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
|
Wang MS, Sussman J, Xu JA, Patel R, Elghawy O, Rawla P. Pharmacological Advancements of PRC2 in Cancer Therapy: A Narrative Review. Life (Basel) 2024; 14:1645. [PMID: 39768352 PMCID: PMC11678550 DOI: 10.3390/life14121645] [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: 08/15/2024] [Revised: 11/25/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Polycomb repressive complex 2 (PRC2) is known to regulate gene expression and chromatin structure as it methylates H3K27, resulting in gene silencing. Studies have shown that PRC2 has dual functions in oncogenesis that allow it to function as both an oncogene and a tumor suppressor. Because of this, nuanced strategies are necessary to promote or inhibit PRC2 activity therapeutically. Given the therapeutic vulnerabilities and associated risks in oncological applications, a structured literature review on PRC2 was conducted to showcase similar cofactor competitor inhibitors of PRC2. Key inhibitors such as Tazemetostat, GSK126, Valemetostat, and UNC1999 have shown promise for clinical use within various studies. Tazemetostat and GSK126 are both highly selective for wild-type and lymphoma-associated EZH2 mutants. Valemetostat and UNC1999 have shown promise as orally bioavailable and SAM-competitive inhibitors of both EZH1 and EZH2, giving them greater efficacy against potential drug resistance. The development of other PRC2 inhibitors, particularly inhibitors targeting the EED or SUZ12 subunit, is also being explored with the development of drugs like EED 226. This review aims to bridge gaps in the current literature and provide a unified perspective on promising PRC2 inhibitors as therapeutic agents in the treatment of lymphomas and solid tumors.
Collapse
Affiliation(s)
- Michael S. Wang
- Hospital of the University of Pennsylvania, HUP 3400 Spruce St., Philadelphia, PA 19104, USA; (M.S.W.)
| | - Jonathan Sussman
- Hospital of the University of Pennsylvania, HUP 3400 Spruce St., Philadelphia, PA 19104, USA; (M.S.W.)
| | - Jessica A. Xu
- Hospital of the University of Pennsylvania, HUP 3400 Spruce St., Philadelphia, PA 19104, USA; (M.S.W.)
| | - Reema Patel
- University of Virginia School of Medicine, Charlottesville, VA 22903, USA;
| | - Omar Elghawy
- Hospital of the University of Pennsylvania, HUP 3400 Spruce St., Philadelphia, PA 19104, USA; (M.S.W.)
| | - Prashanth Rawla
- Parrish Healthcare, 951 North Washington Ave., Titusville, FL 32796, USA
| |
Collapse
|
3
|
Shinde SD, Chhetri A, Ghosh S, Debnath A, Joshi P, Kumar D. Substrate-Induced Cooperative Ionic Catalysis: Difunctionalization of Indole Derivatives Employing Dimethyl Carbonate. J Org Chem 2024; 89:15995-16003. [PMID: 39432396 DOI: 10.1021/acs.joc.4c01619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
The global urge to adopt sustainable chemistry has resulted in the development of more environmentally benign strategies (EBS) that use CO2 and CO2-derived chemicals in a step-economic manner. In this context, we investigated a dual C-H methylation and (C═O)-methoxylation of indole derivatives using dimethyl carbonate (DMC) in the presence of catalytic amounts of Cs2CO3. Mechanistic insights include DMF-assisted, DMC-induced cooperative ionic catalysis, which allows DMC to act as both a nucleophilic and an electrophilic precursor, resulting in (C═O)-methoxylation and C-H methylation of N-benzylindolyl ketones.
Collapse
Affiliation(s)
- Sangita Dattatray Shinde
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar-382355, Gujarat, India
| | - Ashik Chhetri
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar-382355, Gujarat, India
| | - Sayak Ghosh
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar-382355, Gujarat, India
| | - Anusri Debnath
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar-382355, Gujarat, India
| | - Pooja Joshi
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar-382355, Gujarat, India
| | - Dinesh Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmedabad, Palaj, Gandhinagar-382355, Gujarat, India
| |
Collapse
|
4
|
Ye Z, Zhang Y, Guo G, Shao X, Wu JR. Silver-Catalyzed 1,2-Thiosulfonylation of Alkenes: Development of a Nucleophilic d3-Methylthiolating Reagent. J Org Chem 2024; 89:14369-14383. [PMID: 39323108 DOI: 10.1021/acs.joc.4c01787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Development of robust d3-methylthiolating reagents represents an attractive synthetic method to access deuterated molecules in the field of drug discovery. Here, we report a straightforward strategy to prepare electrophilic S-methyl-d3 arylsulfonothioates in one-step without column purification. These reagents exhibit good radical reactivity toward silver-catalyzed vicinal thiosulfonylation of alkenes or 1,6-enynes on water. As a result, simultaneous incorporation of both SCD3 and ArSO2 units into unsaturated carbon-carbon bonds with 100% atom economy has been established. Moreover, the ATRA adducts with >99% D incorporation can serve as nucleophilic d3-methylthiolating synthons via retro-Michael addition under mild basic conditions, providing a good alternative in trideuteromethylthiolating alkyl iodides to access corresponding trideuteromethyl sulfides with high efficiency.
Collapse
Affiliation(s)
- Zhiyong Ye
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, Hangzhou Normal University, Hangzhou 311121, Zhejiang, People's Republic of China
| | - Yan Zhang
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, Hangzhou Normal University, Hangzhou 311121, Zhejiang, People's Republic of China
| | - Guofang Guo
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, Hangzhou Normal University, Hangzhou 311121, Zhejiang, People's Republic of China
| | - Xinxin Shao
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, Hangzhou Normal University, Hangzhou 311121, Zhejiang, People's Republic of China
- Key Laboratory of Organofluorine Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 345 Lingling Road, Shanghai 200032, People's Republic of China
| | - Ji-Rong Wu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, Hangzhou Normal University, Hangzhou 311121, Zhejiang, People's Republic of China
| |
Collapse
|
5
|
Tan JF, Kang YC, Hartwig JF. Catalytic undirected methylation of unactivated C(sp 3)-H bonds suitable for complex molecules. Nat Commun 2024; 15:8307. [PMID: 39333063 PMCID: PMC11437150 DOI: 10.1038/s41467-024-52245-1] [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: 04/23/2024] [Accepted: 08/28/2024] [Indexed: 09/29/2024] Open
Abstract
In pharmaceutical discovery, the "magic methyl" effect describes a substantial improvement in the pharmacological properties of a drug candidate with the incorporation of methyl groups. Therefore, to expedite the synthesis of methylated drug analogs, late-stage, undirected methylations of C(sp3)-H bonds in complex molecules would be valuable. However, current methods for site-selective methylations are limited to activated C(sp3)-H bonds. Here we describe a site-selective, undirected methylation of unactivated C(sp3)-H bonds, enabled by photochemically activated peroxides and a nickel(II) complex whose turnover is enhanced by an ancillary ligand. The methodology displays compatibility with a wide range of functional groups and a high selectivity for tertiary C-H bonds, making it suitable for the late-stage methylation of complex organic compounds that contain multiple alkyl C-H bonds, such as terpene natural products, peptides, and active pharmaceutical ingredients. Overall, this method provides a synthetic tool to explore the "magic methyl" effect in drug discovery.
Collapse
Affiliation(s)
- Jin-Fay Tan
- Department of Chemistry, University of California, Berkeley, CA, USA
| | - Yi Cheng Kang
- Department of Chemistry, University of California, Berkeley, CA, USA
| | - John F Hartwig
- Department of Chemistry, University of California, Berkeley, CA, USA.
| |
Collapse
|
6
|
Luo Y, He X, Jiang Y, Li J, Wu L, Cai Z, He L. Trideuteromethylthiolation through Reaction of Arynes, S-Methyl- d3 Sulfonothioate with Sulfonamides or Amides: Access to Trideuteromethylated Sulfilimines. J Org Chem 2024; 89:11766-11776. [PMID: 39096290 DOI: 10.1021/acs.joc.4c01033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2024]
Abstract
A direct and practical three-component tandem reaction of arynes, S-methyl-d3 sulfonothioate with sulfonamides or amides is developed. The reaction is highly efficient and chemoselective, which allows mild synthesis of trideuteromethylated sulfilimines with broad substrate scope and good functional group compatibility, giving the products in good to excellent yields with 92%-99% deuterium incorporation. Mechanism studies disclosed sulfenamide that generated in situ is the key intermediate for the reaction. This protocol provides potential method for introduction of -SCD3 moiety for deuteration of marked drugs and drug candidates containing sulfilimine skeleton.
Collapse
Affiliation(s)
- Yuping Luo
- School of Chemistry and Chemical Engineering/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Shihezi University, Shihezi 832003, P. R. China
| | - Xiujuan He
- School of Chemistry and Chemical Engineering/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Shihezi University, Shihezi 832003, P. R. China
| | - Yike Jiang
- School of Chemistry and Chemical Engineering/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Shihezi University, Shihezi 832003, P. R. China
| | - Jie Li
- School of Chemistry and Chemical Engineering/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Shihezi University, Shihezi 832003, P. R. China
| | - Leifang Wu
- Analysis and Testing Center of Shihezi University, Shihezi, Xinjiang Uygur Autonomous Region 832000, P. R. China
| | - Zhihua Cai
- School of Chemistry and Chemical Engineering/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Shihezi University, Shihezi 832003, P. R. China
| | - Lin He
- School of Chemistry and Chemical Engineering/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Shihezi University, Shihezi 832003, P. R. China
| |
Collapse
|
7
|
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
|
8
|
La-Ongthong K, Chantarojsiri T, Soorukram D, Leowanawat P, Reutrakul V, Kuhakarn C. Electro-oxidative Methylation of 2-Isocyanobiaryls Using N,N-dimethylformamide (DMF) as Carbon Source: Synthesis of 6-Methylphenanthridines. Chem Asian J 2024; 19:e202400176. [PMID: 38489229 DOI: 10.1002/asia.202400176] [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: 02/19/2024] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 03/17/2024]
Abstract
A benign electrochemical method to access 6-methylphenanthridines from 2-isocyanobiaryls using N,N-dimethylformamide (DMF) as a methyl source is reported. The protocol operates at ambient temperature without the need for harmful methylating reagents. Mechanistic studies suggested that DMF delivered a methylene synthon, followed by reduction at the cathode and tautomerization. The method offers environmental benefits by avoiding metal-based reagents and harsh conditions.
Collapse
Affiliation(s)
- Kannika La-Ongthong
- Department of Chemistry, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
- Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
| | - Teera Chantarojsiri
- Department of Chemistry, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
- Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
| | - Darunee Soorukram
- Department of Chemistry, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
- Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
| | - Pawaret Leowanawat
- Department of Chemistry, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
- Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
| | - Vichai Reutrakul
- Department of Chemistry, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
- Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
| | - Chutima Kuhakarn
- Department of Chemistry, Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
- Center of Excellence for Innovation in Chemistry (PERCH-CIC), Faculty of Science, Mahidol University, Rama VI Road, Bangkok, 10400, Thailand
| |
Collapse
|
9
|
Wu D, Zeng X, Zhao Y, Qin M, Gong P. Discovery of novel pyridone-benzamide derivatives possessing a 1-methyl-2-benzimidazolinone moiety as potent EZH2 inhibitors for the treatment of B-cell lymphomas. Bioorg Med Chem 2024; 105:117725. [PMID: 38640588 DOI: 10.1016/j.bmc.2024.117725] [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: 01/19/2024] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 04/21/2024]
Abstract
Enhancer of zeste homolog 2 (EZH2) is a promising therapeutic target for diffuse large B-cell lymphoma. In this study, based on the binding model of 1 (tazemetostat) with polycomb repressive complex 2 (PRC2), we designed and synthesized a series of tazemetostat analogs bearing a 1-methyl-2-benzimidazolinone moiety to improve the inhibitory activity of EZH2 wild-type (WT) and Y641 mutants and enhance metabolic stability. After the assessment of the structure-activity relationship at enzymatic and cellular levels, compound N40 was identified. Biochemical assays showed that compound N40 (IC50 = 0.32 nM) exhibited superior inhibitory activity against EZH2 WT, compared with 1 (IC50 = 1.20 nM), and high potency against EZH2 Y641 mutants (EZH2 Y641F, IC50 = 0.03 nM; EZH2 Y641N, IC50 = 0.08 nM), which were approximately 10-fold more active than those of 1 (EZH2 Y641F, IC50 = 0.37 nM; EZH2 Y641N, IC50 = 0.85 nM). Furthermore, compound N40 (IC50 = 3.52 ± 1.23 nM) effectively inhibited the proliferation of Karpas-422 cells and was more potent than 1 (IC50 = 35.01 ± 1.28 nM). Further cellular experiments showed that N40 arrested Karpas-422 cells in the G1 phase and induced apoptosis in a dose-dependent manner. Moreover, N40 inhibited the trimethylation of lysine 27 on histone H3 (H3K27Me3) in Karpas-422 cells bearing the EZH2 Y641N mutant. Additionally, N40 (T1/2 = 177.69 min) showed improved metabolic stability in human liver microsomes compared with 1 (T1/2 = 7.97 min). Our findings suggest N40 as a promising EZH2 inhibitor; further investigation remains warranted to confirm our findings and further develop N40.
Collapse
Affiliation(s)
- Di Wu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xiaoyi Zeng
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yuanhao Zhao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Mingze Qin
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Ping Gong
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
10
|
Chen PH, Guo XS, Zhang HE, Dubey GK, Geng ZZ, Fierke CA, Xu S, Hampton JT, Liu WR. Leveraging a Phage-Encoded Noncanonical Amino Acid: A Novel Pathway to Potent and Selective Epigenetic Reader Protein Inhibitors. ACS CENTRAL SCIENCE 2024; 10:782-792. [PMID: 38680566 PMCID: PMC11046469 DOI: 10.1021/acscentsci.3c01419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 05/01/2024]
Abstract
Epigenetic reader proteins interpret histone epigenetic marks to regulate gene expression. Given their vital roles and the link between their dysfunction and various diseases, these proteins present compelling targets for therapeutic interventions. Nevertheless, designing selective inhibitors for these proteins poses significant challenges, primarily due to their unique properties such as shallow binding sites and similarities with homologous proteins. To overcome these challenges, we propose an innovative strategy that uses phage display with a genetically encoded noncanonical amino acid (ncAA) containing an epigenetic mark. This ncAA guides binding to the reader protein's active site, allowing the identification of peptide inhibitors with enhanced affinity and selectivity. In this study, we demonstrate this novel approach's effectiveness by identifying potent inhibitors for the ENL YEATS domain that plays a critical role in leukemogenesis. Our strategy involved genetically incorporating Nε-butyryl-l-lysine (BuK), known for its binding to ENL YEATS, into a phage display library for enriching the pool of potent inhibitors. One resultant hit was further optimized by substituting BuK with other pharmacophores to exploit a unique π-π-π stacking interaction with ENL YEATS. This led to the creation of selective ENL YEATS inhibitors with a KD value of 2.0 nM and a selectivity 28 times higher for ENL YEATS than its close homologue AF9 YEATS. One such inhibitor, tENL-S1f, demonstrated robust cellular target engagement and on-target effects to inhibit leukemia cell growth and suppress the expression of ENL target genes. As a pioneering study, this work opens up extensive avenues for the development of potent and selective peptidyl inhibitors for a broad spectrum of epigenetic reader proteins.
Collapse
Affiliation(s)
- Peng-Hsun
Chase Chen
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Xuejiao Shirley Guo
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Hanyuan Eric Zhang
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Gopal K. Dubey
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Zhi Zachary Geng
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Carol A. Fierke
- Department
of Biochemistry, Brandeis University, Waltham, Massachusetts 02453, United States
| | - Shiqing Xu
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
- Department
of Pharmaceutical Sciences, Texas A&M
University, College
Station, Texas 77843, United States
| | - J. Trae Hampton
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
| | - Wenshe Ray Liu
- Texas
A&M Drug Discovery Center and Department of Chemistry, Texas A&M University, College Station, Texas 77843, United States
- Department
of Pharmaceutical Sciences, Texas A&M
University, College
Station, Texas 77843, United States
- Institute
of Biosciences and Technology and Department of Translational Medical
Sciences, College of Medicine, Texas A&M
University, Houston, Texas 77030, United States
- Department
of Biochemistry and Biophysics, Texas A&M
University, College
Station, Texas 77843, United States
- Department
of Cell Biology and Genetics, College of Medicine, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
11
|
de Sena Murteira Pinheiro P, Franco LS, Montagnoli TL, Fraga CAM. Molecular hybridization: a powerful tool for multitarget drug discovery. Expert Opin Drug Discov 2024; 19:451-470. [PMID: 38456452 DOI: 10.1080/17460441.2024.2322990] [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: 10/24/2023] [Accepted: 02/21/2024] [Indexed: 03/09/2024]
Abstract
INTRODUCTION The current drug discovery paradigm of 'one drug, multiple targets' has gained attention from both the academic medicinal chemistry community and the pharmaceutical industry. This is in response to the urgent need for effective agents to treat multifactorial chronic diseases. The molecular hybridization strategy is a useful tool that has been widely explored, particularly in the last two decades, for the design of multi-target drugs. AREAS COVERED This review examines the current state of molecular hybridization in guiding the discovery of multitarget small molecules. The article discusses the design strategies and target selection for a multitarget polypharmacology approach to treat various diseases, including cancer, Alzheimer's disease, cardiac arrhythmia, endometriosis, and inflammatory diseases. EXPERT OPINION Although the examples discussed highlight the importance of molecular hybridization for the discovery of multitarget bioactive compounds, it is notorious that the literature has focused on specific classes of targets. This may be due to a deep understanding of the pharmacophore features required for target binding, making targets such as histone deacetylases and cholinesterases frequent starting points. However, it is important to encourage the scientific community to explore diverse combinations of targets using the molecular hybridization strategy.
Collapse
Affiliation(s)
- Pedro de Sena Murteira Pinheiro
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucas Silva Franco
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tadeu Lima Montagnoli
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Alberto Manssour Fraga
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Gao Y, Vakoc CR. Therapeutic index of targeting select chromatin complexes in human cancer patients. Curr Opin Genet Dev 2024; 85:102162. [PMID: 38401489 PMCID: PMC11072572 DOI: 10.1016/j.gde.2024.102162] [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: 09/13/2023] [Revised: 01/21/2024] [Accepted: 01/29/2024] [Indexed: 02/26/2024]
Abstract
Aberrant chromatin regulation can promote the initiation and progression of human cancer. An improved understanding of such mechanisms has resulted in the identification of cancers with an enhanced dependency on specific chromatin regulatory proteins relative to nonmalignant cell types. Hence, targeting of such complexes with small molecules has significant therapeutic potential in oncology. In recent years, several drugs have been developed and evaluated in human cancer patients, which can influence tumor biology by reprogramming of chromatin structure. In this review, we summarize several of the known mechanisms that endow cancer cells with a powerful dependency on chromatin regulation that exceeds the requirements for normal tissue homeostasis. We also summarize the remarkable small-molecule inhibitors that exploit chromatin regulator dependencies with a clear therapeutic benefit in human cancer patients.
Collapse
Affiliation(s)
- Yuan Gao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA. https://twitter.com/@yuangao_yg
| | | |
Collapse
|
13
|
Imhoff RD, Patel R, Safdar MH, Jones HBL, Pinto-Fernandez A, Vendrell I, Chen H, Muli CS, Krabill AD, Kessler BM, Wendt MK, Das C, Flaherty DP. Covalent Fragment Screening and Optimization Identifies the Chloroacetohydrazide Scaffold as Inhibitors for Ubiquitin C-terminal Hydrolase L1. J Med Chem 2024; 67:4496-4524. [PMID: 38488146 DOI: 10.1021/acs.jmedchem.3c01661] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Dysregulation of the ubiquitin-proteasome systems is a hallmark of various disease states including neurodegenerative diseases and cancer. Ubiquitin C-terminal hydrolase L1 (UCHL1), a deubiquitinating enzyme, is expressed primarily in the central nervous system under normal physiological conditions, however, is considered an oncogene in various cancers, including melanoma, lung, breast, and lymphoma. Thus, UCHL1 inhibitors could serve as a viable treatment strategy against these aggressive cancers. Herein, we describe a covalent fragment screen that identified the chloroacetohydrazide scaffold as a covalent UCHL1 inhibitor. Subsequent optimization provided an improved fragment with single-digit micromolar potency against UCHL1 and selectivity over the closely related UCHL3. The molecule demonstrated efficacy in cellular assays of metastasis. Additionally, we report a ligand-bound crystal structure of the most potent molecule in complex with UCHL1, providing insight into the binding mode and information for future optimization.
Collapse
Affiliation(s)
- Ryan D Imhoff
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Drug Discovery, West Lafayette, Indiana 47907, United States
| | - Rishi Patel
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
| | - Muhammad Hassan Safdar
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Hannah B L Jones
- Target Discovery Institute, Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford OX3 7FZ, U.K
| | - Adan Pinto-Fernandez
- Target Discovery Institute, Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford OX3 7FZ, U.K
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, U.K
| | - Iolanda Vendrell
- Target Discovery Institute, Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford OX3 7FZ, U.K
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, U.K
| | - Hao Chen
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Christine S Muli
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Aaron D Krabill
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Benedikt M Kessler
- Target Discovery Institute, Nuffield Department of Medicine, Centre for Medicines Discovery, University of Oxford, Oxford OX3 7FZ, U.K
- Chinese Academy for Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, U.K
| | - Michael K Wendt
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Drug Discovery, West Lafayette, Indiana 47907, United States
- Purdue Institute for Cancer Research, West Lafayette, Indiana 47907, United States
| | - Chittaranjan Das
- Purdue Institute for Drug Discovery, West Lafayette, Indiana 47907, United States
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Cancer Research, West Lafayette, Indiana 47907, United States
| | - Daniel P Flaherty
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue Institute for Drug Discovery, West Lafayette, Indiana 47907, United States
- Purdue Institute for Cancer Research, West Lafayette, Indiana 47907, United States
| |
Collapse
|
14
|
Sharma S, Wang SA, Yang WB, Lin HY, Lai MJ, Chen HC, Kao TY, Hsu FL, Nepali K, Hsu TI, Liou JP. First-in-Class Dual EZH2-HSP90 Inhibitor Eliciting Striking Antiglioblastoma Activity In Vitro and In Vivo. J Med Chem 2024; 67:2963-2985. [PMID: 38285511 PMCID: PMC10895674 DOI: 10.1021/acs.jmedchem.3c02053] [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: 11/03/2023] [Revised: 12/22/2023] [Accepted: 01/09/2024] [Indexed: 01/31/2024]
Abstract
Structural analysis of tazemetostat, an FDA-approved EZH2 inhibitor, led us to pinpoint a suitable site for appendage with a pharmacophoric fragment of second-generation HSP90 inhibitors. Resultantly, a magnificent dual EZH2/HSP90 inhibitor was pinpointed that exerted striking cell growth inhibitory efficacy against TMZ-resistant Glioblastoma (GBM) cell lines. Exhaustive explorations of chemical probe 7 led to several revelations such as (i) compound 7 increased apoptosis/necrosis-related gene expression, whereas decreased M phase/kinetochore/spindle-related gene expression as well as CENPs protein expression in Pt3R cells; (ii) dual inhibitor 7 induced cell cycle arrest at the M phase; (iii) compound 7 suppressed reactive oxygen species (ROS) catabolism pathway, causing the death of TMZ-resistant GBM cells; and (iv) compound 7 elicited substantial in vivo anti-GBM efficacy in experimental mice xenografted with TMZ-resistant Pt3R cells. Collectively, the study results confirm the potential of dual EZH2-HSP90 inhibitor 7 as a tractable anti-GBM agent.
Collapse
Affiliation(s)
- Sachin Sharma
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
| | - Shao-An Wang
- School
of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Wen-Bin Yang
- TMU
Research Center of Neuroscience, Taipei
Medical University, Taipei 110, Taiwan
| | - Hong-Yi Lin
- Graduate
Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Mei-Jung Lai
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
| | - Hsien-Chung Chen
- TMU
Research Center of Neuroscience, Taipei
Medical University, Taipei 110, Taiwan
- Department
of Neurosurgery, Shuang Ho Hospital, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research
Institutes, Taipei 110, Taiwan
| | - Tzu-Yuan Kao
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research
Institutes, Taipei 110, Taiwan
| | - Feng-Lin Hsu
- School
of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Kunal Nepali
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Tsung-I Hsu
- TMU
Research Center of Neuroscience, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
- Ph.D.
Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research
Institutes, Taipei 110, Taiwan
- International
Master Program in Medical Neuroscience, College of Medical Science
and Technology, Taipei Medical University, Taipei 110, Taiwan
- TMU
Research Center of Cancer Translational Medicine, Taipei 110 Taiwan
| | - Jing-Ping Liou
- School
of Pharmacy, College of Pharmacy, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center for Drug Discovery, Taipei
Medical University, Taipei 110, Taiwan
- TMU
Research Center of Cancer Translational Medicine, Taipei 110 Taiwan
- Ph.D.
Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
15
|
Ryabukhin SV, Bondarenko DV, Trofymchuk SA, Lega DA, Volochnyuk DM. Aza-Heterocyclic Building Blocks with In-Ring CF 2 -Fragment. CHEM REC 2024; 24:e202300283. [PMID: 37873869 DOI: 10.1002/tcr.202300283] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/27/2023] [Indexed: 10/25/2023]
Abstract
Modern organic chemistry is a titan supporting and reinforcing pharmaceutical, agricultural, food and material science products. Over the past decades, the organic compounds market has been evolving to meet all the research demands. In this regard, medicinal chemistry is especially dependent on available chemical space as subtle tuning of the molecule structure is required to create a drug with relevant physicochemical properties and a remarkable activity profile. The recent rapid evolution of synthetic methodology to deploy fluorine has brought fluorinated compounds to the spotlight of MedChem community. And now unique properties of fluorine still keep fascinating more and more as its justified installation into a molecular framework has a beneficial impact on membrane permeability, lipophilicity, metabolic stability, pharmacokinetic properties, conformation, pKa , etc. The backward influence of medicinal chemistry on organic synthesis has also changed the landscape of the latter towards new fluorinated topologies as well. Such complex relationships create a flexible and ever-changing ecosystem. Given that MedChem investigations strongly lean on the ability to reach suitable building blocks and the existence of reliable synthetic methods in this review we collected advances in the chemistry of respectful, but still enigmatic gem-difluorinated aza-heterocyclic building blocks.
Collapse
Affiliation(s)
- S V Ryabukhin
- Enamine Ltd., 78 Winston Churchill str., 02094, Kyiv, Ukraine
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska str., 01033, Kyiv, Ukraine
- Institute of Organic Chemistry of the, National Academy of Sciences of Ukraine, 5 Akademik Kukhar str., 02094, Kyiv, Ukraine
| | - D V Bondarenko
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska str., 01033, Kyiv, Ukraine
| | - S A Trofymchuk
- Enamine Ltd., 78 Winston Churchill str., 02094, Kyiv, Ukraine
- Institute of Organic Chemistry of the, National Academy of Sciences of Ukraine, 5 Akademik Kukhar str., 02094, Kyiv, Ukraine
| | - D A Lega
- Enamine Ltd., 78 Winston Churchill str., 02094, Kyiv, Ukraine
- National University of Pharmacy of the Ministry of Health of Ukraine, 53 Pushkinska str., 61002, Kharkiv, Ukraine
| | - D M Volochnyuk
- Enamine Ltd., 78 Winston Churchill str., 02094, Kyiv, Ukraine
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska str., 01033, Kyiv, Ukraine
- Institute of Organic Chemistry of the, National Academy of Sciences of Ukraine, 5 Akademik Kukhar str., 02094, Kyiv, Ukraine
| |
Collapse
|
16
|
Hossain M, Habib I, Singha K, Kumar A. FDA-approved heterocyclic molecules for cancer treatment: Synthesis, dosage, mechanism of action and their adverse effect. Heliyon 2024; 10:e23172. [PMID: 38163206 PMCID: PMC10755292 DOI: 10.1016/j.heliyon.2023.e23172] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
As the incorporation of heterocycles increases the physical characteristics and biological activity of pharmacological molecules, heterocyclic scaffolds are commonly discovered as common cores in a wide spectrum of biologically active drugs. In the contemporary context, many heterocycles have arisen, playing vital roles in diverse pharmaceutical compounds that benefit humanity. Over 85 % of FDA-approved medication molecules contain heterocycles, and most importantly, numerous heterocyclic medicinal molecules indicate potential benefits against a range: of malignancies. The unique flexibility and dynamic core scaffold of these compounds have aided anticancer research. These medications are used to treat cancer patients by targeting particular genes, enzymes, and receptors. Aside from the drugs that are now on the market, numerous forms are being researched for their potential anti-cancer activity. Here in this review, we classified some molecules and biologically active heterocycles containing anticancer medicinal moieties approved by the FDA between 2019 and 2021 based on their use in various forms of cancer. We will focus on those that are suitable for cancer treatment, as well as the essential biochemical mechanisms of action, biological targets, synthetic methods, and inherent limiting considerations in their use.
Collapse
Affiliation(s)
- Mossaraf Hossain
- Synthetic Organic Research Laboratory, UGC-HRDC (Chemistry), University of North Bengal, Darjeeling, 734013, India
| | - Imran Habib
- Synthetic Organic Research Laboratory, UGC-HRDC (Chemistry), University of North Bengal, Darjeeling, 734013, India
| | - Koustav Singha
- Synthetic Organic Research Laboratory, UGC-HRDC (Chemistry), University of North Bengal, Darjeeling, 734013, India
| | - Anoop Kumar
- Department of Biotechnology, University of North Bengal, Darjeeling, 734013, India
| |
Collapse
|
17
|
Cao JM, Liu XY, Rao W, Shen SS, Sheng D, Wang SY. Regioselective Thiol-yne Reaction of Thiol with ((Methyl-d 3)sulfonyl)ethyne: Synthesis of (2-((Methyl-d 3)sulfonyl)vinyl)sulfides. J Org Chem 2024; 89:363-372. [PMID: 38085815 DOI: 10.1021/acs.joc.3c02100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
Herein, we have developed a new method for the synthesis of ((methyl-d3)sulfonyl)ethyne, which is cost-effective and environmentally friendly and can be synthesized at the gram level. As an ideal thiol-yne reagent, it can be reacted with various types of thiols to afford (Z)- and (E)-type vinyl sulfides under different conditions with high selectivity. In addition, it can complete the conformational transition from Z- to E-type products under suitable conditions, and can also carry out further derivatization smoothly. The deuterium content of all products was greater than 99%. The preliminary mechanistic studies support the visible light-mediated radical course, and herein provide a novel and efficient synthetic strategy for the direct introduction of deuterated methyl groups, enriching the methods for the construction of C-S bond-containing compounds.
Collapse
Affiliation(s)
- Ji-Min Cao
- Key Laboratory of Organic Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering and Materials Science & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, P. R. China
| | - Xin-Yu Liu
- Key Laboratory of Organic Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering and Materials Science & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, P. R. China
| | - Weidong Rao
- Key Laboratory of Biomass-based Green Fuels and Chemicals, College of Chemical Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Shu-Su Shen
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, No.99, Xuefu road, Huqiu district,Suzhou 215009, PR China
| | - Daopeng Sheng
- Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shun-Yi Wang
- Key Laboratory of Organic Synthesis of Jiangsu Province, College of Chemistry, Chemical Engineering and Materials Science & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
18
|
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
|
19
|
Vejmelkova K, Pokorna P, Noskova K, Faustmannova A, Drabova K, Pavelka Z, Bajciova V, Broz M, Tinka P, Jezova M, Palova H, Kren L, Valik D, Slaby O, Sterba J. Tazemetostat in the therapy of pediatric INI1-negative malignant rhabdoid tumors. Sci Rep 2023; 13:21623. [PMID: 38062114 PMCID: PMC10703767 DOI: 10.1038/s41598-023-48774-2] [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] [Received: 08/29/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
Rhabdoid tumors are aggressive tumors that may arise in the kidney, soft tissue, central nervous system, or other organs. They are defined by SMARCB1 (INI1) or SMARCA4 alterations. Often, very young children are affected, and the prognosis is dismal. Four patients with primary atypical teratoid rhabdoid tumor (AT/RT, a rhabdoid tumor in the central nervous system) were treated by resection and high dose chemotherapy. Tazemetostat was introduced after completion of chemotherapy. Three patients have achieved an event free survival of 32, 34, and 30 months respectively. One progressed and died. His overall survival was 20 months. One patient was treated for a relapsed atypical teratoid rhabdoid tumor. The treatment combined metronomic therapy, radiotherapy, tazemetostat and immunotherapy. This patient died of disease progression, with an overall survival of 37 months. One patient was treated for a rhabdoid tumor of the ovary. Tazemetostat was given as maintenance after resection, chemotherapy, and radiotherapy, concomitantly with immunotherapy. Her event free survival is 44 months. Only approximately 40% of patients with rhabdoid tumors achieve long-term survival. Nearly all relapses occur within two years from diagnosis. The event free survival of four of the six patients in our cohort has exceeded this timepoint. Tazemetostat has been mostly tested as a single agent in the relapsed setting. We present promising results when applied as maintenance or add on in the first line treatment.
Collapse
Affiliation(s)
- Klara Vejmelkova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 62500, Brno, Czechia.
| | - Petra Pokorna
- Department of Biology, Faculty of Medicine and Central European Institute of Technology, Masaryk University, 62500, Brno, Czechia
| | - Kristyna Noskova
- Department of Hospital Pharmacy, University Hospital Brno and Faculty of Medicine, Masaryk University, 62500, Brno, Czechia
- Department of Pharmacology, Faculty of Medicine, Masaryk University, 62500, Brno, Czechia
| | - Anna Faustmannova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Masaryk University, 62500, Brno, Czechia
| | - Klara Drabova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 62500, Brno, Czechia
| | - Zdenek Pavelka
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 62500, Brno, Czechia
| | - Viera Bajciova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 62500, Brno, Czechia
| | - Martin Broz
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 62500, Brno, Czechia
| | - Pavel Tinka
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 62500, Brno, Czechia
| | - Marta Jezova
- Department of Pathology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, 62500, Czechia
| | - Hana Palova
- Department of Biology, Faculty of Medicine and Central European Institute of Technology, Masaryk University, 62500, Brno, Czechia
| | - Leos Kren
- Department of Pathology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, 62500, Czechia
| | - Dalibor Valik
- Department of Laboratory Medicine, University Hospital Brno and Department of Laboratory Methods, Faculty of Medicine, Masaryk University, 62500, Brno, Czechia
| | - Ondrej Slaby
- Department of Biology, Faculty of Medicine and Central European Institute of Technology, Masaryk University, 62500, Brno, Czechia
| | - Jaroslav Sterba
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, 62500, Brno, Czechia
| |
Collapse
|
20
|
Pinheiro PDSM, Franco LS, Fraga CAM. The Magic Methyl and Its Tricks in Drug Discovery and Development. Pharmaceuticals (Basel) 2023; 16:1157. [PMID: 37631072 PMCID: PMC10457765 DOI: 10.3390/ph16081157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/06/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
One of the key scientific aspects of small-molecule drug discovery and development is the analysis of the relationship between its chemical structure and biological activity. Understanding the effects that lead to significant changes in biological activity is of paramount importance for the rational design and optimization of bioactive molecules. The "methylation effect", or the "magic methyl" effect, is a factor that stands out due to the number of examples that demonstrate profound changes in either pharmacodynamic or pharmacokinetic properties. In many cases, this has been carried out rationally, but in others it has been the product of serendipitous observations. This paper summarizes recent examples that provide an overview of the current state of the art and contribute to a better understanding of the methylation effect in bioactive small-molecule drug candidates.
Collapse
Affiliation(s)
- Pedro de Sena Murteira Pinheiro
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (P.d.S.M.P.); (L.S.F.)
| | - Lucas Silva Franco
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (P.d.S.M.P.); (L.S.F.)
- Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), CCS, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil
| | - Carlos Alberto Manssour Fraga
- Laboratório de Avaliação e Síntese de Substâncias Bioativas (LASSBio), Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (P.d.S.M.P.); (L.S.F.)
- Instituto Nacional de Ciência e Tecnologia de Fármacos e Medicamentos (INCT-INOFAR), CCS, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil
- Programa de Pós-Graduação em Farmacologia e Química Medicinal, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Cidade Universitária, Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
21
|
Zhang Q, Wu YX, Yu XQ, Zhang BY, Ma LY. EZH2 serves as a promising therapeutic target for fibrosis. Bioorg Chem 2023; 137:106578. [PMID: 37156135 DOI: 10.1016/j.bioorg.2023.106578] [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: 03/23/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/10/2023]
Abstract
Fibrosis affects the function of many organs and tissues, and its persistent development can lead to tissue sclerosis and cancer, even leading to death further. Recent studies suggested that enhancer of zeste homolog 2 (EZH2), a major regulator of epigenetic repression, played an important role in the occurrence and development of fibrosis through gene silencing or transcriptional activation. As the most studied and powerful pro-fibrotic cytokine closely related to EZH2, TGF-β1 was primarily involved in the regulation of fibrosis along with the typical Smads and non-Smads signaling pathways. In addition, EZH2 inhibitors demonstrated inhibitory effects in several types of fibrosis. This review summarized the relationship underlying the action of EZH2, TGF-β1/Smads, and TGF-β1/non-Smads with fibrosis and described the research progress of EZH2 inhibitors in the treatment of fibrosis.
Collapse
Affiliation(s)
- Qian Zhang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Ya-Xi Wu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Xiao-Qian Yu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Bao-Yin Zhang
- Department of Pharmacy, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Li-Ying Ma
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450001, PR China; China Meheco Topfond Pharmaceutical Co., Key Laboratory of Cardio-cerebrovascular Drug, Zhumadian 463000, PR China.
| |
Collapse
|
22
|
Wan G, Feng H, Su C, Zhu Y, Zhang L, Zhang Q, Yu L. A patent review of EZH2 inhibitors from 2017 and beyond. Expert Opin Ther Pat 2023; 33:293-308. [PMID: 37095742 DOI: 10.1080/13543776.2023.2206018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
INTRODUCTION EZH2 is an important epigenetic regulator that forms the PRC2 complex with SUZ12, EED and RbAp46/48. As the key catalytic subunit of PRC2, EZH2 regulates the trimethylation of histone H3K27, which in turn promotes chromatin condensation and represses the transcription of relevant target genes. EZH2 overexpression and mutations are strictly related to tumor proliferation, invasion and metastasis. Currently, a large number of highly specific EZH2 inhibitors have been developed and some have already been in clinical trials. AREAS COVERED The aim of the present review is to provide an overview of the molecular mechanisms of EZH2 inhibitors and to highlight the research advances in the patent literature published from 2017 to date. A search of the literature and patents for EZH2 inhibitors and degraders was performed using the Web of Science, SCIFinder, WIPO, USPTO, EPO and CNIPA databases. EXPERT OPINION In recent years, a great number of structurally diverse EZH2 inhibitors have been identified, including EZH2 reversible inhibitors, EZH2 irreversible inhibitors, EZH2-based dual inhibitors and EZH2 degraders. Despite the multiple challenges, EZH2 inhibitors offer promising potential for the treatment of various diseases, such as cancers.
Collapse
Affiliation(s)
- Guoquan Wan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section Ren Min South Road, Chengdu 610041, P. R. China
| | - Huan Feng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section Ren Min South Road, Chengdu 610041, P. R. China
| | - Chang Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section Ren Min South Road, Chengdu 610041, P. R. China
| | - Yongxia Zhu
- Department of Clinical Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, P. R. China
| | - Lidan Zhang
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Qiangsheng Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section Ren Min South Road, Chengdu 610041, P. R. China
| | - Luoting Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section Ren Min South Road, Chengdu 610041, P. R. China
| |
Collapse
|
23
|
Chiodi D, Ishihara Y. "Magic Chloro": Profound Effects of the Chlorine Atom in Drug Discovery. J Med Chem 2023; 66:5305-5331. [PMID: 37014977 DOI: 10.1021/acs.jmedchem.2c02015] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2023]
Abstract
Chlorine is one of the most common atoms present in small-molecule drugs beyond carbon, hydrogen, nitrogen, and oxygen. There are currently more than 250 FDA-approved chlorine-containing drugs, yet the beneficial effect of the chloro substituent has not yet been reviewed. The seemingly simple substitution of a hydrogen atom (R = H) with a chlorine atom (R = Cl) can result in remarkable improvements in potency of up to 100,000-fold and can lead to profound effects on pharmacokinetic parameters including clearance, half-life, and drug exposure in vivo. Following the literature terminology of the "magic methyl effect" in drugs, the term "magic chloro effect" has been coined herein. Although reports of 500-fold or 1000-fold potency improvements are often serendipitous discoveries that can be considered "magical" rather than planned, hypotheses made to explain the magic chloro effect can lead to lessons that accelerate the cycle of drug discovery.
Collapse
Affiliation(s)
- Debora Chiodi
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Yoshihiro Ishihara
- Department of Chemistry, Vividion Therapeutics, 5820 Nancy Ridge Drive, San Diego, California 92121, United States
| |
Collapse
|
24
|
Pashenko AE, Gaidai A, Hryhoriev N, Volovenko O, Levandovskiy I, Maksymenko O, Volochnyuk DM, Ryabukhin SV. Scale-Up Synthesis of 1-Methyladamantane and Its Functionalization as a Key Point for Promising Antiviral Agents. Org Process Res Dev 2023. [DOI: 10.1021/acs.oprd.2c00305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023]
Affiliation(s)
- Alexander E. Pashenko
- Enamine Ltd., 78 Chervonotkatska Street, 02660 Kyiv, Ukraine
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska Street, 01033 Kyiv, Ukraine
- Institute of Organic Chemistry, National Academy of Sciences of Ukraine, 5 Murmanska Street, 02660 Kyiv, Ukraine
| | - Alexandr Gaidai
- Enamine Ltd., 78 Chervonotkatska Street, 02660 Kyiv, Ukraine
- Institute of Organic Chemistry, National Academy of Sciences of Ukraine, 5 Murmanska Street, 02660 Kyiv, Ukraine
- National Technical University of Ukraine “Igor Sikorsky Kyiv Polytechnic Institute”, 37 Peremohy Avenue, 03056 Kyiv, Ukraine
| | - Nazar Hryhoriev
- Enamine Ltd., 78 Chervonotkatska Street, 02660 Kyiv, Ukraine
- National Technical University of Ukraine “Igor Sikorsky Kyiv Polytechnic Institute”, 37 Peremohy Avenue, 03056 Kyiv, Ukraine
| | | | - Igor Levandovskiy
- National Technical University of Ukraine “Igor Sikorsky Kyiv Polytechnic Institute”, 37 Peremohy Avenue, 03056 Kyiv, Ukraine
| | - Olga Maksymenko
- Enamine Ltd., 78 Chervonotkatska Street, 02660 Kyiv, Ukraine
| | - Dmitriy M. Volochnyuk
- Enamine Ltd., 78 Chervonotkatska Street, 02660 Kyiv, Ukraine
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska Street, 01033 Kyiv, Ukraine
- Institute of Organic Chemistry, National Academy of Sciences of Ukraine, 5 Murmanska Street, 02660 Kyiv, Ukraine
| | - Sergey V. Ryabukhin
- Enamine Ltd., 78 Chervonotkatska Street, 02660 Kyiv, Ukraine
- Taras Shevchenko National University of Kyiv, 60 Volodymyrska Street, 01033 Kyiv, Ukraine
- Institute of Organic Chemistry, National Academy of Sciences of Ukraine, 5 Murmanska Street, 02660 Kyiv, Ukraine
| |
Collapse
|
25
|
Sutopo NC, Kim JH, Cho JY. Role of histone methylation in skin cancers: Histone methylation-modifying enzymes as a new class of targets for skin cancer treatment. Biochim Biophys Acta Rev Cancer 2023; 1878:188865. [PMID: 36841366 DOI: 10.1016/j.bbcan.2023.188865] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/17/2023] [Accepted: 02/17/2023] [Indexed: 02/27/2023]
Abstract
Histone methylation, one of the most prominent epigenetic modifications, plays a vital role in gene transcription, and aberrant histone methylation levels cause tumorigenesis. Histone methylation is a reversible enzyme-dependent reaction, and histone methyltransferases and demethylases are involved in this reaction. This review addresses the biological and clinical relevance of these histone methylation-modifying enzymes for skin cancer. In particular, the roles of histone lysine methyltransferases, histone arginine methyltransferase, lysine-specific demethylases, and JmjC demethylases in skin cancer are discussed in detail. In addition, we summarize the efficacy of several epigenetic inhibitors targeting histone methylation-modifying enzymes in cutaneous cancers, such as basal cell carcinoma (BCC), squamous cell carcinoma (SCC), and melanoma. In conclusion, we propose histone methylation-modifying enzymes as novel targets for next-generation pharmaceuticals in the treatment of skin cancers and further provide a rationale for the development of epigenetic drugs (epidrugs) that target specific histone methylases/demethylases in cutaneous tumors.
Collapse
Affiliation(s)
| | - Ji Hye Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Jae Youl Cho
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea; Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
26
|
Zhang Q, Chen X, Cao J, Yang W, Wan G, Feng Q, Zhou S, Yang H, Wang N, Liu Z, Xiao H, Zhu Y, Yu L. Discovery of a Novel Covalent EZH2 Inhibitor Based on Tazemetostat Scaffold for the Treatment of Ovarian Cancer. J Med Chem 2023; 66:1725-1741. [PMID: 36692394 DOI: 10.1021/acs.jmedchem.2c01370] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Enhancer of zeste homologue 2 (EZH2) is the enzymatic catalytic subunit of polycomb repressive complex 2 (PRC2), which plays an important role in post-translational modifications of histones. In this study, we designed and synthesized a new series EZH2 covalent inhibitors that have rarely been reported. Biochemical studies and mass spectrometry provide information that SKLB-03220 could covalently bind to the S-adenosylmethionine (SAM) pocket of EZH2. Besides, SKLB-03220 was highly potent for EZH2MUT, while exhibiting weak activities against other tested histone methyltransferases (HMTs) and kinases. Moreover, SKLB-03220 displayed noteworthy potency against ovarian cancer cell lines and continuously abolished H3K27me3 after washing out. Furthermore, oral administration of SKLB-03220 significantly inhibited tumor growth in PA-1 xenograft model without obvious adverse effects. Taken together, SKLB-03220 is a potent, selective EZH2 covalent inhibitor with noteworthy anticancer efficacy both in vitro and in vivo.
Collapse
Affiliation(s)
- Qiangsheng Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, P. R. China
| | - Xinyi Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, P. R. China
| | - Jiaying Cao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, P. R. China
| | - Wan Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, P. R. China
| | - Guoquan Wan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, P. R. China
| | - Qiang Feng
- College of Chemistry and Life Science, Chengdu Normal University, Chengdu 611130, P. R. China
| | - Shuyan Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, P. R. China
| | - Hongling Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, P. R. China
| | - Ningyu Wang
- School of Life Science and Engineering, Southwest JiaoTong University, Chengdu 611756, Sichuan, P. R. China
| | - Zhihao Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, P. R. China
- Laboratory of Emergency Medicine, Department of Emergency Medicine, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Hongtao Xiao
- Department of Clinical Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, P. R. China
| | - Yongxia Zhu
- Department of Clinical Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, P. R. China
| | - Luoting Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, P. R. China
| |
Collapse
|
27
|
Hogg A, Wheatley M, Domingo-Legarda P, Carral-Menoyo A, Cottam N, Larrosa I. Ruthenium-Catalyzed Monoselective C-H Methylation and d 3-Methylation of Arenes. JACS AU 2022; 2:2529-2538. [PMID: 36465534 PMCID: PMC9709947 DOI: 10.1021/jacsau.2c00399] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/21/2022] [Accepted: 10/03/2022] [Indexed: 05/26/2023]
Abstract
Site-selective installation of C-Me bonds remains a powerful and sought-after tool to alter the chemical and pharmacological properties of a molecule. Direct C-H functionalization provides an attractive means of achieving this transformation. Such protocols, however, typically utilize harsh conditions and hazardous methylating agents with poor applicability toward late-stage functionalization. Furthermore, highly monoselective methylation protocols remain scarce. Herein, we report an efficient monoselective, directed ortho-methylation of arenes using N,N,N-trimethylanilinium salts as noncarcinogenic, bench-stable methylating agents. We extend this protocol to d 3-methylation in addition to the late-stage functionalization of pharmaceutically active compounds. Detailed kinetic studies indicate the rate-limiting in situ formation of MeI is integral to the observed reactivity.
Collapse
|
28
|
Lu D, Wang C, Qu L, Yin F, Li S, Luo H, Zhang Y, Liu X, Chen X, Luo Z, Cui N, Kong L, Wang X. Histone Deacetylase and Enhancer of Zeste Homologue 2 Dual Inhibitors Presenting a Synergistic Effect for the Treatment of Hematological Malignancies. J Med Chem 2022; 65:12838-12859. [DOI: 10.1021/acs.jmedchem.2c00673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Dehua Lu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Cheng Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lailiang Qu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Fucheng Yin
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shang Li
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Heng Luo
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yonglei Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xingchen Liu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xinye Chen
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhongwen Luo
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ningjie Cui
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaobing Wang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
29
|
Wu MC, Li MZ, Chen YX, Liu F, Xiao JA, Chen K, Xiang HY, Yang H. Photoredox-Catalyzed C–H Trideuteromethylation of Quinoxalin-2(1 H)-ones with CDCl 3 as the “CD 3” source. Org Lett 2022; 24:6412-6416. [DOI: 10.1021/acs.orglett.2c02439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Mei-Chun Wu
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
- College of Chemistry and Chemical Engineering, Huaihua University, Huaihua 418008, P. R. China
| | - Ming-Zhi Li
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
| | - Yi-Xuan Chen
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
| | - Fang Liu
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
| | - Jun-An Xiao
- College of Chemistry and Materials Science, Nanning Normal University, Nanning 530001, P. R. China
| | - Kai Chen
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
| | - Hao-Yue Xiang
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, P. R. China
| | - Hua Yang
- College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, P. R. China
| |
Collapse
|
30
|
Cao YC, Shan SK, Guo B, Li CC, Li FXZ, Zheng MH, Xu QS, Wang Y, Lei LM, Tang KX, Ou-Yang WL, Duan JY, Wu YY, Ullah MHE, Zhou ZA, Xu F, Lin X, Wu F, Liao XB, Yuan LQ. Histone Lysine Methylation Modification and Its Role in Vascular Calcification. Front Endocrinol (Lausanne) 2022; 13:863708. [PMID: 35784574 PMCID: PMC9243330 DOI: 10.3389/fendo.2022.863708] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/06/2022] [Indexed: 01/10/2023] Open
Abstract
Histone methylation is an epigenetic change mediated by histone methyltransferase, and has been connected to the beginning and progression of several diseases. The most common ailments that affect the elderly are cardiovascular and cerebrovascular disorders. They are the leading causes of death, and their incidence is linked to vascular calcification (VC). The key mechanism of VC is the transformation of vascular smooth muscle cells (VSMCs) into osteoblast-like phenotypes, which is a highly adjustable process involving a variety of complex pathophysiological processes, such as metabolic abnormalities, apoptosis, oxidative stress and signalling pathways. Many researchers have investigated the mechanism of VC and related targets for the prevention and treatment of cardiovascular and cerebrovascular diseases. Their findings revealed that histone lysine methylation modification may play a key role in the various stages of VC. As a result, a thorough examination of the role and mechanism of lysine methylation modification in physiological and pathological states is critical, not only for identifying specific molecular markers of VC and new therapeutic targets, but also for directing the development of new related drugs. Finally, we provide this review to discover the association between histone methylation modification and VC, as well as diverse approaches with which to investigate the pathophysiology of VC and prospective treatment possibilities.
Collapse
Affiliation(s)
- Ye-Chi Cao
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Su-Kang Shan
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bei Guo
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chang-Chun Li
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fu-Xing-Zi Li
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming-Hui Zheng
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiu-Shuang Xu
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Wang
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Li-Min Lei
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ke-Xin Tang
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Lu Ou-Yang
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Yue Duan
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yun-Yun Wu
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Muhammad Hasnain Ehsan Ullah
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhi-Ang Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Xu
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Lin
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Wu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Bo Liao
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ling-Qing Yuan
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
31
|
Xia J, Li J, Tian L, Ren X, Liu C, Liang C. Targeting Enhancer of Zeste Homolog 2 for the Treatment of Hematological Malignancies and Solid Tumors: Candidate Structure–Activity Relationships Insights and Evolution Prospects. J Med Chem 2022; 65:7016-7043. [DOI: 10.1021/acs.jmedchem.2c00047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Juan Xia
- Laboratory of Hematologic Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, P. R. China
| | - Jingyi Li
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi’an 710021, P. R. China
| | - Lei Tian
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi’an 710021, P. R. China
- College of Bioresources Chemical and Materials Engineering, Shaanxi University of Science & Technology, Xi’an 710021, P. R. China
| | - Xiaodong Ren
- Medical College, Guizhou University, Guiyang 550025, P. R. China
| | - Chang Liu
- Zhuhai Jinan Selenium Source Nanotechnology Co., Ltd., Zhuhai 519030, P. R. China
| | - Chengyuan Liang
- Faculty of Pharmacy, Shaanxi University of Science & Technology, Xi’an 710021, P. R. China
| |
Collapse
|
32
|
Abstract
C−H methylation of sp2 and sp3 carbon centers is significant in many biological processes. Methylated drug candidates show unique properties due to the change in solubility, conformation and metabolic activities. Several photo-catalyzed, electrochemical, mechanochemical and metal-free techniques that are widely utilized strategies in medicinal chemistry for methylation of arenes and heteroarenes have been covered in this review.
Collapse
|
33
|
Zeng J, Zhang J, Sun Y, Wang J, Ren C, Banerjee S, Ouyang L, Wang Y. Targeting EZH2 for cancer therapy: From current progress to novel strategies. Eur J Med Chem 2022; 238:114419. [DOI: 10.1016/j.ejmech.2022.114419] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 12/14/2022]
|
34
|
Identification of SET/EED dual binders as innovative PRC2 inhibitors. Future Med Chem 2022; 14:609-621. [DOI: 10.4155/fmc-2022-0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background: The inhibition of PRC2, implicated in the pathogenesis of several tumors, can be a useful therapeutic strategy for cancer treatment. In the literature, two types of PRC2 modulators are reported: competitive inhibitors of S-adenosyl methionine binding to the catalytic subunit EZH2; and allosteric ligands that prevent the interaction of the trimethylated H3K27 lysine in histone 3 to the EED subunit. The lack of dual EZH2/EED modulators drove us to search for compounds capable of recognizing both domains. Materials & methods: This goal was pursued by combining pharmacophore- and docking-based virtual screening of the Multi-Target Ligand Chemotheca database. Prediction tools for absorption, distribution, metabolism and excretion and pan-assay interference compounds were also applied. Results: Finally, five 1,2,3-triazole derivatives were identified as promising dual EZH2/EED modulators. Conclusion: Our multistage screening protocol highlighted the great potential of Chemotheca for identifying polypharmacological agents.
Collapse
|
35
|
Liang R, Tomita D, Sasaki Y, Ginn J, Michino M, Huggins DJ, Baxt L, Kargman S, Shahid M, Aso K, Duggan M, Stamford AW, DeStanchina E, Liverton N, Meinke PT, Foley MA, Phillips RE. A Chemical Strategy toward Novel Brain-Penetrant EZH2 Inhibitors. ACS Med Chem Lett 2022; 13:377-387. [PMID: 35300079 PMCID: PMC8919293 DOI: 10.1021/acsmedchemlett.1c00448] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 01/24/2022] [Indexed: 12/14/2022] Open
Abstract
Aberrant gene-silencing through dysregulation of polycomb protein activity has emerged as an important oncogenic mechanism in cancer, implicating polycomb proteins as important therapeutic targets. Recently, an inhibitor targeting EZH2, the methyltransferase component of PRC2, received U.S. Food and Drug Administration approval following promising clinical responses in cancer patients. However, the current array of EZH2 inhibitors have poor brain penetrance, limiting their use in patients with central nervous system malignancies, a number of which have been shown to be sensitive to EZH2 inhibition. To address this need, we have identified a chemical strategy, based on computational modeling of pyridone-containing EZH2 inhibitor scaffolds, to minimize P-glycoprotein activity, and here we report the first brain-penetrant EZH2 inhibitor, TDI-6118 (compound 5). Additionally, in the course of our attempts to optimize this compound, we discovered TDI-11904 (compound 21), a novel, highly potent, and peripherally active EZH2 inhibitor based on a 7 member ring structure.
Collapse
Affiliation(s)
- Rui Liang
- Tri-Institutional Therapeutics Discovery Institute, 413 East 69th Street, New York, New York 10021, United States
| | - Daisuke Tomita
- Tri-Institutional Therapeutics Discovery Institute, 413 East 69th Street, New York, New York 10021, United States
| | - Yusuke Sasaki
- Tri-Institutional Therapeutics Discovery Institute, 413 East 69th Street, New York, New York 10021, United States
| | - John Ginn
- Tri-Institutional Therapeutics Discovery Institute, 413 East 69th Street, New York, New York 10021, United States
| | - Mayako Michino
- Tri-Institutional Therapeutics Discovery Institute, 413 East 69th Street, New York, New York 10021, United States
| | - David J Huggins
- Tri-Institutional Therapeutics Discovery Institute, 413 East 69th Street, New York, New York 10021, United States.,Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York 10021, United States
| | - Leigh Baxt
- Tri-Institutional Therapeutics Discovery Institute, 413 East 69th Street, New York, New York 10021, United States
| | - Stacia Kargman
- Tri-Institutional Therapeutics Discovery Institute, 413 East 69th Street, New York, New York 10021, United States
| | - Maaz Shahid
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Epigenetics Program, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States.,Abramson Cancer Center, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Kazuyoshi Aso
- Tri-Institutional Therapeutics Discovery Institute, 413 East 69th Street, New York, New York 10021, United States
| | - Mark Duggan
- LifeSci Consulting, LLC., 18243 SE Ridgeview Drive, Tequesta, Florida 33469, United States
| | - Andrew W Stamford
- Tri-Institutional Therapeutics Discovery Institute, 413 East 69th Street, New York, New York 10021, United States
| | - Elisa DeStanchina
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Nigel Liverton
- Tri-Institutional Therapeutics Discovery Institute, 413 East 69th Street, New York, New York 10021, United States
| | - Peter T Meinke
- Tri-Institutional Therapeutics Discovery Institute, 413 East 69th Street, New York, New York 10021, United States.,Department of Pharmacology, Weill Cornell Medical College, New York, New York 10021, United States
| | - Michael A Foley
- Tri-Institutional Therapeutics Discovery Institute, 413 East 69th Street, New York, New York 10021, United States
| | - Richard E Phillips
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Epigenetics Program, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States.,Abramson Cancer Center, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
36
|
Zhang Q, Yang H, Feng Q, Cao J, Zhang Y, Li L, Yu L. Focus on the classical and non-classical functions of EZH2: Guide the development of inhibitors and degraders. Pharmacol Res 2022; 178:106159. [DOI: 10.1016/j.phrs.2022.106159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 12/16/2022]
|
37
|
Xiao X, Huang YQ, Tian HY, Bai J, Cheng F, Wang X, Ke ML, Chen FE. Robust, scalable construction of an electrophilic deuterated methylthiolating reagent: facile access to SCD 3-containing scaffolds. Chem Commun (Camb) 2022; 58:3015-3018. [PMID: 35147615 DOI: 10.1039/d1cc07184j] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We have established a practical and concise method for the straightforward access of a universal deuterated methylthiolating reagent through a one-pot gram-scale operation under mild conditions. This odourless electrophilic SCD3 reagent was widely applied to react with numerous representative nucleophiles and approached various valuable SCD3 analogues with excellent levels of deuterium content (>99% D). The divergent further transformations were smoothly carried out to obtain the significant derivatives with different oxidative states in high efficiency.
Collapse
Affiliation(s)
- Xiao Xiao
- Institute of Pharmaceutical Science and Technology, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China.
| | - Yin-Qiu Huang
- Institute of Pharmaceutical Science and Technology, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China.
| | - Hong-Yu Tian
- Institute of Pharmaceutical Science and Technology, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China.
| | - Jun Bai
- Institute of Pharmaceutical Science and Technology, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China.
| | - Fei Cheng
- Institute of Pharmaceutical Science and Technology, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China.
| | - Xu Wang
- Institute of Pharmaceutical Science and Technology, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China.
| | - Miao-Lin Ke
- Institute of Pharmaceutical Science and Technology, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China.
| | - Fen-Er Chen
- Institute of Pharmaceutical Science and Technology, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China. .,Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry Fudan University, 220 Handan Road, Shanghai 200433, People's Republic of China.,Shanghai Engineering Center of Industrial Asymmetric Catalysis for Chiral Drugs, Shanghai 200433, People's Republic of China
| |
Collapse
|
38
|
Hu X, Zhang J, Zhang Y, Jiao F, Wang J, Chen H, Ouyang L, Wang Y. Dual-target inhibitors of poly (ADP-ribose) polymerase-1 for cancer therapy: Advances, challenges, and opportunities. Eur J Med Chem 2022; 230:114094. [PMID: 34998039 DOI: 10.1016/j.ejmech.2021.114094] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/27/2021] [Accepted: 12/27/2021] [Indexed: 02/08/2023]
Abstract
PARP1 plays a crucial role in DNA damage repair, making it an essential target for cancer therapy. PARP1 inhibitors are widely used to treat BRCA-deficient malignancies, and six PARP inhibitors have been approved for clinical use. However, excluding the great clinical success of PARP inhibitors, the concomitant toxicity, drug resistance, and limited scope of application restrict their clinical efficacy. To find solutions to these problems, dual-target inhibitors have shown great potential. In recent years, several studies have linked PAPR1 to other primary cancer targets. Many dual-target inhibitors have been developed using structural fusion, linkage, or library construction methods, overcoming the defects of many single-target inhibitors of PARP1 and achieving great success in clinical cancer therapy. This review summarizes the advance of dual-target PARP1 inhibitors in recent years, focusing on their structural optimization process, structure-activity relationships (SARs), and in vitro or in vivo analysis results.
Collapse
Affiliation(s)
- Xinyue Hu
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Jifa Zhang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ya Zhang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Fulun Jiao
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, Tennessee, United States
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, 38163, Tennessee, United States
| | - Liang Ouyang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuxi Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
39
|
Abstract
Abstract:
Dimethyl sulfoxide (DMSO) is not only a common and cheap aprotic polar solvent with low toxicity, but also serves as an efficient and multipurpose reactant and has widely been used in organic synthesis. DMSO as an important precursor can effectively introducea broad range of functional fragments into organic molecules, such as -Me, -CH, -CH2, -SMe2, -CH2SMe, -CH2SOMe, -SMe, -SO2Me, -SOMe or as O substituents, and serves as a mild oxidant in organic transformations. Many significant achievements based on DMSO as a synthon in synthetic chemistry have rapidly made over the past several years. To help researchers further understand the recent advances in the field, the review summarizes the applications of DMSO as carbon, sulfur, and oxygen sources and is used as the dual synthon in synthetic transformations.
Collapse
Affiliation(s)
- Su-qian CAI
- School of Pharmaceutical Sciences, Guilin Medical University, Guangxi Guilin 541199, P. R. China,
| | - Ke-feng ZHANG
- School of Pharmaceutical Sciences, Guilin Medical University, Guangxi Guilin 541199, P. R. China,
| | - Xiao-hua CAI
- School of Chemical Engineering, Guizhou Minzu University, Guizhou, Guiyang 550025, P. R., China
| |
Collapse
|
40
|
Feng Q, Zhang Q, He H, Zhang L, Chang B, Yu L, Zhang X. A Facile Total Synthesis of Kilogram-Scale Production of SKLB1039: A Novel and Selective Hexahydroisoquinolin-Containing EZH2 Inhibitor. Curr Org Synth 2022; 19:583-590. [PMID: 34994315 DOI: 10.2174/1570179419666220107161257] [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: 09/16/2021] [Revised: 12/03/2021] [Accepted: 12/17/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND SKLB1039 is a potent, highly selective, and orally bioavailable EZH2 inhibitor, which significantly inhibited breast tumor growth and metastasis, in pre-clinical studies. In a previously reported synthesis of SKLB1039, the yields of several steps were low which led to an overall yield of less than 10%. In addition, flash chromatography was required for the purification of several intermediates using this route. OBJECTIVE To optimize the synthesis and establish an efficient commercial-scale method for the production of SKLB1039. METHODS The reaction time, solvent, reactant ratio, temperature, and mode of addition of reactants in the reductive amination, hydrolysis, hexahydroisoquinoline formation, hydrogenolysis, condensation and Suzuki cross-coupling reactions were optimized. RESULTS A chromatography-free seven-step process starting from a commercially available material was developed that afforded SKLB1039 in 36% overall yield with > 99% purity. CONCLUSION A cost-effective, high-yielding, and convergent kilo-scale synthesis for the EZH2 inhibitor SKLB1039 was developed. The operation was simple, and the pure product was easily obtained without column chromatography. This method will be economical and convenient for the subsequent industrial scale-up production of SKLB1039, which will be conducive for this promising EZH2 inhibitor to enter clinical studies of its anti-tumor effects.
Collapse
Affiliation(s)
- Qiang Feng
- Colledge of Chemistry and Life Science,Sichuan Provincial Key Laboratory for Structural Optimization and Application of Functional Molecules,Chengdu Normal University,Chengdu, 611130,China
| | - Qiangsheng Zhang
- State Key Laboratory of Biotherapy/Collaborative Innovation Centor of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Hualong He
- State Key Laboratory of Biotherapy/Collaborative Innovation Centor of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Lidan Zhang
- State Key Laboratory of Biotherapy/Collaborative Innovation Centor of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Bo Chang
- Colledge 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/Collaborative Innovation Centor of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Xiaoling Zhang
- Colledge of Chemistry and Life Science,Sichuan Provincial Key Laboratory for Structural Optimization and Application of Functional Molecules,Chengdu Normal University,Chengdu, 611130,China
| |
Collapse
|
41
|
Azimi S, Khuttan S, Wu JZ, Pal RK, Gallicchio E. Relative Binding Free Energy Calculations for Ligands with Diverse Scaffolds with the Alchemical Transfer Method. J Chem Inf Model 2022; 62:309-323. [PMID: 34990555 DOI: 10.1021/acs.jcim.1c01129] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We present an extension of the alchemical transfer method (ATM) for the estimation of relative binding free energies of molecular complexes applicable to conventional, as well as scaffold-hopping, alchemical transformations. Named ATM-RBFE, the method is implemented in the free and open-source OpenMM molecular simulation package and aims to provide a simpler and more generally applicable route to the calculation of relative binding free energies than what is currently available. ATM-RBFE is based on sound statistical mechanics theory and a novel coordinate perturbation scheme designed to swap the positions of a pair of ligands such that one is transferred from the bulk solvent to the receptor binding site while the other moves simultaneously in the opposite direction. The calculation is conducted directly in a single solvent box with a system prepared with conventional setup tools, without splitting of electrostatic and nonelectrostatic transformations, and without pairwise soft-core potentials. ATM-RBFE is validated here against the absolute binding free energies of the SAMPL8 GDCC host-guest benchmark set and against protein-ligand benchmark sets that include complexes of the estrogen receptor ERα and those of the methyltransferase EZH2. In each case the method yields self-consistent and converged relative binding free energy estimates in agreement with absolute binding free energies and reference literature values, as well as experimental measurements.
Collapse
Affiliation(s)
- Solmaz Azimi
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, New York 11210, United States.,Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
| | - Sheenam Khuttan
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, New York 11210, United States.,Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
| | - Joe Z Wu
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, New York 11210, United States.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
| | - Rajat K Pal
- Roivant Sciences, Inc., Boston, Massachusetts 02210, United States
| | - Emilio Gallicchio
- Department of Chemistry, Brooklyn College of the City University of New York, Brooklyn, New York 11210, United States.,Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States.,Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
| |
Collapse
|
42
|
Zhang Q, Chen X, Hu X, Duan X, Wan G, Li L, Feng Q, Zhang Y, Wang N, Yu L. Covalent inhibitors of EZH2: Design, synthesis and evaluation. Biomed Pharmacother 2022; 147:112617. [PMID: 34998031 DOI: 10.1016/j.biopha.2022.112617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/31/2021] [Accepted: 01/02/2022] [Indexed: 02/05/2023] Open
Abstract
The histone lysine methyltransferase EZH2 has been implicated as a key component in cancer development. Up to date, there are only a few EZH2 covalent inhibitors. In this study, a new series of 3-acrylamido-2-methyl-N-((2-oxo-1,2-dihydropyridin-3-yl) methyl) benzamide derivatives were designed, synthesized, and demonstrated to act as EZH2 covalent inhibitors, among which SKLB-03176 was the most potent compound. SAM competition experiments, mass spectrometry, and washing-out assays proved that SKLB-03176 could covalently bind to the SAM pocket of EZH2. Remarkably, SKLB-03176 exhibited weak activity against other targets, such as 5 histone methyltransferases and more than 30 kinases. Besides, it could inhibit the activity of a variety of EZH2 mutants and significantly inhibit the expression of H3K27Me3 in cells. Furthermore, SKLB-03176 showed no cytotoxicity to normal cells. Our data suggested that SKLB-03176 could be used as a promising lead compound for the development of new EZH2 covalent inhibitors and a valuable chemical tool to study the biological functions of EZH2 or PRC2.
Collapse
Affiliation(s)
- Qiangsheng Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, PR China
| | - Xinyi Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, PR China
| | - Xi Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, PR China
| | - Xianjie Duan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, PR China
| | - Guoquan Wan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, PR China
| | - Lu Li
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Qiang Feng
- College of Chemistry and Life Science, Chengdu Normal University, Chengdu 611130, PR China
| | - Yiqian Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, PR China
| | - Ningyu Wang
- School of Life Science and Engineering, Southwest JiaoTong University, Chengdu, Sichuan 611756, PR China.
| | - Luoting Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, 17#3rd Section, Ren Min South Road, Chengdu 610041, PR China.
| |
Collapse
|
43
|
Cao Y, Yang Y, Zhai H, Wang J, Zhang S, Wang H, Yang P, Wu C. Synthesis and Antitumor Activity of Novel 5- and 6-Substituted Indazole Derivatives. CHINESE J ORG CHEM 2022. [DOI: 10.6023/cjoc202107049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
44
|
Yang H, Wang Z, An H, Yang Q, Xue W, Li F, Wang Y. Catalyst-free N-methylation of 3-methylxanthine with dimethyl carbonate in water: green synthesis of theobromine. NEW J CHEM 2022. [DOI: 10.1039/d2nj01624a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
3-Methylxanthine conversion of 89.9% with 98.2% theobromine selectivity.
Collapse
Affiliation(s)
- Hansen Yang
- Hebei Provincial Key Laboratory of Green Chemical Technology and High Efficient Energy Saving, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Zhimiao Wang
- Hebei Provincial Key Laboratory of Green Chemical Technology and High Efficient Energy Saving, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
- Tianjin Key Laboratory of Chemical Process Safety, Tianjin 300130, China
| | - Hualiang An
- Hebei Provincial Key Laboratory of Green Chemical Technology and High Efficient Energy Saving, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
- Tianjin Key Laboratory of Chemical Process Safety, Tianjin 300130, China
| | - Qiusheng Yang
- Hebei Provincial Key Laboratory of Green Chemical Technology and High Efficient Energy Saving, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
- Tianjin Key Laboratory of Chemical Process Safety, Tianjin 300130, China
| | - Wei Xue
- Hebei Provincial Key Laboratory of Green Chemical Technology and High Efficient Energy Saving, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
- Tianjin Key Laboratory of Chemical Process Safety, Tianjin 300130, China
| | - Fang Li
- Hebei Provincial Key Laboratory of Green Chemical Technology and High Efficient Energy Saving, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
- Tianjin Key Laboratory of Chemical Process Safety, Tianjin 300130, China
| | - Yanji Wang
- Hebei Provincial Key Laboratory of Green Chemical Technology and High Efficient Energy Saving, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
- Tianjin Key Laboratory of Chemical Process Safety, Tianjin 300130, China
- Hebei Industrial Technology Research Institute of Green Chemical Industry, Huanghua 061100, Hebei, China
| |
Collapse
|
45
|
Gutiérrez JR, Salgadoa ARM, Arias MDÁ, Vergara HSJ, Rada WR, Gómez CMM. Epigenetic Modulators as Treatment Alternative to Diverse Types of Cancer. Curr Med Chem 2021; 29:1503-1542. [PMID: 34963430 DOI: 10.2174/0929867329666211228111036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/17/2021] [Accepted: 10/21/2021] [Indexed: 01/10/2023]
Abstract
DNA is packaged in rolls in an octamer of histones forming a complex of DNA and proteins called chromatin. Chromatin as a structural matrix of a chromosome and its modifications are nowadays considered relevant aspects for regulating gene expression, which has become of high interest in understanding genetic mechanisms regulating various diseases, including cancer. In various types of cancer, the main modifications are found to be DNA methylation in the CpG dinucleotide as a silencing mechanism in transcription, post-translational histone modifications such as acetylation, methylation and others that affect the chromatin structure, the ATP-dependent chromatin remodeling and miRNA-mediated gene silencing. In this review we analyze the main alterations in gene expression, the epigenetic modification patterns that cancer cells present, as well as the main modulators and inhibitors of each epigenetic mechanism and the molecular evolution of the most representative inhibitors, which have opened a promising future in the study of HAT, HDAC, non-glycoside DNMT inhibitors and domain inhibitors.
Collapse
Affiliation(s)
- Jorseth Rodelo Gutiérrez
- Organic and Biomedical Chemistry Research Group, Faculty of Basic Sciences, Universidad del Atlántico, Barranquilla, Colombia
| | - Arturo René Mendoza Salgadoa
- Organic and Biomedical Chemistry Research Group, Faculty of Basic Sciences, Universidad del Atlántico, Barranquilla, Colombia
| | - Marcio De Ávila Arias
- Department of Medicine, Biotechnology Research Group, Health Sciences Division, Universidad del Norte, Barranquilla, Colombia
| | - Homero San- Juan- Vergara
- Department of Medicine, Biotechnology Research Group, Health Sciences Division, Universidad del Norte, Barranquilla, Colombia
| | - Wendy Rosales Rada
- Advanced Biomedicine Research Group. Faculty of Exact and Natural Sciences, Universidad Libre Seccional, Barranquilla, Colombia
- Advanced Biomedicine Research Group. Faculty of Exact and Natural Sciences, Universidad Libre Seccional, Barranquilla, Colombia
| | - Carlos Mario Meléndez Gómez
- Organic and Biomedical Chemistry Research Group, Faculty of Basic Sciences, Universidad del Atlántico, Barranquilla, Colombia
| |
Collapse
|
46
|
Guillade L, Mora P, Villar P, Alvarez R, R de Lera A. Total synthesis of nahuoic acid A via a putative biogenetic intramolecular Diels-Alder (IMDA) reaction. Chem Sci 2021; 12:15157-15169. [PMID: 34909158 PMCID: PMC8612404 DOI: 10.1039/d1sc04524e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/26/2021] [Indexed: 11/21/2022] Open
Abstract
Inspired by the biogenetic proposal of an intramolecular Diels-Alder (IMDA) cycloaddition, the total synthesis of natural product nahuoic acid A, a cofactor-competitive inhibitor of the epigenetic enzyme lysine methyl transferase SETD8, has been carried out. A non-conjugated pentaenal precursor was synthesized with high levels of stereoselectivity at seven stereogenic centers and with the appropriate control of double bond geometries. Although the IMDA reaction of the non-conjugated pentaenal using Me2AlCl for catalysis at -40 °C selectively afforded the trans-fused diastereomer corresponding to the Re-endo mode of cycloaddition, under thermal reaction conditions it gave rise to a mixture of diastereomers, that preferentially formed through the exo mode, including the cis-fused angularly-methylated octahydronaphthalene diastereomer precursor of nahuoic acid A. The natural product could be obtained upon oxidation and overall deprotection of the hydroxyl groups present in the Si-exo IMDA diastereomer.
Collapse
Affiliation(s)
- Lucía Guillade
- Departamento de Química Orgánica, Facultade de Química, CINBIO, IIS Galicia Sur, Universidade de Vigo 36310 Vigo Spain
| | - Paula Mora
- Departamento de Química Orgánica, Facultade de Química, CINBIO, IIS Galicia Sur, Universidade de Vigo 36310 Vigo Spain
| | - Pedro Villar
- Departamento de Química Orgánica, Facultade de Química, CINBIO, IIS Galicia Sur, Universidade de Vigo 36310 Vigo Spain
| | - Rosana Alvarez
- Departamento de Química Orgánica, Facultade de Química, CINBIO, IIS Galicia Sur, Universidade de Vigo 36310 Vigo Spain
| | - Angel R de Lera
- Departamento de Química Orgánica, Facultade de Química, CINBIO, IIS Galicia Sur, Universidade de Vigo 36310 Vigo Spain
| |
Collapse
|
47
|
Holdgate GA, Bardelle C, Lanne A, Read J, O'Donovan DH, Smith JM, Selmi N, Sheppard R. Drug discovery for epigenetics targets. Drug Discov Today 2021; 27:1088-1098. [PMID: 34728375 DOI: 10.1016/j.drudis.2021.10.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 09/19/2021] [Accepted: 10/27/2021] [Indexed: 12/28/2022]
Abstract
Dysregulation of the epigenome is associated with the onset and progression of several diseases, including cancer, autoimmune, cardiovascular, and neurological disorders. Members from the three families of epigenetic proteins (readers, writers, and erasers) have been shown to be druggable using small-molecule inhibitors. Increasing knowledge of the role of epigenetics in disease and the reversibility of these modifications explain why pharmacological intervention is an attractive strategy for tackling epigenetic-based disease. In this review, we provide an overview of epigenetics drug targets, focus on approaches used for initial hit identification, and describe the subsequent role of structure-guided chemistry optimisation of initial hits to clinical candidates. We also highlight current challenges and future potential for epigenetics-based therapies.
Collapse
Affiliation(s)
- Geoffrey A Holdgate
- High-throughput Screening, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Alderley Park, UK.
| | - Catherine Bardelle
- High-throughput Screening, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Alderley Park, UK
| | - Alice Lanne
- High-throughput Screening, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Alderley Park, UK
| | - Jon Read
- Structure and Biophysics, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | | | - Nidhal Selmi
- iLAB, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Robert Sheppard
- Medicinal Chemistry, Cardiovascular, Renal, Metabolism R&D, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
48
|
Zhou B, Liang X, Mei H, Qi S, Jiang Z, Wang A, Zou F, Liu Q, Liu J, Wang W, Hu C, Chen Y, Wang Z, Wang B, Wang L, Liu J, Liu Q. Discovery of IHMT-EZH2-115 as a Potent and Selective Enhancer of Zeste Homolog 2 (EZH2) Inhibitor for the Treatment of B-Cell Lymphomas. J Med Chem 2021; 64:15170-15188. [PMID: 34664960 DOI: 10.1021/acs.jmedchem.1c01154] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The enhancer of zeste homologue 2 (EZH2) is the catalytic subunit of polycomb repressive complex 2 that catalyzes methylation of histone H3 lysine 27 (H3K27). Overexpression or mutation of EZH2 has been identified in hematologic malignancies and solid tumors. Based on the structure of EPZ6438 (1) and the binding model with PRC2, we designed a series of analogues aiming to improve the activities of EZH2 mutants. Structure-activity relationship (SAR) exploration at both enzymatic and cellular levels led to the discovery of inhibitor 29. In the biochemical assay, 29 inhibited EZH2 (IC50 = 26.1 nM) with high selectivity over other histone methyltransferases. It was also potent against EZH2 mutants (EZH2 Y641F, IC50 = 72.3 nM). Furthermore, it showed no apparent inhibitory activity against the human ether-á-go-go related gene (hERG) (IC50 > 30 μM). In vivo, 29 exhibited favorable pharmacokinetic properties for oral administration and showed better efficacy than 1 in both Pfeiffer and Karpas-422 cell-mediated xenograft mouse models, indicating that it might be a new potential therapeutic candidate for EZH2 mutant cancers.
Collapse
Affiliation(s)
- Bin Zhou
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Xiaofei Liang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
| | - Husheng Mei
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Shuang Qi
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
| | - Zongru Jiang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
| | - Aoli Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
| | - Fengming Zou
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
| | - Qingwang Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
| | - Juan Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
| | - Wenliang Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
| | - Chen Hu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
| | - Yongfei Chen
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
| | - Zuowei Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Beilei Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
| | - Li Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
| | - Jing Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China
| | - Qingsong Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,University of Science and Technology of China, Hefei, Anhui 230026, P. R. China.,Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui 230031, P. R. China.,Precision Medicine Research Laboratory of Anhui Province, Hefei, Anhui 230088, P. R. China
| |
Collapse
|
49
|
Vaidergorn MM, da Silva Emery F, Ganesan A. From Hit Seeking to Magic Bullets: The Successful Union of Epigenetic and Fragment Based Drug Discovery (EPIDD + FBDD). J Med Chem 2021; 64:13980-14010. [PMID: 34591474 DOI: 10.1021/acs.jmedchem.1c00787] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
We review progress in the application of fragment-based drug discovery (FBDD) to epigenetic drug discovery (EPIDD) targeted at epigenetic writer and eraser enzymes as well as reader domains over the last 15 years. The greatest successes to date are in prospecting for bromodomain binding ligands. From a diverse array of fragment hits, multiple potent and selective compounds ensued, including the oncology clinical candidates mivebresib, ABBV-744, pelabresib, and PLX51107.
Collapse
Affiliation(s)
- Miguel M Vaidergorn
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 14040-903, Brazil
| | - Flavio da Silva Emery
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 14040-903, Brazil
| | - A Ganesan
- School of Pharmacy, University of East Anglia, Norwich NR4 7TJ, United Kingdom
| |
Collapse
|
50
|
Xiao W, Zhou Q, Wen X, Wang R, Liu R, Wang T, Shi J, Hu Y, Hou J. Small-Molecule Inhibitors Overcome Epigenetic Reprogramming for Cancer Therapy. Front Pharmacol 2021; 12:702360. [PMID: 34603017 PMCID: PMC8484527 DOI: 10.3389/fphar.2021.702360] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/02/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer treatment is a significant challenge for the global health system, although various pharmacological and therapeutic discoveries have been made. It has been widely established that cancer is associated with epigenetic modification, which is reversible and becomes an attractive target for drug development. Adding chemical groups to the DNA backbone and modifying histone proteins impart distinct characteristics on chromatin architecture. This process is mediated by various enzymes modifying chromatin structures to achieve the diversity of epigenetic space and the intricacy in gene expression files. After decades of effort, epigenetic modification has represented the hallmarks of different cancer types, and the enzymes involved in this process have provided novel targets for antitumor therapy development. Epigenetic drugs show significant effects on both preclinical and clinical studies in which the target development and research offer a promising direction for cancer therapy. Here, we summarize the different types of epigenetic enzymes which target corresponding protein domains, emphasize DNA methylation, histone modifications, and microRNA-mediated cooperation with epigenetic modification, and highlight recent achievements in developing targets for epigenetic inhibitor therapy. This article reviews current anticancer small-molecule inhibitors targeting epigenetic modified enzymes and displays their performances in different stages of clinical trials. Future studies are further needed to address their off-target effects and cytotoxicity to improve their clinical translation.
Collapse
Affiliation(s)
- Wenjing Xiao
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China.,Department of Pharmacy, The General Hospital of Western Theater Command of PLA, Chengdu, China
| | - Qiaodan Zhou
- Department of Ultrasonic, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xudong Wen
- Department of Gastroenterology and Hepatology, Chengdu First People's Hospital, Chengdu, China
| | - Rui Wang
- Information Department of Medical Security Center, The General Hospital of Western Theater Command of PLA, Chengdu, China
| | - Ruijie Liu
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Tingting Wang
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yonghe Hu
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China.,Department of Pharmacy, The General Hospital of Western Theater Command of PLA, Chengdu, China
| | - Jun Hou
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China.,Department of Pharmacy, The General Hospital of Western Theater Command of PLA, Chengdu, China
| |
Collapse
|