ArticlePDF Available

VEGFR-1 activation-induced MMP-9-dependent invasion in hepatocellular carcinoma

Authors:

Abstract and Figures

Aim: VEGFR-1 can promote invasion through epithelial-mesenchymal transition induction in hepatocellular carcinoma (HCC). This study aims to elucidate VEGFR-1 impact on proteolytic enzymes profile involved with invasion. Materials & methods: The effect on cell invasion was evaluated by invasive and migration assays with and without VEGFR-1 activation. The mechanism was investigated by real-time PCR, western blot and gelatin zymography using inhibitors for MMP-9. In total, 95 HCC patients were enrolled for its clinical value evaluation. Results: VEGFR-1 activation induced invasion in HCC cells with an increase in the expression and activity of MMP-9 and Snail. MMP-9 blockage effectively inhibited VEGFR-1-induced invasion. High coexpression of both in HCC predicted a worse clinical outcome. Conclusion: Data show a novel VEGFR-1 activation-to-MMP-9 mechanism promoting HCC invasion.
Content may be subject to copyright.
3143
ISSN 1479-6694
Future Oncol. (2015) 11(23), 3143–3157
part of
10.2217/fon.15.263 © 2015 Future Medicine Ltd
RESEARCH ARTICLE
VEGFR-1 activation-induced
MMP-9-dependent invasion in
hepatocellular carcinoma
Tao Li1, Yuhua Zhu1, Lihui Han2, Wanhua Ren1, Hui Liu1 & Chengyong Qin*,1
1Department o f Infectious diseases, Shan dong Provincial Hospital Affiliated to Shandong Univer sity, Jinan 250021, China
2Departm ent of Immunology, Shandong University Scho ol of Medicine, Jinan 250012, China
*Author for correspondence: qchengy@163.com
Aim: VEGFR-1 can promote invasion through epithelial–mesenchymal transition induction in
hepatocellular carcinoma (HCC). This study aims to elucidate VEGFR-1 impact on proteolytic
enzymes prole involved with invasion. Materials & methods: The eect on cell invasion
was evaluated by invasive and migration assays with and without VEGFR-1 activation. The
mechanism was investigated by real-time PCR, western blot and gelatin zymography using
inhibitors for MMP-9. In total, 95 HCC patients were enrolled for its clinical value evaluation.
Results: VEGFR-1 activation induced invasion in HCC cells with an increase in the expression
and activity of MMP-9 and Snail. MMP-9 blockage eectively inhibited VEGFR-1-induced
invasion. High coexpression of both in HCC predicted a worse clinical outcome. Conclusion:
Data show a novel VEGFR-1 activation-to-MMP-9 mechanism promoting HCC invasion.
KEYWORDS
hepatocellular carcinoma
invasion MMP-9 Snail
V EGFR -1
Hepatocellular carcinoma (HCC) is an aggressive disease with a poor prognosis due to the tumor
invasiveness, intrahepatic spread and extrahepatic metastasis [1] . HCC metastasis is frequently associ-
ated with epithelial–mesenchymal transition (EMT) and extracellular matrix (ECM) degradation
using proteases including matrix metalloproteinases (MMPs) [2, 3]. Understanding of the molecular
mechanisms regulating the metastatic and invasive behavior of this malignant tumor is essential
for improving the treatments.
Activation of growth factor receptors is one of the most important mechanisms for survival and
invasion of human cancers. VEGFR-1 is one of three typical membrane-bound tyrosine kinase
receptors that specifically bind VEGF-B and PGF. VEGFR-1-mediated signaling promotes cancer
metastasis via three major mechanisms: promotion of angiogenesis, activation of tumor cell prolifera-
tion and induction of tumor EMT, which endows tumor cell with a more invasive phenotype [4 ,5] .
Activation of VEGFR-1 on the endothelial cells by its ligands results in the enhanced angiogenesis
and metastasis. VEGFR-1 signaling facilitates malignant angiogenesis through the enhancement of
endothelial migration and activity [6 ] . VEGFR-1 activation in breast cancer cells promotes tumor
growth via activation of MAPK and PI3K/Akt signaling [7, 8] . Moreover, activation of VEGFR-1
in cancer cells can also induce EMT, a critical mechanism for acquisition of invasive potential and
promotion of cancer cell metastasis [4,9–10] . Furthermore, it is reported that activation/induction of
proteolytic enzyme-mediated degradation of the ECM is also one of the essential steps in carcinoma
invasion [1]. Many growth factors including TGF-β, EGF and IGF-1 have the ability to enhance the
cancer cell invasiveness through activation of proteolytic enzymes [11 –13 ] . While the mechanisms
of angiogenesis and EMT are well established, the VEGFR-1 signaling pathways that regulate the
activation of proteolytic enzymes are poorly understood.
For reprint orders, please contact: reprints@futuremedicine.com
Future Oncol. (2015) 11(23)
3144
RESEaRch aRticlE Li, Zhu, Han, Ren, Liu & Qin
future science group
MMPs, a family of metalloendopeptidases
that cleave the protein components of the ECM
and endothelial cell basement membrane, play
a pivotal role in tumor-associated angiogenesis
and cancer metastasis [14– 16 ] . MMP-9, a mem-
ber of MMPs family, is particularly interest-
ing, because it is correlated with the tumor
recurrence and survival in patients with HCC.
Quantitative real-time PCR studies have also
shown that the expression of MMP-9 was signifi-
cantly upregulated in the high-metastatic HCC
cells lines [1] . Moreover, animal studies have
confirmed that blocking MMP-9 can inhibit
tumor cell invasion and metastasis. Importantly,
it is hypothesized that MMP-9 enhances tumor
angiogenesis through the VEGF–VEGFR sign-
aling system [17 ] . Recent studies have shown
that VEGFR-1 activation markedly promotes
pulmonary metastasis through the induction of
MMP-9 secretion in premetastatic lung endothe-
lial cells and macrophages [18] . However, the
mechanisms of MMP-9 regulation and its func-
tion of VEGFR-1 activation in HCC remain to
be established.
In this study, we investigated one possible
mechanism of MMP-9 regulation and its role in
activation of VEGFR-1-mediated invasion. We
showed that Snail, an EMT regulator induced
by activation of VEGFR-1, was involved in the
regulation of the expression of MMP-9. We
also showed a strong correlation of high coex-
pression of VEGFR-1 and MMP-9 with the
patient tumor invasion and demonstrated that
high coexpression of VEGFR-1 and MMP-9 is
a prognostic marker for HCC.
Materials & methods
Reagents
Recombinant human VEGF-B167 was purchased
from R&D Systems, Inc. (MN, USA). Purified
human immunoglobulin (Sigma, MO, USA),
a nonspecific IgG, was used as a control. Both
of them were added to cultures at a final con-
centration of 50 ng/ml. The broad spectrum
MMPs inhibitor (GM6001) was purchased from
Chemicon (CA, USA). Anti-MMP-9 monoclo-
nal antibody (Ab-1) for functional blocking
was purchased from Calbiochem (CA, USA).
Monoclonal anti-VEGFR-1 and polyclonal anti-
Snail were purchased from Abcam Cambridge
(MA, USA). Polyclonal anti-MMP-9 and
anti-β-actin were purchased from Santa Cruz
Biotechnology (CA, USA). FITC-conjugated
antirabbit IgG, horseradish peroxidase
(HRP)-conjugated antirabbit IgG and HRP-
conjugated antigoat IgG were purchased from
Zhongshan Biotechnology (Beijing, China).
Cell cultures
All cell lines, except for those with particular
notes, were obtained from Cell bank of Chinese
Academy of Science. MHCC97H was obtained
from the Liver Cancer Institute of Zhongshan
Hospital, Fudan University, China. The human
umbilical vein endothelial cells (which are used
as positive control of VEGFR-1 expression) and
NIH3T3 were kindly provided by Professor Ling
Gao (Central Laboratory of Provincial Hospital
Affiliated to Shandong University, China). HCC
cell line and NIH3T3 was cultured as described
previously [19] . SMMC7721 and HepG2 cell lines
were routinely cultured in Dulbecco’s modified
Eagle’s medium (DMEM; Hyclone, USA) sup-
plemented with 10% FBS (Gibco, NY, USA) and
antibiotics (100 U/ml penicillin and 100 μg/ml
streptomycin; Gibco), at 37°C in 5% CO2 and
95% air. Results from all studies were confirmed
in at least three independent experiments.
Patients, follow up & tissue microarrays
construction
A total of 95 HCC patients who underwent cura-
tive resection treatment at Provincial Hospital
Aff iliated to Shandong University during
October 2006 to August 2007 were analyzed.
The criteria for curative resection were described
previously [2 0] . No patients received any preop-
erative anticancer treatment. All pathological
features such as cirrhosis, tumor encapsulation,
tumor size and tumor number were defined
histologically. Tumor differentiation and stage
were grade by Edmondson grading system and
the 2002 International Union Against Cancer
TNM classification system (6th edition), respec-
tively. This study was approved by the Ethics
Committee of Provincial Hospital Affiliated
to Shandong University, and informed consent
was obtained from all patients. Patients’ clinical
characteristics were summarized in Tab le 1.
All of the patients received follow-up tests
including liver function, tumor markers and
ultrasonography with an interval of 4–7 months
after curative resection. In case of suspicious
tumor recurrence, computed tomography scan-
ning was used immediately. Treatment modali-
ties after relapse were administered depending
on the individual situation, including radi-
ofrequency ablation and selected transcatheter
3145
VEGFR-1 activation-induced MMP-9-dependent invasion in hepatocellular carcinoma RESEaRch aRticlE
future science group www.futuremedicine.com
arterial chemoembolization. The median follow-
up period was 38 months. Overall survival (OS)
and recurrence-free survival (RFS) were defined
as the interval between curative resection and
death or recurrence.
A total of 95 pathology-proven HCC tissues
and 33 peritumoral tissues (only corresponding
to HCC tissues of moderate differentiation) were
collected. After examining HE-stained slides for
the location and differentiation of cancerous tis-
sues and peritumoral tissues, tissue microarrays
(TMAs; collaborated with Shanghai Biochip
Co, Ltd, Shanghai, China) were constructed.
Immunouorescent microscopy
Paraformaldehyde-fixed, permeabilized
SMMC7721 cells cultured on glass slide for 48h
were incubated with primary antibodies over-
night at 4°C followed by incubation with second-
ary fluorescent antibodies. Nuclei were stained
with DAPI (Vector Laboratories, CA, USA),
and the resultant immunof luorescence was
observed under a fluorescent microscope (Leica
Microsystems, Wetzlar, Germany). Negative
controls were treated identically but with the
primary antibodies omitted.
Cell migration assay
Cell migration was examined utilizing a wound
healing assay as described previously [2 1] . Briey,
SMCC7721 cells were digested and plated in
a six-well plate until 100% confluence. Three
parallel wounds with a size of approximately
400 μm were made in the cell monolayer using
a sterile pipette tip. The monolayer was washed
twice with PBS followed by incubation with
VEGF-B167 (R&D Systems, Inc., MN, USA) or
nonspecific human IgG (Sigma) in DMEM + 1%
FBS for 24 h. Relative cell migration was calcu-
lated as the percentage of the remaining cell-free
area among the initial wounded areas. Closure
of the wounded area was monitored using an
inverted microscope (Leica Microsystems,
Germany) attached with a digital camera. Cell
migration distance was determined by meas-
uring the width of the wound and calculated
using the following equation: cell migration
distance = initial half-width of the wound - the
width of the wound after migration/2. Results
are expressed as mean ± standard deviation of
three independent experiments.
Matrigel invasive assay
Invasion assay was performed using 24-well
Transwell chamber with a pore size of 8 μm
(Costar, NY, USA) and the inserts were coated
with Matrigel (BD, Bioscience, MA, USA). Cells
were digested and diluted to 1 × 106 cells/ml in
DMEM + 1% FBS. A total of 100 μl of cells
was then added on the upper chamber of the
insert. Consequently, DMEM + 10% with or
without VEGF-B167 was added to the upper
chamber. After 48 h of incubation, cells on the
upper surface of the filters were removed with
a cotton tip. The migrant cells were fixed with
4% paraformaldehyde and stained with hema-
toxylin. Numbers of cells migrating to the mem-
brane were counted in five randomly chosen fields
under a light microscope (×200). The average
number of migrated cell per microscopic field was
analyzed. Results are expressed as mean ± stand-
ard deviation of three independent experiments.
Table 1. Characteristics and univariate survival analysis of 95 hepatocellular carcinoma patients.
Factor n Mean RFS (95% CI) p-value Mean OS (95% CI) p-value
Age (≤50 years/>50 years) 41/54 31.6 (27.6– 35.5 )/28 .4 (25. 3–31.5) 0.236 44.2 (41.4–47.0)/38.9 (35.8–42.1) 0.087
Gender (male/female) 79/16 30.6 (28.0–33.3)/25.3 (19.3–31.3) 0. 074 40.7 (38.3–43.2)/43.1 (38.2–47.9) 0.293
Liver cirrhosis (no/yes) 15/80 29.6 (23.7–35.5)/29. 8 (27.1–32.5) 0.724 45.8 (42.6 –49.1)/40.4 (37.9–42.9) 0.459
Tumor size (≤5 cm/>5 cm) 38/57 38.6 (35.3– 41.8)/23.7 (21.3–26.1) 0.0001* 47.5 (4 5.4 –4 9.6) /37.0 (3 4. 0– 39. 9) 0.0001*
Tumor number (single/multiple) 50/45 35.5 (32.2–38.9)/23.3 (20.8–32.2) 0.0001* 47.1 (45.1–49.2)/34.8 (31.7–38.0) 0.0001*
Tumor tissue encapsulation (yes/no) 53/42 35.2 (32.2–38.2)/22.5 (19.8–25.3) 0.0001* 45.6 (43.1–48.1)/35.5 (32.3–38.6) 0.0001*
TNM stage (I–II/III–IV) 50/45 36.9 (34.0–39.7)/21.5 (19.1–23.9) 0.0001* 47.6 (45.7–49.4)/34.0 (30.9–37.0) 0.0001*
VEGFR-1 (low/high)46/43 33.2 (29.4 –36.9)/25.1 (22.2–27.9) 0.0001* 44.5 (41.9–47.0)/37.2 (33.6–40.8) 0.023*
Snail (low/high)43/49 34.0 (30.4–37.6)/24.7 (22.0–27.4) 0.0001* 47.4 (45.1–49.8)/35.1 (32.2–37.9) 0.0 01*
MMP-9 (low/high)53/33 30.7 (27.3–34.1)/28.5 (24.6–32.5) 0.327 42.2 (39.0–45.4)/40.1 (36.9–43.4) 0. 274
VEGFR-1/MMP-9 high coexpression
(yes/no)
19/60 23.2 (19.7–26.6)/30.4 (27.3–33.6) 0.003* 35.6 (30.6–40.7)/42.5 (39.8–45.2) 0.02*
Cores of six (V EGFR-1), three (Snail) and nine (MMP-9) patient s were detached from tissues microarrays duri ng immunstaining.
*p < 0.05 was considere d statistically significant .
OS: Overall survival; RFS: Recurrence-free survival.
Future Oncol. (2015) 11(23)
3146
RESEaRch aRticlE Li, Zhu, Han, Ren, Liu & Qin
future science group
Western blot analysis
Cells were grown to 70–80% confluence in
DMEM + 10%FBS and were then serum-
deprived overnight in DMEM + 1% FBS.
Subsequently, cells were treated with VEGF-B167
or nonspecific IgG in DMEM + 1% FBS. Cells
in both groups were collected by scraping, and
the whole cell lysate was prepared using lysis
buffer (1 × PBS, 0.5% sodium dexycholate,
1 mM Na3VO4, 1% NP-40, 0.1% SDS, 5 mM
EDTA, 1 mM PMSF). After centrifugation,
40 μg of total protein from each group were
loaded onto 12% SDS-PAGE gels. After elec-
trophoresis, the proteins were transferred to a
nitrocellulose membrane, which was blocked,
incubated with appropriate antibodies and
developed with chemiluminescence as described
previously [19] . The fluorescence of the protein
bands was detected and quantified with LAS-
3000 system (Fuji Systems, Tokyo, Japan).
β-actin was used as a loading control.
Quantitative real-time PCR
Quantitative real-time PCR (qRT-PCR) was
performed on ABI 7500 Real-time PCR sys-
tem (Applied Biosystems, CA, USA) using the
SYBR Premix Ex Taq (TaKaRa Biotechnology
[Dalian] Co., Ltd, Dalian, China) accord-
ing to the manufacturer’s instructions.
Primer pairs were as follows: Snail: forward
5´-TAGGCCCTGGCTGCTACAAG-3´,
reverse 5´-GAGAAGGTCCGAGC ACACG-3´;
β-actin: forward-TGACGTGG ACATCC
GCAAAG-3´, reverse 5´-CTGGAAGGTGGA
CAGCGAGG-3´; MMP-9: forward 5´-GTGCT
GGGCTGCTTTGCTG-3´, reverse 5´-GT
CGCCCTCAAAGGTTTGGAAT-3. The
data were quantified with the comparative Ct
method for relative gene expression [22] . β-actin
was used as the reference gene.
Gelatin zymography
Cells at 70–80% confluence were treated with
VEGF-B167 or nonspecific IgG after overnight
serum deprivation in DMEM + 1% FBS. These
conditioned media were centrifuged to remove
cell debris and concentrated 15–20-fold using
Spin-x UF concentrators (Costar, NY, USA)
before being utilized as gelatinase. Volumes for
each group were adjusted to 15 μg protein as
determined by BCA. Gelatin zymography was
performed as described previously [23] . Brie y,
samples were mixed with SDS sample buffer
without reducing agent and separated on 10%
SDS-PAGE gels containing 0.1% gelatin. Gels
were incubated at 37°C in digestion buffer
after SDS was removed by washing with 2.5%
Triton X-100-containing buffer. Gels were
stained with Coomassie Brilliant Blue R250
and the gelatinolytic activities were detected as
clear bands against a blue background.
Immunohistochemistry analysis
The immunohistochemistry protocols were
described previously [24 ] . Negative controls
were treated identically but with the primary
antibodies omitted. Immunoreactivity was
evaluated independently by two pathologists.
Immunohistochemical expression of VEGFR-1
and Snail were quantified by determining the
percentage of positive tumor cells at each inten-
sity scores: 1+ (absent), 2+ (weak), 3+ (moder-
ate) and 4+ (strong). The sum of these scores
gave a final score for every core: 1, 1.01~2;
2.01~3; 3.01~4, as reported elsewhere [12 ] .
Positive expression on the endothelial cells was
not counted. Staining of MMP-9 was quan-
tified by log-transformed integrated optical
d ensity as reported previously [2] .
Statistical analysis
For VEGFR-1, Snail and MMP-9, the cutoff
for definition of subgroups was the median
value of 2.3, 1.9 and 5.2, respectively [3]. The
normality test indicated that the raw data
has normal distribution and data were ana-
lyzed using SPSS15.0 and expressed as mean
± standard deviation. Differences between
means were compared using either a standard
two-sided independent t-test for two-group
comparisons or one-way analysis of variance
for multiple comparisons. The association was
assessed using Pearson correlation coefficient.
Survival curves were generated using Kaplan–
Meier method, and Log-rank test was used
to compare patients’ survival between sub-
groups. p < 0.05 was considered as st atistically
significant.
Results
VEGFR-1 expression in HCC cell lines &
surgical specimens
Western blot demonstrated that VEGFR-1
(180 kDa) was expressed in four HCC cell lines
including Hep3B, HepG2, SMMC7721 and
MHCC97H (Figure 1A). human umbilical vein
endothelial cells was used as a positive con-
trol [25]. Immunofluorescence analysis showed
3147
VEGFR-1 activation-induced MMP-9-dependent invasion in hepatocellular carcinoma RESEaRch aRticlE
future science group www.futuremedicine.com
that VEGFR-1 was localized in the cytomem-
brane of SMMC7721 cells (Figure 1B, upper
panel). By contrast, the negative control with-
out addition of primary anti-VEGFR-1 anti-
body did not show specific staining (Figure1B,
lower panel). Immunohistochemical staining
was performed to determine the expression of
VEGFR-1 in the HCC tumor and peritumoral
tissues of surgical specimens. VEGFR-1 was
barely detectable in the hepatocyte of peritu-
moral tissues (Figure1Ci). However, VEGFR-1
expression was distinctly observed on the cell
membrane in the parenchyma (Figure 1Cii) and
also localized at the vascular endothelial cells
and bile duct epithelial cells in the stroma of
HCC tissues (Figure1Ciii).
Figure 1. VEGFR-1 protein expression in hepatocellular carcinoma. (A) Western blot shows
expression of VEGFR-1 protein in all four cell lines. HUVECs were used as a positives control.
(B) Immunoreactivity for the VEGFR-1 was clearly observed (arrow, upper panel). The negative
control was performed similarly without the primary antibody (lower panel). Scale bar: 50 μm.
(C)Immunohistochemical staining of VEGFR-1 was localized in hepatocytes of peritumoral tissues
(i), the cell membrane in the parenchyma (ii) and the vascular endothelial cell (upper arrow) and bile
duct epithelial cells (lower arrow) in stroma of hepatocellular carcinoma tissues (iii). Scale bar:50 μm.
HUVEC: Human umbilical vein endothelial cell.
HUVECs
Hep3B
HepG2
SMMC 7721
MHCC97H
kDa
-180
-43
VEGFR-1
VEGFR-1 DAPI Merged
β-actin
DAPI VEGFR-1
iiii
ii
Future Oncol. (2015) 11(23)
3148
RESEaRch aRticlE Li, Zhu, Han, Ren, Liu & Qin
future science group
100
90
80
70
60
50
40
30
20
10
0
0
0 h
12
12 h
24
24 h
Time (h)
Relative cell migration (%)
70
60
50
40
30
20
10
0
Untreated group
Untreated group
Non-specific IgG-treated group
VEGF-B167-treated group
Untreated group
IgG
IgG
VEGF-B
VEGF-B
Invaded cells/HPF (n)
**
*
*
3149
Figure 2. VEGFR-1 activation enhances SMMC7721 cell migration and invasion (see facing
page). (A) Wound healing assay was performed to analyze the cell migration. Bars indicated
standard deviation; *p < 0.05 versus non-specic IgG group; **p = 0.01 versus untreated group.
(B) Photographs of cell migration at the point of 12 h and 24 h post-scratch. Scale bar: 100 μm.
(C)Matrigel invasive assay was performed to analyze the cell invasion. Data shown are representative
of three independent experiments. Bars indicated standard deviation; *p < 0.0001 versus nonspecic
IgG group. Scale bar:100 μm.
VEGFR-1 activation-induced MMP-9-dependent invasion in hepatocellular carcinoma RESEaRch aRticlE
future science group www.futuremedicine.com
VEGFR-1 activation promotes cell
migration & invasion of SMMC7721 cell line
Having determined that VEGFR-1 is present in
HCC cell lines and tissues, we determined its role
in the tumor progression. We first tested if acti-
vation of VEGFR-1 by VEGF-B167 can enhance
the migration and invasion of SMMC7721 by
wound healing and cell invasion assays. At the
point of 12 h and 24 h postscratch, the relative
cell migration was 60.3 and 12.3%, respectively,
for the untreated group, 60.5 and 8.4%, respec-
tively, for the nonspecific IgG group, but 58.6
and 1.4%, respectively, for the VEGF-B167-treated
group. There was no significant increase in the
migration among these three groups in initial
12 h. However, the motility of the VEGF-B167-
treated cells was significantly increased compared
with the nonspecific IgG-treated cells after 24 h
(Figure2A & B).
Because the activation of VEGFR-1 appeared
to promote the migration of HCC cells, we next
assessed the effect of VEGFR-1 on invasion using
Matrigel Invasive assay. Cells were treated as
described in Material and methods and invasion
was determined at 48 h. As shown in Figure2C,
invasion of cells treated with VEGF-B167 were
significantly increased compared with that of
the cell treated with nonspecific IgG. These
data suggest that the activation of VEGFR-1 by
VEGF-B167 promotes the migration and invasion
of SMMC7721 cells.
VEGFR-1 activation induces MMP-9
expression in HCC cell lines
MMP-9 and MMP-2 are the major proteinases
for the proteolytic degradation of the ECM com-
ponents, leading to the cancer cell invasion [2]. In
HCC metastasis, MMP-9 possesses greater bio-
logical activity than MMP-2 [20,26]. Therefore, we
wondered whether VEGFR-1 activation-induced
cell invasion was due to the elevated expression
of MMP-9. As shown in Figure 3A, the level of
MMP-9 mRNA expression in the cell treated with
VEGF-B167 for 32 h was increased by 4.3-fold and
2.6-fold in SMMC7721 and HepG2 cells, respec-
tively. The expression level was declined at 48 h
post-treatment. Gelatin zymography assay showed
that MMP-9 gelatinolytic activity in the culture
medium from VEGF-B167-treated SMMC7721
cells was significantly higher than that in the non-
specific IgG group or untreated cells (Figure 3B,
left panel). However, MMP-2 activity was barely
detectable (Figure 3B, left panel). In contrast, in
the VEGF-B167-treated HepG2 cells, the activ-
ity of both MMP-9 and MMP-2 were increased
in comparison with the controls (Figure 3B, right
panel). The predominant band was detected at
92 kDa and 72 kDa corresponding to the proac-
tive form that becomes active during zymography.
Western blot indicated that the level of MMP-9
protein, detected as two bands at 92 kDa and
82 kDa, was increased in these representative
two cell lines (Figure 3B). Immunofluorescence
detection was performed to confirm the changes
of MMP-9. As shown in Figure 3C, the immu-
noreactivtity for MMP-9 in the cytoplasm of
both SMMC7721 and HepG2 cells was signifi-
cantly elevated in the VEGF-B167-treated group.
However, there were not significantly differences
in the immunoreactivtity for MMP-2 between
two groups of HepG2 cells (data not shown).
Taken together, these data indicated that MMP-9
expression and activity were increased in HCC
cells upon activation of VEGFR-1.
VEGFR-1 activation promotes
MMP-9-dependent Matrigel invasion
Our data have indicated that VEGFR-1 acti-
vation enhances the invasiveness accompanied
by an increase in expression of MMP-9 in
SMMC7721 cell line. To determine the specific
contribution of MMPs, especially MMP-9 in
this cellular invasion, a broad-spectrum phar-
macologic MMP inhibitor (GM6001), and
a functional blocking anti-MMP-9 antibody
(Ab-1) were used to block the proteinase activ-
ity. VEGFR-1 activation-induced Matrigel inva-
sion was completely blocked after addition of
GM6001, indicating that metalloproteinase
activity is required for the cellular invasion
(Figure 4A). To examine if MMP-9 is involved
in the cellular invasion, functional blocking
Future Oncol. (2015) 11(23)
3150
RESEaRch aRticlE Li, Zhu, Han, Ren, Liu & Qin
future science group
5.0
4.0
3.0
2.0
1.0
0.0
0 h8 h24 h28 h32 h48 h
5.0
4.0
3.0
2.0
1.0
0.0
0 h8 h24 h28 h32 h48 h
MMP-9/β-actin
MMP-9/β-actin
SMMC7721
SMMC7721
SMMC7721
HepG2
HepG2
HepG2
VEGF-B167 treated
Non-specific IgG-
treated group
VEGF-B167-treated group
Non-specific IgG-treated
group
Untreated
group
Untreated
group
IgG IgG
VEGF-B VEGF-B
MMP-9 92 kDa
82 kDa
Fold Upper band
Down band
MMP-9
92 kDa
MMP-2
72 kDa
β-actin
1
1
1.26
1.04
3.58
1.85
1
1
1.53
1.12
2.46
1.24
Non-specific IgG group VEGF-B167-treated group
1
1
1
.
26
1.0
.0
4
4
3
.5
8
1.8
1.8
1
1
1
1
.
53
1.1
2
2
.4
6
1.2
4
DAPI MMP-9
DAPI
M
MP-
9
*
*
3151
Figure 3. VEGFR-1 activation upregulates the expression and activity of MMP-9 in hepatocellular
carcinoma cell lines (see facing page). (A) Relative expression of MMP-9 and β-actin was determined
by quantitative real-time PCR, normalized to non-specic IgG group, and arbitrarily set at 1.0. Bars
indicate standard deviation; *p<0.001 for both versus the zero time point, and the similar results
were obtained in three independent experiments. (B) The expression and activity of MMP-9 in
SMMC7721 and HepG2 cells was analyzed by western blot and gelatin zymography. Data shown are
representative of three independent experiments. (C) Immunouorescence detection of MMP-9 in
SMMC7721 (upper panel) and HepG2 cells (lower panel). Scale bar:50 μm.
VEGFR-1 activation-induced MMP-9-dependent invasion in hepatocellular carcinoma RESEaRch aRticlE
future science group www.futuremedicine.com
antibody against MMP-9 was used and the
results showed that abrogation of MMP-9 activ-
ity significantly reduced cell invasion by approxi-
mately 84% when compared with SMMC7721
cells treated with VEGF-B167 alone (Figure 4B).
These results support a key role for MMP-9 in
VEGFR-1 activation-induced invasion.
VEGFR-1 activation induces Snail
expression in HCC cell lines
Recent reports have shown that Snail promotes
HCC invasion by inducing EMT and/or upregu-
lating MMPs expression [2 ,3] . Moreover, Snail is
activated downstream of several reporters and bio-
active compounds including VEGFRs [1 2 , 27–29] .
Accompanied with activation of VEGFR-1, Snail
expression is significantly elevated in pancreas
cancer cells [4] . Thus, we examined the specific
role of Snail in VEGFR-1 mediated MMP-9
expression in HCC cells. The results showed
that mRNA level of Snail in SMMC7721 and
HepG2 were increased by 8.5-fold and 2.2-fold
after 28 h and 24 h of VEGF-B167 treatment,
respectively (Figure 5A). Western blot analysis also
indicated the Snail protein level was increased
after VEGF-B167 treatment (Figure 5B).
High expression of both VEGFR-1 & MMP-9
correlated with tumor metastasis
The results that activation of VEGFR-1 promotes
MMP-9 expression of in HCC cell lines suggest
that VEGFR-1 and MMP-9 might be involved
in the tumor invasion and metastasis. To test this
hypothesis, we investigated the clinical signi-
cance of VEGFR-1 in HCC and its relationship
to MMP-9 and Snail expression by immunohis-
tochemistry ( Tabl e 1). The representative exam-
ples of VEGFR-1, MMP-9 and Snail staining
in HCC tissues and corresponding peritumoral
tissues were shown in Figure 6A. As expected,
VEGFR-1 was localized on the cell membrane;
Snail was localized in the nucleus, whereas the
MMP-9 was present in both cytoplasmic and
extracellular compartment. High expression of
VEGFR-1, Snail and MMP-9 was confirmed in
48.3, 53.3 and 45.3%, respectively, of all the cases.
High expression of VEGFR-1 was also associated
with a worse prognosis of HCC cases (p < 0.0001
for RFS and p = 0.023 for OS, respectively), and
there was also a trend toward a worse outcome
in patients with high expression of MMP-9
(p = 0.327 for RFS and p = 0.274 for OS, respec-
tively; Ta ble 1). HCC tissues with high expression
of VEGFR-1 also showed increased MMP-9 and
Snail expression with a Pearson correlation coef-
ficient of r = 0.232 (p = 0.036) and r = 0.418,
respectively (p < 0.001) (Figure 6B). To demon-
strate the prognostic significance of expression
pattern of VEGFR-1 and/or MMP-9 in HCC,
we divided the patients in four groups, I: high
expression of both VEGFR-1 and MMP-9, II:
low VEGFR-1 but high MMP-9 expression, III:
high VEGFR-1 but low MMP-9 expression, IV:
low expression of both VEGFR-1 and MMP-9,
and performed a Kaplan–Meier survival analy-
sis. The results showed that patients with high
expression of both VEGFR-1 and MMP-9 had
the worst prognosis when compared with other
groups (p = 0.008 for RFS and p = 0.128 for OS,
respectively) (Figure6C). Therefore, we divided
patients into groups I with high expression of both
VEGFR-1 and MMP-9 and group II without high
expression of both VEGFR-1 and MMP-9. Our
results showed that patients with high expression
of both VEGFR-1 and MMP-9 had a significantly
worse RFS and OS than the patients without high
expression of both VEGFR-1 and MMP-9 (p =
0.003 for RFS and p = 0.02 for OS, respectively)
(Figure 6C). Collectively, these results strongly sup-
port that induction of MMP-9 by VEGFR-1 acti-
vation contributes to HCC invasion and metasta-
sis, and it can be speculated a new model for caner
invasion (Figure 6D).
Discussion
In comparison with the well-established mecha-
nisms of EMT and angiogenesis, the impact of
VEGFR-1 mediated signaling on the expression
profile of proteolytic enzymes involved in inva-
siveness is not well understood. In this study, we
Future Oncol. (2015) 11(23)
3152
RESEaRch aRticlE Li, Zhu, Han, Ren, Liu & Qin
future science group
confirmed that VEGFR-1 activation in HCC
cell lines can regulate the expression of MMP-
9, which might be a novel mechanism for HCC
invasion.
HCC usually expresses high level of VEGF/
VEGFR compared with the peritumoral tis-
sue [16 ,3 0 –3 1] . Among VEGFRs, VEGFR-
1, expressed in cancer cells, is particularly
interesting due to its direct tumor activation
via an autocrine stimulatory pathway [4,7,3 2].
Previously studies have shown that overexpres-
sion of VEGF-B (a sole ligand for VEGFR-1)
in cancerous tissues is associated with tumor
invasion and poor prognosis in HCC. In addi-
tion, isoform VEGF-B167 appears to be the clini-
cally dominant isoform of VEGF-B [33] . Using
VEGF-B167 as the ligand to activate VEGFR-1,
we found that VEGFR-1 activation can sig-
nificantly promote cancer cell migratory and
invasive ability in vitro. These results provided
experimental evidence for the clinical signifi-
cance of VEGF-B in HCC prognosis. In addi-
tion, these results are also supported by the in
vivo results showing that high level of VEGFR-1
was associated with poor prognosis of HCC
patients, which was consistent with the finding
in renal cell cancer [34 ] . However, the clinical
value of VEGFR-1 warrants further evaluation
in larger clinical trials.
Recently, a monoclonal VEGFR-1 blocking
antibody IMC-18F1 was used to block the func-
tion of VEGFR-1 to confirm that the changes in
invasiveness were mediated through VEGFR-1,
but not the aberrant expression of another VEGF
tyrosine kinase receptor or due to the activation
of neuropilin-1 in several studies [31,35] . However,
VEGFR-B167 is thought to be the sole ligand for
VEGFR-1. There are no significant effects of the
nonspecific IgG on the changes of invasiveness.
These two aspects confirmed that the elevation in
migration and invasion of HCC cells was caused
by VEGFR-1 activation rather than unspecific
stimulation.
MMP-9, which is expressed in many types of
human carcinomas including HCC, has been
closely associated with tumor invasion and poor
prognosis [20,36 –37] . Recent studies reported that
MMP-9 was significantly upregulated by many
growth factors, leading to the invasion a vari-
ety of solid cancers in vitro [11– 13] . In present
study, VEGFR-1 activation-to-MMP-9 signal-
ing pathway was investigated and confirmed as
a novel mechanism of HCC invasion in vitro
and in vivo. Additional, these findings were
reinforced by the results of immunohistochem-
istry on TMA showing that VEGFR-1 is posi-
tively correlated with the MMP-9 expression
in cancerous tissues. Moreover, the results that
Figure 4. VEGFR-1 activation promotes MMP-9-dependet Matrigen invasion by SMMC7721 cells. (A) Matrigel invasion by SMMC7721
cells was examined in the absence or presence of a broad-spectrum MMP inhibitor GM6001 (12.5 μmol/l), bars indicate standard
deviation; *p < 0.005, versus nonspecic IgG group. (B) Matrigel invasion by SMMC7721 cells was examined in the presence or absence
of function-blocking MMP-9 antibody Ab-1 (15 μg/ml). Bars indicated standard deviation; **p < 0.05 versus VEGF-B167-treated group. The
similar results were obtained in three independent experiments.
70
60
50
40
30
20
10
0
70
60
50
40
30
20
10
0
IgG VEGF-B IgG VEGF-B
+GM6001
+Ab-1
+Ab-1
+GM6001
IgG VEGF-B IgG VEGF-B
Invaded cells/HPF (n)
# of invaded cells/HPF
***
3153
VEGFR-1 activation-induced MMP-9-dependent invasion in hepatocellular carcinoma RESEaRch aRticlE
future science group www.futuremedicine.com
patients with high coexpression of VEGFR-1
and MMP-9 exhibited the worst clinical out-
come also suggest that overexpression of both
VEGFR-1 and MMP-9 may play a synergistic
role in the progression of HCC and can serve
as a prognostic marker for HCC.
Snail, one of the key regulators of EMT, is
involved in the process of VEGFR-mediated
cancer cell invasion [4 ,29] . Correlative stud-
ies have shown Snail also can induce MMP-9
expression at the level of mRNA and protein
in vitro [1 2 ,23,38] . In this study, we found that
a distinct increase of Snail expression was
detected in the cells with activation of VEGFR-
1. Additionally, a strong positive correlation of
them was also observed in HCC tissue. These
results indicated that Snail may participate in the
regulation of VEGFR-1 activation-to-MMP-9
signaling pathway. VEGFR-1 activation led to
the enhanced expression of EMT regulators
including Snail, Slug and Twist in pancreatic
cancer cells. Among them, Twist appears to be
regulated by VEGFR-1 activation [4] . However,
the disadvantage of the present study was that
only one regulator was detected. Further stud-
ies are required to analyze the other regulators
expression in responding to the activation of
VEGFR-1 and elucidate the exact role in this
pa t hway.
Conclusion
In conclusion, we provide evidence for a new,
stepwise signaling pathway from activation of
VEGFR-1 to MMP-9 and demonstrate the criti-
cal role of MMP-9 in VEGFR-1-mediated HCC
cell invasion. Based on the findings of this study,
VEGFR-1 is present and functional in HCC
cells and its activation significantly enhances the
Figure 5. VEGFR-1 activation promotes Snail expression in hepatocellular carcinoma cell
lines. (A)Relative expression of Snail and β-actin was determined by quantitative real-time PCR,
normalized to nonspecic IgG group, and arbitrarily set at 1.0. Bars indicated standard deviation;
*p < 0.001 for both versus the zero time point, and the similar results were obtained in three
independent experiments. (B) The expression of Snail was detected by western blot. Data shown are
representative of three independent experiments.
10.0
9.0
8.0
7. 0
6.0
5.0
4.0
3.0
2.0
1. 0
0.0
01 24824 28 32 0124
824 28 32
2.5
2.2
2.0
1. 8
1. 5
1. 2
1. 0
0.8
0.5
0.2
0.0
SMMC7721
SMMC7721
HepG2
HepG2
Snail/β-actin
Snail/β-actin
Untreated
group IgG VEGF-B
β-actin
β-actin
Snail
Snail
VEGF-B167 treated
Non-specific IgG-
treated group
VEGF-B167 treated
Non-specific IgG-
treated group
**
Time (h) Time (h)
Future Oncol. (2015) 11(23)
3154
RESEaRch aRticlE Li, Zhu, Han, Ren, Liu & Qin
future science group
4.0
3.5
2.5
1. 5
1. 0
2.0
3.0
4.0
3.5
2.5
1. 5
1. 0
2.0
3.0
1. 01.5 2.52.0 3.0 3.5 3.5 4.0 4.5 5.0 6.05.5
MMP-9(log)value
VEGFR-1
VEGFR-1
Snail
VEGF-B
VEGFR-1 activation
MMP-9
Cancer invasion
Months after surgery
Months after surgery Months after surgery
Months after surgery
1. 0
0.8
0.6
0.4
0.2
0.0
1. 0
0.8
0.6
0.4
0.2
0.0
1. 0
0.8
0.6
0.4
0.2
0.0
1. 0
0.8
0.6
0.4
0.2
0.0
10 20 30 40 50 60 10 20 30 40 50 60
10 20 30 40 50 60
10 20 30 40 50 60
Recurrence-free survival (%)
Overall survival (%)
Recurrence-free survival (%)Overall survival (%)
I (n = 19)
II (n = 16)
III (n = 21)
IIV (n = 23)
I (n = 19)
II (n = 16)
III (n = 21)
IIV (n = 23)
Non-high coexpression
VEGFR-1/MM-9 (n = 60)
VEGFR-1
Case83 Case88
Hepatocellular carcinoma tissuesPeritumoral tissues
Case83
MMP-9
Snail
Non-high coexpression
VEGFR-1/MMP-9 (n = 60)
High coexpression
VEGFR-1/MM-9 (n = 19)
High coexpression
VEGFR-1/MMP-9 (n = 19)
+
+
++ ++
+
++
+
+
+
++
+
+
++
+
+
++
+
+
+
++
++
+
+++
++
++
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
+
3155
Figure 6. High coexpression of VEGFR-1/MMP-9 in hepatocellular carcinoma patients indicates
the worst outcome and a proposed model of VEGFR-1 activation promoting invasion via
induction of MMP-9 (see facing page). (A) Immunohistochemistry staining of VEGFR-1, MMP-9
and Snail in two representative patients with high coexpression of three proteins (Case83) and low
for three proteins (Case88). The black arrows indicated the expression of VEGFR-1; the blue arrows
indicated the expression of MMP-9; the red arrows indicated the expression of Snail. Scale bars,
50μm. (B) Comparison of the recurrence-free survival (upper left panel) and overal survival (lower
left panel) of patients categorized by VEGFR-1/MMP-9 immunohistochemistry result. Dierence of
recurrence-free survival (upper right panel) and overall survival (low right panel) in hepatocellular
carcinoma patients with or without high coexpression VEGFR-1/MMP-9. (C) Pearson correlation
test was performed to analyze the relationship between the VEGFR-1 and Snail (upper panel) and
between VEGFR-1 and MMP-9 (lower panel). (D) A proposed model of VEGFR-1 activation induced
invasion via MMP-9.
VEGFR-1 activation-induced MMP-9-dependent invasion in hepatocellular carcinoma RESEaRch aRticlE
future science group www.futuremedicine.com
invasiveness of cell lines through its regulation of
MMP-9. This study provided a novel pathway
associated with the progression of HCC induced
by VEGFR-1 activation.
Future perspective
It is significant to further confirm the role of
Snail in the novel pathway associated with
VEGFR-1 activation in the future. It is necessary
to verify whether VEGFR-1 could be a target for
the treatment of HCC invasion and metastasis.
Author contributions
T Li and C Qin conceived and designed the study, con-
ducted most of experiments and drafted the manuscript.
Y Zhu and L Han conducted some of the experiments,
supervised the data collection and analysis, interpreted
data and assisted in writing the manuscript. W Ren and
H Liu c onducted some of the experiments and interpreted
the data.
Financial & competing interests disclosure
This study was supported by National Nature Science
Foundation of China (numbers 81472685), Shandong
Outstanding Youth Science Fund (BS2013YY037). The
authors have no other relevant af filiations or financial
involvement with any organization or entity with a finan-
cial interest in or financial conflict with the subject matter
or materials discussed in the manuscript apart from those
disclosed.
No writing assistance was utilized in the production of
this manuscript.
Ethical conduct of research
The authors state that they have obtained appropriate
institutional review board approval or have followed the
principles outlined in the Declaration of Helsinki for all
human or animal experimental investigations. In addi-
tion, for investigations involving human subjects,
informed consent has been obtained from the participants
involved.
EXEcUtiVE SUMMaRY
Epithelial–mesenchymal transition and extracellular matrix degradation are critical for the initiation and progression of
tumor invasion.
VEGFR-1 activation can induce EMT by decreasing the expression of E-cadherin in hepatocellular carcinoma (HCC).
However, the impact of VEGFR-1 activation on the expression prole of proteolytic enzymes involved with HCC
invasion is unknown.
In this study, we found that activation of VEGFR-1 in HCC cell lines can signicantly increase the expression and
activity of MMP-9, and then promoted the cell invasion. Functional blockage of MMP-9 inhibited tumor cell invasion,
suggesting that VEGFR-1-induced cell invasion is dependent on the function of MMP-9.
Snail, an epithelial–mesenchymal transition regulator induced by VEGFR-1, was increased by VEGFR-1 activation,
indicating that Snail is involved in the regulation of the MMP-9 expression and cell invasion.
High coexpression of VEGFR-1 and MMP-9 in HCC, which is associated with the patient tumor invasion and metastasis,
is predictive of a worse clinical outcome.
These data showed a novel VEGFR-1 activation-to-MMP-9 signaling pathway that promotes HCC invasion.
Future Oncol. (2015) 11(23)
3156
RESEaRch aRticlE Li, Zhu, Han, Ren, Liu & Qin
future science group
References
Papers of special note have been highlighted as:
• of interest ; •• of considerable interest
1 Huo TI, Lin HC , Huang YH et al . The model
for end-stage liver disease-based Japan
Integrated Scoring system may have a better
predictive abilit y for patients with
hepatocellular carcinoma undergoing
locoregional therapy. Cancer 107(1), 141–148
(2006).
2 Miyoshi A, Kitajima Y, Kido S et al. Snail
accelerates cancer invasion by upregulating
MMP expression and is associated with poor
prognosis of hepatocellular carcinoma. Br. J.
Cancer 92(2), 252–258 (2005).
• HighlightsthattheupregulatedMMPs
expressioninducedbySnailcansignicantly
promotetheinvasionofhepatocellula r
carcinoma.
3 Sugimachi K, Tana ka S, K ameyama T et al.
Transcriptional repressor snail and progression
of human hepatocellular carcinoma.
Clin. Cancer Res. 9(7), 2657–2664 (2003).
•• Highlightsthatepithelial–mesenchy mal
transitionisacriticalmecha nismin
metastasisofhepatocellularcarcinoma.
4 Yang AD, Camp ER, Fan F et al. Va scu lar
endothelial grow th factor receptor-1
activation mediates epithelial to mesenchymal
transition in human pancreatic carcinoma
cells. Cancer Res. 66(1), 46 –51 (2006 ).
5 Schwartz JD, Rowinsky EK, Youssoufian H,
Pytowski B, Wu Y. Vascular endothelial
growth factor receptor-1 in human cancer:
concise review and rationale for development
of IMC-18F1 (Human antibody targeting
vascular endothelial growth factor receptor-1).
Cancer 116(4 S uppl. ), 1027–1032 (2010) .
•• Anexcellentreviewaboutthefunctionand
clinicalvalueofVEGFR-1inhuma ncancer.
6 Ng IO, Poon RT, Lee JM, Fan ST, Ng M, Tso
WK. Microvessel density, vascular endothelial
growth factor and its receptors Flt-1 and
Flk-1/KDR in hepatocellular carcinoma.
Am. J. Clin . Pathol . 116(6), 838–845 (2001).
7 Wu Y, Hooper AT, Zhong Z et al . The
vascular endothelial growth factor receptor
(VEGFR-1) supports grow th and survival of
human breast ca rcinoma. Int. J. Cancer
119 (7) , 1519 152 9 (20 06 ) .
8 Price DJ, Miralem T, Jiang S, Steinberg R,
Avraham H. Role of vascular endothelial
growth factor in the stimulation of cellular
invasion and signa ling of breast cancer cells.
Cell Growth Differ. 12(3), 129–135 (2001).
9 Bates RC, Goldsmith JD, Bachelder R E et al .
Flt-1-dependent survival characterizes the
epithelia l-mesenchymal transition of colonic
organoids. Curr. Biol. 13 (19 ), 1721 –1727
(2003).
10 Yi ZY, Feng LJ, Xiang Z et al. Va scul ar
endothelial grow th factor receptor-1
activation mediates epithelial to mesenchymal
transition in hepatocellular ca rcinoma cells.
J. Invest. Surg. 24(2), 67–76 (2011).
•• FirstpapertoindicatethatVEGFR-1
activationca ninduceepithelial
mesenchy maltransitioninhepatocellular
carcinomaandpromoteitsmetastasis.
11 Mira E, Mañes S, Lacalle RA, Márquez G,
Martínez-A C. Insulin-like growth factor
I-triggered cell migration and invasion are
mediated by matrix metalloproteinase-9.
Endocrinology 140 (4 ), 1657–166 4 (1999) .
12 Sun L, Diamond ME, Ottaviano AJ, Joseph
MJ, Ananthanarayan V, Munshi HG.
Transforming growth factor-beta 1 promotes
matrix metalloproteinase-9-mediated ora l
cancer invasion through snail expression. Mol.
Cancer Res. 6(1), 10–20 (2008).
13 Zuo JH, Zhu W, Li MY et al. Activation of
EGFR promotes squamous carcinoma
SCC10A cell migration and invasion via
inducing EMT-like phenoty pe cha nge and
MMP-9-mediated degradation of E-cadherin.
J. Cell Biochem. 112(9), 2508–2517 (2011).
14 Der yugina EI, Quigley JP. Matrix
metalloproteinases and tumor metasta sis.
Cancer Metastasis Rev. 25(1), 9 –34 (2006 ).
15 Egeblad M, Werb Z. New functions for the
matrix metalloproteinases in c ancer
progression. Nat. Rev. Cancer 2 (3) , 161–174
(2002).
16 Stamenkovic I. Matrix metalloproteinases in
tumor invasion and metastasis. Semin. Cancer
Biol. 10(6), 415–433 (2000 ).
17 Bergers G, Brekken R, McMahon G et al.
Matrix metalloproteina se-9 triggers the
angiogenic switch during carcinogenesis. Nat.
Cell Biol . 2(10), 737–744 (2000).
18 Hiratsuka S, Nak amura K, Iwai S et al.
MMP9 induction by vascular endothelial
growth factor receptor-1 is involved in
lung-specific metastasis. Cancer Cell 2(4),
289–300 (2002).
•• HighlightsthatMMP-9expressioninduced
byVEGFR-1isinvolvedinca ncermetastasis.
19 Zhang S, Li J, Jiang Y, Xu Y, Qin C.
Programmed cell death 4 (PDCD4)
suppresses metastastic potentia l of human
hepatocellular carcinoma cells. J. Exp. Clin.
Cancer Res. 28, 71(2009).
20 Chen R, Cui J, Xu C et al. The significance of
MMP-9 Over MMP-2 in HCC invasiveness
and recurrence of hepatocellular carcinoma
after curative resection. Ann. Surg. Oncol.
19(Suppl. 3), S375–S384 (2012).
•• HighlightsthatMMP-9ratherthanMMP-2
expressioniscloselyinvolvedwith
hepatocellularcarcinomainvasion.
21 Yung S, Davies M. Response of the human
peritoneal mesothelial cell to injury: an
in vitro model of peritonea l wound hea ling.
Kidney Int. 5 4 (6) , 216 0 –2169 (1998).
22 Ottaviano AJ, Sun L , Ananthanarayanan V,
Munshi HG. Extracellular matrix-mediated
membrane-type 1 matrix metalloproteina se
expression in pancreatic ductal cells is
regulated by transforming growth factor-
bet a1. Cancer Res. 66 (14), 7032 –7040 (2006 ).
23 Jordà M, Olmeda D, Vinyals A et al.
Upregulation of MMP-9 in MDCK epithelial
cell line in response to expression of the Snail
transcription factor. J. Cell Sci. 118(Pt15 ),
3371–3385 (2005).
24 Xu Y, Zhu M, Zhang S, Liu H, Li T, Qin C.
Expression and prognostic value of PRL-3 in
human intrahepatic cholangiocarcinoma.
Pathol. Oncol. Res. 16(2), 169–175 (2010).
25 Toi M, Bando H, Ogawa T, Muta M, Hornig
C, Weich HA. Significance of vascular
endothelial grow th factor (VEGF)/soluble
VEGF receptor-1 relationship in breast
cancer. Int. J. Cancer 98(1), 14–18 (2002) .
26 Tang J, Cui J, Chen R et al. A three-
dimensional cell biology model of human
hepatocellular carcinoma in vitro. Tum our
Biol. 32(3), 469–479 (2011).
27 Fukase K, Ohtsuka H, Onogawa T et al. Bile
acids repress E-cadherin through the
induction of Snail and increase cancer
invasiveness in human hepatobiliary
carcinoma. Cancer Sci. 99 (9) , 17 85 –1792
(2008).
28 Hipp S, Walch A, Schuster T et al. Activation
of epidermal growth factor receptor results in
snail protein but not mR NA overexpression in
endometrial cancer. J. Cell Mol . Med. 13(9B),
3858–3867 (2009).
29 Wanami LS, Chen HY, Peiró S, García de
Herreros A, Bachelder R E. Vascular
endothelial grow th factor-A stimulates Snail
expression in breast tumor cells: implications
for tumor progression. Exp. Cell Res. 314 (13),
2448–2453 (2008).
30 Amaoka N, Saio M, Nonaka K et al.
Expression of vascular endothelial growth
factor receptors is closely related to the
histological grade of hepatocellular
carcinoma. Oncol. Rep. 16(1), 3–10 (2006).
•• HighlightsthatV EGFR-1wasexpressedin
thehepatocellu larcarcinomatissueandwas
3157
VEGFR-1 activation-induced MMP-9-dependent invasion in hepatocellular carcinoma RESEaRch aRticlE
future science group www.futuremedicine.com
VEGFR-1 activation-induced MMP-9-dependent invasion in hepatocellular carcinoma RESEaRch aRticlE
conrmedasafunctionalreceptorassociated
withhistologicalgrade.
31 Jia JB, Zhuang PY, Sun HC et al. Protein
expression profiling of vascular endothelial
growth factor and its receptors identifies
subclasses of hepatocellular carcinoma and
predicts survival. J. Cancer Res. Clin. Oncol.
135(6), 847–854 (2009).
32 Wey JS, Fan F, Gray MJ et al. Vasc ula r
endothelial grow th factor receptor-1 promotes
migration and inva sion in pancreatic carcinoma
cell lines. Cancer 104(2), 427–438 (2005).
33 Kanda M, Nomoto S, Nishikawa Y et al.
Correlations of the expression of vascular
endothelial grow th factor B and its isoforms
in hepatocellula r carcinoma with clinico-
pathological parameters. J. Surg. Oncol. 98(3),
190 –196 (2 0 08) .
34 Laird A, O’Mahony FC, Nanda J et al.
Differential expression of prognostic
proteomic markers in primar y tumour, venous
tumour thrombus a nd metastatic renal cell
cancer tissue a nd correlation with patient
outcome. PLoS ONE 8(4), e60483 (2013).
•• HighlightsthatV EGFR-1couldserveasa
prognosticmarkerforothert ypeof
ma lig nanc y.
35 Fan F, Wey JS, McCarty MF et al. Expression
and function of vascular endothelial growth
factor receptor-1 on human colorectal cancer
cells. Oncogene 24(16), 2647–2653 (2005).
36 Libra M, Scalisi A, Vella N et al. Uterine
cervical carcinoma: role of matrix
metalloproteinases (review). Int. J. Oncol .
34(4), 897–903 (2009).
37 Köhrmann A, Kammerer U, Kapp M et al .
Expression of matrix metalloproteinases
(MMPs) in primary human breast cancer and
breast cancer cell lines: New findings and
review of t he literature. BMC Cancer 9, 188
(2009).
38 Lin CY, Tsai PH, Kandaswami CC et al.
Matrix metalloproteina se-9 cooperates with
transcription factor Snail to induce
epithelial-mesenchymal transition. Cancer
Sci. 102(4), 815–827 (2011).
... Sachie et al. [56] found that the activation of FLT1 could induce macrophages to secrete MMP-9, which signi cantly promoted lung metastasis. Li et al. [57] suggested that the activation of FLT1 in HCC cells could induce the secretion of MMP9 into the extracellular matrix (ECM), thereby promoting tumor invasion and metastasis. These studies might better explain the current study results; the VEGFA/FLT1 ligand-receptor interactions activated TAM_ SPP1-derived MMP9 secretion, thereby promoting tumor angiogenesis. ...
Preprint
Full-text available
Purpose Cytokeratin 19-positive cancer stem cells (CK9 + CSCs) and their tumor-associated macrophages (TAMs) have not been fully explored yet in the hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC). Experimental Design: Single-cell RNA sequencing was performed on the viable cells obtained from 11 treatment-naïve HBV-associated HCC patients, including 8 CK19 + patients, to elucidate their transcriptomic landscape, CK19 + CSC heterogeneity, and immune microenvironment. Two in-house primary HCC cohorts (96 cases-related HBV and 89 cases with recurrence), multiple external cohorts, and in vitro and in vivo experiments were used to validate the results. Results A total of 64,581 single cells derived from the human HCC and adjacent normal tissues were sequenced, and 11 cell types were identified. The result showed that CK19 + CSCs were phenotypically and transcriptionally heterogeneous, co-expressed multiple hepatics CSC markers, and were positively correlated with worse prognosis. Moreover, the SPP1 + TAMs (TAM_SPP1) with strong M2-like features and worse prognosis were specifically enriched in the CK19 + HCC and promoted tumor invasion and metastasis by activating angiogenesis. Importantly, matrix metalloproteinase 9 (MMP9) derived from TAM_SPP1, as the hub gene of CK19 + HCC, was activated by the VEGFA signal. The patients with low TAM_SPP1 enrichment might benefit from trans-arterial chemoembolization. Conclusions This study revealed the heterogeneity and stemness characteristics of CK19 + CSCs and specific immunosuppressive TAM_SPP1 in CK19 + HCC. The VEGFA signal can activate TAM_SPP1-derived MMP9 to promote the invasion and metastasis of CK19 + HCC tumors. This might provide novel insights into the clinical treatment of HCC patients.
... Activation of VEGFR-1 has been shown to be involved in epithelial-mesenchymal transition (EMT), which confers on cancer cells a more invasive phenotype, and to promote metalloprotease activation [11]. Interestingly, PlGF is frequently expressed by melanoma cell lines derived from metastatic lesions compared to those obtained from primary tumors [12]. ...
Article
Full-text available
Placenta growth factor (PlGF) is a member of the vascular endothelial growth factor (VEGF) family involved in tumor-associated angiogenesis and melanoma invasion of the extra-cellular matrix (ECM) through activation of membrane VEGF receptor 1 (VEGFR-1). A soluble VEGFR-1 (sVEGFR-1) form is released in the ECM, where it sequesters proangiogenic factors and stimulates endothelial or tumor cell adhesion and chemotaxis through interaction with α5β1 integrin. The anti-VEGFR-1 monoclonal antibody (D16F7 mAb) inhibits VEGF-A or PlGF-mediated signal transduction without affecting ligand interaction, thus preserving sVEGFR-1 decoy function. The aim of this study was to investigate whether D16F7 mAb hampers melanoma spread by in vitro analysis of cell adhesion to sVEGFR-1, ECM invasion, transmigration through an endothelial cell monolayer and in vivo evaluation of tumor infiltrative potential in a syngeneic murine model. Results indicate that D16F7 mAb significantly inhibits melanoma adhesion to sVEGFR-1 and ECM invasion, as well as transmigration in response to PlGF. Moreover, treatment of melanoma-bearing mice with the anti-VEGFR-1 mAb not only inhibits tumor growth but also induces a significant reduction in bone infiltration associated with a decrease in PlGF-positive melanoma cells. Furthermore, D16F7 mAb reduces PlGF production by melanoma cells. Therefore, blockade of PLGF/VEGFR-1 signaling represents a suitable strategy to counteract the metastatic potential of melanoma.
... Increased levels of circulating VEGF-A in the later phases of ischemic stroke are probably due to the activation of angiogenesis [33]. Alternatively, stroke-induced vascular damage in the blood-brain barrier leads to increased VEGF expression in the ischemic penumbra zone, from where it can enter the bloodstream when the blood-brain barrier is damaged [46,47]. ...
Article
Full-text available
Vascular endothelial growth factors (VEGFs) are important regulators of angiogenesis, neuroprotection, and neurogenesis. Studies have indicated the association of VEGF dysregulation with the development of neurodegenerative and cerebrovascular diseases. We studied the changes in serum levels of VEGF-A, VEGFR-1, and VEGFR-2 in patients at various phases of ischemic and hemorrhagic strokes. Quantitative assessment of VEGF-A, VEGFR-1, and VEGFR-2 in serum of patients with hemorrhagic or ischemic stroke was performed by enzyme immunoassay in the hyper-acute (1–24 h from the onset), acute (up to 1–7 days), and early subacute (7 days to 3 months) phases of stroke, and then compared with the control group and each other. Results of our retrospective study demonstrated different levels of VEGF-A and its receptors at various phases of ischemic and hemorrhagic strokes. In ischemic stroke, increased VEGFR-2 level was found in the hyper-acute (p = 0.045) and acute phases (p = 0.024), while elevated VEGF-A and reduced VEGFR-1 levels were revealed in the early subacute phase (p = 0.048 and p = 0.012, respectively). In hemorrhagic stroke, no significant changes in levels of VEGF-A and its receptors were identified in the hyper-acute phase. In the acute and early subacute phases there was an increase in levels of VEGF-A (p < 0.001 and p = 0.006, respectively) and VEGFR-2 (p < 0.001 and p = 0.012, respectively). Serum levels of VEGF-A and its receptors in patients with hemorrhagic and ischemic stroke indicate different pathogenic pathways depending on the phase of the disease.
... They belong to the same membrane protein family which can catalyze transferring of the γ phosphate of ATP to hydroxyl groups of tyrosinases on target proteins and play important roles in the control of most fundamental cellular processes including cell cycle, migration, metabolism, proliferation, differentiation and survival [14]. We have confirmed activation of VEGFR-1, one of the RTKs which leads to tumor angiogenesis and induces MMP-9dependent invasion in HCC [15]. Some RTKs namely EGFR/MET can participate in metastasis of tumors by forming a molecular complex with integrin, mutation, amplification, and drug resistance [16][17][18]. ...
Article
Full-text available
The receptor tyrosine kinases (RTKs) family is well-recognized as vital targets for the treatment of hepatocarcinoma cancer (HCC) clinically, whereas the survival benefit of target therapy sorafenib is not satisfactory for liver cancer patients due to metastasis. EGFR and MET are two molecules of the RTK family that were related to the survival time of liver cancer patients and resistance to targeted therapy in clinical reports. However, the mechanism and clinical therapeutic value of EGFR/MET in HCC metastasis are still not completely clarified. The study confirmed that EGFR/MET was highly expressed in HCC cells and tissues and the phosphorylation was stable after metastasis. The expression of EGFR/MET was up-regulated in circulating tumor microemboli (CTM) to accelerate IL-8 production and resistance to the lethal effect of leukocytes. Meanwhile, highly expressed EGFR/MET effectively regulated the Ras/MAPK pathway and stabilized suspended HCC cells by facilitating proliferation and inhibiting apoptosis. Moreover, EGFR/MET promoted phosphorylation of hetero-RTKs, which was dependent on high-energy phosphoric acid compounds rather than their direct interactions. In conclusion, highly expressed EGFR/MET could be used in CTM identification and suitable for preventing metastasis of HCC in clinical practice.
... Matrix metalloproteinases (MMPs) are a family of zincdependent proteases that are engaged in ECM degradation and remolding. 5 MMPs perform considerable roles in tumor progression, 6 including HCC. [7][8][9] MMP-21 is a newly identified member of the MMP family. As in other MMPs, it is synthesized as an latent proenzyme and activated as an proteolytic product in the extracellular secretion pathway. ...
Article
Full-text available
MMP-21 is a newly-identified member of Matrix metalloproteinases family and has been reported to regulate both embryonic development and tumor progression. However, roles of MMP-21 in HCC remains largely unclear. In this study, we used western blot, qPCR and IHC to determine the upregulation of MMP-21 in HCC tissues, and showed that the increase of MMP-21 was associated with vascular invasion and poor prognosis. Although changing levels of MMP-21 in HCC cell lines had no significant effects on cell migration or invasion abilities in in vitro transwell tests, both IHC analysis and in vivo mouse model proved that upregulated MMP-21 promoted metastasis. Functional enrichments of MMP-21 using TCGA data suggested that MMP-21 might regulate metastasis via macrophages. Further experiments proved that MMP-21 enhanced macrophage recruitment via increasing CCL-14 levels and promoted M2-type polarization of macrophage via elevating the expression of CSF-1 and FGF-1. Taken together, this study revealed that MMP-21 controlled the tumor microenvironment remodeling and functional regulation of macrophages to regulate HCC metastasis.
... Moreover, studies have demonstrated the functional interplay between MMP-9 and vascular endothelial growth factor (VEGF), another EMT inducer, in HCC, favoring tumor angiogenesis [12]. This can somewhat be explained by studies that demonstrated a positive correlation of MMP-9 and VEGF expression with the progression and recurrence of HCC [57,58]. Angiogenesis has an important role in HCC progression and aggressiveness, being a part of its multifaceted molecular pathogenesis [13]. ...
Article
Full-text available
Hepatocellular carcinoma (HCC) is one of the most burdened tumors worldwide, with a complex and multifactorial pathogenesis. Current treatment approaches involve different molecular targets. Phytochemicals have shown considerable promise in the prevention and treatment of HCC. We investigated the efficacy of two natural components, 1,8 cineole (Cin) and ellagic acid (EA), against diethylnitrosamine/2-acetylaminofluorene (DEN/2-AAF) induced HCC in rats. DEN/2-AAF showed deterioration of hepatic cells with an impaired functional capacity of the liver. In addition, the levels of tumor markers including alpha-fetoprotein, arginase-1, alpha-L-fucosidase, and ferritin were significantly increased, whereas the hepatic miR-122 level was significantly decreased in induced-HCC rats. Interestingly, treatment with Cin (100mg/kg) and EA (60mg/kg) powerfully restored these biochemical alterations. Moreover, Cin and EA treatment exhibited significant downregulation in transforming growth factor beta-1 (TGF-β1), Fascin-1 (FSCN1), vascular endothelial growth factor (VEGF), matrix metalloproteinase-9 (MMP-9), and epithelial-mesenchymal transition (EMT) key marker, vimentin, along with a restoration of histopathological findings compared to HCC group. Such effects were comparable to Doxorubicin (DOX) (2mg/kg); however, a little additive effect was evident through combining these phytochemicals with DOX. Altogether, this study highlighted 1,8 cineole and ellagic acid for the first time as promising phytochemicals for the treatment of hepatocarcinogenesis via regulating multiple targets.
... Endothelial cells express the VEGF receptor (VEGFR), which once activated by the ligand, initiates a transduction cascade, leading to the production of matrix metalloproteases (MMP) [152,153]. Disruption of the matrix allows for more space for endothelial cells to divide and organize into a mature network of new vessels. ...
Article
Full-text available
Extracellular vesicles (EVs) are heterogenous membrane-encapsulated vesicles secreted by every cell into the extracellular environment. EVs carry bioactive molecules, including proteins, lipids, DNA, and different RNA forms, which can be internalized by recipient cells, thus altering their biological characteristics. Given that EVs are commonly found in most body fluids, they have been widely described as mediators of communication in several physiological and pathological processes, including cancer. Moreover, their easy detection in biofluids makes them potentially useful candidates as tumor biomarkers. In this manuscript, we review the current knowledge regarding EVs and non-coding RNAs and their role as drivers of the metastatic process in lung cancer. Furthermore, we present the most recent applications for EVs and non-coding RNAs as cancer therapeutics and their relevance as clinical biomarkers.
Article
Full-text available
Innate and adaptive immune cells patrol and survey throughout the human body and sometimes reside in the tumor microenvironment (TME) with a variety of cell types and nutrients that may differ from those in which they developed. The metabolic pathways and metabolites of immune cells are rooted in cell physiology, and not only provide nutrients and energy for cell growth and survival but also influencing cell differentiation and effector functions. Nowadays, there is a growing awareness that metabolic processes occurring in cancer cells can affect immune cell function and lead to tumor immune evasion and angiogenesis. In order to safely treat cancer patients and prevent immune checkpoint blockade-induced toxicities and autoimmunity, we suggest using anti-angiogenic drugs solely or combined with Immune checkpoint blockers (ICBs) to boost the safety and effectiveness of cancer therapy. As a consequence, there is significant and escalating attention to discovering techniques that target metabolism as a new method of cancer therapy. In this review, a summary of immune-metabolic processes and their potential role in the stimulation of intracellular signaling in TME cells that lead to tumor angiogenesis, and therapeutic applications is provided. Video abstract.
Article
Full-text available
Objective: High altitude heart disease (HAHD) is a common pediatric disease in high altitude areas. It usually occurs in people who have lived for a long time or have lived for more than 2500m above sea level. Its common inducement is respiratory tract infection. The clinical differential diagnosis is difficult because the symptoms of HAHD are similar to those of congenital heart disease; Due to the limitation of medical conditions, many patients are in the state of losing follow-up or not seeking medical treatment, resulting in poor prognosis of HAHD and becoming a high-altitude disease with high mortality. Clarifying the molecular mechanism of HAHD, developing early molecular screening technology and accurate treatment methods of HAHD are the key to improve the ability of prevention and treatment of HAHD. Methods: First, the literature in the PubMed and CNKI databases were screened based on keywords and abstracts. Then, the literature for the study was identified based on the fitness between the content of the literature, the research objectives, and the timeliness of the literature. Finally, a systematic molecular mechanism of HAHD was established by investigating the literature and sorting out the genetic adaptations of Tibetan populations compared with low-altitude populations that migrated to the plateau. Results: With the investigation of the 48 papers screened, it was found that genes capable of enhancing the hypoxic ventilatory response and resistance to pulmonary hypertension were all correlated with the hypoxia-inducible factor (HIF) pathway, consisting mainly of three pathways, HIF-1α, HIF-2α, and NO. Conclusion: The low prevalence of HAHD in Tibetan aboriginal children was mainly due to the genetic adaptation of the Tibetan population to the high altitude environment, which coordinated the cellular response to hypoxia by regulating the downstream hypoxia control genes in the HIF pathway.
Article
Breast cancer is the most common malignant tumor in women and the leading cause of cancer death in women. About 30% of breast cancer patients have metastasis every year, which greatly increases the mortality rate of breast cancer. The main target organs for metastasis are bone, brain, liver and lung. The breast cancer liver metastasis (BCLM) mechanism is not fully clarified. This is a complex process involving multiple factors, which is not only related to the microenvironment of the primary tumor and liver, but also regulated by a variety of signaling pathways. Clarifying these mechanisms is of great help to guide clinical treatment. With the in-depth study of BCLM, a variety of new treatment schemes such as targeted therapy and endocrine therapy provide new ideas for the cure of BCLM. In this review, we will summarize the molecular mechanism and treatment of BCLM.
Article
Full-text available
Renal cell carcinoma (RCC) is the most deadly of urological malignancies. Metastatic disease affects one third of patients at diagnosis with a further third developing metastatic disease after extirpative surgery. Heterogeneity in the clinical course ensures predicting metastasis is notoriously difficult, despite the routine use of prognostic clinico-pathological parameters in risk stratification. With greater understanding of pathways involved in disease pathogenesis, a number of biomarkers have been shown to have prognostic significance, including Ki67, p53, vascular endothelial growth factor receptor 1 (VEGFR1) and ligand D (VEGFD), SNAIL and SLUG. Previous pathway analysis has been from study of the primary tumour, with little attention to the metastatic tumours which are the focus of targeted molecular therapies. As such, in this study a tissue microarray from 177 patients with primary renal tumour, renal vein tumour thrombus and/or RCC metastasis has been created and used with Automated Quantitative Analysis (AQUA) of immunofluorescence to study the prognostic significance of these markers in locally advanced and metastatic disease. Furthermore, this has allowed assessment of differential protein expression between the primary tumours, renal vein tumour thrombi and metastases. The results demonstrate that clinico-pathological parameters remain the most significant predictors of cancer specific survival; however, high VEGFR1 or VEGFD can predict poor cancer specific survival on univariate analysis for locally advanced and metastatic disease. There was significantly greater expression of Ki67, p53, VEGFR1, SLUG and SNAIL in the metastases compared with the primary tumours and renal vein tumour thrombi. With the exception of p53, these differences in protein expression have not been shown previously in RCC. This confirms the importance of proliferation, angiogenesis and epithelial to mesenchymal transition in the pathogenesis and metastasis of RCC. Importantly, this work highlights the need for further pathway analysis of metastatic tumours for overcoming drug resistance and developing new therapies.
Article
MCF-7 cells migrate through vitronectin-coated filters in response to insulin-like growth factor I (IGF-I); migration is inhibited by the matrix metalloproteinase (MMP) inhibitor BB-94, but not by the serine proteinase inhibitor aprotinin. MMP-9 was identified in the conditioned medium of MCF-7 cells; in addition, fluorescence-activated cell sorting analysis revealed its presence on the cell surface, where MMP-9 activity was also found using a specific fluorogenic peptide. Furthermore, the messenger RNA encoding MMP-9 was detected in MCF-7 cells by PCR. The IGF-I concentration leading to maximal MCF-7 invasion produces an increase in cell surface proteolytic activity after short incubation periods. At 18 h, however, preincubation of MCF-7 cells with IGF-I produces at 18 h a dose-dependent decrease in cell-associated MMP-9 activity and an increase in soluble MMP-9. MCF-7 invasion is dependent on the αvβ5 integrin, a vitronectin receptor. The levels of αv- andβ 5-subunits expressed in MCF-7 cells depend on the IGF-I concentration, which triggers an increase in both of these subunits. Based on these results, we suggest that IGF-I-induced MCF-7 cell migration is mediated by the MMP-9 activity on the cell surface and byα vβ5 integrin.
Article
The human vascular endothelial growth factor receptor-1 (VEGFR-1, or Flt-1) is widely expressed in normal and pathologic tissue and contributes to the pathogenesis of both neoplastic and inflammatory diseases. In human cancer, VEGFR-1 mediated signaling is responsible for both direct tumor activation and angiogenesis. VEGFR-1 mediated activation of nonmalignant supporting cells, particularly stromal, dendritic, hematopoietic cells, and macrophages, is also likely important for cancer pathogenesis. VEGFR-1 is also hypothesized to enable the development of cancer metastases by means of activation and premetastatic localization in distant organs of bone marrow-derived hematopoietic progenitor cells, which express VEGFR-1. IMC-18F1 is a fully human IgG1 antibody that binds to VEGFR-1 and has been associated with the inhibition of cancer growth in multiple in vitro and human tumor xenograft models. The preliminary results of phase 1 investigations have also indicated a favorable safety profile for IMC-18F1 at doses that confer antibody concentrations that are associated with relevant antitumor activity in preclinical models. Cancer 2010;116(4 suppl):1027–32. © 2010 American Cancer Society.
Article
Cytokine gene transfer is a current approach in studies of gene therapy of cancer. IFNs represent valuable cytokines for these studies, since they exert multiple biological effects, including anti-tumor activities. Early studies have been focused on IFN- γ. Recently, several reports have shown that the transfer of type I IFN (especially IFN- α) genes represents a powerful approach for inducing tumor suppression. Recent studies have underscored new IFN-induced activities on immune cells. This knowledge adds a further rationale for the use of IFN- α in strategies of gene therapy of cancer and can be exploited for the design of more selective and effective anticancer treatments.
Article
The human vascular endothelial growth factor receptor-1 (VEGFR-1, or Flt-1) is widely expressed in normal and pathologic tissue and contributes to the pathogenesis of both neoplastic and inflammatory diseases. In human cancer, VEGFR-1 mediated signaling is responsible for both direct tumor activation and angiogenesis. VEGFR-1 mediated activation of nonmalignant supporting cells, particularly stromal, dendritic, hematopoietic cells, and macrophages, is also likely important for cancer pathogenesis. VEGFR-1 is also hypothesized to enable the development of cancer metastases by means of activation and premetastatic localization in distant organs of bone marrow-derived hematopoietic progenitor cells, which express VEGFR-1. IMC-18F1 is a fully human IgG(1) antibody that binds to VEGFR-1 and has been associated with the inhibition of cancer growth in multiple in vitro and human tumor xenograft models. The preliminary results of phase 1 investigations have also indicated a favorable safety profile for IMC-18F1 at doses that confer antibody concentrations that are associated with relevant antitumor activity in preclinical models.
Article
The extracellular matrix metalloproteases MMP-9 and MMP-2 are critical for the invasive potential of tumors. However, it is not clear which of the two plays the predominant role in tumor invasion and progression. In the present study, we compared the clinical efficacy of MMP-9 and MMP-2 overexpression for predicting tumor recurrence and survival after surgical resection in HCC patients. MMP-9 and MMP-2 expression in HCC cell lines and in vitro HCC invasion model were detected by quantitative RT-PCR and immunofluorescence. The expression levels of MMP-9 and MMP-2 were assessed by immunohistochemistry in HCC tissue microarrays from HCC patients (study set) who underwent curative resection. The clinicopathological data were retrospectively analyzed. The results were further verified in an independent cohort of 92 HCC patients (validation set). Univariate analysis demonstrated that high expression of MMP-9 was associated with both time to recurrence (TTR, P = .015) and overall survival (OS, P = .024), whereas high expression of MMP-2 was only correlated with TTR (P = .041). Multivariate analysis confirmed that MMP-9 expression was an independent predictor of TTR and OS. The coindex of MMP-9 and preoperative serum AFP levels was significantly correlated with TTR and OS (P = .036 and P = .040), but the coindex of MMP-2 and AFP did not show prognostic significance for either TTR or OS (P = .067 and P = .053). The prognostic value of MMP-9 overexpression was validated in the independent data set. MMP-9 is superior to MMP-2 for the prediction of tumor recurrence and survival in HCC patients after surgical resection.
Article
EGFR is a potent stimulator of invasion and metastasis in head and neck squamous cell carcinomas (HNSCC). However, the mechanism by which EGFR may stimulate tumor cell invasion and metastasis still need to be elucidated. In this study, we showed that activation of EGFR by EGF in HNSCC cell line SCC10A enhanced cell migration and invasion, and induced loss of epitheloid phenotype in parallel with downregulation of E-cadherin and upregulation of N-cadherin and vimentin, indicating that EGFR promoted SCC10A cell migration and invasion possibly by an epithelial to mesenchymal transition (EMT)-like phenotype change. Interestingly, activation of EGFR by EGF induced production of matrix metalloproteinase-9 (MMP-9) and soluble E-cadherin (sE-cad), and knockdown of MMP-9 by siRNA inhibited sE-cad production induced by EGF in SCC10A. Moreover, both MMP-9 knockdown and E-cadherin overexpression inhibited cell migration and invasion induced by EGF in SCC10A. The results indicate that EGFR activation promoted cell migration and invasion through inducing MMP-9-mediated degradation of E-cadherin into sE-cad. Pharmacologic inhibition of EGFR, MEK, and PI3K kinase activity in SCC10A reduced phosphorylated levels of ERK-1/2 and AKT, production of MMP-9 and sE-cad, cell migration and invasion, and expressional changes of EMT markers (E-cadherin and N-cadherin) induced by EGF, indicating that EGFR activation promotes cell migration and invasion via ERK-1/2 and PI3K-regulated MMP-9/E-cadherin signaling pathways. Taken together, the data suggest that EGFR activation promotes HNSCC SCC10A cell migration and invasion by inducing EMT-like phenotype change and MMP-9-mediated degradation of E-cadherin into sE-cad related to activation of ERK-1/2 and PI3K signaling pathways.
Article
To explore the molecular mechanism of Vascular endothelial growth factor receptor-1 (VEGFR-1) in invasion and metastasis of hepatocellular carcinoma. Reverse transcription polymerase chain reaction was performed to test expression of VEGFR-1 and its ligand VEGF-B19 in four hepatoma carcinoma cell. Fluorescent immunohistochemistry and western blotting were used to test the change of expression of E-cadherin or α-catenin. VEGF-B-treated cells exhibited a change in E-cadherin from an organized, membrane-bound structure to a disorganized state that was dispersed throughout the cytoplasm. The maximal changes in E-cadherin were observed 24 hr after treatment of cells with VEGF-B. α-catenin was observed to translocate to the nucleus from its usual membrane-bound location 24 hr after treatment with either VEGF-B. Expression of the epithelial adhesion molecules E-cadherin was observed to decrease 48 hours after VEGF-B treatment. The nuclear expression of α-catenin was observed to increase 24 hr after treatment with VEGF-B. VEGFR-1 on tumor cells may contribute to the aggressive behavior of hepatocellular carcinoma cells by inducing epithelial to mesenchymal transition (EMT). Targeting VEGFR-1 and downstream mediators of EMT may provide the foundation for the development of novel therapeutic approaches for this morbid and lethal disease.