Quick Search:

IDHwt glioblastomas can be stratified by their transcriptional response to standard treatment, with implications for targeted therapy

Tanner, Georgette, Barrow, Rhiannon, Ajaib, Shoaib, Al-Jabri, Muna, Ahmed, Nazia, Pollock, Steven, Finetti, Martina, Rippaus, Nora, Bruns, Alexander, Syed, Khaja, Poulter, James, Matthews, Laura, Hughes, Thomas A ORCID: https://orcid.org/0000-0003-1169-3386, Wilson, Erica, Johnson, Colin, Varn, Frederick, Brüning-Richardson, Anke, Hogg, Catherine, Droop, Alastair, Gusnanto, Arief, Care, Matthew, Cutillo, Luisa, Westhead, David, Susan, Short, Jenkinson, Michael, Brodbelt, Andrew, Chakrabarty, Aruna, Ismail, Azzam, Verhaak, Roel GW and Stead, Lucy F (2024) IDHwt glioblastomas can be stratified by their transcriptional response to standard treatment, with implications for targeted therapy. Genome Biology, 25 (1).

[img]
Preview
Text
s13059-024-03172-3.pdf - Published Version
Available under License Creative Commons Attribution.

| Preview

Abstract

Background
Glioblastoma (GBM) brain tumors lacking IDH1 mutations (IDHwt) have the worst prognosis of all brain neoplasms. Patients receive surgery and chemoradiotherapy but tumors almost always fatally recur.

Results
Using RNA sequencing data from 107 pairs of pre- and post-standard treatment locally recurrent IDHwt GBM tumors, we identify two responder subtypes based on longitudinal changes in gene expression. In two thirds of patients, a specific subset of genes is upregulated from primary to recurrence (Up responders), and in one third, the same genes are downregulated (Down responders), specifically in neoplastic cells. Characterization of the responder subtypes indicates subtype-specific adaptive treatment resistance mechanisms that are associated with distinct changes in the tumor microenvironment. In Up responders, recurrent tumors are enriched in quiescent proneural GBM stem cells and differentiated neoplastic cells, with increased interaction with the surrounding normal brain and neurotransmitter signaling, whereas Down responders commonly undergo mesenchymal transition. ChIP-sequencing data from longitudinal GBM tumors suggests that the observed transcriptional reprogramming could be driven by Polycomb-based chromatin remodeling rather than DNA methylation.

Conclusions
We show that the responder subtype is cancer-cell intrinsic, recapitulated in in vitro GBM cell models, and influenced by the presence of the tumor microenvironment. Stratifying GBM tumors by responder subtype may lead to more effective treatment.

Item Type: Article
Status: Published
DOI: https://doi.org/10.1186/s13059-024-03172-3
Subjects: Q Science > Q Science (General)
School/Department: School of Science, Technology and Health
URI: https://ray.yorksj.ac.uk/id/eprint/9205

University Staff: Request a correction | RaY Editors: Update this record