IBYME   02675
INSTITUTO DE BIOLOGIA Y MEDICINA EXPERIMENTAL
Unidad Ejecutora - UE
congresos y reuniones científicas
Título:
Targeting nuclear ErbB-2 function in triple negative breast cancer
Autor/es:
FLORENCIA CHERVO; FRANCO IZZO; MARA DE MARTINO; LEANDRO VENTURUTTI; SANTIAGO MADERA; JUAN CRUZ GÓMEZ POVIÑA; VIOLETA CHIAUZZI; CECILIA J PROIETTI; PABLO GUZMAN; JUAN C. ROA; EDUARDO H CHARREAU; ROXANA SCHILLACI; PATRICIA V ELIZALDE; ROSALÍA I. CORDO RUSSO
Lugar:
Boston, MA, EEUU.
Reunión:
Congreso; The Endocrine Society 98th Annual Meeting; 2016
Institución organizadora:
The Endocrine Society
Resumen:
riple negative breast cancer (TNBC) refers to the group of tumors with poor prognosis without clinically significant levels of estrogen and progesterone receptors, and which lack membrane ErbB-2 (MErbB-2) overexpression or gene amplification. ErbB-2/HER2 receptor tyrosine kinase is a major player in the breast cancer (BC) scenario. The dogma of ErbB-2 mechanism of action has been challenged by the demonstration that MErbB-2 migrates to the nucleus of BC cells where it acts as a transcription factor. We demonstrated that nuclear ErbB-2 (NErbB-2) acts also as a transcriptional coactivator of the signal transducer and activator of transcription 3 (Stat3) to modulate BC growth (1). Most recently, we have found that ErbB-2 assembles a transcriptional complex with Stat3 and ErbB-3, another member of the ErbBs family, which revealed the first nuclear function of ErbB-2/ErbB-3 dimer (2). Here, we explored NErbB-2 presence in TNBC using immunofluorescence (IF) and confocal microscopy. We found a striking NErbB-2 presence in TN MDA-MB-468, HCC-70, MDA-MB-231, and MDA-MB-453 cells, which belong to different TNBC subtypes (3). We also detected NErbB-2 in TNBC tumors. In accordance with previous findings (4,5), we observed constitutive Stat3 and ErbB-3 nuclear localization in TNBC cells. Notably, we here also found a strong nuclear colocalization of ErbB-2/Stat3 and of ErbB-2/ErbB-3. To explore NErbB-2 role in TNBC proliferation, cells were transfected with the ErbB-2DNLS mutant, which is unable to translocate to the nucleus and acts as dominant negative inhibitor of endogenous NErbB-2 translocation (1,2). Our present findings revealed that transfection of ErbB-2DNLS abolished NErbB-2 presence and proliferation in a series of TNBC cell lines, thus demonstrating that NErbB2 is a major proliferation driver in TNBC. The clinical significance of NErbB-2 was assessed in tissue microarrays from a small cohort of 226 primary invasive breast carcinomas by IF. Among them, 51 (23%) were TNBC. Our results revealed NErbB-2 positivity as a significant predictor of worse overall survival in TNBC patients (p=0.045). These findings for the first time reveal NErbB-2 presence and function in TNBC and highlight NErbB-2 as a novel therapeutic target in TNBC.(1) Béguelin W et al., Mol Cell Biol 2010; 30 (23): 5456-72. (2) Cordo Russo RI et al., Oncogene 2015; 34 (26): 3413-28. (3) Lehmann BD et al., J Clin Invest 2011; 121:2750-2767. (4) Lee HJ et al., Carcinogenesis 2011; 32:804-811. (5) Tao R-Het al., J Cell Sci 2008; 121: 3207-17.- See more at: http://press.endocrine.org/doi/abs/10.1210/endo-meetings.2016.TB.1.FRI-066#sthash.4utkHVAN.dpuf