Ren-Jie Lin, Lin Xie, Tian-Yu Gao, Yi-Zhou Yang, Lan Huang, Kui Cheng, Zhi-Peng Chen
{"title":"Design, synthesis and anti-tumor evaluation of novel pyrimidine and quinazoline analogues","authors":"Ren-Jie Lin, Lin Xie, Tian-Yu Gao, Yi-Zhou Yang, Lan Huang, Kui Cheng, Zhi-Peng Chen","doi":"10.1016/j.ejmech.2024.117057","DOIUrl":"https://doi.org/10.1016/j.ejmech.2024.117057","url":null,"abstract":"Disrupting microtubule dynamics has emerged as a promising strategy for cancer therapy. Novel trimethoxyanilino-substituted pyrimidine and quinazoline derivatives were designed and synthesized to serve as potent microtubule-inhibiting agents with anti-proliferative activity. Compound <strong>2k</strong> demonstrates high efficacy against B16–F10 cancer cells at low nanomolar concentrations, with an IC<sub>50</sub> of 0.098 ± 0.006 μM, which is comparable to colchicine. Mechanistic studies have revealed that <strong>2k</strong> has the ability to inhibit microtubule protein polymerization <em>in vitro</em>, resulting in cell cycle arrest and apoptosis. Furthermore, <strong>2k</strong> inhibits tumor cell migration and exhibits significant anti-tumor efficacy in a melanoma tumor model without causing obvious toxicity. In summary, the pyrimidine derivative <strong>2k</strong> exhibits excellent anticancer activity and provides a new scaffold for the development of novel microtubule inhibitors, which deserves further in-depth research.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"169 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2024-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142665249","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Chengchun Zhu, Leilei Li, Yan Yu, Xiao Wang, Ying Shi, Yiping Gao, Kai Chen, Xiaoyu Liu, Yuqian Cui, Tao Zhang, Zhiyi Yu
{"title":"Optimization of SHP2 Allosteric Inhibitors with Novel Tail Heterocycles and Their Potential as Antitumor Therapeutics","authors":"Chengchun Zhu, Leilei Li, Yan Yu, Xiao Wang, Ying Shi, Yiping Gao, Kai Chen, Xiaoyu Liu, Yuqian Cui, Tao Zhang, Zhiyi Yu","doi":"10.1016/j.ejmech.2024.117078","DOIUrl":"https://doi.org/10.1016/j.ejmech.2024.117078","url":null,"abstract":"SHP2, a non-receptor protein tyrosine phosphatase involved in cancers, plays a pivotal role in numerous cellular signaling cascades, including the MAPK and PD-L1/PD-1 pathways. Although several SHP2 allosteric inhibitors have already entered clinical trials, none have been approved to date. Therefore, the development of new SHP2 allosteric inhibitors with improved efficacy is urgently required. Herein, we report the optimization of tail heterocycles in SHP2 allosteric inhibitors using a structure-based drug design strategy. Four series of compounds with different tail skeletons were synthesized, among which <strong>D13</strong> showed notable inhibitory activity (IC<sub>50</sub> = 1.2 μM) against SHP2. Molecular docking and binding studies indicated that the newly synthesized compounds exerted enzymatic inhibitory effects by directly binding to SHP2 with relatively slow dissociation rates. At the cellular level, Huh7 cells demonstrated heightened sensitivity to the novel SHP2 inhibitors, and <strong>D13</strong> exhibited superior antiproliferative activity (IC<sub>50</sub> = 38 μM) by arresting G0/G1 cell cycle, facilitating cell apoptosis and suppressing the MAPK signaling pathway. In the <em>in vivo</em> study, <strong>D13</strong> displayed significant antitumor activity in a Huh7 xenograft model and possessed favorable druggability with acceptable oral bioavailability (<em>F</em> = 54%) and half-life (<em>t</em><sub>1/2</sub> = 10.57 h). Collectively, this study lays a robust foundation for further optimization of the tail heterocycle skeleton in SHP2 allosteric inhibitors.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"18 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2024-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142665557","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Si Ha, Chenxuan Ji, Jiaqi Yang, Maoxu Xiao, Ziyi Xu, Wei-Wei Pan, Hua Xiang, Guoshun Luo
{"title":"Discovery of a Highly Potent, N-terminal Domain-targeting degrader of AR-FL/AR-V7 for the treatment of Prostate Cancer","authors":"Si Ha, Chenxuan Ji, Jiaqi Yang, Maoxu Xiao, Ziyi Xu, Wei-Wei Pan, Hua Xiang, Guoshun Luo","doi":"10.1016/j.ejmech.2024.117079","DOIUrl":"https://doi.org/10.1016/j.ejmech.2024.117079","url":null,"abstract":"The clinical development of PROTACs targeting the androgen receptor (AR) for degradation has made significant progress. However, effective treatments for metastatic prostate cancers containing the androgen receptor splice variant 7 (AR-V7), a constitutively active mutant without the ligand-binding domain (LBD), are still lacking. Here, we reported the identification of a highly potent, noncovalent PROTAC targeting the N-terminal domain (NTD) of AR, <strong>NP18</strong>, which is developed from the covalent AR-NTD antagonist EPI-002, and effectively degrades both AR-FL and AR-V7 in 22Rv1 cells (DC<sub>50</sub>: 18 and 26 nM respectively). Mechanistically, <strong>NP18</strong> interacts with the N-terminal domain (NTD) of both full-length AR (AR-FL) and splice variant 7 (AR-V7), leading to their selective and proteasomal degradation. Importantly, <strong>NP18</strong> exhibited remarkably superior antitumor activity in both 22Rv1 xenograft and patient-derived xenograft (PDX) models than EPI-002. Taken together, these findings highlight <strong>NP18</strong> as a promising candidate to counteract AR splice variant-driven resistance.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"248 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2024-11-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142665558","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"DNA or Not DNA —That is the Question Determining the Design of Platinum Anticancer Drugs","authors":"Suxing Jin, Chenyao Feng, Xiaoyong Wang","doi":"10.1016/j.ejmech.2024.117077","DOIUrl":"https://doi.org/10.1016/j.ejmech.2024.117077","url":null,"abstract":"Platinum drugs are the most widely used chemotherapeutics to treat various tumors. Their primary mode of action is supposed to be inducing apoptosis of cancer cells via covalent binding to DNA. This mechanism has shackled the design of new platinum drugs for many years. Mounting evidence shows that many platinum complexes form non-covalent adducts with DNA or interact with proteins to exhibit significant antitumor activity, thus implying some distinct mechanisms from that of traditional platinum drugs. These unconventional examples indicate that covalent DNA binding is not the precondition for the antitumor activity of platinum complexes, and diversified reactions or interactions with biomolecules, organelles, signal pathways, or immune system could lead to the antitumor activity of platinum complexes. The atypical mechanisms break the classical DNA-only paradigm and structure−activity relationships, thus opening a wide avenue for the design of innovative platinum anticancer drugs.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"76 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2024-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142665580","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Mingfei Wu, Yiming Jiang, Daoming Zhang, Yiquan Wu, Yuyuan Jin, Tao Liu, Xinfei Mao, Hengyuan Yu, Tengfei Xu, Yong Chen, Wenhai Huang, Jinxin Che, Bo Zhang, Tao Liu, Nengming Lin, Xiaowu Dong
{"title":"Discovery of a potent PARP1 PROTAC as a chemosensitizer for the treatment of colorectal cancer","authors":"Mingfei Wu, Yiming Jiang, Daoming Zhang, Yiquan Wu, Yuyuan Jin, Tao Liu, Xinfei Mao, Hengyuan Yu, Tengfei Xu, Yong Chen, Wenhai Huang, Jinxin Che, Bo Zhang, Tao Liu, Nengming Lin, Xiaowu Dong","doi":"10.1016/j.ejmech.2024.117062","DOIUrl":"https://doi.org/10.1016/j.ejmech.2024.117062","url":null,"abstract":"Given the vulnerability of colorectal cancer (CRC) patients could not obtain a sustained benefit from chemotherapy, combination therapy is frequently employed as a treatment strategy. Targeting PARP1 blockade exhibit specific toxicity towards tumor cells with BRCA1 or BRCA2 mutations through synthetic lethality. This study focuses on developing a series of potent PROTACs targeting PARP1 in order to enhance the sensitivity of CRC cells with BRCA1 or BRCA2 mutations to chemotherapy. Compound <strong>C6</strong>, obtained based on precise structural optimization of the linker, has been shown to effectively degrade PARP1 with a DC<sub>50</sub> value of 58.14 nM. Furthermore, <strong>C6</strong> significantly increased the cytotoxic efficacy of SN-38, an active metabolite of Irinotecan, in BRCA-mutated CRC cells, achieving a favorable combination index (CI) of 0.487. In conclusion, this research underscores the potential benefits of employing a combination therapy that utilizes PAPRP1 degrader <strong>C6</strong> alongside Irinotecan for CRC patients harboring BRCA mutations in CRC.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"34 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2024-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142645848","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Luolong Qing, Zhengzai Cheng, Juan Xu, Ziwei Wang, Yuanyuan Li, Mario Gauthier, Silong Zhang, Huan He
{"title":"Novel potent SOS1 inhibitors containing a tricyclic quinazoline scaffold: a joint view of experiments and simulations","authors":"Luolong Qing, Zhengzai Cheng, Juan Xu, Ziwei Wang, Yuanyuan Li, Mario Gauthier, Silong Zhang, Huan He","doi":"10.1016/j.ejmech.2024.117065","DOIUrl":"https://doi.org/10.1016/j.ejmech.2024.117065","url":null,"abstract":"Small molecules that possess the ability to regulate the interactions between Son of Sevenless 1 (SOS1) and Kristen rat sarcoma (KRAS) offer immense potential in the realm of cancer therapy. In this study, we present a novel series of SOS1 inhibitors featuring a tricyclic quinazoline scaffold. Notably, we have identified compound <strong>8d</strong>, which demonstrates the highest potency with an IC<sub>50</sub> value of 5.1 nM for disrupting the KRAS:SOS1 interaction. Compound <strong>8d</strong> exhibits a promising pharmacokinetic profile and achieves a remarkable 70.5% inhibition of tumor growth in pancreas tumor xenograft models. Furthermore, molecular dynamic simulations have unveiled that the tricyclic quinazoline derivatives exhibit extensive interaction with Tyr884, a crucial residue for the recognition between SOS1 and KRAS. Our findings provide fresh insights into the design of future SOS1 inhibitors, paving the way for innovative therapeutic strategies.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"171 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2024-11-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142665582","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Deng-Gao Zhao, JieYing Liu, Zhengxi Su, Wenbo Zou, Qianwei Zhou, Ting Yin, Tan Jiyao, Yan-Yan Ma
{"title":"Discovery of novel KSP-targeting PROTACs with potent antitumor effects in Vitro and in Vivo","authors":"Deng-Gao Zhao, JieYing Liu, Zhengxi Su, Wenbo Zou, Qianwei Zhou, Ting Yin, Tan Jiyao, Yan-Yan Ma","doi":"10.1016/j.ejmech.2024.117052","DOIUrl":"https://doi.org/10.1016/j.ejmech.2024.117052","url":null,"abstract":"Kinesin spindle protein (KSP) plays a crucial role during mitosis, making it an attractive target for cancer treatment. Herein, we report the design, synthesis, and evaluation of the first series of KSP degraders by using the utilization of the proteolysis-targeting chimera (PROTAC) technology. Compound <strong>21</strong> was identified as a potent KSP degrader with a DC<sub>50</sub> (concentration causing 50% of protein degradation) value of 114.8 nM and a D<sub>max</sub> (maximum degradation) of 90% in the HCT-116 cells. Compound <strong>21</strong> showed strong antiproliferative activity against HCT-116 cells with an IC<sub>50</sub> values of 10 nM. Mechanistic investigations revealed that <strong>21</strong> causes the cell arrest at the G2/M phase and subsequent cell apoptosis. In addition, <strong>21</strong> demonstrated more significant inhibition of tumor growth in an HCT-116 xenograft model compared to its parent compound <strong>1.</strong> Our findings suggest that <strong>21</strong> may become the promising leads for further development.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"12 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2024-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142642895","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Soheil Sojdeh, Moein Safarkhani, Hossein Daneshgar, Abdullah Aldhaher, Golnaz Heidari, Ehsan Nazarzadeh Zare, Siavash Iravani, Ali Zarrabi, Navid Rabiee
{"title":"Promising Breakthroughs in Amyotrophic Lateral Sclerosis Treatment through Nanotechnology's Unexplored Frontier","authors":"Soheil Sojdeh, Moein Safarkhani, Hossein Daneshgar, Abdullah Aldhaher, Golnaz Heidari, Ehsan Nazarzadeh Zare, Siavash Iravani, Ali Zarrabi, Navid Rabiee","doi":"10.1016/j.ejmech.2024.117080","DOIUrl":"https://doi.org/10.1016/j.ejmech.2024.117080","url":null,"abstract":"This article explores the transformative potential of nanotechnology in the treatment and diagnosis of amyotrophic lateral sclerosis (ALS), a progressive neurodegenerative disorder characterized by motor neuron degeneration, muscle weakness, and eventual paralysis. Nanotechnology offers innovative solutions across various domains, including targeted drug delivery, neuroprotection, gene therapy and editing, biomarker detection, advanced imaging techniques, and tissue engineering. By enhancing the precision and efficacy of therapeutic interventions, nanotechnology facilitates key advancements such as crossing the blood-brain barrier, targeting specific cell types, achieving sustained therapeutic release, and enabling combination therapies tailored to the complex pathophysiology of ALS. Despite its immense promise, the clinical translation of these approaches faces challenges, including potential cytotoxicity, biocompatibility, and regulatory compliance, which must be addressed through rigorous research and testing. This review emphasizes the application of nanotechnology in targeted drug delivery and gene therapy/editing for ALS, drawing on the author’s prior work with various nanotechnological platforms to illustrate strategies for overcoming similar obstacles in drug and gene delivery. By bridging the gap between cutting-edge technology and clinical application, this article aims to highlight the vital role of nanotechnology in shaping the future of ALS treatment.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"249 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2024-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142645849","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Synthesis, biological evaluation and mechanism study of a novel indole-pyridine chalcone derivative as antiproliferative agent against tumor cells through dual targeting tubulin and HK2","authors":"Mengzhu Zheng, Guangyuan Liu, Yawei Han, Pengyu Qian, Mingze Wu, Ming Xiang, Yirong Zhou","doi":"10.1016/j.ejmech.2024.117058","DOIUrl":"https://doi.org/10.1016/j.ejmech.2024.117058","url":null,"abstract":"Chalcones have the characteristics of simple structure, easy synthesis and potent anti-tumor activity. Herein, a small library of fifty-five novel indole-chalcone derivatives were rationally designed and facilely synthesized. Consequently, their antiproliferative activity was systematically evaluated. Among which, compound <strong>26</strong> exhibited the most potent antiproliferative activity, with IC<sub>50</sub> value of 0.764 μM against MD-MBA-231 cells. Moreover, it displayed a 5-fold selectivity compared with normal human cells. Further investigation revealed that compound <strong>26</strong> bound at the colchicine binding site of tubulin, disrupted their fibrous structure, thereby blocking the progression of the cell cycle and inducing apoptosis. Molecular docking and cellular thermal shift assay (CETSA) experiments further demonstrated that compound <strong>26</strong> could specifically bind to hexokinase 2 (HK2) and inhibit its activity, leading to impaired mitochondrial function and hindered mitochondrial respiration. Based on the quantitative structure-activity relationship study, further structure modifications were performed. Employing biotin probe pull-down assays, we demonstrated that compound <strong>26</strong> exerted its antiproliferative activity through a dual targeting mechanism, which simultaneously disrupted microtubule function and inhibited HK2 activity. Taken together, these results highlighted that compound <strong>26</strong> might be a promising antiproliferative agent for human cancer therapy.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"37 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2024-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142642978","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Nanomaterials exert biological effects by influencing the ubiquitin-proteasome system","authors":"Zhen Ai, Dan Li, Shuquan Lan, Chao Zhang","doi":"10.1016/j.ejmech.2024.116974","DOIUrl":"10.1016/j.ejmech.2024.116974","url":null,"abstract":"<div><div>The ubiquitin-proteasome system (UPS) is an important type of protein post-translational modification that affects the quantity and quality of various proteins and influences cellular processes such as the cell cycle, transcription, oxidative stress, and autophagy. Nanomaterials (NMs), which exhibit excellent physicochemical properties, can directly interact with the UPS and act as molecular-targeted drugs to induce changes in biological processes. This review provides an overview of the influence of NMs on the UPS of misfolded proteins and key proteins, which are related to cancer, neurodegenerative diseases and oxidative stress. This review also summarizes the role of modification processes involved in ubiquitination the biological effects of NMs and the mechanism of such effects of NMs through regulation of the UPS. This review deepens our understanding of the influence of NMs on the protein degradation process and provides new potential therapeutic targets for disease.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"282 ","pages":"Article 116974"},"PeriodicalIF":6.0,"publicationDate":"2024-11-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142642898","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}