Cargando…

Inhibition of HDACs reduces Ewing sarcoma tumor growth through EWS-FLI1 protein destabilization

Oncogenic transcription factors lacking enzymatic activity or targetable binding pockets are typically considered “undruggable”. An example is provided by the EWS-FLI1 oncoprotein, whose continuous expression and activity as transcription factor are critically required for Ewing sarcoma tumor format...

Descripción completa

Detalles Bibliográficos
Autores principales: Pedot, Gloria, Marques, Joana Graça, Ambühl, Philip P., Wachtel, Marco, Kasper, Stephanie, Ngo, Quy A., Niggli, Felix K., Schäfer, Beat W.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Neoplasia Press 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8971315/
https://www.ncbi.nlm.nih.gov/pubmed/35366465
http://dx.doi.org/10.1016/j.neo.2022.100784
_version_ 1784679606292316160
author Pedot, Gloria
Marques, Joana Graça
Ambühl, Philip P.
Wachtel, Marco
Kasper, Stephanie
Ngo, Quy A.
Niggli, Felix K.
Schäfer, Beat W.
author_facet Pedot, Gloria
Marques, Joana Graça
Ambühl, Philip P.
Wachtel, Marco
Kasper, Stephanie
Ngo, Quy A.
Niggli, Felix K.
Schäfer, Beat W.
author_sort Pedot, Gloria
collection PubMed
description Oncogenic transcription factors lacking enzymatic activity or targetable binding pockets are typically considered “undruggable”. An example is provided by the EWS-FLI1 oncoprotein, whose continuous expression and activity as transcription factor are critically required for Ewing sarcoma tumor formation, maintenance, and proliferation. Because neither upstream nor downstream targets have so far disabled its oncogenic potential, we performed a high-throughput drug screen (HTS), enriched for FDA-approved drugs, coupled to a Global Protein Stability (GPS) approach to identify novel compounds capable to destabilize EWS-FLI1 protein by enhancing its degradation through the ubiquitin-proteasome system. The protein stability screen revealed the dual histone deacetylase (HDAC) and phosphatidylinositol-3-kinase (PI3K) inhibitor called fimepinostat (CUDC-907) as top candidate to modulate EWS-FLI1 stability. Fimepinostat strongly reduced EWS-FLI1 protein abundance, reduced viability of several Ewing sarcoma cell lines and PDX-derived primary cells and delayed tumor growth in a xenograft mouse model, whereas it did not significantly affect healthy cells. Mechanistically, we demonstrated that EWS-FLI1 protein levels were mainly regulated by fimepinostat's HDAC activity. Our study demonstrates that HTS combined to GPS is a reliable approach to identify drug candidates able to modulate stability of EWS-FLI1 and lays new ground for the development of novel therapeutic strategies aimed to reduce Ewing sarcoma tumor progression.
format Online
Article
Text
id pubmed-8971315
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Neoplasia Press
record_format MEDLINE/PubMed
spelling pubmed-89713152022-04-07 Inhibition of HDACs reduces Ewing sarcoma tumor growth through EWS-FLI1 protein destabilization Pedot, Gloria Marques, Joana Graça Ambühl, Philip P. Wachtel, Marco Kasper, Stephanie Ngo, Quy A. Niggli, Felix K. Schäfer, Beat W. Neoplasia Original Research Oncogenic transcription factors lacking enzymatic activity or targetable binding pockets are typically considered “undruggable”. An example is provided by the EWS-FLI1 oncoprotein, whose continuous expression and activity as transcription factor are critically required for Ewing sarcoma tumor formation, maintenance, and proliferation. Because neither upstream nor downstream targets have so far disabled its oncogenic potential, we performed a high-throughput drug screen (HTS), enriched for FDA-approved drugs, coupled to a Global Protein Stability (GPS) approach to identify novel compounds capable to destabilize EWS-FLI1 protein by enhancing its degradation through the ubiquitin-proteasome system. The protein stability screen revealed the dual histone deacetylase (HDAC) and phosphatidylinositol-3-kinase (PI3K) inhibitor called fimepinostat (CUDC-907) as top candidate to modulate EWS-FLI1 stability. Fimepinostat strongly reduced EWS-FLI1 protein abundance, reduced viability of several Ewing sarcoma cell lines and PDX-derived primary cells and delayed tumor growth in a xenograft mouse model, whereas it did not significantly affect healthy cells. Mechanistically, we demonstrated that EWS-FLI1 protein levels were mainly regulated by fimepinostat's HDAC activity. Our study demonstrates that HTS combined to GPS is a reliable approach to identify drug candidates able to modulate stability of EWS-FLI1 and lays new ground for the development of novel therapeutic strategies aimed to reduce Ewing sarcoma tumor progression. Neoplasia Press 2022-03-30 /pmc/articles/PMC8971315/ /pubmed/35366465 http://dx.doi.org/10.1016/j.neo.2022.100784 Text en © 2022 The Authors. Published by Elsevier Inc. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Original Research
Pedot, Gloria
Marques, Joana Graça
Ambühl, Philip P.
Wachtel, Marco
Kasper, Stephanie
Ngo, Quy A.
Niggli, Felix K.
Schäfer, Beat W.
Inhibition of HDACs reduces Ewing sarcoma tumor growth through EWS-FLI1 protein destabilization
title Inhibition of HDACs reduces Ewing sarcoma tumor growth through EWS-FLI1 protein destabilization
title_full Inhibition of HDACs reduces Ewing sarcoma tumor growth through EWS-FLI1 protein destabilization
title_fullStr Inhibition of HDACs reduces Ewing sarcoma tumor growth through EWS-FLI1 protein destabilization
title_full_unstemmed Inhibition of HDACs reduces Ewing sarcoma tumor growth through EWS-FLI1 protein destabilization
title_short Inhibition of HDACs reduces Ewing sarcoma tumor growth through EWS-FLI1 protein destabilization
title_sort inhibition of hdacs reduces ewing sarcoma tumor growth through ews-fli1 protein destabilization
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8971315/
https://www.ncbi.nlm.nih.gov/pubmed/35366465
http://dx.doi.org/10.1016/j.neo.2022.100784
work_keys_str_mv AT pedotgloria inhibitionofhdacsreducesewingsarcomatumorgrowththroughewsfli1proteindestabilization
AT marquesjoanagraca inhibitionofhdacsreducesewingsarcomatumorgrowththroughewsfli1proteindestabilization
AT ambuhlphilipp inhibitionofhdacsreducesewingsarcomatumorgrowththroughewsfli1proteindestabilization
AT wachtelmarco inhibitionofhdacsreducesewingsarcomatumorgrowththroughewsfli1proteindestabilization
AT kasperstephanie inhibitionofhdacsreducesewingsarcomatumorgrowththroughewsfli1proteindestabilization
AT ngoquya inhibitionofhdacsreducesewingsarcomatumorgrowththroughewsfli1proteindestabilization
AT nigglifelixk inhibitionofhdacsreducesewingsarcomatumorgrowththroughewsfli1proteindestabilization
AT schaferbeatw inhibitionofhdacsreducesewingsarcomatumorgrowththroughewsfli1proteindestabilization