Please use this identifier to cite or link to this item: http://hdl.handle.net/2445/124571
Title: Nintedanib selectively inhibits the activation and tumour-promoting effects of fibroblasts from lung adenocarcinoma patients
Author: Gabasa Ferràndez, Marta
Ikemori, Rafael
Hilberg, F.
Reguart, Noemí
Alcaraz Casademunt, Jordi
Keywords: Fibroblasts
Fibrosi pulmonar
Reacció en cadena de la polimerasa
Farmacologia
Càncer de pulmó
Fibroblasts
Pulmonary fibrosis
Polymerase chain reaction
Pharmacology
Lung cancer
Issue Date: 12-Sep-2017
Publisher: Cancer Research UK
Abstract: Background: Nintedanib is a clinically approved multikinase receptor inhibitor to treat non-small cell lung cancer with adenocarcinoma (ADC) histology in combination with docetaxel, based on the clinical benefits reported on ADC but not on squamous cell carcinoma (SCC), which are the two most common histologic lung cancer subtypes.Methods: We examined the potential role of tumour-associated fibroblasts (TAFs) in the differential effects of nintedanib in ADC and SCC. Because TAFs are largely quiescent and activated in histologic sections, we focused on the antifibrotic effects of nintedanib on TAFs stimulated with the potent fibroblast activator TGF-beta 1, which is upregulated in lung cancer.Results: Nintedanib dose-dependently inhibited the TGF-beta 1-induced expression of a panel of pro-fibrotic activation markers in both ADC-TAFs and control fibroblasts derived from uninvolved lung parenchyma, whereas such inhibition was very modest in SCC-TAFs. Remarkably, nintedanib abrogated the stimulation of growth and invasion in a panel of carcinoma cell lines induced by secreted factors from activated TAFs in ADC but not SCC, thereby supporting that TGF-beta signalling and aberrant TAF-carcinoma cross-talk is regulated by different mechanisms in ADC and SCC.Conclusions: These results reveal that nintedanib is an effective inhibitor of fibrosis and its associated tumour-promoting effects in ADC, and that the poor antifibrotic response of SCC-TAFs to nintedanib may contribute to the differential clinical benefit observed in both subtypes. Our findings also support that preclinical models based on carcinoma-TAF interactions may help defining the mechanisms of the poor antifibrotic response of SCC-TAFs to nintedanib and testing new combined therapies to further expand the therapeutic effects of this drug in solid tumours.
Note: Reproducció del document publicat a: https://doi.org/10.1038/bjc.2017.270
It is part of: British Journal of Cancer, 2017, vol. 117, num. 8, p. 1128-1138
URI: http://hdl.handle.net/2445/124571
Related resource: https://doi.org/10.1038/bjc.2017.270
ISSN: 0007-0920
Appears in Collections:Articles publicats en revistes (IDIBAPS: Institut d'investigacions Biomèdiques August Pi i Sunyer)
Articles publicats en revistes (Biomedicina)

Files in This Item:
File Description SizeFormat 
677497.pdf1.65 MBAdobe PDFView/Open


This item is licensed under a Creative Commons License Creative Commons