[HTML][HTML] Myeloid-derived interleukin-1β drives oncogenic KRAS-NF-κΒ addiction in malignant pleural effusion

A Marazioti, I Lilis, M Vreka, H Apostolopoulou… - Nature …, 2018 - nature.com
A Marazioti, I Lilis, M Vreka, H Apostolopoulou, A Kalogeropoulou, I Giopanou
Nature communications, 2018nature.com
Malignant pleural effusion (MPE) is a frequent metastatic manifestation of human cancers.
While we previously identified KRAS mutations as molecular culprits of MPE formation, the
underlying mechanism remained unknown. Here, we determine that non-canonical IKKα-
RelB pathway activation of KRAS-mutant tumor cells mediates MPE development and this is
fueled by host-provided interleukin IL-1β. Indeed, IKKα is required for the MPE-competence
of KRAS-mutant tumor cells by activating non-canonical NF-κB signaling. IL-1β fuels …
Abstract
Malignant pleural effusion (MPE) is a frequent metastatic manifestation of human cancers. While we previously identified KRAS mutations as molecular culprits of MPE formation, the underlying mechanism remained unknown. Here, we determine that non-canonical IKKα-RelB pathway activation of KRAS-mutant tumor cells mediates MPE development and this is fueled by host-provided interleukin IL-1β. Indeed, IKKα is required for the MPE-competence of KRAS-mutant tumor cells by activating non-canonical NF-κB signaling. IL-1β fuels addiction of mutant KRAS to IKKα resulting in increased CXCL1 secretion that fosters MPE-associated inflammation. Importantly, IL-1β-mediated NF-κB induction in KRAS-mutant tumor cells, as well as their resulting MPE-competence, can only be blocked by co-inhibition of both KRAS and IKKα, a strategy that overcomes drug resistance to individual treatments. Hence we show that mutant KRAS facilitates IKKα-mediated responsiveness of tumor cells to host IL-1β, thereby establishing a host-to-tumor signaling circuit that culminates in inflammatory MPE development and drug resistance.
nature.com