1
|
Gong X, Jiao Y, Hu H, Zhang R, Jia W, Zhao J, Liu Z, Xin Y, Han W. A prospective randomized controlled study of multi-intravenous infusion of umbilical cord mesenchymal stem cells in patients with heart failure and reduced ejection fraction (PRIME-HFrEF) trial: Rationale and design. Contemp Clin Trials Commun 2024; 41:101350. [PMID: 39246626 PMCID: PMC11377133 DOI: 10.1016/j.conctc.2024.101350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/12/2024] [Accepted: 08/11/2024] [Indexed: 09/10/2024] Open
Abstract
Background and objective The use of mesenchymal stem cells for heart failure treatment has gained increasing interest. However, most studies have relied on a single injection approach, with no research yet confirming the effects of multiple administrations. The present trial aims to investigate the safety and efficacy of multi-intravenous infusion of umbilical cord-mesenchymal stem cells (UC-MSCs) in patients with heart failure and reduced ejection fraction (HFrEF). Methods The PRIME-HFrEF trial is a single-center, prospective, randomized, triple-blinded, placebo-controlled trial of multi-intravenous infusion of UC-MSCs in HFrEF patients. A total of 40 patients meeting the inclusion criteria for HFrEF were enrolled and randomized 1:1 to the MSC group or the placebo group. Patients enrolled will receive intravenous injections of either UC-MSCs or placebo every 6 weeks for three times. Both groups will be followed up for 12 months. The primary safety endpoint is the incidence of serious adverse events. The primary efficacy endpoint is a change in left ventricular ejection fraction (LVEF) measured by left ventricular opacification (LVO) with contrast echocardiography and magnetic resonance imaging (MRI) at 12 months. The secondary endpoints include a composite of the incidence of death and re-hospitalization caused by heart failure at the 12th month, serum NT-proBNP, growth stimulation expressed gene 2 (ST2), and a change of right ventricular structure and function. Conclusions The PRIME-HFrEF study is designed to shed new light on multiple UC-MSC administration regimens for heart failure treatment.
Collapse
Affiliation(s)
- Xin Gong
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yuheng Jiao
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Hao Hu
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Rongzhen Zhang
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wenwen Jia
- Institute for Regenerative Medicine, National Stem Cell Translational Resource Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200120, China
| | - Jun Zhao
- Department of Nuclear Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhongmin Liu
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China
| | - Yuanfeng Xin
- Department of Cardiovascular Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wei Han
- Department of Heart failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| |
Collapse
|
2
|
Thej C, Kishore R. Epigenetic regulation of sex dimorphism in cardiovascular health. Can J Physiol Pharmacol 2024; 102:498-510. [PMID: 38427976 PMCID: PMC11789622 DOI: 10.1139/cjpp-2023-0406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality, affecting people of all races, ages, and sexes. Substantial sex dimorphism exists in the prevalence, manifestation, and outcomes of CVDs. Understanding the role of sex hormones as well as sex-hormone-independent epigenetic mechanisms could play a crucial role in developing effective and sex-specific cardiovascular therapeutics. Existing research highlights significant disparities in sex hormones, epigenetic regulators, and gene expression related to cardiac health, emphasizing the need for a nuanced understanding of these variations between men and women. Despite these differences, current treatment approaches for CVDs often lack sex-specific considerations. A pivotal shift toward personalized medicine, informed by comprehensive insights into sex-specific DNA methylation, histone modifications, and non-coding RNA dynamics, holds the potential to revolutionize CVD management. By understanding sex-specific epigenetic complexities, independent of sex hormone influence, future cardiovascular research can be tailored to achieve effective diagnostic and therapeutic interventions for both men and women. This review summarizes the current knowledge and gaps in epigenetic mechanisms and sex dimorphism implicated in CVDs.
Collapse
Affiliation(s)
- Charan Thej
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Raj Kishore
- Aging and Cardiovascular Discovery Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
- Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
3
|
Du L, Wang X, Guo Y, Tao T, Wu H, Xu X, Zhang B, Chen T, Xu Q, Guo X. Altered lipid metabolism promoting cardiac fibrosis is mediated by CD34 + cell-derived FABP4 + fibroblasts. Exp Mol Med 2024; 56:1869-1886. [PMID: 39198543 PMCID: PMC11372182 DOI: 10.1038/s12276-024-01309-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/22/2024] [Accepted: 05/21/2024] [Indexed: 09/01/2024] Open
Abstract
Hyperlipidemia and hypertension might play a role in cardiac fibrosis, in which a heterogeneous population of fibroblasts seems important. However, it is unknown whether CD34+ progenitor cells are involved in the pathogenesis of heart fibrosis. This study aimed to explore the mechanism of CD34+ cell differentiation in cardiac fibrosis during hyperlipidemia. Through the analysis of transcriptomes from 50,870 single cells extracted from mouse hearts and 76,851 single cells from human hearts, we have effectively demonstrated the evolving cellular landscape throughout cardiac fibrosis. Disturbances in lipid metabolism can accelerate the development of fibrosis. Through the integration of bone marrow transplantation models and lineage tracing, our study showed that hyperlipidemia can expedite the differentiation of non-bone marrow-derived CD34+ cells into fibroblasts, particularly FABP4+ fibroblasts, in response to angiotensin II. Interestingly, the partial depletion of CD34+ cells led to a notable reduction in triglycerides in the heart, mitigated fibrosis, and improved cardiac function. Furthermore, immunostaining of human heart tissue revealed colocalization of CD34+ cells and fibroblasts. Mechanistically, our investigation of single-cell RNA sequencing data through pseudotime analysis combined with in vitro cellular studies revealed the crucial role of the PPARγ/Akt/Gsk3β pathway in orchestrating the differentiation of CD34+ cells into FABP4+ fibroblasts. Through our study, we generated valuable insights into the cellular landscape of CD34+ cell-derived cells in the hypertrophic heart with hyperlipidemia, indicating that the differentiation of non-bone marrow-derived CD34+ cells into FABP4+ fibroblasts during this process accelerates lipid accumulation and promotes heart failure via the PPARγ/Akt/Gsk3β pathway.
Collapse
Affiliation(s)
- Luping Du
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xuyang Wang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yan Guo
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tingting Tao
- Department of Cardiovascular Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hong Wu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaodong Xu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bohuan Zhang
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Chen
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Health care, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
4
|
Nativi-Nicolau J, Yilmaz A, Dasgupta N, Macey R, Cochrane J, Peatman J, Summers C, Luth J, Zolty R. Six-minute walk test as clinical end point in cardiomyopathy clinical trials, including ATTR-CM: a systematic literature review. J Comp Eff Res 2024; 13:e230158. [PMID: 38869839 PMCID: PMC11234454 DOI: 10.57264/cer-2023-0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 05/20/2024] [Indexed: 06/14/2024] Open
Abstract
Aim: The six-minute walk test (6MWT) is a common measure of functional capacity in patients with heart failure (HF). Primary clinical study end points in cardiomyopathy (CM) trials, including transthyretin-mediated amyloidosis with CM (ATTR-CM), are often limited to hospitalization and mortality. Objective: To investigate the relationship between the 6MWT and hospitalization or mortality in CM, including ATTR-CM. Method: A PRISMA-guided systematic literature review was conducted using search terms for CM, 6MWT, hospitalization and mortality. Results: Forty-one studies were identified that reported 6MWT data and hospitalization or mortality data for patients with CM. The data suggest that a greater 6MWT distance is associated with a reduced risk of hospitalization or mortality in CM. Conclusion: The 6MWT is an accepted alternative end point in CM trials, including ATTR-CM.
Collapse
Affiliation(s)
| | - Ali Yilmaz
- Division of Cardiovascular Imaging, University Hospital Münster, 48149, Münster, Germany
| | - Noel Dasgupta
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Richard Macey
- Adelphi Values PROVE, Bollington, Cheshire, UK, SK10 5JB
| | - James Cochrane
- Adelphi Values PROVE, Bollington, Cheshire, UK, SK10 5JB
| | - Judith Peatman
- Adelphi Values PROVE, Bollington, Cheshire, UK, SK10 5JB
| | - Catherine Summers
- Medical Affairs Department, Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | - Jennifer Luth
- Medical Affairs Department, Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | - Ronald Zolty
- Division of Cardiovascular Medicine, University of Nebraska Medical Center (UNMC), Omaha, NE 68198 USA
| |
Collapse
|
5
|
Fatehi Hassanabad A, Zarzycki AN, Fedak PWM. Cellular and molecular mechanisms driving cardiac tissue fibrosis: On the precipice of personalized and precision medicine. Cardiovasc Pathol 2024; 71:107635. [PMID: 38508436 DOI: 10.1016/j.carpath.2024.107635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024] Open
Abstract
Cardiac fibrosis is a significant contributor to heart failure, a condition that continues to affect a growing number of patients worldwide. Various cardiovascular comorbidities can exacerbate cardiac fibrosis. While fibroblasts are believed to be the primary cell type underlying fibrosis, recent and emerging data suggest that other cell types can also potentiate or expedite fibrotic processes. Over the past few decades, clinicians have developed therapeutics that can blunt the development and progression of cardiac fibrosis. While these strategies have yielded positive results, overall clinical outcomes for patients suffering from heart failure continue to be dire. Herein, we overview the molecular and cellular mechanisms underlying cardiac tissue fibrosis. To do so, we establish the known mechanisms that drive fibrosis in the heart, outline the diagnostic tools available, and summarize the treatment options used in contemporary clinical practice. Finally, we underscore the critical role the immune microenvironment plays in the pathogenesis of cardiac fibrosis.
Collapse
Affiliation(s)
- Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Anna N Zarzycki
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul W M Fedak
- Section of Cardiac Surgery, Department of Cardiac Science, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
6
|
Chowdhury MA, Zhang JJ, Rizk R, Chen WCW. Stem cell therapy for heart failure in the clinics: new perspectives in the era of precision medicine and artificial intelligence. Front Physiol 2024; 14:1344885. [PMID: 38264333 PMCID: PMC10803627 DOI: 10.3389/fphys.2023.1344885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 12/26/2023] [Indexed: 01/25/2024] Open
Abstract
Stem/progenitor cells have been widely evaluated as a promising therapeutic option for heart failure (HF). Numerous clinical trials with stem/progenitor cell-based therapy (SCT) for HF have demonstrated encouraging results, but not without limitations or discrepancies. Recent technological advancements in multiomics, bioinformatics, precision medicine, artificial intelligence (AI), and machine learning (ML) provide new approaches and insights for stem cell research and therapeutic development. Integration of these new technologies into stem/progenitor cell therapy for HF may help address: 1) the technical challenges to obtain reliable and high-quality therapeutic precursor cells, 2) the discrepancies between preclinical and clinical studies, and 3) the personalized selection of optimal therapeutic cell types/populations for individual patients in the context of precision medicine. This review summarizes the current status of SCT for HF in clinics and provides new perspectives on the development of computation-aided SCT in the era of precision medicine and AI/ML.
Collapse
Affiliation(s)
- Mohammed A. Chowdhury
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
- Department of Public Health and Health Sciences, Health Sciences Ph.D. Program, School of Health Sciences, University of South Dakota, Vermillion, SD, United States
- Department of Cardiology, North Central Heart, Avera Heart Hospital, Sioux Falls, SD, United States
| | - Jing J. Zhang
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| | - Rodrigue Rizk
- Department of Computer Science, University of South Dakota, Vermillion, SD, United States
| | - William C. W. Chen
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD, United States
| |
Collapse
|
7
|
Reid A, Hussain M, Veerapen J, Ramaseshan R, Hall R, Bowles R, Jones DA, Mathur A. DCM Support: cell therapy and circulatory support for dilated cardiomyopathy patients with severe ventricular impairment. ESC Heart Fail 2023. [PMID: 37190883 PMCID: PMC10375109 DOI: 10.1002/ehf2.14393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 04/11/2023] [Accepted: 04/24/2023] [Indexed: 05/17/2023] Open
Abstract
AIMS The DCM Support trial (NCT03572660) uses a percutaneous circulatory support device (Impella CP, Abiomed, Danvers, MA, USA) to improve the safety of an intracoronary cell infusion procedure in patients with dilated cardiomyopathy (DCM) and a severely reduced left ventricular ejection fraction (LVEF). METHODS AND RESULTS DCM Support is a single-site, single-arm Phase II trial enrolling 20 symptomatic DCM patients with an LVEF ≤ 35% despite optimal medical and device therapy. After 5 days of granulocyte colony-stimulating factor therapy and a subsequent bone marrow aspiration, patients undergo an intracoronary infusion of autologous bone-marrow-derived mononuclear cells. The Impella CP device is used to provide haemodynamic support during the infusion procedure. The trial's primary endpoint is change in LVEF from baseline at 3 months. Secondary efficacy endpoints are change in LVEF from baseline at 12 months, and change in exercise capacity, New York Heart Association class, quality of life, and N-terminal pro-B-type natriuretic peptide levels from baseline at 3 and 12 months. Safety endpoints include procedural safety and major adverse cardiac events at 3 and 12 months. CONCLUSIONS This is the first trial to assess the safety and efficacy of cytokine and autologous intracoronary cell therapy with a procedural circulatory support device for patients with severe left ventricular impairment. This novel combination may allow us to target a patient population most at need of this therapy.
Collapse
Affiliation(s)
- Alice Reid
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- NIHR Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, London, EC1M 6BQ, United Kingdom
| | - Mohsin Hussain
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Jessry Veerapen
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Rohini Ramaseshan
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Russell Hall
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Ruth Bowles
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- NIHR Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, London, EC1M 6BQ, United Kingdom
| | - Daniel A Jones
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Anthony Mathur
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- NIHR Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, London, EC1M 6BQ, United Kingdom
| |
Collapse
|
8
|
Qayyum AA, van Klarenbosch B, Frljak S, Cerar A, Poglajen G, Traxler-Weidenauer D, Nadrowski P, Paitazoglou C, Vrtovec B, Bergmann MW, Chamuleau SAJ, Wojakowski W, Gyöngyösi M, Kraaijeveld A, Hansen KS, Vrangbaek K, Jørgensen E, Helqvist S, Joshi FR, Johansen EM, Follin B, Juhl M, Højgaard LD, Mathiasen AB, Ekblond A, Haack-Sørensen M, Kastrup J. Effect of allogeneic adipose tissue-derived mesenchymal stromal cell treatment in chronic ischaemic heart failure with reduced ejection fraction - the SCIENCE trial. Eur J Heart Fail 2023; 25:576-587. [PMID: 36644821 DOI: 10.1002/ejhf.2772] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 11/27/2022] [Accepted: 01/08/2023] [Indexed: 01/17/2023] Open
Abstract
AIMS The aim of the SCIENCE trial was to investigate whether a single treatment with direct intramyocardial injections of adipose tissue-derived mesenchymal stromal cells (CSCC_ASCs) was safe and improved cardiac function in patients with chronic ischaemic heart failure with reduced ejection fraction (HFrEF). METHODS AND RESULTS The study was a European multicentre, double-blind, placebo-controlled phase II trial using allogeneic CSCC_ASCs from healthy donors or placebo (2:1 randomization). Main inclusion criteria were New York Heart Association (NYHA) class II-III, left ventricular ejection fraction (LVEF) <45%, and N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels >300 pg/ml. CSCC_ASCs or placebo (isotonic saline) were injected directly into viable myocardium. The primary endpoint was change in left ventricular end-systolic volume (LVESV) at 6-month follow-up measured by echocardiography. A total of 133 symptomatic HFrEF patients were included. The treatment was safe without any drug-related severe adverse events or difference in cardiac-related adverse events during a 3-year follow-up period. There were no significant differences between groups during follow-up in LVESV (0.3 ± 5.0 ml, p = 0.945), nor in secondary endpoints of left ventricular end-diastolic volume (-2.0 ± 6.0 ml, p = 0.736) and LVEF (-1.6 ± 1.0%, p = 0.119). The NYHA class improved slightly within the first year in both groups without any difference between groups. There were no changes in 6-min walk test, NT-proBNP, C-reactive protein or quality of life the first year in any groups. CONCLUSION The SCIENCE trial demonstrated safety of intramyocardial allogeneic CSCC_ASC therapy in patients with chronic HFrEF. However, it was not possible to improve the pre-defined endpoints and induce restoration of cardiac function or clinical symptoms.
Collapse
Affiliation(s)
- Abbas Ali Qayyum
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Bas van Klarenbosch
- Department of Cardiology and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sabina Frljak
- Advanced Heart Failure and Transplantation Center, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Andraz Cerar
- Advanced Heart Failure and Transplantation Center, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Gregor Poglajen
- Advanced Heart Failure and Transplantation Center, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | | | - Pawel Nadrowski
- Department of Cardiology and Structural Heart Diseases, Medical University of Silesia, Katowice, Poland
| | | | - Bojan Vrtovec
- Advanced Heart Failure and Transplantation Center, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Martin W Bergmann
- Department of Cardiology, Asklepios Klinik St. Georg, Hamburg, Germany
| | - Steven A J Chamuleau
- Department of Cardiology and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Wojtek Wojakowski
- Department of Cardiology and Structural Heart Diseases, Medical University of Silesia, Katowice, Poland
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Adriaan Kraaijeveld
- Department of Cardiology and Regenerative Medicine Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kristian Schultz Hansen
- Faculty of Social Sciences and the Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karsten Vrangbaek
- Faculty of Social Sciences and the Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Erik Jørgensen
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Steffen Helqvist
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Francis Richard Joshi
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ellen Mønsted Johansen
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Bjarke Follin
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Morten Juhl
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lisbeth Drozd Højgaard
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Anders Bruun Mathiasen
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Annette Ekblond
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Mandana Haack-Sørensen
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens Kastrup
- Department of Cardiology and Cardiology Stem Cell Centre, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
9
|
Fujita Y, Kawamoto A. Therapeutic Angiogenesis Using Autologous CD34-Positive Cells for Vascular Diseases. Ann Vasc Dis 2022; 15:241-252. [PMID: 36644256 PMCID: PMC9816028 DOI: 10.3400/avd.ra.22-00086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 08/13/2022] [Indexed: 12/25/2022] Open
Abstract
CD34 is a cell surface marker, which is expressed in various somatic stem/progenitor cells such as bone marrow (BM)-derived hematopoietic stem cells and endothelial progenitor cells (EPCs), skeletal muscle satellite cells, epithelial hair follicle stem cells, and adipose tissue mesenchymal stem cells. CD34+ cells in BM and peripheral blood are known as a rich source of EPCs. Thus, vascular regeneration therapy using granulocyte colony stimulating factor (G-CSF) mobilized- or BM CD34+ cells has been carried out in patients with various vascular diseases such as chronic severe lower limb ischemia, acute myocardial infarction, refractory angina, ischemic cardiomyopathy, and dilated cardiomyopathy as well as ischemic stroke. Pilot and randomized clinical trials demonstrated the safety, feasibility, and effectiveness of the CD34+ cell therapy in peripheral arterial, cardiovascular, and cerebrovascular diseases. This review provides an overview of the preclinical and clinical reports of CD34+ cell therapy for vascular regeneration.
Collapse
Affiliation(s)
- Yasuyuki Fujita
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Hyogo, Japan
| | - Atsuhiko Kawamoto
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Hyogo, Japan,Corresponding author: Atsuhiko Kawamoto, MD, PhD. Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, 1-5-4 Minatojima-Minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan Tel: +81-78-304-5772, Fax: +81-78-304-5263, E-mail:
| |
Collapse
|
10
|
Banovic M, Poglajen G, Vrtovec B, Ristic A. Contemporary Challenges of Regenerative Therapy in Patients with Ischemic and Non-Ischemic Heart Failure. J Cardiovasc Dev Dis 2022; 9:jcdd9120429. [PMID: 36547426 PMCID: PMC9783726 DOI: 10.3390/jcdd9120429] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/11/2022] [Accepted: 10/26/2022] [Indexed: 12/02/2022] Open
Abstract
It has now been almost 20 years since first clinical trials of stem cell therapy for heart repair were initiated. While initial preclinical data were promising and suggested that stem cells may be able to directly restore a diseased myocardium, this was never unequivocally confirmed in the clinical setting. Clinical trials of cell therapy did show the process to be feasible and safe. However, the clinical benefits of this treatment modality in patients with ischemic and non-ischemic heart failure have not been consistently confirmed. What is more, in the rapidly developing field of stem cell therapy in patients with heart failure, relevant questions regarding clinical trials' protocol streamlining, optimal patient selection, stem cell type and dose, and the mode of cell delivery remain largely unanswered. Recently, novel approaches to myocardial regeneration, including the use of pluripotent and allogeneic stem cells and cell-free therapeutic approaches, have been proposed. Thus, in this review, we aim to outline current knowledge and highlight contemporary challenges and dilemmas in clinical aspects of stem cell and regenerative therapy in patients with chronic ischemic and non-ischemic heart failure.
Collapse
Affiliation(s)
- Marko Banovic
- Cardiology Department, University Clinical Center of Serbia, 11000 Beograd, Serbia
- Belgrade Medical School, 11000 Belgrade, Serbia
- Correspondence: (M.B.); (G.P.)
| | - Gregor Poglajen
- Advanced Heart Failure and Transplantation Center, Department of Cardiology, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Medical Faculty Ljubljana, University of Ljubljana, 1000 Ljubljana, Slovenia
- Correspondence: (M.B.); (G.P.)
| | - Bojan Vrtovec
- Advanced Heart Failure and Transplantation Center, Department of Cardiology, University Medical Center Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Medical Faculty Ljubljana, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Arsen Ristic
- Cardiology Department, University Clinical Center of Serbia, 11000 Beograd, Serbia
- Belgrade Medical School, 11000 Belgrade, Serbia
| |
Collapse
|
11
|
Sim DS, Jones DA, Davies C, Locca D, Veerapen J, Reid A, Godec T, Martin J, Mathur A. Cell administration routes for heart failure: a comparative re-evaluation of the REGENERATE-DCM and REGENERATE-IHD trials. Regen Med 2022; 17:891-903. [PMID: 36226504 DOI: 10.2217/rme-2022-0138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims: Given the logistical issues surrounding intramyocardial cell delivery, we sought to address the efficacy of the simpler, more accessible intracoronary route by re-evaluating REGENERATE-DCM and REGENERATE-IHD (autologous cell therapy trials for heart failure; n = 150). Methods: A retrospective statistical analysis was performed on the trials' combined data. The following end points were evaluated: left ventricular ejection fraction (LVEF), N-terminal pro brain natriuretic peptide concentration (NT-proBNP), New York Heart Association class (NYHA) and quality of life. Results: This demonstrated a new efficacy signal for intracoronary delivery, with significant benefits to: LVEF (3.7%; p = 0.01), NT-proBNP (median -76 pg/ml; p = 0.04), NYHA class (48% patients; p = 0.01) and quality of life (12 ± 19; p = 0.006). The improvements in LVEF, NYHA and quality of life scores remained significant compared to the control group. Conclusion: The efficacy and logistical simplicity of intracoronary delivery should be taken into consideration for future trials.
Collapse
Affiliation(s)
- Doo Sun Sim
- Centre for Cardiovascular Medicine & Devices, William Harvey Research Institute, Queen Mary University of London, London, UK.,Department of Cardiovascular Medicine, Chonnam National University Hospital, Chonnam National University School of Medicine, Gwanjgu, Republic of Korea
| | - Daniel A Jones
- Centre for Cardiovascular Medicine & Devices, William Harvey Research Institute, Queen Mary University of London, London, UK.,Department of Interventional Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Ceri Davies
- Centre for Cardiovascular Medicine & Devices, William Harvey Research Institute, Queen Mary University of London, London, UK.,Department of Interventional Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Didier Locca
- Centre for Cardiovascular Medicine & Devices, William Harvey Research Institute, Queen Mary University of London, London, UK.,École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jessry Veerapen
- Centre for Cardiovascular Medicine & Devices, William Harvey Research Institute, Queen Mary University of London, London, UK.,Department of Interventional Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| | - Alice Reid
- Centre for Cardiovascular Medicine & Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Thomas Godec
- Centre for Cardiovascular Medicine & Devices, William Harvey Research Institute, Queen Mary University of London, London, UK.,Barts Cardiovascular Clinical Trials Unit, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - Anthony Mathur
- Centre for Cardiovascular Medicine & Devices, William Harvey Research Institute, Queen Mary University of London, London, UK.,Department of Interventional Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
| |
Collapse
|
12
|
Hall RC, Ramaseshan R, Reid A, Jones DA, Mathur A. Case report: Cytokine therapy and an intracoronary autologous bone marrow-derived cell infusion with Impella support in a patient with dilated cardiomyopathy and a severely reduced ejection fraction. Front Cardiovasc Med 2022; 9:1002508. [PMID: 36172585 PMCID: PMC9510980 DOI: 10.3389/fcvm.2022.1002508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/25/2022] [Indexed: 11/24/2022] Open
Abstract
Introduction This is the first reported case of a patient with dilated cardiomyopathy (DCM) and severely impaired left ventricular function to receive a combined treatment of granulocyte colony-stimulating factor therapy and an intracoronary delivery of autologous bone marrow-derived mononuclear cells with percutaneous circulatory assistance (the Impella CP device; Abiomed, Danvers, MA). Main symptoms and outcome Three months post-treatment, the gentleman in his early 70s demonstrated an improvement in left ventricular ejection fraction (13–17%) and a reduction in New York Heart Association class from III to class I. There was also an improvement in his 6-minute walk test (147–357 meters), N-terminal pro-brain natriuretic peptide level (14,099–7,129 ng/l) and quality of life scores. There were no safety concerns during the treatment or follow-up. Conclusion This case report suggests combined cell and cytokine therapy with adjunctive circulatory support could be a safe and promising treatment for patients with DCM and severely reduced ejection fraction.
Collapse
Affiliation(s)
| | - Rohini Ramaseshan
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Alice Reid
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Daniel A. Jones
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Anthony Mathur
- Barts Heart Centre, Barts Health NHS Trust, London, United Kingdom
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- *Correspondence: Anthony Mathur
| |
Collapse
|
13
|
Vrtovec B, Frljak S, Poglajen G, Zemljic G, Cerar A, Sever M, Haddad F, Wu JC. A PILOT CLINICAL TRIAL OF CELL THERAPY IN HEART FAILURE WITH PRESERVED EJECTION FRACTION. Eur J Heart Fail 2022; 24:1441-1449. [PMID: 35775390 PMCID: PMC9540623 DOI: 10.1002/ejhf.2596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/19/2022] [Accepted: 06/20/2022] [Indexed: 12/02/2022] Open
Abstract
Aims We investigated the effects of CD34+ cell therapy in patients with heart failure with preserved ejection fraction (HFpEF). Methods and results In a prospective pilot study, we enrolled 30 patients with HFpEF. In Phase 1, patients were treated with medical therapy for 6 months. Thereafter, all patients underwent CD34+ cell transplantation. Using electroanatomical mapping, we measured local mechanical diastolic delay and myocardial viability to guide the targeting of cell injections. Patients were followed for 6 months after cell transplantation (Phase 2), and the primary endpoint was the difference in change in E/e′ between Phase 1 and Phase 2. In Phase 1, the decrease in E/e′ was significantly less pronounced than in Phase 2 (−0.33 ± 1.72 vs. −3.77 ± 2.66, p = 0.001). During Phase 1, there was no significant change in global systolic strain (GLS; from −12.5 ± 2.4% to −12.8 ± 2.6%, p = 0.77), N‐terminal pro‐B‐type natriuretic peptide (NT‐proBNP; from 1463 ± 1247 pg/ml to 1298 ± 931 pg/ml, p = 0.31), or 6‐min walk test (6MWT; from 391 ± 75 m to 402 ± 93 m, p = 0.42). In Phase 2, an improvement was noted in NT‐proBNP (from 1298 ± 931 pg/ml to 887 ± 809 pg/ml, p = 0.02) and 6MWT (from 402 ± 93 m to 438 ± 72 m, p = 0.02). Although GLS did not change significantly in Phase 2 (from −12.8 ± 2.6% to −13.8 ± 2.7%, p = 0.36), we found improved local systolic strain at cell injection sites (−3.4 ± 6.8%, p = 0.005). Conclusions In this non‐randomized trial, transendocardial CD34+ cell therapy in HFpEF was associated with an improvement in E/e′, NT‐proBNP, exercise capacity, and local myocardial strain at the cell injection sites. Clinical Trial Registration: ClinicalTrials.gov NCT02923609.
Collapse
Affiliation(s)
- Bojan Vrtovec
- Advanced Heart Failure and Transplantation Center, Department of Cardiology, UMC Ljubljana, Slovenia
| | - Sabina Frljak
- Advanced Heart Failure and Transplantation Center, Department of Cardiology, UMC Ljubljana, Slovenia
| | - Gregor Poglajen
- Advanced Heart Failure and Transplantation Center, Department of Cardiology, UMC Ljubljana, Slovenia
| | - Gregor Zemljic
- Advanced Heart Failure and Transplantation Center, Department of Cardiology, UMC Ljubljana, Slovenia
| | - Andraz Cerar
- Advanced Heart Failure and Transplantation Center, Department of Cardiology, UMC Ljubljana, Slovenia
| | - Matjaz Sever
- Department of Hematology, UMC Ljubljana, Slovenia
| | - Francois Haddad
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
14
|
Wang X, Wang R, Jiang L, Xu Q, Guo X. Endothelial repair by stem and progenitor cells. J Mol Cell Cardiol 2021; 163:133-146. [PMID: 34743936 DOI: 10.1016/j.yjmcc.2021.10.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/20/2021] [Accepted: 10/26/2021] [Indexed: 12/19/2022]
Abstract
The integrity of the endothelial barrier is required to maintain vascular homeostasis and fluid balance between the circulatory system and surrounding tissues and to prevent the development of vascular disease. However, the origin of the newly developed endothelial cells is still controversial. Stem and progenitor cells have the potential to differentiate into endothelial cell lines and stimulate vascular regeneration in a paracrine/autocrine fashion. The one source of new endothelial cells was believed to come from the bone marrow, which was challenged by the recent findings. By administration of new techniques, including genetic cell lineage tracing and single cell RNA sequencing, more solid data were obtained that support the concept of stem/progenitor cells for regenerating damaged endothelium. Specifically, it was found that tissue resident endothelial progenitors located in the vessel wall were crucial for endothelial repair. In this review, we summarized the latest advances in stem and progenitor cell research in endothelial regeneration through findings from animal models and discussed clinical data to indicate the future direction of stem cell therapy.
Collapse
Affiliation(s)
- Xuyang Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruilin Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liujun Jiang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
15
|
Diaz-Navarro R, Urrútia G, Cleland JG, Poloni D, Villagran F, Acosta-Dighero R, Bangdiwala SI, Rada G, Madrid E. Stem cell therapy for dilated cardiomyopathy. Cochrane Database Syst Rev 2021; 7:CD013433. [PMID: 34286511 PMCID: PMC8406792 DOI: 10.1002/14651858.cd013433.pub2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Stem cell therapy (SCT) has been proposed as an alternative treatment for dilated cardiomyopathy (DCM), nonetheless its effectiveness remains debatable. OBJECTIVES To assess the effectiveness and safety of SCT in adults with non-ischaemic DCM. SEARCH METHODS We searched CENTRAL in the Cochrane Library, MEDLINE, and Embase for relevant trials in November 2020. We also searched two clinical trials registers in May 2020. SELECTION CRITERIA Eligible studies were randomized controlled trials (RCT) comparing stem/progenitor cells with no cells in adults with non-ischaemic DCM. We included co-interventions such as the administration of stem cell mobilizing agents. Studies were classified and analysed into three categories according to the comparison intervention, which consisted of no intervention/placebo, cell mobilization with cytokines, or a different mode of SCT. The first two comparisons (no cells in the control group) served to assess the efficacy of SCT while the third (different mode of SCT) served to complement the review with information about safety and other information of potential utility for a better understanding of the effects of SCT. DATA COLLECTION AND ANALYSIS Two review authors independently screened all references for eligibility, assessed trial quality, and extracted data. We undertook a quantitative evaluation of data using random-effects meta-analyses. We evaluated heterogeneity using the I² statistic. We could not explore potential effect modifiers through subgroup analyses as they were deemed uninformative due to the scarce number of trials available. We assessed the certainty of the evidence using the GRADE approach. We created summary of findings tables using GRADEpro GDT. We focused our summary of findings on all-cause mortality, safety, health-related quality of life (HRQoL), performance status, and major adverse cardiovascular events. MAIN RESULTS We included 13 RCTs involving 762 participants (452 cell therapy and 310 controls). Only one study was at low risk of bias in all domains. There were many shortcomings in the publications that did not allow a precise assessment of the risk of bias in many domains. Due to the nature of the intervention, the main source of potential bias was lack of blinding of participants (performance bias). Frequently, the format of the continuous data available was not ideal for use in the meta-analysis and forced us to seek strategies for transforming data in a usable format. We are uncertain whether SCT reduces all-cause mortality in people with DCM compared to no intervention/placebo (mean follow-up 12 months) (risk ratio (RR) 0.84, 95% confidence interval (CI) 0.54 to 1.31; I² = 0%; studies = 7, participants = 361; very low-certainty evidence). We are uncertain whether SCT increases the risk of procedural complications associated with cells injection in people with DCM (data could not be pooled; studies = 7; participants = 361; very low-certainty evidence). We are uncertain whether SCT improves HRQoL (standardized mean difference (SMD) 0.62, 95% CI 0.01 to 1.23; I² = 72%; studies = 5, participants = 272; very low-certainty evidence) and functional capacity (6-minute walk test) (mean difference (MD) 70.12 m, 95% CI -5.28 to 145.51; I² = 87%; studies = 5, participants = 230; very low-certainty evidence). SCT may result in a slight functional class (New York Heart Association) improvement (data could not be pooled; studies = 6, participants = 398; low-certainty evidence). None of the included studies reported major adverse cardiovascular events as defined in our protocol. SCT may not increase the risk of ventricular arrhythmia (data could not be pooled; studies = 8, participants = 504; low-certainty evidence). When comparing SCT to cell mobilization with granulocyte-colony stimulating factor (G-CSF), we are uncertain whether SCT reduces all-cause mortality (RR 0.46, 95% CI 0.16 to 1.31; I² = 39%; studies = 3, participants = 195; very low-certainty evidence). We are uncertain whether SCT increases the risk of procedural complications associated with cells injection (studies = 1, participants = 60; very low-certainty evidence). SCT may not improve HRQoL (MD 4.61 points, 95% CI -5.62 to 14.83; studies = 1, participants = 22; low-certainty evidence). SCT may improve functional capacity (6-minute walk test) (MD 140.14 m, 95% CI 119.51 to 160.77; I² = 0%; studies = 2, participants = 155; low-certainty evidence). None of the included studies reported MACE as defined in our protocol or ventricular arrhythmia. The most commonly reported outcomes across studies were based on physiological measures of cardiac function where there were some beneficial effects suggesting potential benefits of SCT in people with non-ischaemic DCM. However, it is unclear if this intermediate effects translates into clinical benefits for these patients. With regard to specific aspects related to the modality of cell therapy and its delivery, uncertainties remain as subgroup analyses could not be performed as planned, making it necessary to wait for the publication of several studies that are currently in progress before any firm conclusion can be reached. AUTHORS' CONCLUSIONS We are uncertain whether SCT in people with DCM reduces the risk of all-cause mortality and procedural complications, improves HRQoL, and performance status (exercise capacity). SCT may improve functional class (NYHA), compared to usual care (no cells). Similarly, when compared to G-CSF, we are also uncertain whether SCT in people with DCM reduces the risk of all-cause mortality although some studies within this comparison observed a favourable effect that should be interpreted with caution. SCT may not improve HRQoL but may improve to some extent performance status (exercise capacity). Very low-quality evidence reflects uncertainty regarding procedural complications. These suggested beneficial effects of SCT, although uncertain due to the very low certainty of the evidence, are accompanied by favourable effects on some physiological measures of cardiac function. Presently, the most effective mode of administration of SCT and the population that could benefit the most is unclear. Therefore, it seems reasonable that use of SCT in people with DCM is limited to clinical research settings. Results of ongoing studies are likely to modify these conclusions.
Collapse
Affiliation(s)
- Rienzi Diaz-Navarro
- Department of Internal Medicine, School of Medicine, Universidad de Valparaiso, Vina del Mar, Chile
| | - Gerard Urrútia
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - John Gf Cleland
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Daniel Poloni
- Department of Internal Medicine, School of Medicine, Universidad de Valparaiso, Vina del Mar, Chile
| | - Francisco Villagran
- Department of Internal Medicine, School of Medicine, Universidad de Valparaiso, Vina del Mar, Chile
| | - Roberto Acosta-Dighero
- Cochrane Chile Associate Centre, Universidad de Valparaíso, Valparaíso, Chile
- School of Physiotherapy, Faculty of Health Sciences, Universidad San Sebastian, Santiago, Chile
| | - Shrikant I Bangdiwala
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Gabriel Rada
- Department of Internal Medicine and Evidence-Based Healthcare Program, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eva Madrid
- Interdisciplinary Centre for Health Studies CIESAL, Universidad de Valparaíso, Viña del Mar, Chile
- Cochrane Chile Associate Centre, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Cheung MM, Jahan N. Can Stem Cells Improve Left Ventricular Ejection Fraction in Heart Failure? A Literature Review of Skeletal Myoblasts and Bone Marrow-Derived Cells. Cureus 2020; 12:e11598. [PMID: 33364119 PMCID: PMC7752736 DOI: 10.7759/cureus.11598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 11/20/2020] [Indexed: 01/05/2023] Open
Abstract
Heart failure is a life-threatening condition that affects millions worldwide and is only expected to get worse with an ageing population. Current treatment regimens rely on medical therapy and heart transplantation as a last resort. Stem cells have been undergoing clinical trials worldwide as a hope for a new and safe clinical treatment. Skeletal myoblasts and bone marrow-derived stem cells are two types of stem cells being tested. The objective is to evaluate the efficacy of these two types of stem cells for heart failure therapy. Data were searched in PubMed using both regular and Medical Subject Heading (MeSH) keywords (stem cells, therapy, heart failure) and then filtered using inclusion/exclusion criteria (language, species, publication date, and age). In total, 31 research articles were reviewed (14 clinical trials, four randomized control trials, nine review articles, one case report, one comparative study, one systematic review, and one categorized as a systematic review and meta-analysis). Both skeletal myoblasts and bone marrow-derived stem cells showed mixed results in improving left ventricular ejection fraction in heart failure patients in the majority of studies. Larger studies need to be done to further investigate the efficacy of stem cells as a therapy for heart failure.
Collapse
Affiliation(s)
- Meghan M Cheung
- Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Nusrat Jahan
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
19
|
Frljak S, Poglajen G, Zemljic G, Cerar A, Haddad F, Terzic A, Vrtovec B. Larger End-Diastolic Volume Associates With Response to Cell Therapy in Patients With Nonischemic Dilated Cardiomyopathy. Mayo Clin Proc 2020; 95:2125-2133. [PMID: 33012343 PMCID: PMC7539130 DOI: 10.1016/j.mayocp.2020.02.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/13/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To investigate the association of left ventricular end-diastolic volume (LVEDV) and the response to cell therapy in patients with nonischemic dilated cardiomyopathy (NICM). PATIENTS AND METHODS Five-year registry data from 133 consecutive patients with NICM who underwent CD34+ cell treatment were analyzed. All patients received granulocyte-colony stimulating factor; CD34+ cells were collected by apheresis and delivered by transendocardial injections. Patients with baseline LVEDV less than 200 mL (group A; n=72) and patients with LVEDV 200 to 370 mL (group B; n=54) were included. Patients with LVEDV greater than 370 mL were excluded (n=7). Favorable ejection fraction response was pre-defined by improvement in left ventricular ejection fraction (LVEF) greater than or equal to 5% at 1 y post-cell therapy. RESULTS At baseline, groups A and B were comparable with regards to age (52±11 y in group A vs 53±10 y in group B; P=.95), sex (male: 79% vs 83%, respectively; P=.55), creatinine (1.07±0.28 mg/dL vs 1.03±0.21 mg/dL, respectively; P=.21), or N-terminal probrain natriuretic peptide (1454±1658 pg/mL vs 1589±1338 pg/mL, respectively; P=.80). Baseline LVEF was higher in group A (32.8±8.7%) than in group B (30.2±8.7%; P=.03). During follow-up, there were four deaths in group A (5.6%), and 2 in group B (3.7%, P=.63). At 1-year post-cell therapy, LVEDV decreased significantly in group B (-56±30 mL; P=.003), but not in group A (+12±97 mL; P=.13). On multivariate analysis, baseline LVEDV was an independent correlate of favorable response in LVEF to therapy (P=.02). CONCLUSION Larger LVEDV was associated with more pronounced increase in LVEF after transendocardial CD34+ cell therapy in NICM patients, informing target individuals with the highest likelihood of regenerative response. TRIAL REGISTRATION clinicaltrials.gov identifier: NCT02445534.
Collapse
Affiliation(s)
- Sabina Frljak
- Advanced Heart Failure and Transplantation Center, UMC Ljubljana, Slovenia
| | - Gregor Poglajen
- Advanced Heart Failure and Transplantation Center, UMC Ljubljana, Slovenia
| | - Gregor Zemljic
- Advanced Heart Failure and Transplantation Center, UMC Ljubljana, Slovenia
| | - Andraz Cerar
- Advanced Heart Failure and Transplantation Center, UMC Ljubljana, Slovenia
| | - Francois Haddad
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA
| | - Andre Terzic
- Center for Regenerative Medicine, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Bojan Vrtovec
- Advanced Heart Failure and Transplantation Center, UMC Ljubljana, Slovenia.
| |
Collapse
|
20
|
Poglajen G, Frljak S, Zemljič G, Cerar A, Okrajšek R, Šebeštjen M, Vrtovec B. Stem Cell Therapy for Chronic and Advanced Heart Failure. Curr Heart Fail Rep 2020; 17:261-270. [PMID: 32783146 DOI: 10.1007/s11897-020-00477-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to discuss recent advances in the field of cell therapy in patients with heart failure with reduced ejection fraction (HFrEF) of ischemic (iCMP) and nonischemic (dCMP) etiology, heart failure with preserved ejection fraction (HFpEF), and in advanced heart failure patients undergoing mechanical circulatory support (LVAD). RECENT FINDINGS In HFrEF patients (iCMP and dCMP cohorts), autologous and/or allogeneic cell therapy was shown to improve myocardial performance, patients' functional capacity, and neurohumoral activation. In HFpEF patient population, the concept of cell therapy in novel and remains largely unexplored. However, initial data are very encouraging and suggest at least a similar benefit in improvements of myocardial performance (also diastolic function of the left ventricle), exercise capacity, and neurohumoral activation. Recently, cell therapy was explored in the sickest population of advanced heart failure patients undergoing LVAD support also showing a potential benefit in promoting myocardial reverse remodeling and recovery. In the past decade, several cell therapy-based clinical trials showed promising results in various chronic and advanced heart failure patient cohorts. Future cell treatment strategies should aim for more personalized therapeutic approaches by defining optimal stem cell type or their combination, dose, and delivery method for an individual patient adjusted for patient's age and stage/duration of heart failure.
Collapse
Affiliation(s)
- Gregor Poglajen
- Department of Cardiology, Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Zaloška cesta 7, 1000, Ljubljana, Slovenia. .,Medical Faculty, University of Ljubljana, Ljubljana, Slovenia.
| | - Sabina Frljak
- Department of Cardiology, Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Zaloška cesta 7, 1000, Ljubljana, Slovenia
| | - Gregor Zemljič
- Department of Cardiology, Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Zaloška cesta 7, 1000, Ljubljana, Slovenia
| | - Andraž Cerar
- Department of Cardiology, Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Zaloška cesta 7, 1000, Ljubljana, Slovenia
| | - Renata Okrajšek
- Department of Cardiology, Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Zaloška cesta 7, 1000, Ljubljana, Slovenia
| | - Miran Šebeštjen
- Department of Cardiology, Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Zaloška cesta 7, 1000, Ljubljana, Slovenia
| | - Bojan Vrtovec
- Department of Cardiology, Advanced Heart Failure and Transplantation Center, University Medical Center Ljubljana, Zaloška cesta 7, 1000, Ljubljana, Slovenia.,Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
21
|
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
|
22
|
Affiliation(s)
- Bojan Vrtovec
- From the Advanced Heart Failure and Transplantation Center, Department of Cardiology, UMC Ljubljana, Slovenia (B.V.)
| | - Roberto Bolli
- Division of Cardiovascular Medicine, Institute of Molecular Cardiology, University of Louisville, KY (R.B.)
| |
Collapse
|
23
|
Diaz-Navarro R, Urrútia G, Cleland JGF, Poloni D, Villagran F, Bangdiwala S, Rada G, Madrid E. Stem cell therapy for dilated cardiomyopathy. Hippokratia 2019. [DOI: 10.1002/14651858.cd013433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Rienzi Diaz-Navarro
- Universidad de Valparaiso; Department of Internal Medicine, School of Medicine; Vina del Mar Chile
| | - Gerard Urrútia
- CIBER Epidemiología y Salud Pública (CIBERESP); Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau); Sant Antoni Maria Claret, 167 Pavilion 18 (D-53) Barcelona Catalonia Spain 08025
| | - John GF Cleland
- Imperial College London; National Heart and Lung Institute; London UK
| | - Daniel Poloni
- Universidad de Valparaiso; Department of Internal Medicine, School of Medicine; Vina del Mar Chile
| | - Francisco Villagran
- Universidad de Valparaiso; Department of Internal Medicine, School of Medicine; Vina del Mar Chile
| | - Shrikant Bangdiwala
- Collaborative Studies Coordinating Center; Department of Biostatistics, Gillings School of Global Public Health; Suite 203, Bank of America Center 137 E. Franklin Street Chapel Hill North Carolina USA 27514-4145
| | - Gabriel Rada
- Pontificia Universidad Católica de Chile; Department of Internal Medicine and Evidence-Based Healthcare Program, Faculty of Medicine; Lira 44, Decanato Primer piso Santiago Chile
| | - Eva Madrid
- Cochrane Centre School of Medicine Universidad de Valparaiso; Interdisciplinary Centre for Health Studies CIESAL; Viña del Mar Chile
- Universidad de Valparaiso; Chilean Cochrane Centre; Valparaiso Chile
| |
Collapse
|
24
|
Williams R. Circulation Research "In This Issue" Anthology. Circ Res 2019; 124:e123-e148. [PMID: 31170049 DOI: 10.1161/res.0000000000000275] [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: 11/16/2022]
|
25
|
Qin X, Han D, Wu JC. Molecular imaging of cardiac regenerative medicine. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2019. [DOI: 10.1016/j.cobme.2019.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|