Sie sind auf Seite 1von 46

 

 
The Clinical Application of Mesenchymal and Cardiac Stem Cells as a
Therapy for Cardiovascular Disease
Jiyeon Kim, Linda Shapiro, Aidan Flynn
PII:
DOI:
Reference:

S0163-7258(15)00047-9
doi: 10.1016/j.pharmthera.2015.02.003
JPT 6762

To appear in:

Pharmacology and Therapeutics

Received date:
Accepted date:

11 February 2015
11 February 2015

Please cite this article as: Kim, J., Shapiro, L. & Flynn, A., The Clinical Application of
Mesenchymal and Cardiac Stem Cells as a Therapy for Cardiovascular Disease, Pharmacology and Therapeutics (2015), doi: 10.1016/j.pharmthera.2015.02.003

This is a PDF le of an unedited manuscript that has been accepted for publication.
As a service to our customers we are providing this early version of the manuscript.
The manuscript will undergo copyediting, typesetting, and review of the resulting proof
before it is published in its nal form. Please note that during the production process
errors may be discovered which could aect the content, and all legal disclaimers that
apply to the journal pertain.

ACCEPTED MANUSCRIPT
P&T #22675
Title: The Clinical Application of Mesenchymal and Cardiac Stem Cells as a Therapy for

RI

PT

Cardiovascular Disease

Center for Vascular Biology, University of Connecticut Health

Center, Farmington, CT, 06030;

MA

Author Affiliations:

NU

SC

Authors: Jiyeon Kim Ph.D.1, Linda Shapiro Ph.D.1, Aidan Flynn M.B., Ph.D.2, 3,*

Department of Echocardiography, Division of

TE

Cardiology, Hartford Hospital, Hartford, CT, 06102; 3 Department of Medicine, University

*:

Address

AC
CE
P

of Connecticut Health Center, Farmington, CT, 06030

for

Correspondence:

Aidan

Flynn

M.B.,

Ph.D.,

Department

of

Echocardiography, S201, Hartford Hospital, Hartford, CT, 06102. Fax: 860-545-5631;


Tel: 860-972-2976; E-mail: aidan.flynn@hhchealth.org.

Word Count: 4849

ACCEPTED MANUSCRIPT

PT

ABSTRACT:

RI

Cardiovascular disease (CVD) can be separated into two broad etiological categories,
based on the presence or absence of ischemia as a causative factor. In both ischemic

SC

and non-ischemic heart disease, myocardial dysfunction or damage frequently results in

NU

the development of heart failure, characterized by dyspnea, fatigue and reduced


survival. As one of the least regenerative organs in the human body, current standards

MA

of care are limited to mitigating loss and preventing recurrence of damage, rather than
stimulating actual regeneration of functional heart tissue. Cell based therapies using

TE

progenitor cells from bone marrow and the heart itself have been evaluated in
preclinical models, and have demonstrated some promise. Accordingly, several clinical

AC
CE
P

trials using autologous stem and progenitor cells have demonstrated that these cells
can be used safely in humans, and some studies suggest that they may improve
relevant clinical parameters in patients with heart disease. Two specific cell populations
that are particularly promising are the bone marrow derived mesenchymal stem cell
(MSC) and the heart muscle derived cardiac stem cell (CSC). This review will
summarize preclinical studies evaluating these stem cell populations and will discuss
the clinical application of these cells in contemporary clinical trials, and potential future
investigations.

ACCEPTED MANUSCRIPT

PT

Key Words: Cardiovascular disease; therapy; mesenchymal stem cell; cardiac stem

AC
CE
P

TE

MA

NU

SC

RI

cell; pre-conditioning; clinical trial

ACCEPTED MANUSCRIPT

PT

Table of Contents:

RI

1. Introduction

SC

2. Stem Cell Populations

NU

2.1 Bone Marrow Derived Mononuclear Cells (BMMNC)


2.2 Bone Marrow Derived Mesenchymal Stem Cells (BMMSC)

MA

2.3 Preconditioned Mesenchymal Stem Cells

TE

2.4 Cardiac Stem Cells

3. Clinical Trials of Stem Cell Populations

AC
CE
P

3.1 Mesenchymal Stem Cells

3.2 Preconditioned Mesenchymal Stem Cells


3.3 Cardiac Stem Cells

4. Future Directions

ACCEPTED MANUSCRIPT

Abbreviations:

PT

BMMNC: Bone Marrow Derived Mononuclear Cells

RI

CSC: Cardiac stem cell


CVD: Cardiovascular disease

AC
CE
P

TE

MA

NU

SC

MSC: Mesenchymal stem cell

ACCEPTED MANUSCRIPT

PT

1. Introduction

RI

Cardiovascular disease (CVD) is the leading cause of death in the United States and
worldwide (Heidenreich et al., 2011; Lopez et al., 2006). Currently affecting one in three

SC

adults, or over 70 million people, in the United States alone, the projected prevalence of

NU

CVD in the US by 2030 is over 40%, costing the nation over $1 trillion in direct and
indirect costs (Heidenreich et al., 2011; Thom et al., 2006). The most common

MA

manifestations of CVD are hypertension, coronary artery disease, heart failure and
stroke. Heart disease and stroke account for almost 35% of the 2.4 million deaths in the

TE

US in 2003, making them the first and third leading causes of mortality (Go et al., 2014).
Indeed, the morbidity and mortality associated with CVD is projected to worsen with the

AC
CE
P

current trends in obesity and aging of the population unless better preventative and
therapeutic modalities can be implemented (Hoyert et al., 2006).
Despite significant advances in the management of heart disease over the last two
decades, there remains an obviously unmet clinical need in treating this large and
expanding patient population. As part of a multi-faceted approach, the field of cell
therapy for advanced heart failure aims to provide an effective therapeutic option with
the goal of improving quality of life and perhaps reducing mortality. Cells such as bone
marrow derived mesenchymal stem cells (BMMSCs), autologous cardiac stem cells
(CSCs), embryonic stem cells (ESCs) and induced pluripotent stem (iPS) cells have
been used in experimental models and show improvement in cardiac function and/or
secretion of bioactive paracrine molecules that are pro-angiogenic and cardio-protective

ACCEPTED MANUSCRIPT
(Williams et al, 2014, Zhou et al., 2012, Yoshida and Yamanaka, 2011). While each of
these cell types have been extensively studied in animal models, until recently, most of

PT

the phase I and II trials have tested the safety and efficacy of autologous adult stem cell

RI

therapies only. These early studies unanimously demonstrate the safety of using
patient-derived cells (Strauer and Steinhoff, 2011). Moreover, some show improvement

SC

in relevant clinical parameters as well as patient-described quality of life (Heldman, et

NU

al. 2014). Further clinical investigation is underway to determine the ideal cell type or
combination of cell types as well as optimal dosage in larger scale phase II/III studies

MA

based on these initial investigations. The purpose of this review is 1) to provide an


overview of the bone marrow derived mononuclear cell (BMMNC) and its use in clinical

TE

trials, and 2) to describe the potential therapeutic option that is offered by the cell
populations for which there is the strongest evidence; namely the bone marrow derived

AC
CE
P

mesenchymal stem cell (MSC) and the heart muscle derived cardiac stem cell (CSC).

2. Stem Cell Populations

2.1 Bone Marrow Derived Mononuclear Cells (BMMNC)

Adult bone marrow harbors multiple cell populations including stem cells and various
lineage committed cell types. Unfractionated bone marrow derived mononuclear cells
(BMMNC) include a small number of stem cells, and a larger number of cells at different
levels of maturation. Early preclinical studies were very promising in demonstrating the

ACCEPTED MANUSCRIPT
effect of BMMNC in treating acute myocardial infarction (AMI), leading to the rapid
transition to phase I/II clinical trials (Orlic et al. 2001, Yoshioka et al. 2005, Chen et al.

PT

2004, Wollert et al. 2004). Encouraging results were reported in two early studies, the

RI

BOne marrOw transfer to enhance ST-elevation infarct regeneration (BOOST) trial


(Wollert et al., 2004), and the Reinfusion of Enriched Progenitor Cells and Infarct

SC

Remodeling in Acute Myocardial Infarction (REPAIR-AMI) trial (Schachinger et al.,

NU

2006). A significant improvement in left ventricular ejection fraction (LVEF) was


observed at 6 month and 4 month follow-up, respectively, with no signals of adverse

MA

events. These findings provided a basis for continuing to investigate the efficacy and
safety of cell therapy, and led on to additional studies evaluating the optimal time of

TE

delivery, the optimal dose of the cell product, and the optimal method for delivery.
Recently, the National Heart, Lung and Blood Institute (NHLBI) Cardiovascular Cell

AC
CE
P

Therapy Research Network (CCTRN) has coordinated trials evaluating the effect of
BMMNCs as an intracoronary therapy. In the Transplantation in Myocardial Infarction
Evaluation (TIME) trial (Traverse, et al., 2012) and LateTIME trial (Traverse, et al.,
2011), two timepoints were selected the first was 3-7 days after intervention for an
acute anterior wall ST-segment elevation myocardial infarction, and the second was 2-3
weeks after such intervention. In these trials, a significant improvement in LVEF was not
observed at 6 month follow-up in either study. As this is in contrast to the findings of
BOOST and REPAIR-AMI, it has caused some re-evaluation of the efficacy of this
particular cell population. One particular consideration is that using LVEF as a measure
of effectiveness may not be the most prudent approach. As the cell product generates
its greatest effect in the border zone of the infarct, anticipating a global improvement

ACCEPTED MANUSCRIPT
may not be realistic, nor may it be necessary for a benefit to be derived. Perhaps more
importantly, BMMNC represent a broad selection of cell types, and this unfractionated

PT

cell population may not represent the optimal cell therapy. Individual constituents are

RI

now considered more likely to be the preferred, or active, therapy. Indeed, in the TACHFT trial, direct comparison of BMMNC and MSC versus placebo demonstrated

SC

improvement in infarct size, regional myocardial function, and 6 minute walk distance

NU

with MSC treatment only, even though treatment with either cell type reduced patient
reported Minnesota Living with Heart Failure scores (Heldman, et al. 2014). Although

MA

the authors acknowledge limitations in sample size due to the multiple parameters being
compared, this study implies that administration of specific cell types within the BMMNC

TE

population will increase therapeutic potency. Of the different constituents that have
been described in the literature, there is increasing evidence that the mesenchymal

attention.

AC
CE
P

stem cell (MSC) is most associated with efficacy, and thus has become a major focus of

2.2 Bone Marrow Derived Mesenchymal Stem Cells (BMMSC)

The MSC is a cell population that was originally isolated from the bone marrow, but
has since been identified in many tissues, including adipose tissue and umbilical cord
blood (da Silva Meirelles et al., 2006). The characteristics which define MSCs are the
ability to adhere to plastic, the expression of surface antigens CD73 and CD90 (and the
absence of CD34 and CD45), and the ability, under appropriate conditions, to
differentiate into osteoblasts, chondrocytes and adipocytes. It has been shown that

ACCEPTED MANUSCRIPT
MSCs may differentiate into cardiomyocytes in vitro and in vivo (Amado et al., 2005;
Miyahara et al., 2006; Shake et al., 2002), albeit with inconsistent rates of differentiation

PT

and engraftment (Shake et al., 2002; Toma et al., 2002). MSC are immune-privileged,

RI

as they do not express MHC class II molecules or Fas ligand and other co-stimulatory
molecules. This allows allogeneic use (Pittenger et al., 1999), with the encouraging

SC

possibility of an off-the-shelf product.

NU

The advantages of using MSCs are widely recognized. Cell isolation from bone
marrow and subsequent transfusion has been clinically practiced safely for years to

MA

treat other conditions. MSCs survive and differentiate in allograft and xenograft animal
models without immune suppression (Amado et al., 2005; Atoui and Chiu, 2012; Le

Blanc and Ringden, 2007; Le Blanc et al., 2003; Nauta and Fibbe, 2007), allowing an

TE

allogeneic donor if the patients bone marrow is compromised by age or other co-

AC
CE
P

morbidities. Unlike other stem cell-derived therapies, MSCs do not have to be


differentiated into a mature cell type prior to administration, and also have powerful
homing capabilities to sites of injury after intravenous administration (Chamberlain et al.,
2007). Indeed, a number of studies have demonstrated the ability of intravenously
administered MSCs to migrate specifically to areas of inflammation from ischemic injury
to provide measurable benefit (Kawada et al., 2004; Price et al., 2006; Wu et al., 2003).
Thus, the ease of isolation and administration coupled with that fact that these cells are
immune-privileged and have been used safely in patients for years make MSC-based
regenerative approaches a very appealing therapeutic.
A large number of preclinical investigations have been performed using MSC, and
nearly uniformly demonstrate a significant beneficial effect on cardiac structure and

10

ACCEPTED MANUSCRIPT
function. For example, Noiseux et al demonstrated that administration of 5x10 5
allogeneic MSC to the border zone of a myocardial infarction in mice resulted in a

PT

significant reduction in infarct size, and significant improvements in left ventricular

RI

volumes, despite transient engraftment and infrequent cellular fusion (Noiseux, et al.,
2006). Similarly, in a large animal model, Quevedo et al demonstrate that administration

SC

of allogeneic MSC to a swine model of chronically infarcted myocardium resulted in

NU

improvements in regional contractility and myocardial blood flow, as well as evidence


supporting engraftment, differentiation and enhanced survival (Quevedo, et al., 2009).

MA

In a similarly well-designed and rigorously performed large animal study, Williams et al


show significant reductions in scar size, improvements in ejection fraction and absence

TE

of further adverse LV chamber remodeling (all quantified by serial cardiac MRI) in


animals receiving IM injections of MSC at 3 months post-MI as compared to controls

AC
CE
P

(Williams, et al. 2013). These studies, and many others, demonstrate that the functional
benefit of MSC therapy is considerable. A consistent observation is that the
considerable functional benefit appears to be out of proportion to what would be
expected from the rates of engraftment and differentiation. This observation has led to
the theory that cell therapy produces factors that act locally or systemically to favorably
impact recovery the paracrine hypothesis.
A wide variety of cytokines, chemokines and growth factors are produced by MSC,
and many are involved in restoring cardiac function or regenerating myocardial tissue.
Administration of conditioned medium from MSC generates similar beneficial effects to
the administration of the cell product itself (Gnecchi, et al., 2005). Factors such as basic
fibroblast growth factor (bFGF), hepatocyte growth factor (HGF) and insulin-like growth

11

ACCEPTED MANUSCRIPT
factor (IGF)-1 have been used to pre-condition MSC, and enhance their reparative
effect (Bartunek, et al., 2007). Additional paracrine factors that are secreted, and have

PT

beneficial effects are vascular endothelial growth factor (VEGF), transforming growth

RI

factor (TGF)-, secreted frizzled-related protein (SFRP)-1 and SFRP-2 (Gnecchi, et al.,
2008). The effects of these, and other paracrine factors, extend beyond their

SC

cardioprotective effects, and include favorable effects on cardiac metabolism,

NU

contractility, regeneration and neovascularization.

MA

2.3 Preconditioned Mesenchymal Stem Cells

TE

As the safety and efficacy of MSCs has clearly been demonstrated by preclinical
work for a variety of organ systems, there has been an increasing focus on enhancing

AC
CE
P

the benefit of MSC therapy. Combining MSC and pharmacotherapy (Yang et al., 2009),
genetically modifying MSCs (Li et al., 2007; Noiseux et al., 2006; Tang et al., 2010) and
pre-conditioning MSCs (Wu et al., 2011; Mylotte et al, 2008) are approaches that are
being explored to augment MSC-mediated cardiac repair. In the realm of combination
MSCs/pharmacologic therapy, a few groups are reporting mixed results. For instance,
co-administration of MSCs and simvastatin improves systolic wall thickening and MSC
engraftment (Yang et al., 2009). On the other hand, supplementing MSC infusion with
hepatocyte growth factor (HGF) does not seem to improve any measure of outcome
over cell therapy alone (Yang et al., 2006); however, this may not be surprising given
that the effective synergy between a cell and a drug may be dose and time dependent,
and may require more than one factor.

12

ACCEPTED MANUSCRIPT
Separate from pharmacologic modification is another process, whereby MSCs
are genetically modified with viruses to express certain enzymes, cytokines or cell

PT

surface molecules that can affect their survival, engraftment or function (Figure 1). The

RI

genes being studied in animal models include anti-apoptotic factors such as Bcl2 and
Akt, angiogenic factors such as VEGF and Ang1, and the stem cell homing factor SDF-

SC

1 (Li et al., 2007; Noiseux et al., 2006; Tang et al., 2010). For instance, MSCs

NU

transfected to overexpress Akt, a kinase involved in cellular activities including


apoptosis and cell proliferation, seem to confer a possible myocardial protective function

MA

(Lim et al., 2006; Mangi et al., 2003; Matsui et al., 2001; Noiseux et al., 2006). Akttransduced MSC secrete a number of proteins in response to hypoxia, including the

TE

recently described Hypoxia and Akt induced Stem cell Factor (HASF). This has been
shown to be an important mediator of cardioprotection following ischemic injury (Huang

AC
CE
P

J, et al., 2014). These studies indicate improved ejection fraction and reduced infarct
size with the administration of the Akt-overexpressing MSCs over that seen with
injection of control MSCs. Furthermore, MSCs engineered to express combinations of
gene products such as Akt and Ang1 are also showing promise in animal models
(Shujia et al., 2008). Interestingly, the combined overexpression of VEGF and SDF-1 in
MSCs also seem to work via Akt activation (Tang et al., 2010). MSCs transfected to
express heme-oxygenase 1 (HO-1), an enzyme that improves MSC tolerance to
hypoxia, and subsequently infused into a cardiac ischemia-reperfusion model
demonstrated improved ejection fraction and lower end systolic volume than plasmid
transfected controls (Tang et al., 2005). Further histologic and molecular analyses of
hearts treated with HO-1 overexpressing MSCs demonstrated increased capillary and

13

ACCEPTED MANUSCRIPT
arteriolar density as well as a cytokine profile that is anti-inflammatory and proangiogenic (Tang et al., 2005). As a whole, these data appear promising, but the safety

PT

of these cells must be carefully and thoroughly addressed before use in humans.

RI

Preconditioning MSCs with physical and chemical manipulations avoids the use
of viral transfection and also may induce certain pathways that improve engraftment and

SC

survival of transplanted cells (Figure 2). Common approaches include heat shock,

NU

hypoxia, anoxia and treatment with pharmacologics (Wu et al., 2011; Mylotte et al,
2008). Brief treatment with hypoxia is known to induce the hypoxia-inducible factor (HIF)

MA

1 and SDF1 pathways which both limit infarct size and improve angiogenesis in the
heart, but interestingly, hypoxia treated MSCs also demonstrate increased survival and

TE

engraftment in the heart (Chacko et al., 2010; Hu et al., 2008; Tang et al., 2009). Brief
periods of anoxia, or ischemia, is also known to reduce apoptosis through activation of

AC
CE
P

Akt and HIF1 pathways (Fenton et al., 2005; Kim et al., 2009). Pretreatment of MSCs
with trimetazidine, a fatty acid oxidation inhibitor commonly used to treat angina, seems
to improve myocardial recovery and decrease tissue fibrosis again by stimulating HIF1,
Akt and Bcl-2 (Wisel et al., 2009). Culturing MSCs with growth factors, such as basic
fibroblast growth factor (bFGF), insulin-like growth factor (IGF)-1 and bone
morphogenetic protein 2 (BMP2) can also improve myocardial repair in rat models of MI
(Hahn et al., 2008). Finally, heat-shocking cells has been shown to increase cellular
fortitude by activating certain heat shock proteins; however, the precise pathways that
get activated in MSCs and how they converge to enhance MSC-mediated therapy have
yet to be determined.

14

ACCEPTED MANUSCRIPT
2.4 Cardiac Stem Cells (CSC)

PT

In addition to BMMSC being a promising cell therapeutic, other cell populations

RI

continue to be evaluated. Adult mammalian myocardium harbors endogenous


populations of cells that can be stimulated in certain circumstances to generate new

SC

cardiomyocytes. One hypothesis is that these are cardiac stem cells (CSC) that have

NU

the ability to differentiate to cardiomyocytes as well as other supporting cell types such
as endothelium and vascular smooth muscle (Beltrami et al., 2003; Laugwitz et al.,

MA

2005; Martin et al., 2004; Smart et al., 2011). A second suggestion is that these cells
are existing cardiomyocytes re-entering the cell cycle, and that they replicate, thus

TE

generating new cardiomyocytes (Senyo et al., 2013). These cells are understood to
maintain normal homeostasis in the heart. With an annual rate of turnover under 1%,

AC
CE
P

endogenous CSCs are unable to completely remedy the massive loss of tissue after
myocardial infarction (Malliaras et al., 2013; Mollova et al., 2013). Because these cells
are already located in the heart and are primed for cardiac repair, protocols to enhance
the endogenous activity of these cells or expand these cells in vitro before re-implanting
them in the heart are currently being tested. Indeed a number of animal studies indicate
that the administration of CSCs can slow left ventricular remodeling and cardiac
improve function after ischemic injury (Beltrami et al., 2003; Linke et al., 2005).
Various groups have identified CSCs with c-kit, Sca1 or Isl1 expression, the
ability to efflux Hoechst dye, or even as a side population upon flow cytometric analysis
(Beltrami et al., 2003; Breitbach et al., 2007; Laugwitz et al., 2005; Martin et al., 2004;
Oh et al., 2003; Pfister et al., 2005). All of these putative cardiac stem cells have been

15

ACCEPTED MANUSCRIPT
isolated from rodent hearts and expanded in vitro under varying culture conditions
before assessing cardiac differentiation via protein expression or rescue of animals with

PT

myocardial infarction and the results have been encouraging. Isl1 positive cells have

RI

been shown to form not only myocardial cells, but also endothelial, endocardial, and
smooth muscle lineages in the embryonic heart, but appear to be restricted to the right

SC

atrium in the adult heart (Laugwitz et al., 2005). Their location in the atrium limits the

NU

ability to exploit them endogenously; however, ex vivo expansion or generation of


similar Isl1 lineage cells from pluripotent stem cells still hold promise (Moretti et al.,

MA

2006). Nkx2.5 expressing cells have also been isolated from the heart, but they seem to
be bipotent progenitors, producing only cardiomyocytes and smooth muscle cells, that

are downstream of the Isl1 population (Yi et al., 2010). The epicardium has also been

TE

found to harbor a quiescent population of Wilms tumor 1 (Wt1) positive cardiac stem

AC
CE
P

cells (Smart et al., 2011). In mice, these cells seemed to increase cardiomyocyte
generation after activation with thymosin 4 and myocardial injury, but later studies
seem to favor a pro-angiogenic mechanism rather than replacement of actual muscle
(Zhou et al., 2012). Further studies are being conducted to elucidate this phenomenon.
While many groups are working to delineate individual cardiac lineages that
reconstitute the heart, other groups have chosen to use cardiospheres, heterogeneous
cell aggregates that can be grown as spheroids in suspension cultures after isolation
from heart tissue. This amalgam of cells has been demonstrated to express markers of
stem-ness such as c-kit and to improve cardiac performance in animal models, which
has prompted human clinical trials using autologous cardiosphere derived cells (Makkar
et al., 2012; Messina et al., 2004). Although the exact types of cells and the correct

16

ACCEPTED MANUSCRIPT
ratios to use are not precisely known, researchers and clinicians are actively pursuing

PT

this line of inquiry not only in animal models, but also in early human trials.

RI

3. Clinical Trials of Stem Cell Populations

NU

SC

3.1 Mesenchymal Stem Cells

As discussed earlier, contemporary clinical trials of BMMNC have produced results

MA

which suggest that unfractionated marrow may not generate the effect necessary for
transition to widespread clinical use. Several studies have evaluated the BMMSC, in the

TE

anticipation that they will have a greater clinical benefit. Chen et al. administered 48-60
billion bone marrow derived MSCs by intracoronary injection into 34 patients and

AC
CE
P

reported a 14% higher ejection fraction compared to placebo-treated controls (Chen et


al., 2004). These patients exhibited a 10% improvement between 3-6 months after
treatment. More recently, the PercutaneOus StEm Cell Injection Delivery Effects On
Neo-myogenesis (POSEIDON) and Prospective Randomized Study of Mesenchymal
Stem Cell Therapy in Patients Undergoing Cardiac Surgery (PROMETHEUS) trials
have utilized a transendocardial or intramyocardial method of delivering MSCs directly
to sites of injury (Karantalis et al., 2014; Suncion et al., 2014). The POSEIDON trial
tested the ability of autologous and allogeneic MSCs to promote cardiac recovery
following transendocardial stem cell injection (TESI) (Suncion et al., 2014). Using
multidetector computed tomography (MDCT) and biplane left ventriculography, this
study reports scar size reductions of approximately 44% in treated groups versus only

17

ACCEPTED MANUSCRIPT
25% in untreated groups with the greatest improvement seen in those who received 20
million

autologous

MSCs.

Furthermore,

MSC-treated

myocardial

segments

PT

demonstrated an over 40% improvement in segmental ejection fraction whereas no

RI

improvement was detected in untreated segments. Thus, this study clearly


demonstrates the importance of the location and delivery of MSCs and also indicates

SC

the safety of using allogeneic MSCs. The smaller PROMETHEUS trial injected

NU

autologous MSCs into akinetic or hypokinetic areas of the hearts that were unsuitable
for surgical revascularization during CABG in 6 patients. Cardiac MRI analysis

MA

demonstrated increased ejection fraction as well as scar reduction and contractile


improvement in areas that received MSC injection over those areas that were surgically

TE

reperfused (Karantalis et al., 2014). While the lack of a placebo control group and the
very small number of recruited patients prevent the establishment of definitive

AC
CE
P

improvement with this treatment, this trial also seems to indicate potential benefits of
MSCs injected directly into non-revascularized myocardium.
Cumulatively, these studies demonstrate the safety and efficacy of using BMDCs
and MSCs for cell therapy. In fact, a meta-analysis of the 16 largest randomized
controlled studies shows variable decreases in infarct size and an average increase in
left ventricular ejection fraction of 11.3% (Strauer and Steinhoff, 2011). Similarly, a
meta-analysis by Jeevanantham et al demonstrated that bone marrow derived cells
were associated with improved ejection fraction (an absolute improvement of almost
4%) and smaller infarct size, as well as reductions in mortality and the incidence of
recurrent myocardial infarction (Jeevanantham et al., 2012). A summary of clinical trials
and their results is presented in Table 1.

18

ACCEPTED MANUSCRIPT

PT

3.2 Preconditioned Mesenchymal Stem Cells

RI

A further development in MSC therapy is the pretreatment of MSCs with certain growth
factors to enhance cardioprotective functions. The Cardiopoietic stem Cell therapy in

SC

heart failure (C-CURE) trial has tested the ability of a cardiogenic cocktail to enhance

NU

the therapeutic benefits to the heart rendered by autologous MSCs (Bartunek et al.,
2013). The rationale for this study stems from the fact that CSCs from heart failure

MA

patients may be impaired and the MSCs from the bone marrow can be coaxed to adopt
a cardiopoietic lineage, which improves therapeutic benefit (Behfar et al., 2010). The C-

TE

CURE trial treated 21 patients suffering from heart failure with an average number of
over 700 million cells in 9-26 electromechanically guided endomyocardial injections. No

AC
CE
P

adverse events or systemic toxicity was observed. Moreover, significant improvements


in left ventricular ejection fraction, end-systolic volume and 6-minute walking test were
reported. The Safety and Efficacy of Autologous Cardiopoietic Cells for Treatment of
Ischemic Heart Failure (CHART-1) trial is powered to evaluate the efficacy of this
therapy, and is currently enrolling patients.

3.3 Cardiac Stem Cells

In line with the promising preclinical work on CSC, this cell type has already been
investigated in clinical studies. There are three clinical trials using cardiac stem cells for
treatment of ischemic cardiomyopathy and even though they use slightly different

19

ACCEPTED MANUSCRIPT
approaches, all report significant improvements in certain measures of cardiac function
(Oldroyd et al., 2012). The CArdiosphere-Derived aUtologous stem CElls to reverse
dySfunction

(CADUCEUS)

trial

evaluated

the

effectiveness

of

PT

ventricUlar

RI

cardiospheres, which, as described above, are clusters of undifferentiated cells


expressing endothelial progenitor markers grown from human heart biopsy subcultures

SC

(Messina et al., 2004). Cardiospheres are heterogenous groups of cells that contain not

NU

only adult CSCs, which are capable of long-term self-renewal and cardiomyocyte
differentiation, but also vascular cells and differentiated progenitor cells. CADUCEUS

MA

analyzed cardiac MRI scans of 25 patients who were given 12.5-25 million autologous
cardiosphere derived cells (Makkar et al., 2012) after successful percutaneous coronary

TE

intervention. The cardiospheres were expanded approximately 36 days in culture from


right ventricular endomyocardial biopsies taken 2-4 weeks after acute myocardial

AC
CE
P

infarction and injected into the previously stented coronary artery between 6-12 weeks
after the heart attack. Despite the lack of improvement in left ventricular ejection fraction
or patient reported outcomes, the scar mass was 7.7% and 12.3% lower at 6 and 12
months respectively and regional wall motion was significantly improved in treated
patients. Serious adverse events were also reported to be three times higher in the
treated group, but the relatively small number of patients prohibited the use of this trial
in ascertaining safety. Furthermore, this study was not blinded due to ethical
considerations surrounding the harvest of cardiac tissue from the control group, but
additional investigations are necessary to determine the safety and potency of
cardiospheres as this initial inquiry seems promising.

20

ACCEPTED MANUSCRIPT
In a slightly different approach, the Stem Cell Infusion in Patients with Ischemic
cardiOmyopathy (SCIPIO) trial isolated autologous CSCs during coronary artery bypass

PT

grafting (CABG) procedures (Bolli et al., 2011). SCIPIO enlisted 23 randomized patients
who had experienced myocardial infarction in the remote past and exhibited an ejection

RI

fraction of under 40%. One million cKit+ lineage- cardiac stem cells (CSCs) were

SC

isolated with magnetic beads from cultures of patient-specific right atrial appendage

NU

tissue and administered via intracoronary infusion one month after CABG. Four months
after this treatment, 14 out of 16 treated patients saw a 24% relative decrease in infarct

MA

size, an 8.2% absolute improvement in left ventricular ejection fraction and reported
improvements in New York Heart Association functional class. The benefits of treatment

TE

were sustained and even increased over time - after one year, eight of these patients
demonstrated an 8% improvement in ejection fraction, which became 12% after 2 years

awaited.

AC
CE
P

(Chugh et al., 2012). Publication of the complete findings of the two-year follow-up is

Analogous to the SCIPIO procedure, the AutoLogous human CArdiac-Derived


stem cell to Treat Ischemic cArdiomyopathy (ALCADIA) trial also harvests patient
cardiac tissue during CABG (Yacoub and Terrovitis, 2013); however, like the
CADUCEUS trial, endomyocardial tissue served as the source of CSCs. With only six
subjects, ALCADIA is the smallest trial and combines the use of stem cells,
bioengineered scaffolds and biologics to create a hybrid therapy. Cells from these
patients were cultured for one month before intramyocardial injection of half a million
cells per kilogram distributed in 20 injection sites, followed by placement of a
biodegradable hydrogel scaffold containing basic fibroblast growth factor (bFGF) over

21

ACCEPTED MANUSCRIPT
those sites. At the 6 month time point, cardiac MRI indicated an increase in ejection
fraction of 12.1%, a 3.3% reduction in infarct size and significant improvement in wall

RI

these results will need to be confirmed in a larger cohort.

PT

motion as well as maximum aerobic exercise capacity. This was a small study and

Lastly, combining the use of MSC and CSC in post-MI treatment may further

SC

enhance the therapeutic effects of each cell type. Indeed, recent work by Williams et al

NU

demonstrate that the combined use of one million human CSCs and 200 million human
MSCs provide greater recovery, almost to baseline, in swine models of anterior wall MI

MA

(Williams AR, et al. 2013). While all stem cell treated animals demonstrated improved
LVEF compared to placebo controls, notably, animals receiving dual cell therapy had 2-

TE

fold greater reductions in scar size (21.1% for CSC/MSC versus 10.4% for CSC alone
or 9.9% for MSC alone) and improved rates of pressure change during diastole. Overall

AC
CE
P

left ventricular chamber dynamics were improved in both the dual therapy and CSC or
MSC alone treated groups. Interestingly, CSC alone treated animals demonstrated
better isovolumic relaxation as compared to controls, while MSC alone treated animals
exhibited improved diastolic compliance, indicating that the enhanced effect of dual
therapy on both systolic and diastolic function may be due to a synergistic effect
between CSC and MSC targeted mechanisms.

4. Future Directions

The heart is one of the most important, but ironically, one of the least
regenerative organs of the body. With the rising trend in heart disease, developing

22

ACCEPTED MANUSCRIPT
methods to enhance cardiac repair has become one of the most important areas of
translational research. Cell therapy continues to be well positioned to fill the void that

PT

currently exists in heart failure management. Before cell therapy can be considered a

RI

widely accepted therapeutic option however, continued research on a variety of fronts is


required (Figure 3). At the forefront of this is an emphasis on safety. To date, no

SC

adverse signals have been identified in the many clinical investigations of cell therapy in

NU

humans. The field must remain vigilant however, as the consequences of unreported or
underreported adverse events would be profound. In light of the promising safety profile

MA

experienced thus far, a particular emphasis is being focused on the optimal cell type. It
appears that CSC and BMMSC are prime candidates, and are certainly worthy of

TE

continued investigation. Indeed, a combination of both cell populations may be more


effective than either one alone, and this consideration remains under investigation.

AC
CE
P

Similarly, pre-conditioning MSC is a hugely promising approach, and further


investigation is eagerly anticipated. Identifying the optimal dose and method of delivery,
as well as the optimal time for delivery are important variables that are being studied.
Perhaps one of the most important advances in the field has been the collaborative
approach that has recently been engendered by CCTRN. This multi-institutional group
of highly respected researchers is ideally positioned to have a very formative role in the
future of cell therapy trials, and aims to address the many remaining questions in an
objective and definitive manner. The evolution of cell therapy for heart disease has
resulted in a refinement of a number of variables that were initially being broadly
investigated particularly the cell population. We will watch with intense interest how
the field will progress over the coming years, with the anticipation that cell therapy will

23

ACCEPTED MANUSCRIPT
become a mainstream treatment for heart disease, free of significant safety concerns,

AC
CE
P

TE

MA

NU

SC

RI

PT

and associated with important functional and perhaps mortality benefits.

24

ACCEPTED MANUSCRIPT
Acknowledgements: The authors would like to acknowledge the assistance of

RI

PT

Amanda Zaleski and Katelyn Zaleski in preparing the illustrations.

AC
CE
P

TE

MA

NU

SC

The authors declare that there are no conflicts of interest.

25

ACCEPTED MANUSCRIPT
Figure Legends:

PT

Figure 1: Pre-Conditioned MSC receive stimuli from various external factors, which

RI

either act directly, or via intermediaries to upregulate genes which act in a protective or

responsible for repair of damaged myocardium.

SC

anti-apoptotic manner. These factors result in production of proteins that are

NU

bFGF: basic fibroblast growth factor; HGF: hepatocyte growth factor; BMP2: bone

MA

morphogenetic protein 2; IGF-1: insulin-like growth factor-1; HIF-1: hypoxia induced


factor-1; SDF-1: stem cell derived factor-1; VEGF: vascular endothelial growth factor;

TE

TGF-: transforming growth factor-; Sfrp: secreted frizzled related peptide.

AC
CE
P

Figure 2: Genetic Modification of MSC. Various factors have been transfected into
MSC, including anti-apoptotic, angiogenic and stem cell homing factors, as well as Akt.
A potentially important factor, secreted frizzled related peptide is produced, as are many
other intermediaries, which result in the secretion of Hypoxia and Akt induced Stem cell
Factor, an important mediator of the reparative process.
HASF: hypoxia and Akt induced stem cell factor; Sfrp: secreted frizzled related peptide;
VEGF: vascular endothelial growth factor; SDF-1: stem cell derived factor-1.

Figure 3: Cardiac regenerative processes are multi-faceted, and approaches to cardiac


repair have included MSC, pre-conditioned MSC, genetically modified MSC and

26

ACCEPTED MANUSCRIPT
CSC/CDC. Questions remain to be answered as to which (or combinations of which) will
be the optimal approach. Some of the remaining questions are highlighted, with safety

PT

and efficacy being the primary factors in identifying the optimal approach.

AC
CE
P

TE

MA

NU

SC

RI

CSC/CDC: cardiac stem cell / cardiospheres; MSC: mesenchymal stem cells

27

ACCEPTED MANUSCRIPT
References

PT

Amado LC, Saliaris AP, Schuleri KH, St John M, Xie JS, Cattaneo S, et al (2005)

RI

Cardiac repair with intramyocardial injection of allogeneic mesenchymal stem


cells after myocardial infarction. Proceedings of the National Academy of

SC

Sciences of the United States of America 102:11474-11479.

NU

Atoui R and Chiu RC (2012) Concise review: immunomodulatory properties of


mesenchymal stem cells in cellular transplantation: update, controversies, and

MA

unknowns. Stem cells translational medicine 1:200-205.


Bartunek J, Croissant JD, Wijns W, Gofflot S, de Lavareille A, Vanderheyden M, et al

TE

(2007) Pretreatment of adult bone marrow mesenchymal stem cells with


cardiomyogenic growth factors and

repair of

the

chronically

infarcted

AC
CE
P

myocardium. Am J Physiol Heart Circ Physiol 292: H1095-104.


Bartunek J, Behfar A, Dolatabadi D, Vanderheyden M, Ostojic M, Dens J, et al (2013)
Cardiopoietic stem cell therapy in heart failure: the C-CURE (Cardiopoietic stem
Cell therapy in heart failURE) multicenter randomized trial with lineage-specified
biologics. Journal of the American College of Cardiology 61:2329-2338.
Behfar A, Yamada S, Crespo-Diaz R, Nesbitt JJ, Rowe LA, Perez-Terzic C, et al (2010)
Guided cardiopoiesis enhances therapeutic benefit of bone marrow human
mesenchymal stem cells in chronic myocardial infarction. Journal of the
American College of Cardiology 56:721-734.

28

ACCEPTED MANUSCRIPT
Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, et al (2003) Adult
cardiac stem cells are multipotent and support myocardial regeneration. Cell

PT

114:763-776.

RI

Bolli R, Chugh AR, D'Amario D, Loughran JH, Stoddard MF, Ikram S, et al (2011)
Cardiac stem cells in patients with ischaemic cardiomyopathy (SCIPIO): initial

SC

results of a randomised phase 1 trial. Lancet 378:1847-1857.

NU

Breitbach M, Bostani T, Roell W, Xia Y, Dewald O, Nygren JM, et al (2007) Potential


risks of bone marrow cell transplantation into infarcted hearts. Blood 110:1362-

MA

1369.

Chacko SM, Ahmed S, Selvendiran K, Kuppusamy ML, Khan M & Kuppusamy P (2010)

TE

Hypoxic preconditioning induces the expression of prosurvival and proangiogenic


markers in mesenchymal stem cells. American journal of physiology Cell

AC
CE
P

physiology 299:C1562-1570.

Chamberlain G, Fox J, Ashton B & Middleton J (2007) Concise review: mesenchymal


stem cells: their phenotype, differentiation capacity, immunological features, and
potential for homing. Stem cells 25:2739-2749.
Chen SL, Fang WW, Ye F, Liu YH, Qian J, Shan SJ, et al (2004) 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 94:92-95.
Chugh AR, Beache GM, Loughran JH, Mewton N, Elmore JB, Kajstura J, et al (2012)
Administration of cardiac stem cells in patients with ischemic cardiomyopathy:

29

ACCEPTED MANUSCRIPT
the SCIPIO trial: surgical aspects and interim analysis of myocardial function and
viability by magnetic resonance. Circulation 126:S54-64.

PT

da Silva Meirelles L, Chagastelles PC & Nardi NB (2006) Mesenchymal stem cells

RI

reside in virtually all post-natal organs and tissues. J Cell Sci ;119(Pt 11):220413.

SC

Fenton RA, Dickson EW & Dobson JG, Jr. (2005) Inhibition of phosphatase activity

NU

enhances preconditioning and limits cell death in the ischemic/reperfused aged


rat heart. Life sciences 77:3375-3388.

MA

Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ, et al, American
Heart Association Statistics C and Stroke Statistics S (2014) Heart disease and

TE

stroke statistics--2014 update: a report from the American Heart Association.


Circulation 129:e28-e292.

AC
CE
P

Gnecchi M, He H, Liang OD, Melo LG, Morello F, Mu H, et al (2005) Paracrine action


accounts for marked protection of ischemic heart by Akt-modified mesenchymal
stem cells. Nat Med11:367-8.
Gnecchi M, Zhang Z, Ni A & Dzau VJ (2008) Paracrine mechanisms in adult stem cell
signaling and therapy. Circ Res 103:1204-19.
Hahn JY, Cho HJ, Kang HJ, Kim TS, Kim MH, Chung JH, et al (2008) Pre-treatment of
mesenchymal stem cells with a combination of growth factors enhances gap
junction formation, cytoprotective effect on cardiomyocytes, and therapeutic
efficacy for myocardial infarction. Journal of the American College of Cardiology
51:933-943.

30

ACCEPTED MANUSCRIPT
Heidenreich PA, Trogdon JG, Khavjou OA, Butler J, Dracup K, Ezekowitz MD, et al
(2011) Forecasting the future of cardiovascular disease in the United States: a

PT

policy statement from the American Heart Association. Circulation 123:933-944.

RI

Heldman AW, DiFede DL, Fishman JE, Zambrano JP, Trachtenberg BH, Karantalis V,
et al (2014) Transendocardial mesenchymal stem cells and mononuclear bone

SC

marrow cells for ischemic cardiomyopathy: the TAC-HFT randomized trial. The

NU

journal of the American Medical Association 311:62-73.


Hoyert DL, Heron MP, Murphy SL & Kung HC (2006) Deaths: final data for 2003.

MA

National vital statistics reports : from the Centers for Disease Control and
Prevention, National Center for Health Statistics, National Vital Statistics System

TE

54:1-120.

Hu X, Yu SP, Fraser JL, Lu Z, Ogle ME, Wang JA et al (2008) Transplantation of

AC
CE
P

hypoxia-preconditioned mesenchymal stem cells improves infarcted heart


function via enhanced survival of implanted cells and angiogenesis. The Journal
of thoracic and cardiovascular surgery 135:799-808.
Huang J, Guo J, Beigi F, Hodgkinson CP, Facundo HT, Zhang Z, et al (2014) HASF is a
stem cell paracrine factor that activates PKC epsilon mediated cytoprotection. J
Mol Cell Cardiol 66:157-64.
Jeevanantham V, Butler M, Saad A, Abdel-Latif A, Zuba-Surma EK & Dawn B (2012)
Adult bone marrow cell therapy improves survival and induces long-term
improvement in cardiac parameters: a systematic review and meta-analysis.
Circulation 126:551-68.

31

ACCEPTED MANUSCRIPT
Karantalis V, DiFede DL, Gerstenblith G, Pham S, Symes J, Zambrano JP, et al (2014)
Autologous mesenchymal stem cells produce concordant improvements in

undergoing

coronary

artery

bypass

grafting:

The

Prospective

RI

patients

PT

regional function, tissue perfusion, and fibrotic burden when administered to

Randomized Study of Mesenchymal Stem Cell Therapy in Patients Undergoing

SC

Cardiac Surgery (PROMETHEUS) trial. Circulation research 114:1302-1310.

NU

Kawada H, Fujita J, Kinjo K, Matsuzaki Y, Tsuma M, Miyatake H, et al (2004)


Nonhematopoietic mesenchymal stem cells can be mobilized and differentiate

MA

into cardiomyocytes after myocardial infarction. Blood 104:3581-3587.


Kim HW, Haider HK, Jiang S & Ashraf M (2009) Ischemic preconditioning augments

TE

survival of stem cells via miR-210 expression by targeting caspase-8-associated


protein 2. The Journal of biological chemistry 284:33161-33168.

AC
CE
P

Laugwitz KL, Moretti A, Lam J, Gruber P, Chen Y, Woodard S, et al (2005) Postnatal


isl1+ cardioblasts enter fully differentiated cardiomyocyte lineages. Nature
433:647-653.

Le Blanc K & Ringden O (2007) Immunomodulation by mesenchymal stem cells and


clinical experience. Journal of internal medicine 262:509-525.
Le Blanc K, Tammik C, Rosendahl K, Zetterberg E & Ringden O (2003) HLA expression
and immunologic properties of differentiated and undifferentiated mesenchymal
stem cells. Experimental hematology 31:890-896.
Li W, Ma N, Ong LL, Nesselmann C, Klopsch C, Ladilov Y, et al (2007) Bcl-2
engineered MSCs inhibited apoptosis and improved heart function. Stem cells
25:2118-2127.

32

ACCEPTED MANUSCRIPT
Lim SY, Kim YS, Ahn Y, Jeong MH, Hong MH, Joo SY, et al (2006) The effects of
mesenchymal stem cells transduced with Akt in a porcine myocardial infarction

PT

model. Cardiovascular research 70:530-542.

RI

Linke A, Muller P, Nurzynska D, Casarsa C, Torella D, Nascimbene A, et al (2005)


Stem cells in the dog heart are self-renewing, clonogenic, and multipotent and

SC

regenerate infarcted myocardium, improving cardiac function. Proceedings of the

NU

National Academy of Sciences of the United States of America 102:8966-8971.


Lopez AD, Mathers CD, Ezzati M, Jamison DT & Murray CJ (2006) Global and regional

data. Lancet 367:1747-1757.

MA

burden of disease and risk factors, 2001: systematic analysis of population health

TE

Makkar RR, Smith RR, Cheng K, Malliaras K, Thomson LE, Berman D, et al (2012)
Intracoronary cardiosphere-derived cells for heart regeneration after myocardial

AC
CE
P

infarction (CADUCEUS): a prospective, randomised phase 1 trial. Lancet


379:895-904.

Malliaras K, Zhang Y, Seinfeld J, Galang G, Tseliou E, Cheng K, et al (2013)


Cardiomyocyte proliferation and progenitor cell recruitment underlie therapeutic
regeneration after myocardial infarction in the adult mouse heart. EMBO
molecular medicine 5:191-209.
Mangi AA, Noiseux N, Kong D, He H, Rezvani M, Ingwall JS et al (2003) Mesenchymal
stem cells modified with Akt prevent remodeling and restore performance of
infarcted hearts. Nature medicine 9:1195-1201.
Martin CM, Meeson AP, Robertson SM, Hawke TJ, Richardson JA, Bates S, et al
(2004) Persistent expression of the ATP-binding cassette transporter, Abcg2,

33

ACCEPTED MANUSCRIPT
identifies cardiac SP cells in the developing and adult heart. Developmental
biology 265:262-275.

PT

Matsui T, Tao J, del Monte F, Lee KH, Li L, Picard M, et al (2001) Akt activation

RI

preserves cardiac function and prevents injury after transient cardiac ischemia in
vivo. Circulation 104:330-335.

SC

Messina E, De Angelis L, Frati G, Morrone S, Chimenti S, Fiordaliso F, et al (2004)

NU

Isolation and expansion of adult cardiac stem cells from human and murine
heart. Circulation research 95:911-921.

MA

Miyahara Y, Nagaya N, Kataoka M, Yanagawa B, Tanaka K, Hao H, et al (2006)


Monolayered mesenchymal stem cells repair scarred myocardium after

TE

myocardial infarction. Nature medicine 12:459-465.


Mollova M, Bersell K, Walsh S, Savla J, Das LT, Park SY, et al (2013) Cardiomyocyte

AC
CE
P

proliferation contributes to heart growth in young humans. Proceedings of the


National Academy of Sciences of the United States of America 110:1446-1451.
Moretti A, Caron L, Nakano A, Lam JT, Bernshausen A, Chen Y, et al (2006)
Multipotent embryonic isl1+ progenitor cells lead to cardiac, smooth muscle, and
endothelial cell diversification. Cell 127:1151-1165.
Mylotte LA, Duffy AM, Murphy M, O'Brien T, Samali A, Barry F, et al (2008) Metabolic
flexibility permits mesenchymal stem cell survival in an ischemic environment.
Stem Cells 26:1325-36.
Nauta AJ & Fibbe WE (2007) Immunomodulatory properties of mesenchymal stromal
cells. Blood 110:3499-3506.

34

ACCEPTED MANUSCRIPT
Noiseux N, Gnecchi M, Lopez-Ilasaca M, Zhang L, Solomon SD, Deb A, et al (2006)
Mesenchymal stem cells overexpressing Akt dramatically repair infarcted

PT

myocardium and improve cardiac function despite infrequent cellular fusion or

RI

differentiation. Molecular therapy : the journal of the American Society of Gene


Therapy 14:840-850.

SC

Oh H, Bradfute SB, Gallardo TD, Nakamura T, Gaussin V, Mishina Y, et al (2003)

NU

Cardiac progenitor cells from adult myocardium: homing, differentiation, and


fusion after infarction. Proceedings of the National Academy of Sciences of the

MA

United States of America 100:12313-12318.

Oldroyd KG, Berry C & Bartunek J (2012) Myocardial repair and regeneration: bone

TE

marrow or cardiac stem cells? Molecular therapy : the journal of the American
Society of Gene Therapy 20:1102-1105.

AC
CE
P

Pfister O, Mouquet F, Jain M, Summer R, Helmes M, Fine A, et al (2005) CD31- but Not
CD31+ cardiac

side population

cells exhibit

functional cardiomyogenic

differentiation. Circulation research 97:52-61.


Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al (1999)
Multilineage potential of adult human mesenchymal stem cells. Science 284:143147.
Price MJ, Chou CC, Frantzen M, Miyamoto T, Kar S, Lee S, et al (2006) Intravenous
mesenchymal stem cell therapy early after reperfused acute myocardial infarction
improves left ventricular function and alters electrophysiologic properties.
International journal of cardiology 111:231-239.

35

ACCEPTED MANUSCRIPT
Quevedo HC, Hatzistergos KE, Oskouei BN, Feigenbaum GS, Rodriguez JE, Valdes D,
et al (2009) Allogeneic mesenchymal stem cells restore cardiac function in

PT

chronic ischemic cardiomyopathy via trilineage differentiating capacity. Proc Natl

RI

Acad Sci USA;106:14022-7.

Schchinger V, Erbs S, Elssser A, Haberbosch W, Hambrecht R, Hlschermann H, et

SC

al; REPAIR-AMI Investigators (2006) Intracoronary bone marrow-derived

NU

progenitor cells in acute myocardial infarction. N Engl J Med 355:1210-21.


Senyo SE, Steinhauser ML, Pizzimenti CL, Yang VK, Cai L, Wang M, et al (2013)

MA

Mammalian heart renewal by pre-existing cardiomyocytes. Nature 493:433-436.


Shake JG, Gruber PJ, Baumgartner WA, Senechal G, Meyers J, Redmond JM, et al

TE

(2002) Mesenchymal stem cell implantation in a swine myocardial infarct model:


engraftment and functional effects. The Annals of thoracic surgery 73:1919-1925;

AC
CE
P

discussion 1926.

Shujia J, Haider HK, Idris NM, Lu G & Ashraf M (2008) Stable therapeutic effects of
mesenchymal stem cell-based multiple gene delivery for cardiac repair.
Cardiovascular research 77:525-533.
Smart N, Bollini S, Dube KN, Vieira JM, Zhou B, Davidson S, et al (2011) De novo
cardiomyocytes from within the activated adult heart after injury. Nature 474:640644.
Strauer BE & Steinhoff G (2011) 10 years of intracoronary and intramyocardial bone
marrow stem cell therapy of the heart: from the methodological origin to clinical
practice. Journal of the American College of Cardiology 58:1095-1104.

36

ACCEPTED MANUSCRIPT
Suncion VY, Ghersin E, Fishman JE, Zambrano JP, Karantalis V, Mandel N, et al
(2014) Does transendocardial injection of mesenchymal stem cells improve

PT

myocardial function locally or globally?: An analysis from the Percutaneous Stem

RI

Cell Injection Delivery Effects on Neomyogenesis (POSEIDON) randomized trial.


Circulation research 114:1292-1301.

SC

Tang J, Wang J, Guo L, Kong X, Yang J, Zheng F, et al (2010) Mesenchymal stem cells

NU

modified with stromal cell-derived factor 1 alpha improve cardiac remodeling via
paracrine activation of hepatocyte growth factor in a rat model of myocardial

MA

infarction. Molecules and cells 29:9-19.

Tang YL, Tang Y, Zhang YC, Qian K, Shen L & Phillips MI (2005) Improved graft

TE

mesenchymal stem cell survival in ischemic heart with a hypoxia-regulated heme


oxygenase-1 vector. Journal of the American College of Cardiology 46:1339-

AC
CE
P

1350.

Tang YL, Zhu W, Cheng M, Chen L, Zhang J, Sun T, et al (2009) Hypoxic


preconditioning enhances the benefit of cardiac progenitor cell therapy for
treatment of myocardial infarction by inducing CXCR4 expression. Circulation
research 104:1209-1216.
Thom T, Haase N, Rosamond W, Howard VJ, Rumsfeld J, Manolio T, et al, American
Heart Association Statistics C and Stroke Statistics S (2006) Heart disease and
stroke statistics--2006 update: a report from the American Heart Association
Statistics Committee and Stroke Statistics Subcommittee. Circulation 113:e85151.

37

ACCEPTED MANUSCRIPT
Toma C, Pittenger MF, Cahill KS, Byrne BJ & Kessler PD (2002) Human mesenchymal
stem cells differentiate to a cardiomyocyte phenotype in the adult murine heart.

PT

Circulation 105:93-98.

RI

Traverse JH, Henry TD, Ellis SG, Pepine CJ, Willerson JT, Zhao DX, et al (2011) Effect
of intracoronary delivery of autologous bone marrow mononuclear cells 2 to 3

SC

weeks following acute myocardial infarction on left ventricular function: the

NU

LateTIME randomized trial. JAMA : the journal of the American Medical


Association 306:2110-2119.

MA

Traverse JH, Henry TD, Pepine CJ, Willerson JT, Zhao DX, Ellis SG, et al (2012) Effect
of the use and timing of bone marrow mononuclear cell delivery on left ventricular

TE

function after acute myocardial infarction: the TIME randomized trial. the journal
of the American Medical Association 308:2380-9.

AC
CE
P

Williams AR, Hatzistergos KE, Addicott B, McCall F, Carvalho D, Suncion V, et al


(2013) Enhanced effect of combining human cardiac stem cells and bone marrow
mesenchymal stem cells to reduce infarct size and to restore cardiac function
after myocardial infarction. Circulation 127:213-23.
Williams AR, Suncion VY, McCall F, Guerra D, Mather J, Zambrano JP, et al (2014)
Durable scar size reduction due to allogeneic mesenchymal stem cell therapy
regulates whole-chamber remodeling. J am heart assoc 3:e000140.
Wisel S, Khan M, Kuppusamy ML, Mohan IK, Chacko SM, Rivera BK, et al (2009)
Pharmacological preconditioning of mesenchymal stem cells with trimetazidine
(1-[2,3,4-trimethoxybenzyl]piperazine) protects hypoxic cells against oxidative
stress and enhances recovery of myocardial function in infarcted heart through

38

ACCEPTED MANUSCRIPT
Bcl-2 expression. The Journal of pharmacology and experimental therapeutics
329:543-550.

PT

Wollert KC, Meyer GP, Lotz J, Ringes-Lichtenberg S, Lippolt P, Breidenbach C, et al

RI

(2004) Intracoronary autologous bone-marrow cell transfer after myocardial


infarction: the BOOST randomised controlled clinical trial. Lancet 364:141-148.

stem

cells

to

heart

allografts

during

chronic

rejection.

NU

mesenchymal

SC

Wu GD, Nolta JA, Jin YS, Barr ML, Yu H, Starnes VA et al (2003) Migration of

Transplantation 75:679-685.

MA

Wu KH, Mo XM, Han ZC & Zhou B (2011) Stem cell engraftment and survival in the
ischemic heart. The Annals of thoracic surgery 92:1917-1925.

TE

Yacoub MH & Terrovitis J (2013) CADUCEUS, SCIPIO, ALCADIA: Cell therapy trials
using cardiac-derived cells for patients with post myocardial infarction LV

AC
CE
P

dysfunction, still evolving. Global cardiology science & practice 1:5-8.


Yang YJ, Qian HY, Huang J, Li JJ, Gao RL, Dou KF, et al (2009) Combined therapy
with simvastatin and bone marrow-derived mesenchymal stem cells increases
benefits in infarcted swine hearts. Arteriosclerosis, thrombosis, and vascular
biology 29:2076-2082.

Yang ZJ, Ma DC, Wang W, Xu SL, Zhang YQ, Chen B, et al (2006) Experimental study
of bone marrow-derived mesenchymal stem cells combined with hepatocyte
growth factor transplantation via noninfarct-relative artery in acute myocardial
infarction. Gene therapy 13:1564-1568.

39

ACCEPTED MANUSCRIPT
Yi BA, Wernet O & Chien KR (2010) Pregenerative medicine: developmental paradigms
in the biology of cardiovascular regeneration. The Journal of clinical investigation

PT

120:20-28.

molecular and cellular cardiology 50:327-32.

RI

Yoshida Y & Yamanaka S (2011) iPS cells: a source of cardiac regeneration. Journal of

SC

Zhou B, Honor LB, Ma Q, Oh JH, Lin RZ, Melero-Martin JM, et al (2012) Thymosin beta

NU

4 treatment after myocardial infarction does not reprogram epicardial cells into

AC
CE
P

TE

MA

cardiomyocytes. Journal of molecular and cellular cardiology 52:43-47.

40

ACCEPTED MANUSCRIPT
Table 1: Comparison of the results of selected clinical trials of cell therapy for cardiac
disease sorted by cell populations. BMMNC: Bone marrow derived mononuclear

PT

cells; CDC: Cardiospheres; CSC: Cardiac stem cells; LVEF: Left ventricular

RI

ejection fraction; MSC: Mesenchymal stem cells; NYHA: New York Heart

BMMNC

Cell number and


method of
administration
48-60x109
intracoronary infusion

TE

24.6x108 intracoronary
infusion
intracoronary infusion

AC
CE
P

150x106 intracoronary
infusion
150x106 intracoronary
infusion
100x106
transendocardial
injection

MSC

Results

MA

Cell Type

NU

SC

Classification.

100-200x106
transendocardial
injection
733x106
endoventricular
injection
20-200x106
intramyocardial
injection

Reference

Improved cardiac
contractility and
perfusion
Increased LVEF and
systolic function
Increased LVEF,
Reduced long term
negative outcomes
No significant
improvement at 6
months
No significant
improvement over
placebo
No significant
improvement at 6
months

Chen et al. 2004

Improvements in
multiple parameters
for both autologous
and allogeneic MSC
treatment groups
Improved LVEF, 6
minute walk
distance and NYHA
functional class
Reduced scar size,
Improved LVEF

Hare et al. 2012

41

Wollert et al. 2004


Schachinger et al.
2006
Traverse et al. 2011

Traverse et al. 2012

Perin et al. 2012

Bartunek et al. 2013

Karantalis et al.
2014

ACCEPTED MANUSCRIPT
Reduced scar size,
Improved regional
wall dynamics,
improved 6 minute
walk distance and
Minnesota Living
with Heart Failure
score
Reduced scar size,
Improved segmental
EF

2-fold higher
reduction in scar
size, improved
systolic and diastolic
measured of
function with
combination

Williams et al. 2013

12.5-25x106
intracoronary infusion

Reduced scar size,


no changes in LVEF

Malliaras et al. 2014

MA

TE

1x106 CSC+200x106
MSC, intramyocardial
injection

AC
CE
P

CDC

SC

Bolli et al. 2011

NU

Reduced scar size,


Improved LVEF
Reduced scar size,
greater regional
contractility. No
changes in LVEF.
Reduced scar size,
Improved LVEF

1x106 intracoronary
infusion
12.5-25x106
intracoronary infusion
1x106 intracoronary
infusion

CSC+
MSC

Suncion et al. 2014

RI

20-100x106
transendocardial
injection
CSC

Heldman et al. 2014

PT

transendocardial
injection

42

Makkar et al. 2012

Chugh et al. 2012

Figure 1

AC
CE
P

TE

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

43

AC
CE
P

TE

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

Figure 2

44

Figure 3

AC
CE
P

TE

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

45

Das könnte Ihnen auch gefallen