Abstract

Despite improvements in modern cardiovascular therapy, the morbidity and mortality of ischaemic heart disease (IHD) and heart failure (HF) remain significant in Europe and worldwide. Patients with IHD may benefit from therapies that would accelerate natural processes of postnatal collateral vessel formation and/or muscle regeneration. Here, we discuss the use of cells in the context of heart repair, and the most relevant results and current limitations from clinical trials using cell-based therapies to treat IHD and HF. We identify and discuss promising potential new therapeutic strategies that include ex vivo cell-mediated gene therapy, the use of biomaterials and cell-free therapies aimed at increasing the success rates of therapy for IHD and HF. The overall aim of this Position Paper of the ESC Working Group Cellular Biology of the Heart is to provide recommendations on how to improve the therapeutic application of cell-based therapies for cardiac regeneration and repair.

Stem cells in the context of heart repair

Stem cells are defined as cells with the ability (i) to self-renew by dividing to make copies of themselves and (ii) to differentiate to at least one other cell type.1 In the context of cell transplantation and heart repair or regeneration [i.e. replacement or regrowth of heart damaged, respectively, including restoration of the (epicardial) vasculature and neovascularization], the term “stem cells” has been widely used but in retrospect, some of the cells used should not strictly be defined as stem cells. Cells with various molecular and functional properties have been isolated from the heart and termed “cardiac stem cells” (CSCs), “cardiac progenitor cells” (CPCs), or “cardiomyocyte progenitor cells” (CMPCs).2,3 These cells can self-renew in culture, and differentiate into different lineages (endothelial cells and mesenchymal cells), but for example have limited cardiogenic differentiation capacities except under exceptional circumstances. By the addition of compounds that induce demethylation, human CMPCs do form cardiomyocytes.4 Otherwise, the only “stem cells” that form cardiomyocytes using mixtures of growth factors, collectively referred to as “cardiogenic cocktails”, are pluripotent stem cells (PSCs). PSCs can be of embryonic origin (embryonic stem cells, ESCs) or created by introducing reprogramming genes into terminally differentiated cells, to make what are called induced pluripotent stem cells (iPSCs).1 Another term now often regarded as incorrectly used is “endothelial progenitor cells” (EPCs). These cells were originally isolated as populations that grew in culture from peripheral blood samples (reviewed in5). They could form networks that resembled vasculature, but they turned out not to be true mature endothelial cells. Finally, cells that adhere onto tissue culture plastic in serum-containing growth medium and have adipogenic, osteogenic, and chondrogenic differentiation potential in culture were termed mesenchymal stem cells (MSCs).6 However, these cells have not been isolated clonally as single cells and could therefore be heterogeneous cell populations. Moreover, with the exception of MSCs derived from bone marrow, these differentiation effects are not observed in vivo. Thus, despite them all expressing a similar set of surface markers, these cells are now called “bone marrow-derived mesenchymal stromal cells” (BM-MSCs) or adipose tissue-derived mesenchymal stromal cells (AT-MSCs).7 These MSCs have not been shown to spontaneously differentiate into cardiomyocytes. For the purpose of this position paper, we use the terminology as in the (historic) literature for the sake of clarity, but we are aware of the caveats in the terminology itself.

Translation of cell therapy: successful preclinical stories with uncertain clinical efficacy

Ischaemic heart disease (IHD) and heart failure (HF) remain major causes of morbidity and mortality worldwide.8,9 Potentially valid clinical strategies aimed at repairing damaged heart muscle and ischaemic tissue and increasing the heart's regenerative potential, are currently being tested in clinical trials.2,10 Despite originally high expectations fueled by exciting scientific progress in preclinical models, and long-term, randomized clinical trials that showed reassuring safety profiles for intracoronary (IC) delivery of cells,2,11–16 regenerative therapy for cardiovascular disease had been inconsistent and shown modest efficacy thus far.10,17–23 Several limitations of most previous clinical trials of cell-based therapies were raised and should be addressed before we can fully understand their true therapeutic potential (see Table 1).

Table 1

Limitations of cell-based therapies

DrawbacksReferences
1. Low engraftment of BMCs and blood-derived EPCs12,24–26
2. Poor survival of transplanted cells in ischaemic tissue27
3. Failure of adult stem cells to differentiate efficiently into mature and functional cardiomyocytes28
4. Inadequate recruitment of circulating or resident CSCs2,29
5. Anomalous electromechanical coupling between the transplanted cells or between the transplanted and host cells with consequent arrhythmias30
6. The use of LVEF for assessing the effects of cell therapy, as this is a load-dependent variable and loss of contractility may be compensated by increases in preload and decrease in afterload that determine changes of the Frank-Starling forces31
7. Incorrect target population of not very sick patients with baseline LVEF ∼50%, with generally a favourable outcome31
8. Existence of well-established alternative therapeutic strategies (PCI, FL, ACE-, and β-blockers) that might mask potential of cell therapy effects32
9. The lack of experimental validation (prove of efficacy) of cell preparation within the trial course
DrawbacksReferences
1. Low engraftment of BMCs and blood-derived EPCs12,24–26
2. Poor survival of transplanted cells in ischaemic tissue27
3. Failure of adult stem cells to differentiate efficiently into mature and functional cardiomyocytes28
4. Inadequate recruitment of circulating or resident CSCs2,29
5. Anomalous electromechanical coupling between the transplanted cells or between the transplanted and host cells with consequent arrhythmias30
6. The use of LVEF for assessing the effects of cell therapy, as this is a load-dependent variable and loss of contractility may be compensated by increases in preload and decrease in afterload that determine changes of the Frank-Starling forces31
7. Incorrect target population of not very sick patients with baseline LVEF ∼50%, with generally a favourable outcome31
8. Existence of well-established alternative therapeutic strategies (PCI, FL, ACE-, and β-blockers) that might mask potential of cell therapy effects32
9. The lack of experimental validation (prove of efficacy) of cell preparation within the trial course

BMCs, bone marrow cells; EPCs, endothelial progenitor cells; CSCs, cardiac stem cells; LVEF, left ventricle ejection fraction; PCI, percutaneous coronary intervention; FL, fibrinolysis; ACE-, angiotensin-converting enzyme inhibition.

Table 1

Limitations of cell-based therapies

DrawbacksReferences
1. Low engraftment of BMCs and blood-derived EPCs12,24–26
2. Poor survival of transplanted cells in ischaemic tissue27
3. Failure of adult stem cells to differentiate efficiently into mature and functional cardiomyocytes28
4. Inadequate recruitment of circulating or resident CSCs2,29
5. Anomalous electromechanical coupling between the transplanted cells or between the transplanted and host cells with consequent arrhythmias30
6. The use of LVEF for assessing the effects of cell therapy, as this is a load-dependent variable and loss of contractility may be compensated by increases in preload and decrease in afterload that determine changes of the Frank-Starling forces31
7. Incorrect target population of not very sick patients with baseline LVEF ∼50%, with generally a favourable outcome31
8. Existence of well-established alternative therapeutic strategies (PCI, FL, ACE-, and β-blockers) that might mask potential of cell therapy effects32
9. The lack of experimental validation (prove of efficacy) of cell preparation within the trial course
DrawbacksReferences
1. Low engraftment of BMCs and blood-derived EPCs12,24–26
2. Poor survival of transplanted cells in ischaemic tissue27
3. Failure of adult stem cells to differentiate efficiently into mature and functional cardiomyocytes28
4. Inadequate recruitment of circulating or resident CSCs2,29
5. Anomalous electromechanical coupling between the transplanted cells or between the transplanted and host cells with consequent arrhythmias30
6. The use of LVEF for assessing the effects of cell therapy, as this is a load-dependent variable and loss of contractility may be compensated by increases in preload and decrease in afterload that determine changes of the Frank-Starling forces31
7. Incorrect target population of not very sick patients with baseline LVEF ∼50%, with generally a favourable outcome31
8. Existence of well-established alternative therapeutic strategies (PCI, FL, ACE-, and β-blockers) that might mask potential of cell therapy effects32
9. The lack of experimental validation (prove of efficacy) of cell preparation within the trial course

BMCs, bone marrow cells; EPCs, endothelial progenitor cells; CSCs, cardiac stem cells; LVEF, left ventricle ejection fraction; PCI, percutaneous coronary intervention; FL, fibrinolysis; ACE-, angiotensin-converting enzyme inhibition.

As a consequence, several strategies have been developed to improve cardiac function in response to cell delivery further. The different strategies and protocols, collectively referred to as ‘cell enhancement’, are discussed in the section “Critical issues on protocols for cell-based therapy”.

In this Position Paper of the ESC WG Cellular Biology of the Heart, we critically review the current approaches using stem cell or cell-based therapies to treat IHD and HF, and discuss promising new strategies for stem cell therapy enhancement, with the aim of increasing the efficacy and outcome of stem cell therapies in the future. The overall objective of this Position Paper is to provide recommendations on how to improve cell-based therapies for cardiac regeneration and repair in IHD and related HF.

Cell sources used in clinical trials

Several types of cells have been used in clinical trials, most of them derived from bone marrow,12,14,15,17–22,33–37 or peripheral blood,38,39 although some studies have used MSCs, cultured from a variety of tissue sources (Table 2). These heterogeneous cell populations used in the early years of regenerative cardiac medicine, have been called “first-generation” stem cells, in contrast to contemporary “second-generation” counterparts. The latter consist of more purified cell populations with a presumed greater potential for cardiac repair and are often derived from non-bone marrow sources, or subjected to genetic and pharmacological “priming” in vitro to enhance their engraftment, survival, plasticity, and paracrine activity. Mesenchymal stem cells exhibit low immunogenicity, making allogeneic application feasible. Since the quality and number of cells may diminish in patients who are older or have comorbidities or genetic defects (reviewed in63), allogeneic MSCs can be used from young healthy individuals. Five systematic reviews and meta-analyses have reported a significant improvement in left ventricle ejection fraction (LVEF) of 2–4% and a reduction in infarct scar size and left ventricular end-systolic volume after intramyocardial transplantation of bone marrow cells.23,31,64–66 To put LVEF into the correct perspective, one must realize that the size of improvement in LVEF determined by cell therapy is comparable, if not higher than what was registered in clinical trials for evaluation of other established therapies for HF, such as angiotensin receptor blockers, aldosterone antagonists, β-blockers, and cardiac resynchronization therapy.67–70 In fact, as summarized in a recent meta-analysis that quantitatively assessed the short-term (4–6 months) therapy-induced changes in LVEF in patients with HF due to left ventricular systolic dysfunction,68 the mean increase in LVEF after subtraction of placebo was 1.3% for angiotensin receptor blockers (valsartan in the Val-Heft trial),67 2.0% for aldosterone antagonists,69 2.7% for cardiac resynchronization therapy,68 and 2.9% for β-blockers (carvedilol).70 Nevertheless, all these therapies are well established to improve clinical outcome in chronic HF. However, biological activity of a cellular product may differ greatly depending on cell source, cell preparation, and cell administration techniques. Therefore, results from meta-analysis should be interpreted with caution, especially in the field of regenerative medicine. Putting together all different trials into one basket becomes more than questionable.

Table 2

Cell source for therapeutic cardiac regeneration

Cell typeNameSources/originCommonly used markersAdvantages/Therapeutic considerationsDisadvantagesReferences
First generationBone marrow and peripheral blood PCsBone marrow
Peripheral blood
CD117+, CD34+Limited regenerative potential
Clinical trial phase 3
Minimal improved cardiac function,
Limited engraftment
Readily cryopreserved
Readily genetically manipulated
Safety profile
Easy accessibility
Lack of ethical or immunological problems
Extensive clinical experience
Limited differentiation potential
Limited yield depending on source
12,14,15,17–22, 33–35,37–39
Mesenchymal SCsFoetal origins (Wharton's jelly and cord blood)
Adult tissues (i.e. bone marrow or adipose tissue)
Developing tooth bud of the mandibular third molar
CD105+, CD117+Limited regenerative potential
Clinical trial phase 3
Minimal improved cardiac function,
Limited engraftment
Readily cryopreserved
Readily genetically manipulated
Source of paracrine factors
Easy accessibility
Safety profile
Limited differentiation potential
Limited yield depending on source
40
Side population cellsCardiac biopsyAbcg2+, Mdr1+Limited regenerative potential
No current clinical strategy
Limited yield depending on source41–43
Epicardial PCsDeveloping heartWt1+, Tbx18+, CD90+, CD44+Limited regenerative potential
No current clinical strategy
Limited yield44–47
Isl+ PCsDeveloping heartIsl1+Limited regenerative potential
No current clinical strategy
Limited yield48–50
Second generationCells from cardiospheresCardiac biopsyc-kit+, Sca-1+
CD105+, CD29+, CD45
High regenerative potential
Clinical trials phases 1 and 2
Improved cardiac function
Limited engraftment
Limited yield3
c-kitCardiac biopsyc-kit+, CD45High regenerative potential
Clinical trials phase 2
Improved cardiac function
Limited engraftment
Limited yield
The paper describing phase 1
SCIPIO trial received
Expression from journal editors at The Lancet, and is still under scrutiny
2,51–54
Sca-1Cardiac biopsyckit+, CD31+, CD14, CD34, CD105+, CD45High regenerative potential
Preclinical stage
Limited engraftment
Limited yield55–57
Embryonic SCsInner mass of blastocystOct4+, Nanog+, SSEA4+High regenerative potential
Preclinical stage
FIM phase 1
Ethically controversial source
Teratoma formation
Immunogenicity
58
Induced PSCsDerived from somatic cells using reprogramming technologiesOct4+, Nanog+, SSEA4+High regenerative potential
Preclinical stage
Improved cardiomyocyte differentiation
Tumorigenicity59–62
Cell typeNameSources/originCommonly used markersAdvantages/Therapeutic considerationsDisadvantagesReferences
First generationBone marrow and peripheral blood PCsBone marrow
Peripheral blood
CD117+, CD34+Limited regenerative potential
Clinical trial phase 3
Minimal improved cardiac function,
Limited engraftment
Readily cryopreserved
Readily genetically manipulated
Safety profile
Easy accessibility
Lack of ethical or immunological problems
Extensive clinical experience
Limited differentiation potential
Limited yield depending on source
12,14,15,17–22, 33–35,37–39
Mesenchymal SCsFoetal origins (Wharton's jelly and cord blood)
Adult tissues (i.e. bone marrow or adipose tissue)
Developing tooth bud of the mandibular third molar
CD105+, CD117+Limited regenerative potential
Clinical trial phase 3
Minimal improved cardiac function,
Limited engraftment
Readily cryopreserved
Readily genetically manipulated
Source of paracrine factors
Easy accessibility
Safety profile
Limited differentiation potential
Limited yield depending on source
40
Side population cellsCardiac biopsyAbcg2+, Mdr1+Limited regenerative potential
No current clinical strategy
Limited yield depending on source41–43
Epicardial PCsDeveloping heartWt1+, Tbx18+, CD90+, CD44+Limited regenerative potential
No current clinical strategy
Limited yield44–47
Isl+ PCsDeveloping heartIsl1+Limited regenerative potential
No current clinical strategy
Limited yield48–50
Second generationCells from cardiospheresCardiac biopsyc-kit+, Sca-1+
CD105+, CD29+, CD45
High regenerative potential
Clinical trials phases 1 and 2
Improved cardiac function
Limited engraftment
Limited yield3
c-kitCardiac biopsyc-kit+, CD45High regenerative potential
Clinical trials phase 2
Improved cardiac function
Limited engraftment
Limited yield
The paper describing phase 1
SCIPIO trial received
Expression from journal editors at The Lancet, and is still under scrutiny
2,51–54
Sca-1Cardiac biopsyckit+, CD31+, CD14, CD34, CD105+, CD45High regenerative potential
Preclinical stage
Limited engraftment
Limited yield55–57
Embryonic SCsInner mass of blastocystOct4+, Nanog+, SSEA4+High regenerative potential
Preclinical stage
FIM phase 1
Ethically controversial source
Teratoma formation
Immunogenicity
58
Induced PSCsDerived from somatic cells using reprogramming technologiesOct4+, Nanog+, SSEA4+High regenerative potential
Preclinical stage
Improved cardiomyocyte differentiation
Tumorigenicity59–62

SCs, stem cells, PSCs, pluripotent stem cells, PCs, progenitor cells.

Table 2

Cell source for therapeutic cardiac regeneration

Cell typeNameSources/originCommonly used markersAdvantages/Therapeutic considerationsDisadvantagesReferences
First generationBone marrow and peripheral blood PCsBone marrow
Peripheral blood
CD117+, CD34+Limited regenerative potential
Clinical trial phase 3
Minimal improved cardiac function,
Limited engraftment
Readily cryopreserved
Readily genetically manipulated
Safety profile
Easy accessibility
Lack of ethical or immunological problems
Extensive clinical experience
Limited differentiation potential
Limited yield depending on source
12,14,15,17–22, 33–35,37–39
Mesenchymal SCsFoetal origins (Wharton's jelly and cord blood)
Adult tissues (i.e. bone marrow or adipose tissue)
Developing tooth bud of the mandibular third molar
CD105+, CD117+Limited regenerative potential
Clinical trial phase 3
Minimal improved cardiac function,
Limited engraftment
Readily cryopreserved
Readily genetically manipulated
Source of paracrine factors
Easy accessibility
Safety profile
Limited differentiation potential
Limited yield depending on source
40
Side population cellsCardiac biopsyAbcg2+, Mdr1+Limited regenerative potential
No current clinical strategy
Limited yield depending on source41–43
Epicardial PCsDeveloping heartWt1+, Tbx18+, CD90+, CD44+Limited regenerative potential
No current clinical strategy
Limited yield44–47
Isl+ PCsDeveloping heartIsl1+Limited regenerative potential
No current clinical strategy
Limited yield48–50
Second generationCells from cardiospheresCardiac biopsyc-kit+, Sca-1+
CD105+, CD29+, CD45
High regenerative potential
Clinical trials phases 1 and 2
Improved cardiac function
Limited engraftment
Limited yield3
c-kitCardiac biopsyc-kit+, CD45High regenerative potential
Clinical trials phase 2
Improved cardiac function
Limited engraftment
Limited yield
The paper describing phase 1
SCIPIO trial received
Expression from journal editors at The Lancet, and is still under scrutiny
2,51–54
Sca-1Cardiac biopsyckit+, CD31+, CD14, CD34, CD105+, CD45High regenerative potential
Preclinical stage
Limited engraftment
Limited yield55–57
Embryonic SCsInner mass of blastocystOct4+, Nanog+, SSEA4+High regenerative potential
Preclinical stage
FIM phase 1
Ethically controversial source
Teratoma formation
Immunogenicity
58
Induced PSCsDerived from somatic cells using reprogramming technologiesOct4+, Nanog+, SSEA4+High regenerative potential
Preclinical stage
Improved cardiomyocyte differentiation
Tumorigenicity59–62
Cell typeNameSources/originCommonly used markersAdvantages/Therapeutic considerationsDisadvantagesReferences
First generationBone marrow and peripheral blood PCsBone marrow
Peripheral blood
CD117+, CD34+Limited regenerative potential
Clinical trial phase 3
Minimal improved cardiac function,
Limited engraftment
Readily cryopreserved
Readily genetically manipulated
Safety profile
Easy accessibility
Lack of ethical or immunological problems
Extensive clinical experience
Limited differentiation potential
Limited yield depending on source
12,14,15,17–22, 33–35,37–39
Mesenchymal SCsFoetal origins (Wharton's jelly and cord blood)
Adult tissues (i.e. bone marrow or adipose tissue)
Developing tooth bud of the mandibular third molar
CD105+, CD117+Limited regenerative potential
Clinical trial phase 3
Minimal improved cardiac function,
Limited engraftment
Readily cryopreserved
Readily genetically manipulated
Source of paracrine factors
Easy accessibility
Safety profile
Limited differentiation potential
Limited yield depending on source
40
Side population cellsCardiac biopsyAbcg2+, Mdr1+Limited regenerative potential
No current clinical strategy
Limited yield depending on source41–43
Epicardial PCsDeveloping heartWt1+, Tbx18+, CD90+, CD44+Limited regenerative potential
No current clinical strategy
Limited yield44–47
Isl+ PCsDeveloping heartIsl1+Limited regenerative potential
No current clinical strategy
Limited yield48–50
Second generationCells from cardiospheresCardiac biopsyc-kit+, Sca-1+
CD105+, CD29+, CD45
High regenerative potential
Clinical trials phases 1 and 2
Improved cardiac function
Limited engraftment
Limited yield3
c-kitCardiac biopsyc-kit+, CD45High regenerative potential
Clinical trials phase 2
Improved cardiac function
Limited engraftment
Limited yield
The paper describing phase 1
SCIPIO trial received
Expression from journal editors at The Lancet, and is still under scrutiny
2,51–54
Sca-1Cardiac biopsyckit+, CD31+, CD14, CD34, CD105+, CD45High regenerative potential
Preclinical stage
Limited engraftment
Limited yield55–57
Embryonic SCsInner mass of blastocystOct4+, Nanog+, SSEA4+High regenerative potential
Preclinical stage
FIM phase 1
Ethically controversial source
Teratoma formation
Immunogenicity
58
Induced PSCsDerived from somatic cells using reprogramming technologiesOct4+, Nanog+, SSEA4+High regenerative potential
Preclinical stage
Improved cardiomyocyte differentiation
Tumorigenicity59–62

SCs, stem cells, PSCs, pluripotent stem cells, PCs, progenitor cells.

Moreover, functional and structural parameters such as LVEF, left ventricular end-systolic volume, and infarct scar size are regarded as surrogate endpoints that cannot substitute hard clinical endpoints.23,31,64–66 Among various possibilities (discussed in Table 1), these modest improvements and the variability between trials have been attributed to the different isolation protocols used, which may profoundly impact the function and number of bone marrow cells or blood-derived EPCs actually delivered to the patient.71,72 Therefore, the general consensus is that assessing cell number and viability along with careful cell characterization and functionality is necessary before delivering cells into patients in any clinical trials. Moreover, the effect of bone marrow mononuclear cells on the incidence of death, recurrent myocardial infarction or stroke and hospitalization for HF remains to be determined in adequately powered prospective clinical trials.

Cardiac-derived progenitor or stem cells have very recently entered the clinical trial arena. Although isolation of these cells from the heart is more invasive than bone marrow, long culture periods are required to obtain sufficient numbers for transplantation, and cell number and functional activity may decline with age, their reported intrinsic paracrine activity is expected to make them potentially good candidates for enhancing myocardial function in HF patients.73–75 Except for the small-scale transendocardial (TE) MSCs and mononuclear bone marrow cells for ischaemic cardiomyopathy trial, comparative clinical data between bone marrow-derived cells (BMCs), MSCs, and CPCs/CSCs is not available in HF patients. A few comparisons have been done in animal models of myocardial infarction (reviewed in76), and MSCs seemed to transfer more benefit on systolic function than BMCs in a chronic large animal model of myocardial infarction.77 Preclinical research thus far suggests the greatest potential functional benefit for CPCs/CSCs from the heart, followed by MSCs, with BMCs having more modest effects on LVEF.78 Conclusions about the effect on mortality of BMC therapy after acute myocardial infarction (AMI) are expected to derive from the ongoing phase III BAMI trial, despite the lack of an placebo control injected group (https://clinicaltrials.gov/ct2/show/NCT01569178).

Of note, there is still no consensus on whether transplanted cell numbers or survival in vivo are crucial for effect size. While trial-based meta-analysis suggested a relationship between cell numbers and effect in clinical trials, individual patient-based meta-analysis have not confirmed this relationship.79

Autologous cells are non-immunogenic and do generally not entail ownership—or ethical issues.80 However, their quality may diminish with age and comorbidities, and genetic defects of the patient will also be present in his/her stem cells and their derivatives. Recent developments now allow the use of allogeneic cells, which can be selected for quality and can be kept ready to use in large quantities “off the shelf” for acute applications.81

Pluripotent stem cells in clinical trials

Another class among the second-generation cells are pluripotent stem cells, both ESCs and iPSCs (Table 2). Prerequisites for clinical application of PSCs for heart regeneration and repair is their efficient and strict differentiation into cardiomyocytes and endothelial cells, which would be directly translated into high effectiveness of the therapy, with minimum risk of undesirable side effects. A clinical trial with ESC-derived cardiomyocytes in severe HF (ESCORT) has been initiated in France and is being monitored with both interest and caution.82,83 Since the same differentiation protocols for ESCs are effective in iPSCs, it may be expected that iPSC-based approaches will also move forward for the treatment of advanced HF. iPSC-derived cardiomyocytes have not yet been tested in humans, largely because of the extra risk of genetic mutation inherent to the reprogramming method as such. The first results of ESC-mediated eye repair are encouraging84 and iPSCs for this aim are in clinical trial since September 2014. The later study is on hold since July 2015 after the identification of a mutation in an oncogene in one of the human iPSC lines (http://www.ipscell.com/2015/07/firstipscstop/). This next-generation iPSC-derived approach is therefore still fraught with uncertainty in the absence of a regulatory framework or guidance about “allowable” levels of mutations and methods of their detection in iPSC products.

Cell-free approaches

A general consensus is that first-generation cells may exert their effects on tissue repair by secretion of paracrine factors. These largely unknown factors may stimulate the myocardium via myocyte salvage, induction of angiogenesis or stimulation of myocyte division. Although the second-generation cells, e.g. CPCs/CSCs and iPSC-derived cardiomyocytes, have been suggested (CSCs) or proven (pluripotent cells) in preclinical trials to have greater regenerative capacity because of their ability to form new myocardium, the contribution of remuscularization vs. “paracrine effects” to overall efficacy has not been demonstrated clinically nor preclinically. Any effects of second-generation cells are thought to be mediated by paracrine mechanisms. Since functional improvement is not necessarily related to cell survival at least in experimental models,85,86 approaches have been developed to mimic the benefit of cell therapy without transplanting the cells. Trialists have attempted to reproduce stem cell-like paracrine effects in ST-elevation myocardial infarction patients using growth factors such as granulocyte-colony stimulating factor (G-CSF).87 All other novel developments in this area are at the preclinical stage. Cell-free strategies include stimulating endogenous repair, e.g. by promoting neovascularization or activating resident progenitor cells.88,89 Mediators of paracrine effects are thought to include growth factors (e.g. erythropoietin, G-CSF),90 episomes,91 and non-coding RNAs,92 mimicking the secretome of donor cells. These factors can also be combined by assembling them in different controlled release formulations, such as microbeads,93 large scaffolds, or injectable biomaterials.94 Recent developments for cell-free approaches that emanate from cells, such as these focus on secreted nano-sized vesicles, called extracellular vesicles, and include microvesicles and exosomes.95,96 These small lipid containing vesicles are capable of transferring proteins, mRNA, and miRNAs between cells, and therefore represent a way for intercellular communication and inducing cardiac repair.97 However, organ selectivity after systemic delivery or inadvertent systemic spread after IC or intramyocardial delivery of these nanoparticles remains unknown and a topic for further scrutiny.

In summary, although the superiority in cardiac repair of one type of cell compared with another has not yet been proven, BMCs continue to be the source of cells most often used in human clinical trials. In cardiac patients, direct comparative data between different cell types is still lacking since adequately powered, randomized clinical trials with head-to-head comparisons of different cell types have not yet been performed. Apart from the risk of immune rejection, which can potentially be circumvented using MSCs, allogeneic somatic/adult cells appear to be safe. To match reported levels of functional cardiac improvement, cell therapy without the cells via paracrine factors may be an interesting alternative. For functional improvement beyond current levels achieved via paracrine actions, new developments will be necessary for proper regeneration of lost tissue.

Critical issues on protocols for cell-based therapy

One major problem for cell therapy is the relatively poor levels of cell retention in the transplanted area, and this may not be limited to first-generation cells, but apply to all cell sources. In fact, ≤10% of injected cells remain at the targeted location. No cells survive when injected into the infarct scar, short-term engraftment is ∼8% regardless of injected cell dose in remote normal myocardium, and in the infarct border zone, the percent survival at 24 h decreases progressively from ∼8% to <1%.27,98

Strategies to improve cell coupling, differentiation, survival, and retention by target area preparation, cell modification, conjugation with biomaterials and/or tissue engineering, and cytoprotection pathways

To improve cell retention, several biomaterial-based approaches have been explored (e.g. hydrogels, cell sheets, prefabricated matrices, microspheres, and injectable nanomatrix).94,99–102 Decellularized extracellular matrices have been shown to promote the healing process via modulation of the host immune response and resistance to bacterial infections.103 An alternative approach, explored in animal models, is the implantation of engineered heart tissue made in vitro from cardiomyocytes and hydrogel.104 Another method is the use of bispecific antibodies that bind to the cells and recognize a cardiac-specific antigen that is only present in injured myocardium.105 Finally, homing can be improved by priming the target organ or tissue with specific treatments, such as extracorporeal shockwaves.106 Localized hypoxia, inflammation, excessive oxidative stress, lack of supporting cells, poor supply of nutrients, and fibrosis promote apoptosis or necrosis of the grafted cells. Thus, the efficiency of cell therapies might be improved by using genetic engineering tools, including overexpression of pro-survival genes (e.g. Akt, Pim-1 kinase, ERK1/2, HIF-1α, haeme-oxygenase 1, GATA4, heat shock protein 27, miRNA-1, myocardin, and protein kinase G1α) or angiogenesis-initiating genes (e.g. VEGF, MYDGF, fibroblast growth factor (FGF)-2, SDF-1, and PDGF) in the cells to be transplanted or by transplanting the cells together with pro-survival or pro-angiogenic factors.76,98,107–113 Interestingly, exposure of cells to sub-lethal hypoxia increased the tolerance of these cells to the harsh environment after transplantation.114 These preconditioned cells also showed increased differentiation, enhanced paracrine effects leading to increased trophic support, and improved homing to the lesion site.114 Transplantation of preconditioned cells helped to suppress inflammatory factors and immune responses, and promoted heart function.114 In addition, transient modulation of cell specification towards myogenic differentiation, e.g. via microRNAs, could also be beneficial in increasing the amount of myocardium. miR-1 and -499 are excellent candidates as they can enhance both differentiation in vitro95 and in vivo.115 Another approach to promote transplanted cell survival is to modulate the inflammatory environment (using TSG-6, IL-1 inhibitor).109,116 Finally, a significant barrier to the therapeutic use of most cell populations with the exception of ESCs and iPSCs, is their limited cardiac differentiation potential despite the use of “cardiogenic cocktails” (containing TGF-β1, BMP-4, activin A, retinoic acid, IGF-1, FGF-2, α-thrombin, and IL-6) and overexpression of cardiac transcription factors.117–119 In addition, these stem cells fail to electromechanically integrate.120 This limitation has been partially solved by overexpressing the two key proteins, N-cadherin, and connexin 43, but clinical translation remains to be fully investigated. In contrast, human PSCs can now be routinely differentiated with high efficiency (>80%) into cardiomyocytes.121 However, the cardiomyocyte populations may contain varying proportions of atrial, ventricular, and nodal cardiomyocytes.122,123 This is a critical issue as they have unique mechanical and electrical properties and thus, the implantation of a mixture of these cells harbors the risk of arrhythmias.124 In addition, all of these cardiomyocyte types are immature and beat spontaneously, another source of arrhythmogenic risk. Consequently, even though many protocols primarily give rise to ventricular-like cardiomyocytes, it is important to refine the differentiation protocols to produce pure populations of defined cardiomyocyte phenotype.125,126 In addition, robust irreversible cardiac lineage differentiation of all transplanted cells is critically important to avoid the formation of teratomas.

Stem cell rejuvenation

Aging or comorbidities may cause a reduction in the number and function of tissue-resident and circulating cells.63,127,128 Several proteins and signaling pathways have been identified that are capable of reverting the process of cell senescence, including Pim-1 kinase,121–124 NOTCH1,129–131 telomerase, and myocardin.132 Pim-1 kinase has anti-senescence and anti-apoptotic effects in CSCs as well as in MSCs.133 Activation of the NOTCH1 signaling pathway results in remarkable rejuvenation of satellite muscle cells associated with enhanced proliferation, increased telomere lengths, and decreased susceptibility to replicative senescence.129 The overexpression of telomerase and myocardin genes increases cell survival, proliferation, cardiomyogenic,132,134 and smooth muscle differentiation in vitro.135 After overexpression of genes encoding for “rejuvenating factors’ and in vitro expansion, genetically modified cells may secrete high amounts of the “regenerating factor”, either transiently or permanently, at the transplantation site.107,136 Most of the approaches for genetic modification of cells requires cell manipulation with some risk of cell contamination, accumulation of mutations during in vitro culture, or insertional activation of other genes, due to the use of viral vectors. Taken together, the design of new protocols for aged cell rejuvenation would allow improved cell preparation and clinical application of cells in older patient populations, taking into account the safety problem of the genetic manipulation of cells.

Enhancing endogenous cardiac regeneration

Recent studies have demonstrated that cardiomyocyte turnover occurs throughout life in mammals, including humans.137–141 While the estimated rate of human cardiomyocyte renewal is controversial, most studies report an annual turnover rate of 1%, which increases after injury. However, the intrinsic capability in humans to regenerate injured myocardium after massive ischaemic cell death is too low to be of functional relevance. It has been suggested that transplanted cells may exert their beneficial effects by secreting cytokines and growth factors promoting cardiomyocyte proliferation, recruitment and activation of CPCs, induction of vessel formation, reduction of fibrotic scars, and inhibition of apoptosis.142 In addition, modulation of macrophage and regulatory T-cell function can improve healing, repair, and regeneration.143,144 Another approach to enhance endogenous cardiac repair is the induction of cardiomyocyte proliferation, a mechanism described in neonatal mice, zebrafish and newts in response to injury,145 although never in adult mammals. However, cardiomyocyte regeneration after myocardial infarction may be promoted also by administration of FGF-1, p38 MAP kinase inhibitor,146 blocking the Hippo pathway or upregulating the downstream Hippo effector Yes-associated protein (Yap),147 activation of Erb-B2 Receptor Tyrosine Kinase 2 signaling, or application of the human Fstl1 protein via an epicardial patch.148 Yet, so far it is unclear whether the observed functional improvement is due to cardiomyocyte proliferation and generation of new cardiomyocytes.149

Recently, promising new directions have been explored that are promoting for in situ regeneration, in which cellular transplantation will become redundant. Hereby, fibroblasts are being directly converted into cardiomyocyte-like cells by using a combination of three cardiac developmental transcription factors, Gata4, Mef2c, and Tbx5.150 These reprogrammed cells express several cardiac-specific markers and exhibit spontaneous contractions. Still in its infancy and efficiency being low, this approach will potentially allow the conversion of extracelluar matrix producing cells—the myofibroblasts in the scarred area—into cardiomyocytes in vivo.151 Follow-up studies are exploring the use of different combinations of transcription factors,152,153 or efficiencies being improved via microRNA-mediated reprogramming.154

Cell-tracking systems

In vivo cell tracking involves either ‘direct’ physical labelling of cells by incubating them with a contrast agent, or ‘indirect’ genetic labelling by transfecting cells with a reporter gene construct. The position of, and signal from these labels can then be tracked using various imaging modalities, including clinical scanners, such as positron emission tomography (PET), single photon emission computed tomography (SPECT) and magnetic resonance imaging (MRI) (reviewed in155,156). All imaging modalities can provide information regarding short-term kinetics of transplanted cells and their effects on cardiac function, but are not capable to assess the long-term engraftment. Given its high anatomical resolution and safety profile allowing serial longitudinal evaluations, MRI has been commonly used to track cells in clinical trials.157 However, MRI might detect macrophages that ingest the marker after the cell (derivative) had lysed. Other limitation could be the loss of signals over the time related to cell death or cell division. Safety concerns regarding the effects of genetic manipulation of cells currently limit the use of genetically modified cells in clinical trials, and thus long-term cell tracking. However, combination approaches relying on the simultaneous co-registration of different imaging modalities (nuclear medicine combined with CT or MRI) might overcome the limitations of individual imaging techniques, and represent powerful tools to gain insight into the delivery, engraftment, survival, off-target, and possible adverse effects of transplanted stem and progenitor cells. Given the indispensable role of cell tracking in clinical trials, the feasibility of imaging should be included in preliminary proof of concept studies, and considered among inclusion or exclusion criteria, but will limit cell transfer studies to only a few centers that have access to multimodal imaging expertise.

Controls, data reproducibility, standardization issue, and data quality

Over the past few years, concerns have been increasingly voiced about experimental reproducibility across the whole biomedical research fields,158,159 not least in cell therapy. For example, a recent paper searching for errors in published cardiac clinical trials using autologous BMCs reported that the greatest enhancement of LVEF was described in those studies with the most discrepancies or errors in factual reporting.160 The pervasive risk of neglecting basic rules of clinical trial design in stem cell trials has been demonstrated in a recent review.161 On the other hand, phase II studies, where the aim is to prove efficacy should be designed to assess several primary end-points, which might include structural evaluations, cardiovascular physiological measurements, biomarkers, functional capacity, and quality of life.162

The choice of appropriate controls and methodological rigor may be more demanding in the field of cell therapy if, for example, the need for a myocardial biopsy to harvest autologous stem cells complicates double-blinding. A pragmatic alternative is to use a crossover study design, in which each patient is randomly assigned to a sequence of treatments. However, where reagents, such as cytokines are administered in conjunction with cells, a control group with cytokines alone should also be included. Another issue is the choice of the right placebo control, which, in some cell therapy trials, simply consisted of transparent saline solution which can be easily be distinguished visually from serum.

Standardization of cell isolation and processing procedures is highly desirable in order to facilitate comparisons between trials and to enable meta-analyses. Sex of donor-cells should also be clearly stated, as sex differences can affect the regenerative potential of transplanted cells. Finally, standardization of patient populations and stratifications should be attempted. It has been proposed that reference MSCs be developed to facilitate comparison between studies.163

In summary, cell-based therapies would benefit significantly from different protocols collectively referred to as cell enhancement, including possible priming of host tissue with cytokines to increase homing, preconditioning of transplanted cells, drugs and pro-survival factors, genetic engineering of cells, and the use of biomaterials. All of these strategies could contribute to improving cell retention and promote cell survival, proliferation, differentiation, and induction of neo-angiogenesis. Nevertheless, irrespective of cell enhancement, pilot studies to understand where the cells go by choosing the best tracking system in vivo, and adherence to well-established rules for the design of robust clinical trials, are minimum requirements for any cell protocol to assess actual effectiveness of cell-based clinical interventions.

Clinical trial design

Safety and ethical issues

The design of randomized controlled clinical trials that are able to ascertain the long-term safety of cell therapy can be challenging from an ethical perspective, and encompass issues related to:164 (i) public perception of cell therapy—heightened expectations may influence the patient's decision to participate in clinical studies with cell therapy and may also affect the randomization procedure, with a preference to be in the treatment arm of the study rather than in the control group; (ii) conflicts of interest—commercial interests may place pressure on researchers to investigate cell therapies which are not yet ready for clinical testing; (iii) risks vs. potential benefits—given the invasive nature and uncertainties surrounding cell therapy, the potential risks may be difficult to define, thereby making the consent procedure all the more challenging; (iv) choice of study outcome measure—there is a fine balance between choosing a surrogate endpoint which provides mechanistic insight, and clinically relevant endpoints.

Patient selection (co-morbidities and co-medications)

When considering efficacy of cell therapy, a better understanding of cell biology and the interaction between treatment and patient-specific cardiovascular risk factors, co-morbidities (such as age, gender, diabetes, hypertension, dyslipidaemia, smoking, depression, and psychological stress), and routine medications is required. All major co-morbidities and co-medications in patients with IHD are potential confounders of the efficacy of cell therapy, because they affect the quality of source cells as well as the response of host tissue to the transplanted cells.32,165,166 Autologous or allogeneic haematopoietic cell transplantation for haematologic diseases was the first type of cell therapy, the outcome of which was correlated with comorbidity indices.167 However, no data are available on comorbidity index or score systems to be used in clinical cell therapy trials in order to objectively and reproducibly assess the possible interference of pre-existing co-morbidities and co-medications with outcome.165 Recently, micro-array platforms identified global transcriptional and functional differences between CPCs/CSCs and BMCs. These tools can help examine the true functionality of stem cells and customize cellular therapy according to specific patient variability in terms of co-morbidities or age.168 In this regard, key points that should be considered are the following: (i) roughly equal stratification of patients into risk groups; (ii) the inclusion of possible confounders in the analyses of outcomes; (iii) evaluation of aging as a three-dimensional variable incorporating chronologic age (which is a poor predictor of cell therapy outcomes, probably due to a lack of data on organ dysfunctions,169), co-morbidities, physical function, nutritional, and cognitive status; (iv) developing useful prognostic biomarkers and co-morbidity index that could help understanding correlations between co-morbidities with either cell biology and host response before any cell therapy; (v) determining the transcriptional and functional variation of adult stem cells for autologous cell therapy, that could help assessing which cell populations are optimal for diverse aged patients with existing co-morbidities and co-medications.

In summary, careful attention must be given to a variety of factors (including age, gender, co-morbidities, concomitant medications, and any other cardiovascular risk factors) that may interfere with the regenerative potential of cell therapy in the setting of IHD and HF. The development of useful prognostic biomarkers and co-morbidity indexes could help to objectively assess the weight of these factors in both preclinical and clinical trials.

Clinically relevant delivery routes, cell dose, and timing of delivery

Catheter-based IC cell infusion using a perfusion balloon catheter during stop flow conditions is the mostly used delivery route in clinical trials, with the following drawbacks: (i) the potential non-selective distribution pattern of the transferred cells, with exclusion of infarcted and border area in the case of an occluded coronary artery; (ii) the need for the cells to transmigrate from the vessel lumen into the myocardium; (iii) the possible occurrence of microembolisms with subsequent myocardial dysfunction. Intravenous administration is limited by entrapment of the donor cells in the capillaries of the lungs. Direct myocardial injection during open chest surgery is the most precise and accurate type of delivery. Transcatheter TE cell injection through the femoral artery and the aortic valve is less invasive.

Direct comparison of IC and TE using electromechanical mapping guidance and surgical delivery of autologous Indium-oxine-labelled bone marrow-derived MSCs was studied in a chronic pig model of ischaemic cardiomyopathy, however, did not show any significant difference in cell delivery efficiency to the myocardium nor in differences in safety profile.170

Recently, retrograde bone marrow cell injection via coronary sinus infusion has been tested as safe and feasible in patients with either ischaemic or non-ischaemic HF.171

With respect to cell dose (reviewed in76), it should be noted that in the vast majority of pre-clinical and clinical studies, dosing has been non-systematic and empirically assessed, guided more by feasibility and accessibility rather than by intentional dosage optimization. This has contributed to the still open question of how many cells should be delivered in order to achieve clinical benefit. Although a dose–response curve is difficult to obtain when using autologous cells from individual patients, the relative number of cells with high functional activity, rather than the absolute number of cells, should be considered to describe the dose response. Mean absolute numbers of cells infused into the coronary circulation of patients with IHD and HF range from 1.2 × 107 to 2.05 ± 110 × 108 bone marrow cells and from 1 × 106 to 25 × 106 CSCs (reviewed in76).

The optimal timing of donor cell delivery also remains debated. Although no consensus has been reached, between 4169 and 8 days172 after AMI onset seemed to be the optimal time point for BMCs or circulating blood-derived progenitor cells delivery into an infarct-related coronary artery, based on limited homing studies and the time course of inflammatory responses after myocardial infarction.

How to assess the clinical benefit of cell therapy (including follow-up)

In the vast majority of trials, the primary endpoint has been the evaluation of left ventricular size and global systolic function before and after treatment (reviewed173). Small (if any) improvements of LVEF have been observed in cell-treated patients by two-dimensional echocardiography, MRI, left ventricular angiography, or radionuclide ventriculography performed at different time points and with different acquisition and analysis protocols (reviewed in173). Given the controversial outcomes of previous clinical trials, future studies should avoid imaging methodologies with poor reproducibility, should standardize timing of image acquisition and analysis protocols, and more comprehensively evaluate the potential benefits deriving from cell therapy. Indeed, implementation and standardization of other techniques, such as 3D echocardiography,174 strain/strain rates,175,176 tissue Doppler echocardiography,177,178 and MRI might be extremely helpful to identify more sensitive markers of cardiac improvement. It is important to emphasize that, at the present time, MRI currently provides the most accurate, comprehensive, and reproducible measurements of cardiac chamber dimensions, volumes, function, and infarct size compared with other techniques,179,180 and therefore should be performed in cell-treated patients enrolled in clinical trials whenever possible at baseline, after treatment, and during follow-up. In addition to MRI, myocardial viability should be determined by 18F-FDG PET assessing glucose metabolism, alone or in association with dobutamine stress echocardiography, since all studies using 18F-FDG have shown an improvement in myocardial viability,181,182 but this beneficial effect has not always been paralleled by an increase in contractile reserve.183 Furthermore, possible limitation of 18F-FDG PET is the radiation exposure of patients undergoing serial imaging. Finally, to precisely determine the effects of cell therapies on vasculogenesis, serial quantitative PET evaluations of global, and regional myocardial perfusion might be extremely valuable.20,182,184 Independent of the specific technology, centralized evaluation by independent, and blinded core labs should be standard.

In addition to the above endpoints, real, clinically relevant endpoints should also be used in future clinical trials, as, e.g. indicated in the BAMI trial that is focused on the effect of IC reinfusion of BMCs on all-cause mortality in AMI (NCT01569178). Although such trials need enough power and are costly, they are essential to demonstrate the net clinical benefit for patients. Additional standard tests that should be considered, include quality of life assessment, number of hospitalizations, 6 min walk tests, and death over several years' follow-up.

In summary, how patient selection takes place, what the best clinically relevant delivery routes are for cell administration, which cell dose and timing of delivery should be used and what clinical endpoint should be analyzed and by which method, are the most crucial aspects in clinical trials investigating the effects of cell therapy. Adequately powered large-scale clinical trials, taking into account all the possible safety and ethical issues, considering cell function as one of major predictors of successful cell therapy, and focusing on hard clinically meaningful endpoints, are mandatory to determine whether the observed functional improvement reported in some studies can be extended to others and indeed translates into increased survival and reduced morbidity.

Figure 1

Flowchart of experimental design starting from preclinical studies and ending to the human clinical trials.

Recommendations

In Figure 1, we provide a flowchart of experimental design starting from non-clinical studies and ending with the human clinical trials. To this translational pathway, we would like to make the following recommendations when assessing the clinical potential of conventional cell-based therapy, as well as novel strategies of cell enhancement for cardiac regeneration and repair in IHD and HF patients:

  • Conventional cell-based therapy have reported efficacy and safety in most experimental myocardial infarction models tested, including those in large animals, but in human clinical trials in IHD and HF patients, only safety of cell therapies has been shown. Therefore, future pre-clinical studies using cell-based therapies should be designed to address specific hypotheses on modes of delivery and mechanisms of efficacy, rather than safety and efficacy endpoints only;

  • Based on the expected clinical trial outcome, a careful selection of cell source is essential. Whereas first-generation cells might be useful for stimulation of endogenous repair mechanisms or angiogenic effects, second-generation cells truly aim at replacing damaged myocardium. A comparison of different cell types, or a combination of cell types in randomized clinical trials has not yet been performed, but are being planned in future trials of chronic ischaemic HF.

  • Assessing cell number and viability along with full cell characterization including cell function, should be done in every clinical trial;

  • Poor cell retention remains a major issue. To further boost both cellular and paracrine effects, effective carrier materials and/or engineering approaches or pre-treatment strategies of cells or target tissues should be further developed.

  • To maximize successful translation of novel cell enhancement strategies, it is of primary importance to ensure that the efficacy of preclinical studies is validated whenever possible in the presence of confounding factors, such as age and gender and common cardiovascular co-morbidities as well as their routine medications. Likewise, for clinical application of genetically manipulated cells, it is important to ensure the safety of the injected cells in terms of genetically and epigenetically stability, efficient and reproducible differentiation, and highly reliable cell purification;

  • The usage of hard clinically meaningful endpoints is mandatory to determine whether functional improvement indeed translates into increased survival and reduced morbidity.

Conclusions

The early promise of cell therapy has not yet been fulfilled. First-generation cells and their secretomes that aim at myocardial salvage and stimulating the endogenous repair mechanisms of the heart through pro-angiogenic or prosurvival activity should be carefully selected depending on the desired effect. Second-generation cells such as pluripotent stem cells are indisputably capable of forming beating contractile cardiomyocytes, but large surviving grafts of injected cells are rarely observed.185 Combining these cell types with biomaterials may enhance the outcome of present cardiac cell transplantation therapy, by truly replacing the damaged myocardium with muscular grafts. Other strategies to empower the donor cells, referred to as cell enhancement, may further stimulate paracrine effects, but new developments will be necessary to achieve cardiac regeneration, e.g. by stimulating endogenous cardiac regeneration. Moreover, the selection of appropriate clinical endpoints, patient population, and delivery strategies are crucial aspects to understand the clinical effects. Furthermore, focusing on hard clinical endpoints in future cell-based trials is mandatory to determine whether any observed functional improvement translates into increased survival and reduced morbidity.

Funding

This work was supported by Cariplo (Rif. 2011-0566) and the Italian Ministry of Research (PRIN 2010YK7Z5K_007) (to R.M. and Prof. Raffaele De Caterina); by Veni (ZonMW) 91612147 and Netherlands Heart Foundation 2013T056 (to L.W.V.L.); by the NIHR Biomedical Research Centre (BRC233/CM/SD/101320), British Heart Foundation (PG/15/52/31598), and Medical Research Council (MR/K002066/1) (to S.M.D.); by the Italian Ministry of Health (GR-2009-1596220) and the Italian Ministry of Research (RBFR124FEN) (to C.P.); by ZonMW (91612147) and Netherlands Heart Foundation (2013T056) (to L.v.L.); by the Emerging Fields Initiative (EFI) for Cell Cycle in Disease and Regeneration (CYDER) from the Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU) (Germany) (to F.B.E.); by the German Centre for Cardiovascular Research (DZHK) and the German Ministry of Education and Research (BMBF), the German Research Foundation (DFG Es 88/12-1), the European Research Council (ERC AG IndivuHeart), and the European Commission (FP7 Biodesign) (to T.E.); by the Netherlands CardioVascular Research Initiative (CVON), the Dutch Heart Foundation, Dutch Federation of University Medical Centers, the Netherlands Organization for Health Research and Development, and the Royal Netherlands Academy of Sciences (to J.P.G.S.); by Norwegian Council on Cardiovascular Diseases (to K.Y.), by National Heart, Lung and Blood Institute NHLBI, rif. UM1 HL087365-10 (to J.T.W.), and by H2020-SMEINST-1-2015 “Infarnosys” (to Pharmahungary). P.F. was a Szentágothai Fellow (TÁMOP-4.2.4.A/2-11/1-2012-0001) of the National Excellence Program of Hungary.

Conflict of interest: T.E. and C.L.M. are co-founders of EHT Technologies GmbH, Hamburg and Pluriomics BV, respectively. P.F. is a founder and CEO of Pharmahungary Group.

References

1

Baker
M
.
Stem cells by any other name
.
Nature
2007
;
449
:
389
.

2

Bolli
R
,
Chugh
AR
,
D'Amario
D
,
Loughran
JH
,
Stoddard
MF
,
Ikram
S
,
Beache
GM
,
Wagner
SG
,
Leri
A
,
Hosoda
T
,
Sanada
F
,
Elmore
JB
,
Goichberg
P
,
Cappetta
D
,
Solankhi
NK
,
Fahsah
I
,
Rokosh
DG
,
Slaughter
MS
,
Kajstura
J
,
Anversa
P
.
Cardiac stem cells in patients with ischaemic cardiomyopathy (SCIPIO): initial results of a randomised phase 1 trial
.
Lancet
2011
;
378
:
1847
1857
.

3

Messina
E
,
De Angelis
L
,
Frati
G
,
Morrone
S
,
Chimenti
S
,
Fiordaliso
F
,
Salio
M
,
Battaglia
M
,
Latronico
MV
,
Coletta
M
,
Vivarelli
E
,
Frati
L
,
Cossu
G
,
Giacomello
A
.
Isolation and expansion of adult cardiac stem cells from human and murine heart
.
Circ Res
2004
;
95
:
911
921
.

4

Boland
MJ
,
Nazor
KL
,
Loring
JF
.
Epigenetic regulation of pluripotency and differentiation
.
Circ Res
2014
;
115
:
311
324
.

5

Basile
DP
,
Yoder
MC
.
Circulating and tissue resident endothelial progenitor cells
.
J Cell Physiol
2014
;
229
:
10
16
.

6

Horwitz
EM
,
Le Blanc
K
,
Dominici
M
,
Mueller
I
,
Slaper-Cortenbach
I
,
Marini
FC
,
Deans
RJ
,
Krause
DS
,
Keating
A
.
Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement
.
Cytotherapy
2005
;
7
:
393
395
.

7

Keating
A
.
Mesenchymal stromal cells
.
Curr Opin Hematol
2006
;
13
:
419
425
.

8

Nichols
M
,
Townsend
N
,
Scarborough
P
,
Rayner
M
.
Cardiovascular disease in Europe: epidemiological update
.
Eur Heart J
2013
;
34
:
3028
3034
.

9

Mnozanffarian
D
,
Benjamin
EJ
,
Go
AS
,
Arnett
DK
,
Blaha
MJ
,
Cushman
M
,
de Ferranti
S
,
Despres
JP
,
Fullerton
HJ
,
Howard
VJ
,
Huffman
MD
,
Judd
SE
,
Kissela
BM
,
Lackland
DT
,
Lichtman
JH
,
Lisabeth
LD
,
Liu
S
,
Mackey
RH
,
Matchar
DB
,
McGuire
DK
,
Mohler
ER
3rd
,
Moy
CS
,
Muntner
P
,
Mussolino
ME
,
Nasir
K
,
Neumar
RW
,
Nichol
G
,
Palaniappan
L
,
Pandey
DK
,
Reeves
MJ
,
Rodriguez
CJ
,
Sorlie
PD
,
Stein
J
,
Towfighi
A
,
Turan
TN
,
Virani
SS
,
Willey
JZ
,
Woo
D
,
Yeh
RW
,
Turner
MB
.
Heart disease and stroke statistics – 2015 update: a report from the American Heart Association
.
Circulation
2015
;
131
:
e29
322
.

10

Perin
EC
,
Borow
KM
,
Silva
GV
,
DeMaria
AN
,
Marroquin
OC
,
Huang
P
,
Traverse
JH
,
Krum
H
,
Skerrett
D
,
Zheng
Y
,
Willerson
JT
,
Itescu
S
,
Henry
TD
.
A Phase II dose-escalation study of allogeneic mesenchymal precursor cells in patients with ischemic or non-ischemic heart failure
.
Circ Res
2015
;
117
:
576
584
.

11

Vrtovec
B
,
Poglajen
G
,
Lezaic
L
,
Sever
M
,
Domanovic
D
,
Cernelc
P
,
Socan
A
,
Schrepfer
S
,
Torre-Amione
G
,
Haddad
F
,
Wu
JC
.
Effects of intracoronary CD34+ stem cell transplantation in nonischemic dilated cardiomyopathy patients: 5-year follow-up
.
Circ Res
2013
;
112
:
165
173
.

12

Schachinger
V
,
Erbs
S
,
Elsasser
A
,
Haberbosch
W
,
Hambrecht
R
,
Holschermann
H
,
Yu
J
,
Corti
R
,
Mathey
DG
,
Hamm
CW
,
Suselbeck
T
,
Werner
N
,
Haase
J
,
Neuzner
J
,
Germing
A
,
Mark
B
,
Assmus
B
,
Tonn
T
,
Dimmeler
S
,
Zeiher
AM
.
Improved clinical outcome after intracoronary administration of bone-marrow-derived progenitor cells in acute myocardial infarction: final 1-year results of the REPAIR-AMI trial
.
Eur Heart J
2006
;
27
:
2775
2783
.

13

Vrtovec
B
,
Poglajen
G
,
Sever
M
,
Lezaic
L
,
Domanovic
D
,
Cernelc
P
,
Haddad
F
,
Torre-Amione
G
.
Effects of intracoronary stem cell transplantation in patients with dilated cardiomyopathy
.
J Card Fail
2011
;
17
:
272
281
.

14

Wollert
KC
,
Meyer
GP
,
Lotz
J
,
Ringes-Lichtenberg
S
,
Lippolt
P
,
Breidenbach
C
,
Fichtner
S
,
Korte
T
,
Hornig
B
,
Messinger
D
,
Arseniev
L
,
Hertenstein
B
,
Ganser
A
,
Drexler
H
.
Intracoronary autologous bone-marrow cell transfer after myocardial infarction: the BOOST randomised controlled clinical trial
.
Lancet
2004
;
364
:
141
148
.

15

Lunde
K
,
Solheim
S
,
Aakhus
S
,
Arnesen
H
,
Abdelnoor
M
,
Egeland
T
,
Endresen
K
,
Ilebekk
A
,
Mangschau
A
,
Fjeld
JG
,
Smith
HJ
,
Taraldsrud
E
,
Grogaard
HK
,
Bjornerheim
R
,
Brekke
M
,
Muller
C
,
Hopp
E
,
Ragnarsson
A
,
Brinchmann
JE
,
Forfang
K
.
Intracoronary injection of mononuclear bone marrow cells in acute myocardial infarction
.
N Engl J Med
2006
;
355
:
1199
1209
.

16

Makkar
RR
,
Smith
RR
,
Cheng
K
,
Malliaras
K
,
Thomson
LE
,
Berman
D
,
Czer
LS
,
Marban
L
,
Mendizabal
A
,
Johnston
PV
,
Russell
SD
,
Schuleri
KH
,
Lardo
AC
,
Gerstenblith
G
,
Marban
E
.
Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase 1 trial
.
Lancet
2012
;
379
:
895
904
.

17

Meyer
GP
,
Wollert
KC
,
Lotz
J
,
Steffens
J
,
Lippolt
P
,
Fichtner
S
,
Hecker
H
,
Schaefer
A
,
Arseniev
L
,
Hertenstein
B
,
Ganser
A
,
Drexler
H
.
Intracoronary bone marrow cell transfer after myocardial infarction: eighteen months’ follow-up data from the randomized, controlled BOOST (BOne marrOw transfer to enhance ST-elevation infarct regeneration) trial
.
Circulation
2006
;
113
:
1287
1294
.

18

Huikuri
HV
,
Kervinen
K
,
Niemela
M
,
Ylitalo
K
,
Saily
M
,
Koistinen
P
,
Savolainen
ER
,
Ukkonen
H
,
Pietila
M
,
Airaksinen
JK
,
Knuuti
J
,
Makikallio
TH
.
Effects of intracoronary injection of mononuclear bone marrow cells on left ventricular function, arrhythmia risk profile, and restenosis after thrombolytic therapy of acute myocardial infarction
.
Eur Heart J
2008
;
29
:
2723
2732
.

19

Tendera
M
,
Wojakowski
W
,
Ruzyllo
W
,
Chojnowska
L
,
Kepka
C
,
Tracz
W
,
Musialek
P
,
Piwowarska
W
,
Nessler
J
,
Buszman
P
,
Grajek
S
,
Breborowicz
P
,
Majka
M
,
Ratajczak
MZ
.
Intracoronary infusion of bone marrow-derived selected CD34+CXCR4+ cells and non-selected mononuclear cells in patients with acute STEMI and reduced left ventricular ejection fraction: results of randomized, multicentre Myocardial Regeneration by Intracoronary Infusion of Selected Population of Stem Cells in Acute Myocardial Infarction (REGENT) Trial
.
Eur Heart J
2009
;
30
:
1313
1321
.

20

Janssens
S
,
Dubois
C
,
Bogaert
J
,
Theunissen
K
,
Deroose
C
,
Desmet
W
,
Kalantzi
M
,
Herbots
L
,
Sinnaeve
P
,
Dens
J
,
Maertens
J
,
Rademakers
F
,
Dymarkowski
S
,
Gheysens
O
,
Van Cleemput
J
,
Bormans
G
,
Nuyts
J
,
Belmans
A
,
Mortelmans
L
,
Boogaerts
M
,
Van de Werf
F
.
Autologous bone marrow-derived stem-cell transfer in patients with ST-segment elevation myocardial infarction: double-blind, randomised controlled trial
.
Lancet
2006
;
367
:
113
121
.

21

Hirsch
A
,
Nijveldt
R
,
van der Vleuten
PA
,
Tijssen
JG
,
van der Giessen
WJ
,
Tio
RA
,
Waltenberger
J
,
ten Berg
JM
,
Doevendans
PA
,
Aengevaeren
WR
,
Zwaginga
JJ
,
Biemond
BJ
,
van Rossum
AC
,
Piek
JJ
,
Zijlstra
F
.
Intracoronary infusion of mononuclear cells from bone marrow or peripheral blood compared with standard therapy in patients after acute myocardial infarction treated by primary percutaneous coronary intervention: results of the randomized controlled HEBE trial
.
Eur Heart J
2010
;
32
:
1736
1747
.

22

Wohrle
J
,
Merkle
N
,
Mailander
V
,
Nusser
T
,
Schauwecker
P
,
von Scheidt
F
,
Schwarz
K
,
Bommer
M
,
Wiesneth
M
,
Schrezenmeier
H
,
Hombach
V
.
Results of intracoronary stem cell therapy after acute myocardial infarction
.
Am J Cardiol
2010
;
105
:
804
812
.

23

Martin-Rendon
E
,
Brunskill
SJ
,
Hyde
CJ
,
Stanworth
SJ
,
Mathur
A
,
Watt
SM
.
Autologous bone marrow stem cells to treat acute myocardial infarction: a systematic review
.
Eur Heart J
2008
;
29
:
1807
1818
.

24

Templin
C
,
Kotlarz
D
,
Marquart
F
,
Faulhaber
J
,
Brendecke
V
,
Schaefer
A
,
Tsikas
D
,
Bonda
T
,
Hilfiker-Kleiner
D
,
Ohl
L
,
Naim
HY
,
Foerster
R
,
Drexler
H
,
Limbourg
FP
.
Transcoronary delivery of bone marrow cells to the infarcted murine myocardium: feasibility, cellular kinetics, and improvement in cardiac function
.
Basic Res Cardiol
2006
;
101
:
301
310
.

25

Hofmann
M
,
Wollert
KC
,
Meyer
GP
,
Menke
A
,
Arseniev
L
,
Hertenstein
B
,
Ganser
A
,
Knapp
WH
,
Drexler
H
.
Monitoring of bone marrow cell homing into the infarcted human myocardium
.
Circulation
2005
;
111
:
2198
2202
.

26

Blocklet
D
,
Toungouz
M
,
Berkenboom
G
,
Lambermont
M
,
Unger
P
,
Preumont
N
,
Stoupel
E
,
Egrise
D
,
Degaute
JP
,
Goldman
M
,
Goldman
S
.
Myocardial homing of nonmobilized peripheral-blood CD34+ cells after intracoronary injection
.
Stem Cells
2006
;
24
:
333
336
.

27

Hou
D
,
Youssef
EA
,
Brinton
TJ
,
Zhang
P
,
Rogers
P
,
Price
ET
,
Yeung
AC
,
Johnstone
BH
,
Yock
PG
,
March
KL
.
Radiolabeled cell distribution after intramyocardial, intracoronary, and interstitial retrograde coronary venous delivery: implications for current clinical trials
.
Circulation
2005
;
112
(9 Suppl.)
:
I150
I156
.

28

Passier
R
,
Mummery
C
.
Cardiomyocyte differentiation from embryonic and adult stem cells
.
Curr Opin Biotechnol
2005
;
16
:
498
502
.

29

Smart
N
,
Riley
PR
.
The stem cell movement
.
Circ Res
2008
;
102
:
1155
1168
.

30

Pijnappels
DA
,
Gregoire
S
,
Wu
SM
.
The integrative aspects of cardiac physiology and their implications for cell-based therapy
.
Ann N Y Acad Sci
2010
;
1188
:
7
14
.

31

Afzal
MR
,
Samanta
A
,
Shah
ZI
,
Jeevanantham
V
,
Abdel-Latif
A
,
Zuba-Surma
EK
,
Dawn
B
.
Adult bone marrow cell therapy for ischemic heart disease: evidence and insights from randomized controlled trials
.
Circ Res
2015
;
117
:
558
575
.

32

Lecour
S
,
Botker
HE
,
Condorelli
G
,
Davidson
SM
,
Garcia-Dorado
D
,
Engel
FB
,
Ferdinandy
P
,
Heusch
G
,
Madonna
R
,
Ovize
M
,
Ruiz-Meana
M
,
Schulz
R
,
Sluijter
JP
,
Van Laake
LW
,
Yellon
DM
,
Hausenloy
DJ
.
ESC working group cellular biology of the heart: position paper: improving the preclinical assessment of novel cardioprotective therapies
.
Cardiovasc Res
2014
;
104
:
399
411
.

33

Zhang
S
,
Sun
A
,
Xu
D
,
Yao
K
,
Huang
Z
,
Jin
H
,
Wang
K
,
Zou
Y
,
Ge
J
.
Impact of timing on efficacy and safety of intracoronary autologous bone marrow stem cells transplantation in acute myocardial infarction: a pooled subgroup analysis of randomized controlled trials
.
Clin Cardiol
2009
;
32
:
458
466
.

34

Yao
K
,
Huang
RC
,
Ge
L
,
Qian
JY
,
Li
YL
,
Xu
SK
,
Zhang
F
,
Zhang
YQ
,
Niu
YH
,
Shi
JH
,
Zhang
SH
,
Fan
B
,
Wang
QB
,
Sun
AJ
,
Zou
YZ
,
Ge
JB
.
[Observation on the safety: clinical trial on intracoronary autologous bone marrow mononuclear cells transplantation for acute myocardial infarction]
.
Zhonghua Xin Xue Guan Bing Za Zhi
2006
;
34
:
577
581
.

35

Traverse
JH
,
Henry
TD
,
Ellis
SG
,
Pepine
CJ
,
Willerson
JT
,
Zhao
DX
,
Forder
JR
,
Byrne
BJ
,
Hatzopoulos
AK
,
Penn
MS
,
Perin
EC
,
Baran
KW
,
Chambers
J
,
Lambert
C
,
Raveendran
G
,
Simon
DI
,
Vaughan
DE
,
Simpson
LM
,
Gee
AP
,
Taylor
DA
,
Cogle
CR
,
Thomas
JD
,
Silva
GV
,
Jorgenson
BC
,
Olson
RE
,
Bowman
S
,
Francescon
J
,
Geither
C
,
Handberg
E
,
Smith
DX
,
Baraniuk
S
,
Piller
LB
,
Loghin
C
,
Aguilar
D
,
Richman
S
,
Zierold
C
,
Bettencourt
J
,
Sayre
SL
,
Vojvodic
RW
,
Skarlatos
SI
,
Gordon
DJ
,
Ebert
RF
,
Kwak
M
,
Moye
LA
,
Simari
RD
.
Effect of intracoronary delivery of autologous bone marrow mononuclear cells 2 to 3 weeks following acute myocardial infarction on left ventricular function: the LateTIME randomized trial
.
J Am Med Assoc
2011
;
306
:
2110
2119
.

36

Perin
EC
,
Willerson
JT
,
Pepine
CJ
,
Henry
TD
,
Ellis
SG
,
Zhao
DX
,
Silva
GV
,
Lai
D
,
Thomas
JD
,
Kronenberg
MW
,
Martin
AD
,
Anderson
RD
,
Traverse
JH
,
Penn
MS
,
Anwaruddin
S
,
Hatzopoulos
AK
,
Gee
AP
,
Taylor
DA
,
Cogle
CR
,
Smith
D
,
Westbrook
L
,
Chen
J
,
Handberg
E
,
Olson
RE
,
Geither
C
,
Bowman
S
,
Francescon
J
,
Baraniuk
S
,
Piller
LB
,
Simpson
LM
,
Loghin
C
,
Aguilar
D
,
Richman
S
,
Zierold
C
,
Bettencourt
J
,
Sayre
SL
,
Vojvodic
RW
,
Skarlatos
SI
,
Gordon
DJ
,
Ebert
RF
,
Kwak
M
,
Moye
LA
,
Simari
RD
.
Effect of transendocardial delivery of autologous bone marrow mononuclear cells on functional capacity, left ventricular function, and perfusion in chronic heart failure: the FOCUS-CCTRN trial
.
J Am Med Assoc
2012
;
307
:
1717
1726
.

37

Perin
EC
,
Silva
GV
,
Henry
TD
,
Cabreira-Hansen
MG
,
Moore
WH
,
Coulter
SA
,
JP
Herlihy
,
Fernandes
MR
,
Cheong
BY
,
Flamm
SD
,
Traverse
JH
,
Zheng
Y
,
Smith
D
,
Shaw
S
,
Westbrook
L
,
Olson
R
,
Patel
D
,
Gahremanpour
A
,
Canales
J
,
Vaughn
WK
,
Willerson
JT
.
A randomized study of transendocardial injection of autologous bone marrow mononuclear cells and cell function analysis in ischemic heart failure (FOCUS-HF)
.
Am Heart J
2011
;
161
:
1078
1087
e1073
.

38

Flores-Ramirez
R
,
Uribe-Longoria
A
,
Rangel-Fuentes
MM
,
Gutierrez-Fajardo
P
,
Salazar-Riojas
R
,
Cervantes-Garcia
D
,
Trevino-Ortiz
JH
,
Benavides-Chereti
GJ
,
Espinosa-Oliveros
LP
,
Limon-Rodriguez
RH
,
Monreal-Puente
R
,
Gonzalez-Trevino
JL
,
Rojas-Martinez
A
.
Intracoronary infusion of CD133+ endothelial progenitor cells improves heart function and quality of life in patients with chronic post-infarct heart insufficiency
.
Cardiovasc Revasc Med
2010
;
11
:
72
78
.

39

Taljaard
M
,
Ward
MR
,
Kutryk
MJ
,
Courtman
DW
,
Camack
NJ
,
Goodman
SG
,
Parker
TG
,
Dick
AJ
,
Galipeau
J
,
Stewart
DJ
.
Rationale and design of Enhanced Angiogenic Cell Therapy in Acute Myocardial Infarction (ENACT-AMI): the first randomized placebo-controlled trial of enhanced progenitor cell therapy for acute myocardial infarction
.
Am Heart J
2010
;
159
:
354
360
.

40

Perin
EC
,
Sanz-Ruiz
R
,
Sanchez
PL
,
Lasso
J
,
Perez-Cano
R
,
Alonso-Farto
JC
,
Perez-David
E
,
Fernandez-Santos
ME
,
Serruys
PW
,
Duckers
HJ
,
Kastrup
J
,
Chamuleau
S
,
Zheng
Y
,
Silva
GV
,
Willerson
JT
,
Fernandez-Aviles
F
.
Adipose-derived regenerative cells in patients with ischemic cardiomyopathy: the PRECISE Trial
.
Am Heart J
2014
;
168
:
88
95
e82
.

41

Hierlihy
AM
,
Seale
P
,
Lobe
CG
,
Rudnicki
MA
,
Megeney
LA
.
The post-natal heart contains a myocardial stem cell population
.
FEBS Lett
2002
;
530
:
239
243
.

42

Martin
CM
,
Meeson
AP
,
Robertson
SM
,
Hawke
TJ
,
Richardson
JA
,
Bates
S
,
Goetsch
SC
,
Gallardo
TD
,
Garry
DJ
.
Persistent expression of the ATP-binding cassette transporter, Abcg2, identifies cardiac SP cells in the developing and adult heart
.
Dev Biol
2004
;
265
:
262
275
.

43

Pfister
O
,
Mouquet
F
,
Jain
M
,
Summer
R
,
Helmes
M
,
Fine
A
,
Colucci
WS
,
Liao
R
.
CD31- but Not CD31+ cardiac side population cells exhibit functional cardiomyogenic differentiation
.
Circ Res
2005
;
97
:
52
61
.

44

Zhou
B
,
Ma
Q
,
Rajagopal
S
,
Wu
SM
,
Domian
I
,
Rivera-Feliciano
J
,
Jiang
D
,
von Gise
A
,
Ikeda
S
,
Chien
KR
,
Pu
WT
.
Epicardial progenitors contribute to the cardiomyocyte lineage in the developing heart
.
Nature
2008
;
454
:
109
113
.

45

Smart
N
,
Bollini
S
,
Dube
KN
,
Vieira
JM
,
Zhou
B
,
Davidson
S
,
Yellon
D
,
Riegler
J
,
Price
AN
,
Lythgoe
MF
,
Pu
WT
,
Riley
PR
.
De novo cardiomyocytes from within the activated adult heart after injury
.
Nature
2011
;
474
:
640
644
.

46

Zhou
B
,
Honor
LB
,
Ma
Q
,
Oh
JH
,
Lin
RZ
,
Melero-Martin
JM
,
von Gise
A
,
Zhou
P
,
Hu
T
,
He
L
,
Wu
KH
,
Zhang
H
,
Zhang
Y
,
Pu
WT
.
Thymosin beta 4 treatment after myocardial infarction does not reprogram epicardial cells into cardiomyocytes
.
J Mol Cell Cardiol
2012
;
52
:
43
47
.

47

Bollini
S
,
Vieira
JM
,
Howard
S
,
Dube
KN
,
Balmer
GM
,
Smart
N
,
Riley
PR
.
Re-activated adult epicardial progenitor cells are a heterogeneous population molecularly distinct from their embryonic counterparts
.
Stem Cells Dev
2014
;
23
:
1719
1730
.

48

Cai
CL
,
Liang
X
,
Shi
Y
,
Chu
PH
,
Pfaff
SL
,
Chen
J
,
Evans
S
.
Isl1 identifies a cardiac progenitor population that proliferates prior to differentiation and contributes a majority of cells to the heart
.
Dev Cell
2003
;
5
:
877
889
.

49

Moretti
A
,
Caron
L
,
Nakano
A
,
Lam
JT
,
Bernshausen
A
,
Chen
Y
,
Qyang
Y
,
Bu
L
,
Sasaki
M
,
Martin-Puig
S
,
Sun
Y
,
Evans
SM
,
Laugwitz
KL
,
Chien
KR
.
Multipotent embryonic isl1+ progenitor cells lead to cardiac, smooth muscle, and endothelial cell diversification
.
Cell
2006
;
127
:
1151
1165
.

50

Laugwitz
KL
,
Moretti
A
,
Lam
J
,
Gruber
P
,
Chen
Y
,
Woodard
S
,
Lin
LZ
,
Cai
CL
,
Lu
MM
,
Reth
M
,
Platoshyn
O
,
Yuan
JX
,
Evans
S
,
Chien
KR
.
Postnatal isl1+ cardioblasts enter fully differentiated cardiomyocyte lineages
.
Nature
2005
;
433
:
647
653
.

51

Beltrami
AP
,
Barlucchi
L
,
Torella
D
,
Baker
M
,
Limana
F
,
Chimenti
S
,
Kasahara
H
,
Rota
M
,
Musso
E
,
Urbanek
K
,
Leri
A
,
Kajstura
J
,
Nadal-Ginard
B
,
Anversa
P
.
Adult cardiac stem cells are multipotent and support myocardial regeneration
.
Cell
2003
;
114
:
763
776
.

52

Zaruba
MM
,
Soonpaa
M
,
Reuter
S
,
Field
LJ
.
Cardiomyogenic potential of C-kit(+)-expressing cells derived from neonatal and adult mouse hearts
.
Circulation
2010
;
121
:
1992
2000
.

53

Jesty
SA
,
Steffey
MA
,
Lee
FK
,
Breitbach
M
,
Hesse
M
,
Reining
S
,
Lee
JC
,
Doran
RM
,
Nikitin
AY
,
Fleischmann
BK
,
Kotlikoff
MI
.
c-kit+ precursors support postinfarction myogenesis in the neonatal, but not adult, heart
.
Proc Natl Acad Sci USA
2012
;
109
:
13380
13385
.

54

van Berlo
JH
,
Kanisicak
O
,
Maillet
M
,
Vagnozzi
RJ
,
Karch
J
,
Lin
SC
,
Middleton
RC
,
Marban
E
,
Molkentin
JD
.
c-kit+ cells minimally contribute cardiomyocytes to the heart
.
Nature
2014
;
509
:
337
341
.

55

Goumans
MJ
,
de Boer
TP
,
Smits
AM
,
van Laake
LW
,
van Vliet
P
,
Metz
CH
,
Korfage
TH
,
Kats
KP
,
Hochstenbach
R
,
Pasterkamp
G
,
Verhaar
MC
,
van der Heyden
MA
,
de Kleijn
D
,
Mummery
CL
,
van Veen
TA
,
Sluijter
JP
,
Doevendans
PA
.
TGF-beta1 induces efficient differentiation of human cardiomyocyte progenitor cells into functional cardiomyocytes in vitro
.
Stem Cell Res
2007
;
1
:
138
149
.

56

Oh
H
,
Chi
X
,
Bradfute
SB
,
Mishina
Y
,
Pocius
J
,
Michael
LH
,
Behringer
RR
,
Schwartz
RJ
,
Entman
ML
,
Schneider
MD
.
Cardiac muscle plasticity in adult and embryo by heart-derived progenitor cells
.
Ann N Y Acad Sci
2004
;
1015
:
182
189
.

57

Leung
HW
,
Moerkamp
AT
,
Padmanabhan
J
,
Ng
SW
,
Goumans
MJ
,
Choo
A
.
mAb C19 targets a novel surface marker for the isolation of human cardiac progenitor cells from human heart tissue and differentiated hESCs
.
J Mol Cell Cardiol
2015
;
82
:
228
237
.

58

Chong
JJ
,
Yang
X
,
Don
CW
,
Minami
E
,
Liu
YW
,
Weyers
JJ
,
Mahoney
WM
,
Van Biber
B
,
Cook
SM
,
Palpant
NJ
,
Gantz
JA
,
Fugate
JA
,
Muskheli
V
,
Gough
GM
,
Vogel
KW
,
Astley
CA
,
Hotchkiss
CE
,
Baldessari
A
,
Pabon
L
,
Reinecke
H
,
Gill
EA
,
Nelson
V
,
Kiem
HP
,
Laflamme
MA
,
Murry
CE
.
Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts
.
Nature
2014
;
510
:
273
277
.

59

Mauritz
C
,
Schwanke
K
,
Reppel
M
,
Neef
S
,
Katsirntaki
K
,
Maier
LS
,
Nguemo
F
,
Menke
S
,
Haustein
M
,
Hescheler
J
,
Hasenfuss
G
,
Martin
U
.
Generation of functional murine cardiac myocytes from induced pluripotent stem cells
.
Circulation
2008
;
118
:
507
517
.

60

Narazaki
G
,
Uosaki
H
,
Teranishi
M
,
Okita
K
,
Kim
B
,
Matsuoka
S
,
Yamanaka
S
,
Yamashita
JK
.
Directed and systematic differentiation of cardiovascular cells from mouse induced pluripotent stem cells
.
Circulation
2008
;
118
:
498
506
.

61

Kawamura
M
,
Miyagawa
S
,
Miki
K
,
Saito
A
,
Fukushima
S
,
Higuchi
T
,
Kawamura
T
,
Kuratani
T
,
Daimon
T
,
Shimizu
T
,
Okano
T
,
Sawa
Y
.
Feasibility, safety, and therapeutic efficacy of human induced pluripotent stem cell-derived cardiomyocyte sheets in a porcine ischemic cardiomyopathy model
.
Circulation
2012
;
126
(11 Suppl. 1)
:
S29
S37
.

62

Kattman
SJ
,
Witty
AD
,
Gagliardi
M
,
Dubois
NC
,
Niapour
M
,
Hotta
A
,
Ellis
J
,
Keller
G
.
Stage-specific optimization of activin/nodal and BMP signaling promotes cardiac differentiation of mouse and human pluripotent stem cell lines
.
Cell Stem Cell
2011
;
8
:
228
240
.

63

Efimenko
AY
,
Kochegura
TN
,
Akopyan
ZA
,
Parfyonova
YV
.
Autologous stem cell therapy: how aging and chronic diseases affect stem and progenitor cells
.
BioResearch Open Access
2015
;
4.1
:
26
38
.

64

Delewi
R
,
Hirsch
A
,
Tijssen
JG
,
Schachinger
V
,
Wojakowski
W
,
Roncalli
J
,
Aakhus
S
,
Erbs
S
,
Assmus
B
,
Tendera
M
,
Goekmen Turan
R
,
Corti
R
,
Henry
T
,
Lemarchand
P
,
Lunde
K
,
Cao
F
,
Huikuri
HV
,
Surder
D
,
Simari
RD
,
Janssens
S
,
Wollert
KC
,
Plewka
M
,
Grajek
S
,
Traverse
JH
,
Zijlstra
F
,
Piek
JJ
.
Impact of intracoronary bone marrow cell therapy on left ventricular function in the setting of ST-segment elevation myocardial infarction: a collaborative meta-analysis
.
Eur Heart J
2014
;
35
:
989
998
.

65

Jeevanantham
V
,
Butler
M
,
Saad
A
,
Abdel-Latif
A
,
Zuba-Surma
EK
,
Dawn
B
.
Adult bone marrow cell therapy improves survival and induces long-term improvement in cardiac parameters: a systematic review and meta-analysis
.
Circulation
2012
;
126
:
551
568
.

66

Zimmet
H
,
Porapakkham
P
,
Sata
Y
,
Haas
SJ
,
Itescu
S
,
Forbes
A
,
Krum
H
.
Short- and long-term outcomes of intracoronary and endogenously mobilized bone marrow stem cells in the treatment of ST-segment elevation myocardial infarction: a meta-analysis of randomized control trials
.
Eur J Heart Fail
2012
;
14
:
91
105
.

67

Wong
M
,
Staszewsky
L
,
Latini
R
,
Barlera
S
,
Glazer
R
,
Aknay
N
,
Hester
A
,
Anand
I
,
Cohn
JN
.
Severity of left ventricular remodeling defines outcomes and response to therapy in heart failure: Valsartan heart failure trial (Val-HeFT) echocardiographic data
.
J Am Coll Cardiol
2004
;
43
:
2022
2027
.

68

Kramer
DG
,
Trikalinos
TA
,
Kent
DM
,
Antonopoulos
GV
,
Konstam
MA
,
Udelson
JE
.
Quantitative evaluation of drug or device effects on ventricular remodeling as predictors of therapeutic effects on mortality in patients with heart failure and reduced ejection fraction: a meta-analytic approach
.
J Am Coll Cardiol
2010
;
56
:
392
406
.

69

Ezekowitz
JA
,
McAlister
FA
.
Aldosterone blockade and left ventricular dysfunction: a systematic review of randomized clinical trials
.
Eur Heart J
2009
;
30
:
469
477
.

70

Cleland
JG
,
Pennell
DJ
,
Ray
SG
,
Coats
AJ
,
Macfarlane
PW
,
Murray
GD
,
Mule
JD
,
Vered
Z
,
Lahiri
A
.
Myocardial viability as a determinant of the ejection fraction response to carvedilol in patients with heart failure (CHRISTMAS trial): randomised controlled trial
.
Lancet
2003
;
362
:
14
21
.

71

Beitnes
JO
,
Gjesdal
O
,
Lunde
K
,
Solheim
S
,
Edvardsen
T
,
Arnesen
H
,
Forfang
K
,
Aakhus
S
.
Left ventricular systolic and diastolic function improve after acute myocardial infarction treated with acute percutaneous coronary intervention, but are not influenced by intracoronary injection of autologous mononuclear bone marrow cells: a 3 year serial echocardiographic sub-study of the randomized-controlled ASTAMI study
.
Eur J Echocardiogr
2011
;
12
:
98
106
.

72

Assmus
B
,
Leistner
DM
,
Schachinger
V
,
Erbs
S
,
Elsasser
A
,
Haberbosch
W
,
Hambrecht
R
,
Sedding
D
,
Yu
J
,
Corti
R
,
Mathey
DG
,
Barth
C
,
Mayer-Wehrstein
C
,
Burck
I
,
Sueselbeck
T
,
Dill
T
,
Hamm
CW
,
Tonn
T
,
Dimmeler
S
,
Zeiher
AM
.
Long-term clinical outcome after intracoronary application of bone marrow-derived mononuclear cells for acute myocardial infarction: migratory capacity of administered cells determines event-free survival
.
Eur Heart J
2014
;
35
:
1275
1283
.

73

Chimenti
I
,
Smith
RR
,
Li
TS
,
Gerstenblith
G
,
Messina
E
,
Giacomello
A
,
Marban
E
.
Relative roles of direct regeneration versus paracrine effects of human cardiosphere-derived cells transplanted into infarcted mice
.
Circ Res
2010
;
106
:
971
980
.

74

Hong
KU
,
Guo
Y
,
Li
QH
,
Cao
P
,
Al-Maqtari
T
,
Vajravelu
BN
,
Du
J
,
Book
MJ
,
Zhu
X
,
Nong
Y
,
Bhatnagar
A
,
Bolli
R
.
c-kit+ Cardiac stem cells alleviate post-myocardial infarction left ventricular dysfunction despite poor engraftment and negligible retention in the recipient heart
.
PLoS ONE
2014
;
9
:
e96725
.

75

Keith
MC
,
Bolli
R
.
“String theory” of c-kit(pos) cardiac cells: a new paradigm regarding the nature of these cells that may reconcile apparently discrepant results
.
Circ Res
2015
;
116
:
1216
1230
.

76

Madonna
R
,
Ferdinandy
P
,
De Caterina
R
,
Willerson
JT
,
Marian
AJ
.
Recent developments in cardiovascular stem cells
.
Circ Res
2014
;
115
:
e71
e78
.

77

van der Spoel
TIG
,
Gathier
WA
,
Koudstaal
S
,
van Slochteren
F
,
Jansen of Lorkeers
S
,
Sluijter
JPG
,
Hoefer
IE
,
Steendijk
P
,
Cramer
MJM
,
Doevendans
PA
,
van Belle
E
,
Chamuleau
SAJ
.
Autologous mesenchymal stem cells show more benefit on systolic function compared to bone marrow mononuclear cells in a porcine model of chronic myocardial infarction
.
J Cardiovasc Transl Res
2015
;
8
:
393
403
.

78

Li
TS
,
Cheng
K
,
Malliaras
K
,
Smith
RR
,
Zhang
Y
,
Sun
B
,
Matsushita
N
,
Blusztajn
A
,
Terrovitis
J
,
Kusuoka
H
,
Marban
L
,
Marban
E
.
Direct comparison of different stem cell types and subpopulations reveals superior paracrine potency and myocardial repair efficacy with cardiosphere-derived cells
.
J Am Coll Cardiol
2012
;
59
:
942
953
.

79

Gyongyosi
M
,
Wojakowski
W
,
Lemarchand
P
,
Lunde
K
,
Tendera
M
,
Bartunek
J
,
Marban
E
,
Assmus
B
,
Henry
TD
,
Traverse
JH
,
Moye
LA
,
Surder
D
,
Corti
R
,
Huikuri
H
,
Miettinen
J
,
Wohrle
J
,
Obradovic
S
,
Roncalli
J
,
Malliaras
K
,
Pokushalov
E
,
Romanov
A
,
Kastrup
J
,
Bergmann
MW
,
Atsma
DE
,
Diederichsen
A
,
Edes
I
,
Benedek
I
,
Benedek
T
,
Pejkov
H
,
Nyolczas
N
,
Pavo
N
,
Bergler-Klein
J
,
Pavo
IJ
,
Sylven
C
,
Berti
S
,
Navarese
EP
,
Maurer
G
.
Meta-Analysis of Cell-based CaRdiac stUdiEs (ACCRUE) in patients with acute myocardial infarction based on individual patient data
.
Circ Res
2015
;
116
:
1346
1360
.

80

Hare
JM
,
Fishman
JE
,
Gerstenblith
G
,
DiFede Velazquez
DL
,
Zambrano
JP
,
Suncion
VY
,
Tracy
M
,
Ghersin
E
,
Johnston
PV
,
Brinker
JA
,
Breton
E
,
Davis-Sproul
J
,
Schulman
IH
,
Byrnes
J
,
Mendizabal
AM
,
Lowery
MH
,
Rouy
D
,
Altman
P
,
Wong Po Foo
C
,
Ruiz
P
,
Amador
A
,
Da Silva
J
,
McNiece
IK
,
Heldman
AW
,
George
R
,
Lardo
A
.
Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial
.
J Am Med Assoc
2012
;
308
:
2369
2379
.

81

Jansen of Lorkeers
SJ
,
Eding
JE
,
Vesterinen
HM
,
van der Spoel
TI
,
Sena
ES
,
Duckers
HJ
,
Doevendans
PA
,
Macleod
MR
,
Chamuleau
SA
.
Similar effect of autologous and allogeneic cell therapy for ischemic heart disease: systematic review and meta-analysis of large animal studies
.
Circ Res
2015
;
116
:
80
86
.

82

Menasche
P
,
Vanneaux
V
,
Fabreguettes
JR
,
Bel
A
,
Tosca
L
,
Garcia
S
,
Bellamy
V
,
Farouz
Y
,
Pouly
J
,
Damour
O
,
Perier
MC
,
Desnos
M
,
Hagege
A
,
Agbulut
O
,
Bruneval
P
,
Tachdjian
G
,
Trouvin
JH
,
Larghero
J
.
Towards a clinical use of human embryonic stem cell-derived cardiac progenitors: a translational experience
.
Eur Heart J
2015
;
36
:
743
750
.

83

Menasche
P
,
Vanneaux
V
,
Hagege
A
,
Bel
A
,
Cholley
B
,
Cacciapuoti
I
,
Parouchev
A
,
Benhamouda
N
,
Tachdjian
G
,
Tosca
L
,
Trouvin
JH
,
Fabreguettes
JR
,
Bellamy
V
,
Guillemain
R
,
Suberbielle Boissel
C
,
Tartour
E
,
Desnos
M
,
Larghero
J
.
Human embryonic stem cell-derived cardiac progenitors for severe heart failure treatment: first clinical case report
.
Eur Heart J
2015
;
36
:
2011
2017
.

84

Schwartz
SD
,
Regillo
CD
,
Lam
BL
,
Eliott
D
,
Rosenfeld
PJ
,
Gregori
NZ
,
Hubschman
JP
,
Davis
JL
,
Heilwell
G
,
Spirn
M
,
Maguire
J
,
Gay
R
,
Bateman
J
,
Ostrick
RM
,
Morris
D
,
Vincent
M
,
Anglade
E
,
Del Priore
LV
,
Lanza
R
.
Human embryonic stem cell-derived retinal pigment epithelium in patients with age-related macular degeneration and Stargardt's macular dystrophy: follow-up of two open-label phase 1/2 studies
.
Lancet
2015
;
385
:
509
516
.

85

van Laake
LW
,
Passier
R
,
Doevendans
PA
,
Mummery
CL
.
Human embryonic stem cell-derived cardiomyocytes and cardiac repair in rodents
.
Circ Res
2008
;
102
:
1008
1010
.

86

Feyen
D
,
Gaetani
R
,
Liu
J
,
Noort
W
,
Martens
A
,
den Ouden
K
,
Doevendans
PA
,
Sluijter
JP
.
Increasing short-term cardiomyocyte progenitor cell (CMPC) survival by necrostatin-1 did not further preserve cardiac function
.
Cardiovasc Res
2013
;
99
:
83
91
.

87

Engelmann
MG
,
Theiss
HD
,
Hennig-Theiss
C
,
Huber
A
,
Wintersperger
BJ
,
Werle-Ruedinger
AE
,
Schoenberg
SO
,
Steinbeck
G
,
Franz
WM
.
Autologous bone marrow stem cell mobilization induced by granulocyte colony-stimulating factor after subacute ST-segment elevation myocardial infarction undergoing late revascularization: final results from the G-CSF-STEMI (Granulocyte Colony-Stimulating Factor ST-Segment Elevation Myocardial Infarction) trial
.
J Am Coll Cardiol
2006
;
48
:
1712
1721
.

88

Timmers
L
,
Lim
SK
,
Hoefer
IE
,
Arslan
F
,
Lai
RC
,
van Oorschot
AA
,
Goumans
MJ
,
Strijder
C
,
Sze
SK
,
Choo
A
,
Piek
JJ
,
Doevendans
PA
,
Pasterkamp
G
,
de Kleijn
DP
.
Human mesenchymal stem cell-conditioned medium improves cardiac function following myocardial infarction
.
Stem Cell Res
2011
;
6
:
206
214
.

89

Sluijter
JP
,
Condorelli
G
,
Davidson
SM
,
Engel
FB
,
Ferdinandy
P
,
Hausenloy
DJ
,
Lecour
S
,
Madonna
R
,
Ovize
M
,
Ruiz-Meana
M
,
Schulz
R
,
Van Laake
LW
.
Novel therapeutic strategies for cardioprotection
.
Pharmacol Ther
2014
;
144
:
60
70
.

90

Zafiriou
MP
,
Noack
C
,
Unsold
B
,
Didie
M
,
Pavlova
E
,
Fischer
HJ
,
Reichardt
HM
,
Bergmann
MW
,
El-Armouche
A
,
Zimmermann
WH
,
Zelarayan
LC
.
Erythropoietin responsive cardiomyogenic cells contribute to heart repair post myocardial infarction
.
Stem Cells
2014
;
32
:
2480
2491
.

91

Barile
L
,
Lionetti
V
,
Cervio
E
,
Matteucci
M
,
Gherghiceanu
M
,
Popescu
LM
,
Torre
T
,
Siclari
F
,
Moccetti
T
,
Vassalli
G
.
Extracellular vesicles from human cardiac progenitor cells inhibit cardiomyocyte apoptosis and improve cardiac function after myocardial infarction
.
Cardiovasc Res
2014
;
103
:
530
541
.

92

Emanueli
C
,
Shearn
AI
,
Angelini
GD
,
Sahoo
S
.
Exosomes and exosomal miRNAs in cardiovascular protection and repair
.
Vascul Pharmacol
2015
;
71
:
24
30
.

93

de Jong
R
,
van Hout
GP
,
Houtgraaf
JH
,
Kazemi
K
,
Wallrapp
C
,
Lewis
A
,
Pasterkamp
G
,
Hoefer
IE
,
Duckers
HJ
.
Intracoronary infusion of encapsulated glucagon-like peptide-1-eluting mesenchymal stem cells preserves left ventricular function in a porcine model of acute myocardial infarction
.
Circ Cardiovasc Interv
2014
;
7
:
673
683
.

94

Gaetani
R
,
Feyen
DA
,
Verhage
V
,
Slaats
R
,
Messina
E
,
Christman
KL
,
Giacomello
A
,
Doevendans
PA
,
Sluijter
JP
.
Epicardial application of cardiac progenitor cells in a 3D-printed gelatin/hyaluronic acid patch preserves cardiac function after myocardial infarction
.
Biomaterials
2015
;
61
:
339
348
.

95

Sluijter
JP
,
van Mil
A
,
van Vliet
P
,
Metz
CH
,
Liu
J
,
Doevendans
PA
,
Goumans
MJ
.
MicroRNA-1 and -499 regulate differentiation and proliferation in human-derived cardiomyocyte progenitor cells
.
Arterioscler Thromb Vasc Biol
2010
;
30
:
859
868
.

96

Yellon
DM
,
Davidson
SM
.
Exosomes: nanoparticles involved in cardioprotection?
Circ Res
2014
;
114
:
325
332
.

97

Sahoo
S
,
Losordo
DW
.
Exosomes and cardiac repair after myocardial infarction
.
Circ Res
2014
;
114
:
333
344
.

98

Hong
KU
,
Bolli
R
.
Cardiac stem cell therapy for cardiac repair
.
Curr Treat Options Cardiovasc Med
2014
;
16
:
324
.

99

Patra
C
,
Boccaccini
AR
,
Engel
FB
.
Vascularisation for cardiac tissue engineering: the extracellular matrix
.
Thromb Haemost
2015
;
113
:
532
547
.

100

Ban
K
,
Park
HJ
,
Kim
S
,
Andukuri
A
,
Cho
KW
,
Hwang
JW
,
Cha
HJ
,
Kim
SY
,
Kim
WS
,
Jun
HW
,
Yoon
YS
.
Cell therapy with embryonic stem cell-derived cardiomyocytes encapsulated in injectable nanomatrix gel enhances cell engraftment and promotes cardiac repair
.
ACS Nano
2014
;
8
:
10815
10825
.

101

Madonna
R
,
Petrov
L
,
Teberino
MA
,
Manzoli
L
,
Karam
JP
,
Renna
FV
,
Ferdinandy
P
,
Montero-Menei
CN
,
Yla-Herttuala
S
,
De Caterina
R
.
Transplantation of adipose tissue mesenchymal cells conjugated with VEGF-releasing microcarriers promotes repair in murine myocardial infarction
.
Cardiovasc Res
2015
;
108
:
39
49
.

102

Madonna
R
,
De Caterina
R
.
Stem cells and growth factor delivery systems for cardiovascular disease
.
J Biotechnol
2011
;
154
:
291
297
.

103

Piterina
AV
,
Cloonan
AJ
,
Meaney
CL
,
Davis
LM
,
Callanan
A
,
Walsh
MT
,
McGloughlin
TM
.
ECM-based materials in cardiovascular applications: Inherent healing potential and augmentation of native regenerative processes
.
Int J Mol Sci
2009
;
10
:
4375
4417
.

104

Zimmermann
WH
,
Melnychenko
I
,
Wasmeier
G
,
Didie
M
,
Naito
H
,
Nixdorff
U
,
Hess
A
,
Budinsky
L
,
Brune
K
,
Michaelis
B
,
Dhein
S
,
Schwoerer
A
,
Ehmke
H
,
Eschenhagen
T
.
Engineered heart tissue grafts improve systolic and diastolic function in infarcted rat hearts
.
Nat Med
2006
;
12
:
452
458
.

105

Zhao
TC
,
Tseng
A
,
Yano
N
,
Tseng
Y
,
Davol
PA
,
Lee
RJ
,
Lum
LG
,
Padbury
JF
.
Targeting human CD34+ hematopoietic stem cells with anti-CD45 x anti-myosin light-chain bispecific antibody preserves cardiac function in myocardial infarction
.
J Appl Physiol (1985)
2008
;
104
:
1793
1800
.

106

Aicher
A
,
Heeschen
C
,
Sasaki
K
,
Urbich
C
,
Zeiher
AM
,
Dimmeler
S
.
Low-energy shock wave for enhancing recruitment of endothelial progenitor cells: a new modality to increase efficacy of cell therapy in chronic hind limb ischemia
.
Circulation
2006
;
114
:
2823
2830
.

107

Siddiqi
S
,
Sussman
MA
.
Cell and gene therapy for severe heart failure patients: the time and place for Pim-1 kinase
.
Expert Rev Cardiovasc Ther
2013
;
11
:
949
957
.

108

Don
CW
,
Murry
CE
.
Improving survival and efficacy of pluripotent stem cell-derived cardiac grafts
.
J Cell Mol Med
2013
;
17
:
1355
1362
.

109

Karantalis
V
,
Hare
JM
.
Use of mesenchymal stem cells for therapy of cardiac disease
.
Circ Res
2015
;
116
:
1413
1430
.

110

Kim
J
,
Shapiro
L
,
Flynn
A
.
The clinical application of mesenchymal stem cells and cardiac stem cells as a therapy for cardiovascular disease
.
Pharmacol Ther
2015
;
151
:
8
15
.

111

Liu
XB
,
Wang
JA
,
Ji
XY
,
Yu
SP
,
Wei
L
.
Preconditioning of bone marrow mesenchymal stem cells by prolyl hydroxylase inhibition enhances cell survival and angiogenesis in vitro and after transplantation into the ischemic heart of rats
.
Stem Cell Res Ther
2014
;
5
:
111
.

112

Ni
NC
,
Li
RK
,
Weisel
RD
.
The promise and challenges of cardiac stem cell therapy
.
Semin Thorac Cardiovasc Surg
2014
;
26
:
44
52
.

113

Samper
E
,
Diez-Juan
A
,
Montero
JA
,
Sepulveda
P
.
Cardiac cell therapy: boosting mesenchymal stem cells effects
.
Stem Cell Rev
2013
;
9
:
266
280
.

114

Yu
SP
,
Wei
Z
,
Wei
L
.
Preconditioning strategy in stem cell transplantation therapy
.
Transl Stroke Res
2013
;
4
:
76
88
.

115

Hosoda
T
,
Zheng
H
,
Cabral-da-Silva
M
,
Sanada
F
,
Ide-Iwata
N
,
Ogorek
B
,
Ferreira-Martins
J
,
Arranto
C
,
D'Amario
D
,
del Monte
F
,
Urbanek
K
,
D'Alessandro
DA
,
Michler
RE
,
Anversa
P
,
Rota
M
,
Kajstura
J
,
Leri
A
.
Human cardiac stem cell differentiation is regulated by a mircrine mechanism
.
Circulation
2011
;
123
:
1287
1296
.

116

Forbes
SJ
,
Rosenthal
N
.
Preparing the ground for tissue regeneration: from mechanism to therapy
.
Nat Med
2014
;
20
:
857
869
.

117

Murry
CE
,
Soonpaa
MH
,
Reinecke
H
,
Nakajima
H
,
Nakajima
HO
,
Rubart
M
,
Pasumarthi
KB
,
Virag
JI
,
Bartelmez
SH
,
Poppa
V
,
Bradford
G
,
Dowell
JD
,
Williams
DA
,
Field
LJ
.
Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts
.
Nature
2004
;
428
:
664
668
.

118

Behfar
A
,
Yamada
S
,
Crespo-Diaz
R
,
Nesbitt
JJ
,
Rowe
LA
,
Perez-Terzic
C
,
Gaussin
V
,
Homsy
C
,
Bartunek
J
,
Terzic
A
.
Guided cardiopoiesis enhances therapeutic benefit of bone marrow human mesenchymal stem cells in chronic myocardial infarction
.
J Am Coll Cardiol
2015
;
56
:
721
734
.

119

Nunez Garcia
A
,
Sanz-Ruiz
R
,
Fernandez Santos
ME
,
Fernandez-Aviles
F
.
“Second-generation” stem cells for cardiac repair
.
World J Stem Cells
2015
;
7
:
352
367
.

120

Almeida
SO
,
Skelton
RJ
,
Adigopula
S
,
Ardehali
R
.
Arrhythmia in stem cell transplantation
.
Card Electrophysiol Clin
2015
;
7
:
357
370
.

121

Birket
MJ
,
Mummery
CL
.
Pluripotent stem cell derived cardiovascular progenitors--a developmental perspective
.
Dev Biol
2015
;
400
:
169
179
.

122

Sahara
M
,
Santoro
F
,
Chien
KR
.
Programming and reprogramming a human heart cell
.
EMBO J
2015
;
34
:
710
738
.

123

Weng
Z
,
Kong
CW
,
Ren
L
,
Karakikes
I
,
Geng
L
,
He
J
,
Chow
MZ
,
Mok
CF
,
Keung
W
,
Chow
H
,
Leung
AY
,
Hajjar
RJ
,
Li
RA
,
Chan
CW
.
A simple, cost-effective but highly efficient system for deriving ventricular cardiomyocytes from human pluripotent stem cells
.
Stem Cells Dev
2014
;
23
:
1704
1716
.

124

Blin
G
,
Nury
D
,
Stefanovic
S
,
Neri
T
,
Guillevic
O
,
Brinon
B
,
Bellamy
V
,
Rucker-Martin
C
,
Barbry
P
,
Bel
A
,
Bruneval
P
,
Cowan
C
,
Pouly
J
,
Mitalipov
S
,
Gouadon
E
,
Binder
P
,
Hagege
A
,
Desnos
M
,
Renaud
JF
,
Menasche
P
,
Puceat
M
.
A purified population of multipotent cardiovascular progenitors derived from primate pluripotent stem cells engrafts in postmyocardial infarcted nonhuman primates
.
J Clin Invest
2010
;
120
:
1125
1139
.

125

Devalla
HD
,
Schwach
V
,
Ford
JW
,
Milnes
JT
,
El-Haou
S
,
Jackson
C
,
Gkatzis
K
,
Elliott
DA
,
Chuva de Sousa Lopes
SM
,
Mummery
CL
,
Verkerk
AO
,
Passier
R
.
Atrial-like cardiomyocytes from human pluripotent stem cells are a robust preclinical model for assessing atrial-selective pharmacology
.
EMBO Mol Med
2015
;
7
:
394
410
.

126

Birket
MJ
,
Ribeiro
MC
,
Verkerk
AO
,
Ward
D
,
Leitoguinho
AR
,
den Hartogh
SC
,
Orlova
VV
,
Devalla
HD
,
Schwach
V
,
Bellin
M
,
Passier
R
,
Mummery
CL
.
Expansion and patterning of cardiovascular progenitors derived from human pluripotent stem cells
.
Nat Biotechnol
2015
;
33
:
970
979
.

127

Rauscher
FM
,
Goldschmidt-Clermont
PJ
,
Davis
BH
,
Wang
T
,
Gregg
D
,
Ramaswami
P
,
Pippen
AM
,
Annex
BH
,
Dong
C
,
Taylor
DA
.
Aging, progenitor cell exhaustion, and atherosclerosis
.
Circulation
2003
;
108
:
457
463
.

128

Madonna
R
,
Engel
FB
,
Davidson
SM
,
Ferdinandy
P
,
Gorbe
A
,
Sluijter
JP
,
Van Laake
LW
.
Stem cell aging and age-related cardiovascular disease: perspectives of treatment by ex-vivo stem cell rejuvenation
.
Curr Drug Targets
2015
;
16
:
780
785
.

129

Nemir
M
,
Metrich
M
,
Plaisance
I
,
Lepore
M
,
Cruchet
S
,
Berthonneche
C
,
Sarre
A
,
Radtke
F
,
Pedrazzini
T
.
The Notch pathway controls fibrotic and regenerative repair in the adult heart
.
Eur Heart J
2014
;
35
:
2174
2185
.

130

Yang
YJ
,
Qian
HY
,
Huang
J
,
Li
JJ
,
Gao
RL
,
Dou
KF
,
Yang
GS
,
Willerson
JT
,
Geng
YJ
.
Combined therapy with simvastatin and bone marrow-derived mesenchymal stem cells increases benefits in infarcted swine hearts
.
Arterioscler Thromb Vasc Biol
2009
;
29
:
2076
2082
.

131

Zhao
L
,
Borikova
AL
,
Ben-Yair
R
,
Guner-Ataman
B
,
MacRae
CA
,
Lee
RT
,
Burns
CG
,
Burns
CE
.
Notch signaling regulates cardiomyocyte proliferation during zebrafish heart regeneration
.
Proc Natl Acad Sci USA
2014
;
111
:
1403
1408
.

132

Madonna
R
,
Taylor
DA
,
Geng
YJ
,
De Caterina
R
,
Shelat
H
,
Perin
EC
,
Willerson
JT
.
Transplantation of mesenchymal cells rejuvenated by the overexpression of telomerase and myocardin promotes revascularization and tissue repair in a murine model of hindlimb ischemia
.
Circ Res
2013
;
113
:
902
914
.

133

Mohsin
S
,
Khan
M
,
Nguyen
J
,
Alkatib
M
,
Siddiqi
S
,
Hariharan
N
,
Wallach
K
,
Monsanto
M
,
Gude
N
,
Dembitsky
W
,
Sussman
MA
.
Rejuvenation of human cardiac progenitor cells with Pim-1 kinase
.
Circ Res
2013
;
113
:
1169
1179
.

134

Madonna
R
,
Willerson
JT
,
Geng
YJ
.
Myocardin a enhances telomerase activities in adipose tissue mesenchymal cells and embryonic stem cells undergoing cardiovascular myogenic differentiation
.
Stem Cells
2008
;
26
:
202
211
.

135

Wong
LS
,
Oeseburg
H
,
de Boer
RA
,
van Gilst
WH
,
van Veldhuisen
DJ
,
van der Harst
P
.
Telomere biology in cardiovascular disease: the TERC-/- mouse as a model for heart failure and ageing
.
Cardiovasc Res
2009
;
81
:
244
252
.

136

Rando
TA
,
Wyss-Coray
T
.
Stem cells as vehicles for youthful regeneration of aged tissues
.
J Gerontol A Biol Sci Med Sci
2014
;
69
(Suppl. 1)
:
S39
S42
.

137

Bergmann
O
,
Bhardwaj
RD
,
Bernard
S
,
Zdunek
S
,
Barnabe-Heider
F
,
Walsh
S
,
Zupicich
J
,
Alkass
K
,
Buchholz
BA
,
Druid
H
,
Jovinge
S
,
Frisen
J
.
Evidence for cardiomyocyte renewal in humans
.
Science
2009
;
324
:
98
102
.

138

Kajstura
J
,
Gurusamy
N
,
Ogorek
B
,
Goichberg
P
,
Clavo-Rondon
C
,
Hosoda
T
,
D'Amario
D
,
Bardelli
S
,
Beltrami
AP
,
Cesselli
D
,
Bussani
R
,
del Monte
F
,
Quaini
F
,
Rota
M
,
Beltrami
CA
,
Buchholz
BA
,
Leri
A
,
Anversa
P
.
Myocyte turnover in the aging human heart
.
Circ Res
2010
;
107
:
1374
1386
.

139

Mollova
M
,
Bersell
K
,
Walsh
S
,
Savla
J
,
Das
LT
,
Park
SY
,
Silberstein
LE
,
Dos Remedios
CG
,
Graham
D
,
Colan
S
,
Kuhn
B
.
Cardiomyocyte proliferation contributes to heart growth in young humans
.
Proc Natl Acad Sci USA
2013
;
110
:
1446
1451
.

140

Senyo
SE
,
Steinhauser
ML
,
Pizzimenti
CL
,
Yang
VK
,
Cai
L
,
Wang
M
,
Wu
TD
,
Guerquin-Kern
JL
,
Lechene
CP
,
Lee
RT
.
Mammalian heart renewal by pre-existing cardiomyocytes
.
Nature
2013
;
493
:
433
436
.

141

Bergmann
O
,
Zdunek
S
,
Felker
A
,
Salehpour
M
,
Alkass
K
,
Bernard
S
,
Sjostrom
SL
,
Szewczykowska
M
,
Jackowska
T
,
Dos Remedios
C
,
Malm
T
,
Andra
M
,
Jashari
R
,
Nyengaard
JR
,
Possnert
G
,
Jovinge
S
,
Druid
H
,
Frisen
J
.
Dynamics of cell generation and turnover in the human heart
.
Cell
2015
;
161
:
1566
1575
.

142

Mirotsou
M
,
Jayawardena
TM
,
Schmeckpeper
J
,
Gnecchi
M
,
Dzau
VJ
.
Paracrine mechanisms of stem cell reparative and regenerative actions in the heart
.
J Mol Cell Cardiol
2011
;
50
:
280
289
.

143

Aurora
AB
,
Olson
EN
.
Immune modulation of stem cells and regeneration
.
Cell Stem Cell
2014
;
15
:
14
25
.

144

Lauden
L
,
Boukouaci
W
,
Borlado
LR
,
Lopez
IP
,
Sepulveda
P
,
Tamouza
R
,
Charron
D
,
Al-Daccak
R
.
Allogenicity of human cardiac stem/progenitor cells orchestrated by programmed death ligand 1
.
Circ Res
2013
;
112
:
451
464
.

145

Zacchigna
S
,
Giacca
M
.
Extra- and intracellular factors regulating cardiomyocyte proliferation in postnatal life
.
Cardiovasc Res
2014
;
102
:
312
320
.

146

Engel
FB
,
Hsieh
PC
,
Lee
RT
,
Keating
MT
.
FGF1/p38 MAP kinase inhibitor therapy induces cardiomyocyte mitosis, reduces scarring, and rescues function after myocardial infarction
.
Proc Natl Acad Sci USA
2006
;
103
:
15546
15551
.

147

Morikawa
Y
,
Zhang
M
,
Heallen
T
,
Leach
J
,
Tao
G
,
Xiao
Y
,
Bai
Y
,
Li
W
,
Willerson
JT
,
Martin
JF
.
Actin cytoskeletal remodeling with protrusion formation is essential for heart regeneration in Hippo-deficient mice
.
Sci Signal
2015
;
8
:
ra41
.

148

Wei
K
,
Serpooshan
V
,
Hurtado
C
,
Diez-Cunado
M
,
Zhao
M
,
Maruyama
S
,
Zhu
W
,
Fajardo
G
,
Noseda
M
,
Nakamura
K
,
Tian
X
,
Liu
Q
,
Wang
A
,
Matsuura
Y
,
Bushway
P
,
Cai
W
,
Savchenko
A
,
Mahmoudi
M
,
Schneider
MD
,
van den Hoff
MJ
,
Butte
MJ
,
Yang
PC
,
Walsh
K
,
Zhou
B
,
Bernstein
D
,
Mercola
M
,
Ruiz-Lozano
P
.
Epicardial FSTL1 reconstitution regenerates the adult mammalian heart
.
Nature
2015
;
525
:
479
485
.

149

Leone
M
,
Magadum
A
,
Engel
FB
.
Cardiomyocyte proliferation in cardiac development and regeneration – a guide to methodologies and interpretations
.
Am J Physiol Heart Circ Physiol
2015
;
309
:
H1237
H1250
.

150

Ieda
M
,
Fu
JD
,
Delgado-Olguin
P
,
Vedantham
V
,
Hayashi
Y
,
Bruneau
BG
,
Srivastava
D
.
Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors
.
Cell
2010
;
42
:
375
386
.

151

Qian
L
,
Huang
Y
,
Spencer
CI
,
Foley
A
,
Vedantham
V
,
Liu
L
,
Conway
SJ
,
Fu
JD
,
Srivastava
D
.
In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes
.
Nature
2012
;
85
:
593
598
.

152

Song
K
,
Nam
YJ
,
Luo
X
,
Qi
X
,
Tan
W
,
Huang
GN
,
Acharya
A
,
Smith
CL
,
Tallquist
MD
,
Neilson
EG
,
Hill
JA
,
Bassel-Duby
R
,
Olson
EN
.
Heart repair by reprogramming non-myocytes with cardiac transcription factors
.
Nature
2012
;
85
:
599
604
.

153

Protze
S
,
Khattak
S
,
Poulet
C
,
Lindemann
D
,
Tanaka
EM
,
Ravens
U
.
A new approach to transcription factor screening for reprogramming of fibroblasts to cardiomyocyte-like cells
.
J Mol Cell Cardiol
2012
;
3
:
323
332
.

154

Jayawardena
TM
,
Egemnazarov
B
,
Finch
EA
,
Zhang
L
,
Payne
JA
,
Pandya
K
,
Zhang
Z
,
Rosenberg
P
,
Mirotsou
M
,
Dzau
VJ
.
MicroRNA-mediated in vitro and in vivo direct reprogramming of cardiac fibroblasts to cardiomyocytes
.
Circ Res
2012
;
10
:
1465
1473.

155

Frangioni
JV
,
Hajjar
RJ
.
In vivo tracking of stem cells for clinical trials in cardiovascular disease
.
Circulation
2004
;
110
:
3378
3383
.

156

Chen
IY
,
Wu
JC
.
Cardiovascular molecular imaging: focus on clinical translation
.
Circulation
2011
;
123
:
425
443
.

157

Zhu
J
,
Zhou
L
,
XingWu
F
.
Tracking neural stem cells in patients with brain trauma
.
N Engl J Med
2006
;
355
:
2376
2378
.

158

Hartung
DM
,
Zarin
DA
,
Guise
JM
,
McDonagh
M
,
Paynter
R
,
Helfand
M
.
Reporting discrepancies between the ClinicalTrials.gov results database and peer-reviewed publications
.
Ann Intern Med
2014
;
160
:
477
483
.

159

Ioannidis
JP
.
Contradicted and initially stronger effects in highly cited clinical research
.
J Am Med Assoc
2005
;
294
:
218
228
.

160

Nowbar
AN
,
Mielewczik
M
,
Karavassilis
M
,
Dehbi
HM
,
Shun-Shin
MJ
,
Jones
S
,
Howard
JP
,
Cole
GD
,
Francis
DP
.
Discrepancies in autologous bone marrow stem cell trials and enhancement of ejection fraction (DAMASCENE): weighted regression and meta-analysis
.
BMJ
2014
;
348
:
g2688
.

161

Rosen
MR
,
Myerburg
RJ
,
Francis
DP
,
Cole
GD
,
Marban
E
.
Translating stem cell research to cardiac disease therapies: pitfalls and prospects for improvement
.
J Am Coll Cardiol
2014
;
64
:
922
937
.

162

Hare
JM
,
Bolli
R
,
Cooke
JP
,
Gordon
DJ
,
Henry
TD
,
Perin
EC
,
March
KL
,
Murphy
MP
,
Pepine
CJ
,
Simari
RD
,
Skarlatos
SI
,
Traverse
JH
,
Willerson
JT
,
Szady
AD
,
Taylor
DA
,
Vojvodic
RW
,
Yang
PC
,
Moye
LA
.
Phase II clinical research design in cardiology: learning the right lessons too well: observations and recommendations from the Cardiovascular Cell Therapy Research Network (CCTRN)
.
Circulation
2013
;
127
:
1630
1635
.

163

Viswanathan
S
,
Keating
A
,
Deans
R
,
Hematti
P
,
Prockop
D
,
Stroncek
DF
,
Stacey
G
,
Weiss
DJ
,
Mason
C
,
Rao
MS
.
Soliciting strategies for developing cell-based reference materials to advance mesenchymal stromal cell research and clinical translation
.
Stem Cells Dev
2014
;
23
:
1157
1167
.

164

Niemansburg
SL
,
Teraa
M
,
Hesam
H
,
van Delden
JJ
,
Verhaar
MC
,
Bredenoord
AL
.
Stem cell trials for cardiovascular medicine: ethical rationale
.
Tissue Eng Part A
2013
;
20
:
2567
2574
.

165

de Groot
V
,
Beckerman
H
,
Lankhorst
GJ
,
Bouter
LM
.
How to measure comorbidity. A critical review of available methods
. J Clin Epidemiol
2003
;
56
:
221
229
.

166

Ferdinandy
P
,
Hausenloy
DJ
,
Heusch
G
,
Baxter
GF
,
Schulz
R
.
Interaction of risk factors, comorbidities, and comedications with ischemia/reperfusion injury and cardioprotection by preconditioning, postconditioning, and remote conditioning
.
Pharmacol Rev
2014
;
66
:
1142
1174
.

167

Sorror
ML
,
Maris
MB
,
Sandmaier
BM
,
Storer
BE
,
Stuart
MJ
,
Hegenbart
U
,
Agura
E
,
Chauncey
TR
,
Leis
J
,
Pulsipher
M
,
McSweeney
P
,
Radich
JP
,
Bredeson
C
,
Bruno
B
,
Langston
A
,
Loken
MR
,
Al-Ali
H
,
Blume
KG
,
Storb
R
,
Maloney
DG
.
Hematopoietic cell transplantation after nonmyeloablative conditioning for advanced chronic lymphocytic leukemia
.
J Clin Oncol
2005
;
23
:
3819
3829
.

168

Dey
D
,
Han
L
,
Bauer
M
,
Sanada
F
,
Oikonomopoulos
A
,
Hosoda
T
,
Unno
K
,
De Almeida
P
,
Leri
A
,
Wu
JC
.
Dissecting the molecular relationship among various cardiogenic progenitor cells
.
Circ Res
2013
;
112
:
1253
1262
.

169

Britten
MB
,
Abolmaali
ND
,
Assmus
B
,
Lehmann
R
,
Honold
J
,
Schmitt
J
,
Vogl
TJ
,
Martin
H
,
Schachinger
V
,
Dimmeler
S
,
Zeiher
AM
.
Infarct remodeling after intracoronary progenitor cell treatment in patients with acute myocardial infarction (TOPCARE-AMI): mechanistic insights from serial contrast-enhanced magnetic resonance imaging
.
Circulation
2003
;
108
:
2212
2218
.

170

van der Spoel
TI
,
Vrijsen
KR
,
Koudstaal
S
,
Sluijter
JP
,
Nijsen
JF
,
de Jong
HW
,
Hoefer
IE
,
Cramer
MJ
,
Doevendans
PA
,
van Belle
E
,
Chamuleau
SA
.
Transendocardial cell injection is not superior to intracoronary infusion in a porcine model of ischaemic cardiomyopathy: a study on delivery efficiency
.
J Cell Mol Med
2012
;
16
:
2768
2776
.

171

Patel
AN
,
Mittal
S
,
Turan
G
,
Winters
AA
,
Henry
TD
,
Ince
H
,
Trehan
N
.
REVIVE Trial: retrograde delivery of autologous bone marrow in patients with heart failure
.
Stem Cells Transl Med
2015
;
4
:
1021
1027
.

172

Strauer
BE
,
Brehm
M
,
Zeus
T
,
Kostering
M
,
Hernandez
A
,
Sorg
RV
,
Kogler
G
,
Wernet
P
.
Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans
.
Circulation
2002
;
106
:
1913
1918
.

173

Nguyen
PK
,
Lan
F
,
Wang
Y
,
Wu
JC
.
Imaging: guiding the clinical translation of cardiac stem cell therapy
.
Circ Res
2011
;
109
:
962
979
.

174

Hung
J
,
Lang
R
,
Flachskampf
F
,
Shernan
SK
,
McCulloch
ML
,
Adams
DB
,
Thomas
J
,
Vannan
M
,
Ryan
T
.
3D echocardiography: a review of the current status and future directions
.
J Am Soc Echocardiogr
2007
;
20
:
213
233
.

175

Dandel
M
,
Lehmkuhl
H
,
Knosalla
C
,
Suramelashvili
N
,
Hetzer
R
.
Strain and strain rate imaging by echocardiography – basic concepts and clinical applicability
.
Curr Cardiol Rev
2009
;
5
:
133
148
.

176

Wakami
K
,
Ohte
N
,
Sakata
S
,
Kimura
G
.
Myocardial radial strain in early diastole is useful for assessing left ventricular early diastolic function: comparison with invasive parameters
.
J Am Soc Echocardiogr
2008
;
21
:
446
451
.

177

Abraham
TP
,
Dimaano
VL
,
Liang
HY
.
Role of tissue Doppler and strain echocardiography in current clinical practice
.
Circulation
2007
;
116
:
2597
2609
.

178

Yu
CM
,
Sanderson
JE
,
Marwick
TH
,
Oh
JK
.
Tissue Doppler imaging a new prognosticator for cardiovascular diseases
.
J Am Coll Cardiol
2007
;
49
:
1903
1914
.

179

Grothues
F
,
Smith
GC
,
Moon
JC
,
Bellenger
NG
,
Collins
P
,
Klein
HU
,
Pennell
DJ
.
Comparison of interstudy reproducibility of cardiovascular magnetic resonance with two-dimensional echocardiography in normal subjects and in patients with heart failure or left ventricular hypertrophy
.
Am J Cardiol
2002
;
90
:
29
34
.

180

Pohost
GM
,
Hung
L
,
Doyle
M
.
Clinical use of cardiovascular magnetic resonance
.
Circulation
2003
;
108
:
647
653
.

181

Dobert
N
,
Britten
M
,
Assmus
B
,
Berner
U
,
Menzel
C
,
Lehmann
R
,
Hamscho
N
,
Schachinger
V
,
Dimmeler
S
,
Zeiher
AM
,
Grunwald
F
.
Transplantation of progenitor cells after reperfused acute myocardial infarction: evaluation of perfusion and myocardial viability with FDG-PET and thallium SPECT
.
Eur J Nucl Med Mol Imaging
2004
;
31
:
1146
1151
.

182

Castellani
M
,
Colombo
A
,
Giordano
R
,
Pusineri
E
,
Canzi
C
,
Longari
V
,
Piccaluga
E
,
Palatresi
S
,
Dellavedova
L
,
Soligo
D
,
Rebulla
P
,
Gerundini
P
.
The role of PET with 13N-ammonia and 18F-FDG in the assessment of myocardial perfusion and metabolism in patients with recent AMI and intracoronary stem cell injection
.
J Nucl Med
2010
;
51
:
1908
1916
.

183

Sloof
GW
,
Knapp
FF
Jr
,
van Lingen
A
,
Eersels
J
,
Poldermans
D
,
Bax
JJ
.
Nuclear imaging is more sensitive for the detection of viable myocardium than dobutamine echocardiography
.
Nucl Med Commun
2003
;
24
:
375
381
.

184

Salerno
M
,
Beller
GA
.
Noninvasive assessment of myocardial perfusion
.
Circ Cardiovasc Imaging
2009
;
2
:
412
424
.

185

Shiba
Y
,
Fernandes
S
,
Zhu
WZ
,
Filice
D
,
Muskheli
V
,
Kim
J
,
Palpant
NJ
,
Gantz
J
,
Moyes
KW
,
Reinecke
H
,
Van Biber
B
,
Dardas
T
,
Mignone
JL
,
Izawa
A
,
Hanna
R
,
Viswanathan
M
,
Gold
JD
,
Kotlikoff
MI
,
Sarvazyan
N
,
Kay
MW
,
Murry
CE
,
Laflamme
MA
.
Human ES-cell-derived cardiomyocytes electrically couple and suppress arrhythmias in injured hearts
.
Nature
2012
;
489
:
322
325
.

Author notes

These authors contributed equally.