miR-221 stimulates breast cancer cells and cancer-associated fibroblasts (CAFs) through selective interference with the A20/c-Rel/CTGF signaling

Maria Francesca Santolla, Rosamaria Lappano, Francesca Cirillo, Damiano Cosimo Rigiracciolo, Anna Sebastiani, Sergio Abonante, Pierfrancesco Tassone, Pierosandro Tagliaferri, Maria Teresa Di Martino, Marcello Maggiolini, Adele Vivacqua, Maria Francesca Santolla, Rosamaria Lappano, Francesca Cirillo, Damiano Cosimo Rigiracciolo, Anna Sebastiani, Sergio Abonante, Pierfrancesco Tassone, Pierosandro Tagliaferri, Maria Teresa Di Martino, Marcello Maggiolini, Adele Vivacqua

Abstract

Background: MicroRNA (miRNAs) are non-coding small RNA molecules that regulate gene expression by inhibiting the translation of target mRNAs. Among several dysregulated miRNAs in human cancer, the up-regulation of miR-221 has been associated with development of a variety of hematologic and solid malignancies. In this study, we investigated the involvement of miR-221 in breast cancer.

Methods: TaqMan microRNA assay was used to detect the miR-221 levels in normal cells and in MDA-MB 231 and SkBr3 breast cancer cells as well as in main players of the tumor microenvironment, namely cancer-associated fibroblasts (CAFs). miR-221 mimic sequence and locked nucleic acid (LNA)-i-miR-221 construct were used to induce or inhibit, respectively, the miR-221 expression in cells used. Quantitative PCR and western blotting analysis were performed to evaluate the levels of the miR-221 target gene A20 (TNFAIP3), as well as the member of the NF-kB complex namely c-Rel and the connective tissue growth factor (CTGF). Chromatin immunoprecipitation (ChIP) assay was performed to ascertain the recruitment of c-Rel to the CTFG promoter. Finally, the cell growth and migration in the presence of LNA-i-miR-221 or silencing c-Rel and CTGF by specific short hairpin were assessed by cell count, colony formation and boyden chambers assays. Statistical analysis was performed by ANOVA.

Results: We first demonstrated that LNA-i-miR-221 inhibits both endogenous and ectopic expression of miR-221 in our experimental models. Next, we found that the A20 down-regulation, as well as the up-regulation of c-Rel induced by miR-221 were no longer evident using LNA-i-miR-221. Moreover, we established that the miR-221 dependent recruitment of c-Rel to the NF-kB binding site located within the CTGF promoter region is prevented by using LNA-i-miR-221. Furthermore, we determined that the up-regulation of CTGF mRNA and protein levels by miR-221 is no longer evident using LNA-i-miR221 and silencing c-Rel. Finally, we assessed that cell growth and migration induced by miR-221 in MDA-MB 231 and SkBr3 breast cancer cells as well as in CAFs are abolished by LNAi-miR-221 and silencing c-Rel or CTGF.

Conclusions: Overall, these data provide novel insights into the stimulatory action of miR-221 in breast cancer cells and CAFs, suggesting that its inhibition may be considered toward targeted therapeutic approaches in breast cancer patients.

Keywords: A20; Breast cancer; C-Rel; CAFs; CTGF; miR-221.

Conflict of interest statement

Ethics approval and consent to participate

Signed informed consent from all the patients was obtained and all samples were collected, identified and used in accordance with approval by the Institutional Ethical Committee Board (Regional Hospital, Cosenza, Italy).

Competing interests

The authors declare that they have no competing interest.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
miR-221 regulates A20 expression in CAFs and breast cancer cells. a Expression of miR-221 in fibroblasts, CAFs, MCF10A, MDA-MB 231 and SkBr3 cells. Raw Ct data were normalized to the housekeeping RNU6 levels and expressed as ΔCt values using the comparative cross threshold (Ct) method. b miR-221 expression in CAFs, MDA-MB 231 and SkBr3 breast cancer cells after transfection for 48 h with 25 nM miR-Ctrl and 25 nM miR-221, alone or in combination with 100 nM LNA-i-miR-221 (LNA-i). c Schematic alignment between the miR-221 sequence and the 3’-UTR mRNA region of A20. d A20 mRNA expression in CAFs, MDA-MB 231 and SkBr3 cells transfected for 48 h with 25 nM miR-Ctrl and 25 nM miR-221, alone or in combination with 100 nM LNA-i-miR-221 (LNA-i). A20 protein expression in CAFs (e), MDA-MB 231 (f) and SkBr3 (g) cells transfected for 48 h with 25 nM miR-Ctrl and 25 nM miR-221, alone or in in combination with 100 nM LNA-i-miR-221 (LNA-i); β-actin serves as a loading control. Below panels show densitometric analysis of the blots normalized to the loading controls. Each column represents the mean ± SD of three independent experiments performed in triplicate. The data are shown as fold induction respect to cells transfected with miR-Ctrl. (*) indicates p < 0.05 and (**) p < 0.01
Fig. 2
Fig. 2
miR-221 prompts the protein expression of c-Rel and its recruitment to the CTGF promoter region. c-Rel protein expression in CAFs (a), MDA-MB 231 (b) and SkBr3 (c) cells transfected for 48 h with 25 nM miR-Ctrl and 25 nM miR-221, alone or in combination with 100 nM LNA-i-miR-221 (LNA-i). β-actin serves as a loading control. Below panels show densitometric analysis of the blots normalized to the loading controls. d Putative NF-kB binding site (capital letters in the rectangle) located within the CTGF promoter sequence (− 625 bp to + 62 bp). The transcriptional start site is indicated as + 1. The position of primers used for ChIP-qPCR analyses is underlined. Recruitment of c-Rel to the NF-kB binding site within the CTGF promoter sequence in CAFs (e), MDA-MB 231 (f) and SkBr3 (g) cells transfected for 48 h with 25 nM miR-Ctrl and 25 nM miR-221, alone or in combination with 100 nM LNA-i-miR-221 (LNA-i). Data were normalized to the Input and reported as fold changes respect to IgG. h Luciferase activity of the CTGF reporter gene in CAFs, MDA-MB 231 and SkBr3 cells transfected for 48 h with 25 nM miR-Ctrl and 25 nM miR-221, alone or in combination with 100 nM LNA-i-miR-221 (LNA-i). i Luciferase activity of the CTGF reporter gene in CAFs, MDA-MB 231 and SkBr3 cells transfected for 8 h with shRNA or shRel and then transfected for 48 h with 25 nM miR-221. The luciferase activity was normalized to the internal transfection control; values of cells receiving scrambled controls were set as 1-fold induction upon which the activity obtained with the indicated effectors was calculated. Each column represents the mean ± SD of three independent experiments performed in triplicate. (**) indicates p < 0.01
Fig. 3
Fig. 3
c-Rel is involved in the up-regulation of the CTGF triggered by miR-221. a CTGF mRNA levels in CAFs, MDA-MB 231 and SkBr3 cells transfected for 48 h with 25 nM miR-Ctrl and 25 nM miR-221, alone or in combination with 100 nM LNA-i-miR-221 (LNA-i). Data are shown as fold changes respect to the scrambled controls. b CTGF mRNA levels in CAFs, MDA-MB 231 and SkBr3 cells transfected for 8 h with shRNA or shRel and then transfected for 48 h with 25 nM miR-221. Data are shown as fold changes respect to the scrambled controls. CTGF protein levels in CAFs (c), MDA-MB 231 (d) and SkBr3 (e) cells transfected for 48 h with 25 nM miR-Ctrl and 25 nM miR-221, alone or in combination with 100 nM LNA-i-miR-221 (LNA-i). β-actin serves as a loading control. Below panels show densitometric analysis of the blots normalized to the loading controls. CTGF protein levels in CAFs (f), MDA-MB 231 (g) and SkBr3 (h) cells transfected for 8 h with shRNA or shRel and then transfected for 48 h with 25 nM miR-221. β-actin serves as a loading control. Below panels show densitometric analysis of the blots normalized to the loading controls. Efficacy of c-Rel silencing in CAFs (i), MDA-MB 231 (j) and SkBr3 (k) cells. β-actin serves as a loading control. Side panels show densitometric analysis of the blots normalized to the loading controls. Results shown are representative of at least three independent experiments. (**) indicates p < 0.01
Fig. 4
Fig. 4
miR-221 induces proliferative effects in CAFs, MDA-MB 231 and SkBr3 cells. Cell proliferation in (a) CAFs, (d) MDA-MB 231 and (g) SkBr3 cells transfected every 2 days with 25 nM miR-Ctrl and 25 nM miR-221, alone or in combination with 100 nM LNA-i-miR-221 (LNA-i) and then counted on day 6. Cell proliferation in CAFs (b) MDA-MB 231 (e) and SkBr3 (h) cells transfected with shRNA or shRel for 8 h and then transfected for 48 h with 25 nM miR-221. Cell proliferation in CAFs (c), MDA-MB 231 (f) and SkBr3 (i) cells transfected with shRNA or shCTGF for 8 h and then transfected for 48 h with 25 nM miR-221. The transfections were renewed every 2 days and cells were counted on day 6. Each data point is the mean ± SD of three independent experiments performed in triplicate. (**) indicates p < 0.01
Fig. 5
Fig. 5
miR-221 promotes colony formation in CAFs, MDA-MB 231 and SkBr3 cells. Colony formation in CAFs (a, b), MDA-MB 231 (c, d) and SkBr3 (e, f) cells transfected every 2 days as indicated, after 10 days of incubation cell colonies were stained and pictures were captured by a digital camera. Colonies were counted using the program WCIF ImageJ for Windows. Each data point is the mean ± SD of three independent experiments performed in triplicate. (**) indicates p < 0.01
Fig. 6
Fig. 6
miR-221 triggers migratory effects in CAFs, MDA-MB 231 and SkBr3 cells. Cell migration as evaluated by Boyden Chamber assay in (a) CAFs, (c) MDA-MB 231 and (e) SkBr3 cells transfected for 48 h with 25 nM miR-Ctrl and 25 nM miR-221, alone or in combination with 100 nM LNA-i-miR-221 (LNA-i). Cell migration, evaluated by Boyden Chamber assay, in CAFs (b) MDA-MB 231 (d) and SkBr3 (f) cells transfected with shRNA or shRel for 8 h and then transfected for 48 h with 25 nM miR-221. Cells were counted in at least 10 random fields at 10× magnification (Scale bar =200 μm) in three independent experiments performed in triplicate. (**) indicates p < 0.01

References

    1. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell. 2009;136:215–233. doi: 10.1016/j.cell.2009.01.002.
    1. Ben-Hamo R, Efroni S. MicroRNA regulation of molecular pathways as a generic mechanism and as a core disease phenotype. Oncotarget. 2015;6:1594–1604. doi: 10.18632/oncotarget.2734.
    1. Wu Z, Huang X, Huang X, Zou Q, Guo Y. The inhibitory role of Mir-29 in growth of breast cancer cells. J Exp Clin Cancer Res. 2013;32:98. doi: 10.1186/1756-9966-32-98.
    1. Wang C, Zheng X, Shen C, Shi Y. MicroRNA-203 suppresses cell proliferation and migration by targeting BIRC5 and LASP1 in human triple-negative breast cancer cells. J Exp Clin Cancer Res. 2012;31:58. doi: 10.1186/1756-9966-31-58.
    1. Paladini L, Fabris L, Bottai G, Raschioni C, Calin GA, Santarpia L. Targeting microRNAs as key modulators of tumor immune response. J Exp Cancer Res. 2016;35:103. doi: 10.1186/s13046-016-0375-2.
    1. Smigal C, Jemal A, Ward E, Cokkinides V, Smith R, Howe HL, et al. Trends in breast cancer by race and ethnicity: update 2006. CA Cancer J Clin. 2006;56:168–183. doi: 10.3322/canjclin.56.3.168.
    1. Iorio MV, Ferracin M, Liu CG, Veronese A, Spizzo R, Sabbioni S, et al. MicroRNA gene expression deregulation in human breast cancer. Cancer Res. 2005;65:7065–7070. doi: 10.1158/0008-5472.CAN-05-1783.
    1. Song B, Wang C, Liu J, Wang X, Lv L, Wei L, et al. MicroRNA-21 regulates breast cancer invasion partly by targeting tissue inhibitor of metalloproteinase 3 expression. J Exp Clin Cancer Res. 2010;29:29. doi: 10.1186/1756-9966-29-29.
    1. Vivacqua A, De Marco P, Santolla MF, Cirillo F, Pellegrino M, Panno ML, et al. Estrogenic GPER signaling regulates mir144 expression in cancer cells and cancer-associated fibroblasts (CAFs) Oncotarget. 2015;6:16573–18711. doi: 10.18632/oncotarget.4117.
    1. Vivacqua A, De Marco P, Belfiore A, Maggiolini M. Recent advances on the role of microRNAs in both insulin resistance and Cancer. Curr Pharm Des. 2017;23:3658–3666. doi: 10.2174/1381612823666170622105123.
    1. Di Martino MT, Rossi M, Caracciolo D, Gullà A, Tagliaferri P, Tassone P. MiR-221 and miR-222 are promising targets for innovative anticancer therapy. Expert Opin Ther Targets. 2016;21:1–10.
    1. Han SH, Kim HJ, Gwak JM, Kim M, Chung YR, Park SY. MicroRNA-222 expression as a predictive marker for tumor progression in hormone receptor-positive breast cancer. J Breast Cancer. 2017;20:35–44. doi: 10.4048/jbc.2017.20.1.35.
    1. Shen H, Wang D, Li L, Yang S, Chen X, Zhou S, et al. Mir-222 promotes drug-resistance of breast cancer cells to adriamycin via modulation of PTEN/Akt/ FOXO1 pathway. Gene. 2017;596:110–118. doi: 10.1016/j.gene.2016.10.016.
    1. Di Martino MT, Gullà A, Cantafio ME, Lionetti M, Leone E, Amodio N, et al. In vitro and in vivo anti-tumor activity of miR-221/222 inhibitors in multiple myeloma. Oncotarget. 2013;4:242–255. doi: 10.18632/oncotarget.820.
    1. Fornari F, Gramantieri L, Ferracin M, Veronese A, Sabbioni S, Calin GA, et al. MiR-221 controls CDKN1C/p57 and CDKN1B/p27 expression in human hepatocellular carcinoma. Oncogene. 2008;27:5651–5661. doi: 10.1038/onc.2008.178.
    1. Zhao D, Zhuang N, Ding Y, Kang Y, Shi L. MiR-221 activates the NF-κB pathway by targeting A20. Biochem Biophys Res Commun. 2016;472:11–18. doi: 10.1016/j.bbrc.2015.11.009.
    1. Lappas M. A20, an essential component of the ubiquitin-editing protein complex, is a negative regulator of inflammation in human myometrium and foetal membranes. Mol Hum Reprod. 2017;23:628–645. doi: 10.1093/molehr/gax041.
    1. Düwel M, Welteke V, Oeckinghaus A, Baens M, Kloo B, Ferch U, et al. A20 negatively regulates T cell receptor signaling to NF-kappaB by cleaving Malt1 ubiquitin chains. J Immunol. 2009;182:7718–7728. doi: 10.4049/jimmunol.0803313.
    1. Karin M, Greten FR. NF-kappaB: linking inflammation and immunity to cancer development and progression. Nat Rev Immunol. 2005;5:749–759. doi: 10.1038/nri1703.
    1. Meteoglu I, Erdogdu IH, Meydan N, Erkus M, Barutca S. NF-kappa B expression correlates with apoptosis and angiogenesis in clear cell renal carcinoma tissues. J Exp Clin Cancer Res. 2008;27:53. doi: 10.1186/1756-9966-27-53.
    1. Leotoing L, Chereau F, Baron S, Hube F, Valencia HJ, Bordereaux D, et al. A20-binding inhibitor of nuclear factorkappaB (NF-kappaB)-2 (ABIN-2) is an activator of inhibitor of NF-kappaB (IkappaB) kinase alpha (IKKalpha)-mediated NF-kappaB transcriptional activity. J Biol Chem. 2011;286:32277–e32288. doi: 10.1074/jbc.M111.236448.
    1. Gao L, Coope H, Grant S, Ma A, Ley SC, Harhaj EW. ABIN1 protein cooperates with TAX1BP1 and A20 proteins to inhibit antiviral signaling. J Biol Chem. 2011;286:36592e36602.
    1. Xia M, Liu J, Wu X, Liu S, Li G, Han C, et al. Histone methyltransferase Ash1l suppresses interleukin-6 production and inflammatory autoimmune diseases by inducing the ubiquitin-editing enzyme A20. Immun. 2013;39:470–e481. doi: 10.1016/j.immuni.2013.08.016.
    1. Zhang X, Dong C, Sun X, Li Z, Zhang M, Guan Z, et al. Induction of the cellular miR-29c by influenza virus inhibits the innate immune response through protection of A20 mRNA. Biochem Biophys Res Commun. 2014;450:755e761.
    1. Liu X, He F, Pang R, Zhao D, Qiu W, Shan K, et al. Interleukin-17 (IL-17)-induced microRNA 873 (miR-873) contributes to the pathogenesis of experimental autoimmune encephalomyelitis by targeting A20 ubiquitin-editing enzyme. J Biol Chem. 2014;289:28971–e28986. doi: 10.1074/jbc.M114.577429.
    1. Kumar M, Sahu SK, Kumar R, Subuddhi A, Maji RK, Jana K, et al. MicroRNA let-7 modulates the immune response to Mycobacterium tuberculosis infection via control of A20, an inhibitor of the NF-kappaB pathway. Cell Host Microbe. 2015;17:345–e356. doi: 10.1016/j.chom.2015.01.007.
    1. Kim SW, Ramasamy K, Bouamar H, Lin AP, Jiang D, Aguiar RC. MicroRNAs miR-125a and miR-125b constitutively activate the NF-kappaB pathway by targeting the tumor necrosis factor alpha-induced protein 3 (TNFAIP3, A20) Proc Natl Acad Sci U S A. 2012;109:7865e. doi: 10.1073/pnas.1200081109.
    1. Cortes Sempere M, Rodrıguez Fanjul V, Sanchez Perez I, Perona R. The role of the NFkappaB signalling pathway in cancer. Clin Transl Oncol. 2008;10:143–147. doi: 10.1007/s12094-008-0171-3.
    1. Giordano M, Roncagalli R, Bourdely P, Chasson L, Buferne M, Yamasaki S. The tumor necrosis factor alpha-induced protein 3 (TNFAIP3, A20) imposes a brake on antitumor activity of CD8 T cells. Proc Natl Acad Sci U S A. 2014;111:11115–11120. doi: 10.1073/pnas.1406259111.
    1. Gilmore TD, Gerondakis S. The c-Rel transcription factor in development and disease. Genes Cancer. 2011;2:695–711. doi: 10.1177/1947601911421925.
    1. Romieu-Mourez R, Kim DW, Shin SM, Demicco EG, Landesman-Bollag E, Seldin DC, et al. Mouse mammary tumor virus c-rel transgenic mice develop mammary tumors. Mol Cell Biol. 2003;23:5738–5754. doi: 10.1128/MCB.23.16.5738-5754.2003.
    1. Xue X, Chen Q, Zhao G, Zhao JY, Duan Z, Zheng PS. The overexpression of TGF-β and CCN2 in intrauterine adhesions involves the NF-κB signaling pathway. PLoS One. 2015;10:e0146159. doi: 10.1371/journal.pone.0146159.
    1. Terada Y, Ueda S, Hamada K, Shimamura Y, Ogata K, Inoue K, et al. Aldosterone stimulates nuclear factor-kappa B activity and transcription of intercellular adhesion molecule-1 and connective tissue growth factor in rat mesangial cells via serum- and glucocorticoid-inducible protein kinase-1. Clin Exp Nephrol. 2012;16:81–88. doi: 10.1007/s10157-011-0498-x.
    1. Cicha I, Goppelt-Struebe M. Connective tissue growth factor: context-dependent functions and mechanisms of regulation. Biofactors. 2009;35:200–208. doi: 10.1002/biof.30.
    1. Vivacqua A, Romeo E, De Marco P, De Francesco EM, Abonante S, Maggiolini M. GPER mediates the Egr-1 expression induced by 17β-estradiol and 4-hydroxitamoxifen in breast and endometrial cancer cells. Breast Cancer Res Treat. 2012;133:1025–1035. doi: 10.1007/s10549-011-1901-8.
    1. Hutchenreuther J, Vincent KM, Carter DE, Postovit LM, Leask A. CCN2 expression by tumor stroma is required for melanoma metastasis. J Invest Dermatol. 2015;135:2805–2813. doi: 10.1038/jid.2015.279.
    1. Liu LY, Han YC, WU SH, ZH LV. Expression of connective tissue growth factor in tumor tissues is an independent predictor of poor prognosis in patients with gastric cancer. World J Gastroenterol. 2008;14:2110–2114. doi: 10.3748/wjg.14.2110.
    1. Bennewith KL, Huang X, Ham CM, Graves EE, Erler JT, Kambham N, et al. The role of tumor cell-derived connective tissue growth factor (CTGF/CCN2) in pancreatic tumor growth. Cancer Res. 2009;69:775–784. doi: 10.1158/0008-5472.CAN-08-0987.
    1. Yang F, Tuxhorn JA, Ressler SJ, McAlhany SJ, Dang TD, Rowley DR. Stromal expression of connective tissue growth factor promotes angiogenesis and prostate cancer tumorigenesis. Cancer Res. 2005;65:8887–8895. doi: 10.1158/0008-5472.CAN-05-1702.
    1. Jia Q, Bu Y, Wang Z, Chen B, Zhang Q, Yu S, et al. Maintenance of stemness is associated with the interaction of LRP6 and heparin-binding protein CCN2 autocrined by hepatocellular carcinoma. J Exp Clin Cancer Res. 2017;36:117. doi: 10.1186/s13046-017-0576-3.
    1. Ubink I, Verhaar ER, Kranenburg O, Goldschmeding R. A potential role for CCN2/CTGF in aggressive colorectal cancer. J Cell Commun Signal. 2016;10:223–227. doi: 10.1007/s12079-016-0347-5.
    1. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006;6:392–401. doi: 10.1038/nrc1877.
    1. Gullà A, Di Martino MT, Gallo Cantafio ME, Morelli E, Amodio N, Botta C, et al. A 13 mer LNA-i-miR-221 inhibitor restores drug sensitivity in Melphalan-refractory multiple myeloma cells. Clin Cancer Res. 2016;22:1222–1233. doi: 10.1158/1078-0432.CCR-15-0489.
    1. Di Martino MT, Gullà A, Gallo Cantafio ME, Altomare E, Amodio N, Leone E, et al. In vitro and in vivo activity of a novel locked nucleic acid (LNA)-inhibitor-miR-221 against multiple myeloma cells. PLoS One. 2014;9:e89659. doi: 10.1371/journal.pone.0089659.
    1. Gallo Cantafio ME, Nielsen BS, Mignogna C, Arbitrio M, Botta C, Frandsen NM, et al. Pharmacokinetics and pharmacodynamics of a 13-mer LNA-inhibitor-miR-221 in mice and non-human primates. Mol Ther Nucleic Acids. 2016;5 10.1038/mtna.2016.36.
    1. Franzoni S, Vezzelli A, Turtoro A, Solazzo L, Greco A, Tassone P, et al. Development and validation of a bioanalytical method for quantification of LNA-i-miR-221, a 13-mer oligonucleotide, in rat plasma using LC–MS/MS. J Pharm Biomed Anal. 2018;150:300–307. doi: 10.1016/j.jpba.2017.12.027.
    1. Chaqour B, Yang R, Sha Q. Mechanical stretch modulates the promoter activity of the profibrotic factor CCN2 through increased actin polymerization and NF-kappaB activation. J Biol Chem. 2006;281:20608–20622. doi: 10.1074/jbc.M600214200.
    1. Khaled WT, Choon Lee S, Stingl J, Chen X, Raza Ali H, Rueda OM, et al. BCL11A is a triple-negative breast cancer gene with critical function in stem and progenitor cells. Nat Commun. 2015;6:5987. doi: 10.1038/ncomms6987.
    1. Blows FM, Driver KE, Schmidt MK, Broeks A, van Leeuwen FE, Wesseling J, et al. Subtyping of breast cancer by immunohistochemistry to investigate a relationship between subtype and short and long term survival: a collaborative analysis of data for 10,159 cases from 12 studies. PLoS Med. 2010;7:e1000279. doi: 10.1371/journal.pmed.1000279.
    1. Knoop AS, Laenkholm AV, Jensen MB, Nielsen KV, Andersen J, Nielsen D, et al. Estrogen receptor, progesterone receptor, HER2 status and Ki67 index and responsiveness to adjuvant tamoxifen in postmenopausal high-risk breast cancer patients enrolled in the DBCG 77C trial. Eur J Cancer. 2014;50:1412–1421. doi: 10.1016/j.ejca.2014.02.022.
    1. Maly JJ, Macrae ER. Pertuzumab in combination with Trastuzumab and chemotherapy in the treatment of HER2-positive metastatic breast Cancer: safety, efficacy, and progression free survival. Breast Cancer (Auckl) 2014;8:81–88.
    1. Yang Q, Liu HY, Liu D, Song YQ. Ultrasonographic features of triple-negative breast cancer: a comparison with other breast cancer subtypes. Asian Pac J Cancer Prev. 2014;16:3229–3232. doi: 10.7314/APJCP.2015.16.8.3229.
    1. Iorfida M, Maiorano E, Orvieto E, Maisonneuve P, Bottiglieri L, Rotmensz N, et al. Invasive lobular breast cancer: subtypes and outcome. Breast Cancer Res Treat. 2012;133:713–723. doi: 10.1007/s10549-012-2002-z.
    1. Zhi K, Shen X, Zhang H, Bi J. Cancer-associated fibroblasts are positively correlated with metastatic potential of human gastric cancers. J Exp Clin Cancer Res. 2010;29:66. doi: 10.1186/1756-9966-29-66.
    1. Dong-Le Bourhis X, Berthois Y, Millot G, Degeorges A, Sylvi M, Martin PM, et al. Effect of stromal and epithelial cells derived from normal and tumorous breast tissue on the proliferation of human breast cancer cell lines in co-culture. Int J Cancer. 1997;71:42–48. doi: 10.1002/(SICI)1097-0215(19970328)71:1<42::AID-IJC9>;2-3.
    1. Frazier KS, Grotendorst GR. Expression of connective tissue growth factor mRNA in the fibrous stroma of mammary tumors. Int J Biochem Cell Biol. 1997;29:153–161. doi: 10.1016/S1357-2725(96)00127-6.
    1. Ellis MJ, Singer C, Hornby A, Rasmussen A, Cullen KJ. Insulin-like growth factor mediated stromal-epithelial interactions in human breast cancer. Breast Cancer Res Treat. 1994;31:249–261. doi: 10.1007/BF00666158.
    1. Erez N, Truitt M, Olson P, Arron ST, Hanahan D. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. Cancer Cell. 2010;17:135–147. doi: 10.1016/j.ccr.2009.12.041.
    1. Chi Y, Zhou D. MicroRNAs in colorectal carcinoma-from pathogenesis to therapy. J Exp Clin Cancer Res. 2016;35:43. doi: 10.1186/s13046-016-0320-4.
    1. Eissa S, Matboli M, Sharawy A, El-Sharkawi F. Prognostic and biological significance of microRNA-221 in breast cancer. Gene. 2015;574:163–167. doi: 10.1016/j.gene.2015.08.004.

Source: PubMed

3
Suscribir