Prognostic significance of the co‑overexpression of fibroblast growth factor receptors 1, 2 and 4 in gastric cancer

  • Authors:
    • Hideaki Murase
    • Mikito Inokuchi
    • Yoko Takagi
    • Keiji Kato
    • Kazuyuki Kojima
    • Kenichi Sugihara
  • View Affiliations

  • Published online on: May 15, 2014     https://doi.org/10.3892/mco.2014.293
  • Pages: 509-517
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The overexpression of fibroblast growth factor receptor (FGFR) 2 is an established prognostic factor and treatment target in gastric cancer. However, the roles of other FGFRs have not been fully elucidated. In this study, we investigated the correlations of the expression of FGFR1‑4 with clinicopathological characteristics and outcomes in gastric cancer. Tumor samples were obtained from 222 patients with gastric adenocarcinoma who underwent gastrectomy between 2003 and 2007. The expression of each FGFR was measured in the tumors by immunohistochemical analysis. The overexpression of FGFR1, FGFR2 or FGFR4 was found to be significantly associated with tumor progression, including depth of invasion, lymph node metastasis, pathological stage and distant metastasis or recurrent disease. Patients exhibiting overexpression of FGFR1, FGFR2 or FGFR4 had a significantly poorer disease‑specific survival (DSS; P<0.001, P=0.008 and P<0.001, respectively). Moreover, the co‑overexpression of all three FGFRs was significantly associated with a poorer DSS compared to the expression of none or only one of the FGFRs (P<0.001 and P=0.001, respectively) and it was found to be an independent prognostic factor (HR=1.71, 95% CI: 1.02‑2.85, p=0.041). In conclusion, high expression of FGFR1, FGFR2 or FGFR4 was associated with tumor progression and poor survival in patients with gastric cancer. Similar to FGFR2, FGFR1 and FGFR4 may be considered as prognostic factors and treatment targets in gastric cancer.

Introduction

Gastric cancer is the second leading cause of cancer-related mortality worldwide, accounting for ~1 in 10 of all deaths from cancer (1). The outcome of gastric cancer is generally poor, with a 5-year relative survival of <30% in most countries (2). Although radical surgery remains the cornerstone of treatment for gastric cancer, surgery alone appears to have reached its limits in terms of local control and survival. The achievement of locoregional control remains difficult in the presence of advanced disease (3). The majority of patients with advanced gastric cancer receive palliative chemotherapy, which is associated with a median survival of 11–12 months (4). In addition to standard cytotoxic regimens, targeted therapies, using small molecules or antibodies designed to disrupt the activity of specific oncogenic signaling pathways, have recently emerged as a promising treatment strategy. A number of receptor tyrosine kinases (RTKs) have been associated with tumor progression and patient outcomes in various types of cancer. RTK inhibitors, such as human epidermal growth factor receptor (HER), have been evaluated and some have been used to treat gastrointestinal cancers. In a recent ToGA trial (5), trastuzumab, a monoclonal antibody against the p185HER2 protein, improved the overall survival of patients with HER2-positive tumors when combined with chemotherapy. However, only 7–17% of gastric cancer patients have HER2-positive tumors and are considered as suitable candidates for anti-HER2 therapy (6,7). Further investigations are required to increase the number of patients with gastric cancer for whom targeted treatments may be a viable clinical option.

The fibroblast growth factor receptor (FGFR) family (FGFR1-4) belongs to the receptor tyrosine kinase superfamily. FGFRs regulate fundamental developmental pathways by interacting with fibroblast growth factors (FGFs) and thereby control a wide range of events, extending from mesoderm patterning in the early embryo to the development of multiple organ systems (8,9). FGF signaling participates in several biological functions in the adult organism, including regulation of angiogenesis and wound repair. FGFRs are expressed on a number of different cell types and regulate key cell activities, such as proliferation, survival, migration and differentiation, which renders FGF signaling susceptible to subversion by cancer cells (10).

FGFR2 amplifications have been reported in 10% of gastric cancers, the majority of which are of the diffuse type (11). FGFR2 amplification may correlate with poor outcomes in patients with diffuse-type gastric cancer (12). Moreover, the presence of FGFR2 gene amplification in gastric cancer is associated with sensitivity to inhibition of FGFR signaling by tyrosine kinase inhibitors and monoclonal antibodies in preclinical models (13,14). Thus, FGFR2 has attracted considerable attention as a novel therapeutic candidate for the development of targeted anticancer agents (15).

In contrast to FGFR2, the roles of FGFR1, FGFR3 and FGFR4 have not been fully elucidated. Overexpression of these FGFRs in gastric cancer was reported by a few small studies (1619). In this study, we aimed to investigate the correlations of FGFR1-4 immunohistochemical expression with clinicopathological characteristics and outcomes in gastric cancer.

Patients and methods

Patients

Our study group comprised 222 patients with primary gastric adenocarcinoma who underwent surgical resection between January, 2003 and December, 2007 in the Department of Esophagogastric Surgery, Tokyo Medical and Dental University. Each tumor was classified according to the tumor-node-metastasis staging system recommended by the International Union Against Cancer. Of the 222 patients, 168 were men and 54 were women. The mean age of the patients was 64.6 years (range, 21–92 years). All the patients were evaluated for recurrent disease by tumor marker analysis or diagnostic imaging (computed tomography, ultrasonography, magnetic resonance imaging and endoscopy) every 3–6 months. Patients with distant metastasis or recurrent disease received chemotherapy with S-1 alone or combined chemotherapy. A total of 20 patients (9%) received adjuvant chemotherapy with S-1 following radical resection. All the patients were followed up until July, 2012. The median follow-up was 60 months (range, 3–111 months). A total of 77 patients (35%) succumbed to gastric cancer, 66 (30%) had recurrent disease and 11 (5%) died from other causes.

Immunostaining of the FGFR family

Immunohistochemical analysis was performed with the use of secondary antibodies conjugated to a peroxidase-labeled polymer [Histofine Simple Stain MAX PO (Multi); Nichirei Co., Tokyo, Japan]. Polyclonal rabbit antibodies against FGFR1, FGFR2, FGFR3 and FGFR4 were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA). All the available hematoxylin and eosin-stained slides of the surgical specimens were reviewed. For each case, representative formalin-fixed, paraffin-embedded tissue blocks were selected for immunohistochemical studies and sliced into 4-μm sections. Following deparaffinization and rehydration, antigen retrieval was performed at 98°C for 30 min, using a pH 6.0, 10 mmol/l sodium citrate buffer (Mitsubishi Chemical Medience Corporation, Tokyo, Japan) in a microwave processor (MI-77; Azumaya, Tokyo, Japan). Endogenous peroxidase was blocked with 3% hydrogen peroxide in methanol. Subsequently, non-specific binding was blocked by treating the slides with 10% normal goat serum for 10 min at room temperature. The slides were incubated with the primary antibodies, including anti-FGFR1 (dilution, 1:100), anti-FGFR2 (dilution, 1:300), anti-FGFR3 (dilution, 1:500) and anti-FGFR4 (dilution, 1:100) in 1% bovine serum albumin/phosphate-buffered saline overnight at 4°C. The sections were then incubated with Simple Stain Max PO (Multi) for 30 min at room temperature. The chromogen substrate was 3,3′-diaminobenzidine tetrahydrochloride solution (Histofine Simple Stain DAB solution; Nichirei Co.). Subsequently, the sections were counterstained with Mayer’s hematoxylin (Wako, Tokyo, Japan). Negative controls were treated similarly, except for the antibodies being replaced by normal rabbit IgG (Santa Cruz Biotechnology, Inc.).

Interpretation of immunostaining results

The staining intensity was scored into four grades as follows: 0, no staining; 1, weakly positive; 2, moderately positive; and 3, strongly positive. The staining extent (positive frequency) was also scored into four grades according to the percentage of stained tumor cells as follows: 0, complete absence of staining; 1, ≤20%; 2, >20 to ≤50%; and 3, >50% stained cells. Composite scores were derived by addition of the intensity score and the staining extent score. For the statistical analysis, composite scores of ≥4 were defined as high expression and scores of <4 as low expression. Two investigators (Hideaki Murase and Yoko Takagi), who were blinded to the patients’ outcomes separately counted the stained cancer cells. Any disagreements between the two investigators were resolved by reassessment and consensus.

Statistical analysis

The statistical analysis was performed using IBM SPSS Statistics 20 software (IBM, Inc., Armonk, NY, USA). The χ2 test was used to investigate the possible associations between the expression of each FGFR receptor and clinicopathological variables. The χ2 test was also used to assess the correlations between FGFR expressions. The Mann-Whitney U test was used to analyze the associations between FGFR expression and patient age. Kaplan-Meier curves were plotted to assess the effects of FGFR expression on disease-specific survival (DSS) and different DSS curves were compared using the log-rank test. Multivariate proportional Cox models were used to assess the prognostic significance of FGFR and of factors associated with DSS. P<0.05 was considered to indicate a statistically significant difference.

Results

Immunohistochemical analysis of the FGFR family

Expression of FGFR1, FGFR2, FGFR3 and FGFR4 was mainly observed in the cytoplasm of cancer cells (Fig. 1) and fibroblasts in cancer tissue. Weak expression was observed in certain regions of the normal epithelium in proximity to the cancer cells. Among the 222 tumors investigated, the number of tumors exhibiting high FGFR expression were 66 (30%) for FGFR1, 114 (51%) for FGFR2, 142 (64%) for FGFR3 and 175 (79%) for FGFR4. High expression of FGFR1, FGFR2, FGFR3 or FGFR4 was significantly correlated with high expression of each of the other three proteins (Table I).

Table I

Correlations among the expressions of FGFR1, FGFR2, FGFR3 and FGFR4.

Table I

Correlations among the expressions of FGFR1, FGFR2, FGFR3 and FGFR4.

Expression levelFGFR2FGFR3FGFR4



LowHighP-valueLowHighP-valueLowHighP-value
FGFR1
 Low9462<0.00164920.02243113<0.001
 High14521650462
FGFR2
 Low49590.0053969<0.001
 High31838106
FGFR3
 Low26540.003
 High21121

[i] FGFR, fibroblast growth factor receptor.

Association with clinicopathological variables

The clinicopathological variables are summarized in Table II. A high expression of FGFR1, FGFR2 and FGFR4 was significantly associated with the depth of tumor invasion (T3–T4 vs. T1–T2: P<0.001, P=0.011 and P<0.001, respectively), lymph node metastasis (P=0.002, P=0.011 and P<0.001, respectively), and tumor stage (III–IV vs. I–II: P=0.001, P=0.012 and P<0.001, respectively). Distant metastasis or recurrence was found in a significantly higher proportion of patients with high expression of FGFR1, FGFR2 and FGFR4 compared to those with low expression of these proteins (P<0.001, P=0.004 and P<0.001, respectively). As the high expression of FGFR1, FGFR2 and FGFR4 was significantly associated with lymph node metastasis, we immunohistochemically evaluated the expression of these proteins in lymph node metastases from 88 patients and compared it to their expression in the primary tumor. A high expression of FGFR1, FGFR2 and FGFR4 was found in 61 (69%), 44 (50%), and 67 (76%) patients, respectively. However, only FGFR4 exhibited a significant association between its expression in the primary tumor and that in metastatic lymph nodes (P=0.017) (Table III).

Table II

Correlations of the expressions of FGFR1, FGFR2, FGFR3 and FGFR4 with clinicopathological factors.

Table II

Correlations of the expressions of FGFR1, FGFR2, FGFR3 and FGFR4 with clinicopathological factors.

Clinicopathological factorsnFGFR1FGFR2FGFR3FGFR4




Low (156)High (66)P-valueLow (108)High (114)P-valueLow (80)High (142)P-valueLow (47)High (175)P-value
Age (years)0.360.0360.380.006
 <70142103397765489438104
 ≥7080532731493248971
Gender1.001.001.000.70
 Female543816262819351044
 Male1681185082866110737131
Main location0.200.0290.390.84
 Middle or lower1771284993846111637140
 Upper452817153019261035
WHO pathological type0.470.0230.0020.74
 Differentiated1067729436327792185
 Undifferentiated1167937655153632690
Depth of invasion<0.0010.0110.58<0.001
 T1/21189622675145734177
 T3/4104604441633569698
Lymphatic invasion0.0010.0200.45<0.001
 Negative695910422722472643
 Positive15397566687589521132
Venous invasion0.0190.0040.66<0.001
 Negative735914462728453043
 Positive14997526287529717132
LN metastasis0.0020.0110.41<0.001
 Negative (N0)1149123654938763678
 Positive (N1/2/3)1086543436542661197
Stage0.0010.0120.47<0.001
 I/II14111031786348934299
 III/IV81463530513249576
Distant metastasis or recurrence<0.0010.0040.29<0.001
 Negative1521193384685110143109
 Positive70373324462941466

[i] FGFR, fibroblast growth factor receptor; WHO, World Health Organization; LN, lymph node.

Table III

Correlations of FGFR1, FGFR2, FGFR3 and FGFR4 expression between primary tumors and metastatic lymph nodes.

Table III

Correlations of FGFR1, FGFR2, FGFR3 and FGFR4 expression between primary tumors and metastatic lymph nodes.

Expression levelMetastatic lymph nodes

LowHighP-value
Primary tumor
 FGFR1
  Low17380.95
  High1023
 FGFR2
  Low21140.13
  High2330
 FGFR4
  Low530.017
  High1664

[i] FGFR, fibroblast growth factor receptor.

Association with DSS

High expression of FGFR1, FGFR2 and FGFR4 in the primary tumor was significantly associated with poorer DSS on the univariate analysis (P<0.001, P=0.008 and P<0.001, respectively) (Fig. 2). The 5-year DSS in patients with high expression of FGFR1, FGFR2 and FGFR4 was 57, 63 and 66%, respectively, compared to 77, 79 and 91%, respectively, in patients with low expression of these proteins (Table IV). On the multivariate analysis, the depth of tumor invasion and lymph node involvement were independent prognostic factors [hazard ratio (HR)=6.80, 95% confidence interval (CI): 2.63–15.6, P<0.001; and HR=4.48, 95% CI: 1.90–10.5, P=0.001, respectively], unlike FGFR1, FGFR2 and FGFR4 (HR=1.20, 95% CI: 0.73–2.00, P=0.47; HR=1.32, 95% CI: 0.77–2.24, P=0.31; and HR=0.96, 95% CI: 0.32–2.90, P=0.95, respectively).

Table IV

Prognostic factors in univariate and multivariate Cox proportional-hazards regression models for DSS in the study group as a whole.

Table IV

Prognostic factors in univariate and multivariate Cox proportional-hazards regression models for DSS in the study group as a whole.

Prognostic factorsUnivariate (log-rank)Multivariate


5-year DSS (%)P-valueHR95% CIP-value
Age (years)
 <7072
 ≥70690.37
Gender
 Female72
 Male710.81
Main location
 Middle or lower74
 Upper620.15
WHO pathological type
 Differentiated801
 Undifferentiated620.0041.420.84–2.400.18
Depth of invasion
 T1/2971
 T3/454<0.0016.802.63–15.6<0.001
LN metastasis
 Negative951
 Positive46<0.0014.481.90–10.50.001
FGFR1
 Low771
 High57<0.0011.200.73–2.000.47
FGFR2
 Low791
 High640.0081.320.77–2.240.31
FGFR3
 Low64
 High750.18
FGFR4
 Low911
 High66<0.0010.960.32–2.900.95

[i] FGFR, fibroblast growth factor receptor; DSS, disease-specific survival; LN, lymph node; HR, hazard ratio; CI, confidence interval.

Co-overexpression of FGFR1, FGFR2 and FGFR4

The co-overexpression of FGFR1, FGFR2 and FGFR4 in the primary tumors was found to be significantly associated with a poorer DSS compared to the expression of none or only one of these proteins (P<0.001 and P=0.001). The 5-year DSS was 55, 65, 78 and 92% in patients exhibiting high expression of all three, two, one and none of these FGFRs, respectively (Fig. 3). Although tumor stage was the most significant prognostic factor (HR=22.3, 95% CI: 10.1–49.6, P<0.001), the co-overexpression of these three FGFRs was also identified as an independent prognostic factor (HR=1.71, 95% CI: 1.02–2.85, P=0.041) (Table V).

Table V

Prognostic factors in multivariate Cox proportional-hazards regression models for disease-specific survival of patients with co-overexpression of FGFR1, FGFR2 and FGFR4.

Table V

Prognostic factors in multivariate Cox proportional-hazards regression models for disease-specific survival of patients with co-overexpression of FGFR1, FGFR2 and FGFR4.

Prognostic factorsMultivariate

HR95% CIP-value
WHO pathological type
 Differentiated1
 Undifferentiated1.290.77–2.190.33
Stage
 Stage I/II1
 Stage III/IV22.310.1–49.6<0.001
Co-overexpression of FGFR1, 2 and 4
 Others1
 All high1.711.02–2.850.041

[i] FGFR, fibroblast growth factor receptor; WHO, World Health Organisation; HR, hazard ratio; CI, confidence interval.

Discussion

Our results suggested that high expression of FGFR1, FGFR2 or FGFR4 may be crucial in tumor progression, metastasis and outcomes in gastric cancer patients. Moreover, the co-overexpression of FGFR1, FGFR2 and FGFR4 was found to be an independent prognostic factor in gastric cancer.

FGFR-dependent signaling occurs through two main pathways: via the intracellular receptor substrates FGFR substrate 2 (FRS2) and phospholipase Cg (PLCg), ultimately upregulating the Ras-dependent mitogen-activated protein kinase, and the Ras-independent phosphoinositide 3-kinase-Akt signaling pathways (15). Other pathways may also be activated by FGFRs, including STAT-dependent signaling (20). Although all four FGFRs generally signal through a similar network of pathways, a number of qualitative and quantitative differences have been identified and FGFR-specific differences in signaling pathways associated with genetic alterations of each FGFR have been confirmed in different types of cancer (2124).

FGFR1 amplification was previously identified in breast (25), ovarian (26), bladder (27) and lung cancer (28). In gastric cancer, a previous study reported the presence of FGFR1 amplifications in 12 (50%) of the 24 cases and FGFR1 protein was overexpressed in 37 (61%) of the 61 specimens on immunohistochemical analysis using a monoclonal antibody that differed from the one used in the present study. However, that study reported no significant correlation between FGFR1 expression and clinicopathological characteristics (17). To the best of our knowledge, the present study is the first to demonstrate that the high expression of FGFR1 is associated with poor survival in gastric cancer. The overexpression of FGFR1 was also found to be correlated with liver metastasis in colorectal cancer (29), whereas the amplification and overexpression of FGFR1 may contribute to poor outcomes in luminal-type breast cancer by driving anchorage-independent proliferation and resistance to endocrine therapy (25). The co-overexpression of FGF1 and FGFR1 has also been associated with poor outcomes in esophageal squamous cell carcinoma (30); however, we did not assess FGF expression in this study.

The FGFR4 Gly388Arg polymorphism has attracted considerable attention since the discovery of this germline polymorphism by Bange et al (31). In the human FGFR4 gene, a single-nucleotide polymorphism (SNP) from G to A at codon 388 at exon 9 changes the amino acid sequence of FGFR4 from glycine to arginine (Gly388 to Arg388). FGFR4 Gly388Arg was found to be associated with poor outcomes in breast (32), ovarian (33), lung (34) and gastric cancer (18). FGFR4 amplification was also found in pancreatic (35), renal cell (36) and gastric cancer (19). Ye et al (19) reported that the high expression of FGFR4 is associated with lymph node metastasis and a trend toward worse survival. Our findings are consistent with the findings of that study.

FGFR2 amplification was previously reported in gastric (37) and breast cancer (38), whereas FGFR2 missense mutations have been identified in gastric (39), lung (40), ovarian (41) and endometrial cancer (42), as well as in melanoma (43). FGFR2 genetic amplification or mutation leads to abnormal activation of the FGFR2 signaling pathway and contributes to carcinogenesis and tumor progression in gastric cancer. Overexpression of FGFR2 protein was detected on immunohistochemical staining in 20 of 38 diffuse-type gastric cancers, but in none of 11 intestinal-type lesions (44). FGFR2 amplifications were found in 10% of gastric cancers, the majority of which were of the undifferentiated type (11). Furthermore, FGFR2 amplification may correlate with poor outcomes in undifferentiated gastric cancer (12). In the present study, a high expression of FGFR2 was observed in undifferentiated as well as in differentiated-type gastric cancer. Although high expression of FGFR2 was not identified as an independent prognostic factor, it was significantly associated with poorer survival. Our findings are consistent with the results of previous studies.

FGFR3 amplification has been rarely reported in cancer (45,46). FGFR3 mutations have been identified in several types of cancer, including cervical cancer (47), multiple myeloma, prostate cancer (48) and spermatocytic seminomas (49). Bladder cancer exhibits the most clearly established association with FGFR3 mutations, which are strongly associated with low-grade non-invasive disease (50). Overexpression of FGFR3 was reportedly associated with low-stage bladder cancers on immunohistochemical analysis (51). However, overexpression of FGFR3 has also been associated with poor differentiation and high nuclear grade in hepatocellular carcinoma (52). The overexpression of FGFR3 in invasive breast cancer was not significantly associated with specific clinicopathological characteristics, although it was suggested to be a candidate marker for a poor prognosis (53). In this study, the expression of FGFR3 was not significantly associated with clinicopathological findings or survival. Shin et al (16) investigated the expression of the FGFRs in gastric cancer tissues and cell lines on northern blot analysis, ribonuclease protection assay and immunohistochemical analysis and reported that the mRNAs of FGFR1, FGFR2 and FGFR4 were upregulated in cancer tissues, whereas FGFR3 mRNA was not. These FGFR mRNAs were coexpressed in various combinations of two or three in the same tissue. The immunohistochemical analysis confirmed specific staining of multiple FGFRs, excluding FGFR3, in cancer specimens. In the present study, a high expression of FGFR3 was detected in addition to that of the other three FGFRs. The discrepancies among studies may be attributed to the differences in disease stage or the techniques used for immunohistochemical analysis.

We also evaluated the expression of FGFR1, FGFR2 and FGFR4 in metastatic lymph nodes. To the best of our knowledge, FGFRs in metastatic sites of gastric cancer had not been previously investigated. The expression of FGFR1 and FGFR2 differed between the primary tumors and lymph node metastases and were significantly correlated with the expression of only FGFR4. The differences in FGFR1 or FGFR2 expression among different tumor sites may represent a challenge regarding chemotherapy against these molecular targets.

FGFR-targeted therapeutics using small-molecule compounds that inhibit binding of FGF to FGFR is an active topic in the field of clinical oncology (54). Ki23057, a FGFR2 inhibitor, was reported to enhance the chemosensitivity of drug-resistant gastric cancer cells (55). Inhibition of FGFR2 signaling by AZD4547, a selective inhibitor of FGFR1, FGFR2 and FGFR3, was shown to significantly inhibit tumor growth in a dose-dependent manner in FGFR2-amplified xenografts (56). AZD4547 is currently being compared to paclitaxel as second-line treatment for patients with gastric cancer whose tumors exhibit FGFR2 gene amplification (NCT01457846, SHINE). Monoclonal antibodies that selectively recognize FGF or FGFR represent additional options for FGFR-targeted cancer therapy. Anti-FGFR2 monoclonal antibodies inhibit the in vivo growth of SNU-16 and OCUM-2M gastric cancer cells with FGFR2 gene amplification (13). Our results suggested that a selective inhibitor of FGFR1, FGFR2 and FGFR4 or the combined use of anti-FGFR1, -FGFR2 and -FGFR4 monoclonal antibodies may represent an effective FGFR-targeted therapy for gastric cancer.

In conclusion, high expression of FGFR1, FGFR2 or FGFR4 may be associated with tumor progression and poor survival in patients with gastric cancer. Similar to FGFR2, FGFR1 and FGFR4 may represent future prognostic factors and treatment targets in gastric cancer.

Acknowledgements

K.M., M.I. and K.S. were responsible for drafting the manuscript. K.M., K.K. and Y.T. contributed to the immunohistochemical analysis. M.I. and K.K. contributed to the analysis and interpretation of data.

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar

2 

Brenner H, Rothenbacher D and Arndt V: Epidemiology of stomach cancer. Methods Mol Biol. 472:467–477. 2009. View Article : Google Scholar

3 

Hartgrink HH, Jansen EP, van Grieken NC and van de Velde CJ: Gastric cancer. Lancet. 374:477–490. 2009. View Article : Google Scholar

4 

Cunningham D, Starling N, Rao S, et al: Capecitabine and oxaliplatin for advanced esophagogastric cancer. N Engl J Med. 358:36–46. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Bang YJ, Van Cutsem E, Feyereislova A, et al: Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 376:687–697. 2010. View Article : Google Scholar

6 

Gravalos C and Jimeno A: HER2 in gastric cancer: a new prognostic factor and a novel therapeutic target. Ann Oncol. 19:1523–1529. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Hofmann M, Stoss O, Shi D, et al: Assessment of a HER2 scoring system for gastric cancer: results from a validation study. Histopathology. 52:797–805. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Kimelman D and Kirschner M: Synergistic induction of mesoderm by FGF and TGF-beta and the identification of an mRNA coding for FGF in the early Xenopus embryo. Cell. 51:869–877. 1987. View Article : Google Scholar : PubMed/NCBI

9 

De Moerlooze L, Spencer-Dene B, Revest JM, Hajihosseini M, Rosewell I and Dickson C: An important role for the IIIb isoform of fibroblast growth factor receptor 2 (FGFR2) in mesenchymal-epithelial signalling during mouse organogenesis. Development. 127:483–492. 2000.PubMed/NCBI

10 

Turner N and Grose R: Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer. 10:116–129. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Kunii K, Davis L, Gorenstein J, et al: FGFR2-amplified gastric cancer cell lines require FGFR2 and Erbb3 signaling for growth and survival. Cancer Res. 68:2340–2348. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Toyokawa T, Yashiro M and Hirakawa K: Co-expression of keratinocyte growth factor and K-sam is an independent prognostic factor in gastric carcinoma. Oncol Rep. 21:875–880. 2009.PubMed/NCBI

13 

Zhao WM, Wang L, Park H, et al: Monoclonal antibodies to fibroblast growth factor receptor 2 effectively inhibit growth of gastric tumor xenografts. Clin Cancer Res. 16:5750–5758. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Zhao G, Li WY, Chen D, et al: A novel, selective inhibitor of fibroblast growth factor receptors that shows a potent broad spectrum of antitumor activity in several tumor xenograft models. Mol Cancer Ther. 10:2200–2210. 2011. View Article : Google Scholar

15 

Brooks AN, Kilgour E and Smith PD: Molecular pathways: fibroblast growth factor signaling: a new therapeutic opportunity in cancer. Clin Cancer Res. 18:1855–1862. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Shin EY, Lee BH, Yang JH, et al: Up-regulation and co-expression of fibroblast growth factor receptors in human gastric cancer. J Cancer Res Clin Oncol. 126:519–528. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Oki M, Yamamoto H, Taniguchi H, Adachi Y, Imai K and Shinomura Y: Overexpression of the receptor tyrosine kinase EphA4 in human gastric cancers. World J Gastroenterol. 14:5650–5656. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Ye Y, Shi Y, Zhou Y, et al: The fibroblast growth factor receptor-4 Arg388 allele is associated with gastric cancer progression. Ann Surg Oncol. 17:3354–3361. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Ye YW, Zhou Y, Yuan L, et al: Fibroblast growth factor receptor 4 regulates proliferation and antiapoptosis during gastric cancer progression. Cancer. 117:5304–5313. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Eswarakumar VP, Lax I and Schlessinger J: Cellular signaling by fibroblast growth factor receptors. Cytokine Growth Factor Rev. 16:139–149. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Vainikka S, Joukov V, Wennström S, Bergman M, Pelicci PG and Alitalo K: Signal transduction by fibroblast growth factor receptor-4 (FGFR-4). Comparison with FGFR-1. J Biol Chem. 269:18320–18326. 1994.PubMed/NCBI

22 

Wang JK, Gao G and Goldfarb M: Fibroblast growth factor receptors have different signaling and mitogenic potentials. Mol Cell Biol. 14:181–188. 1994.PubMed/NCBI

23 

Shaoul E, Reich-Slotky R, Berman B and Ron D: Fibroblast growth factor receptors display both common and distinct signaling pathways. Oncogene. 10:1553–1561. 1995.PubMed/NCBI

24 

Xian W, Schwertfeger KL and Rosen JM: Distinct roles of fibroblast growth factor receptor 1 and 2 in regulating cell survival and epithelial mesenchymal transition. Mol Endocrinol. 21:987–1000. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Turner N, Pearson A, Sharpe R, et al: FGFR1 amplification drives endocrine therapy resistance and is a therapeutic target in breast cancer. Cancer Res. 70:2085–2094. 2010. View Article : Google Scholar

26 

Theillet C, Adelaide J, Louason G, et al: FGFRI and PLAT genes and DNA amplification at 8p12 in breast and ovarian cancers. Genes Chromosomes Cancer. 7:219–226. 1993. View Article : Google Scholar : PubMed/NCBI

27 

Simon R, Richter J, Wagner U, et al: High-throughput tissue microarray analysis of 3p25 (RAF1) and 8p12 (FGFR1) copy number alterations in urinary bladder cancer. Cancer Res. 61:4514–4519. 2001.PubMed/NCBI

28 

Weiss J, Sos ML, Seidel D, et al: Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer. Sci Transl Med. 2:62ra932010.PubMed/NCBI

29 

Sato T, Oshima T, Yoshihara K, et al: Overexpression of the fibroblast growth factor receptor-1 gene correlates with liver metastasis in colorectal cancer. Oncol Rep. 21:211–216. 2009.PubMed/NCBI

30 

Sugiura K, Ozawa S, Kitagawa Y, Ueda M and Kitajima M: Co-expression of aFGF and FGFR-1 is predictive of a poor prognosis in patients with esophageal squamous cell carcinoma. Oncol Rep. 17:557–564. 2007.PubMed/NCBI

31 

Bange J, Prechtl D, Cheburkin Y, et al: Cancer progression and tumor cell motility are associated with the FGFR4 Arg(388) allele. Cancer Res. 62:840–847. 2002.PubMed/NCBI

32 

Thussbas C, Nahrig J, Streit S, et al: FGFR4 Arg388 allele is associated with resistance to adjuvant therapy in primary breast cancer. J Clin Oncol. 24:3747–3755. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Marmé F, Hielscher T, Hug S, et al: Fibroblast growth factor receptor 4 gene (FGFR4) 388Arg allele predicts prolonged survival and platinum sensitivity in advanced ovarian cancer. Int J Cancer. 131:E586–E591. 2012.PubMed/NCBI

34 

Frullanti E, Berking C, Harbeck N, et al: Meta and pooled analyses of FGFR4 Gly388Arg polymorphism as a cancer prognostic factor. Eur J Cancer Prev. 20:340–347. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Leung HY, Gullick WJ and Lemoine NR: Expression and functional activity of fibroblast growth factors and their receptors in human pancreatic cancer. Int J Cancer. 59:667–675. 1994. View Article : Google Scholar : PubMed/NCBI

36 

Takahashi A, Sasaki H, Kim SJ, et al: Identification of receptor genes in renal cell carcinoma associated with angiogenesis by differential hybridization technique. Biochem Biophys Res Commun. 257:855–859. 1999. View Article : Google Scholar : PubMed/NCBI

37 

Hattori Y, Odagiri H, Nakatani H, et al: K-sam, an amplified gene in stomach cancer, is a member of the heparin-binding growth factor receptor genes. Proc Natl Acad Sci USA. 87:5983–5987. 1990. View Article : Google Scholar : PubMed/NCBI

38 

Heiskanen M, Kononen J, Bärlund M, et al: CGH, cDNA and tissue microarray analyses implicate FGFR2 amplification in a small subset of breast tumors. Anal Cell Pathol. 22:229–234. 2001. View Article : Google Scholar : PubMed/NCBI

39 

Jang JH, Shin KH and Park JG: Mutations in fibroblast growth factor receptor 2 and fibroblast growth factor receptor 3 genes associated with human gastric and colorectal cancers. Cancer Res. 61:3541–3543. 2001.PubMed/NCBI

40 

Davies H, Hunter C, Smith R, et al: Somatic mutations of the protein kinase gene family in human lung cancer. Cancer Res. 65:7591–7595. 2005.PubMed/NCBI

41 

Byron SA, Gartside MG, Wellens CL, et al: FGFR2 mutations are rare across histologic subtypes of ovarian cancer. Gynecol Oncol. 117:125–129. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Pollock PM, Gartside MG, Dejeza LC, et al: Frequent activating FGFR2 mutations in endometrial carcinomas parallel germline mutations associated with craniosynostosis and skeletal dysplasia syndromes. Oncogene. 26:7158–7162. 2007. View Article : Google Scholar

43 

Gartside MG, Chen H, Ibrahimi OA, et al: Loss-of-function fibroblast growth factor receptor-2 mutations in melanoma. Mol Cancer Res. 7:41–54. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Hattori Y, Itoh H, Uchino S, et al: Immunohistochemical detection of K-sam protein in stomach cancer. Clin Cancer Res. 2:1373–1381. 1996.PubMed/NCBI

45 

Nord H, Segersten U, Sandgren J, et al: Focal amplifications are associated with high grade and recurrences in stage Ta bladder carcinoma. Int J Cancer. 126:1390–1402. 2010.PubMed/NCBI

46 

Khnykin D, Troen G, Berner JM and Delabie A: The expression of fibroblast growth factors and their receptors in Hodgkin’s lymphoma. J Pathol. 208:431–438. 2006.

47 

Rosty C, Aubriot MH, Cappellen D, et al: Clinical and biological characteristics of cervical neoplasias with FGFR3 mutation. Mol Cancer. 4:152005. View Article : Google Scholar : PubMed/NCBI

48 

Hernández S, de Muga S, Agell L, et al: FGFR3 mutations in prostate cancer: association with low-grade tumors. Modern Pathol. 22:848–856. 2009.PubMed/NCBI

49 

Goriely A, Hansen RMS, Taylor IB, et al: Activating mutations in FGFR3 and HRAS reveal a shared genetic origin for congenital disorders and testicular tumors. Nat Genet. 41:1247–1252. 2009. View Article : Google Scholar : PubMed/NCBI

50 

van Oers JM, Wild PJ, Burger M, et al: FGFR3 mutations and a normal CK20 staining pattern define low-grade noninvasive urothelial bladder tumours. Eur Urol. 52:760–768. 2007.

51 

Mhawrech-Fauceglia P, Cheney RT, Fischer G, Beek A and Herrmann FR: FGFR3 and p53 protein expressions in patients with pTa and pT1 urothelial bladder cancer. Eur J Surg Oncol. 32:231–237. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Qiu WH, Zhou BS, Chu PGG, et al: Over-expression of fibroblast growth factor receptor 3 in human hepatocellular carcinoma. World J Gastroenterol. 11:5266–5272. 2005.PubMed/NCBI

53 

Kuroso K, Imai Y, Kobayashi M, et al: Immunohistochemical detection of fibroblast growth factor receptor 3 in human breast cancer: correlation with clinicopathological/molecular parameters and prognosis. Pathobiology. 77:231–240. 2010. View Article : Google Scholar

54 

Katoh M and Nakagama H: FGF receptors: cancer biology and therapeutics. Med Res Rev. 34:280–300. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Qiu H, Yashiro M, Zhang X, Miwa A and Hirakawa K: A FGFR2 inhibitor, Ki23057, enhances the chemosensitivity of drug-resistant gastric cancer cells. Cancer Lett. 307:47–52. 2011. View Article : Google Scholar : PubMed/NCBI

56 

Xie L, Su X, Zhang L, et al: FGFR2 gene amplification in gastric cancer predicts sensitivity to the selective FGFR inhibitor AZD4547. Clin Cancer Res. 19:2572–2583. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-August 2014
Volume 2 Issue 4

Print ISSN: 2049-9450
Online ISSN:2049-9469

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Murase H, Inokuchi M, Takagi Y, Kato K, Kojima K and Sugihara K: Prognostic significance of the co‑overexpression of fibroblast growth factor receptors 1, 2 and 4 in gastric cancer. Mol Clin Oncol 2: 509-517, 2014
APA
Murase, H., Inokuchi, M., Takagi, Y., Kato, K., Kojima, K., & Sugihara, K. (2014). Prognostic significance of the co‑overexpression of fibroblast growth factor receptors 1, 2 and 4 in gastric cancer. Molecular and Clinical Oncology, 2, 509-517. https://doi.org/10.3892/mco.2014.293
MLA
Murase, H., Inokuchi, M., Takagi, Y., Kato, K., Kojima, K., Sugihara, K."Prognostic significance of the co‑overexpression of fibroblast growth factor receptors 1, 2 and 4 in gastric cancer". Molecular and Clinical Oncology 2.4 (2014): 509-517.
Chicago
Murase, H., Inokuchi, M., Takagi, Y., Kato, K., Kojima, K., Sugihara, K."Prognostic significance of the co‑overexpression of fibroblast growth factor receptors 1, 2 and 4 in gastric cancer". Molecular and Clinical Oncology 2, no. 4 (2014): 509-517. https://doi.org/10.3892/mco.2014.293