Epigenetic SMAD3 repression in tumor-associated fibroblasts impairs fibrosis and response to the antifibrotic drug nintedanib in lung squamous cell carcinoma.

The tumor-promoting fibrotic stroma rich in tumor-associated fibroblasts (TAFs) is drawing increased therapeutic attention. Intriguingly, a trial with the antifibrotic drug nintedanib in non-small cell lung cancer reported clinical benefits in adenocarcinoma (ADC) but not squamous cell carcinoma (SCC), even though the stroma is fibrotic in both histotypes. Likewise, we reported that nintedanib inhibited the tumor-promoting fibrotic phenotype of TAFs selectively in ADC. Here we show that tumor fibrosis is actually higher in ADC-TAFs than SCC-TAFs in vitro and patient samples. Mechanistically, the reduced fibrosis and nintedanib response of SCC-TAFs were associated with increased promoter methylation of the pro-fibrotic TGF-β transcription factor SMAD3 compared to ADC-TAFs, which elicited a compensatory increase in TGF-β1/SMAD2 activation. Consistently, forcing global DNA demethylation of SCC-TAFs with 5-AZA rescued TGF-β1/SMAD3 activation, whereas genetic downregulation of SMAD3 in ADC-TAFs and control fibroblasts increased TGF-β1/SMAD2 activation, and reduced their fibrotic phenotype and antitumor responses to nintedanib in vitro and in vivo. Our results also support that smoking and/or the anatomic location of SCC in the proximal airways, which are more exposed to cigarette smoke particles, may prime SCC-TAFs to stronger SMAD3 epigenetic repression, since cigarette smoke condensate selectively increased SMAD3 promoter methylation. Our results unveil that the histotype-specific regulation of tumor fibrosis in lung cancer is mediated through differential SMAD3 promoter methylation in TAFs, and provide new mechanistic insights on the selective poor response of SCC-TAFs to nintedanib. Moreover, our findings support that ADC patients may be more responsive to antifibrotic drugs targeting their stromal TGF-β1/SMAD3 activation.

[1]  J. Alcaraz,et al.  Stromal markers of activated tumor associated fibroblasts predict poor survival and are associated with necrosis in non-small cell lung cancer. , 2019, Lung cancer.

[2]  P. Carmeliet,et al.  Phenotype molding of stromal cells in the lung tumor microenvironment , 2018, Nature Medicine.

[3]  R. West,et al.  GFPT2-Expressing Cancer-Associated Fibroblasts Mediate Metabolic Reprogramming in Human Lung Adenocarcinoma. , 2018, Cancer Research.

[4]  J. Alcaraz,et al.  Epithelial contribution to the profibrotic stiff microenvironment and myofibroblast population in lung fibrosis , 2017, Molecular biology of the cell.

[5]  J. Alcaraz,et al.  Nintedanib selectively inhibits the activation and tumour-promoting effects of fibroblasts from lung adenocarcinoma patients , 2017, British Journal of Cancer.

[6]  V. Peinado,et al.  Cigarette smoke challenges bone marrow mesenchymal stem cell capacities in guinea pig , 2017, Respiratory Research.

[7]  R. Kalluri The biology and function of fibroblasts in cancer , 2016, Nature Reviews Cancer.

[8]  J. Alcaraz,et al.  Heterotypic paracrine signaling drives fibroblast senescence and tumor progression of large cell carcinoma of the lung , 2016, Oncotarget.

[9]  A. Russo,et al.  Nintedanib in NSCLC: evidence to date and place in therapy , 2016, Therapeutic advances in medical oncology.

[10]  Agnes G Loeffler,et al.  Periductal stromal collagen topology of pancreatic ductal adenocarcinoma differs from that of normal and chronic pancreatitis , 2015, Modern Pathology.

[11]  J. Alcaraz,et al.  Aberrant DNA methylation in non-small cell lung cancer-associated fibroblasts , 2015, Carcinogenesis.

[12]  S. Estrem,et al.  Clinical development of galunisertib (LY2157299 monohydrate), a small molecule inhibitor of transforming growth factor-beta signaling pathway , 2015, Drug design, development and therapy.

[13]  Kevin J. Harrington,et al.  The tumour microenvironment after radiotherapy: mechanisms of resistance and recurrence , 2015, Nature Reviews Cancer.

[14]  Shulian Wang,et al.  Nondosimetric risk factors for radiation-induced lung toxicity. , 2015, Seminars in radiation oncology.

[15]  Alexander Pautsch,et al.  Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis , 2015, European Respiratory Journal.

[16]  A. Jemal,et al.  Global cancer statistics, 2012 , 2015, CA: a cancer journal for clinicians.

[17]  Cheng Huang,et al.  Smad2 protects against TGF-β1/Smad3-mediated collagen synthesis in human hepatic stellate cells during hepatic fibrosis , 2015, Molecular and Cellular Biochemistry.

[18]  Yongbing Chen,et al.  High p-Smad2 expression in stromal fibroblasts predicts poor survival in patients with clinical stage I to IIIA non-small cell lung cancer , 2014, World Journal of Surgical Oncology.

[19]  J. Alcaraz,et al.  Matrix Stiffening and β1 Integrin Drive Subtype-Specific Fibroblast Accumulation in Lung Cancer , 2014, Molecular Cancer Research.

[20]  Daniel Öhlund,et al.  Fibroblast heterogeneity in the cancer wound , 2014, The Journal of experimental medicine.

[21]  Kwok-Kin Wong,et al.  Non-small-cell lung cancers: a heterogeneous set of diseases , 2014, Nature Reviews Cancer.

[22]  Rolf Kaiser,et al.  Docetaxel plus nintedanib versus docetaxel plus placebo in patients with previously treated non-small-cell lung cancer (LUME-Lung 1): a phase 3, double-blind, randomised controlled trial. , 2014, The Lancet. Oncology.

[23]  T. Schmid,et al.  High transforming growth factor β expression represents an important prognostic parameter for surgically resected non-small cell lung cancer. , 2012, Human pathology.

[24]  D. Carbone,et al.  Smoking Attenuates Transforming Growth Factor-β–Mediated Tumor Suppression Function through Downregulation of Smad3 in Lung Cancer , 2012, Cancer Prevention Research.

[25]  Luca Richeldi,et al.  Efficacy of a tyrosine kinase inhibitor in idiopathic pulmonary fibrosis. , 2011, The New England journal of medicine.

[26]  Nikolina Radulovich,et al.  Prognostic gene-expression signature of carcinoma-associated fibroblasts in non-small cell lung cancer , 2011, Proceedings of the National Academy of Sciences.

[27]  E. Bottinger,et al.  Smad2 protects against TGF-beta/Smad3-mediated renal fibrosis. , 2010, Journal of the American Society of Nephrology : JASN.

[28]  N. Probst-Hensch,et al.  Prognostic Significance of Epithelial-Mesenchymal and Mesenchymal-Epithelial Transition Protein Expression in Non–Small Cell Lung Cancer , 2008, Clinical Cancer Research.

[29]  W. Sommergruber,et al.  BIBF 1120: triple angiokinase inhibitor with sustained receptor blockade and good antitumor efficacy. , 2008, Cancer research.

[30]  Liang Zhang,et al.  Identification of the gene transcription and apoptosis mediated by TGF‐β‐Smad2/3‐Smad4 signaling , 2008, Journal of cellular physiology.

[31]  J. Califano,et al.  DNA global hypomethylation in squamous cell head and neck cancer associated with smoking, alcohol consumption and stage , 2007, International journal of cancer.

[32]  R. Chambers,et al.  Molecular targets in pulmonary fibrosis: the myofibroblast in focus. , 2007, Chest.

[33]  A. Thorne,et al.  Regulation of myofibroblast transdifferentiation by DNA methylation and MeCP2: implications for wound healing and fibrogenesis , 2007, Cell Death and Differentiation.

[34]  Brian Bierie,et al.  Tumour microenvironment: TGFβ: the molecular Jekyll and Hyde of cancer , 2006, Nature Reviews Cancer.

[35]  A. Roberts,et al.  Smad3 Null Mice Develop Airspace Enlargement and Are Resistant to TGF-β-Mediated Pulmonary Fibrosis1 , 2004, The Journal of Immunology.

[36]  Ying E. Zhang,et al.  Smad-dependent and Smad-independent pathways in TGF-β family signalling , 2003, Nature.

[37]  A. Reith,et al.  SB-431542 is a potent and specific inhibitor of transforming growth factor-beta superfamily type I activin receptor-like kinase (ALK) receptors ALK4, ALK5, and ALK7. , 2002, Molecular pharmacology.

[38]  Thomas D. Schmittgen,et al.  Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. , 2001, Methods.

[39]  R. Kucherlapati,et al.  Functional Characterization of Transforming Growth Factor β Signaling in Smad2- and Smad3-deficient Fibroblasts* , 2001, The Journal of Biological Chemistry.

[40]  S. Khuder,et al.  Effect of cigarette smoking on major histological types of lung cancer: a meta-analysis. , 2001, Lung cancer.

[41]  Risa J. Robinson,et al.  Deposition of Cigarette Smoke Particles in the Human Respiratory Tract , 2001 .

[42]  J. Ou,et al.  Synergistic Cooperation between Sp1 and Smad3/Smad4 Mediates Transforming Growth Factor β1 Stimulation of α2(I)-Collagen (COL1A2) Transcription* , 2000, The Journal of Biological Chemistry.

[43]  C. Heldin,et al.  Expression of transforming‐growth‐factor (TGF)‐β receptors and Smad proteins in glioblastoma cell lines with distinct responses to TGF‐β1 , 1999 .

[44]  R. Derynck,et al.  Transforming growth factor-beta activation in irradiated murine mammary gland. , 1994, The Journal of clinical investigation.

[45]  A. Jemal,et al.  Cancer statistics, 2018 , 2018, CA: a cancer journal for clinicians.

[46]  V. Thannickal,et al.  Novel Mechanisms for the Antifibrotic Action of Nintedanib. , 2016, American journal of respiratory cell and molecular biology.

[47]  Yuming Liu,et al.  Computational segmentation of collagen fibers from second-harmonic generation images of breast cancer , 2014, Journal of biomedical optics.

[48]  Michael D. Abràmoff,et al.  Image processing with ImageJ , 2004 .

[49]  K. Knoch,et al.  Evidence for the involvement of TGF-beta and PDGF in the regulation of prolyl 4-hydroxylase and lysyloxidase in cultured rat lung fibroblasts. , 2003, Experimental and toxicologic pathology : official journal of the Gesellschaft fur Toxikologische Pathologie.