Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
    • CME
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Clinical Cancer Research
Clinical Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
    • CME
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • CCR Focus Archive
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Breast Cancer
      • Clinical Trials
      • Immunotherapy: Facts and Hopes
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citation
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Cancer Therapy: Preclinical

Resistance to Chemotherapy Is Associated with Fibroblast Growth Factor Receptor 4 Up-Regulation

Andreas Roidl, Hans-Jürgen Berger, Sushil Kumar, Johannes Bange, Pjotr Knyazev and Axel Ullrich
Andreas Roidl
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Hans-Jürgen Berger
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sushil Kumar
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Johannes Bange
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Pjotr Knyazev
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Axel Ullrich
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/1078-0432.CCR-08-0890 Published March 2009
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Purpose: Establishment of antiapoptotic signaling pathways in tumor cells is a major cause for the failure of chemotherapy against cancer. To investigate the underlying mechanisms, we developed an experimental approach that is based on the genetic plasticity of cancer cells and the selection for cell survival on treatment with chemotherapeutic agents.

Experimental Design: Gene expression changes of surviving cell clones were analyzed by macroarrays. Involvement of fibroblast growth factor receptor 4 (FGFR4) in antiapoptotic pathways was elucidated by apoptosis assays, small interfering RNA experiments, and an antagonistic antibody.

Results: We show that FGFR4 gene expression is up-regulated in doxorubicin-treated, apoptosis-resistant cancer cell clones. Ectopic expression of FGFR4 in cancer cells led to reduced apoptosis sensitivity on treatment with doxorubicin or cyclophosphamide, whereas knockdown of endogenous FGFR4 expression in breast cancer cell lines had the opposite effect. FGFR4 overexpression resulted in Bcl-xl up-regulation at both mRNA and protein levels. Knockdown of FGFR4 expression by small interfering RNA caused a decrease in phospho-extracellular signal-regulated kinase 1/2 levels and reduced Bcl-xl expression. Moreover, an antagonistic FGFR4 antibody suppressed the resistance of cancer cells with endogenous FGFR4 expression against apoptosis-inducing chemotherapeutic agents.

Conclusion: Based on these findings, we propose an antiapoptotic signaling pathway that is initiated by FGFR4 and regulating the expression of Bcl-xl through the mitogen-activated protein kinase cascade. Our findings are exemplary for a novel strategy toward the elucidation of diverse signaling pathways that define antiapoptotic potential in cancer cells. These observations open new avenues toward the diagnosis of chemoresistant tumors and therapies targeting FGFR4-overexpressing cancers.

  • cancer
  • FGFR4
  • antiapoptosis
  • chemoresistance
  • Bcl-xl

Translational Relevance

Treatment of patients suffering from breast cancer is still largely based on classic chemotherapy. Beside serious side effects, formation of resistance to these drugs is hampering successful cancer therapy. Here, we describe a novel strategy toward the elucidation of signaling pathways and targets that define chemoresistance in cancer cells. Using our cell genetics-based strategy, we identified the fibroblast growth factor receptor 4 (FGFR4) to contribute to the cancer cell resistance phenotype. We show that doxorubicin-resistant cancer cells become sensitive after being treated with small interfering RNA or a blocking antibody against FGFR4. In a combined application of FGFR4-blocking antibody and the chemotherapeutic drug doxorubicin, we could further increase the apoptosis rate of cancer cell lines, suggesting a combinatorial strategy of FGFR4 inhibitors (e.g., antagonistic antibodies) and chemotherapeutic drugs in FGFR4-overexpressing cancers. These results present new options toward the early diagnosis of chemoresistance and suggest novel combinations of chemotherapy and anti-FGFR4 agents to prevent or significantly delay the onset of therapy resistance.

Breast cancer is the most frequent malignancy among women in the western world.3,4 Although much effort has been invested into designing new therapies, classic chemotherapeutic agents such as doxorubicin (Adriamycin) are still widely employed in the clinic (1). Chemotherapeutic drugs are used as primary or adjuvant therapy with response rates from 60% to 100% (2, 3). A major problem of most of the current therapies are the sometimes severe side effects and the intrinsic or acquired drug resistance of cancer cells to these drugs, which lead to relapse and metastatic progression of the tumor. This is reflected by the decline in the response rates with second-line chemotherapy to 20% to 30% (4).

Motivated by these facts, a comprehensive characterization of genes contributing to a drug resistance phenotype has been done in this study. Based on an approach of Hudziak et al. (5) and Abraham et al. (6), where tumor necrosis factor-α-resistant and Fas ligand-resistant cell clones were established, we respectively took advantage of the genomic instability of breast cancer cell lines to generate doxorubicin-resistant cell clones. When treated with chemotherapeutic drugs, cell clones survived the treatment because of their ability to adapt to the selection pressure by using antiapoptotic mechanisms. Subsequent comparison of gene expression profiles of apoptosis-sensitive breast cancer cells and corresponding apoptosis-resistant clones led to the identification of fibroblast growth factor receptor 4 (FGFR4) as a gene with increased gene expression in response to treatment with doxorubicin or cyclophosphamide.

The four closely related members of the human FGFRs (FGFR1-FGFR4) belong to the receptor tyrosine kinase family and consist of three IG domains, an acidic box, except for FGFR2, a HAV motive, and a split tyrosine kinase domain (7). Currently, 23 ligands are known to activate FGFRs and stimulate a multitude of signaling pathways (8). On ligand binding, FGFRs become transphosphorylated, which results in the recruitment of the central adaptor protein FGFR substrate 2. FGFR substrate 2 is a binding platform for various adaptor proteins such as gab1, shc, grb2, and other signaling molecules (9). FGFR signaling leads among others to activation of mitogen-activated protein kinase (MAPK) in breast cancer cells and thereby stimulates proliferation (10, 11). Other cellular properties such as differentiation, motility (12), and invasiveness (13) can also be regulated by FGFRs. Dysregulation of FGFR signaling can lead to severe diseases such as Apert, Pfeiffer, Jackson-Weiss, or Crouzon syndromes or achondroplasia (14). Importantly, FGFRs have been reported to contribute to oncogenic cell transformation in different human cancers (15, 16). For example, overexpression or selection for constitutively active mutants of FGFR1 and FGFR3 in multiple myelomas leads to an accelerated progression of the disease by enhanced cell survival (17). Recently, we have identified a short nucleotide polymorphism of the transmembrane domain in FGFR4 homologous to that in FGFR3 causing achondroplasia (13). In this study, an involvement of FGFR4 in disease progression and reduced disease-free survival time of breast cancer patients could be shown. Similar findings were also reported in other studies on different cancer types (13, 18).

To identify genes that are critical in the establishment of antiapoptotic mechanisms in cancer cells, we treated breast cancer cells with doxorubicin and isolated resistant clones. We analyzed changes of gene expression and found FGFR4 as a gene with significantly increased expression in chemoresistant clones. In FGFR4 ectopically overexpressing breast cancer cells, the same phenotype was observed. In accordance with these results, small interfering RNA (siRNA)-mediated down-regulation of the endogenous receptor in chemoresistant clones and in other cancer cell lines increased chemosensitivity. Furthermore, we could show that inhibition of FGFR4 with an antagonistic antibody enhanced chemosensitivity of endogenously FGFR4-expressing cell lines as well. We show a positive regulation of the antiapoptotic protein Bcl-xl by FGFR4 via the MAPK cascade. These findings implicate FGFR4 as an important factor for cancer cell resistance formation in oncogenesis and during chemotherapy.

Materials and Methods

Reagents and antibodies. FGF ligands were purchased from TEBU. The antibodies used were polyclonal anti-FGFR4, anti-extracellular signal-regulated kinase (ERK) 2, and anti-Akt1/2 (Santa Cruz Biotechnology), rabbit polyclonal anti-phospho-Akt (Ser473; New England Biolabs), mouse monoclonal anti-phosphotyrosine antibody 4G10 (Upstate Biotechnology), mouse monoclonal anti-tubulin (Sigma), rabbit polyclonal anti-phospho-ERK (Cell Signalling), mouse monoclonal anti-Bcl-xl (BD Biosciences), mouse monoclonal anti-VSV antibody (Sigma), and mouse monoclonal anti-FGFR4 antibody 10F10 (U3 Pharma).

Secondary horseradish peroxidase-conjugated antibodies were goat anti-mouse (Sigma) and goat anti-rabbit (Bio-Rad). If required, cells were starved by total serum withdrawal for 48 h and treated with growth factors as indicated in the figure legends.

Cell culture, plasmids, and retroviral infections. Cell lines 769-P, TE671, MDA-MB-453, BT-474, MDA-MB-361, MDA-MB-231, ZR75-1, and MCF7 were obtained from the American Type Culture Collection and cultivated following the supplier's instruction, except MDA-MB-453 cells, which were maintained in RPMI supplemented with 10% FCS and glutamine (Life Technologies/Invitrogen).

pLXSN vectors containing human FGFR4-encoding cDNA have been described before (13). The polyclonal MCF7 cell lines expressing FGFR4 were generated by retroviral gene transfer. Amphotropic retroviral supernatants were produced by transfection of Phoenix packaging cells using the appropriate vector constructs by the calcium phosphate/chloroquine method as described previously. At 48 h post-transfection, the tissue culture medium was filtered through a 0.45 μm filter, mixed with polybrene (4 μg/mL final), and used for infection of the cells. Cells were infected three times for at least 4 h and allowed to recover for 24 h with fresh medium. Polyclonal cells stably expressing FGFR4 were selected with G418 (2 mg/mL) for 2 weeks.

To target FGFR4 protein for down-regulation by siRNA, we generated pRetroSuper-FGFR4 constructs. The target sequences of siRNAs used in this article were GAGCAGGAGCTGACAGAGT (si66), CTACCTGCTAGATGTGCTG (siCtrl), and GTGCTCGACCTTGATAGCA (si74). Polyclonal cells carrying pRetroSuper-FGFR4 were selected with puromycin (2 μg/mL) for 2 days.

Reverse transcription-PCR and semiquantitative PCR. RNA was isolated using the RNeasy Mini Kit (Qiagen) and reversely transcribed into cDNA using the AMV reverse transcriptase (Roche) and oligo(dT) primer (Life Technologies). The RNA and cDNA concentration was measured using the NanoDrop ND1000 (PeqLab) system. For the semiquantitative PCR amplification, the following primers (MWG) have been used: 5′-ACCACAGTCCATGCCATCAC-3′ (GAPDH forward) and 5′-TCCACCACCCTGTTGCTGTA-3′ (GAPDH reverse) and 5′-AGAATTCTGCCACCATGTCTCAGAGCAACC-3′ (Bcl-xl forward) and 5′-GGGTGATGTGGAGCTGGGATGTC-3′ (Bcl-xl reverse). PCR products were subjected to electrophoresis on a 2% agarose gel and DNA was visualized by ethidium bromide staining.

Cell lysis, immunoprecipitation, and immunoblotting. Cell cultures were washed with PBS and incubated at 4°C with lysis buffer [50 mmol/L HEPES/NaOH (pH 7.5), 150 mmol/L NaCl, 1 mmol/L EDTA, 10% glycerol, and 1% Triton X-100] supplemented with phosphatase and protease inhibitors (10 mmol/L sodium pyrophosphate, 1 mmol/L phenylmethylsulfonyl fluoride, 2 mmol/L sodium orthovanadate, and 10 μg/mL aprotinin). Cellular debris was removed by centrifugation. Protein concentration measurements were done using the Micro-BCA Protein Assay Kit (Pierce). For immunoprecipitations, whole-cell lysates were combined with antibody and 30 μL protein A/G-Sepharose slurry (GE Healthcare). The samples were incubated for 3 h on a rotation wheel at 4°C. The precipitates were washed three times with HNTG buffer [20 mmol/L HEPES/NaOH (pH 7.5), 150 mmol/L NaCl, 10% glycerol, 0.1% Triton X-100, and 10 mmol/L sodium pyrophosphate] suspended in 3× SDS sample buffer, boiled for 3 min, and subjected to SDS-PAGE. For Western blot analysis, proteins were transferred to nitrocellulose membranes and incubated with the appropriate antibodies. Signals were developed via an enhanced chemiluminescence detection system (Perkin-Elmer). Before reprobing, membranes were stripped with 65 mmol/L Tris-HCl (pH 6.8) containing 2% SDS at 50°C for 1 h. For the quantification of immunoblots, scanned images were analyzed with Aida Image Analyzer software (Raytest).

RNA interference. Transfection of 21-nucleotide siRNA duplexes (Ambion) for targeting endogenous genes was carried out using Lipofectamine 2000 (Invitrogen) and 0.84 μg siRNA duplexes per 6-well plate as described. Transfected cells were assayed on indicated time points. Sequences of siRNA used were FGFR4 5′-GGCUCUUCCGGCAAGUCAAtt-3′ and GL2-luciferase 5′-CGUACGCGGAAUACUUCGAtt-3′. Specific silencing of FGFR4 was confirmed by immunoblot analysis as described before.

Cell death assays. Cell death was measured by counting the percentage of hypodiploid cells using flow cytometry as described previously. Propidium iodide fluorescence was measured in the FL2 channel on a FACSCalibur (BD Biosciences). Cell culture supernatant and trypsinized cells were pooled.

cDNA array preparation. Gene expression was analyzed by hybridization of nylon filter arrays with radioactive targets (cDNA). The homemade arrays comprised 1,100 gene inserts (all protein kinases, protein phosphatases, and other signaling proteins). The vector with different gene inserts was spotted with a biogrid roboter in triplicates.

cDNA array hybridization. Radioactive labeling of the cDNAs was achieved using the Megaprime DNA labeling kit (Amersham Biosciences) and 50 μCi [α-33P]ATP per reaction. The labeled cDNA was purified via the Nucleotide Removal Kit (Qiagen) and incubated with 0.5 mg/mL COT-DNA (Invitrogen) in hybridization buffer (5× SSC, 0.1% SDS) for 5 min at 95°C and 30 min at 68°C to block repetitive sequences in the cDNA. The cDNA was added to prewarmed (68°C) hybridization buffer containing 100 μg/mL tRNA (baker's yeast; Roche Applied Science). The cDNA arrays were incubated in pre-hybridization buffer (5× Denhardt's, 5× SSC, 100 mmol/L Na3PO4, 2 mmol/L Na4P2O7, and 100 μg/mL tRNA) for 4 h and subsequently with the labeled cDNA in hybridization buffer for 16 h. The cDNA was removed, and the cDNA arrays were washed with increasing stringency, dried, and exposed on phosphorimaging plates (Fuji). The plates were read on a FujiBas2500 phosphorimaging device, and the raw spot values (volume) were determined using ArrayVision (Raytest).

Statistical analysis. The raw DNA macroarray intensity data were pre-processed using total intensity normalization. Mean gene expression of 10 independent arrays of MDA-MB-453 cell line and at least 3 independent arrays of doxorubicin-treated clones were compared.

Results

Resistance of MDA-MB-453 breast cancer cells to doxorubicin correlates with FGFR4 up-regulation. We performed an assay to identify antiapoptotic mechanisms in the breast cancer cell line MDA-MB-453. Cells were treated with 0.5 μm doxorubicin for 48 h and surviving clones were isolated after 8 weeks.

On average, a 2-fold up-regulation of FGFR4 mRNA was found in clones surviving the doxorubicin treatment when the gene expression profiles generated by array analysis were compared (Fig. 1A ). About half of the clones showed an up to 4-fold increase in FGFR4 mRNA levels, whereas others showed weak or no increase (Fig. 1B). The FGFR4 expression levels were confirmed by semiquantitative PCR analysis (Supplementary Fig. S1).

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

FGFR4 is up-regulated in doxorubicin-treated MDA-MB-453 clones. A, expression of FGFR4 as determined by array analysis. Mean expression of 10 independent drug treatments of MDA-MB-453 and resistant clones. B, expression of each clone assembled by three independent experiments.

FGFR4 expression affects resistance of cancer cells to doxorubicin. To study the role of FGFR4 in chemoresistance, we used two of the resistant MDA-MB-453 clones (453R1 and 453R9) for stable knockdown of FGFR4 by FGFR4 siRNAs (si66 and si74). Transfection of siRNAs 66 and 74 efficiently suppressed receptor expression in 453R1 (Fig. 2A ) and 453R9 (data not shown) cells and exhibited a significant increase in the sensitivity to apoptosis induction by doxorubicin treatment when compared with control siRNA-infected cells (Fig. 2A).

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

FGFR4 expression affects chemoresistance of cancer cells. A, immunoprecipitation of FGFR4 in 453R1 knockdown cells. Propidium iodide staining of doxorubicin-treated (2 μmol/L for 48 h) 453R1 cells and flow cytometry analysis for apoptosis quantification. B, immunoprecipitation of FGFR4 from MCF7-FGFR4 and MCF7-Mock cell lysates. MCF7-FGFR4 and MCF7-Mock cells were treated with 1 μmol/L doxorubicin for 24 h to induce apoptosis. Propidium iodide-stained cells were quantified by fluorescence-activated cell sorting. C, dose-response assay with different concentrations of doxorubicin for 24 h and measurement of apoptotic cells with propidium iodide staining followed by fluorescence-activated cell sorting. D, induction of apoptosis with the chemotherapeutic drugs cyclophosphamide (CPA), Taxotere (TXT), and cisplatin (CP) for 48 h. Rate of apoptosis was measured by propidium iodide staining followed by fluorescence-activated cell sorting.

Furthermore, we stably overexpressed the FGFR4 in MCF7 breast cancer cells, which do not express FGFR4 endogenously (Fig. 2B). As expected, MCF7-FGFR4-overexpressing cells treated with doxorubicin showed less apoptosis than MCF7-parental control cells (Fig. 2B).

In addition, we analyzed the apoptosis rate of MCF7 cells ectopically overexpressing FGFR4 and 453R1 cells after knockdown of FGFR4 expression at different doxorubicin concentrations. In MCF7-FGFR4 cells, apoptosis was reduced at different doxorubicin concentrations. Consequently, the 453R1 FGFR4 knockdown cells showed higher apoptosis rates compared with control 453R1cells (Fig. 2C).

Finally, we analyzed if ectopic overexpression of FGFR4 in MCF7 or a FGFR4 knockdown in 453R1 cells affects sensitivity against other chemotherapeutic drugs such as cyclophosphamide, cisplatin, or Taxotere. Interestingly, only with cyclophosphamide reduced apoptosis sensitivity was observed in MCF7-FGFR4-overexpressing cells. Accordingly, 453R1 siRNA 66 and 74 cells exhibit a higher apoptosis rate than the control cells when treated with cyclophosphamide (Fig. 2D). However, no effect could be observed when cells were treated with Taxotere and cisplatin. These experiments show that overexpression of FGFR4 increases apoptosis resistance, whereas a stable knockdown of FGFR4 sensitizes cancer cells toward specific chemotherapeutic drugs.

Gene expression analysis of ectopic FGFR4-expressing cells reveals increased Bcl-xl levels. To gain further insight into the molecular mechanism mediating the chemoresistance by FGFR4, we performed a gene expression array analysis of MCF7 cells ectopically expressing FGFR4. Here, we could detect the Bcl-xl expression to be 2-fold up-regulated (Fig. 3A ). We validated the differences in expression levels of Bcl-xl by semiquantitative PCR (Fig. 3A). Furthermore, an increased amount of Bcl-xl protein was detected in the FGFR4-expressing MCF7 cell line (data not shown). One of the FGFR4 downstream signaling pathways is the MAPK cascade. As MAPK cascade is also an important regulator of Bcl-xl expression, MCF7-FGFR4 cells were treated with UO126, a specific MEK inhibitor. The semiquantitative PCR showed that UO126 treatment abolished Bcl-xl expression (Fig. 3B). Therefore, we conclude that Bcl-xl expression is dependent on FGFR4 expression and MAPK activity.

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

FGFR4 governs Bcl-xl expression. A, array analysis of Bcl-xl expression in MCF7-Mock and ectopically expressing FGFR4 cells. Validation by semiquantitative reverse transcription-PCR of Bcl-xl expression in MCF7 cells. B, MCF7-FGFR4 cells were treated with 10 μmol/L U0126 for 6 and 12 h. The cDNA of U0126 treated cells was used to perform reverse transcription-PCR with Bcl-xl primers. The integrity and amount of cDNA used in each reverse transcription-PCR were measured by GAPDH amplification.

Knockdown of FGFR4 affects MAPK signaling pathway and Bcl-xl expression. To proof the concept of FGFR4 mediating drug resistance by activating MAPK cascade and thereby increasing Bcl-xl levels, we used different breast cancer cell lines, which are endogenously expressing high amounts of FGFR4. As expected, the transient knockdown with synthetic siRNA directed against FGFR4 (siFGFR4) in BT-474 breast cancer cells showed a reduced apoptosis rate on several time points (Fig. 4A ).

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Analysis of downstream signaling pathways in transient FGFR4 knockdown cells. A, apoptosis assays of transient FGFR4 knockdown in BT-474 cells treated with 2 μmol/L doxorubicin for indicated time points. Propidium iodide-stained cells were quantified by fluorescence-activated cell sorting. B, immunoprecipitation of FGFR4 in BT-474 and ZR75-1 cells transiently transfected with luciferase (control) or FGFR4 siRNA (siFGFR4). TL immunoblots were done to monitor ERK phosphorylation. Quantification of ERK phosphorylation levels compared with total ERK protein levels. C, FGFR4 knockdown in BT-474 and ZR75-1 cells and TL immunoblot of Bcl-xl. Quantification of Bcl-xl protein levels compared with tubulin protein levels.

On FGFR4 knockdown, we could observe lower phospho-ERK levels at 96 h after transfection than in control experiments (Fig. 4B). However, Akt phosphorylation was unchanged by the knockdown (data not shown).

As expected, the transient knockdown of FGFR4 also affected Bcl-xl expression. One hundred forty-four hours after transfection, we could detect reduced Bcl-xl protein levels (Fig. 4C). These results showed that Bcl-xl expression is partially dependent on FGFR4 expression and could be suppressed by FGFR4 knockdown.

A FGFR4-blocking antibody inhibits FGF19-induced MAPK activation and reduces the chemoresistance of doxorubicin-treated cancer cells. Next, we wanted to address the question if the enhanced chemoresistance is linked with the FGFR4 activity in the different cancer cell lines. For this purpose, we generated a FGFR4-blocking antibody (10F10). We stimulated various breast cancer cell lines, which endogenously express FGFR4, with its specific ligand FGF19. When FGF19 treated, phospho-ERK levels were increased compared to not stimulated control cells. After preincubation of the cell lines with the FGFR4-blocking antibody and subsequent FGF19 stimulation, reduced phospho-ERK levels compared with sole FGF19-treated cancer cell lines could be detected (Fig. 5A ). In these experiments, we clearly could show that the FGFR4-blocking antibody (10F10) is inhibiting the FGFR4-mediated downstream signaling.

Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

Usage of a FGFR4-blocking antibody mediates inhibition of FGF19-induced MAPK activation and decreases the chemoresistance of doxorubicin-treated cancer cells. A, different breast cancer cell lines (BT-474, MDA-MB-361, and ZR75-1) were starved 48 h with 0% FCS/medium. Cells were treated with either FGF19 alone or 10F10 plus FGF19 and compared with the untreated control cells. Stimulation and inhibition of the cells is shown by a TL immunoblot with phospho-ERK antibody. B, apoptosis rate of BT-474, MDA-MB-361, and ZR75-1 breast cancer cell line treated with 2 μmol/L doxorubicin for 48 h and/or different stimuli. C, cDNA macroarray analysis shows the relative FGFR4 expression in various breast cancer cell lines. D, induction of apoptosis with doxorubicin in FGF19-treated or FGF19/10F10-treated breast cancer cell lines showing high FGFR4 expression. Quantification of the apoptosis rates by propidium iodide staining and subsequent fluorescence-activated cell sorting analysis. Apoptosis rate of two cancer cell lines with other origin than mammary gland assessed as described above.

To verify if the blocking of the FGFR4 activity results in increased sensitivity to chemotherapeutic drugs, we incubated the breast cancer cell lines MDA-MB-361, BT-474, and ZR75-1 with doxorubicin, VSV/10F10 antibody, or in different combinations and measured the apoptosis rate after 48 h of doxorubicin treatment. Here, we could show that the blocking of FGFR4 by 10F10 antibody resulted in increased apoptosis rate after doxorubicin induction, whereas the VSV antibody incubation had no effect on apoptosis (Fig. 5B).

In cDNA macroarray experiments, we could observe that FGFR4 is highly expressed in ∼20% of the available breast cancer cell lines, which is consistent with other published data in human cancers (refs. 19, 20; Fig. 5C). High expression of the receptor was observed in the cell lines BT-474, ZR75-1, MDA-MB-361, and MDA-MB-453. Therefore, these cell lines were used to inhibit FGFR4 signaling and to sensitize the cancer cells for chemotherapy. We compared the apoptosis rates of FGF19/doxorubicin-treated cancer cell lines with cell lines additionally treated with FGFR4-blocking antibody. Doxorubicin/FGF19 treatment of the different cell lines induced apoptosis in a low to medium extent, whereas the additional treatment with FGFR4-blocking antibody significantly further increased the apoptosis rates (Fig. 5D): ∼40% in MDA-MB-361, ∼29% in ZR75-1, and ∼10% in BT-474. In MDA-MB-453 cells, we could not detect increased apoptosis with 10F10 antibody treatment. This might be due to the constitutive activation of the FGFR4 in this cell line, which could not be blocked by the 10F10 antibody (data not shown). However, in these experiments, we could show that FGFR4 activity supports resistance of cancer cells to doxorubicin, whereas inversely the inhibition of FGFR4 activation by a FGFR4-blocking antibody (10F10) leads to increased chemosensitivity of cancer cells.

The Oncomine database (21) allows searching for genes, which are up-regulated in tumors, in many published array experiments. For FGFR4, we observed a significant increase in expression in oncogenic transformed tissues compared with normal tissues in six different studies (Supplementary Fig. S2). This shows the frequency of FGFR4 overexpression and the relevance of FGFR4 signaling in human cancers. Therefore, we analyzed the efficacy of the blocking antibody 10F10 in cancer cell lines of other origin than breast tissue. As an example, we used the 769-P kidney carcinoma and the TE671 glioblastoma cell lines. Likewise, additional treatment with the blocking antibody increased the apoptosis rate in these cells (Fig. 5D).

Discussion

Mitotic instability is a hallmark of cancer and allows tumor cells to adapt to the changing microenvironment when drugs are applied. Chemotherapy imposes such a selective process and the tumor cells harboring genetic changes benefit under such conditions and survive (22). We tried to simulate the selective process in normal cell culture conditions by treating MDA-MB-453, a breast cancer cell line, with the commonly used chemotherapeutic drug doxorubicin. The doxorubicin-resistant cancer cells were used to analyze the gene expression changes, which could be advantageous for the cells to withstand doxorubicin treatment. FGFR4 expression was significantly higher in the resistant clones, and as FGFRs are known to provide apoptotic resistance (23), we predicted it to be one of the events that take place during the selection process. This change in expression has presumably occurred at transcriptional level as there was found no gene amplification for FGFR4 in resistant clones (data not shown).

The FGFR4 gene is known to be regulated by two transcription factors Ikaros and HNF1 (24–27) where deregulation could result in increased expression. Therefore, it would be interesting to study if such a regulatory mechanism exists in chemoresistant tumors. A comparison of published gene expression studies revealed the increased expression of FGFR4 in several breast cancer cell lines (Fig. 5C) and oncogenically transformed tissues (Supplementary Fig. S2). Various studies found an overexpression of FGFR4 in breast and other cancer types (19, 20, 25, 27, 28). This, for example, leads to an unfavorable disease progression and reduced survival of the patients with prostate cancer and astrocytomas. We could show an antiapoptotic effect of FGFR4 by overexpression as well as repression of the mRNA by siRNA experiments. This effect could only be observed when cells were treated with doxorubicin or cyclophosphamide, both DNA-damaging agents, suggesting common mechanisms of these drugs. Several polymorphisms of FGFR family members have been reported and linked to different diseases (29–31). Up to now, antiapoptotic effects have only been reported for FGFR1 and FGFR3 (23, 32). Translocation or constitutive active mutations of FGFR1 or FGFR3 result in hyperactivity and can lead to activation or up-regulation of antiapoptotic proteins such as STATs, Mcl1, Bcl2, or Bcl-xl (33, 34), which contribute to cancer cell survival. Inhibition of FGFR1 or FGFR3 by inhibitors, antisense approaches, or short hairpin RNA experiments induced apoptosis in cells of different origin (17, 35). Apart from that, we show that FGFR4 also provides apoptotic resistance.

Although FGFR activation is associated with multiple physiologic effects, the induction or repression of apoptosis is increasingly intriguing considering recent reports. FGFR activation leads to MAPK or phosphatidylinositol 3-kinase signaling depending on the cellular context (12). Recently, it has been reported that FGFR signaling converges the HER-2 signaling and modulates translation (10). Although all these observations have been instrumental in our understanding of FGFR signaling, the majority of them struggles with the intrinsic redundancy of FGFRs and cannot associate specific FGFRs with these effects and therefore warrants a candidate-based approach to study their functions. Our studies overcome these problems as we have used overexpression, siRNA knockdown, specific blocking antibody approaches, and the specific FGFR4 ligand FGF19 (36) to implicate the FGFR4 as an important component of antiapoptotic signaling in cancers during the chemotherapeutic treatment. The Bcl-xl and FGFR4 coexpression in endogenous and overexpressing systems and the direct dependence of Bcl-xl expression on FGFR4 expression is of immense relevance. Bcl-xl as a prominent downstream effector molecule in antiapoptosis is also up-regulated in various tumors such as breast, squamous cell, and liver carcinoma (37–39). It is well known that Bcl-xl is responsible for the accomplishment of chemoresistance of cancer cells against, for example, doxorubicin, leading to decreased apoptosis and increased survival (40, 41).

It is shown that MAPK activation via various signaling pathways leads to the expression of antiapoptotic molecules and the rescue of cells from apoptosis. On the other hand, the MAPK has also been implicated in apoptosis induction actively via up-regulation of caspases, which could be reversed by blocking MEK (42). Therefore, it is attractive to hypothesize that certain growth signals converge the MAPK pathway and, in addition to activating it, lead to up-regulation of both apoptotic and antiapoptotic molecules depending on the cellular context. Furthermore, MAPK signaling is indispensable for up-regulation, as Bcl-xl expression has been reduced to basal levels on UO126, a specific MEK inhibitor, treatment (Fig. 4C).

In principle, STAT3 or STAT5, among other transcription factors, is known to be mainly responsible for activating Bcl-xl transcription in epithelial cancers (43). We have not been able to see any difference in phospho-STAT expression levels in our experiments, which let us predict the existence of another alternative mechanism how FGFR4 signaling controls Bcl-xl expression. In another study, it was shown that Bcl-xl expression is dependent on prolactin stimulation (44). Interestingly, FGF19 stimulation of FGFR4 is increasing the prolactin promoter activity (45). These observations might be the basis to study the link between FGFR4 and Bcl-xl expression. As Bcl-xl expression is very often counteracting, the chemotherapy of cancer patients, small peptide antagonists, or antisense oligonucleotides against Bcl-xl are already used in the clinical therapy to extend the disease-free survival time of cancer patients (46–49).

Our findings open up new possibilities in cancer therapy. Patients can be treated with alternative therapeutic drugs or antibodies against FGFR4 to decrease the receptor tyrosine kinase expression or activity, which leads to the expression of antiapoptotic molecules (e.g., Bcl-xl) during the applied chemotherapy. Consequently, FGFR4 could serve as a prognostic marker and target for novel cotherapies in cancer patients.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

We thank Heike Stubbe for excellent technical support and Dr. Peter Czernilofsky for critically reading the manuscript.

Footnotes

  • ↵3 http://www.who.int/ceh/publications/statistics/cehstathq/en/WHO Cancer Mortality Databank.

  • ↵4 http://www.who.int/mediacentre/factsheets/fs297/en/ Media centre-Cancer.

  • Grant support: National Genome Research Network 2/CancerNet grant 01GS0435.

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

  • Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

  • A. Roidl and H-J. Berger contributed equally as first authors.

  • Received April 7, 2008.
  • Revision received August 18, 2008.
  • Accepted September 29, 2008.

References

  1. ↵
    Bange J, Zwick E, Ullrich A. Molecular targets for breast cancer therapy and prevention. Nat Med 2001;7:548–52.
    OpenUrlCrossRefPubMed
  2. ↵
    Fisher B, Bryant J, Dignam JJ, et al. Tamoxifen, radiation therapy, or both for prevention of ipsilateral breast tumor recurrence after lumpectomy in women with invasive breast cancers of one centimeter or less. J Clin Oncol 2002;20:4141–9.
    OpenUrlAbstract/FREE Full Text
  3. ↵
    Wolff AC, Davidson NE. Primary systemic therapy in operable breast cancer. J Clin Oncol 2000;18:1558–69.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    Linn SC, Pinedo HM, van Ark-Otte J, et al. Expression of drug resistance proteins in breast cancer, in relation to chemotherapy. Int J Cancer 1997;71:787–95.
    OpenUrlCrossRefPubMed
  5. ↵
    Hudziak RM, Lewis GD, Holmes WE, Ullrich A, Shepard HM. Selection for transformation and met protooncogene amplification in NIH3T3 fibroblasts using tumor necrosis factor α. Cell Growth Differ 1990;1:129–34.
    OpenUrlAbstract
  6. ↵
    Abraham R, Schaefer J, Rothe M, Bange J, Knyazev P, Ullrich A. Identification of MMP-15 as an anti-apoptotic factor in cancer cells. J Biol Chem 2005;280:34123–32.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Eswarakumar VP, Lax I, Schlessinger J. Cellular signaling by fibroblast growth factor receptors. Cytokine Growth Factor Rev 2005;16:139–49.
    OpenUrlCrossRefPubMed
  8. ↵
    Ornitz DM, Itoh N. Fibroblast growth factors. Genome Biol 2001;2:3005.
    OpenUrlCrossRefPubMed
  9. ↵
    Cross MJ, Claesson-Welsh L. FGF and VEGF function in angiogenesis: signalling pathways, biological responses and therapeutic inhibition. Trends Pharmacol Sci 2001;22:201–7.
    OpenUrlCrossRefPubMed
  10. ↵
    Koziczak M, Hynes NE. Cooperation between fibroblast growth factor receptor-4 and ErbB2 in regulation of cyclin D1 translation. J Biol Chem 2004;279:50004–11.
    OpenUrlAbstract/FREE Full Text
  11. ↵
    Shaoul E, Reich-Slotky R, Berman B, Ron D. Fibroblast growth factor receptors display both common and distinct signaling pathways. Oncogene 1995;10:1553–61.
    OpenUrlPubMed
  12. ↵
    Dailey L, Ambrosetti D, Mansukhani A, Basilico C. Mechanisms underlying differential responses to FGF signaling. Cytokine Growth Factor Rev 2005;16:233–47.
    OpenUrlCrossRefPubMed
  13. ↵
    Bange J, Prechtl D, Cheburkin Y, et al. Cancer progression and tumor cell motility are associated with the FGFR4 Arg(388) allele. Cancer Res 2002;62:840–7.
    OpenUrlAbstract/FREE Full Text
  14. ↵
    Muenke M, Schell U. Fibroblast-growth-factor receptor mutations in human skeletal disorders. Trends Genet 1995;11:308–13.
    OpenUrlCrossRefPubMed
  15. ↵
    Giri D, Ropiquet F, Ittmann M. Alterations in expression of basic fibroblast growth factor (FGF) 2 and its receptor FGFR-1 in human prostate cancer. Clin Cancer Res 1999;5:1063–71.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Yamaguchi F, Saya H, Bruner JM, Morrison RS. Differential expression of two fibroblast growth factor-receptor genes is associated with malignant progression in human astrocytomas. Proc Natl Acad Sci U S A 1994;91:484–8.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Trudel S, Ely S, Farooqi Y, et al. Inhibition of fibroblast growth factor receptor 3 induces differentiation and apoptosis in t(4;14) myeloma. Blood 2004;103:3521–8.
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Wang J, Stockton DW, Ittmann M. The fibroblast growth factor receptor-4 Arg388 allele is associated with prostate cancer initiation and progression. Clin Cancer Res 2004;10:6169–78.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Jaakkola S, Salmikangas P, Nylund S, et al. Amplification of fgfr4 gene in human breast and gynecological cancers. Int J Cancer 1993;54:378–82.
    OpenUrlCrossRefPubMed
  20. ↵
    Penault-Llorca F, Bertucci F, Adelaide J, et al. Expression of FGF and FGF receptor genes in human breast cancer. Int J Cancer 1995;61:170–6.
    OpenUrlPubMed
  21. ↵
    Rhodes DR, Yu J, Shanker K, et al. ONCOMINE: a cancer microarray database and integrated data-mining platform. Neoplasia 2004;6:1–6.
    OpenUrlCrossRefPubMed
  22. ↵
    Kops GJ, Weaver BA, Cleveland DW. On the road to cancer: aneuploidy and the mitotic checkpoint. Nat Rev Cancer 2005;5:773–85.
    OpenUrlCrossRefPubMed
  23. ↵
    L'Hote CG, Knowles MA. Cell responses to FGFR3 signalling: growth, differentiation and apoptosis. Exp Cell Res 2005;304:417–31.
    OpenUrlCrossRefPubMed
  24. ↵
    Ezzat S, Yu S, Asa SL. Ikaros isoforms in human pituitary tumors: distinct localization, histone acetylation, and activation of the 5′ fibroblast growth factor receptor-4 promoter. Am J Pathol 2003;163:1177–84.
    OpenUrlPubMed
  25. ↵
    Shah RN, Ibbitt JC, Alitalo K, Hurst HC. FGFR4 overexpression in pancreatic cancer is mediated by an intronic enhancer activated by HNF1α. Oncogene 2002;21:8251–61.
    OpenUrlCrossRefPubMed
  26. Yu S, Asa SL, Ezzat S. Fibroblast growth factor receptor 4 is a target for the zinc-finger transcription factor Ikaros in the pituitary. Mol Endocrinol 2002;16:1069–78.
    OpenUrlCrossRefPubMed
  27. ↵
    Yamada SM, Yamada S, Hayashi Y, Takahashi H, Teramoto A, Matsumoto K. Fibroblast growth factor receptor (FGFR) 4 correlated with the malignancy of human astrocytomas. Neurol Res 2002;24:244–8.
    OpenUrlCrossRefPubMed
  28. ↵
    Gowardhan B, Douglas DA, Mathers ME, et al. Evaluation of the fibroblast growth factor system as a potential target for therapy in human prostate cancer. Br J Cancer 2005;92:320–7.
    OpenUrlPubMed
  29. ↵
    Reardon W, Winter RM, Rutland P, Pulleyn LJ, Jones BM, Malcolm S. Mutations in the fibroblast growth factor receptor 2 gene cause Crouzon syndrome. Nat Genet 1994;8:98–103.
    OpenUrlCrossRefPubMed
  30. Schell U, Hehr A, Feldman GJ, et al. Mutations in FGFR1 and FGFR2 cause familial and sporadic Pfeiffer syndrome. Hum Mol Genet 1995;4:323–8.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Shiang R, Thompson LM, Zhu YZ, et al. Mutations in the transmembrane domain of FGFR3 cause the most common genetic form of dwarfism, achondroplasia. Cell 1994;78:335–42.
    OpenUrlCrossRefPubMed
  32. ↵
    Xian W, Schwertfeger KL, Vargo-Gogola T, Rosen JM. Pleiotropic effects of FGFR1 on cell proliferation, survival, and migration in a 3D mammary epithelial cell model. J Cell Biol 2005;171:663–73.
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Hart KC, Robertson SC, Donoghue DJ. Identification of tyrosine residues in constitutively activated fibroblast growth factor receptor 3 involved in mitogenesis, Stat activation, and phosphatidylinositol 3-kinase activation. Mol Biol Cell 2001;12:931–42.
    OpenUrlAbstract/FREE Full Text
  34. ↵
    Hart KC, Robertson SC, Kanemitsu MY, Meyer AN, Tynan JA, Donoghue DJ. Transformation and Stat activation by derivatives of FGFR1, FGFR3, and FGFR4. Oncogene 2000;19:3309–20.
    OpenUrlCrossRefPubMed
  35. ↵
    Zhu L, Somlo G, Zhou B, et al. Fibroblast growth factor receptor 3 inhibition by short hairpin RNAs leads to apoptosis in multiple myeloma. Mol Cancer Ther 2005;4:787–98.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Xie MH, Holcomb I, Deuel B, et al. FGF-19, a novel fibroblast growth factor with unique specificity for FGFR4. Cytokine 1999;11:729–35.
    OpenUrlCrossRefPubMed
  37. ↵
    Olopade OI, Adeyanju MO, Safa AR, et al. Overexpression of BCL-x protein in primary breast cancer is associated with high tumor grade and nodal metastases. Cancer J Sci Am 1997;3:230–7.
    OpenUrlPubMed
  38. Pena JC, Thompson CB, Recant W, Vokes EE, Rudin CM. Bcl-xL and Bcl-2 expression in squamous cell carcinoma of the head and neck. Cancer 1999;85:164–70.
    OpenUrlCrossRefPubMed
  39. ↵
    Watanabe J, Kushihata F, Honda K, et al. Prognostic significance of Bcl-xL in human hepatocellular carcinoma. Surgery 2004;135:604–12.
    OpenUrlCrossRefPubMed
  40. ↵
    Williams J, Lucas PC, Griffith KA, et al. Expression of Bcl-xL in ovarian carcinoma is associated with chemoresistance and recurrent disease. Gynecol Oncol 2005;96:287–95.
    OpenUrlCrossRefPubMed
  41. ↵
    Luo D, Cheng SC, Xie H, Xie Y. Effects of Bcl-2 and Bcl-XL protein levels on chemoresistance of hepatoblastoma HepG2 cell line. Biochem Cell Biol 2000;78:119–26.
    OpenUrlCrossRefPubMed
  42. ↵
    Sebolt-Leopold JS, Herrera R. Targeting the mitogen-activated protein kinase cascade to treat cancer. Nat Rev Cancer 2004;4:937–47.
    OpenUrlCrossRefPubMed
  43. ↵
    Yu H, Jove R. The STATs of cancer—new molecular targets come of age. Nat Rev Cancer 2004;4:97–105.
    OpenUrlCrossRefPubMed
  44. ↵
    Kochendoerfer SK, Krishnan N, Buckley DJ, Buckley AR. Prolactin regulation of Bcl-2 family members: increased expression of bcl-xL but not mcl-1 or bad in Nb2-T cells. J Endocrinol 2003;178:265–73.
    OpenUrlAbstract
  45. ↵
    Yu S, Zheng L, Asa SL, Ezzat S. Fibroblast growth factor receptor 4 (FGFR4) mediates signaling to the prolactin but not the FGFR4 promoter. Am J Physiol Endocrinol Metab 2002;283:E490–5.
    OpenUrlAbstract/FREE Full Text
  46. ↵
    Olie RA, Hafner C, Kuttel R, et al. Bcl-2 and bcl-xL antisense oligonucleotides induce apoptosis in melanoma cells of different clinical stages. J Invest Dermatol 2002;118:505–12.
    OpenUrlCrossRefPubMed
  47. Ozvaran MK, Cao XX, Miller SD, Monia BA, Hong WK, Smythe WR. Antisense oligonucleotides directed at the bcl-xl gene product augment chemotherapy response in mesothelioma. Mol Cancer Ther 2004;3:545–50.
    OpenUrlAbstract/FREE Full Text
  48. Simoes-Wust AP, Schurpf T, Hall J, Stahel RA, Zangemeister-Wittke U. Bcl-2/bcl-xL bispecific antisense treatment sensitizes breast carcinoma cells to doxorubicin, paclitaxel and cyclophosphamide. Breast Cancer Res Treat 2002;76:157–66.
    OpenUrlCrossRefPubMed
  49. ↵
    Wang S, Yang D, Lippman ME. Targeting Bcl-2 and Bcl-XL with nonpeptidic small-molecule antagonists. Semin Oncol 2003;30:133–42.
    OpenUrlCrossRefPubMed
View Abstract
PreviousNext
Back to top
Clinical Cancer Research: 15 (6)
March 2009
Volume 15, Issue 6
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Clinical Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Resistance to Chemotherapy Is Associated with Fibroblast Growth Factor Receptor 4 Up-Regulation
(Your Name) has forwarded a page to you from Clinical Cancer Research
(Your Name) thought you would be interested in this article in Clinical Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Resistance to Chemotherapy Is Associated with Fibroblast Growth Factor Receptor 4 Up-Regulation
Andreas Roidl, Hans-Jürgen Berger, Sushil Kumar, Johannes Bange, Pjotr Knyazev and Axel Ullrich
Clin Cancer Res March 15 2009 (15) (6) 2058-2066; DOI: 10.1158/1078-0432.CCR-08-0890

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Resistance to Chemotherapy Is Associated with Fibroblast Growth Factor Receptor 4 Up-Regulation
Andreas Roidl, Hans-Jürgen Berger, Sushil Kumar, Johannes Bange, Pjotr Knyazev and Axel Ullrich
Clin Cancer Res March 15 2009 (15) (6) 2058-2066; DOI: 10.1158/1078-0432.CCR-08-0890
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Disclosure of Potential Conflicts of Interest
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • TAZ is a Potent Mediator of aRMS Tumorigenesis
  • Targeting HER2 with Osimertinib in NSCLC
  • Combined VEGF/EGFR Inhibition
Show more Cancer Therapy: Preclinical
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • CCR Focus Archive
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Clinical Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Clinical Cancer Research
eISSN: 1557-3265
ISSN: 1078-0432

Advertisement