首页 > 最新文献

European Journal of Medicinal Chemistry最新文献

英文 中文
Discovery of new fungal jumonji H3K27 demethylase inhibitors for the treatment of Cryptococcus neoformans and Candida auris infections 发现治疗新生隐球菌和白色念珠菌感染的新型真菌 Jumonji H3K27 去甲基化酶抑制剂
IF 6 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-06 DOI: 10.1016/j.ejmech.2024.117028
Xin Liu , Wang Li , Yang Liu , Xiaoqing Wang , Qiao Shi , Wanzhen Yang , Jie Tu , Yan Wang , Chunquan Sheng , Na Liu
Invasive fungal infections have become a serious public health problem. To tackle the challenges of limited efficacy in antifungal therapy and severe drug resistance, antifungal drugs with new mechanisms of action are urgently needed. Our previous study identified JIB-04 to be an inhibitor of fungal histone demethylase (HDM). To promote target validation and inhibitor design, herein a series of new JIB-04 derivatives were designed and synthesized. After the establishment of structure-activity relationship, compound A4 was identified to possess potent antifungal activity against Cryptococcus neoformans and Candida auris. Compared to lead compound JIB-04, compound A4 was a more potent HDM inhibitor and exhibited better water solubility, virulence factors inhibitory activity and in vivo antifungal potency. Collectively, this study further confirmed that fungal HDMs were potential antifungal targets and compound A4 was a promising antifungal lead compound.
侵袭性真菌感染已成为一个严重的公共卫生问题。为了应对抗真菌治疗疗效有限和严重耐药性的挑战,迫切需要具有新作用机制的抗真菌药物。我们之前的研究发现,JIB-04 是真菌组蛋白去甲基化酶(HDM)的抑制剂。为了促进靶点验证和抑制剂设计,我们在此设计并合成了一系列新的 JIB-04 衍生物。在建立了结构-活性关系后,化合物 A4 被确认对新型隐球菌和白色念珠菌具有强效抗真菌活性。与先导化合物 JIB-04 相比,化合物 A4 是一种更有效的 HDM 抑制剂,具有更好的水溶性、毒力因子抑制活性和体内抗真菌效力。总之,这项研究进一步证实了真菌 HDMs 是潜在的抗真菌靶标,而化合物 A4 则是一种很有前途的抗真菌先导化合物。
{"title":"Discovery of new fungal jumonji H3K27 demethylase inhibitors for the treatment of Cryptococcus neoformans and Candida auris infections","authors":"Xin Liu ,&nbsp;Wang Li ,&nbsp;Yang Liu ,&nbsp;Xiaoqing Wang ,&nbsp;Qiao Shi ,&nbsp;Wanzhen Yang ,&nbsp;Jie Tu ,&nbsp;Yan Wang ,&nbsp;Chunquan Sheng ,&nbsp;Na Liu","doi":"10.1016/j.ejmech.2024.117028","DOIUrl":"10.1016/j.ejmech.2024.117028","url":null,"abstract":"<div><div>Invasive fungal infections have become a serious public health problem. To tackle the challenges of limited efficacy in antifungal therapy and severe drug resistance, antifungal drugs with new mechanisms of action are urgently needed. Our previous study identified <strong>JIB-04</strong> to be an inhibitor of fungal histone demethylase (HDM). To promote target validation and inhibitor design, herein a series of new <strong>JIB-04</strong> derivatives were designed and synthesized. After the establishment of structure-activity relationship, compound <strong>A4</strong> was identified to possess potent antifungal activity against <em>Cryptococcus neoformans</em> and <em>Candida auris</em>. Compared to lead compound <strong>JIB-04</strong>, compound <strong>A4</strong> was a more potent HDM inhibitor and exhibited better water solubility, virulence factors inhibitory activity and <em>in vivo</em> antifungal potency. Collectively, this study further confirmed that fungal HDMs were potential antifungal targets and compound <strong>A4</strong> was a promising antifungal lead compound.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"281 ","pages":"Article 117028"},"PeriodicalIF":6.0,"publicationDate":"2024-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142594537","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}
引用次数: 0
Discovery of pyrrolopyrimidinone derivatives as potent PKMYT1 inhibitors for the treatment of cancer 发现吡咯并嘧啶酮衍生物作为治疗癌症的强效 PKMYT1 抑制剂
IF 6 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-05 DOI: 10.1016/j.ejmech.2024.117025
Chao Wang , Yazhou Wang , Fanye Meng , Tingting Liu , Xiaomin Wang , Xin Cai , Man Zhang , Alex Aliper , Feng Ren , Alex Zhavoronkov , Xiao Ding
The protein kinase PKMYT1 is responsible for inhibitory CDK1 phosphorylation, thus playing a central role in regulating the G2/M cell cycle checkpoint. As many cancers have dysfunctional cell cycle checkpoint signaling, PKMYT1 inhibition is emerging as an attractive target in advanced tumors. PKMYT1 inhibitors, however, have encountered difficulties in balancing biological efficacy, on-target specificity, and favorable stability and other drug-like properties. Herein, we report the design and development of pyrrolopyrimidinone derivatives intended to simultaneously restrict molecular conformation and shield a metabolic site in order to optimize stability. Compound 7 demonstrated strong PKMYT1-specific inhibition, a subsequent decrease in CDK1 phosphorylation, and antitumor efficacy in vitro, as well as enhanced metabolic stability, favorable pharmacokinetic and bioavailability properties, and potent antitumor in vivo efficacy. Our findings indicate that compound 7 is a promising PKMYT1 inhibitor for the treatment of advanced cancers with cell cycle defects.
蛋白激酶 PKMYT1 负责抑制 CDK1 磷酸化,因此在调节 G2/M 细胞周期检查点方面发挥着核心作用。由于许多癌症都存在细胞周期检查点信号传导失调的问题,PKMYT1抑制剂正成为晚期肿瘤的一个诱人靶点。然而,PKMYT1 抑制剂在平衡生物有效性、靶向特异性、良好的稳定性和其他类药物特性方面遇到了困难。在此,我们报告了吡咯并嘧啶酮衍生物的设计和开发情况,这些衍生物旨在同时限制分子构象和保护代谢位点,以优化稳定性。化合物 7 在体外表现出很强的 PKMYT1 特异性抑制作用、CDK1 磷酸化随之降低和抗肿瘤功效,同时还具有更高的代谢稳定性、良好的药代动力学和生物利用度特性以及强大的体内抗肿瘤功效。我们的研究结果表明,化合物 7 是一种很有前景的 PKMYT1 抑制剂,可用于治疗存在细胞周期缺陷的晚期癌症。
{"title":"Discovery of pyrrolopyrimidinone derivatives as potent PKMYT1 inhibitors for the treatment of cancer","authors":"Chao Wang ,&nbsp;Yazhou Wang ,&nbsp;Fanye Meng ,&nbsp;Tingting Liu ,&nbsp;Xiaomin Wang ,&nbsp;Xin Cai ,&nbsp;Man Zhang ,&nbsp;Alex Aliper ,&nbsp;Feng Ren ,&nbsp;Alex Zhavoronkov ,&nbsp;Xiao Ding","doi":"10.1016/j.ejmech.2024.117025","DOIUrl":"10.1016/j.ejmech.2024.117025","url":null,"abstract":"<div><div>The protein kinase PKMYT1 is responsible for inhibitory CDK1 phosphorylation, thus playing a central role in regulating the G2/M cell cycle checkpoint. As many cancers have dysfunctional cell cycle checkpoint signaling, PKMYT1 inhibition is emerging as an attractive target in advanced tumors. PKMYT1 inhibitors, however, have encountered difficulties in balancing biological efficacy, on-target specificity, and favorable stability and other drug-like properties. Herein, we report the design and development of pyrrolopyrimidinone derivatives intended to simultaneously restrict molecular conformation and shield a metabolic site in order to optimize stability. Compound <strong>7</strong> demonstrated strong PKMYT1-specific inhibition, a subsequent decrease in CDK1 phosphorylation, and antitumor efficacy <em>in vitro</em>, as well as enhanced metabolic stability, favorable pharmacokinetic and bioavailability properties, and potent antitumor <em>in vivo</em> efficacy. Our findings indicate that compound <strong>7</strong> is a promising PKMYT1 inhibitor for the treatment of advanced cancers with cell cycle defects.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"281 ","pages":"Article 117025"},"PeriodicalIF":6.0,"publicationDate":"2024-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142580260","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}
引用次数: 0
Design, synthesis, and cytotoxicity screening of novel pyrazolopyrimidines over renal cell carcinoma (UO-31 cells) as p38α inhibitors, and apoptotic cells inducing activities 新型吡唑并嘧啶作为 p38α 抑制剂和细胞凋亡诱导剂对肾细胞癌(UO-31 细胞)的设计、合成和细胞毒性筛选
IF 6 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-05 DOI: 10.1016/j.ejmech.2024.117005
Sara Y. Ewieda , Amr Sonousi , Aliaa M. Kamal , Mohamed K. Abdelhamid
A series of novel molecules with pyrazolopyrimidine-4-amine core were designed and synthesized as potential cytotoxic agents over Renal Cell Carcinoma cells (UO-31). Results of cytotoxic activity against UO-31 cells showed that pyrazolopyrimidines 19 and 31 were found to be more cytotoxic than sorafenib (SOR). The cytotoxic activity of these compounds appeared to correlate with their ability to inhibit p38α MAPK which are 2.53- and 2.27- folds more potent than SOR. Moreover, results of the cell cycle analysis as well as the results of annexin-V on the (UO-31) cells showed that pyrazolopyrimidines 19 and 31 had a pro-apoptotic activity higher than SOR by 1.42- and 1.20- folds, respectively. Furthermore, compounds 19 and 31 were found to be effective in arresting the cell cycle throughout the accumulation of the cells at G2/M phase. Finally, the tested compounds decreased the TNF concentration as well as increased the expression of tumor suppressor gene p53, Bax/BCL-2 ratio and caspase 3/7.
研究人员设计并合成了一系列以吡唑嘧啶-4-胺为核心的新型分子,这些分子对肾细胞癌细胞(UO-31)具有潜在的细胞毒性。对 UO-31 细胞的细胞毒活性研究结果表明,吡唑嘧啶 19 和 31 的细胞毒活性高于索拉非尼(SOR)。这些化合物的细胞毒性活性似乎与它们抑制 p38α MAPK 的能力有关,其抑制能力分别比 SOR 强 2.53 倍和 2.27 倍。此外,细胞周期分析结果以及附件素-V 对(UO-31)细胞的检测结果表明,吡唑并嘧啶 19 和 31 的促凋亡活性分别比 SOR 高 1.42 倍和 1.20 倍。此外,还发现化合物 19 和 31 能有效阻止细胞周期在 G2/M 期的积累。最后,受试化合物降低了 TNF 浓度,并增加了肿瘤抑制基因 p53、Bax/BCL-2 比率和 caspase 3/7 的表达。
{"title":"Design, synthesis, and cytotoxicity screening of novel pyrazolopyrimidines over renal cell carcinoma (UO-31 cells) as p38α inhibitors, and apoptotic cells inducing activities","authors":"Sara Y. Ewieda ,&nbsp;Amr Sonousi ,&nbsp;Aliaa M. Kamal ,&nbsp;Mohamed K. Abdelhamid","doi":"10.1016/j.ejmech.2024.117005","DOIUrl":"10.1016/j.ejmech.2024.117005","url":null,"abstract":"<div><div>A series of novel molecules with pyrazolopyrimidine-4-amine core were designed and synthesized as potential cytotoxic agents over Renal Cell Carcinoma cells (UO-31). Results of cytotoxic activity against UO-31 cells showed that pyrazolopyrimidines <strong>19</strong> and <strong>31</strong> were found to be more cytotoxic than sorafenib (SOR). The cytotoxic activity of these compounds appeared to correlate with their ability to inhibit p38α MAPK which are 2.53- and 2.27- folds more potent than SOR. Moreover, results of the cell cycle analysis as well as the results of annexin-V on the (UO-31) cells showed that pyrazolopyrimidines <strong>19</strong> and <strong>31</strong> had a pro-apoptotic activity higher than SOR by 1.42- and 1.20- folds, respectively. Furthermore, compounds <strong>19</strong> and <strong>31</strong> were found to be effective in arresting the cell cycle throughout the accumulation of the cells at G2/M phase. Finally, the tested compounds decreased the TNF concentration as well as increased the expression of tumor suppressor gene p53, Bax/BCL-2 ratio and caspase 3/7.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"281 ","pages":"Article 117005"},"PeriodicalIF":6.0,"publicationDate":"2024-11-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142588325","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}
引用次数: 0
Discovery of cloxiquine derivatives as potent HDAC inhibitors for the treatment of melanoma via activating PPARγ 发现通过激活 PPARγ 治疗黑色素瘤的强效 HDAC 抑制剂氯喹衍生物
IF 6 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-04 DOI: 10.1016/j.ejmech.2024.117029
Limin Yang , Ran Ding , Xiaojie Tong , Tong Shen , Shuting Jia , Xiqing Yan , Chong Zhang , Liqiang Wu
The combined treatment with histone deacetylase (HDAC) inhibitors with peroxisome proliferator-activated receptor γ (PPARγ) agonists has displayed significant anticancer efficacy. Based on these results, a series of cloxiquine derivatives were prepared as potent HDAC inhibitors for the treatment of melanoma. Among these compounds, CS4 exhibited excellent inhibitory effects on HDAC1 (IC50 = 38 nM) and HDAC6 (IC50 = 12 nM), and had good antiproliferative effects against A375 and SK-MEL-5 melanoma cells (IC50 values, 1.20 and 0.93 μM, respectively). Mechanism research indicated that CS4 inhibited SK-MEL-5 cell growth by promoting α-tubulin and histone 3 (H3) acetylation. At the metabolic level, treatment with BG11 activated PPARγ and blocked glycolysis in SK-MEL-5 cells, which mediated partial antimelanoma effects of CS4. In addition, CS4 also induced cell cycle arrest at G2, suppressed migration and facilitated apoptosis of SK-MEL-5 cells. More importantly, compound CS4 demonstrated significant in vivo anticancer effect compared with SAHA, and exhibited neglectable toxicity. Consequently, CS4 is the potent HDAC inhibitor, which may be developed as the candidate antimelanoma drug.
组蛋白去乙酰化酶(HDAC)抑制剂与过氧化物酶体增殖激活受体γ(PPARγ)激动剂联合治疗显示出显著的抗癌疗效。基于这些结果,研究人员制备了一系列氯喹衍生物,作为治疗黑色素瘤的强效 HDAC 抑制剂。在这些化合物中,CS4 对 HDAC1(IC50 = 38 nM)和 HDAC6(IC50 = 12 nM)有很好的抑制作用,对 A375 和 SK-MEL-5 黑色素瘤细胞有很好的抗增殖作用(IC50 值分别为 1.20 和 0.93 μM)。机理研究表明,CS4通过促进α-微管蛋白和组蛋白3(H3)乙酰化来抑制SK-MEL-5细胞的生长。在代谢水平上,BG11 能激活 PPARγ 并阻断 SK-MEL-5 细胞的糖酵解,从而介导 CS4 的部分抗黑色素瘤作用。此外,CS4 还能诱导细胞周期停滞在 G2 阶段,抑制迁移并促进 SK-MEL-5 细胞凋亡。更重要的是,与 SAHA 相比,化合物 CS4 在体内具有显著的抗癌效果,而且毒性可忽略不计。因此,CS4 是一种有效的 HDAC 抑制剂,可作为抗黑色素瘤的候选药物进行开发。
{"title":"Discovery of cloxiquine derivatives as potent HDAC inhibitors for the treatment of melanoma via activating PPARγ","authors":"Limin Yang ,&nbsp;Ran Ding ,&nbsp;Xiaojie Tong ,&nbsp;Tong Shen ,&nbsp;Shuting Jia ,&nbsp;Xiqing Yan ,&nbsp;Chong Zhang ,&nbsp;Liqiang Wu","doi":"10.1016/j.ejmech.2024.117029","DOIUrl":"10.1016/j.ejmech.2024.117029","url":null,"abstract":"<div><div>The combined treatment with histone deacetylase (HDAC) inhibitors with peroxisome proliferator-activated receptor γ (PPARγ) agonists has displayed significant anticancer efficacy. Based on these results, a series of cloxiquine derivatives were prepared as potent HDAC inhibitors for the treatment of melanoma. Among these compounds, <strong>CS4</strong> exhibited excellent inhibitory effects on HDAC1 (IC<sub>50</sub> = 38 nM) and HDAC6 (IC<sub>50</sub> = 12 nM), and had good antiproliferative effects against A375 and SK-MEL-5 melanoma cells (IC<sub>50</sub> values, 1.20 and 0.93 μM, respectively). Mechanism research indicated that <strong>CS4</strong> inhibited SK-MEL-5 cell growth by promoting α-tubulin and histone 3 (H3) acetylation. At the metabolic level, treatment with <strong>BG11</strong> activated PPARγ and blocked glycolysis in SK-MEL-5 cells, which mediated partial antimelanoma effects of <strong>CS4</strong>. In addition, <strong>CS4</strong> also induced cell cycle arrest at G2, suppressed migration and facilitated apoptosis of SK-MEL-5 cells. More importantly, compound <strong>CS4</strong> demonstrated significant <em>in vivo</em> anticancer effect compared with SAHA, and exhibited neglectable toxicity. Consequently, <strong>CS4</strong> is the potent HDAC inhibitor, which may be developed as the candidate antimelanoma drug.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"281 ","pages":"Article 117029"},"PeriodicalIF":6.0,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142580104","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}
引用次数: 0
Development of hybrid aptamers-engineered PROTACs for degrading VEGF165 in both tumor- and vascular endothelial cells 开发可在肿瘤细胞和血管内皮细胞中降解 VEGF165 的混合适配体工程化 PROTACs
IF 6 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-02 DOI: 10.1016/j.ejmech.2024.117027
Ziting Feng , Duoli Xie , Fang Qiu , Jie Huang , Zhuqian Wang , Chao Liang
Tumors and angiogenesis are connected through a complex interplay. VEGF165, generated from both tumor and vascular endothelial cells, serves as a mutual benefit for both cell types. Therapeutic approaches modulating VEGF165 have been proposed as promising antitumor therapies. PROTACs are bifunctional molecules that exploit the intracellular ubiquitin-proteasome system to degrade specific proteins. To date, there are no targeted PROTACs designed to degrade VEGF165 in both tumor and vascular endothelial cells. The aptamer AS1411 is notable for its ability to selectively recognize and enter both tumor and vascular endothelial cells by targeting the cell surface nucleolin (NCL). Moreover, AS1411 has also been repurposed as an intracellular recruiter of E3 ligase MDM2 via leveraging NCL as a molecular bridge. In this study, we conjugated AS1411 with a VEGF165-specific aptamer V7t1, creating hybrid aptamers-engineered PROTACs. The PROTACs demonstrate remarkable selectivity for both tumor and vascular endothelial cells and facilitate the ubiquitination and proteasomal degradation of VEGF165. The PROTACs inhibit the growth of tumor cells and also impede angiogenesis, without causing toxicity to normal tissues. The hybrid aptamers-engineered PROTACs provide an avenue for disrupting the tumor-angiogenesis interplay through modulation of VEGF165 in both tumor and vascular endothelial cells.
肿瘤和血管生成通过复杂的相互作用联系在一起。由肿瘤细胞和血管内皮细胞产生的 VEGF165 对两种细胞类型都有好处。调控 VEGF165 的治疗方法被认为是有前景的抗肿瘤疗法。PROTACs 是一种双功能分子,可利用细胞内泛素-蛋白酶体系统降解特定蛋白质。迄今为止,还没有设计出同时降解肿瘤细胞和血管内皮细胞中 VEGF165 的靶向 PROTACs。适配体 AS1411 的显著特点是能通过靶向细胞表面的核素蛋白(NCL),选择性地识别并进入肿瘤细胞和血管内皮细胞。此外,AS1411 还通过利用 NCL 作为分子桥,被重新用作 E3 连接酶 MDM2 的细胞内招募剂。在本研究中,我们将 AS1411 与 VEGF165 特异性拟合物 V7t1 共轭,创造出混合拟合物工程化的 PROTACs。PROTACs 对肿瘤细胞和血管内皮细胞都具有显著的选择性,能促进 VEGF165 的泛素化和蛋白酶体降解。PROTACs 可抑制肿瘤细胞的生长,阻碍血管生成,但不会对正常组织造成毒性。这种由混合适配体设计的 PROTACs 为通过调节肿瘤细胞和血管内皮细胞中的 VEGF165 来破坏肿瘤与血管生成之间的相互作用提供了一条途径。
{"title":"Development of hybrid aptamers-engineered PROTACs for degrading VEGF165 in both tumor- and vascular endothelial cells","authors":"Ziting Feng ,&nbsp;Duoli Xie ,&nbsp;Fang Qiu ,&nbsp;Jie Huang ,&nbsp;Zhuqian Wang ,&nbsp;Chao Liang","doi":"10.1016/j.ejmech.2024.117027","DOIUrl":"10.1016/j.ejmech.2024.117027","url":null,"abstract":"<div><div>Tumors and angiogenesis are connected through a complex interplay. VEGF165, generated from both tumor and vascular endothelial cells, serves as a mutual benefit for both cell types. Therapeutic approaches modulating VEGF165 have been proposed as promising antitumor therapies. PROTACs are bifunctional molecules that exploit the intracellular ubiquitin-proteasome system to degrade specific proteins. To date, there are no targeted PROTACs designed to degrade VEGF165 in both tumor and vascular endothelial cells. The aptamer AS1411 is notable for its ability to selectively recognize and enter both tumor and vascular endothelial cells by targeting the cell surface nucleolin (NCL). Moreover, AS1411 has also been repurposed as an intracellular recruiter of E3 ligase MDM2 via leveraging NCL as a molecular bridge. In this study, we conjugated AS1411 with a VEGF165-specific aptamer V7t1, creating hybrid aptamers-engineered PROTACs. The PROTACs demonstrate remarkable selectivity for both tumor and vascular endothelial cells and facilitate the ubiquitination and proteasomal degradation of VEGF165. The PROTACs inhibit the growth of tumor cells and also impede angiogenesis, without causing toxicity to normal tissues. The hybrid aptamers-engineered PROTACs provide an avenue for disrupting the tumor-angiogenesis interplay through modulation of VEGF165 in both tumor and vascular endothelial cells.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"281 ","pages":"Article 117027"},"PeriodicalIF":6.0,"publicationDate":"2024-11-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142566096","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}
引用次数: 0
Recent report on indoles as a privileged anti-viral scaffold in drug discovery 关于吲哚作为药物研发中的抗病毒支架的最新报告
IF 6 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-01 DOI: 10.1016/j.ejmech.2024.117017
Asmita Singh , Charu Bhutani , Pankaj Khanna , Sangeeta Talwar , Sandeep Kumar Singh , Leena Khanna
In recent years, viral infections such as COVID-19, Zika virus, Nipah virus, Ebola, Influenza, Monkeypox, and Dengue have substantially impacted global health. These outbreaks have led to heightened global health initiatives and collaborative efforts to address and mitigate these significant threats effectively. Thus, developing antiviral treatments and research in this field has become highly important. Heterocycles, particularly indole motifs, have been a valuable resource in drug discovery, as they can be used as treatments or inspire the synthesis of new potent candidates. Indole-containing drugs, such as enfuvirtide (T-20), arbidol, and delavirdine, have demonstrated significant efficacy in treating viral diseases. This review aims to comprehensively assess the latest research and developments in novel indoles as potential scaffolds for antiviral activity. We have compiled detailed information about indoles as potential antivirals by conducting a thorough literature survey from the past ten years. The review includes discussions on synthetic protocols, inhibitory concentrations, SAR study, and computational study. This review shall identify new antiviral indoles that may help to combat new viral threats in the future.
近年来,COVID-19、寨卡病毒、尼帕病毒、埃博拉病毒、流感、猴痘和登革热等病毒感染严重影响了全球健康。这些疫情的爆发促使人们加强全球卫生倡议和合作努力,以有效地应对和减轻这些重大威胁。因此,开发抗病毒治疗方法和在这一领域开展研究已变得非常重要。杂环化合物,尤其是吲哚基团,一直是药物发现领域的宝贵资源,因为它们可以用作治疗药物或激发新的强效候选药物的合成。含吲哚的药物,如恩夫韦肽(T-20)、阿比多和德尔维利定,在治疗病毒性疾病方面具有显著疗效。本综述旨在全面评估新型吲哚作为抗病毒活性潜在支架的最新研究和发展。我们通过对过去十年的文献进行全面调查,汇编了有关吲哚作为潜在抗病毒药物的详细信息。综述包括对合成方案、抑制浓度、SAR 研究和计算研究的讨论。这篇综述将发现新的抗病毒吲哚,它们可能有助于对抗未来新的病毒威胁。
{"title":"Recent report on indoles as a privileged anti-viral scaffold in drug discovery","authors":"Asmita Singh ,&nbsp;Charu Bhutani ,&nbsp;Pankaj Khanna ,&nbsp;Sangeeta Talwar ,&nbsp;Sandeep Kumar Singh ,&nbsp;Leena Khanna","doi":"10.1016/j.ejmech.2024.117017","DOIUrl":"10.1016/j.ejmech.2024.117017","url":null,"abstract":"<div><div>In recent years, viral infections such as COVID-19, Zika virus, Nipah virus, Ebola, Influenza, Monkeypox, and Dengue have substantially impacted global health. These outbreaks have led to heightened global health initiatives and collaborative efforts to address and mitigate these significant threats effectively. Thus, developing antiviral treatments and research in this field has become highly important. Heterocycles, particularly indole motifs, have been a valuable resource in drug discovery, as they can be used as treatments or inspire the synthesis of new potent candidates. Indole-containing drugs, such as enfuvirtide (T-20), arbidol, and delavirdine, have demonstrated significant efficacy in treating viral diseases. This review aims to comprehensively assess the latest research and developments in novel indoles as potential scaffolds for antiviral activity. We have compiled detailed information about indoles as potential antivirals by conducting a thorough literature survey from the past ten years. The review includes discussions on synthetic protocols, inhibitory concentrations, SAR study, and computational study. This review shall identify new antiviral indoles that may help to combat new viral threats in the future.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"281 ","pages":"Article 117017"},"PeriodicalIF":6.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142562207","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}
引用次数: 0
Tubulin/HDAC dual-target inhibitors: Insights from design strategies, SARs, and therapeutic potential 微管蛋白/HDAC 双靶点抑制剂:从设计策略、SARs 和治疗潜力中获得的启示
IF 6 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-01 DOI: 10.1016/j.ejmech.2024.117022
Zhen Zhang , Rui Su , Junao Liu , Keyu Chen , Chengjun Wu , Pinghua Sun , Tiemin Sun
Microtubules, one of the cytoskeletons in eukaryotic cells, maintain the proper operation of several cellular functions. Additionally, they are regulated by the acetylation of HDAC6 and SIRT2 which affects microtubule dynamics. Given the fact that tubulin and HDAC inhibitors play a synergistic effect in the treatment of many cancers, the development of tubulin/HDAC dual-target inhibitors is conducive to addressing multiple limitations including drug resistance, dose toxicity, and unpredictable pharmacokinetic properties. At present, tubulin/HDAC dual-target inhibitors have been obtained in three main ways: uncleavable linked pharmacophores, cleavable linked pharmacophores, and modification of single-target drugs. Their therapeutic efficacy has been verified in vivo and in vitro assays. In this article, we reviewed the research progress of tubulin/HDAC dual inhibitors from design strategies, SARs, and biological activities, which may provide help for the discovery of novel tubulin/HDAC dual inhibitors.
微管是真核细胞中的细胞骨架之一,能维持多种细胞功能的正常运行。此外,微管受 HDAC6 和 SIRT2 的乙酰化调节,而乙酰化会影响微管的动力学。鉴于微管蛋白和 HDAC 抑制剂在许多癌症的治疗中发挥着协同作用,开发微管蛋白/HDAC 双靶点抑制剂有利于解决耐药性、剂量毒性和不可预测的药代动力学特性等多重限制。目前,获得微管蛋白/HDAC 双靶点抑制剂的途径主要有三种:不可逆连接药基、可裂解连接药基以及对单靶点药物进行修饰。它们的疗效已在体内和体外实验中得到验证。本文从设计策略、SARs和生物活性等方面综述了微管蛋白/HDAC双重抑制剂的研究进展,以期为新型微管蛋白/HDAC双重抑制剂的发现提供帮助。
{"title":"Tubulin/HDAC dual-target inhibitors: Insights from design strategies, SARs, and therapeutic potential","authors":"Zhen Zhang ,&nbsp;Rui Su ,&nbsp;Junao Liu ,&nbsp;Keyu Chen ,&nbsp;Chengjun Wu ,&nbsp;Pinghua Sun ,&nbsp;Tiemin Sun","doi":"10.1016/j.ejmech.2024.117022","DOIUrl":"10.1016/j.ejmech.2024.117022","url":null,"abstract":"<div><div>Microtubules, one of the cytoskeletons in eukaryotic cells, maintain the proper operation of several cellular functions. Additionally, they are regulated by the acetylation of HDAC6 and SIRT2 which affects microtubule dynamics. Given the fact that tubulin and HDAC inhibitors play a synergistic effect in the treatment of many cancers, the development of tubulin/HDAC dual-target inhibitors is conducive to addressing multiple limitations including drug resistance, dose toxicity, and unpredictable pharmacokinetic properties. At present, tubulin/HDAC dual-target inhibitors have been obtained in three main ways: uncleavable linked pharmacophores, cleavable linked pharmacophores, and modification of single-target drugs. Their therapeutic efficacy has been verified in <em>vivo</em> and in <em>vitro</em> assays. In this article, we reviewed the research progress of tubulin/HDAC dual inhibitors from design strategies, SARs, and biological activities, which may provide help for the discovery of novel tubulin/HDAC dual inhibitors.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"281 ","pages":"Article 117022"},"PeriodicalIF":6.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142562205","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}
引用次数: 0
Advance on the effects of algal carotenoids on inflammatory signaling pathways 藻类类胡萝卜素对炎症信号通路影响的研究进展
IF 6 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-01 DOI: 10.1016/j.ejmech.2024.117020
Yudi Wang , Xinrong Geng , Song Qin , Tuanjie Che , Libo Yan , Biao Yuan , Wenjun Li
The development of inflammation has an indispensable importance in the self-protection of the human body. However, over-inflammation may damage human health, and inflammatory pathways and inflammasomes have a significant impact on the onset of inflammation. Therefore, how to constrain the development of inflammation through inflammatory pathways or inflammasomes becomes a hot research issue. Carotenoids are a natural pigment and an active substance in algae, with anti-inflammatory and antioxidant effects. Many studies have shown that carotenoids have inhibitory effects on the inflammatory pathways and inflammasomes. In this review, we discussed the mechanism of carotenoids targeting those important inflammatory pathways and their effects on common inflammasome NLRP3 and inflammation-related diseases from the perspective of several inflammatory pathways, including p38 MAPK, IL-6/JAK/STAT3, and PI3K, with a focus on the targets and targeting effects of carotenoids on different inflammatory signaling pathways, and at last proposed possible anti-inflammatory targets.
炎症的发展对人体的自我保护具有不可或缺的重要意义。然而,过度炎症会损害人体健康,而炎症通路和炎性体对炎症的发生有重要影响。因此,如何通过炎症通路或炎症小体来制约炎症的发展成为研究热点。类胡萝卜素是一种天然色素,也是藻类中的一种活性物质,具有抗炎和抗氧化作用。许多研究表明,类胡萝卜素对炎症通路和炎症体具有抑制作用。在这篇综述中,我们从p38 MAPK、IL-6/JAK/STAT3和PI3K等几种炎症通路的角度探讨了类胡萝卜素靶向那些重要炎症通路的机制及其对常见炎症体NLRP3和炎症相关疾病的影响,重点讨论了类胡萝卜素对不同炎症信号通路的靶点和靶向作用,最后提出了可能的抗炎靶点。
{"title":"Advance on the effects of algal carotenoids on inflammatory signaling pathways","authors":"Yudi Wang ,&nbsp;Xinrong Geng ,&nbsp;Song Qin ,&nbsp;Tuanjie Che ,&nbsp;Libo Yan ,&nbsp;Biao Yuan ,&nbsp;Wenjun Li","doi":"10.1016/j.ejmech.2024.117020","DOIUrl":"10.1016/j.ejmech.2024.117020","url":null,"abstract":"<div><div>The development of inflammation has an indispensable importance in the self-protection of the human body. However, over-inflammation may damage human health, and inflammatory pathways and inflammasomes have a significant impact on the onset of inflammation. Therefore, how to constrain the development of inflammation through inflammatory pathways or inflammasomes becomes a hot research issue. Carotenoids are a natural pigment and an active substance in algae, with anti-inflammatory and antioxidant effects. Many studies have shown that carotenoids have inhibitory effects on the inflammatory pathways and inflammasomes. In this review, we discussed the mechanism of carotenoids targeting those important inflammatory pathways and their effects on common inflammasome NLRP3 and inflammation-related diseases from the perspective of several inflammatory pathways, including p38 MAPK, IL-6/JAK/STAT3, and PI3K, with a focus on the targets and targeting effects of carotenoids on different inflammatory signaling pathways, and at last proposed possible anti-inflammatory targets.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"281 ","pages":"Article 117020"},"PeriodicalIF":6.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142563217","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}
引用次数: 0
Design, synthesis, and evaluation of 4-(4-methyl-4H-1,2,4-triazol-3-yl)piperidine derivatives as potential glutaminyl cyclase isoenzyme inhibitors for the treatment of cancer 设计、合成和评估 4-(4-甲基-4H-1,2,4-三唑-3-基)哌啶衍生物作为治疗癌症的潜在谷氨酰胺环化酶同工酶抑制剂
IF 6 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-01 DOI: 10.1016/j.ejmech.2024.117019
Qingqing Zhou , Zhenxin Wu , Feixia Qin , Pan He , Zhuoran Wang , Fangyi Zhu , Ying Gao , Wei Xiong , Chenyang Li , Haiqiang Wu
Upregulated glutaminyl cyclase isoenzyme (isoQC) contributes to cancer development by catalyzing pE-CD47 generation and thus enhancing CD47-SIRPα binding and subsequent “don't eat me” signals. We thus consider that isoQC could represent a novel target for cancer therapy. We previously prepared a series of diphenyl conjugated imidazole derivatives (DPCIs) and evaluated their use as glutaminyl cyclase (QC) inhibitors. Here, a new series of DPCIs was rationally designed and synthesized. As anticipated, the analogues exhibited considerably improved inhibitory potency against both QC and isoQC. Crucially, these chemicals exhibited marked selectivity toward isoQC. Further assessments established that one selected compound (27) did not affect the viability of A549, H1299, PC9, or HEK293T cells or the body weight of mice. This compound did, however, reduce pE-CD47 levels in infected A549 cells (isoQC_OE and isoQC_KD) and exhibited apparent anti-cancer effects in vivo by downregulating the level of pE-CD47 via the inhibition of isoQC activity. Taken together, these findings indicated that the compounds synthesized in this study could represent potential QC/isoQC inhibitors for the treatment of cancers.
上调的谷氨酰胺酰环酶同工酶(isoQC)可催化 pE-CD47 的生成,从而增强 CD47-SIRPα 的结合和随后的 "别吃我 "信号,从而促进癌症的发展。因此,我们认为异QC可能是癌症治疗的一个新靶点。我们之前制备了一系列二苯基共轭咪唑衍生物(DPCIs),并评估了它们作为谷氨酰胺酰环化酶(QC)抑制剂的用途。在此,我们合理地设计并合成了一系列新的 DPCIs。正如预期的那样,这些类似物对 QC 和异 QC 的抑制效力都有显著提高。最重要的是,这些化学物质对异QC具有明显的选择性。进一步的评估表明,一种被选中的化合物(27)不会影响 A549、H1299、PC9 或 HEK293T 细胞的活力或小鼠的体重。不过,这种化合物确实降低了受感染的 A549 细胞(isoQC_OE 和 isoQC_KD)中 pE-CD47 的水平,并通过抑制 isoQC 活性下调 pE-CD47 的水平,在体内表现出明显的抗癌效果。综上所述,这些发现表明本研究合成的化合物可能是治疗癌症的潜在 QC/isoQC 抑制剂。
{"title":"Design, synthesis, and evaluation of 4-(4-methyl-4H-1,2,4-triazol-3-yl)piperidine derivatives as potential glutaminyl cyclase isoenzyme inhibitors for the treatment of cancer","authors":"Qingqing Zhou ,&nbsp;Zhenxin Wu ,&nbsp;Feixia Qin ,&nbsp;Pan He ,&nbsp;Zhuoran Wang ,&nbsp;Fangyi Zhu ,&nbsp;Ying Gao ,&nbsp;Wei Xiong ,&nbsp;Chenyang Li ,&nbsp;Haiqiang Wu","doi":"10.1016/j.ejmech.2024.117019","DOIUrl":"10.1016/j.ejmech.2024.117019","url":null,"abstract":"<div><div>Upregulated glutaminyl cyclase isoenzyme (isoQC) contributes to cancer development by catalyzing pE-CD47 generation and thus enhancing CD47-SIRPα binding and subsequent “don't eat me” signals. We thus consider that isoQC could represent a novel target for cancer therapy. We previously prepared a series of diphenyl conjugated imidazole derivatives (DPCIs) and evaluated their use as glutaminyl cyclase (QC) inhibitors. Here, a new series of DPCIs was rationally designed and synthesized. As anticipated, the analogues exhibited considerably improved inhibitory potency against both QC and isoQC. Crucially, these chemicals exhibited marked selectivity toward isoQC. Further assessments established that one selected compound (<strong>27</strong>) did not affect the viability of A549, H1299, PC9, or HEK293T cells or the body weight of mice. This compound did, however, reduce pE-CD47 levels in infected A549 cells (isoQC_OE and isoQC_KD) and exhibited apparent anti-cancer effects <em>in vivo</em> by downregulating the level of pE-CD47 via the inhibition of isoQC activity. Taken together, these findings indicated that the compounds synthesized in this study could represent potential QC/isoQC inhibitors for the treatment of cancers.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"281 ","pages":"Article 117019"},"PeriodicalIF":6.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142562206","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}
引用次数: 0
Discovery of the first selective and potent PROTAC degrader for the pseudokinase TRIB2 发现首个针对伪激酶 TRIB2 的选择性强效 PROTAC 降解剂
IF 6 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2024-11-01 DOI: 10.1016/j.ejmech.2024.117016
Chaowei Wen , Prathibha R. Gajjala , Yihan Liu , Bingzhong Chen , Mehtab S. Bal , Payal Sutaria , Qiao Yuanyuan , Yang Zheng , Yang Zhou , Jinwei Zhang , Weixue Huang , Xiaomei Ren , Zhen Wang , Ke Ding , Arul M. Chinnaiyan , Fengtao Zhou
Pseudokinase TRIB2, a member of the CAMK Ser/Thr protein kinase family, regulates various cellular processes through phosphorylation-independent mechanisms. Dysregulation of TRIB2 has been implicated in promoting tumor growth, metastasis, and therapy resistance, making it a promising target for cancer treatment. In this study, we designed and synthesized a series of TRIB2 PROTAC degraders by conjugating a TRIB2 binder 1 with VHL or CRBN ligands via linkers of varying lengths and compositions. Among these compounds, 5k demonstrated potent TRIB2 degradation with a DC50 value of 16.84 nM (95 % CI: 13.66–20.64 nM) in prostate cancer PC3 cells. Mechanistic studies revealed that 5k directly interacted with TRIB2, selectively inducing its degradation through a CRBN-dependent ubiquitin-proteasomal pathway. Moreover, 5k outperformed the TRIB2 binder alone in inhibiting cell proliferation and inducing apoptosis, confirming that TRIB2 protein degradation could be a promising therapeutic strategy for TRIB2-associated cancers. Additionally, compound 5k also serves as an effective tool for probing TRIB2 biology.
伪激酶 TRIB2 是 CAMK Ser/Thr 蛋白激酶家族的成员,通过磷酸化无关机制调节各种细胞过程。TRIB2 的失调与促进肿瘤生长、转移和耐药性有关,因此是一个很有前景的癌症治疗靶点。在这项研究中,我们设计并合成了一系列 TRIB2 PROTAC 降解剂,方法是通过不同长度和组成的连接体将 TRIB2 结合剂 1 与 VHL 或 CRBN 配体连接。在这些化合物中,5k 对前列腺癌 PC3 细胞的 TRIB2 降解效果显著,DC50 值为 16.84 nM(95% CI:13.66 - 20.64 nM)。机理研究显示,5k 与 TRIB2 直接相互作用,通过依赖 CRBN 的泛素-蛋白酶体途径选择性地诱导其降解。此外,5k 在抑制细胞增殖和诱导细胞凋亡方面的表现优于单独的 TRIB2 结合剂,这证实了 TRIB2 蛋白降解可能是治疗 TRIB2 相关癌症的一种有前景的策略。此外,化合物 5k 还是探究 TRIB2 生物学特性的有效工具。
{"title":"Discovery of the first selective and potent PROTAC degrader for the pseudokinase TRIB2","authors":"Chaowei Wen ,&nbsp;Prathibha R. Gajjala ,&nbsp;Yihan Liu ,&nbsp;Bingzhong Chen ,&nbsp;Mehtab S. Bal ,&nbsp;Payal Sutaria ,&nbsp;Qiao Yuanyuan ,&nbsp;Yang Zheng ,&nbsp;Yang Zhou ,&nbsp;Jinwei Zhang ,&nbsp;Weixue Huang ,&nbsp;Xiaomei Ren ,&nbsp;Zhen Wang ,&nbsp;Ke Ding ,&nbsp;Arul M. Chinnaiyan ,&nbsp;Fengtao Zhou","doi":"10.1016/j.ejmech.2024.117016","DOIUrl":"10.1016/j.ejmech.2024.117016","url":null,"abstract":"<div><div>Pseudokinase TRIB2, a member of the CAMK Ser/Thr protein kinase family, regulates various cellular processes through phosphorylation-independent mechanisms. Dysregulation of TRIB2 has been implicated in promoting tumor growth, metastasis, and therapy resistance, making it a promising target for cancer treatment. In this study, we designed and synthesized a series of TRIB2 PROTAC degraders by conjugating a TRIB2 binder <strong>1</strong> with VHL or CRBN ligands via linkers of varying lengths and compositions. Among these compounds, <strong>5k</strong> demonstrated potent TRIB2 degradation with a DC<sub>50</sub> value of 16.84 nM (95 % CI: 13.66–20.64 nM) in prostate cancer PC3 cells. Mechanistic studies revealed that <strong>5k</strong> directly interacted with TRIB2, selectively inducing its degradation through a CRBN-dependent ubiquitin-proteasomal pathway. Moreover, <strong>5k</strong> outperformed the TRIB2 binder alone in inhibiting cell proliferation and inducing apoptosis, confirming that TRIB2 protein degradation could be a promising therapeutic strategy for TRIB2-associated cancers. Additionally, compound <strong>5k</strong> also serves as an effective tool for probing TRIB2 biology.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"281 ","pages":"Article 117016"},"PeriodicalIF":6.0,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142562190","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}
引用次数: 0
期刊
European Journal of Medicinal Chemistry
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1