1
|
Rogers RG, Antich J, Fournier M, Omidfar A, Sanchez L, Alfaro J, Zarrow J, Manriquez N, Ciullo A, Valle J, Marbán E. Long-term preservation of muscle function and structure by repeated administration of cardiosphere-derived cells in mdx mice. Stem Cell Reports 2025; 20:102468. [PMID: 40118057 DOI: 10.1016/j.stemcr.2025.102468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/23/2025] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive myodegenerative disease that leads to severe muscle weakness and premature death. Mouse cardiosphere-derived cells (mCDCs) and extracellular vesicles (EVs) secreted by human cardiosphere-deriveds (hCDC-EVs) are therapeutic to mice with advanced-stage DMD. Here, we investigated the long-term benefits of monthly dosing when initiated early. At the endpoint, exercise performance and skeletal muscle function were strikingly preserved in mdx mice that had received mCDCs, but not in vehicle control. In contrast, the beneficial effects of hCDC-EVs waned after 6 months, in parallel with the development of anti-hCDC-EV antibodies. Further investigation showed that mCDCs lowered fibrosis and initiated a myogenic response program in mdx skeletal muscle. Thus, early and sustained intervention with mCDCs prevents disease progression for up to 1 year in mdx mice. This discovery offers new insights into how cell therapy can be used to treat DMD and motivates clinical testing of CDCs beginning in newly diagnosed DMD.
Collapse
Affiliation(s)
- Russell G Rogers
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | - Jack Antich
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Mario Fournier
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ariel Omidfar
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Lizbeth Sanchez
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Juliet Alfaro
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jonah Zarrow
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nancy Manriquez
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Alessandra Ciullo
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jackelyn Valle
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
2
|
D’Amario D, Arcudi A, Narducci ML, Novelli V, Canonico F, Parodi A, Dell’Era G, Di Francesco M, Laborante R, Borovac JA, Galli M, Mercuri EM, Vergaro G, Russo AD, D’Amico AT, Bisignani A, Adorisio R, Pompilio G, Patti G. Arrhythmic Risk Stratification and Sudden Cardiac Death Prevention in Duchenne Muscular Dystrophy: A Critical Appraisal. Rev Cardiovasc Med 2025; 26:27089. [PMID: 40160579 PMCID: PMC11951492 DOI: 10.31083/rcm27089] [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: 10/23/2024] [Revised: 11/30/2024] [Accepted: 12/26/2024] [Indexed: 04/02/2025] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic progressive neuromuscular disorder characterized by early-onset proximal muscle weakness and significant long-term pulmonary and cardiac involvement. Due to the early pharmacological treatments and the wider adoption of non-invasive ventilation, life expectancy has significantly increased in recent years, highlighting the relevance of DMD-related cardiomyopathy and fatal arrhythmias, especially in the late stage of the disease. Current guideline-derived evaluation of sudden cardiac death (SCD) in DMD lacks accuracy, leading to inadequate arrhythmic risk stratification and jeopardized SCD prevention strategies. This review aims to outline these critical issues, proposing an integrative approach encompassing manifold tools such as an imaging-derived systematic and comprehensive evaluation (speckle-tracking echocardiography and magnetic resonance imaging), the electrophysiological study, the 3-dimensional electroanatomic mapping, and a multidimensional clinical examination. This approach might lead to more personalized management along with an effective arrhythmia-prevention strategy aiming to balance clinical care goals, patient expectations, and ethical considerations.
Collapse
Affiliation(s)
- Domenico D’Amario
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
- Thoraco-Cardio-Vascular Department, Azienda Ospedaliero-Universitaria Maggiore della Carità, 28100 Novara, Italy
| | - Alessandra Arcudi
- Thoraco-Cardio-Vascular Department, Azienda Ospedaliero-Universitaria Maggiore della Carità, 28100 Novara, Italy
| | - Maria Lucia Narducci
- Department of Cardiovascular Science, Fondazione Policlinico Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Cardiovascular Science, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Valeria Novelli
- Department of Cardiac Surgery, Centro Cardiologico Monzino-IRCCS, 20138 Milan, Italy
| | - Francesco Canonico
- Thoraco-Cardio-Vascular Department, Azienda Ospedaliero-Universitaria Maggiore della Carità, 28100 Novara, Italy
| | - Alessandro Parodi
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
| | - Gabriele Dell’Era
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
| | - Marco Di Francesco
- Department of Cardiovascular Science, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Renzo Laborante
- Department of Cardiovascular Science, Fondazione Policlinico Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Josip Andelo Borovac
- Clinic for Heart and Vascular Diseases, University Hospital of Split, 21000 Split, Croatia
| | - Mattia Galli
- Maria Cecilia Hospital, GVM Care and Research, 48033 Cotignola, Italy
| | - Eugenio Maria Mercuri
- Department of Cardiovascular Science, Fondazione Policlinico Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Cardiovascular Science, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | | | - Antonio Dello Russo
- Cardiology and Arrhythmology Clinic, University Hospital “Ospedali Riuniti Umberto I-Lancisi-Salesi”, 60126 Ancona, Italy
| | - Anthea Tonia D’Amico
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
| | - Antonio Bisignani
- Center of Excellence in Cardiovascular Sciences, Ospedale Isola Tiberina-Gemelli Isola, 00153 Rome, Italy
| | - Rachele Adorisio
- Advanced Cardiovascular Therapy Unit, Bambino Gesù Pediatric Hospital and Research Institute, 00165, Rome, Italy
| | - Giulio Pompilio
- Department of Cardiac Surgery, Centro Cardiologico Monzino-IRCCS, 20138 Milan, Italy
| | - Giuseppe Patti
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy
- Thoraco-Cardio-Vascular Department, Azienda Ospedaliero-Universitaria Maggiore della Carità, 28100 Novara, Italy
| |
Collapse
|
3
|
Hakimi M, Burnham T, Ramsay J, Cheung JW, Goyal NA, Jefferies JL, Donaldson D. Electrophysiologic and cardiovascular manifestations of Duchenne and Becker muscular dystrophies. Heart Rhythm 2025; 22:192-202. [PMID: 38997055 DOI: 10.1016/j.hrthm.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/14/2024]
Abstract
There have been significant advances in the diagnosis and management of the hereditary muscular disorders Duchenne and Becker muscular dystrophy (DMD and BMD). Cardiac electrophysiologic and cardiovascular involvement has long been important in the surveillance, care, and prognosis of patients with both BMD and DMD and is the leading cause of mortality in patients with DMD. With improved long-term prognosis, rhythm disorders and progressive cardiomyopathy with resultant heart failure are increasingly common. This review aimed to provide an overview to electrophysiologists and cardiologists of the cardiac electrophysiologic phenotypes and genetics of BMD and DMD and to highlight the recent discoveries that have advanced clinical course and management. A systematic review was performed of the diagnosis and management of DMD and BMD. The Cochrane Library, PubMed, MEDLINE, Europe PubMed Central, AMED, and Embase databases were accessed for available evidence. The research reported in this paper adhered to Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Evidence from randomized controlled trials and studies cited in expert consensus and practice guidelines are examined. Advanced imaging techniques and a spectrum of rhythm disorders associated with the progressive cardiomyopathy are presented. Early initiation of heart failure therapies, the role of cardiac implantable devices, and novel gene therapies approved for use with the potential to alter the disease course are discussed. When profound cardiac and cardiac electrophysiologic involvement is diagnosed and treated earlier, outcomes for DMD and BMD patients may be improved.
Collapse
Affiliation(s)
- Matthew Hakimi
- Division of Cardiology, Weill Cornell Medical, New York, New York
| | - Tyson Burnham
- Division of Cardiology, Department of Medicine, University of California at Irvine, Irvine Medical Center, Orange, California.
| | - Jay Ramsay
- Division of Cardiology, Department of Medicine, University of California at Irvine, Irvine Medical Center, Orange, California
| | - Jim W Cheung
- Division of Cardiology, Weill Cornell Medical, New York, New York
| | - Namita A Goyal
- Department of Neurology, University of California at Irvine, Irvine Medical Center, Orange, California
| | | | - David Donaldson
- Division of Cardiology, Department of Medicine, University of California at Irvine, Irvine Medical Center, Orange, California
| |
Collapse
|
4
|
Lee J, Park SE, Kim M, Kim H, Kwon JY, Jeon HB, Chang JW, Lee J. Safety and Tolerability of Wharton's Jelly-Derived Mesenchymal Stem Cells for Patients With Duchenne Muscular Dystrophy: A Phase 1 Clinical Study. J Clin Neurol 2025; 21:40-52. [PMID: 39778566 PMCID: PMC11711273 DOI: 10.3988/jcn.2024.0299] [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: 07/01/2024] [Revised: 10/15/2024] [Accepted: 10/31/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND AND PURPOSE This study was an open-label, dose-escalation, phase 1 clinical trial to determine the safety and dose of EN001 for patients with Duchenne muscular dystrophy (DMD). EN001, developed by ENCell, are allogeneic early-passage Wharton's jelly-derived mesenchymal stem cells that originate at the umbilical cord, with preclinical studies demonstrating their high therapeutic efficacy for DMD. METHODS This phase 1 clinical trial explored the safety and tolerability of EN001 as a potential treatment option for patients with DMD. Six pediatric participants with DMD were divided into two subgroups of equal size: low-dose EN001 (5.0×10⁵ cells/kg) and high-dose EN001 (2.5×10⁶ cells/kg). All participants were monitored for 12 weeks after EN001 administration to assess its safety. Dose-limiting toxicity (DLT) was evaluated across 2 weeks post administration. Exploratory efficacy was evaluated by measuring serum creatine kinase levels, and functional evaluations-including spirometry, myometry, the North Star Ambulatory Assessment, and the 6-minute walk test-were conducted at week 12 and compared with the baseline values. RESULTS No participants experienced serious adverse events related to EN001 injection during the 12-week follow-up period. Mild adverse events included injection-related local erythema, edema, parosmia, and headache, but DLT was not observed. Functional evaluations at week 12 revealed no significant changes from baseline. CONCLUSIONS These results demonstrated that EN001 are safe and well tolerated for patients with DMD, and did not cause serious adverse events. The efficacy of EN001 could be confirmed through larger-scale future studies that incorporate repeated dosing and have a randomized controlled trial design.
Collapse
Affiliation(s)
- Jiwon Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang Eon Park
- Cell and Gene Therapy Research Institute, ENCell Co. Ltd., Seoul, Korea
- Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Mira Kim
- Clinical Development Department, ENCell Co. Ltd., Seoul, Korea
| | - Hyeongseop Kim
- Cell and Gene Therapy Research Institute, ENCell Co. Ltd., Seoul, Korea
- Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
| | - Jeong-Yi Kwon
- Department of Physical and Rehabilitation Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hong Bae Jeon
- Cell and Gene Therapy Research Institute, ENCell Co. Ltd., Seoul, Korea
| | - Jong Wook Chang
- Cell and Gene Therapy Research Institute, ENCell Co. Ltd., Seoul, Korea
- Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Jeehun Lee
- Department of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Cell & Gene Therapy Institute, Samsung Medical Center, Seoul, Korea.
| |
Collapse
|
5
|
McDonald CM, Signorovitch J, Mercuri E, Niks EH, Wong B, Fillbrunn M, Sajeev G, Yim E, Dieye I, Miller D, Ward SJ, Goemans N. Functional trajectories before and after loss of ambulation in Duchenne muscular dystrophy and implications for clinical trials. PLoS One 2024; 19:e0304099. [PMID: 38829874 PMCID: PMC11146704 DOI: 10.1371/journal.pone.0304099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 04/24/2024] [Indexed: 06/05/2024] Open
Abstract
This study examined functional trajectories of subjects during the transition phase between ambulatory and non-ambulatory Duchenne muscular dystrophy (DMD) to inform clinical trial designs for new therapeutics. Ambulatory, pulmonary, and upper limb function leading up to loss of ambulation (LoA) and non-ambulatory measures following LoA were quantified; time ordering of pulmonary and upper limb milestones relative to LoA were determined; and the 10-second time threshold for 10-meter walk/run (10MWR) as a marker of approaching LOA was explored. Included in this analysis were 51 subjects aged between 7 and 18 years who experienced LoA during follow-up in the PRO-DMD-01 natural history study. Mean age at LoA was 12.7 (7.1-18.6) years. Mean annual rates of decline in forced vital capacity (FVC) <80%-predicted and performance of upper limb (PUL) 1.2 total score were smaller before than after LoA, but not significantly (FVC %-predicted: 5.6% vs. 10.1%, p = 0.21; PUL 1.2 total score: 2.3 vs. 3.8 units, p = 0.20). More than half of patients experienced clinically significant deficits in FVC %-predicted and PUL 1.2 before experiencing LoA. Among subjects with baseline 10MWR >10 s, those with <1 year to LoA had similar mean ages but significantly worse mean ambulatory function at baseline compared to those with ≥1 year to LoA. Enriching DMD clinical trials for patients with declining pulmonary or upper limb function is achievable without restricting enrollment to non-ambulatory patients. The sequencing of LoA and initial deficits in pulmonary and upper limb function varied across patients and highlights the potential for composite outcomes or multi-outcome trial designs to assess disease-modifying therapies more comprehensively.
Collapse
Affiliation(s)
- Craig M. McDonald
- Department of Physical Medicine and Rehabilitation and Department of Pediatrics, University of California Davis Health System, Sacramento, California, United States of America
| | - James Signorovitch
- Analysis Group Inc., Boston, Massachusetts, United States of America
- Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
| | - Eugenio Mercuri
- Child Neurology Unit e Centro Nemo, IRCCS Fondazione Policlinico Gemelli, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Erik H. Niks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Brenda Wong
- Department of Pediatrics and Neurology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Mirko Fillbrunn
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Gautam Sajeev
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Erica Yim
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Ibrahima Dieye
- Analysis Group Inc., Boston, Massachusetts, United States of America
| | - Debra Miller
- CureDuchenne, Newport Beach, California, United States of America
| | - Susan J. Ward
- Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America
| | | | | |
Collapse
|
6
|
Bryl R, Kulus M, Bryja A, Domagała D, Mozdziak P, Antosik P, Bukowska D, Zabel M, Dzięgiel P, Kempisty B. Cardiac progenitor cell therapy: mechanisms of action. Cell Biosci 2024; 14:30. [PMID: 38444042 PMCID: PMC10913616 DOI: 10.1186/s13578-024-01211-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 02/17/2024] [Indexed: 03/07/2024] Open
Abstract
Heart failure (HF) is an end-stage of many cardiac diseases and one of the main causes of death worldwide. The current management of this disease remains suboptimal. The adult mammalian heart was considered a post-mitotic organ. However, several reports suggest that it may possess modest regenerative potential. Adult cardiac progenitor cells (CPCs), the main players in the cardiac regeneration, constitute, as it may seem, a heterogenous group of cells, which remain quiescent in physiological conditions and become activated after an injury, contributing to cardiomyocytes renewal. They can mediate their beneficial effects through direct differentiation into cardiac cells and activation of resident stem cells but majorly do so through paracrine release of factors. CPCs can secrete cytokines, chemokines, and growth factors as well as exosomes, rich in proteins, lipids and non-coding RNAs, such as miRNAs and YRNAs, which contribute to reparation of myocardium by promoting angiogenesis, cardioprotection, cardiomyogenesis, anti-fibrotic activity, and by immune modulation. Preclinical studies assessing cardiac progenitor cells and cardiac progenitor cells-derived exosomes on damaged myocardium show that administration of cardiac progenitor cells-derived exosomes can mimic effects of cell transplantation. Exosomes may become new promising therapeutic strategy for heart regeneration nevertheless there are still several limitations as to their use in the clinic. Key questions regarding their dosage, safety, specificity, pharmacokinetics, pharmacodynamics and route of administration remain outstanding. There are still gaps in the knowledge on basic biology of exosomes and filling them will bring as closer to translation into clinic.
Collapse
Affiliation(s)
- Rut Bryl
- Section of Regenerative Medicine and Cancer Research, Natural Sciences Club, Faculty of Biology, Adam Mickiewicz University, Poznań, Poznan, 61-614, Poland
| | - Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University, Torun, 87-100, Poland
| | - Artur Bryja
- Department of Human Morphology and Embryology, Division of Anatomy, Wroclaw Medical University, Wroclaw, 50-367, Poland
| | - Dominika Domagała
- Department of Human Morphology and Embryology, Division of Anatomy, Wroclaw Medical University, Wroclaw, 50-367, Poland
| | - Paul Mozdziak
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, 27695, USA
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC, 27695, USA
| | - Paweł Antosik
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University, Torun, 87-100, Poland
| | - Dorota Bukowska
- Department of Diagnostics and Clinical Sciences, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, 87-100, Poland
| | - Maciej Zabel
- Division of Anatomy and Histology, University of Zielona Góra, Zielona Góra, 65-046, Poland
- Department of Human Morphology and Embryology, Division of Histology and Embryology, Wroclaw Medical University, Wroclaw, 50-368, Poland
| | - Piotr Dzięgiel
- Department of Human Morphology and Embryology, Division of Histology and Embryology, Wroclaw Medical University, Wroclaw, 50-368, Poland
| | - Bartosz Kempisty
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University, Torun, 87-100, Poland.
- Department of Human Morphology and Embryology, Division of Anatomy, Wroclaw Medical University, Wroclaw, 50-367, Poland.
- Physiology Graduate Faculty, North Carolina State University, Raleigh, NC, 27695, USA.
- Department of Obstetrics and Gynaecology, University Hospital and Masaryk University, Brno, 62500, Czech Republic.
| |
Collapse
|
7
|
Akat A, Karaöz E. Cell Therapy Strategies on Duchenne Muscular Dystrophy: A Systematic Review of Clinical Applications. Stem Cell Rev Rep 2024; 20:138-158. [PMID: 37955832 DOI: 10.1007/s12015-023-10653-8] [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] [Accepted: 11/03/2023] [Indexed: 11/14/2023]
Abstract
Duchenne Muscular Dystrophy (DMD) is an inherited genetic disorder characterized by progressive degeneration of muscle tissue, leading to functional disability and premature death. Despite extensive research efforts, the discovery of a cure for DMD continues to be elusive, emphasizing the need to investigate novel treatment approaches. Cellular therapies have emerged as prospective approaches to address the underlying pathophysiology of DMD. This review provides an examination of the present situation regarding cell-based therapies, including CD133 + cells, muscle precursor cells, mesoangioblasts, bone marrow-derived mononuclear cells, mesenchymal stem cells, cardiosphere-derived cells, and dystrophin-expressing chimeric cells. A total of 12 studies were found eligible to be included as they were completed cell therapy clinical trials, clinical applications, or case reports with quantitative results. The evaluation encompassed an examination of limitations and potential advancements in this particular area of research, along with an assessment of the safety and effectiveness of cell-based therapies in the context of DMD. In general, the available data indicates that diverse cell therapy approaches may present a new, safe, and efficacious treatment modality for patients diagnosed with DMD. However, further studies are required to comprehensively understand the most advantageous treatment approach and therapeutic capacity.
Collapse
Affiliation(s)
- Ayberk Akat
- Life Park Hospital, Cellular and Biological Products Manufacturing Center, Ragıp Kenan Sok. No:8, Ortakoy, 99010, Nicosia (Lefkosa), Cyprus.
| | - Erdal Karaöz
- Liv Hospital Ulus, Regenerative Medicine and Stem Cell Center, Istanbul, Turkey
| |
Collapse
|
8
|
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
|
9
|
Chepeleva EV. Cell Therapy in the Treatment of Coronary Heart Disease. Int J Mol Sci 2023; 24:16844. [PMID: 38069167 PMCID: PMC10706847 DOI: 10.3390/ijms242316844] [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: 11/07/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Heart failure is a leading cause of death in patients who have suffered a myocardial infarction. Despite the timely use of modern reperfusion therapies such as thrombolysis, surgical revascularization and balloon angioplasty, they are sometimes unable to prevent the development of significant areas of myocardial damage and subsequent heart failure. Research efforts have focused on developing strategies to improve the functional status of myocardial injury areas. Consequently, the restoration of cardiac function using cell therapy is an exciting prospect. This review describes the characteristics of various cell types relevant to cellular cardiomyoplasty and presents findings from experimental and clinical studies investigating cell therapy for coronary heart disease. Cell delivery methods, optimal dosage and potential treatment mechanisms are discussed.
Collapse
Affiliation(s)
- Elena V. Chepeleva
- Federal State Budgetary Institution National Medical Research Center Named after Academician E.N. Meshalkin of the Ministry of Health of the Russian Federation, 15, Rechkunovskaya Str., 630055 Novosibirsk, Russia;
- Research Institute of Clinical and Experimental Lymphology—Branch of the Institute of Cytology and Genetics Siberian Branch of Russian Academy of Sciences, 2, Timakova Str., 630060 Novosibirsk, Russia
| |
Collapse
|
10
|
Ranjan P, Colin K, Dutta RK, Verma SK. Challenges and future scope of exosomes in the treatment of cardiovascular diseases. J Physiol 2023; 601:4873-4893. [PMID: 36398654 PMCID: PMC10192497 DOI: 10.1113/jp282053] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/21/2022] [Indexed: 07/28/2023] Open
Abstract
Exosomes are nanosized vesicles that carry biologically diverse molecules for intercellular communication. Researchers have been trying to engineer exosomes for therapeutic purposes by using different approaches to deliver biologically active molecules to the various target cells efficiently. Recent technological advances may allow the biodistribution and pharmacokinetics of exosomes to be modified to meet scientific needs with respect to specific diseases. However, it is essential to determine an exosome's optimal dosage and potential side effects before its clinical use. Significant breakthroughs have been made in recent decades concerning exosome labelling and imaging techniques. These tools provide in situ monitoring of exosome biodistribution and pharmacokinetics and pinpoint targetability. However, because exosomes are nanometres in size and vary significantly in contents, a deeper understanding is required to ensure accurate monitoring before they can be applied in clinical settings. Different research groups have established different approaches to elucidate the roles of exosomes and visualize their spatial properties. This review covers current and emerging strategies for in vivo and in vitro exosome imaging and tracking for potential studies.
Collapse
Affiliation(s)
- Prabhat Ranjan
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL-35233
| | - Karen Colin
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL-35233
- UAB School of Health Professions, The University of Alabama at Birmingham, Birmingham, AL
| | - Roshan Kumar Dutta
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL-35233
| | - Suresh Kumar Verma
- Department of Medicine, Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL-35233
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
11
|
Roberts TC, Wood MJA, Davies KE. Therapeutic approaches for Duchenne muscular dystrophy. Nat Rev Drug Discov 2023; 22:917-934. [PMID: 37652974 DOI: 10.1038/s41573-023-00775-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2023] [Indexed: 09/02/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a monogenic muscle-wasting disorder and a priority candidate for molecular and cellular therapeutics. Although rare, it is the most common inherited myopathy affecting children and so has been the focus of intense research activity. It is caused by mutations that disrupt production of the dystrophin protein, and a plethora of drug development approaches are under way that aim to restore dystrophin function, including exon skipping, stop codon readthrough, gene replacement, cell therapy and gene editing. These efforts have led to the clinical approval of four exon skipping antisense oligonucleotides, one stop codon readthrough drug and one gene therapy product, with other approvals likely soon. Here, we discuss the latest therapeutic strategies that are under development and being deployed to treat DMD. Lessons from these drug development programmes are likely to have a major impact on the DMD field, but also on molecular and cellular medicine more generally. Thus, DMD is a pioneer disease at the forefront of future drug discovery efforts, with these experimental treatments paving the way for therapies using similar mechanisms of action being developed for other genetic diseases.
Collapse
Affiliation(s)
- Thomas C Roberts
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK.
- Department of Paediatrics, University of Oxford, Oxford, UK.
- MDUK Oxford Neuromuscular Centre, Oxford, UK.
| | - Matthew J A Wood
- Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Department of Paediatrics, University of Oxford, Oxford, UK
- MDUK Oxford Neuromuscular Centre, Oxford, UK
| | - Kay E Davies
- MDUK Oxford Neuromuscular Centre, Oxford, UK.
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
12
|
Wilton-Clark H, Yokota T. CRISPR-Cas9-mediated exon skipping as a cardioprotective strategy in Duchenne muscular dystrophy. Mol Ther Methods Clin Dev 2023; 30:500-501. [PMID: 37693945 PMCID: PMC10491811 DOI: 10.1016/j.omtm.2023.08.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Affiliation(s)
- Harry Wilton-Clark
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
13
|
Bez Batti Angulski A, Hosny N, Cohen H, Martin AA, Hahn D, Bauer J, Metzger JM. Duchenne muscular dystrophy: disease mechanism and therapeutic strategies. Front Physiol 2023; 14:1183101. [PMID: 37435300 PMCID: PMC10330733 DOI: 10.3389/fphys.2023.1183101] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/24/2023] [Indexed: 07/13/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe, progressive, and ultimately fatal disease of skeletal muscle wasting, respiratory insufficiency, and cardiomyopathy. The identification of the dystrophin gene as central to DMD pathogenesis has led to the understanding of the muscle membrane and the proteins involved in membrane stability as the focal point of the disease. The lessons learned from decades of research in human genetics, biochemistry, and physiology have culminated in establishing the myriad functionalities of dystrophin in striated muscle biology. Here, we review the pathophysiological basis of DMD and discuss recent progress toward the development of therapeutic strategies for DMD that are currently close to or are in human clinical trials. The first section of the review focuses on DMD and the mechanisms contributing to membrane instability, inflammation, and fibrosis. The second section discusses therapeutic strategies currently used to treat DMD. This includes a focus on outlining the strengths and limitations of approaches directed at correcting the genetic defect through dystrophin gene replacement, modification, repair, and/or a range of dystrophin-independent approaches. The final section highlights the different therapeutic strategies for DMD currently in clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
14
|
Heydemann A, Siemionow M. A Brief Review of Duchenne Muscular Dystrophy Treatment Options, with an Emphasis on Two Novel Strategies. Biomedicines 2023; 11:biomedicines11030830. [PMID: 36979809 PMCID: PMC10044847 DOI: 10.3390/biomedicines11030830] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Despite the full cloning of the Dystrophin cDNA 35 years ago, no effective treatment exists for the Duchenne Muscular Dystrophy (DMD) patients who have a mutation in this gene. Many treatment options have been considered, investigated preclinically and some clinically, but none have circumvented all barriers and effectively treated the disease without burdening the patients with severe side-effects. However, currently, many novel therapies are in the pipelines of research labs and pharmaceutical companies and many of these have progressed to clinical trials. A brief review of these promising therapies is presented, followed by a description of two novel technologies that when utilized together effectively treat the disease in the mdx mouse model. One novel technology is to generate chimeric cells from the patient’s own cells and a normal donor. The other technology is to systemically transplant these cells into the femur via the intraosseous route.
Collapse
Affiliation(s)
- Ahlke Heydemann
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60607, USA
- Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60607, USA
- Correspondence:
| | - Maria Siemionow
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
15
|
Wilton-Clark H, Yokota T. Biological and genetic therapies for the treatment of Duchenne muscular dystrophy. Expert Opin Biol Ther 2023; 23:49-59. [PMID: 36409820 DOI: 10.1080/14712598.2022.2150543] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Duchenne muscular dystrophy is a lethal genetic disease which currently has no cure, and poor standard treatment options largely focused on symptom relief. The development of multiple biological and genetic therapies is underway across various stages of clinical progress which could markedly affect how DMD patients are treated in the future. AREAS COVERED The purpose of this review is to provide an introduction to the different therapeutic modalities currently being studied, as well as a brief description of their progress to date and relative advantages and disadvantages for the treatment of DMD. This review discusses exon skipping therapy, microdystrophin therapy, stop codon readthrough therapy, CRISPR-based gene editing, cell-based therapy, and utrophin upregulation. Secondary therapies addressing nonspecific symptoms of DMD were excluded. EXPERT OPINION Despite the vast potential held by gene replacement therapy options such as microdystrophin production and utrophin upregulation, safety risks inherent to the adeno-associated virus delivery vector might hamper the clinical viability of these approaches until further improvements can be made. Of the mutation-specific therapies, exon skipping therapy remains the most extensively validated and explored option, and the cell-based CAP-1002 therapy may prove to be a suitable adjunct therapy filling the urgent need for cardiac-specific therapies.
Collapse
Affiliation(s)
- Harry Wilton-Clark
- Faculty of Medicine and Dentistry, Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Toshifumi Yokota
- Faculty of Medicine and Dentistry, Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
16
|
Salerno N, Salerno L, Marino F, Scalise M, Chiefalo A, Panuccio G, De Angelis A, Cianflone E, Urbanek K, Torella D. Myocardial regeneration protocols towards the routine clinical scenario: An unseemly path from bench to bedside. EClinicalMedicine 2022; 50:101530. [PMID: 35799845 PMCID: PMC9253597 DOI: 10.1016/j.eclinm.2022.101530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED Heart failure secondary to cardiomyocyte loss and/or dysfunction is the number one killer worldwide. The field of myocardial regeneration with its far-reaching primary goal of cardiac remuscularization and its hard-to-accomplish translation from bench to bedside, has been filled with ups and downs, steps forward and steps backward, controversies galore and, unfortunately, scientific scandals. Despite the present morass in which cardiac remuscularization is stuck in, the search for clinically effective regenerative approaches remains keenly active. Starting with a concise overview of the still highly debated regenerative capacity of the adult mammalian heart, we focus on the main interventions, that have reached or are close to clinical use, critically discussing key findings, successes, and failures. Finally, some promising and innovative approaches for myocardial repair/regeneration still at the pre-clinical stage are discussed to offer a holistic view on the future of myocardial repair/regeneration for the prevention/management of heart failure in the clinical scenario. FUNDING This research was funded by Grants from the Ministry of University and Research PRIN2015 2015ZTT5KB_004; PRIN2017NKB2N4_005; PON-AIM - 1829805-2.
Collapse
Affiliation(s)
- Nadia Salerno
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
| | - Luca Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Fabiola Marino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Mariangela Scalise
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Antonio Chiefalo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Giuseppe Panuccio
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
| | - Antonella De Angelis
- Department of Experimental Medicine, University of Campania “L. Vanvitelli”, 80138 Naples, Italy
| | - Eleonora Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100, Catanzaro, Italy
| | - Konrad Urbanek
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80125, Naples, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
- Corresponding author.
| |
Collapse
|
17
|
Deng J, Zhang J, Shi K, Liu Z. Drug development progress in duchenne muscular dystrophy. Front Pharmacol 2022; 13:950651. [PMID: 35935842 PMCID: PMC9353054 DOI: 10.3389/fphar.2022.950651] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/28/2022] [Indexed: 12/22/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe, progressive, and incurable X-linked disorder caused by mutations in the dystrophin gene. Patients with DMD have an absence of functional dystrophin protein, which results in chronic damage of muscle fibers during contraction, thus leading to deterioration of muscle quality and loss of muscle mass over time. Although there is currently no cure for DMD, improvements in treatment care and management could delay disease progression and improve quality of life, thereby prolonging life expectancy for these patients. Furthermore, active research efforts are ongoing to develop therapeutic strategies that target dystrophin deficiency, such as gene replacement therapies, exon skipping, and readthrough therapy, as well as strategies that target secondary pathology of DMD, such as novel anti-inflammatory compounds, myostatin inhibitors, and cardioprotective compounds. Furthermore, longitudinal modeling approaches have been used to characterize the progression of MRI and functional endpoints for predictive purposes to inform Go/No Go decisions in drug development. This review showcases approved drugs or drug candidates along their development paths and also provides information on primary endpoints and enrollment size of Ph2/3 and Ph3 trials in the DMD space.
Collapse
Affiliation(s)
- Jiexin Deng
- School of Nursing and Health, Henan University, Kaifeng, China
- *Correspondence: Jiexin Deng, ; Zhigang Liu,
| | - Junshi Zhang
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Keli Shi
- School of Medicine, Henan University, Kaifeng, China
| | - Zhigang Liu
- Department of Orthopedics, First Affiliated Hospital of Henan University, Kaifeng, China
- *Correspondence: Jiexin Deng, ; Zhigang Liu,
| |
Collapse
|
18
|
Emerging therapies for Duchenne muscular dystrophy. Lancet Neurol 2022; 21:814-829. [DOI: 10.1016/s1474-4422(22)00125-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 02/21/2022] [Accepted: 03/18/2022] [Indexed: 12/11/2022]
|
19
|
Vadakke‐Madathil S, Chaudhry HW. Concepts of Cell Therapy and Myocardial Regeneration. Interv Cardiol 2022. [DOI: 10.1002/9781119697367.ch30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
20
|
Long-Term Protective Effect of Human Dystrophin Expressing Chimeric (DEC) Cell Therapy on Amelioration of Function of Cardiac, Respiratory and Skeletal Muscles in Duchenne Muscular Dystrophy. Stem Cell Rev Rep 2022; 18:2872-2892. [PMID: 35590083 PMCID: PMC9622520 DOI: 10.1007/s12015-022-10384-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 12/12/2022]
Abstract
Duchenne Muscular Dystrophy (DMD) is a lethal disease caused by mutations in dystrophin encoding gene, causing progressive degeneration of cardiac, respiratory, and skeletal muscles leading to premature death due to cardiac and respiratory failure. Currently, there is no cure for DMD. Therefore, novel therapeutic approaches are needed for DMD patients. We have previously reported functional improvements which correlated with increased dystrophin expression following administration of dystrophin expressing chimeric (DEC) cells of myoblast origin to the mdx mouse models of DMD. In the current study, we confirmed dose-dependent protective effect of human DEC therapy created from myoblasts of normal and DMD-affected donors, on restoration of dystrophin expression and amelioration of cardiac, respiratory, and skeletal muscle function at 180 days after systemic-intraosseous DEC administration to mdx/scid mouse model of DMD. Functional improvements included maintenance of ejection fraction and fractional shortening levels on echocardiography, reduced enhanced pause and expiration time on plethysmography and improved grip strength and maximum stretch induced contraction of skeletal muscles. Improved function was associated with amelioration of mdx muscle pathology revealed by reduced muscle fibrosis, reduced inflammation and improved muscle morphology confirmed by reduced number of centrally nucleated fibers and normalization of muscle fiber diameters. Our findings confirm the long-term systemic effect of DEC therapy in the most severely affected by DMD organs including heart, diaphragm, and long skeletal muscles. These encouraging preclinical data introduces human DEC as a novel therapeutic modality of Advanced Therapy Medicinal Product (ATMP) with the potential to improve or halt the progression of DMD and enhance quality of life of DMD patients.
Collapse
|
21
|
Birnkrant DJ, Bello L, Butterfield RJ, Carter JC, Cripe LH, Cripe TP, McKim DA, Nandi D, Pegoraro E. Cardiorespiratory management of Duchenne muscular dystrophy: emerging therapies, neuromuscular genetics, and new clinical challenges. THE LANCET RESPIRATORY MEDICINE 2022; 10:403-420. [DOI: 10.1016/s2213-2600(21)00581-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 11/01/2021] [Accepted: 12/14/2021] [Indexed: 01/06/2023]
|
22
|
Davis DR. Cell therapy for patients with Duchenne muscular dystrophy. Lancet 2022; 399:1024-1025. [PMID: 35279246 DOI: 10.1016/s0140-6736(22)00185-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022]
Affiliation(s)
- Darryl R Davis
- University of Ottawa Heart Institute, Division of Cardiology, Department of Medicine, and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1Y 4W7, Canada.
| |
Collapse
|
23
|
McDonald CM, Marbán E, Hendrix S, Hogan N, Ruckdeschel Smith R, Eagle M, Finkel RS, Tian C, Janas J, Harmelink MM, Varadhachary AS, Taylor MD, Hor KN, Mayer OH, Henricson EK, Furlong P, Ascheim DD, Rogy S, Williams P, Marbán L. Repeated intravenous cardiosphere-derived cell therapy in late-stage Duchenne muscular dystrophy (HOPE-2): a multicentre, randomised, double-blind, placebo-controlled, phase 2 trial. Lancet 2022; 399:1049-1058. [PMID: 35279258 DOI: 10.1016/s0140-6736(22)00012-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/28/2021] [Accepted: 12/02/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Cardiosphere-derived cells (CDCs) ameliorate skeletal and cardiac muscle deterioration in experimental models of Duchenne muscular dystrophy. The HOPE-2 trial examined the safety and efficacy of sequential intravenous infusions of human allogeneic CDCs in late-stage Duchenne muscular dystrophy. METHODS In this multicentre, randomised, double-blind, placebo-controlled, phase 2 trial, patients with Duchenne muscular dystrophy, aged 10 years or older with moderate upper limb impairment, were enrolled at seven centres in the USA. Patients were randomly assigned (1:1) using stratified permuted blocks to receive CAP-1002 (1·5 × 108 CDCs) or placebo intravenously every 3 months for a total of four infusions. Clinicians, caregivers, patients, and clinical operations personnel were fully masked to treatment groups. The primary outcome was the change in mid-level elbow Performance of Upper Limb version 1.2 (PUL 1.2) score at 12 months, assessed in the intention-to-treat population. Safety was assessed in all individuals who received an investigational product. This trial is registered with ClinicalTrials.gov, NCT03406780. FINDINGS Between March 1, 2018, and March 31, 2020, 26 male patients with Duchenne muscular dystrophy were enrolled, of whom eight were randomly assigned to the CAP-1002 group and 12 to the placebo group (six were not randomised due to screening failure). In patients who had a post-treatment PUL 1.2 assessment (eight in the CAP-1002 group and 11 in the placebo group), the mean 12-month change from baseline in mid-level elbow PUL1.2 favoured CAP-1002 over placebo (percentile difference 36·2, 95% CI 12·7-59·7; difference of 2·6 points; p=0·014). Infusion-related hypersensitivity reactions without long-term sequelae were observed in three patients, with one patient discontinuing therapy due to a severe allergic reaction. No other major adverse reactions were noted, and no deaths occurred. INTERPRETATION CAP-1002 cell therapy appears to be safe and effective in reducing deterioration of upper limb function in patients with late-stage Duchenne muscular dystrophy. Various measures of cardiac function and structure were also improved in the CAP-1002 group compared with the placebo group. Longer-term extension studies are needed to confirm the therapeutic durability and safety of CAP-1002 beyond 12 months for the treatment of skeletal myopathy and cardiomyopathy in Duchenne muscular dystrophy. FUNDING Capricor Therapeutics.
Collapse
Affiliation(s)
- Craig M McDonald
- University of California Davis School of Medicine, Sacramento, CA, USA.
| | - Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | | | | | | | - Richard S Finkel
- Nemours Children's Hospital, Orlando, FL, USA; St Jude Children's Research Hospital, Memphis, TN, USA
| | - Cuixia Tian
- Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Joanne Janas
- University of Colorado, Children's Hospital Colorado, Denver, CO, USA
| | | | | | - Michael D Taylor
- Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kan N Hor
- Nationwide Children's Hospital, Columbus, OH, USA
| | - Oscar H Mayer
- Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA
| | - Erik K Henricson
- University of California Davis School of Medicine, Sacramento, CA, USA
| | - Pat Furlong
- Parent Project Muscular Dystrophy, Hackensack, NJ, USA
| | | | | | | | | |
Collapse
|
24
|
Marbán E, Liao K. On the cellular origin of cardiosphere-derived cells (CDCs). Basic Res Cardiol 2022; 117:12. [PMID: 35258685 DOI: 10.1007/s00395-022-00914-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 01/31/2023]
Affiliation(s)
- Eduardo Marbán
- Cedars-Sinai Medical Center, Smidt Heart Institute, 127 South San Vicente Boulevard, AHSP A3600, Los Angeles, CA, 90048, USA.
| | - Ke Liao
- Cedars-Sinai Medical Center, Smidt Heart Institute, 127 South San Vicente Boulevard, AHSP A3600, Los Angeles, CA, 90048, USA
| |
Collapse
|
25
|
Bairey Merz CN, Marbán E. Stem Cell Therapy Targets: Repêchage! Circ Res 2022; 130:339-342. [PMID: 35113658 PMCID: PMC8855782 DOI: 10.1161/circresaha.121.320663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- C Noel Bairey Merz
- Barbra Streisand Women's Heart Center, Smidt Heart Institute (C.N.B.M.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Eduardo Marbán
- Smidt Heart Institute (E.M.), Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
26
|
Guo Z, Geng M, Qin L, Hao B, Liao S. Epicardium-Derived Tbx18 + CDCs Transplantation Improve Heart Function in Infarcted Mice. Front Cardiovasc Med 2022; 8:744353. [PMID: 35141286 PMCID: PMC8820322 DOI: 10.3389/fcvm.2021.744353] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022] Open
Abstract
Cardiosphere-derived cells (CDCs) constitute a cardiac stem cell pool, a promising therapeutics in treating myocardial infarction (MI). However, the cell source of CDCs remains unclear. In this study, we isolated CDCs directly from adult mouse heart epicardium named primary epicardium-derived CDCs (pECDCs), which showed a different expression profile compared with primary epicardial cells (pEpiCs). Interestingly, pECDCs highly expressed T-box transcription factor 18 (Tbx18) and showed multipotent differentiation ability in vitro. Human telomerase reverse transcriptase (hTERT) transduction could inhibit aging-induced pECDCs apoptosis and differentiation, thus keeping a better proliferation capacity. Furthermore, immortalized epicardium CDCs (iECDCs) transplantation extensively promote cardiogenesis in the infracted mouse heart. This study demonstrated epicardium-derived CDCs that may derive from Tbx18+ EpiCs, which possess the therapeutic potential to be applied to cardiac repair and regeneration and suggest a new kind of CDCs with identified origination that may be followed in the developing and injured heart.
Collapse
Affiliation(s)
- Zhenglong Guo
- Henan Medical Genetics Institute, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, National Health Commission Key Laboratory of Birth Defects Prevention, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Mengyuan Geng
- School of Medical Laboratory and Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Tianjin Medical University, Tianjin, China
| | - Litao Qin
- Henan Medical Genetics Institute, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, National Health Commission Key Laboratory of Birth Defects Prevention, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| | - Bingtao Hao
- Henan Medical Genetics Institute, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, National Health Commission Key Laboratory of Birth Defects Prevention, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
- School of Basic Medical Sciences, Cancer Research Institute, Southern Medical University, Guangzhou, China
| | - Shixiu Liao
- Henan Medical Genetics Institute, Henan Provincial Key Laboratory of Genetic Diseases and Functional Genomics, National Health Commission Key Laboratory of Birth Defects Prevention, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
27
|
Johnston PV, Raval AN, Henry TD, Traverse JH, Pepine CJ. Dare to dream? Cell-based therapies for heart failure after DREAM-HF: Review and roadmap for future clinical study. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2022; 13:100118. [PMID: 38560073 PMCID: PMC10978179 DOI: 10.1016/j.ahjo.2022.100118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/28/2022] [Indexed: 04/04/2024]
Abstract
Clinical trials of cell-based therapies for heart failure have resulted in significant strides forward in our understanding of the potential the failing heart has for regeneration and repair. Yet, two decades on, the need for novel cell-based therapies for heart failure has never been greater. The DREAM-HF trial, which was presented as a late-breaking trial at the American Heart Association Scientific Sessions 2021 did not meet the primary heart failure outcome, but did show a large, clinically significant reduction in major adverse cardiovascular events (MACE) in patients receiving cells, an effect that was most pronounced in patients with evidence of maladaptive inflammation. These results represent an important step forward in our understanding of how cell-based therapies can exert beneficial effects in patients with heart failure and should serve as a guide for future clinical efforts. In light of the results of DREAM-HF, this review serves to provide an understanding of the current state of cell-based therapies for heart failure, as well as to highlight major knowledge gaps and suggest guiding principles for clinical trials of cell therapy going forward. Using the knowledge gained from DREAM-HF along with the trials that preceded it, the potential for breakthrough cell-based therapies for heart failure in the coming decade is immense.
Collapse
Affiliation(s)
- Peter V. Johnston
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Amish N. Raval
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| | - Timothy D. Henry
- Carl and Edyth Lindner Center for Research at the Christ Hospital, Cincinnati, OH, United States of America
| | - Jay H. Traverse
- Minneapolis Heart Institute Foundation at Abbot Northwestern Hospital, Minneapolis, MN, United States of America
| | - Carl J. Pepine
- Department of Medicine, Division of Cardiovascular Medicine, University of Florida, Gainesville, FL, United States of America
| |
Collapse
|
28
|
Kogan PS, Wirth F, Tomar A, Darr J, Teperino R, Lahm H, Dreßen M, Puluca N, Zhang Z, Neb I, Beck N, Luzius T, de la Osa de la Rosa L, Gärtner K, Hüls C, Zeidler R, Ramanujam D, Engelhardt S, Wenk C, Holdt LM, Mononen M, Sahara M, Cleuziou J, Hörer J, Lange R, Krane M, Doppler SA. Uncovering the molecular identity of cardiosphere-derived cells (CDCs) by single-cell RNA sequencing. Basic Res Cardiol 2022; 117:11. [PMID: 35258704 PMCID: PMC8902493 DOI: 10.1007/s00395-022-00913-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 01/31/2023]
Abstract
Cardiosphere-derived cells (CDCs) generated from human cardiac biopsies have been shown to have disease-modifying bioactivity in clinical trials. Paradoxically, CDCs' cellular origin in the heart remains elusive. We studied the molecular identity of CDCs using single-cell RNA sequencing (sc-RNAseq) in comparison to cardiac non-myocyte and non-hematopoietic cells (cardiac fibroblasts/CFs, smooth muscle cells/SMCs and endothelial cells/ECs). We identified CDCs as a distinct and mitochondria-rich cell type that shared biological similarities with non-myocyte cells but not with cardiac progenitor cells derived from human-induced pluripotent stem cells. CXCL6 emerged as a new specific marker for CDCs. By analysis of sc-RNAseq data from human right atrial biopsies in comparison with CDCs we uncovered transcriptomic similarities between CDCs and CFs. By direct comparison of infant and adult CDC sc-RNAseq data, infant CDCs revealed GO-terms associated with cardiac development. To analyze the beneficial effects of CDCs (pro-angiogenic, anti-fibrotic, anti-apoptotic), we performed functional in vitro assays with CDC-derived extracellular vesicles (EVs). CDC EVs augmented in vitro angiogenesis and did not stimulate scarring. They also reduced the expression of pro-apoptotic Bax in NRCMs. In conclusion, CDCs were disclosed as mitochondria-rich cells with unique properties but also with similarities to right atrial CFs. CDCs displayed highly proliferative, secretory and immunomodulatory properties, characteristics that can also be found in activated or inflammatory cell types. By special culture conditions, CDCs earn some bioactivities, including angiogenic potential, which might modify disease in certain disorders.
Collapse
Affiliation(s)
- Palgit-S. Kogan
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Felix Wirth
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Archana Tomar
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Jonatan Darr
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Raffaele Teperino
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany ,German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Harald Lahm
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Martina Dreßen
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Nazan Puluca
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Zhong Zhang
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Irina Neb
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Nicole Beck
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Tatjana Luzius
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Luis de la Osa de la Rosa
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| | - Kathrin Gärtner
- Research Unit Gene Vectors, Helmholtz Center Munich German Research Center for Environmental Health, Munich, Germany
| | - Corinna Hüls
- Research Unit Gene Vectors, Helmholtz Center Munich German Research Center for Environmental Health, Munich, Germany
| | - Reinhard Zeidler
- Research Unit Gene Vectors, Helmholtz Center Munich German Research Center for Environmental Health, Munich, Germany ,Department of Otorhinolaryngology, Klinikum der Universität (KUM), Munich, Germany
| | - Deepak Ramanujam
- DZHK (German Center for Cardiovascular Research)-Partner Site Munich Heart Alliance, Biedersteiner Straße 29, 80802 Munich, Germany ,Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, 80802 Munich, Germany
| | - Stefan Engelhardt
- DZHK (German Center for Cardiovascular Research)-Partner Site Munich Heart Alliance, Biedersteiner Straße 29, 80802 Munich, Germany ,Institute of Pharmacology and Toxicology, Technische Universität München, Biedersteiner Str. 29, 80802 Munich, Germany
| | - Catharina Wenk
- Institute of Laboratory Medicine, University Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Lesca M. Holdt
- Institute of Laboratory Medicine, University Hospital, Ludwig Maximilians University Munich, Munich, Germany
| | - Mimmi Mononen
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Makoto Sahara
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden ,Department of Surgery, Yale University School of Medicine, CN06510 New Haven, CT USA
| | - Julie Cleuziou
- School of Medicine and Health, Department of Pediatric and Congenital Heart Surgery, Institute Insure, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
| | - Jürgen Hörer
- School of Medicine and Health, Department of Pediatric and Congenital Heart Surgery, Technical University of Munich, German Heart Center Munich, Lazarettstraße 36, 80636 Munich, Germany ,Division of Congenital and Pediatric Heart Surgery, University Hospital of Munich, Ludwig-Maximilians-Universität, Munich, Germany
| | - Rüdiger Lange
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany ,DZHK (German Center for Cardiovascular Research)-Partner Site Munich Heart Alliance, Biedersteiner Straße 29, 80802 Munich, Germany
| | - Markus Krane
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany ,DZHK (German Center for Cardiovascular Research)-Partner Site Munich Heart Alliance, Biedersteiner Straße 29, 80802 Munich, Germany ,Division of Cardiac Surgery, Department of Surgery, Yale University School of Medicine, New Haven, CT USA
| | - Stefanie A. Doppler
- School of Medicine and Health, Department of Cardiovascular Surgery, Institute Insure, Technical University of Munich, German Heart Center Munich, Lazarettstrasse 36, 80636 Munich, Germany
| |
Collapse
|
29
|
Grigoratos C, Aimo A, Barison A, Castiglione V, Todiere G, Ricci G, Siciliano G, Emdin M. Cardiac magnetic resonance in patients with muscular dystrophies. Eur J Prev Cardiol 2021; 28:1526-1535. [PMID: 32418485 DOI: 10.1177/2047487320923052] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/10/2020] [Indexed: 01/15/2023]
Abstract
Muscular dystrophies are inherited disorders sharing similar clinical features and dystrophic changes on muscle biopsy. Duchenne muscular dystrophy is the most common inherited muscle disease of childhood, and Becker muscular dystrophy is a milder allelic variant with a slightly lower prevalence. Myotonic dystrophy is the most frequent form in adults. Cardiac magnetic resonance is the gold standard technique for the quantification of cardiac chamber volumes and function, and also enables a characterisation of myocardial tissue. Most cardiac magnetic resonance studies in the setting of muscular dystrophy were carried out at single centres, evaluated small numbers of patients and used widely heterogeneous protocols. Even more importantly, those studies analysed more or less extensively the patterns of cardiac involvement, but usually did not try to establish the added value of cardiac magnetic resonance to standard echocardiography, the evolution of cardiac disease over time and the prognostic significance of cardiac magnetic resonance findings. As a result, the large and heterogeneous amount of information on cardiac involvement in muscular dystrophies cannot easily be translated into recommendations on the optimal use of cardiac magnetic resonance. In this review, whose targets are cardiologists and neurologists who manage patients with muscular dystrophy, we try to summarise cardiac magnetic resonance findings in patients with muscular dystrophy, and the results of studies evaluating the role of cardiac magnetic resonance as a tool for diagnosis, risk stratification and follow-up. Finally, we provide some practical recommendations about the need and timing of cardiac magnetic resonance examination for the management of patients with muscular dystrophy.
Collapse
Affiliation(s)
| | - Alberto Aimo
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Italy
| | - Andrea Barison
- Fondazione Toscana Gabriele Monasterio, Italy
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Italy
| | | | | | - Giulia Ricci
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Michele Emdin
- Fondazione Toscana Gabriele Monasterio, Italy
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Italy
| |
Collapse
|
30
|
Zhang J, Bolli R, Garry DJ, Marbán E, Menasché P, Zimmermann WH, Kamp TJ, Wu JC, Dzau VJ. Basic and Translational Research in Cardiac Repair and Regeneration: JACC State-of-the-Art Review. J Am Coll Cardiol 2021; 78:2092-2105. [PMID: 34794691 PMCID: PMC9116459 DOI: 10.1016/j.jacc.2021.09.019] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 12/25/2022]
Abstract
This paper aims to provide an important update on the recent preclinical and clinical trials using cell therapy strategies and engineered heart tissues for the treatment of postinfarction left ventricular remodeling and heart failure. In addition to the authors’ own works and opinions on the roadblocks of the field, they discuss novel approaches for cardiac remuscularization via the activation of proliferative mechanisms in resident cardiomyocytes or direct reprogramming of somatic cells into cardiomyocytes. This paper’s main mindset is to present current and future strategies in light of their implications for the design of future patient trials with the ultimate objective of facilitating the translation of discoveries in regenerative myocardial therapies to the clinic.
Collapse
Affiliation(s)
- Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine, School of Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, USA.
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, Louisville, Kentucky, USA
| | - Daniel J Garry
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles California, USA
| | - Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, University of Paris, PARCC, INSERM, F-75015, Paris, France
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, and DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Timothy J Kamp
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Victor J Dzau
- Mandel Center for Hypertension Research, Duke Cardiovascular Center, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
31
|
Affiliation(s)
- Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
32
|
Lin YN, Mesquita T, Sanchez L, Chen YH, Liu W, Li C, Rogers R, Wang Y, Li X, Wu D, Zhang R, Ibrahim A, Marbán E, Cingolani E. Extracellular vesicles from immortalized cardiosphere-derived cells attenuate arrhythmogenic cardiomyopathy in desmoglein-2 mutant mice. Eur Heart J 2021; 42:3558-3571. [PMID: 34345905 PMCID: PMC8442111 DOI: 10.1093/eurheartj/ehab419] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 04/28/2021] [Accepted: 06/25/2021] [Indexed: 01/19/2023] Open
Abstract
AIMS Arrhythmogenic cardiomyopathy (ACM) is characterized by progressive loss of cardiomyocytes, and fibrofatty tissue replacement. Extracellular vesicles (EVs) secreted by cardiosphere-derived cells, immortalized, and engineered to express high levels of β-catenin, exert anti-inflammatory, and anti-fibrotic effects. The aim of the current study was to assess efficacy of EVs in an ACM murine model. METHODS AND RESULTS Four-week-old homozygous knock-in mutant desmoglein-2 (Dsg2mt/mt) were randomized to receive weekly EVs or vehicle for 4 weeks. After 4 weeks, DSG2mt/mt mice receiving EVs showed improved biventricular function (left, P < 0.0001; right, P = 0.0037) and less left ventricular dilation (P < 0.0179). Electrocardiography revealed abbreviated QRS duration (P = 0.0003) and QTc interval (P = 0.0006) in EV-treated DSG2mt/mt mice. Further electrophysiology testing in the EV group showed decreased burden (P = 0.0042) and inducibility of ventricular arrhythmias (P = 0.0037). Optical mapping demonstrated accelerated repolarization (P = 0.0290) and faster conduction (P = 0.0274) in Dsg2mt/mt mice receiving EVs. DSG2mt/mt hearts exhibited reduced fibrosis, less cell death, and preserved connexin 43 expression after EV treatment. Hearts of Dsg2mt/mt mice expressed markedly increased levels of inflammatory cytokines that were, in part, attenuated by EV therapy. The pan-inflammatory transcription factor nuclear factor-κB (NF-κB), the inflammasome sensor NLRP3, and the macrophage marker CD68 were all reduced in EV-treated animals. Blocking EV hsa-miR-4488 in vitro and in vivo reactivates NF-κB and blunts the beneficial effects of EVs. CONCLUSIONS Extracellular vesicle treatment improved cardiac function, reduced cardiac inflammation, and suppressed arrhythmogenesis in ACM. Further studies are needed prior to translating the present findings to human forms of this heterogenous disease.
Collapse
Affiliation(s)
- Yen-Nien Lin
- Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Boulevard, Los Angeles, CA 90048, USA
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University and Hospital, 2, Yu-Der Road, North District, Taichung 40447, Taiwan; and
| | - Thassio Mesquita
- Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Boulevard, Los Angeles, CA 90048, USA
| | - Lizbeth Sanchez
- Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Boulevard, Los Angeles, CA 90048, USA
| | - Yin-Huei Chen
- Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Boulevard, Los Angeles, CA 90048, USA
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University and Hospital, 2, Yu-Der Road, North District, Taichung 40447, Taiwan; and
| | - Weixin Liu
- Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Boulevard, Los Angeles, CA 90048, USA
| | - Chang Li
- Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Boulevard, Los Angeles, CA 90048, USA
| | - Russell Rogers
- Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Boulevard, Los Angeles, CA 90048, USA
| | - Yizhou Wang
- Genomics Core, Cedars-Sinai Medical Center, 8700 Beverly Blvd. Los Angeles, CA 90048, USA
| | - Xinling Li
- Genomics Core, Cedars-Sinai Medical Center, 8700 Beverly Blvd. Los Angeles, CA 90048, USA
| | - Di Wu
- Genomics Core, Cedars-Sinai Medical Center, 8700 Beverly Blvd. Los Angeles, CA 90048, USA
| | - Rui Zhang
- Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Boulevard, Los Angeles, CA 90048, USA
| | - Ahmed Ibrahim
- Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Boulevard, Los Angeles, CA 90048, USA
| | - Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Boulevard, Los Angeles, CA 90048, USA
| | - Eugenio Cingolani
- Smidt Heart Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Boulevard, Los Angeles, CA 90048, USA
| |
Collapse
|
33
|
Davoli GBDQ, Cardoso J, Silva GC, Moreira RDFC, Mattiello-Sverzut AC. Instruments to assess upper-limb function in children and adolescents with neuromuscular diseases: a systematic review. Dev Med Child Neurol 2021; 63:1030-1037. [PMID: 33834485 DOI: 10.1111/dmcn.14887] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/06/2021] [Indexed: 11/29/2022]
Abstract
AIM To synthesize clinical and scientific evidence regarding the instruments available to assess upper-limb function in paediatric patients with neuromuscular disease (NMD). METHOD This systematic review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses and COnsensus-based Standards for the selection of health Measurement INstruments (COSMIN) guidelines (Prospective Registry of Systematic Reviews no. CRD42020140343). Two independent reviewers searched the PubMed/MEDLINE, LILACS, Embase, and Scopus databases. Inclusion criteria were cross-sectional or longitudinal studies or randomized controlled trials that used scales or questionnaires to assess upper-limb function in paediatric patients with NMDs. The COSMIN Risk of Bias checklist and criteria for good measurement properties were applied to assess the methodological quality of the instruments. RESULTS In total, 34 articles and 12 instruments were included. The Brooke Upper Extremity (n=16) and Performance of Upper Limb (PUL) (n=12) instruments were the most used tools. The PUL and Duchenne muscular dystrophy (DMD) Upper Limb patient-reported outcome measures (PROMs) tested more measurement properties and provided higher methodological quality scores for patients with DMD. Likewise, the Revised Upper Limb Module (RULM) was the most suitable instrument for patients with spinal muscular atrophy. No instrument has been devised to assess upper-limb function in patients with Charcot-Marie-Tooth disease and no other disease-specific instruments were found. INTERPRETATION The PUL, DMD Upper Limb PROM, and RULM are the most suitable instruments to assess upper-limb function in the two most prevalent paediatric NMDs. The identified gaps and methodological flaws of the available instruments indicate a need to develop high-quality instruments to assess other types of paediatric NMDs. What this paper adds The most suitable observer-rater instrument to assess upper-limb function in Duchenne muscular dystrophy (DMD) is the Performance of Upper Limb. The most suitable observer-rater instrument to assess upper-limb function in spinal muscular atrophy is the Revised Upper Limb Module. The DMD Upper Limb patient-reported outcome measure is recommended to assess the upper-limb performance of patients with DMD. Literature gaps and methodological flaws indicate the need to develop high-quality instruments to assess other types of paediatric neuromuscular disease.
Collapse
Affiliation(s)
| | - Juliana Cardoso
- Department of Health Science, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Giovanna Constantin Silva
- Department of Health Science, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | | |
Collapse
|
34
|
Leiva-Cepas F, Montaño Martínez A, López-López I. [Update on Duchenne muscular dystrophy]. Semergen 2021; 47:472-481. [PMID: 34417099 DOI: 10.1016/j.semerg.2021.06.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 11/18/2022]
Abstract
Duchenne muscular dystrophy, DMD*(ICD-9-C: 359.1; ICD-10-ES: G71.01, ORPHA: 98896) is a dystrophic type, autosomal recessive myopathy linked to the X chromosome, low incidence 1/3300, with full penetrance and multi-organ involvement (neuro-muscular, respiratory, digestive and metabolic). It has great clinical variability. Symptoms begin in pediatric age (mobility limitation and early respiratory complications). Respiratory complications reduce the life expectancy of those affected. There is no treatment that modifies its evolution, although corticosteroids and new gene therapies are increasing the half-life of this disease. The role of the Primary Care Physician (PCP) is decisive in the monitoring and control of the complications of DMD, either coordinating the different specialties involved in it.
Collapse
Affiliation(s)
- F Leiva-Cepas
- Departamento de Ciencias Morfológicas, Sección de Histología, Facultad de Medicina y Enfermería, Universidad de Córdoba, Córdoba, España; Unidad de Gestión Clínica de Anatomía Patológica, Hospital Universitario Reina Sofía, Córdoba, España; Grupo de Investigación en Regeneración Muscular (REGMUS), Código PAIDI: CTS-285, Córdoba, España; Grupo GC-12 de Investigación Epidemiológica en Atención Primaria, Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, España.
| | - A Montaño Martínez
- Hospital de Montilla, Empresa Pública Hospital Alto Guadalquivir, Montilla, Córdoba, España
| | - I López-López
- Servicio de Nefrología. Hospital Universitario Reina Sofía, Córdoba, España
| |
Collapse
|
35
|
Clinical outcome assessments in Duchenne muscular dystrophy and spinal muscular atrophy: past, present and future. Neuromuscul Disord 2021; 31:1028-1037. [PMID: 34412961 DOI: 10.1016/j.nmd.2021.07.015] [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: 06/30/2021] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 11/23/2022]
Abstract
Scores and scales used in pediatric motor development for neuromuscular disorders have evolved greatly since the beginning of their development. In this review we provide a brief history of scales used in pediatric patients with neuromuscular disorders and an update regarding the advancement of the scales commonly used in patients with spinal muscular atrophy and Duchenne muscular dystrophy. We focus on the collaborative effort that has led to the development of outcomes and speak to the possible future of Clinical Outcome Assessments.
Collapse
|
36
|
Siemionow M, Langa P, Harasymczuk M, Cwykiel J, Sielewicz M, Smieszek J, Heydemann A. Human dystrophin expressing chimeric (DEC) cell therapy ameliorates cardiac, respiratory, and skeletal muscle's function in Duchenne muscular dystrophy. Stem Cells Transl Med 2021; 10:1406-1418. [PMID: 34291884 PMCID: PMC8459641 DOI: 10.1002/sctm.21-0054] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/11/2021] [Accepted: 07/07/2021] [Indexed: 12/28/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive and lethal disease, caused by X‐linked mutations of the dystrophin encoding gene. The lack of dystrophin leads to muscle weakness, degeneration, fibrosis, and progressive loss of skeletal, cardiac, and respiratory muscle function resulting in premature death due to the cardiac and respiratory failure. There is no cure for DMD and current therapies neither cure nor arrest disease progression. Thus, there is an urgent need to develop new approaches and safer therapies for DMD patients. We have previously reported functional improvements which correlated with increased dystrophin expression following transplantation of dystrophin expressing chimeric (DEC) cells of myoblast origin to the mdx mouse models of DMD. In this study, we demonstrated that systemic‐intraosseous transplantation of DEC human cells derived from myoblasts of normal and DMD‐affected donors, increased dystrophin expression in cardiac, respiratory, and skeletal muscles of the mdx/scid mouse model of DMD. DEC transplant correlated with preservation of ejection fraction and fractional shortening on echocardiography, improved respiratory function on plethysmography, and improved strength and function of the limb skeletal muscles. Enhanced function was associated with improved muscle histopathology, revealing reduced mdx pathology, fibrosis, decreased inflammation, and preserved muscle morphology and architecture. Our findings confirm that DECs generate a systemic protective effect in DMD‐affected target organs. Therefore, DECs represents a novel therapeutic approach with the potential to preserve or enhance multiorgan function of the skeletal, cardiac, and respiratory muscles critical for the well‐being of DMD patients.
Collapse
Affiliation(s)
- Maria Siemionow
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Paulina Langa
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Michal Harasymczuk
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Joanna Cwykiel
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Magdalena Sielewicz
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Jaroslaw Smieszek
- Department of Orthopaedics, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Ahlke Heydemann
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, Illinois, USA.,Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
37
|
Akhmerov A, Rogers R, de Couto G, Valle J, Li L, Ibrahim A, Sanchez L, Zhang R, Lin YN, Liu W, Marbán E. Regulatory T cell activation, proliferation, and reprogramming induced by extracellular vesicles. J Heart Lung Transplant 2021; 40:1387-1395. [PMID: 34281778 DOI: 10.1016/j.healun.2021.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 06/04/2021] [Accepted: 06/07/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) from heart stromal/progenitor cells modulate innate immunity, with salutary effects in a variety of cardiac disease models. Little is known, however, about the effects of these EVs on adaptive immunity. METHODS Ex vivo differentiation of naïve CD4+ T cells was conducted to assess the effect of EVs on cytokine production and proliferation of Th1, Th2, Th17, and regulatory T (Treg) cells. These effects were further tested in vivo using the experimental autoimmune myocarditis (EAM) model. RESULTS Using differentiated CD4+ T cells, we show that EVs secreted by human-derived heart stromal/progenitor cells selectively influence the phenotype, activity, and proliferation of regulatory T (Treg) cells. Exposure of Treg cells to EVs results in faster proliferation, augmented production of IL-10, and polarization toward an intermediate FOXP3+RORγt+ phenotype. In experimental autoimmune myocarditis, EVs attenuate cardiac inflammation and functional decline, in association with increased numbers of splenic IL10+-Treg cells. CONCLUSIONS T cell modulation by EVs represents a novel therapeutic approach to inflammation, harnessing endogenous immunosuppressive mechanisms that may be applied in solid organ transplantation, graft-versus-host disease, and autoimmune disorders.
Collapse
Affiliation(s)
| | - Russell Rogers
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Geoffrey de Couto
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jackelyn Valle
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Liang Li
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Ahmed Ibrahim
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Lizbeth Sanchez
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Rui Zhang
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Yen-Nien Lin
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Weixin Liu
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Eduardo Marbán
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
38
|
Hirai K, Ousaka D, Fukushima Y, Kondo M, Eitoku T, Shigemitsu Y, Hara M, Baba K, Iwasaki T, Kasahara S, Ohtsuki S, Oh H. Cardiosphere-derived exosomal microRNAs for myocardial repair in pediatric dilated cardiomyopathy. Sci Transl Med 2021; 12:12/573/eabb3336. [PMID: 33298561 DOI: 10.1126/scitranslmed.abb3336] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 10/15/2020] [Indexed: 12/31/2022]
Abstract
Although cardiosphere-derived cells (CDCs) improve cardiac function and outcomes in patients with single ventricle physiology, little is known about their safety and therapeutic benefit in children with dilated cardiomyopathy (DCM). We aimed to determine the safety and efficacy of CDCs in a porcine model of DCM and translate the preclinical results into this patient population. A swine model of DCM using intracoronary injection of microspheres created cardiac dysfunction. Forty pigs were randomized as preclinical validation of the delivery method and CDC doses, and CDC-secreted exosome (CDCex)-mediated cardiac repair was analyzed. A phase 1 safety cohort enrolled five pediatric patients with DCM and reduced ejection fraction to receive CDC infusion. The primary endpoint was to assess safety, and the secondary outcome measure was change in cardiac function. Improved cardiac function and reduced myocardial fibrosis were noted in animals treated with CDCs compared with placebo. These functional benefits were mediated via CDCex that were highly enriched with proangiogenic and cardioprotective microRNAs (miRNAs), whereas isolated CDCex did not recapitulate these reparative effects. One-year follow-up of safety lead-in stage was completed with favorable profile and preliminary efficacy outcomes. Increased CDCex-derived miR-146a-5p expression was associated with the reduction in myocardial fibrosis via suppression of proinflammatory cytokines and transcripts. Collectively, intracoronary CDC administration is safe and improves cardiac function through CDCex in a porcine model of DCM. The safety lead-in results in patients provide a translational framework for further studies of randomized trials and CDCex-derived miRNAs as potential paracrine mediators underlying this therapeutic strategy.
Collapse
Affiliation(s)
- Kenta Hirai
- Department of Pediatric Cardiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Daiki Ousaka
- Department of Cardiovascular Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Yosuke Fukushima
- Department of Pediatric Cardiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Maiko Kondo
- Department of Pediatric Cardiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Takahiro Eitoku
- Department of Pediatric Cardiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Yusuke Shigemitsu
- Department of Pediatric Cardiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Mayuko Hara
- Department of Pediatric Cardiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Kenji Baba
- Department of Pediatric Cardiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Tatsuo Iwasaki
- Department of Anesthesiology and Resuscitology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Shingo Kasahara
- Department of Cardiovascular Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Shinichi Ohtsuki
- Department of Pediatric Cardiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Hidemasa Oh
- Department of Regenerative Medicine, Center for Innovative Clinical Medicine, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan.
| |
Collapse
|
39
|
Sandonà M, Di Pietro L, Esposito F, Ventura A, Silini AR, Parolini O, Saccone V. Mesenchymal Stromal Cells and Their Secretome: New Therapeutic Perspectives for Skeletal Muscle Regeneration. Front Bioeng Biotechnol 2021; 9:652970. [PMID: 34095095 PMCID: PMC8172230 DOI: 10.3389/fbioe.2021.652970] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent cells found in different tissues: bone marrow, peripheral blood, adipose tissues, skeletal muscle, perinatal tissues, and dental pulp. MSCs are able to self-renew and to differentiate into multiple lineages, and they have been extensively used for cell therapy mostly owing to their anti-fibrotic and immunoregulatory properties that have been suggested to be at the basis for their regenerative capability. MSCs exert their effects by releasing a variety of biologically active molecules such as growth factors, chemokines, and cytokines, either as soluble proteins or enclosed in extracellular vesicles (EVs). Analyses of MSC-derived secretome and in particular studies on EVs are attracting great attention from a medical point of view due to their ability to mimic all the therapeutic effects produced by the MSCs (i.e., endogenous tissue repair and regulation of the immune system). MSC-EVs could be advantageous compared with the parental cells because of their specific cargo containing mRNAs, miRNAs, and proteins that can be biologically transferred to recipient cells. MSC-EV storage, transfer, and production are easier; and their administration is also safer than MSC therapy. The skeletal muscle is a very adaptive tissue, but its regenerative potential is altered during acute and chronic conditions. Recent works demonstrate that both MSCs and their secretome are able to help myofiber regeneration enhancing myogenesis and, interestingly, can be manipulated as a novel strategy for therapeutic interventions in muscular diseases like muscular dystrophies or atrophy. In particular, MSC-EVs represent promising candidates for cell free-based muscle regeneration. In this review, we aim to give a complete picture of the therapeutic properties and advantages of MSCs and their products (MSC-derived EVs and secreted factors) relevant for skeletal muscle regeneration in main muscular diseases.
Collapse
Affiliation(s)
- Martina Sandonà
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione Santa Lucia, Rome, Italy
| | - Lorena Di Pietro
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Federica Esposito
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione Santa Lucia, Rome, Italy
| | - Alessia Ventura
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione Santa Lucia, Rome, Italy
| | - Antonietta Rosa Silini
- Centro di Ricerca "E. Menni", Fondazione Poliambulanza - Istituto Ospedaliero, Brescia, Italy
| | - Ornella Parolini
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Valentina Saccone
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione Santa Lucia, Rome, Italy.,Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
40
|
Proteomic and Glyco(proteo)mic tools in the profiling of cardiac progenitors and pluripotent stem cell derived cardiomyocytes: Accelerating translation into therapy. Biotechnol Adv 2021; 49:107755. [PMID: 33895330 DOI: 10.1016/j.biotechadv.2021.107755] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 03/15/2021] [Accepted: 04/18/2021] [Indexed: 12/14/2022]
Abstract
Research in stem cells paved the way to an enormous amount of knowledge, increasing expectations on cardio regenerative therapeutic approaches in clinic. While the first generation of clinical trials using cell-based therapies in the heart were performed with bone marrow and adipose tissue derived mesenchymal stem cells, second generation cell therapies moved towards the use of cardiac-committed cell populations, including cardiac progenitor cells and pluripotent stem cell derived cardiomyocytes. Despite all these progresses, translating the aptitudes of R&D and pre-clinical data into effective clinical treatments is still highly challenging, partially due to the demanding regulatory and safety concerns but also because of the lack of knowledge on the regenerative mechanisms of action of these therapeutic products. Thus, the need of analytical methodologies that enable a complete characterization of such complex products and a deep understanding of their therapeutic effects, at the cell and molecular level, is imperative to overcome the hurdles of these advanced therapies. Omics technologies, such as proteomics and glyco(proteo)mics workflows based on state of the art mass-spectrometry, have prompted some major breakthroughs, providing novel data on cell biology and a detailed assessment of cell based-products applied in cardiac regeneration strategies. These advanced 'omics approaches, focused on the profiling of protein and glycan signatures are excelling the identification and characterization of cell populations under study, namely unveiling pluripotency and differentiation markers, as well as paracrine mechanisms and signaling cascades involved in cardiac repair. The leading knowledge generated is supporting a more rational therapy design and the rethinking of challenges in Advanced Therapy Medicinal Products development. Herein, we review the most recent methodologies used in the fields of proteomics, glycoproteomics and glycomics and discuss their impact on the study of cardiac progenitor cells and pluripotent stem cell derived cardiomyocytes biology. How these discoveries will impact the speed up of novel therapies for cardiovascular diseases is also addressed.
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW Duchenne muscular dystrophy is one of many neuromuscular disorders, but it frequently causes severe disability early in life and early death. Cardiac involvement is an important cause of morbidity and mortality. RECENT FINDINGS Heart disease in Duchenne muscular dystrophy can include a cardiomyopathy leading to end-stage heart failure along with associated supraventricular and ventricular arrhythmias. This article reviews the diagnosis and treatment of heart disease in Duchenne muscular dystrophy as well as emerging therapies.
Collapse
Affiliation(s)
- Jeffrey A Shih
- University of Massachusetts, 55 Lake Avenue North, Worcester, MA, 01532, USA.
| | - Alejandro Folch
- University of Massachusetts, 55 Lake Avenue North, Worcester, MA, 01532, USA
| | - Brenda L Wong
- University of Massachusetts, 55 Lake Avenue North, Worcester, MA, 01532, USA
| |
Collapse
|
42
|
Spurney CF, Ascheim D, Charnas L, Cripe L, Hor K, King N, Kinnett K, McNally EM, Sauer JM, Sweeney L, Villa C, Markham LW. Current state of cardiac troponin testing in Duchenne muscular dystrophy cardiomyopathy: review and recommendations from the Parent Project Muscular Dystrophy expert panel. Open Heart 2021; 8:openhrt-2021-001592. [PMID: 33762424 PMCID: PMC7993361 DOI: 10.1136/openhrt-2021-001592] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/17/2021] [Accepted: 03/05/2021] [Indexed: 01/06/2023] Open
Abstract
Cardiac disease is now the leading cause of death in Duchenne muscular dystrophy (DMD). Clinical evaluations over time have demonstrated asymptomatic cardiac troponin elevations and acute elevations are associated with symptoms and cardiac dysfunction in DMD. Clinicians require a better understanding of the relationship of symptoms, troponin levels and progression of cardiac disease in DMD. As clinical trials begin to assess novel cardiac therapeutics in DMD, troponin levels in DMD are important for safety monitoring and outcome measures. The Parent Project Muscular Dystrophy convened an expert panel of cardiologists, scientists, and regulatory and industry specialists on 16 December 2019 in Silver Spring, Maryland and reviewed published and unpublished data from their institutions. The panel recommended retrospective troponin data analyses, prospective longitudinal troponin collection using high-sensitivity cardiac troponin I assays, inclusion of troponin in future clinical trial outcomes and future development of clinical guidelines for monitoring and treating troponin elevations in DMD.
Collapse
Affiliation(s)
- Christopher F Spurney
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia, USA
| | | | - Lawrence Charnas
- Clinical Research Rare Neurology Disease, Pfizer, Cambridge, Massachusetts, USA
| | - Linda Cripe
- The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Kan Hor
- The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA
| | | | - Kathi Kinnett
- Parent Project Muscular Dystrophy, Hackensack, New Jersey, USA
| | | | | | - Lee Sweeney
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, USA
| | - Chet Villa
- Cardiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Larry W Markham
- Pediatrics, Division of Pediatric Cardiology, Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana, USA
| |
Collapse
|
43
|
Abstract
Increased understanding of disease pathophysiology and advances in gene therapies and drug technologies are revolutionizing treatment of muscular dystrophies and motor neuron disorders (MNDs). New drugs have been approved for Duchenne muscular dystrophy, spinal muscular atrophy, and amyotrophic lateral sclerosis. For other diseases, new targets have been identified, and new therapies are in clinical trials. The impact of such therapies will be fully understood only in the next decades. Cost burden and accessibility are major challenges in the wide application of new drugs. This article reviews advances in gene therapies, newly approved drugs, and therapeutic promises in muscular dystrophies and MNDs.
Collapse
|
44
|
Povsic TJ, Gersh BJ. Stem Cells in Cardiovascular Diseases: 30,000-Foot View. Cells 2021; 10:cells10030600. [PMID: 33803227 PMCID: PMC8001267 DOI: 10.3390/cells10030600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 12/15/2022] Open
Abstract
Stem cell and regenerative approaches that might rejuvenate the heart have immense intuitive appeal for the public and scientific communities. Hopes were fueled by initial findings from preclinical models that suggested that easily obtained bone marrow cells might have significant reparative capabilities; however, after initial encouraging pre-clinical and early clinical findings, the realities of clinical development have placed a damper on the field. Clinical trials were often designed to detect exceptionally large treatment effects with modest patient numbers with subsequent disappointing results. First generation approaches were likely overly simplistic and relied on a relatively primitive understanding of regenerative mechanisms and capabilities. Nonetheless, the field continues to move forward and novel cell derivatives, platforms, and cell/device combinations, coupled with a better understanding of the mechanisms that lead to regenerative capabilities in more primitive models and modifications in clinical trial design suggest a brighter future.
Collapse
Affiliation(s)
- Thomas J. Povsic
- Department of Medicine, and Duke Clinical Research Institute, Duke University, Durham, NC 27705, USA
- Correspondence:
| | - Bernard J. Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA;
| |
Collapse
|
45
|
Madeddu P. Cell therapy for the treatment of heart disease: Renovation work on the broken heart is still in progress. Free Radic Biol Med 2021; 164:206-222. [PMID: 33421587 DOI: 10.1016/j.freeradbiomed.2020.12.444] [Citation(s) in RCA: 4] [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: 11/12/2020] [Revised: 11/26/2020] [Accepted: 12/29/2020] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease (CVD) continues to be the number one killer in the aging population. Heart failure (HF) is also an important cause of morbidity and mortality in patients with congenital heart disease (CHD). Novel therapeutic approaches that could restore stable heart function are much needed in both paediatric and adult patients. Regenerative medicine holds promises to provide definitive solutions for correction of congenital and acquired cardiac defects. In this review article, we recap some important aspects of cardiovascular cell therapy. First, we report quantifiable data regarding the scientific advancements in the field and how this has been translated into tangible outcomes according clinical studies and related meta-analyses. We then comment on emerging trends and technologies, such as the use of second-generation cell products, including pericyte-like vascular progenitors, and reprogramming of cells by different approaches including modulation of oxidative stress. The more affordable and feasible strategy of repurposing clinically available drugs to awaken the intrinsic healing potential of the heart will be discussed in the light of current social, financial, and ethical context. Cell therapy remains a work in progress field. Uncertainty in the ability of the experts and policy makers to solve urgent medical problems is growing in a world that is significantly influenced by them. This is particularly true in the field of regenerative medicine, due to great public expectations, polarization of leadership and funding, and insufficient translational vision. Cardiovascular regenerative medicine should be contextualized in a holistic program with defined priorities to allow a complete realization. Reshaping the notion of medical expertise is fundamental to fill the current gap in translation.
Collapse
Affiliation(s)
- Paolo Madeddu
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Upper Maudlin Street, BS28HW, Bristol, United Kingdom.
| |
Collapse
|
46
|
Abstract
Duchenne muscular dystrophy is a severe, progressive, muscle-wasting disease that leads to difficulties with movement and, eventually, to the need for assisted ventilation and premature death. The disease is caused by mutations in DMD (encoding dystrophin) that abolish the production of dystrophin in muscle. Muscles without dystrophin are more sensitive to damage, resulting in progressive loss of muscle tissue and function, in addition to cardiomyopathy. Recent studies have greatly deepened our understanding of the primary and secondary pathogenetic mechanisms. Guidelines for the multidisciplinary care for Duchenne muscular dystrophy that address obtaining a genetic diagnosis and managing the various aspects of the disease have been established. In addition, a number of therapies that aim to restore the missing dystrophin protein or address secondary pathology have received regulatory approval and many others are in clinical development.
Collapse
Affiliation(s)
- Dongsheng Duan
- Department of Molecular Microbiology and Immunology and Department of Neurology, School of Medicine; Department of Biomedical Sciences, College of Veterinary Medicine; Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO, USA
| | - Nathalie Goemans
- Department of Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | | | - Eugenio Mercuri
- Centro Clinico Nemo, Policlinico Gemelli, Rome, Italy
- Peadiatric Neurology, Catholic University, Rome, Italy
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
47
|
Fortunato F, Rossi R, Falzarano MS, Ferlini A. Innovative Therapeutic Approaches for Duchenne Muscular Dystrophy. J Clin Med 2021; 10:jcm10040820. [PMID: 33671409 PMCID: PMC7922390 DOI: 10.3390/jcm10040820] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common childhood muscular dystrophy affecting ~1:5000 live male births. Following the identification of pathogenic variations in the dystrophin gene in 1986, the underlining genotype/phenotype correlations emerged and the role of the dystrophin protein was elucidated in skeletal, smooth, and cardiac muscles, as well as in the brain. When the dystrophin protein is absent or quantitatively or qualitatively modified, the muscle cannot sustain the stress of repeated contractions. Dystrophin acts as a bridging and anchoring protein between the sarcomere and the sarcolemma, and its absence or reduction leads to severe muscle damage that eventually cannot be repaired, with its ultimate substitution by connective tissue and fat. The advances of an understanding of the molecular pathways affected in DMD have led to the development of many therapeutic strategies that tackle different aspects of disease etiopathogenesis, which have recently led to the first successful approved orphan drugs for this condition. The therapeutic advances in this field have progressed exponentially, with second-generation drugs now entering in clinical trials as gene therapy, potentially providing a further effective approach to the condition.
Collapse
|
48
|
Biressi S, Filareto A, Rando TA. Stem cell therapy for muscular dystrophies. J Clin Invest 2021; 130:5652-5664. [PMID: 32946430 DOI: 10.1172/jci142031] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Muscular dystrophies are a heterogeneous group of genetic diseases, characterized by progressive degeneration of skeletal and cardiac muscle. Despite the intense investigation of different therapeutic options, a definitive treatment has not been developed for this debilitating class of pathologies. Cell-based therapies in muscular dystrophies have been pursued experimentally for the last three decades. Several cell types with different characteristics and tissues of origin, including myogenic stem and progenitor cells, stromal cells, and pluripotent stem cells, have been investigated over the years and have recently entered in the clinical arena with mixed results. In this Review, we do a roundup of the past attempts and describe the updated status of cell-based therapies aimed at counteracting the skeletal and cardiac myopathy present in dystrophic patients. We present current challenges, summarize recent progress, and make recommendations for future research and clinical trials.
Collapse
Affiliation(s)
- Stefano Biressi
- Department of Cellular, Computational and Integrative Biology (CIBIO) and.,Dulbecco Telethon Institute, University of Trento, Povo, Italy
| | - Antonio Filareto
- Department of Research Beyond Borders, Regenerative Medicine, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Conneticut, USA
| | - Thomas A Rando
- Department of Neurology and Neurological Sciences and.,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, California, USA.,Center for Tissue Regeneration, Repair and Restoration, Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
49
|
Ishigami S, Sano T, Krishnapura S, Ito T, Sano S. An overview of stem cell therapy for paediatric heart failure. Eur J Cardiothorac Surg 2020; 58:881-887. [PMID: 32588055 DOI: 10.1093/ejcts/ezaa155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 11/13/2022] Open
Abstract
Significant achievements in paediatric cardiology, surgical treatment and intensive care of congenital heart disease have drastically changed clinical outcomes for paediatric patients. Nevertheless, late-onset heart failure in children after staged surgeries still remains a serious concern in the medical community. Heart transplantation is an option for treatment; however, the shortage of available organs is a persistent problem in many developed countries. In order to resolve these issues, advanced technologies, such as innovative mechanical circulatory support devices and regenerative therapies, are strongly desired. Accumulated evidence regarding cell-based cardiac regenerative therapies has suggested their safety and efficacy in treating adult heart failure. Given that young children seem to have a higher regenerative capacity than adults, stem cell-based therapies appear a promising treatment option for paediatric heart failure as well. Based on the findings from past trials and studies, we present the potential of various different types of stem cells, ranging from bone marrow mononuclear cells to cardiosphere-derived stem cells for use in paediatric cell-based therapies. Here, we assess both the current challenges associated with cell-based therapies and novel strategies that may be implemented in the future to advance stem cell therapy in the paediatric population.
Collapse
Affiliation(s)
- Shuta Ishigami
- Department of Pediatric Cardiothoracic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Toshikazu Sano
- Department of Pediatric Cardiothoracic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Sunaya Krishnapura
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Tatsuo Ito
- Department of Hygiene, Kawasaki Medical University, Kurashiki, Japan
| | - Shunji Sano
- Department of Pediatric Cardiothoracic Surgery, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
50
|
Menasché P. Cell Therapy With Human ESC-Derived Cardiac Cells: Clinical Perspectives. Front Bioeng Biotechnol 2020; 8:601560. [PMID: 33195177 PMCID: PMC7649799 DOI: 10.3389/fbioe.2020.601560] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/02/2020] [Indexed: 12/12/2022] Open
Abstract
In the ongoing quest for the “ideal” cell type for heart repair, pluripotent stem cells (PSC) derived from either embryonic or reprogrammed somatic cells have emerged as attractive candidates because of their unique ability to give rise to lineage-specific cells and to transplant them at the desired stage of differentiation. The technical obstacles which have initially hindered their clinical use have now been largely overcome and several trials are under way which encompass several different diseases, including heart failure. So far, there have been no safety warning but it is still too early to draw definite conclusions regarding efficacy. In parallel, mechanistic studies suggest that the primary objective of “remuscularizing” the heart with PSC-derived cardiac cells can be challenged by their alternate use as ex vivo sources of a biologically active extracellular vesicle-enriched secretome equally able to improve heart function through harnessing endogenous repair pathways. The exclusive use of this secretome would combine the advantages of a large-scale production more akin to that of a biological medication, the likely avoidance of cell-associated immune and tumorigenicity risks and the possibility of intravenous infusions compatible with repeated dosing.
Collapse
Affiliation(s)
- Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou, Paris, France.,PARCC, INSERM, University of Paris, Paris, France
| |
Collapse
|