide changes. Pathology was characterised quantitatively using histology and measurement of gene expression to determine statistical power and sample sizes appropriate for future work. Results: DE50-MD skeletal muscle exhibits widespread degeneration/regeneration, fibrosis, atrophy and inflammation. Degenerative/inflammatory changes peak during the first year of life, while fibrotic remodelling appears more gradual. Pathology is similar in most skeletal muscles, but in the diaphragm, fibrosis is more prominent, associated with fibre splitting and pathological hypertrophy. Picrosirius red and acid phosphatase staining represent useful quantitative histological biomarkers for fibrosis and inflammation respectively, while qPCR can be used to measure regeneration ( MYH3, MYH8), fibrosis ( COL1A1), inflammation ( SPP1), and stability of DE50-MD dp427 transcripts. Conclusion: The DE50-MD dog is a valuable model of DMD, with pathological features similar to young, ambulant human patients. Sample size and power calculations show that our panel of muscle biomarkers are of strong pre-clinical value, able to detect therapeutic improvements of even 25%, using trials with only six animals per group.
Collapse
Affiliation(s)
- John C.W. Hildyard
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, London, UK
| | - Dominique O. Riddell
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, London, UK
| | - Rachel C.M. Harron
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, London, UK
| | - Faye Rawson
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, London, UK
- Langford Veterinary Services, University of Bristol, Langford, UK
| | - Emma M.A. Foster
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, London, UK
| | - Claire Massey
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, London, UK
| | - Frances Taylor-Brown
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, London, UK
- Cave Veterinary Specialists, George's Farm, West Buckland, UK
| | - Dominic J. Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, London, UK
| | - Richard J. Piercy
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London, London, UK
| |
Collapse
|
6
|
Development of Duchenne Video Assessment scorecards to evaluate ease of movement among those with Duchenne muscular dystrophy. PLoS One 2022; 17:e0266845. [PMID: 35417501 PMCID: PMC9007341 DOI: 10.1371/journal.pone.0266845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/29/2022] [Indexed: 12/02/2022] Open
Abstract
Background Patients with Duchenne muscular dystrophy (DMD) adopt compensatory movement patterns as muscles weaken. The Duchenne Video Assessment (DVA) measures patient ease of movement through identification of compensatory movement patterns. The DVA directs caregivers to video record patients performing specific movement tasks at home using a secure mobile application, and DVA-certified physical therapists (PTs) score the videos using scorecards with prespecified compensatory movement criteria. The goal of this study was to develop and refine the DVA scorecards. Methods To develop the initial scorecards, 4 PTs collaboratively created compensatory movement lists for each task, and researchers structured the lists into scorecards. A 2-round modified Delphi process was used to gather expert opinion on the understandability, comprehensiveness, and clinical meaningfulness of the compensatory movements on the scorecards. Eight PTs who had evaluated ≥50 patients with DMD and participated in ≥10 DMD clinical trials were recruited for the panel. In Round 1, panelists evaluated compensatory movement criteria understandability via questionnaire and tested the scorecards. In Round 2, panelists participated in an in-person meeting to discuss areas of disagreement from Round 1 and reach consensus (≥75% agreement) on all revisions to the scorecards. Results During the Round 1 revisions to the scorecards, there were 67 changes (44%) to the wording of 153 original compensatory movement criteria and 3 criteria were removed. During the Round 2 revisions to the scorecards, there were 47 changes (31%) to the wording of 150 compensatory movement criteria, 20 criteria were added, and 30 criteria were removed. The panel reached 100% agreement on all changes made to scorecards during Round 2. Conclusion PTs with extensive experience evaluating patients with DMD confirmed that the compensatory movement criteria included in the DVA scorecards were understandable, comprehensive, and clinically meaningful.
Collapse
|
7
|
Stefano MED, Ferretti V, Mozzetta C. Synaptic alterations as a neurodevelopmental trait of Duchenne muscular dystrophy. Neurobiol Dis 2022; 168:105718. [PMID: 35390481 DOI: 10.1016/j.nbd.2022.105718] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 01/14/2023] Open
Abstract
Dystrophinopaties, e.g., Duchenne muscular dystrophy (DMD), Becker muscular dystrophy and X-linked dilated cardiomyopathy are inherited neuromuscular diseases, characterized by progressive muscular degeneration, which however associate with a significant impact on general system physiology. The more severe is the pathology and its diversified manifestations, the heavier are its effects on organs, systems, and tissues other than muscles (skeletal, cardiac and smooth muscles). All dystrophinopaties are characterized by mutations in a single gene located on the X chromosome encoding dystrophin (Dp427) and its shorter isoforms, but DMD is the most devasting: muscular degenerations manifests within the first 4 years of life, progressively affecting motility and other muscular functions, and leads to a fatal outcome between the 20s and 40s. To date, after years of studies on both DMD patients and animal models of the disease, it has been clearly demonstrated that a significant percentage of DMD patients are also afflicted by cognitive, neurological, and autonomic disorders, of varying degree of severity. The anatomical correlates underlying neural functional damages are established during embryonic development and the early stages of postnatal life, when brain circuits, sensory and motor connections are still maturing. The impact of the absence of Dp427 on the development, differentiation, and consolidation of specific cerebral circuits (hippocampus, cerebellum, prefrontal cortex, amygdala) is significant, and amplified by the frequent lack of one or more of its lower molecular mass isoforms. The most relevant aspect, which characterizes DMD-associated neurological disorders, is based on morpho-functional alterations of selective synaptic connections within the affected brain areas. This pathological feature correlates neurological conditions of DMD to other severe neurological disorders, such as schizophrenia, epilepsy and autistic spectrum disorders, among others. This review discusses the organization and the role of the dystrophin-dystroglycan complex in muscles and neurons, focusing on the neurological aspect of DMD and on the most relevant morphological and functional synaptic alterations, in both central and autonomic nervous systems, described in the pathology and its animal models.
Collapse
Affiliation(s)
- Maria Egle De Stefano
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy; Center for Research in Neurobiology Daniel Bovet, Sapienza University of Rome, 00185 Rome, Italy.
| | - Valentina Ferretti
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy; Center for Research in Neurobiology Daniel Bovet, Sapienza University of Rome, 00185 Rome, Italy
| | - Chiara Mozzetta
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR) of Italy c/o Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
8
|
Blaschek A, Rodrigues M, Rawer R, Müller C, Ille L, Schröder S, Idriess M, Müller-Felber W, Vill K. Jumping Mechanography is a Suitable Complementary Method to Assess Motor Function in Ambulatory Boys with Duchenne Muscular Dystrophy. Neuropediatrics 2021; 52:455-461. [PMID: 33706402 DOI: 10.1055/s-0041-1722880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE The number of clinical trials for Duchenne muscular dystrophy (DMD) has increased substantially lately, therefore appropriate clinical instruments are needed to measure disease progression and drug efficacy. Jumping mechanography is a medical diagnostic method for motion analysis, which allows to quantify physical parameters. In this study, we compared mechanography with timed function tests (TFTs). METHODS 41 ambulatory DMD patients performed a total of 95 chair rising tests (CRT) and a total of 76 single two-legged jumps (S2LJ) on a mechanography ground reaction force platform. The results were correlated with a 6-minute walk test (6MWT) and the time required to run 10 meters, stand up from a supine position, and climb four stairs, all performed in the same setting. RESULTS Our measurements show a high correlation between mechanography and the TFTs: S2LJ/10-m run, r = 0.62; CRT/10-m run, r = 0.61; S2LJ/standing up from supine, r = 0.48; CRT/standing up from supine, r = 0.58; S2LJ/climb four stairs, r = 0.55; CRT/climb four stairs, r = 0.51. The correlation between mechanography and the 6MWT was only moderate with r = 0.38 for S2LJ/6MWT and r = 0.39 for CRT/6MWT. INTERPRETATION Jumping mechanography is a reliable additional method, which can be used for physical endpoint measurements in clinical trials. We confirmed our assumption, that the method provides additional information concerning performance at movement with higher power output. We suggest using the S2LJ as a first-choice tandem tool combined with the 6MWT. In patients with higher disability, the CRT is an alternative measuring method, because with the progression of the disease this is longer feasible.
Collapse
Affiliation(s)
- Astrid Blaschek
- Department of Paediatric Neurology and Developmental Medicine, LMU - University of Munich, Dr. v. Hauner Children's Hospital, Munich, Germany
| | - Martin Rodrigues
- Department of Paediatric Neurology and Developmental Medicine, LMU - University of Munich, Dr. v. Hauner Children's Hospital, Munich, Germany.,Department of Traumatology, Klinikum Starnberg, Starnberg, Germany
| | | | - Christine Müller
- Department of Paediatric Neurology and Developmental Medicine, LMU - University of Munich, Dr. v. Hauner Children's Hospital, Munich, Germany
| | - Lena Ille
- Department of Paediatric Neurology and Developmental Medicine, LMU - University of Munich, Dr. v. Hauner Children's Hospital, Munich, Germany.,Department for Gynaecology, Städtisches Klinikum München Neuperlach, Munich, Germany
| | - Sebastian Schröder
- Department of Paediatric Neurology and Developmental Medicine, LMU - University of Munich, Dr. v. Hauner Children's Hospital, Munich, Germany
| | - Mohamed Idriess
- Department of Paediatric Neurology and Developmental Medicine, LMU - University of Munich, Dr. v. Hauner Children's Hospital, Munich, Germany
| | - Wolfgang Müller-Felber
- Department of Paediatric Neurology and Developmental Medicine, LMU - University of Munich, Dr. v. Hauner Children's Hospital, Munich, Germany
| | - Katharina Vill
- Department of Paediatric Neurology and Developmental Medicine, LMU - University of Munich, Dr. v. Hauner Children's Hospital, Munich, Germany
| |
Collapse
|
9
|
Animal models for researching approaches to therapy of Duchenne muscular dystrophy. Transgenic Res 2021; 30:709-725. [PMID: 34409525 DOI: 10.1007/s11248-021-00278-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/11/2021] [Indexed: 01/17/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a relatively widespread genetic disease which develops as a result of a mutation in the gene DMD encoding dystrophin. In this review, animal models of DMD are described. These models are used in preclinical studies to elucidate the pathogenesis of the disease or to develop effective treatments; each animal model has its own advantages and disadvantages. For instance, Caenorhabditis elegans, Drosophila melanogaster, and zebrafish (sapje) are suitable for large-scale chemical screening of large numbers of small molecules, but their disease phenotype differs from that of mammals. The use of larger animals is important for understanding of the potential efficacy of various treatments for DMD. While mdx mice have their advantages, they exhibit a milder disease phenotype compared to humans or dogs, making it difficult to evaluate the efficacy of new treatment for DMD. The disease in dogs and pigs is more severe and progresses faster than in mice, but it is more difficult to breed and obtain sufficient numbers of specimens in order to achieve statistically significant results. Moreover, working with large animals is also more labor-intensive. Therefore, when choosing the optimal animal model for research, it is worth considering all the goals and objectives.
Collapse
|
10
|
Shastry A, Aravind S, Sunil M, Ramesh K, Ashley B, T. N, Ramprasad VL, Gupta R, Seshagiri S, Nongthomba U, Phalke S. Matrilineal analysis of mutations in the DMD gene in a multigenerational South Indian cohort using DMD gene panel sequencing. Mol Genet Genomic Med 2021; 9:e1633. [PMID: 33960727 PMCID: PMC8172192 DOI: 10.1002/mgg3.1633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/28/2021] [Accepted: 02/09/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is an X-linked recessive neuromuscular disorder characterised by progressive irreversible muscle weakness, primarily of the skeletal and the cardiac muscles. DMD is characterised by mutations in the dystrophin gene, resulting in the absence or sparse quantities of dystrophin protein. A precise and timely molecular detection of DMD mutations encourages interventions such as carrier genetic counselling and in undertaking therapeutic measures for the DMD patients. RESULTS In this study, we developed a 2.1 Mb custom DMD gene panel that spans the entire DMD gene, including the exons and introns. The panel also includes the probes against 80 additional genes known to be mutated in other muscular dystrophies. This custom DMD gene panel was used to identify single nucleotide variants (SNVs) and large deletions with precise breakpoints in 77 samples that included 24 DMD patients and their matrilineage across four generations. We used this panel to evaluate the inheritance pattern of DMD mutations in maternal subjects representing 24 DMD patients. CONCLUSION Here we report our observations on the inheritance pattern of DMD gene mutations in matrilineage samples across four generations. Additionally, our data suggest that the DMD gene panel designed by us can be routinely used as a single genetic test to identify all DMD gene variants in DMD patients and the carrier mothers.
Collapse
Affiliation(s)
- Arun Shastry
- Dystrophy Annihilation Research Trust (DART)BangaloreIndia
| | - Sankaramoorthy Aravind
- Dystrophy Annihilation Research Trust (DART)BangaloreIndia
- Indian Institute of Science (IISc)BangaloreIndia
| | | | - Keerthi Ramesh
- Dystrophy Annihilation Research Trust (DART)BangaloreIndia
| | - Berty Ashley
- Dystrophy Annihilation Research Trust (DART)BangaloreIndia
| | | | | | | | | | | | - Sameer Phalke
- MedGenome LabsBangaloreIndia
- SciGenom Labs Pvt LtdCochinIndia
| |
Collapse
|
11
|
Peay HL, Fischer R, Mange B, Paquin RS, Smith EC, Sadosky A, Russo L, Ricotti V, Rensch C, Morris C, Martin AS, Ganot A, Beaverson K, Mansfield C. Patients' and caregivers' maximum acceptable risk of death for non-curative gene therapy to treat Duchenne muscular dystrophy. Mol Genet Genomic Med 2021; 9:e1664. [PMID: 33755338 PMCID: PMC8172191 DOI: 10.1002/mgg3.1664] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Gene therapy offers an etiologically targeted treatment for genetic disorders. Little is known about the acceptance of mortality risk among patients with progressive, fatal conditions. We assessed patients' and caregivers' maximum acceptable risk (MAR) of mortality for gene therapy when used to treat Duchenne muscular dystrophy (DMD). METHODS The threshold technique was used to assess tolerance for mortality risks using a hypothetical vignette. Gene therapy was described as non-curative and resulting in a slowing of progression and with a 10-year benefit duration. MAR was analyzed using interval regression for gene therapy initiated "now"; in the last year of walking well; in the last year of being able to bring arms to mouth; and in newborns (for caregivers only). RESULTS Two hundred eighty-five caregivers and 35 patients reported the greatest MAR for gene therapy initiated in last year of being able to lift arms (mean MAR 6.3%), followed by last year of walking well (mean MAR 4.4%), when used "now" (mean MAR 3.5%), and when used in the newborn period (mean MAR 2.1%, caregivers only). About 35% would accept ≥200/2000 risk in the last year of being able to lift arms. Non-ambulatory status predicted accepting 1.8 additional points in MAR "now" compared with ambulatory status (p = 0.010). Respondent type (caregiver or patient) did not predict MAR. CONCLUSION In this first quantitative study to assess MAR associated with first-generation DMD gene therapy, we find relatively high tolerance for mortality risk in response to a non-curative treatment scenario. Risk tolerance increased with disease progression. Patients and caregivers did not have significantly different MAR. These results have implications for protocol development and shared decision making.
Collapse
Affiliation(s)
- Holly L Peay
- RTI International, Research Triangle Park, NC, USA
| | - Ryan Fischer
- Parent Project Muscular Dystrophy, Hackensack, NJ, USA
| | - Brennan Mange
- RTI Health Solutions, Research Triangle Park, NC, USA
| | | | | | | | | | | | - Colin Rensch
- Parent Project Muscular Dystrophy, Hackensack, NJ, USA
| | | | | | | | | | | |
Collapse
|
12
|
Muramatsu H, Kuramochi T, Katada H, Ueyama A, Ruike Y, Ohmine K, Shida-Kawazoe M, Miyano-Nishizawa R, Shimizu Y, Okuda M, Hori Y, Hayashi M, Haraya K, Ban N, Nonaka T, Honda M, Kitamura H, Hattori K, Kitazawa T, Igawa T, Kawabe Y, Nezu J. Novel myostatin-specific antibody enhances muscle strength in muscle disease models. Sci Rep 2021; 11:2160. [PMID: 33495503 PMCID: PMC7835227 DOI: 10.1038/s41598-021-81669-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/08/2021] [Indexed: 11/22/2022] Open
Abstract
Myostatin, a member of the transforming growth factor-β superfamily, is an attractive target for muscle disease therapy because of its role as a negative regulator of muscle growth and strength. Here, we describe a novel antibody therapeutic approach that maximizes the potential of myostatin-targeted therapy. We generated an antibody, GYM329, that specifically binds the latent form of myostatin and inhibits its activation. Additionally, via "sweeping antibody technology", GYM329 reduces or "sweeps" myostatin in the muscle and plasma. Compared with conventional anti-myostatin agents, GYM329 and its surrogate antibody exhibit superior muscle strength-improvement effects in three different mouse disease models. We also demonstrate that the superior efficacy of GYM329 is due to its myostatin specificity and sweeping capability. Furthermore, we show that a GYM329 surrogate increases muscle mass in normal cynomolgus monkeys without any obvious toxicity. Our findings indicate the potential of GYM329 to improve muscle strength in patients with muscular disorders.
Collapse
Affiliation(s)
- Hiroyasu Muramatsu
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Taichi Kuramochi
- Chugai Pharmabody Research Pte. Ltd., 3 Biopolis Drive, #07-11 to 16, Synapse, Singapore, 138623, Singapore
| | - Hitoshi Katada
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Atsunori Ueyama
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Yoshinao Ruike
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Ken Ohmine
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | | | | | - Yuichiro Shimizu
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Momoko Okuda
- Chugai Pharmabody Research Pte. Ltd., 3 Biopolis Drive, #07-11 to 16, Synapse, Singapore, 138623, Singapore
| | - Yuji Hori
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Madoka Hayashi
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Kenta Haraya
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Nobuhiro Ban
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Tatsuya Nonaka
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Masaki Honda
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Hidetomo Kitamura
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Kunihiro Hattori
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Takehisa Kitazawa
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Tomoyuki Igawa
- Chugai Pharmabody Research Pte. Ltd., 3 Biopolis Drive, #07-11 to 16, Synapse, Singapore, 138623, Singapore
| | - Yoshiki Kawabe
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan
| | - Junichi Nezu
- Research Division, Chugai Pharmaceutical Co., Ltd., Tokyo, 103-8324, Japan.
| |
Collapse
|
13
|
Cadilla R, Deaton DN, Do Y, Elkins PA, Ennulat D, Guss JH, Holt J, Jeune MR, King AG, Klapwijk JC, Kramer HF, Kramer NJ, Laffan SB, Masuria PI, McDougal AV, Mortenson PN, Musetti C, Peckham GE, Pietrak BL, Poole C, Price DJ, Rendina AR, Sati G, Saxty G, Shearer BG, Shewchuk LM, Sneddon HF, Stewart EL, Stuart JD, Thomas DN, Thomson SA, Ward P, Wilson JW, Xu T, Youngman MA. The exploration of aza-quinolines as hematopoietic prostaglandin D synthase (H-PGDS) inhibitors with low brain exposure. Bioorg Med Chem 2020; 28:115791. [PMID: 33059303 DOI: 10.1016/j.bmc.2020.115791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 12/15/2022]
Abstract
GlaxoSmithKline and Astex Pharmaceuticals recently disclosed the discovery of the potent H-PGDS inhibitor GSK2894631A 1a (IC50 = 9.9 nM) as part of a fragment-based drug discovery collaboration with Astex Pharmaceuticals. This molecule exhibited good murine pharmacokinetics, allowing it to be utilized to explore H-PGDS pharmacology in vivo. Yet, with prolonged dosing at higher concentrations, 1a induced CNS toxicity. Looking to attenuate brain penetration in this series, aza-quinolines, were prepared with the intent of increasing polar surface area. Nitrogen substitutions at the 6- and 8-positions of the quinoline were discovered to be tolerated by the enzyme. Subsequent structure activity studies in these aza-quinoline scaffolds led to the identification of 1,8-naphthyridine 1y (IC50 = 9.4 nM) as a potent peripherally restricted H-PGDS inhibitor. Compound 1y is efficacious in four in vivo inflammatory models and exhibits no CNS toxicity.
Collapse
Affiliation(s)
- Rodolfo Cadilla
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - David N Deaton
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA.
| | - Young Do
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Patricia A Elkins
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Daniela Ennulat
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Jeffrey H Guss
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Jason Holt
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Michael R Jeune
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Andrew G King
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Jan C Klapwijk
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - H Fritz Kramer
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Nicholas J Kramer
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Susan B Laffan
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Paresh I Masuria
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Alan V McDougal
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Paul N Mortenson
- Astex Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, UK
| | - Caterina Musetti
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Gregory E Peckham
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Beth L Pietrak
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Chuck Poole
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Daniel J Price
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Alan R Rendina
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Girish Sati
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Gordon Saxty
- Astex Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, UK
| | - Barry G Shearer
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Lisa M Shewchuk
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Helen F Sneddon
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, UK
| | - Eugene L Stewart
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - J Darren Stuart
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Dean N Thomas
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Stephen A Thomson
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Paris Ward
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Joseph W Wilson
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Tiahshun Xu
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Mark A Youngman
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| |
Collapse
|
14
|
Alayi T, Tawalbeh SM, Ogundele M, Smith HR, Samsel AM, Barbieri ML, Hathout Y. Tandem Mass Tag-Based Serum Proteome Profiling for Biomarker Discovery in Young Duchenne Muscular Dystrophy Boys. ACS OMEGA 2020; 5:26504-26517. [PMID: 33110978 PMCID: PMC7581259 DOI: 10.1021/acsomega.0c03206] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/23/2020] [Indexed: 06/11/2023]
Abstract
Blood-accessible molecular biomarkers are becoming highly attractive tools to assess disease progression and response to therapies in Duchenne muscular dystrophy (DMD) especially in very young patients for whom other outcome measures remain subjective and challenging. In this study, we have standardized a highly specific and reproducible multiplexing mass spectrometry method using the tandem mass tag (TMT) strategy in combination with depletion of abundant proteins from serum and high-pH reversed-phase peptide fractionation. Differential proteome profiling of 4 year-old DMD boys (n = 9) and age-matched healthy controls (n = 9) identified 38 elevated and 50 decreased serum proteins (adjusted P < 0.05, FDR <0.05) in the DMD group relative to the healthy control group. As expected, we confirmed previously reported biomarkers but also identified novel biomarkers. These included novel muscle injury-associated biomarkers such as telethonin, smoothelin-like protein 1, cofilin-1, and plectin, additional muscle-specific enzymes such as UTP-glucose-1-phosphate uridylyltransferase, aspartate aminotransferase, pyruvate kinase PKM, lactotransferrin, tissue alpha-l-fucosidase, pantetheinase, and ficolin-1, and some pro-inflammatory and cell adhesion-associated biomarkers such as leukosialin, macrophage receptor MARCO, vitronectin, galectin-3-binding protein, and ProSAAS. The workflow including serum depletion, sample processing, and mass spectrometry analysis was found to be reproducible and stable over time with CV < 20%. Furthermore, the method was found to be superior in terms of specificity compared to other multiplexing affinity-based methods. These findings demonstrate the specificity and reliability of TMT-based mass spectrometry methods in detection and identification of serum biomarkers in presymptomatic young DMD patients.
Collapse
Affiliation(s)
- Tchilabalo
D. Alayi
- Department
of Pharmaceutical Science, School of Pharmacy and Pharmaceutical Sciences, Binghamton University−SUNY, Johnson City, New York 13790, United States
| | - Shefa M. Tawalbeh
- Department
of Pharmaceutical Science, School of Pharmacy and Pharmaceutical Sciences, Binghamton University−SUNY, Johnson City, New York 13790, United States
- Department
of Biomedical Engineering, Binghamton University−SUNY, 4400 Vestal Pkwy E, Binghamton, New York 13902, United States
| | - Michael Ogundele
- Department
of Pharmaceutical Science, School of Pharmacy and Pharmaceutical Sciences, Binghamton University−SUNY, Johnson City, New York 13790, United States
- Department
of Biomedical Engineering, Binghamton University−SUNY, 4400 Vestal Pkwy E, Binghamton, New York 13902, United States
| | - Holly R. Smith
- Department
of Pharmaceutical Science, School of Pharmacy and Pharmaceutical Sciences, Binghamton University−SUNY, Johnson City, New York 13790, United States
- Department
of Biochemistry, Binghamton University−SUNY, 4400 Vestal Pkwy E, Binghamton, New York 13902, United States
| | - Alison M. Samsel
- Department
of Pharmaceutical Science, School of Pharmacy and Pharmaceutical Sciences, Binghamton University−SUNY, Johnson City, New York 13790, United States
| | - Marissa L. Barbieri
- Department
of Pharmaceutical Science, School of Pharmacy and Pharmaceutical Sciences, Binghamton University−SUNY, Johnson City, New York 13790, United States
| | - Yetrib Hathout
- Department
of Pharmaceutical Science, School of Pharmacy and Pharmaceutical Sciences, Binghamton University−SUNY, Johnson City, New York 13790, United States
| |
Collapse
|
15
|
Blaschek A, Rodrigues M, Ille L, Idriess M, Well T, Warken B, Müller C, Hannibal I, Tacke M, Müller-Felber W, Vill K. Is Exercise-Induced Fatigue a Problem in Children with Duchenne Muscular Dystrophy? Neuropediatrics 2020; 51:342-348. [PMID: 32369835 DOI: 10.1055/s-0040-1708859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVE Duchenne muscular dystrophy (DMD) is a devastating X-linked muscular disorder. The number of studies investigating new therapeutic approaches is substantially increasing. This study aims to investigate the impact and diagnostic value of exercise-induced fatigue in DMD, which has been proposed as a suitable outcome parameter in other conditions like spinal muscular atrophy. PATIENTS AND METHODS A cohort of 55 DMD patients (49 of them treated with steroids and 9 with ataluren) underwent a total of 241 6MWT (mean 4.4 tests/patient) which were retrospectively analyzed. Exercise-induced fatigue was assessed by the ratio between the distance achieved in the sixth minute and the distance in the second minute of the 6MWT. In previous studies a quotient above 1 was defined as a sign of fatigue. RESULTS The average fatigue quotient in the whole cohort of patients was 1.0. In a further analysis no impact of age, steroid therapy, ataluren therapy, overall disability, and distance in the 6-minute walk test (6MWT) on fatigue in DMD patients could be shown. CONCLUSION Our data show that fatigue does not play a relevant role in DMD. Analysis of fatigue is not a useful outcome parameter in DMD studies. For this reason we suggest the 2MWT, which is better accepted by the patients, as an alternative to the commonly 6MWT.
Collapse
Affiliation(s)
- Astrid Blaschek
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Martin Rodrigues
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Lena Ille
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Mohammed Idriess
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Therese Well
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Birgit Warken
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Christine Müller
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Iris Hannibal
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Moritz Tacke
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Wolfgang Müller-Felber
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Katharina Vill
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| |
Collapse
|
16
|
Population-Wide Duchenne Muscular Dystrophy Carrier Detection by CK and Molecular Testing. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8396429. [PMID: 33029525 PMCID: PMC7537677 DOI: 10.1155/2020/8396429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/27/2020] [Accepted: 09/03/2020] [Indexed: 01/28/2023]
Abstract
Carrier screening of Duchenne muscular dystrophy (DMD) has not been widely evaluated. To identify definite DMD female carriers prior to or in early pregnancy, we studied a large population of reproductive age females and provided informed reproductive options to DMD carriers. 37268 females were recruited from the Hangzhou Family Planning Publicity and Technology Guidance Station/Hangzhou Health Service Center for Children and Women, Hangzhou, China, between October 10, 2017, and December 16, 2018. CK activity was measured with follow-up serum DMD genetic testing in subjects with hyperCKemia, defined as CK > 200 U/L. The calculated upper reference limit (97.5th percentile) of serum creatine kinase (CK) for females aged 20-50 years in this study was near the reference limit recommended by the manufacturer (200 U/L), above which was defined as hyperCKemia. 427 females (1.2%) harbored initially elevated CK, among which 281 females (response rate of 65.8%) accepted CK retesting. DMD genetic testing was conducted on 62 subjects with sustained serum CK > 200 U/L and 16 females with a family history of DMD. Finally, 6 subjects were confirmed to be DMD definite carriers. The estimated DMD female carrier rate in this study was 1 : 4088 (adjusting for response rate), an underestimated rate, since only 50% to 70% of DMD female carriers manifest elevated serum CK, and carriers in this study may have been missed due to lack of follow-up or inability to detect all DMD pathogenic variants by current genetic testing.
Collapse
|
17
|
Interleukin-6: A neuro-active cytokine contributing to cognitive impairment in Duchenne muscular dystrophy? Cytokine 2020; 133:155134. [DOI: 10.1016/j.cyto.2020.155134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/13/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022]
|
18
|
Huang L, Li L, Li Q, Chen J, Lin S, Li K, Fan D, Jin W, Li Y, Yang X, Xiong Y, Li M, Yang X. Different Clinical Phenotypes Caused by Three F8 Missense Mutations in Three Chinese Families with Moderate Hemophilia A. DNA Cell Biol 2020; 39:1685-1690. [PMID: 32589464 DOI: 10.1089/dna.2020.5359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In families with a monogenic disorder, the causal mutation usually cosegregates with the disease phenotype. In rare cases, however, individuals carrying the same mutation within a family may show various phenotypes. This study aimed to analyze the discrepancy between genotype and phenotype in three families with moderate hemophilia A (HA) caused by missense mutation in the F8 gene. Among the 67 noninversion moderate HA families in our cohort, incomplete penetrance was found in three families. In these three families, the grandfathers were asymptomatic, whereas the probands had different clinical phenotypes. Apart from one mutation in the VWF gene (c.1330 G>A) found in one grandfather, only one missense mutation (c.5837A>T, c.6679G>A, and c.6506G>A, in the respective families) in the F8 gene was identified in each proband and their grandfathers. Subsequent Sanger sequencing results combined with bioinformatic analysis indicated that the three missense mutations in the F8 gene were the causative mutations. Our study demonstrated incomplete penetrance of missense mutation within HA families in China. Therefore, genetic counseling and management of such cases need to be reappraised.
Collapse
Affiliation(s)
- Limin Huang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Liyan Li
- Technology Center of Prenatal Diagnosis and Genetic Diseases Diagnosis, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiang Li
- The Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Juanjuan Chen
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Sheng Lin
- Lab of Molecular Medicine, Shenzhen Health Development Research Center, Shenzhen, China
| | - Kun Li
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Dongmei Fan
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Wangjie Jin
- Technology Center of Prenatal Diagnosis and Genetic Diseases Diagnosis, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yihong Li
- Technology Center of Prenatal Diagnosis and Genetic Diseases Diagnosis, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xu Yang
- Clinical Innovation & Research Center, Shenzhen Hospital of Southern Medical University, Shenzhen, China
| | - Yufeng Xiong
- Department of Clinical Laboratory, Guangdong Women and Children's Hospital, Guangzhou, China
| | - Ming Li
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xuexi Yang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| |
Collapse
|
19
|
Bulut N, Aydın G, Alemdaroğlu-Gürbüz İ, Karaduman A, Yılmaz Ö. Pulmonary and upper limbs function in children with early stage Duchenne muscular dystrophy compared to their healthy peers. Braz J Phys Ther 2020; 25:251-255. [PMID: 32553415 DOI: 10.1016/j.bjpt.2020.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 05/20/2020] [Accepted: 05/28/2020] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Pulmonary and upper limbs function of children with Duchenne muscular dystrophy (DMD) are known to deteriorate throughout the disease process. However, there is a lack of information on the extent of impairments in the early stages of DMD when compared to healthy peers. OBJECTIVE To investigate to what extent pulmonary and upper limbs function of children with early stage DMD are impaired. METHODS Sixty-one children participated in the study: 31 with Grade 1 DMD (study group) according to the Brooke Upper and Lower Extremity Functional Classification Systems, and 30 age matched healthy peers (control group). Pulmonary function was determined with pulmonary function tests. The Performance of Upper Limb test was used to evaluate the upper limbs function. RESULTS Study and control groups were homogenous in terms of physical characteristics (p>0.05). Pulmonary and upper limbs function of children with DMD were about 85% and 93% of healthy peers, respectively. CONCLUSION This study provides evidence for deterioration of pulmonary and upper limbs function in children with early stage DMD. Better knowledge of deterioration rate over time may help therapists to better plan and update their plan of care.
Collapse
Affiliation(s)
- Numan Bulut
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey.
| | - Güllü Aydın
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey
| | | | - Ayşe Karaduman
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey
| | - Öznur Yılmaz
- Faculty of Physical Therapy and Rehabilitation, Hacettepe University, Ankara, Turkey
| |
Collapse
|
20
|
He R, Li H, Wang L, Li Y, Zhang Y, Chen M, Zhu Y, Zhang C. Engraftment of human induced pluripotent stem cell-derived myogenic progenitors restores dystrophin in mice with duchenne muscular dystrophy. Biol Res 2020; 53:22. [PMID: 32430065 PMCID: PMC7238630 DOI: 10.1186/s40659-020-00288-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is a devastating genetic muscular disorder with no effective treatment that is caused by the loss of dystrophin. Human induced pluripotent stem cells (hiPSCs) offer a promising unlimited resource for cell-based therapies of muscular dystrophy. However, their clinical applications are hindered by inefficient myogenic differentiation, and moreover, the engraftment of non-transgene hiPSC-derived myogenic progenitors has not been examined in the mdx mouse model of DMD. Methods We investigated the muscle regenerative potential of myogenic progenitors derived from hiPSCs in mdx mice. The hiPSCs were transfected with enhanced green fluorescent protein (EGFP) vector and defined as EGFP hiPSCs. Myogenic differentiation was performed on EGFP hiPSCs with supplementary of basic fibroblast growth factor, forskolin, 6-bromoindirubin-3′-oxime as well as horse serum. EGFP hiPSCs-derived myogenic progenitors were engrafted into mdx mice via both intramuscular and intravenous injection. The restoration of dystrophin expression, the ratio of central nuclear myofibers, and the transplanted cells-derived satellite cells were accessed after intramuscular and systemic transplantation. Results We report that abundant myogenic progenitors can be generated from hiPSCs after treatment with these three small molecules, with consequent terminal differentiation giving rise to mature myotubes in vitro. Upon intramuscular or systemic transplantation into mdx mice, these myogenic progenitors engrafted and contributed to human-derived myofiber regeneration in host muscles, restored dystrophin expression, ameliorated pathological lesions, and seeded the satellite cell compartment in dystrophic muscles. Conclusions This study demonstrates the muscle regeneration potential of myogenic progenitors derived from hiPSCs using non-transgenic induction methods. Engraftment of hiPSC-derived myogenic progenitors could be a potential future therapeutic strategy to treat DMD in a clinical setting.
Collapse
Affiliation(s)
- Ruojie He
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, Guangdong, China
| | - Huan Li
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, Guangdong, China
| | - Liang Wang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.,Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, Guangdong, China
| | - Yaqin Li
- Department of Neurology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yu Zhang
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Menglong Chen
- Department of Neurology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Yuling Zhu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China. .,Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, Guangdong, China.
| | - Cheng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China. .,Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, Guangdong, China.
| |
Collapse
|
21
|
Killian M, Buchowski MS, Donnelly T, Burnette WB, Markham LW, Slaughter JC, Xu M, Crum K, Damon BM, Soslow JH. Beyond ambulation: Measuring physical activity in youth with Duchenne muscular dystrophy. Neuromuscul Disord 2020; 30:277-282. [PMID: 32291149 PMCID: PMC7234926 DOI: 10.1016/j.nmd.2020.02.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/08/2020] [Accepted: 02/13/2020] [Indexed: 01/04/2023]
Abstract
Patients with Duchenne muscular dystrophy (DMD) develop skeletal muscle weakness and cardiomyopathy. Validated skeletal muscle outcome measures are limited to ambulatory patients, but most DMD patients in cardiac trials are non-ambulatory. New objective functional assessments are needed. This study's objective was to assess the correlation and longitudinal change of two measures: quantitative muscle testing (QMT) and accelerometry. Patients with DMD were prospectively enrolled and underwent QMT and wore wrist and ankle accelerometers for seven days at baseline, 1-, and 2-years. QMT measures were indexed to age. Accelerometer recordings were total vector magnitudes and awake vector magnitude. Correlations were assessed using a Spearman correlation, and longitudinal change was evaluated using a paired t-test or a Wilcoxon signed rank test. Forty-eight participants were included. QMT and accelerometry measures had a moderate or strong correlation, particularly indexed arm QMT with total wrist vector magnitude (rho=0.85, p<0.001), total indexed QMT with total wrist vector magnitude (rho=0.8, p<0.001) and indexed leg QMT with total ankle vector magnitude (rho=0.69, p<0.001). QMT and accelerometry measures declined significantly over time. Accelerometry correlates with QMT and indexed QMT in boys with DMD. A combination of QMT and accelerometry may provide a complementary assessment of skeletal muscle function in non-ambulatory boys with DMD.
Collapse
Affiliation(s)
- Mary Killian
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Maciej S. Buchowski
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Thomas Donnelly
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - W. Bryan Burnette
- Division of Neurology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Larry W. Markham
- Division of Cardiology, Department of Pediatrics, Riley Hospital for Children at Indiana Health, Indianapolis, IN
| | - James C. Slaughter
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Meng Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Kimberly Crum
- Thomas P Graham Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Bruce M. Damon
- Departments of Radiology and Radiological Sciences, Molecular Physiology and Biophysics, and Biomedical Engineering, Vanderbilt University Medical Center, Nashville, TN
| | - Jonathan H. Soslow
- Thomas P Graham Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
22
|
Stem Cell Aging in Skeletal Muscle Regeneration and Disease. Int J Mol Sci 2020; 21:ijms21051830. [PMID: 32155842 PMCID: PMC7084237 DOI: 10.3390/ijms21051830] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/04/2020] [Accepted: 03/04/2020] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscle comprises 30-40% of the weight of a healthy human body and is required for voluntary movements in humans. Mature skeletal muscle is formed by multinuclear cells, which are called myofibers. Formation of myofibers depends on the proliferation, differentiation, and fusion of muscle progenitor cells during development and after injury. Muscle progenitor cells are derived from muscle satellite (stem) cells (MuSCs), which reside on the surface of the myofiber but beneath the basement membrane. MuSCs play a central role in postnatal maintenance, growth, repair, and regeneration of skeletal muscle. In sedentary adult muscle, MuSCs are mitotically quiescent, but are promptly activated in response to muscle injury. Physiological and chronological aging induces MuSC aging, leading to an impaired regenerative capability. Importantly, in pathological situations, repetitive muscle injury induces early impairment of MuSCs due to stem cell aging and leads to early impairment of regeneration ability. In this review, we discuss (1) the role of MuSCs in muscle regeneration, (2) stem cell aging under physiological and pathological conditions, and (3) prospects related to clinical applications of controlling MuSCs.
Collapse
|
23
|
Bylo M, Farewell R, Coppenrath VA, Yogaratnam D. A Review of Deflazacort for Patients With Duchenne Muscular Dystrophy. Ann Pharmacother 2020; 54:788-794. [PMID: 32019318 DOI: 10.1177/1060028019900500] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Objective: The objective of this article is to review the pharmacology, pharmacokinetics, efficacy, safety, dosage and administration, and formulary considerations of deflazacort. Data Sources: A search of MEDLINE and EMBASE (1946 to December 31, 2019) was conducted using the terms deflazacort and Duchenne muscular dystrophy (DMD). Results were limited to clinical trials, humans, and English. Additional sources and data were obtained from the references of included articles and prescribing information. Study Selection and Data Extraction: All articles published after July 2014 related to pharmacology, pharmacokinetics, efficacy, or safety of the therapy in human subjects were included. Data Synthesis: Deflazacort 0.9 mg/kg/d is a once-daily oral corticosteroid and is the first drug of its class to be Food and Drug Administration (FDA) approved for DMD. Studies with deflazacort show improved functional outcomes, delayed onset of cardiomyopathy, reduction in scoliosis surgery, and improved survival, but these improvements are supported by relatively weak evidence. Relevance to Patient Care and Clinical Practice: This review presents data from studies published after the most recent DMD 2016 treatment guidelines and offers prescribing considerations, including pharmacology, pharmacokinetics, adverse effects, formulary considerations, and areas of uncertainty. Conclusions: Deflazacort presents an additional, FDA-approved corticosteroid option for patients that offers improved quality of life for DMD patients. However, there is weak evidence to support these benefits; a full risk-benefit analysis considering adverse events, efficacy, cost, and previous trials of steroid therapy is necessary when selecting therapy. Further research will help clarify deflazacort's optimal dose, duration of treatment, and impact on quality of life.
Collapse
Affiliation(s)
- Mary Bylo
- MCPHS University, Worcester, MA, USA
| | | | | | | |
Collapse
|
24
|
Meng J, Sweeney NP, Doreste B, Muntoni F, McClure M, Morgan J. Restoration of Functional Full-Length Dystrophin After Intramuscular Transplantation of Foamy Virus-Transduced Myoblasts. Hum Gene Ther 2020; 31:241-252. [PMID: 31801386 PMCID: PMC7047098 DOI: 10.1089/hum.2019.224] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 11/24/2019] [Indexed: 12/12/2022] Open
Abstract
Stem cell therapy is a promising strategy to treat muscle diseases such as Duchenne muscular dystrophy (DMD). To avoid immune rejection of donor cells or donor-derived muscle, autologous cells, which have been genetically modified to express dystrophin, are preferable to cells derived from healthy donors. Restoration of full-length dystrophin (FL-dys) using viral vectors is extremely challenging, due to the limited packaging capacity of the vectors, but we have recently shown that either a foamy viral or lentiviral vector is able to package FL-dys open-reading frame and transduce myoblasts derived from a DMD patient. Differentiated myotubes derived from these transduced cells produced FL-dys. Here, we transplanted the foamy viral dystrophin-corrected DMD myoblasts intramuscularly into mdx nude mice, and showed that the transduced cells contributed to muscle regeneration, expressing FL-dys in nearly all the muscle fibers of donor origin. Furthermore, we showed that the restored FL-dys recruited members of the dystrophin-associated protein complex and neuronal nitric oxide synthase within donor-derived muscle fibers, evidence that the restored dystrophin protein is functional. Dystrophin-expressing donor-derived muscle fibers expressed lower levels of utrophin than host muscle fibers, providing additional evidence of functional improvement of donor-derived myofibers. This is the first in vivo evidence that foamy virus vector-transduced DMD myoblasts can contribute to muscle regeneration and mediate functional dystrophin restoration following their intramuscular transplantation, representing a promising therapeutic strategy for individual small muscles in DMD.
Collapse
Affiliation(s)
- Jinhong Meng
- Developmental Neuroscience Programme, Molecular Neurosciences Section, Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, United Kingdom
| | - Nathan Paul Sweeney
- Jefferiss Research Trust Laboratories, Imperial College London, London, United Kingdom
| | - Bruno Doreste
- Developmental Neuroscience Programme, Molecular Neurosciences Section, Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, United Kingdom
| | - Francesco Muntoni
- Developmental Neuroscience Programme, Molecular Neurosciences Section, Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, United Kingdom
| | - Myra McClure
- Jefferiss Research Trust Laboratories, Imperial College London, London, United Kingdom
| | - Jennifer Morgan
- Developmental Neuroscience Programme, Molecular Neurosciences Section, Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, United Kingdom
| |
Collapse
|
25
|
Affiliation(s)
| | | | - Indu Mahabeer
- Sandwell & West Birmingham Hospital NHS Trust, Midlands, UK
| | - Henriette van Ruiten
- The Great North Children's Hospital and The John Walton Muscular Dystrophy Research Centre, Newcastle, UK
| |
Collapse
|
26
|
Souza LBD, Maziero C, Lazzarin MC, Quintana HT, Tomé TDC, Baptista VIDA, de Oliveira F. Presence of metalloproteinases 2 and 9 and 8-OHdG in the fibrotic process in skeletal muscle of Mdx mice. Acta Histochem 2020; 122:151458. [PMID: 31699373 DOI: 10.1016/j.acthis.2019.151458] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/13/2019] [Accepted: 10/07/2019] [Indexed: 12/29/2022]
Abstract
Inflammation and oxidative stress occurs in muscle of Duchenne muscular dystrophy (DMD). The relationship between a panel of biomarkers and the DMD outcome is necessary to indicate of disease progression and response to rehabilitation programs. The aim was to analyze the connective tissue of muscle of Mdx mice and immunoexpression of MMP-2, MMP-9, and 8-OHdG, which signalizes oxidative stress related to DNA damage. Biceps brachii of male C57BL/10 and C57BL/10-Dmdmdx mice was submitted to Hematoxylin-Eosin, Sirius red and immunohistochemistry (MMP-2, MMP-9 and 8-OHdG) analysis. Mdx showed focal lesions with intense inflammation and fibrosis related to immunoexpression of MMP-2 and MMP-9, proving the hypothesis that these MMPs are linked to muscular tissue degeneration, which can be regenerated by their inhibition, improving the treatment of DMD carriers. Histopathological findings related to centralized nuclei increase were related to higher 8-OHdG immunomarked nuclei in Mdx, which signalizes oxidative stress associated with DNA damage provoked by DMD. Such result shows that the evaluation of 8-OHdG during the evolution of the disease could be a method to evaluate DMD disease progression.
Collapse
Affiliation(s)
- Lidiane Begalli de Souza
- Departamento de Biociências, Universidade Federal de São Paulo, Campus Baixada Santista, SP, Brazil
| | - Carla Maziero
- Departamento de Biociências, Universidade Federal de São Paulo, Campus Baixada Santista, SP, Brazil
| | - Mariana Cruz Lazzarin
- Departamento de Biociências, Universidade Federal de São Paulo, Campus Baixada Santista, SP, Brazil
| | | | - Tabata de Carvalho Tomé
- Departamento de Biociências, Universidade Federal de São Paulo, Campus Baixada Santista, SP, Brazil
| | | | - Flavia de Oliveira
- Departamento de Biociências, Universidade Federal de São Paulo, Campus Baixada Santista, SP, Brazil.
| |
Collapse
|
27
|
Clay A, Hearle P, Schadt K, Lynch DR. New developments in pharmacotherapy for Friedreich ataxia. Expert Opin Pharmacother 2019; 20:1855-1867. [PMID: 31311349 DOI: 10.1080/14656566.2019.1639671] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Friedreich ataxia (FRDA), a rare disease caused by the deficiency of the mitochondrial matrix protein frataxin, affects roughly 1 in 50,000 individuals worldwide. Current and emerging therapies focus on reversing the deleterious effects of such deficiency including mitochondrial augmentation and increasing frataxin levels, providing the possibility of treatment options for this physiologically complex, multisystem disorder. Areas covered: In this review article, the authors discuss the current and prior in vivo and in vitro research studies related to the treatment of FRDA, with a particular interest in future implications of each therapy. Expert opinion: Since the discovery of FXN in 1996, multiple clinical trials have occurred or are currently occurring; at a rapid pace for a rare disease. These trials have been directed at the augmentation of mitochondrial function and/or alleviation of symptoms and are not regarded as potential cures in FRDA. Either a combination of therapies or a drug that replaces or increases the pathologically low levels of frataxin better represent potential cures in FRDA.
Collapse
Affiliation(s)
- Alexandra Clay
- Division of Neurology, Children's Hospital of Philadelphia , Philadelphia , PA , USA
| | - Patrick Hearle
- Division of Neurology, Children's Hospital of Philadelphia , Philadelphia , PA , USA
| | - Kim Schadt
- Division of Neurology, Children's Hospital of Philadelphia , Philadelphia , PA , USA
| | - David R Lynch
- Division of Neurology, Children's Hospital of Philadelphia , Philadelphia , PA , USA
| |
Collapse
|
28
|
Echevarría L, Aupy P, Goyenvalle A. Exon-skipping advances for Duchenne muscular dystrophy. Hum Mol Genet 2019; 27:R163-R172. [PMID: 29771317 DOI: 10.1093/hmg/ddy171] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 05/03/2018] [Indexed: 12/19/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal genetic disorder characterized by progressive muscle wasting that has currently no cure. Exon-skipping strategy represents one of the most promising therapeutic approaches that aim to restore expression of a shorter but functional dystrophin protein. The antisense field has remarkably progress over the last years with recent accelerated approval of the first antisense oligonucleotide-based therapy for DMD, Exondys 51, though the therapeutic benefit remains to be proved in patients. Despite clinical advances, the poor effective delivery to target all muscle remains the main hurdle for antisense drug therapy. This review describes the antisense-based exon-skipping approach for DMD, from proof-of-concept to first marketed drug. We discuss the main obstacles to achieve a successful exon-skipping therapy and the latest advances of the international community to develop more powerful chemistries and more sophisticated delivery systems in order to increase potency, bioavailability and safety. Finally, we highlight the importance of collaborative efforts and early dialogue between drug developers and regulatory agencies in order to overcome difficulties, find appropriate outcome markers and collect useful data.
Collapse
Affiliation(s)
- Lucía Echevarría
- U1179 INSERM, UFR des Sciences de la Santé, Montigny le Bretonneux, France.,SQY Therapeutics, Université de Versailles St-Quentin, Montigny le Bretonneux, France
| | - Philippine Aupy
- U1179 INSERM, UFR des Sciences de la Santé, Montigny le Bretonneux, France
| | - Aurélie Goyenvalle
- U1179 INSERM, UFR des Sciences de la Santé, Montigny le Bretonneux, France
| |
Collapse
|
29
|
S. Ferreira G, Veening-Griffioen DH, Boon WPC, Moors EHM, Gispen-de Wied CC, Schellekens H, van Meer PJK. A standardised framework to identify optimal animal models for efficacy assessment in drug development. PLoS One 2019; 14:e0218014. [PMID: 31194784 PMCID: PMC6563989 DOI: 10.1371/journal.pone.0218014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 05/23/2019] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Poor translation of efficacy data derived from animal models can lead to clinical trials unlikely to benefit patients-or even put them at risk-and is a potential contributor to costly and unnecessary attrition in drug development. OBJECTIVES To develop a tool to assess, validate and compare the clinical translatability of animal models used for the preliminary assessment of efficacy. DESIGN AND RESULTS We performed a scoping review to identify the key aspects used to validate animal models. Eight domains (Epidemiology, Symptomatology and Natural History-SNH, Genetic, Biochemistry, Aetiology, Histology, Pharmacology and Endpoints) were identified. We drafted questions to evaluate the different facets of human disease simulation. We designed the Framework to Identify Models of Disease (FIMD) to include standardised instructions, a weighting and scoring system to compare models as well as factors to help interpret model similarity and evidence uncertainty. We also added a reporting quality and risk of bias assessment of drug intervention studies in the Pharmacological Validation domain. A web-based survey was conducted with experts from different stakeholders to gather input on the framework. We conducted a pilot study of the validation in two models for Type 2 Diabetes (T2D)-the ZDF rat and db/db mouse. Finally, we present a full validation and comparison of two animal models for Duchenne Muscular Dystrophy (DMD): the mdx mouse and GRMD dog. We show that there are significant differences between the mdx mouse and the GRMD dog, the latter mimicking the human epidemiological, SNH, and histological aspects to a greater extent than the mouse despite the overall lack of published data. CONCLUSIONS FIMD facilitates drug development by serving as the basis to select the most relevant model that can provide meaningful data and is more likely to generate translatable results to progress drug candidates to the clinic.
Collapse
Affiliation(s)
- Guilherme S. Ferreira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Désirée H. Veening-Griffioen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Wouter P. C. Boon
- Copernicus Institute of Sustainable Development, Innovation Studies, Utrecht University, Utrecht, The Netherlands
| | - Ellen H. M. Moors
- Copernicus Institute of Sustainable Development, Innovation Studies, Utrecht University, Utrecht, The Netherlands
| | | | - Huub Schellekens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Peter J. K. van Meer
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Medicines Evaluation Board, Utrecht, The Netherlands
| |
Collapse
|
30
|
Paquin RS, Fischer R, Mansfield C, Mange B, Beaverson K, Ganot A, Martin AS, Morris C, Rensch C, Ricotti V, Russo LJ, Sadosky A, Smith EC, Peay HL. Priorities when deciding on participation in early-phase gene therapy trials for Duchenne muscular dystrophy: a best-worst scaling experiment in caregivers and adult patients. Orphanet J Rare Dis 2019; 14:102. [PMID: 31072340 PMCID: PMC6509771 DOI: 10.1186/s13023-019-1069-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/17/2019] [Indexed: 01/14/2023] Open
Abstract
PURPOSE Several gene therapy trials for Duchenne muscular dystrophy initiated in 2018. Trial decision making is complicated by non-curative, time-limited benefits; the progressive, fatal course; and high unmet needs. Here, caregivers and patients prioritize factors influencing decision making regarding participation in early-phase gene therapy trials. METHODS We conducted a best-worst scaling experiment among U.S. caregivers and adults with Duchenne (N = 274). Participants completed 11 choice sets, choosing features they cared about most and least when deciding whether to participate in a hypothetical gene therapy trial. We analyzed the data using sequential conditional logistic regression. RESULTS Participants prioritized improved muscle function in trial decision making. Concerns about participation limiting later use of gene transfer and editing were also important, as were improved lung and heart function. Low risk of death fell near the middle. Participants cared least about muscle biopsies and potential for randomization to placebo. Adults with Duchenne and caregivers of non-ambulatory children significantly prioritized improved lung function compared to caregivers of ambulatory children. CONCLUSION Our data demonstrate prioritization of anticipated benefits and opportunity costs relative to potential harms and procedures in gene therapy trial decision making. Such data inform protocol development, education and advocacy efforts, and informed consent.
Collapse
Affiliation(s)
- Ryan S Paquin
- RTI International, Research Triangle Park, North Carolina, USA
| | - Ryan Fischer
- Parent Project Muscular Dystrophy, Hackensack, NJ, USA
| | - Carol Mansfield
- RTI Health Solutions, Research Triangle Park, North Carolina, USA
| | - Brennan Mange
- RTI Health Solutions, Research Triangle Park, North Carolina, USA
| | | | | | | | | | - Colin Rensch
- Parent Project Muscular Dystrophy, Hackensack, NJ, USA
| | | | | | | | | | - Holly L Peay
- RTI International, Research Triangle Park, North Carolina, USA.
| |
Collapse
|
31
|
Landrum Peay H, Fischer R, Tzeng JP, Hesterlee SE, Morris C, Strong Martin A, Rensch C, Smith E, Ricotti V, Beaverson K, Wand H, Mansfield C. Gene therapy as a potential therapeutic option for Duchenne muscular dystrophy: A qualitative preference study of patients and parents. PLoS One 2019; 14:e0213649. [PMID: 31042754 PMCID: PMC6493713 DOI: 10.1371/journal.pone.0213649] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/26/2019] [Indexed: 01/14/2023] Open
Abstract
Objectives Duchenne muscular dystrophy (DMD) is a rare neuromuscular disorder that causes progressive weakness and early death. Gene therapy is an area of new therapeutic development. This qualitative study explored factors influencing parents’ and adult patients’ preferences about gene therapy. Methods We report qualitative data from 17 parents of children with DMD and 6 adult patients. Participants responded to a hypothetical gene therapy vignette with features including non-curative stabilizing benefits to muscle, cardiac and pulmonary function; a treatment-related risk of death; and one-time dosing with time-limited benefit of 8–10 years. We used NVivo 11 to code responses and conduct thematic analyses. Results All participants placed high value on benefits to skeletal muscle, cardiac, and pulmonary functioning, with the relative importance of cardiac and pulmonary function increasing with disease progression. More than half tolerated a hypothetical 1% risk of death when balanced against Duchenne progression and limited treatment options. Risk tolerance increased at later stages. Participants perceived a ‘right time’ to initiate gene therapy. Most preferred to wait until a highly-valued function was about to be lost. Conclusion Participants demonstrated a complex weighing of potential benefits against harms and the inevitable decline of untreated Duchenne. Disease progression increased risk tolerance as participants perceived fewer treatment options and placed greater value on maintaining remaining function. In the context of a one-time treatment like gene therapy, our finding that preferences about timing of initiation are influenced by disease state suggest the importance of assessing ‘lifetime’ preferences across the full spectrum of disease progression.
Collapse
Affiliation(s)
- Holly Landrum Peay
- Center for Newborn Screening, Ethics, and Disability Studies, RTI International, Research Triangle Park, North Caroilina, United States of America
- * E-mail:
| | - Ryan Fischer
- Parent Project Muscular Dystrophy, Hackensack, New Jersey, United States of America
| | - Janice P. Tzeng
- Center for Newborn Screening, Ethics, and Disability Studies, RTI International, Research Triangle Park, North Caroilina, United States of America
| | - Sharon E. Hesterlee
- Lion Therapeutics, Asklepios BioPharmaceutical, Inc., Research Triangle Park, North Carolina, United States of America
| | - Carl Morris
- Solid Biosciences, Cambridge, Massachusetts, United States of America
| | - Amy Strong Martin
- Center for Duchenne Muscular Dystrophy at UCLA, University of California Los Angeles, Los Angeles, California, United States of America
| | - Colin Rensch
- Parent Project Muscular Dystrophy, Hackensack, New Jersey, United States of America
| | - Edward Smith
- Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Valeria Ricotti
- Solid Biosciences, Cambridge, Massachusetts, United States of America
| | - Katherine Beaverson
- Rare Disease Research Unit, Pfizer, Inc, Cambridge, Massachusetts, United States of America
| | - Hannah Wand
- Stanford Healthcare and ClinGen, Sanford, California, United States of America
| | - Carol Mansfield
- RTI Health Solutions, RTI International, Research Triangle Park, North Carolina, United States of America
| |
Collapse
|
32
|
Korinthenberg R. A new era in the management of Duchenne muscular dystrophy. Dev Med Child Neurol 2019; 61:292-297. [PMID: 30556126 DOI: 10.1111/dmcn.14129] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2018] [Indexed: 12/27/2022]
Abstract
The management of Duchenne muscular dystrophy (DMD) has changed considerably over the last few decades. Standards of care have recently been updated, based on systematic reviews of the literature and structured, case-based consensus discussions of experts. Besides addressing muscular function, these recommendations cover most areas of organ involvement and psychosocial issues. Studies employing animal models investigating basic disease mechanisms have increased considerably. Development of valid functional outcome measures enabled high-evidence, clinical trials in males with DMD. Stop codon readthrough with ataluren has revealed some effect in patients with moderate disability and has been approved for males with suitable mutations who are still ambulant. Exon 51 skipping with eteplirsen demonstrated a structural and functional effect in a small group of patients and received restricted approval in the USA. Further trials investigating a broad range of mechanisms are underway. However, much more work is needed to develop more active treatments that stop disease progression. Likewise, the distribution of complex and expensive therapies to underprivileged patients and those in poorer regions must be improved. WHAT THIS PAPER ADDS: Updated standards of care covering most clinical aspects of Duchenne muscular dystrophy (DMD) are available. Adequately controlled clinical trials have allowed initial approval of disease-modifying drugs for small groups of patients. Scientific, economic, and political efforts are needed to make effective therapies available more quickly. Effective therapies should be made available more quickly to patients in low-income regions.
Collapse
Affiliation(s)
- Rudolf Korinthenberg
- Department of Neuropediatrics and Muscular Disorders, Medical Faculty, University of Freiburg, Freiburg, Germany
| |
Collapse
|
33
|
Kong R, Laskin OL, Kaushik D, Jin F, Ma J, McIntosh J, Souza M, Almstead N. Ataluren Pharmacokinetics in Healthy Japanese and Caucasian Subjects. Clin Pharmacol Drug Dev 2019; 8:172-178. [PMID: 30629861 PMCID: PMC6590116 DOI: 10.1002/cpdd.645] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/03/2018] [Indexed: 12/11/2022]
Abstract
To evaluate the potential for ethnicity-related differences in ataluren pharmacokinetics (PK) and safety, a phase 1 single-dose study was conducted in 48 healthy (24 Japanese and 24 Caucasian subjects), nonsmoking male volunteers who were equally divided into 3 cohorts of oral doses at 5, 10, and 20 mg/kg. Blood samples were collected until 48 hours postdose. PK results demonstrated rapid absorption of ataluren, with peak plasma levels (Cmax ) being attained between 0.875 and 2.5 hours after dosing. The mean Cmax and area under the concentration-time curve (AUC(0-last) ) increased with each increasing dose level in both Japanese and Caucasian subjects. Although the Cmax was similar across all subjects at each dose regardless of ethnicity, Japanese subjects had a mean AUC(0-last) approximately 14% to 34% lower than that of Caucasian subjects across the 3 dose levels. This difference was likely due to the higher variability of AUC values in Caucasian subjects and the relatively small study population. In conclusion, similar ataluren PK profiles were observed in healthy Japanese and Caucasian subjects following single oral administration of ataluren at all dose levels.
Collapse
Affiliation(s)
- Ronald Kong
- PTC Therapeutics, Inc, South Plainfield, NJ, USA
| | - Oscar L Laskin
- PTC Therapeutics, Inc, South Plainfield, NJ, USA.,R2D Pharma Services LLC, Princeton, NJ, USA
| | | | - Fengbin Jin
- PTC Therapeutics, Inc, South Plainfield, NJ, USA
| | - Jiyuan Ma
- PTC Therapeutics, Inc, South Plainfield, NJ, USA
| | | | - Marcio Souza
- PTC Therapeutics, Inc, South Plainfield, NJ, USA
| | | |
Collapse
|
34
|
Propp R, McAdam L, Davis AM, Salbach NM, Weir S, Encisa C, Narayanan UG. Development and content validation of the Muscular Dystrophy Child Health Index of Life with Disabilities questionnaire for children with Duchenne muscular dystrophy. Dev Med Child Neurol 2019; 61:75-81. [PMID: 30058069 DOI: 10.1111/dmcn.13977] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/22/2018] [Indexed: 02/04/2023]
Abstract
AIM To develop a patient-reported outcome measure that comprehensively captures the health-related priorities of children with Duchenne muscular dystrophy (DMD). METHOD Children with DMD and their parents completed the iteratively revised versions of the Caregiver Priorities and Child Health Index of Life with Disabilities (CPCHILD), followed by a cognitive interview to develop a pilot version of a new measure. Multidisciplinary health care professionals completed an item-by-item analysis of the measure and a 14-item sensibility questionnaire. Minimum content validity ratio for each item of the new measure and the mean score (0-7) for the items of the sensibility questionnaire were calculated. RESULTS The CPCHILD underwent changes over 19 interviews with children and their parents, resulting in the pilot Muscular Dystrophy Child Health Index of Life with Disabilities (MDCHILD). The content validity ratio of each MDCHILD item ranged from 0.85 to 1 based on health care professionals' ratings. The mean score exceeded the threshold of four for all items of the sensibility questionnaire. Based on child, parent, and health care professional recommendations, 16 items were added, six eliminated, and 15 items modified from the original CPCHILD. The MDCHILD consists of 47 items over seven domains. INTERPRETATION The MDCHILD met all sensibility criteria by children with DMD, their parents, and health care professionals, and is ready for psychometric evaluation. WHAT THIS PAPER ADDS The Muscular Dystrophy Child Health Index of Life with Disabilities (MDCHILD) is a new patient-reported outcome measure for Duchenne muscular dystrophy (DMD). The Priority Framework of Outcomes underpins the content for the MDCHILD. The MDCHILD incorporates the health-related priorities of males with DMD and their parents. The MDCHILD was deemed sensible by children, their parents, and health care professionals.
Collapse
Affiliation(s)
- Roni Propp
- Child Health Evaluative Sciences Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada.,Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada
| | - Laura McAdam
- Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada.,Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Aileen M Davis
- Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada.,Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Department of Physical Therapy, University of Toronto, Toronto, ON, Canada.,Institute of Health Policy, Management & Evaluation, University of Toronto, Toronto, ON, Canada
| | - Nancy M Salbach
- Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada.,Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Shannon Weir
- Child Health Evaluative Sciences Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Clarissa Encisa
- Child Health Evaluative Sciences Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada
| | - Unni G Narayanan
- Child Health Evaluative Sciences Program, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada.,Rehabilitation Sciences Institute, University of Toronto, Toronto, ON, Canada.,Holland Bloorview Kids Rehabilitation Hospital, Toronto, ON, Canada.,Department of Surgery, Division of Orthopaedics, The Hospital for Sick, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
35
|
Abstract
The ability to efficiently modify the genome using CRISPR technology has rapidly revolutionized biology and genetics and will soon transform medicine. Duchenne muscular dystrophy (DMD) represents one of the first monogenic disorders that has been investigated with respect to CRISPR-mediated correction of causal genetic mutations. DMD results from mutations in the gene encoding dystrophin, a scaffolding protein that maintains the integrity of striated muscles. Thousands of different dystrophin mutations have been identified in DMD patients, who suffer from a loss of ambulation followed by respiratory insufficiency, heart failure, and death by the third decade of life. Using CRISPR to bypass DMD mutations, dystrophin expression has been efficiently restored in human cells and mouse models of DMD. Here, we review recent progress toward the development of possible CRISPR therapies for DMD and highlight opportunities and potential obstacles in attaining this goal.
Collapse
Affiliation(s)
- Yi-Li Min
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA;
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA;
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA;
| |
Collapse
|
36
|
Dombernowsky NW, Ölmestig JNE, Witting N, Kruuse C. Role of neuronal nitric oxide synthase (nNOS) in Duchenne and Becker muscular dystrophies - Still a possible treatment modality? Neuromuscul Disord 2018; 28:914-926. [PMID: 30352768 DOI: 10.1016/j.nmd.2018.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/07/2018] [Accepted: 09/05/2018] [Indexed: 02/08/2023]
Abstract
Neuronal nitric oxide synthase (nNOS) is involved in nitric oxide (NO) production and suggested to play a crucial role in blood flow regulation of skeletal muscle. During activation of the muscle, NO helps attenuate the sympathetic vasoconstriction to accommodate increased metabolic demands, a phenomenon known as functional sympatholysis. In inherited myopathies such as the dystrophinopathies Duchenne and Becker muscle dystrophies (DMD and BMD), nNOS is lost from the sarcolemma. The loss of nNOS may cause functional ischemia contributing to skeletal and cardiac muscle cell injury. Effects of NO is augmented by inhibiting degradation of the second messenger cyclic guanosine monophosphate (cGMP) using sildenafil and tadalafil, both of which inhibit the enzyme phosphodiesterase 5 (PDE5). In animal models of DMD, PDE5-inhibitors prevent functional ischemia, reduce post-exercise skeletal muscle pathology and fatigue, show amelioration of cardiac muscle cell damage and increase cardiac performance. However, effect on clinical outcomes in DMD and BMD patients have been disappointing with minor effects on upper limb performance and none on ambulation. This review aims to summarize the current knowledge of nNOS function related to functional sympatholysis in skeletal muscle and studies on PDE5-inhibitor treatment in nNOS-deficient animal models and patients.
Collapse
Affiliation(s)
- Nanna W Dombernowsky
- Department of Neurology, Rigshospitalet Glostrup, University of Copenhagen, Denmark
| | - Joakim N E Ölmestig
- Department of Neurology, Neurovascular Research Unit, Herlev Gentofte Hospital, University of Copenhagen, Denmark
| | - Nanna Witting
- Department of Neurology, Rigshospitalet Glostrup, University of Copenhagen, Denmark
| | - Christina Kruuse
- Department of Neurology, Neurovascular Research Unit, Herlev Gentofte Hospital, University of Copenhagen, Denmark; PDE Research Group, Lundbeck Foundation Center for Neurovascular Research (LUCENS), Denmark.
| |
Collapse
|
37
|
Nutrition in Duchenne muscular dystrophy 16–18 March 2018, Zaandam, the Netherlands. Neuromuscul Disord 2018; 28:680-689. [DOI: 10.1016/j.nmd.2018.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 05/09/2018] [Indexed: 11/17/2022]
|
38
|
Matsuo M, Shirakawa T, Awano H, Nishio H. Receiver operating curve analyses of urinary titin of healthy 3-y-old children may be a noninvasive screening method for Duchenne muscular dystrophy. Clin Chim Acta 2018; 486:110-114. [PMID: 30053403 DOI: 10.1016/j.cca.2018.07.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/21/2018] [Accepted: 07/23/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a progressive, fatal muscle wasting disease. Early detection of DMD by mass screening may enable the early treatment of these patients. We have reported that urinary titin concentration, an indicator of severe muscle wasting, is a diagnostic biomarker for DMD. METHODS Urinary titin concentrations were measured in healthy 3-y-old children and, by comparison with concentrations in 4 DMD patients, and validated as a screening biomarker for DMD. Urine samples were obtained from 100 healthy Japanese children, 52 boys and 48 girls, and their urinary titin concentrations measured by ELISA. RESULTS The mean ± SD urinary titin concentration was 1.5 ± 2.5 nmol/l, and the mean urinary titin concentration normalized to creatinine was 2.2 ± 4.1 pmol/mg creatinine, with no differences between boys and girls. Histograms and box-and-whisker plots showed that almost all titin and normalized titin concentrations were in narrow ranges, with one outlier in common. Receiver operating characteristic curve analysis showed that titin and normalized-titin concentrations from healthy 3-y-olds were completely separate from those of 3-y-old DMD patients. CONCLUSIONS These findings indicate that urinary titin may be an excellent non-invasive biomarker to screen for DMD.
Collapse
Affiliation(s)
- Masafumi Matsuo
- Research Center for Locomotion Biology, Kobe Gakuin University, Kobe, Japan; KNC Department of Nucleic Acid Drug Discovery, Faculty of Rehabilitation, Kobe Gakuin University, Kobe, Japan.
| | - Taku Shirakawa
- Research Center for Locomotion Biology, Kobe Gakuin University, Kobe, Japan; KNC Department of Nucleic Acid Drug Discovery, Faculty of Rehabilitation, Kobe Gakuin University, Kobe, Japan.
| | - Hiroyuki Awano
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Hisahide Nishio
- Research Center for Locomotion Biology, Kobe Gakuin University, Kobe, Japan; Department of Occupational Therapy, Faculty of Rehabilitation, Kobe Gakuin University, Kobe, Japan.
| |
Collapse
|
39
|
Mohammed F, Elshafey A, Al-balool H, Alaboud H, Al Ben Ali M, Baqer A, Bastaki L. Mutation spectrum analysis of Duchenne/Becker muscular dystrophy in 68 families in Kuwait: The era of personalized medicine. PLoS One 2018; 13:e0197205. [PMID: 29847600 PMCID: PMC5976149 DOI: 10.1371/journal.pone.0197205] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 04/27/2018] [Indexed: 11/19/2022] Open
Abstract
Duchenne and Becker muscular dystrophies (DMD/BMD) are X-linked recessive neuromuscular disorders characterized by progressive irreversible muscle weakness and atrophy that affect both skeletal and cardiac muscles. DMD/BMD is caused by mutations in the Dystrophin gene on the X chromosome, leading to the absence of the essential muscle protein Dystrophin in DMD. In BMD, Dystrophin is partially functioning with a shorter protein product. Recent advances in molecular therapies for DMD require precise genetic diagnoses because most therapeutic strategies are mutation-specific. Hence, early diagnosis is crucial to allow appropriate planning for patient care and treatment. In this study, data from DMD/BMD patients who attended the Kuwait Medical Genetic Center during the last 20 years was retrieved from a Kuwait neuromuscular registry and analyzed. We combined multiplex PCR and multiplex ligation-dependent probe amplification (MLPA) with Sanger sequencing to detect Dystrophin gene mutations. A total of 35 different large rearrangements, 2 deletion-insertions (Indels) and 4 substitution mutations were identified in the 68 unrelated families. The deletion and duplication rates were 66.2% and 4.4%, respectively. The analyzed data from our registry revealed that 11 (16%) of the DMD families will benefit from newly introduced therapies (Ataluren and exon 51 skipping). At the time of submitting this paper, two cases have already enrolled in Ataluren (Tranlsarna™) therapy, and one case has been enrolled in exon 51 skipping therapy.
Collapse
Affiliation(s)
- Fawziah Mohammed
- Medical Laboratory Sciences, Faculty of Allied Health Sciences, Kuwait University, Jabriah, Kuwait
- * E-mail:
| | - Alaa Elshafey
- Kuwait Medical Genetic Centre, Ministry of Health, Shouaikh, Kuwait
| | - Haya Al-balool
- Kuwait Medical Genetic Centre, Ministry of Health, Shouaikh, Kuwait
| | - Hayat Alaboud
- Kuwait Medical Genetic Centre, Ministry of Health, Shouaikh, Kuwait
| | | | - Adel Baqer
- Kuwait Medical Genetic Centre, Ministry of Health, Shouaikh, Kuwait
| | - Laila Bastaki
- Kuwait Medical Genetic Centre, Ministry of Health, Shouaikh, Kuwait
| |
Collapse
|
40
|
Hildyard JC, Taylor-Brown F, Massey C, Wells DJ, Piercy RJ. Determination of qPCR Reference Genes Suitable for Normalizing Gene Expression in a Canine Model of Duchenne Muscular Dystrophy. J Neuromuscul Dis 2018; 5:177-191. [DOI: 10.3233/jnd-170267] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- John C.W. Hildyard
- Department of Clinical Science and Services, Comparative Neuromuscular Diseases Laboratory, Royal Veterinary College, London, UK
| | - Frances Taylor-Brown
- Department of Clinical Science and Services, Comparative Neuromuscular Diseases Laboratory, Royal Veterinary College, London, UK
| | - Claire Massey
- Department of Clinical Science and Services, Comparative Neuromuscular Diseases Laboratory, Royal Veterinary College, London, UK
| | - Dominic J. Wells
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Richard J. Piercy
- Department of Clinical Science and Services, Comparative Neuromuscular Diseases Laboratory, Royal Veterinary College, London, UK
| |
Collapse
|
41
|
Cordova G, Negroni E, Cabello-Verrugio C, Mouly V, Trollet C. Combined Therapies for Duchenne Muscular Dystrophy to Optimize Treatment Efficacy. Front Genet 2018; 9:114. [PMID: 29692797 PMCID: PMC5902687 DOI: 10.3389/fgene.2018.00114] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/22/2018] [Indexed: 01/01/2023] Open
Abstract
Duchene Muscular Dystrophy (DMD) is the most frequent muscular dystrophy and one of the most severe due to the absence of the dystrophin protein. Typical pathological features include muscle weakness, muscle wasting, degeneration, and inflammation. At advanced stages DMD muscles present exacerbated extracellular matrix and fat accumulation. Recent progress in therapeutic approaches has allowed new strategies to be investigated, including pharmacological, gene-based and cell-based therapies. Gene and cell-based therapies are still limited by poor targeting and low efficiency in fibrotic dystrophic muscle, therefore it is increasingly evident that future treatments will have to include “combined therapies” to reach maximal efficiency. The scope of this mini-review is to provide an overview of the current literature on such combined therapies for DMD. By “combined therapies” we mean those that include both a therapy to correct the genetic defect and an additional one to address one of the secondary pathological features of the disease. In this mini-review, we will not provide a comprehensive view of the literature on therapies for DMD, since many such reviews already exist, but we will focus on the characteristics, efficiency, and potential of such combined therapeutic strategies that have been described so far for DMD.
Collapse
Affiliation(s)
- Gonzalo Cordova
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, Paris, France
| | - Elisa Negroni
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, Paris, France
| | - Claudio Cabello-Verrugio
- Laboratorio de Patologías Musculares, Fragilidad y Envejecimiento, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andres Bello, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Vincent Mouly
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, Paris, France
| | - Capucine Trollet
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Association Institut de Myologie, Centre de Recherche en Myologie, UMRS974, Paris, France
| |
Collapse
|
42
|
Ismail HM, Dorchies OM, Scapozza L. The potential and benefits of repurposing existing drugs to treat rare muscular dystrophies. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1452733] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Hesham M. Ismail
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| | - Olivier M. Dorchies
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| | - Leonardo Scapozza
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| |
Collapse
|
43
|
Barnard AM, Willcocks RJ, Finanger EL, Daniels MJ, Triplett WT, Rooney WD, Lott DJ, Forbes SC, Wang DJ, Senesac CR, Harrington AT, Finkel RS, Russman BS, Byrne BJ, Tennekoon GI, Walter GA, Sweeney HL, Vandenborne K. Skeletal muscle magnetic resonance biomarkers correlate with function and sentinel events in Duchenne muscular dystrophy. PLoS One 2018; 13:e0194283. [PMID: 29554116 PMCID: PMC5858773 DOI: 10.1371/journal.pone.0194283] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/28/2018] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To provide evidence for quantitative magnetic resonance (qMR) biomarkers in Duchenne muscular dystrophy by investigating the relationship between qMR measures of lower extremity muscle pathology and functional endpoints in a large ambulatory cohort using a multicenter study design. METHODS MR spectroscopy and quantitative imaging were implemented to measure intramuscular fat fraction and the transverse magnetization relaxation time constant (T2) in lower extremity muscles of 136 participants with Duchenne muscular dystrophy. Measures were collected at 554 visits over 48 months at one of three imaging sites. Fat fraction was measured in the soleus and vastus lateralis using MR spectroscopy, while T2 was assessed using MRI in eight lower extremity muscles. Ambulatory function was measured using the 10m walk/run, climb four stairs, supine to stand, and six minute walk tests. RESULTS Significant correlations were found between all qMR and functional measures. Vastus lateralis qMR measures correlated most strongly to functional endpoints (|ρ| = 0.68-0.78), although measures in other rapidly progressing muscles including the biceps femoris (|ρ| = 0.63-0.73) and peroneals (|ρ| = 0.59-0.72) also showed strong correlations. Quantitative MR biomarkers were excellent indicators of loss of functional ability and correlated with qualitative measures of function. A VL FF of 0.40 was an approximate lower threshold of muscle pathology associated with loss of ambulation. DISCUSSION Lower extremity qMR biomarkers have a robust relationship to clinically meaningful measures of ambulatory function in Duchenne muscular dystrophy. These results provide strong supporting evidence for qMR biomarkers and set the stage for their potential use as surrogate outcomes in clinical trials.
Collapse
Affiliation(s)
- Alison M. Barnard
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Rebecca J. Willcocks
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Erika L. Finanger
- Departments of Pediatrics and Neurology, Oregon Health & Science University, Portland, OR, United States of America
| | - Michael J. Daniels
- Department of Statistics, University of Florida, Gainesville, FL, United States of America
| | - William T. Triplett
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - William D. Rooney
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, United States of America
| | - Donovan J. Lott
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Sean C. Forbes
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Dah-Jyuu Wang
- Department of Radiology, Division of Neurology, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Claudia R. Senesac
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
| | - Ann T. Harrington
- The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | | | - Barry S. Russman
- Departments of Pediatrics and Neurology, Oregon Health & Science University, Portland, OR, United States of America
| | - Barry J. Byrne
- Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center, University of Florida, Gainesville, FL, United States of America
| | - Gihan I. Tennekoon
- The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Glenn A. Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States of America
| | - H. Lee Sweeney
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, United States of America
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, FL, United States of America
- * E-mail:
| |
Collapse
|
44
|
Lozanoska-Ochser B, Benedetti A, Rizzo G, Marrocco V, Di Maggio R, Fiore P, Bouche M. Targeting early PKCθ-dependent T-cell infiltration of dystrophic muscle reduces disease severity in a mouse model of muscular dystrophy. J Pathol 2018; 244:323-333. [PMID: 29214629 DOI: 10.1002/path.5016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/09/2017] [Accepted: 11/30/2017] [Indexed: 12/17/2022]
Abstract
Chronic muscle inflammation is a critical feature of Duchenne muscular dystrophy and contributes to muscle fibre injury and disease progression. Although previous studies have implicated T cells in the development of muscle fibrosis, little is known about their role during the early stages of muscular dystrophy. Here, we show that T cells are among the first cells to infiltrate mdx mouse dystrophic muscle, prior to the onset of necrosis, suggesting an important role in early disease pathogenesis. Based on our comprehensive analysis of the kinetics of the immune response, we further identify the early pre-necrotic stage of muscular dystrophy as the relevant time frame for T-cell-based interventions. We focused on protein kinase C θ (PKCθ, encoded by Prkcq), a critical regulator of effector T-cell activation, as a potential target to inhibit T-cell activity in dystrophic muscle. Lack of PKCθ not only reduced the frequency and number of infiltrating T cells but also led to quantitative and qualitative changes in the innate immune cell infiltrate in mdx/Prkcq-/- muscle. These changes were due to the inhibition of T cells, since PKCθ was necessary for T-cell but not for myeloid cell infiltration of acutely injured muscle. Targeting T cells with a PKCθ inhibitor early in the disease process markedly diminished the size of the inflammatory cell infiltrate and resulted in reduced muscle damage. Moreover, diaphragm necrosis and fibrosis were also reduced following treatment. Overall, our findings identify the early T-cell infiltrate as a therapeutic target and highlight the potential of PKCθ inhibition as a therapeutic approach to muscular dystrophy. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Biliana Lozanoska-Ochser
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences (DAHFMO), Sapienza University of Rome, Rome, Italy
| | - Anna Benedetti
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences (DAHFMO), Sapienza University of Rome, Rome, Italy
| | - Giuseppe Rizzo
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences (DAHFMO), Sapienza University of Rome, Rome, Italy
| | - Valeria Marrocco
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences (DAHFMO), Sapienza University of Rome, Rome, Italy
| | - Rosanna Di Maggio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences (DAHFMO), Sapienza University of Rome, Rome, Italy
| | - Piera Fiore
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences (DAHFMO), Sapienza University of Rome, Rome, Italy
| | - Marina Bouche
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences (DAHFMO), Sapienza University of Rome, Rome, Italy
| |
Collapse
|
45
|
Forsythe E, Kenny J, Bacchelli C, Beales PL. Managing Bardet-Biedl Syndrome-Now and in the Future. Front Pediatr 2018; 6:23. [PMID: 29487844 PMCID: PMC5816783 DOI: 10.3389/fped.2018.00023] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 01/25/2018] [Indexed: 11/13/2022] Open
Abstract
Bardet-Biedl syndrome is a rare autosomal recessive multisystem disorder caused by defects in genes encoding for proteins that localize to the primary cilium/basal body complex. Twenty-one disease-causing genes have been identified to date. It is one of the most well-studied conditions in the family of diseases caused by defective cilia collectively known as ciliopathies. In this review, we provide an update on diagnostic developments, clinical features, and progress in the management of Bardet-Biedl syndrome. Advances in diagnostic technologies including exome and whole genome sequencing are expanding the spectrum of patients who are diagnosed with Bardet-Biedl syndrome and increasing the number of cases with diagnostic uncertainty. As a result of the diagnostic developments, a small number of patients with only one or two clinical features of Bardet-Biedl syndrome are being diagnosed. Our understanding of the syndrome-associated renal disease has evolved and is reviewed here. Novel interventions are developing at a rapid pace and are explored in this review including genetic therapeutics such as gene therapy, exon skipping therapy, nonsense suppression therapy, and gene editing. Other non-genetic therapies such as gene repurposing, targeted therapies, and non-pharmacological interventions are also discussed.
Collapse
Affiliation(s)
- Elizabeth Forsythe
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Joanna Kenny
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Chiara Bacchelli
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Philip L Beales
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
46
|
Welch RD, Billon C, Valfort AC, Burris TP, Flaveny CA. Pharmacological inhibition of REV-ERB stimulates differentiation, inhibits turnover and reduces fibrosis in dystrophic muscle. Sci Rep 2017; 7:17142. [PMID: 29215066 PMCID: PMC5719458 DOI: 10.1038/s41598-017-17496-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/27/2017] [Indexed: 12/21/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a debilitating X-linked disorder that is fatal. DMD patients lack the expression of the structural protein dystrophin caused by mutations within the DMD gene. The absence of functional dystrophin protein results in excessive damage from normal muscle use due to the compromised structural integrity of the dystrophin associated glycoprotein complex. As a result, DMD patients exhibit ongoing cycles of muscle destruction and regeneration that promote inflammation, fibrosis, mitochondrial dysfunction, satellite cell (SC) exhaustion and loss of skeletal and cardiac muscle function. The nuclear receptor REV-ERB suppresses myoblast differentiation and recently we have demonstrated that the REV-ERB antagonist, SR8278, stimulates muscle regeneration after acute injury. Therefore, we decided to explore whether the REV-ERB antagonist SR8278 could slow the progression of muscular dystrophy. In mdx mice SR8278 increased lean mass and muscle function, and decreased muscle fibrosis and muscle protein degradation. Interestingly, we also found that SR8278 increased the SC pool through stimulation of Notch and Wnt signaling. These results suggest that REV-ERB is a potent target for the treatment of DMD.
Collapse
Affiliation(s)
- Ryan D Welch
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA
| | - Cyrielle Billon
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA
| | - Aurore-Cecile Valfort
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA
| | - Thomas P Burris
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA
| | - Colin A Flaveny
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, MO, 63104, USA.
| |
Collapse
|
47
|
McMorran BJ, Miceli MC, Baum LG. Lectin-binding characterizes the healthy human skeletal muscle glycophenotype and identifies disease-specific changes in dystrophic muscle. Glycobiology 2017; 27:1134-1143. [PMID: 28973355 PMCID: PMC6283322 DOI: 10.1093/glycob/cwx073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/08/2017] [Accepted: 08/21/2017] [Indexed: 12/27/2022] Open
Abstract
Our understanding of muscle glycosylation to date has derived from studies in mouse models and a limited number of human lectin histochemistry studies. As various therapeutic approaches aimed at treating patients with muscular dystrophies are being translated from rodent models to human, it is critical to better understand human muscle glycosylation and relevant disease-specific differences between healthy and dystrophic muscle. Here, we report the first quantitative characterization of human muscle glycosylation, and identify differentiation- and disease-specific differences in human muscle glycosylation. Utilizing a panel of 13 lectins with varying glycan specificities, we surveyed lectin binding to primary and immortalized myoblasts and myotubes from healthy and dystrophic sources. Following differentiation of primary and immortalized healthy human muscle cells, we observed increased binding of Narcissus pseudonarcissus agglutinin (NPA), PNA, MAA-II and WFA to myotubes compared to myoblasts. Following differentiation of immortalized healthy and dystrophic human muscle cells, we observed disease-specific differences in binding of NPA, Jac and Tricosanthes japonica agglutinin-I (TJA-I) to differentiated myotubes. We also observed differentiation- and disease-specific differences in binding of NPA, Jac, PNA, TJA-I and WFA to glycoprotein receptors in muscle cells. Additionally, Jac, PNA and WFA precipitated functionally glycosylated α-DG, that bound laminin, while NPA and TJA-I did not. Lectin histochemistry of healthy and dystrophic human muscle sections identified disease-specific differences in binding of O-glycan and sialic acid-specific lectins between healthy and dystrophic muscle. These results indicate that specific and discrete changes in glycosylation occur following differentiation, and identify specific lectins as potential biomarkers sensitive to changes in healthy human muscle glycosylation.
Collapse
Affiliation(s)
- Brian J McMorran
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Ave., CHS 14-127, Los Angeles, CA 90095, USA
| | - M Carrie Miceli
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, 609 Charles E Young Dr E, Los Angeles, CA 90095, USA
| | - Linda G Baum
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Ave., CHS 14-127, Los Angeles, CA 90095, USA
| |
Collapse
|
48
|
Guimbellot J, Sharma J, Rowe SM. Toward inclusive therapy with CFTR modulators: Progress and challenges. Pediatr Pulmonol 2017; 52:S4-S14. [PMID: 28881097 PMCID: PMC6208153 DOI: 10.1002/ppul.23773] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/29/2017] [Indexed: 12/29/2022]
Abstract
Cystic fibrosis is caused by gene mutations that result in an abnormal Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) protein on the surface of cells. CFTR modulators are a novel class of drugs that directly target the molecular defect. CFTR modulators include potentiators that result in improved activity of the channel; correctors that help the protein traffic to the cell surface properly; and readthrough agents that restore full-length CFTR by suppression of premature termination codons, among other novel classes more recently established. While some of these drugs, CFTR potentiators in particular, have provided remarkable improvements for CF patients, others have yet to achieve profoundly improved outcomes, and many CF patients are not yet impacted by CFTR modulators due to lack of knowledge regarding susceptibility of their mutations to treatment. One limitation to expanding these types of therapies to the maximum number of patients with CF is the lack of rigorously validated clinical biomarkers that can determine efficacy on an individual basis, as well as few pre-clinical tools that can predict whether an individual with a rare combination of mutant alleles will respond to a particular CFTR modulator regimen. In this review, we discuss the various groups of CFTR modulators and their status in clinical development, as well as address the current literature on biomarkers, pre-clinical cell-based tools, and the role of pharmacometrics in creating therapeutic strategies to improve the lives of all patients with cystic fibrosis, regardless of their specific mutation.
Collapse
Affiliation(s)
- Jennifer Guimbellot
- Departments of Pediatrics, The University of Alabama at Birmingham, Birmingham, Alabama
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Jyoti Sharma
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, Alabama
- Cell Developmental and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Steven M. Rowe
- Departments of Pediatrics, The University of Alabama at Birmingham, Birmingham, Alabama
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, Alabama
- Cell Developmental and Integrative Biology, The University of Alabama at Birmingham, Birmingham, Alabama
- Departments of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
49
|
Irwin AN, Herink MC. Eteplirsen for the Treatment of Duchenne Muscular Dystrophy: Quality of Evidence Concerns-an Alternative Viewpoint. Pharmacotherapy 2017; 37:e109-e111. [PMID: 28741800 DOI: 10.1002/phar.1996] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Adriane N Irwin
- College of Pharmacy, Oregon State University/Oregon Health and Science University, Corvallis, Oregon
| | - Megan C Herink
- College of Pharmacy, Oregon State University/Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
50
|
Abstract
This commentary will focus on how we can use our knowledge about the complexity of human disease and its pathogenesis to identify novel approaches to therapy. We know that even for single gene Mendelian disorders, patients with identical mutations often have different presentations and outcomes. This lack of genotype-phenotype correlation led us and others to examine the roles of modifier genes in the context of biological networks. These investigations have utilized vertebrate and invertebrate model organisms. Since one of the goals of research on modifier genes and networks is to identify novel therapeutic targets, the challenges to patient access and compliance because of the high costs of medications for rare genetic diseases must be recognized. A recent article explored protective modifiers, including plastin 3 (PLS3) and coronin 1C (CORO1C), in spinal muscular atrophy (SMA). SMA is an autosomal recessive deficit of survival motor neuron protein (SMN) caused by mutations in SMN1. However, the severity of SMA is determined primarily by the number of SMN2 copies, and this results in significant phenotypic variability. PLS3 was upregulated in siblings who were asymptomatic compared with those who had SMA2 or SMA3, but identical homozygous SMN1 deletions and equal numbers of SMN2 copies. CORO1C was identified by interrogation of the PLS3 interactome. Overexpression of these proteins rescued endocytosis in SMA models. In addition, antisense RNA for upregulation of SMN2 protein expression is being developed as another way of modifying the SMA phenotype. These investigations suggest the practical application of protective modifiers to rescue SMA phenotypes. Other examples of the potential therapeutic value of novel protective modifiers will be discussed, including in Duchenne muscular dystrophy and glycerol kinase deficiency. This work shows that while we live in an exciting era of genomic sequencing, a functional understanding of biology, the impact of its disruption, and possibilities for its repair have never been more important as we search for new therapies.
Collapse
Affiliation(s)
- Edward R B McCabe
- March of Dimes Foundation, United States; Department of Pediatrics, David Geffen School of Medicine at UCLA, United States.
| |
Collapse
|