Oops, you're using an old version of your browser so some of the features on this page may not be displaying properly.

MINIMAL Requirements: Google Chrome 24+Mozilla Firefox 20+Internet Explorer 11Opera 15–18Apple Safari 7SeaMonkey 2.15-2.23

Poster session 12

1722P - The subtype-specific cell fitness of head and neck cancer is regulated by the YBX1 integration of oncogenic PI3K/mTOR signalling

Date

10 Sep 2022

Session

Poster session 12

Topics

Tumour Site

Head and Neck Cancers

Presenters

Yuchen Bai

Citation

Annals of Oncology (2022) 33 (suppl_7): S772-S784. 10.1016/annonc/annonc1079

Authors

Y. Bai, C. Darido

Author affiliations

  • Cancer Research Department, Peter MacCallum Cancer Centre, 3000 - Melbourne/AU

Resources

Login to get immediate access to this content.

If you do not have an ESMO account, please create one for free.

Abstract 1722P

Background

Molecular subtyping of heterogeneous cancers has gained substantial biological interest although with limited translation to the clinic. In heterogeneous head and neck cancer (HNC), subtype-specific treatment regimens are currently missing.

Methods

A multi-omic approach integrating whole-genome and single-cell transcriptome sequencing with reverse-phase protein array was employed in patient primary and metastatic tumours and human cell lines to investigate the subtype-specific molecular mechanisms as the therapeutic vulnerabilities in HNC.

Results

An integrated analysis of HNC subtypes using single-cell sequencing and proteome profiles revealed a hallmark epithelial-mesenchymal transition (EMT) signature within the epithelial cancer-cell population. The EMT signature was evident in the invasive mesenchymal subtype and surprisingly coincided with PI3K/mTOR pathway inactivation. Conversely, the EMT signature was suppressed in epithelial cancer cells of the basal subtype which exhibited hyperactive PI3K/mTOR oncogenic signalling. We further identified that phosphorylation of Y Box binding protein 1 (YBX1), downstream of the PI3K/mTOR pathway, restrained the basal-like cancer cell proliferation. In contrast, YBX1 acted as a safeguard against the proliferation to invasion switch in mesenchymal-like epithelial cancer cells, and its loss accentuated partial EMT acquisition and in vivo invasion. Interestingly, the proteomic analysis of patient HNC identified phospho-YBX1 as a prognostic marker for overall patient outcomes, the loss of which correlated with HNC metastasis and poor prognosis.

Conclusions

Our results indicate that HNC cells adopt a PI3K-phospho-YBX1-dependent proliferation program that is mutually exclusive to partial EMT involvement which relies on the lack of phosphorylation of YBX1 in PI3K-inactive cancer cells. These findings create a unique opportunity to sensitise mesenchymal cancer cells to PI3K/mTOR inhibitors by shifting them towards a basal-like subtype, constituting a promising therapeutic approach against HNC.

Clinical trial identification

Editorial acknowledgement

Legal entity responsible for the study

The authors.

Funding

Australian Government National Health and Medical Research Council.

Disclosure

All authors have declared no conflicts of interest.

This site uses cookies. Some of these cookies are essential, while others help us improve your experience by providing insights into how the site is being used.

For more detailed information on the cookies we use, please check our Privacy Policy.

Customise settings
  • Necessary cookies enable core functionality. The website cannot function properly without these cookies, and you can only disable them by changing your browser preferences.