Cargando…

Selective inhibition of EZH2 by ZLD1039 blocks H3K27methylation and leads to potent anti-tumor activity in breast cancer

Enhancer of zeste homolog 2 (EZH2) is a candidate oncogenic driver due to its prevalent overexpression and aberrant repression of tumor suppressor genes in diverse cancers. Therefore, blocking EZH2 enzyme activity may present a valid therapeutic strategy for the treatment of cancers with EZH2 overex...

Descripción completa

Detalles Bibliográficos
Autores principales: Song, Xuejiao, Gao, Tiantao, Wang, Ningyu, Feng, Qiang, You, Xinyu, Ye, Tinghong, Lei, Qian, Zhu, Yongxia, Xiong, Menghua, Xia, Yong, Yang, Fangfang, Shi, Yaojie, Wei, Yuquan, Zhang, Lidan, Yu, Luoting
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group 2016
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4751454/
https://www.ncbi.nlm.nih.gov/pubmed/26868841
http://dx.doi.org/10.1038/srep20864
_version_ 1782415584471810048
author Song, Xuejiao
Gao, Tiantao
Wang, Ningyu
Feng, Qiang
You, Xinyu
Ye, Tinghong
Lei, Qian
Zhu, Yongxia
Xiong, Menghua
Xia, Yong
Yang, Fangfang
Shi, Yaojie
Wei, Yuquan
Zhang, Lidan
Yu, Luoting
author_facet Song, Xuejiao
Gao, Tiantao
Wang, Ningyu
Feng, Qiang
You, Xinyu
Ye, Tinghong
Lei, Qian
Zhu, Yongxia
Xiong, Menghua
Xia, Yong
Yang, Fangfang
Shi, Yaojie
Wei, Yuquan
Zhang, Lidan
Yu, Luoting
author_sort Song, Xuejiao
collection PubMed
description Enhancer of zeste homolog 2 (EZH2) is a candidate oncogenic driver due to its prevalent overexpression and aberrant repression of tumor suppressor genes in diverse cancers. Therefore, blocking EZH2 enzyme activity may present a valid therapeutic strategy for the treatment of cancers with EZH2 overexpression including breast cancers. Here, we described ZLD1039 a potent, highly selective, and orally bioavailable small molecule inhibitor of EZH2, which inhibited breast tumor growth and metastasis. ZLD1039 considerably inhibited EZH2 methyltransferase activity with nanomolar potency, decreased global histone-3 lysine-27 (H3K27) methylation, and reactivated silenced tumor suppressors connected to increased survival of patients with breast cancer. Comparable to conditional silencing of EZH2, its inhibition by ZLD1039 decreased cell proliferation, cell cycle arrest, and induced apoptosis. Comparably, treatment of xenograft-bearing mice with ZLD1039 led to tumor growth regression and metastasis inhibition. These data confirmed the dependency of breast cancer progression on EZH2 activity and the usefulness of ZLD1039 as a promising treatment for breast cancer.
format Online
Article
Text
id pubmed-4751454
institution National Center for Biotechnology Information
language English
publishDate 2016
publisher Nature Publishing Group
record_format MEDLINE/PubMed
spelling pubmed-47514542016-02-22 Selective inhibition of EZH2 by ZLD1039 blocks H3K27methylation and leads to potent anti-tumor activity in breast cancer Song, Xuejiao Gao, Tiantao Wang, Ningyu Feng, Qiang You, Xinyu Ye, Tinghong Lei, Qian Zhu, Yongxia Xiong, Menghua Xia, Yong Yang, Fangfang Shi, Yaojie Wei, Yuquan Zhang, Lidan Yu, Luoting Sci Rep Article Enhancer of zeste homolog 2 (EZH2) is a candidate oncogenic driver due to its prevalent overexpression and aberrant repression of tumor suppressor genes in diverse cancers. Therefore, blocking EZH2 enzyme activity may present a valid therapeutic strategy for the treatment of cancers with EZH2 overexpression including breast cancers. Here, we described ZLD1039 a potent, highly selective, and orally bioavailable small molecule inhibitor of EZH2, which inhibited breast tumor growth and metastasis. ZLD1039 considerably inhibited EZH2 methyltransferase activity with nanomolar potency, decreased global histone-3 lysine-27 (H3K27) methylation, and reactivated silenced tumor suppressors connected to increased survival of patients with breast cancer. Comparable to conditional silencing of EZH2, its inhibition by ZLD1039 decreased cell proliferation, cell cycle arrest, and induced apoptosis. Comparably, treatment of xenograft-bearing mice with ZLD1039 led to tumor growth regression and metastasis inhibition. These data confirmed the dependency of breast cancer progression on EZH2 activity and the usefulness of ZLD1039 as a promising treatment for breast cancer. Nature Publishing Group 2016-02-12 /pmc/articles/PMC4751454/ /pubmed/26868841 http://dx.doi.org/10.1038/srep20864 Text en Copyright © 2016, Macmillan Publishers Limited http://creativecommons.org/licenses/by/4.0/ This work is licensed under a Creative Commons Attribution 4.0 International License. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in the credit line; if the material is not included under the Creative Commons license, users will need to obtain permission from the license holder to reproduce the material. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/
spellingShingle Article
Song, Xuejiao
Gao, Tiantao
Wang, Ningyu
Feng, Qiang
You, Xinyu
Ye, Tinghong
Lei, Qian
Zhu, Yongxia
Xiong, Menghua
Xia, Yong
Yang, Fangfang
Shi, Yaojie
Wei, Yuquan
Zhang, Lidan
Yu, Luoting
Selective inhibition of EZH2 by ZLD1039 blocks H3K27methylation and leads to potent anti-tumor activity in breast cancer
title Selective inhibition of EZH2 by ZLD1039 blocks H3K27methylation and leads to potent anti-tumor activity in breast cancer
title_full Selective inhibition of EZH2 by ZLD1039 blocks H3K27methylation and leads to potent anti-tumor activity in breast cancer
title_fullStr Selective inhibition of EZH2 by ZLD1039 blocks H3K27methylation and leads to potent anti-tumor activity in breast cancer
title_full_unstemmed Selective inhibition of EZH2 by ZLD1039 blocks H3K27methylation and leads to potent anti-tumor activity in breast cancer
title_short Selective inhibition of EZH2 by ZLD1039 blocks H3K27methylation and leads to potent anti-tumor activity in breast cancer
title_sort selective inhibition of ezh2 by zld1039 blocks h3k27methylation and leads to potent anti-tumor activity in breast cancer
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4751454/
https://www.ncbi.nlm.nih.gov/pubmed/26868841
http://dx.doi.org/10.1038/srep20864
work_keys_str_mv AT songxuejiao selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer
AT gaotiantao selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer
AT wangningyu selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer
AT fengqiang selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer
AT youxinyu selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer
AT yetinghong selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer
AT leiqian selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer
AT zhuyongxia selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer
AT xiongmenghua selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer
AT xiayong selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer
AT yangfangfang selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer
AT shiyaojie selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer
AT weiyuquan selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer
AT zhanglidan selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer
AT yuluoting selectiveinhibitionofezh2byzld1039blocksh3k27methylationandleadstopotentantitumoractivityinbreastcancer