Anda di halaman 1dari 16

MINERVA CARDIOANGIOL 2005;53:549-64

Cardiac stem cell therapy

A
for myocardial regeneration
A clinical perspective

C
T ® DI
B. DAWN, E. K. ZUBA-SURMA, A. ABDEL-LATIF, S. TIWARI, R. BOLLI

H E
Myocardial infarction and other pathologic Institute of Molecular Cardiology
IG M
conditions of the heart result in loss of car- University of Louisville, Louisville, KY, USA
diomyocytes, scar formation, ventricular
remodeling, and eventually heart failure. Since
pharmacologic and interventional strategies
fail to regenerate dead myocardium, heart fail-
the potential utility of cardiac stem cells for
R A
ure continues to be a major health problem
worldwide. Recent studies in animal models therapeutic myocardial regeneration.
of myocardial infarction and heart failure have Key words: Cardiac stem cell - Myocardial regen-
Y V

demonstrated that various subsets of adult eration - Myocardial infarction - Cardiomyo-


primitive cells can regenerate functional car- pathies.
diomyocytes and cardiac vasculature with
P R

improvement in cardiac structure and func-


tion. Small clinical trials of cell therapy in
patients with myocardial infarction and
ischemic cardiomyopathy have recapitulated M yocardial infarction (MI) leads to loss
of cardiomyocytes and scar formation
O E

these beneficial effects in humans with infarct which over time result in progressive left
size reduction and improvement in ejection ventricular (LV) remodeling and congestive
fraction, myocardial perfusion, and wall
C IN

heart failure.1 Until recently, the heart was


motion. Several phenotypically distinct cell
populations have been utilized for cardiac viewed as a terminally differentiated organ
regeneration, and the relative merits of one that lacked the ability to heal itself 2 and car-
cell over another remain to be determined. The diac regeneration with functional new car-
recent discovery of adult cardiac stem cells has diomyocytes was considered impossible.
M

sparked intense hope for myocardial regener- Thus, therapy of heart failure has focused
ation with cells that are from the heart itself
and are thereby inherently programmed to predominantly on improving patient symp-
reconstitute cardiac tissue. The purpose of this toms with pharmacotherapy, restoring blood
review is to summarize the evidence regard- supply via interventional strategies, and using
ing the feasibility of cardiac repair in humans assist devices and cardiac transplantation in
via adult stem/progenitor cells, and to discuss end-stage disease.3 However, while these
This publication was supported in part by the National therapeutic approaches can only ameliorate
Institutes of Health grants R01 HL-72410 (B.D.), HL-55757, HL- the symptoms of heart failure, they fail to
68088, HL-70897, HL-76794, and HL-78825 (R.B.) and National
AHA Scientist Development Grant 0130146N (B.D.). reconstitute dead myocardium with func-
tional new cardiomyocytes.
Over the past decade, several studies have
Address reprint requests to: B. Dawn, MD, Division of reported the ability of adult stem/progenitor
Cardiology, University of Louisville, Louisville, KY 40292,
USA. E-mail: buddha@louisville.edu cells to differentiate into cardiomyocytes as

Vol. 53, N. 6 MINERVA CARDIOANGIOLOGICA 549


DAWN CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION

well as cells of the vascular lineage both in adult stem/progenitor cell populations for
vitro and in vivo.4-11 Consistent with these cardiac regeneration in humans. These stud-
observations, in animal models of MI and ies may be broadly divided into several cat-
heart failure, it has been repeatedly demon- egories based on the cell types used.
strated that therapy with adult primitive cells
induces cardiac repair, improves LV function, Bone marrow mononuclear cells

A
and halts LV remodeling.5, 6, 12-16 Collectively,
these findings have generated tremendous Since bone marrow mononuclear cells
enthusiasm toward formulating a safe and (BMMNCs) can be isolated relatively quickly

C
effective strategy to achieve therapeutic car- by density gradient centrifugation from total
diac repair by cell therapy. bone marrow cells, this fraction can be pre-
pared with relative ease and speed. In a small

T ® DI
In 2003, Beltrami et al.8 isolated and char-
acterized a lin-/c-kit+ resident cardiac stem study by Hamano et al.,28 intramyocardial
cell (CSC) population from the adult heart injection of BMMNCs during coronary artery
that can differentiate into all of the 3 major bypass graft (CABG) surgery in patients with
functional cell types in the heart, cardiomy- ischemic heart disease improved myocardial

H E
ocytes, smooth muscle cells, and endothe- perfusion. In the study by Strauer et al.29
lial cells, and repair infarcted myocardium autologous BMMNCs harvested from patients
IG M
in vivo with improvement in cardiac func- with AMI were injected following percuta-
tion and anatomy. Since these cells fulfill the neous coronary intervention (PCI). Compared
criteria for adult organ-specific stem cells,17- with patients that received standard therapy,
19 including clonogenicity and multipoten- cell-treated patients showed improvement in
tiality,8, 20 it is hoped that CSCs will offer a regional wall motion and contractility, small-
R A
therapeutic advantage for cardiac repair in er infarct size, and improved myocardial per-
humans. Following a summary of the data fusion. No significant improvement was not-
Y V

from the initial clinical trials with different ed in LVEF. No significant adverse effect due
types of adult stem/progenitor cells, this to cell therapy was observed.29 Several stud-
review will focus on the advances made in ies that used BMMNCs for cardiac repair have
P R

the identification and characterization of CSCs since been reported, and all of these studies
and their potential therapeutic utility for 30-36, 38, 40, 41 except two 37, 39 have shown
improvements in one or more parameters of
O E

myocardial regeneration.
LV contractility, anatomy, and perfusion,
including global LV ejection fraction (LVEF),
C IN

Myocardial regeneration: regional wall motion/thickening, LV end-sys-


bench to bedside tolic volume (LVESV), and myocardial per-
fusion. Improvements in patient symptoms,
Despite continued skepticism,21-25 cell ther- exercise capacity, and New York Heart
M

apy for acute MI (AMI) and ischemic car- Association functional class have been report-
diomyopathy has progressed at an unprece- ed.33, 34 Increased myocardial microvessel
dented translational rapidity. Because of iso- density at the site of BMMNC injection has
lated negative results from animal studies,24, also been noted at autopsy.40 Interestingly,
26 the mechanistic underpinnings of clinical BMMNC therapy has been used successfully
trials were questioned,26 and it was suggest- via the intracoronary route,29, 32, 35 as well as
ed that clinical trials should be deferred until transepicardial 28, 40 and transendocardial 33, 34,
the unresolved mechanistic issues are 36, 38 routes in patients with AMI, 29, 32, 35

addressed.21, 24 Fortunately, fueled by encour- ischemic heart disease,28, 34, 40, 41 and ischemic
aging results from small clinical studies that cardiomyopathy.33, 36, 38 Despite these diverse
showed improvement in LV function and per- patient subsets and modes of delivery, no
fusion, there has been an explosion of cell significant adverse effect of BMMNC therapy
therapy studies in humans.27 Table I 28-53 sum- has been reported thus far. However, since
marizes studies that have employed various bone marrow-derived cells can contribute to

550 MINERVA CARDIOANGIOLOGICA Dicembre 2005


CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION DAWN

tumor neovascularization,54 the potential for regional wall motion and global LVEF and
angiomatous neoplasms should be examined reduction in myocardial perfusion defect.
in longterm studies. Also, the total number of However, 1 patient experienced spontaneous
cells injected in these studies has varied wide- sustained ventricular tachycardia (VT) and
ly (Table I) and the optimal number of cells sustained VT could be induced in another
for different modes of delivery needs to be with poor LV function. Importantly, during

A
determined in dosing studies. Further, despite follow-up, a higher rate of in-stent restenosis
documented improvement in LV function and was observed in the cell-treated group.45
perfusion, the mechanistic underpinnings of

C
BMMNC therapy in humans, i.e., differentia- Circulating progenitor cells
tion of BMMNCs into cardiomyocytes and
In the TOPCARE-AMI trial,30-32 in addition

T ® DI
vascular cells vs cell fusion vs favorable alter-
ations in myocardial matrix and scar due to to autologous BMMNCs, the investigators
paracrine effects, remain largely unknown. also utilized CPCs harvested from patients’
peripheral blood followed by expansion ex
vivo. Although heterogeneous in nature,

H E
Unfractionated bone marrow cells approximately 90% of these circulating prog-
Unfractionated bone marrow cells (BMCs) enitor cells (CPCs) exhibited an endothelial
IG M
have also been shown to improve LV function phenotype.30-32 The overall beneficial effects
and perfusion following transendocardial 42 noted in CPC-treated patients were similar to
as well as intracoronary 43 injection. In the those observed in BMMNC-treated patients.
study by Fuchs et al.,42 electromechanical In a recent randomized double-blind place-
bo-controlled study, Erbs et al.47 have report-
R A
mapping (EMM) guided transendocardial
injection of BMCs improved anginal symp- ed reduction in infarct size, improvement
toms and myocardial perfusion. In a rela- in global LVEF, decrease in the number of
Y V

tively large randomized clinical trial of cell hibernating myocardial segments, and
therapy in humans, Wollert et al.43 reported increase in coronary flow reserve following
significant improvement in regional and glob- intracoronary CPC delivery in patients with
P R

al LVEF and regional wall motion in cell-treat- ischemic heart disease with chronic total
ed patients. No increase in the incidence of occlusion of coronary artery. These data
suggest that the benefits of late reperfusion
O E

arrhythmia or restenosis was noted. Despite


the proven efficacy and safety, the underly- (the open-artery hypothesis) 55 may possibly
ing mechanism of these beneficial effects be enhanced by concomitant cell therapy. In
C IN

with BMC therapy remains to be elucidated. this study,47 the chronicity of the lesion was
greater than 30 days. No increase in resteno-
AC133+ BMCs sis was observed during the three-month
follow-up period.47 However, in a random-
M

AC133+ BMCs exhibit both hematopietic ized trial of cell therapy with granulocyte-
and endothelial differentiation potential and colony stimulating factor (G-CSF)-mobilized
thereby, may represent a subpopulation suit- peripheral blood stem cells, Kang et al.56
able for inducing angiogenesis. In the first reported a very high rate of in-stent resteno-
study, transepicardial injection of AC133+ sis. The trial was stopped prematurely due
BMCs during CABG resulted in improved to safety concerns and despite some bene-
myocardial perfusion and LVEF and smaller ficial effects, data remain essentially incom-
LV end-diastolic volume (LVEDV).44 Two plete without a sizable control group. 56
patients experienced supraventricular tach- Importantly, Schober et al.57 has recently
yarhythmia, a relatively common occurrence reported a significant correlation between
during the postperative period following a greater number of circulating CD34+ cells
CABG. In a subsequent larger study with a following elective stenting and a higher rate
control group,45 intracoronary delivery of of restenosis in patients with coronary artery
AC133+ BMCs resulted in improvement in disease.

Vol. 53, N. 6 MINERVA CARDIOANGIOLOGICA 551


DAWN CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION

TABLE I.—Clinical trials with different types of stem/progenitor cells.


Study Disease Patients treated Controls Route of delivery Number of cells (millions)

BMMNC
Hamano et al.28 IHD 5 None Transepicardial 300-2 200
(during CABG)
Strauer et al.29 AMI 10 10 Intracoronary 28±22

A
TOPCARE-AMI30-32 AMI 29 BMMNC, 11 Intracoronary 213±75 BMMNCs
30 CPC 16±12 CPCs

C
Perin et al.33 ICM 14 7 Transendocardial 25.5±6.3
(EMM-guided)
Tse et al.34 IHD 8 None Transendocardial BMMNC from 40 ml BM

T ® DI
(EMM-guided)
Fernandez-Aviles AMI 20 13 Intracoronary 78±41
et al.35
Perin et al.36 ICM 11 9 Transendocardial 30
(EMM-guided)

H E
Kuethe et al.37 AMI 5 None Intracoronary 39±23

Silva et al.38 ICM 5 None Transendocardial 30


(EMM-guided)
IG M
Kuethe et al.39 ICM 5 None Intracoronary 29±9

Yaoita et al.40 IHD 10 None Transepicardial 3 400±1 200 BMMNCs (5.2±1.6


(during off-pump CD34+ cells)
CABG)
R A
Strauer et al.41 Chronic IHD 18 18 Intracoronary 60-132
(prior MI)
Y V

BMC
Fuchs et al.42 IHD 10 None Transendocardial 78±66
(EMM-guided)
P R

Wollert et al.43 AMI 30 30 Intracoronary 2 500±900

AC133+ BMC
O E

Stamm et al.44 ICM 6 None Transepicardial 1.02-1.57


(during CABG)
C IN

Bartunek et al.45 AMI 19 16 Intracoronary 12.6±2.2

MSC
Chen et al.46 AMI 34 35 Intracoronary 48 000-60 000
M

CPC
TOPCARE-AMI30-32 AMI 29 BMMNC, 11 Intracoronary 213±75 BMMNCs
30 CPC 16±12 CPCs
Erbs et al.47 CTO 12 11 Intracoronary 69±14

Skeletal myoblasts
Menasche et al.48 ICM 10 None Transepicardial 500-1 150
(during CABG)
Smits et al.49 ICM 5 None Transendocardial 196±105
(EMM-guided)

Herreros et al.50 IHD 11 None Transepicardial 100-393


(during CABG)
Siminiak et al.51 ICM 10 None Transepicardial 0.4-50
None (during CABG)

552 MINERVA CARDIOANGIOLOGICA Dicembre 2005


CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION DAWN

ABLE I.—Clinical trials with different types of stem/progenitor cells.


Timing Follow-up duration Results Adverse events

NR 12 months Perfusion ↑ in 3 of 5 patients None

5-9 days after 3 months Infarct size ↓; regional wall motion ↑; perfusion ↑; LVE- None

A
AMI SV ↓
4.9±1.5 days 12 months Infarct size ↓; global LVEF ↑; regional wall motion ↑; None
after AMI perfusion defect ↓; LVEDV ↓; LVESV ↓

C
NR 4 months Angina ↓; NYHA class ↓; perfusion ↑; regional wall None
motion ↑; global LVEF ↑; LVESV ↓
NR 3 months Angina ↓; perfusion ↑; regional wall motion ↑; regional None
wall thickening ↑

T ® DI
13.5±5.5 days 6 months Regional wall motion ↑; global LVEF↑; regional EF↑; None
after AMI LVESV↓
NR 12 months Exercise capacity ↑; perfusion ↑; no improvement in None
LVEF

H E
6.3±0.4 days 12 months No improvement in regional wall None
after PCI Motion, contractility, and global LVEF
NR 6 months Exercise capacity ↑; no change in global LVEF and per- None
fusion
IG M
1.3±0.5 years 12 months No improvement in regional wall None
after AMI motion or global LVEF
NR 5-32 months Perfusion ↑; myocardial microvessel density ↑ at autopsy 1 sudden death after 27
months
R A
27±31 3 months Infarct size ↓; regional wall motion ↑; No significant increase in reste-
months after global LVEF ↑; VO2max ↑; FDG-PET uptake ↑ nosis
AMI
Y V

NR 3 months Angina ↓; perfusion ↑ None


P R

4.8±1.3 days 6 months Regional wall motion ↑; global and regional LVEF ↑ None
after PCI
O E

>10 days but 3-10 months Global LVEF ↑; perfusion ↑; LVEDV ↓ 2 patients had SVT
<3 months
after AMI
C IN

11.6±1.4 days 4 months Regional wall motion ↑; global LVEF ↑; perfusion ↑ 1 patient had sustained VT;
after PCI higher in-stent restenosis

18 days after 6 months Infarct size ↓; global LVEF ↑; regional wall motion ↑; None
M

PCI perfusion ↑; LVEDV ↓

4.9±1.5 days 12 months Infarct size ↓; global LVEF ↑; regional wall motion ↑; None
after AMI perfusion defect ↓; LVEDV ↓; LVESV ↓
10±1 days 3 months Infarct size ↓; global LVEF ↑; coronary flow reserve ↑; None
after PCI hibernating myocardial segments ↓

3-228 months 10.9 months NYHA class ↓; global LVEF ↑; regional wall thickening ↑ 4 patients had sustained VT
after AMI
24-132 6 months Global LVEF ↑; regional wall thickening ↑ 1 patient had nonsustained VT
months after
AMI
3-168 months 6.5 months Global LVEF ↑; regional wall None
after AMI motion ↑; viability in infarct area ↑
4-108 months 12 months Global LVEF ↑; regional wall 4 patients had sustained VT
after AMI motion ↑ None
(Continued)

Vol. 53, N. 6 MINERVA CARDIOANGIOLOGICA 553


DAWN CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION

TABLE I.—Clinical trials with different types of stem/progenitor cells.


Study Disease Patients treated Controls Route of delivery Number of cells (millions)

Chachques et al.52 ICM 20 None Transepicardial 300±20


(during CABG) maximum
Siminiak et al.53 ICM 10 None Intramyocardial 17-106
(via cardiac veins)

A
AMI: acute myocardial infarction; BMC: bone marrow cells; BMMNC: bone marrow mononuclear cells; CABG: coronary artery bypass
FDG-PET: 18fluorodeoxyglucose-positron emission tomography; ICM: ischemic cardiomyopathy; IHD: ischemic heart disease; LV: left
tion; NR: data not reported; NYHA: New York Heart Association; PCI: percutaneous coronary intervention; SVT: supraventricular

C
T ® DI
Mesenchymal stem cells cation may render skeletal myoblasts safer
for clinical use in the future.
In a relatively large randomized trial, Chen
et al.46 injected bone marrow-derived mes-
enchymal stem cells (MSCs) in patients with

H E
AMI. MSCs were expanded in culture for 10 Cardiac stem cells: isolation and
days and injected via the coronary artery 18 characterization
days (average) after PCI. Treated patients
IG M
exhibited improved regional wall motion and Adult stem cells have been identified in
global LVEF, smaller perfusion defect, and many organs, including bone marrow, skele-
smaller LVEDV and LVESV.46 No significant tal muscle, skin, central nervous system, liv-
adverse effect was noted. er, and gastrointestinal tract.17, 18 Under phys-
R A
iologic and pathologic conditions, these res-
Skeletal myoblasts ident stem cells can differentiate into appro-
priate organ-specific lineages and reconsti-
Y V

Following the initial reports of myocardial tute dead or damaged tissue.17, 18 Although the
repair with skeletal myoblasts,58 several clin- heart has traditionally been viewed as a post-
ical studies 48-53 have tested the effect of these
P R

mitotic organ incapable of intrinsic repair,2


cells in humans. In the first clinical study,48 elegant work from Anversa’s laboratory has
transplantation of autologous skeletal documented that death and regeneration of
O E

myoblasts during CABG improved patient cardiac myocytes indeed play significant roles
symptoms, increased systolic wall thicken- in cardiac homeostasis.62-68 According to this
ing and LVEF. However, 4/10 patients expe- new paradigm, a small population of cycling
C IN

rienced delayed sustained VT needing myocytes are continuously regenerated via


implantable cardioverter defibrillator (ICD) the differentiation of CSCs.66-68
implantation.48 Increased incidence of VT fol-
lowing myoblast transplantation has been Lin-/c-kit+ CSCs
M

reported in subsequent studies.49, 51, 53 This


arrhythmogenic potential of myoblasts is not Anversa et al. recently isolated and charac-
surprising as studies in animal models indi- terized a resident CSC population in the adult
cate that these cells do not differentiate into heart that expresses markers for primitive cells,
cardiomyocytes 58 and do not connect with including c-kit, Sca-1, and MDR1, but do not
neighboring cells via gap junctions.59, 60 express any of the markers of hematopoietic
Intriguingly, despite these limitations, lineages (lin-).8, 20, 69 In vitro, CSCs are clono-
improvement in global LVEF and/or patient genic and are capable of self-renewal through
symptom was noted in all of these studies.48- multiple passages.8, 69 By virtue of their multi-
53 In coculture studies of skeletal myoblasts potential nature, CSCs are also able to differ-
and cardiomyocytes, lentiviral transduction entiate in vitro into all of the 3 functionally
of connexin 43 in skeletal myoblasts has important types of cells in the heart, car-
recently been shown to decrease arrhyth- diomyocytes, smooth muscle cells, and
mogenicity in vitro.61 Similar genetic modifi- endothelial cells identified by the expression

554 MINERVA CARDIOANGIOLOGICA Dicembre 2005


CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION DAWN

ABLE I.—Clinical trials with different types of stem/progenitor cells.


Timing Follow-up duration Results Adverse events

NR 14±5 NYHA class ?; global LVEF ?; regional wall motion ? None


months
5-96 months 6 months NYHA class ? 1 patient had sustained VT
after AMI

A
graft surgery; CPC: circulating progenitor cells; CTO: chronic total occlusion; EF: ejection fraction; EMM: electromechanical mapping;
ventricular; LVEDV: LV end-diastolic volume; LVESV: LV end-systolic volume; MSC: mesenchymal stem cells; LVEF: LV ejection frac-
tachyarrhythmia; VT: ventricular tachycardia

C
T ® DI
of cell-specific markers.8 In these studies,8, 20, ent system, these cells were able to generate
69 CSC therapy after AMI induced myocardial cardiomyocytes both with and without cell
repair with formation of new cardiac myocytes, fusion.73 Identification of similar Sca-1+ car-
smooth muscle cells, and endothelial cells.8 diac progenitors have been reported by other
The newly formed BrdU+ myocytes expressed investigators.74 Upon oxytocin stimulation,

H E
cardiac-specific proteins, including α-sar- adherent cardiac Sca-1+ cells differentiated into
comeric actin, cardiac myosin heavy chain a cardiomyocytic phenotype and exhibited
IG M
(MHC), α-cardiac actinin, and proteins asso- spontaneous contractions.74 Interestingly, 90%
ciated with intercellular gap junctions, N-cad- of these Sca-1+ cells did not express c-kit.74
herin and connexin 43.8 It was also demon- However, the clonogenicity and in vivo regen-
strated that CSCs express CXCR4 and can effec- erative capacity of these Sca-1+ progenitors 74
tively cross the vascular barrier following intra- remain to be demonstrated.
R A
aortic injection, inducing cardiac repair.20
Further characterization revealed that the CSCs CARDIAC SIDE POPULATION CELLS
Y V

express insulin-like growth factor (IGF)-1 Side population (SP) cells have been
receptor and c-Met (receptor for stem cell fac- described in several organs, including bone
tor) and are able to migrate in vivo along a
P R

marrow, skeletal muscle, kidney, lung, and


growth factor gradient.70 The number of CSCs small intestine.80, 81 Although SP cells are typ-
increases with the formation of numerous new ically identified by their ability to extrude
O E

myocytes in humans with aortic stenosis, a Hoechst 33342 dye, the majority of SP cells
valvular condition associated with cardiac from the bone marrow and muscle are Sca-
hypertrophy.71 Activation of CSCs in humans 1+.82, 83 Martin et al.75 reported the identifi-
C IN

with ischemic cardiomyopathy with resultant cation of an SP cell in the heart of embryon-
cardiac repair has also been documented.72 ic as well as adult mice that expresses the
ATP-binding cassette transporter Abcg2, an
Other cardiac progenitors integral feature of the SP cell phenotype.84
M

Several other investigators have since Interestingly, these cardiac Abcg2-express-


reported the identification of distinctly dif- ing SP cells are Sca-1high, c-kitlow, CD34low,
ferent populations of primitive cells in the CD45low, and distinct from endothelial cells.75
heart that are able to differentiate into car- Subsequently, Pfister et al.77 reported a cardiac
diomyocytes and/or regenerate infarcted SP cell that expresses Sca-1 and is negative for
myocardium.73-79 CD31. Although these SP cells 77 differentiate
into cardiomyocytes in vitro, their ability to
SCA1+ CARDIAC PROGENITORS survive in vivo and induce myocardial repair
remains to be proven. Subsequent work from
Oh et al.73 described a Sca-1+ cardiac prog- these authors 85 has demonstrated that the
enitor population that also expresses CD31, number of cardiac SP cells decreases acute-
but not c-kit or other markers of hematopoietic ly following AMI and is replenished by cir-
lineage and endothelial cells. When injected i.v. culating bone marrow-derived primitive cells
into infarcted mice in a Cre-lox donor/recipi- that lose the CD45 antigen following myocar-

Vol. 53, N. 6 MINERVA CARDIOANGIOLOGICA 555


DAWN CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION

dial homing. These interesting results 85 sug- with minimal adverse effects, and efficacious
gest that primitive bone marrow cells may with significant health benefits. Although no
partake in the turnover of cardiac progenitors data are available regarding the safety and
during pathologic conditions. benefits of potential CSC therapy in humans,
based on results from animals studies, ther-
CARDIOSPHERE-FORMING PROGENITORS apy with CSCs may potentially offer several

A
distinct advantages over other cell types.
Messina et al.76 described the isolation of
primitive cells from human and murine hearts

C
Regeneration of both muscle and vessel
that formed cardiosphere (CS) in culture. These
Sca-1+, c-kit+, KDR/flk-1+, and CD31+ CSs An organ-specific stem cell should be able
beat spontaneously in culture and exhibit self- to replenish not only one type of cell, but

T ® DI
renewal capacity.76 In addition, they express several important components of the organ.17,
18, 86 Further, it is important to recognize that
cardiac differentiation markers, including tro-
ponin I, MHC, atrial natriuretic peptide, and newly formed myocytes in the middle of an
endothelial marker von Willebrand factor. infarct scar will not survive in the absence

H E
These CSs survive in vivo following trans- of adjoining vascular structures. On the oth-
plantation into the heart and express markers er hand, regeneration of blood vessels alone
will not enhance contractility unless accom-
IG M
of cardiac lineages.76 A detailed characterization
of proliferating CSs grown in vitro from human panied by formation of new myocytes. Thus
endocardial biopsies revealed that they are far, different results have been reported by
positive for c-kit, MDR1, connexin 43, and the various laboratories with reports confirming
cardiac transcription factor Nkx2.5.79 the regeneration of either muscle or vessels,
R A
but infrequently of both. In this regard, c-
kit+ CSCs offer a greater therapeutic poten-
ISL-1+ CARDIAC PROGENITORS
Y V

tial as they express markers specific for car-


Using a tamoxifen-inducible Cre-lox sys- diomyocytes, smooth muscle cells, and
tem, Laugwitz et al.78 described a primitive endothelial cells in vitro,8 and are able to
P R

cell population in the heart that expresses generate not only new myocytes but also
the homeobox gene isl-1 but does not new blood vessels securing blood supply to
extrude Hoechst 33342 and does not express the newly formed cells in vivo.8, 20, 69
O E

c-kit. These cells acquire cardiomyocytic phe-


notype without fusion when cocultured with Connection with other myocytes
C IN

neonatal cardiomyocytes in vitro and show Another important hurdle for cell trans-
electrical as well as contractile properties plantation has emerged over the past few
reminiscent of neonatal cardiomyocytes.78 years during clinical trials with skeletal
However, these isl-1+ cells have only been myoblasts - the increased incidence of arrhyth-
isolated from very young animals and
M

mias.48, 49, 51, 53 Importantly, skeletal myoblasts


humans,78 and their presence and signifi- do not differentiate into cardiomyocytes 58
cance in cardiac homeostasis in adult organ- and do not connect with the surrounding
isms remain to be ascertained. Further, myocardium.59, 60 These islands of electrical-
whether these isl-1+ progenitors are multi- ly active yet dissimilar cells may potentially
potential and can indeed induce cardiac serve as arrhythmogenic foci. In contrast, fol-
repair in vivo remains to be proven. lowing CSC transplantation in vivo, the regen-
erated myocytes express on their surface not
only N-cadherin, which is necessary for
Cardiac stem cells: therapeutic anatomic connection with neighboring
potential myocytes, but also connexin 43, which con-
nects them to other myocytes electrically.8, 20
As with any other form of therapy, cell These favorable features predict less arrhyth-
therapy in humans should ideally be safe mic events with CSC transplantation.

556 MINERVA CARDIOANGIOLOGICA Dicembre 2005


CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION DAWN

Clonogenicity be tested in humans. CSCs may potentially be


harvested during an open-heart procedure
Since the beneficial effects of cell therapy
(e.g., CABG, LV assist device implantation,
are dose dependent,87 the number of trans-
valve repair/replacement) or catheter-based
planted cells is important for cardiac repair in
myocardial biopsy. Because of their clono-
humans. Although the use of unselected bone
genicity,8, 69 CSCs can then be grown in vit-

A
marrow cells and BMMNCs has resulted in
improvement in LV function and myocardial ro to generate sufficient number of multipo-
perfusion in clinical trials, perhaps the ben- tent cells. Because of the reported success
with cardiac repair following intramyocar-

C
efits would be much greater if the appropri-
ate cells, isolated in small numbers, could be dial injection of CSCs,8 these cells can be
expanded in vitro prior to cell therapy and injected directly into the myocardium dur-

T ® DI
administered in sufficient numbers. Although ing CABG or via the intraventricular approach
self-renewal is a cardinal feature of a stem guided by an EMM system. These stategies
cell,17, 19 many of the primitive cell popula- may be utilized for cardiac repair in patients
tions are difficult to expand in vitro and with chronic ischemic as well as nonischemic

H E
undergo a phenotypic shift during this cardiomyopathy. However, following AMI,
process. In contrast, CSCs 8, 20, 69 are clono- a transcoronary route of delivery during PCI
genic and retain their phenotype during may represent the most convenient mode of
IG M
expansion in vitro. These CSCs may be har- delivery. Although AMI and ischemic car-
vested from patients with ischemic heart dis- diomyopathy would constitute the most com-
ease and expanded in vitro to obtain a large mon clinical conditions for therapeutic use of
number of cells for future therapeutic use. CSCs, potentially, CSCs may also be used to
R A
reverse the progressive deterioration of ven-
tricular function in nonischemic cardiomy-
Efficacy with transcoronary delivery opathies. Importantly, in all of these scenar-
Y V

In the setting of an AMI, the most conve- ios, stringent criteria must be formulated to
nient mode of delivery is via the coronary assess the benefits of CSC therapy by rigor-
P R

artery during primary PCI or as an elective ous measurements of LV function by echocar-


procedure a few days later. However, deliv- diography, left ventriculography, and cardiac
ery of MSCs via the intracoronary route has magnetic resonance imaging studies if pos-
O E

been reported to cause microinfarctions in sible. Myocardial perfusion should be quan-


dogs, possibly because of the relatively large tified by radionuclide perfusion imaging.
cell size.88 This problem may potentially be Finally, any impact on survival should be
C IN

encountered during intracoronary delivery assessed during longterm follow-up.


of any large cell. In our experience, delivery
of culture expanded CSCs via the intracoro- Safety issues
nary route in rats and pigs was not associat-
M

ed with microinfarctions (unpublished obser- Major adverse effects of cell therapy iden-
vation). Importantly, following the intra- tified in recent studies in humans include
coronary delivery, CSCs were able to cross the potentially fatal ventricular arrhythmias and
vessel wall relatively quickly to home into higher incidence of restenosis (Table I). In
the injured myocardium and regenerate new light of this knowledge, the safety of CSC
cardiomyocytes and vasculature.20 It was also therapy in humans needs to be closely mon-
shown that CSCs express CXCR 4,20 the recep- itored. However, sustained VT has been not-
tor for stromal cell-derived factor-1, that plays ed primarily following transplantation of
a critical role in stem cell homing.89 skeletal myoblasts,48, 49, 51, 53 which are unable
to connect electrically to the neighboring
myocytes.59, 60 In contrast, in animal models,
Potential therapeutic uses
CSC-derived cardiomyocytes have been
Based on the evidence discussed above, it shown to express N-cadherin and connexin
is apparent that CSCs are eminently poised to 43,8, 20 which are necessary for establishing

Vol. 53, N. 6 MINERVA CARDIOANGIOLOGICA 557


DAWN CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION

mechanical and electrical connection, respec- tion. Third, the ideal cell should be able to
tively. A higher rate of in-stent restenosis has escape from the vasculature and home into
been reported with intracoronary injection the infarcted or noninfarcted myocardium.
of AC133+ progenitors 45 and G-CSF-mobi- Expression of CXCR4 and other adhesion
lized peripheral blood cells.56 Restenosis is molecules will identify cells with this capa-
multifactorial in etiology and involves smooth bility. This will greatly improve efficacy of

A
muscle cell activation and neointimal thick- cell delivery via the intracoronary route, and
ening.90 Several growth factors also play crit- improve retention of cells following intramy-
ical roles in restenosis,90 and although coro- ocardial delivery. Fourth, this cell should be

C
nary stenting eliminates elastic recoil and easy to harvest in a timely fashion, and/or
remodeling, it accentuates neointimal hyper- be suitable for longterm storage with or with-

T ® DI
plasia. Since CSCs have been shown to out expansion following harvest at an oppor-
express growth factor systems 70 and are able tune time beforehand. Fifth, the ideal cell
to differentiate into smooth muscle cells as should be able to self-renew in vitro without
well as endothelial cells,8, 20 the rate of a change in phenotype or loss of the multi-

H E
restenosis following intracoronary injection of potential nature. Finally, the adverse effects
CSCs following PCI will need to be moni- of therapy should be minimal with this cell.
tored carefully. Cell transplantation should not cause the
IG M development of teratomas or other neoplas-
tic growth, and intracoronary administration
Cell therapy: optimizing the variables should not cause hyperplasia involving the
arterial wall causing restenosis or a de novo
R A
Despite the initial encouraging results from atheromatous lesion. Although numerous cell
clinical trials, many questions remain unan- types have been utilized in animal models,
swered regarding the optimal cell therapy each with somewhat distinct phenotype and
Y V

strategy for myocardial repair. As summa- properties, no cell has been definitively
rized in Table I, diverse types of cells, num- shown to fulfill all of these criteria. Although
P R

bers, and routes have thus far been employed CSCs fulfill several of these criteria, the safe-
in patients with AMI, ischemic cardiomy- ty profile has not been tested in the clinical
opathy, and chronic ischemic heart disease. setting.
O E

However, several major issues regarding the


cell type, route, and timing of therapy remain The most efficacious route
to be addressed.
C IN

Based on patient demographics and pathol-


ogy, several different approaches can be uti-
The ideal cell lized for cell delivery. For chronic ischemic
Based on current evidence, the most cardiomyopathy, transepicardial injection dur-
M

appropriate cell for cardiac regeneration ing CABG offers a very effective route, which
should have several important properties. has been employed in several clinical studies.
First, it should be able to differentiate into Transendocardial delivery has also been suc-
both cardiomyocytes and cells of the vascu- cessfully employed in a number of studies.
lar lineage, so that not only new contractile With this method, the use of an EMM system
units are formed, but also blood supply to enables the identification of the scar area and
these newly formed myocytes is secured. enables precise cell injection. The use of a
Second, the newly formed myocytes should transendocardial route may potentially be
couple mechanically as well as electrically limited by the availability of this system.
with native myocytes (contracting in syn- Alternatively, in patients with significant coro-
chrony with adjacent cells). Mechanical cou- nary artery disease, cells may be injected in
pling will preclude ineffectual contraction, the coronary artery following PCI or as an
and electrical coupling may prevent the elective procedure. Interestingly, intracoro-
occurrence of arrhythmias and improve func- nary delivery of CPCs in patients with revas-

558 MINERVA CARDIOANGIOLOGICA Dicembre 2005


CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION DAWN

cularized chronic (>30 days) total occlusion Perspectives and future directions
47 and BMMNCs in patients with old myocar-
dial infarction 41 has been associated with AMI and ischemic cardiomyopathy con-
improvement in LV function. These results tinue to be major health problems with
suggest that intracoronary route of cell deliv- increasing incidence, and the benefits of phar-
ery may be utilized even when myocardial macologic therapy appears to be limited to

A
inflammation is subacute or minimal. symptomatic improvement with only mod-
In the setting of an AMI, cell delivery imme- est effects on disease progression. Because of
diately following PCI is likely to be most cost- the compelling need to formulate a better

C
effective. However, the ready availability of strategy for treating millions of patients suf-
cells is a significant issue in this regard. Another fering from these debilitating conditions

T ® DI
important issue for the intracoronary delivery worldwide, and because of the unprece-
relates to the ability of the cell to cross the dented promise of stem cell therapy, clini-
vessel barrier. Primitive cells that express cal application of cell therapy has progressed
CXCR4 (for example, CSCs20) and other adhe- at a rapid pace. Thus far, data from these rel-
atively small trials have generally been pos-

H E
sion molecules may be better suited for this
mode of delivery. Hofmann et al.91 reported itive in terms of improvements in LV function,
greater myocardial retention of CD34+ BMCs myocardial perfusion, and even decrease in
IG M
compared with unselected BMCs following infarct size.
intracoronary delivery 5-10 days after PCI fol- It should be noted that many of these ear-
lowing AMI. However, large cells may not be ly clinical trials did not have a control group
suitable for intracoronary administration and (Table I). In the few that did have a control
R A
intracoronary delivery of MSCs in dogs has group, control patients received standard care
been reported to cause microinfarction.88 - no control cell was injected. Further, only a
Further, injection of circulating AC133+ prog- few studies were randomized.30, 32, 43, 46, 47
Y V

enitors 45 and G-CSF-mobilized progenitors 56 Even in the animal models, very few studies
has been associated with a higher rate of compared the benefits of more than one cell
type. In humans, the TOPCARE-AMI trial 30-
P R

restenosis. Thus, the method of delivery needs


32 compared 2 cell populations; the benefits
to be tailored according to the cell injected.
of cell therapy were similar in patients treat-
O E

The optimal time ed with BMMNCs and CPCs.30-32 To resolve


the issue of the ideal cell type, future basic
For nonacute conditions, including and clinical studies should be designed to
C IN

ischemic heart disease and cardiomyopathy, compare the beneficial effects of different
the time of delivery may be synchronized types of cells, especially different subpopu-
with a planned procedure. However, since lations from the bone marrow and CSCs.
the inflammatory reaction persists for a pro- Although intramyocardial injection may be
M

longed period of time after AMI,92 timing of employed during surgical procedures or
cell delivery may prove critical in the setting when an EMM system is available, perhaps
of an AMI. On one hand, the increased intravenous or intracoronary delivery would
expression of adhesion molecules and be more suitable for general use. However,
chemoattractants in the infarcted myocardium myocardial homing/retention following injec-
93 as well as the viable myocardium may tion into the blood stream has been subop-
improve cell retention.94 On the other hand, timal. Four days after LV intracavitary injec-
delivery of cells into an inflammatory milieu tion, only 0.44% of the injected MSCs were
may cause excessive cell death. Thus far, cell detected in the myocardium.7 This problem
delivery after AMI in humans has been ben- is even greater with intravenous injection,
eficial at various time points (Table I). Future with only a small fraction of injected cells
studies will be necessary to identify the most homing to the myocardium.91, 94, 95 Even with
appropriate time for cell delivery in general intracoronary injection, less than 3% of uns-
and CSC delivery following AMI. elected BMCs and 14-39% of CD34+ BMCs

Vol. 53, N. 6 MINERVA CARDIOANGIOLOGICA 559


DAWN CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION

were retained in the myocardium.91 To objec- sion. If one considers new vessel formation
tively address cell kinetics, cells labeled with as a surrogate marker for improved perfu-
enhanced green fluorescent protein, CM-DiI, sion, in preclinical studies CSCs have been
and radionuclides have been used. However, shown to achieve all of these end-points.8, 20
each of these methods has specific limita- Thus, the time seems ripe for testing the car-
tions and newer labeling techniques with diac regenerative efficacy of CSCs in patients

A
greater precision need to be devised to accu- with AMI, ischemic cardiomyopathy, and oth-
rately assess cell retention and viability fol- er cardiac pathologies that result in heart fail-
lowing transplantation. ure.

C
Low survival rate of grafted cells is a poten-
tial problem with cell transplantation in gen-

T ® DI
eral.96 The inflammatory milieu in the infarct- Conclusions
ed myocardium 92 may cause death of trans-
planted cells and progressive apoptosis may Therapy with stem/progenitor cells holds
also contribute to cell loss.97 However, the great promise for millions of patients with

H E
true extent of this problem with bone mar- AMI, ischemic cardiomyopathy, and heart
row-derived cells and CSCs needs further failure. Although many details regarding the
assessment. In an elegant study by Mangi et ideal cell type, route, and timing remain to be
IG M
al.,98 transplanted MSCs overexpressing the optimized, the collective data from initial clin-
antiapoptotic gene Akt restored four-fold ical trials indicate that cell therapy can
greater myocardial volume compared with improve LV function and perfusion without
equal number of cells transduced with a significant adverse effects. In this regard, the
R A
reporter gene. Similar genetic modification discovery of CSCs represents a major
of CSCs may potentially offer significant addi- advance. Based on the evidence generated
tional benefits. from extensive basic studies, CSCs appear to
Y V

Augmentation of the beneficial effects of be ideally suited for myocardial regeneration


cell therapy may also be achieved by express- as they exhibit many of the necessary pre-
ing angiogenic factors in the transplanted
P R

requisites for successful cellular transplanta-


cell. Tranfection of skeletal myoblasts with tion for cardiac repair. Although CSCs have
vascular endothelial growth factor (VEGF) not been tested for cardiac regeneration in
O E

enhanced myocardial angiogenesis and humans, they may potentially be utilized in


improved cardiac function.99 In another ischemic and nonischemic cardiomyopathies,
study,100 transfection of endothelial progen- myocardial infarction, and other pathologic
C IN

itor cells (EPCs) with VEGF reduced the num- states that culminate in loss of myocytes.
ber of cells necessary to improve ischemic However, the safety and efficacy of CSC ther-
limb salvage. CSCs are able to differentiate apy in humans should be monitored with
into endothelial cells and vascular smooth stringent measures and compared with those
M

muscle cells, thereby regenerating the vas- achieved with other forms of adult primitive
culature necessary to sustain cell survival.8, 20 cells.
However, since VEGF has been shown to
enhance the efficacy of EPCs,100 transfection
of CSCs with VEGF or other angiogenic fac- Riassunto
tors may merit further investigation.
It is obvious that cell therapy for myocar- Terapia con cellule staminali per la rigenerazione
dial regeneration is in its infancy and the miocardica: una prospettiva clinica
mechanism underlying the observed bene- Terapia con cellule staminali per la rigenerazione
fits remains to be elucidated. From a clinical miocardica: una prospettiva clinica L’infarto miocar-
dico ed altre patologie cardiache determinano perdita
standpoint, however, what is more impor- netta di cardiomiociti, sviluppo di tessuto cicatrizia-
tant is to achieve infarct size reduction, atten- le, rimodellamento ventricolare ed infine scompenso
uation of LV remodeling, improvement in LV cardiaco. Poiche’ le strategie farmacologiche ed inter-
function, and increase in myocardial perfu- ventistiche non sono in grado di rigenerare il mio-

560 MINERVA CARDIOANGIOLOGICA Dicembre 2005


CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION DAWN

cardio perduto, lo scompenso cardiaco rimane una tionally active cardiomyocytes. Circulation 2003;107:
delle piu’ importanti patologie a livello mondiale. 1024-32.
Recenti studi su modelli animali di infarto miocardi- 10. Kawada H, Fujita J, Kinjo K, Matsuzaki Y, Tsuma M,
Miyatake H et al. Nonhematopoietic mesenchymal
co e di scompenso cardiaco hanno dimostrato come stem cells can be mobilized and differentiate into car-
vari sottotipi di cellule dell’adulto possano rigenera- diomyocytes after myocardial infarction. Blood
re cardiomiociti funzionali e tessuto vascolare car- 2004;104:3581-7.
diaco, con miglioramento della struttura e della fun- 11. Hattan N, Kawaguchi H, Ando K, Kuwabara E, Fujita

A
zione cardiaca. Piccoli trial clinici di terapia cellulare J, Murata M et al. Purified cardiomyocytes from bone
marrow mesenchymal stem cells produce stable intrac-
in pazienti con infarto miocardico e cardiomiopatia ardiac grafts in mice. Cardiovasc Res 2005;65:334-44.
ischemica hanno confermato questi effetti benefici 12. Kocher AA, Schuster MD, Szabolcs MJ, Takuma S,

C
anche nell’uomo, con riduzione delle dimensioni Burkhoff D, Wang J et al. Neovascularization of
infartuali e miglioramento di frazione di eiezione, ischemic myocardium by human bone-marrow-derived
perfusione miocardica e cinetica di parete. Diverse angioblasts prevents cardiomyocyte apoptosis, reduces
remodeling and improves cardiac function. Nat Med

T ® DI
popolazioni cellulari fenotipicamente distinte sono 2001;7:430-6.
state impiegate per la rigenerazione miocardica e i 13. Kawamoto A, Gwon HC, Iwaguro H, Yamaguchi JI,
meriti relativi di ciascun tipo cellulare restano anco- Uchida S, Masuda H et al. Therapeutic potential of ex
ra da determinare. La recente scoperta di cellule sta- vivo expanded endothelial progenitor cells for myocar-
minali cardiache dell’adulto ha alimentato un’intensa dial ischemia. Circulation 2001;103:634-7.

H E
speranza per la rigenerazione cardiaca con cellule 14. Shake JG, Gruber PJ, Baumgartner WA, Senechal G,
Meyers J, Redmond JM et al. Mesenchymal stem cell
del cuore stesso e che sono pertanto intrinsecamen- implantation in a swine myocardial infarct model:
te programmate per la ricostituzione del tessuto car- engraftment and functional effects. Ann Thorac Surg
IG M
diaco. Lo scopo di questa revisione e’ di riassumere
le evidenze sulla fattibilita’ della riparazione tissuta- 15.
2002;73:1919-25; discussion 1926.
Kawamoto A, Tkebuchava T, Yamaguchi J, Nishimura
le cardiaca nell’uomo mediante l’uso di cellule adul- H, Yoon YS, Milliken C et al. Intramyocardial trans-
te staminali o progenitrici e di discutere la potenzia- plantation of autologous endothelial progenitor cells for
therapeutic neovascularization of myocardial ischemia.
le utilita’ delle cellule staminali cardiache per la rige- Circulation 2003;107:461-8.
R A
nerazione miocardica. 16. Yoon YS, Wecker A, Heyd L, Park JS, Tkebuchava T,
Parole chiave: Cellule staminali cardiache - Kusano K et al. Clonally expanded novel multipotent
stem cells from human bone marrow regenerate
Rigenerazione miocardica - Infarto miocardico - myocardium after myocardial infarction. J Clin Invest
Y V

Cardiomiopatie. 2005;115:326-38.
17. Fuchs E, Segre JA. Stem cells: a new lease on life. Cell
2000;100:143-55.
P R

18. Poulsom R, Alison MR, Forbes SJ, Wright NA. Adult


References stem cell plasticity. J Pathol 2002;197:441-56.
19. Verfaillie CM, Pera MF, Lansdorp PM. Stem cells: hype
1. Pfeffer MA, Braunwald E. Ventricular remodeling after and reality. Hematology (Am Soc Hematol Educ
O E

myocardial infarction. Experimental observations and Program) 2002;1:369-91.


clinical implications. Circulation 1990;81:1161-72. 20. Dawn B, Stein AB, Urbanek K, Rota M, Whang B,
2. MacLellan WR, Schneider MD. Genetic dissection of Rastaldo R et al. Cardiac stem cells delivered intravas-
cardiac growth control pathways. Annu Rev Physiol cularly traverse the vessel barrier, regenerate infarcted
C IN

2000;62:289-319. myocardium, and improve cardiac function. Proc Natl


3. McMurray JJ, Pfeffer MA. Heart failure. Lancet Acad Sci U S A 2005;102:3766-71.
2005;365:1877-89. 21. Chien KR. Stem cells: lost in translation. Nature
4. Makino S, Fukuda K, Miyoshi S, Konishi F, Kodama 2004;428:607-8.
H, Pan J et al. Cardiomyocytes can be generated from 22. Matsubara H. Risk to the coronary arteries of intra-
marrow stromal cells in vitro. J Clin Invest 1999;103:697- coronary stem cell infusion and G-CSF cytokine ther-
M

705. apy. Lancet 2004;363:746-7.


5. Tomita S, Li RK, Weisel RD, Mickle DA, Kim EJ, Sakai 23. Wagers AJ, Sherwood RI, Christensen JL, Weissman IL.
T et al. Autologous transplantation of bone marrow Little evidence for developmental plasticity of adult
cells improves damaged heart function. Circulation hematopoietic stem cells. Science 2002;297:2256-9.
1999;100(19 Suppl):II247-56. 24. Balsam LB, Wagers AJ, Christensen JL, Kofidis T,
6. Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson Weissman IL, Robbins RC. Haematopoietic stem cells
SM, Li B et al. Bone marrow cells regenerate infarcted adopt mature haematopoietic fates in ischaemic
myocardium. Nature 2001;410:701-5. myocardium. Nature 2004;428:668-73.
7. Toma C, Pittenger MF, Cahill KS, Byrne BJ, Kessler PD. 25. Bauersachs J, Thum T, Frantz S, Ertl G. Cardiac regen-
Human mesenchymal stem cells differentiate to a car- eration by progenitor cells--bedside before bench? Eur
diomyocyte phenotype in the adult murine heart. J Clin Invest 2005;35:417-20.
Circulation 2002;105:93-8. 26. Murry CE, Soonpaa MH, Reinecke H, Nakajima H,
8. Beltrami AP, Barlucchi L, Torella D, Baker M, Limana Nakajima HO, Rubart M et al. Haematopoietic stem
F, Chimenti S et al. Adult cardiac stem cells are multi- cells do not transdifferentiate into cardiac myocytes in
potent and support myocardial regeneration. Cell myocardial infarcts. Nature 2004;428:664-8.
2003;114:763-76. 27. Bolli R, Jneid H, Dawn B. Bone marrow cell-mediated
9. Badorff C, Brandes RP, Popp R, Rupp S, Urbich C, cardiac regeneration a veritable revolution. J Am Coll
Aicher A et al. Transdifferentiation of blood-derived Cardiol 2005;46:1659-61.
human adult endothelial progenitor cells into func- 28. Hamano K, Nishida M, Hirata K, Mikamo A, Li TS,

Vol. 53, N. 6 MINERVA CARDIOANGIOLOGICA 561


DAWN CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION

Harada M et al. Local implantation of autologous bone Baffour R et al. Catheter-based autologous bone mar-
marrow cells for therapeutic angiogenesis in patients row myocardial injection in no-option patients with
with ischemic heart disease: clinical trial and prelimi- advanced coronary artery disease: a feasibility study. J
nary results. Jpn Circ J 2001;65:845-7. Am Coll Cardiol 2003;41:1721-4.
29. Strauer BE, Brehm M, Zeus T, Kostering M, Hernandez 43. Wollert KC, Meyer GP, Lotz J, Ringes-Lichtenberg S,
A, Sorg RV et al. Repair of infarcted myocardium by Lippolt P, Breidenbach C et al. Intracoronary autologous
autologous intracoronary mononuclear bone marrow bone-marrow cell transfer after myocardial infarction:
cell transplantation in humans. Circulation 2002;106: the BOOST randomised controlled clinical trial. Lancet

A
1913-8. 2004;364:141-8.
30. Assmus B, Schachinger V, Teupe C, Britten M, Lehmann 44. Stamm C, Westphal B, Kleine HD, Petzsch M, Kittner C,
R, Dobert N et al. Transplantation of Progenitor Cells Klinge H et al. Autologous bone-marrow stem-cell
and Regeneration Enhancement in Acute Myocardial transplantation for myocardial regeneration. Lancet

C
Infarction (TOPCARE-AMI). Circulation 2002;106: 2003;361:45-6.
3009-17. 45. Bartunek J, Vanderheyden M, Vandekerckhove B,
31. Britten MB, Abolmaali ND, Assmus B, Lehmann R, Mansour S, De Bruyne B, De Bondt P et al. Intracoro-
Honold J, Schmitt J et al. Infarct remodeling after intra- nary injection of CD133-positive enriched bone marrow

T ® DI
coronary progenitor cell treatment in patients with progenitor cells promotes cardiac recovery after recent
acute myocardial infarction (TOPCARE-AMI): mecha- myocardial infarction: feasibility and safety. Circulation
nistic insights from serial contrast-enhanced magnetic 2005;112:I178-83.
resonance imaging. Circulation 2003;108:2212-8. 46. Chen SL, Fang WW, Ye F, Liu YH, Qian J, Shan SJ et al.
32. Schachinger V, Assmus B, Britten MB, Honold J, Effect on left ventricular function of intracoronary trans-

H E
Lehmann R, Teupe C et al. Transplantation of progen- plantation of autologous bone marrow mesenchymal
itor cells and regeneration enhancement in acute stem cell in patients with acute myocardial infarction.
myocardial infarction: final one-year results of the TOP- Am J Cardiol 2004;94:92-5.
CARE-AMI Trial. J Am Coll Cardiol 2004;44:1690-9. 47. Erbs S, Linke A, Adams V, Lenk K, Thiele H, Diederich
33.
IG M
Perin EC, Dohmann HF, Borojevic R, Silva SA, Sousa AL, KW et al. Transplantation of blood-derived progenitor
Mesquita CT et al. Transendocardial, autologous bone cells after recanalization of chronic coronary artery
marrow cell transplantation for severe, chronic ischemic occlusion: first randomized and placebo-controlled
heart failure. Circulation 2003;107:2294-302. study. Circ Res 2005;97:756-62.
34. Tse HF, Kwong YL, Chan JK, Lo G, Ho CL, Lau CP. 48. Menasche P, Hagege AA, Vilquin JT, Desnos M, Abergel
Angiogenesis in ischaemic myocardium by intramy- E, Pouzet B et al. Autologous skeletal myoblast trans-
R A
ocardial autologous bone marrow mononuclear cell plantation for severe postinfarction left ventricular dys-
implantation. Lancet 2003;361:47-9. function. J Am Coll Cardiol 2003;41:1078-83.
35. Fernandez-Aviles F, San Roman JA, Garcia-Frade J, 49. Smits PC, van Geuns RJ, Poldermans D, Bountioukos
Fernandez ME, Penarrubia MJ, de la Fuente L et al. M, Onderwater EE, Lee CH et al. Catheter-based
Y V

Experimental and clinical regenerative capability of intramyocardial injection of autologous skeletal


human bone marrow cells after myocardial infarction. myoblasts as a primary treatment of ischemic heart
Circ Res 2004;95:742-8. failure: clinical experience with six-month follow-up.
P R

36. Perin EC, Dohmann HF, Borojevic R, Silva SA, Sousa AL, J Am Coll Cardiol 2003;42:2063-9.
Silva GV et al. Improved exercise capacity and ischemia 50. Herreros J, Prosper F, Perez A, Gavira JJ, Garcia-Velloso
6 and 12 months after transendocardial injection of MJ, Barba J et al. Autologous intramyocardial injection
autologous bone marrow mononuclear cells for of cultured skeletal muscle-derived stem cells in patients
O E

ischemic cardiomyopathy. Circulation 2004;110: with non-acute myocardial infarction. Eur Heart J
II213-8. 2003;24:2012-20.
37. Kuethe F, Richartz BM, Sayer HG, Kasper C, Werner GS, 51. Siminiak T, Kalawski R, Fiszer D, Jerzykowska O,
Hoffken K et al. Lack of regeneration of myocardium Rzezniczak J, Rozwadowska N et al. Autologous skele-
C IN

by autologous intracoronary mononuclear bone mar- tal myoblast transplantation for the treatment of postin-
row cell transplantation in humans with large anterior farction myocardial injury: phase I clinical study with
myocardial infarctions. Int J Cardiol 2004;97:123-7. 12 months of follow-up. Am Heart J 2004;148:531-7.
38. Silva GV, Perin EC, Dohmann HF, Borojevic R, Silva SA, 52. Chachques JC, Herreros J, Trainini J, Juffe A, Rendal E,
Sousa AL et al. Catheter-based transendocardial deliv- Prosper F et al. Autologous human serum for cell cul-
ery of autologous bone-marrow-derived mononuclear ture avoids the implantation of cardioverter-defibrilla-
M

cells in patients listed for heart transplantation. Tex tors in cellular cardiomyoplasty. Int J Cardiol 2004;95
Heart Inst J 2004;31:214-9. Suppl 1:S29-33.
39. Kuethe F, Richartz BM, Kasper C, Sayer HG, Hoeffken 53. Siminiak T, Fiszer D, Jerzykowska O, Grygielska B,
K, Werner GS et al. Autologous intracoronary mononu- Rozwadowska N, Kalmucki P et al. Percutaneous trans-
clear bone marrow cell transplantation in chronic coronary-venous transplantation of autologous skele-
ischemic cardiomyopathy in humans. Int J Cardiol tal myoblasts in the treatment of post-infarction myocar-
2005;100:485-91. dial contractility impairment: the POZNAN trial. Eur
40. Yaoita H, Takase S, Maruyama Y, Sato Y, Satokawa H, Heart J 2005;26:1188-95.
Hoshi N et al. Scintigraphic assessment of the effects of 54. Davidoff AM, Ng CY, Brown P, Leary MA, Spurbeck
bone marrow-derived mononuclear cell transplanta- WW, Zhou J et al. Bone marrow-derived cells con-
tion combined with off-pump coronary artery bypass tribute to tumor neovasculature and, when modified to
surgery in patients with ischemic heart disease. J Nucl express an angiogenesis inhibitor, can restrict tumor
Med 2005;46:1610-7. growth in mice. Clin Cancer Res 2001;7:2870-9.
41. Strauer BE, Brehm M, Zeus T, Bartsch T, Schannwell C, 55. Kim CB, Braunwald E. Potential benefits of late reper-
Antke C et al. Regeneration of human infarcted heart fusion of infarcted myocardium. The open artery
muscle by intracoronary autologous bone marrow cell hypothesis. Circulation 1993;88(5 Pt 1):2426-36.
transplantation in chronic coronary artery disease: the 56. Kang HJ, Kim HS, Zhang SY, Park KW, Cho HJ, Koo BK
IACT Study. J Am Coll Cardiol 2005;46:1651-8. et al. Effects of intracoronary infusion of peripheral
42. Fuchs S, Satler LF, Kornowski R, Okubagzi P, Weisz G, blood stem-cells mobilised with granulocyte-colony

562 MINERVA CARDIOANGIOLOGICA Dicembre 2005


CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION DAWN

stimulating factor on left ventricular systolic function 74. Matsuura K, Nagai T, Nishigaki N, Oyama T, Nishi J,
and restenosis after coronary stenting in myocardial Wada H et al. Adult cardiac Sca-1-positive cells differ-
infarction: the MAGIC cell randomised clinical trial. entiate into beating cardiomyocytes. J Biol Chem 2004;
Lancet 2004;363:751-6. 279:11384-91.
57. Schober A, Hoffmann R, Opree N, Knarren S, Iofina E, 75. Martin CM, Meeson AP, Robertson SM, Hawke TJ,
Hutschenreuter G et al. Peripheral CD34+ Cells and Richardson JA, Bates S et al. Persistent expression of the
the Risk of In-Stent Restenosis in Patients With Coronary ATP-binding cassette transporter, Abcg2, identifies car-
Heart Disease. Am J Cardiol 2005;96:1196-22. diac SP cells in the developing and adult heart. Dev Biol

A
58. Murry CE, Wiseman RW, Schwartz SM, Hauschka SD. 2004;265:262-75.
Skeletal myoblast transplantation for repair of myocar- 76. Messina E, De Angelis L, Frati G, Morrone S, Chimenti
dial necrosis. J Clin Invest 1996;98:2512-23. S, Fiordaliso F et al. Isolation and expansion of adult
59. Leobon B, Garcin I, Menasche P, Vilquin JT, Audinat E, cardiac stem cells from human and murine heart. Circ

C
Charpak S. Myoblasts transplanted into rat infarcted Res 2004;95:911-21.
myocardium are functionally isolated from their host. 77. Pfister O, Mouquet F, Jain M, Summer R, Helmes M,
Proc Natl Acad Sci U S A 2003;100:7808-11. Fine A et al. CD31- but not CD31+ cardiac side popu-
60. Al Attar N, Carrion C, Ghostine S, Garcin I, Vilquin JT, lation cells exhibit functional cardiomyogenic differ-

T ® DI
Hagege AA et al. Long-term (1 year) functional and entiation. Circ Res 2005;97:52-61.
histological results of autologous skeletal muscle cells 78. Laugwitz KL, Moretti A, Lam J, Gruber P, Chen Y,
transplantation in rat. Cardiovasc Res 2003;58:142-8. Woodard S et al. Postnatal isl1+ cardioblasts enter ful-
61. Abraham MR, Henrikson CA, Tung L, Chang MG, Aon ly differentiated cardiomyocyte lineages. Nature 2005;
M, Xue T et al. Antiarrhythmic engineering of skeletal 433:647-53.

H E
myoblasts for cardiac transplantation. Circ Res 79. Smith RR, Barile L, Cho HC, Abraham MR, Messina E,
2005;97:159-67. Giacomello A et al. Unique phenotype of cardios-
62. Beltrami CA, Di Loreto C, Finato N, Rocco M, Artico D, pheres derived from human endomyocardial biopsies.
Cigola E et al. Proliferating cell nuclear antigen (PCNA), Circulation 2005;112(suppl II):II-51.
IG M
DNA synthesis and mitosis in myocytes following car-
diac transplantation in man. J Mol Cell Cardiol 1997;
80. Goodell MA, Brose K, Paradis G, Conner AS, Mulligan
RC. Isolation and functional properties of murine
29:2789-802. hematopoietic stem cells that are replicating in vivo. J
63. Kajstura J, Leri A, Finato N, Di Loreto C, Beltrami CA, Exp Med 1996;183:1797-806.
Anversa P. Myocyte proliferation in end-stage cardiac 81. Asakura A, Rudnicki MA. Side population cells from
failure in humans. Proc Natl Acad Sci USA 1998;95: diverse adult tissues are capable of in vitro
R A
8801-5. hematopoietic differentiation. Exp Hematol 2002;
64. Anversa P, Leri A, Beltrami CA, Guerra S, Kajstura J. 30:1339-45.
Myocyte death and growth in the failing heart. Lab 82. Asakura A, Seale P, Girgis-Gabardo A, Rudnicki MA.
Invest 1998;78:767-86. Myogenic specification of side population cells in skele-
Y V

65. Beltrami AP, Urbanek K, Kajstura J, Yan SM, Finato N, tal muscle. J Cell Biol 2002;159:123-34.
Bussani R et al. Evidence that human cardiac myocytes 83. Pearce DJ, Ridler CM, Simpson C, Bonnet D.
divide after myocardial infarction. N Engl J Med 2001; Multiparameter analysis of murine bone marrow side
P R

344:1750-7. population cells. Blood 2004;103:2541-6.


66. Anversa P, Nadal-Ginard B. Myocyte renewal and ven- 84. Zhou S, Schuetz JD, Bunting KD, Colapietro AM,
tricular remodelling. Nature 2002;415:240-3. Sampath J, Morris JJ et al. The ABC transporter
67. Anversa P, Leri A, Kajstura J, Nadal-Ginard B. Myocyte Bcrp1/ABCG2 is expressed in a wide variety of stem
O E

growth and cardiac repair. J Mol Cell Cardiol 2002;34: cells and is a molecular determinant of the side-pop-
91-105. ulation phenotype. Nat Med 2001;7:1028-34.
68. Nadal-Ginard B, Kajstura J, Leri A, Anversa P. Myocyte 85. Mouquet F, Pfister O, Jain M, Oikonomopoulos A,
death, growth, and regeneration in cardiac hypertrophy Ngoy S, Summer R et al. Restoration of cardiac prog-
C IN

and failure. Circ Res 2003;92:139-50. enitor cells following myocardial infarction by self-
69. Linke A, Muller P, Nurzynska D, Casarsa C, Torella D, proliferation and selective homing of bone marrow-
Nascimbene A et al. Stem cells in the dog heart are derived stem cells. Circ Res 2005 Nov 3; [Epub ahead
self-renewing, clonogenic, and multipotent and regen- of print].
erate infarcted myocardium, improving cardiac function. 86. Wright NA. Epithelial stem cell repertoire in the gut:
Proc Natl Acad Sci U S A 2005;102:8966-71. clues to the origin of cell lineages, proliferative units
M

70. Urbanek K, Rota M, Cascapera S, Bearzi C, Nascimbene and cancer. Int J Exp Pathol 2000;81:117-43.
A, De Angelis A et al. Cardiac stem cells possess growth 87. Schuster MD, Kocher AA, Seki T, Martens TP, Xiang
factor-receptor systems that after activation regenerate the G, Homma S et al. Myocardial neovascularization by
infarcted myocardium, improving ventricular function bone marrow angioblasts results in cardiomyocyte
and long-term survival. Circ Res 2005;97:663-73. regeneration. Am J Physiol 2004;287:H525-32.
71. Urbanek K, Quaini F, Tasca G, Torella D, Castaldo C, 88. Vulliet PR, Greeley M, Halloran SM, MacDonald KA,
Nadal-Ginard B et al. Intense myocyte formation from Kittleson MD. Intra-coronary arterial injection of mes-
cardiac stem cells in human cardiac hypertrophy. Proc enchymal stromal cells and microinfarction in dogs.
Natl Acad Sci U S A 2003;100:10440-5. Epub 2003 Aug Lancet 2004;363:783-4.
19. 89. Lapidot T. Mechanism of human stem cell migration and
72. Urbanek K, Torella D, Sheikh F, De Angelis A, repopulation of NOD/SCID and B2mnull NOD/SCID
Nurzynska D, Silvestri F et al. Myocardial regeneration mice. The role of SDF-1/CXCR4 interactions. Ann N Y
by activation of multipotent cardiac stem cells in Acad Sci 2001;938:83-95.
ischemic heart failure. Proc Natl Acad Sci U S A 90. Bauters C, Isner JM. The biology of restenosis. Prog
2005;102:8692-7. Cardiovasc Dis 1997;40:107-16.
73. Oh H, Bradfute SB, Gallardo TD, Nakamura T, Gaussin 91. Hofmann M, Wollert KC, Meyer GP, Menke A, Arseniev
V, Mishina Y et al. Cardiac progenitor cells from adult L, Hertenstein B et al. Monitoring of bone marrow cell
myocardium: homing, differentiation, and fusion after homing into the infarcted human myocardium.
infarction. Proc Natl Acad Sci U S A 2003;100:12313-8. Circulation 2005;111:2198-202.

Vol. 53, N. 6 MINERVA CARDIOANGIOLOGICA 563


DAWN CARDIAC STEM CELL THERAPY FOR MYOCARDIAL REGENERATION

92. Nian M, Lee P, Khaper N, Liu P. Inflammatory cytokines ual myoblasts after transplantation into muscles of
and postmyocardial infarction remodeling. Circ Res DMD patients. Nat Med 1997;3:970-7.
2004;94:1543-53. 97. Zhang M, Methot D, Poppa V, Fujio Y, Walsh K, Murry
93. Kucia M, Dawn B, Hunt G, Guo Y, Wysoczynski M, CE. Cardiomyocyte grafting for cardiac repair: graft
Majka M et al. Cells expressing early cardiac markers cell death and anti-death strategies. J Mol Cell Cardiol
reside in the bone marrow and are mobilized into the 2001;33:907-21.
peripheral blood following myocardial infarction. Circ 98. Mangi AA, Noiseux N, Kong D, He H, Rezvani M,
Res 2004;95:1191-9. Ingwall JS et al. Mesenchymal stem cells modified

A
94. Barbash IM, Chouraqui P, Baron J, Feinberg MS, Etzion with Akt prevent remodeling and restore performance
S, Tessone A et al. Systemic delivery of bone mar- of infarcted hearts. Nat Med 2003;9:1195-201.
row-derived mesenchymal stem cells to the infarcted 99. Suzuki K, Murtuza B, Smolenski RT, Sammut IA, Suzuki
myocardium: feasibility, cell migration, and body dis- N, Kaneda Y et al. Cell transplantation for the treatment

C
tribution. Circulation 2003;108:863-8. of acute myocardial infarction using vascular endothe-
95. Chin BB, Nakamoto Y, Bulte JW, Pittenger MF, Wahl lial growth factor-expressing skeletal myoblasts.
R, Kraitchman DL. 111In oxine labelled mesenchy- Circulation 2001;104(12 Suppl 1):I207-12.
mal stem cell SPECT after intravenous administration 100. Iwaguro H, Yamaguchi J, Kalka C, Murasawa S,

T ® DI
in myocardial infarction. Nucl Med Commun Masuda H, Hayashi S et al. Endothelial progenitor cell
2003;24:1149-54. vascular endothelial growth factor gene transfer for
96. Gussoni E, Blau HM, Kunkel LM. The fate of individ- vascular regeneration. Circulation 2002;105:732-8.

H E
IG M
R A
Y V
P R
O E
C IN
M

564 MINERVA CARDIOANGIOLOGICA Dicembre 2005

Anda mungkin juga menyukai