1
|
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
|
2
|
A comprehensive, multiscale framework for evaluation of arrhythmias arising from cell therapy in the whole post-myocardial infarcted heart. Sci Rep 2019; 9:9238. [PMID: 31239508 PMCID: PMC6592890 DOI: 10.1038/s41598-019-45684-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 06/12/2019] [Indexed: 12/19/2022] Open
Abstract
Direct remuscularization approaches to cell-based heart repair seek to restore ventricular contractility following myocardial infarction (MI) by introducing new cardiomyocytes (CMs) to replace lost or injured ones. However, despite promising improvements in cardiac function, high incidences of ventricular arrhythmias have been observed in animal models of MI injected with pluripotent stem cell-derived cardiomyocytes (PSC-CMs). The mechanisms of arrhythmogenesis remain unclear. Here, we present a comprehensive framework for computational modeling of direct remuscularization approaches to cell therapy. Our multiscale 3D whole-heart modeling framework integrates realistic representations of cell delivery and transdifferentiation therapy modalities as well as representation of spatial distributions of engrafted cells, enabling simulation of clinical therapy and the prediction of emergent electrophysiological behavior and arrhythmogenensis. We employ this framework to explore how varying parameters of cell delivery and transdifferentiation could result in three mechanisms of arrhythmogenesis: focal ectopy, heart block, and reentry.
Collapse
|
3
|
Cell Therapy for Refractory Angina: A Reappraisal. Stem Cells Int 2017; 2017:5648690. [PMID: 29375624 PMCID: PMC5742462 DOI: 10.1155/2017/5648690] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/05/2017] [Indexed: 12/23/2022] Open
Abstract
Cardiac cell-based therapy has emerged as a novel therapeutic option for patients dealing with untreatable refractory angina (RA). However, after more than a decade of controlled studies, no definitive consensus has been reached regarding clinical efficacy. Although positive results in terms of surrogate endpoints have been suggested by early and phase II clinical studies as well as by meta-analyses, the more recent reports lacked the provision of definitive response in terms of hard clinical endpoints. Regrettably, pivotal trials designed to conclusively determine the efficacy of cell-based therapeutics in such a challenging clinical condition are therefore still missing. Considering this, a comprehensive reappraisal of cardiac cell-based therapy role in RA seems warranted and timely, since a number of crucial cell- and patient-related aspects need to be systematically analysed. As an example, the large variability in efficacy endpoint selection appears to be a limiting factor for the advancement of cardiac cell-based therapy in the field. This review will provide an overview of the key elements that may have influenced the results of cell-based trials in the context of RA, focusing in particular on the understanding at which the extent of angina-related endpoints may predict cell-based therapeutic efficacy.
Collapse
|
4
|
Abstract
Despite that advances in medical treatment and interventional procedures have reduced the mortality rate in patients with coronary artery disease, the number of patients with refractory myocardial ischemia and congestive heart failure is rapidly increasing. Experimental studies have demonstrated that bone marrow (BM) contains adult stem cells that can induce neovascularization and improve heart function in ischemic myocardium. Recent insights into the understanding of the mechanisms involved in proliferation, recruitment, mobilization, and incorporation of BM-derived stem cells into the myocardium and blood vessels have prompted development of cellular transplantation therapy for heart diseases refractory to conventional therapy. Initial preliminary clinical studies indicated potential clinical benefit of BM therapy in patients with acute myocardial infarction and chronic myocardial ischemia. Nevertheless, many obstacles remain, such as long-term safety and optimal timing and treatment strategies for BM cell therapy, and these issues need to be addressed in rationally designed, randomized clinical trials.
Collapse
Affiliation(s)
- Hung-Fat Tse
- Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China. hftse@ hkucc.hku.hk
| | | |
Collapse
|
5
|
Jimenez-Quevedo P, Gonzalez-Ferrer JJ, Sabate M, Garcia-Moll X, Delgado-Bolton R, Llorente L, Bernardo E, Ortega-Pozzi A, Hernandez-Antolin R, Alfonso F, Gonzalo N, Escaned J, Bañuelos C, Regueiro A, Marin P, Fernandez-Ortiz A, Neves BD, del Trigo M, Fernandez C, Tejerina T, Redondo S, Garcia E, Macaya C. Selected CD133+Progenitor Cells to Promote Angiogenesis in Patients With Refractory Angina. Circ Res 2014; 115:950-60. [DOI: 10.1161/circresaha.115.303463] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Pilar Jimenez-Quevedo
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Juan Jose Gonzalez-Ferrer
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Manel Sabate
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Xavier Garcia-Moll
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Roberto Delgado-Bolton
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Leopoldo Llorente
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Esther Bernardo
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Aranzazu Ortega-Pozzi
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Rosana Hernandez-Antolin
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Fernando Alfonso
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Nieves Gonzalo
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Javier Escaned
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Camino Bañuelos
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Ander Regueiro
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Pedro Marin
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Antonio Fernandez-Ortiz
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Barbara Das Neves
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Maria del Trigo
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Cristina Fernandez
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Teresa Tejerina
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Santiago Redondo
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Eulogio Garcia
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| | - Carlos Macaya
- From the Cardiology and Hematology Department, Hospital Clínico San Carlos, IdISSC, Madrid, Spain (P.J.-Q., J.J.G.-F., L.L., E.B., A.O.-P., R.H.-A., F.A., N.G., J.E., C.B., A.F.-O., B.D.N., M.d.T., E.G., C.M.); Cardiology and Hematology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain (M.S., A.R., P.M.); Cardiology Department, Hospital Sant Pau, Barcelona, Spain (X.G.-M.); Department of Radiology and Nuclear Medicine, San Pedro Hospital and Centre for Biomedical Research of La Rioja (CIBIR),
| |
Collapse
|
6
|
Ghodsizad A, Ruhparwar A, Bordel V, Mirsaidighazi E, Klein HM, Koerner MM, Karck M, El-Banayosy A. Clinical application of adult stem cells for therapy for cardiac disease. Cardiovasc Ther 2014; 31:323-34. [PMID: 23773460 DOI: 10.1111/1755-5922.12032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION Cardiovascular disease is a major cause of death worldwide. Different medical and surgical therapeutic options are well established, but a significant number of patients are not amenable to standard therapeutic options. Cell-based therapies after clinical application have shown different results in recent years. Here, we are giving a comprehensive overview on major available clinical data regarding cell therapy. BACKGROUND Cell-based therapies and tissue engineering provide new promising platforms to develop upcoming therapeutic options. Initial clinical trials were able to generate promising results. A variety of different stem cell types have been used for the clinical application. Different adult cardiac stem cells and progenitor cells, including mesenchymal, CD34(+) and CD133(+) autologous human bone marrow-derived stem cells (BMCs), human myoblasts, and peripheral blood-derived stem and progenitor cells (PBSCs) have been used for the therapy for end-stage heart failure. Future experiments will show the importance of novel cell populations and clarify the mechanism causing cell therapy-mediated observed effects. CONCLUSION Several clinical trials have reported on sole therapy, as well as combined application of autologous adult stem cells with conventional revascularization. The reported promising findings encourage further research in the field of the translational research.
Collapse
Affiliation(s)
- Ali Ghodsizad
- Heart and Vascular Institute, Milton S. Hershey Medical Center, College of Medicine, The Pennsylvania State University, Hershey, PA, USA; Department of Cardiac Surgery, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Pavo N, Charwat S, Nyolczas N, Jakab A, Murlasits Z, Bergler-Klein J, Nikfardjam M, Benedek I, Benedek T, Pavo IJ, Gersh BJ, Huber K, Maurer G, Gyöngyösi M. Cell therapy for human ischemic heart diseases: critical review and summary of the clinical experiences. J Mol Cell Cardiol 2014; 75:12-24. [PMID: 24998410 DOI: 10.1016/j.yjmcc.2014.06.016] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 05/23/2014] [Accepted: 06/26/2014] [Indexed: 12/24/2022]
Abstract
A decade ago, stem or progenitor cells held the promise of tissue regeneration in human myocardium, with the expectation that these therapies could rescue ischemic myocyte damage, enhance vascular density and rebuild injured myocardium. The accumulated evidence in 2014 indicates, however, that the therapeutic success of these cells is modest and the tissue regeneration involves much more complex processes than cell-related biologics. As the quest for the ideal cell or combination of cells continues, alternative cell types, such as resident cardiac cells, adipose-derived or phenotypic modified stem or progenitor cells have also been applied, with the objective of increasing both the number and the retention of the reparative cells in the myocardium. Two main delivery routes (intracoronary and percutaneous intramyocardial) of stem cells are currently used preferably for patients with recent acute myocardial infarction or ischemic cardiomyopathy. Other delivery modes, such as surgical or intravenous via peripheral veins or coronary sinus have also been utilized with less success. Due to the difficult recruitment of patients within conceivable timeframe into cardiac regenerative trials, meta-analyses of human cardiac cell-based studies have tried to gather sufficient number of subjects to present a statistical compelling statement, reporting modest success with a mean increase of 0.9-6.1% in left ventricular global ejection fraction. Additionally, nearly half of the long-term studies reported the disappearance of the initial benefit of this treatment. Beside further extensive efforts to increase the efficacy of currently available methods, pre-clinical experiments using new techniques such as tissue engineering or exploiting paracrine effect hold promise to regenerate injured human cardiac tissue.
Collapse
Affiliation(s)
- Noemi Pavo
- Department of Cardiology, Medical University of Vienna, Austria
| | - Silvia Charwat
- Department of Cardiology, Medical University of Vienna, Austria
| | - Noemi Nyolczas
- Department of Cardiology, Medical University of Vienna, Austria
| | - András Jakab
- Department of Biomedical Laboratory and Imaging Science, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsolt Murlasits
- Exercise Biochemistry Laboratory, The University of Memphis, Department of Health and Sport Sciences, Memphis, TN, USA
| | | | | | - Imre Benedek
- Department of Cardiology, University of Medicine and Pharmacy Tirgu Mures, Romania
| | - Teodora Benedek
- Department of Cardiology, University of Medicine and Pharmacy Tirgu Mures, Romania
| | - Imre J Pavo
- Department of Cardiology, Medical University of Vienna, Austria
| | - Bernard J Gersh
- Internal Medicine, Mayo Graduate School of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Kurt Huber
- 3(rd) Dept. Cardiology and Emergency Medicine, Wilhelminen hospital, Vienna, Austria
| | - Gerald Maurer
- Department of Cardiology, Medical University of Vienna, Austria
| | | |
Collapse
|
8
|
Hossne NA, Cruz E, Buffolo E, Coimbra ACTDSMD, Machado J, Goldenberg RCDS, Regazzi G, Azevedo S, Invitti AL, Rodrigues Branco JN, Rodrigues de Oliveira JS, Stolf NAG, Miller LW, Sanberg PR. Long-Term and Sustained Therapeutic Results of a Specific Promonocyte Cell Formulation in Refractory Angina: ReACT(®) (Refractory Angina Cell Therapy) Clinical Update and Cost-Effective Analysis. Cell Transplant 2014; 24:955-70. [PMID: 24819720 DOI: 10.3727/096368914x681595] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mononuclear stem cells have been studied for their potential in myocardial ischemia. In our previous published article, ReACT(®) phase I/II clinical trial, our results suggest that a certain cell population, promonocytes, directly correlated with the perceived angiogenesis in refractory angina patients. This study is ReACT's clinical update, assessing long-term sustained efficacy. The ReACT phase IIA/B noncontrolled, open-label, clinical trial enrolled 14 patients with refractory angina and viable ischemic myocardium, without ventricular dysfunction, who were not suitable for myocardial revascularization. The procedure consisted of direct myocardial injection of a specific mononuclear cell formulation, with a certain percentage of promonocytes, in a single series of multiple injections (24-90; 0.2 ml each) into specific areas of the left ventricle. Primary endpoints were Canadian Cardiovascular Society Angina Classification (CCSAC) improvement at the 12-month follow-up and ischemic area reduction (scintigraphic analysis) at the 12-month follow-up, in correlation with ReACT's formulation. A recovery index (for patients with more than 1 year follow-up) was created to evaluate CCSAC over time, until April 2011. Almost all patients presented progressive improvement in CCSAC beginning 3 months (p=0.002) postprocedure, which was sustained at the 12-month follow-up (p=0.002), as well as objective myocardium ischemic area reduction at 6 months (decrease of 15%, p<0.024) and 12 months (decrease of 100%, p<0.004) The recovery index (n=10) showed that the patients were graded less than CCSAC 4 for 73.9 ± 24.2% over a median follow-up time of 46.8 months. After characterization, ReACT's promonocyte concentration suggested a positive correlation with CCSAC improvement (r=-0.575, p=0.082). Quality of life (SF-36 questionnaire) improved significantly in almost all domains. Cost-effectiveness analysis showed decrease in angina-related direct costs. Refractory angina patients presented a sustained long-term improvement in CCSAC and myocardium ischemic areas after the procedure. The long-term follow-up and strong improvement in quality of life reinforce effectiveness. Promonocytes may play a key role in myocardial neoangiogenesis. ReACT dramatically decreased direct costs.
Collapse
Affiliation(s)
- Nelson Americo Hossne
- Cardiovascular Surgery Division, Surgery Department, Paulista School of Medicine, Federal University of São Paulo, São Paulo, SP, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Stem Cell Therapy Is a Promising Tool for Refractory Angina: A Meta-analysis of Randomized Controlled Trials. Can J Cardiol 2013; 29:908-14. [DOI: 10.1016/j.cjca.2012.12.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 11/19/2012] [Accepted: 12/04/2012] [Indexed: 12/26/2022] Open
|
10
|
Potential benefits of cell therapy in coronary heart disease. J Cardiol 2013; 62:267-76. [PMID: 23834957 DOI: 10.1016/j.jjcc.2013.05.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 05/21/2013] [Accepted: 05/24/2013] [Indexed: 12/31/2022]
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in the world. In recent years, there has been an increasing interest both in basic and clinical research regarding the field of cell therapy for coronary heart disease (CHD). Several preclinical models of CHD have suggested that regenerative properties of stem and progenitor cells might help restoring myocardial functions in the event of cardiac diseases. Here, we summarize different types of stem/progenitor cells that have been tested in experimental and clinical settings of cardiac regeneration, from embryonic stem cells to induced pluripotent stem cells. Then, we provide a comprehensive description of the most common cell delivery strategies with their major pros and cons and underline the potential of tissue engineering and injectable matrices to address the crucial issue of restoring the three-dimensional structure of the injured myocardial region. Due to the encouraging results from preclinical models, the number of clinical trials with cell therapy is continuously increasing and includes patients with CHD and congestive heart failure. Most of the already published trials have demonstrated safety and feasibility of cell therapies in these clinical conditions. Several studies have also suggested that cell therapy results in improved clinical outcomes. Numerous ongoing clinical trials utilizing this therapy for CHD will address fundamental issues concerning cell source and population utilized, as well as the use of imaging techniques to assess cell homing and survival, all factors that affect the efficacy of different cell therapy strategies.
Collapse
|
11
|
Hatt CR, Jain AK, Parthasarathy V, Lang A, Raval AN. MRI-3D ultrasound-X-ray image fusion with electromagnetic tracking for transendocardial therapeutic injections: in-vitro validation and in-vivo feasibility. Comput Med Imaging Graph 2013; 37:162-73. [PMID: 23561056 DOI: 10.1016/j.compmedimag.2013.03.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 03/12/2013] [Accepted: 03/14/2013] [Indexed: 11/17/2022]
Abstract
Myocardial infarction (MI) is one of the leading causes of death in the world. Small animal studies have shown that stem-cell therapy offers dramatic functional improvement post-MI. An endomyocardial catheter injection approach to therapeutic agent delivery has been proposed to improve efficacy through increased cell retention. Accurate targeting is critical for reaching areas of greatest therapeutic potential while avoiding a life-threatening myocardial perforation. Multimodal image fusion has been proposed as a way to improve these procedures by augmenting traditional intra-operative imaging modalities with high resolution pre-procedural images. Previous approaches have suffered from a lack of real-time tissue imaging and dependence on X-ray imaging to track devices, leading to increased ionizing radiation dose. In this paper, we present a new image fusion system for catheter-based targeted delivery of therapeutic agents. The system registers real-time 3D echocardiography, magnetic resonance, X-ray, and electromagnetic sensor tracking within a single flexible framework. All system calibrations and registrations were validated and found to have target registration errors less than 5 mm in the worst case. Injection accuracy was validated in a motion enabled cardiac injection phantom, where targeting accuracy ranged from 0.57 to 3.81 mm. Clinical feasibility was demonstrated with in-vivo swine experiments, where injections were successfully made into targeted regions of the heart.
Collapse
Affiliation(s)
- Charles R Hatt
- University of Wisconsin - Madison, College of Engineering, Department of Biomedical Engineering, 1415 Engineering Drive, Madison, WI 53706, USA.
| | | | | | | | | |
Collapse
|
12
|
Abdelwahid E, Siminiak T, Guarita-Souza LC, Teixeira de Carvalho KA, Gallo P, Shim W, Condorelli G. Stem cell therapy in heart diseases: a review of selected new perspectives, practical considerations and clinical applications. Curr Cardiol Rev 2013; 7:201-12. [PMID: 22758618 PMCID: PMC3263484 DOI: 10.2174/157340311798220502] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2010] [Revised: 01/31/2011] [Accepted: 02/03/2011] [Indexed: 12/13/2022] Open
Abstract
Degeneration of cardiac tissues is considered a major cause of mortality in the western world and is expected to be a greater problem in the forthcoming decades. Cardiac damage is associated with dysfunction and irreversible loss of cardiomyocytes. Stem cell therapy for ischemic heart failure is very promising approach in cardiovascular medicine. Initial trials have indicated the ability of cardiomyocytes to regenerate after myocardial injury. These preliminary trials aim to translate cardiac regeneration strategies into clinical practice. In spite of advances, current therapeutic strategies to ischemic heart failure remain very limited. Moreover, major obstacles still need to be solved before stem cell therapy can be fully applied. This review addresses the current state of research and experimental data regarding embryonic stem cells (ESCs), myoblast transplantation, histological and functional analysis of transplantation of co-cultured myoblasts and mesenchymal stem cells, as well as comparison between mononuclear and mesenchymal stem cells in a model of myocardium infarction. We also discuss how research with stem cell transplantation could translate to improvement of cardiac function.
Collapse
Affiliation(s)
- Eltyeb Abdelwahid
- CBRC, Massachusetts General Hospital/Harvard Medical School, Charlestown, Massachusetts 02129, USA.
| | | | | | | | | | | | | |
Collapse
|
13
|
Sheng CC, Zhou L, Hao J. Current stem cell delivery methods for myocardial repair. BIOMED RESEARCH INTERNATIONAL 2012; 2013:547902. [PMID: 23509740 PMCID: PMC3591183 DOI: 10.1155/2013/547902] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 09/14/2012] [Indexed: 12/25/2022]
Abstract
Heart failure commonly results from an irreparable damage due to cardiovascular diseases (CVDs), the leading cause of morbidity and mortality in the United States. In recent years, the rapid advancements in stem cell research have garnered much praise for paving the way to novel therapies in reversing myocardial injuries. Cell types currently investigated for cellular delivery include embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and adult stem cell lineages such as skeletal myoblasts, bone-marrow-derived stem cells (BMSCs), mesenchymal stem cells (MSCs), and cardiac stem cells (CSCs). To engraft these cells into patients' damaged myocardium, a variety of approaches (intramyocardial, transendocardial, transcoronary, venous, intravenous, intracoronary artery and retrograde venous administrations and bioengineered tissue transplantation) have been developed and explored. In this paper, we will discuss the pros and cons of these delivery modalities, the current state of their therapeutic potentials, and a multifaceted evaluation of their reported clinical feasibility, safety, and efficacy. While the issues of optimal delivery approach, the best progenitor stem cell type, the most effective dose, and timing of administration remain to be addressed, we are highly optimistic that stem cell therapy will provide a clinically viable option for myocardial regeneration.
Collapse
Affiliation(s)
- Calvin C. Sheng
- School of Medicine, Vanderbilt University, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Li Zhou
- School of Medicine, Vanderbilt University, 2220 Pierce Avenue, Nashville, TN 37232, USA
| | - Jijun Hao
- College of Veterinary Medicine, Western University of Health Sciences, 309 East Second Street, Pomona, CA 91766, USA
| |
Collapse
|
14
|
Giordano C, Kuraitis D, Beanlands RSB, Suuronen EJ, Ruel M. Cell-based vasculogenic studies in preclinical models of chronic myocardial ischaemia and hibernation. Expert Opin Biol Ther 2012; 13:411-28. [PMID: 23256710 DOI: 10.1517/14712598.2013.748739] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Coronary artery disease commonly leads to myocardial ischaemia and hibernation. Relevant preclinical models of these conditions are essential to evaluate new therapeutic options such as cell-based vasculogenic therapies. AREAS COVERED In this article, the authors first review basic concepts of myocardial ischaemia/hibernation and relevant techniques to assess myocardial viability. Then, preclinical models of chronic myocardial ischaemia and hibernation, induced by devices such as ameroid constrictors, Delrin stenosis, hydraulic occluders, and coils/stents are described. Lastly, the authors discuss cell-based vasculogenic therapy, and summarise studies conducted in large animal models of chronic myocardial ischaemia and hibernation. EXPERT OPINION Approximately one-third of patients with viable myocardium do not undergo revascularisation; however, this population is at high risk for cardiac events and would surely benefit from effective cell-based therapy. Because of the modest benefits in clinical studies, preclinical models accurately representing clinical myocardial ischemia/hibernation are necessary to better understand and appropriately direct regenerative therapy research.
Collapse
Affiliation(s)
- Céline Giordano
- University of Ottawa Heart Institute, Division of Cardiac Surgery, 40 Ruskin Street, Suite 3403, Ottawa, Ontario, K1Y 4W7, Canada
| | | | | | | | | |
Collapse
|
15
|
Rodrigo SF, van Ramshorst J, Beeres SL, Al Younis I, Dibbets-Schneider P, de Roos A, Fibbe WE, Zwaginga JJ, Schalij MJ, Bax JJ, Atsma DE. Intramyocardial injection of bone marrow mononuclear cells in chronic myocardial ischemia patients after previous placebo injection improves myocardial perfusion and anginal symptoms: an intra-patient comparison. Am Heart J 2012; 164:771-8. [PMID: 23137509 DOI: 10.1016/j.ahj.2012.08.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 08/24/2012] [Indexed: 12/21/2022]
Abstract
BACKGROUND We recently demonstrated in a randomized, double-blind, placebo-controlled trial that intramyocardial bone marrow cell (BMC) injection is associated with improvements in myocardial perfusion and anginal symptoms in chronic myocardial ischemia patients. In the present study the results of the crossover phase of this trial, in which patients previously treated with placebo received autologous BMC injections are reported. This allows a unique intra-patient comparison on the effect of BMC versus placebo injection with elimination of patient-related confounding factors. METHODS In 16 patients (14 male, 64 ± 10 years), who previously received intramyocardial placebo injections in the setting of a randomized trial, 100 × 10(6) BMC were injected using the NOGA-system. Canadian Cardiovascular Society angina score and quality of life were evaluated at baseline, 3 and 6 months. Tc-99m single photon emission computed tomography and magnetic resonance imaging were performed at baseline and 3 months to assess myocardial perfusion and left ventricular (LV) function. RESULTS Canadian Cardiovascular Society score and quality of life improved significantly after BMC injection as compared to placebo (P = 0.01 and P = 0.02, respectively). Single photon emission computed tomography revealed a significant greater improvement (P = 0.03) in summed stress score after BMC injection as compared to placebo. LV end-systolic volume significantly decreased after BMC injection but not after placebo injection. LV end-diastolic volume and LV ejection fraction did not change. CONCLUSION Intramyocardial BMC injection in patients with chronic myocardial ischemia who previously received intramyocardial placebo treatment resulted in significant improvement in angina symptoms and myocardial perfusion. These results confirm the outcome of our previously reported randomized trial.
Collapse
Affiliation(s)
- Sander F Rodrigo
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Szöke K, Brinchmann JE. Concise review: therapeutic potential of adipose tissue-derived angiogenic cells. Stem Cells Transl Med 2012. [PMID: 23197872 DOI: 10.5966/sctm.2012-0069] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Inadequate blood supply to tissues is a leading cause of morbidity and mortality today. Ischemic symptoms caused by obstruction of arterioles and capillaries are currently not treatable by vessel replacement or dilatation procedures. Therapeutic angiogenesis, the treatment of tissue ischemia by promoting the proliferation of new blood vessels, has recently emerged as one of the most promising therapies. Neovascularization is most often attempted by introduction of angiogenic cells from different sources. Emerging evidence suggests that adipose tissue (AT) is an excellent reservoir of autologous cells with angiogenic potential. AT yields two cell populations of importance for neovascularization: AT-derived mesenchymal stromal cells, which likely act predominantly as pericytes, and AT-derived endothelial cells (ECs). In this concise review we discuss different physiological aspects of neovascularization, briefly present cells isolated from the blood and bone marrow with EC properties, and then discuss isolation and cell culture strategies, phenotype, functional capabilities, and possible therapeutic applications of angiogenic cells obtained from AT.
Collapse
|
17
|
Wei F, Wang TZ, Zhang J, Yuan ZY, Tian HY, Ni YJ, Zhuo XZ, Han K, Liu Y, Lu Q, Bai HY, Ma AQ. Mesenchymal stem cells neither fully acquire the electrophysiological properties of mature cardiomyocytes nor promote ventricular arrhythmias in infarcted rats. Basic Res Cardiol 2012; 107:274. [PMID: 22744762 DOI: 10.1007/s00395-012-0274-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 04/30/2012] [Accepted: 05/25/2012] [Indexed: 12/28/2022]
Abstract
Electrophysiological properties of implanted mesenchymal stem cells (MSCs) in infarcted hearts remain unclear, and their proarrhythmic effect is still controversial. The intent of this study was to investigate electrophysiological properties and proarrhythmic effects of MSCs in infarcted hearts. Rats were randomly divided into a myocardial infarction (MI) group, a MI-DMEM group (received DMEM medium injection) and MI-MSCs group (received MSCs injection). Survival analysis showed that the majority of engrafted MSCs died at day 9 after transplantation. Engrafted MSCs expressed cardiac markers (MYH, cTnI, Cx43), cardiac ion channel genes (Kv1.4, Kv4.2 and Kir2.1) and potassium currents (I (to), I (K1) and I (KDR)), but did not express Nav1.5, Cav1.2, Na(+) current and Ca(2+) current during their survival. When induced by Ca(2+), implanted MSCs exhibited no contraction ability after being isolated from the heart. Following 8-week electrocardiography monitoring, the cumulative occurrence of ventricular arrhythmias (VAs) was not different among the three groups. However, the prolonged QRS duration in infarcted rats without VAs was significantly decreased in the MI-MSCs group compared with the other two groups. The inducibility of VAs in the MI-MSCs group was much lower than that in the MI and MI-DMEM groups (41.20 vs. 86.67 % and 92.86 %; P < 0.0125). The ventricular effective refractory period in MI-MSCs group was prolonged in comparison with that in the MI and MI-DMEM groups (56.0 ± 8.8 vs. 47.7 ± 8.8 ms and 45.7 ± 6.2 ms; P < 0.01). These results demonstrate that MSCs do not acquire the electrophysiological properties of mature cardiomyocytes during the survival period in the infarcted hearts. However, they can alleviate the electrical vulnerability and do not promote ventricular arrhythmias.
Collapse
Affiliation(s)
- Feng Wei
- Department of Cardiovascular Medicine, First Affiliated Hospital of the Xi'an Jiaotong University School of Medicine, Shaanxi, 710061, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Perin EC, Willerson JT, Pepine CJ, Henry TD, Ellis SG, Zhao DX, Silva GV, Lai D, Thomas JD, Kronenberg MW, Martin AD, Anderson RD, Traverse JH, Penn MS, Anwaruddin S, Hatzopoulos AK, Gee AP, Taylor DA, Cogle CR, Smith D, Westbrook L, Chen J, Handberg E, Olson RE, Geither C, Bowman S, Francescon J, Baraniuk S, Piller LB, Simpson LM, Loghin C, Aguilar D, Richman S, Zierold C, Bettencourt J, Sayre SL, Vojvodic RW, Skarlatos SI, Gordon DJ, Ebert RF, Kwak M, Moyé LA, Simari RD. Effect of transendocardial delivery of autologous bone marrow mononuclear cells on functional capacity, left ventricular function, and perfusion in chronic heart failure: the FOCUS-CCTRN trial. JAMA 2012; 307:1717-26. [PMID: 22447880 PMCID: PMC3600947 DOI: 10.1001/jama.2012.418] [Citation(s) in RCA: 342] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CONTEXT Previous studies using autologous bone marrow mononuclear cells (BMCs) in patients with ischemic cardiomyopathy have demonstrated safety and suggested efficacy. OBJECTIVE To determine if administration of BMCs through transendocardial injections improves myocardial perfusion, reduces left ventricular end-systolic volume (LVESV), or enhances maximal oxygen consumption in patients with coronary artery disease or LV dysfunction, and limiting heart failure or angina. DESIGN, SETTING, AND PATIENTS A phase 2 randomized double-blind, placebo-controlled trial of symptomatic patients (New York Heart Association classification II-III or Canadian Cardiovascular Society classification II-IV) with a left ventricular ejection fraction of 45% or less, a perfusion defect by single-photon emission tomography (SPECT), and coronary artery disease not amenable to revascularization who were receiving maximal medical therapy at 5 National Heart, Lung, and Blood Institute-sponsored Cardiovascular Cell Therapy Research Network (CCTRN) sites between April 29, 2009, and April 18, 2011. INTERVENTION Bone marrow aspiration (isolation of BMCs using a standardized automated system performed locally) and transendocardial injection of 100 million BMCs or placebo (ratio of 2 for BMC group to 1 for placebo group). MAIN OUTCOME MEASURES Co-primary end points assessed at 6 months: changes in LVESV assessed by echocardiography, maximal oxygen consumption, and reversibility on SPECT. Phenotypic and functional analyses of the cell product were performed by the CCTRN biorepository core laboratory. RESULTS Of 153 patients who provided consent, a total of 92 (82 men; average age: 63 years) were randomized (n = 61 in BMC group and n = 31 in placebo group). Changes in LVESV index (-0.9 mL/m(2) [95% CI, -6.1 to 4.3]; P = .73), maximal oxygen consumption (1.0 [95% CI, -0.42 to 2.34]; P = .17), and reversible defect (-1.2 [95% CI, -12.50 to 10.12]; P = .84) were not statistically significant. There were no differences found in any of the secondary outcomes, including percent myocardial defect, total defect size, fixed defect size, regional wall motion, and clinical improvement. CONCLUSION Among patients with chronic ischemic heart failure, transendocardial injection of autologous BMCs compared with placebo did not improve LVESV, maximal oxygen consumption, or reversibility on SPECT. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT00824005.
Collapse
Affiliation(s)
| | | | | | - Timothy D. Henry
- Minneapolis Heart Institute at Abbott Northwestern Hospital, Minneapolis, Minnesota
- University of Minnesota School of Medicine, Minneapolis
| | | | - David X.M. Zhao
- Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - Dejian Lai
- The University of Texas School of Public Health, Houston
| | | | | | - A. Daniel Martin
- University of Florida College of Public Health and Health Professions, Gainesville
| | | | - Jay H. Traverse
- Minneapolis Heart Institute at Abbott Northwestern Hospital, Minneapolis, Minnesota
- University of Minnesota School of Medicine, Minneapolis
| | | | - Saif Anwaruddin
- Penn Heart and Vascular Hospital of the University of Pennsylvania, Philadelphia
| | | | | | | | | | - Deirdre Smith
- Texas Heart Institute, St. Luke’s Episcopal Hospital, Houston
| | | | - James Chen
- Texas Heart Institute, St. Luke’s Episcopal Hospital, Houston
| | | | - Rachel E. Olson
- Minneapolis Heart Institute at Abbott Northwestern Hospital, Minneapolis, Minnesota
| | | | - Sherry Bowman
- Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Judy Francescon
- Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Sarah Baraniuk
- The University of Texas School of Public Health, Houston
| | | | | | | | | | | | | | | | | | | | | | - David J. Gordon
- National Heart, Lung and Blood Institute, Bethesda, Maryland
| | - Ray F. Ebert
- National Heart, Lung and Blood Institute, Bethesda, Maryland
| | - Minjung Kwak
- National Heart, Lung and Blood Institute, Bethesda, Maryland
| | - Lemuel A. Moyé
- The University of Texas School of Public Health, Houston
| | | |
Collapse
|
19
|
Perin EC, Silva GV, Zheng Y, Gahremanpour A, Canales J, Patel D, Fernandes MR, Keller LH, Quan X, Coulter SA, Moore WH, Herlihy JP, Willerson JT. Randomized, double-blind pilot study of transendocardial injection of autologous aldehyde dehydrogenase-bright stem cells in patients with ischemic heart failure. Am Heart J 2012; 163:415-21, 421.e1. [PMID: 22424012 DOI: 10.1016/j.ahj.2011.11.020] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 11/30/2011] [Indexed: 01/19/2023]
Abstract
BACKGROUND The optimal type of stem cell for use in patients with ischemic heart disease has not been determined. A primitive population of bone marrow-derived hematopoietic cells has been isolated by the presence of the enzyme aldehyde dehydrogenase and comprises a multilineage mix of stem and progenitor cells. Aldehyde dehydrogenase-bright (ALDH(br)) cells have shown promise in promoting angiogenesis and providing perfusion benefits in preclinical ischemia studies. We hypothesize that ALDH(br) cells may be beneficial in treating ischemic heart disease and thus conducted the first randomized, controlled, double-blind study to assess the safety of the transendocardial injection of autologous ALDH(br) cells isolated from the bone marrow in patients with advanced ischemic heart failure. METHODS Aldehyde dehydrogenase-bright cells were isolated from patients' bone marrow on the basis of the expression of a functional (aldehyde dehydrogenase) marker. We enrolled 20 patients (treatment, n = 10; control, n = 10). Safety (primary end point) and efficacy (secondary end point) were assessed at 6 months. RESULTS No major adverse cardiovascular or cerebrovascular events occurred in ALDH(br)-treated patients in the periprocedural period (up to 1 month); electromechanical mapping-related ventricular tachycardia (n = 2) and fibrillation (n = 1) occurred in control patients. Aldehyde dehydrogenase-bright-treated patients showed a significant decrease in left ventricular end-systolic volume at 6 months (P = .04) and a trend toward improved maximal oxygen consumption. The single photon emission computed tomography delta analysis showed a trend toward significant improvement in reversibility in cell-treated patients (P = .053). CONCLUSIONS We provide preliminary evidence that treatment with the novel cell population, ALDH(br) cells, is safe and may provide perfusion and functional benefits in patients with chronic myocardial ischemia.
Collapse
Affiliation(s)
- Emerson C Perin
- Stem Cell Center at the Texas Heart Institute at St Luke's Episcopal Hospital, Houston, TX, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Cell delivery in cardiac regenerative therapy. Ageing Res Rev 2012; 11:32-40. [PMID: 21736956 DOI: 10.1016/j.arr.2011.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 06/09/2011] [Accepted: 06/15/2011] [Indexed: 01/16/2023]
Abstract
There is a growing interest in the clinical application of stem cells as a novel therapeutic approach for treatment of myocardial infarction and prevention of subsequent heart failure. Transplanted stem cells improve cardiac functions through multiple mechanisms, which include but are not limited to promoting angiogenesis, replacing dead cardiomyocytes, modulating cardiac remodeling. Most of the results obtained so far are exciting and very promising, spawning an increasing number of clinical trials recently. However, many problems still remain to be resolved such as the best delivery method for transplantation of cells to the injured myocardium and the issue of how to optimize the delivery of targeted cells is of exceptional clinical relevance. In this review, we focus on the different delivery strategies in cardiac regenerative therapy, as well as provide a brief overview of current clinical trials utilizing cell-based therapy in patients with ischemic heart disease.
Collapse
|
21
|
|
22
|
Tuma J, Fernández-Viña R, Carrasco A, Castillo J, Cruz C, Carrillo A, Ercilla J, Yarleque C, Cunza J, Henry TD, Patel AN. Safety and feasibility of percutaneous retrograde coronary sinus delivery of autologous bone marrow mononuclear cell transplantation in patients with chronic refractory angina. J Transl Med 2011; 9:183. [PMID: 22029669 PMCID: PMC3215661 DOI: 10.1186/1479-5876-9-183] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Accepted: 10/26/2011] [Indexed: 12/19/2022] Open
Abstract
Background Chronic refractory angina is a challenging clinical problem with limited treatment options. The results of early cardiovascular stem cell trials using ABMMC have been promising but have utilized intracoronary or intramyocardial delivery. The goal of the study was to evaluate the safety and early efficacy of autologous bone marrow derived mononuclear cells (ABMMC) delivered via percutaneous retrograde coronary sinus perfusion (PRCSP) to treat chronic refractory angina (CRA). Methods From May 2005 to October 2006, 14 patients, age 68 +/- 20 years old, with CRA and ischemic stress-induced myocardial segments assessed by SPECT received a median 8.19*108 ± 4.3*108 mononuclear and 1.65*107 ± 1.42*107 CD34+ cells by PRCSP.. Results ABMMC delivery was successful in all patients with no arrhythmias, elevated cardiac enzymes or complications related to the delivery. All but one patient improved by at least one Canadian Cardiovascular Society class at 2 year follow-up compared to baseline (p < 0.001). The median baseline area of ischemic myocardium by SPECT of 38.2% was reduced to 26.5% at one year and 23.5% at two years (p = 0.001). The median rest left ventricular ejection fraction by SPECT at baseline was 31.2% and improved to 35.5% at 2 year follow up (p = 0.019). Conclusions PRCSP should be considered as an alternative method of delivery for cell therapy with the ability to safely deliver large number of cells regardless of coronary anatomy, valvular disease or myocardial dysfunction. The clinical improvement in angina, myocardial perfusion and function in this phase 1 study is encouraging and needs to be confirmed in randomized placebo controlled trials.
Collapse
Affiliation(s)
- Jorge Tuma
- Division of Interventional Cardiology and Regenerative Medicine, Clínica Maisón de Santé, Lima, Peru
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Psaltis PJ, Simari RD, Rodriguez-Porcel M. Emerging roles for integrated imaging modalities in cardiovascular cell-based therapeutics: a clinical perspective. Eur J Nucl Med Mol Imaging 2011; 39:165-81. [PMID: 21901381 DOI: 10.1007/s00259-011-1925-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 08/18/2011] [Indexed: 12/20/2022]
Abstract
Despite preclinical promise, the progress of cell-based therapy to clinical cardiovascular practice has been slowed by several challenges and uncertainties that have been highlighted by the conflicting results of human trials. Most telling has been the revelation that current strategies fall short of achieving sufficient retention and engraftment of cells to meet the ambitious objective of myocardial regeneration. This has sparked novel research into the refinement of cell biology and delivery to overcome these shortcomings. Within this context, molecular imaging has emerged as a valuable tool for providing noninvasive surveillance of cell fate in vivo. Direct and indirect labelling of cells can be coupled with clinically relevant imaging modalities, such as radionuclide single photon emission computed tomography and positron emission tomography, and magnetic resonance imaging, to assess their short- and long-term distributions, along with their viability, proliferation and functional interaction with the host myocardium. This review details the strengths and limitations of the different cell labelling and imaging techniques and their potential application to the clinical realm. We also consider the broader, multifaceted utility of imaging throughout the cell therapy process, providing a discussion of its considerable value during cell delivery and its importance during the evaluation of cardiac outcomes in clinical studies.
Collapse
Affiliation(s)
- Peter J Psaltis
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | | |
Collapse
|
24
|
Immediate Intramyocardial Bone Marrow-Derived Mononuclear Cells Implantation in Minipig Myocardium After Permanent Coronary Artery Ligation. Invest Radiol 2011; 46:495-503. [DOI: 10.1097/rli.0b013e318214a63f] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
25
|
A randomized study of transendocardial injection of autologous bone marrow mononuclear cells and cell function analysis in ischemic heart failure (FOCUS-HF). Am Heart J 2011; 161:1078-87.e3. [PMID: 21641354 DOI: 10.1016/j.ahj.2011.01.028] [Citation(s) in RCA: 147] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Accepted: 01/13/2011] [Indexed: 01/12/2023]
Abstract
BACKGROUND Autologous bone marrow mononuclear cell (ABMMNC) therapy has shown promise in patients with heart failure (HF). Cell function analysis may be important in interpreting trial results. METHODS In this prospective study, we evaluated the safety and efficacy of the transendocardial delivery of ABMMNCs in no-option patients with chronic HF. Efficacy was assessed by maximal myocardial oxygen consumption, single photon emission computed tomography, 2-dimensional echocardiography, and quality-of-life assessment (Minnesota Living with Heart Failure and Short Form 36). We also characterized patients' bone marrow cells by flow cytometry, colony-forming unit, and proliferative assays. RESULTS Cell-treated (n = 20) and control patients (n = 10) were similar at baseline. The procedure was safe; adverse events were similar in both groups. Canadian Cardiovascular Society angina score improved significantly (P = .001) in cell-treated patients, but function was not affected. Quality-of-life scores improved significantly at 6 months (P = .009 Minnesota Living with Heart Failure and P = .002 physical component of Short Form 36) over baseline in cell-treated but not control patients. Single photon emission computed tomography data suggested a trend toward improved perfusion in cell-treated patients. The proportion of fixed defects significantly increased in control (P = .02) but not in treated patients (P = .16). Function of patients' bone marrow mononuclear cells was severely impaired. Stratifying cell results by age showed that younger patients (≤60 years) had significantly more mesenchymal progenitor cells (colony-forming unit fibroblasts) than patients >60 years (20.16 ± 14.6 vs 10.92 ± 7.8, P = .04). Furthermore, cell-treated younger patients had significantly improved maximal myocardial oxygen consumption (15 ± 5.8, 18.6 ± 2.7, and 17 ± 3.7 mL/kg per minute at baseline, 3 months, and 6 months, respectively) compared with similarly aged control patients (14.3 ± 2.5, 13.7 ± 3.7, and 14.6 ± 4.7 mL/kg per minute, P = .04). CONCLUSIONS ABMMNC therapy is safe and improves symptoms, quality of life, and possibly perfusion in patients with chronic HF.
Collapse
|
26
|
Gyöngyösi M, Dib N. Diagnostic and prognostic value of 3D NOGA mapping in ischemic heart disease. Nat Rev Cardiol 2011; 8:393-404. [DOI: 10.1038/nrcardio.2011.64] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
27
|
van Ramshorst J, Rodrigo SF, Schalij MJ, Beeres SLMA, Bax JJ, Atsma DE. Bone marrow cell injection for chronic myocardial ischemia: the past and the future. J Cardiovasc Transl Res 2011; 4:182-91. [PMID: 21213093 PMCID: PMC3047688 DOI: 10.1007/s12265-010-9249-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 11/17/2010] [Indexed: 01/12/2023]
Abstract
Intramyocardial bone marrow cell injection is currently being investigated as a new therapeutic option for the treatment of chronic myocardial ischemia. Experimental studies and early phase clinical trials established a favorable safety profile of this approach and suggested that bone marrow cell injection was associated with clinical and functional improvements. Recently, a randomized, double-blind, placebo-controlled trial demonstrated that intramyocardial bone marrow cell injection was associated with beneficial effects on myocardial perfusion and anginal symptoms. However, the mechanisms by which bone marrow cells may improve myocardial perfusion are only partially understood, and several issues remain to be addressed. This review aims to provide a summary of the current experience with bone marrow cell therapy as a novel treatment option for patients with chronic myocardial ischemia. Therefore, the most frequently used cell types will be reviewed along with the mechanisms through which bone marrow cells may improve myocardial perfusion and function. In addition, possible routes of delivery are compared, and the results of currently available experimental and clinical studies are discussed.
Collapse
Affiliation(s)
- Jan van Ramshorst
- Department of Cardiology, Leiden University Medical Centre, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Sander F. Rodrigo
- Department of Cardiology, Leiden University Medical Centre, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Martin J. Schalij
- Department of Cardiology, Leiden University Medical Centre, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Saskia L. M. A. Beeres
- Department of Cardiology, Leiden University Medical Centre, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Jeroen J. Bax
- Department of Cardiology, Leiden University Medical Centre, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Douwe E. Atsma
- Department of Cardiology, Leiden University Medical Centre, P.O. Box 9600, 2300 RC Leiden, The Netherlands
- Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
28
|
Combination stem cell therapy for the treatment of medically refractory coronary ischemia: a Phase I study. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2010; 12:29-34. [PMID: 21241969 DOI: 10.1016/j.carrev.2010.01.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 01/04/2010] [Accepted: 01/07/2010] [Indexed: 11/20/2022]
Abstract
PURPOSE Infusion of a source of endothelial progenitor cells (EPC) into the ischemic myocardium is emerging as a promising therapy for coronary ischemia, probably mediated by the formation of new blood vessels. Studies have shown that while the procedure is safe and feasible, efficacy results are contentious. The investigators hypothesized that the infusion of a combination cell product consisting of a source of EPC and mesenchymal stem cells (MSC) is safe and promotes the formation of more stable and mature blood vessels resulting in improved clinical outcomes. METHODS Ten patients with stable angina pectoris (class III to IV) on maximal medical therapy were included. All patients had ≥ 70% stenosis in at least one coronary artery, and none was considered a candidate for percutaneous coronary intervention or coronary artery bypass graft. End points were feasibility and safety of intracoronary infusion of the combination cell product and assessment of myocardial ischemia, left ventricular ejection fraction (LVEF), and quality of life at 6 months postinfusion. RESULTS Six months after cell infusion there were no adverse clinical events. Functional cardiac evaluation during the same period showed significant improvements in LVEF (average increase: 11%, P = .02) and myocardial ischemia (average decrease: 1.8 fold, P = .02). Additionally, all patients described significant improvements in quality of life. CONCLUSIONS Despite the inherent limitations associated with a Phase I clinical trial, this study demonstrates that the intracoronary infusion of the combination cell product is feasible and safe and also insinuates that this form of therapy may be beneficial.
Collapse
|
29
|
Abstract
In patients with ischemic heart disease, the goal of cell therapy is to improve perfusion and function of the damaged heart muscle. For this review, we selected articles that reported the findings from the major clinical studies of cardiovascular stem cell therapy in patients with chronic ischemic heart disease. Because of the current status of development of clinical investigation in this field, all relevant studies were included. Initial clinical trials have shown that adult cell-based therapy is safe and may improve the quality of life and the functional status of patients with chronic myocardial ischemia. Adult bone marrow mononuclear cells have been most frequently used in cardiac cell therapy trials to date, but new cell types are now being assessed in both preclinical and clinical studies. Although not well defined, mechanisms underlying the benefits associated with cell therapy are most likely multiple and include a paracrine effect. Cell therapy in patients with chronic ischemic heart disease has been shown to be safe and feasible. Initial data have shown that cell therapy with autologous bone marrow cells is associated with modest functional improvements. This finding needs to be confirmed in subsequent phase 2 and 3 trials.
Collapse
Affiliation(s)
- Emerson C Perin
- Stem Cell Center, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, TX, USA.
| | | |
Collapse
|
30
|
Non-surgical stem cell delivery strategies and in vivo cell tracking to injured myocardium. Int J Cardiovasc Imaging 2010; 27:367-83. [PMID: 20577813 PMCID: PMC3092059 DOI: 10.1007/s10554-010-9658-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Accepted: 06/11/2010] [Indexed: 01/10/2023]
Abstract
Heart failure is a major economic and public health problem. Despite the recent advances in drug therapy and coronary revascularization, the lost cardiomyocytes due to necrosis and apoptosis are not replaced by new myocardial tissue. Cell therapy is an interesting therapeutic option as it potentially improves contractility and restores regional ventricular function. Early clinical data demonstrated that cell transplantation, mainly delivered through non-surgical methods, is safe and feasible. However, several important issues need to be elucidated. This includes, next to determining the best cell type, the optimal delivery strategy, the biodistribution and the survival of implanted stem cells after transplantation. In this view, pre-clinical animal experiments are indispensable. Reporter genes, magnetic or radioactive labeling of stem cells have been developed to observe the fate and the distribution of transplanted cells using non-invasive imaging techniques. Several studies have demonstrated that these direct and non-direct labeling techniques may become an important tool in cell therapy. Integration of cell delivery and cell tracking will probably be a key for the success of cell therapy in patients. This review will provide a comprehensive overview on the various cell tracking and non-surgical cell delivery techniques, which are highly important in view of experimental and clinical studies.
Collapse
|
31
|
Chan CWS, Kwong YL, Kwong RY, Lau CP, Tse HF. Improvement of myocardial perfusion reserve detected by cardiovascular magnetic resonance after direct endomyocardial implantation of autologous bone marrow cells in patients with severe coronary artery disease. J Cardiovasc Magn Reson 2010; 12:6. [PMID: 20100336 PMCID: PMC2845579 DOI: 10.1186/1532-429x-12-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Accepted: 01/25/2010] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Recent studies suggested that bone marrow (BM) cell implantation in patients with severe chronic coronary artery disease (CAD) resulted in modest improvement in symptoms and cardiac function. This study sought to investigate the functional changes that occur within the chronic human ischaemic myocardium after direct endomyocardial BM cells implantation by cardiovascular magnetic resonance (CMR). METHODS AND RESULTS We compared the interval changes of left ventricular ejection fraction (LVEF), myocardial perfusion reserve and the extent of myocardial scar by using late gadolinium enhancement CMR in 12 patients with severe CAD. CMR was performed at baseline and at 6 months after catheter-based direct endomyocardial autologous BM cell (n = 12) injection to viable ischaemic myocardium as guided by electromechanical mapping. In patients randomized to receive BM cell injection, there was significant decrease in percentage area of peri-infarct regions (-23.6%, P = 0.04) and increase in global LVEF (+9.0%, P = 0.02), the percentage of regional wall thickening (+13.1%, P= 0.04) and MPR (+0.25%, P = 0.03) over the target area at 6-months compared with baseline. CONCLUSIONS Direct endomyocardial implantation of autologous BM cells significantly improved global LVEF, regional wall thickening and myocardial perfusion reserve, and reduced percentage area of peri-infarct regions in patients with severe CAD.
Collapse
Affiliation(s)
- Carmen Wing-Sze Chan
- Cardiology Division, Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Yok-Lam Kwong
- Haematology Division, Department of Medicine, the University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Raymond Y Kwong
- Cardiac Magnetic Resonance Imaging, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Chu-Pak Lau
- Cardiology Division, Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Hung-Fat Tse
- Cardiology Division, Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong
- Haematology Division, Department of Medicine, the University of Hong Kong, Queen Mary Hospital, Hong Kong
| |
Collapse
|
32
|
Hossne NA, Invitti AL, Buffolo E, Azevedo S, Rodrigues de Oliveira JS, Stolf NG, Cruz LE, Sanberg PR. Refractory angina cell therapy (ReACT) involving autologous bone marrow cells in patients without left ventricular dysfunction: a possible role for monocytes. Cell Transplant 2009; 18:1299-310. [PMID: 20149298 DOI: 10.3727/096368909x484671] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Autologous bone marrow mononuclear cell (BMMC) transplantation has emerged as a potential therapeutic option for refractory angina patients. Previous studies have shown conflicting myocardium reperfusion results. The present study evaluated safety and efficacy of CellPraxis Refractory Angina Cell Therapy Protocol (ReACT), in which a specific BMMC formulation was administered as the sole therapy for these patients. The phase I/IIa noncontrolled, open label, clinical trial, involved eight patients with refractory angina and viable ischemic myocardium, without left ventricular dysfunction and who were not suitable for conventional myocardial revascularization. ReACT is a surgical procedure involving a single series of multiple injections (40-90 injections, 0.2 ml each) into ischemic areas of the left ventricle. Primary endpoints were Canadian Cardiovascular Society Angina Classification (CCSAC) improvement at 18 months follow-up and myocardium ischemic area reduction (assessed by scintigraphic analysis) at 12 months follow-up, in correlation with a specific BMMC formulation. Almost all patients presented progressive improvement in angina classification beginning 3 months (p = 0.008) postprocedure, which was sustained at 18 months follow-up (p = 0.004), as well as objective myocardium ischemic area reduction at 12 months (decrease of 84.4%, p < 0.004). A positive correlation was found between monocyte concentration and CCSAC improvement (r = -0.759, p < 0.05). Improvement in CCSAC, followed by correlated reduction in scintigraphic myocardium ischemic area, strongly suggests neoangiogenesis as the main stem cell action mechanism. The significant correlation between number of monocytes and improvement strongly supports a cell-related effect of ReACT. ReACT appeared safe and effective.
Collapse
Affiliation(s)
- Nelson Americo Hossne
- Cardiovascular Surgery Division, Surgery Department, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Sun L, Bai Y, Du G. Endothelial dysfunction--an obstacle of therapeutic angiogenesis. Ageing Res Rev 2009; 8:306-13. [PMID: 19389489 DOI: 10.1016/j.arr.2009.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Revised: 04/14/2009] [Accepted: 04/14/2009] [Indexed: 01/14/2023]
Abstract
Due to ageing populations and improvements in survival, increasing numbers of patients suffering from ischemic cardiovascular disease are not amenable to revascularization. Hence, interests are currently focused on "therapeutic angiogenesis", which is the clinical use of growth factors to enhance or promote the development of collateral blood vessels in ischemic tissue. Several growth factors (or genes encoding these growth factors) are now available for therapeutic vascular growth in normal and ischemic tissues. However, the successes of angiogenic therapy observed in pre-clinical studies have not been realized in clinical trials. Most animal studies demonstrating the physiologic effectiveness of angiogenic therapies have been performed in normal young animals, while clinical trials typically enroll older patients with various endothelial disruptive risk factors. The promising results of trials using endothelial function-improving strategies support the hypothesis that the decreased effectiveness of growth factor therapy due to endothelial dysfunction could be a principle reason for failure of trials using growth factors. We will have a retrospection of therapeutic angiogenesis trials and discuss the mechanisms that contribute to an impaired angiogenic response in the setting of endothelial dysfunction. We also briefly explore endothelial function-improving procedures that have the potentially therapeutic benefit of enhancing the angiogenic response.
Collapse
Affiliation(s)
- Lan Sun
- National Center for Pharmaceutical Screening, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| | | | | |
Collapse
|
34
|
Sanberg PR, Park DH, Kuzmin-Nichols N, Cruz E, Hossne NA, Buffolo E, Willing AE. Monocyte transplantation for neural and cardiovascular ischemia repair. J Cell Mol Med 2009; 14:553-63. [PMID: 19754667 PMCID: PMC3823455 DOI: 10.1111/j.1582-4934.2009.00903.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Neovascularization is an integral process of inflammatory reactions and subsequent repair cascades in tissue injury. Monocytes/macrophages play a key role in the inflammatory process including angiogenesis as well as the defence mechanisms by exerting microbicidal and immunomodulatory activity. Current studies have demonstrated that recruited monocytes/macrophages aid in regulating angiogenesis in ischemic tissue, tumours and chronic inflammation. In terms of neovascularization followed by tissue regeneration, monocytes/macrophages should be highly attractive for cell-based therapy compared to any other stem cells due to their considerable advantages: non-oncogenic, non-teratogenic, multiple secretary functions including pro-angiogenic and growth factors, straightforward cell harvesting procedure and non-existent ethical controversy. In addition to adult origins such as bone marrow or peripheral blood, umbilical cord blood (UCB) can be a potential source for autologous or allogeneic monocytes/macrophages. Especially, UCB monocytes should be considered as the first candidate owing to their feasibility, low immune rejection and multiple characteristic advantages such as their anti-inflammatory properties by virtue of their unique immune and inflammatory immaturity, and their pro-angiogenic ability. In this review, we present general characteristics and potential of monocytes/macrophages for cell-based therapy, especially focusing on neovascularization and UCB-derived monocytes.
Collapse
Affiliation(s)
- Paul R Sanberg
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL 33612, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Neoangiogenesis after direct intramyocardial implantation of bone marrow-derived stem cells in a patient with severe coronary artery disease ineligible for percutaneous or surgical revascularization. Open Med (Wars) 2009. [DOI: 10.2478/s11536-009-0035-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractBone marrow-derived stem cells (BMSC) may be an alternative for the treatment of patients with severe coronary artery disease ineligible for either percutaneous or surgical revascularization. This case report presents a 65-year-old male patient with untreatable angina pectoris (Canadian Cardiovascular Society Class III) and severe coronary artery disease. A mixture of BMSC containing approximately 3×106 CD34+ cells was directly injected into preoperatively determined ischemic regions of the myocardium by median sternotomy. At baseline, at 3 months, and at 1 year of follow-up, echocardiography (demonstrating wall motions of 16 segments), single-photon emission computed tomography, and coronary angiography (at baseline and at 1 year) were performed to assess myocardial perfusion, left ventricular (LV) function and coronary anatomy. The patient reached Canadian Cardiovascular Society Class I after 6 months of cell implantation. The ejection fraction increased from 34% to 37% at the third month and 40% at 1 year of follow-up. At 1 year of follow-up, preoperatively akinetic mid-base septum and anteroseptal regions progressed to mild hipokinesia and severe hypokinetic mid-base-apical anterior regions and apical lateral-inferior regions became normokinesia. Single-photon emission computed tomography revealed a visible improvement in anterior and lateral segments at 1 year of follow-up. Coronary angiography showed newly developed collateral arteries at 1 year of follow-up. BMSC transplantation in a patient with severe coronary artery disease resulted in increase of LV ejection fraction, an increase of the perfusion of ischemic myocardial regions, and improvement in wall motion defects without any adverse events.
Collapse
|
36
|
Beeres SLMA, Bax JJ, Roes SD, Lamb HJ, Fibbe WE, De Roos A, Van Der Wall EE, Schalij MJ, Atsma DE. Intramyocardial bone marrow cell transplantation and the progression of coronary atherosclerosis in patients with chronic myocardial ischemia. ACTA ACUST UNITED AC 2009; 9:243-51. [DOI: 10.1080/17482940701639385] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
37
|
Affiliation(s)
- Ester Macia
- Department of Pharmacology, Center for Molecular Therapeutics, Columbia University, New York, NY, USA
| | | |
Collapse
|
38
|
Fuh E, Brinton TJ. Bone Marrow Stem Cells for the Treatment of Ischemic Heart Disease: A Clinical Trial Review. J Cardiovasc Transl Res 2009; 2:202-18. [DOI: 10.1007/s12265-009-9095-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Accepted: 02/23/2009] [Indexed: 01/14/2023]
|
39
|
Bone marrow-derived stem/progenitor cells: their use in clinical studies for the treatment of myocardial infarction. Heart Lung Circ 2008; 18:171-80. [PMID: 19081302 DOI: 10.1016/j.hlc.2008.09.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Revised: 09/09/2008] [Accepted: 09/10/2008] [Indexed: 01/08/2023]
Abstract
Over the last six years, several centres around the world have started clinical trials to investigate the utilisation of bone marrow-derived cells for myocardial infarction. Different types and numbers of cells have been used assuming they possess a potential to originate new endothelial cells and/or cardiomyocytes to repair/regenerate the ailed heart. Despite diversity in number, clinical status of subjects, route of cell administration, and criteria to evaluate efficacy, the main conclusion drawn from these clinical studies was that such therapies were safe. However, attempts to unify efficacy data have yielded no clear answers, so far. This review offers an in-depth and critical analysis of these trials and intends to evaluate from the cellular biology and clinical cardiology viewpoints, the significant information that has been published since 2002, as well as that emerging from ongoing clinical trials. Emphasis will be placed on cellular types, research designs and methods to evaluate efficacy of each particular treatment modality.
Collapse
|
40
|
Beeres SLMA, Lamb HJ, Roes SD, Holman ER, Kaandorp TAM, Fibbe WE, de Roos A, van der Wall EE, Schalij MJ, Bax JJ, Atsma DE. Effect of intramyocardial bone marrow cell injection on diastolic function in patients with chronic myocardial ischemia. J Magn Reson Imaging 2008; 27:992-7. [PMID: 18425845 DOI: 10.1002/jmri.21081] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
PURPOSE To evaluate the effect of intramyocardial bone marrow cell injection on diastolic function in patients with chronic myocardial ischemia. MATERIALS AND METHODS In 24 patients (19 male; 65 +/- 9 years) with refractory angina (Canadian Cardiovascular Society [CCS] class III-IV) 84.6 +/- 28.7 x 10(6) bone marrow-derived mononuclear cells were injected intramyocardially (using the NOGA system) in regions with ischemia on Tc-99m tetrofosmin single photon emission computed tomography (SPECT). Diastolic function was evaluated at baseline and at three months using magnetic resonance imaging (MRI) and tissue Doppler imaging (TDI). RESULTS MRI revealed an increased early (E) peak filling rate (374 +/- 121 mL/second vs. 412 +/- 102 mL/second; P = 0.04), whereas the atrial (A) peak filling rate remained unchanged (340 +/- 81 mL/second vs. 334 +/- 93 mL/second; P = not significant [NS]). The E/A peak flow ratio increased from 1.09 +/- 0.33 to 1.23 +/- 0.47 at three months (P = 0.02). TDI demonstrated a significant improvement in early diastolic velocity (E') from 4.4 +/- 1.7 cm/second to 4.8 +/- 1.6 cm/second at three months (P = 0.03), whereas the late diastolic velocity (A') remained unchanged (6.0 +/- 1.6 cm/second vs. 6.0 +/- 1.7 cm/second; P = NS). Consequently, the E'/A' ratio increased from 0.74 +/- 0.19 to 0.84 +/- 0.28 at three months (P = 0.02). CONCLUSION Intramyocardial bone marrow cell injection in patients with chronic myocardial ischemia improved MRI and TDI-derived parameters of diastolic function.
Collapse
Affiliation(s)
- Saskia L M A Beeres
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Baldazzi F, Jørgensen E, Ripa RS, Kastrup J. Release of biomarkers of myocardial damage after direct intramyocardial injection of genes and stem cells via the percutaneous transluminal route. Eur Heart J 2008; 29:1819-26. [PMID: 18524811 DOI: 10.1093/eurheartj/ehn233] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS We aimed to quantify the release of biomarkers of myocardial damage in relation to direct intramyocardial injections of genes and stem cells in patients with severe coronary artery disease. METHODS AND RESULTS We studied 71 patients with 'no-option' coronary artery disease. Patients had, via the percutaneous transluminal route, a total of 11 +/- 1 (mean +/- SD) intramyocardial injections of vascular endothelial growth factor genes (n = 56) or mesenchymal stromal cells (n = 15). Injections were guided to an ischaemic area by electromechanical mapping, using the NOGA/Myostar catheter system. Plasma CKMB (upper normal laboratory limit = 5 microg/L) was 2 microg/L (2-3) at baseline; increased to 6 (5-9) after 8 h (P < 0.0001) and normalized to 4 (3-5) after 24 h. A total of eight patients (17%), receiving a volume of 0.3 mL per injection, had CKMB rises exceeding three times the upper limit, whereas no patient in the group receiving 0.2 mL had a more than two-fold CKMB increase. No patient developed new ECG changes. There were no clinically ventricular arrhythmias and no death. CONCLUSION NOGA mapping followed by direct intramyocardial injections of stem cells or genes lead to measurable release of cardiac biomarkers compared with NOGA mapping alone. The increase in biomarkers was related to the injected volume.
Collapse
Affiliation(s)
- Federica Baldazzi
- Department of Cardiology, Cardiac Catheterisation Laboratory 2014, The Heart Centre, Rigshospitalet, University Hospital Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen Ø, Denmark
| | | | | | | |
Collapse
|
42
|
GUERRERO MAYRA, ATHOTA KRISHNA, MOY JASON, MEHTA LAXMIS, LAGUENS RUBEN, CROTTOGINI ALBERTO, BORRELLI MICHAEL, CORRY PETER, SCHOENHERR DIANE, GENTRY RALPH, BOURA JUDITH, GRINES CINDYL, RAFF GILBERTL, SHANLEY CHARLESJ, O'NEILL WILLIAMW. Vascular Endothelial Growth Factor-165 Gene Therapy Promotes Cardiomyogenesis in Reperfused Myocardial Infarction. J Interv Cardiol 2008; 21:242-51. [DOI: 10.1111/j.1540-8183.2008.00358.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
43
|
Charwat S, Gyöngyösi M, Lang I, Graf S, Beran G, Hemetsberger R, Nyolczas N, Sochor H, Glogar D. Role of adult bone marrow stem cells in the repair of ischemic myocardium: current state of the art. Exp Hematol 2008; 36:672-80. [PMID: 18358589 DOI: 10.1016/j.exphem.2008.01.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Revised: 10/30/2007] [Accepted: 01/22/2008] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To review the milestones in stem cell therapy for ischemic heart disease from early basic science to large clinical studies and new therapeutic approaches. MATERIALS AND METHODS Basic research and clinical trials (systematic review) were used. The heart has the ability to regenerate through activation of resident cardiac stem cells or through recruitment of a stem cell population from other tissues, such as bone marrow. Although the underlying mechanism is yet to be made clear, numerous studies in animals have documented that transplantation of bone marrow-derived stem cells or circulating progenitor cells following acute myocardial infarction and ischemic cardiomyopathy is associated with a reduction in infarct scar size and improvements in left ventricular function and myocardial perfusion. RESULTS Cell-based cardiac therapy has expanded considerably in recent years and is on its way to becoming an established cardiovascular therapy for patients with ischemic heart disease. There have been recent insights into the understanding of mechanisms involved in the mobilization and homing of the imported cells, as well as into the paracrine effect, growth factors, and bioactive molecules. Additional information has been obtained regarding new stem cell sources, cell-based gene therapy, cell-enhancement strategies, and tissue engineering, all of which should enhance the efficacy of human cardiac stem cell therapy. CONCLUSIONS The recently published trials using bone marrow-origin stem cells in cardiac repair reported a modest but significant benefit from this therapy. Further clinical research should aim to optimize the cell types utilized and their delivery mode, and pinpoint optimal time of cell transplantation.
Collapse
Affiliation(s)
- Silvia Charwat
- Department of Cardiology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Refractory angina as a result of chronic myocardial ischemia is a common and disabling condition. Adult progenitor cells have emerged as a potential revolutionary treatment for cardiovascular disease. Locally administered adult progenitor cells with particular vasculogenic potential may offer treatment hope for those with chronic ischemia; however, the optimal cell type, dose, delivery mode, and target patient population has not been defined. Preclinical cell therapy studies have shown safety and efficacy sufficient to warrant human trials. Early, small-scale human trials exploring various cell types and delivery modes have shown that most approaches are safe, with modest early efficacy. This overview discusses the rationale and early results for ongoing larger cardiovascular disease trials, with a special emphasis on refractory angina and chronic myocardial ischemia.
Collapse
Affiliation(s)
- Amish N Raval
- Division of Cardiovascular Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53792-3248, USA.
| |
Collapse
|
45
|
Ichim TE, Solano F, Brenes R, Glenn E, Chang J, Chan K, Riordan NH. Placental mesenchymal and cord blood stem cell therapy for dilated cardiomyopathy. Reprod Biomed Online 2008; 16:898-905. [DOI: 10.1016/s1472-6483(10)60159-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
46
|
Al Sabti H. Therapeutic angiogenesis in cardiovascular disease. J Cardiothorac Surg 2007; 2:49. [PMID: 18021404 PMCID: PMC2169246 DOI: 10.1186/1749-8090-2-49] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2007] [Accepted: 11/16/2007] [Indexed: 01/13/2023] Open
Abstract
Atherosclerotic disease of the arteries is a major cause of coronary artery disease, peripheral vascular disease and stroke. Some patients are however not candidate for the standard treatment of angioplasty or bypass surgery. Hence there is tremendous enthusiasm for the utilization of angiogenesis as a therapeutic modality for atherosclerotic arterial disease. This augmentation of physiological neo-vascularization in cardiovascular disease can be achieved through different pathways. In this article we are reviewing the Use of Gene therapy, Protein therapy and cellular therapy.
Collapse
Affiliation(s)
- Hilal Al Sabti
- Department of surgery, Sultan Qaboos University Hospital, Code 123, P.Box 35, Al Khod, Sultanate of Oman.
| |
Collapse
|
47
|
Tse HF, Thambar S, Kwong YL, Rowlings P, Bellamy G, McCrohon J, Bastian B, Chan JKF, Lo G, Ho CL, Parker A, Hauser TH, Lau CP. Comparative evaluation of long-term clinical efficacy with catheter-based percutaneous intramyocardial autologous bone marrow cell implantation versus laser myocardial revascularization in patients with severe coronary artery disease. Am Heart J 2007; 154:982.e1-6. [PMID: 17967607 DOI: 10.1016/j.ahj.2007.07.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2007] [Accepted: 07/23/2007] [Indexed: 11/17/2022]
Abstract
BACKGROUND Catheter-based percutaneous laser myocardial revascularization (PMR) and intramyocardial direct bone marrow (BM) cell implantation have been investigated to treat patients with severe coronary artery disease (CAD). In both therapeutic approaches, direct local myocardial injury might be a common mechanism to induce therapeutic angiogenesis. METHODS We studied the long-term clinical outcome in 16 patients with severe CAD who received either catheter-based PMR (n = 8) or intramyocardial autologous BM cell implantation (n = 8) as guided by electromechanical mapping. RESULTS There were no significant differences in the baseline characteristics and the number of injection versus the number of laser pulse delivered between the 2 groups (P > .05). As compared with baseline, the New York Heart Association functional class and the number of anginal episodes were significantly reduced at 3- and 6-month follow-up in both BM and PMR groups (P < .05). However, the improvement in the New York Heart Association class and the reduction in anginal episodes at 18 months were only persisted in the BM group (P < .05) but not in the PMR group (P > .05). Furthermore, there were significant improvements in exercise time at 6- and 18-month follow-up, and the extent of stress-induced perfusion single-photon emission computed tomography defects at 6-month follow-up in BM group, as compared with baseline (all P < .05), but not in the PMR group (all P > .05). As compared with baseline, there were no significant changes in the total quality of life scores during follow-up in both groups (all P > .05). CONCLUSIONS The results of this study demonstrated that the catheter-based intramyocardial autologous BM cell implantation might be more effective than PMR in improving symptoms and exercise capacity in patients with severe CAD. The beneficial effect of direct intramyocardial injection was over and beyond those noted in patients treated with PMR, suggesting a potential direct therapeutic effect of BM cells, rather than local myocardial injury alone on chronic ischemic myocardium.
Collapse
Affiliation(s)
- Hung-Fat Tse
- Department of Medicine, University of Hong Kong, Queen Mary Hospital, Hong Kong, China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Tse HF, Siu CW, Zhu SG, Songyan L, Zhang QY, Lai WH, Kwong YL, Nicholls J, Lau CP. Paracrine effects of direct intramyocardial implantation of bone marrow derived cells to enhance neovascularization in chronic ischaemic myocardium. Eur J Heart Fail 2007; 9:747-53. [PMID: 17481945 DOI: 10.1016/j.ejheart.2007.03.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2006] [Revised: 02/24/2007] [Accepted: 03/19/2007] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVE To determine the optimal bone marrow (BM) cell types, and their potential mechanisms of action for neovascularization in chronic ischaemic myocardium. METHODS AND RESULTS The functional effects, angiogenic potential and cytokine expression of direct intramyocardial implantation of autologous BM CD31-positive endothelial progenitor cells (EPC, n=9), BM mononuclear cells (MNCs, n=9), and saline (n=9) were compared in a swine model of chronic ischaemic myocardium. Autologous BM cells were harvested and catheter-based electromechanical mapping-guided direct intramyocardial injection was performed to target ischaemic myocardium. After 12 weeks, injection of BM-MNC resulted in significant improvements in left ventricular dP/dt (+21+/-8%, P=0.032), left ventricular pressure (+17+/-4%, P=0.048) and regional microsphere myocardial perfusion over ischaemic endocardium (+74+/-28%, P<0.05) and epicardium (+73+/-29%, P<0.05). No significant effects were observed following injection of BM-EPC or saline. Capillary density (1132+/-69 versus 903+/-44 per mm(2), P=0.047) and expression of mRNA of vascular endothelial growth factor (VEGF, 32.3+/-5.6 versus 13.1+/-3.7, P<0.05,) and angiopoietin-2 (23.9+/-3.6 versus 13.7+/-3.1, P<0.05) in ischaemic myocardium was significantly greater in the BM-MNC group than the saline group. The capillary density in ischaemic myocardium demonstrated a significant positive correlation with VEGF expression (r=0.61, P<0.001). CONCLUSION Catheter-based direct intramyocardial injection of BM-MNC enhanced angiogenesis more effectively than BM-EPC or saline, possibly via a paracrine effect, with increased expression of VEGF that subsequently improved cardiac performance of ischaemic myocardium.
Collapse
Affiliation(s)
- Hung-Fat Tse
- Division of Cardiology, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Chachques JC, Azarine A, Mousseaux E, El Serafi M, Cortes-Morichetti M, Carpentier AF. MRI Evaluation of Local Myocardial Treatments: Epicardial Versus Endocardial (Cell-Fix Catheter) Injections. J Interv Cardiol 2007; 20:188-96. [PMID: 17524110 DOI: 10.1111/j.1540-8183.2007.00255.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
AIMS We compared two procedures for local myocardial treatment: transepicardial versus transendocardial catheter injection. Transepicardial injections were performed under direct surgical visualization whereas transendocardial injections were performed using electrophysiological guidance. METHODS A left ventricle (LV) myocardial infarction (MI) was surgically created in 14 sheep. At 3 months, gadolinium was injected IV followed by the injection of super paramagnetic iron oxide (SPIO) into MI. Animals were divided in two groups: transepicardial injection (Group I) versus transendocardial (Group II) using "Cell-Fix" catheter injection. This catheter was developed to identify by electrophysiology the infarcted area and to stabilize injections suctioning the device to the endocardium. Postgadolinium delayed-enhancement magnetic resonance imaging (MRI) was performed to stain the infarct size. SPIO injections were used to assess the magnitude of the treated area. The ratio between SPIO black stained treatment areas and white gadolinium stained infarcted areas was calculated using MRI. RESULTS The electrophysiological recordings by the catheter for the MI versus normal LV wall were: R wave amplitude 4.16 versus 12.08 mV (P = 0.003), slew rate (slope of the signal) 0.36 V/s versus 1.04 V/s (P = 0.008). The ratio of the SPIO diffusion into the MI was 41.2 +/- 8.1% for surgical and 63.7 +/- 8.2% for percutaneous endocardial injections (P = 0.0132). CONCLUSION MRI allows evaluation of the extent of local myocardial treatments. The differences shown between epicardial and endocardial injections concerning the distribution of SPIO can be justified by the methodology of injection and by a more precise MI detection by electrophysiology. In conclusion, electrophysiological recordings to guide injections is superior to direct surgical visualization in terms of injecting into infarcted tissue.
Collapse
Affiliation(s)
- Juan C Chachques
- Department of Cardiovascular Surgery, Pompidou and Bicetre Hospitals, Paris, France.
| | | | | | | | | | | |
Collapse
|
50
|
Beeres SLMA, Zeppenfeld K, Bax JJ, Dibbets-Schneider P, Stokkel MPM, Fibbe WE, van der Wall EE, Atsma DE, Schalij MJ. Electrophysiological and arrhythmogenic effects of intramyocardial bone marrow cell injection in patients with chronic ischemic heart disease. Heart Rhythm 2007; 4:257-65. [PMID: 17341383 DOI: 10.1016/j.hrthm.2006.10.033] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2006] [Accepted: 10/30/2006] [Indexed: 10/23/2022]
Abstract
BACKGROUND Bone marrow cell injection has been introduced to treat patients with ischemic heart disease. However, focal application of bone marrow cells may generate an arrhythmogenic substrate. OBJECTIVES To assess the electrophysiological and arrhythmogenic effects of intramyocardial bone marrow cell injection in patients with chronic myocardial ischemia. METHODS Bone marrow was aspirated in 20 patients (65+/-11 years, 19 male) with drug-refractory angina and myocardial ischemia. Electroanatomical mapping (NOGA, Biosense-Webster, Waterloo, Belgium) was performed during mononuclear cell isolation. Areas for cell injection were selected based on the localization of ischemia on SPECT. These areas were mapped in detail to evaluate local bipolar electrogram duration, amplitude and fragmentation. Mononuclear cells were injected in the ischemic area with the NOGA system. SPECT and electroanatomical mapping were repeated at 3 months. Holter monitoring was repeated at 3 and 6 months. RESULTS SPECT revealed a decrease in the number of segments with ischemia (3.5+/-2.5 vs. 1.1+/-1.0 at 3 months; P<0.01) and an increased left ventricular ejection fraction (44+/-13% vs. 49+/-17% at 3 months; P=0.02). The number of ventricular premature beats remained unchanged (10+/-24x10(2)/24h vs. 8+/-23x10(2)/24h at 3 months (P=NS) and 12+/-30x10(2)/24h at 6 months (P=NS)). At 3 months follow-up, bone marrow cell injection did not prolong electrogram duration (15.9+/-4.6 ms vs. 15.6+/-4.0 ms; P=NS), decrease electrogram amplitude (3.8+/-1.5 mV vs. 3.8+/-1.5 mV; P=NS), or increase fragmentation (2.0+/-0.5 vs. 1.9+/-0.4; P=NS). CONCLUSION Intramyocardial bone marrow cell injection does not increase the incidence of ventricular arrhythmias and does not alter the electrophysiological properties of the injected myocardium.
Collapse
Affiliation(s)
- Saskia L M A Beeres
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|