1
|
Yahyazadeh R, Baradaran Rahimi V, Askari VR. Stem cell and exosome therapies for regenerating damaged myocardium in heart failure. Life Sci 2024; 351:122858. [PMID: 38909681 DOI: 10.1016/j.lfs.2024.122858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Finding novel treatments for cardiovascular diseases (CVDs) is a hot topic in medicine; cell-based therapies have reported promising news for controlling dangerous complications of heart disease such as myocardial infarction (MI) and heart failure (HF). Various progenitor/stem cells were tested in various in-vivo, in-vitro, and clinical studies for regeneration or repairing the injured tissue in the myocardial to accelerate the healing. Fetal, adult, embryonic, and induced pluripotent stem cells (iPSC) have revealed the proper potency for cardiac tissue repair. As an essential communicator among cells, exosomes with specific contacts (proteins, lncRNAs, and miRNAs) greatly promote cardiac rehabilitation. Interestingly, stem cell-derived exosomes have more efficiency than stem cell transplantation. Therefore, stem cells induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), cardiac stem cells (CDC), and skeletal myoblasts) and their-derived exosomes will probably be considered an alternative therapy for CVDs remedy. In addition, stem cell-derived exosomes have been used in the diagnosis/prognosis of heart diseases. In this review, we explained the advances of stem cells/exosome-based treatment, their beneficial effects, and underlying mechanisms, which will present new insights in the clinical field in the future.
Collapse
Affiliation(s)
- Roghayeh Yahyazadeh
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Zhang J, Suo M, Wang J, Liu X, Huang H, Wang K, Liu X, Sun T, Li Z, Liu J. Standardisation is the key to the sustained, rapid and healthy development of stem cell-based therapy. Clin Transl Med 2024; 14:e1646. [PMID: 38572666 PMCID: PMC10993161 DOI: 10.1002/ctm2.1646] [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: 12/08/2023] [Revised: 02/20/2024] [Accepted: 03/17/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Stem cell-based therapy (SCT) is an important component of regenerative therapy that brings hope to many patients. After decades of development, SCT has made significant progress in the research of various diseases, and the market size has also expanded significantly. The transition of SCT from small-scale, customized experiments to routine clinical practice requires the assistance of standards. Many countries and international organizations around the world have developed corresponding SCT standards, which have effectively promoted the further development of the SCT industry. METHODS We conducted a comprehensive literature review to introduce the clinical application progress of SCT and focus on the development status of SCT standardization. RESULTS We first briefly introduced the types and characteristics of stem cells, and summarized the current clinical application and market development of SCT. Subsequently, we focused on the development status of SCT-related standards as of now from three levels: the International Organization for Standardization (ISO), important international organizations, and national organizations. Finally, we provided perspectives and conclusions on the significance and challenges of SCT standardization. CONCLUSIONS Standardization plays an important role in the sustained, rapid and healthy development of SCT.
Collapse
Affiliation(s)
- Jing Zhang
- Department of OrthopedicsFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoning ProvinceChina
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic DiseasesDalianLiaoning ProvinceChina
| | - Moran Suo
- Department of OrthopedicsFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoning ProvinceChina
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic DiseasesDalianLiaoning ProvinceChina
| | - Jinzuo Wang
- Department of OrthopedicsFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoning ProvinceChina
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic DiseasesDalianLiaoning ProvinceChina
| | - Xin Liu
- Department of OrthopedicsFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoning ProvinceChina
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic DiseasesDalianLiaoning ProvinceChina
| | - Huagui Huang
- Department of OrthopedicsFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoning ProvinceChina
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic DiseasesDalianLiaoning ProvinceChina
| | - Kaizhong Wang
- Department of OrthopedicsFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoning ProvinceChina
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic DiseasesDalianLiaoning ProvinceChina
| | - Xiangyan Liu
- Department of OrthopedicsFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoning ProvinceChina
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic DiseasesDalianLiaoning ProvinceChina
| | - Tianze Sun
- Department of OrthopedicsFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoning ProvinceChina
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic DiseasesDalianLiaoning ProvinceChina
| | - Zhonghai Li
- Department of OrthopedicsFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoning ProvinceChina
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic DiseasesDalianLiaoning ProvinceChina
- Stem Cell Clinical Research CenterNational Joint Engineering LaboratoryFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoning ProvinceChina
- Dalian Innovation Institute of Stem Cell and Precision MedicineDalianLiaoning ProvinceChina
| | - Jing Liu
- Stem Cell Clinical Research CenterNational Joint Engineering LaboratoryFirst Affiliated Hospital of Dalian Medical UniversityDalianLiaoning ProvinceChina
- Dalian Innovation Institute of Stem Cell and Precision MedicineDalianLiaoning ProvinceChina
| |
Collapse
|
3
|
Kishino Y, Tohyama S. An Approach That Brings Out the Potential of Regenerative Therapies in Heart Failure. Circ J 2023; 87:487-489. [PMID: 36642512 DOI: 10.1253/circj.cj-22-0781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine
| |
Collapse
|
4
|
Banovic M, Poglajen G, Vrtovec B, Ristic A. Contemporary Challenges of Regenerative Therapy in Patients with Ischemic and Non-Ischemic Heart Failure. J Cardiovasc Dev Dis 2022; 9:jcdd9120429. [PMID: 36547426 PMCID: PMC9783726 DOI: 10.3390/jcdd9120429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/11/2022] [Accepted: 10/26/2022] [Indexed: 12/02/2022] Open
Abstract
It has now been almost 20 years since first clinical trials of stem cell therapy for heart repair were initiated. While initial preclinical data were promising and suggested that stem cells may be able to directly restore a diseased myocardium, this was never unequivocally confirmed in the clinical setting. Clinical trials of cell therapy did show the process to be feasible and safe. However, the clinical benefits of this treatment modality in patients with ischemic and non-ischemic heart failure have not been consistently confirmed. What is more, in the rapidly developing field of stem cell therapy in patients with heart failure, relevant questions regarding clinical trials' protocol streamlining, optimal patient selection, stem cell type and dose, and the mode of cell delivery remain largely unanswered. Recently, novel approaches to myocardial regeneration, including the use of pluripotent and allogeneic stem cells and cell-free therapeutic approaches, have been proposed. Thus, in this review, we aim to outline current knowledge and highlight contemporary challenges and dilemmas in clinical aspects of stem cell and regenerative therapy in patients with chronic ischemic and non-ischemic heart failure.
Collapse
Affiliation(s)
- Marko Banovic
- Cardiology Department, University Clinical Center of Serbia, 11000 Beograd, Serbia
- Belgrade Medical School, 11000 Belgrade, Serbia
- Correspondence: (M.B.); (G.P.)
| | - Gregor Poglajen
- Advanced Heart Failure and Transplantation Center, Department of Cardiology, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Medical Faculty Ljubljana, University of Ljubljana, 1000 Ljubljana, Slovenia
- Correspondence: (M.B.); (G.P.)
| | - Bojan Vrtovec
- Advanced Heart Failure and Transplantation Center, Department of Cardiology, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Medical Faculty Ljubljana, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Arsen Ristic
- Cardiology Department, University Clinical Center of Serbia, 11000 Beograd, Serbia
- Belgrade Medical School, 11000 Belgrade, Serbia
| |
Collapse
|
5
|
Gyöngyösi M, Pokushalov E, Romanov A, Perin E, Hare JM, Kastrup J, Fernández-Avilés F, Sanz-Ruiz R, Mathur A, Wojakowski W, Martin-Rendon E, Pavo N, Pavo IJ, Hemetsberger R, Traxler D, Spannbauer A, Haller PM. Meta-Analysis of Percutaneous Endomyocardial Cell Therapy in Patients with Ischemic Heart Failure by Combination of Individual Patient Data (IPD) of ACCRUE and Publication-Based Aggregate Data. J Clin Med 2022; 11:jcm11113205. [PMID: 35683592 PMCID: PMC9181462 DOI: 10.3390/jcm11113205] [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: 05/03/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 12/10/2022] Open
Abstract
Individual patient data (IPD)-based meta-analysis (ACCRUE, meta-analysis of cell-based cardiac studies, NCT01098591) revealed an insufficient effect of intracoronary cell-based therapy in acute myocardial infarction. Patients with ischemic heart failure (iHF) have been treated with reparative cells using percutaneous endocardial, surgical, transvenous or intracoronary cell delivery methods, with variable effects in small randomized or cohort studies. The objective of this meta-analysis was to investigate the safety and efficacy of percutaneous transendocardial cell therapy in patients with iHF. Two investigators extracted the data. Individual patient data (IPD) (n = 8 studies) and publication-based (n = 10 studies) aggregate data were combined for the meta-analysis, including patients (n = 1715) with chronic iHF. The data are reported in accordance with PRISMA guidelines. The primary safety and efficacy endpoints were all-cause mortality and changes in global ejection fraction. The secondary safety and efficacy endpoints were major adverse events, hospitalization and changes in end-diastolic and end-systolic volumes. Post hoc analyses were performed using the IPD of eight studies to find predictive factors for treatment safety and efficacy. Cell therapy was significantly (p < 0.001) in favor of survival, major adverse events and hospitalization during follow-up. A forest plot analysis showed that cell therapy presents a significant benefit of increasing ejection fraction with a mean change of 2.51% (95% CI: 0.48; 4.54) between groups and of significantly decreasing end-systolic volume. The analysis of IPD data showed an improvement in the NYHA and CCS classes. Cell therapy significantly decreased the end-systolic volume in male patients; in patients with diabetes mellitus, hypertension or hyperlipidemia; and in those with previous myocardial infarction and baseline ejection fraction ≤ 45%. The catheter-based transendocardial delivery of regenerative cells proved to be safe and effective for improving mortality and cardiac performance. The greatest benefit was observed in male patients with significant atherosclerotic co-morbidities.
Collapse
Affiliation(s)
- Mariann Gyöngyösi
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.P.); (R.H.); (D.T.); (A.S.)
- Correspondence: ; Tel.: +43-1-40400-46140
| | - Evgeny Pokushalov
- Center of the New and Modern Medical Technologies, 630090 Novosibirsk, Russia;
| | - Aleksander Romanov
- E. Meshalkin National Medical Research Center, 630055 Novosibirsk, Russia;
| | - Emerson Perin
- Stem Cell Center and Adult Cardiology, Texas Heart Institute, Houston, TX 37660, USA;
| | - Joshua M. Hare
- Interdisciplinary Stem Cell Institute, Cardiovascular Division, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Jens Kastrup
- Cardiology Stem Cell Centre, The Centre for Cardiac, Vascular, Pulmonary and Infectious Diseases, Copenhagen University Hospital Rigshospitalet, 2100 Copenhagen, Denmark;
| | | | - Ricardo Sanz-Ruiz
- CIBERCV, Instituto de Salud Carlos III, 28029 Madrid, Spain; (F.F.-A.); (R.S.-R.)
| | - Anthony Mathur
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK;
| | - Wojcieh Wojakowski
- Department of Cardiology and Structural Heart Diseases, Medical University of Silesia, 40-635 Katowice, Poland;
| | - Enca Martin-Rendon
- R&D Division, National Health Service (NHS)-Blood and Transplant, Oxford Centre, Oxford OX3 9DU, UK;
| | - Noemi Pavo
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.P.); (R.H.); (D.T.); (A.S.)
| | - Imre J. Pavo
- Department of Pediatrics, Medical University of Vienna, 1090 Vienna, Austria;
| | - Rayyan Hemetsberger
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.P.); (R.H.); (D.T.); (A.S.)
| | - Denise Traxler
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.P.); (R.H.); (D.T.); (A.S.)
| | - Andreas Spannbauer
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria; (N.P.); (R.H.); (D.T.); (A.S.)
| | - Paul M. Haller
- Department of Cardiology, University Heart and Vascular Center UKE Hamburg, 20246 Hamburg, Germany;
| |
Collapse
|
6
|
Afjeh-Dana E, Naserzadeh P, Moradi E, Hosseini N, Seifalian AM, Ashtari B. Stem Cell Differentiation into Cardiomyocytes: Current Methods and Emerging Approaches. Stem Cell Rev Rep 2022; 18:2566-2592. [PMID: 35508757 DOI: 10.1007/s12015-021-10280-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 12/26/2022]
Abstract
Cardiovascular diseases (CVDs) are globally known to be important causes of mortality and disabilities. Common treatment strategies for CVDs, such as pharmacological therapeutics impose serious challenges due to the failure of treatments for myocardial necrosis. By contrast, stem cells (SCs) based therapies are seen to be promising approaches to CVDs treatment. In such approaches, cardiomyocytes are differentiated from SCs. To fulfill SCs complete potential, the method should be appointed to generate cardiomyocytes with more mature structure and well-functioning operations. For heart repairing applications, a greatly scalable and medical-grade cardiomyocyte generation must be used. Nonetheless, there are some challenges such as immune rejection, arrhythmogenesis, tumorigenesis, and graft cell death potential. Herein, we discuss the types of potential SCs, and commonly used methods including embryoid bodies related techniques, co-culture, mechanical stimulation, and electrical stimulation and their applications, advantages and limitations in this field. An estimated 17.9 million people died from CVDs in 2019, representing 32 % of all global deaths. Of these deaths, 85 % were due to heart attack and stroke.
Collapse
Affiliation(s)
- Elham Afjeh-Dana
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Parvaneh Naserzadeh
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Moradi
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Nasrin Hosseini
- Neuroscience Research Centre, Iran University of Medical Sciences, Tehran, Iran.
| | - Alexander Marcus Seifalian
- Nanotechnology & Regenerative Medicine Commercialisation Centre (NanoRegMed Ltd), London BioScience Innovation Centre, London, UK
| | - Behnaz Ashtari
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran. .,Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran. .,Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Xu Z, Neuber S, Nazari-Shafti T, Liu Z, Dong F, Stamm C. Impact of procedural variability and study design quality on the efficacy of cell-based therapies for heart failure - a meta-analysis. PLoS One 2022; 17:e0261462. [PMID: 34986181 PMCID: PMC8730409 DOI: 10.1371/journal.pone.0261462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 12/02/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Cell-based therapy has long been considered a promising strategy for the treatment of heart failure (HF). However, its effectiveness in the clinical setting is now doubted. Because previous meta-analyses provided conflicting results, we sought to review all available data focusing on cell type and trial design. METHODS AND FINDINGS The electronic databases PubMed, Cochrane library, ClinicalTrials.gov, and EudraCT were searched for randomized controlled trials (RCTs) utilizing cell therapy for HF patients from January 1, 2000 to December 31, 2020. Forty-three RCTs with 2855 participants were identified. The quality of the reported study design was assessed by evaluating the risk-of-bias (ROB). Primary outcomes were defined as mortality rate and left ventricular ejection fraction (LVEF) change from baseline. Secondary outcomes included both heart function data and clinical symptoms/events. Between-study heterogeneity was assessed using the I2 index. Subgroup analysis was performed based on HF type, cell source, cell origin, cell type, cell processing, type of surgical intervention, cell delivery routes, cell dose, and follow-up duration. Only 10 of the 43 studies had a low ROB for all method- and outcome parameters. A higher ROB was associated with a greater increase in LVEF. Overall, there was no impact on mortality for up to 12 months follow-up, and a clinically irrelevant average LVEF increase by LVEF (2.4%, 95% CI = 0.75-4.05, p = 0.004). Freshly isolated, primary cells tended to produce better outcomes than cultured cell products, but there was no clear impact of the cell source tissue, bone marrow cell phenotype or cell chricdose (raw or normalized for CD34+ cells). A meaningful increase in LVEF was only observed when cell therapy was combined with myocardial revascularization. CONCLUSIONS The published results suggest a small increase in LVEF following cell therapy for heart failure, but publication bias and methodologic shortcomings need to be taken into account. Given that cardiac cell therapy has now been pursued for 20 years without real progress, further efforts should not be made. STUDY REGISTRY NUMBER This meta-analysis is registered at the international prospective register of systematic reviews, number CRD42019118872.
Collapse
Affiliation(s)
- Zhiyi Xu
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Neuber
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
| | - Timo Nazari-Shafti
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Zihou Liu
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Fengquan Dong
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Christof Stamm
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- Helmholtz Zentrum Geesthacht, Institut für Aktive Polymere, Teltow, Germany
| |
Collapse
|
8
|
Bhawnani N, Ethirajulu A, Alkasabera A, Onyali CB, Anim-Koranteng C, Shah HE, Mostafa JA. Effectiveness of Stem Cell Therapies in Improving Clinical Outcomes in Patients With Heart Failure. Cureus 2021; 13:e17236. [PMID: 34540463 PMCID: PMC8447853 DOI: 10.7759/cureus.17236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/16/2021] [Indexed: 11/15/2022] Open
Abstract
Heart failure (HF), continuing to be a notable cause of morbidity and mortality worldwide, also is a noteworthy economic burden to the patients. Current medical management of HF has poor efficacy to completely arrest or reverse the progression to end-stage disease. As the option of cardiac transplantation remains limited to few patients, the stem cell approach continues to be a promising one in developing a novel therapy in the treatment of HF. This review attempts to discuss and compare the outcomes of numerous clinical trials that involved treatment of HF of variable etiologies with stem cells of numerous lineages such as bone marrow-derived cells (BMCs), mesenchymal stem cells (MSCs), cardiosphere derived progenitor cells (CDCs), etc. We reviewed articles and randomized controlled trials (RCT) that used stem cells to treat heart failure. The articles and RCT studies were obtained through a search on PubMed and Medline databases and performed using regular and medical subject heading (MeSH) keyword search strategy. A total of 17 trial-based studies, along with other articles that met the aim of the review, were selected. A discussion of the findings from major clinical trials such as the C-CURE, CHART-1, POSEIDON, POSEIDON-DCM, TAC-HFT, and other small scale trials highlights the change in functional and mechanical parameters of HF, namely, left ventricular ejection fraction (LVEF), end-diastolic volume (EDV), end-systolic volume (ESV), 6-minute walking test distance (6MWTD), N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels and assessment of New York heart association (NYHA) class of heart failure, and Minnesota Living with Heart Failure Questionnaire (MLHFQ) score to reflect improvement in quality of life (QoL) of patients. Out of the studies analyzed, the majority reported significant improvements in at least two of the parameters mentioned above. However, more phase three randomized trials are required to compare the efficacy of multiple lineages of stem cells, factoring in molecular and dosage factors to develop a standardized therapy.
Collapse
Affiliation(s)
- Nitin Bhawnani
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Aarthi Ethirajulu
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Almothana Alkasabera
- General Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Chike B Onyali
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | | | - Hira E Shah
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Jihan A Mostafa
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
9
|
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
|
10
|
Evolution of Stem Cells in Cardio-Regenerative Therapy. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
11
|
Cheung MM, Jahan N. Can Stem Cells Improve Left Ventricular Ejection Fraction in Heart Failure? A Literature Review of Skeletal Myoblasts and Bone Marrow-Derived Cells. Cureus 2020; 12:e11598. [PMID: 33364119 PMCID: PMC7752736 DOI: 10.7759/cureus.11598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/20/2020] [Indexed: 01/05/2023] Open
Abstract
Heart failure is a life-threatening condition that affects millions worldwide and is only expected to get worse with an ageing population. Current treatment regimens rely on medical therapy and heart transplantation as a last resort. Stem cells have been undergoing clinical trials worldwide as a hope for a new and safe clinical treatment. Skeletal myoblasts and bone marrow-derived stem cells are two types of stem cells being tested. The objective is to evaluate the efficacy of these two types of stem cells for heart failure therapy. Data were searched in PubMed using both regular and Medical Subject Heading (MeSH) keywords (stem cells, therapy, heart failure) and then filtered using inclusion/exclusion criteria (language, species, publication date, and age). In total, 31 research articles were reviewed (14 clinical trials, four randomized control trials, nine review articles, one case report, one comparative study, one systematic review, and one categorized as a systematic review and meta-analysis). Both skeletal myoblasts and bone marrow-derived stem cells showed mixed results in improving left ventricular ejection fraction in heart failure patients in the majority of studies. Larger studies need to be done to further investigate the efficacy of stem cells as a therapy for heart failure.
Collapse
Affiliation(s)
- Meghan M Cheung
- Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Nusrat Jahan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
12
|
Selvakumar D, Clayton ZE, Chong JJH. Robust Cardiac Regeneration: Fulfilling the Promise of Cardiac Cell Therapy. Clin Ther 2020; 42:1857-1879. [PMID: 32943195 DOI: 10.1016/j.clinthera.2020.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE We review the history of cardiac cell therapy, highlighting lessons learned from initial adult stem cell (ASC) clinical trials. We present pluripotent stem cell-derived cardiomyocytes (PSC-CMs) as a leading candidate for robust regeneration of infarcted myocardium but identify several issues that must be addressed before successful clinical translation. METHODS We conducted an unstructured literature review of PubMed-listed articles, selecting the most comprehensive and relevant research articles, review articles, clinical trials, and basic or translation articles in the field of cardiac cell therapy. Articles were identified using the search terms adult stem cells, pluripotent stem cells, cardiac stem cell, and cardiac regeneration or from references of relevant articles, Articles were prioritized and selected based on their impact, originality, or potential clinical applicability. FINDINGS Since its inception, the ASC therapy field has been troubled by conflicting preclinical data, academic controversies, and inconsistent trial designs. These issues have damaged perceptions of cardiac cell therapy among investors, the academic community, health care professionals, and, importantly, patients. In hindsight, the key issue underpinning these problems was the inability of these cell types to differentiate directly into genuine cardiomyocytes, rendering them unable to replace damaged myocardium. Despite this, beneficial effects through indirect paracrine or immunomodulatory effects remain possible and continue to be investigated. However, in preclinical models, PSC-CMs have robustly remuscularized infarcted myocardium with functional, force-generating cardiomyocytes. Hence, PSC-CMs have now emerged as a leading candidate for cardiac regeneration, and unpublished reports of first-in-human delivery of these cells have recently surfaced. However, the cardiac cell therapy field's history should serve as a cautionary tale, and we identify several translational hurdles that still remain. Preclinical solutions to issues such as arrhythmogenicity, immunogenicity, and poor engraftment rates are needed, and next-generation clinical trials must draw on robust knowledge of mechanistic principles of the therapy. IMPLICATIONS The clinical transplantation of functional stem cell-derived heart tissue with seamless integration into native myocardium is a lofty goal. However, considerable advances have been made during the past 2 decades. Currently, PSC-CMs appear to be the best prospect to reach this goal, but several hurdles remain. The history of adult stem cell trials has taught us that shortcuts cannot be taken without dire consequences, and it is essential that progress not be hurried and that a worldwide, cross-disciplinary approach be used to ensure safe and effective clinical translation.
Collapse
Affiliation(s)
- Dinesh Selvakumar
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Zoe E Clayton
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - James J H Chong
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia.
| |
Collapse
|
13
|
Tamaki T. Biomedical applications of muscle-derived stem cells: from bench to bedside. Expert Opin Biol Ther 2020; 20:1361-1371. [PMID: 32643444 DOI: 10.1080/14712598.2020.1793953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Skeletal muscle-derived stem cells (Sk-MDSCs) are considered promising sources of adult stem cell therapy. Skeletal muscle comprises approximately 40-50% of the total body mass with marked potential for postnatal adaptive response, such as muscle hypertrophy, hyperplasia, atrophy, and regenerative capacity. This strongly suggests that skeletal muscle contains various stem/progenitor cells related to muscle-nerve-vascular tissues, which would support the above postnatal events even in adulthood. AREA COVERED The focus of this review is the therapeutic potential of the Sk-MDSCs as an adult stem cell autograft. For this purpose, the validity of cell isolation and purification, tissue reconstitution capacity in vivo after transplantation, comparison of the results of basic mouse and preclinical human studies, potential problematic and beneficial aspects, and effective usage have been discussed following the history of clinical applications. EXPERT OPINION Although the clinical application of Sk-MDSCs began as a therapy for the systemic disease of Duchenne muscular dystrophy, here, through the unique local injection method, therapy for severely damaged peripheral nerves, particularly the long-gap nerve transection, has been introduced. The beneficial aspects of the use of Sk-MDSCs as the source of local tissue transplantation therapy have also been discussed.
Collapse
Affiliation(s)
- Tetsuro Tamaki
- Muscle Physiology and Cell Biology Unit, Department of Physiology, Tokai University School of Medicine , Isehara, Kanagawa ,Japan
| |
Collapse
|
14
|
White SJ, Chong JJH. Growth factor therapy for cardiac repair: an overview of recent advances and future directions. Biophys Rev 2020; 12:805-815. [PMID: 32691300 DOI: 10.1007/s12551-020-00734-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/08/2020] [Indexed: 12/21/2022] Open
Abstract
Heart disease represents a significant public health burden and is associated with considerable morbidity and mortality at the level of the individual. Current therapies for pathologies such as myocardial infarction, cardiomyopathy and heart failure are unable to repair damaged tissue to an extent that provides restoration of function approaching that of the pre-diseased state. Novel approaches to repair and regenerate the injured heart include cell therapy and the use of exogenous factors. Improved understanding of the role of growth factors in endogenous cardiac repair processes has motivated the investigation of their potential as therapeutic agents for cardiac pathology. Despite the disappointing performance of other growth factors in historical clinical trials, insulin-like growth factor 1 (IGF-1), neuregulin and platelet-derived growth factor (PDGF) have recently emerged as new candidate therapies. These growth factors elicit tissue repair through anti-apoptotic, pro-angiogenic and fibrosis-modulating mechanisms and have produced clinically significant functional improvement in preclinical studies. Early human trials suggest that IGF-1 and neuregulin are well tolerated and yield dose-dependent benefit, warranting progression to later phase studies. However, outstanding challenges such as short growth factor serum half-life and insufficient target-organ specificity currently necessitate the development of novel delivery strategies.
Collapse
Affiliation(s)
- Samuel J White
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - James J H Chong
- Centre for Heart Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia.
- Department of Cardiology, Westmead Hospital, Westmead, NSW, 2145, Australia.
| |
Collapse
|
15
|
Raval AN, Pepine CJ. Clinical Safety Profile of Transendocardial Catheter Injection Systems: A Plea for Uniform Reporting. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2020; 22:100-108. [PMID: 32651159 DOI: 10.1016/j.carrev.2020.06.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The aim of this study was to characterize the clinical safety profile of transendocardial injection catheters (TIC) reported in the published literature. BACKGROUND Transendocardial delivery is a minimally invasive approach to deliver potential therapeutic agents directly into the myocardium. The rate of adverse events across TIC is uncertain. METHODS A systematic search was performed for trial publications using TIC. Procedure-associated adverse event data were abstracted, pooled and compared across catheters for active treatment and placebo injected patients. The transendocardial injection associated serious adverse events (TEI-SAE) was defined as the composite of death, myocardial infarction, stroke or transient ischemic attack within 30 days and cardiac perforation causing death or requiring evacuation, serious intraprocedural arrhythmias and serious coronary artery or peripheral vascular complications. RESULTS The search identified 4 TIC systems: a helical needle (HN), an electro-anatomically tracked straight needle (EAM-SN), a straight needle without tracking elements (SN), and a curved needle (CN). Of 1799 patients who underwent transendocardial injections, the combined TEI-SAE was 3.4% across all catheters, and 1.1%, 3.3%, 7.1%, and 8.3% for HN, EAM-SN, SN and CN, respectively. However, TIC procedure duration and post procedural cardiac biomarker levels were reported in only 24% and 36% of published trials, respectively. CONCLUSIONS Transendocardial injection is associated with varied TEI-SAE but the data are very limited. The HN catheter appeared to be associated with lower TEI-SAE, versus other catheters. Procedure duration and post procedure cardiac biomarker levels were under-reported. Clearly, standardized, procedure-related event reporting for trials involving transcatheter delivery would improve our understanding of complications across different systems.
Collapse
Affiliation(s)
- Amish N Raval
- Department of Medicine and Biomedical Engineering, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - Carl J Pepine
- Division of Cardiovascular Medicine, University of Florida, Gainsville, FL, USA
| |
Collapse
|
16
|
Prat-Vidal C, Rodríguez-Gómez L, Aylagas M, Nieto-Nicolau N, Gastelurrutia P, Agustí E, Gálvez-Montón C, Jorba I, Teis A, Monguió-Tortajada M, Roura S, Vives J, Torrents-Zapata S, Coca MI, Reales L, Cámara-Rosell ML, Cediel G, Coll R, Farré R, Navajas D, Vilarrodona A, García-López J, Muñoz-Guijosa C, Querol S, Bayes-Genis A. First-in-human PeriCord cardiac bioimplant: Scalability and GMP manufacturing of an allogeneic engineered tissue graft. EBioMedicine 2020; 54:102729. [PMID: 32304998 PMCID: PMC7163319 DOI: 10.1016/j.ebiom.2020.102729] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/24/2020] [Accepted: 03/05/2020] [Indexed: 02/07/2023] Open
Abstract
Background Small cardiac tissue engineering constructs show promise for limiting post-infarct sequelae in animal models. This study sought to scale-up a 2-cm2 preclinical construct into a human-size advanced therapy medicinal product (ATMP; PeriCord), and to test it in a first-in-human implantation. Methods The PeriCord is a clinical-size (12–16 cm2) decellularised pericardial matrix colonised with human viable Wharton's jelly-derived mesenchymal stromal cells (WJ-MSCs). WJ-MSCs expanded following good manufacturing practices (GMP) met safety and quality standards regarding the number of cumulative population doublings, genomic stability, and sterility. Human decellularised pericardial scaffolds were tested for DNA content, matrix stiffness, pore size, and absence of microbiological growth. Findings PeriCord implantation was surgically performed on a large non-revascularisable scar in the inferior wall of a 63-year-old male patient. Coronary artery bypass grafting was concomitantly performed in the non-infarcted area. At implantation, the 16-cm2 pericardial scaffold contained 12·5 × 106 viable WJ-MSCs (85·4% cell viability; <0·51 endotoxin units (EU)/mL). Intraoperative PeriCord delivery was expeditious, and secured with surgical glue. The post-operative course showed non-adverse reaction to the PeriCord, without requiring host immunosuppression. The three-month clinical follow-up was uneventful, and three-month cardiac magnetic resonance imaging showed ~9% reduction in scar mass in the treated area. Interpretation This preliminary report describes the development of a scalable clinical-size allogeneic PeriCord cardiac bioimplant, and its first-in-human implantation. Funding La Marató de TV3 Foundation, Government of Catalonia, Catalan Society of Cardiology, “La Caixa” Banking Foundation, Spanish Ministry of Science, Innovation and Universities, Institute of Health Carlos III, and the European Regional Development Fund.
Collapse
Affiliation(s)
- Cristina Prat-Vidal
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain; Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica de Bellvitge - IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Luciano Rodríguez-Gómez
- Cell Therapy Service, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Miriam Aylagas
- Cell Therapy Service, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain; Transfusional Medicine Group, Vall d'Hebron Research Institute, Autonomous University of Barcelona, Barcelona, Spain
| | | | - Paloma Gastelurrutia
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain; Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain; Institut d'Investigació Biomèdica de Bellvitge - IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Elba Agustí
- Barcelona Tissue Bank (BTB), Banc de Sang i Teixits (BST), Barcelona, Spain
| | - Carolina Gálvez-Montón
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain; Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| | - Ignasi Jorba
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, CIBERES, University of Barcelona, Barcelona, Spain
| | - Albert Teis
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain
| | - Marta Monguió-Tortajada
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain; REMAR-IVECAT Group, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain
| | - Santiago Roura
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain; Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain
| | - Joaquim Vives
- Cell Therapy Service, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain; Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Autonomous University of Barcelona, Passeig de la Vall d'Hebron 129-139, 08035 Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035 Barcelona, Spain
| | - Silvia Torrents-Zapata
- Cell Therapy Service, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - María Isabel Coca
- Cell Therapy Service, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Laura Reales
- Cell Therapy Service, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - María Luisa Cámara-Rosell
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain
| | - Germán Cediel
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain
| | - Ruth Coll
- Research and Education. Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Ramon Farré
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona-IDIBAPS-CIBERES, Barcelona, Spain
| | - Daniel Navajas
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, CIBERES, University of Barcelona, Barcelona, Spain
| | - Anna Vilarrodona
- Barcelona Tissue Bank (BTB), Banc de Sang i Teixits (BST), Barcelona, Spain
| | - Joan García-López
- Research and Education. Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain
| | - Christian Muñoz-Guijosa
- Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain
| | - Sergi Querol
- Cell Therapy Service, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005 Barcelona, Spain; Transfusional Medicine Group, Vall d'Hebron Research Institute, Autonomous University of Barcelona, Barcelona, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Germans Trias i Pujol Health Science Research Institute, Can Ruti Campus, Badalona, Spain; Heart Institute (iCor), Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916 Badalona, Spain; CIBER Cardiovascular, Instituto de Salud Carlos III, Madrid, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035 Barcelona, Spain.
| |
Collapse
|
17
|
Samak M, Hinkel R. Stem Cells in Cardiovascular Medicine: Historical Overview and Future Prospects. Cells 2019; 8:cells8121530. [PMID: 31783680 PMCID: PMC6952821 DOI: 10.3390/cells8121530] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/19/2019] [Accepted: 11/23/2019] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases remain the leading cause of death in the developed world, accounting for more than 30% of all deaths. In a large proportion of these patients, acute myocardial infarction is usually the first manifestation, which might further progress to heart failure. In addition, the human heart displays a low regenerative capacity, leading to a loss of cardiomyocytes and persistent tissue scaring, which entails a morbid pathologic sequela. Novel therapeutic approaches are urgently needed. Stem cells, such as induced pluripotent stem cells or embryonic stem cells, exhibit great potential for cell-replacement therapy and an excellent tool for disease modeling, as well as pharmaceutical screening of novel drugs and their cardiac side effects. This review article covers not only the origin of stem cells but tries to summarize their translational potential, as well as potential risks and clinical translation.
Collapse
Affiliation(s)
- Mostafa Samak
- Department of Laboratory Animal Science, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Rabea Hinkel
- Department of Laboratory Animal Science, Leibnitz-Institut für Primatenforschung, Deutsches Primatenzentrum GmbH, Kellnerweg 4, 37077 Göttingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
18
|
Fernandes GC, Fernandes ADF, Rivera M, Khan A, Schulman IH, Lambrakos LK, Myerburg RJ, Goldberger JJ, Hare JM, Mitrani RD. A meta‐analysis of arrhythmia endpoints in randomized controlled trials of transendocardial stem cell injections for chronic ischemic heart disease. J Cardiovasc Electrophysiol 2019; 30:2492-2500. [DOI: 10.1111/jce.14185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 09/02/2019] [Accepted: 09/11/2019] [Indexed: 02/01/2023]
Affiliation(s)
- Gilson C. Fernandes
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
| | | | - Manuel Rivera
- Division of Cardiology and Cardiovascular Washington University in Saint Louis Saint Louis Missouri
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute University of Miami Miller School of Medicine Miami Florida
| | - Ivonne H. Schulman
- Department of Medicine University of Miami Miller School of Medicine Miami Florida
- Interdisciplinary Stem Cell Institute University of Miami Miller School of Medicine Miami Florida
| | - Litsa K. Lambrakos
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
| | - Robert J. Myerburg
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
| | | | - Joshua M. Hare
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
- Interdisciplinary Stem Cell Institute University of Miami Miller School of Medicine Miami Florida
| | - Raul D. Mitrani
- Division of Cardiology University of Miami Miller School of Medicine Miami Florida
- Interdisciplinary Stem Cell Institute University of Miami Miller School of Medicine Miami Florida
| |
Collapse
|
19
|
Normand C, Kaye DM, Povsic TJ, Dickstein K. Beyond pharmacological treatment: an insight into therapies that target specific aspects of heart failure pathophysiology. Lancet 2019; 393:1045-1055. [PMID: 30860030 DOI: 10.1016/s0140-6736(18)32216-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/30/2018] [Accepted: 09/05/2018] [Indexed: 12/14/2022]
Abstract
Heart failure is a common syndrome associated with substantial morbidity and mortality. The management of symptoms and the strategies for improving prognosis have largely been based on pharmacological treatments. The pathophysiology of heart failure is complex because of the multiple causes responsible for this syndrome. This Series paper presents some examples of advances in heart failure management, in which the treatment specifically targets the underlying pathophysiological mechanisms responsible for the symptoms. These treatments include treatment of electromechanical dyssynchrony and dysrhythmia by cardiac resynchronisation and implantable cardioverter-defibrillators; neurohumoral modification by baroreflex and vagal stimulation; prevention of adverse cardiac remodelling by interatrial shunts; and finally targeting the myocardium directly by cell therapy in an attempt to regenerate new myocardial cells.
Collapse
Affiliation(s)
- Camilla Normand
- Department of Cardiology, Stavanger University Hospital, Stavanger, Norway; Institute of Internal Medicine, University of Bergen, Bergen, Norway
| | - David M Kaye
- Department of Cardiology, Alfred Hospital, Melbourne, VIC, Australia
| | - Thomas J Povsic
- Duke Clinical Research Institute, Duke Department of Medicine, Durham, NC, USA
| | - Kenneth Dickstein
- Department of Cardiology, Stavanger University Hospital, Stavanger, Norway; Institute of Internal Medicine, University of Bergen, Bergen, Norway.
| |
Collapse
|
20
|
Rikhtegar R, Pezeshkian M, Dolati S, Safaie N, Afrasiabi Rad A, Mahdipour M, Nouri M, Jodati AR, Yousefi M. Stem cells as therapy for heart disease: iPSCs, ESCs, CSCs, and skeletal myoblasts. Biomed Pharmacother 2018; 109:304-313. [PMID: 30396088 DOI: 10.1016/j.biopha.2018.10.065] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 10/04/2018] [Accepted: 10/12/2018] [Indexed: 01/14/2023] Open
Abstract
Heart Diseases are serious and global public health concern. In spite of remarkable therapeutic developments, the prediction of patients with Heart Failure (HF) is weak, and present therapeutic attitudes do not report the fundamental problem of the cardiac tissue loss. Innovative therapies are required to reduce mortality and limit or abolish the necessity for cardiac transplantation. Stem cell-based therapies applied to the treatment of heart disease is according to the understanding that natural self-renewing procedures are inherent to the myocardium, nonetheless may not be adequate to recover the infarcted heart muscle. Following the first account of cell therapy in heart diseases, examination has kept up to rapidity; besides, several animals and human clinical trials have been conducted to preserve the capacity of numerous stem cell population in advance cardiac function and decrease infarct size. The purpose of this study was to censoriously evaluate the works performed regarding the usage of four major subgroups of stem cells, including induced Pluripotent Stem Cells (iPSC), Embryonic Stem Cells (ESCs), Cardiac Stem Cells (CDC), and Skeletal Myoblasts, in heart diseases, at the preclinical and clinical studies. Moreover, it is aimed to argue the existing disagreements, unsolved problems, and prospect directions.
Collapse
Affiliation(s)
- Reza Rikhtegar
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Pezeshkian
- Department of Cardiac Surgery, Tabriz University of Medical, Tabriz, Iran; Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Dolati
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naser Safaie
- Department of Cardiac Surgery, Tabriz University of Medical, Tabriz, Iran; Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Afrasiabi Rad
- Department of Cardiac Surgery, Tabriz University of Medical, Tabriz, Iran; Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Reza Jodati
- Department of Cardiac Surgery, Tabriz University of Medical, Tabriz, Iran; Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Gurusamy N, Alsayari A, Rajasingh S, Rajasingh J. Adult Stem Cells for Regenerative Therapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 160:1-22. [PMID: 30470288 DOI: 10.1016/bs.pmbts.2018.07.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell therapy has been identified as an effective method to regenerate damaged tissue. Adult stem cells, also known as somatic stem cells or resident stem cells, are a rare population of undifferentiated cells, located within a differentiated organ, in a specialized structure, called a niche, which maintains the microenvironments that regulate the growth and development of adult stem cells. The adult stem cells are self-renewing, clonogenic, and multipotent in nature, and their main role is to maintain the tissue homeostasis. They can be activated to proliferate and differentiate into the required type of cells, upon the loss of cells or injury to the tissue. Adult stem cells have been identified in many tissues including blood, intestine, skin, muscle, brain, and heart. Extensive preclinical and clinical studies have demonstrated the structural and functional regeneration capabilities of these adult stem cells, such as bone marrow-derived mononuclear cells, hematopoietic stem cells, mesenchymal stromal/stem cells, resident adult stem cells, induced pluripotent stem cells, and umbilical cord stem cells. In this review, we focus on the human therapies, utilizing adult stem cells for their regenerative capabilities in the treatment of cardiac, brain, pancreatic, and eye disorders.
Collapse
Affiliation(s)
- Narasimman Gurusamy
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Sheeja Rajasingh
- Department of Internal Medicine, University of Kansas Medical Center, Kansas, KS, United States
| | - Johnson Rajasingh
- Department of Internal Medicine, University of Kansas Medical Center, Kansas, KS, United States.
| |
Collapse
|
22
|
Banovic M, Pusnik-Vrckovnik M, Nakou E, Vardas P. Myocardial regeneration therapy in heart failure: Current status and future therapeutic implications in clinical practice. Int J Cardiol 2018; 260:124-130. [DOI: 10.1016/j.ijcard.2018.01.144] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 01/25/2018] [Accepted: 01/31/2018] [Indexed: 12/16/2022]
|
23
|
Sterner RM, Sterner RC, Brenes-Salazar JA, Yu Ballard AC. Cellular therapies for chronic ischemic heart failure. Hellenic J Cardiol 2018; 59:78-90. [PMID: 29355725 DOI: 10.1016/j.hjc.2018.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/10/2018] [Accepted: 01/12/2018] [Indexed: 12/16/2022] Open
Abstract
The development of stem cell therapies for chronic ischemic heart failure is highly sought after to attempt to improve morbidity and mortality of this prevalent disease. This article reviews clinical trials that investigate stem cell therapy for chronic ischemic heart failure. To generate this review article, PubMed was searched using keywords "stem cell therapy heart failure" with the article type "Clinical Trial" selected on 10/04/2016. The raw search yielded 156 articles; 53 articles were selected for inclusion in the review between the original literature search and manual research/cross-referencing. Additional reviews and original articles were also manually researched and cross-referenced. Cellular-based therapies utilizing peripheral blood progenitor cells, bone marrow cells, mesenchymal stem cells, cells of cardiac origin, and embryonic stem cells have yielded mixed results, but some studies have shown modest efficacy. Skeletal myoblasts raised concerns about safety due to arrhythmias. Optimizing cell type and delivery method will be of critical importance in enhancing efficacy of therapy within various subsets of chronic ischemic heart failure patients. Although much more work needs to be done to optimize treatment strategies, developing stem cell therapies for chronic ischemic heart failure could be of critical importance to lessen the impactful health burden that heart failure has on patients and society.
Collapse
Affiliation(s)
- Rosalie M Sterner
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| | - Robert C Sterner
- University of Wisconsin-Madison Medical Scientist Training Program, 750 Highland Avenue, Madison, WI, 53726, USA.
| | | | - Aimee C Yu Ballard
- Primary Care Internal Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
24
|
Poulin MF, Deka A, Mohamedali B, Schaer GL. Clinical Benefits of Stem Cells for Chronic Symptomatic Systolic Heart Failure: A Systematic Review of the Existing Data and Ongoing Trials. Cell Transplant 2018; 25:1911-1923. [PMID: 27349212 DOI: 10.3727/096368916x692087] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The benefits of stem cell therapy for patients with chronic symptomatic systolic heart failure due to ischemic and nonischemic cardiomyopathy (ICM and NICM, respectively) are unclear. We performed a systematic review of major published and ongoing trials of stem cell therapy for systolic heart failure and compared measured clinical outcomes for both types of cardiomyopathy. The majority of the 29 published studies demonstrated clinical benefits of autologous bone marrow-derived mesenchymal stem cells (BM-MSCs). Left ventricular ejection fraction (LVEF) was improved in the majority of trials after therapy. Cell delivery combined with coronary artery bypass grafting was associated with the greatest improvement in LVEF. Left ventricular end-systolic volume (or diameter), New York Heart Association functional classification, quality of life, and exercise capacity were also improved in most studies after cell therapy. Most ICM trials demonstrated a significant improvement in perfusion defects, infarct size, and myocardial viability. Several larger clinical trials that are in progress employ alternative delivery modes, cell types, and longer follow-up periods. Stem cells are a promising therapeutic modality for patients with heart failure due to ICM or NICM. More data are required from larger blinded trials to determine which combination of cell type and delivery mode will yield the most benefit with avoidance of harm in these patient populations.
Collapse
Affiliation(s)
- Marie-France Poulin
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Anjan Deka
- Division of Cardiology, Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Burhan Mohamedali
- Division of Cardiology, Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Gary L Schaer
- Division of Cardiology, Department of Medicine, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
25
|
Povsic TJ. Emerging Therapies for Congestive Heart Failure. Clin Pharmacol Ther 2017; 103:77-87. [DOI: 10.1002/cpt.913] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/06/2017] [Accepted: 10/06/2017] [Indexed: 01/02/2023]
Affiliation(s)
- Thomas J. Povsic
- Duke Clinical Research Institute; Duke University Medical Center; Durham North Carolina USA
| |
Collapse
|
26
|
Gwizdala A, Rozwadowska N, Kolanowski TJ, Malcher A, Cieplucha A, Perek B, Seniuk W, Straburzynska-Migaj E, Oko-Sarnowska Z, Cholewinski W, Michalak M, Grajek S, Kurpisz M. Safety, feasibility and effectiveness of first in-human administration of muscle-derived stem/progenitor cells modified with connexin-43 gene for treatment of advanced chronic heart failure. Eur J Heart Fail 2017; 19:148-157. [PMID: 28052545 DOI: 10.1002/ejhf.700] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 09/29/2016] [Accepted: 11/02/2016] [Indexed: 12/22/2022] Open
Abstract
AIMS To assess the safety and efficacy of transendocardial delivery of muscle-derived stem/progenitor cells with connexin-43 overexpression (Cx-43-MDS/PC) in advanced heart failure (HF). METHODS AND RESULTS Thirteen subjects with advanced HF, New York Heart Association (NYHA) class II-III were enrolled and treated with targeted injection of Cx-43-MDS/PCs and then monitored for at least 6 months. Overexpression of Cx43 (Cx43+) was significantly higher in all but one subject (Cx43-). Injection of MDS/PCs was associated with significant improvement of exercise capacity: NYHA (3 ± 0 vs. 1.8 ± 0.7, P = 0.003), exercise duration (388.69 ± 141.83 s vs. 462.08 ± 176.69 s, P = 0.025), peak oxygen consumption (14.38 ± 3.97 vs. 15.83 ± 3.74 ml/kg.min, P = 0.022) and oxygen pulse (10.58 ± 2.89 vs. 18.88 ± 22.63 mLO2 /heart rate, P = 0.012). Levels of BNP, left ventricular (LV) ejection fraction and LV end-diastolic volumes tended to improve. There was a significant improvement of the mean unipolar voltage amplitudes measured for the injected segments and the entire left ventricle (9.62 ± 2.64 vs. 11.62 ± 3.50 mV, P = 0.014 and 8.83 ± 2.80 vs. 10.22 ± 3.41 mV, P = 0.041, respectively). No deaths were documented, Cx43+ (n = 12) subjects presented no significant ventricular arrhythmia; one Cx43- subject suffered from ventricular tachycardia (successfully treated with amiodarone). CONCLUSIONS Injection of Cx-43-MDS/PCs in patients with severe HF led to significant improvement in exercise capacity and myocardial viability of the injected segments while inducing no significant ventricular arrhythmia. This may arise from improved electrical coupling of the injected cells and injured myocardium and thus better in-situ mechanical cooperation of both cell types. Therefore, further clinical studies with Cx43+ MDS/PCs are warranted.
Collapse
Affiliation(s)
- Adrian Gwizdala
- Poznan University of Medical Sciences, 1st Department of Cardiology, Poznan, Poland
| | - Natalia Rozwadowska
- Department of Reproductive Biology and Stem Cells, Institute of Human Genetics Polish Academy of Sciences, ul. Strzeszynska 32, 60-479, Poznan, Poland
| | - Tomasz Jan Kolanowski
- Department of Reproductive Biology and Stem Cells, Institute of Human Genetics Polish Academy of Sciences, ul. Strzeszynska 32, 60-479, Poznan, Poland
| | - Agnieszka Malcher
- Department of Reproductive Biology and Stem Cells, Institute of Human Genetics Polish Academy of Sciences, ul. Strzeszynska 32, 60-479, Poznan, Poland
| | - Aleksandra Cieplucha
- Poznan University of Medical Sciences, 1st Department of Cardiology, Poznan, Poland
| | - Bartlomiej Perek
- Poznan University of Medical Sciences, Department of Cardiac Surgery, Poznan, Poland
| | - Wojciech Seniuk
- Poznan University of Medical Sciences, 1st Department of Cardiology, Poznan, Poland
| | | | - Zofia Oko-Sarnowska
- Poznan University of Medical Sciences, 1st Department of Cardiology, Poznan, Poland
| | - Witold Cholewinski
- Greater Poland Cancer Centre, Nuclear Medicine Department, Poznan, Poland
| | - Michal Michalak
- Poznan University of Medical Sciences, Department of Statistics, Poznan, Poland
| | - Stefan Grajek
- Poznan University of Medical Sciences, 1st Department of Cardiology, Poznan, Poland
| | - Maciej Kurpisz
- Department of Reproductive Biology and Stem Cells, Institute of Human Genetics Polish Academy of Sciences, ul. Strzeszynska 32, 60-479, Poznan, Poland
| |
Collapse
|
27
|
Fernández-Avilés F, Sanz-Ruiz R, Climent AM, Badimon L, Bolli R, Charron D, Fuster V, Janssens S, Kastrup J, Kim HS, Lüscher TF, Martin JF, Menasché P, Simari RD, Stone GW, Terzic A, Willerson JT, Wu JC. Global position paper on cardiovascular regenerative medicine. Eur Heart J 2017; 38:2532-2546. [PMID: 28575280 PMCID: PMC5837698 DOI: 10.1093/eurheartj/ehx248] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/13/2017] [Accepted: 04/20/2017] [Indexed: 12/11/2022] Open
Affiliation(s)
- Francisco Fernández-Avilés
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Ricardo Sanz-Ruiz
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Andreu M Climent
- Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain
- CIBERCV, ISCIII, Madrid, Spain
| | - Lina Badimon
- CIBERCV, ISCIII, Madrid, Spain
- Cardiovascular Research Center (CSIC-ICCC), Hospital de la Santa Creu i Sant Pau (HSCSP), Barcelona, Spain
| | - Roberto Bolli
- Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville School of Medicine, Louisville, Kentucky
| | - Dominique Charron
- LabEx TRANSPLANTEX; HLA & Médecine "Jean Dausset" Laboratory Network, Hôpital Saint-Louis AP-HP, Université Paris Diderot, 75013, France
| | - Valentin Fuster
- CIBERCV, ISCIII, Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of medicine at Mount Sinai, New York, NY, USA
| | - Stefan Janssens
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Jens Kastrup
- Department of Cardiology, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Hyo-Soo Kim
- National Research Laboratory for Stem Cell Niche, Center for Medical Innovation, Seoul National University Hospital, Seoul, Korea; Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea
| | - Thomas F Lüscher
- Department of Cardiology, University Heart Center Zurich, Zurich, Switzerland; Center for Molecular Cardiology, University of Zurich, Zurich, Switzerland
| | | | - Philippe Menasché
- Department of Cardiovascular Surgery Hôpital Européen Georges Pompidou; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Robert D Simari
- School of Medicine, University of Kansas, 3901 Rainbow Boulevard, Kansas City, KS, USA
| | - Gregg W Stone
- Center for Clinical Trials, Cardiovascular Research Foundation, New York, New York; Center for Clinical Trials, NewYork-Presbyterian Hospital, Columbia University Medical Center, New York, NY, USA
| | - Andre Terzic
- Center for Regenerative Medicine, Department of Cardiovascular Diseases, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, NY, USA
| | - James T Willerson
- Department of Regenerative Medicine Research, Texas Heart Institute, Houston, TX, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Division of Cardiovascular Medicine, Department of Medicine and Department of Radiology, Stanford University School of Medicine, CA, USA
| |
Collapse
|
28
|
Bianconi V, Sahebkar A, Kovanen P, Bagaglia F, Ricciuti B, Calabrò P, Patti G, Pirro M. Endothelial and cardiac progenitor cells for cardiovascular repair: A controversial paradigm in cell therapy. Pharmacol Ther 2017; 181:156-168. [PMID: 28827151 DOI: 10.1016/j.pharmthera.2017.08.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Stem cells have the potential to differentiate into cardiovascular cell lineages and to stimulate tissue regeneration in a paracrine/autocrine manner; thus, they have been extensively studied as candidate cell sources for cardiovascular regeneration. Several preclinical and clinical studies addressing the therapeutic potential of endothelial progenitor cells (EPCs) and cardiac progenitor cells (CPCs) in cardiovascular diseases have been performed. For instance, autologous EPC transplantation and EPC mobilization through pharmacological agents contributed to vascular repair and neovascularization in different animal models of limb ischemia and myocardial infarction. Also, CPC administration and in situ stimulation of resident CPCs have been shown to improve myocardial survival and function in experimental models of ischemic heart disease. However, clinical studies using EPC- and CPC-based therapeutic approaches have produced mixed results. In this regard, intracoronary, intra-myocardial or intramuscular injection of either bone marrow-derived or peripheral blood progenitor cells has improved pathological features of tissue ischemia in humans, despite modest or no clinical benefit has been observed in most cases. Also, the intriguing scientific background surrounding the potential clinical applications of EPC capture stenting is still waiting for a confirmatory proof. Moreover, clinical findings on the efficacy of CPC-based cell therapy in heart diseases are still very preliminary and based on small-size studies. Despite promising evidence, widespread clinical application of both EPCs and CPCs remains delayed due to several unresolved issues. The present review provides a summary of the different applications of EPCs and CPCs for cardiovascular cell therapy and underlies their advantages and limitations.
Collapse
Affiliation(s)
- Vanessa Bianconi
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Francesco Bagaglia
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy
| | - Biagio Ricciuti
- Department of Medical Oncology, S. Maria della Misericordia Hospital, Perugia, Italy
| | - Paolo Calabrò
- Division of Cardiology, Second University of Naples, Department of Cardio-Thoracic and Respiratory Sciences, Italy
| | - Giuseppe Patti
- Unit of Cardiovascular Science, Campus Bio-Medico University of Rome, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Perugia, Italy.
| |
Collapse
|
29
|
Psaltis PJ, Schwarz N, Toledo-Flores D, Nicholls SJ. Cellular Therapy for Heart Failure. Curr Cardiol Rev 2016; 12:195-215. [PMID: 27280304 PMCID: PMC5011188 DOI: 10.2174/1573403x12666160606121858] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 12/18/2015] [Accepted: 12/31/1969] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis of cardiomyopathy and heart failure (HF) is underpinned by complex changes at subcellular, cellular and extracellular levels in the ventricular myocardium. For all of the gains that conventional treatments for HF have brought to mortality and morbidity, they do not adequately address the loss of cardiomyocyte numbers in the remodeling ventricle. Originally conceived to address this problem, cellular transplantation for HF has already gone through several stages of evolution over the past two decades. Various cell types and delivery routes have been implemented to positive effect in preclinical models of ischemic and nonischemic cardiomyopathy, with pleiotropic benefits observed in terms of myocardial remodeling, systolic and diastolic performance, perfusion, fibrosis, inflammation, metabolism and electrophysiology. To a large extent, these salubrious effects are now attributed to the indirect, paracrine capacity of transplanted stem cells to facilitate endogenous cardiac repair processes. Promising results have also followed in early phase human studies, although these have been relatively modest and somewhat inconsistent. This review details the preclinical and clinical evidence currently available regarding the use of pluripotent stem cells and adult-derived progenitor cells for cardiomyopathy and HF. It outlines the important lessons that have been learned to this point in time, and balances the promise of this exciting field against the key challenges and questions that still need to be addressed at all levels of research, to ensure that cell therapy realizes its full potential by adding to the armamentarium of HF management.
Collapse
Affiliation(s)
- Peter J Psaltis
- Co-Director of Vascular Research Centre, Heart Health Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia 5000.
| | | | | | | |
Collapse
|
30
|
Abstract
Improvements in the care of patients with ischemic cardiovascular disease have led to improved survival but also a burgeoning population of patients with advanced ischemic heart disease. Cell therapies offer a novel approach toward cardiac "rejuvenation" via stimulation of new blood vessel growth, enhancing tissue perfusion, and via preservation or even regeneration of myocardial tissue, leading to improvements in cardiac performance after myocardial infarction and in patients with advanced heart failure. Here, we summarize and offer some thoughts on the state of the field of cell therapy for ischemic heart disease, targeting three separate conditions that have been the subject of significant clinical research: enhancing left ventricular recovery after MI, improving outcomes and symptoms in patients with congestive heart failure (CHF), and treatment of patients with refractory angina, despite maximal medical therapy.
Collapse
Affiliation(s)
- Thomas J Povsic
- Duke Clinical Research Institute and Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC, 27708, USA.
| |
Collapse
|
31
|
Shadrin IY, Khodabukus A, Bursac N. Striated muscle function, regeneration, and repair. Cell Mol Life Sci 2016; 73:4175-4202. [PMID: 27271751 PMCID: PMC5056123 DOI: 10.1007/s00018-016-2285-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/20/2016] [Accepted: 05/26/2016] [Indexed: 12/18/2022]
Abstract
As the only striated muscle tissues in the body, skeletal and cardiac muscle share numerous structural and functional characteristics, while exhibiting vastly different size and regenerative potential. Healthy skeletal muscle harbors a robust regenerative response that becomes inadequate after large muscle loss or in degenerative pathologies and aging. In contrast, the mammalian heart loses its regenerative capacity shortly after birth, leaving it susceptible to permanent damage by acute injury or chronic disease. In this review, we compare and contrast the physiology and regenerative potential of native skeletal and cardiac muscles, mechanisms underlying striated muscle dysfunction, and bioengineering strategies to treat muscle disorders. We focus on different sources for cellular therapy, biomaterials to augment the endogenous regenerative response, and progress in engineering and application of mature striated muscle tissues in vitro and in vivo. Finally, we discuss the challenges and perspectives in translating muscle bioengineering strategies to clinical practice.
Collapse
Affiliation(s)
- I Y Shadrin
- Department of Biomedical Engineering, Duke University, 3000 Science Drive, Hudson Hall 136, Durham, NC, 27708-90281, USA
| | - A Khodabukus
- Department of Biomedical Engineering, Duke University, 3000 Science Drive, Hudson Hall 136, Durham, NC, 27708-90281, USA
| | - N Bursac
- Department of Biomedical Engineering, Duke University, 3000 Science Drive, Hudson Hall 136, Durham, NC, 27708-90281, USA.
| |
Collapse
|
32
|
Ghafarzadeh M, Namdari M, Eatemadi A. Stem cell therapies for congenital heart disease. Biomed Pharmacother 2016; 84:1163-1171. [PMID: 27780147 DOI: 10.1016/j.biopha.2016.10.055] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/16/2016] [Accepted: 10/17/2016] [Indexed: 01/15/2023] Open
Abstract
Congenital heart disease (CHD) is the most prevalent congenital anomaly in newborn babies. Cardiac malformations have been induced in different animal model experiments, by perturbing some molecules that take part in the developmental pathways associated with myocyte differentiation, specification, or cardiac morphogenesis. The exact epigenetic, environmental, or genetic, basis for these molecules perturbations is yet to be understood. But, scientist have bridged this gap by introducing autologous stem cell into the defective hearts to treat CHD. The choice of stem cells to use has also raised an issue. In this review, we explore different stem cells that have been recently used, as an update into the pool of this knowledge and we suggested the future perspective into the choice of stem cells to control this disease. We propose that isolating mesenchymal stem cells from neonate will give a robust heart regeneration as compared to adults. This source are easily isolated. To unveil stem cell therapy beyond its possibility and safety, further study is required, including largescale randomized, and clinical trials to certify the efficacy of stem cell therapy.
Collapse
Affiliation(s)
- Masoumeh Ghafarzadeh
- Assalian Hospital, Center for Obstetrics and Gynecology, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Mehrdad Namdari
- Department of Cardiology, Lorestan University of Medical Sciences, Postal address: 6997118544, Khoramabad, Iran.
| | - Ali Eatemadi
- Department of Medical Biotechnology, School of advance Science in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Lorestan, Iran
| |
Collapse
|
33
|
Maulik SK, Wilson V, Seth S, Bhargava B, Dua P, Ramakrishnan S, Katiyar CK. Clinical efficacy of water extract of stem bark of Terminalia arjuna (Roxb. ex DC.) Wight & Arn. in patients of chronic heart failure: a double-blind, randomized controlled trial. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:1211-1219. [PMID: 26988798 DOI: 10.1016/j.phymed.2016.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 06/05/2023]
Abstract
BACKGROUND The stem bark of Terminalia arjuna (Roxb. ex DC.) Wight and Arn. (Arjuna) is used in Indian system of medicine (Ayurveda) for treatment of various cardiac diseases, including heart failure. However, well designed clinical trials exploring its efficacy and safety in chronic heart failure (CHF) are lacking. PURPOSE To ascertain the add-on efficacy and safety of a standardized water extract of stem bark of Arjuna (Arjuna extract) in CHF patients on standard pharmacotherapy. STUDY DESIGN Double-blind, parallel, randomized, placebo-controlled add-on clinical trial. METHODS After approval of institutional ethics committee, 100 patients of CHF of New York Heart Association (NYHA) functional class II on standard pharmacotherapy having an echocardiographic left ventricular ejection fraction (LVEF) ≤ 40% were consecutively recruited with informed consent and randomized 1:1 to Arjuna extract 750 mg or matching placebo twice daily. The primary outcome measure was change in LVEF at 12 weeks. Secondary outcome measures included changes in (i) NYHA functional class, (ii) distance covered in 6 min walk test (6MWT), (iii) quality of life (QoL), as determined by the Kansas City Cardiomyopathy Questionnaire (KCCQ), (iv) plasma brain natriuretic peptide, (v) plasma cytokines (interleukin-6, high sensitivity C-reactive protein and tumour necrosis factor-α) and (vi) oxidative stress markers [serum thiobarbituric acid reactive substances (TBARS), red blood cell (RBC) superoxide dismutase (SOD), RBC catalase and RBC glutathione (GSH)] at 6 and 12 weeks. Safety assessment was done by adverse event monitoring and laboratory investigations. Results were expressed as mean ± SD or median (interquartile range) and analysed with intention-to- treat principle using appropriate two-sided statistical tests. A p-value < 0.05 was considered significant. RESULTS Arjuna extract was well-tolerated, but did not change LVEF (24.3 ± 7.1 versus 25.5 ± 7.7%; p = 0.4) or secondary outcome measures except preservation of RBC catalase activity [1275(104, 10350) versus 1243.5(104, 10350) U/g haemoglobin; p = 0.01] compared to placebo. Significantly greater percentage increases occurred in distance covered in 6 MWT, RBC-SOD, RBC catalase, RBC GSH and in symptom severity and stability domains of KCCQ in patients on Arjuna extract versus those on placebo, on a post-hoc analysis, between subgroups of patients who improved in these outcomes. CONCLUSION Arjuna extract did not improve LVEF in CHF patients over 12 weeks, although there was improvement in functional capacity, antioxidant reserves and symptom-related QoL domains in some patients.
Collapse
Affiliation(s)
- Subir K Maulik
- Department of Pharmacology, All India Institute of Medical Sciences (A.I.I.M.S.), Ansari Nagar, 110029 New Delhi, India..
| | - Vinu Wilson
- Department of Pharmacology, All India Institute of Medical Sciences (A.I.I.M.S.), Ansari Nagar, 110029 New Delhi, India.; Department of Pharmacology, Sree Gokulam Medical College & Research Foundation, Venjaramoodu, Trivandrum, 695607 Kerala, India
| | - Sandeep Seth
- Department of Cardiology, All India Institute of Medical Sciences (A.I.I.M.S.), Ansari Nagar, 110029 New Delhi, India
| | - Balram Bhargava
- Department of Cardiology, All India Institute of Medical Sciences (A.I.I.M.S.), Ansari Nagar, 110029 New Delhi, India
| | - Pamila Dua
- Department of Pharmacology, All India Institute of Medical Sciences (A.I.I.M.S.), Ansari Nagar, 110029 New Delhi, India
| | - Sivasubramanian Ramakrishnan
- Department of Cardiology, All India Institute of Medical Sciences (A.I.I.M.S.), Ansari Nagar, 110029 New Delhi, India
| | - Chandra K Katiyar
- Dabur India Limited, Ghaziabad, 201010 Uttar Pradesh, India.; Emami Limited, Kolkata, 700107 West Bengal, India
| |
Collapse
|
34
|
Feasibility study of particulate extracellular matrix (P-ECM) and left ventricular assist device (HVAD) therapy in chronic ischemic heart failure bovine model. ASAIO J 2016; 61:161-9. [PMID: 25423120 DOI: 10.1097/mat.0000000000000178] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Myocardial recovery with left ventricular assist device (LVAD) support is uncommon and unpredictable. We tested the hypothesis that injectable particulate extracellular matrix (P-ECM) with LVAD support promotes cell proliferation and improves cardiac function. LVAD, P-ECM, and P-ECM + LVAD therapies were investigated in chronic ischemic heart failure (IHF) calves induced using coronary embolization. Particulate extracellular matrix emulsion (CorMatrix, Roswell, GA) was injected intramyocardially using a 7 needle pneumatic delivery tool. Left ventricular assist devices (HVAD, HeartWare) were implanted in a left ventricle (LV) apex to proximal descending aorta configuration. Cell proliferation was identified using BrdU (5 mg/kg) injections over the last 45 treatment days. Echocardiography was performed weekly. End-organ regional blood flow (RBF) was quantified at study endpoints using fluorescently labeled microspheres. Before treatment, IHF calves had an ejection fraction (EF) of 33 ± 2% and left ventricular end-diastolic volume of 214 ± 18 ml with cardiac cachexia (0.69 ± 0.06 kg/day). Healthy weight gain was restored in all groups (0.89 ± 0.03 kg/day). EF increased with P-ECM + HVAD from 36 ± 5% to 75 ± 2%, HVAD 38 ± 4% to 58 ± 5%, and P-ECM 27 ± 1% to 66 ± 6%. P-ECM + HVAD demonstrated the largest increase in cell proliferation and end-organ RBF. This study demonstrates the feasibility of combined LVAD support with P-ECM injection to stimulate new cell proliferation and improve cardiac function, which warrants further investigation.
Collapse
|
35
|
Takanari H, Miwa K, Fu X, Nakai J, Ito A, Ino K, Honda H, Tonomura W, Konishi S, Opthof T, van der Heyden MA, Kodama I, Lee JK. A New In Vitro Co-Culture Model Using Magnetic Force-Based Nanotechnology. J Cell Physiol 2016; 231:2249-56. [PMID: 26873862 DOI: 10.1002/jcp.25342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 02/10/2016] [Indexed: 11/07/2022]
Abstract
Skeletal myoblast (SkMB) transplantation has been conducted as a therapeutic strategy for severe heart failure. However, arrhythmogenicity following transplantation remains unsolved. We developed an in vitro model of myoblast transplantation with "patterned" or "randomly-mixed" co-culture of SkMBs and cardiomyocytes enabling subsequent electrophysiological, and arrhythmogenic evaluation. SkMBs were magnetically labeled with magnetite nanoparticles and co-cultured with neonatal rat ventricular myocytes (NRVMs) on multi-electrode arrays. SkMBs were patterned by a magnet beneath the arrays. Excitation synchronicity was evaluated by Ca(2+) imaging using a gene-encoded Ca(2+) indicator, G-CaMP2. In the monoculture of NRVMs (control), conduction was well-organized. In the randomly-mixed co-culture of NRVMs and SkMBs (random group), there was inhomogeneous conduction from multiple origins. In the "patterned" co-culture where an en bloc SKMB-layer was inserted into the NRVM-layer, excitation homogenously propagated although conduction was distorted by the SkMB-area. The 4-mm distance conduction time (CT) in the random group was significantly longer (197 ± 126 ms) than in control (17 ± 3 ms). In the patterned group, CT through NRVM-area did not change (25 ± 3 ms), although CT through the SkMB-area was significantly longer (132 ± 77 ms). The intervals between spontaneous excitation varied beat-to-beat in the random group, while regular beating was recorded in the control and patterned groups. Synchronized Ca(2+) transients of NRVMs were observed in the patterned group, whereas those in the random group were asynchronous. Patterned alignment of SkMBs is feasible with magnetic nanoparticles. Using the novel in vitro model mimicking cell transplantation, it may become possible to predict arrhythmogenicity due to heterogenous cell transplantation. J. Cell. Physiol. 231: 2249-2256, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hiroki Takanari
- Department of Pathophysiology, Oita University School of Medicine, Oita, Japan.,Department of Cardiovascular Research, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Keiko Miwa
- Department of Cardiovascular Research, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - XianMing Fu
- Department of Cardiovascular Research, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Cardiothoracic Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Department of Cardiovascular Surgery, Central South University The Second Xiangya Hospital, Changsha, China
| | - Junichi Nakai
- Saitama University Brain Science Institute, Saitama, Japan
| | - Akira Ito
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Kousuke Ino
- Department of Biotechnology, School of Engineering, Nagoya University, Nagoya, Japan
| | - Hiroyuki Honda
- Department of Biotechnology, School of Engineering, Nagoya University, Nagoya, Japan
| | - Wataru Tonomura
- Department of Mechanical Engineering, Ritsumeikan University, Kusatsu, Japan
| | - Satoshi Konishi
- Department of Mechanical Engineering, Ritsumeikan University, Kusatsu, Japan
| | - Tobias Opthof
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands.,Experimental Cardiology Group, Center for Heart Failure Research, Academic Medical Center, Amsterdam, The Netherlands
| | - Marcel Ag van der Heyden
- Division of Heart and Lungs, Department of Medical Physiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Jong-Kook Lee
- Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
36
|
Sawa Y, Yoshikawa Y, Toda K, Fukushima S, Yamazaki K, Ono M, Sakata Y, Hagiwara N, Kinugawa K, Miyagawa S. Safety and Efficacy of Autologous Skeletal Myoblast Sheets (TCD-51073) for the Treatment of Severe Chronic Heart Failure Due to Ischemic Heart Disease. Circ J 2016; 79:991-9. [PMID: 25912561 DOI: 10.1253/circj.cj-15-0243] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Poor survival outcomes for patients with severe heart failure (HF) and the donor shortage for heart transplantation warrant the development of myocardial regenerative therapy. We performed a multicenter, phase II study to evaluate the safety and efficacy of autologous skeletal myoblast sheets (TCD-51073). METHODS AND RESULTS In 3 study sites, we enrolled 7 patients with severe chronic HF due to ischemic heart disease despite maximal therapy, all of whom underwent transplantation of TCD-51073. No serious arrhythmia was reported, and no changes were noted in the frequency of ventricular extrasystole frequency. The primary efficacy endpoint of the change in left ventricular ejection fraction (LVEF) on gated blood-pool scintigraphy at 26 weeks after transplantation showed that 5 subjects were responders (classified as "improved" or "unchanged"). In addition, LVEF on echocardiography improved over time, with a change in LVEF of 7.1±2.8% at 26 weeks posttransplantation. Among the 7 subjects, 6 showed improvement in New York Heart Association functional class by at least 1 class. The 6-min walk distance was 410.1±136.1 m before transplantation and 455.4±103.7 m at 26 weeks after transplantation. CONCLUSIONS This study demonstrated the feasibility and safety of the transplantation of TCD-51073 in the patients with severe chronic HF due to ischemic heart disease, suggesting that TCD-51073 might maintain or improve cardiac function, symptoms, and physical function.
Collapse
Affiliation(s)
- Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW The aim of this review was to discuss recent advances in clinical aspects of stem cell therapy in heart failure with emphasis on patient selection, stem cell types and delivery methods. RECENT FINDINGS Several stem cell types have been considered for the treatment of patients with heart failure. In nonischemic heart failure, transplantation of CD34 cells improved myocardial performance, functional capacity and neurohumoral activation. In ischemic heart failure, cardiosphere-derived cells were shown to reduce myocardial scar burden with concomitant increase in viable tissue and regional systolic wall thickening. Both autologous and allogeneic mesenchymal stem cells were shown to be effective in improving heart function in patients with ischemic heart failure; this may represent an important step toward the development of a standardized stem cell product for widespread clinical use. SUMMARY Although trials of stem cell therapy in heart failure have shown promising results, the findings are not consistent. Given the wide spectrum of heart failure, it may be difficult to define a uniform stem cell therapy for all subsets of patients; instead, future stem cell therapeutic strategies should aim for a more personalized approach by establishing optimal stem cell type, dose and delivery method for an individual patient and disease state.
Collapse
|
38
|
Banovic M, Loncar Z, Behfar A, Vanderheyden M, Beleslin B, Zeiher A, Metra M, Terzic A, Bartunek J. Endpoints in stem cell trials in ischemic heart failure. Stem Cell Res Ther 2015; 6:159. [PMID: 26319401 PMCID: PMC4552990 DOI: 10.1186/s13287-015-0143-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Despite multimodal regimens and diverse treatment options alleviating disease symptoms, morbidity and mortality associated with advanced ischemic heart failure remain high. Recently, technological innovation has led to the development of regenerative therapeutic interventions aimed at halting or reversing the vicious cycle of heart failure progression. Driven by the unmet patient need and fueled by encouraging experimental studies, stem cell-based clinical trials have been launched over the past decade. Collectively, these trials have enrolled several thousand patients and demonstrated the clinical feasibility and safety of cell-based interventions. However, the totality of evidence supporting their efficacy in ischemic heart failure remains limited. Experience from the early randomized stem cell clinical trials underscores the key points in trial design ranging from adequate hypothesis formulation to selection of the optimal patient population, cell type and delivery route. Importantly, to translate the unprecedented promise of regenerative biotherapies into clinical benefit, it is crucial to ensure the appropriate choice of endpoints along the regulatory path. Accordingly, we here provide considerations relevant to the choice of endpoints for regenerative clinical trials in the ischemic heart failure setting.
Collapse
Affiliation(s)
- Marko Banovic
- Cardiology Department, University Clinical Center of Serbia, Belgrade Medical School, 11000, Belgrade, Serbia.
| | - Zlatibor Loncar
- Cardiology Department, University Clinical Center of Serbia, Belgrade Medical School, 11000, Belgrade, Serbia.
| | | | | | - Branko Beleslin
- Cardiology Department, University Clinical Center of Serbia, Belgrade Medical School, 11000, Belgrade, Serbia.
| | - Andreas Zeiher
- Cardiology Department, Goethe University of Frankfurt, 60590, Frankfurt, Germany.
| | - Marco Metra
- Cardiology Department, University of Brescia, 25123, Brescia, Italy.
| | | | - Jozef Bartunek
- Cardiovascular Center, OLV Hospital, 9300, Aalst, Belgium.
| |
Collapse
|
39
|
Hayashi E, Hosoda T. Myocyte renewal and therapeutic myocardial regeneration using various progenitor cells. Heart Fail Rev 2015; 19:789-97. [PMID: 24743881 DOI: 10.1007/s10741-014-9430-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Whereas the demand on effective treatment options for chronic heart failure is dramatically increasing, the recent recognition of physiological and pathological myocyte turnover in the adult human heart provided a fundamental basis for the therapeutic regeneration. Divergent modalities were experimentally introduced to this field, and selected ones have been applied clinically; the history began with skeletal myoblasts and bone-marrow-derived cells, and lately mesenchymal stem/stromal cells and resident cardiac cells joined the repertoire. Among them, autologous transplantation of c-kit-positive cardiac stem cells in patients with chronic ventricular dysfunction resulted in an outstanding outcome with long-lasting effects without increasing major adverse events. To further optimize currently available approaches, we have to consider multiple factors, such as the targeting disease, the cell population and number to be administered, and the timing and the route of cell delivery. Exploration of the consequence of the previous clinical trials would allow us to envision an ideal cellular therapy for various cardiovascular disorders.
Collapse
Affiliation(s)
- Emiko Hayashi
- Tokai University Institute of Innovative Science and Technology, 143 Shimokasuya, Isehara, 259-1193, Kanagawa, Japan
| | | |
Collapse
|
40
|
Alrefai MT, Murali D, Paul A, Ridwan KM, Connell JM, Shum-Tim D. Cardiac tissue engineering and regeneration using cell-based therapy. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2015; 8:81-101. [PMID: 25999743 PMCID: PMC4437607 DOI: 10.2147/sccaa.s54204] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Stem cell therapy and tissue engineering represent a forefront of current research in the treatment of heart disease. With these technologies, advancements are being made into therapies for acute ischemic myocardial injury and chronic, otherwise nonreversible, myocardial failure. The current clinical management of cardiac ischemia deals with reestablishing perfusion to the heart but not dealing with the irreversible damage caused by the occlusion or stenosis of the supplying vessels. The applications of these new technologies are not yet fully established as part of the management of cardiac diseases but will become so in the near future. The discussion presented here reviews some of the pioneering works at this new frontier. Key results of allogeneic and autologous stem cell trials are presented, including the use of embryonic, bone marrow-derived, adipose-derived, and resident cardiac stem cells.
Collapse
Affiliation(s)
- Mohammad T Alrefai
- Division of Cardiac Surgery, McGill University Health Center, Montreal, QC, Canada ; Division of Surgical Research, McGill University Health Center, Montreal, QC, Canada ; King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| | - Divya Murali
- Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, KS, USA
| | - Arghya Paul
- Department of Chemical and Petroleum Engineering, School of Engineering, University of Kansas, Lawrence, KS, USA
| | - Khalid M Ridwan
- Division of Cardiac Surgery, McGill University Health Center, Montreal, QC, Canada ; Division of Surgical Research, McGill University Health Center, Montreal, QC, Canada
| | - John M Connell
- Division of Cardiac Surgery, McGill University Health Center, Montreal, QC, Canada ; Division of Surgical Research, McGill University Health Center, Montreal, QC, Canada
| | - Dominique Shum-Tim
- Division of Cardiac Surgery, McGill University Health Center, Montreal, QC, Canada ; Division of Surgical Research, McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
41
|
Fisher SA, Doree C, Mathur A, Martin-Rendon E. Meta-Analysis of Cell Therapy Trials for Patients With Heart Failure. Circ Res 2015; 116:1361-77. [DOI: 10.1161/circresaha.116.304386] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 01/20/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Sheila A Fisher
- From the Systematic Review Group, R&D Department, NHS Blood and Transplant, Oxford, UK (S.A.F., C.D.); Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK (S.A.F., C.D., E.M.-R.); Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK (A.M.); and Stem Cell Research Laboratory, R&D Department, NHS Blood and Transplant, Oxford, UK (E.M.-R.)
| | - Carolyn Doree
- From the Systematic Review Group, R&D Department, NHS Blood and Transplant, Oxford, UK (S.A.F., C.D.); Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK (S.A.F., C.D., E.M.-R.); Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK (A.M.); and Stem Cell Research Laboratory, R&D Department, NHS Blood and Transplant, Oxford, UK (E.M.-R.)
| | - Anthony Mathur
- From the Systematic Review Group, R&D Department, NHS Blood and Transplant, Oxford, UK (S.A.F., C.D.); Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK (S.A.F., C.D., E.M.-R.); Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK (A.M.); and Stem Cell Research Laboratory, R&D Department, NHS Blood and Transplant, Oxford, UK (E.M.-R.)
| | - Enca Martin-Rendon
- From the Systematic Review Group, R&D Department, NHS Blood and Transplant, Oxford, UK (S.A.F., C.D.); Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK (S.A.F., C.D., E.M.-R.); Department of Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK (A.M.); and Stem Cell Research Laboratory, R&D Department, NHS Blood and Transplant, Oxford, UK (E.M.-R.)
| |
Collapse
|
42
|
Young PP, Schäfer R. Cell-based therapies for cardiac disease: a cellular therapist's perspective. Transfusion 2015; 55:441-51; quiz 440. [PMID: 25145464 PMCID: PMC5771653 DOI: 10.1111/trf.12826] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 07/14/2014] [Accepted: 07/14/2014] [Indexed: 12/14/2022]
Abstract
Cell-based therapy is an exciting, promising, and a developing new treatment for cardiac diseases. Stem cell-based therapies have the potential to fundamentally transform the treatment of ischemic cardiac injury and heart failure by achieving what would have been unthinkable only a few years ago-the Holy Grail of myocardial regeneration. Recent therapeutic approaches involve bone marrow (BM)-derived mononuclear cells and their subsets such as mesenchymal stem/stromal cells (MSCs), endothelial progenitor cells as well as adipose tissue-derived MSCs, cardiac tissue-derived stem cells, and cell combinations. Clinical trials employing these cells have demonstrated that cellular therapy is feasible and safe. Regarding delivery methods, the safety of catheter-based, transendocardial and -epicardial stem cell injection has been established. However, the results, while variable, suggest rather modest clinical efficacy overall in both heart failure and ischemic heart disease, such as in acute myocardial infarction. Future studies will focus on determining the most efficacious cell type(s) and/or cell combinations and the most reasonable indications and optimal timing of transplantation, as well as the mechanisms underlying their therapeutic effects. We will review and summarize the clinical trial results to date. In addition, we discuss challenges and operational issues in cell processing for cardiac applications.
Collapse
Affiliation(s)
- Pampee P. Young
- Department of Pathology, Microbiology and Immunology, Nashville, Tennessee
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Veterans Affairs, Nashville, Tennessee
| | - Richard Schäfer
- Institute for Transfusion Medicine and Immunohematology, German Red Cross Blood Donor Service Baden-Württemberg-Hessen gGmbH, Johann-Wolfgang-Goethe-University Hospital, Frankfurt am Main, Germany
| |
Collapse
|
43
|
Angelini G, Caputo M, Madeddu P. Extending flaps lifts an infarcted heart toward repair. Mol Ther 2015; 23:223-5. [PMID: 25633173 DOI: 10.1038/mt.2014.252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Gianni Angelini
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, United Kingdom
| | - Massimo Caputo
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, United Kingdom
| | - Paolo Madeddu
- Bristol Heart Institute, School of Clinical Sciences, University of Bristol, United Kingdom
| |
Collapse
|
44
|
Reinhardt M, Bader A, Giri S. Devices for stem cell isolation and delivery: current need for drug discovery and cell therapy. Expert Rev Med Devices 2014; 12:353-64. [DOI: 10.1586/17434440.2015.995094] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
45
|
Squiers JJ, Hutcheson KA, Thatcher JE, DiMaio JM. Cardiac stem cell therapy: checkered past, promising future? J Thorac Cardiovasc Surg 2014; 148:3188-93. [PMID: 25433891 DOI: 10.1016/j.jtcvs.2014.10.077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 10/17/2014] [Indexed: 12/29/2022]
Affiliation(s)
- John J Squiers
- University of Texas Southwestern Medical Center, Dallas, Tex
| | | | | | - J Michael DiMaio
- Spectral MD, Dallas, Tex; Baylor University Medical Center, Dallas, Tex.
| |
Collapse
|
46
|
Dauwe DF, Nuyens D, De Buck S, Claus P, Gheysens O, Koole M, Coudyzer W, Vanden Driessche N, Janssens L, Ector J, Dymarkowski S, Bogaert J, Heidbuchel H, Janssens S. Three-dimensional rotational angiography fused with multimodal imaging modalities for targeted endomyocardial injections in the ischaemic heart. Eur Heart J Cardiovasc Imaging 2014; 15:900-7. [PMID: 24596052 DOI: 10.1093/ehjci/jeu019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM Biological therapies for ischaemic heart disease require efficient, safe, and affordable intramyocardial delivery. Integration of multiple imaging modalities within the fluoroscopy framework can provide valuable information to guide these procedures. We compared an anatomo-electric method (LARCA) with a non-fluoroscopic electromechanical mapping system (NOGA(®)). LARCA integrates selective three-dimensional-rotational angiograms with biplane fluoroscopy. To identify the infarct region, we studied LARCA-fusion with pre-procedural magnetic resonance imaging (MRI), dedicated CT, or (18)F-FDG-PET/CT. METHODS AND RESULTS We induced myocardial infarction in 20 pigs by 90-min LAD occlusion. Six weeks later, we compared peri-infarct delivery accuracy of coloured fluospheres using sequential NOGA(®)- and LARCA-MRI-guided vs. LARCA-CT- and LARCA-(18)F-FDG-PET/CT-guided intramyocardial injections. MRI after 6 weeks revealed significant left ventricular (LV) functional impairment and remodelling (LVEF 31 ± 3%, LVEDV 178 ± 15 mL, infarct size 17 ± 2% LV mass). During NOGA(®)-procedures, three of five animals required DC-shock for major ventricular arrhythmias vs. one of ten during LARCA-procedures. Online procedure time was shorter for LARCA than NOGA(®) (77 ± 6 vs. 130 ± 3 min, P < 0.0001). Absolute distance of injection spots to the infarct border was similar for LARCA-MRI (4.8 ± 0.5 mm) and NOGA(®) (5.4 ± 0.5 mm). LARCA-CT-integration allowed closer approximation of the targeted border zone than LARCA-PET (4.0 ± 0.5 mm vs. 6.2 ± 0.6 mm, P < 0.05). CONCLUSION Three-dimensional -rotational angiography fused with multimodal imaging offers a new, cost-effective, and safe strategy to guide intramyocardial injections. Endoventricular procedure times and arrhythmias compare favourably to NOGA(®), without compromising injection accuracy. LARCA-based fusion imaging is a promising enabling technology for cardiac biological therapies.
Collapse
Affiliation(s)
- Dieter Frans Dauwe
- Department of Cardiovascular Sciences, Clinical Cardiology, KULeuven, Leuven, Belgium
| | - Dieter Nuyens
- Department of Cardiovascular Sciences, Clinical Cardiology, KULeuven, Leuven, Belgium
| | - Stijn De Buck
- Department of Cardiovascular Sciences, Clinical Cardiology, KULeuven, Leuven, Belgium
| | - Piet Claus
- Department of Cardiovascular Sciences, Imaging and Cardiovascular Dynamics, KULeuven, Leuven, Belgium
| | - Olivier Gheysens
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KULeuven, Leuven, Belgium
| | - Michel Koole
- Department of Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KULeuven, Leuven, Belgium
| | - Walter Coudyzer
- Clinical Radiology, Gasthuisberg University Hospitals Leuven, Leuven, Belgium
| | - Nina Vanden Driessche
- Department of Cardiovascular Sciences, Clinical Cardiology, KULeuven, Leuven, Belgium
| | - Laurens Janssens
- Department of Cardiovascular Sciences, Clinical Cardiology, KULeuven, Leuven, Belgium
| | - Joris Ector
- Department of Cardiovascular Sciences, Clinical Cardiology, KULeuven, Leuven, Belgium
| | - Steven Dymarkowski
- Clinical Radiology, Gasthuisberg University Hospitals Leuven, Leuven, Belgium Department of Imaging and Pathology, Translational MRI, KULeuven, Leuven, Belgium
| | - Jan Bogaert
- Clinical Radiology, Gasthuisberg University Hospitals Leuven, Leuven, Belgium Department of Imaging and Pathology, Translational MRI, KULeuven, Leuven, Belgium
| | - Hein Heidbuchel
- Department of Cardiovascular Sciences, Clinical Cardiology, KULeuven, Leuven, Belgium
| | - Stefan Janssens
- Department of Cardiovascular Sciences, Clinical Cardiology, KULeuven, Leuven, Belgium
| |
Collapse
|
47
|
Malliaras K, Marbán E. Moving beyond surrogate endpoints in cell therapy trials for heart disease. Stem Cells Transl Med 2014; 3:2-6. [PMID: 24292794 PMCID: PMC3902289 DOI: 10.5966/sctm.2013-0104] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/16/2013] [Indexed: 12/19/2022] Open
Abstract
Cell therapy for heart disease began clinically more than a decade ago. Since then, numerous trials have been performed, but the studies have been underpowered, focusing primarily on low-risk patients with a recent myocardial infarction. Many data have accumulated on surrogate endpoints such as ejection fraction, but few clinical conclusions can be drawn from such studies. We argue here that the time is right for targeting larger and/or higher-risk populations for whom there is some expectation of being able to influence mortality or rehospitalization.
Collapse
|
48
|
Hassan N, Tchao J, Tobita K. Concise review: skeletal muscle stem cells and cardiac lineage: potential for heart repair. Stem Cells Transl Med 2013; 3:183-93. [PMID: 24371329 DOI: 10.5966/sctm.2013-0122] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Valuable and ample resources have been spent over the last two decades in pursuit of interventional strategies to treat the unmet demand of heart failure patients to restore myocardial structure and function. At present, it is clear that full restoration of myocardial structure and function is outside our reach from both clinical and basic research studies, but it may be achievable with a combination of ongoing research, creativity, and perseverance. Since the 1990s, skeletal myoblasts have been extensively investigated for cardiac cell therapy of congestive heart failure. Whereas the Myoblast Autologous Grafting in Ischemic Cardiomyopathy (MAGIC) trial revealed that transplanted skeletal myoblasts did not integrate into the host myocardium and also did not transdifferentiate into cardiomyocytes despite some beneficial effects on recipient myocardial function, recent studies suggest that skeletal muscle-derived stem cells have the ability to adopt a cardiomyocyte phenotype in vitro and in vivo. This brief review endeavors to summarize the importance of skeletal muscle stem cells and how they can play a key role to surpass current results in the future and enhance the efficacious implementation of regenerative cell therapy for heart failure.
Collapse
Affiliation(s)
- Narmeen Hassan
- Department of Developmental Biology, Department of Bioengineering, and McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
49
|
Mathiasen AB, Haack-Sørensen M, Jørgensen E, Kastrup J. Autotransplantation of mesenchymal stromal cells from bone-marrow to heart in patients with severe stable coronary artery disease and refractory angina--final 3-year follow-up. Int J Cardiol 2013; 170:246-51. [PMID: 24211066 DOI: 10.1016/j.ijcard.2013.10.079] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 10/21/2013] [Indexed: 01/16/2023]
Abstract
BACKGROUND The study assessed long-term safety and efficacy of intramyocardial injection of autologous bone-marrow derived mesenchymal stromal cells (BMMSCs) in patients with severe stable coronary artery disease (CAD) and refractory angina. METHODS Thirty-one patients with severe stable CAD and refractory angina were included. Patients had reversible myocardial ischemia and no further revascularization options. Autologous BMMSCs were isolated, culture expanded and stimulated with vascular endothelial growth-factor to facilitate endothelial differentiation. BMMSCs were injected into an ischemic, viable region of the myocardium. Patients were followed for 3 years. RESULTS We found significant clinical improvements in exercise time (p=0.0016), angina class (CCS) (p<0.0001), weekly number of angina attacks (p<0.0001) and use of nitroglycerine from (p=0.0017). In the Seattle Angina Questionnaire there were significant improvements in physical limitation score, angina stability score, angina frequency score and quality of life score (all p<0.0001). When comparing all hospital admissions from 3 years before to 3 years after treatment, we observed highly reduced admission rates for stable angina (p<0.0001), revascularization (p=0.003) and overall cardiovascular disease (p<0.0001). No early or late side-effects of the treatment were observed. CONCLUSIONS The final 3-year follow-up data after intramyocardial injection of autologous BMMSCs, in patients with severe CAD and refractory angina, demonstrated sustained clinical effects, reduced hospital admissions for cardiovascular disease and excellent long-term safety. The results indicate that autotransplantation of BMMSCs to the heart does not only improve symptoms but also slows down disease progression.
Collapse
Affiliation(s)
- Anders Bruun Mathiasen
- Cardiac Stem Cell Laboratory, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Cardiac Catheterization Laboratory 2014, The Heart Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.
| | | | | | | |
Collapse
|
50
|
Sanganalmath SK, Bolli R. Cell therapy for heart failure: a comprehensive overview of experimental and clinical studies, current challenges, and future directions. Circ Res 2013; 113:810-34. [PMID: 23989721 PMCID: PMC3892665 DOI: 10.1161/circresaha.113.300219] [Citation(s) in RCA: 434] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 06/07/2013] [Indexed: 12/28/2022]
Abstract
Despite significant therapeutic advances, the prognosis of patients with heart failure (HF) remains poor, and current therapeutic approaches are palliative in the sense that they do not address the underlying problem of the loss of cardiac tissue. Stem cell-based therapies have the potential to fundamentally transform the treatment of HF by achieving what would have been unthinkable only a few years ago-myocardial regeneration. For the first time since cardiac transplantation, a therapy is being developed to eliminate the underlying cause of HF, not just to achieve damage control. Since the initial report of cell therapy (skeletal myoblasts) in HF in 1998, research has proceeded at lightning speed, and numerous preclinical and clinical studies have been performed that support the ability of various stem cell populations to improve cardiac function and reduce infarct size in both ischemic and nonischemic cardiomyopathy. Nevertheless, we are still at the dawn of this therapeutic revolution. Many important issues (eg, mechanism(s) of action of stem cells, long-term engraftment, optimal cell type(s), and dose, route, and frequency of cell administration) remain to be resolved, and no cell therapy has been conclusively shown to be effective. The purpose of this article is to critically review the large body of work performed with respect to the use of stem/progenitor cells in HF, both at the experimental and clinical levels, and to discuss current controversies, unresolved issues, challenges, and future directions. The review focuses specifically on chronic HF; other settings (eg, acute myocardial infarction, refractory angina) are not discussed.
Collapse
Affiliation(s)
- Santosh K Sanganalmath
- Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville, KY, USA
| | | |
Collapse
|