1
|
Demkes E, Cervera-Barea A, Ebner-Peking P, Wolf M, Hochmann S, Scheren AS, Bijsterveld M, van Oostveen CM, Jansen M, Visser J, Triebert W, Halloin C, Dobbe JGG, de Vos J, Schürz M, Danmayr J, Aalders MCG, Boink GJJ, Neef K, Strunk D, Zweigerdt R, de Jager SCA, Sluijter JPG. Human Cardiac Microtissues Display Improved Engraftment and Survival in a Porcine Model of Myocardial Infarction. J Cardiovasc Transl Res 2025:10.1007/s12265-025-10596-0. [PMID: 40082315 DOI: 10.1007/s12265-025-10596-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 01/20/2025] [Indexed: 03/16/2025]
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) constitute a promising therapy for myocardial infarction (MI). The lack of an effective immunosuppressive regimen, combined with single-cell transplantations, results in suboptimal outcomes, such as poor engraftment and compromised therapeutic efficacy. This study aimed to confirm the increased retention of hiPSC-CMs microtissues (CMTs) over single-cell grafts. To ensure the long-term survival of CMTs for potential cardiac applications, CMTs were transplanted in a porcine model of MI using a triple immunosuppression protocol designed to limit immune cell infiltration. Acute evaluation of spherical hiPSC-CMs aggregates and dissociated aggregates followed by the development of a triple immunosuppression protocol were performed in healthy animals. Long-term survival of CMTs was later examined in pigs that underwent a transient coronary occlusion. Two weeks post-MI, the immunosuppression treatment was initiated and on day 28 the animals were transplanted with CMTs and followed for four more weeks. Acutely, CMTs showed superior retention compared to their dissociated counterparts. The immunosuppression regimen led to no organ damage and stable levels of circulating drugs once optimal dose was achieved. Two weeks post-xenotransplantation in healthy pigs, histology revealed that immunosuppressed animals displayed a significant decrease in total cellular infiltrates, particularly in CD3+ T cells. Pigs that underwent coronary occlusion, which later were immunosuppressed and treated with CMTs (5 × 107 cells), showed cell engraftment onto the native myocardium four weeks post-transplantation. This study supports the use of a triple immunosuppression cocktail to ensure long-term survival of CMTs for the treatment of MI.
Collapse
Affiliation(s)
- Evelyne Demkes
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Aina Cervera-Barea
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| | | | - Martin Wolf
- Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria
| | - Sarah Hochmann
- Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria
| | - Amy S Scheren
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Mayke Bijsterveld
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - C Marlies van Oostveen
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marlijn Jansen
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Joyce Visser
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wiebke Triebert
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
| | - Caroline Halloin
- Cell Therapy Process Development, Novo Nordisk A/S, Maaloev, Denmark
| | - Johannes G G Dobbe
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Judith de Vos
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Melanie Schürz
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Joachim Danmayr
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Maurice C G Aalders
- Department of Biomedical Engineering and Physics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Gerard J J Boink
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Department of Clinical and Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Klaus Neef
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Dirk Strunk
- Cell Therapy Institute, Paracelsus Medical University, Salzburg, Austria
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs, Hannover Medical School, Hannover, Germany
| | - Saskia C A de Jager
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Joost P G Sluijter
- Experimental Cardiology Laboratory, Department of Cardiology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
2
|
Wulfse M, Vervoorn MT, Amelink JJGJ, Ballan EM, de Jager SCA, Sluijter JPG, Doevendans PA, Zwetsloot PPM, Van der Kaaij NP. Past Trends and Future Directions of Cardiac Regenerative Medicine - A Systematic Analysis of Clinical Trial Registries. J Cardiovasc Transl Res 2025; 18:209-220. [PMID: 39361114 PMCID: PMC11885401 DOI: 10.1007/s12265-024-10563-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/12/2024] [Indexed: 03/09/2025]
Abstract
Cell therapy, gene therapy, and tissue engineering have been explored as potential strategies to repair or regenerate damaged cardiac tissue. Despite the presence of encouraging preclinical data, clinical trials of regenerative cardiac therapies have yielded mixed results. Our study aimed to investigate the fate of all registered clinical trials within regenerative cardiac medicine, with the purpose of exploring the potential role of publication bias (or trial-completion bias), how published and unpublished research affects the field, and to draw lessons and recommendations for future clinical trials. In this analysis, we show that only a third of all registered trials has yielded results and that a significant number of trials are not completed. Furthermore, we identified significant heterogeneity in study design, study phase, funding, specific therapies used, primary outcome measures and methods of outcome assessment. These observations might hinder the successful translation of cardiac regenerative therapies into clinical practice.
Collapse
Affiliation(s)
- Maaike Wulfse
- Department of Cardiothoracic Surgery, Division of Heart & Lungs, University Medical Center Utrecht, P.O. Box 85500, Utrecht, 3508, GA, The Netherlands
| | - Mats T Vervoorn
- Department of Cardiothoracic Surgery, Division of Heart & Lungs, University Medical Center Utrecht, P.O. Box 85500, Utrecht, 3508, GA, The Netherlands
| | - Jantijn J G J Amelink
- Department of Cardiothoracic Surgery, Division of Heart & Lungs, University Medical Center Utrecht, P.O. Box 85500, Utrecht, 3508, GA, The Netherlands
| | - Elisa M Ballan
- Department of Cardiothoracic Surgery, Division of Heart & Lungs, University Medical Center Utrecht, P.O. Box 85500, Utrecht, 3508, GA, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
- Department of Cardiology, Laboratory of Experimental Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Saskia C A de Jager
- Department of Cardiology, Laboratory of Experimental Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
- Circulatory Health Research Center, Regenerative Medicine Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Pieter A Doevendans
- Netherlands Heart Institute, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Peter-Paul M Zwetsloot
- Department of Cardiology, Laboratory of Experimental Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart & Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Niels P Van der Kaaij
- Department of Cardiothoracic Surgery, Division of Heart & Lungs, University Medical Center Utrecht, P.O. Box 85500, Utrecht, 3508, GA, The Netherlands.
| |
Collapse
|
3
|
Li YL, Chen EG, Ren BB. Umbilical cord-derived mesenchymal stromal cells: Promising therapy for heart failure. World J Cardiol 2025; 17:101153. [PMID: 39866217 PMCID: PMC11755126 DOI: 10.4330/wjc.v17.i1.101153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/06/2024] [Accepted: 12/02/2024] [Indexed: 01/21/2025] Open
Abstract
Heart failure (HF) is a complex syndrome characterized by the reduced capacity of the heart to adequately fill or eject blood. Currently, HF remains a leading cause of morbidity and mortality worldwide, imposing a substantial burden on global healthcare systems. Recent advancements have highlighted the therapeutic potential of mesenchymal stromal cells (MSCs) in managing HF. Notably, umbilical cord-derived MSCs (UC-MSCs) have demonstrated superior clinical potential compared to traditional bone marrow-derived MSCs; this is evident in their non-invasive collection process, higher proliferation efficacy, and lower immunogenicity and tumorigenicity, as substantiated by preclinical studies. Although the feasibility and safety of UC-MSCs have been tested in animal models, the application of UC-MSCs in HF treatment remains challenged by issues such as inaccurate targeted migration and low survival rates of UC-MSCs. Therefore, further research and clinical trials are imperative to advance the clinical application of UC-MSCs.
Collapse
Affiliation(s)
- Ya-Lun Li
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Disease, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
- Medical College, Zhejiang University, Hangzhou 310063, Zhejiang Province, China
| | - En-Guo Chen
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Disease, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China
| | - Bing-Bing Ren
- Department of Pulmonary and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Disease, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, Zhejiang Province, China.
| |
Collapse
|
4
|
Sajjad MW, Muzamil F, Sabir M, Ashfaq UA. Regenerative Medicine and Nanotechnology Approaches against Cardiovascular Diseases: Recent Advances and Future Prospective. Curr Stem Cell Res Ther 2025; 20:50-71. [PMID: 38343052 DOI: 10.2174/011574888x263530230921074827] [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: 05/22/2023] [Revised: 07/07/2023] [Accepted: 07/14/2023] [Indexed: 01/31/2025]
Abstract
Regenerative medicine refers to medical research focusing on repairing, replacing, or regenerating damaged or diseased tissues or organs. Cardiovascular disease (CVDs) is a significant health issue globally and is the leading cause of death in many countries. According to the Centers for Disease Control and Prevention (CDC), one person dies every 34 seconds in the United States from cardiovascular diseases, and according to a World Health Organization (WHO) report, cardiovascular diseases are the leading cause of death globally, taking an estimated 17.9 million lives each year. Many conventional treatments are available using different drugs for cardiovascular diseases, but these treatments are inadequate. Stem cells and nanotechnology are promising research areas for regenerative medicine treating CVDs. Regenerative medicines are a revolutionary strategy for advancing and successfully treating various diseases, intending to control cardiovascular disorders. This review is a comprehensive study of different treatment methods for cardiovascular diseases using different types of biomaterials as regenerative medicines, the importance of different stem cells in therapeutics, the expanded role of nanotechnology in treatment, the administration of several types of stem cells, their tracking, imaging, and the final observation of clinical trials on many different levels as well as it aims to keep readers up to pace on emerging therapeutic applications of some specific organs and disorders that may improve from regenerative medicine shortly.
Collapse
Affiliation(s)
- Muhammad Waseem Sajjad
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Fatima Muzamil
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Maida Sabir
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| |
Collapse
|
5
|
Garmany A, Terzic A. Artificial intelligence powers regenerative medicine into predictive realm. Regen Med 2024; 19:611-616. [PMID: 39660914 PMCID: PMC11703382 DOI: 10.1080/17460751.2024.2437281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024] Open
Abstract
The expanding regenerative medicine toolkit is reaching a record number of lives. There is a pressing need to enhance the precision, efficiency, and effectiveness of regenerative approaches and achieve reliable outcomes. While regenerative medicine has relied on an empiric paradigm, availability of big data along with advances in informatics and artificial intelligence offer the opportunity to inform the next generation of regenerative sciences along the discovery, translation, and application pathway. Artificial intelligence can streamline discovery and development of optimized biotherapeutics by aiding in the interpretation of readouts associated with optimal repair outcomes. In advanced biomanufacturing, artificial intelligence holds potential in ensuring quality control and assuring scalability through automated monitoring of process-critical variables mandatory for product consistency. In practice application, artificial intelligence can guide clinical trial design, patient selection, delivery strategies, and outcome assessment. As artificial intelligence transforms the regenerative horizon, caution is necessary to reduce bias, ensure generalizability, and mitigate ethical concerns with the goal of equitable access for patients and populations.
Collapse
Affiliation(s)
- Armin Garmany
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Department of Molecular Pharmacology & Experimental Therapeutics, Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Alix School of Medicine, Regenerative Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
| | - Andre Terzic
- Marriott Heart Disease Research Program, Department of Cardiovascular Medicine, Department of Molecular Pharmacology & Experimental Therapeutics, Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
6
|
Garmany A, Arrell DK, Yamada S, Jeon R, Behfar A, Park S, Terzic A. Decoded cardiopoietic cell secretome linkage to heart repair biosignature. Stem Cells Transl Med 2024; 13:1144-1159. [PMID: 39259666 PMCID: PMC11555478 DOI: 10.1093/stcltm/szae067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/03/2024] [Indexed: 09/13/2024] Open
Abstract
Cardiopoiesis-primed human stem cells exert sustained benefit in treating heart failure despite limited retention following myocardial delivery. To assess potential paracrine contribution, the secretome of cardiopoiesis conditioned versus naïve human mesenchymal stromal cells was decoded by directed proteomics augmented with machine learning and systems interrogation. Cardiopoiesis doubled cellular protein output generating a distinct secretome that segregated the conditioned state. Altering the expression of 1035 secreted proteins, cardiopoiesis reshaped the secretome across functional classes. The resolved differential cardiopoietic secretome was enriched in mesoderm development and cardiac progenitor signaling processes, yielding a cardiovasculogenic profile bolstered by upregulated cardiogenic proteins. In tandem, cardiopoiesis enhanced the secretion of immunomodulatory proteins associated with cytokine signaling, leukocyte migration, and chemotaxis. Network analysis integrated the differential secretome within an interactome of 1745 molecules featuring prioritized regenerative processes. Secretome contribution to the repair signature of cardiopoietic cell-treated infarcted hearts was assessed in a murine coronary ligation model. Intramyocardial delivery of cardiopoietic cells improved the performance of failing hearts, with undirected proteomics revealing 50 myocardial proteins responsive to cell therapy. Pathway analysis linked the secretome to cardiac proteome remodeling, pinpointing 17 cardiopoiesis-upregulated secretome proteins directly upstream of 44% of the cell therapy-responsive cardiac proteome. Knockout, in silico, of this 22-protein secretome-dependent myocardial ensemble eliminated indices of the repair signature. Accordingly, in vivo, cell therapy rendered the secretome-dependent myocardial proteome of an infarcted heart indiscernible from healthy counterparts. Thus, the secretagogue effect of cardiopoiesis transforms the human stem cell secretome, endows regenerative competency, and upregulates candidate paracrine effectors of cell therapy-mediated molecular restitution.
Collapse
Affiliation(s)
- Armin Garmany
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic Alix School of Medicine, Regenerative Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States
| | - D Kent Arrell
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Satsuki Yamada
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Section of Geriatric Medicine & Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Ryounghoon Jeon
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Atta Behfar
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, MN, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Sungjo Park
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
| | - Andre Terzic
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Rochester, MN, United States
- Marriott Heart Disease Research Program, Mayo Clinic, Rochester, MN, United States
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
- Department of Medical Genetics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
7
|
Tao S, Yu L, Li J, Wu J, Yang D, Xue T, Zhang L, Xie Z, Huang X. Stem cell therapy for non-ischemic dilated cardiomyopathy: a systematic review and meta-analysis. Syst Rev 2024; 13:276. [PMID: 39516841 PMCID: PMC11546504 DOI: 10.1186/s13643-024-02701-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Stem cell therapy is the transplantation of human cells to aid the healing of damaged or wounded tissues and cells. Only a few small-scale trials have been conducted to investigate stem cell therapy for non-ischemic dilated cardiomyopathy (DCM). We aimed to perform a systematic review and meta-analysis to assess the efficacy and safety of stem cell therapy for DCM. METHODS A comprehensive search of the databases of PubMed, Embase, Web of Science Core Collection, Cochrane Library, and ProQuest was conducted from their inception to June 30, 2024, to access randomized controlled trials (RCTs) that were centered on stem cell therapy for DCM. The primary outcome was left ventricular ejection fraction (LVEF), and the secondary outcomes included left ventricular end-diastolic dimension (LVEDD), left ventricular end-diastolic volume (LVEDV), 6-min walk test (6MWT), NYHA functional classification, quality of life (QoL) such as Minnesota Living with Heart Failure Questionnaire (MLHFQ) and Kansas City Cardiomyopathy Questionnaire (KCCQ), N-terminal pro-brain natriuretic peptide (NT-proBNP), and VO2 peak. Moreover, major adverse cardiovascular events (MACEs) were also recorded. The Cochrane risk-of-bias assessment tool was used to evaluate the quality of the included RCTs, and the certainty of the evidence was assessed using the GRADE method. Sensitivity analysis was taken into consideration to determine the stability of the results. This review was registered with PROSPERO (CRD42024568912). RESULTS Eleven RCTs involving 637 participants were included in the quantitative analysis. The results indicated that there was a significant increase in mean LVEF (MD = 4.84, 95% CI 3.25-6.42, P < 0.00001) and considerable decrease in LVEDV (MD = - 29.51, 95% CI - 58.07 to - 0.95, P = 0.04) and NT-proBNP (MD = - 737.55, 95% CI - 904.28 to - 570.82, P < 0.00001) in DCM patients treated with stem cell therapy compared with controls. Stem cell therapy was also related to the improvement in functional capacity, as evaluated by 6MWT (MD = 44.32, 95% CI 34.70 - 53.94, P < 0.00001) and NYHA functional classification (MD = - 0.63, 95% CI - 0.96 to - 0.30, P = 0.0002). It also had positive effects on improving QoL, including significantly decreasing MLHFQ score (MD = - 16.60, 95% CI - 26.57 to - 6.63, P = 0.001) and increasing the KCCQ score (MD = 14.76, 95% CI 7.76 - 21.76, P < 0.0001). No significant differences were observed in LVEDD, VO2 peak, and MACEs between the two groups. The GRADE analysis revealed that the evidence was graded from low to moderate. Sensitivity analysis of the results suggested that the results were stable. CONCLUSION The systematic review and meta-analysis indicates that stem cell therapy may be an effective and safe approach to improve cardiac function and quality of life in DCM patients. Nevertheless, given the limitations of existing studies, larger well-designed RCTs are required to confirm and support our findings.
Collapse
Affiliation(s)
- Shiyi Tao
- Department of Cardiology, Guang'anmen Hospital, China, Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Lintong Yu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Jun Li
- Department of Cardiology, Guang'anmen Hospital, China, Academy of Chinese Medical Sciences, Beijing, China.
| | - Ji Wu
- Department of Cardiology, Guang'anmen Hospital, China, Academy of Chinese Medical Sciences, Beijing, China
| | - Deshuang Yang
- Department of Integrative Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Tiantian Xue
- Department of Cardiology, Guang'anmen Hospital, China, Academy of Chinese Medical Sciences, Beijing, China
| | - Lanxin Zhang
- Department of Cardiology, Guang'anmen Hospital, China, Academy of Chinese Medical Sciences, Beijing, China
| | - Zicong Xie
- Department of Cardiology, Guang'anmen Hospital, China, Academy of Chinese Medical Sciences, Beijing, China
| | - Xuanchun Huang
- Department of Cardiology, Guang'anmen Hospital, China, Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
8
|
Smadja DM. Extracellular Microvesicles vs. Mitochondria: Competing for the Top Spot in Cardiovascular Regenerative Medicine. Stem Cell Rev Rep 2024; 20:1813-1818. [PMID: 38976143 DOI: 10.1007/s12015-024-10758-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/09/2024]
Abstract
Regenerative medicine aims to restore, replace, and regenerate human cells, tissues, and organs. Despite significant advancements, many cell therapy trials for cardiovascular diseases face challenges like cell survival and immune compatibility, with benefits largely stemming from paracrine effects. Two promising therapeutic tools have been recently emerged in cardiovascular diseases: extracellular vesicles (EVs) and mitochondrial transfer. Concerning EVs, the first pivotal study with EV-enriched secretome derived from cardiovascular progenitor cells has been done treating heart failure. This first in man demonstrated the safety and feasibility of repeated intravenous infusions and highlighted significant clinical improvements, including enhanced cardiac function and reduced symptoms in heart failure patients. The second study uncovered a novel mechanism of endothelial regeneration through mitochondrial transfer via tunneling nanotubes (TNTs). This research showed that mesenchymal stromal cells (MSCs) transfer mitochondria to endothelial cells, significantly enhancing their bioenergetics and vessel-forming capabilities. This mitochondrial transfer was crucial for endothelial cell engraftment and function, offering a new strategy for vascular regeneration without the need for additional cell types. Combining EV and mitochondrial strategies presents new clinical opportunities. These approaches could revolutionize regenerative medicine, offering new hope for treating cardiovascular and other degenerative diseases. Continued research and clinical trials will be crucial in optimizing these therapies, potentially leading to personalized medicine approaches that enhance patient outcomes.
Collapse
Affiliation(s)
- David M Smadja
- Université Paris Cité, INSERM, Innovative Therapies in Hemostasis, Paris, F-75006, France.
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Inserm UMR-S1140, 56 rue Leblanc, Paris, F-75015, France.
| |
Collapse
|
9
|
Fatehi Hassanabad A, Zarzycki AN, Fedak PWM. Cellular and molecular mechanisms driving cardiac tissue fibrosis: On the precipice of personalized and precision medicine. Cardiovasc Pathol 2024; 71:107635. [PMID: 38508436 DOI: 10.1016/j.carpath.2024.107635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024] Open
Abstract
Cardiac fibrosis is a significant contributor to heart failure, a condition that continues to affect a growing number of patients worldwide. Various cardiovascular comorbidities can exacerbate cardiac fibrosis. While fibroblasts are believed to be the primary cell type underlying fibrosis, recent and emerging data suggest that other cell types can also potentiate or expedite fibrotic processes. Over the past few decades, clinicians have developed therapeutics that can blunt the development and progression of cardiac fibrosis. While these strategies have yielded positive results, overall clinical outcomes for patients suffering from heart failure continue to be dire. Herein, we overview the molecular and cellular mechanisms underlying cardiac tissue fibrosis. To do so, we establish the known mechanisms that drive fibrosis in the heart, outline the diagnostic tools available, and summarize the treatment options used in contemporary clinical practice. Finally, we underscore the critical role the immune microenvironment plays in the pathogenesis of cardiac fibrosis.
Collapse
Affiliation(s)
- Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Anna N Zarzycki
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul W M Fedak
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
10
|
Yang B, Qiao Y, Yan D, Meng Q. Targeting Interactions between Fibroblasts and Macrophages to Treat Cardiac Fibrosis. Cells 2024; 13:764. [PMID: 38727300 PMCID: PMC11082988 DOI: 10.3390/cells13090764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 05/13/2024] Open
Abstract
Excessive extracellular matrix (ECM) deposition is a defining feature of cardiac fibrosis. Most notably, it is characterized by a significant change in the concentration and volume fraction of collagen I, a disproportionate deposition of collagen subtypes, and a disturbed ECM network arrangement, which directly affect the systolic and diastolic functions of the heart. Immune cells that reside within or infiltrate the myocardium, including macrophages, play important roles in fibroblast activation and consequent ECM remodeling. Through both direct and indirect connections to fibroblasts, monocyte-derived macrophages and resident cardiac macrophages play complex, bidirectional, regulatory roles in cardiac fibrosis. In this review, we discuss emerging interactions between fibroblasts and macrophages in physiology and pathologic conditions, providing insights for future research aimed at targeting macrophages to combat cardiac fibrosis.
Collapse
Affiliation(s)
- Bo Yang
- Center for Organoid and Regeneration Medicine, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Guangzhou 511466, China;
| | - Yan Qiao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010021, China;
| | - Dong Yan
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China;
| | - Qinghang Meng
- Center for Organoid and Regeneration Medicine, Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Guangzhou 511466, China;
| |
Collapse
|
11
|
Shiraishi M, Sasaki D, Hibino M, Takeda A, Harashima H, Yamada Y. Human cardiosphere-derived cells with activated mitochondria for better myocardial regenerative therapy. J Control Release 2024; 367:486-499. [PMID: 38295995 DOI: 10.1016/j.jconrel.2024.01.058] [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: 11/29/2023] [Revised: 01/04/2024] [Accepted: 01/27/2024] [Indexed: 02/06/2024]
Abstract
Cell transplantation is a promising therapeutic strategy for myocardial regeneration therapy. To improve therapeutic effects, we developed a culture medium additive that enhances the mitochondrial function of cardiomyocytes for transplantation. A mitochondrial targeting drug delivery system (MITO-Porter system) was used to deliver mitochondrial activation molecules to mouse-derived cardiac progenitor cells. In this study, we investigated whether the mitochondrial function of human-derived myocardial precursor cells could be enhanced using MITO-Porter. Human cardiosphere-derived cells (CDCs) were isolated from myocardium which was excised during surgery for congenital heart disease. MITO-Porter was added to the cell culture medium to generate mitochondrial activated CDCs (human MITO cells). The human MITO cells were transplanted into myocardial ischemia-reperfusion model rat, and the effect was investigated. The transplanted human MITO cells improved the cardiac function and suppressed myocardial fibrosis compared to conventional cell transplantation methods. These effects were observed not only with myocardial administration but also by intravenous administration of human MITO cells. This study is the first study that assessed whether the mitochondrial delivery of functional compounds improved the outcome of human-derived myocardial cell transplantation therapy.
Collapse
Affiliation(s)
- Masahiro Shiraishi
- Department of Pediatrics, Graduate School of Medicine, Hokkaido University, Kita-15, Nishi 7, Kita-ku, Sapporo 060-8638, Japan
| | - Daisuke Sasaki
- Department of Pediatrics, Graduate School of Medicine, Hokkaido University, Kita-15, Nishi 7, Kita-ku, Sapporo 060-8638, Japan
| | - Mitsue Hibino
- Faculty of Engineering, Hokkaido University, Kita-13, Nishi-8, Kita-ku, Sapporo 060-0812, Japan
| | - Atsuhito Takeda
- Department of Pediatrics, Graduate School of Medicine, Hokkaido University, Kita-15, Nishi 7, Kita-ku, Sapporo 060-8638, Japan
| | - Hideyoshi Harashima
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan
| | - Yuma Yamada
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12, Nishi-6, Kita-ku, Sapporo 060-0812, Japan; Fusion Oriented REsearch for disruptive Science and Technology (FOREST) Program, Japan Science and Technology Agency (JST) Japan, Kawaguchi Center Building, 4-1-8, Honcho, Kawaguchi-shi, Saitama 332-0012, Japan.
| |
Collapse
|
12
|
Badimon L, Arderiu G, Vilahur G, Padro T, Cordero A, Mendieta G. Perivascular and epicardial adipose tissue. Vascul Pharmacol 2024; 154:107254. [PMID: 38072220 DOI: 10.1016/j.vph.2023.107254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/29/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023]
Affiliation(s)
- Lina Badimon
- Cardiovascular-Program ICCC; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Ciber CV, Instituto Carlos III, Madrid, Spain; Red TERAV, Instituto Carlos III, Madrid, Spain.
| | - Gemma Arderiu
- Cardiovascular-Program ICCC; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Ciber CV, Instituto Carlos III, Madrid, Spain; Red TERAV, Instituto Carlos III, Madrid, Spain
| | - Gemma Vilahur
- Cardiovascular-Program ICCC; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Ciber CV, Instituto Carlos III, Madrid, Spain; Red TERAV, Instituto Carlos III, Madrid, Spain
| | - Teresa Padro
- Cardiovascular-Program ICCC; Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain; Ciber CV, Instituto Carlos III, Madrid, Spain; Red TERAV, Instituto Carlos III, Madrid, Spain
| | - Alberto Cordero
- Ciber CV, Instituto Carlos III, Madrid, Spain; Cardiology Department, Hospital IMED Elche, Alicante, Spain
| | - Guiomar Mendieta
- Cardiology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain
| |
Collapse
|
13
|
Paz Y, Levy Y, Grosman-Rimon L, Shinfeld A. Nonpharmacological interventions for 'no-option' refractory angina patients. J Cardiovasc Med (Hagerstown) 2024; 25:13-22. [PMID: 37942734 DOI: 10.2459/jcm.0000000000001566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Refractory angina pectoris (RAP) defined as chronic anginal chest pain because of coronary artery disease (CAD) is a major problem. The increase in the number of patients with RAP in recent years is because of the increasing aging population and improved survival rates among patients with CAD. Management of patients with RAP is often extremely challenging. In this review, we present several interventional approaches for RAP, including device therapies, lifestyle intervention, and cell therapies. Some of these treatments are currently used in the management of RAP, whereas other treatments are under investigation.
Collapse
Affiliation(s)
- Yoav Paz
- General Intensive Care Unit, Sourasky Medical Center, Tel Aviv, Israel, affiliated with Sackler Faculty of Medicine, Tel Aviv University
| | - Yair Levy
- Department of Medicine, Meir Hospital, Kfar-Saba, Israel
| | - Liza Grosman-Rimon
- School of Graduate Studies, Levinsky-Wingate Academic College, Wingate Institute, Netanya, Israel
| | - Amihay Shinfeld
- Department of Cardiac Surgery, Sheba Medical Center, Tel Hashomer, Tel Aviv, Israel
| |
Collapse
|
14
|
Manescu (Paltanea) V, Antoniac I, Antoniac A, Laptoiu D, Paltanea G, Ciocoiu R, Nemoianu IV, Gruionu LG, Dura H. Bone Regeneration Induced by Patient-Adapted Mg Alloy-Based Scaffolds for Bone Defects: Present and Future Perspectives. Biomimetics (Basel) 2023; 8:618. [PMID: 38132557 PMCID: PMC10742271 DOI: 10.3390/biomimetics8080618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/09/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Treatment of bone defects resulting after tumor surgeries, accidents, or non-unions is an actual problem linked to morbidity and the necessity of a second surgery and often requires a critical healthcare cost. Although the surgical technique has changed in a modern way, the treatment outcome is still influenced by patient age, localization of the bone defect, associated comorbidities, the surgeon approach, and systemic disorders. Three-dimensional magnesium-based scaffolds are considered an important step because they can have precise bone defect geometry, high porosity grade, anatomical pore shape, and mechanical properties close to the human bone. In addition, magnesium has been proven in in vitro and in vivo studies to influence bone regeneration and new blood vessel formation positively. In this review paper, we describe the magnesium alloy's effect on bone regenerative processes, starting with a short description of magnesium's role in the bone healing process, host immune response modulation, and finishing with the primary biological mechanism of magnesium ions in angiogenesis and osteogenesis by presenting a detailed analysis based on a literature review. A strategy that must be followed when a patient-adapted scaffold dedicated to bone tissue engineering is proposed and the main fabrication technologies are combined, in some cases with artificial intelligence for Mg alloy scaffolds, are presented with examples. We emphasized the microstructure, mechanical properties, corrosion behavior, and biocompatibility of each study and made a basis for the researchers who want to start to apply the regenerative potential of magnesium-based scaffolds in clinical practice. Challenges, future directions, and special potential clinical applications such as osteosarcoma and persistent infection treatment are present at the end of our review paper.
Collapse
Affiliation(s)
- Veronica Manescu (Paltanea)
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.A.); (R.C.)
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (G.P.); (I.V.N.)
| | - Iulian Antoniac
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.A.); (R.C.)
- Academy of Romanian Scientists, 54 Splaiul Independentei, RO-050094 Bucharest, Romania
| | - Aurora Antoniac
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.A.); (R.C.)
| | - Dan Laptoiu
- Department of Orthopedics and Trauma I, Colentina Clinical Hospital, 19-21 Soseaua Stefan cel Mare, RO-020125 Bucharest, Romania;
| | - Gheorghe Paltanea
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (G.P.); (I.V.N.)
| | - Robert Ciocoiu
- Faculty of Material Science and Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (V.M.); (A.A.); (R.C.)
| | - Iosif Vasile Nemoianu
- Faculty of Electrical Engineering, National University of Science and Technology Politehnica Bucharest, 313 Splaiul Independentei, District 6, RO-060042 Bucharest, Romania; (G.P.); (I.V.N.)
| | - Lucian Gheorghe Gruionu
- Faculty of Mechanics, University of Craiova, 13 Alexandru Ioan Cuza, RO-200585 Craiova, Romania;
| | - Horatiu Dura
- Faculty of Medicine, Lucian Blaga University of Sibiu, RO-550169 Sibiu, Romania;
| |
Collapse
|
15
|
Chepeleva EV. Cell Therapy in the Treatment of Coronary Heart Disease. Int J Mol Sci 2023; 24:16844. [PMID: 38069167 PMCID: PMC10706847 DOI: 10.3390/ijms242316844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Heart failure is a leading cause of death in patients who have suffered a myocardial infarction. Despite the timely use of modern reperfusion therapies such as thrombolysis, surgical revascularization and balloon angioplasty, they are sometimes unable to prevent the development of significant areas of myocardial damage and subsequent heart failure. Research efforts have focused on developing strategies to improve the functional status of myocardial injury areas. Consequently, the restoration of cardiac function using cell therapy is an exciting prospect. This review describes the characteristics of various cell types relevant to cellular cardiomyoplasty and presents findings from experimental and clinical studies investigating cell therapy for coronary heart disease. Cell delivery methods, optimal dosage and potential treatment mechanisms are discussed.
Collapse
Affiliation(s)
- Elena V. Chepeleva
- Federal State Budgetary Institution National Medical Research Center Named after Academician E.N. Meshalkin of the Ministry of Health of the Russian Federation, 15, Rechkunovskaya Str., 630055 Novosibirsk, Russia;
- Research Institute of Clinical and Experimental Lymphology—Branch of the Institute of Cytology and Genetics Siberian Branch of Russian Academy of Sciences, 2, Timakova Str., 630060 Novosibirsk, Russia
| |
Collapse
|
16
|
Pompilio G, Sanz-Ruiz R, Sluijter JPG, Janssens S. The ESC Working Group on cardiovascular regenerative and reparative medicine. Eur Heart J 2023; 44:4503-4505. [PMID: 37642557 PMCID: PMC10645042 DOI: 10.1093/eurheartj/ehad538] [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: 08/31/2023] Open
Affiliation(s)
- Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milano, Italy
- Dipartimento di Scienze Cliniche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Milano, Italy
| | - Ricardo Sanz-Ruiz
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Joost P G Sluijter
- Department of Cardiology, Experimental Cardiology Laboratory, Circulatory Health Research Center, Regenerative medicine Center Utrecht, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Stefan Janssens
- Department of Cardiovascular Diseases, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
17
|
Deszcz I. Stem Cell-Based Therapy and Cell-Free Therapy as an Alternative Approach for Cardiac Regeneration. Stem Cells Int 2023; 2023:2729377. [PMID: 37954462 PMCID: PMC10635745 DOI: 10.1155/2023/2729377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/21/2023] [Accepted: 10/10/2023] [Indexed: 11/14/2023] Open
Abstract
The World Health Organization reports that cardiovascular diseases (CVDs) represent 32% of all global deaths. The ineffectiveness of conventional therapies in CVDs encourages the development of novel, minimally invasive therapeutic strategies for the healing and regeneration of damaged tissue. The self-renewal capacity, multilineage differentiation, lack of immunogenicity, and immunosuppressive properties of mesenchymal stem cells (MSCs) make them a promising option for CVDs. However, growing evidence suggests that myocardial regeneration occurs through paracrine factors and extracellular vesicle (EV) secretion, rather than through differentiation into cardiomyocytes. Research shows that stem cells secrete or surface-shed into their culture media various cytokines, chemokines, growth factors, anti-inflammatory factors, and EVs, which constitute an MSC-conditioned medium (MSC-CM) or the secretome. The use of MSC-CM enhances cardiac repair through resident heart cell differentiation, proliferation, scar mass reduction, a decrease in infarct wall thickness, and cardiac function improvement comparable to MSCs without their side effects. This review highlights the limitations and benefits of therapies based on stem cells and their secretome as an innovative treatment of CVDs.
Collapse
Affiliation(s)
- Iwona Deszcz
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Borowska 211, 50-556, Wroclaw, Poland
| |
Collapse
|
18
|
Sundin A, Ionescu SI, Balkan W, Hare JM. Mesenchymal STRO-1/STRO-3 + precursor cells for the treatment of chronic heart failure with reduced ejection fraction. Future Cardiol 2023; 19:567-581. [PMID: 37933628 PMCID: PMC10652293 DOI: 10.2217/fca-2023-0081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/30/2023] [Indexed: 11/08/2023] Open
Abstract
The heart is susceptible to proinflammatory and profibrotic responses after myocardial injury, leading to further worsening of cardiac dysfunction. Important developments in the management of heart failure with reduced ejection fraction have reduced morbidity and mortality; however, these therapies focus on optimizing cardiac function through hemodynamic and neurohormonal pathways and not by repairing the underlying cardiac injury. The potential of cell-based therapy to reverse cardiac injury has received substantial attention. Herein are examined the phase II and III studies of bone marrow-derived mesenchymal STRO-1+ or STRO-1/STRO-3+ precursor cells in patients with ischemic and nonischemic heart failure with reduced ejection fraction, addressing the safety and efficacy of cell-based therapy throughout multiple clinical trials, the optimal dose and the steps toward revolutionizing the treatment of heart failure.
Collapse
Affiliation(s)
- Andrew Sundin
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Simona I Ionescu
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
19
|
Berkeley B, Tang MNH, Brittan M. Mechanisms regulating vascular and lymphatic regeneration in the heart after myocardial infarction. J Pathol 2023; 260:666-678. [PMID: 37272582 PMCID: PMC10953458 DOI: 10.1002/path.6093] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/14/2023] [Accepted: 04/27/2023] [Indexed: 06/06/2023]
Abstract
Myocardial infarction, caused by a thrombus or coronary vascular occlusion, leads to irreversible ischaemic injury. Advances in early reperfusion strategies have significantly reduced short-term mortality after myocardial infarction. However, survivors have an increased risk of developing heart failure, which confers a high risk of death at 1 year. The capacity of the injured neonatal mammalian heart to regenerate has stimulated extensive research into whether recapitulation of developmental regeneration programmes may be beneficial in adult cardiovascular disease. Restoration of functional blood and lymphatic vascular networks in the infarct and border regions via neovascularisation and lymphangiogenesis, respectively, is a key requirement to facilitate myocardial regeneration. An improved understanding of the endogenous mechanisms regulating coronary vascular and lymphatic expansion and function in development and in adult patients after myocardial infarction may inform future therapeutic strategies and improve translation from pre-clinical studies. In this review, we explore the underpinning research and key findings in the field of cardiovascular regeneration, with a focus on neovascularisation and lymphangiogenesis, and discuss the outcomes of therapeutic strategies employed to date. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Bronwyn Berkeley
- Centre for Cardiovascular Science, The Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Michelle Nga Huen Tang
- Centre for Cardiovascular Science, The Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
| | - Mairi Brittan
- Centre for Cardiovascular Science, The Queen's Medical Research InstituteUniversity of EdinburghEdinburghUK
| |
Collapse
|
20
|
Clavellina D, Balkan W, Hare JM. Stem cell therapy for acute myocardial infarction: Mesenchymal Stem Cells and induced Pluripotent Stem Cells. Expert Opin Biol Ther 2023; 23:951-967. [PMID: 37542462 PMCID: PMC10837765 DOI: 10.1080/14712598.2023.2245329] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 08/07/2023]
Abstract
INTRODUCTION Acute myocardial infarction (AMI) remains a leading cause of death in the United States. The limited capacity of cardiomyocytes to regenerate and the restricted contractility of scar tissue after AMI are not addressed by current pharmacologic interventions. Mesenchymal stem/stromal cells (MSCs) have emerged as a promising therapeutic approach due to their low antigenicity, ease of harvesting, and efficacy and safety in preclinical and clinical studies, despite their low survival and engraftment rates. Other stem cell types, such as induced pluripotent stem cells (iPSCs) also show promise, and optimizing cardiac repair requires integrating emerging technologies and strategies. AREAS COVERED This review offers insights into advancing cell-based therapies for AMI, emphasizing meticulously planned trials with a standardized definition of AMI, for a bench-to-bedside approach. We critically evaluate fundamental studies and clinical trials to provide a comprehensive overview of the advances, limitations and prospects for cell-based therapy in AMI. EXPERT OPINION MSCs continue to show potential promise for treating AMI and its sequelae, but addressing their low survival and engraftment rates is crucial for clinical success. Integrating emerging technologies such as pluripotent stem cells and conducting well-designed trials will harness the full potential of cell-based therapy in AMI management. Collaborative efforts are vital to developing effective stem cell therapies for AMI patients.
Collapse
Affiliation(s)
- Diana Clavellina
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
21
|
Wu X, Vacante F, Wu JC. Lnc-ing epicardium-derived cells to cardiac remodelling: lncRNA-TARID as a novel antifibrotic option. Eur Heart J 2023; 44:1761-1763. [PMID: 36928295 PMCID: PMC10182884 DOI: 10.1093/eurheartj/ehad058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
Affiliation(s)
- Xuekun Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Francesca Vacante
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Division of Cardiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
22
|
Ning Y, Huang P, Chen G, Xiong Y, Gong Z, Wu C, Xu J, Jiang W, Li X, Tang R, Zhang L, Hu M, Xu J, Xu J, Qian H, Jin C, Yang Y. Atorvastatin-pretreated mesenchymal stem cell-derived extracellular vesicles promote cardiac repair after myocardial infarction via shifting macrophage polarization by targeting microRNA-139-3p/Stat1 pathway. BMC Med 2023; 21:96. [PMID: 36927608 PMCID: PMC10022054 DOI: 10.1186/s12916-023-02778-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/10/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) derived from bone marrow mesenchymal stem cells (MSCs) pretreated with atorvastatin (ATV) (MSCATV-EV) have a superior cardiac repair effect on acute myocardial infarction (AMI). The mechanisms, however, have not been fully elucidated. This study aims to explore whether inflammation alleviation of infarct region via macrophage polarization plays a key role in the efficacy of MSCATV-EV. METHODS MSCATV-EV or MSC-EV were intramyocardially injected 30 min after coronary ligation in AMI rats. Macrophage infiltration and polarization (day 3), cardiac function (days 0, 3, 7, 28), and infarct size (day 28) were measured. EV small RNA sequencing and bioinformatics analysis were conducted for differentially expressed miRNAs between MSCATV-EV and MSC-EV. Macrophages were isolated from rat bone marrow for molecular mechanism analysis. miRNA mimics or inhibitors were transfected into EVs or macrophages to analyze its effects on macrophage polarization and cardiac repair in vitro and in vivo. RESULTS MSCATV-EV significantly reduced the amount of CD68+ total macrophages and increased CD206+ M2 macrophages of infarct zone on day 3 after AMI compared with MSC-EV group (P < 0.01-0.0001). On day 28, MSCATV-EV much more significantly improved the cardiac function than MSC-EV with the infarct size markedly reduced (P < 0.05-0.0001). In vitro, MSCATV-EV also significantly reduced the protein and mRNA expressions of M1 markers but increased those of M2 markers in lipopolysaccharide-treated macrophages (P < 0.05-0.0001). EV miR-139-3p was identified as a potential cardiac repair factor mediating macrophage polarization. Knockdown of miR-139-3p in MSCATV-EV significantly attenuated while overexpression of it in MSC-EV enhanced the effect on promoting M2 polarization by suppressing downstream signal transducer and activator of transcription 1 (Stat1). Furthermore, MSCATV-EV loaded with miR-139-3p inhibitors decreased while MSC-EV loaded with miR-139-3p mimics increased the expressions of M2 markers and cardioprotective efficacy. CONCLUSIONS We uncovered a novel mechanism that MSCATV-EV remarkably facilitate cardiac repair in AMI by promoting macrophage polarization via miR-139-3p/Stat1 pathway, which has the great potential for clinical translation.
Collapse
Affiliation(s)
- Yu Ning
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
- National Health Commission Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Peisen Huang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
- National Health Commission Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Guihao Chen
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Yuyan Xiong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Zhaoting Gong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Chunxiao Wu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Junyan Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Wenyang Jiang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Xiaosong Li
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Ruijie Tang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Lili Zhang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Mengjin Hu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Jing Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Jun Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Haiyan Qian
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Chen Jin
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, No.167 North Lishi Road, Xicheng District, Beijing, 100037, China.
| |
Collapse
|
23
|
Yu Y, Tham SK, Roslan FF, Shaharuddin B, Yong YK, Guo Z, Tan JJ. Large animal models for cardiac remuscularization studies: A methodological review. Front Cardiovasc Med 2023; 10:1011880. [PMID: 37008331 PMCID: PMC10050756 DOI: 10.3389/fcvm.2023.1011880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 02/20/2023] [Indexed: 03/17/2023] Open
Abstract
Myocardial infarction is the most common cause of heart failure, one of the most fatal non-communicable diseases worldwide. The disease could potentially be treated if the dead, ischemic heart tissues are regenerated and replaced with viable and functional cardiomyocytes. Pluripotent stem cells have proven the ability to derive specific and functional cardiomyocytes in large quantities for therapy. To test the remuscularization hypothesis, the strategy to model the disease in animals must resemble the pathophysiological conditions of myocardial infarction as in humans, to enable thorough testing of the safety and efficacy of the cardiomyocyte therapy before embarking on human trials. Rigorous experiments and in vivo findings using large mammals are increasingly important to simulate clinical reality and increase translatability into clinical practice. Hence, this review focus on large animal models which have been used in cardiac remuscularization studies using cardiomyocytes derived from human pluripotent stem cells. The commonly used methodologies in developing the myocardial infarction model, the choice of animal species, the pre-operative antiarrhythmics prophylaxis, the choice of perioperative sedative, anaesthesia and analgesia, the immunosuppressive strategies in allowing xenotransplantation, the source of cells, number and delivery method are discussed.
Collapse
Affiliation(s)
- Yuexin Yu
- USM-ALPS Cardiac Research Laboratory, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, China
| | | | - Fatin Fazrina Roslan
- USM-ALPS Cardiac Research Laboratory, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Bakiah Shaharuddin
- USM-ALPS Cardiac Research Laboratory, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Yoke Keong Yong
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Zhikun Guo
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, China
- Correspondence: Jun Jie Tan Zhikun Guo
| | - Jun Jie Tan
- USM-ALPS Cardiac Research Laboratory, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
- Correspondence: Jun Jie Tan Zhikun Guo
| |
Collapse
|
24
|
Lang CI, Dahmen A, Vasudevan P, Lemcke H, Gäbel R, Öner A, Ince H, David R, Wolfien M. Cardiac cell therapies for the treatment of acute myocardial infarction in mice: systematic review and meta-analysis. Cytotherapy 2023; 25:640-652. [PMID: 36890093 DOI: 10.1016/j.jcyt.2023.01.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 03/08/2023]
Abstract
Backgound Aims: This meta-analysis aims at summarizing the whole body of research on cell therapies for acute myocardial infarction (MI) in the mouse model to bring forward ongoing research in this field of regenerative medicine. Despite rather modest effects in clinical trials, pre-clinical studies continue to report beneficial effects of cardiac cell therapies for cardiac repair following acute ischemic injury. Results: The authors' meta-analysis of data from 166 mouse studies comprising 257 experimental groups demonstrated a significant improvement in left ventricular ejection fraction of 10.21% after cell therapy compared with control animals. Subgroup analysis indicated that second-generation cell therapies such as cardiac progenitor cells and pluripotent stem cell derivatives had the highest therapeutic potential for minimizing myocardial damage post-MI. Conclusions: Whereas the vision of functional tissue replacement has been replaced by the concept of regional scar modulation in most of the investigated studies, rather basic methods for assessing cardiac function were most frequently used. Hence, future studies will highly benefit from integrating methods for assessment of regional wall properties to evolve a deeper understanding of how to modulate cardiac healing after acute MI.
Collapse
Affiliation(s)
| | - Anika Dahmen
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Praveen Vasudevan
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Heiko Lemcke
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Ralf Gäbel
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Alper Öner
- Department of Cardiology, Rostock University Medical Center, Rostock, Germany
| | - Hüseyin Ince
- Department of Cardiology, Rostock University Medical Center, Rostock, Germany
| | - Robert David
- Department of Cardiac Surgery, Rostock University Medical Center, Rostock, Germany; Department of Life, Light and Matter, University of Rostock, Rostock, Germany
| | - Markus Wolfien
- Institute of Medical Informatics and Biometry, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
25
|
Sanz-Ruiz R, Perin EC, Fernández-Avilés F. Cell therapy for heart failure: lessons learned from SCIENCE. Eur J Heart Fail 2023; 25:588-590. [PMID: 36823785 DOI: 10.1002/ejhf.2807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 02/19/2023] [Indexed: 02/25/2023] Open
Affiliation(s)
- Ricardo Sanz-Ruiz
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), Centro de Investigación Biomédica en Red - Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Francisco Fernández-Avilés
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañon (IiSGM), Centro de Investigación Biomédica en Red - Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
26
|
Qayyum AA, van Klarenbosch B, Frljak S, Cerar A, Poglajen G, Traxler-Weidenauer D, Nadrowski P, Paitazoglou C, Vrtovec B, Bergmann MW, Chamuleau SAJ, Wojakowski W, Gyöngyösi M, Kraaijeveld A, Hansen KS, Vrangbaek K, Jørgensen E, Helqvist S, Joshi FR, Johansen EM, Follin B, Juhl M, Højgaard LD, Mathiasen AB, Ekblond A, Haack-Sørensen M, Kastrup J. Effect of allogeneic adipose tissue-derived mesenchymal stromal cell treatment in chronic ischaemic heart failure with reduced ejection fraction - the SCIENCE trial. Eur J Heart Fail 2023; 25:576-587. [PMID: 36644821 DOI: 10.1002/ejhf.2772] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 11/27/2022] [Accepted: 01/08/2023] [Indexed: 01/17/2023] Open
Abstract
AIMS The aim of the SCIENCE trial was to investigate whether a single treatment with direct intramyocardial injections of adipose tissue-derived mesenchymal stromal cells (CSCC_ASCs) was safe and improved cardiac function in patients with chronic ischaemic heart failure with reduced ejection fraction (HFrEF). METHODS AND RESULTS The study was a European multicentre, double-blind, placebo-controlled phase II trial using allogeneic CSCC_ASCs from healthy donors or placebo (2:1 randomization). Main inclusion criteria were New York Heart Association (NYHA) class II-III, left ventricular ejection fraction (LVEF) <45%, and N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels >300 pg/ml. CSCC_ASCs or placebo (isotonic saline) were injected directly into viable myocardium. The primary endpoint was change in left ventricular end-systolic volume (LVESV) at 6-month follow-up measured by echocardiography. A total of 133 symptomatic HFrEF patients were included. The treatment was safe without any drug-related severe adverse events or difference in cardiac-related adverse events during a 3-year follow-up period. There were no significant differences between groups during follow-up in LVESV (0.3 ± 5.0 ml, p = 0.945), nor in secondary endpoints of left ventricular end-diastolic volume (-2.0 ± 6.0 ml, p = 0.736) and LVEF (-1.6 ± 1.0%, p = 0.119). The NYHA class improved slightly within the first year in both groups without any difference between groups. There were no changes in 6-min walk test, NT-proBNP, C-reactive protein or quality of life the first year in any groups. CONCLUSION The SCIENCE trial demonstrated safety of intramyocardial allogeneic CSCC_ASC therapy in patients with chronic HFrEF. However, it was not possible to improve the pre-defined endpoints and induce restoration of cardiac function or clinical symptoms.
Collapse
Affiliation(s)
- Abbas Ali Qayyum
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Bas van Klarenbosch
- Department of Cardiology and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sabina Frljak
- Advanced Heart Failure and Transplantation Center, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Andraz Cerar
- Advanced Heart Failure and Transplantation Center, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Gregor Poglajen
- Advanced Heart Failure and Transplantation Center, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | | | - Pawel Nadrowski
- Department of Cardiology and Structural Heart Diseases, Medical University of Silesia, Katowice, Poland
| | | | - Bojan Vrtovec
- Advanced Heart Failure and Transplantation Center, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Martin W Bergmann
- Department of Cardiology, Asklepios Klinik St. Georg, Hamburg, Germany
| | - Steven A J Chamuleau
- Department of Cardiology and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wojtek Wojakowski
- Department of Cardiology and Structural Heart Diseases, Medical University of Silesia, Katowice, Poland
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Adriaan Kraaijeveld
- Department of Cardiology and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kristian Schultz Hansen
- Faculty of Social Sciences and the Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karsten Vrangbaek
- Faculty of Social Sciences and the Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Erik Jørgensen
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Steffen Helqvist
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Francis Richard Joshi
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ellen Mønsted Johansen
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Bjarke Follin
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Morten Juhl
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lisbeth Drozd Højgaard
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anders Bruun Mathiasen
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Annette Ekblond
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Mandana Haack-Sørensen
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens Kastrup
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
27
|
Qayyum AA, Mouridsen M, Nilsson B, Gustafsson I, Schou M, Nielsen OW, Hove JD, Mathiasen AB, Jørgensen E, Helqvist S, Joshi FR, Johansen EM, Follin B, Juhl M, Højgaard LD, Haack-Sørensen M, Ekblond A, Kastrup J. Danish phase II trial using adipose tissue derived mesenchymal stromal cells for patients with ischaemic heart failure. ESC Heart Fail 2023; 10:1170-1183. [PMID: 36638837 PMCID: PMC10053281 DOI: 10.1002/ehf2.14281] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 12/10/2022] [Accepted: 12/15/2022] [Indexed: 01/15/2023] Open
Abstract
AIMS Patients suffering from chronic ischaemic heart failure with reduced left ventricular ejection fraction (HFrEF) have reduced quality-of-life, repetitive hospital admissions, and reduced life expectancy. Allogeneic cell therapy is currently investigated as a potential treatment option after initially encouraging results from clinical autologous and allogeneic trials in patients with HFrEF. We aimed to investigate the allogeneic Cardiology Stem Cell Centre Adipose tissue derived mesenchymal Stromal Cell product (CSCC_ASC) as an add-on therapy in patients with chronic HFrEF. METHODS AND RESULTS This is a Danish multi-centre double-blinded placebo-controlled phase II study with direct intra-myocardial injections of allogeneic CSCC_ASC. A total of 81 HFrEF patients were included and randomized 2:1 to CSCC_ASC or placebo injections. The inclusion criteria were reduced left ventricular ejection fraction (LVEF ≤ 45%), New York Heart Association (NYHA) class II-III despite optimal anti-congestive heart failure medication and no further revascularization options. Injections of 0.3 mL CSCC_ASC (total cell dose 100 × 106 ASCs) (n = 54) or isotonic saline (n = 27) were performed into the viable myocardium in the border zone of infarcted tissue using the NOGA Myostar® catheter (Biological Delivery System, Cordis, Johnson & Johnson, USA). The primary endpoint, left ventricular end systolic volume (LVESV), was evaluated at 6-month follow-up. The safety was measured during a 3-years follow-up period. RESULTS Mean age was 67.0 ± 9.0 years and 66.6 ± 8.1 years in the ASC and placebo groups, respectively. LVESV was unchanged from baseline to 6-month follow-up in the ASC (125.7 ± 68.8 mL and 126.3 ± 72.5 mL, P = 0.827) and placebo (134.6 ± 45.8 mL and 135.3 ± 49.6 mL, P = 0.855) group without any differences between the groups (0.0 mL (95% CI -9.1 to 9.0 mL, P = 0.992). Neither were there significant changes in left ventricular end diastolic volume or LVEF within the two groups or between groups -5.7 mL (95% CI -16.7 to 5.3 mL, P = 0.306) and -1.7% (95% CI -4.4. to 1.0, P = 0.212), respectively). NYHA classification and 6-min walk test did not alter significantly in the two groups (P > 0.05). The quality-of-life, total symptom, and overall summary score improved significantly only in the ASC group but not between groups. There were 24 serious adverse events (SAEs) in the ASC group and 11 SAEs in the placebo group without any significant differences between the two groups at 1-year follow-up. Kaplan-Meier plot using log-rank test of combined cardiac events showed an overall mean time to event of 30 ± 2 months in the ASC group and 29 ± 2 months in the placebo group without any differences between the groups during the 3 years follow-up period (P = 0.994). CONCLUSIONS Intramyocardial CSCC_ASC injections in patients with chronic HFrEF were safe but did not improve myocardial function or structure, nor clinical symptoms.
Collapse
Affiliation(s)
- Abbas Ali Qayyum
- Department of Cardiology and Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Department of Cardiology, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Mette Mouridsen
- Department of Cardiology, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Brian Nilsson
- Department of Cardiology, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ida Gustafsson
- Department of Cardiology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Morten Schou
- Department of Cardiology, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Olav Wendelboe Nielsen
- Department of Cardiology, Bispebjerg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jens Dahlgaard Hove
- Department of Cardiology, Hvidovre Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Anders Bruun Mathiasen
- Department of Cardiology and Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Erik Jørgensen
- Department of Cardiology and Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Steffen Helqvist
- Department of Cardiology and Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Francis Richard Joshi
- Department of Cardiology and Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ellen Mønsted Johansen
- Department of Cardiology and Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Bjarke Follin
- Department of Cardiology and Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Morten Juhl
- Department of Cardiology and Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lisbeth Drozd Højgaard
- Department of Cardiology and Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Mandana Haack-Sørensen
- Department of Cardiology and Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Annette Ekblond
- Department of Cardiology and Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens Kastrup
- Department of Cardiology and Cardiology Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
28
|
Whole-Heart Tissue Engineering and Cardiac Patches: Challenges and Promises. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010106. [PMID: 36671678 PMCID: PMC9855348 DOI: 10.3390/bioengineering10010106] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Despite all the advances in preventing, diagnosing, and treating cardiovascular disorders, they still account for a significant part of mortality and morbidity worldwide. The advent of tissue engineering and regenerative medicine has provided novel therapeutic approaches for the treatment of various diseases. Tissue engineering relies on three pillars: scaffolds, stem cells, and growth factors. Gene and cell therapy methods have been introduced as primary approaches to cardiac tissue engineering. Although the application of gene and cell therapy has resulted in improved regeneration of damaged cardiac tissue, further studies are needed to resolve their limitations, enhance their effectiveness, and translate them into the clinical setting. Scaffolds from synthetic, natural, or decellularized sources have provided desirable characteristics for the repair of cardiac tissue. Decellularized scaffolds are widely studied in heart regeneration, either as cell-free constructs or cell-seeded platforms. The application of human- or animal-derived decellularized heart patches has promoted the regeneration of heart tissue through in vivo and in vitro studies. Due to the complexity of cardiac tissue engineering, there is still a long way to go before cardiac patches or decellularized whole-heart scaffolds can be routinely used in clinical practice. This paper aims to review the decellularized whole-heart scaffolds and cardiac patches utilized in the regeneration of damaged cardiac tissue. Moreover, various decellularization methods related to these scaffolds will be discussed.
Collapse
|
29
|
Segovia F, Garcia H, Alkhateeb H, Mukherjee D, Nickel N. Updates in the Pharmacotherapy of Pulmonary Hypertension in Patients with Heart Failure with Preserved Ejection Fraction. Cardiovasc Hematol Disord Drug Targets 2023; 23:215-225. [PMID: 37921162 DOI: 10.2174/011871529x258234230921112507] [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: 05/31/2023] [Revised: 07/24/2023] [Accepted: 08/24/2023] [Indexed: 11/04/2023]
Abstract
Pulmonary hypertension (PH) associated with left heart disease (LHD) is a complex cardiopulmonary condition where a variable degree of pulmonary congestion, arterial vasoconstriction and vascular remodeling can lead to PH and right heart strain. Right heart dysfunction has a significant prognostic impact on these patients. Therefore, preserving right ventricular (RV) function is an important treatment goal. However, the treatment of PH in patients with left heart disease has produced conflicting evidence. The transition from pure LHD to LHD with PH is a continuum and clinically challenging. The heart failure with preserved ejection fraction (HFpEF) patient population is heterogeneous when it comes to PH and RV function. Appropriate clinical and hemodynamic phenotyping of patients with HFpEF and concomitant PH is paramount to making the appropriate treatment decision. This manuscript will summarize the current evidence for the use of pulmonary arterial vasodilators in patients with HFpEF.
Collapse
Affiliation(s)
- Fernando Segovia
- Department of Internal Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Hernando Garcia
- Pulmonary and Critical Care, Mount Sinai Medical Center, Miami, Florida, USA
| | - Haider Alkhateeb
- Division of Cardiovascular Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Debabrata Mukherjee
- Division of Cardiovascular Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| | - Nils Nickel
- Division of Pulmonary and Critical Care Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, USA
| |
Collapse
|
30
|
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
|
31
|
Cardiovascular Diseases in the Digital Health Era: A Translational Approach from the Lab to the Clinic. BIOTECH 2022; 11:biotech11030023. [PMID: 35892928 PMCID: PMC9326743 DOI: 10.3390/biotech11030023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/19/2022] [Accepted: 06/27/2022] [Indexed: 11/16/2022] Open
Abstract
Translational science has been introduced as the nexus among the scientific and the clinical field, which allows researchers to provide and demonstrate that the evidence-based research can connect the gaps present between basic and clinical levels. This type of research has played a major role in the field of cardiovascular diseases, where the main objective has been to identify and transfer potential treatments identified at preclinical stages into clinical practice. This transfer has been enhanced by the intromission of digital health solutions into both basic research and clinical scenarios. This review aimed to identify and summarize the most important translational advances in the last years in the cardiovascular field together with the potential challenges that still remain in basic research, clinical scenarios, and regulatory agencies.
Collapse
|
32
|
Gara E, Ong SG, Winkler J, Zlabinger K, Lukovic D, Merkely B, Emmert MY, Wolint P, Hoerstrup SP, Gyöngyösi M, Wu JC, Pavo N. Cell-Based HIF1α Gene Therapy Reduces Myocardial Scar and Enhances Angiopoietic Proteome, Transcriptomic and miRNA Expression in Experimental Chronic Left Ventricular Dysfunction. Front Bioeng Biotechnol 2022; 10:767985. [PMID: 35646882 PMCID: PMC9133350 DOI: 10.3389/fbioe.2022.767985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Recent preclinical investigations and clinical trials with stem cells mostly studied bone-marrow-derived mononuclear cells (BM-MNCs), which so far failed to meet clinically significant functional study endpoints. BM-MNCs containing small proportions of stem cells provide little regenerative potential, while mesenchymal stem cells (MSCs) promise effective therapy via paracrine impact. Genetic engineering for rationally enhancing paracrine effects of implanted stem cells is an attractive option for further development of therapeutic cardiac repair strategies. Non-viral, efficient transfection methods promise improved clinical translation, longevity and a high level of gene delivery. Hypoxia-induced factor 1α is responsible for pro-angiogenic, anti-apoptotic and anti-remodeling mechanisms. Here we aimed to apply a cellular gene therapy model in chronic ischemic heart failure in pigs. A non-viral circular minicircle DNA vector (MiCi) was used for in vitro transfection of porcine MSCs (pMSC) with HIF1α (pMSC-MiCi-HIF-1α). pMSCs-MiCi-HIF-1α were injected endomyocardially into the border zone of an anterior myocardial infarction one month post-reperfused-infarct. Cell injection was guided via 3D-guided NOGA electro-magnetic catheter delivery system. pMSC-MiCi-HIF-1α delivery improved cardiac output and reduced myocardial scar size. Abundances of pro-angiogenic proteins were analyzed 12, 24 h and 1 month after the delivery of the regenerative substances. In a protein array, the significantly increased angiogenesis proteins were Activin A, Angiopoietin, Artemin, Endothelin-1, MCP-1; and remodeling factors ADAMTS1, FGFs, TGFb1, MMPs, and Serpins. In a qPCR analysis, increased levels of angiopeptin, CXCL12, HIF-1α and miR-132 were found 24 h after cell-based gene delivery, compared to those in untreated animals with infarction and in control animals. Expression of angiopeptin increased already 12 h after treatment, and miR-1 expression was reduced at that time point. In total, pMSC overexpressing HIF-1α showed beneficial effects for treatment of ischemic injury, mediated by stimulation of angiogenesis.
Collapse
Affiliation(s)
- Edit Gara
- Heart and Vascular Centre, Semmelweis University, Budapest, Hungary
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Sang-Ging Ong
- Stanford Cardiovascular Institute, Stanford, CA, United States
| | - Johannes Winkler
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Katrin Zlabinger
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Dominika Lukovic
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Bela Merkely
- Heart and Vascular Centre, Semmelweis University, Budapest, Hungary
| | - Maximilian Y. Emmert
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- Department of Cardiovascular Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Wolint
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Simon P. Hoerstrup
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Mariann Gyöngyösi
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford, CA, United States
| | - Noemi Pavo
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
33
|
Chimenti I, Picchio V, Pagano F, Schirone L, Schiavon S, D'Ambrosio L, Valenti V, Forte M, di Nonno F, Rubattu S, Peruzzi M, Versaci F, Greco E, Calogero A, De Falco E, Frati G, Sciarretta S. The impact of autophagy modulation on phenotype and survival of cardiac stromal cells under metabolic stress. Cell Death Discov 2022; 8:149. [PMID: 35365624 PMCID: PMC8975847 DOI: 10.1038/s41420-022-00924-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 01/31/2022] [Accepted: 02/17/2022] [Indexed: 01/18/2023] Open
Abstract
Cardiac stromal cells (CSCs) embrace multiple phenotypes and are a contributory factor in tissue homeostasis and repair. They can be exploited as therapeutic mediators against cardiac fibrosis and remodeling, but their survival and cardioprotective properties can be decreased by microenvironmental cues. We evaluated the impact of autophagy modulation by different pharmacological/genetic approaches on the viability and phenotype of murine CSCs, which had been subjected to nutrient deprivation or hyperglycemia, in order to mimic relevant stress conditions and risk factors of cardiovascular diseases. Our results show that autophagy is activated in CSCs by nutrient deprivation, and that autophagy induction by trehalose or autophagy-related protein 7 (ATG7)-overexpression can significantly preserve CSC viability. Furthermore, autophagy induction is associated with a higher proportion of primitive, non-activated stem cell antigen 1 (Sca1)-positive cells, and with a reduced fibrotic fraction (positive for the discoidin domain-containing receptor 2, DDR2) in the CSC pool after nutrient deprivation. Hyperglycemia, on the other hand, is associated with reduced autophagic flux in CSCs, and with a significant reduction in primitive Sca1+ cells. Autophagy induction by adenoviral-mediated ATG7-overexpression maintains a cardioprotective, anti-inflammatory and pro-angiogenic paracrine profile of CSCs exposed to hyperglycemia for 1 week. Finally, autophagy induction by ATG7-overexpression during hyperglycemia can significantly preserve cell viability in CSCs, which were subsequently exposed to nutrient deprivation, reducing hyperglycemia-induced impairment of cell resistance to stress. In conclusion, our results show that autophagy stimulation preserves CSC viability and function in response to metabolic stressors, suggesting that it may boost the beneficial functions of CSCs in cardiac repair mechanisms.
Collapse
Affiliation(s)
- Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.
- Mediterranea Cardiocentro, Napoli, Italy.
| | - Vittorio Picchio
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Francesca Pagano
- Biochemistry and Cellular Biology Istitute, CNR, Monterotondo, Italy
| | - Leonardo Schirone
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Department of Clinical, Internal Medicine, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Sonia Schiavon
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Luca D'Ambrosio
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Valentina Valenti
- Haemodynamic and Cardiology Unit, "Santa Maria Goretti" Hospital, Latina, Italy
| | | | | | - Speranza Rubattu
- IRCCS Neuromed, Pozzilli, Italy
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Mariangela Peruzzi
- Mediterranea Cardiocentro, Napoli, Italy
- Department of Clinical, Internal Medicine, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesco Versaci
- Haemodynamic and Cardiology Unit, "Santa Maria Goretti" Hospital, Latina, Italy
- Department of System Medicine, "Tor Vergata" University, Rome, Italy
| | - Ernesto Greco
- Department of Clinical, Internal Medicine, Anaesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Antonella Calogero
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Elena De Falco
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Mediterranea Cardiocentro, Napoli, Italy
| | - Giacomo Frati
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Sebastiano Sciarretta
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
34
|
Johnston PV, Raval AN, Henry TD, Traverse JH, Pepine CJ. Dare to dream? Cell-based therapies for heart failure after DREAM-HF: Review and roadmap for future clinical study. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2022; 13:100118. [PMID: 38560073 PMCID: PMC10978179 DOI: 10.1016/j.ahjo.2022.100118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/28/2022] [Indexed: 04/04/2024]
Abstract
Clinical trials of cell-based therapies for heart failure have resulted in significant strides forward in our understanding of the potential the failing heart has for regeneration and repair. Yet, two decades on, the need for novel cell-based therapies for heart failure has never been greater. The DREAM-HF trial, which was presented as a late-breaking trial at the American Heart Association Scientific Sessions 2021 did not meet the primary heart failure outcome, but did show a large, clinically significant reduction in major adverse cardiovascular events (MACE) in patients receiving cells, an effect that was most pronounced in patients with evidence of maladaptive inflammation. These results represent an important step forward in our understanding of how cell-based therapies can exert beneficial effects in patients with heart failure and should serve as a guide for future clinical efforts. In light of the results of DREAM-HF, this review serves to provide an understanding of the current state of cell-based therapies for heart failure, as well as to highlight major knowledge gaps and suggest guiding principles for clinical trials of cell therapy going forward. Using the knowledge gained from DREAM-HF along with the trials that preceded it, the potential for breakthrough cell-based therapies for heart failure in the coming decade is immense.
Collapse
Affiliation(s)
- Peter V. Johnston
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Amish N. Raval
- Department of Medicine, Division of Cardiovascular Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| | - Timothy D. Henry
- Carl and Edyth Lindner Center for Research at the Christ Hospital, Cincinnati, OH, United States of America
| | - Jay H. Traverse
- Minneapolis Heart Institute Foundation at Abbot Northwestern Hospital, Minneapolis, MN, United States of America
| | - Carl J. Pepine
- Department of Medicine, Division of Cardiovascular Medicine, University of Florida, Gainesville, FL, United States of America
| |
Collapse
|
35
|
Cenko E, Badimon L, Bugiardini R, Claeys MJ, De Luca G, de Wit C, Derumeaux G, Dorobantu M, Duncker DJ, Eringa EC, Gorog DA, Hassager C, Heinzel FR, Huber K, Manfrini O, Milicic D, Oikonomou E, Padro T, Trifunovic-Zamaklar D, Vasiljevic-Pokrajcic Z, Vavlukis M, Vilahur G, Tousoulis D. Cardiovascular disease and COVID-19: a consensus paper from the ESC Working Group on Coronary Pathophysiology & Microcirculation, ESC Working Group on Thrombosis and the Association for Acute CardioVascular Care (ACVC), in collaboration with the European Heart Rhythm Association (EHRA). Cardiovasc Res 2021; 117:2705-2729. [PMID: 34528075 PMCID: PMC8500019 DOI: 10.1093/cvr/cvab298] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/10/2021] [Indexed: 01/08/2023] Open
Abstract
The cardiovascular system is significantly affected in coronavirus disease-19 (COVID-19). Microvascular injury, endothelial dysfunction, and thrombosis resulting from viral infection or indirectly related to the intense systemic inflammatory and immune responses are characteristic features of severe COVID-19. Pre-existing cardiovascular disease and viral load are linked to myocardial injury and worse outcomes. The vascular response to cytokine production and the interaction between severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and angiotensin-converting enzyme 2 receptor may lead to a significant reduction in cardiac contractility and subsequent myocardial dysfunction. In addition, a considerable proportion of patients who have been infected with SARS-CoV-2 do not fully recover and continue to experience a large number of symptoms and post-acute complications in the absence of a detectable viral infection. This conditions often referred to as 'post-acute COVID-19' may have multiple causes. Viral reservoirs or lingering fragments of viral RNA or proteins contribute to the condition. Systemic inflammatory response to COVID-19 has the potential to increase myocardial fibrosis which in turn may impair cardiac remodelling. Here, we summarize the current knowledge of cardiovascular injury and post-acute sequelae of COVID-19. As the pandemic continues and new variants emerge, we can advance our knowledge of the underlying mechanisms only by integrating our understanding of the pathophysiology with the corresponding clinical findings. Identification of new biomarkers of cardiovascular complications, and development of effective treatments for COVID-19 infection are of crucial importance.
Collapse
Affiliation(s)
- Edina Cenko
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Via Giuseppe Massarenti 9, 40134 Bologna, Italy
| | - Lina Badimon
- Cardiovascular Program ICCC-Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, CiberCV, Barcelona, Spain
| | - Raffaele Bugiardini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Via Giuseppe Massarenti 9, 40134 Bologna, Italy
| | - Marc J Claeys
- Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| | - Giuseppe De Luca
- Cardiovascular Department of Cardiology, Ospedale “Maggiore della Carità”, Eastern Piedmont University, Novara, Italy
| | - Cor de Wit
- Institut für Physiologie, Universität zu Lübeck, Lübeck, Germany
- Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V. (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, Lübeck, Germany
| | - Geneviève Derumeaux
- IMRB U955, UPEC, Créteil, France
- Department of Physiology, AP-HP, Henri-Mondor Teaching Hospital, Créteil, France
- Fédération Hospitalo-Universitaire « SENEC », Créteil, France
| | - Maria Dorobantu
- “Carol Davila” University of Medicine and Pharmacy, Bucharest, Romania
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Etto C Eringa
- Department of Physiology, Amsterdam Cardiovascular Science Institute, Amsterdam University Medical Centres, Amsterdam, The Netherlands
- Department of Physiology, Maastricht University, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, The Netherlands
| | - Diana A Gorog
- Faculty of Medicine, National Heart and Lung Institute, Imperial College, London, UK
- Department of Postgraduate Medicine, University of Hertfordshire, Hatfield, UK
| | - Christian Hassager
- Department of Cardiology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Frank R Heinzel
- Department of Cardiology, Charité-Universitaetsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Kurt Huber
- 3rd Medical Department, Cardiology and Intensive Care Medicine, Wilhelminen Hospital, Vienna, Austria
- Medical School, Sigmund Freud University, Vienna, Austria
| | - Olivia Manfrini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Via Giuseppe Massarenti 9, 40134 Bologna, Italy
| | - Davor Milicic
- Department of Cardiovascular Diseases, University Hospital Centre Zagreb, University of Zagreb, Zagreb, Croatia
| | - Evangelos Oikonomou
- Department of Cardiology, ‘Hippokration’ General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Teresa Padro
- Cardiovascular Program ICCC-Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, CiberCV, Barcelona, Spain
| | - Danijela Trifunovic-Zamaklar
- Cardiology Department, Clinical Centre of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | - Marija Vavlukis
- University Clinic of Cardiology, Medical Faculty, Ss' Cyril and Methodius University in Skopje, Skopje, Republic of Macedonia
| | - Gemma Vilahur
- Cardiovascular Program ICCC-Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, CiberCV, Barcelona, Spain
| | - Dimitris Tousoulis
- Department of Cardiology, ‘Hippokration’ General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
36
|
Yamada S, Bartunek J, Behfar A, Terzic A. Mass Customized Outlook for Regenerative Heart Failure Care. Int J Mol Sci 2021; 22:11394. [PMID: 34768825 PMCID: PMC8583673 DOI: 10.3390/ijms222111394] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/01/2021] [Accepted: 10/18/2021] [Indexed: 11/16/2022] Open
Abstract
Heart failure pathobiology is permissive to reparative intent. Regenerative therapies exemplify an emerging disruptive innovation aimed at achieving structural and functional organ restitution. However, mixed outcomes, complexity in use, and unsustainable cost have curtailed broader adoption, mandating the development of novel cardio-regenerative approaches. Lineage guidance offers a standardized path to customize stem cell fitness for therapy. A case in point is the molecular induction of the cardiopoiesis program in adult stem cells to yield cardiopoietic cell derivatives designed for heart failure treatment. Tested in early and advanced clinical trials in patients with ischemic heart failure, clinical grade cardiopoietic cells were safe and revealed therapeutic improvement within a window of treatment intensity and pre-treatment disease severity. With the prospect of mass customization, cardiopoietic guidance has been streamlined from the demanding, recombinant protein cocktail-based to a protein-free, messenger RNA-based single gene protocol to engineer affordable cardiac repair competent cells. Clinical trial biobanked stem cells enabled a systems biology deconvolution of the cardiopoietic cell secretome linked to therapeutic benefit, exposing a paracrine mode of action. Collectively, this new knowledge informs next generation regenerative therapeutics manufactured as engineered cellular or secretome mimicking cell-free platforms. Launching biotherapeutics tailored for optimal outcome and offered at mass production cost would contribute to advancing equitable regenerative care that addresses population health needs.
Collapse
Affiliation(s)
- Satsuki Yamada
- Center for Regenerative Medicine, Marriott Family Comprehensive Cardiac Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (S.Y.); (A.B.)
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jozef Bartunek
- Cardiovascular Center, OLV Hospital, 9300 Aalst, Belgium
| | - Atta Behfar
- Center for Regenerative Medicine, Marriott Family Comprehensive Cardiac Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (S.Y.); (A.B.)
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Andre Terzic
- Center for Regenerative Medicine, Marriott Family Comprehensive Cardiac Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA; (S.Y.); (A.B.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
37
|
Tripathi A, Khan MS, Khan AR, Vaughn VM, Bolli R. Cell therapy for nonischemic dilated cardiomyopathy: A systematic review and meta-analysis of randomized controlled trials. Stem Cells Transl Med 2021; 10:1394-1405. [PMID: 34346555 PMCID: PMC8459637 DOI: 10.1002/sctm.21-0094] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/18/2021] [Accepted: 06/16/2021] [Indexed: 01/03/2023] Open
Abstract
Cell therapy involves transplantation of human cells to promote repair of diseased or injured tissues and/or cells. Only a limited number of mostly small-scale trials have studied cell therapy in nonischemic cardiomyopathy (NICM). We performed a meta-analysis of randomized clinical trials (RCTs) to assess the safety and efficacy of cell therapy in NICM. Electronic databases were searched for relevant RCTs from inception until August 2020. Outcomes assessed were left ventricular ejection fraction (LVEF), left ventricular end-diastolic diameter or volume (LVEDD), quality of life (QoL) indices, and major adverse cardiac events (MACEs). Weighted mean differences (MDs) and standardized mean differences (SMDs) were calculated using random-effects methods. Eleven RCTs with 574 participants were included in the analysis. There was a significant increase in mean LVEF (MD, 4.17%; 95% confidence interval [CI] = 1.66-6.69) and modest decrease in LVEDD (SMD, -0.50; 95% CI = -0.95 to -0.06) in patients treated with cell therapy compared with controls. Cell therapy was also associated with improvement in functional capacity, as assessed by the 6-minute walking distance (MD, 72.49 m; 95% CI = 3.44-141.53). No significant differences were seen in MACEs and QoL indices between treated and control groups. This meta-analysis suggests that cell therapy may improve LV systolic function and may be associated with improvement in LVEDD and functional capacity compared with maximal medical therapy. Cell therapy was safe, with no significant difference in MACEs between treatment and control groups. However, given the limitations of current studies, larger well-designed RCTs are needed to evaluate the efficacy of cell therapy in patients with NICM.
Collapse
Affiliation(s)
- Avnish Tripathi
- Division of CardiologyUniversity of Kentucky College of MedicineBowling GreenKentuckyUSA
| | - Mohammad Saud Khan
- Division of CardiologyUniversity of Kentucky College of MedicineBowling GreenKentuckyUSA
- Department of CardiologyCheyenne Regional Medical CenterCheyenneWyomingUSA
| | - Abdur Rahman Khan
- Kornhauser Health Science LibraryUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Vida M. Vaughn
- Kornhauser Health Science LibraryUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Roberto Bolli
- Institute of Molecular CardiologyUniversity of LouisvilleLouisvilleKentuckyUSA
| |
Collapse
|
38
|
Hesselbarth R, Esser TU, Roshanbinfar K, Schrüfer S, Schubert DW, Engel FB. CHIR99021 Promotes hiPSC-Derived Cardiomyocyte Proliferation in Engineered 3D Microtissues. Adv Healthc Mater 2021; 10:e2100926. [PMID: 34499814 PMCID: PMC11468594 DOI: 10.1002/adhm.202100926] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/30/2021] [Indexed: 02/05/2023]
Abstract
Cardiac tissue engineering is a promising strategy to generate human cardiac tissues for modeling cardiac diseases, screening for therapeutic drugs, and repairing the injured heart. Yet, several issues remain to be resolved including the generation of tissues with high cardiomyocyte density. Here, it is shown that the integration of the glycogen synthase kinase-3β inhibitor CHIR99021 in collagen I hydrogels promotes proliferation of human-induced pluripotent stem cell-derived (hiPSC) cardiomyocytes post-fabrication improving contractility of and calcium flow in engineered 3D cardiac microtissues. CHIR99021 has no effect on the gelation kinetics or the mechanical properties of collagen I hydrogels. Analysis of cell density and proliferation based on Ki-67 staining indicates that integration of CHIR99021 together with external CHIR99021 stimulation increases hiPSC-cardiomyocyte number by ≈2-fold within 7 d post-fabrication. Analysis of the contractility of engineered cardiac tissues after another 3 d in the absence of external CHIR99021 shows that CHIR99021-induced hiPSC-cardiomyocyte proliferation results in synchronized calcium flow, rhythmic beating, increased speed of contraction and contraction amplitude, and reduced peak-to-peak time. The CHIR99021-stimulated engineered cardiac microtissues exhibit spontaneous rhythmic contractions for at least 35 d. Collectively, the data demonstrate the potential of induced cardiomyocyte proliferation to enhance engineered cardiac microtissues by increasing cardiomyocyte density.
Collapse
Affiliation(s)
- Ramona Hesselbarth
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of PathologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Muscle Research Center Erlangen (MURCE)Erlangen91054Germany
| | - Tilman U. Esser
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of PathologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Muscle Research Center Erlangen (MURCE)Erlangen91054Germany
| | - Kaveh Roshanbinfar
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of PathologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Muscle Research Center Erlangen (MURCE)Erlangen91054Germany
| | - Stefan Schrüfer
- Institute of Polymer MaterialsFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Martensstraße 7Erlangen91058Germany
| | - Dirk W. Schubert
- Institute of Polymer MaterialsFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Martensstraße 7Erlangen91058Germany
| | - Felix B. Engel
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of PathologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Muscle Research Center Erlangen (MURCE)Erlangen91054Germany
| |
Collapse
|
39
|
Garmany A, Yamada S, Terzic A. Longevity leap: mind the healthspan gap. NPJ Regen Med 2021; 6:57. [PMID: 34556664 PMCID: PMC8460831 DOI: 10.1038/s41536-021-00169-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 09/02/2021] [Indexed: 02/08/2023] Open
Abstract
Life expectancy has increased by three decades since the mid-twentieth century. Parallel healthspan expansion has however not followed, largely impeded by the pandemic of chronic diseases afflicting a growing older population. The lag in quality of life is a recognized challenge that calls for prioritization of disease-free longevity. Contemporary communal, clinical and research trends aspiring to extend the health horizon are here outlined in the context of an evolving epidemiology. A shared action integrating public and societal endeavors with emerging interventions that target age-related multimorbidity and frailty is needed. A multidimensional buildout of a curative perspective, boosted by modern anti-senescent and regenerative technology with augmented decision making, would require dedicated resources and cost-effective validation to responsibly bridge the healthspan-lifespan gap for a future of equitable global wellbeing.
Collapse
Affiliation(s)
- Armin Garmany
- Center for Regenerative Medicine, Marriott Family Comprehensive Cardiac Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Alix School of Medicine, Regenerative Sciences Track, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
| | - Satsuki Yamada
- Center for Regenerative Medicine, Marriott Family Comprehensive Cardiac Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, MN, USA
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Andre Terzic
- Center for Regenerative Medicine, Marriott Family Comprehensive Cardiac Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, MN, USA.
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA.
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
40
|
The Essential Need for a Validated Potency Assay for Cell-Based Therapies in Cardiac Regenerative and Reparative Medicine. A Practical Approach to Test Development. Stem Cell Rev Rep 2021; 17:2235-2244. [PMID: 34463902 PMCID: PMC8599250 DOI: 10.1007/s12015-021-10244-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2021] [Indexed: 01/04/2023]
Abstract
Biological treatments are one of the medical breakthroughs in the twenty-first century. The initial enthusiasm pushed the field towards indiscriminatory use of cell therapy regardless of the pathophysiological particularities of underlying conditions. In the reparative and regenerative cardiovascular field, the results of the over two decades of research in cell-based therapies, although promising still could not be translated into clinical scenario. Now, when we identified possible deficiencies and try to rebuild its foundations rigorously on scientific evidence, development of potency assays for the potential therapeutic product is one of the steps which will bring our goal of clinical translation closer. Although, highly challenging, the potency tests for cell products are considered as a priority by the regulatory agencies. In this paper we describe the main characteristics and challenges for a cell therapy potency test focusing on the cardiovascular field. Moreover, we discuss different steps and types of assays that should be taken into consideration for an eventual potency test development by tying together two fundamental concepts: target disease and expected mechanism of action.
Collapse
|
41
|
Deuse T, Tediashvili G, Hu X, Gravina A, Tamenang A, Wang D, Connolly A, Mueller C, Mallavia B, Looney MR, Alawi M, Lanier LL, Schrepfer S. Hypoimmune induced pluripotent stem cell-derived cell therapeutics treat cardiovascular and pulmonary diseases in immunocompetent allogeneic mice. Proc Natl Acad Sci U S A 2021; 118:e2022091118. [PMID: 34244428 PMCID: PMC8285900 DOI: 10.1073/pnas.2022091118] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The emerging field of regenerative cell therapy is still limited by the few cell types that can reliably be differentiated from pluripotent stem cells and by the immune hurdle of commercially scalable allogeneic cell therapeutics. Here, we show that gene-edited, immune-evasive cell grafts can survive and successfully treat diseases in immunocompetent, fully allogeneic recipients. Transplanted endothelial cells improved perfusion and increased the likelihood of limb preservation in mice with critical limb ischemia. Endothelial cell grafts transduced to express a transgene for alpha1-antitrypsin (A1AT) successfully restored physiologic A1AT serum levels in mice with genetic A1AT deficiency. This cell therapy prevented both structural and functional changes of emphysematous lung disease. A mixture of endothelial cells and cardiomyocytes was injected into infarcted mouse hearts, and both cell types orthotopically engrafted in the ischemic areas. Cell therapy led to an improvement in invasive hemodynamic heart failure parameters. Our study supports the development of hypoimmune, universal regenerative cell therapeutics for cost-effective treatments of major diseases.
Collapse
Affiliation(s)
- Tobias Deuse
- Division of Cardiothoracic Surgery, Department of Surgery, Transplant and Stem Cell Immunobiology Laboratory, University of California, San Francisco, CA 94143
| | - Grigol Tediashvili
- Division of Cardiothoracic Surgery, Department of Surgery, Transplant and Stem Cell Immunobiology Laboratory, University of California, San Francisco, CA 94143
- Department of Cardiovascular Surgery, University Heart Center Hamburg, 20246 Hamburg, Germany
| | - Xiaomeng Hu
- Division of Cardiothoracic Surgery, Department of Surgery, Transplant and Stem Cell Immunobiology Laboratory, University of California, San Francisco, CA 94143
- Department of Cardiovascular Surgery, University Heart Center Hamburg, 20246 Hamburg, Germany
- German Center for Cardiovascular Research (DZHK) partner site Hamburg/Kiel/Luebeck, 20246 Hamburg, Germany
- Sana Biotechnology Inc., South San Francisco, CA 94080
| | - Alessia Gravina
- Division of Cardiothoracic Surgery, Department of Surgery, Transplant and Stem Cell Immunobiology Laboratory, University of California, San Francisco, CA 94143
| | - Annika Tamenang
- Division of Cardiothoracic Surgery, Department of Surgery, Transplant and Stem Cell Immunobiology Laboratory, University of California, San Francisco, CA 94143
- Department of Cardiovascular Surgery, University Heart Center Hamburg, 20246 Hamburg, Germany
| | - Dong Wang
- Division of Cardiothoracic Surgery, Department of Surgery, Transplant and Stem Cell Immunobiology Laboratory, University of California, San Francisco, CA 94143
| | - Andrew Connolly
- Department of Pathology, University of California, San Francisco, CA 94143
| | - Christian Mueller
- Horae Gene Therapy Center, University of Massachusetts, Worcester, MA 01605
- Department of Pediatrics, University of Massachusetts, Worcester, MA 01605
| | - Beñat Mallavia
- Department of Medicine, University of California, San Francisco, CA 94143
| | - Mark R Looney
- Department of Medicine, University of California, San Francisco, CA 94143
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143
| | - Malik Alawi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lewis L Lanier
- Department of Microbiology and Immunology and the Parker Institute for Cancer Immunotherapy, University of California, San Francisco, CA 94143
| | - Sonja Schrepfer
- Division of Cardiothoracic Surgery, Department of Surgery, Transplant and Stem Cell Immunobiology Laboratory, University of California, San Francisco, CA 94143;
- Sana Biotechnology Inc., South San Francisco, CA 94080
| |
Collapse
|
42
|
Yamada S, Jeon R, Garmany A, Behfar A, Terzic A. Screening for regenerative therapy responders in heart failure. Biomark Med 2021; 15:775-783. [PMID: 34169733 PMCID: PMC8252977 DOI: 10.2217/bmm-2020-0683] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/23/2021] [Indexed: 12/20/2022] Open
Abstract
Risk of outcome variability challenges therapeutic innovation. Selection of the most suitable candidates is predicated on reliable response indicators. Especially for emergent regenerative biotherapies, determinants separating success from failure in achieving disease rescue remain largely unknown. Accordingly, (pre)clinical development programs have placed increased emphasis on the multi-dimensional decoding of repair capacity and disease resolution, attributes defining responsiveness. To attain regenerative goals for each individual, phenotype-based patient selection is poised for an upgrade guided by new insights into disease biology, translated into refined surveillance of response regulators and deep learning-amplified clinical decision support.
Collapse
Affiliation(s)
- Satsuki Yamada
- Department of Cardiovascular Medicine, Mayo Clinic, Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Rochester, MN 55905, USA
- Department of Medicine, Division of Geriatric Medicine & Gerontology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ryounghoon Jeon
- Department of Cardiovascular Medicine, Mayo Clinic, Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Rochester, MN 55905, USA
| | - Armin Garmany
- Department of Cardiovascular Medicine, Mayo Clinic, Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Rochester, MN 55905, USA
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine, Regenerative Sciences Track, Rochester, MN 55905, USA
| | - Atta Behfar
- Department of Cardiovascular Medicine, Mayo Clinic, Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Rochester, MN 55905, USA
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Andre Terzic
- Department of Cardiovascular Medicine, Mayo Clinic, Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Rochester, MN 55905, USA
- Department of Molecular Pharmacology & Experimental Therapeutics, Department of Clinical Genomics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
43
|
Liu M, López de Juan Abad B, Cheng K. Cardiac fibrosis: Myofibroblast-mediated pathological regulation and drug delivery strategies. Adv Drug Deliv Rev 2021; 173:504-519. [PMID: 33831476 PMCID: PMC8299409 DOI: 10.1016/j.addr.2021.03.021] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/16/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023]
Abstract
Cardiac fibrosis remains an unresolved problem in heart diseases. After initial injury, cardiac fibroblasts (CFs) are activated and subsequently differentiate into myofibroblasts (myoFbs) that are major mediator cells in the pathological remodeling. MyoFbs exhibit proliferative and secretive characteristics, and contribute to extracellular matrix (ECM) turnover, collagen deposition. The persistent functions of myoFbs lead to fibrotic scars and cardiac dysfunction. The anti-fibrotic treatment is hindered by the elusive mechanism of fibrosis and lack of specific targets on myoFbs. In this review, we will outline the progress of cardiac fibrosis and its contributions to the heart failure. We will also shed light on the role of myoFbs in the regulation of adverse remodeling. The communication between myoFbs and other cells that are involved in the heart injury and repair respectively will be reviewed in detail. Then, recently developed therapeutic strategies to treat fibrosis will be summarized such as i) chimeric antigen receptor T cell (CAR-T) therapy with an optimal target on myoFbs, ii) direct reprogramming from stem cells to quiescent CFs, iii) "off-target" small molecular drugs. The application of nano/micro technology will be discussed as well, which is involved in the construction of cell-based biomimic platforms and "pleiotropic" drug delivery systems.
Collapse
Affiliation(s)
- Mengrui Liu
- Department of Molecular Biomedical Sciences, North Carolina State University, NC, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
| | - Blanca López de Juan Abad
- Department of Molecular Biomedical Sciences, North Carolina State University, NC, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences, North Carolina State University, NC, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA.
| |
Collapse
|
44
|
Arrell DK, Crespo-Diaz RJ, Yamada S, Jeon R, Garmany A, Park S, Adolf JP, Livia C, Hillestad ML, Bartunek J, Behfar A, Terzic A. Secretome signature of cardiopoietic cells echoed in rescued infarcted heart proteome. Stem Cells Transl Med 2021; 10:1320-1328. [PMID: 34047493 PMCID: PMC8380441 DOI: 10.1002/sctm.20-0509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/17/2021] [Accepted: 04/20/2021] [Indexed: 12/27/2022] Open
Abstract
Stem cell paracrine activity is implicated in cardiac repair. Linkage between secretome functionality and therapeutic outcome was here interrogated by systems analytics of biobanked human cardiopoietic cells, a regenerative biologic in advanced clinical trials. Protein chip array identified 155 proteins differentially secreted by cardiopoietic cells with clinical benefit, expanded into a 520 node network, collectively revealing inherent vasculogenic properties along with cardiac and smooth muscle differentiation and development. Next generation RNA sequencing, refined by pathway analysis, pinpointed miR-146 dependent regulation upstream of the decoded secretome. Intracellular and extracellular integration unmasked commonality across cardio-vasculogenic processes. Mirroring the secretome pattern, infarcted hearts benefiting from cardiopoietic cell therapy restored the disease proteome engaging cardiovascular system functions. The cardiopoietic cell secretome thus confers a therapeutic molecular imprint on recipient hearts, with response informed by predictive systems profiling.
Collapse
Affiliation(s)
- D Kent Arrell
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA
| | - Ruben J Crespo-Diaz
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA.,Cardiovascular Division, University of Minnesota, Minneapolis, Minnesota, USA
| | - Satsuki Yamada
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Division of Geriatric & Gerontology Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Ryounghoon Jeon
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Armin Garmany
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Alix School of Medicine, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Sungjo Park
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jeffrey P Adolf
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Christopher Livia
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Mayo Clinic Alix School of Medicine, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Matthew L Hillestad
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Atta Behfar
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA.,Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Andre Terzic
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota, USA.,Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
45
|
Popescu S, Preda MB, Marinescu CI, Simionescu M, Burlacu A. Dual Stem Cell Therapy Improves the Myocardial Recovery Post-Infarction through Reciprocal Modulation of Cell Functions. Int J Mol Sci 2021; 22:ijms22115631. [PMID: 34073327 PMCID: PMC8199446 DOI: 10.3390/ijms22115631] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/21/2021] [Accepted: 05/23/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal cells (MSC) are promising candidates for regenerative therapy of the infarcted heart. However, poor cell retention within the transplantation site limits their potential. We hypothesized that MSC benefits could be enhanced through a dual-cell approach using jointly endothelial colony forming cells (ECFC) and MSC. To assess this, we comparatively evaluated the effects of the therapy with MSC and ECFC versus MSC-only in a mouse model of myocardial infarction. Heart function was assessed by echocardiography, and the molecular crosstalk between MSC and ECFC was evaluated in vitro through direct or indirect co-culture systems. We found that dual-cell therapy improved cardiac function in terms of ejection fraction and stroke volume. In vitro experiments showed that ECFC augmented MSC effector properties by increasing Connexin 43 and Integrin alpha-5 and the secretion of healing-associated molecules. Moreover, MSC prompted the organization of ECFC into vascular networks. This indicated a reciprocal modulation in the functionality of MSC and ECFC. In conclusion, the crosstalk between MSC and ECFC augments the therapeutic properties of MSC and enhances the angiogenic properties of ECFC. Our data consolidate the dual-cell therapy as a step forward for the development of effective treatments for patients affected by myocardial infarction.
Collapse
|
46
|
Bolli R, Solankhi M, Tang XL, Kahlon A. Cell Therapy in Patients with Heart Failure: A Comprehensive Review and Emerging Concepts. Cardiovasc Res 2021; 118:951-976. [PMID: 33871588 PMCID: PMC8930075 DOI: 10.1093/cvr/cvab135] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 04/15/2021] [Indexed: 12/16/2022] Open
Abstract
This review summarizes the results of clinical trials of cell therapy in patients with heart failure (HF). In contrast to acute myocardial infarction (where results have been consistently negative for more than a decade), in the setting of HF the results of Phase I–II trials are encouraging, both in ischaemic and non-ischaemic cardiomyopathy. Several well-designed Phase II studies have met their primary endpoint and demonstrated an efficacy signal, which is remarkable considering that only one dose of cells was used. That an efficacy signal was seen 6–12 months after a single treatment provides a rationale for larger, rigorous trials. Importantly, no safety concerns have emerged. Amongst the various cell types tested, mesenchymal stromal cells derived from bone marrow (BM), umbilical cord, or adipose tissue show the greatest promise. In contrast, embryonic stem cells are not likely to become a clinical therapy. Unfractionated BM cells and cardiosphere-derived cells have been abandoned. The cell products used for HF will most likely be allogeneic. New approaches, such as repeated cell treatment and intravenous delivery, may revolutionize the field. As is the case for most new therapies, the development of cell therapies for HF has been slow, plagued by multifarious problems, and punctuated by many setbacks; at present, the utility of cell therapy in HF remains to be determined. What the field needs is rigorous, well-designed Phase III trials. The most important things to move forward are to keep an open mind, avoid preconceived notions, and let ourselves be guided by the evidence.
Collapse
Affiliation(s)
- Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY 40292
| | - Mitesh Solankhi
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY 40292
| | - Xiang-Liang Tang
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY 40292
| | - Arunpreet Kahlon
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY 40292
| |
Collapse
|
47
|
Bolli R, Mitrani RD, Hare JM, Pepine CJ, Perin EC, Willerson JT, Traverse JH, Henry TD, Yang PC, Murphy MP, March KL, Schulman IH, Ikram S, Lee DP, O’Brien C, Lima JA, Ostovaneh MR, Ambale-Venkatesh B, Lewis G, Khan A, Bacallao K, Valasaki K, Longsomboon B, Gee AP, Richman S, Taylor DA, Lai D, Sayre SL, Bettencourt J, Vojvodic RW, Cohen ML, Simpson L, Aguilar D, Loghin C, Moyé L, Ebert RF, Davis BR, Simari RD. A Phase II study of autologous mesenchymal stromal cells and c-kit positive cardiac cells, alone or in combination, in patients with ischaemic heart failure: the CCTRN CONCERT-HF trial. Eur J Heart Fail 2021; 23:661-674. [PMID: 33811444 PMCID: PMC8357352 DOI: 10.1002/ejhf.2178] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/21/2022] Open
Abstract
AIMS CONCERT-HF is an NHLBI-sponsored, double-blind, placebo-controlled, Phase II trial designed to determine whether treatment with autologous bone marrow-derived mesenchymal stromal cells (MSCs) and c-kit positive cardiac cells (CPCs), given alone or in combination, is feasible, safe, and beneficial in patients with heart failure (HF) caused by ischaemic cardiomyopathy. METHODS AND RESULTS Patients were randomized (1:1:1:1) to transendocardial injection of MSCs combined with CPCs, MSCs alone, CPCs alone, or placebo, and followed for 12 months. Seven centres enrolled 125 participants with left ventricular ejection fraction of 28.6 ± 6.1% and scar size 19.4 ± 5.8%, in New York Heart Association class II or III. The proportion of major adverse cardiac events (MACE) was significantly decreased by CPCs alone (-22% vs. placebo, P = 0.043). Quality of life (Minnesota Living with Heart Failure Questionnaire score) was significantly improved by MSCs alone (P = 0.050) and MSCs + CPCs (P = 0.023) vs. placebo. Left ventricular ejection fraction, left ventricular volumes, scar size, 6-min walking distance, and peak oxygen consumption did not differ significantly among groups. CONCLUSIONS This is the first multicentre trial assessing CPCs and a combination of two cell types from different tissues in HF patients. The results show that treatment is safe and feasible. Even with maximal guideline-directed therapy, both CPCs and MSCs were associated with improved clinical outcomes (MACE and quality of life, respectively) in ischaemic HF without affecting left ventricular function or structure, suggesting possible systemic or paracrine cellular mechanisms. Combining MSCs with CPCs was associated with improvement in both these outcomes. These results suggest potential important beneficial effects of CPCs and MSCs and support further investigation in HF patients.
Collapse
Affiliation(s)
- Roberto Bolli
- University of Louisville, School of Medicine, Louisville, KY, USA
| | - Raul D. Mitrani
- University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Joshua M. Hare
- University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Carl J. Pepine
- University of Florida College of Medicine, Gainesville, FL, USA
| | - Emerson C. Perin
- Texas Heart Institute, CHI St. Luke’s Health Baylor College of Medicine Medical Center, Houston, TX, USA
| | - James T. Willerson
- Texas Heart Institute, CHI St. Luke’s Health Baylor College of Medicine Medical Center, Houston, TX, USA
| | - Jay H. Traverse
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, and University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Timothy D. Henry
- The Carl and Edyth Lindner Center for Research and Education, The Christ Hospital, Cincinnati, OH, USA
| | | | | | - Keith L. March
- University of Florida College of Medicine, Gainesville, FL, USA
| | | | - Sohail Ikram
- University of Louisville, School of Medicine, Louisville, KY, USA
| | - David P. Lee
- Stanford University School of Medicine, Stanford, CA, USA
| | - Connor O’Brien
- Stanford University School of Medicine, Stanford, CA, USA
| | - Joao A. Lima
- Johns Hopkins University, Cardiovascular Imaging, Baltimore, MD, USA
| | | | | | - Gregory Lewis
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aisha Khan
- University of Miami, Miller School of Medicine, Interdisciplinary Stem Cell Institute, Miami, FL, USA
| | - Ketty Bacallao
- University of Miami, Miller School of Medicine, Interdisciplinary Stem Cell Institute, Miami, FL, USA
| | - Krystalenia Valasaki
- University of Miami, Miller School of Medicine, Interdisciplinary Stem Cell Institute, Miami, FL, USA
| | - Bangon Longsomboon
- University of Miami, Miller School of Medicine, Interdisciplinary Stem Cell Institute, Miami, FL, USA
| | - Adrian P. Gee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Sara Richman
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Doris A. Taylor
- Texas Heart Institute, CHI St. Luke’s Health Baylor College of Medicine Medical Center, Houston, TX, USA
| | - Dejian Lai
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Shelly L. Sayre
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Judy Bettencourt
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Rachel W. Vojvodic
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Michelle L. Cohen
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Lara Simpson
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - David Aguilar
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
- UTHealth University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - Catalin Loghin
- UTHealth University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - Lem Moyé
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Ray F. Ebert
- NIH, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Barry R. Davis
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | | | | |
Collapse
|
48
|
Yamada S, Behfar A, Terzic A. Regenerative medicine clinical readiness. Regen Med 2021; 16:309-322. [PMID: 33622049 PMCID: PMC8050983 DOI: 10.2217/rme-2020-0178] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/05/2021] [Indexed: 02/06/2023] Open
Abstract
Regenerative medicine, poised to transform 21st century healthcare, has aspired to enrich care options by bringing cures to patients in need. Science-driven responsible and regulated translation of innovative technology has enabled the launch of previously unimaginable care pathways adopted prudently for select serious diseases and disabilities. The collective resolve to advance the design, manufacture and validity of affordable regenerative solutions aims to democratize such health benefits for all. The objective of this Review is to outline the framework and prerequisites that underpin clinical readiness of regenerative care. Integrated research and development, specialized workforce education and accessible evidence-based practice implementation are at the core of realizing an equitable regenerative medicine vision.
Collapse
Affiliation(s)
- Satsuki Yamada
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, 55905 MN, USA
- Division of Geriatric Medicine & Gerontology, Department of Medicine, Mayo Clinic, Rochester, 55905 MN, USA
| | - Atta Behfar
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, 55905 MN, USA
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, 55905 MN, USA
| | - Andre Terzic
- Center for Regenerative Medicine, Marriott Heart Disease Research Program, Van Cleve Cardiac Regenerative Medicine Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, 55905 MN, USA
- Department of Molecular Pharmacology & Experimental Therapeutics, Department of Clinical Genomics, Mayo Clinic, Rochester, 55905 MN, USA
| |
Collapse
|
49
|
Madeddu P. Cell therapy for the treatment of heart disease: Renovation work on the broken heart is still in progress. Free Radic Biol Med 2021; 164:206-222. [PMID: 33421587 DOI: 10.1016/j.freeradbiomed.2020.12.444] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/26/2020] [Accepted: 12/29/2020] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease (CVD) continues to be the number one killer in the aging population. Heart failure (HF) is also an important cause of morbidity and mortality in patients with congenital heart disease (CHD). Novel therapeutic approaches that could restore stable heart function are much needed in both paediatric and adult patients. Regenerative medicine holds promises to provide definitive solutions for correction of congenital and acquired cardiac defects. In this review article, we recap some important aspects of cardiovascular cell therapy. First, we report quantifiable data regarding the scientific advancements in the field and how this has been translated into tangible outcomes according clinical studies and related meta-analyses. We then comment on emerging trends and technologies, such as the use of second-generation cell products, including pericyte-like vascular progenitors, and reprogramming of cells by different approaches including modulation of oxidative stress. The more affordable and feasible strategy of repurposing clinically available drugs to awaken the intrinsic healing potential of the heart will be discussed in the light of current social, financial, and ethical context. Cell therapy remains a work in progress field. Uncertainty in the ability of the experts and policy makers to solve urgent medical problems is growing in a world that is significantly influenced by them. This is particularly true in the field of regenerative medicine, due to great public expectations, polarization of leadership and funding, and insufficient translational vision. Cardiovascular regenerative medicine should be contextualized in a holistic program with defined priorities to allow a complete realization. Reshaping the notion of medical expertise is fundamental to fill the current gap in translation.
Collapse
Affiliation(s)
- Paolo Madeddu
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol Royal Infirmary, Upper Maudlin Street, BS28HW, Bristol, United Kingdom.
| |
Collapse
|
50
|
Grigorian-Shamagian L, Sanz-Ruiz R, Climent A, Badimon L, Barile L, Bolli R, Chamuleau S, Grobbee DE, Janssens S, Kastrup J, Kragten-Tabatabaie L, Madonna R, Mathur A, Menasché P, Pompilio G, Prosper F, Sena E, Smart N, Zimmermann WH, Fernández-Avilés F. Insights into therapeutic products, preclinical research models, and clinical trials in cardiac regenerative and reparative medicine: where are we now and the way ahead. Current opinion paper of the ESC Working Group on Cardiovascular Regenerative and Reparative Medicine. Cardiovasc Res 2020; 117:1428-1433. [PMID: 33258961 DOI: 10.1093/cvr/cvaa337] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 10/08/2020] [Accepted: 11/17/2020] [Indexed: 01/04/2023] Open
Abstract
Great expectations have been set around the clinical potential of regenerative and reparative medicine in the treatment of cardiovascular diseases [i.e. in particular, heart failure (HF)]. Initial excitement, spurred by encouraging preclinical data, resulted in a rapid translation into clinical research. The sobering outcome of the resulting clinical trials suggests that preclinical testing may have been insufficient to predict clinical outcome. A number of barriers for clinical translation include the inherent variability of the biological products and difficulties to develop potency and quality assays, insufficient rigour of the preclinical research and reproducibility of the results, manufacturing challenges, and scientific irregularities reported in the last years. The failure to achieve clinical success led to an increased scrutiny and scepticism as to the clinical readiness of stem cells and gene therapy products among clinicians, industry stakeholders, and funding bodies. The present impasse has attracted the attention of some of the most active research groups in the field, which were then summoned to analyse the position of the field and tasked to develop a strategy, to re-visit the undoubtedly promising future of cardiovascular regenerative and reparative medicine, based on lessons learned over the past two decades. During the scientific retreat of the ESC Working Group on Cardiovascular Regenerative and Reparative Medicine (CARE) in November 2018, the most relevant and timely research aspects in regenerative and/or reparative medicine were presented and critically discussed, with the aim to lay out a strategy for the future development of the field. We report herein the main ideas and conclusions of that meeting.
Collapse
Affiliation(s)
- Lilian Grigorian-Shamagian
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Doctor Esquerdo 46, 28007 Madrid, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Ricardo Sanz-Ruiz
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Doctor Esquerdo 46, 28007 Madrid, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Andreu Climent
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Doctor Esquerdo 46, 28007 Madrid, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Lina Badimon
- CIBERCV, Instituto de Salud Carlos III, Madrid, Spain.,Cardiovascular Program-ICCC, IR-Hospital de la Santa Creu i Sant Pau, and Cardiovascular Research Chair, Autonomous University of Barcelona, Spain
| | - Lucio Barile
- Laboratory for Cardiovascular Theranostics, Cardiocentro Ticino Foundation and Faculty of Biomedical Sciences Università Svizzera Italiana, Lugano, Switzerland
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, 550 S. Jackson St., ACB, 3rd Floor, Louisville, KY 40292, USA
| | - Steven Chamuleau
- Department of Cardiology, Amsterdam UMC, Location AMC
- B2-239
- Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Diederick E Grobbee
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands
| | - Stefan Janssens
- Department of Cardiovascular Medicine, UZ Leuven Campus Gasthuisberg, Herestraat 49, B-3000 Leuven, Belgium
| | - Jens Kastrup
- Department of Cardiology, The heart Centre, Rigshospitalet University of Copenhagen, Denmark
| | | | | | - Anthony Mathur
- Centre for Cardiovascular Medicine and Device Innovation, Queen Mary University of London, Barts Heart Centre, St Bartholomew's Hospital, UK
| | - Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou 20, Université de Paris, PARCC, INSERM, rue Leblanc 75015 Paris, F-75015, Paris, France
| | - Giulio Pompilio
- Pompilio G Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, Italy.,Department of Clinical Sciences and Community Health, University of Milano, Italy
| | - Felipe Prosper
- Hematology and Cell Therapy, Clinica Universidad de Navarra, Pamplona and Tercel, Instituto de Salud Carlos III, Madrid, Spain
| | - Emily Sena
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Nicola Smart
- Department of Physiology, Anatomy & Genetics, University of Oxford, UK
| | - Wolfgram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center, Georg-August University, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Potsdamer Str. 58 10785 Berlin, Germany
| | - Francisco Fernández-Avilés
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Doctor Esquerdo 46, 28007 Madrid, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|