EZH2 抑制剂 Tulmimetostat 的全面靶向作用可用于 ARID1A 突变癌症的靶向治疗。

IF 12.5 1区 医学 Q1 ONCOLOGY
Patricia J Keller, Elizabeth J Adams, Rentian Wu, Alexandre Côté, Shilpi Arora, Nico Cantone, Rosana Meyer, Jennifer A Mertz, Victor Gehling, Jike Cui, Jacob I Stuckey, Avinash Khanna, Feng Zhao, Zehua Chen, Ziyang Yu, Richard T Cummings, Mohammed Taimi, Nehal J Lakhani, Drew Rasco, Martin Gutierrez, Linda Duska, Michael Devitt, Ronda Rippley, Julian Levell, Jennifer Truong, Jing Wang, Kaiming Sun, Patrick Trojer
{"title":"EZH2 抑制剂 Tulmimetostat 的全面靶向作用可用于 ARID1A 突变癌症的靶向治疗。","authors":"Patricia J Keller, Elizabeth J Adams, Rentian Wu, Alexandre Côté, Shilpi Arora, Nico Cantone, Rosana Meyer, Jennifer A Mertz, Victor Gehling, Jike Cui, Jacob I Stuckey, Avinash Khanna, Feng Zhao, Zehua Chen, Ziyang Yu, Richard T Cummings, Mohammed Taimi, Nehal J Lakhani, Drew Rasco, Martin Gutierrez, Linda Duska, Michael Devitt, Ronda Rippley, Julian Levell, Jennifer Truong, Jing Wang, Kaiming Sun, Patrick Trojer","doi":"10.1158/0008-5472.CAN-24-0398","DOIUrl":null,"url":null,"abstract":"<p><p>Recurrent somatic mutations in the BRG1/BRM-associated factor (BAF) chromatin remodeling complex subunit ARID1A occur frequently in advanced urothelial, endometrial, and ovarian clear cell carcinomas, creating an alternative chromatin state that may be exploited therapeutically. The histone methyltransferase EZH2 has been previously identified as targetable vulnerability in the context of ARID1A mutations. In this study, we describe the discovery of tulmimetostat, an orally available, clinical stage EZH2 inhibitor, and it elucidates the aspects of its application potential in ARID1A mutant tumors. Tulmimetostat administration achieved efficacy in multiple ARID1A mutant bladder, ovarian, and endometrial tumor models and improved cisplatin response in chemotherapy-resistant models. Consistent with its comprehensive and durable level of target coverage, tulmimetostat demonstrated greater efficacy than other PRC2-targeted inhibitors at comparable or lower exposures in a bladder cancer xenograft mouse model. Tulmimetostat mediated extensive changes in gene expression, in addition to a profound reduction in global H3K27me3 levels in tumors. Phase I clinical pharmacokinetic and pharmacodynamic data indicated that tulmimetostat exhibits durable exposure and profound target engagement. Importantly, a tulmimetostat controlled gene expression signature identified in whole blood from a cohort of 32 patients with cancer correlated with tulmimetostat exposure, representing a pharmacodynamic marker for the assessment of target coverage for PRC2-targeted agents in the clinic. Collectively, these data suggest that tulmimetostat has the potential to achieve clinical benefit in solid tumors as a monotherapy but also in combination with chemotherapeutic agents, and may be beneficial in various indications with recurrent ARID1A mutations. Significance: The EZH2 inhibitor tulmimetostat achieves comprehensive target inhibition in ARID1A mutant solid tumor models and cancer patients that can be assessed with a pharmacodynamic gene signature in peripheral blood.</p>","PeriodicalId":9441,"journal":{"name":"Cancer research","volume":null,"pages":null},"PeriodicalIF":12.5000,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11292196/pdf/","citationCount":"0","resultStr":"{\"title\":\"Comprehensive Target Engagement by the EZH2 Inhibitor Tulmimetostat Allows for Targeting of ARID1A Mutant Cancers.\",\"authors\":\"Patricia J Keller, Elizabeth J Adams, Rentian Wu, Alexandre Côté, Shilpi Arora, Nico Cantone, Rosana Meyer, Jennifer A Mertz, Victor Gehling, Jike Cui, Jacob I Stuckey, Avinash Khanna, Feng Zhao, Zehua Chen, Ziyang Yu, Richard T Cummings, Mohammed Taimi, Nehal J Lakhani, Drew Rasco, Martin Gutierrez, Linda Duska, Michael Devitt, Ronda Rippley, Julian Levell, Jennifer Truong, Jing Wang, Kaiming Sun, Patrick Trojer\",\"doi\":\"10.1158/0008-5472.CAN-24-0398\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Recurrent somatic mutations in the BRG1/BRM-associated factor (BAF) chromatin remodeling complex subunit ARID1A occur frequently in advanced urothelial, endometrial, and ovarian clear cell carcinomas, creating an alternative chromatin state that may be exploited therapeutically. The histone methyltransferase EZH2 has been previously identified as targetable vulnerability in the context of ARID1A mutations. In this study, we describe the discovery of tulmimetostat, an orally available, clinical stage EZH2 inhibitor, and it elucidates the aspects of its application potential in ARID1A mutant tumors. Tulmimetostat administration achieved efficacy in multiple ARID1A mutant bladder, ovarian, and endometrial tumor models and improved cisplatin response in chemotherapy-resistant models. Consistent with its comprehensive and durable level of target coverage, tulmimetostat demonstrated greater efficacy than other PRC2-targeted inhibitors at comparable or lower exposures in a bladder cancer xenograft mouse model. Tulmimetostat mediated extensive changes in gene expression, in addition to a profound reduction in global H3K27me3 levels in tumors. Phase I clinical pharmacokinetic and pharmacodynamic data indicated that tulmimetostat exhibits durable exposure and profound target engagement. Importantly, a tulmimetostat controlled gene expression signature identified in whole blood from a cohort of 32 patients with cancer correlated with tulmimetostat exposure, representing a pharmacodynamic marker for the assessment of target coverage for PRC2-targeted agents in the clinic. Collectively, these data suggest that tulmimetostat has the potential to achieve clinical benefit in solid tumors as a monotherapy but also in combination with chemotherapeutic agents, and may be beneficial in various indications with recurrent ARID1A mutations. Significance: The EZH2 inhibitor tulmimetostat achieves comprehensive target inhibition in ARID1A mutant solid tumor models and cancer patients that can be assessed with a pharmacodynamic gene signature in peripheral blood.</p>\",\"PeriodicalId\":9441,\"journal\":{\"name\":\"Cancer research\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":12.5000,\"publicationDate\":\"2024-08-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11292196/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Cancer research\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1158/0008-5472.CAN-24-0398\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"ONCOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cancer research","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1158/0008-5472.CAN-24-0398","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

在晚期尿路上皮癌、子宫内膜癌和卵巢透明细胞癌中,BAF染色质重塑复合体亚基ARID1A经常发生体细胞突变,从而产生了一种可用于治疗的替代染色质状态。组蛋白甲基转移酶 EZH2 此前已被确定为 ARID1A 突变背景下的可靶向易损性。在此,我们介绍了一种口服的、处于临床阶段的 EZH2 抑制剂 tulmimetostat 的发现,并阐明了它在治疗 ARID1A 突变肿瘤方面的治疗潜力。Tulmimetostat 在多个 ARID1A 突变膀胱、卵巢和子宫内膜肿瘤模型中都取得了疗效,并改善了化疗耐药模型对顺铂的反应。在膀胱癌异种移植小鼠模型中,与其他 PRC2 靶向抑制剂相比,图利美司他在相似或更低的暴露量下显示出更高的疗效,这与其全面而持久的靶点覆盖水平相一致。除了显著降低肿瘤中的全局 H3K27me3 水平外,Tulmimetostat 还介导了基因表达的广泛变化。I 期临床药代动力学和药效学数据表明,曲美司他具有持久的暴露和深度靶向作用。重要的是,在一组 32 名癌症患者的全血中发现的曲美司他控制基因表达特征与曲美司他暴露相关,是评估临床中 PRC2 靶向药物靶点覆盖情况的药效学标志物。总之,这些数据表明,度米司他不仅有可能作为一种单药治疗实体瘤,也有可能与化疗药物联合使用,并在复发性ARID1A突变的各种适应症中发挥有益作用。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
Comprehensive Target Engagement by the EZH2 Inhibitor Tulmimetostat Allows for Targeting of ARID1A Mutant Cancers.

Recurrent somatic mutations in the BRG1/BRM-associated factor (BAF) chromatin remodeling complex subunit ARID1A occur frequently in advanced urothelial, endometrial, and ovarian clear cell carcinomas, creating an alternative chromatin state that may be exploited therapeutically. The histone methyltransferase EZH2 has been previously identified as targetable vulnerability in the context of ARID1A mutations. In this study, we describe the discovery of tulmimetostat, an orally available, clinical stage EZH2 inhibitor, and it elucidates the aspects of its application potential in ARID1A mutant tumors. Tulmimetostat administration achieved efficacy in multiple ARID1A mutant bladder, ovarian, and endometrial tumor models and improved cisplatin response in chemotherapy-resistant models. Consistent with its comprehensive and durable level of target coverage, tulmimetostat demonstrated greater efficacy than other PRC2-targeted inhibitors at comparable or lower exposures in a bladder cancer xenograft mouse model. Tulmimetostat mediated extensive changes in gene expression, in addition to a profound reduction in global H3K27me3 levels in tumors. Phase I clinical pharmacokinetic and pharmacodynamic data indicated that tulmimetostat exhibits durable exposure and profound target engagement. Importantly, a tulmimetostat controlled gene expression signature identified in whole blood from a cohort of 32 patients with cancer correlated with tulmimetostat exposure, representing a pharmacodynamic marker for the assessment of target coverage for PRC2-targeted agents in the clinic. Collectively, these data suggest that tulmimetostat has the potential to achieve clinical benefit in solid tumors as a monotherapy but also in combination with chemotherapeutic agents, and may be beneficial in various indications with recurrent ARID1A mutations. Significance: The EZH2 inhibitor tulmimetostat achieves comprehensive target inhibition in ARID1A mutant solid tumor models and cancer patients that can be assessed with a pharmacodynamic gene signature in peripheral blood.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Cancer research
Cancer research 医学-肿瘤学
CiteScore
16.10
自引率
0.90%
发文量
7677
审稿时长
2.5 months
期刊介绍: Cancer Research, published by the American Association for Cancer Research (AACR), is a journal that focuses on impactful original studies, reviews, and opinion pieces relevant to the broad cancer research community. Manuscripts that present conceptual or technological advances leading to insights into cancer biology are particularly sought after. The journal also places emphasis on convergence science, which involves bridging multiple distinct areas of cancer research. With primary subsections including Cancer Biology, Cancer Immunology, Cancer Metabolism and Molecular Mechanisms, Translational Cancer Biology, Cancer Landscapes, and Convergence Science, Cancer Research has a comprehensive scope. It is published twice a month and has one volume per year, with a print ISSN of 0008-5472 and an online ISSN of 1538-7445. Cancer Research is abstracted and/or indexed in various databases and platforms, including BIOSIS Previews (R) Database, MEDLINE, Current Contents/Life Sciences, Current Contents/Clinical Medicine, Science Citation Index, Scopus, and Web of Science.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信