Skip to main content
Top
Published in: Breast Cancer Research and Treatment 1/2015

01-11-2015 | Preclinical study

Differential expression of epithelial–mesenchymal transition and stem cell markers in intrinsic subtypes of breast cancer

Authors: Victoria Pomp, Cornelia Leo, Andrea Mauracher, Dimitri Korol, Wenjun Guo, Zsuzsanna Varga

Published in: Breast Cancer Research and Treatment | Issue 1/2015

Login to get access

Abstract

The transcription factors SLUG and SOX9 have been shown to define mammary stem cell state. Similarly, epithelial–mesenchymal transition (EMT) markers (E-Cadherin, mTOR) have been shown to play a role in tumor-progression and metastatic potential in breast cancer. Finally, SOX10 is known to be expressed in breast cancer as well. The overexpressions of EMT and stem cell markers have been shown to correlate with poor overall survival. In this study, we examined whether the expression of these markers correlates with intrinsic subtypes of breast cancer and whether there is a prognostic difference in their expression-profile. We analyzed 617 breast cancer samples from two tissue micro arrays. Breast cancer samples were categorized into three groups according to hormone receptor expression and HER2-status as Luminal A/B, HER2-positive, and triple negative subgroup. Immunohistochemical expressions of SLUG, SOX9, SOX10, E-Cadherin, and mTOR were semi-quantitatively analyzed using a two-tiered and three-tiered scoring system in which cytoplasmic and nuclear stains were considered. Strong nuclear expression of SLUG was observed preferentially in triple negative but not in Luminal A/B or HER2-positive cases (24 vs. 3 and 0 %, p < 0.001). Loss of SOX9 in the nuclear stain was less frequent in triple negative than in Luminal A/B or HER2-positive cases (4 vs. 9 vs. 13 %, p < 0.001). Expression of nuclear SOX10 was lower in triple negative than in Luminal A/B and HER2-positive cases (67 vs.78 and 79 %, p = 0.012). E-Cadherin loss was observed only in Luminal A/B tumors (p = 0.016), no difference in the mTOR expression was seen between any of the three groups. No correlation to conventional histopathological-parameters or stage could be established in our cohort. Our study shows an inversed preferential nuclear expression of SLUG, SOX10, and SOX9 in triple negative and non-triple negative cases. This information is important in understanding the biology of triple negative breast cancer, also in terms of future studies dealing with targeted therapies based on the alterations of EMT and stem cell markers.
Literature
1.
go back to reference Alkatout I, Wiedermann M, Bauer M, Wenners A, Jonat W, Klapper W (2013) Transcription factors associated with epithelial–mesenchymal transition and cancer stem cells in the tumor centre and margin of invasive breast cancer. Exp Mol Pathol 94:168–173. doi:10.1016/j.yexmp.2012.09.003 CrossRefPubMed Alkatout I, Wiedermann M, Bauer M, Wenners A, Jonat W, Klapper W (2013) Transcription factors associated with epithelial–mesenchymal transition and cancer stem cells in the tumor centre and margin of invasive breast cancer. Exp Mol Pathol 94:168–173. doi:10.​1016/​j.​yexmp.​2012.​09.​003 CrossRefPubMed
3.
go back to reference Chakravarty G, Moroz K, Makridakis NM, Lloyd SA, Galvez SE, Canavello PR, Lacey MR, Agrawal K, Mondal D (2011) Prognostic significance of cytoplasmic SOX9 in invasive ductal carcinoma and metastatic breast cancer. Exp Biol Med (Maywood) 236:145–155. doi:10.1258/ebm.2010.010086236/2/145 CrossRef Chakravarty G, Moroz K, Makridakis NM, Lloyd SA, Galvez SE, Canavello PR, Lacey MR, Agrawal K, Mondal D (2011) Prognostic significance of cytoplasmic SOX9 in invasive ductal carcinoma and metastatic breast cancer. Exp Biol Med (Maywood) 236:145–155. doi:10.​1258/​ebm.​2010.​010086236/​2/​145 CrossRef
4.
go back to reference Cheang MC, Voduc D, Bajdik C, Leung S, McKinney S, Chia SK, Perou CM, Nielsen TO (2008) Basal-like breast cancer defined by five biomarkers has superior prognostic value than triple-negative phenotype. Clin Cancer Res 14:1368–1376. doi:10.1158/1078-0432.CCR-07-1658 CrossRefPubMed Cheang MC, Voduc D, Bajdik C, Leung S, McKinney S, Chia SK, Perou CM, Nielsen TO (2008) Basal-like breast cancer defined by five biomarkers has superior prognostic value than triple-negative phenotype. Clin Cancer Res 14:1368–1376. doi:10.​1158/​1078-0432.​CCR-07-1658 CrossRefPubMed
7.
go back to reference Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, Senn HJ, Panel Members (2013) Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol 24:2206–2223. doi:10.1093/annonc/mdt303 PubMedCentralCrossRefPubMed Goldhirsch A, Winer EP, Coates AS, Gelber RD, Piccart-Gebhart M, Thurlimann B, Senn HJ, Panel Members (2013) Personalizing the treatment of women with early breast cancer: highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann Oncol 24:2206–2223. doi:10.​1093/​annonc/​mdt303 PubMedCentralCrossRefPubMed
8.
go back to reference Granados-Principal S, Liu Y, Guevara ML, Blanco E, Choi DS, Qian W, Patel T, Rodriguez AA, Cusimano J, Weiss HL, Zhao H, Landis MD, Dave B, Gross SS, Chang JC (2015) Inhibition of iNOS as a novel effective targeted therapy against triple-negative breast cancer. Breast Cancer Res 17:25. doi:10.1186/s13058-015-0527-x PubMedCentralCrossRefPubMed Granados-Principal S, Liu Y, Guevara ML, Blanco E, Choi DS, Qian W, Patel T, Rodriguez AA, Cusimano J, Weiss HL, Zhao H, Landis MD, Dave B, Gross SS, Chang JC (2015) Inhibition of iNOS as a novel effective targeted therapy against triple-negative breast cancer. Breast Cancer Res 17:25. doi:10.​1186/​s13058-015-0527-x PubMedCentralCrossRefPubMed
10.
go back to reference Gupta P, Srivastava SK (2014) Inhibition of Integrin-HER2 signaling by Cucurbitacin B leads to in vitro and in vivo breast tumor growth suppression. Oncotarget 5:1812–1828PubMedCentralCrossRefPubMed Gupta P, Srivastava SK (2014) Inhibition of Integrin-HER2 signaling by Cucurbitacin B leads to in vitro and in vivo breast tumor growth suppression. Oncotarget 5:1812–1828PubMedCentralCrossRefPubMed
11.
go back to reference Hugo HJ, Kokkinos MI, Blick T, Ackland ML, Thompson EW, Newgreen DF (2011) Defining the E-cadherin repressor interactome in epithelial–mesenchymal transition: the PMC42 model as a case study. Cells Tissues Organs 193:23–40. doi:10.1159/000320174 CrossRefPubMed Hugo HJ, Kokkinos MI, Blick T, Ackland ML, Thompson EW, Newgreen DF (2011) Defining the E-cadherin repressor interactome in epithelial–mesenchymal transition: the PMC42 model as a case study. Cells Tissues Organs 193:23–40. doi:10.​1159/​000320174 CrossRefPubMed
12.
go back to reference Ito M, Shien T, Omori M, Mizoo T, Iwamoto T, Nogami T, Motoki T, Taira N, Doihara H, Miyoshi S (2015) Evaluation of aldehyde dehydrogenase 1 and transcription factors in both primary breast cancer and axillary lymph node metastases as a prognostic factor. Breast Cancer. doi:10.1007/s12282-015-0583-1 Ito M, Shien T, Omori M, Mizoo T, Iwamoto T, Nogami T, Motoki T, Taira N, Doihara H, Miyoshi S (2015) Evaluation of aldehyde dehydrogenase 1 and transcription factors in both primary breast cancer and axillary lymph node metastases as a prognostic factor. Breast Cancer. doi:10.​1007/​s12282-015-0583-1
14.
go back to reference Kononen J, Bubendorf L, Kallioniemi A, Bärlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP (1998) Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 4:844–847CrossRefPubMed Kononen J, Bubendorf L, Kallioniemi A, Bärlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP (1998) Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med 4:844–847CrossRefPubMed
15.
go back to reference Li Y, Wu Y, Abbatiello TC, Wu WL, Kim JR, Sarkissyan M, Sarkissyan S, Chung SS, Elshimali Y, Vadgama JV (2015) Slug contributes to cancer progression by direct regulation of ERalpha signaling pathway. Int J Oncol 46:1461–1472. doi:10.3892/ijo.2015.2878 PubMedCentralPubMed Li Y, Wu Y, Abbatiello TC, Wu WL, Kim JR, Sarkissyan M, Sarkissyan S, Chung SS, Elshimali Y, Vadgama JV (2015) Slug contributes to cancer progression by direct regulation of ERalpha signaling pathway. Int J Oncol 46:1461–1472. doi:10.​3892/​ijo.​2015.​2878 PubMedCentralPubMed
17.
18.
go back to reference Mego M, Mani SA, Lee BN, Li C, Evans KW, Cohen EN, Gao H, Jackson SA, Giordano A, Hortobagyi GN, Cristofanilli M, Lucci A, Reuben JM (2012) Expression of epithelial–mesenchymal transition-inducing transcription factors in primary breast cancer: the effect of neoadjuvant therapy. Int J Cancer 130:808–816. doi:10.1002/ijc.26037 PubMedCentralCrossRefPubMed Mego M, Mani SA, Lee BN, Li C, Evans KW, Cohen EN, Gao H, Jackson SA, Giordano A, Hortobagyi GN, Cristofanilli M, Lucci A, Reuben JM (2012) Expression of epithelial–mesenchymal transition-inducing transcription factors in primary breast cancer: the effect of neoadjuvant therapy. Int J Cancer 130:808–816. doi:10.​1002/​ijc.​26037 PubMedCentralCrossRefPubMed
23.
24.
go back to reference Theurillat JP, Zürrer-Härdi U, Varga Z, Storz M, Probst-Hensch NM, Seifert B, Fehr MK, Fink D, Ferrone S, Pestalozzi B, Jungbluth AA, Chen YT, Jäger D, Knuth A, Moch H (2007) NY-BR-1 protein expression in breast carcinoma: a mammary gland differentiation antigen as target for cancer immunotherapy. Cancer Immunol Immunother 56:1723–1731. doi:10.1007/s00262-007-0316-1 CrossRefPubMed Theurillat JP, Zürrer-Härdi U, Varga Z, Storz M, Probst-Hensch NM, Seifert B, Fehr MK, Fink D, Ferrone S, Pestalozzi B, Jungbluth AA, Chen YT, Jäger D, Knuth A, Moch H (2007) NY-BR-1 protein expression in breast carcinoma: a mammary gland differentiation antigen as target for cancer immunotherapy. Cancer Immunol Immunother 56:1723–1731. doi:10.​1007/​s00262-007-0316-1 CrossRefPubMed
27.
Metadata
Title
Differential expression of epithelial–mesenchymal transition and stem cell markers in intrinsic subtypes of breast cancer
Authors
Victoria Pomp
Cornelia Leo
Andrea Mauracher
Dimitri Korol
Wenjun Guo
Zsuzsanna Varga
Publication date
01-11-2015
Publisher
Springer US
Published in
Breast Cancer Research and Treatment / Issue 1/2015
Print ISSN: 0167-6806
Electronic ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-015-3598-6

Other articles of this Issue 1/2015

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

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

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

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