Scolaris Content Display Scolaris Content Display

Tratamiento con células madre para la cardiopatía isquémica crónica y la insuficiencia cardíaca congestiva

Contraer todo Desplegar todo

Referencias

Ang 2008 {published data only}

Ang KL. Intramuscular or intracoronary administration of autologous BMC fails to improve contractility of scarred myocardium: IC/IM‐BMC Study. Clinical Research in Cardiology 2008;97(1):10. CENTRAL
Ang KL, Chin D, Leyva F, Foley P, Kubal C, Chalil S, et al. Randomized, controlled trial of intramuscular or intracoronary injection of autologous bone marrow cells into scarred myocardium during CABG versus CABG alone. Nature Clinical Practice. Cardiovascular Medicine 2008;5(10):663‐70. [PUBMED: 18711405]CENTRAL
ISRCTN47591706. Efficacy of the mode of delivery of autologous bone marrow cells into heart scar muscle for the recovery of contractile function. www.isrctn.com/ISRCTN47591706 First received 30 September 2005. CENTRAL
NCT00560742. Efficacy study of intramuscular or intracoronary injection of autologous bone marrow cells to treat scarred myocardium. clinicaltrials.gov/show/NCT00560742 First received 16 November 2007. CENTRAL

Assmus 2006 {published data only}

Assmus B, Honold J, Fischer‐Rasokat U, Martin H, Schachinger V, Zeiher AM. Intraconorary cell transplantation in patients with chronic myocardial infarction: a randomized intrapatient comparison of bone marrow‐ versus blood‐derived progenitor cells. Circulation 2005;112 (17 Suppl):U644. Abstract 2756. CENTRAL
Assmus B, Honold J, Lehmann R, Pistorius K, Hoffmann WK, Martin H, et al. Transcoronary transplantation of progenitor cells and recovery of left ventricular function in patients with chronic ischemic heart disease: results of a randomized, controlled trial. Circulation 2004;110 (17 Suppl):238. Abstract 1142. CENTRAL
Assmus B, Honold J, Schachinger V, Britten MB, Fischer‐Rasokat U, Lehmann R, et al. Transcoronary transplantation of progenitor cells after myocardial infarction. New England Journal of Medicine 2006;355(12):1222‐32. [PUBMED: 16990385]CENTRAL
Assmus B, Honold J, Schachinger V, Britten MB, Fischer‐Rasokat U, Lehmann R, et al. Transcoronary transplantation of progenitor cells in patients with persistent left ventricular dysfunction after myocardial infarction: a randomized controlled trial (Topcare‐CHD). Circulation 20015;112 (17 Suppl):U694. Abstract 2984. CENTRAL
Assmus B, Honold J, Schaechinger V, Britten MB, Fischer‐Rasokat U, Lehmann R, et al. Transcoronary transplantation of progenitor cells for left ventricular dysfunction after healed myocardial infarction: a direct comparison of different cell types (TOPCARE‐CHD crossover trial). World Congress of Cardiology, 2006 September 2‐6, Barcelona, Spain. European Heart Journal 2006;27 (Suppl 1):282 Abstract P1687. CENTRAL
Bellera Gotarda MN, Schaechinger V, Fischer‐Rasokat U, Honold J, Seeger FH, Dimmeler S, et al. Impaired microvascular function as a predictor of improvement in patients with chronic post‐infarction heart failure receiving intracoronary progenitor cells ‐ results of the TOPCARE‐CHD Doppler substudy. Circulation 2008;118 (18 Suppl):Abstract 3416. CENTRAL
NCT00289822. Cell therapy for coronary heart disease. clinicaltrials.gov/show/NCT00289822 First received 8 February 2006. CENTRAL

Assmus 2013 {published data only}

Assmus B, Klotsche J, Walter DH, Seeger FH, Lutz A, Khaled W, et al. Sustained clinical benefit in patients with chronic post‐infarction heart failure treated with shockwave‐facilitated intracoronary administration of bone marrow‐derived cells: long term follow‐up of the randomized, placebo‐controlled CELLWAVE trial. American Heart Association's 2014 Scientific Sessions and Resuscitation Science Symposium, 2014 November 15‐18, Chicago, IL. Circulation 2014;130. CENTRAL
Assmus B, Walter DH, Seeger FH, Leistner DM, Lutz A, Khaled W, et al. Cardiac extracorporal shock wave application to enhance the efficiency of intracoronary cell therapy in chronic heart failure ‐ final results of the randomized, double‐blind, placebo‐controlled CELLWAVE trial. American Heart Association 2012 Scientific Sessions and Resuscitation Science Symposium, 2012 November 3‐6, Los Angeles, CA. Circulation 2012;126 (21 Suppl 1):Abstract 13050. CENTRAL
Assmus B, Walter DH, Seeger FH, Leistner DM, Lutz A, Khaled W, et al. Cardiac extracorporal shock wave application to enhance the efficiency of intracoronary cell therapy in chronic heart failure ‐ results of the randomized, double‐blind, placebo‐controlled CELLWAVE trial. American Heart Association's Scientific Sessions 2011, 2011 November 12‐16, Orlando, FL. Circulation 2011;124 (21):2372. CENTRAL
Assmus B, Walter DH, Seeger FH, Leistner DM, Steiner J, Ziegler I, et al. Effect of shock wave‐facilitated intracoronary cell therapy on LVEF in patients with chronic heart failure: the CELLWAVE randomized clinical trial. JAMA 2013;309(15):1622‐31. CENTRAL
Assmus B, Walter DH, Seeger FH, Leistner DM, Steiner J, Ziegler I, et al. Effect of shock wave‐facilitated intracoronary cell therapy on LVEF in patients with chronic heart failure: the CELLWAVE randomized clinical trial [Erratum]. JAMA 2013;309(19):1994. CENTRAL
NCT00326989. Cell‐Wave Study: Combined extracorporal shock wave therapy and intracoronary cell therapy in chronic ischemic myocardium. clinicaltrials.gov/show/NCT00326989 First received 16 May 2006. CENTRAL
Steiner JK, Ziegler I, Assmus B, Seeger F, Walter F, Walter D, et al. Cardiac extracorporal shock wave‐facilitated cell therapy in patients with chronic heart failure (Cellwave trial) ‐ mechanistic insights by magnetic resonance imaging. American Heart Association 2012 Scientific Sessions and Resuscitation Science Symposium, 2012 November 3‐6, Los Angeles, CA. Circulation 2012;126 (21 Suppl 1):Abstract 14838. CENTRAL

Bartunek 2012 {published data only}

Bartunek J, Behfar A, Dolatabadi D, Ostojic M, Dens J, Vanderheyden M, et al. Cardiopoietic stem cell therapy in heart failure: The multicenter randomized c‐cure trial. American Heart Association 2012 Scientific Sessions and Resuscitation Science Symposium, 2012 November 3‐6, Los Angeles, CA. Circulation 2012;126 (Suppl 1):Abstract 18117. CENTRAL
Bartunek J, Behfar A, Dolatabadi D, Vanderheyden M, Ostojic M, Dens J, et al. Cardiopoietic stem cell therapy in heart failure: The C‐CURE (Cardiopoietic stem Cell therapy in heart failURE) multicenter randomized trial with lineage‐specified biologics [Erratum]. Journal of the American College of Cardiology 2013;62(25):2457‐8. CENTRAL
Bartunek J, Behfar A, Dolatabadi D, Vanderheyden M, Ostojic M, Dens J, et al. Cardiopoietic stem cell therapy in heart failure: The C‐CURE (cardiopoietic stem cell therapy in heart failure) multicenter randomized trial with lineage‐specified biologics. Journal of the American College of Cardiology 2013;61(23):2329‐38. CENTRAL
Bartunek J, Behfar A, Dolatabadi D, Vanderheyden M, Ostojic M, Dens J, et al. Reply: The C‐CURE randomized clinical trial (cardiopoietic stem cell therapy in heart failure). Journal of the American College of Cardiology 2013;62(25):2454‐6. CENTRAL
Bartunek J, Dolatabadi D, Vanderheyden M, Dens J, Ostojic M, Behfar A, et al. Cardiopoietic mesenchymal stem cells for treatment of ischemic cardiomyopathy: First‐in‐man phase II multicentre clinical trial. European Heart Journal 2011;32:815. CENTRAL
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. Journal of the American College of Cardiology 2011;57 (14 Suppl 1):E200. CENTRAL
NCT00810238. C‐Cure clinical trial. clinicaltrials.gov/ct2/show/NCT00810238 First received 17 December 2008. CENTRAL

Chen 2006 {published data only}

Chen S, Liu Z, Tian N, Zhang J, Yei F, Duan B, et al. Intracoronary transplantation of autologous bone marrow mesenchymal stem cells for ischemic cardiomyopathy due to isolated chronic occluded left anterior descending artery. Journal of Invasive Cardiology 2006;18(11):552‐6. [PUBMED: 17090821]CENTRAL

Erbs 2005 {published data only}

Erbs S, Adams V, Thiele H, Emmrich F, Kluge R, Kendziorra K, et al. Intracoronary transplantation of circulating progenitor cells after recanalisation of chronic coronary artery occlusions: impact on coronary vasomotion and left ventricular remodelling. European Society of Cardiology Congress, 2005 September 3‐7, Stockholm, Sweden. European Heart Journal 2005;26 (Suppl 1):532, Abstract P3148. CENTRAL
Erbs S, Linke A, Adams V, Lenk K, Thiele H, Diederich KW, et al. Transplantation of blood‐derived progenitor cells after recanalization of chronic coronary artery occlusion: first randomized and placebo‐controlled study. Circulation Research 2005;97(8):756‐62. [PUBMED: 16151021]CENTRAL
Erbs S, Thiele H, Linke A, Adams V, Lenk K, Emmrich F, et al. Intracoronary infusion of blood‐derived progenitor cells after recanalization of chronic coronary occlusions: long term effects on cardiac function and infarct size. World Congress of Cardiology, 2006 September 2‐6, Barcelona, Spain. European Heart Journal 2006;27 (Suppl 1):274, Abstract P1656. CENTRAL
Thiele H, Schuster A, Erbs S, Adams V, Lenk K, Linke A, et al. Effects on myocardial perfusion at 3 and 15 months in recanalized chronic total occlusions ‐ randomized comparison of blood‐derived progenitor cells and inactive serum. American Heart Association Scientific Sessions 2007, 2007 November 3‐7, Orlando, FL. Circulation 2007;116 (16 Suppl):Abstract 3420. CENTRAL
Thiele H, Schuster A, Erbs S, Adams V, Linke A, Schuler G, et al. Mechanistic insights from serial cardiac magnetic resonance imaging at 3 and 15 months after application of blood‐derived progenitor cells in recanalized chronic coronary total occlusions (CTO). American Heart Association Scientific Sessions 2006, 2006 November 12‐15, Chicago, IL. Circulation 2006;114 (18 Suppl):Abstract 2616. CENTRAL
Thiele H, Schuster A, Erbs S, Linke A, Lenk K, Adams V, et al. Cardiac magnetic resonance imaging at 3 and 15 months after application of circulating progenitor cells in recanalised chronic total occlusions. International Journal of Cardiology 2009;135(3):287‐95. [PUBMED: 18584897]CENTRAL

Hamshere 2015_IC {published and unpublished data}

Hamshere S, Choudhury T, Mozid A, Agarwal S, Jones DA, Martin J, et al. Safety and efficacy of G‐CSF and autologous bone marrow‐derived cells in ischaemic cardiomyopathy: Results of the REGENERATE‐IHD Phase II trial. European Society of Cardiology, ESC Congress 2015, 2015 August 29 ‐ September 2, London, United Kingdom. European Heart Journal 2015;36 (Suppl 1):667‐8. CENTRAL
NCT00747708. Bone marrow derived adult stem cells for chronic heart failure (REGEN‐IHD). clinicaltrials.gov/show/NCT00747708 First received 4 September 2008. CENTRAL

Hamshere 2015_IM {published and unpublished data}

Hamshere S, Choudhury T, Mozid A, Agarwal S, Jones DA, Martin J, et al. Safety and efficacy of G‐CSF and autologous bone marrow‐derived cells in ischaemic cardiomyopathy: Results of the REGENERATE‐IHD Phase II trial. European Society of Cardiology, ESC Congress 2015, 2015 August 29 ‐ September 2, London, United Kingdom. European Heart Journal 2015;36 (Suppl 1):667‐8. CENTRAL
NCT00747708. Bone marrow derived adult stem cells for chronic heart failure (REGEN‐IHD). clinicaltrials.gov/show/NCT00747708 First received 4 September 2008. CENTRAL

Heldman 2014_BMMNC {published data only}

Hare JM, Heldman AW, DiFede DL, Zambrano JP, Fishman JE, Trachtenberg BH, et al. Assessment of safety and efficacy of autologous mesenchymal stem cells and whole bone marrow in patients with ischemic cardiomyopathy: the TAC‐HFT trial. American Heart Association's Scientific Sessions 2013, 2013 November 16‐20, Dallas, TX. Circulation 2013;128 (24):2713. CENTRAL
Heldman AW, DiFede DL, Fishman JE, Zambrano JP, Trachtenberg BH, Karantalis V, et al. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: the TAC‐HFT randomized trial. JAMA 2014;311(1):62‐73. CENTRAL
NCT00768066. The Transendocardial Autologous Cells (hMSC or hBMC) in Ischemic Heart Failure Trial (TAC‐HFT). clinicaltrials.gov/show/NCT00768066 First received 3 October 2008. CENTRAL
Ramireddy A, Brodt CR, DiFede DL, Mendizabal AM, Coffey JO, Viles‐Gonzalez JF. Arrhythmogenic effects of cardiac mesenchymal stem cell implantation: Results from the POSEIDON and TAC‐HFT trials. American Heart Association's 2014 Scientific Sessions and Resuscitation Science Symposium, 2014 November 15‐18, Chicago, IL. Circulation 2014;130. CENTRAL
Ramireddy A, Brodt CR, DiFede DL, Mendizabal AM, Coffey JO, Viles‐Gonzalez JF, et al. Arrhythmogenic effects of cardiac mesenchymal stem cell implantation: Results from the POSEIDON and TAC‐HFT trials. Circulation 2014;130. CENTRAL
Trachtenberg B, Velazquez DL, Williams AR, McNiece I, Fishman J, Nguyen K, et al. Rational and design of the transendocardial injection of autologous human cells (bone marrow or mesenchymal) in chronic ischemic left ventricular dysfunction and heart failure secondary to myocardial infarction (TAC‐HFT) trial: a randomized, double‐blind, placebo‐controlled study of safety and efficacy. American Heart Journal 2011;161(3):487‐93. CENTRAL
Williams AR, Trachtenberg B, Velazquez DL, Altman P, Rouy D, Mendizabal A, et al. Preliminary results from the transendocardial injections of autologous whole bone marrow and mesenchymal stem cells in ischemic heart failure (TAC‐HFT) trial. Journal of the American College of Cardiology 2011;57 (14 Suppl 1):E242. CENTRAL
Wong Po Foo C, Rouy D, Hare J, Heldman A, DiFede D, McNiece I, et al. The transendocardial autologous cells in ischemic heart failure trial bone marrow mononuclear cells (TAC‐HFT‐BMC) randomized placebo controlled blinded study. World Conference on Regenerative Medicine 2015, 2015 October 21‐23, Leipzig, Germany. Regenerative Medicine 2015;10 (7 Suppl 1):169. CENTRAL

Heldman 2014_BM‐MSC {published data only}

Hare JM, Heldman AW, DiFede DL, Zambrano JP, Fishman JE, Trachtenberg BH, et al. Assessment of safety and efficacy of autologous mesenchymal stem cells and whole bone marrow in patients with ischemic cardiomyopathy: the TAC‐HFT trial. American Heart Association's Scientific Sessions 2013, 2013 November 16‐20, Dallas, TX. Circulation 2013;128 (24):2713. CENTRAL
Heldman AW, DiFede DL, Fishman JE, Zambrano JP, Trachtenberg BH, Karantalis V, et al. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: the TAC‐HFT randomized trial. JAMA 2014;311(1):62‐73. CENTRAL
NCT00768066. The Transendocardial Autologous Cells (hMSC or hBMC) in Ischemic Heart Failure Trial (TAC‐HFT). clinicaltrials.gov/show/NCT00768066 First received 3 October 2008. CENTRAL
Ramireddy A, Brodt CR, DiFede DL, Mendizabal AM, Coffey JO, Viles‐Gonzalez JF. Arrhythmogenic effects of cardiac mesenchymal stem cell implantation: Results from the POSEIDON and TAC‐HFT trials. American Heart Association's 2014 Scientific Sessions and Resuscitation Science Symposium, 2014 November 15‐18, Chicago, IL. Circulation 2014;130. CENTRAL
Ramireddy A, Brodt CR, DiFede DL, Mendizabal AM, Coffey JO, Viles‐Gonzalez JF, et al. Arrhythmogenic effects of cardiac mesenchymal stem cell implantation: Results from the POSEIDON and TAC‐HFT trials. Circulation 2014;130. CENTRAL
Trachtenberg B, Velazquez DL, Williams AR, McNiece I, Fishman J, Nguyen K, et al. Rationale and design of the Transendocardial Injection of Autologous Human Cells (bone marrow or mesenchymal) in Chronic Ischemic Left Ventricular Dysfunction and Heart Failure Secondary to Myocardial Infarction (TAC‐HFT) trial: a randomized, double‐blind, placebo‐controlled study of safety and efficacy. American Heart Journal 2011;161(3):487‐93. CENTRAL
Williams AR, Trachtenberg B, Velazquez DL, Altman P, Rouy D, Mendizabal A, et al. Preliminary results from the transendocardial injections of autologous whole bone marrow and mesenchymal stem cells in ischemic heart failure (TAC‐HFT) trial. Journal of the American College of Cardiology 2011;57 (14 Suppl 1):E242. CENTRAL
Wong Po Foo C, Rouy D, Hare J, Heldman A, DiFede D, McNiece I, et al. The transendocardial autologous cells in ischemic heart failure trial bone marrow mononuclear cells (TAC‐HFT‐BMC) randomized placebo controlled blinded study. World Conference on Regenerative Medicine 2015, 2015 October 21‐23, Leipzig, Germany. Regenerative Medicine 2015;10 (7 Suppl 1):169. CENTRAL

Hendrikx 2006 {published data only}

Hendrikx M, Hensen K, Clijsters C, Jongen H, Koninckx R, Bijnens E, et al. Recovery of regional but not global contractile function by the direct intramyocardial autologous bone marrow transplantation: results from a randomized controlled clinical trial. Circulation2006; Vol. 114, issue 1 Suppl:I101‐7. [PUBMED: 16820557]CENTRAL

Honold 2012 {published data only}

Honold J, Fischer‐Rasokat U, Lehmann R, Leistner DM, Seeger FH, Schachinger V, et al. G‐CSF stimulation and coronary reinfusion of mobilized circulating mononuclear proangiogenic cells in patients with chronic ischemic heart disease: Five‐year results of the TOPCARE‐G‐CSF trial. Cell Transplantation 2012;21(11):2325‐37. CENTRAL

Hu 2011 {published data only}

Duan F, Qi Z, Liu S, Lv X, Wang H, Gao Y, et al. Effectiveness of bone marrow mononuclear cells delivered through a graft vessel for patients with previous myocardial infarction and chronic heart failure: an echocardiographic study of left ventricular modeling. Medical Ultrasonography 2015;17(2):160‐6. CENTRAL
Hu S, Liu S, Zheng Z, Yuan X, Li L, Lu M, et al. Isolated coronary artery bypass graft combined with bone marrow mononuclear cells delivered through a graft vessel for patients with previous myocardial infarction and chronic heart failure: a single‐center, randomized, double‐blind, placebo‐controlled clinical trial. Journal of the American College of Cardiology 2011;57(24):2409‐15. [PUBMED: 21658561]CENTRAL
Lu M, Liu S, Zheng Z, Yin G, Song L, Chen H, et al. A pilot trial of autologous bone marrow mononuclear cell transplantation through grafting artery: A sub‐study focused on segmental left ventricular function recovery and scar reduction. International Journal of Cardiology 2013;168(3):2221‐7. CENTRAL
Lu MJ, Zhao SH, Liu S, Zhang PH, Jiang SL, Zhang Y, et al. Assessment of therapeutic effects of stem cell transplantation in heart failure patients with old myocardial infarction by magnetic resonance imaging. Chinese Journal of Cardiology 2008;36(11):969‐74. CENTRAL
NCT00395811. Stem cell therapy to improve myocardial function in patients undergoing coronary artery bypass grafting (CABG). clinicaltrials.gov/ct2/show/NCT00395811 First received 1 November 2006. CENTRAL
Qi Z, Duan F, Liu S, Lv X, Wang H, Gao Y. Effect of bone marrow mononuclear cells delivered through a graft vessel for patients with previous myocardial infarction and chronic heart failure: An echocardiographic study of left atrium function. Echocardiography 2015;32(6):937‐46. CENTRAL

Jimenez‐Quevedo 2011 {published data only}

Jimenez‐Quevedo P, Gonzalez FJ, Llorente L, Sabate M, Garcia MX, Hernandez‐Antolin R, et al. Selected CD133+ endothelial progenitor cells to create angiogenesis in no‐option patients: The design of the PROGENITOR randomized trial. European Heart Journal 2011;32:816: Abstract P4654. CENTRAL
Jimenez‐Quevedo P, Gonzalez‐Ferrer JJ, Sabat M, Garcia‐Mol X, Llorent L, Hernandez‐Antoli R, et al. Selected CD133+ endothelial progenitor cells to create angiogenesis in no‐option patients: Preliminary 3‐months results of the progenitor trial. American Heart Association 2012 Scientific Sessions and Resuscitation Science Symposium, 2012 November 3‐6, Los Angeles, CA. Circulation 2012;126(21 (Suppl 1)). CENTRAL
Jimenez‐Quevedo P, Gonzalez‐Ferrer JJ, Sabate M, Garcia‐Moll X, Alfonso F, Hernandez‐Antolin R, et al. Selected CD133+ endothelial progenitor cells to create angiogenesis in no‐option patients: Preliminary results of safety and feasibility. Journal of the American College of Cardiology 2012;60:B112. CENTRAL
Jimenez‐Quevedo P, Gonzalez‐Ferrer JJ, Sabate M, Garcia‐Moll X, Delgado‐Bolton R, Llorente L, et al. Selected CD133+ progenitor cells to promote angiogenesis in patients with refractory angina: final results of the PROGENITOR randomized trial. Circulation Research 2014;115(11):950‐60. CENTRAL
Jimenez‐Quevedo P, Gonzalez‐Ferrer JJ, Sabate M, Moll XG, Hernandez‐Antolin R, Delgado‐Bolton R, et al. Selected CD133+ endothelial progenitor cells to create angiogenesis in patients with refractory angina. Final results of the PROGENITOR trial. Circulation 2013;128 (22 Suppl):Abstract 14006. CENTRAL
NCT00694642. Safety and efficacy of autologous endothelial progenitor cell CD 133 for therapeutic angiogenesis (PROGENITOR). clinicaltrials.gov/ct2/show/NCT00694642 First received 5 June 2008. CENTRAL

Losordo 2007 {published data only}

Losordo DW, Henry TD, Schatz RA, Sup Lee J, Costa M, Bass T, et al. Randomized, double‐blind, placebo‐controlled trial of autologous CD34+ cell therapy for refractory angina: 2‐year safety analysis. American Heart Association Scientific Sessions, 2010 November 13‐17, Chicago, IL. Circulation 2010;122 (21 Suppl 1):Abstract A15621. CENTRAL
Losordo DW, Kearney M, Patel S, Poh K‐K, Shah P, Welt F, et al. Randomized, double‐blind, placebo controlled pilot trial of intramyocardial autologous CD34 cell therapy for intractable angina. American Heart Association Scientific Sessions 2006, 2006 November 12‐15, Chicago, IL. Circulation 2006;114 (18 Suppl):Abstract 3361. CENTRAL
Losordo DW, Schatz RA, White CJ, Udelson JE, Veereshwarayya V, Durgin M, et al. Intramyocardial transplantation of autologous CD34+ stem cells for intractable angina: a phase I/IIa double‐blind, randomized controlled trial. Circulation 2007;115(25):3165‐72. [PUBMED: 17562958]CENTRAL

Losordo 2011 {published data only}

Junge CE, Motlagh D, Debelak J, Cohen A, Hammel S, Nada A, et al. Clinical parameters influencing cell mobilization and impact of mobilization ability on efficacy outcomes: An analysis from ACT34‐CMI. American Heart Association 2012 Scientific Sessions and Resuscitation Science Symposium, 2012 November 3‐6, Los Angeles, CA. Circulation 2012;126 (21 Suppl 1):Abstract 12015. CENTRAL
Livingston D, Motlagh D, Debelak J, Cohen A, Story K, Hammel S, et al. Phase 2 study of intramyocardial injection of autologous CD34+ cells to treat subjects with refractory chronic myocardial ischemia (CMI): Factors influencing mobilization and apheresis. 51st Annual Meeting of the American Society of Hematology, ASH, 2009 December 5‐8, New Orleans, LA. Blood 2009;114:Abstract 3227. CENTRAL
Losordo D, Motlagh D, Cohen A, Junge C, Nada A, Story K. Impact of mobilization ability on cell functionality and comparison of normal and CMI subject samples: An analysis from ACT34‐CMI. 62nd Annual Scientific Session of the American College of Cardiology and i2 Summit: Innovation in Intervention, ACC.13, 2013 March 9‐11, San Francisco, CA. Journal of the American College of Cardiology 2013;61 (10 Suppl 1):E1817. CENTRAL
Losordo DW, Henry T, Schatz RA, Lee JS, Costa M, Bass T, et al. Autologous CD34+ cell therapy for refractory angina: 12 month results of the phase II ACT34‐CMI study. American Heart Association Scientific Sessions 2009, 2009 November 14‐18, Orlando, FL. Circulation 2009;120 (18 Suppl):Abstract 5638. CENTRAL
Losordo DW, Henry TD, Davidson C, Sup Lee J, Costa MA, Bass T, et al. ACT34‐CMI Investigators. Intramyocardial, autologous CD34+ cell therapy for refractory angina. Circulation Research 2011;109(4):428‐36. CENTRAL
NCT00300053. ACT34‐CMI ‐ Adult autologous CD34+ stem cells. clinicaltrials.gov/ct2/show/NCT00300053 First received 6 March 2006. CENTRAL
Povsic TJ, Losordo DW, Story K, Junge CE, Schatz RA, Harrington RA, et al. Cardiac biomarker elevation during stem cell mobilisation, apheresis and intramyocardial delivery is common but does not impact incidence of long‐term MACE: An analysis from ACT34‐CMI. American Heart Association 2012 Scientific Sessions and Resuscitation Science Symposium, 2012 November 3‐6, Los Angeles, CA. Circulation 2012;126 (21 Suppl 1):Abstract 11770. CENTRAL
Povsik TJ, Losordo DW, Story K, Junge CE, Schatz RA, Harrington RA, et al. Incidence and clinical significance of cardiac biomarker elevation during stem cell mobilization, apheresis, and intramyocardial delivery: An analysis from ACT34‐CMI. American Heart Journal 2012;164(5):689‐97.e3. CENTRAL

Mathiasen 2015 {published data only}

Mathiasen AB, Jorgensen E, Qayyum E, Haack‐Sorensen M, Ekblond A, Kastrup J. Rationale and design of the first randomized, double‐blind, placebo‐controlled trial of intramyocardial injection of autologous bone‐marrow derived Mesenchymal Stromal Cells in chronic ischemic Heart Failure (MSC‐HF Trial). American Heart Journal 2012;164(3):285‐91. CENTRAL
Mathiasen AB, Qayyum AA, Jorgensen E, Helqvist S, Fischer‐Nielsen A, Kofoed KF. Bone marrow‐derived mesenchymal stromal cell treatment in patients with severe ischaemic heart failure: a randomized placebo‐controlled trial (MSC‐HF trial). European Heart Journal 2015;36(27):1744‐53. CENTRAL
NCT00644410. Autologous mesenchymal stromal cell therapy in heart failure. clinicaltrials.gov/show/NCT00644410 First received 20 March 2008. CENTRAL

Mozid 2014_IC {published data only}

Mozid A, Arnous S, Yeo C, Brookman P, Preston M, Archbold A, et al. Head‐to‐head comparison of different delivery methods of autologous bone marrow progenitor cells in chronic ischaemic heart failure. European Society of Cardiology Congress, 2010 August 28‐ September 1, Stockholm, Sweden. European Heart Journal 2011;32 (16 Suppl 1):456. CENTRAL
Mozid A, Yeo C, Arnous S, Ako E, Saunders N, Locca D, et al. Safety and feasibility of intramyocardial versus intracoronary delivery of autologous cell therapy in advanced heart failure: the REGENERATE‐IHD pilot study. Regenerative Medicine 2014;9(3):269‐78. CENTRAL
Mozid AM, Holstensson M, Choudhury T, Ben‐Haim S, Allie R, Martin J, et al. Clinical feasibility study to detect angiogenesis following bone marrow stem cell transplantation in chronic ischaemic heart failure. Nuclear Medicine Communications 2014;35(8):839‐48. CENTRAL
Papalia F, Mozid AM, Davies LC, Mathur A. Incidence of left ventricular thrombus in patients with severe ischaemic left ventricular systolic dysfunction. Heart Failure 2012, 2012 May 19‐22, Belgrade, Serbia. European Journal of Heart Failure, Supplement 2012;11:161‐2. CENTRAL
Yeo C, Locca D, Wong J, Burchell T, Preston M, Brookman P, et al. Ejection fraction and NYHA class in heart failure in the REGENERATE‐IHD stem cell study: A comparison between the intramyocardial intracoronary arms. American Journal of Cardiology 2009;104 (6 Suppl 1):190D‐1D. CENTRAL
Yeo C, Mathur A. Autologous bone marrow‐derived stem cells for ischemic heart failure: REGENERATE‐IHD trial. Regenerative Medicine 2009;4(1):119‐27. CENTRAL

Mozid 2014_IM {published data only}

Mozid A, Arnous S, Yeo C, Brookman P, Preston M, Archbold A, et al. Head‐to‐head comparison of different delivery methods of autologous bone marrow progenitor cells in chronic ischaemic heart failure. European Society of Cardiology Congress, 2010 August 28‐September 1, Stockholm, Sweden. European Heart Journal 2011;32 (16 Suppl 1):456. CENTRAL
Mozid A, Yeo C, Arnous S, Ako E, Saunders N, Locca D, et al. Safety and feasibility of intramyocardial versus intracoronary delivery of autologous cell therapy in advanced heart failure: the REGENERATE‐IHD pilot study. Regenerative Medicine 2014;9(3):269‐78. CENTRAL
Mozid AM, Holstensson M, Choudhury T, Ben‐Haim S, Allie R, Martin J, et al. Clinical feasibility study to detect angiogenesis following bone marrow stem cell transplantation in chronic ischaemic heart failure. Nuclear Medicine Communications 2014;35(8):839‐48. CENTRAL
Papalia F, Mozid AM, Davies LC, Mathur A. Incidence of left ventricular thrombus in patients with severe ischaemic left ventricular systolic dysfunction. Heart Failure 2012, 2012 May 19‐22, Belgrade, Serbia. European Journal of Heart Failure, Supplement 2012;11:161‐2. CENTRAL
Yeo C, Locca D, Wong J, Burchell T, Preston M, Brookman P, et al. Ejection fraction and NYHA class in heart failure in the REGENERATE‐IHD stem cell study: A comparison between the intramyocardial intracoronary arms. American Journal of Cardiology 2009;104 (6 Suppl 1):190D‐1D. CENTRAL
Yeo C, Mathur A. Autologous bone marrow‐derived stem cells for ischemic heart failure: REGENERATE‐IHD trial. Regenerative Medicine 2009;4(1):119‐27. CENTRAL

Nasseri 2012 {published data only}

NCT00462774. Bypass surgery and CD133 marrow cell injection for treatment of ischemic heart failure (Cardio133). www.clinicaltrials.gov/ct2/show/NCT00462774 First received 18 April 2007. CENTRAL
Nasseri BA, Ebell W, Dandel M, Kukucka M, Gebker R, Doltra A, et al. Autologous CD133+ bone marrow cells and bypass grafting for regeneration of ischaemic myocardium: The CARDIO133 trial. European Heart Journal 2014;35(19):1263‐74. CENTRAL
Nasseri BA, Kikucha M, Dandel M, Ebell W, Hetzer R, Stamm C. Autologous CD133+ bone marrow cells and bypass grafting for regeneration of ischemic myocardium: Results of the CARDIO133 trial. Journal of the American College of Cardiology 2012;59 (13 Suppl 1):E864. CENTRAL
Nasseri BA, Klose K, Ebell W, Dandel M, Kukucka M, Gebker R, et al. Improved regional contractile function and reduced scar size after clinical cell therapy with CD133‐positive cells. 3rd EACTS Meeting on Cardiac and Pulmonary Regeneration, 2012 December 14‐15, Berlin, Germany. Interactive Cardiovascular and Thoracic Surgery 2013;16 (2):S233. CENTRAL
Nasseri BA, Kukucka M, Dandel M, Ebell W, Gebker R, Hetzer R, et al. Results of the Cardio133 trial: A randomized double‐blinded controlled trial of intramyocardial injection of autologous CD133+ bone marrow cells during bypass grafting. 42nd Annual Meeting of the German Society for Cardiovascular and Thoracic Surgery, 2013 February 17‐20, Freiburg, Germany. Thoracic and Cardiovascular Surgeon 2013;61. CENTRAL

Patel 2005 {published data only}

Patel AN, Geffner L, Vina RF, Saslavsky J, Urschel HC, Kormos R, et al. Surgical treatment for congestive heart failure with autologous adult stem cell transplantation: a prospective randomized study. Journal of Thoracic and Cardiovascular Surgery 2005;130(6):1631‐8. [PUBMED: 16308009]CENTRAL
Patel AN, Mittal S, Vina RF, Benetti F, Trehan N. Long term follow‐up of coronary artery bypass grafting with autologous bone marrow cell therapy. 20th Annual Meeting of the International Society for Cell Therapy, ISCT 2014, 23‐26 April 2014, Paris, France. Cytotherapy 2014;16:S39. CENTRAL

Patel 2015 {published data only}

NCT01299324. Retrograde delivery of BMAC (bone marrow aspirate concentrate) for congestive heart failure. clinicaltrials.gov/show/NCT01299324 First received 10 January 2011. CENTRAL
Patel AN, Ince H, Mittal S, Turan G, Trehan N. Retrograde delivery of autologous concentrated bone marrow cell therapy for patients with congestive heart failure ‐ REVIVE‐1 a prospective randomized study. 20th Annual Meeting of the International Society for Cell Therapy, ISCT 2014, 2014 April 23‐26, Paris, France. Cytotherapy 2014;16:S39. CENTRAL
Patel AN, Mittal S, Turan G, Winters AA, Henry TD, Ince H, et al. REVIVE Trial: Retrograde delivery of autologous bone marrow in patients with heart failure. Stem Cells in Translational Medicine 2015;4(9):1021‐7. CENTRAL

Patila 2014 {published data only}

Lehtinen M, Patila T, Kankuri E, Lauerma K, Sinisalo J, Laine M, et al. Intramyocardial bone marrow mononuclear cell transplantation in ischemic heart failure: Long‐term follow‐up. Journal of Heart and Lung Transplantation 2015;34(7):899‐905. CENTRAL
Lehtinen M, Patila T, Vento A, Kankuri E, Suojaranta‐Ylinen R, Poyhia R. Prospective, randomized, double‐blinded trial of bone marrow cell transplantation combined with coronary surgery ‐ perioperative safety study. Interactive Cardiovascular and Thoracic Surgery 2014;19(6):990‐6. CENTRAL
NCT00418418. Combined CABG and stem‐cell transplantation for heart failure. clinicaltrials.gov/show/NCT00418418 First received 3 January 2007. CENTRAL
Patila T, Lehtinen M, Vento A, Schildt J, Sinisalo J, Laine M. Bone marrow mononuclear cells for ischemic cardiac failure ‐ A prospective, controlled, randomized, double‐blinded study of cell transplantation combined with coronary bypass surgery. 20th Annual Meeting of the International Society for Cell Therapy, ISCT 2014, 2014 April 23‐26, Paris, France. Cytotherapy 2014;16:S9‐S10. CENTRAL
Patila T, Lehtinen M, Vento A, Schildt J, Sinisalo J, Laine M, et al. Autologous bone marrow mononuclear cell transplantation in ischemic heart failure: A prospective, controlled, randomized, double‐blind study of cell transplantation combined with coronary bypass. Journal of Heart and Lung Transplantation 2014;33(6):567‐74. CENTRAL

Perin 2011 {published data only}

NCT00203203. Autologous stem cells for cardiac angiogenesis (FOCUS HF). clinicaltrials.gov/show/NCT00203203 First received 12 September 2005. CENTRAL
Perin EC, Silva GV, Fernandes MR, Henry T, Moore W, Coulter S, et al. FOCUS‐HF: The first US randomized blinded controlled trial of transendocardial injection of bone marrow mononuclear cells in chronic severe ischemic heart failure patients. American College of Cardiology 58th Annual Scientific Session and i2 Summit: Innovation in Intervention, 2009 September 29‐31, Orlando, FL. Journal of the American College of Cardiology 2009;53 (10 Suppl):A193, Abstract 1051‐191. CENTRAL
Perin EC, Silva GV, Henry TD, Cabreira‐Hansen MG, Moore WH, Coulter SA, et al. A randomized study of transendocardial injection of autologous bone marrow mononuclear cells and cell function analysis in ischemic heart failure (FOCUS‐HF). American Heart Journal 2011;161(6):1078‐87.e3. [PUBMED: 21641354]CENTRAL

Perin 2012a {published data only}

NCT00824005. Effectiveness of stem cell treatment for adults with ischemic cardiomyopathy (The FOCUS Study). clinicaltrials.gov/ct2/show/NCT00824005 First received 15 January 2009. CENTRAL
Perin EC, Willerson TJ, Pepine CJ, Henry TD, Ellis SG, Zhao DX, et al. Cardiovascular Cell Therapy Research Network CCTRN. 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. CENTRAL
Willerson JT, Perin EC, Ellis SG, Pepine CJ, Henry TD, Zhao DX, et al. Intramyocardial injection of autologous bone marrow mononuclear cells for patients with chronic ischemic heart disease and left ventricular dysfunction (First Mononuclear Cells injected in the US [FOCUS]): Rationale and design. American Heart Journal 2010;160(2):215‐23. [PUBMED: 20691824]CENTRAL

Perin 2012b {published data only}

NCT00314366. Injection of autologous aldehyde dehydrogenase‐bright stem cells for therapeutic angiogenesis (FOCUS Br). clinicaltrials.gov/show/NCT00314366 First received 10 April 2006. CENTRAL
Perin EC, Silva GV, Zheng Y, Fernandez MR, Moore W, Coulter S, et al. First in man transendocardial injection of autologous aldehyde dehydrogenase‐bright cells in heart failure patients (FOCUS‐Bright). American Heart Association Scientific Sessions 2009; 2009 November 14‐18, Orlando, FL. Circulation 2009;120 (18 Suppl):Abstract 3502. CENTRAL
Perin EC, Silva GV, Zheng Y, Gahremanpour A, Canales J, Patel D, et al. Randomized, double‐blind pilot study of transendocardial injection of autologous aldehyde dehydrogenase‐bright stem cells in patients with ischemic heart failure. American Heart Journal 2012;163(3):415‐21, 421.e1. [PUBMED: 22424012]CENTRAL

Pokushalov 2010 {published data only}

Pokushalov E, Romanov A, Artemenko S, Cherniavskiy A, Larionov P, Terehov I, et al. Efficacy of intramyocardial injections of autologous bone marrow mononuclear stem cells in patients with ischemic heart failure: Long term results: ACC poster contributions. Amercan College of Cardiology's 59th Annual Scientific Session and i2 Summit: Innovation in Intervention, 2010 March 14‐16, Atlanta, GA. Journal of the American College of Cardiology 2010;55 (10 Suppl 1):A24: Abstract E228. CENTRAL
Pokushalov E, Romanov A, Artemenko S, Larionov P, Cherniavskiy A. Efficiency of intramyocardial injections of autologous bone marrow mononuclear stem cells in patients with ischemic heart failure: Long‐term results. 13th Annual Scientific Meeting of the Heart Failure Society of America, 2009 September 13‐16, Boston, MA. Journal of Cardiac Failure 2009;15 (6 Suppl 1):S44, Abstract 138. CENTRAL
Pokushalov E, Romanov A, Cherniavskiy A, Artemenko S, Larionov P, Terehov I, et al. Efficiency of intramyocardial injections of autologous bone marrow mononuclear stem cells in patients with ischemic heart failure: Long‐term results. Transcatheter Cardiovascular Therapeutics Symposium, 2009 September 21‐15, San Francisco, CA. American Journal of Cardiology 2009;104 (6 Suppl 1):74D‐5D, Abstract TCT‐194. CENTRAL
Pokushalov E, Romanov A, Chernyavsky A, Larionov P, Terekhov I, Artyomenko S, et al. Efficiency of intramyocardial injections of autologous bone marrow mononuclear cells in patients with ischemic heart failure: a randomized study. Journal of Cardiovascular Translational Research 2010;3(2):160‐8. [PUBMED: 20560030]CENTRAL
Romanov A, Pokushalov E, Artemenko S, Larionov P, Terehov I, Kliver E, et al. Efficiency of intramyocardial injections of autologous bone marrow mononuclear stem cells in patients with ischemic heart failure: long‐term results. European Society of Cardiology Congress 2009, 2009 August 29‐September 2, Barcelona, Spain. European Heart Journal 2009;30 (Suppl 1):504, Abstract P3097. CENTRAL
Romanov A, Pokushalov E, Cherniavskiy A, Artemenko S, Larionov P, Terehov I, et al. Efficiency of intramyocardial injections of autologous bone marrow mononuclear cells in patients with ischemic heart failure: A randomized study. 31st Annual Scientific Sessions of the Heart Rhythm Society, 2010 May 12‐15, Denver, CO. Heart Rhythm 2010;7 (5 Suppl 1):S348: Abstract P05‐82. CENTRAL
Romanov A, Pokushalov E, Cherniavskiy A, Larionov P, Artemenko S, Terekhov I, et al. Efficiency of intramyocardial injections of autologous bone marrow mononuclear stem cells in patients with ischemic heart failure: long‐term results. Heart Failure Congress, 2009 May 30‐June 2, Nice, France. European Journal of Heart Failure 2009;8 (Suppl 2):ii705, Abstract 1401. CENTRAL
Romanov A, Pokushalov E, Cherniavskiy A, Larionov P, Terekhov I, Kilver E, et al. Efficiency of intramyocardial injections of autologous bone marrow mononuclear cells in patients with ischemic heart failure: A randomized study. Heart Failure Congress 2010, 2010 May 29‐June 1, Berlin, Germany. European Journal of Heart Failure 2010;9 (Suppl 1):S60‐S61, Abstract 404. CENTRAL
Romanov A, Pokushalov E, Cherniavskiy A, Larionov P, Terekhov I, Poveschenko O, et al. Efficiency of intramyocardial injections of autologous bone marrow mononuclear cells in patients with ischemic heart failure: a randomized study. European Society of Cardiology Congress, 2010 August 28‐September 1, Stockholm, Sweden. European Heart Journal 2010;31 (Abstract Supplement):323, Abstract 2024. CENTRAL
Romanov A, Pokushalov E, Prohorova D, Chemyavsky A, Larionov P, Terekhov I, et al. Cardiac resynchronization therapy and bone marrow cell transplantation in patients with ischemic heart failure and electro‐mechanical dyssynchrony: A randomized pilot study. 17th World Congress in Cardiac Electrophysiology and Cardiac Techniques, Cardiostim 2010, 2010 June 16‐19, Nice, France. Europace 2010;12:i58. CENTRAL
Romanov A, Pokushalov E, Prokhorova D, Cherniavskiy A, Artemenko S, Shirokova N, et al. Cardiac resynchronization therapy and bone marrow transplantation in patients with ischemic heart failure and electro‐mechanical dyssynchrony. A randomized study. European Society of Cardiology Congress, 2010 August 28‐September 1, Stockholm, Sweden. European Heart Journal 2010;31 (17 Suppl 1):591; Abstract 3476. CENTRAL
Romanov AB, Pokushalov E, Cherniavskiy A, Kliver E, Karaskov A, Dib N. Efficiency of intramyocardial injections of autologous bone marrow mononuclear cells in patients with ischemic heart failure: A randomized study. 60th Annual Scientific Session and Expo ACC, 2011 April 2‐5, New Orleans, LA. Journal of the American College of Cardiology 2011;57 (14 Suppl 1):Abstract E241. CENTRAL

Santoso 2014 {published data only}

ACTRN12611000219987. A study of the effect of direct endomyocardial injection of autologous bone marrow cells on left ventricular ejection function in patients with "end‐stage" ischaemic heart failure. https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=335320 First received 30 March 2010. CENTRAL
HKUCTR‐763. Direct endomyocardial injection of autologous bone marrow cells for treatment of patients with "end‐stage" ischemic heart failure. www.hkuctr.com First received 16 October 2008. CENTRAL
NCT01150175. Direct endomyocardial injection of autologous bone marrow cells to treat ischaemic heart failure (END‐HF). clinicaltrials.gov/show/NCT01150175 First received 24 May 2010. CENTRAL
Santoso T, Siu CW, Irawan C, Chan WS, Alwi I, Yiu KH, et al. A randomized placebo controlled trial of endomyocardial implantation of autologous bone marrow mononuclear cells in advanced ischemic heart failure (END‐HF). European Society of Cardiology, ESC Congress 2013, 2013 August 31‐September 4, Amsterdam, Netherlands. European Heart Journal 2013;34:652. CENTRAL
Santoso T, Siu CW, Irawan C, Chan WS, Alwi I, Yiu KH, et al. Endomyocardial implantation of autologous bone marrow mononuclear cells in advanced ischemic heart failure: a randomized placebo‐controlled trial (END‐HF). Journal of Cardiovascular Translational Research 2014;7(6):545‐52. CENTRAL

Trifunovic 2015 {published data only}

Trifunovic Z, Obradovic S, Balint B, Ilic R, Vukic Z, Sisic M, et al. Functional recovery of patients with ischemic cardiomyopathy treated with coronary artery bypass surgery and concomitant intramyocardial bone marrow mononuclear cell implantation ‐ a long‐term follow‐up study. Vojnosanit Pregl 2015;72(3):225‐32. CENTRAL

Tse 2007 {published data only}

Chan CW, 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. Journal of Cardiovascular Magnetic Resonance 2010;12:6. [PUBMED: 20100336]CENTRAL
Tse HF, Thambar S, Kwong YL, Rowlings P, Bellamy G, McCrohon J, et al. Prospective randomized trial of direct endomyocardial implantation of bone marrow cells for treatment of severe coronary artery diseases (PROTECT‐CAD trial). European Heart Journal 2007;28(24):2998‐3005. [PUBMED: 17984132]CENTRAL

Turan 2011 {published data only}

Turan RG, Bozdag‐Turan I, Ortak J, Akin I, Kische S, Schneider H, et al. Improved mobilization of the CD34(+) and CD133(+) bone marrow‐derived circulating progenitor cells by freshly isolated intracoronary bone marrow cell transplantation in patients with ischemic heart disease. Stem Cells and Development 2011;20(9):1491‐501. [PUBMED: 21190450]CENTRAL
Turan RG, Bozdag‐Turan I, Ortak J, Kische S, Akin I, Schneider H, et al. Improved functional activity of bone marrow derived circulating progenitor cells after intra coronary freshly isolated bone marrow cells transplantation in patients with ischemic heart disease. Stem Cell Reviews 2011;7(3):646‐56. [PUBMED: 21188654]CENTRAL

Van Ramshorst 2009 {published data only}

ISRCTN58194927. Randomised, double blind, placebo controlled trial of intramyocardial injection of autologous bone marrow cells in no‐option patients with refractory angina pectoris and documented ischaemia. www.isrctn.com/ISRCTN58194927 First received 27 January 2006. CENTRAL
NTR400. Randomized, double blind, placebo controlled trial of intramyocardial injection of autologous bone marrow cells in no‐option patients with refractory angina pectoris and documented ischemia. www.trialregister.nl/trialreg/admin/rctview.asp?TC=400 First received 27 January 2006. CENTRAL
Rodrigo S, Van Ramshorst J, Beeres SL, Al Younis I, Dibbets‐Schneider P, De Roos A, et al. 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. American Heart Journal 2012;164(5):771‐8. CENTRAL
Rodrigo S, Van Ramshorst J, Beeres SL, Dibbets‐Schneider P, De Roos A, Fibbe WE, et al. Intramyocardial injection of bone marrow mononuclear cells in patients with chronic myocardial ischemia: An intra‐patient comparison. Circulation 2011;124(Suppl 1):A15175. CENTRAL
Rodrigo S, Van Ramshorst J, Beeres SL, Dibbets‐Schneider P, Stokkel M, Fibbe WE, et al. Intramyocardial bone marrow cell injection in patients with chronic myocardial ischemia: Results long term follow‐up. American Heart Association Scientific Sessions, 2011 November 12‐16, Orlando, FL. Circulation 2011;124 (21 Suppl 1):Abstract A15175. CENTRAL
Rodrigo S, Van Ramshorst J, Beeres SL, Dibbets‐Schneider P, Stokkel M, Zwaginga JJ, et al. Intramyocardial bone marrow cell injection in patients with chronic myocardial ischemia: Results long term follow‐up. European Society of Cardiology Congress 2011, 2011 Augst 27‐31, Paris, France. European Heart Journal 2011;32:820, Abstract P4671. CENTRAL
Van Ramshorst J, Antoni ML, Beeres SL, Roes SD, Delgado V, Rodrigo SF, et al. Intramyocardial bone marrow‐derived mononuclear cell injection for chronic myocardial ischemia: the effect on diastolic function. Circulation: Cardiovascular Imaging 2011;4(2):122‐9. [PUBMED: 21209073]CENTRAL
Van Ramshorst J, Bax JJ, Beeres SL, Dibbets‐Schneider P, Roes SD, Stokkel MP, et al. Intramyocardial bone marrow cell injection for chronic myocardial ischemia: a randomized controlled trial. JAMA 2009;301(19):1997‐2004. [PUBMED: 19454638]CENTRAL
Van Ramshorst J, Bax JJ, Beeres SL, Dibbets‐Schneider P, Roes SD, Stokkel MPM, et al. Intramyocardial injection of bone marrow‐derived mononuclear cells for chronic myocardial ischemia: a randomized, double‐blind, placebo‐controlled trial. European Society of Cardiology Congress, 2009 August 29‐September 2, Barcelona, Spain. European Heart Journal 2009;30 (Suppl 1):452, Abstract 2819. CENTRAL
Van Ramshorst J, Bax JJ, Beeres SL, Roes SD, Dibbets P, De Roos A, et al. Intramyocardial autologous bone marrow cell injection in no‐option patients with refractory angina pectoris and documented ischemia: A randomized, double blinded, placebo‐controlled trial. American College of Cardiology 58th Annual Scientific Session and i2 Summit: Innovation in Intervention, 2009 March 29‐31, Orlando, FL. Journal of the American College of Cardiology 2009;53 (10 Suppl):A340, Abstract 1041‐145. CENTRAL
van Ramshorst J, Beeres SL, Rodrigo SF, Dibbets‐Schneider P, Scholte AJ, Fibbe WE, et al. Effect of intramyocardial bone marrow‐derived mononuclear cell injection on cardiac sympathetic innervation in patients with chronic myocardial ischemia. International Journal of Cardiovascular Imaging 2014;30(3):583‐9. CENTRAL

Wang 2009 {published data only}

Wang S‐H, Cui J‐Y, Lu M, Wang X, Li, X‐M, Tan C, et al. Intracoronary transplantation with autologous bone marrow CD34+ stem cells for angina: a randomized controlled clinical analysis. Journal of Clinical Rehabilitative Tissue Engineering Research 2009;13(14):2623‐6. CENTRAL

Wang 2010 {published data only}

Shihong W. Intracoronary autologous CD34+ stem cell therapy for intractable angina. Heart 2012;98 (Suppl 2):E42‐3. CENTRAL
Wang S, Cui J, Peng W, Lu M. Intracoronary autologous CD34+ stem cell therapy for intractable angina. Cardiology 2010;117(2):140‐7. [PUBMED: 20975266]CENTRAL

Wang 2014 {published data only}

Wang X, Bai M, Huang S, Jie Q. Autologous CD133+ bone marrow cells for regeneration of ischaemic myocardium. 25th Great Wall International Congress of Cardiology, Asia Pacific Heart Congress 2014, and the International Congress Cardiovascular Prevention and Rehabilitation 2014, 2014 October 16‐19, Beijing, China. Journal of the American College of Cardiology 2014;64 (16 Suppl 1):C116. CENTRAL

Wang 2015 {published data only}

Wang H, Wang Z, Jiang H, Ma D, Zhou W, Zhang G, et al. Effect of autologous bone marrow cell transplantation combined with off‐pump coronary artery bypass grafting on cardiac function in patients with chronic myocardial infarction. Cardiology 2015;130(1):27‐33. CENTRAL

Yao 2008 {published data only}

Yao K, Huang R, Qian J, Cui J, Ge L, Li Y, et al. Administration of intracoronary bone marrow mononuclear cells on chronic myocardial infarction improves diastolic function. Heart (British Cardiac Society) 2008;94(9):1147‐53. [PUBMED: 18381377]CENTRAL

Zhao 2008 {published data only}

Zhao Q, Sun Y, Xia L, Chen A, Wang Z. Randomized study of mononuclear bone marrow cell transplantation in patients with coronary surgery. Annals of Thoracic Surgery 2008;86(6):1833‐40. [PUBMED: 19021989]CENTRAL

Ascheim 2014 {published data only}

Ascheim DD, Gelijns AC, Goldstein D, Moye LA, Smedira N, Lee S, et al. Mesenchymal precursor cells as adjunctive therapy in recipients of contemporary left ventricular assist devices. Circulation 2014;129(22):2287‐96. CENTRAL
NCT01442129. The effect of intramyocardial injection of mesenchymal precursor cells on myocardial function in patients undergoing LVAD implantation. clinicaltrials.gov/ct2/show/NCT01442129 First received 26 September 2011. CENTRAL

Assmann 2014 {published data only}

Assmann A, Heke M, Kropil P, Ptok L, Hafner D, Ohmann C, et al. Laser‐supported CD133+ cell therapy in patients with ischemic cardiomyopathy: Initial results from a prospective phase I multicenter trial. PLoS ONE 2014;9(7):e101449. CENTRAL

Beeres 2006 {published data only}

Beeres SL, Bax JJ, Dibbets‐Schneider P, Stokkel MP, Fibbe WE, Van der Wall EE, et al. Sustained effect of autologous bone marrow mononuclear cell injection in patients with refractory angina pectoris and chronic myocardial ischemia: twelve‐month follow‐up results. American Heart Journal 2006;152(4):684.e11‐6. [PUBMED: 16996834]CENTRAL
Rodrigo SF, van Ramshorst J, Mann I, Leong DP, Cannegieter SC, Al Younis I, et al. Predictors of response to intramyocardial bone marrow cell treatment in patients with refractory angina and chronic myocardial ischemia. International Journal of Cardiology 2014;175(3):539‐44. CENTRAL

Beeres 2007 {published data only}

Beeres SL, Bax JJ, Zeppenfeld K, Dibbets‐Schneider P, Stokkel MP, Fibbe WE, et al. Feasibility of trans‐endocardial cell transplantation in chronic ischaemia. Heart (British Cardiac Society)2007; Vol. 93, issue 1:113‐4. [PUBMED: 17170348]CENTRAL

Beeres 2007a {published data only}

Beeres SL, Bax JJ, Dibbets‐Schneider P, Stokkel MP, Fibbe WE, Van der Wall EE, et al. Intramyocardial injection of autologous bone marrow mononuclear cells in patients with chronic myocardial infarction and severe left ventricular dysfunction. American Journal of Cardiology 2007;100(7):1094‐8. [PUBMED: 17884369]CENTRAL

Beeres 2007b {published data only}

Beeres SL, Bengel FM, Bartunek J, Atsma DE, Hill JM, Vanderheyden M, et al. Role of imaging in cardiac stem cell therapy. Journal of the American College of Cardiology 2007;49(11):1137‐48. [PUBMED: 17367656]CENTRAL

Bittencourt 2008 {published data only}

Bittencourt MS, Schettert IT, Grupi CJ, Cesar LAM, Krieger JE, Dallan LAO, et al. Resting electrocardiographic variables of patients with severe coronary artery disease undergoing coronary artery bypass graft surgery plus intramyocardial bone marrow cell injection: A one‐year follow up study. Circulation 2008;118(12):E225. CENTRAL

Bolli 2011 {published data only}

Bolli R, Chugh A, D'Amario D, Loughran JH, Stoddard MF, Ikram S, et al. Effect of cardiac stem cells in patients with ischemic cardiomyopathy: Interim results of the SCIPIO trial up to 2 years after therapy. American Heart Association 2012 Scientific Sessions and Resuscitation Science Symposium, 2012 November 3‐6, Los Angeles, CA. Circulation 2012;126 (23)(23):2784. CENTRAL
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. CENTRAL
NCT00474461. Cardiac stem cell infusion in patients with ischemic cardiomyopathy (SCIPIO). clinicaltrials.gov/ct2/show/NCT00474461 First received 15 May 2007. CENTRAL

Chang 2006 {published data only}

Chang SA, Kim HK, Lee HY, Kang HJ, Kim YJ, Zo JH, et al. Restoration of synchronicity of the left ventricular myocardial contraction with stem cell therapy: New insights into the therapeutic implication of stem cell therapy in myocardial infarction. American Heart Association Scientific Sessions 2006 2006 November 12‐15, Chicago, IL. Circulation 2006;114 (18 Suppl):Abstract 2718. CENTRAL

Charwat 2010 {published data only}

Charwat S, Lang I, Dettke M, Graf S, Nyolczas N, Hemetsberger R, et al. Effect of intramyocardial delivery of autologous bone marrow mononuclear stem cells on the regional myocardial perfusion. NOGA‐guided subanalysis of the MYSTAR prospective randomised study. Thrombosis and Haemostasis 2010;103(3):564‐71. [PUBMED: 20076851]CENTRAL

Chen 2014 {published data only}

Chen XM, Cui DY, Zhang M. Clinical observation of stem cell transplantation in patients with acute myocardial infarction complicated with heart failure. Journal of Dalian Medical University 2014;36(2):157‐9. CENTRAL

Chin 2010 {published data only}

Chin SP, Poey AE, Chang SK, Wong CY, Lam KH, Cheong SK. Safety and efficacy of autologous mesenchymal stem cells for the treatment of end‐stage dilated cardiomyopathy ‐ a comparison of intracoronary and direct intramyocardial injection. European Society of Cardiology Congress 2010 August 28‐September 1, Stockholm, Sweden. European Heart Journal 2010;31 (17 Suppl 1):79‐80, Abstract P601. CENTRAL

EUCTR2006‐005628‐17‐ES {published data only}

EUCTR2006‐005628‐17‐ES. Open study with blind regulator on the effectiveness of autologous bone marrow mononuclear cells in patients with left ventricular dysfunction after myocardia infarction [Estudio abierto con evaluador ciego de la eficacia de las células mononucleares autólogas de médula ósea en la regeneración muscular y vascular en pacientes con disfunción ventricular izquierda tras infarto de miocardio]. https://www.clinicaltrialsregister.eu/ctr‐search/search?query=2006‐005628‐17 First received 19 February 2007. CENTRAL

EUCTR2009‐017924‐18‐NL {published data only}

EUCTR2009‐017924‐18‐NL. Efficacy assessment of repeat intramyocardial injection of autologous bone marrow cells in previously responding no‐option patients with residual refractory angina pectoris and documented ischemia. https://www.clinicaltrialsregister.eu/ctr‐search/search?query=2009‐017924‐18 First received 17 June 2010. CENTRAL

Fuchs 2004 {published data only}

Fuchs S, Kornowski R, Weisz G, Satler LF, Smits PC, Okubagzi P, et al. Transendocardial autologous bone marrow cell transplantation in patients with advanced ischemic heart disease: Final results from a multi‐center feasibility study. Journal of the American College of Cardiology 2004;5 (Suppl A):99A. CENTRAL

Gu 2011 {published data only}

Gu X, Xie Y, Gu J, Sun L, He S, Xu R, et al. Repeated intracoronary infusion of peripheral blood stem cells with G‐CSF in patients with refractory ischemic heart failure ‐ a pilot study. Circulation Journal 2011;75(4):955‐63. [PUBMED: 21325723]CENTRAL

Haack‐Sorensen 2013 {published data only}

Haack‐Sorensen M, Friis T, Mathiasen AB, Jorgensen E, Hansen L, Dickmeiss E, et al. Direct intramyocardial mesenchymal stromal cell injections in patients with severe refractory angina: one‐year follow‐up. Cell Transplantation 2013;22(3):521‐8. CENTRAL

Jimenez‐Quevedo 2008 {published data only}

Jimenez‐Quevedo P, Silva GV, Sanz‐Ruiz R, Oliveira EM, Fernandes MR, Angeli F, et al. Diabetic and nondiabetic patients respond differently to transendocardial injection of bone marrow mononuclear cells: Findings from prospective clinical trials in "no‐option" patients. Revista Espanola de Cardiologia 2008;61(6):635‐9. CENTRAL

Kang 2006 {published data only}

Kang HJ, Kim MK, Lee HY, Park KW, Lee W, Cho YS, et al. Five‐year results of intracoronary infusion of the mobilized peripheral blood stem cells by granulocyte colony‐stimulating factor in patients with myocardial infarction. European Heart Journal 2012;33(24):3062‐9. CENTRAL
Kang HJ, Lee HY, Na SH, Chang SA, Park KW, Kim HK, et al. Differential effect of intracoronary infusion of mobilized peripheral blood stem cells by granulocyte colony‐stimulating factor on left ventricular function and remodeling in patients with acute myocardial infarction versus old myocardial infarction: the MAGIC Cell‐3‐DES randomized, controlled trial. Circulation 2006;114(1 Suppl):I145‐51. [PUBMED: 16820564]CENTRAL

Kang 2006b {published data only}

Kang H‐J, Kim H‐S, Na S‐H, Zhang S‐Y, Kang WJ, Youn T‐J, et al. Six months follow up results of "granulocytes‐colony stimulating factor" based stem cell therapy in patients with myocardial infarction: MAGIC cell randomized controlled trial. Korean Circulation Journal 2006;36(2):99‐107. CENTRAL

Karantalis 2014 {published data only}

Karantalis V, DiFede DL, Gerstenblith G, Pham S, Symes J, Zambrano JP, et al. Autologous mesenchymal stem cells produce concordant improvements in regional function, tissue perfusion, and fibrotic burden when administered to patients undergoing coronary artery bypass grafting. American Heart Association; 2013 Scientific Sessions and Resuscitation Science Symposium, 2013 November 16‐20, Dallas, TX. Circulation 2013;128 (22 Suppl 1). CENTRAL
Karantalis V, DiFede DL, Gerstenblith G, Pham S, Symes J, Zambrano JP, et al. Autologous mesenchymal stem cells produce concordant improvements in regional function, tissue perfusion, and fibrotic burden when administered to patients undergoing coronary artery bypass grafting: The Prospective Randomized Study of Mesenchymal Stem Cell Therapy in Patients Undergoing Cardiac Surgery (PROMETHEUS) trial. Circulation Research 2014;114(8):1302‐10. CENTRAL
NCT00587990. Prospective randomized study of mesenchymal stem cell therapy in patients undergoing cardiac surgery (PROMETHEUS). clinicaltrials.gov/ct2/show/NCT00587990 First received 2 January 2008. CENTRAL

Koestering 2005 {published data only}

Brehm M, Koestering M, Zeus T, Bartsch T, Turan G, Antke C, et al. Improvement of heart function in chronic coronary heart disease with chronic myocardial infarction: Controlled study with intracoronary autologous mononuclear bone marrow cell transplantation (IACT‐Study). Circulation 2005;111 (13):1721. CENTRAL
Koestering M, Bartsch T, Brehm M, Zeus T, Turan RG, Schannwell CM. Long term follow‐up of improvement of human infarcted heart muscle by intracoronary autologous mononuclear bone marrow cells in chronic coronary artery disease (IACT‐Study). European Heart Journal 2006;27 (Suppl 1):281‐2. CENTRAL
Koestering M, Bartsch T, Zeus T, Brehm M, Scharmwell CM, Strauer BE. Long term (2,5 year's) follow‐up by intracoronary infusion of autologous mononuclear bone marrow cells in chronic coronary artery disease after is associated with improvement of left ventricular function in human infarcted heart muscle (IACT‐Study). Circulation 2007;116 (16 Suppl):289. Abstract 1406. CENTRAL
Koestering M, Brehm M, Zeus T, Antke C, Koegler G, Wernet P, et al. Regeneration of human heart function in chronic coronary heart disease with chronic myocardial infarction: Controlled study with intracoronary autologous mononuclear bone marrow cell transplantation. European Heart Journal 2005;26 (Suppl 1):532. CENTRAL
Koestering M, Zeus T, Brehm M, Bartsch T, Scharmwell C, Strauer BE. Regeneration of human infarcted heart muscle in chronic coronary artery disease after myocardial infarction: Controlled study with intracoronary autologous mononuclear bone marrow cell transplantation (IACT‐study). Circulation 2007;116 (Suppl):767‐8. Abstract 3400. CENTRAL
Koestering M, Zeus T, Brehm M, Bartsch T, Turan G, Mohammadi H, et al. Regeneration of human infarcted heart function by intracoronary autologous bone marrow cell transplantation in chronic coronary artery disease: Controlled study with intracoronary autologous mononuclear bone marrow cell transplantation (IACT‐Study). Circulation 2006;114 (18 Suppl):139. CENTRAL
Koestering M, Zeus T, Brehm M, Bartsch T, Turan GR, Schannwell MC, et al. Improvement of heart function in chronic coronary heart disease with chronic myocardial infarction: Controlled study with intracoronary autologous mononuclear bone marrow cell transplantation (IACT‐Study). Circulation 2005;112 (17 Suppl):U334. CENTRAL
Koestering M, Zeus T, Brehm M, Bartsche T, Schannwell CM, Mueller HW, et al. Improvement of heart function in chronic coronary heart disease with chronic myocardial infarction: Controlled study with intracoronary autologous mononuclear bone marrow cell transplantation. European Heart Journal 2007;27 (Suppl 1):276‐7. CENTRAL

Lai 2009 {published data only}

Ang KL, Lai VK, Rathbone WE, Harvey NH, Galinanes M. Randomized controlled trial on the cardioprotective effect of bone marrow cells in patients undergoing coronary artery bypass graft surgery. European Journal of Heart Failure 2009;8:ii707. CENTRAL
Lai VK, Ang KL, Rathbone W, Harvey NJ, Galinanes M. Randomized controlled trial on the cardioprotective effect of bone marrow cells in patients undergoing coronary bypass graft surgery. European Heart Journal 2009;30(19):2354‐9. [PUBMED: 19561024]CENTRAL

Lee 2015 {published data only}

ISRCTN26002902. Intra‐coronary transfusion of autologous CD34+ cells improves left ventricular function in patients with diffuse coronary artery disease (CAD) and non‐candidates for coronary artery intervention. www.isrctn.com/ISRCTN26002902 First received 12 August 2013. CENTRAL
ISRCTN72853206. Intra‐coronary transfusion of autologous CD34+ cells improves left ventricular function in patients with diffuse coronary artery disease and non candidates for coronary artery intervention. www.isrctn.com/ISRCTN72853206 First received 30 January 2012. CENTRAL
Lee F, Chen Y, Chua S, Fu M, Pei S, Yip H. Intra‐coronary transfusion of circulatory derived CD34+ cells improves left ventricular function in patients with diffuse coronary artery disease and non candidates for coronary artery intervention. 35th Annual Meeting and Scientific Sessions of the International Society for Heart and Lung Transplantation, ISHLT 2015, 2015 April 15‐18, Nice, France. Journal of Heart and Lung Transplantation 2015;34 (Suppl 1):S183. CENTRAL
Lee FY, Chen YL, Sung PH, Ma MC, Pei SN, Wu CJ, et al. Intracoronary transfusion of circulation‐derived CD34+ cells improves left ventricular function in patients with end‐stage diffuse coronary artery disease unsuitable for coronary intervention. Critical Care Medicine 2015;43(10):2117‐32. CENTRAL

Makkar 2011 {published data only}

Makkar R, Smith RR, Cheng K, Malliaras K, Marban L, Thomson L, et al. The CADUCIUS (cardiosphere‐derived autologous stem cells to reverse ventricular dysfunction) trial. American Heart Association's Scientific Sessions 2011, 2011 November 12‐16, Orlando, FL. Circulation 2011;124 (21):2373. CENTRAL

Mann 2015 {published data only}

Mann I, Rodrigo SF, van Ramshorst J, Beeres SL, Dibbets‐Schneider P, de Roos A, et al. Repeated intramyocardial bone marrow cell injection in previously responding patients with refractory angina again improves myocardial perfusion, anginal complaints and quality of life. Circulation: Cardiovascular Interventions 2015;8:8. CENTRAL

Maroto 2010 {published data only}

Maroto L, San Roman A, Di Stefano S, Rey J, Fulquet E, Arevalo A, et al. Transplantation of unselected autologous bone marrow mononuclear cells in subacute myocardial infarction. 59th International Congress of the European Society for Cardiovascular Surgery, ESCVS 2010, 2010 April 15‐18, Izmir, Turkey. Interactive Cardiovascular and Thoracic Surgery 2010;10:S170‐1. CENTRAL

Maureira 2012 {published data only}

Maureira P, Tran N, Djaballah W, Angioi M, Bensoussan D, Didot N, et al. Residual viability is a predictor of the perfusion enhancement obtained with the cell therapy of chronic myocardial infarction: a pilot multimodal imaging study. Clinical Nuclear Medicine 2012;37(8):738‐42. [PUBMED: 22785499]CENTRAL

Mocini 2006 {published data only}

Mocini D, Staibano M, Mele L, Giannantoni P, Menichella G, Colivicchi F, et al. Autologous bone marrow mononuclear cell transplantation in patients undergoing coronary artery bypass grafting. American Heart Journal 2006;151(1):192‐7. CENTRAL

Nagaya 2007 {published data only}

Nagaya N, Ohgushi H, Shimizu W, Yamagishi M, Noguchi T, Noda T, et al. Clinical trial of autologous bone marrow mesenchymal stem cell transplantation for severe chronic heart failure. Circulation 2007;116 (16 Suppl):453. CENTRAL

NCT00285454 {published data only}

NCT00285454. Cell repair in heart failure. clinicaltrials.gov/show/NCT00285454 First received 27 January 2006. CENTRAL

NCT00289822 {published data only}

NCT00289822. Cell therapy for coronary heart disease. clinicaltrials.gov/show/NCT00289822 First received 8 February 2006. CENTRAL

NCT00362388 {published data only}

NCT00362388. Cell therapy in chronic ischemic heart disease. clinicaltrials.gov/show/NCT00362388 First received 8 August 2006. CENTRAL

NCT01074099 {published data only}

NCT01074099. Feasibility study of BMAC enhanced CABG. clinicaltrials.gov/show/NCT01074099 First received 1 February 2010. CENTRAL

NCT01337011 {published data only}

NCT01337011. Intra‐coronary versus intramyocardial application of enriched CD133pos autologous bone marrow derived stem cells (AlsterMACS). clinicaltrials.gov/show/NCT01337011 First received 14 April 2011. CENTRAL

NCT01666132 {published data only}

NCT01666132. METHOD ‐ Bone marrow derived mononuclear cells in chronic ischemic disease. clinicaltrials.gov/show/NCT01666132 First received 27 June 2011. CENTRAL
Surder D, Radrizzani M, Turchetto L, Cicero VL, Soncin S, Muzzarelli S, et al. Combined delivery of bone marrow‐derived mononuclear cells in chronic ischemic heart disease: rationale and study design. Clinical Cardiology 2013;36(8):435‐41. CENTRAL

NCT01693042 {unpublished data only}

Assmus B, Alakmeh S, De Rosa S, Bonig H, Hermann E, Levy WC, et al. Improved outcome with repeated intracoronary injection of bone marrow‐derived cells within a registry: rationale for the randomized outcome trial REPEAT. European Heart Journal 2015 Oct 29 [Epub ahead of print]. CENTRAL
NCT01693042. Compare the effects of single versus repeated intracoronary application of autologous bone marrow‐derived mononuclear cells on mortality in patients with chronic post‐infarction heart failure (REPEAT). clinicaltrials.gov/ct2/show/NCT01693042 First received 19 September 2012. CENTRAL

NCT01721902 {published data only}

NCT01721902. Stem cell implantation in patients undergoing CABG. clinicaltrials.gov/show/NCT01721902 First received 1 October 2012. CENTRAL

Perin 2003 {published data only}

Dohmann HFR, Perin EC, Borojevic H, Sousa ALS, Silva SA, Carvalho AC, et al. Transendocardial autologous bone‐marrow cell transplantation in severe, chronic ischaemic heart failure: Prospective controlled study. European Heart Journal 2003;24 (Suppl):715. CENTRAL
Dohmann HFR, Perin EC, Borojevic R, Silva SA, Souza ALS, Silva GV, et al. Sustained improvement in symptoms and exercise capacity up to six months after autologous transendocardial transplantation of bone marrow mononuclear cells in patients with severe ischemic heart disease. Arquivos Brasileiros de Cardiologia 2005;84(5):360‐6. CENTRAL
Perin EC, Dohmann HF, Borojevic R, Silva SA, Sousa AL, Mesquita CT, et al. Transendocardial, autologous bone marrow cell transplantation for severe, chronic ischemic heart failure. Circulation 2003;107(18):2294‐302. [PUBMED: 12707230]CENTRAL
Perin EC, Dohmann HF, Borojevic R, Silva SA, Sousa AL, Silva GV, et al. Improved exercise capacity and ischemia 6 and 12 months after transendocardial injection of autologous bone marrow mononuclear cells for ischemic cardiomyopathy. Circulation 2004;110(11 Suppl 1):II213‐8. [PUBMED: 15364865]CENTRAL
Perin EC, Dohmann HF, Borojevic R, Sousa ALS, Dohmann H, Carvalho AC, et al. Improvement in symptoms and exercise capacity at eight weeks in a controlled study of autologous bone marrow cell transplant in humans with severe ischemic heart failure. Journal of the American College of Cardiology 2003;6 (Suppl A):44A. CENTRAL

Peruga 2009 {published data only}

Peruga J, Plewka M, Kasprzak J, Jezewski T, Wierzbicka A, Robak T, et al. Intracoronary administration of stem cells in patients with acute myocardial infarction ‐ angiographic follow‐up. Kardiologia Polska 2009;67(5):477‐84. [PUBMED: 19521932]CENTRAL

Poglajen 2013 {published data only}

Poglajen G, Sever M, Cukjati M, Cernelc P, Knezevic I, Zemljic G, et al. Effects of transendocardial CD34+ cell transplantation in patients with ischemic cardiomyopathy. Circulation: Cardiovascular Interventions 2014;7(4):552‐9. CENTRAL
Poglajen G, Sever M, Mali P, Haddad F, Wu JC, Vrtovec B. Effects of transendocardial CD34+ stem cell transplantation in patients with ischaemic cardiomyopathy. American Heart Association 2013 Scientific Sessions and Resuscitation Science Symposium, 2013 November 16‐20, Dallas, TX. Circulation 2013;128 (22 Suppl 1). CENTRAL
Poglajen G, Zemljic G, Sever M, Cukjati M, Haddad F, Wu JC, et al. Comparison of clinical effects following multi‐site versus single‐site CD34+ cell injections in patients with ischemic cardiomyopathy. American Heart Association's 2014 Scientific Sessions and Resuscitation Science Symposium, 2014 November 15‐18, Chicago, IL. Circulation 2014;130. CENTRAL

Pokushalov 2011 {published data only}

Pokushalov E, Romanov A, Corbucci G, Prohorova D, Chernyavsky A, Larionov P, et al. Cardiac resynchronization therapy and bone marrow cell transplantation in patients with ischemic heart failure and electromechanical dyssynchrony: a randomized pilot study. Journal of Cardiovascular Translational Research 2011;4(6):767‐78. [PUBMED: 21547598]CENTRAL
Pokushalov E, Romanov A, Prohorova D, Artemenko S, Cheriniavskiy A, Karaskov A, et al. Cardiac resynchronization therapy in patients with ischemic heart failure after bone marrow cell transplantation. Journal of the American College of Cardiology 2011;57 (14 Suppl 1):Abstract E103. CENTRAL

Premer 2014 {published data only}

Premer C, Blum A, Bellio M, Schulman IH, Sierra J, Delgado C, et al. Intracardiac mesenchymal stem cells restore endothelial function in heart failure by stimulating the release of endothelial progenitor cells. American Heart Association's 2014 Scientific Sessions and Resuscitation Science Symposium, 2014 November 15‐18, Chicago, IL. Circulation 2014;130. CENTRAL
Premer C, Blum A, Bellio MA, Schulman IH, Hurwitz BE, Parker M, et al. Allogeneic mesenchymal stem cells restore endothelial function in heart failure by stimulating endothelial progenitor cells. EBioMedicine 2015;2(5):467‐75. CENTRAL

Qin 2015 {published data only}

Qin J, Guo Y, Chen X, Liu X. Intracoronary cardiosphere‐derived cells for heart regeneration after myocardial infarction. 26th Great Wall International Congress of Cardiology, Asia Pacific Heart Congress 2015 and the International Congress of Cardiovascular Prevention and Rehabilitation 2015, 2015 October 29‐November 1, Beijing, China. Journal of the American College of Cardiology 2015;66(16 (Suppl 1)):C30. CENTRAL

Rivas‐Plata 2010 {published data only}

Rivas‐Plata A, Castillo J, Pariona M, Chunga A. Bypass grafts and cell transplant in heart failure with low ejection fraction. Asian Cardiovascular & Thoracic Annals 2010;18(5):425‐9. [PUBMED: 20947595]CENTRAL

Shen 2007 {published data only}

Shen ZY, Yu GP, Hu YQ. The experimental study on the effect to cardiac function by intramyocardial transplantation of mobilized autologous bone marrow stem cells after myocardial infarction. Xenotransplantation 2007;14(5):539. CENTRAL

Stamm 2007a {published data only}

Stamm C, Kleine HD, Choi YH, Dunkelmann S, Lauffs JA, Lorenzen B, et al. Intramyocardial delivery of CD133+ bone marrow cells and coronary artery bypass grafting for chronic ischemic heart disease: safety and efficacy studies. Journal of Thoracic and Cardiovascular Surgery 2007;133(3):717‐25. [PUBMED: 17320570]CENTRAL

Suncion 2014 {published data only}

Suncion VY, Ghersin E, Fishman JE, Zambrano JP, Karantalis V, Mandel N, et al. Does transendocardial injection of mesenchymal stem cells improve myocardial function locally or globally? An analysis from the percutaneous stem cell injection delivery effects on neomyogenesis (POSEIDON) randomized trial. Circulation Research 2014;14(8):1292‐301. CENTRAL

Takehara 2012 {published data only}

Takehara N, Ogata T, Nakata M, Kami D, Nakamura T, Matoba S, et al. The ALCADIA (autologous human cardiac‐derived stem cell to treat ischemic cardiomyopathy) trial. Circulation 2012;126(23):2783. CENTRAL

Tuma 2010 {published data only}

Tuma J, Fernandez R, Cruz C, Carrillo A, Erchilla J, Inga L, et al. Long term benefit of autologous bone marrow transplantation by retrograde technique in terminal heart failure (LIBERTY study). Journal of the American College of Cardiology 2010;56 (13 Suppl 1):B71. CENTRAL

Tuma 2011 {published data only}

Tuma J, Carrasco A, Vina RF, Chirinos S, Curz C, Patel AN. Five year follow‐up of coronary sinus delivery of bone marrow cells for congestive heart failure. 20th Annual Meeting of the International Society for Cell Therapy, ISCT 2014, 2014 April 23‐26, Paris, France. Cytotherapy 2014;16:S42. CENTRAL
Tuma J, Carrasco A, Winters AA, Chirinos S, Patel AN. Long term follow‐up of coronary sinus delivery of bone marrow cells for congestive heart failure. 35th Annual Meeting and Scientific Sessions of the International Society for Heart and Lung Transplantation, ISHLT 2015, 2015 April 15‐18, Nice, France. Journal of Heart and Lung Transplantation 2015;34 (4 Suppl 1):S181‐2. CENTRAL
Tuma J, Fernandez‐Vina R, Carrasco A, Castillo J, Cruz C, Carrillo A, et al. Safety and feasibility of percutaneous retrograde coronary sinus delivery of autologous bone marrow mononuclear cell transplantation in patients with chronic refractory angina. Journal of Translational Medicine 2011;9:183. [PUBMED: 22029669]CENTRAL

Vicario 2004 {published data only}

Vicario J, Campo C, Piva J, Faccio F, Gerardo L, Becker C, et al. One‐year follow‐up of transcoronary sinus administration of autologous bone marrow in patients with chronic refractory angina. Cardiovascular Revascularization Medicine: including Molecular Interventions 2005;6(3):99‐107. CENTRAL
Vicario J, Campos C, Piva J, Faccio F, Gerardo L, Becker C, et al. Transcoronary sinus administration of autologous bone marrow in patients with chronic refractory stable angina Phase 1. Cardiovascular Radiation Medicine 2004;5(2):71‐6. [PUBMED: 15464943]CENTRAL

Vrtovec 2015 {published data only}

Vrtovec B, Poglajen G, Zemljic G, Sever M, Cukhati M, Haddad F, et al. Response to CD34+ cell therapy is associated with myocardial scar burden in patients with ischemic and non‐ischemic chronic heart failure. 35th Annual Meeting and Scientific Sessions of the International Society for Heart and Lung Transplantation, ISHLT 2015, 2015 April 14‐18, Nice, France. Journal of Heart and Lung Transplantation 2015;34 (Suppl 1):S90‐1. CENTRAL

Wang 2006 {published data only}

Wang WM, Sun NL, Liu J, Zhang P, Liu KY, Wang Q, et al. Effects of intracoronary autologous bone marrow mononuclear cells transplantation in patients with anterior myocardial infarction. Zhonghua Xin Xue Guan Bing Za Zhi 2006;34(2):103‐6. [PUBMED: 16626572]CENTRAL

Referencias de los estudios en espera de evaluación

Ahmadi 2010 {published data only}

Ahmadi S, Soleymani M, Sahebjam M, Zorofian A, Ahmadbeigi N, Karimi A, et al. Treatment of heart failure with expanded autologous bone marrow‐derived mesenchymal/CD133+ stem/progenitor cell transplantation during CABG. 59th International Congress of the European Society for Cardiovascular Surgery, ESCVS 2010, 2010 April 15‐18, Izmir, Turkey. Interactive Cardiovascular and Thoracic Surgery 2010;10 (Suppl 1):S80. CENTRAL

Ahmadi 2015 {published data only}

Ahmadi SH, Soleymani M, Sahebjam M, Karimi AA, Madani CM. Which stem cell type is more efficient as an adjunctive to surgical treatment of severe ischemic heart failure: expanded mesenchymal or recycling stem cells? International Academy of Cardiology 20th World Congress on Heart Disease Annual Scientific Sessions 2015, 2015 July 25‐27, Vancouver, Canada. Cardiology (Switzerland) 2015;131:100. CENTRAL

Cuzzola 2007 {published data only}

Cuzzola M, Irrera G, Pontari A, Callea I, Pucci G, Martinelli G, et al. Progenitor cell trafficking in patients with infarcted myocardium undergoing autologous bone marrow mononuclear cell injection. Interim analysis of a double blind randomised phase II clinical trial. European Group for Blood and Marrow Transplantation Annual Congress, 2007 March 25‐28, Lyon, France. Bone Marrow Transplantation 2007;39 (Suppl 1s):S215. CENTRAL

Grynberg 2008 {published data only}

Grynberg L, Balino NP, Riccitelli M, Dupont L, Caccione R, Traverso S, et al. Intracoronary bone marrow stem cell transplantation: Feasibility, safety, and effect on ventricular function and myocardial perfusion. Circulation 2008;118(12):E482‐3. CENTRAL

Jie 2014 {published data only}

Jie Q, Wang X, Bai M, Huang S, Qin J. A prospective study on autologous bone marrow mononuclear cell transplantation in ischemic heart failure. 25th Great Wall International Congress of Cardiology, Asia Pacific Heart Congress 2014, and the International Congress Cardiovascular Prevention and Rehabilitation 2014, 2014 October 16‐19, Beijing, China. Journal of the American College of Cardiology 2014;64 (Suppl 1):C182. CENTRAL

Kakuchaya 2011 {published data only}

Kakuchaya T. Influence of bone‐marrow derived progenitor stem cells on cardiac remodelling in a placebo‐controlled clinical trial involving patients with congestive heart failure. 20th World Congress of the World Society of Cardio‐Thoracic Surgeons, WSCTS, 2010 October 20‐23, Chennai, India. Heart Surgery Forum 2010;13:S121. CENTRAL
Kakuchaya T, Golukhova E, Eremeeva M, Chigogidze N, Aslanidi I, Nikitina T, et al. Bone‐marrow progenitor stem cells for the treatment of patients with congestive heart failure of different etiology in a placebo controlled clinical trial. 60th International Congress of the European Society for Cardiovascular Surgery, ESCVS 2011, 2011 May 20‐22, Moscow, Russia. Interactive Cardiovascular and Thoracic Surgery 2011;12:S68. CENTRAL
Kakuchaya T, Golukhova E, Eremeeva M, Chigogidze N, Aslanidi I, Shurupove I, et al. Accurate design of randomized placebo‐controlled clinical trials for assessment of stem cell effects on cardiac regeneration. European Heart Journal 2011;32:290: Abstract P1758. CENTRAL

Minjie 2011 {published data only}

Minjie L, Shihua Z, Sheng L, Shiliang J, Gang Y. Effects of autologous bone marrow mononuclear cells transplantation through coronary artery bypass grafting in patients with old myocardial infarction assessed by MRI: A randomised, double‐blind, placebo‐controlled pilot trial. Heart 2011;97:A137‐8. CENTRAL

Pourrajab 2013 {published data only}

Pourrajab F, Hekmatimoghaddam SH, Forouzannia SK, Baqhiyazdi M, Babaeezarch M, Ebrahimi H. Design of a combinatorial and feasible protocol for autologous bone marrow stem cell transplantation in patients candidate for CABG. 9th Congress on Stem Cell Biology and Technology of the Royan International Twin Congress, 2013 September 4‐6, Tehran, Iran. Cell Journal 2013;15:61, Abstract Ps96. CENTRAL

Stefanelli 2015 {published data only}

Stefanelli GG, Perro F, Trevisan D, Olaru A, Bia E, Meli M, et al. One step direct subendocardial implant of autologous stem cells during left ventricular restoration for ischemic heart failure. Heart Failure 2015 and the 2nd World Congress on Acute Heart Failure, 2015 May 23‐26, Seville, Spain. European Journal of Heart Failure 2015;17:410. CENTRAL

Zverev 2006 {published data only}

Zverev O, Boldueva S, Nemkov A, Shloydo E, Tsurupa S, Rigkova D, et al. Improvement of cardiomyocyte function after transplantation of autologous bone marrow mesenchymal stem cells in patients with non‐acute ischemic heart disease. World Congress of Cardiology, 2006 September 2‐6, Barcelona, Spain. European Heart Journal 2006;27 (Suppl 1):276: Abstract P1663. CENTRAL

EUCTR2009‐016364‐36‐NL {published data only}

EUCTR2009‐016364‐36‐NL. Injection of autologous bone marrow cells into damaged myocardium of no‐option patients with ischemic heart failure: a randomized placebo controlled trial. https://www.clinicaltrialsregister.eu/ctr‐search/search?query=2009‐016364‐36 First received 17 December 2009. CENTRAL
NTR2516. Injection of autologous bone marrow cells into damaged myocardium of no‐option patients with ischemic heart failure, a randomized placebo‐controlled trial. apps.who.int/trialsearch/trial.aspx?trialid=NTR2516 (accessed 7 September 2016). CENTRAL

ISRCTN71717097 {published data only}

ISRCTN71717097. Bone‐marrow derived stem cell transplantation in patients undergoing left ventricular restoration surgery for dilated ischaemic end‐stage heart failure: a randomised blinded controlled trial (TransACT 2). www.isrctn.com/ISRCTN71717097 First received 27 July 2009. CENTRAL

ISRCTN75217135 {published data only}

ISRCTN75217135. A pilot study to evaluate the efficacy of combined transplantation of progenitor cells and coronary artery bypass grafting (TOPCABG) N/A. www.isrctn.com/ISRCTN75217135 First received 30 September 2004. CENTRAL

NCT00690209 {published data only}

NCT00690209. By pass surgery with stem cell therapy in chronic ischemic cardiopathy. clinicaltrials.gov/show/NCT00690209 First received 30 May 2008. CENTRAL

NCT00790764 {published data only}

NCT00790764. Phase II combination stem cell therapy for the treatment of severe coronary ischemia (CI). clinicaltrials.gov/show/NCT00790764 First received 12 November 2008. CENTRAL

NCT00820586 {published data only}

NCT00820586. Intramyocardial delivery of autologous bone marrow. clinicaltrials.gov/show/NCT00820586 First received 8 January 2009. CENTRAL

NCT00950274 {published data only}

Donndorf P, Kaminski A, Tiedemann G, Kindt G, Steinhoff G. Validating intramyocardial bone marrow stem cell therapy in combination with coronary artery bypass grafting, the PERFECT Phase III randomized multicentre trial: study protocol for a randomized controlled trial. Trials 2012;13:99. CENTRAL
NCT00950274. Intramyocardial transplantation of bone marrow stem cells in addition to coronary artery bypass graft (CABG) surgery. clinicaltrials.gov/show/NCT00950274 First received 30 July 2009. CENTRAL

NCT01033617 {published data only}

NCT01033617. IMPACT‐CABG Trial: IMPlantation of Autologous CD133+ sTem Cells in Patients Undergoing CABG. clinicaltrials.gov/show/NCT01033617 First received 14 December 2009. CENTRAL

NCT01214499 {published data only}

NCT01214499. Prospective, controlled and randomized clinical trial on cardiac cell regeneration with laser and autologous bone marrow stem cells, in patients with coronary disease and refractory angina. clinicaltrials.gov/show/NCT01214499 First received 3 October 2010. CENTRAL

NCT01267331 {published data only}

NCT01267331. Cell therapy in patients with chronic ischemic heart disease undergoing cardiac surgery. clinicaltrials.gov/show/NCT01267331 First received 23 December 2010. CENTRAL

NCT01354678 {published data only}

NCT01354678. Intramyocardial multiple precision injection of bone marrow mononuclear cells in myocardial ischemia (IMPI). clinicaltrials.gov/show/NCT01354678 First received 13 May 2011. CENTRAL
Shlyakhto EV, Lebedev DS, Kryzhanovsky DV, Anisimov SV, Kozlenok AV, Berezina AV, et al. First experience of the study "Intramyocardial multiple precision administration of mononuclear bone marrow cells in the treatment of myocardial ischemia". Kardiologiia 2013;53(3):4‐8. CENTRAL

NCT01467232 {published data only}

NCT01467232. IMPACT‐CABG Trial: IMPlantation of Autologous CD133+ sTem Cells in Patients Undergoing Coronary Artery Bypass Grafting. clinicaltrials.gov/show/NCT01467232 First received 28 October 2011. CENTRAL

NCT01508910 {published data only}

NCT01508910. Efficacy and safety of targeted intramyocardial delivery of auto CD34+ stem cells for improving exercise capacity in subjects with refractory angina (RENEW). clinicaltrials.gov/show/NCT01508910 First received 10 January 2012. CENTRAL
Povsic TJ, Junge C, Nada A, Schatz RA, Harrington RA, Davidson CA, et al. A phase 3, randomized, double‐blinded, active‐controlled, unblinded standard of care study assessing the efficacy and safety of intramyocardial autologous CD34+ cell administration in patients with refractory angina: Design of the RENEW study. American Heart Journal 2013;165(6):854‐61. CENTRAL
Povsic TJ, Losordo DW, Story K, Junge CE, Schatz RA, Harrington RA, et al. A phase 3, randomized, partially blinded, active‐controlled, study assessing the efficacy and safety of intramyocardial autologous CD34+ cell administration in patients with refractory angina: Design of the RENEW study. American Heart Association 2012 Scientific Sessions and Resuscitation Science Symposium, 2012 November 3‐6, Los Angeles, CA. Circulation 2012;126 (21 Suppl 1):Abstract 11777. CENTRAL

NCT01615250 {published data only}

NCT01615250. Implantation of peripheral stem cells in patient with ischemic cardiomyopathy (ISCIC). clinicaltrials.gov/show/NCT01615250 First received 6 June 2012. CENTRAL

NCT01660581 {published data only}

NCT01660581. Intracardiac CD133+ cells in patients with no‐option resistant angina (Regent Vsel). clinicaltrials.gov/show/NCT01660581 First received 6 August 2012. CENTRAL

NCT01720888 {published data only}

NCT01720888. Intracoronary autologous mesenchymal stem cells implantation in patients with ischemic dilated cardiomyopathy. clinicaltrials.gov/show/NCT01720888 First received 1 November 2012. CENTRAL

NCT01727063 {published data only}

NCT01727063. Cell therapy in severe chronic ischemic heart disease (MiHeart). clinicaltrials.gov/show/NCT01727063 First received 12 November 2012. CENTRAL

NCT01758406 {published data only}

NCT01758406. Transplantation of autologous cardiac stem cells in ischemic heart failure. clinicaltrials.gov/show/NCT01758406 First received 24 December 2012. CENTRAL

NCT01768702 {published data only}

Bartunek J, Davison B, Sherman W, Povsic T, Henry TD, Gersh B, et al. Congestive Heart Failure Cardiopoietic Regenerative Therapy (CHART‐1) trial design. European Journal of Heart Failure 2016;18(2):160‐8. CENTRAL
EUCTR2011_001117‐13‐GB. Research study aiming at investigating the potential effectiveness and safety of a treatment for chronic advanced heart failure of ischemic origin. The treatment is based on patient own stem cells that will be collected and guided to the cardiac cells lineage before being injected into the heart muscle. https://www.clinicaltrialsregister.eu/ctr‐search/search?query=2011‐001117‐13 First received 11 July 2012. CENTRAL
NCT01768702. Safety and efficacy of autologous cardiopoietic cells for treatment of ischemic heart failure. (CHART‐1). clinicaltrials.gov/show/NCT01768702 First received 21 December 2012. CENTRAL

NCT02022514 {published data only}

EUCTR2013‐000915‐26‐ES. Clinical trial phase III single‐center, open‐label efficacy of intracoronary infusion of bone marrow mononuclear cells in patients with occlusion autologous chronic coronary revascularization and ventricular dysfunction previously [Ensayo Clínico Fase III Unicéntrico, Abierto, Aleatorizado sobre eficacia de la Infusión Intracoronaria de células Mononucleadas de Médula Ósea Autóloga en Pacientes Con Oclusión Coronaria Crónica y Disfunción Ventricular previamente revascularizados]. https://www.clinicaltrialsregister.eu/ctr‐search/search?query=2013‐000915‐26 First received 9 April 2013. CENTRAL
NCT02022514. Intracoronary infusion of mononuclear cells autologous bone marrow in patients with chronic coronary occlusion and ventricular dysfunction, previously revascularized. clinicaltrials.gov/show/NCT02022514 First received 20 December 2013. CENTRAL

NCT02059512 {published data only}

NCT02059512. Autologous bone marrow mononuclear cells in the combined treatment of coronary heart disease. clinicaltrials.gov/show/NCT02059512 First received 1 February 2014. CENTRAL

NCT02317458 {published data only}

NCT02317458. Congestive Heart Failure Cardiopoietic Regenerative Therapy (CHART‐2) Trial. clinicaltrials.gov/show/NCT02317458 First received 9 December 2014. CENTRAL

NCT02362646 {published data only}

NCT02362646. Safety & efficacy of intramyocardial injection of mesenchymal precursor cells on myocardial function in LVAD recipients. clinicaltrials.gov/show/NCT02362646 First received 9 February 2015. CENTRAL

NCT02438306 {published data only}

NCT02438306. CardiAMP™ Heart Failure Trial. clinicaltrials.gov/ct2/show/NCT02438306 First received 5 May 2015. CENTRAL

NCT02462330 {published data only}

NCT02462330. Administration of mesenchymal stem cells in patients with chronic ischemic cardiomyopathy (MESAMI2). clinicaltrials.gov/ct2/show/NCT02462330 First received 27 May 2015. CENTRAL

NCT02501811 {published data only}

NCT02501811. Combination of Mesenchymal and C‐kit+ Cardiac Stem Cells as Regenerative Therapy for Heart Failure (CONCERT‐HF). clinicaltrials.gov/ct2/show/NCT02501811 First received 15 July 2015. CENTRAL

NCT02503280 {published data only}

NCT02503280. The Transendocardial Autologous Cells (hMSC or hMSC and hCSC) in Ischemic Heart Failure Trial (TAC‐HFT II). clinicaltrials.gov/ct2/show/NCT02503280 First received 4 May 2015. CENTRAL

NCT02504437 {published data only}

NCT02504437. Therapy of Preconditioned Autologous BMMSCs for Patients With Ischemic Heart Disease (TPAABPIHD). clinicaltrials.gov/ct2/show/NCT02504437 First received 14 July 2015. CENTRAL

Abdel‐Latif 2007

Abdel‐Latif A, Bolli R, Tleyjeh IM, Montori VM, Perin EC, Hornung CA, et al. Adult bone marrow‐derived cells for cardiac repair: a systematic review and meta‐analysis. Archives of Internal Medicine 2007;167(10):989‐97.

Afzal 2015

Afzal MR, Samanta A, Shah ZI, Jeevanantham V, Abdel‐Latif A, Zuba‐Surma EK, et al. Adult bone marrow cell therapy for ischemic heart disease: evidence and insights from randomized controlled trials. Circulation Research 2015;117(6):558‐75.

Ambrosy 2014

Ambrosy AP, Fonarow GC, Butler J, Chioncel O, Greene SJ, Vaduganathan M, et al. The global health and economic burden of hospitalizations for heart failure: lessons learned from hospitalized heart failure registries. Journal of the American College of Cardiology 2014;63(12):1123‐33.

Arnesen 2007

Arnesen H, Lunde K, Aakhus S, Forfang K. Cell therapy in myocardial infarction. Lancet 2007;369(9580):2142‐3.

Arnold 2014

Arnold SV, Kosiborod M, Li Y, Jones PG, Yue P, Belardinelli L, et al. Comparison of the Seattle Angina Questionnaire with daily angina diary in the TERISA clinical trial. Circulation. Cardiovascular Quality Outcomes 2014;7(6):844‐50.

Behfar 2014

Behfar A, Crespo‐Diaz R, Terzic A, Gersh BJ. Cell therapy for cardiac repair ‐ lessons from clinical trials. Nature Reviews. Cardiology 2014;11(4):232‐46.

Beltrami 2003

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.

Benedetto 2016

Benedetto U, Gaudino M, Ng C, Biondi‐Zoccai G, D'Ascenzo F, Frati G, et al. Coronary surgery is superior to drug eluting stents in multivessel disease. Systematic review and meta‐analysis of contemporary randomized controlled trials. International Journal of Cardiology 2016;210:19‐24.

BHF 2014

British Heart Foundation. Cardiovascular Disease Statistics 2014. www.bhf.org.uk/publications/statistics/cardiovascular‐disease‐statistics‐2014 (accessed 5 April 2016).

Borm 2009

Borm GF, Donders AR. Updating meta‐analyses leads to larger type I errors than publication bias. Journal of Clinical Epidemiology 2009;62(8):825‐30.

Carr 2008

Carr CA, Stuckey DJ, Tatton L, Tyler DJ, Hale SJ, Sweeney D, et al. Bone marrow‐derived stromal cells home to and remain in the infarcted rat heart but fail to improve function: an in vivo cine‐MRI study. American Journal of Physiology. Heart and Circulatory Physiology 2008;295(2):H533‐42.

Clifford 2012a

Clifford DM, Fisher SA, Brunskill SJ, Doree C, Mathur A, Watt S, et al. Stem cell treatment for acute myocardial infarction. Cochrane Database of Systematic Reviews 2012, Issue 2. [DOI: 10.1002/14651858.CD006536.pub3]

de Jong 2014

de Jong R, Houtgraaf JH, Samiei S, Boersma E, Duckers HJ. Intracoronary stem cell infusion after acute myocardial infarction: a meta‐analysis and update on clinical trials. Circulation. Cardiovascular Interventions 2014;7:156‐67.

Delewi 2014

Delewi R, Hirsch A, Tijssen JG, Schächinger V, Wojakowski W, Roncalli J, et al. Impact of intracoronary bone marrow cell therapy on left ventricular function in the setting of ST‐segment elevation myocardial infarction: a collaborative meta‐analysis. European Heart Journal 2014;35(15):989‐98.

Egger 1987

Egger M, Smith GD, Schneider M, Minder C. Bias in meta‐analysis detected by a simple, graphical test. BMJ 1997;315:629‐34.

Fisher 2013

Fisher SA, Doree C, Brunskill JS, Mathur A, Martin‐Rendon E. Bone marrow stem cell treatment for ischemic heart disease patients with no option of revascularization: a systematic review and meta‐analysis. PLoS ONE 2013;8(6):e64669.

Fisher 2015a

Fisher SA, Zhang H, Doree C, Mathur A, Martin‐Rendon E. Stem cell treatment for acute myocardial infarction. Cochrane Database of Systematic Reviews 2015, Issue 9. [DOI: 10.1002/14651858.CD006536.pub4]

Fisher 2015b

Fisher SA, Doree C, Mathur A, Martin‐Rendon E. Meta‐analysis of cell therapy trials for patients with heart failure. Circulation Research 2015;116(8):1361‐77.

Fisher 2016

Fisher SA, Doree C, Taggart DP, Mathur A, Martin‐Rendon E. Cell therapy for heart disease: Trial sequential analyses of two Cochrane reviews. Clinical Pharmacology and Therapeutics 2016 Jan 28 [Epub ahead of print]. [DOI: 10.1002/cpt.344]

Go 2014

Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ, et al. Heart disease and stroke statistics ‐ 2014 update: a report from the American Heart Association. Circulation 2014;129(3):e28‐e292.

GRADEpro GDT [Computer program]

McMaster University. GRADEpro GDT: GRADEpro Guideline Development Tool. Hamilton, ON: McMaster University, 2015 (developed by Evidence Prime, Inc.) Available from gradepro.org.

Gyöngyösi 2015

Gyöngyösi M, Wojakowski W, Lemarchand P, Lunde K, Tendera M, Bartunek J, et al. Meta‐Analysis of Cell‐based CaRdiac stUdiEs (ACCRUE) in patients with acute myocardial infarction based on individual patient data. Circulation Research 2015;116(8):1346‐60.

Hamshere 2015

Hamshere S, Choudhury T, Mozid A, Agarwal S, Jones DA, Martin J, et al. Safety and efficacy of G‐CSF and autologous bone marrow‐derived cells in ischaemic cardiomyopathy: Results of the REGENERATE‐IHD Phase II trial. European Heart Journal 2015;36 (Suppl 1):P3774.

Hartwell 2005

Hartwell D, Colquitt J, Loveman E, Clegg AJ, Brodin H, Waugh N, et al. Clinical effectiveness and cost‐effectiveness of immediate angioplasty for acute myocardial infarction: systematic review and economic evaluation. Health Technology Assessment 2005;9(17):1‐99.

Harvey 2015

Harvey E, Fisher SA, Doree C, Taggart DP, Martin‐Rendon E. Current evidence of the efficacy of cell‐based therapies in heart failure. Circulation Journal 2015;79(2):229‐36.

Heldman 2014

Heldman AW, DiFede DL, Fishman JE, Zambrano JP, Trachtenberg BH, Karantalis V, et al. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: the TAC‐HFT randomized trial. JAMA 2014;311(1):62‐73.

Higgins 2002

Higgins JPT, Thompson SG. Quantifying heterogeneity in a meta‐analysis. Statistics in Medicine 2002;21(11):1539‐58.

Higgins 2003

Higgins JPT, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta‐analyses. BMJ 2003;327(7414):557‐60.

Higgins 2011

Higgins JPT, Green S (editors). Cochrane Handbook for Systematic Reviews of Interventions Version 5.1.0 [updated March 2011]. The Cochrane Collaboration, 2011. Available from handbook.cochrane.org.

Hu 2007

Hu M, Cappeleri J, Lan KK. Applying the law of the iterated logarithm to control type I error in cumulative meta‐analysis of binary outcomes. Clinical Trials 2007;4(4):329‐40.

Huedo‐Medina 2006

Huedo‐Medina TB, Sánchez‐Meca J, Marín‐Martínez F, Botella J. Assessing heterogeneity in meta‐analysis: Q statistic or I2 index?. Psychological Methods 2006;11(2):193‐206.

Ibrahim 2016

Ibrahim A, Marbán E. Exosomes: Fundamental biology and roles in cardiovascular physiology. Annual Review of Physiology 2016;10(78):67‐83.

Ioannidis 2007

Ioannidis JP, Patsopoulos NA, Evangelou E. Assessing heterogeneity in meta‐analysis: Q statistic or I2 index?. BMJ 2007;335(7626):914‐6.

Jeevanantham 2012

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.

Khan 2016

Khan AR, Farid TA, Pathan A, Tripathi A, Ghafghazi S, Wysoczynski M, et al. Impact of cell therapy on myocardial perfusion and cardiovascular outcomes in patients with angina refractory to medical therapy: A systematic review and meta‐analysis. Circulation Research 2016;118(6):984‐93.

Lan 2003

Lan KK, Hu M, Cappelieri J. Applying the law of the iterated logarithm to cumulative meta‐analysis of a continuous endpoint. Statistica Sinica 2003;13:1135‐45.

Lehtinen 2014

Lehtinen M, Patila T, Vento A, Kankuri E, Suojaranta‐Ylinen R, Poyhia R. Prospective, randomized, double‐blinded trial of bone marrow cell transplantation combined with coronary surgery ‐ perioperative safety study. Interactive Cardiovascular and Thoracic Surgery 2014;19(6):990‐6.

Li 2012

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. Journal of the American College of Cardiology 2012;59(10):942‐53.

Lloyd‐Jones 2002

Lloyd‐Jones DM, Larson MG, Leip EP, Beiser A, D'Agostino RB, Kannel WB, et al. Lifetime risk for developing congestive heart failure: the Framingham Heart Study. Circulation 2002;106(24):3068‐72.

Martin‐Rendon 2008a

Martin‐Rendon E, Sweeney D, Lu FJ, Girdlestone J, Navarrete C, Watt SM. 5‐Azacytidine‐treated human mesenchymal stem/progenitor cells derived from umbilical cord, cord blood and bone marrow do not generate cardiomyocytes in vitro at high frequencies. Vox Sanguinis 2008;95(2):137‐48.

Mathur 2004

Mathur A, Martin JF. Stem cells and repair of the heart. Lancet 2004;364(9429):183‐92.

NCT00747708

NCT00747708. Bone marrow derived adult stem cells for chronic heart failure (REGEN‐IHD). clinicaltrials.gov/show/NCT00747708 (accessed 14 October 2015).

R Core Team 2013 [Computer program]

R Core Team. R: A language and environment for statistical computing (www.R‐project.org/). Vienna, Austria: R Foundation for Statistical Computing, 2013.

Review Manager 2014 [Computer program]

The Nordic Cochrane Centre, The Cochrane Collaboration. Review Manager (RevMan). Version 5.3. Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration, 2014.

Schünemann 2013

Schünemann H, Brożek J, Guyatt G, Oxman A (editors). GRADE Handbook for Grading Quality of Evidence and Strength of Recommendations. The GRADE Working Group, updated October 2013. Available from guidelinedevelopment.org/handbook.

Skinner 2011

Skinner JS, Cooper A. Secondary prevention of ischaemic cardiac events. BMJ Clinical Evidence 2011;Aug 30:0206.

Stuckey 2006

Stuckey DJ, Carr CA, Martin‐Rendon E, Tyler DJ, Willmott C, Cassidy PJ, et al. Iron particles for noninvasive monitoring of bone marrow stromal cell engraftment into, and isolation of viable engrafted donor cells from, the heart. Stem Cells 2006;24(8):1968‐75.

Taggart 2012

Taggart DP. Incomplete revascularization: appropriate and inappropriate. European Journal of Cardiothoracic Surgery 2012;41(3):542‐3.

TSA 2011 [Computer program]

Copenhagen Trial Unit. Trial Sequential Analysis (TSA) program. Denmark: Copenhagen Trial Unit, 2011. (www.ctu.dk/tsa).

Wen 2011

Wen Y, Meng L, Xie J, Ouyang J. Direct autologous bone marrow‐derived stem cell transplantation for ischemic heart disease: A meta‐analysis. Expert Opinion on Biological Therapy 2011;11(5):559‐67.

Wettersley 2008

Wettersley J, Thorlund K, Brok J, Gluud C. Trial sequential analysis may establish when firm evidence is reached in cumulative meta‐analysis. Journal of Clinical Epidemiology 2008;61:64‐75.

Wu 2009

Wu T, Li Y, Bian Z, Liu G, Moher D. Randomized trials published in some Chinese journals: how many are randomized?. Trials 2009;20:46.

Xu 2014

Xu R, Ding S, Zhao Y, Pu J, He B. Autologous transplantation of bone marrow/blood‐derived cells for chronic ischemic heart disease: A systematic review and meta‐analysis. Canadian Journal of Cardiology 2014;30(11):1370‐7.

Yoon 2005

Yoon YS, Wecker A, Heyd L, Parks JS, Tkebuchava T, Kusano K, et al. Clonally expanded novel multipotent stem cells from human bone marrow regenerate myocardium after myocardial infarction. Journal of Clinical Investigation 2005;115(2):326‐38.

Yusuf 2002

Yusuf S. Two decades of progress in preventing vascular disease. Lancet 2002;360(9326):2‐3.

Referencias de otras versiones publicadas de esta revisión

Fisher 2014

Fisher SA, Brunskill SJ, Doree C, Mathur A, Taggart DP, Martin‐Rendon E. Stem cell therapy for chronic ischaemic heart disease and congestive heart failure. Cochrane Database of Systematic Reviews 2014, Issue 4. [DOI: 10.1002/14651858.CD007888.pub2]

Martin‐Rendon 2009

Martin‐Rendon E, Brunskill S, Hyde C, Doree C, Mathur A, Taggart DP. Stem cell therapy for ischaemic heart disease and congestive heart failure. Cochrane Database of Systematic Reviews 2009, Issue 3. [DOI: 10.1002/14651858.CD007888]

Characteristics of studies

Characteristics of included studies [ordered by study ID]

Ang 2008

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: British Heart Foundation (grant PG04050)

Study setting: Leicester, UK
Number of centres: 1
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: Intramyocardial BMSC (IM): 21; intracoronary BMSC (IC): 21; Controls: 21
Number (N) of participants analysed (primary outcome) in each arm: 17 IM, 16 IC, 15 C

Participants

Description: Chronic IHD (aged 18 to 80 years; presence of at least 1 chronic, irreversible myocardial scar; elective cardiac surgery).
Age distribution (SD) in each arm: IM: 64.7 (8.7) years; IC: 62.1 (8.7) years; Controls: 61.3 (8.3) years.
Sex (% male) in each arm: IM: 71.4%; IC: 90.5%; Controls: 90%.

Number of diseased vessels: n/r (multivessel).
Time from symptom onset to initial treatment: At least 6 weeks.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arms: BMSC‐IM or BMSC‐IC

Type of stem cells: Mononuclear cells administered intramyocardially (IM) or intracoronarily (IC)
Summary of stem cell isolation and type and route of delivery: Bone marrow aspiration followed by density gradient centrifugation to enrich in mononuclear cells, infused via the coronary artery (IC) or injected into the myocardium (IM)
Dose of stem cells: 8.6 (5.6) x 107 cells (IM); 11.5 (73) x 107 cells (IC)
Timing of stem cell procedure: Concomitant to CABG

G‐CSF details: No G‐CSF administered.

Comparator arm: Control (no BM aspiration; no placebo or sham procedure)

Co‐intervention: CABG

Outcomes

Primary outcome:

Improvement in contractile function of treated scar areas at 6 months.
Secondary outcomes:

Global left ventricular functions at 6 months (infarct size, global end‐diastolic volume and end‐systolic volume, and improvement in stroke volume and LVEF).

Additional outcomes: Postoperative complications, troponin I levels within 24 hours of surgery, and clinical evaluation (assessment of functional status and adverse events).
Outcome assessment points: Baseline and 6 months
Method(s) of outcome measurement: MRI

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The trial was described as randomised, but the method of randomisation was not reported.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo was administered; participants and clinicians were not blinded.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Physicians treating the participants during the postoperative period and the investigators performing the examinations and interpreting the results were blind to which group participants had been assigned.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

2 controls (2/21) were excluded from analysis of mortality and morbidity (1x withdrawal before surgery, 1x deemed unsuitable for follow‐up). 12 participants were lost to follow‐up (4 IM, 4 IC, 3 controls), and 2 died within 30 days. MRI was performed in 33 participants, of which 4 were “not suitable for accurate analysis”. However, MRI results were only reported for 25 participants; this discrepancy was unexplained.

Selective reporting (reporting bias)

Low risk

All outcomes reported in the trial protocol (NCT00560742) were reported.

Other bias

Low risk

No other sources of bias were reported or identified.

Assmus 2006

Methods

Type of study: Cross‐over RCT
Type of publication: Full paper
Source of funding: Supported by the Deutsche ForschungsGemeinschaft (FOR 501‐1: WA 146/2‐1), Fondation Leducq Transatlantic Network of Excellence for Cardiac Regeneration, European Union European Vascular Genomics Network (LSHM‐CT‐2003‐503254), and Alfried Krupp Stiftung.

Study setting: Frankfurt, Germany
Number of centres: 1
Length of follow‐up: 3 months
Number (N) of participants randomised to each arm: BMSC: 28; CPC: 24; Controls: 23
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 24; CPC: 19, Controls: 18

Participants

Description: Chronic IHD (aged 18 to 80 years; MI at least 3 months previously; well‐demarcated region of left ventricular dysfunction and a patent infarct‐related artery).
Age distribution in each arm: BMSC: 59 ± 12 years; CPC: 54 ± 12 years; Controls: 61 ± 9 years.
Sex (% male) in each arm: BMSC: 89%; CPC: 79%; Controls: 100%.

Number of diseased vessels: BMSC: 1 (n = 7), 2 (n = 13), 3 (n = 8); CPC: 1 (n = 7), 2 (n = 4), 3 (n = 12); Controls: 1 (n = 2), 2 (n = 9), 3 (n = 12).
Time from symptom onset to initial treatment: Previous MI at least 3 months earlier. 100% participants with previous MI.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC or CPC
Type of stem cells: Mononuclear cells or circulating progenitor cells
Summary of stem cell isolation and type and route of delivery: BMSC: 50 mL of bone marrow aspirate was obtained under local anaesthesia on the morning of cell transplantation. Mononuclear cells were isolated by Ficoll‐gradient centrifugation. CPC: Mononuclear cell fraction was isolated by Ficoll‐gradient centrifugation of 270 mL of venous blood and cultured for 3 days ex vivo. Cells were delivered intracoronarily in both arms of the trial.
Dose of stem cells: BMSC arm: 2.05 ± 1.1 x 108 mononuclear cells. On average less than 1% were CD34‐positive cells. CPC arm: 2.2 ± 1.1 x 107 mononuclear cells. No measure of CD34‐positive cells in this fraction.
Timing of stem cell procedure: At least 3 months after last MI. In some cases concomitant PCI.

G‐CSF details: No G‐CSF administered.

Comparator arm: Control (no BM aspiration; no placebo or sham procedure)

Co‐intervention: Standard medical therapy; PCI (in 27% of participants)

Outcomes

Primary outcome:

Change in global left ventricular function (measured by quantitative left ventricular angiography).
Secondary outcomes:

  1. Quantitative parameters of regional left ventricular function of the target area

  2. Changes in left ventricular volumes

  3. Functional status as assessed by NYHA classification

  4. Event‐free survival (defined as freedom from death, MI, stroke, or rehospitalisation for worsening HF) after 4 months' follow‐up

Outcome assessment points: Baseline and 3 months.
Method(s) of outcome measurement: LV angiography and MRI

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was performed using computerised simple random allocation with known N. No blockwise randomisation was performed.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo was administered; participants and clinicians were not blinded.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Quantitative analysis of angiograms and MRI images was performed by an investigator who was blinded to the individual participant's treatment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants were included in the analysis of mortality and morbidity. 14 participants (4x cell therapy, 5x CPC, 5x controls) were excluded from MRI/angiography and functional status at follow‐up, with reasons given for all exclusions.

Selective reporting (reporting bias)

Low risk

All outcomes reported in the trial protocol (NCT00289822) were reported.

Other bias

Low risk

No other sources of bias were reported or identified.

Assmus 2013

Methods

Type of study: Parallel RCT
Type of publication: Full paper; abstract (long‐term follow‐up)
Source of funding: Supported by an unrestricted grant to the Goethe University Frankfurt from t2cure GmbH.

Study setting: Langen, Germany
Number of centres: 1
Length of follow‐up: 45.7 (17) months
Number (N) of participants randomised to each arm: BMSC: 43 (22 LD (low‐dose shockwave), 21 HD (high‐dose shockwave)); Controls: 39 (20 LD, 19 HD)
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 41 (21 LD, 20 HD); Controls: 38 (19 LD, 19 HD)

Participants

Description: Chronic ischaemic HF (aged 18 to 80 years; anterior MI occurring 3 months or more prior to inclusion and stable chronic postinfarction HF defined by LVEF less than 50% or symptoms of NYHA class II or greater; a patent vessel supplying the target region).

Age distribution in each arm: BMSC: 65 (12) (LD), 58 (11) (HD); Controls: 60 (10) (LD), 63 (10) (HD).
Sex (% male) in each arm: BMSC: 77% (LD), 86% (HD); Controls: 80% (LD), 90% (HD).

Number of diseased vessels: Not reported.
Time from symptom onset to initial treatment: Not reported.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: 50 mL of bone marrow was aspirated into heparin‐containing syringes from the iliac crest under local anaesthesia. Mononuclear cells were isolated and enriched with the use of Ficoll‐Hypaque centrifugation procedures. The cell suspension consisted of a heterogeneous cell population including hematopoietic, mesenchymal, and other progenitor cells. The cells were suspended in 10 mL of X‐VIVO 10 medium, including 2 mL of the participant's own serum.

Dose of stem cells: 123 (69) x 106 (HD), 150 (77) x 106 (LD).
Timing of stem cell procedure: 24 hours following shockwave.

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; 10 mL of X‐VIVO 10 medium, including 2 mL of the participant's own serum).

Co‐intervention: Shockwave (HD or LD)

Outcomes

Primary outcome:

Improvement in global LVEF on quantitative LV angiography at 4 months' follow‐up.
Secondary outcomes:

  1. Global LV volumes, regional LV function, and late enhancement volume measured by MRI at 4 months and 1 year

  2. NYHA class at 4 months

  3. NT BNP levels at 4 months

  4. Major adverse clinical events (death, model of death, rehospitalisation for worsening HF, recurrent MI, ventricular tachycardia, revascularisation, and stroke) at 4 months

  5. Quality of life at 4 months

Outcome assessment points: Baseline, 4 months, and mean 45.7 (17) months (clinical outcomes only).

Method(s) of outcome measurement: LV angiography; MRI

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was performed by a simple random allocation using a computer list with known N. No blockwise randomisation was performed.

Allocation concealment (selection bias)

Low risk

Randomisation codes were generated at the cell‐processing centre for the entire study cohort.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The trial was reported as double‐blind. All participants underwent BM aspiration, and the control group received a placebo. Participants were blinded; blinding of clinicians was not specifically reported.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Investigators were blinded for the intracoronary infusion of the study medication.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of mortality and morbidity. All participants were included in angiography analyses on an intention‐to‐treat basis. MRI was performed in a subset of participants (15 cell therapy and 12 controls).

Selective reporting (reporting bias)

Low risk

All outcomes reported in the trial protocol (NCT00326989) were reported.

Other bias

High risk

Supported by an unrestricted grant from t2cure GmbH. No other sources of bias were reported or identified.

Bartunek 2012

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: Cardio3 BioSciences

Study setting: Belgium, Serbia, Switzerland
Number of centres: 9 (Belgium, Serbia, Switzerland)
Length of follow‐up: 2 years
Number (N) of participants randomised to each arm: BMSC: 32; Controls: 15
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 21; Controls: 15

Participants

Description: IHD (aged 18 to 75 years; stable HF population with a history of MI; baseline LVEF 15% to 40%; ischaemic event at least 2 months prior to recruitment; optimally managed and revascularised).

Age distribution in each arm: BMSC: 55.3 (SE 10.4) years; Controls: 58.7 (SE 8.2) years
Sex (% male) in each arm: BMSC: 90.5%; Controls: 86.7%

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: time from infarction to cell delivery, mean 1540 (range 192 to 7515) days.
Statistically significant baseline imbalances between the groups? None

Interventions

Intervention arm: BMSC
Type of stem cells: Cardiopoietic cells (mesenchymal stem cells)
Summary of stem cell isolation and type and route of delivery: Human BM was harvested from the iliac crest, cultured at 37°C/5% CO2 in 175 cm2 flasks to purify MSCs. After 24 h, nonadherent BM and cellular debris were discarded, and adherent MSCs were washed with PBS solution. A 1‐to‐1 passage was performed to dissociate colony‐forming units and allow for expansion for up to 6 days in a culture medium supplemented with 5% human pooled platelet lysate media to generate a monolayer whereby 50 x 106 cells were obtained. Lineage specification was achieved by MSC exposure to a cardiogenic cocktail regimen triggering expression and nuclear translocation of cardiac transcription factors while maintaining clonal proliferation. Passage P1 marked the start of cardiogenic cocktail treatment in which cells were cultured for 5 days in 5% platelet lysate‐supplemented high glucose medium containing cardiogenic growth factors (TGF‐b, BMP‐4, Activin A, FGF‐2, cardiotrophin, a‐thrombin, diaminopyrimidine). Cell density was 4000 cells/cm2 during MSC culture and 1500 cells/cm2 during cardiopoietic induction. Passage P2/P3 marked the end of the cardiogenic cocktail treatment followed by expansion to yield 600 to 1200 x 106 cells. Harvest involved trypsinisation and concentration in a preservation solution. Cells were centrally manufactured in a GMP facility. Cells packaged for transportation were transplanted within 72 h of derivation.

Dose of stem cells: mean 733 x 106 (range 605 x 1168 x 106) cells
Timing of stem cell procedure: BM harvest ‐ 24 hrs ‐ (P0) (up to 6 days) ‐ P1 (5 days cell culture) ‐ 4 to 6 weeks.

G‐CSF details: No G‐CSF administered.

Comparator arm: Control (no BM aspiration; no placebo or sham procedure)

Co‐intervention: Standard medical therapy

Outcomes

Primary outcome:

Changes in LVEF at 6 months

Note: Main study publication reports primary endpoint as feasibility and safety at 2‐year follow‐up (Bartunek 2012).
Secondary outcomes:

  1. 6‐min walking distance (6 months, 1 and 2 years)

  2. Quality of life (6 months, 1 and 2 years)

  3. All‐cause mortality (at each follow‐up)

  4. Cardiovascular events (at each follow‐up)

Note: Main study publication reports secondary endpoints as including cardiac structure and function (Bartunek 2012).

Outcome assessment points: Baseline, 6 months, and 2 years (clinical outcomes only).
Method(s) of outcome measurement: Echocardiography

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was conducted through a site‐independent, centralised process after exclusion of participants that did not meet the inclusion criteria.

Allocation concealment (selection bias)

Low risk

Allocation concealment was not fully described, but randomisation was conducted in a site‐independent manner through a centralised process.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo was administered; neither participants nor clinicians were blinded.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

An independent core laboratory masked to study arm assignment and chronology of clinical evaluation provided data analysis.

Incomplete outcome data (attrition bias)
All outcomes

High risk

11 participants in the cell therapy group were excluded from analysis (2x clinical inclusion criteira not met; 2x BM inclusion criteria not met; 5x cell release inclusion criteria not met; 2x cell growth inclusion criteria not met). In primary analyses described in the paper, these participants were analysed as part of the control group (although they are excluded from analysis in the results of this review). All other randomised participants were included in all analyses.

Selective reporting (reporting bias)

Unclear risk

The study protocol (NCT00810238) defined the primary outcome as change in LVEF at 6 months, whereas the study publication defined the primary outcome as feasibility and safety at 2 years. Follow‐up of exercise capacity and quality of life at 1 and 2 years was also defined as an outcome according to the trial protocol but was not reported. All other outcomes described in the trial protocol were reported in results.

Other bias

High risk

This study received commercial funding from Cardio3 BioSciences, although the authors reported that they had no relationships relevant to the contents of the paper to disclose. No other sources of bias were reported or identified.

Chen 2006

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: Not reported

Study setting: China
Number of centres: 1
Length of follow‐up: 12 months
Number (N) of participants randomised to each arm: BMSC: 24; Controls: 24
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 22; Controls: 23

Participants

Description: Severe ischaemic HF (isolated, chronic, total, or subtotal occluded LAD due to previous anterior wall infarction untreated by either thrombolysis or primary PCI; reversible perfusion defect detectable by SPECT; LVEF < 40%).

Age distribution in each arm: BMSC: 59.3 ± 6.8 years; Controls: 57.8 ± 7.2 years.
Sex (% male) in each arm: BMSC: 88%; Controls: 92%.

Number of diseased vessels: Not reported.
Time from symptom onset to initial treatment: 14 days following successful PCI.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC
Type of stem cells: Mesenchymal stem cells

Summary of stem cell isolation and type and route of delivery: 60 mL of autologous bone marrow were aspirated under local anaesthesia from the ilium of all participants during the morning of the 8th day after the PCI procedure and then cultured for 7 days. BM mesenchymal stem cells were harvested and washed 3 to 4 times with heparinised saline. 2 hours before transplantation, the stem cell suspension was mixed with heparin, filtered, and prepared for implantation. Cell viability was > 92%.
Dose of stem cells: 5 x 106 cells
Timing of stem cell procedure: 14 days following successful PCI and 7 days after bone marrow aspiration.

G‐CSF details: No G‐CSF administered.

Comparator arm: Control (no placebo).

Co‐intervention: PCI

Outcomes

Primary outcome:

None reported.
Secondary outcomes:

Reversible defects, metabolic equivalents, exercise, LVEF, NYHA, mortality.
Outcome assessment points: Baseline and 12 months.
Method(s) of outcome measurement: SPECT

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

This Chinese trial was described as randomised, but the method of randomisation was not reported.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo was used; neither participants nor clinicians were blinded.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

Blinding of outcome assessors was not reported.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

2 cell therapy participants and 1 control were excluded from all analyses due to failed PCI. All remaining participants were included in the analysis of mortality and morbidity; all surviving participants were included in the analysis of LVEF, NYHA class, and exercise tolerance at follow‐up.

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

Erbs 2005

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: Supported by Heart Center Leipzig GmbH, University of Leipzig.

Study setting: Leipzig, Germany
Number of centres: 1
Length of follow‐up: 15 months
Number (N) of participants randomised to each arm: BMSC: 14; Controls: 14
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 12; Controls: 11

Participants

Description: Chronic total artery occlusion with clinical signs of myocardial ischaemia and local wall motion abnormalities.
Age distribution in each arm: BMSC: 63 ± 7 years; Controls: 61 ± 9 years.
Sex (% male) in each arm: BMSC: 71%; Controls: 86%.

Number of diseased vessels: BMSC: 1 (n = 8), 2 (n = 4), 3 (n = 2); Control arm: 1 (n = 6), 2 (n = 5), 3 (n = 3).
Time from symptom onset to initial treatment: Complete total obstruction defined as an obstruction of a native coronary artery for more than 30 days with no luminal continuity and with TIMI flow grade 0 or 1.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: G‐CSF + BMSC
Type of stem cells: Circulating progenitor cells
Summary of stem cell isolation and type and route of delivery: All participants subcutaneously injected twice a day with filgrastim (G‐CSF, 300 mcg) over 4 days to increase the amount of CPC in the blood. At day 4, 400 mL of venous blood were collected from all participants, MNC were purified and ex vivo‐cultured for 4 days in endothelial‐specific medium to select CPC. MNC were isolated from 400 mL of venous blood by density gradient centrifugation (Histopaque‐1077). Immediately after isolation, total MNC were plated on gelatin‐coated cell culture flasks with a cell density of 1 x 106 cells/cm2. Cells were maintained for 4 days in endothelial basal medium supplemented with EGM SingleQuots and 10% human serum, collected from each individual participant. Additionally, the cell culture medium was supplemented with ascorbic acid (final concentration 75 ng/mL) and hydrocortisone (0.2 mcg/mL). After 4 days of culture, non‐adherent cells were removed by a thorough washing with PBS, and the adherent cells were detached with trypsin/EDTA. The collected cells were washed twice with PBS containing 2 mmol/L EDTA and resuspended in a final volume of 20 mL physiological sodium chloride supplemented with 10% autologous participant serum. Cells were administered intracoronarily.
Dose of stem cells: 69 ± 14 x 106 CPC (range 22 x 106 to 200 x 106).
Timing of stem cell procedure: 10 ± 1 days following successful recanalisation.

G‐CSF details: 300 mg of G‐CSF administered for 4 days to all participants.

Comparator arm: G‐CSF + placebo (BM aspiration; cell‐free serum solution).

Co‐intervention: PCI

Outcomes

Primary outcomes:

LV function
Secondary outcomes:

Assessment of coronary endothelial function, myocardial viability (number of myocardial segments with hibernation), regional wall motion, LV mass (myocardial mass; infarct size). Clinical outcomes, restenosis, coronary endothelium function, myocardial viability, number of hibernating segments in myocardium.
Outcome assessment points: Baseline, 3 and 15 months.
Method(s) of outcome measurement: MRI

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The trial was described as randomised, but the method of randomisation was not reported.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

G‐CSF was administered and blood was taken from all participants. Control participants received a placebo. Participants and clinicians were blinded to treatment.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Image analysis assessors remained blinded after the results at 3 months' follow‐up. Other assessors were blinded to 3 months only.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

1 cell therapy participant and 2 controls were excluded from all analyses (1x reocclusion of target vessel, 2x withdrawal of consent). MRI was performed in 23 participants (12 cell therapy, 11 controls) at 3 months and 22 participants (12 cell therapy and 10 controls) at 15 months.

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

Hamshere 2015_IC

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: National Institute of Health Research (UK), Heart Cells Foundation, Barts and The London Charity, Chugai Pharma UK, and Cordis Corporation

Study setting: London, UK
Number of centres: 1
Length of follow‐up: 12 months
Number (N) of participants randomised to each arm: BMSC: 15; Controls: 15
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 15; Controls: 15

Participants

Description: Advanced HF (NYHA class II‐IV; optimal medical therapy and device therapy with no further treatment options).

Age distribution in each arm: n/r
Sex (% male) in each arm: n/r

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: n/r
Statistically significant baseline imbalances between the groups? None.

Interventions

Intervention arm: G‐CSF + BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: 50 mL BM was obtained from the posterior iliac crest. The BMSC fraction was obtained from the BM samples, and cells were resuspended in 10 mL autologous serum. All samples were maintained at room temperature for the entire procedure. Following arterial access, a weight‐adjusted (70 IU/kg) bolus dose of unfractionated heparin was administered. A coronary angiogram was performed to expose the largest possible area of the left ventricle to the injectate via the intact coronary circulation. The total 10 mL volume of injectate was divided equally and injected down patent coronary arteries or grafts, or both through an over‐the‐wire balloon catheter (Medtronic, Galway, Ireland). The balloon was inflated at low pressure to occlude blood flow, while the appropriate volume of injectate was delivered distally over 3 minutes. This procedure was repeated in the remaining target vessels.

Dose of stem cells: n/r
Timing of stem cell procedure: n/r

G‐CSF details: 10 ug/kg/day for 5 days

Comparator arm: G‐CSF + placebo (BM aspiration; 10 mL autologous serum)

Co‐intervention: standard medical therapy

Outcomes

Primary outcomes:

Change in global LVEF from baseline (12 months)
Secondary outcomes:

Change in quality of life (6 and 12 months), NT‐proBNP (6 months); major adverse cardiac events (12 months), change in NYHA class (12 months), change in CCS class (12 months).

Outcome assessment points: Baseline, 6 and 12 months.
Method(s) of outcome measurement: NYHA class; MRI, computed tomography

Notes

Outcome data for this trial were obtained directly from the study authors.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were enrolled in a 1:1 computer‐generated randomisation list.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants received G‐CSF, underwent bone marrow aspiration, and received a placebo infusion. Blinding of clinicians was not specifically reported, but the trial was described as "double‐blind".

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The endpoints of NYHA and CCS classifications were measured by an investigator blinded to the participant's treatment assignment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of mortality and morbidity on an intention‐to‐treat basis.

Selective reporting (reporting bias)

Low risk

Information for all outcomes requested from the authors was provided.

Other bias

High risk

Partially sponsored by Chugai Pharma UK and the Cordis Corporation. The primary investigator of this trial is an author of this Cochrane review. No other sources of bias were reported or identified.

Hamshere 2015_IM

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: National Institute of Health Research (UK), Heart Cells Foundation, Barts and The London Charity, Chugai Pharma UK, and Cordis Corporation

Study setting: London, UK
Number of centres: 1
Length of follow‐up: 12 months
Number (N) of participants randomised to each arm: BMSC: 15; Controls: 15
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 15; Controls: 15

Participants

Description: Advanced HF (NYHA class II‐IV; optimal medical therapy and device therapy with no further treatment options).

Age distribution in each arm: n/r
Sex (% male) in each arm: n/r

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: n/r
Statistically significant baseline imbalances between the groups? None.

Interventions

Intervention arm: G‐CSF + BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: 50 mL BM was obtained from the posterior iliac crest. The BMSC fraction was obtained from the BM samples and cells were resuspended in 10 mL autologous serum. All samples were maintained at room temperature for the entire procedure. After femoral arterial access, a weight‐adjusted (70 IU/kg) bolus dose of heparin was administered. Participants underwent left ventricular electromechanical mapping using NOGA XP Cardiac Navigation System (Biologics Delivery Systems Group, Cordis Corporation, CA, USA) and direct intramyocardial injection with a MyoStar injection catheter. The number of sampling points for the mapping procedure varied between 86 and 110. The target areas for injection were the border zones around the scar tissue based on voltage criteria obtained using the NOGA map (areas greater than 6.9 mV). Areas of the myocardium with a wall thickness of < 5 mm were avoided. The total 2 mL volume of injectate was divided and delivered equally to 10 target areas at approximately 1‐centimetre intervals.

Dose of stem cells: n/r
Timing of stem cell procedure: n/r

G‐CSF details: 10 ug/kg/day for 5 days

Comparator arm: G‐CSF + placebo (BM aspiration; 2 mL autologous serum)

Co‐intervention: standard medical therapy

Outcomes

Primary outcomes:

Change in global LVEF from baseline (12 months)
Secondary outcomes:

Change in quality of life (6 and 12 months), NT‐proBNP (6 months); major adverse cardiac events (12 months), change in NYHA class (12 months), change in CCS class (12 months).

Outcome assessment points: Baseline, 6 and 12 months.
Method(s) of outcome measurement: NYHA class; MRI, computed tomography

Notes

Outcome data for this trial were obtained directly from the study authors.

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were enrolled in a 1:1 computer‐generated randomisation list.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants received G‐CSF, underwent bone marrow aspiration, and received a placebo infusion. Blinding of clinicians was not specifically reported, but the trial was described as "double‐blind".

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The endpoints of NYHA and CCS classifications were measured by an investigator blinded to the participant's treatment assignment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of mortality and morbidity on an intention‐to‐treat basis.

Selective reporting (reporting bias)

Low risk

Information for all outcomes requested from the authors was provided.

Other bias

High risk

Partially sponsored by Chugai Pharma UK and the Cordis Corporation. The primary investigator of this trial is an author of this Cochrane review. No other sources of bias were reported or identified.

Heldman 2014_BM‐MSC

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: Partially funded by the Interdisciplinary Stem Cell Institute, Miller School of Medicine, BioCardia, and grant U54HL081028 from the National Heart, Lung, and Blood Institute Specialized Centers for Cell‐Based Therapy. Helical infusion catheters were provided by BioCardia Inc and one trial investigator (J. Hare) was supported by grants from the National Institutes of Health.

Study setting: Florida, USA
Number of centres: 1
Length of follow‐up: 12 months
Number (N) of participants randomised to each arm: BM‐MSC: 22; Controls: 11
Number (N) of participants analysed (primary outcome) in each arm: BM‐MSC: 19; Controls: 11

Participants

Description: Chronic MI and LV dysfunction (aged 21 to 90 years; ischaemic cardiomyopathy with LV dysfunction resulting from chronic MI, as documented by confirmed coronary artery disease with a corresponding area of myocardial akinesis, dyskinesis, or severe hypokinesis and had LVEF < 50% within 6 months of screening while taking maximally tolerated doses of beta‐adrenergic blocking and angiotensin‐converting enzyme or angiotensin II receptor blocking drugs and not during or recently after an ischaemic event).

Age distribution in each arm: BM‐MSC: 57.1 (10.6) years; Controls: 60.0 (12.0) years
Sex (% male) in each arm: BM‐MSC: 94.7%; Controls: 90.9%

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: n/r
Statistically significant baseline imbalances between the groups? Significantly higher baseline stroke volume in participants who had received MSC compared with placebo.

Interventions

Intervention arm: BMSC
Type of stem cells: Mesenchymal stem cells
Summary of stem cell isolation and type and route of delivery: All participants had bone marrow harvested. MSC were cultured from bone marrow aspirates. Delivery was by injection at 10 LV sites using TESI during left heart catheterisation using the helical infusion catheter (BioCardia). Injections were targeted to encircle the border zone of a chronically infarcted myocardial territory and defined by MRI and CT imaging, echocardiography, and well‐pacified biplane left ventriculography.

Dose of stem cells: n/r

Timing of stem cell procedure: 4 to 6 weeks from BM aspiration to cell administration.

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; vehicle placebo)

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes:

Incidence of treatment‐emergent serious adverse events (defined as composite of death, non‐fatal MI, stroke, hospitalisation for worsening HF, cardiac perforation, pericardial tamponade, ventricular arrhythmias > 15 sec, or with haemodynamic compromise or atrial fibrillation) at 1 month

Secondary outcomes:

  1. Serial troponin values (every 12 hours for the first 48 hours postcatheterisation)

  2. Serial creatine kinase values (every 12 hours for the first 48 hours postcatheterisation)

  3. Incidence of major adverse cardiac events (MACE) (defined as the composite incidence of (1) death, (2) hospitalisation for HF, or (3) non‐fatal recurrent MI) at 12 months

  4. Ectopic tissue formation (12 months)

  5. Number of deaths at 12 months

  6. Change from baseline in distance walked in 6 minutes (12 months)

  7. Change from baseline in the MLHFQ total score (12 months)

  8. Change from baseline in scar mass as a fraction of left ventricle mass by cardiac MRI or CT (12 months)

Additional outcomes:

Infarct size, regional wall motion at the sites of study agent injection, global LV size and function, exercise peak O2 consumption, NYHA class, quality of life measured at 3 and 6 months
Outcome assessment points: Baseline, 3 and 6 months (quality of life only), 12 months
Method(s) of outcome measurement: MRI

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

An electronic data entry system was used for randomisation. Participants were randomised to the MSC or BMMNC group, and further randomised within groups to cell therapy or placebo.

Allocation concealment (selection bias)

Low risk

Participants were randomised (unblinded) to the MSC or BMMNC group. Participants were further randomised (blinded) within groups to cell therapy or placebo.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants underwent BM harvest, and control participants received a placebo. Neither participants nor clinicians were aware of treatment allocation.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Preparation and administration of the study product was blinded to investigators outside the cell‐processing laboratory.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

3 cell therapy participants were excluded from the analysis of mortality and morbidity (2x withdrew consent, 1x cell‐processing failure). MRI analysis included 16 cell therapy participants and 17 controls (BMSC and MSC control groups combined); missing data were unexplained.

Selective reporting (reporting bias)

Unclear risk

All outcomes reported in the trial protocol (NCT00768066) were reported; some additional outcomes were reported in the publication of results.

Other bias

High risk

Received partial funding from BioCardia. No other sources of bias were reported or identified.

Heldman 2014_BMMNC

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: Partially funded by the Interdisciplinary Stem Cell Institute, Miller School of Medicine, BioCardia, and grant U54HL081028 from the National Heart, Lung, and Blood Institute Specialized Centers for Cell‐Based Therapy. Helical infusion catheters were provided by BioCardia Inc and one trial investigator (J. Hare) was supported by grants from the National Institutes of Health.

Study setting: Florida, USA
Number of centres: 1
Length of follow‐up: 12 months
Number (N) of participants randomised to each arm: BMSC: 22; Controls: 10
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 19; Controls: 10

Participants

Description: Chronic MI and LV dysfunction (aged 21 to 90 years; ischaemic cardiomyopathy with LV dysfunction resulting from chronic MI, as documented by confirmed coronary artery disease with a corresponding area of myocardial akinesis, dyskinesis, or severe hypokinesis and had LVEF < 50% within 6 months of screening while taking maximally tolerated doses of beta‐adrenergic blocking and angiotensin‐converting enzyme or angiotensin II receptor blocking drugs and not during or recently after an ischaemic event).

Age distribution in each arm: BMSC: 61.1 (8.4) years; Controls: 61.3 (9.0) years
Sex (% male) in each arm: BMSC: 89.5%; Controls: 100%

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: n/r
Statistically significant baseline imbalances between the groups? None.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: All participants underwent BM aspiration from the iliac crest. BMMNC were prepared by centrifugation of whole BM against a low‐density gradient using Ficoll‐Paque PREMIUM according to manufacturer's protocol. Cells were collected at the interface. Delivery was by injection at 10 LV sites using TESI during left heart catheterisation using the helical infusion catheter (BioCardia). Injections were targeted to encircle the border zone of a chronically infarcted myocardial territory and defined by MRI and CT imaging, echocardiography, and well‐pacified biplane left ventriculography.

Dose of stem cells: n/r
Timing of stem cell procedure: 4 to 6 weeks from BM aspiration to cell administration.

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; vehicle placebo)

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes:

Incidence of treatment‐emergent serious adverse events (defined as composite of death, non‐fatal MI, stroke, hospitalisation for worsening HF, cardiac perforation, pericardial tamponade, ventricular arrhythmias > 15 sec, or with haemodynamic compromise or atrial fibrillation) at 1 month

Secondary outcomes:

  1. Serial troponin values (every 12 hours for the first 48 hours postcatheterisation)

  2. Serial creatine kinase values (every 12 hours for the first 48 hours postcatheterisation)

  3. Incidence of the major adverse cardiac events (MACE) (defined as the composite incidence of (1) death, (2) hospitalisation for HF, or (3) non‐fatal recurrent MI) at 12 months

  4. Ectopic tissue formation (12 months)

  5. Number of deaths at 12 months

  6. Change from baseline in distance walked in 6 minutes (12 months)

  7. Change from baseline in MLHFQ total score (12 months)

  8. Change from baseline in scar mass as a fraction of left ventricle mass by cardiac MRI or CT (12 months)

Additional outcomes:

Infarct size, regional wall motion at the sites of study agent injection, global LV size and function, exercise peak O2 consumption, NYHA class, quality of life measured at 3 and 6 months
Outcome assessment points: Baseline, 3 and 6 months (quality of life only), 12 months
Method(s) of outcome measurement: MRI

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

An electronic data entry system was used for randomisation. Participants were randomised to the MSC or BMMNC group, and further randomised within groups to cell therapy or placebo.

Allocation concealment (selection bias)

Low risk

Participants were randomised (unblinded) to the MSC or BMMNC group. Participants were further randomised (blinded) within groups to cell therapy or placebo.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants underwent BM harvest, and control participants received a placebo. Neither participants nor clinicians were aware of treatment allocation.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Preparation and administration of the study product was blinded to investigators outside the cell‐processing laboratory.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

3 cell therapy participants were excluded from the analysis of mortality and morbidity (2x withdrew consent, 1x ineligible before BM aspiration). MRI analysis included 16 cell therapy participants and 17 controls (BMSC and MSC control groups combined); missing data were unexplained.

Selective reporting (reporting bias)

Unclear risk

All outcomes reported in the trial protocol (NCT00768066) were reported; some additional outcomes were reported in the publication of results.

Other bias

High risk

Received partial funding from BioCardia. No other sources of bias were reported or identified.

Hendrikx 2006

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: Not reported

Study setting: Hasselt, Belgium
Number of centres: 1
Length of follow‐up: 4 months
Number (N) of participants randomised to each arm: BMSC: 11; Controls: 12
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 10; Controls: 10

Participants

Description: Elective CABG surgery; transmural MI on ECG and akinesia or dyskinesia in part of the left ventricle as shown by angiography.
Age distribution in each arm: BMSC: 63.2 ± 8.5 years; Controls: 66.8 ± 9.2 years.
Sex (% male) in each arm: BMSC: 100%; Controls: 70%.

Number of diseased vessels: BMSC: 1 (n = 0), 2 (n = 2), 3 (n = 8); Controls: 1 (n = 1), 2 (n = 2), 3 (n = 7).
Time from symptom onset to initial treatment: BMSC arm: 217 (162) days and control arm: 213 (145) days between occurrence of MI and time of CABG (and treatment).
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: 40 mL of bone marrow was aspirated under local anaesthesia from the participant's iliac crest, the day before surgery. BMSC were immediately isolated by density gradient centrifugation using Lymphoprep. Isolated cells were washed twice with saline and subsequently resuspended in X‐VIVO 15 medium (Cambrex) supplemented with 2% autologous serum. This cell suspension was transferred to Teflon bags at a concentration of approximately 1 x 106 cells/mL for overnight cultivation. The next day cells were harvested and washed 3 times before finally being suspended in 10 mL heparinised saline. 10 mL of cell suspension were injected into the border zone of the infarct with 29‐gauge myoinjector syringes containing 0.5 mL of cell suspension. Multiple punctures were performed with prevent needles to make injections parallel to the epicardium and avoid delivery of cells into the ventricular cavity.
Dose of stem cells: 60.25 (31.35) x 106 cells with > 95% viability and over 73% recovery. Containing 1.42% (0.99%) CD34‐positive cells and 76.37 (44.47) CFU‐GM/105 mononuclear cells.
Timing of stem cell procedure: Approximately 24 hours following bone marrow aspiration; 217 (162) days post‐AMI.

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; heparinised saline)

Co‐intervention: CABG

Outcomes

Primary outcomes:

Global LVEF change and regional wall‐thickening changes in the infarct area.
Secondary outcomes:

Changes in metabolic activity measured by thallium scintigraphy.
Outcome assessment points: Baseline, postoperative (9 to 14 days), and 4 months
Method(s) of outcome measurement: MRI

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation (1:1) was carried out using sequentially numbered, sealed envelopes.

Allocation concealment (selection bias)

Low risk

Sealed envelopes were used.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Both treatment groups underwent BM aspiration: the BM group had bone marrow isolated the day before surgery from the iliac crest, and the control group had bone marrow aspirated from the sternum during the operation. Controls received a placebo. Both participants and the surgeon were unaware of whether cells or only saline was injected.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Cardiac MR images were analysed by an investigator blinded to treatment assignment. For thallium scintigraphy, 2 investigators independently analysed data and were blinded to treatment assignment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of mortality and morbidity. 3 participants (1x cell therapy and 1x control) were excluded from MRI analysis (2x death, 1x acute psychiatric illness).

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

Honold 2012

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: Not reported

Study setting: Frankfurt/Main, Germany
Number of centres: 1
Length of follow‐up: 60 months
Number (N) of participants randomised to each arm: BMSC: 23; Controls: 10
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 23; Controls: 9

Participants

Description: Coronary artery disease (aged > 18 years; previous MI at least 3 months prior to cell therapy and well demarcated LV regional wall motion abnormality; receiving constant state‐of‐the‐art pharmacotherapy for at least 3 months prior to enrolment).

Age distribution in each arm: BMSC: 53.4 ± 12.3 years; Controls: 58.8 ± 7.3 years.
Sex (% male) in each arm: BMSC: 82%; Controls: 100%.

Number of diseased vessels: BMSC: 1 (n = 10), 2 (n = 6), 3 (n = 6); Controls: 1 (n = 4), 2 (n = 2), 3 (n = 4).
Time from symptom onset to initial treatment: At least 3 months from previous MI.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: G‐CSF + BMSC
Type of stem cells: Circulating progenitor cells.
Summary of stem cell isolation and type and route of delivery: G‐CSF was administered to the participants for 5 days. 270 mL of peripheral blood was drawn. Mononuclear cells were isolated using a Ficoll gradient centrifugation, and cells were resuspended in X‐VIVO 15 medium with 1 ng/mL carrier‐free human recombinant VEGF, atorvastatin, and 20% human serum drawn from each individual participant. Cells were cultured ex vivo for 4 days to enrich in endothelial progenitor cells (uptake of LDL).
Dose of stem cells: 29 ± 12 x 106.
Timing of stem cell procedure: % days following G‐SCF administration and 4 days following bone marrow aspiration and cell culture.

G‐CSF details: 5 ug/kg/day (first 12 participants) or 10 ug/kg/day (20 participants) for 5 days.

Comparator arm: G‐CSF + control (no BM aspiration, no placebo)

Co‐intervention: Standard medical therapy; PCI (in 33% of participants)

Outcomes

Primary outcomes:

Safety and efficacy.
Secondary outcomes:

Global and regional LV function and volumes after 3 months, determined by both LV angiography and MRI. Clinical parameters like functional NYHA class, cardiopulmonary exercise testing, and NT‐proBNP serum levels were obtained during a 5‐year follow‐up period.
Outcome assessment points: Baseline and 3, 12, 60 months
Method(s) of outcome measurement: MRI

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The trial was described as randomised, but the method of randomisation was not reported.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Blinding of clinicians and participants was not specifically reported, but no placebo was administered to the control group.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

MRI independent observers were blinded.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

All randomised participants were included in the analysis of mortality and morbidity, although 1 participant randomised to (but who did not receive) cell therapy was analysed in the control group. Angiography was carried out at follow‐up in 26 participants (21 cell therapy, 5 controls). MRI was performed in a subset of participants (9 cell therapy, 4 controls).

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

Hu 2011

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: Key project in the National Science and Technology Pillar programme during the 11th 5‐year plan period (2006BAJ01A09), basic scientific research fund of the National Scientific Institute 2009‐2011.

Study setting: Beijing, China
Number of centres: 1
Length of follow‐up: 12 months
Number (N) of participants randomised to each arm: BMSC: 31; Controls: 29
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 31; Controls: 28

Participants

Description: Chronic HF (aged 18 to 75 years; at least 3 months since last MI; severe ischaemic cardiomyopathy with LVEF < 30% by MRI and suitable for CABG; no evidence of surviving myocardium in the infarct area, as shown by SPECT and LV angiography; without LV aneurysm or valvular diseases requiring surgical intervention).

Age distribution in each arm: BMSC: 56.6 ± 9.7 years; Controls: 58.3 ± 8.9 years.
Sex (% male) in each arm: 93.3% (both arms pooled).

Number of diseased vessels: BMSC: 3; Controls: 3.
Time from symptom onset to initial treatment: At least 3 months from last MI.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: After anaesthesia but before CABG, 60 mL of BM was aspirated from the participant's iliac crest and diluted with normal saline solution. The mononuclear cells were isolated using Ficoll density gradient centrifugation according to good manufacturing practice regulations and resuspended in 10 mL of saline solution. The cell suspension was filtered by a 70‐micrometre cell strainer before transplantation. The cells were counted under a light microscope, and the viability was assessed by trypan blue dye. The final suspension of BMMNC contained 107 mL MNC. Cells were delivered via the grafted vessel (saphenous vein graft).
Dose of stem cells: Mean 13.17 ± 10.66 x 107.
Timing of stem cell procedure: Within 24 hours and during CABG.

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; mixture of 8 mL of saline solution and 2 mL of the participant's own serum)

Co‐intervention: CABG

Outcomes

Primary outcomes:

Changes in LVEF from baseline to 6 months' follow‐up.
Secondary outcomes:

None reported.

Additional outcomes:

LVEDV index (MRI; echocardiograpy); LVESV index (MRI); wall motion score index (echocardiography); perfusion score (SPECT), 6‐min walking test, and BNP value.
Outcome assessment points: Baseline, 6 and 12 months
Method(s) of outcome measurement: MRI; echocardiography

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A randomisation table was generated by statistical software.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants underwent BM harvest, and control participants received a placebo. The study processes were blinded to surgeons and participants.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The study processes were blinded to investigators who were responsible for participant assessments.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants were included in the analysis of mortality and morbidity; 1 control participant did not attend follow‐up at 6 months. MRI at 12 months included 25 participants in each group. Echocardiography at 12 months included 42 participants (24 cell therapy and 18 controls); missing data were explained.

Selective reporting (reporting bias)

Low risk

All outcomes reported in the trial protocol (NCT00395811) were reported.

Other bias

Low risk

No other sources of bias were reported or identified.

Jimenez‐Quevedo 2011

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: Funded by an independent research grant from the Spanish National Ministry of Health and Social Policy (Direccion general de Terapias Avanzadas y Transplante) and an unrestricted grant from Mutua Madrileña Foundation.

Study setting: Spain
Number of centres: 3 (Madrid, Barcelona, Logrono)
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 19; Controls: 9
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 19; Controls: 9

Participants

Description: Refractory angina (CCS class II‐IV; optimal medical therapy; not suitable for surgical/percutaneous revascularisation; and with reversible perfusion defect measured by SPECT).
Age distribution in each arm: BMSC: median 70 years; Controls: median 58.2 years
Sex (% male) in each arm: BMSC: 78.9%; Controls: 100%

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: n/r (no AMI within preceding 3 months)
Statistically significant baseline imbalances between the groups? Median age significantly higher in treated group.

Interventions

Intervention arm: G‐CSF; BMSC
Type of stem cells: CD133+ progenitor cells from mobilised peripheral blood
Summary of stem cell isolation and type and route of delivery: All participants underwent leukapheresis to isolate the mononuclear fraction from the peripheral blood. Only those participants allocated to the cell group CD133+ PC were isolated by immunomagnetic selection with CliniMACS cell separation system (Miltenyi Biotec, Bergisch‐Gladback, Germany). Sterility tests (Gram stain and culture) were performed on the final cell preparation. The cells were suspended in normal saline and concentrated in 3 mL for the injection.

Dose of stem cells: 20 to 30 × 106 cells
Timing of stem cell procedure: At last 5 days after G‐CSF.

G‐CSF details: 5 μg/kg per 12 hours for 4 days

Comparator arm: Control (BM aspiration; sham procedure but no placebo administered)

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes:

Major adverse cardiac and cerebrovascular events, defined as cardiovascular death, non‐fatal MI, ischaemic stroke, need for revascularisation, or procedure‐related complications (pericardial effusion/cardiac tamponade, vascular complications, and sustained ventricular arrhythmias) at 6, 12, and 24 months.
Secondary outcomes:
Efficacy of the transendocardial injection of PC CD133+ assessed by means of the following variables: the change in the myocardial perfusion defect as measured by SPECT, symptom‐limited treadmill test, quality of life, CCS angina classification, and antianginal medication requirement.
Outcome assessment points: Baseline, 6 months
Method(s) of outcome measurement: Echocardiography, SPECT, LV angiography

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A centralised telephone randomisation was performed using a computer‐generated code before the index procedure.

Allocation concealment (selection bias)

Low risk

Randomisation was performed using a centralised telephone procedure.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Both groups were treated with G‐CSF, underwent an apheresis and electromechanical mapping; however, transendocardial injections were not performed in the control group but were simulated to keep the participant and all the investigators except the 2 operators who performed the injections blinded.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

A blinded investigator analysed angiograms with the use of a computer‐based system. All the analyses were centralised in an independent core laboratory blinded to the randomisation. All investigators except 2 operators who performed the injections were blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All participants were included in the analysis of all outcomes on an intention‐to‐treat basis.

Selective reporting (reporting bias)

Low risk

All outcomes reported in the trial protocol (NCT00694642) were reported.

Other bias

Low risk

No other sources of bias were reported or identified.

Losordo 2007

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: Supported in part by National Institutes of Health grants and by a grant from Baxter Healthcare. Biosense Webster provided the mapping and injection catheters for this study at no extra cost.

Study setting: USA
Number of centres: n/r (multicentre)

Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC‐high dose (HD): 6; BMSC‐medium dose (MD): 6; BMSC‐low dose (LD): 6; and Controls: 6
Number (N) of participants analysed (primary outcome) in each arm: BMSC‐HD: 6; BMSC‐MD: 6; BMSC‐LD: 6; and Controls: 6

Participants

Description: Chronic refractory angina (aged > 21 years; CCS class III–IV; optimal medical therapy; not suitable for revascularisation; ischaemia on nuclear perfusion imaging, to complete at least 1 minute but no more than 6 mins of a standard Bruce protocol, and to experience angina/angina equivalent during the baseline exercise test).

Age distribution in each arm: Mean 62.4 (range 48 to 84 years) for all groups.
Sex (% male) in each arm: 80% for all arms.

Number of diseased vessels: Not reported.
Time from symptom onset to initial treatment: Not reported, not applicable.
Statistically significant baseline imbalances between the groups? None reported.

Interventions

Intervention arm: G‐CSF, BMSC at low dose, medium dose, or high dose.
Type of stem cells: CD34+ cells from mobilised peripheral blood.
Summary of stem cell isolation and type and route of delivery: Leukoapheresis was performed on the 5th day for collection of mononuclear cells. The cells were stored overnight at 4°C, and the following morning the CD34+ fraction was purified on a commercially available device (Isolex 300i, Baxter Healthcare) according to manufacturer's instructions. Cells were then subjected to testing and were required to meet lot‐release criteria. Once passed, the participants underwent NOGA electromechanical mapping and intramyocardial injection of CD34+ cells suspended in saline plus 5% autologous serum, versus cell diluent using the NOGA MyoStar catheter. The dose was divided into 10 injections of 0.2 mL per injection.
Dose of stem cells: 5 x 104 CD34 cells/kg (LD); 1 x 105 CD34 cells/kg (MD); 5 x 105 CD34 cells/kg (HD).
Timing of stem cell procedure: On day 6 following G‐CSF administration and within 24 hours of cell isolation.

G‐CSF details: G‐CSF was given to all participants at 5 μg/kg for 4 to 5 days.

Comparator arm: G‐CSF; placebo (BM aspiration; saline (0.9% sodium chloride) with 5% autologous plasma).

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes: Not reported.
Secondary outcomes: Safety analysis (AEs), efficacy (angina frequency, NTG use, exercise tolerance, CCS class, SPECT perfusion imaging, quality of life testing).
Outcome assessment points: Baseline and 6 months
Method(s) of outcome measurement: Angina frequency and CCS angina class

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation codes were established by the study statistician.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants were administered G‐CSF prior to treatment and had CD34+ cells collected. Controls received a placebo solution in a syringe that was identical for all treatment arms.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Randomisation codes were only revealed to the stem cell laboratory technician responsible for separating the cells into aliquots or preparing the placebo material.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses of all outcomes at follow‐up.

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

High risk

Partially funded by a grant from Baxter Healthcare. No other sources of bias were reported or identified.

Losordo 2011

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: Baxter Healthcare sponsored the study and was responsible for the conduct of the investigation, with oversight provided by the principal investigator and the scientific advisory board.

Study setting: USA
Number of centres: 26
Length of follow‐up: 12 months
Number (N) of participants randomised to each arm: BMSC‐low dose (LD): 56; BMSC‐high dose (HD): 56; Controls: 56
Number (N) of participants analysed (primary outcome) in each arm: BMSC‐LD: 55; BMSC‐HD: 56; Controls: 56

Participants

Description: Chronic refractory angina (aged 21 to 80 years; CCS class III–IV; optimum medical management; not suitable for revascularisation; SPECT imaging to document the presence of reversible ischaemia; patients required to walk a minimum of 3 mins but no longer than 10 mins on a modified Bruce protocol exercise tolerance test and had to experience angina or their angina equivalent during exercise testing.

Age distribution in each arm: BMSC‐LD: 61.3 (9.1) years; BMSC‐HD: 59.8 ± 9.2 years; Controls: 61.8 ± 8.5 years.
Sex (% male) in each arm: BMSC‐LD: 83.6%; BMSC‐HD: 87.5%; Controls: 89.3%.

Number of diseased vessels: Not reported.
Time from symptom onset to initial treatment: At least 40 days from previous MI.
Statistically significant baseline imbalances between the groups? Cardiovascular risk factors (HTN, smoking, DM); angina episodes per week.

Interventions

Intervention arm: G‐CSF, BMSC at low dose or high dose.
Type of stem cells: CD34+ cells from mobilised peripheral blood.
Summary of stem cell isolation and type and route of delivery: On day 5 leukapheresis was performed. The following day mononuclear cells were collected and CD34+ cells enriched using a commercially available device (Isolex 300im) magnetic cell separation system. Cell suspension with > 70% viability and > 50% CD34+ cells were given at 2 doses of body weight with a maximum of 100 kg. Cell suspension was diluted in saline (0.9% sodium chloride) with 5% autologous plasma. Cells were injected into the myocardium. The injection was performed by NOGA mapping and at 10 sites (0.2 cm3/site) using a NOGA MyoStar catheter.
Dose of stem cells: 1 x 105 CD34 cells/kg (LD) or 5 x 105 CD34 cells/kg (HD).
Timing of stem cell procedure: At least 3 months following MI.

G‐CSF details: G‐CSF was given to all participants at 5 μg/kg for 4 to 5 days.

Comparator arm: G‐CSF; placebo (BM aspiration; saline (0.9% sodium chloride) with 5% autologous plasma).

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes:

Angina frequency 6 months after treatment.
Secondary outcomes:

None reported in study protocol.

Additional outcomes:

Efficacy endpoints including exercise tolerance testing; use of antianginal medication; CCS functional class; health‐related QOL (Seattle Angina Questionnaire, SF‐36, Dyspnea Questionnaire, EQ‐5D); combined rate of MACE, SPECT, cardiac MRI (in a substudy). Safety endpoints including adverse event reporting, chest X‐ray, and echocardiology and laboratory screening.
Outcome assessment points: Baseline, 6 and 12 months
Method(s) of outcome measurement: CCS functional class

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were randomly assigned to 1 of 3 treatment groups via a telephone call‐in and an interactive voice‐response system.

Allocation concealment (selection bias)

Low risk

The cell‐processing laboratory at each centre was responsible for making the randomisation call and preparing the CD34+ cells or control injection accordingly.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants were administered G‐CSF prior to treatment and had CD34+ cells collected. Controls received a placebo solution in a syringe that was identical for both treatment arms.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

An independent committee conducted the analysis. All study personnel remained blinded until the end of the study.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in analyses at follow‐up on an intention‐to‐treat basis.

Selective reporting (reporting bias)

Low risk

All outcomes reported in the trial protocol (NCT00300053) were reported.

Other bias

High risk

Baxter Healthcare sponsored the study and was responsible for the conduct of the investigation. No other sources of bias were reported or identified.

Mathiasen 2015

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: Supported by the Arvid Nilsson's Foundation, Aase and Ejnar Danielsen's Foundation, Agustinus Foundation, the Research Foundation at Rigshospitalet, Axel Muusfeldt Foundation, Eva and Henry Fraenkel's Foundation, Gangsted Foundation, Vera and Fleming Westerberg's Foundation, Jeppe and Ovita Juhl's Foundation, Sophus and Astrid Jacobsen Foundation.

Study setting: Copenhagen, Denmark
Number of centres: 1
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 40; Controls: 20
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 40; Controls: 20

Participants

Description: Severe ischaemic HF (aged 30 to 80 years; optimal medical therapy with no change in medication for 2 months; no revascularisation options, LVEF < 45%; NYHA class II‐III).

Age distribution in each arm: BMSC: 66.1 (7.7) years; Controls: 64.2 (10.6) years
Sex (% male) in each arm: BMSC: 90%; Controls: 70%

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: n/r
Statistically significant baseline imbalances between the groups? None.

Interventions

Intervention arm: BMSC
Type of stem cells: Mesenchymal stem cells
Summary of stem cell isolation and type and route of delivery: A total of 50 mL bone marrow aspirate was obtained from the iliac crest by needle aspiration under local anaesthesia. The marrow sample was diluted 1:2 with PBS. MNC were harvested by gradient centrifugation on lymphoprep (density 1077 g/cm3), then primary cell cultures were established by seeding 2 × 107 BMMNC using a T75 culture flask in complete medium (DMEM low glucose (1 g/L) with 25 mM HEPES and L‐Glutamine, 1% penicillin/streptomycin and 10% fetal bovine serum). The cells were incubated at 37°C in humid air with 5% CO2. The medium was changed 5 days after plating and subsequently every 3 or 4 days. After 2, 3, 4, and 5 weeks of cultivation, cells were harvested. Mesenchymal stromal cells were successfully culture expanded under good manufacturing practice conditions for 46.9 + 10.5 days. Participants were treated with the number of cells reached after 2 passages.

Dose of stem cells: mean 77.5 (67.9) x 10e6
Timing of stem cell procedure: Mesenchymal stromal cells were successfully culture expanded under good manufacturing practice conditions for 46.9 (10.5) days following BM aspiration.

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; PBS mixed with a drop of the participant’s blood to maintain blinded appearance of placebo solution).

Co‐intervention: Standard medical therapy

Outcomes

Primary outcome:

Changes in LVESV at 6 months' follow‐up
Secondary outcome:

Clinical improvements at 6 and 12 months

Note: the main study publication reports secondary outcomes as LVEDV, LVEF, SV, cardiac output, LV myocardial mass, wall thickness, wall thickening, scar volume, NYHA class, CCS class, 6MWT, weekly angina attacks and weekly use of nitroglycerine, biomarkers, the Seattle Angina Questionnaire and Kansas City Cardiomyopathy Questionnaire, and safety (Mathiasen 2015).
Outcome assessment points: Baseline, 6 months
Method(s) of outcome measurement: MRI; computed tomography

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were enrolled in a 2:1 computer‐generated randomisation list blocks of 6.

Allocation concealment (selection bias)

Low risk

The randomisation list was generated by a person unrelated to the study group.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

The trial investigators, study nurses, and participants were blinded to treatment allocation. To maintain blinding, a drop of the participant's blood was mixed into the syringe containing MSC or placebo by the stem cell laboratory to make the MSC solution and placebo identical.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The trial investigators and experienced physicians who performed the MRI analyses were blinded to treatment allocation.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in all analyses at follow‐up on an intention‐to‐treat basis.

Selective reporting (reporting bias)

Low risk

All outcomes reported in the trial protocol (NCT00644410) were reported.

Other bias

Low risk

No other sources of bias were reported or identified.

Mozid 2014_IC

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: National Institute of Health Research (UK), Heart Cells Foundation, Barts and The London Charity, Chugai Pharma UK, and Cordis Corporation.

Study setting: London, UK
Number of centres: 1
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 14; Controls: 2
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 14; Controls: 2

Participants

Description: Advanced HF (NYHA class II‐IV; optimal medical therapy and device therapy with no further treatment options).

Age distribution in each arm: Mean 70 (10) years
Sex (% male) in each arm: 94%

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: Duration since last MI: 11 (7) years
Statistically significant baseline imbalances between the groups? None.

Interventions

Intervention arm: G‐CSF + BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: 50 mL BM was obtained from the posterior iliac crest. The BMSC fraction was obtained from the BM samples, and cells were resuspended in 10 mL autologous serum. All samples were maintained at room temperature for the entire procedure. Following arterial access, a weight‐adjusted (70 IU/kg) bolus dose of unfractionated heparin was administered. A coronary angiogram was performed to expose the largest possible area of the left ventricle to the injectate via the intact coronary circulation. The total 10 mL volume of injectate was divided equally and injected down patent coronary arteries or grafts, or both through an over‐the‐wire balloon catheter (Medtronic, Galway, Ireland). The balloon was inflated at low pressure to occlude blood flow, while the appropriate volume of injectate was delivered distally over 3 minutes. This procedure was repeated in the remaining target vessels.

Dose of stem cells: Mean 8.6 (11.0) x 107
Timing of stem cell procedure: n/r

G‐CSF details: 10 ug/kg/day for 5 days

Comparator arm: G‐CSF + placebo (BM aspiration; 10 mL autologous serum)

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes:

None (change in global LVEF from baseline (12 months) is a primary outcome in the main REGENERATE‐IHD trial but not included in the pilot study)
Secondary outcomes:

Change in quality of life (6 and 12 months); NT‐proBNP (6 months); major adverse cardiac events (12 months); change in NYHA class (12 months)

Outcome assessment points: Baseline and 6 months
Method(s) of outcome measurement: NYHA class; MRI, computed tomography

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were enrolled in a 1:1 computer‐generated randomisation list.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants received G‐CSF, underwent bone marrow aspiration, and received a placebo infusion. Blinding of clinicians was not specifically reported, but the trial was described as "double‐blind".

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The endpoints of NYHA and CCS classifications were measured by an investigator blinded to the participant's treatment assignment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of mortality and morbidity on an intention‐to‐treat basis.

Selective reporting (reporting bias)

Unclear risk

This is a pilot study report that only reports 6‐month follow‐up of the secondary outcomes described in the protocol (NCT00747708).

Other bias

High risk

Partially sponsored by Chugai Pharma UK and the Cordis Corporation. The primary investigator of this trial is an author of this Cochrane review. No other sources of bias were reported or identified.

Mozid 2014_IM

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: National Institute of Health Research (UK), Heart Cells Foundation, Barts and The London Charity, Chugai Pharma UK, and Cordis Corporation.

Study setting: London, UK
Number of centres: 1
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 10; Controls: 8
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 10; Controls: 8

Participants

Description: Advanced HF (NYHA class II‐IV; optimal medical therapy and device therapy with no further treatment options).
Age distribution in each arm: Mean 64 (9) years
Sex (% male) in each arm: 100%

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: Duration since last MI: 7 (5) years
Statistically significant baseline imbalances between the groups? None.

Interventions

Intervention arm: G‐CSF + BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: 50 mL BM was obtained from the posterior iliac crest. The BMSC fraction was obtained from the BM samples, and cells were resuspended in 10 mL autologous serum. All samples were maintained at room temperature for the entire procedure. After femoral arterial access, a weight‐adjusted (70 IU/kg) bolus dose of heparin was administered. Participants underwent left ventricular electromechanical mapping using NOGA XP Cardiac Navigation System (Biologics Delivery Systems Group, Cordis Corporation, CA, USA) and direct intramyocardial injection with a MyoStar injection catheter. The number of sampling points for the mapping procedure varied between 86 and 110. The target areas for injection were the border zones around the scar tissue based on voltage criteria obtained using the NOGA map (areas greater than 6.9 mV). Areas of the myocardium with a wall thickness of < 5 mm were avoided. The total 2 mL volume of injectate was divided and delivered equally to 10 target areas at approximately 1‐centimetre intervals.

Dose of stem cells: Mean 5.2 (5.3) x 107
Timing of stem cell procedure: n/r

G‐CSF details: 10 ug/kg/day for 5 days

Comparator arm: G‐CSF + placebo (BM aspiration; 2 mL autologous serum)

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes:

None (change in global LVEF from baseline (12 months) is a primary outcome in the main REGENERATE‐IHD trial but not included in the pilot study)
Secondary outcomes:

Change in quality of life (6 and 12 months); NT‐proBNP (6 months); major adverse cardiac events (12 months); change in NYHA class (12 months)

Outcome assessment points: Baseline and 6 months
Method(s) of outcome measurement: NYHA class; MRI, computed tomography

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Participants were enrolled in a 1:1 computer‐generated randomisation list.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants received G‐CSF, underwent bone marrow aspiration, and received a placebo infusion. Blinding of clinicians was not specifically reported, but the trial was described as "double‐blind".

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The endpoints of NYHA and CCS classifications were measured by an investigator blinded to the participant's treatment assignment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of mortality and morbidity on an intention‐to‐treat basis.

Selective reporting (reporting bias)

Unclear risk

This is a pilot study report that only reports 6‐month follow‐up of the secondary outcomes described in the protocol (NCT00747708).

Other bias

High risk

Partially sponsored by Chugai Pharma UK and the Cordis Corporation. The primary investigator of this trial is an author of this Cochrane review. No other sources of bias were reported or identified.

Nasseri 2012

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: Supported in part by Miltenyi Biotec and by the German Bundesministerium fur Bildung und Forschung.

Study setting: Berlin, Germany
Number of centres: 1
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 30; Controls: 30
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 28; Controls: 26

Participants

Description: Chronic IHD (indication for CABG surgery; reduced global LVEF by transthoracic echocardiography at rest (LVEF ≤ 35); presence of akinetic or hypokinetic and hypoperfused LV myocardium on MRI for defining the target area).
Age distribution in each arm: BMSC: 61.9 (7.3) years; Controls: 62.7 (10.6) years
Sex (% male) in each arm: BMSC: 93%; Controls: 97%

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: Duration since last MI: BMSC: 2.6 months (range 17 days to 17.1 years); Controls: 1.5 months (range 14 days to 28.5 years).
Statistically significant baseline imbalances between the groups? None.

Interventions

Intervention arm: BMSC
Type of stem cells: CD133+ progenitor cells
Summary of stem cell isolation and type and route of delivery: All participants underwent BM aspiration from the left posterior iliac crest with local anaesthesia and analgosedation. An average BM volume of 196 +/‐ 28 mL was harvested and diluted with 20 mL saline solution containing 1000 U heparin. The BM solution was filtered, transferred into a plastic bag, and washed with PBS/EDTA solution containing 0.5% human serum albumin. This cell suspension was incubated with human IgG 5% as blocking reagent, labelled with 7.5mL reconstituted CD133 MicroBeads, murine anti‐human CD133 monoclonal antibodies conjugated to superparamagnetic iron dextran particles. Then CD133+ cells were separated using the CliniMACS Magnetic Separation device. The enriched cell fraction was reconstituted with 13 mL saline containing 10% autologous serum. Samples were drawn for cell numbers, purity, viability, and proof of sterility. Finally, cells were aliquoted into 1‐millilitre syringes and stored at 4°C without heparin.

Dose of stem cells: Median 5.1 (IQR 3.0 to 9.1) x 106 CD133+ cells
Timing of stem cell procedure: 36 hours after BM aspiration

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; isotonic sodium chloride solution containing 10% autologous serum)

Co‐intervention: CABG

Outcomes

Primary outcomes:

LVEF at rest, measured 6 months' postoperatively by MRI.
Secondary outcomes:

  1. Change in LVEF compared with preoperatively and early postoperatively

  2. Regional contractility in the area of interest

  3. Physical exercise capacity determined by 6‐minute walk test

  4. Perfusion in the AOI

  5. Change in LV dimensions

  6. NYHA and CCS class

  7. MLHFQ

  8. Death, MI, or need for reintervention during follow‐up

Post‐hoc outcome:

Infarct scar size.
Outcome assessment points: Baseline, 6 months
Method(s) of outcome measurement: MRI

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was conducted in the cell preparation facility. Group allocation was performed according to a predefined non‐block‐wise 1:1 randomisation plan.

Allocation concealment (selection bias)

Low risk

The randomisation plan was accessible only to the external cell processing team that prepared the cell product or placebo.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants underwent bone marrow aspiration. Syringes were prepared with either cells or placebo solution, and an ID number was added so that participants, the surgical team, and all investigators were unaware of treatment allocation.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Syringes were prepared with either cells or placebo solution, and an ID number was added so that participants, the surgical team, and all investigators were unaware of treatment allocation.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of mortality and morbidity (3x cell therapy and 1x control were excluded from exercise testing). In MRI analysis, the number of withdrawals was low (treatment: 2/30 vs control: 4/30), and reasons for exclusion were given. 4 participants (3 cell therapy, 1 control) did not undergo exercise tests.

Selective reporting (reporting bias)

Low risk

All outcomes reported in the trial protocol (NCT00462774) were reported. One post‐hoc outcome was clearly defined as such.

Other bias

High risk

Supported in part by Miltenyi Biotec and the German Bundesministerium fur Bildung und Forshcung. Two authors received lecture fees from Miltenyi Biotec. No other sources of bias were reported or identified.

Patel 2005

Methods

Type of study: Parallel RCT
Type of publication: Full paper (6 months); abstract (10 years)
Source of funding: Not reported.

Study setting: Rosario, Argentina
Number of centres: 1
Length of follow‐up: 10 years
Number (N) of participants randomised to each arm: BMSC: 10, Controls: 10 (pilot study); BMSC: 25, Controls: 25 (long‐term follow‐up)
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 10, Control: 10 (pilot study); BMSC: 25, Controls: 25 (long‐term follow‐up)

Participants

Description: Ischaemic HF (LVEF < 35% by echocardiography and multiplanar cardiac catheterisation; NYHA class III or IV; requiring revascularisation, undergoing off‐pump CABG).

Age distribution in each arm: BMSC arm: 64.8 ± 7.1 years old; Control arm: 63.6 ± 5.2 years old (pilot study data).
Sex (% male) in each arm: BMSC arm: 80%; Control arm: 80% (pilot study data).

Number of diseased vessels: Not reported.
Time from symptom onset to initial treatment: At least 7 days after the last MI, all participants had history of MI and revascularisation by PCI.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC
Type of stem cells: CD34+ progenitor cells
Summary of stem cell isolation and type and route of delivery: Bone marrow was harvested from the iliac bone in a sterile fashion after achievement of general anaesthesia. A special multihold harvest needle with a 60‐millilitre syringe was designed to minimise the anaesthetic time. It was introduced into the iliac bone between both posterior iliac spines at both sides. 500 mL to 600 mL of BM with a minimal number of puncture sites was harvested. At least 250 mL BM must have been harvested to continue with the protocol. Harvested BM was placed in a blood bag with 10,000 U of heparin sulfate and 400 μm of lysine acetylsalicylate to avoid platelet clumping. The BM was filtered on a 500‐micrometre filter followed by a 200‐micrometre filter. The resulting solution was mixed with hydroethylstarch 6%. The supernatant was centrifuged at 400 g for 15 mins. The cellular pellet was resuspended in PBS. The cell solution was mixed 3:1 with a solution of 155 mmol/L ammonium chloride, 10 mmol/L potassium bicarbonate, and 0.1 mmol/L EDTA and set for 5 mins at room temperature. The solution was then centrifuged at 400 g for 10 mins. The pellet was washed with PBS and resuspended. The cell suspension was placed over Ficoll‐Paque (1.077 density) 4:1 and centrifuged at 400 g for 30 mins. The upper layer was aspirated, leaving the mononuclear cell layer at the interphase. The interphase cells were transferred to a new conical tube with PBS and centrifuged at 300 g for 10 mins. The supernatant was completely removed, and the cell pellet was resuspended in PBS. Cell counts were performed, and the magnetic labelling with Isolex 300i was performed to obtain an enriched product of at least 70% CD34+ cells. The resulting cell solution was resuspended in 30 mL of the participant's own plasma and 10,000 U of heparin sulphate. 30 mL of cell preparation was delivered in 1 mL aliquots over a 2‐second period. The injections into the myocardium were spaced 1 cm apart and spaced to avoid coronary vessels. Injections were 3 mm to 5 mm in depth.
Dose of stem cells: Median 22 x 106 CD34+ cells.
Timing of stem cell procedure: At least 7 days following last MI.

G‐CSF details: No G‐CSF administered.

Comparator arm: Control (no BM aspiration, no placebo)

Co‐intervention: CABG

Outcomes

Primary outcomes: Not reported.
Secondary outcomes: Global LVEF, LVEDV, NYHA class (6 months only), arrhythmias (6 months) only.
Outcome assessment points: Baseline and 1, 3, and 6 months; 1, 5, and 10 years
Method(s) of outcome measurement: SPECT; echocardiography

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A person who did not participate in the trial had the choice of picking a coloured ball (red = BMSC arm; blue = control arm).

Allocation concealment (selection bias)

Unclear risk

The person undertaking the randomisation procedure did not participate in the trial.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Clinicians were not blinded. The authors report that participants were blinded, although the control group did not undergo bone marrow aspiration and no placebo was used.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The reviewers of imaging studies (cardiologists) were blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analyses of all outcomes at follow‐up.

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

Patel 2015

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: Funded by Harvest Technologies, Plymouth, MA. One study investigator (A. Patel) has "compensated honoraria" from Cook Medical Inc.

Study setting: Utah and California, USA; Rostock and Berlin, Germany; and Grugoan, India
Number of centres: 5
Length of follow‐up: 12 months
Number (N) of participants randomised to each arm: BMSC: 24; Controls: 6
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 22; Controls: 6

Participants

Description: CHF (aged > 18 years; LVEF < 40% by contrast echocardiography, NYHA class III or IV, stable with standard medical therapy for at least 1 month before screening, and a life expectancy of 6 months or longer).

Age distribution in each arm: BMSC: 58.5 (12.7) years; Controls: 52.7 (8.5) years.
Sex (% male) in each arm: BMSC: 91.7%; Controls: 100%.

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: n/r (at least 7 days since last MI).
Statistically significant baseline imbalances between the groups? None.

Interventions

Intervention arm: BMSC
Type of stem cells: Bone marrow cell concentrate
Summary of stem cell isolation and type and route of delivery: Approximately 240 mL of bone marrow was aspirated from the posterior iliac crest. The samples was concentrated to a volume of 60 mL over 15 mins using the Harvest Bone Marrow Aspirate Concentrate System (from Harvest Technologies, Plymouth, MA). The concentrate comprised mononuclear cells. Retrograde delivery through the coronary sinus, accessed via the right femoral vein using a 7F 8 mm x 40 mm balloon catheter (Cook Medical Inc) under fluoroscopic guidance. The 60 mL of BM concentrate was infused continuously over a 5‐minute period. The balloon was kept inflated for 10 min afterwards to allow the migration of cells into the cardiac tissue.

Dose of stem cells: Mean 3.7 (0.9) x 109 nucleated cells.
Timing of stem cell procedure: 29 (14) minutes from venous access.

G‐CSF details: No G‐CSF administered.

Comparator arm: Control (no BM aspiration; no placebo)

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes:

Number of participants with adverse events as a measure of safety and tolerability (12 months)
Secondary outcomes:

To assess the effect of the infusion of bone marrow nucleated cells on the clinical course of angina as measured by QOL questionnaire, MLHFQ, NYHA and CCS classification, and SPECT (12 months)

Assess the effect of the infusion of bone marrow nucleated cells on the clinical course of HF (12 months)
Outcome assessment points: Baseline; 1, 3, 6, and 12 months
Method(s) of outcome measurement: SPECT, NYHA class, CCS class, MLHFQ

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

"Electronic randomisation" was performed.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo was used; neither participants nor clinicians were blinded.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All imaging and follow‐up information was blinded to the reviewers.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

With the exception of 2 early withdrawals with reasons clearly defined, all randomised participants were included in the analysis of mortality and morbidity on an intention‐to‐treat basis.

Selective reporting (reporting bias)

Low risk

All outcomes reported in the trial protocol (NCT01299324) were reported.

Other bias

High risk

Commercially sponsored study (funded by Harvest Technologies). No other sources of bias were reported or identified.

Patila 2014

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: Supported by the Heart Research Foundation, the Academy of Finland and government subsidies for medical research block grants.

Study setting: Helsinki, Finland
Number of centres: 1
Length of follow‐up: 12 months
Number (N) of participants randomised to each arm: BMSC: 20; Controls: 19
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 18; Controls: 17

Participants

Description: Ischaemic HF (aged 18 to 75 years; managed by optimal medical care; undergoing CABG; LVEF between 15% and 45%; NYHA class II‐IV HF symptoms).
Age distribution in each arm: BMSC: median 65 (IQR 57 to 73) years; Controls: median 64 (IQR 58 to 70) years.
Sex (% male) in each arm: BMSC: 94.7%; Controls: 95%

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: n/r
Statistically significant baseline imbalances between the groups? None.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: After anaesthesia induction, 100 mL of BM aspirated from each participant's posterior iliac crests was collected into a sterile bag containing heparin (final concentration 20 units/mL). The aliquots were filtered and density‐gradient centrifuged (Ficoll‐Paque PREMIUM; GE Healthcare Bio‐Sciences AB; COBE 2991 Cell Processing Centrifuge) to obtain the mononuclear cell traction, according to standard methods. The cells were washed with medium 199 containing human serum albumin 0.5% and heparin (20 unit/mL) and finally suspended in 6 mL of the same medium. The cell suspension was divided into six 1‐millilitre syringes for each participant in the treatment group. After BM aspiration, standard CABG operation was performed under cardiac arrest, cardiopulmonary bypass, cardioplegia protection, and mild hypothermia. After completion of bypass anastomoses, each participant received, under cardiac arrest, 15 to 20 0.2 mL injections into the infarction border area through a small 24G needle into sites chosen before surgery using imaging data. Injection procedure was carefully photographed during each surgery, and segments injected were specified in participants' documentation for analysis.

Dose of stem cells: Median 8.4 (range 5.2 to 13.5) x 108 cells
Timing of stem cell procedure: n/r

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; vehicle medium)

Co‐intervention: CABG

Outcomes

Primary outcomes:

Change in LVEF after 1‐year follow‐up measured by MRI (12 months)
Secondary outcomes:

Changes in any other cardiac parameters as measured by echocardiography, MRI, or PET ischaemia area (6 months; 1 year)

Change in plasma concentrations of proBNP (6 months; 1 year)

Primary hospitalisation or days stayed in hospital

Correlation between pericardial fluid growth factor concentrations and left ventricular function improvement (up to 1 year)

Correlation between autologous cardiac stem cell quality and left ventricular function improvement (6 months; 1 year)
Outcome assessment points: Baseline, 1 week, 1 year.
Method(s) of outcome measurement: MRI, PET

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Before examination, numbered randomisation envelopes were sealed by stem cell laboratory personnel blinded to other participants. After delivery of the BM harvest to the stem cell laboratory, randomisation of each participant was done at the time of operation.

Allocation concealment (selection bias)

Low risk

Numbered, sealed envelopes were prepared by the stem cell laboratory before examinations.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants underwent bone marrow aspiration, and the control group received a placebo. Syringes containing cell therapy or placebo were masked using a non‐transparent tape.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

1 investigator analysed all MRI imaging data in a random order. Areas of scar and ischaemic myocardium were assessed by 2 study‐blind, experienced nuclear medicine physicians.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of mortality and morbidity. The number of withdrawals from MRI analysis was low (treatment: 2/20 vs control: 2/19), and reasons for withdrawals were reported in detail.

Selective reporting (reporting bias)

Unclear risk

All outcomes reported in the trial protocol (NCT00418418) were reported, although results were reported for 12 months only and not at 6 months as specified in the trial protocol.

Other bias

Low risk

No other sources of bias were reported or identified.

Perin 2011

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: No extramural funding was used to support this work; the authors have no disclosures, no funding, and no relationship with industry to report.

Study setting: Texas and Minneapolis, USA
Number of centres: 2
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 20; Controls: 10
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 20; Controls: 10

Participants

Description: Ischaemic HF (functional class III or IV (angina) and/or HF symptoms (NYHA) on maximal medical therapy; chronic CAD with a reversible perfusion defect ≥ 7% (SPECT); LVEF < 40%; MVO2 < 21 mL/kg/min; ineligible for percutaneous or surgical revascularisation).
Age distribution in each arm: BMSC: 56.3 ± 8.6 years; Controls: 60.5 ± 6.4 years.
Sex (% male) in each arm: BMSC: 50%; Controls: 80%.

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: n/r
Statistically significant baseline imbalances between the groups? None.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: 50 mL of BM was aspirated from the posterior iliac crest, approximately 4 hours before the cells were injected into the heart. Mononuclear cells were isolated using a density gradient centrifugation, washed in heparinised saline containing 5% human serum albumin and passed through a mesh. 3 x 107 cells were resuspended in 3 mL saline containing serum albumin (5%). 3 mL were preserved for further studies. 3 hours after bone marrow aspiration, participants underwent an electromechanical mapping to select myocardial segments for cell injection. Cells were injected into viable myocardium (> 6.9 mV unipolar voltage). Electromechanical maps comprised an average of 87 ± 16 points. Each injection of 2 million cells was delivered in a volume of 0.2 mL. Participants received an average of 15 cell injections in a mean of 6 ± 1 segments.
Dose of stem cells: 2 x 106 cells.
Timing of stem cell procedure: Within 24 hours of harvesting the bone marrow.

G‐CSF details: No G‐CSF administered.

Comparator arm: Control (no BM aspiration; sham procedure performed but no placebo administered).

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes:

Safety of cell injections at 3 time points: i) early safety (periprocedural and up to 2 weeks); ii) 3 months; and iii) 6 months: major adverse events (hospitalisation, arrhythmia, exacerbation of CHF, acute coronary syndrome, MI, stroke, or death).
Secondary outcomes:

  1. CCS angina score (baseline, 3 months, 6 months)

  2. NYHA classification (baseline, 3 months, 6 months)

  3. Myocardial oxygen consumption (baseline, 3 months, 6 months)

  4. Ejection fraction measured by echocardiography (baseline, 3 months, 6 months)

  5. Minute ventilation ‐ CO2 production relationship (VE/VCO2 slope) (baseline, 3 months)

  6. Wall motion score index measured by echocardiography (baseline, 3 months)

  7. LVEF measured by SPECT (baseline, 3 months, 6 months)

  8. LVEF measured by angiography (baseline, 6 months)

  9. LVEDV and LVESV (baseline and 6 months)

  10. Endocardial unipolar voltages (UPV) (baseline, 6 months)

  11. Linear local shortening (baseline, 6 months)

Outcome assessment points: Baseline, 3 and 6 months
Method(s) of outcome measurement: Not applicable.

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation tables were prepared by the statistical department.

Allocation concealment (selection bias)

Low risk

Numbered, sealed envelopes were used.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Clinicians were not blinded, but participants received a simulated mock injection procedure (although it was unclear whether BM aspiration was undertaken in control group).

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Efficacy studies were read by an independent, blinded investigator. Blinding was maintained until the end of the assessment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of all outcomes at follow‐up.

Selective reporting (reporting bias)

Low risk

All outcomes reported in the trial protocol (NCT00203203) were reported.

Other bias

Low risk

No other sources of bias were reported or identified.

Perin 2012a

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: NHLBI under co‐operative agreement 5 U01 HL087318‐04. In part by NHLBI contracts N01‐HB37164 and HHSN268201000008C awarded to the Molecular and Cellular Therapeutics Facility, University of Minnesota and NO1‐HB‐37163 and HHSN268201000007C awarded to the Cell Processing Facility, Baylor College of Medicine and National Center for Research Resources CTSA grant UL1 TR000064 awarded to the University of Florida. The CCTRN also acknowledges its industry partners, BioSafe, Biologics Delivery Systems Group, and Cordis Corporation for their contributions of equipment and technical support during the conduct of the trial. (Full details and conflict of interest declarations in the paper.)

Study setting: USA
Number of centres: 5
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 61; Controls: 31
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 54; Controls: 28

Participants

Description: Chronic IHD (aged > 18 years; clinically stable coronary artery disease, LVEF ≤ 45%, limiting angina (CCS class II‐IV) and/or CHF (NYHA class II‐III), a perfusion defect by SPECT, and no revascularisation options while receiving guideline‐based medical therapy).

Age distribution in each arm: BMSC: 63.95 ± 10.90 years; Controls: 62.32 ± 8.25 years.
Sex (% male) in each arm: BMSC: 86.89%; Controls: 93.65%.

Number of diseased vessels: Not reported.
Time from symptom onset to initial treatment: Not reported.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: Approximately 80 mL to 100 mL of BM was aspirated from the iliac crest using standard techniques. The aspirate was processed using Ficoll with a closed, automated cell processing system (Sepax). Composition of CD34 and CD133 cells was determined by flow cytometry. Cells passed stipulated lot release criteria, included viability (> 70%) and sterility. The target dose was 100 x 106 total BMC. The BMC final product was suspended in normal saline containing 5% human serum albumin and adjusted to a concentration of 100 x 106 cells in 3 mL distributed into three 1‐millilitre syringes. The placebo group received a cell‐free suspension in the same volume. Mean (SD) volume of BM harvested was 93.7 (8.3) mL. Total dose of 100 x 106 contained an average of 2.6% of CD34 cells and 1.2% of CD133 cells. Cells were delivered by intramyocardial injection. The cell‐containing or cell‐free preparation was delivered to viable myocardial regions identified during electromechanical mapping of the LV endocardial surface (NOGA).
Dose of stem cells: 100 x 106 BMSC.
Timing of stem cell procedure: Within 12 hours of cell harvest.

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; cell‐free suspension in the same volume).

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes:

  1. Change in LVESV (baseline, 6 months)

  2. Change in maximal oxygen consumption (baseline, 6 months)

  3. Change in reversible defect size on SPECT (baseline, 6 months)

Secondary outcomes:

  1. Regional wall motion by MRI (baseline, 6 months)

  2. Regional blood flow improvement by MRI (baseline, 6 months)

  3. Regional wall motion by echocardiography (baseline, 6 months)

  4. Clinical improvement in CCS classification (baseline, 6 months)

  5. Clinical improvement in NYHA classification (baseline, 6 months)

  6. Number of participants with a decrease in antianginal medication (baseline, 6 months)

  7. Exercise time and level (6MWT) (baseline, 6 months)

  8. Serum BNP levels in participants with CHF (baseline, 6 months)

  9. LV diastolic dimension measured by echocardiography (baseline, 6 months)

  10. Incidence of a major adverse cardiac event (baseline, 6 months)

  11. Reduction in fixed perfusion defect via SPECT (baseline, 6 months)

Outcome assessment points: Baseline and 6 months
Method(s) of outcome measurement: Echocardiography and SPECT

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was computer generated and used variable block sizes of 6 or 9, randomly selected and stratified by centre.

Allocation concealment (selection bias)

Low risk

Treatment assignment was masked to all but 1 designated cell‐processing team member at each centre not involved in participant care.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants underwent BM aspiration, and the control group received a placebo injection. All caregivers and participants were masked to treatment.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The study was described as “double‐blind”. Major adverse clinical events were assessed by 2 independent cardiologists not affiliated with any clinical site and masked to treatment assignment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of mortality and morbidity outcomes. Reasons for loss to follow‐up and withdrawals from echocardiography and SPECT analysis were given, with similar attrition rates in both treatment arms.

Selective reporting (reporting bias)

Low risk

All outcomes reported in the trial protocol (NCT00824005) were reported.

Other bias

Low risk

No other sources of bias were reported or identified.

Perin 2012b

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: "This work was supported solely by Aldagen, Inc, Durham, NC".

Study setting: Texas, USA
Number of centres: 1
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 10; Controls: 10
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 10; Controls: 10

Participants

Description: Advanced ischaemic HF (CCS class II‐IV angina or NYHA class II‐III HF; optimal medical therapy, LVEF < 45% by echocardiography; presence of a reversible perfusion defect on SPECT, ineligible for percutaneous or surgical revascularisation).
Age distribution in each arm: BMSC: 58.2 ± 6.1 years; Controls: 57.8 ± 5.5 years.
Sex (% male) in each arm: BMSC: 90%; Controls: 80%.

Number of diseased vessels: Not reported.
Time from symptom onset to initial treatment: At least 1 month from the last MI.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC
Type of stem cells: ALDH+ cells
Summary of stem cell isolation and type and route of delivery: 100 mL (± 20) BM was harvested from the iliac crest under local anaesthesia unless institutional guidelines required general anaesthesia. Bone marrow cells were depleted of CD15 and glycophorin‐A‐expressing cells using immunomagnetic beads (EasySep). The cells were reacted with ALDH substrate and ALDH bright (+) cells were isolated using a cell sorter (MoFlo or FACSAria). After centrifugation, the cells were resuspended in 3.5 mL 5% pharmaceutical grade human serum albumin. The final products were transferred to a 3‐millilitre fluorinated ethylene propylene bag with a needles entry port. ALDH (+) cells were administered intramyocardially via a NOGA MyoStar catheter. Cells comprised a mean of 0.74% (0.28%) of the nucleared BM cells in the unprocessed aspirates from participants (median 0.73%, range 0.35% to 1.16%). Cell injections were targeted at areas of the myocardium identified as ischaemic or SPECT and as viable by EMM.
Dose of stem cells: 15 injections in a volume of 0.2 mL per injection. Mean number of nucleated cells administered to the treatment group was 2.94 ± 1.58 x 106 cells (median 2.78 x 106, range 0.53 to 5.42 x 106). When the total cell doses were corrected for the proportion of ALDH (+) cells in the cell product, the mean number of ALDH (+) cells administered to the cell treatment group was 2.37 ± 1.31 x 106 (median 2.27 x 106, range 0.35 to 4.42 x 106).
Timing of stem cell procedure: Products manufactured at Aldagen were administered within 50 to 55 hours of BM aspiration, whereas those produced locally at the University of Texas were administered within 30 to 36 hours of aspiration.

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; 5% albumin).

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes:

Safety (combined early and late adverse events) (baseline, 6 months)

Secondary outcomes:

  1. NYHA classification (baseline, 6 months)

  2. CCS score (baseline, 6 months)

  3. EF measured by echocardiography (baseline, 6 months)

  4. LVESV; LVEDV (baseline, 6 months)

  5. Wall motion score index measured by echocardiography (baseline, 6 months)

  6. Myocardial oxygen consumption (baseline, 6 months)

  7. Total severity score (stress, rest, and reversible) (baseline, 6 months)

Outcome assessment points: Baseline and 6 months
Method(s) of outcome measurement: SPECT

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

A computer‐generated randomised sequence was used.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Control participants underwent an identical bone marrow harvest procedure, including insertion of the needle, except that BM was not aspirated. Control participants received transendocardial injections of placebo solution instead of cell preparation. All personnel involved were blinded. Personnel involved in the harvesting procedure acted independently of the study team, thus maintaining blinding.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The trial was described as “double‐blind”. Two blinded, independent echocardiologists reviewed the echocardiograms, and the average of the 2 readings was reported.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of all outcomes.

Selective reporting (reporting bias)

Low risk

All outcomes reported in the trial protocol (NCT00314366) were reported.

Other bias

High risk

This work was supported solely by Aldagen Inc, Durham, NC. No other sources of bias were reported or identified.

Pokushalov 2010

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: Not reported.

Study setting: Russia
Number of centres: 1
Length of follow‐up: 12 months
Number (N) of participants randomised to each arm: BMSC: 55; Controls: 54
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 49; Controls: 33 at the end of study

Participants

Description: Chronic MI and end‐stage chronic HF (history of MI > 12 months before enrolment and fixed perfusion defect on technetium‐99m tetrofosmin SPECT; clinical symptoms of HF; ineligible for revascularisation; LVEF < 35% as determined by 2‐dimensional echocardiography).
Age distribution in each arm: BMSC: 61 ± 9 years; Controls: 62 ± 5 years.
Sex (% male) in each arm: BMSC: 87%; Controls: 85%.

Number of diseased vessels: BMSC: 1 (n = 2), 2 (n = 1), 3 (n = 52); Controls: 1 (n = 3), 2 (n = 3), 3 (n = 48).
Time from symptom onset to initial treatment: History of MI > 12 months before enrolment.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: On the day of surgery, BM was aspirated from the iliac crest under local anaesthesia by the standard technique. BMMNC were isolated by Ficoll density gradient centrifugation. 3 washing steps were performed, and the cells were resuspended in heparinised saline for further use. Cell viability was tested by trypan blue (exclusion method) and estimated at more than 98% for each transplant. Intramyocardial injection. Non‐fluroscopic mapping with the NOGA system via femoral artery access and retrograde aortic approach using a 7‐Fr NOGA‐Star catheter. An area of interest located by technetium‐99m tetrofosmin SPECT was delineated in detail by means of NOGA mapping, including ischaemic but viable myocardium. Immediately before injection, the catheter was positioned perpendicularly to endocardium with excellent loop stability and the extension of the needle to induce premature ventricular contraction. 10 successive intramyocardial injections (roughly 0.2 mL each) were administered into the infarction border zone.
Dose of stem cells: 41 ± 16 x 106 BMSC, with 2.5 (1.6)% being CD34‐positive cells.
Timing of stem cell procedure: Within 24 hours after cell harvesting.

G‐CSF details: No G‐CSF administered.

Comparator arm: Control (no BM aspiration or placebo administration reported).

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes: Efficacy of the intramyocardial injection of autologous bone marrow mononuclear cells, measured by change in myocardial perfusion defects at rest and under pharmacological stress.
Secondary outcomes: Safety of the intramyocardial BMMNC therapy, quality of life, CCS angina class, NYHA functional class, LV functions, life‐threatening arrhythmias, mortality between 2 groups, NOGA change in voltage assessed by NOGA follow‐up endocardial mapping.
Outcome assessment points: Baseline, 6 and 12 months
Method(s) of outcome measurement: SPECT

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was carried out using an electronic system.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

No placebo was administered; participants and clinicians were not blinded.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

SPECT imaging was carried out though consensus by 2 readers blinded to the type of study and clinical data. Other blinding was not reported.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of all outcomes (other than deaths prior to follow‐up).

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

Santoso 2014

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: Supported by the S.K. Yee Medical Foundation Grant (208207); Research Grant Council of Hong Kong: General Research Fund (no. HKU 780110M); the Collaborative Research Fund (HKU 8/CRF/09); and Theme Based Research Scheme (T12‐705/11).

Study setting: Jakarta (Indonesia) and Hong Kong (China)
Number of centres: 2
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 19; Controls: 9
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 19; Controls: 9

Participants

Description: Advanced ischaemic HF (NYHA class III or IV; HF refractory to conventional medical therapy not eligible for conventional percutaneous or surgical revascularisation; existence of 1 or 2 coronary territories of viable, ischaemic myocardium as documented by dipyridamole single‐photon emission computed tomographic perfusion study; and LVEF < 40% measured by transthoracic echocardiogram).

Age distribution in each arm: BMSC: 58 (5.9) years; Controls: 60 (5.6) years
Sex (% male) in each arm: BMSC: 95%; Controls: 100%

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: At least 3 months since last MI.
Statistically significant baseline imbalances between the groups? None.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: On the day of the procedure, BMC were harvested from every participant by an experienced haematologist via posterior iliac crest puncture under local anaesthetic. A total of 80 mL to 100 mL of BM blood was aspirated, and an adequate trephine biopsy was performed. Mononuclear cells were isolated by Ficoll density gradient centrifugation. Bone marrow cells were washed twice in PBS, resuspended in PBS enriched with 10% autologous plasma to 100 × 106 mononuclear cells/mL, and returned directly to cardiac catheterisation laboratory for use. In the preparation for the control group, BM cells were not included in the final suspension, which comprised merely phosphate‐buffered saline with 10% autologous plasma. Bone marrow suspensions were tested by flow cytometry (Elite, Beckman Coulter, Fullerton, CA, USA) with directly conjugated antibodies. Cells administered by electromechanical mapping and endocardial injection (e.g. NOGA system).

Dose of stem cells: n/r
Timing of stem cell procedure: At least 3 to 4 hours after BM harvest.

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; PBS with 10% autologous plasma).

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes:

Change in LVEF from baseline to 6 months' follow‐up measured by MRI

Secondary outcomes:

Changes in exercise duration and MVO2 (treadmill modified Bruce protocol) (baseline and 6 months)

Note: main study publication reports secondary endpoints as changes in NYHA classification, LVESV, LV infarct and peri‐infarct ischaemic volume, and exercise performance (6MWT) (Santoso 2014).
Outcome assessment points: Baseline, 6 months; mean 23 (8) months (mortality only)
Method(s) of outcome measurement: MRI, NYHA, 6MWT

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation codes were generated using a randomisation table; randomisation was constrained, stratified by study centre.

Allocation concealment (selection bias)

Low risk

Randomisation was conducted via a system of sealed and numbered envelopes provided to each investigation centre.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

Bone marrow cells were harvested from all participants. The control group received an identical final suspension but without cells. After randomisation, participants were blinded to the study processes.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

After randomisation, investigators who were responsible for participant assessment were blinded to study processes.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of all outcomes on an intention‐to‐treat basis.

Selective reporting (reporting bias)

Unclear risk

All outcomes reported in the trial protocol (NCT01150175) were reported; additional outcomes were reported in the publication of results.

Other bias

Low risk

No other sources of bias were reported or identified.

Trifunovic 2015

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: n/r

Study setting: Belgrade, Serbia
Number of centres: 1
Length of follow‐up: median 5 years (IQR 2.5 to 7.5)
Number (N) of participants randomised to each arm: BMSC: 15; Controls: 15
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 15; Controls: 15

Participants

Description: IHD (aged 35 to 72 years; scheduled for CABG surgery due to LAD occlusion or multivessel coronary disease; previous MI older than 30 days; established diagnosis of ischaemic cardiomyopathy with LVEF < 40% and in the NYHA III‐IV functional class, full medical treatment for HF).
Age distribution in each arm: BMSC: 53.8 (10.1) years; Controls: 60 (6.8) years.
Sex (% male) in each arm: BMSC: 93.3%; Controls: 93.3%

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: Duration since last MI, BMSC: mean 3.2 (range 6 to 12) months; Controls: mean 3.07 (range 6 to 12) months

Statistically significant baseline imbalances between the groups? Significantly higher hypercholesterolaemia in BMSC group than in control group.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: Bone marrow was obtained by multiple aspirations from the posterior iliac crest in the amount of 150 mL, mixed with 25 mL of heparinised saline and transferred into a sterile bag. The BMMNC fraction was isolated by gradient centrifugation. All procedures from harvesting to cell injection were performed in a closed‐circuit system using sterile connection equipment with a sterile plastic bag system designated for cell transplantation in preoperative conditions. After finishing revascularisation with LIMA to LAD and sufficient number of autovenous aortocoronary bypass grafts to achieve total targeted revascularisation (either “on pump” or “off pump”, and if “on pump” when heart resumed its function from cardiopulmonary bypass), intramyocardial injection of BMMNC was carried out with a 1‐millilitre insulin syringe through a 27‐gauge needle. Bone marrow mononuclear cells injection was targeted into the hypocontractile peri‐infarcted viable myocardium visually identified and performed transepicardially in 30‐ to 45‐degree manner by multiple injections (17.5 (3.8) injections) injecting 0.2 mL to 0.5 mL in each injection to a final volume of 5.7 (1.5) mL.

Dose of stem cells: Mean 70.7 (32.4) x 106 cells
Timing of stem cell procedure: n/r

G‐CSF details: No G‐CSF administered.

Comparator arm: Control (no BM aspiration, no placebo).

Co‐intervention: CABG

Outcomes

Primary outcomes:

Postoperative functional capacity and cardiac‐related mortality in the median follow‐up of 5 years.
Secondary outcomes:

Cardiovascular mortality, NYHA, 6MWT, LVEF, perfusion defect; BNP levels.
Outcome assessment points: Baseline, 2 and 4 months, 1 year and subsequent years (clinical f/up); preoperatively and 6 months and at 2‐year intervals (SPECT) or at each follow‐up visit (echocardiography); baseline, 6 months and yearly (exercise capacity). Median follow‐up 5 years (range 2.5 to 7.5 years).

Method(s) of outcome measurement: SPECT, echocardiography, 6MWT

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The trial was described as randomised, but the method of randomisation was not reported.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

This was an open‐label trial; no blinding was carried out.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

The heart team consisting of an interventional cardiologist/radiologist, heart surgeon, and clinical cardiologist evaluated coronary angiography and all clinical and imaging data and made decisions on coronary revascularisation and stem cell implantation. This was an open‐label trial; no blinding was reported.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of all outcomes at follow‐up.

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

Tse 2007

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: This study was partially supported by the Sun Chieh Yeh Heart Foundation Fund; S K Ye Medical Foundation Grant (project no 203217), and The Research Grants Council of Hong Kong (HKU 7357/02M). Two authors received consultant fee from Biosense Webster, CA, USA. All other authors declare that they have no conflict of interest.

Study setting: Hong Kong (China) and Newcastle (Australia)
Number of centres: 2
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 19; Controls: 9
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 19; Controls: 9

Participants

Description: Refractory angina (CCS class III or IV; no conventional percutaneous or surgical revascularisation option; ability to complete > 3 min but < 10 min of treadmill exercise using modified Bruce protocol, and 1 or 2 coronary territories of viable, ischaemic myocardium as documented by dipyridamole SPECT perfusion study).

Age distribution in each arm: BMSC: 65.2 ± 8.3 years; Controls: 68.9 ± 6.3 years.
Sex (% male) in each arm: BMSC: 79%; Controls: 88%.

Number of diseased vessels: Not reported.
Time from symptom onset to initial treatment: Not reported.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: Bone marrow was harvested via posterior iliac crest puncture under local anaesthesia. A total of 40 mL of BM blood was aspirated, and an adequate trephine biopsy was performed. Bone marrow mononuclear cells were isolated by Ficoll density gradient centrifugation. Bone marrow cells were washed twice in phosphate‐buffered saline, resuspended in phosphate‐buffered saline enriched with 10% autologous plasma to either 1 or 2 x 107 MNC/mL and returned directly to cardiac catheterisation laboratory for use. Bone marrow suspensions were tested by flow cytometry with directly conjugated antibodies against CD34. Intramyocardial injection. Non‐fluoroscopic LV electromechanical mapping (NOGA) to identify the foci of ischaemic myocardium. During the procedure, systemic anticoagulation was achieved with intravenous heparin to maintain an activated clotting time of 250 to 300 s throughout the procedure. The targeted injection regions were selected by matching the area of ischaemic myocardium identified by SPECT. After completion of the LV electromechanical mapping, the mapping catheter was replaced by a modified mapping catheter incorporated with a 27G needle at the tip that could be used for direct endomyocardial injection.
Dose of stem cells: 1.5 x 107 BMMNC.
Timing of stem cell procedure: Within 3 to 4 hours from cell harvest.

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; 8 to 12 injections of 0.1 mL of phosphate buffered saline with 10% autologous serum).

Co‐intervention: CABG

Outcomes

Primary outcomes: Change from baseline in total exercise time on a modified Bruce protocol at 6 months' follow‐up.
Secondary outcomes: Changes in LVEF, NYHA, and CCS angina classification and sum of different scores on SPECT, global LVEF, LVEDV and LVESV by MRI.
Outcome assessment points: Baseline and 6 months
Method(s) of outcome measurement: SPECT and MRI

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was performed using a randomisation table and was constrained, stratified by the study centre.

Allocation concealment (selection bias)

Low risk

Sealed, numbered envelopes were provided by the study centre (centralised) to each investigational centre.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All randomised participants underwent BM aspiration, and the control group received a placebo injection. After randomisation, the study processes were blinded to participants. No details were given of the blinding of clinicians.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

After randomisation, the study processes were blinded to study co‐ordinators and investigators responsible for participants' assessment. Blinding was maintained until the end of the study.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of all outcomes on an intention‐to‐treat basis.

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

Turan 2011

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: Not reported

Study setting: Germany
Number of centres: 1
Length of follow‐up: 12 months
Number (N) of participants randomised to each arm: BMSC: 38; Controls: 18
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 33; Controls: 16

Participants

Description: IHD (aged 18 to 80 years; documented MI at least 3 months previously; clear‐cut demarcated region of left ventricular dysfunction with an open infarct‐related coronary artery at the time of stem cell therapy).

Age distribution in each arm: BMSC: 62 ± 10 years old; Controls: 60 ± 9 years old.
Sex (% male) in each arm: BMSC: 52.6%; Controls: 55.6%.

Number of diseased vessels: BMSC: 1.5 ± 0.5; Controls: 2.0 ± 0.6.
Time from symptom onset to initial treatment: Transmural MI 28 ± 14 months before treatment.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC.
Type of stem cells: Mononuclear cells.
Summary of stem cell isolation and type and route of delivery: 120 mL bone marrow was aspirated from the participant's own iliac crest, mononuclear cells were isolated using Harvest BMAC System (Germany) (most likely by density gradient centrifugation) and concentrated into 20 mL of cell suspension. Cell transplantation was performed via the coronary artery using 4 fractional infusions parallel to balloon inflation over 2 to 4 mins of 5 mL cell suspension. Cells were infused directly into the infarcted artery via an angioplasty balloon catheter that was inflated at a low pressure and was located within the previously stented coronary artery. Intracoronary infusion. This prevented back flow of cells and produced stop flow beyond the site of balloon inflation to facilitate high‐pressure infiltration of cells into the infarcted zone with prolonged contact time for cellular migration. After undergoing arterial puncture, all participants received 7500 to 10,000 units of heparin.
Dose of stem cells: 99 x 106 (± 25) mononuclear cells.
Timing of stem cell procedure: Within 24 hours from cell harvest.

G‐CSF details: No G‐CSF administered.

Comparator arm: Control (no BM aspiration; no placebo administered).

Co‐intervention: PCI

Outcomes

Primary outcomes: Change in global EF as well as the size of infarcted area measured by left ventriculography.
Secondary outcomes: Functional activity of BMSC immediately pre‐procedure and 3, 6, and 12 months after procedure; functional status assessed by NYHA classification and brain natriuretic peptide level in peripheral blood in both groups.
Outcome assessment points: Baseline, 3 and 12 months
Method(s) of outcome measurement: Left ventriculography

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Unclear risk

The trial was described as randomised, but the method of randomisation was not reported.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

High risk

Participants in the control group did not undergo BM aspiration, and no placebo was administered. Blinding was not reported.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Haemodynamic investigations and laboratory results were obtained independently by 2 investigators.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of mortality and morbidity outcomes; 7 participants (5x cell therapy, 2x controls) were excluded from LVEF and functional capacity at follow‐up due to restenosis.

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

Van Ramshorst 2009

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: This study is an academia‐initiated exploratory Phase II study. No external sponsor was involved in study design, data collection, data analysis, data interpretation, or writing of the report. No external funding was applicable for this study.

Study setting: Leiden, the Netherlands
Number of centres: 1
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 25; Controls: 25
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 25; Controls: 25

Participants

Description: Severe angina (CCS class II‐IV, myocardial ischaemia in at least 1 myocardial segment on Tc099m tetrofosmin SPECT, ineligible for CABG or PCI).

Age distribution in each arm: BMSC: 64 ± 8 years; Controls: 62 ± 9 years.
Sex (% male) in each arm: BMSC: 92%; Controls: 80%.

Number of diseased vessels: Not reported.
Time from symptom onset to initial treatment: At least 6 months from the last MI.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: Bone marrow was aspirated from the iliac crest under local anaesthesia and placed in a heparinised Hanks balanced salt solution. The MNC were isolated using Ficoll density gradient centrifugation, washed in phosphate‐buffered saline with 0.5% human serum albumin and resuspended in phosphate‐buffered saline with 0.5% human serum albumin. The final suspension of BMMNC contained 40 x 102 mL. The filtered bone marrow was checked for the presence of clots, and the BM cell population was analysed by fluorescence‐activated cell sorting using anti‐CD34 and anti‐CD35 antibodies. Intramyocardial injection. During cell isolation and randomisation, a 3D electromechanical map of the LV was obtained using the NOGA system. The ischaemic regions on SPECT were visually matched with the 3D electromechanical map based on anatomical landmarks including LV long axis, position of apex, mitral valve area, aortic valve location, and basal inferoseptal point. Cross‐referencing was also performed using fluoroscopic identification of anterior, septal, lateral, and inferior orientations.
Dose of stem cells: The cell suspension contained 98 ± 6 x 106 BM cells with a cell viability of 98% (1%) and a CD34+ cell fraction of 2.4% (0.9%).
Timing of stem cell procedure: Within 2 hours of BM aspiration.

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; sodium chloride 0.9% with 0.5% human serum albumin).

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes:

Change in myocardial perfusion (SPECT) at 3 months' follow‐up relative to baseline.
Secondary outcomes:

  1. Angina frequency

  2. CCS score

  3. Quality of life

  4. Exercise capacity

  5. Change in LVEF at 3 months

  6. Regional myocardial function on a segmental base at 3 months

  7. Occurrence of arrhythmias

  8. Pericardial effusion greater than 5 mm (echocardiography)

  9. Myocardial damage

  10. Severe inflammation

Outcome assessment points: Baseline and 6 months
Method(s) of outcome measurement: SPECT

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was carried out using sequentially numbered, sealed envelopes provided by the Department of Medical Statistics and Bioinformatics. A block size of 4 was used without further stratification.

Allocation concealment (selection bias)

Low risk

Sequentially numbered, sealed envelopes were used.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants underwent BM aspiration, and the control group received a placebo injection; participants were unaware of group assignment. A blinded syringe with either cell suspension or placebo was used.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Participants, study co‐ordinators, and investigators involved in participant assessments were unaware of group assignment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of all outcomes on an intention‐to‐treat basis.

Selective reporting (reporting bias)

Low risk

All outcomes reported in the trial protocol (ISRCTN58194927) were reported.

Other bias

Low risk

No other sources of bias were reported or identified.

Wang 2009

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: Not reported.

Study setting: Beijing, China
Number of centres: 1
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 16; Controls: 16
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 16; Controls: 16

Participants

Description: Angina (no AMI in the month prior to transplantation).

Age distribution in each arm: BMSC: 60.6 years; Controls: 60 years.

Sex (% male) in each arm: BMSC: 56.25%; Controls: 63.25%.

Number of diseased vessels: Not reported.
Time from symptom onset to initial treatment: At least 1 month from the last AMI.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC
Type of stem cells: CD34+ progenitor cells
Summary of stem cell isolation and type and route of delivery: 150 mL of BM was aspirated from the iliac crest. CD34+ cells were enriched by a cell separation device under GMP conditions. CD34+ cells were resuspended in normal saline and kept at room temperature. Cells were transported to the catheterisation lab. Cells were delivered using a microcatheter following PCI.
Dose of stem cells: 1.0 ‐ 6.1 x 106 CD34+ cells.
Timing of stem cell procedure: Unclear, not reported.

G‐CSF details: No G‐CSF administered.

Comparator arm: Control (no BM aspiration, no placebo administered).

Co‐intervention: PCI

Outcomes

Primary outcomes: Not reported.
Secondary outcomes: Myocardial perfusion defect area, wall motion, angina frequency change, nitrate triglycerine dose change, angina classification by CCS class.
Outcome assessment points: Baseline and 6 months.
Method(s) of outcome measurement: SPECT

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

This Chinese trial was described as randomised, but the method of randomisation was not reported.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

Blinding of clinicians and participants was not reported.

Blinding of outcome assessment (detection bias)
All outcomes

High risk

Outcome assessors were not blinded.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of all outcomes at follow‐up.

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

Wang 2010

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: Not reported.

Study setting: China
Number of centres: 1
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 56; Controls: 56
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 56; Controls: 56

Participants

Description: Intractable angina (aged > 30 years; diffuse triple‐vessel disease; CCS class III or IV; optimal medical therapy and considered ineligible for revascularisation, no ischaemia or nuclear perfusion imaging according to the Bruce protocol; angina experienced during baseline exercise test).
Age distribution in each arm: BMSC: 42 to 80 years; Controls: 43 to 80 years.
Sex (% male) in each arm: BMSC: 51.79%; Controls: 50%.

Number of diseased vessels: 3
Time from symptom onset to initial treatment: Not reported.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC
Type of stem cells: CD34+ progenitor cells
Summary of stem cell isolation and type and route of delivery: 120 mL to 150 mL bone marrow aspirates from the posterior iliac crest were obtained from all participants. CD34+ cells were isolated by labelling with the appropriate CD34 antibody and separating them magnetically using a CliniMACS (Miltenyi Biotec). CD34+ cells were resuspended in 15 mL of saline + human serum albumin. Only the saline + human serum albumin was infused in the control group, using the same protocol as in the BMSC group. The cells were infused into the coronary artery using a GE Innova 2000 DSA with 3000 units of heparin. Approximately 1 to 2 hours after cell separation, 10 mL of cells and 5 mL of saline were infused into the left coronary artery and right coronary artery separately by an over‐the‐wire balloon.
Dose of stem cells: 5.6 ± 2.3 x 107 CD34 cells.
Timing of stem cell procedure: Within 2 hours of cell harvest.

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; saline + human serum albumin)

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes: Safety (mortality and morbidities).
Secondary outcomes: Arrythmias, angina frequency, nitroglycerine use, exercise tolerance, CCS class, perfusion effect or myocardial perfusion.
Outcome assessment points: Baseline and 6 months
Method(s) of outcome measurement: Treadmill test, CCS class

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

This Chinese trial was described as randomised, but the method of randomisation was not reported.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants underwent BM aspiration, and the control group received a placebo injection. Participants were unaware of the treatment received. Blinding of clinicians was not reported.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All researchers were unaware of the treatments.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of all outcomes at follow‐up.

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

Wang 2014

Methods

Type of study: Parallel RCT

Type of publication: Conference abstract
Source of funding: Not reported.

Study setting: Guangzhou, China
Number of centres: 1
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 35; Controls: 35
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 35; Controls: 35

Participants

Description: Chronic IHD (impaired LV function: LVEF < 35%).
Age distribution in each arm: n/r
Sex (% male) in each arm: n/r

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: n/r
Statistically significant baseline imbalances between the groups? None reported.

Interventions

Intervention arm: BMSC
Type of stem cells: CD133+ progenitor cells
Summary of stem cell isolation and type and route of delivery: Injection of cells into the non‐transmural hypokinetic infract border zone. No further details reported.

Dose of stem cells: n/r
Timing of stem cell procedure: n/r

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration not reported; no further details)

Co‐intervention: Standard medical therapy

Outcomes

Primary outcomes:

LVEF measured by cMRI.
Secondary outcomes:

5‐min walk test; NYHA, regional wall motion, scar mass, LVESV, LVEDV.
Outcome assessment points: Baseline, 6 months
Method(s) of outcome measurement: MRI, NYHA class, 5‐min walk test

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

This Chinese trial was described as randomised, but the method of randomisation was not reported.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

Unclear risk

The trial was reported as "double‐blind", but no details of blinding were reported.

Blinding of outcome assessment (detection bias)
All outcomes

Unclear risk

The trial was reported as "double‐blind", but no details of blinding were reported.

Incomplete outcome data (attrition bias)
All outcomes

Unclear risk

No withdrawals were reported in this conference abstract.

Selective reporting (reporting bias)

High risk

All outcomes mentioned in the methods were reported in the results, although selective reporting would be considered likely in this conference abstract. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

Wang 2015

Methods

Type of study: Parallel RCT

Type of publication: Full paper
Source of funding: Supported by funding from the Major Project of Clinical Advanced Technology from PLA (2010gxjs002 to H.W.) and funded in part by the National Natural Science Foundation of China (30960379 to J.H.)

Study setting: Shenyang, China
Number of centres: 1
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 45; Controls: 45
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 45; Controls: 45

Participants

Description: Chronic MI (multivessel disease; admitted for elective OPCAB surgery at least 4 weeks after a cardiac infarction).
Age distribution in each arm: BMSC: 61.4 (7.45) years; Controls: 62.9 (6.93) years.
Sex (% male) in each arm: BMSC: 82%; Controls: 78%.

Number of diseased vessels: n/r (multivessel)
Time from symptom onset to initial treatment: Duration since last MI, mean 18.2 (16.8) months (BMSC) or 20.1 (19.0) months (Controls).
Statistically significant baseline imbalances between the groups? None.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: Bone marrow aspiration (100 mL) took place from the sternum of all participants. Bone marrow mononuclear cells were isolated by gradient centrifugation using Lymphoprep, washed twice with saline, and resuspended in 2 mL of heparinised saline. The 2 mL suspension or an equivalent volume of saline was injected with 8 punctures from a 22‐gauge Myjector syringe at the border zone of the infarct scar after finishing the revascularisation of the infarct‐related area. In cases where the infarct border could not be visualised, the cells were injected in an area of myocardium that corresponded to the perfusion defect on SPECT or scintigraphy. The injections were made parallel to the epicardium to avoid leakage of cells or delivery into the ventricular cavity, and depth was controlled with a plate stabiliser.

Dose of stem cells: mean 5.21 (0.44) x 108 cells
Timing of stem cell procedure: n/r

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration, saline solution)

Co‐intervention: CABG

Outcomes

Primary outcomes:

Incidence of emergent adverse events within the follow‐up period (6 months).
Secondary outcomes:

LVEF, wall motion score index, LVEDV/LVESV, LVEDD/LVESD, arrhythmias, atrial fibrillation, non‐sustained ventricular tachycardia, ventricular premature beats, sustained ventricular tachycardia, troponin T levels.
Outcome assessment points: Baseline, 6 months
Method(s) of outcome measurement: Echocardiography

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

This Chinese trial was described as randomised, but the method of randomisation was not reported.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants underwent BM aspiration and received cells or a placebo injection. The surgeon performing the OPCAB and injections was unaware of whether the light‐resistant syringe contained saline or BMC.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

All preoperative baseline and follow‐up echocardiography was performed by an experienced cardiologist blinded to treatment assignment.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of all outcomes at follow‐up.

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

Yao 2008

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: This work was supported by Shanghai Scientific Research Fund (06DJ14001), Program for Shanghai Outstanding Medical Academic Leader (LJ06008), and National Key Program (2006CB943704).

Study setting: Shanghai, China
Number of centres: 1
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 24; Controls: 23
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 24; Controls: 23

Participants

Description: IHD (aged < 75 years; history of transmural MI and revascularisation plus stent implantation at least 6 months earlier; patent infarct‐related artery at the time of stem cell transplantation).
Age distribution in each arm: BMSC: 54.8 ± 11.5 years; Controls: 56.3 ± 7.9 years.
Sex (% male) in each arm: BMSC: 96%; Controls: 96%.

Number of diseased vessels: BMSC: 1 (67%), 2 (29%), 3 (4%); Controls: 1 (70%), 2 (26%), 3 (4%).
Time from symptom onset to initial treatment: At least 6 months from last MI. 13 ± 8 months before entry into study.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: Bone marrow (95 (20) mL) was collected under local anaesthesia from the posterior superior iliac spine. Bone marrow cells were isolated and enriched with the use of Ficoll‐Hypaque gradient centrifugation procedures. Bone marrow aspirates were diluted with 0.9% sodium chloride (1:5), and mononuclear cells were isolated by density gradient centrifugation using Ficoll (800 g x 25 mins). Mononuclear cells were washed (800 g x 5 mins) 3 times with phosphate‐buffered saline and then resuspended in 16 mL of heparin‐treated plasma at a density of 2.4 (1.2 x 107) cells/mL at room temperature. Before intracoronary injection, the mononuclear cells were filtered (Falcon) and counted. These cells were used for therapy. To ensure that a certain % of stem cells was present in the infused MNC, a 1 mL suspension was subjected to FACS analysis after incubation with anti‐human monoclonal antibodies: anti‐human CD34 conjugated with FITC, or CD133 antibodies conjugated with APC. The FACS analysis revealed that 2.4% (0.9%) of BMC was positive for CD34, and 0.75% (0.2%) was positive for CD133. Intracoronary infusion. An over‐the‐wire angioplasty balloon catheter was inserted into the stent previously implanted during the acute reperfusion procedure. The balloon was inflated with low pressure (2 atm to 4 atm) to completely block blood flow for 2 mins; this was repeated 5 times. During each balloon inflation, 3 mL of BMC suspensions was infused distal to the occluding balloon into the infarct‐related artery.
Dose of stem cells: 7.2 x 107 cells.
Timing of stem cell procedure: Within 6 hours after bone marrow puncture.

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (0.9% sodium chloride containing heparin). Unclear whether BM aspiration was performed.

Co‐intervention: Standard medical therapy, PCI (in 30% of participants)

Outcomes

Primary outcomes: Improvement of LV function.
Secondary outcomes: LVEF, LVED diameter, LVES diameter (echocardiography).
LVEF, LVESV, LVEDV, infarct size (MRI).
Myocardial perfusion (SPECT); mortality and morbidities.
Outcome assessment points: Baseline and 6 months
Method(s) of outcome measurement: Echocardiography, MRI, and SPECT

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

High risk

This Chinese trial was described as randomised, but the method of randomisation was not reported.

Allocation concealment (selection bias)

Unclear risk

Allocation concealment was not reported.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants underwent BM aspiration, and the control group received a placebo injection. Blinding of clinicians was not reported.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

Outcome assessors (MRI, echocardiography, SPECT) were blinded to the assigned therapy.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of all outcomes at follow‐up.

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

Zhao 2008

Methods

Type of study: Parallel RCT
Type of publication: Full paper
Source of funding: Shanghai Medical Development Research Fund, Grant Number 2000I‐2D002

Study setting: Shanghai, China
Number of centres: 1
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: BMSC: 18; Controls: 18
Number (N) of participants analysed (primary outcome) in each arm: BMSC: 18; Controls: 18

Participants

Description: Ischaemic HF (aged 18 to 75 years; admitted for elective CABG; history of transmural old MI with akinesis or dyskinesis of the left ventricle shown by echocardiography; multivessel disease with a reversible perfusion defect detected by SPECT; LVEF less than 40%).

Age distribution in each arm: BMSC: 60.3 ± 10.4 years; Controls: 59.1 ± 15.7 years.
Sex (% male) in each arm: BMSC: 83.3%; Controls: 83.3%.

Number of diseased vessels: multivessel, 2 or more
Time from symptom onset to initial treatment: Not reported.
Statistically significant baseline imbalances between the groups? No.

Interventions

Intervention arm: BMSC
Type of stem cells: Mononuclear cells
Summary of stem cell isolation and type and route of delivery: After heparinisation and median sternotomy, BM (about 30 mL) was aspirated from the sternum by a special suction appliance in both groups. The BMMNC were immediately isolated by density gradient centrifugation using Ficoll. Isolated cells were washed twice with heparinised saline and subsequently resuspended in 5 mL saline. The cells were counted and the viability was assessed by trypan blue dye exclusion. The cell suspension was filtered by a 70‐micron cell strainer before transplantation. During CABG, intramyocardial injection in and around the infarct area at 10 points (approximately 0.5 mL per injection) with a 29‐gauge syringe.
Dose of stem cells: 6.59 ± 5.12 x 108 (cell viability 96.48% ± 3.10%).
Timing of stem cell procedure: Within 24 hours following cell harvest.

G‐CSF details: No G‐CSF administered.

Comparator arm: Placebo (BM aspiration; saline).

Co‐intervention: CABG

Outcomes

Primary outcomes: Death, MI, and recurrence of HF.
Secondary outcomes: Echo: infarction wall thickness; infarction wall motion velocity; LVEDD/LVESD; global LVEF; LV shortening fraction; mitral valve regurgitation.
SPECT: LV SRS; infarcted area SRS; clinical parameters; NYHA, CCS classification; 24‐hour Holter analysis.
Outcome assessment points: Baseline and 6 months
Method(s) of outcome measurement: Echocardiography and SPECT

Notes

Risk of bias

Bias

Authors' judgement

Support for judgement

Random sequence generation (selection bias)

Low risk

Randomisation was achieved using a computer‐generated sequence of random numbers.

Allocation concealment (selection bias)

Unclear risk

No method of allocation concealment was reported.

Blinding of participants and personnel (performance bias)
All outcomes

Low risk

All participants underwent BM aspiration, and the control group received a placebo injection. Blinding of clinicians was not reported.

Blinding of outcome assessment (detection bias)
All outcomes

Low risk

The results were analysed by 2 independent, experienced observers; investigators (echocardiography, SPECT) were blinded to the randomisation scheme.

Incomplete outcome data (attrition bias)
All outcomes

Low risk

All randomised participants were included in the analysis of all outcomes at follow‐up (other than deaths).

Selective reporting (reporting bias)

Unclear risk

All outcomes mentioned in the methods were reported in the results, although it would be difficult to rule out selective reporting. No prospectively registered or published trial protocol was identified.

Other bias

Low risk

No other sources of bias were reported or identified.

6MWT: 6‐minute walk test
AEs: adverse events
ALDH: aldehyde dehydrogenase
AMI: acute myocardial infarction
AOI: area of interest
APC: allophycocyanin
BM: bone marrow
BMC: bone marrow cells
BMMNC: bone marrow mononuclear cells
BMSC: bone marrow stem cells
BNP: brain natriuretic peptide
CABG: coronary artery bypass grafting
CAD: coronary artery disease
CCS: Canadian Cardiovascular Society
CCTRN: Cardiovascular Cell Therapy Research Network
CFU‐GM: colony forming unit‐granulocyte macrophage
CHF: congestive heart failure
cMRI: cardiovascular magnetic resonance imaging
CPC: circulating progenitor cells
CT: computed tomography
DM: diabetes mellitus
ECG: electrocardiogram
EDTA: ethylenediaminetetraacetic acid
EF: ejection fraction
EMM: electromechanical mapping
EPC: endothelial progenitor cells
FACS: fluorescence‐activated cell sorting
FITC: fluorescein isothiocyanate
G‐CSF: granulocyte colony‐stimulating factor
GMP: good manufacturing practices
HF: heart failure
HTN: hypertension
IC: intracoronarily
IgG: immunoglobulin G
IHD: ischaemic heart disease
IM: intramyocardial
IQR: interquartile range
LAD: left anterior descending
LDL: low‐density lipoprotein
LIMA: left internal mammary artery
LV: left ventricular
LVEDD: left ventricular end‐diastolic diameter
LVEDV: left ventricular end‐diastolic volume
LVEF: left ventricular ejection fraction
LVESD: left ventricular end‐systolic diameter
LVESV: left ventricular end‐systolic volume
MACE: major adverse clinical events
MI: myocardial infarction
MLHFQ: Minnesota Living with Heart Failure Questionnaire
MNC: mononuclear cells
MRI: magnetic resonance imaging
MSC: mesenchymal stem cells
MVO2: myocardial oxygen consumption
NHLBI: National Heart, Lung, and Blood Institute
n/r: not reported
NTG: nitroglycerine
NT‐proBNP: N‐terminal pro b‐type natriuretic peptide
NYHA: New York Heart Association
OPCAB: off‐pump coronary artery bypass
PBS: phosphate‐buffered saline
PBSC: peripheral blood stem cell
PC: progenitor cells
PCI: percutaneous coronary intervention
PET: positron emission tomography
QOL: quality of life
RCT: randomised controlled trial
SD: standard deviation
SE: standard error
SF‐36: 13‐Item Short Form Health Survey
SPECT: single‐photon emission computed tomography
SRS: segmental resting score
SV: stroke volume
STEMI: ST elevation myocardial infarction
TIMI: Thrombolysis In Myocardial Infarction
VEGF: vascular endothelial growth factor

Characteristics of excluded studies [ordered by study ID]

Study

Reason for exclusion

Ascheim 2014

RCT of allogeneic mesenchymal precursor cells.

Assmann 2014

Single‐arm trial of CD133+ in ischaemic cardiomyopathy, no control arm included.

Beeres 2006

Single‐arm substudy of BMMNC in refractory angina and chronic myocardial ischaemia, no control arm included.

Beeres 2007

Single‐arm trial of BMMNC in chronic ischaemia, no control arm included.

Beeres 2007a

Single‐arm trial of BMMNC in chronic myocardial infarction and severe left ventricular dysfunction, no control arm included.

Beeres 2007b

Review of imaging techniques for cardiac stem cell therapy.

Bittencourt 2008

Single‐arm trial of BMSC in severe coronary artery disease, no control arm included.

Bolli 2011

RCT of cardiac stem cells in ischaemic cardiomyopathy, no BMSC administered.

Chang 2006

RCT of peripheral blood stem cells in AMI.

Charwat 2010

Randomised trial of early versus late administration of BMMNC in AMI.

Chen 2014

RCT of G‐CSF‐mobilised PBSC in heart failure; G‐CSF was not administered to the control group.

Chin 2010

Single‐arm trial of autologous MSC in end‐stage dilated cardiomyopathy.

EUCTR2006‐005628‐17‐ES

RCT of BMMNC in people with AMI.

EUCTR2009‐017924‐18‐NL

A follow‐on study of people with refractory angina and documented ischaemia who received bone marrow‐derived cells in 2 previous trials, no control arm included.

Fuchs 2004

Single‐arm trial of bone marrow cells in advanced ischaemic heart disease, no control arm included.

Gu 2011

Non‐randomised trial of single or repeated infusion of PBSC and G‐CSF compared with a control group in refractory ischaemic heart failure.

Haack‐Sorensen 2013

A single‐arm trial of autologous MSC in stable coronary artery disease and refractory angina, no control arm included.

Jimenez‐Quevedo 2008

A comparison of outcomes in diabetic/non‐diabetic patients with end‐stage heart failure who received BMMNC in a previous trial.

Kang 2006

RCT of G‐CSF‐mobilised PBSC in people with acute and old myocardial infarction; G‐CSF was not administered to the control group.

Kang 2006b

RCT of G‐CSF‐mobilised PBSC in people with acute myocardial infarction.

Karantalis 2014

RCT of autologous MSC in people undergoing CABG; trial suspended due to low accrual and no control participants included.

Koestering 2005

Non‐randomised trial of BMMNC in chronic coronary artery disease.

Lai 2009

RCT of autologous MSC in people undergoing CABG; study of cardiac enzyme outcomes measured within 24 to 48 hours of treatment, which are not relevant to this review. No further publications have been identified.

Lee 2015

RCT of CD34+ in end‐stage diffuse coronary artery disease comparing 2 cell doses, no control arm included.

Makkar 2011

RCT of cardiosphere‐derived cells in AMI.

Mann 2015

A follow‐on single‐arm substudy of 3 previous cell therapy trials, no control group included.

Maroto 2010

RCT of BMMNC in sub‐acute myocardial infarction (within 15 days).

Maureira 2012

Although this study was described as randomised, the 7 participants in each treatment arm (14 in total) were matched by age and sex.

Mocini 2006

Non‐randomised study of BMSC in AMI.

Nagaya 2007

Non‐randomised study of BMMSC for severe chronic heart failure.

NCT00285454

Study withdrawn prior to enrolment.

NCT00289822

Trial terminated (reason not given), no relevant publications identified.

NCT00362388

Trial terminated (reason not given), no relevant publications identified.

NCT01074099

Trial terminated due to pilot study resulting in changes to protocol and new study required.

NCT01337011

RCT of intramyocardial versus intracoronary administration of enriched CD133+ cells, no control arm included.

NCT01666132

Trial terminated after phase I due to slow recruitment.

NCT01693042

RCT of single versus repeated administration of BMMNC in chronic postinfarction HF, no control arm included.

NCT01721902

Trial terminated due to lack of recruitment, no relevant publications identified.

Perin 2003

Non‐randomised controlled trial of BMMNC in chronic ischaemic heart failure.

Peruga 2009

Non‐randomised trial of BMMNC in AMI.

Poglajen 2013

Single‐arm trial of CD34+ cell in ischaemic cardiomyopathy.

Pokushalov 2011

Randomised cross‐over trial of cardiac resynchronisation and BMMNC in ischaemic heart failure, no control arm included.

Premer 2014

Randomised trial of autologous versus allogeneic MSC in dilated cardiomyopathy.

Qin 2015

RCT of cardiosphere‐derived cells for heart regeneration after myocardial infarction; no bone marrow‐derived cells were administered.

Rivas‐Plata 2010

Non‐RCT of BMMNC in people with heart failure.

Shen 2007

Pre‐clinical animal study of BMMSC after AMI.

Stamm 2007a

Non‐randomised trial of CD133+ cells in chronic ischaemic heart disease.

Suncion 2014

A single‐arm substudy of the POSEIDON (Prevention of Contrast Renal Injury with Different Hydration Strategies) trial, no control arm.

Takehara 2012

A single‐arm trial of autologous human cardiac‐derived stem cells in ischaemic cardiomyopathy.

Tuma 2010

A comparison of outcomes in people with ischaemic and non‐ischaemic heart failure who received CD34+ and MSC.

Tuma 2011

A single‐arm trial of BMMNC in refractory angina.

Vicario 2004

A single‐arm trial of autologous unfractionated bone marrow in refractory angina.

Vrtovec 2015

A comparison of the effects of CD34+ cell therapy in ischaemic and non‐ischaemic HF, no control arm included.

Wang 2006

Non‐randomised trial of BMMNC in AMI.

AMI: acute myocardial infarction
BMMNC: bone marrow mononuclear cells
BMMSC: bone marrow mesenchymal stem cells
BMSC: bone marrow stem cells
CABG: coronary artery bypass grafting
G‐CSF: granulocyte colony‐stimulating factor
MSC: mesenchymal stem cells
PBSC: peripheral blood stem cell
RCT: randomised controlled trial

Characteristics of studies awaiting assessment [ordered by study ID]

Ahmadi 2010

Methods

Type of study: Parallel RCT

Type of publication: Abstract
Source of funding: n/r

Study setting: Tehran, Iran
Number of centres: n/r
Length of follow‐up: 6 months
Number (N) of participants randomised to each arm: n/r
Number (N) of participants analysed (primary outcome) in each arm: 10 (cell therapy), 10 (controls)

Participants

Description: Ischaemic cardiomyopathy and low global ejection fraction (< 35%)
Age distribution in each arm: n/r
Sex (% male) in each arm: n/r

Number of diseased vessels: n/r
Time from symptom onset to initial treatment: n/r
Statistically significant baseline imbalances between the groups? n/r

Interventions

Intervention arm: CABG + intramyocardial mesenchymal/CD133+ stem cells
Type of cells: Intramyocardial mesenchymal/CD133+ stem cells

Dose of cells: n/r

Timing of stem cell procedure: Bone marrow was harvested from iliac crest 2 weeks prior to surgery, and purified expanded mesenchymal/CD133+ stem/progenitor cells were injected in the ischaemic border zone of the heart during CABG.

Comparator arm: CABG only

Outcomes

LV function, wall motion score index, mortality, morbidities

Outcome assessment points: Baseline, 6 months

Method of measurement: Echocardiography

Notes

To our knowledge, this trial has published in abstract form only with insufficient data reporting for inclusion. Should further publications be identified, this study will be incorporated into future updates to this review.

Ahmadi 2015

Methods

Type of study: Parallel RCT

Type of publication: Abstract

Source of funding: n/r

Study setting: n/r

Number of centres: n/r

Length of follow‐up: 35 months

Number (N) of participants randomised to each arm: n/r (total 27)
Number (N) of participants analysed (primary outcome) in each arm: n/r

Participants

Description: Severe ischaemic HF undergoing CABG

Age distribution in each arm: n/r
Sex (% male) in each arm: n/r

Number of diseased vessels: n/r

Time from symptom onset to initial treatment: n/r

Interventions

Intervention arm 1: CABG + BMMSC

Intervention arm 2: CABG + recycling stem cells

Type of cells: BMMNC

Dose of cells: n/r

Timing of stem cell procedure: Cells were injected in the border zone of the infarcted myocardium.

Comparator arm: CABG

Outcomes

Morbidy, mortality, LVEF wall motion score index.

Outcome assessment points: Baseline, 36 months

Method of measurement: Echocardiography

Notes

To our knowledge, this trial has published in abstract form only with insufficient data reporting for inclusion. Should further publications be identified, this study will be incorporated into future updates to this review.

Cuzzola 2007

Methods

Type of study: Parallel RCT

Type of publication: Abstract

Source of funding: n/r

Study setting: Italy

Number of centres: n/r

Length of follow‐up: 12 months

Number (N) of participants randomised to each arm: n/r (total 37 randomised 1:1)
Number (N) of participants analysed (primary outcome) in each arm: n/r (total 16)

Participants

Description: Ischaemic cardiomyopathy (acute transmural MI less than 6 months prior to admission and LVEF lower than 35%).

Age distribution in each arm: n/r
Sex (% male) in each arm: n/r

Number of diseased vessels: n/r

Time from symptom onset to initial treatment: < 6 months prior to admission

Interventions

Intervention arm: CABG + BMMNC

Type of cells: BMMNC

Dose of cells: n/r

Timing of stem cell procedure: Bone marrow mononuclear cells were isolated from bone marrow aspirates and injected intramyocardially during cardiac surgery (CABG).

Comparator arm: CABG + placebo

Outcomes

Periprocedural adverse events, mortality, LVEF and LV volumes; flow cytometry measures

Outcome assessment points: 6 and 12 months

Method of measurement: Not reported.

Notes

To our knowledge, this trial has published in abstract form only with insufficient data reporting for inclusion. Should further publications be identified, this study will be incorporated into future updates to this review.

Grynberg 2008

Methods

Type of study: Parallel RCT

Type of publication: Abstract

Source of funding: n/r

Study setting: Buenos Aires, Argentina

Number of centres: n/r

Length of follow‐up: 12 months

Number (N) of participants randomised to each arm: n/r
Number (N) of participants analysed (primary outcome) in each arm: 7 cell therapy, 8 controls

Participants

Description: Chronic MI with patent infarct‐related artery, extensive necrosis, no development of ischaemia, and impaired ventricular function.

Age distribution in each arm: n/r
Sex (% male) in each arm: n/r

Number of diseased vessels: n/r

Time from symptom onset to initial treatment: < 3 months

Interventions

Intervention arm: BMSC

Type of cells CD34+ and CD133+ cells

Dose of cells: 4 ‐ 10 x 106

Timing of stem cell procedure: Bone marrow aspiration, with filter and cell processing to obtain CD34+ and CD133+, and latter intracoronary injection of this preparation through the infarct‐related artery.

Comparator arm: Control (optimal medical therapy)

Outcomes

Major cardiovascular events, rehospitalisation, target vessel revascularisation, LVEF, myocardial perfusion (summed rest score)

Outcome assessment points: Baseline, 12 months

Method of measurement: Radioisotopic ventriculography

Notes

Participants are from the chronic branch of the "RECUPERAR" study. To our knowledge, this trial has published in abstract form only with insufficient reporting of methodology to determine whether this substudy is randomised. Should further publications be identified confirming this, the study will be incorporated into future updates to this review.

Jie 2014

Methods

Type of study: Parallel RCT

Type of publication: Abstract

Source of funding: n/r

Study setting: China

Number of centres: n/r

Length of follow‐up: 12 months

Number (N) of participants randomised to each arm: n/r (110 in total)
Number (N) of participants analysed (primary outcome) in each arm: 42x cell therapy (assumed 68 controls)

Participants

Description: Ischaemic HF with LVEF < 45%

Age distribution in each arm: n/r
Sex (% male) in each arm: n/r

Number of diseased vessels: n/r

Time from symptom onset to initial treatment: n/r

Interventions

Intervention arm: BMSC

Type of cells: Mononuclear cells

Dose of cells: 8.6 x 108

Timing of stem cell procedure: Cells or placebo were injected intraoperatively into the MI border area.

Comparator arm: Placebo (no details)

Outcomes

Myocardial scar size, LVEF, cell viability, wall thickening

Outcome assessment points: Baseline, 12 months

Method of measurement: MRI, PET

Notes

To our knowledge, this trial has published in abstract form only with insufficient data reporting for inclusion. Should further publications be identified, this study will be incorporated into future updates to this review.

Kakuchaya 2011

Methods

Type of study: Parallel RCT

Type of publication: Abstract

Source of funding: n/r

Study setting: Moscow, Russia

Number of centres: n/r

Length of follow‐up: 6 months

Number (N) of participants randomised to each arm: n/r
Number (N) of participants analysed (primary outcome) in each arm: n/r (total 50 participants)

Participants

Description: Chronic HF patients in NYHA class III‐IV (24 participants with ischaemic dilated cardiomyopathy and 26 participants with idiopathic dilated cardiomyopathy)

Age distribution in each arm: n/r
Sex (% male) in each arm: n/r

Number of diseased vessels: n/r

Time from symptom onset to initial treatment: n/r

Interventions

Intervention arm: BMSC

Type of cells: CD133+

Dose of cells: n/r

Timing of stem cell procedure: CD133+ were obtained by CliniMACS technology of magnetic separation.

Comparator arm: Placebo

Outcomes

LVEF, LV volumes, LV mass

Outcome assessment points: Baseline, 6 months

Method of measurement: SPECT

Notes

To our knowledge, this trial has published in abstract form only with insufficient data reporting for inclusion. Should further publications be identified, this study will be incorporated into future updates to this review.

Minjie 2011

Methods

Type of study: Parallel RCT

Type of publication: Abstract

Source of funding: n/r

Study setting: Beijing, China

Number of centres: n/r

Length of follow‐up: 12 months

Number (N) of participants randomised to each arm: n/r
Number (N) of participants analysed (primary outcome) in each arm: n/r (50 in total)

Participants

Description: Old MI

Age distribution in each arm: total: 57.48 ± 7.98 years
Sex (% male) in each arm: total: 94%

Number of diseased vessels: n/r

Time from symptom onset to initial treatment: n/r

Interventions

Intervention arm: CABG + BMSC

Type of cells: BMMNC

Dose of cells: n/r

Timing of stem cell procedure: No details of cell isolation or cell delivery method reported.

Comparator arm: CABG + placebo

Outcomes

LVEF, LV volumes, cardiac output, cardiac index, cardiac mass, infarct size

Outcome assessment points: Baseline, 12 months

Method of measurement: MRI

Notes

To our knowledge, this trial has published in abstract form only with insufficient data reporting for inclusion. Should further publications be identified, this study will be incorporated into future updates to this review.

Pourrajab 2013

Methods

Type of study: Controlled trial (unclear whether randomised)

Type of publication: Abstract

Source of funding: n/r

Study setting: Yazd, Iran

Number of centres: n/r

Length of follow‐up: n/r

Number (N) of participants randomised to each arm: n/r (15 recruited in total)
Number (N) of participants analysed (primary outcome) in each arm: n/r

Participants

Description: Severe ischaemic cardiomyopathy requiring CABG

Age distribution in each arm: n/r
Sex (% male) in each arm: n/r

Number of diseased vessels: n/r

Time from symptom onset to initial treatment: n/r

Interventions

Intervention arm: CABG + BMSC

Type of cells: BMSC

Dose of cells: median 5 x 107 (SD 1 x 106) cells

Timing of stem cell procedure: No details given.

Comparator arm: CABG + placebo

Outcomes

Periprocedural adverse events, angina frequency, LVEF, LV volumes, wall motion

Outcome assessment points: n/r

Method of measurement: n/r

Notes

To our knowledge, this trial has published in abstract form only with insufficient data reporting for inclusion. Should further publications be identified, this study will be incorporated into future updates to this review.

Stefanelli 2015

Methods

Type of study: Parallel RCT

Type of publication: Abstract

Source of funding: n/r

Study setting: Modena, Italy

Number of centres: n/r

Length of follow‐up: 12 months

Number (N) of participants randomised to each arm: 19x BMSC, 11x controls
Number (N) of participants analysed (primary outcome) in each arm: 10x BMSC, controls n/r

Participants

Description: Ischaemic HF

Age distribution in each arm: n/r
Sex (% male) in each arm: n/r

Number of diseased vessels: n/r

Time from symptom onset to initial treatment: n/r

Interventions

Intervention arm: LV restoration + BMSC

Type of cells: BMMNC

Dose of cells: 5 cm3 to 8 cm3

Timing of stem cell procedure: Mononuclear cells were derived from the sternal bone marrow and processed in a sterile mini‐lab before injection by direct visualisation into the infarcted areas of the myocardium before LV reconstruction.

Comparator arm: LV restoration only

Outcomes

Mortality, rehospitalisation, change in LVEF diameter, change in LVEF and NYHA, change in infarcted area

Outcome assessment points: Baseline, 6 and 12 months

Method of measurement: Echocardiography, PET

Notes

To our knowledge, this trial has published in abstract form only with insufficient data reporting for inclusion. Should further publications be identified, this study will be incorporated into future updates to this review.

Zverev 2006

Methods

Type of study: Parallel RCT

Type of publication: Abstract

Source of funding: n/r

Study setting: St Petersburg, Russia

Number of centres: n/r

Length of follow‐up: 12 months

Number (N) of participants randomised to each arm: 18x BMMSC, 38x BMMNC, 13x controls
Number (N) of participants analysed (primary outcome) in each arm: 8x BMMSC, 38x BMMNC, 13x controls

Participants

Description: Non‐acute IHD

Age distribution in each arm: n/r
Sex (% male) in each arm: n/r

Number of diseased vessels: n/r

Time from symptom onset to initial treatment: n/r

Interventions

Intervention arm: BMMSC or BMMNC

Type of cells: BMMSC or BMMNC

Dose of cells: 12 x 108 cells

Timing of stem cell procedure: Mesenchymal stem cells were isolated from bone marrow and cultured in vitro. Bone marrow mononuclear cells were also isolated from bone marrow. Cells were delivered via the coronary artery.

Comparator arm: Placebo

Outcomes

Angina episodes, nitroglycerine consumption, myocardial viability and perfusion, LVEF

Outcome assessment points: Baseline, 6, 9, and 12 months

Method of measurement: PET, SPECT, echocardiography

Notes

To our knowledge, this trial has published in abstract form only with insufficient data reporting for inclusion. Should further publications be identified, this study will be incorporated into future updates to this review.

AMI: acute myocardial infarction
BMMNC: bone marrow mononuclear cells
BMMSC: bone marrow mesenchymal stem cells
BMSC: bone marrow stem cells
BNP: brain natriuretic peptide
CABG: coronary artery bypass grafting
CCS: Canadian Cardiovascular Society
CPC: circulating progenitor cells
EPC: endothelial progenitor cells
G‐CSF: granulocyte colony‐stimulating factor
HF: heart failure
IC: intracoronary
IM: intramyocardial
LV: left ventricular
LVEF: left ventricular ejection fraction
LVEDV: left ventricular end‐diastolic volume
LVESV: left ventricular end‐systolic volume
MACE: major adverse clinical events
MI: myocardial infarction
MRI: magnetic resonance imaging
MSC: mesenchymal stem cells
MVO2: myocardial oxygen consumption
NYHA: New York Heart Association
PCI: percutaneous coronary intervention
PET: positron emission tomography
RCT: randomised controlled trial
SD: standard deviation
SPECT: single‐photon emission computed tomography

Characteristics of ongoing studies [ordered by study ID]

EUCTR2009‐016364‐36‐NL

Trial name or title

Injection of autologous bone marrow cells into damaged myocardium of no‐option patients with ischaemic heart failure: a randomised placebo controlled trial ‐ cell therapy for ischaemic heart failure

Methods

A randomised, double‐blind, cross‐over, placebo‐controlled trial

Participants

Ischaemic HF:

  1. Ischaemic HF NYHA class III or IV despite optimal pharmacological and non‐pharmacological therapy.

  2. No candidate for (repeat) surgery (revascularisation, valve repair, or ventricular reconstruction).

  3. No candidate for (repeat) percutaneous revascularisation.

  4. Optimal resynchronisation therapy, or no candidate for resynchronisation therapy.

  5. Male or female > 18 and < 75 years old.

  6. Life expectancy more than 6 months.

  7. Able to perform an exercise tolerance test prior to therapy.

  8. Able and willing to undergo all the tests used in this protocol including the travelling involved.

  9. Written informed consent.

Interventions

Intervention arm: Intracardiac administration of bone marrow mononuclear cells

Comparator arm: Intracardiac administration of placebo

Outcomes

Primary outcomes:

  1. Left ventricular global ejection fraction as assessed by gated SPECT.

  2. LV regional wall motion by echocardiography.

  3. FDG‐SPECT for assessment of viability and hibernation.

  4. Myocardial innervation imaging (MIBG‐SPECT) for assessment of myocardial innervation.

  5. Exercise capacity by bicycle exercise testing with VO2 measurement.

  6. Quality of life assessed using the MLHFQ.

Secondary outcomes:

Safety (incidence of arrhythmias via Holter monitoring, inflammation and myocardial damage)

Starting date

July 2010

Contact information

None identified.

Notes

Planned enrolment: 64

Estimated completion date: The status of this trial is "completed", but no publications have been identified.

ISRCTN71717097

Trial name or title

Bone‐marrow derived stem cell transplantation in patients undergoing left ventricular restoration surgery for dilated ischaemic end‐stage heart failure: a randomised blinded controlled trial (TransACT 2)

Methods

A double‐blind, randomised, placebo‐controlled trial

Participants

End‐stage HF:

  1. Previous anterior MI (with evidence of large surgically excludible scar at cardiac MRI).

  2. Significant LV dilation (LVESV index greater than or equal to 60 mL/m2).

  3. LVEF less than or equal to 35%.

  4. NYHA class III/IV and 1 episode of CHF requiring medical attention.

  5. Elective left ventricular restoration surgery indicated.

  6. Elective CABG indicated to bypass stenoses or occlusions of coronary arteries.

  7. Participant aged ≥ 16 and < 80 years old, either sex.

Interventions

Intervention arm: Surgical ventricular restoration and transplantation of autologous CD133+

Comparator arm: Surgical ventricular restoration and injection of placebo, i.e. autologous plasma

Outcomes

Primary outcome:

Regional LV thickening of the 'affected' segments 6 months after surgery.

Secondary outcomes:

  1. Mid‐term generic and cardiac‐specific health status and quality of life, measured at baseline and 6 months' follow‐up.

  2. End‐systolic volume and stroke volume quantified by cardiac MRI, measured at baseline (3 to 5 days postoperatively), and 6 months' follow‐up.

  3. Myocardial injury throughout the duration of the study by measuring troponin I levels (24 hours preoperatively; surgery; 4, 12, 24 hours postoperatively; 6 weeks' and 6 months' follow‐up).

Starting date

August 2009

Contact information

University of Bristol, Bristol Royal Infirmary. Contact: Mr R Ascione ([email protected])

Notes

Planned enrolment: 40

Estimated completion date: The status of this trial is "completed", but no publications have been identified.

ISRCTN75217135

Trial name or title

A pilot study to evaluate the efficacy of combined transplantation of progenitor cells and coronary artery bypass grafting (TOPCABG)

Methods

RCT

Participants

Participants undergoing CABG

Interventions

Intervention arm: Stem cells (5 participants)

Comparator arm: Heparinised saline (5 participants)

Outcomes

Primary outcome: To show improvements in myocardial function, regional wall motion, and myocardial perfusion.

Starting date

January 2004

Contact information

Southampton University Hospitals NHS Trust, Level D, East Wing, Southampton General Hospital, Tremona Road, Southampton, UK SO16 6YD. Contact: Mr D Varghese ([email protected])

Notes

Planned enrolment: 10

Estimated completion date: The status of this trial is "completed", but no publications have been identified.

NCT00690209

Trial name or title

Bypass surgery with stem cell therapy in chronic ischemic cardiopathy

Methods

A phase II, parallel, randomised, single‐blind (participant) controlled study

Participants

IHD:

  1. Aged 18 to 75 years.

  2. Chronic IHD.

  3. LVESV > 140 mL.

  4. Poor global contractile function (LVEF < 40%).

  5. Substantial amount of residual viability (> 30% of left ventricle).

Interventions

Intervention arm: Surgical revascularisation associated with autologous bone marrow‐derived stem cells injection in viable territories.

Comparator arm: Surgical revascularisation alone.

Outcomes

Primary outcome: Evolution of left ventricular volumes and contractility.

Secondary outcome: Functional status.

Starting date

May 2008

Contact information

Departments of Cardiac Surgery, Cardiology and Radiology, University Hospital, Clermont‐Ferrand, France (Principal Investigator: Dr J Lipiecki. Contact: Patrick Lacarin (placarin@chu‐clermontferrand.fr)

Notes

Planned enrolment: 12

Estimated completion date: June 2011

The status of this trial is "completed", but no publications have been identified.

NCT00790764

Trial name or title

Phase II combination stem cell therapy for the treatment of severe coronary ischemia (CI)

Methods

A phase II, randomised, placebo‐controlled, safety/efficacy study

Participants

Severe coronary ischaemia:

  1. Age 18 to 80.

  2. Men or women.

  3. Angina pectoris: CCS class III or IV or symptoms consistent with angina equivalent (dyspnoea) CCS class III or IV (Functional Class).

  4. Chronic coronary artery disease in at least 1 epicardial vessel with stenosis > 70% by coronary angiography within the last 6 months.

  5. Stable medical therapy for at least 1 month.

  6. Reversible perfusion defects by SPECT.

  7. Not a candidate for coronary artery bypass surgery due to poor targets or small vessels and not a candidate for percutaneous intervention due to small vessels or unreachable coronary lesions due to complicated anatomy.

Interventions

Enrolled individuals (60) will be divided into 2 treatment groups for the infusion of the cell/placebo product:

  1. Intervention arm A: 30 individuals, including patients and placebo controls, will receive the product by intracoronary infusion.

  2. Intervention arm B: 30 individuals, including patients and placebo controls, will receive the product by transendocardial injections.

In turn, each treatment group will consist of 2 subgroups of individuals that will receive the infusion of 1 of the 2 doses established of the cell product:

  1. In subgroup 1, 10 individuals will receive the 'low dose' of the cell product, and 5 individuals will receive the placebo product.

  2. In subgroup 2, 10 individuals will receive the 'high dose' of the cell product, and 5 individuals will receive the placebo product.

For the cell product, proper aliquots of each cell type will be taken to fulfil the doses established for this protocol. The 2 aliquots will be mixed and resuspended to a final volume of 3 mL in the "final suspension medium", which consists of Dulbecco's Phosphate Buffered Saline, containing 5% HSA.

For placebo, 3 mL of the "final suspension medium", which consists of Dulbecco's Phosphate Buffered Saline, containing 5% HSA will be transferred to a 5‐millilitre syringe.

Outcomes

Primary outcome: Safety as measured by laboratory assessments, ECG, and temperature (2 weeks).
Secondary outcome: Efficacy as measured by SPECT scan, MUGA scan, and 2D echocardiogram (6 months).

Starting date

November 2008

Contact information

TCA Cellular Therapy, Covington, LA, United States, 70433 (Principal Investigator: Dr Patrick Lacarin)

Notes

Planned enrolment: 60

Estimated completion date: November 2011

This study has suspended participant recruitment due to lack of funding.

NCT00820586

Trial name or title

Intramyocardial delivery of autologous bone marrow

Methods

A phase II, parallel, randomised, double‐blind (participant, investigator), safety/efficacy study

Participants

Refractory angina:

  1. Participants > 21 years old.

  2. Participants with functional class (CCS) III or IV angina.

  3. Participants with LVEF < 30%.

  4. Attempted 'best' tolerated medical therapy.

  5. Clinical signs and symptoms of myocardial ischaemia with reversible ischaemia on perfusion imaging.

  6. Participant deemed to be a poor candidate or at high surgical risk.

  7. Participant must be able to complete a minimum of 2 minutes but no more than 10 minutes exercise test (Bruce protocol).

  8. Participant (or their legal guardian) understands the nature of the procedure and provides written consent prior to the procedure.

  9. Participant is willing to comply with specified follow‐up evaluations.

  10. Participant must develop angina and a horizontal or down‐sloping ST segment depression of < 1 mm during exercise, compared to pre‐exercise ST segment, 80 ms from the J point or moderate angina with or without the above ST segment changes.

Angiographic inclusion criteria:

  1. Severe obstruction (lumen diameter stenosis > 70%) in a coronary or surgical conduit believed to be solely or partially responsible for angina and myocardial ischaemia.

  2. At least 1 coronary or surgical conduit with < 70% diameter stenosis.

  3. Poor candidate for PCI of treatment zone.

  4. Poor candidates for surgical revascularisation procedures, such as inadequate target coronary anatomy or lack of potential surgical conduits.

Interventions

Intervention arm: Direct intramyocardial percutaneous delivery of autologous total bone marrow‐derived total mononuclear cells or selected CD34+ bone marrow‐derived cells.

Comparator arm: Not specified.

Outcomes

Primary outcome:

Incidence of MACE, defined as a combined endpoint of death, acute MI (Q wave and non‐Q wave), revascularisation procedures (percutaneous or surgical), and periprocedural complications (i.e. left ventricular perforation with haemodynamic consequences requiring pericardiocentesis, and stroke) (1/3/6/12 months).

Secondary outcomes:

  1. Change in CCS angina classification score from baseline (12 months).

  2. Changes in the quality of life, as assessed according to the Seattle Angina Questionnaire.

  3. Change in exercise duration and exercise tolerance using standardised treadmill exercise testing from baseline (6/12 months).

  4. Cumulative number of hospitalisations for coronary ischaemia and CHF (12 months).

  5. SPECT changes in global and regional radionuclide perfusion at rest, peak stress, and redistribution from baseline (1/6/12 months).

  6. Change in angiographic collateral score (6 months).

  7. Change in global and regional myocardial contractility (assessed by echocardiography) from baseline (6/12 months).

Starting date

January 2009

Contact information

Antonio Colombo, Director of Invasive Cardiology Unit, IRCCS San Raffaele, Milan, Italy

Notes

Planned enrolment: 13

Estimated completion date: February 2012

This study has suspended participant recruitment due to lack of further funding support.

NCT00950274

Trial name or title

Intramyocardial transplantation of bone marrow stem cells in addition to coronary artery bypass graft (CABG) surgery (PERFECT)

Methods

A phase III, randomised, parallel, double‐blind (participant, caregiver, investigator, outcomes assessor) efficacy study

Participants

Chronic ischaemic coronary artery disease:

  1. Coronary artery disease after MI with indication for CABG surgery.

  2. Currently reduced global LVEF assessed at site by cardiac MRI at rest (25% ≤ LVEF ≤ 50%).

  3. Presence of a localised akinetic/hypokinetic/hypoperfused area of LV myocardium for defining the target area.

  4. Informed consent of the participant.

  5. Aged ≥ 18 and < 80 years.

  6. Not pregnant and do not plan to become pregnant during the study. Women with childbearing potential must provide a negative pregnancy test within 1 to 7 days before operation and must be using oral or injectable contraception (non‐childbearing potential is defined as postmenopausal for at least 1 year or surgical sterilisation or hysterectomy at least 3 months before study start).

Interventions

Intervention arm: Intramyocardial injection of 5 mL CD133+ cells (0.5 ‐ 5 x 106 cells) suspended in physiological saline + 10% autologous serum intramyocardially during CABG surgery.

Comparator arm: Intramyocardial injection of 5 mL of physiological saline + 10% autologous serum intramyocardially during CABG surgery.

Outcomes

Primary outcome:

LVEF at rest, measured by MRI (6 months).
Secondary outcomes:

  1. Change in LVEF as assessed by MRI and echocardiography (early postoperatively and 6 months).

  2. Regional contractility in the AOI/change in LVESD, LVEDD as assessed by echocardiography (early postoperatively (discharge), 6 months).

  3. Physical exercise capacity determined by 6‐minute walk test (early postoperatively (discharge), 6 months).

  4. NYHA and CCS class (early postoperatively (discharge), 6 months).

  5. MACE (cardiac death, MI, secondary intervention/reoperation, ventricular arrhythmia) (6 months).

  6. QOL score: MLHFQ, SF‐36, EQ‐5D (3 months, 6 months).

Starting date

July 2009

Contact information

University of Rostock, Germany, 18057 (Principal Investigator: Dr G Steinhoff ([email protected]‐rostock.de))

Notes

Planned enrolment: 142

Estimated completion date: December 2013

The status of this trial is "terminated"; no further details are provided, and no publications have been identified.

NCT01033617

Trial name or title

IMPACT‐CABG trial: IMPlantation of Autologous CD133+ sTem Cells in Patients Undergoing CABG (IMPACT‐CABG)

Methods

A phase II, parallel, randomised, double‐blind (participant, caregiver, investigator, outcomes assessor), placebo‐controlled safety/efficacy study

Participants

Myocardial infarct; HF:

  1. Age ≥ 18 and ≤ 75 years.

  2. People with severe chronic ischaemic cardiomyopathy manifested by CCS class II or greater angina or NYHA class II or greater, or both, and who have undergone diagnostic coronary angiography demonstrating ≥ 70% diameter narrowing of at least 2 major coronary arteries or branches or ≥ 50% diameter narrowing of the left main coronary artery.

  3. A significant left ventricular systolic dysfunction evaluated by echocardiography or LV angiography (LVEF ≤ 45% but ≥ 25%) due to prior MI. This area of left ventricular dysfunction should be akinetic or severely hypokinetic, not dyskinetic or aneurysmal, when assessed by echocardiography or LV angiogram. This territory should be irrigated by 1 or a branch of the 3 major vascular territories (i.e. right coronary artery, left circumflex, or left anterior descending artery distribution) that will be bypassed during the surgical procedure.

  4. No contraindications or exclusions (see below).

  5. Willingness to participate and ability to provide informed consent.

Interventions

Intervention arm: Autologous CD133+ stem cells (total 2 mL with 10 to 15 injections) injected into the myocardium.

Comparator arm: Placebo solution containing plasma injected into the myocardium.

Outcomes

Primary outcomes:

  1. Freedom from MACE: cardiac death, myocardial infarct, repeat coronary bypass grafting or percutaneous intervention of bypassed artery (6 months).

  2. Freedom from major arrhythmia: sustained ventricular tachycardia or survived sudden death (6 months).

Secondary outcomes:

  1. Regional myocardial perfusion and function assessed by magnetic resonance scans (6 months).

  2. Device performance endpoint: feasibility to produce from 100 mL of bone marrow aspiration a final cell product that contains a target CD133+ cells higher than 0.5 million with a purity superior to 30% and a recovery superior to 10% (baseline).

  3. Symptom severity and quality of life after CABG surgery (6 months).

Starting date

December 2009

Contact information

Centre de recherche du CHUM (CRCHUM), Montreal, Quebec, Canada, H2W 1T8 (Principal Investigators: Dr N Noiseux, Dr S Mansour, Dr D‐C Roy). Contact: Nicolas Noiseux, MD, MSc, FRCSC, ([email protected])

Notes

Planned enrolment: 20

Estimated completion date: July 2013

The recruitment status of this study is unknown because the information has not been recently verified.

NCT01214499

Trial name or title

Prospective, controlled and randomized clinical trial on cardiac cell regeneration with laser and autologous bone marrow stem cells, in patients with coronary disease and refractory angina

Methods

A phase II, randomised, single‐blind (outcome assessor), parallel safety/efficacy study

Participants

Coronary disease and refractory angina:

  1. Aged > 18 years of age.

  2. At least 1 area of myocardial ischaemia or chronic MI of the left ventricle demonstrated by any imaging technique not amenable to conventional revascularisation and angina refractory to medical treatment.

  3. LVEF > 25% measured in the 6 months prior to the procedure.

  4. Participants must be mentally competent to give consent for inclusion in the clinical trial.

Interventions

Intervention arm: Transmyocardial revascularisation with Holmium YAG laser plus the participant's own stem cells extracted from bone marrow.

Comparator arm: Transmyocardial revascularisation with Holmium YAG laser.

Outcomes

Primary outcome:

NYHA classification for angina (12 months).
Secondary outcomes:

  1. The demographic, intra‐operative, and postoperative variables (12 months).

  2. Percentage of ischaemic area (SPECT) and maximum effort capacity before the occurrence of the angina (12 months).

  3. LVEF, LVESV, LVEDV will be examined through an echocardiogram and a pre‐ and postoperative cardiac magnetic resonance imaging study (12 months).

  4. The EQ‐5D (standardised instrument for use as a measure of health outcome) will be completed by the participant for the subjective assessment of quality of life (12 months).

Starting date

October 2010

Contact information

Cardiovascular Surgery Service, Hospital Universitario de La Princesa, Madrid, Spain, 28006 (Principal Investigator: Dr GR Copa ([email protected]))

Notes

Planned enrolment: 20

Estimated completion date: October 2012

The recruitment status of this study is unknown because the information has not been recently verified.

NCT01267331

Trial name or title

Cell therapy in patients with chronic ischemic heart disease undergoing cardiac surgery

Methods

A phase I/II, randomised, double‐blind (participant, caregiver), parallel safety/efficacy study

Participants

Severe, chronic ischaemic disease:

  1. Aged between 18 and 75 years.

  2. Scheduled to undergo CABG.

  3. At least 3 months since last episode of MI.

  4. Echocardiogram‐assessed LVEF between 15% and 40% (Simpson's rule).

  5. Abnormal wall motion of at least 1 segment due to prior MI shown by echocardiography or left ventriculography.

  6. Abnormal myocardial perfusion in infarcted area by SPECT.

  7. Willingness to participate and ability to provide written informed consent.

Interventions

Intervention arm: Direct intramyocardial injection of autologous bone marrow mononuclear cells during CABG.

Comparator arm: Between 10 and 15 placebo injections consisting of saline and 5% HSA during CABG.

Outcomes

Primary outcome:

Major adverse cardiac events (6 months)
Secondary outcomes:

Left ventricular function (global function, regional myocardial perfusion, and function assessed by magnetic resonance imaging and echocardiogram) (6 months)

Starting date

December 2010

Contact information

Chinese PLA General Hospital, Beijing, China (Principal Investigator: Dr C Gao ([email protected]) and Dr L Zhang ([email protected]))

Notes

Planned enrolment: 60

Estimated completion date: June 2013

The recruitment status of this study is unknown because the information has not been recently verified.

NCT01354678

Trial name or title

Intramyocardial Multiple Precision Injection of Bone Marrow Mononuclear Cells in Myocardial Ischemia (IMPI)

Methods

A phase I, parallel, randomised, double‐blind (participant, caregiver, investigator) safety/efficacy study

Participants

Ischaemic HF:

  1. Participants with coronary artery disease and HF NYHA class II‐III.

  2. MI > 6 months before the study.

  3. LVEF < 35%.

  4. Absence of indication to coronary revascularisation.

  5. Optimal pharmacological therapy no less than 8 weeks.

  6. Heart transplantation is contraindicated.

  7. Participants with implantable cardioverter‐defibrillator or cardiac resynchronisation therapy defibrillator.

  8. Participants giving informed consent.

Interventions

Intervention arm: Intramyocardial multiple precision injection of bone marrow mononuclear cells.

Comparator arm: Intramyocardial multiple precision injection with placebo.

Outcomes

Primary outcome: Change in global LVEF and regional wall motion score index (6/12 months).
Secondary outcomes: Incidence of the major adverse cardiac events (6/12 months).

Starting date

May 2011

Contact information

Almazov Federal Center of Heart, Blood and Endocrinology (Principal Investigator: Prof EV Shlyakhto). Contact: Prof DS Lebedev ([email protected]); Prof OM Moiseeva ([email protected])

Notes

Planned enrolment: 30

Estimated completion date: May 2015

This study is marked as ongoing but is not currently recruiting participants.

First identified publication in Russian (Shlyakhto 2013) confirmed by translator as early report of safety in 1 participant.

NCT01467232

Trial name or title

IMPACT‐CABG trial: IMPlantation of Autologous CD133+ sTem Cells in Patients Undergoing Coronary Artery Bypass Grafting

Methods

A phase II, randomised, parallel, double‐blind (participant, caregiver, investigator, outcomes assessor) safety/efficacy study

Participants

Chronic ischaemic cardiomyopathy:

  1. Age ≥ 18 and ≤ 75 years.

  2. People with severe chronic ischaemic cardiomyopathy manifested by CCS class II or greater angina or NYHA class II or greater dyspnoea, or both, AND who have undergone diagnostic coronary angiography demonstrating ≥ 70% diameter narrowing of at least 2 major coronary arteries or branches or ≥ 50% diameter narrowing of the left main coronary artery.

  3. Significant left ventricular systolic dysfunction evaluated by echocardiography or LV angiography (LVEF ≤ 45% but ≥ 25%) due to a prior MI. This area of left ventricular dysfunction should be akinetic or severely hypokinetic, not dyskinetic or aneurysmal, when assessed by echocardiography or LV angiogram.

  4. No contraindications or exclusions.

  5. Willingness to participate and ability to provide informed consent.

Interventions

Intervention arm: Autologous CD133+ stem cells injected into the mycardium.

Comparator arm: Placebo (saline solution containing autologous plasma without CD133+).

Outcomes

Primary outcomes:

  1. Freedom from major adverse cardiac event at 6 months (cardiac death, myocardial infarct, repeat coronary bypass grafting or percutaneous intervention of bypassed artery).

  2. Freedom from major arrhythmia at 6 months (sustained ventricular tachycardia or survived sudden death).

Secondary outcomes:

  1. Regional myocardial perfusion and function assessed by magnetic resonance scans.

  2. Global ventricular function assessed by echocardiographic measures of ejection fraction.

  3. Relief of symptom severity after CABG surgery.

  4. Device performance endpoint.

  5. Feasibility to produce from 100 mL of bone marrow aspiration a final cell product that contains a target CD133+ cells higher than 0.5 million with a purity superior to 30% and a recovery superior to 10%.

  6. Quality of life after CABG surgery.

Starting date

September 2011

Contact information

Terrence M Yau, MD, Peter Munk Cardiac Center/University Health Network, Toronto, Ontario, Canada, M5G 2C4

Notes

Planned enrolment: 20

Estimated completion date: October 2015

This study is marked as completed, but no publications have been identified.

NCT01508910

Trial name or title

Efficacy and Safety of Targeted Intramyocardial Delivery of Auto CD34+ Stem Cells for Improving Exercise Capacity in Subjects With Refractory Angina (RENEW)

Methods

A phase III, randomised, parallel, double‐blind (participant, investigator) safety/efficacy study

Participants

Refractory angina and chronic myocardial ischaemia:

  1. Men or women aged 21 to 80 years at the time of signing the informed consent.

  2. Participants with CCS class III or IV chronic refractory angina.

  3. Participants without control of their angina symptoms despite maximal tolerated doses of antiangina drugs. Participants must be on optimal therapy for their angina and have been on a stable antianginal medication regimen for at least 4 weeks before signing the informed consent form.

  4. Participants with obstructive coronary disease unsuitable for conventional revascularisation due to unsuitable anatomy or comorbidity as determined at the site and confirmed by an independent adjudication committee.

  5. Participants must have evidence of inducible myocardial ischaemia.

  6. Participants must experience angina episodes.

  7. Participants must be able to complete 2 exercise tolerance tests on the treadmill within 3 weeks of randomisation.

  8. If a woman of childbearing potential, she must not be pregnant and must agree to employ adequate birth control measures for the duration of the study.

Interventions

Intervention arm: Targeted intramyocardial delivery of 1 x 105 Auto‐CD34+ cells after G‐CSF mobilisation and apheresis.

Comparator arm: Targeted intramyocardial delivery of placebo after G‐CSF mobilisation and apheresis.

Outcomes

Primary outcome:

Change from baseline in total exercise time on exercise tolerance test using the modified Bruce protocol (12 months).
Secondary outcomes:

  1. Angina frequency (episodes per week) (3/6/12 months).

  2. Change from baseline in total exercise time on exercise tolerance test (6 months).

  3. Incidence of MACE and other serious adverse events in all participants (24 months).

Starting date

April 2012

Contact information

Baxter Healthcare Corporation (Study Director: Dr A Nada). Contact: Lauren Davis, Clinical Project Manager ([email protected])

Notes

Planned enrolment: 291

Estimated completion date: June 2016

This study is marked as completed, but no publications have been identified.

NCT01615250

Trial name or title

Implantation of Peripheral Stem Cells in Patient With Ischemic Cardiomyopathy (ISCIC)

Methods

A phase I, randomised, parallel, open‐label safety/efficacy study

Participants

Ischaemic cardiomyopathy:

  1. People with ischaemic cardiomyopathy and HF NYHA class II‐IV.

  2. MI > 6 months before the study.

  3. LVEF < 35%.

  4. Absence of effect of coronary revascularisation during 6 months.

  5. Optimal pharmacological therapy no less than 8 weeks.

  6. Heart transplantation is contraindicated.

  7. Participants with implantable cardioverter‐defibrillator or cardiac resynchronisation therapy defibrillator.

  8. Participants giving informed consent.

Interventions

Intervention arm: Intramyocardial implantation of peripheral mononuclear cells with CD34+ stem cells in participant with ischaemic cardiomyopathy after preparatory course of shockwave therapy.

Comparator arm: Cardiospec shockwave therapy only.

Outcomes

Primary outcomes:

Change in global LVEF and regional wall motion score index (6/12 months).
Secondary outcomes:

Incidence of MACE (6/12 months).

Starting date

January 2012

Contact information

Odessa Regional Clinical Hospital, Odessa, Ukraine, 65025 (Principal Investigator: Prof II Karpenko ([email protected]))

Notes

Planned enrolment: 50

Estimated completion date: January 2016

The recruitment status of this study is unknown because the information has not been recently verified.

NCT01660581

Trial name or title

Intracardiac CD133+ cells in patients with no‐option resistant angina (RegentVsel)

Methods

A phase II, randomised, parallel, double‐blind (participant, investigator) efficacy study

Participants

Stable angina:

  1. Stable angina CCS II‐IV despite maximum pharmacotherapy for at least 2 weeks since last medications change.

  2. Presence of ≥ 1 myocardial segment with ischaemia features in Tc‐99m SPECT.

  3. Participants disqualified from revascularisation procedures by Heart Team.

  4. Aged over 18 and less than 75 years old.

  5. Person must provide written informed consent for participation in study.

Interventions

Intervention arm: Intramyocardial injection (electromechanical mapping based) of autological CD133+ cells, isolated from bone marrow.

Comparator arm: Intramyocardial injection (electromechanical mapping based) of placebo (0.9% sodium chloride plus 0.5% solution of participant's serum).

Outcomes

Primary outcome:

Myocardial perfusion change (4 months).
Secondary outcomes:

  1. Global and segmental contractility change and myocardial perfusion change (MRI: 4 months; echocardiography: 4/12 months).

  2. Exercise tolerance (4/12 months).

  3. Occurrence of symptomatic angina (1/4/6/12 months).

  4. Quality of life (1/4/6/12 months).

  5. Occurrence of ventricular arrhythmia (1/4/6/12 months).

  6. Occurrence of in‐stent restenosis and progression of atherosclerotic lesions in remained coronary artery segments (4 months).

Starting date

June 2012

Contact information

Samodzielny Publiczny Szpital Kliniczny nr 7 Śląskiego Uniwersytetu Medycznego w Katowicach Górnośląskie Centrum Medyczne im. prof. Leszka Gieca, Katowice‐Ochojec, Silesian, Poland, 40‐635 (Principal Investigator: Prof W Wojakowski ([email protected]))

Notes

Planned enrolment: 60

Estimated completion date: June 2014

This study is currently recruiting participants.

NCT01720888

Trial name or title

Intracoronary autologous mesenchymal stem cells implantation in patients with ischemic dilated cardiomyopathy

Methods

A phase II, randomised, parallel, open‐label efficacy study

Participants

Ischaemic dilated cardiomyopathy:

  1. Aged between 35 and 75 years.

  2. Diagnosed as having ischaemic cardiomyopathy confirmed by previous coronary angiogram showing significant coronary artery disease > 70% or history of previous MI.

  3. Myocardial infarction event occurred 6 months or longer from time of screening.

  4. Left ventricular ejection fraction of ≤ 40% by echocardiogram or cardiac MRI.

Interventions

Intervention arm: Intracoronary implantation of autologous bone marrow‐derived mesenchymal stem cells.

Comparator arm: Control (optimal medical therapy).

Outcomes

Primary outcome:

Change in LV ejection fraction as measured by echocardiogram and cardiac MRI after implantation (measured at 1, 3, 6, 9, and 12 months).

Secondary outcomes:

  1. Changes in functional status (12 months).

  2. Improvement in other LV parameters as assessed by echocardiogram and cardiovascular magnetic resonance (1, 3, 6, 9, 12 months).

  3. Resolution of scar tissue volume/area on cardiac MRI (6, 12 months).

  4. Change in serum NT‐proBNP level (1, 6, 12 months).

  5. Freedom from major adverse cardiac events as defined by MI, hospitalisation for angina, MI or HF, or death (all‐cause mortality) (1, 3, 6, 9, 12 months).

Other outcomes:

  1. No periprocedural complications (1, 3, 6, 9, 12 months).

  2. Significant improvement in overall left ventricular function (12 months).

  3. Resolution of scar tissue (6, 12 months).

  4. Reduction of major adverse cardiac events (1, 3, 6, 9, 12 months).

Starting date

July 2012

Contact information

Oteh Maskon, MB Bch (Principal Investigator), UKM Medical Centre, Cheras, Kuala Lumpur, Malaysia, 56000

Notes

Planned enrolment: 80

Estimated completion date: December 2015

This study is ongoing but not recruiting participants.

NCT01727063

Trial name or title

Cell therapy in severe chronic ischemic heart disease (MiHeart)

Methods

A phase II/III, randomised, parallel, double‐blind (participant, investigator) safety/efficacy study

Participants

Chronic IHD:

  1. Symptoms of angina or angina equivalent.

  2. Documented coronary artery disease (invasive angiography).

  3. Documented myocardial ischaemia (stress echo, cardiac scintigraphy, or MRI).

  4. Unsuitable for complete myocardial revascularisation (PCI or CABG) OR even if a complete procedure is feasible, it is anticipated that myocardial perfusion may not be restored due to poor distal beds.

Interventions

Intervention arm: Intramyocardial injection of autologous bone marrow‐derived cells.

Comparator arm: Saline injection.

Outcomes

Primary outcome:

Increase in myocardial perfusion assessed by MRI (1/6/12 months).
Secondary outcomes:

  1. Improvement in LV function assessed by MRI (1/6/12 months).

  2. Improvement in angina functional class determined using the CCS classification (1/6/12 months).

Starting date

January 2006

Contact information

Heart Institute, Sao Paulo, SP, Brazil 05403‐000 (Principal Investigator: Prof LHW Gowdak). Contact: Meyrielli A Vieira ([email protected]); Prof LHW Gowdak ([email protected])

Notes

Planned enrolment: 200

Estimated completion date: July 2013

The recruitment status of this study is unknown because the information has not been recently verified.

NCT01758406

Trial name or title

Transplantation of autologous cardiac stem cells in ischemic heart failure

Methods

A phase II, randomised, parallel, double‐blind (participant, investigator) safety/efficacy study

Participants

Ischaemic HF:

  1. EF ≤ 40 (by echocardiography).

  2. Not responding to standard therapies for HF for over 1 month.

  3. NYHA class III or greater.

  4. MI due to coronary artery atherosclerotic disease.

  5. An area of regional dysfunction, i.e. hypokinetic, akinetic, or dyskinetic (echocardiography or MRI).

  6. No HIV/viral hepatitis.

  7. Normal liver function (SGPT < 3 times the upper reference range).

  8. No or controlled diabetes (glycated haemoglobin < 8.5%).

  9. Ability to provide informed consent and follow‐up with protocol procedures.

Interventions

Intervention arm: Autologous cardiac stem cell intracoronary injection.

Comparator arm: Placebo (no details).

Outcomes

Primary outcomes:

Rate of mortality, arrhythmia, and hospitalisation at 18 months.

Secondary outcomes:

  1. Ejection fraction changes (18 months).

  2. NT‐proBNP changes (18 months).

  3. NYHA functional class (18 months).

  4. 6‐minute walk test (18 months).

Starting date

December 2013

Contact information

Hoda Madani, MD (Principal Investigator), Royan Institute, Tehran, Iran. Contact: Nasser Aghdami, MD, PhD +982123562000 ext 504 ([email protected])

Notes

Planned enrolment: 50

Estimated completion date: December 2017

This study is currently recruiting participants.

NCT01768702

Trial name or title

Safety and efficacy of autologous cardiopoietic cells for treatment of ischemic heart failure (CHART‐1)

Methods

A phase III, randomised, parallel, double‐blind (participant, outcomes assessor) safety/efficacy study

Participants

Ischaemic HF:

  1. Age ≥ 18 and < 80 years.

  2. Systolic dysfunction with LVEF ≤ 30% as assessed by echocardiography.

  3. Ischaemic HF without known need for revascularisation.

  4. MLHFQ score > 30.

  5. Ability to perform a 6‐minute walk test > 100 m and ≤ 400 m.

  6. History of hospitalisation for HF within 12 months prior to screening.

  7. NYHA class III or IV despite optimal standard of care or INTERMACS class 4, 5, 6, or 7.

  8. Use of ACE inhibitor and/or ARB and beta blocker for at least 3 months prior to screening visit, unless intolerant or contraindicated.

  9. Stable dosing of ACE inhibitor, ARB, beta blocker, aldosterone blocker, and diuretics for at least 1 month prior to screening visit, defined as ≤ 50% change in total dose of each agent.

  10. Willing and able to give written informed consent.

Interventions

Intervention arm: Injection of C3BS‐CQR‐1 cardiopoietic cells using the C‐Cath injection catheter.

Comparator arm: Mimic injection procedure through insertion of a sham catheter. No injection actually performed.

Outcomes

Primary outcome:

Efficacy between groups post‐index procedure: change between groups from baseline in a hierarchical composite outcome comprising, from most‐ to least‐severe outcome, days to death from any cause, number of worsening of HF events, change in score for the MLHFQ (10‐point deterioration, no meaningful change, 10‐point improvement), change in 6‐minute walk distance (40 m deterioration, no meaningful change, 40 m improvement), and change in LVESV (15 mL deterioration, no meaningful change, 15 mL improvement) and LVEF (4% absolute deterioration, no meaningful change, 4% absolute improvement) (39 weeks).
Secondary outcomes:

  1. Efficacy (time to all‐cause mortality, time to worsening of HF, and time to aborted sudden death) and safety (number and cause of deaths and readmissions, number of cardiac transplantations, number of MIs, number of strokes, incidence of serious AEs and non‐serious AEs) between groups post‐index procedure (52/104 weeks).

  2. Efficacy and safety between groups post‐index procedure (time to all‐cause mortality, time to cardiovascular mortality, and rate of worsening HF requiring outpatient IV therapy for HF or readmission for HF, and other) (39/52 weeks post‐index).

Starting date

November 2012

Contact information

Multicentre study: Cardio3 BioSciences (Study Chairs: Dr A Terzic, Mayo Clinic, Division of Cardiovascular Diseases, Rochester, MN, USA and Dr J Bartunek, OLV Ziekenhuiz Aalst, Belgium). Contact: Dr Christian Homsy ([email protected])

Notes

Planned enrolment: 240

Estimated completion date: March 2017

This study is ongoing but not recruiting participants.

NCT02022514

Trial name or title

Intracoronary infusion of mononuclear cells autologous bone marrow in patients with chronic coronary occlusion and ventricular dysfunction, previously revascularized

Methods

Open‐label randomised controlled trial

Participants

Chronic coronary artery occlusion:

  1. People of both sexes with atherosclerotic coronary disease and chronic occlusions older than 3 months in which successful recanalisation was achieved, medicated stents implanted, and where it persists despite ventricular dysfunction.

  2. Age between 18 and 80 years.

  3. The baseline ventricular function recanalisation catheterisation (performed approximately 3 months earlier) should be less than 45% ejection fraction.

  4. The ejection fraction of the person should remain below 45% in the MRI performed at 3 months of recanalisation.

Interventions

Intervention arm: Bone marrow mononuclear cells by intra‐arterial administration.

Comparator arm: Control (no placebo).

Outcomes

Primary outcome:

Change in ejection fraction measured by MRI between inclusion and 6 months' follow‐up.

Secondary outcomes:

  1. Changes in NYHA functional grade between groups.

  2. Possible cardiac events during follow‐up (death, MI, repeat revascularisation).

  3. Need for hospitalisation or major arrhythmias.

  4. Changes in global and segmental left ventricular function.

Starting date

November 2013

Contact information

None identified.

Notes

Planned enrolment: 66

Estimated completion date: May 2017

This study is currently recruiting participants.

NCT02059512

Trial name or title

Autologous bone marrow mononuclear cells in the combined treatment of coronary heart disease (TAMIS)

Methods

A phase III, randomised, parallel, single‐blind (investigator) efficacy study

Participants

Coronary heart disease:

  1. Men and women from 18 to 80 years.

  2. People with angina pectoris III‐IV functional class.

  3. People signed informed consent.

Interventions

Intervention arm 1: Bone marrow mononuclear cells (intramyocardial).

Intervention arm 2: Bone marrow mononuclear cells (intramyocardial and intracoronary).

Comparator arm: Placebo (intramyocardial administration of 0.9% sodium chloride 0.2 mL).

Outcomes

Primary outcome:

All‐cause mortality associated with the progression of basic disease (60 months).

Secondary outcomes:

Quality of life (12 months).

Other outcomes:

  1. Percentage of functioning grafts in participants with implantation of autologous bone marrow mononuclear cells (60 months).

  2. Estimation of efficiency: (i) assessment of myocardial perfusion and metabolism (before and after treatment); (ii) evaluation of systolic and diastolic myocardial function; (iii) speckle tracking echocardiography; (iv) patency of grafts within a specified time of treatment (angiography); (v) dependence and duration of positive clinical effect on the amount of injected cell material; (vi) evaluation of the quality of life (Minnesota questionnaire, Seattle questionnaire, SF‐36 questionnaire).

Starting date

February 2013

Contact information

Alexander S Nemkov, MD, PhD (Principal Investigator), First Pavlov State Medical University of St Petersburg, St Petersburg, Russia, 197089 ([email protected])

Notes

Planned enrolment: 100

Estimated completion date: February 2018

This study is ongoing but not recruiting participants.

NCT02317458

Trial name or title

Congestive Heart Failure Cardiopoietic Regenerative Therapy (CHART‐2) trial

Methods

A phase III, randomised, parallel, double‐blind (participant, caregiver, investigator, outcomes assessor) safety/efficacy study

Participants

Advanced chronic ischaemic HF:

  1. Age ≥ 18 and < 80 years.

  2. Chronic HF, NYHA class II or greater, without need for revascularisation.

  3. Systolic dysfunction with LVEF ≤ 35%.

  4. Total MLHFQ score > 30.

  5. 6‐minute walk test distance > 100 m and < 400 m.

  6. Hospitalisation or outpatient with intravenous therapy for HF within the previous 12 months.

  7. Stable medical regimen, including ACE inhibitor or ARB, or both; beta blocker, aldosterone blocker and diuretic for at least 1 month.

  8. Willing and able to give written informed consent.

Interventions

Intervention arm: BM‐derived mesenchymal cardiopoietic cells (C3BR‐CQR‐1) using intramyocardial injection.

Comparator arm: Control (standard of care with sham procedure).

Outcomes

Primary outcome:

6‐minute walk test at 39 weeks' postprocedure.

Secondary outcomes:

None reported.

Starting date

December 2014

Contact information

Celyad (formerly named Cardio3 BioSciences)

Notes

Planned enrolment: 240

Estimated completion date: August 2018

This study is not yet open for participant recruitment.

NCT02362646

Trial name or title

Safety & efficacy of intramyocardial injection of mesenchymal precursor cells on myocardial function in LVAD recipients

Methods

A phase II, randomised, parallel, double‐blind (participant, caregiver, investigator, outcomes assessor) safety/efficacy study

Participants

Left ventricular assist device recipients:

  1. Age 18 years or older.

  2. If the participant or partner is of childbearing potential, he or she must be willing to use adequate contraception (hormonal or barrier method or abstinence) from the time of screening and for a period of at least 16 weeks after procedure.

  3. Female participants of childbearing potential must have a negative serum pregnancy test at screening.

  4. Admitted to the clinical center at the time of randomisation.

  5. Clinical indication and accepted candidate for implantation of an FDA‐approved (US sites only) or Health Canada‐approved (Canadian sites only) implantable, non‐pulsatile LVAD as a bridge to transplantation or for destination therapy.

Interventions

Intervention arm: Mensenchymal precursor cells (intramyocardial injection).

Comparator arm: Placebo (50% Alpha‐Minimum Essential Medium/42.5% ProFreeze NAO Freeze Medium/7.5% DMSO)

Outcomes

Primary outcomes:

  1. Functional status (6 months).

  2. Adverse events (12 months).

Secondary outcomes:

  1. Physiologic assessments (12 months).

  2. Histopathological assessments of myocardial tissue (12 months).

  3. Overall survival (12 months).

  4. Change in quality of life (6, 12 months).

  5. Hopkins Verbal Learning Test (3, 12 months).

  6. Trail Making Tests A and B (3, 12 months).

  7. Medical College of Georgia (MCG) complex figures (3, 12 months).

  8. Digit span (3, 12 months).

  9. Digit symbol substitution test (3, 12 months).

  10. Controlled Oral Word Association Test (3, 12 months).

  11. Length of hospital stay (12 months).

  12. Hospitalisations (12 months).

  13. Hospital costs (12 months).

  14. Functional status (12 months).

Starting date

July 2015

Contact information

Michael Bowdish, MD (Principal Investigator), University of Southern California, Los Angeles, California, United States, 90033 ([email protected]); Joseph Woo, MD (Principal Investigator), Stanford University School of Medicine, Stanford, California, United States, 94305 ([email protected])

Notes

Planned enrolment: 120

Estimated completion date: August 2016

This study is currently recruiting participants.

NCT02438306

Trial name or title

CardiAMP Heart Failure Trial

Methods

A phase III, randomised, parallel, double‐blind (participant, outcome assessor) safety/efficacy study

Participants

Post‐MI HF:

  1. Older than 21 and younger than 90 years of age.

  2. NYHA class II or III.

  3. Diagnosis of chronic ischaemic left ventricular dysfunction secondary to MI.

  4. Have an ejection fraction ≥ 20% and ≤ 40%.

  5. On stable evidence‐based medical and device therapy for HF or postinfarction left ventricular dysfunction, per the 2013 ACC/AHA heart failure guidelines, for at least 3 months prior to randomisation.

  6. Cell potency assay score of 3, as determined by the Cell Analysis Core Lab results.

  7. Provide written informed consent.

Interventions

Intervention arm: Autologous CardiAMP cell therapy.

Comparator arm: Placement of an introducer and performance of LV gram only.

Outcomes

Primary outcome:

Change in 6‐minute walk distance at 12 months from baseline.

Secondary outcomes:

  1. Overall survival as a non‐inferiority outcome (12 months).

  2. Freedom from MACE (composite of all‐cause death, hospitalisation for worsening HF, non‐fatal MI, [LVAD], or heart transplantation) as a non‐inferiority outcome (12 months).

  3. Change in quality of life as measured by MLHFQ as a superiority outcome (12 months).

  4. Time to first MACE as a superiority outcome (12 months).

  5. Overall survival as a superiority outcome (12 months).

  6. Survival at 2 years.

  7. HF death (12 months).

  8. Treatment‐emergent serious adverse event at 30 days.

  9. HF hospitalisation (12 months).

  10. All‐cause hospitalisation (12 months).

  11. Days alive out of hospital (12 months).

  12. Freedom from serious adverse events (12 months).

  13. NYHA Functional Class (12 months).

  14. 6‐minute walk distance repeated measure analysis (12 months).

  15. Echocardiographic measures of change in ejection fraction, left ventricular end‐systolic and end‐diastolic volumes, left ventricular end‐systolic and end‐diastolic dimensions, mitral regurgitation (composite) (baseline).

  16. Technical success defined as successful delivery of ABM MNC, at the time of the procedure.

Starting date

January 2016

Contact information

Cheryl Wong Po Foo, BioCardia Inc (Email [email protected]). Principal Investigators: Carl Pepine, University of Florida and Amish Raval, University of Wisconsin

Notes

Planned enrolment: 250

Estimated completion date: April 2019

This study is not yet open for participant recruitment.

NCT02462330

Trial name or title

Administration of mesenchymal stem cells in patients with chronic ischemic cardiomyopathy (MESAMI2)

Methods

A phase II, randomised, parallel, double‐blind (participant, investigator) efficacy study

Participants

Chronic ischaemic cardiomyopathy and left ventricular dysfunction:

  1. Aged 18 to 75 years.

  2. Signed the informed consent.

  3. Chronic, stable ischaemic cardiomyopathy for at least 1 month with a NYHA class II‐IV or angina pectoris CCS class III or IV, or both.

  4. Not a candidate for revascularisation by coronary artery bypass surgery or angioplasty.

  5. Left ventricular function ≤ 45%.

  6. Presence of ischaemia or myocardial viability on the myocardial perfusion imaging.

  7. VO2 max ≤ 20 mL/min/kg.

  8. Optimal medical therapy.

  9. Optimal interventional therapy (implantable cardioverter defibrillator, effort rehabilitation).

Interventions

Intervention arm: BMMSC (isolated and cultured during 17 ± 2 days by the French Blood Establishment; administered by intramyocardial injections of MSC using the electromechanical NOGA‐XP system).

Comparator arm: Human albumin 4%.

Outcomes

Primary outcome:

Change in VO2max (or peak VO2) before injection and at 3 months' postinjection.

Secondary outcomes:

  1. Left ventricular viability (3 and 12 months).

  2. Change in NYHA/CCS class (3 and 12 months).

  3. Change on quality of life test score (3 and 12 months).

  4. Change in VO2max (or peak VO2) at 3 and 12 months postinjection.

  5. 6‐minute walk test (3 and 12 months).

  6. Volume of myocardium and measurement of ejection fraction (echocardiography) (3 and 12 months).

  7. Myocardial perfusion imaging (3 and 12 months).

Other outcomes:

  1. Adverse event related to cell administration (12 months).

  2. Complication related to cell administration (12 months).

  3. Control of the implantable cardioverter defibrillator (12 months).

  4. Analysis of major cardiovascular events (12 months).

Starting date

June 2015

Contact information

Jerome Roncalli, MD, PhD (Principal Investigator), Cardiology Department of Rangueil Hospital, Toulouse, France, 31059 (roncalli.j@chu‐toulouse.fr)

Notes

Planned enrolment: 90

Estimated completion date: May 2017

This study is currently recruiting participants.

NCT02501811

Trial name or title

Combination of mesenchymal and c‐kit+ cardiac stem cells as regenerative therapy for heart failure (CONCERT‐HF)

Methods

A phase II, randomised, parallel, double‐blind (participant, caregiver, investigator, outcomes assessor) safety/efficacy study

Participants

Ischaemic cardiomyopathy:

  1. ≥ 21 and < 80 years of age.

  2. Documented coronary artery disease with evidence of myocardial injury, LV dysfunction, and clinical evidence of HF.

  3. "Detectable" area of myocardial injury defined as ≥ 5% LV involvement (infarct volume) and any subendocardial involvement by cMRI.

  4. EF ≤ 40% by cMRI.

  5. Receiving guideline‐driven medical therapy for HF at stable and tolerated doses for ≥ 1 month prior to consent. For beta‐blockade, "stable" is defined as no greater than a 50% reduction in dose or no more than a 100% increase in dose.

  6. Candidate for cardiac catheterisation.

  7. NYHA class II or III HF symptoms.

  8. If a female of childbearing potential, be willing to use one form of birth control for the duration of the study, and undergo a pregnancy test at baseline and within 36 hours prior to injection.

Interventions

Intervention arm 1: 15 transendocardial injections of 0.4 mL BMMSC administered to the left ventricle via NOGA MyoStar injection catheter (single procedure) (target dose 150 million MSC).

Intervention arm 2: 15 transendocardial injections of 0.4 mL cardiac stem cells (c‐kit+ cells) administered to the left ventricle via NOGA MyoStar injection catheter (single procedure) (target dose 5 million CSC).

Intervention arm 3: Combo: 15 transendocardial injections of 0.4 mL MSC administered to the left ventricle via NOGA MyoStar injection catheter (single procedure) and 15 transendocardial injections of 0.4 mL CSC administered to the left ventricle via NOGA MyoStar injection catheter (single procedure) (target dose 150 million MSC and 5 million CSC).

Comparator arm: 15 transendocardial injections of 0.4 mL placebo (Plasma‐Lyte A) administered to the left ventricle via NOGA MyoStar injection catheter (single procedure).

Outcomes

Primary outcomes:

  1. Change in LVEF as measured by cMRI (12 months).

  2. Change in global strain (HARP MRI) as measured by cMRI (12 months).

  3. Change in regional strain (HARP MRI) as measured by cMRI (12 months).

  4. Change in Left Ventricular End‐Diastolic Volume Index (LVEDVI) as measured by cMRI (12 months).

  5. Change in Left Ventricular End‐Systolic Volume Index (LVESVI) as measured by cMRI (12 months).

  6. Change in Left Ventricular Sphericity Index as measured by cMRI (12 months).

  7. Change in infarct/scar volume (delayed enhancement MRI) as measured by cMRI (12 months).

  8. Change in maximal oxygen consumption (VO2 max) as measured by treadmill (12 months).

  9. Change in exercise tolerance as measured by the 6‐minute walk test (12 months).

  10. Change in MLHFQ score (12 months).

  11. Incidence rate of MACE (12 months).

  12. Cumulative days alive and out of hospital (12 months).

  13. Change in NT‐proBNP as measured by blood draw (12 months).

Secondary outcomes:

  1. Change in LVEF as measured by cMRI (6 months).

  2. Change in global strain (HARP MRI) as measured by cMRI (6 months).

  3. Change in regional strain (HARP MRI) as measured by cMRI (6 months).

  4. Change in Left Ventricular End‐Diastolic Volume Index (LVEDVI) as measured by cMRI (6 months).

  5. Change in Left Ventricular End‐Systolic Volume Index (LVESVI) as measured by cMRI (6 months).

  6. Change in Left Ventricular Sphericity Index as measured by cMRI (6 months).

  7. Change in infarct/scar volume (delayed enhancement MRI) as measured by cMRI (6 months).

  8. Change in maximal oxygen consumption (VO2 max) as measured by treadmill (6 months).

  9. Change in exercise tolerance as measured by the 6‐minute walk test (6 months).

  10. Change in MLHFQ score (6 months).

  11. Incidence rate of MACE (6 months).

  12. Cumulative days alive and out of hospital (6 months).

  13. Change in NT‐proBNP as measured by blood draw (6 months).

Other outcomes:

  1. Change in LVEF as measured by cMRI (6 to 12 months).

  2. Change in global strain (HARP MRI) as measured by cMRI (6 to 12 months).

  3. Change in regional strain (HARP MRI) as measured by cMRI (6 to 12 months).

  4. Change in Left Ventricular End‐Diastolic Volume Index (LVEDVI) as measured by cMRI (6 to 12 months).

  5. Change in Left Ventricular End‐Systolic Volume Index (LVESVI) as measured by cMRI (6 to 12 months).

  6. Change in Left Ventricular Sphericity Index as measured by cMRI (6 to 12 months).

  7. Change in infarct/scar volume (delayed enhancement MRI) as measured by cMRI (6 to 12 months).

  8. Change in maximal oxygen consumption (VO2 max) as measured by treadmill (6 to 12 months).

  9. Change in exercise tolerance as measured by the 6‐minute walk test (6 to 12 months).

  10. Change in MLHFQ score (6 to 12 months).

  11. The difference in the incident rate of MACE (6 to 12 months).

  12. The difference in the cumulative days alive and out of hospital (6 to 12 months).

  13. Change in NT‐proBNP as measured by blood draw (6 to 12 months).

Starting date

October 2015

Contact information

The University of Texas Health Science Center, Houston, TX, United States; National Heart, Lung, and Blood Institute. Contact: Rachel Vojvodic, MPH ([email protected]); Lemuel Moye, MD, PhD ([email protected])

Notes

Planned enrolment: 144

Estimated completion date: February 2019

This study is currently recruiting participants.

NCT02503280

Trial name or title

The Transendocardial Autologous Cells (hMSC or hMSC and hCSC) in Ischemic Heart Failure Trial (TAC‐HFT II)

Methods

A phase I/II, randomised, parallel, single‐blind (participant) safety/efficacy study

Participants

Chronic ischaemic left ventricular dysfunction and HF secondary to MI:

  1. ≥ 21 and < 90 years of age.

  2. Provide written informed consent.

  3. Diagnosis of chronic ischaemic left ventricular dysfunction secondary to MI as defined by the following: Screening MRI must show an area of akinesis, dyskinesis, or severe hypokinesis associated with evidence of myocardial scarring based on delayed hyperenhancement following gadolinium infusion.

  4. Been treated with appropriate maximal medical therapy for HF or postinfarction left ventricular dysfunction. For beta‐blockade, the person must have been on a stable dose of a clinically appropriate beta‐blocker for 3 months. For angiotensin‐converting enzyme inhibition, the person must have been on a stable dose of a clinically appropriate agent for 1 month.

  5. Candidate for cardiac catheterisation.

  6. Ejection fraction ≤ 50% by gated blood pool scan, 2‐dimensional echocardiogram, cardiac MRI, or left ventriculogram within the prior 6 months and not in the setting of a recent ischaemic event.

Interventions

Treatment arm 1: Autologous hMSC: 40 million cells/mL delivered in 0.5 mL injection volumes times 10 injections for a total of 2 x 108 (200 million) hMSC.

Treatment arm 2: Autologous hMSC PLUS autologous c‐kit hCSC: mixture of 39.8 million hMSC and 0.2 million c‐kit hCSC/mL delivered in 0.5 mL injection volumes times 10 injections for a total of 1.99 x 108 (199 million) hMSC and 1 million c‐kit hCSC.

Comparator arm: Placebo (10 0.5 mL injections of phosphate‐buffered saline and 1% human serum albumin).

Outcomes

Primary outcomes:

  1. Incidence of any TE‐SAEs 1 month postcatherisation.

  2. Incidence (at 1 month postcatheterisation) of any TE‐SAEs, defined as the composite of: death, non‐fatal MI, stroke, hospitalisation for worsening HF, cardiac perforation, pericardial tamponade, sustained ventricular arrhythmias (characterised by ventricular arrhythmias lasting longer than 15 seconds or with haemodynamic compromise), or atrial fibrillation.

Secondary outcomes:

  1. Treatment emergent adverse event rates (6 and 12 months).

  2. Ectopic tissue formation (6 and 12 months).

  3. 48‐hour ambulatory electrocardiogram recordings (6 and 12 months).

  4. Urinalysis results changes postcatheterisation (6 and 12 months).

  5. Clinical chemistry values postcatheterisation (6 and 12 months).

  6. Pulmonary function ‐ forced expiratory volume in 1 second (FEV1) results.

  7. Serial troponin I values (every 12 hours for first 48 hours post‐cardiac catheterisation).

  8. Creatine kinase‐MB values (every 12 hours for first 48 hours post‐cardiac catheterisation).

  9. Post‐cardiac catheterisation echocardiogram (6 and 12 months).

  10. MRI measures of infarct scar size (6 and 12 months).

  11. Echocardiographic measures of infarct scar size (6 and 12 months).

  12. Left regional and global ventricular function (6 and 12 months).

  13. Global ventricular function (6 and 12 months).

  14. Tissue perfusion measured by MRI (6 and 12 months).

  15. Peak oxygen consumption (peak VO2) (by treadmill determination) (6 and 12 months).

  16. 6‐minute walk test (6 and 12 months).

  17. NYHA Functional Class (6 and 12 months).

  18. MLHFQ (6 and 12 months).

  19. Incidence of MACE (6 and 12 months).

  20. Incidence of MACE, defined as the composite incidence of death, hospitalisation for worsening HF, or non‐fatal recurrent MI (6 and 12 months).

Starting date

March 2020

Contact information

ISCI/University of Miami Miller School of Medicine, Miami, Florida, United States, 33136. Principal Investigator: Joshua M Hare, MD. Contact: Darcy L DiFede ([email protected])

Notes

Planned enrolment: 55

Estimated completion date: March 2030

This study is not yet open for participant recruitment.

NCT02504437

Trial name or title

Therapy of Preconditioned Autologous BMMSCs for Patients With Ischemic Heart Disease (TPAABPIHD)

Methods

A phase I/II, randomised, parallel, double‐blind (participant, outcome assessor) safety/efficacy study

Participants

IHD:

  1. Under 75 years of age.

  2. Clinical diagnosis of AMI, chronic MI, and ischaemic cardiomyopathy.

  3. NYHA grade III‐IV, LVEF 25% to 50%.

  4. No infection diseases including hepatitis B virus, hepatitis C virus, syphilis, and AIDS.

  5. No psychiatric illnesses and speaking dysfunction.

  6. Informed consent.

Interventions

Intervention arm 1: Autologous BMMSC with hypoxia pre‐condition and endothelial pre‐induction.

Intervention arm 2: Autologous BMMSC without pre‐condition.

Comparator: Standard therapy without autologous BMMSC infusion.

Outcomes

Primary outcome: LVEF at 12 months.

Secondary outcomes: None reported.

Starting date

November 2015

Contact information

Academy Military Medical Science, China; Sun Yat‐sen University. Contact: Xuetao Pei, MD, PhD ([email protected]); Junnian Zhou, PhD ([email protected])

Notes

Planned enrolment: 200

Estimated completion date: December 2017

This study is not yet open for participant recruitment.

ABM MNC: autologous bone marrow mononuclear cell
ACE: angiotensin converting enzyme
AEs: adverse events
ACC/AHA: American College of Cardiology/American Heart Association
AICD: automatic implantable cardioverter defibrillator
AMI: acute myocardial infarction
AOI:
ARB: angiotensin receptor blocker
AST: aspartate transaminase
BMAC: bone marrow aspirate concentrate
BMMNC: bone marrow mononuclear cells
BMMSC: bone marrow mesenchymal stem cells
BMSC: bone marrow stem cells
BNP: brain natriuretic peptide
CABG: coronary artery bypass grafting
CCS: Canadian Cardiovascular Society
CHF: congestive heart failure
CMR: cardiac magnetic resonance
cMRI: cardiovascular magnetic resonance imaging
CPC: circulating progenitor cells
CSC: cardiac stem cells
DMSO: dimethyl sulfoxide
ECG: electrocardiogram
EDTA: ethylene‐diamine‐tetraacetic acid
EF: ejection fraction
EPC: endothelial progenitor cells
FDA: US Food and Drug Administration
G‐CSF: granulocyte‐colony stimulating factor
HARP: harmonic phase
HcG: human chorionic gonadotrophin
hCSC: human cardiac stem cells
HF: heart failure
hMSC: human mesenchymal stem cells
HSA: human serum albumin
IC: intracoronary
IHD: ischaemic heart disease
IM: intramuscular
INTERMACS: Interagency Registry for Mechanically Assisted Circulatory Support
IV: intravenous
LV: left ventricular
LVAD: left ventricular assist device
LVEDD: left ventricular end‐diastolic diameter
LVEDV: left ventricular end‐diastolic volume
LVEF: left ventricular ejection fraction
LVESD: left ventricular end‐systolic diameter
LVESV: left ventricular end‐systolic volume
MACE: major adverse clinical events
MI: myocardial infarction
MLHFQ: Minnesota Living with Heart Failure Questionnaire
MRI: magnetic resonance imaging
MSC: mesenchymal stem cells
MUGA: multigated radionuclide angiography
MVO₂: myocardial oxygen consumption
NT‐proBNP: N‐terminal pro b‐type natriuretic peptide
NYHA: New York Heart Association
PCI: percutaneous coronary intervention
QOL: quality of life
PET: positron emission tomography
RCT: randomised controlled trial
SF‐36: 13‐Item Short Form Health Survey
SGOT: serum aspartic aminotransferase
SGPT: serum glutamate pyruvate transaminase
SPECT: single‐photon emission computed tomography
TE‐SAEs: treatment emergent serious adverse events

Data and analyses

Open in table viewer
Comparison 1. Cells versus no cells

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause) Show forest plot

37

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.1

Comparison 1 Cells versus no cells, Outcome 1 Mortality (all‐cause).

Comparison 1 Cells versus no cells, Outcome 1 Mortality (all‐cause).

1.1 Short term follow‐up (< 12 months)

33

1637

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.26, 0.87]

1.2 Long term follow‐up (≥ 12 months)

21

1010

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.25, 0.58]

2 Non‐fatal myocardial infarction Show forest plot

25

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.2

Comparison 1 Cells versus no cells, Outcome 2 Non‐fatal myocardial infarction.

Comparison 1 Cells versus no cells, Outcome 2 Non‐fatal myocardial infarction.

2.1 Short term follow‐up (< 12 months)

20

881

Risk Ratio (M‐H, Random, 95% CI)

0.60 [0.17, 2.15]

2.2 Long term follow‐up (≥ 12 months)

9

461

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.17, 0.93]

3 Rehospitalisation due to heart failure Show forest plot

16

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.3

Comparison 1 Cells versus no cells, Outcome 3 Rehospitalisation due to heart failure.

Comparison 1 Cells versus no cells, Outcome 3 Rehospitalisation due to heart failure.

3.1 Short term follow‐up (< 12 months)

10

482

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.36, 1.12]

3.2 Long term follow‐up (≥ 12 months)

10

495

Risk Ratio (M‐H, Random, 95% CI)

0.62 [0.36, 1.04]

4 Arrhythmias Show forest plot

24

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.4

Comparison 1 Cells versus no cells, Outcome 4 Arrhythmias.

Comparison 1 Cells versus no cells, Outcome 4 Arrhythmias.

4.1 Short term follow‐up (< 12 months)

22

959

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.33, 1.45]

4.2 Long term follow‐up (≥ 12 months)

7

363

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.22, 0.97]

5 Composite MACE Show forest plot

9

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 1.5

Comparison 1 Cells versus no cells, Outcome 5 Composite MACE.

Comparison 1 Cells versus no cells, Outcome 5 Composite MACE.

5.1 Short term follow‐up (< 12 months)

8

288

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.18, 1.42]

5.2 Long term follow‐up (≥ 12 months)

5

201

Risk Ratio (M‐H, Random, 95% CI)

0.68 [0.41, 1.12]

6 MLHFQ: short term follow‐up (< 12 months) Show forest plot

4

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.6

Comparison 1 Cells versus no cells, Outcome 6 MLHFQ: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 6 MLHFQ: short term follow‐up (< 12 months).

6.1 Mean value at endpoint

2

125

Mean Difference (IV, Random, 95% CI)

‐29.52 [‐33.76, ‐25.27]

6.2 Mean change from baseline

2

72

Mean Difference (IV, Random, 95% CI)

‐9.07 [‐22.09, 3.95]

6.3 Combined

4

197

Mean Difference (IV, Random, 95% CI)

‐18.96 [‐31.97, ‐5.94]

7 MLHFQ: long term follow‐up (≥ 12 months) Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.7

Comparison 1 Cells versus no cells, Outcome 7 MLHFQ: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 7 MLHFQ: long term follow‐up (≥ 12 months).

7.1 Mean value at endpoint

1

82

Mean Difference (IV, Random, 95% CI)

‐36.5 [‐42.21, ‐30.79]

7.2 Mean change from baseline

2

69

Mean Difference (IV, Random, 95% CI)

‐7.63 [‐16.35, 1.09]

7.3 Combined

3

151

Mean Difference (IV, Random, 95% CI)

‐17.80 [‐39.87, 4.26]

8 Seattle Angina Questionnaire: short term follow‐up (< 12 months) Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.8

Comparison 1 Cells versus no cells, Outcome 8 Seattle Angina Questionnaire: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 8 Seattle Angina Questionnaire: short term follow‐up (< 12 months).

8.1 Mean value at endpoint

1

49

Mean Difference (IV, Random, 95% CI)

5.0 [‐3.21, 13.21]

8.2 Mean change from baseline

2

211

Mean Difference (IV, Random, 95% CI)

9.34 [2.62, 16.07]

8.3 Combined

2

211

Mean Difference (IV, Random, 95% CI)

9.34 [2.62, 16.07]

9 Angina episodes per week: short term follow‐up (< 12 months) Show forest plot

5

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.9

Comparison 1 Cells versus no cells, Outcome 9 Angina episodes per week: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 9 Angina episodes per week: short term follow‐up (< 12 months).

9.1 Mean value at endpoint

4

396

Mean Difference (IV, Random, 95% CI)

‐6.96 [‐11.99, ‐1.93]

9.2 Mean change from baseline

3

167

Mean Difference (IV, Random, 95% CI)

‐1.77 [‐14.61, 11.08]

9.3 Combined

5

428

Mean Difference (IV, Random, 95% CI)

‐5.11 [‐11.30, 1.09]

10 NYHA classification: short‐term follow‐up (< 12 months) Show forest plot

17

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.10

Comparison 1 Cells versus no cells, Outcome 10 NYHA classification: short‐term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 10 NYHA classification: short‐term follow‐up (< 12 months).

10.1 Mean value at endpoint

16

658

Mean Difference (IV, Random, 95% CI)

‐0.42 [‐0.84, ‐0.00]

10.2 Mean change from baseline

4

239

Mean Difference (IV, Random, 95% CI)

‐0.56 [‐1.49, 0.36]

10.3 Combined

17

741

Mean Difference (IV, Random, 95% CI)

‐0.44 [‐0.84, ‐0.05]

11 NYHA classification: long term follow‐up (≥ 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.11

Comparison 1 Cells versus no cells, Outcome 11 NYHA classification: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 11 NYHA classification: long term follow‐up (≥ 12 months).

11.1 Mean value at endpoint

9

346

Mean Difference (IV, Random, 95% CI)

‐0.57 [‐1.03, ‐0.10]

11.2 Mean change from baseline

1

39

Mean Difference (IV, Random, 95% CI)

‐2.2 [‐2.70, ‐1.70]

11.3 Combined

9

346

Mean Difference (IV, Random, 95% CI)

‐0.81 [‐1.23, ‐0.39]

12 CCS class: short term follow‐up (< 12 months) Show forest plot

13

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.12

Comparison 1 Cells versus no cells, Outcome 12 CCS class: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 12 CCS class: short term follow‐up (< 12 months).

12.1 Mean value at endpoint

10

486

Mean Difference (IV, Random, 95% CI)

‐0.32 [‐0.82, 0.18]

12.2 Mean change from baseline

6

318

Mean Difference (IV, Random, 95% CI)

‐0.62 [‐1.40, 0.17]

12.3 Combined

13

608

Mean Difference (IV, Random, 95% CI)

‐0.43 [‐0.92, 0.06]

13 CCS class: long term follow‐up (≥ 12 months) Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.13

Comparison 1 Cells versus no cells, Outcome 13 CCS class: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 13 CCS class: long term follow‐up (≥ 12 months).

13.1 Mean value at endpoint

3

142

Mean Difference (IV, Random, 95% CI)

‐0.58 [‐2.04, 0.88]

14 Exercise capacity: short term follow‐up (< 12 months) Show forest plot

16

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.14

Comparison 1 Cells versus no cells, Outcome 14 Exercise capacity: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 14 Exercise capacity: short term follow‐up (< 12 months).

14.1 Mean value at endpoint

11

563

Std. Mean Difference (IV, Random, 95% CI)

0.56 [0.19, 0.93]

14.2 Mean change from baseline

9

535

Std. Mean Difference (IV, Random, 95% CI)

0.33 [0.05, 0.61]

15 Exercise capacity: long term follow‐up (≥ 12 months) Show forest plot

8

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.15

Comparison 1 Cells versus no cells, Outcome 15 Exercise capacity: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 15 Exercise capacity: long term follow‐up (≥ 12 months).

15.1 Mean value at endpoint

5

178

Std. Mean Difference (IV, Random, 95% CI)

1.14 [0.04, 2.25]

15.2 Mean change from baseline

3

227

Std. Mean Difference (IV, Random, 95% CI)

0.34 [0.07, 0.62]

16 LVEF (%) measured by MRI: short term follow‐up (< 12 months) Show forest plot

12

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.16

Comparison 1 Cells versus no cells, Outcome 16 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 16 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

16.1 Mean value at endpoint

10

352

Mean Difference (IV, Random, 95% CI)

3.01 [‐0.05, 6.07]

16.2 Mean change from baseline

9

308

Mean Difference (IV, Random, 95% CI)

4.05 [2.55, 5.55]

16.3 Combined

12

439

Mean Difference (IV, Random, 95% CI)

2.92 [1.03, 4.82]

17 LVEF (%) measured by MRI: long term follow‐up (≥ 12 months) Show forest plot

4

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.17

Comparison 1 Cells versus no cells, Outcome 17 LVEF (%) measured by MRI: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 17 LVEF (%) measured by MRI: long term follow‐up (≥ 12 months).

17.1 Mean value at endpoint

4

110

Mean Difference (IV, Random, 95% CI)

2.37 [‐1.54, 6.29]

17.2 Mean change from baseline

3

97

Mean Difference (IV, Random, 95% CI)

3.83 [‐0.42, 8.08]

17.3 Combined

4

110

Mean Difference (IV, Random, 95% CI)

4.38 [0.82, 7.93]

18 LVEF (%) measured by echocardiography: short term follow‐up (< 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.18

Comparison 1 Cells versus no cells, Outcome 18 LVEF (%) measured by echocardiography: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 18 LVEF (%) measured by echocardiography: short term follow‐up (< 12 months).

18.1 Mean value at endpoint

8

388

Mean Difference (IV, Random, 95% CI)

5.16 [2.87, 7.44]

18.2 Mean change from baseline

3

161

Mean Difference (IV, Random, 95% CI)

3.47 [1.59, 5.34]

18.3 Combined

9

470

Mean Difference (IV, Random, 95% CI)

5.71 [4.29, 7.13]

19 LVEF (%) measured by echocardiography: long term follow‐up (≥ 12 months) Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.19

Comparison 1 Cells versus no cells, Outcome 19 LVEF (%) measured by echocardiography: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 19 LVEF (%) measured by echocardiography: long term follow‐up (≥ 12 months).

19.1 Mean value at endpoint

3

154

Mean Difference (IV, Random, 95% CI)

7.69 [6.47, 8.92]

19.2 Mean change from baseline

1

82

Mean Difference (IV, Random, 95% CI)

6.1 [‐1.27, 13.47]

19.3 Combined

3

154

Mean Difference (IV, Random, 95% CI)

7.96 [6.39, 9.54]

20 LVEF (%) measured by SPECT: short term follow‐up (< 12 months) Show forest plot

4

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.20

Comparison 1 Cells versus no cells, Outcome 20 LVEF (%) measured by SPECT: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 20 LVEF (%) measured by SPECT: short term follow‐up (< 12 months).

20.1 Mean value at endpoint

4

145

Mean Difference (IV, Random, 95% CI)

2.41 [‐2.65, 7.46]

20.2 Mean change from baseline

1

30

Mean Difference (IV, Random, 95% CI)

‐2.3 [‐17.33, 12.73]

20.3 Combined

4

145

Mean Difference (IV, Random, 95% CI)

5.22 [2.60, 7.85]

21 LVEF (%) measured by SPECT: long term follow‐up (≥ 12 months) Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.21

Comparison 1 Cells versus no cells, Outcome 21 LVEF (%) measured by SPECT: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 21 LVEF (%) measured by SPECT: long term follow‐up (≥ 12 months).

21.1 Mean value at endpoint

2

88

Mean Difference (IV, Random, 95% CI)

0.37 [‐2.30, 3.04]

21.2 Mean change from baseline

1

49

Mean Difference (IV, Random, 95% CI)

4.0 [‐6.48, 14.48]

21.3 Combined

2

88

Mean Difference (IV, Random, 95% CI)

0.28 [‐2.48, 3.03]

22 LVEF (%) measured by LV angiography: short term follow‐up (< 12 months) Show forest plot

6

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.22

Comparison 1 Cells versus no cells, Outcome 22 LVEF (%) measured by LV angiography: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 22 LVEF (%) measured by LV angiography: short term follow‐up (< 12 months).

22.1 Mean value at endpoint

6

265

Mean Difference (IV, Random, 95% CI)

3.18 [0.39, 5.97]

22.2 Mean change from baseline

4

181

Mean Difference (IV, Random, 95% CI)

1.72 [0.50, 2.95]

22.3 Combined

6

250

Mean Difference (IV, Random, 95% CI)

2.00 [0.53, 3.46]

23 LVEF (%) measured by LV angiography: long term follow‐up (≥ 12 months) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 1.23

Comparison 1 Cells versus no cells, Outcome 23 LVEF (%) measured by LV angiography: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 23 LVEF (%) measured by LV angiography: long term follow‐up (≥ 12 months).

23.1 Mean value at endpoint

1

49

Mean Difference (IV, Random, 95% CI)

6.0 [0.81, 11.19]

Open in table viewer
Comparison 2. Cell dose: subgroup analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause): short term follow‐up (< 12 months) Show forest plot

30

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.1

Comparison 2 Cell dose: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

Comparison 2 Cell dose: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

1.1 < 107 cells

6

334

Risk Ratio (M‐H, Random, 95% CI)

0.18 [0.02, 1.63]

1.2 107 < 108 cells

18

771

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.15, 0.79]

1.3 ≥ 108 cells

8

487

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.35, 1.94]

2 Mortality (all‐cause): long term follow‐up (≥ 12 months) Show forest plot

16

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 2.2

Comparison 2 Cell dose: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

Comparison 2 Cell dose: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

2.1 < 107 cells

4

297

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.10, 1.09]

2.2 107 < 108 cells

7

330

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.17, 0.53]

2.3 ≥ 108 cells

5

236

Risk Ratio (M‐H, Random, 95% CI)

0.62 [0.30, 1.26]

3 NYHA classification: short term follow‐up (< 12 months) Show forest plot

15

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 2.3

Comparison 2 Cell dose: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

Comparison 2 Cell dose: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

3.1 < 107 cells

4

149

Mean Difference (IV, Random, 95% CI)

‐0.29 [‐0.94, 0.36]

3.2 107 < 108 cells

8

309

Mean Difference (IV, Random, 95% CI)

‐0.65 [‐1.22, ‐0.08]

3.3 ≥ 108 cells

4

241

Mean Difference (IV, Random, 95% CI)

‐0.41 [‐0.72, ‐0.11]

4 CCS class: short term follow‐up (< 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 2.4

Comparison 2 Cell dose: subgroup analysis, Outcome 4 CCS class: short term follow‐up (< 12 months).

Comparison 2 Cell dose: subgroup analysis, Outcome 4 CCS class: short term follow‐up (< 12 months).

4.1 < 107 cells

4

288

Mean Difference (IV, Random, 95% CI)

‐0.87 [‐1.92, 0.19]

4.2 107 < 108 cells

5

160

Mean Difference (IV, Random, 95% CI)

‐0.54 [‐1.40, 0.32]

5 Exercise capacity: short term follow‐up (< 12 months) Show forest plot

10

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 2.5

Comparison 2 Cell dose: subgroup analysis, Outcome 5 Exercise capacity: short term follow‐up (< 12 months).

Comparison 2 Cell dose: subgroup analysis, Outcome 5 Exercise capacity: short term follow‐up (< 12 months).

5.1 107 < 108 cells

7

357

Std. Mean Difference (IV, Random, 95% CI)

0.56 [‐0.03, 1.14]

5.2 ≥ 108 cells

3

161

Std. Mean Difference (IV, Random, 95% CI)

0.43 [0.10, 0.77]

6 LVEF (%) measured by MRI: short term follow‐up (< 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 2.6

Comparison 2 Cell dose: subgroup analysis, Outcome 6 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

Comparison 2 Cell dose: subgroup analysis, Outcome 6 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

6.1 107 < 108 cells

7

199

Mean Difference (IV, Random, 95% CI)

5.23 [3.91, 6.54]

6.2 ≥ 108 cells

3

101

Mean Difference (IV, Random, 95% CI)

2.37 [‐0.92, 5.66]

Open in table viewer
Comparison 3. Baseline cardiac function: subgroup analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause): short term follow‐up (< 12 months) Show forest plot

28

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.1

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

1.1 < 30%

11

508

Risk Ratio (M‐H, Random, 95% CI)

0.23 [0.09, 0.59]

1.2 30 ‐ 50%

13

642

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.36, 2.11]

1.3 > 50%

4

271

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.11, 3.35]

2 Mortality (all‐cause): long term follow‐up (≥ 12 months) Show forest plot

16

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 3.2

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

2.1 < 30%

9

426

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.20, 0.64]

2.2 30 ‐ 50%

7

289

Risk Ratio (M‐H, Random, 95% CI)

0.57 [0.27, 1.21]

3 NYHA classification: short term follow‐up (< 12 months) Show forest plot

15

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 3.3

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

3.1 < 30%

6

273

Mean Difference (IV, Random, 95% CI)

‐0.40 [‐1.22, 0.43]

3.2 30 ‐ 50%

9

420

Mean Difference (IV, Random, 95% CI)

‐0.32 [‐0.54, ‐0.10]

4 NYHA classification: long term follow‐up (≥ 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 3.4

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 4 NYHA classification: long term follow‐up (≥ 12 months).

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 4 NYHA classification: long term follow‐up (≥ 12 months).

4.1 < 30%

5

202

Mean Difference (IV, Random, 95% CI)

‐0.66 [‐1.28, ‐0.04]

4.2 30 ‐ 50%

4

144

Mean Difference (IV, Random, 95% CI)

‐0.98 [‐1.72, ‐0.25]

5 CCS class: short term follow‐up (< 12 months) Show forest plot

8

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 3.5

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 5 CCS class: short term follow‐up (< 12 months).

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 5 CCS class: short term follow‐up (< 12 months).

5.1 < 30%

4

213

Mean Difference (IV, Random, 95% CI)

‐0.25 [‐1.47, 0.97]

5.2 30 ‐ 50%

4

150

Mean Difference (IV, Random, 95% CI)

‐0.11 [‐0.31, 0.09]

6 Exercise capacity: short term follow‐up (< 12 months) Show forest plot

7

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 3.6

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 6 Exercise capacity: short term follow‐up (< 12 months).

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 6 Exercise capacity: short term follow‐up (< 12 months).

6.1 < 30%

4

225

Std. Mean Difference (IV, Random, 95% CI)

0.96 [0.37, 1.56]

6.2 30 ‐ 50%

3

127

Std. Mean Difference (IV, Random, 95% CI)

0.38 [‐0.57, 1.33]

7 LVEF (%) measured by MRI: short term follow‐up (< 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 3.7

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 7 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 7 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

7.1 < 30%

6

290

Mean Difference (IV, Random, 95% CI)

1.54 [‐1.96, 5.03]

7.2 30 ‐ 50%

3

60

Mean Difference (IV, Random, 95% CI)

3.31 [0.88, 5.75]

Open in table viewer
Comparison 4. Route of cell administration: subgroup analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause): short term follow‐up (< 12 months) Show forest plot

33

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.1

Comparison 4 Route of cell administration: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

Comparison 4 Route of cell administration: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

1.1 Intramyocardial

22

1049

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.21, 1.03]

1.2 Intracoronary

12

607

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.21, 1.23]

2 Mortality (all‐cause): long term follow‐up (≥ 12 months) Show forest plot

21

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 4.2

Comparison 4 Route of cell administration: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

Comparison 4 Route of cell administration: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

2.1 Intramyocardial

13

652

Risk Ratio (M‐H, Random, 95% CI)

0.29 [0.17, 0.50]

2.2 Intracoronary

8

358

Risk Ratio (M‐H, Random, 95% CI)

0.57 [0.30, 1.09]

3 NYHA classification: short term follow‐up (< 12 months) Show forest plot

17

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 4.3

Comparison 4 Route of cell administration: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

Comparison 4 Route of cell administration: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

3.1 Intramyocardial

11

445

Mean Difference (IV, Random, 95% CI)

‐0.48 [‐0.99, 0.03]

3.2 Intracoronary

6

296

Mean Difference (IV, Random, 95% CI)

‐0.38 [‐0.76, 0.00]

4 NYHA classification: long term follow‐up (≥ 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 4.4

Comparison 4 Route of cell administration: subgroup analysis, Outcome 4 NYHA classification: long term follow‐up (≥ 12 months).

Comparison 4 Route of cell administration: subgroup analysis, Outcome 4 NYHA classification: long term follow‐up (≥ 12 months).

4.1 Intramyocardial

4

181

Mean Difference (IV, Random, 95% CI)

‐1.09 [‐1.76, ‐0.41]

4.2 Intracoronary

5

165

Mean Difference (IV, Random, 95% CI)

‐0.61 [‐0.92, ‐0.30]

5 CCS class: short term follow‐up (< 12 months) Show forest plot

13

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 4.5

Comparison 4 Route of cell administration: subgroup analysis, Outcome 5 CCS class: short term follow‐up (< 12 months).

Comparison 4 Route of cell administration: subgroup analysis, Outcome 5 CCS class: short term follow‐up (< 12 months).

5.1 Intramyocardial

10

434

Mean Difference (IV, Random, 95% CI)

‐0.33 [‐0.87, 0.22]

5.2 Intracoronary

3

174

Mean Difference (IV, Random, 95% CI)

‐1.00 [‐2.87, 0.86]

6 Exercise capacity: short term follow‐up (< 12 months) Show forest plot

11

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 4.6

Comparison 4 Route of cell administration: subgroup analysis, Outcome 6 Exercise capacity: short term follow‐up (< 12 months).

Comparison 4 Route of cell administration: subgroup analysis, Outcome 6 Exercise capacity: short term follow‐up (< 12 months).

6.1 Intramyocardial

6

310

Std. Mean Difference (IV, Random, 95% CI)

0.78 [0.19, 1.36]

6.2 Intracoronary

5

253

Std. Mean Difference (IV, Random, 95% CI)

0.33 [‐0.06, 0.72]

7 LVEF (%) measured by MRI: short term follow‐up (< 12 months) Show forest plot

12

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 4.7

Comparison 4 Route of cell administration: subgroup analysis, Outcome 7 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

Comparison 4 Route of cell administration: subgroup analysis, Outcome 7 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

7.1 Intramyocardial

8

309

Mean Difference (IV, Random, 95% CI)

2.18 [‐0.41, 4.77]

7.2 Intracoronary

5

137

Mean Difference (IV, Random, 95% CI)

3.72 [0.86, 6.57]

Open in table viewer
Comparison 5. Cell type: subgroup analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause): short term follow‐up (< 12 months) Show forest plot

33

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.1

Comparison 5 Cell type: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

Comparison 5 Cell type: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

1.1 Mononuclear cells

20

966

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.28, 1.04]

1.2 Circulating progenitor cells

3

104

Risk Ratio (M‐H, Random, 95% CI)

0.32 [0.01, 7.48]

1.3 Haematopoietic progenitor cells

8

464

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.05, 1.46]

1.4 Mesenchymal stem cells

3

126

Risk Ratio (M‐H, Random, 95% CI)

0.5 [0.03, 7.59]

2 Mortality (all‐cause): long term follow‐up (≥ 12 months) Show forest plot

19

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 5.2

Comparison 5 Cell type: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

Comparison 5 Cell type: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

2.1 Mononuclear cells

12

540

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.25, 0.70]

2.2 Haematopoietic progenitor cells

4

302

Risk Ratio (M‐H, Random, 95% CI)

0.26 [0.10, 0.69]

2.3 Mesenchymal stem cells

3

111

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.15, 1.57]

3 NYHA classification: short term follow‐up (< 12 months) Show forest plot

15

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 5.3

Comparison 5 Cell type: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

Comparison 5 Cell type: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

3.1 Mononuclear cells

12

547

Mean Difference (IV, Random, 95% CI)

‐0.42 [‐0.86, 0.02]

3.2 Haematopoietic progenitor cells

3

94

Mean Difference (IV, Random, 95% CI)

‐0.47 [‐1.95, 1.02]

4 CCS class: short term follow‐up (< 12 months) Show forest plot

13

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 5.4

Comparison 5 Cell type: subgroup analysis, Outcome 4 CCS class: short term follow‐up (< 12 months).

Comparison 5 Cell type: subgroup analysis, Outcome 4 CCS class: short term follow‐up (< 12 months).

4.1 Mononuclear cells

8

366

Mean Difference (IV, Random, 95% CI)

‐0.39 [‐0.99, 0.21]

4.2 Haematopoietic progenitor cells

5

242

Mean Difference (IV, Random, 95% CI)

‐0.54 [‐1.55, 0.46]

Open in table viewer
Comparison 6. Participant diagnosis: subgroup analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause): short term follow‐up (< 12 months) Show forest plot

33

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 6.1

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

1.1 Chronic IHD

11

550

Risk Ratio (M‐H, Random, 95% CI)

0.65 [0.26, 1.62]

1.2 HF (secondary to IHD)

15

645

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.14, 0.82]

1.3 Refractory/intractable angina

7

442

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.11, 3.35]

2 Mortality (all‐cause): long term follow‐up (≥ 12 months) Show forest plot

21

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 6.2

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

2.1 Chronic IHD

9

389

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.27, 0.99]

2.2 HF (secondary to IHD)

9

401

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.19, 0.58]

2.3 Refractory/intractable angina

3

220

Risk Ratio (M‐H, Random, 95% CI)

0.11 [0.01, 0.91]

3 NYHA classification: short term follow‐up (< 12 months) Show forest plot

16

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 6.3

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

3.1 Chronic IHD

6

296

Mean Difference (IV, Random, 95% CI)

‐0.43 [‐0.78, ‐0.07]

3.2 HF (secondary to IHD)

10

417

Mean Difference (IV, Random, 95% CI)

‐0.47 [‐1.02, 0.09]

4 NYHA classification: long term follow‐up (≥ 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 6.4

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 4 NYHA classification: long term follow‐up (≥ 12 months).

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 4 NYHA classification: long term follow‐up (≥ 12 months).

4.1 Chronic IHD

3

105

Mean Difference (IV, Random, 95% CI)

‐0.66 [‐0.91, ‐0.42]

4.2 HF (secondary to IHD)

6

241

Mean Difference (IV, Random, 95% CI)

‐0.92 [‐1.47, ‐0.37]

5 CCS class: short term follow‐up (< 12 months) Show forest plot

13

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 6.5

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 5 CCS class: short term follow‐up (< 12 months).

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 5 CCS class: short term follow‐up (< 12 months).

5.1 HF (secondary to IHD)

8

363

Mean Difference (IV, Random, 95% CI)

‐0.25 [‐0.90, 0.40]

5.2 Refractory/intractable angina

5

245

Mean Difference (IV, Random, 95% CI)

‐0.78 [‐1.44, ‐0.11]

6 Exercise capacity: short term follow‐up (< 12 months) Show forest plot

11

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 6.6

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 6 Exercise capacity: short term follow‐up (< 12 months).

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 6 Exercise capacity: short term follow‐up (< 12 months).

6.1 Chronic IHD

4

114

Std. Mean Difference (IV, Random, 95% CI)

0.48 [‐0.26, 1.22]

6.2 HF (secondary to IHD)

4

260

Std. Mean Difference (IV, Random, 95% CI)

0.79 [0.04, 1.53]

6.3 Refractory/intractable angina

3

189

Std. Mean Difference (IV, Random, 95% CI)

0.26 [‐0.03, 0.55]

7 LVEF (%) measured by MRI: short term follow‐up (< 12 months) Show forest plot

10

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 6.7

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 7 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 7 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

7.1 Chronic IHD

6

178

Mean Difference (IV, Random, 95% CI)

2.58 [‐0.16, 5.31]

7.2 HF (secondary to IHD)

4

195

Mean Difference (IV, Random, 95% CI)

2.50 [‐1.97, 6.97]

Open in table viewer
Comparison 7. Co‐interventions: subgroup analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause): short term follow‐up (< 12 months) Show forest plot

33

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 7.1

Comparison 7 Co‐interventions: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

Comparison 7 Co‐interventions: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

1.1 Co‐interventions

8

432

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.32, 1.70]

1.2 No co‐interventions

25

1205

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.13, 0.72]

2 Mortality (all‐cause): long term follow‐up (≥ 12 months) Show forest plot

21

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 7.2

Comparison 7 Co‐interventions: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

Comparison 7 Co‐interventions: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

2.1 Co‐interventions

6

312

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.26, 0.88]

2.2 No co‐interventions

15

698

Risk Ratio (M‐H, Random, 95% CI)

0.32 [0.19, 0.56]

3 NYHA classification: short term follow‐up (< 12 months) Show forest plot

17

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 7.3

Comparison 7 Co‐interventions: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

Comparison 7 Co‐interventions: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

3.1 Co‐interventions

6

233

Mean Difference (IV, Random, 95% CI)

‐0.57 [‐1.20, 0.05]

3.2 No co‐interventions

11

508

Mean Difference (IV, Random, 95% CI)

‐0.37 [‐0.87, 0.13]

4 LVEF (%) measured by MRI: short term follow‐up (< 12 months) Show forest plot

12

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 7.4

Comparison 7 Co‐interventions: subgroup analysis, Outcome 4 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

Comparison 7 Co‐interventions: subgroup analysis, Outcome 4 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

4.1 Co‐interventions

5

179

Mean Difference (IV, Random, 95% CI)

2.01 [‐0.26, 4.29]

4.2 No co‐interventions

7

260

Mean Difference (IV, Random, 95% CI)

3.55 [0.82, 6.27]

Open in table viewer
Comparison 8. Sensitivity analysis: excluding studies with high/unclear risk of selection bias

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause) Show forest plot

15

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 8.1

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 1 Mortality (all‐cause).

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 1 Mortality (all‐cause).

1.1 Short term follow‐up (< 12 months)

14

744

Risk Ratio (M‐H, Random, 95% CI)

0.69 [0.32, 1.50]

1.2 Long term follow‐up (≥ 12 months)

9

491

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.21, 0.87]

2 Non‐fatal myocardial infarction Show forest plot

11

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 8.2

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 2 Non‐fatal myocardial infarction.

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 2 Non‐fatal myocardial infarction.

2.1 Short term follow‐up (< 12 months)

6

288

Risk Ratio (M‐H, Random, 95% CI)

0.50 [0.05, 4.58]

2.2 Long term follow‐up (≥ 12 months)

5

345

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.15, 0.97]

3 Rehospitalisation due to heart failure Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 8.3

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 3 Rehospitalisation due to heart failure.

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 3 Rehospitalisation due to heart failure.

3.1 Short term follow‐up (< 12 months)

3

234

Risk Ratio (M‐H, Random, 95% CI)

0.65 [0.32, 1.32]

3.2 Long term follow‐up (≥ 12 months)

6

375

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.36, 1.09]

4 Arrhythmias Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 8.4

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 4 Arrhythmias.

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 4 Arrhythmias.

4.1 Short term follow‐up (< 12 months)

6

224

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.18, 3.21]

4.2 Long term follow‐up (≥ 12 months)

1

82

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.18, 0.99]

5 Composite MACE Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 8.5

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 5 Composite MACE.

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 5 Composite MACE.

5.1 Short term follow‐up (< 12 months)

2

59

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

5.2 Long term follow‐up (≥ 12 months)

3

141

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.38, 1.08]

6 NYHA classification: short term follow‐up (< 12 months) Show forest plot

5

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 8.6

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 6 NYHA classification: short term follow‐up (< 12 months).

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 6 NYHA classification: short term follow‐up (< 12 months).

6.1 Combined

5

277

Mean Difference (IV, Random, 95% CI)

‐0.26 [‐0.59, 0.07]

7 NYHA classification: long term follow‐up (≥ 12 months) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 8.7

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 7 NYHA classification: long term follow‐up (≥ 12 months).

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 7 NYHA classification: long term follow‐up (≥ 12 months).

7.1 Combined

1

39

Mean Difference (IV, Random, 95% CI)

‐2.2 [‐2.70, ‐1.70]

8 LVEF (%) measured by MRI: short term follow‐up (< 12 months) Show forest plot

7

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 8.8

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 8 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 8 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

8.1 Combined

7

249

Mean Difference (IV, Random, 95% CI)

2.92 [0.67, 5.17]

9 LVEF (%) measured by MRI: long term follow‐up (≥ 12 months) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

Analysis 8.9

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 9 LVEF (%) measured by MRI: long term follow‐up (≥ 12 months).

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 9 LVEF (%) measured by MRI: long term follow‐up (≥ 12 months).

9.1 Combined

1

25

Mean Difference (IV, Random, 95% CI)

‐1.60 [‐8.70, 5.50]

Open in table viewer
Comparison 9. Sensitivity analysis: excluding studies with high/unclear risk of performance bias

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause) Show forest plot

26

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 9.1

Comparison 9 Sensitivity analysis: excluding studies with high/unclear risk of performance bias, Outcome 1 Mortality (all‐cause).

Comparison 9 Sensitivity analysis: excluding studies with high/unclear risk of performance bias, Outcome 1 Mortality (all‐cause).

1.1 Short term follow‐up (< 12 months)

25

1216

Risk Ratio (M‐H, Random, 95% CI)

0.58 [0.29, 1.16]

1.2 Long term follow‐up (≥ 12 months)

13

624

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.21, 0.86]

Open in table viewer
Comparison 10. Sensitivity analysis: excluding studies with high/unclear risk of attrition bias

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause) Show forest plot

32

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

Analysis 10.1

Comparison 10 Sensitivity analysis: excluding studies with high/unclear risk of attrition bias, Outcome 1 Mortality (all‐cause).

Comparison 10 Sensitivity analysis: excluding studies with high/unclear risk of attrition bias, Outcome 1 Mortality (all‐cause).

1.1 Short term follow‐up (< 12 months)

28

1449

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.26, 0.89]

1.2 Long term follow‐up (≥ 12 months)

17

883

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.25, 0.60]

PRISMA flow diagram.
Figuras y tablas -
Figure 1

PRISMA flow diagram.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 2

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Funnel plot of comparison: 1 Stem cells versus no stem cells, outcome: 1.1 Mortality.
Figuras y tablas -
Figure 3

Funnel plot of comparison: 1 Stem cells versus no stem cells, outcome: 1.1 Mortality.

Trial sequential analysis: Mortality at long‐term follow‐up (≥ 12 months). TSMB = trial sequential monitoring boundary; horizontal red lines indicate conventional significance threshold.
Figuras y tablas -
Figure 4

Trial sequential analysis: Mortality at long‐term follow‐up (≥ 12 months). TSMB = trial sequential monitoring boundary; horizontal red lines indicate conventional significance threshold.

Trial sequential analysis: Non‐fatal myocardial infarction at long‐term follow‐up (≥ 12 months). TSMB = trial sequential monitoring boundary; horizontal red lines indicate conventional significance threshold.
Figuras y tablas -
Figure 5

Trial sequential analysis: Non‐fatal myocardial infarction at long‐term follow‐up (≥ 12 months). TSMB = trial sequential monitoring boundary; horizontal red lines indicate conventional significance threshold.

Trial sequential analysis: Rehospitalisation due to heart failure at long‐term follow‐up (≥ 12 months). TSMB = trial sequential monitoring boundary; horizontal red lines indicate conventional significance threshold.
Figuras y tablas -
Figure 6

Trial sequential analysis: Rehospitalisation due to heart failure at long‐term follow‐up (≥ 12 months). TSMB = trial sequential monitoring boundary; horizontal red lines indicate conventional significance threshold.

Trial sequential analysis: Arrhythmias at long‐term follow‐up (≥ 12 months). TSMB = trial sequential monitoring boundary; horizontal red lines indicate conventional significance threshold.
Figuras y tablas -
Figure 7

Trial sequential analysis: Arrhythmias at long‐term follow‐up (≥ 12 months). TSMB = trial sequential monitoring boundary; horizontal red lines indicate conventional significance threshold.

Trial sequential analysis: Composite MACE at long‐term follow‐up (≥ 12 months). TSMB = trial sequential monitoring boundary; horizontal red lines indicate conventional significance threshold.
Figuras y tablas -
Figure 8

Trial sequential analysis: Composite MACE at long‐term follow‐up (≥ 12 months). TSMB = trial sequential monitoring boundary; horizontal red lines indicate conventional significance threshold.

Trial sequential analysis: Left ventricular ejection fraction measured by MRI at long‐term follow‐up (≥ 12 months). TSMB = trial sequential monitoring boundary; horizontal red lines indicate conventional significance threshold.
Figuras y tablas -
Figure 9

Trial sequential analysis: Left ventricular ejection fraction measured by MRI at long‐term follow‐up (≥ 12 months). TSMB = trial sequential monitoring boundary; horizontal red lines indicate conventional significance threshold.

Comparison 1 Cells versus no cells, Outcome 1 Mortality (all‐cause).
Figuras y tablas -
Analysis 1.1

Comparison 1 Cells versus no cells, Outcome 1 Mortality (all‐cause).

Comparison 1 Cells versus no cells, Outcome 2 Non‐fatal myocardial infarction.
Figuras y tablas -
Analysis 1.2

Comparison 1 Cells versus no cells, Outcome 2 Non‐fatal myocardial infarction.

Comparison 1 Cells versus no cells, Outcome 3 Rehospitalisation due to heart failure.
Figuras y tablas -
Analysis 1.3

Comparison 1 Cells versus no cells, Outcome 3 Rehospitalisation due to heart failure.

Comparison 1 Cells versus no cells, Outcome 4 Arrhythmias.
Figuras y tablas -
Analysis 1.4

Comparison 1 Cells versus no cells, Outcome 4 Arrhythmias.

Comparison 1 Cells versus no cells, Outcome 5 Composite MACE.
Figuras y tablas -
Analysis 1.5

Comparison 1 Cells versus no cells, Outcome 5 Composite MACE.

Comparison 1 Cells versus no cells, Outcome 6 MLHFQ: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 1.6

Comparison 1 Cells versus no cells, Outcome 6 MLHFQ: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 7 MLHFQ: long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 1.7

Comparison 1 Cells versus no cells, Outcome 7 MLHFQ: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 8 Seattle Angina Questionnaire: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 1.8

Comparison 1 Cells versus no cells, Outcome 8 Seattle Angina Questionnaire: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 9 Angina episodes per week: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 1.9

Comparison 1 Cells versus no cells, Outcome 9 Angina episodes per week: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 10 NYHA classification: short‐term follow‐up (< 12 months).
Figuras y tablas -
Analysis 1.10

Comparison 1 Cells versus no cells, Outcome 10 NYHA classification: short‐term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 11 NYHA classification: long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 1.11

Comparison 1 Cells versus no cells, Outcome 11 NYHA classification: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 12 CCS class: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 1.12

Comparison 1 Cells versus no cells, Outcome 12 CCS class: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 13 CCS class: long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 1.13

Comparison 1 Cells versus no cells, Outcome 13 CCS class: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 14 Exercise capacity: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 1.14

Comparison 1 Cells versus no cells, Outcome 14 Exercise capacity: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 15 Exercise capacity: long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 1.15

Comparison 1 Cells versus no cells, Outcome 15 Exercise capacity: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 16 LVEF (%) measured by MRI: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 1.16

Comparison 1 Cells versus no cells, Outcome 16 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 17 LVEF (%) measured by MRI: long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 1.17

Comparison 1 Cells versus no cells, Outcome 17 LVEF (%) measured by MRI: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 18 LVEF (%) measured by echocardiography: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 1.18

Comparison 1 Cells versus no cells, Outcome 18 LVEF (%) measured by echocardiography: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 19 LVEF (%) measured by echocardiography: long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 1.19

Comparison 1 Cells versus no cells, Outcome 19 LVEF (%) measured by echocardiography: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 20 LVEF (%) measured by SPECT: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 1.20

Comparison 1 Cells versus no cells, Outcome 20 LVEF (%) measured by SPECT: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 21 LVEF (%) measured by SPECT: long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 1.21

Comparison 1 Cells versus no cells, Outcome 21 LVEF (%) measured by SPECT: long term follow‐up (≥ 12 months).

Comparison 1 Cells versus no cells, Outcome 22 LVEF (%) measured by LV angiography: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 1.22

Comparison 1 Cells versus no cells, Outcome 22 LVEF (%) measured by LV angiography: short term follow‐up (< 12 months).

Comparison 1 Cells versus no cells, Outcome 23 LVEF (%) measured by LV angiography: long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 1.23

Comparison 1 Cells versus no cells, Outcome 23 LVEF (%) measured by LV angiography: long term follow‐up (≥ 12 months).

Comparison 2 Cell dose: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 2.1

Comparison 2 Cell dose: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

Comparison 2 Cell dose: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 2.2

Comparison 2 Cell dose: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

Comparison 2 Cell dose: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 2.3

Comparison 2 Cell dose: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

Comparison 2 Cell dose: subgroup analysis, Outcome 4 CCS class: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 2.4

Comparison 2 Cell dose: subgroup analysis, Outcome 4 CCS class: short term follow‐up (< 12 months).

Comparison 2 Cell dose: subgroup analysis, Outcome 5 Exercise capacity: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 2.5

Comparison 2 Cell dose: subgroup analysis, Outcome 5 Exercise capacity: short term follow‐up (< 12 months).

Comparison 2 Cell dose: subgroup analysis, Outcome 6 LVEF (%) measured by MRI: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 2.6

Comparison 2 Cell dose: subgroup analysis, Outcome 6 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 3.1

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 3.2

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 3.3

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 4 NYHA classification: long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 3.4

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 4 NYHA classification: long term follow‐up (≥ 12 months).

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 5 CCS class: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 3.5

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 5 CCS class: short term follow‐up (< 12 months).

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 6 Exercise capacity: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 3.6

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 6 Exercise capacity: short term follow‐up (< 12 months).

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 7 LVEF (%) measured by MRI: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 3.7

Comparison 3 Baseline cardiac function: subgroup analysis, Outcome 7 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

Comparison 4 Route of cell administration: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 4.1

Comparison 4 Route of cell administration: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

Comparison 4 Route of cell administration: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 4.2

Comparison 4 Route of cell administration: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

Comparison 4 Route of cell administration: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 4.3

Comparison 4 Route of cell administration: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

Comparison 4 Route of cell administration: subgroup analysis, Outcome 4 NYHA classification: long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 4.4

Comparison 4 Route of cell administration: subgroup analysis, Outcome 4 NYHA classification: long term follow‐up (≥ 12 months).

Comparison 4 Route of cell administration: subgroup analysis, Outcome 5 CCS class: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 4.5

Comparison 4 Route of cell administration: subgroup analysis, Outcome 5 CCS class: short term follow‐up (< 12 months).

Comparison 4 Route of cell administration: subgroup analysis, Outcome 6 Exercise capacity: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 4.6

Comparison 4 Route of cell administration: subgroup analysis, Outcome 6 Exercise capacity: short term follow‐up (< 12 months).

Comparison 4 Route of cell administration: subgroup analysis, Outcome 7 LVEF (%) measured by MRI: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 4.7

Comparison 4 Route of cell administration: subgroup analysis, Outcome 7 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

Comparison 5 Cell type: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 5.1

Comparison 5 Cell type: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

Comparison 5 Cell type: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 5.2

Comparison 5 Cell type: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

Comparison 5 Cell type: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 5.3

Comparison 5 Cell type: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

Comparison 5 Cell type: subgroup analysis, Outcome 4 CCS class: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 5.4

Comparison 5 Cell type: subgroup analysis, Outcome 4 CCS class: short term follow‐up (< 12 months).

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 6.1

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 6.2

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 6.3

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 4 NYHA classification: long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 6.4

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 4 NYHA classification: long term follow‐up (≥ 12 months).

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 5 CCS class: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 6.5

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 5 CCS class: short term follow‐up (< 12 months).

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 6 Exercise capacity: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 6.6

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 6 Exercise capacity: short term follow‐up (< 12 months).

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 7 LVEF (%) measured by MRI: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 6.7

Comparison 6 Participant diagnosis: subgroup analysis, Outcome 7 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

Comparison 7 Co‐interventions: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 7.1

Comparison 7 Co‐interventions: subgroup analysis, Outcome 1 Mortality (all‐cause): short term follow‐up (< 12 months).

Comparison 7 Co‐interventions: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 7.2

Comparison 7 Co‐interventions: subgroup analysis, Outcome 2 Mortality (all‐cause): long term follow‐up (≥ 12 months).

Comparison 7 Co‐interventions: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 7.3

Comparison 7 Co‐interventions: subgroup analysis, Outcome 3 NYHA classification: short term follow‐up (< 12 months).

Comparison 7 Co‐interventions: subgroup analysis, Outcome 4 LVEF (%) measured by MRI: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 7.4

Comparison 7 Co‐interventions: subgroup analysis, Outcome 4 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 1 Mortality (all‐cause).
Figuras y tablas -
Analysis 8.1

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 1 Mortality (all‐cause).

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 2 Non‐fatal myocardial infarction.
Figuras y tablas -
Analysis 8.2

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 2 Non‐fatal myocardial infarction.

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 3 Rehospitalisation due to heart failure.
Figuras y tablas -
Analysis 8.3

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 3 Rehospitalisation due to heart failure.

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 4 Arrhythmias.
Figuras y tablas -
Analysis 8.4

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 4 Arrhythmias.

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 5 Composite MACE.
Figuras y tablas -
Analysis 8.5

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 5 Composite MACE.

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 6 NYHA classification: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 8.6

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 6 NYHA classification: short term follow‐up (< 12 months).

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 7 NYHA classification: long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 8.7

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 7 NYHA classification: long term follow‐up (≥ 12 months).

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 8 LVEF (%) measured by MRI: short term follow‐up (< 12 months).
Figuras y tablas -
Analysis 8.8

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 8 LVEF (%) measured by MRI: short term follow‐up (< 12 months).

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 9 LVEF (%) measured by MRI: long term follow‐up (≥ 12 months).
Figuras y tablas -
Analysis 8.9

Comparison 8 Sensitivity analysis: excluding studies with high/unclear risk of selection bias, Outcome 9 LVEF (%) measured by MRI: long term follow‐up (≥ 12 months).

Comparison 9 Sensitivity analysis: excluding studies with high/unclear risk of performance bias, Outcome 1 Mortality (all‐cause).
Figuras y tablas -
Analysis 9.1

Comparison 9 Sensitivity analysis: excluding studies with high/unclear risk of performance bias, Outcome 1 Mortality (all‐cause).

Comparison 10 Sensitivity analysis: excluding studies with high/unclear risk of attrition bias, Outcome 1 Mortality (all‐cause).
Figuras y tablas -
Analysis 10.1

Comparison 10 Sensitivity analysis: excluding studies with high/unclear risk of attrition bias, Outcome 1 Mortality (all‐cause).

Summary of findings for the main comparison. Bone marrow‐derived cell therapy for people with chronic ischaemic heart disease and congestive heart failure

Bone marrow‐derived cell therapy for people with chronic ischaemic heart disease and congestive heart failure

Patient or population: people with chronic ischaemic heart disease and congestive heart failure
Settings: hospitalisation
Intervention: bone marrow‐derived cell therapy

Comparison: no cell therapy

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect
(95% CI)

No of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk

Corresponding risk

No cell therapy

Bone marrow‐derived cell therapy

Mortality (all cause)

Long‐term follow‐up (≥ 12 months)

102 per 1000

43 per 1000
(21 to 89)

RR 0.42
(0.21 to 0.87)

491
(9 studies)

⊕⊕⊝⊝
low1,2

The required information size of 1899 participants to detect a RRR of 35% has not been reached.

Periprocedural adverse events

See comment

See comment

Not estimable

1695

(34 studies)

See comment

Adverse events occurring during the mapping or cell/placebo injection procedure included ventricular tachycardia (7), ventricular fibrillation (1), atrial fibrillation (1), transient complete heart block (1), transient pulmonary oedema (3), thrombus on mapping catheter tip (1), visual disturbances (2), myocardial perforation (2), limited retrograde catheter‐related dissection of the abdominal aorta (1).

Non‐fatal myocardial infarction

Long‐term follow‐up (≥ 12 months)

83 per 1000

31 per 1000
(12 to 80)

RR 0.38
(0.15 to 0.97)

345
(5 studies)

⊕⊕⊝⊝
low2,3

The required information size of 2383 participants to detect a RRR of 35% has not been reached.

Rehospitalisation due to heart failure

Long‐term follow‐up (≥ 12 months)

155 per 1000

98 per 1000
(56 to 169)

RR 0.63
(0.36 to 1.09)

375
(6 studies)

⊕⊕⊝⊝
low2,4

The required information size of 1193 participants to detect a RRR of 35% has not been reached.

Arrhythmias

Long‐term follow‐up (≥ 12 months)

333 per 1000

140 per 1000
(60 to 330)

RR 0.42
(0.18 to 0.99)

82
(1 study)

⊕⊕⊝⊝
low5,6

The required information size of 461 participants to detect a RRR of 35% has not been reached.

Composite MACE

Long‐term follow‐up (≥ 12 months)

350 per 1000

224 per 1000
(133 to 378)

RR 0.64
(0.38 to 1.08)

141
(3 studies)

⊕⊕⊝⊝
low7,8

The required information size of 431 participants to detect a RRR of 35% has not been reached.

LVEF (%) measured by MRI

Long‐term follow‐up (≥ 12 months)

The mean LVEF (%) measured by MRI in the intervention groups was 1.6 lower (8.7 lower to 5.5 higher).

25
(1 study)

⊕⊕⊝⊝
low6,7

The required information size of 322 participants to detect a mean difference of 4% has not been reached.

*The risk in the intervention group (and its 95% confidence interval) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

Only studies with a low risk of selection bias are included.
CI: confidence interval; LVEF: left ventricular ejection fraction; MACE: major adverse clinical events; MD: mean difference; MRI: magnetic resonance imaging; NYHA: New York Heart Assocation; OR: odds ratio; RCT: randomised controlled trial; RR: risk ratio; RRR: relative risk reduction

GRADE Working Group grades of evidence
High quality: Further research is very unlikely to change our confidence in the estimate of effect.
Moderate quality: Further research is likely to have an important impact on our confidence in the estimate of effect and may change the estimate.
Low quality: Further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate.
Very low quality: We are very uncertain about the estimate.

1Six trials received full or partial commercial funding, which could have resulted in a biased assessment of the intervention effect and were therefore deemed to have a high risk of bias. One trial was not blinded (high risk of performance bias) and had a high risk of attrition bias.
2The number of observed events was low, leading to imprecision.
3Four studies received full or partial commercial funding with a high risk of bias.
4Five trials received full or partial commercial funding with a high risk of bias.
5The included trial received partial commercial funding with a high risk of bias.
6Only one trial with a low number of observed events was included in the analysis, leading to imprecision.
7All three included trials received partial commercial funding with a high risk of bias.
8The number of included studies was low with a low number of observed events, leading to imprecision.

Figuras y tablas -
Summary of findings for the main comparison. Bone marrow‐derived cell therapy for people with chronic ischaemic heart disease and congestive heart failure
Table 1. Characteristics of study participants

Study ID

Country of study

Patient population

Mean (SD) age of participants (years)

% Male

No. randomised participants receiving intervention

No. randomised participants receiving comparator

Mean duration of follow‐up

Ang 2008

UK

CIHD (> 1 chronic myocardial scar; elective CABG)

BMMNC‐IM: 64.7 (8.7)

BMMNC‐IC: 62.1 (8.7)

Controls: 61.3 (8.3)

BMMNC‐IM: 71.4%

BMMNC‐IC: 90.5%

Controls: 90.0%

42 (21 IM, 21 IC)

21

6 months

Assmus 2006

Germany

CIHD (MI > 3 months; LV dysfunction)

BMMNC: 59 (12)

CPC: 54 (12)

Controls: 61 (9)

BMMNC: 89%

CPC: 79%

Controls: 100%

52 (28 MNC, 24 CPC)

23

3 months

Assmus 2013

Germany

CIHD (MI > 3 months; LVEF < 50%; NYHA class II or greater)

BMMNC‐LDSW: 65 (12)

BMMNC‐HDSW: 58 (11)

Controls‐LDSW: 60 (10)

Controls‐HDSW: 63 (10)

BMMNC‐LDSW: 77%

BMMNC‐HDSW: 86%

Controls‐LDSW: 80%

Controls‐HDSW: 90%

43 (22 LDSW, 21 HDSW)

39 (20 LDSW, 19 HDSW)

45.7 (17) months

Bartunek 2012

Belgium/

Serbia/

Switzerland

HF (LVEF 15% to 40%; ischaemic event > 2 months)

BM‐MSC: 55.3 (SE 10.4)

Controls: 58.7 (SE 8.2)

BM‐MSC: 90.5%

Controls: 86.7%

32

15

24 months

Chen 2006

China

CIHD (isolated, chronic LAD; LVEF < 40%)

BM‐MSC: 59.3 (6.8)

Controls: 57.8 (7.2)

BM‐MSC: 88%

Controls: 92%

24

24

12 months

Erbs 2005

Germany

CIHD (chronic total occlusion; myocardial ischaemia)

CPC: 63 (7)

Controls: 61 (9)

CPC: 71%

Controls: 86%

14

14

15 months

Hamshere 2015_IC

UK

HF (NYHA class II‐IV; no revascularisation options)

BMMNC: n/r

Controls: n/r

BMMNC: n/r

Controls: n/r

15

15

12 months

Hamshere 2015_IM

UK

HF (NYHA class II‐IV; no revascularisation options)

BMMNC: n/r

Controls: n/r

BMMNC: n/r

Controls: n/r

15

15

12 months

Heldman 2014_BMMNC

USA

CIHD (chronic MI; LV dysfunction)

BMMNC: 61.1 (8.4)

Controls: 61.3 (9.0)

BMMNC: 89.5%

Controls: 100%

22

10

12 months

Heldman 2014_BM‐MSC

USA

CIHD (chronic MI; LV dysfunction)

BM‐MSC: 57.1 (10.6)

Controls: 60.0 (12.0)

BM‐MSC: 94.7%

Controls: 90.9%

22

11

12 months

Hendrikx 2006

Belgium

CIHD (transmural MI; LV dysfunction; elective CABG)

BMMNC: 63.2 (8.5)

Controls: 66.8 (9.2)

BMMNC: 100%

Controls: 70%

11

12

4 months

Honold 2012

Germany

CIHD (MI > 3 months; LV regional wall motion abnormality)

CPC: 53.4 (12.3)

Controls: 58.8 (7.3)

CPC: 82%

Controls: 100%

23

10

60 months

Hu 2011

China

HF (MI > 3 months; LVEF < 30%; elective CABG)

BMMNC: 56.6 (9.7)

Controls: 58.3 (8.9)

BMMNC: 88%

Controls: 96%

31

29

12 months

Jimenez‐Quevedo 2011

Spain

Refractory angina (CCS class II‐IV)

CD133+: median 70.0

Controls: median 58.2

CD133+: 78.9%

Controls: 100%

19

9

6 months

Losordo 2007

USA

Refractory angina (CCS class III‐IV)

CD34+/controls pooled: 62.4 (range 48 to 84)

CD34+/controls pooled: 80%

18 (6 LD, 6 MD 6, HD)

6

6 months

Losordo 2011

USA

Refractory angina (CCS class III‐IV)

CD34+/LD: 61.3 (9.1)

CD34+/HD: 59.8 (9.2)

Controls: 61.8 (8.5)

CD34+/LD: 83.6%

CD34+/HD: 87.5%

Controls: 89.3%

112 (56 LD, 56 HD)

56

12 months

Mathiasen 2015

Denmark

HF (NYHA class II‐III; LVEF < 45%; no revascularisation options)

BM‐MSC: 66.1 (7.7)

Controls: 64.2 (10.6)

BM‐MSC: 90%

Controls: 70%

40

20

6 months

Mozid 2014_IC

UK

HF (NYHA class II‐IV; no revascularisation options)

BMMNC/controls pooled (16 participants): 70 (10)

BMMNC/controls pooled (16 participants): 94%

14

2

6 months

Mozid 2014_IM

UK

HF (NYHA class II‐IV; no revascularisation options)

BMMNC/controls pooled (18 participants): 64 (9)

BMMNC/controls pooled (18 participants): 100%

10

8

6 months

Nasseri 2012

Germany

HF (LVEF < 35%; elective CABG)

CD133+: 61.9 (7.3)

Controls: 62.7 (10.6)

CD133+: 93%

Controls: 97%

30

30

6 months

Patel 2005

Argentina

HF (LVEF < 35%; NYHA class III‐IV; elective CABG)

CD34+: 64.8 (7.1)

Controls: 63.6 (5.2)

CD34+: 80%

Controls: 80%

25

25

10 years

Patel 2015

USA/Germany/India

HF (LVEF < 40%; NYHA class III‐IV)

BMAC: 58.5 (12.7)

Controls: 52.7 (8.5)

BMAC: 91.7%

Controls: 100%

24

6

12 months

Patila 2014

Finland

HF (LVEF 15% to 40%; NYHA class II‐IV; elective CABG)

BMMNC: median 65 (range 57 to 73)

Controls: median 64 (range 58 to 70)

BMMNC: 94.7%

Controls: 95.0%

20

19

12 months

Perin 2011

USA

HF (angina/HF symptoms; chronic CAD; LVEF < 40%; no revascularisation options)

BMMNC: 56.3 (8.6)

Controls: 60.5 (6.4)

BMMNC: 50%

Controls: 80%

20

10

6 months

Perin 2012a

USA

HF (CCS class II‐IV or NYHA class II‐III, or both; LVEF < 45%; no revascularisation options)

BMMNC: 64.0 (10.9)

Controls: 62.3 (8.3)

BMMNC: 86.9%

Controls: 93.7%

61

31

6 months

Perin 2012b

USA

HF (CCS class II‐IV or NYHA class II‐III, or both; LVEF < 45%; no revascularisation options)

ALDH+: 58.2 (6.1)

Controls: 57.8 (5.5)

ALDH+: 90%

Controls: 80%

10

10

6 months

Pokushalov 2010

Russia

HF (LVEF < 35%; no revascularisation options)

BMMNC: 61 (9)

Controls: 62 (5)

BMMNC: 87%

Controls: 85%

55

54

12 months

Santoso 2014

Indonesia/China

HF (NYHA class III‐IV; LVEF < 40%; no revascularisation options)

BMMNC: 58 (5.9)

Controls: 60 (5.6)

BMMNC: 95%

Controls: 100%

19

9

6 months

Trifunovic 2015

Serbia

CIHD (MI < 30 days; LVEF < 40%; NYHA class III‐IV; elective CABG)

BMMNC: 53.8 (10.1)

Controls: 60.0 (6.8)

BMMNC: 93.3%

Controls: 93.3%

15

15

Median 5 years (IQR 2.5 to 7.5)

Tse 2007

China/Australia

Refractory angina (CCS class III‐IV)

BMMNC: 65.2 (8.3)

Controls: 68.9 (6.3)

BMMNC: 79%

Controls: 88%

19

9

6 months

Turan 2011

Germany

CIHD (MI > 3 months; LV dysfunction)

BMMNC: 62 (10)

Controls: 60 (9)

BMMNC: 52.6%

Controls: 55.6%

38

18

12 months

Van Ramshorst 2009

The Netherlands

Refractory angina (CCS class II‐IV)

BMMNC: 64 (8)

Controls: 62 (9)

BMMNC: 92%

Controls: 80%

25

25

6 months

Wang 2009

China

Refractory angina (MI > 1 month)

CD34+: 60.6 (n/r)

Controls: 60.0 (n/r)

CD34+: 56.3%

Controls: 63.3%

16

16

6 months

Wang 2010

China

Refractory angina (CCS class III‐IV)

CD34+: range 42 to 80

Controls: range 43 to 80

CD34+: 51.8%

Controls: 50.0%

56

56

6 months

Wang 2014

China

CIHD (LVEF < 35%)

CD133+: n/r

Controls: n/r

CD133+: n/r

Controls: n/r

35

35

6 months

Wang 2015

China

CIHD (multivessel disease; MI > 4 weeks; elective CABG)

BMMNC: 61.4 (7.5)

Controls: 62.9 (6.9)

BMMNC: 82%

Controls: 78%

45

45

6 months

Yao 2008

China

CIHD (MI > 6 months)

BMMNC: 54.8 (11.5)

Controls: 56.3 (7.9)

BMMNC: 96%

Controls: 96%

24

23

6 months

Zhao 2008

China

HF (LVEF < 40%; elective CABG)

BMMNC: 60.3 (10.4)

Controls: 59.1 (15.7)

BMMNC: 83.3%

Controls: 83.3%

18

18

6 months

ALDH: aldehyde dehydrogenase
BMAC: bone marrow aspirate concentrate
BMMNC: bone marrow mononuclear cells
BM‐MSC: bone marrow‐derived mesenchymal stem cells
CABG: coronary artery bypass grafting
CCS: Canadian Cardiovascular Society
CIHD: chronic ischaemic heart disease
CPC: circulating progenitor cells
EF: ejection fraction
HD: high dose
HDSW: high dose shockwave
HF: heart failure
IC: intracoronary
IM: intramyocardial
IQR: interquartile range
LAD: left ventricular assist device
LD: low dose
LDSW: low dose shockwave
LV: left ventricular
LVEF: left ventricular ejection fraction
MD: medium dose
MI: myocardial infarction
MNC: mononuclear cells
n/r: not reported
NYHA: New York Heart Association
SD: standard deviation
SE: standard error
SW: shockwave

Figuras y tablas -
Table 1. Characteristics of study participants
Table 2. Characteristics of study interventions

Study ID

Co‐intervention

Intervention given by:

Route of cell administration

Intervention cell type

How are cells obtained?

What were they resuspended in?

Dose administered?

Comparator arm (placebo or control)

Ang 2008

CABG

Cardiothoracic surgeon

IC or IM

BMMNC

BM aspiration (**)

Autologous serum

IM: 84 (56) million cells

IC: 115 (73) million cells

No additional therapy (control)

Assmus 2006

Standard medical therapy

Cardiologist

IC

BMMNC or CPC

BM aspiration (**) for BMMNC. Vein puncture, mononuclear cell isolation by gradient centrifugation and culture for 3 days for CPC

n/r

BMMNC: 205 (110) million cells

CPC: 22 (11) million cells

No additional therapy (control)

Assmus 2013

Shockwave

Cardiologist

IC

BMMNC

BM aspiration (**)

X‐VIVO 10 medium and autologous serum

HDSW: 123 (69) million cells

LDSW: 150 (77) million cells

Placebo (10 mL X‐VIVO 10 medium and autologous serum)

Bartunek 2012

Standard medical therapy

Cardiologist

IC

BM‐MSC (cardiopoietic cells)

BM aspiration (**), culture for 6 days and exposure to cardiopoietic factors

Preservation solution (no details)

733 (range 605 to 1168) million cells

No additional therapy (control)

Chen 2006

Standard medical therapy

Cardiologist

IC

BM‐MSC

BM aspiration (**), culture for 7 days to select MSC

Heparinised saline

5 million cells

No additional therapy (control)

Erbs 2005

G‐CSF

Cardiologist

IC

CPC

G‐CSF infusion for 4 days prior to vein puncture, mononuclear cell isolation by gradient centrifugation and culture for 3 days for CPC

Saline and 10% autologous serum

69 (14) million cells

Placebo (cell‐free serum solution)

Hamshere 2015_IC

G‐CSF

Cardiologist

IC

BMMNC

G‐CSF infusion for 5 days and BM aspiration (**)

Autologous serum

n/r

Placebo (10 mL autologous serum)

Hamshere 2015_IM

G‐CSF

Cardiologist

IM

BMMNC

G‐CSF infusion for 5 days and BM aspiration (**)

Autologous serum

n/r

Placebo (2 mL autologous serum)

Heldman 2014_BMMNC

Standard medical therapy

Cardiologist

IM

BMMNC

BM aspiration (**)

n/r

n/r

Placebo (vehicle medium)

Heldman 2014_BM‐MSC

Standard medical therapy

Cardiologist

IM

BM‐MSC

BM aspiration (**), culture to select MSC

n/r

n/r

Placebo (vehicle medium)

Hendrikx 2006

CABG

Cardiothoracic surgeon

IM

BMMNC

BM aspiration (**)

Heparinised saline

60 (31) million cells

Placebo (heparinised saline)

Honold 2012

G‐CSF

Cardiologist

IC

CPC

G‐CSF infusion for 5 days prior to vein puncture, mononuclear cell isolation by gradient centrifugation and culture for 4 days for CPC

n/r

29 (12) million cells

No additional therapy (control)

Hu 2011

CABG

Cardiothoracic surgeon

IC

BMMNC

BM aspiration (**)

Saline solution and 20% autologous serum

132 (107) million cells

Placebo (8 mL saline; 2 mL autologous serum)

Jimenez‐Quevedo 2011

G‐CSF

Cardiologist

IM

CD133+

G‐CSF infusion for 5 days prior to leukapheresis, mononuclear cell isolation by gradient centrifugation immunomagnetic selection to isolate CD133+ cells

Normal saline solution

20 to 30 million cells

No additional therapy (control)

Losordo 2007

G‐CSF

Cardiologist

IM

CD34+

G‐CSF infusion for 5 days prior to leukapheresis, mononuclear cell isolation by gradient centrifugation immunomagnetic selection to isolate CD34+ cells

Saline solution and 5% autologous serum

LD: 0.05 million cells

MD: 0.1 million cells

HD: 0.5 million cells

Placebo (0.9% sodium chloride; 5% autologous plasma)

Losordo 2011

G‐CSF

Cardiologist

IM

CD34+

G‐CSF infusion for 5 days prior to leukapheresis, mononuclear cell isolation by gradient centrifugation immunomagnetic selection to isolate CD34+ cells

Saline solution and 5% autologous serum

LD: 0.1 million cells

HD: 0.5 million cells

Placebo (0.9% sodium chloride; 5% autologous plasma)

Mathiasen 2015

Standard medical therapy

Cardiologist

IM

BM‐MSC

BM aspiration (**), culture for 14 to 35 days to select MSC

Phosphate buffered saline with a drop of the participant’s blood

77.5 (68) million cells

Placebo (phosphate buffered saline mixed with drop of participant’s blood)

Mozid 2014_IC

G‐CSF

Cardiologist

IC

BMMNC

G‐CSF infusion for 5 days and BM aspiration (**)

Autologous serum

86 (110) million cells

Placebo (10 mL autologous serum)

Mozid 2014_IM

G‐CSF

Cardiologist

IM

BMMNC

G‐CSF infusion for 5 days and BM aspiration (**)

Autologous serum

52 (53) million cells

Placebo (2 mL autologous serum)

Nasseri 2012

CABG

Cardiothoracic surgeon

IM

CD133+

BM aspiration (**), immunomagnetic selection to isolate CD133+ cells

Sodium chloride and 10% autologous serum

Median 5.1 million cells

Placebo (isotonic saline solution; 10% autologous serum)

Patel 2005

CABG

Cardiothoracic surgeon

IM

CD34+

BM aspiration (**), immunomagnetic selection to isolate CD34+ cells

Heparinised saline and autologous serum

Median 22 million cells

No additional therapy (control)

Patel 2015

Standard medical therapy

Cardiologist

IC

BMAC

BM aspiration (**) and concentration

Autologous serum

3700 (900) million cells

No additional therapy (control)

Patila 2014

CABG

Cardiothoracic surgeon

IM

BMMNC

BM aspiration (**)

Medium 199 containing albumin, heparin

Median 840 (range 52 to 135) million cells

Placebo (vehicle medium)

Perin 2011

Standard medical therapy

Cardiologist

IM

BMMNC

BM aspiration (**)

Saline containing 5% human serum albumin

2 million cells

No additional therapy (control)

Perin 2012a

Standard medical therapy

Cardiologist

IM

BMMNC

BM aspiration (**)

Saline containing 5% human serum albumin

100 million cells

Placebo (cell‐free suspension in same volume)

Perin 2012b

Standard medical therapy

Cardiologist

IM

ALDH+

BM aspiration (**) and cell sorting

Pharmaceutical grade human serum albumin

2.4 (1.3) million cells

Placebo (5% pharmaceutical serum albumin)

Pokushalov 2010

Standard medical therapy

Cardiologist

IM

BMMNC

BM aspiration (**)

Heparinised saline

41 (16) million cells

No additional therapy (control)

Santoso 2014

Standard medical therapy

Cardiologist

IM

BMMNC

BM aspiration (**)

Phosphate buffered saline with 10% autologous plasma

n/r

Placebo (phosphate buffered saline; 10% autologous plasma)

Trifunovic 2015

CABG

Cardiothoracic surgeon

IM

BMMNC

BM aspiration (**)

n/r

70.7 (32.4) million cells

No additional therapy (control)

Tse 2007

Standard medical therapy

Cardiologist

IM

BMMNC

BM aspiration (**)

Phosphate buffered saline with 10% autologous plasma

15 million cells

Placebo (8 ‐ 12 x 0.1 mL phosphate buffered saline with 10% autologous serum)

Turan 2011

Standard medical therapy

Cardiologist

IC

BMMNC

BM aspiration (**)

n/r

99 (25) million cells

No additional therapy (control)

Van Ramshorst 2009

Standard medical therapy

Cardiologist

IM

BMMNC

BM aspiration (**)

Phosphate buffered saline with 0.5% human serum albumin

98 (6) million cells

Placebo (0.9% sodium chloride; 0.5% human serum albumin)

Wang 2009

Standard medical therapy

Cardiologist

IC

CD34+

BM aspiration (**), immunomagnetic selection to isolate CD34+ cells

Normal saline

Range 1.0 to 6.1 million cells

No additional therapy (control)

Wang 2010

Standard medical therapy

Cardiologist

IC

CD34+

BM aspiration (**), immunomagnetic selection to isolate CD34+ cells

Saline and human serum albumin

56 (23) million cells

Placebo (saline; human serum albumin)

Wang 2014

Standard medical therapy

Cardiologist

IM

CD133+

n/r

n/r

n/r

Placebo (n/r)

Wang 2015

CABG

Cardiothoracic surgeon

IM

BMMNC

BM aspiration (**)

Heparinised saline

521 (44) million cells

Placebo (saline solution)

Yao 2008

Standard medical therapy

Cardiologist

IC

BMMNC

BM aspiration (**)

Heparinised saline

72 million cells

Placebo (0.9% sodium chloride containing heparin)

Zhao 2008

CABG

Cardiothoracic surgeon

IM

BMMNC

BM aspiration (**)

Heparinised saline

659 (512) million cells

Placebo (saline)

**BM aspiration ‐ bone marrow aspiration and isolation of bone marrow mononuclear cells by gradient centrifugation.

ALDH: aldehyde dehydrogenase
BM: bone marrow
BMAC: bone marrow aspirate concentrate
BMMNC: bone marrow mononuclear cells
BM‐MSC: bone marrow‐derived mesenchymal stem cells
CABG: coronary artery bypass grafting
CPC: circulating progenitor cells
G‐CSF: granulocyte colony‐stimulating factor
HD: high dose
HDSW: high dose shockwave
IC: intracoronary
IM: intramyocardial
LD: low dose
LDSW: low dose shockwave
MD: medium dose
MSC: mesenchymal stem cells
n/r: not reported
SW: shockwave

Figuras y tablas -
Table 2. Characteristics of study interventions
Table 3. Summary of outcome reporting

Study ID

Primary outcomes

Secondary outcomes

All‐cause mortality

Non‐fatal MI

Hospital readmission for HF

Composite MACEa

Arrhythmias

NYHA class

CCS class

Angina frequency

Exercise tolerance

Quality of life

LVEFb

ST

LT

ST

LT

ST

LT

ST

LT

ST

LT

ST

LT

ST

LT

ST

LT

ST

LT

ST

LT

ST

LT

Ang 2008

FR

NR

PR*

NR

NR

NR

NR

NR

PR*

NR

PR

NR

PR

NR

NR

NR

NR

NR

NR

NR

FR

NR

Assmus 2006

FR

NR

FR

NR

FR

NR

FR

NR

FR

NR

FR

NR

NR

NR

NR

NR

NR

NR

NR

NR

FR

NR

Assmus 2013

FR

FR

NR

FR

FR

FR

NR

FR

NR

FR

FR

NR

NR

NR

NR

NR

NR

NR

NR

NR

FR

NR

Bartunek 2012

PR*

FR

NR

NR

NR

FR

NR

NR

PR

PR

PR

NR

NR

NR

NR

NR

FR

NR

PR

NR

FR

NR

Chen 2006

NR

FR

NR

NR

NR

NR

NR

NR

PR*

NR

FR

FR

NR

NR

NR

NR

FR

FR

NR

NR

FR

FR

Erbs 2005

PR*

FR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

FR

FR

NR

NR

FR

FR

Hamshere 2015_IC

PR*

PR*

PR*

FR

PR*

PR*

PR*

FR

FR

FR

FR

FR

FR

FR

NR

NR

NR

NR

NR

NR

PR

PR

Hamshere 2015_IM

PR*

PR*

PR*

PR*

FR

FR

FR

FR

FR

FR

FR

FR

FR

FR

NR

NR

NR

NR

NR

NR

PR

PR

Heldman 2014_BMMNC

PR*

PR*

NR

PR*

NR

FR

PR*

FR

NR

NR

NR

PR

NR

NR

NR

NR

FR

FR

FR

FR

NR

PR

Heldman 2014_BM‐MSC

PR*

FR

NR

PR*

NR

PR*

PR*

FR

NR

NR

NR

PR

NR

NR

NR

NR

FR

FR

FR

FR

NR

PR

Hendrikx 2006

FR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

FR

NR

Honold 2012

PR*

FR

FR

FR

PR*

FR

NR

NR

NR

NR

FR

FR

NR

NR

NR

NR

FR

FR

NR

NR

FR

FR

Hu 2011

FR

FR

PR*

NR

NR

NR

FR

NR

PR*

FR

NR

NR

NR

NR

NR

NR

FR

NR

NR

NR

FR

FR

Jimenez‐Quevedo 2011

FR

NR

PR*

NR

NR

NR

PR

NR

FR

NR

NR

NR

PR

NR

PR

NR

PR

NR

PR

NR

PR

NR

Losordo 2007

PR*

PR*

PR*

PR*

NR

NR

NR

NR

FR

FR

NR

NR

FR

NR

FR

NR

FR

NR

PR

NR

NR

NR

Losordo 2011

FR

FR

NR

FR

NR

FR

NR

PR

NR

NR

NR

NR

PR

PR

FR

NR

FR

FR

FR

FR

NR

NR

Mathiasen 2015

FR

NR

PR*

NR

FR

NR

NR

NR

FR

NR

PR

NR

PR

NR

PR

NR

PR

NR

PR

NR

FR

NR

Mozid 2014_IC

FR

NR

PR*

NR

FR

NR

FR

NR

PR*

NR

FR

NR

FR

NR

NR

NR

NR

NR

NR

NR

NR

NR

Mozid 2014_IM

FR

NR

PR*

NR

PR*

NR

FR

NR

FR

NR

FR

NR

FR

NR

NR

NR

NR

NR

NR

NR

NR

NR

Nasseri 2012

FR

FR

NR

NR

NR

NR

NR

NR

NR

NR

FR

NR

FR

NR

NR

NR

PR

NR

PR

NR

FR

NR

Patel 2005

PR*

FR

NR

NR

NR

NR

NR

NR

PR*

NR

FR

NR

NR

NR

NR

NR

NR

NR

NR

NR

PR

PR

Patel 2015

NR

FR

NR

NR

NR

FR

NR

NR

NR

PR*

NR

FR

NR

PR

NR

NR

NR

NR

NR

PR

PR

PR

Patila 2014

NR

PR*

NR

PR*

NR

FR

NR

NR

NR

NR

NR

FR

NR

NR

NR

NR

NR

NR

NR

PR

NR

FR

Perin 2011

PR*

NR

PR*

NR

NR

NR

NR

NR

PR*

NR

FR

NR

FR

NR

NR

NR

NR

NR

FR

NR

FR

NR

Perin 2012a

FR

NR

FR

NR

FR

NR

NR

NR

NR

NR

FR

NR

FR

NR

NR

NR

FR

NR

NR

NR

FR

NR

Perin 2012b

PR*

NR

FR

NR

NR

NR

NR

NR

FR

NR

FR

NR

FR

NR

NR

NR

NR

NR

NR

NR

FR

NR

Pokushalov 2010

FR

FR

NR

NR

NR

NR

NR

NR

PR*

PR*

FR

FR

FR

FR

FR

FR

FR

FR

FR

FR

FR

FR

Santoso 2014

PR*

FR

NR

NR

NR

NR

NR

NR

FR

NR

PR

NR

NR

NR

NR

NR

PR

NR

NR

NR

FR

NR

Trifunovic 2015

NR

FR

NR

NR

NR

NR

NR

NR

NR

NR

FR

FR

NR

NR

NR

NR

FR

FR

NR

NR

FR

FR

Tse 2007

PR*

FR

FR

NR

NR

NR

NR

NR

PR*

NR

FR

NR

FR

NR

NR

NR

FR

NR

NR

NR

FR

NR

Turan 2011

PR*

PR*

NR

NR

NR

NR

NR

NR

NR

NR

FR

FR

NR

NR

NR

NR

NR

NR

NR

NR

FR

FR

Van Ramshorst 2009

FR

NR

PR*

NR

NR

NR

NR

NR

PR*

NR

NR

NR

FR

NR

NR

NR

FR

NR

FR

NR

FR

FR

Wang 2009

PR*

NR

PR*

NR

NR

NR

NR

NR

PR*

NR

NR

NR

FR

NR

FR

NR

FR

NR

NR

NR

NR

NR

Wang 2010

PR*

NR

PR*

NR

NR

NR

NR

NR

FR

NR

NR

NR

FR

NR

FR

NR

FR

NR

NR

NR

NR

NR

Wang 2014

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

PR

NR

NR

NR

NR

NR

PR

NR

NR

NR

FR

NR

Wang 2015

PR*

NR

NR

NR

NR

NR

NR

NR

PR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

FR

NR

Yao 2008

PR*

NR

FR

NR

FR

NR

NR

NR

PR*

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

NR

PR

NR

Zhao 2008

FR

NR

PR*

NR

NR

NR

NR

NR

FR

NR

FR

NR

FR

NR

NR

NR

NR

NR

NR

NR

FR

NR

Total (%) analysedc

1637

(85.8)

1010

(53.0)

881

(46.2)

461 (24.2)

482

(25.3)

495

(26.0)

288 (15.1)

201

(10.5)

959

(50.3)

363

(19.0)

741

(38.9)

346

(18.1)

608

(31.9)

142 (7.4)

428

(22.4)

82 (4.3)d

535

(28.1)

227

(11.9)

197

(10.3)e

151

(7.9)e

439

(23.0)f

110

(5.8)f

CCS: Canadian Cardiovascular Society; FR: full reporting, outcome included in analysis; HF: heart failure; LT: long‐term follow‐up (≥ 12 months); LVEF: left ventricular ejection fraction; MACE: major adverse clinical events; MI: myocardial infarction; NR: outcome not reported; NYHA: New York Heart Association; PR: partial reporting with insufficient information on outcome reported for inclusion in analysis; PR*: no incidence of outcome observed; ST: short‐term follow‐up (< 12 months)

aComposite measure of mortality, reinfarction, or rehospitalisation for heart failure.
bLVEF measured by any method.
cTotal number of participants included in meta‐analysis of outcome (% of total number of participants from all included studies).
dNo meta‐analysis was performed, as only one study reported values suitable for inclusion.
eMinnesota Living with Heart Failure Questionnaire.
fTotal number analysed given for LVEF measured by magnetic resonance imaging.

Figuras y tablas -
Table 3. Summary of outcome reporting
Table 4. Clinical (dichotomous) outcomes

Study ID

Number of analysed participants

All‐cause mortality events

Non‐fatal MI events

Hospital readmission for HF

Composite MACEa

Arrhythmia events

Cells

No cells

Cells

No cells

Length of follow‐up

Cells

No cells

Length of follow‐up

Cells

No cells

Length of follow‐up

Cells

No cells

Length of follow‐up

Cells

No cells

Length of follow‐up

Ang 2008

42

19

1

1

6 mthsa

0

0

6 mths

n/r

n/r

n/r

n/r

n/r

n/r

0

0

6 mths

Assmus 2006

52

23

0

1

3 mths

1

0

3 mths

1

1

3 mths

1

1

3 mths

0

1

3 mths

Assmus 2013

43

39

6

8

45.7 (17) mths

1

4

45.7 (17) mths

8

13

45.7 (17) mths

14

19

45.7 (17) mths

6

13

45.7 (17) mths

Bartunek 2012

21

15

1

2

24 mths

n/r

n/r

n/r

6

4

24 mths

n/r

n/r

n/r

n/r

n/r

n/r

Chen 2006

22

23

2

4

12 mths

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

0

0

6 mths

Erbs 2005

13

12

0

1

15 mths

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

Hamshere 2015_IC

15

15

0

0

12 mths

1

0

12 mths

0

0

12 mths

1

0

12 mths

1

1

12 mths

Hamshere 2015_IM

15

15

0

0

12 mths

0

0

12 mths

1

1

12 mths

1

1

12 mths

0

1

12 mths

Heldman 2014_BMMNC

19

10

0

0

12 mths

0

0

12 mths

0

1

12 mths

0

1

12 mths

n/r

n/r

n/r

Heldman 2014_BM‐MSC

19

11

1

1

12 mths

0

0

12 mths

0

0

12 mths

1

1

12 mths

n/r

n/r

n/r

Hendrikx 2006

11

12

1

1

4 mths

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

Honold 2012

23

9

0

1

60 mths

1

2

60 mths

0

2

60 mths

n/r

n/r

n/r

n/r

n/r

n/r

Hu 2011

31

29

1

2

12 mths

0

0

6 mths

n/r

n/r

n/r

3

4

6 mths

1

0

12 mths

Jimenez‐Quevedo 2011

19

9

1

1

6 mths

0

0

6 mths

n/r

n/r

n/r

n/r

n/r

n/r

1

1

6 mths

Losordo 2007

18

6

0

0

12 mths

0

0

12 mths

n/r

n/r

n/r

n/r

n/r

n/r

0

1

12 mths

Losordo 2011

112

56

0

3

12 mths

6

7

12 mths

3

4

12 mths

n/r

n/r

n/r

n/r

n/r

n/r

Mathiasen 2015

40

20

1

1

6 mths

0

0

6 mths

6

2

6 mths

n/r

n/r

n/r

3

1

6 mths

Mozid 2014_IC

14

2

0

1

6 mths

0

0

6 mths

1

0

6 mths

1

1

6 mths

0

0

6 mths

Mozid 2014_IM

10

8

0

3

6 mths

0

0

6 mths

0

0

6 mths

0

3

6 mths

2

2

6 mths

Nasseri 2012

30

30

1

3

34 mthsb

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

Patel 2005

25

25

3

10

10 yrs

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

0

0

6 mths

Patel 2015

22

6

5

2

12 mths

n/r

n/r

n/r

2

0

12 mths

n/r

n/r

n/r

0

0

12 mths

Patila 2014

13c

17c

0

0

Median 60 mths

0

0

Median 60 mths

1

1

Median 60 mths

n/r

n/r

n/r

n/r

n/r

n/r

Perin 2011

20

10

0

0

6 mths

0

0

6 mths

n/r

n/r

n/r

n/r

n/r

n/r

0

0

6 mths

Perin 2012a

61

31

1

0

6 mths

1

0

6 mths

3

5

6 mths

n/r

n/r

n/r

n/r

n/r

n/r

Perin 2012b

10

10

0

0

6 mths

1

0

6 mths

n/r

n/r

n/r

n/r

n/r

n/r

3

2

6 mths

Pokushalov 2010

55

54

6

21

12 mths

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

0

0

12 mths

Santoso 2014

19

9

0

2

23 (8) mths

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

1

1

6 mths

Trifunovic 2015

15

15

2

4

Median 5 yrs

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

Tse 2007

19

9

0

1

19 (9) mths

0

1

3 mths

n/r

n/r

n/r

n/r

n/r

n/r

0

0

6 mths

Turan 2011

38

18

0

0

12 mths

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

Van Ramshorst 2009

25

25

1

0

6 mths

0

0

6 mths

n/r

n/r

n/r

n/r

n/r

n/r

0

0

6 mths

Wang 2009

16

16

0

0

6 mths

0

0

6 mths

n/r

n/r

n/r

n/r

n/r

n/r

0

0

6 mths

Wang 2010

56

56

0

0

6 mths

0

0

6 mths

n/r

n/r

n/r

n/r

n/r

n/r

0

1

6 mthsd

Wang 2014

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

Wang 2015

45

45

0

0

6 mths

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

n/r

Yao 2008

24

23

0

0

6 mths

0

1

6 mths

1

2

6 mths

n/r

n/r

n/r

0

0

6 mths

Zhao 2008

18

18

2

0

6 mths

0

0

6 mths

n/r

n/r

n/r

n/r

n/r

n/r

1

0

6 mths

HF: heart failure; MACE: major adverse clinical events; MI: myocardial infarction; n/r: not reported

aAng 2008: participants followed up for six months; mortality reported as “death within 30 days of treatment”.
bNasseri 2012: deaths reported “beyond follow‐up period” occurred at 31 and 34 months.
cPatila 2014: mortality rates reported in 20/19 participants at 12 months and 13/17 participants at 60 months.
dWang 2010: values are for ventricular arrhythmia (atrial arrhythmia also reported but unclear whether any participant overlap).

Figuras y tablas -
Table 4. Clinical (dichotomous) outcomes
Table 5. Periprocedural adverse events

Study ID

Periprocedural adverse events

Ang 2008

2 deaths (1 control, 1 intracoronary cell therapy) occurred within 30 days of treatment. Reasons were not given, but neither was considered to be related to cell therapy.

Assmus 2006

In‐hospital events: MI occurred in 1 CPC participant and ventricular arrhythmia detected during monitoring in 1 control participant.

Assmus 2013

n/r (only safety of shockwave procedure reported)

Bartunek 2012

In the cell therapy group, 1 participant had ventricular tachycardia during procedure which was resolved by cardioversion, and 1 participant had blurred vision after intervention (participant had pre‐existing ophthalmic migraines). Other reported adverse events (gastrointestinal, hepatobiliary, respiratory, thoracic, mediastinal, and peripheral vascular disorders) were not considered to be related to cell therapy.

Chen 2006

3 participants in cell therapy group experienced a transient episode of pulmonary oedema during the injection of stem cells. No sustained arrhythmias were monitored during the procedure.

Erbs 2005

1 cell therapy and 1 control participant reported headache, and 1 control participant developed fever during G‐CSF stimulation. G‐CSF resulted in comparable increases in serum C‐reactive protein levels and blood leukocyte count in both CPC and control groups (returned to baseline values within 4 days after G‐CSF). Neither G‐CSF injection nor intracoronary transplantation of CPC caused any elevation in troponin T levels.

Hamshere 2015_IC

n/r

Hamshere 2015_IM

n/r

Heldman 2014_BMMNC

No participant had significant postprocedural pericardial effusion. Small transient increases in CK‐MB and serum troponin I were observed. There were no treatment emergent serious adverse events among any of participants who received cell therapy.

Heldman 2014_BM‐MSC

No participant had significant postprocedural pericardial effusion. Small transient increases in CK‐MB and serum troponin I were observed. There were no treatment emergent serious adverse events among any of participants who received cell therapy.

Hendrikx 2006

1 cell therapy participant died on postoperative day 7 from a perforated oesophageal ulcer complicated by mediastinitis. 1 control participant died on the 5th postoperative day from multiorgan failure secondary to low cardiac output syndrome.

Honold 2012

Mild cephalgies and episodes of mild to moderate bone and muscular pain were reported during 5‐day course of G‐CSF. No participant developed chest pain episodes or clinical signs of decompensated HF. No novel ischaemia‐related ECG changes were observed during G‐CSF treatment and after intracoronary CPC infusion. Troponin T levels remained unchanged. Moreover, no specific G‐CSF‐mediated severe complications occurred. Intracoronary infusions were successfully performed without any procedural complications.

Hu 2011

2 participants (unclear which treatment arm) had neurological complications but recovered and were discharged. No participants had arrhythmia.

Jimenez‐Quevedo 2011

G‐CSF treatment was well tolerated, all participants presented bone pain as the only symptom. After cell injection, none of the participants had a significant rise in creatine phosphokinase, symptoms, ECG changes, or echocardiographic abnormalities.

Losordo 2007

13 participants reported transient increase in angina frequency after administration of G‐CSF. There were no cardiac enzyme elevations, MIs, acute coronary syndromes, or deaths. 1 participant in the placebo group developed ventricular tachycardia during the mapping procedure. No arrhythmias were detected by implantable cardioverter defibrillator, LifeVest, or Holter monitoring in any participant during or after the injection procedure.

Losordo 2011

Administration of G‐CSF was associated with bone pain (20.1%), angina (17.4%), CHF (2 participants), and 8 participants had troponin elevations consistent with non‐STEMI. In 1 participant a thrombus was observed on the mapping catheter tip as it was removed. 2 participants experienced an apparent myocardial perforation during the injection procedure (1 resulted in haemothorax, which was successfully treated; 1 resulted in cardiac tamponade; this participant died after unsuccessful pericardiocentesis procedure). Elevated troponin levels were observed in 28% of participants at some point during the mobilisation and injection period, all of which were minor and subclinical except for those mentioned above.

Mathiasen 2015

1 participant with a history of episodic ventricular tachycardia developed ventricular tachycardia during the NOGA mapping procedure. Another participant experienced double vision and dizziness during the injection procedure; cerebral‐CT afterwards was normal, but the incident was diagnosed as a minor stroke by the neurologist. 1 participant from the treatment group suffered a stroke 12 days after treatment.

Mozid 2014_IC

The most common side effects from G‑CSF were bone pain (22%) and low grade pyrexia (65%) (reported in all G‐CSF groups combined). Bleeding from the arterial access site did not differ significantly between the 2 intervention arms. All episodes were minor and resolved with conservative treatment within 24 h of the procedure. As expected, there were increases in troponin and creatine kinase levels postprocedure in both arms.

Mozid 2014_IM

The most common side effects from G‑CSF were bone pain (22%) and low grade pyrexia (65%) (reported in all G‐CSF groups combined). There were 3 cases of arrhythmia during the intramyocardial procedure that required treatment. Of these, 1 participant developed atrial fibrillation, which reverted to sinus rhythm within 24 h of the procedure. Another participant developed transient complete heart block periprocedure requiring temporary pacing only. The final participant suffered an episode of pulse­less ventricular tachycardia following intramyocardial injection, which was successfully cardioverted with a single 200 J external defibrillation and remained haemodynamically stable afterwards. 1 participant died from suspected acute LV failure 6 days after discharge. Bleeding from the arterial access site did not differ significantly between the two intervention arms. All episodes were minor and resolved with conservative treatment within 24 h of the procedure. As expected, there were increases in troponin and creatine kinase levels postprocedure in both arms.

Nasseri 2012

2 participants in the placebo group died early postoperatively: 1 died on day 8 after developing Candida sepsis following LV failure despite intra‐aortic balloon pump and catecholamine treatment and mechanical assist device implantation, and 1 died on day 22 (reason not given).

Patel 2005

1 participant in the OPCAB plus stem cell therapy group had a haematoma at the bone marrow harvest site. There were no other adverse events in either group (i.e. neurologic, haematologic, vascular, death, or infection events). No participants had any postoperative arrhythmias.

Patel 2015

5 participants who received BMAC experienced “non‐serious adverse events possibly related to the procedure”. Procedure‐related complications included haematomas at the catheterisation site and elevated serum creatinine levels.

Patila 2014

There were no differences between treatment groups in participants’ haemodynamics, arterial blood gases, systemic vein oxygen level, blood glucose, acid–base balance, lactate, haemoglobin, body temperature, and diuresis, as well as medications needed. Perioperative measures are reported in detail in Lehtinen 2014.

Perin 2011

No perforations or arrhythmias were associated with cell injection procedures. Postprocedural transient left bundle‐branch block (resolved in 24 h) was seen in 1 treated and 1 control participant. 1 treated participant had non‐significant pericardial effusion. No sustained ventricular arrhythmias were observed by Holter monitoring in any participant. Transient fever but no sepsis occurred in 1 control participant.

Perin 2012a

1 participant experienced a limited retrograde catheter‐related dissection of the abdominal aorta (withdrawn from study). 1 participant experienced recurrent ventricular tachycardia with hypotension (and received only a small volume of cell product).

Perin 2012b

No major adverse clinical cardiac events were associated with the cell injection procedures, including no perforations. Electromechanical mapping–related ventricular tachycardia occurred in 2 control participants, and ventricular fibrillation occurred in 1 control participant. No deaths occurred, and HF was not exacerbated in any participant. Holter monitoring showed no sustained ventricular arrhythmia in any participant.

Pokushalov 2010

No periprocedural complications occurred in participants who received cell therapy. 2‐dimensional echocardiography did not reveal postprocedural pericardial effusion. Creatine kinase activity and peak troponin T level remained unaltered. No new periprocedural arrhythmias were recorded during 24 h of consecutive electrocardiographic monitoring. An implantable cardioverter defibrillator was implanted to 2 participants with ventricular tachycardia prior to cell injections.

Santoso 2014

There were no acute procedural‐related complications, including stroke, transient ischaemic attack, ECG changes, sustained ventricular or atrial arrhythmias, and elevation of CPK‐MB. There was also no echocardiographic evidence of pericardial effusion in any participant within the first 24 h of the procedure.

Trifunovic 2015

The early postoperative course was uneventful in both groups with no significant differences between them with regard to adverse side effects during hospital stay. There were no significant differences in cardiac‐specific enzymes activities after the operation or the number of atrial fibrillation episodes or appearance of pericardial effusion between the groups.

Tse 2007

There were no acute procedure‐related complications, including stroke, transient ischaemic attack, ECG changes, sustained ventricular or atrial arrhythmias, elevation of CPK‐MB, or echocardiographic evidence of pericardial effusion within the first 24 h after the procedure.

Turan 2011

There was no inflammatory response or myocardial reaction (white blood cell count, C‐reactive protein, CK, troponin) after cell therapy. There were no immediate pre‐ or postprocedure adverse complications, new electrocardiographic changes, or significant elevations in CK or troponin, and no inflammatory response was observed in participants with bone marrow cell transplant.

Van Ramshorst 2009

In the placebo group, a greater than 0.5‐centimetre pericardial effusion was detected on 2‐dimensional echocardiography in an asymptomatic participant 2 days after the injection procedure, and pericardiocentesis was subsequently performed.

Wang 2009

No periprocedural adverse events; cardiac proteins in normal range.

Wang 2010

No increase in angina frequency or usage of sublingual NTG was observed in participants of either group. There were no cardiac enzyme elevations, MIs, acute coronary syndromes, or deaths. No participants from either group developed ventricular tachycardia during the cell or saline infusion procedure. No arrhythmias were detected by Holter monitoring in any participant during or after the infusion process.

Wang 2014

n/r

Wang 2015

Predischarge arrhythmias were reported (as number of events) in both cell therapy and control participants.

Yao 2008

Intracoronary application of BMC was performed without any acute or long‐term side effects. There was no inflammatory response or myocardial reaction (i.e. white blood cell count, C‐reactive protein, and creatinine phosphokinase) after cell therapy.

Zhao 2008

In the perioperative period, sporadic ventricular premature beats and self terminating bouts of rapid atrial fibrillation were observed in both groups. However, 2 participants developed VF, and 1 died in the BMMNC group: 1 participant developed VF on the 5th day postoperatively but was successfully resuscitated and VF well‐controlled, and the other developed refractory VF 5 hours' postoperatively with death on postoperative day 3. There were no ventricular arrhythmias in the control group.

AMI: acute myocardial infarction
BM: bone marrow
BMAC: bone marrow aspirate concentrate
BMC: bone marrow cells

BMMNC: bone marrow mononuclear cells
CHF: congestive heart failure
CK‐MB: creatine kinase‐MB
CPC: circulating progenitor cells
CPK‐MB: creatine phosphokinase‐MB
CT: computed tomography
ECG: electrocardiogram
G‐CSF: granulocyte colony‐stimulating factor
HF: heart failure
LV: left ventricular
MI: myocardial infarction
MSC: mesenchymal stem cells
non‐STEMI: non‐ST elevation myocardial infarction
n/r: not reported
NTG: nitroglycerine
OPCAB: off‐pump coronary artery bypass
PCI: percutaneous coronary intervention
ULN: upper limit of normal
VF: ventricular fibrillation

Figuras y tablas -
Table 5. Periprocedural adverse events
Table 6. Quality of life and performance measures

Study ID

No. analysed participants

Performance assessment

Mean follow‐up

No. analysed participants

Quality of life assessment

Mean follow‐up

Cells

No cells

ST

LT

Cells

No cells

ST

LT

Ang 2008

21

21

NYHA class (SR)a

6 mths

n/r

21

21

CCS class (SR)b

6 mths

n/r

Assmus 2006

43

18

NYHA class (EP)

3 mths

n/r

Assmus 2013

43

39

NYHA class (EP/MC)

4 mths

n/r

Bartunek 2012

21

15

NYHA class (SR)c

6 mths

n/r

21

15

MLHFQ (SR)c

6 mths

n/r

21

15

6MWT (distance) (EP)

6 mths

n/r

Chen 2006

22d

23d

NYHA class (EP)

6 mths

12 mths

22d

23d

ETT (METs) (EP)

6 mths

12 mths

Erbs 2005

12

10

Bike test (max O2 update) (EP)

3 mths

15 mths

Hamshere 2015_IC

15

15

NYHA class (EP)

6 mths

12 mths

15

15

CCS class (EP)

6 mths

12 mths

Hamshere 2015_IM

15

15

NYHA class (EP)

6 mths

12 mths

15

15

CCS class (EP)

6 mths

12 mths

Heldman 2014_BMMNC

17

16

NYHA class (SR)e

n/r

12 mths

15

19

MLHFQ (MC)

6 mths

12 mths

15f

19f

6MWT (distance) (MC)

6 mths

12 mths

Heldman 2014_BM‐MSC

17

16

NYHA class (SR)e

n/r

12 mths

19g

19g

MLHFQ (MC)

6 mths

12 mths

18h

19h

6MWT (distance) (MC)

6 mths

12 mths

Honold 2012

21j

10j

NYHA class (EP)

3 mths

60 mths

12k

5k

Bike test (sec) (EP)

3 mths

12 mths

Hu 2011

30

27

6MWT (distance) (EP/MC)

6 mths

n/r

Jimenez‐Quevedo 2011

19

9

CCS class (median)m

6 mths

n/r

n/r

n/r

SAQ (median)m

6 mths

n/r

15

7

ETT (time; METs) (median)m

6 mths

n/r

19

9

Angina frequency (median)n

6 mths

n/r

Losordo 2007

18

6

CCS class (MC)

6 mths

n/r

18

6

SAQ (SR)p

6 mths

n/r

18

6

ETT (time) (MC)

6 mths

n/r

17

6

Angina frequency (EP/MC)

6 mths

n/r

Losordo 2011

109q

53q

CCS class (SR)r

6 mths

12 mths

109q

53q

SAQ (MC)

6 mths

12 mths

109q

53q

ETT (time) (MC)

6 mths

12 mths

109

53

Angina frequency (EP)

6 mths

n/r

Mathiasen 2015

40

40

NYHA class (SR)s

6 mths

n/r

40

40

KCCQ‐QOL (SR)s

6 mths

n/r

40

40

CCS class (SR)s

6 mths

n/r

40

40

SAQ (SR)s

6 mths

n/r

40

40

6MWT (SR)s

6 mths

n/r

40

40

Angina frequency (SR)s

6 mths

n/r

Mozid 2014_IC

14

2

NYHA class (EP)

6 mths

n/r

14

2

CCS class (SR)

6 mths

n/r

Mozid 2014_IM

10

8

NYHA class (EP)

6 mths

n/r

10

8

CCS class (SR)

6 mths

n/r

Nasseri 2012

28

26

NYHA class (EP/MC)t

6 mths

n/r

28

26

MLHFQu

6 mths

n/r

28

26

6MWTu

6 mths

n/r

28

26

CCS class (EP/MC)t

6 mths

n/r

Patel 2005

10

10

NYHA class (EP/MC)t

6 mths

n/r

Patel 2015

17

4

NYHA class (EP)t

n/r

12 mths

17

4

MLHFQ (SR)

n/r

12 mths

17

4

CCS class (SR)

n/r

12 mths

Patila 2014

20

19

NYHA class (EP/MC)

n/r

12 mthsv

20

19

SF‐36w

n/r

60 mths

Perin 2011

20

10

NYHA class (EP)

6 mths

n/r

17

9

MLHFQ (EP)

6 mths

n/r

20

10

CCS class (EP/MC)

6 mths

n/r

13

10

SF‐36 (physical/mental) (EP)

6 mths

n/r

Perin 2012a

55

30

NYHA class (MC)

6 mths

n/r

44

22

CCS class (MC)

6 mths

n/r

51

29

6MWT (distance) (EP)

6 mths

n/r

Perin 2012b

10

10

NYHA class (EP)

6 mths

n/r

10

10

CCS class (EP)

6 mths

n/r

Pokushalov 2010

53x

46x

NYHA class (EP)

6 mths

12 mths

53x

46x

MLHFQ (EP)

6 mths

12 mths

53x

46x

CCS class (EP)

6 mths

12 mths

53x

46x

Angina frequency (EP)

6 mths

12 mths

53x

46x

6MWT (distance) (EP)

6 mths

12 mths

Santoso 2014

19

9

NYHA class (EP)y

6 mths

n/r

19

9

6MWT (distance) (EP)y

6 mths

n/r

Trifunovic 2015

15

15

NYHA class (EP)

6 mths

12 mths

15

15

6MWT (distance) (EP)

6 mths

12 mths

Tse 2007

19

9

NYHA class (EP)t

6 mths

n/r

19

9

CCS class (EP)t

6 mths

n/r

19

9

Treadmill test (time; METs) (EP/MC)

6 mths

n/r

Turan 2011

33

16

NYHA class (EP)

6 mths

12 mths

Van Ramshorst 2009

24

25

CCS class (EP)

6 mths

n/r

24

25

SAQ (EP/MC)

6 mths

n/r

24

25

Bike test (workload) (EP/MC)

6 mths

n/r

Wang 2009

16

16

CCS class (MC)

6 mths

n/r

16

16

Angina frequency (MC)

6 mths

n/r

16

16

ETT (min) (MC)

6 mths

n/r

Wang 2010

56

56

CCS class (EP/MC)

6 mths

n/r

56

56

Angina frequency (EP/MC)

6 mths

n/r

56

56

ETT (min) (EP/MC)

6 mths

n/r

Wang 2014

n/r

n/r

NYHA class (SR)

6 mths

n/r

n/r

n/r

5MWT (distance) (SR)

6 mths

n/r

Zhao 2008

16

18

NYHA class (EP)

6 mths

n/r

16

18

CCS class (EP)

6 mths

n/r

CCS: Canadian Cardiovascular Society; EP: endpoint; ETT: exercise tolerance test; KCCQ‐QOL: Kansas City Cardiomyopathy Questionnaire – Quality of Life; LT: long term; MC: mean change from baseline; MET: metabolic equivalent test (mL/kg/min); MLHFQ: Minnesota Living with Heart Failure Questionnaire; n/r: not reported; NYHA: New York Heart Association; SAQ: Seattle Angina Questionnaire; SF‐36: 36‐Item Short Form Health Survey; SR: summary results; ST: short term; 5MWT: 5‐minute walk test; 6MWT: 6‐minute walk test

aReported as number of participants in NYHA class III/IV.
bReported as number of participants in CCS class II or greater.
cReported graphically as percentage of participants showing improvement or deterioration.
d20/19 at 12 months.
eReported as number who improved/did not change/deteriorated.
f17/19 at 12 months.
g16/19 at 12 months.
h16/19 at 12 months.
j20/6 at 5 years.
k10/5 at 12 months.
mReported as median absolute difference with 95% confidence interval.
nMedian time to onset of angina also reported.
pResults presented graphically.
q106/50 at 12 months.
rReported as percentage of participants changed.
sResults presented graphically with P values for differences between groups.
tCalculated from frequency data.
uUnclear whether mean or median values are reported.
vAlso reported: median values at 60 months.
wReported graphically for each of eight components of SF‐36 at 60 months.
x49/33 at 12 months.
yReported as difference between groups at endpoint.

Figuras y tablas -
Table 6. Quality of life and performance measures
Table 7. Surrogate (continous) outcome: LVEF

Study ID

No. randomised participants

No. analysed participants

Baseline LVEF: Mean (SD)

Mean follow‐up of LVEF

Cells

No cells

Cells

No cells

Cells

No cells

ST

LT

Measured by MRI

Ang 2008

42

21

18

7

IM: 25.4 (8.1)

IC: 28.5 (6.5)

20.9 (8.9)

6 mths

Assmus 2013

43

39

15

12

n/r

n/r

4 mths

Erbs 2005

14

14

12a

11a

51.0 (12.1)

55.8 (12.4)

3 mths

15 mths

Hendrikx 2006

11

12

10

10

42.9 (10.3)

39.5 (5.5)

4 mths

Honold 2012

23

10

9

4

33.4 (SEM 12.7)

23.3 (SEM 7.2)

3 mths

12 mths

Hu 2011

31

29

31b

28b

23.5 (6.7)

24.8 (5.2)

6 mths

12 mths

Mathiasen 2015

40

20

40

20

28.2 (9.3)

25.1 (8.5)

6 mths

Nasseri 2012

30

30

26

22

27 (6)

26 (6)

6 mths

Patila 2014

20

19

18

7

37.1 (9.5)

38.5 (13.5)

60 mths

Santoso 2014

19

9

19

9

23.6 (8.4)

26.8 (8.8)

6 mths

Tse 2007

19

9

18

8

51.9 (8.5)

45.7 (8.3)

6 mths

Van Ramshorst 2009

25

25

22

18

56 (12)

54 (10)

6 mths

Wang 2014

35

35

35

35

29 (7)

28 (6)

6 mths

Measured by echocardiography

Bartunek 2012

32

15

21

15

27.5 (95% CI 25.5, 29.5)

27.8 (95% CI 25.9, 29.8)

6 mths

Hu 2011

31

29

24

18

36.0 (1.2)

34.7 (1.4)

12 mths

Perin 2011

20

10

20

10

37.0 (10.6)

39.0 (9.1)

6 mths

Perin 2012a

61

31

54

28

34.7 (8.8)

32.3 (8.6)

6 mths

Perin 2012b

10

10

10

10

36.1 (10.9)

32.1 (10.6)

6 mths

Pokushalov 2010

55

54

53c

46c

27.8 (3.4)

26.8 (3.8)

6 mths

12 mths

Trifunovic 2015

15

15

15

15

35.3 (3.9)

36.5 (5.3)

6 mths

12 mths

Van Ramshorst 2009

25

25

24

25

50 (5)

52 (5)

6 mths

Wang 2015

45

45

45

45

39.3 (6.2)

38.2 (8.0)

6 mths

Zhao 2008

18

18

16

18

35.8 (7.3)

36.7 (9.2)

6 mths

Measured by SPECT

Chen 2006

24

24

22d

23d

26 (6)

23 (8)

6 mths

12 mths

Perin 2011

20

10

20

10

41.5 (11.2)

43.0 (10.4)

6 mths

Van Ramshorst 2009

25

25

24

25

53 (12)

54 (12)

6 mths

12 mths

Measured by LV angiography

Assmus 2006

52

23

43

18

BMMNC: 41 (11)

CPC: 39 (10)

43 (13)

3 mths

Assmus 2013

43

39

41

38

LDSW: 37.2 (95% CI 31.7, 42.7)

HDSW: 32.4 (95% CI 26.9, 37.9)

LDSW: 29.9 (95% CI 24.0, 35.7)

HDSW: 32.3 (95% CI 26.5, 38.1)

4 mths

Honold 2012

23

10

21

5

37.5 (SEM 12.9)

37.6 (SEM 7.5)

3 mths

Perin 2011

20

10

20

10

37.5 (8.2)

40.0 (3.2)

6 mths

Perin 2012b

10

10

10

10

38.0 (17.5)

41.9 (11.8)

6 mths

Turan 2011

38

18

33

16

46 (10)

46 (10)

3 mths

12 mths

95% CI: 95% confidence interval; BMMNC: bone marrow mononuclear cells; CPC: circulating progenitor cells; HDSW: high‐dose shockwave; IC: intracoronary; IM: intramyocardial; LDSW: low‐dose shockwave; LT: long term; LV: left ventricular; LVEF: left ventricular ejection fraction; SD: standard deviation; SEM: standard error of the mean; SPECT: single‐photon emission computed tomography; ST: short term

a12/10 at 15 months.
b25/25 at 12 months.
c20/19 at 12 months.
d49/33 at 12 months.

Figuras y tablas -
Table 7. Surrogate (continous) outcome: LVEF
Comparison 1. Cells versus no cells

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause) Show forest plot

37

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Short term follow‐up (< 12 months)

33

1637

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.26, 0.87]

1.2 Long term follow‐up (≥ 12 months)

21

1010

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.25, 0.58]

2 Non‐fatal myocardial infarction Show forest plot

25

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Short term follow‐up (< 12 months)

20

881

Risk Ratio (M‐H, Random, 95% CI)

0.60 [0.17, 2.15]

2.2 Long term follow‐up (≥ 12 months)

9

461

Risk Ratio (M‐H, Random, 95% CI)

0.40 [0.17, 0.93]

3 Rehospitalisation due to heart failure Show forest plot

16

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 Short term follow‐up (< 12 months)

10

482

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.36, 1.12]

3.2 Long term follow‐up (≥ 12 months)

10

495

Risk Ratio (M‐H, Random, 95% CI)

0.62 [0.36, 1.04]

4 Arrhythmias Show forest plot

24

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 Short term follow‐up (< 12 months)

22

959

Risk Ratio (M‐H, Random, 95% CI)

0.70 [0.33, 1.45]

4.2 Long term follow‐up (≥ 12 months)

7

363

Risk Ratio (M‐H, Random, 95% CI)

0.46 [0.22, 0.97]

5 Composite MACE Show forest plot

9

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 Short term follow‐up (< 12 months)

8

288

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.18, 1.42]

5.2 Long term follow‐up (≥ 12 months)

5

201

Risk Ratio (M‐H, Random, 95% CI)

0.68 [0.41, 1.12]

6 MLHFQ: short term follow‐up (< 12 months) Show forest plot

4

Mean Difference (IV, Random, 95% CI)

Subtotals only

6.1 Mean value at endpoint

2

125

Mean Difference (IV, Random, 95% CI)

‐29.52 [‐33.76, ‐25.27]

6.2 Mean change from baseline

2

72

Mean Difference (IV, Random, 95% CI)

‐9.07 [‐22.09, 3.95]

6.3 Combined

4

197

Mean Difference (IV, Random, 95% CI)

‐18.96 [‐31.97, ‐5.94]

7 MLHFQ: long term follow‐up (≥ 12 months) Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

7.1 Mean value at endpoint

1

82

Mean Difference (IV, Random, 95% CI)

‐36.5 [‐42.21, ‐30.79]

7.2 Mean change from baseline

2

69

Mean Difference (IV, Random, 95% CI)

‐7.63 [‐16.35, 1.09]

7.3 Combined

3

151

Mean Difference (IV, Random, 95% CI)

‐17.80 [‐39.87, 4.26]

8 Seattle Angina Questionnaire: short term follow‐up (< 12 months) Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

8.1 Mean value at endpoint

1

49

Mean Difference (IV, Random, 95% CI)

5.0 [‐3.21, 13.21]

8.2 Mean change from baseline

2

211

Mean Difference (IV, Random, 95% CI)

9.34 [2.62, 16.07]

8.3 Combined

2

211

Mean Difference (IV, Random, 95% CI)

9.34 [2.62, 16.07]

9 Angina episodes per week: short term follow‐up (< 12 months) Show forest plot

5

Mean Difference (IV, Random, 95% CI)

Subtotals only

9.1 Mean value at endpoint

4

396

Mean Difference (IV, Random, 95% CI)

‐6.96 [‐11.99, ‐1.93]

9.2 Mean change from baseline

3

167

Mean Difference (IV, Random, 95% CI)

‐1.77 [‐14.61, 11.08]

9.3 Combined

5

428

Mean Difference (IV, Random, 95% CI)

‐5.11 [‐11.30, 1.09]

10 NYHA classification: short‐term follow‐up (< 12 months) Show forest plot

17

Mean Difference (IV, Random, 95% CI)

Subtotals only

10.1 Mean value at endpoint

16

658

Mean Difference (IV, Random, 95% CI)

‐0.42 [‐0.84, ‐0.00]

10.2 Mean change from baseline

4

239

Mean Difference (IV, Random, 95% CI)

‐0.56 [‐1.49, 0.36]

10.3 Combined

17

741

Mean Difference (IV, Random, 95% CI)

‐0.44 [‐0.84, ‐0.05]

11 NYHA classification: long term follow‐up (≥ 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

11.1 Mean value at endpoint

9

346

Mean Difference (IV, Random, 95% CI)

‐0.57 [‐1.03, ‐0.10]

11.2 Mean change from baseline

1

39

Mean Difference (IV, Random, 95% CI)

‐2.2 [‐2.70, ‐1.70]

11.3 Combined

9

346

Mean Difference (IV, Random, 95% CI)

‐0.81 [‐1.23, ‐0.39]

12 CCS class: short term follow‐up (< 12 months) Show forest plot

13

Mean Difference (IV, Random, 95% CI)

Subtotals only

12.1 Mean value at endpoint

10

486

Mean Difference (IV, Random, 95% CI)

‐0.32 [‐0.82, 0.18]

12.2 Mean change from baseline

6

318

Mean Difference (IV, Random, 95% CI)

‐0.62 [‐1.40, 0.17]

12.3 Combined

13

608

Mean Difference (IV, Random, 95% CI)

‐0.43 [‐0.92, 0.06]

13 CCS class: long term follow‐up (≥ 12 months) Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

13.1 Mean value at endpoint

3

142

Mean Difference (IV, Random, 95% CI)

‐0.58 [‐2.04, 0.88]

14 Exercise capacity: short term follow‐up (< 12 months) Show forest plot

16

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

14.1 Mean value at endpoint

11

563

Std. Mean Difference (IV, Random, 95% CI)

0.56 [0.19, 0.93]

14.2 Mean change from baseline

9

535

Std. Mean Difference (IV, Random, 95% CI)

0.33 [0.05, 0.61]

15 Exercise capacity: long term follow‐up (≥ 12 months) Show forest plot

8

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

15.1 Mean value at endpoint

5

178

Std. Mean Difference (IV, Random, 95% CI)

1.14 [0.04, 2.25]

15.2 Mean change from baseline

3

227

Std. Mean Difference (IV, Random, 95% CI)

0.34 [0.07, 0.62]

16 LVEF (%) measured by MRI: short term follow‐up (< 12 months) Show forest plot

12

Mean Difference (IV, Random, 95% CI)

Subtotals only

16.1 Mean value at endpoint

10

352

Mean Difference (IV, Random, 95% CI)

3.01 [‐0.05, 6.07]

16.2 Mean change from baseline

9

308

Mean Difference (IV, Random, 95% CI)

4.05 [2.55, 5.55]

16.3 Combined

12

439

Mean Difference (IV, Random, 95% CI)

2.92 [1.03, 4.82]

17 LVEF (%) measured by MRI: long term follow‐up (≥ 12 months) Show forest plot

4

Mean Difference (IV, Random, 95% CI)

Subtotals only

17.1 Mean value at endpoint

4

110

Mean Difference (IV, Random, 95% CI)

2.37 [‐1.54, 6.29]

17.2 Mean change from baseline

3

97

Mean Difference (IV, Random, 95% CI)

3.83 [‐0.42, 8.08]

17.3 Combined

4

110

Mean Difference (IV, Random, 95% CI)

4.38 [0.82, 7.93]

18 LVEF (%) measured by echocardiography: short term follow‐up (< 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

18.1 Mean value at endpoint

8

388

Mean Difference (IV, Random, 95% CI)

5.16 [2.87, 7.44]

18.2 Mean change from baseline

3

161

Mean Difference (IV, Random, 95% CI)

3.47 [1.59, 5.34]

18.3 Combined

9

470

Mean Difference (IV, Random, 95% CI)

5.71 [4.29, 7.13]

19 LVEF (%) measured by echocardiography: long term follow‐up (≥ 12 months) Show forest plot

3

Mean Difference (IV, Random, 95% CI)

Subtotals only

19.1 Mean value at endpoint

3

154

Mean Difference (IV, Random, 95% CI)

7.69 [6.47, 8.92]

19.2 Mean change from baseline

1

82

Mean Difference (IV, Random, 95% CI)

6.1 [‐1.27, 13.47]

19.3 Combined

3

154

Mean Difference (IV, Random, 95% CI)

7.96 [6.39, 9.54]

20 LVEF (%) measured by SPECT: short term follow‐up (< 12 months) Show forest plot

4

Mean Difference (IV, Random, 95% CI)

Subtotals only

20.1 Mean value at endpoint

4

145

Mean Difference (IV, Random, 95% CI)

2.41 [‐2.65, 7.46]

20.2 Mean change from baseline

1

30

Mean Difference (IV, Random, 95% CI)

‐2.3 [‐17.33, 12.73]

20.3 Combined

4

145

Mean Difference (IV, Random, 95% CI)

5.22 [2.60, 7.85]

21 LVEF (%) measured by SPECT: long term follow‐up (≥ 12 months) Show forest plot

2

Mean Difference (IV, Random, 95% CI)

Subtotals only

21.1 Mean value at endpoint

2

88

Mean Difference (IV, Random, 95% CI)

0.37 [‐2.30, 3.04]

21.2 Mean change from baseline

1

49

Mean Difference (IV, Random, 95% CI)

4.0 [‐6.48, 14.48]

21.3 Combined

2

88

Mean Difference (IV, Random, 95% CI)

0.28 [‐2.48, 3.03]

22 LVEF (%) measured by LV angiography: short term follow‐up (< 12 months) Show forest plot

6

Mean Difference (IV, Random, 95% CI)

Subtotals only

22.1 Mean value at endpoint

6

265

Mean Difference (IV, Random, 95% CI)

3.18 [0.39, 5.97]

22.2 Mean change from baseline

4

181

Mean Difference (IV, Random, 95% CI)

1.72 [0.50, 2.95]

22.3 Combined

6

250

Mean Difference (IV, Random, 95% CI)

2.00 [0.53, 3.46]

23 LVEF (%) measured by LV angiography: long term follow‐up (≥ 12 months) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

23.1 Mean value at endpoint

1

49

Mean Difference (IV, Random, 95% CI)

6.0 [0.81, 11.19]

Figuras y tablas -
Comparison 1. Cells versus no cells
Comparison 2. Cell dose: subgroup analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause): short term follow‐up (< 12 months) Show forest plot

30

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 < 107 cells

6

334

Risk Ratio (M‐H, Random, 95% CI)

0.18 [0.02, 1.63]

1.2 107 < 108 cells

18

771

Risk Ratio (M‐H, Random, 95% CI)

0.34 [0.15, 0.79]

1.3 ≥ 108 cells

8

487

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.35, 1.94]

2 Mortality (all‐cause): long term follow‐up (≥ 12 months) Show forest plot

16

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 < 107 cells

4

297

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.10, 1.09]

2.2 107 < 108 cells

7

330

Risk Ratio (M‐H, Random, 95% CI)

0.30 [0.17, 0.53]

2.3 ≥ 108 cells

5

236

Risk Ratio (M‐H, Random, 95% CI)

0.62 [0.30, 1.26]

3 NYHA classification: short term follow‐up (< 12 months) Show forest plot

15

Mean Difference (IV, Random, 95% CI)

Subtotals only

3.1 < 107 cells

4

149

Mean Difference (IV, Random, 95% CI)

‐0.29 [‐0.94, 0.36]

3.2 107 < 108 cells

8

309

Mean Difference (IV, Random, 95% CI)

‐0.65 [‐1.22, ‐0.08]

3.3 ≥ 108 cells

4

241

Mean Difference (IV, Random, 95% CI)

‐0.41 [‐0.72, ‐0.11]

4 CCS class: short term follow‐up (< 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 < 107 cells

4

288

Mean Difference (IV, Random, 95% CI)

‐0.87 [‐1.92, 0.19]

4.2 107 < 108 cells

5

160

Mean Difference (IV, Random, 95% CI)

‐0.54 [‐1.40, 0.32]

5 Exercise capacity: short term follow‐up (< 12 months) Show forest plot

10

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

5.1 107 < 108 cells

7

357

Std. Mean Difference (IV, Random, 95% CI)

0.56 [‐0.03, 1.14]

5.2 ≥ 108 cells

3

161

Std. Mean Difference (IV, Random, 95% CI)

0.43 [0.10, 0.77]

6 LVEF (%) measured by MRI: short term follow‐up (< 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

6.1 107 < 108 cells

7

199

Mean Difference (IV, Random, 95% CI)

5.23 [3.91, 6.54]

6.2 ≥ 108 cells

3

101

Mean Difference (IV, Random, 95% CI)

2.37 [‐0.92, 5.66]

Figuras y tablas -
Comparison 2. Cell dose: subgroup analysis
Comparison 3. Baseline cardiac function: subgroup analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause): short term follow‐up (< 12 months) Show forest plot

28

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 < 30%

11

508

Risk Ratio (M‐H, Random, 95% CI)

0.23 [0.09, 0.59]

1.2 30 ‐ 50%

13

642

Risk Ratio (M‐H, Random, 95% CI)

0.87 [0.36, 2.11]

1.3 > 50%

4

271

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.11, 3.35]

2 Mortality (all‐cause): long term follow‐up (≥ 12 months) Show forest plot

16

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 < 30%

9

426

Risk Ratio (M‐H, Random, 95% CI)

0.36 [0.20, 0.64]

2.2 30 ‐ 50%

7

289

Risk Ratio (M‐H, Random, 95% CI)

0.57 [0.27, 1.21]

3 NYHA classification: short term follow‐up (< 12 months) Show forest plot

15

Mean Difference (IV, Random, 95% CI)

Subtotals only

3.1 < 30%

6

273

Mean Difference (IV, Random, 95% CI)

‐0.40 [‐1.22, 0.43]

3.2 30 ‐ 50%

9

420

Mean Difference (IV, Random, 95% CI)

‐0.32 [‐0.54, ‐0.10]

4 NYHA classification: long term follow‐up (≥ 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 < 30%

5

202

Mean Difference (IV, Random, 95% CI)

‐0.66 [‐1.28, ‐0.04]

4.2 30 ‐ 50%

4

144

Mean Difference (IV, Random, 95% CI)

‐0.98 [‐1.72, ‐0.25]

5 CCS class: short term follow‐up (< 12 months) Show forest plot

8

Mean Difference (IV, Random, 95% CI)

Subtotals only

5.1 < 30%

4

213

Mean Difference (IV, Random, 95% CI)

‐0.25 [‐1.47, 0.97]

5.2 30 ‐ 50%

4

150

Mean Difference (IV, Random, 95% CI)

‐0.11 [‐0.31, 0.09]

6 Exercise capacity: short term follow‐up (< 12 months) Show forest plot

7

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

6.1 < 30%

4

225

Std. Mean Difference (IV, Random, 95% CI)

0.96 [0.37, 1.56]

6.2 30 ‐ 50%

3

127

Std. Mean Difference (IV, Random, 95% CI)

0.38 [‐0.57, 1.33]

7 LVEF (%) measured by MRI: short term follow‐up (< 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

7.1 < 30%

6

290

Mean Difference (IV, Random, 95% CI)

1.54 [‐1.96, 5.03]

7.2 30 ‐ 50%

3

60

Mean Difference (IV, Random, 95% CI)

3.31 [0.88, 5.75]

Figuras y tablas -
Comparison 3. Baseline cardiac function: subgroup analysis
Comparison 4. Route of cell administration: subgroup analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause): short term follow‐up (< 12 months) Show forest plot

33

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Intramyocardial

22

1049

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.21, 1.03]

1.2 Intracoronary

12

607

Risk Ratio (M‐H, Random, 95% CI)

0.51 [0.21, 1.23]

2 Mortality (all‐cause): long term follow‐up (≥ 12 months) Show forest plot

21

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Intramyocardial

13

652

Risk Ratio (M‐H, Random, 95% CI)

0.29 [0.17, 0.50]

2.2 Intracoronary

8

358

Risk Ratio (M‐H, Random, 95% CI)

0.57 [0.30, 1.09]

3 NYHA classification: short term follow‐up (< 12 months) Show forest plot

17

Mean Difference (IV, Random, 95% CI)

Subtotals only

3.1 Intramyocardial

11

445

Mean Difference (IV, Random, 95% CI)

‐0.48 [‐0.99, 0.03]

3.2 Intracoronary

6

296

Mean Difference (IV, Random, 95% CI)

‐0.38 [‐0.76, 0.00]

4 NYHA classification: long term follow‐up (≥ 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 Intramyocardial

4

181

Mean Difference (IV, Random, 95% CI)

‐1.09 [‐1.76, ‐0.41]

4.2 Intracoronary

5

165

Mean Difference (IV, Random, 95% CI)

‐0.61 [‐0.92, ‐0.30]

5 CCS class: short term follow‐up (< 12 months) Show forest plot

13

Mean Difference (IV, Random, 95% CI)

Subtotals only

5.1 Intramyocardial

10

434

Mean Difference (IV, Random, 95% CI)

‐0.33 [‐0.87, 0.22]

5.2 Intracoronary

3

174

Mean Difference (IV, Random, 95% CI)

‐1.00 [‐2.87, 0.86]

6 Exercise capacity: short term follow‐up (< 12 months) Show forest plot

11

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

6.1 Intramyocardial

6

310

Std. Mean Difference (IV, Random, 95% CI)

0.78 [0.19, 1.36]

6.2 Intracoronary

5

253

Std. Mean Difference (IV, Random, 95% CI)

0.33 [‐0.06, 0.72]

7 LVEF (%) measured by MRI: short term follow‐up (< 12 months) Show forest plot

12

Mean Difference (IV, Random, 95% CI)

Subtotals only

7.1 Intramyocardial

8

309

Mean Difference (IV, Random, 95% CI)

2.18 [‐0.41, 4.77]

7.2 Intracoronary

5

137

Mean Difference (IV, Random, 95% CI)

3.72 [0.86, 6.57]

Figuras y tablas -
Comparison 4. Route of cell administration: subgroup analysis
Comparison 5. Cell type: subgroup analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause): short term follow‐up (< 12 months) Show forest plot

33

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Mononuclear cells

20

966

Risk Ratio (M‐H, Random, 95% CI)

0.54 [0.28, 1.04]

1.2 Circulating progenitor cells

3

104

Risk Ratio (M‐H, Random, 95% CI)

0.32 [0.01, 7.48]

1.3 Haematopoietic progenitor cells

8

464

Risk Ratio (M‐H, Random, 95% CI)

0.27 [0.05, 1.46]

1.4 Mesenchymal stem cells

3

126

Risk Ratio (M‐H, Random, 95% CI)

0.5 [0.03, 7.59]

2 Mortality (all‐cause): long term follow‐up (≥ 12 months) Show forest plot

19

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Mononuclear cells

12

540

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.25, 0.70]

2.2 Haematopoietic progenitor cells

4

302

Risk Ratio (M‐H, Random, 95% CI)

0.26 [0.10, 0.69]

2.3 Mesenchymal stem cells

3

111

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.15, 1.57]

3 NYHA classification: short term follow‐up (< 12 months) Show forest plot

15

Mean Difference (IV, Random, 95% CI)

Subtotals only

3.1 Mononuclear cells

12

547

Mean Difference (IV, Random, 95% CI)

‐0.42 [‐0.86, 0.02]

3.2 Haematopoietic progenitor cells

3

94

Mean Difference (IV, Random, 95% CI)

‐0.47 [‐1.95, 1.02]

4 CCS class: short term follow‐up (< 12 months) Show forest plot

13

Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 Mononuclear cells

8

366

Mean Difference (IV, Random, 95% CI)

‐0.39 [‐0.99, 0.21]

4.2 Haematopoietic progenitor cells

5

242

Mean Difference (IV, Random, 95% CI)

‐0.54 [‐1.55, 0.46]

Figuras y tablas -
Comparison 5. Cell type: subgroup analysis
Comparison 6. Participant diagnosis: subgroup analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause): short term follow‐up (< 12 months) Show forest plot

33

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Chronic IHD

11

550

Risk Ratio (M‐H, Random, 95% CI)

0.65 [0.26, 1.62]

1.2 HF (secondary to IHD)

15

645

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.14, 0.82]

1.3 Refractory/intractable angina

7

442

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.11, 3.35]

2 Mortality (all‐cause): long term follow‐up (≥ 12 months) Show forest plot

21

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Chronic IHD

9

389

Risk Ratio (M‐H, Random, 95% CI)

0.52 [0.27, 0.99]

2.2 HF (secondary to IHD)

9

401

Risk Ratio (M‐H, Random, 95% CI)

0.33 [0.19, 0.58]

2.3 Refractory/intractable angina

3

220

Risk Ratio (M‐H, Random, 95% CI)

0.11 [0.01, 0.91]

3 NYHA classification: short term follow‐up (< 12 months) Show forest plot

16

Mean Difference (IV, Random, 95% CI)

Subtotals only

3.1 Chronic IHD

6

296

Mean Difference (IV, Random, 95% CI)

‐0.43 [‐0.78, ‐0.07]

3.2 HF (secondary to IHD)

10

417

Mean Difference (IV, Random, 95% CI)

‐0.47 [‐1.02, 0.09]

4 NYHA classification: long term follow‐up (≥ 12 months) Show forest plot

9

Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 Chronic IHD

3

105

Mean Difference (IV, Random, 95% CI)

‐0.66 [‐0.91, ‐0.42]

4.2 HF (secondary to IHD)

6

241

Mean Difference (IV, Random, 95% CI)

‐0.92 [‐1.47, ‐0.37]

5 CCS class: short term follow‐up (< 12 months) Show forest plot

13

Mean Difference (IV, Random, 95% CI)

Subtotals only

5.1 HF (secondary to IHD)

8

363

Mean Difference (IV, Random, 95% CI)

‐0.25 [‐0.90, 0.40]

5.2 Refractory/intractable angina

5

245

Mean Difference (IV, Random, 95% CI)

‐0.78 [‐1.44, ‐0.11]

6 Exercise capacity: short term follow‐up (< 12 months) Show forest plot

11

Std. Mean Difference (IV, Random, 95% CI)

Subtotals only

6.1 Chronic IHD

4

114

Std. Mean Difference (IV, Random, 95% CI)

0.48 [‐0.26, 1.22]

6.2 HF (secondary to IHD)

4

260

Std. Mean Difference (IV, Random, 95% CI)

0.79 [0.04, 1.53]

6.3 Refractory/intractable angina

3

189

Std. Mean Difference (IV, Random, 95% CI)

0.26 [‐0.03, 0.55]

7 LVEF (%) measured by MRI: short term follow‐up (< 12 months) Show forest plot

10

Mean Difference (IV, Random, 95% CI)

Subtotals only

7.1 Chronic IHD

6

178

Mean Difference (IV, Random, 95% CI)

2.58 [‐0.16, 5.31]

7.2 HF (secondary to IHD)

4

195

Mean Difference (IV, Random, 95% CI)

2.50 [‐1.97, 6.97]

Figuras y tablas -
Comparison 6. Participant diagnosis: subgroup analysis
Comparison 7. Co‐interventions: subgroup analysis

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause): short term follow‐up (< 12 months) Show forest plot

33

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Co‐interventions

8

432

Risk Ratio (M‐H, Random, 95% CI)

0.74 [0.32, 1.70]

1.2 No co‐interventions

25

1205

Risk Ratio (M‐H, Random, 95% CI)

0.31 [0.13, 0.72]

2 Mortality (all‐cause): long term follow‐up (≥ 12 months) Show forest plot

21

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Co‐interventions

6

312

Risk Ratio (M‐H, Random, 95% CI)

0.47 [0.26, 0.88]

2.2 No co‐interventions

15

698

Risk Ratio (M‐H, Random, 95% CI)

0.32 [0.19, 0.56]

3 NYHA classification: short term follow‐up (< 12 months) Show forest plot

17

Mean Difference (IV, Random, 95% CI)

Subtotals only

3.1 Co‐interventions

6

233

Mean Difference (IV, Random, 95% CI)

‐0.57 [‐1.20, 0.05]

3.2 No co‐interventions

11

508

Mean Difference (IV, Random, 95% CI)

‐0.37 [‐0.87, 0.13]

4 LVEF (%) measured by MRI: short term follow‐up (< 12 months) Show forest plot

12

Mean Difference (IV, Random, 95% CI)

Subtotals only

4.1 Co‐interventions

5

179

Mean Difference (IV, Random, 95% CI)

2.01 [‐0.26, 4.29]

4.2 No co‐interventions

7

260

Mean Difference (IV, Random, 95% CI)

3.55 [0.82, 6.27]

Figuras y tablas -
Comparison 7. Co‐interventions: subgroup analysis
Comparison 8. Sensitivity analysis: excluding studies with high/unclear risk of selection bias

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause) Show forest plot

15

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Short term follow‐up (< 12 months)

14

744

Risk Ratio (M‐H, Random, 95% CI)

0.69 [0.32, 1.50]

1.2 Long term follow‐up (≥ 12 months)

9

491

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.21, 0.87]

2 Non‐fatal myocardial infarction Show forest plot

11

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

2.1 Short term follow‐up (< 12 months)

6

288

Risk Ratio (M‐H, Random, 95% CI)

0.50 [0.05, 4.58]

2.2 Long term follow‐up (≥ 12 months)

5

345

Risk Ratio (M‐H, Random, 95% CI)

0.38 [0.15, 0.97]

3 Rehospitalisation due to heart failure Show forest plot

8

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

3.1 Short term follow‐up (< 12 months)

3

234

Risk Ratio (M‐H, Random, 95% CI)

0.65 [0.32, 1.32]

3.2 Long term follow‐up (≥ 12 months)

6

375

Risk Ratio (M‐H, Random, 95% CI)

0.63 [0.36, 1.09]

4 Arrhythmias Show forest plot

7

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

4.1 Short term follow‐up (< 12 months)

6

224

Risk Ratio (M‐H, Random, 95% CI)

0.77 [0.18, 3.21]

4.2 Long term follow‐up (≥ 12 months)

1

82

Risk Ratio (M‐H, Random, 95% CI)

0.42 [0.18, 0.99]

5 Composite MACE Show forest plot

3

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

5.1 Short term follow‐up (< 12 months)

2

59

Risk Ratio (M‐H, Random, 95% CI)

0.0 [0.0, 0.0]

5.2 Long term follow‐up (≥ 12 months)

3

141

Risk Ratio (M‐H, Random, 95% CI)

0.64 [0.38, 1.08]

6 NYHA classification: short term follow‐up (< 12 months) Show forest plot

5

Mean Difference (IV, Random, 95% CI)

Subtotals only

6.1 Combined

5

277

Mean Difference (IV, Random, 95% CI)

‐0.26 [‐0.59, 0.07]

7 NYHA classification: long term follow‐up (≥ 12 months) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

7.1 Combined

1

39

Mean Difference (IV, Random, 95% CI)

‐2.2 [‐2.70, ‐1.70]

8 LVEF (%) measured by MRI: short term follow‐up (< 12 months) Show forest plot

7

Mean Difference (IV, Random, 95% CI)

Subtotals only

8.1 Combined

7

249

Mean Difference (IV, Random, 95% CI)

2.92 [0.67, 5.17]

9 LVEF (%) measured by MRI: long term follow‐up (≥ 12 months) Show forest plot

1

Mean Difference (IV, Random, 95% CI)

Subtotals only

9.1 Combined

1

25

Mean Difference (IV, Random, 95% CI)

‐1.60 [‐8.70, 5.50]

Figuras y tablas -
Comparison 8. Sensitivity analysis: excluding studies with high/unclear risk of selection bias
Comparison 9. Sensitivity analysis: excluding studies with high/unclear risk of performance bias

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause) Show forest plot

26

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Short term follow‐up (< 12 months)

25

1216

Risk Ratio (M‐H, Random, 95% CI)

0.58 [0.29, 1.16]

1.2 Long term follow‐up (≥ 12 months)

13

624

Risk Ratio (M‐H, Random, 95% CI)

0.43 [0.21, 0.86]

Figuras y tablas -
Comparison 9. Sensitivity analysis: excluding studies with high/unclear risk of performance bias
Comparison 10. Sensitivity analysis: excluding studies with high/unclear risk of attrition bias

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Mortality (all‐cause) Show forest plot

32

Risk Ratio (M‐H, Random, 95% CI)

Subtotals only

1.1 Short term follow‐up (< 12 months)

28

1449

Risk Ratio (M‐H, Random, 95% CI)

0.48 [0.26, 0.89]

1.2 Long term follow‐up (≥ 12 months)

17

883

Risk Ratio (M‐H, Random, 95% CI)

0.39 [0.25, 0.60]

Figuras y tablas -
Comparison 10. Sensitivity analysis: excluding studies with high/unclear risk of attrition bias