Skip to main content
Top
Published in: Current Heart Failure Reports 2/2013

01-06-2013 | Management of Heart Failure (TE Meyer, Section Editor)

Current Status of Cell-Based Therapy for Heart Failure

Authors: Philipp Jakob, Ulf Landmesser

Published in: Current Heart Failure Reports | Issue 2/2013

Login to get access

Abstract

In the last two decades, morbidity and mortality of patients with chronic heart failure could be further reduced by improved pharmacological and cardiac device therapies. However, despite these advances, there is a substantial unmet need for novel therapies, ideally specifically addressing repair and regeneration of the damaged or lost myocardium and its vasculature, given the limited endogenous potential for renewal of cardiomyocytes in adults. In this respect, cardiac cell-based therapies have gained substantial attention and have entered clinical feasibility and safety studies a decade ago. Different cell-types have been used, including bone marrow–derived mononuclear cells, bone marrow–derived mesenchymal stem cells, mobilized CD34+ cells, and more recently cardiac-derived c-kit+ stem cells and cardiosphere-derived cells. Some of these studies have suggested a potential of cell-based therapies to reduce cardiac scar size and to improve cardiac function in patients with ischemic cardiomyopathy. While first clinical trials examining the impact of cardiac cell–based therapy on clinical outcome have now been initiated, improved understanding of underlying mechanisms of action of cell-based therapies may lead to strategies for optimization of the cardiac repair potential of the applied cells. In experimental studies, direct in vivo reprogramming of cardiac fibroblasts towards cardiomyocytes, and microRNA-based promotion of cardiomyocyte proliferation and cardiac repair have recently been reported that may represent novel therapeutic approaches for cardiac regeneration that would not need cell-administration but rather directly stimulate endogenous cardiac regeneration. This review will focus mainly on recently completed clinical trials (within the last 2 years) investigating cardiac cell-based therapies and the current status of experimental studies for cardiac cell-based repair and regeneration with a potential for later translation into clinical studies in the future.
Literature
2.
go back to reference Jopling C, Sleep E, Raya M, Marti M, Raya A, Izpisua Belmonte JC. Zebrafish heart regeneration occurs by cardiomyocyte dedifferentiation and proliferation. Nature. 2010;464(7288):606–9. doi:10.1038/nature08899.PubMedCrossRef Jopling C, Sleep E, Raya M, Marti M, Raya A, Izpisua Belmonte JC. Zebrafish heart regeneration occurs by cardiomyocyte dedifferentiation and proliferation. Nature. 2010;464(7288):606–9. doi:10.​1038/​nature08899.PubMedCrossRef
7.
go back to reference Gepstein L. Derivation and potential applications of human embryonic stem cells. Circ Res. 2002;91(10):866–76.PubMedCrossRef Gepstein L. Derivation and potential applications of human embryonic stem cells. Circ Res. 2002;91(10):866–76.PubMedCrossRef
8.
go back to reference Laflamme MA, Chen KY, Naumova AV, Muskheli V, Fugate JA, Dupras SK, et al. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat Biotechnol. 2007;25(9):1015–24. doi:10.1038/nbt1327.PubMedCrossRef Laflamme MA, Chen KY, Naumova AV, Muskheli V, Fugate JA, Dupras SK, et al. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat Biotechnol. 2007;25(9):1015–24. doi:10.​1038/​nbt1327.PubMedCrossRef
9.
10.
go back to reference Yamashita J, Itoh H, Hirashima M, Ogawa M, Nishikawa S, Yurugi T, et al. Flk1-positive cells derived from embryonic stem cells serve as vascular progenitors. Nature. 2000;408(6808):92–6. doi:10.1038/35040568.PubMedCrossRef Yamashita J, Itoh H, Hirashima M, Ogawa M, Nishikawa S, Yurugi T, et al. Flk1-positive cells derived from embryonic stem cells serve as vascular progenitors. Nature. 2000;408(6808):92–6. doi:10.​1038/​35040568.PubMedCrossRef
16.
go back to reference Templin C, Zweigerdt R, Schwanke K, Olmer R, Ghadri JR, Emmert MY, et al. Transplantation and tracking of human-induced pluripotent stem cells in a pig model of myocardial infarction: assessment of cell survival, engraftment, and distribution by hybrid single photon emission computed tomography/computed tomography of sodium iodide symporter transgene expression. Circulation. 2012;126(4):430–9. doi:10.1161/CIRCULATIONAHA.111.087684.PubMedCrossRef Templin C, Zweigerdt R, Schwanke K, Olmer R, Ghadri JR, Emmert MY, et al. Transplantation and tracking of human-induced pluripotent stem cells in a pig model of myocardial infarction: assessment of cell survival, engraftment, and distribution by hybrid single photon emission computed tomography/computed tomography of sodium iodide symporter transgene expression. Circulation. 2012;126(4):430–9. doi:10.​1161/​CIRCULATIONAHA.​111.​087684.PubMedCrossRef
18.
go back to reference • Qian L, Huang Y, Spencer CI, Foley A, Vedantham V, Liu L, et al. In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes. Nature. 2012;485(7400):593–8. doi:10.1038/nature11044. This experimental study shows that systemic delivery of cardiac transcription factors can directly reprogram resident cardiac fibroblasts into cardiomyocyte–like cells.PubMedCrossRef • Qian L, Huang Y, Spencer CI, Foley A, Vedantham V, Liu L, et al. In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes. Nature. 2012;485(7400):593–8. doi:10.​1038/​nature11044. This experimental study shows that systemic delivery of cardiac transcription factors can directly reprogram resident cardiac fibroblasts into cardiomyocyte–like cells.PubMedCrossRef
20.
go back to reference Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114(6):763–76.PubMedCrossRef Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114(6):763–76.PubMedCrossRef
22.
24.
go back to reference Oh H, Bradfute SB, Gallardo TD, Nakamura T, Gaussin V, Mishina Y, et al. Cardiac progenitor cells from adult myocardium: homing, differentiation, and fusion after infarction. Proc Natl Acad Sci U S A. 2003;100(21):12313–8. doi:10.1073/pnas.2132126100.PubMedCrossRef Oh H, Bradfute SB, Gallardo TD, Nakamura T, Gaussin V, Mishina Y, et al. Cardiac progenitor cells from adult myocardium: homing, differentiation, and fusion after infarction. Proc Natl Acad Sci U S A. 2003;100(21):12313–8. doi:10.​1073/​pnas.​2132126100.PubMedCrossRef
25.
go back to reference Matsuura K, Honda A, Nagai T, Fukushima N, Iwanaga K, Tokunaga M, et al. Transplantation of cardiac progenitor cells ameliorates cardiac dysfunction after myocardial infarction in mice. J Clin Invest. 2009;119(8):2204–17. doi:10.1172/JCI37456.PubMed Matsuura K, Honda A, Nagai T, Fukushima N, Iwanaga K, Tokunaga M, et al. Transplantation of cardiac progenitor cells ameliorates cardiac dysfunction after myocardial infarction in mice. J Clin Invest. 2009;119(8):2204–17. doi:10.​1172/​JCI37456.PubMed
28.
go back to reference Li TS, Cheng K, Malliaras K, Smith RR, Zhang Y, Sun B, et al. Direct comparison of different stem cell types and subpopulations reveals superior paracrine potency and myocardial repair efficacy with cardiosphere-derived cells. J Am Coll Cardiol. 2012;59(10):942–53. doi:10.1016/j.jacc.2011.11.029.PubMedCrossRef Li TS, Cheng K, Malliaras K, Smith RR, Zhang Y, Sun B, et al. Direct comparison of different stem cell types and subpopulations reveals superior paracrine potency and myocardial repair efficacy with cardiosphere-derived cells. J Am Coll Cardiol. 2012;59(10):942–53. doi:10.​1016/​j.​jacc.​2011.​11.​029.PubMedCrossRef
29.
go back to reference Johnston PV, Sasano T, Mills K, Evers R, Lee ST, Smith RR, et al. Engraftment, differentiation, and functional benefits of autologous cardiosphere-derived cells in porcine ischemic cardiomyopathy. Circulation. 2009;120(12):1075–83. doi:10.1161/CIRCULATIONAHA.108.816058. 7 p following 83.PubMedCrossRef Johnston PV, Sasano T, Mills K, Evers R, Lee ST, Smith RR, et al. Engraftment, differentiation, and functional benefits of autologous cardiosphere-derived cells in porcine ischemic cardiomyopathy. Circulation. 2009;120(12):1075–83. doi:10.​1161/​CIRCULATIONAHA.​108.​816058. 7 p following 83.PubMedCrossRef
30.
go back to reference Chimenti I, Smith RR, Li TS, Gerstenblith G, Messina E, Giacomello A, et al. Relative roles of direct regeneration versus paracrine effects of human cardiosphere-derived cells transplanted into infarcted mice. Circ Res. 2010;106(5):971–80. doi:10.1161/CIRCRESAHA.109.210682.PubMedCrossRef Chimenti I, Smith RR, Li TS, Gerstenblith G, Messina E, Giacomello A, et al. Relative roles of direct regeneration versus paracrine effects of human cardiosphere-derived cells transplanted into infarcted mice. Circ Res. 2010;106(5):971–80. doi:10.​1161/​CIRCRESAHA.​109.​210682.PubMedCrossRef
31.
go back to reference Welt FG, Gallegos R, Connell J, Kajstura J, D'Amario D, Kwong RY, et al. Effect of cardiac stem cells on left ventricular remodeling in a canine model of chronic myocardial infarction. Circ Heart Fail. 2012. doi:10.1161/CIRCHEARTFAILURE.112.972273. Welt FG, Gallegos R, Connell J, Kajstura J, D'Amario D, Kwong RY, et al. Effect of cardiac stem cells on left ventricular remodeling in a canine model of chronic myocardial infarction. Circ Heart Fail. 2012. doi:10.​1161/​CIRCHEARTFAILURE​.​112.​972273.
32.
go back to reference •• Bolli R, Chugh AR, D'Amario D, Loughran JH, Stoddard MF, Ikram S, et al. Cardiac stem cells in patients with ischaemic cardiomyopathy (SCIPIO): initial results of a randomised phase 1 trial. Lancet. 2011;378(9806):1847–57. doi:10.1016/S0140-6736(11)61590-0. This is the first clinical study using c-kit+ cardiac stem cells in patients with ischemic cardiomyopathy.PubMedCrossRef •• Bolli R, Chugh AR, D'Amario D, Loughran JH, Stoddard MF, Ikram S, et al. Cardiac stem cells in patients with ischaemic cardiomyopathy (SCIPIO): initial results of a randomised phase 1 trial. Lancet. 2011;378(9806):1847–57. doi:10.​1016/​S0140-6736(11)61590-0. This is the first clinical study using c-kit+ cardiac stem cells in patients with ischemic cardiomyopathy.PubMedCrossRef
33.
go back to reference Chugh AR, Beache GM, Loughran JH, Mewton N, Elmore JB, Kajstura J, et al. Administration of cardiac stem cells in patients with ischemic cardiomyopathy: the SCIPIO trial: surgical aspects and interim analysis of myocardial function and viability by magnetic resonance. Circulation. 2012;126(11 Suppl 1):S54–64. doi:10.1161/CIRCULATIONAHA.112.092627.PubMedCrossRef Chugh AR, Beache GM, Loughran JH, Mewton N, Elmore JB, Kajstura J, et al. Administration of cardiac stem cells in patients with ischemic cardiomyopathy: the SCIPIO trial: surgical aspects and interim analysis of myocardial function and viability by magnetic resonance. Circulation. 2012;126(11 Suppl 1):S54–64. doi:10.​1161/​CIRCULATIONAHA.​112.​092627.PubMedCrossRef
34.
go back to reference •• Makkar RR, Smith RR, Cheng K, Malliaras K, Thomson LE, Berman D, et al. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase 1 trial. Lancet. 2012;379(9819):895–904. doi:10.1016/S0140-6736(12)60195-0. This is the first clinical trial using cardiosphere-derived cells (CDCs) in patients with ischemic cardiomyopathy.PubMedCrossRef •• Makkar RR, Smith RR, Cheng K, Malliaras K, Thomson LE, Berman D, et al. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase 1 trial. Lancet. 2012;379(9819):895–904. doi:10.​1016/​S0140-6736(12)60195-0. This is the first clinical trial using cardiosphere-derived cells (CDCs) in patients with ischemic cardiomyopathy.PubMedCrossRef
36.
go back to reference Jakob P, Doerries C, Briand S, Mocharla P, Krankel N, Besler C, et al. Loss of AngiomiR-126 and 130a in angiogenic early outgrowth cells from patients with chronic heart failure: role for impaired in vivo neovascularization and cardiac repair capacity. Circulation. 2012. doi:10.1161/CIRCULATIONAHA.112.093906. Jakob P, Doerries C, Briand S, Mocharla P, Krankel N, Besler C, et al. Loss of AngiomiR-126 and 130a in angiogenic early outgrowth cells from patients with chronic heart failure: role for impaired in vivo neovascularization and cardiac repair capacity. Circulation. 2012. doi:10.​1161/​CIRCULATIONAHA.​112.​093906.
39.
go back to reference Murry CE, Soonpaa MH, Reinecke H, Nakajima H, Nakajima HO, Rubart M, et al. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature. 2004;428(6983):664–8. doi:10.1038/nature02446.PubMedCrossRef Murry CE, Soonpaa MH, Reinecke H, Nakajima H, Nakajima HO, Rubart M, et al. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature. 2004;428(6983):664–8. doi:10.​1038/​nature02446.PubMedCrossRef
40.
go back to reference Nygren JM, Jovinge S, Breitbach M, Sawen P, Roll W, Hescheler J, et al. Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nat Med. 2004;10(5):494–501. doi:10.1038/nm1040.PubMedCrossRef Nygren JM, Jovinge S, Breitbach M, Sawen P, Roll W, Hescheler J, et al. Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nat Med. 2004;10(5):494–501. doi:10.​1038/​nm1040.PubMedCrossRef
41.
go back to reference • Gnecchi M, Zhang Z, Ni A, Dzau VJ. Paracrine mechanisms in adult stem cell signaling and therapy. Circ Res. 2008;103(11):1204–19. doi:10.1161/CIRCRESAHA.108.176826. This comprehensive review describes and discusses one of the most important mechanism - namely paracrine signaling - by which adult stem cells exert their effects.PubMedCrossRef • Gnecchi M, Zhang Z, Ni A, Dzau VJ. Paracrine mechanisms in adult stem cell signaling and therapy. Circ Res. 2008;103(11):1204–19. doi:10.​1161/​CIRCRESAHA.​108.​176826. This comprehensive review describes and discusses one of the most important mechanism - namely paracrine signaling - by which adult stem cells exert their effects.PubMedCrossRef
43.
go back to reference Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D, Wang J, et al. Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med. 2001;7(4):430–6. doi:10.1038/86498.PubMedCrossRef Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D, Wang J, et al. Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med. 2001;7(4):430–6. doi:10.​1038/​86498.PubMedCrossRef
44.
go back to reference Urbich C, Aicher A, Heeschen C, Dernbach E, Hofmann WK, Zeiher AM, et al. Soluble factors released by endothelial progenitor cells promote migration of endothelial cells and cardiac resident progenitor cells. J Mol Cell Cardiol. 2005;39(5):733–42. doi:10.1016/j.yjmcc.2005.07.003.PubMedCrossRef Urbich C, Aicher A, Heeschen C, Dernbach E, Hofmann WK, Zeiher AM, et al. Soluble factors released by endothelial progenitor cells promote migration of endothelial cells and cardiac resident progenitor cells. J Mol Cell Cardiol. 2005;39(5):733–42. doi:10.​1016/​j.​yjmcc.​2005.​07.​003.PubMedCrossRef
47.
go back to reference Assmus B, Schachinger V, Teupe C, Britten M, Lehmann R, Dobert N, et al. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction (TOPCARE-AMI). Circulation. 2002;106(24):3009–17.PubMedCrossRef Assmus B, Schachinger V, Teupe C, Britten M, Lehmann R, Dobert N, et al. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction (TOPCARE-AMI). Circulation. 2002;106(24):3009–17.PubMedCrossRef
48.
go back to reference Wollert KC, Meyer GP, Lotz J, Ringes-Lichtenberg S, Lippolt P, Breidenbach C, et al. Intracoronary autologous bone-marrow cell transfer after myocardial infarction: the BOOST randomised controlled clinical trial. Lancet. 2004;364(9429):141–8. doi:10.1016/S0140-6736(04)16626-9.PubMedCrossRef Wollert KC, Meyer GP, Lotz J, Ringes-Lichtenberg S, Lippolt P, Breidenbach C, et al. Intracoronary autologous bone-marrow cell transfer after myocardial infarction: the BOOST randomised controlled clinical trial. Lancet. 2004;364(9429):141–8. doi:10.​1016/​S0140-6736(04)16626-9.PubMedCrossRef
49.
go back to reference Janssens S, Dubois C, Bogaert J, Theunissen K, Deroose C, Desmet W, et al. Autologous bone marrow-derived stem-cell transfer in patients with ST-segment elevation myocardial infarction: double-blind, randomised controlled trial. Lancet. 2006;367(9505):113–21. doi:10.1016/S0140-6736(05)67861-0.PubMedCrossRef Janssens S, Dubois C, Bogaert J, Theunissen K, Deroose C, Desmet W, et al. Autologous bone marrow-derived stem-cell transfer in patients with ST-segment elevation myocardial infarction: double-blind, randomised controlled trial. Lancet. 2006;367(9505):113–21. doi:10.​1016/​S0140-6736(05)67861-0.PubMedCrossRef
50.
go back to reference Lunde K, Solheim S, Aakhus S, Arnesen H, Abdelnoor M, Egeland T, et al. Intracoronary injection of mononuclear bone marrow cells in acute myocardial infarction. N Engl J Med. 2006;355(12):1199–209. doi:10.1056/NEJMoa055706.PubMedCrossRef Lunde K, Solheim S, Aakhus S, Arnesen H, Abdelnoor M, Egeland T, et al. Intracoronary injection of mononuclear bone marrow cells in acute myocardial infarction. N Engl J Med. 2006;355(12):1199–209. doi:10.​1056/​NEJMoa055706.PubMedCrossRef
51.
go back to reference •• Schachinger V, Erbs S, Elsasser A, Haberbosch W, Hambrecht R, Holschermann H, et al. Intracoronary bone marrow-derived progenitor cells in acute myocardial infarction. N Engl J Med. 2006;355(12):1210–21. doi:10.1056/NEJMoa060186. This landmark study demonstrates improvement of cardiac function after BM-MNCs transplantation in patients with acute myocardial infarction.PubMedCrossRef •• Schachinger V, Erbs S, Elsasser A, Haberbosch W, Hambrecht R, Holschermann H, et al. Intracoronary bone marrow-derived progenitor cells in acute myocardial infarction. N Engl J Med. 2006;355(12):1210–21. doi:10.​1056/​NEJMoa060186. This landmark study demonstrates improvement of cardiac function after BM-MNCs transplantation in patients with acute myocardial infarction.PubMedCrossRef
53.
go back to reference Zimmet H, Porapakkham P, Sata Y, Haas SJ, Itescu S, Forbes A, et al. Short- and long-term outcomes of intracoronary and endogenously mobilized bone marrow stem cells in the treatment of ST-segment elevation myocardial infarction: a meta-analysis of randomized control trials. Eur J Heart Fail. 2012;14(1):91–105. doi:10.1093/eurjhf/hfr148.PubMedCrossRef Zimmet H, Porapakkham P, Sata Y, Haas SJ, Itescu S, Forbes A, et al. Short- and long-term outcomes of intracoronary and endogenously mobilized bone marrow stem cells in the treatment of ST-segment elevation myocardial infarction: a meta-analysis of randomized control trials. Eur J Heart Fail. 2012;14(1):91–105. doi:10.​1093/​eurjhf/​hfr148.PubMedCrossRef
54.
go back to reference Lipinski MJ, Biondi-Zoccai GG, Abbate A, Khianey R, Sheiban I, Bartunek J, et al. Impact of intracoronary cell therapy on left ventricular function in the setting of acute myocardial infarction: a collaborative systematic review and meta-analysis of controlled clinical trials. J Am Coll Cardiol. 2007;50(18):1761–7. doi:10.1016/j.jacc.2007.07.041.PubMedCrossRef Lipinski MJ, Biondi-Zoccai GG, Abbate A, Khianey R, Sheiban I, Bartunek J, et al. Impact of intracoronary cell therapy on left ventricular function in the setting of acute myocardial infarction: a collaborative systematic review and meta-analysis of controlled clinical trials. J Am Coll Cardiol. 2007;50(18):1761–7. doi:10.​1016/​j.​jacc.​2007.​07.​041.PubMedCrossRef
55.
56.
go back to reference Clifford DM, Fisher SA, Brunskill SJ, Doree C, Mathur A, Clarke MJ, et al. Long-term effects of autologous bone marrow stem cell treatment in acute myocardial infarction: factors that may influence outcomes. PLoS One. 2012;7(5):e37373. doi:10.1371/journal.pone.0037373.PubMedCrossRef Clifford DM, Fisher SA, Brunskill SJ, Doree C, Mathur A, Clarke MJ, et al. Long-term effects of autologous bone marrow stem cell treatment in acute myocardial infarction: factors that may influence outcomes. PLoS One. 2012;7(5):e37373. doi:10.​1371/​journal.​pone.​0037373.PubMedCrossRef
57.
go back to reference Perin EC, Willerson JT, Pepine CJ, Henry TD, Ellis SG, Zhao DX, et al. 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(16):1717–26. doi:10.1001/jama.2012.418.PubMedCrossRef Perin EC, Willerson JT, Pepine CJ, Henry TD, Ellis SG, Zhao DX, et al. 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(16):1717–26. doi:10.​1001/​jama.​2012.​418.PubMedCrossRef
58.
go back to reference • Traverse JH, Henry TD, Pepine CJ, Willerson JT, Zhao DX, Ellis SG et al. effect of the use and timing of bone marrow mononuclear cell delivery on left ventricular function after acute myocardial infarction: the TIME randomized trial. Jama. 2012:1–10. doi:10.1001/jama.2012.28726. • Traverse JH, Henry TD, Pepine CJ, Willerson JT, Zhao DX, Ellis SG et al. effect of the use and timing of bone marrow mononuclear cell delivery on left ventricular function after acute myocardial infarction: the TIME randomized trial. Jama. 2012:1–10. doi:10.1001/jama.2012.28726.
59.
go back to reference Wang X, Takagawa J, Lam VC, Haddad DJ, Tobler DL, Mok PY, et al. Donor myocardial infarction impairs the therapeutic potential of bone marrow cells by an interleukin-1-mediated inflammatory response. Sci Transl Med. 2011;3(100):100ra90. doi:10.1126/scitranslmed.3002814.PubMedCrossRef Wang X, Takagawa J, Lam VC, Haddad DJ, Tobler DL, Mok PY, et al. Donor myocardial infarction impairs the therapeutic potential of bone marrow cells by an interleukin-1-mediated inflammatory response. Sci Transl Med. 2011;3(100):100ra90. doi:10.​1126/​scitranslmed.​3002814.PubMedCrossRef
60.
go back to reference • Traverse JH, Henry TD, Ellis SG, Pepine CJ, Willerson JT, Zhao DX, et al. Effect of intracoronary delivery of autologous bone marrow mononuclear cells 2 to 3 weeks following acute myocardial infarction on left ventricular function: the LateTIME randomized trial. Jama. 2011;306(19):2110–9. doi:10.1001/jama.2011.1670.PubMedCrossRef • Traverse JH, Henry TD, Ellis SG, Pepine CJ, Willerson JT, Zhao DX, et al. Effect of intracoronary delivery of autologous bone marrow mononuclear cells 2 to 3 weeks following acute myocardial infarction on left ventricular function: the LateTIME randomized trial. Jama. 2011;306(19):2110–9. doi:10.​1001/​jama.​2011.​1670.PubMedCrossRef
61.
go back to reference • Surder D. Intracoronary infusion of BM-MNC early or late after AMI – 4 months results of the SWISS-AMI trial. Scientific Sessions of the AHA – late braking trials. 2012. References 58, 60, and 61 are all well-designed clinical trials that investigated BM-MNCs administration at different time points in patients with acute myocardial infarction and left ventricular dysfunction. • Surder D. Intracoronary infusion of BM-MNC early or late after AMI – 4 months results of the SWISS-AMI trial. Scientific Sessions of the AHA – late braking trials. 2012. References 58, 60, and 61 are all well-designed clinical trials that investigated BM-MNCs administration at different time points in patients with acute myocardial infarction and left ventricular dysfunction.
62.
go back to reference Seeger FH, Tonn T, Krzossok N, Zeiher AM, Dimmeler S. Cell isolation procedures matter: a comparison of different isolation protocols of bone marrow mononuclear cells used for cell therapy in patients with acute myocardial infarction. Eur Heart J. 2007;28(6):766–72. doi:10.1093/eurheartj/ehl509.PubMedCrossRef Seeger FH, Tonn T, Krzossok N, Zeiher AM, Dimmeler S. Cell isolation procedures matter: a comparison of different isolation protocols of bone marrow mononuclear cells used for cell therapy in patients with acute myocardial infarction. Eur Heart J. 2007;28(6):766–72. doi:10.​1093/​eurheartj/​ehl509.PubMedCrossRef
63.
go back to reference Seeger FH, Rasper T, Fischer A, Muhly-Reinholz M, Hergenreider E, Leistner DM, et al. Heparin disrupts the CXCR4/SDF-1 axis and impairs the functional capacity of bone marrow-derived mononuclear cells used for cardiovascular repair. Circ Res. 2012;111(7):854–62. doi:10.1161/CIRCRESAHA.112.265678.PubMedCrossRef Seeger FH, Rasper T, Fischer A, Muhly-Reinholz M, Hergenreider E, Leistner DM, et al. Heparin disrupts the CXCR4/SDF-1 axis and impairs the functional capacity of bone marrow-derived mononuclear cells used for cardiovascular repair. Circ Res. 2012;111(7):854–62. doi:10.​1161/​CIRCRESAHA.​112.​265678.PubMedCrossRef
65.
go back to reference Marban E, Malliaras K. Mixed results for bone marrow-derived cell therapy for ischemic heart disease. Jama. 2012:1–2. doi:10.1001/jama.2012.64751. Marban E, Malliaras K. Mixed results for bone marrow-derived cell therapy for ischemic heart disease. Jama. 2012:1–2. doi:10.1001/jama.2012.64751.
66.
go back to reference Fleissner F, Jazbutyte V, Fiedler J, Gupta SK, Yin X, Xu Q, et al. Short communication: asymmetric dimethylarginine impairs angiogenic progenitor cell function in patients with coronary artery disease through a microRNA-21-dependent mechanism. Circ Res. 2010;107(1):138–43. doi:10.1161/CIRCRESAHA.110.216770.PubMedCrossRef Fleissner F, Jazbutyte V, Fiedler J, Gupta SK, Yin X, Xu Q, et al. Short communication: asymmetric dimethylarginine impairs angiogenic progenitor cell function in patients with coronary artery disease through a microRNA-21-dependent mechanism. Circ Res. 2010;107(1):138–43. doi:10.​1161/​CIRCRESAHA.​110.​216770.PubMedCrossRef
68.
go back to reference Penn MS, Ellis S, Gandhi S, Greenbaum A, Hodes Z, Mendelsohn FO, et al. Adventitial delivery of an allogeneic bone marrow-derived adherent stem cell in acute myocardial infarction: phase I clinical study. Circ Res. 2012;110(2):304–11. doi:10.1161/CIRCRESAHA.111.253427.PubMedCrossRef Penn MS, Ellis S, Gandhi S, Greenbaum A, Hodes Z, Mendelsohn FO, et al. Adventitial delivery of an allogeneic bone marrow-derived adherent stem cell in acute myocardial infarction: phase I clinical study. Circ Res. 2012;110(2):304–11. doi:10.​1161/​CIRCRESAHA.​111.​253427.PubMedCrossRef
69.
go back to reference Wang J, Zhang S, Rabinovich B, Bidaut L, Soghomonyan S, Alauddin MM, et al. Human CD34+ cells in experimental myocardial infarction: long-term survival, sustained functional improvement, and mechanism of action. Circ Res. 2010;106(12):1904–11. doi:10.1161/CIRCRESAHA.110.221762.PubMedCrossRef Wang J, Zhang S, Rabinovich B, Bidaut L, Soghomonyan S, Alauddin MM, et al. Human CD34+ cells in experimental myocardial infarction: long-term survival, sustained functional improvement, and mechanism of action. Circ Res. 2010;106(12):1904–11. doi:10.​1161/​CIRCRESAHA.​110.​221762.PubMedCrossRef
70.
go back to reference • Losordo DW, Henry TD, Davidson C, Sup Lee J, Costa MA, Bass T, et al. Intramyocardial, autologous CD34+ cell therapy for refractory angina. Circ Res. 2011;109(4):428–36. doi:10.1161/CIRCRESAHA.111.245993. This clinical study demonstrates a reduction of angina pectoris frequency in patients with Canadian Cardiovascular Society (CCS) class III–IV refractory angina after intramyocardial delivery of CD34+ cells and emphasizes that relief of symptoms may emerge as an important target of cell-based therapies.PubMedCrossRef • Losordo DW, Henry TD, Davidson C, Sup Lee J, Costa MA, Bass T, et al. Intramyocardial, autologous CD34+ cell therapy for refractory angina. Circ Res. 2011;109(4):428–36. doi:10.​1161/​CIRCRESAHA.​111.​245993. This clinical study demonstrates a reduction of angina pectoris frequency in patients with Canadian Cardiovascular Society (CCS) class III–IV refractory angina after intramyocardial delivery of CD34+ cells and emphasizes that relief of symptoms may emerge as an important target of cell-based therapies.PubMedCrossRef
72.
go back to reference • Jeevanantham V, Butler M, Saad A, Abdel-Latif A, Zuba-Surma EK, Dawn B. Adult bone marrow cell therapy improves survival and induces long-term improvement in cardiac parameters: a systematic review and meta-analysis. Circulation. 2012;126(5):551–68. doi:10.1161/CIRCULATIONAHA.111.086074. This up-to-date meta-analysis highlights beneficial short- and long-term effects after BM-MNCs administration in 2625 patients with ischemic heart disease.PubMedCrossRef • Jeevanantham V, Butler M, Saad A, Abdel-Latif A, Zuba-Surma EK, Dawn B. Adult bone marrow cell therapy improves survival and induces long-term improvement in cardiac parameters: a systematic review and meta-analysis. Circulation. 2012;126(5):551–68. doi:10.​1161/​CIRCULATIONAHA.​111.​086074. This up-to-date meta-analysis highlights beneficial short- and long-term effects after BM-MNCs administration in 2625 patients with ischemic heart disease.PubMedCrossRef
74.
go back to reference Toma C, Pittenger MF, Cahill KS, Byrne BJ, Kessler PD. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation. 2002;105(1):93–8.PubMedCrossRef Toma C, Pittenger MF, Cahill KS, Byrne BJ, Kessler PD. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation. 2002;105(1):93–8.PubMedCrossRef
75.
go back to reference Gnecchi M, He H, Liang OD, Melo LG, Morello F, Mu H, et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat Med. 2005;11(4):367–8. doi:10.1038/nm0405-367.PubMedCrossRef Gnecchi M, He H, Liang OD, Melo LG, Morello F, Mu H, et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat Med. 2005;11(4):367–8. doi:10.​1038/​nm0405-367.PubMedCrossRef
80.
go back to reference Griffin MD, Ryan AE, Alagesan S, Lohan P, Treacy O, Ritter T. Anti-donor immune responses elicited by allogeneic mesenchymal stem cells: what have we learned so far? Immunol Cell Biol. 2012. doi:10.1038/icb.2012.67. Griffin MD, Ryan AE, Alagesan S, Lohan P, Treacy O, Ritter T. Anti-donor immune responses elicited by allogeneic mesenchymal stem cells: what have we learned so far? Immunol Cell Biol. 2012. doi:10.​1038/​icb.​2012.​67.
81.
go back to reference Quevedo HC, Hatzistergos KE, Oskouei BN, Feigenbaum GS, Rodriguez JE, Valdes D, et al. Allogeneic mesenchymal stem cells restore cardiac function in chronic ischemic cardiomyopathy via trilineage differentiating capacity. Proc Natl Acad Sci U S A. 2009;106(33):14022–7. doi:10.1073/pnas.0903201106.PubMedCrossRef Quevedo HC, Hatzistergos KE, Oskouei BN, Feigenbaum GS, Rodriguez JE, Valdes D, et al. Allogeneic mesenchymal stem cells restore cardiac function in chronic ischemic cardiomyopathy via trilineage differentiating capacity. Proc Natl Acad Sci U S A. 2009;106(33):14022–7. doi:10.​1073/​pnas.​0903201106.PubMedCrossRef
82.
go back to reference Chen SL, Fang WW, Qian J, Ye F, Liu YH, Shan SJ, et al. Improvement of cardiac function after transplantation of autologous bone marrow mesenchymal stem cells in patients with acute myocardial infarction. Chin Med J (Engl). 2004;117(10):1443–8. Chen SL, Fang WW, Qian J, Ye F, Liu YH, Shan SJ, et al. Improvement of cardiac function after transplantation of autologous bone marrow mesenchymal stem cells in patients with acute myocardial infarction. Chin Med J (Engl). 2004;117(10):1443–8.
83.
go back to reference Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, et al. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol. 2009;54(24):2277–86. doi:10.1016/j.jacc.2009.06.055.PubMedCrossRef Hare JM, Traverse JH, Henry TD, Dib N, Strumpf RK, Schulman SP, et al. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (prochymal) after acute myocardial infarction. J Am Coll Cardiol. 2009;54(24):2277–86. doi:10.​1016/​j.​jacc.​2009.​06.​055.PubMedCrossRef
84.
go back to reference •• Hare JM, Fishman JE, Gerstenblith G, Difede Velazquez DL, Zambrano JP, Suncion VY et al. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. Jama. 2012:1–11. doi:10.1001/jama.2012.25321. This clinical trial investigated transendocardial delivery of allogeneic and autologous MSCs head-to-head in patients with ischemic cardiomyopathy (ICM). •• Hare JM, Fishman JE, Gerstenblith G, Difede Velazquez DL, Zambrano JP, Suncion VY et al. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. Jama. 2012:1–11. doi:10.1001/jama.2012.25321. This clinical trial investigated transendocardial delivery of allogeneic and autologous MSCs head-to-head in patients with ischemic cardiomyopathy (ICM).
85.
86.
go back to reference Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation. 1986;74(5):1124–36.PubMedCrossRef Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation. 1986;74(5):1124–36.PubMedCrossRef
88.
go back to reference Aicher A, Heeschen C, Mildner-Rihm C, Urbich C, Ihling C, Technau-Ihling K, et al. Essential role of endothelial nitric oxide synthase for mobilization of stem and progenitor cells. Nat Med. 2003;9(11):1370–6. doi:10.1038/nm948.PubMedCrossRef Aicher A, Heeschen C, Mildner-Rihm C, Urbich C, Ihling C, Technau-Ihling K, et al. Essential role of endothelial nitric oxide synthase for mobilization of stem and progenitor cells. Nat Med. 2003;9(11):1370–6. doi:10.​1038/​nm948.PubMedCrossRef
89.
go back to reference Landmesser U, Engberding N, Bahlmann FH, Schaefer A, Wiencke A, Heineke A, et al. Statin-induced improvement of endothelial progenitor cell mobilization, myocardial neovascularization, left ventricular function, and survival after experimental myocardial infarction requires endothelial nitric oxide synthase. Circulation. 2004;110(14):1933–9. doi:10.1161/01.CIR.0000143232.67642.7A.PubMedCrossRef Landmesser U, Engberding N, Bahlmann FH, Schaefer A, Wiencke A, Heineke A, et al. Statin-induced improvement of endothelial progenitor cell mobilization, myocardial neovascularization, left ventricular function, and survival after experimental myocardial infarction requires endothelial nitric oxide synthase. Circulation. 2004;110(14):1933–9. doi:10.​1161/​01.​CIR.​0000143232.​67642.​7A.PubMedCrossRef
90.
go back to reference Bauersachs J, Bouloumie A, Fraccarollo D, Hu K, Busse R, Ertl G. Endothelial dysfunction in chronic myocardial infarction despite increased vascular endothelial nitric oxide synthase and soluble guanylate cyclase expression: role of enhanced vascular superoxide production. Circulation. 1999;100(3):292–8.PubMedCrossRef Bauersachs J, Bouloumie A, Fraccarollo D, Hu K, Busse R, Ertl G. Endothelial dysfunction in chronic myocardial infarction despite increased vascular endothelial nitric oxide synthase and soluble guanylate cyclase expression: role of enhanced vascular superoxide production. Circulation. 1999;100(3):292–8.PubMedCrossRef
91.
go back to reference Jujo K, Ii M, Sekiguchi H, Klyachko E, Misener S, Tanaka T, et al. CXCR4 Antagonist AMD3100 promotes cardiac functional recovery after ischemia-reperfusion injury via eNOS-dependent mechanism. Circulation. 2012. doi:10.1161/CIRCULATIONAHA.112.099242. Jujo K, Ii M, Sekiguchi H, Klyachko E, Misener S, Tanaka T, et al. CXCR4 Antagonist AMD3100 promotes cardiac functional recovery after ischemia-reperfusion injury via eNOS-dependent mechanism. Circulation. 2012. doi:10.​1161/​CIRCULATIONAHA.​112.​099242.
92.
go back to reference Taljaard M, Ward MR, Kutryk MJ, Courtman DW, Camack NJ, Goodman SG, et al. Rationale and design of enhanced angiogenic cell therapy in acute myocardial infarction (ENACT-AMI): the first randomized placebo-controlled trial of enhanced progenitor cell therapy for acute myocardial infarction. Am Heart J. 2010;159(3):354–60. doi:10.1016/j.ahj.2009.12.021.PubMedCrossRef Taljaard M, Ward MR, Kutryk MJ, Courtman DW, Camack NJ, Goodman SG, et al. Rationale and design of enhanced angiogenic cell therapy in acute myocardial infarction (ENACT-AMI): the first randomized placebo-controlled trial of enhanced progenitor cell therapy for acute myocardial infarction. Am Heart J. 2010;159(3):354–60. doi:10.​1016/​j.​ahj.​2009.​12.​021.PubMedCrossRef
94.
go back to reference Takehara N, Tsutsumi Y, Tateishi K, Ogata T, Tanaka H, Ueyama T, et al. Controlled delivery of basic fibroblast growth factor promotes human cardiosphere-derived cell engraftment to enhance cardiac repair for chronic myocardial infarction. J Am Coll Cardiol. 2008;52(23):1858–65. doi:10.1016/j.jacc.2008.06.052.PubMedCrossRef Takehara N, Tsutsumi Y, Tateishi K, Ogata T, Tanaka H, Ueyama T, et al. Controlled delivery of basic fibroblast growth factor promotes human cardiosphere-derived cell engraftment to enhance cardiac repair for chronic myocardial infarction. J Am Coll Cardiol. 2008;52(23):1858–65. doi:10.​1016/​j.​jacc.​2008.​06.​052.PubMedCrossRef
95.
go back to reference Behfar A, Yamada S, Crespo-Diaz R, Nesbitt JJ, Rowe LA, Perez-Terzic C, et al. Guided cardiopoiesis enhances therapeutic benefit of bone marrow human mesenchymal stem cells in chronic myocardial infarction. J Am Coll Cardiol. 2010;56(9):721–34. doi:10.1016/j.jacc.2010.03.066.PubMedCrossRef Behfar A, Yamada S, Crespo-Diaz R, Nesbitt JJ, Rowe LA, Perez-Terzic C, et al. Guided cardiopoiesis enhances therapeutic benefit of bone marrow human mesenchymal stem cells in chronic myocardial infarction. J Am Coll Cardiol. 2010;56(9):721–34. doi:10.​1016/​j.​jacc.​2010.​03.​066.PubMedCrossRef
96.
go back to reference Bartunek J, Wijns W, Dolatabadi D, Vanderheyden M, Dens J, Ostojic M, et al. C-cure multicenter trial: lineage specified bone marrow derived cardiopoietic mesenchymal stem cells for treatment of ischemic cardiomyopathy. J Am Coll Cardiol. 2011;57:E200.CrossRef Bartunek J, Wijns W, Dolatabadi D, Vanderheyden M, Dens J, Ostojic M, et al. C-cure multicenter trial: lineage specified bone marrow derived cardiopoietic mesenchymal stem cells for treatment of ischemic cardiomyopathy. J Am Coll Cardiol. 2011;57:E200.CrossRef
98.
100.
go back to reference Xu Q, Seeger FH, Castillo J, Iekushi K, Boon RA, Farcas R, et al. Micro-RNA-34a contributes to the impaired function of bone marrow-derived mononuclear cells from patients with cardiovascular disease. J Am Coll Cardiol. 2012;59(23):2107–17. doi:10.1016/j.jacc.2012.02.033.PubMedCrossRef Xu Q, Seeger FH, Castillo J, Iekushi K, Boon RA, Farcas R, et al. Micro-RNA-34a contributes to the impaired function of bone marrow-derived mononuclear cells from patients with cardiovascular disease. J Am Coll Cardiol. 2012;59(23):2107–17. doi:10.​1016/​j.​jacc.​2012.​02.​033.PubMedCrossRef
102.
go back to reference Williams AR, Hatzistergos KE, Addicott B, McCall F, Carvalho D, Suncion V, et al. Enhanced effect of human cardiac stem cells and bone marrow mesenchymal stem cells to reduce infarct size and restore cardiac function after myocardial infarction. Circulation. 2012. doi:10.1161/CIRCULATIONAHA.112.131110 [Epub ahead of print]. Williams AR, Hatzistergos KE, Addicott B, McCall F, Carvalho D, Suncion V, et al. Enhanced effect of human cardiac stem cells and bone marrow mesenchymal stem cells to reduce infarct size and restore cardiac function after myocardial infarction. Circulation. 2012. doi:10.​1161/​CIRCULATIONAHA.​112.​131110 [Epub ahead of print].
Metadata
Title
Current Status of Cell-Based Therapy for Heart Failure
Authors
Philipp Jakob
Ulf Landmesser
Publication date
01-06-2013
Publisher
Current Science Inc.
Published in
Current Heart Failure Reports / Issue 2/2013
Print ISSN: 1546-9530
Electronic ISSN: 1546-9549
DOI
https://doi.org/10.1007/s11897-013-0134-z

Other articles of this Issue 2/2013

Current Heart Failure Reports 2/2013 Go to the issue

Pathophysiology: Neuroendocrine, Vascular, and Metabolic Factors (SD Katz, Section Editor)

Chemohypersensitivity and Autonomic Modulation of Venous Capacitance in the Pathophysiology of Acute Decompensated Heart Failure

Pathophysiology: Neuroendocrine, Vascular, and Metabolic Factors (S.D. Katz, Section Editor)

Platelet Activating Factor in Heart Failure: Potential Role in Disease Progression and Novel Target for Therapy

Pathophysiology: Neuroendocrine, Vascular, and Metabolic Factors (S.D. Katz, Section Editor)

High-Intensity Aerobic Interval Exercise in Chronic Heart Failure