Skip to main content
Top
Published in: BMC Cancer 1/2015

Open Access 01-12-2015 | Research article

The role of HGF/MET and FGF/FGFR in fibroblast-derived growth stimulation and lapatinib-resistance of esophageal squamous cell carcinoma

Authors: Shin Saito, Kazue Morishima, Takashi Ui, Hiroko Hoshino, Daisuke Matsubara, Shumpei Ishikawa, Hiroyuki Aburatani, Masashi Fukayama, Yoshinori Hosoya, Naohiro Sata, Alan K Lefor, Yoshikazu Yasuda, Toshiro Niki

Published in: BMC Cancer | Issue 1/2015

Login to get access

Abstract

Background

Although advanced esophageal squamous-cell carcinoma (ESCC) is treated using a multidisciplinary approach, outcomes remain unsatisfactory. The microenvironment of cancer cells has recently been shown to strongly influence the biologic properties of malignancies. We explored the effect of supernatant from esophageal fibroblasts on the cell growth and chemo-resistance of ESCC cell lines.

Methods

We used 22 ESCC cell lines, isolated primary human esophageal fibroblasts and immortalized fibroblasts. We first examined cell proliferation induced by fibroblast supernatant. The effect of supernatant was evaluated to determine whether paracrine signaling induced by fibroblasts can influence the proliferation of cancer cells. Next, we examined the effects of adding growth factors HGF, FGF1, FGF7, and FGF10, to the culture medium of cancer cells. These growth factors are assumed to be present in the culture supernatants of fibroblasts and may exert a paracrine effect on the proliferation of cancer cells. We also examined the intrinsic role of HGF/MET and FGFs/FGFR in ESCC proliferation. In addition, we examined the inhibitory effect of lapatinib on ESCC cell lines and studied whether the fibroblast supernatants affect the inhibitory effect of lapatinib on ESCC cell proliferation. Finally, we tested whether the FGFR inhibitor PD-173074 could eliminate the rescue effect against lapatinib that was induced by fibroblast supernatants.

Results

The addition of fibroblast supernatant induces cell proliferation in the majority of cell lines tested. The results of experiments to evaluate the effects of adding growth factors and kinase inhibitors suggests that the stimulating effect of fibroblasts was attributable in part to HGF/MET or FGF/FGFR. The results also indicate diversity in the degree of dependence on HGF/MET and FGF/FGFR among the cell lines. Though lapanitib at 1 μM inhibits cell proliferation by more than 50% in the majority of the ESCC cell lines, fibroblast supernatant can rescue the growth inhibition of ESCC cells. However, the rescue effect is abrogated by co-treatment with FGFR inhibitor.

Conclusion

These results demonstrate that cell growth of ESCC depends on diverse receptor tyrosine kinase signaling, in both cell-autonomous and cell-non-autonomous manners. The combined inhibition of these signals may hold promise for the treatment of ESCC.
Literature
1.
go back to reference Pennathur A, Gibson MK, Jobe BA, Luketich JD. Oesophageal carcinoma. Lancet. 2013;381(9864):400–12.CrossRefPubMed Pennathur A, Gibson MK, Jobe BA, Luketich JD. Oesophageal carcinoma. Lancet. 2013;381(9864):400–12.CrossRefPubMed
2.
3.
go back to reference Enzinger PC, Mayer RJ. Esophageal cancer. N Eng J Med. 2003;349(23):2241–52.CrossRef Enzinger PC, Mayer RJ. Esophageal cancer. N Eng J Med. 2003;349(23):2241–52.CrossRef
4.
go back to reference Ozawa S, Tachimori Y, Baba H, Fujishiro M, Matsubara H, Numasaki H, et al. Comprehensive Registry of Esophageal Cancer in Japan, 2003. Esophagus. 2011;8(1):9–29.CrossRef Ozawa S, Tachimori Y, Baba H, Fujishiro M, Matsubara H, Numasaki H, et al. Comprehensive Registry of Esophageal Cancer in Japan, 2003. Esophagus. 2011;8(1):9–29.CrossRef
5.
go back to reference Chang CY, Cook MB, Lee YC, Lin JT, Ando T, Bhatia S, et al. Current status of Barrett’s esophagus research in Asia. J Gastroenterol Hepatol. 2011;26(2):240–6.CrossRefPubMedPubMedCentral Chang CY, Cook MB, Lee YC, Lin JT, Ando T, Bhatia S, et al. Current status of Barrett’s esophagus research in Asia. J Gastroenterol Hepatol. 2011;26(2):240–6.CrossRefPubMedPubMedCentral
6.
go back to reference Miyazaki T, Inose T, Tanaka N, Yokobori T, Suzuki S, Ozawa D, et al. Management of Barrett’s esophageal carcinoma. Surg Today. 2013;43(4):353–60.CrossRefPubMed Miyazaki T, Inose T, Tanaka N, Yokobori T, Suzuki S, Ozawa D, et al. Management of Barrett’s esophageal carcinoma. Surg Today. 2013;43(4):353–60.CrossRefPubMed
7.
go back to reference Kroep S, Lansdorp-Vogelaar I, Rubenstein JH, Lemmens VE, van Heijningen EB, Aragones N, et al. Comparing trends in esophageal adenocarcinoma incidence and lifestyle factors between the United States, Spain, and the Netherlands. Am J Gastroenterol. 2014;109(3):336–43. quiz 335, 344.CrossRefPubMed Kroep S, Lansdorp-Vogelaar I, Rubenstein JH, Lemmens VE, van Heijningen EB, Aragones N, et al. Comparing trends in esophageal adenocarcinoma incidence and lifestyle factors between the United States, Spain, and the Netherlands. Am J Gastroenterol. 2014;109(3):336–43. quiz 335, 344.CrossRefPubMed
8.
go back to reference American Cancer Society. Cancer Facts & Figures 2013. Atlanta: American Cancer Society; 2013. American Cancer Society. Cancer Facts & Figures 2013. Atlanta: American Cancer Society; 2013.
9.
go back to reference Su C, Chen Z, Luo H, Su Y, Liu W, Cai L, et al. Different patterns of NF-kappaB and Notch1 signaling contribute to tumor-induced lymphangiogenesis of esophageal squamous cell carcinoma. J Exp Clin Cancer Res. 2011;30:85.CrossRefPubMedPubMedCentral Su C, Chen Z, Luo H, Su Y, Liu W, Cai L, et al. Different patterns of NF-kappaB and Notch1 signaling contribute to tumor-induced lymphangiogenesis of esophageal squamous cell carcinoma. J Exp Clin Cancer Res. 2011;30:85.CrossRefPubMedPubMedCentral
10.
go back to reference Chen JW, Xie JD, Ling YH, Li P, Yan SM, Xi SY, et al. The prognostic effect of perineural invasion in esophageal squamous cell carcinoma. BMC Cancer. 2014;14(1):313.CrossRefPubMedPubMedCentral Chen JW, Xie JD, Ling YH, Li P, Yan SM, Xi SY, et al. The prognostic effect of perineural invasion in esophageal squamous cell carcinoma. BMC Cancer. 2014;14(1):313.CrossRefPubMedPubMedCentral
11.
go back to reference Fujita H. Present status of esophageal cancer and its treatment in Japan. Ann Thorac Crdiovasc Surg. 2004;10(3):135–9. Fujita H. Present status of esophageal cancer and its treatment in Japan. Ann Thorac Crdiovasc Surg. 2004;10(3):135–9.
12.
go back to reference Higuchi K, Koizumi W, Tanabe S, Sasaki T, Katada C, Azuma M, et al. Current management of esophageal squamous-cell carcinoma in Japan and other countries. Gastrointest Cancer Res: GCR. 2009;3(4):153–61.PubMedPubMedCentral Higuchi K, Koizumi W, Tanabe S, Sasaki T, Katada C, Azuma M, et al. Current management of esophageal squamous-cell carcinoma in Japan and other countries. Gastrointest Cancer Res: GCR. 2009;3(4):153–61.PubMedPubMedCentral
13.
go back to reference Mueller MM, Fusenig NE. Friends or foes - bipolar effects of the tumour stroma in cancer. Nat Rev Cancer. 2004;4(11):839–49.CrossRefPubMed Mueller MM, Fusenig NE. Friends or foes - bipolar effects of the tumour stroma in cancer. Nat Rev Cancer. 2004;4(11):839–49.CrossRefPubMed
14.
go back to reference Liotta LA, Kohn EC. The microenvironment of the tumour-host interface. Nature. 2001;411(6835):375–9.CrossRefPubMed Liotta LA, Kohn EC. The microenvironment of the tumour-host interface. Nature. 2001;411(6835):375–9.CrossRefPubMed
15.
go back to reference De Wever O, Mareel M. Role of tissue stroma in cancer cell invasion. J Pathol. 2003;200(4):429–47.CrossRefPubMed De Wever O, Mareel M. Role of tissue stroma in cancer cell invasion. J Pathol. 2003;200(4):429–47.CrossRefPubMed
16.
go back to reference Noguchi M, Morikawa A, Kawasaki M, Matsuno Y, Yamada T, Hirohashi S, et al. Small Adenocarcinoma of the Lung. Histologic characteristics and prognosis. Cancer. 1995;75(12):2844–52.CrossRefPubMed Noguchi M, Morikawa A, Kawasaki M, Matsuno Y, Yamada T, Hirohashi S, et al. Small Adenocarcinoma of the Lung. Histologic characteristics and prognosis. Cancer. 1995;75(12):2844–52.CrossRefPubMed
17.
go back to reference Hasebe T, Tsuda H, Hirohashi S, Shimosato Y, Iwai M, Imoto S, et al. Fibrotic focus in invasive ductal carcinoma: an indicator of high tumor aggressiveness. Jpn J Cancer Res. 1996;87(4):385–94.CrossRefPubMed Hasebe T, Tsuda H, Hirohashi S, Shimosato Y, Iwai M, Imoto S, et al. Fibrotic focus in invasive ductal carcinoma: an indicator of high tumor aggressiveness. Jpn J Cancer Res. 1996;87(4):385–94.CrossRefPubMed
18.
go back to reference Halvorsen TB, Seim E. Association between invasiveness, inflammatory reaction, desmoplasia and survival in colorectal cancer. J Clin Pathol. 1989;42(2):162–6.CrossRefPubMedPubMedCentral Halvorsen TB, Seim E. Association between invasiveness, inflammatory reaction, desmoplasia and survival in colorectal cancer. J Clin Pathol. 1989;42(2):162–6.CrossRefPubMedPubMedCentral
19.
go back to reference Noma K, Smalley KS, Lioni M, Naomoto Y, Tanaka N, El-Deiry W, et al. The essential role of fibroblasts in esophageal squamous cell carcinoma-induced angiogenesis. Gastroenterology. 2008;134(7):1981–93.CrossRefPubMedPubMedCentral Noma K, Smalley KS, Lioni M, Naomoto Y, Tanaka N, El-Deiry W, et al. The essential role of fibroblasts in esophageal squamous cell carcinoma-induced angiogenesis. Gastroenterology. 2008;134(7):1981–93.CrossRefPubMedPubMedCentral
20.
go back to reference Okawa T, Michaylira CZ, Kalabis J, Stairs DB, Nakagawa H, Andl CD, et al. The functional interplay between EGFR overexpression, hTERT activation, and p53 mutation in esophageal epithelial cells with activation of stromal fibroblasts induces tumor development, invasion, and differentiation. Genes Dev. 2007;21(21):2788–803.CrossRefPubMedPubMedCentral Okawa T, Michaylira CZ, Kalabis J, Stairs DB, Nakagawa H, Andl CD, et al. The functional interplay between EGFR overexpression, hTERT activation, and p53 mutation in esophageal epithelial cells with activation of stromal fibroblasts induces tumor development, invasion, and differentiation. Genes Dev. 2007;21(21):2788–803.CrossRefPubMedPubMedCentral
22.
go back to reference Wojta J, Kaun C, Breuss JM, Koshelnick Y, Beckmann R, Hattey E, et al. Hepatocyte growth factor increases expression of vascular endothelial growth factor and plasminogen activator inhibitor-1 in human keratinocytes and the vascular endothelial growth factor receptor flk-1 in human endothelial cells. Lab Invest. 1999;79(4):427–38.PubMed Wojta J, Kaun C, Breuss JM, Koshelnick Y, Beckmann R, Hattey E, et al. Hepatocyte growth factor increases expression of vascular endothelial growth factor and plasminogen activator inhibitor-1 in human keratinocytes and the vascular endothelial growth factor receptor flk-1 in human endothelial cells. Lab Invest. 1999;79(4):427–38.PubMed
23.
go back to reference Grugan KD, Miller CG, Yao Y, Michaylira CZ, Ohashi S, Klein-Szanto AJ, et al. Fibroblast-secreted hepatocyte growth factor plays a functional role in esophageal squamous cell carcinoma invasion. Proc Natl Acad Sci U S A. 2010;107(24):11026–31.CrossRefPubMedPubMedCentral Grugan KD, Miller CG, Yao Y, Michaylira CZ, Ohashi S, Klein-Szanto AJ, et al. Fibroblast-secreted hepatocyte growth factor plays a functional role in esophageal squamous cell carcinoma invasion. Proc Natl Acad Sci U S A. 2010;107(24):11026–31.CrossRefPubMedPubMedCentral
24.
go back to reference Yoshino M, Ishiwata T, Watanabe M, Matsunobu T, Komine O, Ono Y, et al. Expression and roles of keratinocyte growth factor and its receptor in esophageal cancer cells. Int J Oncol. 2007;31(4):721–8.PubMed Yoshino M, Ishiwata T, Watanabe M, Matsunobu T, Komine O, Ono Y, et al. Expression and roles of keratinocyte growth factor and its receptor in esophageal cancer cells. Int J Oncol. 2007;31(4):721–8.PubMed
25.
go back to reference Mohammadi M, Olsen SK, Ibrahimi OA. Structural basis for fibroblast growth factor receptor activation. Cytokine Growth Factor Rev. 2005;16(2):107–37.CrossRefPubMed Mohammadi M, Olsen SK, Ibrahimi OA. Structural basis for fibroblast growth factor receptor activation. Cytokine Growth Factor Rev. 2005;16(2):107–37.CrossRefPubMed
26.
go back to reference Igarashi M, Finch PW, Aaronson SA. Characterization of recombinant human fibroblast growth factor (FGF)-10 reveals functional similarities with keratinocyte growth factor (FGF-7). J Biol Chem. 1998;273(21):13230–5.CrossRefPubMed Igarashi M, Finch PW, Aaronson SA. Characterization of recombinant human fibroblast growth factor (FGF)-10 reveals functional similarities with keratinocyte growth factor (FGF-7). J Biol Chem. 1998;273(21):13230–5.CrossRefPubMed
27.
go back to reference Zhang C, Fu L, Fu J, Hu L, Yang H, Rong TH, et al. Fibroblast growth factor receptor 2-positive fibroblasts provide a suitable microenvironment for tumor development and progression in esophageal carcinoma. Clin Cancer Res. 2009;15(12):4017–27.CrossRefPubMed Zhang C, Fu L, Fu J, Hu L, Yang H, Rong TH, et al. Fibroblast growth factor receptor 2-positive fibroblasts provide a suitable microenvironment for tumor development and progression in esophageal carcinoma. Clin Cancer Res. 2009;15(12):4017–27.CrossRefPubMed
28.
go back to reference Baselga J. Targeting tyrosine kinases in cancer: the second wave. Science. 2006;312(5777):1175–8.CrossRefPubMed Baselga J. Targeting tyrosine kinases in cancer: the second wave. Science. 2006;312(5777):1175–8.CrossRefPubMed
29.
go back to reference Wood ER, Truesdale AT, McDonald OB, Yuan D, Hassell A, Dickerson SH, et al. A unique structure for epidermal growth factor receptor bound to GW572016 (Lapatinib): relationships among protein conformation, inhibitor off-rate, and receptor activity in tumor cells. Cancer Res. 2004;64(18):6652–9.CrossRefPubMed Wood ER, Truesdale AT, McDonald OB, Yuan D, Hassell A, Dickerson SH, et al. A unique structure for epidermal growth factor receptor bound to GW572016 (Lapatinib): relationships among protein conformation, inhibitor off-rate, and receptor activity in tumor cells. Cancer Res. 2004;64(18):6652–9.CrossRefPubMed
30.
go back to reference Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 2006;355(26):2733–43.CrossRefPubMed Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, et al. Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 2006;355(26):2733–43.CrossRefPubMed
31.
go back to reference Konecny GE, Pegram MD, Venkatesan N, Finn R, Yang G, Rahmeh M, et al. Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer Res. 2006;66(3):1630–9.CrossRefPubMed Konecny GE, Pegram MD, Venkatesan N, Finn R, Yang G, Rahmeh M, et al. Activity of the dual kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing and trastuzumab-treated breast cancer cells. Cancer Res. 2006;66(3):1630–9.CrossRefPubMed
32.
go back to reference Oshima Y, Tanaka H, Murakami H, Ito Y, Furuya T, Kondo E, et al. Lapatinib sensitivities of two novel trastuzumab-resistant HER2 gene-amplified gastric cancer cell lines. Gastric Cancer. 2013. doi:10.1007/s10120-013-0290-6. Oshima Y, Tanaka H, Murakami H, Ito Y, Furuya T, Kondo E, et al. Lapatinib sensitivities of two novel trastuzumab-resistant HER2 gene-amplified gastric cancer cell lines. Gastric Cancer. 2013. doi:10.​1007/​s10120-013-0290-6.
33.
go back to reference Mimura K, Kono K, Maruyama T, Watanabe M, Izawa S, Shiba S, et al. Lapatinib inhibits receptor phosphorylation and cell growth and enhances antibody-dependent cellular cytotoxicity of EGFR- and HER2-overexpressing esophageal cancer cell lines. Int J Cancer. 2011;129(10):2408–16.CrossRefPubMed Mimura K, Kono K, Maruyama T, Watanabe M, Izawa S, Shiba S, et al. Lapatinib inhibits receptor phosphorylation and cell growth and enhances antibody-dependent cellular cytotoxicity of EGFR- and HER2-overexpressing esophageal cancer cell lines. Int J Cancer. 2011;129(10):2408–16.CrossRefPubMed
34.
go back to reference Saito S, Morishima K, Ui T, Matsubara D, Tamura T, Oguni S, et al. Stromal fibroblasts are predictors of disease-related mortality in esophageal squamous cell carcinoma. Oncol Rep. 2014;32(1):348–54.PubMed Saito S, Morishima K, Ui T, Matsubara D, Tamura T, Oguni S, et al. Stromal fibroblasts are predictors of disease-related mortality in esophageal squamous cell carcinoma. Oncol Rep. 2014;32(1):348–54.PubMed
35.
go back to reference Underwood TJ, Derouet MF, White MJ, Noble F, Moutasim KA, Smith E, et al. A comparison of primary oesophageal squamous epithelial cells with HET-1A in organotypic culture. Biol Cell. 2010;102(12):635–44.CrossRefPubMed Underwood TJ, Derouet MF, White MJ, Noble F, Moutasim KA, Smith E, et al. A comparison of primary oesophageal squamous epithelial cells with HET-1A in organotypic culture. Biol Cell. 2010;102(12):635–44.CrossRefPubMed
36.
go back to reference Andl CD, Mizushima T, Nakagawa H, Oyama K, Harada H, Chruma K, et al. Epidermal growth factor receptor mediates increased cell proliferation, migration, and aggregation in esophageal keratinocytes in vitro and in vivo. J Bio Chem. 2003;278(3):1824–30.CrossRef Andl CD, Mizushima T, Nakagawa H, Oyama K, Harada H, Chruma K, et al. Epidermal growth factor receptor mediates increased cell proliferation, migration, and aggregation in esophageal keratinocytes in vitro and in vivo. J Bio Chem. 2003;278(3):1824–30.CrossRef
37.
go back to reference Haga K, Ohno S, Yugawa T, Narisawa-Saito M, Fujita M, Sakamoto M, et al. Efficient immortalization of primary human cells by p16INK4a-specific short hairpin RNA or Bmi-1, combined with introduction of hTERT. Cancer Sci. 2007;98(2):147–54.CrossRefPubMed Haga K, Ohno S, Yugawa T, Narisawa-Saito M, Fujita M, Sakamoto M, et al. Efficient immortalization of primary human cells by p16INK4a-specific short hairpin RNA or Bmi-1, combined with introduction of hTERT. Cancer Sci. 2007;98(2):147–54.CrossRefPubMed
38.
go back to reference Matsubara D, Ishikawa S, Sachiko O, Aburatani H, Fukayama M, Niki T. Co-activation of epidermal growth factor receptor and c-MET defines a distinct subset of lung adenocarcinomas. Am J Pathol. 2010;177(5):2191–204.CrossRefPubMedPubMedCentral Matsubara D, Ishikawa S, Sachiko O, Aburatani H, Fukayama M, Niki T. Co-activation of epidermal growth factor receptor and c-MET defines a distinct subset of lung adenocarcinomas. Am J Pathol. 2010;177(5):2191–204.CrossRefPubMedPubMedCentral
39.
go back to reference Edgar R, Domrachev M, Lash AE. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30(1):207–10.CrossRefPubMedPubMedCentral Edgar R, Domrachev M, Lash AE. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30(1):207–10.CrossRefPubMedPubMedCentral
40.
go back to reference Ren Y, Cao B, Law S, Xie Y, Lee PY, Cheung L, et al. Hepatocyte growth factor promotes cancer cell migration and angiogenic factors expression: a prognostic marker of human esophageal squamous cell carcinomas. Clin Cancer Res. 2005;11(17):6190–7.CrossRefPubMed Ren Y, Cao B, Law S, Xie Y, Lee PY, Cheung L, et al. Hepatocyte growth factor promotes cancer cell migration and angiogenic factors expression: a prognostic marker of human esophageal squamous cell carcinomas. Clin Cancer Res. 2005;11(17):6190–7.CrossRefPubMed
42.
go back to reference Shaul YD, Seger R. The MEK/ERK cascade: from signaling specificity to diverse functions. Biochim Biophys Acta. 2007;1773(8):1213–26.CrossRefPubMed Shaul YD, Seger R. The MEK/ERK cascade: from signaling specificity to diverse functions. Biochim Biophys Acta. 2007;1773(8):1213–26.CrossRefPubMed
43.
go back to reference Fournier NM, Lee B, Banasr M, Elsayed M, Duman RS. Vascular endothelial growth factor regulates adult hippocampal cell proliferation through MEK/ERK- and PI3K/Akt-dependent signaling. Neuropharmacology. 2012;63(4):642–52.CrossRefPubMedPubMedCentral Fournier NM, Lee B, Banasr M, Elsayed M, Duman RS. Vascular endothelial growth factor regulates adult hippocampal cell proliferation through MEK/ERK- and PI3K/Akt-dependent signaling. Neuropharmacology. 2012;63(4):642–52.CrossRefPubMedPubMedCentral
44.
go back to reference Li BS, Ma W, Zhang L, Barker JL, Stenger DA, Pant HC. Activation of phosphatidylinositol-3 kinase (PI-3 K) and extracellular regulated kinases (Erk1/2) is involved in muscarinic receptor-mediated DNA synthesis in neural progenitor cells. J Neurosci. 2001;21(5):1569–79.PubMed Li BS, Ma W, Zhang L, Barker JL, Stenger DA, Pant HC. Activation of phosphatidylinositol-3 kinase (PI-3 K) and extracellular regulated kinases (Erk1/2) is involved in muscarinic receptor-mediated DNA synthesis in neural progenitor cells. J Neurosci. 2001;21(5):1569–79.PubMed
45.
go back to reference Wang B, Gao Y, Xiao Z, Chen B, Han J, Zhang J, et al. Erk1/2 promotes proliferation and inhibits neuronal differentiation of neural stem cells. Neurosci Lett. 2009;461(3):252–7.CrossRefPubMed Wang B, Gao Y, Xiao Z, Chen B, Han J, Zhang J, et al. Erk1/2 promotes proliferation and inhibits neuronal differentiation of neural stem cells. Neurosci Lett. 2009;461(3):252–7.CrossRefPubMed
46.
go back to reference Bhosle J, Kiakos K, Porter AC, Wu J, Makris A, Hartley JA, et al. Treatment with gefitinib or lapatinib induces drug resistance through downregulation of topoisomerase IIalpha expression. Mol Cancer Ther. 2013;12(12):2897–908.CrossRefPubMed Bhosle J, Kiakos K, Porter AC, Wu J, Makris A, Hartley JA, et al. Treatment with gefitinib or lapatinib induces drug resistance through downregulation of topoisomerase IIalpha expression. Mol Cancer Ther. 2013;12(12):2897–908.CrossRefPubMed
47.
go back to reference Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2(2):127–37.CrossRefPubMed Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2(2):127–37.CrossRefPubMed
48.
go back to reference 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(21):6630–8.CrossRefPubMed 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(21):6630–8.CrossRefPubMed
49.
go back to reference Chen CT, Kim H, Liska D, Gao S, Christensen JG, Weiser MR. MET activation mediates resistance to lapatinib inhibition of HER2-amplified gastric cancer cells. Mol Cancer Ther. 2012;11(3):660–9.CrossRefPubMedPubMedCentral Chen CT, Kim H, Liska D, Gao S, Christensen JG, Weiser MR. MET activation mediates resistance to lapatinib inhibition of HER2-amplified gastric cancer cells. Mol Cancer Ther. 2012;11(3):660–9.CrossRefPubMedPubMedCentral
50.
go back to reference Mueller KL, Madden JM, Zoratti GL, Kuperwasser C, List K, Boerner JL. Fibroblast-secreted hepatocyte growth factor mediates epidermal growth factor receptor tyrosine kinase inhibitor resistance in triple-negative breast cancers through paracrine activation of Met. Breast Cancer Res: BCR. 2012;14(4):R104.CrossRefPubMedPubMedCentral Mueller KL, Madden JM, Zoratti GL, Kuperwasser C, List K, Boerner JL. Fibroblast-secreted hepatocyte growth factor mediates epidermal growth factor receptor tyrosine kinase inhibitor resistance in triple-negative breast cancers through paracrine activation of Met. Breast Cancer Res: BCR. 2012;14(4):R104.CrossRefPubMedPubMedCentral
51.
go back to reference Johansson AC, Ansell A, Jerhammar F, Lindh MB, Grenman R, Munck-Wikland E, et al. Cancer-associated fibroblasts induce matrix metalloproteinase-mediated cetuximab resistance in head and neck squamous cell carcinoma cells. Mol Cancer Res. 2012;10(9):1158–68.CrossRefPubMed Johansson AC, Ansell A, Jerhammar F, Lindh MB, Grenman R, Munck-Wikland E, et al. Cancer-associated fibroblasts induce matrix metalloproteinase-mediated cetuximab resistance in head and neck squamous cell carcinoma cells. Mol Cancer Res. 2012;10(9):1158–68.CrossRefPubMed
52.
go back to reference Kharaishvili G, Simkova D, Bouchalova K, Gachechiladze M, Narsia N, Bouchal J. The role of cancer-associated fibroblasts, solid stress and other microenvironmental factors in tumor progression and therapy resistance. Cancer Cell Int. 2014;14:41.CrossRefPubMedPubMedCentral Kharaishvili G, Simkova D, Bouchalova K, Gachechiladze M, Narsia N, Bouchal J. The role of cancer-associated fibroblasts, solid stress and other microenvironmental factors in tumor progression and therapy resistance. Cancer Cell Int. 2014;14:41.CrossRefPubMedPubMedCentral
53.
go back to reference Maurer J, Schopp M, Thurau K, Haier J, Kohler G, Hummel R. Immunohistochemical analysis on potential new molecular targets for esophageal cancer. Dis Esophagus. 2014;27(1):93–100.CrossRefPubMed Maurer J, Schopp M, Thurau K, Haier J, Kohler G, Hummel R. Immunohistochemical analysis on potential new molecular targets for esophageal cancer. Dis Esophagus. 2014;27(1):93–100.CrossRefPubMed
54.
go back to reference Kono K, Mimura K, Fujii H, Shabbir A, Yong W-P, Jimmy So A. Potential Therapeutic Significance of HER-Family in Esophageal Squamous Cell Carcinoma. Ann Thorac Cardiovasc Surg. 2012;18(6):506–13.CrossRefPubMed Kono K, Mimura K, Fujii H, Shabbir A, Yong W-P, Jimmy So A. Potential Therapeutic Significance of HER-Family in Esophageal Squamous Cell Carcinoma. Ann Thorac Cardiovasc Surg. 2012;18(6):506–13.CrossRefPubMed
Metadata
Title
The role of HGF/MET and FGF/FGFR in fibroblast-derived growth stimulation and lapatinib-resistance of esophageal squamous cell carcinoma
Authors
Shin Saito
Kazue Morishima
Takashi Ui
Hiroko Hoshino
Daisuke Matsubara
Shumpei Ishikawa
Hiroyuki Aburatani
Masashi Fukayama
Yoshinori Hosoya
Naohiro Sata
Alan K Lefor
Yoshikazu Yasuda
Toshiro Niki
Publication date
01-12-2015
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2015
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-015-1065-8

Other articles of this Issue 1/2015

BMC Cancer 1/2015 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine