1
|
Koehler S, Kuhm J, Huffaker T, Young D, Tandon A, André F, Frey N, Greil G, Hussain T, Engelhardt S. Deep Learning-based Aligned Strain from Cine Cardiac MRI for Detection of Fibrotic Myocardial Tissue in Patients with Duchenne Muscular Dystrophy. Radiol Artif Intell 2025; 7:e240303. [PMID: 40008976 DOI: 10.1148/ryai.240303] [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] [Indexed: 02/27/2025]
Abstract
Purpose To develop a deep learning (DL) model that derives aligned strain values from cine (noncontrast) cardiac MRI and evaluate performance of these values to predict myocardial fibrosis in patients with Duchenne muscular dystrophy (DMD). Materials and Methods This retrospective study included 139 male patients with DMD who underwent cardiac MRI at a single center between February 2018 and April 2023. A DL pipeline was developed to detect five key frames throughout the cardiac cycle and respective dense deformation fields, allowing for phase-specific strain analysis across patients and from one key frame to the next. Effectiveness of these strain values in identifying abnormal deformations associated with fibrotic segments was evaluated in 57 patients (mean age [± SD], 15.2 years ± 3.1), and reproducibility was assessed in 82 patients by comparing the study method with existing feature-tracking and DL-based methods. Statistical analysis compared strain values using t tests, mixed models, and more than 2000 machine learning models; accuracy, F1 score, sensitivity, and specificity are reported. Results DL-based aligned strain identified five times more differences (29 vs five; P < .01) between fibrotic and nonfibrotic segments compared with traditional strain values and identified abnormal diastolic deformation patterns often missed with traditional methods. In addition, aligned strain values enhanced performance of predictive models for myocardial fibrosis detection, improving specificity by 40%, overall accuracy by 17%, and accuracy in patients with preserved ejection fraction by 61%. Conclusion The proposed aligned strain technique enables motion-based detection of myocardial dysfunction at noncontrast cardiac MRI, facilitating detailed interpatient strain analysis and allowing precise tracking of disease progression in DMD. Keywords: Pediatrics, Image Postprocessing, Heart, Cardiac, Convolutional Neural Network (CNN) Duchenne Muscular Dystrophy Supplemental material is available for this article. © RSNA, 2025.
Collapse
Affiliation(s)
- Sven Koehler
- Department of Internal Medicine III, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Sites Heidelberg and Mannheim, Germany
- Medical Faculty of University Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Julian Kuhm
- Department of Internal Medicine III, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Sites Heidelberg and Mannheim, Germany
| | - Tyler Huffaker
- Division of Pediatric Cardiology, Department of Pediatrics, UT Southwestern/Children's Health, Dallas, Tex
| | - Daniel Young
- Division of Pediatric Cardiology, Department of Pediatrics, UT Southwestern/Children's Health, Dallas, Tex
| | - Animesh Tandon
- Department of Heart, Vascular, and Thoracic, Children's Institute; Cleveland Clinic Children's Centre for Artificial Intelligence (C4AI); and Cardiovascular Innovation Research Centre, Cleveland Children's Clinic, Cleveland, Ohio
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Florian André
- Department of Internal Medicine III, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Sites Heidelberg and Mannheim, Germany
- Medical Faculty of University Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Norbert Frey
- Department of Internal Medicine III, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Sites Heidelberg and Mannheim, Germany
- Medical Faculty of University Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Gerald Greil
- Division of Pediatric Cardiology, Department of Pediatrics, UT Southwestern/Children's Health, Dallas, Tex
| | - Tarique Hussain
- Division of Pediatric Cardiology, Department of Pediatrics, UT Southwestern/Children's Health, Dallas, Tex
| | - Sandy Engelhardt
- Department of Internal Medicine III, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Sites Heidelberg and Mannheim, Germany
- Medical Faculty of University Heidelberg, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
2
|
Lupu M, Marcu MA, Epure DA, Vladacenco OA, Severin EM, Teleanu RI. Lost in Transition: Challenges in the Journey from Pediatric to Adult Care for a Romanian DMD Patient. Healthcare (Basel) 2025; 13:830. [PMID: 40218127 PMCID: PMC11988853 DOI: 10.3390/healthcare13070830] [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: 01/26/2025] [Revised: 03/05/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND The transition from pediatric to adult care in Duchenne Muscular Dystrophy (DMD) is challenging due to the disease's complexity and the need for lifelong, comprehensive management. In Romania, ongoing efforts aim to enhance multidisciplinary collaboration, though systemic barriers, such as fragmented healthcare services, persist. Nonsense mutations, including those in exon 30 described here, are often associated with more severe disease progression. METHODS We present the case of a 17-year-old Romanian DMD patient with a nonsense mutation in exon 30 of the dystrophin gene. The patient received multidisciplinary pediatric care addressing his medical needs, including neuromuscular, respiratory, cardiac, and orthopedic management. Transition readiness was assessed using the Transition Readiness Assessment Questionnaire (TRAQ), and the patient's perspective on the process was documented. RESULTS Care followed international standards, but the disease progressed predictably, with gradual loss of ambulation, respiratory decline, and cardiac complications. The TRAQ revealed strengths in communication with healthcare providers but moderate confidence in self-management tasks. From the patient's perspective, fragmented adult services and difficulty accessing specialized neuromuscular support remain major obstacles, underscoring the importance of early, structured transition planning and patient-centered approaches. CONCLUSIONS Transitioning to adult services requires strong communication between pediatric and adult teams and integration of specialized care. Tailored follow-up plans ensure continuity of care and effective disease management. This case reflects broader needs in similar healthcare contexts, highlighting the necessity of robust transition frameworks to respond to patient-specific challenges and ultimately support long-term quality of life.
Collapse
Affiliation(s)
- Maria Lupu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.L.); (O.A.V.); (E.M.S.); (R.I.T.)
| | - Maria-Alexandra Marcu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.L.); (O.A.V.); (E.M.S.); (R.I.T.)
| | - Diana Anamaria Epure
- Department of Paediatric Neurology, Dr Victor Gomoiu Children’s Hospital, 022102 Bucharest, Romania;
| | - Oana Aurelia Vladacenco
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.L.); (O.A.V.); (E.M.S.); (R.I.T.)
- Department of Paediatric Neurology, Dr Victor Gomoiu Children’s Hospital, 022102 Bucharest, Romania;
| | - Emilia Maria Severin
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.L.); (O.A.V.); (E.M.S.); (R.I.T.)
| | - Raluca Ioana Teleanu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.L.); (O.A.V.); (E.M.S.); (R.I.T.)
- Department of Paediatric Neurology, Dr Victor Gomoiu Children’s Hospital, 022102 Bucharest, Romania;
| |
Collapse
|
3
|
Conway KM, Thomas S, Neyaz T, Ciafaloni E, Mann JR, Staron‐Ehlinger M, Beasley GS, Romitti PA, Mathews KD, the Muscular Dystrophy Surveillance, Tracking and Research Network (MD STAR net). Prophylactic Use of Cardiac Medications and Survival in Duchenne Muscular Dystrophy. Muscle Nerve 2025; 71:574-582. [PMID: 39853770 PMCID: PMC11887528 DOI: 10.1002/mus.28353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/26/2025]
Abstract
INTRODUCTION/AIMS Prophylactic treatment of left ventricular dysfunction (LVD) in Duchenne muscular dystrophy (DMD) delays onset of LVD, but there is limited data showing impact on survival. Our aim was to describe survival among treated and untreated individuals with DMD. METHODS Retrospective, population-based surveillance data from the Muscular Dystrophy Surveillance, Tracking and Research Network (MD STARnet) were used. We analyzed 327 males with DMD born between 1982 and 2009 who were at least 6 years old at the last visit and who initiated cardiac prophylactic medication before age 14 years. Death status was ascertained through vital record linkages and medical record review. Prophylaxis was defined as cardiac medication use at least 1 year before LVD onset (ejection fraction < 55% or shortening fraction < 28%). Age at first visit, corticosteroid use, scoliosis surgery, initiation of noninvasive ventilation, and loss of ambulation were also coded. Cox Proportional Hazard modeling with time-varying covariates describes associations. RESULTS Prophylactic cardiac treatment was documented for 27.7% (n = 90); corticosteroids were used by 60.9% (n = 157). Adjusting for age at first visit and MD STARnet site, prophylactic treatment was associated with a 54% lower hazard of death (HR = 0.46, 95% CI = 0.22-0.93) compared to no prophylaxis. Adjusting for selected clinical covariates did not appreciably change the estimate (HR = 0.46, 95% CI = 0.22-0.99). DISCUSSION Initiation of cardiac medication when left ventricular function is normal was associated with prolonged survival in this study of males with DMD. Only one-quarter of individuals received this treatment, however, indicating a topic of focus for improving care.
Collapse
Affiliation(s)
- Kristin M. Conway
- Department of Epidemiology, College of Public HealthThe University of IowaIowa CityIowaUSA
| | - Shiny Thomas
- New York State Department of HealthAlbanyNew YorkUSA
| | - Tahereh Neyaz
- Department of Epidemiology, College of Public HealthThe University of IowaIowa CityIowaUSA
| | - Emma Ciafaloni
- Department of Neurology, School of Medicine and DentistryUniversity of Rochester Medical CenterRochesterNew YorkUSA
| | - Joshua R. Mann
- Department of Preventive Medicine, School of Medicine and John D. Bower School of Population HealthUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Michelle Staron‐Ehlinger
- Stead Family Department of Pediatrics, Roy J. and Lucille A. Carver College of MedicineThe University of IowaIowa CityIowaUSA
| | - Gary S. Beasley
- Stead Family Department of Pediatrics, Roy J. and Lucille A. Carver College of MedicineThe University of IowaIowa CityIowaUSA
| | - Paul A. Romitti
- Department of Epidemiology, College of Public HealthThe University of IowaIowa CityIowaUSA
| | - Katherine D. Mathews
- Stead Family Department of Pediatrics, Roy J. and Lucille A. Carver College of MedicineThe University of IowaIowa CityIowaUSA
| | | |
Collapse
|
4
|
Etxaniz U, Marks I, Albin T, Diaz M, Bhardwaj R, Anderson A, Tyaglo O, Hoang T, Missinato MA, Svensson K, Badillo B, Kovach PR, Leung L, Cochran M, Kwon HW, Ahad Shah MN, Maruyama R, Yokota T, Doppalapudi VR, Darimont B, Younis H, Flanagan WM, Levin AA, Huang H, Karamanlidis G. AOC 1044 induces exon 44 skipping and restores dystrophin protein in preclinical models of Duchenne muscular dystrophy. Nucleic Acids Res 2025; 53:gkaf241. [PMID: 40183632 PMCID: PMC11969676 DOI: 10.1093/nar/gkaf241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 03/05/2025] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe disorder caused by mutations in the dystrophin gene, resulting in loss of functional dystrophin protein in muscle. While phosphorodiamidate morpholino oligomers (PMOs) are promising exon-skipping therapeutics aimed at restoring dystrophin expression, their effectiveness is often limited by poor muscle delivery. We developed AOC 1044, an antibody-oligonucleotide conjugate (AOC) that combines a PMO-targeting exon 44 with an antibody against the transferrin receptor (TfR1), enhancing delivery to muscle tissues for patients with DMD amenable to exon 44 skipping (DMD44). AOC 1044 induces dose-dependent exon 44 skipping and its mouse-active variant elicited dose-dependent dystrophin restoration in skeletal and cardiac muscle in a DMD mouse model. This treatment also reduced muscle damage, as evidenced by decreases in serum creatine kinase and key liver enzymes, suggesting that restored dystrophin is functionally active. In nonhuman primates, single or repeated AOC 1044 doses resulted in dose-dependent increases in PMO concentration and exon 44 skipping across a range of muscle tissues, including the heart. Collectively, these findings highlight AOC 1044 as a promising therapeutic candidate for patients with DMD44, offering improved muscle targeting and meaningful dystrophin restoration, with potential clinical benefits in reducing muscle degeneration.
Collapse
MESH Headings
- Animals
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/therapy
- Dystrophin/genetics
- Dystrophin/metabolism
- Exons
- Mice
- Disease Models, Animal
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/pathology
- Morpholinos
- Humans
- Mice, Inbred mdx
- Male
- Receptors, Transferrin/immunology
- Receptors, Transferrin/antagonists & inhibitors
Collapse
Affiliation(s)
- Usue Etxaniz
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Isaac Marks
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Tyler Albin
- Seawolf Therapeutics, 9880 Campus Point Drive, Suite 210, San Diego, CA 92121, United States
| | - Matthew Diaz
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Raghav Bhardwaj
- Broad Institute of MIT and Harvard, 75 Ames Street, Cambridge, MA 02142, United States
| | - Aaron Anderson
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Olecya Tyaglo
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Tiffany Hoang
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Maria Azzurra Missinato
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Kristoffer Svensson
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Ben Badillo
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Philip R Kovach
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Laura Leung
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Michael Cochran
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Hae Won Kwon
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Md Nur Ahad Shah
- Yokota Lab, Department of Medical Genetics, University of Alberta, Edmonton,T6G 2H, Canada
| | - Rika Maruyama
- Yokota Lab, Department of Medical Genetics, University of Alberta, Edmonton,T6G 2H, Canada
| | - Toshifumi Yokota
- Yokota Lab, Department of Medical Genetics, University of Alberta, Edmonton,T6G 2H, Canada
| | - Venkata R Doppalapudi
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Beatrice Darimont
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Husam S Younis
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - W Michael Flanagan
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Arthur A Levin
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Hanhua Huang
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| | - Georgios Karamanlidis
- Avidity Biosciences, Inc., 10578 Science Drive, Suite 125, San Diego, CA 92121, United States
| |
Collapse
|
5
|
Blackstone WZ, Malaguit SE, Shwaish NS, Frandsen EL. Contiguous Xp21 deletion involving Duchenne muscular dystrophy and McLeod neuroacanthocytosis syndrome results in rapidly progressive and fatal cardiomyopathy. Cardiol Young 2025; 35:358-360. [PMID: 39749981 DOI: 10.1017/s1047951124036370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Dilated cardiomyopathy is an expected manifestation and common cause of death in patients with Duchenne muscular dystrophy. We present an unusually rapid progression of cardiomyopathy in a boy with Duchenne muscular dystrophy. Expanded genetic testing revealed a contiguous Xp21 deletion involving dystrophin and XK genes, responsible for Duchenne muscular dystrophy and McLeod neuroacanthocytosis syndrome, respectively, resulting in a more severe cardiac phenotype.
Collapse
Affiliation(s)
| | - Seth E Malaguit
- Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Natalie S Shwaish
- Loma Linda Children's Hospital, Department of Pediatric Cardiology, Loma Linda, CA, USA
| | - Erik L Frandsen
- Loma Linda Children's Hospital, Department of Pediatric Cardiology, Loma Linda, CA, USA
| |
Collapse
|
6
|
Liu ZQ, Maforo NG, Magrath P, Prosper A, Renella P, Halnon N, Wu HH, Ennis DB. MRI-Based Circumferential Strain in Boys with Early Duchenne Muscular Dystrophy Cardiomyopathy. Diagnostics (Basel) 2024; 14:2673. [PMID: 39682580 DOI: 10.3390/diagnostics14232673] [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: 10/11/2024] [Revised: 11/14/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Background: In boys with Duchenne muscular dystrophy (DMD), cardiomyopathy has become the primary cause of death. Although both positive late gadolinium enhancement (LGE) and reduced left ventricular ejection fraction (LVEF) are late findings in a DMD cohort, LV end-systolic circumferential strain at middle wall (Ecc) serves as a biomarker for detecting early impairment in cardiac function associated with DMD. However, Ecc derived from cine Displacement Encoding with Stimulated Echoes (DENSE) has not been quantified in boys with DMD. We aim to: (1) use cine DENSE to quantify regional Ecc in LGE negative (-) boys with DMD and healthy controls; and (2) compare Ecc with LVEF in terms of differentiating DMD boys with LGE (-) from healthy boys. Methods: 10 LGE (-) boys with DMD and 12 healthy boys were enrolled prospectively in an IRB-approved study for CMR at 3T. Navigator-gated cine DENSE was used to obtain short-axis mid-ventricular data and estimate global and regional Ecc. Group-wise differences were tested via a Wilcoxon rank-sum test. Within-group differences were tested via a Skillings-Mack test followed by pairwise Wilcoxon signed-rank tests. A binomial logistic regression model was adopted to differentiate between DMD boys with LGE (-) and healthy boys. Results: When compared to healthy boys, LGE (-) boys with DMD demonstrated significantly impaired septal Ecc [-0.13 (0.01) vs. -0.16 (0.03), p = 0.019]. In comparison to the Ecc in other segments, both groups of boys exhibited significantly reduced septal Ecc and significantly elevated lateral Ecc. Septal Ecc outperformed LVEF in distinguishing DMD boys with LGE (-) from healthy boys. Conclusions: Reduced septal Ecc may serve as an early indicator of cardiac involvement in LGE (-) DMD boys prior to reduced LVEF and a positive LGE finding.
Collapse
Affiliation(s)
- Zhan-Qiu Liu
- Department of Radiology, Stanford University, Palo Alto, CA 94305, USA
- Cardiovascular Institute, Stanford University, Palo Alto, CA 94305, USA
| | - Nyasha G Maforo
- Physics and Biology in Medicine Interdepartmental Program, University of California, Los Angeles, CA 90095, USA
- Department of Radiological Sciences, University of California, Los Angeles, CA 90095, USA
| | - Patrick Magrath
- Department of Radiological Sciences, University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Ashley Prosper
- Department of Radiological Sciences, University of California, Los Angeles, CA 90095, USA
| | - Pierangelo Renella
- Department of Radiological Sciences, University of California, Los Angeles, CA 90095, USA
- Department of Medicine, Division of Pediatric Cardiology, CHOC Children's Hospital, Orange, CA 92868, USA
| | - Nancy Halnon
- Department of Pediatrics, University of California, Los Angeles, CA 90095, USA
| | - Holden H Wu
- Physics and Biology in Medicine Interdepartmental Program, University of California, Los Angeles, CA 90095, USA
- Department of Radiological Sciences, University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Daniel B Ennis
- Department of Radiology, Stanford University, Palo Alto, CA 94305, USA
- Cardiovascular Institute, Stanford University, Palo Alto, CA 94305, USA
- Maternal & Child Health Research Institute, Stanford University, Palo Alto, CA 94305, USA
| |
Collapse
|
7
|
Palm A, Ekström M, Emilsson Ö, Ersson K, Ljunggren M, Sundh J, Grote L. Control of hypercapnia and mortality in home mechanical ventilation: the population-based DISCOVERY study. ERJ Open Res 2024; 10:00461-2024. [PMID: 39655175 PMCID: PMC11626622 DOI: 10.1183/23120541.00461-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 06/26/2024] [Indexed: 12/12/2024] Open
Abstract
Background Studies on the survival of patients with home mechanical ventilation (HMV) are sparse. We aimed to analyse the impact of controlled hypercapnia on survival over 27 years among patients with HMV in Sweden. Study design and methods Population-based cohort study of adult patients starting HMV in the Swedish Registry for Respiratory Failure (Swedevox) during 1996-2022 cross-linked with the National Cause of Death registry. Mortality risk factors were analysed using crude and multivariable Cox regression models, including adjustments for anthropometrics, comorbidities, the underlying diagnosis causing chronic hypercapnic respiratory failure (CRF) and the control of hypercapnia (P aCO2 ≤6.0 kPa) at follow-up. Results We included 10 190 patients (50.1% women, age 62.9±14.5 years). Control of hypercapnia at follow-up after 1.3±0.9 years was associated with lower mortality, hazard ratio (HR) 0.74 (95% CI 0.68-0.80) and the association was strongest in those with pulmonary disease, restrictive thoracal disease (RTD), obesity hypoventilation syndrome (OHS) and amyotrophic lateral sclerosis (ALS). Predictors for increased mortality included age, Charlson Comorbidity Index, supplemental oxygen therapy and acute start of HMV therapy. Median survival varied between 0.8 years (95% CI 0.8-0.9 (n=1401)) for ALS and 7.6 years (95% CI 6.9-8.6 (n=1061)) for neuromuscular disease. Three-year survival decreased from 76% (95% CI 71-80) between 1996 and 1998 to 52% (95% CI 50-55) between 2017 and 2019. When adjusting for underlying diagnosis and age, the association between start year and decreased survival disappeared, HR 1.00 (95% CI 0.99-1.01). Conclusion Controlling P aCO2 is a key treatment goal for survival in HMV therapy. Survival differed markedly between diagnosis and age groups, and survival rates have declined as the patient group has aged.
Collapse
Affiliation(s)
- Andreas Palm
- Department of Medical Sciences, Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Magnus Ekström
- Department of Clinical Sciences, Respiratory Medicine, Allergology and Palliative Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Össur Emilsson
- Department of Medical Sciences, Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Karin Ersson
- Department of Medical Sciences, Clinical Physiology, Uppsala University, Uppsala, Sweden
- Department of Women's and Children's Health, Physiotherapy, Uppsala University, Uppsala, Sweden
| | - Mirjam Ljunggren
- Department of Medical Sciences, Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Josefin Sundh
- Department of Respiratory Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Ludger Grote
- Centre for Sleep and Wake Disorders, Sahlgrenska Academy, Gothenburg University, Gothenburg, Sweden
- Pulmonary Department, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
8
|
Henzi BC, Lava SAG, Spagnuolo C, Putananickal N, Donner BC, Pfluger M, Burkhardt B, Fischer D. Tamoxifen may contribute to preserve cardiac function in Duchenne muscular dystrophy. Eur J Pediatr 2024; 183:4057-4062. [PMID: 38960907 PMCID: PMC11322393 DOI: 10.1007/s00431-024-05670-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/20/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
Duchenne muscular dystrophy is life-limiting. Cardiomyopathy, which mostly ensues in the second decade of life, is the main cause of death. Treatment options are still limited. The TAMDMD (NCT03354039) trial assessed motor function, muscle strength and structure, laboratory biomarkers, and safety in 79 ambulant boys with genetically confirmed Duchenne muscular dystrophy, 6.5-12 years of age, receiving either daily tamoxifen 20 mg or placebo for 48 weeks. In this post-hoc analysis, available echocardiographic data of ambulant patients recruited at one study centre were retrieved and compared before and after treatment. Data from 14 patients, median 11 (interquartile range, IQR, 11-12) years of age was available. Baseline demographic characteristics were similar in participants assigned to placebo (n = 7) or tamoxifen (n = 7). Left ventricular end-diastolic diameter in the placebo group (median and IQR) was 39 (38-41) mm at baseline and 43 (38-44) mm at study end, while it was 44 (41-46) mm at baseline and 41 (37-46) mm after treatment in the tamoxifen group. Left ventricular fractional shortening in the placebo group was 35% (32-38%) before and 33% (32-36%) after treatment, while in the tamoxifen group it was 34% (33-34%) at baseline and 35% (33-35%) at study end. No safety signals were detected. CONCLUSION This hypothesis-generating post-hoc analysis suggests that tamoxifen over 48 weeks is well tolerated and may help preserving cardiac structure and function in Duchenne muscular dystrophy. Further studies are justified. CLINICALTRIALS gov Identifier: EudraCT 2017-004554-42, NCT03354039 What is known: • Duchenne muscular dystrophy (DMD) is life-limiting. Cardiomyopathy ensues in the second decade of life and is the main cause of death. Treatment options are still limited. • Tamoxifen reduced cardiac fibrosis in mice and improved cardiomyocyte function in human-induced pluripotent stem cell-derived cardiomyocytes. WHAT IS NEW • In this post-hoc analysis of the TAMDMD trial among 14 boys, median 11 years of age, treated with either tamoxifen or placebo for 48 weeks, treatment was well-tolerated. • A visual trend of improved left-ventricular dimensions and better systolic function preservation generates the hypothesis of a potential beneficial effect of tamoxifen in DMD cardiomyopathy.
Collapse
Affiliation(s)
- Bettina C Henzi
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel, University of Basel, Basel, Switzerland
- Division of Neuropediatrics, Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Sebastiano A G Lava
- Pediatric Cardiology Unit, Department of Pediatrics, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 46, Lausanne, 1011, Switzerland.
- Heart Failure and Transplantation, Department of Paediatric Cardiology, Great Ormond Street Hospital, London, UK.
- Clinical Pharmacology and Therapeutics Group, University College London, London, UK.
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Sciences of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland.
| | - Carlos Spagnuolo
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Niveditha Putananickal
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Birgit C Donner
- Pediatric Cardiology, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Marc Pfluger
- Department of Cardiology, Center for Congenital Heart Disease, University Hospital of Bern, Bern, Switzerland
| | - Barbara Burkhardt
- Department of Pediatric Cardiology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Dirk Fischer
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
9
|
Bourke J, Tynan M, Stevenson H, Bremner L, Gonzalez-Fernandez O, McDiarmid AK. Arrhythmias and cardiac MRI associations in patients with established cardiac dystrophinopathy. Open Heart 2024; 11:e002590. [PMID: 38569668 PMCID: PMC10989184 DOI: 10.1136/openhrt-2023-002590] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/21/2024] [Indexed: 04/05/2024] Open
Abstract
AIMS Some patients with cardiac dystrophinopathy die suddenly. Whether such deaths are preventable by specific antiarrhythmic management or simply indicate heart failure overwhelming medical therapies is uncertain. The aim of this prospective, cohort study was to describe the occurrence and nature of cardiac arrhythmias recorded during prolonged continuous ECG rhythm surveillance in patients with established cardiac dystrophinopathy and relate them to abnormalities on cardiac MRI. METHODS AND RESULTS A cohort of 10 patients (36.3 years; 3 female) with LVEF<40% due to Duchenne (3) or Becker muscular (4) dystrophy or Duchenne muscular dystrophy-gene carrying effects in females (3) were recruited, had cardiac MRI, ECG signal-averaging and ECG loop-recorder implants. All were on standard of care heart medications and none had prior history of arrhythmias.No deaths or brady arrhythmias occurred during median follow-up 30 months (range 13-35). Self-limiting episodes of asymptomatic tachyarrhythmia (range 1-29) were confirmed in 8 (80%) patients (ventricular only 2; ventricular and atrial 6). Higher ventricular arrhythmia burden correlated with extent of myocardial fibrosis (extracellular volume%, p=0.029; native T1, p=0.49; late gadolinium enhancement, p=0.49), but not with LVEF% (p=1.0) on MRI and atrial arrhythmias with left atrial dilatation. Features of VT episodes suggested various underlying arrhythmia mechanisms. CONCLUSIONS The overall prevalence of arrhythmias was low. Even in such a small sample size, higher arrhythmia counts occurred in those with larger scar burden and greater ventricular volume, suggesting key roles for myocardial stretch as well as disease progression in arrhythmogenesis. These features overlap with the stage of left ventricular dysfunction when heart failure also becomes overt. The findings of this pilot study should help inform the design of a definitive study of specific antiarrhythmic management in dystrophinopathy. TRIAL REGISTRATION NUMBER ISRCTN15622536.
Collapse
Affiliation(s)
- John Bourke
- Department of Cardiology, NUTH NHS Hospitals Foundation Trust, Newcastle upon Tyne, UK
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle upon Tyne, UK
| | - Margaret Tynan
- Department of Cardiology, NUTH NHS Hospitals Foundation Trust, Newcastle upon Tyne, UK
| | - Hannah Stevenson
- Cardiology Research, NUTH NHS Hospitals Foundation Trust, Newcastle upon Tyne, UK
| | - Leslie Bremner
- Cardiology Research, NUTH NHS Hospitals Foundation Trust, Newcastle upon Tyne, UK
| | | | - Adam K McDiarmid
- Department of Cardiology, NUTH NHS Hospitals Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
10
|
Gharibi S, Vaillend C, Lindsay A. The unconditioned fear response in vertebrates deficient in dystrophin. Prog Neurobiol 2024; 235:102590. [PMID: 38484964 DOI: 10.1016/j.pneurobio.2024.102590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 01/31/2024] [Accepted: 03/05/2024] [Indexed: 03/19/2024]
Abstract
Dystrophin loss due to mutations in the Duchenne muscular dystrophy (DMD) gene is associated with a wide spectrum of neurocognitive comorbidities, including an aberrant unconditioned fear response to stressful/threat stimuli. Dystrophin-deficient animal models of DMD demonstrate enhanced stress reactivity that manifests as sustained periods of immobility. When the threat is repetitive or severe in nature, dystrophinopathy phenotypes can be exacerbated and even cause sudden death. Thus, it is apparent that enhanced sensitivity to stressful/threat stimuli in dystrophin-deficient vertebrates is a legitimate cause of concern for patients with DMD that could impact neurocognition and pathophysiology. This review discusses our current understanding of the mechanisms and consequences of the hypersensitive fear response in preclinical models of DMD and the potential challenges facing clinical translatability.
Collapse
Affiliation(s)
- Saba Gharibi
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Cyrille Vaillend
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, Saclay 91400, France.
| | - Angus Lindsay
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia; School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand; Department of Medicine, University of Otago, Christchurch 8014, New Zealand.
| |
Collapse
|
11
|
Dabaj I, Ducatez F, Marret S, Bekri S, Tebani A. Neuromuscular disorders in the omics era. Clin Chim Acta 2024; 553:117691. [PMID: 38081447 DOI: 10.1016/j.cca.2023.117691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023]
Abstract
Neuromuscular disorders encompass a spectrum of conditions characterized by primary lesions within the peripheral nervous system, which include the anterior horn cell, peripheral nerve, neuromuscular junction, and muscle. In pediatrics, most of these disorders are linked to genetic causes. Despite the considerable progress, the diagnosis of these disorders remains a challenging due to wide clinical presentation, disease heterogeneity and rarity. It is noteworthy that certain neuromuscular disorders, once deemed untreatable, can now be effectively managed through novel therapies. Biomarkers emerge as indispensable tools, serving as objective measures that not only refine diagnostic accuracy but also provide guidance for therapeutic decision-making and the ongoing monitoring of long-term outcomes. Herein a comprehensive review of biomarkers in neuromuscular disorders is provided. We highlight the role of omics-based technologies that further characterize neuromuscular pathophysiology as well as identify potential therapeutic targets to guide treatment strategies.
Collapse
Affiliation(s)
- Ivana Dabaj
- Normandie Univ, UNIROUEN, INSERM U1245, Nord/Est/Ile de France Neuromuscular Reference Center CHU Rouen, Department of Neonatalogy, Pediatric Intensive Care, and Neuropediatrics, F-76000 Rouen, France.
| | - Franklin Ducatez
- Normandie Univ, UNIROUEN, INSERM U1245, Nord/Est/Ile de France Neuromuscular Reference Center CHU Rouen, Department of Neonatalogy, Pediatric Intensive Care, and Neuropediatrics, F-76000 Rouen, France
| | - Stéphane Marret
- Normandie Univ, UNIROUEN, INSERM U1245, Nord/Est/Ile de France Neuromuscular Reference Center CHU Rouen, Department of Neonatalogy, Pediatric Intensive Care, and Neuropediatrics, F-76000 Rouen, France
| | - Soumeya Bekri
- Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, Department of Metabolic Biochemistry, F-76000 Rouen, France
| | - Abdellah Tebani
- Normandie Univ, UNIROUEN, INSERM U1245, CHU Rouen, Department of Metabolic Biochemistry, F-76000 Rouen, France
| |
Collapse
|
12
|
Koehler S, Kuhm J, Huffaker T, Young D, Tandon A, André F, Frey N, Greil G, Hussain T, Engelhardt S. Artificial Intelligence to derive aligned strain in cine CMR to detect patients with myocardial fibrosis: an open and scrutinizable approach. RESEARCH SQUARE 2024:rs.3.rs-3785677. [PMID: 38260274 PMCID: PMC10802696 DOI: 10.21203/rs.3.rs-3785677/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Cine Cardiac Magnetic Resonance (CMR) is the gold standard for cardiac function evaluation, incorporating ejection fraction (EF) and strain as vital indicators of abnormal deformation. Rare pathologies like Duchenne muscular dystrophies (DMD) are monitored with repeated late gadolinium-enhanced (LGE) CMR for identification of myocardial fibrosis. However, it is judicious to reduce repeated gadolinium exposure and rather employ strain analysis from cine CMR. This solution is limited so far since full strain curves are not comparable between individual cardiac cycles and current practice mainly neglects diastolic deformation patterns. Our novel Deep Learning-based approach derives strain values aligned by key frames throughout the cardiac cycle. In a reproducibility scenario (57+82 patients), our results reveal five times more significant differences (22 vs. 4) between patients with scar and without, enhancing scar detection by +30%, improving detection of patients with preserved EF by +61%, with an overall sensitivity/specificity of 82/81%.
Collapse
Affiliation(s)
- Sven Koehler
- Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partnersites Heidelberg and Mannheim, Germany
- University Heidelberg, Heidelberg, Germany
| | - Julian Kuhm
- Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partnersites Heidelberg and Mannheim, Germany
| | - Tyler Huffaker
- Division of Pediatric Cardiology, Department of Pediatrics, UT Southwestern /Children’s Health, 1935 Medical District Drive B3.09, Dallas, TX 75235, USA
| | - Daniel Young
- Division of Pediatric Cardiology, Department of Pediatrics, UT Southwestern /Children’s Health, 1935 Medical District Drive B3.09, Dallas, TX 75235, USA
| | - Animesh Tandon
- Department of Heart, Vascular, and Thoracic, Children’s Institute; Cleveland Clinic Children’s Center for Artificial Intelligence (C4AI); and Cardiovascular Innovation Research Center, Cleveland Clinic Children’s, Cleveland, Ohio, USA
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Florian André
- Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partnersites Heidelberg and Mannheim, Germany
- University Heidelberg, Heidelberg, Germany
| | - Norbert Frey
- Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partnersites Heidelberg and Mannheim, Germany
- University Heidelberg, Heidelberg, Germany
| | - Gerald Greil
- Division of Pediatric Cardiology, Department of Pediatrics, UT Southwestern /Children’s Health, 1935 Medical District Drive B3.09, Dallas, TX 75235, USA
| | - Tarique Hussain
- Division of Pediatric Cardiology, Department of Pediatrics, UT Southwestern /Children’s Health, 1935 Medical District Drive B3.09, Dallas, TX 75235, USA
| | - Sandy Engelhardt
- Department of Internal Medicine III, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partnersites Heidelberg and Mannheim, Germany
- University Heidelberg, Heidelberg, Germany
| |
Collapse
|
13
|
Conway KM, Thomas S, Ciafaloni E, Khan RS, Mann JR, Romitti PA, Mathews KD. Prophylactic use of cardiac medications for delay of left ventricular dysfunction in Duchenne muscular dystrophy. Birth Defects Res 2024; 116:e2260. [PMID: 37850663 DOI: 10.1002/bdr2.2260] [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: 06/27/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Epidemiological support for prophylactic treatment of left ventricular dysfunction (LVD) in Duchenne muscular dystrophy is limited. We used retrospective, population-based surveillance data from the Muscular Dystrophy Surveillance, Tracking and Research Network to evaluate whether prophylaxis delays LVD onset. METHODS We analyzed 455 males born during 1982-2009. Age at first abnormal echocardiogram (ejection fraction <55% or shortening fraction <28%) determined LVD onset. Prophylaxis was defined as cardiac medication use at least 1 year prior to LVD. Corticosteroid use was also coded. Kaplan-Meier curve estimation and Cox Proportional Hazard modeling with time-varying covariates describe associations. RESULTS LVD was identified among 40.7%; average onset age was 14.2 years. Prophylaxis was identified for 20.2% and corticosteroids for 57.4%. Prophylaxis showed delayed LVD onset (p < .001) and lower hazard of dysfunction (adjusted hazard ratio [aHR] = 0.39, 95%CL = 0.22, 0.65) compared to untreated. Compared to no treatment, continuous corticosteroids only (aHR = 1.01, 95%CL = 0.66, 1.53) and prophylaxis only (aHR = 0.67, 95%CL = 0.25, 1.50) were not cardioprotective, but prophylaxis plus continuous corticosteroids were associated with lower hazard of dysfunction (aHR = 0.37, 95%CL = 0.15, 0.80). CONCLUSIONS Proactive cardiac treatment and monitoring are critical aspects of managing Duchenne muscular dystrophy. Consistent with clinical care guidelines, this study supports clinical benefit from cardiac medications initiated prior to documented LVD and suggests further benefit when combined with corticosteroids.
Collapse
Affiliation(s)
- Kristin M Conway
- Department of Epidemiology, College of Public Health, The University of Iowa, Iowa City, Iowa, USA
| | - Shiny Thomas
- New York State Department of Health, Albany, New York, USA
| | - Emma Ciafaloni
- Department of Neurology, School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, New York, USA
| | - Rabia S Khan
- Department of Pediatrics, UCLA Health Sciences, Los Angeles, California, USA
- Department of Pediatrics, Roy J and Lucille A Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Joshua R Mann
- Department of Preventive Medicine, School of Medicine and John D. Bower School of Population Health, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Paul A Romitti
- Department of Epidemiology, College of Public Health, The University of Iowa, Iowa City, Iowa, USA
| | - Katherine D Mathews
- Department of Pediatrics, Roy J and Lucille A Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
14
|
McDonald C, Camino E, Escandon R, Finkel RS, Fischer R, Flanigan K, Furlong P, Juhasz R, Martin AS, Villa C, Sweeney HL. Draft Guidance for Industry Duchenne Muscular Dystrophy, Becker Muscular Dystrophy, and Related Dystrophinopathies - Developing Potential Treatments for the Entire Spectrum of Disease. J Neuromuscul Dis 2024; 11:499-523. [PMID: 38363616 DOI: 10.3233/jnd-230219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Background Duchenne muscular dystrophy (DMD) and related dystrophinopathies are neuromuscular conditions with great unmet medical needs that require the development of effective medical treatments. Objective To aid sponsors in clinical development of drugs and therapeutic biological products for treating DMD across the disease spectrum by integrating advancements, patient registries, natural history studies, and more into a comprehensive guidance. Methods This guidance emerged from collaboration between the FDA, the Duchenne community, and industry stakeholders. It entailed a structured approach, involving multiple committees and boards. From its inception in 2014, the guidance underwent revisions incorporating insights from gene therapy studies, cardiac function research, and innovative clinical trial designs. Results The guidance provides a deeper understanding of DMD and its variants, focusing on patient engagement, diagnostic criteria, natural history, biomarkers, and clinical trials. It underscores patient-focused drug development, the significance of dystrophin as a biomarker, and the pivotal role of magnetic resonance imaging in assessing disease progression. Additionally, the guidance addresses cardiomyopathy's prominence in DMD and the burgeoning field of gene therapy. Conclusions The updated guidance offers a comprehensive understanding of DMD, emphasizing patient-centric approaches, innovative trial designs, and the importance of biomarkers. The focus on cardiomyopathy and gene therapy signifies the evolving realm of DMD research. It acts as a crucial roadmap for sponsors, potentially leading to improved treatments for DMD.
Collapse
Affiliation(s)
| | - Eric Camino
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Rafael Escandon
- DGBI Consulting, LLC, Bainbridge Island, Washington, DC, USA
| | | | - Ryan Fischer
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Kevin Flanigan
- Center for Experimental Neurotherapeutics, Department of Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Pat Furlong
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Rose Juhasz
- Nationwide Children's Hospital, Columbus, OH, USA
| | - Ann S Martin
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Chet Villa
- Trinity Health Michigan, Grand Rapids, MI, USA
| | - H Lee Sweeney
- Cincinnati Children's Hospital Medical Center within the UC Department of Pediatrics, Cincinnati, OH, USA
| |
Collapse
|
15
|
Zhou ZQ, Xu HY, Fu H, Xu K, Xu R, Cai XT, Guo YK. Derivation and validation of diagnostic models for myocardial fibrosis in duchenne muscular dystrophy: assessed by multi-parameter cardiovascular magnetic resonance. Orphanet J Rare Dis 2023; 18:388. [PMID: 38082428 PMCID: PMC10714650 DOI: 10.1186/s13023-023-02931-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 09/25/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Gadolinium-enhanced cardiovascular magnetic resonance (CMR) is the most widely used approach for diagnosing myocardial fibrosis with late gadolinium enhancement (LGE) in cardiomyopathy associated with Duchenne muscular dystrophy. Given the limitations and safety of gadolinium use, we wanted to develop and evaluate multi-parametric pre-contrast CMR models for the diagnosis of LGE and investigate whether they could be utilised as surrogates for LGE in DMD patients. METHODS A total of 136 DMD patients were prospectively recruited and separated into LGE - and LGE + groups. In the first subset of patients (derivation cohort), regression models for the diagnosis of LGE were built by logistic regression using pre-contrast sequence parameters. In a validation cohort of other patients, the models' performances were evaluated. RESULTS EF, native T1 and longitudinal strain alone, as well as their combinations form seven models. The model that included EF, native T1 and longitudinal strain had the best diagnostic value, but there was no significant difference in diagnostic accuracy among the other models except EF. In the validation cohort, the diagnosis outcomes of models were moderate consistent with the existence of LGE. The longitudinal strain outperformed the other models in terms of diagnostic value (sensitivity: 83.33%, specificity: 54.55%). CONCLUSIONS Pre-contrast sequences have a moderate predictive value for LGE. Thus, pre-contrast parameters may be considered only in a specific subset of DMD patients who cannot cooperate for long-time examinations and have contradiction of contrast agent to help predict the presence of LGE. TRIAL REGISTRATION NUMBER (TRN) ChiCTR1800018340 DATE OF REGISTRATION: 20180107.
Collapse
Grants
- 82120108015, 81971586, 81771897, 82102020, 82071874, 81901712, 82271981 and 81771887 National Natural Science Foundation of China
- 2020YFS0050, 2020YJ0029, 2017TD0005, 21ZDYF1967 and 2021YFS0175 Sichuan Science and Technology Program
- SCU2020D4132 Fundamental Research Funds for the Central Universities
- No. HFCSC2019B01 Clinical Research Finding of Chinese Society of Cardiovascular Disease (CSC) of 2019
- ZYGD18019 1•3•5 project for disciplines of excellence, West China Hospital, Sichuan University
Collapse
Affiliation(s)
- Zi-Qi Zhou
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, China
| | - Hua-Yan Xu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, China
| | - Hang Fu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, China
| | - Ke Xu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, China
| | - Rong Xu
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, China
| | - Xiao-Tang Cai
- Department of Rehabilitation, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Department of Rehabilitation, Chengdu, China.
| | - Ying-Kun Guo
- Department of Radiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital, Sichuan University, 20# Section 3 South Renmin Road, Chengdu, 610041, China.
| |
Collapse
|
16
|
Yang TL, Ting J, Lin MR, Chang WC, Shih CM. Identification of Genetic Variants Associated with Severe Myocardial Bridging through Whole-Exome Sequencing. J Pers Med 2023; 13:1509. [PMID: 37888120 PMCID: PMC10608235 DOI: 10.3390/jpm13101509] [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: 09/06/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023] Open
Abstract
Myocardial bridging (MB) is a congenital coronary artery anomaly and an important cause of angina. The genetic basis of MB is currently unknown. This study used a whole-exome sequencing technique and analyzed genotypic differences. Eight coronary angiography-confirmed cases of severe MB and eight age- and sex-matched control patients were investigated. In total, 139 rare variants that are potentially pathogenic for severe MB were identified in 132 genes. Genes with multiple rare variants or co-predicted by ClinVar and CADD/REVEL for severe MB were collected, from which heart-specific genes were selected under the guidance of tissue expression levels. Functional annotation indicated significant genetic associations with abnormal skeletal muscle mass, cardiomyopathies, and transmembrane ion channels. Candidate genes were reviewed regarding the functions and locations of each individual gene product. Among the gene candidates for severe MB, rare variants in DMD, SGCA, and TTN were determined to be the most crucial. The results suggest that altered anchoring proteins on the cell membrane and intracellular sarcomere unit of cardiomyocytes play a role in the development of the missed trajectory of coronary vessels. Additional studies are required to support the diagnostic application of cardiac sarcoglycan and dystroglycan complexes in patients with severe MB.
Collapse
Affiliation(s)
- Tsung-Lin Yang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
| | - Jafit Ting
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan; (J.T.); (M.-R.L.)
| | - Min-Rou Lin
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan; (J.T.); (M.-R.L.)
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan; (J.T.); (M.-R.L.)
- Master’ Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
- Integrative Research Center for Critical Care, Wan Fang Hospital, Taipei Medical University, Taipei 11696, Taiwan
| | - Chun-Ming Shih
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
17
|
Soslow JH, Xu M, Slaughter JC, Crum K, Kaslow JA, George-Durrett K, Raucci FJ, Wilkinson JD, Cripe L, Hor K, Spurney CF, Markham LW. Cardiovascular Measures of All-Cause Mortality in Duchenne Muscular Dystrophy. Circ Heart Fail 2023; 16:e010040. [PMID: 37288563 PMCID: PMC10524475 DOI: 10.1161/circheartfailure.122.010040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 03/30/2023] [Indexed: 06/09/2023]
Abstract
BACKGROUND Cardiopulmonary failure is the leading cause of death in Duchenne muscular dystrophy (DMD). Research into DMD-specific cardiovascular therapies is ongoing, but there are no Food and Drug Administration-approved cardiac end points. To adequately power a therapeutic trial, appropriate end points must be chosen and the rate of change for these end points reported. The objective of this study was to evaluate rate of change for cardiac magnetic resonance and blood biomarkers and to determine which measures associate with all-cause mortality in DMD. METHODS Seventy-eight DMD subjects underwent 211 cardiac magnetic resonance studies analyzed for left ventricular (LV) ejection fraction, indexed LV end diastolic and systolic volumes, circumferential strain, late gadolinium enhancement presence and severity (global severity score, and full width half maximum), native T1 mapping, T2 mapping, and extracellular volume. Blood samples were analyzed for BNP (brain natriuretic peptide), NT-proBNP (N-terminal pro-B-type natriuretic peptide), and troponin I. Cox proportional hazard regression modeling was performed with all-cause mortality as the outcome. RESULTS Fifteen subjects (19%) died. LV ejection fraction, indexed end systolic volumes, global severity score, and full width half maximum worsened at 1 and 2 years while circumferential strain and indexed LV end diastolic volumes worsened at 2 years. LV ejection fraction, indexed LV end diastolic and systolic volumes, late gadolinium enhancement full width half maximum, and circumferential strain associated with all-cause mortality (P<0.05). NT-proBNP was the only blood biomarker that associated with all-cause mortality (P<0.05). CONCLUSIONS LV ejection fraction, indexed LV volumes, circumferential strain, late gadolinium enhancement full width half maximum, and NT-proBNP are associated with all-cause mortality in DMD and may be the best end points for use in cardiovascular therapeutic trials. We also report change over time of cardiac magnetic resonance and blood biomarkers.
Collapse
Affiliation(s)
- Jonathan H Soslow
- Division or Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Meng Xu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - James C Slaughter
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kimberly Crum
- Division or Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jacob A Kaslow
- Division of Pediatric Pulmonology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kristen George-Durrett
- Division or Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Frank J Raucci
- Division of Pediatric Cardiology, Department of Pediatrics, Children’s Hospital of Richmond at Virginia Commonwealth University Health System, Richmond, VA 23298, USA
| | - James D Wilkinson
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Linda Cripe
- Division of Pediatric Cardiology, Nationwide Children’s Hospital and The Ohio State University, Columbus, OH 43205 USA
| | - Kan Hor
- Division of Pediatric Cardiology, Nationwide Children’s Hospital and The Ohio State University, Columbus, OH 43205 USA
| | - Christopher F Spurney
- Division of Cardiology, Children’s National Heart Institute, Children’s National Hospital, Washington, D.C. 20010, USA
| | - Larry W Markham
- Division of Cardiology, Department of Pediatrics, Riley Hospital for Children at Indiana University Health, Indianapolis, IN, USA
| |
Collapse
|
18
|
Baskan S, Karaca Ozer P, Orta H, Ozbingol D, Yavuz ML, Ayduk Govdeli E, Nisli K, Oztarhan K. Prognostic Value of Tpeak-Tend Interval in Early Diagnosis of Duchenne Muscular Dystrophy Cardiomyopathy. Diagnostics (Basel) 2023; 13:2381. [PMID: 37510124 PMCID: PMC10377932 DOI: 10.3390/diagnostics13142381] [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: 06/04/2023] [Revised: 07/02/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
The most common cause of death in patients with Duchenne muscular dystrophy (DMD) is cardiomyopathy. Our aim was to investigate the relationship between the Tpeak-Tend (Tp-e) interval and the premature ventricular contraction (PVC) burden and therefore early arrhythmic risk and cardiac involvement in DMD patients. Twenty-five patients with DMD followed by pediatric cardiology were included in the study. Those with a frequency of <1% PVC in the 24 h Holter were assigned to Group 1 (n = 15), and those with >1% were assigned to Group 2 (n = 10). Comparisons were made with healthy controls (n = 27). Left ventricular ejection fraction (LVEF) was lowest in Group 2 and highest in the control group (p < 0.001). LV end-diastolic diameter was greater in Group 2 than in Group 1 and the control group (p = 0.005). Pro-BNP and troponin levels were higher in Group 1 and Group 2 than in the control group (p = 0.001 and p < 0.001, respectively). Tp-e interval was longer in Group 2 compared to Group 1 and the control group (p < 0.001). The LVEF (OR 0.879, 95% CI 0.812-0.953; p = 0.002) and Tp-e interval (OR 1.181, 95% CI 1.047-1.332; p = 0.007) were independent predictors of PVC/24 h frequency of >1%. A Tp-e interval > 71.65 ms predicts PVC > 1%, with a sensitivity of 80% and a specificity of 90% (AUC = 0.842, 95% CI (0.663-1.000), p = 0.001). Determination of Tp-e prolongation from ECG data may help in the determination of cardiac involvement and early diagnosis of arrhythmic risk in DMD.
Collapse
Affiliation(s)
- Serra Baskan
- Department of Pediatric Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34134, Turkey
| | - Pelin Karaca Ozer
- Department of Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34134, Turkey
| | - Huseyin Orta
- Department of Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34134, Turkey
| | - Doruk Ozbingol
- Department of Pediatric Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34134, Turkey
| | - Mustafa Lutfi Yavuz
- Department of Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34134, Turkey
| | - Elif Ayduk Govdeli
- Department of Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34134, Turkey
| | - Kemal Nisli
- Department of Pediatric Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34134, Turkey
| | - Kazim Oztarhan
- Department of Pediatric Cardiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul 34134, Turkey
| |
Collapse
|
19
|
George TG, Hanft LM, Krenz M, Domeier TL, McDonald KS. Dystrophic cardiomyopathy: role of the cardiac myofilaments. Front Physiol 2023; 14:1207658. [PMID: 37362434 PMCID: PMC10288979 DOI: 10.3389/fphys.2023.1207658] [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: 04/17/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023] Open
Abstract
Dystrophic cardiomyopathy arises from mutations in the dystrophin gene. Dystrophin forms part of the dystrophin glycoprotein complex and is postulated to act as a membrane stabilizer, protecting the sarcolemma from contraction-induced damage. Duchenne muscular dystrophy (DMD) is the most severe dystrophinopathy, caused by a total absence of dystrophin. Patients with DMD present with progressive skeletal muscle weakness and, because of treatment advances, a cardiac component of the disease (i.e., dystrophic cardiomyopathy) has been unmasked later in disease progression. The role that myofilaments play in dystrophic cardiomyopathy is largely unknown and, as such, this study aimed to address cardiac myofilament function in a mouse model of muscular dystrophy. To assess the effects of DMD on myofilament function, isolated permeabilized cardiomyocytes of wild-type (WT) littermates and Dmdmdx-4cv mice were attached between a force transducer and motor and subjected to contractile assays. Maximal tension and rates of force development (indexed by the rate constant, k tr) were similar between WT and Dmdmdx-4cv cardiac myocyte preparations. Interestingly, Dmdmdx-4cv cardiac myocytes exhibited greater sarcomere length dependence of peak power output compared to WT myocyte preparations. These results suggest dystrophin mitigates length dependence of activation and, in the absence of dystrophin, augmented sarcomere length dependence of myocyte contractility may accelerate ventricular myocyte contraction-induced damage and contribute to dystrophic cardiomyopathy. Next, we assessed if mavacamten, a small molecule modulator of thick filament activation, would mitigate contractile properties observed in Dmdmdx-4cv permeabilized cardiac myocyte preparations. Mavacamten decreased maximal tension and k tr in both WT and Dmdmdx-4cv cardiac myocytes, while also normalizing the length dependence of peak power between WT and Dmdmdx-4cv cardiac myocyte preparations. These results highlight potential benefits of mavacamten (i.e., reduced contractility while maintaining exquisite sarcomere length dependence of power output) as a treatment for dystrophic cardiomyopathy associated with DMD.
Collapse
|
20
|
Kaufman BD, Garcia A, He Z, Tesi-Rocha C, Buu M, Rosenthal D, Gordish-Dressman H, Almond CS, Duong T. Major Adverse Dystrophinopathy Events (MADE) Score as Marker of Cumulative Morbidity and Risk for Mortality in Boys with Duchenne Muscular Dystrophy. PROGRESS IN PEDIATRIC CARDIOLOGY 2023; 69:101639. [PMID: 37990740 PMCID: PMC10659574 DOI: 10.1016/j.ppedcard.2023.101639] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Background Overlapping symptoms from cardiomyopathy, respiratory insufficiency, and skeletal myopathy confound assessment of heart failure in Duchenne Muscular Dystrophy. We developed an ordinal scale of multiorgan clinical variables that reflect cumulative disease burden-the Major Adverse Dystrophinopathy Event (MADE) Score. We hypothesized that a higher MADE score would be associated with increased mortality in boys with Duchenne Muscular Dystrophy. The Cooperative International Neuromuscular Research Group Duchenne Natural History Study dataset was utilized for validation. Methods Duchenne Natural History Study variables were selected based on clinical relevance to prespecified domains: Cardiac, Pulmonary, Myopathy, Nutrition. Severity points (0-4) were assigned and summed for study visits. MADE score for cohorts defined by age, ambulatory status, and survival were compared at enrollment and longitudinally.Associations between MADE score and mortality were examined. Results Duchenne Natural History Study enrolled 440 males, 12.6 ±6.1 years old, with 3,559 visits over 4.6 ±2.8 years, 45 deaths. MADE score increased with age and nonambulatory status. Mean MADE score per visit was 19 ±10 for those who died vs. 9.8 ±9.3 in survivors p=0.03. Baseline MADE score >12 predicted mortality independent of age (78% sensitivity, CPE.70). Rising MADE score trajectory was associated with mortality in models adjusted for enrollment age, follow-up time, and ambulatory status, all p<.001. Conclusion A multiorgan severity score, MADE, was developed to track cumulative morbidities that impact heart failure in Duchenne muscular dystrophy. MADE score predicted Duchenne Natural History Study mortality. MADE score can be used for serial heart failure assessment in males and may serve as an endpoint for Duchenne muscular dystrophy clinical research.
Collapse
Affiliation(s)
- Beth D Kaufman
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Palo Alto, CA
| | - Ariadna Garcia
- Quantitative Sciences Unit, Stanford University, Palo Alto, CA
| | - Zihuai He
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA
| | - Carolina Tesi-Rocha
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA
| | - MyMy Buu
- Department of Pediatrics (Pulmonary Medicine), Stanford University School of Medicine, Palo Alto, CA
| | - David Rosenthal
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Palo Alto, CA
| | | | - Christopher S Almond
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Palo Alto, CA
| | - Tina Duong
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Palo Alto, CA
| |
Collapse
|
21
|
Lupu M, Ioghen M, Perjoc RȘ, Scarlat AM, Vladâcenco OA, Roza E, Epure DAM, Teleanu RI, Severin EM. The Importance of Implementing a Transition Strategy for Patients with Muscular Dystrophy: From Child to Adult-Insights from a Tertiary Centre for Rare Neurological Diseases. CHILDREN (BASEL, SWITZERLAND) 2023; 10:959. [PMID: 37371191 DOI: 10.3390/children10060959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023]
Abstract
Progress in the field of muscular dystrophy (MD) using a multidisciplinary approach based on international standards of care has led to a significant increase in the life expectancy of patients. The challenge of transitioning from pediatric to adult healthcare has been acknowledged for over a decade, yet it continues to be a last-minute concern. Currently, there is no established consensus on how to evaluate the effectiveness of the transition process. Our study aimed to identify how well patients are prepared for the transition and to determine their needs. We conducted a descriptive, cross-sectional study on 15 patients aged 14 to 21 years. The patients completed a sociodemographic and a Transition Readiness Assessment Questionnaire (TRAQ). We also analyzed the comorbidities of these patients. Our study revealed that only 46.7% of the patients had engaged in a conversation with a medical professional, namely, a child neurologist, about transitioning. A total of 60% of the participants expressed having confidence in their self-care ability. However, the median TRAQ score of 3.6 shows that these patients overestimate themselves. We emphasize the necessity for a slow, personalized transition led by a multidisciplinary team to ensure the continuity of state-of-the-art care from pediatric to adult healthcare services and the achievement of the highest possible quality of life for these patients.
Collapse
Affiliation(s)
- Maria Lupu
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Mihaela Ioghen
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Radu-Ștefan Perjoc
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Andra-Maria Scarlat
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Oana Aurelia Vladâcenco
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Paediatric Neurology, Dr Victor Gomoiu Children's Hospital, 022102 Bucharest, Romania
| | - Eugenia Roza
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Paediatric Neurology, Dr Victor Gomoiu Children's Hospital, 022102 Bucharest, Romania
| | - Diana Ana-Maria Epure
- Department of Paediatric Neurology, Dr Victor Gomoiu Children's Hospital, 022102 Bucharest, Romania
| | - Raluca Ioana Teleanu
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Paediatric Neurology, Dr Victor Gomoiu Children's Hospital, 022102 Bucharest, Romania
| | - Emilia Maria Severin
- Clinical Neurosciences Department, Peadiatric Neurology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
22
|
Sarkozy A, Quinlivan R, Bourke JP, Ferlini A. 263rd ENMC International Workshop: Focus on female carriers of dystrophinopathy: refining recommendations for prevention, diagnosis, surveillance, and treatment. Hoofddorp, The Netherlands, 13-15 May 2022. Neuromuscul Disord 2023; 33:274-284. [PMID: 36804616 DOI: 10.1016/j.nmd.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 01/08/2023] [Indexed: 01/13/2023]
Affiliation(s)
- Anna Sarkozy
- Dubowitz Neuromuscular Centre, Great Ormond Street Hospital, Institute of Child Health, London, UK.
| | - Rosaline Quinlivan
- Queen Square Centre for Neuromuscular Diseases, UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK.
| | - John P Bourke
- Department of Cardiology, Freeman Hospital, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, UK and John Walton Muscular Dystrophy Research Centre, Newcastle University.
| | - Alessandra Ferlini
- Medical Genetics Unit, Department of Medical Science, University of Ferrara, Italy.
| |
Collapse
|
23
|
Nandi D, Auerbach SR, Bansal N, Buchholz H, Conway J, Esteso P, Kaufman BD, Lal AK, Law SP, Lorts A, May LJ, Mehegan M, Mokshagundam D, Morales DLS, O'Connor MJ, Rosenthal DN, Shezad MF, Simpson KE, Sutcliffe DL, Vanderpluym C, Wittlieb-Weber CA, Zafar F, Cripe L, Villa CR. Initial multicenter experience with ventricular assist devices in children and young adults with muscular dystrophy: An ACTION registry analysis. J Heart Lung Transplant 2023; 42:246-254. [PMID: 36270923 DOI: 10.1016/j.healun.2022.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 08/19/2022] [Accepted: 09/06/2022] [Indexed: 01/18/2023] Open
Abstract
PURPOSE Cardiac disease results in significant morbidity and mortality in patients with muscular dystrophy (MD). Single centers have reported their ventricular assist device (VAD) experience in specific MDs and in limited numbers. This study sought to describe the outcomes associated with VAD therapy in an unselected population across multiple centers. METHODS We examined outcomes of patients with MD and dilated cardiomyopathy implanted with a VAD at Advanced Cardiac Therapies Improving Outcomes Network (ACTION) centers from 9/2012 to 9/2020. RESULTS A total of 19 VADs were implanted in 18 patients across 12 sites. The majority of patients had dystrophinopathy (66%) and the median age at implant was 17.2 years (range 11.7-29.5). Eleven patients were non-ambulatory (61%) and 6 (33%) were on respiratory support pre-VAD. Five (28%) patients were implanted as a bridge to transplant, 4 of whom survived to transplant. Of 13 patients implanted as bridge to decision or destination therapy, 77% were alive at 1 year and 69% at 2 years. The overall frequencies of positive outcome (transplanted or alive on device) at 1 year and 2 years were 84% and 78%, respectively. Two patients suffered a stroke, 2 developed sepsis, 1 required tracheostomy, and 1 experienced severe right heart failure requiring right-sided VAD. CONCLUSIONS This study demonstrates the potential utility of VAD therapies in patients with muscular dystrophy. Further research is needed to further improve outcomes and better determine which patients may benefit most from VAD therapy in terms of survival and quality of life.
Collapse
Affiliation(s)
| | - Scott R Auerbach
- University of Colorado Denver, Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado
| | - Neha Bansal
- Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York
| | | | | | - Paul Esteso
- Boston Children's Hospital, Boston, Massachusetts
| | - Beth D Kaufman
- Lucile Packard Children's Hospital, Palo Alto, California
| | - Ashwin K Lal
- Primary Children's Hospital, Salt Lake City, Utah
| | - Sabrina P Law
- Morgan Stanley Children's Hospital of New York-Presbyterian, Columbia University Irving Medical Center, New York, New York
| | - Angela Lorts
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | | | - Mary Mehegan
- St. Louis Children's Hospital, St Louis, Missouri
| | | | | | | | | | | | - Kathleen E Simpson
- University of Colorado Denver, Anschutz Medical Campus and Children's Hospital Colorado, Aurora, Colorado
| | | | | | | | - Farhan Zafar
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Linda Cripe
- Nationwide Children's Hospital, Columbus, Ohio
| | - Chet R Villa
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
24
|
Birch SM, Lawlor MW, Conlon TJ, Guo LJ, Crudele JM, Hawkins EC, Nghiem PP, Ahn M, Meng H, Beatka MJ, Fickau BA, Prieto JC, Styner MA, Struharik MJ, Shanks C, Brown KJ, Golebiowski D, Bettis AK, Balog-Alvarez CJ, Clement N, Coleman KE, Corti M, Pan X, Hauschka SD, Gonzalez JP, Morris CA, Schneider JS, Duan D, Chamberlain JS, Byrne BJ, Kornegay JN. Assessment of systemic AAV-microdystrophin gene therapy in the GRMD model of Duchenne muscular dystrophy. Sci Transl Med 2023; 15:eabo1815. [PMID: 36599002 PMCID: PMC11107748 DOI: 10.1126/scitranslmed.abo1815] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 12/09/2022] [Indexed: 01/06/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle wasting disease caused by the absence of dystrophin, a membrane-stabilizing protein encoded by the DMD gene. Although mouse models of DMD provide insight into the potential of a corrective therapy, data from genetically homologous large animals, such as the dystrophin-deficient golden retriever muscular dystrophy (GRMD) model, may more readily translate to humans. To evaluate the clinical translatability of an adeno-associated virus serotype 9 vector (AAV9)-microdystrophin (μDys5) construct, we performed a blinded, placebo-controlled study in which 12 GRMD dogs were divided among four dose groups [control, 1 × 1013 vector genomes per kilogram (vg/kg), 1 × 1014 vg/kg, and 2 × 1014 vg/kg; n = 3 each], treated intravenously at 3 months of age with a canine codon-optimized microdystrophin construct, rAAV9-CK8e-c-μDys5, and followed for 90 days after dosing. All dogs received prednisone (1 milligram/kilogram) for a total of 5 weeks from day -7 through day 28. We observed dose-dependent increases in tissue vector genome copy numbers; μDys5 protein in multiple appendicular muscles, the diaphragm, and heart; limb and respiratory muscle functional improvement; and reduction of histopathologic lesions. As expected, given that a truncated dystrophin protein was generated, phenotypic test results and histopathologic lesions did not fully normalize. All administrations were well tolerated, and adverse events were not seen. These data suggest that systemically administered AAV-microdystrophin may be dosed safely and could provide therapeutic benefit for patients with DMD.
Collapse
Affiliation(s)
- Sharla M. Birch
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, TX; 77843
| | | | - Thomas J. Conlon
- University of Florida, Powell Gene Therapy Center, Gainesville, FL; 32610
| | - Lee-Jae Guo
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, TX; 77843
| | | | - Eleanor C. Hawkins
- North Carolina State University, College of Veterinary Medicine, Raleigh, NC; 27606
| | - Peter P. Nghiem
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, TX; 77843
| | - Mihye Ahn
- University of Nevada-Reno, Reno, NV; 89557
| | - Hui Meng
- Medical College of Wisconsin, Milwaukee, WI; 53226
| | | | | | | | | | | | | | | | | | - Amanda K. Bettis
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, TX; 77843
| | - Cynthia J. Balog-Alvarez
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, TX; 77843
| | - Nathalie Clement
- University of Florida, Powell Gene Therapy Center, Gainesville, FL; 32610
| | - Kirsten E. Coleman
- University of Florida, Powell Gene Therapy Center, Gainesville, FL; 32610
| | - Manuela Corti
- University of Florida, Powell Gene Therapy Center, Gainesville, FL; 32610
| | - Xiufang Pan
- University of Missouri, School of Medicine, Columbia, MO 65212
| | | | | | | | | | - Dongsheng Duan
- University of Missouri, School of Medicine, Columbia, MO 65212
| | | | - Barry J. Byrne
- University of Florida, Powell Gene Therapy Center, Gainesville, FL; 32610
| | - Joe. N. Kornegay
- Texas A&M University, College of Veterinary Medicine and Biomedical Sciences, College Station, TX; 77843
| |
Collapse
|
25
|
Clinical outcomes of continuous flow left ventricular assist device therapy as bridge to transplant strategy in muscular dystrophy: a single-center study. Gen Thorac Cardiovasc Surg 2022; 71:347-353. [PMID: 36348232 DOI: 10.1007/s11748-022-01889-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/27/2022] [Indexed: 11/10/2022]
Abstract
OBJECTIVE In muscular dystrophies (MD) patients with end-stage heart failure (HF), continuous flow left ventricular assist device (cf-LVAD) therapy is still controversial due to a progressive nature of MD-associated muscle weakness. METHODS All the MD patients who had cf- VAD implants between March 2013 and August 2019 in our hospital were retrospectively studied. Study end points were death, major LVAD-associated complications or respiratory dysfunction caused by muscular weakness. RESULTS A total of 11 MD patients (Becker type: n = 6; Emery-Dreifuss Myodystrophy: n = 2; Fukuyama subtype: n = 1; Limb-girdle 1B: n = 2) were enrolled. DEMOGRAPHICS median age 41 years (IQR; 29-47); median Japanese Registry for Mechanically Assisted Circulatory Support: level 3 (2-3); a median interval between MD diagnosis and LVAD implantation 9 years (6-18). The pulmonary function test at LVAD implantation showed a median of %VC; 62% (45-82), FEV1%, 82% (81-88). Survival to discharge was 100% without pulmonary complication and early VAD-related complications. During a median follow-up of 38 months (27-53), re-admissions were needed due to device infection (n = 2), cerebrovascular accidents (disabling, n = 2 and non-disabling, n = 2), ventricular tachycardia (n = 4), and right HF (n = 3), respectively. 7 patients received successful heart transplant after a median waiting time of 44 months (34-61); 3 patients are still on the waiting list (waiting time: 21, 38, and 39 months). One patient died of right HF 15 months after VAD implantation. No one had overt pulmonary dysfunction during LVAD support. CONCLUSION In selected MD patients with end-stage HF, cf-LVAD therapy is a viable therapeutic option as bridge to heart transplant.
Collapse
|
26
|
Brashear SE, Wohlgemuth RP, Hu LY, Jbeily EH, Christiansen BA, Smith LR. Collagen cross-links scale with passive stiffness in dystrophic mouse muscles, but are not altered with administration of a lysyl oxidase inhibitor. PLoS One 2022; 17:e0271776. [PMID: 36302059 PMCID: PMC9612445 DOI: 10.1371/journal.pone.0271776] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/10/2022] [Indexed: 11/19/2022] Open
Abstract
In Duchenne muscular dystrophy (DMD), a lack of functional dystrophin leads to myofiber instability and progressive muscle damage that results in fibrosis. While fibrosis is primarily characterized by an accumulation of extracellular matrix (ECM) components, there are changes in ECM architecture during fibrosis that relate more closely to functional muscle stiffness. One of these architectural changes in dystrophic muscle is collagen cross-linking, which has been shown to increase the passive muscle stiffness in models of fibrosis including the mdx mouse, a model of DMD. We tested whether the intraperitoneal injections of beta-aminopropionitrile (BAPN), an inhibitor of the cross-linking enzyme lysyl oxidase, would reduce collagen cross-linking and passive stiffness in young and adult mdx mice compared to saline-injected controls. We found no significant differences between BAPN treated and saline treated mice in collagen cross-linking and stiffness parameters. However, we observed that while collagen cross-linking and passive stiffness scaled positively in dystrophic muscles, collagen fiber alignment scaled with passive stiffness distinctly between muscles. We also observed that the dystrophic diaphragm showed the most dramatic fibrosis in terms of collagen content, cross-linking, and stiffness. Overall, we show that while BAPN was not effective at reducing collagen cross-linking, the positive association between collagen cross-linking and stiffness in dystrophic muscles still show cross-linking as a viable target for reducing passive muscle stiffness in DMD or other fibrotic muscle conditions.
Collapse
Affiliation(s)
- Sarah E. Brashear
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Davis, California, United States of America
| | - Ross P. Wohlgemuth
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Davis, California, United States of America
| | - Lin-Ya Hu
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Davis, California, United States of America
| | - Elias H. Jbeily
- Department of Orthopaedic Surgery, University of California Davis, Sacramento, California, United States of America
| | - Blaine A. Christiansen
- Department of Orthopaedic Surgery, University of California Davis, Sacramento, California, United States of America
| | - Lucas R. Smith
- Department of Neurobiology, Physiology, and Behavior, University of California Davis, Davis, California, United States of America
- Department of Physical Medicine and Rehabilitation, University of California Davis, Sacramento, California, United States of America
- * E-mail:
| |
Collapse
|
27
|
Lin BL, Shin JY, Jeffreys WP, Wang N, Lukban CA, Moorer MC, Velarde E, Hanselman OA, Kwon S, Kannan S, Riddle RC, Ward CW, Pullen SS, Filareto A, Kass DA. Pharmacological TRPC6 inhibition improves survival and muscle function in mice with Duchenne muscular dystrophy. JCI Insight 2022; 7:e158906. [PMID: 36099033 PMCID: PMC9675567 DOI: 10.1172/jci.insight.158906] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022] Open
Abstract
Gene mutations causing loss of dystrophin result in the severe muscle disease known as Duchenne muscular dystrophy (DMD). Despite efforts at genetic repair, DMD therapy remains largely palliative. Loss of dystrophin destabilizes the sarcolemmal membrane, inducing mechanosensitive cation channels to increase calcium entry and promote cell damage and, eventually, muscle dysfunction. One putative channel is transient receptor potential canonical 6 (TRPC6); we have shown that TRPC6 contributed to abnormal force and calcium stress-responses in cardiomyocytes from mice lacking dystrophin that were haplodeficient for utrophin (mdx/utrn+/- [HET] mice). Here, we show in both the HET mouse and the far more severe homozygous mdx/utrn-/- mouse that TRPC6 gene deletion or its selective pharmacologic inhibition (by BI 749327) prolonged survival 2- to 3-fold, improving skeletal and cardiac muscle and bone defects. Gene pathways reduced by BI 749327 treatment most prominently regulated fat metabolism and TGF-β1 signaling. These results support the testing of TRPC6 inhibitors in human trials for other diseases as a novel DMD therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Esteban Velarde
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | | | | | | | | | | | | | - Antonio Filareto
- Research Beyond Borders, Boehringer Ingelheim Pharmaceuticals, Ridgefield, Connecticut, USA
| | - David A. Kass
- Department of Cardiology
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
28
|
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: 1.0] [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
|
29
|
Al Hajri HS, El Husseiny EM, Qayyum H. Chest Pain and Electrocardiographic Changes in a Child With Duchenne Muscular Dystrophy. Cureus 2022; 14:e26105. [PMID: 35747106 PMCID: PMC9207991 DOI: 10.7759/cureus.26105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2022] [Indexed: 11/12/2022] Open
Abstract
A 12-year-old boy known to have Duchenne muscular dystrophy presented to our Emergency Department with acute onset central chest pain. A 12-lead electrocardiogram (ECG) was performed showing ST-segment elevation with reciprocal changes. An echocardiogram showed reduced left ventricular systolic function with an ejection fraction of 45%. Initial cardiac biomarkers were significantly elevated, with troponin-T result recorded at 7,065 ng/L (reference range: 0-14 ng/L). The patient was admitted to the pediatric intensive care unit with a differential diagnosis of acute myocardial infarction or acute myocardial injury related to cardiomyopathy and commenced on an ACE (angiotensin-converting enzyme) inhibitor. Computed tomography (CT) of the coronary arteries was performed, which showed normal coronary arteries and cardiac anatomy. The patient was discharged on day 5 and continues to follow up in the pediatric cardiology clinic. He was commenced on a beta blocker at one-month follow-up when he was asymptomatic.
Collapse
|
30
|
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: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [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
|
31
|
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: 2.3] [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
|
32
|
Abstract
PURPOSE OF REVIEW This review highlights the key studies investigating various types of biomarkers in Duchenne muscular dystrophy (DMD). RECENT FINDINGS Several proteomic and metabolomic studies have been undertaken in both human DMD patients and animal models of DMD that have identified potential biomarkers in DMD. Although there have been a number of proteomic and metabolomic studies that have identified various potential biomarkers in DMD, more definitive studies still need to be undertaken in DMD patients to firmly correlate these biomarkers with diagnosis, disease progression, and monitoring the effects of novel treatment strategies being developed.
Collapse
Affiliation(s)
- Theo Lee-Gannon
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Xuan Jiang
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tara C Tassin
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Pradeep P A Mammen
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
- Heart Failure, Ventricular Assist Device & Heart Transplant Program, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
- Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
33
|
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: 2.3] [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
|
34
|
Tang YC, Tsui PH, Wang CY, Chien YH, Weng HL, Yang CY, Weng WC. Hepatic Steatosis Assessment as a New Strategy for the Metabolic and Nutritional Management of Duchenne Muscular Dystrophy. Nutrients 2022; 14:nu14040727. [PMID: 35215377 PMCID: PMC8875407 DOI: 10.3390/nu14040727] [Citation(s) in RCA: 3] [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: 01/02/2022] [Revised: 02/04/2022] [Accepted: 02/04/2022] [Indexed: 02/04/2023] Open
Abstract
Growing evidence suggests that patients with Duchenne muscular dystrophy (DMD) have an increased risk of obesity and metabolic syndrome (MetS). The aim of this study was to investigate the potential risk factors for MetS and hepatic steatosis in patients with different stages of DMD. A total of 48 patients with DMD were enrolled and classified into three stages according to ambulatory status. Body mass index (BMI), serum fasting glucose, insulin, and lipid profiles including triglycerides (TG) and high-density lipoprotein were measured, and the homeostatic model assessment for insulin resistance (HOMA-IR) index was evaluated. Ultrasound examinations of the liver were performed to assess hepatic steatosis using the Nakagami parameter index (NPI). The results showed that BMI, TG, HOMA-IR, and ultrasound NPI differed significantly among DMD stages (p < 0.05). In contrast to the low rates of conventional MetS indices, including disturbed glucose metabolism (0%), dyslipidemia (14.28%), and insulin resistance (4.76%), a high proportion (40.48%) of the patients had significant hepatic steatosis. The ultrasound NPI increased with DMD progression, and two thirds of the non-ambulatory patients had moderate to severe hepatic steatosis. Steroid treatment was a risk factor for hepatic steatosis in ambulatory patients (p < 0.05). We recommend that DMD patients should undergo ultrasound evaluations for hepatic steatosis for better metabolic and nutritional management.
Collapse
Affiliation(s)
- Ya-Chun Tang
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou, Taoyuan 333423, Taiwan;
| | - Po-Hsiang Tsui
- Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan; (P.-H.T.); (C.-Y.W.)
- Division of Pediatric Gastroenterology, Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, Taoyuan 333423, Taiwan
| | - Chiao-Yin Wang
- Department of Medical Imaging and Radiological Sciences, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan; (P.-H.T.); (C.-Y.W.)
| | - Yin-Hsiu Chien
- Department of Pediatrics, National Taiwan University Hospital, Taipei 100225, Taiwan;
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei 100233, Taiwan
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Hui-Ling Weng
- Department of Dietetics, National Taiwan University Hospital, Taipei 100225, Taiwan;
- School of Nursing, College of Nursing, National Taipei University of Nursing and Health Sciences, Taipei 112303, Taiwan
| | - Chung-Yi Yang
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 824005, Taiwan;
- Department of Medical Imaging, E-Da Hospital, Kaohsiung 824410, Taiwan
| | - Wen-Chin Weng
- Department of Pediatrics, National Taiwan University Hospital, Taipei 100225, Taiwan;
- Department of Pediatrics, College of Medicine, National Taiwan University, Taipei 100233, Taiwan
- Department of Pediatric Neurology, National Taiwan University Children’s Hospital, Taipei 100226, Taiwan
- Correspondence: ; Tel.: +886-2-23123456 (ext. 71609)
| |
Collapse
|
35
|
Laurila PP, Luan P, Wohlwend M, Zanou N, Crisol B, Imamura de Lima T, Goeminne LJE, Gallart-Ayala H, Shong M, Ivanisevic J, Place N, Auwerx J. Inhibition of sphingolipid de novo synthesis counteracts muscular dystrophy. SCIENCE ADVANCES 2022; 8:eabh4423. [PMID: 35089797 PMCID: PMC8797791 DOI: 10.1126/sciadv.abh4423] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 11/01/2021] [Indexed: 05/29/2023]
Abstract
Duchenne muscular dystrophy (DMD), the most common muscular dystrophy, is a severe muscle disorder, causing muscle weakness, loss of independence, and premature death. Here, we establish the link between sphingolipids and muscular dystrophy. Transcripts of sphingolipid de novo biosynthesis pathway are up-regulated in skeletal muscle of patients with DMD and other muscular dystrophies, which is accompanied by accumulation of metabolites of the sphingolipid pathway in muscle and plasma. Pharmacological inhibition of sphingolipid synthesis by myriocin in the mdx mouse model of DMD ameliorated the loss in muscle function while reducing inflammation, improving Ca2+ homeostasis, preventing fibrosis of the skeletal muscle, heart, and diaphragm, and restoring the balance between M1 and M2 macrophages. Myriocin alleviated the DMD phenotype more than glucocorticoids. Our study identifies inhibition of sphingolipid synthesis, targeting multiple pathogenetic pathways simultaneously, as a strong candidate for treatment of muscular dystrophies.
Collapse
Affiliation(s)
- Pirkka-Pekka Laurila
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Peiling Luan
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Martin Wohlwend
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences, Department of Physiology, Faculty of Biology-Medicine, University of Lausanne, Lausanne, Switzerland
| | - Barbara Crisol
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tanes Imamura de Lima
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Ludger J. E. Goeminne
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Nicolas Place
- Institute of Sport Sciences, Department of Physiology, Faculty of Biology-Medicine, University of Lausanne, Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
36
|
A Dystrophin Exon-52 Deleted Miniature Pig Model of Duchenne Muscular Dystrophy and Evaluation of Exon Skipping. Int J Mol Sci 2021; 22:ijms222313065. [PMID: 34884867 PMCID: PMC8657897 DOI: 10.3390/ijms222313065] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal X-linked recessive disorder caused by mutations in the DMD gene and the subsequent lack of dystrophin protein. Recently, phosphorodiamidate morpholino oligomer (PMO)-antisense oligonucleotides (ASOs) targeting exon 51 or 53 to reestablish the DMD reading frame have received regulatory approval as commercially available drugs. However, their applicability and efficacy remain limited to particular patients. Large animal models and exon skipping evaluation are essential to facilitate ASO development together with a deeper understanding of dystrophinopathies. Using recombinant adeno-associated virus-mediated gene targeting and somatic cell nuclear transfer, we generated a Yucatan miniature pig model of DMD with an exon 52 deletion mutation equivalent to one of the most common mutations seen in patients. Exon 52-deleted mRNA expression and dystrophin deficiency were confirmed in the skeletal and cardiac muscles of DMD pigs. Accordingly, dystrophin-associated proteins failed to be recruited to the sarcolemma. The DMD pigs manifested early disease onset with severe bodywide skeletal muscle degeneration and with poor growth accompanied by a physical abnormality, but with no obvious cardiac phenotype. We also demonstrated that in primary DMD pig skeletal muscle cells, the genetically engineered exon-52 deleted pig DMD gene enables the evaluation of exon 51 or 53 skipping with PMO and its advanced technology, peptide-conjugated PMO. The results show that the DMD pigs developed here can be an appropriate large animal model for evaluating in vivo exon skipping efficacy.
Collapse
|
37
|
Blaszczyk E, Gröschel J, Schulz-Menger J. Role of CMR Imaging in Diagnostics and Evaluation of Cardiac Involvement in Muscle Dystrophies. Curr Heart Fail Rep 2021; 18:211-224. [PMID: 34319529 PMCID: PMC8342365 DOI: 10.1007/s11897-021-00521-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 11/18/2022]
Abstract
PURPOSE OF REVIEW This review aims to outline the utility of cardiac magnetic resonance (CMR) in patients with different types of muscular dystrophies for the assessment of myocardial involvement, risk stratification and in guiding therapeutic decisions. RECENT FINDINGS In patients suffering from muscular dystrophies (MD), even mild initial dysfunction may lead to severe heart failure over a time course of years. CMR plays an increasing role in the diagnosis and clinical care of these patients, mostly due to its unique capability to precisely characterize subclinical and progressive changes in cardiac geometry, function in order to differentiate myocardial injury it allows the identification of inflammation, focal and diffuse fibrosis as well as fatty infiltration. CMR may provide additional information in addition to the physical examination, laboratory tests, ECG, and echocardiography. Further trials are needed to investigate the potential impact of CMR on the therapeutic decision-making as well as the assessment of long-term prognosis in different forms of muscular dystrophies. In addition to the basic cardiovascular evaluation, CMR can provide a robust, non-invasive technique for the evaluation of subclinical myocardial tissue injury like fat infiltration and focal and diffuse fibrosis. Furthermore, CMR has a unique capability to detect the progression of myocardial tissue damage in patients with a preserved systolic function.
Collapse
Affiliation(s)
- Edyta Blaszczyk
- Working Group on Cardiovascular Magnetic Resonance, Experimental and Clinical Research Center a joint cooperation between the Charité – Universitätsmedizin Berlin, Department of Internal Medicine and Cardiology and the Max-Delbrueck Center for Molecular Medicine, and HELIOS Klinikum Berlin Buch, Department of Cardiology and Nephrology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Jan Gröschel
- Working Group on Cardiovascular Magnetic Resonance, Experimental and Clinical Research Center a joint cooperation between the Charité – Universitätsmedizin Berlin, Department of Internal Medicine and Cardiology and the Max-Delbrueck Center for Molecular Medicine, and HELIOS Klinikum Berlin Buch, Department of Cardiology and Nephrology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Jeanette Schulz-Menger
- Working Group on Cardiovascular Magnetic Resonance, Experimental and Clinical Research Center a joint cooperation between the Charité – Universitätsmedizin Berlin, Department of Internal Medicine and Cardiology and the Max-Delbrueck Center for Molecular Medicine, and HELIOS Klinikum Berlin Buch, Department of Cardiology and Nephrology, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| |
Collapse
|
38
|
Staunton H, Trennery C, Arbuckle R, Guridi M, Zhuravleva E, Furlong P, Fischer R, Hall R. Development of a Clinical Global Impression of Change (CGI-C) and a Caregiver Global Impression of Change (CaGI-C) measure for ambulant individuals with Duchenne muscular dystrophy. Health Qual Life Outcomes 2021; 19:184. [PMID: 34311756 PMCID: PMC8314490 DOI: 10.1186/s12955-021-01813-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 06/28/2021] [Indexed: 01/14/2023] Open
Abstract
Background In clinical trials for rare diseases, such as Duchenne muscular dystrophy, clinical outcome assessments (COA) used to assess treatment benefit are often generic and may not be sensitive enough to detect change in specific patient populations. Thus, there is a need for disease specific COAs that track meaningful change among individuals. When developing such measures, input from clinicians, caregivers and patients is critical for assessing clinically relevant concepts and ensuring validity of the measure. Method The aim of this study was to develop two Duchenne-specific global impression items for use in clinical trials. The development of the Duchenne Clinical Global Impression of Change (CGI-C) and Caregiver Global Impression of Change (CaGI-C) was informed by findings from concept elicitation (CE) interviews with clinicians, caregivers and individuals with Duchenne. Through cognitive debriefing (CD) interviews, clinicians and caregivers evaluated draft CGI-C and CaGI-C items to ensure relevance and understanding of the items and instructions. Suggestions made during the CD interviews were incorporated into the finalized CGI-C and CaGI-C measures. Results The symptoms most frequently reported by clinicians, caregivers and individuals with Duchenne were muscle weakness, fatigue, cardiac difficulties and pain. Regarding physical functioning, all three populations noted that small changes in functional ability were meaningful, particularly when independence was impacted. Caregivers and clinicians reported that changes in speed, endurance and quality of movement were important, as was improvement in the ability of individuals to keep up with their peers. A change in the ability to complete everyday activities was also significant to families. These results were used to create two global impression of change items and instruction documents for use by clinicians (CGI-C) and caregivers (CaGI-C). Overall, both items were well understood by participants. The descriptions and examples developed from the CE interviews were reported to be relevant and appropriate for illustrating different levels of meaningful change in patients with Duchenne. Modifications were made based on caregiver and clinician CD feedback . Conclusions As part of a holistic measurement strategy, such COA can be incorporated into the clinical trial setting to assess global changes in relevant symptoms and functional impacts associated with Duchenne. Supplementary Information The online version contains supplementary material available at 10.1186/s12955-021-01813-w.
Collapse
Affiliation(s)
| | | | - Rob Arbuckle
- Adelphi Values, Patient-Centered Outcomes, Bollington, UK
| | | | | | - Pat Furlong
- Parent Project Muscular Dystrophy, Hackensack, NJ, USA
| | - Ryan Fischer
- Parent Project Muscular Dystrophy, Hackensack, NJ, USA
| | - Rebecca Hall
- Adelphi Values, Patient-Centered Outcomes, Bollington, UK
| |
Collapse
|
39
|
Starosta A, Konieczny P. Therapeutic aspects of cell signaling and communication in Duchenne muscular dystrophy. Cell Mol Life Sci 2021; 78:4867-4891. [PMID: 33825942 PMCID: PMC8233280 DOI: 10.1007/s00018-021-03821-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/26/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating chromosome X-linked disease that manifests predominantly in progressive skeletal muscle wasting and dysfunctions in the heart and diaphragm. Approximately 1/5000 boys and 1/50,000,000 girls suffer from DMD, and to date, the disease is incurable and leads to premature death. This phenotypic severity is due to mutations in the DMD gene, which result in the absence of functional dystrophin protein. Initially, dystrophin was thought to be a force transducer; however, it is now considered an essential component of the dystrophin-associated protein complex (DAPC), viewed as a multicomponent mechanical scaffold and a signal transduction hub. Modulating signal pathway activation or gene expression through epigenetic modifications has emerged at the forefront of therapeutic approaches as either an adjunct or stand-alone strategy. In this review, we propose a broader perspective by considering DMD to be a disease that affects myofibers and muscle stem (satellite) cells, as well as a disorder in which abrogated communication between different cell types occurs. We believe that by taking this systemic view, we can achieve safe and holistic treatments that can restore correct signal transmission and gene expression in diseased DMD tissues.
Collapse
Affiliation(s)
- Alicja Starosta
- Faculty of Biology, Institute of Human Biology and Evolution, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland
| | - Patryk Konieczny
- Faculty of Biology, Institute of Human Biology and Evolution, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland.
| |
Collapse
|
40
|
LoMauro A, Gandossini S, Russo A, Velardo D, Comi GP, Turconi AC, Bresolin N, Aliverti A, D'Angelo MG. A Multidisciplinary Evaluation of Patients with DMD in An Italian Tertiary Care Center. J Neuromuscul Dis 2020; 8:235-249. [PMID: 33361606 DOI: 10.3233/jnd-190417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
With more widespread prolonged survival, Duchenne muscular dystrophy patients progressively experience multisystem complications. We retrospectively reviewed the charts of 132 Duchenne patients (112 alive/20 dead, age 3.5÷32.3 years) with the aims: 1) to provide a comprehensive description of the clinical status considering different aspects of the disease; 2) to propose a new scoring tool able to consider and pool together heterogeneous different functional. Five functions were analyzed: cardiac, respiratory, nutritional, ambulation and scoliosis. For each function, different items were considered and classified according to clinical severity (as indicated by international guidelines) and an incremental scoring was assigned. In addition, a global score incorporating all functions was defined. The scoring system confirmed that despite the significant protective role of steroids, all functions deteriorated with age. The severity of the global score became significantly higher since the age of 13 years. The severity of cardiac, respiratory and nutritional dysfunction was higher since 18 years. Deceased patients were characterized by significantly worse cardiac function, absence of steroid therapy and later use of respiratory assistive devices. The index proposed in this pilot study is a promising tool able to aggregate and correlate heterogeneous functions. It could become either an individual prognostic indicator of decline or a global score to evaluate changes in clinical trials therefore allowing multicenter studies, optimizing the management of both the primary and the secondary complications of the disease and understanding their relative impact.
Collapse
Affiliation(s)
- A LoMauro
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano, Italy
| | - S Gandossini
- Scientific Institute IRCCS E. Medea, Neuromuscular Unit, Bosisio Parini (Lecco) Italy
| | - A Russo
- Scientific Institute IRCCS E. Medea, Neuromuscular Unit, Bosisio Parini (Lecco) Italy
| | - D Velardo
- Scientific Institute IRCCS E. Medea, Neuromuscular Unit, Bosisio Parini (Lecco) Italy
| | - G P Comi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan
| | - A C Turconi
- Scientific Institute, IRCCS E. Medea- via don Luigi Monza, Bosisio P (Lecco) Italy
| | - N Bresolin
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), Neurology Unit, IRCCS Foundation Ca' Granda Ospedale Maggiore Policlinico, University of Milan
| | - A Aliverti
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Milano, Italy
| | - M G D'Angelo
- Scientific Institute IRCCS E. Medea, Neuromuscular Unit, Bosisio Parini (Lecco) Italy
| |
Collapse
|
41
|
de Souza F, Bittar Braune C, Dos Santos Nucera APC. Duchenne muscular dystrophy: an overview to the cardiologist. Expert Rev Cardiovasc Ther 2020; 18:867-872. [PMID: 32985912 DOI: 10.1080/14779072.2020.1828065] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is the most common form of muscular dystrophy in children, affecting approximately one in 3,500-5,000 liveborn boys. The main signs and symptoms include gait disturbances beginning in early childhood, with later onset of respiratory and cardiac function disorders, both directly affecting the prognosis. AREAS COVERED The recent improvement of mechanical ventilation increased the mean DMD survival age; however, there has been little progress in the treatment and prevention of cardiac complications, which currently predominantly impact survival. Cardiological evaluation with imaging methods, such as echocardiography and magnetic resonance imaging, can improve the understanding and detect changes in cardiac function early. EXPERT OPINION Close monitoring by the cardiologists and early treatment, with adequate heart disease stratification, may be the key to prolong the lives of these patients until more promising therapies are available and can predict DMD prognosis and progression more accurately. The objective of this brief review is to update the cardiologists by highlighting the most relevant aspects of treatment and follow-up, in a practical and concise way.
Collapse
Affiliation(s)
- Fabio de Souza
- Cardiology Section, Department of Specialized Medicine, School of Medicine and Surgery, Federal University of the State of Rio De Janeiro , Rio de Janeiro, Brazil
| | | | - Ana Paula Cassetta Dos Santos Nucera
- Cardiology Section, Department of Specialized Medicine, School of Medicine and Surgery, Federal University of the State of Rio De Janeiro , Rio de Janeiro, Brazil
| |
Collapse
|
42
|
Pradhan S, Das A, Singh H, Chaturvedi S, Singh K, Mishra R, Misra DP, Agarwal V. Steroid Responsiveness in Duchenne Muscular Dystrophy - Predictive Value of Epigenetic Regulator Histone Deacetylase 2. Indian J Pediatr 2020; 87:692-698. [PMID: 32314166 DOI: 10.1007/s12098-020-03183-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 01/02/2020] [Indexed: 01/04/2023]
Abstract
OBJECTIVE To find the role of an epigenetic regulator histone deacetylase (HDAC-2) in predicting steroid responsiveness in patients with Duchenne muscular dystrophy (DMD). METHODS Peripheral blood mononuclear cells were isolated from three milliliters of venous blood sample of patients with DMD for estimation of HDAC-2 levels. The patients were then given prednisolone at 0.75 mg/kg/d and followed up for response. Improvement in muscle power and timed function tests were evaluated 2 monthly for 6 mo. Side- effects of steroids were also noted. RESULTS HDAC-2 values showed a linear correlation with improvement in muscle power at 6 mo as reflected in direct assessment and in various functional tests. When ≥3 point change from baseline was considered in power of various muscle groups at 6 mo as cut off to distinguish good responders (GR) from poor responders (PR) of steroid, a HDAC-2 value of ≥4.40 seemed to be 92.6% sensitive and 100% specific to distinguish GRs. CONCLUSIONS The degree of improvement in power and various timed function tests at 6 mo could be predicted from the HDAC-2 value of the individual patient recorded at baseline. HDAC-2 levels at baseline might be a good biomarker for predicting steroid response in patients with DMD.
Collapse
Affiliation(s)
- Sunil Pradhan
- Department of Neurology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226014, India.
| | - Animesh Das
- Department of Neurology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, 226014, India
| | - Harshit Singh
- Department of Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Saurabh Chaturvedi
- Department of Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Kritika Singh
- Department of Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Ravi Mishra
- Department of Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Durga Prassana Misra
- Department of Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Vikas Agarwal
- Department of Immunology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| |
Collapse
|
43
|
Al-Khalili Szigyarto C. Duchenne Muscular Dystrophy: recent advances in protein biomarkers and the clinical application. Expert Rev Proteomics 2020; 17:365-375. [PMID: 32713262 DOI: 10.1080/14789450.2020.1773806] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Early biomarker discovery studies have praised the value of their emerging results, predicting an unprecedented impact on health care. Biomarkers are expected to provide tests with increased specificity and sensitivity compared to existing measures, improve the decision-making process, and accelerate the development of therapies. For rare disorders, like Duchenne Muscular Dystrophy (DMD) such biomarkers can assist the development of therapies, therefore also helping to find a cure for the disease. AREA COVERED State-of-the-art technologies have been used to identify blood biomarkers for DMD and efforts have been coordinated to develop and promote translation of biomarkers for clinical practice. Biomarker translation to clinical practice is however, adjoined by challenges related to the complexity of the disease, involving numerous biological processes, and the limited sample resources. This review highlights the current progress on the development of biomarkers, describing the proteomics technologies used, the most promising findings and the challenges encountered. EXPERT OPINION Strategies for effective use of samples combined with orthogonal proteomics methods for protein quantification are essential for translating biomarkers to the patient's bed side. Progress is achieved only if strong evidence is provided that the biomarker constitutes a reliable indicator of the patient's health status for a specific context of use.
Collapse
Affiliation(s)
- Cristina Al-Khalili Szigyarto
- Science for Life Laboratory, KTH - Royal Institute of Technology , Solna, Sweden.,School of Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology , Stockholm, Sweden
| |
Collapse
|
44
|
Nikhanj A, Nichols BM, Wang K, Siddiqi ZA, Oudit GY. Evaluating the Diagnostic and Prognostic Value of Biomarkers for Heart Disease and Major Adverse Cardiac Events in Patients With Muscular Dystrophy. EUROPEAN HEART JOURNAL. QUALITY OF CARE & CLINICAL OUTCOMES 2020; 7:564-573. [PMID: 32687175 DOI: 10.1093/ehjqcco/qcaa059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022]
Abstract
AIMS Heart disease is recognized as the leading cause of morbidity and mortality in patients with muscular dystrophy (MD). Our study demonstrates the clinical utility of cardiac biomarkers to improve the diagnosis of cardiomyopathy and prognostication of major adverse cardiac events (MACE) in these vulnerable patients. METHODS AND RESULTS We prospectively followed 117 patients (median age, 42 (interquartile range [IQR], 26-50) years; 49 [41.9%] women) at the Neuromuscular Multidisciplinary clinic diagnosed with a dystrophinopathy, limb-girdle MD, type 1 myotonic dystrophy, or facioscapulohumeral MD. We determined that B-type natriuretic peptide (BNP) and high-sensitive troponin I (hsTnI) were effective diagnostic markers of cardiomyopathy (area under the curve [AUC], 0.64; P=0.017; and AUC, 0.69; P=0.001, respectively). Patient risk stratification for MACE was based on cutoff values of BNP and hsTnI defined a priori as 30.5000 pg/mL and 7.6050 ng/L, respectively. Over a median follow-up period of 2.09 (IQR, 1.17-2.81) years there were 36 confirmed MACE. Multivariate regression analyses showed that patients with BNP and hsTnI levels above the respective cutoff values had a 3.70-fold (P=0.001) and 3.24-fold (P=0.002) greater risk of MACE, respectively, compared to patients with biomarker levels below. Furthermore, patients with biomarker levels above both cutoff values had a 4.08-fold (P=0.001) greater risk of MACE. Inflammatory biomarkers did not show clinical utility for heart disease in these patients. CONCLUSION Our study demonstrates important diagnostic and prognostic value of BNP and hsTnI as part of a comprehensive cardiac assessment to augment the management and treatment of heart disease in patients with MD.
Collapse
Affiliation(s)
- Anish Nikhanj
- Division of Cardiology, University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Bailey Miskew Nichols
- Division of Cardiology, University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Kaiming Wang
- Division of Cardiology, University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Zaeem A Siddiqi
- Division of Neurology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Gavin Y Oudit
- Division of Cardiology, University of Alberta, Edmonton, Canada.,Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
45
|
Meyers TA, Heitzman JA, Townsend D. DMD carrier model with mosaic dystrophin expression in the heart reveals complex vulnerability to myocardial injury. Hum Mol Genet 2020; 29:944-954. [PMID: 31976522 PMCID: PMC7158376 DOI: 10.1093/hmg/ddaa015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/26/2019] [Accepted: 01/20/2020] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating neuromuscular disease that causes progressive muscle wasting and cardiomyopathy. This X-linked disease results from mutations of the DMD allele on the X-chromosome resulting in the loss of expression of the protein dystrophin. Dystrophin loss causes cellular dysfunction that drives the loss of healthy skeletal muscle and cardiomyocytes. As gene therapy strategies strive toward dystrophin restoration through micro-dystrophin delivery or exon skipping, preclinical models have shown that incomplete restoration in the heart results in heterogeneous dystrophin expression throughout the myocardium. This outcome prompts the question of how much dystrophin restoration is sufficient to rescue the heart from DMD-related pathology. Female DMD carrier hearts can shed light on this question, due to their mosaic cardiac dystrophin expression resulting from random X-inactivation. In this work, a dystrophinopathy carrier mouse model was derived by breeding male or female dystrophin-null mdx mice with a wild type mate. We report that these carrier hearts are significantly susceptible to injury induced by one or multiple high doses of isoproterenol, despite expressing ~57% dystrophin. Importantly, only carrier mice with dystrophic mothers showed mortality after isoproterenol. These findings indicate that dystrophin restoration in approximately half of the heart still allows for marked vulnerability to injury. Additionally, the discovery of divergent stress-induced mortality based on parental origin in mice with equivalent dystrophin expression underscores the need for better understanding of the epigenetic, developmental, and even environmental factors that may modulate vulnerability in the dystrophic heart.
Collapse
Affiliation(s)
- Tatyana A Meyers
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Jackie A Heitzman
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - DeWayne Townsend
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Paul and Sheila Wellstone Muscular Dystrophy Center, University of Minnesota Medical School, Minneapolis, MN 55455, USA
- Lillehei Heart Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
46
|
Wittlieb‑Weber CA, Knecht KR, Villa CR, Cunningham C, Conway J, Bock MJ, Gambetta KE, Lal AK, Schumacher KR, Law SP, Deshpande SR, West SC, Friedland‑Little JM, Lytrivi ID, McCulloch MA, Butts RJ, Weber DR, Johnson JN. Risk Factors for Cardiac and Non-cardiac Causes of Death in Males with Duchenne Muscular Dystrophy. Pediatr Cardiol 2020; 41:764-771. [PMID: 32016582 PMCID: PMC7328368 DOI: 10.1007/s00246-020-02309-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/22/2020] [Indexed: 01/14/2023]
Abstract
As survival and neuromuscular function in Duchenne muscular dystrophy (DMD) have improved with glucocorticoid (GC) therapy and ventilatory support, cardiac deaths are increasing. Little is known about risk factors for cardiac and non-cardiac causes of death in DMD. A multi-center retrospective cohort study of 408 males with DMD, followed from January 1, 2005 to December 31, 2015, was conducted to identify risk factors for death. Those dying of cardiac causes were compared to those dying of non-cardiac causes and to those alive at study end. There were 29 (7.1%) deaths at a median age of 19.5 (IQR: 16.9-24.6) years; 8 (27.6%) cardiac, and 21 non-cardiac. Those living were younger [14.9 (IQR: 11.0-19.1) years] than those dying of cardiac [18 (IQR 15.5-24) years, p = 0.03] and non-cardiac [19 (IQR: 16.5-23) years, p = 0.002] causes. GC use was lower for those dying of cardiac causes compared to those living [2/8 (25%) vs. 304/378 (80.4%), p = 0.001]. Last ejection fraction prior to death/study end was lower for those dying of cardiac causes compared to those living (37.5% ± 12.8 vs. 54.5% ± 10.8, p = 0.01) but not compared to those dying of non-cardiac causes (37.5% ± 12.8 vs. 41.2% ± 19.3, p = 0.58). In a large DMD cohort, approximately 30% of deaths were cardiac. Lack of GC use was associated with cardiac causes of death, while systolic dysfunction was associated with death from any cause. Further work is needed to ensure guideline adherence and to define optimal management of systolic dysfunction in males with DMD with hopes of extending survival.
Collapse
Affiliation(s)
- Carol A. Wittlieb‑Weber
- Golisano Children’s Hospital, University of Rochester Medical Center, Rochester, NY, USA,Present Address: Division of Cardiology, The Children’s Hospital of Philadelphia, 34th and Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Kenneth R. Knecht
- Arkansas Children’s Hospital, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Chet R. Villa
- The Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Chentel Cunningham
- Stollery Children’s Hospital, University of Alberta, Edmonton, AB, Canada
| | - Jennifer Conway
- Stollery Children’s Hospital, University of Alberta, Edmonton, AB, Canada
| | - Matthew J. Bock
- Loma Linda University Children’s Hospital, Loma Linda, CA, USA
| | - Katheryn E. Gambetta
- Ann & Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ashwin K. Lal
- Primary Children’s Hospital, University of Utah, Salt Lake City, UT, USA
| | - Kurt R. Schumacher
- C.S. Mott Children’s Hospital, University of Michigan, Ann Arbor, MI, USA
| | - Sabrina P. Law
- Morgan Stanley Children’s Hospital of New York Presbyterian, New York, NY, USA
| | | | - Shawn C. West
- Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA
| | | | - Irene D. Lytrivi
- Morgan Stanley Children’s Hospital of New York Presbyterian, New York, NY, USA
| | | | - Ryan J. Butts
- Children’s Medical Center of Dallas, UT Southwestern Medical Center, Dallas, TX, USA
| | - David R. Weber
- Golisano Children’s Hospital, University of Rochester Medical Center, Rochester, NY, USA
| | | |
Collapse
|
47
|
Razzoli M, Lindsay A, Law ML, Chamberlain CM, Southern WM, Berg M, Osborn J, Engeland WC, Metzger JM, Ervasti JM, Bartolomucci A. Social stress is lethal in the mdx model of Duchenne muscular dystrophy. EBioMedicine 2020; 55:102700. [PMID: 32192914 PMCID: PMC7251247 DOI: 10.1016/j.ebiom.2020.102700] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/10/2020] [Accepted: 02/18/2020] [Indexed: 12/19/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is caused by the loss of dystrophin. Severe and ultimately lethal, DMD progresses relatively slowly in that patients become wheelchair bound only around age twelve with a survival expectancy reaching the third decade of life. Methods The mildly-affected mdx mouse model of DMD, and transgenic DysΔMTB-mdx and Fiona-mdx mice expressing dystrophin or utrophin, respectively, were exposed to either mild (scruffing) or severe (subordination stress) stress paradigms and profiled for their behavioral and physiological responses. A subgroup of mdx mice exposed to subordination stress were pretreated with the beta-blocker metoprolol. Findings Subordination stress caused lethality in ∼30% of mdx mice within 24 h and ∼70% lethality within 48 h, which was not rescued by metoprolol. Lethality was associated with heart damage, waddling gait and hypo-locomotion, as well as marked up-regulation of the hypothalamus-pituitary-adrenocortical axis. A novel cardiovascular phenotype emerged in mdx mice, in that scruffing caused a transient drop in arterial pressure, while subordination stress caused severe and sustained hypotension with concurrent tachycardia. Transgenic expression of dystrophin or utrophin in skeletal muscle protected mdx mice from scruffing and social stress-induced responses including mortality. Interpretation We have identified a robust new stress phenotype in the otherwise mildly affected mdx mouse that suggests relatively benign handling may impact the outcome of behavioural experiments, but which should also expedite the knowledge-based therapy development for DMD. Funding Greg Marzolf Jr. Foundation, Summer's Wish Fund, NIAMS, Muscular Dystrophy Association, University of Minnesota and John and Cheri Gunvalson Trust.
Collapse
Affiliation(s)
- Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Angus Lindsay
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Michelle L Law
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Christopher M Chamberlain
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - William M Southern
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Madeleine Berg
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - John Osborn
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - William C Engeland
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States.
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States.
| |
Collapse
|
48
|
Life expectancy at birth in Duchenne muscular dystrophy: a systematic review and meta-analysis. Eur J Epidemiol 2020; 35:643-653. [PMID: 32107739 PMCID: PMC7387367 DOI: 10.1007/s10654-020-00613-8] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 02/05/2020] [Indexed: 12/29/2022]
Abstract
Several studies indicate that prognosis for survival in Duchenne muscular dystrophy (DMD) has improved in recent decades. However, published evidence is inconclusive and some estimates may be obsolete due to improvements in standards of care, in particular the routine use of mechanical ventilatory support in advanced stages of the disease. In this systematic review and meta-analysis (PROSPERO identifier: CRD42019121800), we searched MEDLINE (through PubMed), CINAHL, Embase, PsycINFO, and Web of Science for studies published from inception up until December 31, 2018, reporting results of life expectancy in DMD. We pooled median survival estimates from individual studies using the median of medians, and weighted median of medians, methods. Risk of bias was established with the Newcastle–Ottawa Scale. Results were stratified by ventilatory support and risk of bias. We identified 15 publications involving 2662 patients from 12 countries from all inhabited continents except Africa. Median life expectancy without ventilatory support ranged between 14.4 and 27.0 years (pooled median: 19.0 years, 95% CI 18.0–20.9; weighted pooled median: 19.4 years, 18.2–20.1). Median life expectancy with ventilatory support, introduced in most settings in the 1990s, ranged between 21.0 and 39.6 years (pooled median: 29.9 years, 26.5–30.8; weighted pooled median: 31.8 years, 29.3–36.2). Risk of bias had little impact on pooled results. In conclusion, median life expectancy at birth in DMD seems to have improved considerably during the last decades. With current standards of care, many patients with DMD can now expect to live into their fourth decade of life.
Collapse
|
49
|
Li JM, Chen H. Recurrent hypotension induced by sacubitril/valsartan in cardiomyopathy secondary to Duchenne muscular dystrophy: A case report. World J Clin Cases 2019; 7:4098-4105. [PMID: 31832414 PMCID: PMC6906571 DOI: 10.12998/wjcc.v7.i23.4098] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/17/2019] [Accepted: 11/19/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD), which is caused by a mutation/deletion in the dystrophin gene on the X-chromosome, is the most common type of neuromuscular disorder in pediatrics. Skeletal muscle weakness progressively develops in DMD patients and usually leads to respiratory failure in the early adolescent years. Cardiac muscle is frequently affected in DMD patients, which leads to a high burden of cardiomyopathy and heart failure. In the era of improved respiratory care, cardiac deaths are becoming the major cause of mortality in DMD patients.
CASE SUMMARY We report the case of a 15-year-old boy who presented to the hospital due to recurrent orthopnea for 6 mo and palpitations for 4 mo. He was diagnosed with progressive muscular dystrophy at the age of 3 years and was confined to a wheelchair at 12 years. He was prescribed diuretics and digoxin at the outpatient clinic; however, his symptoms did not resolve. Sacubitril/valsartan was added 1 mo prior to presentation, but he experienced recurrent episodes of palpitations. The electrocardiogram showed atrial tachycardia with a heart rate of 201 bpm, and he was then hospitalized. Hypotension was found following the administration of sacubitril/valsartan tablets; he could not tolerate even a small dose, always developing tachyarrhythmia. His symptoms were relieved after discontinuing sacubitril/valsartan, and his heart rate was controlled by a small dose of metoprolol tartrate and digoxin. Atrial tachycardia spontaneously converted in this patient, and his symptoms attenuated in the following 6 mo, without palpitation episodes.
CONCLUSION Blood pressure should be closely monitored in DMD patients with advanced heart failure when taking sacubitril/valsartan.
Collapse
Affiliation(s)
- Jia-Min Li
- Department of Cardiology, Zhejiang Provincial Key Laboratory of Cardiovascular Disease Diagnosis and Treatment, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| | - Han Chen
- Department of Cardiology, Zhejiang Provincial Key Laboratory of Cardiovascular Disease Diagnosis and Treatment, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang Province, China
| |
Collapse
|
50
|
Mercuri E, Bönnemann CG, Muntoni F. Muscular dystrophies. Lancet 2019; 394:2025-2038. [PMID: 31789220 DOI: 10.1016/s0140-6736(19)32910-1] [Citation(s) in RCA: 307] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 09/02/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022]
Abstract
Muscular dystrophies are primary diseases of muscle due to mutations in more than 40 genes, which result in dystrophic changes on muscle biopsy. Now that most of the genes responsible for these conditions have been identified, it is possible to accurately diagnose them and implement subtype-specific anticipatory care, as complications such as cardiac and respiratory muscle involvement vary greatly. This development and advances in the field of supportive medicine have changed the standard of care, with an overall improvement in the clinical course, survival, and quality of life of affected individuals. The improved understanding of the pathogenesis of these diseases is being used for the development of novel therapies. In the most common form, Duchenne muscular dystrophy, a few personalised therapies have recently achieved conditional approval and many more are at advanced stages of clinical development. In this Seminar, we concentrate on clinical manifestations, molecular pathogenesis, diagnostic strategy, and therapeutic developments for this group of conditions.
Collapse
Affiliation(s)
- Eugenio Mercuri
- Pediatric Neurology Unit, Università Cattolica del Sacro Cuore Roma, Rome, Italy; Nemo Clinical Centre, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London, UK; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|