Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Jun 27.
Published in final edited form as: J Am Coll Cardiol. 2010 Oct 12;56(16):1287–1297. doi: 10.1016/j.jacc.2010.05.039

Rebuilding the Damaged Heart

The Potential of Cytokines and Growth Factors in the Treatment of Ischemic Heart Disease

Nirat Beohar *,, Jonathan Rapp †,, Sanjay Pandya *, Douglas W Losordo *,†,§
PMCID: PMC3123891  NIHMSID: NIHMS304822  PMID: 20888519

Abstract

Cytokine therapy promises to provide a noninvasive treatment option for ischemic heart disease. Cytokines are thought to influence angiogenesis directly via effects on endothelial cells or indirectly through progenitor cell-based mechanisms or by activating the expression of other angiogenic agents. Several cytokines mobilize progenitor cells from the bone marrow or are involved in the homing of mobilized cells to ischemic tissue. The recruited cells contribute to myocardial regeneration both as a structural component of the regenerating tissue and by secreting angiogenic or antiapoptotic factors, including cytokines. To date, randomized, controlled clinical trials have not reproduced the efficacy observed in pre-clinical and small-scale clinical investigations. Neverthe-less, the list of promising cytokines continues to grow, and combinations of cytokines, with or without concurrent progenitor cell therapy, warrant further investigation.

Keywords: angiogenesis, cytokines, heart failure, regenerative medicine


Cytokine therapy is a promising, noninvasive treatment approach that may prevent cardiomyocyte loss or regenerate damaged tissue. It may also be useful as an adjunctive treatment to mechanical revascularization or cell therapy and for patients with disabling ischemia despite optimal medical treatment. Cytokines are thought to benefit the damaged heart through direct effects in the myocardium and indirectly by stimulating progenitor cells. Progenitor and stem cells reside in the myocardium or are mobilized from the bone marrow in response to cardiac ischemia, and numerous reports indicate that these cells can be mobilized therapeutically by cytokines (1-14). In the ischemic heart, progenitor cells can form a structural component of the regenerating tissue— e.g., resident side-population cells (15) and lineage-negative, c-kit–positive cells (4) differentiated into endothelial cells, smooth-muscle cells, and cardiomyocytes in a murine model of myocardial infarction (MI). The recruited cells also secrete angiogenic or anti-apoptotic factors (16,17) that can maintain myocardial viability, accelerate the recovery of ischemic myocardium, or amplify function in nonischemic regions (18,19). These observations have led to the development of strategies that mobilize progenitor cells and enhance progenitor cell recruitment, thereby preserving or replacing injured tissue (4). Importantly, cytokines are pleiotropic, and their effects can be either beneficial or detrimental (Table 1) depending on the dose and timing of administration. This review focuses on several cytokines that have displayed potentially beneficial effects in pre-clinical or clinical studies of ischemic heart disease.

Table 1. Agents.

Agent Molecular Targets Effects/Mechanism Potential Detrimental Effects
VEGF VEGF receptors on endothelial cells,
 monocytes, and HSCs
Stimulates proliferation, migration, and
 tube formation
Tumorigenesis
Retinopathy
Mobilizes EPCs; improves EPC survival
 and differentiation
Flushing (protein infusion)
Hypotension (protein infusion)
PlGF VEGF receptor 1 Cross-talk with VEGF receptor 2
Mobilizes EPCs and HSCs
Associated with carotid atherosclerotic plaque
 destabilization and adverse outcomes
 in ACS
Associated with depressed LV function in
 ischemic cardiomyopathy (exact role
 undefined)
FGF FGF receptors on endothelial cells,
 smooth muscle cells, and myoblasts
Stimulates proliferation Membranous nephropathy
Dose-related hypotension
Coronary plaque destabilization
Accelerated atherosclerosis
GCSF GCSF receptors on hematopoietic and
 nonhematopoietic cells
Inhibits apoptosis
Activates the JAK-STAT pathway
Synergistic with SDF-1
Mobilizes HSCs and granulocytes
Accelerates wound healing post-MI
Possible effects on resident cardiac
 stem cells
Tumorigenesis
Dysregulated inflammation leading to
 impaired wound healing or plaque
 destabilization
Medullary bone pain
GMCSF GMCSF receptors on granulocyte and
 monocyte precursor cells and
 monocytes
Stimulates arteriogenesis
Activates monocytic cells
Mobilizes EPCs and HSCs
Infarct expansion through alteration of
 inflammation (i.e., dendritic cell function)
Plaque destabilization/acute MI
“First-dose reaction”: dyspnea, hypotension,
 hypoxia, tachycardia, or syncope
Bone pain
Peripheral edema, pericardial effusion
SCF c-kit Synergistic with colony-stimulating
 factors
Mobilizes bone marrow precursor cells
Mast cell degranulation/wheal formation
 at injection site
Hyperpigmentation
Allergic-like reaction including respiratory
 distress
Angiopoietin-1 TIE2 receptors on endothelial cells Enhances vessel maturation and
 stability
Mobilizes EPCs and HPCs
Enhanced renal inflammation and fibrosis
Pulmonary hypertension
Adverse vascular remodeling or angiogenesis
HGF c-Met receptor on numerous cells (e.g.,
 endothelial cells, cardiac myocytes,
progenitor cells)
Attracts resident cardiac stem cells Tumorigenesis
Retinopathy
GH/IGF-1 IGF receptor on vascular and satellite
cells, cardiac stem cells
Enhances skeletal and cardiac muscle

regeneration
Retinopathy
Diarrhea
Hypotension
Hypoglycemia
Erythropoietin Erythropoietin receptor on HSCs, EPCs,
 endothelial cells, and cardiac
myocytes
Promotes cell survival
Mobilizes EPCs
May accelerate death in patients with cancer
Hypertension, headache, arthralgias, and
 nausea
MI (rare)

ACS = acute coronary syndrome; EPC = endothelial progenitor cell; FGF = fibroblast growth factor; GCSF = granulocyte colony-stimulating factor; GH = growth hormone; GMCSF = granulocyte-macrophage colony-stimulating factor; HGF = hepatocyte growth factor; HPC = hematopoietic progenitor cell; HSC = hematopoietic stem cell; IGF = insulin-like growth factor; JAK = Janus kinase; LV = left ventricular; MI = myocardial infarction; PlGF = placental growth factor; SCF = stem cell factor; SDF = stromal cell-derived factor; STAT = signal transducers and activators of transcription; TIE2 = Tyrosine kinase with Ig-like loops and Epidermal growth factor homology domains-2; VEGF = vascular endothelial growth factor.

Cytokine Agents

Fibroblast growth factor (FGF)

The FGF family comprises 22 polypeptides that promote angiogenesis and arteriogenesis by modulating the phenotypes of endothelial cells and vascular smooth muscle cells. FGF4 is encoded by the heparin-binding secretory-transforming proto-oncogene and is not expressed in normal adult tissue. However, when administered to ischemic hearts, FGF4 stimulates endothelial cell proliferation and the secretion of metalloproteinases, urokinase-type plasminogen activator, and vascular endothelial growth factor (VEGF), which subsequently stimulate angiogenesis (20).

The AGENT (Angiogenic Gene Therapy) trials (Table 2) examined the effects of FGF in patients with coronary artery disease (CAD) and medically refractory angina. In the first AGENT trial, intracoronary delivery of an adenovirus coding for FGF4 transcription (Ad5FGF-4) was safe and appeared to improve exercise treadmill times (21). The phase 2 AGENT 2 trial tested whether Ad5FGF-4 treatment improved regional myocardial perfusion (22). Patients administered Ad5FGF-4, but not placebo, experienced a substantial reduction in reversible and total perfusion defect, but the results were not statistically significant. The phase 3 AGENT 3 and 4 trials (23,24) enrolled patients with Canadian Cardiovascular Society (CCS) class 2 to 4 angina who were unsuitable for mechanical revascularization. Both trials were halted prematurely when a planned interim analysis of the AGENT 3 cohort indicated that the between-group difference in the primary end point (treadmill exercise duration 12 weeks after treatment) would not reach statistical significance. Differences in secondary outcomes (such as change in CCS class or other clinical variables) were also nonsignificant.

Table 2. Randomized, Double-Blind Trials of FGF.

Grines et al. (21)
Agent
Grines et al. (23)
Agent 2
Henry et al. (24)
Agent 3
Henry et al. (24)
Agent 4
Phase 1 2 3 3
Patients Chronic CAD, LVEF ≥40%,
CCS class 2-3
Chronic CAD, LVEF ≥30%,
CCS class 4
CAD not requiring immediate
 revascularization,
 LVEF ≥30%, CCS class 2–4
 (U.S.)
“No-option” CAD patients,
 LVEF ≥30%, CCS class 2-4
 (Europe, Latin America,
 Canada)
Therapy Ad5FGF-4 Ad5FGF-4 Ad5FGF-4 Ad5FGF-4
Delivery route Intracoronary Intracoronary Intracoronary Intracoronary
Duration of follow-up 12 weeks 8 weeks 12 weeks 12 weeks
Primary end point Safety and feasibility Δ RPDS Δ ETT Δ ETT
Assessment modality Clinical and exercise treadmill test Adenosine SPECT Exercise treadmill test Exercise treadmill test
Secondary end point Δ Exercise time Δ Defect size Δ CCS class, coronary events, or death at 1 year, Δ QOL, time to
 ST-segment depression during exercise, proportion of patients
 with ≥30% increase in ETT
Treatment groups FGF (n = 60)
Placebo (n = 19)
FGF (n = 35)
Placebo (n = 17)
High-dose (n = 140)
Low-dose (n = 137)
Placebo (n = 139)
High-dose (n = 35)
Low-dose (n = 43)
Placebo (n = 38)
Dose 108.5-1010.5 vp 1010 vp High-dose: 1010 vp
Low-dose: 109 vp
Placebo = NA
High-dose: 1010 vp
Low-dose: 109 vp
Placebo = NA
Findings Trend toward ↑ in exercise time
 with therapy
Lower rates of revascularization
 at 1 year with therapy. ↓ in
 RPDS with therapy from
 baseline to follow-up*; no
 change in CCS score (not
 primary end point)
Potential therapeutic benefit
 among older patients with
 more severe angina
Pooled analysis: placebo effect much greater in men than women.
 Women had improved CCS class and ETT with therapy*
Notes One outlier in placebo group
 had 50% ↓ in RPDS
Enrollment ended early when interim analysis indicated that the
 primary end point was unlikely to differ significantly between
 groups
*

p ≤ 0.05.

Ad = adenoviral; CAD = coronary artery disease; CCS = Canadian Cardiovascular Society; ETT = exercise treadmill time; FGF = fibroblast growth factor; LVEF = left ventricular ejection fraction; NA = not applicable; QOL = quality of life; RPDS = reversible perfusion defect size; SPECT = single-photon emission computed tomography; vp = viral particles; Δ = change; ↑ = increase; ↓ = decline.

Vascular endothelial growth factor (VEGF)

The VEGF cytokines were among the first to display protective or regenerative effects in cardiac tissue. During hypoxia, the therapeutic benefit of VEGF occurs primarily through the stimulation of endothelial cell proliferation, migration, and survival, which subsequently leads to neovascularization (25-30). The extent of regeneration is determined by the tissue retention of the administered isoform and by the isoform’s affinity for VEGF receptors. Both endothelial cells and hematopoietic stem cells (HSCs) express VEGF receptors 1 and 2 (also known as Flt-1 and Flk-1, respectively) (31,32), and the expression of VEGF receptors on HSCs appears to be critical for VEGF-dependent regulation of endothelial progenitor cells (EPCs) (33,34).

Treatment with VEGF protein-enhanced collateral blood flow in animal models of chronic myocardial ischemia (35-38) and the safety and feasibility of VEGF therapy for angiogenesis has been assessed in several phase 1 studies (39-44). The VIVA (Vascular Endothelial Growth Factor in Ischemia for Vascular Angiogenesis) trial was among the first large, phase 2 trials (Table 3) completed. Patients with myocardial ischemia who were considered unsuitable for mechanical revascularization were randomized to receive placebo, low-dose recombinant human vascular endothelial growth factor (rhVEGF), or high-dose rhVEGF by intracoronary infusion, followed by peripheral infusions 3, 6, and 9 days later (45). VEGF therapy was safe, but changes in exercise treadmill time (the primary end point) did not differ significantly between groups at the 2-month follow-up visit. Four months after treatment, the only statistically significant finding was the proportion of high-dose VEGF patients who improved by at least 1 CCS class (p = 0.05 vs. placebo). In the Euroinject One trial (46), patients with CCS class 3 or 4 angina were randomized to receive 0.5-mg injections of either VEGF-A165 plasmid or a placebo plasmid into myocardial regions that displayed stress-induced perfusion defects. At the 3-month follow-up visit, the perfusion defects did not differ between treatment groups, but VEGF treatment was associated with improvements in local wall motion. Intracoronary VEGF gene transfer was also evaluated in the KAT (Kuopio Angiogenesis Trial) (47). Upon initiation of percutaneous coronary intervention (PCI), patients with CCS class 2 or 3 angina (90% of whom received stents) were randomized to receive intracoronary injections of adenovirus-encoded VEGF165, VEGF165 plasmid liposome, or Ringer’s lactate. Six months after treatment, there were no significant differences among the 3 treatment groups in functional status, but myocardial perfusion improved in patients treated with adenoviral VEGF.

Table 3. Phase 2 VEGF Trials.

Henry et al. (45) VIVA Hedman et al. (47) KAT Kastrup et al. (46) Euroinject One
Patients Stable angina, “no-option” CAD Stable CAD undergoing PCI Severe, stable angina; “no-option” CAD
Therapy Recombinant human VEGF Ad or PL VEGF Plasmid VEGF
Duration of follow-up 60 days, 120 days 6 months 3 months
Primary end point Δ ETT at 60 days Minimal lumen diameter, % stenosis Δ Myocardial perfusion
Secondary end points Δ ETT, angina, and myocardial perfusion at
day 120
Myocardial perfusion, exercise tolerance,
 incidence of new cardiac events,
 revascularization, functional class
Safety, wall motion, LVEF, angina
Treatment groups High-dose (n = 59)
Low-dose (n = 56)
Placebo (n = 63)
Adv (n = 37)
PL (n = 28)
Placebo (n = 38)
VEGF-A165 (n = 40)
Placebo (n = 40)
Dosage High-dose: 50 ng/kg per min IC for 20 min
Low-dose: 17 ng/kg per min IC for 20 min
Adv: 2 × 1010 PFU IC
PL: 2,000 μg IC
VEGF-A165: 0.5 mg
Findings ↓ in angina class at day 120 in high-dose
 group compared with placebo*;
 no benefit in ΔETT
↑ in perfusion at 6 months compared
 with baseline in Ad group*; no
 differences among groups
Improved regional wall motion in VEGF
 group compared with placebo*
*

p ≤ 0.05.

Followed by 4-h infusions on days 3, 6, and 9.

Adv = adenovirus vector; IC = intracoronary; PCI = percutaneous coronary intervention; PFU = plaque-forming unit; PL = plasmid liposome; VEGF = vascular endothelial growth factor; other abbreviations as in Table 2.

Granulocyte colony-stimulating factor (GCSF)

GCSF is a potent hematopoietic cytokine that influences the development and function of granulocytes and mobilizes progenitor cells from the bone marrow (48). Progenitor cell mobilization appears to be initiated when GCSF binds to receptors on the cell surface, which leads to the release of enzymes that digest adhesion molecules (49). GCSF also directly influences the activity of some nonhematopoietic cells, such as cardiomyocytes and endothelial cells (50). When administered shortly after MI, GCSF activates the Janus kinase/signal transducer and activator of transcription pathway, which stimulates the production of several antiapoptotic proteins, decreases cardiomyocyte death, and limits infarct size (51). In a murine model of MI, GCSF treatment was associated with improvements in left ventricular function and enhanced arteriogenesis (52). However, GCSF can also stimulate the differentiation of lineage-committed progenitor cells into neutrophils and macrophages (53), which could worsen inflammation and cardiac remodeling (54).

In phase 1 trials, GCSF administration after PCI for acute MI appeared to improve cardiac function (50,55-58). Patients in the randomized, open-label FIRSTLINE-AMI (Front-Integrated Revascularization and Stem Cell Liberation in Evolving Acute Myocardial Infarction) trial received 6 daily subcutaneous GCSF injections starting within 90 min after primary PCI for ST-segment elevation MI; the control group did not receive placebo injections but had identical post-interventional care. Four months (56) and 1 year (55) after the PCI procedure, improvements in left ventricular ejection fraction (LVEF) were significantly greater in GCSF-treated patients than in the control group. Unfortunately, these promising results were not reproduced in subsequent double-blind, placebo-controlled trials (59-61) (Table 4).

Table 4. Randomized Controlled GCSF Trials* in Patients With Acute MI.

Valgimigli
et al. (58)
Ince et al. (56)
FIRSTLINE-AMI
Ripa et al. (60)
 STEMMI
Zohlnhöfer et al. (61)
 REVIVAL-2
Engelmann et al. (59)
 G-CSF-STEMI

Treatment Group GCSF Control GCSF Control GCSF Control GCSF Control GCSF Control
GCSF dosage 5 /μg/kg/day for
 4 days beginning
37 ± 66 h after
symptom onset
10 /μg/kg/day for
 6 days beginning
85 ± 30 min after
reperfusion
10 /μg/kg/day for
 6 days beginning
1–2 days after AMI
10 /μg/kg/day for
 5 days beginning
5 days after AMI
10 /μg/kg/day for
 5 days beginning
6 h to 7 days after
symptom onset
Duration of follow-up 6 months 4 months 6 months 4–6 months 3 months
n 10 10 25 25 39 39 56 58 23 21
LVEF Increased Increased Increased Unchanged Increased Increased Increased Increased Increased Increased
Perfusion Increased Increased NR NR NR NR Increased Increased Unchanged Unchanged
LVEDV Unchanged Unchanged Unchanged Declined Increased Increased Decreased Decreased Increased Increased
LVESV NR NR NR NR Decreased Decreased Decreased Decreased Decreased Decreased
Wall thickening NR NR Increased Increased Increased Increased NR NR Increased Increased

Parameters displaying significantly better performance than was observed in the alternative treatment group are identified with bold italicized text. Within-group changes from baseline to follow-up were either insignificant (p > 0.05) or the significance was not reported.

*

Double-blind: Ripa et al. (60), Zohlnhöfer et al. (61), and Engelmann et al. (59); single-blind: Valgimigli et al. (58); open-label: Ince et al. (56).

No placebo treatment.

Decreased infarct size.

AMI = acute myocardial infarction; GCSF = granulocyte colony-stimulating factor; LVEDV = left ventricular end-diastolic volume; LVEF = left ventricular ejection fraction; LVESV = left ventricular end-systolic volume; NR = not reported.

Several nonrandomized studies have investigated the use of GCSF for treatment of chronic ischemic heart disease (Table 5). Hill et al. (62) administered 5 daily subcutaneous injections of GCSF to patients with CAD and angina. The treatment produced large increases in the number of circulating progenitor cells, but there was no improvement in LVEF, left ventricular wall motion, myocardial perfusion, or treadmill exercise time 1 month after treatment. Wang et al. (63) administered subcutaneous injections of GCSF for 6 days to 13 prospectively selected patients with severe occlusive CAD. CCS class improved from baseline to the 2-month follow-up visit, but the number of single-photon emission computed tomography image segments with perfusion defects either at rest or under stress was unchanged and LVEFs declined, perhaps because GCSF-mobilized leukocytes increased inflammation and fibrosis. Notably, progenitor cell mobilization was considerably lower in 4 patients who were considered “poor mobilizers” than in the 9 remaining patients who were considered “mobilizers.” The mobilizers, but not the poor mobilizers, experienced significant improvements in nitroglycerin use and angina frequency. These findings indicate that the potential benefit of GCSF therapy requires progenitor cell mobilization.

Table 5. GCSF Trials in Patients With Chronic Ischemic Heart Disease.

Wang et al. (63)
Hill et al. (62)
Boyle et al. (66)
Treatment Groups GCSF Control GCSF GCSF
GCSF dosage 5 /μg/kg/day for 6 days 10 /μg/kg/day for 5 days 10 /μg/kg/day for 4 days
Cell therapy None None Intracoronary infusion
Duration of follow-up 2 months 1 month 12 months
End point assessment SPECT, MRI, echocardiography MRI Angiography
n 13 16 16 5
LVEF Decreased Decreased Decreased Increased
Perfusion Unchanged Unchanged Increased Increased
LVEDV Decreased Increased NR NR
LVESV Increased Increased NR NR
Myocardial ischemia Decreased Increased Increased Decreased

Significant (p < 0.05), within-group changes from baseline to follow-up are identified with italicized text.

MRI = magnetic resonance imaging; SPECT = single-photon emission computed tomography; other abbreviations as in Table 4.

GCSF treatment did not worsen inflammation in several studies of patients with acute MI (50,55,56,5861), and the rate of restenosis did not differ significantly between GCSF and control patients in the randomized, open-label, FIRSTLINE-AMI trial (55,56) or in 3 randomized, double-blind, placebo-controlled studies (i.e., the STEMMI [Stem Cells in Myocardial Infarction], REVIVAL-2 [22 Regenerate Vital Myocardium by Vigorous Activation of Bone Marrow Stem Cells], and G-CSF-STEMI [Granulocyte Colony-Stimulating Factor ST-Segment Elevation Myocardial Infarction] trials) (59-61). However, Kang et al. (64) found restenosis in 2 of 3 patients who received GCSF alone as adjunctive therapy to primary PCI and in 5 of 7 patients who received both GCSF and infused progenitor cells as adjunctive treatments. This may have occurred because PCI was performed 4 days after GCSF injection, when GCSF-induced leukocytosis and the subsequent inflammatory response was peaking. In an uncontrolled study (65), patients received GCSF therapy 2 days after primary PCI, followed 4 days later by the collection and intracoronary injection of mobilized progenitor cells; restenosis was reported in 8 of 20 patients, and 2 patients experienced MI between 2 and 6 months after treatment.

Among 122 patients in 4 studies who received GCSF therapy soon after acute MI (50,57,60,61), 2 died during the follow-up periods (50,61), another experienced subacute stent thrombosis (60), and a fourth patient underwent emergency splenectomy for spontaneous splenic rupture (57). Wang et al. (63) found no serious vascular adverse events in 13 patients who received GCSF therapy for treatment of severe ischemic heart disease. However, Hill et al. (62) and Boyle et al. (66) reported serious vascular adverse events in 2 of 16 patients with CAD and in 1 of 5 patients with chronic ischemic heart disease, respectively, after GCSF treatment. There is no evidence of GCSF-induced arrhythmia in clinical trials of patients with ischemic heart disease, and the results from some animal studies suggest that GCSF treatment may improve electrical stability (67).

Granulocyte-macrophage colony-stimulating factor (GMCSF)

GMCSF stimulates the growth and differentiation of granulocyte and macrophage precursor cells, induces peripheral monocytosis, impedes monocyte apoptosis (5,68), and increases the number of circulating EPCs, which may then participate in regenerative activity (69). In patients with CAD who received a single intracoronary injection of GMCSF, followed by subcutaneous injections every other day for 2 weeks afterward, GMCSF was associated with significant improvement in electrocardiographic signs of myocardial ischemia during coronary balloon occlusion and with significant improvement in collateral flow index (CFI). CFI was unchanged in patients who received placebo injections. These benefits likely evolved from cytokine-induced angiogenesis or changes in collateral vascular tone, rather than through a progenitor cell–mediated mechanism, because the mobilization of progenitor cells was low. Improvements in CFI were also reported in a subsequent study of GMCSF therapy, but 2 of 7 GMCSF-treated patients experienced acute MI during the 2-week course of therapy (70). Furthermore, in animal studies of MI, cardiac remodeling worsened after treatment with romurtide to induce GMCSF expression (68,71). Thus the mechanism of action and potential inflammatory consequences associated with GMCSF must be better understood and controlled before larger human trials can be considered.

Erythropoietin (EPO)

EPO is involved in both angiogenesis and progenitor/stem cell development. Numerous tissues produce EPO in response to hypoxia and metabolic stress though a mechanism mediated by hypoxia-inducible factor (HIF)-1, and activation of the EPO receptor inhibits apoptosis (72). EPO is also believed to enhance angiogenesis by increasing the proliferation of endothelial cells and by mobilizing bone marrow–derived cells, including EPCs (73). A long-acting EPO analog, darbepoetin alpha, was safely administered to patients with acute MI but provided no functional benefit (74,75). The administration of EPO to patients with acute MI continues to be investigated in the ongoing HEBE III (Intracoronary Infusion of Autologous Mononuclear Bone Marrow Cells or Peripheral Mononuclear Blood Cells After Primary Percutaneous Coronary Intervention) and REVEAL (Reduction of Infarct Expansion and Ventricular Remodeling With Erythropoietin After Large Myocardial Infarction) trials (NCT00378352, ClinicalTrials.gov) (76).

Growth hormone (GH) and insulin-like growth factor (IGF)-1

GH is synthesized by the anterior pituitary gland, and the binding of GH to receptors in the myocardium increases IGF-1 synthesis (77). Experimental evidence has long suggested a role for the GH/IGF signaling in the myocardium (8). In experimental models of MI, subcutaneous injections of GH significantly increased hypertrophy of the viable myocardium and improved left ventricular systolic function without increasing collagen deposition or fibrosis (78,79). Moreover, cardiac stem cells have been shown to possess growth factor receptors that may participate in the activation of these cells for cardiac repair (9). The effect of GH on myocardial growth, cardiac function, and IGF-1 levels in patients with nonischemic or ischemic cardiomyopathy, and in mixed patient populations, has been investigated in several small studies (80-85). Collectively, the findings suggest that additional investigations with GH or IGF-1 are warranted, despite concerns about retinopathy and other potential long-term side effects.

Angiopoietin (Ang)

Ang1, Ang2, and Ang3/4 are believed to control the remodeling and stabilization of vessels during the later stages of adult vascular development (86,87). Ang1 is a Tyrosine kinase with Ig-like loops and Epidermal growth factor homology domains-2 (TIE2) agonist that promotes vessel survival, inhibits vascular damage, suppresses inflammatory gene expression, and stimulates vessel remodeling and angiogenesis (88). In rats, Ang1 administration after acute MI increased vascular density, and the vessels appeared to be relatively mature, with larger-sized lumens (89). In a swine model of chronic ischemia, perfusion improved significantly 4 weeks after Ang1 administration, and the improvement was sustained through week 12 (90). Curiously, Ang1 activity seems to oppose VEGF-induced angiogenesis (91). Ang2 acts as a TIE2 agonist in EPCs, which increases angiogenesis (92), but primarily antagonizes TIE2 in vascular endothelial cells, thereby reducing endothelial integrity, increasing vessel permeability, and inducing vessel destabilization and remodeling through, in part, the suppression of Ang1-mediated activity (93). Thus the influence of Ang2 on endothelial cell activity and, by extension, angiogenesis is complex and context-dependent. To date, none of the angiopoietins have been investigated for treatment of ischemic heart disease in clinical studies.

Hepatocyte growth factor (HGF)

HGF is a pluripotent growth factor synthesized by the liver. Both HGF and the HGF receptor c-Met can be found in the heart, and cardiac HGF levels are up-regulated after MI in both animal models and human subjects (94). HGF can induce both pro-angiogenic and antiapoptotic effects and is believed to reduce detrimental remodeling after MI (95). HGF has yet to be investigated for cardiac repair in humans.

Placental growth factor (PlGF)

PlGF is a member of the VEGF family of cytokines (96) and directly influences angiogenesis by binding to VEGF receptor-1, transactivating VEGF receptor-2, and enhancing VEGF activity (97). PlGF mediates endothelial cell growth, survival, and migration (98,99); mobilizes hematopoietic progenitor cells from the bone marrow; is chemotactic for monocytes and macrophages (2,100,101); and may induce monocytes to release cytokines that increase the homing of stem cells to the injured myocardium (102). Results from pre-clinical investigations suggest that PlGF can improve myocardial perfusion (11,103); however, adenoviral PlGF therapy was associated with greater atherosclerotic intimal thickening and adventitial neovascularization (104). Before clinical trials can be initiated, additional pre-clinical studies must demonstrate that PlGF can induce neovascularization without worsening atherosclerosis.

Stem cell factor (SCF)

SCF is the ligand of c-kit (CD117), a proto-oncogene receptor tyrosine kinase that is expressed on adult HSCs (105-107). Activation of the c-kit receptor (107-110) is required for the mobilization of c-kit–positive HSCs and EPCs, and c-kit–positive cells favorably impact cardiac remodeling both directly, through the repair and regeneration of infarcted myocardium, and indirectly, through an increase in neoangiogenesis (4,111-117). SCF acts synergistically with colony-stimulating factors to mobilize bone marrow-derived stem cells (106,118), improves the homing of c-kit–positive bone marrow-derived cells (118), and enhances the migration of resident cardiac stem cells to the peri-infarct zone (119). Combined SCF and GCSF therapy increased myocardial blood flow, but not function, after circumflex artery ligation in baboons (120), and treatment with SCF, GCSF, or both (compared with placebo) reduced mortality in a murine model of MI, but SCF therapy alone did not improve left ventricular performance or remodeling (121). The benefits of combined SCF and GCSF treatment have also been reported in a mouse infarct-reperfusion model (122).

Practical Considerations

Timing of therapy

The cellular environment of infarcted myocardium is dynamic, so the timing of cytokine administration is a necessary consideration during treatment. For example, the number of GCSF receptors on cardiomyocytes increases markedly soon after MI in rats (51), and prompt GCSF administration has been associated with declines in cardiomyocyte apoptosis, smaller infarct areas, and decreased ventricular dilation (51,123,124), whereas delayed GSCF treatment aggravated left ventricular remodeling in a porcine myocardial-reperfusion model (124). Four clinical trials of GCSF therapy administered at different time points and for different durations after MI yielded different patterns of efficacy (55,60,61,64).

The time-dependent effects of cytokine administration after MI could be either direct or related to the recruitment of bone marrow-derived progenitor cells. The activity of bone marrow-derived cells may differ depending on the state of the infarcted myocardium, and the expression of cell-signaling molecules may change over time. Wang et al. (30) found that the expression of VEGF and stromal cell-derived factor (SDF)-1, which is involved in the homing of progenitor cells to ischemic tissue, is not acutely elevated in ischemic myocardium (30), and that human plasma levels of SDF-1, VEGF-A, and FGF-2 reach maximum concentrations 2 to 3 weeks after MI (125). In a murine MI model, cytokines involved in progenitor-cell homing, including SDF-1, were up-regulated immediately after MI, then down-regulated over a 7-day period, and the delayed administration of GCSF (56 days after MI) improved remodeling only when SDF-1 expression was enhanced in the infarcted tissue. These observations imply that the effectiveness of cell-mobilizing agents for treatment of MI may depend on the early, cytokine-mediated recruitment of mobilized stem cells to the injured myocardium (126,127).

Safety

Because angiogenic inhibitors can reverse or reduce the formation of atherosclerotic plaques in animal models (128,129), the pro-angiogenic activity of cytokines could, in theory, worsen plaque formation or contribute to plaque destabilization (as noted in the studies of GCSF, GMCSF, and PlGF described above). However, successful angiogenic therapy may enhance re-endothelialization after vascular injury and, consequently, impede plaque formation by suppressing neointimal thickening (130). The long-term theoretical concerns associated with therapeutic angiogenesis include an increased risk for neoplastic disease and the induction or worsening of retinopathy. No evidence of a link between angiogenic cytokines and tumor development or retinopathy has been reported in clinical trials (131), but candidate patients must be adequately screened and monitored for malignancies, premalignant conditions, and retinopathy.

Reperfusion injury

In most published animal studies, MI was induced by ligating the coronary artery, which permanently halts blood flow. However, blood flow is restored in clinical presentations of MI, so the ischemia-reperfusion models used by Beohar et al. (124) and Dawn et al. (122) more closely reproduce the clinical experience. The release of cell-homing factors may differ in chronically ischemic and reperfused cells and could influence the recruitment of progenitor cells to the injured tissue. In addition, early, sustained reperfusion has been shown to reduce left ventricular remodeling after MI (62,132), and this benefit could obscure the effects of cytokine administration in clinical trials.

Summary and Future Directions

Cytokine therapy could provide an attractive treatment option for many cardiovascular diseases. Although the outcomes from clinical trials of cytokine therapy have failed to meet expectations, combinations of cytokines, with or without concurrent progenitor cell therapy, or the administration of agents (e.g., HIF-1 alpha, Sonic hedgehog) that up-regulate several angiogenic factors simultaneously, warrant further investigation. For patients with chronic CAD, therapy that combines progenitor cell-mobilizing agents with the elevated expression of cell-homing factors (e.g., SDF-1) in ischemic tissue may be particularly valuable. The effectiveness of regenerative therapies could also be improved by a more complete understanding of the cellular environment after MI. The growth factors IGF-1 (133) and HGF (134) induce expression of collagen-degrading metalloproteinases that could make the extracellular matrix more amenable to progenitor cell migration, and researchers have begun to investigate other agents that modulate the interstitial matrix, including the matrix protein Del-1, which coordinates integrin expression (135), and Cyr61, which induces angiogenesis by binding to avβ5 (136). Approaches that combine cytokine and cell therapy can be refined by continuing to characterize the relevant signaling pathways, the optimal magnitude of progenitor cell mobilization, the relative efficiency of subpopulations of bone marrow–derived cells, and the potential importance of resident cardiac progenitor cells (9).

Acknowledgments

This work was supported in part by National Institutes of Health grants HL-53354, HL-57516, HL-77428, HL-63414, HL-80137, and HL-95874. The authors have reported that they have no relationships to disclose.

Abbreviations and Acronyms

Ad5FGF

adenoviral fibroblast growth factor

Ang

angiopoietin

CAD

coronary artery disease

CCS

Canadian Cardiovascular Society

CFI

collateral flow index

EPC

endothelial progenitor cell

EPO

erythropoietin

FGF

fibroblast growth factor

GCSF

granulocyte colony-stimulating factor

GH

growth hormone

GMCSF

granulocyte-macrophage colony-stimulating factor

HGF

hepatocyte growth factor

HIF

hypoxia-inducible factor

HSC

hematopoietic stem cell

IGF

insulin-like growth factor

LVEF

left ventricular ejection fraction

MI

myocardial infarction

PCI

percutaneous coronary intervention

PlGF

placental growth factor

rhVEGF

recombinant human vascular endothelial growth factor

SCF

stem cell factor

SDF

stromal cell-derived factor

VEGF

vascular endothelial growth factor

REFERENCES

  • 1.Asahara T, Masuda H, Takahashi T, et al. Bone marrow origin of endothelial progenitor cells responsible for postnatal vasculogenesis in physiological and pathological neovascularization. Circ Res. 1999;85:221–8. doi: 10.1161/01.res.85.3.221. [DOI] [PubMed] [Google Scholar]
  • 2.Hattori K, Heissig B, Wu Y, et al. Placental growth factor reconstitutes hematopoiesis by recruiting VEGFR1(+) stem cells from bone-marrow microenvironment. Nat Med. 2002;8:841–9. doi: 10.1038/nm740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kalka C, Masuda H, Takahashi T, et al. Vascular endothelial growth factor(165) gene transfer augments circulating endothelial progenitor cells in human subjects. Circ Res. 2000;86:1198–202. doi: 10.1161/01.res.86.12.1198. [DOI] [PubMed] [Google Scholar]
  • 4.Orlic D, Kajstura J, Chimenti S, et al. Mobilized bone marrow cells repair the infarcted heart, improving function and survival. Proc Natl Acad Sci U S A. 2001;98:10344–9. doi: 10.1073/pnas.181177898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Seiler C, Pohl T, Wustmann K, et al. Promotion of collateral growth by granulocyte-macrophage colony-stimulating factor in patients with coronary artery disease: a randomized, double-blind, placebo-controlled study. Circulation. 2001;104:2012–7. doi: 10.1161/hc4201.097835. [DOI] [PubMed] [Google Scholar]
  • 6.Takahashi T, Kalka C, Masuda H, et al. Ischemia- and cytokine-induced mobilization of bone marrow-derived endothelial progenitor cells for neovascularization. Nat Med. 1999;5:434–8. doi: 10.1038/7434. [DOI] [PubMed] [Google Scholar]
  • 7.Kajstura J, Fiordaliso F, Andreoli AM, et al. IGF-1 overexpression inhibits the development of diabetic cardiomyopathy and angiotensin II-mediated oxidative stress. Diabetes. 2001;50:1414–24. doi: 10.2337/diabetes.50.6.1414. [DOI] [PubMed] [Google Scholar]
  • 8.Reiss K, Kajstura J, Capasso JM, Marino TA, Anversa P. Impairment of myocyte contractility following coronary artery narrowing is associated with activation of the myocyte IGF1 autocrine system, enhanced expression of late growth related genes, DNA synthesis, and myocyte nuclear mitotic division in rats. Exp Cell Res. 1993;207:348–60. doi: 10.1006/excr.1993.1202. [DOI] [PubMed] [Google Scholar]
  • 9.Urbanek K, Rota M, Cascapera S, et al. Cardiac stem cells possess growth factor-receptor systems that after activation regenerate the infarcted myocardium, improving ventricular function and long-term survival. Circ Res. 2005;97:663–73. doi: 10.1161/01.RES.0000183733.53101.11. [DOI] [PubMed] [Google Scholar]
  • 10.Welch S, Plank D, Witt S, et al. Cardiac-specific IGF-1 expression attenuates dilated cardiomyopathy in tropomodulin-overexpressing transgenic mice. Circ Res. 2002;90:641–8. doi: 10.1161/01.res.0000013780.77774.75. [DOI] [PubMed] [Google Scholar]
  • 11.Luttun A, Tjwa M, Moons L, et al. Revascularization of ischemic tissues by PlGF treatment, and inhibition of tumor angiogenesis, arthritis and atherosclerosis by anti-Flt1. Nat Med. 2002;8:831–40. doi: 10.1038/nm731. [DOI] [PubMed] [Google Scholar]
  • 12.Bearzi C, Rota M, Hosoda T, et al. Human cardiac stem cells. Proc Natl Acad Sci U S A. 2007;104:14068–73. doi: 10.1073/pnas.0706760104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bearzi C, Leri A, Lo Monaco F, et al. Identification of a coronary vascular progenitor cell in the human heart. Proc Natl Acad Sci U S A. 2009;106:15885–90. doi: 10.1073/pnas.0907622106. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 14.Hosoda T, D’Amario D, Cabral-Da-Silva MC, et al. Clonality of mouse and human cardiomyogenesis in vivo. Proc Natl Acad Sci U S A. 2009;106:17169–74. doi: 10.1073/pnas.0903089106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Jackson KA, Majka SM, Wang H, et al. Regeneration of ischemic cardiac muscle and vascular endothelium by adult stem cells. J Clin Invest. 2001;107:1395–402. doi: 10.1172/JCI12150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wartiovaara U, Salven P, Mikkola H, et al. Peripheral blood platelets express VEGF-C and VEGF which are released during platelet activation. Thromb Haemost. 1998;80:171–5. [PubMed] [Google Scholar]
  • 17.Huang YQ, Li JJ, Karpatkin S. Identification of a family of alternatively spliced mRNA species of angiopoietin-1. Blood. 2000;95:1993–9. [PubMed] [Google Scholar]
  • 18.Gnecchi M, He H, Liang OD, et al. Paracrine action accounts for marked protection of ischemic heart by Akt-modified mesenchymal stem cells. Nat Med. 2005;11:367–8. doi: 10.1038/nm0405-367. [DOI] [PubMed] [Google Scholar]
  • 19.Kamihata H, Matsubara H, Nishiue T, et al. Implantation of bone marrow mononuclear cells into ischemic myocardium enhances collateral perfusion and regional function via side supply of angioblasts, angiogenic ligands, and cytokines. Circulation. 2001;104:1046–52. doi: 10.1161/hc3501.093817. [DOI] [PubMed] [Google Scholar]
  • 20.Kapur NK, Rade JJ. Fibroblast growth factor 4 gene therapy for chronic ischemic heart disease. Trends Cardiovasc Med. 2008;18:133–41. doi: 10.1016/j.tcm.2008.03.002. [DOI] [PubMed] [Google Scholar]
  • 21.Grines CL, Watkins MW, Helmer G, et al. Angiogenic Gene Therapy (AGENT) trial in patients with stable angina pectoris. Circulation. 2002;105:1291–7. doi: 10.1161/hc1102.105595. [DOI] [PubMed] [Google Scholar]
  • 22.Grines C, Rubanyi GM, Kleiman NS, Marrott P, Watkins MW. Angiogenic gene therapy with adenovirus 5 fibroblast growth factor-4 (Ad5FGF-4): a new option for the treatment of coronary artery disease. Am J Cardiol. 2003;92:24N–31N. doi: 10.1016/s0002-9149(03)00965-2. [DOI] [PubMed] [Google Scholar]
  • 23.Grines CL, Watkins MW, Mahmarian JJ, et al. A randomized, double-blind, placebo-controlled trial of Ad5FGF-4 gene therapy and its effect on myocardial perfusion in patients with stable angina. J Am Coll Cardiol. 2003;42:1339–47. doi: 10.1016/s0735-1097(03)00988-4. [DOI] [PubMed] [Google Scholar]
  • 24.Henry TD, Grines CL, Watkins MW, et al. Effects of Ad5FGF-4 in patients with angina: an analysis of pooled data from the AGENT-3 and AGENT-4 trials. J Am Coll Cardiol. 2007;50:1038–46. doi: 10.1016/j.jacc.2007.06.010. [DOI] [PubMed] [Google Scholar]
  • 25.Matsunaga T, Warltier DC, Tessmer J, Weihrauch D, Simons M, Chilian WM. Expression of VEGF and angiopoietins-1 and−2 during ischemia-induced coronary angiogenesis. Am J Physiol Heart Circ Physiol. 2003;285:H352–8. doi: 10.1152/ajpheart.00621.2002. [DOI] [PubMed] [Google Scholar]
  • 26.Shweiki D, Itin A, Soffer D, Keshet E. Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis. Nature. 1992;359:843–5. doi: 10.1038/359843a0. [DOI] [PubMed] [Google Scholar]
  • 27.Takeshita S, Zheng LP, Brogi E, et al. Therapeutic angiogenesis. A single intraarterial bolus of vascular endothelial growth factor augments revascularization in a rabbit ischemic hind limb model. J Clin Invest. 1994;93:662–70. doi: 10.1172/JCI117018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Vandervelde S, van Luyn MJ, Rozenbaum MH, Petersen AH, Tio RA, Harmsen MC. Stem cell-related cardiac gene expression early after murine myocardial infarction. Cardiovasc Res. 2007;73:783–93. doi: 10.1016/j.cardiores.2006.11.030. [DOI] [PubMed] [Google Scholar]
  • 29.Voo S, Eggermann J, Dunaeva M, Ramakers-van Oosterhoud C, Waltenberger J. Enhanced functional response of CD133+ circulating progenitor cells in patients early after acute myocardial infarction. Eur Heart J. 2008;29:241–50. doi: 10.1093/eurheartj/ehm542. [DOI] [PubMed] [Google Scholar]
  • 30.Wang Y, Gabrielsen A, Lawler PR, et al. Myocardial gene expression of angiogenic factors in human chronic ischemic myocardium: influence of acute ischemia/cardioplegia and reperfusion. Microcirculation. 2006;13:187–97. doi: 10.1080/10739680600556811. [DOI] [PubMed] [Google Scholar]
  • 31.Katoh O, Tauchi H, Kawaishi K, Kimura A, Satow Y. Expression of the vascular endothelial growth factor (VEGF) receptor gene KDR, in hematopoietic cells and inhibitory effect of VEGF on apoptotic cell death caused by ionizing radiation. Cancer Research. 1995;55:5687–92. [PubMed] [Google Scholar]
  • 32.Neufeld G, Cohen T, Gengrinovitch S, Poltorak Z. Vascular endothelial growth factor (VEGF) and its receptors. FASEB J. 1999;13:9–22. [PubMed] [Google Scholar]
  • 33.Kaplan RN, Riba RD, Zacharoulis S, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438:820–7. doi: 10.1038/nature04186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Friedrich EB, Walenta K, Scharlau J, Nickenig G, Werner N. CD34−/CD133+/VEGFR-2+ endothelial progenitor cell subpopulation with potent vasoregenerative capacities. Circ Res. 2006;98:e20–5. doi: 10.1161/01.RES.0000205765.28940.93. [DOI] [PubMed] [Google Scholar]
  • 35.Banai S, Jaklitsch MT, Shou M, et al. Angiogenic-induced enhancement of collateral blood flow to ischemic myocardium by vascular endothelial growth factor in dogs. Circulation. 1994;89:2183–9. doi: 10.1161/01.cir.89.5.2183. [DOI] [PubMed] [Google Scholar]
  • 36.Harada K, Friedman M, Lopez JJ, et al. Vascular endothelial growth factor administration in chronic myocardial ischemia. Am J Physiol. 1996;270:H1791–802. doi: 10.1152/ajpheart.1996.270.5.H1791. [DOI] [PubMed] [Google Scholar]
  • 37.Lopez JJ, Laham RJ, Stamler A, et al. VEGF administration in chronic myocardial ischemia in pigs. Cardiovasc Res. 1998;40:272–81. doi: 10.1016/s0008-6363(98)00136-9. [DOI] [PubMed] [Google Scholar]
  • 38.Pearlman JD, Hibberd MG, Chuang ML, et al. Magnetic resonance mapping demonstrates benefits of VEGF-induced myocardial angiogenesis. Nat Med. 1995;1:1085–9. doi: 10.1038/nm1095-1085. [DOI] [PubMed] [Google Scholar]
  • 39.Henry TD, Rocha-Singh K, Isner JM, et al. Intracoronary administration of recombinant human vascular endothelial growth factor to patients with coronary artery disease. Am Heart J. 2001;142:872–80. doi: 10.1067/mhj.2001.118471. [DOI] [PubMed] [Google Scholar]
  • 40.Losordo DW, Vale PR, Symes JF, et al. Gene therapy for myocardial angiogenesis: initial clinical results with direct myocardial injection of phVEGF165 as sole therapy for myocardial ischemia. Circulation. 1998;98:2800–4. doi: 10.1161/01.cir.98.25.2800. [DOI] [PubMed] [Google Scholar]
  • 41.Rosengart TK, Lee LY, Patel SR, et al. Angiogenesis gene therapy: phase I assessment of direct intramyocardial administration of an adenovirus vector expressing VEGF121 cDNA to individuals with clinically significant severe coronary artery disease. Circulation. 1999;100:468–74. doi: 10.1161/01.cir.100.5.468. [DOI] [PubMed] [Google Scholar]
  • 42.Symes JF, Losordo DW, Vale PR, et al. Gene therapy with vascular endothelial growth factor for inoperable coronary artery disease. Ann Thorac Surg. 1999;68:830–6. doi: 10.1016/s0003-4975(99)00807-3. discussion 6–7. [DOI] [PubMed] [Google Scholar]
  • 43.Vale PR, Losordo DW, Milliken CE, et al. Left ventricular electromechanical mapping to assess efficacy of phVEGF(165) gene transfer for therapeutic angiogenesis in chronic myocardial ischemia. Circulation. 2000;102:965–74. doi: 10.1161/01.cir.102.9.965. [DOI] [PubMed] [Google Scholar]
  • 44.Vale PR, Losordo DW, Milliken CE, et al. Randomized, single-blind, placebo-controlled pilot study of catheter-based myocardial gene transfer for therapeutic angiogenesis using left ventricular electromechanical mapping in patients with chronic myocardial ischemia. Circulation. 2001;103:2138–43. doi: 10.1161/01.cir.103.17.2138. [DOI] [PubMed] [Google Scholar]
  • 45.Henry TD, Annex BH, McKendall GR, et al. The VIVA trial: Vascular endothelial growth factor in Ischemia for Vascular Angiogenesis. Circulation. 2003;107:1359–65. doi: 10.1161/01.cir.0000061911.47710.8a. [DOI] [PubMed] [Google Scholar]
  • 46.Kastrup J, Jorgensen E, Ruck A, et al. Direct intramyocardial plasmid vascular endothelial growth factor-A165 gene therapy in patients with stable severe angina pectoris. A randomized double-blind placebo-controlled study: the Euroinject One trial. J Am Coll Cardiol. 2005;45:982–8. doi: 10.1016/j.jacc.2004.12.068. [DOI] [PubMed] [Google Scholar]
  • 47.Hedman M, Hartikainen J, Syvanne M, et al. Safety and feasibility of catheter-based local intracoronary vascular endothelial growth factor gene transfer in the prevention of postangioplasty and in-stent restenosis and in the treatment of chronic myocardial ischemia: phase II results of the Kuopio Angiogenesis Trial (KAT) Circulation. 2003;107:2677–83. doi: 10.1161/01.CIR.0000070540.80780.92. [DOI] [PubMed] [Google Scholar]
  • 48.Anderlini P, Donato M, Chan KW, et al. Allogeneic blood progenitor cell collection in normal donors after mobilization with filgrastim: the M.D. Anderson Cancer Center experience. Transfusion. 1999;39:555–60. doi: 10.1046/j.1537-2995.1999.39060555.x. [DOI] [PubMed] [Google Scholar]
  • 49.Levesque JP, Hendy J, Takamatsu Y, Simmons PJ, Bendall LJ. Disruption of the CXCR4/CXCL12 chemotactic interaction during hematopoietic stem cell mobilization induced by GCSF or cyclo-phosphamide. J Clin Invest. 2003;111:187–96. doi: 10.1172/JCI15994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kuethe F, Figulla HR, Herzau M, et al. Treatment with granulocyte colony-stimulating factor for mobilization of bone marrow cells in patients with acute myocardial infarction. Am Heart J. 2005;150:115. doi: 10.1016/j.ahj.2005.04.030. [DOI] [PubMed] [Google Scholar]
  • 51.Harada M, Qin Y, Takano H, et al. G-CSF prevents cardiac remodeling after myocardial infarction by activating the Jak-Stat pathway in cardiomyocytes. Nat Med. 2005;11:305–11. doi: 10.1038/nm1199. [DOI] [PubMed] [Google Scholar]
  • 52.Deindl E, Zaruba MM, Brunner S, et al. G-CSF administration after myocardial infarction in mice attenuates late ischemic cardiomyopathy by enhanced arteriogenesis. FASEB J. 2006;20:956–8. doi: 10.1096/fj.05-4763fje. [DOI] [PubMed] [Google Scholar]
  • 53.Suda T, Suda J, Kajigaya S, et al. Effects of recombinant murine granulocyte colony-stimulating factor on granulocyte-macrophage and blast colony formation. Exp Hematol. 1987;15:958–65. [PubMed] [Google Scholar]
  • 54.Vandervelde S, van Luyn MJ, Tio RA, Harmsen MC. Signaling factors in stem cell-mediated repair of infarcted myocardium. J Mol Cell Cardiol. 2005;39:363–76. doi: 10.1016/j.yjmcc.2005.05.012. [DOI] [PubMed] [Google Scholar]
  • 55.Ince H, Petzsch M, Kleine HD, et al. Prevention of left ventricular remodeling with granulocyte colony-stimulating factor after acute myocardial infarction: final 1-year results of the Front-Integrated Revascularization and Stem Cell Liberation in Evolving Acute Myocardial Infarction by Granulocyte Colony-Stimulating Factor (FIRSTLINE-AMI) Trial. Circulation. 2005;112:I73–80. doi: 10.1161/CIRCULATIONAHA.104.524827. [DOI] [PubMed] [Google Scholar]
  • 56.Ince H, Petzsch M, Kleine HD, et al. Preservation from left ventricular remodeling by front-integrated revascularization and stem cell liberation in evolving acute myocardial infarction by use of granulocyte-colony-stimulating factor (FIRSTLINE-AMI) Circulation. 2005;112:3097–106. doi: 10.1161/CIRCULATIONAHA.105.541433. [DOI] [PubMed] [Google Scholar]
  • 57.Suarez de Lezo J, Torres A, Herrera I, et al. Effects of stem-cell mobilization with recombinant human granulocyte colony stimulating factor in patients with percutaneously revascularized acute anterior myocardial infarction. Rev Esp Cardiol. 2005;58:253–61. [PubMed] [Google Scholar]
  • 58.Valgimigli M, Rigolin GM, Cittanti C, et al. Use of granulocytecolony stimulating factor during acute myocardial infarction to enhance bone marrow stem cell mobilization in humans: clinical and angiographic safety profile. Eur Heart J. 2005;26:1838–45. doi: 10.1093/eurheartj/ehi289. [DOI] [PubMed] [Google Scholar]
  • 59.Engelmann MG, Theiss HD, Hennig-Theiss C, et al. Autologous bone marrow stem cell mobilization induced by granulocyte colony-stimulating factor after subacute ST-segment elevation myocardial infarction undergoing late revascularization: final results from the G-CSF-STEMI (Granulocyte Colony-Stimulating Factor ST-Segment Elevation Myocardial Infarction) trial. J Am Coll Cardiol. 2006;48:1712–21. doi: 10.1016/j.jacc.2006.07.044. [DOI] [PubMed] [Google Scholar]
  • 60.Ripa RS, Jorgensen E, Wang Y, et al. Stem cell mobilization induced by subcutaneous granulocyte-colony stimulating factor to improve cardiac regeneration after acute ST-elevation myocardial infarction: result of the double-blind, randomized, placebo-controlled stem cells in myocardial infarction (STEMMI) trial. Circulation. 2006;113:1983–92. doi: 10.1161/CIRCULATIONAHA.105.610469. [DOI] [PubMed] [Google Scholar]
  • 61.Zohlnhöfer D, Ott I, Mehilli J, et al. Stem cell mobilization by granulocyte colony-stimulating factor in patients with acute myocardial infarction: a randomized controlled trial. JAMA. 2006;295:1003–10. doi: 10.1001/jama.295.9.1003. [DOI] [PubMed] [Google Scholar]
  • 62.Hill JM, Syed MA, Arai AE, et al. Outcomes and risks of granulocyte colony-stimulating factor in patients with coronary artery disease. J Am Coll Cardiol. 2005;46:1643–8. doi: 10.1016/j.jacc.2005.01.067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Wang Y, Tagil K, Ripa RS, et al. Effect of mobilization of bone marrow stem cells by granulocyte colony stimulating factor on clinical symptoms, left ventricular perfusion and function in patients with severe chronic ischemic heart disease. Int J Cardiol. 2005;100:477–83. doi: 10.1016/j.ijcard.2004.12.006. [DOI] [PubMed] [Google Scholar]
  • 64.Kang HJ, Kim HS, Zhang SY, et al. Effects of intracoronary infusion of peripheral blood stem-cells mobilised with granulocyte-colony stimulating factor on left ventricular systolic function and restenosis after coronary stenting in myocardial infarction: the MAGIC cell randomised clinical trial. Lancet. 2004;363:751–6. doi: 10.1016/S0140-6736(04)15689-4. [DOI] [PubMed] [Google Scholar]
  • 65.Steinwender C, Hofmann R, Kammler J, et al. Effects of peripheral blood stem cell mobilization with granulocyte-colony stimulating factor and their transcoronary transplantation after primary stent implantation for acute myocardial infarction. Am Heart J. 2006;151:1296.e7–13. doi: 10.1016/j.ahj.2006.03.012. [DOI] [PubMed] [Google Scholar]
  • 66.Boyle AJ, Whitbourn R, Schlicht S, et al. Intra-coronary high-dose CD34+ stem cells in patients with chronic ischemic heart disease: a 12-month follow-up. Int J Cardiol. 2006;109:21–7. doi: 10.1016/j.ijcard.2005.05.024. [DOI] [PubMed] [Google Scholar]
  • 67.Kuhlmann MT, Kirchhof P, Klocke R, et al. G-CSF/SCF reduces inducible arrhythmias in the infarcted heart potentially via increased connexin43 expression and arteriogenesis. J Exp Med. 2006;203:87–97. doi: 10.1084/jem.20051151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Maekawa Y, Anzai T, Yoshikawa T, et al. Effect of granulocyte-macrophage colony-stimulating factor inducer on left ventricular remodeling after acute myocardial infarction. J Am Coll Cardiol. 2004;44:1510–20. doi: 10.1016/j.jacc.2004.05.083. [DOI] [PubMed] [Google Scholar]
  • 69.Deng Z, Yang C, Deng H, et al. Effects of GM-CSF on the stem cells mobilization and plasma C-reactive protein levels in patients with acute myocardial infarction. Int J Cardiol. 2006;113:92–6. doi: 10.1016/j.ijcard.2006.06.014. [DOI] [PubMed] [Google Scholar]
  • 70.Zbinden S, Zbinden R, Meier P, Windecker S, Seiler C. Safety and efficacy of subcutaneous-only granulocyte-macrophage colony-stimulating factor for collateral growth promotion in patients with coronary artery disease. J Am Coll Cardiol. 2005;46:1636–42. doi: 10.1016/j.jacc.2005.01.068. [DOI] [PubMed] [Google Scholar]
  • 71.Naito K, Anzai T, Sugano Y, et al. Differential effects of GM-CSF and G-CSF on infiltration of dendritic cells during early left ventricular remodeling after myocardial infarction. J Immunol. 2008;181:5691–701. doi: 10.4049/jimmunol.181.8.5691. [DOI] [PubMed] [Google Scholar]
  • 72.van der Meer P, Lipsic E, Henning RH, et al. Erythropoietin induces neovascularization and improves cardiac function in rats with heart failure after myocardial infarction. J Am Coll Cardiol. 2005;46:125–33. doi: 10.1016/j.jacc.2005.03.044. [DOI] [PubMed] [Google Scholar]
  • 73.Heeschen C, Aicher A, Lehmann R, et al. Erythropoietin is a potent physiologic stimulus for endothelial progenitor cell mobilization. Blood. 2003;102:1340–6. doi: 10.1182/blood-2003-01-0223. [DOI] [PubMed] [Google Scholar]
  • 74.Ruifrok WP, de Boer RA, Westenbrink BD, van Veldhuisen DJ, van Gilst WH. Erythropoietin in cardiac disease: new features of an old drug. Eur J Pharmacol. 2008;585:270–7. doi: 10.1016/j.ejphar.2008.01.054. [DOI] [PubMed] [Google Scholar]
  • 75.Lipsic E, van der Meer P, Voors AA, et al. A single bolus of a long-acting erythropoietin analogue darbepoetin alfa in patients with acute myocardial infarction: a randomized feasibility and safety study. Cardiovasc Drugs Ther. 2006;20:135–41. doi: 10.1007/s10557-006-7680-5. [DOI] [PubMed] [Google Scholar]
  • 76.Belonje AM, Voors AA, van Gilst WH, et al. Effects of erythropoietin after an acute myocardial infarction: rationale and study design of a prospective, randomized, clinical trial (HEBE III) Am Heart J. 2008;155:817–22. doi: 10.1016/j.ahj.2007.12.036. [DOI] [PubMed] [Google Scholar]
  • 77.Fazio S, Palmieri EA, Biondi B, Cittadini A, Sacca L. The role of the GH-IGF-I axis in the regulation of myocardial growth: from experimental models to human evidence. Eur J Endocrinol. 2000;142:211–6. doi: 10.1530/eje.0.1420211. [DOI] [PubMed] [Google Scholar]
  • 78.Duerr RL, Huang S, Miraliakbar HR, Clark R, Chien KR, Ross J., Jr. Insulin-like growth factor-1 enhances ventricular hypertrophy and function during the onset of experimental cardiac failure. J Clin Invest. 1995;95:619–27. doi: 10.1172/JCI117706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Grimm D, Cameron D, Griese DP, Riegger GA, Kromer EP. Differential effects of growth hormone on cardiomyocyte and extra-cellular matrix protein remodeling following experimental myocardial infarction. Cardiovasc Res. 1998;40:297–306. doi: 10.1016/s0008-6363(98)00181-3. [DOI] [PubMed] [Google Scholar]
  • 80.Osterziel KJ, Strohm O, Schuler J, et al. Randomised, double-blind, placebo-controlled trial of human recombinant growth hormone in patients with chronic heart failure due to dilated cardiomyopathy. Lancet. 1998;351:1233–7. doi: 10.1016/S0140-6736(97)11329-0. [DOI] [PubMed] [Google Scholar]
  • 81.Volterrani M, Desenzani P, Lorusso R, d’Aloia A, Manelli F, Giustina A. Haemodynamic effects of intravenous growth hormone in congestive heart failure. Lancet. 1997;349:1067–8. doi: 10.1016/S0140-6736(97)24015-8. [DOI] [PubMed] [Google Scholar]
  • 82.Isgaard J, Bergh CH, Caidahl K, Lomsky M, Hjalmarson A, Bengtsson BA. A placebo-controlled study of growth hormone in patients with congestive heart failure. Eur Heart J. 1998;19:1704–11. doi: 10.1053/euhj.1998.1123. [DOI] [PubMed] [Google Scholar]
  • 83.Fazio S, Sabatini D, Capaldo B, et al. A preliminary study of growth hormone in the treatment of dilated cardiomyopathy. N Engl J Med. 1996;334:809–14. doi: 10.1056/NEJM199603283341301. [DOI] [PubMed] [Google Scholar]
  • 84.Genth-Zotz S, Zotz R, Geil S, Voigtlander T, Meyer J, Darius H. Recombinant growth hormone therapy in patients with ischemic cardiomyopathy: effects on hemodynamics, left ventricular function, and cardiopulmonary exercise capacity. Circulation. 1999;99:18–21. doi: 10.1161/01.cir.99.1.18. [DOI] [PubMed] [Google Scholar]
  • 85.O’Driscoll JG, Green DJ, Ireland M, Kerr D, Larbalestier RI. Treatment of end-stage cardiac failure with growth hormone. Lancet. 1997;349:1068. doi: 10.1016/s0140-6736(05)62290-8. [DOI] [PubMed] [Google Scholar]
  • 86.Hattori K, Dias S, Heissig B, et al. Vascular endothelial growth factor and angiopoietin-1 stimulate postnatal hematopoiesis by recruitment of vasculogenic and hematopoietic stem cells. J Exp Med. 2001;193:1005–14. doi: 10.1084/jem.193.9.1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Brindle NP, Saharinen P, Alitalo K. Signaling and functions of angiopoietin-1 in vascular protection. Circ Res. 2006;98:1014–23. doi: 10.1161/01.RES.0000218275.54089.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Gamble JR, Drew J, Trezise L, et al. Angiopoietin-1 is an antipermeability and anti-inflammatory agent in vitro and targets cell junctions. Circ Res. 2000;87:603–7. doi: 10.1161/01.res.87.7.603. [DOI] [PubMed] [Google Scholar]
  • 89.Takahashi K, Ito Y, Morikawa M, et al. Adenoviral-delivered angiopoietin-1 reduces the infarction and attenuates the progression of cardiac dysfunction in the rat model of acute myocardial infarction. Mol Ther. 2003;8:584–92. doi: 10.1016/s1525-0016(03)00230-2. [DOI] [PubMed] [Google Scholar]
  • 90.Shim WS, Li W, Zhang L, et al. Angiopoietin-1 promotes functional neovascularization that relieves ischemia by improving regional reperfusion in a swine chronic myocardial ischemia model. J Biomed Sci. 2006;13:579–91. doi: 10.1007/s11373-006-9082-x. [DOI] [PubMed] [Google Scholar]
  • 91.Visconti RP, Richardson CD, Sato TN. Orchestration of angiogenesis and arteriovenous contribution by angiopoietins and vascular endothelial growth factor (VEGF) Proc Natl Acad Sci U S A. 2002;99:8219–24. doi: 10.1073/pnas.122109599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kim KL, Shin IS, Kim JM, et al. Interaction between Tie receptors modulates angiogenic activity of angiopoietin2 in endothelial progenitor cells. Cardiovasc Res. 2006;72:394–402. doi: 10.1016/j.cardiores.2006.08.002. [DOI] [PubMed] [Google Scholar]
  • 93.Roviezzo F, Tsigkos S, Kotanidou A, et al. Angiopoietin-2 causes inflammation in vivo by promoting vascular leakage. J Pharmacol Exp Ther. 2005;314:738–44. doi: 10.1124/jpet.105.086553. [DOI] [PubMed] [Google Scholar]
  • 94.Yasuda S, Goto Y, Baba T, et al. Enhanced secretion of cardiac hepatocyte growth factor from an infarct region is associated with less severe ventricular enlargement and improved cardiac function. J Am Coll Cardiol. 2000;36:115–21. doi: 10.1016/s0735-1097(00)00675-6. [DOI] [PubMed] [Google Scholar]
  • 95.Bussolino F, DiRenzo MF, Ziche M, et al. Hepatocyte growth factor is a potent angiogenic factor which stimulates endothelial cell mobility and growth. J Cell Biol. 1992;119:629–41. doi: 10.1083/jcb.119.3.629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Carmeliet P, Moons L, Luttun A, et al. Synergism between vascular endothelial growth factor and placental growth factor contributes to angiogenesis and plasma extravasation in pathological conditions. Nat Med. 2001;7:575–83. doi: 10.1038/87904. [DOI] [PubMed] [Google Scholar]
  • 97.Autiero M, Waltenberger J, Communi D, et al. Role of PlGF in the intra- and intermolecular cross talk between the VEGF receptors Flt1 and Flk1. Nat Med. 2003;9:936–43. doi: 10.1038/nm884. [DOI] [PubMed] [Google Scholar]
  • 98.Mattei MG, Borg JP, Rosnet O, Marme D, Birnbaum D. Assignment of vascular endothelial growth factor (VEGF) and placenta growth factor (PLGF) genes to human chromosome 6p12-p21 and 14q24-q31 regions, respectively. Genomics. 1996;32:168–9. doi: 10.1006/geno.1996.0098. [DOI] [PubMed] [Google Scholar]
  • 99.Autiero M, Luttun A, Tjwa M, Carmeliet P. Placental growth factor and its receptor, vascular endothelial growth factor receptor-1: novel targets for stimulation of ischemic tissue revascularization and inhibition of angiogenic and inflammatory disorders. J Thromb Haemost. 2003;1:1356–70. doi: 10.1046/j.1538-7836.2003.00263.x. [DOI] [PubMed] [Google Scholar]
  • 100.Rafii S, Avecilla S, Shmelkov S, et al. Angiogenic factors reconstitute hematopoiesis by recruiting stem cells from bone marrow microenvironment. Ann N Y Acad Sci. 2003;996:49–60. doi: 10.1111/j.1749-6632.2003.tb03232.x. [DOI] [PubMed] [Google Scholar]
  • 101.Pilarczyk K, Sattler KJ, Galili O, et al. Placenta growth factor expression in human atherosclerotic carotid plaques is related to plaque destabilization. Atherosclerosis. 2008;196:333–40. doi: 10.1016/j.atherosclerosis.2006.10.038. [DOI] [PubMed] [Google Scholar]
  • 102.Iwama H, Uemura S, Naya N, et al. Cardiac expression of placental growth factor predicts the improvement of chronic phase left ventricular function in patients with acute myocardial infarction. J Am Coll Cardiol. 2006;47:1559–67. doi: 10.1016/j.jacc.2005.11.064. [DOI] [PubMed] [Google Scholar]
  • 103.Kolakowski S, Jr., Berry MF, Atluri P, et al. Placental growth factor provides a novel local angiogenic therapy for ischemic cardiomyopathy. J Card Surg. 2006;21:559–64. doi: 10.1111/j.1540-8191.2006.00296.x. [DOI] [PubMed] [Google Scholar]
  • 104.Khurana R, Moons L, Shafi S, et al. Placental growth factor promotes atherosclerotic intimal thickening and macrophage accumulation. Circulation. 2005;111:2828–36. doi: 10.1161/CIRCULATIONAHA.104.495887. [DOI] [PubMed] [Google Scholar]
  • 105.Zsebo K, Williams D, Geissler EN, et al. Stem cell factor is encoded at the Sl locus of the mouse and is the ligand for the c-kit tyrosine kinase receptor. Cell. 1990:213–24. doi: 10.1016/0092-8674(90)90302-u. [DOI] [PubMed] [Google Scholar]
  • 106.Broudy VC. Stem cell factor and hematopoiesis. Blood. 1997;90:1345–64. [PubMed] [Google Scholar]
  • 107.Chabot B, Stephenson DA, Chapman VM, Besmer P, Bernstein A. The proto-oncogene c-kit encoding a transmembrane tyrosine kinase receptor maps to the mouse W locus. Nature. 1988;335:88–9. doi: 10.1038/335088a0. [DOI] [PubMed] [Google Scholar]
  • 108.Heissig B, Hattori K, Dias S, et al. Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand. Cell. 2002;109:625–37. doi: 10.1016/s0092-8674(02)00754-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Fazel SS, Chen L, Angoulvant D, et al. Activation of c-kit is necessary for mobilization of reparative bone marrow progenitor cells in response to cardiac injury. FASEB J. 2008;22:930–40. doi: 10.1096/fj.07-8636com. [DOI] [PubMed] [Google Scholar]
  • 110.Broudy VC, Lin NL, Priestley GV, Nocka K, Wolf NS. Interaction of stem cell factor and its receptor c-kit mediates lodgment and acute expansion of hematopoietic cells in the murine spleen. Blood. 1996;88:75–81. [PubMed] [Google Scholar]
  • 111.Balsam LB, Wagers AJ, Christensen JL, Kofidis T, Weissman IL, Robbins RC. Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium. Nature. 2004;428:668–73. doi: 10.1038/nature02460. [DOI] [PubMed] [Google Scholar]
  • 112.Beltrami AP, Barlucchi L, Torella D, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114:763–76. doi: 10.1016/s0092-8674(03)00687-1. [DOI] [PubMed] [Google Scholar]
  • 113.Kajstura J, Rota M, Whang B, et al. Bone marrow cells differentiate in cardiac cell lineages after infarction independently of cell fusion. Circ Res. 2005;96:127–37. doi: 10.1161/01.RES.0000151843.79801.60. [DOI] [PubMed] [Google Scholar]
  • 114.Li TS, Hayashi M, Ito H, et al. Regeneration of infarcted myocardium by intramyocardial implantation of ex vivo transforming growth factor-beta-preprogrammed bone marrow stem cells. Circulation. 2005;111:2438–45. doi: 10.1161/01.CIR.0000167553.49133.81. [DOI] [PubMed] [Google Scholar]
  • 115.Orlic D, Kajstura J, Chimenti S, et al. Bone marrow cells regenerate infarcted myocardium. Nature. 2001;410:701–5. doi: 10.1038/35070587. [DOI] [PubMed] [Google Scholar]
  • 116.Li TS, Hamano K, Nishida M, et al. CD117+ stem cells play a key role in therapeutic angiogenesis induced by bone marrow cell implantation. Am J Physiol Heart Circ Physiol. 2003;285:H931–7. doi: 10.1152/ajpheart.01146.2002. [DOI] [PubMed] [Google Scholar]
  • 117.Li TS, Ito H, Hayashi M, Furutani A, Matsuzaki M, Hamano K. Cellular expression of integrin-beta 1 is of critical importance for inducing therapeutic angiogenesis by cell implantation. Cardiovasc Res. 2005;65:64–72. doi: 10.1016/j.cardiores.2004.08.019. [DOI] [PubMed] [Google Scholar]
  • 118.Lutz M, Rosenberg M, Kiessling F, et al. Local injection of stem cell factor (SCF) improves myocardial homing of systemically delivered c-kit + bone marrow-derived stem cells. Cardiovasc Res. 2008;77:143–50. doi: 10.1093/cvr/cvm027. [DOI] [PubMed] [Google Scholar]
  • 119.Kuang D, Zhao X, Xiao G, et al. Stem cell factor/c-kit signaling mediated cardiac stem cell migration via activation of p38 MAPK. Basic Res Cardiol. 2008;103:265–73. doi: 10.1007/s00395-007-0690-z. [DOI] [PubMed] [Google Scholar]
  • 120.Norol F, Merlet P, Isnard R, et al. Influence of mobilized stem cells on myocardial infarct repair in a nonhuman primate model. Blood. 2003;102:4361–8. doi: 10.1182/blood-2003-03-0685. [DOI] [PubMed] [Google Scholar]
  • 121.Ohtsuka M, Takano H, Zou Y, et al. Cytokine therapy prevents left ventricular remodeling and dysfunction after myocardial infarction through neovascularization. FASEB J. 2004;18:851–3. doi: 10.1096/fj.03-0637fje. [DOI] [PubMed] [Google Scholar]
  • 122.Dawn B, Guo Y, Rezazadeh A, et al. Postinfarct cytokine therapy regenerates cardiac tissue and improves left ventricular function. Circ Res. 2006;98:1098–105. doi: 10.1161/01.RES.0000218454.76784.66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Minatoguchi S, Takemura G, Chen XH, et al. Acceleration of the healing process and myocardial regeneration may be important as a mechanism of improvement of cardiac function and remodeling by postinfarction granulocyte colony-stimulating factor treatment. Circulation. 2004;109:2572–80. doi: 10.1161/01.CIR.0000129770.93985.3E. [DOI] [PubMed] [Google Scholar]
  • 124.Beohar N, Flaherty J, Davidson CJ, et al. Granulocyte-colony stimulating factor administration after myocardial infarction in a porcine ischemia-reperfusion model: functional and pathological effects of dose timing. Catheter Cardiovasc Interv. 2007;69:257–65. doi: 10.1002/ccd.20925. [DOI] [PubMed] [Google Scholar]
  • 125.Wang Y, Johnsen HE, Mortensen S, et al. Changes in circulating mesenchymal stem cells, stem cell homing factor, and vascular growth factors in patients with acute ST elevation myocardial infarction treated with primary percutaneous coronary intervention. Heart. 2006;92:768–74. doi: 10.1136/hrt.2005.069799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Askari AT, Unzek S, Popovic ZB, et al. Effect of stromal-cell-derived factor 1 on stem-cell homing and tissue regeneration in ischaemic cardiomyopathy. Lancet. 2003;362:697–703. doi: 10.1016/S0140-6736(03)14232-8. [DOI] [PubMed] [Google Scholar]
  • 127.Lee MS, Lill M, Makkar RR. Stem cell transplantation in myocardial infarction. Rev Cardiovasc Med. 2004;5:82–98. [PubMed] [Google Scholar]
  • 128.Moulton KS, Heller E, Konerding MA, Flynn E, Palinski W, Folkman J. Angiogenesis inhibitors endostatin or TNP-470 reduce intimal neovascularization and plaque growth in apolipoprotein E-deficient mice. Circulation. 1999;99:1726–32. doi: 10.1161/01.cir.99.13.1726. [DOI] [PubMed] [Google Scholar]
  • 129.Moulton KS, Vakili K, Zurakowski D, et al. Inhibition of plaque neovascularization reduces macrophage accumulation and progression of advanced atherosclerosis. Proc Natl Acad Sci U S A. 2003;100:4736–41. doi: 10.1073/pnas.0730843100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Van Belle E, Tio FO, Chen D, Maillard L, Kearney M, Isner JM. Passivation of metallic stents following arterial gene transfer of phVEGF165 inhibits thrombus formation and intimal thickening. J Am Coll Cardiol. 1997;29:1371–9. doi: 10.1016/s0735-1097(97)00049-1. [DOI] [PubMed] [Google Scholar]
  • 131.Vale PR, Rauh G, Wuensch DI, Pieczek A, Schainfeld RM. Influence of vascular endothelial growth factor on diabetic retinopathy. Circulation. 1998;17:I353. [Google Scholar]
  • 132.Stone GW, Grines CL, Cox DA, et al. Comparison of angioplasty with stenting, with or without abciximab, in acute myocardial infarction. N Engl J Med. 2002;346:957–66. doi: 10.1056/NEJMoa013404. [DOI] [PubMed] [Google Scholar]
  • 133.Zhang D, Brodt P. Type 1 insulin-like growth factor regulates MT1-MMP synthesis and tumor invasion via PI 3-kinase/Akt signaling. Oncogene. 2003;22:974–82. doi: 10.1038/sj.onc.1206197. [DOI] [PubMed] [Google Scholar]
  • 134.Hamasuna R, Kataoka H, Moriyama T, Itoh H, Seiki M, Koono M. Regulation of matrix metalloproteinase-2 (MMP-2) by hepatocyte growth factor/scatter factor (HGF/SF) in human glioma cells: HGF/SF enhances MMP-2 expression and activation accompanying up-regulation of membrane type-1 MMP. Int J Cancer. 1999;82:274–81. doi: 10.1002/(sici)1097-0215(19990719)82:2<274::aid-ijc19>3.0.co;2-2. [DOI] [PubMed] [Google Scholar]
  • 135.Zhong J, Eliceiri B, Stupack D, et al. Neovascularization of ischemic tissues by gene delivery of the extracellular matrix protein Del-1. J Clin Invest. 2003;112:30–41. doi: 10.1172/JCI17034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Babic AM, Kireeva ML, Kolesnikova TV, Lau LF. CYR61, a product of a growth factor-inducible immediate early gene, promotes angiogenesis and tumor growth. Proc Natl Acad Sci U S A. 1998;95:6355–60. doi: 10.1073/pnas.95.11.6355. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES