首页 > 最新文献

European Journal of Medicinal Chemistry最新文献

英文 中文
Targeting paracaspase1 (MALT1) for cancer therapy: Updated progress and study on structure-activity relationships (SARs) 靶向Paracaspase1 (MALT1)治疗肿瘤:结构-活性关系(SARs)研究进展
IF 5.9 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-11-13 DOI: 10.1016/j.ejmech.2025.118372
Yinglong Li, Yanyun Hong, Jianye Zhang, Shan Xu
As a paracaspase, mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) plays a crucial role in the NF-κB signaling pathway. It typically exerts its function by forming a ternary complex with CARMA and BCL. The structure of MALT1 endows it with dual roles as a protease and a scaffold protein, both of which are of great significance for the regulation of the signaling pathway. Activated B-cell like diffuse large B-cell lymphoma (ABC-DLBCL) is characterized by high complication rates and mortality. Studies have demonstrated that its pathogenesis relies on the NF-κB pathway mediated by the CBM complex (CARMA-Bcl10-MALT1 complex). As the only human cysteine-aspartic acid protease (caspase-like protease) in the CBM complex, MALT1 has thus become a key target, making the research on MALT1 inhibitors of substantial importance. This review provides an overview of the research progress and structure-activity relationships (SARs) of MALT1 inhibitors, including covalent inhibitors, allosteric inhibitors, PROTACs (Proteolysis-Targeting Chimeras), and activity-based probes. Additionally, it discusses the future opportunities and challenges in this field, aiming to provide insights for the future development of MALT1-targeted drugs.
粘膜相关淋巴组织淋巴瘤易位蛋白1 (MALT1)作为一种副半乳糖酶,在NF-κB信号通路中起着至关重要的作用。它通常通过与CARMA和BCL形成三元配合物来发挥作用。MALT1的结构使其具有蛋白酶和支架蛋白的双重作用,对信号通路的调控具有重要意义。活化b细胞样弥漫性大b细胞淋巴瘤(ABC-DLBCL)的特点是高并发症发生率和死亡率。研究表明其发病机制依赖于CBM复合物介导的NF-κB通路。作为CBM复合体(CARMA-Bcl10-MALT1复合体)中唯一的人半胱氨酸-天门氨酸蛋白酶(caspase样蛋白酶),MALT1因此成为关键靶点,因此对MALT1抑制剂的研究具有重要意义。本文综述了MALT1抑制剂的研究进展和构效关系,包括共价抑制剂、变构抑制剂、Proteolysis-Targeting Chimeras (Proteolysis-Targeting Chimeras)和基于活性的探针。并讨论了该领域未来的机遇和挑战,旨在为malt1靶向药物的未来发展提供见解。
{"title":"Targeting paracaspase1 (MALT1) for cancer therapy: Updated progress and study on structure-activity relationships (SARs)","authors":"Yinglong Li,&nbsp;Yanyun Hong,&nbsp;Jianye Zhang,&nbsp;Shan Xu","doi":"10.1016/j.ejmech.2025.118372","DOIUrl":"10.1016/j.ejmech.2025.118372","url":null,"abstract":"<div><div>As a paracaspase, mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) plays a crucial role in the NF-κB signaling pathway. It typically exerts its function by forming a ternary complex with CARMA and BCL. The structure of MALT1 endows it with dual roles as a protease and a scaffold protein, both of which are of great significance for the regulation of the signaling pathway. Activated B-cell like diffuse large B-cell lymphoma (ABC-DLBCL) is characterized by high complication rates and mortality. Studies have demonstrated that its pathogenesis relies on the NF-κB pathway mediated by the CBM complex (CARMA-Bcl10-MALT1 complex). As the only human cysteine-aspartic acid protease (caspase-like protease) in the CBM complex, MALT1 has thus become a key target, making the research on MALT1 inhibitors of substantial importance. This review provides an overview of the research progress and structure-activity relationships (SARs) of MALT1 inhibitors, including covalent inhibitors, allosteric inhibitors, PROTACs (Proteolysis-Targeting Chimeras), and activity-based probes. Additionally, it discusses the future opportunities and challenges in this field, aiming to provide insights for the future development of MALT1-targeted drugs.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"302 ","pages":"Article 118372"},"PeriodicalIF":5.9,"publicationDate":"2025-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145509048","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
Pyrazolo[1,5-a]pyrimidine scaffold-based small molecules: From bench to FDA-approved TRK kinase inhibitors (Part 1) Pyrazolo[1,5-a]嘧啶支架小分子:从实验室到fda批准的TRK激酶抑制剂(上)
IF 5.9 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-11-13 DOI: 10.1016/j.ejmech.2025.118348
Mustafa A. Al-Qadhi , Tawfeek A.A. Yahya , Anas A. Alqadhi , Reem K. Arafa
The pyrazolo[1,5-a]pyrimidine scaffold has emerged as a privileged structure in medicinal chemistry, particularly in designing kinase-targeted therapeutics, such as tropomyosin receptor kinase (TRK) inhibitors, for targeted cancer therapy. This review provides a critical analysis of small molecules bearing this heterocyclic motif, focusing on their chemical evolution, structure-activity relationship (SAR)- driven optimization, mechanistic insights into TRK inhibition, and the translational journey of these compounds from preclinical studies to FDA-approved drugs, such as Larotrectinib. The review highlights the strategic chemical modifications that improved selectivity, pharmacokinetics, and safety profiles of TRK inhibitors. Additionally, it outlines the evolution of kinase inhibitor design strategies leveraging this scaffold and discusses current challenges and future directions in optimizing these compounds for broader clinical applications. This work aims to serve as a foundation for ongoing drug discovery efforts utilizing the pyrazolo[1,5-a]pyrimidine core in oncogenic kinase targeting. Collectively, this work underscores the significance of the pyrazolo[1,5-a]pyrimidine scaffold as a privileged chemotype and sets the stage for the rational development of next-generation TRK inhibitors with enhanced clinical utility.
吡唑啉[1,5-a]嘧啶支架在药物化学中已经成为一种特殊的结构,特别是在设计激酶靶向治疗中,如原肌球蛋白受体激酶(TRK)抑制剂,用于靶向癌症治疗。这篇综述提供了具有这种杂环基序的小分子的关键分析,重点是它们的化学进化,结构-活性关系(SAR)驱动的优化,TRK抑制的机制见解,以及这些化合物从临床前研究到fda批准的药物(如larorectinib)的转化过程。这篇综述强调了战略性的化学修饰可以提高TRK抑制剂的选择性、药代动力学和安全性。此外,它概述了利用这种支架的激酶抑制剂设计策略的演变,并讨论了优化这些化合物以实现更广泛临床应用的当前挑战和未来方向。这项工作旨在为利用pyrazolo[1,5-a]嘧啶核心靶向致癌激酶的药物发现工作奠定基础。总的来说,这项工作强调了吡唑[1,5-a]嘧啶支架作为一种特殊化学型的重要性,并为合理开发具有增强临床实用性的下一代TRK抑制剂奠定了基础。
{"title":"Pyrazolo[1,5-a]pyrimidine scaffold-based small molecules: From bench to FDA-approved TRK kinase inhibitors (Part 1)","authors":"Mustafa A. Al-Qadhi ,&nbsp;Tawfeek A.A. Yahya ,&nbsp;Anas A. Alqadhi ,&nbsp;Reem K. Arafa","doi":"10.1016/j.ejmech.2025.118348","DOIUrl":"10.1016/j.ejmech.2025.118348","url":null,"abstract":"<div><div>The pyrazolo[1,5-a]pyrimidine scaffold has emerged as a privileged structure in medicinal chemistry, particularly in designing kinase-targeted therapeutics, such as tropomyosin receptor kinase (TRK) inhibitors, for targeted cancer therapy. This review provides a critical analysis of small molecules bearing this heterocyclic motif, focusing on their chemical evolution, structure-activity relationship (SAR)- driven optimization, mechanistic insights into TRK inhibition, and the translational journey of these compounds from preclinical studies to FDA-approved drugs, such as Larotrectinib. The review highlights the strategic chemical modifications that improved selectivity, pharmacokinetics, and safety profiles of TRK inhibitors. Additionally, it outlines the evolution of kinase inhibitor design strategies leveraging this scaffold and discusses current challenges and future directions in optimizing these compounds for broader clinical applications. This work aims to serve as a foundation for ongoing drug discovery efforts utilizing the pyrazolo[1,5-a]pyrimidine core in oncogenic kinase targeting. Collectively, this work underscores the significance of the pyrazolo[1,5-a]pyrimidine scaffold as a privileged chemotype and sets the stage for the rational development of next-generation TRK inhibitors with enhanced clinical utility.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"302 ","pages":"Article 118348"},"PeriodicalIF":5.9,"publicationDate":"2025-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145498837","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
Natural alkaloids with therapeutic potential against Alzheimer's disease through cholinesterase inhibition 天然生物碱通过抑制胆碱酯酶对阿尔茨海默病具有治疗潜力
IF 5.9 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-11-12 DOI: 10.1016/j.ejmech.2025.118371
Umam Khan , Moazzam Ahmad , Mariam Tuba , Raabia Naaz , Fayeqa Qayum , Shahnaaz Khatoon , Sanobar , Shakir Ahamad , Mohammad Saquib , Mohd Kamil Hussain
Alzheimer's disease (AD) is a progressive neurodegenerative disorder in which the decline of cholinergic neurotransmission plays a central role in cognitive impairment. Cholinesterase inhibition remains an established therapeutic approach to enhance acetylcholine levels and provide symptomatic relief. Natural alkaloids have long served as an important source of cholinesterase inhibitors, offering diverse molecular scaffolds, stereochemical complexity, and strong biological relevance. This review provides a comprehensive overview of natural alkaloids reported as acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibitors, highlighting their structural diversity, mechanisms of inhibition, and structure–activity relationships. In addition, the review presents a brief analysis on the emerging pseudo-natural product (PNP) framework as a complementary strategy for designing new alkaloid-inspired potential inhibitors that combine nature-derived functionality with synthetic innovation. Overall, the manuscript underscores the enduring significance of alkaloids in cholinesterase inhibitor discovery and their potential in developing next-generation therapeutics for AD.
阿尔茨海默病(AD)是一种进行性神经退行性疾病,其中胆碱能神经传递的下降在认知障碍中起着核心作用。胆碱酯酶抑制仍然是一种既定的治疗方法,以提高乙酰胆碱水平和提供症状缓解。天然生物碱长期以来一直是胆碱酯酶抑制剂的重要来源,具有分子支架的多样性、立体化学的复杂性和很强的生物学相关性。本文综述了作为乙酰胆碱酯酶(AChE)和丁基胆碱酯酶(BChE)抑制剂的天然生物碱,重点介绍了它们的结构多样性、抑制机制和构效关系。此外,该综述简要分析了新兴的伪天然产物(PNP)框架,作为设计新的生物碱启发的潜在抑制剂的补充策略,将自然衍生的功能与合成创新相结合。总的来说,该论文强调了生物碱在发现胆碱酯酶抑制剂中的持久意义,以及它们在开发下一代阿尔茨海默病治疗方法方面的潜力。
{"title":"Natural alkaloids with therapeutic potential against Alzheimer's disease through cholinesterase inhibition","authors":"Umam Khan ,&nbsp;Moazzam Ahmad ,&nbsp;Mariam Tuba ,&nbsp;Raabia Naaz ,&nbsp;Fayeqa Qayum ,&nbsp;Shahnaaz Khatoon ,&nbsp;Sanobar ,&nbsp;Shakir Ahamad ,&nbsp;Mohammad Saquib ,&nbsp;Mohd Kamil Hussain","doi":"10.1016/j.ejmech.2025.118371","DOIUrl":"10.1016/j.ejmech.2025.118371","url":null,"abstract":"<div><div>Alzheimer's disease (AD) is a progressive neurodegenerative disorder in which the decline of cholinergic neurotransmission plays a central role in cognitive impairment. Cholinesterase inhibition remains an established therapeutic approach to enhance acetylcholine levels and provide symptomatic relief. Natural alkaloids have long served as an important source of cholinesterase inhibitors, offering diverse molecular scaffolds, stereochemical complexity, and strong biological relevance. This review provides a comprehensive overview of natural alkaloids reported as acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibitors, highlighting their structural diversity, mechanisms of inhibition, and structure–activity relationships. In addition, the review presents a brief analysis on the emerging pseudo-natural product (PNP) framework as a complementary strategy for designing new alkaloid-inspired potential inhibitors that combine nature-derived functionality with synthetic innovation. Overall, the manuscript underscores the enduring significance of alkaloids in cholinesterase inhibitor discovery and their potential in developing next-generation therapeutics for AD.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"302 ","pages":"Article 118371"},"PeriodicalIF":5.9,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145498228","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
TREM2 activation by first-in-class direct small molecule agonists: DEL screening, optimization, biophysical validation, and functional characterization 一流的直接小分子激动剂激活TREM2: DEL筛选,优化,生物物理验证和功能表征
IF 5.9 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-11-12 DOI: 10.1016/j.ejmech.2025.118358
Hossam Nada , Shaoren Yuan , Farida El Gaamouch , Sungwoo Cho , Katarzyna Kuncewicz , Laura Calvo-Barreiro , Moustafa T. Gabr
Triggering receptor expressed on myeloid cells 2 (TREM2) is a key regulator of microglial function, and its loss-of-function variants are linked to Alzheimer's disease (AD) and neurodegenerative disorders. While TREM2 activation is a promising therapeutic strategy, no small molecule agonists acting via direct TREM2 binding have been reported to date. Here, we describe the discovery of first-in-class, direct small molecule TREM2 agonists identified through DNA-encoded library (DEL) screening. The DEL hit (4a) demonstrated TREM2 binding affinity, as validated by three biophysical screening platforms (TRIC, MST, and SPR), induced Syk phosphorylation, luciferase assay and enhanced microglial phagocytosis. Pre-liminary optimization yielded 4i, which maintained TREM2 engagement with improved selectivity over TREM1 and no cytotoxicity. Molecular dynamics simulations predicted that 4a stabilizes a transient binding pocket on TREM2, indicating the possibility of a novel mechanism for receptor activation. These findings provide the first proof-of-concept for direct pharmacological TREM2 agonism, offering a foundation for developing therapeutics against AD and related disorders.
髓样细胞上表达的触发受体2 (TREM2)是小胶质细胞功能的关键调节因子,其功能丧失变体与阿尔茨海默病(AD)和神经退行性疾病有关。虽然激活TREM2是一种很有前景的治疗策略,但迄今为止还没有报道过通过直接结合TREM2起作用的小分子激动剂。在这里,我们描述了通过dna编码文库(DEL)筛选鉴定的一流的直接小分子TREM2激动剂的发现。通过三种生物物理筛选平台(TRIC、MST和SPR)验证,DEL hit (4a)显示出TREM2结合亲和力,诱导Syk磷酸化、荧光素酶测定和增强小胶质细胞吞噬。初步优化得到了4i,它保持了TREM2的结合,比TREM1具有更高的选择性,并且没有细胞毒性。分子动力学模拟预测4a稳定了TREM2上的瞬时结合袋,这表明可能存在一种新的受体激活机制。这些发现首次提供了直接药理TREM2激动作用的概念证明,为开发针对AD和相关疾病的治疗方法提供了基础。
{"title":"TREM2 activation by first-in-class direct small molecule agonists: DEL screening, optimization, biophysical validation, and functional characterization","authors":"Hossam Nada ,&nbsp;Shaoren Yuan ,&nbsp;Farida El Gaamouch ,&nbsp;Sungwoo Cho ,&nbsp;Katarzyna Kuncewicz ,&nbsp;Laura Calvo-Barreiro ,&nbsp;Moustafa T. Gabr","doi":"10.1016/j.ejmech.2025.118358","DOIUrl":"10.1016/j.ejmech.2025.118358","url":null,"abstract":"<div><div>Triggering receptor expressed on myeloid cells 2 (TREM2) is a key regulator of microglial function, and its loss-of-function variants are linked to Alzheimer's disease (AD) and neurodegenerative disorders. While TREM2 activation is a promising therapeutic strategy, no small molecule agonists acting via direct TREM2 binding have been reported to date. Here, we describe the discovery of first-in-class, direct small molecule TREM2 agonists identified through DNA-encoded library (DEL) screening. The DEL hit (4a) demonstrated TREM2 binding affinity, as validated by three biophysical screening platforms (TRIC, MST, and SPR), induced Syk phosphorylation, luciferase assay and enhanced microglial phagocytosis. Pre-liminary optimization yielded 4i, which maintained TREM2 engagement with improved selectivity over TREM1 and no cytotoxicity. Molecular dynamics simulations predicted that 4a stabilizes a transient binding pocket on TREM2, indicating the possibility of a novel mechanism for receptor activation. These findings provide the first proof-of-concept for direct pharmacological TREM2 agonism, offering a foundation for developing therapeutics against AD and related disorders.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"302 ","pages":"Article 118358"},"PeriodicalIF":5.9,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145498833","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 pyrimidine-based KRAS-G12C inhibitors with potent anti-NSCLC activity via virtual screening and structure optimization 通过虚拟筛选和结构优化发现具有有效抗nsclc活性的新型嘧啶基KRAS-G12C抑制剂
IF 5.9 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-11-12 DOI: 10.1016/j.ejmech.2025.118354
Jian-Tao Shi , Su-Juan Hou , Cheng-Long Xu , Lei Cheng , Feng-Ya Ge , Xiu-Juan Liu , Yi-Ru Wang , Xi-Bo Wang , Yao-Sheng Zhang , Junmin Zhang , Shi-Wu Chen
KRAS-G12C is a validated therapeutic target for KRAS-mutant cancers. However, current KRAS-G12C inhibitors face limitations due to structural homogeneity and the emergence of drug resistance. To address this, we employed virtual screening to identify novel pyrimidine-based KRAS-G12C inhibitors, followed by rational structural optimization. Among the optimized compounds, KD36 significantly inhibited the proliferation of KRAS-G12C mutant NSCLC cell lines (NCI–H23 and NCI–H358) in a dose-dependent manner. Mechanistically, KD36 suppressed the phosphorylation of KRAS downstream effectors ERK and AKT. Importantly, KD36 induced intrinsic (mitochondrial) apoptosis in NCI–H23 cells. Critically, in an NCI–H358 xenograft mouse model, KD36 (at 30 mg/kg) exhibited significant tumor growth inhibition with 54.6 % tumor growth inhibition (TGI), without apparent systemic toxicity. These findings establish KD36 as a promising, structurally novel pyrimidine-based KRAS-G12C inhibitor lead compound with potent anti-tumor efficacy against KRAS-G12C mutant NSCLC, demonstrating the success of our virtual screening and optimization strategy in overcoming scaffold limitations.
KRAS-G12C是kras突变型癌症的有效治疗靶点。然而,目前的KRAS-G12C抑制剂由于结构同质性和耐药的出现而面临局限性。为了解决这个问题,我们采用虚拟筛选方法鉴定了新型嘧啶基KRAS-G12C抑制剂,然后进行了合理的结构优化。在优化的化合物中,KD36显著抑制KRAS-G12C突变体NSCLC细胞株(NCI-H23和NCI-H358)的增殖,且呈剂量依赖性。在机制上,KD36抑制KRAS下游效应物ERK和AKT的磷酸化。重要的是,KD36诱导NCI-H23细胞的内在(线粒体)凋亡。关键的是,在NCI-H358异种移植小鼠模型中,KD36 (30 mg/kg)表现出显著的肿瘤生长抑制作用,肿瘤生长抑制率为54.6% (TGI),无明显的全身毒性。这些发现表明KD36是一种有前景的、结构新颖的基于嘧啶的KRAS-G12C抑制剂先导化合物,对KRAS-G12C突变体NSCLC具有强大的抗肿瘤功效,表明我们的虚拟筛选和优化策略在克服支架限制方面取得了成功。
{"title":"Discovery of novel pyrimidine-based KRAS-G12C inhibitors with potent anti-NSCLC activity via virtual screening and structure optimization","authors":"Jian-Tao Shi ,&nbsp;Su-Juan Hou ,&nbsp;Cheng-Long Xu ,&nbsp;Lei Cheng ,&nbsp;Feng-Ya Ge ,&nbsp;Xiu-Juan Liu ,&nbsp;Yi-Ru Wang ,&nbsp;Xi-Bo Wang ,&nbsp;Yao-Sheng Zhang ,&nbsp;Junmin Zhang ,&nbsp;Shi-Wu Chen","doi":"10.1016/j.ejmech.2025.118354","DOIUrl":"10.1016/j.ejmech.2025.118354","url":null,"abstract":"<div><div>KRAS-G12C is a validated therapeutic target for KRAS-mutant cancers. However, current KRAS-G12C inhibitors face limitations due to structural homogeneity and the emergence of drug resistance. To address this, we employed virtual screening to identify novel pyrimidine-based KRAS-G12C inhibitors, followed by rational structural optimization. Among the optimized compounds, <strong>KD36</strong> significantly inhibited the proliferation of KRAS-G12C mutant NSCLC cell lines (NCI–H23 and NCI–H358) in a dose-dependent manner. Mechanistically, <strong>KD36</strong> suppressed the phosphorylation of KRAS downstream effectors ERK and AKT. Importantly, <strong>KD36</strong> induced intrinsic (mitochondrial) apoptosis in NCI–H23 cells. Critically, in an NCI–H358 xenograft mouse model, <strong>KD36</strong> (at 30 mg/kg) exhibited significant tumor growth inhibition with 54.6 % tumor growth inhibition (TGI), without apparent systemic toxicity. These findings establish <strong>KD36</strong> as a promising, structurally novel pyrimidine-based KRAS-G12C inhibitor lead compound with potent anti-tumor efficacy against KRAS-G12C mutant NSCLC, demonstrating the success of our virtual screening and optimization strategy in overcoming scaffold limitations.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"302 ","pages":"Article 118354"},"PeriodicalIF":5.9,"publicationDate":"2025-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145498841","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
Beyond traditional antibacterial agents: Novel approaches to combat resistant pathogens 超越传统抗菌剂:对抗耐药病原体的新方法
IF 5.9 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-11-10 DOI: 10.1016/j.ejmech.2025.118362
Wei Zong , Shengjie Xie , Hongtao Chu , Shuang Han , Xunan Zhang
As bacterial infections caused by antibiotic-resistant strains become increasingly prevalent, traditional antibacterial therapies face mounting challenges. These resilient pathogens not only complicate the treatment of common infections but also undermine the efficacy of therapies for major diseases. This growing threat underscores the urgent need for innovative therapeutic strategies. In recent years, the combinatorial use of antibacterial agents has emerged as a promising approach to enhance efficacy and combat resistant bacteria. This review first provides an overview of antibacterial classifications and their mechanisms of action against bacterial infections. It then explores two combined treatment strategies: antibacterial–antibacterial combinations and antibacterial–non-antibacterial pairings, alongside a drug delivery technology: antibacterial-loaded liposomes. Furthermore, we highlight emerging frontiers in antimicrobial strategies, including CRISPR-Cas technologies, AI-driven discovery platforms, nanomaterials beyond liposomes, microbiota-based therapies, and immunotherapeutic approaches. Finally, we offer a forward-looking perspective on the challenges and opportunities shaping the future of antibacterial development in the biomedical field.
随着耐药菌株引起的细菌感染日益普遍,传统的抗菌疗法面临越来越大的挑战。这些具有弹性的病原体不仅使普通感染的治疗复杂化,而且还破坏了对主要疾病的治疗效果。这一日益严重的威胁凸显了对创新治疗策略的迫切需要。近年来,联合使用抗菌药物已成为提高疗效和对抗耐药细菌的一种有前途的方法。本文首先综述了抗菌药物的分类及其抗细菌感染的作用机制。然后探讨了两种联合治疗策略:抗菌-抗菌组合和抗菌-非抗菌配对,以及一种药物输送技术:抗菌负载脂质体。此外,我们还重点介绍了抗菌策略的新兴前沿,包括CRISPR-Cas技术、人工智能驱动的发现平台、脂质体以外的纳米材料、基于微生物群的疗法和免疫治疗方法。最后,我们对未来生物医学领域抗菌药物发展的挑战和机遇进行了前瞻性的展望。
{"title":"Beyond traditional antibacterial agents: Novel approaches to combat resistant pathogens","authors":"Wei Zong ,&nbsp;Shengjie Xie ,&nbsp;Hongtao Chu ,&nbsp;Shuang Han ,&nbsp;Xunan Zhang","doi":"10.1016/j.ejmech.2025.118362","DOIUrl":"10.1016/j.ejmech.2025.118362","url":null,"abstract":"<div><div>As bacterial infections caused by antibiotic-resistant strains become increasingly prevalent, traditional antibacterial therapies face mounting challenges. These resilient pathogens not only complicate the treatment of common infections but also undermine the efficacy of therapies for major diseases. This growing threat underscores the urgent need for innovative therapeutic strategies. In recent years, the combinatorial use of antibacterial agents has emerged as a promising approach to enhance efficacy and combat resistant bacteria. This review first provides an overview of antibacterial classifications and their mechanisms of action against bacterial infections. It then explores two combined treatment strategies: antibacterial–antibacterial combinations and antibacterial–non-antibacterial pairings, alongside a drug delivery technology: antibacterial-loaded liposomes. Furthermore, we highlight emerging frontiers in antimicrobial strategies, including CRISPR-Cas technologies, AI-driven discovery platforms, nanomaterials beyond liposomes, microbiota-based therapies, and immunotherapeutic approaches. Finally, we offer a forward-looking perspective on the challenges and opportunities shaping the future of antibacterial development in the biomedical field.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"302 ","pages":"Article 118362"},"PeriodicalIF":5.9,"publicationDate":"2025-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145485399","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
Brain-targeted Jedi1 and Jedi2 derivatives as potent enhancers of Piezo1 ion channel activity 脑靶向Jedi1和Jedi2衍生物作为Piezo1离子通道活性的有效增强剂
IF 5.9 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-11-10 DOI: 10.1016/j.ejmech.2025.118359
Landry Anamea , Adéla Králová , Janne Tampio , Anastasia Shakirzyanova , Milka Hänninen , Rashid Giniatullin , Aaro J. Jalkanen , Tarja Malm , Kristiina M. Huttunen
Mechanosensitive Piezo1 channel is highly expressed in the eukaryotic central nervous system and involved in many important physiological functions that maintain cell homeostasis. This membrane ion channel is opened by different forms of mechanical and chemical stimulation. However, pharmacological tools for Piezo1 channel manipulations and in vivo application are limited. In the present study, to address this need, 12 L-type amino acid transporter 1 (LAT1)-utilizing brain-targeted conjugates of Piezo1 activators, Jedi1 and Jedi2, were synthesized and evaluated for their physicochemical and pharmaceutical properties. As a result, the novel compounds were more effectively transported via LAT1 into mouse BV2 microglia cells. Moreover, compared to their parent compounds, they had a higher ability to activate Piezo1 measured by a fluorescent live calcium imaging assay. Furthermore, the pharmacokinetic study revealed that the compounds were delivered into the mouse brain and were distributed to other peripheral tissues to a lesser extent than their parent compound, Jedi2. Therefore, these novel conjugates can be considered as potential compounds to achieve brain- and microglia-targeted delivery and potential neuroprotective effects via Piezo1 ion channel activation, being beneficial in the treatment of neurodegenerative disorders.
机械敏感的Piezo1通道在真核生物中枢神经系统中高度表达,参与维持细胞内稳态的许多重要生理功能。这种膜离子通道通过不同形式的机械和化学刺激而打开。然而,用于Piezo1通道操作和体内应用的药理学工具是有限的。在本研究中,为了满足这一需求,我们合成了12个l型氨基酸转运蛋白1 (LAT1)-利用Piezo1激活剂的脑靶向偶联物Jedi1和Jedi2,并对其物理化学和药物性质进行了评估。结果,新化合物更有效地通过LAT1转运到小鼠BV2小胶质细胞中。此外,与它们的母体化合物相比,通过荧光活钙成像测定,它们具有更高的激活Piezo1的能力。此外,药代动力学研究表明,与母体化合物Jedi2相比,这些化合物进入小鼠大脑并分布到其他外周组织的程度较低。因此,这些新的缀合物可以被认为是潜在的化合物,通过Piezo1离子通道激活实现脑和小胶质细胞靶向递送和潜在的神经保护作用,有利于神经退行性疾病的治疗。
{"title":"Brain-targeted Jedi1 and Jedi2 derivatives as potent enhancers of Piezo1 ion channel activity","authors":"Landry Anamea ,&nbsp;Adéla Králová ,&nbsp;Janne Tampio ,&nbsp;Anastasia Shakirzyanova ,&nbsp;Milka Hänninen ,&nbsp;Rashid Giniatullin ,&nbsp;Aaro J. Jalkanen ,&nbsp;Tarja Malm ,&nbsp;Kristiina M. Huttunen","doi":"10.1016/j.ejmech.2025.118359","DOIUrl":"10.1016/j.ejmech.2025.118359","url":null,"abstract":"<div><div>Mechanosensitive Piezo1 channel is highly expressed in the eukaryotic central nervous system and involved in many important physiological functions that maintain cell homeostasis. This membrane ion channel is opened by different forms of mechanical and chemical stimulation. However, pharmacological tools for Piezo1 channel manipulations and <em>in vivo</em> application are limited. In the present study, to address this need, 12 L-type amino acid transporter 1 (LAT1)-utilizing brain-targeted conjugates of Piezo1 activators, Jedi1 and Jedi2, were synthesized and evaluated for their physicochemical and pharmaceutical properties. As a result, the novel compounds were more effectively transported via LAT1 into mouse BV2 microglia cells. Moreover, compared to their parent compounds, they had a higher ability to activate Piezo1 measured by a fluorescent live calcium imaging assay. Furthermore, the pharmacokinetic study revealed that the compounds were delivered into the mouse brain and were distributed to other peripheral tissues to a lesser extent than their parent compound, Jedi2. Therefore, these novel conjugates can be considered as potential compounds to achieve brain- and microglia-targeted delivery and potential neuroprotective effects via Piezo1 ion channel activation, being beneficial in the treatment of neurodegenerative disorders.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"302 ","pages":"Article 118359"},"PeriodicalIF":5.9,"publicationDate":"2025-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145485278","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
Synthesis and structure-activity relationship study of novel quinazolin-4(3H)-one derivatives as Toll-like receptor 7 and 8 agonists with immunomodulatory activity 新型toll样受体7和8激动剂喹唑啉-4(3H)- 1衍生物的合成及构效关系研究
IF 5.9 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-11-10 DOI: 10.1016/j.ejmech.2025.118360
Nika Strašek Benedik , David Lukić , Tjaša Slokan , Ana Dolšak , Urban Švajger , Simona Kranjc Brezar , Valerij Talagayev , Gerhard Wolber , Maja Čemažar , Izidor Sosič , Matej Sova
Toll-like receptors (TLRs) 7 and 8 are intracellular pattern recognition receptors that play a crucial role in the innate immune system, making them promising targets for the treatment of viral infections, autoimmune diseases and cancer. In this study, we present the synthesis and biological evaluation of quinazolin-4(3H)-one derivatives as a new class of dual TLR7/8 agonists. In a comprehensive structure-activity relationship (SAR) study, different substitutions on the quinazoline ring and modifications of the aliphatic side chain were investigated. Several compounds showed significantly improved potency compared to the original hit compound, with EC50 values in the nanomolar and low micromolar range for TLR7 and TLR8, respectively. The most potent compounds significantly increased the secretion of the proinflammatory cytokines TNF-α, IL-1β, IL-8 and interferon γ in peripheral blood mononuclear cells (PBMCs). In addition, increased secretion of TNF-α and upregulated CD86 expression in dendritic cells were also observed, indicating their immunomodulatory properties. Notably, the most potent compound 69 significantly suppressed tumor growth in vivo in the CT26 mouse tumor model after intratumoral administration. These results highlight the potential of quinazolinone-based compounds as promising candidates for further development of new immunomodulatory agents targeting TLR7 and TLR8.
toll样受体(TLRs) 7和8是细胞内模式识别受体,在先天免疫系统中起着至关重要的作用,使它们成为治疗病毒感染、自身免疫性疾病和癌症的有希望的靶点。本文报道了一类新型TLR7/8双受体激动剂喹唑啉-4(3H)- 1衍生物的合成及生物学评价。在一项全面的构效关系(SAR)研究中,研究了喹唑啉环上的不同取代和脂肪侧链的修饰。TLR7和TLR8的EC50值分别在纳摩尔和低微摩尔范围内。最有效的化合物显著增加外周血单核细胞(PBMCs)促炎细胞因子TNF-α、IL-1β、IL-8和干扰素γ的分泌。此外,树突状细胞中TNF-α分泌增加,CD86表达上调,表明其免疫调节特性。值得注意的是,在肿瘤内给药后,最有效的化合物69在体内显著抑制CT26小鼠肿瘤模型中的肿瘤生长。这些结果突出了喹唑啉酮类化合物作为进一步开发靶向TLR7和TLR8的新型免疫调节剂的潜力。
{"title":"Synthesis and structure-activity relationship study of novel quinazolin-4(3H)-one derivatives as Toll-like receptor 7 and 8 agonists with immunomodulatory activity","authors":"Nika Strašek Benedik ,&nbsp;David Lukić ,&nbsp;Tjaša Slokan ,&nbsp;Ana Dolšak ,&nbsp;Urban Švajger ,&nbsp;Simona Kranjc Brezar ,&nbsp;Valerij Talagayev ,&nbsp;Gerhard Wolber ,&nbsp;Maja Čemažar ,&nbsp;Izidor Sosič ,&nbsp;Matej Sova","doi":"10.1016/j.ejmech.2025.118360","DOIUrl":"10.1016/j.ejmech.2025.118360","url":null,"abstract":"<div><div>Toll-like receptors (TLRs) 7 and 8 are intracellular pattern recognition receptors that play a crucial role in the innate immune system, making them promising targets for the treatment of viral infections, autoimmune diseases and cancer. In this study, we present the synthesis and biological evaluation of quinazolin-4(3<em>H</em>)-one derivatives as a new class of dual TLR7/8 agonists. In a comprehensive structure-activity relationship (SAR) study, different substitutions on the quinazoline ring and modifications of the aliphatic side chain were investigated. Several compounds showed significantly improved potency compared to the original hit compound, with EC<sub>50</sub> values in the nanomolar and low micromolar range for TLR7 and TLR8, respectively. The most potent compounds significantly increased the secretion of the proinflammatory cytokines TNF-α, IL-1β, IL-8 and interferon γ in peripheral blood mononuclear cells (PBMCs). In addition, increased secretion of TNF-α and upregulated CD86 expression in dendritic cells were also observed, indicating their immunomodulatory properties. Notably, the most potent compound <strong>69</strong> significantly suppressed tumor growth <em>in vivo</em> in the CT26 mouse tumor model after intratumoral administration. These results highlight the potential of quinazolinone-based compounds as promising candidates for further development of new immunomodulatory agents targeting TLR7 and TLR8.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"302 ","pages":"Article 118360"},"PeriodicalIF":5.9,"publicationDate":"2025-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145478019","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
Identification of EEF1A2 as a potential therapy target of osteosarcoma using novel compound 8e 利用新型化合物8e鉴定EEF1A2作为骨肉瘤潜在治疗靶点
IF 5.9 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-11-08 DOI: 10.1016/j.ejmech.2025.118355
Jian Xue , Meng Li , Ying Wang , Donghui Sun , Shilong Hao , Zhuochao Liu , Weibin Zhang , Lei Li , Tong Zhu , Shunying Liu
Osteosarcoma (OS) is a rare malignant tumor and has the second-highest mortality rate of malignant tumors in children. Due to its unclear pathogenesis and therapeutic targets, there has been no significant progress in the targeted therapy of OS in the past 50 years. Therefore, it is critically important to develop new drug targets for OS. In this study, a covalent molecule library consisting of 61 small molecules was constructed based on our previous research. Phenotypic screening revealed that small molecule 8e effectively inhibited the proliferation of OS 143B cells, with an IC50 value of 0.73 μM. Compound 8e also showed good antitumor effects and low toxicity in a xenograft model (30.1 % inhibition of OS growth in BALB/c nude mice). Using compound 9a as an efficient activity-based protein profiling (ABPP) probe, eukaryotic protein elongation factor 1 alpha 2 (EEF1A2) was then enriched and conveniently identified as a potential target. The potential target was validated by pull-down assay, cellular thermal shift assay (CETSA), mass spectrometry analysis, molecular docking, and in vitro and in vivo functional studies. Mechanistic studies suggest that compound 8e-induced 143B cell apoptotic is mediated by EEF1A2 inhibition of the AKT signaling pathway and EEF1A2 serves as a potential candidate for targeted OS therapy.
骨肉瘤(OS)是一种罕见的恶性肿瘤,是儿童恶性肿瘤中死亡率第二高的肿瘤。由于其发病机制和治疗靶点不明确,在过去的50年里,OS的靶向治疗没有取得重大进展。因此,开发新的OS药物靶点至关重要。本研究在前期研究的基础上,构建了由61个小分子组成的共价分子文库。表型筛选显示,小分子8e能有效抑制OS 143B细胞的增殖,IC50值为0.73 μM。化合物8e在异种移植模型中也显示出良好的抗肿瘤作用和低毒性(抑制BALB/c裸鼠OS生长30.1%)。利用化合物9a作为高效的基于活性的蛋白谱分析(ABPP)探针,对真核蛋白延伸因子1 α 2 (EEF1A2)进行富集并方便地鉴定为潜在靶标。通过下拉实验、细胞热移实验(CETSA)、质谱分析、分子对接以及体外和体内功能研究对潜在靶点进行了验证。机制研究表明,化合物8e诱导的143B细胞凋亡是通过EEF1A2抑制AKT信号通路介导的,EEF1A2可作为靶向OS治疗的潜在候选药物。
{"title":"Identification of EEF1A2 as a potential therapy target of osteosarcoma using novel compound 8e","authors":"Jian Xue ,&nbsp;Meng Li ,&nbsp;Ying Wang ,&nbsp;Donghui Sun ,&nbsp;Shilong Hao ,&nbsp;Zhuochao Liu ,&nbsp;Weibin Zhang ,&nbsp;Lei Li ,&nbsp;Tong Zhu ,&nbsp;Shunying Liu","doi":"10.1016/j.ejmech.2025.118355","DOIUrl":"10.1016/j.ejmech.2025.118355","url":null,"abstract":"<div><div>Osteosarcoma (OS) is a rare malignant tumor and has the second-highest mortality rate of malignant tumors in children. Due to its unclear pathogenesis and therapeutic targets, there has been no significant progress in the targeted therapy of OS in the past 50 years. Therefore, it is critically important to develop new drug targets for OS. In this study, a covalent molecule library consisting of 61 small molecules was constructed based on our previous research. Phenotypic screening revealed that small molecule <strong>8e</strong> effectively inhibited the proliferation of OS 143B cells, with an IC<sub>50</sub> value of 0.73 μM. Compound <strong>8e</strong> also showed good antitumor effects and low toxicity in a xenograft model (30.1 % inhibition of OS growth in BALB/c nude mice). Using compound <strong>9a</strong> as an efficient activity-based protein profiling (ABPP) probe, eukaryotic protein elongation factor 1 alpha 2 (EEF1A2) was then enriched and conveniently identified as a potential target. The potential target was validated by pull-down assay, cellular thermal shift assay (CETSA), mass spectrometry analysis, molecular docking, and in vitro and in vivo functional studies. Mechanistic studies suggest that compound <strong>8e</strong>-induced 143B cell apoptotic is mediated by EEF1A2 inhibition of the AKT signaling pathway and EEF1A2 serves as a potential candidate for targeted OS therapy.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"302 ","pages":"Article 118355"},"PeriodicalIF":5.9,"publicationDate":"2025-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145462276","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
Novel imidazo[1,2-a]pyridine-based tubulin polymerization inhibitors: Structure-activity relationships and anti-tumor immune potentiation 新型咪唑[1,2-a]吡啶基微管蛋白聚合抑制剂:构效关系及抗肿瘤免疫增强作用
IF 5.9 2区 医学 Q1 CHEMISTRY, MEDICINAL Pub Date : 2025-11-08 DOI: 10.1016/j.ejmech.2025.118356
Binbin Cheng , Chao Li , Xixiang Yang , Yinrong Wu , Yong Ruan , Yichang Ren , Zhenhong Su , Shanhe Wan , Xin Li , Dulin Kong , Jianjun Chen
Building on our previous research, a series of novel imidazo[1,2-a]pyridine derivatives were rationally designed and synthesized as tubulin polymerization inhibitors. Among these analogues, compound 5b exhibited the strongest antiproliferative activity against Jurkat, B16–F10, HCT116, and MDA-MB-231 cells, with IC50 values of 60 nM, 380 nM, 138 nM, and 1.054 μM, respectively. Further functional assays revealed that 5b can effectively suppress the migration and colony-forming capacity of B16–F10 cells. Mechanistically, compound 5b induced apoptosis and arrested the cell cycle in the G2/M phase by inhibiting tubulin polymerization. Molecular docking simulations revealed that 5b efficiently binds to the colchicine-binding pocket of tubulin, providing a structural basis for its activity. In vivo, compound 5b (10 mg/kg) demonstrated potent anti-tumor efficacy in a melanoma model without obvious systemic toxicity. Notably, 5b markedly potentiated the in vivo anti-tumor immune response through its combination with a PD-L1 monoclonal antibody (mAb), as evidenced by increased infiltration of cytotoxic CD8+ effector T cells in tumor tissues. Collectively, these findings identify 5b as a promising tubulin polymerization inhibitor with immune-modulatory potential, meriting further investigation.
在前人研究的基础上,合理设计合成了一系列新型咪唑[1,2-a]吡啶衍生物作为微管蛋白聚合抑制剂。其中,化合物5b对Jurkat、B16-F10、HCT116和MDA-MB-231细胞的抑制活性最强,IC50值分别为60 nM、380 nM、138 nM和1.054 μM。进一步的功能实验表明,5b能有效抑制B16-F10细胞的迁移和集落形成能力。机制上,化合物5b通过抑制微管蛋白聚合,诱导细胞凋亡并将细胞周期阻滞在G2/M期。分子对接模拟显示,5b有效结合到微管蛋白的秋水仙碱结合口袋,为其活性提供了结构基础。在体内,化合物5b (10 mg/kg)在黑色素瘤模型中显示出强大的抗肿瘤功效,无明显的全身毒性。值得注意的是,5b通过与PD-L1单克隆抗体(mAb)结合,显著增强了体内抗肿瘤免疫应答,这可以通过肿瘤组织中细胞毒性CD8+效应T细胞的浸润增加来证明。总的来说,这些发现确定5b是一种有希望的微管蛋白聚合抑制剂,具有免疫调节潜力,值得进一步研究。
{"title":"Novel imidazo[1,2-a]pyridine-based tubulin polymerization inhibitors: Structure-activity relationships and anti-tumor immune potentiation","authors":"Binbin Cheng ,&nbsp;Chao Li ,&nbsp;Xixiang Yang ,&nbsp;Yinrong Wu ,&nbsp;Yong Ruan ,&nbsp;Yichang Ren ,&nbsp;Zhenhong Su ,&nbsp;Shanhe Wan ,&nbsp;Xin Li ,&nbsp;Dulin Kong ,&nbsp;Jianjun Chen","doi":"10.1016/j.ejmech.2025.118356","DOIUrl":"10.1016/j.ejmech.2025.118356","url":null,"abstract":"<div><div>Building on our previous research, a series of novel imidazo[1,2-<em>a</em>]pyridine derivatives were rationally designed and synthesized as tubulin polymerization inhibitors. Among these analogues, compound <strong>5b</strong> exhibited the strongest antiproliferative activity against Jurkat, B16–F10, HCT116, and MDA-MB-231 cells, with IC<sub>50</sub> values of 60 nM, 380 nM, 138 nM, and 1.054 μM, respectively. Further functional assays revealed that <strong>5b</strong> can effectively suppress the migration and colony-forming capacity of B16–F10 cells. Mechanistically, compound <strong>5b</strong> induced apoptosis and arrested the cell cycle in the G2/M phase by inhibiting tubulin polymerization. Molecular docking simulations revealed that <strong>5b</strong> efficiently binds to the colchicine-binding pocket of tubulin, providing a structural basis for its activity. <em>In vivo</em>, compound <strong>5b</strong> (10 mg/kg) demonstrated potent anti-tumor efficacy in a melanoma model without obvious systemic toxicity. Notably, <strong>5b</strong> markedly potentiated the <em>in vivo</em> anti-tumor immune response through its combination with a PD-L1 monoclonal antibody (mAb), as evidenced by increased infiltration of cytotoxic CD8<sup>+</sup> effector T cells in tumor tissues. Collectively, these findings identify <strong>5b</strong> as a promising tubulin polymerization inhibitor with immune-modulatory potential, meriting further investigation.</div></div>","PeriodicalId":314,"journal":{"name":"European Journal of Medicinal Chemistry","volume":"302 ","pages":"Article 118356"},"PeriodicalIF":5.9,"publicationDate":"2025-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145462275","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