Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2021

Open Access 01-12-2021 | Crizotinib | Research

The FAM3C locus that encodes interleukin-like EMT inducer (ILEI) is frequently co-amplified in MET-amplified cancers and contributes to invasiveness

Authors: Ulrike Schmidt, Gerwin Heller, Gerald Timelthaler, Petra Heffeter, Zsolt Somodi, Norbert Schweifer, Maria Sibilia, Walter Berger, Agnes Csiszar

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2021

Login to get access

Abstract

Background

Gene amplification of MET, which encodes for the receptor tyrosine kinase c-MET, occurs in a variety of human cancers. High c-MET levels often correlate with poor cancer prognosis. Interleukin-like EMT inducer (ILEI) is also overexpressed in many cancers and is associated with metastasis and poor survival. The gene for ILEI, FAM3C, is located close to MET on chromosome 7q31 in an amplification “hotspot”, but it is unclear whether FAMC3 amplification contributes to elevated ILEI expression in cancer. In this study we have investigated FAMC3 copy number gain in different cancers and its potential connection to MET amplifications.

Methods

FAMC3 and MET copy numbers were investigated in various cancer samples and 200 cancer cell lines. Copy numbers of the two genes were correlated with mRNA levels, with relapse-free survival in lung cancer patient samples as well as with clinicopathological parameters in primary samples from 49 advanced stage colorectal cancer patients. ILEI knock-down and c-MET inhibition effects on proliferation and invasiveness of five cancer cell lines and growth of xenograft tumors in mice were then investigated.

Results

FAMC3 was amplified in strict association with MET amplification in several human cancers and cancer cell lines. Increased FAM3C and MET copy numbers were tightly linked and correlated with increased gene expression and poor survival in human lung cancer and with extramural invasion in colorectal carcinoma. Stable ILEI shRNA knock-down did not influence proliferation or sensitivity towards c-MET-inhibitor induced proliferation arrest in cancer cells, but impaired both c-MET-independent and -dependent cancer cell invasion. c-MET inhibition reduced ILEI secretion, and shRNA mediated ILEI knock-down prevented c-MET-signaling induced elevated expression and secretion of matrix metalloproteinase (MMP)-2 and MMP-9. Combination of ILEI knock-down and c-MET-inhibition significantly reduced the invasive outgrowth of NCI-H441 and NCI-H1993 lung tumor xenografts by inhibiting proliferation, MMP expression and E-cadherin membrane localization.

Conclusions

These novel findings suggest MET amplifications are often in reality MET-FAM3C co-amplifications with tight functional cooperation. Therefore, the clinical relevance of this frequent cancer amplification hotspot, so far dedicated purely to c-MET function, should be re-evaluated to include ILEI as a target in the therapy of c-MET-amplified human carcinomas.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Hnisz D, Weintraub AS, Day DS, Valton AL, Bak RO, Li CH, et al. Activation of proto-oncogenes by disruption of chromosome neighborhoods. Science. 2016;351(6280):1454–8.PubMedPubMedCentralCrossRef Hnisz D, Weintraub AS, Day DS, Valton AL, Bak RO, Li CH, et al. Activation of proto-oncogenes by disruption of chromosome neighborhoods. Science. 2016;351(6280):1454–8.PubMedPubMedCentralCrossRef
4.
go back to reference Blumenschein GR Jr, Mills GB, Gonzalez-Angulo AM. Targeting the hepatocyte growth factor-cMET axis in cancer therapy. J Clin Oncol. 2012;30(26):3287–96.PubMedPubMedCentralCrossRef Blumenschein GR Jr, Mills GB, Gonzalez-Angulo AM. Targeting the hepatocyte growth factor-cMET axis in cancer therapy. J Clin Oncol. 2012;30(26):3287–96.PubMedPubMedCentralCrossRef
5.
go back to reference Hyman E, Kauraniemi P, Hautaniemi S, Wolf M, Mousses S, Rozenblum E, et al. Impact of DNA amplification on gene expression patterns in breast cancer. Cancer Res. 2002;62(21):6240–5.PubMed Hyman E, Kauraniemi P, Hautaniemi S, Wolf M, Mousses S, Rozenblum E, et al. Impact of DNA amplification on gene expression patterns in breast cancer. Cancer Res. 2002;62(21):6240–5.PubMed
6.
go back to reference Waerner T, Alacakaptan M, Tamir I, Oberauer R, Gal A, Brabletz T, et al. ILEI: a cytokine essential for EMT, tumor formation, and late events in metastasis in epithelial cells. Cancer Cell. 2006;10(3):227–39.PubMedCrossRef Waerner T, Alacakaptan M, Tamir I, Oberauer R, Gal A, Brabletz T, et al. ILEI: a cytokine essential for EMT, tumor formation, and late events in metastasis in epithelial cells. Cancer Cell. 2006;10(3):227–39.PubMedCrossRef
7.
go back to reference Jechlinger M, Grunert S, Tamir IH, Janda E, Ludemann S, Waerner T, et al. Expression profiling of epithelial plasticity in tumor progression. Oncogene. 2003;22(46):7155–69.PubMedCrossRef Jechlinger M, Grunert S, Tamir IH, Janda E, Ludemann S, Waerner T, et al. Expression profiling of epithelial plasticity in tumor progression. Oncogene. 2003;22(46):7155–69.PubMedCrossRef
8.
go back to reference Du B, Shim JS. Targeting epithelial-Mesenchymal transition (EMT) to overcome drug resistance in cancer. Molecules. 2016;21:7. Du B, Shim JS. Targeting epithelial-Mesenchymal transition (EMT) to overcome drug resistance in cancer. Molecules. 2016;21:7.
9.
go back to reference Micalizzi DS, Farabaugh SM, Ford HL. Epithelial-mesenchymal transition in cancer: parallels between normal development and tumor progression. J Mammary Gland Biol Neoplasia. 2010;15(2):117–34.PubMedPubMedCentralCrossRef Micalizzi DS, Farabaugh SM, Ford HL. Epithelial-mesenchymal transition in cancer: parallels between normal development and tumor progression. J Mammary Gland Biol Neoplasia. 2010;15(2):117–34.PubMedPubMedCentralCrossRef
10.
go back to reference Liao TT, Yang MH. Revisiting epithelial-mesenchymal transition in cancer metastasis: the connection between epithelial plasticity and stemness. Mol Oncol. 2017;11(7):792–804.PubMedPubMedCentralCrossRef Liao TT, Yang MH. Revisiting epithelial-mesenchymal transition in cancer metastasis: the connection between epithelial plasticity and stemness. Mol Oncol. 2017;11(7):792–804.PubMedPubMedCentralCrossRef
11.
go back to reference Lahsnig C, Mikula M, Petz M, Zulehner G, Schneller D, van Zijl F, et al. ILEI requires oncogenic Ras for the epithelial to mesenchymal transition of hepatocytes and liver carcinoma progression. Oncogene. 2009;28(5):638–50.PubMedCrossRef Lahsnig C, Mikula M, Petz M, Zulehner G, Schneller D, van Zijl F, et al. ILEI requires oncogenic Ras for the epithelial to mesenchymal transition of hepatocytes and liver carcinoma progression. Oncogene. 2009;28(5):638–50.PubMedCrossRef
12.
go back to reference Song Q, Sheng W, Zhang X, Jiao S, Li F. ILEI drives epithelial to mesenchymal transition and metastatic progression in the lung cancer cell line A549. Tumour Biol. 2014;35(2):1377–82.PubMedCrossRef Song Q, Sheng W, Zhang X, Jiao S, Li F. ILEI drives epithelial to mesenchymal transition and metastatic progression in the lung cancer cell line A549. Tumour Biol. 2014;35(2):1377–82.PubMedCrossRef
13.
go back to reference Csiszar A, Kutay B, Wirth S, Schmidt U, Macho-Maschler S, Schreiber M, et al. Interleukin-like epithelial-to-mesenchymal transition inducer activity is controlled by proteolytic processing and plasminogen inverted question mark urokinase plasminogen activator receptor system-regulated secretion during breast cancer progression. Breast Cancer Res. 2014;16(5):433.PubMedPubMedCentralCrossRef Csiszar A, Kutay B, Wirth S, Schmidt U, Macho-Maschler S, Schreiber M, et al. Interleukin-like epithelial-to-mesenchymal transition inducer activity is controlled by proteolytic processing and plasminogen inverted question mark urokinase plasminogen activator receptor system-regulated secretion during breast cancer progression. Breast Cancer Res. 2014;16(5):433.PubMedPubMedCentralCrossRef
14.
go back to reference Gao ZH, Lu C, Wang ZN, Song YX, Zhu JL, Gao P, et al. ILEI: a novel marker for epithelial-mesenchymal transition and poor prognosis in colorectal cancer. Histopathology. 2014;65(4):527–38.PubMedCrossRef Gao ZH, Lu C, Wang ZN, Song YX, Zhu JL, Gao P, et al. ILEI: a novel marker for epithelial-mesenchymal transition and poor prognosis in colorectal cancer. Histopathology. 2014;65(4):527–38.PubMedCrossRef
15.
go back to reference Weksberg R, Hughes S, Moldovan L, Bassett AS, Chow EW, Squire JA. A method for accurate detection of genomic microdeletions using real-time quantitative PCR. BMC Genomics. 2005;6:180.PubMedPubMedCentralCrossRef Weksberg R, Hughes S, Moldovan L, Bassett AS, Chow EW, Squire JA. A method for accurate detection of genomic microdeletions using real-time quantitative PCR. BMC Genomics. 2005;6:180.PubMedPubMedCentralCrossRef
16.
go back to reference Smolen GA, Sordella R, Muir B, Mohapatra G, Barmettler A, Archibald H, et al. Amplification of MET may identify a subset of cancers with extreme sensitivity to the selective tyrosine kinase inhibitor PHA-665752. Proc Natl Acad Sci U S A. 2006;103(7):2316–21.PubMedPubMedCentralCrossRef Smolen GA, Sordella R, Muir B, Mohapatra G, Barmettler A, Archibald H, et al. Amplification of MET may identify a subset of cancers with extreme sensitivity to the selective tyrosine kinase inhibitor PHA-665752. Proc Natl Acad Sci U S A. 2006;103(7):2316–21.PubMedPubMedCentralCrossRef
17.
go back to reference Kral M, Klimek C, Kutay B, Timelthaler G, Lendl T, Neuditschko B, et al. Covalent dimerization of interleukin-like epithelial-to-mesenchymal transition (EMT) inducer (ILEI) facilitates EMT, invasion, and late aspects of metastasis. FEBS J. 2017;284(20):3484–505.PubMedCrossRef Kral M, Klimek C, Kutay B, Timelthaler G, Lendl T, Neuditschko B, et al. Covalent dimerization of interleukin-like epithelial-to-mesenchymal transition (EMT) inducer (ILEI) facilitates EMT, invasion, and late aspects of metastasis. FEBS J. 2017;284(20):3484–505.PubMedCrossRef
18.
19.
go back to reference McDermott U, Sharma SV, Dowell L, Greninger P, Montagut C, Lamb J, et al. Identification of genotype-correlated sensitivity to selective kinase inhibitors by using high-throughput tumor cell line profiling. Proc Natl Acad Sci U S A. 2007;104(50):19936–41.PubMedPubMedCentralCrossRef McDermott U, Sharma SV, Dowell L, Greninger P, Montagut C, Lamb J, et al. Identification of genotype-correlated sensitivity to selective kinase inhibitors by using high-throughput tumor cell line profiling. Proc Natl Acad Sci U S A. 2007;104(50):19936–41.PubMedPubMedCentralCrossRef
20.
go back to reference Ikebuchi F, Oka K, Mizuno S, Fukuta K, Hayata D, Ohnishi H, et al. Dissociation of c-met phosphotyrosine sites in human cells in response to mouse hepatocyte growth factor but not human hepatocyte growth factor: the possible roles of different amino acids in different species. Cell Biochem Funct. 2013;31(4):298–304.PubMedCrossRef Ikebuchi F, Oka K, Mizuno S, Fukuta K, Hayata D, Ohnishi H, et al. Dissociation of c-met phosphotyrosine sites in human cells in response to mouse hepatocyte growth factor but not human hepatocyte growth factor: the possible roles of different amino acids in different species. Cell Biochem Funct. 2013;31(4):298–304.PubMedCrossRef
21.
go back to reference Rong S, Bodescot M, Blair D, Dunn J, Nakamura T, Mizuno K, et al. Tumorigenicity of the met proto-oncogene and the gene for hepatocyte growth factor. Mol Cell Biol. 1992;12(11):5152–8.PubMedPubMedCentralCrossRef Rong S, Bodescot M, Blair D, Dunn J, Nakamura T, Mizuno K, et al. Tumorigenicity of the met proto-oncogene and the gene for hepatocyte growth factor. Mol Cell Biol. 1992;12(11):5152–8.PubMedPubMedCentralCrossRef
22.
go back to reference Winer A, Adams S, Mignatti P. Matrix metalloproteinase inhibitors in cancer therapy: turning past failures into future successes. Mol Cancer Ther. 2018;17(6):1147–55.PubMedPubMedCentralCrossRef Winer A, Adams S, Mignatti P. Matrix metalloproteinase inhibitors in cancer therapy: turning past failures into future successes. Mol Cancer Ther. 2018;17(6):1147–55.PubMedPubMedCentralCrossRef
23.
go back to reference Kermorgant S, Aparicio T, Dessirier V, Lewin MJ, Lehy T. Hepatocyte growth factor induces colonic cancer cell invasiveness via enhanced motility and protease overproduction. Evidence for PI3 kinase and PKC involvement. Carcinogenesis. 2001;22(7):1035–42.PubMedCrossRef Kermorgant S, Aparicio T, Dessirier V, Lewin MJ, Lehy T. Hepatocyte growth factor induces colonic cancer cell invasiveness via enhanced motility and protease overproduction. Evidence for PI3 kinase and PKC involvement. Carcinogenesis. 2001;22(7):1035–42.PubMedCrossRef
24.
go back to reference Fujiuchi Y, Nagakawa O, Murakami K, Fuse H, Saiki I. Effect of hepatocyte growth factor on invasion of prostate cancer cell lines. Oncol Rep. 2003;10(4):1001–6.PubMed Fujiuchi Y, Nagakawa O, Murakami K, Fuse H, Saiki I. Effect of hepatocyte growth factor on invasion of prostate cancer cell lines. Oncol Rep. 2003;10(4):1001–6.PubMed
25.
go back to reference Bure IV, Nemtsova MV, Zaletaev DV. Roles of E-cadherin and noncoding RNAs in the epithelial-mesenchymal transition and progression in gastric cancer. Int J Mol Sci. 2019;20:12.CrossRef Bure IV, Nemtsova MV, Zaletaev DV. Roles of E-cadherin and noncoding RNAs in the epithelial-mesenchymal transition and progression in gastric cancer. Int J Mol Sci. 2019;20:12.CrossRef
26.
go back to reference Zou HY, Li Q, Lee JH, Arango ME, McDonnell SR, Yamazaki S, et al. An orally available small-molecule inhibitor of c-met, PF-2341066, exhibits cytoreductive antitumor efficacy through antiproliferative and antiangiogenic mechanisms. Cancer Res. 2007;67(9):4408–17.PubMedCrossRef Zou HY, Li Q, Lee JH, Arango ME, McDonnell SR, Yamazaki S, et al. An orally available small-molecule inhibitor of c-met, PF-2341066, exhibits cytoreductive antitumor efficacy through antiproliferative and antiangiogenic mechanisms. Cancer Res. 2007;67(9):4408–17.PubMedCrossRef
27.
go back to reference Miura H, Nishimura K, Tsujimura A, Matsumiya K, Matsumoto K, Nakamura T, et al. Effects of hepatocyte growth factor on E-cadherin-mediated cell-cell adhesion in DU145 prostate cancer cells. Urology. 2001;58(6):1064–9.PubMedCrossRef Miura H, Nishimura K, Tsujimura A, Matsumiya K, Matsumoto K, Nakamura T, et al. Effects of hepatocyte growth factor on E-cadherin-mediated cell-cell adhesion in DU145 prostate cancer cells. Urology. 2001;58(6):1064–9.PubMedCrossRef
28.
go back to reference Menakongka A, Suthiphongchai T. Involvement of PI3K and ERK1/2 pathways in hepatocyte growth factor-induced cholangiocarcinoma cell invasion. World J Gastroenterol. 2010;16(6):713–22.PubMedPubMedCentralCrossRef Menakongka A, Suthiphongchai T. Involvement of PI3K and ERK1/2 pathways in hepatocyte growth factor-induced cholangiocarcinoma cell invasion. World J Gastroenterol. 2010;16(6):713–22.PubMedPubMedCentralCrossRef
29.
go back to reference Wang Y, Jiang Z, Xu C, Wang H, Tan L, Su J, et al. Increased MET gene copy number negatively affects the survival of esophageal squamous cell carcinoma patients. BMC Cancer. 2019;19(1):240.PubMedPubMedCentralCrossRef Wang Y, Jiang Z, Xu C, Wang H, Tan L, Su J, et al. Increased MET gene copy number negatively affects the survival of esophageal squamous cell carcinoma patients. BMC Cancer. 2019;19(1):240.PubMedPubMedCentralCrossRef
30.
go back to reference Cappuzzo F, Marchetti A, Skokan M, Rossi E, Gajapathy S, Felicioni L, et al. Increased MET gene copy number negatively affects survival of surgically resected non-small-cell lung cancer patients. J Clin Oncol. 2009;27(10):1667–74.PubMedPubMedCentralCrossRef Cappuzzo F, Marchetti A, Skokan M, Rossi E, Gajapathy S, Felicioni L, et al. Increased MET gene copy number negatively affects survival of surgically resected non-small-cell lung cancer patients. J Clin Oncol. 2009;27(10):1667–74.PubMedPubMedCentralCrossRef
31.
go back to reference Macher-Goeppinger S, Keith M, Endris V, Penzel R, Tagscherer KE, Pahernik S, et al. MET expression and copy number status in clear-cell renal cell carcinoma: prognostic value and potential predictive marker. Oncotarget. 2017;8(1):1046–57.PubMedCrossRef Macher-Goeppinger S, Keith M, Endris V, Penzel R, Tagscherer KE, Pahernik S, et al. MET expression and copy number status in clear-cell renal cell carcinoma: prognostic value and potential predictive marker. Oncotarget. 2017;8(1):1046–57.PubMedCrossRef
32.
go back to reference Kim WY, Shim SH, Jung HY, Dong M, Kim SN, Lee SJ. The gene copy number of c-MET has a significant impact on progression-free survival in Korean patients with ovarian carcinoma. Hum Pathol. 2017;64:98–105.PubMedCrossRef Kim WY, Shim SH, Jung HY, Dong M, Kim SN, Lee SJ. The gene copy number of c-MET has a significant impact on progression-free survival in Korean patients with ovarian carcinoma. Hum Pathol. 2017;64:98–105.PubMedCrossRef
33.
go back to reference Lee SJ, Lee J, Sohn I, Mao M, Kai W, Park CK, et al. A survey of c-MET expression and amplification in 287 patients with hepatocellular carcinoma. Anticancer Res. 2013;33(11):5179–86.PubMed Lee SJ, Lee J, Sohn I, Mao M, Kai W, Park CK, et al. A survey of c-MET expression and amplification in 287 patients with hepatocellular carcinoma. Anticancer Res. 2013;33(11):5179–86.PubMed
34.
go back to reference Sivakumar M, Jayakumar M, Seedevi P, Sivasankar P, Ravikumar M, Surendar S, et al. Meta-analysis of functional expression and mutational analysis of c-met in various cancers. Curr Probl Cancer. 2019;44:100515.PubMedCrossRef Sivakumar M, Jayakumar M, Seedevi P, Sivasankar P, Ravikumar M, Surendar S, et al. Meta-analysis of functional expression and mutational analysis of c-met in various cancers. Curr Probl Cancer. 2019;44:100515.PubMedCrossRef
35.
go back to reference Mignard X, Ruppert AM, Antoine M, Vasseur J, Girard N, Mazieres J, et al. C-MET overexpression as a poor predictor of MET amplifications or exon 14 mutations in lung Sarcomatoid carcinomas. J Thorac Oncol. 2018;13(12):1962–7.PubMedCrossRef Mignard X, Ruppert AM, Antoine M, Vasseur J, Girard N, Mazieres J, et al. C-MET overexpression as a poor predictor of MET amplifications or exon 14 mutations in lung Sarcomatoid carcinomas. J Thorac Oncol. 2018;13(12):1962–7.PubMedCrossRef
36.
go back to reference Kwak Y, Yun S, Nam SK, Seo AN, Lee KS, Shin E, et al. Comparative analysis of the EGFR, HER2, c-MYC, and MET variations in colorectal cancer determined by three different measures: gene copy number gain, amplification status and the 2013 ASCO/CAP guideline criterion for HER2 testing of breast cancer. J Transl Med. 2017;15(1):167.PubMedPubMedCentralCrossRef Kwak Y, Yun S, Nam SK, Seo AN, Lee KS, Shin E, et al. Comparative analysis of the EGFR, HER2, c-MYC, and MET variations in colorectal cancer determined by three different measures: gene copy number gain, amplification status and the 2013 ASCO/CAP guideline criterion for HER2 testing of breast cancer. J Transl Med. 2017;15(1):167.PubMedPubMedCentralCrossRef
37.
go back to reference Nalesnik MA, Michalopoulos GK. Growth factor pathways in development and progression of hepatocellular carcinoma. Front Biosci (Scholar edition). 2012;4:1487–515.CrossRef Nalesnik MA, Michalopoulos GK. Growth factor pathways in development and progression of hepatocellular carcinoma. Front Biosci (Scholar edition). 2012;4:1487–515.CrossRef
38.
go back to reference Bouattour M, Raymond E, Qin S, Cheng AL, Stammberger U, Locatelli G, et al. Recent developments of c-met as a therapeutic target in hepatocellular carcinoma. Hepatology (Baltimore, Md). 2018;67(3):1132–49.CrossRef Bouattour M, Raymond E, Qin S, Cheng AL, Stammberger U, Locatelli G, et al. Recent developments of c-met as a therapeutic target in hepatocellular carcinoma. Hepatology (Baltimore, Md). 2018;67(3):1132–49.CrossRef
39.
go back to reference Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S, Turke AB, Fidias P, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med. 2011;3(75):75ra26.PubMedPubMedCentralCrossRef Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S, Turke AB, Fidias P, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med. 2011;3(75):75ra26.PubMedPubMedCentralCrossRef
41.
go back to reference Lindemann K, Resau J, Nahrig J, Kort E, Leeser B, Annecke K, et al. Differential expression of c-met, its ligand HGF/SF and HER2/neu in DCIS and adjacent normal breast tissue. Histopathology. 2007;51(1):54–62.PubMedCrossRef Lindemann K, Resau J, Nahrig J, Kort E, Leeser B, Annecke K, et al. Differential expression of c-met, its ligand HGF/SF and HER2/neu in DCIS and adjacent normal breast tissue. Histopathology. 2007;51(1):54–62.PubMedCrossRef
42.
go back to reference Woosley AN, Dalton AC, Hussey GS, Howley BV, Mohanty BK, Grelet S, et al. TGFbeta promotes breast cancer stem cell self-renewal through an ILEI/LIFR signaling axis. Oncogene. 2019;38(20):3794–811.PubMedPubMedCentralCrossRef Woosley AN, Dalton AC, Hussey GS, Howley BV, Mohanty BK, Grelet S, et al. TGFbeta promotes breast cancer stem cell self-renewal through an ILEI/LIFR signaling axis. Oncogene. 2019;38(20):3794–811.PubMedPubMedCentralCrossRef
43.
go back to reference Jansson AM, Csiszar A, Maier J, Nystrom AC, Ax E, Johansson P, et al. The interleukin-like epithelial-mesenchymal transition inducer ILEI exhibits a non-interleukin-like fold and is active as a domain-swapped dimer. J Biol Chem. 2017;292(37):15501–11.PubMedPubMedCentralCrossRef Jansson AM, Csiszar A, Maier J, Nystrom AC, Ax E, Johansson P, et al. The interleukin-like epithelial-mesenchymal transition inducer ILEI exhibits a non-interleukin-like fold and is active as a domain-swapped dimer. J Biol Chem. 2017;292(37):15501–11.PubMedPubMedCentralCrossRef
44.
go back to reference Noguchi K, Dincman TA, Dalton AC, Howley BV, McCall BJ, Mohanty BK, et al. Interleukin-like EMT inducer (ILEI) promotes melanoma invasiveness and is transcriptionally up-regulated by upstream stimulatory factor-1 (USF-1). J Biol Chem. 2018;293(29):11401–14.PubMedPubMedCentralCrossRef Noguchi K, Dincman TA, Dalton AC, Howley BV, McCall BJ, Mohanty BK, et al. Interleukin-like EMT inducer (ILEI) promotes melanoma invasiveness and is transcriptionally up-regulated by upstream stimulatory factor-1 (USF-1). J Biol Chem. 2018;293(29):11401–14.PubMedPubMedCentralCrossRef
45.
go back to reference Lustosa SA, Viana Lde S, Affonso RJ Jr, Silva SR, Denadai MV, de Toledo SR, et al. Expression profiling using a cDNA array and immunohistochemistry for the extracellular matrix genes FN-1, ITGA-3, ITGB-5, MMP-2, and MMP-9 in colorectal carcinoma progression and dissemination. ScientificWorldJournal. 2014;2014:102541.PubMedPubMedCentralCrossRef Lustosa SA, Viana Lde S, Affonso RJ Jr, Silva SR, Denadai MV, de Toledo SR, et al. Expression profiling using a cDNA array and immunohistochemistry for the extracellular matrix genes FN-1, ITGA-3, ITGB-5, MMP-2, and MMP-9 in colorectal carcinoma progression and dissemination. ScientificWorldJournal. 2014;2014:102541.PubMedPubMedCentralCrossRef
46.
go back to reference Bruner HC, Derksen PWB. Loss of E-cadherin-dependent cell-cell adhesion and the development and progression of cancer. Cold Spring Harb Perspect Biol. 2018;10:3.CrossRef Bruner HC, Derksen PWB. Loss of E-cadherin-dependent cell-cell adhesion and the development and progression of cancer. Cold Spring Harb Perspect Biol. 2018;10:3.CrossRef
47.
go back to reference Wong SHM, Fang CM, Chuah LH, Leong CO, Ngai SC. E-cadherin: its dysregulation in carcinogenesis and clinical implications. Crit Rev Oncol Hematol. 2018;121:11–22.PubMedCrossRef Wong SHM, Fang CM, Chuah LH, Leong CO, Ngai SC. E-cadherin: its dysregulation in carcinogenesis and clinical implications. Crit Rev Oncol Hematol. 2018;121:11–22.PubMedCrossRef
48.
go back to reference Ghafoory S, Mehrabi A, Hafezi M, Cheng X, Breitkopf-Heinlein K, Hick M, et al. Nuclear accumulation of CDH1 mRNA in hepatocellular carcinoma cells. Oncogenesis. 2015;4:e152.PubMedPubMedCentralCrossRef Ghafoory S, Mehrabi A, Hafezi M, Cheng X, Breitkopf-Heinlein K, Hick M, et al. Nuclear accumulation of CDH1 mRNA in hepatocellular carcinoma cells. Oncogenesis. 2015;4:e152.PubMedPubMedCentralCrossRef
Metadata
Title
The FAM3C locus that encodes interleukin-like EMT inducer (ILEI) is frequently co-amplified in MET-amplified cancers and contributes to invasiveness
Authors
Ulrike Schmidt
Gerwin Heller
Gerald Timelthaler
Petra Heffeter
Zsolt Somodi
Norbert Schweifer
Maria Sibilia
Walter Berger
Agnes Csiszar
Publication date
01-12-2021
Publisher
BioMed Central
Keyword
Crizotinib
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2021
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-021-01862-5

Other articles of this Issue 1/2021

Journal of Experimental & Clinical Cancer Research 1/2021 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