295
Views
13
CrossRef citations to date
0
Altmetric
Review Article

Present and future of stem cells for cardiovascular therapy

, , , , , & show all
Pages 412-427 | Published online: 08 Jul 2009

Abstract

In this review we summarize the available evidence regarding the application of stem cell therapy for human cardiovascular repair, going over the principal concepts that will help us understand the present and future of this therapy: first the different types of cells available in clinical practice, second the delivery approaches, and third highlighting the most important clinical studies and their efficacy and safety results.

In addition, we also speculate on the value of current clinical data to gain an insight into the mechanism of stem cell‐based cardiac repair and to design clinical trials in the future.

Introduction

Prognosis for patients with myocardial infarction has improved spectacularly in the last 25 years thanks to the success of strategies that limit necrosis, increase electrical stability of the myocardium and improve ventricular function Citation1.Since the main underlying cause of postinfarction left ventricle remodeling and dysfunction is irreversible damage to the infarcted wall, the development of treatments aimed at regenerating its muscular and vascular components is now considered a main therapeutic challenge.

The rationale for such an approach includes recent evidence demonstrating the remarkable ability of adult stem cells to produce differentiated cells from embryologically unrelated tissues Citation2, as well as convincing data supporting the fact that the heart has a potent intrinsic regenerative capacity Citation3, with the existence of cardiac stem cells able to regenerate myocytes and vasculature after ageing or damage Citation4–6. In spite of concerns, initial studies suggest that this is also possible in humans Citation7.

In this review we summarize the available evidence regarding the application of this therapy for human cardiovascular repair, going over the principal concepts that will help us understand the present and future of this therapy; first the different types of cells available in clinical practice, second the delivery approaches, and third highlighting the most important clinical studies and their efficacy and safety results. In addition, we also speculate on the value of current clinical data to gain an insight into the mechanism of stem cell‐based cardiac repair and to design clinical trials in the future.

Concept of stem cell, types and mechanism of action

Stem cells are defined as cells that remain in an early stage of development (immature) capable of dividing indefinitely (self‐renewable) and of giving rise to replica cells (clonogenic) as well as to differentiate to various cell lineages (multipotency).

The most characteristic stem cells are the embryonic stem cells, present in the fertilized oocyte or in the inner cell mass of the blastocyst. As well as in the embryo, stem cells remain in adult tissues (adult stem cells) as skin, skeletal muscle, intestinal mucosa, etc. Adult stem cells stay within the tissues in niches with a specific microenvironment and begin to divide and differentiate into specialized cells of the same germ layer when particular conditions are present. Therefore, two main stem cell sources are available: embryonic and adult stem cells Citation7. To date, no cardiovascular clinical experience with embryonic cells is available, while the most used adult stem cell sources in clinical trials have been skeletal myoblasts and bone marrow‐derived stem cells (Figure ).

Figure 1 The most used adult stem cell sources in clinical trials have been skeletal myoblasts(top) and mononuclear bone marrow (bottom) stem cells.

Figure 1 The most used adult stem cell sources in clinical trials have been skeletal myoblasts(top) and mononuclear bone marrow (bottom) stem cells.

Embryonic stem cells

Embryonic cells are highly proliferative and totipotent, giving rise to all tissues of the adult body. As they are obtained from the inner cell mass of embryos in blastocyst stage, the potential use of human embryonic cells has several problems: the ethical issue, the need of immunosuppression when using allogenic cells for implanting, and its tumorigenic capacity.

Adult stem cells

Skeletal myoblast

These cells are resident satellite stem cells in the muscle and constitute a source of cells with a contractile phenotype. To obtain a sufficient number of cells, myoblasts need to be cultured and expanded for approximately 3 weeks before transplantation Citation8. Thus, myoblasts have been used only out of the acute clinical setting. It is admitted that myoblasts, which lack electromechanical integration with the host cardiomyocytes, could provide a putative arrhythmogenic substrate Citation8–10.

Bone marrow‐derived stem cells

Bone marrow mononuclear stem cells

Bone marrow mononuclear stem cells include several cell subpopulations with different morphologic and phenotype characteristics (Table ) Citation11, Citation12. Experimental work has shown their capacity to transdifferentiate into myocytes and to promote neovascularization Citation13. These cells can be used either in the acute or chronic phase of the disease, as their collection is easy and their preparation is not very time‐consuming Citation7, Citation14. Only a Ficoll density separation in autologous serum is recommended Citation15. The great inconvenience is that using whole mononuclear cells it is difficult to analyze the exact role of the different subpopulations in the clinical process.

Table I. Flow cytometry surface expression profile of the different bone marrow cell types used in clinical practice.

Mobilized peripheral blood progenitor cells

Under steady state conditions hematopoietic progenitors are in the bone marrow but can occasionally be detected in circulation. The use of hematopoietic growth factors (for example granulocyte colony‐stimulating factor (G‐CSF)) facilitates the egress of hematopoietic progenitors from bone marrow to peripheral blood. Mobilizing with growth factors could even avoid any direct delivery approach in the coronary artery system or the myocardium.

Selected bone marrow or peripheral blood subpopulations

The expression of some surface antigens CD34, CD133, CD117, characterizes a heterogeneous population of cells including hematopoietic progenitor cells and endothelial progenitor cells. These antigens have been used to isolate specific subpopulations of hematopoietic stem cells. It has been claimed that a subset of CD133+ cells represents endothelial progenitor cells, and the selection method has also been used to enrich the progenitors after mobilization or from bone marrow mononuclear cells.

Bone marrow mesenchymal stem cells

These are a rare population (0.01%–0.001%) of cells from bone marrow stroma, isolated for their capacity of plastic adhesion and their immunophenotype. Mesenchymal stem cells are negative for the markers of hematopoietic stem cells (CD34 and CD133); in fact, these cells lack specific surface markers and have an immunophenotype positive for many adhesion proteins. Bone marrow mesenchymal cells need to be expanded in culture and could only be considered for use in patients with acute myocardial infarction in the allogeneic setting if it is shown that they really are immune‐privileged and will not be rejected.

Adipose tissue‐derived mesenchymal stem cells

Recently, the existence of a population of cells has been seen, located in the adipose tissue, which are able to differentiate into multiple cell lineages. The phenotype of adipose stem cells is also similar to that of bone marrow mesenchymal cells. What makes this source so attractive is the possibility to extract large volumes of tissue and cells avoiding culture.

Cardiac resident stem cells

Several reports have demonstrated the existence of cycling ventricular myocytes in both the normal and the pathologic adult heart, challenging the classic dogma which stated that the adult mammalian heart is an organ without regenerative capacity. Their capability to differentiate into cardiomyocytes or vascular lineages suggests that these cells could be used for cardiac tissue repair Citation16. To date, several subpopulations have been identified: c‐kit cells, stem cell antigen‐1 (sca‐1), side population (SP) cells, cardiospheres, and Isl‐1+ cells Citation16. In spite of their presence in adult heart, spontaneous total recovery after myocardial injury does not occur since a very insufficient number of cardiac stem cells is available. However, they could potentially be expanded in culture or stimulated with growth factors.

Although different types of stem cells can be used in clinical practice, we do not know at present which cell subtype is the more efficient. Each subtype has advantages and pitfalls, but one important issue is the time needed for their preparation: if it is time‐consuming they cannot be used in the acute clinical setting. Moreover, in conditions where early microvascular reperfusion or chronic ischemia prevails, the angiogenic potential of bone marrow‐derived cells may be of high priority. However, in patients where additional restoration of contractile function is the clinical goal, such as those with ischemic cardiomyopathy and heart failure or early post infarction when blood flow has been restored but cardiocytes have died, delivering cells with contractile potential seems a more reasonable approach; under these conditions, skeletal myoblasts, or any progenitor cells driven down a muscle lineage appear a better first choice.

Key messages

  • The principal differences between the available clinical studies have been the type of patients studied, the type of cell used, the delivery approach, and the method of measurement.

  • Two main stem cell sources are available: embryonic and adult stem cells. To date, no cardiovascular clinical experience with embryonic cells is available, while the currently used adult stem cell sources in clinical trials have been skeletal myoblasts and bone marrow‐derived stem cells.

  • Clinical data show that this therapy is a feasible and safe procedure that seems to diminish significantly the extent of left ventricular remodeling, promote recovery of both global and segmental cardiac function, and improve myocardial perfusion. At this point experts advocate conducting intermediate‐size, randomized controlled trials to establish the effects of stem cell therapy using surrogate end points, not forgetting safety concerns.

Mechanism of action

According to the classical view, adult stem or progenitor cells seemed to have more limited potential than embryonic cells, being able to generate cells only within the same lineage. However, in recent years several studies have shown that circumstances such as tissue injury may act as a trigger for adult stem cell differentiation into specialized cells from embryological unrelated tissues. When metaplasia (conversion of one cell type into another) happens to a differentiated cell the process is named transdifferentiation. Bone marrow‐derived stem cells (hematopoietic and mesenchymal) appear as a representative example of plasticity given their outstanding capability to give rise to a wide range of cell types from different organs. Indeed, some authors suggest that the mechanism by which partially or totally differentiated cells change their phenotype might be a process of in vivo stem cell fusion with differentiated host cells Citation17. To date it is unknown whether transdifferentiation or cell fusion is the main mechanism of cardiac repair. Nevertheless, the positive influence of stem cell therapy could hardly be explained only thanks to cell replacement, as the number of cells (approximately 3%) that remain in the cardiac tissue is limited and insufficient in hearts with large injuries Citation18. A paracrine effect of extracardiac progenitors on cardiac resident stem cells might have something to do with the benefits Citation19. In this way, such mechanisms seem to be complementary rather than opposite, and their understanding becomes the main challenge in the future. Therefore, although regeneration does exist, it is yet to be determined whether cardiac regeneration or repair is responsible for the benefit.

Delivery approach strategies

The aim of any route of stem cell delivery is to implant a sufficient number of cells to gain the maximum possible retention in the region of the myocardium to be treated. The local host is a crucial determinant of the cellular retention since it determines cellular survival in the short term, cellular adhesion and transmigration across the vascular wall, and cellular tissue invasion.

To date, two principal routes of administration have been used: the transvascular approach and a direct injection in the ventricular wall.

Transvascular approaches

Intracoronary infusion

Obviously, to administer the cells via the coronary arteries they have to be patent. Thus, an intracoronary percutaneous revascularization of the culprit artery is a prerequisite to ensure a vascular access through which the stem cells will be given. An over‐the‐wire angioplasty balloon is used to deliver the cells into the coronary artery. Alternating periods of occlusion of the artery and infusion of the cells (2–3 minutes) with periods of reperfusion (1–3 minutes) are used until the total dose is given. Currently, intracoronary administration of hematopoietic progenitors represents the best‐tried method of delivery in the acute setting.

Intravenous infusion

In animal models the intravenous delivery of progenitor and mesenchymal cells has shown to improve left ventricular function after an acute coronary syndrome Citation4, Citation20. However, the application in clinical studies is limited considering seeding in organs other than the heart. In a recent study myocardial homing was not observed after intravenous administration when compared to intracoronary infusion Citation21.

Mobilization with growth factors

The pharmacological mobilization of bone marrow stem cells with growth colony‐stimulating factor is a very attractive alternative considering its easy administration (subcutaneously) avoiding any invasive procedure. The process by which this mobilization takes place is not completely understood, but it is well known that it is mediated by adhesion molecules and through trophic chemokines Citation22.

Direct injection in the ventricular wall

This approach should be preferred when the coronary vascularization avoids intracoronary infusion. However, the direct injection in the ischemic myocardium could create cell islets with scarce blood flow that could limit cell survival Citation23. This technique allows big‐size cells to be injected (mesenchymal and myoblasts) that could produce microembolisms with the intracoronary delivery Citation24.

Transendocardial injection

The electromechanical mapping of the endocardial surface using the NOGA® system (Cordis Corporation's Biologics Delivery Systems) allows viable, ischemic, and scar myocardium to be defined before the injection through a catheter positioned in the left ventricle. This approach has been used in patients with chronic heart disease.

Transepicardial injection

This approach is used in patients going to coronary artery bypass grafting surgery (CABG). The injection procedure is generally performed after all the graft sutures have been done, before removing the extracorporeal circulation, and while the heart is initiating spontaneous heartbeat. The injection area is often easily identifiable as a patchy fibrotic area with a dyskinetic or akinetic wall motion assessed by the specific imaging techniques. This approach allows a large amount of stem cells to be delivered, but there are myocardial regions not accessible (i.e. the interventricular septum).

Transvenous injection

This approach has been experimented in a pilot study in patients with ischemic cardiomyopathy using myoblasts and guided with intravascular ultrasound. In contrast to the transepicardial injection, with this method cells are injected in parallel to the interventricular septum.

Stem cell delivery is a critical step toward the optimization of the therapy and techniques to find out the destiny of stem cells will be crucial if we want to understand the mechanisms of action and to improve the short‐ and long‐term efficacy of this treatment. Stem cells labeled with various substances have been tracked in animal models by means of magnetic resonance, positron emission tomography, single‐photon emission computed tomography, and bioluminescent imaging Citation25. However, stem cells infused into humans have been tracked in only one study to date Citation21. Unfractionated cells and CD34+ cells were labeled with 18F‐fluorodeoxyglucose and infused via an antecubital vein or a coronary artery after a first myocardial infarction in nine patients. After intracoronary transfer only 1.3%–2.6% of unselected bone marrow stem cells and 14%–39% of CD34 cells were detected in the infarcted myocardium; the remaining activity was found primarily in liver and spleen. Furthermore, only background activity was detected in the infarcted myocardium after intravenous transfer. Integration of cell imaging into studies of cardiac cell therapy is necessary to further growth of the field towards optimizing routes of stem cell delivery.

Clinical studies—efficacy and safety

The evidence that stem cells may reconstitute necrotic myocardium and improve cardiac function in animals has led to an initiation of clinical studies, which have been mainly focused on addressing the feasibility and the safety of this therapy. The vast majority of studies have been performed in patients with acute myocardial infarction (Table ). Other important groups of studies have been performed in patients with chronic ischemic cardiomyopathy (Tables  and ). Thus, the principal differences between these studies were the clinical scenario, type of cell employed, the delivery approach, and the method of efficacy measurement (Figure ).

Table II. Cell therapy clinical trials in humans after acute myocardial infarction.

Table III. Clinical trials with direct intramyocardial stem cell injection in humans with chronic ischemic heart disease.

Table IV. Clinical trials with intracoronary administration and/or intravascular mobilization of stem cells in humans with chronic ischemic heart disease.

Figure 2 The principal differences between cardiovascular stem cell therapy clinical studies have been the type of patients studied, the type of cell used, the delivery approach, and the method of measurement. Three different clinical scenarios have been investigated to date: acute myocardial infarction, myocardial ischemia without revascularization possibilities, and ischemic cardiomyopathy with a depressed contractile function. The type of stem cell used could be classified in two groups: those bone marrow‐derived and myoblast. In conditions where reperfusion is the primary objective or chronic ischemia prevails, the angiogenic potential of bone marrow‐derived may be of high priority. In contrast, in patients where additional restoration of contractile function is the clinical goal, delivering cells with contractile potential like myoblast seems a more reasonable approach. The routes of stem cell delivery experimented have been percutaneous intracoronary infusion, transendocardial delivery through a left ventricle catheter, transvenous catheter approach, transepicardial delivery during surgery, and pharmacological mobilization with stimulating factors. Finally, to assess the efficacy of stem cell therapy, different imaging techniques have been used either to measure perfusion or left ventricular function.

Figure 2 The principal differences between cardiovascular stem cell therapy clinical studies have been the type of patients studied, the type of cell used, the delivery approach, and the method of measurement. Three different clinical scenarios have been investigated to date: acute myocardial infarction, myocardial ischemia without revascularization possibilities, and ischemic cardiomyopathy with a depressed contractile function. The type of stem cell used could be classified in two groups: those bone marrow‐derived and myoblast. In conditions where reperfusion is the primary objective or chronic ischemia prevails, the angiogenic potential of bone marrow‐derived may be of high priority. In contrast, in patients where additional restoration of contractile function is the clinical goal, delivering cells with contractile potential like myoblast seems a more reasonable approach. The routes of stem cell delivery experimented have been percutaneous intracoronary infusion, transendocardial delivery through a left ventricle catheter, transvenous catheter approach, transepicardial delivery during surgery, and pharmacological mobilization with stimulating factors. Finally, to assess the efficacy of stem cell therapy, different imaging techniques have been used either to measure perfusion or left ventricular function.

Among the trials performed in patients with acute myocardial infarction, all cases have been performed with the culprit artery patent. Most have been phase I studies analyzing feasibility and safety. The delivery approaches used have essentially been two: the intracoronary injection and the mobilization of stem cells using G‐CSF. The types of stem cells that have been used are bone marrow‐derived. Regarding the design of the trials, most studies have not been randomized, though randomized trials involving a considerable number of patients exist, and although general controversial results have been found, some benefits in ejection fraction improvement or remodeling related parameters have been observed. Recently, the largest study of cardiac cell therapy to date administering hematopoietic progenitors intracoronarily after acute myocardial infarction has been published Citation26. The REPAIR‐AMI trial, designed as a randomized, double‐blinded study, including 204 patients from 17 European centers, showed a significant increase in the left ventricular ejection fraction (5.5±0.7% versus 3±0.7%; P = 0.014) among patients treated with cells compared to those given placebo. However, other contemporary studies, although reinforcing the message that bone marrow stem cell infusion is not only feasible but also safe, failed to show significant improvement in left ventricular function Citation27. Importantly, technical differences in the characteristics or handling of the infused stem cells might explain the different outcomes observed Citation28.

These conflicting results are similar to those found in patients receiving G‐CSF. Four studies published within the last year did not find any differences in the left ventricular function among patients treated with the factor or placebo Citation29–32. Possible causes to argue these differences could be absence of homing signals, timing of drug administration (in the study which showed a benefit, G‐CSF was administered immediately after myocardial infarction, while in others administration was delayed for 3–5 days after revascularization), subpopulations of stem cells mobilized not effective, and objectives of the study not well established.

Simultaneously to the studies performed in patients with acute myocardial infarction, some groups have tried to avoid established left ventricular remodeling as it happens in the chronic ischemic cardiomyopathy, or to improve myocardial perfusion as it happens in patients with chronic ischemia and no revascularization options. In this chronic clinical scenario the variability of stem cell and delivery approaches used is higher, but in general direct intramyocardial injection (Table ) or a transvascular approach (Table ) have been used. The first trials were done using myoblasts and bone marrow‐derived progenitors directly injected in the infarcted myocardium during CABG surgery. Other studies have evaluated the transendocardial delivery safety also using myoblasts or bone marrow progenitors, and recently experiences have been reported on infusing progenitor cells intracoronarily after opening the coronary artery chronically occluded and with myoblasts transvenously (Tables  and ). However, only one double‐blinded, randomized clinical study in this setting has been reported to date Citation33. But despite the positive results observed in this study, it is important to notice that no definitive conclusions can be reached due to study design limitations. Thus, we will have to wait until randomized, double‐blinded ongoing studies are fully reported.

Safety

When a new therapeutic strategy gets started, safety becomes the main goal of every single investigation testing that treatment. Undoubtedly, that long‐term stem cell therapy is safe is far from being proved. Therefore, current ongoing randomized mechanistic intermediate‐size trials on stem cell regenerative therapy must focus not only on remodeling and perfusion but also on safety. A shared database with all complications of stem cell treatment in humans should be a priority of all investigators involved in this field. Trials undertaken so far shed light on several complications which cast doubts on regenerative therapy safety in humans: arrhythmias, restenosis and atherosclerosis progression, and nontarget organ seeding.

Arrhythmias have been mainly reported in clinical trials using myoblasts for cardiac repair. It is admitted that myoblasts do not become coupled with the surrounding myocytes providing a putative arrhythmogenic substrate Citation9, Citation10. However, it is not clear that these in vitro observations are translated to the clinical setting. Although the first study by Menache observed a high incidence of ventricular arrhythmias in patients with severely depressed ejection fraction Citation8, no arrhythmias have been reported when myoblasts were cultured without bovine serum Citation34. Furthermore, in the MAGIC trial the incidence of arrythmias was similar in the active group compared with the placebo group (personal communication). Thus far, this adverse effect could not be exclusive of myoblast. In our ongoing experience Citation35, we have intracoronarily transplanted autologous bone marrow mononuclear cells in 72 patients following ST‐segment elevation acute myocardial infarction. Four patients showed delayed episodes of ventricular arrhythmias, and three were implanted with an internal defibrillator. Recently, Bartunek et al., intracoronarily delivering CD133+ in patients with acute myocardial infarction, have reported two patients presenting ventricular tachycardia Citation36. Such adverse events required us to be extremely cautious until future larger controlled trials assist us in identifying the risk attributable to cell replacement therapy and to the arrhythmogenic substrate.

A small clinical trial with ten patients showed a high rate of in‐stent restenosis after G‐CSF administration Citation37. Whether this finding is related to a wrong timing of G‐CSF injection 4 days before stent implantation remains a matter of conjecture. In this regard, further randomized trials of G‐CSF in patients with acute myocardial infarction treated with primary angioplasty with stent have showed a similar binary restenosis rate in both the active and the control groups Citation29–32, Citation38, Citation39. Regarding bone marrow stem cells, a low restenosis rate has been published in initial studies with angiographic follow‐up Citation27, Citation30. A recent nonrandomized matched trial in acute myocardial infarction raises concerns on this issue, though. After intracoronary injection of CD133+ cells, despite a significant improvement in remodeling and perfusion parameters, unexpected rates of 37% in‐stent restenosis and 11% reocclusion were found Citation36. The use of bare metal stents and CD133+ cells which carry a high angiogenic potential may in part account for these apparent discrepancies.

A rather high incidence of de novo lesions at nonstented arteries has been found after stem cell transplantation Citation15, Citation36. The capability of stem cells on angiographically smooth arteries to induce or accelerate atherosclerosis cannot be neglected. Only randomized trials using angiographic and intravascular ultrasound end points will provide information on this topic.

Molecular imaging with radioactively labeled stem cells in humans showed that the percentage cells homing to the heart is quite low with other organs (kidney, liver, and spleen) being the receptors of the vast majority of progenitors Citation21. The clinical consequences, if any, of this unintended nontarget organ homing are not known.

Stem cell therapy for cardiac repair into perspective

Much excitement has surrounded recent breakthroughs in the field of adult stem cell research and their implications for cell therapy in patients with acute and chronic ischemic heart disease. In the last 5 years numerous clinical studies have been performed in order to evaluate the feasibility and safety of the different stem cell techniques in the acute or chronic cardiovascular setting, with most studies being small‐size and nonrandomized.

Available clinical data show that this therapy is a feasible and safe procedure (with unresolved concerns on arrythmias and restenosis) that seems to diminish significantly the extent of left ventricular remodeling, promote recovery of both global and segmental cardiac function, and improve myocardial perfusion. At this point experts advocate to no longer perform studies involving small numbers of patients, but rather to conduct intermediate‐size, randomized controlled trials to establish the effects of stem cell therapy using surrogate end points, not forgetting safety concerns Citation40.

These trials should be compatible with simultaneous mechanistic and basic research aimed at addressing some crucial questions still unsettled: How do adult stem cells contribute to cardiac repair? At least six different mechanisms might be proposed: transdifferentiation, de‐differentiation, transdetermination, cell fusion, true pluripotent stem cell behavior, and the production of trophic factors. It is likely that our current knowledge of cell markers is inadequate to define cell populations accurately, so it is possible that cells with true multipotency may have contaminated experiments previously reported as examples of transdifferentiation. What will be the ideal source of adult stem cells? Bone marrow stem cells would seem simple, cheap, and apparently widely applicable. What is the best route of administration? When should cell transplantation be performed? Experimental studies suggest that stem cell transplantation is most successful after the inflammatory reaction is resolved but before scar expansion. How many times should cell transplantation be performed? Several clinical studies have demonstrated that stem cells can improve cardiac function in patients with old myocardial infarction, so successive transplantations may improve the benefit of this therapy.

We are optimistic that stem cell transplantation is likely to be of future benefit and that new tools, such as magnetic resonance, will help to refine patient selection and lead to a better assessment of results. In this sense, as pointed out by others Citation41, there is no doubt that the success of stem cell therapy will rest on its ability to show clinical efficacy. Thus, the question is whether it is time to perform large‐scale, randomized clinical trials, involving more than a thousand patients, to establish the impact on patient outcomes. In our point of view, that time has not yet come, considering that most studies have included patients with good prognosis, out of the hyperacute phase, and considering the variability in the sources of cells used and the number and quality of cells obtained. Therefore, too much hurry and neutral results could irreversibly damage this promising field of investigation that is currently far from its optimal application method.

References

  • Rogers W. J., Canto J. G., Lambrew C. T., Tiefenbrunn A. J., Kinkaid B., Shoultz D. A., et al. Temporal trends in the treatment of over 1.5 million patients with myocardial infarction in the US from 1990 through 1999: the National Registry of Myocardial Infarction 1, 2 and 3. J Am Coll Cardiol 2000; 36: 2056–63
  • Beltrami A. P., Urbanek K., Kajstura J., Yan S. M., Finato N., Bussani R., et al. Evidence that human cardiac myocytes divide after myocardial infarction. N Engl J Med 2001; 344: 1750–7
  • Anversa P., Nadal‐Ginard B. Myocyte renewal and ventricular remodelling. Nature 2002; 415: 240–3
  • Kocher A. A., Schuster M. D., Szabolcs M. J., Takuma S., Burkhoff D., Wang J., et al. Neovascularization of ischemic myocardium by human bone‐marrow‐derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med 2001; 7: 430–6
  • Orlic D., Kajstura J., Chimenti S., Jakoniuk I., Anderson S. M., Li B., et al. Bone marrow cells regenerate infarcted myocardium. Nature 2001; 410: 701–5
  • Orlic D., Kajstura J., Chimenti S., Limana F., Jakoniuk I., Quaini F., et al. Mobilized bone marrow cells repair the infarcted heart, improving function and survival. Proc Natl Acad Sci U S A 2001; 98: 10344–9
  • Sanchez P. L., San Roman J. A., Villa A., Fernandez M. E., Fernandez‐Aviles F. Contemplating the bright future of stem cell therapy for cardiovascular disease. Nat Clin Pract Cardiovasc Med 2006; 3 Suppl 1: S138–51
  • Menasche P., Hagege A. A., Vilquin J. T., Desnos M., Abergel E., Pouzet B., et al. Autologous skeletal myoblast transplantation for severe postinfarction left ventricular dysfunction. J Am Coll Cardiol 2003; 41: 1078–83
  • Fernandes S., Amirault J. C., Lande G., Nguyen J. M., Forest V., Bignolais O., et al. Autologous myoblast transplantation after myocardial infarction increases the inducibility of ventricular arrhythmias. Cardiovasc Res 2006; 69: 348–58
  • Fouts K., Fernandes B., Mal N., Liu J., Laurita K. R. Electrophysiological consequence of skeletal myoblast transplantation in normal and infarcted canine myocardium. Heart Rhythm 2006; 3: 452–61
  • de la Fuente R., Abad J. L., Garcia‐Castro J., Fernandez‐Miguel G., Petriz J., Rubio D., et al. Dedifferentiated adult articular chondrocytes: a population of human multipotent primitive cells. Exp Cell Res 2004; 297: 313–28
  • Villaron E. M., Almeida J., Lopez‐Holgado N., Alcoceba M., Sanchez‐Abarca L. I., Sanchez‐Guijo F. M., et al. Mesenchymal stem cells are present in peripheral blood and can engraft after allogeneic hematopoietic stem cell transplantation. Haematologica 2004; 89: 1421–7
  • Schatteman G. C., Awad O. Hemangioblasts, angioblasts, and adult endothelial cell progenitors. Anat Rec A Discov Mol Cell Evol Biol 2004; 276: 13–21
  • Sanchez P. L., Sanchez‐Guijo F. M., Villa A., Del Canizo C., Arnold R., San Roman J. A., et al. Launching a clinical program of stem cell therapy for cardiovascular repair. Nat Clin Pract Cardiovasc Med 2007; 4 Suppl 1: S123–9
  • Fernandez‐Aviles F., San Roman J. A., Garcia‐Frade J., Fernandez M. E., Penarrubia M. J., de la Fuente L., et al. Experimental and clinical regenerative capability of human bone marrow cells after myocardial infarction. Circ Res 2004; 95: 742–8
  • Torella D., Ellison G. M., Mendez‐Ferrer S., Ibanez B., Nadal‐Ginard B. Resident human cardiac stem cells: role in cardiac cellular homeostasis and potential for myocardial regeneration. Nat Clin Pract Cardiovasc Med 2006; 3 Suppl 1: S8–13
  • Fuchs E., Tumbar T., Guasch G. Socializing with the neighbors: stem cells and their niche. Cell 2004; 116: 769–78
  • Beltrami A. P., 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: 763–76
  • Chien K. R. Lost and found: cardiac stem cell therapy revisited. J Clin Invest 2006; 116: 1838–40
  • Pittenger M. F., Martin B. J. Mesenchymal stem cells and their potential as cardiac therapeutics. Circ Res 2004; 95: 9–20
  • Hofmann M., Wollert K. C., Meyer G. P., Menke A., Arseniev L., Hertenstein B., et al. Monitoring of bone marrow cell homing into the infarcted human myocardium. Circulation 2005; 111: 2198–202
  • Flomenberg N., DiPersio J., Calandra G. Role of CXCR4 chemokine receptor blockade using AMD3100 for mobilization of autologous hematopoietic progenitor cells. Acta Haematol 2005; 114: 198–205
  • Bel A., Messas E., Agbulut O., Richard P., Samuel J. L., Bruneval P., et al. Transplantation of autologous fresh bone marrow into infarcted myocardium: a word of caution. Circulation 2003; 108 Suppl 1: II247–52
  • Vulliet P. R., Greeley M., Halloran S. M., MacDonald K. A., Kittleson M. D. Intra‐coronary arterial injection of mesenchymal stromal cells and microinfarction in dogs. Lancet 2004; 363: 783–4
  • Zhou R., Acton P. D., Ferrari V. A. Imaging stem cells implanted in infarcted myocardium. J Am Coll Cardiol 2006; 48: 2094–106
  • Schachinger V., Erbs S., Elsasser A., Haberbosch W., Hambrecht R., Holschermann H., et al. Intracoronary bone marrow‐derived progenitor cells in acute myocardial infarction. N Engl J Med 2006; 355: 1210–21
  • Lunde K., Solheim S., Aakhus S., Arnesen H., Abdelnoor M., Egeland T., et al. Intracoronary injection of mononuclear bone marrow cells in acute myocardial infarction. N Engl J Med 2006; 355: 1199–209
  • Seeger F. H., Tonn T., Krzossok N., Zeiher A. M., Dimmeler S. Cell isolation procedures matter: a comparison of different isolation protocols of bone marrow mononuclear cells used for cell therapy in patients with acute myocardial infarction. Eur Heart J 2007; 28: 766–72
  • Ripa R. S., Jorgensen E., Wang Y., Thune J. J., Nilsson J. C., Sondergaard L., et al. Stem cell mobilization induced by subcutaneous granulocyte‐colony stimulating factor to improve cardiac regeneration after acute ST‐elevation myocardial infarction: result of the double‐blind, randomized, placebo‐controlled stem cells in myocardial infarction (STEMMI) trial. Circulation 2006; 113: 1983–92
  • Zohlnhofer D., Ott I., Mehilli J., Schomig K., Michalk F., Ibrahim T., et al. Stem cell mobilization by granulocyte colony‐stimulating factor in patients with acute myocardial infarction: a randomized controlled trial. JAMA 2006; 295: 1003–10
  • Ellis S. G., Penn M. S., Bolwell B., Garcia M., Chacko M., Wang T., et al. Granulocyte colony stimulating factor in patients with large acute myocardial infarction: results of a pilot dose‐escalation randomized trial. Am Heart J 2006; 152: 1051 e9–14
  • Engelmann M. G., Theiss H. D., Hennig‐Theiss C., Huber A., Wintersperger B. J., Werle‐Ruedinger A. E., 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: 1712–21
  • Assmus B., Honold J., Schachinger V., Britten M. B., Fischer‐Rasokat U., Lehmann R., et al. Transcoronary transplantation of progenitor cells after myocardial infarction. N Engl J Med 2006; 355: 1222–32
  • Herreros J., Prosper F., Perez A., Gavira J. J., Garcia‐Velloso M. J., Barba J., et al. Autologous intramyocardial injection of cultured skeletal muscle‐derived stem cells in patients with non‐acute myocardial infarction. Eur Heart J 2003; 24: 2012–20
  • Bartunek J., Vanderheyden M., Wijns W., Timmermans F., Vandekerkhove B., Villa A., et al. Bone‐marrow‐derived cells for cardiac stem cell therapy: safe or still under scrutiny?. Nat Clin Pract Cardiovasc Med 2007; 4 Suppl 1: S100–5
  • Bartunek J., Vanderheyden M., Vandekerckhove B., Mansour S., De Bruyne B., De Bondt P., et al. Intracoronary injection of CD133‐positive enriched bone marrow progenitor cells promotes cardiac recovery after recent myocardial infarction: feasibility and safety. Circulation 2005; 112 Suppl: I178–83
  • Kang H. J., Kim H. S., Zhang S. Y., Park K. W., Cho H. J., Koo B. K., 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: 751–6
  • Ince H., Petzsch M., Kleine H. D., Eckard H., Rehders T., Burska D., 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 Suppl: I73–80
  • Kang H. J., Kim H. S., Koo B. K., Kim Y. J., Lee D., Sohn D. W., et al. 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: 237.e1–8
  • Bartunek J., Dimmeler S., Drexler H., Fernandez‐Aviles F., Galinanes M., Janssens S., et al. The consensus of the task force of the European Society of Cardiology concerning the clinical investigation of the use of autologous adult stem cells for repair of the heart. Eur Heart J 2006; 27: 1338–40
  • Rosenzweig A. Cardiac cell therapy—mixed results from mixed cells. N Engl J Med 2006; 355: 1274–7
  • Strauer B. E., Brehm M., Zeus T., Gattermann N., Hernandez A., Sorg R. V., et al. [Intracoronary, human autologous stem cell transplantation for myocardial regeneration following myocardial infarction]. Dtsch Med Wochenschr 2001; 126: 932–8
  • Strauer B. E., Brehm M., Zeus T., Kostering M., Hernandez A., Sorg R. V., et al. Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans. Circulation 2002; 106: 1913–8
  • Assmus B., Schachinger V., Teupe C., Britten M., Lehmann R., Dobert N., et al. Transplantation of Progenitor Cells and Regeneration Enhancement in Acute Myocardial Infarction (TOPCARE‐AMI). Circulation 2002; 106: 3009–17
  • Schachinger V., Assmus B., Britten M. B., Honold J., Lehmann R., Teupe C., 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: 1690–9
  • Chen S. L., Fang W. W., Ye F., Liu Y. H., Qian J., Shan S. J., 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: 92–5
  • Kuethe F., Richartz B. M., Sayer H. G., Kasper C., Werner G. S., Hoffken K., et al. Lack of regeneration of myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans with large anterior myocardial infarctions. Int J Cardiol 2004; 97: 123–7
  • Aviles F. F., San Roman J. A., Garcia Frade J., Valdes M., Sanchez A., de la Fuente L., et al. [Intracoronary stem cell transplantation in acute myocardial infarction]. Rev Esp Cardiol 2004; 57: 201–8
  • Wollert K. C., Meyer G. P., Lotz J., Ringes‐Lichtenberg S., Lippolt P., Breidenbach C., et al. Intracoronary autologous bone‐marrow cell transfer after myocardial infarction: the BOOST randomised controlled clinical trial. Lancet 2004; 364: 141–8
  • Meyer G. P., Wollert K. C., Lotz J., Steffens J., Lippolt P., Fichtner S., 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: 1287–94
  • Katritsis D. G., Sotiropoulou P. A., Karvouni E., Karabinos I., Korovesis S., Perez S. A., et al. Transcoronary transplantation of autologous mesenchymal stem cells and endothelial progenitors into infarcted human myocardium. Catheter Cardiovasc Interv 2005; 65: 321–9
  • Ge J., Li Y., Qian J., Shi J., Wang Q., Niu Y., et al. Efficacy of emergent transcatheter transplantation of stem cells for treatment of acute myocardial infarction (TCT‐STAMI). Heart 2006; 92: 1764–7
  • Janssens S., Dubois C., Bogaert J., Theunissen K., Deroose C., Desmet W., et al. Autologous bone marrow‐derived stem‐cell transfer in patients with ST‐segment elevation myocardial infarction: double‐blind, randomised controlled trial. Lancet 2006; 367: 113–21
  • Schachinger V., Erbs S., Elsasser A., Haberbosch W., Hambrecht R., Holschermann H., 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: 2775–83
  • Kuethe F., Figulla H. R., Voth M., Richartz B. M., Opfermann T., Sayer H. G., et al. [Mobilization of stem cells by granulocyte colony‐stimulating factor for the regeneration of myocardial tissue after myocardial infarction]. Dtsch Med Wochenschr 2004; 129: 424–8
  • Kuethe F., Figulla H. R., Herzau M., Voth M., Fritzenwanger M., Opfermann T., et al. Treatment with granulocyte colony‐stimulating factor for mobilization of bone marrow cells in patients with acute myocardial infarction. Am Heart J 2005; 150: 115
  • Valgimigli M., Rigolin G. M., Cittanti C., Malagutti P., Curello S., Percoco G., et al. Use of granulocyte‐colony stimulating factor during acute myocardial infarction to enhance bone marrow stem cell mobilization in humans: clinical and angiographic safety profile. Eur Heart J 2005; 26: 1838–45
  • Suarez de Lezo J., Torres A., Herrera I., Pan M., Romero M., Pavlovic D., et al. [Effects of stem‐cell mobilization with recombinant human granulocyte colony stimulating factor in patients with percutaneously revascularized acute anterior myocardial infarction]. Rev Esp Cardiol 2005; 58: 253–61
  • Ince H., Petzsch M., Kleine H. D., Schmidt H., Rehders T., Korber T., et al. Preservation from left ventricular remodeling by front‐integrated revascularization and stem cell liberation in evolving acute myocardial infarction by use of granulocyte‐colony‐stimulating factor (FIRSTLINE‐AMI). Circulation 2005; 112: 3097–106
  • Jorgensen E., Ripa R. S., Helqvist S., Wang Y., Johnsen H. E., Grande P., 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
  • Kang H. J., Lee H. Y., Na S. H., Chang S. A., Park K. W., Kim H. K., 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 Suppl: I145–51
  • Steinwender C., Hofmann R., Kammler J., Kypta A., Pichler R., Maschek W., et al. Effects of peripheral blood stem cell mobilization with granulocyte‐colony stimulating factor and their transcoronary transplantation after primary stent implantation for acute myocardial infarction. Am Heart J 2006; 151: 1296.e7–13
  • Hagege A. A., Marolleau J. P., Vilquin J. T., Alheritiere A., Peyrard S., Duboc D., et al. Skeletal myoblast transplantation in ischemic heart failure: long‐term follow‐up of the first phase I cohort of patients. Circulation 2006; 114 Suppl: I108–13
  • Gavira J. J., Herreros J., Perez A., Garcia‐Velloso M. J., Barba J., Martin‐Herrero F., et al. Autologous skeletal myoblast transplantation in patients with nonacute myocardial infarction: 1‐year follow‐up. J Thorac Cardiovasc Surg 2006; 131: 799–804
  • Siminiak T., Kalawski R., Fiszer D., Jerzykowska O., Rzezniczak J., Rozwadowska N., et al. Autologous skeletal myoblast transplantation for the treatment of postinfarction myocardial injury: phase I clinical study with 12 months of follow‐up. Am Heart J 2004; 148: 531–7
  • Chachques J. C., Herreros J., Trainini J., Juffe A., Rendal E., Prosper F., et al. Autologous human serum for cell culture avoids the implantation of cardioverter‐defibrillators in cellular cardiomyoplasty. Int J Cardiol 2004; 95 Suppl 1: S29–33
  • Dib N., Michler R. E., Pagani F. D., Wright S., Kereiakes D. J., Lengerich R., et al. Safety and feasibility of autologous myoblast transplantation in patients with ischemic cardiomyopathy: four‐year follow‐up. Circulation 2005; 112: 1748–55
  • Stamm C., Westphal B., Kleine H. D., Petzsch M., Kittner C., Klinge H., et al. Autologous bone‐marrow stem‐cell transplantation for myocardial regeneration. Lancet 2003; 361: 45–6
  • Stamm C., Kleine H. D., Westphal B., Petzsch M., Kittner C., Nienaber C. A., et al. CABG and bone marrow stem cell transplantation after myocardial infarction. Thorac Cardiovasc Surg 2004; 52: 152–8
  • 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. Am Heart J 2006; 151: 192–7
  • Patel A. N., Geffner L., Vina R. F., Saslavsky J., Urschel H. C Jr., Kormos R., et al. Surgical treatment for congestive heart failure with autologous adult stem cell transplantation: a prospective randomized study. J Thorac Cardiovasc Surg 2005; 130: 1631–8
  • Archundia A., Aceves J. L., Lopez‐Hernandez M., Alvarado M., Rodriguez E., Diaz Quiroz G., et al. Direct cardiac injection of G‐CSF mobilized bone‐marrow stem‐cells improves ventricular function in old myocardial infarction. Life Sci 2005; 78: 279–83
  • Galinanes M., Loubani M., Davies J., Chin D., Pasi J., Bell P. R. Autotransplantation of unmanipulated bone marrow into scarred myocardium is safe and enhances cardiac function in humans. Cell Transplant 2004; 13: 7–13
  • Hamano K., Nishida M., Hirata K., Mikamo A., Li T. S., Harada M., et al. Local implantation of autologous bone marrow cells for therapeutic angiogenesis in patients with ischemic heart disease: clinical trial and preliminary results. Jpn Circ J 2001; 65: 845–7
  • Li T. S., Hamano K., Hirata K., Kobayashi T., Nishida M. The safety and feasibility of the local implantation of autologous bone marrow cells for ischemic heart disease. J Card Surg 2003; 18 Suppl 2: S69–75
  • Ozbaran M., Omay S. B., Nalbantgil S., Kultursay H., Kumanlioglu K., Nart D., et al. Autologous peripheral stem cell transplantation in patients with congestive heart failure due to ischemic heart disease. Eur J Cardiothorac Surg 2004; 25: 342–50; discussion 350–1
  • Pompilio G., Cannata A., Peccatori F., Bertolini F., Nascimbene A., Capogrossi M. C., et al. Autologous peripheral blood stem cell transplantation for myocardial regeneration: a novel strategy for cell collection and surgical injection. Ann Thorac Surg 2004; 78: 1808–12
  • Siminiak T., Fiszer D., Jerzykowska O., Grygielska B., Rozwadowska N., Kalmucki P., 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: 1188–95
  • Smits P. C., van Geuns R. J., Poldermans D., Bountioukos M., Onderwater E. E., Lee C. H., et al. Catheter‐based intramyocardial injection of autologous skeletal myoblasts as a primary treatment of ischemic heart failure: clinical experience with six‐month follow‐up. J Am Coll Cardiol 2003; 42: 2063–9
  • Ince H., Petzsch M., Rehders T. C., Chatterjee T., Nienaber C. A. Transcatheter transplantation of autologous skeletal myoblasts in postinfarction patients with severe left ventricular dysfunction. J Endovasc Ther 2004; 11: 695–704
  • Perin E. C., Dohmann H. F., Borojevic R., Silva S. A., Sousa A. L., Mesquita C. T., et al. Transendocardial, autologous bone marrow cell transplantation for severe, chronic ischemic heart failure. Circulation 2003; 107: 2294–302
  • Perin E. C., Dohmann H. F., Borojevic R., Silva S. A., Sousa A. L., Silva G. V., 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 Suppl 1: II213–8
  • Silva G. V., Perin E. C., Dohmann H. F., Borojevic R., Silva S. A., Sousa A. L., et al. Catheter‐based transendocardial delivery of autologous bone‐marrow‐derived mononuclear cells in patients listed for heart transplantation. Tex Heart Inst J 2004; 31: 214–9
  • Tse H. F., Kwong Y. L., Chan J. K., Lo G., Ho C. L., Lau C. P. Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet 2003; 361: 47–9
  • Tse H. F., Thambar S., Kwong Y. L., Rowlings P., Bellamy G., McCrohon J., et al. Safety of catheter‐based intramyocardial autologous bone marrow cells implantation for therapeutic angiogenesis. Am J Cardiol 2006; 98: 60–2
  • Fuchs S., Satler L. F., Kornowski R., Okubagzi P., Weisz G., Baffour 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: 1721–4
  • Fuchs S., Kornowski R., Weisz G., Satler L. F., Smits P. C., Okubagzi P., et al. Safety and feasibility of transendocardial autologous bone marrow cell transplantation in patients with advanced heart disease. Am J Cardiol 2006; 97: 823–9
  • Briguori C., Reimers B., Sarais C., Napodano M., Pascotto P., Azzarello G., et al. Direct intramyocardial percutaneous delivery of autologous bone marrow in patients with refractory myocardial angina. Am Heart J 2006; 151: 674–80
  • Kuethe F., Richartz B. M., Kasper C., Sayer H. G., Hoeffken K., Werner G. S., et al. Autologous intracoronary mononuclear bone marrow cell transplantation in chronic ischemic cardiomyopathy in humans. Int J Cardiol 2005; 100: 485–91
  • Strauer B. E., Brehm M., Zeus T., Bartsch T., Schannwell C., Antke C., 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: 1651–8
  • Blatt A., Cotter G., Leitman M., Krakover R., Kaluski E., Milo‐Cotter O., et al. Intracoronary administration of autologous bone marrow mononuclear cells after induction of short ischemia is safe and may improve hibernation and ischemia in patients with ischemic cardiomyopathy. Am Heart J 2005; 150: 986
  • Erbs S., Linke A., Adams V., Lenk K., Thiele H., Diederich K. W., et al. Transplantation of blood‐derived progenitor cells after recanalization of chronic coronary artery occlusion: first randomized and placebo‐controlled study. Circ Res 2005; 97: 756–62
  • Seiler C., Pohl T., Wustmann K., Hutter D., Nicolet P. A., Windecker S., et al. Promotion of collateral growth by granulocyte‐macrophage colony‐stimulating factor in patients with coronary artery disease: a randomized, double‐blind, placebo‐controlled study. Circulation 2001; 104: 2012–7
  • Wang Y., Tagil K., Ripa R. S., Nilsson J. C., Carstensen S., Jorgensen E., et al. Effect of mobilization of bone marrow stem cells by granulocyte colony stimulating factor on clinical symptoms, left ventricular perfusion and function in patients with severe chronic ischemic heart disease. Int J Cardiol 2005; 100: 477–83
  • Hill J. M., Syed M. A., Arai A. E., Powell T. M., Paul J. D., Zalos G., et al. Outcomes and risks of granulocyte colony‐stimulating factor in patients with coronary artery disease. J Am Coll Cardiol 2005; 46: 1643–8
  • Joseph J., Rimawi A., Mehta P., Cottler‐Fox M., Sinha A., Singh B. K., et al. Safety and effectiveness of granulocyte‐colony stimulating factor in mobilizing stem cells and improving cytokine profile in advanced chronic heart failure. Am J Cardiol 2006; 97: 681–4

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.