1
|
Zhang Y, Shi Q, Fang W, Liu L, Yang H, Liu X, Huang Y, Zhang Y, Huang X, Wang Y. Discovery of Highly Potent and Selective EZH2 Covalent Inhibitors via Incorporating Basic Amines. J Med Chem 2025; 68:10365-10383. [PMID: 40340349 DOI: 10.1021/acs.jmedchem.5c00545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Targeted covalent inhibition is a promising strategy to address the high dose and acquired drug resistance issues of the first-generation EZH2 noncovalent inhibitors. Recently we have reported a new generation of highly potent EZH2 covalent inhibitors, but further optimization to enhance aqueous solubility is required. Here, we described the systematic optimization of EPZ-6438 by preserving the aqueous groups, resulting in the identification of a highly potent and selective EZH2 covalent inhibitor 13, which displayed nanomolar potency in biochemical and cellular assays. Moreover, SAM competition experiments preliminarily confirmed that 13 was noncompetitive with SAM, leading to the remarkable reduction of the H3K27Me3 marker. In addition, 13 exhibited superior cell growth inhibition in the EZH2 mutant cancer cell lines. The discovery of 13 holds promise for the development of highly potent EZH2 covalent inhibitors.
Collapse
Affiliation(s)
- Yi Zhang
- Balance-Based Drug Discovery Laboratory, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Qiongyu Shi
- Lingang Laboratory, Shanghai 200031, P. R. China
| | - Wei Fang
- Balance-Based Drug Discovery Laboratory, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Li Liu
- Balance-Based Drug Discovery Laboratory, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Hong Yang
- Lingang Laboratory, Shanghai 200031, P. R. China
| | - Xinqiao Liu
- Balance-Based Drug Discovery Laboratory, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yuting Huang
- Lingang Laboratory, Shanghai 200031, P. R. China
- School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ying Zhang
- Lingang Laboratory, Shanghai 200031, P. R. China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xun Huang
- Lingang Laboratory, Shanghai 200031, P. R. China
- School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuanxiang Wang
- Balance-Based Drug Discovery Laboratory, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
2
|
Xu M, Xu C, Wang R, Tang Q, Zhou Q, Wu W, Wan X, Mo H, Pan J, Wang S. Treating human cancer by targeting EZH2. Genes Dis 2025; 12:101313. [PMID: 40028035 PMCID: PMC11870178 DOI: 10.1016/j.gendis.2024.101313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 03/05/2025] Open
Abstract
Enhancer of zeste homolog 2 (EZH2), an epigenetic regulator that primarily inhibits downstream gene expression by tri-methylating histone H3, which is usually overexpressed in tumors and participates in many processes such as tumor occurrence and development, invasion, migration, drug resistance, and anti-tumor immunity as an oncogene, making it an important biomarker in cancer therapy. Collectively, several transcription factors and RNAs cooperate to facilitate the elevated expression of EZH2 in cancer. Although the significance of blocking EZH2 in cancer for inhibiting cancer progression is widely recognized, the clinical application of EZH2 inhibitors continues to encounter numerous challenges. In this review, drawing upon our comprehensive understanding of the factual underpinnings of EZH2's role in cancer, we aim to clarify the crucial importance of targeting EZH2 in cancer treatment. Furthermore, we summarize the current research landscape surrounding targeted EZH2 inhibitors and offer insights into potential future applications of these inhibitors.
Collapse
Affiliation(s)
- Mengfei Xu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Chunyan Xu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Rui Wang
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Qing Tang
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Qichun Zhou
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Wanyin Wu
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Xinliang Wan
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Handan Mo
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Jun Pan
- Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510120, China
| | - Sumei Wang
- Department of Oncology, Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, Guangdong 510120, China
| |
Collapse
|
3
|
Zheng J, Ma Z, Liu P, Wei J, Min S, Shan Y, Zhang J, Li Y, Xue L, Tan Z, Wang D. EZH2 inhibits senescence-associated inflammation and attenuates intervertebral disc degeneration by regulating the cGAS/STING pathway via H3K27me3. Osteoarthritis Cartilage 2025; 33:548-559. [PMID: 39938633 DOI: 10.1016/j.joca.2025.02.771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 01/28/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
OBJECTIVE Senescent nucleus pulposus mesenchymal stem cells (NPMSCs) are key instigators of local chronic inflammation and disruptions in nucleus pulposus tissue repair in intervertebral disc degeneration (IVDD). This study aimed to investigate the interplay between EZH2 and NPMSCs senescence-associated inflammation. METHODS Nucleus pulposus samples were collected from IVDD patients (n = 15, F/M = 7/8, average age 47.9 (21-72) year-old). Multiplex immunohistochemistry was conducted to detect the expression of EZH2 and the cGAS/STING pathway. Subsequently, NPMSCs were isolated from 7 patients (n = 7, F/M = 4/3, average age 49.4 (36-68) year-old). After treatment with tert-butyl hydroperoxide and lentivirus-overexpression-EZH2 (Lv-OE-EZH2), real time fluorescent quantitative PCR, immunofluorescence, western blot, and ChIP were used to detect the expression of EZH2 and the cGAS/STING pathway. Micro-CT, magnetic resonance imaging, and histological staining were performed to assess the therapeutic effects of Lv-OE-EZH2 and a STING inhibitor on rat IVDD. All experiment designs were independent. RESULTS In both human nucleus pulposus tissues and an in vitro cell model, EZH2 expression decreased while the cGAS/STING pathway became activated in senescent NPMSCs. ChIP assays and Lv-OE-EZH2 experiments validated that EZH2 epigenetically inhibited STING expression via H3K27me3, thereby impairing the cGAS/STING pathway and attenuating senescence-associated inflammation. Moreover, overexpression of EZH2 (Pfirrmann grade means difference -1.375, p = 0.0089) and inhibition of STING effectively attenuated rat IVDD. CONCLUSION The decreased expression of EZH2 in senescent NPMSCs promotes senescence-associated inflammation and the progression of IVDD, possibly by relieving the transcriptional inhibition of STING and enabling the activation of the cGAS/STING pathway.
Collapse
Affiliation(s)
- Jianrui Zheng
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, PR China.
| | - Zetao Ma
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, PR China.
| | - Pei Liu
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, PR China.
| | - Jiewen Wei
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, PR China; Shantou University Medical College, Shantou 515000, PR China.
| | - Shaoxiong Min
- Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, PR China.
| | - Ying Shan
- Clinical Research Academy, Peking University Shenzhen Hospital, Shenzhen 518000, PR China.
| | - Jianlin Zhang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100000, PR China.
| | - Ye Li
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, 999077, Hong Kong.
| | - Lixiang Xue
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100000, PR China.
| | - Zhen Tan
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, PR China.
| | - Deli Wang
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen 518000, PR China.
| |
Collapse
|
4
|
Zeng Y, Xiao J, Shi L, Li Y, Xu Y, Zhou J, Dong X, Hou H, Zhong C, Cheng G, Chen Y, Zhang N, Fang Y, Hu Y. Discovery of 2,4-quinazolinedione derivatives as LC3B recruiters in the facilitation of protein complex degradations. Eur J Med Chem 2025; 287:117293. [PMID: 39923533 DOI: 10.1016/j.ejmech.2025.117293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/11/2025]
Abstract
Targeted protein degradation through autophagosome-tethering compounds (ATTECs) that bypasses the ubiquitination process has garnered increasing attention. LC3B, a key protein in autophagosome formation, recruits substrates into the autophagy-lysosome system for degradation. In this study, we systematically optimized 2,4-quinazolinedione derivatives as LC3B-recruiting fragments, utilizing the CDK9 indicator. By attaching the designed LC3B-recruiting fragment to CDK9 inhibitor SNS-032 through a linker, the resulting bifunctional ATTEC molecule simultaneously degraded CDK9 and its associated Cyclin T1. Two-dimensional NMR experiments confirmed the direct interaction between the novel LC3B-recruiting fragments and LC3B. Mechanistic studies elucidated that degradation occurred via an LC3B-dependent autophagy-lysosomal pathway. Additionally, the general applicability of leveraging LC3B-recruiting fragments linked to inhibitors for the targeted degradation of protein complexes was validated with PRC2 and CDK2/4/6 along with their respective Cyclins. This work provides a series of novel LC3B-recruiting fragments that enrich the ATTEC toolbox and can be applied to the degradation of diverse intracellular disease-causing proteins.
Collapse
Affiliation(s)
- Yanping Zeng
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, 1 Xiangshanzhi Road, Hangzhou, 310024, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China
| | - Jian Xiao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 110039, China
| | - Li Shi
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China
| | - Yangsha Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 110039, China
| | - Yuanxin Xu
- Nanjing University of Chinese Medicine, School of Chinese Materia Medica, 138 Xianlin Road, Nanjing, 210046, China
| | - Jiayun Zhou
- School of Life Sciences, Fudan University (Jiangwan Campus), 2005 Songhu Road, Yangpu District, Shanghai, 200433, China
| | - Xiao Dong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 110039, China
| | - Haiyang Hou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 110039, China
| | - Chao Zhong
- School of Life Sciences, Fudan University (Jiangwan Campus), 2005 Songhu Road, Yangpu District, Shanghai, 200433, China
| | - Gang Cheng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Yi Chen
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 110039, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China
| | - Naixia Zhang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China.
| | - Yanfen Fang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China.
| | - Youhong Hu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, 1 Xiangshanzhi Road, Hangzhou, 310024, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 ZuChongZhi Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 110039, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China.
| |
Collapse
|
5
|
Huang H, Ding C, Zhao WH, Zhang HB, Zhao ZX, Li XG, Wang YJ, Chen PJ, Li BS, Li XB, Li YW, Liu HY, Chen J. Nicotine promotes the progression and metastasis of non-small cell lung cancer by modulating the OTUB1-c-Myc-EZH2 axis. Acta Pharmacol Sin 2025:10.1038/s41401-025-01527-5. [PMID: 40169782 DOI: 10.1038/s41401-025-01527-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 02/26/2025] [Indexed: 04/03/2025]
Abstract
Smoking has been identified as a major risk factor for the development and progression of non-small cell lung cancer (NSCLC). As a key component of tobacco smoke, nicotine is believed to play a significant role in promoting NSCLC growth and progression. EZH2 is an epigenetic regulator highly expressed in the tumor tissues of smokers. However, whether and how nicotine regulates the expression of EZH2 and the underlying mechanisms remain unclear. Bioinformatics analysis and immunohistochemistry were used to compare the expression of EZH2 in NSCLC samples between smokers and nonsmokers. Western blotting, real-time quantitative PCR, and immunofluorescence were employed to confirm the effects of nicotine on EZH2 expression. Cell Counting Kit-8 assays, colony formation assays, 5-ethynyl-2-deoxyuridine staining, and Transwell assays were conducted to analyze the proliferation and metastasis of A549 and H1650 cells treated with siRNA or EZH2 inhibitors. Real-time quantitative PCR and chromatin immunoprecipitation assays were performed to assess the regulatory effect of nicotine on EZH2 transcript levels via c-Myc. Coimmunoprecipitation and ubiquitination assays were used to assess the deubiquitination of c-Myc by OTUB1. Finally, a nude mouse model was used to evaluate the impact of combined c-Myc and EZH2 inhibitors on tumor proliferation and metastasis in vivo. EZH2 is expressed at relatively high levels in NSCLC patients, as determined by both bioinformatic and IHC analyses. Nicotine upregulates EZH2 expression and promotes the proliferation and metastatic ability of lung cancer cells. Inhibition of EZH2 with either DZNep or EPZ6438, EZH2 inhibitors, or siRNA significantly decreased the proliferative and metastatic capacity of NSCLC cells induced by nicotine treatment. Moreover, the study revealed that nicotine induces OTUB1 expression, stabilizes the c-Myc protein via deubiquitination, and enables c-Myc-mediated transcriptional activation of EZH2. Furthermore, the c-Myc inhibitor 10058-F4 exhibited synergistic effects with the EZH2 inhibitor DZNep in suppressing NSCLC cell proliferation and metastasis both in vitro and in vivo.Nicotine regulates the c-Myc/EZH2 signaling pathway via OTUB1-mediated deubiquitination, thereby promoting the proliferation and metastasis of NSCLC cells. This research reveals novel molecular mechanisms of nicotine in the development of NSCLC, providing a theoretical foundation for future therapeutic strategies.
Collapse
Affiliation(s)
- Hua Huang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Chen Ding
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Wen-Hao Zhao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hong-Bing Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Ze-Xia Zhao
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xuan-Guang Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Ying-Jie Wang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Pei-Jie Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Bo-Shi Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xue-Bing Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yong-Wen Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hong-Yu Liu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
6
|
Guo Y, Huang J, Lin M, Yin Q, Zhang T, Guo Z, Tang Y, Cheng R, Wang Y, Peng Y, Cao X, Wang Y, Qi X, Liu Y, Xue L. Nano particle loaded EZH2 inhibitors: Increased efficiency and reduced toxicity for malignant solid tumors. J Transl Int Med 2025; 13:156-169. [PMID: 40443399 PMCID: PMC12116265 DOI: 10.1515/jtim-2025-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2025] Open
Abstract
Background and Objectives Aberrant upregulation or mutations of EZH2 frequently occur in human cancers. However, the clinical benefits of EZH2 inhibitors (EZH2i) remain unsatisfactory for majority of solid tumors. Therefore, there is an urgent need to develop new strategies to expand the therapeutic benefits of EZH2i. Nanocarriers have gained increased attention due to their advantages of prolonged blood circulation, enhanced cellular uptake, and active targeting capabilities. This study aims to address the challenges of EZH2i GSK126's limited efficacy and severe adverse effects against solid tumors. Methods A nano delivery system was developed by encapsulating GSK126 within albumin nanoparticles (GSK126 NPs). Results The prepared GSK126 NPs exhibited a small spherical core with an average diameter of 30.09 nm ± 1.55 nm, high drug loading capacity (16.59% ± 2.86%) and good entrapment efficiency (99.53% ± 0.208%). GSK126 NPs decreased tumor weight and volume in the B16F10 xenograft mice, while such effects were not observed in the free GSK126 group. Subsequently, histological analysis demonstrated that GSK126 NPs significantly alleviated lipid-associated liver toxicity. Additionally, GSK126 NPs can partially counteract the effects of GSK126 on MDSCs, particularly by decreasing the infiltration of M-MDSCs into tumors. Conclusions Albumin-based EZH2i NPs have potent anti-cancer efficacy with tolerable adverse effects, providing promising opportunity for future clinical translation in treating solid tumors.
Collapse
Affiliation(s)
- Yunyun Guo
- Cancer Center of Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Peking University Third Hospital, Beijing, China
| | - Jiaqi Huang
- Department of Radiation, Peking University People's Hospital, Beijing, China
| | - Meng Lin
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qianqian Yin
- Biobank of Peking University Third Hospital, Beijing, China
| | - Tengrui Zhang
- Cancer Center of Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Peking University Third Hospital, Beijing, China
| | - Zhengyang Guo
- Cancer Center of Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Peking University Third Hospital, Beijing, China
| | - Yuanjun Tang
- Cancer Center of Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Peking University Third Hospital, Beijing, China
| | - Rui Cheng
- Cancer Center of Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Peking University Third Hospital, Beijing, China
| | - Yan Wang
- Cancer Center of Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Peking University Third Hospital, Beijing, China
| | - Yiwei Peng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xuedi Cao
- Cancer Center of Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Peking University Third Hospital, Beijing, China
| | - Yuqing Wang
- Cancer Center of Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Peking University Third Hospital, Beijing, China
| | - Xianrong Qi
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yang Liu
- Cancer Center of Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Peking University Third Hospital, Beijing, China
| | - Lixiang Xue
- Cancer Center of Peking University Third Hospital, Beijing, China
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Peking University Third Hospital, Beijing, China
| |
Collapse
|
7
|
Li Z, Zhu T, Wu Y, Yu Y, Zang Y, Yu L, Zhang Z. Functions and mechanisms of non-histone post-translational modifications in cancer progression. Cell Death Discov 2025; 11:125. [PMID: 40164592 PMCID: PMC11958777 DOI: 10.1038/s41420-025-02410-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/19/2025] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
Protein post-translational modifications (PTMs) refer to covalent and enzymatic alterations to folded or nascent proteins during or after protein biosynthesis to alter the properties and functions of proteins. PTMs are modified in a variety of types and affect almost all aspects of cell biology. PTMs have been reported to be involved in cancer progression by influencing multiple signaling pathways. The mechanism of action of histone PTMs in cancer has been extensively studied. Notably, evidence is mounting that PTMs of non-histone proteins also play a vital role in cancer progression. In this review, we provide a systematic description of main non-histone PTMs associated with cancer progression, including acetylation, lactylation, methylation, ubiquitination, phosphorylation, and SUMOylation, based on recent studies.
Collapse
Affiliation(s)
- Zongyang Li
- Department of Urology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, 261041, China
- School of Clinical Medicine, Shandong Second Medical University, Weifang, 261041, China
| | - Tao Zhu
- Department of Urology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, 261041, China
| | - Yushu Wu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, 261041, China
| | - Yongbo Yu
- Department of Urology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, 261041, China
| | - Yunjiang Zang
- Department of Urology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, 261041, China
| | - Lebo Yu
- Department of Urology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, 261041, China
| | - Zhilei Zhang
- Department of Urology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, 261041, China.
| |
Collapse
|
8
|
Wan G, Li S, Tang Q, Qiu H, Zhang Q, Yu L. An updated patent review of EZH2 inhibitors (2024-present). Expert Opin Ther Pat 2025:1-14. [PMID: 40116819 DOI: 10.1080/13543776.2025.2483399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 03/23/2025]
Abstract
INTRODUCTION EZH2 forms the PRC2 complex with SUZ12 and EED. As a crucial catalytic subunit of PRC2, EZH2 modifies histone H3K27 via its SET domain, resulting in chromatin condensation and suppressing the transcription of related target genes. EZH2 not only functions in PRC2-dependent transcriptional repression but can also activate gene expression in PRC2-independent circumstances or regulate the activity of downstream genes via its own activating mutations. On the basis of the critical role of EZH2 in cancer, the development of inhibitors targeting EZH2 provides a new strategy for cancer therapy. AREAS COVERED The purpose of this review is to summarize the molecular mechanisms of EZH2 inhibitors and emphasize the research progress on EZH2 inhibitors published in the patent literature in recent years. The literature and patent databases of PubMed, Web of Science, SCIFinder, WIPO, USPTO, EPO, and CNIPA were combined to search for more effective EZH2 inhibitors. EXPERT OPINION Recently, a wide range of structurally diverse EZH2 inhibitors, particularly EZH2 degraders, have been identified. These EZH2 modulators have demonstrated significant potential in treating various diseases, with cancer being a primary focus. The development of small molecules targeting EZH2 with distinct pharmacological effects is poised with numerous opportunities.
Collapse
Affiliation(s)
- Guoquan Wan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Siyan Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Qifan Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Huapei Qiu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| | - Qiangsheng Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Luoting Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Jiang M, Huang F, Hong X, Xu C, Zhang B, Hu S, Wang G, Hu D, Sun W, Lu Q, Liu H, Cai D, Yang X, Lin T, Chen S. PQQ Inhibits PRC2 Methyltransferase Activity and Suppresses the Proliferation of B-Cell Lymphoma In Vitro. Chem Biodivers 2025:e202500198. [PMID: 40024903 DOI: 10.1002/cbdv.202500198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/04/2025]
Abstract
Polycomb repressive complex 2 (PRC2) is a multi-subunit complex that catalyzes the tri-methylation of histone H3 at lysine 27 (H3K27me3), serving as an epigenetic marker of gene silencing. PRC2 plays a crucial role in numerous fundamental biological processes, and its dysregulation is closely linked to cancer and developmental disorders. EZH2, a key component of PRC2, is aberrantly overexpressed in various human cancers. Inhibition of EZH2 enzymatic activity has been shown to effectively reduce cancer cell proliferation and tumorigenesis. Consequently, EZH2 is widely recognized as a driver of cancer, and the development of EZH2-specific inhibitors has become an active area of research. In this study, we screened over 2000 compounds from solid libraries using a PRC2 enzymatic activity assay and identified pyrroloquinoline quinone (PQQ) as a potent inhibitor of PRC2 methyltransferase activity in vitro. We evaluated the antitumor effects of PQQ across different tumor cell lines and found that it exhibited strong anticancer activity, specifically against B-cell lymphoma cells, which demonstrate elevated EZH2 activity. We used a combination of biochemical assays, cellular assays, and molecular docking studies to thoroughly investigate the inhibitory effects of PQQ on PRC2 activity. Furthermore, PQQ is a naturally occurring compound with various biological activities, including antioxidant and neuroprotective effects, and it has been approved as a nutritional supplement and health product in the United States. This study demonstrates, for the first time, that PQQ, a dietary supplement, selectively inhibits PRC2 methyltransferase activity, therefore providing new insights for targeted anti-lymphoma therapies involving PRC2.
Collapse
Affiliation(s)
- Meizhi Jiang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Fangfang Huang
- Department of Hematology, School of Medicine, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Xiuli Hong
- Department of Hematology, School of Medicine, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Chenyu Xu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Bin Zhang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Shengwei Hu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Guijiang Wang
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Die Hu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Wenxin Sun
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Quanyi Lu
- Department of Hematology, School of Medicine, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Huiheng Liu
- Emergency Department of Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Dachuan Cai
- Department of Infectious Diseases, Institute for Viral Hepatitis, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Xianwen Yang
- Hainan Academy of Medical Sciences, Hainan Medical University, Haikou, China
| | - Ting Lin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Siming Chen
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
- Shenzhen Research Institute of Xiamen University, Shenzhen, Guangdong, China
| |
Collapse
|
10
|
Kumari P, Garg S, Arya AK, Kaur J, Sachdeva NK, Saikia UN, Dahiya D, Bhadada SK, Rao SD. Enhancer of zeste homolog 2 (EZH2) in endocrine tumors: current knowledge and future directions. Expert Opin Ther Targets 2025; 29:159-169. [PMID: 40111060 DOI: 10.1080/14728222.2025.2482555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/06/2025] [Accepted: 03/18/2025] [Indexed: 03/22/2025]
Abstract
INTRODUCTION Enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase that orchestrates gene expression via epigenetic and non-epigenetic mechanisms. EZH2 performs epigenetic functions by methylating histones and/or non-histone proteins and suppressing or activating target genes. Moreover, non-epigenetic functions involve dysregulation of target genes independent of histone methylation, thereby impacting multiple signaling pathways. AREAS COVERED EZH2 has emerged as a pivotal player in the initiation of various cancers. EZH2 overexpression facilitated by H3K27me3 is the principal driver. However, the consequent dysregulation of target genes resulting from EZH2 overexpression has emerged as a secondary instigator of tumorigenesis, leading to metastasis and poor prognosis. Further complexity arises from somatic mutations in EZH2 and downstream target genes such as BRAF and RASSF1A. However, understanding its effects on endocrine tumors/cancers remains an underexplored with the potential to significantly enhance clinical outcomes and contribute to human health. Therefore, the present review focuses on the multifaceted functions of EZH2 and its pathophysiological mechanisms in tumor proliferation, with a specific emphasis on endocrine tumors. EXPERT OPINION Investigating EZH2 mechanisms and targeting with inhibitors and drugs is an active area of research that could offer a promising avenue for treatment and a better understanding of molecular therapeutic interventions.
Collapse
Affiliation(s)
- Poonam Kumari
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Sheenam Garg
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Ashutosh Kumar Arya
- Department of Endocrinology & Metabolism, All India Institute of Medical Sciences, New Delhi, India
| | - Jyotdeep Kaur
- Department of Biochemistry, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Naresh Kumar Sachdeva
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Uma Nahar Saikia
- Department of Histopathology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Divya Dahiya
- Department of General Surgery, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Sanjay Kumar Bhadada
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Sudhaker D Rao
- Bone and Mineral Research Laboratory, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
11
|
He J, Wang L, Lv M, Yuan Y. GGCT participates in the malignant process of hepatocellular cancer cells by regulating the PTEN/PI3K/AKT pathway through binding to EZH2. Discov Oncol 2025; 16:129. [PMID: 39918720 PMCID: PMC11806167 DOI: 10.1007/s12672-025-01882-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 02/03/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND γ-Glutamylcyclotransferase (GGCT) is implicated in multiple types of cancer diseases. Nevertheless, the roles and relevant mechanisms of GGCT in hepatocellular carcinoma (HCC) remain vague. METHODS GGCT expression in HCC and its effect on patient survival curve in HCC were evaluated utilizing the UALCAN database, along with western blot. CCK-8, EdU, and wound healing, together with transwell and western blot assays were adopted to assess the capabilities of cells to proliferate, migrate, invade, and epithelial-mesenchymal transition (EMT). Cell apoptosis was appraised utilizing TUNEL as well as western blot. Glycolysis was measured by western blot and kits. Enhancer of zeste homolog 2 (EZH2) expression in HCC cells was detected by western blot. Co-IP verified the combination of GGCT and EZH2. Moreover, PI3K/AKT pathway-related proteins were assessed employing western blot. RESULTS GGCT expression was conspicuously upregulated in HCC samples and HCC cells. GGCT silencing repressed HuH-7 cell proliferative, invasive, and migratory capabilities as well as EMT, whereas facilitated cell apoptosis. In addition, GGCT silencing inhibited PTEN/PI3K/AKT pathway-mediated glycolysis. EZH2 was highly expressed in HCC cells and the interaction of GGCT and EZH2 was verified. Overexpression of EZH2 reversed the effects of GGCT silencing on HuH-7 cell proliferation, migration, invasion, cell apoptosis, and glycolysis. Moreover, the PTEN inhibitor SF1670 reversed the effects of GGCT silencing and EZH2 overexpression on the glycolysis and malignant process in HuH-7 cells. CONCLUSION In conclusion, GGCT silencing restrained the proliferation and metastasis, and promoted apoptotic levels of HCC cells via regulating PTEN/PI3K/AKT pathway-mediated glycolysis, which might offer a prospective candidate in treating HCC.
Collapse
Affiliation(s)
- Junbo He
- Department of General Practice, The Third Affiliated Hospital of Soochow University, No. 185, Jiuqian Street, Changzhou, 213003, Jiangsu, China
| | - Liangzhi Wang
- Department of General Practice, The Third Affiliated Hospital of Soochow University, No. 185, Jiuqian Street, Changzhou, 213003, Jiangsu, China
| | - Mengjia Lv
- Department of General Practice, The Third Affiliated Hospital of Soochow University, No. 185, Jiuqian Street, Changzhou, 213003, Jiangsu, China
| | - Yiming Yuan
- Department of General Practice, The Third Affiliated Hospital of Soochow University, No. 185, Jiuqian Street, Changzhou, 213003, Jiangsu, China.
| |
Collapse
|
12
|
Bao Y, Xu Z, Cheng K, Li X, Chen F, Yuan D, Zhang F, Che ARY, Zeng X, Zhao YD, Xia J. Staudinger Reaction-Responsive Coacervates for Cytosolic Antibody Delivery and TRIM21-Mediated Protein Degradation. J Am Chem Soc 2025; 147:3830-3839. [PMID: 39805770 PMCID: PMC11783599 DOI: 10.1021/jacs.4c17054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
A low-molecular-weight compound whose structure strikes a fine balance between hydrophobicity and hydrophilicity may form coacervates via liquid-liquid phase separation in an aqueous solution. These coacervates may encapsulate and convoy proteins across the plasma membrane into the cell. However, releasing the cargo from the vehicle to the cytosol is challenging. Here, we address this issue by designing phase-separating coacervates, which are disassembled by the bioorthogonal Staudinger reaction. We constructed and selected triphenylphosphine-based compounds that formed phase-separated coacervates in an aqueous solution. Reacting the coacervates with azides resulted in microdroplet dissolution, so they received the name Staudinger Reaction-Responsive Coacervates, SR-Coa. SR-Coa could encapsulate proteins, including antibodies, and translocate them across the plasma membrane into the cell. Further treatment of the cell with ethyl azidoacetate induced the cargo dispersion from the puncta to the cytosolic distribution. We showcased an application of the SR-Coa/ethyl azidoacetate system in facilitating the translocation of the EGFR/antibody complex into the cell, which induced EGFR degradation via the TRIM21-dependent pathway both in vitro and in vivo. Besides the membrane protein EGFR, this system could also degrade endogenous protein EZH2. Taken together, here we report a strategy of controlling molecular coacervates by a bioorthogonal reaction in the cell for cytosolic protein delivery and demonstrate its use in promoting targeted protein degradation via the proteasome-dependent pathway.
Collapse
Affiliation(s)
- Yishu Bao
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, Hong Kong SAR 99999, China
| | - Zhiyi Xu
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, Hong Kong SAR 99999, China
| | - Kai Cheng
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, Hong Kong SAR 99999, China
| | - Xiaojing Li
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, Hong Kong SAR 99999, China
| | - Fangke Chen
- Department
of Physics, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR 99999, China
| | - Dingdong Yuan
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, Hong Kong SAR 99999, China
| | - Fang Zhang
- Britton
Chance Center for Biomedical Photonics at Wuhan National Laboratory
for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key
Laboratory, Department of Biomedical Engineering, College of Life
Science and Technology, Huazhong University
of Science and Technology, Wuhan 430074, Hubei, China
| | - Audrey Run-Yu Che
- Department
of Natural Sciences, Pitzer and Scripps
Colleges, 925 N. Mills
Ave, Claremont, California 91711, United States
| | - Xiangze Zeng
- Department
of Physics, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR 99999, China
| | - Yuan-Di Zhao
- Britton
Chance Center for Biomedical Photonics at Wuhan National Laboratory
for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key
Laboratory, Department of Biomedical Engineering, College of Life
Science and Technology, Huazhong University
of Science and Technology, Wuhan 430074, Hubei, China
| | - Jiang Xia
- Department
of Chemistry, The Chinese University of
Hong Kong, Shatin, Hong Kong SAR 99999, China
| |
Collapse
|
13
|
Cao R, Ni J, Zhang X, Qi M, Zhang Z, Liu Z, Chen G. Recent Advances in enhancer of zeste homolog 2 Inhibitors: Structural insights and therapeutic applications. Bioorg Chem 2025; 154:108070. [PMID: 39709735 DOI: 10.1016/j.bioorg.2024.108070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/11/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024]
Abstract
Enhancer of Zeste Homolog 2 (EZH2) is overexpressed in many malignancies and plays a critical role in cancer progression. Therefore, it is considered a promising target for therapeutic intervention. Although several EZH2 inhibitors have entered clinical trials, only one has received FDA approval. In this review, we focus on the latest advancements in highly selective and potent dual-targeting EZH2 inhibitors, as well as proteolysis-targeted chimeras (PROTACs) and hydrophobic tagging (HYT) degraders. These novel compounds have been developed to address the existing gaps in the management of abnormal EZH2 expression. Notably, EZH2 inhibitors have shown great efficacy in antitumor therapy and have also demonstrated promising results in antiviral, anti-inflammatory, antisclerotic, bone protection, and nerve injury pain applications. The insights gained from this analysis could provide valuable guidance for future drug design and optimization of EZH2 inhibitors, potentially expediting the discovery of new inhibitors or degraders targeting EZH2.
Collapse
Affiliation(s)
- Ruolin Cao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Jiayang Ni
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xiaoyu Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Minggang Qi
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zhen Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zhongbo Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
14
|
Wang Z, Guo Z, Wang X, Chai Y, Wang Z, Liao H, Chen F, Xia Y, Wang X, Wang Z. EZH2 contributes to sepsis-induced acute lung injury through regulating macrophage polarization. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167554. [PMID: 39471914 DOI: 10.1016/j.bbadis.2024.167554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 09/08/2024] [Accepted: 10/24/2024] [Indexed: 11/01/2024]
Abstract
BACKGROUND Zeste enhancer homolog 2 (EZH2) is a pivotal regulator of gene dynamics implicated in the progression of sepsis-induced acute lung injury (SALI). EZH2 regulates aberrant inflammatory and immune responses in macrophages via unconventional biochemical interactions. However, the mechanisms driving atypical behavior of EZH2 during sepsis remain elusive, and therapeutic strategies targeting EZH2 are currently underutilized. PURPOSE This study aimed to investigate how EZH2 regulates macrophage polarization through the AKT pathway to improve SALI and to explore therapeutic drugs targeting EZH2. METHODS We used Western blotting, hematoxylin-eosin stainin, immunofluorescence, flow cytometry, qRT-PCR, RNA sequencing, and chromatin immunoprecipitation sequencing methods to investigate regulation of macrophage immune response by EZH2 and explored its specific mechanism. These methods were also used to examine the protective effects of MS177 against SALI. RESULTS Specific deletion of EZH2 in macrophages reduced the level of AKTIP, downregulated the M1 macrophage markers CD86 and cytotoxic T cell marker CD8+, upregulated the M2 macrophage marker CD206 and regulatory T cell marker FOXP3+, decreased the levels of pro-inflammatory cytokines IL-6, TNF-α, and IL-β, and increased the level of anti-inflammatory cytokine IL-10. This ultimately improved lung injury and mortality in SALI mice. EZH2 promoted the M1 polarization of macrophages by activating AKT2 via direct binding to the promoter region of AKTIP in a SALI mouse model. Furthermore, MS177 alleviated SALI by degrading EZH2 both in vitro and in vivo. CONCLUSION EZH2 regulates macrophage polarization via the AKTIP-AKT2 pathway. Our findings suggest that MS177 is a promising therapeutic agent for EZH2-dependent SALI.
Collapse
Affiliation(s)
- Ziyi Wang
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, PR China; Department of Anaesthesiology, Peking University First Hospital, Beijing, PR China
| | - Zhe Guo
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, PR China
| | - Xuesong Wang
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, PR China
| | - Yan Chai
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, PR China
| | - Ziwen Wang
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, PR China
| | - Haiyan Liao
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, PR China
| | - Feng Chen
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, PR China
| | - Yuxiang Xia
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, PR China
| | - Xinrui Wang
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, PR China
| | - Zhong Wang
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, PR China.
| |
Collapse
|
15
|
Feng Q, Yu L, Li L, Zhang Q. Covalent inhibitors meet epigenetics: New opportunities. Eur J Med Chem 2024; 280:116951. [PMID: 39406112 DOI: 10.1016/j.ejmech.2024.116951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/09/2024] [Accepted: 09/23/2024] [Indexed: 11/25/2024]
Abstract
Epigenetic intervention has become an important therapeutic strategy for a variety of diseases, such as cancer. Although a small number of epigenetic drugs have been marketed, most of these inhibitors are limited by their poor efficacy, dose-dependent toxicity, poor selectivity, and drug resistance. The development of covalent inhibitors has progressed from questioning to resurgence. Its slow dissociation is expected to inject new vitality into epigenetic drugs. In this review, more than 40 covalent inhibitors of 29 epigenetic targets were collated, focusing on their design strategies, reaction mechanisms, covalent warheads and targeted amino acids, and covalent verification methods. Furthermore, this review presented new opportunities based on the current development of covalent inhibitors targeting epigenetic regulators. It is believed that epigenetic covalent inhibitors will lead to more breakthroughs.
Collapse
Affiliation(s)
- Qiang Feng
- College of Chemistry and Life Science, Sichuan Provincial Key Laboratory for Structural Optimization and Application of Functional Molecules, Chengdu Normal University, Chengdu, 611130, China
| | - Luoting Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, And Collaborative Innovation Center for Biotherapy, 17#3rd Section, Ren Min South Road, Chengdu 610041, China
| | - Lu Li
- Department of Pharmacy, NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Clinical Trial Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qiangsheng Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, And Collaborative Innovation Center for Biotherapy, 17#3rd Section, Ren Min South Road, Chengdu 610041, China; Department of Pharmacy, West China Second University Hospital, Sichuan University, Children's Medicine Key Laboratory of Sichuan Province, Chengdu, 610041, China.
| |
Collapse
|
16
|
Gilardini Montani MS, Benedetti R, Cirone M. Targeting EZH2 in Cancer: Mechanisms, Pathways, and Therapeutic Potential. Molecules 2024; 29:5817. [PMID: 39769907 PMCID: PMC11678268 DOI: 10.3390/molecules29245817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Enhancer of zeste homolog 2 (EZH2) is a methyltransferase involved in cell cycle regulation, cell differentiation, and cell death and plays a role in modulating the immune response. Although it mainly functions by catalyzing the tri-methylation of H3 histone on K27 (H3K27), to inhibit the transcription of target genes, EZH2 can directly methylate several transcription factors or form complexes with them, regulating their functions. EZH2 expression/activity is often dysregulated in cancer, contributing to carcinogenesis and immune escape, thereby representing an important target in anti-cancer therapy. This review summarizes some of the mechanisms through which EZH2 regulates the expression and function of tumor suppressor genes and oncogenic molecules such as STAT3, mutant p53, and c-Myc and how it modulates the anti-cancer immune response. The influence of posttranslational modifications on EZH2 activity and stability and the possible strategies leading to its inhibition are also reviewed.
Collapse
Affiliation(s)
| | | | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| |
Collapse
|
17
|
Zhang J, Yang M, Liu Q, Xue X, Tian S, Hu X, Li M, Li J, Chai Q, Liu F, You X, Zhang Y. Discovery of epigenetic modulators targeting HDACs and EZH2 simultaneously for the treatment of hematological malignancies. Bioorg Chem 2024; 153:107964. [PMID: 39571304 DOI: 10.1016/j.bioorg.2024.107964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/01/2024] [Accepted: 11/13/2024] [Indexed: 12/14/2024]
Abstract
Epigenetic-targeted therapy has been applied in the treatment of several types of cancer. Herein, based on the synergistic antitumor effects of co-targeting HDACs and EZH2 in some hematological malignancies, a novel series of tazemetostat-based HDACs/EZH2 dual inhibitors were rationally designed, synthesized, and biologically evaluated. Satisfyingly, compounds 22a and 22b were identified as potent HDACs/EZH2 dual inhibitors with robust antiproliferative activities against one diffuse large-cell B cell lymphomas (DLBCL) cell line harboring EZH2 mutation and multiple acute myeloid leukemia (AML) cell lines. Notably, after a short-term treatment in the EZH2 mutant DLBCL cell line (SU-DHL-6), 22a and 22b displayed much stronger antiproliferative activities than the approved EZH2 inhibitor tazemetostat, while after a long-term treatment in SU-DHL-6 cells, 22a and 22b displayed comparable or even superior antiproliferative activities to the approved HDACs inhibitor SAHA. In AML cells, compounds 22a and 22b displayed much more potent antiproliferative activities than tazemetostat, as well as distinctive differentiation-inducing abilities and superior apoptosis-inducing abilities relative to tazemetostat and SAHA. Moreover, the synergistic anti-AML effects of HDACs/EZH2 dual inhibitors combined with various anti-AML drugs were demonstrated.
Collapse
Affiliation(s)
- Jinwei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China
| | - Maoshuo Yang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China
| | - Qian Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China
| | - Xintong Xue
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China
| | - Sijia Tian
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China
| | - Xi Hu
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China
| | - Mengzhe Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China
| | - Jintao Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China
| | - Qipeng Chai
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China
| | - Fabao Liu
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China.
| | - Xiaona You
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China.
| | - Yingjie Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong 250012, PR China.
| |
Collapse
|
18
|
Yang J. Emerging roles of long non-coding RNA FOXP4-AS1 in human cancers: From molecular biology to clinical application. Heliyon 2024; 10:e39857. [PMID: 39539976 PMCID: PMC11558633 DOI: 10.1016/j.heliyon.2024.e39857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Forkhead box P4 antisense RNA 1 (FOXP4-AS1) is a long non-coding RNA (lncRNA) situated on the human chromosome 6p21.1 locus. Previous research has demonstrated that FOXP4-AS1 is dysregulated in various cancers and exhibits a dual purpose as a tumor suppressor or oncogene in specific types of cancer. The levels of FOXP4-AS1 are significantly correlated with clinical features of cancer as well as prognosis. Additionally, FOXP4-AS1 is stimulated by transcription factors ATF3, YY1, PAX5, and SP4. The molecular mechanisms of FOXP4-AS1 in cancer are quite complex. It competitively sponges multiple miRNAs, bidirectionally regulates the levels of host gene FOXP4, activates the PI3K/AKT, Wnt/β-catenin, and ERK/MAPK signaling pathways, and recruits chromatin-modifying enzymes or interacts with other proteins to regulate malignant phenotypes of tumors, including proliferation, invasion, epithelial-mesenchymal transition (EMT), and angiogenesis. In this review, we provide an overview of the latest developments in FOXP4-AS1 oncology research, outlines its molecular regulatory networks in cancer, and discusses its prospective relevance as a cancer therapeutic target as well as a biomarker for prognosis and diagnosis.
Collapse
Affiliation(s)
- Jingjie Yang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, 443002, China
- College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| |
Collapse
|
19
|
Li X, Song Y. Perspective for Drug Discovery Targeting SARS Coronavirus Methyltransferases: Function, Structure and Inhibition. J Med Chem 2024; 67:18642-18655. [PMID: 39478665 PMCID: PMC11787806 DOI: 10.1021/acs.jmedchem.4c01749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Severe acute respiratory syndrome-associated coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), is highly contagious and caused a catastrophic pandemic. It has infected billions of people worldwide with >6 million deaths. With expedited development of effective vaccines and antiviral drugs, there have been significantly reduced SARS-CoV-2 infections and associated mortalities and morbidities. The virus is closely related to SARS-CoV, which emerged in 2003 and infected several thousand people with a higher mortality rate of ∼10%. Because of continued viral evolution and drug-induced resistance, as well as the possibility of a new coronavirus in the future, studies for new therapies are needed. The viral methyltransferases play critical roles in SARS coronavirus replication and are therefore promising drug targets. This review summarizes the function, structure and inhibition of methyltransferases of SARS-CoV-2 and SARS-CoV. Challenges and perspectives of targeting the viral methyltransferases to treat viral infections are discussed.
Collapse
Affiliation(s)
- Xin Li
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Yongcheng Song
- Verna and Marrs McLean Department of Biochemistry and Molecular Pharmacology
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
20
|
Zhang Y, Yu Y, Yuan L, Zhang B. EZH2 Promotes Glioma Cell Proliferation, Invasion, and Migration via Mir-142-3p/KCNQ1OT1/HMGB3 Axis : Running Title: EZH2 Promotes Glioma cell Malignant Behaviors. Mol Neurobiol 2024; 61:8668-8687. [PMID: 38556567 DOI: 10.1007/s12035-024-04080-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/28/2024] [Indexed: 04/02/2024]
Abstract
This study investigates the role and molecular mechanism of EZH2 in glioma cell proliferation, invasion, and migration. EZH2, miR-142-3p, lncRNA KCNQ1OT1, LIN28B, and HMGB3 expressions in glioma tissues and cells were determined using qRT-PCR or Western blot, followed by CCK-8 assay detection of cell viability, Transwell detection of invasion and migration, ChIP analysis of the enrichment of EZH2 and H3K27me3 on miR-142-3p promoter, dual-luciferase reporter assay and RIP validation of the binding of miR-142-3p-KCNQ1OT1 and KCNQ1OT1-LIN28B, and actinomycin D detection of KCNQ1OT1 and HMGB3 mRNA stability. A nude mouse xenograft model and a lung metastasis model were established. EZH2, KCNQ1OT1, LIN28B, and HMGB3 were highly expressed while miR-142-3p was poorly expressed in gliomas. EZH2 silencing restrained glioma cell proliferation, invasion, and migration. EZH2 repressed miR-142-3p expression by elevating the H3K27me3 level. miR-142-3p targeted KCNQ1OT1 expression, and KCNQ1OT1 bound to LIN28B to stabilize HMGB3 mRNA, thereby promoting its protein expression. EZH2 silencing depressed tumor growth and metastasis in nude mice via the miR-142-3p/KCNQ1OT1/HMGB3 axis. In conclusion, EZH2 curbed miR-142-3p expression, thereby relieving the inhibition of KCNQ1OT1 expression by miR-142-3p, enhancing the binding of KCNQ1OT1 to LIN28B, elevating HMGB3 expression, and ultimately accelerating glioma cell proliferation, invasion, and migration.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Neurosurgery, Beijing Fengtai You'anmen Hospital, Beijing, 100069, China
| | - Yong Yu
- Epilepsy Center, Beijing Fengtai You'anmen Hospital, Beijing, 100069, China
| | - Lei Yuan
- Department of Neurosurgery, PLA Rocket Force Characteristic Medical Center, No. 16, Xin Jie Kou Wai Street, Beijing, 100088, China.
| | - Baozhong Zhang
- Department of Neurosurgery, He Bei Hua Ao Hospital, No. 11, the Changcheng West Street, Zhangjiakou, 075000, Hebei Province, China.
| |
Collapse
|
21
|
Yuan K, Tang Y, Ding Z, Peng L, Zeng J, Wu H, Yi Q. Mutant ATRX: pathogenesis of ATRX syndrome and cancer. Front Mol Biosci 2024; 11:1434398. [PMID: 39479502 PMCID: PMC11521912 DOI: 10.3389/fmolb.2024.1434398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/04/2024] [Indexed: 11/02/2024] Open
Abstract
The transcriptional regulator ATRX, a genetic factor, is associated with a range of disabilities, including intellectual, hematopoietic, skeletal, facial, and urogenital disabilities. ATRX mutations substantially contribute to the pathogenesis of ATRX syndrome and are frequently detected in gliomas and many other cancers. These mutations disrupt the organization, subcellular localization, and transcriptional activity of ATRX, leading to chromosomal instability and affecting interactions with key regulatory proteins such as DAXX, EZH2, and TERRA. ATRX also functions as a transcriptional regulator involved in the pathogenesis of neuronal disorders and various diseases. In conclusion, ATRX is a central protein whose abnormalities lead to multiple diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Huaying Wu
- Key Laboratory of Model Animals and Stem Cell Biology, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Qi Yi
- Key Laboratory of Model Animals and Stem Cell Biology, Hunan Normal University School of Medicine, Changsha, Hunan, China
| |
Collapse
|
22
|
Jiang W, Liu L, Wang M, Li X, Zhou T, Hou X, Qiao L, Chen C, Zuo D, Liu J, Ren L. KLF15 suppresses stemness of pancreatic cancer by decreasing USP21-mediated Nanog stability. Cell Mol Life Sci 2024; 81:417. [PMID: 39367978 PMCID: PMC11455850 DOI: 10.1007/s00018-024-05442-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/08/2024] [Accepted: 08/22/2024] [Indexed: 10/07/2024]
Abstract
The existence of cancer stem cells (CSCs) in pancreatic ductal adenocarcinoma (PDAC) is considered to be the key factor for metastasis and chemoresistance. Thus, novel therapeutic strategies for eradicating CSCs are urgently needed. Here we aimed to explore the role of KLF15 in stemness and the feasibility of using KLF15 to inhibit CSCs and improve chemotherapy sensitivity in PDAC. In this study, we report that KLF15 is negatively associated with poor survival and advanced pathological staging of PDAC. Moreover, tumorous KLF15 suppresses the stemness of PDAC by promoting the degradation of Nanog, and KLF15 directly interacts with Nanog, inhibiting interaction between Nanog with USP21. We also demonstrate that the KLF15/Nanog complex inhibit the stemness in vivo and in PDX cells. Tazemetostat suppresses stemness and sensitizes PDAC cells to gemcitabine by promoting KLF15 expression in PDAC. In summary, the findings of our study confirm the value of KLF15 level in diagnosis and prognosis of PDAC, it is the first time to explore the inhibition role of KLF15 in stemness of PDAC and the regulation mechanism of Nanog, contributing to provide a new therapeutic strategy that using Tazemetostat sensitizes PDAC cells to gemcitabine by promoting KLF15 expression for PDAC.
Collapse
Affiliation(s)
- Wenna Jiang
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Lin Liu
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Meng Wang
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Xueyang Li
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Tianxing Zhou
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Xupeng Hou
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Lu Qiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences of Tianjin Medical University, Tianjin, China
| | - Chong Chen
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Duo Zuo
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China
| | - Jing Liu
- Department of Breast Oncoplastic Surgery, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
| | - Li Ren
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, Tianjin Key Laboratory of Digestive Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Hexi District, Huanhuxi Road, Tianjin, 300060, China.
| |
Collapse
|
23
|
Murga M, Lopez-Pernas G, Soliva R, Fueyo-Marcos E, Amor C, Faustino I, Serna M, Serrano AG, Díaz L, Martínez S, Blanco-Aparicio C, Antón ME, Seashore-Ludlow B, Pastor J, Jafari R, Lafarga M, Llorca O, Orozco M, Fernández-Capetillo O. SETD8 inhibition targets cancer cells with increased rates of ribosome biogenesis. Cell Death Dis 2024; 15:694. [PMID: 39341827 PMCID: PMC11438997 DOI: 10.1038/s41419-024-07106-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
SETD8 is a methyltransferase that is overexpressed in several cancers, which monomethylates H4K20 as well as other non-histone targets such as PCNA or p53. We here report novel SETD8 inhibitors, which were discovered while trying to identify chemicals that prevent 53BP1 foci formation, an event mediated by H4K20 methylation. Consistent with previous reports, SETD8 inhibitors induce p53 expression, although they are equally toxic for p53 proficient or deficient cells. Thermal stability proteomics revealed that the compounds had a particular impact on nucleoli, which was confirmed by fluorescent and electron microscopy. Similarly, Setd8 deletion generated nucleolar stress and impaired ribosome biogenesis, supporting that this was an on-target effect of SETD8 inhibitors. Furthermore, a genome-wide CRISPR screen identified an enrichment of nucleolar factors among those modulating the toxicity of SETD8 inhibitors. Accordingly, the toxicity of SETD8 inhibition correlated with MYC or mTOR activity, key regulators of ribosome biogenesis. Together, our study provides a new class of SETD8 inhibitors and a novel biomarker to identify tumors most likely to respond to this therapy.
Collapse
Affiliation(s)
- Matilde Murga
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), Madrid, 28029, Spain
| | - Gema Lopez-Pernas
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), Madrid, 28029, Spain
| | - Robert Soliva
- Nostrum Biodiscovery, Av. Josep Tarradellas 8-10, 3-2, 08029, Barcelona, Spain
| | - Elena Fueyo-Marcos
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), Madrid, 28029, Spain
| | - Corina Amor
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), Madrid, 28029, Spain
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ignacio Faustino
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028, Barcelona, Spain
| | - Marina Serna
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Alicia G Serrano
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), Madrid, 28029, Spain
| | - Lucía Díaz
- Nostrum Biodiscovery, Av. Josep Tarradellas 8-10, 3-2, 08029, Barcelona, Spain
| | - Sonia Martínez
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Carmen Blanco-Aparicio
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Marta Elena Antón
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), Madrid, 28029, Spain
| | - Brinton Seashore-Ludlow
- Department of Oncology-Pathology, Karolinska Institutet, Science for Life Laboratory, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Chemical Biology Consortium Sweden (CBCS), Science for Life Laboratory, Karolinska Institute, S-171 21, Stockholm, Sweden
| | - Joaquín Pastor
- Experimental Therapeutics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Rozbeh Jafari
- Department of Oncology-Pathology, Karolinska Institutet, Science for Life Laboratory, Stockholm, Sweden
| | - Miguel Lafarga
- Departament of Anatomy and Cell Biology, Neurodegenerative diseases network (CIBERNED), University of Cantabria-IDIVAL, Santander, Spain
| | - Oscar Llorca
- Structural Biology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Modesto Orozco
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, 08028, Barcelona, Spain
- Departament de Bioquímica i Biomedicina, Facultat de Biologia, Universitat de Barcelona, 08028, Barcelona, Spain
| | - Oscar Fernández-Capetillo
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), Madrid, 28029, Spain.
- Science for Life Laboratory, Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, S-171 21, Stockholm, Sweden.
| |
Collapse
|
24
|
Zhang Y, Zhang Y. Knockdown of USP7 alleviates atherosclerosis in ApoE-deficient mice by regulating EZH2 expression. Open Life Sci 2024; 19:20220929. [PMID: 39310812 PMCID: PMC11416069 DOI: 10.1515/biol-2022-0929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/10/2024] [Accepted: 06/24/2024] [Indexed: 09/25/2024] Open
Abstract
Atherosclerosis (AS) is a chronic vascular disease associated with lipid accumulation. Understanding the molecular mechanisms of AS is essential. Ubiquitin-specific protease 7 (USP7) is a deubiquitination enzyme involved in various cellular processes, including lipid metabolism. In this study, we aimed to elucidate the role of USP7 in AS progression and its underlying mechanism using ApoE-deficient mice. We found that USP7 ablation improved the morphological characteristics of AS in these mice. USP7 knockdown reduced inflammation, evidenced by decreases in inflammatory markers IL-6, TNF-α, and IL-1β by 35, 40, and 38%, respectively (p < 0.01). Additionally, USP7 depletion reduced oxidative stress, indicated by a 30% reduction in malondialdehyde levels and increases in superoxide dismutase and glutathione peroxidase levels by 25 and 28%, respectively (p < 0.01). Moreover, USP7 knockdown blocked lipid accumulation in aortic tissue cells. Mechanistically, USP7 knockdown inhibited enhancer of Zeste Homolog 2 (EZH2) expression, thereby suppressing AS progression. In conclusion, USP7 depletion alleviated AS progression in ApoE-deficient mice by targeting EZH2 expression. USP7 may serve as a therapeutic target for AS.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, 214000, China
| | - Yanchun Zhang
- Department of Cardiology, Shuyang Hospital of Traditional Chinese Medicine, No. 28 Shanghai Road, Shuyang, Jiangsu, 223600, China
| |
Collapse
|
25
|
Zhu J, Dai G, Chen T, Zhou Y, Zang Y, Xu L, Jin L, Zhu J. Ailanthone suppresses cell proliferation of renal cell carcinoma partially via inhibition of EZH2. Discov Oncol 2024; 15:464. [PMID: 39298003 DOI: 10.1007/s12672-024-01347-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/13/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND Ailanthone (Ail) extracted from medicinal plants has played an anticancer role in multiple cancers, while there is no research about Ail in renal cell carcinoma (RCC). METHODS In the present study, we performed CCK-8 and flow cytometry to assess the effect of Ail on cell viability, apoptosis and cycle. We also performed tandem mass tags (TMT)-labeled quantitative proteomic technology and bioinformatic analysis to identify the functional pathway and proteins of Ail in RCC. RESULTS The results showed Ail could inhibit cell viability and induce cell apoptosis. Proteomic profiling identified 1732 differentially expressed proteins in cells treated with Ail, compared to the negative control group. Gene ontology function annotation and Gene Set Enrichment Analysis (GSEA) were performed to identified the involved biological processes, molecular function and pathway. Results of GSEA proved the enrichment of Deps in EZH2 targets. The comparison between Deps and EZH2 co-expressed genes revealed 44 overlapped genes and we identified 4 hub genes (CDC20, CEP55, TOP2A, and UBE2C) associated with RCC progression. The molecular docking study revealed a moderate to tight binding potential of Ail and EZH2, and western blotting showed EZH2 was suppressed after cells treated with Ail. CONCLUSION Altogether, we identified the anticancer role of Ail in RCC, including inhibition of cell proliferation and induction of apoptosis. The results also screened the key proteins mediate the function of Ail, which have laid a theoretical foundation for elucidating the applications of Ail in clinical research.
Collapse
Affiliation(s)
- Jianbing Zhu
- Department of Radiology, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
- Key Laboratory of Functional Genomic and Molecular Diagnosis of Gansu Province, Lanzhou, Gansu, China
| | - Guangcheng Dai
- Department of Urology, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Road, Suzhou, Jiangsu, China
| | - Ting Chen
- Department of Pathology, Children's Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yibin Zhou
- Department of Urology, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Road, Suzhou, Jiangsu, China
| | - Yachen Zang
- Department of Urology, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Road, Suzhou, Jiangsu, China
| | - Lijun Xu
- Department of Urology, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Road, Suzhou, Jiangsu, China
| | - Lu Jin
- Department of Urology, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Road, Suzhou, Jiangsu, China.
| | - Jin Zhu
- Department of Urology, The Second Affiliated Hospital of Soochow University, No.1055, Sanxiang Road, Suzhou, Jiangsu, China.
| |
Collapse
|
26
|
Zhang L, Pan H, Yao Y, Gu X, Ge T, Cui J, Chai P, Xu X, Jia R, Zhuang A, Fan X. Gain of chromosome 8q and high expression of EZH2 may predict poor prognosis in Chinese patients with uveal melanoma. Asia Pac J Ophthalmol (Phila) 2024; 13:100108. [PMID: 39395714 DOI: 10.1016/j.apjo.2024.100108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 10/14/2024] Open
Abstract
PURPOSE To explore risk factors predicting poor prognosis of uveal melanoma in a Chinese population, with specific emphasis on monosomy 3, 8q gain, and EZH2 staining. METHODS Eighty-nine patients with uveal melanoma from 2012 to 2021 were reviewed. Clinical and pathological records were collected and analyzed. Immunohistochemical staining of EZH2, monosomy 3 and 8q gain were respectively conducted in 45, 54, and 57 cases. Survival was evaluated by Kaplan-Meier analysis and log-rank test. Cox proportional hazard regressions were employed to predict risk factors of distant metastasis. RESULTS The median follow-up was 44 months. Altogether, 16 % of patients developed distant metastases and died from disease-related causes. Disease-specific survival at one and three years was 96.6 % and 88.4 % while distant metastasis rates were 7.9 % and 12 %. Univariate Cox regression analysis revealed that age (HR: 1.04), tumor largest basal diameter (HR: 1.21), tumor thickness (HR: 1.21), ciliary body involvement (HR: 3.50), AJCC stage (HR: 5.68), epithelioid cell type (HR: 7.71), 8q gain (HR: 7.48), and high expression of EZH2 (HR: 6.09) were associated with distant metastasis. 8q gain was associated with epithelioid cell type and thicker tumor while EZH2 was correlated with epithelioid cell type. Monosomy 3 lacked a significant correlation with other factors. CONCLUSION EZH2 and 8q gain could be taken into consideration when calculating poor prognosis in Chinese patients with uveal melanoma. Monosomy 3 showed no significance in distant metastasis, but this may be due to a small sample size.
Collapse
Affiliation(s)
- Lingyu Zhang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, PR China
| | - Hui Pan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, PR China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, PR China
| | - Xiang Gu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, PR China
| | - Tongxin Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, PR China
| | - Junqi Cui
- Department of Pathology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China
| | - Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, PR China
| | - Xiaofang Xu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, PR China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, PR China.
| | - Ai Zhuang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, PR China.
| | - Xianqun Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, PR China; Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai 200011, PR China.
| |
Collapse
|
27
|
Leone GM, Mangano K, Caponnetto S, Fagone P, Nicoletti F. Identification of Poliovirus Receptor-like 3 Protein as a Prognostic Factor in Triple-Negative Breast Cancer. Cells 2024; 13:1299. [PMID: 39120328 PMCID: PMC11312209 DOI: 10.3390/cells13151299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/07/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024] Open
Abstract
Triple-negative breast cancer (TNBC) represents an aggressive subtype of breast cancer, with a bad prognosis and lack of targeted therapeutic options. Characterized by the absence of estrogen receptors, progesterone receptors, and HER2 expression, TNBC is often associated with a significantly lower survival rate compared to other breast cancer subtypes. Our study aimed to explore the prognostic significance of 83 immune-related genes, by using transcriptomic data from the TCGA database. Our analysis identified the Poliovirus Receptor-Like 3 protein (PVRL3) as a critical negative prognostic marker in TNBC patients. Furthermore, we found that the Enhancer of Zeste Homolog 2 (EZH2), a well-known epigenetic regulator, plays a pivotal role in modulating PVRL3 levels in TNBC cancer cell lines expressing EZH2 along with high levels of PVRL3. The elucidation of the EZH2-PVRL3 regulatory axis provides valuable insights into the molecular mechanisms underlying TNBC aggressiveness and opens up potential pathways for personalized therapeutic intervention.
Collapse
Affiliation(s)
- Gian Marco Leone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (G.M.L.); (K.M.); (F.N.)
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (G.M.L.); (K.M.); (F.N.)
| | - Salvatore Caponnetto
- Medical Oncology Unit B, Department of Radiology, Oncology and Pathology, Policlinico Umberto I, Sapienza University of Rome, 00161 Rome, Italy;
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (G.M.L.); (K.M.); (F.N.)
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95123 Catania, Italy; (G.M.L.); (K.M.); (F.N.)
| |
Collapse
|
28
|
Wei L, Mei D, Hu S, Du S. Dual-target EZH2 inhibitor: latest advances in medicinal chemistry. Future Med Chem 2024; 16:1561-1582. [PMID: 39082677 PMCID: PMC11370917 DOI: 10.1080/17568919.2024.2380243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/09/2024] [Indexed: 09/03/2024] Open
Abstract
Enhancer of zeste homolog 2 (EZH2), a histone methyltransferase, plays a crucial role in tumor progression by regulating gene expression. EZH2 inhibitors have emerged as promising anti-tumor agents due to their potential in cancer treatment strategies. However, single-target inhibitors often face limitations such as drug resistance and side effects. Dual-target inhibitors, exemplified by EZH1/2 inhibitor HH-2853(28), offer enhanced efficacy and reduced adverse effects. This review highlights recent advancements in dual inhibitors targeting EZH2 and other proteins like BRD4, PARP1, and EHMT2, emphasizing rational design, structure-activity relationships, and safety profiles, suggesting their potential in clinical applications.
Collapse
Affiliation(s)
- Lai Wei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Department of Orthodontics, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Dan Mei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Department of Orthodontics, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Sijia Hu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Department of Orthodontics, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Shufang Du
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology Department of Orthodontics, Sichuan University, Chengdu, 610041, Sichuan, China
| |
Collapse
|
29
|
Cai Y, Li L, Shao C, Chen Y, Wang Z. Therapeutic Strategies for Angiogenesis Based on Endothelial Cell Epigenetics. J Cardiovasc Transl Res 2024; 17:816-827. [PMID: 38294628 DOI: 10.1007/s12265-024-10485-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/17/2024] [Indexed: 02/01/2024]
Abstract
With the in-depth investigation of various diseases, angiogenesis has gained increasing attention. Among the contributing factors to angiogenesis research, endothelial epigenetics has emerged as an influential player. Endothelial epigenetic therapy exerts its regulatory effects on endothelial cells by controlling gene expression, RNA, and histone modification within these cells, which subsequently promotes or inhibits angiogenesis. As a result, this therapeutic approach offers potential strategies for disease treatment. The purpose of this review is to outline the pertinent mechanisms of endothelial cell epigenetics, encompassing glycolysis, lactation, amino acid metabolism, non-coding RNA, DNA methylation, histone modification, and their connections to specific diseases and clinical applications. We firmly believe that endothelial cell epigenetics has the potential to become an integral component of precision medicine therapy, unveiling novel therapeutic targets and providing new directions and opportunities for disease treatment.
Collapse
Affiliation(s)
- Yue Cai
- Department of Cardiology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China
| | - Yiliu Chen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, No. 438 Jiefang Road, Jingkou District, Zhenjiang, 212000, Jiangsu Province, China.
| |
Collapse
|
30
|
Su Y, Mei L, Jiang T, Wang Z, Ji Y. Novel role of lncRNAs regulatory network in papillary thyroid cancer. Biochem Biophys Rep 2024; 38:101674. [PMID: 38440062 PMCID: PMC10909982 DOI: 10.1016/j.bbrep.2024.101674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/06/2024] Open
Abstract
Papillary thyroid cancer (PTC) is the most common endocrine malignancy. The incidence of PTC has increased annually worldwide. Thus, PTC diagnosis and treatment attract more attention. Noncoding RNAs (lncRNAs) play crucial roles in PTC progression and act as prognostic biomarkers. Moreover, microRNAs (miRNAs) and epithelial-mesenchymal transition (EMT)-associated proteins have potential biomarkers for diagnosing and treating PTC. However, the correlation of lncRNAs with miRNAs and EMT-associated proteins needs further clarification. The present review highlights the recent advances of lncRNAs in PTC. We significantly summarized the two molecular regulatory mechanisms in PTC progress, including lncRNAs-miRNAs-protein signaling axes and lncRNAs-EMT pathways. This review will help our understanding of the association between lncRNAs and PTC and may assist us in evaluating the prognosis for PTC patients. Taken together, targeting the lncRNAs regulatory network has promising applications in diagnosing and treating PTC.
Collapse
Affiliation(s)
- Yuanhao Su
- Department of General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong, University, Xi'an, 710004, China
| | - Lin Mei
- Scientific Research Center and Precision Medical Institute, The Second Affiliated, Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Tiantian Jiang
- Department of General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong, University, Xi'an, 710004, China
| | - Zhidong Wang
- Department of General Surgery, The Second Affiliated Hospital, Xi'an Jiaotong, University, Xi'an, 710004, China
| | - Yuanyuan Ji
- Scientific Research Center and Precision Medical Institute, The Second Affiliated, Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| |
Collapse
|
31
|
Chen Y, Zhu H, Luo Y, Tong S, Liu Y. EZH2: The roles in targeted therapy and mechanisms of resistance in breast cancer. Biomed Pharmacother 2024; 175:116624. [PMID: 38670045 DOI: 10.1016/j.biopha.2024.116624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/04/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Drug resistance presents a formidable challenge in the realm of breast cancer therapy. Accumulating evidence suggests that enhancer of zeste homolog 2 (EZH2), a component of the polycomb repressive complex 2 (PRC2), may serve as a key regulator in controlling drug resistance. EZH2 overexpression has been observed in breast cancer and many other malignancies, showing a strong correlation with poor outcomes. This review aims to summarize the mechanisms by which EZH2 regulates drug resistance, with a specific focus on breast cancer, in order to provide a comprehensive understanding of the underlying molecular processes. Additionally, we will discuss the current strategies and outcomes of targeting EZH2 using both single agents and combination therapies, with the goal of offering improved guidance for the clinical treatment of breast cancer patients who have developed drug resistance.
Collapse
Affiliation(s)
- Yun Chen
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Hongyan Zhu
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Yi Luo
- Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Biotheus Inc., Guangdong Province, Zhuhai 519080, PR China.
| | - Shuangmei Tong
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Yan Liu
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China; Clinical Pharmacy Innovation Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| |
Collapse
|
32
|
Mi L, Xing Z, Zhang Y, He T, Su A, Wei T, Li Z, Wu W. Unveiling Gambogenic Acid as a Promising Antitumor Compound: A Review. PLANTA MEDICA 2024; 90:353-367. [PMID: 38295847 DOI: 10.1055/a-2258-6663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Gambogenic acid is a derivative of gambogic acid, a polyprenylated xanthone isolated from Garcinia hanburyi. Compared with the more widely studied gambogic acid, gambogenic acid has demonstrated advantages such as a more potent antitumor effect and less systemic toxicity than gambogic acid according to early investigations. Therefore, the present review summarizes the effectiveness and mechanisms of gambogenic acid in different cancers and highlights the mechanisms of action. In addition, drug delivery systems to improve the bioavailability of gambogenic acid and its pharmacokinetic profile are included. Gambogenic acid has been applied to treat a wide range of cancers, such as lung, liver, colorectal, breast, gastric, bladder, and prostate cancers. Gambogenic acid exerts its antitumor effects as a novel class of enhancer of zeste homolog 2 inhibitors. It prevents cancer cell proliferation by inducing apoptosis, ferroptosis, and necroptosis and controlling the cell cycle as well as autophagy. Gambogenic acid also hinders tumor cell invasion and metastasis by downregulating metastasis-related proteins. Moreover, gambogenic acid increases the sensitivity of cancer cells to chemotherapy and has shown effects on multidrug resistance in malignancy. This review adds insights for the prevention and treatment of cancers using gambogenic acid.
Collapse
Affiliation(s)
- Li Mi
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Zhichao Xing
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yujie Zhang
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Ting He
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Anping Su
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Wei
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihui Li
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Wenshuang Wu
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
33
|
Zhao R, Cheng S, Bai X, Zhang D, Fang H, Che W, Zhang W, Zhou Y, Duan W, Liang Q, Xiao L, Nie G, Hou Y. Development of an efficient liposomal DOX delivery formulation for HCC therapy by targeting CK2α. Biotechnol J 2024; 19:e2400050. [PMID: 38651271 DOI: 10.1002/biot.202400050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/11/2024] [Accepted: 03/23/2024] [Indexed: 04/25/2024]
Abstract
Hepatocellular carcinoma (HCC) is a digestive tract cancer with high mortality and poor prognosis, especially in China. Current chemotherapeutic drugs lead to poor prognosis, low efficacy, and high side effects due to weak targeting specificity and rapidly formed multidrug resistance (MDR). Based on the previous studies on the doxorubicin (DOX) formulation for cancer targeting therapy, we developed a novel DOX delivery formulation for the targeting chemotherapy of HCC and DOX resistant HCC. HCSP4 was previously screened and casein kinase 2α (CK2α) was predicted as its specific target on HCC cells in our lab. In the study, miR125a-5p was firstly predicted as an MDR inhibiting miRNA, and then CK2α was validated as the target of HCSP4 and miR125a-5p using CK2α-/-HepG2 cells. Based on the above, an HCC targeting and MDR inhibiting DOX delivery liposomal formulation, HCSP4/Lipo-DOX/miR125a-5p was synthesized and tested for its HCC therapeutic efficacy in vitro. The results showed that the liposomal DOX delivery formulation targeted to HCC cells specifically and sensitively, and presented the satisfied therapeutic efficacy for HCC, particularly for DOX resistant HCC. The potential therapeutic mechanism of the DOX delivery formulation was explored, and the formulation inhibited the expression of MDR-relevant genes including ATP-binding cassette subfamily B member 1 (ABCB1, also known as P-glycoprotein), ATP-binding cassette subfamily C member 5 (ABCC5), enhancer of zeste homolog 2 (EZH2), and ATPase Na+/K+ transporting subunit beta 1 (ATP1B1). Our study presents a novel targeting chemotherapeutic drug formulation for the therapy of HCC, especially for drug resistant HCC, although it is primarily and needs further study in vivo, but provided a new strategy for the development of novel anticancer drugs.
Collapse
Affiliation(s)
- Ruixia Zhao
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Sinan Cheng
- Changzhi Medical College, Changzhi, Shanxi, China
| | - Xue Bai
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Danying Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Hongming Fang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Wanlin Che
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Wenxuan Zhang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Yujuan Zhou
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Wei Duan
- School of Medicine, Deakin University, Waurn Ponds, VIC, Australia
| | - Qiumin Liang
- Guangxi Key Laboratory of Agricultural Resource Chemistry and Biotechnology, Yulin, Guangxi, China
| | - Li Xiao
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
- Xi'an Medical University, Xi'an, Shaanxi, China
| | - Guochao Nie
- Guangxi Key Laboratory of Agricultural Resource Chemistry and Biotechnology, Yulin, Guangxi, China
| | - Yingchun Hou
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| |
Collapse
|
34
|
Nguyen HD, Lin CC. Viscoelastic stiffening of gelatin hydrogels for dynamic culture of pancreatic cancer spheroids. Acta Biomater 2024; 177:203-215. [PMID: 38354874 PMCID: PMC10958777 DOI: 10.1016/j.actbio.2024.02.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/24/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
The tumor microenvironment (TME) in pancreatic adenocarcinoma (PDAC) is a complex milieu of cellular and non-cellular components. Pancreatic cancer cells (PCC) and cancer-associated fibroblasts (CAF) are two major cell types in PDAC TME, whereas the non-cellular components are enriched with extracellular matrices (ECM) that contribute to high stiffness and fast stress-relaxation. Previous studies have suggested that higher matrix rigidity promoted aggressive phenotypes of tumors, including PDAC. However, the effects of dynamic viscoelastic matrix properties on cancer cell fate remain largely unexplored. The focus of this work was to understand the effects of such dynamic matrix properties on PDAC cell behaviors, particularly in the context of PCC/CAF co-culture. To this end, we engineered gelatin-norbornene (GelNB) based hydrogels with a built-in mechanism for simultaneously increasing matrix elastic modulus and viscoelasticity. Two GelNB-based macromers, namely GelNB-hydroxyphenylacetic acid (GelNB-HPA) and GelNB-boronic acid (GelNB-BA), were modularly mixed and crosslinked with 4-arm poly(ethylene glycol)-thiol (PEG4SH) to form elastic hydrogels. Treating the hybrid hydrogels with tyrosinase not only increased the elastic moduli of the gels (due to HPA dimerization) but also concurrently produced 1,2-diols that formed reversible boronic acid-diol bonding with the BA groups on GelNB-BA. We employed patient-derived CAF and a PCC cell line COLO-357 to demonstrate the effect of increasing matrix stiffness and viscoelasticity on CAF and PCC cell fate. Our results indicated that in the stiffened environment, PCC underwent epithelial-mesenchymal transition. In the co-culture PCC and CAF spheroid, CAF enhanced PCC spreading and stimulated collagen 1 production. Through mRNA-sequencing, we further showed that stiffened matrices, regardless of the degree of stress-relaxation, heightened the malignant phenotype of PDAC cells. STATEMENT OF SIGNIFICANCE: The pancreatic cancer microenvironment is a complex milieu composed of various cell types and extracellular matrices. It has been suggested that stiffer matrices could promote aggressive behavior in pancreatic cancer, but the effect of dynamic stiffening and matrix stress-relaxation on cancer cell fate remains largely undefined. This study aimed to explore the impact of dynamic changes in matrix viscoelasticity on pancreatic ductal adenocarcinoma (PDAC) cell behavior by developing a hydrogel system capable of simultaneously increasing stiffness and stress-relaxation on demand. This is achieved by crosslinking two gelatin-based macromers through orthogonal thiol-norbornene photochemistry and post-gelation stiffening with mushroom tyrosinase. The results revealed that higher matrix stiffness, regardless of the degree of stress relaxation, exacerbated the malignant characteristics of PDAC cells.
Collapse
Affiliation(s)
- Han D Nguyen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Chien-Chi Lin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA.
| |
Collapse
|
35
|
Rhodes CT, Asokumar D, Sohn M, Naskar S, Elisha L, Stevenson P, Lee DR, Zhang Y, Rocha PP, Dale RK, Lee S, Petros TJ. Loss of Ezh2 in the medial ganglionic eminence alters interneuron fate, cell morphology and gene expression profiles. Front Cell Neurosci 2024; 18:1334244. [PMID: 38419656 PMCID: PMC10899338 DOI: 10.3389/fncel.2024.1334244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction Enhancer of zeste homolog 2 (Ezh2) is responsible for trimethylation of histone 3 at lysine 27 (H3K27me3), resulting in repression of gene expression. Here, we explore the role of Ezh2 in forebrain GABAergic interneuron development. Methods We removed Ezh2 in the MGE by generating Nkx2-1Cre;Ezh2 conditional knockout mice. We then characterized changes in MGE-derived interneuron fate and electrophysiological properties in juvenile mice, as well as alterations in gene expression, chromatin accessibility and histone modifications in the MGE. Results Loss of Ezh2 increases somatostatin-expressing (SST+) and decreases parvalbumin-expressing (PV+) interneurons in the forebrain. We observe fewer MGE-derived interneurons in the first postnatal week, indicating reduced interneuron production. Intrinsic electrophysiological properties in SST+ and PV+ interneurons are normal, but PV+ interneurons display increased axonal complexity in Ezh2 mutant mice. Single nuclei multiome analysis revealed differential gene expression patterns in the embryonic MGE that are predictive of these cell fate changes. Lastly, CUT&Tag analysis revealed that some genomic loci are particularly resistant or susceptible to shifts in H3K27me3 levels in the absence of Ezh2, indicating differential selectivity to epigenetic perturbation. Discussion Thus, loss of Ezh2 in the MGE alters interneuron fate, morphology, and gene expression and regulation. These findings have important implications for both normal development and potentially in disease etiologies.
Collapse
Affiliation(s)
- Christopher T Rhodes
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Dhanya Asokumar
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
- Unit on Genome Structure and Regulation, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Mira Sohn
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Shovan Naskar
- Unit on Functional Neural Circuits, National Institute of Mental Health (NIMH), NIH, Bethesda, MD, United States
| | - Lielle Elisha
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Parker Stevenson
- Unit on Functional Neural Circuits, National Institute of Mental Health (NIMH), NIH, Bethesda, MD, United States
| | - Dongjin R Lee
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Yajun Zhang
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Pedro P Rocha
- Unit on Genome Structure and Regulation, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
- National Cancer Institute (NCI), NIH, Bethesda, MD, United States
| | - Ryan K Dale
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| | - Soohyun Lee
- Unit on Functional Neural Circuits, National Institute of Mental Health (NIMH), NIH, Bethesda, MD, United States
| | - Timothy J Petros
- Unit on Cellular and Molecular Neurodevelopment, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, United States
| |
Collapse
|
36
|
Guo P, Lim RC, Rajawasam K, Trinh T, Sun H, Zhang H. A methylation-phosphorylation switch controls EZH2 stability and hematopoiesis. eLife 2024; 13:e86168. [PMID: 38346162 PMCID: PMC10901513 DOI: 10.7554/elife.86168] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/11/2024] [Indexed: 02/29/2024] Open
Abstract
The Polycomb Repressive Complex 2 (PRC2) methylates H3K27 to regulate development and cell fate by transcriptional silencing. Alteration of PRC2 is associated with various cancers. Here, we show that mouse Kdm1a deletion causes a dramatic reduction of PRC2 proteins, whereas mouse null mutation of L3mbtl3 or Dcaf5 results in PRC2 accumulation and increased H3K27 trimethylation. The catalytic subunit of PRC2, EZH2, is methylated at lysine 20 (K20), promoting EZH2 proteolysis by L3MBTL3 and the CLR4DCAF5 ubiquitin ligase. KDM1A (LSD1) demethylates the methylated K20 to stabilize EZH2. K20 methylation is inhibited by AKT-mediated phosphorylation of serine 21 in EZH2. Mouse Ezh2K20R/K20R mutants develop hepatosplenomegaly associated with high GFI1B expression, and Ezh2K20R/K20R mutant bone marrows expand hematopoietic stem cells and downstream hematopoietic populations. Our studies reveal that EZH2 is regulated by methylation-dependent proteolysis, which is negatively controlled by AKT-mediated S21 phosphorylation to establish a methylation-phosphorylation switch to regulate the PRC2 activity and hematopoiesis.
Collapse
Affiliation(s)
- Pengfei Guo
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, United States
| | - Rebecca C Lim
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, United States
| | - Keshari Rajawasam
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, United States
| | - Tiffany Trinh
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, United States
| | - Hong Sun
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, United States
| | - Hui Zhang
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, United States
| |
Collapse
|
37
|
Guo Y, Cheng R, Wang Y, Gonzalez ME, Zhang H, Liu Y, Kleer CG, Xue L. Regulation of EZH2 protein stability: new mechanisms, roles in tumorigenesis, and roads to the clinic. EBioMedicine 2024; 100:104972. [PMID: 38244292 PMCID: PMC10835131 DOI: 10.1016/j.ebiom.2024.104972] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/13/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
The importance of EZH2 as a key methyltransferase has been well documented theoretically. Practically, the first EZH2 inhibitor Tazemetostat (EPZ6438), was approved by FDA in 2020 and is used in clinic. However, for most solid tumors it is not as effective as desired and the scope of clinical indications is limited, suggesting that targeting its enzymatic activity may not be sufficient. Recent technologies focusing on the degradation of EZH2 protein have drawn attention due to their potential robust effects. This review focuses on the molecular mechanisms that regulate EZH2 protein stability via post-translational modifications (PTMs), mainly including ubiquitination, phosphorylation, and acetylation. In addition, we discuss recent advancements of multiple proteolysis targeting chimeras (PROTACs) strategies and the latest degraders that can downregulate EZH2 protein. We aim to highlight future directions to expand the application of novel EZH2 inhibitors by targeting both EZH2 enzymatic activity and protein stability.
Collapse
Affiliation(s)
- Yunyun Guo
- Cancer Center of Peking University Third Hospital, Beijing, China; Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Rui Cheng
- Cancer Center of Peking University Third Hospital, Beijing, China; Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Yuqing Wang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Maria E Gonzalez
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Hongshan Zhang
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Yang Liu
- Cancer Center of Peking University Third Hospital, Beijing, China; Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
| | - Celina G Kleer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| | - Lixiang Xue
- Cancer Center of Peking University Third Hospital, Beijing, China; Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
38
|
Peng X, Hu Z, Zeng L, Zhang M, Xu C, Lu B, Tao C, Chen W, Hou W, Cheng K, Bi H, Pan W, Chen J. Overview of epigenetic degraders based on PROTAC, molecular glue, and hydrophobic tagging technologies. Acta Pharm Sin B 2024; 14:533-578. [PMID: 38322348 PMCID: PMC10840439 DOI: 10.1016/j.apsb.2023.09.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/21/2023] [Accepted: 08/30/2023] [Indexed: 02/08/2024] Open
Abstract
Epigenetic pathways play a critical role in the initiation, progression, and metastasis of cancer. Over the past few decades, significant progress has been made in the development of targeted epigenetic modulators (e.g., inhibitors). However, epigenetic inhibitors have faced multiple challenges, including limited clinical efficacy, toxicities, lack of subtype selectivity, and drug resistance. As a result, the design of new epigenetic modulators (e.g., degraders) such as PROTACs, molecular glue, and hydrophobic tagging (HyT) degraders has garnered significant attention from both academia and pharmaceutical industry, and numerous epigenetic degraders have been discovered in the past decade. In this review, we aim to provide an in-depth illustration of new degrading strategies (2017-2023) targeting epigenetic proteins for cancer therapy, focusing on the rational design, pharmacodynamics, pharmacokinetics, clinical status, and crystal structure information of these degraders. Importantly, we also provide deep insights into the potential challenges and corresponding remedies of this approach to drug design and development. Overall, we hope this review will offer a better mechanistic understanding and serve as a useful guide for the development of emerging epigenetic-targeting degraders.
Collapse
Affiliation(s)
- Xiaopeng Peng
- College of Pharmacy, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 314000, China
| | - Zhihao Hu
- College of Pharmacy, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 314000, China
| | - Limei Zeng
- College of Basic Medicine, Gannan Medical University, Ganzhou 314000, China
| | - Meizhu Zhang
- College of Pharmacy, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 314000, China
| | - Congcong Xu
- College of Pharmacy, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 314000, China
| | - Benyan Lu
- College of Pharmacy, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 314000, China
| | - Chengpeng Tao
- College of Pharmacy, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 314000, China
| | - Weiming Chen
- College of Pharmacy, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 314000, China
| | - Wen Hou
- College of Pharmacy, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 314000, China
| | - Kui Cheng
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Huichang Bi
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wanyi Pan
- College of Pharmacy, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, Ganzhou 314000, China
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
39
|
Xiao B, Shi Z, Liu J, Huang Q, Shu K, Liu F, Zhi C, Zhang D, Wu L, Yang S, Zeng X, Fan T, Liu Z, Jiang Y. Design, synthesis, and evaluation of VHL-based EZH2 degraders for breast cancer. Bioorg Chem 2024; 143:107078. [PMID: 38181661 DOI: 10.1016/j.bioorg.2023.107078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/26/2023] [Accepted: 12/27/2023] [Indexed: 01/07/2024]
Abstract
EZH2 (enhancer of zeste homolog 2) is one of the most important histone methyltransferases (HMTs), and overexpression of EZH2 can lead to proliferation, migration and angiogenesis of tumor cells. But most of EZH2 inhibitors are only effective against some hematologic malignancies and have poor efficacy against solid tumors. Here, we report the design, synthesis, and evaluation of highly potent proteolysis targeting chimeric (PROTACs) small molecules targeting EZH2. We developed a potent and effective EZH2 degrader P4, which effectively induced EZH2 protein degradation and inhibited breast cancer cell growth. Further studies showed that P4 can significantly decrease the degree of H3K27me3 in MDA-MB-231 cell line, induce apoptosis and G0/G1 phase arrest in Pfeiffer and MDA-MB-231 cell lines. Therefore, P4 is a potential anticancer molecule for breast cancer treatment.
Collapse
Affiliation(s)
- Boren Xiao
- Department of Chemistry, Tsinghua University, Beijing 100084, China; The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Zhichao Shi
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Jiaqi Liu
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Qiuhua Huang
- Department of Chemistry, Tsinghua University, Beijing 100084, China; The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Kaifei Shu
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Funian Liu
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Cailian Zhi
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Dandan Zhang
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Lihong Wu
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Shiqi Yang
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Xiliang Zeng
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Tingting Fan
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| | - Zijian Liu
- Shenzhen Kivita Innovative Drug Discovery Institute, Shenzhen 518057, China; Shenzhen Winkey Technology Co., Ltd., Shenzhen 518000, China.
| | - Yuyang Jiang
- The State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Biology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China; School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
40
|
Van der Meer JMR, Bulder I, Kuijk C, Kleijer M, Verheij MW, Omar SZ, Haverkate NJE, Dolstra H, Blom B, Hazenberg MD, Voermans C. Generation of human ILC3 from allogeneic and autologous CD34 + hematopoietic progenitors toward adoptive transfer. Cytotherapy 2024; 26:136-144. [PMID: 38149947 DOI: 10.1016/j.jcyt.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 11/10/2023] [Accepted: 11/21/2023] [Indexed: 12/28/2023]
Abstract
Type 3 innate lymphoid cells (ILC3) are important in tissue homeostasis. In the gut, ILC3 repair damaged epithelium and suppress inflammation. In allogeneic hematopoietic cell transplantation (HCT), ILC3 protect against graft-versus-host disease (GvHD), most likely by restoring tissue damage and preventing inflammation. We hypothesize that supplementing HCT grafts with interleukin-22 (IL-22)-producing ILC3 may prevent acute GvHD. We therefore explored ex vivo generation of human IL-22-producing ILC3 from hematopoietic stem and progenitor cells (HSPC) obtained from adult, neonatal and fetal sources. We established a stroma-free system culturing human cord blood-derived CD34+ HSPC with successive cytokine mixes for 5 weeks. We analyzed the presence of phenotypically defined ILC, their viability, proliferation and IL-22 production (after stimulation) by flow cytometry and enzyme-linked immunosorbent assay (ELISA). We found that the addition of recombinant human IL-15 and the enhancer of zeste homolog 1/2 inhibitor UNC1999 promoted ILC3 generation. Similar results were demonstrated when UNC1999 was added to CD34+ HSPC derived from healthy adult granulocyte colony-stimulating factor mobilized peripheral blood and bone marrow, but not fetal liver. UNC1999 did not negatively impact IL-22 production in any of the HSPC sources. Finally, we observed that autologous HSPC mobilized from the blood of adults with hematological malignancies also developed into ILC3, albeit with a significantly lower capacity. Together, we developed a stroma-free protocol to generate large quantities of IL-22-producing ILC3 from healthy adult human HSPC that can be applied for adoptive transfer to prevent GvHD after allogeneic HCT.
Collapse
Affiliation(s)
- Jolien M R Van der Meer
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Ingrid Bulder
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Carlijn Kuijk
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Marion Kleijer
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Myrddin W Verheij
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands
| | - Said Z Omar
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, The Netherlands
| | - Nienke J E Haverkate
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, The Netherlands
| | - Harry Dolstra
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bianca Blom
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, The Netherlands
| | - Mette D Hazenberg
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Department of Hematology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Carlijn Voermans
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, the Netherlands; Department of Hematology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
41
|
Li D, Peng X, Hu Z, Li S, Chen J, Pan W. Small molecules targeting selected histone methyltransferases (HMTs) for cancer treatment: Current progress and novel strategies. Eur J Med Chem 2024; 264:115982. [PMID: 38056296 DOI: 10.1016/j.ejmech.2023.115982] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/17/2023] [Accepted: 11/19/2023] [Indexed: 12/08/2023]
Abstract
Histone methyltransferases (HMTs) play a critical role in gene post-translational regulation and diverse physiological processes, and are implicated in a plethora of human diseases, especially cancer. Increasing evidences demonstrate that HMTs may serve as a potential therapeutic target for cancer treatment. Thus, the development of HMTs inhibitor have been pursued with steadily increasing interest over the past decade. However, the disadvantages such as insufficient clinical efficacy, moderate selectivity, and propensity for acquired resistance have hindered the development of conventional HMT inhibitors. New technologies and methods are imperative to enhance the anticancer activity of HMT inhibitors. In this review, we first review the structure and biological functions of the several essential HMTs, such as EZH2, G9a, PRMT5, and DOT1L. The internal relationship between these HMTs and cancer is also expounded. Next, we mainly focus on the latest progress in the development of HMT modulators encompassing dual-target inhibitors, targeted protein degraders and covalent inhibitors from perspectives such as rational design, pharmacodynamics, pharmacokinetics, and clinical status. Lastly, we also discuss the challenges and future directions for HMT-based drug discovery for cancer therapy.
Collapse
Affiliation(s)
- Deping Li
- Department of Pharmacy, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, PR China
| | - Xiaopeng Peng
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Zhihao Hu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Shuqing Li
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, 516000, PR China.
| | - Wanyi Pan
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China.
| |
Collapse
|
42
|
Xu X, Chen J, Wang G, Zhang X, Li Q, Zhou X, Guo F, Li M. Conformationally constrained potent inhibitors for enhancer of zeste homolog 2 (EZH2). Bioorg Med Chem Lett 2024; 97:129558. [PMID: 37956762 DOI: 10.1016/j.bmcl.2023.129558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/27/2023] [Accepted: 11/10/2023] [Indexed: 11/15/2023]
Abstract
The enhancer of zeste homolog 2 (EZH2) plays the role of the main catalytic subunit of polycomb repressive complex 2 (PRC2) that catalyzes the methylation of histone H3 lysine 27 (H3K27). Overexpression or mutation of EZH2 has been observed in many types of hematologic malignancies and solid tumors, such as myeloma, lymphoma, prostate, breast, kidney, and lung cancers. EZH2 has been demonstrated as a promising therapeutic target for the treatment of tumors. Based on the structure of 1 (EPZ-6438), a series of novel conformationally constrained derivatives were designed and synthesized aiming to improve the EZH2 inhibition activity, especially for mutated EZH2. Structure and activity relationship (SAR) exploration and optimization at both enzymatic and cellular levels led to the discovery of 28. In vitro, 28 displayed potent EZH2 inhibition activity with an IC50 value of 0.95 nM, which is comparable to EPZ-6438 (1). 28 exhibited high anti-proliferation activity against different lymphoma cell lines including WSU-DLCL2, Pfeiffer and Karpas-422 (IC50 = 2.36, 1.73, and 1.82 nM, respectively). In vivo, 28 showed acceptable pharmacokinetic characteristics (oral bioavailability F = 36.9%) and better efficacy than 1 in both Pfeiffer and Karpas-422 xenograft mouse models, suggesting that it can be further developed as a potential therapeutic candidate for EZH2-associated cancers.
Collapse
Affiliation(s)
- Xin Xu
- Shanghai Synergy Pharmaceutical Sciences Co., Ltd., Shanghai, China.
| | - Jia Chen
- Shanghai Synergy Pharmaceutical Sciences Co., Ltd., Shanghai, China
| | - Guan Wang
- Shanghai Synergy Pharmaceutical Sciences Co., Ltd., Shanghai, China
| | - Xiaojuan Zhang
- Shanghai Synergy Pharmaceutical Sciences Co., Ltd., Shanghai, China
| | - Qiang Li
- Shanghai Synergy Pharmaceutical Sciences Co., Ltd., Shanghai, China
| | - Xiaobo Zhou
- Shanghai Synergy Pharmaceutical Sciences Co., Ltd., Shanghai, China
| | - Fengying Guo
- Shanghai Synergy Pharmaceutical Sciences Co., Ltd., Shanghai, China
| | - Min Li
- Shanghai Synergy Pharmaceutical Sciences Co., Ltd., Shanghai, China
| |
Collapse
|
43
|
Hasan A, Khan NA, Uddin S, Khan AQ, Steinhoff M. Deregulated transcription factors in the emerging cancer hallmarks. Semin Cancer Biol 2024; 98:31-50. [PMID: 38123029 DOI: 10.1016/j.semcancer.2023.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 11/25/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Cancer progression is a multifaceted process that entails several stages and demands the persistent expression or activation of transcription factors (TFs) to facilitate growth and survival. TFs are a cluster of proteins with DNA-binding domains that attach to promoter or enhancer DNA strands to start the transcription of genes by collaborating with RNA polymerase and other supporting proteins. They are generally acknowledged as the major regulatory molecules that coordinate biological homeostasis and the appropriate functioning of cellular components, subsequently contributing to human physiology. TFs proteins are crucial for controlling transcription during the embryonic stage and development, and the stability of different cell types depends on how they function in different cell types. The development and progression of cancer cells and tumors might be triggered by any anomaly in transcription factor function. It has long been acknowledged that cancer development is accompanied by the dysregulated activity of TF alterations which might result in faulty gene expression. Recent studies have suggested that dysregulated transcription factors play a major role in developing various human malignancies by altering and rewiring metabolic processes, modifying the immune response, and triggering oncogenic signaling cascades. This review emphasizes the interplay between TFs involved in metabolic and epigenetic reprogramming, evading immune attacks, cellular senescence, and the maintenance of cancer stemness in cancerous cells. The insights presented herein will facilitate the development of innovative therapeutic modalities to tackle the dysregulated transcription factors underlying cancer.
Collapse
Affiliation(s)
- Adria Hasan
- Molecular Cell Biology Laboratory, Integral Information and Research Centre-4 (IIRC-4), Integral University, Lucknow 226026, India; Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow 226026, India
| | - Naushad Ahmad Khan
- Department of Surgery, Trauma and Vascular Surgery Clinical Research, Hamad General Hospital, Doha 3050, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Biosciences, Integral University, Lucknow 226026, India; Animal Research Center, Qatar University, Doha, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Abdul Q Khan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar.
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Animal Research Center, Qatar University, Doha, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar; Department of Medicine, Weill Cornell Medicine Qatar, Qatar Foundation-Education City, Doha 24144, Qatar; Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; College of Medicine, Qatar University, Doha 2713, Qatar
| |
Collapse
|
44
|
Hosokawa M, Tetsumoto S, Yasui M, Kono Y, Ogawara KI. 3-deazaneplanocin A, a histone methyltransferase inhibitor, improved the chemoresistance induced under hypoxia in melanoma cells. Biochem Biophys Res Commun 2023; 677:26-30. [PMID: 37542772 DOI: 10.1016/j.bbrc.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023]
Abstract
One of common characteristics of solid tumors is low O2 level, so-called hypoxia, which plays a critical role in chemoresistance. Epigenetic mechanism such as DNA methylation and histone modification is involved in cancer development and progression. There is ample evidence that epigenetic drugs reversed acquired chemoresistance in cancer cells under normal O2 level, normoxia. However, it remains unknown whether epigenetic drugs improve acquired chemoresistance under hypoxia. The aim of our study was to investigate whether epigenetic drugs can improve the chemoresistance induced under hypoxia in cancer cells. In murine melanoma B16-BL6 (B16) cells, the culture under hypoxia, 1%O2 caused the elevated expression of hypoxia-inducible factor-1α (HIF-1α) and its target genes. The chemoresistance to 7-ethyl-10-hydroxycamptothecin (SN-38, the active metabolite of irinotecan) was also acquired under hypoxia in B16 cells. In addition, as epigenetic mechanisms, the protein expression of the enhancer of zeste homolog 2 (EZH2), histone methyltransferase and its target histone H3 trimethylation at lysine 27 (H3K27Me3) level increased under hypoxia. The induction of H3K27Me3 under hypoxia was suppressed by EZH2 siRNA and 3-deazaneplanocin A (DZNep), an EZH2 inhibitor. Furthermore, both EZH2 siRNA and DZNep significantly reduced the cell viability after SN-38 treatment and improved the chemoresistance to SN-38 under hypoxia. These results indicated that the chemoresistance to SN-38 under hypoxia would arise from epigenetic mechanism, H3K27Me3 elevation due to EZH2 induction. In conclusion, a histone methyltransferase EZH2 inhibitor, DZNep was capable of tackling acquired chemoresistance via the suppression of histone methylation induced under hypoxic tumor microenvironment.
Collapse
Affiliation(s)
- Mika Hosokawa
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan.
| | - Sekai Tetsumoto
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan
| | - Mirano Yasui
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan
| | - Yusuke Kono
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan
| | - Ken-Ichi Ogawara
- Laboratory of Pharmaceutics, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe, 658-8558, Japan
| |
Collapse
|
45
|
Wu Y, Xu Z, Chen X, Fu G, Tian J, Jin B. RCC1 functions as a tumor facilitator in clear cell renal cell carcinoma by dysregulating cell cycle, apoptosis, and EZH2 stability. Cancer Med 2023; 12:19889-19903. [PMID: 37747077 PMCID: PMC10587970 DOI: 10.1002/cam4.6594] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/03/2023] [Accepted: 09/14/2023] [Indexed: 09/26/2023] Open
Abstract
BACKGROUND RCC1 functions as a pivotal guanine nucleotide exchange factor and was reported to be involved in mitosis, the assembly of the nuclear envelope, nucleocytoplasmic transport in cell physiological processes. Recent studies reported that RCC1 could regulate immunological pathways and promote the growth of some malignant solid tumors. However, the prognostic value and exact function of RCC1 remain unknown in patients with clear cell renal cell carcinoma (cRCC). METHODS The UALCAN and KM plotter portals were used to analyze the expression profile and related tumor prognosis of RCC1 in ccRCC using data from TCGA. The expression profile of RCC1 was also confirmed in clinical samples using qRT-PCR, western blotting, and immunohistochemistry. The role of RCC1 on ccRCC cells in vitro was confirmed by a series of functional assays. Animal experiments were performed to verify the suppressive effect of RCC1 knockdown on tumor growth in vivo. The correlation of RCC1 expression with that of EZH2 was explored in clinical samples using IHC. The interaction between RCC1 and EZH2 was further verified using a CO-IP assay and a protein stability assay. RESULTS RCC1 was upregulated in ccRCC tissues compared with normal tissues in TCGA dataset and paired clinical samples. RCC1 promoted ccRCC progression by accelerating the cell cycle and suppressing apoptosis. In addition, RCC1 could bind EZH2 and regulate its expression at the posttranscriptional level. RCC1 and EZH2 expression showed a strong correlation in clinical samples. Further investigation proved that RCC1 regulated EZH2 protein stability through the ubiquitin-proteasome pathway. CONCLUSIONS RCC1 could be a potential therapeutic target in ccRCC. The RCC1/EZH2 axis takes part in the development of ccRCC.
Collapse
Affiliation(s)
- Yunfei Wu
- Department of Urology, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and TreatmentHangzhouChina
| | - Zhijie Xu
- Department of Urology, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and TreatmentHangzhouChina
| | - Xiaoyi Chen
- Department of Urology, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and TreatmentHangzhouChina
| | - Guanghou Fu
- Department of Urology, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and TreatmentHangzhouChina
| | - Junjie Tian
- Department of Urology, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and TreatmentHangzhouChina
| | - Baiye Jin
- Department of Urology, The First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and TreatmentHangzhouChina
| |
Collapse
|
46
|
Kim SB, Yang CE, Jeong Y, Yu M, Choi WS, Lim JY, Jeon Y. Dual Targeting of EZH2 Degradation and EGFR/HER2 Inhibition for Enhanced Efficacy against Burkitt's Lymphoma. Cancers (Basel) 2023; 15:4472. [PMID: 37760442 PMCID: PMC10526300 DOI: 10.3390/cancers15184472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
EZH2, a histone methyltransferase, contributes significantly to cancer cell survival and proliferation. Although various EZH2 inhibitors have demonstrated promise in treating lymphoma, they have not fully managed to curb lymphoma cell proliferation despite effective reduction of the H3K27me3 mark. We used MS1943, an EZH2 selective degrader, which successfully diminishes EZH2 levels in lymphoma cells. Additionally, lapatinib, a dual inhibitor of the epidermal growth factor receptor (EGFR) and human epidermal growth factor receptor 2 (HER2) tyrosine kinases, targets a receptor protein that regulates cell growth and division. The overexpression of this protein is often observed in lymphoma cells. Our study aims to combine these two therapeutic targets to stimulate apoptosis pathways and potentially suppress Burkitt's lymphoma cell survival and proliferation in a complementary and synergistic manner. We observed that a combination of MS1943 and lapatinib induced apoptosis in Daudi cells and caused cell cycle arrest at the S and G2/M phases in both Ramos and Daudi cells. This strategy, using a combination of MS1943 and lapatinib, presents a promising therapeutic approach for treating lymphoma and potentially Burkitt's lymphoma.
Collapse
Affiliation(s)
- Se Been Kim
- Department of Biomedical Laboratory Science, Inje University, Gimhae 50834, Republic of Korea; (S.B.K.); (C.-E.Y.); (Y.J.); (M.Y.); (W.-S.C.)
- Lymphoma and Cell-Therapy Research Center, Yeouido St. Mary Hospital, School of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea
- JL’s Lymphoma Origins & Clinical Applications Lab (JL-LOCAL), The Catholic University of Korea, Seoul 07345, Republic of Korea
| | - Chae-Eun Yang
- Department of Biomedical Laboratory Science, Inje University, Gimhae 50834, Republic of Korea; (S.B.K.); (C.-E.Y.); (Y.J.); (M.Y.); (W.-S.C.)
- Lymphoma and Cell-Therapy Research Center, Yeouido St. Mary Hospital, School of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea
- JL’s Lymphoma Origins & Clinical Applications Lab (JL-LOCAL), The Catholic University of Korea, Seoul 07345, Republic of Korea
| | - Yurim Jeong
- Department of Biomedical Laboratory Science, Inje University, Gimhae 50834, Republic of Korea; (S.B.K.); (C.-E.Y.); (Y.J.); (M.Y.); (W.-S.C.)
- Lymphoma and Cell-Therapy Research Center, Yeouido St. Mary Hospital, School of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea
- JL’s Lymphoma Origins & Clinical Applications Lab (JL-LOCAL), The Catholic University of Korea, Seoul 07345, Republic of Korea
| | - Minseo Yu
- Department of Biomedical Laboratory Science, Inje University, Gimhae 50834, Republic of Korea; (S.B.K.); (C.-E.Y.); (Y.J.); (M.Y.); (W.-S.C.)
- Lymphoma and Cell-Therapy Research Center, Yeouido St. Mary Hospital, School of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea
- JL’s Lymphoma Origins & Clinical Applications Lab (JL-LOCAL), The Catholic University of Korea, Seoul 07345, Republic of Korea
| | - Wan-Su Choi
- Department of Biomedical Laboratory Science, Inje University, Gimhae 50834, Republic of Korea; (S.B.K.); (C.-E.Y.); (Y.J.); (M.Y.); (W.-S.C.)
- Department of Digital Anti-Aging Health Care, Inje University, Gimhae 50834, Republic of Korea
| | - Jung-Yeon Lim
- Department of Biomedical Laboratory Science, Inje University, Gimhae 50834, Republic of Korea; (S.B.K.); (C.-E.Y.); (Y.J.); (M.Y.); (W.-S.C.)
- Lymphoma and Cell-Therapy Research Center, Yeouido St. Mary Hospital, School of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea
- JL’s Lymphoma Origins & Clinical Applications Lab (JL-LOCAL), The Catholic University of Korea, Seoul 07345, Republic of Korea
| | - Youngwoo Jeon
- Lymphoma and Cell-Therapy Research Center, Yeouido St. Mary Hospital, School of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea
- JL’s Lymphoma Origins & Clinical Applications Lab (JL-LOCAL), The Catholic University of Korea, Seoul 07345, Republic of Korea
- Department of Hematology, Yeouido St. Mary Hospital, School of Medicine, The Catholic University of Korea, Seoul 07345, Republic of Korea
| |
Collapse
|
47
|
Zheng X, Dozmorov MG, Strohlein CE, Bastacky S, Sawalha AH. Ezh2 Knockout in B Cells Impairs Plasmablast Differentiation and Ameliorates Lupus-like Disease in MRL/lpr Mice. Arthritis Rheumatol 2023; 75:1395-1406. [PMID: 36897808 PMCID: PMC10492897 DOI: 10.1002/art.42492] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 01/25/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023]
Abstract
OBJECTIVES EZH2 regulates B cell development and differentiation. We previously demonstrated increased EZH2 expression in peripheral blood mononuclear cells from lupus patients. The goal of this study was to evaluate the role of EZH2 expression in B cells in the pathogenesis of lupus. METHODS We generated an MRL/lpr mouse with floxed Ezh2, which was crossed with CD19-Cre mice to examine the effect of B cell EZH2 deficiency in MRL/lpr lupus-prone mice. Differentiation of B cells was assessed using flow cytometry. Single-cell RNA sequencing and single-cell B cell receptor sequencing were performed. In vitro B cell culture with an X-box binding protein 1 (XBP1) inhibitor was performed. EZH2 and XBP1 messenger RNA levels in CD19+ B cells isolated from lupus patients and healthy controls were analyzed. RESULTS We show that Ezh2 deletion in B cells significantly decreased autoantibody production and improved glomerulonephritis. B cell development was altered in the bone marrow and spleen of EZH2-deficient mice. Differentiation of germinal center B cells and plasmablasts was impaired. Single-cell RNA sequencing showed that XBP1, a key transcription factor in B cell development, is down-regulated in the absence of EZH2. Inhibiting XBP1 in vitro impairs plasmablast development similar to EZH2 deficiency in mice. Single-cell B cell receptor RNA sequencing revealed defective immunoglobulin class-switch recombination in EZH2-deficient mice. In human lupus B cells, we observed a strong correlation between EZH2 and XBP1 messenger RNA expression levels. CONCLUSION EZH2 overexpression in B cells contributes to disease pathogenesis in lupus.
Collapse
Affiliation(s)
- Xiaoqing Zheng
- Department of Pediatrics, Division of Rheumatology, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mikhail G Dozmorov
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia
| | - Colleen E Strohlein
- Department of Pediatrics, Division of Rheumatology, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sheldon Bastacky
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Amr H Sawalha
- Department of Pediatrics, Division of Rheumatology, Children's Hospital of Pittsburgh, Department of Medicine, Division of Rheumatology and Clinical Immunology, Lupus Center of Excellence, and Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
48
|
Celikdemir B, Houben R, Kervarrec T, Samimi M, Schrama D. Current and preclinical treatment options for Merkel cell carcinoma. Expert Opin Biol Ther 2023; 23:1015-1034. [PMID: 37691397 DOI: 10.1080/14712598.2023.2257603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 09/04/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION Merkel cell carcinoma (MCC) is a rare, highly aggressive form of skin cancer with neuroendocrine features. The origin of this cancer is still unclear, but research in the last 15 years has demonstrated that MCC arises via two distinct etiologic pathways, i.e. virus and UV-induced. Considering the high mortality rate and the limited therapeutic options available, this review aims to highlight the significance of MCC research and the need for advancement in MCC treatment. AREAS COVERED With the advent of the immune checkpoint inhibitor therapies, we now have treatment options providing a survival benefit for patients with advanced MCC. However, the issue of primary and acquired resistance to these therapies remains a significant concern. Therefore, ongoing efforts seeking additional therapeutic targets and approaches for MCC therapy are a necessity. Through a comprehensive literature search, we provide an overview on recent preclinical and clinical studies with respect to MCC therapy. EXPERT OPINION Currently, the only evidence-based therapy for MCC is immune checkpoint blockade with anti-PD-1/PD-L1 for advanced patients. Neoadjuvant, adjuvant and combined immune checkpoint blockade are promising treatment options.
Collapse
Affiliation(s)
- Büke Celikdemir
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Roland Houben
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Thibault Kervarrec
- Department of Pathology, Centre Hospitalier Universitaire De Tours, Tours, France
| | - Mahtab Samimi
- Department of Dermatology, University Hospital of Tours, Tours, France
| | - David Schrama
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
49
|
Wang B, Liu Y, Liao Z, Wu H, Zhang B, Zhang L. EZH2 in hepatocellular carcinoma: progression, immunity, and potential targeting therapies. Exp Hematol Oncol 2023; 12:52. [PMID: 37268997 PMCID: PMC10236851 DOI: 10.1186/s40164-023-00405-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/15/2023] [Indexed: 06/04/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the leading cause of cancer-related death. The accumulation of genetic and epigenetic changes is closely related to the occurrence and development of HCC. Enhancer of zeste homolog 2 (EZH2, a histone methyltransferase) is suggested to be one of the principal factors that mediates oncogenesis by acting as a driver of epigenetic alternation. Recent studies show that EZH2 is widely involved in proliferation and metastasis of HCC cells. In this review, the functions of EZH2 in HCC progression, the role of EZH2 in tumor immunity and the application of EZH2-related inhibitors in HCC therapy are summarized.
Collapse
Affiliation(s)
- Bohan Wang
- Hepatic Surgery Center, Institute of Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yachong Liu
- Hepatic Surgery Center, Institute of Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhibin Liao
- Hepatic Surgery Center, Institute of Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haofeng Wu
- Hepatic Surgery Center, Institute of Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Institute of Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Lei Zhang
- Hepatic Surgery Center, Institute of Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Department of Hepatobiliary Surgery, Shanxi Tongji Hospital, Tongji Medical College, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Shanxi Medical University, Huazhong University of Science and Technology, Taiyuan, 030032, China.
- Key Laboratory of Hepatobiliary and Pancreatic Diseases of Shanxi Province (Preparatory), Shanxi Tongji Hospital, Tongji Medical College, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Shanxi Medical University, Huazhong University of Science and Technology, Taiyuan, 030032, China.
| |
Collapse
|
50
|
Yang X, Xu L, Yang L. Recent advances in EZH2-based dual inhibitors in the treatment of cancers. Eur J Med Chem 2023; 256:115461. [PMID: 37156182 DOI: 10.1016/j.ejmech.2023.115461] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/04/2023] [Accepted: 05/04/2023] [Indexed: 05/10/2023]
Abstract
The enhancer of zeste homolog 2 (EZH2) protein is the catalytic subunit of one of the histone methyltransferases. EZH2 catalyzes the trimethylation of lysine 27 of histone H3 (H3K27me3) and further alters downstream target levels. EZH2 is upregulated in cancer tissues, wherein its levels correlate strongly with cancer genesis, progression, metastasis, and invasion. Consequently, it has emerged as a novel anticancer therapeutic target. Nonetheless, developing EZH2 inhibitors (EZH2i) has encountered numerous difficulties, such as pre-clinical drug resistance and poor therapeutic effect. The EZH2i synergistically suppresses cancers when used in combination with additional antitumor drugs, such as PARP inhibitors, HDAC inhibitors, BRD4 inhibitors, EZH1 inhibitors, and EHMT2 inhibitors. Typically, the use of dual inhibitors of two different targets mediated by one individual molecule has been recognized as the preferred approach for overcoming the limitations of EZH2 monotherapy. The present review discusses the theoretical basis for designing EZH2-based dual-target inhibitors, and also describes some in vitro and in vivo analysis results.
Collapse
Affiliation(s)
- Xiaojuan Yang
- School of Pharmacy, Xinxiang University, Xinxiang, 453003, China.
| | - Lu Xu
- School of Pharmacy, Xinxiang University, Xinxiang, 453003, China
| | - Li Yang
- School of Pharmacy, Xinxiang University, Xinxiang, 453003, China
| |
Collapse
|