A growing body of evidence suggests that components of the tumor microenvironment, including cancer-associated fibroblasts (CAF), may modulate the treatment sensitivity of tumor cells. Here, we investigated the possible influence of CAFs on the sensitivity of head and neck squamous cell carcinoma (HNSCC) cell lines to cetuximab, an antagonistic epidermal growth factor receptor (EGFR) antibody. Cetuximab treatment caused a reduction in the proliferation rate of HNSCC cell lines, whereas the growth of HNSCC-derived CAF cultures was unaffected. When tumor cells were cocultured with CAFs in a transwell system, the cetuximab-induced growth inhibition was reduced, and a complete protection from growth inhibition was observed in one of the tumor cell lines investigated. Media that had been conditioned by CAFs offered protection from cetuximab treatment in a concentration-dependent manner, suggesting that the resistance to treatment was mediated by CAF-derived soluble factors. The coculture of HNSCC cell lines with CAFs resulted in an elevated expression of matrix metalloproteinase-1 (MMP-1) in both the tumor cells and CAFs. Moreover, the CAF-induced resistance was partly abolished by the presence of an MMP inhibitor. However, CAFs treated with siRNA targeting MMP-1 still protected tumor cells from cetuximab treatment, suggesting that several MMPs may cooperate to facilitate resistance or that the protective effect is mediated by another member of the MMP family. These results identify a novel CAF-dependent modulation of cetuximab sensitivity and suggest that inhibiting MMPs may improve the effects of EGFR-targeted therapy. Mol Cancer Res; 10(9); 1158–68. ©2012 AACR.

Cancer of the head and neck is the sixth most common form of cancer worldwide with 650,000 new cases each year. The majority of all head and neck cancers are squamous cell carcinomas (HNSCC) with origins in the mucosa of the oral cavity, throat, nose or sinuses. Platinum-based chemoradiotherapy is considered to be the standard treatment for locally advanced HNSCC. However, clinical drug resistance remains a major problem, and the overall survival rate for patients with HNSCC has not increased over the past decades. This treatment deficiency stresses the need for new therapeutic strategies and biomarkers that are predictive of the treatment response.

The EGF receptor (EGFR; also called ErbB1 or HER1) is a transmembrane receptor tyrosine kinase that belongs to the HER family of receptors. Upon ligand binding, the receptor dimerizes, which leads to the autophosphorylation and activation of the intracellular tyrosine kinase domain (1). Kinase activation triggers multiple downstream signaling pathways involving MAPK, PI3K/Akt, STATs, Src, and PLCγ, which regulate key cellular processes including proliferation and survival. EGFR is frequently overexpressed in a number of human solid tumors, including HNSCC, where the high expression of EGFR correlates with a poor clinical outcome (2–3).

Due to the relationship between the overexpression of EGFR and the aggressive behavior of tumor cells, therapies aiming to prevent EGFR signaling, including monoclonal antibodies and small molecule tyrosine kinase inhibitors, have been developed. Cetuximab (Erbitux) is a monoclonal antibody that inhibits tumor growth by binding to the extracellular domain of EGFR to prevent ligand binding (4–6). In addition, cetuximab may also stimulate the internalization of EGFR, which leads to the downregulation of its cell surface expression, and may also trigger antibody-dependent, cell-mediated cytotoxicity (7–8). Cetuximab treatment has been proven to be effective in recurrent or metastatic HNSCC both as first-line treatment in combination with platinum-based chemotherapy (9–12), and as second-line treatment in patients with platinum-refractory disease (13–15). Cetuximab was also shown to improve locoregional control and overall survival when combined with radiotherapy in the first-line treatment of patients with locoregionally advanced HNSCC (16). However, the cetuximab response rate is generally not higher than 20%, highlighting the need for predictive markers of cetuximab treatment response. Increased knowledge regarding the molecular mechanisms by which tumor cells resist cetuximab treatment would enable the selection of patients that would benefit from treatment. Ideally, these resistance mechanisms would also be targetable and would thus provide a possibility to increase the efficacy of cetuximab treatment.

For many years, cancer research has focused mainly on the cancer cell and its molecular changes; however, the stromal microenvironment is now generally accepted to contribute to tumorigenesis in cancers of an epithelial origin (17). Already in premalignant dysplasia, fibroblasts, endothelial cells, and immune cells are recruited to the stroma. The progression from dysplasia into invasive cancer involves the disruption of the basement membrane barrier to enable direct contact between the tumor cells, stromal cells, and extracellular matrix (ECM). During this progression, the stroma undergoes a transformation into a reactive tumor stroma that supports tumor growth and metastasis. Cancer-associated fibroblasts (CAF), the major cellular component of the tumor stroma, may derive from different subsets of cells and likely display functional differences depending on their cancer type of origin. CAFs provide protumorigenic signals that lead to increased tumor growth via the stimulation of tumor cell proliferation and angiogenesis as well as the formation of metastases by the enhancement of the migratory and invasive potential of the tumor cells (18–25). As a number of publications suggest that CAFs could also modulate the drug sensitivity of cancer cells (26–34), this study was undertaken to evaluate the influence of CAFs on the cetuximab response in HNSCC cell lines.

Cells and culture conditions

Head and neck squamous cell carcinomas cell lines (UT-SCC-9, UT-SCC-24A, UT-SCC-19A, and UT-SCC-76A used in passages 28–39, 16–29, 12–15, and 15–18, respectively) were established at the Grénman laboratory in Turku and cultured in Dulbecco's Modified Eagle's Medium supplemented with 2 mmol/L glutamine, 1% nonessential amino acids, 50 IU/mL penicillin, 50 μg/mL streptomycin, and 5% fetal bovine serum (FBS; all from GIBCO). The cells were incubated in humidified air with 5% CO2 at 37°C and subcultured twice a week using 0.25% trypsin with 0.02% EDTA.

Cancer-associated fibroblasts were isolated from 7 patients with HNSCC that had given their informed consent (approved by the Linköping University Ethical Committee). Small pieces of tumor tissue were distributed at the bottom of 25 cm2 cell culture flasks that had been precoated with serum, and 2 mL of keratinocyte serum-free media (GIBCO) supplemented with 50 IU/mL penicillin, 50 μg/mL streptomycin, 2.5 μg/mL amphotericine B (Fungizone; GIBCO), and 20% FBS (Integro B.V.) was added. The tissue cultures were incubated at 37°C in humidified air with 5% CO2, and the media were changed every 3 to 4 days. CAFs were detached and separated from the epithelial cells by differential trypsinization and subsequently cultured in Dulbecco's Modified Eagle's Medium supplemented with 2 mmol/L glutamine, 1% nonessential amino acids, 50 IU/mL penicillin, 50 μg/mL streptomycin, and 5% FBS. All CAF cultures showed homogenous, positive immunofluorescent staining for vimentin and negative staining for cytokeratin (data not shown). Seven different CAF cultures originating from tumors of the tongue, larynx, tonsil, gingiva, and buccal mucosa were used in this study. For all experiments, CAFs were used at passages 2 to 7.

CAF-conditioned media were collected from confluent cultures 72 hours after the media were changed, precleared by a 5-minute centrifugation at 2,060 × g, and stored at −20°C until use. For size-fractionation of the conditioned media, centrifugal filter units (Amicon ultra-4, Millipore, Billerica) were used with molecular weight cutoffs of 50 and 100 kDa (3,000 × g for 45 minutes).

The tumor cells were seeded in 12-well plates (BD Falcon, Franklin Lakes) at a density of 200 (UT-SCC-24A), 625 (UT-SCC-76A), or 750 (UT-SCC-9 and -19A) cells/cm2 depending on the plating efficiency of each cell line. After 48 hours, cetuximab (30 nmol/L; Erbitux) or gefitinib (50 nmol/L; Iressa; AstraZeneca) was added, and the cells were incubated for another 8 days before the cytostatic/cytotoxic effect of the drug was evaluated by crystal violet staining. Where indicated, the cultures were pretreated with 1 μmol/L of MMP inhibitor III (Calbiochem), 100 nmol/L Met inhibitor PHA-665752 (Sigma-Aldrich), or 0.1 to 5 ng/mL of recombinant human hepatocyte growth factor (HGF; Sigma).

The effect of CAFs on the sensitivity of the tumor cells to cetuximab and gefitinib treatment was evaluated by coculture using transwell polycarbonate membrane filters with a 0.4 μm pore size (Falcon, BD Biosciences) or by the addition of fibroblast conditioned media (25% v/v). The tumor cells were allowed to attach before the CAFs were seeded in the filters at a density of 3,300 cells/cm2, which resulted in approximately a 1:1 ratio of CAFs to tumor cells.

Crystal violet staining

The cells were fixed in 4% paraformaldehyde (PFA) for 20 minutes at room temperature (RT) and stained with 0.04% crystal violet in 1% ethanol (20 minutes, RT). The plates were then washed extensively under running tap water and air dried. After solubilization of the samples in 1% SDS, the optical density values were read using a Victor plate reader (EG & G Wallac) at 550 nm.

Immunofluorescent staining

Fibroblasts grown on glass coverslips were fixed with 4% PFA in phosphate-buffered saline (PBS) for 20 minutes at 4°C and subsequently incubated with 0.1% saponin and 5% FBS in PBS for 20 minutes at RT. The cells were then incubated with a monoclonal mouse antihuman vimentin antibody (1:50; Santa Cruz Biotechnology) or a monoclonal mouse antihuman multicytokeratin antibody (1:50; Novacastra) followed by incubation with an Alexa Fluor 488 monoclonal goat antimouse antibody (1:400; Invitrogen). Finally, cells were rinsed in PBS and distilled water, mounted in Vectashield Mounting Medium (Vector Laboratories), and examined with an Olympus photomicroscope (Olympus) using a blue excitation light and a green barrier filter. The controls incubated without the antivimentin or anticytokeratin antibodies did not show any staining.

Western blotting

The cells were washed in PBS and lysed in 63 mmol/L Tris-HCl buffer (pH 6.8) containing 10% glycerol, 2% SDS, 20 μmol/L dithiotreitol, 1% bromphenol blue (all from Sigma-Aldrich), Protease arrest (G-Biosciences), and phosSTOP (Roche Diagnostics GmbH). The protein concentration was determined as described by Lowry and colleagues (35), and the samples were subjected to gel electrophoresis. The proteins were then transferred to a nitrocellulose membrane using an iBlot system (Invitrogen). The proteins were detected using rabbit anti-EGFR (Santa Cruz Biotechnology) and mouse antiphospho-EGFR antibodies (Tyr1173; Millipore) and visualized by HRP-conjugated goat antirabbit and goat antimouse antibodies (Santa Cruz Biotechnology). Equal loading was verified using a HRP-conjugated goat antiactin antibody (Santa Cruz Biotechnology).

ELISA

The quantitative determination of HGF and MMP-1 levels in conditioned media was carried out according to the manufacturer's instructions using the Quantikine (human HGF) and Flourokine E Enzyme Activity Assay (human active MMP1; both from RnD Systems). The optical density for HGF and the relative fluorescence for MMP-1 were measured using the Versa Max (Molecular Devices Corp) and Victor (EG & G Wallac) microplate readers, respectively. All analyses were carried out twice in duplicate, and the mean value was used for further investigations.

RNA extraction, cDNA synthesis, and quantitative real-time PCR

Total RNA was extracted with the RNeasy Mini Kit (Qiagen), and cDNA was obtained using the High Capacity RNA-to-cDNA Kit (Applied Biosystems). The mRNA expression of HGF and MMP-1 was analyzed using a 7,500 Fast Real-Time PCR system and FAM/MGB probes (Applied Biosystems). All reactions and conditions were carried out according to the manufacturer's instructions.

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was amplified as an internal standard. The data were calculated using the comparative Ct method to present the data as a fold difference in the expression level relative to a control sample (36).

Custom RT2 Profiler PCR array

The expression of 87 genes (Supplementary Table S1) primarily involved in the epithelial to mesenchymal transition, EGFR signaling, and treatment resistance was analyzed using a custom-made PCR array plate from SABiosciences. The PCR reaction was carried out according to the manufacturer's protocol.

Total RNA was extracted as described above, and cDNA was acquired using the RT2 First Strand Kit (SABiosciences). DNA and RNA contamination was assessed using a genomic DNA control and a positive PCR control, respectively. The average of 5 housekeeping genes [beta-2-mictoglobulin (B2M), hypoxanthine phosphribosyltransferase 1 (HPRT1), ribosomal protein L13a (RPL13A), GAPDH, and beta-actin (ACTB)] was used to normalize the Ct-values. The relative change in mRNA expression was calculated by the comparative Ct method.

RNA interference

Cells were seeded at a density of 6,700 cells/cm2 in 6-well plates and transfected 24 hours later with siRNA targeting HGF or MMP-1 or a nontargeting siRNA with no homology to any known human gene (AllStars Negative Control siRNA; all from Qiagen) using the HiPerFect transfection reagent (Qiagen). The final concentration of siRNA in the culture medium was 10 nmol/L. Gene silencing was verified by quantitative real-time PCR (qPCR) and ELISA at 48 and 96 hours after transfection, respectively. A 70% knockdown was considered acceptable for further studies.

For cetuximab sensitivity studies, the cells were reseeded in 12-well plates, as described above, 24 hours after transfection. To obtain media conditioned by siRNA-treated cultures, the media were replaced 24 hours after transfection, and the conditioned media were collected after an additional 72 hours.

CAFs induce cetuximab resistance in HNSCC cell lines

Cetuximab treatment (30 nmol/L) resulted in a 40% and 60% reduction of cell growth in the HNSCC UT-SCC-9 (larynx) and UT-SCC-24A (tongue) cell lines, respectively, suggesting that these cells, in part, rely on EGFR signaling to maintain a high proliferation rate. However, when the tumor cells were cocultured with HNSCC-derived CAFs in a transwell system, they were less susceptible to cetuximab treatment. All investigated CAFs completely abolished the cetuximab-induced growth inhibition in the UT-SCC-9 cell line (Fig. 1A and Supplementary Fig. 1A). Interestingly, during the coculture of UT-SCC-9 cells with LK0826Fib, LK0850Fib, or LK0862Fib, cetuximab treatment even stimulated cell growth compared with the respective untreated cocultures. A more modest effect on the treatment response was observed in the UT-SCC-24A cell line where 4 of 7 CAFs significantly reduced the cetuximab sensitivity, but a complete resistance to treatment was not achieved (Fig. 1B and Supplementary Fig. 1B). The results from the UT-SCC-19A and UT-SCC-76A cell lines, which are derived from the larynx and tongue, respectively, showed that the magnitude of the protection conferred by CAFs was not dependent on the tumor location (Fig. 1C and D). Interestingly, CAF cultures were completely resistant to cetuximab treatment, which indicates that mitogenic pathways other than EGFR are dominant in these cells (Fig. 1E). Collectively, these data suggest that under the influence of CAFs tumor cells are less susceptible or even resistant to the growth inhibition caused by cetuximab treatment.

Figure 1.

CAFs induce cetuximab resistance in HNSCC cell lines. HNSCC cell lines (A) UT-SCC-9 (larynx), (B) UT-SCC-24A (tongue), (C) UT-SCC-19A (larynx), and (D) UT-SCC-76A (tongue) were either grown alone or cocultured with HNSCC-derived CAFs and subjected to cetuximab treatment (30 nmol/L). At day 10, the effect of treatment was evaluated by crystal violet staining. The cell growth of cetuximab-treated cultures is shown relative to their respective untreated controls. E, the cetuximab response of CAF monocultures. The data represent the means ± SD from at least 3 experiments in triplicate. The statistical analyses were conducted using one-way ANOVA and Dunnett or Bonferroni post hoc tests (*, P < 0.05). #, a significant increase in the cell growth relative to the cetuximab untreated coculture control.

Figure 1.

CAFs induce cetuximab resistance in HNSCC cell lines. HNSCC cell lines (A) UT-SCC-9 (larynx), (B) UT-SCC-24A (tongue), (C) UT-SCC-19A (larynx), and (D) UT-SCC-76A (tongue) were either grown alone or cocultured with HNSCC-derived CAFs and subjected to cetuximab treatment (30 nmol/L). At day 10, the effect of treatment was evaluated by crystal violet staining. The cell growth of cetuximab-treated cultures is shown relative to their respective untreated controls. E, the cetuximab response of CAF monocultures. The data represent the means ± SD from at least 3 experiments in triplicate. The statistical analyses were conducted using one-way ANOVA and Dunnett or Bonferroni post hoc tests (*, P < 0.05). #, a significant increase in the cell growth relative to the cetuximab untreated coculture control.

Close modal

CAF-mediated growth stimulation is not a prerequisite for the induction of cetuximab resistance

In UT-SCC-9 cultures, which showed the most pronounced CAF-mediated induction of cetuximab resistance, CAFs did not alter the proliferation rate in the absence of cetuximab (Fig. 2A). However, in the other 3 cell lines that showed a more modest protection from cetuximab, the coculture of the tumor cells with CAFs resulted in a significant stimulation of cell growth (Fig. 2B–D). These data indicate that the cetuximab resistance conferred by CAFs is not dependent on the ability of the fibroblasts to stimulate tumor cell proliferation.

Figure 2.

The influence of CAFs on the growth rate of HNSCC cell lines. The HNSCC cell lines (A) UT-SCC-9 (larynx), (B) UT-SCC-24A (tongue), (C) UT-SCC-19A (larynx), and (D) UT-SCC-76A (tongue) were either grown alone or cocultured with HNSCC-derived CAFs. The cell growth was evaluated by crystal violet staining. The growth of the cocultures is shown relative to the tumor cell monocultures. The data represent the means ± SD from at least 3 experiments in triplicate. The statistical analyses were conducted using one-way ANOVA and Dunnett post hoc test (*, P < 0.05).

Figure 2.

The influence of CAFs on the growth rate of HNSCC cell lines. The HNSCC cell lines (A) UT-SCC-9 (larynx), (B) UT-SCC-24A (tongue), (C) UT-SCC-19A (larynx), and (D) UT-SCC-76A (tongue) were either grown alone or cocultured with HNSCC-derived CAFs. The cell growth was evaluated by crystal violet staining. The growth of the cocultures is shown relative to the tumor cell monocultures. The data represent the means ± SD from at least 3 experiments in triplicate. The statistical analyses were conducted using one-way ANOVA and Dunnett post hoc test (*, P < 0.05).

Close modal

CAFs protect tumor cells from EGFR inhibition in general

To establish whether CAFs specifically interfere with the function of cetuximab or rather if they suppress the effect of EGFR inhibition in general, the cells were treated with the EGFR-selective tyrosine kinase inhibitor gefitinib (50 nmol/L). Gefitinib reduced cell growth by 50% and 75%, in UT-SCC-9 and UT-SCC-24A monocultures, respectively (Fig. 3A and B). Similar to the results observed for cetuximab, CAFs provided almost complete protection against the effects of gefitinib treatment in UT-SCC-9 cultures but showed a less pronounced effect in UT-SCC-24A cells. As expected, CAFs did not respond to gefitinib treatment (Fig. 3C). These data suggest that CAFs do not interfere with the interaction between cetuximab and EGFR but rather trigger signaling events that counteract the effects of general EGFR inhibition.

Figure 3.

CAFs induce gefitinib resistance in HNSCC cell lines. HNSCC cell lines (A) UT-SCC-9 and (B) UT-SCC-24A were grown alone or cocultured with HNSCC-derived CAFs and subjected to gefitinib treatment (50 nmol/L). At day 10, the effect of treatment was evaluated by crystal violet staining. The cell growth of gefitinib-treated cultures is shown relative to their respective untreated controls. C, the gefitinib response of CAF monocultures. The data represent the means ± SD from at least 3 experiments in triplicate. The statistical analyses were conducted using one-way ANOVA and Dunnett or Bonferroni post hoc tests (*, P < 0.05).

Figure 3.

CAFs induce gefitinib resistance in HNSCC cell lines. HNSCC cell lines (A) UT-SCC-9 and (B) UT-SCC-24A were grown alone or cocultured with HNSCC-derived CAFs and subjected to gefitinib treatment (50 nmol/L). At day 10, the effect of treatment was evaluated by crystal violet staining. The cell growth of gefitinib-treated cultures is shown relative to their respective untreated controls. C, the gefitinib response of CAF monocultures. The data represent the means ± SD from at least 3 experiments in triplicate. The statistical analyses were conducted using one-way ANOVA and Dunnett or Bonferroni post hoc tests (*, P < 0.05).

Close modal

Protection from cetuximab treatment is provided by CAF-derived soluble factors

Media conditioned by CAFs induced cetuximab resistance in UT-SCC-9 and UT-SCC-24A monocultures, suggesting that CAF-derived soluble factors mediate the protective effect of these cells (Fig. 4A and B and Supplementary Fig. 2). The cetuximab resistance in UT-SCC-9 increased with increasing amounts of media conditioned by LK0862Fib (Fig. 4C). When tumor cell cultures were treated with cetuximab in the presence of size-fractioned conditioned media, the protective effect was observed in the fraction containing factors larger than 50 kDa (Fig. 4D). Interestingly, both the less than 100 kDa and more than 100 kDa fractions conferred cetuximab resistance albeit at a lower level than the unfractionated conditioned media. These results suggest that several resistance factors may cooperate to induce the cetuximab resistance in tumor cells.

Figure 4.

Protection from cetuximab treatment is provided by CAF-derived soluble factors. The HNSCC (A) UT-SCC-9 and (B) UT-SCC-24A cell lines were subjected to cetuximab treatment (30 nmol/L) in the presence or absence of media conditioned by HNSCC-derived CAFs. At day 10, the effect of treatment was evaluated by crystal violet staining. The cell growth of the cetuximab-treated cultures is shown relative to their respective untreated controls. C, UT-SCC-9 cultures were subjected to cetuximab treatment in the presence or absence of LK0862Fib conditioned media (CM) at various concentrations. D, UT-SCC-9 cultures were subjected to cetuximab treatment in the presence or absence of size-fractionated LK0862Fib conditioned media. The data presented in A–C represent the means ± SD from at least 3 experiments in triplicate, and the data in D show the results from 1 representative experiment out of 3 in triplicate. The statistical analyses were conducted using a one-way ANOVA and Dunnett post hoc test (*, P < 0.05).

Figure 4.

Protection from cetuximab treatment is provided by CAF-derived soluble factors. The HNSCC (A) UT-SCC-9 and (B) UT-SCC-24A cell lines were subjected to cetuximab treatment (30 nmol/L) in the presence or absence of media conditioned by HNSCC-derived CAFs. At day 10, the effect of treatment was evaluated by crystal violet staining. The cell growth of the cetuximab-treated cultures is shown relative to their respective untreated controls. C, UT-SCC-9 cultures were subjected to cetuximab treatment in the presence or absence of LK0862Fib conditioned media (CM) at various concentrations. D, UT-SCC-9 cultures were subjected to cetuximab treatment in the presence or absence of size-fractionated LK0862Fib conditioned media. The data presented in A–C represent the means ± SD from at least 3 experiments in triplicate, and the data in D show the results from 1 representative experiment out of 3 in triplicate. The statistical analyses were conducted using a one-way ANOVA and Dunnett post hoc test (*, P < 0.05).

Close modal

CAFs do not alter the expression or phosphorylation of EGFR or its downstream effectors AKT and ERK 1/2

To further examine the protective mechanisms underlying the CAF-induced cetuximab resistance, the mRNA and protein levels of EGFR and its downstream effectors were examined. For these studies, UT-SSC-9 and LK0862Fib cultures were used as the LK0862Fib offered the UT-SCC-9 cells a high level of protection from cetuximab treatment in both coculture and conditioned media experiments.

No changes in the EGFR mRNA or protein levels were found in the UT-SSC-9 cells upon coculture with CAFs (Fig. 5A and B). The amount of phosphorylated EGFR, which was very low in untreated cultures, increased in response to EGF stimulation. In cetuximab-treated cultures, the EGF-induced increase in phosphorylated EGFR was prevented. Importantly, cetuximab also blocked EGFR phosphorylation in the coculture, showing that coculture did not confer resistance by preventing the EGFR-antagonistic effect of cetuximab. Furthermore, no changes in the expression or phosphorylation status of AKT or ERK 1/2 were seen upon coculture with CAFs that could explain the treatment resistance observed (data not shown).

Figure 5.

CAFs do not alter the expression or phosphorylation status of EGFR. UT-SCC-9 HNSCC cells were grown alone or cocultured with HNSCC-derived LK0862Fib CAFs, subjected to cetuximab treatment (30 nmol/L) for 4 hours, and where indicated, stimulated with EGF (10 ng/mL) for 10 minutes before sampling. A, the quantitative PCR results for the EGFR mRNA expression levels. B, a Western blot analysis of total and phosphorylated EGFR. The blots shown are representative of 3 independent experiments. The data represent the means ± SD from 3 experiments. The statistical analyses were conducted using one-way ANOVA and Bonferroni post hoc test (*, P < 0.05).

Figure 5.

CAFs do not alter the expression or phosphorylation status of EGFR. UT-SCC-9 HNSCC cells were grown alone or cocultured with HNSCC-derived LK0862Fib CAFs, subjected to cetuximab treatment (30 nmol/L) for 4 hours, and where indicated, stimulated with EGF (10 ng/mL) for 10 minutes before sampling. A, the quantitative PCR results for the EGFR mRNA expression levels. B, a Western blot analysis of total and phosphorylated EGFR. The blots shown are representative of 3 independent experiments. The data represent the means ± SD from 3 experiments. The statistical analyses were conducted using one-way ANOVA and Bonferroni post hoc test (*, P < 0.05).

Close modal

HGF offers protection from cetuximab treatment but is not responsible for the CAF-induced resistance

As CAFs were previously shown to induce resistance to EGFR tyrosine kinase inhibitors by secretion of HGF (29, 37), the possible involvement of this growth factor in the CAF-mediated cetuximab resistance was investigated. Tumor cells, which lacked HGF expression, stimulated the secretion of HGF when cocultured with CAFs (Fig. 6A). Adding recombinant HGF confirmed that HGF could act as a resistance factor at relevant concentrations (Fig. 6B). However, the media collected from CAFs transfected with siRNA targeting HGF still protected tumor cells from cetuximab treatment (Fig. 6C). Moreover, a Met inhibitor also failed to prevent treatment resistance, thus, the possibility that HGF contributes to the CAF-induced cetuximab resistance was excluded (Fig. 6D).

Figure 6.

HGF offers protection from cetuximab treatment but is not responsible for the CAF-induced resistance. A, the levels of HGF as determined by ELISA in media conditioned by UT-SCC-9 HNSCC cells, HNSCC-derived LK0862Fib CAFs, or a coculture of UT-SCC-9 and LK0862Fib. B, UT-SCC-9 cultures were treated with various concentrations of HGF and subjected to cetuximab treatment (30 nmol/L). At day 10, the effect of treatment was evaluated by crystal violet staining. The cell growth of cetuximab-treated cultures is shown relative to their respective untreated controls. C, UT-SCC-9 cultures were subjected to cetuximab treatment in the presence or absence of conditioned media (CM) collected from LK0862Fib cultures treated with nontargeting or HGF-targeting siRNA (72% and 70% knockdown on mRNA and protein level in cell-culture media, respectively). D, the UT-SCC-9 cultures were treated with cetuximab in the presence or absence of LK0862Fib CM, 5 ng/mL HGF and a Met inhibitor (PHA-665752, 100 nmol/L). The data presented are from 1 representative experiment of 2. The means ± SD of triplicate samples are shown in B–D. The statistical analyses were conducted using one-way ANOVA and Bonferroni post hoc test (*, P < 0.05).

Figure 6.

HGF offers protection from cetuximab treatment but is not responsible for the CAF-induced resistance. A, the levels of HGF as determined by ELISA in media conditioned by UT-SCC-9 HNSCC cells, HNSCC-derived LK0862Fib CAFs, or a coculture of UT-SCC-9 and LK0862Fib. B, UT-SCC-9 cultures were treated with various concentrations of HGF and subjected to cetuximab treatment (30 nmol/L). At day 10, the effect of treatment was evaluated by crystal violet staining. The cell growth of cetuximab-treated cultures is shown relative to their respective untreated controls. C, UT-SCC-9 cultures were subjected to cetuximab treatment in the presence or absence of conditioned media (CM) collected from LK0862Fib cultures treated with nontargeting or HGF-targeting siRNA (72% and 70% knockdown on mRNA and protein level in cell-culture media, respectively). D, the UT-SCC-9 cultures were treated with cetuximab in the presence or absence of LK0862Fib CM, 5 ng/mL HGF and a Met inhibitor (PHA-665752, 100 nmol/L). The data presented are from 1 representative experiment of 2. The means ± SD of triplicate samples are shown in B–D. The statistical analyses were conducted using one-way ANOVA and Bonferroni post hoc test (*, P < 0.05).

Close modal

CAF-induced cetuximab resistance is dependent on the matrix metalloproteinases

In an attempt to identify the cause of the reduced sensitivity of tumor cells to cetuximab treatment, a PCR array analyzing the expression of 87 genes was carried out in mono- and cocultured tumor cells (Supplementary Table S1). The expression levels of 2 proteins, matrix metalloproteinase-1 (MMP-1) and the EGFR ligand amphiregulin, were found to increase by more than 2-fold in cells cocultured with CAFs. Recombinant amphiregulin failed to protect tumor cells from cetuximab treatment and was therefore ruled out as a possible resistance factor (data not shown). Using quantitative PCR, the increased expression of MMP-1 in tumor cells cocultured with CAFs was confirmed (Fig. 7A). Furthermore, an even greater upregulation of MMP-1 mRNA expression was observed in cocultured CAFs. We failed to detect any active MMP-1 secreted by the tumor cells; however, in accordance with the mRNA data, a higher level of active MMP-1 was found in the media collected from the cocultures compared with the media conditioned by CAFs alone (Fig. 7B). An inhibitor of MMP-1 significantly reduced the protective effect of CAF-conditioned media (Fig. 7C); however, media collected from CAFs transfected with siRNA targeting MMP-1was as effective in inducing resistance as media from MMP-1 expressing CAFs (Fig. 7D). The downregulation of MMP-1 expression in tumor cells also failed to increase their cetuximab sensitivity but rather resulted in the opposite effect (Fig. 7E).

Figure 7.

CAF-induced cetuximab resistance is dependent on the MMPs. A, the MMP-1 mRNA expression in mono- or cocultured UT-SCC-9 HNSCC cells and HNSCC-derived LK0862Fib CAFs was evaluated. B, the active MMP-1 in the culture media conditioned by UT-SCC-9, LK0862Fib, or cocultures of UT-SCC-9 and LK0862Fib. C, the UT-SCC-9 cultures were subjected to cetuximab treatment (30 nmol/L) in the presence or absence of LK0862Fib-conditioned media (CM) and an MMP inhibitor. At day 10, the effect of treatment was evaluated by crystal violet staining. The cell growth of cetuximab-treated cultures is shown relative to their respective untreated controls. D, the UT-SCC-9 cultures were subjected to cetuximab treatment in the presence or absence of media collected from LK0862Fib cultures transfected with nontargeting or MMP-1 targeting siRNA (88% and 81% knockdown on the mRNA and protein level, respectively). E, the cetuximab treatment response of UT-SSC-9 cultures transfected with nontargeting or MMP-1 targeting siRNA (83% knockdown on the mRNA level). The data presented are from 1 representative experiment out of 2. The means ± SD of triplicate samples are shown in Fig. 6 C–E. The statistical analyses were conducted using one-way ANOVA and Bonferroni post hoc test (*, P < 0.05).

Figure 7.

CAF-induced cetuximab resistance is dependent on the MMPs. A, the MMP-1 mRNA expression in mono- or cocultured UT-SCC-9 HNSCC cells and HNSCC-derived LK0862Fib CAFs was evaluated. B, the active MMP-1 in the culture media conditioned by UT-SCC-9, LK0862Fib, or cocultures of UT-SCC-9 and LK0862Fib. C, the UT-SCC-9 cultures were subjected to cetuximab treatment (30 nmol/L) in the presence or absence of LK0862Fib-conditioned media (CM) and an MMP inhibitor. At day 10, the effect of treatment was evaluated by crystal violet staining. The cell growth of cetuximab-treated cultures is shown relative to their respective untreated controls. D, the UT-SCC-9 cultures were subjected to cetuximab treatment in the presence or absence of media collected from LK0862Fib cultures transfected with nontargeting or MMP-1 targeting siRNA (88% and 81% knockdown on the mRNA and protein level, respectively). E, the cetuximab treatment response of UT-SSC-9 cultures transfected with nontargeting or MMP-1 targeting siRNA (83% knockdown on the mRNA level). The data presented are from 1 representative experiment out of 2. The means ± SD of triplicate samples are shown in Fig. 6 C–E. The statistical analyses were conducted using one-way ANOVA and Bonferroni post hoc test (*, P < 0.05).

Close modal

Because the MMP-1 inhibitor also blocks several other MMPs (MMP-2, -3, -7, and -13), these results collectively indicate that one of the other MMPs may confer protection to cetuximab treatment or that several CAF-regulated MMPs cooperate in the induction of drug resistance.

The importance of the tumor microenvironment for tumor growth and metastasis has been recognized for many years. In 1990, Teicher and colleagues proposed that something other than the inherent properties of tumor cells must affect the ability of these cells to resist cytotoxic drugs (38). Since then, several studies have shown that fibroblasts can induce resistance to various anticancer treatments, such as tamoxifen in the treatment of breast cancer (28, 39), gemcitabine and radiotherapy in the treatment of pancreatic adenocarcinoma (26), and paclitaxel in the treatment of non–small cell lung cancer (30). Conversely, fibroblasts may also sensitize tumor cells to therapy as was shown for RAF and MEK1 inhibitors in breast cancer (33). However, the influence of CAFs on the cetuximab treatment response in HNSCC has not previously been investigated.

Here, we show that CAFs isolated from patients with HNSCC desensitize HNSCC cell lines to the EGFR-targeted therapies cetuximab and gefitinib by the secretion of unidentified soluble factors. The protective effect was partially dependent on MMP activity, which suggests that CAFs may release MMPs or factors that influence the expression or activation of tumor cell-derived MMPs. To our knowledge, this is the first paper that shows a CAF-dependent induction of tumor cell cetuximab resistance. In agreement with our findings regarding the outcome of gefitinib treatment, it was recently reported that human lung embryonic fibroblasts and CAFs from patients with lung cancer reduce the effect of this drug in lung cancer cell lines harboring EGFR-activating mutations (29). These mutations are very rare in HNSCC (40–41). Thus, our data provide evidence that CAF-mediated resistance might also operate in tumors lacking EGFR-activating mutations.

Full protection from cetuximab-induced growth inhibition was observed in UT-SCC-9 cells, whereas the influence of CAFs on the treatment response was less pronounced in the 3 other cell lines tested. This discrepancy in the magnitude of the protection conferred by CAFs was independent of the location of the tumor from which the cell lines originate. As similar results were obtained using CAF-conditioned media, it is unlikely that the particular sensitivity of UT-SCC-9 involves a greater capacity of these tumor cells to induce the secretion of CAF-derived resistance factors. Thus, other inherent properties of the tumor cells must determine the extent to which CAFs influence the treatment response. When identified, these properties could be useful for the prediction of the cetuximab treatment outcome.

Our data suggest that factors associated with both CAFs and tumor cells determine the outcome of EGFR-targeted therapy. This indicates that successful individualized treatment of patients with HNSCC is possible only if predictive factors of the treatment response are identified both in tumor cells and the tumor microenvironment. The identification of stromal resistance factors that hamper the clinical response of EGFR-targeted therapy may not only provide useful biomarkers, but also open new treatment possibilities. If one could target the crosstalk between cancer cells and CAFs, the efficacy of cetuximab treatment in patients with HNSCC could be greatly improved.

Importantly, in the presence of 3 (LK0826Fib, LK0850Fib, and LK0862Fib) of 7 CAFs tested, the growth inhibition caused by cetuximab treatment in the UT-SCC-9 cell line was not only abolished, but also replaced by a growth stimulation. This result further highlights the need for biomarkers to predict the cetuximab treatment response. These biomarkers would enable not only the identification of patients that would benefit, but also those that could potentially be harmed by cetuximab treatment.

Much effort has been put into the development of MMP inhibitors to treat cancer. Although an impressive effect was observed for MMP inhibitors in in vitro and mouse models, the results from clinical trials have been disappointing (42–43). The failure of these drugs may in part be explained by the fact that many of the patients recruited in these trials were at the most advanced, metastatic stages of cancer. As is the case for most anticancer drugs, MMP inhibitors may be more effective when given at earlier stages of cancer progression. Our data suggest that MMPs are involved in the development of stroma-induced resistance to EGFR-targeted therapies. Furthermore, previous studies have indicated a correlation between MMPs and the resistance to cytotoxic agents (44–46). Thus, when combined with other anticancer therapies at earlier stages of cancer, MMP inhibitors may improve the clinical outcome.

In summary, we have shown that CAFs induce cetuximab resistance in HNSCC cells in an MMP-dependent manner. These results suggest that targeting the crosstalk between CAFs and tumor cells may circumvent the resistance to EGFR-targeted therapy in patients with HNSCC.

Studies in the A. Östman laboratory was partially funded by an unrestricted research grant from Merck Serono, Stockholm, Sweden. A. Östman is on the advisory board of Pfizer, ImClone, and Merck Serono. No potential conflicts of interest were disclosed by the other authors.

Conception and design: A.C. Johansson, A. Ansell, F. Jerhammar, R. Grénman, A. Östman, K. Roberg

Development of methodology: M. Bradic Lindh, R. Grénman, K.B. Roberg

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A.C. Johansson, A. Ansell, Eva Munck-Wikland, R. Grénman

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A.C. Johansson, A. Ansell, M. Bradic Lindh, A. Östman

Writing, review, and/or revision of the manuscript: A.C. Johansson, A. Ansell, F. Jerhammar, Eva Munck-Wikland, R. Grénman, A. Östman, K. Roberg

Study supervision: A. Östman

In loving memory of Dr. Cathrine Nilsson. The authors thank Camilla Janefjord for technical assistance.

A.C. Johansson received financial support from the Johan and Jakob Söderberg Foundation, the Foundation Olle Engqvist Byggmästare, the Swedish Laryng Foundation, Borgholm Rotary Club, the Swedish National Board of Health and Welfare, Lions Research Foundation, the Lars Hierta Memorial Foundation, the Tore Nilsson Foundation for Medical Research, the Swedish Society for medical research, the County Council of Östergötland, and the Cancer Foundation of Östergötland. A. Östman received support from Merck Serono, the Swedish Research Council (349-2008-6578), and the Swedish Cancer Society (CAN 2009/1136). K. Roberg received support from the Swedish Cancer Society (CAN 2010/545).

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.

1.
Ono
M
,
Kuwano
M
. 
Molecular mechanisms of epidermal growth factor receptor (EGFR) activation and response to gefitinib and other EGFR-targeting drugs
.
Clin Cancer Res
2006
;
12
:
7242
51
.
2.
Dassonville
O
,
Formento
JL
,
Francoual
M
,
Ramaioli
A
,
Santini
J
,
Schneider
M
, et al
Expression of epidermal growth factor receptor and survival in upper aerodigestive tract cancer
.
J Clin Oncol
1993
;
11
:
1873
8
.
3.
Grandis
JR
,
Melhem
MF
,
Barnes
EL
,
Tweardy
DJ
. 
Quantitative immunohistochemical analysis of transforming growth factor-alpha and epidermal growth factor receptor in patients with squamous cell carcinoma of the head and neck
.
Cancer
1996
;
78
:
1284
92
.
4.
Goldstein
NI
,
Prewett
M
,
Zuklys
K
,
Rockwell
P
,
Mendelsohn
J
. 
Biological efficacy of a chimeric antibody to the epidermal growth factor receptor in a human tumor xenograft model
.
Clin Cancer Res
1995
;
1
:
1311
8
.
5.
Li
S
,
Schmitz
KR
,
Jeffrey
PD
,
Wiltzius
JJ
,
Kussie
P
,
Ferguson
KM
. 
Structural basis for inhibition of the epidermal growth factor receptor by cetuximab
.
Cancer Cell
2005
;
7
:
301
11
.
6.
Sato
JD
,
Kawamoto
T
,
Le
AD
,
Mendelsohn
J
,
Polikoff
J
,
Sato
GH
. 
Biological effects in vitro of monoclonal antibodies to human epidermal growth factor receptors
.
Mol Biol Med
1983
;
1
:
511
29
.
7.
Kimura
H
,
Sakai
K
,
Arao
T
,
Shimoyama
T
,
Tamura
T
,
Nishio
K
. 
Antibody-dependent cellular cytotoxicity of cetuximab against tumor cells with wild-type or mutant epidermal growth factor receptor
.
Cancer Sci
2007
;
98
:
1275
80
.
8.
Zhang
W
,
Gordon
M
,
Schultheis
AM
,
Yang
DY
,
Nagashima
F
,
Azuma
M
, et al
FCGR2A and FCGR3A polymorphisms associated with clinical outcome of epidermal growth factor receptor expressing metastatic colorectal cancer patients treated with single-agent cetuximab
.
J Clin Oncol
2007
;
25
:
3712
8
.
9.
Bourhis
J
,
Rivera
F
,
Mesia
R
,
Awada
A
,
Geoffrois
L
,
Borel
C
, et al
Phase I/II study of cetuximab in combination with cisplatin or carboplatin and fluorouracil in patients with recurrent or metastatic squamous cell carcinoma of the head and neck
.
J Clin Oncol
2006
;
24
:
2866
72
.
10.
Burtness
B
,
Goldwasser
MA
,
Flood
W
,
Mattar
B
,
Forastiere
AA
. 
Phase III randomized trial of cisplatin plus placebo compared with cisplatin plus cetuximab in metastatic/recurrent head and neck cancer: an Eastern Cooperative Oncology Group study
.
J Clin Oncol
2005
;
23
:
8646
54
.
11.
Shin
DM
,
Donato
NJ
,
Perez-Soler
R
,
Shin
HJ
,
Wu
JY
,
Zhang
P
, et al
Epidermal growth factor receptor-targeted therapy with C225 and cisplatin in patients with head and neck cancer
.
Clin Cancer Res
2001
;
7
:
1204
13
.
12.
Vermorken
JB
,
Mesia
R
,
Rivera
F
,
Remenar
E
,
Kawecki
A
,
Rottey
S
, et al
Platinum-based chemotherapy plus cetuximab in head and neck cancer
.
N Engl J Med
2008
;
359
:
1116
27
.
13.
Baselga
J
,
Trigo
JM
,
Bourhis
J
,
Tortochaux
J
,
Cortes-Funes
H
,
Hitt
R
, et al
Phase II multicenter study of the antiepidermal growth factor receptor monoclonal antibody cetuximab in combination with platinum-based chemotherapy in patients with platinum-refractory metastatic and/or recurrent squamous cell carcinoma of the head and neck
.
J Clin Oncol
2005
;
23
:
5568
77
.
14.
Herbst
RS
,
Arquette
M
,
Shin
DM
,
Dicke
K
,
Vokes
EE
,
Azarnia
N
, et al
Phase II multicenter study of the epidermal growth factor receptor antibody cetuximab and cisplatin for recurrent and refractory squamous cell carcinoma of the head and neck
.
J Clin Oncol
2005
;
23
:
5578
87
.
15.
Vermorken
JB
,
Trigo
J
,
Hitt
R
,
Koralewski
P
,
Diaz-Rubio
E
,
Rolland
F
, et al
Open-label, uncontrolled, multicenter phase II study to evaluate the efficacy and toxicity of cetuximab as a single agent in patients with recurrent and/or metastatic squamous cell carcinoma of the head and neck who failed to respond to platinum-based therapy
.
J Clin Oncol
2007
;
25
:
2171
7
.
16.
Bonner
JA
,
Harari
PM
,
Giralt
J
,
Azarnia
N
,
Shin
DM
,
Cohen
RB
, et al
Radiotherapy plus cetuximab for squamous-cell carcinoma of the head and neck
.
N Engl J Med
2006
;
354
:
567
78
.
17.
McAllister
SS
,
Weinberg
RA
. 
Tumor-host interactions: a far-reaching relationship
.
J Clin Oncol
2010
;
28
:
4022
8
.
18.
Orimo
A
,
Gupta
PB
,
Sgroi
DC
,
Arenzana-Seisdedos
F
,
Delaunay
T
,
Naeem
R
, et al
Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion
.
Cell
2005
;
121
:
335
48
.
19.
Camps
JL
,
Chang
SM
,
Hsu
TC
,
Freeman
MR
,
Hong
SJ
,
Zhau
HE
, et al
Fibroblast-mediated acceleration of human epithelial tumor growth in vivo
.
Proc Natl Acad Sci U S A
1990
;
87
:
75
9
.
20.
Picard
O
,
Rolland
Y
,
Poupon
MF
. 
Fibroblast-dependent tumorigenicity of cells in nude mice: implication for implantation of metastases
.
Cancer Res
1986
;
46
:
3290
4
.
21.
De Wever
O
,
Demetter
P
,
Mareel
M
,
Bracke
M
. 
Stromal myofibroblasts are drivers of invasive cancer growth
.
Int J Cancer
2008
;
123
:
2229
38
.
22.
Kalluri
R
,
Zeisberg
M
. 
Fibroblasts in cancer
.
Nat Rev Cancer
2006
;
6
:
392
401
.
23.
Orimo
A
,
Weinberg
RA
. 
Stromal fibroblasts in cancer: a novel tumor-promoting cell type
.
Cell Cycle
2006
;
5
:
1597
601
.
24.
Ostman
A
,
Augsten
M
. 
Cancer-associated fibroblasts and tumor growth—bystanders turning into key players
.
Curr Opin Genet Dev
2009
;
19
:
67
73
.
25.
Rasanen
K
,
Vaheri
A
. 
Activation of fibroblasts in cancer stroma
.
Exp Cell Res
2010
;
316
:
2713
22
.
26.
Hwang
RF
,
Moore
T
,
Arumugam
T
,
Ramachandran
V
,
Amos
KD
,
Rivera
A
, et al
Cancer-associated stromal fibroblasts promote pancreatic tumor progression
.
Cancer Res
2008
;
68
:
918
26
.
27.
Muerkoster
S
,
Wegehenkel
K
,
Arlt
A
,
Witt
M
,
Sipos
B
,
Kruse
ML
, et al
Tumor stroma interactions induce chemoresistance in pancreatic ductal carcinoma cells involving increased secretion and paracrine effects of nitric oxide and interleukin-1beta
.
Cancer Res
2004
;
64
:
1331
7
.
28.
Shekhar
MP
,
Santner
S
,
Carolin
KA
,
Tait
L
. 
Direct involvement of breast tumor fibroblasts in the modulation of tamoxifen sensitivity
.
Am J Pathol
2007
;
170
:
1546
60
.
29.
Wang
W
,
Li
Q
,
Yamada
T
,
Matsumoto
K
,
Matsumoto
I
,
Oda
M
, et al
Crosstalk to stromal fibroblasts induces resistance of lung cancer to epidermal growth factor receptor tyrosine kinase inhibitors
.
Clin Cancer Res
2009
;
15
:
6630
8
.
30.
Bartling
B
,
Hofmann
HS
,
Silber
RE
,
Simm
A
. 
Differential impact of fibroblasts on the efficient cell death of lung cancer cells induced by paclitaxel and cisplatin
.
Cancer Biol Ther
2008
;
7
:
1250
61
.
31.
Martinez-Outschoorn
UE
,
Lin
Z
,
Ko
YH
,
Goldberg
AF
,
Flomenberg
N
,
Wang
C
, et al
Understanding the metabolic basis of drug resistance: therapeutic induction of the Warburg effect kills cancer cells
.
Cell Cycle
2011
;
10
:
2521
8
.
32.
Liles
JS
,
Arnoletti
JP
,
Kossenkov
AV
,
Mikhaylina
A
,
Frost
AR
,
Kulesza
P
, et al
Targeting ErbB3-mediated stromal-epithelial interactions in pancreatic ductal adenocarcinoma
.
Br J Cancer
2011
;
105
:
523
33
.
33.
Dittmer
A
,
Fuchs
A
,
Oerlecke
I
,
Leyh
B
,
Kaiser
S
,
Martens
JW
, et al
Mesenchymal stem cells and carcinoma-associated fibroblasts sensitize breast cancer cells in 3D cultures to kinase inhibitors
.
Int J Oncol
2011
;
39
:
689
96
.
34.
Manshouri
T
,
Estrov
Z
,
Quintas-Cardama
A
,
Burger
J
,
Zhang
Y
,
Livun
A
, et al
Bone marrow stroma-secreted cytokines protect JAK2(V617F)-mutated cells from the effects of a JAK2 inhibitor
.
Cancer Res
2011
;
71
:
3831
40
.
35.
Lowry
OH
,
Rosebrough
NJ
,
Farr
AL
,
Randall
RJ
. 
Protein measurement with the folin phenol reagent
.
J Biol Chem
1951
;
193
:
265
75
.
36.
Leutenegger
CM
,
Mislin
CN
,
Sigrist
B
,
Ehrengruber
MU
,
Hofmann-Lehmann
R
,
Lutz
H
. 
Quantitative real-time PCR for the measurement of feline cytokine mRNA
.
Vet Immunol Immunopathol
1999
;
71
:
291
305
.
37.
Yamada
T
,
Matsumoto
K
,
Wang
W
,
Li
Q
,
Nishioka
Y
,
Sekido
Y
, et al
Hepatocyte growth factor reduces susceptibility to an irreversible epidermal growth factor receptor inhibitor in EGFR-T790M mutant lung cancer
.
Clin Cancer Res
2010
;
16
:
174
83
.
38.
Teicher
BA
,
Herman
TS
,
Holden
SA
,
Wang
YY
,
Pfeffer
MR
,
Crawford
JW
, et al
Tumor resistance to alkylating agents conferred by mechanisms operative only in vivo
.
Science
1990
;
247
:
1457
61
.
39.
Martinez-Outschoorn
UE
,
Lin
Z
,
Ko
YH
,
Goldberg
AF
,
Flomenberg
N
,
Wang
C
, et al
Understanding the metabolic basis of drug resistance: therapeutic induction of the Warburg effect kills cancer cells
.
Cell Cycle
2011
;
10
:
2521
8
.
40.
Sheikh
Ali MA
,
Gunduz
M
,
Nagatsuka
H
,
Gunduz
E
,
Cengiz
B
,
Fukushima
K
, et al
Expression and mutation analysis of epidermal growth factor receptor in head and neck squamous cell carcinoma
.
Cancer Sci
2008
;
99
:
1589
94
.
41.
Lemos-Gonzalez
Y
,
Paez de la Cadena
M
,
Rodriguez-Berrocal
FJ
,
Rodriguez-Pineiro
AM
,
Pallas
E
,
Valverde
D
. 
Absence of activating mutations in the EGFR kinase domain in Spanish head and neck cancer patients
.
Tumour Biol
2007
;
28
:
273
9
.
42.
Coussens
LM
,
Fingleton
B
,
Matrisian
LM
. 
Matrix metalloproteinase inhibitors and cancer: trials and tribulations
.
Science
2002
;
295
:
2387
92
.
43.
Pavlaki
M
,
Zucker
S
. 
Matrix metalloproteinase inhibitors (MMPIs): the beginning of phase I or the termination of phase III clinical trials
.
Cancer Metastasis Rev
2003
;
22
:
177
203
.
44.
Ansell
A
,
Jerhammar
F
,
Ceder
R
,
Grafstrom
R
,
Grenman
R
,
Roberg
K
. 
Matrix metalloproteinase-7 and -13 expression associate to cisplatin resistance in head and neck cancer cell lines
.
Oral Oncol
2009
;
45
:
866
71
.
45.
Almendro
V
,
Ametller
E
,
Garcia-Recio
S
,
Collazo
O
,
Casas
I
,
Auge
JM
, et al
The role of MMP7 and its cross-talk with the FAS/FASL system during the acquisition of chemoresistance to oxaliplatin
.
PLoS One
2009
;
4
:
e4728
.
46.
Mitsiades
N
,
Yu
WH
,
Poulaki
V
,
Tsokos
M
,
Stamenkovic
I
. 
Matrix metalloproteinase-7-mediated cleavage of Fas ligand protects tumor cells from chemotherapeutic drug cytotoxicity
.
Cancer Res
2001
;
61
:
577
81
.