Skip to main content
Top
Published in: Clinical & Experimental Metastasis 6/2008

Open Access 01-10-2008 | Original Article

Bypassing cellular EGF receptor dependence through epithelial-to-mesenchymal-like transitions

Authors: Sharon Barr, Stuart Thomson, Elizabeth Buck, Suzanne Russo, Filippo Petti, Izabela Sujka-Kwok, Alexandra Eyzaguirre, Maryland Rosenfeld-Franklin, Neil W. Gibson, Mark Miglarese, David Epstein, Kenneth K. Iwata, John D. Haley

Published in: Clinical & Experimental Metastasis | Issue 6/2008

Login to get access

Abstract

Over 90% of all cancers are carcinomas, malignancies derived from cells of epithelial origin. As carcinomas progress, these tumors may lose epithelial morphology and acquire mesenchymal characteristics which contribute to metastatic potential. An epithelial-to-mesenchymal transition (EMT) similar to the process critical for embryonic development is thought to be an important mechanism for promoting cancer invasion and metastasis. Epithelial-to-mesenchymal transitions have been induced in vitro by transient or unregulated activation of receptor tyrosine kinase signaling pathways, oncogene signaling and disruption of homotypic cell adhesion. These cellular models attempt to mimic the complexity of human carcinomas which respond to autocrine and paracrine signals from both the tumor and its microenvironment. Activation of the epidermal growth factor receptor (EGFR) has been implicated in the neoplastic transformation of solid tumors and overexpression of EGFR has been shown to correlate with poor survival. Notably, epithelial tumor cells have been shown to be significantly more sensitive to EGFR inhibitors than tumor cells which have undergone an EMT-like transition and acquired mesenchymal characteristics, including non-small cell lung (NSCLC), head and neck (HN), bladder, colorectal, pancreas and breast carcinomas. EGFR blockade has also been shown to inhibit cellular migration, suggesting a role for EGFR inhibitors in the control of metastasis. The interaction between EGFR and the multiple signaling nodes which regulate EMT suggest that the combination of an EGFR inhibitor and other molecular targeted agents may offer a novel approach to controlling metastasis.
Literature
1.
go back to reference Peinado H, Olmeda D, Cano A (2007) Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer 7(6):415–428PubMed Peinado H, Olmeda D, Cano A (2007) Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer 7(6):415–428PubMed
2.
go back to reference Maeda M, Johnson KR, Wheelock MJ (2005) Cadherin switching: essential for behavioral but not morphological changes during an epithelium-to-mesenchyme transition. J Cell Sci 118(Pt 5):873–887PubMed Maeda M, Johnson KR, Wheelock MJ (2005) Cadherin switching: essential for behavioral but not morphological changes during an epithelium-to-mesenchyme transition. J Cell Sci 118(Pt 5):873–887PubMed
3.
go back to reference Brabletz T, Jung A, Reu S et al (2001) Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc Natl Acad Sci USA 98(18):10356–10361PubMed Brabletz T, Jung A, Reu S et al (2001) Variable beta-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc Natl Acad Sci USA 98(18):10356–10361PubMed
4.
go back to reference Willipinski-Stapelfeldt B, Riethdorf S, Assmann V et al (2005) Changes in cytoskeletal protein composition indicative of an epithelial-mesenchymal transition in human micrometastatic and primary breast carcinoma cells. Clin Cancer Res 11(22):8006–8014PubMed Willipinski-Stapelfeldt B, Riethdorf S, Assmann V et al (2005) Changes in cytoskeletal protein composition indicative of an epithelial-mesenchymal transition in human micrometastatic and primary breast carcinoma cells. Clin Cancer Res 11(22):8006–8014PubMed
5.
go back to reference Buck E, Eyzaguirre A, Barr S et al (2007) Loss of homotypic cell adhesion by epithelial-mesenchymal transition or mutation limits sensitivity to epidermal growth factor receptor inhibition. Mol Cancer Ther 6(2):532–541PubMed Buck E, Eyzaguirre A, Barr S et al (2007) Loss of homotypic cell adhesion by epithelial-mesenchymal transition or mutation limits sensitivity to epidermal growth factor receptor inhibition. Mol Cancer Ther 6(2):532–541PubMed
6.
go back to reference Umbas R, Schalken JA, Aalders TW et al (1992) Expression of the cellular adhesion molecule E-cadherin is reduced or absent in high-grade prostate cancer. Cancer Res 52(18):5104–5109PubMed Umbas R, Schalken JA, Aalders TW et al (1992) Expression of the cellular adhesion molecule E-cadherin is reduced or absent in high-grade prostate cancer. Cancer Res 52(18):5104–5109PubMed
7.
go back to reference Yin T, Wang C, Liu T et al (2007) Expression of snail in pancreatic cancer promotes metastasis and chemoresistance. J Surg Res 141(2):196–203PubMed Yin T, Wang C, Liu T et al (2007) Expression of snail in pancreatic cancer promotes metastasis and chemoresistance. J Surg Res 141(2):196–203PubMed
8.
go back to reference Kajiyama H, Shibata K, Terauchi M et al (2007) Chemoresistance to paclitaxel induces epithelial-mesenchymal transition and enhances metastatic potential for epithelial ovarian carcinoma cells. Int J Oncol 31(2):277–283PubMed Kajiyama H, Shibata K, Terauchi M et al (2007) Chemoresistance to paclitaxel induces epithelial-mesenchymal transition and enhances metastatic potential for epithelial ovarian carcinoma cells. Int J Oncol 31(2):277–283PubMed
9.
go back to reference Cheng GZ, Chan J, Wang Q et al (2007) Twist transcriptionally up-regulates AKT2 in breast cancer cells leading to increased migration, invasion, and resistance to paclitaxel. Cancer Res 67(5):1979–1987PubMed Cheng GZ, Chan J, Wang Q et al (2007) Twist transcriptionally up-regulates AKT2 in breast cancer cells leading to increased migration, invasion, and resistance to paclitaxel. Cancer Res 67(5):1979–1987PubMed
10.
go back to reference Zhang Z, Xie D, Li X et al (2007) Significance of TWIST expression and its association with E-cadherin in bladder cancer. Hum Pathol 38(4):598–606PubMed Zhang Z, Xie D, Li X et al (2007) Significance of TWIST expression and its association with E-cadherin in bladder cancer. Hum Pathol 38(4):598–606PubMed
11.
go back to reference Hosono S, Kajiyama H, Terauchi M et al (2007) Expression of Twist increases the risk for recurrence and for poor survival in epithelial ovarian carcinoma patients. Br J Cancer 96(2):314–320PubMed Hosono S, Kajiyama H, Terauchi M et al (2007) Expression of Twist increases the risk for recurrence and for poor survival in epithelial ovarian carcinoma patients. Br J Cancer 96(2):314–320PubMed
12.
go back to reference Leroy P, Mostov KE (2007) Slug is required for cell survival during partial epithelial-mesenchymal transition of HGF-induced tubulogenesis. Mol Biol Cell 18(5):1943–1952PubMed Leroy P, Mostov KE (2007) Slug is required for cell survival during partial epithelial-mesenchymal transition of HGF-induced tubulogenesis. Mol Biol Cell 18(5):1943–1952PubMed
13.
go back to reference Cho HJ, Baek KE, Saika S et al (2007) Snail is required for transforming growth factor-beta-induced epithelial-mesenchymal transition by activating PI3 kinase/Akt signal pathway. Biochem Biophys Res Commun 353(2):337–343PubMed Cho HJ, Baek KE, Saika S et al (2007) Snail is required for transforming growth factor-beta-induced epithelial-mesenchymal transition by activating PI3 kinase/Akt signal pathway. Biochem Biophys Res Commun 353(2):337–343PubMed
14.
go back to reference Laux H, Tomer R, Mader MT et al (2004) Tumor-associated E-cadherin mutations do not induce Wnt target gene expression, but affect E-cadherin repressors. Lab Invest 84(10):1372–1386PubMed Laux H, Tomer R, Mader MT et al (2004) Tumor-associated E-cadherin mutations do not induce Wnt target gene expression, but affect E-cadherin repressors. Lab Invest 84(10):1372–1386PubMed
15.
go back to reference Lombaerts M, van Wezel T, Philippo K et al (2006) E-cadherin transcriptional downregulation by promoter methylation but not mutation is related to epithelial-to-mesenchymal transition in breast cancer cell lines. Br J Cancer 94(5):661–671PubMed Lombaerts M, van Wezel T, Philippo K et al (2006) E-cadherin transcriptional downregulation by promoter methylation but not mutation is related to epithelial-to-mesenchymal transition in breast cancer cell lines. Br J Cancer 94(5):661–671PubMed
16.
go back to reference Perl AK, Wilgenbus P, Dahl U et al (1998) A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature 392(6672):190–193PubMed Perl AK, Wilgenbus P, Dahl U et al (1998) A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature 392(6672):190–193PubMed
17.
go back to reference Vleminckx K, Vakaet L Jr, Mareel M et al (1991) Genetic manipulation of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell 66(1):107–119PubMed Vleminckx K, Vakaet L Jr, Mareel M et al (1991) Genetic manipulation of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell 66(1):107–119PubMed
18.
go back to reference Yang J, Mani SA, Donaher JL et al (2004) Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117(7):927–939PubMed Yang J, Mani SA, Donaher JL et al (2004) Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117(7):927–939PubMed
19.
go back to reference Cano A, Perez-Moreno MA, Rodrigo I et al (2000) The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol 2(2):76–83PubMed Cano A, Perez-Moreno MA, Rodrigo I et al (2000) The transcription factor snail controls epithelial-mesenchymal transitions by repressing E-cadherin expression. Nat Cell Biol 2(2):76–83PubMed
20.
go back to reference Batlle E, Sancho E, Franci C et al (2000) The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol 2(2):84–89PubMed Batlle E, Sancho E, Franci C et al (2000) The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol 2(2):84–89PubMed
21.
go back to reference Hennig G, Lowrick O, Birchmeier W et al (1996) Mechanisms identified in the transcriptional control of epithelial gene expression. J Biol Chem 271(1):595–602PubMed Hennig G, Lowrick O, Birchmeier W et al (1996) Mechanisms identified in the transcriptional control of epithelial gene expression. J Biol Chem 271(1):595–602PubMed
22.
go back to reference Hennig G, Behrens J, Truss M et al (1995) Progression of carcinoma cells is associated with alterations in chromatin structure and factor binding at the E-cadherin promoter in vivo. Oncogene 11(3):475–484PubMed Hennig G, Behrens J, Truss M et al (1995) Progression of carcinoma cells is associated with alterations in chromatin structure and factor binding at the E-cadherin promoter in vivo. Oncogene 11(3):475–484PubMed
23.
go back to reference Birchmeier W, Behrens J (1994) Cadherin expression in carcinomas: role in the formation of cell junctions and the prevention of invasiveness. Biochim Biophys Acta 1198(1):11–26PubMed Birchmeier W, Behrens J (1994) Cadherin expression in carcinomas: role in the formation of cell junctions and the prevention of invasiveness. Biochim Biophys Acta 1198(1):11–26PubMed
24.
go back to reference Nieto MA (2002) The snail superfamily of zinc-finger transcription factors. Nat Rev Mol Cell Biol 3(3):155–166PubMed Nieto MA (2002) The snail superfamily of zinc-finger transcription factors. Nat Rev Mol Cell Biol 3(3):155–166PubMed
25.
go back to reference Aigner K, Dampier B, Descovich L et al (2007) The transcription factor ZEB1 (deltaEF1) promotes tumour cell dedifferentiation by repressing master regulators of epithelial polarity. Oncogene 26(49):6979–6988PubMed Aigner K, Dampier B, Descovich L et al (2007) The transcription factor ZEB1 (deltaEF1) promotes tumour cell dedifferentiation by repressing master regulators of epithelial polarity. Oncogene 26(49):6979–6988PubMed
26.
go back to reference Eger A, Aigner K, Sonderegger S et al (2005) DeltaEF1 is a transcriptional repressor of E-cadherin and regulates epithelial plasticity in breast cancer cells. Oncogene 24(14):2375–2385PubMed Eger A, Aigner K, Sonderegger S et al (2005) DeltaEF1 is a transcriptional repressor of E-cadherin and regulates epithelial plasticity in breast cancer cells. Oncogene 24(14):2375–2385PubMed
27.
go back to reference Perez-Moreno MA, Locascio A, Rodrigo I et al (2001) A new role for E12/E47 in the repression of E-cadherin expression and epithelial-mesenchymal transitions. J Biol Chem 276(29):27424–27431PubMed Perez-Moreno MA, Locascio A, Rodrigo I et al (2001) A new role for E12/E47 in the repression of E-cadherin expression and epithelial-mesenchymal transitions. J Biol Chem 276(29):27424–27431PubMed
28.
go back to reference Guaita S, Puig I, Franci C et al (2002) Snail induction of epithelial to mesenchymal transition in tumor cells is accompanied by MUC1 repression and ZEB1 expression. J Biol Chem 277(42):39209–39216PubMed Guaita S, Puig I, Franci C et al (2002) Snail induction of epithelial to mesenchymal transition in tumor cells is accompanied by MUC1 repression and ZEB1 expression. J Biol Chem 277(42):39209–39216PubMed
29.
go back to reference Tamagnone L, Comoglio PM (1997) Control of invasive growth by hepatocyte growth factor (HGF) and related scatter factors. Cytokine Growth Factor Rev 8(2):129–142PubMed Tamagnone L, Comoglio PM (1997) Control of invasive growth by hepatocyte growth factor (HGF) and related scatter factors. Cytokine Growth Factor Rev 8(2):129–142PubMed
30.
go back to reference Montesano R, Matsumoto K, Nakamura T et al (1991) Identification of a fibroblast-derived epithelial morphogen as hepatocyte growth factor. Cell 67(5):901–908PubMed Montesano R, Matsumoto K, Nakamura T et al (1991) Identification of a fibroblast-derived epithelial morphogen as hepatocyte growth factor. Cell 67(5):901–908PubMed
31.
go back to reference Thiery JP (2002) Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2(6):442–454PubMed Thiery JP (2002) Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer 2(6):442–454PubMed
32.
go back to reference Thiery JP (2003) Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol 15(6):740–746PubMed Thiery JP (2003) Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol 15(6):740–746PubMed
33.
go back to reference Vincent-Salomon A, Thiery JP (2003) Host microenvironment in breast cancer development: epithelial-mesenchymal transition in breast cancer development. Breast Cancer Res 5(2):101–106PubMed Vincent-Salomon A, Thiery JP (2003) Host microenvironment in breast cancer development: epithelial-mesenchymal transition in breast cancer development. Breast Cancer Res 5(2):101–106PubMed
34.
go back to reference Onoue T, Uchida D, Begum NM et al (2006) Epithelial-mesenchymal transition induced by the stromal cell-derived factor-1/CXCR4 system in oral squamous cell carcinoma cells. Int J Oncol 29(5):1133–1138PubMed Onoue T, Uchida D, Begum NM et al (2006) Epithelial-mesenchymal transition induced by the stromal cell-derived factor-1/CXCR4 system in oral squamous cell carcinoma cells. Int J Oncol 29(5):1133–1138PubMed
35.
go back to reference Waerner T, Alacakaptan M, Tamir I et al (2006) ILEI: a cytokine essential for EMT, tumor formation, and late events in metastasis in epithelial cells. Cancer Cell 10(3):227–239PubMed Waerner T, Alacakaptan M, Tamir I et al (2006) ILEI: a cytokine essential for EMT, tumor formation, and late events in metastasis in epithelial cells. Cancer Cell 10(3):227–239PubMed
36.
go back to reference Petersen OW, Nielsen HL, Gudjonsson T et al (2003) Epithelial to mesenchymal transition in human breast cancer can provide a nonmalignant stroma. Am J Pathol 162(2):391–402PubMed Petersen OW, Nielsen HL, Gudjonsson T et al (2003) Epithelial to mesenchymal transition in human breast cancer can provide a nonmalignant stroma. Am J Pathol 162(2):391–402PubMed
37.
go back to reference Derynck R, Akhurst RJ, Balmain A (2001) TGF-beta signaling in tumor suppression and cancer progression. Nat Genet 29(2):117–129PubMed Derynck R, Akhurst RJ, Balmain A (2001) TGF-beta signaling in tumor suppression and cancer progression. Nat Genet 29(2):117–129PubMed
38.
go back to reference Moses HL, Yang EY, Pietenpol JA (1990) TGF-beta stimulation and inhibition of cell proliferation: new mechanistic insights. Cell 63(2):245–247PubMed Moses HL, Yang EY, Pietenpol JA (1990) TGF-beta stimulation and inhibition of cell proliferation: new mechanistic insights. Cell 63(2):245–247PubMed
39.
go back to reference Valdes F, Alvarez AM, Locascio A et al (2002) The epithelial mesenchymal transition confers resistance to the apoptotic effects of transforming growth factor Beta in fetal rat hepatocytes. Mol Cancer Res 1(1):68–78PubMed Valdes F, Alvarez AM, Locascio A et al (2002) The epithelial mesenchymal transition confers resistance to the apoptotic effects of transforming growth factor Beta in fetal rat hepatocytes. Mol Cancer Res 1(1):68–78PubMed
40.
go back to reference Sanchez A, Alvarez AM, Lopez Pedrosa JM et al (1999) Apoptotic response to TGF-beta in fetal hepatocytes depends upon their state of differentiation. Exp Cell Res 252(2):281–291PubMed Sanchez A, Alvarez AM, Lopez Pedrosa JM et al (1999) Apoptotic response to TGF-beta in fetal hepatocytes depends upon their state of differentiation. Exp Cell Res 252(2):281–291PubMed
41.
go back to reference Robson EJ, Khaled WT, Abell K et al (2006) Epithelial-to-mesenchymal transition confers resistance to apoptosis in three murine mammary epithelial cell lines. Differentiation; Res Biol Divers 74(5):254–264 Robson EJ, Khaled WT, Abell K et al (2006) Epithelial-to-mesenchymal transition confers resistance to apoptosis in three murine mammary epithelial cell lines. Differentiation; Res Biol Divers 74(5):254–264
42.
go back to reference Gal A, Sjoblom T, Fedorova L et al (2007) Sustained TGFbeta exposure suppresses Smad and non-Smad signalling in mammary epithelial cells, leading to EMT and inhibition of growth arrest and apoptosis. Oncogene. doi:10.1038/sj.onc.1210741 Gal A, Sjoblom T, Fedorova L et al (2007) Sustained TGFbeta exposure suppresses Smad and non-Smad signalling in mammary epithelial cells, leading to EMT and inhibition of growth arrest and apoptosis. Oncogene. doi:10.​1038/​sj.​onc.​1210741
43.
go back to reference Vega S, Morales AV, Ocana OH et al (2004) Snail blocks the cell cycle and confers resistance to cell death. Genes Dev 18(10):1131–1143PubMed Vega S, Morales AV, Ocana OH et al (2004) Snail blocks the cell cycle and confers resistance to cell death. Genes Dev 18(10):1131–1143PubMed
44.
go back to reference Inoue A, Seidel MG, Wu W et al (2002) Slug, a highly conserved zinc finger transcriptional repressor, protects hematopoietic progenitor cells from radiation-induced apoptosis in vivo. Cancer Cell 2(4):279–288PubMed Inoue A, Seidel MG, Wu W et al (2002) Slug, a highly conserved zinc finger transcriptional repressor, protects hematopoietic progenitor cells from radiation-induced apoptosis in vivo. Cancer Cell 2(4):279–288PubMed
45.
go back to reference Wu WS, Heinrichs S, Xu D et al (2005) Slug antagonizes p53-mediated apoptosis of hematopoietic progenitors by repressing puma. Cell 123(4):641–653PubMed Wu WS, Heinrichs S, Xu D et al (2005) Slug antagonizes p53-mediated apoptosis of hematopoietic progenitors by repressing puma. Cell 123(4):641–653PubMed
46.
go back to reference Fujita Y, Krause G, Scheffner M et al (2002) Hakai, a c-Cbl-like protein, ubiquitinates and induces endocytosis of the E-cadherin complex. Nat Cell Biol 4(3):222–231PubMed Fujita Y, Krause G, Scheffner M et al (2002) Hakai, a c-Cbl-like protein, ubiquitinates and induces endocytosis of the E-cadherin complex. Nat Cell Biol 4(3):222–231PubMed
47.
go back to reference Kim HJ, Litzenburger BC, Cui X et al (2007) Constitutively active type I insulin-like growth factor receptor causes transformation and xenograft growth of immortalized mammary epithelial cells and is accompanied by an epithelial-to-mesenchymal transition mediated by NF-kappaB and snail. Mol Cell Biol 27(8):3165–3175PubMed Kim HJ, Litzenburger BC, Cui X et al (2007) Constitutively active type I insulin-like growth factor receptor causes transformation and xenograft growth of immortalized mammary epithelial cells and is accompanied by an epithelial-to-mesenchymal transition mediated by NF-kappaB and snail. Mol Cell Biol 27(8):3165–3175PubMed
48.
go back to reference Lopez T, Hanahan D (2002) Elevated levels of IGF-1 receptor convey invasive and metastatic capability in a mouse model of pancreatic islet tumorigenesis. Cancer Cell 1(4):339–353PubMed Lopez T, Hanahan D (2002) Elevated levels of IGF-1 receptor convey invasive and metastatic capability in a mouse model of pancreatic islet tumorigenesis. Cancer Cell 1(4):339–353PubMed
49.
go back to reference Doerr ME, Jones JI (1996) The roles of integrins and extracellular matrix proteins in the insulin-like growth factor I-stimulated chemotaxis of human breast cancer cells. J Biol Chem 271(5):2443–2447PubMed Doerr ME, Jones JI (1996) The roles of integrins and extracellular matrix proteins in the insulin-like growth factor I-stimulated chemotaxis of human breast cancer cells. J Biol Chem 271(5):2443–2447PubMed
50.
go back to reference Stracke ML, Engel JD, Wilson LW et al (1989) The type I insulin-like growth factor receptor is a motility receptor in human melanoma cells. J Biol Chem 264(36):21544–21549PubMed Stracke ML, Engel JD, Wilson LW et al (1989) The type I insulin-like growth factor receptor is a motility receptor in human melanoma cells. J Biol Chem 264(36):21544–21549PubMed
51.
go back to reference Bornfeldt KE, Raines EW, Nakano T et al (1994) Insulin-like growth factor-I and platelet-derived growth factor-BB induce directed migration of human arterial smooth muscle cells via signaling pathways that are distinct from those of proliferation. J Clin Invest 93(3):1266–1274PubMed Bornfeldt KE, Raines EW, Nakano T et al (1994) Insulin-like growth factor-I and platelet-derived growth factor-BB induce directed migration of human arterial smooth muscle cells via signaling pathways that are distinct from those of proliferation. J Clin Invest 93(3):1266–1274PubMed
52.
go back to reference Leventhal PS, Shelden EA, Kim B et al (1997) Tyrosine phosphorylation of paxillin and focal adhesion kinase during insulin-like growth factor-I-stimulated lamellipodial advance. J Biol Chem 272(8):5214–5218PubMed Leventhal PS, Shelden EA, Kim B et al (1997) Tyrosine phosphorylation of paxillin and focal adhesion kinase during insulin-like growth factor-I-stimulated lamellipodial advance. J Biol Chem 272(8):5214–5218PubMed
53.
go back to reference Playford MP, Bicknell D, Bodmer WF et al (2000) Insulin-like growth factor 1 regulates the location, stability, and transcriptional activity of beta-catenin. Proc Natl Acad Sci USA 97(22):12103–12108PubMed Playford MP, Bicknell D, Bodmer WF et al (2000) Insulin-like growth factor 1 regulates the location, stability, and transcriptional activity of beta-catenin. Proc Natl Acad Sci USA 97(22):12103–12108PubMed
54.
go back to reference Zhang D, Brodt P (2003) Type 1 insulin-like growth factor regulates MT1-MMP synthesis and tumor invasion via PI 3-kinase/Akt signaling. Oncogene 22(7):974–982PubMed Zhang D, Brodt P (2003) Type 1 insulin-like growth factor regulates MT1-MMP synthesis and tumor invasion via PI 3-kinase/Akt signaling. Oncogene 22(7):974–982PubMed
55.
go back to reference Shen MR, Hsu YM, Hsu KF et al (2006) Insulin-like growth factor 1 is a potent stimulator of cervical cancer cell invasiveness and proliferation that is modulated by alphavbeta3 integrin signaling. Carcinogenesis 27(5):962–971PubMed Shen MR, Hsu YM, Hsu KF et al (2006) Insulin-like growth factor 1 is a potent stimulator of cervical cancer cell invasiveness and proliferation that is modulated by alphavbeta3 integrin signaling. Carcinogenesis 27(5):962–971PubMed
56.
go back to reference Bohula EA, Playford MP, Macaulay VM (2003) Targeting the type 1 insulin-like growth factor receptor as anti-cancer treatment. Anticancer Drugs 14(9):669–682PubMed Bohula EA, Playford MP, Macaulay VM (2003) Targeting the type 1 insulin-like growth factor receptor as anti-cancer treatment. Anticancer Drugs 14(9):669–682PubMed
57.
go back to reference Price JT, Wilson HM, Haites NE (1996) Epidermal growth factor (EGF) increases the in vitro invasion, motility and adhesion interactions of the primary renal carcinoma cell line, A704. Eur J Cancer 32A(11):1977–1982PubMed Price JT, Wilson HM, Haites NE (1996) Epidermal growth factor (EGF) increases the in vitro invasion, motility and adhesion interactions of the primary renal carcinoma cell line, A704. Eur J Cancer 32A(11):1977–1982PubMed
58.
go back to reference Shibata T, Kawano T, Nagayasu H et al (1996) Enhancing effects of epidermal growth factor on human squamous cell carcinoma motility and matrix degradation but not growth. Tumour Biol 17(3):168–175PubMedCrossRef Shibata T, Kawano T, Nagayasu H et al (1996) Enhancing effects of epidermal growth factor on human squamous cell carcinoma motility and matrix degradation but not growth. Tumour Biol 17(3):168–175PubMedCrossRef
59.
go back to reference Lu Z, Jiang G, Blume-Jensen P et al (2001) Epidermal growth factor-induced tumor cell invasion and metastasis initiated by dephosphorylation and downregulation of focal adhesion kinase. Mol Cell Biol 21(12):4016–4031PubMed Lu Z, Jiang G, Blume-Jensen P et al (2001) Epidermal growth factor-induced tumor cell invasion and metastasis initiated by dephosphorylation and downregulation of focal adhesion kinase. Mol Cell Biol 21(12):4016–4031PubMed
60.
go back to reference Salamanca CM, Maines-Bandiera SL, Leung PC et al (2004) Effects of epidermal growth factor/hydrocortisone on the growth and differentiation of human ovarian surface epithelium. J Soc Gynecol Invest 11(4):241–251 Salamanca CM, Maines-Bandiera SL, Leung PC et al (2004) Effects of epidermal growth factor/hydrocortisone on the growth and differentiation of human ovarian surface epithelium. J Soc Gynecol Invest 11(4):241–251
61.
go back to reference Lu Z, Ghosh S, Wang Z et al (2003) Downregulation of caveolin-1 function by EGF leads to the loss of E-cadherin, increased transcriptional activity of beta-catenin, and enhanced tumor cell invasion. Cancer Cell 4(6):499–515PubMed Lu Z, Ghosh S, Wang Z et al (2003) Downregulation of caveolin-1 function by EGF leads to the loss of E-cadherin, increased transcriptional activity of beta-catenin, and enhanced tumor cell invasion. Cancer Cell 4(6):499–515PubMed
62.
go back to reference Cole GW Jr, Alleva AM, Reddy RM et al (2005) The selective epidermal growth factor receptor tyrosine kinase inhibitor PD153035 suppresses expression of prometastasis phenotypes in malignant pleural mesothelioma cells in vitro. J Thorac Cardiovasc Surg 129(5):1010–1017PubMed Cole GW Jr, Alleva AM, Reddy RM et al (2005) The selective epidermal growth factor receptor tyrosine kinase inhibitor PD153035 suppresses expression of prometastasis phenotypes in malignant pleural mesothelioma cells in vitro. J Thorac Cardiovasc Surg 129(5):1010–1017PubMed
63.
go back to reference Lal A, Glazer CA, Martinson HM et al (2002) Mutant epidermal growth factor receptor up-regulates molecular effectors of tumor invasion. Cancer Res 62(12):3335–3339PubMed Lal A, Glazer CA, Martinson HM et al (2002) Mutant epidermal growth factor receptor up-regulates molecular effectors of tumor invasion. Cancer Res 62(12):3335–3339PubMed
64.
go back to reference Seton-Rogers SE, Lu Y, Hines LM et al (2004) Cooperation of the ErbB2 receptor and transforming growth factor beta in induction of migration and invasion in mammary epithelial cells. Proc Natl Acad Sci USA 101(5):1257–1262PubMed Seton-Rogers SE, Lu Y, Hines LM et al (2004) Cooperation of the ErbB2 receptor and transforming growth factor beta in induction of migration and invasion in mammary epithelial cells. Proc Natl Acad Sci USA 101(5):1257–1262PubMed
65.
go back to reference Mateus AR, Seruca R, Machado JC et al (2007) EGFR regulates RhoA-GTP dependent cell motility in E-cadherin mutant cells. Hum Mol Genet 16(13):1639–1647PubMed Mateus AR, Seruca R, Machado JC et al (2007) EGFR regulates RhoA-GTP dependent cell motility in E-cadherin mutant cells. Hum Mol Genet 16(13):1639–1647PubMed
66.
go back to reference Lorch JH, Klessner J, Park JK et al (2004) Epidermal growth factor receptor inhibition promotes desmosome assembly and strengthens intercellular adhesion in squamous cell carcinoma cells. J Biol Chem 279(35):37191–37200PubMed Lorch JH, Klessner J, Park JK et al (2004) Epidermal growth factor receptor inhibition promotes desmosome assembly and strengthens intercellular adhesion in squamous cell carcinoma cells. J Biol Chem 279(35):37191–37200PubMed
67.
go back to reference Wells A (1999) EGF receptor. Int J Biochem Cell Biol 31(6):637–643PubMed Wells A (1999) EGF receptor. Int J Biochem Cell Biol 31(6):637–643PubMed
68.
go back to reference Grandis JR, Sok JC (2004) Signaling through the epidermal growth factor receptor during the development of malignancy. Pharmacol Ther 102(1):37–46PubMed Grandis JR, Sok JC (2004) Signaling through the epidermal growth factor receptor during the development of malignancy. Pharmacol Ther 102(1):37–46PubMed
69.
go back to reference Shepherd FA, Rodrigues Pereira J, Ciuleanu T et al (2005) Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med 353(2):123–132PubMed Shepherd FA, Rodrigues Pereira J, Ciuleanu T et al (2005) Erlotinib in previously treated non-small-cell lung cancer. N Engl J Med 353(2):123–132PubMed
70.
go back to reference Thomson S, Buck E, Petti F et al (2005) Epithelial to mesenchymal transition is a determinant of sensitivity of non-small-cell lung carcinoma cell lines and xenografts to epidermal growth factor receptor inhibition. Cancer Res 65(20):9455–9462PubMed Thomson S, Buck E, Petti F et al (2005) Epithelial to mesenchymal transition is a determinant of sensitivity of non-small-cell lung carcinoma cell lines and xenografts to epidermal growth factor receptor inhibition. Cancer Res 65(20):9455–9462PubMed
71.
go back to reference Yauch RL, Januario T, Eberhard DA et al (2005) Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients. Clin Cancer Res 11(24 Pt 1):8686–8698PubMed Yauch RL, Januario T, Eberhard DA et al (2005) Epithelial versus mesenchymal phenotype determines in vitro sensitivity and predicts clinical activity of erlotinib in lung cancer patients. Clin Cancer Res 11(24 Pt 1):8686–8698PubMed
72.
go back to reference Witta SE, Gemmill RM, Hirsch FR et al (2006) Restoring E-cadherin expression increases sensitivity to epidermal growth factor receptor inhibitors in lung cancer cell lines. Cancer Res 66(2):944–950PubMed Witta SE, Gemmill RM, Hirsch FR et al (2006) Restoring E-cadherin expression increases sensitivity to epidermal growth factor receptor inhibitors in lung cancer cell lines. Cancer Res 66(2):944–950PubMed
73.
go back to reference Frederick BA, Helfrich BA, Coldren CD et al (2007) Epithelial to mesenchymal transition predicts gefitinib resistance in cell lines of head and neck squamous cell carcinoma and non-small cell lung carcinoma. Mol Cancer Ther 6(6):1683–1691PubMed Frederick BA, Helfrich BA, Coldren CD et al (2007) Epithelial to mesenchymal transition predicts gefitinib resistance in cell lines of head and neck squamous cell carcinoma and non-small cell lung carcinoma. Mol Cancer Ther 6(6):1683–1691PubMed
74.
go back to reference Shrader M, Pino MS, Brown G et al (2007) Molecular correlates of gefitinib responsiveness in human bladder cancer cells. Mol Cancer Ther 6(1):277–285PubMed Shrader M, Pino MS, Brown G et al (2007) Molecular correlates of gefitinib responsiveness in human bladder cancer cells. Mol Cancer Ther 6(1):277–285PubMed
75.
go back to reference Buck E, Eyzaguirre A, Franklin M et al (2007) The EGFR inhibitor erlotinib sensitizes tumor cells to IGF-1 receptor inhibition by promoting Akt signaling through the IGF-1R-IRS1-Akt axis. Proceedings of American Association for Cancer Research Annual Meeting, Los Angeles, CA Buck E, Eyzaguirre A, Franklin M et al (2007) The EGFR inhibitor erlotinib sensitizes tumor cells to IGF-1 receptor inhibition by promoting Akt signaling through the IGF-1R-IRS1-Akt axis. Proceedings of American Association for Cancer Research Annual Meeting, Los Angeles, CA
76.
go back to reference Herbst RS, Prager D, Hermann R et al (2005) TRIBUTE: a phase III trial of erlotinib hydrochloride (OSI-774) combined with carboplatin and paclitaxel chemotherapy in advanced non-small-cell lung cancer. J Clin Oncol 23(25):5892–5899PubMed Herbst RS, Prager D, Hermann R et al (2005) TRIBUTE: a phase III trial of erlotinib hydrochloride (OSI-774) combined with carboplatin and paclitaxel chemotherapy in advanced non-small-cell lung cancer. J Clin Oncol 23(25):5892–5899PubMed
77.
go back to reference Ono M, Hirata A, Kometani T et al (2004) Sensitivity to gefitinib (Iressa, ZD1839) in non-small cell lung cancer cell lines correlates with dependence on the epidermal growth factor (EGF) receptor/extracellular signal-regulated kinase 1/2 and EGF receptor/Akt pathway for proliferation. Mol Cancer Ther 3(4):465–472PubMed Ono M, Hirata A, Kometani T et al (2004) Sensitivity to gefitinib (Iressa, ZD1839) in non-small cell lung cancer cell lines correlates with dependence on the epidermal growth factor (EGF) receptor/extracellular signal-regulated kinase 1/2 and EGF receptor/Akt pathway for proliferation. Mol Cancer Ther 3(4):465–472PubMed
78.
go back to reference Arteaga CL (2002) Epidermal growth factor receptor dependence in human tumors: more than just expression? Oncologist 7(Suppl 4):31–39PubMed Arteaga CL (2002) Epidermal growth factor receptor dependence in human tumors: more than just expression? Oncologist 7(Suppl 4):31–39PubMed
79.
go back to reference Moulder SL, Yakes FM, Muthuswamy SK et al (2001) Epidermal growth factor receptor (HER1) tyrosine kinase inhibitor ZD1839 (Iressa) inhibits HER2/neu (erbB2)-overexpressing breast cancer cells in vitro and in vivo. Cancer Res 61(24):8887–8895PubMed Moulder SL, Yakes FM, Muthuswamy SK et al (2001) Epidermal growth factor receptor (HER1) tyrosine kinase inhibitor ZD1839 (Iressa) inhibits HER2/neu (erbB2)-overexpressing breast cancer cells in vitro and in vivo. Cancer Res 61(24):8887–8895PubMed
80.
go back to reference Buck E, Eyzaguirre A, Haley JD et al (2006) Inactivation of Akt by the epidermal growth factor receptor inhibitor erlotinib is mediated by HER-3 in pancreatic and colorectal tumor cell lines and contributes to erlotinib sensitivity. Mol Cancer Ther 5(8):2051–2059PubMed Buck E, Eyzaguirre A, Haley JD et al (2006) Inactivation of Akt by the epidermal growth factor receptor inhibitor erlotinib is mediated by HER-3 in pancreatic and colorectal tumor cell lines and contributes to erlotinib sensitivity. Mol Cancer Ther 5(8):2051–2059PubMed
81.
go back to reference Del Castillo G, Murillo MM, Alvarez-Barrientos A et al (2006) Autocrine production of TGF-beta confers resistance to apoptosis after an epithelial-mesenchymal transition process in hepatocytes: role of EGF receptor ligands. Exp Cell Res 312(15):2860–2871PubMed Del Castillo G, Murillo MM, Alvarez-Barrientos A et al (2006) Autocrine production of TGF-beta confers resistance to apoptosis after an epithelial-mesenchymal transition process in hepatocytes: role of EGF receptor ligands. Exp Cell Res 312(15):2860–2871PubMed
82.
go back to reference Murillo MM, del Castillo G, Sanchez A et al (2005) Involvement of EGF receptor and c-Src in the survival signals induced by TGF-beta1 in hepatocytes. Oncogene 24(28):4580–4587PubMed Murillo MM, del Castillo G, Sanchez A et al (2005) Involvement of EGF receptor and c-Src in the survival signals induced by TGF-beta1 in hepatocytes. Oncogene 24(28):4580–4587PubMed
83.
go back to reference Engelman JA, Janne PA, Mermel C et al (2005) ErbB-3 mediates phosphoinositide 3-kinase activity in gefitinib-sensitive non-small cell lung cancer cell lines. Proc Natl Acad Sci USA 102(10):3788–3793PubMed Engelman JA, Janne PA, Mermel C et al (2005) ErbB-3 mediates phosphoinositide 3-kinase activity in gefitinib-sensitive non-small cell lung cancer cell lines. Proc Natl Acad Sci USA 102(10):3788–3793PubMed
84.
go back to reference Desbois-Mouthon C, Cacheux W, Blivet-Van Eggelpoel MJ et al (2006) Impact of IGF-1R/EGFR cross-talks on hepatoma cell sensitivity to gefitinib. Int J Cancer 119(11):2557–2566PubMed Desbois-Mouthon C, Cacheux W, Blivet-Van Eggelpoel MJ et al (2006) Impact of IGF-1R/EGFR cross-talks on hepatoma cell sensitivity to gefitinib. Int J Cancer 119(11):2557–2566PubMed
85.
go back to reference Barr SM, Buck EA, Thomson S et al (2007) The combination of small molecule inhibitors of EGFR and IGF-1R is synergistic in HNSCC and ovarian cancer cell lines. Proceedings of American Association of Cancer Research Annual Meeting. Los Angeles, CA Barr SM, Buck EA, Thomson S et al (2007) The combination of small molecule inhibitors of EGFR and IGF-1R is synergistic in HNSCC and ovarian cancer cell lines. Proceedings of American Association of Cancer Research Annual Meeting. Los Angeles, CA
86.
go back to reference Chakravarti A, Loeffler JS, Dyson NJ (2002) Insulin-like growth factor receptor I mediates resistance to anti-epidermal growth factor receptor therapy in primary human glioblastoma cells through continued activation of phosphoinositide 3-kinase signaling. Cancer Res 62(1):200–207PubMed Chakravarti A, Loeffler JS, Dyson NJ (2002) Insulin-like growth factor receptor I mediates resistance to anti-epidermal growth factor receptor therapy in primary human glioblastoma cells through continued activation of phosphoinositide 3-kinase signaling. Cancer Res 62(1):200–207PubMed
87.
go back to reference Jones HE, Goddard L, Gee JM et al (2004) Insulin-like growth factor-I receptor signalling and acquired resistance to gefitinib (ZD1839; Iressa) in human breast and prostate cancer cells. Endocr Relat Cancer 11(4):793–814PubMed Jones HE, Goddard L, Gee JM et al (2004) Insulin-like growth factor-I receptor signalling and acquired resistance to gefitinib (ZD1839; Iressa) in human breast and prostate cancer cells. Endocr Relat Cancer 11(4):793–814PubMed
88.
go back to reference Sutter AP, Hopfner M, Huether A et al (2006) Targeting the epidermal growth factor receptor by erlotinib (Tarceva) for the treatment of esophageal cancer. Int J Cancer 118(7):1814–1822PubMed Sutter AP, Hopfner M, Huether A et al (2006) Targeting the epidermal growth factor receptor by erlotinib (Tarceva) for the treatment of esophageal cancer. Int J Cancer 118(7):1814–1822PubMed
89.
go back to reference Slomiany MG, Black LA, Kibbey MM et al (2007) Insulin-like growth factor-1 receptor and ligand targeting in head and neck squamous cell carcinoma. Cancer Lett 248(2):269–279PubMed Slomiany MG, Black LA, Kibbey MM et al (2007) Insulin-like growth factor-1 receptor and ligand targeting in head and neck squamous cell carcinoma. Cancer Lett 248(2):269–279PubMed
90.
go back to reference Desbois-Mouthon C, Danan C, Amselem S et al (1996) Severe resistance to insulin and insulin-like growth factor-I in cells from a patient with leprechaunism as a result of two mutations in the tyrosine kinase domain of the insulin receptor. Metabolism 45(12):1493–1500PubMed Desbois-Mouthon C, Danan C, Amselem S et al (1996) Severe resistance to insulin and insulin-like growth factor-I in cells from a patient with leprechaunism as a result of two mutations in the tyrosine kinase domain of the insulin receptor. Metabolism 45(12):1493–1500PubMed
91.
go back to reference Morgillo F, Woo JK, Kim ES et al (2006) Heterodimerization of insulin-like growth factor receptor/epidermal growth factor receptor and induction of survivin expression counteract the antitumor action of erlotinib. Cancer Res 66(20):10100–10111PubMed Morgillo F, Woo JK, Kim ES et al (2006) Heterodimerization of insulin-like growth factor receptor/epidermal growth factor receptor and induction of survivin expression counteract the antitumor action of erlotinib. Cancer Res 66(20):10100–10111PubMed
92.
go back to reference Dunn SE, Ehrlich M, Sharp NJ et al (1998) A dominant negative mutant of the insulin-like growth factor-I receptor inhibits the adhesion, invasion, and metastasis of breast cancer. Cancer Res 58(15):3353–3361PubMed Dunn SE, Ehrlich M, Sharp NJ et al (1998) A dominant negative mutant of the insulin-like growth factor-I receptor inhibits the adhesion, invasion, and metastasis of breast cancer. Cancer Res 58(15):3353–3361PubMed
93.
go back to reference Samani AA, Fallavollita L, Jaalouk DE et al (2001) Inhibition of carcinoma cell growth and metastasis by a vesicular stomatitis virus G-pseudotyped retrovector expressing type I insulin-like growth factor receptor antisense. Hum Gene Ther 12(16):1969–1977PubMed Samani AA, Fallavollita L, Jaalouk DE et al (2001) Inhibition of carcinoma cell growth and metastasis by a vesicular stomatitis virus G-pseudotyped retrovector expressing type I insulin-like growth factor receptor antisense. Hum Gene Ther 12(16):1969–1977PubMed
94.
go back to reference Choi YJ, Nam SJ, Son MJ et al (2006) Erlotinib prevents pulmonary metastasis in curatively resected breast carcinoma using a mouse model. Oncol Rep 16(1):119–122PubMed Choi YJ, Nam SJ, Son MJ et al (2006) Erlotinib prevents pulmonary metastasis in curatively resected breast carcinoma using a mouse model. Oncol Rep 16(1):119–122PubMed
95.
go back to reference Angelucci A, Gravina GL, Rucci N et al (2006) Suppression of EGF-R signaling reduces the incidence of prostate cancer metastasis in nude mice. Endocr Relat Cancer 13(1):197–210PubMed Angelucci A, Gravina GL, Rucci N et al (2006) Suppression of EGF-R signaling reduces the incidence of prostate cancer metastasis in nude mice. Endocr Relat Cancer 13(1):197–210PubMed
96.
go back to reference Shintani S, Li C, Mihara M et al (2003) Gefitinib (‘Iressa’), an epidermal growth factor receptor tyrosine kinase inhibitor, mediates the inhibition of lymph node metastasis in oral cancer cells. Cancer Lett 201(2):149–155PubMed Shintani S, Li C, Mihara M et al (2003) Gefitinib (‘Iressa’), an epidermal growth factor receptor tyrosine kinase inhibitor, mediates the inhibition of lymph node metastasis in oral cancer cells. Cancer Lett 201(2):149–155PubMed
97.
go back to reference Thomson S, Petti F, Haley J (2007) Multiple mechanisms of insensitivity to Erlotinib in mesenchymal-like NSCLC cell lines. Proceedings of the American Association for Cancer Research Anual Meeting, Los Angeles, CA; 48 Thomson S, Petti F, Haley J (2007) Multiple mechanisms of insensitivity to Erlotinib in mesenchymal-like NSCLC cell lines. Proceedings of the American Association for Cancer Research Anual Meeting, Los Angeles, CA; 48
98.
go back to reference Heldin CH, Westermark B, Wasteson A (1981) Specific receptors for platelet-derived growth factor on cells derived from connective tissue and glia. Proc Natl Acad Sci USA 78(6):3664–3668PubMed Heldin CH, Westermark B, Wasteson A (1981) Specific receptors for platelet-derived growth factor on cells derived from connective tissue and glia. Proc Natl Acad Sci USA 78(6):3664–3668PubMed
99.
go back to reference Westermark B, Heldin CH, Nister M (1995) Platelet-derived growth factor in human glioma. Glia 15(3):257–263PubMed Westermark B, Heldin CH, Nister M (1995) Platelet-derived growth factor in human glioma. Glia 15(3):257–263PubMed
100.
go back to reference Dai C, Celestino JC, Okada Y et al (2001) PDGF autocrine stimulation dedifferentiates cultured astrocytes and induces oligodendrogliomas and oligoastrocytomas from neural progenitors and astrocytes in vivo. Genes Dev 15(15):1913–1925PubMed Dai C, Celestino JC, Okada Y et al (2001) PDGF autocrine stimulation dedifferentiates cultured astrocytes and induces oligodendrogliomas and oligoastrocytomas from neural progenitors and astrocytes in vivo. Genes Dev 15(15):1913–1925PubMed
101.
go back to reference Matei D, Emerson RE, Lai YC et al (2006) Autocrine activation of PDGFRalpha promotes the progression of ovarian cancer. Oncogene 25(14):2060–2069PubMed Matei D, Emerson RE, Lai YC et al (2006) Autocrine activation of PDGFRalpha promotes the progression of ovarian cancer. Oncogene 25(14):2060–2069PubMed
102.
go back to reference Ko YJ, Small EJ, Kabbinavar F et al (2001) A multi-institutional phase ii study of SU101, a platelet-derived growth factor receptor inhibitor, for patients with hormone-refractory prostate cancer. Clin Cancer Res 7(4):800–805PubMed Ko YJ, Small EJ, Kabbinavar F et al (2001) A multi-institutional phase ii study of SU101, a platelet-derived growth factor receptor inhibitor, for patients with hormone-refractory prostate cancer. Clin Cancer Res 7(4):800–805PubMed
103.
go back to reference Carvalho I, Milanezi F, Martins A et al (2005) Overexpression of platelet-derived growth factor receptor alpha in breast cancer is associated with tumour progression. Breast Cancer Res 7(5):R788–R795PubMed Carvalho I, Milanezi F, Martins A et al (2005) Overexpression of platelet-derived growth factor receptor alpha in breast cancer is associated with tumour progression. Breast Cancer Res 7(5):R788–R795PubMed
104.
go back to reference Jechlinger M, Sommer A, Moriggl R et al (2006) Autocrine PDGFR signaling promotes mammary cancer metastasis. J Clin Invest 116(6):1561–1570PubMed Jechlinger M, Sommer A, Moriggl R et al (2006) Autocrine PDGFR signaling promotes mammary cancer metastasis. J Clin Invest 116(6):1561–1570PubMed
105.
go back to reference Kalluri R, Zeisberg M (2006) Fibroblasts in cancer. Nat Rev Cancer 6(5):392–401PubMed Kalluri R, Zeisberg M (2006) Fibroblasts in cancer. Nat Rev Cancer 6(5):392–401PubMed
Metadata
Title
Bypassing cellular EGF receptor dependence through epithelial-to-mesenchymal-like transitions
Authors
Sharon Barr
Stuart Thomson
Elizabeth Buck
Suzanne Russo
Filippo Petti
Izabela Sujka-Kwok
Alexandra Eyzaguirre
Maryland Rosenfeld-Franklin
Neil W. Gibson
Mark Miglarese
David Epstein
Kenneth K. Iwata
John D. Haley
Publication date
01-10-2008
Publisher
Springer Netherlands
Published in
Clinical & Experimental Metastasis / Issue 6/2008
Print ISSN: 0262-0898
Electronic ISSN: 1573-7276
DOI
https://doi.org/10.1007/s10585-007-9121-7

Other articles of this Issue 6/2008

Clinical & Experimental Metastasis 6/2008 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