首页 > 最新文献

European Journal of Medicinal Chemistry最新文献

英文 中文
Target Validation and Drug Discovery for TNBC: Targeting the Lin28B/Let-7/PBK with the Ponicidin TNBC的靶点验证和药物发现:poniciidin靶向Lin28B/Let-7/PBK
IF 6.7 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-12-11 DOI: 10.1016/j.ejmech.2025.118474
Xingpeng Wang, Lian Zhong, Zhaodi Wang, Wenwen Xia, Dongning Shen, Yuan Sun, Bo Liu, Tingxiu Zhao, Yunshan Wu
Triple-negative breast cancer (TNBC), characterized by high aggressiveness and limited targeted therapeutic options, urgently requires novel therapeutic targets and drugs. Although the Lin28/Let-7 axis has shown great therapeutic potential in refractory tumors, its role and specific regulatory mechanisms in TNBC remain unclear. This study first demonstrated through multi-dimensional bioinformatic analyses that Lin28B is preferentially overexpressed in TNBC, closely associated with poor patient prognosis, and exhibits a distinct expression pattern compared to other breast cancer subtypes, establishing its potential as a TNBC-specific therapeutic target. Subsequently, through tiered computational screening of a natural compound library, ponicidin was identified as a potential Lin28B inhibitor. Molecular docking, molecular dynamics simulations, and surface plasmon resonance (SPR) assays suggested that ponicidin binds to the cold-shock domain (CSD) of Lin28B with high affinity, which may competitively block the interaction between Lin28B and Let-7 and potentially promote Lin28B degradation via the ubiquitin-proteasome pathway, thereby relieving Let-7 suppression. Functional experiments demonstrated that ponicidin dose-dependently inhibits proliferation, invasion, and induces apoptosis in TNBC cells (MDA-MB-231, 4T1), with low toxicity to normal mammary epithelial cells. Through bioinformatic analyses including Weighted Gene Co-expression Network Analysis (WGCNA) and Protein-Protein Interaction (PPI) network analysis, PDZ-binding kinase (PBK) was predicted as a core downstream hub gene of the Lin28B/Let-7 axis. Experimental validation confirmed that ponicidin upregulates the expression of Let-7 family members, thereby downregulating multiple oncoproteins including PBK, C-MYC, RAS, and HMGA2. Further verification using a 4T1 cell orthotopic tumor mouse model showed that ponicidin dose-dependently inhibits in vivo tumor growth, reduces Lin28B expression in tumor tissues, increases apoptotic regions. This study is the first to tentatively explore the potential mechanism by which ponicidin inhibits TNBC by targeting the Lin28B/Let-7/PBK axis, clarifying its dual mode of action ("binding inhibition + protein degradation"). It provides a promising candidate compound with both efficacy and safety for TNBC, while laying a theoretical and experimental foundation for the development of Lin28B-targeted drugs.
三阴性乳腺癌(TNBC)具有侵袭性强、靶向治疗选择有限的特点,迫切需要新的治疗靶点和药物。尽管Lin28/Let-7轴在难治性肿瘤中显示出巨大的治疗潜力,但其在TNBC中的作用和具体调控机制尚不清楚。本研究首次通过多维生物信息学分析证明了Lin28B在TNBC中优先过表达,与患者预后不良密切相关,并且与其他乳腺癌亚型相比表现出不同的表达模式,确立了其作为TNBC特异性治疗靶点的潜力。随后,通过对天然化合物文库的分层计算筛选,ponicidin被鉴定为潜在的Lin28B抑制剂。分子对接、分子动力学模拟和表面等离子体共振(SPR)实验表明,ponicidin以高亲和力结合Lin28B的冷休克结构域(CSD),可能竞争性地阻断Lin28B与Let-7的相互作用,并可能通过泛素-蛋白酶体途径促进Lin28B的降解,从而缓解Let-7的抑制。功能实验表明,ponicidin在TNBC细胞(mda - mb - 231,4t1)中具有剂量依赖性抑制增殖、侵袭和诱导凋亡,对正常乳腺上皮细胞具有低毒性。通过加权基因共表达网络分析(WGCNA)和蛋白-蛋白相互作用网络分析(PPI)等生物信息学分析,预测pdz结合激酶(PBK)是Lin28B/Let-7轴的核心下游枢纽基因。实验验证证实poniciidin上调Let-7家族成员的表达,从而下调PBK、C-MYC、RAS和HMGA2等多种癌蛋白。4T1细胞原位肿瘤小鼠模型进一步验证表明,ponicidin剂量依赖性地抑制体内肿瘤生长,降低肿瘤组织中Lin28B的表达,增加凋亡区域。本研究首次初步探讨了ponicidin通过靶向Lin28B/Let-7/PBK轴抑制TNBC的潜在机制,阐明了其“结合抑制+蛋白降解”的双重作用模式。为TNBC提供了一种既有效又安全的有前景的候选化合物,同时为开发lin28b靶向药物奠定了理论和实验基础。
{"title":"Target Validation and Drug Discovery for TNBC: Targeting the Lin28B/Let-7/PBK with the Ponicidin","authors":"Xingpeng Wang, Lian Zhong, Zhaodi Wang, Wenwen Xia, Dongning Shen, Yuan Sun, Bo Liu, Tingxiu Zhao, Yunshan Wu","doi":"10.1016/j.ejmech.2025.118474","DOIUrl":"https://doi.org/10.1016/j.ejmech.2025.118474","url":null,"abstract":"Triple-negative breast cancer (TNBC), characterized by high aggressiveness and limited targeted therapeutic options, urgently requires novel therapeutic targets and drugs. Although the Lin28/<em>Let-7</em> axis has shown great therapeutic potential in refractory tumors, its role and specific regulatory mechanisms in TNBC remain unclear. This study first demonstrated through multi-dimensional bioinformatic analyses that <em>Lin28B</em> is preferentially overexpressed in TNBC, closely associated with poor patient prognosis, and exhibits a distinct expression pattern compared to other breast cancer subtypes, establishing its potential as a TNBC-specific therapeutic target. Subsequently, through tiered computational screening of a natural compound library, ponicidin was identified as a potential Lin28B inhibitor. Molecular docking, molecular dynamics simulations, and surface plasmon resonance (SPR) assays suggested that ponicidin binds to the cold-shock domain (CSD) of Lin28B with high affinity, which may competitively block the interaction between Lin28B and <em>Let-7</em> and potentially promote Lin28B degradation via the ubiquitin-proteasome pathway, thereby relieving <em>Let-7</em> suppression. Functional experiments demonstrated that ponicidin dose-dependently inhibits proliferation, invasion, and induces apoptosis in TNBC cells (MDA-MB-231, 4T1), with low toxicity to normal mammary epithelial cells. Through bioinformatic analyses including Weighted Gene Co-expression Network Analysis (WGCNA) and Protein-Protein Interaction (PPI) network analysis, <strong>PDZ-binding kinase</strong> (PBK) was predicted as a core downstream hub gene of the Lin28B/<em>Let-7</em> axis. Experimental validation confirmed that ponicidin upregulates the expression of <em>Let-7</em> family members, thereby downregulating multiple oncoproteins including <em>PBK</em>, <em>C-MYC</em>, <em>RAS</em>, and <em>HMGA2</em>. Further verification using a 4T1 cell orthotopic tumor mouse model showed that ponicidin dose-dependently inhibits in vivo tumor growth, reduces <em>Lin28B</em> expression in tumor tissues, increases apoptotic regions. This study is the first to tentatively explore the potential mechanism by which ponicidin inhibits TNBC by targeting the Lin28B/<em>Let-7</em>/PBK axis, clarifying its dual mode of action (\"binding inhibition + protein degradation\"). It provides a promising candidate compound with both efficacy and safety for TNBC, while laying a theoretical and experimental foundation for the development of Lin28B-targeted drugs.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"144 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2025-12-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145718070","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
Corrigendum to “Thiol esters as chemical warheads of SARS-CoV-2 main protease (3CLpro) peptide-like inhibitors” [Eur. J. Med. Chem. 293 (2025) 117709] “硫醇酯作为SARS-CoV-2主要蛋白酶(3CLpro)肽样抑制剂的化学弹头”的勘误表[欧洲]。医学化学杂志。293 (2025)117709]
IF 6.7 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-12-10 DOI: 10.1016/j.ejmech.2025.118470
Xuehong Qiao, Menghan Cui, Zhiwei Yu, Ling Ma, Hailong Liu, Xingxing Yang, Yuan Chen, Dahong Li, Jinjing Che, Linxiang Zhao, Ruibin Su, Xuhong Ren, Shan Cen, Bin Lin, Xinhua He
The authors regret that the correct number for the National Natural Science Foundation of China for this article is 82341090, not 82273909 as stated in the original article.
很抱歉,本文国家自然科学基金的正确编号是82341090,而不是原文所述的82273909。
{"title":"Corrigendum to “Thiol esters as chemical warheads of SARS-CoV-2 main protease (3CLpro) peptide-like inhibitors” [Eur. J. Med. Chem. 293 (2025) 117709]","authors":"Xuehong Qiao, Menghan Cui, Zhiwei Yu, Ling Ma, Hailong Liu, Xingxing Yang, Yuan Chen, Dahong Li, Jinjing Che, Linxiang Zhao, Ruibin Su, Xuhong Ren, Shan Cen, Bin Lin, Xinhua He","doi":"10.1016/j.ejmech.2025.118470","DOIUrl":"https://doi.org/10.1016/j.ejmech.2025.118470","url":null,"abstract":"The authors regret that the correct number for the National Natural Science Foundation of China for this article is 82341090, not 82273909 as stated in the original article.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"11 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2025-12-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145718057","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 Glucagon-Like Peptide-1 Lysosomal Targeting Chimeras for Degradation of Extracellular and Membrane Proteins 胰高血糖素样肽-1溶酶体靶向嵌合体降解细胞外和膜蛋白的研究进展
IF 6.7 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-12-09 DOI: 10.1016/j.ejmech.2025.118473
Liquan Zhu, Haotian Liu, Xin Zeng, Ke Liu, Xiaozhen Liu, Zhuotao Yang, Yuchen Duan, Da Qian, Chaoqi He, Xuli Meng
Lysosome-targeting chimeras (LYTAC) represent a revolutionary technology for targeted protein degradation. However, the scarcity of highly efficient lysosomal transport receptors poses a major bottleneck to LYTAC advancement. In previous studies, we identified the glucagon-like peptide-1 receptor (GLP-1R) as a novel lysosomal transport receptor that promotes the degradation of membrane proteins. Here, leveraging the stability of semaglutide, we developed GLP-1R-mediated lysosomal-targeting chimeras (g-LYTAC), including antibody–peptide conjugate-based chimeras (APCTACs) targeting epidermal growth factor receptor (EGFR) and programmed cell death ligand 1 (PD-L1), and peptide–peptide conjugate-based chimeras (PPCTACs) targeting integrin. Optimized g-LYTAC exhibit enhanced potency in eliminating cell-surface targets, with efficacy dependent on GLP-1R expression and lysosomal activity. Specifically, APCTAC enhances T cell-mediated cytotoxicity against tumor cells. In mouse models, APCTAC mediates robust PD-L1 degradation, driving tumor-specific immune responses by converting the immunosuppressive tumor microenvironment into an immunostimulatory one. Notably, immune-checkpoint degradation therapy with APCTAC achieves comparable or superior antitumor efficacy while causing significantly less inflammatory damage than antibody therapy. This peptide-based LYTAC system offers a safer, minimally invasive strategy for cancer immunotherapy by combining immune-checkpoint degradation with immunomodulation, laying the groundwork for the development of peptide-based LYTAC as an effective cancer therapy.
溶酶体靶向嵌合体(LYTAC)是一种革命性的靶向蛋白质降解技术。然而,高效溶酶体转运受体的缺乏是LYTAC发展的主要瓶颈。在之前的研究中,我们发现胰高血糖素样肽-1受体(GLP-1R)是一种促进膜蛋白降解的新型溶酶体转运受体。利用semaglutide的稳定性,我们开发了glp - 1r介导的溶酶体靶向嵌合体(g-LYTAC),包括靶向表皮生长因子受体(EGFR)和程序性细胞死亡配体1 (PD-L1)的抗体-肽偶联基嵌合体(APCTACs),以及靶向整合素的肽-肽偶联基嵌合体(PPCTACs)。优化后的g-LYTAC在消除细胞表面靶标方面表现出更强的效力,其效力取决于GLP-1R的表达和溶酶体活性。具体来说,APCTAC增强了T细胞介导的对肿瘤细胞的细胞毒性。在小鼠模型中,APCTAC介导强大的PD-L1降解,通过将免疫抑制的肿瘤微环境转化为免疫刺激的微环境来驱动肿瘤特异性免疫反应。值得注意的是,与抗体治疗相比,APCTAC免疫检查点降解治疗具有相当或更好的抗肿瘤疗效,同时引起的炎症损伤明显减少。这种基于肽的LYTAC系统通过结合免疫检查点降解和免疫调节,为癌症免疫治疗提供了一种更安全、微创的策略,为基于肽的LYTAC作为一种有效的癌症治疗方法的发展奠定了基础。
{"title":"Development of Glucagon-Like Peptide-1 Lysosomal Targeting Chimeras for Degradation of Extracellular and Membrane Proteins","authors":"Liquan Zhu, Haotian Liu, Xin Zeng, Ke Liu, Xiaozhen Liu, Zhuotao Yang, Yuchen Duan, Da Qian, Chaoqi He, Xuli Meng","doi":"10.1016/j.ejmech.2025.118473","DOIUrl":"https://doi.org/10.1016/j.ejmech.2025.118473","url":null,"abstract":"Lysosome-targeting chimeras (LYTAC) represent a revolutionary technology for targeted protein degradation. However, the scarcity of highly efficient lysosomal transport receptors poses a major bottleneck to LYTAC advancement. In previous studies, we identified the glucagon-like peptide-1 receptor (GLP-1R) as a novel lysosomal transport receptor that promotes the degradation of membrane proteins. Here, leveraging the stability of semaglutide, we developed GLP-1R-mediated lysosomal-targeting chimeras (g-LYTAC), including antibody–peptide conjugate-based chimeras (APCTACs) targeting epidermal growth factor receptor (EGFR) and programmed cell death ligand 1 (PD-L1), and peptide–peptide conjugate-based chimeras (PPCTACs) targeting integrin. Optimized g-LYTAC exhibit enhanced potency in eliminating cell-surface targets, with efficacy dependent on GLP-1R expression and lysosomal activity. Specifically, APCTAC enhances T cell-mediated cytotoxicity against tumor cells. In mouse models, APCTAC mediates robust PD-L1 degradation, driving tumor-specific immune responses by converting the immunosuppressive tumor microenvironment into an immunostimulatory one. Notably, immune-checkpoint degradation therapy with APCTAC achieves comparable or superior antitumor efficacy while causing significantly less inflammatory damage than antibody therapy. This peptide-based LYTAC system offers a safer, minimally invasive strategy for cancer immunotherapy by combining immune-checkpoint degradation with immunomodulation, laying the groundwork for the development of peptide-based LYTAC as an effective cancer therapy.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"22 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145704012","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 Biological Evaluation of Hybrids from Both Nitrones with eNOS-Mimicking Activity and Selective iNOS Inhibitors for the Treatment of Ischemic Stroke 具有模拟iNOS活性和选择性iNOS抑制剂的nitro酮复合物治疗缺血性卒中的设计、合成和生物学评价
IF 6.7 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-12-08 DOI: 10.1016/j.ejmech.2025.118428
Weijie Jiao, Hui Ye, Duorui Ji, Mengshuang Huang, Ruichen Li, Jian Jia, Bowen Wang, Shurui Wang, Zhen Lei, Nan qin, Hong Wu, Xiaokun Li, Guiyue Wu, Yinglin Cui, Yihua Zhang, Jianbing Wu, Zhangjian Huang
Ischemic stroke (IS) involves complex pathologies such as excitotoxicity, oxidative stress, and inflammation. Targeting inducible nitric oxide synthase (iNOS) which produces damaging levels of NO, while sparing neuroprotective endothelial NOS (eNOS) activity, represents a promising therapeutic strategy. We designed and synthesized a series of hybrids from both nitrones with eNOS-mimicking activity and iNOS inhibitors. Among them, compound 13h exhibited selectivity for iNOS (49.2- and 43.3-fold selectivity over nNOS and eNOS, respectively). And 13h demonstrated significant neuroprotective effects across multiple in vitro models, including oxygen-glucose deprivation/reoxygenation (OGD/R) and H2O2-induced damage in neuronal and endothelial cells. In a transient middle cerebral artery occlusion (tMCAO) rat model, 13h (30 mg/kg, i.v.) markedly reduced cerebral infarction volume (>80%) and improved neurological function. Mechanistic studies suggest its efficacy stems from anti-oxidant, selective iNOS inhibition, and 3-nitrotyrosine (3-NT) suppression activities. Thus, 13h serves as a novel compound with a multi-target activity against IS.
缺血性中风(IS)涉及复杂的病理,如兴奋毒性、氧化应激和炎症。靶向诱导型一氧化氮合酶(iNOS)产生损伤水平的一氧化氮,同时保留神经保护性内皮一氧化氮合酶(eNOS)的活性,是一种很有前途的治疗策略。我们设计并合成了一系列具有模拟iNOS活性和iNOS抑制剂的硝基化合物。其中,化合物13h对iNOS具有选择性(分别是nNOS和eNOS的49.2倍和43.3倍)。13h在多个体外模型中显示出显著的神经保护作用,包括氧-葡萄糖剥夺/再氧化(OGD/R)和h2o2诱导的神经元和内皮细胞损伤。在短暂性大脑中动脉闭塞(tMCAO)大鼠模型中,13h (30 mg/kg,静脉注射)显著减少脑梗死体积(80%),改善神经功能。机制研究表明其功效源于抗氧化、选择性抑制iNOS和3-硝基酪氨酸(3-NT)抑制活性。因此,13h作为一种具有抗IS多靶点活性的新型化合物。
{"title":"Design, Synthesis, and Biological Evaluation of Hybrids from Both Nitrones with eNOS-Mimicking Activity and Selective iNOS Inhibitors for the Treatment of Ischemic Stroke","authors":"Weijie Jiao, Hui Ye, Duorui Ji, Mengshuang Huang, Ruichen Li, Jian Jia, Bowen Wang, Shurui Wang, Zhen Lei, Nan qin, Hong Wu, Xiaokun Li, Guiyue Wu, Yinglin Cui, Yihua Zhang, Jianbing Wu, Zhangjian Huang","doi":"10.1016/j.ejmech.2025.118428","DOIUrl":"https://doi.org/10.1016/j.ejmech.2025.118428","url":null,"abstract":"Ischemic stroke (IS) involves complex pathologies such as excitotoxicity, oxidative stress, and inflammation. Targeting inducible nitric oxide synthase (iNOS) which produces damaging levels of NO, while sparing neuroprotective endothelial NOS (eNOS) activity, represents a promising therapeutic strategy. We designed and synthesized a series of hybrids from both nitrones with eNOS-mimicking activity and iNOS inhibitors. Among them, compound <strong>13h</strong> exhibited selectivity for iNOS (49.2- and 43.3-fold selectivity over nNOS and eNOS, respectively). And <strong>13h</strong> demonstrated significant neuroprotective effects across multiple <em>in vitro</em> models, including oxygen-glucose deprivation/reoxygenation (OGD/R) and H<sub>2</sub>O<sub>2</sub>-induced damage in neuronal and endothelial cells. In a transient middle cerebral artery occlusion (tMCAO) rat model, <strong>13h</strong> (30 mg/kg, <em>i.v.</em>) markedly reduced cerebral infarction volume (&gt;80%) and improved neurological function. Mechanistic studies suggest its efficacy stems from anti-oxidant, selective iNOS inhibition, and 3-nitrotyrosine (3-NT) suppression activities. Thus, <strong>13h</strong> serves as a novel compound with a multi-target activity against IS.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"10 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145696969","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
Exploring novel aromatase cytochrome P450-binding ligands: Molecular docking and QSAR study of phorbol esters as aromatase inhibitors 探索新的芳香化酶细胞色素p450结合配体:酚酯作为芳香化酶抑制剂的分子对接和QSAR研究
IF 5.9 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-12-06 DOI: 10.1016/j.ejmech.2025.118458
Abdellah Ezzanad , Felipe Escobar-Montaño , Jesus Sánchez-Márquez , David Zorrilla , Victor Coca , Glen J.P. McCann , Neill Horley , José Manuel Botubol-Ares , Rosa Durán-Patrón , Antonio J. Macías-Sánchez , Alan Christy Hunter , Rosario Hernández-Galán
A library comprising thirty-three natural and semi-synthetic phorbol derivatives was constructed and subsequently evaluated for aromatase (CYP19A1) inhibitory activity. The primary goal was to identify novel aromatase inhibitors through selective esterifications at the C-12, C-13 and C-20 positions of the phorbol skeleton. Twenty-two phorbol esters, fifteen of which were previously undescribed (59, 12 and 1624), were obtained via semi-synthetic modifications. Additionally, ten 12-deoxyphorbol esters (2534), four bearing an extra acyloxy chain at C-16, isolated from Euphorbia resinifera, were tested. Inhibition studies revealed that 12,13-diesters bearing medium-sized ester moieties (such as isobutyrate, tigliate or phenylacetate) exhibited the highest potency (IC50 = 0.9–6.4 μM). Conversely, shorter or longer ester chains significantly reduced activity (IC50 values ranging from 22.4 to 29.8 μM). An enhancement of potency was observed upon reduction of the α,β-unsaturated ketone at C-3 to an allylic alcohol. Molecular docking suggested that the phorbol derivatives bind within the aromatase activator-binding pocket, exerting their inhibitory actions through hydrogen-bond interactions with protein residues, independent of the heme group. The inhibitory potency correlated with the ability to establish these interactions. The activities of the most potent compounds are comparable to nonsteroidal, non-heme-binding aromatase inhibitors, such as stilbene and coumarin derivatives. Moreover, 3D-QSAR models were developed, with the most robust model (R2 = 0.908) highlighting the molecular descriptors NQC14 and AP as crucial modulators of the anti-aromatase activity. These findings provide a framework for future optimization of phorbol ester as selective aromatase inhibitors.
构建了一个包含33种天然和半合成phorbol衍生物的文库,并随后评估了芳香化酶(CYP19A1)抑制活性。主要目的是通过在phorbol骨架的C-12, C-13和C-20位置选择性酯化鉴定新的芳香酶抑制剂。通过半合成改性得到了22种佛波酯,其中15种以前未被描述过(5-9、12和16-24)。此外,从大戟树脂中分离出10个12-脱氧酚酯(25-34),其中4个在C-16上有一个额外的酰基链。抑制研究表明,含有中等酯段的12,13-二酯(如异丁酸酯、替格酸酯或苯乙酸酯)具有最高的抑制效力(IC50 = 0.9-6.4 μM)。相反,较短或较长的酯链显著降低活性(IC50值为22.4 ~ 29.8 μM)。在C-3处α,β-不饱和酮还原成烯丙醇后,药效增强。分子对接表明,phorbol衍生物结合在芳香酶激活物结合口袋内,通过与蛋白质残基的氢键相互作用发挥其抑制作用,独立于血红素基团。抑制效力与建立这些相互作用的能力相关。最有效的化合物的活性可与非甾体、非血红素结合的芳香酶抑制剂相媲美,如苯乙烯和香豆素衍生物。此外,我们还建立了3D-QSAR模型,其中最稳健的模型(R2 = 0.908)突出了分子描述子NQC14和AP是抗芳香酶活性的关键调节剂。这些发现为未来优化佛波酯作为选择性芳香酶抑制剂提供了一个框架。
{"title":"Exploring novel aromatase cytochrome P450-binding ligands: Molecular docking and QSAR study of phorbol esters as aromatase inhibitors","authors":"Abdellah Ezzanad ,&nbsp;Felipe Escobar-Montaño ,&nbsp;Jesus Sánchez-Márquez ,&nbsp;David Zorrilla ,&nbsp;Victor Coca ,&nbsp;Glen J.P. McCann ,&nbsp;Neill Horley ,&nbsp;José Manuel Botubol-Ares ,&nbsp;Rosa Durán-Patrón ,&nbsp;Antonio J. Macías-Sánchez ,&nbsp;Alan Christy Hunter ,&nbsp;Rosario Hernández-Galán","doi":"10.1016/j.ejmech.2025.118458","DOIUrl":"10.1016/j.ejmech.2025.118458","url":null,"abstract":"<div><div>A library comprising thirty-three natural and semi-synthetic phorbol derivatives was constructed and subsequently evaluated for aromatase (CYP19A1) inhibitory activity. The primary goal was to identify novel aromatase inhibitors through selective esterifications at the C-12, C-13 and C-20 positions of the phorbol skeleton. Twenty-two phorbol esters, fifteen of which were previously undescribed (<strong>5</strong>–<strong>9</strong>, <strong>12</strong> and <strong>16</strong>–<strong>24</strong>), were obtained via semi-synthetic modifications. Additionally, ten 12-deoxyphorbol esters (<strong>25</strong>–<strong>34</strong>), four bearing an extra acyloxy chain at C-16, isolated from <em>Euphorbia resinifera</em>, were tested. Inhibition studies revealed that 12,13-diesters bearing medium-sized ester moieties (such as isobutyrate, tigliate or phenylacetate) exhibited the highest potency (IC<sub>50</sub> = 0.9–6.4 μM). Conversely, shorter or longer ester chains significantly reduced activity (IC<sub>50</sub> values ranging from 22.4 to 29.8 μM). An enhancement of potency was observed upon reduction of the <em>α</em>,<em>β</em>-unsaturated ketone at C-3 to an allylic alcohol. Molecular docking suggested that the phorbol derivatives bind within the aromatase activator-binding pocket, exerting their inhibitory actions through hydrogen-bond interactions with protein residues, independent of the heme group. The inhibitory potency correlated with the ability to establish these interactions. The activities of the most potent compounds are comparable to nonsteroidal, non-heme-binding aromatase inhibitors, such as stilbene and coumarin derivatives. Moreover, 3D-QSAR models were developed, with the most robust model (R<sup>2</sup> = 0.908) highlighting the molecular descriptors NQC<sub>14</sub> and AP as crucial modulators of the anti-aromatase activity. These findings provide a framework for future optimization of phorbol ester as selective aromatase inhibitors.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"303 ","pages":"Article 118458"},"PeriodicalIF":5.9,"publicationDate":"2025-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145690090","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 Novel Small Molecule Inhibitor Targeting the Tumor Promoting Effect of Transcription Factor PLAGL2 靶向转录因子PLAGL2促肿瘤作用的新型小分子抑制剂的发现
IF 6.7 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-12-06 DOI: 10.1016/j.ejmech.2025.118471
Yushi Ding, Zhi Wang, Xingyu Xia, Jiayi Wang, Zhenyu Li, Congying Gu, Chenyan Hao, Kang Xu, Mengkang Gao, Hao Yin, Siheng Chen, Xin Gong, Wang Zhou, Min Zhang, Yong Yang, Dayong Zhang, Beiying Dai, Menghan Zhang
Pleiomorphic adenoma-like protein 2 (PLAGL2) is a key player in the development of hepatocellular carcinoma (HCC) and other malignant tumors. High levels of PLAGL2 expression are associated with poor prognosis in cancer. While the potential of PLAGL2 as a therapeutic target for HCC has been recognized, there is a lack of research on small-molecule inhibitors targeting PLAGL2. In this study, a series of small-molecule inhibitors of PLAGL2 transcriptional activity were developed through virtual screening and structure optimization. Among which, compounds C7 and C8 potently suppressed PLAGL2 transcriptional activity, leading to reduced proliferation, colony formation, migration, invasion, cell cycle arrest, and apoptosis in HCC cells. Compound C8 exhibited stronger binding affinity to PLALG2 than compound C7. Furthermore, C8 disrupted extracellular matrix organization and suppressed the PI3K-AKT pathway by reducing AKT phosphorylation. It effectively inhibited tumor growth in HCCLM3 xenograft tumor models while demonstrating a favorable safety profile. Taken together, this study introduces a promising 3-(Phenylsulfonamido) benzamide derivative as a novel approach to targeting PLAGL2 transcriptional activity, laying a foundation for future investigations in anti-tumor therapy.
多形性腺瘤样蛋白2 (PLAGL2)在肝细胞癌(HCC)和其他恶性肿瘤的发展中起着关键作用。高水平的PLAGL2表达与癌症预后不良相关。虽然人们已经认识到PLAGL2作为HCC治疗靶点的潜力,但缺乏针对PLAGL2的小分子抑制剂的研究。本研究通过虚拟筛选和结构优化,开发了一系列PLAGL2转录活性的小分子抑制剂。其中化合物C7和C8能有效抑制PLAGL2转录活性,导致HCC细胞增殖、集落形成、迁移、侵袭、细胞周期阻滞和细胞凋亡减少。化合物C8比化合物C7对PLALG2具有更强的结合亲和力。此外,C8破坏细胞外基质组织,通过降低AKT磷酸化抑制PI3K-AKT通路。它在HCCLM3异种移植肿瘤模型中有效抑制肿瘤生长,同时显示出良好的安全性。综上所述,本研究介绍了一种有前景的3-(苯基磺胺)苯酰胺衍生物,作为靶向PLAGL2转录活性的新方法,为未来抗肿瘤治疗的研究奠定了基础。
{"title":"Discovery of Novel Small Molecule Inhibitor Targeting the Tumor Promoting Effect of Transcription Factor PLAGL2","authors":"Yushi Ding, Zhi Wang, Xingyu Xia, Jiayi Wang, Zhenyu Li, Congying Gu, Chenyan Hao, Kang Xu, Mengkang Gao, Hao Yin, Siheng Chen, Xin Gong, Wang Zhou, Min Zhang, Yong Yang, Dayong Zhang, Beiying Dai, Menghan Zhang","doi":"10.1016/j.ejmech.2025.118471","DOIUrl":"https://doi.org/10.1016/j.ejmech.2025.118471","url":null,"abstract":"Pleiomorphic adenoma-like protein 2 (PLAGL2) is a key player in the development of hepatocellular carcinoma (HCC) and other malignant tumors. High levels of PLAGL2 expression are associated with poor prognosis in cancer. While the potential of PLAGL2 as a therapeutic target for HCC has been recognized, there is a lack of research on small-molecule inhibitors targeting PLAGL2. In this study, a series of small-molecule inhibitors of PLAGL2 transcriptional activity were developed through virtual screening and structure optimization. Among which, compounds C7 and C8 potently suppressed PLAGL2 transcriptional activity, leading to reduced proliferation, colony formation, migration, invasion, cell cycle arrest, and apoptosis in HCC cells. Compound C8 exhibited stronger binding affinity to PLALG2 than compound C7. Furthermore, C8 disrupted extracellular matrix organization and suppressed the PI3K-AKT pathway by reducing AKT phosphorylation. It effectively inhibited tumor growth in HCCLM3 xenograft tumor models while demonstrating a favorable safety profile. Taken together, this study introduces a promising 3-(Phenylsulfonamido) benzamide derivative as a novel approach to targeting PLAGL2 transcriptional activity, laying a foundation for future investigations in anti-tumor therapy.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"1 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2025-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145690088","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
Ligand-Based Design and Optimization of Tecovirimat Derivatives as Potent Antiviral Agents Against Monkeypox Virus 基于配体的抗猴痘病毒特可维美衍生物的设计与优化
IF 6.7 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-12-05 DOI: 10.1016/j.ejmech.2025.118447
Yuanguang Chen, Sumei Yang, Xingyu Zhou, Hongyang Yi, Dizhen Liang, Guanguan Li, Xiang Liu, Peisen Zheng, Yongqing Liu, Xinshan Deng, Xumu Zhang, Qifan Zhou, Hongzhou Lu
Among orthopoxviruses, monkeypox virus (MPXV) is currently the most significant human pathogen due to ongoing outbreaks and global spread. Tecovirimat, an FDA-approved antiviral for orthopoxviruses, inhibits the viral p37 protein essential for viral egress and is used under expanded access for monkeypox treatment. To develop more accessible inhibitors and enhance structure-activity relationship (SAR) research, a new library of tecovirimat derivatives has been designed and synthesized. These compounds underwent phenotype determination to evaluate their antiviral properties, leading to the identification of C12 (EC50 = 12.14 nM, CC50 = 304.63 μM) as a promising lead compound. C12 demonstrated stable high plasma exposure following oral administration in mice, achieving a bioavailability (F) of 162% and extensive distribution within the plasma. It also demonstrated low toxicity and good tolerability in mice, with a single dose of 2000 mg/kg or repeated doses of 100 mg/kg once daily for 14 days. Additionally, the P37 protein structure of VACN was employed for molecular docking to investigate potential binding interactions. Computational simulations suggest that C12 targets the same protein as tecovirimat, offering valuable insights for the rational design of next-generation analogs.
在正痘病毒中,猴痘病毒(MPXV)由于持续暴发和全球传播,目前是最重要的人类病原体。Tecovirimat是一种fda批准的用于正痘病毒的抗病毒药物,可抑制病毒输出所必需的病毒p37蛋白,并被广泛用于猴痘治疗。为了开发更容易获得的抑制剂和加强结构-活性关系(SAR)的研究,设计和合成了一个新的tecovirimat衍生物库。这些化合物通过表型测定来评估其抗病毒性能,最终确定C12 (EC50 = 12.14 nM, CC50 = 304.63 μM)为有希望的先导化合物。C12在小鼠口服给药后表现出稳定的高血浆暴露,生物利用度(F)达到162%,并在血浆中广泛分布。它在小鼠中也显示出低毒性和良好的耐受性,单次剂量为2000 mg/kg或重复剂量为100 mg/kg,每天一次,持续14天。此外,利用VACN的P37蛋白结构进行分子对接,研究潜在的结合相互作用。计算模拟表明,C12与tecovirimat靶向相同的蛋白质,为下一代类似物的合理设计提供了有价值的见解。
{"title":"Ligand-Based Design and Optimization of Tecovirimat Derivatives as Potent Antiviral Agents Against Monkeypox Virus","authors":"Yuanguang Chen, Sumei Yang, Xingyu Zhou, Hongyang Yi, Dizhen Liang, Guanguan Li, Xiang Liu, Peisen Zheng, Yongqing Liu, Xinshan Deng, Xumu Zhang, Qifan Zhou, Hongzhou Lu","doi":"10.1016/j.ejmech.2025.118447","DOIUrl":"https://doi.org/10.1016/j.ejmech.2025.118447","url":null,"abstract":"Among <em>orthopoxviruses</em>, monkeypox virus (MPXV) is currently the most significant human pathogen due to ongoing outbreaks and global spread. Tecovirimat, an FDA-approved antiviral for <em>orthopoxviruses</em>, inhibits the viral p37 protein essential for viral egress and is used under expanded access for monkeypox treatment. To develop more accessible inhibitors and enhance structure-activity relationship (SAR) research, a new library of tecovirimat derivatives has been designed and synthesized. These compounds underwent phenotype determination to evaluate their antiviral properties, leading to the identification of C12 (EC<sub>50</sub> = 12.14 nM, CC<sub>50</sub> = 304.63 μM) as a promising lead compound. C12 demonstrated stable high plasma exposure following oral administration in mice, achieving a bioavailability (F) of 162% and extensive distribution within the plasma. It also demonstrated low toxicity and good tolerability in mice, with a single dose of 2000 mg/kg or repeated doses of 100 mg/kg once daily for 14 days. Additionally, the P37 protein structure of VACN was employed for molecular docking to investigate potential binding interactions. Computational simulations suggest that C12 targets the same protein as tecovirimat, offering valuable insights for the rational design of next-generation analogs.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"11 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2025-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145690086","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 α-Carboline fused benzofuran-Containing Anticancer Agents as Rapid Apoptosis Inducers through Intrinsic Pathway α-卡波林融合含苯并呋喃抗癌药物通过内在途径作为细胞凋亡快速诱导因子的发现
IF 6.7 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-12-05 DOI: 10.1016/j.ejmech.2025.118464
Fang Lin, Zhangshun Luo, Qinyu Wu, Lijie Lv, Zhonghong Chen, Yi He, Hai-Xin Yuan, Yahui Zhao, Shenyou Nie
Apoptosis, a programmed form of cell death, typically requires several hours to initiate cytochrome c release following stimuli. Facilitating apoptosis plays a decisive role in eliminating detrimental cells as well as in cancer treatment. In this study, diverse novel α-carboline-fused benzofuran collections were efficiently achieved via C-H activation/Lossen rearrangement/ [4+2] cyclization cascades. Among them, compound 10 was found to rapidly induce apoptosis through the intrinsic pathway by releasing cytochrome c within 10 minutes, followed by cleavage and activation of caspase-9, caspase-3, and PARP1. Mechanistically, compound 10 was found to potently inhibit several signaling cascades that are important for cell proliferation and survival, including AKT, ERK1/2, STAT1, STAT3, and S6K, suggesting its role as a multi-kinase inhibitor in inducing rapid apoptosis. Furthermore, compound 10 significantly inhibited tumor growth in a mouse xenograft model. Overall, this novel inducer of rapid apoptosis may serve as a powerful tool for further elucidation of apoptotic mechanisms and holds promising therapeutic potential in clinical cancer treatment.
细胞凋亡是细胞死亡的一种程序化形式,通常在刺激后需要数小时启动细胞色素c的释放。促进细胞凋亡在清除有害细胞和癌症治疗中起着决定性的作用。在本研究中,通过C-H活化/Lossen重排/[4+2]环化级联有效地实现了多种新型α-羰基融合苯并呋喃的收集。其中化合物10在10分钟内释放细胞色素c,随后裂解和激活caspase-9、caspase-3和PARP1,通过内在途径快速诱导细胞凋亡。在机制上,化合物10被发现能有效抑制几种对细胞增殖和存活至关重要的信号级联反应,包括AKT、ERK1/2、STAT1、STAT3和S6K,这表明它是一种多激酶抑制剂,可诱导细胞快速凋亡。此外,化合物10在小鼠异种移植瘤模型中显著抑制肿瘤生长。总之,这种新型的快速凋亡诱导剂可以作为进一步阐明细胞凋亡机制的有力工具,在临床癌症治疗中具有良好的治疗潜力。
{"title":"Discovery of α-Carboline fused benzofuran-Containing Anticancer Agents as Rapid Apoptosis Inducers through Intrinsic Pathway","authors":"Fang Lin, Zhangshun Luo, Qinyu Wu, Lijie Lv, Zhonghong Chen, Yi He, Hai-Xin Yuan, Yahui Zhao, Shenyou Nie","doi":"10.1016/j.ejmech.2025.118464","DOIUrl":"https://doi.org/10.1016/j.ejmech.2025.118464","url":null,"abstract":"Apoptosis, a programmed form of cell death, typically requires several hours to initiate cytochrome c release following stimuli. Facilitating apoptosis plays a decisive role in eliminating detrimental cells as well as in cancer treatment. In this study, diverse novel α-carboline-fused benzofuran collections were efficiently achieved via C-H activation/Lossen rearrangement/ [4+2] cyclization cascades. Among them, compound <strong>10</strong> was found to rapidly induce apoptosis through the intrinsic pathway by releasing cytochrome c within 10 minutes, followed by cleavage and activation of caspase-9, caspase-3, and PARP1. Mechanistically, compound <strong>10</strong> was found to potently inhibit several signaling cascades that are important for cell proliferation and survival, including AKT, ERK1/2, STAT1, STAT3, and S6K, suggesting its role as a multi-kinase inhibitor in inducing rapid apoptosis. Furthermore, compound <strong>10</strong> significantly inhibited tumor growth in a mouse xenograft model. Overall, this novel inducer of rapid apoptosis may serve as a powerful tool for further elucidation of apoptotic mechanisms and holds promising therapeutic potential in clinical cancer treatment.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"1 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2025-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145690089","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
Benzoheterocyclic analogues of dotinurad as orally efficacious human urate transporter 1 inhibitors: Design, synthesis, structure-activity relationships, and pharmacokinetic studies 多替努酸的苯并杂环类似物作为口服有效的人类尿酸转运蛋白1抑制剂:设计、合成、构效关系和药代动力学研究
IF 5.9 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-12-05 DOI: 10.1016/j.ejmech.2025.118459
Linjian Zhang , Kaijun Su , Haoshu Zhang , Guowei Zhang , Jian Zhou , Jiaqi Liang , Xiang Li , Xiaojin Zhang
A series of benzoheterocyclic analogues of Dotinurad were designed and synthesized as URAT1 inhibitors via bioisosterism and linker modification strategies. Among the 19 synthesized derivatives, compounds 9, 13, and 14 (bearing benzimidazole, benzotetrahydropyrrole, and benzo[b]thiazine 1,1-dioxide scaffolds, respectively) exhibited acceptable inhibitory potency (IC50: 69.81–441.51 nM), favorable metabolic stability, and improved pharmacokinetic profiles compared to Dotinurad. Notably, despite showing weaker in vitro URAT1 inhibition than the Dotinurad, these three candidates demonstrated significantly superior uricosuric activity in hyperuricemic rats. Mechanistic investigation revealed that the substantial excretion of unchanged prototype compounds into the urine ensured a high local concentration within the renal tubule lumen, which effectively compensated for their moderate in vitro affinity. These findings validated the design strategy and highlighted compounds 9, 13, and 14 as promising candidates for hyperuricemia treatment.
通过生物等构和连接体修饰等策略,设计并合成了一系列苯并杂环Dotinurad类似物作为URAT1抑制剂。在合成的19个衍生物中,化合物9、13和14(分别含有苯并咪唑、苯并四氢吡罗和苯并[b]噻嗪1,1-二氧化氮支架)与Dotinurad相比具有良好的抑制效力(IC50: 69.81 ~ 441.51 nM)、良好的代谢稳定性和更好的药代动力学特征。值得注意的是,尽管体外URAT1抑制作用弱于Dotinurad,但这三种候选物在高尿酸血症大鼠中表现出明显优越的尿酸活性。机制研究表明,未改变的原型化合物大量排泄到尿液中,确保了肾小管腔内的高局部浓度,这有效地补偿了它们在体外的中等亲和力。这些发现验证了设计策略,并强调化合物9、13和14是治疗高尿酸血症的有希望的候选者。
{"title":"Benzoheterocyclic analogues of dotinurad as orally efficacious human urate transporter 1 inhibitors: Design, synthesis, structure-activity relationships, and pharmacokinetic studies","authors":"Linjian Zhang ,&nbsp;Kaijun Su ,&nbsp;Haoshu Zhang ,&nbsp;Guowei Zhang ,&nbsp;Jian Zhou ,&nbsp;Jiaqi Liang ,&nbsp;Xiang Li ,&nbsp;Xiaojin Zhang","doi":"10.1016/j.ejmech.2025.118459","DOIUrl":"10.1016/j.ejmech.2025.118459","url":null,"abstract":"<div><div>A series of benzoheterocyclic analogues of Dotinurad were designed and synthesized as URAT1 inhibitors via bioisosterism and linker modification strategies. Among the 19 synthesized derivatives, compounds <strong>9</strong>, <strong>13</strong>, and <strong>14</strong> (bearing benzimidazole, benzotetrahydropyrrole, and benzo[<em>b</em>]thiazine 1,1-dioxide scaffolds, respectively) exhibited acceptable inhibitory potency (IC<sub>50</sub>: 69.81–441.51 nM), favorable metabolic stability, and improved pharmacokinetic profiles compared to Dotinurad. Notably, despite showing weaker <em>in vitro</em> URAT1 inhibition than the Dotinurad, these three candidates demonstrated significantly superior uricosuric activity in hyperuricemic rats. Mechanistic investigation revealed that the substantial excretion of unchanged prototype compounds into the urine ensured a high local concentration within the renal tubule lumen, which effectively compensated for their moderate <em>in vitro</em> affinity. These findings validated the design strategy and highlighted compounds <strong>9</strong>, <strong>13</strong>, and <strong>14</strong> as promising candidates for hyperuricemia treatment.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"303 ","pages":"Article 118459"},"PeriodicalIF":5.9,"publicationDate":"2025-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145690087","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 biological evaluation of novel FXIa inhibitors featuring five-membered heterocycles as P2’ fragments 以五元杂环为P2片段的新型FXIa抑制剂的设计、合成及生物学评价
IF 6.7 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-12-04 DOI: 10.1016/j.ejmech.2025.118457
Jie Wu, Xiaoqian Wang, Huachao Zhu, Ping Gong, Tonghui Wang
Thrombus formation within blood vessels poses a serious threat to human health and is closely associated with various cardiovascular disorders. Therefore, developing novel anticoagulants with reduced bleeding risk holds significant clinical importance. Factor XIa (FXIa), a key enzyme in the intrinsic coagulation pathway, has emerged as an attractive target for safer anticoagulant therapy. In this study, a series of novel small-molecule FXIa inhibitors were designed based on the scaffold of Asundexian through a bioisosteric replacement strategy. Starting from compound F22, structural optimization at the P2’ region was conducted by replacing the amide with non-classical heterocycles, aiming to improve inhibitory potency, selectivity, and metabolic stability. Among the synthesized analogs, compound FE12 exhibited potent FXIa inhibition (IC50 = 4.4 nM), high selectivity over PKal (SI = 60.3), and favorable metabolic stability (T1/2 = 38.6 min in HLMs). Consistent with the in vitro enzyme assay, FE12 significantly prolonged aPTT in a dose-dependent manner, comparable to Asundexian. Molecular docking indicated that FE12 retains key hydrogen-bonding and water-mediated interactions within the FXIa active site, whereas its conformation in PKal shifts and loses these interactions, explaining its potent FXIa inhibitory activity and weak PKal inhibition. In the FeCl3-induced rat thrombosis model, FE12 effectively inhibited thrombus formation comparable to Asundexian. In the mouse tail bleeding assay, FE12 did not cause a significant prolongation of bleeding time, indicating a minimal effect on hemostasis. Furthermore, acute toxicity evaluation demonstrated its good safety and tolerability. Overall, FE12 exhibits an excellent balance of potency, selectivity and safety, representing a promising lead compound for the development of small-molecule FXIa inhibitors.
血管内血栓的形成严重威胁着人类的健康,与各种心血管疾病密切相关。因此,开发具有降低出血风险的新型抗凝剂具有重要的临床意义。因子XIa (FXIa)是内在凝血途径中的关键酶,已成为更安全的抗凝治疗的一个有吸引力的靶点。本研究基于assundexian支架,通过生物等构置换策略设计了一系列新型小分子FXIa抑制剂。从化合物F22开始,以非经典杂环取代酰胺,对P2′区进行结构优化,以提高抑制效力、选择性和代谢稳定性。在所合成的类似物中,化合物FE12对FXIa具有较强的抑制作用(IC50 = 4.4 nM),对PKal具有较高的选择性(SI = 60.3),代谢稳定性较好(T1/2 = 38.6 min)。与体外酶分析一致,FE12以剂量依赖的方式显著延长aPTT,与assundexian相当。分子对接表明,FE12在FXIa活性位点保留了关键的氢键和水介导的相互作用,而其在PKal中的构象发生了变化并失去了这些相互作用,这解释了其强大的FXIa抑制活性和弱PKal抑制作用。在fecl3诱导的大鼠血栓模型中,FE12对血栓形成的抑制作用与Asundexian相当。在小鼠尾部出血实验中,FE12没有引起出血时间的显著延长,表明对止血的影响很小。急性毒性评价表明其具有良好的安全性和耐受性。总的来说,FE12在效价、选择性和安全性方面表现出良好的平衡,是开发小分子FXIa抑制剂的一个很有前景的先导化合物。
{"title":"Design, synthesis and biological evaluation of novel FXIa inhibitors featuring five-membered heterocycles as P2’ fragments","authors":"Jie Wu, Xiaoqian Wang, Huachao Zhu, Ping Gong, Tonghui Wang","doi":"10.1016/j.ejmech.2025.118457","DOIUrl":"https://doi.org/10.1016/j.ejmech.2025.118457","url":null,"abstract":"Thrombus formation within blood vessels poses a serious threat to human health and is closely associated with various cardiovascular disorders. Therefore, developing novel anticoagulants with reduced bleeding risk holds significant clinical importance. Factor XIa (FXIa), a key enzyme in the intrinsic coagulation pathway, has emerged as an attractive target for safer anticoagulant therapy. In this study, a series of novel small-molecule FXIa inhibitors were designed based on the scaffold of Asundexian through a bioisosteric replacement strategy. Starting from compound <strong>F22</strong>, structural optimization at the P2’ region was conducted by replacing the amide with non-classical heterocycles, aiming to improve inhibitory potency, selectivity, and metabolic stability. Among the synthesized analogs, compound <strong>FE12</strong> exhibited potent FXIa inhibition (IC<sub>50</sub> = 4.4 nM), high selectivity over PKal (SI = 60.3), and favorable metabolic stability (T<sub>1/2</sub> = 38.6 min in HLMs). Consistent with the <em>in vitro</em> enzyme assay, <strong>FE12</strong> significantly prolonged aPTT in a dose-dependent manner, comparable to Asundexian. Molecular docking indicated that <strong>FE12</strong> retains key hydrogen-bonding and water-mediated interactions within the FXIa active site, whereas its conformation in PKal shifts and loses these interactions, explaining its potent FXIa inhibitory activity and weak PKal inhibition. In the FeCl<sub>3</sub>-induced rat thrombosis model, <strong>FE12</strong> effectively inhibited thrombus formation comparable to Asundexian. In the mouse tail bleeding assay, <strong>FE12</strong> did not cause a significant prolongation of bleeding time, indicating a minimal effect on hemostasis. Furthermore, acute toxicity evaluation demonstrated its good safety and tolerability. Overall, <strong>FE12</strong> exhibits an excellent balance of potency, selectivity and safety, representing a promising lead compound for the development of small-molecule FXIa inhibitors.","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"1 1","pages":""},"PeriodicalIF":6.7,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145674045","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:604180095
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