Skip to main content
Top
Published in: Breast Cancer Research and Treatment 2/2017

01-11-2017 | Preclinical study

Metalloprotease-dependent activation of EGFR modulates CD44+/CD24 populations in triple negative breast cancer cells through the MEK/ERK pathway

Authors: Randi Wise, Anna Zolkiewska

Published in: Breast Cancer Research and Treatment | Issue 2/2017

Login to get access

Abstract

Purpose

The CD44+/CD24 cell phenotype is enriched in triple negative breast cancers, is associated with tumor invasive properties, and serves as a cell surface marker profile of breast cancer stem-like cells. Activation of Epidermal Growth Factor Receptor (EGFR) promotes the CD44+/CD24 phenotype, but the specific signaling pathway downstream of EGFR responsible for this effect is not clear. The purpose of this study was to determine the role of the MEK/ERK pathway in the expansion of CD44+/CD24 populations in TNBC cells in response to EGFR activation.

Methods

Representative TNBC cell lines SUM159PT (claudin-low) and SUM149PT (basal) were used to evaluate cell surface expression of CD44 and CD24 by flow cytometry in response to EGFR and MEK inhibition or activation. EGFR and ERK phosphorylation levels were analyzed by Western blotting. The relationship between EGFR phosphorylation and MEK activation score in basal and claudin-low tumors from the TCGA database was examined.

Results

Inhibition of ERK activation with selumetinib, a MEK1/2 inhibitor, blocked EGF-induced expansion of CD44+/CD24 populations. Sustained activation of ERK by overexpression of constitutively active MEK1 was sufficient to expand CD44+/CD24 populations in cells in which EGFR activity was blocked by either erlotinib, an EGFR kinase inhibitor, or BB-94, a metalloprotease inhibitor that prevents generation of soluble EGFR ligands. In basal and claudin-low tumors from the TCGA database, there was a positive correlation between EGFR_pY1068 and MEK activation score in tumors without genomic loss of DUSP4, a negative regulator of ERK, but not in tumors harboring DUSP4 deletion.

Conclusion

Our results demonstrate that ERK activation is a key event in EGFR-dependent regulation of CD44+/CD24 populations. Furthermore, our findings highlight the role of ligand-mediated EGFR signaling in the control of MEK/ERK pathway output in TNBC tumors without DUSP4 loss.
Appendix
Available only for authorised users
Literature
1.
go back to reference Masuda H, Zhang D, Bartholomeusz C, Doihara H, Hortobagyi GN, Ueno NT (2012) Role of epidermal growth factor receptor in breast cancer. Breast Cancer Res Treat 136:331–345CrossRefPubMed Masuda H, Zhang D, Bartholomeusz C, Doihara H, Hortobagyi GN, Ueno NT (2012) Role of epidermal growth factor receptor in breast cancer. Breast Cancer Res Treat 136:331–345CrossRefPubMed
2.
go back to reference Williams CB, Soloff AC, Ethier SP, Yeh ES (2015) Perspectives on epidermal growth factor receptor regulation in triple-negative breast cancer: ligand-mediated mechanisms of receptor regulation and potential for clinical targeting. Adv Cancer Res 127:253–281CrossRefPubMed Williams CB, Soloff AC, Ethier SP, Yeh ES (2015) Perspectives on epidermal growth factor receptor regulation in triple-negative breast cancer: ligand-mediated mechanisms of receptor regulation and potential for clinical targeting. Adv Cancer Res 127:253–281CrossRefPubMed
3.
go back to reference Hsu JL, Hung MC (2016) The role of HER2, EGFR, and other receptor tyrosine kinases in breast cancer. Cancer Metastasis Rev 35:575–588CrossRefPubMed Hsu JL, Hung MC (2016) The role of HER2, EGFR, and other receptor tyrosine kinases in breast cancer. Cancer Metastasis Rev 35:575–588CrossRefPubMed
4.
go back to reference Verbeek BS, Adriaansen-Slot SS, Vroom TM, Beckers T, Rijksen G (1998) Overexpression of EGFR and c-erbB2 causes enhanced cell migration in human breast cancer cells and NIH3T3 fibroblasts. FEBS Lett 425:145–150CrossRefPubMed Verbeek BS, Adriaansen-Slot SS, Vroom TM, Beckers T, Rijksen G (1998) Overexpression of EGFR and c-erbB2 causes enhanced cell migration in human breast cancer cells and NIH3T3 fibroblasts. FEBS Lett 425:145–150CrossRefPubMed
5.
go back to reference Mukhopadhyay P, Lakshmanan I, Ponnusamy MP, Chakraborty S, Jain M, Pai P et al (2013) MUC4 overexpression augments cell migration and metastasis through EGFR family proteins in triple negative breast cancer cells. PLoS ONE 8:e54455CrossRefPubMedPubMedCentral Mukhopadhyay P, Lakshmanan I, Ponnusamy MP, Chakraborty S, Jain M, Pai P et al (2013) MUC4 overexpression augments cell migration and metastasis through EGFR family proteins in triple negative breast cancer cells. PLoS ONE 8:e54455CrossRefPubMedPubMedCentral
6.
go back to reference Maretzky T, Evers A, Zhou W, Swendeman SL, Wong PM, Rafii S et al (2011) Migration of growth factor-stimulated epithelial and endothelial cells depends on EGFR transactivation by ADAM17. Nat Commun 2:229CrossRefPubMedPubMedCentral Maretzky T, Evers A, Zhou W, Swendeman SL, Wong PM, Rafii S et al (2011) Migration of growth factor-stimulated epithelial and endothelial cells depends on EGFR transactivation by ADAM17. Nat Commun 2:229CrossRefPubMedPubMedCentral
7.
go back to reference Reginato MJ, Mills KR, Paulus JK, Lynch DK, Sgroi DC, Debnath J et al (2003) Integrins and EGFR coordinately regulate the pro-apoptotic protein Bim to prevent anoikis. Nat Cell Biol 5:733–740CrossRefPubMed Reginato MJ, Mills KR, Paulus JK, Lynch DK, Sgroi DC, Debnath J et al (2003) Integrins and EGFR coordinately regulate the pro-apoptotic protein Bim to prevent anoikis. Nat Cell Biol 5:733–740CrossRefPubMed
8.
go back to reference Ahmed N, Maines-Bandiera S, Quinn MA, Unger WG, Dedhar S, Auersperg N (2006) Molecular pathways regulating EGF-induced epithelio-mesenchymal transition in human ovarian surface epithelium. Am J Physiol Cell Physiol 290:C1532–C1542CrossRefPubMed Ahmed N, Maines-Bandiera S, Quinn MA, Unger WG, Dedhar S, Auersperg N (2006) Molecular pathways regulating EGF-induced epithelio-mesenchymal transition in human ovarian surface epithelium. Am J Physiol Cell Physiol 290:C1532–C1542CrossRefPubMed
9.
go back to reference Lo HW, Hsu SC, Xia W, Cao X, Shih JY, Wei Y et al (2007) Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelial-mesenchymal transition in cancer cells via up-regulation of TWIST gene expression. Cancer Res 67:9066–9076CrossRefPubMedPubMedCentral Lo HW, Hsu SC, Xia W, Cao X, Shih JY, Wei Y et al (2007) Epidermal growth factor receptor cooperates with signal transducer and activator of transcription 3 to induce epithelial-mesenchymal transition in cancer cells via up-regulation of TWIST gene expression. Cancer Res 67:9066–9076CrossRefPubMedPubMedCentral
10.
go back to reference Zhang D, LaFortune TA, Krishnamurthy S, Esteva FJ, Cristofanilli M, Liu P et al (2009) Epidermal growth factor receptor tyrosine kinase inhibitor reverses mesenchymal to epithelial phenotype and inhibits metastasis in inflammatory breast cancer. Clin Cancer Res 15:6639–6648CrossRefPubMedPubMedCentral Zhang D, LaFortune TA, Krishnamurthy S, Esteva FJ, Cristofanilli M, Liu P et al (2009) Epidermal growth factor receptor tyrosine kinase inhibitor reverses mesenchymal to epithelial phenotype and inhibits metastasis in inflammatory breast cancer. Clin Cancer Res 15:6639–6648CrossRefPubMedPubMedCentral
11.
go back to reference Bartholomeusz C, Xie X, Pitner MK, Kondo K, Dadbin A, Lee J et al (2015) MEK inhibitor Selumetinib (AZD6244; ARRY-142886) prevents lung metastasis in a triple-negative breast cancer xenograft model. Mol Cancer Ther 14:2773–2781CrossRefPubMedPubMedCentral Bartholomeusz C, Xie X, Pitner MK, Kondo K, Dadbin A, Lee J et al (2015) MEK inhibitor Selumetinib (AZD6244; ARRY-142886) prevents lung metastasis in a triple-negative breast cancer xenograft model. Mol Cancer Ther 14:2773–2781CrossRefPubMedPubMedCentral
12.
go back to reference Wang M, Kern AM, Hulskotter M, Greninger P, Singh A, Pan Y et al (2014) EGFR-mediated chromatin condensation protects KRAS-mutant cancer cells against ionizing radiation. Cancer Res 74:2825–2834CrossRefPubMedPubMedCentral Wang M, Kern AM, Hulskotter M, Greninger P, Singh A, Pan Y et al (2014) EGFR-mediated chromatin condensation protects KRAS-mutant cancer cells against ionizing radiation. Cancer Res 74:2825–2834CrossRefPubMedPubMedCentral
13.
go back to reference Wei Y, Zou Z, Becker N, Anderson M, Sumpter R, Xiao G et al (2013) EGFR-mediated Beclin 1 phosphorylation in autophagy suppression, tumor progression, and tumor chemoresistance. Cell 154:1269–1284CrossRefPubMedPubMedCentral Wei Y, Zou Z, Becker N, Anderson M, Sumpter R, Xiao G et al (2013) EGFR-mediated Beclin 1 phosphorylation in autophagy suppression, tumor progression, and tumor chemoresistance. Cell 154:1269–1284CrossRefPubMedPubMedCentral
14.
go back to reference Honeth G, Bendahl PO, Ringner M, Saal LH, Gruvberger-Saal SK, Lovgren K et al (2008) The CD44+/CD24− phenotype is enriched in basal-like breast tumors. Breast Cancer Res 10:R53CrossRefPubMedPubMedCentral Honeth G, Bendahl PO, Ringner M, Saal LH, Gruvberger-Saal SK, Lovgren K et al (2008) The CD44+/CD24 phenotype is enriched in basal-like breast tumors. Breast Cancer Res 10:R53CrossRefPubMedPubMedCentral
15.
go back to reference Prat A, Adamo B, Cheang MC, Anders CK, Carey LA, Perou CM (2013) Molecular characterization of basal-like and non-basal-like triple-negative breast cancer. Oncologist 18:123–133CrossRefPubMedPubMedCentral Prat A, Adamo B, Cheang MC, Anders CK, Carey LA, Perou CM (2013) Molecular characterization of basal-like and non-basal-like triple-negative breast cancer. Oncologist 18:123–133CrossRefPubMedPubMedCentral
16.
go back to reference Blick T, Hugo H, Widodo E, Waltham M, Pinto C, Mani SA et al (2010) Epithelial mesenchymal transition traits in human breast cancer cell lines parallel the CD44hi/CD24lo/− stem cell phenotype in human breast cancer. J Mammary Gland Biol Neoplasia 15:235–252CrossRefPubMed Blick T, Hugo H, Widodo E, Waltham M, Pinto C, Mani SA et al (2010) Epithelial mesenchymal transition traits in human breast cancer cell lines parallel the CD44hi/CD24lo/− stem cell phenotype in human breast cancer. J Mammary Gland Biol Neoplasia 15:235–252CrossRefPubMed
17.
go back to reference Sheridan C, Kishimoto H, Fuchs RK, Mehrotra S, Bhat-Nakshatri P, Turner CH et al (2006) CD44+/CD24− breast cancer cells exhibit enhanced invasive properties: an early step necessary for metastasis. Breast Cancer Res 8:R59CrossRefPubMedPubMedCentral Sheridan C, Kishimoto H, Fuchs RK, Mehrotra S, Bhat-Nakshatri P, Turner CH et al (2006) CD44+/CD24 breast cancer cells exhibit enhanced invasive properties: an early step necessary for metastasis. Breast Cancer Res 8:R59CrossRefPubMedPubMedCentral
18.
go back to reference Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF (2003) Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA 100:3983–3988CrossRefPubMedPubMedCentral Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF (2003) Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA 100:3983–3988CrossRefPubMedPubMedCentral
19.
go back to reference Tanei T, Choi DS, Rodriguez AA, Liang DH, Dobrolecki L, Ghosh M et al (2016) Antitumor activity of Cetuximab in combination with Ixabepilone on triple negative breast cancer stem cells. Breast Cancer Res 18:6CrossRefPubMedPubMedCentral Tanei T, Choi DS, Rodriguez AA, Liang DH, Dobrolecki L, Ghosh M et al (2016) Antitumor activity of Cetuximab in combination with Ixabepilone on triple negative breast cancer stem cells. Breast Cancer Res 18:6CrossRefPubMedPubMedCentral
20.
go back to reference Henson E, Chen Y, Gibson S (2017) EGFR family members’ regulation of autophagy is at a crossroads of cell survival and death in cancer. Cancers (Basel) 9:27CrossRef Henson E, Chen Y, Gibson S (2017) EGFR family members’ regulation of autophagy is at a crossroads of cell survival and death in cancer. Cancers (Basel) 9:27CrossRef
21.
go back to reference Chen Y, Henson ES, Xiao W, Huang D, McMillan-Ward EM, Israels SJ et al (2016) Tyrosine kinase receptor EGFR regulates the switch in cancer cells between cell survival and cell death induced by autophagy in hypoxia. Autophagy 12:1029–1046CrossRefPubMedPubMedCentral Chen Y, Henson ES, Xiao W, Huang D, McMillan-Ward EM, Israels SJ et al (2016) Tyrosine kinase receptor EGFR regulates the switch in cancer cells between cell survival and cell death induced by autophagy in hypoxia. Autophagy 12:1029–1046CrossRefPubMedPubMedCentral
22.
23.
go back to reference Li X, Fan Z (2010) The epidermal growth factor receptor antibody cetuximab induces autophagy in cancer cells by downregulating HIF-1α and Bcl-2 and activating the beclin 1/hVps34 complex. Cancer Res 70:5942–5952CrossRefPubMedPubMedCentral Li X, Fan Z (2010) The epidermal growth factor receptor antibody cetuximab induces autophagy in cancer cells by downregulating HIF-1α and Bcl-2 and activating the beclin 1/hVps34 complex. Cancer Res 70:5942–5952CrossRefPubMedPubMedCentral
24.
go back to reference Balko JM, Schwarz LJ, Bhola NE, Kurupi R, Owens P, Miller TW et al (2013) Activation of MAPK pathways due to DUSP4 loss promotes cancer stem cell-like phenotypes in basal-like breast cancer. Cancer Res 73:6346–6358CrossRefPubMedPubMedCentral Balko JM, Schwarz LJ, Bhola NE, Kurupi R, Owens P, Miller TW et al (2013) Activation of MAPK pathways due to DUSP4 loss promotes cancer stem cell-like phenotypes in basal-like breast cancer. Cancer Res 73:6346–6358CrossRefPubMedPubMedCentral
26.
go back to reference Blobel CP (2005) ADAMs: key components in EGFR signalling and development. Nat Rev Mol Cell Biol 6:32–43CrossRefPubMed Blobel CP (2005) ADAMs: key components in EGFR signalling and development. Nat Rev Mol Cell Biol 6:32–43CrossRefPubMed
27.
go back to reference Kataoka H (2009) EGFR ligands and their signaling scissors, ADAMs, as new molecular targets for anticancer treatments. J Dermatol Sci 56:148–153CrossRefPubMed Kataoka H (2009) EGFR ligands and their signaling scissors, ADAMs, as new molecular targets for anticancer treatments. J Dermatol Sci 56:148–153CrossRefPubMed
28.
go back to reference Giltnane JM, Balko JM (2014) Rationale for targeting the Ras/MAPK pathway in triple-negative breast cancer. Discov Med 17:275–283PubMed Giltnane JM, Balko JM (2014) Rationale for targeting the Ras/MAPK pathway in triple-negative breast cancer. Discov Med 17:275–283PubMed
29.
go back to reference Mazumdar A, Poage GM, Shepherd J, Tsimelzon A, Hartman ZC, Den Hollander P et al (2016) Analysis of phosphatases in ER-negative breast cancers identifies DUSP4 as a critical regulator of growth and invasion. Breast Cancer Res Treat 158:441–454CrossRefPubMedPubMedCentral Mazumdar A, Poage GM, Shepherd J, Tsimelzon A, Hartman ZC, Den Hollander P et al (2016) Analysis of phosphatases in ER-negative breast cancers identifies DUSP4 as a critical regulator of growth and invasion. Breast Cancer Res Treat 158:441–454CrossRefPubMedPubMedCentral
30.
go back to reference Creighton CJ, Hilger AM, Murthy S, Rae JM, Chinnaiyan AM, El-Ashry D (2006) Activation of mitogen-activated protein kinase in estrogen receptor & #x03B1;-positive breast cancer cells in vitro induces an in vivo molecular phenotype of estrogen receptor & #x03B1;-negative human breast tumors. Cancer Res 66:3903–3911CrossRefPubMed Creighton CJ, Hilger AM, Murthy S, Rae JM, Chinnaiyan AM, El-Ashry D (2006) Activation of mitogen-activated protein kinase in estrogen receptor & #x03B1;-positive breast cancer cells in vitro induces an in vivo molecular phenotype of estrogen receptor & #x03B1;-negative human breast tumors. Cancer Res 66:3903–3911CrossRefPubMed
31.
go back to reference Bhat-Nakshatri P, Appaiah H, Ballas C, Pick-Franke P, Goulet R Jr, Badve S et al (2010) SLUG/SNAI2 and tumor necrosis factor generate breast cells with CD44+/CD24− phenotype. BMC Cancer 10:411CrossRefPubMedPubMedCentral Bhat-Nakshatri P, Appaiah H, Ballas C, Pick-Franke P, Goulet R Jr, Badve S et al (2010) SLUG/SNAI2 and tumor necrosis factor generate breast cells with CD44+/CD24 phenotype. BMC Cancer 10:411CrossRefPubMedPubMedCentral
32.
33.
go back to reference Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al (2013) Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6:pl1 Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al (2013) Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6:pl1
34.
go back to reference Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA et al (2012) The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2:401–404CrossRefPubMed Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA et al (2012) The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2:401–404CrossRefPubMed
35.
go back to reference Li H, Duhachek-Muggy S, Qi Y, Hong Y, Behbod F, Zolkiewska A (2012) An essential role of metalloprotease-disintegrin ADAM12 in triple-negative breast cancer. Breast Cancer Res Treat 135:759–769CrossRefPubMedPubMedCentral Li H, Duhachek-Muggy S, Qi Y, Hong Y, Behbod F, Zolkiewska A (2012) An essential role of metalloprotease-disintegrin ADAM12 in triple-negative breast cancer. Breast Cancer Res Treat 135:759–769CrossRefPubMedPubMedCentral
36.
go back to reference Li H, Duhachek-Muggy S, Dubnicka S, Zolkiewska A (2013) Metalloproteinase-disintegrin ADAM12 is associated with a breast tumor-initiating cell phenotype. Breast Cancer Res Treat 139:691–703CrossRefPubMed Li H, Duhachek-Muggy S, Dubnicka S, Zolkiewska A (2013) Metalloproteinase-disintegrin ADAM12 is associated with a breast tumor-initiating cell phenotype. Breast Cancer Res Treat 139:691–703CrossRefPubMed
37.
go back to reference Duhachek-Muggy S, Qi Y, Wise R, Alyahya L, Li H, Hodge J et al (2017) Metalloprotease-disintegrin ADAM12 actively promotes the stem cell-like phenotype in claudin-low breast cancer. Mol Cancer 16:32CrossRefPubMedPubMedCentral Duhachek-Muggy S, Qi Y, Wise R, Alyahya L, Li H, Hodge J et al (2017) Metalloprotease-disintegrin ADAM12 actively promotes the stem cell-like phenotype in claudin-low breast cancer. Mol Cancer 16:32CrossRefPubMedPubMedCentral
38.
go back to reference Creighton CJ, Li X, Landis M, Dixon JM, Neumeister VM, Sjolund A et al (2009) Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc Natl Acad Sci USA 106:13820–13825CrossRefPubMedPubMedCentral Creighton CJ, Li X, Landis M, Dixon JM, Neumeister VM, Sjolund A et al (2009) Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc Natl Acad Sci USA 106:13820–13825CrossRefPubMedPubMedCentral
39.
go back to reference Prat A, Parker JS, Karginova O, Fan C, Livasy C, Herschkowitz JI et al (2010) Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res 12:R68CrossRefPubMedPubMedCentral Prat A, Parker JS, Karginova O, Fan C, Livasy C, Herschkowitz JI et al (2010) Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res 12:R68CrossRefPubMedPubMedCentral
40.
go back to reference Haglund K, Dikic I (2012) The role of ubiquitylation in receptor endocytosis and endosomal sorting. J Cell Sci 125:265–275CrossRefPubMed Haglund K, Dikic I (2012) The role of ubiquitylation in receptor endocytosis and endosomal sorting. J Cell Sci 125:265–275CrossRefPubMed
41.
go back to reference Sorkin A, Goh LK (2009) Endocytosis and intracellular trafficking of ErbBs. Exp Cell Res 315:396–683CrossRef Sorkin A, Goh LK (2009) Endocytosis and intracellular trafficking of ErbBs. Exp Cell Res 315:396–683CrossRef
42.
go back to reference Hollestelle A, Elstrodt F, Nagel JH, Kallemeijn WW, Schutte M (2007) Phosphatidylinositol-3-OH kinase or RAS pathway mutations in human breast cancer cell lines. Mol Cancer Res 5:195–201CrossRefPubMed Hollestelle A, Elstrodt F, Nagel JH, Kallemeijn WW, Schutte M (2007) Phosphatidylinositol-3-OH kinase or RAS pathway mutations in human breast cancer cell lines. Mol Cancer Res 5:195–201CrossRefPubMed
43.
go back to reference Fillmore CM, Kuperwasser C (2008) Human breast cancer cell lines contain stem-like cells that self-renew, give rise to phenotypically diverse progeny and survive chemotherapy. Breast Cancer Res 10:R25CrossRefPubMedPubMedCentral Fillmore CM, Kuperwasser C (2008) Human breast cancer cell lines contain stem-like cells that self-renew, give rise to phenotypically diverse progeny and survive chemotherapy. Breast Cancer Res 10:R25CrossRefPubMedPubMedCentral
44.
go back to reference Keller PJ, Arendt LM, Skibinski A, Logvinenko T, Klebba I, Dong S et al (2011) Defining the cellular precursors to human breast cancer. Proc Natl Acad Sci USA 109:2772–2777CrossRefPubMedPubMedCentral Keller PJ, Arendt LM, Skibinski A, Logvinenko T, Klebba I, Dong S et al (2011) Defining the cellular precursors to human breast cancer. Proc Natl Acad Sci USA 109:2772–2777CrossRefPubMedPubMedCentral
45.
go back to reference Boehm JS, Zhao JJ, Yao J, Kim SY, Firestein R, Dunn IF et al (2007) Integrative genomic approaches identify IKBKE as a breast cancer oncogene. Cell 129:1065–1079CrossRefPubMed Boehm JS, Zhao JJ, Yao J, Kim SY, Firestein R, Dunn IF et al (2007) Integrative genomic approaches identify IKBKE as a breast cancer oncogene. Cell 129:1065–1079CrossRefPubMed
46.
go back to reference Brady DC, Crowe MS, Turski ML, Hobbs GA, Yao X, Chaikuad A et al (2014) Copper is required for oncogenic BRAF signalling and tumorigenesis. Nature 509:492–496CrossRefPubMedPubMedCentral Brady DC, Crowe MS, Turski ML, Hobbs GA, Yao X, Chaikuad A et al (2014) Copper is required for oncogenic BRAF signalling and tumorigenesis. Nature 509:492–496CrossRefPubMedPubMedCentral
47.
go back to reference Nyati MK, Morgan MA, Feng FY, Lawrence TS (2006) Integration of EGFR inhibitors with radiochemotherapy. Nat Rev Cancer 6:876–885CrossRefPubMed Nyati MK, Morgan MA, Feng FY, Lawrence TS (2006) Integration of EGFR inhibitors with radiochemotherapy. Nat Rev Cancer 6:876–885CrossRefPubMed
48.
49.
go back to reference Baselga J, Gomez P, Greil R, Braga S, Climent MA, Wardley AM et al (2013) Randomized phase II study of the anti-epidermal growth factor receptor monoclonal antibody cetuximab with cisplatin versus cisplatin alone in patients with metastatic triple-negative breast cancer. J Clin Oncol 31:2586–2592CrossRefPubMed Baselga J, Gomez P, Greil R, Braga S, Climent MA, Wardley AM et al (2013) Randomized phase II study of the anti-epidermal growth factor receptor monoclonal antibody cetuximab with cisplatin versus cisplatin alone in patients with metastatic triple-negative breast cancer. J Clin Oncol 31:2586–2592CrossRefPubMed
50.
go back to reference Carey LA, Rugo HS, Marcom PK, Mayer EL, Esteva FJ, Ma CX et al (2012) TBCRC 001: randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer. J Clin Oncol 30:2615–2623CrossRefPubMedPubMedCentral Carey LA, Rugo HS, Marcom PK, Mayer EL, Esteva FJ, Ma CX et al (2012) TBCRC 001: randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer. J Clin Oncol 30:2615–2623CrossRefPubMedPubMedCentral
51.
go back to reference Tredan O, Campone M, Jassem J, Vyzula R, Coudert B, Pacilio C et al (2015) Ixabepilone alone or with cetuximab as first-line treatment for advanced/metastatic triple-negative breast cancer. Clin Breast Cancer 15:8–15CrossRefPubMed Tredan O, Campone M, Jassem J, Vyzula R, Coudert B, Pacilio C et al (2015) Ixabepilone alone or with cetuximab as first-line treatment for advanced/metastatic triple-negative breast cancer. Clin Breast Cancer 15:8–15CrossRefPubMed
53.
go back to reference Costa R, Shah AN, Santa-Maria CA, Cruz MR, Mahalingam D, Carneiro BA et al (2017) Targeting Epidermal Growth Factor Receptor in triple negative breast cancer: new discoveries and practical insights for drug development. Cancer Treat Rev 53:111–119CrossRefPubMed Costa R, Shah AN, Santa-Maria CA, Cruz MR, Mahalingam D, Carneiro BA et al (2017) Targeting Epidermal Growth Factor Receptor in triple negative breast cancer: new discoveries and practical insights for drug development. Cancer Treat Rev 53:111–119CrossRefPubMed
54.
go back to reference Nakai K, Hung MC, Yamaguchi H (2016) A perspective on anti-EGFR therapies targeting triple-negative breast cancer. Am J Cancer Res 6:1609–1623PubMedPubMedCentral Nakai K, Hung MC, Yamaguchi H (2016) A perspective on anti-EGFR therapies targeting triple-negative breast cancer. Am J Cancer Res 6:1609–1623PubMedPubMedCentral
55.
go back to reference Fleisher B, Clarke C, Ait-Oudhia S (2016) Current advances in biomarkers for targeted therapy in triple-negative breast cancer. Breast Cancer 8:183–197PubMedPubMedCentral Fleisher B, Clarke C, Ait-Oudhia S (2016) Current advances in biomarkers for targeted therapy in triple-negative breast cancer. Breast Cancer 8:183–197PubMedPubMedCentral
56.
go back to reference Farnie G, Clarke RB, Spence K, Pinnock N, Brennan K, Anderson NG et al (2007) Novel cell culture technique for primary ductal carcinoma in situ: role of Notch and epidermal growth factor receptor signaling pathways. J Natl Cancer Inst 99:616–627CrossRefPubMed Farnie G, Clarke RB, Spence K, Pinnock N, Brennan K, Anderson NG et al (2007) Novel cell culture technique for primary ductal carcinoma in situ: role of Notch and epidermal growth factor receptor signaling pathways. J Natl Cancer Inst 99:616–627CrossRefPubMed
57.
go back to reference Cufi S, Vazquez-Martin A, Oliveras-Ferraros C, Martin-Castillo B, Vellon L, Menendez JA (2011) Autophagy positively regulates the CD44(+) CD24(-/low) breast cancer stem-like phenotype. Cell Cycle 10:3871–3885CrossRefPubMed Cufi S, Vazquez-Martin A, Oliveras-Ferraros C, Martin-Castillo B, Vellon L, Menendez JA (2011) Autophagy positively regulates the CD44(+) CD24(-/low) breast cancer stem-like phenotype. Cell Cycle 10:3871–3885CrossRefPubMed
58.
go back to reference Choi DS, Blanco E, Kim YS, Rodriguez AA, Zhao H, Huang TH et al (2014) Chloroquine eliminates cancer stem cells through deregulation of Jak2 and DNMT1. Stem Cells 32:2309–2323CrossRefPubMedPubMedCentral Choi DS, Blanco E, Kim YS, Rodriguez AA, Zhao H, Huang TH et al (2014) Chloroquine eliminates cancer stem cells through deregulation of Jak2 and DNMT1. Stem Cells 32:2309–2323CrossRefPubMedPubMedCentral
59.
go back to reference Liang DH, Choi DS, Ensor JE, Kaipparettu BA, Bass BL, Chang JC (2016) The autophagy inhibitor chloroquine targets cancer stem cells in triple negative breast cancer by inducing mitochondrial damage and impairing DNA break repair. Cancer Lett 376:249–258CrossRefPubMedPubMedCentral Liang DH, Choi DS, Ensor JE, Kaipparettu BA, Bass BL, Chang JC (2016) The autophagy inhibitor chloroquine targets cancer stem cells in triple negative breast cancer by inducing mitochondrial damage and impairing DNA break repair. Cancer Lett 376:249–258CrossRefPubMedPubMedCentral
60.
go back to reference Qiang L, Zhao B, Ming M, Wang N, He TC, Hwang S et al (2014) Regulation of cell proliferation and migration by p62 through stabilization of Twist1. Proc Natl Acad Sci USA 111:9241–9246CrossRefPubMedPubMedCentral Qiang L, Zhao B, Ming M, Wang N, He TC, Hwang S et al (2014) Regulation of cell proliferation and migration by p62 through stabilization of Twist1. Proc Natl Acad Sci USA 111:9241–9246CrossRefPubMedPubMedCentral
62.
go back to reference Bayliss J, Hilger A, Vishnu P, Diehl K, El-Ashry D (2007) Reversal of the estrogen receptor negative phenotype in breast cancer and restoration of antiestrogen response. Clin Cancer Res 13:7029–7036CrossRefPubMed Bayliss J, Hilger A, Vishnu P, Diehl K, El-Ashry D (2007) Reversal of the estrogen receptor negative phenotype in breast cancer and restoration of antiestrogen response. Clin Cancer Res 13:7029–7036CrossRefPubMed
63.
go back to reference Loi S, Dushyanthen S, Beavis PA, Salgado R, Denkert C, Savas P et al (2016) RAS/MAPK activation is associated with reduced tumor-infiltrating lymphocytes in triple-negative breast cancer: therapeutic cooperation between MEK and PD-1/PD-L1 immune checkpoint inhibitors. Clin Cancer Res 22:1499–1509CrossRefPubMed Loi S, Dushyanthen S, Beavis PA, Salgado R, Denkert C, Savas P et al (2016) RAS/MAPK activation is associated with reduced tumor-infiltrating lymphocytes in triple-negative breast cancer: therapeutic cooperation between MEK and PD-1/PD-L1 immune checkpoint inhibitors. Clin Cancer Res 22:1499–1509CrossRefPubMed
Metadata
Title
Metalloprotease-dependent activation of EGFR modulates CD44+/CD24− populations in triple negative breast cancer cells through the MEK/ERK pathway
Authors
Randi Wise
Anna Zolkiewska
Publication date
01-11-2017
Publisher
Springer US
Published in
Breast Cancer Research and Treatment / Issue 2/2017
Print ISSN: 0167-6806
Electronic ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-017-4440-0

Other articles of this Issue 2/2017

Breast Cancer Research and Treatment 2/2017 Go to the issue

Letter to the Editor

Reply to Kopans

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