Abstract

Abstract Despite remarkable progress in diagnosis and treatment, metastasis from the primary tumor to distant organs is responsible for over 90% of cancer deaths. Therefore, there is a need to understand the drivers of the process to detect, target, and develop therapeutics to treat metastases better and extend their lifespan. Aurora-A Kinase (AURKA) is a serine/threonine kinase that typically localizes in the cytoplasm. However, the nuclear localization of AURKA (N-AURKA) has been reported in multiple cancers and correlates with poor survival and the advanced/metastatic stage of the disease. AURKA is often present in the nucleus of metastatic Triple Negative Breast Cancer (TNBC) cells and patient biopsies compared to non-metastatic cells. Moreover, induced translocation of AURKA to the nucleus in experimental models of TNBC results in a 5-fold increase in metastases to the bones, lungs, and liver compared to AURKA in the cytoplasm. We have previously shown that N-AURKA under normoxic conditions binds to HIF1A/1B and leads to induction of hypoxia signaling via transactivation of Hypoxia Inducible Factors (HIFs)-dependent genes, including migration/invasion, survival/death, and stemness, promoting early cancer dissemination. Transactivation of HIF1-genes requires cofactor p300/CBP. It is currently unknown if N-AURKA/HIF1 transcription is p300 dependent. In this study, we used the small molecular inhibitor Chetomin to disrupt the interaction of HIF1A and co-activator CBP/p300 in experimental TNBC and animal models combined with AURKA inhibitor Alisertib. Disruption of the hypoxia signaling pathway significantly decreases the transactivation of HIF-dependent genes, including migration/invasion, survival/death, and stemness in N-AURKA cells. Moreover, in mouse models, inhibition of HIF1 activity via treatment with Chetomin in combination with AURKA inhibitor Alisertib significantly decreased metastatic burden suggesting that p300 binding to AURKA/HIF complex is critical for gene expression and enabling metastatic dissemination/colonization. Our studies show the mechanistic relationship between N-AURKA and hypoxia signaling and lay the foundation for developing novel combination treatment strategies to improve patient outcomes. Citation Format: Abha Maskey, Kristina Marinak Whately, Elena N. Pugacheva. Targeting nuclear AURKA and hypoxia signaling pathway attenuates metastatic burden in triple negative breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 1296.

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.