Skip to main content
Top
Published in: Breast Cancer Research 1/2019

Open Access 01-12-2019 | Breast Cancer | Research article

Epigenetic silencing of TGFBI confers resistance to trastuzumab in human breast cancer

Authors: Sònia Palomeras, Ángel Diaz-Lagares, Gemma Viñas, Fernando Setien, Humberto J. Ferreira, Glòria Oliveras, Ana B. Crujeiras, Alejandro Hernández, David H. Lum, Alana L. Welm, Manel Esteller, Teresa Puig

Published in: Breast Cancer Research | Issue 1/2019

Login to get access

Abstract

Background

Acquired resistance to trastuzumab is a major clinical problem in the treatment of HER2-positive (HER2+) breast cancer patients. The selection of trastuzumab-resistant patients is a great challenge of precision oncology. The aim of this study was to identify novel epigenetic biomarkers associated to trastuzumab resistance in HER2+ BC patients.

Methods

We performed a genome-wide DNA methylation (450K array) and a transcriptomic analysis (RNA-Seq) comparing trastuzumab-sensitive (SK) and trastuzumab-resistant (SKTR) HER2+ human breast cancer cell models. The methylation and expression levels of candidate genes were validated by bisulfite pyrosequencing and qRT-PCR, respectively. Functional assays were conducted in the SK and SKTR models by gene silencing and overexpression. Methylation analysis in 24 HER2+ human BC samples with complete response or non-response to trastuzumab-based treatment was conducted by bisulfite pyrosequencing.

Results

Epigenomic and transcriptomic analysis revealed the consistent hypermethylation and downregulation of TGFBI, CXCL2, and SLC38A1 genes in association with trastuzumab resistance. The DNA methylation and expression levels of these genes were validated in both sensitive and resistant models analyzed. Of the genes, TGFBI presented the highest hypermethylation-associated silencing both at the transcriptional and protein level. Ectopic expression of TGFBI in the SKTR model suggest an increased sensitivity to trastuzumab treatment. In primary tumors, TGFBI hypermethylation was significantly associated with trastuzumab resistance in HER2+ breast cancer patients.

Conclusions

Our results suggest for the first time an association between the epigenetic silencing of TGFBI by DNA methylation and trastuzumab resistance in HER2+ cell models. These results provide the basis for further clinical studies to validate the hypermethylation of TGFBI promoter as a biomarker of trastuzumab resistance in HER2+ breast cancer patients.
Appendix
Available only for authorised users
Literature
1.
go back to reference Global Burden of Disease Cancer Collaboration, Fitzmaurice C, Allen C, Barber RM, Barregard L, Bhutta ZA, et al. Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 32 cancer groups, 1990 to 2015: a systematic analysis for the global burden of disease study. JAMA Oncol. 2017;3(4):524–48.PubMedCentralCrossRef Global Burden of Disease Cancer Collaboration, Fitzmaurice C, Allen C, Barber RM, Barregard L, Bhutta ZA, et al. Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 32 cancer groups, 1990 to 2015: a systematic analysis for the global burden of disease study. JAMA Oncol. 2017;3(4):524–48.PubMedCentralCrossRef
2.
go back to reference Bauer K, Parise C, Caggiano V. Use of ER/PR/HER2 subtypes in conjunction with the 2007 St Gallen consensus statement for early breast cancer. BMC Cancer. 2010;10(1):228.PubMedPubMedCentralCrossRef Bauer K, Parise C, Caggiano V. Use of ER/PR/HER2 subtypes in conjunction with the 2007 St Gallen consensus statement for early breast cancer. BMC Cancer. 2010;10(1):228.PubMedPubMedCentralCrossRef
3.
go back to reference Hudis CA. Trastuzumab--mechanism of action and use in clinical practice. N Engl J Med. 2007;357(1):39–51.PubMedCrossRef Hudis CA. Trastuzumab--mechanism of action and use in clinical practice. N Engl J Med. 2007;357(1):39–51.PubMedCrossRef
4.
go back to reference Luque-Cabal M, García-Teijido P, Fernández-Pérez Y, Sánchez-Lorenzo L, Palacio-Vázquez I. Mechanisms behind the resistance to trastuzumab in HER2-amplified breast cancer and strategies to overcome it. Clin Med Insights Oncol. 2016;10(Suppl 1):21–30.PubMedPubMedCentral Luque-Cabal M, García-Teijido P, Fernández-Pérez Y, Sánchez-Lorenzo L, Palacio-Vázquez I. Mechanisms behind the resistance to trastuzumab in HER2-amplified breast cancer and strategies to overcome it. Clin Med Insights Oncol. 2016;10(Suppl 1):21–30.PubMedPubMedCentral
5.
go back to reference Nakashoji A, Hayashida T, Yokoe T, Maeda H, Toyota T, Kikuchi M, et al. The updated network meta-analysis of neoadjuvant therapy for HER2-positive breast cancer. Cancer Treat Rev. 2018;62:9–17.PubMedCrossRef Nakashoji A, Hayashida T, Yokoe T, Maeda H, Toyota T, Kikuchi M, et al. The updated network meta-analysis of neoadjuvant therapy for HER2-positive breast cancer. Cancer Treat Rev. 2018;62:9–17.PubMedCrossRef
6.
go back to reference Carter P, Presta L, Gorman CM, Ridgway JB, Henner D, Wong WL, et al. Humanization of an anti-p185HER2 antibody for human cancer therapy. Proc Natl Acad Sci U S A. 1992;89(10):4285–9.PubMedPubMedCentralCrossRef Carter P, Presta L, Gorman CM, Ridgway JB, Henner D, Wong WL, et al. Humanization of an anti-p185HER2 antibody for human cancer therapy. Proc Natl Acad Sci U S A. 1992;89(10):4285–9.PubMedPubMedCentralCrossRef
7.
go back to reference Nahta R, Yu D, Hung M-C, Hortobagyi GN, Esteva FJ. Mechanisms of disease: understanding resistance to HER2-targeted therapy in human breast cancer. Nat Clin Pract Oncol. 2006;3(5):269–80.PubMedCrossRef Nahta R, Yu D, Hung M-C, Hortobagyi GN, Esteva FJ. Mechanisms of disease: understanding resistance to HER2-targeted therapy in human breast cancer. Nat Clin Pract Oncol. 2006;3(5):269–80.PubMedCrossRef
8.
go back to reference Scaltriti M, Rojo F, Ocaña A, Anido J, Guzman M, Cortes J, et al. Expression of p95HER2, a truncated form of the HER2 receptor, and response to anti-HER2 therapies in breast cancer. J Natl Cancer Inst. 2007;99(8):628–38.PubMedCrossRef Scaltriti M, Rojo F, Ocaña A, Anido J, Guzman M, Cortes J, et al. Expression of p95HER2, a truncated form of the HER2 receptor, and response to anti-HER2 therapies in breast cancer. J Natl Cancer Inst. 2007;99(8):628–38.PubMedCrossRef
10.
go back to reference Paluszczak J, Baer-Dubowska W. Epigenetic diagnostics of cancer — the application of DNA methylation markers. J Appl Genet. 2006;47(4):365–75.PubMedCrossRef Paluszczak J, Baer-Dubowska W. Epigenetic diagnostics of cancer — the application of DNA methylation markers. J Appl Genet. 2006;47(4):365–75.PubMedCrossRef
11.
go back to reference Duffy MJ, Napieralski R, Martens JWM, Span PN, Spyratos F, Sweep FCGJ, et al. Methylated genes as new cancer biomarkers. Eur J Cancer. 2009;45(3):335–46.PubMedCrossRef Duffy MJ, Napieralski R, Martens JWM, Span PN, Spyratos F, Sweep FCGJ, et al. Methylated genes as new cancer biomarkers. Eur J Cancer. 2009;45(3):335–46.PubMedCrossRef
12.
go back to reference Rice JC, Ozcelik H, Maxeiner P, Andrulis I, Futscher BW. Methylation of the BRCA1 promoter is associated with decreased BRCA1 mRNA levels in clinical breast cancer specimens. Carcinogenesis. 2000;21(9):1761–5.PubMedCrossRef Rice JC, Ozcelik H, Maxeiner P, Andrulis I, Futscher BW. Methylation of the BRCA1 promoter is associated with decreased BRCA1 mRNA levels in clinical breast cancer specimens. Carcinogenesis. 2000;21(9):1761–5.PubMedCrossRef
13.
go back to reference Dammann R, Schagdarsurengin U, Strunnikova M, Rastetter M, Seidel C, Liu L, et al. Epigenetic inactivation of the Ras-association domain family 1 (RASSF1A) gene and its function in human carcinogenesis. Histol Histopathol. 2003;18(2):665–77.PubMed Dammann R, Schagdarsurengin U, Strunnikova M, Rastetter M, Seidel C, Liu L, et al. Epigenetic inactivation of the Ras-association domain family 1 (RASSF1A) gene and its function in human carcinogenesis. Histol Histopathol. 2003;18(2):665–77.PubMed
14.
go back to reference Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;13(7):484–92.PubMedCrossRef Jones PA. Functions of DNA methylation: islands, start sites, gene bodies and beyond. Nat Rev Genet. 2012;13(7):484–92.PubMedCrossRef
15.
go back to reference Trempe GL. Human breast cancer in culture. Recent Results Cancer Res. 1976;57(57):33–41. Trempe GL. Human breast cancer in culture. Recent Results Cancer Res. 1976;57(57):33–41.
16.
go back to reference Blancafort A, Giró-Perafita A, Oliveras G, Palomeras S, Turrado C, Campuzano Ò, et al. Dual fatty acid synthase and HER2 signaling blockade shows marked antitumor activity against breast cancer models resistant to anti-HER2 drugs. PLoS One. 2015;10(6):e0131241.PubMedPubMedCentralCrossRef Blancafort A, Giró-Perafita A, Oliveras G, Palomeras S, Turrado C, Campuzano Ò, et al. Dual fatty acid synthase and HER2 signaling blockade shows marked antitumor activity against breast cancer models resistant to anti-HER2 drugs. PLoS One. 2015;10(6):e0131241.PubMedPubMedCentralCrossRef
17.
go back to reference Puig T, Aguilar H, Cufí S, Oliveras G, Turrado C, Ortega-gutiérrez S, et al. A novel inhibitor of fatty acid synthase shows activity against HER2 + breast cancer xenografts and is active in anti-HER2 drug-resistant cell lines. Breast Cancer Res. 2011;13(6):R131.PubMedPubMedCentralCrossRef Puig T, Aguilar H, Cufí S, Oliveras G, Turrado C, Ortega-gutiérrez S, et al. A novel inhibitor of fatty acid synthase shows activity against HER2 + breast cancer xenografts and is active in anti-HER2 drug-resistant cell lines. Breast Cancer Res. 2011;13(6):R131.PubMedPubMedCentralCrossRef
18.
go back to reference Edge SB, Compton CC. The American Joint Committee on Cancer: the 7th edition of the AJCC Cancer staging manual and the future of TNM. Ann Surg Oncol. 2010;17(6):1471–4.CrossRefPubMed Edge SB, Compton CC. The American Joint Committee on Cancer: the 7th edition of the AJCC Cancer staging manual and the future of TNM. Ann Surg Oncol. 2010;17(6):1471–4.CrossRefPubMed
19.
go back to reference Bender CM, Pao MM, Jones PA. Inhibition of DNA methylation by 5-aza-2′-deoxycytidine suppresses the growth of human tumor cell lines. Cancer Res. 1998;58(1):95–101.PubMed Bender CM, Pao MM, Jones PA. Inhibition of DNA methylation by 5-aza-2′-deoxycytidine suppresses the growth of human tumor cell lines. Cancer Res. 1998;58(1):95–101.PubMed
20.
go back to reference Sandoval J, Heyn H, Moran S, Serra-Musach J, Pujana MA, Bibikova M, et al. Validation of a DNA methylation microarray for 450,000 CpG sites in the human genome. Epigenetics. 2011;6(6):692–702.CrossRefPubMed Sandoval J, Heyn H, Moran S, Serra-Musach J, Pujana MA, Bibikova M, et al. Validation of a DNA methylation microarray for 450,000 CpG sites in the human genome. Epigenetics. 2011;6(6):692–702.CrossRefPubMed
21.
go back to reference Naumov VA, Generozov EV, Zaharjevskaya NB, Matushkina DS, Larin AK, Chernyshov SV, et al. Genome-scale analysis of DNA methylation in colorectal cancer using Infinium HumanMethylation450 BeadChips. Epigenetics. 2013;8(9):921–34.PubMedPubMedCentralCrossRef Naumov VA, Generozov EV, Zaharjevskaya NB, Matushkina DS, Larin AK, Chernyshov SV, et al. Genome-scale analysis of DNA methylation in colorectal cancer using Infinium HumanMethylation450 BeadChips. Epigenetics. 2013;8(9):921–34.PubMedPubMedCentralCrossRef
22.
go back to reference Ween MP, Oehler MK, Ricciardelli C. Transforming growth factor-beta-induced protein (TGFBI)/(βig-H3): a matrix protein with dual functions in ovarian cancer. Int J Mol Sci. 2012;13(8):10461–77.PubMedPubMedCentralCrossRef Ween MP, Oehler MK, Ricciardelli C. Transforming growth factor-beta-induced protein (TGFBI)/(βig-H3): a matrix protein with dual functions in ovarian cancer. Int J Mol Sci. 2012;13(8):10461–77.PubMedPubMedCentralCrossRef
23.
go back to reference Diaz-Lagares A, Crujeiras AB, Lopez-Serra P, Soler M, Setien F, Goyal A, et al. Epigenetic inactivation of the p53-induced long noncoding RNA TP53 target 1 in human cancer. Proc Natl Acad Sci. 2016;113(47):E7535–44.PubMedCrossRefPubMedCentral Diaz-Lagares A, Crujeiras AB, Lopez-Serra P, Soler M, Setien F, Goyal A, et al. Epigenetic inactivation of the p53-induced long noncoding RNA TP53 target 1 in human cancer. Proc Natl Acad Sci. 2016;113(47):E7535–44.PubMedCrossRefPubMedCentral
24.
go back to reference Diaz-Lagares A, Mendez-Gonzalez J, Hervas D, Saigi M, Pajares MJ, Garcia D, et al. A novel epigenetic signature for early diagnosis in lung cancer. Clin Cancer Res. 2016;22(13):3361–71.PubMedCrossRef Diaz-Lagares A, Mendez-Gonzalez J, Hervas D, Saigi M, Pajares MJ, Garcia D, et al. A novel epigenetic signature for early diagnosis in lung cancer. Clin Cancer Res. 2016;22(13):3361–71.PubMedCrossRef
25.
go back to reference Wen G, Partridge MA, Li B, Hong M, Liao W, Cheng SK, et al. TGFBI expression reduces in vitro and in vivo metastatic potential of lung and breast tumor cells. Cancer Lett. 2011;308(1):23–32.PubMedPubMedCentralCrossRef Wen G, Partridge MA, Li B, Hong M, Liao W, Cheng SK, et al. TGFBI expression reduces in vitro and in vivo metastatic potential of lung and breast tumor cells. Cancer Lett. 2011;308(1):23–32.PubMedPubMedCentralCrossRef
26.
go back to reference Kronski E, Fiori ME, Barbieri O, Astigiano S, Mirisola V, Killian PH, et al. miR181b is induced by the chemopreventive polyphenol curcumin and inhibits breast cancer metastasis via down-regulation of the inflammatory cytokines CXCL1 and −2. Mol Oncol. 2014;8(3):581–95.PubMedPubMedCentralCrossRef Kronski E, Fiori ME, Barbieri O, Astigiano S, Mirisola V, Killian PH, et al. miR181b is induced by the chemopreventive polyphenol curcumin and inhibits breast cancer metastasis via down-regulation of the inflammatory cytokines CXCL1 and −2. Mol Oncol. 2014;8(3):581–95.PubMedPubMedCentralCrossRef
27.
go back to reference Wang K, Cao F, Fang W, Hu Y, Chen Y, Ding H, et al. Activation of SNAT1/SLC38A1 in human breast cancer: correlation with p-Akt overexpression. BMC Cancer. 2013;13:343.PubMedPubMedCentralCrossRef Wang K, Cao F, Fang W, Hu Y, Chen Y, Ding H, et al. Activation of SNAT1/SLC38A1 in human breast cancer: correlation with p-Akt overexpression. BMC Cancer. 2013;13:343.PubMedPubMedCentralCrossRef
28.
go back to reference Dejeux E, El Abdalaoui H, Gut IG, Tost J. Identification and quantification of differentially methylated loci by the PyrosequencingTM technology. Methods Mol Biol. 2009;507:189–205.PubMedCrossRef Dejeux E, El Abdalaoui H, Gut IG, Tost J. Identification and quantification of differentially methylated loci by the PyrosequencingTM technology. Methods Mol Biol. 2009;507:189–205.PubMedCrossRef
29.
go back to reference Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands (DNA methylation/tumor suppressor genes/pl6/p15). Proc Natl Acad Sci U S A. 1996;93:9821–6.PubMedPubMedCentralCrossRef Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB. Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands (DNA methylation/tumor suppressor genes/pl6/p15). Proc Natl Acad Sci U S A. 1996;93:9821–6.PubMedPubMedCentralCrossRef
30.
go back to reference Hashimoto K, Noshiro M, Ohno S, Kawamoto T, Satakeda H, Akagawa Y, et al. Characterization of a cartilage-derived 66-kDa protein (RGD-CAP/βig-h3) that binds to collagen. Biochim Biophys Acta - Mol Cell Res. 1997;1355(3):303–14.CrossRef Hashimoto K, Noshiro M, Ohno S, Kawamoto T, Satakeda H, Akagawa Y, et al. Characterization of a cartilage-derived 66-kDa protein (RGD-CAP/βig-h3) that binds to collagen. Biochim Biophys Acta - Mol Cell Res. 1997;1355(3):303–14.CrossRef
31.
go back to reference Kim J-E, Jeong H-W, Nam J-O, Lee B-H, Choi J-Y, Park R-W, et al. Identification of motifs in the fasciclin domains of the transforming growth factor-β-induced matrix protein βig-h3 that interact with the αvβ5 integrin. J Biol Chem. 2002;277(48):46159–65.PubMedCrossRef Kim J-E, Jeong H-W, Nam J-O, Lee B-H, Choi J-Y, Park R-W, et al. Identification of motifs in the fasciclin domains of the transforming growth factor-β-induced matrix protein βig-h3 that interact with the αvβ5 integrin. J Biol Chem. 2002;277(48):46159–65.PubMedCrossRef
32.
go back to reference Billings PC, Charles Whitbeck J, Adams CS, Abrams WR, Cohen AJ, Engelsberg BN, et al. The transforming growth factor-β-inducible matrix protein βig-h3 interacts with fibronectin. J Biol Chem. 2002;277(31):28003–9.PubMedCrossRef Billings PC, Charles Whitbeck J, Adams CS, Abrams WR, Cohen AJ, Engelsberg BN, et al. The transforming growth factor-β-inducible matrix protein βig-h3 interacts with fibronectin. J Biol Chem. 2002;277(31):28003–9.PubMedCrossRef
33.
go back to reference Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344(11):783–92.PubMedCrossRef Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344(11):783–92.PubMedCrossRef
34.
go back to reference Nahta R, Yuan LXHH, Zhang B, Kobayashi R, Esteva FJ. Insulin-like growth factor-I receptor/human epidermal growth factor receptor 2 heterodimerization contributes to trastuzumab resistance of breast cancer cells. Cancer Res. 2005;65(23):11118–28.PubMedCrossRef Nahta R, Yuan LXHH, Zhang B, Kobayashi R, Esteva FJ. Insulin-like growth factor-I receptor/human epidermal growth factor receptor 2 heterodimerization contributes to trastuzumab resistance of breast cancer cells. Cancer Res. 2005;65(23):11118–28.PubMedCrossRef
35.
go back to reference Neve RM, Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell. 2006;10(6):515–27.PubMedPubMedCentralCrossRef Neve RM, Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell. 2006;10(6):515–27.PubMedPubMedCentralCrossRef
36.
go back to reference Weigelt B, Lo AT, Park CC, Gray JW, Bissell MJ. HER2 signaling pathway activation and response of breast cancer cells to HER2-targeting agents is dependent strongly on the 3D microenvironment. Breast Cancer Res Treat. 2010;122(1):35–43.PubMedCrossRef Weigelt B, Lo AT, Park CC, Gray JW, Bissell MJ. HER2 signaling pathway activation and response of breast cancer cells to HER2-targeting agents is dependent strongly on the 3D microenvironment. Breast Cancer Res Treat. 2010;122(1):35–43.PubMedCrossRef
37.
go back to reference Shao G, Berenguer J, Borczuk AC, Powell CA, Hei TK, Zhao Y. Epigenetic inactivation of Betaig-h3 gene in human cancer cells. Cancer Res. 2006;66(9):4566–73.PubMedCrossRef Shao G, Berenguer J, Borczuk AC, Powell CA, Hei TK, Zhao Y. Epigenetic inactivation of Betaig-h3 gene in human cancer cells. Cancer Res. 2006;66(9):4566–73.PubMedCrossRef
38.
go back to reference Wang N, Zhang H, Yao Q, Wang Y, Dai S, Yang X. TGFBI promoter hypermethylation correlating with paclitaxel chemoresistance in ovarian cancer. J Exp Clin Cancer Res. 2012;31(1):6.PubMedPubMedCentralCrossRef Wang N, Zhang H, Yao Q, Wang Y, Dai S, Yang X. TGFBI promoter hypermethylation correlating with paclitaxel chemoresistance in ovarian cancer. J Exp Clin Cancer Res. 2012;31(1):6.PubMedPubMedCentralCrossRef
39.
go back to reference Skonier J, Neubauer M, Madisen L, Bennett K, Plowman GD, Purchio AF. cDNA cloning and sequence analysis of βig-h3, a novel gene induced in a human adenocarcinoma cell line after treatment with transforming growth factor-β. DNA Cell Biol. 1992;11(7):511–22.PubMedCrossRef Skonier J, Neubauer M, Madisen L, Bennett K, Plowman GD, Purchio AF. cDNA cloning and sequence analysis of βig-h3, a novel gene induced in a human adenocarcinoma cell line after treatment with transforming growth factor-β. DNA Cell Biol. 1992;11(7):511–22.PubMedCrossRef
40.
go back to reference Calaf GM, Echiburú-Chau C, Zhao YL, Hei TK. BigH3 protein expression as a marker for breast cancer. Int J Mol Med. 2008;21(5):561–8.PubMed Calaf GM, Echiburú-Chau C, Zhao YL, Hei TK. BigH3 protein expression as a marker for breast cancer. Int J Mol Med. 2008;21(5):561–8.PubMed
41.
go back to reference Shah JN, Shao G, Hei TK, Zhao Y. Methylation screening of the TGFBI promoter in human lung and prostate cancer by methylation-specific PCR. BMC Cancer. 2008;8(1):284.PubMedPubMedCentralCrossRef Shah JN, Shao G, Hei TK, Zhao Y. Methylation screening of the TGFBI promoter in human lung and prostate cancer by methylation-specific PCR. BMC Cancer. 2008;8(1):284.PubMedPubMedCentralCrossRef
42.
go back to reference Ahmed AA, Mills AD, Ibrahim AEKK, Temple J, Blenkiron C, Vias M, et al. The extracellular matrix protein TGFBI induces microtubule stabilization and sensitizes ovarian cancers to paclitaxel. Cancer Cell. 2007;12(6):514–27.PubMedPubMedCentralCrossRef Ahmed AA, Mills AD, Ibrahim AEKK, Temple J, Blenkiron C, Vias M, et al. The extracellular matrix protein TGFBI induces microtubule stabilization and sensitizes ovarian cancers to paclitaxel. Cancer Cell. 2007;12(6):514–27.PubMedPubMedCentralCrossRef
43.
go back to reference Kitahara O, Furukawa Y, Tanaka T, Kihara C, Ono K, Yanagawa R, et al. Alterations of gene expression during colorectal carcinogenesis revealed by cDNA microarrays after laser-capture microdissection of tumor tissues and normal epithelia. Cancer Res. 2001;61(9):3544–9.PubMed Kitahara O, Furukawa Y, Tanaka T, Kihara C, Ono K, Yanagawa R, et al. Alterations of gene expression during colorectal carcinogenesis revealed by cDNA microarrays after laser-capture microdissection of tumor tissues and normal epithelia. Cancer Res. 2001;61(9):3544–9.PubMed
44.
go back to reference Schneider D, Kleeff J, Berberat PO, Zhu Z, Korc M, Friess H, et al. Induction and expression of betaig-h3 in pancreatic cancer cells. Biochim Biophys Acta. 2002;1588(1):1–6.PubMedCrossRef Schneider D, Kleeff J, Berberat PO, Zhu Z, Korc M, Friess H, et al. Induction and expression of betaig-h3 in pancreatic cancer cells. Biochim Biophys Acta. 2002;1588(1):1–6.PubMedCrossRef
45.
go back to reference Guo S-K, Shen M-F, Yao H-W, Liu Y-S. Enhanced expression of TGFBI promotes the proliferation and migration of glioma cells. Cell Physiol Biochem. 2018;49(3):1097–109.PubMedCrossRef Guo S-K, Shen M-F, Yao H-W, Liu Y-S. Enhanced expression of TGFBI promotes the proliferation and migration of glioma cells. Cell Physiol Biochem. 2018;49(3):1097–109.PubMedCrossRef
46.
go back to reference Chen W-Y, Tsai Y-C, Yeh H-L, Suau F, Jiang K-C, Shao A-N, et al. Loss of SPDEF and gain of TGFBI activity after androgen deprivation therapy promote EMT and bone metastasis of prostate cancer. Sci Signal. 2017;10(492):eaam6826.PubMedCrossRef Chen W-Y, Tsai Y-C, Yeh H-L, Suau F, Jiang K-C, Shao A-N, et al. Loss of SPDEF and gain of TGFBI activity after androgen deprivation therapy promote EMT and bone metastasis of prostate cancer. Sci Signal. 2017;10(492):eaam6826.PubMedCrossRef
47.
48.
go back to reference Soung YH, Clifford JL, Chung J. Crosstalk between integrin and receptor tyrosine kinase signaling in breast carcinoma progression. BMB Rep. 2010;43(5):311–8.PubMedCrossRef Soung YH, Clifford JL, Chung J. Crosstalk between integrin and receptor tyrosine kinase signaling in breast carcinoma progression. BMB Rep. 2010;43(5):311–8.PubMedCrossRef
49.
go back to reference Lesniak D, Xu Y, Deschenes J, Lai R, Thoms J, Murray D, et al. 1-integrin circumvents the antiproliferative effects of trastuzumab in human epidermal growth factor receptor-2-positive breast cancer. Cancer Res. 2009;69(22):8620–8.PubMedCrossRef Lesniak D, Xu Y, Deschenes J, Lai R, Thoms J, Murray D, et al. 1-integrin circumvents the antiproliferative effects of trastuzumab in human epidermal growth factor receptor-2-positive breast cancer. Cancer Res. 2009;69(22):8620–8.PubMedCrossRef
50.
go back to reference Guo W, Pylayeva Y, Pepe A, Yoshioka T, Muller WJ, Inghirami G, et al. β4 integrin amplifies ErbB2 signaling to promote mammary tumorigenesis. Cell. 2006;126(3):489–502.PubMedCrossRef Guo W, Pylayeva Y, Pepe A, Yoshioka T, Muller WJ, Inghirami G, et al. β4 integrin amplifies ErbB2 signaling to promote mammary tumorigenesis. Cell. 2006;126(3):489–502.PubMedCrossRef
51.
go back to reference Nam J-O, Jeong H-W, Lee B-H, Park R-W, Kim I-S. Regulation of tumor angiogenesis by fastatin, the fourth FAS1 domain of βig-h3, via αvβ3 integrin. Cancer Res. 2005;65(10):4153–61.PubMedCrossRef Nam J-O, Jeong H-W, Lee B-H, Park R-W, Kim I-S. Regulation of tumor angiogenesis by fastatin, the fourth FAS1 domain of βig-h3, via αvβ3 integrin. Cancer Res. 2005;65(10):4153–61.PubMedCrossRef
52.
go back to reference Zhao YL, Piao CQ, Hei TK. Overexpression of Betaig-h3 gene downregulates integrin alpha5beta1 and suppresses tumorigenicity in radiation-induced tumorigenic human bronchial epithelial cells. Br J Cancer. 2002;86(12):1923–8.PubMedPubMedCentralCrossRef Zhao YL, Piao CQ, Hei TK. Overexpression of Betaig-h3 gene downregulates integrin alpha5beta1 and suppresses tumorigenicity in radiation-induced tumorigenic human bronchial epithelial cells. Br J Cancer. 2002;86(12):1923–8.PubMedPubMedCentralCrossRef
53.
go back to reference Kim J-E, Kim S-J, Jeong H-W, Lee B-H, Choi J-Y, Park R-W, et al. RGD peptides released from βig-h3, a TGF-β-induced cell-adhesive molecule, mediate apoptosis. Oncogene. 2003;22(13):2045–53.PubMedCrossRef Kim J-E, Kim S-J, Jeong H-W, Lee B-H, Choi J-Y, Park R-W, et al. RGD peptides released from βig-h3, a TGF-β-induced cell-adhesive molecule, mediate apoptosis. Oncogene. 2003;22(13):2045–53.PubMedCrossRef
54.
go back to reference Wen G, Hong M, Li B, Liao W, Cheng SK, Hu B, et al. Transforming growth factor-β-induced protein (TGFBI) suppresses mesothelioma progression through the Akt/mTOR pathway. Int J Oncol. 2011;39(4):1001–9.PubMed Wen G, Hong M, Li B, Liao W, Cheng SK, Hu B, et al. Transforming growth factor-β-induced protein (TGFBI) suppresses mesothelioma progression through the Akt/mTOR pathway. Int J Oncol. 2011;39(4):1001–9.PubMed
55.
go back to reference Becker J, Volland S, Noskova I, Schramm A, Schweigerer LL, Wilting J. Keratoepithelin reverts the suppression of tissue factor pathway inhibitor 2 by MYCN in human neuroblastoma: a mechanism to inhibit invasion. Int J Oncol. 2008;32(1):235–40.PubMed Becker J, Volland S, Noskova I, Schramm A, Schweigerer LL, Wilting J. Keratoepithelin reverts the suppression of tissue factor pathway inhibitor 2 by MYCN in human neuroblastoma: a mechanism to inhibit invasion. Int J Oncol. 2008;32(1):235–40.PubMed
56.
go back to reference Bissey P-A, Law JH, Bruce JP, Shi W, Renoult A, Chua MLK, et al. Dysregulation of the MiR-449b target TGFBI alters the TGFβ pathway to induce cisplatin resistance in nasopharyngeal carcinoma. Oncogenesis. 2018;7(5):40.PubMedPubMedCentralCrossRef Bissey P-A, Law JH, Bruce JP, Shi W, Renoult A, Chua MLK, et al. Dysregulation of the MiR-449b target TGFBI alters the TGFβ pathway to induce cisplatin resistance in nasopharyngeal carcinoma. Oncogenesis. 2018;7(5):40.PubMedPubMedCentralCrossRef
57.
go back to reference Beyer I, Li Z, Persson J, Liu Y, van Rensburg R, Yumul R, et al. Controlled extracellular matrix degradation in breast cancer tumors improves therapy by trastuzumab. Mol Ther. 2011;19(3):479–89.PubMedCrossRef Beyer I, Li Z, Persson J, Liu Y, van Rensburg R, Yumul R, et al. Controlled extracellular matrix degradation in breast cancer tumors improves therapy by trastuzumab. Mol Ther. 2011;19(3):479–89.PubMedCrossRef
58.
go back to reference Pajares MJ, Agorreta J, Salvo E, Behrens C, Wistuba II, Montuenga LM, et al. TGFBI expression is an independent predictor of survival in adjuvant-treated lung squamous cell carcinoma patients. Br J Cancer. 2014;110(6):1545–51.PubMedPubMedCentralCrossRef Pajares MJ, Agorreta J, Salvo E, Behrens C, Wistuba II, Montuenga LM, et al. TGFBI expression is an independent predictor of survival in adjuvant-treated lung squamous cell carcinoma patients. Br J Cancer. 2014;110(6):1545–51.PubMedPubMedCentralCrossRef
59.
go back to reference Schettini F, Buono G, Cardalesi C, Desideri I, De Placido S, Del Mastro L. Hormone receptor/human epidermal growth factor receptor 2-positive breast cancer: where we are now and where we are going. Cancer Treat Rev. 2016;46:20–6.PubMedCrossRef Schettini F, Buono G, Cardalesi C, Desideri I, De Placido S, Del Mastro L. Hormone receptor/human epidermal growth factor receptor 2-positive breast cancer: where we are now and where we are going. Cancer Treat Rev. 2016;46:20–6.PubMedCrossRef
60.
go back to reference Gianni L, Pienkowski T, Im Y-H, Tseng L-M, Liu M-C, Lluch A, et al. 5-year analysis of neoadjuvant pertuzumab and trastuzumab in patients with locally advanced, inflammatory, or early-stage HER2-positive breast cancer (NeoSphere): a multicentre, open-label, phase 2 randomised trial. Lancet Oncol. 2016;17(6):791–800.PubMedCrossRef Gianni L, Pienkowski T, Im Y-H, Tseng L-M, Liu M-C, Lluch A, et al. 5-year analysis of neoadjuvant pertuzumab and trastuzumab in patients with locally advanced, inflammatory, or early-stage HER2-positive breast cancer (NeoSphere): a multicentre, open-label, phase 2 randomised trial. Lancet Oncol. 2016;17(6):791–800.PubMedCrossRef
Metadata
Title
Epigenetic silencing of TGFBI confers resistance to trastuzumab in human breast cancer
Authors
Sònia Palomeras
Ángel Diaz-Lagares
Gemma Viñas
Fernando Setien
Humberto J. Ferreira
Glòria Oliveras
Ana B. Crujeiras
Alejandro Hernández
David H. Lum
Alana L. Welm
Manel Esteller
Teresa Puig
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 1/2019
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-019-1160-x

Other articles of this Issue 1/2019

Breast Cancer Research 1/2019 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