1
|
Muntoni F, Goemans N, Posner N, Signorovitch J, Johnson M, He C, Dorling P, Beaverson K, Alvir J, Mahn M, Ward SJ, McDonald CM, Mercuri E. Characterization of patients with Duchenne muscular dystrophy across previously developed health states. PLoS One 2024; 19:e0307118. [PMID: 39475941 PMCID: PMC11524485 DOI: 10.1371/journal.pone.0307118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/29/2024] [Indexed: 11/02/2024] Open
Abstract
Project HERCULES has developed a natural history model (NHM) of disease progression in Duchenne muscular dystrophy (DMD) that comprises eight ordered health states (two ambulatory states, one transfer state indicating increased caregiver burden in which patients cannot walk/run 10m or rise from floor but can still support their own weight, and five non-ambulatory states). The current study used data from nine sources (clinical trial placebo arms, one real-world dataset, and three natural history datasets) to further characterize patients with DMD according to these health states. The study included 1,173 patients across 5,306 visits. Patients were on average older and exhibited worse ambulatory, pulmonary, upper limb, and cardiac functions with each successive health state. Mean±SE ages increased monotonically across health states, starting with 8.47±0.07 for early ambulatory, 10.86±0.13 for late ambulatory, 11.65±0.35 for transfer state, and ranging from 13.17±0.32 to 16.84±0.37 for the non-ambulatory states. North Star Ambulatory Assessment (NSAA) total score, which measures motor function and ranges from 34 (best) to 0 (worst), was 23.7 (interquartile range [IQR]: 20-30) for early ambulatory patients, 12.7 (IQR: 9-16) for late ambulatory patients, and 3.9 (IQR: 2-5) for transfer patients. Pulmonary function as measured by mean±SE of forced vital capacity percent predicted (FVC%p) was 94.5±0.8 for early ambulatory, 89.1±1.4 for late ambulatory, and 80.2±2.8 for transfer states, and decreased from 77.2±1.7 to 20.6±1.6 across the five non-ambulatory health states. In summary, these findings further characterize health states and their interpretation in economic modeling and decision-making in DMD management.
Collapse
Affiliation(s)
- Francesco Muntoni
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, London, United Kingdom
| | | | - Nate Posner
- Pfizer, Inc., New York, NY, United States of America
| | - James Signorovitch
- Analysis Group, Inc., Boston, MA, United States of America
- collaborative Trajectory Analysis Project (cTAP), Cambridge, MA, United States of America
| | | | - Chujun He
- Analysis Group, Inc., Boston, MA, United States of America
| | | | | | - Jose Alvir
- Pfizer, Inc., New York, NY, United States of America
| | - Matthias Mahn
- Pfizer, Inc., New York, NY, United States of America
| | - Susan J. Ward
- collaborative Trajectory Analysis Project (cTAP), Cambridge, MA, United States of America
| | - Craig M. McDonald
- Department of Physical Medicine and Rehabilitation, University of California, Davis, Sacramento, CA, United States of America
| | - Eugenio Mercuri
- Pediatric Neurology, Catholic University, Rome, Italy
- Centro Clinico NeMO, Fondazione Policlinico Gemelli IRCCS, Rome, Italy
| | | |
Collapse
|
2
|
Budzynska K, Bozyk KT, Jarosinska K, Ziemiecka A, Siemionow K, Siemionow M. Developing Advanced Chimeric Cell Therapy for Duchenne Muscular Dystrophy. Int J Mol Sci 2024; 25:10947. [PMID: 39456730 PMCID: PMC11507628 DOI: 10.3390/ijms252010947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/06/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is a lethal, X-linked disorder leading to muscle degeneration and premature death due to cardiopulmonary complications. Currently, there is no cure for DMD. We previously confirmed the efficacy of human Dystrophin-Expressing Chimeric (DEC) cells created via the fusion of myoblasts from normal and DMD-affected donors. The current study aimed to optimize the development of DEC therapy via the polyethylene glycol (PEG)-mediated fusion protocol of human myoblasts derived from normal, unrelated donors. The optimization of cell fusion assessed different factors influencing fusion efficacy, including myoblast passage number, the efficacy of PKH myoblast staining, the ratio of the single-stained myoblasts in the MIX, and PEG administration time. Additionally, the effect of PEG fusion procedure on cell viability was assessed. A correlation was found between the number of cells used for PKH staining and staining efficacy. Furthermore, the ratio of single-stained myoblasts in the MIX and PEG administration time correlated with fusion efficacy. There was no correlation found between the myoblast passage number and fusion efficacy. This study successfully optimized the myoblast fusion protocol for creation of human DEC cells, introducing DEC as a new Advanced Therapy Medicinal Product (ATMP) for DMD patients.
Collapse
Affiliation(s)
- Katarzyna Budzynska
- Dystrogen Therapeutics Technology Polska sp. z o.o., 00-777 Warsaw, Poland; (K.B.); (K.T.B.); (K.J.); (A.Z.); (K.S.)
| | - Katarzyna T. Bozyk
- Dystrogen Therapeutics Technology Polska sp. z o.o., 00-777 Warsaw, Poland; (K.B.); (K.T.B.); (K.J.); (A.Z.); (K.S.)
| | - Klaudia Jarosinska
- Dystrogen Therapeutics Technology Polska sp. z o.o., 00-777 Warsaw, Poland; (K.B.); (K.T.B.); (K.J.); (A.Z.); (K.S.)
| | - Anna Ziemiecka
- Dystrogen Therapeutics Technology Polska sp. z o.o., 00-777 Warsaw, Poland; (K.B.); (K.T.B.); (K.J.); (A.Z.); (K.S.)
| | - Krzysztof Siemionow
- Dystrogen Therapeutics Technology Polska sp. z o.o., 00-777 Warsaw, Poland; (K.B.); (K.T.B.); (K.J.); (A.Z.); (K.S.)
| | - Maria Siemionow
- Dystrogen Therapeutics Technology Polska sp. z o.o., 00-777 Warsaw, Poland; (K.B.); (K.T.B.); (K.J.); (A.Z.); (K.S.)
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL 60607, USA
- Chair and Department of Traumatology, Orthopaedics, and Surgery of the Hand, Poznan University of Medical Sciences, 61-545 Poznan, Poland
| |
Collapse
|
3
|
Fontaine Carbonnel S, Dabaj I, de Montferrand C, Rippert P, Laugel V, De Lucia S, Ravelli C, Seferian A, Ropars J, Cances C. Choice of compound, dosage, and management of side effects for long-term corticosteroid treatment in Duchenne muscular dystrophy: Guidelines from the Neuromuscular Commission of the French Society of Pediatric Neurology. Arch Pediatr 2024; 31:410-418. [PMID: 39332946 DOI: 10.1016/j.arcped.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 05/26/2024] [Indexed: 09/29/2024]
Abstract
The French Society of Pediatric Neurology and the FILNEMUS network created a working group on corticosteroid therapy in children with Duchenne muscular dystrophy in order to analyze the literature review and current French practices. The aim of this work was to produce guidelines regarding treatment initiation, pre-therapeutic interventions, choice between available compounds, and treatment monitoring (dosage, duration, and discontinuation). The treatment side effects and their management are also detailed: osteoporosis, endocrinological anomaly (growth delay, weight gain, pubertal delay), cataract, arterial hypertension, behavioral disorders, management of immunosuppression and vaccines, and management of gastrointestinal and metabolic complications.
Collapse
Affiliation(s)
- Stéphanie Fontaine Carbonnel
- Department of Pediatric Physical Medicine and Rehabilitation, Hôpital Mère Enfant, Hospices Civils de Lyon, 59 Boulevard Pinel, Bron F-69677, CEDEX, France.
| | - Ivana Dabaj
- UNIROUEN, Inserm U1245, Department of Neonatology, Pediatric Intensive Care and Neuropediatrics, Normandie University, CHU Rouen F-76000, Rouen, France
| | - Camille de Montferrand
- Department of Pediatric Physical Medicine and Rehabilitation, Hôpital Mère Enfant, Hospices Civils de Lyon, 59 Boulevard Pinel, Bron F-69677, CEDEX, France
| | - Pascal Rippert
- Service Recherche et Epidémiologie Cliniques, Hospices Civils de Lyon, Pôle Santé Publique, 162 Avenue Lacassagne, Lyon F-69003, France
| | - Vincent Laugel
- Department of pediatric neurology, Strasbourg University Hospital, 1 avenue Moliere, Strasbourg F-67098, France
| | - Silvana De Lucia
- Assistance Publique Hôpitaux de Paris, Sorbonne Université, Institut de Myologie, AFM-Téléthon, Essais cliniques I-Motion Enfants, Hôpital Armand Trousseau, Paris F-75012, France
| | - Claudia Ravelli
- Assistance Publique Hôpitaux de Paris, Sorbonne Université, Institut de Myologie, AFM-Téléthon, Essais cliniques I-Motion Enfants, Hôpital Armand Trousseau, Paris F-75012, France
| | - Andreea Seferian
- Assistance Publique Hôpitaux de Paris, Sorbonne Université, Institut de Myologie, AFM-Téléthon, Essais cliniques I-Motion Enfants, Hôpital Armand Trousseau, Paris F-75012, France
| | - Juliette Ropars
- Service de pédiatrie, CHU Brest, 2 Avenue Foch, Brest F-29609, France
| | - Claude Cances
- Unit of Pediatric Neurology, French Greater South-West Reference Center for Neuromuscular Diseases, Hôpital des Enfants, University Hospital Center Toulouse, 330 Avenue de Grande Bretagne-TSA, Toulouse Cedex F-31059, France
| |
Collapse
|
4
|
Landfeldt E, Alemán A, Abner S, Zhang R, Werner C, Tomazos I, Lochmüller H, Quinlivan RM, Wahbi K. Predictors of cardiac disease in duchenne muscular dystrophy: a systematic review and evidence grading. Orphanet J Rare Dis 2024; 19:359. [PMID: 39342355 PMCID: PMC11439250 DOI: 10.1186/s13023-024-03372-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a rare disease that causes progressive muscle degeneration resulting in life-threatening cardiac complications. The objective of this systematic literature review was to describe and grade the published evidence of predictors of cardiac disease in DMD. METHODS The review encompassed searches of Embase, MEDLINE ALL, and the Cochrane Database of Systematic Reviews from January 1, 2000, to December 31, 2022, for predictors of cardiac disease in DMD. The certainty of evidence (i.e., very low to high) was assessed using the Grading of Recommendations, Assessment, Development and Evaluations (GRADE) framework. RESULTS We included 33 publications encompassing 9,232 patients with DMD. We found moderate- to high-quality evidence that cardiac medication (i.e., ACE inhibitors [enalapril and perindopril], β-blockers [carvedilol], and mineralocorticoid receptor antagonists [eplerenone]) are significantly associated with preserved left ventricular ejection fraction (LVEF), left ventricular end-systolic volume (LVESV), and left ventricular circumferential strain (LVCS). DMD mutations in exons 51 and 52 were found to be significantly associated with lower risk of cardiomyopathy; deletions treatable by exon 53 skipping and mutations in the Dp116 coding region with improved LVEF and prolonged cardiac dysfunction-free survival; and exons 45-50 and 52 with early left ventricular systolic dysfunction (low/very low-quality evidence). We found high-quality evidence that glucocorticoids (deflazacort) are significantly associated with preserved LVEF and improved fractional shortening (FS), and low-quality evidence that glucocorticoids (deflazacort, prednisone, and/or prednisolone) are associated with improved ejection fraction (EF) and lower risk of cardiomyopathy, ventricular dysfunction, and heart failure-related mortality. Full-time mechanical ventilation was found to be significantly correlated with LVEF (low-quality evidence), muscle strength with FS (low-quality evidence), and genetic modifiers (i.e., LTBP4 rs10880 and ACTN3) with LVEF, lower risk of cardiomyopathy and left ventricular dilation (low-quality evidence). CONCLUSION Several sources of cardiac disease heterogeneity are well-studied in patients with DMD. Yet, the certainty of evidence is generally low, and little is known of the contribution of non-pharmacological interventions, as well as the impact of different criteria for initiation of specific treatments. Our findings help raise awareness of prevailing unmet needs, shape expectations of treatment outcomes, and inform the design of future research.
Collapse
Affiliation(s)
- Erik Landfeldt
- IQVIA, Pyramidvägen 7, 169 56, Solna, Stockholm, Sweden.
| | - Alberto Alemán
- Division of Neurology, Department of Pediatrics, Children's Hospital of Eastern Ontario, Research Institute, University of Ottawa, Ottawa, ON, Canada
- Division of Neurology, Department of Medicine, The Ottawa Hospital, Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | | | | | | | | | - Hanns Lochmüller
- Division of Neurology, Department of Pediatrics, Children's Hospital of Eastern Ontario, Research Institute, University of Ottawa, Ottawa, ON, Canada
- Division of Neurology, Department of Medicine, The Ottawa Hospital, Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Ros M Quinlivan
- Centre for Neuromuscular Diseases, UCL Institute of Neurology, National Hospital, London, UK
| | - Karim Wahbi
- Cardiology Department, AP-HP, Cochin Hospital, Paris, France
- Université de Paris, Paris, France
| |
Collapse
|
5
|
Siemionow M, Ziemiecka A, Bożyk K, Siemionow K. Mechanisms of Chimeric Cell Therapy in Duchenne Muscular Dystrophy. Biomedicines 2024; 12:1996. [PMID: 39335509 PMCID: PMC11428244 DOI: 10.3390/biomedicines12091996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/21/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
Despite scientific efforts, there is no cure for Duchenne muscular dystrophy (DMD), a lethal, progressive, X-linked genetic disorder caused by mutations in the dystrophin gene. DMD leads to cardiac and skeletal muscle weakness, resulting in premature death due to cardio-pulmonary complications. We have developed Dystrophin Expressing Chimeric (DEC) cell therapy, DT-DEC01, by fusing human myoblasts from healthy donors and from DMD patients. Preclinical studies on human DEC cells showed increased dystrophin expression and improved cardiac, pulmonary, and skeletal muscle function after intraosseous administration. Our clinical study confirmed the safety and efficacy of DT-DEC01 therapy up to 24 months post-administration. In this study, we conducted in vitro assays to test the composition and potency of DT-DEC01, assessing chimerism level and the presence of dystrophin, desmin, and myosin heavy chain. Myoblast fusion resulted in the transfer of healthy donor mitochondria and the creation of chimeric mitochondria within DT-DEC01. The Pappenheim assay confirmed myotube formation in the final product. This study highlights the unique properties of DT-DEC01 therapy and their relevance to DMD treatment mechanisms.
Collapse
Affiliation(s)
- Maria Siemionow
- Dystrogen Therapeutics Technology Polska sp. z o.o., 00-777 Warsaw, Poland
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL 60612, USA
- Chair and Department of Traumatology, Orthopedics and Surgery of the Hand, Poznan University of Medical Sciences, 61-545 Poznan, Poland
| | - Anna Ziemiecka
- Dystrogen Therapeutics Technology Polska sp. z o.o., 00-777 Warsaw, Poland
| | - Katarzyna Bożyk
- Dystrogen Therapeutics Technology Polska sp. z o.o., 00-777 Warsaw, Poland
| | - Krzysztof Siemionow
- Dystrogen Therapeutics Technology Polska sp. z o.o., 00-777 Warsaw, Poland
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
6
|
Schiava M, Lofra RM, Bourke JP, James MK, Díaz-Manera J, Elseed MA, Michel-Sodhi J, Moat D, Mccallum M, Mayhew A, Ghimenton E, Díaz CFB, Malinova M, Wong K, Richardson M, Tasca G, Grover E, Robinson EJ, Tanner S, Eglon G, Behar L, Eagle M, Turner C, Verdú-Díaz J, Heslop E, Straub V, Bettolo CM, Guglieri M. Disease-associated comorbidities, medication records and anthropometric measures in adults with Duchenne muscular dystrophy. Neuromuscul Disord 2024; 41:8-19. [PMID: 38865917 DOI: 10.1016/j.nmd.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024]
Abstract
We investigated the comorbidities, associated factors, and the relationship between anthropometric measures and respiratory function and functional abilities in adults with Duchenne muscular dystrophy (DMD). This was a single-centre cross-sectional study in genetically diagnosed adults with DMD (>16 years old). Univariate and multivariate analyses identified factors associated with dysphagia, constipation, Body Mass Index (BMI), and weight. Regression analysis explored associations between BMI, weight, and respiratory/motor abilities. We included 112 individuals (23.4 ± 5.2 years old), glucocorticoid-treated 66.1 %. The comorbidities frequency was 61.6 % scoliosis (61.0 % of them had spinal surgery), 36.6 % dysphagia, 36.6 % constipation, and 27.8 % urinary conditions. The use of glucocorticoids delayed the time to spinal surgery. The univariate analysis revealed associations between dysphagia and constipation with age, lack of glucocorticoid treatment, and lower respiratory and motor function. In the multivariate analysis, impaired cough ability remained as the factor consistently linked to both conditions. Constipation associated with lower BMI and weight. BMI and weight positively correlated with respiratory parameters, but they did not associate with functional abilities. Glucocorticoids reduce the frequency of comorbidities in adults with DMD. The ability to cough can help identifying dysphagia and constipation. Lower BMI and weight in individuals with DMD with compromised respiratory function may suggest a higher calories requirement.
Collapse
Affiliation(s)
- Marianela Schiava
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Robert Muni Lofra
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - John P Bourke
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Meredith K James
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Jordi Díaz-Manera
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Maha A Elseed
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Jassi Michel-Sodhi
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Dionne Moat
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Michelle Mccallum
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Anna Mayhew
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Elisabetta Ghimenton
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Carla Florencia Bolaño Díaz
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Monika Malinova
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Karen Wong
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Mark Richardson
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Giorgio Tasca
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Emma Grover
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Emma-Jayne Robinson
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Stephanie Tanner
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Gail Eglon
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Laura Behar
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | | | - Catherine Turner
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - José Verdú-Díaz
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Emma Heslop
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Chiara Marini Bettolo
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK
| | - Michela Guglieri
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne, UK.
| |
Collapse
|
7
|
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
|
8
|
Benemei S, Gatto F, Boni L, Pane M. "If you cannot measure it, you cannot improve it". Outcome measures in Duchenne Muscular Dystrophy: current and future perspectives. Acta Neurol Belg 2024:10.1007/s13760-024-02600-2. [PMID: 39080230 DOI: 10.1007/s13760-024-02600-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/11/2024] [Indexed: 09/04/2024]
Abstract
Duchenne Muscular Dystrophy (DMD) is an X-linked recessive neuromuscular disorder primarily affecting males, caused by mutations in the dystrophin gene. The absence of dystrophin protein leads to progressive skeletal muscle degeneration. Recent advances in the therapeutic landscape underscore the need to identify appropriate outcome measures to assess treatment efficacy in ambulant and non-ambulant DMD patients, across clinical and research settings. This is essential for accurately evaluating new treatments and attributing therapeutic benefits.It is crucial to establish a robust correlation between outcome scores and disease progression patterns. This task is challenging since functional test performance may be influenced by different patient's characteristics, including the physiological evolution of the neurodevelopment together with the disease progression. While widely used DMD outcomes such as the North Star Ambulatory Assessment, the 6-Minute Walking Test, the 4 stairs climbed, and the Performance of the Upper Limb exhibit reliability and validity, their clinical significance is influenced by the wide phenotype and progression variability of the disease.We present and discuss the features (relevance, quantifiability, validity, objectivity, reliability, sensitivity, specificity, precision) of available DMD outcome measures, including new potential measures that may be provided by digital tools and artificial intelligence.
Collapse
Affiliation(s)
| | | | - Luca Boni
- U.O. Epidemiologia Clinica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Marika Pane
- Nemo Clinical Centre, Fondazione Policlinico Universitario A. Gemelli IRCSS, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
9
|
Schiava M, Lofra RM, Bourke JP, Díaz‐Manera J, James MK, Elseed MA, Malinova M, Michel‐Sodhi J, Moat D, Ghimenton E, Mccallum M, Díaz CFB, Mayhew A, Wong K, Richardson M, Tasca G, Eglon G, Eagle M, Turner C, Heslop E, Straub V, Bettolo CM, Guglieri M. Functional abilities, respiratory and cardiac function in a large cohort of adults with Duchenne muscular dystrophy treated with glucocorticoids. Eur J Neurol 2024; 31:e16267. [PMID: 38556893 PMCID: PMC11236054 DOI: 10.1111/ene.16267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 01/18/2024] [Accepted: 02/14/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND AND PURPOSE The transition to adult services, and subsequent glucocorticoid management, is critical in adults with Duchenne muscular dystrophy. This study aims (1) to describe treatment, functional abilities, respiratory and cardiac status during transition to adulthood and adult stages; and (2) to explore the association between glucocorticoid treatment after loss of ambulation (LOA) and late-stage clinical outcomes. METHODS This was a retrospective single-centre study on individuals with Duchenne muscular dystrophy (≥16 years old) between 1986 and 2022. Logistic regression, Cox proportional hazards models and survival analyses were conducted utilizing data from clinical records. RESULTS In all, 112 individuals were included. Mean age was 23.4 ± 5.2 years and mean follow-up was 18.5 ± 5.5 years. At last assessment, 47.2% were on glucocorticoids; the mean dose of prednisone was 0.38 ± 0.13 mg/kg/day and of deflazacort 0.43 ± 0.16 mg/kg/day. At age 16 years, motor function limitations included using a manual wheelchair (89.7%), standing (87.9%), transferring from a wheelchair (86.2%) and turning in bed (53.4%); 77.5% had a peak cough flow <270 L/min, 53.3% a forced vital capacity percentage of predicted <50% and 40.3% a left ventricular ejection fraction <50%. Glucocorticoids after LOA reduced the risk and delayed the time to difficulties balancing in the wheelchair, loss of hand to mouth function, forced vital capacity percentage of predicted <30% and forced vital capacity <1 L and were associated with lower frequency of left ventricular ejection fraction <50%, without differences between prednisone and deflazacort. Glucocorticoid dose did not differ by functional, respiratory or cardiac status. CONCLUSION Glucocorticoids after LOA preserve late-stage functional abilities, respiratory and cardiac function. It is suggested using functional abilities, respiratory and cardiac status at transition stages for adult services planning.
Collapse
Affiliation(s)
- Marianela Schiava
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Robert Muni Lofra
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - John P. Bourke
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Jordi Díaz‐Manera
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Meredith K. James
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Maha A. Elseed
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Monika Malinova
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Jassi Michel‐Sodhi
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Dionne Moat
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Elisabetta Ghimenton
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Michelle Mccallum
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Carla Florencia Bolaño Díaz
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Anna Mayhew
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Karen Wong
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Mark Richardson
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Giorgio Tasca
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Gail Eglon
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | | | - Cathy Turner
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Emma Heslop
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Volker Straub
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Chiara Marini Bettolo
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| | - Michela Guglieri
- John Walton Muscular Dystrophy Research CentreNewcastle University and Newcastle Hospitals NHS Foundation TrustsNewcastle Upon TyneUK
| |
Collapse
|
10
|
Qian C, Klimchak AC, Szabo SM, Popoff E, Iannaccone ST, Gooch KL. Observing the Clinical Course of Duchenne Muscular Dystrophy in Medicaid Real-World Healthcare Data. Adv Ther 2024; 41:2519-2530. [PMID: 38698169 PMCID: PMC11133018 DOI: 10.1007/s12325-024-02865-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/03/2024] [Indexed: 05/05/2024]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is a rare, severe progressive neuromuscular disease. Health insurance claims allow characterization of population-level real-world outcomes, based on observed healthcare resource use. An analysis of data specific to those with Medicaid insurance is presently unavailable. The objective was to describe the real-world clinical course of DMD based on claims data from Medicaid-insured individuals in the USA. METHODS Individuals with DMD were identified from the MarketScan Multi-State Medicaid datasets (2013-2018). Diagnosis and procedure codes from healthcare claims were used to characterize the occurrence of DMD-relevant clinical observations; categories were scoliosis, cardiovascular-related, respiratory and severe respiratory-related, and neurologic/neuropsychiatric. Age-restricted analyses were conducted to focus on the ages at which DMD-relevant clinical observations were more likely to be captured, and to better understand the impact of both age and follow-up time. RESULTS Of 2007 patients with DMD identified, median (interquartile range) age at index was 14 (9-20) years, and median follow-up was 3.1 (1.6-4.7) years. Neurologic and neuropsychiatric observations were most frequently identified, among 49.3% of the cohort; followed by cardiovascular (48.5%), respiratory (38.1%), scoliosis (36.3%), and severe respiratory (25.0%). Prevalence estimates for each category were higher when analyzed within age-restricted subgroups; and increased as follow-up time increased. CONCLUSIONS This study is the first to use diagnosis and procedure codes from real-world Medicaid claims to document the clinical course in DMD. Findings were consistent with previously published estimates from commercially insured populations and clinical registries; and contribute to the expanding body of real-world evidence around clinical progression of patients with DMD.
Collapse
Affiliation(s)
- Christina Qian
- Broadstreet Health Economics and Outcomes Research, 201-343 Railway Street, Vancouver, BC, V6A 1A4, Canada
| | - Alexa C Klimchak
- Sarepta Therapeutics, Inc, 215 First Street, Cambridge, MA, 02142, USA
| | - Shelagh M Szabo
- Broadstreet Health Economics and Outcomes Research, 201-343 Railway Street, Vancouver, BC, V6A 1A4, Canada.
| | - Evan Popoff
- Broadstreet Health Economics and Outcomes Research, 201-343 Railway Street, Vancouver, BC, V6A 1A4, Canada
| | - Susan T Iannaccone
- University of Texas Southwestern, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Katherine L Gooch
- Sarepta Therapeutics, Inc, 215 First Street, Cambridge, MA, 02142, USA
| |
Collapse
|
11
|
Schiava M, McDermott MP, Broomfield J, Abrams KR, Mayhew AG, McDonald CM, Martens WB, Gregory SJ, Griggs RC, Guglieri M. Factors Associated With Early Motor Function Trajectories in DMD After Glucocorticoid Initiation: Post Hoc Analysis of the FOR-DMD Trial. Neurology 2024; 102:e209206. [PMID: 38710006 DOI: 10.1212/wnl.0000000000209206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Clinical trials in Duchenne muscular dystrophy (DMD) require 3-6 months of stable glucocorticoids, and the primary outcome is explored at 48-52 weeks. The factors that influence the clinical outcome assessment (COA) trajectories soon after glucocorticoid initiation are relevant for the design and analysis of clinical trials of novel drugs. We describe early COA trajectories, associated factors, and the time from glucocorticoid initiation to COA peak. METHODS This was a prospective 18-month analysis of the Finding the Optimum Corticosteroid Regimen for Duchenne Muscular Dystrophy study. Four COAs were investigated: rise from supine velocity (RFV), 10-meter walk/run velocity (10MWRV), North Star Ambulatory Assessment (NSAA) total score, and 6-minute walk test distance (6MWT). The relationships of baseline age (4-5 vs 6-7 years), COA baseline performance, genotype, and glucocorticoid regimen (daily vs intermittent) with the COA trajectories were evaluated using linear mixed-effects models. RESULTS One hundred ninety-six glucocorticoid-naïve boys with DMD aged 4-7 years were enrolled. The mean age at baseline was 5.9 ± 1.0 years, 66% (n = 130) were on daily regimens, 55% (n = 107) showed a 6MWT distance >330 metres; 41% (n = 78) showed RFV >0.2 rise/s; 76% (n = 149) showed 10MWRV >0.142 10m/s, and 41.0% (n = 79) showed NSAA total score >22 points. Mean COA trajectories differed by age at glucocorticoid initiation (p < 0.01 for RFV, 10MWRV, and NSAA; p < 0.05 for 6MWT) and regimen (p < 0.01 for RFV, 10MWRV, and NSAA). Boys younger than 6 years reached their peak performance 12-18 months after glucocorticoid initiation. Boys aged 6 years or older on a daily regimen peaked between months 9 and 12 and those on an intermittent regimen by 9 months. The baseline COA performance was associated with the NSAA (p < 0.01) and the 6MWT trajectory in boys younger than 6 years on a daily regimen (p < 0.01). Differences in the mean trajectories by genotype were not significant. DISCUSSION Glucocorticoid regimen, age, duration of glucocorticoid exposure, and baseline COA performance need to be considered in the design and analysis of clinical trials in young boys with DMD.
Collapse
Affiliation(s)
- Marianela Schiava
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Michael P McDermott
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Jonathan Broomfield
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Keith R Abrams
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Anna G Mayhew
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Craig M McDonald
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - William B Martens
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Stephanie J Gregory
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Robert C Griggs
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| | - Michela Guglieri
- From the John Walton Muscular Dystrophy Research Centre (M.S., A.G.M., M.G.), Clinical and Translational Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, United Kingdom; Department of Biostatistics and Computational Biology (M.P.M.); Department of Neurology, University of Rochester Medical Centre, NY; Department of Health Sciences (J.B., K.R.A.), University of Leicester, United Kingdom; Department of Physical Medicine and Rehabilitation (C.M.M.), University of California, Davis, Sacramento; and Department of Neurology (W.B.M., S.J.G., R.C.G.), University of Rochester Medical Centre, NY
| |
Collapse
|
12
|
Gatto F, Benemei S, Piluso G, Bello L. The complex landscape of DMD mutations: moving towards personalized medicine. Front Genet 2024; 15:1360224. [PMID: 38596212 PMCID: PMC11002111 DOI: 10.3389/fgene.2024.1360224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/26/2024] [Indexed: 04/11/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe genetic disorder characterized by progressive muscle degeneration, with respiratory and cardiac complications, caused by mutations in the DMD gene, encoding the protein dystrophin. Various DMD mutations result in different phenotypes and disease severity. Understanding genotype/phenotype correlations is essential to optimize clinical care, as mutation-specific therapies and innovative therapeutic approaches are becoming available. Disease modifier genes, trans-active variants influencing disease severity and phenotypic expressivity, may modulate the response to therapy, and become new therapeutic targets. Uncovering more disease modifier genes via extensive genomic mapping studies offers the potential to fine-tune prognostic assessments for individuals with DMD. This review provides insights into genotype/phenotype correlations and the influence of modifier genes in DMD.
Collapse
Affiliation(s)
| | | | - Giulio Piluso
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Luca Bello
- Department of Neurosciences DNS, University of Padova, Padova, Italy
| |
Collapse
|
13
|
Atalaia A, Wandrei D, Lalout N, Thompson R, Tassoni A, 't Hoen PAC, Athanasiou D, Baker SA, Sakellariou P, Paliouras G, D'Angelo C, Horvath R, Mancuso M, van der Beek N, Kornblum C, Kirschner J, Pareyson D, Bassez G, Blacas L, Jacoupy M, Eng C, Lamy F, Plançon JP, Haberlova J, Brusse E, Hoeijmakers JGJ, de Visser M, Claeys KG, Paradas C, Toscano A, Silani V, Gyenge M, Reviers E, Hamroun D, Vroom E, Wilkinson MD, Lochmuller H, Evangelista T. EURO-NMD registry: federated FAIR infrastructure, innovative technologies and concepts of a patient-centred registry for rare neuromuscular disorders. Orphanet J Rare Dis 2024; 19:66. [PMID: 38355534 PMCID: PMC10865673 DOI: 10.1186/s13023-024-03059-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 02/03/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND The EURO-NMD Registry collects data from all neuromuscular patients seen at EURO-NMD's expert centres. In-kind contributions from three patient organisations have ensured that the registry is patient-centred, meaningful, and impactful. The consenting process covers other uses, such as research, cohort finding and trial readiness. RESULTS The registry has three-layered datasets, with European Commission-mandated data elements (EU-CDEs), a set of cross-neuromuscular data elements (NMD-CDEs) and a dataset of disease-specific data elements that function modularly (DS-DEs). The registry captures clinical, neuromuscular imaging, neuromuscular histopathology, biological and genetic data and patient-reported outcomes in a computer-interpretable format using selected ontologies and classifications. The EURO-NMD registry is connected to the EURO-NMD Registry Hub through an interoperability layer. The Hub provides an entry point to other neuromuscular registries that follow the FAIR data stewardship principles and enable GDPR-compliant information exchange. Four national or disease-specific patient registries are interoperable with the EURO-NMD Registry, allowing for federated analysis across these different resources. CONCLUSIONS Collectively, the Registry Hub brings together data that are currently siloed and fragmented to improve healthcare and advance research for neuromuscular diseases.
Collapse
Affiliation(s)
- Antonio Atalaia
- Inserm Center of Research in Myology, Neuro-Myology Service G.H. Pitié-Salpêtrière, Sorbonne Université, Paris, France.
| | - Dagmar Wandrei
- Clinical Trials Unit, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nawel Lalout
- Medical BioSciences Department, Radboud University Medical Center, Nijmegen, Netherlands
- Duchenne Parent Project, Veenendaal, The Netherlands
| | - Rachel Thompson
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada
| | - Adrian Tassoni
- Clinical Trials Unit, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter A C 't Hoen
- Medical BioSciences Department, Radboud University Medical Center, Nijmegen, Netherlands
| | | | | | | | | | - Carla D'Angelo
- European Reference Network for Rare Neuromuscular Diseases EURO-NMD, Institute of Myology, University Hospital Pitie-Salpetriere-APHP, Paris, France
| | - Rita Horvath
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Michelangelo Mancuso
- Department of Clinical and Experimental Medicine, Neurological Institute, University of Pisa, Pisa, Italy
| | - Nadine van der Beek
- Department of Neurology/Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Cornelia Kornblum
- Department of Neurology, Neuromuscular Diseases Section, University Hospital Bonn, Bonn, Germany
| | - Janbernd Kirschner
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Davide Pareyson
- Unit of Rare Neurological Diseases. Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Guillaume Bassez
- Neuromuscular Diseases Reference Center, Pitié-Salpêtrière University Hospital, APHP Paris, Paris, France
| | - Laura Blacas
- Association Institute of Myology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Maxime Jacoupy
- Association Institute of Myology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Catherine Eng
- Association Française Contre Les Myopathies, AFM-Téléthon, Evry, France
| | - François Lamy
- Association Française Contre Les Myopathies, AFM-Téléthon, Evry, France
| | - Jean-Philippe Plançon
- European Patient Organisation for Dysimmune and Inflammatory Neuropathies, Paris, France
| | - Jana Haberlova
- Neuromuscular Center, University Hospital Motol, Prague, Czech Republic
| | - Esther Brusse
- Department of Neurology/Center for Lysosomal and Metabolic Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Janneke G J Hoeijmakers
- Department of Neurology, Maastricht University Medical Center+, and MHeNS, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Marianne de Visser
- Department of Neurology, Amsterdam University Medical Center, Location Academic Medical Center, Amsterdam, The Netherlands
| | - Kristl G Claeys
- Department of Neurology, University Hospitals Leuven, and Laboratory for Muscle Diseases and Neuropathies, Department of Neurosciences, KU Leuven, and Leuven Brain Institute (LBI), Louvain, Belgium
| | - Carmen Paradas
- Hospital Universitario Virgen del Rocío/IBiS, Avda Manuel Siurot S/N, 41013, Seville, Andalucía, Spain
| | - Antonio Toscano
- Department of Clinical and Experimental Medicine, AOU G. Martino Di Messina, University of Messina, Messina, Italy
| | - Vincenzo Silani
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Melinda Gyenge
- Neuromuscular Diseases Reference Center, Pitié-Salpêtrière University Hospital, APHP Paris, Paris, France
| | | | - Dalil Hamroun
- CHRU de Montpellier, Direction de la Recherche et de L'Innovation, Hôpital La Colombière, Montpellier, France
| | | | - Mark D Wilkinson
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, Centro de Biotecnología y Genómica de Plantas UPM-INIA, Universidad Politécnica de Madrid (UPM), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA/CSIC), 28223, Madrid, ES, Spain
| | - Hanns Lochmuller
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Canada
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Teresinha Evangelista
- Neuromuscular Pathology Functional Unit; Neuropathology Service, Institute of Myology, University Hospital Pitié-Salpêtrière-APHP, Paris, France
| |
Collapse
|
14
|
Arshad A, Tsirikos AI. Survival of patients with Duchenne muscular dystrophy who underwent spinal deformity correction. Dev Med Child Neurol 2024; 66:187-194. [PMID: 37482906 DOI: 10.1111/dmcn.15711] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 07/25/2023]
Abstract
AIM We investigated survival rates after scoliosis correction in individuals with Duchenne muscular dystrophy (DMD) and evaluated factors that can affect them. METHOD This was a retrospective cohort study from 2000 to 2022 with a minimum 2-year postoperative follow-up. We reviewed the hospital records/spinal radiographs and analysed data with XLSTAT. Kaplan-Meier and multivariate Cox regression survival analysis was performed. RESULTS Forty-three patients had a mean age at surgery of 14 years 5 months. Mean postoperative follow-up was 10 years 10 months. There was no operative or 30-day postoperative mortality in this group. Twenty-four patients died because of cardiorespiratory failure. Median survivorship was 14 years 2 months, with the longest observed survival being 22 years 6 months given the limitation of the length of postoperative follow-up. The degree of preoperative coronal imbalance and pelvic obliquity, as well as intraoperative blood loss, were factors that significantly affected survival. The impact of preoperative sagittal imbalance and extension of the fusion to the sacrum/pelvis trended towards significance. In contrast, age at surgery, preoperative/postoperative scoliosis, thoracic kyphosis, lumbar lordosis, scoliosis and pelvic obliquity flexibility or correction indices, postoperative coronal/sagittal balance, need of preoperative non-invasive ventilation, preoperative feeding disorders, development of surgical complications, and length of hospital and intensive care unit stay were exposure variables that did not affect postsurgical survival in patients with DMD. INTERPRETATION Survival of up to two decades or more was possible among young patients with DMD after scoliosis correction. This was affected by factors that related to disease and deformity severity, as well as surgical morbidity. WHAT THIS PAPER ADDS There was no operative or 30-day postoperative mortality in this group of patients with Duchenne muscular dystrophy (DMD) undergoing scoliosis correction. Survival probabilities at 5-year, 10-year, 15-year, and 20-year intervals post-surgery were 92%, 80%, 33%, and 12% respectively. Scoliosis surgery achieved good deformity correction and a balanced spine that was maintained at follow-up. Respiratory failure, severe pneumonia, and left ventricular failure were the leading causes of death in the study participants. Preoperative global coronal imbalance, pelvic obliquity, and intraoperative blood loss significantly predicted survival. Factors that affected survival after scoliosis surgery were associated with perioperative morbidity and disease or deformity severity in the DMD group.
Collapse
Affiliation(s)
- Ayesha Arshad
- Scottish National Spine Deformity Centre, Royal Hospital for Children and Young People, Edinburgh, UK
| | - Athanasios I Tsirikos
- Scottish National Spine Deformity Centre, Royal Hospital for Children and Young People, Edinburgh, UK
| |
Collapse
|
15
|
Zhou K, Yuan M, Sun J, Zhang F, Zong X, Li Z, Tang D, Zhou L, Zheng J, Xiao X, Wu X. Sildenafil increases AAV9 transduction after a systemic administration and enhances AAV9-dystrophin therapeutic effect in mdx mice. Gene Ther 2024; 31:19-30. [PMID: 37500816 DOI: 10.1038/s41434-023-00411-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 07/07/2023] [Accepted: 07/17/2023] [Indexed: 07/29/2023]
Abstract
Adeno-associated virus (AAV) vectors have been successfully used to deliver genes for treating rare diseases. However, the systemic administration of high AAV vector doses triggers several adverse effects, including immune response, the asymptomatic elevation of liver transaminase levels, and complement activation. Thus, improving AAV transduction and reducing AAV dosage for treatment is necessary. Recently, we found that a phosphodiesterase-5 inhibitor significantly promoted AAV9 transduction in vitro by regulating the caveolae and macropinocytosis pathways. When AAV9-Gaussian luciferase (AAV9-Gluc) and AAV9-green fluorescent protein (AAV9-GFP) were injected intravenously into mice pre-treated with sildenafil, the expressions of Gluc in the plasma and GFP in muscle tissues significantly increased (P < 0.05). Sildenafil also improved Evans blue permeation in tissues. Additionally, we found that sildenafil promoted Treg proliferation, inhibited B-cell activation, and decreased anti-AAV9 IgG levels (P < 0.05). Furthermore, sildenafil significantly promoted Duchenne muscular dystrophy gene therapy efficacy using AAV9 in mdx mice; it increased micro-dystrophin gene expression, forelimb grip strength, and time spent on the rotarod test, decreased serum creatine kinase levels, and ameliorated histopathology by improving muscle cell morphology and reducing fibrosis (P < 0.05). These results show that sildenafil significantly improved AAV transduction, suppressed the levels of anti-AAV9 IgG, and enhanced the efficacy of gene therapy.
Collapse
Affiliation(s)
- Kaiyi Zhou
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Meng Yuan
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jiabao Sun
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Feixu Zhang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Xiaoying Zong
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Zhanao Li
- School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Dingyue Tang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, China
| | - Lichen Zhou
- The General Hospital of Western Theater Command PLA, Sichuan Province, China
| | - Jing Zheng
- Belief BioMed, Xuhui District, Shanghai, China
| | - Xiao Xiao
- School of Pharmacy, East China University of Science and Technology, Shanghai, China.
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27517, USA.
| | - Xia Wu
- School of Pharmacy, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
16
|
Landfeldt E, Aleman A, Abner S, Zhang R, Werner C, Tomazos I, Lochmüller H, Quinlivan R. Factors Associated with Respiratory Health and Function in Duchenne Muscular Dystrophy: A Systematic Review and Evidence Grading. J Neuromuscul Dis 2024; 11:25-57. [PMID: 37980679 PMCID: PMC10789346 DOI: 10.3233/jnd-230094] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Despite advances in the medical management of the disease, respiratory involvement remains a significant source of morbidity and mortality in children and adults with Duchenne muscular dystrophy (DMD). OBJECTIVE The objective of this systematic literature review was to synthesize and grade published evidence of factors associated with respiratory health and function in DMD. METHODS We searched MEDLINE, Embase, and the Cochrane Library for records of studies published from January 1, 2000 (to ensure relevance to current care practices), up until and including December 31, 2022, reporting evidence of prognostic indicators and predictors of disease progression in DMD. The quality of evidence (i.e., very low to high) was assessed using the Grading of Recommendations, Assessment, Development and Evaluations (GRADE) framework. RESULTS The bibliographic search strategy resulted in the inclusion of 29 articles. In total, evidence of 10 factors associated with respiratory health and function in patients with DMD was identified: glucocorticoid exposure (high- to very low-quality evidence), DMD mutations (low-quality evidence), DMD genetic modifiers (low-quality evidence), other pharmacological interventions (i.e., ataluren, eteplirsen, idebenone, and tamoxifen) (moderate- to very low-quality evidence), body mass index and weight (low-quality evidence), and functional ability (low-quality evidence). CONCLUSIONS In conclusion, we identified a total of 10 factors associated with respiratory health in function in DMD, encompassing both pharmacological therapies, genetic mutations and modifiers, and patient clinical characteristics. Yet, more research is needed to further delineate sources of respiratory heterogeneity, in particular the genotype-phenotype association and the impact of novel DMD therapies in a real-world setting. Our synthesis and grading should be helpful to inform clinical practice and future research of this heavily burdened patient population.
Collapse
Affiliation(s)
| | - A. Aleman
- Department of Pediatrics, Division of Neurology, Children’s Hospital of Eastern Ontario, Research Institute, University of Ottawa, Ottawa, ON, Canada
- Department of Medicine, Division of Neurology, The Ottawa Hospital, Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
| | | | - R. Zhang
- PTC Therapeutics Sweden AB, Askim, Sweden
| | - C. Werner
- PTC Therapeutics Germany GmbH, Frankfurt, Germany
| | - I. Tomazos
- PTC Therapeutics Inc, South Plainfield, NJ, USA
| | - H. Lochmüller
- Department of Pediatrics, Division of Neurology, Children’s Hospital of Eastern Ontario, Research Institute, University of Ottawa, Ottawa, ON, Canada
- Department of Medicine, Division of Neurology, The Ottawa Hospital, Brain and Mind Research Institute, University of Ottawa, Ottawa, ON, Canada
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - R.M. Quinlivan
- Centre for Neuromuscular Diseases, UCL Institute of Neurology, National Hospital, London, UK
| |
Collapse
|
17
|
Landfeldt E, Alemán A, Abner S, Zhang R, Werner C, Tomazos I, Ferizovic N, Lochmüller H, Kirschner J. Predictors of Loss of Ambulation in Duchenne Muscular Dystrophy: A Systematic Review and Meta-Analysis. J Neuromuscul Dis 2024; 11:579-612. [PMID: 38669554 DOI: 10.3233/jnd-230220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
Objective The objective of this study was to describe predictors of loss of ambulation in Duchenne muscular dystrophy (DMD). Methods This systematic review and meta-analysis included searches of MEDLINE ALL, Embase, and the Cochrane Database of Systematic Reviews from January 1, 2000, to December 31, 2022, for predictors of loss of ambulation in DMD. Search terms included "Duchenne muscular dystrophy" as a Medical Subject Heading or free text term, in combination with variations of the term "predictor". Risk of bias was assessed using the Newcastle-Ottawa Scale. We performed meta-analysis pooling of hazard ratios of the effects of glucocorticoids (vs. no glucocorticoid therapy) by fitting a common-effect inverse-variance model. Results The bibliographic searches resulted in the inclusion of 45 studies of children and adults with DMD from 17 countries across Europe, Asia, and North America. Glucocorticoid therapy was associated with delayed loss of ambulation (overall meta-analysis HR deflazacort/prednisone/prednisolone: 0.44 [95% CI: 0.40-0.48]) (n = 25 studies). Earlier onset of first signs or symptoms, earlier loss of developmental milestones, lower baseline 6MWT (i.e.,<350 vs. ≥350 metres and <330 vs. ≥330 metres), and lower baseline NSAA were associated with earlier loss of ambulation (n = 5 studies). Deletion of exons 3-7, proximal mutations (upstream intron 44), single exon 45 deletions, and mutations amenable of skipping exon 8, exon 44, and exon 53, were associated with prolonged ambulation; distal mutations (intron 44 and downstream), deletion of exons 49-50, and mutations amenable of skipping exon 45, and exon 51 were associated with earlier loss of ambulation (n = 13 studies). Specific single-nucleotide polymorphisms in CD40 gene rs1883832, LTBP4 gene rs10880, SPP1 gene rs2835709 and rs11730582, and TCTEX1D1 gene rs1060575 (n = 7 studies), as well as race/ethnicity and level of family/patient deprivation (n = 3 studies), were associated with loss of ambulation. Treatment with ataluren (n = 2 studies) and eteplirsen (n = 3 studies) were associated with prolonged ambulation. Magnetic resonance biomarkers (MRI and MRS) were identified as significant predictors of loss of ambulation (n = 6 studies). In total, 33% of studies exhibited some risk of bias. Conclusion Our synthesis of predictors of loss of ambulation in DMD contributes to the understanding the natural history of disease and informs the design of new trials of novel therapies targeting this heavily burdened patient population.
Collapse
Affiliation(s)
| | - A Alemán
- Department of Pediatrics, Division of Neurology, Children's Hospital of Eastern Ontario, Research Institute, University of Ottawa, Ottawa, ON, Canada
- Department of Medicine, Division of Neurology, The Ottawa Hospital, Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | | | - R Zhang
- PTC Therapeutics Sweden AB, Askim, Sweden
| | - C Werner
- PTC Therapeutics Germany GmbH, Frankfurt, Germany
| | - I Tomazos
- PTC Therapeutics Inc, South Plainfield, NJ, USA
| | | | - H Lochmüller
- Department of Pediatrics, Division of Neurology, Children's Hospital of Eastern Ontario, Research Institute, University of Ottawa, Ottawa, ON, Canada
- Department of Neuropediatrics and Muscle Disorders, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany
- Department of Medicine, Division of Neurology, The Ottawa Hospital, Brain and Mind Research Institute, University of Ottawa, Ottawa, Canada
| | - J Kirschner
- Department of Neuropediatrics and Muscle Disorders, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| |
Collapse
|
18
|
Sarker S, Eshaque TB, Soorajkumar A, Nassir N, Zehra B, Kanta SI, Rahaman MA, Islam A, Akter S, Ali MK, Mim RA, Uddin KMF, Chowdhury MSJ, Shams N, Baqui MA, Lim ET, Akter H, Woodbury-Smith M, Uddin M. Mutational spectrum and phenotypic variability of Duchenne muscular dystrophy and related disorders in a Bangladeshi population. Sci Rep 2023; 13:21547. [PMID: 38057384 PMCID: PMC10700514 DOI: 10.1038/s41598-023-48982-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 12/02/2023] [Indexed: 12/08/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe rare neuromuscular disorder caused by mutations in the X-linked dystrophin gene. Several mutations have been identified, yet the full mutational spectrum, and their phenotypic consequences, will require genotyping across different populations. To this end, we undertook the first detailed genotype and phenotype characterization of DMD in the Bangladeshi population. We investigated the rare mutational and phenotypic spectrum of the DMD gene in 36 DMD-suspected Bangladeshi participants using an economically affordable diagnostic strategy involving initial screening for exonic deletions in the DMD gene via multiplex PCR, followed by testing PCR-negative patients for mutations using whole exome sequencing. The deletion mapping identified two critical DMD gene hotspot regions (near proximal and distal ends, spanning exons 8-17 and exons 45-53, respectively) that comprised 95% (21/22) of the deletions for this population cohort. From our exome analysis, we detected two novel pathogenic hemizygous mutations in exons 21 and 42 of the DMD gene, and novel pathogenic recessive and loss of function variants in four additional genes: SGCD, DYSF, COL6A3, and DOK7. Our phenotypic analysis showed that DMD suspected participants presented diverse phenotypes according to the location of the mutation and which gene was impacted. Our study provides ethnicity specific new insights into both clinical and genetic aspects of DMD.
Collapse
Affiliation(s)
- Shaoli Sarker
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
- Bangladesh Shishu Hospital and Institute, Dhaka, Bangladesh
| | | | - Anjana Soorajkumar
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Nasna Nassir
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Binte Zehra
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | | | - Md Atikur Rahaman
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Amirul Islam
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
- GenomeArc Inc., Mississauga, Ontario, Canada
| | - Shimu Akter
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Mohammad Kawsar Ali
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Rabeya Akter Mim
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - K M Furkan Uddin
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | | | - Nusrat Shams
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Md Abdul Baqui
- Department of Biochemistry, Holy Family Red Crescent Medical College and Hospital, Dhaka, Bangladesh
| | - Elaine T Lim
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Hosneara Akter
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Marc Woodbury-Smith
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| | - Mohammed Uddin
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE.
- GenomeArc Inc., Mississauga, Ontario, Canada.
| |
Collapse
|
19
|
Tones M, Zeps N, Wyborn Y, Smith A, Barrero RA, Heussler H, Cross M, McGree J, Bellgard M. Does the registry speak your language? A case study of the Global Angelman Syndrome Registry. Orphanet J Rare Dis 2023; 18:330. [PMID: 37858180 PMCID: PMC10588126 DOI: 10.1186/s13023-023-02904-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 08/31/2023] [Indexed: 10/21/2023] Open
Abstract
Global disease registries are critical to capturing common patient related information on rare illnesses, allowing patients and their families to provide information about their condition in a safe, accessible, and engaging manner that enables researchers to undertake critical research aimed at improving outcomes. Typically, English is the default language of choice for these global digital health platforms. Unfortunately, language barriers can significantly inhibit participation from non-English speaking participants. In addition, there is potential for compromises in data quality and completeness. In contrast, multinational commercial entities provide access to their websites in the local language of the country they are operating in, and often provide multiple options reflecting ethnic diversity. This paper presents a case study of how the Global Angelman Syndrome Registry (GASR) has used a novel approach to enable multiple language translations for its website. Using a "semi-automated language translation" approach, the GASR, which was originally launched in English in September 2016, is now available in several other languages. In 2020, the GASR adopted a novel approach using crowd-sourcing and machine translation tools leading to the availability of the GASR in Spanish, Traditional Chinese, Italian, and Hindi. As a result, enrolments increased by 124% percent for Spain, 67% percent for Latin America, 46% percent for Asia, 24% for Italy, and 43% for India. We describe our approach here, which we believe presents an opportunity for cost-effective and timely translations responsive to changes to the registry and helps build and maintain engagement with global disease communities.
Collapse
Affiliation(s)
- Megan Tones
- Office of eResearch, Queensland University of Technology, Brisbane, QLD, 4000, Australia.
| | - Nikolajs Zeps
- Office of eResearch, Queensland University of Technology, Brisbane, QLD, 4000, Australia
| | - Yvette Wyborn
- Office of eResearch, Queensland University of Technology, Brisbane, QLD, 4000, Australia
| | - Adam Smith
- Office of eResearch, Queensland University of Technology, Brisbane, QLD, 4000, Australia
| | - Roberto A Barrero
- Office of eResearch, Queensland University of Technology, Brisbane, QLD, 4000, Australia
| | - Helen Heussler
- Centre for Clinical Trials in Rare Neurodevelopmental Disorders, Child Development Program, Children's Health Queensland, Child Health Research Centre University of Queensland, Brisbane, QLD, 4101, Australia
| | - Meagan Cross
- Foundation for Angelman Syndrome Therapeutics Australia, Salisbury, QLD, 4107, Australia
| | - James McGree
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Matthew Bellgard
- Office of eResearch, Queensland University of Technology, Brisbane, QLD, 4000, Australia.
- University of East London, London, UK.
| |
Collapse
|
20
|
Le Guiner C, Xiao X, Larcher T, Lafoux A, Huchet C, Toumaniantz G, Adjali O, Anegon I, Remy S, Grieger J, Li J, Farrokhi V, Neubert H, Owens J, McIntyre M, Moullier P, Samulski RJ. Evaluation of an AAV9-mini-dystrophin gene therapy candidate in a rat model of Duchenne muscular dystrophy. Mol Ther Methods Clin Dev 2023; 30:30-47. [PMID: 37746247 PMCID: PMC10512999 DOI: 10.1016/j.omtm.2023.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 05/15/2023] [Indexed: 09/26/2023]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked disease caused by loss-of-function mutations in the dystrophin gene and is characterized by muscle wasting and early mortality. Adeno-associated virus-mediated gene therapy is being investigated as a treatment for DMD. In the nonclinical study documented here, we determined the effective dose of fordadistrogene movaparvovec, a clinical candidate adeno-associated virus serotype 9 vector carrying a human mini-dystrophin transgene, after single intravenous injection in a dystrophin-deficient (DMDmdx) rat model of DMD. Overall, we found that transduction efficiency, number of muscle fibers expressing the human mini-dystrophin polypeptide, improvement of the skeletal and cardiac muscle tissue architecture, correction of muscle strength and fatigability, and improvement of diastolic and systolic cardiac function were directly correlated with the amount of vector administered. The effective dose was then tested in older DMDmdx rats with a more dystrophic phenotype similar to the pathology observed in older patients with DMD. Except for a less complete rescue of muscle function in the oldest cohort, fordadistrogene movaparvovec was also found to be therapeutically effective in older DMDmdx rats, suggesting that this product may be appropriate for evaluation in patients with DMD at all stages of disease.
Collapse
Affiliation(s)
- Caroline Le Guiner
- Nantes Université, CHU Nantes, INSERM, TaRGeT, UMR 1089, Translational Research for Gene Therapies, 44200 Nantes, France
| | - Xiao Xiao
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC 27599-7352, USA
| | | | - Aude Lafoux
- Therassay Platform, Capacités, Nantes Université, 44007 Nantes, France
| | - Corinne Huchet
- Nantes Université, CHU Nantes, INSERM, TaRGeT, UMR 1089, Translational Research for Gene Therapies, 44200 Nantes, France
- Therassay Platform, Capacités, Nantes Université, 44007 Nantes, France
| | - Gilles Toumaniantz
- Therassay Platform, Capacités, Nantes Université, 44007 Nantes, France
- Nantes Université, CHU Nantes, CNRS, L’Institut du Thorax, 44007 Nantes, France
| | - Oumeya Adjali
- Nantes Université, CHU Nantes, INSERM, TaRGeT, UMR 1089, Translational Research for Gene Therapies, 44200 Nantes, France
| | - Ignacio Anegon
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, ITUN, 44093 Nantes, France
| | - Séverine Remy
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, ITUN, 44093 Nantes, France
| | - Josh Grieger
- Bamboo Therapeutics, Pfizer, Chapel Hill, NC 27514, USA
| | - Juan Li
- Gene Therapy Center, Eshelman School of Pharmacy DPMP, University of North Carolina, Chapel Hill, NC 27599-7352, USA
| | | | | | | | | | - Philippe Moullier
- Nantes Université, CHU Nantes, INSERM, TaRGeT, UMR 1089, Translational Research for Gene Therapies, 44200 Nantes, France
| | - R. Jude Samulski
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC 27599-7352, USA
| |
Collapse
|
21
|
Rok M, Wong TWY, Maino E, Ahmed A, Yang G, Hyatt E, Lindsay K, Fatehi S, Marks R, Delgado-Olguín P, Ivakine EA, Cohn RD. Prevention of early-onset cardiomyopathy in Dmd exon 52-54 deletion mice by CRISPR-Cas9-mediated exon skipping. Mol Ther Methods Clin Dev 2023; 30:246-258. [PMID: 37545481 PMCID: PMC10403712 DOI: 10.1016/j.omtm.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/12/2023] [Indexed: 08/08/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a disease with a life-threatening trajectory resulting from mutations in the dystrophin gene, leading to degeneration of skeletal muscle and fibrosis of cardiac muscle. The overwhelming majority of mutations are multiexonic deletions. We previously established a dystrophic mouse model with deletion of exons 52-54 in Dmd that develops an early-onset cardiac phenotype similar to DMD patients. Here we employed CRISPR-Cas9 delivered intravenously by adeno-associated virus (AAV) vectors to restore functional dystrophin expression via excision or skipping of exon 55. Exon skipping with a solitary guide significantly improved editing outcomes and dystrophin recovery over dual guide excision. Some improvements to genomic and transcript editing levels were observed when the guide dose was enhanced, but dystrophin restoration did not improve considerably. Editing and dystrophin recovery were restricted primarily to cardiac tissue. Remarkably, our exon skipping approach completely prevented onset of the cardiac phenotype in treated mice up to 12 weeks. Thus, our results demonstrate that intravenous delivery of a single-cut CRISPR-Cas9-mediated exon skipping therapy can prevent heart dysfunction in DMD in vivo.
Collapse
Affiliation(s)
- Matthew Rok
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Tatianna Wai Ying Wong
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Eleonora Maino
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Abdalla Ahmed
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Grace Yang
- Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Elzbieta Hyatt
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Kyle Lindsay
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Sina Fatehi
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Ryan Marks
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Paul Delgado-Olguín
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Heart & Stroke Richard Lewar Centre of Excellence, Toronto, ON, Canada
| | - Evgueni A. Ivakine
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Ronald D. Cohn
- Program in Genetics and Genome Biology, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
22
|
Stephenson AA, Nicolau S, Vetter TA, Dufresne GP, Frair EC, Sarff JE, Wheeler GL, Kelly BJ, White P, Flanigan KM. CRISPR-Cas9 homology-independent targeted integration of exons 1-19 restores full-length dystrophin in mice. Mol Ther Methods Clin Dev 2023; 30:486-499. [PMID: 37706184 PMCID: PMC10495553 DOI: 10.1016/j.omtm.2023.08.009] [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: 03/09/2023] [Accepted: 08/15/2023] [Indexed: 09/15/2023]
Abstract
Duchenne muscular dystrophy is an X-linked disorder typically caused by out-of-frame mutations in the DMD gene. Most of these are deletions of one or more exons, which can theoretically be corrected through CRISPR-Cas9-mediated knockin. Homology-independent targeted integration is a mechanism for achieving such a knockin without reliance on homology-directed repair pathways, which are inactive in muscle. We designed a system based on insertion into intron 19 of a DNA fragment containing a pre-spliced mega-exon encoding DMD exons 1-19, along with the MHCK7 promoter, and delivered it via a pair of AAV9 vectors in mice carrying a Dmd exon 2 duplication. Maximal efficiency was achieved using a Cas9:donor adeno-associated virus (AAV) ratio of 1:5, with Cas9 under the control of the SPc5-12 promoter. This approach achieved editing of 1.4% of genomes in the heart, leading to 30% correction at the transcript level and restoration of 11% of normal dystrophin levels. Treatment efficacy was lower in skeletal muscles. Sequencing additionally revealed integration of fragmentary and recombined AAV genomes at the target site. These data provide proof of concept for a gene editing system that could restore full-length dystrophin in individuals carrying mutations upstream of intron 19, accounting for approximately 25% of Duchenne muscular dystrophy patients.
Collapse
Affiliation(s)
- Anthony A. Stephenson
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Stefan Nicolau
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Tatyana A. Vetter
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Gabrielle P. Dufresne
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Emma C. Frair
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Jessica E. Sarff
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Gregory L. Wheeler
- The Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Benjamin J. Kelly
- The Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Peter White
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
- The Institute for Genomic Medicine, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Kevin M. Flanigan
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
- Department of Neurology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
23
|
Sunthankar SD, George-Durrett K, Crum K, Slaughter JC, Kasten J, Raucci FJ, Markham LW, Soslow JH. Comprehensive cardiac magnetic resonance T1, T2, and extracellular volume mapping to define Duchenne cardiomyopathy. J Cardiovasc Magn Reson 2023; 25:44. [PMID: 37517994 PMCID: PMC10388519 DOI: 10.1186/s12968-023-00951-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 06/30/2023] [Indexed: 08/01/2023] Open
Abstract
BACKGROUND Cardiomyopathy is the leading cause of death in Duchenne muscular dystrophy (DMD). Cardiac magnetic resonance (CMR) parametric mapping sequences offer insights into disease pathophysiology. We propose a novel approach by leveraging T2 mapping in conjunction with T1 and extracellular volume (ECV) mapping to perform a virtual myocardial biopsy. While previous work has attempted to describe myocardial changes in DMD, our inclusion of T2 mapping enables comprehensive categorization of myocardial tissue characteristics of fibrosis, edema, and fat to better understand the pathological composition of the myocardium with disease progression. METHODS DMD patients (n = 49; median: 12 years-old) underwent CMR, including T1, T2, and ECV. Categories were defined as normal, isolated high T1 (normal ECV, high T1, normal T2), fibrosis (high ECV, normal or high T1, normal T2), edema (normal or high ECV, normal or high T1, high T2), fat (normal ECV, low T1, high T2) or fibrofatty (high ECV, low T1, high T2). RESULTS Median left ventricular ejection fraction (LVEF) was 59% with 27% having LVEF < 55%. Those with normal LVEF and no late gadolinium enhancement (37%) were younger in age (10.5 ± 2.6 vs. 15.0 ± 4.3 years-old, p < 0.001). Native T1 was elevated in at least one slice in 82% of patients. Those with high T2 at any slice (27%) were older (p = 0.005) and had lower LVEF (p = 0.005) compared with subjects with normal T2 (73%). The most common myocardial characterization was fibrosis (43%) followed by isolated high T1 (24%). Of the 13 with high T2, ten were categorized as edema, two as fibrofatty, and one as fat. CONCLUSION CMR parametric mapping sequences offer insights into Duchenne cardiomyopathy pathophysiology, which should drive development of therapeutic interventions aimed at these targets. Myocardial fibrosis is common in DMD. Patients with elevated T2 were older and had lower LVEF. Though fat infiltration was present, the majority of subjects with elevated T2 met criteria for myocardial edema.
Collapse
Affiliation(s)
- Sudeep D Sunthankar
- Thomas P. Graham Jr Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Monroe Carell Jr Children's Hospital at Vanderbilt, 2220 Children's Way, Suite 5230, TN, 37232, Nashville, USA.
| | - Kristen George-Durrett
- Thomas P. Graham Jr Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Monroe Carell Jr Children's Hospital at Vanderbilt, 2220 Children's Way, Suite 5230, TN, 37232, Nashville, USA
| | - Kimberly Crum
- Thomas P. Graham Jr Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Monroe Carell Jr Children's Hospital at Vanderbilt, 2220 Children's Way, Suite 5230, TN, 37232, Nashville, USA
| | - James C Slaughter
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jennifer Kasten
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Frank J Raucci
- Division of Pediatric Cardiology, Department of Pediatrics, Children's Hospital of Richmond at Virginia Commonwealth University Health System, Richmond, VA, 23219, USA
| | - Larry W Markham
- Division of Cardiology, Department of Pediatrics, Riley Hospital for Children at Indiana University Health, Indianapolis, IN, 46202, USA
| | - Jonathan H Soslow
- Thomas P. Graham Jr Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Monroe Carell Jr Children's Hospital at Vanderbilt, 2220 Children's Way, Suite 5230, TN, 37232, Nashville, USA
| |
Collapse
|
24
|
Bello L, Hoffman EP, Pegoraro E. Is it time for genetic modifiers to predict prognosis in Duchenne muscular dystrophy? Nat Rev Neurol 2023; 19:410-423. [PMID: 37308617 DOI: 10.1038/s41582-023-00823-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/14/2023]
Abstract
Patients with Duchenne muscular dystrophy (DMD) show clinically relevant phenotypic variability, despite sharing the same primary biochemical defect (dystrophin deficiency). Factors contributing to this clinical variability include allelic heterogeneity (specific DMD mutations), genetic modifiers (trans-acting genetic polymorphisms) and variations in clinical care. Recently, a series of genetic modifiers have been identified, mostly involving genes and/or proteins that regulate inflammation and fibrosis - processes increasingly recognized as being causally linked with physical disability. This article reviews genetic modifier studies in DMD to date and discusses the effect of genetic modifiers on predicting disease trajectories (prognosis), clinical trial design and interpretation (inclusion of genotype-stratified subgroup analyses) and therapeutic approaches. The genetic modifiers identified to date underscore the importance of progressive fibrosis, downstream of dystrophin deficiency, in driving the disease process. As such, genetic modifiers have shown the importance of therapies aimed at slowing this fibrotic process and might point to key drug targets.
Collapse
Affiliation(s)
- Luca Bello
- Department of Neurosciences (DNS), University of Padova, Padova, Italy
| | - Eric P Hoffman
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University (State University of New York), Binghamton, NY, USA
| | - Elena Pegoraro
- Department of Neurosciences (DNS), University of Padova, Padova, Italy.
| |
Collapse
|
25
|
Viggiano E, Picillo E, Passamano L, Onore ME, Piluso G, Scutifero M, Torella A, Nigro V, Politano L. Spectrum of Genetic Variants in the Dystrophin Gene: A Single Centre Retrospective Analysis of 750 Duchenne and Becker Patients from Southern Italy. Genes (Basel) 2023; 14:214. [PMID: 36672955 PMCID: PMC9859256 DOI: 10.3390/genes14010214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/29/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Dystrophinopathies are X-linked recessive muscle disorders caused by mutations in the dystrophin (DMD) gene that include deletions, duplications, and point mutations. Correct diagnosis is important for providing adequate patient care and family planning, especially at this time when mutation-specific therapies are available. We report a large single-centre study on the spectrum of DMD gene variants observed in 750 patients analyzed for suspected Duchenne (DMD) or Becker (BMD) muscular dystrophy, over the past 30 years, at the Cardiomyology and Medical Genetics of the University of Campania. We found 534 (71.21%) large deletions, 73 (9.73%) large duplications, and 112 (14.93%) point mutations, of which 44 (5.9%) were small ins/del causing frame-shifts, 57 (7.6%) nonsense mutations, 8 (1.1%) splice site and 3 (0.4%) intronic mutations, and 31 (4.13%) non mutations. Moreover, we report the prevalence of the different types of mutations in patients with DMD and BMD according to their decade of birth, from 1930 to 2020, and correlate the data to the different techniques used over the years. In the most recent decades, we observed an apparent increase in the prevalence of point mutations, probably due to the use of Next-Generation Sequencing (NGS). In conclusion, in southern Italy, deletions are the most frequent variation observed in DMD and BMD patients followed by point mutations and duplications, as elsewhere in the world. NGS was useful to identify point mutations in cases of strong suspicion of DMD/BMD negative on deletions/duplications analyses. In the era of personalized medicine and availability of new causative therapies, a collective effort is necessary to enable DMD and BMD patients to have timely genetic diagnoses and avoid late implementation of standard of care and late initiation of appropriate treatment.
Collapse
Affiliation(s)
- Emanuela Viggiano
- Department of Prevention, Hygiene and Public Health Service, ASL Roma 2, 00157 Rome, Italy
| | - Esther Picillo
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Luigia Passamano
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Maria Elena Onore
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Giulio Piluso
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Marianna Scutifero
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Annalaura Torella
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Vincenzo Nigro
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy
| | - Luisa Politano
- Cardiomyology and Medical Genetics, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| |
Collapse
|
26
|
Araujo APDQC, Saute JAM, Fortes CPDD, França MC, Pereira JA, Albuquerque MAVD, Carvalho AADS, Cavalcanti EBU, Covaleski APPM, Fagondes SC, Gurgel-Giannetti J, Gonçalves MVM, Martinez ARM, Coimbra Neto AR, Neves FR, Nucci A, Nucera APCDS, Pessoa ALS, Rebel MF, Santos FND, Scola RH, Sobreira CFDR. Update of the Brazilian consensus recommendations on Duchenne muscular dystrophy. ARQUIVOS DE NEURO-PSIQUIATRIA 2023; 81:81-94. [PMID: 36918011 PMCID: PMC10014210 DOI: 10.1055/s-0043-1761466] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
In the last few decades, there have been considerable improvements in the diagnosis and care of Duchenne muscular dystrophy (DMD), the most common childhood muscular dystrophy. International guidelines have been published and recently reviewed. A group of Brazilian experts has developed a standard of care based on a literature review with evidence-based graded recommendations in a two-part publication. Implementing best practice management has helped change the natural history of this chronic progressive disorder, in which the life expectancy for children of the male sex in the past used to be very limited. Since the previous publication, diagnosis, steroid treatment, rehabilitation, and systemic care have gained more significant insights with new original work in certain fields. Furthermore, the development of new drugs is ongoing, and some interventions have been approved for use in certain countries. Therefore, we have identified the need to review the previous care recommendations for Brazilian patients with DMD. Our objective was to create an evidence-based document that is an update on our previous consensus on those topics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Flavio Reis Neves
- Instituto de Puericultura e Pediatria Martagão Gesteira, Equipe de Pesquisa em Doenças Neuromusculares, Rio de Janeiro RJ, Brazil
| | - Anamarli Nucci
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Campinas SP, Brazil
| | | | | | - Marcos Ferreira Rebel
- Universidade Federal do Rio de Janeiro, Faculdade de Fisioterapia, Rio de Janeiro RJ, Brazil
| | | | | | | |
Collapse
|
27
|
Liu J, Barrett JS, Leonardi ET, Lee L, Roychoudhury S, Chen Y, Trifillis P. Natural History and Real-World Data in Rare Diseases: Applications, Limitations, and Future Perspectives. J Clin Pharmacol 2022; 62 Suppl 2:S38-S55. [PMID: 36461748 PMCID: PMC10107901 DOI: 10.1002/jcph.2134] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/28/2022] [Indexed: 12/04/2022]
Abstract
Rare diseases represent a highly heterogeneous group of disorders with high phenotypic and genotypic diversity within individual conditions. Due to the small numbers of people affected, there are unique challenges in understanding rare diseases and drug development for these conditions, including patient identification and recruitment, trial design, and costs. Natural history data and real-world data (RWD) play significant roles in defining and characterizing disease progression, final patient populations, novel biomarkers, genetic relationships, and treatment effects. This review provides an introduction to rare diseases, natural history data, RWD, and real-world evidence, the respective sources and applications of these data in several rare diseases. Considerations for data quality and limitations when using natural history and RWD are also elaborated. Opportunities are highlighted for cross-sector collaboration, standardized and high-quality data collection using new technologies, and more comprehensive evidence generation using quantitative approaches such as disease progression modeling, artificial intelligence, and machine learning. Advanced statistical approaches to integrate natural history data and RWD to further disease understanding and guide more efficient clinical study design and data analysis in drug development in rare diseases are also discussed.
Collapse
Affiliation(s)
- Jing Liu
- Pfizer, Inc., Groton, Connecticut, USA
| | - Jeffrey S Barrett
- Critical Path Institute, Rare Disease Cures Accelerator Data Analytics Platform, Tucson, Arizona, USA
| | | | - Lucy Lee
- PTC Therapeutics, Inc., South Plainfield, New Jersey, USA
| | | | - Yong Chen
- Pfizer, Inc., Groton, Connecticut, USA
| | | |
Collapse
|
28
|
Kariyawasam D, D’Silva A, Mowat D, Russell J, Sampaio H, Jones K, Taylor P, Farrar M. Incidence of Duchenne muscular dystrophy in the modern era; an Australian study. Eur J Hum Genet 2022; 30:1398-1404. [PMID: 35754057 PMCID: PMC9712523 DOI: 10.1038/s41431-022-01138-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 04/20/2022] [Accepted: 06/13/2022] [Indexed: 11/09/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), an X-linked recessive condition is maternally inherited in two-thirds of affected boys. It is important to establish carrier status of female relatives to restore reproductive confidence for non-carriers and facilitate reproductive options and cardiac surveillance for carriers. This study investigates disease incidence within an Australian model of cascade screening and evolving genetic diagnostic technologies. A retrospective population-based cohort study of all genetically and/or histopathologically confirmed males with DMD, born in New South Wales and the Australian Capital Territory was undertaken from 2002-2012. Cases were identified using state-wide molecular laboratory and clinical databases. The annual disease incidence and "theoretically" preventable cases were extrapolated over the study period. Proband genotype/phenotype, pedigree analysis, carrier-risk and extent of cascade screening were also determined. The cumulative incidence of disease was 19.7 per 100,000 male live births and 1 in 5076 live born males were diagnosed with DMD. Differences in disease incidence were not statistically different when compared between 2002-2007 and 2008-2012 (incidence rate ratio = 1.13, 95% CI 0.76-1.69, p = 0.52). The incidence rate ratio of theoretically preventable cases did not significantly change between 2002-2007 and 2008-2012 (incidence rate ratio = 2.07, 95% CI 0.58-9.21, p = 0.23). Current diagnostic and cascade screening models have limitations in their impact on disease incidence, due to a spectrum of logistical, patient and condition related factors. Innovative approaches to reduce DMD incidence may be better achieved by preconception or early pregnancy carrier screening, prenatal exome sequencing and newborn screening.
Collapse
Affiliation(s)
- Didu Kariyawasam
- Department of Neurology, Sydney Children's Hospital, Randwick, Sydney, NSW, Australia. .,School of Clinical Medicine, UNSW Medicine and Health, Randwick Clinical Campus, Discipline of Paediatrics, University of New South Wales, Sydney, NSW, Australia.
| | - Arlene D’Silva
- grid.1005.40000 0004 4902 0432School of Clinical Medicine, UNSW Medicine and Health, Randwick Clinical Campus, Discipline of Paediatrics, University of New South Wales, Sydney, NSW Australia
| | - David Mowat
- grid.1005.40000 0004 4902 0432School of Clinical Medicine, UNSW Medicine and Health, Randwick Clinical Campus, Discipline of Paediatrics, University of New South Wales, Sydney, NSW Australia ,grid.414009.80000 0001 1282 788XCentre for Clinical Genetics, Sydney Children’s Hospital, Randwick, Sydney, NSW Australia
| | - Jacqui Russell
- grid.414009.80000 0001 1282 788XCentre for Clinical Genetics, Sydney Children’s Hospital, Randwick, Sydney, NSW Australia
| | - Hugo Sampaio
- grid.414009.80000 0001 1282 788XDepartment of Neurology, Sydney Children’s Hospital, Randwick, Sydney, NSW Australia
| | - Kristi Jones
- grid.413973.b0000 0000 9690 854XDepartment of Clinical Genetics, Children’s Hospital Westmead, Westmead, NSW Australia ,grid.1013.30000 0004 1936 834XPaediatrics and Child Health, Sydney Medical School, University of Sydney NSW Australia, Sydney, NSW Australia
| | - Peter Taylor
- Genomic Diagnostics, Healius Pathology, Melbourne, Vic Australia
| | - Michelle Farrar
- grid.414009.80000 0001 1282 788XDepartment of Neurology, Sydney Children’s Hospital, Randwick, Sydney, NSW Australia ,grid.1005.40000 0004 4902 0432School of Clinical Medicine, UNSW Medicine and Health, Randwick Clinical Campus, Discipline of Paediatrics, University of New South Wales, Sydney, NSW Australia
| |
Collapse
|
29
|
Wojciechowski J, Purohit VS, Harnisch LO, Dua P, Tan B, Nicholas T. Population PK and PD Analysis of Domagrozumab in Pediatric Patients with Duchenne Muscular Dystrophy. Clin Pharmacol Ther 2022; 112:1291-1302. [PMID: 36104012 PMCID: PMC9828399 DOI: 10.1002/cpt.2747] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 08/22/2022] [Indexed: 01/31/2023]
Abstract
Myostatin, a negative regulator of skeletal muscle growth, is a therapeutic target in muscle-wasting diseases. Domagrozumab, a humanized recombinant monoclonal antibody, binds myostatin and inhibits activity. Domagrozumab was investigated in a phase II trial (NCT02310763) as a potential treatment for boys with Duchenne muscular dystrophy (DMD). Pharmacokinetic/pharmacodynamic (PK/PD) modeling is vital in clinical trial design, particularly for determining dosing regimens in pediatric populations. This analysis sought to establish the PK/PD relationship between free domagrozumab and total myostatin concentrations in pediatric patients with DMD using a prior semimechanistic model developed from a phase I study in healthy adult volunteers (NCT01616277) and following inclusion of phase II data. The refined model was developed using a multiple-step approach comprising structural, random effects, and covariate model development; assessment of model adequacy (goodness-of-fit); and predictive performance. Differences in PKs/PDs between healthy adult volunteers and pediatric patients with DMD were quantitatively accounted for and evaluated by predicting myostatin coverage (the percentage of myostatin bound by domagrozumab). The final model parameter estimates and semimechanistic target-mediated drug disposition structure sufficiently described both domagrozumab and myostatin concentrations in pediatric patients with DMD, and most population parameters were comparable with the prior model (in healthy adult volunteers). Predicted myostatin coverage for phase II patients with DMD was consistently > 90%. Baseline serum myostatin was ~ 65% lower than in healthy adult volunteers. This study provides insights into the regulation of myostatin in healthy adults and pediatric patients with DMD. Clinicaltrials.gov identifiers: NCT01616277 and NCT02310763.
Collapse
|
30
|
High-Throughput Screening to Identify Modulators of Sarcospan. Methods Mol Biol 2022; 2587:479-493. [PMID: 36401045 DOI: 10.1007/978-1-0716-2772-3_25] [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: 11/19/2022]
Abstract
High-throughput screening enables the discovery of disease-modifying small molecules. Here, we describe the development of a scalable, cell-based assay to screen for small molecules that modulate sarcospan for the treatment of Duchenne muscular dystrophy. We detail the hit validation pipeline, which includes secondary screening, gene/protein quantification, and an in vitro membrane stability assay.
Collapse
|
31
|
Szabo SM, Klimchak AC, Qian C, Iannaccone S, Popoff E, Gooch KL. Characterizing the Occurrence of Key Clinical Milestones in Duchenne Muscular Dystrophy in the United States Using Real-World Data. J Neuromuscul Dis 2022; 9:689-699. [DOI: 10.3233/jnd-220816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Background: Data on the clinical course of Duchenne muscular dystrophy (DMD) exist from well-characterized clinical cohorts but estimates from real-world populations are fewer. Objective: The objective was to estimate the prevalence of key clinical milestones by age, among real-world commercially-insured DMD patients in the United States. Methods: MarketScan claims (2013–2018) were used to identify males with DMD. The percentages with wheelchair use or experiencing scoliosis, neurologic/neuropsychiatric involvement, cardiomyopathy, and respiratory involvement were tabulated; as were the median (interquartile range [IQR]) ages at first observed occurrence within the claims data. Results: Among DMD patients (n = 1,964), the median (IQR) baseline age was 15 (9–21) years, and median follow-up was 1.7 years. Wheelchair use was observed in 55% of those aged 8 to 13 years at cohort entry; scoliosis, among 38% of those 8 to 10 and 52% of those 11 to 13 years; neurologic/neuropsychiatric involvement, among 41–43% of those 8 to 13 years; respiratory involvement, among 45% of those 14 to 19 years; and cardiomyopathy, among 68% of those 14 to 16 and 58% of those 17 to 19 years. Conclusions: The prevalence of key clinical milestones across ages was broadly consistent with published findings. Variability in estimates reflect clinical heterogeneity; these contemporary estimates from real-world data help characterize clinical outcomes in DMD.
Collapse
Affiliation(s)
- Shelagh M. Szabo
- Broadstreet Health Economics & Outcomes Research, Vancouver BC, Canada
| | | | - Christina Qian
- Broadstreet Health Economics & Outcomes Research, Vancouver BC, Canada
| | - Susan Iannaccone
- University of Texas Southwestern, Harry Hines Blvd, Dallas TX, USA
| | - Evan Popoff
- Broadstreet Health Economics & Outcomes Research, Vancouver BC, Canada
| | | |
Collapse
|
32
|
Gan S, Liu S, Yang H, Wu L. Clinical and genetic characteristics of Chinese Duchenne/Becker muscular dystrophy patients with small mutations. Front Neurosci 2022; 16:992546. [DOI: 10.3389/fnins.2022.992546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022] Open
Abstract
BackgroundDuchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are amongst the inherited neuromuscular diseases with the highest incidence. Small mutations are less common and therefore have been poorly studied in China.Materials and methodsThe clinical data of 150 patients diagnosed with DMD/BMD by genetic analysis in Hunan Children’s Hospital from 2009 to 2021 were analyzed. The patients were followed up for an average of 3.42 years and their clinical characteristics were collected. Loss of ambulation (LOA) was used to evaluate the severity of disease progression. The correlation among clinical features, different variants, and glucocorticoid (GC) treatment was analyzed by Cox regression analysis.Results150 different variants were detected in this study, including 21 (14%) novel mutations, 88 (58.7%) non-sense mutations, 33 (22.0%) frameshift mutations, 22 (14.7%) splicing mutations, and 7 (4.7%) missense mutations. Single-exon skipping and single- or double-exon (double/single-exon) skipping strategies covered more than 90% of patients with small mutations. A case with frameshift mutation combined with Klinefelter’s syndrome (47, XXY) and another one with missense mutation combined with epilepsy was found in our study. De novo mutations accounted for 30.0% of all patients. The mean onset age was 4.19 ± 1.63 years old, and the mean diagnosed age was 5.60 ± 3.13 years old. The mean age of LOA was 10.4 years old (40 cases). 60.7% of them received GC treatment at 7.0 ± 2.7 years old. The main causes of complaints were muscle weakness, high creatine kinase (CK), motor retardation, and family history. The risk factors of LOA were positive family history (HR 5.52, CI 1.26–24.18), short GC treatment duration (HR 0.54, CI 0.36–0.82) and frameshift mutation (HR 14.58, CI 1.74–121.76). DMD patients who treated with GC after 7 years old had a higher risk of earlier LOA compared to those receiving treatment before the age of 7 (HR 0.083, CI 0.009–0.804). Moreover, an earlier onset age, a higher CK value, and a larger LOA population were found in the DMD patients compared to the BMD ones. Finally, the locations of the most frequent mutation were in exon 70 and exon 22.ConclusionIn conclusion, 150 small mutations were identified in this study, and 21 of them were discovered for the first time. We found the hotspots of small mutations were in exon 70 and exon 20. Also, the analysis showed that positive family history, frameshift mutation, short duration of GC treatment, and delayed GC treatment resulted in earlier LOA for the DMD patients. Taken together, our findings complement the mutation spectrum of DMD/BMD, benefit us understanding to the DMD disease, and lay foundations for the clinical trials.
Collapse
|
33
|
Farrokhi V, Walsh J, Palandra J, Brodfuehrer J, Caiazzo T, Owens J, Binks M, Neelakantan S, Yong F, Dua P, Le Guiner C, Neubert H. Dystrophin and mini-dystrophin quantification by mass spectrometry in skeletal muscle for gene therapy development in Duchenne muscular dystrophy. Gene Ther 2022; 29:608-615. [PMID: 34737451 PMCID: PMC9068826 DOI: 10.1038/s41434-021-00300-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 01/09/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, degenerative muscle disorder caused by mutations in the DMD gene, leading to severe reduction or absence of the protein dystrophin. Gene therapy strategies that aim to increase expression of a functional dystrophin protein (mini-dystrophin) are under investigation. The ability to accurately quantify dystrophin/mini-dystrophin is essential in assessing the level of gene transduction. We demonstrated the validation and application of a novel peptide immunoaffinity liquid chromatography-tandem mass spectrometry (IA-LC-MS/MS) assay. Data showed that dystrophin expression in Becker muscular dystrophy and DMD tissues, normalized against the mean of non-dystrophic control tissues (n = 20), was 4-84.5% (mean 32%, n = 20) and 0.4-24.1% (mean 5%, n = 20), respectively. In a DMD rat model, biceps femoris tissue from dystrophin-deficient rats treated with AAV9.hCK.Hopti-Dys3978.spA, an adeno-associated virus vector containing a mini-dystrophin transgene, showed a dose-dependent increase in mini-dystrophin expression at 6 months post-dose, exceeding wildtype dystrophin levels at high doses. Validation data showed that inter- and intra-assay precision were ≤20% (≤25% at the lower limit of quantification [LLOQ]) and inter- and intra-run relative error was within ±20% (±25% at LLOQ). IA-LC-MS/MS accurately quantifies dystrophin/mini-dystrophin in human and preclinical species with sufficient sensitivity for immediate application in preclinical/clinical trials.
Collapse
Affiliation(s)
- Vahid Farrokhi
- Biomedicine Design, Worldwide Research & Development, Pfizer Inc, 1 Burtt Road, Andover, MA, 01810, USA
| | - Jason Walsh
- Biomedicine Design, Worldwide Research & Development, Pfizer Inc, 1 Burtt Road, Andover, MA, 01810, USA
| | - Joe Palandra
- Biomedicine Design, Worldwide Research & Development, Pfizer Inc, 1 Burtt Road, Andover, MA, 01810, USA
| | - Joanne Brodfuehrer
- Biomedicine Design, Worldwide Research & Development, Pfizer Inc, 610 Main Street, Cambridge, MA, 02139, USA
| | - Teresa Caiazzo
- Biomedicine Design, Worldwide Research & Development, Pfizer Inc, 1 Burtt Road, Andover, MA, 01810, USA
| | - Jane Owens
- Rare Disease Research Unit, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, MA, 02139, USA
| | - Michael Binks
- Rare Disease Research Unit, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, MA, 02139, USA
| | - Srividya Neelakantan
- Clinical Pharmacology, Early Clinical Development, Worldwide Research & Development, Pfizer Inc, 1 Portland St, Cambridge, MA, 02139, USA
| | - Florence Yong
- Biostatistics, Worldwide Research & Development, Pfizer Inc, Cambridge, MA, 02139, USA
| | - Pinky Dua
- Early Clinical Development, Clinical Pharmacology, Pfizer R&D UK Limited, Cambridge, UK
| | - Caroline Le Guiner
- Translational Gene Therapy Laboratory, University of Nantes, INSERM UMR1089, CHU de Nantes, IRS 2 Nantes Biotech, 22 Boulevard Benoni Goulin, 44200, Nantes, France
| | - Hendrik Neubert
- Biomedicine Design, Worldwide Research & Development, Pfizer Inc, 1 Burtt Road, Andover, MA, 01810, USA.
| |
Collapse
|
34
|
Barboni MTS, Joachimsthaler A, Roux MJ, Nagy ZZ, Ventura DF, Rendon A, Kremers J, Vaillend C. Retinal dystrophins and the retinopathy of Duchenne muscular dystrophy. Prog Retin Eye Res 2022:101137. [DOI: 10.1016/j.preteyeres.2022.101137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 10/25/2022] [Accepted: 11/03/2022] [Indexed: 11/21/2022]
|
35
|
Zhang S, Wang S, Dong Y, Chen X, Hu M, Kou S, Zhang C, Wu S, Tian Y. Current care practices for patients with Duchenne muscular dystrophy in China. Brain Dev 2022; 44:623-629. [PMID: 35691779 PMCID: PMC9181563 DOI: 10.1016/j.braindev.2022.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/18/2022] [Accepted: 05/23/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND The coronavirus disease (COVID-19) pandemic has presented challenges in the care of patients with chronic diseases. We identified the challenges faced by Chinese patients with Duchenne muscular dystrophy (DMD) during the pandemic. METHODS An online cross-sectional survey was conducted between March 27 and June 30, 2021. RESULTS Of the 2105 valid questionnaire responses, 2,056 patients were from non-lockdown areas. In these areas, 42.8% reduced outside daily activities, 49.4% reduced rehabilitation service use, 39.7% postponed regular follow-ups, and 40.8% reported accelerated motor function decline. These figures generally increased for patients from lockdown areas-67.3% reduced outside daily activities, 44.9% reduced rehabilitation service use, 79.6% postponed regular follow-ups, and 55.1% reported accelerated motor function decline. Ambulation loss was most commonly reported in September and March before 2020; however, this trend was absent in 2020. Regarding the informed prices of disease-modifying drugs in Europe and the United States, 86.7% could afford a maximum of one-twentieth of the prices, 8.0% could afford one-tenth of the prices, and only 0.6% of the patients could afford the full prices. CONCLUSIONS Implementation of standardized care for DMD in China is consistent with global practices, and the COVID-19 pandemic has affected the care of patients with chronic diseases worldwide, particularly in lockdown areas. Telemedicine is an effective model for providing healthcare to such patients. Healthcare workers should assist patients and establish more robust chronic disease management systems. Collaboration between governmental and non-governmental entities could address the cost of disease-modifying drugs in China and other developing countries.
Collapse
Affiliation(s)
- Shu Zhang
- Research Center for Birth Defects Prevention and Control Technology, Chinese PLA General Hospital, 28 Fu-Xing Road, Beijing 100853, China,Department of Neurology, First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Shaoxia Wang
- Department of Neurology, Third Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Yuru Dong
- Department of Radiology, Third Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Xinyu Chen
- Department of Neurology, Third Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Miao Hu
- Department of Neurology, Third Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Sen Kou
- Department of Neurology, Third Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Chunyan Zhang
- Research Center for Birth Defects Prevention and Control Technology, Chinese PLA General Hospital, 28 Fu-Xing Road, Beijing 100853, China
| | - Shiwen Wu
- Department of Neurology, First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Yaping Tian
- Research Center for Birth Defects Prevention and Control Technology, Chinese PLA General Hospital, 28 Fu-Xing Road, Beijing 100853, China.
| |
Collapse
|
36
|
Weber FJ, Latshang TD, Blum MR, Kohler M, Wertli MM. Prognostic factors, disease course, and treatment efficacy in Duchenne muscular dystrophy: A systematic review and meta-analysis. Muscle Nerve 2022; 66:462-470. [PMID: 35860996 PMCID: PMC9804574 DOI: 10.1002/mus.27682] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 07/10/2022] [Accepted: 07/17/2022] [Indexed: 01/05/2023]
Abstract
INTRODUCTION/AIMS Prognostic factors in Duchenne muscular dystrophy (DMD) predict the disease course and may help individualize patient care. The aim was to summarize the evidence on prognostic factors that may support treatment decisions. METHODS We searched six databases for prospective studies that each included ≥50 DMD patients with a minimum follow-up of 1 y. Primary outcomes were age at loss of ambulation (LoA), pulmonary function (forced vital capacity percent of predicted, FVC%p), and heart failure. RESULTS Out of 5074 references, 59 studies were analyzed. Corticosteroid use was associated with a delayed LoA (pooled effect hazard ratio [HR] 0.42, 95% confidence interval [CI] 0.23-0.75, I2 94%), better pulmonary function tests (higher peak FVC%, prolonged time with FVC%p > 50%, and reduced need for assisted ventilation) and delayed cardiomyopathy. Longer corticosteroid treatment was associated with later LoA (>1 y compared to <1 y; pooled HR: 0.50, 95% CI 0.27-0.90) and early treatment start (aged <5 y) may be associated with early cardiomyopathy and higher fracture risk. Genotype appeared to be an independent driver of LoA in some studies. Higher baseline physical function tests (e.g., 6-minute walk test) were associated with delayed LoA. Left ventricular dysfunction and FVC <1 L increased and the use of angiotensin-converting enzyme (ACE) inhibitors reduced the risk of heart failure and death. Fusion surgery in scoliosis may potentially preserve pulmonary function. DISCUSSION Prognostic factors that may inform clinical decisions include age at corticosteroid treatment initiation and treatment duration, ACE-inhibitor use, baseline physical function tests, pulmonary function, and cardiac dysfunction.
Collapse
Affiliation(s)
- Fabio J Weber
- Sleep Disorders Center and Pulmonary Division, University Hospital Zurich, Zurich, Switzerland
| | - Tsogyal D Latshang
- Sleep Disorders Center and Pulmonary Division, University Hospital Zurich, Zurich, Switzerland.,Sleep Disorders Center and Pulmonary Division, Kantonsspital Graubuenden, Chur, Switzerland
| | - Manuel R Blum
- Department of General Internal Medicine, University Hospital Bern, University of Bern, Bern, Switzerland.,Institute of Primary Health Care (BIHAM), University of Bern, Bern, Switzerland
| | - Malcolm Kohler
- Sleep Disorders Center and Pulmonary Division, University Hospital Zurich, Zurich, Switzerland.,Zurich Center for Interdisciplinary Sleep Research, University of Zurich, Zurich, Switzerland
| | - Maria M Wertli
- Department of General Internal Medicine, University Hospital Bern, University of Bern, Bern, Switzerland.,Department of Internal Medicine, Cantonal Hospital Baden, Baden, Switzerland
| |
Collapse
|
37
|
Gosselin MRF, Mournetas V, Borczyk M, Verma S, Occhipinti A, Róg J, Bozycki L, Korostynski M, Robson SC, Angione C, Pinset C, Gorecki DC. Loss of full-length dystrophin expression results in major cell-autonomous abnormalities in proliferating myoblasts. eLife 2022; 11:e75521. [PMID: 36164827 PMCID: PMC9514850 DOI: 10.7554/elife.75521] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 09/02/2022] [Indexed: 12/05/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) affects myofibers and muscle stem cells, causing progressive muscle degeneration and repair defects. It was unknown whether dystrophic myoblasts-the effector cells of muscle growth and regeneration-are affected. Using transcriptomic, genome-scale metabolic modelling and functional analyses, we demonstrate, for the first time, convergent abnormalities in primary mouse and human dystrophic myoblasts. In Dmdmdx myoblasts lacking full-length dystrophin, the expression of 170 genes was significantly altered. Myod1 and key genes controlled by MyoD (Myog, Mymk, Mymx, epigenetic regulators, ECM interactors, calcium signalling and fibrosis genes) were significantly downregulated. Gene ontology analysis indicated enrichment in genes involved in muscle development and function. Functionally, we found increased myoblast proliferation, reduced chemotaxis and accelerated differentiation, which are all essential for myoregeneration. The defects were caused by the loss of expression of full-length dystrophin, as similar and not exacerbated alterations were observed in dystrophin-null Dmdmdx-βgeo myoblasts. Corresponding abnormalities were identified in human DMD primary myoblasts and a dystrophic mouse muscle cell line, confirming the cross-species and cell-autonomous nature of these defects. The genome-scale metabolic analysis in human DMD myoblasts showed alterations in the rate of glycolysis/gluconeogenesis, leukotriene metabolism, and mitochondrial beta-oxidation of various fatty acids. These results reveal the disease continuum: DMD defects in satellite cells, the myoblast dysfunction affecting muscle regeneration, which is insufficient to counteract muscle loss due to myofiber instability. Contrary to the established belief, our data demonstrate that DMD abnormalities occur in myoblasts, making these cells a novel therapeutic target for the treatment of this lethal disease.
Collapse
Affiliation(s)
- Maxime RF Gosselin
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
| | | | - Malgorzata Borczyk
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology PASKrakowPoland
| | - Suraj Verma
- School of Computing, Engineering and Digital Technologies, Teesside UniversityMiddlesbroughUnited Kingdom
| | - Annalisa Occhipinti
- School of Computing, Engineering and Digital Technologies, Teesside UniversityMiddlesbroughUnited Kingdom
| | - Justyna Róg
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental BiologyWarsawPoland
| | - Lukasz Bozycki
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental BiologyWarsawPoland
| | - Michal Korostynski
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology PASKrakowPoland
| | - Samuel C Robson
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
- Centre for Enzyme Innovation, University of PortsmouthPortsmouthUnited Kingdom
| | - Claudio Angione
- School of Computing, Engineering and Digital Technologies, Teesside UniversityMiddlesbroughUnited Kingdom
| | | | - Dariusz C Gorecki
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
| |
Collapse
|
38
|
Yoon JA, Park HE, Kim J, Son J, Shin YB. Cardiac and Pulmonary Management Status of Duchenne Muscular Dystrophy in South Korea Based on Data From the National Health Insurance Database. J Clin Neurol 2022; 18:522-528. [PMID: 36062769 PMCID: PMC9444560 DOI: 10.3988/jcn.2022.18.5.522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/14/2022] [Accepted: 02/14/2022] [Indexed: 11/18/2022] Open
Abstract
Background and Purpose The purpose of this study was to determine the cardiac and pulmonary management status of patients with Duchenne muscular dystrophy (DMD) in South Korea based on the Korean National Health Insurance database. Methods This study used data of patients with code G71.0 in the National Health Information database, and also those with the special case of code V012. Cardiac function was assessed based on whether echocardiography and 24-h electrocardiography were performed, as well as the frequency of these investigations. Furthermore, information on the use of angiotensin-converting enzyme inhibitors (ACEi), angiotensin II receptor blockers (ARB), and beta blockers (BB) in the drug benefit list was checked. Medical charge records were also checked regarding the prescription of respiratory functional assessments and treatments. Results The diagnostic criteria were met by 479 patients, with 41% of these patients receiving a cardiac evaluation, 29.8% being prescribed ACEi at 14.4±3.7 years of age, 59.5% undergoing pulmonary function tests, and 42.1% received pulmonary rehabilitation. The age at receiving ventilator support was 19.4±2.7 years. The frequency of cardiac and respiratory function tests increased with age, but the interval between tests was longer than the recent DMD care recommendations. The trend of taking ACEi, ARB, and BB for cardiac management in South Korea did not change during the study analysis period. Conclusions The findings of this study will contribute to recognizing the current status and the importance of applying an anticipatory approach to cardiopulmonary function in DMD patients.
Collapse
Affiliation(s)
- Jin A Yoon
- Department of Rehabilitation Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Ho Eun Park
- Department of Rehabilitation Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Jinmi Kim
- Department of Biostatistics, Clinical Trial Center, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Jungmin Son
- Department of Biostatistics, Clinical Trial Center, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Yong Beom Shin
- Department of Rehabilitation Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea.
| |
Collapse
|
39
|
Kosac A, Pesovic J, Radenkovic L, Brkusanin M, Radovanovic N, Djurisic M, Radivojevic D, Mladenovic J, Ostojic S, Kovacevic G, Kravljanac R, Savic Pavicevic D, Milic Rasic V. LTBP4, SPP1, and CD40 Variants: Genetic Modifiers of Duchenne Muscular Dystrophy Analyzed in Serbian Patients. Genes (Basel) 2022; 13:1385. [PMID: 36011296 PMCID: PMC9407083 DOI: 10.3390/genes13081385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Clinical course variability in Duchenne muscular dystrophy (DMD) is partially explained by the mutation location in the DMD gene and variants in modifier genes. We assessed the effect of the SPP1, CD40, and LTBP4 genes and DMD mutation location on loss of ambulation (LoA). METHODS SNPs in SPP1-rs28357094, LTBP4-rs2303729, rs1131620, rs1051303, rs10880, and CD40-rs1883832 were genotyped, and their effect was assessed by survival and hierarchical cluster analysis. RESULTS Patients on glucocorticoid corticosteroid (GC) therapy experienced LoA one year later (p = 0.04). The modifying effect of SPP1 and CD40 variants, as well as LTBP4 haplotypes, was not observed using a log-rank test and multivariant Cox regression analysis. Cluster analysis revealed two subgroups with statistical trends in differences in age at LoA. Almost all patients in the cluster with later LoA had the protective IAAM LTBP4 haplotype and statistically significantly fewer CD40 genotypes with harmful T allele and "distal" DMD mutations. CONCLUSIONS The modifying effect of SPP1, CD40, and LTBP4 was not replicated in Serbian patients, although our cohort was comparable in terms of its DMD mutation type distribution, SNP allele frequencies, and GC-positive effect with other European cohorts. Cluster analysis may be able to identify patient subgroups carrying a combination of the genetic variants that modify LoA.
Collapse
Affiliation(s)
- Ana Kosac
- Department of Neurology, Clinic of Neurology and Psychiatry for Children and Youth, 11000 Belgrade, Serbia
| | - Jovan Pesovic
- Centre for Human Molecular Genetics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Lana Radenkovic
- Centre for Human Molecular Genetics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Milos Brkusanin
- Centre for Human Molecular Genetics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Nemanja Radovanovic
- Centre for Human Molecular Genetics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | - Marina Djurisic
- Laboratory of Medical Genetics, Mother and Child Health Care Institute of Serbia “Dr Vukan Cupic”, 11000 Belgrade, Serbia
| | - Danijela Radivojevic
- Laboratory of Medical Genetics, Mother and Child Health Care Institute of Serbia “Dr Vukan Cupic”, 11000 Belgrade, Serbia
| | - Jelena Mladenovic
- Department of Neurology, Clinic of Neurology and Psychiatry for Children and Youth, 11000 Belgrade, Serbia
| | - Slavica Ostojic
- Department of Neurology, Mother and Child Health Care Institute of Serbia “Dr Vukan Cupic”, 11000 Belgrade, Serbia
| | - Gordana Kovacevic
- Department of Neurology, Mother and Child Health Care Institute of Serbia “Dr Vukan Cupic”, 11000 Belgrade, Serbia
| | - Ruzica Kravljanac
- Department of Neurology, Mother and Child Health Care Institute of Serbia “Dr Vukan Cupic”, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Dusanka Savic Pavicevic
- Centre for Human Molecular Genetics, Faculty of Biology, University of Belgrade, 11000 Belgrade, Serbia
| | | |
Collapse
|
40
|
Yoon JA, Park HE, Kim J, Son J, Shin YB. Corticosteroid use and bone health management for Duchenne muscular dystrophy in South Korea. Sci Rep 2022; 12:11300. [PMID: 35788153 PMCID: PMC9253151 DOI: 10.1038/s41598-022-15510-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/24/2022] [Indexed: 11/09/2022] Open
Abstract
This study aimed to determine the current corticosteroid use and bone health management status of patients with Duchenne muscular dystrophy (DMD) in South Korea. This is a national population-based study utilized information from the databased of Korean National Health Insurance Database. Database regarding bone status, spine radiography findings, bone mineral density, and laboratory test results were obtained, as well as the proportion of patients with spine and lower limb prostheses, occurrence of scoliosis, and age at scoliosis surgery. Deflazacort dose in the ambulant group (aged < 15 years) increased by age and year. The maintenance dose of prednisolone and deflazacort for the 15-19 years group decreased by year. Among the patients, 12.47% underwent spine radiography, 23.11% underwent dual-energy X-ray absorptiometry, and 22.7% underwent vitamin D tests. Moreover, 40.9% of the patients were prescribed vitamin D at a mean age of 14.6 ± 6.1 years, while 10.22% were prescribed bisphosphonate at 17.92 ± 3.4 years. Further, 16.1% of the patients underwent posterior spinal instrumentation and fusion at 14.4 ± 2.3 years and 5.3% underwent anterior spinal instrumentation and fusion at 14.4 ± 2.3 years. Ankle-foot orthosis and spine orthosis prescriptions were noted in 4.91% and 1.84% of patients, respectively. In this our study, the current corticosteroid use and bone health management status of DMD in South Korea has been presented. The dose prescription for corticosteroid and bone health monitoring did not reach to current recommendation.
Collapse
Affiliation(s)
- Jin A Yoon
- Department of Rehabilitation Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, 179 Gudeok-Ro Seo-Gu, Busan, 602-739, Republic of Korea
| | - Ho Eun Park
- Department of Rehabilitation Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, 179 Gudeok-Ro Seo-Gu, Busan, 602-739, Republic of Korea
| | - Jinmi Kim
- Department of Biostatistics, Clinical Trial Center, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Jungmin Son
- Department of Biostatistics, Clinical Trial Center, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Yong Beom Shin
- Department of Rehabilitation Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, 179 Gudeok-Ro Seo-Gu, Busan, 602-739, Republic of Korea.
| |
Collapse
|
41
|
Biggar WD, Skalsky A, McDonald CM. Comparing Deflazacort and Prednisone in Duchenne Muscular Dystrophy. J Neuromuscul Dis 2022; 9:463-476. [PMID: 35723111 PMCID: PMC9398085 DOI: 10.3233/jnd-210776] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Deflazacort and prednisone/prednisolone are the current standard of care for patients with Duchenne muscular dystrophy (DMD) based on evidence that they improve muscle strength, improve timed motor function, delay loss of ambulation, improve pulmonary function, reduce the need for scoliosis surgery, delay onset of cardiomyopathy, and increase survival. Both have been used off-label for many years (choice dependent on patient preference, cost, and geographic location) before FDA approval of deflazacort for DMD in 2017. In this review, we compare deflazacort and prednisone/prednisolone in terms of their key pharmacological features, relative efficacy, and safety profiles in patients with DMD. Differentiating features include lipid solubility, pharmacokinetics, changes in gene expression profiles, affinity for the mineralocorticoid receptor, and impact on glucose metabolism. Evidence from randomized clinical trials, prospective studies, meta-analyses, and post-hoc analyses suggests that patients receiving deflazacort experience similar or slower rates of functional decline compared with those receiving prednisone/prednisolone. Regarding side effects, weight gain and behavior side effects appear to be greater with prednisone/prednisolone than with deflazacort, whereas bone health, growth parameters, and cataracts appear worse with deflazacort.
Collapse
Affiliation(s)
- W Douglas Biggar
- University of Toronto, 15583 22nd Side Road, Georgetown, Ontario, Canada
| | - Andrew Skalsky
- University of California San Diego, Rady Children's Hospital San Diego, MC, San Diego, CA, USA
| | - Craig M McDonald
- University of California Davis Health, Departments of Physical Medicine & Rehabilitation and Pediatrics, Lawrence J. Ellison Ambulatory Care Center, Sacramento, CA, USA
| |
Collapse
|
42
|
Iff J, Zhong Y, Gupta D, Paul X, Tuttle E, Henricson E, Schrader R. Disease Progression Stages and Burden in Patients with Duchenne Muscular Dystrophy Using Administrative Claims Supplemented by Electronic Medical Records. Adv Ther 2022; 39:2906-2919. [PMID: 35460510 DOI: 10.1007/s12325-022-02117-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/07/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION This study aims to identify stages of Duchenne muscular dystrophy (DMD) and assess the disease burden by progression stage using real-world administrative claims supplemented by relevant electronic medical record (EMR) data. METHODS Claims and EMR data from the Decision Resources Group's Real World Data Repository (2011-2020) were used to identify patients with DMD by diagnosis code and to stratify them into four disease stages by diagnosis and procedure markers reflective of DMD progression. Clinical and medical history data from the Cooperative International Neuromuscular Research Group (CINRG) were used to validate the developed claims-based staging algorithm. The distribution and drivers by disease stage, as well as disease burden, were examined. RESULTS A total of 938 (94%) of patients with DMD identified in claims/EMR data had sufficient information for stage classification. Patients were classified by stage based on patient characteristics and the presence or absence of progression markers such as genetic testing, wheelchair usage, scoliosis treatment, or ventilation assistance. Average ages at stages 1-4 are 7, 13, 18, and 23 years, respectively. Using natural history data, the claims-based staging algorithm was validated with high sensitivity and specificity rates. Both healthcare resource utilization and medical charges increased by stage. For example, the average annualized total charges were $17,688 (stage 1), $36,868 (stage 2), $72,801 (stage 3), and $167,285 (stage 4). CONCLUSIONS Large-scale claims data supplemented by EMR data can be used to characterize DMD progression and evaluate disease burden which may inform the design of future real-world studies about DMD.
Collapse
Affiliation(s)
- Joel Iff
- Sarepta Therapeutics, Inc, Cambridge, MA, USA
| | - Yi Zhong
- Analysis Group, Inc., 1010 El Camino Real #310, Menlo Park, CA, 94025, USA.
| | - Deepshekhar Gupta
- Analysis Group, Inc., 1010 El Camino Real #310, Menlo Park, CA, 94025, USA
| | - Xander Paul
- Analysis Group, Inc., 1010 El Camino Real #310, Menlo Park, CA, 94025, USA
| | - Edward Tuttle
- Analysis Group, Inc., 1010 El Camino Real #310, Menlo Park, CA, 94025, USA
| | | | | |
Collapse
|
43
|
Cha JJ, Kim IS, Kim JY, Choi EY, Min PK, Yoon YW, Lee BK, Hong BK, Kwon HM, Cho HE, Choi WA, Kang SW, Rim SJ. The association between cardiac involvement and long-term clinical outcomes in patients with Duchenne muscular dystrophy. ESC Heart Fail 2022; 9:2199-2206. [PMID: 35579098 PMCID: PMC9288783 DOI: 10.1002/ehf2.13970] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 03/29/2022] [Accepted: 05/04/2022] [Indexed: 01/14/2023] Open
Abstract
Aims Despite advances in contemporary cardiopulmonary therapies, cardiomyopathy remains the leading cause of death in patients with Duchenne muscular dystrophy (DMD). Also, the long‐term clinical outcomes of patients with DMD and cardiomyopathy is unknown. This study investigated long‐term clinical outcomes and their associated factors in patients with late‐stage DMD. Methods and results A total of 116 patients with late‐stage DMD (age > 15 years) were enrolled in this retrospective study. All enrolled patients were followed up at a single tertiary referral hospital. LV systolic dysfunction was dichotomously defined as reduced [left ventricular ejection fraction (LVEF) ≤ 40%] vs. preserved [>40%] based on the initial echocardiographic result. The primary endpoint was all‐cause death. The secondary endpoint was a composite event defined as death or unexpected hospitalization due to cardiovascular reasons including chest pain, dyspnoea, and generalized oedema. The patients were divided into preserved (n = 84, 72.4%) and reduced LVEF groups (n = 32, 27.6%). The mean age was 20.8 ± 5.9 years, the mean disease duration, 8.8 ± 3.7 years, and the mean follow‐up duration, 1708 ± 659 days. For primary endpoint, the reduced LVEF group showed a lower rate of overall survival (Reduced LVEF vs. Preserved LVEF; 81.3% vs. 98.8%, log‐rank P = 0.005). In the multivariable Cox regression analysis, brain‐natriuretic peptide (BNP) level (adjusted hazard ratio [HR] 1.088, 95% confidence interval [CI] 1.019–1.162, P = 0.011) and diuretic use (adjusted HR 9.279, 95%CI 1.651–52.148, P = 0.011) were significant predictors of all‐cause death in patients with DMD. For the secondary endpoint, the reduced LVEF group had a lower rate of freedom from composite events than the preserved LVEF group (65.6% vs. 86.9%, log‐rank P = 0.005). In the multivariable Cox regression analysis, BNP level (adjusted HR 1.057, 95%CI 1.005–1.112, P = 0.032) and diuretic use (adjusted HR 4.189, 95% CI 1.704–10.296, P = 0.002) were significant predictors of the composite event in patients with DMD. Conclusions Patients with DMD and reduced LVEF had worse clinical outcomes than those with preserved LVEF. BNP level and diuretic use were associated with adverse clinical outcomes in patients with late‐stage DMD, irrespective of LVEF.
Collapse
Affiliation(s)
- Jung-Joon Cha
- Department of Cardiology, Cardiovascular Centre, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea.,Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - In-Soo Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.,Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Jong-Youn Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.,Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Eui-Young Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.,Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Pil-Ki Min
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.,Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Young Won Yoon
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.,Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Byoung Kwon Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.,Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Bum-Kee Hong
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.,Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hyuck Moon Kwon
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.,Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Han Eol Cho
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Korea.,Department of Rehabilitation Medicine and Pulmonary Rehabilitation Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Won Ah Choi
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Korea.,Department of Rehabilitation Medicine and Pulmonary Rehabilitation Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Seong-Woong Kang
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Korea.,Department of Rehabilitation Medicine and Pulmonary Rehabilitation Center, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Se-Joong Rim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea.,Division of Cardiology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
44
|
Mejia EJ, Lin KY, Okunowo O, Iacobellis KA, Matesanz SE, Brandsema JF, Wittlieb-Weber CA, Katcoff H, Griffis H, Edelson JB. Health Care Use of Cardiac Specialty Care in Children With Muscular Dystrophy in the United States. J Am Heart Assoc 2022; 11:e024722. [PMID: 35411787 PMCID: PMC9238456 DOI: 10.1161/jaha.121.024722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background Duchenne and Becker muscular dystrophy are progressive disorders associated with cardiac mortality. Guidelines recommend routine surveillance; we assess cardiac resource use and identify gaps in care delivery. Methods and Results Male patients, aged 1 to 18 years, with Duchenne and Becker muscular dystrophy between January 2013 and December 2017 were identified in the IBM MarketScan Research Database. The cohort was divided into <10 and 10 to 18 years of age. The primary outcome was rate of annual health care resource per person year. Resource use was assessed for place of service, cardiac testing, and medications. Adjusted incidence rate ratios (IRRs) were estimated using a Poisson regression model. Medication use was measured by proportion of days covered. There were 1386 patients with a median follow‐up time of 3.0 years (interquartile range, 1.9–4.7 years). Patients in the 10 to 18 years group had only 0.40 (95% CI, 0.35–0.45) cardiology visits per person year and 0.66 (95% CI, 0.62–0.70) echocardiography/magnetic resonance imaging per person year. Older patients had higher rates of inpatient admissions (IRR, 1.46; 95% CI, 1.03–2.09), outpatient cardiology visits (IRR, 2.0; 95% CI, 1.66–2.40), cardiac imaging (IRR, 1.59; 95% CI, 1.40–1.80), and Holter monitoring (IRR, 3.33; 95% CI, 2.35–4.73). A proportion of days covered >80% for angiotensin‐converting enzyme inhibitors/angiotensin receptor blockers was observed in 13.6% (419/3083) of total person years among patients in the 10 to 18 years group. Conclusions Children 10 to 18 years of age have higher rates of cardiac resource use compared with those <10 years of age. However, rates in both age groups fall short of guidelines. Opportunities exist to identify barriers to resource use and optimize cardiac care for patients with Duchenne and Becker muscular dystrophy.
Collapse
Affiliation(s)
- Erika J Mejia
- Division of Cardiology Children's Hospital of PhiladelphiaUniversity of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| | - Kimberly Y Lin
- Division of Cardiology Children's Hospital of PhiladelphiaUniversity of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| | - Oluwatimilehin Okunowo
- Data Science & Biostatistics Unit Department of Biomedical and Health Informatics Children's Hospital of Philadelphia University of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| | - Katherine A Iacobellis
- Division of Cardiology Children's Hospital of PhiladelphiaUniversity of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| | - Susan E Matesanz
- Division of Neurology Children's Hospital of Philadelphia University of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| | - John F Brandsema
- Division of Neurology Children's Hospital of Philadelphia University of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| | - Carol A Wittlieb-Weber
- Division of Cardiology Children's Hospital of PhiladelphiaUniversity of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| | - Hannah Katcoff
- Data Science & Biostatistics Unit Department of Biomedical and Health Informatics Children's Hospital of Philadelphia University of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| | - Heather Griffis
- Data Science & Biostatistics Unit Department of Biomedical and Health Informatics Children's Hospital of Philadelphia University of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| | - Jonathan B Edelson
- Division of Cardiology Children's Hospital of PhiladelphiaUniversity of Pennsylvania Perelman School of Medicine Philadelphia Pennsylvania
| |
Collapse
|
45
|
Diversity of Dystrophin Gene Mutations and Disease Progression in a Contemporary Cohort of Duchenne Muscular Dystrophy. Pediatr Cardiol 2022; 43:855-867. [PMID: 35064276 DOI: 10.1007/s00246-021-02797-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 11/24/2021] [Indexed: 01/14/2023]
Abstract
Abnormal dystrophin production due to mutations in the dystrophin gene causes Duchenne Muscular Dystrophy (DMD). Cases demonstrate considerable genetic and disease progression variability. It is unclear if specific gene mutations are prognostic of outcomes in this population. We conducted a retrospective cohort study of DMD patients followed at 17 centers across the USA and Canada from 2005 to 2015 with goal of understanding the genetic variability of DMD and its impact on clinical outcomes. Cumulative incidence of clinically relevant outcomes was stratified by genetic mutation type, exon mutation location, and extent of exon deletion. Of 436 males with DMD, 324 (74.3%) underwent genetic testing. Deletions were the most common mutation type (256, 79%), followed by point mutations (45, 13.9%) and duplications (23, 7.1%). There were 131 combinations of mutations with most mutations located along exons 45 to 52. The number of exons deleted varied between 1 and 52 with a median of 3 exons deleted (IQR 1-6). Subjects with mutations starting at exon positions 40-54 had a later onset of arrhythmias occurring at median age 25 years (95% CI 18-∞), p = 0.01. Loss of ambulation occurred later at median age of 13 years (95% CI 12-15) in subjects with mutations that started between exons 55-79, p = 0.01. There was no association between mutation type or location and onset of cardiac dysfunction. We report the genetic variability in DMD and its association with timing of clinical outcomes. Genetic modifiers may explain some phenotypic variability.
Collapse
|
46
|
Ferizovic N, Summers J, de Zárate IBO, Werner C, Jiang J, Landfeldt E, Buesch K. Prognostic indicators of disease progression in Duchenne muscular dystrophy: A literature review and evidence synthesis. PLoS One 2022; 17:e0265879. [PMID: 35333888 PMCID: PMC8956179 DOI: 10.1371/journal.pone.0265879] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 03/09/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a rare, severely debilitating, and fatal neuromuscular disease characterized by progressive muscle degeneration. Like in many orphan diseases, randomized controlled trials are uncommon in DMD, resulting in the need to indirectly compare treatment effects, for example by pooling individual patient-level data from multiple sources. However, to derive reliable estimates, it is necessary to ensure that the samples considered are comparable with respect to factors significantly affecting the clinical progression of the disease. To help inform such analyses, the objective of this study was to review and synthesise published evidence of prognostic indicators of disease progression in DMD. We searched MEDLINE (via Ovid), Embase (via Ovid) and the Cochrane Library (via Wiley) for records published from inception up until April 23 2021, reporting evidence of prognostic indicators of disease progression in DMD. Risk of bias was established with the grading system of the Centre for Evidence-Based Medicine (CEBM). RESULTS Our search included 135 studies involving 25,610 patients from 18 countries across six continents (Africa, Asia, Australia, Europe, North America and South America). We identified a total of 23 prognostic indicators of disease progression in DMD, namely age at diagnosis, age at onset of symptoms, ataluren treatment, ATL1102, BMI, cardiac medication, DMD genetic modifiers, DMD mutation type, drisapersen, edasalonexent, eteplirsen, glucocorticoid exposure, height, idebenone, lower limb surgery, orthoses, oxandrolone, spinal surgery, TAS-205, vamorolone, vitlolarsen, ventilation support, and weight. Of these, cardiac medication, DMD genetic modifiers, DMD mutation type, and glucocorticoid exposure were designated core prognostic indicators, each supported by a high level of evidence and significantly affecting a wide range of clinical outcomes. CONCLUSION This study provides a current summary of prognostic indicators of disease progression in DMD, which will help inform the design of comparative analyses and future data collection initiatives in this patient population.
Collapse
Affiliation(s)
- Nermina Ferizovic
- MAP BioPharma Ltd, Cambridge, England, United Kingdom
- BresMed Health Solutions, Sheffield, England, United Kingdom
| | | | | | | | - Joel Jiang
- PTC Therapeutics, South Plainfield, New Jersey, United States of America
| | | | | |
Collapse
|
47
|
Handberg C, Werlauff U, Højberg AL. Perspectives on Everyday Life Challenges of Danish Young People With Duchenne Muscular Dystrophy (DMD) on Corticosteroids. Glob Qual Nurs Res 2022; 9:23333936221094858. [PMID: 35493771 PMCID: PMC9052227 DOI: 10.1177/23333936221094858] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 12/03/2022] Open
Abstract
This study aimed to investigate perspectives on everyday life challenges of young persons with Duchenne muscular dystrophy in Denmark treated with corticosteroids perceived by young persons and their parents to improve rehabilitation interventions. Nineteen semi-structured interviews were conducted: 10 individual interviews with 10 persons with DMD and six individual interviews with parents (five mothers and one father) and three couple interviews (three mothers and three fathers). The analysis was guided by interpretive description methodology and Antonovsky’s Sense of Coherence theory. The results indicated that persons with Duchenne muscular dystrophy existed in a flux between experiencing greater Sense of Coherence revolving around normality and less Sense of Coherence exposing their vulnerability which unfolded in four opposing themes: (1) bodily ability and disability, (2) content and anxious, (3) sociable and lonely, and (4) independent and dependent. Future rehabilitation should aim at supporting resistance resources promoting bodily ability, being content, sociable, and independent.
Collapse
Affiliation(s)
- Charlotte Handberg
- National Rehabilitation Center for Neuromuscular Diseases, Aarhus, Denmark.,Department of Public Health, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Ulla Werlauff
- National Rehabilitation Center for Neuromuscular Diseases, Aarhus, Denmark
| | | |
Collapse
|
48
|
Influence of β2 Adrenergic Receptor Genotype on Longitudinal Measures of Forced Vital Capacity in Patients with Duchenne Muscular Dystrophy. Neuromuscul Disord 2022; 32:150-158. [DOI: 10.1016/j.nmd.2021.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 11/23/2021] [Accepted: 12/29/2021] [Indexed: 11/21/2022]
|
49
|
Abdelgalil Ali Ahmed S, Adam Essa ME, Ahmed AF, Elagib EM, Ahmed Eltahir NI, Awadallah H, Hassan A, Khair ASM, Ebad MAB. Incidence and Clinical Pattern of Mixed Connective Tissue Disease in Sudanese Patients at Omdurman Military Hospital: Hospital-Based Study. Open Access Rheumatol 2021; 13:333-341. [PMID: 34916856 PMCID: PMC8668256 DOI: 10.2147/oarrr.s335206] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022] Open
Abstract
Background Mixed connective tissue disease (MCTD) is a rare autoimmune disease, characterized by the production of specific autoantibody anti-RNP, which presents with varied overlapping symptoms of different connective tissue disorders. The aim of this study is to identify the frequency and patterns of MCTD. Methods This is a descriptive cross-sectional hospital-based study conducted at the rheumatology clinic at Omdurman Military Hospital between February 2019 and July 2019. The study included 30 patients and data were collected using a designated questionnaire. Results The study showed that the majority of patients (96.7%) were females and only 3.3% was male. About 30% of the patients aged between 30 and 39 years were the most affected. As a first diagnosis, 10% of the patients had a MCTD fulfilling the Alarcon-Segovia criteria. The remaining 90% of the patients were diagnosed with other diseases before evolving into MCTD. The most common clinical presentation was arthralgia in 100% of the patients, 90% were symmetrically followed by myositis in 70% of the patients, arthritis in 63.3% of the patients, puffy fingers in 63.3% of the patients, and hand swelling in 60% as major musculoskeletal symptoms. Regarding the initial results in immunological profile, the most common positive autoantibodies among the patients were anti-RNP titer in 96.7% of the patients, ANA in 90%, anti-Sm in 50%, RF in 50%, anti-Ds DNA in 46.7%, and anti-Ro in 43.3%. Conclusion This study showed that MCTD is more common in females, only 10% of patients presented with a fulfilling criteria of the disease at diagnosis, and the rest of the patients presented with other rheumatologic diseases before evolving into MCTD.
Collapse
Affiliation(s)
- Sulafah Abdelgalil Ali Ahmed
- Department of Medicine, Faculty of Medicine, Ahfad University, Khartoum, Sudan.,Department of Rheumatology, Omdurman Military Hospital, Khartoum, Sudan
| | - Mohammed Elmujtba Adam Essa
- Department of Clinical Medicine, Medical and Cancer Research Institute, Nyala, Sudan.,Faculty of Medicine, Al Fashir University, Al Fashir, Sudan
| | - Amar F Ahmed
- Department of Internal Medicine, Wayne State University, Detroit, MI, USA
| | - Elnour Mohammed Elagib
- Department of Rheumatology, Omdurman Military Hospital, Khartoum, Sudan.,Department of Medicine, Faculty of Medicine, Karrary University, Khartoum, Sudan
| | - Noha Ibrahim Ahmed Eltahir
- Department of Rheumatology, Omdurman Military Hospital, Khartoum, Sudan.,Department of Medicine, Faculty of Medicine, Karrary University, Khartoum, Sudan
| | - Huyam Awadallah
- Department of Internal Medicine, Detroit Medical Centre, Detroit, MI, USA
| | - Abubakr Hassan
- Department of Clinical Medicine, Medical and Cancer Research Institute, Nyala, Sudan
| | | | | |
Collapse
|
50
|
Szabo SM, Gooch KL, Mickle AT, Salhany RM, Connolly AM. The impact of genotype on outcomes in individuals with Duchenne muscular dystrophy: A systematic review. Muscle Nerve 2021; 65:266-277. [PMID: 34878187 DOI: 10.1002/mus.27463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 11/07/2022]
Abstract
Duchenne muscular dystrophy (DMD) is associated with progressive muscle weakness, loss of ambulation (LOA), and early mortality. In this review we have synthesized published data on the clinical course of DMD by genotype. Using a systematic search implemented in Medline and Embase, 53 articles were identified that describe the clinical course of DMD, with pathogenic variants categorizable by exon skip or stop-codon readthrough amenability and outcomes presented by age. Outcomes described included those related to ambulatory, cardiac, pulmonary, or cognitive function. Estimates of the mean (95% confidence interval) age at LOA ranged from 9.1 (8.7-9.6) years among 90 patients amenable to skipping exon 53 to 11.5 (9.5-13.5) years among three patients amenable to skipping exon 8. Although function worsened with age, the impact of genotype was less clear for other outcomes (eg, forced vital capacity and left ventricular ejection fraction). Understanding the distribution of pathogenic variants is important for studies in DMD, as this research suggests major differences in the natural history of disease. In addition, specific details of the use of key medications, including corticosteroids, antisense oligonucleotides, and cardiac medications, should be reported.
Collapse
Affiliation(s)
- Shelagh M Szabo
- Broadstreet Heath Economics & Outcomes Research, Vancouver, British Columbia, Canada
| | | | - Alexis T Mickle
- Broadstreet Heath Economics & Outcomes Research, Vancouver, British Columbia, Canada
| | | | - Anne M Connolly
- Division of Neurology, Nationwide Children's Hospital, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|