Purpose: Angiogenic behavior is a critical aspect of cancer biology and subject to regulation by multiple molecular pathways. Because the signal transducer and activator of transcription 3 (Stat3) transcription factor regulates multiple genes important to angiogenesis, we sought to determine whether Stat3 expression is related to vascular endothelial growth factor (VEGF) expression and microvessel density (MVD) in gastric cancer and whether these factors predict survival in gastric cancer patients.

Experimental Design: The expression of Stat3 and VEGF was determined by immunohistochemistry using archival tissues from 86 cases of resected human gastric cancer and confirmed by Western blot analysis. Angiogenic phenotype was determined by CD34 staining and microvessel counting.

Results: Stat3 expression correlated with VEGF expression and MVD. In univariate survival analyses, Stat3 expression (P = 0.013) and MVD (P = 0.036) were associated with inferior survival. However, when Stat3 expression, VEGF expression, MVD, stage, completeness of resection, Lauren's histologic classification, and age were entered into a Cox proportional hazards model, only strong Stat3 expression (P = 0.049) and advanced stage (P < 0.01) were independently prognostic of poor survival. Furthermore, genetically enforced alterations of activated Stat3 expression led to altered VEGF expression and angiogenic potential in human gastric cancer cells.

Conclusion: Dysregulated Stat3 activation may play an important role in VEGF overexpression and elevated angiogenic phenotype in gastric cancer and contribute to gastric cancer development and progression.

Gastric cancer remains the second most frequently diagnosed malignancy worldwide (1), and its aggressive nature is related to mutations of various oncogenes and tumor suppressor genes (2–5) and abnormalities in several growth factors and their receptors (3, 4). These abnormalities affect the downstream signal transduction pathways involved in the control of many aspects of cancer biology, including angiogenesis. Specifically, these perturbations may confer a phenotype of increased angiogenesis to gastric cancer cells (3–5). Previous studies indicated involvement of several angiogenic factors in gastric cancer angiogenesis, including interleukin-8, epidermal growth factor, fibroblast growth factor, insulin-like growth factor, matrix metalloproteinase-2, transforming growth factor, and vascular endothelial growth factor (VEGF; refs. 6–10). Previous studies also showed that microvessel density (MVD) and various angiogenic factors related to tumor development and progression are predictive of patient survival in gastric cancer (3, 11–15). However, the molecular mechanisms behind the abnormal expression of many of these angiogenic factors in gastric cancer, including VEGF, remain unclear.

Increasing evidence suggests that VEGF expression is regulated by various hormones and growth factors, including epidermal growth factor and fibroblast growth factor (5), and by oncogenic proteins such as Src and Ras (16, 17). Significantly, many tumors often exhibit overexpression of these growth factors and oncogenic proteins, and these molecules often transmit signals through the signal transducer and activator of transcription 3 (Stat3), a member of the Janus-activated kinase/STAT signaling pathway (16, 17). Therefore, it is highly probable that Stat3 activation is a common signaling intermediate leading to the overexpression of VEGF in malignant tumors. This notion is apparently supported by the fact that constitutively activated Stat3 protein has been found in various types of tumors, including leukemia; cancers of the breast, head and neck, pancreas, and prostate; and melanoma (16–19) . Our recent studies of human pancreatic cancer and melanoma have linked abnormal Stat3 activation to overexpression of multiple genes downstream of Stat3, including VEGF(17, 18) and MMP-2(19). These findings led us to hypothesize that abnormal activation of Stat3 causes overexpression of multiple angiogenic molecules, which in turn render tumor cells highly angiogenic, as manifested by increased MVD. Recent studies also have revealed that altered Stat3 can contribute to oncogenesis, presumably through its critical role in the expression of many other genes key to the regulation of multiple aspects of tumor cell survival, growth, and angiogenesis (17–21). At present, it is not known whether abnormal Stat3 expression and activation critically contribute to gastric cancer development and progression. Because tumor MVD predicts patient survival (22), we asked whether Stat3 predicts tumor MVD, given the evidence that Stat3 may control the expression of many genes key to tumor angiogenesis, such as VEGF.

In the present study, we sought to determine whether Stat3 expression is related to VEGF expression and tumor MVD in gastric cancer specimens and whether these factors predict survival in gastric cancer patients. We found that elevated Stat3 activation and concomitant VEGF overexpression occurred in human gastric cancer and were directly correlated with tumor MVD and inversely correlated with patient survival. Furthermore, genetically enforced alterations of activated Stat3 expression led to altered VEGF expression and angiogenic potential in human gastric cancer cells. Therefore, abnormally activated Stat3 expression may be a potential molecular marker for poor prognosis and directly contribute to gastric cancer angiogenesis and aggressive gastric cancer biology.

Human Tissue Specimens and Patient Information. We used human gastric cancer tissue specimens preserved in the Gastric Cancer Tissue Bank at and obtained information about the patients from the University of Texas M.D. Anderson Cancer Center Upper Gastrointestinal Carcinoma Database. Primary gastric cancer in these patients was diagnosed and treated at M.D. Anderson Cancer Center from 1985 to 1998. The patients had well-documented clinical histories and follow-up information. None of them underwent preoperative chemotherapy and/or radiation therapy. Eighty-six patients with primary gastric cancer were randomly selected for this study. All of the patients had undergone gastrectomy with lymph node dissection. Gastric cancer centered at or above the gastroesophageal junction was not included in this study. Proximal gastric cancer centered below the gastroesophageal junction was included. Gastroesophageal junction cases associated with Barrett's were not included. All patients also were observed at M.D. Anderson Cancer Center through the end of 1999.The median follow-up period for the 86 patients was 25.7 months. At the end of 1999, 30 patients were still alive, whereas 56 patients had died. Fifty-three lymph node metastasis specimens from the 86 patients with gastric cancer and 57 normal gastric tissue specimens obtained from patients without gastric cancer were also included in this study.

Immunohistochemistry. Sections (5 μm thick) of formalin-fixed, paraffin-embedded tumor specimens were deparaffinized in xylene and rehydrated in graded alcohol. Antigen retrieval was done with 0.05% saponin for 30 minutes at room temperature. Endogenous peroxidase was blocked using 3% hydrogen peroxide in PBS for 12 minutes. The specimens were incubated for 20 minutes at room temperature with a protein-blocking solution consisting of PBS (pH 7.5) containing 5% normal horse serum and 1% normal goat serum and then incubated at 4°C in a 1:200 dilution of rabbit polyclonal antibody against human Stat3 (Phospho-Stat3 [tyr-705]; Cell Signaling Technology, Beverly, MA), or a 1:100 dilution of rabbit polyclonal antibody against human VEGF (clone A-20, SC-152, Santa Cruz Biotechnology, Santa Cruz, CA). The samples were then rinsed and incubated for 1 hour at room temperature with peroxidase-conjugated anti-rabbit IgG. Next, the slides were rinsed with PBS and incubated for 5 minutes with diaminobenzidine (Research Genetics, Huntsville, AL). The sections were washed thrice with distilled water, counterstained with Mayer's hematoxylin (Biogenex Laboratories, San Ramon, CA), and washed once each with distilled water and PBS. Afterward, the slides were mounted using Universal Mount (Research Genetics) and examined using a bright-field microscope. A positive reaction was indicated by a reddish-brown precipitate in the nuclei (Stat3) or cytoplasm (VEGF; ref. 18).

Stat3 and VEGF staining were classified as negative, weak positive, or strong positive according to the percentage of positive cells and staining intensity. Scores for percentage of positive cells were assigned as follows: ≤10% of cells positive, 0; 11% to 25% of cells positive, 1; 26% to 50% of cells positive, 2; 51% to 75% of cells positive, 3; and >75% of cells positive, 4. Scores for staining intensity were assigned as follows: no staining, 0; light brown, 1; brown, 2; and dark brown, 3. Overall scores were obtained by multiplying the percentage score by the intensity score. Overall scores ≤3 were defined as negative, overall scores >3 but ≤6 were defined as weak positive, and overall scores >6 were defined as strong positive. Two independent pathologists examined 5 random fields (1 field = 0.159 mm2, at ×100 magnification) of each sample, and scored each sample without knowledge of patient outcome (double-blinded). An average value of the two scores was presented in the present study (23).

Quantification of Tumor Microvessel Density. For CD34 staining, tissue sections were processed and stained with a 1:100 dilution of monoclonal goat anti-CD34 (PECAM1-M20; Santa Cruz Biotechnology), stained with peroxidase-conjugated anti-goat IgG, and then counterstained with Mayer's hematoxylin (Biogenex Laboratories). The slides were mounted and examined using a bright-field microscope. A positive reaction was indicated by a reddish-brown precipitate in the cytoplasm. For quantification of tumor MVD, highly vascular areas were initially identified by scanning tumor sections using light microscopy at low power. Vessels count was assessed in areas of the tumor containing the highest numbers of capillaries and small venules, based on the criteria of Weidner et al. (22). Vessels in five high-power fields (×200 magnification [×20 objective and ×10 ocular]) were counted by two independent investigators without knowledge of the patient outcome (double-blinded). An average value of the two scores was presented in the present study MVD was divided into three groups: low (<50 vessels per five high-power fields), moderate (50-100 vessels), and high (>100 vessels) as described previously (23).

Western Blot Analysis. Whole cell lysates were prepared from human gastric cancer cell lines and human normal and gastric cancer tissue specimens. Standard Western blotting was done using a polyclonal rabbit antibody against activated human Stat3 (Phospho-Stat3 [tyr-705]; Cell Signaling Technology), a polyclonal rabbit antibody against human VEGF; and anti-rabbit IgG, a horseradish peroxidase-linked F(ab′)2 fragment obtained from a donkey (Amersham Life Sciences, Arlington Heights, IL). Equal protein sample loading was monitored by incubating the same membrane filter with an anti-β-actin antibody. The probe proteins were detected using the Amersham enhanced chemiluminescence system according to the manufacturer's instructions (23).

Stable Dominant-Negative Stat3 and Constitutive Active Stat3 Transfection. Human gastric cancer AGS cells were transfected with dominant-negative Stat3 (Stat3-DN), constitutive active Stat3 (Stat3-CA), green fluorescent protein expression vector (GFP), and vector alone (control) using LipofectAMINE (Life Technologies, Inc., Rockville, MD). Stat3-CA was generated by substitution of the cysteine residues within the COOH-terminal loop of the SH2 domain of Stat3 (18, 19, 21). Stat3-DN construct was produced by a phenylalanine substitution of the tyrosine residue position at 705 resulting in a reduction of tyrosine phosphorylation of wild-type Stat3 and inhibition of endogenous Stat3 (18, 19, 24). Successful stable transfection was verified using anti-tag antibodies as described previously (18). Three independent clones of pStat3-CA (CA1, CA2, and CA3) and pStat3-DN transfection (DN1, DN2, and DN3) were used in this study. The dominant-negative Stat3 and control vector expression constructs were provided by Dr. Koichi Nakajima. (Osaka City University Graduate School of Medicine, Osaka, Japan).

Northern Blot Analysis. Total RNA was extracted using TRIzol reagent (Invitrogen, San Diego, CA). RNA (12 μg) was separated electrophoretically on a 1% denaturing formaldehyde agarose gel, transferred to a GeneScreen nylon membrane (DuPont, Boston, MA) in 20× SSC, and UV cross-linked using a UV-Stratalinker 1800 (Stratagene, La Jolla, CA). Additionally, the VEGF probe was labeled with [32P]-dCTP using a random labeling kit (Boehringer Mannheim Biochemicals, Indianapolis, IN). Equal loading of RNA samples was monitored by hybridizing the same membrane filter with a human β-actin cDNA probe (18).

Vascular Endothelial Growth Factor Protein Measurement. The VEGF protein levels in the culture supernatants were determined using the Quantikine VEGF ELISA kit (R&D Systems, Minneapolis, MN), which is a quantitative immunometric sandwich enzyme immunoassay. A curve of the absorbance of VEGF versus its concentration in the standard wells was plotted. By comparing the absorbance of the samples with the standard curve, we determined the VEGF concentration in the unknown samples (18).

Endothelial Cell Tube Formation Assay. The tube formation assay was done as described previously (25). Briefly, 250 μL of growth factor-reduced Matrigel (Collaborative Biomedical Products, Bedford, MA) were pipetted into each well of a 24-well plate and polymerized for 30 minutes at 37°C. Human umbilical vein endothelial cells were harvested after trypsin treatment and suspended in conditioned medium from either 1 × 106 AGS-Stat-DN cells or 1 × 106 AGS-Neo cells cultured for 48 hours in modified Eagle's medium containing 1% fetal bovine serum. Then 2 × 104 human umbilical vein endothelial cells in 300 μL of conditioned medium were added to each well and incubated at 37°C, 5% CO2, for 20 hours. The cultures were photographed with bright-field microscopy using a Sony digital camera equipped with an Optimas 6.2 program. The degree of tube formation was assessed as the percentage of cell surface area versus total surface area. Control cell cultures were given arbitrary percentage values of 100 (25).

Statistical Analysis. The two-tailed χ2 test was done to determine the significance of the difference between the covariates. Survival durations were calculated using the Kaplan-Meier method. The log-rank test was used to compare cumulative survival of patient groups. A Cox proportional hazards model was used to provide univariate and multivariate hazard ratios for the study variables. The expression levels of Stat3 and VEGF, MVD, age, sex, Lauren's histologic classification, stage (American Joint Committee on Cancer system), and completeness of surgical resection (R0 versus R1 or R2) were included in the model. In all of the tests, P < 0.05 was defined as statistically significant. SPSS version 11.05 (SPSS, Inc., Chicago, IL) was used for analyses.

Patient Characteristics. Eighty-six cases were selected to represent all stages and histologic types of malignant gastric cancer. The study included tissue samples from 56 men and 30 women. The mean patient age was 62 years. Proximal cancer localization was observed in 20 cases. Fifty-three patients had intestinal type and 33 had diffuse type cancer. Details of patient characteristics are provided in Table 1.

Table 1

Patient characteristics and pStat3 expression

VariableAll patientsStat3 expression, n
NegativeWeakStrong
Sex     
    Male 56 14 28 14 
    Female 30 15 
Stage     
    I 14 
    II 28 20 
    III 30 14 
    IV 14 
Residual disease     
    R0 69 19 34 16 
    R1, R2 17 
Pathology type     
    Papillary 12 
    Tubular 28 12 
    Diffuse 
    Mucinous 
    Signet ring 21 11 
    Mixed 12 
Lauren's histologic type     
    Intestinal 53 14 28 11 
    Diffuse 33 15 
VariableAll patientsStat3 expression, n
NegativeWeakStrong
Sex     
    Male 56 14 28 14 
    Female 30 15 
Stage     
    I 14 
    II 28 20 
    III 30 14 
    IV 14 
Residual disease     
    R0 69 19 34 16 
    R1, R2 17 
Pathology type     
    Papillary 12 
    Tubular 28 12 
    Diffuse 
    Mucinous 
    Signet ring 21 11 
    Mixed 12 
Lauren's histologic type     
    Intestinal 53 14 28 11 
    Diffuse 33 15 

NOTE. No statistically significant differences in the patient characteristics according to Stat3 group were observed.

Signal Transducer and Activator of Transcription 3 Expression and its Association with Vascular Endothelial Growth Factor Expression and Microvessel Density in Human Gastric Tumor Tissue. Stat3 expression and MVD were evaluated by immunohistochemistry in the primary cancer tissue of 86 patients. Strong Stat3 expression was observed in 20 cases (23%). Stat3 expression was classified as weak or negative in 43 cases (50%) and 23 cases (27%), respectively. No significant differences were detected between the three Stat3 expression categories in the distribution of gender, race or ethnicity, tumor location, type of resection, residual disease status, extent of lymphadenectomy, or Lauren's histologic classification (Table 1). High MVD was observed in 31 cases (36%). Low or moderate MVD was observed in 45 cases (52%) and 10 cases (12%), respectively. Stat3 expression significantly correlated with VEGF expression and MVD. This finding was further confirmed by analysis of consecutive tissue sections, in which the Stat3 expression pattern was consistent with patterns of VEGF expression (Fig. 1), and by Western blot analysis of Stat3 and VEGF protein expression (Fig. 2). Therefore, these data provided clinical evidence to support our hypothesis that aberrant Stat3 activation causes increased angiogenesis through the overexpression of various angiogenic molecules such as VEGF.

Fig. 1

pStat3 and VEGF protein expression in human gastric cancer tissue. Two sets of consecutive tissue sections of high and low Stat3 expression were prepared from formalin-fixed, paraffin-embedded specimens of gastric cancers. Immunohistochemical staining done using specific antibodies against phospho-Stat3 and VEGF. A1, B1, C1, and D1 (original magnification: ×200), representative areas from A, B, C, and D (original magnification: ×50), respectively. Of note is that high Stat3 expression directly correlated with increased VEGF expression.

Fig. 1

pStat3 and VEGF protein expression in human gastric cancer tissue. Two sets of consecutive tissue sections of high and low Stat3 expression were prepared from formalin-fixed, paraffin-embedded specimens of gastric cancers. Immunohistochemical staining done using specific antibodies against phospho-Stat3 and VEGF. A1, B1, C1, and D1 (original magnification: ×200), representative areas from A, B, C, and D (original magnification: ×50), respectively. Of note is that high Stat3 expression directly correlated with increased VEGF expression.

Close modal
Fig. 2

pStat3 and VEGF protein expression in human gastric cancer tissue. Whole-cell protein extracts were prepared from four paired normal gastric (N) and gastric tumor tissue (T) specimens obtained from the patients with pStat3 and VEGF expression detected via immunostaining [immunohistochemical (IHC) scores in italics]. The level of expression of pStat3 and VEGF protein was determined using Western blot analysis with 10-μg whole cell protein extracts. Equal protein sample loading was monitored by hybridizing the same membrane filter with an anti-β-actin antibody. Of note is that the levels of both pStat3 and VEGF protein expression were significantly elevated in tumor tissue compared with normal tissue.

Fig. 2

pStat3 and VEGF protein expression in human gastric cancer tissue. Whole-cell protein extracts were prepared from four paired normal gastric (N) and gastric tumor tissue (T) specimens obtained from the patients with pStat3 and VEGF expression detected via immunostaining [immunohistochemical (IHC) scores in italics]. The level of expression of pStat3 and VEGF protein was determined using Western blot analysis with 10-μg whole cell protein extracts. Equal protein sample loading was monitored by hybridizing the same membrane filter with an anti-β-actin antibody. Of note is that the levels of both pStat3 and VEGF protein expression were significantly elevated in tumor tissue compared with normal tissue.

Close modal

Effects of Signal Transducer and Activator of Transcription 3 Expression, Vascular Endothelial Growth Factor Expression, and Microvessel Density on Patient Survival. Median survival durations according to Stat3 and VEGF expression are presented in Table 2. Elevated Stat3 expression, but not increased VEGF, was associated with inferior survival (Table 2). The median survival durations in patients with low, moderate, and high MVD were 64, 37, and 14 months, respectively. The effect of MVD expression on survival duration was statistically significant (Table 2). Other variables that affected survival in univariate analyses included stage and completeness of resection (Table 2). Age at diagnosis (as a continuous variable by univariate Cox proportional hazard analysis) and Lauren's histologic type did not have a statistically significant affect on survival.

Table 2

Univariate and multivariate survival analyses

VariableUnivariate median survival, moPMultivariate hazard ratio* (95% confidence interval)P
Stat3 expression     
    Negative 43 0.013 1.0 (reference)  
    Weak 53  0.82 (0.38-1.76) 0.630 
    Strong 20  2.28 (0.99-4.58) 0.049 
VEGF expression     
    Negative 29 0.074 1.0 (reference)  
    Weak 41  1.34 (0.62-3.28) 0.436 
    Strong 14  1.89 (0.73-4.47) 0.158 
MVD     
    Low 64 0.036 1.0 (reference)  
    Moderate 37  1.31 (0.49-3.08) 0.663 
    High 14  2.88 (0.84-7.56) 0.071 
Stage     
    I 91 <0.001 1.0 (reference)  
    II 64  1.48 (0.43-4.83) 0.518 
    III 23  6.44 (2.13-19.40) 0.001 
    IV  8.69 (2.25-33.88) 0.001 
Resection status     
    R0 41 <0.001 1.0 (reference)  
    R1 or R2 11  1.18 (0.52-2.57) 0.713 
Lauren's histologic type     
    Intestinal 41 0.061 1.0 (reference)  
    Diffuse 15  1. 37 (0.77-2.49) 0.274 
Age (continuous variable) Hazard ratio 1.004 0.701 1.0 (0.98-1.02) 0.966 
VariableUnivariate median survival, moPMultivariate hazard ratio* (95% confidence interval)P
Stat3 expression     
    Negative 43 0.013 1.0 (reference)  
    Weak 53  0.82 (0.38-1.76) 0.630 
    Strong 20  2.28 (0.99-4.58) 0.049 
VEGF expression     
    Negative 29 0.074 1.0 (reference)  
    Weak 41  1.34 (0.62-3.28) 0.436 
    Strong 14  1.89 (0.73-4.47) 0.158 
MVD     
    Low 64 0.036 1.0 (reference)  
    Moderate 37  1.31 (0.49-3.08) 0.663 
    High 14  2.88 (0.84-7.56) 0.071 
Stage     
    I 91 <0.001 1.0 (reference)  
    II 64  1.48 (0.43-4.83) 0.518 
    III 23  6.44 (2.13-19.40) 0.001 
    IV  8.69 (2.25-33.88) 0.001 
Resection status     
    R0 41 <0.001 1.0 (reference)  
    R1 or R2 11  1.18 (0.52-2.57) 0.713 
Lauren's histologic type     
    Intestinal 41 0.061 1.0 (reference)  
    Diffuse 15  1. 37 (0.77-2.49) 0.274 
Age (continuous variable) Hazard ratio 1.004 0.701 1.0 (0.98-1.02) 0.966 
*

Hazard ratios estimated using multivariate Cox proportional hazards modeling.

Estimated using univariate Cox proportional hazards model.

Next, Stat3 expression, VEGF expression, MVD, stage, completeness of resection, age, and Lauren's histologic classification were entered into a Cox proportional hazards model for multivariate analysis (Table 2). After adjustment for the effect of covariates, strong Stat3 expression [P = 0.049; hazard ratio, 2.28; 95% confidence interval, 0.99-4.58] and advanced stage (P < 0.01) were independent predictors of poor survival. In multivariate analyses, trends toward poorer survival were observed among patients who had high MVD and strong VEGF expression. However, these affects on survival were not statistically significant.

Suppression of Gastric Cancer Angiogenesis by Blockade of Activated Signal Transducer and Activator of Transcription 3 Expression in Human Gastric Cancer Cells. To provide direct evidence of whether Stat3 regulates the angiogenic phenotype of gastric cancer, AGS human gastric cancer cells were stably transfected with pStat3-DN expression vector as we previously reported (18, 19). We found that AGS cells transfected with pStat3-DN (AGS-DN1, AGS-DN2, and AGS-DN3), but not AGS cells transfected with control vector (AGS-Neo1 and AGS-Neo2), exhibited a decreased expression of VEGF as determined using Northern blotting (Fig. 3A) and ELISA (Fig. 3B). Furthermore, consistent with decreased expression of VEGF, the supernatant from pStat3-DN-transfected (DN1 and DN2) cells seemed to be less angiogenic than that from control vector-transfected (Vector) cells as determined by an endothelial cell tube formation assay (Fig. 3C). Addition of exogenous VEGF to the conditioned medium at least partially overcame the Stat3-DN inhibition of tube formation (Fig. 3C). The representative pictures were taken in situ for tube formation in the supernatant of control transfected AGS cells (Fig. 3D,1), Stat3-DN-transfected AGS cells (Fig. 3D,2) and Stat3-DN-transfected AGS cells with the addition of 1 ng/mL VEGF (Fig. 3D 3). We found that Stat3-DN suppressed tube formation, which was partially overcome by addition of exogenous VEGF. These results suggest that blockade of Stat3 activity suppresses VEGF expression and impairs the angiogenic phenotype of gastric cancer cells.

Fig. 3

Genetically enforced down-regulation of activated Stat3 expression suppresses VEGF expression and impairs angiogenic potential in human gastric cancer cells. Cellular mRNA was extracted from AGS cells (lane 1), AGS cells transfected with a control vector (AGS-Neo1, lane 2 and AGS-Neo2, lane 3), or AGS cells transfected with pStat3-DN expression vector (AGS-DN1, lane 4; AGS-DN2, lane 5; and AGS-DN3, lane 6). VEGF mRNA expression was determined using Northern blot analysis (A). Equal loading of RNA samples was monitored by hybridizing the same membrane filter with a human β-actin cDNA probe. Culture supernatant was harvested after 48 hours of incubation of 2 × 106 cells in 10 mL medium. VEGF protein secretion was determined by ELISA (B). The angiogenic potentials of the supernatant of control transfected AGS cells (vector) and pStat3-DN-transfected AGS cells (DN1 and DN2) with or without the addition of 10 ng/mL VEGF were determined by an endothelial cell tube formation assay. The degree of tube formation was assessed as the percentage of cell surface area versus total surface area. Columns, % angiogenesis; bars, SE. Empty vector-transfected cell cultures were given arbitrary percentage values of 100 (C). Representative pictures were taken in situ for tube formation in the supernatant of control transfected AGS cells (D1), pStat3-DN-transfected AGS cells (D2), and pStat3-DN-transfected AGS cells with the addition of 1 ng/mL VEGF (D3). Note that blockade of Stat3 activity suppressed VEGF expression and impaired the angiogenic phenotype of gastric cancer cells. *, P < 0.01, statistical significance as comparisons were made between the Stat3-DN-transfected cells and respective control groups.

Fig. 3

Genetically enforced down-regulation of activated Stat3 expression suppresses VEGF expression and impairs angiogenic potential in human gastric cancer cells. Cellular mRNA was extracted from AGS cells (lane 1), AGS cells transfected with a control vector (AGS-Neo1, lane 2 and AGS-Neo2, lane 3), or AGS cells transfected with pStat3-DN expression vector (AGS-DN1, lane 4; AGS-DN2, lane 5; and AGS-DN3, lane 6). VEGF mRNA expression was determined using Northern blot analysis (A). Equal loading of RNA samples was monitored by hybridizing the same membrane filter with a human β-actin cDNA probe. Culture supernatant was harvested after 48 hours of incubation of 2 × 106 cells in 10 mL medium. VEGF protein secretion was determined by ELISA (B). The angiogenic potentials of the supernatant of control transfected AGS cells (vector) and pStat3-DN-transfected AGS cells (DN1 and DN2) with or without the addition of 10 ng/mL VEGF were determined by an endothelial cell tube formation assay. The degree of tube formation was assessed as the percentage of cell surface area versus total surface area. Columns, % angiogenesis; bars, SE. Empty vector-transfected cell cultures were given arbitrary percentage values of 100 (C). Representative pictures were taken in situ for tube formation in the supernatant of control transfected AGS cells (D1), pStat3-DN-transfected AGS cells (D2), and pStat3-DN-transfected AGS cells with the addition of 1 ng/mL VEGF (D3). Note that blockade of Stat3 activity suppressed VEGF expression and impaired the angiogenic phenotype of gastric cancer cells. *, P < 0.01, statistical significance as comparisons were made between the Stat3-DN-transfected cells and respective control groups.

Close modal

In contrast, AGS cells transfected with pStat3-CA (AGS-CA1, AGS-CA2, and AGS-CA3), but not AGS cells transfected with control GFP expression vector (AGS-GFP), exhibited an increased expression of VEGF as determined using Northern blotting (Fig. 4A) and ELISA (Fig. 4B). Furthermore, consistent with increased expression of VEGF, the supernatant from pStat3-CA-transfected (AGS-CA) cells seemed to be more angiogenic than that from control vector-transfected (AGS-GFP) cells as determined by an endothelial cell tube formation assay (Fig. 4C).

Fig. 4

Genetically enforced up-regulation of activated Stat3 expression increased VEGF expression and angiogenic potential in human gastric cancer cells. Cellular mRNA was extracted from AGS cells (lane 1), AGS cells transfected with a control GFP expression vector (AGS-GFP1, lane 2 and AGS-GFP2, lane 3), or AGS cells transfected with pStat3-CA expression vector (AGS-CA1, lane 4; AGS-CA2, lane 5; and AGS-CA3, lane 6). VEGF mRNA expression (A), VEGF protein secretion assay (B), and the angiogenic potentials of the supernatants of tumor cell cultures (C) were determined as described in Fig. 3. Note that increased Stat3 activity up-regulated VEGF expression and increased the angiogenic phenotype of gastric cancer cells. *, P < 0.01, statistical significance as comparisons were made between the Stat3-CA-transfected cells and respective control groups.

Fig. 4

Genetically enforced up-regulation of activated Stat3 expression increased VEGF expression and angiogenic potential in human gastric cancer cells. Cellular mRNA was extracted from AGS cells (lane 1), AGS cells transfected with a control GFP expression vector (AGS-GFP1, lane 2 and AGS-GFP2, lane 3), or AGS cells transfected with pStat3-CA expression vector (AGS-CA1, lane 4; AGS-CA2, lane 5; and AGS-CA3, lane 6). VEGF mRNA expression (A), VEGF protein secretion assay (B), and the angiogenic potentials of the supernatants of tumor cell cultures (C) were determined as described in Fig. 3. Note that increased Stat3 activity up-regulated VEGF expression and increased the angiogenic phenotype of gastric cancer cells. *, P < 0.01, statistical significance as comparisons were made between the Stat3-CA-transfected cells and respective control groups.

Close modal

Angiogenesis is essential for tumor growth and metastasis (26). Numerous lines of evidence have shown that angiogenesis, as quantitated according to MVD, plays significant clinicopathologic roles in cancer patients (22, 27). Notably, MVD can be an independent, highly significant, accurate prognostic indicator in cancer patients (22). In the present study, we found direct clinical evidence of a strong correlation between Stat3 expression and VEGF expression and MVD in human gastric cancer. Although VEGF, MVD, and Stat3 were all associated with poor patient survival, Stat3 was apparently more powerful than VEGF and MVD in predicting clinical outcome of gastric cancer patients as indicated by multivariate analysis showing that only Stat3 was an independent prognostic factor.

Tumor angiogenesis has been considered the most important predictor of overall survival in gastric cancer (6–10). Moreover, some key angiogenic factors have also been indicated to be independent prognostic factors in gastric cancer (3, 11–15). However, the clinicopathologic role of MVD and its major regulatory molecules in gastric cancer development and progression remain largely unclear. Angiogenesis is regulated by multiple factors, including VEGF (6–10), and many of these factors may individually predict MVD. For example, VEGF, which is considered critical to angiogenesis, has a close association with MVD (3, 11–15). The present study showed an overall close correlation between VEGF and MVD, which reflects the importance of VEGF in tumor angiogenesis and is clearly consistent with the earlier findings. However, expression of VEGF and its receptors has not correlated with MVD or malignancy in many other types of tumors, including digestive endocrine tumors (28). Presumably, different types of tumors may use angiogenic factors other than VEGF to induce vascular formation. Many lines of evidence from studies of gastric cancer and other tumor types indicate a relationship between MVD and other angiogenic molecules (4, 12, 13). Therefore, MVD should have better prognostic value than any particular angiogenic molecule, because increased MVD might result from overexpression of any major angiogenic factors. In the present study, Stat3 seemed to be powerful predictor of MVD. However, more studies are needed to determine whether Stat3 is a better MVD-predicting factor than individual angiogenic factors, considering that Stat3 is a critical transcription factor and may control the expression of many genes key to tumor angiogenesis, such as VEGF(17, 18).

Studies on the association between MVD and prognosis in gastric cancer have produced inconsistent results; some have found that MVD is a predictor of poor prognosis, whereas others have found that MVD predicts good prognosis (11, 13, 15, 29–35). In the present study, overall survival was significantly lower in patients with a high MVD than in patients with a low MVD, which is consistent with several previous reports (11, 14, 32, 33, 36, 37). However, multivariate analysis revealed that MVD was not an independent prognostic indicator, whereas Stat3 was. This superiority of Stat3 may underline the fact that prognosis is directly related to tumor biology manifested by its multiple aspects, including the angiogenic phenotype, invasive capacity, apoptosis resistance, and proliferation. Thus, an elevated MVD may not necessarily predict all other aspects of cancer biology (i.e., the overall biological behavior), besides the angiogenic phenotype, whereas Stat3 might most likely do so, because it is an important transcription factor for many genes that may regulate all aspects of cancer biology (20, 38). However, more studies are needed to substantiate whether that the Stat3 expression and activation status is a powerful and practical predictor of patient outcome.

Furthermore, Stat3 may be not only a useful molecular marker for selecting patients with a poor prognosis to receive more aggressive preoperative or adjuvant therapy in the setting of a clinical trial but also an effective therapeutic target for gastric cancer. In gastric cancer, multiple growth factor pathways are involved though multiple tyrosine kinases, such as epidermal growth factor receptor, HER2, VEGF receptor-1, VEGF receptor-2, platelet-derived growth factor receptor, and c-Met. Inhibition of one such pathway may not be sufficient for antitumor activity. With its central regulatory role in many of these pathways (20), Stat3 represents an attractive target for the development of effective therapies as evidenced by our recent studies in human pancreatic cancer and melanoma (18, 19).

Our current study provided the first evidence for a critical role of activated Stat3 in gastric cancer angiogenesis. Since activated Stat3 up-regulates matrix metalloproteinase-2 (19), cyclin D1 (21), c-myc, Bcl-xL, and Mcl-1(17, 39), but down-regulates death receptor Fas (40), Stat3 may enhance tumor progression inclusively by affecting the expression of various genes related to cell survival, the cell cycle, invasion, and angiogenesis (39–43). Further exploring those molecular mechanisms that result in Stat3 overactivation may not only shed more light on abnormal Stat3 activation but also help improve understanding of Stat3's value as a prognostic factor and aid in the development of effective therapies targeting Stat3.

In summary, we discovered that the level of Stat3 expression in gastric cancer was directly related to VEGF expression level and MVD, which are closely related to the postoperative prognosis for gastric cancer. This study further showed that abnormally activated Stat3 expression represents a potential risk factor for poor prognosis and directly contributes to gastric cancer angiogenesis and progression. Therefore, preoperative determination of the level of Stat3 activity may be useful in deciding on the modality and extent of postoperative therapy. We are currently investigating the molecular mechanism by which the Stat3 signaling pathway regulates gastric cancer development and progression and whether this pathway is a therapeutic target for controlling gastric cancer growth and metastasis.

Grant support: American Cancer Society Research Scholar Grant CSM-106659 (S. Huang) and National Cancer Institute, NIH Cancer Center Support Grant CA 16672.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Note: W. Gong and L. Wang contributed equally to this work.

We thank Lydia Soto for help in the preparation of this article and Stephanie P. Deming for editorial comments.

1
Parkin DM, Pisani P, Ferlay J. Global cancer statistics.
CA Cancer J Clin
1999
;
49
:
33
–64.
2
Tahara E. Molecular aspects of invasion and metastasis of stomach cancer.
Verh Dtsch Ges Pathol
2000
;
84
:
43
–9.
3
Chan AO, Luk JM, Hui WM, Lam SK. Molecular biology of gastric carcinoma: from laboratory to bedside.
J Gastroenterol Hepatol
1999
;
14
:
1150
–60.
4
Xie K, Huang S. Regulation of cancer metastasis by stress pathways.
Clin Exp Metastasis
2003
;
20
:
31
–43.
5
Siewert JR, Bottcher K, Stein HJ, Roder JD. Relevant prognostic factors in gastric cancer: ten-year results of the German Gastric Cancer Study.
Ann Surg
1998
;
228
:
449
–61.
6
Xie K. Interleukin-8 and human cancer biology.
Cytokine Growth Factor Rev
2001
;
12
:
375
–91.
7
Brown LF, Berse B, Jackman RW, et al. Expression of vascular permeability factor (vascular endothelial growth factor) and its receptors in adenocarcinomas of the gastrointestinal tract.
Cancer Res
1993
;
53
:
4727
–35.
8
Guo YS, Beauchamp RD, Jin GF, Townsend CM Jr, Thompson JC. Insulin-like growth factor-binding protein modulates the growth response to insulinlike growth factor 1 by human gastric cancer cells.
Gastroenterology
1993
;
104
:
1595
–604.
9
Yoshikawa T, Tsuburaya A, Kobayashi O, et al. Plasma concentrations of VEGF and bFGF in patients with gastric carcinoma.
Cancer Lett
2000
;
153
:
7
–12.
10
Sier CF, Kuben FJ, Ganesh S, et al. Tissue levels of matrix metalloproteinases MMP-2 and MMP-9 are related to the overall survival of patients with gastric carcinoma.
Br J Cancer
1996
;
74
:
413
–7.
11
Du JR, Jiang Y, Zhang YM, Fu H. Vascular endothelial growth factor and microvascular density in esophageal and gastric carcinomas.
World J Gastroenterol
2003
;
9
:
1604
–6.
12
Li HX, Chang XM, Song ZJ, He SX. Correlation between expression of cyclooxygenase-2 and angiogenesis in human gastric adenocarcinoma.
World J Gastroenterol
2003
;
9
:
674
–7.
13
Kaneko T, Konno H, Baba M, Tanaka T, Nakamura S. Urokinase-type plasminogen activator expression correlates with tumor angiogenesis and poor outcome in gastric cancer.
Cancer Sci
2003
;
94
:
43
–9.
14
Sanz-Ortega J, Steinberg SM, Moro E, et al. Comparative study of tumor angiogenesis and immunohistochemistry for p53, c-ErbB2, c-myc and EGFr as prognostic factors in gastric cancer.
Histol Histopathol
2000
;
15
:
455
–2.
15
Chen CN, Cheng YM, Lin MT, Hsieh FJ, Lee PH, Chang KJ. Association of color Doppler vascularity index and microvessel density with survival in patients with gastric cancer.
Ann Surg
2002
;
235
:
512
–8.
16
Yu CL, Meyer DJ, Campbell GS, et al. Enhanced DNA-binding activity of a Stat3-related protein in cells transformed by the Src oncoprotein.
Science
1995
;
269
:
81
–3.
17
Niu G, Bowman T, Huang M, et al. Roles of activated Src and Stat3 signaling in melanoma tumor cell growth.
Oncogene
2002
;
21
:
7001
–10.
18
Wei D, Le X, Zheng L, et al. Stat3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis.
Oncogene
2003
;
22
:
319
–29.
19
Xie TX, Wei D, Liu M, et al. Stat3 activation regulates the expression of matrix metalloproteinase-2 and tumor invasion and metastasis.
Oncogene
2004
;
23
:
3550
–60.
20
Darnell JE Jr. STATs and gene regulation.
Science
1997
;
277
:
1630
–5.
21
Bromberg JF, Wrzeszczynska MH, Devgan G, et al. Stat3 as an oncogene.
Cell
1999
;
98
:
295
–303.
22
Weidner N, Semple JP, Welch WR, Folkman J. Tumor angiogenesis and metastasis: correlation in invasive breast carcinoma.
N Engl J Med
1991
;
324
:
1
–8.
23
Wang L, Wei D, Huang S, et al. Transcription factor Sp1 expression is a significant predictor of survival in human gastric cancer.
Clin Cancer Res
2003
;
9
:
6371
–80.
24
Kaptein A, Paillard V, Saunders M. Dominant negative stat3 mutant inhibits interleukin-6- induced lak-STAT signal transduction.
J Biol Chem
1996
;
271
:
5961
–4.
25
Huang S, Mills L, Mian B, et al. Fully humanized neutralizing antibodies to interleukin-8 (ABX-IL8) inhibit angiogenesis, tumor growth, and metastasis of human melanoma.
Am J Pathol
2002
;
161
:
125
–34.
26
Folkman J. What is the evidence that tumors are angiogenesis dependent?
J Natl Cancer Inst
1990
;
82
:
4
–6.
27
Weidner N, Folkman J, Pozza F, et al. Tumor angiogenesis: a new significant and independent prognostic indicator in early-stage breast carcinoma.
J Natl Cancer Inst
1992
;
84
:
1875
–87.
28
La Rosa S, Uccella S, Finzi G, Albarello L, Sessa F, Capella C. Localization of vascular endothelial growth factor and its receptors in digestive endocrine tumors: correlation with microvessel density and clinicopathologic features.
Hum Pathol
2003
:
34
:
18
–27.
29
Shi H, Xu JM, Hu NZ, Xie HJ. Prognostic significance of expression of cyclooxygenase-2 and vascular endothelial growth factor in human gastric carcinoma.
World J Gastroenterol
2003
;
9
:
1421
–6.
30
Joo YE, Rew JS, Seo YH, et al. Cyclooxygenase-2 overexpression correlates with vascular endothelial growth factor expression and tumor angiogenesis in gastric cancer.
J Clin Gastroenterol
2003
;
37
:
28
–33.
31
Takahashi R, Tanaka S, Hiyama T, et al. Hypoxia-inducible factor-1α expression and angiogenesis in gastrointestinal stromal tumor of the stomach.
Oncol Rep
2003
:
10
:
797
–802.
32
Joo YE, Sohn YH, Joo SY, et al. The role of vascular endothelial growth factor (VEGF) and p53 status for angiogenesis in gastric cancer.
Korean J Intern Med
2002
;
17
:
211
–9.
33
Tenderenda M, Rutkowski P, Jesionek-Kupnicka D, Kubiak R. Expression of CD34 in gastric cancer and its correlation with histology, stage, proliferation activity, p53 expression and apoptotic index.
Pathol Oncol Res
2001
;
7
:
129
–34.
34
Koide N, Watanabe H, Shimozawa N, et al. Four resections for hepatic metastasis from gastric cancer: histochemical analysis of cell proliferation, apoptosis, and angiogenesis.
J Gastroenterol
2000
;
35
:
150
–4.
35
Ikeguchi M, Oka S, Saito H, et al. The expression of vascular endothelial growth factor and proliferative activity of cancer cells in gastric cancer.
Langenbecks Arch Surg
1999
;
384
:
264
–70.
36
Lu M, Jiang Y, Wang R. [The relationship of vascular endothelial growth factor and angiogenesis to the progression of gastric carcinoma].
Zhonghua Bing Li Xue Za Zhi
1998
;
27
:
278
–81.
37
Yu H, Jove R. The stats of cancer: new molecular targets come of age.
Nat Rev Cancer
2004
;
4
:
97
–105.
38
Takahashi R, Tanaka S, Kitadai Y, et al. Expression of vascular endothelial growth factor and angiogenesis in gastrointestinal stromal tumor of the stomach.
Oncology
2003
;
64
:
266
–74.
39
Catlett-Falcone R, Landowski TH, Oshiro MM, et al. Constitutive activation of Stat3 signaling confers resistance to apoptosis in human U266 myeloma cells.
Immunity
1999
;
10
:
105
–15.
40
Ivanov VN, Bhoumik A, Krasilnikov M, et al. Cooperation between STAT3 and c-jun suppresses Fas transcription.
Mol Cell
2001
;
7
:
517
–28.
41
Gao B, Shen X, Kunos G, et al. Constitutive activation of JAK-STAT3 signaling by BRCA1 in human prostate cancer cells.
FEBS Lett
2001
;
488
:
179
–84.
42
Schindler C, Darnell JE Jr. Transcriptional responses to polypeptide ligands: the JAK-STAT pathway.
Annu Rev Biochem
1995
;
64
:
621
–51.
43
Ni Z, Lou W, Leman ES, Gao AC. Inhibition of constitutively activated Stat3 signaling pathway suppresses growth of prostate cancer cells.
Cancer Res
2000
;
60
:
1225
–8.