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

Size: px
Start display at page:

Download "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"

Transcription

1 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 worldwide. There is an unmet need for treatment of ischemic cardiomyopathy caused by postmyocardial infarction, left ventricular remodeling or chronic myocardial ischemia. As a result, there is tremendous interest in developing stem cell therapies for prevention and treatment of ischemic cardiomyopathy. Although the mechanism remains unclear, the majority of preclinical studies demonstrate that different sources of cell-based therapy reduce the infarct size, and improve myocardial perfusion and contractile function. Recent results from randomized controlled clinical trials suggest that intracoronary or intramyocardial injection of bone marrow-derived cells may be a safe and feasible strategy for treatment of acute myocardial infarction as well as chronic myocardial ischemia. Furthermore, these studies have also provided initial evidence of clinical efficacy with modest, but significant improvement in left ventricular ejection fraction and functional status. However, most of these studies are limited by relatively small sample sizes and a short duration of follow-up, and the clinical efficacy of stem cell therapy in coronary artery disease needs to be confirmed by ongoing major clinical trials. In addition, further studies are also required to refine the optimal timing, cell types and mode of delivery, and to develop strategies to overcome the potential hurdles related to cell-based therapy. Keywords: acute myocardial infarction bone marrow-derived cells cardiac progenitor cells embryonic stem cells induced pluripotent stem cells myocardial ischemia skeletal myoblasts Coronary artery disease (CAD) remains a major cause of morbidity and mortality worldwide [1]. Despite recent advances in reperfusion therapy for acute myocardial infarction (MI) and pharma cotherapy for post-mi left ventricular (LV) remodeling, the incidence and mortality of post-mi heart failure is increasing [2]. As a result, there is a major unmet need for treatment of post-mi heart failure caused by progressive LV remodeling after initial insult to the myocardium. Therefore, there is tremendous interest in developing novel stem cell therapies for prevention and treatment of post-mi LV remodeling and dysfunction. Recent studies demonstrate that the heart is not a terminally differentiated organ but contains a pool of progenitor cells capable of self-renewal [3]. However, in a typical MI involving 25% of the LV, there would be approximately a cardio myocyte (CM) loss, for which this intrinsic repair mechanism will not be able to restore the damaged myocardial contractile function [4,5]. The promise of stem cell therapies is to mobilize and/or introduce exogenous stem cells to repair this deficit of CMs. This article reviews the current state, potential hurdles and future perspective of stem cell therapies for cardiac regeneration. Potential mechanisms for stem cell therapies The concept of cardiac regeneration relies on the belief that exogenous cells or mobilized endogenous cells can transdifferentiate into mature CMs after transplanting into the microenvironment of the damaged myocardium. Furthermore, they should also integrate both electrically and mechanically with host CMs, thereby contributing to improvement of cardiac function [6]. Experimental studies show that several types of stem cells can improve LV function in animal models of MI or myocardial ischemia [7,8]. The potential mechanisms of stem cell therapy for cardiac regeneration are summarized in Figure 1. Clinical studies confirm that new endogenous or exogenous stem cells can incorporate into host myocardium via transdifferentiation and/or cell fusion [9]. However, emerging evidence suggests that cellular mediated paracrine effects are likely to be the major mechanism that contributes to the improvement in LV function [10], as there is lack of significant CM transdifferentiation [11,12] and long-term survival of transplanted cells [13]. Experimental studies have shown that the transplanted cells can secrete proangiogenic cytokines Yee Ki Lee 1, Chung-Wah Siu 1,2, Yang Zhang 1 & Hung-Fat Tse 1,2 1 Cardiology Division, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong SAR, China 2 Research Center of Heart, Brain, Hormone & Healthy Aging, The University of Hong Kong, Hong Kong SAR, China Author for correspondence: Tel.: Fax: hftse@hkucc.hku.hk /THY Future Medicine Ltd Therapy (2011) 8(1), ISSN

2 Review Lee, Siu, Zhang & Tse Stem cell therapies Transdifferentiation Cell fusion Paracrine effects Cardiomyogenesis Neovascularization Recruitment of stem cells Anti-apoptosis Matrix remodeling Cardiac regeneration Figure 1. Potential mechanisms of stem cell therapies for cardiac regeneration. to enhance neovascularization [14]. Furthermore, the paracrine factors secreted by the transplanted cells exert anti-apoptotic effects, alter the restoration of extracellular matrix and recruit endogenous stem cells [10,14]. Indeed, recent studies demonstrate that direct injection of bone marrow (BM) cell extract can provide comparable benefits as intact cell therapy in limiting LV remodeling and improve the LV function after MI [15]. Cell types for cardiac regeneration A variety of stem or progenitor cells, including skeletal myoblast, BM-derived cells, placental/cord blood-derived cells, adipose tissue-derived cells, resident cardiac progenitor cells, embryonic stem cells (ESCs) and, recently, induced pluripotent stem (ips) cells, have been investigated for cardiac regeneration (Figure 2). Table 1 summarizes the advantages and disadvantages of various cell types used for cardiac regeneration. Skeletal myoblasts Skeletal myoblasts, also known as muscle satellite cells, are skeletal muscle precursor cells found between the basal lamina and sarcolemma of skeletal muscle. They can be isolated from skeletal muscle biopsies and expanded ex vivo to the quantities sufficient for auto logous transplantation [16]. Therefore, skeletal myoblasts appear to be an attractive candidate for myocardial regeneration. Preclinical experiments have demonstrated that transplanted skeletal myoblasts successfully engraft, differentiate into myotubules and improve post-mi cardiac function in animal models of MI [17 23]. These initial encouraging results paved the way for the first Phase I human trial in 2000 [24] followed by a number of pilot clinical trials [25 29]. However, the enthusiasm for using skeletal myoblasts for cardiac regeneration has faltered with concerns regarding an increased incidence of ventricular tachyarrhythmia after transplantation. The lack of connexin 43 expression in skeletal myo blasts after transplantation leads to the failure of electrical integration with the host myo cardium and may contribute to an increased risk of proarrhythmias [30,31]. BM-derived cells Bone marrow-derived cells are a heterogeneous cellular population consisting of approximately 98% differentiated cells, including hematopoietic cells, vascular cells, adipocytes, osteoblasts and osteoclasts, and approximately 2% stem or progenitor cells. The undifferentiated stem or progenitor cells in the BM are a mixed population of hematopoietic stem cells, endothelial progenitor cells (EPCs) and mesenchymal stem cells (MSCs). BM-derived stem or progenitor cells have attracted attention owing to 70 Therapy (2011) 8(1)

3 Current perspective of stem cell therapies for cardiac regeneration Review Skeletal myoblast Bone marrow cells Adipose cells Placental/cord blood cells Mesenchymal stem cells Embryonic stem cells Cardiac progenitor cells Induced pluripotent stem cells Figure 2. Different types of stem/progenitor cells for cardiac regeneration. their potential to transdifferentiate into various cell types in the body, including neurons, hepatocytes, skeletal muscle and CM. Furthermore, they can be readily harvested and expanded ex vivo for autologous transplantation without any immune rejection. Prior experimental studies suggest that transplantation of BM-derived cells into the border zone of acute MI results in in vivo myogenesis and neoangiogenesis by the transplanted cell, which subsequently contributes to an improvement in cardiac function [32]. However, recent studies using genetic rather than immunofluorescence techniques have demonstrated that transdifferentiation of BM cells into CMs is only a rare event [11,12,33]. Therefore, paracrine effects of BM cells have been postulated as the major mechanism for the improvement of cardiac function after transplantation [34]. In general, BM-derived cells can be broadly divided into two main groups according to the expression of the surface antigen CD34. CD34 + stem/progenitor cells are precursors of blood cells and endothelial cells in BM. Among Table 1. Comparison of the advantages and disadvantages of different types of cells for engraftment. Cell source Advantages Disadvantages Ref. Skeletal myoblasts Easy to obtain by isolation from skeletal muscle biopsies Using ex vivo expansion it is possible to produce sufficient quantities for autologous transplantation BM-derived cells Using ex vivo expansion it is possible to produce sufficient quantities for autologous transplantation without immune rejection Paracrine effects of BM EPCs and MSCs improve cardiac function ESCs Able to undergo self-renewal in undifferentiated state without karyotypic alteration ips cells Patient specific and feasible for autologous transplantation A lack of connexin 43 expression after in vitro differentiation results in failure of electrical integration with the host myocardium Transdifferentiation into CMs may only be a rare event Efficiency of spontaneous in vitro cardiac differentiation is very low (<1%) Potential risk of immune rejection and teratoma formation Unethical with sacrifice of early embryo Low yield of cardiac differentiation Requires the use of viral vectors, which may lead to oncogenesis and genome abnormality BM: Bone marrow; CM: Cardiomyocyte; EPC: Endothelial progenitor cell; ESC: Embryonic stem cell; ips: Induced pluripotent stem; MSC: Mesenchymal stem cell. [27,101] [10 12,14,33] [25,70,102] [28,70] 71

4 Review Lee, Siu, Zhang & Tse them, EPCs are characterized by their coexpression of CD133 and/or kinase insert domain receptor [35]. Experimental studies suggest that BM-derived EPCs can be mobilized into systematic circulation, and contribute to postnatal neovascularization [36,37] and endothelial repair [38]. Therefore, the use of EPCs as selected by CD34 + or CD133 + may have the greatest potential for enhancement of neovascularization. By contrast, CD34 - cells are less well defined. They consist of MSC and multi potent adult progenitor cells. BM-derived MSCs are precursors of stromal cells in BM, which possess the capacity of multilineage differentiation, including CMs [39,40]. Despite the fact that a panel of markers including CD29, CD90, CD44 and CD73 (SH3 or SH4) has been described, there are no specific markers for their selection. Experimental studies have also shown that MSC transplantation increased myo cardial perfusion, reduced apoptosis and infarct size, and improved LV function [41]. Compared with other BM-derived stem cells, MSCs appear to have immunoprivilege and may have the potential to be used for allogenic transplantation [42]. Although the transplanted MSCs in host myocardium have the potential to transdifferentiate into CMs, in vitro differentiation of MSCs into functional CMs after transplantation into patients with MI is rarely observed. Multilineage differentiation capacity has also been described in a subset of multi potent adult progenitor cells, but specific markers for their identification are lacking [43,44]. The potential of differentiation of MSCs is rather limited and selective, and clinical use of this cell type remains to be discussed. The encouraging results from these preclinical studies using different types of BM-derived cells have prompted a series of clinical trials investigating the potential therapeutic use of BM-derived cells in CAD patients. Indeed, the majority of recently completed or ongoing clinical trials in CAD patients are based on the use of BM-derived cells. Adipose tissue-derived cells Adipose tissue represents another abundant and readily available source of adult stem cells for cardiac regeneration. In addition to adipocytes, adipose tissue also contains MSCs and EPCs. Similar to BM-derived MSCs, adipose tissuederived MSCs have also been shown to differentiate into CM [45] and endothelial lineages [46]. Experimental studies also demonstrate that adipose tissue-derived MSCs can engraft and acquire cardiac phenotypes in post-mi myocardium and improve cardiac function [47,48]. However, their therapeutic potential as compared with other sources of MSCs remains unclear. Placental or cord blood-derived cells Placental or cord blood are also a rich source of stem cells, including hematopoietic stem cells, MSCs and unrestricted somatic stem cells. In general, they have a higher proliferative potential compared with similar cell types that are derived from adult BM. Recently, a nanofiberbased ex vivo human umbilical cord blood stem cell expansion technology made it feasible to obtain an appropriate number of therapeutic stem cells. Additionally, proangiogenic growth factor overexpression in progenitor cells can potentially improve autologous or allogeneic stem cell therapy for ischemic diseases [49]. Another study suggested that human umbilical cord blood cells specifically migrate to infarcted rather than normal myocardium, where engraftment and neoangiogenesis occur, and beneficially influence remodeling processes [50]. Furthermore, these cells can be easily obtained and cryopreserved after birth for future autologous transplantation. However, there are very limited data on the use of placental or cord blood-derived cells for cardiac regeneration [51]. Resident cardiac stem cells The presence of resident cardiac stem cells in the adult mammalian heart has been increasingly recognized [3]. Several investigators confirmed the existence of resident cardiac stem cells, which are self-renewing, clonogenic, and can differentiate into CMs, smooth muscle and endothelial cells [52]. These cells can be identified by the expression of c kit and sca 1, as well as their phenotype and ability to differentiate in cell culture [53]. Resident cardiac stem cells have been successfully isolated from surgical and endomyocardial biopsy in a clinical setting, and expanded ex vivo for autologous trans plantation. In experimental models of MI, transplantation or mobili zation of resident cardiac stem cells can engraft and acquire cardiac and vascular phenotypes in infarcted myocardium, and improve cardiac function [54]. Recently, a method has been presented for the isolation of adult human stem cells from endomyocardial biopsy specimens. Cardiosphere-derived cells are cardiogenic in vitro, and have promoted cardiac regeneration in a murine model [55]. However, by eliminating the presence of myogenic tissue 72 Therapy (2011) 8(1)

5 Current perspective of stem cell therapies for cardiac regeneration Review in the cardac biopsy, Andersen et al. argued that cardiospheres cultured from neonatal and adult rats/mice are indeed without cardiomyogenic potential [56], suggesting a murine model may be unsuitable as a source of cardiac stem cells. Embryonic stem cells Compared with adult stem cells, ESCs possess two unique properties. First, ESCs can be cultured indefinitely (i.e., they self-renew) in an undifferentiated state without karyotypic alteration. Second, ESCs maintain the potential to differentiate into all cell types of the body (pluripotency) [57,58]. In fact, genuine CMs with cardiac-specific structural and functional properties can be consistently differentiated from ESCs using various methods [59]. Furthermore, various subtypes of CMs including pacemaker, atrial and ventricular CMs have been identified [60]. As a result, ESCs have been regarded as a potentially unlimited ex vivo cell source for cell-based therapies. Numerous preclinical studies involving transplantation of undifferentiated ESCs or their cardiac derivatives into infarcted myocardium have been reported [61]. Consistently, transplantation of undifferentiated ESCs or ESC-derived CMs improved LV function post-mi (Table 2). However, there are many obstacles to the clinical use of ESCs for cardiac regeneration. First, the efficiency of in vitro cardiac differentiation from ESCs remains low (typically <1%), thus making it very difficult to achieve the number of CMs needed for therapeutic application. Second, cardiac differentiation from ESCs results in a heterogeneous population of CMs (pacemakers, atrial and ventricular cells), which could give rise to arrhythmia caused by a mismatch between the transplanted cells and the host CMs [62]. Third, CMs derived from ESCs are in fact structurally and functionally immature relative to their adult counterparts. The immature electrophysiological properties of ESC-derived CMs may contribute to proarrhythmia [63]. Fourth, the potential risks of tumor formation and immunogenicity of ESCs and ESC-derived cells also need to be addressed [64,65]. Finally, the most important disadvantages of ESC usage are the controversial ethical issues, as ESCs are isolated from the blastocyst stage of the embryo. ips cells In the last few years, several laboratories have successfully generated ips cells from mouse and human fibroblasts by exogenous introduction of a combination of transcription factors (Oct3/4, Sox2, Klf4 and c Myc), which are expressed at high levels in ESCs [66 68]. These ips cells, generated from the reprogramming process, exhibit essential features of ESCs including morphology, surface markers, gene expression profiles and telomerase activity. More importantly, ips cells also acquire two essential criteria that need to be met for stemness : the capacity to selfrenew and to maintain pluripotency as ESCs. Indeed, CMs can also be successfully differentiated from ips cells and showed typical features that resemble ESC-derived CMs [69]. As ips cells are generated from adult somatic cells, patient-specific stem cells can be obtained for autologous transplantation to avoid the ethical and immunological issues related to ESCs. However, ips cells still share some of the problems associated with ESCs, including the low yield, as well as the immature and heterogenous phenotypes with cardiac differentiation. Furthermore, the use of viral vectors to create ips cells may lead to insertional mutagenesis causing uncontrolled cellular proliferation, oncogenesis or abnormal development [70]. Expression of oncogenic factors should be carefully monitored in the ips cells to avoid teratoma formation upon ips cell therapy. However, such problems can be solved by nonviral administration of the four pluripotencyinducible factors, which include the use of nonintegrating genetic elements [71,72], excision of genetic elements after ips formation [73 76] or recombinant transduction-tagged proteins [77]. Mode of cell delivery Successful cell therapy depends on engraftment and survival of the transplanted cell. Furthermore, systemic administration of stem cells such as intravenous administration relies on nonorgan-specific cell delivery and may increase the risk of adverse systemic side effects. The homing and engraftment of stem or progenitor cells after administration relies on the method of cell delivery, the characteristics of transplanted cells and the host environ ment [34]. One of the practical approaches to select the mode of delivery is based on the physiologic or disease status of the host myocardium (Figure 3). In the setting of acute MI, the upregulation of local homing signals, such as stem cell factor, stromal cell-derived factor 1 and VEGF, enhances extravasation, migration and retention of transplanted cells in the infarcted myocardium [78]. Therefore, stem cells can be delivered by intravenous or intracoronary 73

6 Review Lee, Siu, Zhang & Tse Table 2. Summary of previous studies of embryonic stem cell transplantation into the myocardium. Author (year) Cell type Animal model Time to injection Klug et al. (1996) mesc-d3-derived CMs Min et al. (2002) mesc-d3-derived CMs Behfar et al. (2002) mesc-cgr8 (undifferentiated) Yang et al. (2002) mesc-d3-derived CMs Min et al. (2003) mesc-d3-derived CMs Hodgson et al. (2004) mesc-cgr8- derived CMs Kehat et al. (2004) hesc-h9.2- derived CMs Hime et al. (2004) mesc (undifferentiated) Kofidis et al. mesc (2004) (undifferentiated) Kofidis et al. mesc (2004) (undifferentiated) Naito et al. (2004) mesc-derived CMs Laflamme et al. hesc-h1- and (2005) H7-derived CMs Ke et al. (2005) mesc-d3 (undifferentiated) Menard et al. mesc-cgr8- (2005) derived CMs Swijnenburg et al. (2005) mesc-d3 (undifferentiated) Adult dystrophic mice with uninjured myocardium Rat myocardial infarct model with left anterior descending artery ligation Rat myocardial infarct model with left coronray artery ligation Mouse myocardial infarct model with left coronary artery ligation Rat myocardial infarct model with left anterior descending artery ligation Rat myocardial infarct model with left anterior descending artery ligation Swine complete atrioventricular block model Follow-up Findings Ref. NA 7 weeks Grafted cells formed intracardiac graft in the mouse myocardium 30 min 6 weeks Improved left ventricular systolic function; grafted cells transformed into CMs 4 weeks 5 weeks Grafted cells transformed into CMs; improved left ventricular systolic function 15 weeks 6 weeks Reduced infarct size and reduced postinfarct remodeling; better improvement in VEGF with substantial angiogenesis 20 min 32 weeks Improved survival rate; reduced infarct size; improved left ventricular systolic function; grafted cells transformed into CMs; increased capillary densities 8 weeks 12 weeks Improved left ventricular ejection fraction and reduced postinfarct remodeling; grafted cells transformed into CMs Same session 1 3 weeks Pacemaker activity from the grafted cells [109] Mouse myocardial infarct model Same 12 h 36 days Tracking with MRI [110] session Rat heterotopically transplanted heart NA 2 weeks Improved left ventricular fractional shortening and reduced [111] with/without myocardial infarct postinfarct remodeling Mouse myocardial infarct model with Same 2 weeks Improved left ventricular fractional shortening and reduced [112] left anterior descending artery ligation session postinfarct remodeling Rats with uninjured myocardium NA 1 week Grafted cell survival [113] and cyclosporine [114] Athymic rats with uninjured myocardium Mouse myocardial infarct model with left coronary artery ligation Sheep myocardial infarct model with left circumflex artery embolization Mouse myocardial infarct model with coronary artery ligation NA 4 weeks Grafted cells formed stable expanding graft in the rat myocardium; graft fiber strands aligned in parallel with host myocardium 15 min 8 weeks Grafted cells were harbored into host myocardium; increase in wall thickness and reduced infarct size in the patch-transplanted group 2 weeks 1 month Grafted cells survived and differentiated into mature CMs in postinfarct myocardium in both immunosuppressed and immunocompetent sheep; improved left ventricular ejection fraction 5 min 1 8 weeks Teratoma formation at 2 weeks; vigorous inflammatory infiltrate at 4 and 8 weeks Xue et al. (2005) hesc-h1 Uninjured guinea pig myocardium NA NA Electrical coupling between the transplanted hesc-derived pacemaker and the myocardium Singla et al. (2006) mesc-r1 and HM1 (undifferentiated) Mouse myocardial infarct model with coronary artery ligation 3 5 h post-mi CM: Cardiomyocyte; hesc: Human embryonic stem cell; mesc: Mouse embryonic stem cell; MI: Myocardial infarction; NA: Not applicable. 2 weeks Grafted cells survived in postinfarct myocardium, but no noninjured hearts; grafted cells transformed into CMs, vascular smooth muscle and endothelial cells demonstrated by colocalization immunolabeling; improved cardiac function and reduced postinfarct remodeling [103] [104] [105] [106] [107] [108] [115] [116] [117] [118] [119] 74 Therapy (2011) 8(1)

7 Current perspective of stem cell therapies for cardiac regeneration Review Acute myocardial infarction Myocardial ischemia/hibernation Myocardial scar Cellular homing signal Intravenous/intracoronary delivery Intramyocardial delivery? Myocardial patch Figure 3. Modes of stem cell delivery based on the diseased status of the myocardium. The number of + represent the relative amount of cellular homing signal, while - represents no cellular homing signal. routes after coronary revascularization. The intravenous infusion is the simplest approach, but has a low efficacy for cellular engraftment (<1%) due to cell trapping in the other organs. Selective intracoronary cell injection via an overthe-wire infusion balloon catheter enables the maximum dose of cells to be delivered to the infarcted region. However, neither intravenous nor intracoronary routes are suitable for patients with an occluded artery or for delivery of stem cells of a larger size or with a limited migration ability, such as skeletal myoblasts, owing to the risk of microembolization. For patients with chronic myocardial ischemia, the homing signals are expressed at low levels such that intravenous or intracoronary delivery will provide limited cell engraftment. In this setting, direct intramyocardial injection via either surgical epicardial or transcatheter endocardial approaches is needed for optimal cell delivery. Epicardial injection allows direct visualization of the targeted regions but is only applicable to patients requiring coronary artery bypass grafting. By contrast, catheter-based intramyocardial injection can be performed as a standalone procedure, but requires the use of specially designed injection catheters, with or without the use of 3D electromechanical mapping systems to guide the injection into the targeted regions. These techniques allow direct cell delivery into the targeted regions, even in patients with occluded arteries and when using certain larger cell types. Finally, in patients with post-mi heart failure, owing to the large area of infarcted and nonviable myocardium, direct injection of stem cells into the myocardium scar will result in low graft survival and differentiation. This is caused by the lack of blood supply as well as paracrine supports from adjacent host CMs. Furthermore, transplanting viable cells into the scar may also increase the risk of proarrhythmias [62]. Therefore, the use of bioengineering approaches, such as cardiac patches and injectable delivery matrices, may be needed to improve cell retention, survival and differentiation [79]. However, the safety and efficacy of this approach has not been tested in clinical trials owing to the lack of optimal techniques to create viable 3D cellular patches. Clinical experiences The clinical use of stem cell therapy for treatment of cardiac diseases is still in its infancy. To date, only pilot cohort studies or small scale Phase I II randomized trials have been reported for the treatment of acute MI and chronic myocardial ischemia with congestive heart failure and/or refractory angina. In this article, only data from randomized controlled trials or meta-ana lyses will be discussed. Acute MI In patients with acute MI, the majority of randomized controlled clinical trials have focused on the use of intracoronary administration 75

8 Review Lee, Siu, Zhang & Tse of autologous BM cells in patients who had undergone successful percutaneous coronary intervention at the infarct region. These studies yielded mixed results regarding the effect of intracoronary administration of BM cells owing to the relatively small patient sample size (<200), differences in the study population, the dosage, preparation and types of cells, timing of cell transfer, and the methodology of functional assessment. Nevertheless, none of these studies have shown major adverse effects after intracoronary BM cell delivery [60 68]. As a result, several meta-analyses, including randomized studies only, or both randomized and cohort studies, have been performed to determine the potential therapeutic benefits of intracoronary BM cell therapy during acute MI [80 82]. All these ana lyses demonstrated a modest (~3 4%) but significant improvement in LV ejection fraction, and a small reduction in infarct scar size and LV dimension after intracoronary stem cell therapy as compared with controls. This improvement in LV ejection fraction was similar to those (~2.5%) observed in the Remodeling in Acute Myocardial Infarction (REPAIR-AMI) trial, which is the largest randomized, controlled clinical study reported [83]. Interestingly, the benefit of stem cell therapy appears to be greater in those patients with more severe LV dysfunction after acute MI [83,84]. Indeed, the magnitude of benefits on LV function observed in stem cell therapy is comparable to those of other established therapies for acute MI, such as reperfusion therapies, angiotensin-converting enzyme inhibitors, angiotensin receptor blockers and b blockers [85]. Several larger ongoing studies, such as Swiss Multicenter Intracoronary Stem Cells Study in Acute Myocardial Infarction (SWISS-AMI) and the Bone Marrow Transfer to Enhance ST-Elevation Infarct Regeneration (BOOST)-2 trials, will confirm the results of these meta-analyses, and should provide more information on the optimal timing and dosage of cell administration. Finally, the observed beneficial effects of stem cell therapy on surrogate markers, such as LV ejection fraction and dimension, will also need to be validated in subsequent clinical trials using clinical outcomes as study end points. Chronic myocardial ischemia In contrast to acute MI, there are only a few small randomized controlled clinical trials on stem cell therapy for the treatment of refractory angina caused by chronic myocardial ischemia. These trials mainly focused on the use of intramyocardial injections of BM cells into chronic ischemic myocardium that are not amenable to conventional coronary revascularization [76 78]. In these studies, cell transplantation was performed by 3D electromechancial mapping-guided catheter-based intra myocardial injection into the ischemic myocardium, as described previously [79,80]. Losordo et al. studied the use of three different doses of granulocyte colony-stimulating factor mobilizing CD34 + cells in patients with chronic myocardial ischemia [86]. Overall, they demonstrated a reduction in angina frequency and an increased exercise capacity without any change in SPECT perfusion [76]. The other two trials on the use of BM mononuclear cells showed significant but modest improvement in exercise capacity and LV ejection fraction [77,78]. Furthermore, a significant improvement in SPECT perfusion was also observed in the study by van Ramshorst et al. [87]. The reasons for the conflicting results on SPECT perfusion observed in these trials remain unclear, but are likely to be caused by sampling variation, dosage of cells used and the method of assessment of SPECT perfusion. Furthermore, no significant adverse events, such as cardiac arrhythmias, were observed after direct intramyocardial injection of BM cells [76 80]. Interestingly, assessment of cardiac function by cardiac MRI in two of the larger trials, which included patients with relatively normal LV ejection fraction, consistently showed a similar degree of improvement (3 5%) after BM cell transplantation [77,78]. The effects of BM cell transplantation in patients with ischemic LV dysfunction need to be addressed in future clinical trials. Congestive heart failure There are only two randomized controlled clinical trials on the use of autologous BM or skeletal myoblast cells in patients with congestive heart failure after MI. In the Transplantation of Progenitor Cells in Patients With Persistent Left Ventricular Function After Myocardial Infarction (TOPCARE CHD) study, patients who had chronic MI for more than 3 months were randomized to either receive an intra coronary transplantation of BM cells or circulating progenitor cells from peripheral blood [81]. Patients who received intracoronary BM cell transplantation but not circulating progenitor cells showed a modest but significant improvement in LV ejection fraction (2.9%) at 76 Therapy (2011) 8(1)

9 Current perspective of stem cell therapies for cardiac regeneration Review 3 months compared with controls. However, there was no difference in LV dimension after cell transplantation. In patients with congestive heart failure caused by the loss of CM after MI, it is conceivable that transplantation of cells with contractile properties to repopulate the akinetic nonviable myocardium may restore cardiac mechanical function. Therefore, the use of skeletal myoblasts has been investigated in patients with ischemic cardiomyopathy. The Myoblast Autologous Grafting in Ischemic Cardiomyopathy (MAGIC) trial investigated the safety and efficacy of low-dose ( ) or high-dose ( ) autologous skeletal myoblast cell transplantation versus placebo in patients with ischemic cardiomyopathy and indication for coronary artery bypass grafting [88]. Owing to the safety concern regarding proarrhythmia in the early pilot studies [89], all patients were treated with implantable cardioverter-defibrillators before transplantation. At 6 months, there were no significant differences in regional or global LV function as determined by echocardiogram and arrhythmia events among these three groups. However, patients receiving the highest dose of cells had a significant decrease in LV end-diastolic and end-systolic volume compared with controls, suggesting the possibility of reverse remodeling [82]. Potential hurdles & future perspective Although stem cell therapies appear to be a promising therapeutic approach in patients with severe CAD, there are several major issues that need to be resolved. First, current evidence supports the notion that stem cell-mediated paracrine effects, including enhancement of neovascularization and extracellular matrix remodeling, prevention of apoptosis and recruitment of endogenous cardiac stem cells by multiple cytokines that could enhance homing signal from the injury site [90], rather than direct replacement of damaged myocardium, are the major mechanisms of action of stem cell therapies, especially with the use of adult stem cells. Nevertheless, the optimal cell types will also depend on the clinical setting. In patients with acute MI or chronic myocardial ischemia, the primary objectives are to salvage the ischemic myocardium by improving myocardial perfusion; and to preserve cardiac function by limiting LV remodeling. Therefore, the paracrine effects mediated by BM- or MSCderived cells may be sufficient to achieve the therapeutic target. In addition, some authors suggested that MSC-derived cells may have immuno modulatory effects, which may subsequently modulate the inflammatory response to acute MI, thereby limiting the amount of initial tissue damage [91]. Recent studies have failed to demonstrate any differences in clinical efficacy of direct intracoronary administration of unselected mononuclear BM cells versus CD34/CXCR4 + BM cells [84]. However, a similar improvement in LV ejection fraction was achieved with selected CD34/CXC + BM cells despite the fact that the total number of cells injected was approximately 100 times lower than unselected BM cells. Whether the use of a selected BM cell population at a higher dosage may further improve the efficacy of the stem cell therapy remains unclear. Second, the clinical application of autologous BM-derived cells, including MSCs, is still limited by the depletion and/or functional impairment of those stem cells associated with aging, diabetes [92] and severe CAD [93]. Furthermore, the process of cell preparation and selection can also significantly impact on the clinical efficacy [94]. The potential solutions for these hurdles include the use of standardized allogenic cell products or extracts from healthy donors with limited immunogenicity, such as MSCs derived from different tissues or conditional medium with cytokines secreted from different cell sources during culturing [95]. In addition, genetical manipulation of autologous cells to enhance their functions, such as overexpression of endothelial nitric oxide synthase, may also overcome hereditary defects of patients stem cells [96]. Third, structural regeneration of damaged myocardium is required for post-mi heart failure caused by massive loss of CMs. In this situation, transplantation of stem cells that can differentiate into functional CMs and integrate with host CMs is essential to achieve the therapeutic target. Unfortunately, the majority of adult stem cells rarely transdifferentiate into functional CMs and fail to integrate fully with host CMs. Among different cell types, resident cardiac stem cells, ESCs and ips cells have better potential than other adult stem cells for proliferation and differentiation into functional CMs. Nevertheless, these cell types also have their own limitation as discussed previously. Most importantly, the generation of terminally differentiated and homogeneous functional CMs from these different types of stem cells may be needed to avoid the issues of teratoma formation and proarrhythmias. 77

10 Review Lee, Siu, Zhang & Tse Finally, the rate of sustained cell engraftment and integration after transplantation remains low. In preclinical studies, less than 10% of injected cells remain in the host myocardium after the first day and less than 1% of transplanted cells can still be detected after 4 weeks [97]. Indeed, a histological study also confirmed that the majority of stem cells did not engraft or survive after transplantation [13]. As a result, enhancement of cell engraftment, as well as survival, is mandatory for optimizing the therapeutic benefits of stem cell therapy for post-mi heart failure. The low rate of cell engraftment is caused by the loss of transplanted cells during the cell delivery procedure and mechanical contraction. The potential solutions to improve cell engraftment included tissue engineering of transplanted cells to enhance cell homing, such as overexpression of stromal cell-derived factor 1 [98], or nitric oxide synthase enhancers [99], and to improve cell delivery using cardiac patches and injectable delivery matrices [79]. On the other hand, multiple mechanisms, including ischemia, inflammatory reactions, apoptosis and loss of cell cell interactions, are likely to contribute to the poor survival of transplanted cells. As a result, different therapeutic strategies to enhance angiogensis via co-administration of EPCs or growth factors [92], and to prevent apoptosis by cellular preconditioning, such as hypoxia [93] or coexpression of Akt [100], have been investigated to improve cell survival. Furthermore, embedding cells into biomaterial as cardiac patches or injectable matrices can also improve cell survival by promoting cell-to-cell contact and act as barriers to reduce inflammatory reactions. Conclusion The lack of optimal therapeutic options for the treatment of organ failure has motivated the pursuit for an alternative biological therapeutic approach. In the last decade, preclinical experimental data, as well as pilot human clinical trials, suggest that damaged myocardium can be repaired by cell-based therapies using different types of stem cells. Despite much hetero geneity in clinical trials, including cell types, dosage, study design, mode of delivery and methodology, it has been demonstrated that there is a modest beneficial effect on the LV ejection fraction with the use of BM-derived cellular therapy in patients with acute MI or chronic myocardial ischemia, which goes beyond the magnitude of conventional therapy with a reasonable safety margin. Future challenges in the field include optimizing the cell type, dosage, modes of delivery and the choice of relevant functional assessment. Financial & competing interests disclosure This work was supported by the Hong Kong Research Grant Council General Research Fund (HKU M and HKU 7747/08M to CW Siu and HF Tse) and the CC Wong Stem Cell Fund (to HF Tse). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilized in the production of this manuscript. Executive summary Background Clinical applications of stem cell therapies have been investigated as a novel therapy for the treatment of acute myocardial infarction and chronic myocardial ischemia. Mechanisms of action Current evidence suggests that cell-mediated paracrine effects, rather than direct cardiac transdifferentiation, after different types of stem cell transplantation, are more likely to contribute to the observed beneficial effects of stem cell therapies. Cell types Among the different types of stem cells being investigated, bone marrow-derived cells are the most commonly tested in clinical trials. Clinical experiences Phase I/II randomized controlled clinical trials suggest that intracoronary or intramyocardial injection of bone marrow-derived cells may be a safe and feasible strategy for the treatment of acute myocardial infarction as well as chronic myocardial ischemia. Initial results show that these cell-based therapies are associated with a modest but significant improvement in left ventricular ejection fraction and the clinical status of patients after cell transplantation. The clinical efficacy of stem cell therapies needs to be confirmed by future clinical trials. Future perspective Future investigations should address the optimal timing, cell types and modes of delivery, and develop strategies to improve cell survival and engraftment in order to overcome the potential hurdles related to the current applications of cell-based therapies. The possible reason for improved cardiac function and survival of engrafted stem cells by paracrine mechanisms should not be ignored either. Identification of these cell-derived paracrine factors could lead to effective therapies without delivery of the cells themselves. Local intramyocardial delivery of cytokines or growth factors could be more reproducible with defined composition. 78 Therapy (2011) 8(1)

11 Current perspective of stem cell therapies for cardiac regeneration Review Bibliography 1 Dean TJ, Creese A, Prentice T: The double burden: emerging epidemics and persistent problems. In: The World Health Report WHO, Geneva, Switzerland, (1999). 2 Velagaleti RS, Pencina MJ, Murabito JM et al.: Long-term trends in the incidence of heart failure after myocardial infarction. Circulation 118, (2008). 3 Bergmann O, Bhardwaj RD, Bernard S et al.: Evidence for cardiomyocyte renewal in humans. Science 324, (2009). 4 Nadal-Ginard B, Kajstura J, Leri A, Anversa P: Myocyte death, growth, and regeneration in cardiac hypertrophy and failure. Circ. Res. 92, (2003). 5 Gepstein L: Derivation and potential applications of human embryonic stem cells. Circ. Res. 91, (2002). 6 Segers VF, Lee RT: Stem-cell therapy for cardiac disease. Nature 451, (2008). 7 Cheng AS, Yau TM: Paracrine effects of cell transplantation: strategies to augment the efficacy of cell therapies. Semin. Thorac. Cardiovasc. Surg. 20, (2008). 8 Tse HF, Thambar S, Kwong YL et al.: Prospective randomized trial of direct endomyocardial implantation of bone marrow cells for treatment of severe coronary artery diseases (PROTECT CAD trial). Eur. Heart J. 28, (2007). 9 Quaini F, Urbanek K, Beltrami AP et al.: Chimerism of the transplanted heart. N. Engl. J. Med. 346, 5 15 (2002). 10 Gnecchi M, Zhang Z, Ni A, Dzau VJ: Paracrine mechanisms in adult stem cell signaling and therapy. Circ. Res. 103, (2008). 11 Balsam LB, Wagers AJ, Christensen JL, Kofidis T, Weissman IL, Robbins RC: Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium. Nature 428, (2004). 12 Murry CE, Soonpaa MH, Reinecke H et al.: Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature 428, (2004). 13 van der Bogt KE, Sheikh AY, Schrepfer S et al.: Comparison of different adult stem cell types for treatment of myocardial ischemia. Circulation 118, S121 S129 (2008). 14 Fedak PW: Paracrine effects of cell transplantation: modifying ventricular remodeling in the failing heart. Semin. Thorac. Cardiovasc. Surg. 20, (2008). 15 Yeghiazarians Y, Zhang Y, Prasad M et al.: Injection of bone marrow cell extract into infarcted hearts results in functional improvement comparable to intact cell therapy. Mol. Ther. 17(7), (2009). 16 Pagani FD, DerSimonian H, Zawadzka A et al.: Autologous skeletal myoblasts transplanted to ischemia-damaged myocardium in humans. Histological ana lysis of cell survival and differentiation. J. Am. Coll. Cardiol. 41, (2003). 17 Chiu RC, Zibaitis A, Kao RL: Cellular cardiomyoplasty: myocardial regeneration with satellite cell implantation. Ann. Thorac. Surg. 60, (1995). 18 Jain M, DerSimonian H, Brenner DA et al.: Cell therapy attenuates deleterious ventricular remodeling and improves cardiac performance after myocardial infarction. Circulation 103, (2001). 19 Murry CE, Wiseman RW, Schwartz SM, Hauschka SD: Skeletal myoblast transplantation for repair of myocardial necrosis. J. Clin. Invest. 98, (1996). 20 Pouzet B, Vilquin JT, Hagege AA et al.: Intramyocardial transplantation of autologous myoblasts: can tissue processing be optimized? Circulation 102, III210 III215 (2000). 21 Rajnoch C, Chachques JC, Berrebi A, Bruneval P, Benoit MO, Carpentier A: Cellular therapy reverses myocardial dysfunction. J. Thorac. Cardiovasc. Surg. 121, (2001). 22 Scorsin M, Hagege A, Vilquin JT et al.: Comparison of the effects of fetal cardiomyocyte and skeletal myoblast transplantation on postinfarction left ventricular function. J. Thorac. Cardiovasc. Surg. 119, (2000). 23 Taylor DA, Atkins BZ, Hungspreugs P et al.: Regenerating functional myocardium: improved performance after skeletal myoblast transplantation. Nat. Med. 4, (1998). 24 Menasche P, Hagege AA, Scorsin M et al.: Myoblast transplantation for heart failure. Lancet 357, (2001). 25 Dib N, Michler RE, Pagani FD et al.: Safety and feasibility of autologous myoblast transplantation in patients with ischemic cardiomyopathy: four-year follow-up. Circulation 112, (2005). 26 Gavira JJ, Herreros J, Perez A et al.: Autologous skeletal myoblast transplantation in patients with nonacute myocardial infarction: 1 year follow-up. J. Thorac. Cardiovasc. Surg. 131, (2006). 27 Menasche P, Hagege AA, Vilquin JT et al.: Autologous skeletal myoblast transplantation for severe postinfarction left ventricular dysfunction. J. Am. Coll. Cardiol. 41, (2003). 28 Siminiak T, Fiszer D, Jerzykowska O, Grygielska B, Kalmucki P, Kurpisz M: Percutaneous autologous myoblast transplantation in the treatment of post-infarction myocardial contractility impairment report on two cases. Kardiol. Pol. 59, (2003). 29 Siminiak T, Kalawski R, Fiszer D et al.: Autologous skeletal myoblast transplantation for the treatment of postinfarction myocardial injury: Phase I clinical study with 12 months of follow-up. Am. Heart J. 148, (2004). 30 Suzuki K, Brand NJ, Allen S et al.: Overexpression of connexin 43 in skeletal myoblasts: relevance to cell transplantation to the heart. J. Thorac. Cardiovasc. Surg. 122, (2001). 31 Thompson RB, Emani SM, Davis BH et al.: Comparison of intracardiac cell transplantation: autologous skeletal myoblasts versus bone marrow cells. Circulation 108(Suppl. 1), II264 II271 (2003). 32 Orlic D, Kajstura J, Chimenti S et al.: Bone marrow cells regenerate infarcted myocardium. Nature 410, (2001). 33 Nygren JM, Jovinge S, Breitbach M et al.: Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nat. Med. 10, (2004). 34 Tse HF, Yiu KH, Lau CP: Bone marrow stem cell therapy for myocardial angiogenesis. Curr. Vasc. Pharmacol. 5, (2007). 35 Shantsila E, Watson T, Tse HF, Lip GY: New insights on endothelial progenitor cell subpopulations and their angiogenic properties. J. Am. Coll. Cardiol. 51, (2008). 36 Asahara T, Murohara T, Sullivan A et al.: Isolation of putative progenitor endothelial cells for angiogenesis. Science 275, (1997). 37 Reyes M, Dudek A, Jahagirdar B, Koodie L, Marker PH, Verfaillie CM: Origin of endothelial progenitors in human postnatal bone marrow. J. Clin. Invest. 109, (2002). 38 Shi Q, Rafii S, Wu MH et al.: Evidence for circulating bone marrow-derived endothelial cells. Blood 92, (1998). 79

12 Review Lee, Siu, Zhang & Tse 39 Pittenger MF, Mackay AM, Beck SC et al.: Multilineage potential of adult human mesenchymal stem cells. Science 284, (1999). 40 Makino S, Fukuda K, Miyoshi S et al.: Cardiomyocytes can be generated from marrow stromal cells in vitro. J. Clin. Invest. 103, (1999). 41 Amado LC, Saliaris AP, Schuleri KH et al.: Cardiac repair with intramyocardial injection of allogeneic mesenchymal stem cells after myocardial infarction. Proc. Natl Acad. Sci. USA 102, (2005). 42 Pittenger MF, Martin BJ: Mesenchymal stem cells and their potential as cardiac therapeutics. Circ. Res. 95, 9 20 (2004). 43 Jiang Y, Jahagirdar BN, Reinhardt RL et al.: Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 418, (2002). 44 Yoon YS, Wecker A, Heyd L et al.: Clonally expanded novel multipotent stem cells from human bone marrow regenerate myocardium after myocardial infarction. J. Clin. Invest. 115, (2005). 45 Planat-Benard V, Menard C, Andre M et al.: Spontaneous cardiomyocyte differentiation from adipose tissue stroma cells. Circ. Res. 94, (2004). 46 Planat-Benard V, Silvestre JS, Cousin B et al.: Plasticity of human adipose lineage cells toward endothelial cells: physiological and therapeutic perspectives. Circulation 109, (2004). 47 Miyahara Y, Nagaya N, Kataoka M et al.: Monolayered mesenchymal stem cells repair scarred myocardium after myocardial infarction. Nat. Med. 12, (2006). 48 Gimble JM, Katz AJ, Bunnell BA: Adipose-derived stem cells for regenerative medicine. Circ. Res. 100, (2007). 49 Das H, Abdulhameed N, Joseph M, Sakthivel R, Mao HQ, Pompili VJ: Ex vivo nanofiber expansion and genetic modification of human cord blood-derived progenitor/stem cells enhances vasculogenesis. Cell Transplant. 18, (2009). 50 Ma N, Stamm C, Kaminski A et al.: Human cord blood cells induce angiogenesis following myocardial infarction in NOD/scid-mice. Cardiovasc. Res. 66, (2005). 51 Ghodsizad A, Niehaus M, Kogler G et al.: Transplanted human cord blood-derived unrestricted somatic stem cells improve left-ventricular function and prevent left-ventricular dilation and scar formation after acute myocardial infarction. Heart 95, (2009). 52 Beltrami AP, Barlucchi L, Torella D et al.: Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 114, (2003). 53 Kajstura J, Urbanek K, Rota M et al.: Cardiac stem cells and myocardial disease. J. Mol. Cell. Cardiol. 45, (2008). 54 Li Z, Lee A, Huang M et al.: Imaging survival and function of transplanted cardiac resident stem cells. J. Am. Coll. Cardiol. 53, (2009). 55 Smith RR, Barile L, Cho HC et al.: Regenerative potential of cardiosphere derived cells expanded from percutaneous endomyocardial biopsy specimens. Circulation 115, (2007). 56 Andersen DC, Andersen P, Schneider M, Jensen HB, Sheikh SP: Murine cardiospheres are not a source of stem cells with cardiomyogenic potential. Stem Cells 27, (2009). 57 Odorico JS, Kaufman DS, Thomson JA: Multilineage differentiation from human embryonic stem cell lines. Stem Cells 19, (2001). 58 Reubinoff BE, Pera MF, Fong CY, Trounson A, Bongso A: Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat. Biotechnol. 18, (2000). 59 Kehat I, Kenyagin-Karsenti D, Snir M et al.: Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J. Clin. Invest. 108, (2001). 60 He JQ, Ma Y, Lee Y, Thomson JA, Kamp TJ: Human embryonic stem cells develop into multiple types of cardiac myocytes: action potential characterization. Circ. Res. 93, (2003). 61 Habib M, Caspi O, Gepstein L: Human embryonic stem cells for cardiomyogenesis. J. Mol. Cell. Cardiol. 45, (2008). 62 Macia E, Boyden PA: Stem cell therapy is proarrhythmic. Circulation 119, (2009). 63 Siu CW, Moore JC, Li RA: Human embryonic stem cell-derived cardiomyocytes for heart therapies. Cardiovasc. Hematol. Disord. Drug Targets 7, (2007). 64 Shih CC, Forman SJ, Chu P, Slovak M: Human embryonic stem cells are prone to generate primitive, undifferentiated tumors in engrafted human fetal tissues in severe combined immunodeficient mice. Stem Cells Dev. 16, (2007). 65 Cao F, van der Bogt KE, Sadrzadeh A et al.: Spatial and temporal kinetics of teratoma formation from murine embryonic stem cell transplantation. Stem Cells Dev. 16, (2007). 66 Park IH, Zhao R, West JA et al.: Reprogramming of human somatic cells to pluripotency with defined factors. Nature 451, (2008). 67 Takahashi K, Tanabe K, Ohnuki M et al.: Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, (2007). 68 Yu J, Vodyanik MA, Smuga-Otto K et al.: Induced pluripotent stem cell lines derived from human somatic cells. Science 318, (2007). 69 Mauritz C, Schwanke K, Reppel M et al.: Generation of functional murine cardiac myocytes from induced pluripotent stem cells. Circulation 118, (2008). 70 Okita K, Ichisaka T, Yamanaka S: Generation of germline-competent induced pluripotent stem cells. Nature 448, (2007). 71 Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S: Generation of mouse induced pluripotent stem cells without viral vectors. Science 322, (2008). 72 Stadtfeld M, Nagaya M, Utikal J, Weir G, Hochedlinger K: Induced pluripotent stem cells generated without viral integration. Science 322, (2008). 73 Yusa K, Rad R, Takeda J, Bradley A: Generation of transgene-free induced pluripotent mouse stem cells by the piggybac transposon. Nat. Methods 6, (2009). 74 Woltjen K, Michael IP, Mohseni P et al.: piggybac transposition reprograms fibroblasts to induced pluripotent stem cells. Nature 458, (2009). 75 Kaji K, Norrby K, Paca A, Mileikovsky M, Mohseni P, Woltjen K: Virus-free induction of pluripotency and subsequent excision of reprogramming factors. Nature 458, (2009). 76 Soldner F, Hockemeyer D, Beard C et al.: Parkinson s disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell 136, (2009). 77 Kim D, Kim CH, Moon JI et al.: Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cell 4, (2009). 78 Smart N, Riley PR: The stem cell movement. Circ. Res. 102, (2008). 80 Therapy (2011) 8(1)

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

List of publications Curriculum Vitae Acknowledgements

List of publications Curriculum Vitae Acknowledgements List of publications Curriculum Vitae Acknowledgements List of publications 153 LIST OF PUBLICATIONS 1. Van der Bogt KE, Sheik AY, Schrepfer S, Hoyt G, Cao F, Ransohoff K, Swijnenburg RJ, Pearl JI, Lee

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

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

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

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

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

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

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

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

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

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

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

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

JACC: CARDIOVASCULAR INTERVENTIONS VOL. 2, NO. 1, PUBLISHED BY ELSEVIER INC. DOI: /j.jcin

JACC: CARDIOVASCULAR INTERVENTIONS VOL. 2, NO. 1, PUBLISHED BY ELSEVIER INC. DOI: /j.jcin JACC: CARDIOVASCULAR INTERVENTIONS VOL. 2, NO. 1, 2009 2009 BY THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION ISSN 1936-8798/09/$36.00 PUBLISHED BY ELSEVIER INC. DOI: 10.1016/j.jcin.2008.11.003 CLINICAL

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

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

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

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

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

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

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

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

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

Review Article Present and Future Perspectives on Cell Sheet-Based Myocardial Regeneration Therapy

Review Article Present and Future Perspectives on Cell Sheet-Based Myocardial Regeneration Therapy BioMed Volume 2013, Article ID 583912, 6 pages http://dx.doi.org/10.1155/2013/583912 Review Article Present and Future Perspectives on Cell Sheet-Based Myocardial Regeneration Therapy Yoshiki Sawa and

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

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

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

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

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

Transcoronary Infusion of Cardiac Progenitor Cells in Hypoplastic Left Heart Syndrome: 3-year Follow-up of the TICAP Trial

Transcoronary Infusion of Cardiac Progenitor Cells in Hypoplastic Left Heart Syndrome: 3-year Follow-up of the TICAP Trial Transcoronary Infusion of Cardiac Progenitor Cells in Hypoplastic Left Heart Syndrome: 3-year Follow-up of the TICAP Trial Shunji Sano, Shuta Ishigami, Takuya Goto, Daiki Ousaka, Suguru Tarui, Michihiro

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

Masashi Tanaka, Tanaka M Tanaka M Tanaka M Tanaka M Tanaka M

Masashi Tanaka,  Tanaka M Tanaka M Tanaka M Tanaka M Tanaka M 2010 1 1996 1996 2000 2002 2004 2005 2007 2009 2004 1996 2004 2007 2 2007 United States Patent Application Serial No. 11/271,285 for Methods and Compositions for Reducing Injury to a Transplanted Organ

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

Cardiac Stem Cells and Mechanisms of Myocardial Regeneration

Cardiac Stem Cells and Mechanisms of Myocardial Regeneration Physiol Rev 85: 1373 1416, 2005; doi:10.1152/physrev.00013.2005. Cardiac Stem Cells and Mechanisms of Myocardial Regeneration ANNAROSA LERI, JAN KAJSTURA, AND PIERO ANVERSA Cardiovascular Research Institute,

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

Journal Club Semmler Lorenz

Journal Club Semmler Lorenz Beer et al. 2015 - Analysis of the Secretome of Apoptotic Peripheral Blood Mononuclear Cells: Impact of Released Proteins and Exosomes for Tissue Regeneration Journal Club 13.11.2017 1 Introduction to

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

University of Milano-Bicocca School of medicine and faculty of science

University of Milano-Bicocca School of medicine and faculty of science University of Milano-Bicocca School of medicine and faculty of science Isolation, characterization and ex vivo amplification of heartderived c-kit progenitor cells for cellular therapy Coordinator: Prof.

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

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

Stem cells for cardiac repair

Stem cells for cardiac repair Stem cells for cardiac repair Master Thesis Christine van de Werken Cancer Genomics and Developmental Biology Stem cells for cardiac repair 19/12/2008 Master Thesis Christine van de Werken (0424919) Master

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

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

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

1. LV function and remodeling. 2. Contribution of myocardial ischemia due to CAD, and

1. LV function and remodeling. 2. Contribution of myocardial ischemia due to CAD, and 1 The clinical syndrome of heart failure in adults is commonly associated with the etiologies of ischemic and non-ischemic dilated cardiomyopathy, hypertrophic cardiomyopathy, hypertensive heart disease,

More information

Myocardial regeneration autologous stem cell therapy

Myocardial regeneration autologous stem cell therapy Myocardial regeneration autologous stem cell therapy 1 SPL/Agentur Focus Index Main patient cohort 3 Introduction 3 Cell therapy using adult, autologous stem cells 5 Clinical study results 6 Methodology

More information

TRANSLATIONAL AND CLINICAL RESEARCH

TRANSLATIONAL AND CLINICAL RESEARCH TRANSLATIONAL AND CLINICAL RESEARCH A Fibrin Patch-Based Enhanced Delivery of Human Embryonic Stem Cell-Derived Vascular Cell Transplantation in a Porcine Model of Postinfarction Left Ventricular Remodeling

More information

Gated blood pool ventriculography: Is there still a role in myocardial viability?

Gated blood pool ventriculography: Is there still a role in myocardial viability? Gated blood pool ventriculography: Is there still a role in myocardial viability? Oliver C. Alix, MD Adult Clinical and Nuclear Cardiology St. Luke s Medical Centre - Global City Case Presentation A 62-year-old

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

CD34+ Cells: A Comparison of Stem and Progenitor Cells in Cord Blood, Peripheral Blood, and the Bone Marrow

CD34+ Cells: A Comparison of Stem and Progenitor Cells in Cord Blood, Peripheral Blood, and the Bone Marrow White Paper September 2016 CD34+ Cells: A Comparison of Stem and Progenitor Cells in Cord Blood, Peripheral Blood, and the Bone Marrow Lily C. Trajman, PhD Introduction: Hematopoietic Stem Cells (HSCs)

More information

Cardiomyocyte progenitor cell-derived exosomes stimulate migration of endothelial cells

Cardiomyocyte progenitor cell-derived exosomes stimulate migration of endothelial cells Translational Cardiology Most Recent Points of View J. Cell. Mol. Med. Vol 14, No 5, 2010 pp. 1064-1070 Guest Editor: P. Doevendans Cardiomyocyte progenitor cell-derived exosomes stimulate migration of

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

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

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

MEDICAL POLICY SUBJECT: TRANSMYOCARDIAL REVASCULARIZATION

MEDICAL POLICY SUBJECT: TRANSMYOCARDIAL REVASCULARIZATION MEDICAL POLICY SUBJECT: TRANSMYOCARDIAL 7/21/05, 05/18/06, 03/15/07, 02/21/08,, PAGE: 1 OF: 5 If a product excludes coverage for a service, it is not covered, and medical policy criteria do not apply.

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

Stem cell therapy of cardiac disease: an update

Stem cell therapy of cardiac disease: an update Nephrol Dial Transplant (2004) 19: Editorial Comments 1673 PS analyses, the authors found that the results between these two techniques were not materially different in most of these studies. Even if the

More information

Regeneration Gaps Observations on Stem Cells and Cardiac Repair

Regeneration Gaps Observations on Stem Cells and Cardiac Repair Journal of the American College of Cardiology Vol. 47, No. 9, 2006 2006 by the American College of Cardiology Foundation ISSN 0735-1097/06/$32.00 Published by Elsevier Inc. doi:10.1016/j.jacc.2006.02.002

More information

SUPPLEMENTARY INFORMATION

SUPPLEMENTARY INFORMATION doi:10.1038/nature10188 Supplementary Figure 1. Embryonic epicardial genes are down-regulated from midgestation stages and barely detectable post-natally. Real time qrt-pcr revealed a significant down-regulation

More information

Myocardial Infarction

Myocardial Infarction Myocardial Infarction MI = heart attack Defined as necrosis of heart muscle resulting from ischemia. A very significant cause of death worldwide. of these deaths, 33% -50% die before they can reach the

More information

ENGINEERING THREE DIMENSIONAL CARDIOSPHERES FROM PLURIPOTENT STEM CELLS

ENGINEERING THREE DIMENSIONAL CARDIOSPHERES FROM PLURIPOTENT STEM CELLS ENGINEERING THREE DIMENSIONAL CARDIOSPHERES FROM PLURIPOTENT STEM CELLS A Thesis Presented to The Academic Faculty by Nicole Votaw In Partial Fulfillment of the Requirements for the Degree B.S. in Biomedical

More information

Methods Study population and design. Eligibility criteria. Randomization, cell preparation, and study intervention

Methods Study population and design. Eligibility criteria. Randomization, cell preparation, and study intervention A double-blind, randomized, controlled, multicenter study to assess the safety and cardiovascular effects of skeletal myoblast implantation by catheter delivery in patients with chronic heart failure after

More information

Reprogramming through micrornas Stefanie Dimmeler

Reprogramming through micrornas Stefanie Dimmeler Klinikum der Johann Wolfgang Goethe Universität Frankfurt am Main Reprogramming through micrornas Stefanie Dimmeler Conflict of interest: T2cure GmbH, Miragen Non-coding DNA & RNA and micrornas Human Genome

More information

Haematopoietic stem cells

Haematopoietic stem cells Haematopoietic stem cells Neil P. Rodrigues, DPhil NIH Centre for Biomedical Research Excellence in Stem Cell Biology Boston University School of Medicine neil.rodrigues@imm.ox.ac.uk Haematopoiesis: An

More information

Coronary heart disease and heart failure continue to be

Coronary heart disease and heart failure continue to be 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

More information

Cardiac repair with thymosin 4 and cardiac reprogramming factors

Cardiac repair with thymosin 4 and cardiac reprogramming factors Ann. N.Y. Acad. Sci. ISSN 0077-8923 ANNALS OF THE NEW YORK ACADEMY OF SCIENCES Issue: Thymosins in Health and Disease Cardiac repair with thymosin 4 and cardiac reprogramming factors Deepak Srivastava,

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

In the name of GOD. Animal models of cardiovascular diseases: myocardial infarction & hypertension

In the name of GOD. Animal models of cardiovascular diseases: myocardial infarction & hypertension In the name of GOD Animal models of cardiovascular diseases: myocardial infarction & hypertension 44 Presentation outline: Cardiovascular diseases Acute myocardial infarction Animal models for myocardial

More information

European Society of Cardiology Congress DONOR AGE NEGATIVELY INFLUENCES THE CYTOPROTECTIVE PARACRINE EFFECTS EXERTED BY HUMAN MESENCHYMAL STEM CELLS

European Society of Cardiology Congress DONOR AGE NEGATIVELY INFLUENCES THE CYTOPROTECTIVE PARACRINE EFFECTS EXERTED BY HUMAN MESENCHYMAL STEM CELLS European Society of Cardiology Congress 28 Aug - 01 Sep 2009, Stockholm - Sweden DONOR AGE NEGATIVELY INFLUENCES THE CYTOPROTECTIVE PARACRINE EFFECTS EXERTED BY HUMAN MESENCHYMAL STEM CELLS Massimiliano

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

Accelerate Your Research with Conversant Bio

Accelerate Your Research with Conversant Bio Accelerate Your Research with Conversant Bio 400+ Participating MDs 50+ Partner sites for tissue procurement Continuous expansion of sourcing capabilities Closely monitored chain of custody Full regulatory

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

Review. Mesenchymal Stem Cells: Biology, Pathophysiology, Translational Findings, and Therapeutic Implications for Cardiac Disease

Review. Mesenchymal Stem Cells: Biology, Pathophysiology, Translational Findings, and Therapeutic Implications for Cardiac Disease Review This Review is part of a thematic series on Stem Cells, which includes the following articles: Stem Cells Review Series: An Introduction [Circ Res. 2011;109:907 909] Biomaterials to Enhance Stem

More information

Follow-up of the CUPID Gene Therapy Trials

Follow-up of the CUPID Gene Therapy Trials Follow-up of the CUPID Gene Therapy Trials Roger J. Hajjar, MD Icahn School of Medicine at Mount Sinai New York, NY Pathway to Heart Failure Injury / Damage Mature Dysfunctional Dying New Coronary Disease

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

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

Chapter. Department of Cardiology, 2 Department of Nuclear Medicine and the 3

Chapter. Department of Cardiology, 2 Department of Nuclear Medicine and the 3 Chapter 9 Saskia L.M.A. Beeres 1 Jeroen J. Bax 1 Petra Dibbets-Schneider 2 Marcel P.M. Stokkel 2 Willem E. Fibbe 3 Ernst E. van der Wall 1 Martin J. Schalij 1 Douwe E. Atsma 1 1 Department of Cardiology,

More information

Full Record.

Full Record. 第 1 頁, 共 2 頁 Full Record Record 1 of 6 (Set #2) Title: Insulin-like growth factor-1 mediates stretch-induced upregulation of myostatin expression in neonatal rat cardiomyocytes Author(s): Shyu KG, Ko WH,

More information

Effective activity of cytokine-induced killer cells against autologous metastatic melanoma including cells with stemness features

Effective activity of cytokine-induced killer cells against autologous metastatic melanoma including cells with stemness features Effective activity of cytokine-induced killer cells against autologous metastatic melanoma including cells with stemness features Loretta Gammaitoni, Lidia Giraudo, Valeria Leuci, et al. Clin Cancer Res

More information