Abstract

Dysregulated epidermal growth factor receptor (EGFR) is an oncogenic driver of many human cancers, promoting aberrant cell proliferation, migration, and survival. Pharmacological targeting of EGFR is often challenged by acquired mechanisms of resistance. Ligand-dependent mechanisms in EGFR wild-type cells rely on ligand or receptor overexpression, allowing cells to outcompete inhibitors and perpetuate signaling in an autocrine manner. Importantly, EGFR ligands are synthesized as membrane-bound precursors that must be solubilized to enable receptor-ligand interactions. The A disintegrin and metalloproteinase 17 (ADAM17) is considered the main sheddase of several EGFR ligands, and a potential pharmacological target. However, its broad substrate range and ubiquitous expression complicate its therapeutic targeting. Here, we present a novel bispecific fusion protein construct consisting of the inhibitory prodomain of ADAM17 (TPD), fused to an EGFR-targeting designed ankyrin repeat protein (DARPin). TPD is a natural inhibitor of ADAM17, maintaining the protease in a zymogen-like form. Meanwhile, the high affinity anti-EGFR DARPin E01 binds to EGFR and inhibits ligand binding. The resulting fusion protein E01-GS-TPD retained binding ability to both molecular targets EGFR and ADAM17. The large difference in affinity for each target resulted in enrichment of the fusion protein in EGFR-positive cells compared to EGFR-negative cells, suggesting a possible application in autocrine signaling inhibition. Accordingly, E01-GS-TPD decreased migration and proliferation of EGFR-dependent cell lines with no significant increase in apoptotic cell death. Finally, inhibition of proliferation was observed through EGFR ligand-dependent mechanisms as growth inhibition was not observed in EGFR mutant or KRAS mutant cell lines. The use of bispecific proteins targeting the EGFR/ADAM17 axis could be an innovative strategy for the treatment of EGFR-dependent cancers.

Highlights

  • Dysregulated of the epidermal growth factor receptor (EGFR) is an oncogenic driver of many human cancers, promoting aberrant cell proliferation, migration, and survival [1]

  • In this study we aimed to disrupt oncogenic EGFR/A disintegrin and metalloproteinase 17 (ADAM17) signaling using a novel bispecific fusion protein consisting of the inhibitory prodomain of ADAM17 (TPD) fused to an anti-EGFR

  • We opted for the reported designed ankyrin repeat protein (DARPin) E01, capable of binding domain III of the EGFR ectodomain at low nanomolar previously reported DARPin E01, capable of binding domain III of the EGFR ectodomain at low affinity in a specific manner, as well as preventing ligand binding and EGFR activation [16,17]

Read more

Summary

Introduction

Dysregulated of the epidermal growth factor receptor (EGFR) is an oncogenic driver of many human cancers, promoting aberrant cell proliferation, migration, and survival [1]. Dysregulated EGFR signaling and acquired drug resistance can occur in a Cancers 2020, 12, 411; doi:10.3390/cancers12020411 www.mdpi.com/journal/cancers. Cancers 2020, 12, 411 ligand-independent fashion via mutations leading to constitutive activation of the EGFR receptor [2] or that of downstream signaling components such as KRAS [3]. EGFR ligands are synthesized as membrane-bound immature precursors and their proteolytic cleavage from the cell surface enables solubilization, diffusion, and receptor binding. The shedding of EGFR ligands, constitutes an additional layer of regulation in establishing receptor-ligand interactions [5]. EGFR-ligand shedding is primarily catalyzed by transmembrane proteinases of the

Objectives
Results
Discussion
Conclusion
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