Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Mar 14.
Published in final edited form as: JAMA Cardiol. 2016 Oct 1;1(7):831–841. doi: 10.1001/jamacardio.2016.2225

Adult stem cell therapy and heart failure, 2000 to 2016: a systematic review

Patricia K Nguyen 1,2,3,*, June-Wha Rhee 1,2,*, Joseph C Wu 1,2,4
PMCID: PMC5349705  NIHMSID: NIHMS853907  PMID: 27557438

Abstract

Importance

Stem cell therapy is a promising treatment strategy for patients with heart failure, which accounts for over 10% of deaths in the U.S. annually. Despite over a decade of research, further investigation is still needed to determine whether stem cell regenerative therapy is clinically effective and can be routinely implemented in clinical practice.

Objective

The purpose of this review is to describe the current progress in cardiac stem cell regenerative therapy using adult stem cells and highlight the merits and limitations of clinical trials performed to date.

Evidence Review

Information for this review was obtained through a search of PubMed and the Cochrane database for English language studies published between January 1, 2000 and April 20, 2016. Twenty-nine randomized clinical trials and 7 systematic reviews and meta-analyses were included in this review.

Findings

Although adult stem cells were once believed to have the ability to create new heart tissue or grow blood vessels, preclinical studies suggest instead that these cells release cardio-protective paracrine factors that activate endogenous pathways, leading to myocardial repair. Subsequent randomized controlled clinical trials, the majority of which used autologous bone marrow mononuclear cells, have found only a modest benefit in patients receiving stem cell therapy. The lack of a significant benefit may result from variations in trial methodology, discrepancies in reporting, and an over-reliance on surrogate endpoints.

Conclusions and Relevance

Although stem cell therapy for cardiovascular disease is not yet ready for routine clinical application, significant progress continues to be made. Physicians should be aware of the current status of this treatment so that they can better inform their patients who may be in search of alternative therapies.

Introduction

Heart failure (HF) is a devastating disease that causes significant morbidity and mortality, accounting for one in nine deaths in the US.1 Patients who suffer from coronary artery disease (CAD), valvular heart disease, and other cardiac disorders are at risk of developing HF. Because therapeutic options for advanced HF remain limited to organ transplantation and left ventricular assist device (LVAD), there is a strong impetus to develop alternative treatment strategies. Stem cell regenerative medicine is a promising therapeutic strategy to repair or replace injured and nonviable myocardium. Effective clinical translation, however, remains challenging due to inconclusive study results regarding stem cell regenerative capacity and their ability to improve cardiac function.26 Here we will review the proposed mechanisms of action for stem cell regenerative therapy, compare various stem cell sources, and discuss the merits and limitations of recently published adult stem cell clinical trials.

Proposed Mechanisms of Action to Improve Heart Function

Over the last decade, investigators have proposed three basic mechanisms to support the assertion that stem cell therapy can be used as an effective treatment for HF (Figure 1). Although it was once believed that adult stem cells could generate new cardiac tissue,7,8 a process termed cardiogenesis, further investigation has revealed that few if any adult stem cells differentiate into cardiomyocytes and engraft into the myocardium.9 The second proposed mechanism of action suggests that stem cells could generate vasculature via angiogenesis or vasculogenesis by activating endogenous endothelial progenitor cells (EPCs) or recruiting them from the vasculature. The existence of EPCs, however, remains controversial due to a lack of unique surface markers to identify these cells.10 Moreover, only a subset of EPCs may be of true endothelial lineage capable of neovasculogenesis, and these populations are rare and likely of insufficient number to produce measureable improvement in heart function.11

Figure 1. Schematic of the proposed mechanism of action of stem cell therapy.

Figure 1

The figure illustrates the theoretical mechanisms of action of various stem cell populations proposed in the literature. Although stem cells can potentially repair the injured myocardium by increasing angiogenesis, releasing factors that reduce cell death or modulate the immune system (e.g., paracrine activation), and/or creating new heart tissue, thus far only paracrine activation has been proven while the other hypotheses remain controversial. Stem cell sources include: 1) the bone marrow which contains the most diverse group of cells (e.g., HSCs, EPCs, MSCs, and specific stromal cell subpopulations) and factors (e.g., cytokine and growth factors) that can potentially regenerate the myocardium; 2) other sources of MSCs such as adipose tissue and the umbilical cord; and 3) cardiac tissue that may contain cardiac progenitor cells or cardiospheres. HSCs: hematopoietic stem cells, EPCs: endothelial progenitor cells, BM: bone marrow, SCs: stem cells, GFs: growth factors, MSCs: mesenchymal stem cells, CSCs: cardiac stem cells, CDCs: cardiosphere-derived cells.

While these two hypotheses remain controversial, mounting evidence now suggests that adult stem cells may exert paracrine effects by secreting cardio-protective factors. These secreted factors may stimulate vascular growth and remodeling, attenuate fibrosis, modulate inflammation, regulate cell differentiation and survival, and recruit resident stem or progenitor cells.12,13 Activation of these pathways may blunt reperfusion injury or attenuate adverse remodeling in patients suffering from acute myocardial infarction (AMI) or HF, respectively. Interestingly, recent studies have shown that these factors may be clustered into extracellular membrane vesicles, including exosomes and microsomes, which can then transfer proteins, lipids, RNA, and microRNAs to mediate cardioprotection.14,15 Although further studies are needed to confirm that these vesicles can substitute for stem cell therapy, delivering these vesicles rather than cells themselves may present a clinically attractive therapeutic option from a regulatory and commercial perspective.

Stem Cells Utilized in Clinical Trials For Heart Diseases

Although animal studies support the idea that the favorable effects observed from treatment with adult stem cells are attributable to paracrine effect,13,16,17, the exact mechanism of action in humans remains unclear. Despite this incomplete knowledge, ample clinical experience has been accumulated from the numerous clinical trials using various adult stem cell populations, including bone marrow-derived mononuclear cells (BMMNCs), mesenchymal stem cells (MSCs), and stem cells isolated from cardiac tissue (Figure 1). The clinical translatability of each of these adult stem cell populations is discussed below.

Bone marrow mononuclear cells (BMMNCs)

The human bone marrow contains a small fraction of various stem cell populations, including hematopoietic stem cells (HSCs), EPCs, and MSCs, which can be isolated using a Ficoll density gradient centrifugation and purified to obtain a final product that is commonly known as BMMNCs.18 Several studies have also used a magnetic separation device to isolate subpopulations of BMMNCs expressing CD34 or CD133 surface markers to further enhance efficacy.19,20 Interestingly, recent studies have suggested that the reparative capabilities of these cells may also be dependent on the age as well as the health of the donor. Vrtovec et al. showed that CD34+ cells obtained from younger patients had greater myocardial homing than those obtained from older patients, resulting in greater improvement in LVEF in the former.21 The reparative capacity of BMMNCs may even decline post-MI, as shown by Cogle et al., who analyzed the bone marrow obtained from patients enrolled in the TIME, LateTIME, and FOCUS-CCTRN trials.22 While all patients had a heterogeneous mixture of bone marrow cell subsets, bone marrow obtained from those patients post-MI had decreased angiogenic and vasculogenic capabilities. Patients with a higher number of CD34+ had greater improvement in LVEF. Collectively, these studies support the notion that heterogeneity in cell number and viability can affect the therapeutic response of BMMNCS. Nevertheless, the relative abundance of stem cells in the bone marrow, low cost of isolation, and ease of procurement have allowed these cells to be used in more than 100 pre-clinical and clinical studies thus far,23 making BMMNCs the most researched stem cell source.

Mesenchymal Stem Cells (MSCs)

Mesenchymal stem cells are mesoderm-derived stem cells that exist in various tissues, including the bone marrow, umbilical cord blood, adipose tissues, and muscles.24 Although it remains unclear how biologically similar MSCs from various tissue sources are, both BM- and non-BM-derived (e.g., adipose tissue) MSCs, as well as “pre-conditioned” cardiopoietic MSCs, have been increasingly tested in cell therapy studies.25,26 Isolation, expansion, and purification of MSCs, however, can be a long and tedious process, which may limit the large-scale production of these cells for clinical transplantation.

Cardiac-derived stem cells

While still controversial, several investigators have reported the existence of resident populations of cardiac progenitor cells in post-natal hearts, challenging the notion that the myocardium is terminally differentiated.27,28 Isolated from adult heart tissue, c-kit-positive cardiac stem cells (CSCs) have been reported to differentiate into cardiomyocytes when transplanted into the heart after MI. Similarly, cells migrating out of cardiac tissue fragments to form spheres, commonly known as cardiosphere-derived cells (CDCs),29 have been reported to give rise to cardiomyocytes in vitro and in vivo after transplantation. Isolation of CSCs and CDCs requires harvesting cardiac tissue via percutaneous endomyocardial biopsies or surgical extraction, followed by digestion, expansion, and purification to the desired cell types, a process that can take over a month from the time of tissue sampling to generate a sufficient number of cells for therapy.30

Results from Clinical Trials

Over the last decade, researchers have evaluated the safety and efficacy of various stem cell populations in a number of clinical trials, with trials using autologous BMMNCs leading the charge (Table 1 and 2).3,4,31,32 Although initial results were encouraging, subsequent large-scale randomized placebo controlled trials have shown only a modest benefit, as confirmed in recently published meta-analyses.31,32 The following section summarizes a select list of clinical trials that have evaluated the safety and efficacy of transplanting adult stem cells in patients with CAD and HF. Due to space limitations, we are unable to detail the specifics of each trial and refer the reader to more comprehensive reviews of stem cell trials for further detail.2,6,3135

Table 1.

Published Stem Cell Clinical Trials for Ischemic Heart Disease

STUDY NAME YEAR DISEASE TRIAL DESIGN # OF PTS CELLS USED CELL HARVESTING DELIVERY MEAN F/U LVEF OTHER RESULTS
Acute MI
TOPCARE-AMI63 2002 Acute MI s/p PCI non-randomized, matched control 31 autologous BMMNCs/PBSCs BM iliac crest IC 4 mo improved perfusion + viability
Chen et al.47 2004 Acute MI s/p PCI Randomized, DB, PC 69 autologous BMMNCs BM iliac crest IC 6 mo improved perfusion
BOOST37 2004 STEMI s/p PCI randomized, open-label, single-center 60 autologous BMMNCs BM iliac crest IC 18 mo no sig improvement
LEUVEN-AMI46 2006 STEMI s/p PCI, LV dysfunction Randomized, DB, PC 67 autologous BMMNCs BM iliac crest IC 4 mo decreased infarct size, NS
REPAIR-AMI39 2006 Acute MI s/p PCI, LVEF <45% Randomized, DB, PC 204 autologous BMMNCs BM iliac crest IC 1 yr decreased LVEF, improved regional infarct contractility, improved MACE
ASTAMI40 2006 Ant STEMI randomized, open-label, single-center 97 autologous BMMNCs BM iliac crest IC 6 mo improved exercise capacity
FINCELL64 2008 STEMI s/p thrombolysis + PCI Randomized, DB, PC 80 autologous BMMNCs BM iliac crest IC 6 mo
Hare et al.65 2009 Acute MI s/p PCI Randomized, DB, PC, dose-escalating 53* hMSCs n/a IC 6 mo improved symptoms
MYSTAR66 2009 STEMI s/p PCI, LVEF <45% SB, no-placebo 60 autologous BMMNCs BM iliac crest IC & TEN 1 year improved infarct size, viability, perfusion
REGENT20 2009 ant STEMI s/p PCI, LVEF <40% Randomized, DB, PC 200 autologous BMMNCs UNSEL or SEL (CD34+CXCR4+) BM iliac crest IC 6 mo no sig improvement
Cao et al.67 2009 STEMI s/p PCI Randomized, DB, PC 86 autologous BMMNCs BM iliac crest IC 4 yr no diff in viability
BONAMI68 2011 STEMI s/p PCI, LVEF <45% Randomized, DB, PC 101 autologous BMMNCs BM iliac crest IC 3 mo trend to improved viability
HEBE45 2011 Acute MI s/p PCI multicenter, randomized, 3-arm open trial 200 autologous BMMNCs BM iliac crest or peripheral blood IC 1yr no significant improvement
LATE-TIME41 2011 Acute MI s/p PCI, LVEF <45% Randomized, DB, PC 87 autologous BMMNCs BM iliac crest IC 6 mo no significant diff in LVEF, infarct size, wall motion
TIME42 2012 anterior STEMI s/p PCI, LV dysfunction Randomized, DB, PC 120 autologous BMMNCs BM iliac crest IC 1yr no significant improvement
APOLLO69 2012 STEMI s/p PCI Randomized, DB, PC 14 autologous ADSCs liposuction IC 6 mo improved perfusion & scar formation
CADUCEUS30 2012 AMI s/p PCI, LVEF <45% Randomized, no placebo 25 autologous CDCs Endomyocardial Bx IC 6 mo reductions in scar, increases in viable mass and regional contractility
SWISS-AMI43 2013 Acute MI s/p PCI randomized, 3-arm trial, DB, PC 192 autologous BMMNCs BM iliac crest IC 1 yr no significant improvement
Gao et al.25 2015 STEMI s/p PCI Randomized, DB, PC 116* Allogenic WJMSCs Allogenic IC 18 mo Improved LVEF, LV volumes and perfusion
TECAM44 2015 STEMI s/p PCI randomized, open-label, SB, PC 120 autologous BMMNCs, G-CSFs, both Autologous IC 12 mo no significant improvement
Chronic Ischemia with Refractory Angina
Losordo et al.51 2007 intractable angina, ischemia Randomized, DB, PC 24 peripheral CD34+ after G-CSF × 5d peripheral blood TE 6 mo n/a significant improvement in angina
Ramshorst et al.70 2009 severe angina, ischemia, LVEF >35% Randomized, DB, PC 50 BMMNCs BM iliac crest TE 6 mo significant improvement in perfusion and angina
ACT34-CMI52 2011 refractory angina, ischemia Randomized, DB, PC 167 peripheral CD34+ after G-CSF × 5d peripheral blood TE 3/6 mo n/a Improvement in angina and exercise
*

Only a handful of trials use MSCs and these trials include only a small number of patients; thus, their conclusions may not be statistically significant.

Abbreviation: MI: myocardial infarction, SB: single-blinded, DB: double-blinded, PC: placebo-controlled, BM: bone marrow, BMMNCs: bone marrow derived mononuclear cells, MSCs: mesenchymal stem cells, ADSCs: adipose-derived stem cells, CDCs: cardiosphere-derived cells, IC: intracoronary, TEN: trans-endocardial, SM: skeletal myoblasts, TEP: transepicardial, AR: arrhythmogenicity

Table 2.

Published Stem Cell Clinical Trials for Heart Failure

STUDY NAME YR DISEASE TRIAL DESIGN # OF PTS CELLS USED CELL HARVESTING DELIVERY MEAN F/U LV EF OTHER RESULS ADVERSE EVENTS
Menasche et al71 2003 ICM, LVEF <35%, WMA, pre-CABG non-randomized 10 Autologous SM skeletal muscle bx TEP 10.9 mo N/A AR
Perin et al54 2003 ICM, LVEF < 40%, ischemia non-randomized, open-label 21 Autologous BMMNCs BM iliac crest TEN 4 mo improved perfusion none
TOPCARE -CHD55 2006 Sub-acute MI with WMA randomized, open-label, single-center 75 Autologous BMMNCs/PBSCs BM iliac crest IC 3 mo improved regional contractility none
MAGIC72 2008 ICM, LVEF <35%, WMA, pre-CABG randomized, DB, PC 97 Autologous SM skeletal muscle bx from thigh TEP 6 mo no diff in MACE AR
SCIPIO60 2011 ICM, LVEF <40, pre-CABG open-label, randomized 56 Autologous CSCs right atrial appendage IC 4mo/12 mo decreased infarct size none
FOCUS-CCTRN73 2012 ICM, LVEF <45% multicenter, randomized, DB, PC 92 Autologous BMMNCs BM iliac crest TEN 6 mo no significant diff in LVEF, infarct size, wall motion none
POSEIDON48 2012 ICM, LVEF <40 randomized, NO placebo controlled 30 Autologous and Allogenic hMSCs* EMBX TEN 30d/1 yr Autologous MSC improved 6 min walk test, infarct size. Allogenic MSC reduced LVEDV and infarct size none
C-CURE74 2013 ICM with LVEF <40%, NYHA >=2 randomized, DB, PC 36 Autologous Cardiopoietic hMSCs* BM iliac crest TEN 2 yr safe, improved LVEF, LVESV, symptoms none
CELLWAVE75 2013 ICM with LVEF <50% + NYHA>=2 randomized, DB, PC 103 Autologous BMMNCs BM iliac crest SW/IC 4 mo Improved LVEF, regional wall thickening, and MACE none
PRECISE59 2014 ICM with LVEF <45%, NYHA >=2 randomized, DB, PC 27 Autologous ADRCs liposuction TEN 36 mo N/A none
TAC-HFT57 2014 ICM, LVEF <50% randomized, DB, PC 59 Autologous BMMNC and MSCs BM iliac crest TEN 30 d MSC - improved functional capacity, infarct size. BMNC improved functional capacity. none
Vrtovec et al.21 2013 NICM (DCM) randomized, DB, PC 110 peripheral CD34+ after G-CSF × 5d peripheral blood IC 5yr improved LVEF, 6min walk distance, and decreased NT-proBNP. No diff mortality none
*

Only a handful of trials use MSCs and these trials include only a small number of patients; thus, their conclusions may not be statistically significant.

Abbreviation: MI: myocardial infarction, SB: single-blinded, DB: double-blinded, PC: placebo-controlled, BM: bone marrow, BMMNCs: bone marrow derived mononuclear cells, MSCs: mesenchymal stem cells, ADSCs: adipose-derived stem cells, CDCs: cardiosphere-derived cells, IC: intracoronary, TEN: trans-endocardial, SM: skeletal myoblasts, TEP: transepicardial, AR: arrhythmogenicity

Acute Myocardial Infarction

The use of stem cells at the time of AMI was supported by animal studies that suggested stem cell therapy could limit the extent of myocardial injury and potentially restore damaged myocardium.36 The initial observation of improved ventricular function after AMI following BMMNC therapy led to the initiation of the BOne marrOw transfer to enhance ST-elevation infarct regeneration (BOOST) trial published in 2004,37 which enrolled 60 patients randomized to receive intracoronary BMMNC therapy versus conventional medical therapy after percutaneous coronary intervention (PCI) for AMI. Although the study reported an improvement in left ventricular ejection fraction (LVEF) at 6 months following the BMMNC therapy, the observed benefit largely disappeared at 18 months except in those with significant infarct size (>60%) and depressed LVEF <50% at the time of therapy.38 This study was then followed by The Reinfusion of Enriched Progenitor cells And Infarct Remodeling in Acute Myocardial Infarction (REPAIR-AMI) trial,39 the first double-blinded, randomized trial enrolling over 200 patients to receive placebo or autologous BMMNCs. A significantly greater increase in LVEF was reported in the cell therapy group versus control at 6 months (mean of 5.5% versus 3.0%, P = 0.01), with a mortality benefit and sustained LVEF improvement at the 2-year follow up. Similar to the BOOST trial, those with depressed LVEF <50% appeared to have derived the most benefit. Despite the initial encouraging results with BMMNC therapies, many subsequent studies utilizing BMMNCs did not find a significant benefit (Table 1).20,4046 Similarly, the most recent Cochrane Database Systematic Review published in 2015 that included 41 randomized controlled trials using BMMNCs and a total of 2,732 participants showed that cell treatment was safe but found no significant improvements in quality of life or LVEF in the short or long term. The mean difference in LVEF between the treated and control group was 2 – 5%, which was not considered clinically relevant given the inherent variability of imaging tests.32

Because only a very small fraction of BMMNCs are actually stem cells (e.g., ~2–4% HSCs/EPCs and <0.01% MSCs), several studies have utilized a more select subgroup of BMMNCs, namely, CD34+ cells and MSCs, to eliminate biological variability and augment efficacy by removing bystander cells. In a study comparing the effects of intracoronary treatment with BMMNCs vs. BM-derived CD34+/CXCR4+ cells for patients after AMI with reduced LVEF <40%,20 Tendera et al. found no significant differences in LVEF or in the incidence of major adverse cardiovascular events (MACE), including death, re-infarction, stroke, or target vessel revascularization, between the two groups. In 2004, Chen et al. reported the first randomized trial delivering autologous MSCs via intracoronary injection in patients after AMI, finding a significant improvement in LVEF following cell therapy.47 Subsequently, Hare et al. administered intravenous allogeneic MSCs vs. placebo to AMI patients approximately 7–10 days after PCI, and found similar adverse event rates between the two groups, but also a trend toward improved LVEF and functional capacity in the treatment group.48 Despite a previous preclinical study reporting a high risk of micro-infarction from intracoronary delivery,49 available randomized placebo-controlled human studies have shown no significant adverse events after intracoronary or intravenous delivery of MSCs.34

After these encouraging initial results with BM-derived MSCs, investigators then turned their attention to the application of MSCs from adipose tissue and the umbilical cord. In 2004, Vulliet et al. randomized 14 patients with anterior AMI to receive intracoronary infusion of adipose-derived MSCs versus placebo (APOLLO) and showed that the treatment was safe with a trend towards a small reduction in infarct size.49 Further information on the efficacy and safety of these cells in patients with acute MI will be available after the release of findings from a multicenter, prospective, randomized, placebo-controlled phase IIb/III clinical sponsored by Cytori Therapeutics (ADVANCE study, NCT01216995). More recently, Gao et al. reported that intracoronary infusion of MSCs derived from umbilical cord obtained from healthy donors after full term birth resulted in a significant improvement in LVEF, LV volumes, and perfusion compared to controls at 18 months.25 It should be emphasized, however, that given the limited number of studies performed using MSCs and the relatively small sample size in each study, definitive conclusions regarding the efficacy of this cell population cannot be made at the present. Additional studies with larger sample sizes using MSCs are needed to validate these findings.

Finally, in a recent study CArdiosphere-Derived aUtologous stem CElls to reverse ventricUlar dysfunction (CADUCEUS) trial, Makkar et al. reported the first randomized, phase I trial evaluating the therapeutic effects of CDCs in the treatment of AMI.30 Patients with recent AMI and LVEF 25–45% were randomized to receive CDC therapy (n=23) or standard therapy (n = 8). Varying doses of CDCs were injected via intracoronary infusion approximately at 65 days after AMI. Results at 6 and 12 months of follow up showed that no patients died or developed cardiac tumors. However, at 12 months, one patients suffered a non-ST elevation MI and another patient required coronary vascularization.50 Although there was a significant decrease in scar size, increase in viability, and improvement in regional wall function after CDC treatment, there were no significant improvements in global systolic function or quality of life assessments. Subsequent studies such as the REgenerative CardiOsphere iNjection to STRengthen dysfUnCTional Hearts (RECONSTRUCT) trial and the ALLogeneic Heart STem Cells to Achieve Myocardial Regeneration (ALLSTAR) trial are underway to evaluate the efficacy of autologous and allogenic CSCs, respectively, in treating AMI.

Chronic Ischemia with Intractable Angina

Patients with intractable angina due to ischemia that is otherwise not amenable to revascularization may also benefit from therapy with stem cells. Losordo et al. published the first Phase I/IIa trial evaluating the safety and efficacy of intramyocardial transplantation of autologous CD34+ stem cells in 24 patients with intractable angina.51 Although results were not significant due to a small sample size, improvements in angina and exercise time were noted in the treatment group without safety issues. Importantly, in a follow-up study that included a larger sample of patients (n=167),52 the cell therapy group reported less frequent angina with improved exercise tolerance. Cardiac enzyme elevation, however, was reported in both the control and treatment after cell mobilization with G-CSF. More recently, Wang et al. reported similar efficacy results with the use of an intracoronary method for delivering EPCs.53

Ischemic Cardiomyopathy

The initial proposed mechanism of cardiogenesis made ischemic cardiomyopathy an attractive indication for adult stem cell therapy to replace the significant cardiomyocyte loss that occurs. A pilot study in 2003 by Perin et al. randomized 27 patients with ICM to receive transendocardial injection of BMMNCs or placebo.54 At 4 months, there was a significant improvement in LVEF in patients with cell therapy versus placebo (29% vs. 20%; P=0.003) without an increase in serious adverse events. This study validated the safety of transendocardial injection of BMMNCs, leading to two large trials of BMMNC-based therapy for ICM. In the first trial, the Transplantation Of Progenitor Cells And REcovery of LV function in patients with Chronic ischemic Heart Disease (TOPCARE-CHD) study, Assmus et al. randomized ICM patients to intracoronary infusion of BMMNCs vs. “circulating progenitor cells (CPCs)” derived from peripheral blood vs. placebo and found that transplantation of BMMNCs, as opposed to CPCs or placebo, resulted in a modest but significant improvement in LVEF at 3 months.55 These results, however, were not replicated in a second study sponsored by the National Heart, Lung, and Blood Institute (NHLBI), First Mononuclear Cells injected in the United States conducted by the Cardiovascular Cell Therapy Research Network (FOCUS-CCTRN),56 which did not show a significant improvement in LVEF at 6 months following transendocardial delivery of BMMNCs for treating ICM patients with LVEF <45%. Consistent with these findings, a recent Cochrane meta-analysis review that included 23 randomized control trials and 1,255 participants found no short-term benefits (<12 months) associated with treatment with autologous BMMNCs.31 Longer-term results (≥12 months), however, showed that autologous BMMNCs reduced the incidence of mortality (RR=0.28, 95% CI 0.14–0.53, p=0.0001) and HF re-hospitalization (RR 0.26, 95% CI 0.07–0.94, P=0.04).

Several studies have compared the efficacy of BMMNCs versus MSCs. In 2014, Heldman et al. reported a phase I/II study on the transendocardial delivery of BMMNCs versus BM-derived autologous MSCs versus placebo for ICM patients with LVEF <50%.57 Although small in size, significant improvements in HF symptoms, infarct size, and regional myocardial function were observed in patients treated with MSCs. Because BM-derived MSCs require significant preparation time, Hare et al. conducted a study comparing the safety and efficacy of using allogenic instead of autologous MSCs in a non-placebo controlled, randomized, dose-escalating trial (POSEIDON).26 While treatment with both cell types resulted in reduction in the infarct size, only autologous MSCs led to improved quality of life score and 6-minute walk test. Ascheim et al. also reported the use of allogenic MSCs for patients with advanced HF randomized to myocardial injections of MSC versus placebo at the time of LVAD implantation.58 Although imaging surrogates for improvement failed to reach statistical significance, the study demonstrated no increase in incidence of HLA antibodies to indicate immunologic reactions. Recently, Perin et al. reported improvement in exercise tolerance with higher peak VO2 but not in in LVEF for 27 ICM patients receiving adipose-derived MSCs, suggesting that the injected MSCs may promote angiogenesis through its paracrine effects.59 Taken together, these studies support the safety of BMMNCs and MSCs in ICM patients and suggest that MSCs may be more beneficial than BMMNCs. It should be noted, however, that three patients with ICM enrolled in one of two sister trials sponsored by Cytori Inc. (ATHENA I and II: NCT01556022 and NCT02052427) developed reversible, cerebrovascular events after receiving intramyocardial injection of adipose derived stem cells in one of two sister randomized, controlled trials evaluating the safety and efficacy of these cells delivered through intra-myocardial injection in patients with ICM.

With the introduction of potential residential cardiac progenitor cells, the use of CSCs for ICM was studied in the Stem Cell Infusion in Patients with Ischemic cardiOmyopathy (SCIPIO) trial.60 In this phase I trial, CSCs were isolated from the right atrial appendage at the time of coronary artery bypass graft surgery. The cultured CSCs were then delivered to the patients via intracoronary injection at a later time (mean of 113 days after surgery). Published findings indicate that patients had improved LVEF, regional contractility, and HF symptoms when treated with CSCs versus no treatment.

Non-ischemic Cardiomyopathy

Limited but increasing research effort has been made to evaluate the safety and efficacy of cell therapy for treating non-ischemic cardiomyopathy (NICM). One of the largest studies to date on cell therapy for NICM randomized 110 patients with dilated cardiomyopathy in a 1:1 ratio to receive intracoronary infusion of autologous peripheral CD34+ cells or no therapy.21 In this study, CD34+ cells mobilized by G-CSF were injected into the coronary artery territory with the greatest perfusion defect as identified by myocardial scintigraphy. At 1 year and 5 years, the treatment group showed a significant improvement in LVEF and 6-minute walk distance.21

Limitations of Adult Stem Cell Trials Performed To Date

As discussed above and outlined in Table 1 and 2, there are significant differences in the reported benefit between the early randomized controlled trials compared to more recent randomized controlled trials using adult stem cells. The reasons for these differences in efficacy remain unclear, but may be partially due to variations in trial design and methodology, including differences in the chosen cell source, dosing or concentration, the route of administration, timing of delivery, and clinical characteristics of patients recruited in the trial (Figure 2). Furthermore, the lack of standardized protocols limits our ability to evaluate the relative effectiveness of these approaches and most importantly the optimal cell source for each. While a handful of studies performed head-to-head comparisons of efficacy between two different cell types or among multiple doses,48,52 additional studies are needed to identify the optimal strategy for clinical application.

Figure 2. Overview of the various patient cohorts, cell types, doses, routes of delivery, and clinical endpoints used in adult stem cell trials.

Figure 2

A significant difference in efficacy has been observed in earlier randomized, controlled trials versus later randomized, controlled trials, which may be partially explained by variations in patient cohorts, cell types, doses, routes of delivery, and clinical endpoints evaluated in adult stem cell trials. Trials have been conducted in patients with various cardiac diseases including AMI, chronic ischemia/angina, ischemic CM, and non-ischemic CM. Within each cohort, certain patient characteristics may also affect efficacy such as transplanting bone marrow acquired from young versus old patients, delivering cells to patients immediately versus weeks/months post MI, and treating patients who have suffered small versus large infarct or who have mild (<45%) versus significant (<35%) impairment in LVEF. Various adult stem cell types have also been evaluated, including bone-marrow derived cells (e.g., BMMNCs, CD34+ or CD133+ cells, and MSCs) and adipose/umbilical derived SCs, as well as stem cells derived from cardiac tissue (e.g., CSCs and CDCs). These cells have been delivered in multiple doses and with different delivery approaches. Finally, most studies have used surrogate endpoints like LVEF, infarct size, and perfusion defects, which do not always correlate with more definite endpoints such as death, myocardial infarction, revascularization, heart failure readmission, and other major adverse cardiovascular events (MACE).

Discrepancies in reporting have also been recently identified as another potential source of inconsistency. Nowbar et al. performed a recent meta-analysis of 49 trials to determine if the number of discrepancies in trial design, methods and results was associated with improvement in LVEF.61 They found a positive relationship between the number of discrepancies and improvement in LVEF, raising questions about the validity of earlier studies. Results from this study emphasize the need to adhere to rigorous standards in conducting and reporting future stem cell clinical trials.

Another possible source of difference in reported efficacy is the over-reliance on surrogate imaging endpoints, which have inherent inter- and intra-observer variability. Earlier trials have relied on imaging endpoints such as changes in LVEF, infarct size, and perfusion abnormalities because they are more easily measured, require smaller sample size and shorter follow-up. Surrogate endpoints, however, are generally limited by their degree of correlation with MACE, which casts significant doubt as to the clinical relevance of the outcomes observed. Even the most well-studied surrogate endpoint (i.e., global LVEF) does not consistently parallel survival.62 Surrogate endpoints are also not accepted as primary endpoints by the US Food and Drug Administration for Phase III trials.

More recently, stem cell trials in AMI and HF have incorporated more definitive endpoints such as the incidence of death, infarction, HF, and arrhythmias,31,32 but these were low-frequency events, resulting in inadequate power to detect significant differences. To address this limitation, European investigators have begun recruiting for the effect of intracoronary reinfusion of Bone marrow-derived mononuclear cells on all-cause mortality in Acute Myocardial Infarction (BAMI: NCT01569178) study, the largest stem cell trial using BMMNCs to date. The trial is recruiting 3,000 patients with AMI and LVEF <45% and is powered to detect a 25% decrease in 2-year all cause mortality after treatment. Results from this trial will help us determine whether further investment in BMMNCs is warranted.

Conclusion

In the past two decades, researchers have achieved significant milestones toward their goal of bringing stem cell regenerative medicine to the bedside. First, evidence suggests the benefit of adult stem cell therapy is likely mediated by the release of cardio-protective factors that activate endogenous pathways to repair the myocardium rather than de novo cardiomyocyte or blood vessel formation. Second, safe delivery of cells has been demonstrated in both preclinical and clinical trials, although the question of clinical efficacy of adult stem cell therapies remains elusive. Future research efforts should focus on developing new strategies to expand our knowledge of stem cell biology, following the fate of stem cells post-delivery, and designing larger trials using clinically meaningful endpoints rather than surrogate endpoints to better investigate their therapeutic potential.

Acknowledgments

We thank Blake Wu and Karim Sallam for critical reading of the manuscript and Amy Morris for assisting with the illustrations. We thank funding support from American Heart Association 13EIA14420025, Burroughs Wellcome Foundation Innovation in Regulatory Science Award, National Institutes of Health (NIH) R01 HL113006, NIH R01 HL130020, NIH R01 Hl126527, California Institute of Regenerative Medicine (CIRM) DR2A-05394 and RT3-07798 (JCW), and Stanford Cardiovascular Institute Seed Grant (PKN).

References

  • 1.Mozaffarian D, Benjamin EJ, Go AS, et al. Heart disease and stroke statistics--2015 update: a report from the American Heart Association. Circulation. 2015;131(4):e29–322. doi: 10.1161/CIR.0000000000000152. [DOI] [PubMed] [Google Scholar]
  • 2.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. doi: 10.1161/CIRCULATIONAHA.111.086074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Fisher Sa, Doree C, Mathur A, Martin-Rendon E. Meta-analysis of cell therapy trials for patients with heart failure. Circulation Research. 2015;116:1361–1377. doi: 10.1161/CIRCRESAHA.116.304386. [DOI] [PubMed] [Google Scholar]
  • 4.Gyöngyösi M, Wojakowski W, Lemarchand P, et al. Meta-Analysis of Cell-based CaRdiac stUdiEs (ACCRUE) in patients with acute myocardial infarction based on individual patient data. Circulation Research. 2015;116:1346–1360. doi: 10.1161/CIRCRESAHA.116.304346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Delewi R, Hirsch A, Tijssen JG, et al. Impact of intracoronary bone marrow cell therapy on left ventricular function in the setting of ST-segment elevation myocardial infarction: a collaborative meta-analysis. European Heart Journal. 2014;35:989–998. doi: 10.1093/eurheartj/eht372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Wollert KC, Drexler H. Cell therapy for the treatment of coronary heart disease: a critical appraisal. Nature Reviews Cardiology. 2010;7:204–215. doi: 10.1038/nrcardio.2010.1. [DOI] [PubMed] [Google Scholar]
  • 7.Nygren JM, Jovinge S, Breitbach M, et al. Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nature Medicine. 2004;10:494–501. doi: 10.1038/nm1040. [DOI] [PubMed] [Google Scholar]
  • 8.Orlic D, Kajstura J, Chimenti S, et al. Bone marrow cells regenerate infarcted myocardium. Nature. 2001;410:701–705. doi: 10.1038/35070587. [DOI] [PubMed] [Google Scholar]
  • 9.Balsam LB, Wagers AJ, Christensen JL, Kofidis T, Weissman IL, Robbins RC. Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium. Nature. 2004;428:668–673. doi: 10.1038/nature02460. [DOI] [PubMed] [Google Scholar]
  • 10.Fadini GP, Losordo D, Dimmeler S. Critical reevaluation of endothelial progenitor cell phenotypes for therapeutic and diagnostic use. Circulation Research. 2012;110(4):624–637. doi: 10.1161/CIRCRESAHA.111.243386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ingram DA, Mead LE, Tanaka H, et al. Identification of a novel hierarchy of endothelial progenitor cells using human peripheral and umbilical cord blood. Blood. 2004;104(9):2752–2760. doi: 10.1182/blood-2004-04-1396. [DOI] [PubMed] [Google Scholar]
  • 12.Gnecchi M, He H, Liang OD, et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat Med. 2005;11(4):367–368. doi: 10.1038/nm0405-367. [DOI] [PubMed] [Google Scholar]
  • 13.Stempien-Otero A, Helterline D, Plummer T, et al. Mechanisms of bone marrow-derived cell therapy in ischemic cardiomyopathy with left ventricular assist device bridge to transplant. Journal of the American College of Cardiology. 2015;65:1424–1434. doi: 10.1016/j.jacc.2015.01.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Ibrahim AG, Cheng K, Marban E. Exosomes as critical agents of cardiac regeneration triggered by cell therapy. Stem Cell Reports. 2014;2(5):606–619. doi: 10.1016/j.stemcr.2014.04.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kishore R, Khan M. More than tiny sacks: stem cell exosomes as cell-free modality for cardiac repair. Circulation Research. 2016;118(2):330–343. doi: 10.1161/CIRCRESAHA.115.307654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Duran JM, Makarewich CA, Sharp TE, et al. Bone-derived stem cells repair the heart after myocardial infarction through transdifferentiation and paracrine signaling mechanisms. Circulation Research. 2013;113(5):539–552. doi: 10.1161/CIRCRESAHA.113.301202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gnecchi M, Zhang Z, Ni A, Dzau VJ. Paracrine mechanisms in adult stem cell signaling and therapy. Circulation Research. 2008;103(11):1204–1219. doi: 10.1161/CIRCRESAHA.108.176826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bianco P, Riminucci M, Gronthos S, Robey PG. Bone marrow stromal stem cells: nature, biology, and potential applications. Stem Cells. 2001;19(3):180–192. doi: 10.1634/stemcells.19-3-180. [DOI] [PubMed] [Google Scholar]
  • 19.Bartunek J, Vanderheyden M, Vandekerckhove B, et al. Intracoronary injection of CD133-positive enriched bone marrow progenitor cells promotes cardiac recovery after recent myocardial infarction: feasibility and safety. Circulation. 2005;112:I178–183. doi: 10.1161/CIRCULATIONAHA.104.522292. [DOI] [PubMed] [Google Scholar]
  • 20.Tendera M, Wojakowski W, Ruzyłło W, et al. 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 Intracor. European Heart Journal. 2009;30:1313–1321. doi: 10.1093/eurheartj/ehp073. [DOI] [PubMed] [Google Scholar]
  • 21.Vrtovec B, Poglajen G, Lezaic L, et al. Effects of intracoronary CD34+ stem cell transplantation in nonischemic dilated cardiomyopathy patients: 5-year follow-up. Circulation Research. 2013;112:165–173. doi: 10.1161/CIRCRESAHA.112.276519. [DOI] [PubMed] [Google Scholar]
  • 22.Cogle CR, Wise E, Meacham AM, et al. Detailed analysis of bone marrow from patients with ischemic heart disease and left ventricular dysfunction: BM CD34, CD11b, and clonogenic capacity as biomarkers for clinical outcomes. Circulation Research. 2014;115(10):867–874. doi: 10.1161/CIRCRESAHA.115.304353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Strauer B-E, Steinhoff G. 10 Years of intracoronary and intramyocardial bone marrow stem cell therapy of the heart. Journal of the American College of Cardiology. 2011;58:1095–1104. doi: 10.1016/j.jacc.2011.06.016. [DOI] [PubMed] [Google Scholar]
  • 24.Williams AR, Hare JM. Mesenchymal stem cells: biology, pathophysiology, translational findings, and therapeutic implications for cardiac disease. Circulation Research. 2011;109(8):923–940. doi: 10.1161/CIRCRESAHA.111.243147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gao LR, Chen Y, Zhang NK, et al. Intracoronary infusion of Wharton’s jelly-derived mesenchymal stem cells in acute myocardial infarction: double-blind, randomized controlled trial. BMC Medicine. 2015;13:162. doi: 10.1186/s12916-015-0399-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hare JM, Fishman JE, Gerstenblith G, et al. Comparison of allogeneic vs autologous bone marrow – derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy. JAMA. 2012;308:2369–2379. doi: 10.1001/jama.2012.25321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bearzi C, Rota M, Hosoda T, et al. Human cardiac stem cells. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(35):14068–14073. doi: 10.1073/pnas.0706760104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Beltrami AP, Barlucchi L, Torella D, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114(6):763–776. doi: 10.1016/s0092-8674(03)00687-1. [DOI] [PubMed] [Google Scholar]
  • 29.Smith RR, Barile L, Cho HC, et al. Regenerative potential of cardiosphere-derived cells expanded from percutaneous endomyocardial biopsy specimens. Circulation. 2007;115(7):896–908. doi: 10.1161/CIRCULATIONAHA.106.655209. [DOI] [PubMed] [Google Scholar]
  • 30.Makkar RR, Smith RR, Cheng K, et al. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase 1 trial. Lancet. 2012;379:895–904. doi: 10.1016/S0140-6736(12)60195-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Fisher Sheila A, Brunskill Susan J, Doree C, Mathur A, Taggart David P, Martin-Rendon E. Stem cell therapy for chronic ischaemic heart disease and congestive heart failure. Cochrane Database of Systematic Reviews. 2014 doi: 10.1002/14651858.CD007888.pub2. [DOI] [PubMed] [Google Scholar]
  • 32.Fisher SA, Zhang H, Doree C, Mathur A, Martin-Rendon E. Stem cell treatment for acute myocardial infarction. The Cochrane Database of Systematic Reviews. 2015;9:CD006536. doi: 10.1002/14651858.CD006536.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Pavo N, Charwat S, Nyolczas N, et al. Cell therapy for human ischemic heart diseases: critical review and summary of the clinical experiences. J Mol Cell Cardiol. 2014;75:12–24. doi: 10.1016/j.yjmcc.2014.06.016. [DOI] [PubMed] [Google Scholar]
  • 34.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(15):1807–1818. doi: 10.1093/eurheartj/ehn220. [DOI] [PubMed] [Google Scholar]
  • 35.Golpanian S, Schulman IH, Ebert RF, et al. Concise review: review and perspective of cell dosage and routes of administration from preclinical and clinical studies of stem cell therapy for heart disease. Stem Cells Translational Medicine. 2016;5(2):186–191. doi: 10.5966/sctm.2015-0101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Zwetsloot PP, Vegh AM, Jansen Of Lorkeers SJ, et al. Cardiac stem cell treatment in myocardial infarction: a systematic review and meta-analysis of preclinical studies. Circulation Research. 2016;118(8):1223–1232. doi: 10.1161/CIRCRESAHA.115.307676. [DOI] [PubMed] [Google Scholar]
  • 37.Wollert KC, Meyer GP, Lotz J, et al. Intracoronary autologous bone-marrow cell transfer after myocardial infarction: The BOOST randomised controlled clinical trial. Lancet. 2004;364:141–148. doi: 10.1016/S0140-6736(04)16626-9. [DOI] [PubMed] [Google Scholar]
  • 38.Meyer GP, Wollert KC, Lotz J. 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. doi: 10.1161/CIRCULATIONAHA.105.575118. [DOI] [PubMed] [Google Scholar]
  • 39.Schächinger V, Erbs S, Elsässer A, et al. Intracoronary bone marrow-derived progenitor cells in acute myocardial infarction. New England Journal of Medicine. 2006;355:1210–1221. doi: 10.1056/NEJMoa060186. [DOI] [PubMed] [Google Scholar]
  • 40.Lunde K, Solheim S, Aakhus S, et al. Intracoronary injection of mononuclear bone marrow cells in acute myocardial infarction. New England Journal of Medicine. 2006;355:2239–2246. doi: 10.1056/NEJMoa055706. [DOI] [PubMed] [Google Scholar]
  • 41.Traverse JH, Henry TD, Ellis SG, et al. 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. JAMA. 2011;306(19):2110–2119. doi: 10.1001/jama.2011.1670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Traverse JH, Henry TD, Pepine CJ, et al. Effect of the use and timing of bone marrow mononuclear cell delivery on left ventricular function after acute myocardial infarction: the TIME randomized trial. JAMA. 2012;308(22):2380–2389. doi: 10.1001/jama.2012.28726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Sürder D, Manka R, Cicero VL, et al. Intracoronary injection of bone marrow – derived mononuclear cells early or late after acute effects on global left ventricular function. Circulation. 2013;127:1968–1979. doi: 10.1161/CIRCULATIONAHA.112.001035. [DOI] [PubMed] [Google Scholar]
  • 44.Roman JAS, Sanchez PL, Villa A, Sanz-ruiz R, Fernandez-Santos ME, Gimeno F. Comparison of different bone marrow-derived stem cell approaches in perfused STEMI (TECAM Trial) Journal of the American College of Cardiology. 2015:65. doi: 10.1016/j.jacc.2015.03.563. [DOI] [PubMed] [Google Scholar]
  • 45.Hirsch A, Nijveldt R, van der Vleuten PA, et al. 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. European Heart Journal. 2011;32:1736–1747. doi: 10.1093/eurheartj/ehq449. [DOI] [PubMed] [Google Scholar]
  • 46.Janssens S, Dubois C, Bogaert J, et al. Autologous bone marrow-derived stem-cell transfer in patients with ST-segment elevation myocardial infarction: double-blind, randomised controlled trial. Lancet. 2006;367:113–121. doi: 10.1016/S0140-6736(05)67861-0. [DOI] [PubMed] [Google Scholar]
  • 47.Chen S-l, Fang W-w, Ye F, et al. Effect on left ventricular function of intracoronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarction. The American Journal of Cardiology. 2004;94:92–95. doi: 10.1016/j.amjcard.2004.03.034. [DOI] [PubMed] [Google Scholar]
  • 48.Hare JM, Fishman JE, Gerstenblith G, et al. Comparison of allogeneic vs autologous bone marrow-derived mesenchymal stem cells delivered by transendocardial injection in patients with ischemic cardiomyopathy: the POSEIDON randomized trial. JAMA. 2012;308(22):2369–2379. doi: 10.1001/jama.2012.25321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Vulliet PR, Greeley M, Halloran SM, MacDonald KA, Kittleson MD. Intra-coronary arterial injection of mesenchymal stromal cells and microinfarction in dogs. Lancet. 2004;363(9411):783–784. doi: 10.1016/S0140-6736(04)15695-X. [DOI] [PubMed] [Google Scholar]
  • 50.Malliaras K, Makkar RR, Smith RR, et al. Intracoronary cardiosphere-derived cells after myocardial infarction. Journal of the American College of Cardiology. 2014;63:110–122. doi: 10.1016/j.jacc.2013.08.724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Losordo DW, Schatz RA, White CJ, et al. Intramyocardial transplantation of autologous CD34+ stem cells for intractable angina. Circulation. 2007;115:3165–3172. doi: 10.1161/CIRCULATIONAHA.106.687376. [DOI] [PubMed] [Google Scholar]
  • 52.Losordo DW, Henry TD, Davidson C, et al. Intramyocardial, autologous CD34+ cell therapy for refractory angina. Circulation Research. 2011;109:428–436. doi: 10.1161/CIRCRESAHA.111.245993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Wang S, Cui J, Peng W, Lu M. Intracoronary autologous CD34+ stem cell therapy for intractable angina. Cardiology. 2010;117(2):140–147. doi: 10.1159/000320217. [DOI] [PubMed] [Google Scholar]
  • 54.Perin EC, Dohmann HFR, Borojevic R, et al. Transendocardial, autologous bone marrow cell transplantation for severe, chronic ischemic heart failure. Circulation. 2003:2294–2303. doi: 10.1161/01.CIR.0000070596.30552.8B. [DOI] [PubMed] [Google Scholar]
  • 55.Assmus B, Honold J, Schachinger V, et al. Transcoronary transplantation of progenitor cells after myocardial infarction. New England Journal of Medicine. 2006;355:1222–1232. doi: 10.1056/NEJMoa051779. [DOI] [PubMed] [Google Scholar]
  • 56.Perin EC, Willerson JT, Pepine CJ, et al. 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. JAMA. 2012;307:1717–1726. doi: 10.1001/jama.2012.418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Heldman AW, DiFede DL, Fishman JE, et al. Transendocardial mesenchymal stem cells and mononuclear bone marrow cells for ischemic cardiomyopathy: the TAC-HFT randomized trial. JAMA. 2014;311:62–73. doi: 10.1001/jama.2013.282909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ascheim DD, Gelijns AC, Goldstein D, et al. Mesenchymal precursor cells as adjunctive therapy in recipients of contemporary left ventricular assist devices. Circulation. 2014;129(22):2287–2296. doi: 10.1161/CIRCULATIONAHA.113.007412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Perin EC, Sanz-ruiz R, Sánchez PL, Lasso J. Adipose-derived regenerative cells in patients with ischemic cardiomyopathy: the PRECISE Trial. American Heart Journal. 2014;168:88–95. e82. doi: 10.1016/j.ahj.2014.03.022. [DOI] [PubMed] [Google Scholar]
  • 60.Bolli R, Chugh AR, D’Amario D, et al. Cardiac stem cells in patients with ischaemic cardiomyopathy (SCIPIO): initial results of a randomised phase 1 trial. The Lancet. 2011;378:1847–1857. doi: 10.1016/S0140-6736(11)61590-0. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 61.Nowbar AN, Mielewczik M, Karavassilis M, et al. Discrepancies in autologous bone marrow stem cell trials and enhancement of ejection fraction (DAMASCENE): weighted regression and meta-analysis. BMJ. 2014;348:g2688. doi: 10.1136/bmj.g2688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Malliaras K, Marban E. Moving beyond surrogate endpoints in cell therapy trials for heart disease. Stem Cells Translational Medicine. 2014;3(1):2–6. doi: 10.5966/sctm.2013-0104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Assmus B, Schachinger V, Teupe C, et al. Transplantation of progenitor cells and regeneration enhancement in acute myocardial infarction (TOPCARE-AMI) Circulation. 2002;106:3009–3017. doi: 10.1161/01.cir.0000043246.74879.cd. [DOI] [PubMed] [Google Scholar]
  • 64.Huikuri HV, Kervinen K, Niemela M, et al. 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(22):2723–2732. doi: 10.1093/eurheartj/ehn436. [DOI] [PubMed] [Google Scholar]
  • 65.Hare JM, Traverse JH, Henry TD, et al. A randomized, double-blind, placebo-controlled, dose-escalation study of intravenous adult human mesenchymal stem cells (Prochymal) after acute myocardial infarction. Journal of the American College of Cardiology. 2009;54:2277–2286. doi: 10.1016/j.jacc.2009.06.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Gyöngyösi M, Lang I, Dettke M, et al. Combined delivery approach of bone marrow mononuclear stem cells early and late after myocardial infarction: the MYSTAR prospective, randomized study. Nature Clinical Practice Cardiovascular Medicine. 2009;6:70–81. doi: 10.1038/ncpcardio1388. [DOI] [PubMed] [Google Scholar]
  • 67.Cao F, Sun D, Li C, et al. Long-term myocardial functional improvement after autologous bone marrow mononuclear cells transplantation in patients with ST-segment elevation myocardial infarction: 4 years follow-up. European Heart Journal. 2009;30:1986–1994. doi: 10.1093/eurheartj/ehp220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Roncalli J, Mouquet F, Piot C, et al. Intracoronary autologous mononucleated bone marrow cell infusion for acute myocardial infarction: Results of the randomized multicenter BONAMI trial. European Heart Journal. 2011;32:1748–1757. doi: 10.1093/eurheartj/ehq455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Houtgraaf JH, Dekker WKD, Dalen BMV, et al. First experience in humans using adipose tissue – derived regenerative cells in the treatment of patients with ST-segment elevation myocardial infarction. Journal of the American College of Cardiology. 2012;59:539–540. doi: 10.1016/j.jacc.2011.09.065. [DOI] [PubMed] [Google Scholar]
  • 70.Ramshorst Jv, Bax JJ, Beeres SLMA, et al. Intramyocardial bone marrow cell injection for chronic myocardial ischemia. JAMA. 2009;301:1997–2004. doi: 10.1001/jama.2009.685. [DOI] [PubMed] [Google Scholar]
  • 71.Menasche P, Hage AA, Desnos M, et al. Novel approaches to LV dysfunction autologous skeletal myoblast transplantation for severe postinfarction left ventricular dysfunction. Journal of the American College of Cardiology. 2003;41:1078–1083. doi: 10.1016/s0735-1097(03)00092-5. [DOI] [PubMed] [Google Scholar]
  • 72.Menasché P, Alfieri O, Janssens S, et al. The Myoblast Autologous Grafting in Ischemic Cardiomyopathy (MAGIC) Trial. Circulation. 2008;117:1189–1200. doi: 10.1161/CIRCULATIONAHA.107.734103. [DOI] [PubMed] [Google Scholar]
  • 73.Perin EC, Willerson JT, Pepine CJ, et al. 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. JAMA. 2012;307(16):1717–1726. doi: 10.1001/jama.2012.418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Bartunek J, Behfar A, Dolatabadi D, et al. The C-CURE randomized clinical trial (Cardiopoietic stem Cell therapy in heart failURE) Journal of the American College of Cardiology. 2013;62:2454–2456. doi: 10.1016/j.jacc.2013.09.014. [DOI] [PubMed] [Google Scholar]
  • 75.Assmus B, Walter DH, Seeger FH, et al. Effect of shock wave-facilitated intracoronary cell therapy on LVEF in patients with chronic heart failure: the CELLWAVE randomized clinical trial. JAMA. 2013;309:1622–1631. doi: 10.1001/jama.2013.3527. [DOI] [PubMed] [Google Scholar]

RESOURCES