101
|
Chen C, Peng H, Zeng Y, Dong G. CD14, CD163, and CCR1 are involved in heart and blood communication in ischemic cardiac diseases. J Int Med Res 2021; 48:300060520951649. [PMID: 32967511 PMCID: PMC7521061 DOI: 10.1177/0300060520951649] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Objective Cardiac diseases lead to heart failure (HF), but the progression can take several years. Using blood samples to monitor changes in the heart before clinical symptoms begin may help to improve patient management. Methods Microarray data GSE42955 and GSE9128 were used as study datasets and GSE16499, GSE57338, and GSE59867 were used as validation groups. The “limma” package from R Language was used to identify differentially expressed genes. Functional enrichment analyses of gene ontology terms and Kyoto Encyclopedia of Genes and Genomes pathways were performed using Database for Annotation, Visualization and Integrated Discovery. We also investigated the correlation between the heart and blood using the mRNA expression level. Results Three hub genes, CD14, CD163, and CCR1, were identified. Functional enrichment analyses showed their involvement in the immune response and in the inflammatory response, which are the critical biochemical processes in ischemic HF. The mRNA expression level further demonstrated that a special model may exist to help to predict the mRNA level in the heart based on that in blood. Conclusions Our study identified three biomarkers that can connect the heart and blood in ischemic heart diseases, which may be a new approach to help better manage ischemic cardiac disease patients.
Collapse
Affiliation(s)
- Chengcong Chen
- Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Hong Peng
- Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Yongmei Zeng
- Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China
| | - Guoqing Dong
- Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China
| |
Collapse
|
102
|
Rheault-Henry M, White I, Grover D, Atoui R. Stem cell therapy for heart failure: Medical breakthrough, or dead end? World J Stem Cells 2021; 13:236-259. [PMID: 33959217 PMCID: PMC8080540 DOI: 10.4252/wjsc.v13.i4.236] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/22/2020] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
Heart failure continues to be one of the leading causes of morbidity and mortality worldwide. Myocardial infarction is the primary causative agent of chronic heart failure resulting in cardiomyocyte necrosis and the subsequent formation of fibrotic scar tissue. Current pharmacological and non-pharmacological therapies focus on managing symptoms of heart failure yet remain unable to reverse the underlying pathology. Heart transplantation usually cannot be relied on, as there is a major discrepancy between the availability of donors and recipients. As a result, heart failure carries a poor prognosis and high mortality rate. As the heart lacks significant endogenous regeneration potential, novel therapeutic approaches have incorporated the use of stem cells as a vehicle to treat heart failure as they possess the ability to self-renew and differentiate into multiple cell lineages and tissues. This review will discuss past, present, and future clinical trials, factors that influence stem cell therapy outcomes as well as ethical and safety considerations. Preclinical and clinical studies have shown a wide spectrum of outcomes when applying stem cells to improve cardiac function. This may reflect the infancy of clinical trials and the limited knowledge on the optimal cell type, dosing, route of administration, patient parameters and other important variables that contribute to successful stem cell therapy. Nonetheless, the field of stem cell therapeutics continues to advance at an unprecedented pace. We remain cautiously optimistic that stem cells will play a role in heart failure management in years to come.
Collapse
Affiliation(s)
| | - Ian White
- Northern Ontario School of Medicine, Sudbury P3E 2C6, Ontario, Canada
| | - Diya Grover
- Ross University School of Medicine, St. Michael BB11093, Barbados
| | - Rony Atoui
- Division of Cardiac Surgery, Health Sciences North, Northern Ontario School of Medicine, Sudbury P3E 3Y9, Ontario, Canada
| |
Collapse
|
103
|
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: 5.5] [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
|
104
|
Xie DM, Chen Y, Liao Y, Lin W, Dai G, Lu DH, Zhu S, Yang K, Wu B, Chen Z, Peng C, Jiang MH. Cardiac Derived CD51-Positive Mesenchymal Stem Cells Enhance the Cardiac Repair Through SCF-Mediated Angiogenesis in Mice With Myocardial Infarction. Front Cell Dev Biol 2021; 9:642533. [PMID: 33968928 PMCID: PMC8098770 DOI: 10.3389/fcell.2021.642533] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/15/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: Many tissues contained resident mesenchymal stromal/stem cells (MSCs) that facilitated tissue hemostasis and repair. However, there is no typical marker to identify the resident cardiac MSCs. We aimed to determine if CD51 could be an optimal marker of cardiac MSCs and assess their therapeutic potential for mice with acute myocardial infarction (AMI). Methods: Cardiac-derived CD51+CD31–CD45–Ter119– cells (named CD51+cMSCs) were isolated from C57BL/6 mice(7-day-old) by flow cytometry. The CD51+cMSCs were characterized by proliferation capacity, multi-differentiation potential, and expression of typical MSC-related markers. Adult C57BL/6 mice (12-week-old) were utilized for an AMI model via permanently ligating the left anterior descending coronary artery. The therapeutic efficacy of CD51+cMSCs was estimated by echocardiography and pathological staining. To determine the underlying mechanism, lentiviruses were utilized to knock down gene (stem cell factor [SCF]) expression of CD51+cMSCs. Results: In this study, CD51 was expressed in the entire layers of the cardiac wall in mice, including endocardium, epicardium, and myocardium, and its expression was decreased with age. Importantly, the CD51+cMSCs possessed potent self-renewal potential and multi-lineage differentiation capacity in vitro and also expressed typical MSC-related surface proteins. Furthermore, CD51+cMSC transplantation significantly improved cardiac function and attenuated cardiac fibrosis through pro-angiogenesis activity after myocardial infarction in mice. Moreover, SCF secreted by CD51+cMSCs played an important role in angiogenesis both in vivo and in vitro. Conclusions: Collectively, CD51 is a novel marker of cardiac resident MSCs, and CD51+cMSC therapy enhances cardiac repair at least partly through SCF-mediated angiogenesis.
Collapse
Affiliation(s)
- Dong Mei Xie
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Stem Cells and Tissue Engineering, Center for Stem Cell Biology and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Yang Chen
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Liao
- Shenzhen Beike Biotechnology Co., Ltd., Shenzhen, China
| | - Wanwen Lin
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Gang Dai
- NHC Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Di Han Lu
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shuanghua Zhu
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ke Yang
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bingyuan Wu
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihong Chen
- Key Laboratory for Stem Cells and Tissue Engineering, Center for Stem Cell Biology and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Chaoquan Peng
- Department of Cardiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mei Hua Jiang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory for Stem Cells and Tissue Engineering, Center for Stem Cell Biology and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
105
|
Li J, Hu S, Zhu D, Huang K, Mei X, López de Juan Abad B, Cheng K. All Roads Lead to Rome (the Heart): Cell Retention and Outcomes From Various Delivery Routes of Cell Therapy Products to the Heart. J Am Heart Assoc 2021; 10:e020402. [PMID: 33821664 PMCID: PMC8174178 DOI: 10.1161/jaha.120.020402] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the past decades, numerous preclinical studies and several clinical trials have evidenced the feasibility of cell transplantation in treating heart diseases. Over the years, different delivery routes of cell therapy have emerged and broadened the width of the field. However, a common hurdle is shared by all current delivery routes: low cell retention. A myriad of studies confirm that cell retention plays a crucial role in the success of cell-mediated cardiac repair. It is important for any delivery route to maintain donor cells in the recipient heart for enough time to not only proliferate by themselves, but also to send paracrine signals to surrounding damaged heart cells and repair them. In this review, we first undertake an in-depth study of primary theories of cell loss, including low efficiency in cell injection, "washout" effects, and cell death, and then organize the literature from the past decade that focuses on cell transplantation to the heart using various cell delivery routes, including intracoronary injection, systemic intravenous injection, retrograde coronary venous injection, and intramyocardial injection. In addition to a recapitulation of these approaches, we also clearly evaluate their strengths and weaknesses. Furthermore, we conduct comparative research on the cell retention rate and functional outcomes of these delivery routes. Finally, we extend our discussion to state-of-the-art bioengineering techniques that enhance cell retention, as well as alternative delivery routes, such as intrapericardial delivery. A combination of these novel strategies and more accurate assessment methods will help to address the hurdle of low cell retention and boost the efficacy of cell transplantation to the heart.
Collapse
Affiliation(s)
- Junlang Li
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Shiqi Hu
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Dashuai Zhu
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Ke Huang
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Xuan Mei
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Blanca López de Juan Abad
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| | - Ke Cheng
- Department of Molecular Biomedical SciencesNorth Carolina State UniversityRaleighNC
- Joint Department of Biomedical EngineeringNorth Carolina State University and University of North Carolina at Chapel HillRaleighNC
| |
Collapse
|
106
|
Bolli R, Solankhi M, Tang XL, Kahlon A. Cell Therapy in Patients with Heart Failure: A Comprehensive Review and Emerging Concepts. Cardiovasc Res 2021; 118:951-976. [PMID: 33871588 PMCID: PMC8930075 DOI: 10.1093/cvr/cvab135] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 04/15/2021] [Indexed: 12/16/2022] Open
Abstract
This review summarizes the results of clinical trials of cell therapy in patients with heart failure (HF). In contrast to acute myocardial infarction (where results have been consistently negative for more than a decade), in the setting of HF the results of Phase I–II trials are encouraging, both in ischaemic and non-ischaemic cardiomyopathy. Several well-designed Phase II studies have met their primary endpoint and demonstrated an efficacy signal, which is remarkable considering that only one dose of cells was used. That an efficacy signal was seen 6–12 months after a single treatment provides a rationale for larger, rigorous trials. Importantly, no safety concerns have emerged. Amongst the various cell types tested, mesenchymal stromal cells derived from bone marrow (BM), umbilical cord, or adipose tissue show the greatest promise. In contrast, embryonic stem cells are not likely to become a clinical therapy. Unfractionated BM cells and cardiosphere-derived cells have been abandoned. The cell products used for HF will most likely be allogeneic. New approaches, such as repeated cell treatment and intravenous delivery, may revolutionize the field. As is the case for most new therapies, the development of cell therapies for HF has been slow, plagued by multifarious problems, and punctuated by many setbacks; at present, the utility of cell therapy in HF remains to be determined. What the field needs is rigorous, well-designed Phase III trials. The most important things to move forward are to keep an open mind, avoid preconceived notions, and let ourselves be guided by the evidence.
Collapse
Affiliation(s)
- Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY 40292
| | - Mitesh Solankhi
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY 40292
| | - Xiang-Liang Tang
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY 40292
| | - Arunpreet Kahlon
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY 40292
| |
Collapse
|
107
|
Cardiac Differentiation of Mesenchymal Stem Cells: Impact of Biological and Chemical Inducers. Stem Cell Rev Rep 2021; 17:1343-1361. [PMID: 33864233 DOI: 10.1007/s12015-021-10165-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2021] [Indexed: 02/07/2023]
Abstract
Cardiovascular disorders (CVDs) are the leading cause of global death, widely occurs due to irreparable loss of the functional cardiomyocytes. Stem cell-based therapeutic approaches, particularly the use of Mesenchymal Stem Cells (MSCs) is an emerging strategy to regenerate myocardium and thereby improving the cardiac function after myocardial infarction (MI). Most of the current approaches often employ the use of various biological and chemical factors as cues to trigger and modulate the differentiation of MSCs into the cardiac lineage. However, the recent advanced methods of using specific epigenetic modifiers and exosomes to manipulate the epigenome and molecular pathways of MSCs to modify the cardiac gene expression yield better profiled cardiomyocyte like cells in vitro. Hitherto, the role of cardiac specific inducers triggering cardiac differentiation at the cellular and molecular level is not well understood. Therefore, the current review highlights the impact and recent trends in employing biological and chemical inducers on cardiac differentiation of MSCs. Thereby, deciphering the interactions between the cellular microenvironment and the cardiac inducers will help us to understand cardiomyogenesis of MSCs. Additionally, the review also provides an insight on skeptical roles of the cell free biological factors and extracellular scaffold assisted mode for manipulation of native and transplanted stem cells towards translational cardiac research.
Collapse
|
108
|
Bolli R, Mitrani RD, Hare JM, Pepine CJ, Perin EC, Willerson JT, Traverse JH, Henry TD, Yang PC, Murphy MP, March KL, Schulman IH, Ikram S, Lee DP, O’Brien C, Lima JA, Ostovaneh MR, Ambale-Venkatesh B, Lewis G, Khan A, Bacallao K, Valasaki K, Longsomboon B, Gee AP, Richman S, Taylor DA, Lai D, Sayre SL, Bettencourt J, Vojvodic RW, Cohen ML, Simpson L, Aguilar D, Loghin C, Moyé L, Ebert RF, Davis BR, Simari RD. A Phase II study of autologous mesenchymal stromal cells and c-kit positive cardiac cells, alone or in combination, in patients with ischaemic heart failure: the CCTRN CONCERT-HF trial. Eur J Heart Fail 2021; 23:661-674. [PMID: 33811444 PMCID: PMC8357352 DOI: 10.1002/ejhf.2178] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/21/2022] Open
Abstract
AIMS CONCERT-HF is an NHLBI-sponsored, double-blind, placebo-controlled, Phase II trial designed to determine whether treatment with autologous bone marrow-derived mesenchymal stromal cells (MSCs) and c-kit positive cardiac cells (CPCs), given alone or in combination, is feasible, safe, and beneficial in patients with heart failure (HF) caused by ischaemic cardiomyopathy. METHODS AND RESULTS Patients were randomized (1:1:1:1) to transendocardial injection of MSCs combined with CPCs, MSCs alone, CPCs alone, or placebo, and followed for 12 months. Seven centres enrolled 125 participants with left ventricular ejection fraction of 28.6 ± 6.1% and scar size 19.4 ± 5.8%, in New York Heart Association class II or III. The proportion of major adverse cardiac events (MACE) was significantly decreased by CPCs alone (-22% vs. placebo, P = 0.043). Quality of life (Minnesota Living with Heart Failure Questionnaire score) was significantly improved by MSCs alone (P = 0.050) and MSCs + CPCs (P = 0.023) vs. placebo. Left ventricular ejection fraction, left ventricular volumes, scar size, 6-min walking distance, and peak oxygen consumption did not differ significantly among groups. CONCLUSIONS This is the first multicentre trial assessing CPCs and a combination of two cell types from different tissues in HF patients. The results show that treatment is safe and feasible. Even with maximal guideline-directed therapy, both CPCs and MSCs were associated with improved clinical outcomes (MACE and quality of life, respectively) in ischaemic HF without affecting left ventricular function or structure, suggesting possible systemic or paracrine cellular mechanisms. Combining MSCs with CPCs was associated with improvement in both these outcomes. These results suggest potential important beneficial effects of CPCs and MSCs and support further investigation in HF patients.
Collapse
Affiliation(s)
- Roberto Bolli
- University of Louisville, School of Medicine, Louisville, KY, USA
| | - Raul D. Mitrani
- University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Joshua M. Hare
- University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Carl J. Pepine
- University of Florida College of Medicine, Gainesville, FL, USA
| | - Emerson C. Perin
- Texas Heart Institute, CHI St. Luke’s Health Baylor College of Medicine Medical Center, Houston, TX, USA
| | - James T. Willerson
- Texas Heart Institute, CHI St. Luke’s Health Baylor College of Medicine Medical Center, Houston, TX, USA
| | - Jay H. Traverse
- Minneapolis Heart Institute Foundation at Abbott Northwestern Hospital, and University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Timothy D. Henry
- The Carl and Edyth Lindner Center for Research and Education, The Christ Hospital, Cincinnati, OH, USA
| | | | | | - Keith L. March
- University of Florida College of Medicine, Gainesville, FL, USA
| | | | - Sohail Ikram
- University of Louisville, School of Medicine, Louisville, KY, USA
| | - David P. Lee
- Stanford University School of Medicine, Stanford, CA, USA
| | - Connor O’Brien
- Stanford University School of Medicine, Stanford, CA, USA
| | - Joao A. Lima
- Johns Hopkins University, Cardiovascular Imaging, Baltimore, MD, USA
| | | | | | - Gregory Lewis
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Aisha Khan
- University of Miami, Miller School of Medicine, Interdisciplinary Stem Cell Institute, Miami, FL, USA
| | - Ketty Bacallao
- University of Miami, Miller School of Medicine, Interdisciplinary Stem Cell Institute, Miami, FL, USA
| | - Krystalenia Valasaki
- University of Miami, Miller School of Medicine, Interdisciplinary Stem Cell Institute, Miami, FL, USA
| | - Bangon Longsomboon
- University of Miami, Miller School of Medicine, Interdisciplinary Stem Cell Institute, Miami, FL, USA
| | - Adrian P. Gee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Sara Richman
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Doris A. Taylor
- Texas Heart Institute, CHI St. Luke’s Health Baylor College of Medicine Medical Center, Houston, TX, USA
| | - Dejian Lai
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Shelly L. Sayre
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Judy Bettencourt
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Rachel W. Vojvodic
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Michelle L. Cohen
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Lara Simpson
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - David Aguilar
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
- UTHealth University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - Catalin Loghin
- UTHealth University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, USA
| | - Lem Moyé
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | - Ray F. Ebert
- NIH, National Heart, Lung and Blood Institute, Bethesda, MD, USA
| | - Barry R. Davis
- UTHealth University of Texas Health Science Center at Houston School of Public Health, Houston, TX, USA
| | | | | |
Collapse
|
109
|
Povsic TJ, Gersh BJ. Stem Cells in Cardiovascular Diseases: 30,000-Foot View. Cells 2021; 10:cells10030600. [PMID: 33803227 PMCID: PMC8001267 DOI: 10.3390/cells10030600] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 12/15/2022] Open
Abstract
Stem cell and regenerative approaches that might rejuvenate the heart have immense intuitive appeal for the public and scientific communities. Hopes were fueled by initial findings from preclinical models that suggested that easily obtained bone marrow cells might have significant reparative capabilities; however, after initial encouraging pre-clinical and early clinical findings, the realities of clinical development have placed a damper on the field. Clinical trials were often designed to detect exceptionally large treatment effects with modest patient numbers with subsequent disappointing results. First generation approaches were likely overly simplistic and relied on a relatively primitive understanding of regenerative mechanisms and capabilities. Nonetheless, the field continues to move forward and novel cell derivatives, platforms, and cell/device combinations, coupled with a better understanding of the mechanisms that lead to regenerative capabilities in more primitive models and modifications in clinical trial design suggest a brighter future.
Collapse
Affiliation(s)
- Thomas J. Povsic
- Department of Medicine, and Duke Clinical Research Institute, Duke University, Durham, NC 27705, USA
- Correspondence:
| | - Bernard J. Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA;
| |
Collapse
|
110
|
Cardiac Cell Therapy: Insights into the Mechanisms of Tissue Repair. Int J Mol Sci 2021; 22:ijms22031201. [PMID: 33530466 PMCID: PMC7865339 DOI: 10.3390/ijms22031201] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Stem cell-based cardiac therapies have been extensively studied in recent years. However, the efficacy of cell delivery, engraftment, and differentiation post-transplant remain continuous challenges and represent opportunities to further refine our current strategies. Despite limited long-term cardiac retention, stem cell treatment leads to sustained cardiac benefit following myocardial infarction (MI). This review summarizes the current knowledge on stem cell based cardiac immunomodulation by highlighting the cellular and molecular mechanisms of different immune responses to mesenchymal stem cells (MSCs) and their secretory factors. This review also addresses the clinical evidence in the field.
Collapse
|
111
|
Liu Z, Naveed M, Baig MMFA, Mikrani R, Li C, Saeed M, Zhang Q, Farooq MA, Zubair HM, Xiaohui Z. Therapeutic approach for global myocardial injury using bone marrow-derived mesenchymal stem cells by cardiac support device in rats. Biomed Microdevices 2021; 23:5. [PMID: 33415464 DOI: 10.1007/s10544-020-00538-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2020] [Indexed: 02/07/2023]
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) have been considered a promising therapeutic approach to cardiovascular disease. This study intends to compare the effect of BMSCs through a standard active cardiac support device (ASD) and intravenous injection on global myocardial injury induced by isoproterenol. BMSCs were cultured in vitro, and the transplanted cells were labeled with a fluorescent dye CM-Dil. Isoproterenol (ISO) was injected into the rats; 2 weeks later, the labeled cells were transplanted into ISO-induced heart-jury rats through the tail vein or ASD device for 5 days. The rats were sacrificed on the first day, the third day, and the fifth day after transplantation to observe the distribution of cells in the myocardium by fluorescence microscopy. The hemodynamic indexes of the left ventricle were measured before sacrificing. H&E staining and Masson's trichrome staining were used to evaluate the cardiac histopathology. In the ASD groups, after 3 days of transplantation, there were a large number of BMSCs on the epicardial surface, and after 5 days of transplantation, BMSCs were widely distributed in the ventricular muscle. But in the intravenous injection group, there were no labeled-BMSCs distributed. In the ASD + BMSCs-three days treated group and ASD + BMSCs -five days-treated group, left ventricular systolic pressure (LVSP), the maximum rate of left ventricular pressure rise (+dP/dt), the maximum rate of left ventricular pressure decline (-dP/dt) increased compared with model group and intravenous injection group (P < 0.05). By giving BMSCs through ASD device, cells can rapidly and widely distribute in the myocardium and significantly improve heart function.
Collapse
Affiliation(s)
- Ziwei Liu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, School of Pharmacy, Nanjing, Jiangsu Province, 211198, People's Republic of China
| | - Muhammad Naveed
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, School of Pharmacy, Nanjing, Jiangsu Province, 211198, People's Republic of China.,School of Pharmacy, Nanjing Medical University, Nanjing, 211166, Jiangsu Province, China
| | - Mirza Muhammad Faran Ashraf Baig
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Reyaj Mikrani
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, School of Pharmacy, Nanjing, Jiangsu Province, 211198, People's Republic of China
| | - Cuican Li
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, School of Pharmacy, Nanjing, Jiangsu Province, 211198, People's Republic of China
| | - Muhammad Saeed
- Faculty of Animal Production and Technology, The Cholistan University of Veterinary and Animal Sciences, Bahawalpur, 6300, Pakistan
| | - Qin Zhang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, School of Pharmacy, Nanjing, Jiangsu Province, 211198, People's Republic of China
| | - Muhammad Asim Farooq
- Department of Pharmacy, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, People's Republic of China
| | | | - Zhou Xiaohui
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, School of Pharmacy, Nanjing, Jiangsu Province, 211198, People's Republic of China. .,Department of Heart Surgery, Nanjing Shuiximen Hospital, Nanjing, Jiangsu Province, 2110017, People's Republic of China. .,Department of Cardiothoracic Surgery, Zhongda Hospital affiliated with Southeast University, Nanjing, Jiangsu Province, 210017, People's Republic of China.
| |
Collapse
|
112
|
Saltzman RG, Jayaweera DT, Caceres LV, Tovar JA, Vidro-Casiano M, Karakeshishyan V, Soto J, Khan A, Mitrani RD, Schulman IH, Hare JM. Demographic representation in clinical trials for cell-based therapy. Contemp Clin Trials Commun 2021; 21:100702. [PMID: 33511300 PMCID: PMC7817424 DOI: 10.1016/j.conctc.2021.100702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/09/2020] [Accepted: 01/01/2021] [Indexed: 01/22/2023] Open
Abstract
Inclusion of women and minorities in clinical research is critical to fully assess the safety and efficacy of innovative therapies. With inadequate representation of demography, generalizability is impaired since pharmacokinetics and pharmacodynamics differ in these patient populations. This study was designed to analyze the voluntary participation rates of different demographic groups in cell-based therapy clinical trials conducted by the Interdisciplinary Stem Cell Institute (ISCI) at the University of Miami, Miller School of Medicine. ISCI conducted eight clinical trials between 2007 and 2017. The trials enrolled patients with ischemic and non-ischemic cardiomyopathy, idiopathic pulmonary fibrosis (IPF), aging-frailty, and Type-2 Diabetes. Participants received cell-based therapy (n = 218) or placebo (n = 33). Among the 251 participants, 29.5% were Hispanic and 20% were women. The proportion of individuals participating in each trial was compared to that of the respective disease populations attending University of Miami Health System clinics to calculate the participation to prevalence ratio (PPR). Distribution of women accurately reflected the population attending the University of Miami Health System in trials for dilated cardiomyopathy (DCM) and aging-frailty but was under-represented in others. Similarly, Hispanics and whites were accurately represented in three of the five disease fields, with Hispanics under-represented in frailty and diabetes, and whites over-represented in DCM and IPF. Black patients were accurately represented in the diabetes trial but were under-represented in all others. This study provides insight into challenges of achieving representative inclusion in research. Novel community engagement strategies are necessary to improve inclusion of women and under-represented minorities in clinical research of cell-based therapy.
Collapse
Affiliation(s)
- Russell G Saltzman
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dushyantha T Jayaweera
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lina V Caceres
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jairo A Tovar
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mayra Vidro-Casiano
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Vela Karakeshishyan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jeanette Soto
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Aisha Khan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Raul D Mitrani
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ivonne H Schulman
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
113
|
Abstract
Traumatic injuries are a leading cause of death and disability in both military and civilian populations. Given the complexity and diversity of traumatic injuries, novel and individualized treatment strategies are required to optimize outcomes. Cellular therapies have potential benefit for the treatment of acute or chronic injuries, and various cell-based pharmaceuticals are currently being tested in preclinical studies or in clinical trials. Cellular therapeutics may have the ability to complement existing therapies, especially in restoring organ function lost due to tissue disruption, prolonged hypoxia or inflammatory damage. In this article we highlight the current status and discuss future directions of cellular therapies for the treatment of traumatic injury. Both published research and ongoing clinical trials are discussed here.
Collapse
|
114
|
Berezin AE, Berezin AA. Stem-Cell-Based Cardiac Regeneration: Is There a Place For Optimism in the Future? Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
115
|
Evolution of Stem Cells in Cardio-Regenerative Therapy. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
116
|
Subbiah R, Sridharan D, Duairaj K, Rajan KS, Khan M, Garikipati VNS. Emerging Roles of Extracellular Vesicles Derived Non-Coding RNAs in the Cardiovascular System. Subcell Biochem 2021; 97:437-453. [PMID: 33779927 DOI: 10.1007/978-3-030-67171-6_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality all over the world. Emerging evidence emphasize the importance of extracellular vesicles (EVs) in the cell to cell communication in the cardiovascular system which is majorly mediated through non-coding RNA cargo. Advancement in sequencing technologies revealed a major proportion of human genome is composed of non-coding RNAs viz., miRNAs, lncRNAs, tRNAs, snoRNAs, piRNAs and rRNAs. However, our understanding of the role of ncRNAs-containing EVs in cardiovascular health and disease is still in its infancy. This book chapter provides a comprehensive update on our understanding on the role of EVs derived ncRNAs in the cardiovascular pathophysiology and their therapeutic potential.
Collapse
Affiliation(s)
- Ramasamy Subbiah
- Cardiac Hypertrophy Laboratory, Department of Molecular Biology, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | - Divya Sridharan
- Department of Emergency Medicine, Dorothy M. Davis Heart Lung and Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Karthika Duairaj
- Cardiac Hypertrophy Laboratory, Department of Molecular Biology, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | - K Shanmugha Rajan
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Mahmood Khan
- Department of Emergency Medicine, Dorothy M. Davis Heart Lung and Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Venkata Naga Srikanth Garikipati
- Department of Emergency Medicine, Dorothy M. Davis Heart Lung and Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
117
|
Rajendran RL, Jogalekar MP, Gangadaran P, Ahn BC. Noninvasive in vivo cell tracking using molecular imaging: A useful tool for developing mesenchymal stem cell-based cancer treatment. World J Stem Cells 2020; 12:1492-1510. [PMID: 33505597 PMCID: PMC7789123 DOI: 10.4252/wjsc.v12.i12.1492] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/05/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Mounting evidence has emphasized the potential of cell therapies in treating various diseases by restoring damaged tissues or replacing defective cells in the body. Cell therapies have become a strong therapeutic modality by applying noninvasive in vivo molecular imaging for examining complex cellular processes, understanding pathophysiological mechanisms of diseases, and evaluating the kinetics/dynamics of cell therapies. In particular, mesenchymal stem cells (MSCs) have shown promise in recent years as drug carriers for cancer treatment. They can also be labeled with different probes and tracked in vivo to assess the in vivo effect of administered cells, and to optimize therapy. The exact role of MSCs in oncologic diseases is not clear as MSCs have been shown to be involved in tumor progression and inhibition, and the exact interactions between MSCs and specific cancer microenvironments are not clear. In this review, a multitude of labeling approaches, imaging modalities, and the merits/demerits of each strategy are outlined. In addition, specific examples of the use of MSCs and in vivo imaging in cancer therapy are provided. Finally, present limitations and future outlooks in terms of the translation of different imaging approaches in clinics are discussed.
Collapse
Affiliation(s)
| | | | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
| | - Byeong-Cheol Ahn
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu 41944, South Korea
| |
Collapse
|
118
|
Human Mesenchymal Stem Cells: The Present Alternative for High-Incidence Diseases, Even SARS-Cov-2. Stem Cells Int 2020; 2020:8892189. [PMID: 33414832 PMCID: PMC7769649 DOI: 10.1155/2020/8892189] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/06/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022] Open
Abstract
Mesenchymal stem cells (MSCs), defined as plastic adherent cells with multipotent differentiation capacity in vitro, are an emerging and valuable tool to treat a plethora of diseases due to their therapeutic mechanisms such as their paracrine activity, mitochondrial and organelle transfer, and transfer of therapeutic molecules via exosomes. Nowadays, there are more than a thousand registered clinical trials related to MSC application around the world, highlighting MSC role on difficult-to-treat high-incidence diseases such as the current COVID-19, HIV infections, and autoimmune and metabolic diseases. Here, we summarize a general overview of MSCs and their therapeutic mechanisms; also, we discuss some of the novel clinical trial protocols and their results as well as a comparison between the number of registries, countries, and search portals.
Collapse
|
119
|
Ellison-Hughes GM, Colley L, O'Brien KA, Roberts KA, Agbaedeng TA, Ross MD. The Role of MSC Therapy in Attenuating the Damaging Effects of the Cytokine Storm Induced by COVID-19 on the Heart and Cardiovascular System. Front Cardiovasc Med 2020; 7:602183. [PMID: 33363221 PMCID: PMC7756089 DOI: 10.3389/fcvm.2020.602183] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/17/2020] [Indexed: 01/08/2023] Open
Abstract
The global pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that causes coronavirus disease 2019 (COVID-19) has led to 47 m infected cases and 1. 2 m (2.6%) deaths. A hallmark of more severe cases of SARS-CoV-2 in patients with acute respiratory distress syndrome (ARDS) appears to be a virally-induced over-activation or unregulated response of the immune system, termed a "cytokine storm," featuring elevated levels of pro-inflammatory cytokines such as IL-2, IL-6, IL-7, IL-22, CXCL10, and TNFα. Whilst the lungs are the primary site of infection for SARS-CoV-2, in more severe cases its effects can be detected in multiple organ systems. Indeed, many COVID-19 positive patients develop cardiovascular complications, such as myocardial injury, myocarditis, cardiac arrhythmia, and thromboembolism, which are associated with higher mortality. Drug and cell therapies targeting immunosuppression have been suggested to help combat the cytokine storm. In particular, mesenchymal stromal cells (MSCs), owing to their powerful immunomodulatory ability, have shown promise in early clinical studies to avoid, prevent or attenuate the cytokine storm. In this review, we will discuss the mechanistic underpinnings of the cytokine storm on the cardiovascular system, and how MSCs potentially attenuate the damage caused by the cytokine storm induced by COVID-19. We will also address how MSC transplantation could alleviate the long-term complications seen in some COVID-19 patients, such as improving tissue repair and regeneration.
Collapse
Affiliation(s)
- Georgina M. Ellison-Hughes
- Faculty of Life Sciences & Medicine, Centre for Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, King's College London Guy's Campus, London, United Kingdom
| | - Liam Colley
- School of Sport, Health, and Exercise Sciences, Bangor University, Bangor, United Kingdom
| | - Katie A. O'Brien
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Kirsty A. Roberts
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Thomas A. Agbaedeng
- Faculty of Health & Medical Sciences, Centre for Heart Rhythm Disorders, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Mark D. Ross
- School of Applied Sciences, Edinburgh Napier University, Edinburgh, United Kingdom
| |
Collapse
|
120
|
Pepine CJ, Raval AN. Cell Therapy Strategies With No Safety Concerns and Demonstrated Benefits Warrant Study. Circ J 2020; 84:2120-2121. [PMID: 33041288 DOI: 10.1253/circj.cj-19-1175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Carl J Pepine
- Division of Cardiovascular Medicine, University of Florida
| | - Amish N Raval
- Medicine and Biomedical Engineering, University of Wisconsin School of Medicine and Public Health
| |
Collapse
|
121
|
Point of care, bone marrow mononuclear cell therapy in ischemic heart failure patients personalized for cell potency: 12-month feasibility results from CardiAMP heart failure roll-in cohort. Int J Cardiol 2020; 326:131-138. [PMID: 33091520 DOI: 10.1016/j.ijcard.2020.10.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 09/24/2020] [Accepted: 10/15/2020] [Indexed: 01/09/2023]
Abstract
AIM Heart failure following myocardial infarction (MI) is a potentially lethal problem with a staggering incidence. The CardiAMP Heart Failure trial represents the first attempt to personalize marrow-derived cell-based therapy to individuals with cell characteristics associated with beneficial responses in prior trials. Before the initiation of the randomized pivotal trial, an open-label "roll-in cohort" was completed to ensure the feasibility of the protocol's procedures. METHODS Patients with chronic post-MI heart failure (NYHA class II-III) receiving stable, guideline-directed medical therapy with a left ventricular ejection fraction between 20 and 40% were eligible. Two weeks prior to treatment, a ~ 5 mL bone marrow aspiration was performed to examine "cell potency". On treatment day, a 60 mL bone marrow aspiration, bone marrow mononuclear cell (BM MNC) enrichment and transendocardial injection of 200 million BM MNC's was performed in a single, point of care encounter. Patients were then followed to assess clinical outcomes. RESULTS The cell potency small volume bone marrow aspirate, the 60 mL bone marrow aspirate, and transendocardial injections were well tolerated in 10 patients enrolled. There were no serious adverse events related to bone marrow aspiration or cell delivery. Improvement in 6-min walk distance was observed at 6 months (+47.8 m, P = 0.01) and trended to improvement at 12 months (+46.4, P = 0.06). Similarly, trends to improved NYHA heart failure functional class, quality of life, left ventricular ejection fraction and recruitment of previously akinetic left ventricular wall segments were observed. CONCLUSION All CardiAMP HF protocol procedures were feasible and well tolerated. Favorable functional, echo and quality of life trends suggest this approach may offer promise for patients with post MI heart failure. The randomized CardiAMP Heart Failure pivotal trial is underway to confirm the efficacy of this approach. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/ct2/show/NCT02438306.
Collapse
|
122
|
Yamamoto S, Ding N, Matsumoto SI, Hirabayashi H. Highly specific, quantitative polymerase chain reaction probe for the quantification of human cells in cynomolgus monkeys. Drug Metab Pharmacokinet 2020; 36:100359. [PMID: 33348238 DOI: 10.1016/j.dmpk.2020.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 09/17/2020] [Accepted: 09/23/2020] [Indexed: 10/23/2022]
Abstract
Quantification of human cells may be performed using quantitative polymerase chain reaction (qPCR). In preclinical studies, the human Alu sequence is widely used as biomarker for human DNA. However, because the Alu gene is shared by primates, its use is limited to non-primate studies. The biodistribution of human cells in primates is also necessary for translational studies. Therefore, we aimed to design a novel, human-specific primer/probe that enables the quantification of human cells in primates and other animal models. A novel primer/probe set was successfully designed based on highly repetitive LINE1 sequences. qPCR efficiency (94.95-99.21%) and linearity of calibration curves (r2 = 0.996-0.999) were confirmed in tissue homogenates of cynomolgus monkey. The lower limit of detection was 10 cells per 15-mg tissue sample, a sensitivity that is equivalent to existing Alu primers/probes. The set was also effective in other animal models such as mice, rabbits, pigs, and common marmosets. To our knowledge, this is the first study describing the successful design of a human-specific qPCR primer/probe for human cell quantification in various animals, including non-human primates, using LINE1 sequence. The excellent selectivity, sensitivity, and versatility of the LINE1 primers/probes make it a promising quantification tool in preclinical biodistribution studies.
Collapse
Affiliation(s)
- Syunsuke Yamamoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan.
| | - Ning Ding
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan.
| | - Shin-Ichi Matsumoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan.
| | - Hideki Hirabayashi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa, Japan.
| |
Collapse
|
123
|
Li C, Naveed M, Dar K, Liu Z, Baig MMFA, Lv R, Saeed M, Dingding C, Feng Y, Xiaohui Z. Therapeutic advances in cardiac targeted drug delivery: from theory to practice. J Drug Target 2020; 29:235-248. [PMID: 32933319 DOI: 10.1080/1061186x.2020.1818761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The most commonly used administration methods in clinics and life are oral administration, intravenous injection, and other systemic administration methods. Targeted administration must be an essential long-term development direction due to the limited availability and a high incidence of systemic side effects. Cardiovascular diseases (CVD) are the leading cause of death all over the world. Targeted drug delivery (TDD) methods with the heart as the target organ have developed rapidly and are diversified. This article reviews the research progress of various TDD methods around the world with a heart as the target organ. It is mainly divided into two parts: the targeting vector represented by nanoparticles and various TDD methods such as intracoronary injection, ventricular wall injection, pericardial injection, and implantable medical device therapy and put forward some suggestions on the development of targeting. Different TDD methods described in this paper have not been widely used in clinical practice, and some have not even completed preclinical studies. Targeted drug delivery still requires long-term efforts by many researchers to realize the true meaning of the heart. HIGHLIGHTS Targeted administration can achieve a better therapeutic effect and effectively reduce the occurrence of adverse reactions. Parenteral administration or medical device implantation can be used for targeted drug delivery. Combined with new dosage forms or new technologies, better-targeted therapy can be achieved. Clinical trials have confirmed the safety and effectiveness of several administration methods.
Collapse
Affiliation(s)
- Cuican Li
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Muhammad Naveed
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China.,School of Pharmacy, Nanjing Medical University, Nanjing, P. R. China
| | - Kashif Dar
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Medical University, Nanjing, P. R. China
| | - Ziwei Liu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Mirza Muhammad Faran Ashraf Baig
- State Key Laboratory of Analytical Chemistry for Life Sciences, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, P. R. China
| | - Rundong Lv
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Muhammad Saeed
- Faculty of Animal Production and Technology, The Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Chen Dingding
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Yu Feng
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| | - Zhou Xiaohui
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P. R. China.,Department of Heart Surgery, Nanjing Shuiximen Hospital, Nanjing, P. R. China.,Department of Cardiothoracic Surgery, Zhongda Hospital affiliated with Southeast University, Nanjing, P. R. China
| |
Collapse
|
124
|
Rodríguez-Fuentes DE, Fernández-Garza LE, Samia-Meza JA, Barrera-Barrera SA, Caplan AI, Barrera-Saldaña HA. Mesenchymal Stem Cells Current Clinical Applications: A Systematic Review. Arch Med Res 2020; 52:93-101. [PMID: 32977984 DOI: 10.1016/j.arcmed.2020.08.006] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/16/2020] [Accepted: 08/18/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Human Mesenchymal Stem Cells (hMSCs) are multipotent stem cells capable of renewing themselves and differentiation in vitro into different kinds of tissues. In vivo hMSCs are sources of trophic factors modulating the immune system and inducing intrinsic stem cells to repair damaged tissues. Currently, there are multiple clinical trials (CT) using hMSCs for therapeutic purposes in a large number of clinical settings. MATERIAL AND METHODS The search strategy on clinicaltrials.gov has focused on the key term "Mesenchymal Stem Cells", and the inclusion and exclusion criteria were separated into two stages. Stage 1, CT on phases 1-4: location, the field of application, phase, and status. For stage 2, CT that have published outcome results: field of application, treatment, intervention model, source, preparation methods, and results. RESULTS By July 2020, there were a total of 1,138 registered CT. Most studies belong to either phase 2 (61.0%) or phase 1 (30.8%); most of them focused in the fields of traumatology, neurology, cardiology, and immunology. Only 18 clinical trials had published results: the most common source of isolation was bone marrow; the treatment varied from 1-200 M hMSCs; all of them have similar preparation methods; all of them have positive results with no serious adverse effects. CONCLUSIONS There appears to be a broad potential for the clinical use of hMSCs with no reported serious adverse events. There are many trials in progress, their future results will help to explore the therapeutic potential of these promising cellular sources of medicinal signals.
Collapse
Affiliation(s)
| | - Luis E Fernández-Garza
- Innbiogem SC en el Laboratorio Nacional de Servicios Especializados de Investigación, Desarrollo e Innovación en Medicamentos Químicos y Biotecnológicos CONACyT, Monterrey, NL, México
| | - John A Samia-Meza
- Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Monterrey, NL, México
| | | | - Arnold I Caplan
- Skeletal Research Center, Department of Biology Case Western Reserve University, Cleveland, Ohio, USA
| | - Hugo A Barrera-Saldaña
- Innbiogem SC en el Laboratorio Nacional de Servicios Especializados de Investigación, Desarrollo e Innovación en Medicamentos Químicos y Biotecnológicos CONACyT, Monterrey, NL, México.
| |
Collapse
|
125
|
Selvakumar D, Clayton ZE, Chong JJH. Robust Cardiac Regeneration: Fulfilling the Promise of Cardiac Cell Therapy. Clin Ther 2020; 42:1857-1879. [PMID: 32943195 DOI: 10.1016/j.clinthera.2020.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE We review the history of cardiac cell therapy, highlighting lessons learned from initial adult stem cell (ASC) clinical trials. We present pluripotent stem cell-derived cardiomyocytes (PSC-CMs) as a leading candidate for robust regeneration of infarcted myocardium but identify several issues that must be addressed before successful clinical translation. METHODS We conducted an unstructured literature review of PubMed-listed articles, selecting the most comprehensive and relevant research articles, review articles, clinical trials, and basic or translation articles in the field of cardiac cell therapy. Articles were identified using the search terms adult stem cells, pluripotent stem cells, cardiac stem cell, and cardiac regeneration or from references of relevant articles, Articles were prioritized and selected based on their impact, originality, or potential clinical applicability. FINDINGS Since its inception, the ASC therapy field has been troubled by conflicting preclinical data, academic controversies, and inconsistent trial designs. These issues have damaged perceptions of cardiac cell therapy among investors, the academic community, health care professionals, and, importantly, patients. In hindsight, the key issue underpinning these problems was the inability of these cell types to differentiate directly into genuine cardiomyocytes, rendering them unable to replace damaged myocardium. Despite this, beneficial effects through indirect paracrine or immunomodulatory effects remain possible and continue to be investigated. However, in preclinical models, PSC-CMs have robustly remuscularized infarcted myocardium with functional, force-generating cardiomyocytes. Hence, PSC-CMs have now emerged as a leading candidate for cardiac regeneration, and unpublished reports of first-in-human delivery of these cells have recently surfaced. However, the cardiac cell therapy field's history should serve as a cautionary tale, and we identify several translational hurdles that still remain. Preclinical solutions to issues such as arrhythmogenicity, immunogenicity, and poor engraftment rates are needed, and next-generation clinical trials must draw on robust knowledge of mechanistic principles of the therapy. IMPLICATIONS The clinical transplantation of functional stem cell-derived heart tissue with seamless integration into native myocardium is a lofty goal. However, considerable advances have been made during the past 2 decades. Currently, PSC-CMs appear to be the best prospect to reach this goal, but several hurdles remain. The history of adult stem cell trials has taught us that shortcuts cannot be taken without dire consequences, and it is essential that progress not be hurried and that a worldwide, cross-disciplinary approach be used to ensure safe and effective clinical translation.
Collapse
Affiliation(s)
- Dinesh Selvakumar
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Zoe E Clayton
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - James J H Chong
- Centre for Heart Research, The Westmead Institute for Medical Research, The University of Sydney, Sydney, New South Wales, Australia; Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia.
| |
Collapse
|
126
|
Wang S, Huang S, Johnson S, Rosin V, Lee J, Colomb E, Witt R, Jaworski A, Weiss SJ, Si M. Tissue-specific angiogenic and invasive properties of human neonatal thymus and bone MSCs: Role of SLIT3-ROBO1. Stem Cells Transl Med 2020; 9:1102-1113. [PMID: 32470195 PMCID: PMC7445019 DOI: 10.1002/sctm.19-0448] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/05/2020] [Accepted: 05/03/2020] [Indexed: 12/13/2022] Open
Abstract
Although mesenchymal stem/stromal cells (MSCs) are being explored in numerous clinical trials as proangiogenic and proregenerative agents, the influence of tissue origin on the therapeutic qualities of these cells is poorly understood. Complicating the functional comparison of different types of MSCs are the confounding effects of donor age, genetic background, and health status of the donor. Leveraging a clinical setting where MSCs can be simultaneously isolated from discarded but healthy bone and thymus tissues from the same neonatal patients, thereby controlling for these confounding factors, we performed an in vitro and in vivo paired comparison of these cells. We found that both neonatal thymus (nt)MSCs and neonatal bone (nb)MSCs expressed different pericytic surface marker profiles. Further, ntMSCs were more potent in promoting angiogenesis in vitro and in vivo and they were also more motile and efficient at invading ECM in vitro. These functional differences were in part mediated by an increased ntMSC expression of SLIT3, a factor known to activate endothelial cells. Further, we discovered that SLIT3 stimulated MSC motility and fibrin gel invasion via ROBO1 in an autocrine fashion. Consistent with our findings in human MSCs, we found that SLIT3 and ROBO1 were expressed in the perivascular cells of the neonatal murine thymus gland and that global SLIT3 or ROBO1 deficiency resulted in decreased neonatal murine thymus gland vascular density. In conclusion, ntMSCs possess increased proangiogenic and invasive behaviors, which are in part mediated by the paracrine and autocrine effects of SLIT3.
Collapse
Affiliation(s)
- Shuyun Wang
- Department of Cardiac Surgery, Section of Pediatric Cardiovascular SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Shan Huang
- Department of Cardiac Surgery, Section of Pediatric Cardiovascular SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Sean Johnson
- Department of Cardiac Surgery, Section of Pediatric Cardiovascular SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Vadim Rosin
- Department of Cardiac Surgery, Section of Pediatric Cardiovascular SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Jeffrey Lee
- Department of Cardiac Surgery, Section of Pediatric Cardiovascular SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Eric Colomb
- Department of Cardiac Surgery, Section of Pediatric Cardiovascular SurgeryUniversity of MichiganAnn ArborMichiganUSA
| | - Russell Witt
- Department of General SurgeryBrigham and Women's HospitalMassachusettsUSA
| | | | - Stephen J. Weiss
- Department of Internal MedicineUniversity of MichiganAnn ArborMichiganUSA
| | - Ming‐Sing Si
- Department of Cardiac Surgery, Section of Pediatric Cardiovascular SurgeryUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
127
|
Qu W, Wang Z, Hare JM, Bu G, Mallea JM, Pascual JM, Caplan AI, Kurtzberg J, Zubair AC, Kubrova E, Engelberg‐Cook E, Nayfeh T, Shah VP, Hill JC, Wolf ME, Prokop LJ, Murad MH, Sanfilippo FP. Cell-based therapy to reduce mortality from COVID-19: Systematic review and meta-analysis of human studies on acute respiratory distress syndrome. Stem Cells Transl Med 2020; 9:1007-1022. [PMID: 32472653 PMCID: PMC7300743 DOI: 10.1002/sctm.20-0146] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/27/2020] [Accepted: 05/03/2020] [Indexed: 12/17/2022] Open
Abstract
Severe cases of COVID-19 infection, often leading to death, have been associated with variants of acute respiratory distress syndrome (ARDS). Cell therapy with mesenchymal stromal cells (MSCs) is a potential treatment for COVID-19 ARDS based on preclinical and clinical studies supporting the concept that MSCs modulate the inflammatory and remodeling processes and restore alveolo-capillary barriers. The authors performed a systematic literature review and random-effects meta-analysis to determine the potential value of MSC therapy for treating COVID-19-infected patients with ARDS. Publications in all languages from 1990 to March 31, 2020 were reviewed, yielding 2691 studies, of which nine were included. MSCs were intravenously or intratracheally administered in 117 participants, who were followed for 14 days to 5 years. All MSCs were allogeneic from bone marrow, umbilical cord, menstrual blood, adipose tissue, or unreported sources. Combined mortality showed a favorable trend but did not reach statistical significance. No related serious adverse events were reported and mild adverse events resolved spontaneously. A trend was found of improved radiographic findings, pulmonary function (lung compliance, tidal volumes, PaO2 /FiO2 ratio, alveolo-capillary injury), and inflammatory biomarker levels. No comparisons were made between MSCs of different sources.
Collapse
Affiliation(s)
- Wenchun Qu
- Department of Pain MedicineMayo ClinicJacksonvilleFloridaUSA
- Center for Regenerative MedicineMayo ClinicJacksonvilleFloridaUSA
| | - Zhen Wang
- Evidence‐Based Practice CenterMayo ClinicRochesterMinnesotaUSA
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery Mayo ClinicRochesterMinnesotaUSA
| | - Joshua M. Hare
- Interdisciplinary Stem Cell Institute and Cardiology Division, Department of MedicineUniversity of Miami, Miller School of MedicineMiamiFloridaUSA
| | - Guojun Bu
- Center for Regenerative MedicineMayo ClinicJacksonvilleFloridaUSA
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | - Jorge M. Mallea
- Division of Pulmonary, Allergy and Sleep Medicine, Department of MedicineMayo ClinicJacksonvilleFloridaUSA
| | - Jorge M. Pascual
- Division of Pulmonary, Allergy and Sleep Medicine, Department of MedicineMayo ClinicJacksonvilleFloridaUSA
| | - Arnold I. Caplan
- Skeletal Research Center, Biology DepartmentCase Western Reserve UniversityClevelandOhioUSA
| | - Joanne Kurtzberg
- Marcus Center for Cellular CuresDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Abba C. Zubair
- Center for Regenerative MedicineMayo ClinicJacksonvilleFloridaUSA
- Transfusion Medicine and Stem Cell Therapy, Department of Laboratory Medicine and PathologyMayo ClinicJacksonvilleFloridaUSA
| | - Eva Kubrova
- Department of Physical Medicine and Rehabilitation, Department of Orthopedic SurgeryMayo ClinicRochesterMinnesotaUSA
| | | | - Tarek Nayfeh
- Evidence‐Based Practice CenterMayo ClinicRochesterMinnesotaUSA
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery Mayo ClinicRochesterMinnesotaUSA
| | - Vishal P. Shah
- Department of Preventative, Occupational, and Aerospace MedicineMayo ClinicRochesterMinnesotaUSA
| | - James C. Hill
- Department of Preventative, Occupational, and Aerospace MedicineMayo ClinicRochesterMinnesotaUSA
| | - Michael E. Wolf
- Department of Preventative, Occupational, and Aerospace MedicineMayo ClinicRochesterMinnesotaUSA
| | | | - M. Hassan Murad
- Evidence‐Based Practice CenterMayo ClinicRochesterMinnesotaUSA
- Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery Mayo ClinicRochesterMinnesotaUSA
- Department of Preventative, Occupational, and Aerospace MedicineMayo ClinicRochesterMinnesotaUSA
| | - Fred P. Sanfilippo
- Department of Pathology and Laboratory Medicine, Department of Health Policy and ManagementRollins School of Public Health, Emory University, The Marcus FoundationAtlantaGeorgiaUSA
| |
Collapse
|
128
|
Lopes GM, Grudzinski PB, Beyer Nardi N, Leguisamo NM. Cell Therapy Improves Cardiac Function in Anthracycline-Induced Cardiomyopathy Preclinical Models: A Systematic Review and Meta-Analysis. Stem Cells Dev 2020; 29:1247-1265. [PMID: 32741268 DOI: 10.1089/scd.2020.0044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Although anthracycline (ANT)-based treatment strongly contributes to cancer survivorship, the use of these agents is limited by the risk of cardiotoxicity. For those patients who evolve to heart failure, myocardial regenerative approaches are of particular interest, and a growing body of preclinical studies has been investigating the use of cell therapy for ANT-induced cardiomyopathy (AIC). However, since animal models and modalities of cell therapy are highly heterogeneous between studies, the efficacy of cell therapy for AIC is not clear. Thus, we conducted a systematic review and meta-analysis of experimental studies reporting the use of cell therapy with mesenchymal stromal cells (MSC) or bone marrow mononuclear cells (BMMNC) in animal models of AIC with regard to global cardiac function. The Medline, EMBASE, and Web of Science databases were searched from inception to November 2019. Two reviewers independently extracted data on study quality and the results of left ventricular ejection fraction (LVEF) and fractional shortening (FS) obtained by echocardiography. The quality of outcomes was assessed using the Cochrane, Collaborative Approach to Meta-Analysis and Review of Animal Data from Experimental Studies (CAMARADES), and SYRCLE bias risk tools. Pooled random-effects modeling was used to calculate pooled mean differences (MD) and 95% confidence intervals (CIs). Twenty-two studies comprising 381 small animals (rabbits and rodents) were included. A pooled meta-analysis of all treatments showed that cell therapy increased LVEF by 9.87% (95% CI 7.25-12.50, P < 0.00001) and FS by 7.80% (95% CI 5.68-9.92, P < 0.00001) in small animals with AIC. Cell therapy with MSC/BMMNC is effective to mitigate the deleterious effects of ANT on cardiac function in preclinical models. Nevertheless, due to the small number of studies and considerable heterogeneity, future translational studies must be designed to diminish between-study discrepancies and increase similarity to the clinical landscape.
Collapse
Affiliation(s)
- Gabriela Maciel Lopes
- Institute of Cardiology of Rio Grande do Sul/University Foundation of Cardiology (IC/FUC), Porto Alegre, Brazil.,Graduate Program in Health Sciences (Cardiology), University Foundation of Cardiology, Porto Alegre, Brazil
| | - Patrícia Bencke Grudzinski
- Institute of Cardiology of Rio Grande do Sul/University Foundation of Cardiology (IC/FUC), Porto Alegre, Brazil
| | - Nance Beyer Nardi
- Institute of Cardiology of Rio Grande do Sul/University Foundation of Cardiology (IC/FUC), Porto Alegre, Brazil.,Graduate Program in Health Sciences (Cardiology), University Foundation of Cardiology, Porto Alegre, Brazil
| | - Natalia Motta Leguisamo
- Institute of Cardiology of Rio Grande do Sul/University Foundation of Cardiology (IC/FUC), Porto Alegre, Brazil.,Graduate Program in Health Sciences (Cardiology), University Foundation of Cardiology, Porto Alegre, Brazil
| |
Collapse
|
129
|
Mesenchymal Stem/Progenitor Cells: The Prospect of Human Clinical Translation. Stem Cells Int 2020; 2020:8837654. [PMID: 33953753 PMCID: PMC8063852 DOI: 10.1155/2020/8837654] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/19/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/progenitor cells (MSCs) are key players in regenerative medicine, relying principally on their differentiation/regeneration potential, immunomodulatory properties, paracrine effects, and potent homing ability with minimal if any ethical concerns. Even though multiple preclinical and clinical studies have demonstrated remarkable properties for MSCs, the clinical applicability of MSC-based therapies is still questionable. Several challenges exist that critically hinder a successful clinical translation of MSC-based therapies, including but not limited to heterogeneity of their populations, variability in their quality and quantity, donor-related factors, discrepancies in protocols for isolation, in vitro expansion and premodification, and variability in methods of cell delivery, dosing, and cell homing. Alterations of MSC viability, proliferation, properties, and/or function are also affected by various drugs and chemicals. Moreover, significant safety concerns exist due to possible teratogenic/neoplastic potential and transmission of infectious diseases. Through the current review, we aim to highlight the major challenges facing MSCs' human clinical translation and shed light on the undergoing strategies to overcome them.
Collapse
|
130
|
Wu X, Jiang J, Gu Z, Zhang J, Chen Y, Liu X. Mesenchymal stromal cell therapies: immunomodulatory properties and clinical progress. Stem Cell Res Ther 2020; 11:345. [PMID: 32771052 PMCID: PMC7414268 DOI: 10.1186/s13287-020-01855-9] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/09/2020] [Accepted: 07/27/2020] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are a subset of heterogeneous non-hematopoietic fibroblast-like cells that can differentiate into cells of multiple lineages, such as chondrocytes, osteoblasts, adipocytes, myoblasts, and others. These multipotent MSCs can be found in nearly all tissues but mostly located in perivascular niches, playing a significant role in tissue repair and regeneration. Additionally, MSCs interact with immune cells both in innate and adaptive immune systems, modulating immune responses and enabling immunosuppression and tolerance induction. Understanding the biology of MSCs and their roles in clinical treatment is crucial for developing MSC-based cellular therapy for a variety of pathological conditions. Here, we review the progress in the study on the mechanisms underlying the immunomodulatory and regenerative effects of MSCs; update the medical translation of MSCs, focusing on the registration trials leading to regulatory approvals; and discuss how to improve therapeutic efficacy and safety of MSC applications for future.
Collapse
Affiliation(s)
- Xiaomo Wu
- Dermatology Institute of Fuzhou, Dermatology Hospital of Fuzhou, Xihong Road 243, Fuzhou, 350025, China.,Department of Biomedicine, University of Basel, Klingelbergstr 70, CH-4056, Basel, Switzerland
| | - Ju Jiang
- Dermatology Institute of Fuzhou, Dermatology Hospital of Fuzhou, Xihong Road 243, Fuzhou, 350025, China
| | - Zhongkai Gu
- The Institute of Biomedical Sciences, Fudan University, Mingdao Building, Dongan Road 131, Shanghai, 200032, China
| | - Jinyan Zhang
- Dermatology Institute of Fuzhou, Dermatology Hospital of Fuzhou, Xihong Road 243, Fuzhou, 350025, China
| | - Yang Chen
- Dermatology Institute of Fuzhou, Dermatology Hospital of Fuzhou, Xihong Road 243, Fuzhou, 350025, China.
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Xihong Road 312, Fuzhou, 350025, China.
| |
Collapse
|
131
|
Gao L, Yi M, Xing M, Li H, Zhou Y, Xu Q, Zhang Z, Wen Z, Chang J. In situ activated mesenchymal stem cells (MSCs) by bioactive hydrogels for myocardial infarction treatment. J Mater Chem B 2020; 8:7713-7722. [PMID: 32724972 DOI: 10.1039/d0tb01320j] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Stem-cell therapy has been proved as a promising strategy for myocardial infarction (MI) treatment. However, the therapeutic efficacy is mainly limited by the cellular activity of transplanted mesenchymal stem cells (MSCs). In this study, a novel bioglass (BG)/γ-polyglutamic acid (γ-PGA)/chitosan (CS) hydrogel was obtained by in situ adding BG to stimulate the imine bond formation. And the effect of the composite hydrogel on MI therapeutic efficacy was evaluated in a rat acute myocardial infarction (AMI) model in vivo and the possible mechanism of the BG/γ-PGA/CS hydrogel for the stimulation of the intercellular interaction between MSCs and cardiomyocytes (CMs) was explored by a MSC and CM co-culture experiment in vitro. The implantation of the MSC loaded BG/γ-PGA/CS composite hydrogel in the mice AMI model showed a significant improvement in the therapeutic efficacy with improved cardiac function, attenuation of heart remodeling, reduced cardiomyocyte apoptosis and accelerated vascularization. The in vitro cell experiments demonstrated that the BG/γ-PGA/CS hydrogel activated the intercellular interaction between MSCs and CMs, which resulted in reduced cell apoptosis and enhanced angiogenesis. Silicate based bioactive hydrogels activated MSCs and cell-cell interactions in cardiac tissue after AMI and significantly enhanced the efficacy, which suggests that this bioactive hydrogel based approach is an effective way to enhance stem-cell therapy.
Collapse
Affiliation(s)
- Long Gao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Shanghai, 200050, China
| | | | | | | | | | | | | | | | | |
Collapse
|
132
|
Raval AN, Pepine CJ. Clinical Safety Profile of Transendocardial Catheter Injection Systems: A Plea for Uniform Reporting. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2020; 22:100-108. [PMID: 32651159 DOI: 10.1016/j.carrev.2020.06.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES The aim of this study was to characterize the clinical safety profile of transendocardial injection catheters (TIC) reported in the published literature. BACKGROUND Transendocardial delivery is a minimally invasive approach to deliver potential therapeutic agents directly into the myocardium. The rate of adverse events across TIC is uncertain. METHODS A systematic search was performed for trial publications using TIC. Procedure-associated adverse event data were abstracted, pooled and compared across catheters for active treatment and placebo injected patients. The transendocardial injection associated serious adverse events (TEI-SAE) was defined as the composite of death, myocardial infarction, stroke or transient ischemic attack within 30 days and cardiac perforation causing death or requiring evacuation, serious intraprocedural arrhythmias and serious coronary artery or peripheral vascular complications. RESULTS The search identified 4 TIC systems: a helical needle (HN), an electro-anatomically tracked straight needle (EAM-SN), a straight needle without tracking elements (SN), and a curved needle (CN). Of 1799 patients who underwent transendocardial injections, the combined TEI-SAE was 3.4% across all catheters, and 1.1%, 3.3%, 7.1%, and 8.3% for HN, EAM-SN, SN and CN, respectively. However, TIC procedure duration and post procedural cardiac biomarker levels were reported in only 24% and 36% of published trials, respectively. CONCLUSIONS Transendocardial injection is associated with varied TEI-SAE but the data are very limited. The HN catheter appeared to be associated with lower TEI-SAE, versus other catheters. Procedure duration and post procedure cardiac biomarker levels were under-reported. Clearly, standardized, procedure-related event reporting for trials involving transcatheter delivery would improve our understanding of complications across different systems.
Collapse
Affiliation(s)
- Amish N Raval
- Department of Medicine and Biomedical Engineering, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - Carl J Pepine
- Division of Cardiovascular Medicine, University of Florida, Gainsville, FL, USA
| |
Collapse
|
133
|
Ng NN, Thakor AS. Locoregional delivery of stem cell-based therapies. Sci Transl Med 2020; 12:eaba4564. [PMID: 32522806 DOI: 10.1126/scitranslmed.aba4564] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/24/2020] [Accepted: 05/20/2020] [Indexed: 12/13/2022]
Abstract
Interventional regenerative medicine (IRM) uses image-guided, minimally invasive procedures for the targeted delivery of stem cell-based therapies to regenerate, replace, or repair damaged organs. Although many cellular therapies have shown promise in the preclinical setting, clinical results have been suboptimal. Most intravenously delivered cells become trapped in the lungs and reticuloendothelial system, resulting in little therapy reaching target tissues. IRM aims to increase the efficacy of cell-based therapies by locoregional stem cell delivery via endovascular, endoluminal, or direct injection into tissues. This review highlights routes of delivery, disease states, and mechanisms of action involved in the targeted delivery of stem cells.
Collapse
Affiliation(s)
- Nathan Norton Ng
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Avnesh Sinh Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Stanford, CA 94304, USA.
| |
Collapse
|
134
|
Abstract
Stem cell therapy offers a breakthrough opportunity for the improvement of ischemic heart diseases. Numerous clinical trials and meta-analyses appear to confirm its positive but variable effects on heart function. Whereas these trials widely differed in design, cell type, source, and doses reinjected, cell injection route and timing, and type of cardiac disease, crucial key factors that may favour the success of cell therapy emerge from the review of their data. Various types of cell have been delivered. Injection of myoblasts does not improve heart function and is often responsible for severe ventricular arrythmia occurrence. Using bone marrow mononuclear cells is a misconception, as they are not stem cells but mainly a mix of various cells of hematopoietic lineages and stromal cells, only containing very low numbers of cells that have stem cell-like features; this likely explain the neutral results or at best the modest improvement in heart function reported after their injection. The true existence of cardiac stem cells now appears to be highly discredited, at least in adults. Mesenchymal stem cells do not repair the damaged myocardial tissue but attenuate post-infarction remodelling and contribute to revascularization of the hibernated zone surrounding the scar. CD34+ stem cells - likely issued from pluripotent very small embryonic-like (VSEL) stem cells - emerge as the most convincing cell type, inducing structural and functional repair of the ischemic myocardial area, providing they can be delivered in large amounts via intra-myocardial rather than intra-coronary injection, and preferentially after myocardial infarct rather than chronic heart failure.
Collapse
Affiliation(s)
- Philippe Hénon
- CellProthera SAS and Institut de Recherche en Hématologie et Transplantation, CellProthera SAS 12 rue du Parc, 68100, Mulhouse, France.
| |
Collapse
|
135
|
Wang J, Chen Z, Dai Q, Zhao J, Wei Z, Hu J, Sun X, Xie J, Xu B. Intravenously delivered mesenchymal stem cells prevent microvascular obstruction formation after myocardial ischemia/reperfusion injury. Basic Res Cardiol 2020; 115:40. [PMID: 32451935 DOI: 10.1007/s00395-020-0800-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/15/2020] [Indexed: 12/11/2022]
Abstract
Microvascular obstruction (MVO) after primary percutaneous coronary intervention (pPCI) is identified as an independent risk factor for poor prognosis in patients with acute myocardial infarction (AMI). The inflammatory response induced by ischemia and reperfusion (I/R) injury is considered one of the main mechanisms of MVO. Mesenchymal stem cells (MSCs) are a unique stromal cell type that confers an immunomodulatory effect in cardiac disease. The present study aimed to investigate whether immediate intravenous delivery of MSCs could be used as a potential therapeutic method to attenuate MVO formation. A cardiac catheterization-induced porcine model of myocardial I/R injury was established, and allograft MSCs were immediately delivered intravenously. Cardiac magnetic resonance (CMR) imaging was performed on days 2 and 7 after the operation to determine the infarct area, MVO, and cardiac function. The pigs with allograft MSCs showed decreased MVO and infarct size, as well as an improved left ventricular ejection fraction (LVEF). Histological analysis revealed decreased myocyte area, fibrosis, and inflammatory cell infiltration in the peri-infarct zone of pigs with allograft MSCs. Moreover, the concentrations of interleukin-1β (IL-1β), interleukin-6 (IL-6) and C-reactive protein (CRP) in the serum were reduced in the allograft MSC group compared to the control group. Flow cytometry indicated decreased natural killer (NK) cells in the peripheral blood and ischemic heart tissue in the pigs with allograft MSCs. In summary, allograft MSCs delivered intravenously and immediately after myocardial I/R injury can attenuate MVO formation in a porcine model through a decline in the number of NK cells in the myocardium.
Collapse
Affiliation(s)
- Junzhuo Wang
- Department of Cardiology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Ziwei Chen
- Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Qing Dai
- Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Jinxuan Zhao
- Department of Cardiology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Zilun Wei
- Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Jiaxin Hu
- Department of Cardiology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Xuan Sun
- Department of Cardiology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China
| | - Jun Xie
- Department of Cardiology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China.
| | - Biao Xu
- Department of Cardiology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, China. .,Department of Cardiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
136
|
Turner D, Rieger AC, Balkan W, Hare JM. Clinical-based Cell Therapies for Heart Disease-Current and Future State. Rambam Maimonides Med J 2020; 11:RMMJ.10401. [PMID: 32374254 PMCID: PMC7202446 DOI: 10.5041/rmmj.10401] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Patients have an ongoing unmet need for effective therapies that reverse the cellular and functional damage associated with heart damage and disease. The discovery that ~1%-2% of adult cardiomyocytes turn over per year provided the impetus for treatments that stimulate endogenous repair mechanisms that augment this rate. Preclinical and clinical studies provide evidence that cell-based therapy meets these therapeutic criteria. Recent and ongoing studies are focused on determining which cell type(s) works best for specific patient population(s) and the mechanism(s) by which these cells promote repair. Here we review clinical and preclinical stem cell studies and anticipate future directions of regenerative medicine for heart disease.
Collapse
Affiliation(s)
- Darren Turner
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Angela C. Rieger
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Joshua M. Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
137
|
Liew LC, Ho BX, Soh BS. Mending a broken heart: current strategies and limitations of cell-based therapy. Stem Cell Res Ther 2020; 11:138. [PMID: 32216837 PMCID: PMC7098097 DOI: 10.1186/s13287-020-01648-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/02/2020] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
The versatility of pluripotent stem cells, attributable to their unlimited self-renewal capacity and plasticity, has sparked a considerable interest for potential application in regenerative medicine. Over the past decade, the concept of replenishing the lost cardiomyocytes, the crux of the matter in ischemic heart disease, with pluripotent stem cell-derived cardiomyocytes (PSC-CM) has been validated with promising pre-clinical results. Nevertheless, clinical translation was hemmed in by limitations such as immature cardiac properties, long-term engraftment, graft-associated arrhythmias, immunogenicity, and risk of tumorigenicity. The continuous progress of stem cell-based cardiac therapy, incorporated with tissue engineering strategies and delivery of cardio-protective exosomes, provides an optimistic outlook on the development of curative treatment for heart failure. This review provides an overview and current status of stem cell-based therapy for heart regeneration, with particular focus on the use of PSC-CM. In addition, we also highlight the associated challenges in clinical application and discuss the potential strategies in developing successful cardiac-regenerative therapy.
Collapse
Affiliation(s)
- Lee Chuen Liew
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Beatrice Xuan Ho
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| | - Boon-Seng Soh
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore. .,Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore. .,Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
138
|
Seetharaman R, Mahmood A, Kshatriya P, Patel D, Srivastava A. An Overview on Stem Cells in Tissue Regeneration. Curr Pharm Des 2020; 25:2086-2098. [PMID: 31298159 DOI: 10.2174/1381612825666190705211705] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Deteriorations in tissues and decline in organ functions, due to chronic diseases or with advancing age or sometimes due to infections or injuries, can severely compromise the quality of life of an individual. Regenerative medicine, a field of medical research focuses on replacing non-functional or dead cells or repairing or regenerating tissues and organs to restore normal functions of an impaired organ. Approaches used in regenerative therapy for achieving the objective employ a number of means which include soluble biomolecules, stem cell transplants, tissue engineering, gene therapy and reprogramming of cells according to target tissue types. Stem cells transplant and tissue regeneration methods for treating various diseases have rapidly grown in usage over the past decades or so. There are different types of stem cells such as mesenchymal, hematopoietic, embryonic, mammary, intestinal, endothelial, neural, olfactory, neural crest, testicular and induced pluripotent stem cells. METHODS This review covers the recent advances in tissue regeneration and highlights the application of stem cell transplants in treating many life-threatening diseases or in improving quality of life. RESULTS Remarkable progress in stem cell research has established that the cell-based therapy could be an option for treating diseases which could not be cured by conventional medical means till recent. Stem cells play major roles in regenerative medicine with its exceptional characteristics of self-renewal capacity and potential to differentiate into almost all types of cells of a body. CONCLUSION Vast number of reports on preclinical and clinical application of stem cells revealed its vital role in disease management and many pharmacological industries around the globe working to achieve effective stem cell based products.
Collapse
Affiliation(s)
| | | | | | | | - Anand Srivastava
- Global Institute of Stem Cell Therapy and Research, 4660 La Jolla Village Drive, San Diego, CA 92122, United States
| |
Collapse
|
139
|
Liu Z, Mikrani R, Zubair HM, Taleb A, Naveed M, Baig MMFA, Zhang Q, Li C, Habib M, Cui X, Sembatya KR, Lei H, Zhou X. Systemic and local delivery of mesenchymal stem cells for heart renovation: Challenges and innovations. Eur J Pharmacol 2020; 876:173049. [PMID: 32142771 DOI: 10.1016/j.ejphar.2020.173049] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 02/20/2020] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
In the beginning stage of heart disease, the blockage of blood flow frequently occurs due to the persistent damage and even death of myocardium. Cicatricial tissue developed after the death of myocardium can affect heart function, which ultimately leads to heart failure. In recent years, several studies carried out about the use of stem cells such as embryonic, pluripotent, cardiac and bone marrow-derived stem cells as well as myoblasts to repair injured myocardium. Current studies focus more on finding appropriate measures to enhance cell homing and survival in order to increase paracrine function. Until now, there is no universal delivery route for mesenchymal stem cells (MSCs) for different diseases. In this review, we summarize the advantages and challenges of the systemic and local pathways of MSC delivery. In addition, we also describe some advanced measures of cell delivery to improve the efficiency of transplantation. The combination of cells and therapeutic substances could be the most reliable method, which allows donor cells to deliver sufficient amounts of paracrine factors and provide long-lasting effects. The cardiac support devices or tissue engineering techniques have the potential to facilitate the controlled release of stem cells on local tissue for a sustained period. A novel promising epicardial drug delivery system is highlighted here, which not only provides MSCs with a favorable environment to promote retention but also increases the contact area and a number of cells recruited in the heart muscle.
Collapse
Affiliation(s)
- Ziwei Liu
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Reyaj Mikrani
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | | | - Abdoh Taleb
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China
| | - Muhammad Naveed
- School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China
| | - Mirza Muhammad Faran Asraf Baig
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, Jiangsu, 210023, PR China
| | - Qin Zhang
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Cuican Li
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Murad Habib
- Department of Surgery, Ayub Teaching Hospital, Abbottabad, Pakistan
| | - Xingxing Cui
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Kiganda Raymond Sembatya
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Han Lei
- Department of Pharmacy, Jiangsu Worker Medical University, Nanjing, Jiangsu Province, 211198, PR China
| | - Xiaohui Zhou
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, 211198, PR China; Department of Surgery, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu Province, 210017, PR China; Department of Surgery, Nanjing Shuiximen Hospital, Nanjing, Jiangsu Province, 210017, PR China.
| |
Collapse
|
140
|
Bolli R, Kahlon A. Time to end the war on cell therapy. Eur J Heart Fail 2020; 22:893-897. [PMID: 32100951 DOI: 10.1002/ejhf.1767] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/12/2020] [Accepted: 01/27/2020] [Indexed: 01/18/2023] Open
Affiliation(s)
- Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
| | - Arunpreet Kahlon
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY, USA
| |
Collapse
|
141
|
Ory J, Saltzman RG, Blachman-Braun R, Dadoun S, DiFede DL, Premer C, Hurwitz B, Hare JM, Ramasamy R. The Effect of Transendocardial Stem Cell Injection on Erectile Function in Men With Cardiomyopathy: Results From the TRIDENT, POSEIDON, and TAC-HFT Trials. J Sex Med 2020; 17:695-701. [PMID: 32059936 DOI: 10.1016/j.jsxm.2020.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 12/30/2019] [Accepted: 01/04/2020] [Indexed: 01/25/2023]
Abstract
BACKGROUND Despite limited human data, there is a growing interest in the use of stem cell therapy (SCT) for erectile dysfunction (ED). AIM To determine the effect of transendocardial stem cell injection on erectile function on men with cardiomyopathy and ED. METHODS We used International Index of Erectile Function (IIEF) scores collected from men enrolled in 3 separate randomized controlled trials: Comparison of Allogeneic vs Autologous Bone Marrow-Derived Mesenchymal Stem Cells Delivered by Transendocardial Injection in Patients With Ischemic Cardiomyopathy (POSEIDON), Transendocardial Mesenchymal Stem Cells and Mononuclear Bone Marrow Cells for Ischemic Cardiomyopathy (TAC-HFT), and Dose Comparison Study of Allogeneic Mesenchymal Stem Cells in Patients With Ischemic Cardiomyopathy (TRIDENT). These trials recruited patients with ischemic cardiomyopathy and ejection fraction less than 50%. Inclusion and exclusion criteria were identical in all 3 trials. The primary intervention in these trials included transendocardial stem cell injection of stem cells or placebo via cardiac catheterization. The follow-up period was 1 year. IIEF data were collected at baseline and at multiple time points in each trial. OUTCOMES We investigated erectile function over time based on cell dose, cell source (autologous vs allogenic), cell type (mesenchymal stem cells vs bone marrow mononuclear cells), and comparing men who received SCT with those who received placebo. RESULTS A total of 36 men were identified with complete IIEF data. 8 men received placebo injection, and 28 received SCT. The median age was 66.5 years. Comorbidities were similar among all men. Analysis was performed on men with ED, defined by an IIEF-EF score of 24 or less. In the placebo and all-comer SCT group, the median IIEF-EF score was 5 [1-8] and 5 [1-15] at baseline and was 3.5 [3-5.8] and 7 [1-18] at 12 months (P > .05). When analyzed by cell dose, the IIEF-EF score in men who received 200 million cells increased significantly over 12 months (14 [4-23] to 20 [15-24.5], P = .014.) Similarly, an autologous cell source resulted in a similar increase from baseline to 12 months (14 [3.8-23.3] to 20 [12-22], P = .030). CLINICAL IMPLICATIONS Erectile function may improve after systemic delivery of SCT in men with ischemic cardiomyopathy and at least mild ED. STRENGTHS & LIMITATIONS This post hoc analysis is the first to investigate the effect of SCT on erectile function using randomized, placebo-controlled data. Weaknesses include that ED was not a primary end point, and men were not originally recruited based on erectile function. CONCLUSION Future trials on systemic delivery of SCT for ED should focus on high cell dose and autologous cell source, as these seem to provide the best response in men with at least mild ED. Ory J, Saltzman RG, Blachman-Braun R, et al. The Effect of Transendocardial Stem Cell Injection on Erectile Function in Men With Cardiomyopathy: Results From the TRIDENT, POSEIDON, and TAC-HFT Trials. J Sex Med 2020;17:695-701.
Collapse
Affiliation(s)
- Jesse Ory
- Department of Urology, Dalhousie University, Halifax, NS, Canada; Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Russell G Saltzman
- Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL, USA
| | - Ruben Blachman-Braun
- Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL, USA
| | - Simon Dadoun
- Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL, USA
| | - Darcy L DiFede
- Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL, USA
| | - Courtney Premer
- Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL, USA
| | - Barry Hurwitz
- Behavioral Medicine Research Center, University of Miami, Miami, FL, USA
| | - Joshua M Hare
- Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL, USA
| | - Ranjith Ramasamy
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA; Miller School of Medicine, Interdisciplinary Stem Cell Institute, University of Miami, Miami, FL, USA.
| |
Collapse
|
142
|
Yang D, O’Brien CG, Ikeda G, Traverse JH, Taylor DA, Henry TD, Bolli R, Yang PC. Meta-analysis of short- and long-term efficacy of mononuclear cell transplantation in patients with myocardial infarction. Am Heart J 2020; 220:155-175. [PMID: 31821904 PMCID: PMC7173405 DOI: 10.1016/j.ahj.2019.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 09/04/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND Mononuclear cells (MNCs) have been tested in clinical trials across multiple cardiovascular pathologies with mixed results. Major adverse cardiac events (MACE) and markers of cardiovascular capacity have been particularly challenging to interpret because of small size. This meta-analysis is aimed to assess the efficacy of MNC therapy in randomized clinical trials to identify the markers of efficiency that could influence future trial design. METHODS PubMed, Embase, Cochrane library, and ClinicalTrials.gov were searched from inception through November 8, 2018. Changes in left ventricular ejection fraction (LVEF) and infarct size from baseline to follow-up were selected as primary outcomes. Changes in the left ventricular end-systolic volume, left ventricular end-diastolic volume, brain natriuretic peptide/N-terminal pro-B-type natriuretic peptide, 6-minute walk test, New York Heart Association class, and MACE incidences were considered secondary outcomes. RESULTS In short-term follow-up, patients treated with MNCs demonstrated a significant increase in absolute LVEF of 2.21% (95% CI 1.59-2.83; P < .001; I2 = 32%) and 6.01% (95% CI 4.45-7.57; P < .001; I2 = 0%) in acute myocardial infarction (AMI) and ischemic cardiomyopathy studies, respectively. This effect was sustained in long-term follow-up. MNC therapy significantly reduced left ventricular end-systolic volume; however, infarct size, 6-minute walk test, New York Heart Association class, and MACE rates were comparable. CONCLUSIONS MNC therapy may convey a modest but sustained increase in LVEF in ischemic cardiomyopathy patients, supporting further investigation. AMI trials, however, demonstrated minimal improvement in LVEF of unclear clinical significance, suggesting a limited role for MNC therapy in AMI.
Collapse
Affiliation(s)
- Dan Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China,Division of Cardiovascular Medicine, Department of Medicine, and Cardiovascular Institute, Stanford University School of Medicine, CA94305, USA
| | - Connor Galen O’Brien
- Division of Cardiovascular Medicine, Department of Medicine, and Cardiovascular Institute, Stanford University School of Medicine, CA94305, USA
| | - Gentaro Ikeda
- Division of Cardiovascular Medicine, Department of Medicine, and Cardiovascular Institute, Stanford University School of Medicine, CA94305, USA
| | - Jay H. Traverse
- Minneapolis Heart Institute Foundation, Abbott Northwestern Hospital, Minneapolis, MN55407, USA
| | - Doris A. Taylor
- Regenerative Medicine Research, Texas Heart Institute, PO Box 20345, Houston, TX 77225-0345 USA
| | - Timothy D. Henry
- The Carl and Edyth Lindner Center for Research and Education, The Christ Hospital Health Network, Cincinnati, OH45219, USA
| | - Roberto Bolli
- Institute of Molecular Cardiology, University of Louisville, KY40202, USA
| | - Phillip C. Yang
- Division of Cardiovascular Medicine, Department of Medicine, and Cardiovascular Institute, Stanford University School of Medicine, CA94305, USA
| |
Collapse
|
143
|
Mesenchymal Stromal Cells from Patients with Cyanotic Congenital Heart Disease are Optimal Candidate for Cardiac Tissue Engineering. Biomaterials 2020; 230:119574. [DOI: 10.1016/j.biomaterials.2019.119574] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 09/12/2019] [Accepted: 10/18/2019] [Indexed: 12/22/2022]
|
144
|
Maslovaric M, Fatic N, Delević E. State of the art of stem cell therapy for ischaemic cardiomyopathy. Part 2. ANGIOLOGII︠A︡ I SOSUDISTAI︠A︡ KHIRURGII︠A︡ = ANGIOLOGY AND VASCULAR SURGERY 2020; 25:7-26. [PMID: 31855197 DOI: 10.33529/angio2019414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Ischemic cardiomyopathy is becoming a leading cause of morbidity and mortality in the whole world. Stem cell-based therapy is emerging as a promising option for treatment of ischemic cardiomyopathy. Several stem cell types, including cardiac-derived stem cells, bone marrow-derived stem cells, mesenchymal stem cells, skeletal myoblasts, CD34+ and CD133+ stem cells have been used in clinical trials. Clinical effects mostly depend on transdifferentiation and paracrine factors. One important issue is that a low survival and residential rate of transferred stem cells blocks the effective advances in cardiac improvement. Many other factors associated with the efficacy of cell replacement therapy for ischemic cardiomyopathy mainly including the route of delivery, the type and number of stem cell infusion, the timing of injection, patient's physical conditions, the particular microenvironment onto which the cells are delivered, and clinical conditions remain to be addressed. Here we provide an overview of modern methods of stem cell delivery, types of stem cells and discuss the current state of their therapeutic potential.
Collapse
Affiliation(s)
- Milica Maslovaric
- Prona-Montenegrin Science Promotion Foundation, Podgorica, Montenegro
| | - Nikola Fatic
- Department of Vascular Surgery, Clinical Centre of Montenegro, Podgorica, Montenegro
| | - Emilija Delević
- Medical Faculty in Podgorica, University of Montenegro, Podgorica, Montenegro
| |
Collapse
|
145
|
Attia N, Mashal M. Mesenchymal Stem Cells: The Past Present and Future. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1312:107-129. [PMID: 33159306 DOI: 10.1007/5584_2020_595] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The biomedical applications of mesenchymal stem cells (MSCs) have gained expanding attention over the past three decades. MSCs are easily obtained from various tissue types (e.g. bone marrow, fat, cord blood, etc.), are capable of self-renewal, and could be induced to differentiate into several cell lineages for countless biomedical applications. In addition, when transplanted, MSCs are not detected by immune surveillance, thus do not lead to graft rejection. Moreover, they can home towards affected tissues and induce their therapeutic effect in a cell-base and/or a cell-free manner. These properties, and many others, have made MSCs appealing therapeutic cell candidates (for cell and/or gene therapy) in myriad clinical conditions. However, similar to any other therapeutic tool, MSCs still have their own limitations and grey areas that entail more research for better understanding and optimization. Herein, we present a brief overview of various pre-clinical/clinical applications of MSCs in regenerative medicine and discuss limitations and future challenges.
Collapse
Affiliation(s)
- Noha Attia
- Department of Basic Sciences, The American University of Antigua-College of Medicine, Coolidge, Antigua and Barbuda. .,The Center of research and evaluation, The American University of Antigua-College of Medicine, Coolidge, Antigua and Barbuda. .,Histology and Cell Biology Department, Faculty of Medicine, University of Alexandria, Alexandria, Egypt. .,NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.
| | - Mohamed Mashal
- The Center of research and evaluation, The American University of Antigua-College of Medicine, Coolidge, Antigua and Barbuda.,NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| |
Collapse
|
146
|
Khoei SG, Dermani FK, Malih S, Fayazi N, Sheykhhasan M. The Use of Mesenchymal Stem Cells and their Derived Extracellular Vesicles in Cardiovascular Disease Treatment. Curr Stem Cell Res Ther 2020; 15:623-638. [PMID: 32357818 DOI: 10.2174/1574888x15666200501235201] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 03/03/2020] [Accepted: 04/07/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Cardiovascular disease (CVD), including disorders of cardiac muscle and vascular, is the major cause of death globally. Many unsuccessful attempts have been made to intervene in the disease's pathogenesis and treatment. Stem cell-based therapies, as a regeneration strategy, cast a new hope for CVD treatment. One of the most well-known stem cells is mesenchymal stem cells (MSCs), classified as one of the adult stem cells and can be obtained from different tissues. These cells have superior properties, such as proliferation and highly specialized differentiation. On the other hand, they have the potential to modulate the immune system and anti-inflammatory activity. One of their most important features is the secreting the extracellular vesicles (EVs) like exosomes (EXOs) as an intercellular communication system mediating the different physiological and pathophysiological affairs. METHODS In this review study, the importance of MSC and its secretory exosomes for the treatment of heart disease has been together and specifically addressed and the use of these promising natural and accessible agents is predicted to replace the current treatment modalities even faster than we imagine. RESULTS MSC derived EXOs by providing a pro-regenerative condition allowing innate stem cells to repair damaged tissues successfully. As a result, MSCs are considered as the appropriate cellular source in regenerative medicine. In the plethora of experiments, MSCs and MSC-EXOs have been used for the treatment and regeneration of heart diseases and myocardial lesions. CONCLUSION Administration of MSCs has been provided a replacement therapeutic option for heart regeneration, obtaining great attention among the basic researcher and the medical doctors.
Collapse
Affiliation(s)
- Saeideh Gholamzadeh Khoei
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fateme Karimi Dermani
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sara Malih
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nashmin Fayazi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohsen Sheykhhasan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Mesenchymal Stem Cell, the Academic Center for Education, Culture and Research, Qom, Iran
| |
Collapse
|
147
|
Mathiasen AB, Qayyum AA, Jørgensen E, Helqvist S, Kofoed KF, Haack-Sørensen M, Ekblond A, Kastrup J. Bone marrow-derived mesenchymal stromal cell treatment in patients with ischaemic heart failure: final 4-year follow-up of the MSC-HF trial. Eur J Heart Fail 2019; 22:884-892. [PMID: 31863561 DOI: 10.1002/ejhf.1700] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/17/2019] [Accepted: 11/08/2019] [Indexed: 12/17/2022] Open
Abstract
AIMS The study assessed 4-year outcomes of intramyocardial injections of autologous bone marrow-derived mesenchymal stromal cells (MSCs) in patients with ischaemic heart failure. METHODS AND RESULTS The MSC-HF trial was a randomized, double-blind, placebo-controlled trial. Patients were randomized 2:1 to intramyocardial injections of MSCs or placebo. The primary endpoint was change in left ventricular end-systolic volume (LVESV), measured by magnetic resonance imaging or computed tomography. Sixty patients aged 30-80 years with ischaemic heart failure, New York Heart Association class II-III, left ventricular ejection fraction (LVEF) <45% and no further treatment options were randomized. Patients were followed clinically for 12 months and in addition 4-year data of hospitalizations and survival were retrieved. After 12 months, LVESV was significantly reduced in the MSC group and not in the placebo group, with difference between groups of 17.0 ± 16.2 mL (95% confidence interval 8.3-25.7, P = 0.0002). There were also significant improvements in LVEF of 6.2% (P < 0.0001), stroke volume of 16.1 mL (P < 0.0001) and myocardial mass (P = 0.009) between groups. A significant dose-response effect was also observed. Moreover, a significant reduction in the amount of scar tissue and quality of life score in the MSC group but not in the placebo group was observed. After 4 years, there were significantly fewer hospitalizations for angina in the MSC group and otherwise no differences in hospitalizations or survival. No side effects were identified. CONCLUSIONS Intramyocardial injections of autologous bone marrow-derived MSCs improved myocardial function and myocardial mass in patients with ischaemic heart failure.
Collapse
Affiliation(s)
- Anders B Mathiasen
- Department of Cardiology, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Abbas A Qayyum
- Department of Cardiology, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Erik Jørgensen
- Department of Cardiology, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Steffen Helqvist
- Department of Cardiology, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Klaus F Kofoed
- Department of Cardiology, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Mandana Haack-Sørensen
- Cardiac Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Annette Ekblond
- Cardiac Stem Cell Centre, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens Kastrup
- Department of Cardiology, The Heart Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
148
|
Song YL, Jiang H, Jiang NG, Jin YM, Zeng TT. Mesenchymal Stem Cell–Platelet Aggregates Increased in the Peripheral Blood of Patients with Acute Myocardial Infarction and Might Depend on the Stromal Cell-Derived Factor 1/CXCR4 Axis. Stem Cells Dev 2019; 28:1607-1619. [PMID: 31650891 DOI: 10.1089/scd.2019.0154] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Affiliation(s)
- Ya-Li Song
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Hong Jiang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Neng-Gang Jiang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Yong-Mei Jin
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Ting-Ting Zeng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
149
|
Abstract
The investment of nearly 2 decades of clinical investigation into cardiac cell therapy has yet to change cardiovascular practice. Recent insights into the mechanism of cardiac regeneration help explain these results and provide important context in which we can develop next-generation therapies. Non-contractile cells such as bone marrow or adult heart derivatives neither engraft long-term nor induce new muscle formation. Correspondingly, these cells offer little functional benefit to infarct patients. In contrast, preclinical data indicate that transplantation of bona fide cardiomyocytes derived from pluripotent stem cells induces direct remuscularization. This new myocardium beats synchronously with the host heart and induces substantial contractile benefits in macaque monkeys, suggesting that regeneration of contractile myocardium is required to fully recover function. Through a review of the preclinical and clinical trials of cardiac cell therapy, distinguishing the primary mechanism of benefit as either contractile or non-contractile helps appreciate the barriers to cardiac repair and establishes a rational path to optimizing therapeutic benefit.
Collapse
Affiliation(s)
- Kenta Nakamura
- Institute for Stem Cell and Regenerative Medicine, University of Washington
- Center for Cardiovascular Biology, University of Washington
- Department of Medicine/Cardiology, University of Washington
| | - Charles E Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington
- Center for Cardiovascular Biology, University of Washington
- Department of Medicine/Cardiology, University of Washington
- Department of Pathology, University of Washington
- Department of Bioengineering, University of Washington
| |
Collapse
|
150
|
Rockel JS, Rabani R, Viswanathan S. Anti-fibrotic mechanisms of exogenously-expanded mesenchymal stromal cells for fibrotic diseases. Semin Cell Dev Biol 2019; 101:87-103. [PMID: 31757583 DOI: 10.1016/j.semcdb.2019.10.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/11/2019] [Accepted: 10/30/2019] [Indexed: 12/17/2022]
Abstract
Most chronic diseases involving inflammation have a fibrotic component that involves remodeling and excess accumulation of extracellular matrix components. Left unchecked, fibrosis leads to organ failure and death. Mesenchymal stromal cells (MSCs) are emerging as a potent cell-based therapy for a wide spectrum of fibrotic conditions due to their immunomodulatory, anti-inflammatory and anti-fibrotic properties. This review provides an overview of known mechanisms by which MSCs mediate their anti-fibrotic actions and in relation to animal models of pulmonary, liver, renal and cardiac fibrosis. Recent MSC clinical trials results in liver, lung, skin, kidney and hearts are discussed and next steps for future MSC-based therapies including pre-activated or genetically-modified cells, or extracellular vesicles are also considered.
Collapse
Affiliation(s)
- Jason S Rockel
- Arthritis Program, University Health Network, Toronto, ON, Canada; Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.
| | - Razieh Rabani
- Arthritis Program, University Health Network, Toronto, ON, Canada; Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Sowmya Viswanathan
- Arthritis Program, University Health Network, Toronto, ON, Canada; Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada; Division of Hematology, Department of Medicine, University of Toronto, Toronto, Canada
| |
Collapse
|