Coronary heart disease and heart failure continue to be

Size: px
Start display at page:

Download "Coronary heart disease and heart failure continue to be"

Transcription

1 CONTROVERSIES IN CARDIOVASCULAR MEDICINE Is stem cell therapy ready for patients? Stem Cell Therapy for Cardiac Repair Ready for the Next Step Andrew J. Boyle, MBBS, PhD; Steven P. Schulman, MD; Joshua M. Hare, MD Coronary heart disease and heart failure continue to be significant burdens to healthcare systems in the Western world. In the United States alone, there are 7.1 million survivors of myocardial infarction (MI) and 4.9 million people living with congestive heart failure (CHF). 1 Despite recent advances in medical and device therapy for heart failure, the incidence, hospitalization, and mortality rates continue to rise. After receiving a diagnosis of CHF, 1 in 5 patients will be dead within 12 months. 1 Therefore, any new treatment modality that benefits heart failure patients has the potential to result in a dramatic improvement in health outcomes and substantial cost savings for the community. Response by Oettgen p 352 The possibility of using stem cell based therapies for people suffering an acute MI or living with CHF has captured the imagination of both the medical and popular communities. Since early reports in animal models 10 years ago, 2,3 the stem cell field has made enormous advances in moving toward clinically applicable treatment options, and we now stand at the dawn of a new therapeutic era. An abundance of preclinical data demonstrate safety, feasibility, and efficacy, justifying the current entry into clinical trials of stem cell therapy in humans. 4 8 This position, however, is extremely controversial, with some arguing that trials are premature because mechanistic insights are insufficiently addressed. 9,10 Here, we argue that properly conducted rigorous clinical trials are a key and appropriate next step not only to start the long process of therapeutic development but also as an essential component in the process of understanding the scientific underpinnings of cardiac regeneration and its therapeutic utilization. The field of regenerative medicine will advance through the parallel conduct of in vitro/animal model studies and clinical trials, the latter frequently guiding the former. The publication of Menasche et al 11 describing the first patients to receive skeletal myoblasts spawned a profusion of small clinical studies investigating cellular therapy for cardiac repair. At present, a number of early clinical studies have been published and are summarized in Table 1. Several points are immediately apparent from this table. First, 400 patients have completed these published studies, yet most of them are small pilot studies that lack randomization or control groups. Second, despite the fact that several cell types have been studied using different delivery methods, the overwhelming message from all of these studies is that cell therapy is safe and feasible. In addition, the results of these studies provide encouraging, albeit preliminary, signs of efficacy. Finally, although these trials represent the currently published data, they have formed the basis for numerous larger, ongoing trials accruing more patient data (Table 2). Mechanism of Action of Stem Cells One of the major obstacles in progressing to large-scale clinical trials of cardiac stem cell therapy is the ongoing debate regarding the mechanism of action by which stem cell therapy leads to cardiac repair. The classic idea that provided the primary motivation for stem cell therapy is that delivery of the appropriate stem cells would repair a damaged heart via The opinions expressed in this article are not necessarily those of the editors or of the American Heart Association. From the Department of Medicine, Division of Cardiology, Institute for Cell Engineering, and Specialized Center for Cell-Based Therapy, The Johns Hopkins University School of Medicine, Baltimore, Md. Correspondence to Joshua M. Hare, MD, Johns Hopkins Medical Institutions and Institute for Cell Engineering (ICE), Division of Cardiology, BRB 651, 733 N Broadway, Baltimore, MD jhare@mail.jhmi.edu (Circulation. 2006;114: ) 2006 American Heart Association, Inc. Circulation is available at DOI: /CIRCULATIONAHA

2 340 Circulation July 25, 2006 TABLE 1. Clinical Studies of Stem Cell Therapy for Cardiac Repair Study Treated, n Cell Type Mode of Delivery Randomized Primary End Point P Strauer et al BMMNCs Intracoronary Safety NA TOPCARE-AMI 51,52 59 BMMNCs/CPCs Intracoronary / Safety and feasibility NA Stamm et al 103, BMMNCs Open heart surgery Safety and feasibility NA Tse et al BMMNCs Endomyocardial Safety and feasibility NA Perin et al BMMNCs Endomyocardial Safety NA BOOST BMMNCs Intracoronary LVEF Fernandez-Aviles et al BMMNCs Intracoronary Safety and feasibility NA Li et al BMMNCs Open heart surgery Safety and feasibility NA IACT study BMMNCs Intracoronary Not stated NA Janssens et al BMMNCs Intracoronary LVEF 0.36 Chen et al MSCs Intracoronary LVEF 0.01 Katritsis et al MSCs EPCs Intracoronary Scar size 0.02 Galinanes et al Bone marrow Open heart surgery Safety NA Fuchs et al Bone marrow Endomyocardial Safety and feasibility NA MAGIC cell GCSF-mobilized PBMNCs Intracoronary Safety and feasibility NA Ozbaran et al GCSF-mobilized PBMNCs Open heart surgery Safety and feasibility NA Erb et al GCSF-mobilized CPCs Intracoronary CFR and LVEF 0.05 Boyle et al GCSF-mobilized CD34 Intracoronary Safety and feasibility NA Pompilio et al GCSF-mobilized AC133 Open heart surgery Safety and feasibility NA Dib et al Skeletal myoblasts Open heart surgery Safety and feasibility NA POZNAN Skeletal myoblasts Transcoronary-venous Safety and feasibility NA Ince et al Skeletal myoblasts Endomyocardial Safety and feasibility NA Siminiak et al Skeletal myoblasts Open heart surgery Safety and feasibility NA Smits et al Skeletal myoblasts Endomyocardial Safety and feasibility NA Menasche et al Skeletal myoblasts Open heart surgery Safety and feasibility NA Pagani et al Skeletal myoblasts Open heart surgery Histology NA Herreros et al Skeletal myoblasts Open heart surgery Safety and feasibility NA CPC indicates circulating progenitor cell; GCSF, granulocyte colony stimulating factor; PBMNC, peripheral blood mononuclear cell; CFR, coronary flow reserve; and LVEF, LV ejection fraction. active myocardial regeneration resulting from transdifferentiation of the administered stem cells. 4,12 14 However, new data have led to the recognition of alternative mechanisms of action (Figure 1): Exogenous stem cells may also stimulate proliferation of endogenous cardiac precursors or stem cells through neovascularization 6,15 or paracrine signaling actions. 16 In fact, observations in preclinical and clinical scenarios that all of these events occur allow us to generate a new concept that cellular therapy contributes to the restoration of stem cell niches, facilitating the ability of the heart to heal itself. 17 Still other mechanisms are proposed: Exogenous stem cells may lead to cardiac repair via fusion of donor cells with host cardiomyocytes. 18 Finally, other investigators suggest that the effects of stem cells are mediated by altering mechanical properties to strengthen the MI scar, thereby preventing deterioration in cardiac function 19 (see Figure 1). This ongoing debate about mechanism fuels the case for slowing the pace of clinical trials; ie, we should not pursue work in patients until we thoroughly understand, with a high degree of scientific precision, the outcome of cell therapy in in vitro systems and animal models. 9,10 We would suggest that we have reached the appropriate point in the development of cellular therapeutics to enter into the clinic; in fact, entry into the clinic is the next step that will guide our understanding of the mechanistic underpinning for effective cellular therapeutics. Types of Cells Contemplated for Cellular Regenerative Therapy Embryonic Stem Cell Embryonic stem (ES) cells are derived from the inner cell mass of the blastocyst-stage embryo, late in the first week after fertilization. They are considered to be pluripotent, able to give rise to many different cell lineages. For cardiac regeneration therapy, there is a growing body of knowledge from animal models regarding the steps of isolation, differentiation, and clinical application. Human ES cells differentiate into spontaneously beating cells with a cardiomyocyte phenotype. The morphology and ultrastructure of these cells are organized with sarcomeric

3 Stem Cell Therapy 341 TABLE 2. Clinical Trials of Stem Cell Therapy for Cardiac Repair Registered With ClinicalTrials.gov Cell Type Used Trial Design, Phase Condition Treated Randomized structures, formation of intercalated disks, desmosomes, and gap junctions, characteristic of cardiomyocytes, 20,21 and they demonstrate the presence of a functional syncitium with action potential propagation. 21,22 When transplanted into infarcted myocardium, ES cell derived cardiomyocytes engraft and improve cardiac function in several rodent models In the failing heart, in addition to replenishing cardiomyocytes by ES derived cells, a simultaneous increase in the blood supply may be necessary for optimal and prolonged engraftment. Hence, it is of interest that ES cells differentiate to all cell lines necessary for formation of new Total Patients to Be Enrolled, n Primary Outcome Sponsor Region Intramyocardial autologous I/II Myocardial ischemia 40 Perfusion Rigshospitalet Copenhagen, Denmark MSCs Autologous BMMNCs I CHF undergoing CABG 75 LV function University of Pittsburgh Pittsburgh, Pa Autologous BMMNCs I CHF undergoing VAD implantation 10 LV function University of Pittsburgh Pittsburgh, Pa Intramyocardial BMMNCs I Ischemic cardiomyopathy 30 Safety Texas Heart Institute Texas Intracoronary BMMNCs I Acute MI 60 Safety Minneapolis Heart Minneapolis, Minn Institute Foundation Intravenous MSCs I Acute MI 48 Safety Osiris Therapeutics Multicenter, United States Autologous Intramyocardial CD34 I Chronic myocardial ischemia 24 Not stated St Elizabeth s Medical Center Boston, Mass Autologous CD34 I/II Chronic myocardial ischemia Intramyocardial autologous BMMNCs Intracoronary autologous BMMNCs Intramyocardial autologous skeletal myoblasts Intramyocardial autologous skeletal myoblasts Intramyocardial autologous skeletal myoblasts 10 Perfusion Translational Research Informatics Center, Japan Kobe, Japan II Ischemic cardiomyopathy 35 LV function Odense University Hospital Odense, Denmark II Acute MI 100 Myocardial viability Nantes University Nantes, France Hospital I Ischemic cardiomyopathy 15 Safety Bioheart Multicenter, United States II Ischemic cardiomyopathy Not stated LV function Genzyme Multicenter, Europe/United Kingdom I Ischemic cardiomyopathy 15 Safety Bioheart Multicenter, United States undergoing CABG CABG indicates coronary artery bypass grafting; VAD, ventricular assist device. blood vessels. Both murine and human ES cells spontaneously differentiate to form endothelial and smooth muscle cells in vitro 27 and in vivo. 28,29 To date, no human clinical studies have been initiated because of both the ethical issues surrounding access to embryos and the possibility of teratoma formation, suggested by a study injecting ES cells in skeletal muscle. 30 Resident Cardiac Stem Cells In recent years, compelling evidence has accumulated suggesting that the heart has endogenous regenerative potential. Figure 1. Possible mechanisms for successful cardiac regenerative therapy. See text for details.

4 342 Circulation July 25, 2006 Recent studies have isolated, from both human and murine hearts, undifferentiated cells that are clonogenic, express stem and endothelial progenitor cell (EPC) antigens/markers, and appear to have the properties of adult cardiac stem cells. 31,32 These cells most likely mediate endogenous mechanisms for minor repair and for replacement of ongoing cell turnover within the adult heart. More importantly, they may represent a therapeutic target that, if enhanced, could induce cardiac self-repair. These cells have been phenotyped using different antigenic approaches. In a seminal report published in 2003, Beltrami and colleages 31 separated c-kit positive cells from the rat heart and demonstrated their clonogenicity and multipotency in vitro, as well as their capacity to participate in cardiomyocyte and blood vessel regeneration after MI. Subsequently, Oh and coworkers 32 separated resident murine cardiac stem cells on the basis of the presence of stem cell antigen-1. They also demonstrated in vitro and in vivo myocardial differentiation of these cells and provided evidence supporting the fusion of these cells. In 2004, Messina and coworkers 33 reported on the identification of cardiospheres, clusters of self-adherent cells that grew from cultured adult cardiac tissue derived from both human and murine hearts. These cells were shown to be clonogenic and capable of transdifferentiation in vitro, and they induced both myocardial and vascular regeneration after MI. Laugwitz and colleagues 34 isolated a population of cardiac precursor cells from postnatal mouse hearts using isl-1 transcription factor as a cell marker. These cells are c-kit and stem cell antigen-1 negative but are capable of differentiation into cardiomyocytes with electrical and contractile properties. Finally, Martin et al 35 identified a side population of cells (SP cells) in the developing heart and adult heart that are capable of proliferating and differentiating into cardiac and hematopoietic lineages in vitro. These cells were identified on the basis of expressing Abcg2, an ATP-binding cassette transporter, rather than by detection of surface markers. Cardiac stem cells can be harvested from patients and expanded ex vivo to generate large numbers of cells. A recent report by Urbanek and colleagues 36 demonstrated that cardiac stem cells increase in number immediately after MI, but in the chronic phase, the numbers fall, and the remaining cardiac stem cells have less regenerative potential. This suggests that the left ventricular (LV) dysfunction in ischemic cardiomyopathy may be due to a defect in or deficiency of functionally competent cardiac stem cells. In addition, Mouquet et al 37 have recently shown in an experimental study that bone marrow may represent a reservoir for cardiac stem cells and suggested that depletion of this reservoir could contribute to diminished reparative capacity. To date, there are no clinical trials of human cardiac stem cells. However, Smith et al 38 demonstrated that cardiospheres could be grown from human endomyocardial biopsy specimens. These cardiospheres represent an easily accessible option for autologous stem cell therapy, making the possibility of clinical testing of this approach feasible. The Specialized Centers for Cell-Based Therapy initiative of the NHLBI has funded clinical trials of cardiac stem cells that should begin in the near future. Skeletal Myoblasts Autologous skeletal myoblasts are another potential source for cardiac repair because of their biological properties and lack of ethical and immunological problems. Skeletal myoblasts or satellite cells are the reservoir of regenerative cells for skeletal muscle tissue; they have the ability for selfrenewal and differentiation if muscle injury occurs. 39 They have many desirable features as donor cells, including the ability to be amplified in an undifferentiated state in vitro and high resistance to tissue ischemia. They also have been shown to continue proliferation in vivo to a certain extent, which gives rise to a larger graft size. 3 Satellite cells are committed solely to the myogenic lineage. Therefore, regardless of environmental influences, even if implanted into a scar made up mainly of fibroblasts, myoblasts differentiate into functional muscle cells. A growing body of experimental data and initial clinical studies has shown not only engraftment of donor cells but also improvement in global cardiac pump function. 8,11,40 42 However, the exact mechanism by which they improve LV function is still debated. There may be beneficial effects of contracting noncardiac myocytes, some paracrine actions and an effect on scar strengthening to prevent LV dilatation and remodeling. 43 The critical importance of progressing to clinical trials is underscored by the early clinical experience with skeletal myoblasts. Myoblast transfer in early studies 44,45 was associated with sustained ventricular tachycardia, a life-threatening arrhythmia. This finding guided changes in future protocols involving skeletal myoblasts, which now require prophylactic cardioverterdefibrillator implantation and/or amiodarone therapy to prevent ventricular tachycardia. This protocol change resulted in less frequent clinically evident arrhythmias and is a powerful demonstration of how clinical trials are integral in guiding and refining the way in which stem cell therapy should be administered in patients. Phase II studies of skeletal myoblast therapy are presently underway. Human Adult Bone Marrow Derived Stem Cells The observation that bone marrow elements contribute to cardiac repair in the infarcted heart served as the rationale for adult bone marrow cell therapy after MI. Jackson and coworkers 46 transplanted wild-type mice with green fluorescent protein (GFP) positive bone marrow and then induced MI through coronary artery ligation and reperfusion. They demonstrated that bone marrow elements contributed to cardiomyocyte and endothelial cell formation after MI by finding GFP-positive cardiomyocytes and endothelial cells. It appeared that there is an intrinsic repair mechanism for minor cardiac damage within the bone marrow but that it is not adequate to repair larger amounts of damage such as that after MI. Substantial effort has been expended to try to enhance

5 Stem Cell Therapy 343 this endogenous repair mechanism and use bone marrow as a potential source of stem cells for cardiac repair. Orlic and coworkers 4 have shown that lineage-negative, c-kit positive bone marrow derived cells differentiate into new cardiomyocytes after MI. This regenerative therapy can be harnessed by either direct injection into the peri-infarct rim of functioning myocardium or by using chemoattractant cytokines to mobilize the cells from bone marrow. In 1 experiment, bone marrow was harvested from male mice, labeled with GFP, and injected into the peri-infarct rim of female mice. This resulted in a substantial increase in myocytes in the infarct zone, and the myocytes were of donor origin (on the basis of Y chromosome staining and GFP expression). There was a corresponding improvement in hemodynamic parameters after only 9 days. The same group showed that mobilization of lineagenegative, c-kit cells with granulocyte colony-stimulating factor and stem cell factor before and after MI in mice resulted in growth of new cardiomyocytes in the infarct zone and improved ventricular function and led to substantial improvement in survival of the treated group. Endothelial and smooth muscle cells also were proliferating, but new myocyte growth predominated. 47 However, in contrast to these findings, Murry and colleagues 48 and Balsam and coworkers 49 reported that lineage-negative, c-kit cells did not differentiate into cardiomyocytes. In the latter study, bone marrow cells transplanted into ischemic myocardium adopted hematopoietic fates rather than transdifferentiating into myocardium yet interestingly still prevented LV dilatation and dysfunction associated with postinfarction remodeling. Despite the conflicting evidence regarding the ability of bone marrow derived cells to transdifferentiate, their efficacy in preventing remodeling has been demonstrated in many laboratories; therefore, clinical efficacy trials have progressed in parallel with ongoing mechanistic laboratory trials to determine the precise molecular mechanism by which these cells exert their beneficial effects. Kamihata et al 7 isolated mononuclear cells from swine bone marrow and injected them into the infarct zone and peri-infarct region of pigs after MI induced by left anterior descending coronary artery ligation. This was associated with an improvement in myocardial perfusion by contrast echocardiography, increased numbers of capillaries, increased coronary collateral circulation on angiography, improved ejection fraction, and reduction in infarct size compared with controls. Of note, the endothelial lining of newly formed capillaries was derived from the donor bone marrow cells, but the fibroblasts in the fibrotic area of the infarct were not. This suggests that the microenvironment of MI is proangiogenic rather than fibrosisinducing for this population of cells. This evidence that precursors of both cardiomyocytes and endothelial cells exist within the mononuclear cell fraction of adult bone marrow forms the basis for the use of bone marrow mononuclear cells (BMMNCs) in clinical trials (Table 1). In just the past 3 years, BMMNC transplantation has become the most widely studied cell-based therapy for human applications (Table 1). Several studies using autologous bone marrow warrant special mention. The initial studies used intracoronary delivery of BMMNCs in the post-mi setting because animal studies demonstrate that tissue damage in MI resulted in bone marrow stem cell homing to the infarcted myocardium. 6 In the first published human trial, Strauer et al 50 aspirated BMMNCs and reinfused them into the infarctrelated artery 7 days after MI in 10 patients and had a control group comprising 10 patients who refused the treatment. This method resulted in significantly improved myocardial perfusion and wall motion indexes. The Transplantation of Progenitor Cells and Regeneration Enhancement in Acute Myocardial Infarction (TOPCARE-AMI) investigators 51,52 randomized 59 patients after acute MI to receive intracoronary infusion of BMMNCs or ex vivo expanded circulating progenitor cells. They delivered the cells into the infarctrelated artery 4 days after MI and showed improvement in LV ejection fraction from 51% to 58% (P 0.001), as well as significantly enhanced myocardial viability and regional wall motion in the infarct area. Interestingly, they were unable to show a difference between the 2 active cell treatment groups. In the BOne marrow transfer to enhance ST-elevation infarct regeneration (BOOST) trial, Wollert and coworkers 53 randomized 60 patients after successful percutaneous coronary intervention for acute MI to receive either intracoronary BMMNCs or standard therapy. They demonstrated an improvement in LV ejection fraction of 6.7% in the treatment group and 0.7% in the control group at 6 months (P ). Taken together, these studies with control groups suggest that BMMNCs are safe and may improve cardiac function by a substantial and clinically meaningful degree following MI. However, more recently, Janssens and colleagues presented their findings that intracoronary transfer of BMMNCs failed to achieve their primary end point, improvement in global LV function. 54 They demonstrated a significant reduction in scar size and an improvement in regional function, but there was no improvement in LV ejection fraction (P 0.36). Their patient population differed from the BOOST trial in that they were reperfused earlier and may therefore have gained only a small benefit from cell therapy because they derived maximal benefit from earlier reperfusion. In the 18-month follow-up to the BOOST study, the improvement in LV ejection fraction in the cell therapy group was sustained. 55 However, the control group also had improved by this time, and the difference between the 2 groups was no longer significantly different. This catch-up phenomenon in the control group suggests that, rather than the effect of cell therapy being transient, cell therapy may in fact accelerate the postinfarction LV functional recovery that is achievable with standard medical therapy. In contrast to the acute MI setting, patients with chronic ischemic cardiomyopathy are unlikely to release signals from damaged myocardium to induce stem cell homing. Therefore, an alternative approach to intracoronary infusion of cells in

6 344 Circulation July 25, 2006 TABLE 3. Clinical Studies Using MSCs Study Treated, n Autologous/ Allogeneic Indication Randomized Chen et al Autologous MI Bang et al Autologous Stroke Garcia-Olmo et al Autologous Crohn s disease fistula Horwitz et al Allogeneic Osteogenesis imperfecta LeBlanc et al Allogeneic GVHD GVHD indicates graft-versus-host disease. this setting is endomyocardial injection of cells to deliver them to the exact location where their effect is required. Perin et al 56 enrolled 14 subjects and 7 control subjects with ischemic cardiomyopathy. The treatment group received endomyocardial injection of 30 million BMMNCs. They showed a significant reduction in reversible myocardial perfusion defects and a significant improvement in overall LV function. Notably, they enrolled subjects with significant LV dysfunction at baseline; therefore, their results may be more clinically relevant to the CHF patient group than previous studies that enrolled patients with normal or mildly impaired LV function after MI. Currently, a number of phase II studies are ongoing with BMMNCs (Table 2). Mesenchymal Stem Cells Mesenchymal stem cells (MSCs) are found in bone marrow, muscle, skin, and adipose tissue and are characterized by the potential to differentiate into any tissue of mesenchymal origin, including muscle, fibroblasts, bone, tendon, ligament, and adipose tissue. 57 MSCs from adult bone marrow can be separated by density gradient centrifugation and adhering-cell culture in defined serum-containing medium. 58 The cells isolated after adherence in culture are negative for CD34 and CD45, unlike hematopoietic progenitors from bone marrow, and characteristically express CD29, CD44, CD71, CD90, CD105, CD106, CD120a, CD124, SH2, SH3, and SH Some studies demonstrate that MSCs transdifferentiate into cardiomyocytes and vascularlike structures MSCs differentiate into cardiomyocytes and endothelial cells in vivo when transplanted to the heart in both noninjury and MI models. These cells have been strictly characterized by immunohistochemistry and stain positively for cardiac and endothelial specific markers, as well as gap junction proteins. 64,65,67,68 Myocardial function and capillary formation are significantly increased in experimental groups treated with MSCs compared with controls. 69,70 These results suggest that MSCs act by regenerating functionally effective, integrated cardiomyocytes and possibly new blood vessels. MSCs also have been injected into infarcted myocardium via a catheter-based approach in pigs, resulting in regeneration of myocardium, reduced infarct size, and improved regional and global cardiac contractile function. 5 Importantly, the latter study used allogeneic MSCs, which did not produce evidence of rejection. Whereas autologous cell-based therapy poses no risk of rejection, an off the shelf allogeneic cell product would be much more cost effective and much easier to administer and could potentially allow delivery of greater numbers of cells than autologous cell therapy. MSCs appear to avoid the problem of rejection by being hypoimmunogenic. These cells lack MHC-II and B-7 costimulatory molecule expression, thereby preventing T-cell responses, and induce a suppressive local microenvironment through the production of prostaglandins and other soluble mediators. 58,71 As such, MSCs may allow allogeneic cell therapy while avoiding rejection. MSCs also have been studied clinically. Chen and colleagues 72 randomized 69 patients after MI to receive intracoronary autologous MSCs or placebo. They demonstrated a significant improvement in global and regional LV function and a significant reduction in the size of the perfusion defect, suggesting that MSC therapy can regenerate infarcted myocardium or protect against LV remodeling. Currently, several studies have been published using MSCs for noncardiac disorders (see Table 3), and allogeneic MSCs are in clinical trials for myocardial regeneration in the United States under the sponsorship of Osiris Therapeutics (Table 2). In addition, the Specialized Centers for Cell-Based Therapy program also plans to conduct clinical trials of MSCs for patients with CHF. Endothelial Progenitor Cells Cells with phenotypic and functional characteristics similar to the fetal angioblast also are present in adult human bone marrow. 6 These cells, known as EPCs, express some, but not all, cell surface markers characteristic of mature endothelium, certain surface markers of hematopoietic cells, and transcription factors that identify them as precursor cells. 27,73 75 In addition to endothelial cell surface markers, EPCs also express markers of immature cells, including AC133, a novel hematopoietic stem cell marker 76 not expressed on mature endothelial cells. 77 After MI, intravenously injected EPCs homed to the infarct region within 48 hours. 6 At 14 days, there was a marked increase in the number of capillaries in the infarct zone and the peri-infarct rim resulting from the induction of both vasculogenesis and angiogenesis, but there

7 Stem Cell Therapy 345 was no change in the noninfarcted regions of the heart. There was a significant reduction in collagen deposition and apoptosis of cardiomyocytes and an improvement in cardiac function on echocardiography. 6 It appears that neovascularization induced by these cells leads to the prevention of apoptosis and LV remodeling and may lead to some degree of cardiomyocyte regeneration. 15 Erb and colleagues 78 randomized patients with recanalized, chronically occluded coronary arteries to receive intracoronary progenitor cells or placebo. They mobilized bone marrow cells using granulocyte colony-stimulating factor, harvested them from peripheral blood, expanded them ex vivo, and reinfused them via the coronary artery. This treatment resulted in significant improvements in coronary flow reserve and cardiac function and a significant reduction in infarct size. Currently, clinical trials of EPC therapy for angiogenesis and myocardial regeneration are in progress that use CD34 cells from bone marrow that are enriched for EPCs. These cells can be immunoselected from the mononuclear fraction of bone marrow (Table 2). Umbilical Cord Blood Stem Cells Umbilical cord blood (UCB) contains both hematopoietic stem cells and mesenchymal precursor cells. 79 Because stem cells in UCB exist in higher numbers than in adult human blood or bone marrow, 80 several populations of cells derived from UCB are possible sources of stem cells for cardiac repair. Kogler and colleagues 81 have described a population of cells from human UCB called unrestricted somatic stem cells. These cells, which are fibroblastlike in appearance, adhere to culture dishes; are negative for c-kit, CD34, and CD45; and are capable of differentiating, both in vitro and in vivo, into a variety of tissues, including cardiomyocytes. Human unrestricted somatic stem cells, 82 when delivered by direct injection at thoracotomy in immunosuppressed pigs after MI, improved perfusion and wall motion, reduced infarct scar size, and enhanced global cardiac function. Ma et al 83 injected human mononuclear UCB cells, a small fraction ( 1%) of which were CD34, intravenously 1 day after MI in NOD/scid mice. The cells homed to the infarcted hearts, reduced infarct size, and enhanced neovascularization with capillary endothelial cells of both human and mouse origin. Interestingly, they found no evidence of myocytes of human origin, arguing against cardiomyogenic differentiation. In a rat model of MI, 84 UCB CD34 improved cardiac function when injected into the peri-infarct rim immediately after MI compared with control animals that received injection of medium. At present, no clinical studies of UCB have been reported. Guiding Principle for Adopting New Therapies As outlined above, a number of promising cell types for cardiac regenerative therapy have accumulating preclinical and early clinical data supporting their potential. Nevertheless, controversy abounds as to whether ongoing clinical trials of these novel strategies are warranted. We believe that fundamental criteria supporting ongoing trials have been met. The World Medical Association in the Declaration of Helsinki has set down principles for clinician researchers to follow. 85 These state: In the treatment of a patient, where proven prophylactic, diagnostic and therapeutic methods do not exist or have been ineffective, the physician, with informed consent from the patient, must be free to use unproven or new prophylactic, diagnostic and therapeutic measures, if in the physician s judgment it offers hope of saving life, re-establishing health or alleviating suffering. In the case of CHF, it is clear that there are unmet needs for new treatments to save lives and alleviate suffering. Accordingly, this principle supports clinical trials of new cell-based therapies. In assessing the possibility of adopting a new treatment into clinical practice, rigorous scientific and ethical standards must be followed. Stem cell therapy has presented us with a number of unique challenges. Not only is the optimal cell type not clear, but how to best deliver these cells also is not immediately apparent, and these questions can be addressed only by prospectively designed clinical trials. Our understanding of the exact mechanisms by which stem cells exert their beneficial effects on cardiac function has evolved substantially, initially assuming transdifferentiation as the only mode of action and subsequently expanding to include, among other possibilities, effects on strengthening the MI scar 19 and paracrine effects. 16 Importantly, we propose that all of these features may be important for an integrated and orchestrated mechanism of repair that involves restoring endogenous stem cell niches. Despite incomplete understanding of mechanism, many independent laboratories have consistently demonstrated the beneficial effects on cardiac function in preclinical and clinical studies. These beneficial findings cannot be ignored and ongoing clinical studies delayed on the basis of needing better understanding of the molecular underpinnings of the observed effects because these functional improvements may translate to clinical benefit for patients. These ethical and logistic issues, not encountered with most previous drug or device trials, may appear daunting. However, despite the difficulties of stem cell research, the basic tenets of clinical research still apply: Patient safety is paramount. The risk of depriving patients of a potential new therapy must be balanced against the inherent risk of testing the new therapy. It is with these guiding principles in mind that we must decide whether stem cell therapy is ready for clinical trials. Patient Safety Is Paramount The main issue in going forward into clinical trials is patient safety. Primum non nocere (first do no harm) is,

8 346 Circulation July 25, 2006 as always, our overriding concern. But how much safety data are enough to justify advancing to the next stage of clinical investigation? Autologous stem cell therapy begins with the premise that cellular repair occurs naturally in the human body and that enhancing that process should be safe. In fact, numerous animal studies have demonstrated that this is the case, with no adverse effects of cellular therapy for cardiac repair being observed; this has served as the platform for launching into human safety studies. Several postulated safety issues about stem cell therapy were raised. There was concern regarding the intracoronary delivery route, fearing that cells administered this way may result in blockage of the coronary arteries and further damage to the myocardium. One study using autologous MSCs from dog bone marrow injected into normal coronary arteries demonstrated microinfarction in the region supplied by that artery, suggesting that these cells aggregate in vivo, leading to arterial or arteriolar obstruction. 86 Nevertheless, intracoronary MSC therapy in 1 human trial resulted in improved cardiac function and was safe. 72 Differences in cell numbers or handling or delivery techniques may account for these differing clinical outcomes. A number of studies have now demonstrated safety of the intracoronary route using BMMNCs obtained by direct bone marrow aspiration 50,52,53 and MSCs. 72 Another theoretical safety concern was dysregulated angiogenesis or angioma formation within the myocardium seen with some angiogenic gene therapies. 87 Could this occur with stem cell therapy designed to enhance neovascularization? Or could myogenic therapy result in the formation of myoma or myosarcoma? Tumor formation has not been reported in animal or human studies with stem cell therapies to date when passaged for standard periods such as 6 to 8 weeks. However, there has been 1 report of late-passaged MSCs (16 to 20 weeks) developing chromosomal abnormalities and spontaneous cancerous transformation in vitro. 88 It is therefore essential that human trials use rigorous safety analysis of transplanted cells to detect any possible malignant change or chromosomal abnormalities. Additionally, the problems encountered with gene therapy may relate to factors not associated with stem cells. These include the need for viral or other vectors to deliver the genes, problems with short biological half-lives of the gene products, and problems delivering the precise dose required. Although there may be similarities between gene and stem cell therapies as novel biological therapeutics, the practicalities and logistic difficulties are different (and substantially less) with stem cell therapy. Regardless of the type of stem cell used in clinical trials, it is critically important to be vigilant against ex vivo contamination of the specimen. Infected or contaminated cells injected into the heart could potentially lead to devastating consequences of endocarditis and myocarditis. As such, all stem cell trial protocols should use good manufacturing practice facilities to minimize the risk of such outcomes. Rigorous follow-up also is essential after stem cell therapy to monitor for complications. Arrhythmias must be actively looked for both by continuous ECG monitoring around the time of the procedure and by ambulatory monitoring in follow-up. Additionally, patients enrolled in stem cell trials using allogeneic cells must be closely followed up for signs of allograft rejection. A number of cells have been tested so far in preliminary trials (Table 1). More than 400 patients have been tested, and the totality of evidence from these studies supports safety of each cell therapy and the general approach. This evidence strongly supports ongoing efforts to advance to large, randomized, placebo-controlled, double-blind studies with clinical end points. Balance of Risk The risk of exposing patients to possible adverse outcomes of a new treatment must be weighed against the risk of depriving all patients of a new and possibly effective treatment to alleviate suffering or to prolong life. There is now sufficient preclinical and clinical data to warrant larger randomized controlled trials evaluating stem cell therapy. The argument that these trials should be delayed until mechanisms are further understood will unnecessarily deprive large numbers of patients of therapeutic approaches that may improve their clinical outcome. Only clinical trials can lead to optimization of stem cell therapy with recognition of the best type of cell and the best delivery method for our patients. Another compelling argument for initiating clinical research is that the results of these investigations often provide pivotal insights that allow a new field to advance. Several examples in cardiovascular medicine illustrate this concept. ACE inhibitors 89 and 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins), 90 widely used drugs with solid mechanistic underpinning supporting their use, were both appreciated after entry into clinical practice to have pharmacological effects that extended beyond their initially intended design. The additional mechanistic effects were suggested by results obtained in clinical trails, demonstrating the important synergy between in vitro/ animal studies and clinical trials conducted in parallel in guiding clinical therapies. Another important example of how mechanistic, novel, pathophysiological, and therapeutic insights can progress in parallel during clinical development is percutaneous transluminal coronary angioplasty (PTCA). In 1977, pioneer Andreas Gruntzig published results obtained in 8 dogs, then progressed to human postmortem experiments 91 and next to human clinical trials, publishing his first series of 5 patients only 1 year later. 92 The first study of 50 patients followed in This translational research program introduced PTCA as a new clinical treatmen, but was not free of serious complications. Soon the problem of restenosis, the Achilles heel of PTCA, was appreciated,

9 Stem Cell Therapy 347 prompting substantial new basic research and the development of the coronary artery stent, which reduced but did not eliminate the incidence of restenosis. As is now well known, drug-coated stents that nearly eliminate restensosis have been developed. 94 Before the publication of this seminal article, safety and feasibility data had been published on 100 patients, substantially less than the number who have been enrolled in preliminary stem cell trials. This stepwise progression to clinical trials was appropriate and timely, with patients receiving the benefits of the proven therapies, while clinical trial results guided further basic and preclinical research to eventually refine and improve these clinical therapies. We anticipate a similar iterative pathway for the development of stem cell therapy (see Figure 2). Importantly, clinical trials do not always produce positive outcomes, yet unexpectedly negative results are often pivotal to the understanding of disease pathophysiology. For example, in the Cardiac Arrhythmia Suppression Trial (CAST), administration of antiarrhythmic drugs for asymptomatic nonsustained ventricular tachycardia after MI resulted in an excess of deaths in the treatment arms. 98 No animal study had suggested that this would happen. In fact, further laboratory investigations and animal studies would have pursued a wrong path and would have wasted valuable resources. Although the outcome was unfortunate and unexpected, a properly designed prospective clinical trial discovered this deadly complication of antiarrhythmic therapy and changed future therapeutic approaches. The appropriate progression into prospective, randomized, controlled clinical trials will prevent exposing more patients to potentially dangerous therapies in uncontrolled trials or in the proliferation of delivering the therapy out of the clinical trial context. 99 Additionally, the transition from Figure 2. Clinical development of novel therapies. Pivotal role of clinical trials in developing stem cell therapeutics. Current phase I and II clinical trials of cell-based cardiac regenerative therapy are based on basic science observations and preclinical observations. As indicated, the basis for proceeding with clinical trials includes adequate safety data and a rational scientific basis. Trials themselves offer novel mechanistic insights and may be negative even if a mechanistic basis is well understood or, as frequently occurs, may be positive for reasons other than that posited by the initial hypothesis. Even when a therapy proves to be of clinical value, unanticipated adverse side effects may be detected, eliminating the therapy from the therapeutic armamentarium. In either case, results of clinical trials offer critical insights and guide new scientific hypotheses. Clinical trials are rationally and ethically conducted experiments that are a necessary step in the development of any new therapeutic principal. The majority of clinical trials are negative. preliminary studies to randomized controlled trials will demonstrate efficacy and safety of therapies that are indeed safe and effective, thereby allowing them to become incorporated into routine clinical practice. The time to take that step is now. Clinical Trial Design In the United States, the Food and Drug Administration (FDA) Modernization Act, section 113 (FDAMA 113), mandates the registration of all clinical trials that test effectiveness for serious or life-threatening conditions submitted to the FDA under investigational-new-drug applications. 100 A Web-based registry, clinicaltrials.gov, was established in 2000 by the National Library of Medicine on behalf of the National Institutes of Health as a result of this law. This database permits registration of all clinical trials, including phase I studies that are not mandated to be registered; the number of registered trials has been increasing. 101 The International Committee of Medical Journal Editors now demands registration of clinical trials as a prerequisite for publication. 102 Currently, a number of clinical trials of stem cell therapy for cardiac repair are registered with clinicaltrials.gov; they are listed in Table 2. All of these trials are phase I or II trials rather than large phase III trials, and a number continue to be nonrandomized. We have not included trials of cytokines/growth factors alone. Although current data do not support routine clinical use of stem cell therapy for cardiac repair, the results of preliminary studies are promising. Many treatments that improve cardiac function after MI in animal models have failed to translate into clinical benefit. A number of reasons may account for this disparity between preclinical and clinical results, but the most important difference

10 348 Circulation July 25, 2006 between the 2 situations is the complex biology and the multitude of factors in clinical situations that can never fully be replicated in experimental animal models. At some point in the discovery of any new treatment modality, only translation into clinical trials will provide the necessary data to move forward. There is also a need for ongoing basic science research in parallel to advance stem cell therapy toward clinical application. Clinical studies of stem cell therapy should use rigorous trial designs and appropriate built-in safeguard mechanisms (Data Safety and Monitoring boards). In this regard, they should be prospective, double-blind studies with randomization on a background of best conventional therapy. Clinical trials should be designed to go beyond surrogate end points and should have adequate power to detect differences in relevant clinical end points such as survival, hospitalization, or reinfarction. Furthermore, future studies should be designed not only to assess safety and efficacy but also to gain further insight into mechanism of action of stem cells. One method of achieving this is to take advantage of state-of-the-art imaging modalities to assess myocardial function and to assess myocardial regeneration or neovascularization. 126 Many issues in the burgeoning field of regenerative medicine still need to be addressed, and clinical trials will participate in a fundamental way. First, it is necessary to determine the optimal cell number required to gain maximal clinical effect; this will, in turn, guide how to access the cells (eg, bone marrow aspirate versus cytokine mobilization) and whether they need to be expanded ex vivo. Second, the optimal cell type for different patient populations remains unknown. Finally, effective delivery mechanisms must be determined and will likely be cell and/or disease specific. From these types of studies, not only will it be possible to determine whether cell therapy for cardiac repair is effective and safe in humans, but it is also probable that a great deal of mechanistic information will be derived that will guide further basic science and protocol design for subsequent clinical trials. Conclusions Although at this time no data support either cell-based or specific stem cell therapies as standard clinical practice for cardiac applications, there is a wealth of preclinical and early clinical data showing safety, feasibility, and early efficacy of adult cell-based therapy. Adult stem cell therapies should therefore proceed into randomized, placebo-controlled, double-blind clinical trials. The ongoing rigorously designed trials will contribute greatly to this emerging and exciting new therapeutic approach for diseases of the cardiovascular system. Funding This work was supported by The Johns Hopkins Specialized Center for Cell-Based Therapy (U54 HL081028), The Johns Hopkins University School of Medicine Institute for Cell Engineering (ICE), the John Hopkins University, and University of Texas, Southwestern Donald W. Reynolds Centers, and NIH grants HL-65455, HL-72185, and AG Disclosures Dr Hare received a research grant from Osiris Therapeutics. The other authors report no conflicts. References 1. American Heart Association. Heart and Stroke Statistics: 2005 Update. Available at: HDSStats2005Update.pdf. Accessed December 3, Marelli D, Desrosiers C, el-alfy M, Kao RL, Chiu RC. Cell transplantation for myocardial repair: an experimental approach. Cell Transplant. 1992;1: Koh GY, Klug MG, Soonpaa MH, Field LJ. Differentiation and long-term survival of C2C12 myoblast graft in heart. J Clin Invest. 1993;92: Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Picke lj, Mackay R, Nadal-Ginard B, Bodine DM, Leri A, Anversa P. Bone marrow cells regenerate infarcted myocardium. Nature. 2001;410: Amado LC, Saliaris AP, Schuleri KH, St. John M, Xie J-S, Cattaneo S, Durand DJ, Fitton T, Kuang JQ, Stewart G, Lehrke S, Baumgartner WW, Martin BJ, Heldman AW, Hare JM. Cardiac repair with intramyocardial injection of allogeneic mesenchymal stem cells after myocardial infarction. Proc Natl Acad Sci U S A. 2005;102: Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D, Wang J, Homma S, Edwards NM, Itescu S. Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodelling and improves cardiac function. Nat Med. 2001;7: Kamihata H, Matsubara H, Nishiue T, Fujiyama S, Tsutsumi Y, Ozono R, Masaki H, Mori Y, Iba O, Tateishi E, Kosaki A, Shintani S, Murohara T, Imaizumi T, Iwasaka T. Implantation of bone marrow mononuclear cells into ischemic myocardium enhances collateral perfusion and regional function via side supply of angioblasts, angiogenic ligands, and cytokines. Circulation. 2001;104: Ghostine S, Carrion C, Souza LCG, Richard P, Bruneval P, Vilquin J-T, Pouzet B, Schwartz K, Menasche P, Hagege AA. Long-term efficacy of myoblast transplantation on regional structure and function after myocardial infarction. Circulation. 2002;106(suppl I):I-131 I Chien KR. Stem cells: lost in translation. Nature. 2004;428: No consensus on stem cells. Nature. 2004;428: Menasche P, Hagege AA, Scorsin M, Pouzet B, Desnos M, Duboc D, Schwartz K, Vilquin J-T, Marolleau J-P. Myoblast transplantation for heart failure. Lancet. 2001;357: Condorelli G, Borello U, De Angelis L, Latronico M, Sirabella D, Coletta M, Galli R, Balconi G, Follenzi A, Frati G, Cusella De Angelis MG, Gioglio L, Amuchastegui S, Adorini L, Naldini L, Vescovi A, Dejana E, Cossu G. Cardiomyocytes induce endothelial cells to transdifferentiate into cardiac muscle: implications for myocardium regeneration. Proc Natl Acad Sci U S A. 2001;98: Kudo M, Wang Y, Wani MA, Xu M, Ayub A, Ashraf M. Implantation of bone marrow stem cells reduces the infarction and fibrosis in ischemic mouse heart. J Mol Cell Cardiol. 2003;35: Olivares EL, Ribeiro VP, Werneck de Castro JPS, Ribeiro KC, Mattos EC, Goldenberg RCS, Mill JG, Dohmann HF, dos Santos RR, de Carvalho ACC, Masuda MO. Bone marrow stromal cells improve cardiac performance in healed infarcted rat hearts. Am J Physiol Heart Circ Physiol. 2004;287:H464 H Schuster MD, Kocher AA, Seki T, Martens TP, Xiang G, Homma S, Itescu S. Myocardial neovascularization by bone marrow angioblasts results in cardiomyocyte regeneration. Am J Physiol Heart Circ Physiol. 2004;287:H525 H Gnecchi M, He H, Liang OD, Melo LG, Morello F, Mu H, Noiseux N, Zhang L, Pratt RE, Ingwall JS, Dzau VJ. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat Med. 2005;11:

11 Stem Cell Therapy Moore KA, Lemischka IR. Stem cells and their niches. Science. 2006; 311: Nygren JM, Jovinge S, Breitbach M, Sawen P, Roll W, Hescheler J, Taneera J, Fleischmann BK, Jacobsen SEW. Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nat Med. 2004;10: Fujii T, Yau TM, Weisel RD, Ohno N, Mickle DAG, Shiono N, Ozawa T, Matsubayashi K, Li R-K. Cell transplantation to prevent heart failure: a comparison of cell types. Ann Thorac Surg. 2003;76: Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M, Gepstein A, Livne E, Binah O, Itskovitz-Eldor J, Gepstein L. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J Clin Invest. 2001;108: Westfall MV, Pasyk KA, Yule DI, Samuelson LC, Metzger JM. Ultrastructure and cell-cell coupling of cardiac myocytes differentiating in embryonic stem cell cultures. Cell Motil Cytoskel. 1998;36: Kehat I, Gepstein A, Spira A, Itskovitz-Eldor J, Gepstein L. Highresolution electrophysiological assessment of human embryonic stem cell derived cardiomyocytes: a novel in vitro model for the study of conduction. Circ Res. 2002;91: Yang Y, Min J-Y, Rana JS, Ke Q, Cai J, Chen Y, Morgan JP, Xiao Y-F. VEGF enhances functional improvement of postinfarcted hearts by transplantation of ESC-differentiated cells. J Appl Physiol. 2002;93: Behfar A, Zingman LV, Hodgson DM, Rauzier J-M, Kane GC, Terzic A, Puceat M. Stem cell differentiation requires a paracrine pathway in the heart. FASEB J. 2002;16: Klug MG, Soonpaa MH, Koh GY, Field LJ. Genetically selected cardiomyocytes from differentiating embryonic stem cells form stable intracardiac grafts. J Clin Invest. 1996;98: Min J-Y, Yang Y, Converso KL, Liu L, Huang Q, Morgan JP, Xiao Y-F. Transplantation of embryonic stem cells improves cardiac function in postinfarcted rats. J Appl Physiol. 2002;92: Vittet D, Prandini MH, Berthier R, Schweitzer A, Martin-Sisteron H, Uzan G, Dejana E. Embryonic stem cells differentiate in vitro to endothelial cells through successive maturation steps. Blood. 1996;88: Yamashita J, Itoh H, Hirashima M, Ogawa M, Nishikawa S, Yurugi T, Naito M, Nakao K, Nishikawa S-I. Flk1-positive cells derived from embryonic stem cells serve as vascular progenitors. Nature. 2000;408: Marchetti S, Gimond C, Iljin K, Bourcier C, Alitalo K, Pouyssegur J, Pages G. Endothelial cells genetically selected from differentiating mouse embryonic stem cells incorporate at sites of neovascularization in vivo. J Cell Sci. 2002;115: Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282: Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Kasahara H, Rota M, Musso E, Urbanek K, Leri A, Kajstura J, Nadal- Ginard B, Anversa P. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114: Oh H, Bradfute SB, Gallardo TD, Nakamura T, Gaussin V, Mishina Y, Pocius J, Michael LH, Behringer RR, Garry DJ, Entman ML, Schneider MD. Cardiac progenitor cells from adult myocardium: homing, differentiation, and fusion after infarction. Proc Natl Acad Sci U S A. 2003; 100: Messina E, De Angelis L, Frati G, Morrone S, Chimenti S, Fiordaliso F, Salio M, Battaglia M, Latronico MVG, Coletta M, Vivarelli E, Frati L, Cossu G, Giacomello A. Isolation and expansion of adult cardiac stem cells from human and murine heart. Circ Res. 2004;95: Laugwitz K-L, Moretti A, Lam J, Gruber P, Chen Y, Woodard S, Lin L-Z, Cai C-L, Lu MM, Reth M, Platoshyn O, Yuan JXJ, Evans S, Chien KR. Postnatal isl1 cardioblasts enter fully differentiated cardiomyocyte lineages. Nature. 2005;433: Martin CM, Meeson AP, Robertson SM, Hawke TJ, Richardson JA, Bates S, Goetsch SC, Gallardo TD, Garry DJ. Persistent expression of the ATP-binding cassette transporter, Abcg2, identifies cardiac SP cells in the developing and adult heart. Dev Biol. 2004;265: Urbanek K, Torella D, Sheikh F, Angelis AD, Nurzynska D, Silvestri F, Beltrami CA, Bussani R, Beltrami AP, Quaini F, Bolli R, Leri A, Kajstura J, Anversa P. Myocardial regeneration by activation of multipotent cardiac stem cells in ischemic heart failure. Proc Natl Acad Sci USA. 2005;102: Mouquet F, Pfister O, Jain M, Oikonomopoulos A, Ngoy S, Summer R, Fine A, Liao R. Restoration of cardiac progenitor cells after myocardial infarction by self-proliferation and selective homing of bone marrowderived stem cells. Circ Res. 2005;97: Smith RR, Barile L, Cho HC, Abraham MR, Messina E, Giacomello A, Marban E. Unique phenotype of cardiospheres derived from human endomyocardial biopsies. Circulation. 2005;112(suppl II):II-334. Abstract. 39. Siminiak T, Kalmucki P, Kurpisz M. Autologous skeletal myoblasts for myocardial regeneration. J Interv Cardiol. 2004;17: Murry CE, Wiseman RW, Schwartz SM, Hauschka SD. Skeletal myoblast transplantation for repair of myocardial necrosis. J Clin Invest. 1996;98: Hagege AA, Carrion C, Menasche P, Vilquin J-T, Duboc D, Marolleau J-P, Desnos M, Bruneval P. Viability and differentiation of autologous skeletal myoblast grafts in ischaemic cardiomyopathy. Lancet. 2003; 361: Taylor DA, Atkins BZ, Hungspreugs P, Jones TR, Reedy MC, Hutcheson KA, Glower DD, Kraus WE. Regenerating functional myocardium: improved performance after skeletal myoblast transplantation. Nat Med. 1998;4: Menasche P. Skeletal myoblast transplantation for cardiac repair. Exp Rev Cardiovasc Ther. 2004;2: Menasche P, Hagege AA, Vilquin J-T, Desnos M, Abergel E, Pouzet B, Bel A, Sarateanu S, Scorsin M, Schwartz K. Autologous skeletal myoblast transplantation for severe postinfarction left ventricular dysfunction. J Am Coll Cardiol. 2003;41: Siminiak T, Kalawski R, Fiszer D, Jerzykowska O, Rzeniczak J, Rozwadowska N, Kurpisz M. 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: Jackson KA, Majka SM, Wang H, Pocius J, Hartley CJ, Majesky MW, Entman ML, Michael LH, Hirschi KK, Goodell MA. Regeneration of ischemic cardiac muscle and vascular endothelium by adult stem cells. J Clin Invest. 2001;107: Orlic D, Kajstura J, Chimenti S, Limana F, Jakoniuk I, Quaini F, Nadal-Ginard B, Bodine DM, Leri A, Anversa P. Mobilized bone marrow cells repair the infarcted heart, improving function and survival. Proc Natl Acad Sci U S A. 2001;98: Murry CE, Soonpaa MH, Reinecke H, Nakajima H, Nakajima HO, Rubart M, Pasumarthi KBS, Ismail Virag J, Bartelmez SH, Poppa V, Bradford G, Dowell JD, Williams DA, Field LJ. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature. 2004;428: Balsam LB, Wagers AJ, Christensen JL, Kofidis T, Weissman IL, Robbins RC. Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium. Nature. 2004;428: Strauer BE, Brehm M, Zeus T, Kostering M, Hernandez A, Sorg RV, Kogler G, Wernet P. Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans. Circulation. 2002;106: Assmus B, Schachinger V, Teupe C, Britten M, Lehmann R, Dobert N, Grunwald F, Aicher A, Urbich C, Martin H, Hoelzer D, Dimmeler S, Zeiher A. Transplantation of Progenitor Cells and Regeneration Enhancement in Acute Myocardial Infarction (TOPCARE-AMI). Circulation. 2002;106: Schachinger V, Assmus B, Britten MB, Honold J, Lehmann R, Teupe C, Abolmaali ND, Vogl TJ, Hofmann W-K, Martin H. 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: Wollert KC, Meyer GP, Lotz J, Ringes-Lichtenberg S, Lippolt P, Breidenbach C, Fichtner S, Korte T, Hornig B, Messinger D. Intracoronary autologous bone-marrow cell transfer after myocardial infarction: the BOOST randomised controlled clinical trial. Lancet. 2004;364:

12 350 Circulation July 25, Janssens S, Dubois C, Bogaert J, Theunissen K, Deroose C, Desmet W, Kalantzi M, Herbots L, Sinnaeve P, Dens J. Autologous bone marrowderived stem-cell transfer in patients with ST-segment elevation myocardial infarction: double-blind, randomised controlled trial. Lancet. 2005;367: Meyer GP, Wollert KC, Lotz J, Steffens J, Lippolt P, Fichtner S, Hecker H, Schaefer A, Arseniev L, Hertenstein B, Ganser A, Drexler H. 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: Perin EC, Dohmann HFR, Borojevic R, Silva SA, Sousa ALS, Mesquita CT, Rossi MID, Carvalho AC, Dutra HS, Dohmann HJF, Silva GV, Belem L, Vivacqua R, Rangel FOD, Esporcatte R, Geng YJ, Vaughn WK, Assad JAR, Mesquita ET, Willerson JT. Transendocardial, autologous bone marrow cell transplantation for severe, chronic ischemic heart failure. Circulation. 2003;107: Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9: Zimmet J, Hare J. Emerging role for bone marrow derived mesenchymal stem cells in myocardial regenerative therapy. Basic Res Cardiol. 2005; 100: Majumdar MK, Keane-Moore M, Buyaner D, Hardy WB, Moorman MA, McIntosh KR, Mosca JD. Characterization and functionality of cell surface molecules on human mesenchymal stem cells. J Biomed Sci. 2003;10: Haynesworth SE, Baber MA, Caplan AI. Cell surface antigens on human marrow-derived mesenchymal cells are detected by monoclonal antibodies. Bone Marrow Transplant. 1992;13: Pittenger MF, Martin BJ. Mesenchymal stem cells and their potential as cardiac therapeutics. Circ Res. 2004;95: Makino S, Fukuda K, Miyoshi S, Konishi F, Kodama H, Pan J, Sano M, Takahashi T, Hori S, Abe H, Hata J-I, Umezawa A, Ogawa S. Cardiomyocytes can be generated from marrow stromal cells in vitro. J Clin Invest. 1999;103: Min J-Y, Sullivan MF, Yang Y, Zhang J-P, Converso KL, Morgan JP, Xiao Y-Fu. Significant improvement of heart function by cotransplantation of human mesenchymal stem cells and fetal cardiomyocytes in postinfarcted pigs. Ann Thorac Surg. 2002;74: Shake JG, Gruber PJ, Baumgartner WA, Senechal G, Meyers J, Redmond JM, Pittenger MF, Martin BJ. Mesenchymal stem cell implantation in a swine myocardial infarct model: engraftment and functional effects. Ann Thorac Surg. 2002;73: Toma C, Pittenger MF, Cahill KS, Byrne BJ, Kessler PD. Human mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart. Circulation. 2002;105: Thompson CA, Nasseri BA, Makower J, Houser S, McGarry M, Lamson T, Pomerantseva I, Chang JY, Gold HK, Vacanti JP, Oesterle SN. Percutaneous transvenous cellular cardiomyoplasty: a novel nonsurgical approach for myocardial cell transplantation. J Am Coll Cardiol. 2003; 41: Rangappa S, Reddy V, Bongso A, Lee EH, Sim EKW. Transformation of the adult human mesenchymal stem cells into cardiomyocyte-like cells in vivo. Cardiovasc Eng. 2002;2: Gojo S, Gojo N, Takeda Y, Mori T, Abe H, Kyo S, Hata J-i, Umezawa A. In vivo cardiovasculogenesis by direct injection of isolated adult mesenchymal stem cells. Exp Cell Res. 2003;288: Mangi AA, Noiseux N, Kong D, He H, Rezvani M, Ingwall JS, Dzau V. Mesenchymal stem cells modified with Akt prevent remodeling and restore performance of infarcted hearts. Nat Med. 2003;9: Davani S, Marandin A, Mersin N, Royer B, Kantelip B, Herve P, Etievent J-P, Kantelip J-P. Mesenchymal progenitor cells differentiate into an endothelial phenotype, enhance vascular density, and improve heart function in a rat cellular cardiomyoplasty model. Circulation. 2003;108(suppl II):II-253 II Ryan JM, Barry FP, Murphy JM, Mahon BP. Mesenchymal stem cells avoid allogeneic rejection. J Inflamm (Lond). 2005;2: Chen S-l, Fang W-W, Ye F, Liu Y-H, Qian J, Shan S-J, Zhang J-J, Chunhua RZ, Liao L-M, Lin S, Sun J-P. 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: Eichmann A, Corbel C, Nataf V, Vaigot P, Breant C, Le Douarin NM. Ligand-dependent development of the endothelial and hemopoietic lineages from embryonic mesodermal cells expressing vascular endothelial growth factor receptor 2. Proc Natl Acad Sci U S A. 1997;94: Sato TN, Qin Y, Kozak CA, Audus KL Tie-1 and Tie-2 define another class of putative receptor tyrosine kinase genes expressed in early embryonic vascular system [erratum in Proc Natl Acad Sci U S A. 1993;90:12056]. Proc Natl Acad Sci U S A. 1993;90: Suri C, Jones PF, Patan S, Bartunkova S, Maisonpierre PC, Davis S, Sato TN, Yancopoulos GD. Requisite role of angiopoietin-1, a ligand for the TIE2 receptor, during embryonic angiogenesis. Cell. 1996;87: Yin AH, Miraglia S, Zanjani ED, Almeida-Porada G, Ogawa M, Leary AG, Olweus J, Kearney J, Buck DW. AC133, a novel marker for human hematopoietic stem and progenitor cells. Blood. 1997;90: Peichev M, Naiyer AJ, Pereira D, Zhu Z, Lane WJ, Williams M, Oz MC, Hicklin DJ, Witte L, Moore MA, Rafii S. Expression of VEGFR-2 and AC133 by circulating human CD34 cells identifies a population of functional endothelial precursors. Blood. 2000;95: Erb S, Linke A, Adams V, Lenk K, Thiele H, Diederich K-W, Emmrich F, Kluge R, Kendziorra K, Sabri O, Schuler G, Hambrecht R. Transplantation of blood-derived progenitor cells after recanalization of chronic coronary artery occlusion: first randomized and placebocontrolled study. Circ Res. 2005;97: Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol. 2000;109: Mayani H, Lansdorp PM. Biology of human umbilical cord bloodderived hematopoietic stem/progenitor cells. Stem Cells. 1998;16: Kogler G, Sensken S, Airey JA, Trapp T, Muschen M, Feldhahn N, Liedtke S, Sorg RV, Fischer J, Rosenbaum C, Greschat S, Knipper A, Bender J, Degistirici O, Gao J, Caplan AI, Colletti EJ, Almeida-Porada G, Muller HW, Zanjani E, Wernet P. A new human somatic stem cell from placental cord blood with intrinsic pluripotent differentiation potential. J Exp Med. 2004;200: Kim B-O, Tian H, Prasongsukarn K, Wu J, Angoulvant D, Wnendt S, Muhs A, Spitkovsky D, Li R-K. Cell transplantation improves ventricular function after a myocardial infarction: a preclinical study of human unrestricted somatic stem cells in a porcine model. Circulation. 2005; 112(suppl I):I-96 I Ma N, Stamm C, Kaminski A, Li W, Kleine H-D, Muller-Hilke B, Zhang L, Ladilov Y, Egger D, Steinhoff G. Human cord blood cells induce angiogenesis following myocardial infarction in NOD/scid-mice. Cardiovasc Res. 2005;66: Hirata Y, Sata M, Motomura N, Takanashi M, Suematsu Y, Ono M, Takamoto S. Human umbilical cord blood cells improve cardiac function after myocardial infarction. Biochem Biophys Res Commun. 2005;327: World Medical Association. Declaration of Helsinki. Available at: Accessed December 3, Vulliet PR, Greeley M, Halloran SM, MacDonald KA, Kittleson MD. Intra-coronary arterial injection of mesenchymal stromal cells and microinfarction in dogs. Lancet. 2004;363: Schwarz ER, Speakman MT, Patterson M, Hale SS, Isner JM, Kedes LH, Kloner RA. Evaluation of the effects of intramyocardial injection of DNA expressing vascular endothelial growth factor (VEGF) in a myocardial infarction model in the rat: angiogenesis and angioma formation. J Am Coll Cardiol. 2000;35: Rubio D, Garcia-Castro J, Martin MC, de la Fuente R, Cigudosa JC, Lloyd AC, Bernad A. Spontaneous human adult stem cell transformation. Cancer Res. 2005;65: Faggiotto A, Paoletti R. Statins and blockers of the renin-angiotensin system: vascular protection beyond their primary mode of action. Hypertension. 1999;34: Calabrò P, Yeh ETH. The pleiotropic effects of statins. Curr Opin Cardiol. 2005;20: Gruntzig A, Schneider HJ. The percutaneous dilatation of chronic coronary stenoses-experiments and morphology [in German]. Schweiz Med Wochenschr. 1977;107:1588.

13 Stem Cell Therapy Gruntzig A. Transluminal dilatation of coronary artery stenosis. Lancet. 1978;311: Gruntzig AR, Senning A, Siegenthaler WE. Nonoperative dilatation of coronary-artery stenosis: percutaneous transluminal coronary angioplasty. N Engl J Med. 1979;301: Morice M-C, Serruys PW, Sousa JE, Fajadet J, Ban Hayashi E, Perin M, Colombo A, Schuler G, Barragan P, Guagliumi G, Molnar F, Falotico R, for the RAVEL Study Group. A randomized comparison of a sirolimuseluting stent with a standard stent for coronary revascularization. N Engl J Med. 2002;346: Sousa JE, Costa MA, Abizaid A, Abizaid AS, Feres F, Pinto IMF, Seixas AC, Staico R, Mattos LA, Sousa AGMR, Falotico R, Jaeger J, Popma JJ, Serruys PW. Lack of neointimal proliferation after implantation of sirolimus-coated stents in human coronary arteries: a quantitative coronary angiography and three-dimensional intravascular ultrasound study. Circulation. 2001;103: Sousa JE, Costa MA, Abizaid AC, Rensing BJ, Abizaid AS, Tanajura LF, Kozuma K, Van Langenhove G, Sousa AGMR, Falotico R, Jaeger J, Popma JJ, Serruys PW. Sustained suppression of neointimal proliferation by sirolimus-eluting stents: one-year angiographic and intravascular ultrasound follow-up. Circulation. 2001;104: Rensing BJ, Vos J, Smits PC, Foley DP, van den Brand MJBM, van der Giessen WJ, de Feijter PJ, Serruys PW. Coronary restenosis elimination with a sirolimus eluting stent: first European human experience with 6-month angiographic and intravascular ultrasonic follow-up. Eur Heart J. 2001;22: Cardiac Arrhythmia Suppression Trial (CAST) Investigators. Preliminary report: effect of encainide and flecainide on mortality in a randomized trial of arrhythmia suppression after myocardial infarction. N Engl J Med. 1989;321: Baker M. Stem cell therapy or snake oil? Nat Biotechnol. 2005;23: FDA. Food and Drug Administration Modernization Act of 1997, publication L , 113, information program on clinical trials for serious or life-threatening diseases. Available at: cber/fdama.htm. Accessed March 1, Zarin DA, Tse T, Ide NC. Trial registration at ClinicalTrials.gov between May and October N Engl J Med. 2005;353: De Angelis C, Drazen JM, Frizelle FA, Haug C, Hoey J, Horton R, Kotzin S, Laine C, Marusic A, Overbeke AJPM, Schroeder TV, Sox HC, Van Der Weyden MB. Clinical trial registration: a statement from the International Committee of Medical Journal Editors. N Engl J Med. 2004;351: Stamm C, Westphal B, Kleine H-D, Petzsch M, Kittner C, Klinge H, Schümichen C, Nienaber CA, Freund M, Steinhoff G. Autologous bonemarrow stem-cell transplantation for myocardial regeneration. Lancet. 2003;361: Stamm C, Kleine H-D, Westphal B, Petzsch M, Kittner C, Nienaber CA, Freund M, Steinhoff G. CABG and bone marrow stem cell transplantation after myocardial infarction. Thorac Cardiovasc Surg. 2004;52: Tse H-F, Kwong Y-L, Chan JK, Lo G, Ho C-L, Lau C-P. Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet. 2003;361: Fernandez-Aviles F, San Roman JA, Garcia-Frade J, Fernandez ME, Penarrubia MJ, de la Fuente L, Gomez-Bueno M, Cantalapiedra A, Fernandez J, Gutierrez O, Sanchez PL, Hernandez C, Sanz R, Garcia- Sancho J, Sanchez A. Experimental and clinical regenerative capability of human bone marrow cells after myocardial infarction. Circ Res. 2004;95: 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 Cardiac Surg. 2003;18:S69 S Strauer BE, Brehm M, Zeus T, Bartsch T, Schannwell C, Antke C, Sorg RV, Kogler G, Wernet P, Muller H-W, Kostering M. 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: Katritsis DG, Sotiropoulou PA, Karvouni E, Karabinos I, Korovesis S, Perez SA, Voridis EM, Papamichail M. Transcoronary transplantation of autologous mesenchymal stem cells and endothelial progenitors into infarcted human myocardium. Catheter Cardiovasc Interv. 2005;65: Galinanes M, Loubani M, Davies J, Chin D, Pasi J, Bell PR. Autotransplantation of unmanipulated bone marrow into scarred myocardium is safe and enhances cardiac function in humans. Cell Transplany. 2004; 13: Fuchs S, Satler LF, Kornowski R, Okubagzi P, Weisz G, Baffour R, Waksman R, Weissman NJ, Cerqueira M, Leon MB, Epstein SE. 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: Kang H-J, Kim H-S, Zhang S-Y, Park K-W, Cho H-J, Koo B-K, Kim Y-J, Lee DS, Sohn D-W, Han K-S, Oh B-H, Lee M-M, Park Y-B. 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: Ozbaran M, Omay SB, Nalbantgil S, Kultursay H, Kumanlioglu K, Nart D, Pektok E. Autologous peripheral stem cell transplantation in patients with congestive heart failure due to ischemic heart disease. Eur J Cardiothorac Surg. 2004;25: Boyle AJ, Whitbourn R, Schlicht S, Krum H, Kocher A, Nandurkar H, Bergmann S, Daniell M, O Day J, Skerrett D, Haylock D, Gilbert RE, Itescu S. Intra-coronary high-dose CD34 stem cells in patients with chronic ischemic heart disease: A 12-month follow-up. Int J Cardiol. 2006;109: Pompilio G, Cannata A, Peccatori F, Bertolini F, Nascimbene A, Capogrossi MC, Biglioli P. Autologous peripheral blood stem cell transplantation for myocardial regeneration: a novel strategy for cell collection and surgical injection. Ann Thorac Surg. 2004;78: Dib N, Michler RE, Pagani FD, Wright S, Kereiakes DJ, Lengerich R, Binkley P, Buchele D, Anand I, Swingen C, Di Carli MF, Thomas JD, Jaber WA, Opie SR, Campbell A, McCarthy P, Yeager M, Dilsizian V, Griffith BP, Korn R, Kreuger SK, Ghazoul M, MacLellan WR, Fonarow G, Eisen HJ, Dinsmore J, Diethrich E. Safety and feasibility of autologous myoblast transplantation in patients with ischemic cardiomyopathy: four-year follow-up. Circulation. 2005;112: Siminiak T, Fiszer D, Jerzykowska O, Grygielska B, Rozwadowska N, Kalmucki P, Kurpisz M. Percutaneous trans-coronary-venous transplantation of autologous skeletal myoblasts in the treatment of postinfarction myocardial contractility impairment: the POZNAN trial. Eur Heart J. 2005;26: Ince H, Petzsch M, Rehders TC, Chatterjee T, Nienaber CA. Transcatheter transplantation of autologous skeletal myoblasts in postinfarction patients with severe left ventricular dysfunction. J Endovasc Ther. 2004;11: Smits PC, van Geuns R-JM, Poldermans D, Bountioukos M, Onderwater EEM, Lee CH, Maat APWM, Serruys PW. 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: Pagani FD, DerSimonian H, Zawadzka A, Wetzel K, Edge ASB, Jacoby DB, Dinsmore JH, Wright S, Aretz TH, Eisen HJ, Aaronson KD. Autologous skeletal myoblasts transplanted to ischemia-damaged myocardium in humans: histological analysis of cell survival and differentiation. J Am Coll Cardiol. 2003;41: Herreros J, Prosper F, Perez A, Gavira JJ, Garcia-Velloso MJ, Barba J, Sanchez PL, Canizo C, Rabago G, Marti-Climent JM, Hernandez M, Lopez-Holgado N, Gonzalez-Santos JM, Martin-Luengo C, Alegria E. Autologous intramyocardial injection of cultured skeletal musclederived stem cells in patients with non-acute myocardial infarction. Eur Heart J. 2003;24: Bang OY, Lee JS, Lee PH, Lee G. Autologous mesenchymal stem cell transplantation in stroke patients. Ann Neurol. 2005;57: Garcia-Olmo D, Garcia-Arranz M, Herreros D, Pascual I, Peiro C, Rodriguez-Montes JA. A phase I clinical trial of the treatment of Crohn s fistula by adipose mesenchymal stem cell transplantation. Dis Colon Rectum. 2005;48:

14 352 Circulation July 25, Horwitz EM, Gordon PL, Koo WKK, Marx JC, Neel MD, McNall RY, Muul L, Hofmann T. Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: implications for cell therapy of bone. Proc Natl Acad Sci. 2002;99: Le Blanc K, Rasmusson I, Sundberg B, Gotherstrom C, Hassan M, Uzunel M, Ringden O. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet. 2004;363: Amado LC, Schuleri KH, Saliaris AP, Boyle AJ, Helm R, Oskouei B, Centola M, Eneboe V, Young R, Lima JAC, Lardo AC, Heldman AW, Hare JM. Multi-modality non-invasive imaging demonstrates in-vivo cardiac regeneration following mesenchymal stem cell therapy. JAm Coll Cardiol. In press. Response to Boyle et al Peter Oettgen, MD Boyle et al raise several important points about the current status of stem cell therapy for cardiac repair and nicely review the results of multiple clinical trials to date. The vast majority of these trials were conducted using small numbers of patients with mixed results. Larger, recently completed, prospective, randomized, clinical trials have had similarly mixed results. Significant gaps remain with respect to our understanding of which stem cells may ultimately be the most therapeutically useful to promote cardiac tissue regeneration and how best to deliver the stem cells. Because the vast majority of stem cells do not remain within the heart, even after local injection, it is critical that better ways be developed to promote retention of the delivered cells within the heart and to be able to track and monitor the stem cells that do not remain within the heart after delivery. Other safety concerns include the progression of vascular disease and the development of arrhythmias. Do the results of the completed studies warrant initiation of larger additional clinical trials? The financial resources available for conducting stem cell research are limited. Large clinical trials, in particular, are costly. Although a significant unmet clinical need remains in patients with cardiovascular disease, we must carefully weigh how much of these resources should be devoted to additional basic research in stem cell biology and studies aimed at optimizing their therapeutic potential in animal models of cardiovascular disease versus conducting additional large clinical trials.

15 CONTROVERSIES IN CARDIOVASCULAR MEDICINE Cardiac Stem Cell Therapy Need for Optimization of Efficacy and Safety Monitoring Peter Oettgen, MD Cardiovascular disease remains the number one cause of morbidity and mortality in the United States and Europe. In the United States alone, 1 million patients suffer a myocardial infarction every year, with an associated mortality of 25% at 3 years. 1 A more sobering statistic is the fact that there are 5 million Americans with congestive heart failure, with an associated 20% mortality per year. This remains the case despite advances in pharmacotherapy, cardiac resynchronization therapies, and the use of implantable cardioverter-defibrillators. 2 Some patients with end-stage congestive heart failure are considered for cardiac transplantation, but the demand for this therapeutic approach greatly outweighs the availability of donor hearts. Over the past few years, several animal studies and a few clinical trials have supported the use of stem cells as a potential therapeutic modality to address this unmet clinical need. Response by Boyle et al p 358 Type of Cells Used for Cardiac Transplantation Several different types of cells have been used in both animal studies and patients to promote the repair of damaged myocardium. The 2 main sources of stem cells are adult stem and embryonic stem (ES) cells. ES Cells ES cells are derived from the inner mass of developing embryos during the blastocyst stage. Characteristic features of ES cells include their proliferative and self-renewing properties and their ability to differentiate into a wide variety of cell types, including cardiac myocytes. 3 The major concerns with their use in human trials include the formation of teratomas when ES cells are injected into immunocompromised animals. This is particularly important because the ES cells currently available for use in humans would be of allogeneic origin and therefore would require immunosuppression. As nuclear transfer techniques improve, they will provide a way of generating an unlimited supply of histocompatible ES cells using the nuclei of cells obtained directly from the recipient patients with heart disease. 4 Adult Stem Cells Bone Marrow Derived Stem Cells Several different types of stem cells can be isolated from adult bone marrow. Examples of some of these subpopulations of cells include hematopoietic stem cells, endothelial progenitors, and mesenchymal stem cells. Several investigators have chosen to deliver unfractionated bone marrow derived cells, a technique that has the advantage of minimizing extensive ex vivo manipulation of the cells to isolate and expand a selected population of cells. The potential disadvantage of delivering a mixture of cells is that the percentage of cells that are therapeutically useful may be small. An alternative strategy is to isolate The opinions expressed in this article are not necessarily those of the editors or of the American Heart Association. From the Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, Mass. Correspondence to Peter Oettgen, MD, Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA joettgen@bidmc.harvard.edu (Circulation. 2006;114: ) 2006 American Heart Association, Inc. Circulation is available at DOI: /CIRCULATIONAHA

16 354 Circulation July 25, 2006 purer populations of cells that express specific antigens. For example, endothelial progenitors express the cell surface marker CD133. These cells have a greater potential to promote angiogenesis but are more technically challenging to isolate in significant quantities. 5 Mesenchymal cells represent a rare population of bone marrow derived cells that do not express CD34 or CD133. Mesenchymal cells can differentiate into bone, cartilage, adipocytes, and, under certain culture conditions, cardiac myocytes. 6 One advantage of using mesenchymal cells is that clones of these cells can easily be expanded in vitro, exhibit relatively low immunogenicity, and might be particularly useful when autologous stem cells are not readily available. 7 Skeletal Myoblasts Skeletal myoblasts are a population of progenitor cells that can be isolated from skeletal muscle biopsies and expanded in vitro. These myoblasts can differentiate into myotubes and exhibit skeletal muscle phenotype after transplantation, leading to improvements in left ventricular (LV) systolic and diastolic function. 8 However, the transplanted skeletal myocytes are not electrically coupled to surrounding cardiomyocytes and thus may lead to the development of arrhythmias. Resident Cardiac Stem Cells Several investigators have recently identified a population of stem cells within the myocardium that are capable of differentiating into cardiac myocytes. It has recently been reported that these cells can be harvested from cardiac biopsies. Injecting these cells in the setting of myocardial infarction can promote cardiomyocyte formation with associated improvements in systolic function. 9 At present, these cells are limited in number and require ex vivo separation and expansion over several weeks. Methods of Stem Cell Delivery A major goal of cardiac stem cell therapy is to transplant enough cells into the myocardium at the site of injury or infarction to maximize restoration of function. Several different approaches currently are being used to deliver stem cells. Transvascular Route A transvascular approach is particularly well suited to treat patients with acutely infarcted and reperfused myocardium. Stem cells can be infused directly into the coronary arteries and have a greater likelihood of remaining in the injured myocardium as a result of the activation of adhesion molecules and chemokines. 10 The advantage of an intracoronary infusion is that the cells can be directed to a particular territory. An alternative approach is to inject stem cells intravenously. 11 In the setting of myocardial infarction, circulating stem cells have been shown to home to sites of injury, but the number of cells that home to the heart in this way is significantly less than by local injection. Direct Injection Into the Ventricular Wall Direct injection of stem cells is used in patients presenting with established cardiac dysfunction in whom a transvascular approach may not be possible because of total occlusion or poor flow within the vessel of the affected territory. There are 3 different approaches to direct injection. A transendocardial approach can be used in which a needle catheter is advanced across the aortic valve and positioned against the endocardial surface. 12 Cells can then be injected directly into the left ventricle. Electrophysiological mapping can be used to differentiate sites of viable, ischemic, or scarred myocardium. In a transepicardial approach, cells are injected during open heart surgery. The advantage of this approach is that it allows direct visualization of the myocardium and easier identification of regions of scar and border zones of infarcted tissues. A third approach involves the delivery of cells through one of the cardiac veins directly into the myocardium. 13 The limitation of this approach is that positioning the catheter within a particular coronary vein may be considerably more time consuming and technically challenging. Safety Concerns Arrhythmias Over the past few years, some of the early-phase clinical studies have suggested the possibility of a proarrhythmic effect associated with stem cell transplantation. In 1 study, skeletal myoblasts were injected transepicardially at the time of coronary artery bypass surgery. Four patients had documented ventricular tachycardia at 11, 12, 13, and 22 days after stem cell implantation. 14 Interestingly, these events occurred early and were not observed in treated patients later after several months of follow-up. A similar proarrhythmic effect was observed when autologous skeletal myoblasts were delivered via a transvascular route. 15 Other studies have similarly reported an increased frequency of nonsustained ventricular tachycardia in patients treated with skeletal myoblasts, peaking 11 to 30 days after stem cell transplantation. 16,17 A proposed mechanism for the increased incidence of arrhythmias is that the injected stem cells do not communicate electrically with neighboring cardiac myocytes and/or result in slowed conduction, thereby promoting reentrant arrhythmias. It has recently been suggested that skeletal myoblasts that have been genetically engineered to express gap junction protein connexin 43 exhibited decreased arrhythmogenicity. 18 Although proarrhythmic effects have been observed predominantly in patients receiving skeletal myoblast transplantation, they also have been observed recently in 2 patients shortly after transplantation of CD133 cells. 19,20 Restenosis, Accelerated Atherosclerosis, and Coronary Obstruction There have been conflicting reports regarding the potential for increased restenosis after stem cell transplantation. In 1 study, a high rate of restenosis was observed after intracoronary delivery of peripheral blood stem cells mobilized with granulocyte colony-stimulating factor in the setting of myocardial infarction and stent placement. 21 In another study, CD133 cells were delivered via intracoronary injection in

17 Stem Cell Therapy 355 the setting of myocardial infarction, with in-stent restenosis rates of 37% and reocclusion rates of 11%. 19 Relatively low rates of restenosis were observed in earlier studies using bone marrow derived stem cells. 10,20 In addition to restenosis, it is also possible that stem cell transplantation may promote the formation of de novo lesions or atherosclerotic plaque progression. In 2 recent studies, there was a fairly high proportion of new lesions identified in the nonstented vessels after stem cell transplantation. 19,22 It is also possible that if the cells are delivered at a high enough concentration via the coronary circulation, they may adhere to each other, form aggregates, and thereby lead to the occlusion of microvessels. In 1 study in which mesenchymal stem cells were administered by intracoronary injection in pigs, there was associated occlusion of microvessels and macrovessels. 23 Abnormal Cellular Differentiation Fortunately, no clinical trials to date that have used stem cells to promote cardiac tissue regeneration have demonstrated an increased frequency of tumor formation. However, most of the clinical trials have been conducted on small numbers of patients. Furthermore, it is not clear how adequate testing would be conducted to monitor for this potential side effect. Because stem cells are known to migrate to several other organs after delivery to the heart, it is conceivable that aberrant cellular differentiation with the potential of tumor formation could occur in any of these organs. Tracking of Stem Cells One of the major concerns regarding the delivery of stem cells is determining which cells remain in the heart and which cells ultimately end up in other organs as a result of a washout effect. 24 Within a few hours after transplantation, stem cells injected locally within the heart also are observed within the lungs, spleen, liver, and kidney. One day after transplantation of neonatal cardiac myocytes into rat hearts, only 24% of the originally injected cells remained in the heart. 25 Given the small fraction of stem cells that remain within the heart after injection and the multiple organs to which the stem cells migrate, it is imperative that better methods of tracking stem cells be developed to determine the fate of these cells after transplantation. Several potential methods have been developed to label and track stem cells in animal models, including scintigraphy, PET, and MRI PET scanning and MRI also have been tested recently in humans to track stem cells. 29,30 One hour after injection of 18 F-fluorodeoxyglucose labeled CD34 cells, only 5.5% of the cells were detectable in the heart by PET scanning. Unfortunately, because of the short half-life of 18 F-fluorodeoxyglucose, other isotopes with a longer half-life may need to be evaluated for optimal longterm tracking of stem cells. Evidence for Tissue Regeneration The ultimate goal of stem cell therapy is to promote cardiac tissue regeneration so that the regenerated cardiac tissue leads to improvements in cardiac function in a fashion that is synchronized with the rest of the functioning heart in the absence of proarrhythmic or other adverse effects. More recently, however, there is evidence that stem cells may lead to improvements in cardiac function that are independent of tissue regeneration. Although early studies supported the ability of bone marrow derived mononuclear cells to differentiate into cardiac myocytes, subsequent studies failed to support these initial observations It has been suggested that the locally injected cells can act in a paracrine fashion to improve ventricular function through the release of growth factors or other paracrine mediators. These mediators may act to directly augment systolic function, prevent apoptosis of ischemic myocardial cells, or limit injury by promoting angiogenesis. 34 The locally injected stem cells would promote the salvage of injured myocardium rather than tissue regeneration. Additional long-term studies are needed to determine whether the improvements observed after weeks to a few months are generally sustained over longer periods of time. These paracrine effects are more likely to be useful in patients with acute myocardial ischemia or with hibernating myocardium and less likely to be beneficial in patients with chronically infarcted myocardium with significant scar formation. Significant challenges remain with regard to cardiac tissue regeneration. Future studies are needed to identify the best stem cell type to use. To promote cardiac tissue regeneration, sufficient numbers of cells will need to be delivered and maintained within the heart at the site of LV dysfunction, and the new tissue needs to be vascularized, electrically and mechanically coupled with the rest of the myocardium. The hope is that the strategies will include ways of replacing scarred or fibrotic tissue in regions of LV dysfunction. Unless autologous cells can be used to generate the cardiac tissue, potential graft rejection needs to be addressed. Real progress toward this goal will require the collaborative interaction of investigators with expertise in tissue engineering, molecular biology, electrophysiology, cardiac physiology, immunology, and vascular biology. Recent Clinical Trials Several small clinical studies using a variety of different cell types have shown some initial promise regarding the benefit of stem cell therapy, but these small clinical studies have several limitations. In addition to being small, some of the studies lacked adequate controls or randomization in a blinded fashion. Furthermore, some of the studies failed to assess infarct size or ventricular function before administration of the stem cells, and the follow-up period often was short. Results of recent randomized clinical trials evaluating the therapeutic effect of administering bone marrow derived mononuclear cells via intracoronary injection at the time of myocardial infarction have recently been reported (Table). Results of these studies have been mixed. The primary end point of these studies was LV ejection fraction. The administration of stem cells in 2 studies, BOne marrow transfer to

18 356 Circulation July 25, 2006 Randomized Clinical Trials of Stem Cell Therapy for MI Study Treated, n Cell Type/Treatment Mode of Delivery End Point P Janssens et al BMMNCs Intracoronary LV ejection fraction 0.36 BOOST BMMNCs Intracoronary LV ejection fraction ASTAMI 36, BMMNCs Intracoronary LV ejection fraction 0.05* REPAIR-AMI BMMNCs Intracoronary LV ejection fraction Zohlnhoefer et al G-CSF Subcutaneous Infarct size 0.79 STEMMI G-CSF Subcutaneous LV systolic wall thickening 1.00 BMMNCs indicates bone marrow mononuclear cells; G-CSF, granulocyte colony-stimulating factor; and STEMMI, Stem Cells in Myocardial Infarction. *LV ejection fraction was higher in the control group. enhance ST-elevation infarct regeneration (BOOST) and Reinfusion of Enriched Progenitor cells And Infarct Remodelling in Acute Myocardial Infarction (REPAIR-AMI), resulted in significant increases in LV ejection fraction. 35,36 The differences in ejection fraction of the treated and control groups at 6 months in the BOOST trial were 56.7% and 52.0%; in the REPAIR-AMI treated and control groups, the differences were 54% versus 50%. In contrast, in the study by Janssens et al, 37 no difference between control and treated groups was observed, and in the Autologous Stem cell Transplantation in Acute Myocardial Infarction (ASTAMI) trial, the LV ejection fraction was higher in the control group. 36,38 The cause of these differences is unclear but may relate to how the cells were prepared before delivery or to the fact that the baseline ejection fractions at the time of myocardial infarction were only mildly diminished. There are ongoing additional randomized trials. BOOST-II will enroll 200 patients with large myocardial infarctions and depressed ejection fractions to receive bone marrow derived mononuclear cells or placebo. The Myoblast Autologous Grafting in Ischemic Cardiomyopathy (MAGIC) trial will evaluate the effect of autologous skeletal muscle myoblasts in patients with chronic heart failure who are undergoing coronary bypass and defibrillator implantation. One attractive alternative to delivering autologous stem cells via local injection is to identify the mechanisms by which stem cells are recruited to the heart and try to augment mobilization of stem cells, particularly in the setting of acute infarction. Small clinical studies suggested that cytokines such as granulocyte colony-stimulating factor could promote the mobilization of bone marrow derived stem cells in the setting of myocardial infarction, leading to improvements in myocardial function. 39,40 The advantage of this approach is that the cells are autologous and can be mobilized via systemic injections of granulocyte colony-stimulating factor. Two larger randomized trials, consisting of 78 and 114 patients, similarly used granulocyte colonystimulating factor to mobilize stem cells in the setting of myocardial infarction. 41,42 Unfortunately, neither of these studies demonstrated a significant benefit with respect to cardiac function after 6 months. Furthermore, another smaller study in which intracoronary injections of stem cells isolated after stimulation with granulocyte colony-stimulating factor were administered in the setting of myocardial infarction resulted in an increased rate of restenosis. 21 Conclusions Stem cells remain a highly promising therapeutic modality that could address the large, unmet clinical need of treating patients throughout the world with significant cardiac dysfunction that cannot be adequately treated with conventional therapeutic approaches or cardiac transplantation because of the limited availability of this resource. On the basis of the mixed results of more recent larger clinical trials, we should err on the side of caution before committing precious resources to conduct additional large clinical trials. Several questions need to be addressed. First, have we identified which stem cell type to use? Second, have we determined the mechanisms by which stem cells promote myocardial function or repair? At present, there is limited evidence to support that stem cells used thus far in patients promote significant cardiac tissue regeneration. Third, can the stem cell be retained efficiently within the heart? Finally, can clinical trials be done in such a way that important safety issues will be adequately addressed? What methods will be used to monitor the development of life-threatening arrhythmias and to track injected stem cells throughout the body? Because the financial resources available for clinical and basic stem cell research are not unlimited and because of the high cost associated with conducting larger clinical trials, it is particularly important that we address the aforementioned questions before proceeding with larger clinical trials. It is clear that additional basic research is needed to optimize ways to promote cardiac tissue regeneration, to improve methods by which delivered stem cells will remain in the heart, and to optimize the way in which stem cells are tracked after delivery. Funding This work was supported by National Institutes of Health grant PO1-HL Disclosures Dr Oettgen has received a received a research grant from the NIH.

19 Stem Cell Therapy 357 References Chartbook on Cardiovascular Lung and Blood Diseases. Bethesda, Md: National Heart, Lung, and Blood Institute; Bardy GH, Lee KL, Mark DB, Poole JE, Packer DL, Boineau R, Domanski M, Troutman C, Anderson J, Johnson G, McNulty SE, Clapp- Channing N, Davidson-Ray LD, Fraulo ES, Fishbein DP, Luceri RM, Ip JH. Amiodarone or an implantable cardioverter-defibrillator for congestive heart failure. N Engl J Med. 2005;352: Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M, Gepstein A, Livne E, Binah O, Itskovitz-Eldor J, Gepstein L. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J Clin Invest. 2001;108: Lanza R, Moore MA, Wakayama T, Perry AC, Shieh JH, Hendrikx J, Leri A, Chimenti S, Monsen A, Nurzynska D, West MD, Kajstura J, Anversa P. Regeneration of the infarcted heart with stem cells derived by nuclear transplantation. Circ Res. 2004;94: Rafii S, Lyden D. Therapeutic stem and progenitor cell transplantation for organ vascularization and regeneration. Nat Med. 2003;9: Makino S, Fukuda K, Miyoshi S, Konishi F, Kodama H, Pan J, Sano M, Takahashi T, Hori S, Abe H, Hata J, Umezawa A, Ogawa S. Cardiomyocytes can be generated from marrow stromal cells in vitro. J Clin Invest. 1999;103: Pittenger MF, Martin BJ. Mesenchymal stem cells and their potential as cardiac therapeutics. Circ Res. 2004;95: Dowell JD, Rubart M, Pasumarthi KB, Soonpaa MH, Field LJ. Myocyte and myogenic stem cell transplantation in the heart. Cardiovasc Res. 2003;58: Messina E, De Angelis L, Frati G, Morrone S, Chimenti S, Fiordaliso F, Salio M, Battaglia M, Latronico MV, Coletta M, Vivarelli E, Frati L, Cossu G, Giacomello A. Isolation and expansion of adult cardiac stem cells from human and murine heart. Circ Res. 2004;95: Strauer BE, Brehm M, Zeus T, Kostering M, Hernandez A, Sorg RV, Kogler G, Wernet P. Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans. Circulation. 2002;106: Kocher AA, Schuster MD, Szabolcs MJ, Takuma S, Burkhoff D, Wang J, Homma S, Edwards NM, Itescu S. Neovascularization of ischemic myocardium by human bone-marrow-derived angioblasts prevents cardiomyocyte apoptosis, reduces remodeling and improves cardiac function. Nat Med. 2001;7: Tse HF, Kwong YL, Chan JK, Lo G, Ho CL, Lau CP. Angiogenesis in ischaemic myocardium by intramyocardial autologous bone marrow mononuclear cell implantation. Lancet. 2003;361: Thompson CA, Nasseri BA, Makower J, Houser S, McGarry M, Lamson T, Pomerantseva I, Chang JY, Gold HK, Vacanti JP, Oesterle SN. Percutaneous transvenous cellular cardiomyoplasty: a novel nonsurgical approach for myocardial cell transplantation. J Am Coll Cardiol. 2003; 41: Menasche P, Hagege AA, Vilquin JT, Desnos M, Abergel E, Pouzet B, Bel A, Sarateanu S, Scorsin M, Schwartz K, Bruneval P, Benbunan M, Marolleau JP, Duboc D. Autologous skeletal myoblast transplantation for severe postinfarction left ventricular dysfunction. J Am Coll Cardiol. 2003;41: Smits PC, van Geuns RJ, Poldermans D, Bountioukos M, Onderwater EE, Lee CH, Maat AP, Serruys PW. 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: Dib N, McCarthy P, Campbell A, Yeager M, Pagani FD, Wright S, MacLellan WR, Fonarow G, Eisen HJ, Michler RE, Binkley P, Buchele D, Korn R, Ghazoul M, Dinsmore J, Opie SR, Diethrich E. Feasibility and safety of autologous myoblast transplantation in patients with ischemic cardiomyopathy. Cell Transplant. 2005;14: Pagani FD, DerSimonian H, Zawadzka A, Wetzel K, Edge AS, Jacoby DB, Dinsmore JH, Wright S, Aretz TH, Eisen HJ, Aaronson KD. Autologous skeletal myoblasts transplanted to ischemia-damaged myocardium in humans: histological analysis of cell survival and differentiation. J Am Coll Cardiol. 2003;41: Abraham MR, Henrikson CA, Tung L, Chang MG, Aon M, Xue T, Li RA, O Rourke B, Marban E. Antiarrhythmic engineering of skeletal myoblasts for cardiac transplantation. Circ Res. 2005;97: Bartunek J, Vanderheyden M, Vandekerckhove B, Mansour S, De Bruyne B, De Bondt P, Van Haute I, Lootens N, Heyndrickx G, Wijns W. Intracoronary injection of CD133-positive enriched bone marrow progenitor cells promotes cardiac recovery after recent myocardial infarction: feasibility and safety. Circulation. 2005;112(suppl I):I-178 II Britten MB, Abolmaali ND, Assmus B, Lehmann R, Honold J, Schmitt J, Vogl TJ, Martin H, Schachinger V, Dimmeler S, Zeiher AM. Infarct remodeling after intracoronary progenitor cell treatment in patients with acute myocardial infarction (TOPCARE-AMI): mechanistic insights from serial contrast-enhanced magnetic resonance imaging. Circulation. 2003; 108: Kang HJ, Kim HS, Zhang SY, Park KW, Cho HJ, Koo BK, Kim YJ, Soo Lee D, Sohn DW, Han KS, Oh BH, Lee MM, Park YB. 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: Fernandez-Aviles F, San Roman JA, Garcia-Frade J, Fernandez ME, Penarrubia MJ, de la Fuente L, Gomez-Bueno M, Cantalapiedra A, Fernandez J, Gutierrez O, Sanchez PL, Hernandez C, Sanz R, Garcia-Sancho J, Sanchez A. Experimental and clinical regenerative capability of human bone marrow cells after myocardial infarction. Circ Res. 2004;95: Vulliet PR, Greeley M, Halloran SM, MacDonald KA, Kittleson MD. Intra-coronary arterial injection of mesenchymal stromal cells and microinfarction in dogs. Lancet. 2004;363: Dow J, Simkhovich BZ, Kedes L, Kloner RA. Washout of transplanted cells from the heart: a potential new hurdle for cell transplantation therapy. Cardiovasc Res. 2005;67: Muller-Ehmsen J, Whittaker P, Kloner RA, Dow JS, Sakoda T, Long TI, Laird PW, Kedes L. Survival and development of neonatal rat cardiomyocytes transplanted into adult myocardium. J Mol Cell Cardiol. 2002;34: Wu JC, Chen IY, Sundaresan G, Min JJ, De A, Qiao JH, Fishbein MC, Gambhir SS. Molecular imaging of cardiac cell transplantation in living animals using optical bioluminescence and positron emission tomography. Circulation. 2003;108: Himes N, Min JY, Lee R, Brown C, Shea J, Huang X, Xiao YF, Morgan JP, Burstein D, Oettgen P. In vivo MRI of embryonic stem cells in a mouse model of myocardial infarction. Magn Reson Med. 2004;52: Hill JM, Dick AJ, Raman VK, Thompson RB, Yu ZX, Hinds KA, Pessanha BS, Guttman MA, Varney TR, Martin BJ, Dunbar CE, McVeigh ER, Lederman RJ. Serial cardiac magnetic resonance imaging of injected mesenchymal stem cells. Circulation. 2003;108: Kraitchman DL, Heldman AW, Atalar E, Amado LC, Martin BJ, Pittenger MF, Hare JM, Bulte JW. In vivo magnetic resonance imaging of mesenchymal stem cells in myocardial infarction. Circulation. 2003;107: Blocklet D, Toungouz M, Berkenboom G, Lambermont M, Unger P, Preumont N, Stoupel E, Egrise D, Degaute JP, Goldman M, Goldman S. Myocardial homing of nonmobilized peripheral-blood CD34 cells after intracoronary injection. Stem Cells. 2006;24: Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, Pickel J, McKay R, Nadal-Ginard B, Bodine DM, Leri A, Anversa P. Bone marrow cells regenerate infarcted myocardium. Nature. 2001;410: Balsam LB, Wagers AJ, Christensen JL, Kofidis T, Weissman IL, Robbins RC. Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium. Nature. 2004;428: Murry CE, Soonpaa MH, Reinecke H, Nakajima H, Nakajima HO, Rubart M, Pasumarthi KB, Virag JI, Bartelmez SH, Poppa V, Bradford G, Dowell JD, Williams DA, Field LJ. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature. 2004;428: Yoshioka T, Ageyama N, Shibata H, Yasu T, Misawa Y, Takeuchi K, Matsui K, Yamamoto K, Terao K, Shimada K, Ikeda U, Ozawa K, Hanazono Y. Repair of infarcted myocardium mediated by transplanted bone marrowderived CD34 stem cells in a nonhuman primate model. Stem Cells. 2005;23: Wollert KC, Meyer GP, Lotz J, Ringes-Lichtenberg S, Lippolt P, Breidenbach C, Fichtner S, Korte T, Hornig B, Messinger D, Arseniev L,

20 358 Circulation July 25, 2006 Hertenstein B, Ganser A, Drexler H. Intracoronary autologous bonemarrow cell transfer after myocardial infarction: the BOOST randomised controlled clinical trial. Lancet. 2004;364: Cleland JG, Freemantle N, Coletta AP, Clark AL. Clinical trials update from the American Heart Association: REPAIR-AMI, ASTAMI, JELIS, MEGA, REVIVE-II, SURVIVE, and PROACTIVE. Eur J Heart Fail. 2006;8: Janssens S, Dubois C, Bogaert J, Theunissen K, Deroose C, Desmet W, Kalantzi M, Herbots L, Sinnaeve P, Dens J, Maertens J, Rademakers F, Dymarkowski S, Gheysens O, Van Cleemput J, Bormans G, Nuyts J, Belmans A, Mortelmans L, Boogaerts M, Van de Werf F. Autologous bone marrow-derived stem-cell transfer in patients with ST-segment elevation myocardial infarction: double-blind, randomised controlled trial. Lancet. 2006;367: Lunde K, Solheim S, Aakhus S, Arnesen H, Abdelnoor M, Forfang K. Autologous stem cell transplantation in acute myocardial infarction: the ASTAMI randomized controlled trial: intracoronary transplantation of autologous mononuclear bone marrow cells, study design and safety aspects. Scand Cardiovasc J. 2005;39: Kuethe F, Figulla HR, Herzau M, Voth M, Fritzenwanger M, Opfermann T, Pachmann K, Krack A, Sayer HG, Gottschild D, Werner GS. Response to Oettgen Treatment with granulocyte colony-stimulating factor for mobilization of bone marrow cells in patients with acute myocardial infarction. Am Heart J. 2005;150: Kuethe F, Figulla HR, Voth M, Richartz BM, Opfermann T, Sayer HG, Krack A, Fritzenwanger M, Hoffken K, Gottschild D, Werner GS. Mobilization of stem cells by granulocyte colony-stimulating factor for the regeneration of myocardial tissue after myocardial infarction [in German]. Dtsch Med Wochenschr. 2004;129: Zohlnhofer D, Ott I, Mehilli J, Schomig K, Michalk F, Ibrahim T, Meisetschlager G, von Wedel J, Bollwein H, Seyfarth M, Dirschinger J, Schmitt C, Schwaiger M, Kastrati A, Schomig A. Stem cell mobilization by granulocyte colonystimulating factor in patients with acute myocardial infarction: a randomized controlled trial. JAMA. 2006;295: Ripa RS, Jorgensen E, Wang Y, Thune JJ, Nilsson JC, Sondergaard L, Johnsen HE, Kober L, Grande P, Kastrup J. 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: Andrew J. Boyle, MBBS, PhD; Steven P. Schulman, MD; Joshua M. Hare, MD We agree with Dr Oettgen on many issues surrounding cell therapy. Preclinical studies demonstrate considerable promise for many cell types to effect cardiac repair, yet the initial promise from small animal work is far more difficult to demonstrate in humans. Moreover, early clinical trials of skeletal myoblasts and granulocyte colony-stimulating factor have discovered unexpected side effects not apparent in animal models. These rigorously designed early clinical studies and their unanticipated findings have now guided not only the design of next phase of clinical studies but also the future of basic and animal studies. Most importantly, we agree that understanding the mechanistic underpinnings of cell-based therapy is essential. Our area of fundamental disagreement relates to the role of the clinical trial in advancing the burgeoning field of cell-based therapy. We believe that these trials must proceed but must be conducted by responsible investigators concerned with patient safety and committed to incorporating mechanistic studies into the trials. Clinical development of any new therapy begins with a phase I study aimed at proving safety, and major trials contain independent Data Safety and Monitoring boards. Every detail regarding the mechanism of action of these cells cannot be appropriately determined in animal models because, by definition, that which happens in controlled animal experiments may not translate into humans, who are much more heterogeneous and have many comorbidities that affect responsiveness to novel therapies. There exists, by necessity, a synergy between basic science and clinical research. An incomplete understanding of molecular/cellular mechanisms should not halt the progression of clinical trials; quite the opposite is the case. The success or failure of clinical trials guides future basic science research, which, in turn, informs future clinical trials to achieve improvements in health outcomes for patients.

21 Stem Cell Therapy for Cardiac Repair: Ready for the Next Step Andrew J. Boyle, Steven P. Schulman and Joshua M. Hare Circulation. 2006;114: doi: /CIRCULATIONAHA Circulation is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX Copyright 2006 American Heart Association, Inc. All rights reserved. Print ISSN: Online ISSN: The online version of this article, along with updated information and services, is located on the World Wide Web at: Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published in Circulation can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial Office. Once the online version of the published article for which permission is being requested is located, click Request Permissions in the middle column of the Web page under Services. Further information about this process is available in the Permissions and Rights Question and Answer document. Reprints: Information about reprints can be found online at: Subscriptions: Information about subscribing to Circulation is online at:

Resident cardiac stem cells: how to find and use them

Resident cardiac stem cells: how to find and use them Resident cardiac stem cells: how to find and use them G. Hasenfuß Cardiology and Pneumology Heart Research Center Göttingen Georg-August-University Göttingen Definition: Stem cell Selfrenewal Stem cell

More information

Paracrine Mechanisms in Adult Stem Cell Signaling and Therapy

Paracrine Mechanisms in Adult Stem Cell Signaling and Therapy Paracrine Mechanisms in Adult Stem Cell Signaling and Therapy Massimiliano Gnecchi, Zhiping Zhang, Aiguo Ni, Victor J. Dzau Circulation Research 2008 Nov 21;103(11):1204-19 Introduction(1) After AMI all

More information

DOWNLOAD PDF CARDIAC REMODELING AND CELL DEATH IN HEART FAILURE

DOWNLOAD PDF CARDIAC REMODELING AND CELL DEATH IN HEART FAILURE Chapter 1 : The fibrosis-cell death axis in heart failure Remodeling may be defined as changes in the morphology, structure, and function of the heart related to alterations in loading conditions and/or

More information

Protocol. Progenitor Cell Therapy for the Treatment of Damaged Myocardium due to Ischemia

Protocol. Progenitor Cell Therapy for the Treatment of Damaged Myocardium due to Ischemia (20218) Medical Benefit Effective Date: 01/01/11 Next Review Date: 07/18 Preauthorization No Review Dates: 09/10, 07/11, 07/12, 07/13, 07/14, 07/15, 07/16, 07/17 This protocol considers this test or procedure

More information

Supplemental Table 1 Clinical trials of cell-based cardiac repair without controls or with nonrandomized study design

Supplemental Table 1 Clinical trials of cell-based cardiac repair without controls or with nonrandomized study design Cell-Based Therapy for Myocar Ischemia and Infarction: Pathophysiological Mechanisms Supplemental Table 1 Clinical trials of cell-based cardiac repair without s or with nonrandom study design Head-tohead

More information

Stem Cells. Keith Channon. Department of Cardiovascular Medicine University of Oxford John Radcliffe Hospital, Oxford

Stem Cells. Keith Channon. Department of Cardiovascular Medicine University of Oxford John Radcliffe Hospital, Oxford Stem Cells Keith Channon Department of Cardiovascular Medicine University of Oxford John Radcliffe Hospital, Oxford Adult Stem Cells Unique cells that are capable of self-renewal Have the ability to differentiate

More information

Mesenchymal Stem Cells to Repair Vascular Damage after Chemotherapy: Past, Present and Future

Mesenchymal Stem Cells to Repair Vascular Damage after Chemotherapy: Past, Present and Future Mesenchymal Stem Cells to Repair Vascular Damage after Chemotherapy: Past, Present and Future Cell Therapy 2014 Las Vegas, NV, USA Sulaiman Al-Hashmi, PhD Sultan Qaboos University Oman What are MSCs? Stem

More information

Regenerative Medicine for Cardiomyocytes

Regenerative Medicine for Cardiomyocytes Regenerative Medicine Regenerative Medicine for JMAJ 47(7): 328 332, 2004 Keiichi FUKUDA Assistant Professor, Institute for Advanced Cardiac Therapeutics, Keio University School of Medicine Abstract: Heart

More information

Progenitor Cell Therapy for the Treatment of Damaged Myocardium due to Ischemia. Original Policy Date

Progenitor Cell Therapy for the Treatment of Damaged Myocardium due to Ischemia. Original Policy Date MP 2.02.14 Progenitor Cell Therapy for the Treatment of Damaged Myocardium due to Ischemia Medical Policy Section Medicine Issue 12:2013 Original Policy Date 12:2013 Last Review Status/Date Reviewed with

More information

ENDOGENOUS CARDIAC STEM CELLS IN THE REGENERATION OF ACUTE AND CHRONIC ISCHEMIC MYOCARDIUM

ENDOGENOUS CARDIAC STEM CELLS IN THE REGENERATION OF ACUTE AND CHRONIC ISCHEMIC MYOCARDIUM ENDOGENOUS CARDIAC STEM CELLS IN THE REGENERATION OF ACUTE AND CHRONIC ISCHEMIC MYOCARDIUM Bernardo Nadal-Ginard, M.D., Ph.D. New York Medical College Angioplasty Summit 2004, Seoul 04/29/04 MYOCARDIAL

More information

Zachary I. Hodes, M.D., Ph.D., F.A.C.C.

Zachary I. Hodes, M.D., Ph.D., F.A.C.C. Zachary I. Hodes, M.D., Ph.D., F.A.C.C. Disclamer: I personally have no financial relationship with any company mentioned today. The Care Group, LLC does have a contract with Cardium to participate in

More information

Dr. Alexander Lyon Senior Lecturer and Consultant Cardiologist Clinical Lead in Cardio-Oncology Royal Brompton Hospital, London UK

Dr. Alexander Lyon Senior Lecturer and Consultant Cardiologist Clinical Lead in Cardio-Oncology Royal Brompton Hospital, London UK Advanced heart failure - devices, mechanical circulatory support and cardiac transplantation Monday 30 January 2017 Stem cell and gene therapies for heart failure Dr. Alexander Lyon Senior Lecturer and

More information

Cardiovascular Stem Cell Therapy

Cardiovascular Stem Cell Therapy Cardiovascular Stem Cell Therapy Jeffrey A Southard, MD Assistant Clinical Professor, Medicine Division of Cardiovascular Medicine May 20, 2011 Cardiovascular Disease Effects 81.1 million Americans More

More information

Medical Coverage Policy Progenitor Cell Therapy for the Treatment of Damaged Myocardium due to Ischemia

Medical Coverage Policy Progenitor Cell Therapy for the Treatment of Damaged Myocardium due to Ischemia Medical Coverage Policy Progenitor Cell Therapy for the Treatment of Damaged Myocardium due to Ischemia EFFECTIVE DATE: 02 01 2017 POLICY LAST UPDATED: 02 20 2018 OVERVIEW Progenitor cell therapy describes

More information

Mesenchymal Stem Cells

Mesenchymal Stem Cells Mesenchymal Stem Cells Science and therapeutic applications Dirk Büscher (Former VP-R&D Cellerix) GRIFOLS SA May 10 th, 2010 EMA 1 Discovery and Definition of Mesenchymal Stem Cells MSC must be plastic-adherent

More information

WIEF-AFF ROUNDTABLE Tokyo, Japan 26 May 2015

WIEF-AFF ROUNDTABLE Tokyo, Japan 26 May 2015 WIEF-AFF ROUNDTABLE 2015 Tokyo, Japan 26 May 2015 Regenerative Medicine Goal: - To restore organ +/- tissue function - For pts with serious injuries or chronic disease where the body unable to heal & restore

More information

Cardiac Gene Therapy: Beyond the Mouse. David M Kaye Heart Failure Research Group Baker IDI, Melbourne, AUSTRALIA

Cardiac Gene Therapy: Beyond the Mouse. David M Kaye Heart Failure Research Group Baker IDI, Melbourne, AUSTRALIA Cardiac Gene Therapy: Beyond the Mouse David M Kaye Heart Failure Research Group Baker IDI, Melbourne, AUSTRALIA Presenter Disclosure Information FINANCIAL DISCLOSURE: Equity: Osprey Medical Grants/Research

More information

Protocol. Progenitor Cell Therapy for the Treatment of Damaged Myocardium Due to Ischemia

Protocol. Progenitor Cell Therapy for the Treatment of Damaged Myocardium Due to Ischemia (20218) Medical Benefit Effective Date: 01/01/11 Next Review Date: 07/15 Preauthorization No Review Dates: 09/10, 07/11, 07/12, 07/13, 07/14 The following Protocol contains medical necessity criteria that

More information

Citation for published version (APA): Velde, S. V. D. (2006). Stem cell-mediated regeneration of the infarcted heart: inflammation rules?. s.n.

Citation for published version (APA): Velde, S. V. D. (2006). Stem cell-mediated regeneration of the infarcted heart: inflammation rules?. s.n. University of Groningen Stem cell-mediated regeneration of the infarcted heart Velde, Susanne van der IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to

More information

Cell implantation after myocardial infarction: a 10 years experience from the ICREC laboratory

Cell implantation after myocardial infarction: a 10 years experience from the ICREC laboratory Cell implantation after myocardial infarction: a 10 years experience from the ICREC laboratory BANFF-SCT Joint Scientific Meeting 2017 Barcelona, 29 th March Santi Roura, PhD Grup ICREC IGTP HuGTiP (Badalona)

More information

Promoting Fracture Healing Through Systemic or Local Administration of Allogeneic Mesenchymal Stem Cells

Promoting Fracture Healing Through Systemic or Local Administration of Allogeneic Mesenchymal Stem Cells Promoting Fracture Healing Through Systemic or Local Administration of Allogeneic Mesenchymal Stem Cells Gang Li Dept. of Orthopaedics and Traumatology School of Biomedical Sciences, The Chinese University

More information

Ventricular tachycardia and ischemia. Martin Jan Schalij Department of Cardiology Leiden University Medical Center

Ventricular tachycardia and ischemia. Martin Jan Schalij Department of Cardiology Leiden University Medical Center Ventricular tachycardia and ischemia Martin Jan Schalij Department of Cardiology Leiden University Medical Center Disclosure: Research grants from: Boston Scientific Medtronic Biotronik Sudden Cardiac

More information

The concept of regenerative medicine using the body s

The concept of regenerative medicine using the body s MINI-REVIEW: EXPERT OPINIONS Stem Cell Therapy in Perspective Bodo E. Strauer, MD; Ran Kornowski, MD The concept of regenerative medicine using the body s own stem cells and growth factors to repair tissues

More information

Professor Harvey White. Interventional Cardiologist Auckland

Professor Harvey White. Interventional Cardiologist Auckland Professor Harvey White Interventional Cardiologist Auckland Stem cells and the heart Harvey White Director of Coronary Care Unit and Cardiovascular Research Unit Green Lane Cardiovascular Service Auckland

More information

Heart Failure Compendium

Heart Failure Compendium Heart Failure Compendium Circulation Research Compendium on Heart Failure Research Advances in Heart Failure: A Compendium Epidemiology of Heart Failure Genetic Cardiomyopathies Causing Heart Failure Non-coding

More information

Use of Adipose-Derived Stem Cells in Regenerative Therapy. David Euhus, MD Professor of Surgery UT Southwestern Medical Center at Dallas

Use of Adipose-Derived Stem Cells in Regenerative Therapy. David Euhus, MD Professor of Surgery UT Southwestern Medical Center at Dallas Use of Adipose-Derived Stem Cells in Regenerative Therapy David Euhus, MD Professor of Surgery UT Southwestern Medical Center at Dallas Use of Adipose-Derived Stem Cells in Regenerative Therapy David Euhus,

More information

Stem Cell Therapy Concept. Pleuripotent Stromal Cells 8/8/2011. Use of Adipose-Derived Stem Cells in Regenerative Therapy

Stem Cell Therapy Concept. Pleuripotent Stromal Cells 8/8/2011. Use of Adipose-Derived Stem Cells in Regenerative Therapy Use of Adipose-Derived Stem Cells in Regenerative Therapy Use of Adipose-Derived Stem Cells in Regenerative Therapy David Euhus, MD Professor of Surgery UT Southwestern Medical Center at Dallas David Euhus,

More information

FEP Medical Policy Manual

FEP Medical Policy Manual FEP Medical Policy Manual Effective Date: October 15, 2018 Related Policies: 8.01.52 Orthopedic Applications of Stem Cell Therapy (Including Allografts and Bone Substitutes Used With Autologous Bone Marrow)

More information

International Graduate Research Programme in Cardiovascular Science

International Graduate Research Programme in Cardiovascular Science 1 International Graduate Research Programme in Cardiovascular Science This work has been supported by the European Community s Sixth Framework Programme under grant agreement n LSHM-CT-2005-01883 EUGeneHeart.

More information

Cell-based therapy for prevention and reversal of myocardial remodeling

Cell-based therapy for prevention and reversal of myocardial remodeling Cell-based therapy for prevention and reversal of myocardial remodeling Vasileios Karantalis, Wayne Balkan, Ivonne H. Schulman, Konstantinos E. Hatzistergos and Joshua M. Hare Am J Physiol Heart Circ Physiol

More information

The Jay Cohn Lecture 2012: Therapeutic regeneration of the injured heart

The Jay Cohn Lecture 2012: Therapeutic regeneration of the injured heart The Jay Cohn Lecture 2012: Therapeutic regeneration of the injured heart Eduardo Marbán, M.D., Ph.D. Director, Cedars-Sinai Heart Institute Los Angeles, CA Disclosure: founder & stockholder, Capricor Inc.

More information

Section: Medicine Last Reviewed Date: October Policy No: 100 Effective Date: January 1, 2014

Section: Medicine Last Reviewed Date: October Policy No: 100 Effective Date: January 1, 2014 Medical Policy Manual Topic: Progenitor Cell Therapy for the Treatment of Damaged Myocardium Due to Ischemia Date of Origin: August 3, 2004 Section: Medicine Last Reviewed Date: October 2013 Policy No:

More information

Cell Combination Therapy. Disclosures

Cell Combination Therapy. Disclosures Cell Combination Therapy Joshua M. Hare, M.D. Louis Lemberg Professor Senior Associate Dean Chief Science Officer Interdisciplinary Stem Cell Institute The Miller School of Medicine, University of Miami

More information

Cell therapy: enhancing the therapeutic potential of cardiac progenitors for delivery post myocardial infarction. Rita Alonaizan

Cell therapy: enhancing the therapeutic potential of cardiac progenitors for delivery post myocardial infarction. Rita Alonaizan Cell therapy: enhancing the therapeutic potential of cardiac progenitors for delivery post myocardial infarction Rita Alonaizan Department of Physiology, Anatomy & Genetics St Catherine s College Supervisor:

More information

Strategic Research Development in Stem Cell and Regenerative Medicine in HKU Professor Sum-ping Lee Dean HKU Li Ka Shing Faculty of Medicine

Strategic Research Development in Stem Cell and Regenerative Medicine in HKU Professor Sum-ping Lee Dean HKU Li Ka Shing Faculty of Medicine Strategic Research Development in Stem Cell and Regenerative Medicine in HKU Professor Sum-ping Lee Dean HKU Li Ka Shing Faculty of Medicine Adult stem cells Cells that are capable of self renewal and

More information

Indications of Coronary Angiography Dr. Shaheer K. George, M.D Faculty of Medicine, Mansoura University 2014

Indications of Coronary Angiography Dr. Shaheer K. George, M.D Faculty of Medicine, Mansoura University 2014 Indications of Coronary Angiography Dr. Shaheer K. George, M.D Faculty of Medicine, Mansoura University 2014 Indications for cardiac catheterization Before a decision to perform an invasive procedure such

More information

Devices are So Old School: The New World of Myocardial Regeneration

Devices are So Old School: The New World of Myocardial Regeneration Devices are So Old School: The New World of Myocardial Regeneration Todd K. Rosengart, M.D. Professor and Chairman DeBakey-Bard Chair of Surgery Michael E. DeBakey Department of Surgery Professor, Texas

More information

Review Article Stem Cell Therapy in Acute Myocardial Infarction: A Pot of Gold or Pandora s Box

Review Article Stem Cell Therapy in Acute Myocardial Infarction: A Pot of Gold or Pandora s Box SAGE-Hindawi Access to Research Stem Cells International Volume 2011, Article ID 536758, 20 pages doi:10.4061/2011/536758 Review Article Stem Cell Therapy in Acute Myocardial Infarction: A Pot of Gold

More information

Mesenchymal Stem Cells and Cancer: Their Interplay

Mesenchymal Stem Cells and Cancer: Their Interplay Mesenchymal Stem Cells and Cancer: Their Interplay Gang Li, MBBS, DPhil (Oxon) Stem Cell and Regeneration Program School of Biomedical Sciences Li Ka Shing Institute of Health Sciences Department of Orthopaedics

More information

Re: National Coverage Analysis (NCA) for Implantable Cardioverter Defibrillators (CAG R4)

Re: National Coverage Analysis (NCA) for Implantable Cardioverter Defibrillators (CAG R4) December 20, 2017 Ms. Tamara Syrek-Jensen Director, Coverage & Analysis Group Centers for Medicare & Medicaid Services 7500 Security Boulevard Baltimore, MD 21244 Re: National Coverage Analysis (NCA) for

More information

Tissue renewal and Repair. Nisamanee Charoenchon, PhD Department of Pathobiology, Faculty of Science

Tissue renewal and Repair. Nisamanee Charoenchon, PhD   Department of Pathobiology, Faculty of Science Tissue renewal and Repair Nisamanee Charoenchon, PhD Email: nisamanee.cha@mahidol.ac.th Department of Pathobiology, Faculty of Science Topic Objectives 1. Describe processes of tissue repair, regeneration

More information

Children's Hospital of Pittsburgh Annual Progress Report: 2011 Formula Grant

Children's Hospital of Pittsburgh Annual Progress Report: 2011 Formula Grant Children's Hospital of Pittsburgh Annual Progress Report: 2011 Formula Grant Reporting Period July 1, 2012 June 30, 2013 Formula Grant Overview The Children's Hospital of Pittsburgh received $228,401 in

More information

Remodeling of the left ventricle after myocardial infarction

Remodeling of the left ventricle after myocardial infarction Clinical Investigation and Reports Repair of Infarcted Myocardium by Autologous Intracoronary Mononuclear Bone Marrow Cell Transplantation in Humans Bodo E. Strauer, MD; Michael Brehm, MD; Tobias Zeus,

More information

NIH Public Access Author Manuscript Regen Med. Author manuscript; available in PMC 2013 March 21.

NIH Public Access Author Manuscript Regen Med. Author manuscript; available in PMC 2013 March 21. NIH Public Access Author Manuscript Published in final edited form as: Regen Med. 2012 November ; 7(6 Suppl): 17 24. Key Developments in Stem Cell Therapy in Cardiology Ivonne H. Schulman, MD 1,2 and Joshua

More information

Cardiac Cell Repair Therapy: A Clinical Perspective

Cardiac Cell Repair Therapy: A Clinical Perspective REVIEW CARDIAC CELL REPAIR THERAPY: A CLINICAL PERSPECTIVE Cardiac Cell Repair Therapy: A Clinical Perspective Bernard J. Gersh, MBChB, DPhil, FRCP; Robert D. Simari, MD, PhD; Atta Behfar, MD, PhD; Carmen

More information

What are stem cells? A stem cell can differentiate into any one of 220 different specialised cells in the body STEM CELLS

What are stem cells? A stem cell can differentiate into any one of 220 different specialised cells in the body STEM CELLS What are stem cells? Stem cells are often called MASTER CELLS, and form the foundation for your entire body as building blocks for the blood, immune system, tissue and organs. They can REPLICATE or REGENERATE

More information

Regenerative Cardiology Compendium

Regenerative Cardiology Compendium Regenerative Cardiology Compendium Clinical Studies of Cell Therapy in Cardiovascular Medicine Recent Developments and Future Directions Monisha N. Banerjee, Roberto Bolli, Joshua M. Hare Abstract: Given

More information

Journal Club WS 2012/13 Stefanie Nickl

Journal Club WS 2012/13 Stefanie Nickl Journal Club WS 2012/13 Stefanie Nickl Background Mesenchymal Stem Cells First isolation from bone marrow 30 ys ago Isolation from: spleen, heart, skeletal muscle, synovium, amniotic fluid, dental pulp,

More information

Review. Current perspective of stem cell therapies for cardiac regeneration. Yee Ki Lee 1, Chung-Wah Siu 1,2, Yang Zhang 1 & Hung-Fat Tse 1,2

Review. Current perspective of stem cell therapies for cardiac regeneration. Yee Ki Lee 1, Chung-Wah Siu 1,2, Yang Zhang 1 & Hung-Fat Tse 1,2 Review Current perspective of stem cell therapies for cardiac regeneration Despite recent advances in prevention and treatment of coronary artery disease, it remains the major cause of morbidity and mortality

More information

IHCP bulletin INDIANA HEALTH COVERAGE PROGRAMS BT JANUARY 24, 2012

IHCP bulletin INDIANA HEALTH COVERAGE PROGRAMS BT JANUARY 24, 2012 IHCP bulletin INDIANA HEALTH COVERAGE PROGRAMS BT201203 JANUARY 24, 2012 The IHCP to reimburse implantable cardioverter defibrillators separately from outpatient implantation Effective March 1, 2012, the

More information

Iliac Crest: The Gold Standard

Iliac Crest: The Gold Standard Iliac Crest: The Gold Standard Iliac crest is often considered the gold standard for harvesting. The iliac crest contains bone marrow which is a rich source of regenerative cells, including: Endothelial

More information

Recovering Hearts. Saving Lives.

Recovering Hearts. Saving Lives. Recovering Hearts. Saving Lives ṬM The Door to Unload (DTU) STEMI Safety & Feasibility Pilot Trial November 218 Recovering Hearts. Saving Lives. LEGAL DISCLAIMERS This presentation includes select slides

More information

Biomarkers for Personalized Medicine and Regenerative Therapy

Biomarkers for Personalized Medicine and Regenerative Therapy Biomarkers for Personalized Medicine and Regenerative Therapy Bettina Heidecker, MD University of Miami Division of Cardiology Interdisciplinary Stem Cell Institute Biomarkers in Heart Failure Growing

More information

Multimodality Imaging in Cardiac Stem Cell Research

Multimodality Imaging in Cardiac Stem Cell Research Multimodality Imaging in Cardiac Stem Cell Research IL SUK SOHN, MD, PhD Department of Cardiology Kyung Hee University Hospital at Gangdong Kyung Hee University School of Medicine, Seoul, Korea Stem Cell

More information

Stem cell therapy aimed at restoring organ function,

Stem cell therapy aimed at restoring organ function, The : Insights and Lessons From Clinical Trials Kartik S. Telukuntla, BS; Viky Y. Suncion, MD; Ivonne H. Schulman, MD; Joshua M. Hare, MD Stem cell therapy aimed at restoring organ function, notably myocardial

More information

Reperfusion Injury: How Can We Reduce It?

Reperfusion Injury: How Can We Reduce It? MI/CAD: Practical Question in Management of AMI Patients Reperfusion Injury: How Can We Reduce It? Hyun-Jai Cho, M.D., Ph.D Cardiovascular Center & Department of Internal Medicine Seoul National University

More information

Stem cell therapy for chronic ischaemic heart disease and congestive heart failure (Review)

Stem cell therapy for chronic ischaemic heart disease and congestive heart failure (Review) Stem cell therapy for chronic ischaemic heart disease and congestive heart failure (Review) Fisher SA, Brunskill SJ, Doree C, Mathur A, Taggart DP, Martin-Rendon E This is a reprint of a Cochrane review,

More information

ALTERNATIVE APPROACH FOR END-STAGED ISCHEMIC CARDIOMYOPATHY WITH LV ANEURYSM COMBINING SURGICAL AND CELL THERAPY THESSALONIKI, 2018

ALTERNATIVE APPROACH FOR END-STAGED ISCHEMIC CARDIOMYOPATHY WITH LV ANEURYSM COMBINING SURGICAL AND CELL THERAPY THESSALONIKI, 2018 ALTERNATIVE APPROACH FOR END-STAGED ISCHEMIC CARDIOMYOPATHY WITH LV ANEURYSM COMBINING SURGICAL AND CELL THERAPY THESSALONIKI, 2018 Kostas Katsavrias Sotirios N. Prapas A Cardiac Surgery Dpt Henry Dunant

More information

Ischemic heart disease

Ischemic heart disease Ischemic heart disease Introduction In > 90% of cases: the cause is: reduced coronary blood flow secondary to: obstructive atherosclerotic vascular disease so most of the time it is called: coronary artery

More information

Progenitor Cell Therapy for the Treatment of Damaged Myocardium Due to Ischemia

Progenitor Cell Therapy for the Treatment of Damaged Myocardium Due to Ischemia Last Review Status/Date: September 2015 Page: 1 of 17 Damaged Myocardium Due to Ischemia Description Progenitor cell therapy describes the use of multipotent cells of various cell lineages (autologous

More information

In Vivo Animal Models of Heart Disease. Why Animal Models of Disease? Timothy A Hacker, PhD Department of Medicine University of Wisconsin-Madison

In Vivo Animal Models of Heart Disease. Why Animal Models of Disease? Timothy A Hacker, PhD Department of Medicine University of Wisconsin-Madison In Vivo Animal Models of Heart Disease Timothy A Hacker, PhD Department of Medicine University of Wisconsin-Madison Why Animal Models of Disease? Heart Failure (HF) Leading cause of morbidity and mortality

More information

STEM CELL RESEARCH: MEDICAL PROGRESS WITH RESPONSIBILITY

STEM CELL RESEARCH: MEDICAL PROGRESS WITH RESPONSIBILITY STEM CELL RESEARCH: MEDICAL PROGRESS WITH RESPONSIBILITY A REPORT FROM THE CHIEF MEDICAL OFFICER S EXPERT GROUP REVIEWING THE POTENTIAL OF DEVELOPMENTS IN STEM CELL RESEARCH AND CELL NUCLEAR REPLACEMENT

More information

c-kit Stem Cell Therapy of Ischemic Cardiomyopathy

c-kit Stem Cell Therapy of Ischemic Cardiomyopathy American Heart Association Scientific Sessions American Heart Association Scientific Sessions c-kit Stem Cell Therapy of Ischemic Cardiomyopathy Roberto Bolli, M.D. Institute of Molecular Cardiology University

More information

Stem Cell Therapy for Ischemic Heart Disease : A Status Report

Stem Cell Therapy for Ischemic Heart Disease : A Status Report Stem Cell Therapy for Ischemic Heart Disease : A Status Report Do Sun Lim, M.D. Department of Internal Medicine Korea University College of Medicine Anam Hospital E mail : dslmd@kumc.or.kr Abstract Myocardial

More information

Myocardial infarction

Myocardial infarction NEW CARDIAC MARKERS AND CARDIAC REGENERATION Päivi Lakkisto, MD, PhD Specialist in Clinical Chemistry Clinical lecturer University of Helsinki and HUSLAB Minerva Institute for Medical Research Myocardial

More information

Radical treatment for leukemia under way 17 January 2011, By Fumihiko Ishikawa

Radical treatment for leukemia under way 17 January 2011, By Fumihiko Ishikawa Radical treatment for leukemia under way 17 January 2011, By Fumihiko Ishikawa Figure 1: Major blood cells that differentiate from hematopoietic stem cells. Hematopoietic stem cells differentiate into

More information

National Imaging Associates, Inc. Clinical guidelines CARDIAC CATHETERIZATION -LEFT HEART CATHETERIZATION. Original Date: October 2015 Page 1 of 5

National Imaging Associates, Inc. Clinical guidelines CARDIAC CATHETERIZATION -LEFT HEART CATHETERIZATION. Original Date: October 2015 Page 1 of 5 National Imaging Associates, Inc. Clinical guidelines CARDIAC CATHETERIZATION -LEFT HEART CATHETERIZATION CPT Codes: 93451, 93452, 93453, 93454, 93455, 93456, 93457, 93458, 93459, 93460, 93461 LCD ID Number:

More information

Review Article Therapeutic Application of Cardiac Stem Cells and Other Cell Types

Review Article Therapeutic Application of Cardiac Stem Cells and Other Cell Types BioMed Research International Volume 2013, Article ID 736815, 6 pages http://dx.doi.org/10.1155/2013/736815 Review Article Therapeutic Application of Cardiac Stem Cells and Other Cell Types Emiko Hayashi

More information

REVIEW Hematopoietic, vascular and cardiac fates of bone marrow-derived stem cells

REVIEW Hematopoietic, vascular and cardiac fates of bone marrow-derived stem cells (2002) 9, 648 652 2002 Nature Publishing Group All rights reserved 0969-7128/02 $25.00 www.nature.com/gt REVIEW Hematopoietic, vascular and cardiac fates of bone marrow-derived stem cells KK Hirschi 1

More information

Pathology of Cardiovascular Interventions. Body and Disease 2011

Pathology of Cardiovascular Interventions. Body and Disease 2011 Pathology of Cardiovascular Interventions Body and Disease 2011 Coronary Artery Atherosclerosis Intervention Goals: Acute Coronary Syndromes: Treat plaque rupture and thrombosis Significant Disease: Prevent

More information

Myocardial Regeneration and Stem Cell Repair

Myocardial Regeneration and Stem Cell Repair Myocardial Regeneration and Stem Cell Repair Annarosa Leri, MD, Jan Kajstura, MD, Piero Anversa, MD, and William H. Frishman, MD Abstract: Recent evidence would suggest that the heart is not a terminally

More information

A New Treatment Paradigm Outside-In

A New Treatment Paradigm Outside-In A New Treatment Paradigm Outside-In 2 2013 Mercator MedSystems OneMedForum Jan 8, 2013 Outside-In Response: Signal Amplification Signal convection Signal amplification Modified from Michel 2007, ATVB 3

More information

Review Article Functional Multipotency of Stem Cells: What Do We Need from Them in the Heart?

Review Article Functional Multipotency of Stem Cells: What Do We Need from Them in the Heart? Stem Cells International Volume 2012, Article ID 817364, 12 pages doi:10.1155/2012/817364 Review Article Functional Multipotency of Stem Cells: What Do We Need from Them in the Heart? Pablo DíezVillanueva,

More information

Research progress of adult stem cells and clinical applications

Research progress of adult stem cells and clinical applications 18 4 2006 8 Chinese Bulletin of Life Sciences Vol. 18, No. 4 Aug., 2006 1004-0374(2006)04-0328-05 100850 Q831 A Research progress of adult stem cells and clinical applications XI Jia-Fei, WANG Yun-Fang,

More information

Atrial fibrillation (AF) is a disorder seen

Atrial fibrillation (AF) is a disorder seen This Just In... An Update on Arrhythmia What do recent studies reveal about arrhythmia? In this article, the authors provide an update on atrial fibrillation and ventricular arrhythmia. Beth L. Abramson,

More information

Automatic External Defibrillators

Automatic External Defibrillators Last Review Date: April 21, 2017 Number: MG.MM.DM.10dC3v4 Medical Guideline Disclaimer Property of EmblemHealth. All rights reserved. The treating physician or primary care provider must submit to EmblemHealth

More information

Progress in clinical applications of PSCs in The bench to bedside pathway

Progress in clinical applications of PSCs in The bench to bedside pathway Progress in clinical applications of PSCs in 2017 The bench to bedside pathway Trounson A. et al. Nature Reviews Molecular Cell Biology 17, 194 200 (2016) doi:10.1038/nrm.2016.10 Knoepfler PS Adv Drug

More information

1. Cardiomyocytes and nonmyocyte. 2. Extracellular Matrix 3. Vessels שאלה 1. Pathobiology of Heart Failure Molecular and Cellular Mechanism

1. Cardiomyocytes and nonmyocyte. 2. Extracellular Matrix 3. Vessels שאלה 1. Pathobiology of Heart Failure Molecular and Cellular Mechanism Pathobiology of Heart Failure Molecular and Cellular Mechanism Jonathan Leor Neufeld Cardiac Research Institute Tel-Aviv University Sheba Medical Center, Tel-Hashomer שאלה 1 התא הנפוץ ביותר (75%~) בלב

More information

Prevention of sudden cardiac death: With an emphasis on sudden cardiac death from ventricular arrhythmias

Prevention of sudden cardiac death: With an emphasis on sudden cardiac death from ventricular arrhythmias Prevention of sudden cardiac death: With an emphasis on sudden cardiac death from ventricular arrhythmias The Toronto ACS Summit Toronto, March 1, 2014 Andrew C.T. Ha, MD, MSc, FRCPC Cardiac Electrophysiology

More information

E. Cervio, P. Danieli, C. Ciuffreda, F. Pisano, M. Roccio, M. Gnecchi. The authors have no financial disclosures to declare

E. Cervio, P. Danieli, C. Ciuffreda, F. Pisano, M. Roccio, M. Gnecchi. The authors have no financial disclosures to declare 16 th ISCT Annual Meeting SOLUBLE FACTORS RELEASED BY HUMAN MESENCHYMAL STEM CELLS OF FETAL ORIGIN LEAD TO CARDIOMYOCYTE PROTECTION THROUGH THE INHIBITION OF PRO-APOPTOTIC SIGNALING E. Cervio, P. Danieli,

More information

Cardiac Drugs: Chapter 9 Worksheet Cardiac Agents. 1. drugs affect the rate of the heart and can either increase its rate or decrease its rate.

Cardiac Drugs: Chapter 9 Worksheet Cardiac Agents. 1. drugs affect the rate of the heart and can either increase its rate or decrease its rate. Complete the following. 1. drugs affect the rate of the heart and can either increase its rate or decrease its rate. 2. drugs affect the force of contraction and can be either positive or negative. 3.

More information

Most cells in the human body have an assigned purpose. They are liver cells, fat cells, bone cells,

Most cells in the human body have an assigned purpose. They are liver cells, fat cells, bone cells, What is a Stem Cell? Most cells in the human body have an assigned purpose. They are liver cells, fat cells, bone cells, and so on. These cells can replicate more of their own kind of cell, but they cannot

More information

Conflict of Interest Slide

Conflict of Interest Slide Comparison of six- month clinical outcomes, event free survival rates of patients undergoing enhanced external counterpulsation (EECP) for coronary artery disease in the United States and Europe Ozlem

More information

Title: Automatic External Defibrillators Division: Medical Management Department: Utilization Management

Title: Automatic External Defibrillators Division: Medical Management Department: Utilization Management Retired Date: Page 1 of 7 1. POLICY DESCRIPTION: Automatic External Defibrillators 2. RESPONSIBLE PARTIES: Medical Management Administration, Utilization Management, Integrated Care Management, Pharmacy,

More information

Cardiac evaluation for the noncardiac. Nathaen Weitzel MD University of Colorado Denver Dept of Anesthesiology

Cardiac evaluation for the noncardiac. Nathaen Weitzel MD University of Colorado Denver Dept of Anesthesiology Cardiac evaluation for the noncardiac patient Nathaen Weitzel MD University of Colorado Denver Dept of Anesthesiology Objectives! Review ACC / AHA guidelines as updated for 2009! Discuss new recommendations

More information

Leading the Way in Cardiovascular Regenerative Medicine

Leading the Way in Cardiovascular Regenerative Medicine Slide 1 Leading the Way in Cardiovascular Regenerative Medicine Leading the Way in Cardiovascular Regenerative Medicine This slide set presents the current work in cell therapy in treating cardiovascular

More information

Hormonal Alterations in Heart Failure: Anabolic Impairment in Chronic Heart Failure Diagnostic, Prognostic and Therapeutic Issues

Hormonal Alterations in Heart Failure: Anabolic Impairment in Chronic Heart Failure Diagnostic, Prognostic and Therapeutic Issues Hormonal Alterations in Heart Failure: Anabolic Impairment in Chronic Heart Failure Diagnostic, Prognostic and Therapeutic Issues Michele Arcopinto Antonio Cittadini Department of Translational Medical

More information

Review Article Autologous Stem Cell Transplantation for Regeneration of Infarcted Myocardium: Clinical Trials

Review Article Autologous Stem Cell Transplantation for Regeneration of Infarcted Myocardium: Clinical Trials Hellenic J Cardiol 2008; 49: 163-168 Review Article Autologous Stem Cell Transplantation for Regeneration of Infarcted Myocardium: Clinical Trials GEORGIOS PAXINOS 1, DEMOSTHENES KATRITSIS 2 1 Cardiology

More information

Myoblast Transplantation. Advanced Angioplasty London, May 18, 2003

Myoblast Transplantation. Advanced Angioplasty London, May 18, 2003 Myoblast Transplantation Advanced Angioplasty London, May 18, 2003 Heart Failure Epidemiology -- High incidence (~ 500,000 per year - in the U.S.) -- High mortality (40% at 1 year in - Class III-IV patients)

More information

ΘΕΡΑΠΕΥΤΙΚΕΣ ΕΞΕΛΙΞΕΙΣ Ιωάννης Α. Παρασκευαίδης

ΘΕΡΑΠΕΥΤΙΚΕΣ ΕΞΕΛΙΞΕΙΣ Ιωάννης Α. Παρασκευαίδης ΘΕΡΑΠΕΥΤΙΚΕΣ ΕΞΕΛΙΞΕΙΣ 2017 Ιωάννης Α. Παρασκευαίδης Myocardial Recovery in Pts With Systolic HF and Autoantibodies Against b1-adrenergic Receptors The IMAC-2 Study Y. Nagatomo et al. J Am Coll Cardiol

More information

OPTIMISING CELL THERAPY FOR TREATING HEART FAILURE

OPTIMISING CELL THERAPY FOR TREATING HEART FAILURE OPTIMISING CELL THERAPY FOR TREATING HEART FAILURE by Satsuki Fukushima A thesis submitted to Imperial College London for the degree of Doctor of Philosophy Supervisors: Professor Sir Magdi H Yacoub Professor

More information

Stem Cells and Sport Medicine

Stem Cells and Sport Medicine Stem Cells and Sport Medicine Rehal Abbas Bhojani, MD CAQSM Memorial Hermann Medical Group 2014 Sports Medicine Symposium of the Americas Stem cell biology Overview Potential applications of stem cells

More information

Heart failure (HF) is a complex clinical syndrome that results in the. impairment of the heart s ability to fill or to pump out blood.

Heart failure (HF) is a complex clinical syndrome that results in the. impairment of the heart s ability to fill or to pump out blood. Introduction: Heart failure (HF) is a complex clinical syndrome that results in the impairment of the heart s ability to fill or to pump out blood. As of 2013, an estimated 5.8 million people in the United

More information

Therapeutic Potential of Human Umbilical Cord Derived Stem Cells in a Rat Myocardial Infarction Model

Therapeutic Potential of Human Umbilical Cord Derived Stem Cells in a Rat Myocardial Infarction Model Therapeutic Potential of Human Umbilical Cord Derived Stem Cells in a Rat Myocardial Infarction Model Kai Hong Wu, MD, PhD,* Bin Zhou, PhD,* Cun Tao Yu, MD, Bin Cui, MD, Shi Hong Lu, BS, Zhong Chao Han,

More information

Journal of the American College of Cardiology Vol. 35, No. 5, by the American College of Cardiology ISSN /00/$20.

Journal of the American College of Cardiology Vol. 35, No. 5, by the American College of Cardiology ISSN /00/$20. Journal of the American College of Cardiology Vol. 35, No. 5, 2000 2000 by the American College of Cardiology ISSN 0735-1097/00/$20.00 Published by Elsevier Science Inc. PII S0735-1097(00)00546-5 CLINICAL

More information

Chapter 7 Conclusions

Chapter 7 Conclusions VII-1 Chapter 7 Conclusions VII-2 The development of cell-based therapies ranging from well-established practices such as bone marrow transplant to next-generation strategies such as adoptive T-cell therapy

More information

Autologous Bone Marrow Mononuclear Cells Transplant in Patients With Critical Leg Ischemia: Preliminary Clinical Results

Autologous Bone Marrow Mononuclear Cells Transplant in Patients With Critical Leg Ischemia: Preliminary Clinical Results ARTIcle Autologous Bone Marrow Mononuclear Cells Transplant in Patients With Critical Leg Ischemia: Preliminary Clinical Results Min Li, 1 Hua Zhou, 2 Xing Jin, 2 Mo Wang, 2 Shiyi Zhang, 2 Lei Xu 2 Abstract

More information

OUTCOME OF THROMBOLYTIC AND NON- THROMBOLYTIC THERAPY IN ACUTE MYOCARDIAL INFARCTION

OUTCOME OF THROMBOLYTIC AND NON- THROMBOLYTIC THERAPY IN ACUTE MYOCARDIAL INFARCTION OUTCOME OF THROMBOLYTIC AND NON- THROMBOLYTIC THERAPY IN ACUTE MYOCARDIAL INFARCTION FEROZ MEMON*, LIAQUAT CHEEMA**, NAND LAL RATHI***, RAJ KUMAR***, NAZIR AHMED MEMON**** OBJECTIVE: To compare morbidity,

More information

Optimization of Generx (Ad5FGF-4) Clinical Trial Design for Refractory Angina: Interim Results of the Phase 3 ASPIRE Trial

Optimization of Generx (Ad5FGF-4) Clinical Trial Design for Refractory Angina: Interim Results of the Phase 3 ASPIRE Trial Optimization of Generx (Ad5FGF-4) Clinical Trial Design for Refractory Angina: Interim Results of the Phase 3 ASPIRE Trial Gabor M Rubanyi, MD. PhD. Angionetics Inc., San Diego, CA ASGCT, New Orleans,

More information