Abstract

Simple SummaryTriple negative breast cancer is an aggressive subtype of breast cancer that frequently metastasizes. Because the transcription factor FOXM1 is highly upregulated in triple negative breast cancer and controls many cell activities that lead to cancer progression and metastasis, we sought to determine if FOXM1 inhibitory compounds could effectively suppress the invasiveness and progression of triple negative breast cancer cells and tumors. Our findings show that these compounds inhibit cell motility, invasiveness, and the expression of important proteins associated with epithelial to mesenchymal transition. These compounds also suppressed the proliferation and metastatic outgrowth of triple negative breast tumors. Thus, these findings highlight the crucial role of FOXM1 in promoting the progression and metastasis of these cancers, and suggest that FOXM1 inhibitory compounds may have therapeutic potential and prove beneficial in intervention against triple negative breast cancer.Metastasis-related complications account for the overwhelming majority of breast cancer mortalities. Triple negative breast cancer (TNBC), the most aggressive breast cancer subtype, has a high propensity to metastasize to distant organs, leading to poor patient survival. The forkhead transcription factor, FOXM1, is especially upregulated and overexpressed in TNBC and is known to regulate multiple signaling pathways that control many key cancer properties, including proliferation, invasiveness, stem cell renewal, and therapy resistance, making FOXM1 a critical therapeutic target for TNBC. In this study, we test the effectiveness of a novel class of 1,1-diarylethylene FOXM1 inhibitory compounds in suppressing TNBC cell migration, invasion, and metastasis using in vitro cell culture and in vivo tumor models. We show that these compounds inhibit the motility and invasiveness of TNBC MDA-MB-231 and DT28 cells, along with reducing the expression of important epithelial to mesenchymal transition (EMT) associated genes. Further, orthotopic tumor studies in NOD-SCID-gamma (NSG) mice demonstrate that these compounds reduce FOXM1 expression and suppress TNBC tumor growth as well as distant metastasis. Gene expression and protein analyses confirm the decreased levels of EMT factors and FOXM1-regulated target genes in tumors and metastatic lesions in the inhibitor-treated animals. The findings suggest that these FOXM1 suppressive compounds may have therapeutic potential in treating triple negative breast cancer, with the aim of reducing tumor progression and metastatic outgrowth.

Highlights

  • Triple negative breast cancers (TNBCs) represent a highly aggressive breast cancer subtype that very frequently develops treatment resistance and metastasizes to distant organs

  • As a prelude to examining the role of FOXM1 in TNBC metastasis, we first investigated the effect of our FOXM1 inhibitory compounds on the migration of TNBC cells

  • Having observed the effect of the FOXM1 inhibitory compounds on TNBC cell motility and invasion, we investigated their effect on expression of important epithelial to mesenchymal transition (EMT)-related genes known to be involved in breast cancer metastasis

Read more

Summary

Introduction

Triple negative breast cancers (TNBCs) represent a highly aggressive breast cancer subtype that very frequently develops treatment resistance and metastasizes to distant organs. This recurrence and metastatic spread is a leading cause of morbidity and mortality in patients with these breast cancers [1,2]. FOXM1 transcriptionally regulates the expression of key players in epithelial to mesenchymal transition (EMT) These factors, including Snail, Slug, Vimentin, and MMPs, are directly involved in the early stages of cancer metastasis by allowing the conversion of adhesive epithelial cells to a more migratory phenotype, along with a change in the stromal compartment, thereby facilitating cell spread [16,17,18,19]. Targeting FOXM1 could prove to be a useful new therapeutic strategy, allowing the suppression of multiple key cancer regulatory pathways

Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.