Association of amphiregulin with the cetuximab sensitivity of gastric cancer cell lines

  • Authors:
    • Julia Kneissl
    • Simone Keller
    • Thomas Lorber
    • Stefan Heindl
    • Gisela Keller
    • Ingo Drexler
    • Alexander Hapfelmeier
    • Heinz Höfler
    • Birgit Luber
  • View Affiliations

  • Published online on: May 14, 2012     https://doi.org/10.3892/ijo.2012.1479
  • Pages: 733-744
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The therapeutic activity of the epidermal growth factor receptor (EGFR)-directed monoclonal antibody cetuximab in gastric cancer is currently being investigated in clinical studies. Reliable biomarkers for the identification of patients who are likely to benefit from this treatment are not available. In this study, we assessed the activity of cetuximab in five gastric cancer cell lines (AGS, AZ521, Hs746T, LMSU and MKN1). The viability of two of these cell lines, AZ521 and MKN1, was significantly reduced by cetuximab treatment. High expression and secretion levels of the EGFR-binding ligand, amphiregulin (AREG), were associated with cetuximab responsiveness. MET activation and mutations in Kirsten-Ras gene (KRAS) were associated with cetuximab resistance. By introducing a hierarchy between these markers, we established a model that facilitated the correct classification of all five gastric cancer cell lines as cetuximab responsive or non-responsive. The highest priority was allocated to activating KRAS mutations, followed by MET activation and finally by the levels of secreted AREG. In order to validate these results, we used three additional human gastric cancer cell lines (KATOIII, MKN28 and MKN45). In conclusion, we propose that our model allows the response of gastric cancer cell lines to cetuximab treatment to be predicted.

Introduction

With approximately 738,000 deaths per year and an overall five-year survival rate below 30%, gastric cancer is the second most common cause of cancer mortality worldwide (13). This high level of mortality is due to the fact that most gastric cancer patients are diagnosed with locally advanced or metastatic disease. Therefore, the treatment options are limited, and new therapeutic approaches are urgently needed.

The dysregulation of the expression of the epidermal growth factor receptor (EGFR) is considered an important step in tumorigenesis. As EGFR is overexpressed in approximately 27 to 42% of gastric tumors (4,5), it represents an interesting target for therapeutic intervention. One therapeutic approach is the use of monoclonal antibodies that target EGFR. Cetuximab is one such monoclonal antibody and is approved for the treatment of recurrent colorectal cancer (CRC) and squamous cell carcinoma of the head and neck (SCCHN). Furthermore, a number of phase II studies on gastric cancer patients treated with various combinations of cetuximab with chemotherapy have shown promising results (69). A multinational phase III trial of cetuximab plus chemotherapy is on-going.

Experience with CRC and SCCHN therapies has shown that only a subgroup of patients benefits from cetuximab-based therapy. Well-known cetuximab resistance mechanisms include activating mutations in the Kirsten-Ras gene (KRAS), leading to a constantly activated EGFR pathway (10). Nevertheless, in many cases, the underlying resistance mechanism remains unclear. For optimal patient selection, it is therefore necessary to identify additional markers that are predictive of the cetuximab response.

On the molecular level, cetuximab inhibits EGFR activation by binding to the EGFR ligand binding site with a significantly higher affinity than the members of the family of EGFR-binding ligands (11). This group of EGFR-binding ligands consists of the epidermal growth factor (EGF), amphiregulin (AREG), epiregulin (EREG), epigen (EPGN), transforming growth factor α (TGFα), betacellulin (BTC) and heparin-binding EGF (HB-EGF) (1217). Ligand binding results in EGFR dimerization, the activation of its tyrosine kinase domain and subsequent signal transduction via a cascade of EGFR signaling pathway intermediates. Finally, processes that play an important role in tumorigenesis are activated; these processes include cell survival, proliferation and migration. In contrast to ligand binding, cetuximab binding results in EGFR internalization without further activation of the receptor and the downstream signaling pathway (18).

Previously, it was shown that AREG expression and EREG expression are positive predictive markers for the outcome of CRC patients treated with cetuximab in combination with chemotherapy (19,20). Among KRAS wild-type tumors, patients with high expression levels of AREG and EREG are highly likely to respond to these therapy regimens. In addition to KRAS, AREG and EREG, a predictive role in EGFR-inhibitory therapy has been discussed for EGFR, EGFR signaling intermediates, such as PI3K, PTEN or BRAF, and other EGFR ligands (21). A number of studies have suggested a negative predictive value for other receptor tyrosine kinases (RTKs), such as the hepatocyte growth factor receptor, MET (2224).

In gastric cancer, the search for predictive markers for the response to cetuximab-based therapies is on-going. As the prevalence of KRAS mutations in gastric cancer is low (25), a correlation between KRAS mutations and therapy response is difficult to establish. In contrast to CRC, no significant correlation between AREG expression and the response rate was found in gastric cancer patients treated with cetuximab in combination with oxaliplatin, leucovorin and 5-fluorouracil in a recent clinical phase II trial (26).

In the present study, we analyzed the cetuximab responsiveness of several gastric cancer cell lines (AGS, AZ521, Hs746T, LMSU and MKN1) and assessed the predictive value of EGFR and its ligands AREG and EGF alone and in combination with activating KRAS mutations and MET activation. We introduced a hierarchy between these markers and established a model that facilitates the correct classification of all five gastric cancer cell lines as cetuximab-responsive or -non-responsive. The model was validated with three other human gastric cancer cell lines, KATOIII, MKN28 and MKN45.

Materials and methods

Cell lines and cultivation conditions

The human gastric cancer cell lines AGS, KATOIII, MKN1, MKN28 and MKN45 were cultured in RPMI-1640 medium (Invitrogen/Gibco, Darmstadt, Germany) supplemented with 10% fetal bovine serum Sera Plus (PAN Biotech, Aidenbach, Germany), 2 mM l-glutamine (Invitrogen/Gibco) and penicillin-streptomycin (PAA Laboratories, Pasching, Austria; 100 international units (IU)/ml, 100 μg/ml) as reported previously (24). LMSU cells were grown in Nutrient Mixture F-10 Ham medium (Sigma-Aldrich Chemie GmbH, Steinheim, Germany), AZ521 cells were cultured in Minimum Essential Medium Eagle (MEM, Sigma-Aldrich Chemie GmbH), and Hs746T cells were grown in Dulbecco’s modified Eagle’s medium (DMEM) with GlutaMAX™ I (Invitrogen/Gibco), 4500 mg/l d-glucose and sodium pyruvate; all three media were supplemented with 10% fetal bovine serum Sera Plus and penicillin-streptomycin (100 IU/ml, 100 μg/ml). The cells were grown at 37°C in a humidified 5% CO2 atmosphere. The absence of mycoplasma was ensured in the conditioned medium after thawing frozen cells. Cell lines were used until passage 30.

Cell line source and cell validation testing

The AGS and KATOIII cells were obtained from the European Collection of Cell Cultures (ECACC), a Health Protection Agency Culture Collection Supplier of authenticated and quality controlled cell lines and nucleic acids (Porton Down, Salisbury, UK; http://www.hpacultures.org.uk/collections/ecacc.jsp). MKN1, AZ521 and LMSU cells were supplied by the Cell Bank RIKEN BioResource Center (Tsukuba, Japan). MKN28 cells were kindly provided by Dr V. Wachek (Medical University of Vienna, Vienna, Austria). MKN45 cells were obtained from Professor M. Ebert (Technische Universität München, Klinikum rechts der Isar, Munich, Germany). Hs746T cells were obtained from the ATCC Cell Biology Collection (LGC Standards GmbH, Wesel, Germany). The cell validation testing was performed as described previously (24). In addition, authentication of the Hs746T cell line was performed by short tandem repeat profiling using the Cell ID™ system (Promega, Mannheim, Germany).

XTT cell proliferation assay

The XTT cell proliferation kit II (Roche Diagnostics, Mannheim, Germany) was used according to the manufacturer’s instructions to assess growth inhibitory effects, as described previously (24). A modification of the US National Cancer Institute (NCI) protocol for in vitro cancer screens was used to determine cellular sensitivity to cetuximab (27,28). According to the individual doubling times of the different cell lines, cells were plated at densities between 1×103 and 4×103 cells per well in 80 μl of culture medium.

After 24 h of incubation at 37°C and 5% CO2, cetuximab (Merck, Darmstadt, Germany) was added at concentrations between 0 and 200 μg/ml in 20 μl of culture medium. A cetuximab concentration of 100 μg/ml is comparable to the active drug concentrations achieved in cancer patients (2931). After 48 h of incubation, 50 μl of XTT labeling mixture was added per well, and after 2 h at 37°C and 5% CO2, the absorbance of the samples was measured using a microplate reader (Asys Expert Plus, Biochrom, Berlin, Germany).

DNA isolation and sequencing

DNA isolation was performed using a standard protocol. Cells (1×106) were harvested, washed twice with phosphate-buffered saline (PBS) [centrifugation at 1,000 rounds per minute (rpm), 3 min] and transferred to 1.5 ml reaction tubes. The cell sediment was resuspended in 200 μl of 50 mM Tris-HCl pH 8.5, 1 mM EDTA, 0.5 % Tween-20 and 0.2 mg/ml proteinase K. After a 3-h incubation at 55°C, proteinase K was inactivated by boiling for 10 min, and aliquots were used for the mutation analysis.

The sequences of the primers used for the polymerase chain reaction (PCR) were as follows (from 5′ to 3′): BRAF forward (F), ACAGTAAAAATAGGTGATTTTGGTCTAGCTACAGA; BRAF reverse (R), CTATGAAAATACTATAGTTGAGACCTTCAATGACTTTC; EGFR exon 18 F, AGGGCTGAGGTGACCCTTGT; EGFR exon 18 R, TCCCCACCAGACCATGAGAG; EGFR exon 19 F, GCACCATCTCACAATTGCCAGTTA; EGFR exon 19 R, AAAAGGTGGGCCTGAGGTTCA; EGFR exon 21 F, CCTCACAGCAGGGTCTTCTCTGT; EGFR exon 21 R, TCAGGAAAATGCTGGCTGACCTA; KRAS exon 2 F, GGTGGAGTATTTGATAGTGTATTAACC; KRAS exon 2 R, CCTCTATTGTTGGATCATATTCG; PIK3CA exon 9 F, TTGCTTTTTCTGTAAATCATCTGTG; PIK3CA exon 9 R, CTGCTTTATTTATTCCAATAGGTATG; PIK3CA exon 20 F, TGACATTTGAGCAAA GACCTG; PIK3CA exon 20 R, CCTATGCAATCGGTCTTTGC.

The BRAF hotspot mutation, V600E, was analyzed using allele-specific PCR following an established protocol (32). Positive and negative controls that had been verified by direct sequencing were included in this analysis. The primers for the EGFR mutation analysis were described previously (33,34). The KRAS mutation analysis was performed as reported previously (7). The EGFR, KRAS and PIK3CA mutation analysis was performed using PCR amplification followed by direct sequencing.

DNA sequencing analysis was performed using the BigDye Terminator v1.1 Cycle Sequencing kit (Applied Biosystems, Foster City, CA, USA), and the products were separated using an automated sequencing system (3130 Genetic Analyzer, Applied Biosystems).

Enzyme-linked immunosorbent assay (ELISA)

For the ELISA-based detection of AREG and EGF, DuoSet ELISA kits (R&D Systems, Minneapolis, MN, USA) were used according to the manufacturer’s instructions. The ligand concentrations were measured in the conditioned medium and in the cellular extract.

For the determination of the AREG and EGF levels in the conditioned medium of the cell cultures, 1×106 cells were seeded into cell culture plates (10 cm in diameter) and cultured in 10 ml of medium. EGF and cetuximab were added 2 h after the cultures were seeded. At the indicated time-points, the conditioned medium was harvested and centrifuged to remove the cell debris (13,000 rpm, 4°C, 10 min). The supernatant was used for subsequent analysis.

For the determination of AREG and EGF levels in the cellular extract, 5×105 cells (AGS, AZ521), 8×105 cells (MKN28) or 1×106 cells (Hs746T, KATOIII, LMSU, MKN1 and MKN45) were seeded as described above and cultured for 48 h. Subsequently, the cells were lysed in 350 μl of lysis buffer (7X lysis buffer: 20 mM Tris-HCl, pH 7.5; 1 mM EDTA; Complete mini protease inhibitor cocktail tablets, Roche Applied Science, concentration according to the manufacturer’s instructions) and sonicated (25 sec, amplitude 70%). The cell debris was removed by centrifugation as described above. A volume of 100 μl of total protein (100 μg/ml) was used for each sample.

Western blot analysis

The expression levels of EGFR, phosphorylated EGFR (Y1068) and phosphorylated MET (Y1234/1235) were determined using a standard protocol. For the western blot analysis, cells were seeded at a density of 1×106 cells per 10-cm tissue culture dish. After 48 h, the cells were washed with ice-cold PBS and lysed on ice with 150 μl of L-CAM lysis buffer as described previously (35). Between 10 and 30 μg of total protein were separated by SDS-polyacrylamide gel electrophoresis and transferred to PVDF membranes (GE Healthcare, Munich, Germany, no. RPN303F).

After 1 h of blocking, the membranes were probed with appropriate primary antibodies overnight at 4°C: anti-EGFR rabbit polyclonal antibody (no. 2232, Cell Signaling Technology, distributed by New England Biolabs, Frankfurt, Germany; dilution 1:2,000), anti-phosphorylated EGFR (pEGFR) rabbit polyclonal antibody directed against tyrosine residue 1068 (no. 44788G, Invitrogen, Karlsruhe, Germany; dilution 1:2,000), anti-phosphorylated MET (pMET) rabbit monoclonal antibody directed against tyrosine residues 1234 and 1235 (no. 3077, Cell Signaling Technology, dilution 1:1,000), anti-α-tubulin mouse monoclonal antibody (no. T6199, Sigma-Aldrich Chemie GmbH; dilution 1:10,000) and anti-β-actin mouse monoclonal antibody (no. A1978, Sigma-Aldrich Chemie GmbH; dilution 1:5,000).

Detection was performed using horseradish peroxidase-conjugated secondary antibodies by enhanced chemiluminescence (Amersham, Braunschweig, Germany). For signal quantification, blots were scanned and densitometrically analyzed using Image J software 1.42q (National Institute of Health, MD, USA).

Flow cytometry analysis

Cells were seeded at densities of 3×105 to 1×106 cells per 10-cm tissue dish and cultured for 72 h. After washing with PBS (without Ca2+/Mg2+), the cells were detached using 1 ml of Versene (Invitrogen/Gibco) for 10–15 min at 37°C. Detached cells were resuspended in ice-cold FACS buffer [1% (w/v) bovine serum albumin (BSA; Sigma-Aldrich Chemie GmbH) in PBS (without Ca2+/Mg2+)]. After two additional washing steps [each comprising i) resuspension of the cells in FACS buffer, ii) sedimentation at 300 g (4°C) for 3 min and iii) discarding the supernatant], the cells (1×105) were incubated with monoclonal antibody directed against EGFR (Ab-1, clone 528, Thermo Fisher Scientific, Ulm, Germany; 2.5 μg/ml) in 50 μl of FACS buffer in 96-well microtiter plates. Additionally, an isotype control antibody [IgG2a, clone PPV-04, Exbio, Prague, Czech Republic, distributed by Biozol (Eching, Germany; 2.5 μg/ml)] was routinely applied. After incubation for 1 h at 4°C in the dark, the cells were washed twice as described above and incubated with 50 μl of secondary antibody solution [Alexa Fluor 647 F(ab)2-fragment (H+L), Invitrogen, 5 μg/ml] for 1 h (4°C) in the dark. Two washing steps were performed to remove unbound secondary antibodies. The cells were then resuspended in 300 μl of FACS buffer and subsequently analyzed using a FACSCanto flow cytometer (BD Bioscience, Heidelberg, Germany). For live-dead discrimination, propidium iodide was added to each sample at a final concentration of 1 μg/ml directly before measurement.

Statistical analysis

The data are presented as the means ± standard deviation (SD). Pairwise comparisons of samples were performed by two-sided Welch’s t-tests. In the XTT cell proliferation assay, one sample t-tests were used to test the activity ratio of treated samples to untreated samples against a reference value of 100% which indicates equality of activity. All analyses were performed on an explorative significance level of 0.05 using the statistical software package R (The R Foundation for Statistical Computing, Vienna, Austria). P-values at a significance level of <0.05 are indicated by (*) and <0.01 by (**). A summary of all statistical data is available from the authors upon request.

Results

Cetuximab responsiveness of gastric cancer cell lines

The EGFR signaling pathway is involved in the regulation of tumor cell proliferation. To determine the cetuximab responsiveness of a panel of five human gastric cancer cell lines (AGS, AZ521, Hs746T, LMSU and MKN1), cells were treated with varying concentrations of the therapeutic antibody (0–200 μg/ml cetuximab). The metabolic activity of the cell lines as a surrogate marker for cell viability was analyzed with the XTT cell viability assay.

The concentration-response curves indicate that the metabolic activities of AZ521 and MKN1 cells were significantly reduced by cetuximab in a concentration-dependent manner (Fig. 1). Significant effects on MKN1 cells were observed at a cetuximab concentration of 1 μg/ml and on AZ521 cells at a cetuximab concentration of 100 μg/ml. The cell lines, AGS, LMSU and Hs746T, were not cetuximab-responsive.

Based on these results, the gastric cancer cell lines, AZ521 and MKN1, were classified as cetuximab-responsive, whereas the other considered cell lines (AGS, Hs746T and LMSU) were regarded as cetuximab-resistant.

Association of cetuximab responsiveness with the AREG and EGF levels

Previously, a correlation between the levels of EGFR ligands and the response to cetuximab-based therapy was demonstrated. The ligand, AREG, was shown to have a high predictive value for the response to EGFR inhibitory therapy in CRC patients (19), whereas a low EGF level was associated with a higher probability to respond to cetuximab plus chemotherapy in gastric cancer patients (26). To clarify the predictive role of EGFR ligands in our gastric cancer model, we determined the amounts of AREG and EGF secreted into the conditioned medium and in the cellular extract by ELISA. Using this approach, it was possible to distinguish between proteolytically released soluble ligands and membrane-bound ligands.

As shown in Fig. 2, AREG was expressed and secreted by several cell lines. AZ521 cells displayed the highest amount of AREG, with a mean concentration of 1,375 pg/ml in the conditioned medium and 1,017 pg/mg in the cellular extract. A high level of AREG was also detected in the MKN1 cells (519 pg/ml in the conditioned medium and 746 pg/mg in the cellular extract). In comparison, only low amounts of AREG were observed for the cell line Hs746T. In these cells, the AREG concentrations were 135 pg/ml in the conditioned medium and 100 pg/mg in the cellular extract, levels 10-fold lower than those in the AZ521 cells. It was not possible to detect significant concentrations of AREG in the conditioned media of the AGS and LMSU cell cultures. Furthermore, analysis of the cellular extract revealed only marginal amounts of the ligand in these two cell lines.

EGF was not found in significant concentrations in the cell extracts or in the conditioned medium of most of the cell lines. Only in MKN1 cells was a low amount of EGF detected in the cellular extract; this EGF level was very close to the detection limit of the assay.

These findings indicate that AREG is expressed in the cetuximab-sensitive cell lines, AZ521 and MKN1, suggesting a positive predictive value of the ligand in gastric cancer cells. By contrast, due to the low detected EGF levels, no correlation could be established between the presence of EGF and the cetuximab responsiveness of the cell lines that were studied.

Effect of cetuximab treatment on AREG levels in AZ521 and MKN1 cells

To examine the role of the EGFR signaling pathway in the regulation of AREG, the levels of secreted AREG were determined after the treatment of cells with cetuximab or combinations of EGF and cetuximab. The AREG concentrations were determined in the conditioned medium of the cetuximab-sensitive cell lines, AZ521 and MKN1, using ELISA.

As shown in Fig. 3, cetuximab treatment caused a significant decrease in the level of secreted AREG in the MKN1 cells, but no such decrease was observed in the AZ521 cells. In both cell lines, the addition of exogenous EGF to the cells resulted in an increase in AREG secretion. This EGF-induced AREG secretion was blocked by simultaneous treatment of the cells with increasing concentrations of cetuximab to some extent in the MKN1 cells and almost completely in the AZ521 cells. By contrast, Hs746T cells did not respond to EGF or cetuximab treatment (data not shown).

These results demonstrate that the EGFR signaling pathway itself is involved in the regulation of the ligand AREG. This important finding suggests that there exists an autoregulatory mechanism in gastric cancer cell lines.

Analysis of EGFR expression, localization and activation

Ligand binding results in EGFR dimerization, stimulation of its tyrosine kinase activity and activation of downstream signaling cascades. The total and activated levels of EGFR were determined in the five gastric cancer cell lines using immunoblot and flow cytometry analyses.

The analysis of the expression level of EGFR in the different cell lines by western blotting revealed the following order: MKN1 > LMSU = Hs746T > AGS = AZ521 (Fig. 4A). Essentially the same order was obtained when the surface localization of EGFR was analyzed with flow cytometry (Fig. 4B).

The activation level of EGFR was determined by western blot analysis of the level of EGFR phosphorylation on tyrosine residue Y1068 (Fig. 4C). The following order of the EGFR activation levels was obtained: Hs746T >> AZ521 = MKN1 > AGS = LMSU. Together, these findings demonstrate that the studied cell lines express EGFR at considerably different levels and that the expression and activation levels of EGFR are not correlated.

Effect of EGF and cetuximab treatment on the phosphorylation of EGFR

The effects of EGF and cetuximab on the expression and activation of EGFR were determined using western blot analysis.

The detection of EGFR phosphorylated on tyrosine residue Y1068 revealed that EGFR was activated by EGF in the AGS, AZ521, LMSU and MKN1 cell lines and that this EGF-induced activation of EGFR was blocked by cetuximab in a concentration-dependent manner (Fig. 5A, B, D and E). By contrast, Hs746T cells were EGF- and cetuximab-non-responsive (Fig. 5C). In all cell lines, the EGFR expression levels remained essentially unchanged during all treatment conditions.

Together, EGF and/or cetuximab had only minor effects on the degree of EGFR phosphorylation in the Hs746T cell line, whereas EGFR activation was modulated by the treatment in the four other cell lines (AGS, AZ521, LMSU and MKN1).

Analysis of the MET activation

A number of studies have suggested a predictive role for MET in EGFR inhibitory therapy (3638). To determine the role of MET activation in our gastric cancer model, the phosphorylation status of the MET receptor was determined using western blot analysis. Detection of MET phosphorylated on tyrosine residue Y1234/1235 revealed that the activation of MET was very strong in Hs746T cells, whereas no signals were detected in the AGS, AZ521, LMSU and MKN1 cell lines (Fig. 6).

Mutation analysis

EGFR, KRAS, BRAF and PI3K are key components of the EGFR-signaling pathway, and oncogenic alterations in these genes are related to the response to EGFR-targeting therapeutics. Therefore, hotspot mutation regions in these genes were analyzed in the panel of gastric cancer cell lines.

In detail, for BRAF, the cell lines were screened for the activating mutation V600E. Exons 18, 19 and 21 of EGFR, exon 2 of KRAS and exons 9 and 20 of PIK3CA were sequenced.

The presence of the KRAS mutation G12D in AGS cells has been reported previously, and we confirmed this finding. We also confirmed the presence of the recently described PIK3CA mutation, E545K, in MKN1 cells. No further genetic alterations were identified (Table I).

Table I

Genetic alterations in hotspot mutation regions of key components of the EGFR signaling pathway in gastric cancer cell lines.

Table I

Genetic alterations in hotspot mutation regions of key components of the EGFR signaling pathway in gastric cancer cell lines.

MutationAGSAZ521Hs746TLMSUMKN1
EGFRaWTWTWTWTWT
KRASbG12DcWTWTWTWT
PIK3CAdWTWTWTWT E545Ke
BRAFfWTWTWTWTWT

a Exons 18, 19 and 21;

b exon 2;

c previously described by Kim et al (47);

d exons 9 and 20;

e previously described by the Sanger Institute (www.sanger.ac.uk);

f V600E. WT, wild-type. Bold letters indicate the mutations: KRAS G12D in AGS cells and PIK3CA E545K in MKN1 cells.

Model for predicting cetuximab sensitivity in gastric cancer cell lines

In our study, we found that the gastric cancer cell lines, AZ521 and MKN1, were cetuximab-sensitive, whereas the AGS, Hs746T and LMSU cell lines were found to be cetuximab-resistant based on the results of the XTT cell viability assay.

The cetuximab responsiveness of the AZ521 and MKN1 cells was associated with AREG expression and secretion. The cetuximab resistance of the AGS cells is most likely due to the KRAS mutation, whereas the cetuximab insensitivity of Hs746T cells may be explained by MET activation. Together, we were able to explain the cetuximab response of four of the five investigated cell lines.

We propose the model presented in Fig. 7 to predict the cetuximab response of gastric cancer cell lines. To validate this model, we measured the amount of secreted AREG in three additional human gastric cancer cell lines, KATOIII (523 pg/ml in the conditioned medium and 927 pg/mg in the cellular extract), MKN28 (918 pg/ml in the conditioned medium and 106 pg/mg in the cellular extract) and MKN45 (7 pg/ml in the conditioned medium and 168 pg/mg in the cellular extract) (Table II). Among these three cell lines, there was one cetuximab-responsive cell line (MKN28) and two cetuximab-non-responsive cell lines (KATOIII, MKN45) (24). These cell lines have previously been characterized with respect to the KRAS mutation and MET activation status (24). MET has been shown to be activated in the cetuximab-resistant cell lines, KATOIII and MKN45, but not in the MKN28 cell line (24). All three cell lines were correctly classified by the model.

Table II

Molecular characteristics of the gastric cancer cell lines.

Table II

Molecular characteristics of the gastric cancer cell lines.

Cell line KRAS-statusMET status
AREG secretionCetuximab sensitivitya
cMET amplificationMET activation
AGSG12D (25) (39)
AZ521WT (48) (39)++++++
Hs746TWT++ (39)++++
KATOIIIWT (48)+ (49)+ (24)++ (24)
LMSUWTND
MKN1WT (48) (39,49)++++
MKN28WT (48) (49) (24)+++++ (24)
MKN45WT (48)++ (39,49)+++ (24) (24)

a Determined via XTT cell proliferation assay.

−, Not detectable;

+/++/+++, detectable at low/intermediate/high levels. ND, not done; WT, wild-type. Bold letters indicate the mutation: KRAS G12D in AGS cells.

Discussion

Responsiveness of gastric cancer cell lines to cetuximab treatment

In this study, the predictive value of several molecular markers for the cetuximab responsiveness of the human gastric cancer cell lines, AGS, AZ521, Hs746T, LMSU and MKN1, was analyzed. The analysis of cell viability after cetuximab treatment identified two cetuximab-responsive cell lines, AZ521 and MKN1, whose metabolic activity was significantly reduced by cetuximab in a dose-dependent manner. The other three cell lines, AGS, Hs746T and LMSU, were not responsive to cetuximab. For the MKN1 and AGS cells, these results are in agreement with the results reported previously by our group (24). All considered cell lines expressed EGFR at different levels.

Model for the prediction of cetuximab responsiveness

In the present study, we performed a detailed molecular analysis of different putative predictive markers for cetuximab responsiveness, including the expression and secretion of the ligands, AREG and EGF, activation of the RTK, MET, and the presence of activating mutations in KRAS.

The cetuximab responsiveness of AZ521 and MKN1 cells was associated with AREG expression and secretion. By contrast, the cetuximab resistance of AGS cells was most likely due to the KRAS mutation (24). As a number of studies have suggested an association between MET activation and resistance to cetuximab (2224), one possible explanation for the cetuximab insensitivity of Hs746T cells is the high level of MET tyrosine kinase activity. Notably, the Hs746T cell line harbors a splice site mutation in cMET, resulting in the deletion of the juxtamembrane domain (39).

We established a model to facilitate the correct classification of gastric cancer cell lines as cetuximab-responsive and -non-responsive cell lines. In an attempt to establish a hierarchy of predictive molecular markers, the highest priority was allocated to activating KRAS mutations. This decision was based on the experience that patients with colorectal cancers lacking oncogenic activation of the EGFR downstream effectors, KRAS, BRAF, PIK3CA and PTEN, are the most likely to benefit from anti-EGFR therapies (21). The second place in the hierarchy was assigned to MET activation. Finally, gastric cancer cell lines lacking activated KRAS mutations and MET activation were classified according to their level of secreted AREG. Using this approach, it was possible to correctly classify the cetuximab responsiveness of all five cell lines included in this study (AGS, AZ521, Hs746T, LMSU and MKN1). The reason for the cetuximab resistance of LMSU cells is presently unknown.

The model was validated with three other human gastric cancer cell lines, KATOIII, MKN28 and MKN45, among which one was cetuximab-responsive (MKN28) and two were cetuximab-non-responsive (KATOIII and MKN45) (24). None of these three cell lines harbors a known activating KRAS mutation (24). We have previously shown that MET is activated in the cetuximab-resistant cell lines, KATOIII and MKN45, but not in the MKN28 cell line (24). In the present study, we detected high levels of secreted AREG in the MKN28 cells and used the model to classify this cell line as cetuximab-responsive. AREG secretion was not detectable in the cetuximab-resistant cell line MKN45, which falls into the non-responder category due to the elevated MET activation status. Despite the high levels of secreted AREG, the KATOIII cell line was classified as non-responsive due to the high level of activated MET.

In this study, the advantage of the model as a tool to determine the cetuximab sensitivity of gastric cancer cell lines becomes evident. However, it also becomes clear that it is difficult to predict the therapy response by evaluation of a single marker. In order to ensure a correct classification, we suggest defining a panel of predictive markers and establishing a hierarchy among them. Of course, we make no claim of completeness as regards this study.

Positive predictive role of AREG secretion for cetuximab responsiveness

In the present study, we found a positive predictive value of AREG secretion for cetuximab responsiveness when the KRAS mutation status and MET activation were taken into account.

This finding is in agreement with the positive predictive role that was attributed to AREG and EREG as markers for the outcome of CRC patients treated with cetuximab and chemotherapy who have no KRAS mutations in their tumors (19,20). In contrast to the situation in CRC, in 38 advanced gastric cancer patients treated with cetuximab in combination with oxaliplatin, leucovorin and 5-fluorouracil in a recent clinical phase II trial, no significant correlation between AREG expression and the response rate was found (26). Notably, the level of activated MET was not determined in that study.

One main technical difference between the CRC study and the gastric cancer study is that the AREG mRNA expression level was examined in the CRC tumors, whereas the serum levels of AREG were determined in the gastric cancer patients. When the correlation between tumor mRNA expression and the level of protein in the blood (measured by ELISA) was assessed, only a modest correlation between the systemic protein and tumor mRNA levels of AREG was found (19).

Cetuximab is approved for the treatment of recurrent SCCHN in addition to CRC. For these tumors, the findings regarding the predictive value for AREG are contradictory. In one study, high EGFRvIII and AREG expression levels identified SCCHN patients who were less likely to benefit from combination treatment with cetuximab and docetaxel (40). In a different study that included SCCHN tumors and cell lines, autocrine production of AREG was found to predict sensitivity to both gefitinib and cetuximab in EGFR wild-type cancers (41).

A number of studies have suggested that AREG produced by tumor cells may be involved in the pathogenesis and/or progression of human gastric carcinoma (42,43). Notably, 20 out of 32 tumors (62.5%) expressed AREG mRNA at higher levels than their corresponding normal mucosas. By contrast, no obvious correlation was observed between the AREG mRNA levels and the histological types or tumor staging of gastric carcinoma (43). Additionally, an association between AREG expression and the development of peritoneal carcinomatosis in gastric cancer patients was reported (42).

Recently, it was shown that low levels of the EGFR ligands, EGF and TGFα, in combination with EGFR expression positively correlated with the response rates of gastric cancer patients to a cetuximab/modified FOLFOX6 regimen (26). Considering the lack of EGF expression in both cetuximab-sensitive and cetuximab-resistant gastric cancer cell lines, we conclude that EGF expression is not of predictive value in our study. This lack of soluble EGF suggests that although EGF is expressed in all cell lines to a certain extent, it is not proteolytically released. Previously, it was proposed that the proteolytic release of EGF is essential for its activity as it cannot act in a juxtacrine manner (44). Due to the absence of soluble EGF, the EGF-based induction of the EGFR signaling pathway most likely plays no role or only a minor role in our gastric cancer cell lines.

Association of MET activation and cetuximab resistance

As mentioned above, a number of studies have suggested a predictive role for MET in EGFR inhibitory therapy (3638,44,45). As reported previously, the activation of MET was accompanied with the activation of EGFR in the KATOIII and MKN45 cell lines (24). A similar observation was made in the present study: Hs746T cells showed high levels of MET and EGFR activation. Notably, cetuximab had only minor effects on the degree of EGFR phosphorylation in this cell line, whereas EGFR activation was modulated by the treatment in the four other considered cell lines (AGS, AZ521, LMSU and MKN1).

As discussed previously (24), there is a close correlation between MET and EGFR, including physical interaction and ligand-dependent or -independent transactivation, and the co-activation of multiple RTKs in cancer cells results in resistance to single-agent therapy (37,46). Consequently, the co-activation of MET would explain the failure of anti-EGFR therapy in the non-responsive cell lines, Hs746T, KATOIII and MKN45.

In conclusion, in this study, to our knowledge, we present the first model that allows the response of gastric cancer cell lines to cetuximab treatment to be predicted. The considered markers were the status of KRAS mutation, MET activation and AREG secretion. Even if cell culture models are too simple to explain the complex in vivo situation in patients, we believe that the inclusion of a reasonable number of cell lines and accurate statistical analysis will allow for the generation of hypotheses that can be tested in clinical studies.

Abbreviations:

AREG

amphiregulin

CRC

colorectal cancer

DMEM

Dulbecco’s modified Eagle’s medium

ECACC

European Collection of Cell Cultures

EGF

epidermal growth factor

EGFR

epidermal growth factor receptor

ELISA

enzyme-linked immunosorbent assay

EREG

epiregulin

IU

international units

KRAS

Kirsten-Ras gene

MEM

Minimum Essential Medium Eagle

PBS

phosphate-buffered saline

PCR

polymerase chain reaction

rpm

rounds per minute

RTK

receptor tyrosine kinase

SCCHN

squamous cell carcinoma of the head and neck

SD

standard deviation

Acknowledgements

This study was supported by the German Federal Ministry of Education and Research and the Austrian Federal Ministry for Science and Research as part of the program ‘Medizinische Systembiologie-MedSys’ (CANCERMOTISYS project, www.cancermotisys.eu). We thank Birgit Geist and Susanne Plaschke for their excellent technical assistance.

References

1 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Wang SJ, Emery R, Fuller CD, Kim JS, Sittig DF and Thomas CR: Conditional survival in gastric cancer: a SEER database analysis. Gastric Cancer. 10:153–158. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Verdecchia A, Santaquilani M and Sant M: Survival for cancer patients in Europe. Ann Ist Super Sanita. 45:315–324. 2009.PubMed/NCBI

4 

Kim MA, Lee HS, Lee HE, Jeon YK, Yang HK and Kim WH: EGFR in gastric carcinomas: prognostic significance of protein overexpression and high gene copy number. Histopathology. 52:738–746. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Liang Z, Zeng X, Gao J, et al: Analysis of EGFR, HER2, and TOP2A gene status and chromosomal polysomy in gastric adenocarcinoma from Chinese patients. BMC Cancer. 8:3632008. View Article : Google Scholar : PubMed/NCBI

6 

Pinto C, Di Fabio F, Barone C, et al: Phase II study of cetuximab in combination with cisplatin and docetaxel in patients with untreated advanced gastric or gastro-oesophageal junction adenocarcinoma (DOCETUX study). Br J Cancer. 101:1261–1268. 2009. View Article : Google Scholar

7 

Lordick F, Luber B, Lorenzen S, et al: Cetuximab plus oxaliplatin/leucovorin/5-fluorouracil in first-line metastatic gastric cancer: a phase II study of the Arbeitsgemeinschaft Internistische Onkologie (AIO). Br J Cancer. 102:500–505. 2010. View Article : Google Scholar

8 

Kim C, Lee JL, Ryu MH, et al: A prospective phase II study of cetuximab in combination with XELOX (capecitabine and oxaliplatin) in patients with metastatic and/or recurrent advanced gastric cancer. Invest New Drugs. 29:366–373. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Luber B, Deplazes J, Keller G, et al: Biomarker analysis of cetuximab plus oxaliplatin/leucovorin/5-fluorouracil in first-line metastatic gastric and oesophago-gastric junction cancer: results from a phase II trial of the Arbeitsgemeinschaft Internistische Onkologie (AIO). BMC Cancer. 11:5092011. View Article : Google Scholar

10 

Lievre A, Bachet JB, Le Corre D, et al: KRAS mutation status is predictive of response to cetuximab therapy in colorectal cancer. Cancer Res. 66:3992–3995. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Goldstein NI, Prewett M, Zuklys K, Rockwell P and Mendelsohn J: Biological efficacy of a chimeric antibody to the epidermal growth factor receptor in a human tumor xenograft model. Clin Cancer Res. 1:1311–1318. 1995.PubMed/NCBI

12 

Harris RC, Chung E and Coffey RJ: EGF receptor ligands. Exp Cell Res. 284:2–13. 2003. View Article : Google Scholar

13 

Higashiyama S, Abraham JA, Miller J, Fiddes JC and Klagsbrun M: A heparin-binding growth factor secreted by macrophage-like cells that is related to EGF. Science. 251:936–939. 1991. View Article : Google Scholar : PubMed/NCBI

14 

Komurasaki T, Toyoda H, Uchida D and Morimoto S: Epiregulin binds to epidermal growth factor receptor and ErbB-4 and induces tyrosine phosphorylation of epidermal growth factor receptor, ErbB-2, ErbB-3 and ErbB-4. Oncogene. 15:2841–2848. 1997. View Article : Google Scholar : PubMed/NCBI

15 

Shoyab M, Plowman GD, McDonald VL, Bradley JG and Todaro GJ: Structure and function of human amphiregulin: a member of the epidermal growth factor family. Science. 243:1074–1076. 1989. View Article : Google Scholar : PubMed/NCBI

16 

Strachan L, Murison JG, Prestidge RL, Sleeman MA, Watson JD and Kumble KD: Cloning and biological activity of epigen, a novel member of the epidermal growth factor superfamily. J Biol Chem. 276:18265–18271. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Watanabe T, Shintani A, Nakata M, et al: Recombinant human betacellulin. Molecular structure, biological activities, and receptor interaction. J Biol Chem. 269:9966–9973. 1994.PubMed/NCBI

18 

Prewett M, Rockwell P, Rockwell RF, et al: The biologic effects of C225, a chimeric monoclonal antibody to the EGFR, on human prostate carcinoma. J Immunother Emphasis Tumor Immunol. 19:419–427. 1996. View Article : Google Scholar : PubMed/NCBI

19 

Khambata-Ford S, Garrett CR, Meropol NJ, et al: Expression of epiregulin and amphiregulin and K-ras mutation status predict disease control in metastatic colorectal cancer patients treated with cetuximab. J Clin Oncol. 25:3230–3237. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Jacobs B, De Roock W, Piessevaux H, et al: Amphiregulin and epiregulin mRNA expression in primary tumors predicts outcome in metastatic colorectal cancer treated with cetuximab. J Clin Oncol. 27:5068–5074. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Bardelli A and Siena S: Molecular mechanisms of resistance to cetuximab and panitumumab in colorectal cancer. J Clin Oncol. 28:1254–1261. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Krumbach R, Schuler J, Hofmann M, Giesemann T, Fiebig HH and Beckers T: Primary resistance to cetuximab in a panel of patient-derived tumour xenograft models: activation of MET as one mechanism for drug resistance. Eur J Cancer. 47:1231–1243. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Liska D, Chen CT, Bachleitner-Hofmann T, Christensen JG and Weiser MR: HGF rescues colorectal cancer cells from EGFR inhibition via MET activation. Clin Cancer Res. 17:472–482. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Heindl S, Eggenstein E, Keller S, et al: Relevance of MET activation and genetic alterations of KRAS and E-cadherin for cetuximab sensitivity of gastric cancer cell lines. J Cancer Res Clin Oncol. Jan 31–2012.(Epub ahead of print).

25 

Kim IJ, Park JH, Kang HC, et al: Mutational analysis of BRAF and K-ras in gastric cancers: absence of BRAF mutations in gastric cancers. Hum Genet. 114:118–120. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Han SW, Oh DY, Im SA, et al: Phase II study and biomarker analysis of cetuximab combined with modified FOLFOX6 in advanced gastric cancer. Br J Cancer. 100:298–304. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Paull KD, Shoemaker RH, Hodes L, et al: Display and analysis of patterns of differential activity of drugs against human tumor cell lines: development of mean graph and COMPARE algorithm. J Natl Cancer Inst. 81:1088–1092. 1989. View Article : Google Scholar : PubMed/NCBI

28 

Boyd MR and Paull KD: Some practical considerations and applications of the National Cancer Institute in vitro anticancer drug discovery screen. Drug Dev Res. 34:91–109. 1995. View Article : Google Scholar

29 

Luo FR, Yang Z, Dong H, et al: Correlation of pharmacokinetics with the antitumor activity of Cetuximab in nude mice bearing the GEO human colon carcinoma xenograft. Cancer Chemother Pharmacol. 56:455–464. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Robert F, Ezekiel MP, Spencer SA, et al: Phase I study of anti-epidermal growth factor receptor antibody cetuximab in combination with radiation therapy in patients with advanced head and neck cancer. J Clin Oncol. 19:3234–3243. 2001.PubMed/NCBI

31 

Baselga J, Pfister D, Cooper MR, et al: Phase I studies of anti-epidermal growth factor receptor chimeric antibody C225 alone and in combination with cisplatin. J Clin Oncol. 18:904–914. 2000.PubMed/NCBI

32 

Loughrey MB, Waring PM, Tan A, et al: Incorporation of somatic BRAF mutation testing into an algorithm for the investigation of hereditary non-polyposis colorectal cancer. Fam Cancer. 6:301–310. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Pan Q, Pao W and Ladanyi M: Rapid polymerase chain reaction-based detection of epidermal growth factor receptor gene mutations in lung adenocarcinomas. J Mol Diagn. 7:396–403. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Marchetti A, Martella C, Felicioni L, et al: EGFR mutations in non-small-cell lung cancer: analysis of a large series of cases and development of a rapid and sensitive method for diagnostic screening with potential implications on pharmacologic treatment. J Clin Oncol. 23:857–865. 2005. View Article : Google Scholar

35 

Bremm A, Walch A, Fuchs M, et al: Enhanced activation of epidermal growth factor receptor caused by tumor-derived E-cadherin mutations. Cancer Res. 68:707–714. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Engelman JA, Zejnullahu K, Mitsudomi T, et al: MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 316:1039–1043. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Agarwal S, Zerillo C, Kolmakova J, et al: Association of constitutively activated hepatocyte growth factor receptor (Met) with resistance to a dual EGFR/Her2 inhibitor in non-small-cell lung cancer cells. Br J Cancer. 100:941–949. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Guo A, Villen J, Kornhauser J, et al: Signaling networks assembled by oncogenic EGFR and c-Met. Proc Natl Acad Sci USA. 105:692–697. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Asaoka Y, Tada M, Ikenoue T, et al: Gastric cancer cell line Hs746T harbors a splice site mutation of c-Met causing juxta-membrane domain deletion. Biochem Biophys Res Commun. 394:1042–1046. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Tinhofer I, Klinghammer K, Weichert W, et al: Expression of amphiregulin and EGFRvIII affect outcome of patients with squamous cell carcinoma of the head and neck receiving cetuximab-docetaxel treatment. Clin Cancer Res. 17:5197–5204. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Yonesaka K, Zejnullahu K, Lindeman N, et al: Autocrine production of amphiregulin predicts sensitivity to both gefitinib and cetuximab in EGFR wild-type cancers. Clin Cancer Res. 14:6963–6973. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Yasumoto K, Yamada T, Kawashima A, et al: The EGFR ligands amphiregulin and heparin-binding egf-like growth factor promote peritoneal carcinomatosis in CXCR4-expressing gastric cancer. Clin Cancer Res. 17:3619–3630. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Kitadai Y, Yasui W, Yokozaki H, et al: Expression of amphiregulin, a novel gene of the epidermal growth factor family, in human gastric carcinomas. Jpn J Cancer Res. 84:879–884. 1993. View Article : Google Scholar : PubMed/NCBI

44 

Dong J, Opresko LK, Chrisler W, et al: The membrane-anchoring domain of epidermal growth factor receptor ligands dictates their ability to operate in juxtacrine mode. Mol Biol Cell. 16:2984–2998. 2005. View Article : Google Scholar : PubMed/NCBI

45 

Inno A, Salvatore MD, Cenci T, et al: Is there a role for IGF1R and c-MET pathways in resistance to cetuximab in metastatic colorectal cancer? Clin Colorectal Cancer. 10:325–332. 2011. View Article : Google Scholar : PubMed/NCBI

46 

Stommel JM, Kimmelman AC, Ying H, et al: Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies. Science. 318:287–290. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Kim IJ, Park JH, Kang HC, et al: Mutational analysis of BRAF and K-ras in gastric cancers: absence of BRAF mutations in gastric cancers. Hum Genet. 114:118–120. 2003. View Article : Google Scholar : PubMed/NCBI

48 

Mita H, Toyota M, Aoki F, et al: A novel method, digital genome scanning detects KRAS gene amplification in gastric cancers: involvement of overexpressed wild-type KRAS in downstream signaling and cancer cell growth. BMC Cancer. 9:1982009. View Article : Google Scholar : PubMed/NCBI

49 

Kuniyasu H, Yasui W, Kitadai Y, Yokozaki H, Ito H and Tahara E: Frequent amplification of the c-met gene in scirrhous type stomach cancer. Biochem Biophys Res Commun. 189:227–232. 1992. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August 2012
Volume 41 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kneissl J, Keller S, Lorber T, Heindl S, Keller G, Drexler I, Hapfelmeier A, Höfler H and Luber B: Association of amphiregulin with the cetuximab sensitivity of gastric cancer cell lines. Int J Oncol 41: 733-744, 2012
APA
Kneissl, J., Keller, S., Lorber, T., Heindl, S., Keller, G., Drexler, I. ... Luber, B. (2012). Association of amphiregulin with the cetuximab sensitivity of gastric cancer cell lines. International Journal of Oncology, 41, 733-744. https://doi.org/10.3892/ijo.2012.1479
MLA
Kneissl, J., Keller, S., Lorber, T., Heindl, S., Keller, G., Drexler, I., Hapfelmeier, A., Höfler, H., Luber, B."Association of amphiregulin with the cetuximab sensitivity of gastric cancer cell lines". International Journal of Oncology 41.2 (2012): 733-744.
Chicago
Kneissl, J., Keller, S., Lorber, T., Heindl, S., Keller, G., Drexler, I., Hapfelmeier, A., Höfler, H., Luber, B."Association of amphiregulin with the cetuximab sensitivity of gastric cancer cell lines". International Journal of Oncology 41, no. 2 (2012): 733-744. https://doi.org/10.3892/ijo.2012.1479