In order to bring you the best possible user experience, this site uses Javascript. If you are seeing this message, it is likely that the Javascript option in your browser is disabled. For optimal viewing of this site, please ensure that Javascript is enabled for your browser.
Did you know that your browser is out of date? To get the best experience using our website we recommend that you upgrade to a newer version. Learn more.

Cardiac stem cell therapy

An article from the e-journal of the ESC Council for Cardiology Practice

Cell therapy is a potential new treatment for post MI patients, but the main challenges are : the necessity to preserve immune competency and to gain adequate nutrition and homing signals necessary for stem cells’ engraftment and survival. A marginal benefit has been observed in many preclinical studies, however it is also a challenge to understand the mechanisms of these benefits. As such, stem cell therapy cannot be regarded as a valid therapeutic option for patients with cardiovascular disease in the present era of evidenced-based medicine, yet it holds great hope for the future.



I - INTRODUCTION

    The idea of myocardial regeneration with stem cell transplantation after myocardial infarction receives tremendous interest. Preclinical and some small-scaled clinical trials have suggested feasibility and safety of cardiac stem cell therapy. However, there are still questions waiting for answers and results of large-scale randomized trials are needed for the appearance of myocardial cellular therapy as a clinical therapeutic option for patients with ischemic heart disease.

Acute myocardial infarction and resultant heart failure is a major cause of mortality and morbidity worldwide (1). Left ventricular remodeling after acute myocardial infarction (MI) is a complex pathological process producing alterations in topography of both the infarcted and non-infarcted regions of the ventricle leading to progressive dilatation and systolic dysfunction (2). Current developments in revascularization strategies and medical therapies such as beta-blockers and medications interfering with the action of renin-angiotensin aldosterone system have focused on remodeling. However these therapies are of limited value for restoration of cardiac functions after myocardial infarction. Cell therapy is currently emerging as a potential new treatment for post MI patients with the assumption that recolonization of the areas of scarred myocardium with exogenously supplied surrogates or precursors of cardiomyocytes can restore function and ultimately affect clinical outcomes.

Various cell types have been tested experimentally for cardiac repair so far, but only those of autologous origin have yet undergone clinical testing due to immune competency. These include bone marrow-derived cells, peripheral blood derived stem cells and skeletal myoblasts (3).

  Several preliminary reports have demonstrated that local stem cell transplantation in patients with acute myocardial infarction is safe and may lead to improved myocardial function and perfusion. The bone morrow contains several stem cell types including hematopoetic stem cells, endothelial progenitor cells, mesenchymal (stromal) stem cells and multipotent adult progenitor cells. In experimental and clinical studies unselected mononuclear bone marrow cells as well as specific subpopulations have been used for transplantation. Although there is no definitive evidence, combinations of progenitor cells seem to be more benefical than specific stem cell type for cardiac repair (3). Interestingly, contradictory data exist for transformation of bone marrow derived progenitor cells (BMPC) into new cardiomyocytes and alternative mechanisms as enhanced neovascularization, enhanced scar tissue formation due to augmented inflammatory response and decreased apoptosis have been suggested for the benefical effects of these cells on myocardial function after myocardial infarction (4).

    Repair of scar tissue constitutes a challenge for cardiac stem cell therapy due to lack of adequate nutrition and homing signals necessary for stem cells’ engraftment and survival. The resistant state of skeletal myoblasts to ischemia renders these cells suitable candidates for repair of chronically infracted and failing heart (5). However, in spite of their resistance to hypoxia only a small portion of cells survive when transplanted into an infarct scar (6). Injection of myoblasts into infarcted myocardium has been shown to augment systolic and diastolic functions in experimental animal studies (7). Myoblasts retain skeletal muscle properties and couple only sporadically to resident cardiomyocytes precluding synchronized contraction as mechanism of benefit (8). Moreover, it seems that paracrine action of skeletal myoblasts facilitate neighboring cardiomyocytes to maintain their replicative potential and/or stimulate differentiation of native cardiac stem cells (8,9). Stimulation of angiogenesis and stabilization of extracellular matrix have also been suggested to contribute to the benefit (9). However, lack of electromechanical coupling may create an arrhythmogenic substrate and is the main limitation of cellular therapy with myoblasts (10).

Modes of Cell Delivery

  So far, progenitor cells for cardiac repair have been delivered via an intracoronary arterial route or by injection of the ventricular wall via percutaneous endocardial, percutaneous transcoronary venous or surgical epicardial approach.

  Intracoronary application permits application of a maximum concentration of cells homogenously to the site of injury. Skeletal myoblasts are not suitable for this mode of delivery due to their large size and potential for distal obstruction of microcirculation. This mode of delivery has mostly been used for patients with acute myocardial infarction through the central lumen of an over-the-wire balloon catheter during transient balloon inflations to maximize the contact time of the cells with the microcirculation of the infarct related artery.
However, for the unperfused regions of the myocardium this mode of delivery may not be efficient.

   Direct injection is the preferred route for chronic ischemic heart failure patients with considerable scar tissues. Cell homing signals as ‘vascular endothelial growth factor (VEGF)’ and ‘stromal cell derived factor-1 (SDF-1)’ are expressed at low levels late in disease process precluding successful engraftment of cells applied via intracoronary route (11). Direct injection techniques are especially suited for the application of large cells as myoblasts and mesenchymal stem cells. Direct delivery of progenitor cells into scar tissue or areas of hibernating myocardium by catheter-based endocardial injection or direct injection during open-heart surgery techniques are not limited by cell uptake from the circulation or by embolic risk. Endocardial electromechanical mapping techniques are often used as an adjunct to endocardial injection to identify viable but hibernating myocardial regions, which are preferred areas for stem cell injection (12). Transepicardial cell injection has been performed as an adjunct to coronary artery bypass grafting (CABG). A novel technique for direct myocardial cell delivery is via transvenous approach using coronary veins with a special catheter system under fluoroscopic and intravascular ultrasound guidance (13).

II - CLINICAL TRIALS OF CARDIAC STEM CELL THERAPY

1. Acute Myocardial Infarction

    The first small clinical trial justifying the safety and feasibility of the intracoronary application of autologous BM-SCs in patients after recent myocardial infarction (MI) was published in 2002. Strauer et al. (14) applied bone marrow mononuclear cells after AMI via the intracoronary route five to nine days after acute myocardial infarction in 10 patients. After 3 months of follow up the investigators found that the infarct region significantly decreased and cardiac functions were significantly improved in the treatment group without any significant adverse effects. Afterwards, results of small clinical trials as TOPCARE-AMI (Transplantation of Progenitor Cells and Regeneration Enhancement in Acute Myocardial Infarction) and BOOST (Bone Marrow Transfer to Enhance ST-Elevation Infarct Regeneration) studies were reported (15-18). Overall, these trials showed marginal improvement of cardiac functions and myocardial perfusion after 3 to 6 months, without significant adverse effects. The beneficial effects of progenitor cell transplantation persisted during a 1-year follow-up period without significant adverse events in TOPCARE-AMI trial (19). However, no significant differences between patient and control groups regarding improvement of cardiac functions were reported at 18 months’ follow-up of BOOST trial (20).

   Observations from initial clinical studies encouraged investigators to perform larger scale randomized trials. Recently published randomized, double-blind, placebo controlled REPAIR-AMI trial randomly assigned 204 patients with acute myocardial infarction to receive an intracoronary infusion of progenitor cells derived from bone marrow or placebo into the infarct artery 3 to 7 days after successful reperfusion therapy. At 4 months, the absolute improvement in the global left ventricular ejection fraction (LVEF) was significantly greater in the BMPC group than in the placebo group (21). At 1 year, intracoronary infusion of BMPC was associated with a reduction in the prespecified combined clinical end point of death, recurrence of myocardial infarction, and any revascularization procedure (22). Contradictory to these results, another recently published randomized study; ASTAMI trial showed no effects of intracoronary injection of autologous mononuclear BMPC on global left ventricular function could be observed during 6-month follow-up period (23). One hundred patients with a reperfused anterior myocardial infarction patients were randomly assigned to stem cell implant or not, 5-8 days after an acute event in this study. The reasons for the conflict between these two studies are not clear. However, different cell isolation protocols used in these studies may have contributed to this discrepancy (24).

    Due to technical difficulties and the invasive nature of harvesting stem cells from bone marrow, investigators also tried peripheral blood derived progenitor cells (PBPC’s) for cardiac repair after myocardial infarction. These cells were equally effective as BMPC’s for improving cardiac functions in TOPCARE-AMI trial (15). The major concern is the insufficient number of these cells in circulation and need for ex vivo proliferation or utilization of stem cell mobilization factors as ‘stem cell factor (SCF)’ or ‘granulocyte colony-stimulating factor (G-CSF)’. Both of these factors have been proposed to stimulate myogenesis and angiogenesis in the infracted area and to improve cardiac function after AMI in experimental studies (25,26). Granulocyte colony stimulating factor (G-CSF) is frequently used to mobilize marrow stem cells into circulation but is associated with mobilization of other immune cells, which leads to nonspecific inflammation. In the first clinical investigation, the MAGIC cell randomized clinical trial, Kang et al. (27) prospectively randomised 27 patients with myocardial infarction who underwent coronary stenting for the culprit lesion of infarction into three groups; cell infusion group in which G-CSF mobilized peripheral blood-derived progenitor cells were used (n=10), G-CSF alone group (n=10) and control group (n=7). Changes in left ventricular systolic function and perfusion were assessed after 6 months. G-CSF injection and intracoronary infusion of the mobilised PBSC did not aggravate inflammation and ischaemia during the periprocedural period. Exercise capacity, myocardial perfusion and systolic function improved significantly in patients who received cell infusion. However, an unexpectedly high rate of in-stent restenosis was observed at culprit lesion in patients who received G-CSF, which prompted a premature termination of the study. Local augmentation of inflammation by G-CSF was suggested to contribute to this observation but no such effect was observed in other clinical trials integrating G-CSF therapy into primary percutaneous coronary intervention (PCI) management of patients with AMI (28,29). The FIRSTLINE-AMI (Front-Integrated Revascularization and Stem Cell Liberation in Evolving Acute Myocardial Infarction by Granulocyte Colony-Stimulating Factor) trial reported benefical effect of G-CSF therapy given 85 ± 30 min after primary PCI. At 12 months, treatment with G-CSF resulted in a significant improvement of EF of %8 while EF of control subjects decreased by %5 (28). No adverse events as enhanced inflammation, thrombosis, electrical instability or accelerated restenosis were observed throughout the study. However, results of another randomized, prospective, double-blinded, placebo controlled phase II trial indicate no benefit of G-CSF therapy when PCI is performed late (30).

   Two-year follow-up results of MAGIC Cell trial indicate better results with intracoronary cell infusion with mobilized PBSC’s by G-CSF than G-CSF therapy alone, which may indicate inability to achieve sufficient homing and integration of stem cells in the infarct territory unless stem cells are infused locally (31). In a recently published trial Zhan-Quan et al. (32) reported significant improvement of global left ventricular function and significant decrease of ventricular volumes at 6th month with intracoronary autologous PBSCs transplantation pre-mobilized by G-CSF therapy.

2. Chronic Ischemic Heart Disease

  In the first clinical trial concerning autologous skeletal myoblast transplantation in patients with severe ischemic cardiomyopathy, Menasche et al. (10) transplanted skeletal myoblasts to 10 patients at the time of CABG surgery. At an average follow-up of 10.9 months, the functional capacity of patients was significantly improved and ejection fractions were significantly increased (%24 ± 1 to %32± 1, p<0.02). Four out of ten patients experienced delayed episodes of sustained ventricular tachycardia and were implanted with an internal defibrillator. Simultaneous effect of revascularization obscured the analysis about the efficiency of myoblasts. One of the patients died 17 months after transplantation because of a non-cardiac cause and detailed histological examination of the heart revealed engrafted skeletal muscle cells demonstrating the survival ability of these cells in hostile environment (33). The improvement of both clinical status and EF persisted during a median of 56 months of follow up period with a strikingly low incidence of hospitalizations for heart failure. Arrhythmic risk of patients could be controlled by medical therapy and/or ICD implantation (34). Regardless of the injection route, several phase I studies have demonstrated the survival, feasibility, and safety of autologous myoblast transplantation and have suggested that this modality offers a potential therapeutic treatment for end-stage heart disease with arrhythmogenic potential (35-36). In a multicenter, randomized, double blind, placebo-controlled phase II trial 97 patients were assigned to low-dose myoblast, high-dose myoblast or placebo transplantation groups during CABG. There were no improvements in contractility of cell-transplanted segments or in global left ventricular function. However, there were no statistical increases in arrhythmic risk and there was reversal of remodeling and improved scintigraphic LVEF in the high cell dose group (37). These results justify further exploration of this strategy with larger scale controlled trials.

    Besides the skeletal myoblasts, BMPCs were used for cellular therapy of chronic ischemic cardiomyopathy as well. In the first pilot trial Stamm et al (38) injected CD133+ bone marrow stem cells into the infarct border zones of 12 patients during the CABG operation. Scintigraphic imaging demonstrated significantly improved local perfusion in the stem cell-treated infarct area. LV dimensions and LV ejection fraction (%39.7 ± 9 vs %48.7 ± 6, p=0.007) improved without any significant adverse events as new ventricular arrhythmia or neoplasia. After this pilot trial, results of a randomized controlled trial were reported (Regeneration of Human Infarcted Heart Muscle by Intracoronary Autologous Bone Marrow Transplantation in Chronic Coronary Artery Disease: the IACT study) (39). In this study autologous bone marrow mononuclear cells were transplanted via intracoronary route to 18 patients with chronic MI (5 months to 8.5 years old). After three months, infarct size was reduced by 30% and global left ventricular ejection fraction and infarction wall movement velocity increased significantly in the transplantation group. However, no significant changes were observed in infarct size, left ventricular ejection fraction, or wall movement velocity of infarcted area in the control group. No significant adverse events as significant arrhythmias and restenosis of the culprit artery were observed during follow-up. After bone marrow cell transplantation, there was an improvement of maximum oxygen uptake and of regional 18F-fluoro-desoxy-glucose uptake into infarct tissue. These results demonstrated the possibility of functional and metabolic regeneration of infarcted and chronically avital tissue by bone marrow mononuclear cell transplantation in humans. Some phase I studies also investigated the feasibility and efficacy of percutaneous endocardial transplantation of BMPCs guided by electromechanical mapping (40-42). Improvements in symptoms, myocardial function and function at the ischemic regions were demonstrated without significant adverse events as arrhythmias, evidence of myocardial inflammation or increased scar formation.  

III - UNRESOLVED ISSUES IN CARDIAC STEM CELL THERAPY

   Experience with preclinical studies have raised important questions that will hopefully be resolved by larger scale clinical studies (43,44). These trials will further clarify the effects of stem cell therapy on surrogate markers of cardiac functions as LVEF, myocardial perfusion, or exercise capacity and long-term safety of the procedure. 

   Although a marginal benefit was observed in many preclinical studies with stem cell therapy, the mechanisms of benefit are still poorly understood. Optimal cell type and cell dosage to be used for different situations of ischemic heart disease and timing of cell transfer are also important issues that need to be further clarified. Theoretically, timing of cell infusion may be unsuitable - either too early or too late, since early administration causes implanted cells to be killed by an inflammatory environment and late administration can lead to disconnection between implanted cells and cardiomyocytes by fibrosis scar. Long-term experimental data regarding the fate of transplanted cells are still missing. Transdifferentiation and homing mechanisms are poorly understood. Cell labeling and imaging techniques need to be developed to track stem cell fate in patients and correlate cell retention and engraftment with functional outcomes. Pharmacological and genetic strategies seem to offer enhancement of stem cell retention, engraftment, differentiation and paracrine capability, which deserves further exploration (45).

The content of this article reflects the personal opinion of the author/s and is not necessarily the official position of the European Society of Cardiology.

 

Conclusion:

Cardiac stem cell therapy appears as a promising mode of therapy for ischemic heart disease. Although data from preclinical experiments and small clinical studies point to the potential for restoring cardiac functions following myocardial infarction; there are still uncertainties about the technique, safety and long-term clinical outcome. As such, stem cell therapy can not be regarded as a valid therapeutic option for patients with cardiovascular disease in the present era of evidenced-based medicine. Data from large, randomized controlled trials are needed to clarify the short and long term effects of myocardial cellular therapy and suitability for clinical application.  

References


1. Cohn JN, Bristow MR, Chien KR, et al. Report of the National Heart, Lung and Blood Institute special emphasis panel on heart failure research. Circulation 1997; 95: 766-770.

2. Pfeffer MA, Braunwald E. Ventricular remodeling after myocardial infarction: experimental observations and clinical implications. Circulation 1990; 81: 1161-72.

3. Wollert KC, Drexler H. Cell based therapy for heart failure. Curr Opin Cardiol 2006; 21: 234-239.

4.  Murry CE, Wiseman RW, Schwartz SM, et al. Skeletal myoblast transplantation for repair of myocardial necrosis. J Clin Invest 1996; 98: 2512–23.

5. Pagani FD, DerSimonian H, Zawadzka A, et al. Autologous skeletal myoblasts transplanted to ischemia-damaged myocardium in humans. J Am Coll Cardiol 2003; 41: 879-888.

6. Dowell JD, Rubart M, Pasumarthi KB, et al. Myocyte and myogenic stem cell transplantation in the heart. Cardiovasc Res 2003; 58: 336-350.

7. Leobon B, Garcin I, Menasche P, et al. Myoblasts transplanted into rat infarcted myocardium are functionally isolated from their host. Proc Natl Acad Sci USA 2003; 100: 7808-7811.
8. Memon IA, Sawa Y, Fukushima N, et al. Repair of impaired myocardium by means of implantation of engineered autologous myoblast sheets. J Thorac Cardiovasc Surg 2005; 130(5): 1333-41.

9. Menasché P, Hagége AA, Vilquin JT, et al. Autologous skeletal myoblast transplantation for severe postinfarction left ventricular dysfunction. J Am Coll Cardiol 2003; 41: 1078-1083.

10. Lee MS, Lill M, Makkar RR. Stem cell transplantation in myocardial infarction. Rev Cardiovasc Med 2004; 5(2): 82-98.

11. Perin EC, Dohmann HF, Borojevic R, et al. Transendocardial, autologous bone marrow cell transplantation for severe, chronic ischemic heart failure. Circulation 2003; 107: 2294-2302.

12. Siminiak T, Fiszer D, Jerzykowska O, et al. Percutaneous trans-coronary-venous transplantation of autologous skeletal myoblasts in the treatment of post-infarction myocardial contractility impairment: the POZNAN trial. Eur Heart J 2005; 26(12):1188-95.

13. Strauer BE, Brehm M, Zeus T, et al. Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans. Circulation 2002; 106: 1913-1918.

14. Assmus B, Schachinger V, Teupe C, et al. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction (TOPCARE-AMI). Circulation 2002; 106: 3009-3017.

15. Meyer GP, Wollert KC, Lotz J, et al.Intracoronary bone marrow cell transfer after myocardial infarction: eighteen months' follow-up data from the randomized, controlled BOOST (Bone marrow transfer to enhance ST-elevation infarct regeneration) trial. Circulation 2006; 113(10): 1287-94.
 
16. Chen SL, Fang WW, Ye F, et al. Effect on left ventricular function of intracoronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarction. Am J Cardiol 2004; 94(1): 92-5.

17. Fernandez-Aviles F, San Roman JA, Garcia-Frade J, et al. Experimental and clinical regenerative capability of human bone marrow cells after myocardial infarction. Circ Res 2004; 95(7): 742-8.

18. Schachinger V, Assmus B, Britten MB, et al. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction: final one-year results of the TOPCARE-AMI Trial. J Am Coll Cardiol 2004; 44(8): 1690-9.

19. Wollert KC, Meyer GP, Lotz J, et al. Intracoronary autologous bone-marrow cell transfer after myocardial infarction:the BOOST randomised controlled clinical trial. Lancet 2004; 364: 141–148.

20. Schachinger V, Erbs S, Elsasser A, et al. Intracoronary bone marrow-derived progenitor cells in acute myocardial infarction. N Engl J Med 2006; 355(12): 1210-21.

21. Schachinger V, Erbs S, Elsasser A, et al. Improved clinical outcome after intracoronary administration of bone-marrow-derived progenitor cells in acute myocardial infarction: final 1-year results of the REPAIR-AMI trial. Eur Heart J 2006; 27(23): 2775-83.

22. Lunde K, Solheim S, Aakhus S, et al. Intracoronary injection of mononuclear bone marrow cells in acute myocardial infarction. N Engl J Med 2006; 355(12): 1199-209.

23. Seeger FH, Tonn T, Krzossok N, et al. Cell isolation procedures matter: a comparison of different isolation protocols of bone marrow mononuclear cells used for cell therapy in patients with acute myocardial infarction. Eur Heart J 2007; 28(6): 766-72.

24. Ohtsuka M, Takano H, Zou Y, et al. Cytokine therapy prevents left ventricular remodeling and dysfunction after myocardial infarction through neovascularization. FASEB J 2004; 18: 851–3.

25. Ohki Y, Heissig B, Sato Y, et al. Granulocyte colony-stimulating factor promotes neovascularization by releasing vascular endothelial growth factor from neutrophils. FASEB J 2005; 19: 2005–7.

26. Kang HJ, Kim HS, Zhang SY, et al. Effects of intracoronary infusion of peripheral blood stem-cells mobilised with granulocyte-colony stimulating factor on left ventricular systolic function and restenosis after coronary stenting in myocardial infarction: the magic cell randomised clinical trial. Lancet 2004; 363: 751–756.

27. Ince H, Petzsch M, Kleine HD, et al. Prevention of left ventricular remodeling with granulocyte colony-stimulating factor after acute myocardial infarction: final 1-year results of the Front-Integrated Revascularization and Stem Cell Liberation in Evolving Acute Myocardial Infarction by Granulocyte Colony-Stimulating Factor (FIRSTLINE-AMI) Trial. Circulation 2005; 112: I73– 80.

28. Jorgensen E, Ripa RS, Helqvist S, et al. In-stent neo-intimal hyperplasia after stem cell mobilization by granulocyte-colony stimulating factor. Preliminary intracoronary ultrasound results from a double-blind randomized placebo-controlled study of patients treated with percutaneous coronary intervention for ST-elevation myocardial infarction (STEMMI Trial). Int J Cardiol 2006; 111:174 –7.

29. Engelmann MG, Theiss HD, Hennig-Theiss C, et al. Autologous bone marrow stem cell mobilization induced by granulocyte colony-stimulating factor after subacute ST-segment elevation myocardial infarction undergoing late revascularization: final results from the G-CSF-STEMI (Granulocyte Colony-Stimulating Factor ST-Segment Elevation Myocardial Infarction) trial. J Am Coll Cardiol 2006; 48(8): 1712-21.

30. Kang HJ, Kim HS, Koo BK, Kim YJ, Lee D, Sohn DW, Oh BH, Park YB. Intracoronary infusion of the mobilized peripheral blood stem cell by G-CSF is better than mobilization alone by G-CSF for improvement of cardiac function and remodeling: 2-year follow-up results of the Myocardial Regeneration and Angiogenesis in Myocardial Infarction with G-CSF and Intra-Coronary Stem Cell Infusion (MAGIC Cell) 1 trial. Am Heart J 2007; 153(2): 237.e1-8.

31. Zhan-quan L, Ming Z, Yuan-zhe J, et al. The Clinical Study of autologous peripheral blood stem cell transplantation by intracoronary infusion in patients with acute myocardial infarction. Int J Cardiol 2007; 115: 52-56.
32. Hagege AA, Carrion C, Menasche P, et al. Viability and differentiation of autologous skeletal myoblast grafts in ischaemic cardiomyopathy. Lancet 2003; 361(9356): 491-2.

33. Hagege AA, Marolleau JP, Vilquin JT, et al. Skeletal myoblast transplantation in ischemic heart failure: long-term follow-up of the first phase I cohort of patients.
Circulation 2006; 114(1 Suppl): I108-13.

34. Pagani FP, DerSimonian H, Zawadzka A, et al. Autologous skeletal myoblasts transplanted to ischemia-damaged myocardium in humans. J Am Coll Cardiol 2003; 41: 879-888.
35. Smits PC, Van Geuns RJ, Poldermans D, et al. Catheter-based intramyocardial injection of autologous skeletal myoblasts as a primary treatment of ischemic heart failure. J Am Cardiol 2003; 42: 2063-2069.

36. Menasce P, et al. Myoblast Autologous Grafting in Ischemic Cardiomyopathy (MAGIC) trial. Clin Cardiol 2007; 30(2): 98.
37. Stamm C, Westphal B, Hans-Dieter K, et al. Autologous bone-marrow stem-cell transplantation for myocardial regeneration. Lancet 2003; 361: 45-46.

38. Strauer BE, Brehm M, Zeus T, et al. Regeneration of human infarcted heart muscle by intracoronary autologous bone marrow cell transplantation in chronic coronary artery disease: the IACT Study. J Am Coll Cardiol 2005; 46(9): 1651-8.

39. Perin EC, Dohmann H, Borojevic R, et al. Transendocardial, autologous bone marrow cell transplantation for severe, chronic ischemic heart failure. Circulation 2003; 107: 2294-2302.
40. Tse HF, Kwong YL, Chan J, et al. Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet 2003; 361: 47-49.
41. Fuchs S, Satler LF, Kornowski R et al. Catheter-based autologous bone marrow myocardial injection in no-option patients with advanced coronary artery disease: a feasibility study. J Am Coll Cardiol 2003; 41: 1721–1724.

42. Nyolczas N, Gyongyosi M, Beran G, et al. Design and rationale for the Myocardial Stem Cell Administration After Acute Myocardial Infarction (MYSTAR) Study: a multicenter, prospective, randomized, single-blind trial comparing early and late intracoronary or combined (percutaneous intramyocardial and intracoronary) administration of nonselected autologous bone marrow cells to patients after acute myocardial infarction. Am Heart J 2007; 153(2): 212.e1-7.

43. Hirsch A, Nijveldt R, van der Vleuten PA, et al. Intracoronary infusion of autologous mononuclear bone marrow cells or peripheral mononuclear blood cells after primary percutaneous coronary intervention: rationale and design of the HEBE trial- a prospective, multicenter, randomized trial. Am Heart J 2006; 152(3): 434-41.

44. Fazel S, Tang GH, Angoulvant D, et al. Current status of cellular therapy for ischemic heart disease. Ann Thorac Surg 2005; 79(6): S2238-47.

 

 

VolumeNumber:

Vol5 N°31

Notes to editor


Dr. Alper Kepez, *Dr. Ali Oto
Ankara, Turkey
*Member of the EBSC and EHRA

The content of this article reflects the personal opinion of the author/s and is not necessarily the official position of the European Society of Cardiology.