1
|
Gong X, Jiao Y, Hu H, Zhang R, Jia W, Zhao J, Liu Z, Xin Y, Han W. A prospective randomized controlled study of multi-intravenous infusion of umbilical cord mesenchymal stem cells in patients with heart failure and reduced ejection fraction (PRIME-HFrEF) trial: Rationale and design. Contemp Clin Trials Commun 2024; 41:101350. [PMID: 39246626 PMCID: PMC11377133 DOI: 10.1016/j.conctc.2024.101350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/12/2024] [Accepted: 08/11/2024] [Indexed: 09/10/2024] Open
Abstract
Background and objective The use of mesenchymal stem cells for heart failure treatment has gained increasing interest. However, most studies have relied on a single injection approach, with no research yet confirming the effects of multiple administrations. The present trial aims to investigate the safety and efficacy of multi-intravenous infusion of umbilical cord-mesenchymal stem cells (UC-MSCs) in patients with heart failure and reduced ejection fraction (HFrEF). Methods The PRIME-HFrEF trial is a single-center, prospective, randomized, triple-blinded, placebo-controlled trial of multi-intravenous infusion of UC-MSCs in HFrEF patients. A total of 40 patients meeting the inclusion criteria for HFrEF were enrolled and randomized 1:1 to the MSC group or the placebo group. Patients enrolled will receive intravenous injections of either UC-MSCs or placebo every 6 weeks for three times. Both groups will be followed up for 12 months. The primary safety endpoint is the incidence of serious adverse events. The primary efficacy endpoint is a change in left ventricular ejection fraction (LVEF) measured by left ventricular opacification (LVO) with contrast echocardiography and magnetic resonance imaging (MRI) at 12 months. The secondary endpoints include a composite of the incidence of death and re-hospitalization caused by heart failure at the 12th month, serum NT-proBNP, growth stimulation expressed gene 2 (ST2), and a change of right ventricular structure and function. Conclusions The PRIME-HFrEF study is designed to shed new light on multiple UC-MSC administration regimens for heart failure treatment.
Collapse
Affiliation(s)
- Xin Gong
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yuheng Jiao
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Hao Hu
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Rongzhen Zhang
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wenwen Jia
- Institute for Regenerative Medicine, National Stem Cell Translational Resource Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, China
| | - Jun Zhao
- Department of Nuclear Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhongmin Liu
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| | - Yuanfeng Xin
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wei Han
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| |
Collapse
|
2
|
Niroomand A, Nita GE, Lindstedt S. Machine Perfusion and Bioengineering Strategies in Transplantation-Beyond the Emerging Concepts. Transpl Int 2024; 37:13215. [PMID: 39267617 PMCID: PMC11390383 DOI: 10.3389/ti.2024.13215] [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: 05/01/2024] [Accepted: 08/19/2024] [Indexed: 09/15/2024]
Abstract
Solid organ transplantation has progressed rapidly over the decades from the first experimental procedures to its role in the modern era as an established treatment for end-stage organ disease. Solid organ transplantation including liver, kidney, pancreas, heart, and lung transplantation, is the definitive option for many patients, but despite the advances that have been made, there are still significant challenges in meeting the demand for viable donor grafts. Furthermore, post-operatively, the recipient faces several hurdles, including poor early outcomes like primary graft dysfunction and acute and chronic forms of graft rejection. In an effort to address these issues, innovations in organ engineering and treatment have been developed. This review covers efforts made to expand the donor pool including bioengineering techniques and the use of ex vivo graft perfusion. It also covers modifications and treatments that have been trialed, in addition to research efforts in both abdominal organs and thoracic organs. Overall, this article discusses recent innovations in machine perfusion and organ bioengineering with the aim of improving and increasing the quality of donor organs.
Collapse
Affiliation(s)
- Anna Niroomand
- Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden
| | - George Emilian Nita
- Department of Transplantation Surgery, Liverpool University Hospitals NHS Foundation Trust, Liverpool, United Kingdom
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
- Division of Transplantation Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Sandra Lindstedt
- Department of Clinical Sciences, Lund University, Lund, Sweden
- Lund Stem Cell Center, Lund University, Lund, Sweden
- Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
3
|
Safwan M, Bourgleh MS, Aldoush M, Haider KH. Tissue-source effect on mesenchymal stem cells as living biodrugs for heart failure: Systematic review and meta-analysis. World J Cardiol 2024; 16:469-483. [PMID: 39221190 PMCID: PMC11362808 DOI: 10.4330/wjc.v16.i8.469] [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: 05/06/2024] [Revised: 06/24/2024] [Accepted: 07/23/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs), as living biodrugs, have entered advanced phases of clinical assessment for cardiac function restoration in patients with myocardial infarction and heart failure. While MSCs are available from diverse tissue sources, bone-marrow-derived MSCs (BM-MSCs) remain the most well-studied cell type, besides umbilical-cord-derived MSCs (UC-MSCs). The latter offers advantages, including noninvasive availability without ethical considerations.
AIM To compare the safety and efficacy of BM-MSCs and UC-MSCs in terms of left ventricular ejection fraction (LVEF), 6-min walking distance (6MWD), and major adverse cardiac events (MACEs).
METHODS Five databases were systematically searched to identify randomized controlled trials (RCTs). Thirteen RCTs (693 patients) were included using predefined eligibility criteria. Weighted mean differences and odds ratio (OR) for the changes in the estimated treatment effects.
RESULTS UC-MSCs significantly improved LVEF vs controls by 5.08% [95% confidence interval (CI): 2.20%-7.95%] at 6 mo and 2.78% (95%CI: 0.86%-4.70%) at 12 mo. However, no significant effect was observed for BM-MSCs vs controls. No significant changes were observed in the 6MWD with either of the two cell types. Also, no differences were observed for MACEs, except rehospitalization rates, which were lower only with BM-MSCs (odds ratio 0.48, 95%CI: 0.24-0.97) vs controls.
CONCLUSION UC-MSCs significantly improved LVEF compared with BM-MSCs. Their advantageous characteristics position them as a promising alternative to MSC-based therapy.
Collapse
Affiliation(s)
- Moaz Safwan
- Department of Basic Sciences, Sulaiman Al Rajhi University, Al Bukairiyah 51941, Saudi Arabia
| | - Mariam Safwan Bourgleh
- Department of Basic Sciences, Sulaiman Al Rajhi University, Al Bukairiyah 51941, Saudi Arabia
| | - Mohamed Aldoush
- Department of Basic Sciences, Sulaiman Al Rajhi University, Al Bukairiyah 51941, Saudi Arabia
| | - Khawaja Husnain Haider
- Department of Basic Sciences, Sulaiman Al Rajhi University, Al Bukairiyah 51941, Saudi Arabia
| |
Collapse
|
4
|
Kavousi S, Hosseinpour A, Bahmanzadegan Jahromi F, Attar A. Efficacy of mesenchymal stem cell transplantation on major adverse cardiovascular events and cardiac function indices in patients with chronic heart failure: a meta-analysis of randomized controlled trials. J Transl Med 2024; 22:786. [PMID: 39174960 PMCID: PMC11342608 DOI: 10.1186/s12967-024-05352-y] [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: 02/11/2024] [Accepted: 05/29/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND The effects of mesenchymal stem cells (MSCs) on heart failure (HF) have been controversial. This study was conducted to investigate whether the transplantation of MSCs after HF could help improve clinical outcomes and myocardial performance indices. METHODS Using a systematic approach, electronic databases were searched for randomized controlled trials (RCTs), which evaluated the transplantation of MSCs after HF. The outcomes owf interest included clinical outcomes and myocardial function indices. We also assessed the role of age, cause of heart failure, cell origin, cell number, type of donor (autologous/allogeneic), and route of cell delivery on these outcomes. Using the random-effects method, a relative risk (RR) or mean difference (MD) and their corresponding 95% confidence intervals (CI) were pooled. RESULTS Seventeen RCTs including 1684 patients (927 and 757 patients in the intervention and control arms, respectively) were enrolled. The RR (95% CI) of mortality was 0.78 (0.62; 0.99, p = 0.04) in the MSC group compared to the controls. HF rehospitalization decreased in the MSC group (RR = 0.85 (0.71-1.01), p = 0.06), but this was only significant in those who received autologous MSCs (RR = 0.67 (0.49; 0.90), p = 0.008). LVEF was significantly increased among those who received MSC (MD = 3.38 (1.89; 4.87), p < 0.001). LVESV (MD = -9.14 (-13.25; -5.03), p < 0.001), LVEDV (MD = -8.34 -13.41; -3.27), p < 0.001), and scar size (standardized MD = -0.32 (-0.60; -0.05), p = 0.02) were significantly decreased. NYHA class (MD = -0.19 (-0.34; -0.06), p = 0.006), BNP level (standardized MD = -0.28 (-0.50; -0.06), p = 0.01), and MLHFQ (MD = -11.55 (-16.77; -6.33), p = 0.005) significantly decreased and 6-min walk test significantly improved (MD = 36.86 (11.22; 62.50), p = 0.001) in the MSC group. Trials were not affected by the participants' etiology of heart failure, while trials with the autologous source of cells, MSC doses lower than 100 million cells, and intracoronary injection performed significantly better in some of the outcomes. CONCLUSION Transplantation of MSCs for ischemic or dilated heart failure patients may reduce all-cause mortality and improve clinical condition. Moreover, this treatment would improve left ventricular function indices and reduce scar size.
Collapse
Affiliation(s)
- Shahin Kavousi
- Shiraz University of Medical Sciences and Health Services, Shiraz, Iran
| | | | | | - Armin Attar
- Department of Cardiovascular Medicine, TAHA Clinical Trial Group, School of Medicine, Shiraz University of Medical Sciences, Zand Street, Shiraz, Iran.
| |
Collapse
|
5
|
Yahyazadeh R, Baradaran Rahimi V, Askari VR. Stem cell and exosome therapies for regenerating damaged myocardium in heart failure. Life Sci 2024; 351:122858. [PMID: 38909681 DOI: 10.1016/j.lfs.2024.122858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/25/2024]
Abstract
Finding novel treatments for cardiovascular diseases (CVDs) is a hot topic in medicine; cell-based therapies have reported promising news for controlling dangerous complications of heart disease such as myocardial infarction (MI) and heart failure (HF). Various progenitor/stem cells were tested in various in-vivo, in-vitro, and clinical studies for regeneration or repairing the injured tissue in the myocardial to accelerate the healing. Fetal, adult, embryonic, and induced pluripotent stem cells (iPSC) have revealed the proper potency for cardiac tissue repair. As an essential communicator among cells, exosomes with specific contacts (proteins, lncRNAs, and miRNAs) greatly promote cardiac rehabilitation. Interestingly, stem cell-derived exosomes have more efficiency than stem cell transplantation. Therefore, stem cells induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), cardiac stem cells (CDC), and skeletal myoblasts) and their-derived exosomes will probably be considered an alternative therapy for CVDs remedy. In addition, stem cell-derived exosomes have been used in the diagnosis/prognosis of heart diseases. In this review, we explained the advances of stem cells/exosome-based treatment, their beneficial effects, and underlying mechanisms, which will present new insights in the clinical field in the future.
Collapse
Affiliation(s)
- Roghayeh Yahyazadeh
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vafa Baradaran Rahimi
- Department of Cardiovascular Diseases, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Vahid Reza Askari
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Hosseinpour A, Kamalpour J, Dehdari Ebrahimi N, Mirhosseini SA, Sadeghi A, Kavousi S, Attar A. Comparative effectiveness of mesenchymal stem cell versus bone-marrow mononuclear cell transplantation in heart failure: a meta-analysis of randomized controlled trials. Stem Cell Res Ther 2024; 15:202. [PMID: 38971816 PMCID: PMC11227704 DOI: 10.1186/s13287-024-03829-7] [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: 06/04/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024] Open
Abstract
BACKGROUND There is no clear evidence on the comparative effectiveness of bone-marrow mononuclear cell (BMMNC) vs. mesenchymal stromal cell (MSC) stem cell therapy in patients with chronic heart failure (HF). METHODS Using a systematic approach, eligible randomized controlled trials (RCTs) of stem cell therapy (BMMNCs or MSCs) in patients with HF were retrieved to perform a meta-analysis on clinical outcomes (major adverse cardiovascular events (MACE), hospitalization for HF, and mortality) and echocardiographic indices (including left ventricular ejection fraction (LVEF)) were performed using the random-effects model. A risk ratio (RR) or mean difference (MD) with corresponding 95% confidence interval (CI) were pooled based on the type of the outcome and subgroup analysis was performed to evaluate the potential differences between the types of cells. RESULTS The analysis included a total of 36 RCTs (1549 HF patients receiving stem cells and 1252 patients in the control group). Transplantation of both types of cells in patients with HF resulted in a significant improvement in LVEF (BMMNCs: MD (95% CI) = 3.05 (1.11; 4.99) and MSCs: MD (95% CI) = 2.82 (1.19; 4.45), between-subgroup p = 0.86). Stem cell therapy did not lead to a significant change in the risk of MACE (MD (95% CI) = 0.83 (0.67; 1.06), BMMNCs: RR (95% CI) = 0.59 (0.31; 1.13) and MSCs: RR (95% CI) = 0.91 (0.70; 1.19), between-subgroup p = 0.12). There was a marginally decreased risk of all-cause death (MD (95% CI) = 0.82 (0.68; 0.99)) and rehospitalization (MD (95% CI) = 0.77 (0.61; 0.98)) with no difference among the cell types (p > 0.05). CONCLUSION Both types of stem cells are effective in improving LVEF in patients with heart failure without any noticeable difference between the cells. Transplantation of the stem cells could not decrease the risk of major adverse cardiovascular events compared with controls. Future trials should primarily focus on the impact of stem cell transplantation on clinical outcomes of HF patients to verify or refute the findings of this study.
Collapse
Affiliation(s)
- Alireza Hosseinpour
- Department of Cardiovascular Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jahangir Kamalpour
- Department of Cardiovascular Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Alireza Sadeghi
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahin Kavousi
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Armin Attar
- Department of Cardiovascular Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
7
|
Nativi-Nicolau J, Yilmaz A, Dasgupta N, Macey R, Cochrane J, Peatman J, Summers C, Luth J, Zolty R. Six-minute walk test as clinical end point in cardiomyopathy clinical trials, including ATTR-CM: a systematic literature review. J Comp Eff Res 2024; 13:e230158. [PMID: 38869839 PMCID: PMC11234454 DOI: 10.57264/cer-2023-0158] [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: 10/19/2023] [Accepted: 05/20/2024] [Indexed: 06/14/2024] Open
Abstract
Aim: The six-minute walk test (6MWT) is a common measure of functional capacity in patients with heart failure (HF). Primary clinical study end points in cardiomyopathy (CM) trials, including transthyretin-mediated amyloidosis with CM (ATTR-CM), are often limited to hospitalization and mortality. Objective: To investigate the relationship between the 6MWT and hospitalization or mortality in CM, including ATTR-CM. Method: A PRISMA-guided systematic literature review was conducted using search terms for CM, 6MWT, hospitalization and mortality. Results: Forty-one studies were identified that reported 6MWT data and hospitalization or mortality data for patients with CM. The data suggest that a greater 6MWT distance is associated with a reduced risk of hospitalization or mortality in CM. Conclusion: The 6MWT is an accepted alternative end point in CM trials, including ATTR-CM.
Collapse
Affiliation(s)
| | - Ali Yilmaz
- Division of Cardiovascular Imaging, University Hospital Münster, 48149, Münster, Germany
| | - Noel Dasgupta
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Richard Macey
- Adelphi Values PROVE, Bollington, Cheshire, UK, SK10 5JB
| | - James Cochrane
- Adelphi Values PROVE, Bollington, Cheshire, UK, SK10 5JB
| | - Judith Peatman
- Adelphi Values PROVE, Bollington, Cheshire, UK, SK10 5JB
| | - Catherine Summers
- Medical Affairs Department, Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | - Jennifer Luth
- Medical Affairs Department, Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | - Ronald Zolty
- Division of Cardiovascular Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE 68198 USA
| |
Collapse
|
8
|
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
|
9
|
Jihwaprani MC, Sula I, Charbat MA, Haider KH. Establishing delivery route-dependent safety and efficacy of living biodrug mesenchymal stem cells in heart failure patients. World J Cardiol 2024; 16:339-354. [PMID: 38993584 PMCID: PMC11235206 DOI: 10.4330/wjc.v16.i6.339] [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: 02/22/2024] [Revised: 05/21/2024] [Accepted: 05/24/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) as living biopharmaceuticals with unique properties, i.e., stemness, viability, phenotypes, paracrine activity, etc., need to be administered such that they reach the target site, maintaining these properties unchanged and are retained at the injury site to participate in the repair process. Route of delivery (RoD) remains one of the critical determinants of safety and efficacy. This study elucidates the safety and effectiveness of different RoDs of MSC treatment in heart failure (HF) based on phase II randomized clinical trials (RCTs). We hypothesize that the RoD modulates the safety and efficacy of MSC-based therapy and determines the outcome of the intervention. AIM To investigate the effect of RoD of MSCs on safety and efficacy in HF patients. METHODS RCTs were retrieved from six databases. Safety endpoints included mortality and serious adverse events (SAEs), while efficacy outcomes encompassed changes in left ventricular ejection fraction (LVEF), 6-minute walk distance (6MWD), and pro-B-type natriuretic peptide (pro-BNP). Subgroup analyses on RoD were performed for all study endpoints. RESULTS Twelve RCTs were included. Overall, MSC therapy demonstrated a significant decrease in mortality [relative risk (RR): 0.55, 95% confidence interval (95%CI): 0.33-0.92, P = 0.02] compared to control, while SAE outcomes showed no significant difference (RR: 0.84, 95%CI: 0.66-1.05, P = 0.11). RoD subgroup analysis revealed a significant difference in SAE among the transendocardial (TESI) injection subgroup (RR = 0.71, 95%CI: 0.54-0.95, P = 0.04). The pooled weighted mean difference (WMD) demonstrated an overall significant improvement of LVEF by 2.44% (WMD: 2.44%, 95%CI: 0.80-4.29, P value ≤ 0.001), with only intracoronary (IC) subgroup showing significant improvement (WMD: 7.26%, 95%CI: 5.61-8.92, P ≤ 0.001). Furthermore, the IC delivery route significantly improved 6MWD by 115 m (WMD = 114.99 m, 95%CI: 91.48-138.50), respectively. In biochemical efficacy outcomes, only the IC subgroup showed a significant reduction in pro-BNP by -860.64 pg/mL (WMD: -860.64 pg/Ml, 95%CI: -944.02 to -777.26, P = 0.001). CONCLUSION Our study concluded that all delivery methods of MSC-based therapy are safe. Despite the overall benefits in efficacy, the TESI and IC routes provided better outcomes than other methods. Larger-scale trials are warranted before implementing MSC-based therapy in routine clinical practice.
Collapse
Affiliation(s)
| | - Idris Sula
- Basic Sciences, SRC, Al Bukayriyah 52736, AlQaseem, Saudi Arabia
| | | | | |
Collapse
|
10
|
Ahmed ZT, Zain Al-Abeden MS, Al Abdin MG, Muqresh MA, Al Jowf GI, Eijssen LMT, Haider KH. Dose-response relationship of MSCs as living Bio-drugs in HFrEF patients: a systematic review and meta-analysis of RCTs. Stem Cell Res Ther 2024; 15:165. [PMID: 38867306 PMCID: PMC11170815 DOI: 10.1186/s13287-024-03713-4] [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: 02/28/2024] [Accepted: 04/01/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have emerged as living biodrugs for myocardial repair and regeneration. Recent randomized controlled trials (RCTs) have reported that MSC-based therapy is safe and effective in heart failure patients; however, its dose-response relationship has yet to be established. We aimed to determine the optimal MSC dose for treating HF patients with reduced ejection fraction (EF) (HFrEF). METHODS The preferred reporting items for systematic reviews and meta-analyses (PRISMA) and Cochrane Handbook guidelines were followed. Four databases and registries, i.e., PubMed, EBSCO, clinicaltrials.gov, ICTRP, and other websites, were searched for RCTs. Eleven RCTs with 1098 participants (treatment group, n = 606; control group, n = 492) were selected based on our inclusion/exclusion criteria. Two independent assessors extracted the data and performed quality assessments. The data from all eligible studies were plotted for death, major adverse cardiac events (MACE), left ventricular ejection fraction (LVEF), left ventricular end-systolic volume (LVESV), and 6-minute walk distance (6-MWD) as safety, efficacy, and performance parameters. For dose-escalation assessment, studies were categorized as low-dose (< 100 million cells) or high-dose (≥ 100 million cells). RESULTS MSC-based treatment is safe across low and high doses, with nonsignificant effects. However, low-dose treatment had a more significant protective effect than high-dose treatment. Subgroup analysis revealed the superiority of low-dose treatment in improving LVEF by 3.01% (95% CI; 0.65-5.38%) compared with high-dose treatment (-0.48%; 95% CI; -2.14-1.18). MSC treatment significantly improved the 6-MWD by 26.74 m (95% CI; 3.74-49.74 m) in the low-dose treatment group and by 36.73 m (95% CI; 6.74-66.72 m) in the high-dose treatment group. The exclusion of studies using ADRCs resulted in better safety and a significant improvement in LVEF from low- and high-dose MSC treatment. CONCLUSION Low-dose MSC treatment was safe and superior to high-dose treatment in restoring efficacy and functional outcomes in heart failure patients, and further analysis in a larger patient group is warranted.
Collapse
Affiliation(s)
- Ziyad T Ahmed
- College of Medicine, Sulaiman Al Rajhi University, Al-Bukairiyah, 52726, Saudi Arabia
| | | | | | - Mohamad Ayham Muqresh
- College of Medicine, Sulaiman Al Rajhi University, Al-Bukairiyah, 52726, Saudi Arabia
| | - Ghazi I Al Jowf
- Department of Public Health, College of Applied Medical Sciences, King Faisal University, Al-Ahsa, 31982, Saudi Arabia
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Centre, Maastricht, 6200 MD, The Netherlands
- European Graduate School of Neuroscience, Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - Lars M T Eijssen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Centre, Maastricht, 6200 MD, The Netherlands
- Department of Bioinformatics- BiGCaT, School of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, 6200 MD, The Netherlands
- European Graduate School of Neuroscience, Maastricht University, Maastricht, 6200 MD, The Netherlands
| | | |
Collapse
|
11
|
Paresishvili T, Kakabadze Z. Freeze-Dried Mesenchymal Stem Cells: From Bench to Bedside. Review. Adv Biol (Weinh) 2024; 8:e2300155. [PMID: 37990389 DOI: 10.1002/adbi.202300155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/09/2023] [Indexed: 11/23/2023]
Abstract
This review describes the freeze-dried mesenchymal stem cells (MSCs) and their ability to restore damaged tissues and organs. An analysis of the literature shows that after the lyophilization MSCs retain >80% of paracrine factors and that the mechanism of their action on the restoration of damaged tissues and organs is similar to the mechanism of action of paracrine factors in fresh and cryopreserved mesenchymal stem cells. Based on the own materials, the use of paracrine factors of freeze-dried MSCs in vivo and in vitro for the treatment of various diseases of organs and tissues has shown to be effective. The study also discusses about the advantages and disadvantages of freeze-dried MSCs versus cryopreserved MSCs. However, for the effective use of freeze-dried MSCs in clinical practice, a more detailed study of the mechanism of interaction of paracrine factors of freeze-dried MSCs with target cells and tissues is required. It is also necessary to identify possible other specific paracrine factors of freeze-dried MSCs. In addition, develop new therapeutic strategies for the use of freeze-dried MSCs in regenerative medicine and tissue bioengineering.
Collapse
Affiliation(s)
- Teona Paresishvili
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, 0186, Georgia
| | - Zurab Kakabadze
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, 0186, Georgia
| |
Collapse
|
12
|
Chowdhury MA, Zhang JJ, Rizk R, Chen WCW. Stem cell therapy for heart failure in the clinics: new perspectives in the era of precision medicine and artificial intelligence. Front Physiol 2024; 14:1344885. [PMID: 38264333 PMCID: PMC10803627 DOI: 10.3389/fphys.2023.1344885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 12/26/2023] [Indexed: 01/25/2024] Open
Abstract
Stem/progenitor cells have been widely evaluated as a promising therapeutic option for heart failure (HF). Numerous clinical trials with stem/progenitor cell-based therapy (SCT) for HF have demonstrated encouraging results, but not without limitations or discrepancies. Recent technological advancements in multiomics, bioinformatics, precision medicine, artificial intelligence (AI), and machine learning (ML) provide new approaches and insights for stem cell research and therapeutic development. Integration of these new technologies into stem/progenitor cell therapy for HF may help address: 1) the technical challenges to obtain reliable and high-quality therapeutic precursor cells, 2) the discrepancies between preclinical and clinical studies, and 3) the personalized selection of optimal therapeutic cell types/populations for individual patients in the context of precision medicine. This review summarizes the current status of SCT for HF in clinics and provides new perspectives on the development of computation-aided SCT in the era of precision medicine and AI/ML.
Collapse
Affiliation(s)
- Mohammed A. Chowdhury
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
- Department of Public Health and Health Sciences, Health Sciences Ph.D. Program, School of Health Sciences, University of South Dakota, Vermillion, SD, United States
- Department of Cardiology, North Central Heart, Avera Heart Hospital, Sioux Falls, SD, United States
| | - Jing J. Zhang
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Rodrigue Rizk
- Department of Computer Science, University of South Dakota, Vermillion, SD, United States
| | - William C. W. Chen
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| |
Collapse
|
13
|
Krishna Mohan GV, Tirumandyam G, Vemulapalli HS, Vajje J, Asif H, Saleem F. Mesenchymal Stem Cell Therapy for a Better Prognosis of Heart Failure: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Cureus 2023; 15:e43037. [PMID: 37674948 PMCID: PMC10479956 DOI: 10.7759/cureus.43037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2023] [Indexed: 09/08/2023] Open
Abstract
Mesenchymal stem cell (MSC) therapy is a frequently used treatment option for achieving a better prognosis in patients with heart failure (HF). However, due to reported adverse effects, patients are often hesitant to consider this treatment. Consequently, the aim of this systemic review and meta-analysis is to further investigate the effects of MSCs on survival outcomes, hospital readmissions, and left ventricular ejection fraction (LVEF) in individuals with pre-existing HF. We systematically searched PubMed, Web of Science, Embase, and Cochrane Library to review studies published up until July 16, 2023. Risk ratios were generated using the extracted data for all the outcomes except LVEF. The mean difference was generated for LVEF. Sensitivity analysis was performed to investigate heterogeneity, and the risk of bias tool was used to assess the quality of the included studies. Fourteen randomized controlled trials were included in the meta-analysis. Pooled results revealed that the MSC therapy group did not significantly affect the outcomes of cardiovascular death, rehospitalization rate, myocardial infarction, recurrence of HF, and total death when compared to a control group. However, MSC therapy was significantly associated with an increased LVEF (RR = 3.35; 95% CI: 0.79-5.72; p = 0.010; I2 = 95%). Upon sensitivity analysis, MSC therapy was significantly associated with a decreased hospitalization rate (RR = 0.46; 95% CI: 0.34-0.64; p < 0.00001; I2 = 0%). MSC transplantation results in a significantly improved LVEF and rehospitalization rate.
Collapse
Affiliation(s)
| | - Gayathri Tirumandyam
- Internal Medicine, Siddhartha Medical College, Dr. YSR University of Health Sciences, Vijayawada, IND
| | | | - Jaahnavi Vajje
- Internal Medicine, Dr. Pinnamaneni Siddhartha Institute of Medical Sciences & Research Foundation, Vijayawada, IND
| | - Hamza Asif
- Pulmonology, Khyber Teaching Hospital, Peshawar, PAK
| | - Faraz Saleem
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
- Internal Medicine, Akhtar Saeed Medical and Dental College, Lahore, PAK
| |
Collapse
|
14
|
Kalou Y, Al-Khani AM, Haider KH. Bone Marrow Mesenchymal Stem Cells for Heart Failure Treatment: A Systematic Review and Meta-Analysis. Heart Lung Circ 2023; 32:870-880. [PMID: 36872163 DOI: 10.1016/j.hlc.2023.01.012] [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: 06/22/2022] [Revised: 12/22/2022] [Accepted: 01/02/2023] [Indexed: 03/06/2023]
Abstract
AIM Bone marrow-derived mesenchymal stem cells (BM-MSCs) are the most well-studied and characterised stem cell types. This review was undertaken of the current available phase II/III randomised clinical trials (RCTs) that delivered BM-MSCs to treat patients with cardiomyopathy, and to assess their performance. METHODS The Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines were followed during the systematic review and meta-analysis. Eligible studies were reviewed, and their data charted. To assess the efficacy of BM-MSCs, the outcome was improvement in left ventricular ejection fraction (LVEF) and 6-minute walking distance (6MWD). RESULTS The pooled weighted mean difference (WMD) showed that BM-MSCs treatment improved the 6MWD by 27.86 m (95% CI 0.11-55.6 m) compared with the control groups. The pooled WMD showed that BM-MSCs treatment improved the LVEF by 6.37% (95% CI 5.48%-7.26%) compared with the control groups. CONCLUSION BM-MSCs treatment is an effective intervention for managing patients with heart failure, but it requires larger and more robust clinical trials to support its routine use in clinics.
Collapse
Affiliation(s)
- Yazan Kalou
- College of Medicine, Sulaiman Al Rajhi University, Bukairyah, Al-Qassim, Saudi Arabia. https://twitter.com/yazka16
| | - Abdullah Murhaf Al-Khani
- College of Medicine, Sulaiman Al Rajhi University, Bukairyah, Al-Qassim, Saudi Arabia. https://twitter.com/Al_khani_95
| | | |
Collapse
|
15
|
Reid A, Hussain M, Veerapen J, Ramaseshan R, Hall R, Bowles R, Jones DA, Mathur A. DCM Support: cell therapy and circulatory support for dilated cardiomyopathy patients with severe ventricular impairment. ESC Heart Fail 2023. [PMID: 37190883 PMCID: PMC10375109 DOI: 10.1002/ehf2.14393] [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: 12/05/2022] [Revised: 04/11/2023] [Accepted: 04/24/2023] [Indexed: 05/17/2023] Open
Abstract
AIMS The DCM Support trial (NCT03572660) uses a percutaneous circulatory support device (Impella CP, Abiomed, Danvers, MA, USA) to improve the safety of an intracoronary cell infusion procedure in patients with dilated cardiomyopathy (DCM) and a severely reduced left ventricular ejection fraction (LVEF). METHODS AND RESULTS DCM Support is a single-site, single-arm Phase II trial enrolling 20 symptomatic DCM patients with an LVEF ≤ 35% despite optimal medical and device therapy. After 5 days of granulocyte colony-stimulating factor therapy and a subsequent bone marrow aspiration, patients undergo an intracoronary infusion of autologous bone-marrow-derived mononuclear cells. The Impella CP device is used to provide haemodynamic support during the infusion procedure. The trial's primary endpoint is change in LVEF from baseline at 3 months. Secondary efficacy endpoints are change in LVEF from baseline at 12 months, and change in exercise capacity, New York Heart Association class, quality of life, and N-terminal pro-B-type natriuretic peptide levels from baseline at 3 and 12 months. Safety endpoints include procedural safety and major adverse cardiac events at 3 and 12 months. CONCLUSIONS This is the first trial to assess the safety and efficacy of cytokine and autologous intracoronary cell therapy with a procedural circulatory support device for patients with severe left ventricular impairment. This novel combination may allow us to target a patient population most at need of this therapy.
Collapse
Affiliation(s)
- Alice Reid
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- NIHR Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, London, EC1M 6BQ, United Kingdom
| | - Mohsin Hussain
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Jessry Veerapen
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Rohini Ramaseshan
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Russell Hall
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Ruth Bowles
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- NIHR Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, London, EC1M 6BQ, United Kingdom
| | - Daniel A Jones
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Anthony Mathur
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- NIHR Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, London, EC1M 6BQ, United Kingdom
| |
Collapse
|
16
|
Petrosyan A, Martins PN, Solez K, Uygun BE, Gorantla VS, Orlando G. Regenerative medicine applications: An overview of clinical trials. Front Bioeng Biotechnol 2022; 10:942750. [PMID: 36507264 PMCID: PMC9732032 DOI: 10.3389/fbioe.2022.942750] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 11/11/2022] [Indexed: 11/27/2022] Open
Abstract
Insights into the use of cellular therapeutics, extracellular vesicles (EVs), and tissue engineering strategies for regenerative medicine applications are continually emerging with a focus on personalized, patient-specific treatments. Multiple pre-clinical and clinical trials have demonstrated the strong potential of cellular therapies, such as stem cells, immune cells, and EVs, to modulate inflammatory immune responses and promote neoangiogenic regeneration in diseased organs, damaged grafts, and inflammatory diseases, including COVID-19. Over 5,000 registered clinical trials on ClinicalTrials.gov involve stem cell therapies across various organs such as lung, kidney, heart, and liver, among other applications. A vast majority of stem cell clinical trials have been focused on these therapies' safety and effectiveness. Advances in our understanding of stem cell heterogeneity, dosage specificity, and ex vivo manipulation of stem cell activity have shed light on the potential benefits of cellular therapies and supported expansion into clinical indications such as optimizing organ preservation before transplantation. Standardization of manufacturing protocols of tissue-engineered grafts is a critical first step towards the ultimate goal of whole organ engineering. Although various challenges and uncertainties are present in applying cellular and tissue engineering therapies, these fields' prospect remains promising for customized patient-specific treatments. Here we will review novel regenerative medicine applications involving cellular therapies, EVs, and tissue-engineered constructs currently investigated in the clinic to mitigate diseases and possible use of cellular therapeutics for solid organ transplantation. We will discuss how these strategies may help advance the therapeutic potential of regenerative and transplant medicine.
Collapse
Affiliation(s)
- Astgik Petrosyan
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Division of Urology, Children’s Hospital Los Angeles, Saban Research Institute, Los Angeles, CA, United States
| | - Paulo N. Martins
- Department of Surgery, Transplant Division, UMass Memorial Medical Center, University of Massachusetts, Worcester, MA, United States
| | - Kim Solez
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Basak E. Uygun
- Massachusetts General Hospital, Shriners Hospitals for Children in Boston and Harvard Medical School, Boston, MA, United States
| | - Vijay S. Gorantla
- Wake Forest Baptist Medical Center and Wake Forest Institute for Regenerative Medicine, Winston Salem, NC, United States
| | - Giuseppe Orlando
- Wake Forest Baptist Medical Center and Wake Forest Institute for Regenerative Medicine, Winston Salem, NC, United States
| |
Collapse
|
17
|
Margiana R, Markov A, Zekiy AO, Hamza MU, Al-Dabbagh KA, Al-Zubaidi SH, Hameed NM, Ahmad I, Sivaraman R, Kzar HH, Al-Gazally ME, Mustafa YF, Siahmansouri H. Clinical application of mesenchymal stem cell in regenerative medicine: a narrative review. Stem Cell Res Ther 2022; 13:366. [PMID: 35902958 PMCID: PMC9330677 DOI: 10.1186/s13287-022-03054-0] [Citation(s) in RCA: 115] [Impact Index Per Article: 57.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/18/2022] [Indexed: 12/16/2022] Open
Abstract
The multipotency property of mesenchymal stem cells (MSCs) has attained worldwide consideration because of their immense potential for immunomodulation and their therapeutic function in tissue regeneration. MSCs can migrate to tissue injury areas to contribute to immune modulation, secrete anti-inflammatory cytokines and hide themselves from the immune system. Certainly, various investigations have revealed anti-inflammatory, anti-aging, reconstruction, and wound healing potentials of MSCs in many in vitro and in vivo models. Moreover, current progresses in the field of MSCs biology have facilitated the progress of particular guidelines and quality control approaches, which eventually lead to clinical application of MSCs. In this literature, we provided a brief overview of immunoregulatory characteristics and immunosuppressive activities of MSCs. In addition, we discussed the enhancement, utilization, and therapeutic responses of MSCs in neural, liver, kidney, bone, heart diseases, and wound healing.
Collapse
Affiliation(s)
- Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.,Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Alexander Markov
- Tyumen State Medical University, Tyumen, Russian Federation.,Tyumen Industrial University, Tyumen, Russian Federation
| | - Angelina O Zekiy
- Department of Prosthetic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | | | | | - Noora M Hameed
- Anesthesia Techniques, Al-Nisour University College, Baghdad, Iraq
| | - Irshad Ahmad
- Department of Medical Rehabilitation Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - R Sivaraman
- Department of Mathematics, Dwaraka Doss Goverdhan Doss Vaishnav College, Arumbakkam, University of Madras, Chennai, India
| | - Hamzah H Kzar
- Veterinary Medicine College, Al-Qasim Green University, Al-Qasim, Iraq
| | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Homayoon Siahmansouri
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
18
|
da Silva JS, Gonçalves RGJ, Vasques JF, Rocha BS, Nascimento-Carlos B, Montagnoli TL, Mendez-Otero R, de Sá MPL, Zapata-Sudo G. Mesenchymal Stem Cell Therapy in Diabetic Cardiomyopathy. Cells 2022; 11:cells11020240. [PMID: 35053356 PMCID: PMC8773977 DOI: 10.3390/cells11020240] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/17/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023] Open
Abstract
The incidence and prevalence of diabetes mellitus (DM) are increasing worldwide, and the resulting cardiac complications are the leading cause of death. Among these complications is diabetes-induced cardiomyopathy (DCM), which is the consequence of a pro-inflammatory condition, oxidative stress and fibrosis caused by hyperglycemia. Cardiac remodeling will lead to an imbalance in cell survival and death, which can promote cardiac dysfunction. Since the conventional treatment of DM generally does not address the prevention of cardiac remodeling, it is important to develop new alternatives for the treatment of cardiovascular complications induced by DM. Thus, therapy with mesenchymal stem cells has been shown to be a promising approach for the prevention of DCM because of their anti-apoptotic, anti-fibrotic and anti-inflammatory effects, which could improve cardiac function in patients with DM.
Collapse
Affiliation(s)
- Jaqueline S. da Silva
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-902, RJ, Brazil; (J.S.d.S.); (B.S.R.); (B.N.-C.); (T.L.M.)
- Instituto do Coração Edson Saad, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Street Prof. Rodolpho Paulo Rocco, 255, Rio de Janeiro 21941-617, RJ, Brazil;
| | - Renata G. J. Gonçalves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-170, RJ, Brazil; (R.G.J.G.); (R.M.-O.)
| | - Juliana F. Vasques
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-170, RJ, Brazil;
| | - Bruna S. Rocha
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-902, RJ, Brazil; (J.S.d.S.); (B.S.R.); (B.N.-C.); (T.L.M.)
- Instituto do Coração Edson Saad, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Street Prof. Rodolpho Paulo Rocco, 255, Rio de Janeiro 21941-617, RJ, Brazil;
| | - Bianca Nascimento-Carlos
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-902, RJ, Brazil; (J.S.d.S.); (B.S.R.); (B.N.-C.); (T.L.M.)
| | - Tadeu L. Montagnoli
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-902, RJ, Brazil; (J.S.d.S.); (B.S.R.); (B.N.-C.); (T.L.M.)
| | - Rosália Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-170, RJ, Brazil; (R.G.J.G.); (R.M.-O.)
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-902, RJ, Brazil
| | - Mauro P. L. de Sá
- Instituto do Coração Edson Saad, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Street Prof. Rodolpho Paulo Rocco, 255, Rio de Janeiro 21941-617, RJ, Brazil;
| | - Gisele Zapata-Sudo
- Programa de Pesquisa em Desenvolvimento de Fármacos, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro 21941-902, RJ, Brazil; (J.S.d.S.); (B.S.R.); (B.N.-C.); (T.L.M.)
- Instituto do Coração Edson Saad, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Street Prof. Rodolpho Paulo Rocco, 255, Rio de Janeiro 21941-617, RJ, Brazil;
- Correspondence: or ; Tel.: +55-21-39386505
| |
Collapse
|
19
|
Xu Z, Neuber S, Nazari-Shafti T, Liu Z, Dong F, Stamm C. Impact of procedural variability and study design quality on the efficacy of cell-based therapies for heart failure - a meta-analysis. PLoS One 2022; 17:e0261462. [PMID: 34986181 PMCID: PMC8730409 DOI: 10.1371/journal.pone.0261462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 12/02/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Cell-based therapy has long been considered a promising strategy for the treatment of heart failure (HF). However, its effectiveness in the clinical setting is now doubted. Because previous meta-analyses provided conflicting results, we sought to review all available data focusing on cell type and trial design. METHODS AND FINDINGS The electronic databases PubMed, Cochrane library, ClinicalTrials.gov, and EudraCT were searched for randomized controlled trials (RCTs) utilizing cell therapy for HF patients from January 1, 2000 to December 31, 2020. Forty-three RCTs with 2855 participants were identified. The quality of the reported study design was assessed by evaluating the risk-of-bias (ROB). Primary outcomes were defined as mortality rate and left ventricular ejection fraction (LVEF) change from baseline. Secondary outcomes included both heart function data and clinical symptoms/events. Between-study heterogeneity was assessed using the I2 index. Subgroup analysis was performed based on HF type, cell source, cell origin, cell type, cell processing, type of surgical intervention, cell delivery routes, cell dose, and follow-up duration. Only 10 of the 43 studies had a low ROB for all method- and outcome parameters. A higher ROB was associated with a greater increase in LVEF. Overall, there was no impact on mortality for up to 12 months follow-up, and a clinically irrelevant average LVEF increase by LVEF (2.4%, 95% CI = 0.75-4.05, p = 0.004). Freshly isolated, primary cells tended to produce better outcomes than cultured cell products, but there was no clear impact of the cell source tissue, bone marrow cell phenotype or cell chricdose (raw or normalized for CD34+ cells). A meaningful increase in LVEF was only observed when cell therapy was combined with myocardial revascularization. CONCLUSIONS The published results suggest a small increase in LVEF following cell therapy for heart failure, but publication bias and methodologic shortcomings need to be taken into account. Given that cardiac cell therapy has now been pursued for 20 years without real progress, further efforts should not be made. STUDY REGISTRY NUMBER This meta-analysis is registered at the international prospective register of systematic reviews, number CRD42019118872.
Collapse
Affiliation(s)
- Zhiyi Xu
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Neuber
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
| | - Timo Nazari-Shafti
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Zihou Liu
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Fengquan Dong
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Christof Stamm
- Berlin Institute of Health Center for Regenerative Therapies, Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Berlin, Germany
- German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- Helmholtz Zentrum Geesthacht, Institut für Aktive Polymere, Teltow, Germany
| |
Collapse
|
20
|
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: 2.7] [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
|
21
|
Diaz-Navarro R, Urrútia G, Cleland JG, Poloni D, Villagran F, Acosta-Dighero R, Bangdiwala SI, Rada G, Madrid E. Stem cell therapy for dilated cardiomyopathy. Cochrane Database Syst Rev 2021; 7:CD013433. [PMID: 34286511 PMCID: PMC8406792 DOI: 10.1002/14651858.cd013433.pub2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Stem cell therapy (SCT) has been proposed as an alternative treatment for dilated cardiomyopathy (DCM), nonetheless its effectiveness remains debatable. OBJECTIVES To assess the effectiveness and safety of SCT in adults with non-ischaemic DCM. SEARCH METHODS We searched CENTRAL in the Cochrane Library, MEDLINE, and Embase for relevant trials in November 2020. We also searched two clinical trials registers in May 2020. SELECTION CRITERIA Eligible studies were randomized controlled trials (RCT) comparing stem/progenitor cells with no cells in adults with non-ischaemic DCM. We included co-interventions such as the administration of stem cell mobilizing agents. Studies were classified and analysed into three categories according to the comparison intervention, which consisted of no intervention/placebo, cell mobilization with cytokines, or a different mode of SCT. The first two comparisons (no cells in the control group) served to assess the efficacy of SCT while the third (different mode of SCT) served to complement the review with information about safety and other information of potential utility for a better understanding of the effects of SCT. DATA COLLECTION AND ANALYSIS Two review authors independently screened all references for eligibility, assessed trial quality, and extracted data. We undertook a quantitative evaluation of data using random-effects meta-analyses. We evaluated heterogeneity using the I² statistic. We could not explore potential effect modifiers through subgroup analyses as they were deemed uninformative due to the scarce number of trials available. We assessed the certainty of the evidence using the GRADE approach. We created summary of findings tables using GRADEpro GDT. We focused our summary of findings on all-cause mortality, safety, health-related quality of life (HRQoL), performance status, and major adverse cardiovascular events. MAIN RESULTS We included 13 RCTs involving 762 participants (452 cell therapy and 310 controls). Only one study was at low risk of bias in all domains. There were many shortcomings in the publications that did not allow a precise assessment of the risk of bias in many domains. Due to the nature of the intervention, the main source of potential bias was lack of blinding of participants (performance bias). Frequently, the format of the continuous data available was not ideal for use in the meta-analysis and forced us to seek strategies for transforming data in a usable format. We are uncertain whether SCT reduces all-cause mortality in people with DCM compared to no intervention/placebo (mean follow-up 12 months) (risk ratio (RR) 0.84, 95% confidence interval (CI) 0.54 to 1.31; I² = 0%; studies = 7, participants = 361; very low-certainty evidence). We are uncertain whether SCT increases the risk of procedural complications associated with cells injection in people with DCM (data could not be pooled; studies = 7; participants = 361; very low-certainty evidence). We are uncertain whether SCT improves HRQoL (standardized mean difference (SMD) 0.62, 95% CI 0.01 to 1.23; I² = 72%; studies = 5, participants = 272; very low-certainty evidence) and functional capacity (6-minute walk test) (mean difference (MD) 70.12 m, 95% CI -5.28 to 145.51; I² = 87%; studies = 5, participants = 230; very low-certainty evidence). SCT may result in a slight functional class (New York Heart Association) improvement (data could not be pooled; studies = 6, participants = 398; low-certainty evidence). None of the included studies reported major adverse cardiovascular events as defined in our protocol. SCT may not increase the risk of ventricular arrhythmia (data could not be pooled; studies = 8, participants = 504; low-certainty evidence). When comparing SCT to cell mobilization with granulocyte-colony stimulating factor (G-CSF), we are uncertain whether SCT reduces all-cause mortality (RR 0.46, 95% CI 0.16 to 1.31; I² = 39%; studies = 3, participants = 195; very low-certainty evidence). We are uncertain whether SCT increases the risk of procedural complications associated with cells injection (studies = 1, participants = 60; very low-certainty evidence). SCT may not improve HRQoL (MD 4.61 points, 95% CI -5.62 to 14.83; studies = 1, participants = 22; low-certainty evidence). SCT may improve functional capacity (6-minute walk test) (MD 140.14 m, 95% CI 119.51 to 160.77; I² = 0%; studies = 2, participants = 155; low-certainty evidence). None of the included studies reported MACE as defined in our protocol or ventricular arrhythmia. The most commonly reported outcomes across studies were based on physiological measures of cardiac function where there were some beneficial effects suggesting potential benefits of SCT in people with non-ischaemic DCM. However, it is unclear if this intermediate effects translates into clinical benefits for these patients. With regard to specific aspects related to the modality of cell therapy and its delivery, uncertainties remain as subgroup analyses could not be performed as planned, making it necessary to wait for the publication of several studies that are currently in progress before any firm conclusion can be reached. AUTHORS' CONCLUSIONS We are uncertain whether SCT in people with DCM reduces the risk of all-cause mortality and procedural complications, improves HRQoL, and performance status (exercise capacity). SCT may improve functional class (NYHA), compared to usual care (no cells). Similarly, when compared to G-CSF, we are also uncertain whether SCT in people with DCM reduces the risk of all-cause mortality although some studies within this comparison observed a favourable effect that should be interpreted with caution. SCT may not improve HRQoL but may improve to some extent performance status (exercise capacity). Very low-quality evidence reflects uncertainty regarding procedural complications. These suggested beneficial effects of SCT, although uncertain due to the very low certainty of the evidence, are accompanied by favourable effects on some physiological measures of cardiac function. Presently, the most effective mode of administration of SCT and the population that could benefit the most is unclear. Therefore, it seems reasonable that use of SCT in people with DCM is limited to clinical research settings. Results of ongoing studies are likely to modify these conclusions.
Collapse
Affiliation(s)
- Rienzi Diaz-Navarro
- Department of Internal Medicine, School of Medicine, Universidad de Valparaiso, Vina del Mar, Chile
| | - Gerard Urrútia
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - John Gf Cleland
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Daniel Poloni
- Department of Internal Medicine, School of Medicine, Universidad de Valparaiso, Vina del Mar, Chile
| | - Francisco Villagran
- Department of Internal Medicine, School of Medicine, Universidad de Valparaiso, Vina del Mar, Chile
| | - Roberto Acosta-Dighero
- Cochrane Chile Associate Centre, Universidad de Valparaíso, Valparaíso, Chile
- School of Physiotherapy, Faculty of Health Sciences, Universidad San Sebastian, Santiago, Chile
| | - Shrikant I Bangdiwala
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Gabriel Rada
- Department of Internal Medicine and Evidence-Based Healthcare Program, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eva Madrid
- Interdisciplinary Centre for Health Studies CIESAL, Universidad de Valparaíso, Viña del Mar, Chile
- Cochrane Chile Associate Centre, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
22
|
Mesenchymal Stem Cell Transplantation for Ischemic Diseases: Mechanisms and Challenges. Tissue Eng Regen Med 2021; 18:587-611. [PMID: 33884577 DOI: 10.1007/s13770-021-00334-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/07/2021] [Accepted: 02/16/2021] [Indexed: 12/20/2022] Open
Abstract
Ischemic diseases are conditions associated with the restriction or blockage of blood supply to specific tissues. These conditions can cause moderate to severe complications in patients, and can lead to permanent disabilities. Since they are blood vessel-related diseases, ischemic diseases are usually treated with endothelial cells or endothelial progenitor cells that can regenerate new blood vessels. However, in recent years, mesenchymal stem cells (MSCs) have shown potent bioeffects on angiogenesis, thus playing a role in blood regeneration. Indeed, MSCs can trigger angiogenesis at ischemic sites by several mechanisms related to their trans-differentiation potential. These mechanisms include inhibition of apoptosis, stimulation of angiogenesis via angiogenic growth factors, and regulation of immune responses, as well as regulation of scarring to suppress blood vessel regeneration when needed. However, preclinical and clinical trials of MSC transplantation in ischemic diseases have shown some limitations in terms of treatment efficacy. Such studies have emphasized the current challenges of MSC-based therapies. Treatment efficacy could be enhanced if the limitations were better understood and potentially resolved. This review will summarize some of the strategies by which MSCs have been utilized for ischemic disease treatment, and will highlight some challenges of those applications as well as suggesting some strategies to improve treatment efficacy.
Collapse
|
23
|
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: 49] [Impact Index Per Article: 16.3] [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
|
24
|
The efficacy of bone marrow mononuclear stem cell transplantation in patients with non-ischemic dilated cardiomyopathy-a meta analysis. Heart Fail Rev 2021; 27:811-820. [PMID: 33587248 DOI: 10.1007/s10741-021-10082-0] [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] [Accepted: 02/03/2021] [Indexed: 10/22/2022]
Abstract
Cardiomyopathy refers to a wide spectrum of heart pathologies that interfere with normal heart function. Management options of patients with cardiomyopathy depended mainly on the severity of the condition. Lifestyle modifications and regular exercise together with a healthy diet is compatible for mild conditions. Severe conditions, however, rely on medications or surgery. Here, we aim to investigate the efficacy of bone marrow mononuclear stem cell transplantation in patients with dilated cardiomyopathy. We searched PubMed, Scopus, and Cochrane CENTRAL for relevant clinical trials and excluded observational studies. We performed the quality assessment of this study following GRADE guidelines. The assessment of the risk of bias was performed by the Cochrane's risk of bias tool. We present an analysis of the following outcomes: left ventricular ejection fraction (LVEF), left ventricular end-diastolic diameter (LVEDD), and six minutes walking test. Data were pooled as mean differences (MD) and relative confidence intervals (CI). The analysis of 667 patients from 11 studies receiving autologous bone marrow cell therapy for non-ischemic dilated cardiomyopathy is presented. A total of 338 patients were allocated to the treatment group, and 329 participants entered the control group. The mean age of the patients in the treatment group is 52.4 ± 4.3 years, while that of the control is 53.7 ± 3.7 years. Seven studies (14.18-23) reported transplantation through the intracoronary route. Table 1 shows a summary of the baseline characteristics of the included studies and participants, the number of injected cells, and the type of injected cells in each trial. Table 2 summarizes and illustrates the previous treatment history of included patients in each trial, as well as the baseline values of different scores used as outcome measures in this analysis. We found that bone marrow mononuclear stem cell therapy leads to significantly increased LVEF (MD = 4.54%, 95% CI [3.52, 5.56], P < 0.0001). Patients in the transplant group experienced less left ventricular end-diastolic diameter (millimeter) than the control arm (MD = -1.86 mm, 95% CI [-4.01, 0.29], P = 0.09). Additionally, Patients in the transplant group could walk 28.53 m more than the controls (MD = 28.53 m, 95% CI [2.51, 54.55], P = 0.03). Transplantation of bone marrow stem cells yields acceptable results regarding left ventricular ejection fraction and lowers the left ventricular end-diastolic diameter. Additionally, the six minutes walking test is improved in the transplant group. Table 1 Demographic data about the included participants Study Year Sample size Age, years Males, n (%) Diabetics, n (%) Route of administration Number of injected cells Type of injected cells TTT Control TTT Control TTT Control TTT Control Bartolucci 2015 12 11 58 ± 14 57 ± 11 8 (66.7) 9 (81.8) 2 (16.7) 1 (9.1) Intracoronary 1.94 × 10^6 CD34 + Bocchi 2010 8 15 51 ± 15 NR NR NR NR Intracoronary NR NR Frljak 2018 30 30 56 ± 9 54 ± 11 27 (90) 26 (87) 3 (10) 2 (6) Trans-endocardial NR CD34 + Hamshere 2015 15 14 57.67 ± 12.32 56.79 ± 9.8 10 12 9(59.9%) 8(57.1%) Intracoronary 4.91 × 10^6 CD34 + Hu 2011 31 29 56.61 ± 9.72 58.27 ± 8.86 NR NR NR NR NR NR NR Matrino 2015 82 78 51 ± 11.1 49.6 ± 11.1 73.1 68.3 NR NR Intracoronary 10^8 TTT, CD45, CD105, and CD133 Sant'Anna 2014 20 10 48.3 ± 8.71 51.6 ± 7.79 13(65) 5 (50%) NR NR Intra-myocardial 1.06 × 108 CD3, CD4, CD14, CD34, CD38, and CD45 Seth 2010 41 40 45 ± 15 49 ± 9 33 35 NR NR NR 168 × 10^6 Bone marrow mononuclear cells Vrtovec 2011 28 27 52 ± 8 54 ± 7 26 (93) 23 (85) NR NR Intracoronary 123 × 10^6 CD34 + Vrtovec 2013 55 55 53 ± 8 55 ± 7 45 (82) 44 (80) NR NR Intracoronary NR NR Xiao 2017 16 20 49.5 ± 11.6 54.4 ± 11.6 9 (56.3) 14 (70.0) 6 (37.5) 5 (29.4) Intracoronary infusion (4.9 ± 1.7) × 108 (CD29, CD34, CD44, CD45, and CD166) Data are reported as mean ± SD or n (%) unless proved otherwise TTT treatment group, NR not reported Table 2 Previous history of treatment and drug intake by the patients Study Year Medical therapy, n (%) Baseline scores, mean (SD) Beta blockers ACE inhibitors Digoxin Diuretics LVEF, % LVEDD, mm Six minutes-walk test* TTT Control TTT Control TTT Control TTT Control TTT Control TTT Control TTT Control Bartolucci 2015 10 (83.3) 8 (72.7) NR NR 3 (25) 3 (27.3) 11 (91.6) 10 (90.9) 26.8 ± 4.9 30.3 ± 6.3 NR NR NR NR Bocchi 2010 NR NR NR NR NR NR NR NR 21.8 ± 3.8 30.6 ± 7.3 79 (10) 78 (12) NR NR Frljak 2018 30 (100) 30 (100) 31 (100) 32 (100) 2 (7) 3 (10) 32 (100) 33 (100) 32.2 ± 9.3 31.1 ± 7.8 NR NR NR NR Hamshere 2015 13 14 15 13 6 2 9 8 32.93 ± 16.46 29.75 ± 9.2 NR NR NR NR Hu* 2011 NR NR NR NR NR NR NR NR NR NR NR NR 466 (402, 495) 448 (383, 497) Matrino 2015 9 (11) 8 (10.2) 53 (64.1) 48 (61.1) 63 (77) 62 (79) 74 (89.7) 69 (88.9) 23.8 ± 7.2 24.7 ± 7.0 NR NR 347.3(146.7) 349.8(139.7) Sant'Anna 2014 NR NR NR NR NR NR NR NR NR NR NR NR 358.5 (88.69) 353 (86.67) Seth 2010 29 (70) 29 (72) 41 (100) 40 (100) NR NR NR NR NR NR NR NR NR NR Vrtovec 2011 21 (75) 22 (81) NR NR 5 (18) 6 (22) 26 (93) 24 (88) 25.6 ± 5.1 26.7 ± 3.9 69 ± 10 70 ± 7 NR NR Vrtovec 2013 43 (79) 46 (84) 51 (93) 54 (98) 9 (16) 11 (20) 51 (93) 20 (91) 24.3 ± 6.5 25.7 ± 4.1 69 ± 10 70 ± 7 NR NR Xiao 2017 16 (100) 20 (100) 16 (100) 19 (95) 4 (25.0) 8 (40.0) 5 (31.3) 6 (30.0) 33.1 ± 3.9 33.7 ± 4.0 NR NR 355.0 ± 91.2 323.3 ± 89.4 Data are reported as mean ± SD or n (%) unless proved otherwise TTT treatment group, NR not reported *Data are reported as median (IQR).
Collapse
|
25
|
Sim WS, Park BW, Ban K, Park HJ. In Situ Preconditioning of Human Mesenchymal Stem Cells Elicits Comprehensive Cardiac Repair Following Myocardial Infarction. Int J Mol Sci 2021; 22:1449. [PMID: 33535594 PMCID: PMC7867207 DOI: 10.3390/ijms22031449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 01/04/2023] Open
Abstract
Human bone marrow-derived mesenchymal stem cells (BM-MSCs), represented as a population of adult stem cells, have long been considered as one of the most promising sources for cell-based cardiac regenerative therapy. However, their clinical use has been significantly hampered by low survival and poor retention following administration into failing hearts. Here, to improve the therapeutic effectiveness of BM-MSCs, we examined a novel therapeutic platform named in situ preconditioning in a rat myocardial infarction (MI) model. In situ preconditioning was induced by a combinatory treatment of BM-MSCs with genetically engineered hepatocyte growth factor-expressing MSCs (HGF-eMSCs) and heart-derived extracellular matrix (hdECM) hydrogel. Subsequently, our results demonstrated that in situ preconditioning with cell mixture substantially improved the survival/retention of BM-MSCs in the MI-induced rat hearts. Enhanced retention of BM-MSCs ultimately led to a significant cardiac function improvement, which was derived from the protection of myocardium and enhancement of vessel formation in the MI hearts. The results provide compelling evidence that in situ preconditioning devised to improve the therapeutic potential of BM-MSCs can be an effective strategy to achieve cardiac repair of MI hearts.
Collapse
Affiliation(s)
- Woo-Sup Sim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea; (W.-S.S.); (B.-W.P.)
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea
| | - Bong-Woo Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea; (W.-S.S.); (B.-W.P.)
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Hun-Jun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea; (W.-S.S.); (B.-W.P.)
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 137701, Korea
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
26
|
Bui TVA, Hwang JW, Lee JH, Park HJ, Ban K. Challenges and Limitations of Strategies to Promote Therapeutic Potential of Human Mesenchymal Stem Cells for Cell-Based Cardiac Repair. Korean Circ J 2021; 51:97-113. [PMID: 33525065 PMCID: PMC7853896 DOI: 10.4070/kcj.2020.0518] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) represent a population of adult stem cells residing in many tissues, mainly bone marrow, adipose tissue, and umbilical cord. Due to the safety and availability of standard procedures and protocols for isolation, culturing, and characterization of these cells, MSCs have emerged as one of the most promising sources for cell-based cardiac regenerative therapy. Once transplanted into a damaged heart, MSCs release paracrine factors that nurture the injured area, prevent further adverse cardiac remodeling, and mediate tissue repair along with vasculature. Numerous preclinical studies applying MSCs have provided significant benefits following myocardial infarction. Despite promising results from preclinical studies using animal models, MSCs are not up to the mark for human clinical trials. As a result, various approaches have been considered to promote the therapeutic potency of MSCs, such as genetic engineering, physical treatments, growth factor, and pharmacological agents. Each strategy has targeted one or multi-potentials of MSCs. In this review, we will describe diverse approaches that have been developed to promote the therapeutic potential of MSCs for cardiac regenerative therapy. Particularly, we will discuss major characteristics of individual strategy to enhance therapeutic efficacy of MSCs including scientific principles, advantages, limitations, and improving factors. This article also will briefly introduce recent novel approaches that MSCs enhanced therapeutic potentials of other cells for cardiac repair.
Collapse
Affiliation(s)
- Thi Van Anh Bui
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Ji Won Hwang
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Korea.,Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Jung Hoon Lee
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Hun Jun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Korea.,Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.,Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
27
|
Efficacy and Mode of Action of Mesenchymal Stem Cells in Non-Ischemic Dilated Cardiomyopathy: A Systematic Review. Biomedicines 2020; 8:biomedicines8120570. [PMID: 33291410 PMCID: PMC7762005 DOI: 10.3390/biomedicines8120570] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 02/06/2023] Open
Abstract
Non-ischemic dilated cardiomyopathy (NIDCM) constitutes one of the most common causes to non-ischemic heart failure. Despite treatment, the disease often progresses, causing severe morbidity and mortality, making novel treatment strategies necessary. Due to the regenerative actions of mesenchymal stem cells (MSCs), they have been proposed as a treatment for NIDCM. This systematic review aims to evaluate efficacy and mode of action (MoA) of MSC-based therapies in NIDCM. A systematic literature search was conducted in Medline (Pubmed) and Embase. A total of 27 studies were included (3 clinical trials and 24 preclinical studies). MSCs from different tissues and routes of delivery were reported, with bone marrow-derived MSCs and direct intramyocardial injections being the most frequent. All included clinical trials and 22 preclinical trials reported an improvement in cardiac function following MSC treatment. Furthermore, preclinical studies demonstrated alterations in tissue structure, gene, and protein expression patterns, primarily related to fibrosis and angiogenesis. Consequently, MSC treatment can improve cardiac function in NIDCM patients. The MoA underlying this effect involves anti-fibrosis, angiogenesis, immunomodulation, and anti-apoptosis, though these processes seem to be interdependent. These encouraging results calls for larger confirmatory clinical studies, as well as preclinical studies utilizing unbiased investigation of the potential MoA.
Collapse
|
28
|
Nasser MI, Qi X, Zhu S, He Y, Zhao M, Guo H, Zhu P. Current situation and future of stem cells in cardiovascular medicine. Biomed Pharmacother 2020; 132:110813. [PMID: 33068940 DOI: 10.1016/j.biopha.2020.110813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular disease (CVD) is one of the leading causes of death worldwide. Currently, many methods have been proposed by researchers for the prevention and treatment of CVD; among them, stem cell-based therapies are the most promising. As the cells of origin for various mature cells, stem cells have the ability to self-renew and differentiate. Stem cells have a powerful ability to regenerate biologically, self-repair, and enhance damaged functional tissues or organs. Allogeneic stem cells and somatic stem cells are two types of cells that can be used for cardiac repair. Theoretically, dilated cardiomyopathy and acute myocardial infarction can be treated with such cells. In addition, stem cell transplantation procedures, including intravenous, epicardial, cardiac, and endocardial injections, have been reported to provide significant benefits in clinical practice; however, there are still a number of issues that need further study and consideration, such as the form and quantity of transplanted cells and post-transplantation health. The goal of this analysis was to summarize the recent advances in stem cell-based therapies and their efficacy in cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- M I Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Xiao Qi
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Shuoji Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Yin He
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Mingyi Zhao
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Huiming Guo
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, 510100, China. Address: 106 Zhongshan Er Road, Guangzhou, 510080, PR China.
| |
Collapse
|
29
|
Xia L, Zeng L, Pan J, Ding Y. Effects of stem cells on non-ischemic cardiomyopathy: a systematic review and meta-analysis of randomized controlled trials. Cytotherapy 2020; 22:699-711. [PMID: 32893120 DOI: 10.1016/j.jcyt.2020.06.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/01/2020] [Accepted: 06/19/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND AIMS To assess the impacts of stem cell therapy on clinical outcomes in patients with non-ischemic cardiomyopathy (NICM). The effect of stem cell therapy on prognosis is unclear and controversial. METHODS The authors performed a systematic review and meta-analysis of the effects of autologous stem cell transplantation in patients with NICM on a composite outcome of all-cause mortality and heart transplantation, left ventricular ejection fraction (LVEF), left ventricular end-diastolic diameter (LVEDD), New York Heart Association (NYHA) classification, 6-minute walk test (6-MWT) distance and serum brain natriuretic peptide (BNP) level, considering studies published before March 19, 2020. RESULTS Twelve trials with 623 subjects met inclusion criteria. Compared with the control group, stem cell therapy improved LVEF (weighted mean difference [WMD], 4.08%, 95% confidence interval [CI], 1.93-6.23, P = 0.0002) and 6-MWT distance (WMD, 101.49 m, 95% CI, 45.62-157.35, P = 0.0004) and reduced BNP level (-294.94 pg/mL, 95% CI, -383.97 to -205.90, P < 0.00001) and NYHA classification (-0.70, 95% CI, -0.98 to -0.43, P < 0.00001). However, LVEDD showed no significant difference between the two groups (WMD, -0.09 cm, 95% CI, -0.23 to 0.06, P = 0.25). In 10 studies (535 subjects) employing the intracoronary route for cell delivery, mortality and heart transplantation were decreased (risk ratio [RR], 0.73, 95% CI, 0.52-1.00, P = 0.05). Furthermore, in four studies (248 subjects) with peripheral CD34+ cells, either all-cause mortality (RR, 0.44, 95% CI, 0.23-0.86, P = 0.02) or mortality and heart transplantation (RR, 0.45, 95% CI, 0.27-0.77, P = 0.003) improved in the treatment group compared with the control. The trial sequential analysis suggested the information size of LVEF, 6-WMT and BNP has been adequate for evidencing the benefits of stem cells on NICM. However, to determine the potential survival benefit, more clinical data are required to make the statistical significance in meta-analysis more conclusive. CONCLUSIONS This meta-analysis demonstrates that stem cell therapy may improve survival, exercise capacity and cardiac ejection fraction in NICM, which suggests that stem cells are a promising option for NICM treatment.
Collapse
Affiliation(s)
- Liang Xia
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, China.
| | - LingHui Zeng
- Department of Pharmacology, School of Medicine, Zhejiang University City College, Hangzhou, China
| | - JianPing Pan
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, China
| | - YueMin Ding
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, China
| |
Collapse
|
30
|
Thompson M, Mei SH, Wolfe D, Champagne J, Fergusson D, Stewart DJ, Sullivan KJ, Doxtator E, Lalu M, English SW, Granton J, Hutton B, Marshall J, Maybee A, Walley KR, Santos CD, Winston B, McIntyre L. Cell therapy with intravascular administration of mesenchymal stromal cells continues to appear safe: An updated systematic review and meta-analysis. EClinicalMedicine 2020; 19:100249. [PMID: 31989101 PMCID: PMC6970160 DOI: 10.1016/j.eclinm.2019.100249] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/04/2019] [Accepted: 12/17/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Characterization of the mesenchymal stromal cell (MSC) safety profile is important as this novel therapy continues to be evaluated in clinical trials for various inflammatory conditions. Due to an increase in published randomized controlled trials (RCTs) from 2012-2019, we performed an updated systematic review to further characterize the MSC safety profile. METHODS MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials and Web of Science (to May 2018) were searched. RCTs that compared intravascular delivery of MSCs to controls in adult populations were included. Pre-specified adverse events were grouped according to: (1) immediate, (2) infection, (3) thrombotic/embolic, and (4) longer-term events (mortality, malignancy). Adverse events were pooled and meta-analyzed by fitting inverse-variance binary random effects models. Primary and secondary clinical efficacy endpoints were summarized descriptively. FINDINGS 7473 citations were reviewed and 55 studies met inclusion criteria (n = 2696 patients). MSCs as compared to controls were associated with an increased risk of fever (Relative Risk (RR) = 2·48, 95% Confidence Interval (CI) = 1·27-4·86; I2 = 0%), but not non-fever acute infusional toxicity, infection, thrombotic/embolic events, death, or malignancy (RR = 1·16, 0·99, 1·14, 0·78, 0·93; 95% CI = 0·70-1·91, 0·81-1·21, 0·67-1·95, 0·65-0·94, 0·60-1·45; I2 = 0%, 0%, 0%, 0%, 0%). No included trials were ended prematurely due to safety concerns. INTERPRETATIONS MSC therapy continues to exhibit a favourable safety profile. Future trials should continue to strengthen study rigor, reporting of MSC characterization, and adverse events. FUNDING Stem Cell Network, Ontario Institute for Regenerative Medicine and Ontario Research Fund.
Collapse
Affiliation(s)
- Mary Thompson
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Shirley H.J. Mei
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Dianna Wolfe
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Josée Champagne
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Dean Fergusson
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Duncan J. Stewart
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Katrina J. Sullivan
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Emily Doxtator
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Manoj Lalu
- Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Ottawa, Ontario, Canada
| | - Shane W. English
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - John Granton
- Department of Medicine (Critical Care), University of Toronto, Toronto, Ontario, Canada
| | - Brian Hutton
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - John Marshall
- Department of Surgery (Critical Care), University of Toronto, Toronto, Ontario, Canada
| | - Alies Maybee
- Patient Advisors Network, Toronto, Ontario, Canada
| | - Keith R. Walley
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Claudia Dos Santos
- Department of Surgery (Critical Care), University of Toronto, Toronto, Ontario, Canada
| | - Brent Winston
- Department of Critical Care, Medicine, and Biochemistry and Microbiology, University of Calgary, Calgary, Alberta, Canada
| | - Lauralyn McIntyre
- Clinical Epidemiology Program (CEP), Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
31
|
Banerjee MN, Bolli R, Hare JM. Clinical Studies of Cell Therapy in Cardiovascular Medicine: Recent Developments and Future Directions. Circ Res 2019; 123:266-287. [PMID: 29976692 DOI: 10.1161/circresaha.118.311217] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Given the rising prevalence of cardiovascular disease worldwide and the limited therapeutic options for severe heart failure, novel technologies that harness the regenerative capacity of the heart are sorely needed. The therapeutic use of stem cells has the potential to reverse myocardial injury and improve cardiac function, in contrast to most current medical therapies that only mitigate heart failure symptoms. Nearly 2 decades and >200 trials for cardiovascular disease have revealed that most cell types are safe; however, their efficacy remains controversial, limiting the transition of this therapy from investigation to practice. Lessons learned from these initial studies are driving the design of new clinical trials; higher fidelity of cell isolation techniques, standardization of conditions, more consistent use of state of the art measurement techniques, and assessment of multiple end points to garner insights into the efficacy of stem cells. Translation to clinical trials has almost outpaced our mechanistic understanding, and individual patient factors likely play a large role in stem cell efficacy. Therefore, careful analysis of dosing, delivery methods, and the ideal patient populations is necessary to translate cell therapy from research to practice. We are at a pivotal stage in the field in which information from many relatively small clinical trials must guide carefully executed efficacy trials. Larger efficacy trials are being launched to answer questions about older, first-generation stem cell therapeutics, while novel, second-generation products are being introduced into the clinical realm. This review critically examines the current state of clinical research on cell-based therapies for cardiovascular disease, highlighting the controversies in the field, improvements in clinical trial design, and the application of exciting new cell products.
Collapse
Affiliation(s)
- Monisha N Banerjee
- From the Interdisciplinary Stem Cell Institute (M.N.B., J.M.H.).,Department of Surgery (M.N.B)
| | - Roberto Bolli
- University of Miami Miller School of Medicine, FL; and Institute of Molecular Cardiology, University of Louisville, KY (R.B.)
| | - Joshua M Hare
- From the Interdisciplinary Stem Cell Institute (M.N.B., J.M.H.) .,Department of Medicine (J.M.H.)
| |
Collapse
|
32
|
Diaz-Navarro R, Urrútia G, Cleland JGF, Poloni D, Villagran F, Bangdiwala S, Rada G, Madrid E. Stem cell therapy for dilated cardiomyopathy. Hippokratia 2019. [DOI: 10.1002/14651858.cd013433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Rienzi Diaz-Navarro
- Universidad de Valparaiso; Department of Internal Medicine, School of Medicine; Vina del Mar Chile
| | - Gerard Urrútia
- CIBER Epidemiología y Salud Pública (CIBERESP); Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau); Sant Antoni Maria Claret, 167 Pavilion 18 (D-53) Barcelona Catalonia Spain 08025
| | - John GF Cleland
- Imperial College London; National Heart and Lung Institute; London UK
| | - Daniel Poloni
- Universidad de Valparaiso; Department of Internal Medicine, School of Medicine; Vina del Mar Chile
| | - Francisco Villagran
- Universidad de Valparaiso; Department of Internal Medicine, School of Medicine; Vina del Mar Chile
| | - Shrikant Bangdiwala
- Collaborative Studies Coordinating Center; Department of Biostatistics, Gillings School of Global Public Health; Suite 203, Bank of America Center 137 E. Franklin Street Chapel Hill North Carolina USA 27514-4145
| | - Gabriel Rada
- Pontificia Universidad Católica de Chile; Department of Internal Medicine and Evidence-Based Healthcare Program, Faculty of Medicine; Lira 44, Decanato Primer piso Santiago Chile
| | - Eva Madrid
- Cochrane Centre School of Medicine Universidad de Valparaiso; Interdisciplinary Centre for Health Studies CIESAL; Viña del Mar Chile
- Universidad de Valparaiso; Chilean Cochrane Centre; Valparaiso Chile
| |
Collapse
|
33
|
Rong SL, Wang ZK, Zhou XD, Wang XL, Yang ZM, Li B. Efficacy and safety of stem cell therapy in patients with dilated cardiomyopathy: a systematic appraisal and meta-analysis. J Transl Med 2019; 17:221. [PMID: 31296244 PMCID: PMC6624954 DOI: 10.1186/s12967-019-1966-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/27/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The clinical significance of stem cell therapy in the treatment of dilated cardiomyopathy remains unclear. This systemic appraisal and meta-analysis aimed to assess the efficacy and safety of stem cell therapy in patients with dilated cardiomyopathy. After searching the PubMed, Embase, and Cochrane library databases until November 2017, we conducted a meta-analysis to evaluate the efficacy and safety of stem cell therapy in patients with dilated cardiomyopathy. METHODS The weighted mean difference (WMD), standard mean difference (SMD), relative risk (RR), and 95% confidence interval (CI) were summarized in this meta-analysis. Both fixed effects and random effects models were used to combine the data. Sensitivity analyses were conducted to evaluate the impact of an individual dataset on the pooled results. RESULTS A total of eight randomized controlled trials, which involved 531 participants, met the inclusion criteria in this systematic appraisal and meta-analysis. Our meta-analysis showed that stem cell therapy improves left ventricular ejection fraction (SMD = 1.09, 95% CI 0.29 to 1.90, I2 = 92%) and reduces left ventricular end-systolic volume (SMD = - 0.36, 95% CI - 0.61 to - 0.10, I2 = 20.5%) and left ventricular end-diastolic chamber size (SMD = - 0.48, 95% CI - 0.89 to - 0.07, I2 = 64.8%) in patients with dilated cardiomyopathy. However, stem cell therapy has no effect on mortality (RR = 0.72, 95% CI 0.50 to 1.02, I2 = 30.2%) and 6-min-walk test (WMD = 51.52, 95% CI - 24.52 to 127.55, I2 = 94.8%). CONCLUSIONS This meta-analysis suggests that stem cell therapy improves left ventricular ejection fraction and reduces left ventricular end-systolic volume and left ventricular end-diastolic chamber size in patients with dilated cardiomyopathy. However, future well-designed large studies might be necessary to clarify the effect of stem cell therapy in patients with dilated cardiomyopathy.
Collapse
Affiliation(s)
- Shu-Ling Rong
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi People’s Republic of China
| | - Ze-Kun Wang
- State Key Laboratory of Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Xue-Dong Zhou
- State Key Laboratory of Oral Diseases, Department of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan People’s Republic of China
| | - Xiao-Lin Wang
- Department of Neonatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi People’s Republic of China
| | - Zhi-Ming Yang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi People’s Republic of China
| | - Bao Li
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi People’s Republic of China
| |
Collapse
|
34
|
Stem cell therapy in heart failure: Where do we stand today? Biochim Biophys Acta Mol Basis Dis 2019; 1866:165489. [PMID: 31199998 DOI: 10.1016/j.bbadis.2019.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 02/06/2023]
Abstract
Heart failure is a global epidemic that drastically cuts short longevity and compromises quality of life. Approximately 6 million Americans ≥20 years of age carry a diagnosis of heart failure. Worldwide, about 40 million adults are affected. The treatment of HF depends on the etiology. If left untreated it rapidly progresses and compromises quality of life. One of the newer technologies still in its infancy is stem cell therapy for heart failure. This review attempts to highlight the clinical studies done in ischemic cardiomyopathy, dilated cardiomyopathy and restrictive cardiomyopathy. A combined approach of simultaneous revascularization and stem cell therapy appears to produce maximum benefit in ischemic cardiomyopathy. Treatment of dilated cardiomyopathy with stem cells also holds promise but needs more definition with regards to timing, route of cell delivery and type of cell used to achieve reproducible results. The variability noted in response to stem cell therapy in patients could also be secondary to their co-morbidities. Abnormalities of glucose metabolism and diabetes in particular impair stem cell and angiogenic cell mobilization. This opens up a whole new area of investigation into exploring the biochemical microenvironment which could influence the efficacy of stem cell therapy. This article is part of a Special Issue entitled: Stem Cells and Their Applications to Human Diseases edited by Hemachandra Reddy.
Collapse
|
35
|
Fan M, Huang Y, Chen Z, Xia Y, Chen A, Lu D, Wu Y, Zhang N, Qian J. Efficacy of mesenchymal stem cell therapy in systolic heart failure: a systematic review and meta-analysis. Stem Cell Res Ther 2019; 10:150. [PMID: 31151406 PMCID: PMC6544951 DOI: 10.1186/s13287-019-1258-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 12/28/2022] Open
Abstract
Background Heart failure (HF) is the end stage of most heart disease. Mesenchymal stem cells (MSCs), with their specific biological effects, have been applied in several clinical trials to evaluate the efficacy in HF therapy. We performed this meta-analysis to review the clinical evidence of their therapeutic effect on HF. Methods Three databases were searched. The outcomes of interest were death, readmission, the 6-min walk test (6MWT), New York Heart Association (NYHA) class and left ventricular ejection fraction (LVEF). The relative risk (RR) and weighted mean difference (WMD) were calculated to evaluate the effects of MSCs on HF compared to placebo. Results A total of nine studies were included, involving 612 patients who underwent MSCs or placebo treatment. The overall rate of death showed a trend of reduction of 36% (RR [CI] = 0.64 [0.35, 1.16], p = 0.143) in the MSC treatment group. The incidence of readmission was reduced by 34% (RR [CI] = 0.66 [0.51, 0.85], p = 0.001). The patients in the MSC treatment group realised an average of 40.44 m (WMD [95% CI] = 40.44 m [19.07, 61.82], p < 0.0001) improvement in 6MWT. The NYHA class was reduced obviously in the MSC group (WMD [95% CI] = − 0.42 [− 0.64, − 0.20], p < 0.0001). The changes of LVEF from baseline were significantly more than 5.25% (WMD [95% CI] = 5.25 [3.58, 6.92], p < 0.0001) in the MSCs group, unlike in the placebo group. Conclusions Our results suggested that MSC treatment is an effective therapy for HF by improving the prognosis and exercise capacity. SCs derived from allosomes have superior therapeutic effects, and intracoronary injection is the optimum MSC delivery approach. Short-term cryopreservation is feasible in MSCs storage or transport.
Collapse
Affiliation(s)
- Mengkang Fan
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yin Huang
- Department of Geriatric Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Zhangwei Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yan Xia
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Ao Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Danbo Lu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yuan Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Ning Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Juying Qian
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
36
|
Wang C, Li J, Zhang B, Li Y. Safety and efficacy of bone marrow-derived cells therapy on cardiomyopathy: a meta-analysis. Stem Cell Res Ther 2019; 10:137. [PMID: 31109372 PMCID: PMC6528271 DOI: 10.1186/s13287-019-1238-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Controversial results still existed on the clinical utility of bone marrow-derived cells (BMCs) for cardiomyopathy (CMP). This study aims to reveal the true power of this promising approach by synthesizing all the available data on this subject matter. METHODS Twenty studies including 1418 patients were identified from systematic search. Weighted mean differences for changes in left ventricular ejection fraction (LVEF), left ventricular end-diastolic volume (LVEDV), left ventricular end-systolic volume (LVESV), 6-min walk distance, and NYHA functional class were estimated with a random-effects model. Major adverse cardiovascular event (MACE), rehospitalization, all-cause mortality, and patients' quality of life were also calculated. RESULTS Compared with the control group, BMC therapy resulted in greater LVEF (3.72%, 95% CI 2.31 to 5.13, P < 0.0001), 6-min walk distance (53.16, 95% CI 25.17 to 81.10, P = 0.0002), NYHA functional class (- 0.48, 95% CI - 0.65 to - 0.31, P < 0.0001), and smaller LVESV (- 16.79, 95% CI - 27.21 to - 6.38, P = 0.002). BMC treatment significantly reduced the mortality rate and improved patients' quality of life. No significant difference was found between the BMCs and control group in LVEDV, MACE, and rehospitalization rate. However, the outcomes showed a clear trend in favor of the BMC group. Subgroup analysis showed that LVEF improved greater in a subgroup of intracoronary infusion, BMSC, or higher cell dose. CONCLUSION The results of the current meta-analysis suggest that BMC treatment for CMP is safe and feasible. This therapy was associated with persistent improvements in LV function, LV remodeling, functional class, patients' survival, and quality of life. Intracoronary infusion of high-dose (> 108) BMSC might be a better therapeutic option for CMP patients. Further evidences are needed to verify our results.
Collapse
Affiliation(s)
- Chao Wang
- Department of Cardiology, Tianjin Nankai Hospital, No. 6 Changjiang Road, Nankai District, Tianjin, China.
| | - Jingzhao Li
- Department of Cardiology, Tianjin Nankai Hospital, No. 6 Changjiang Road, Nankai District, Tianjin, China
| | - Boya Zhang
- Department of Cardiology, Tianjin Nankai Hospital, No. 6 Changjiang Road, Nankai District, Tianjin, China
| | - Yongjian Li
- Department of Cardiology, Tianjin Nankai Hospital, No. 6 Changjiang Road, Nankai District, Tianjin, China
| |
Collapse
|
37
|
Yagyu T, Yasuda S, Nagaya N, Doi K, Nakatani T, Satomi K, Shimizu W, Kusano K, Anzai T, Noguchi T, Ohgushi H, Kitamura S, Kangawa K, Ogawa H. Long-Term Results of Intracardiac Mesenchymal Stem Cell Transplantation in Patients With Cardiomyopathy. Circ J 2019; 83:1590-1599. [PMID: 31105128 DOI: 10.1253/circj.cj-18-1179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs), which have the potential to differentiate into cardiomyocytes or vascular endothelial cells, have been used clinically as therapy for cardiomyopathy. In this study, we aimed to evaluate the long-term follow-up results.Methods and Results:We studied 8 patients with symptomatic heart failure (HF) on guideline-directed therapy (ischemic cardiomyopathy, n=3; nonischemic cardiomyopathy, n=5) who underwent intracardiac MSC transplantation using a catheter-based injection method between May 2004 and April 2006. Major adverse events and hospitalizations were investigated up to 10 years afterward. Compared with baseline, there were no significant differences in B-type natriuretic peptide (BNP) (from 211 to 173 pg/mL), left ventricular ejection fraction (LVEF) (from 24% to 26%), and peak oxygen uptake (from 16.5 to 19.2 mL/min/kg) at 2 months. During the follow-up period, no patients experienced serious adverse events such as arrhythmias. Three patients died of pneumonia in the 1st year, liver cancer in the 6th year, and HF in the 7th year. Of the remaining 5 patients, 3 patients were hospitalized for exacerbated HF, 1 of whom required heart transplantation in the 2nd year; 2 patients survived for 10 years without worsening HF. CONCLUSIONS The results of this exploratory study of intracardiac MSCs administration suggest further research regarding the feasibility and efficacy is warranted.
Collapse
Affiliation(s)
- Takeshi Yagyu
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center.,Department of Cardiovascular Medicine, Kumamoto University Graduate School of Medical Sciences
| | - Satoshi Yasuda
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center.,Department of Cardiovascular Medicine, Kumamoto University Graduate School of Medical Sciences
| | | | - Kaori Doi
- Research Institute, National Cerebral and Cardiovascular Center
| | - Takeshi Nakatani
- Department of Transplantation, National Cerebral and Cardiovascular Center
| | - Kazuhiro Satomi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center.,Department of Cardiology, Tokyo Medical University
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center.,Department of Cardiovascular Medicine, Nippon Medical School
| | - Kengo Kusano
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | - Toshihisa Anzai
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center.,Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine
| | - Teruo Noguchi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center
| | - Hajime Ohgushi
- National Institute of Advanced Industrial Science and Technology
| | - Soichiro Kitamura
- Department of Transplantation, National Cerebral and Cardiovascular Center
| | - Kenji Kangawa
- Research Institute, National Cerebral and Cardiovascular Center
| | - Hisao Ogawa
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center.,Research Institute, National Cerebral and Cardiovascular Center
| |
Collapse
|
38
|
Streeter BW, Davis ME. Therapeutic Cardiac Patches for Repairing the Myocardium. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1144:1-24. [DOI: 10.1007/5584_2018_309] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
39
|
Poltavtseva RA, Poltavtsev AV, Lutsenko GV, Svirshchevskaya EV. Myths, reality and future of mesenchymal stem cell therapy. Cell Tissue Res 2018; 375:563-574. [PMID: 30456646 DOI: 10.1007/s00441-018-2961-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 11/05/2018] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cell (MSC) therapy represents an alternative approach for tissue regeneration and inflammation control. In spite of a huge amount of preclinical data that has been accumulated on the therapeutic properties of MSCs, there are many conflicting results, possibly due to differences in the properties of MSCs obtained from different sources or underestimated mechanisms of MSC in vivo behavior. This review consolidates the in vivo effects of MSC therapy, discusses the fate of MSCs after intravascular and local delivery and proposes possible trends in MSC therapy.
Collapse
Affiliation(s)
- R A Poltavtseva
- Federal State Budget Institution "Research Center for Obstetrics, Gynecology and Perinatology" Ministry of Healthcare of the Russian Federation, Oparin St, 4, Moscow, Russian Federation, 117997
| | - A V Poltavtsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya St, 16/10, Moscow, Russian Federation, 117997
| | - G V Lutsenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya St, 16/10, Moscow, Russian Federation, 117997
| | - E V Svirshchevskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, Miklukho-Maklaya St, 16/10, Moscow, Russian Federation, 117997.
| |
Collapse
|
40
|
Cell-Based Therapies for Cardiac Regeneration: A Comprehensive Review of Past and Ongoing Strategies. Int J Mol Sci 2018; 19:ijms19103194. [PMID: 30332812 PMCID: PMC6214096 DOI: 10.3390/ijms19103194] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/11/2018] [Accepted: 10/12/2018] [Indexed: 12/20/2022] Open
Abstract
Despite considerable improvements in the treatment of cardiovascular diseases, heart failure (HF) still represents one of the leading causes of death worldwide. Poor prognosis is mostly due to the limited regenerative capacity of the adult human heart, which ultimately leads to left ventricular dysfunction. As a consequence, heart transplantation is virtually the only alternative for many patients. Therefore, novel regenerative approaches are extremely needed, and several attempts have been performed to improve HF patients’ clinical conditions by promoting the replacement of the lost cardiomyocytes and by activating cardiac repair. In particular, cell-based therapies have been shown to possess a great potential for cardiac regeneration. Different cell types have been extensively tested in clinical trials, demonstrating consistent safety results. However, heterogeneous efficacy data have been reported, probably because precise end-points still need to be clearly defined. Moreover, the principal mechanism responsible for these beneficial effects seems to be the paracrine release of antiapoptotic and immunomodulatory molecules from the injected cells. This review covers past and state-of-the-art strategies in cell-based heart regeneration, highlighting the advantages, challenges, and limitations of each approach.
Collapse
|
41
|
Kobayashi K, Suzuki K. Mesenchymal Stem/Stromal Cell-Based Therapy for Heart Failure ― What Is the Best Source? ―. Circ J 2018; 82:2222-2232. [DOI: 10.1253/circj.cj-18-0786] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kazuya Kobayashi
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London
| | - Ken Suzuki
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London
| |
Collapse
|
42
|
Barwinska D, Traktuev DO, Merfeld-Clauss S, Cook TG, Lu H, Petrache I, March KL. Cigarette Smoking Impairs Adipose Stromal Cell Vasculogenic Activity and Abrogates Potency to Ameliorate Ischemia. Stem Cells 2018; 36:856-867. [PMID: 29589872 DOI: 10.1002/stem.2813] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 02/05/2018] [Accepted: 02/05/2018] [Indexed: 12/17/2022]
Abstract
Cigarette smoking (CS) adversely affects the physiologic function of endothelial progenitor, hematopoietic stem and progenitor cells. However, the effect of CS on the ability of adipose stem/stromal cells (ASC) to promote vasculogenesis and rescue perfusion in the context of ischemia is unknown. To evaluate this, ASC from nonsmokers (nCS-ASC) and smokers (CS-ASC), and their activity to promote perfusion in hindlimb ischemia models, as well as endothelial cell (EC) survival and vascular morphogenesis in vitro were assessed. While nCS-ASC improved perfusion in ischemic limbs, CS-ASC completely lost this therapeutic effect. In vitro vasculogenesis assays revealed that human CS-ASC and ASC from CS-exposed mice showed compromised support of EC morphogenesis into vascular tubes, and the CS-ASC secretome was less potent in supporting EC survival/proliferation. Comparative secretome analysis revealed that CS-ASC produced lower amounts of hepatocyte growth factor (HGF) and stromal cell-derived growth factor 1 (SDF-1). Conversely, CS-ASC secreted the angiostatic/pro-inflammatory factor Activin A, which was not detected in nCS-ASC conditioned media (CM). Furthermore, higher Activin A levels were measured in EC/CS-ASC cocultures than in EC/nCS-ASC cocultures. CS-ASC also responded to inflammatory cytokines with 5.2-fold increase in Activin A secretion, whereas nCS-ASC showed minimal Activin A induction. Supplementation of EC/CS-ASC cocultures with nCS-ASC CM or with recombinant vascular endothelial growth factor, HGF, or SDF-1 did not rescue vasculogenesis, whereas inhibition of Activin A expression or activity improved network formation up to the level found in EC/nCS-ASC cocultures. In conclusion, ASC of CS individuals manifest compromised in vitro vasculogenic activity as well as in vivo therapeutic activity. Stem Cells 2018;36:856-867.
Collapse
Affiliation(s)
- Daria Barwinska
- Department of Cellular and Integrative Physiology.,Krannert Institute of Cardiology.,Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA
| | - Dmitry O Traktuev
- Krannert Institute of Cardiology.,Division of Cardiology.,Department of Medicine, Indiana University, Indianapolis, Indiana, USA.,Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA.,Center for Regenerative Medicine, Department of Medicine University of Florida, Gainesville, Florida, USA
| | - Stephanie Merfeld-Clauss
- Krannert Institute of Cardiology.,Division of Cardiology.,Department of Medicine, Indiana University, Indianapolis, Indiana, USA.,Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA.,Center for Regenerative Medicine, Department of Medicine University of Florida, Gainesville, Florida, USA
| | - Todd G Cook
- Krannert Institute of Cardiology.,Division of Cardiology.,Department of Medicine, Indiana University, Indianapolis, Indiana, USA.,Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA
| | - Hongyan Lu
- Krannert Institute of Cardiology.,Division of Cardiology.,Department of Medicine, Indiana University, Indianapolis, Indiana, USA.,Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA
| | - Irina Petrache
- Department of Medicine, Indiana University, Indianapolis, Indiana, USA.,Department of Medicine, National Jewish Health and University of Colorado, Denver, Colorado, USA
| | - Keith L March
- Department of Cellular and Integrative Physiology.,Krannert Institute of Cardiology.,Division of Cardiology.,Department of Medicine, Indiana University, Indianapolis, Indiana, USA.,Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana, USA.,Center for Regenerative Medicine, Department of Medicine University of Florida, Gainesville, Florida, USA
| |
Collapse
|
43
|
Abstract
During the past decades, stem cell-based therapy has acquired a promising role in regenerative medicine. The application of novel cell therapeutics for the treatment of cardiovascular diseases could potentially achieve the ambitious aim of effective cardiac regeneration. Despite the highly positive results from preclinical studies, data from phase I/II clinical trials are inconsistent and the improvement of cardiac remodeling and heart performance was found to be quite limited. The major issues which cardiac stem cell therapy is facing include inefficient cell delivery to the site of injury, accompanied by low cell retention and weak effectiveness of remaining stem cells in tissue regeneration. According to preclinical and clinical studies, various stem cells (adult stem cells, embryonic stem cells, and induced pluripotent stem cells) represent the most promising cell types so far. Beside the selection of the appropriate cell type, researchers have developed several strategies to produce “second-generation” stem cell products with improved regenerative capacity. Genetic and nongenetic modifications, chemical and physical preconditioning, and the application of biomaterials were found to significantly enhance the regenerative capacity of transplanted stem cells. In this review, we will give an overview of the recent developments in stem cell engineering with the goal to facilitate stem cell delivery and to promote their cardiac regenerative activity.
Collapse
|
44
|
Regenerative Medicine Applications of Mesenchymal Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1089:115-141. [PMID: 29767289 DOI: 10.1007/5584_2018_213] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A major research challenge is to develop therapeutics that assist with healing damaged tissues and organs because the human body has limited ability to restore the majority of these tissues and organs to their original state. Tissue engineering (TE) and regenerative medicine (RM) promises to offer efficient therapeutic biological strategies that use mesenchymal stem cells (MSCs). MSCs possess the capability for self-renewal, multilineage differentiation, and immunomodulatory properties that make them attractive for clinical applications. They have been extensively investigated in numerous preclinical and clinical settings in an attempt to overcome their challenges and promote tissue regeneration and repair. This review explores the exciting opportunities afforded by MSCs, their desirable properties as cellular therapeutics in RM, and implicates their potential use in clinical practice. Here, we attempt to identify challenges and issues that determine the clinical efficacy of MSCs as treatment for skeletal and non-skeletal tissues.
Collapse
|