Abstract

Abstract Background: Ewing sarcoma (ES) is a malignant bone tumor characterized by the oncogenic fusion protein EWS-FLI1. EWS-FLI1 acts as an aberrant transcription factor, inducing and silencing genes involved with increasing cell proliferation and survival. We have previously shown that lurbinectedin, a trabectedin analog, effectively inactivates the transcriptional activity of EWS-FLI1 by redistributing it within the nucleus to the nucleolus. Interestingly, this process activates a gene signature like that of the DNA damage response (DDR) to UV light, which was also shown to induce nucleolar translocation of WT Ewing sarcoma breakpoint 1 (EWSR1). However, the mechanism governing the nucleolar translocation of EWS-FLI1 induced by these drugs has yet to be elucidated. In addition, novel approaches to exploit this relocalization mechanism as a means to broaden the therapeutic window have yet to be established. Methods: We used immunoblotting, RT-qPCR, viability assays, cell fractionation, confocal microscopy, proximity ligation assays (PLA), mass spectrometry, genomics, and immunoprecipitation to investigate the mechanism of action of trabectedin/lurbinectedin-induced nucleolar relocalization of EWS-FLI1. Results: Through interrogation of the pathways triggered by trabectedin/lurbinectedin, we found that this mechanism is in part mediated by HSP70. Viability assays also revealed profound synergy between HSP70 inhibitors (HSP70is) and lurbinectedin. Immunoprecipitated EWS-FLI1 subjected to mass spectrometry revealed an association of EWS-FLI1 and several HSP70 isoforms after trabectedin treatment which was confirmed with PLA. Trabectedin/lurbinectedin treatment also revealed an upregulation of EWS-FLI1-suppressed targets and suppression of induced targets. Confocal microscopy showed that EWS-FLI1 “trapping” in the nucleolus was potentiated by combination treatment with HSP70is. Conclusion: Our results underscore the synergistic effect of combining HSP70is with trabectedin or lurbinectedin treatment to attenuate several aspects of ES tumorigenesis in vitro. In vivo experiments with patient-derived xenografts are underway to corroborate the synergy of HSP70is and trabectedin/lurbinectedin. Together, our data highlights the potential for a novel “EWS-FLI1 nucleolar trap” as a means to inhibit and sustain EWS-FLI1 repression with this class of compounds. Citation Format: Sridhar M. Veluvolu, Raphael D. Lopez, Jenna M. Gedminas, Elissa A. Boguslawski, Elizabeth R. Wilson, Benjamin P. Caiello, Maureen E. Murphy, Patrick J. Grohar. Exploiting lurbinectedin-driven nucleolar relocalization of EWS-FLI1 to develop novel combination therapies for Ewing sarcoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 1082.

Full Text
Published version (Free)

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