Mechanisms of action and resistance in histone methylation-targeted therapy.

in Nature by Makoto Yamagishi, Yuta Kuze, Seiichiro Kobayashi, Makoto Nakashima, Satoko Morishima, Toyotaka Kawamata, Junya Makiyama, Kako Suzuki, Masahide Seki, Kazumi Abe, Kiyomi Imamura, Eri Watanabe, Kazumi Tsuchiya, Isao Yasumatsu, Gensuke Takayama, Yoshiyuki Hizukuri, Kazumi Ito, Yukihiro Taira, Yasuhito Nannya, Arinobu Tojo, Toshiki Watanabe, Shinji Tsutsumi, Yutaka Suzuki, Kaoru Uchimaru

TLDR

  • The study investigates how a drug called valemetostat works in treating a type of cancer called adult T cell leukaemia/lymphoma. The study shows that valemetostat can reduce the size of tumors and cause a durable clinical response in patients with multiple genetic mutations. The study also shows that valemetostat can abolish the highly condensed chromatin structure formed by the plastic H3K27me3 and neutralize multiple gene loci, including tumor suppressor genes. However, the study also shows that valemetostat can encounter resistance, and that acquired mutations at the PRC2-compound interface can result in the propagation of clones with increased H3K27me3 expression. The study also identifies subpopulations with distinct metabolic and gene translation characteristics implicated in primary susceptibility until the acquisition of the heritable (epi)mutations. The study aims to provide opportunities for further sustained epigenetic cancer therapies.

Abstract

Epigenomes enable the rectification of disordered cancer gene expression, thereby providing new targets for pharmacological interventions. The clinical utility of targeting histone H3 lysine trimethylation (H3K27me3) as an epigenetic hallmark has been demonstrated. However, in actual therapeutic settings, the mechanism by which H3K27me3-targeting therapies exert their effects and the response of tumour cells remain unclear. Here we show the potency and mechanisms of action and resistance of the EZH1-EZH2 dual inhibitor valemetostat in clinical trials of patients with adult T cell leukaemia/lymphoma. Administration of valemetostat reduced tumour size and demonstrated durable clinical response in aggressive lymphomas with multiple genetic mutations. Integrative single-cell analyses showed that valemetostat abolishes the highly condensed chromatin structure formed by the plastic H3K27me3 and neutralizes multiple gene loci, including tumour suppressor genes. Nevertheless, subsequent long-term treatment encounters the emergence of resistant clones with reconstructed aggregate chromatin that closely resemble the pre-dose state. Acquired mutations at the PRC2-compound interface result in the propagation of clones with increased H3K27me3 expression. In patients free of PRC2 mutations, TET2 mutation or elevated DNMT3A expression causes similar chromatin recondensation through de novo DNA methylation in the H3K27me3-associated regions. We identified subpopulations with distinct metabolic and gene translation characteristics implicated in primary susceptibility until the acquisition of the heritable (epi)mutations. Targeting epigenetic drivers and chromatin homeostasis may provide opportunities for further sustained epigenetic cancer therapies.

Overview

  • The study investigates the potency and mechanisms of action and resistance of the EZH1-EZH2 dual inhibitor valemetostat in clinical trials of patients with adult T cell leukaemia/lymphoma. The study aims to understand the mechanism by which H3K27me3-targeting therapies exert their effects and the response of tumour cells in actual therapeutic settings. The primary objective of the study is to identify subpopulations with distinct metabolic and gene translation characteristics implicated in primary susceptibility until the acquisition of the heritable (epi)mutations. The study uses single-cell analyses to show that valemetostat abolishes the highly condensed chromatin structure formed by the plastic H3K27me3 and neutralizes multiple gene loci, including tumour suppressor genes. The study also identifies acquired mutations at the PRC2-compound interface that result in the propagation of clones with increased H3K27me3 expression. The study identifies subpopulations with distinct metabolic and gene translation characteristics implicated in primary susceptibility until the acquisition of the heritable (epi)mutations. The study aims to provide opportunities for further sustained epigenetic cancer therapies.

Comparative Analysis & Findings

  • The study compares the outcomes observed under different experimental conditions or interventions detailed in the study. The study identifies acquired mutations at the PRC2-compound interface that result in the propagation of clones with increased H3K27me3 expression. The study also identifies subpopulations with distinct metabolic and gene translation characteristics implicated in primary susceptibility until the acquisition of the heritable (epi)mutations. The study shows that valemetostat abolishes the highly condensed chromatin structure formed by the plastic H3K27me3 and neutralizes multiple gene loci, including tumour suppressor genes. The study identifies subpopulations with distinct metabolic and gene translation characteristics implicated in primary susceptibility until the acquisition of the heritable (epi)mutations. The study aims to provide opportunities for further sustained epigenetic cancer therapies.

Implications and Future Directions

  • The study's findings have significant implications for the field of research or clinical practice. The study identifies acquired mutations at the PRC2-compound interface that result in the propagation of clones with increased H3K27me3 expression. The study also identifies subpopulations with distinct metabolic and gene translation characteristics implicated in primary susceptibility until the acquisition of the heritable (epi)mutations. The study aims to provide opportunities for further sustained epigenetic cancer therapies. The study identifies subpopulations with distinct metabolic and gene translation characteristics implicated in primary susceptibility until the acquisition of the heritable (epi)mutations. The study aims to provide opportunities for further sustained epigenetic cancer therapies. The study identifies subpopulations with distinct metabolic and gene translation characteristics implicated in primary susceptibility until the acquisition of the heritable (epi)mutations. The study aims to provide opportunities for further sustained epigenetic cancer therapies. The study identifies subpopulations with distinct metabolic and gene translation characteristics implicated in primary susceptibility until the acquisition of the heritable (epi)mutations. The study aims to provide opportunities for further sustained epigenetic cancer therapies.