Abstract

The epidermal growth factor receptor (EGFR) contributes to the pathogenesis of head&neck squamous cell carcinoma (HNSCC). However, only a subset of HNSCC patients benefit from anti-EGFR targeted therapy. By performing an unbiased proteomics screen, we found that the calcium-activated chloride channel ANO1 interacts with EGFR and facilitates EGFR-signaling in HNSCC. Using structural mutants of EGFR and ANO1 we identified the trans/juxtamembrane domain of EGFR to be critical for the interaction with ANO1. Our results show that ANO1 and EGFR form a functional complex that jointly regulates HNSCC cell proliferation. Expression of ANO1 affected EGFR stability, while EGFR-signaling elevated ANO1 protein levels, establishing a functional and regulatory link between ANO1 and EGFR. Co-inhibition of EGFR and ANO1 had an additive effect on HNSCC cell proliferation, suggesting that co-targeting of ANO1 and EGFR could enhance the clinical potential of EGFR-targeted therapy in HNSCC and might circumvent the development of resistance to single agent therapy. HNSCC cell lines with amplification and high expression of ANO1 showed enhanced sensitivity to Gefitinib, suggesting ANO1 overexpression as a predictive marker for the response to EGFR-targeting agents in HNSCC therapy. Taken together, our results introduce ANO1 as a promising target and/or biomarker for EGFR-directed therapy in HNSCC.

Highlights

  • Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide, with a predicted 40,000 new cases and 8,000 deaths in the USA in 2014 [1]

  • epidermal growth factor receptor (EGFR) coimmunoprecipitated when ANO1 was captured with an ANO1-specific antibody and showed no binding when an unspecific antibody was used as a control

  • These findings support the conclusion that ANO1 and EGFR form a complex in Te11 cells

Read more

Summary

INTRODUCTION

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide, with a predicted 40,000 new cases and 8,000 deaths in the USA in 2014 [1]. Activation of EGFR by specific ligands including EGF, TGF-alpha, HB-EGF and amphiregulin induces the homo- and hetero-dimerization of EGFR with other ErbB proteins and leads to the phosphorylation of tyrosine residues in the cytoplasmic domain of the receptor These phosphotyrosine residues serve as docking sites for proteins which initiate the activation of key signaling pathways like the mitogenactivated protein kinase (MAPK)- or the PI3K-activated www.impactjournals.com/oncotarget protein kinase B (AKT)-pathway; key regulators of HNSCC cell proliferation, tumor growth, invasion and metastasis [4, 6]. Consistent with ANO1 being a regulator of EGFR-signaling, HNSCC cell lines with amplification and high expression of ANO1 showed enhanced sensitivity to Gefitinib, suggesting ANO1 overexpression as a predictive marker for the response to EGFR-targeting agents in HNSCC therapies. Our results introduce ANO1 as a promising target and/or biomarker for EGFR-directed therapy in HNSCC

RESULTS
DISCUSSION
EXPERIMENTAL PROCEDURES
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call