首页 > 最新文献

Chemical Biology & Drug Design最新文献

英文 中文
Screening Potential Drugs From Nitric Oxide Donors for Treating Alopecia Based on Network Pharmacology and Experimental Validation 基于网络药理学和实验验证筛选一氧化氮供体治疗脱发的潜在药物。
IF 3.3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-09 DOI: 10.1111/cbdd.70220
Pan Guo, Mengke Sun, Hong Cui, Jingyu Zhang, Junkai Huang, Jing Luo, Qianyu Zhu, Bingxin Zhang, Lizhi Hu

Nitric oxide (NO) was previously thought to have a beneficial role in hair growth, but it is unclear whether NO donors can encourage hair development. Therefore, this study aims to systematically investigate the physiological mechanisms underlying the therapeutic effects of NO donors in alopecia. We first screened the chemical targets of seven types of NO donors from DrugBank and SwissTargetPrediction, and further identified the differentially expressed genes (DEGs) of three types of alopecia from the GEO database. Integrated bioinformatic analysis was subsequently performed to comprehensively reveal potential pathways. A total of 339 chemical targets of NO donors were collected and 237, 266, and 1158 DEGs of androgenic alopecia (AGA), alopecia areata (AA) and central centrifugal cicatricial alopecia (CCCA) were identified in this study. We further extracted 20 CCCA-related targets from the intersection of 339 targets and 1158 DEGs of CCCA for the following analysis. Through integrative analysis, we found that hesperidin (HP) may exert dual regulatory effects on apoptosis and senescence pathways in hair follicle cells through its specific interactions with BCL2, MAPK8, MMP13 and AURKB, effectively mitigating hair loss. Overall, our work elucidates the molecular mechanisms of NO donors-mediated hair follicle protection, which could pave the way for future investigations into NO-based interventions and their potential clinical applications.

一氧化氮(NO)以前被认为对头发生长有益,但NO捐赠者是否能促进头发生长尚不清楚。因此,本研究旨在系统探讨NO供体治疗脱发的生理机制。我们首先筛选了来自DrugBank和SwissTargetPrediction的7种NO供体的化学靶点,并进一步从GEO数据库中鉴定了3种类型脱发的差异表达基因(DEGs)。随后进行综合生物信息学分析,以全面揭示潜在的途径。本研究共收集NO供体339个化学靶点,鉴定出雄激素性脱发(AGA)、斑秃(AA)和中心性瘢痕性脱发(CCCA)分别为237、266和1158℃。我们进一步从339个靶点和1158个CCCA度的交集中提取了20个CCCA相关靶点用于以下分析。通过综合分析,我们发现橙皮苷(hesperidin, HP)可能通过与BCL2、MAPK8、MMP13和AURKB的特异性相互作用,对毛囊细胞的凋亡和衰老通路发挥双重调控作用,有效缓解脱发。总之,我们的工作阐明了一氧化氮供体介导的毛囊保护的分子机制,这可能为未来研究基于一氧化氮的干预措施及其潜在的临床应用铺平道路。
{"title":"Screening Potential Drugs From Nitric Oxide Donors for Treating Alopecia Based on Network Pharmacology and Experimental Validation","authors":"Pan Guo,&nbsp;Mengke Sun,&nbsp;Hong Cui,&nbsp;Jingyu Zhang,&nbsp;Junkai Huang,&nbsp;Jing Luo,&nbsp;Qianyu Zhu,&nbsp;Bingxin Zhang,&nbsp;Lizhi Hu","doi":"10.1111/cbdd.70220","DOIUrl":"10.1111/cbdd.70220","url":null,"abstract":"<div>\u0000 \u0000 <p>Nitric oxide (NO) was previously thought to have a beneficial role in hair growth, but it is unclear whether NO donors can encourage hair development. Therefore, this study aims to systematically investigate the physiological mechanisms underlying the therapeutic effects of NO donors in alopecia. We first screened the chemical targets of seven types of NO donors from DrugBank and SwissTargetPrediction, and further identified the differentially expressed genes (DEGs) of three types of alopecia from the GEO database. Integrated bioinformatic analysis was subsequently performed to comprehensively reveal potential pathways. A total of 339 chemical targets of NO donors were collected and 237, 266, and 1158 DEGs of androgenic alopecia (AGA), alopecia areata (AA) and central centrifugal cicatricial alopecia (CCCA) were identified in this study. We further extracted 20 CCCA-related targets from the intersection of 339 targets and 1158 DEGs of CCCA for the following analysis. Through integrative analysis, we found that hesperidin (HP) may exert dual regulatory effects on apoptosis and senescence pathways in hair follicle cells through its specific interactions with BCL2, MAPK8, MMP13 and AURKB, effectively mitigating hair loss. Overall, our work elucidates the molecular mechanisms of NO donors-mediated hair follicle protection, which could pave the way for future investigations into NO-based interventions and their potential clinical applications.</p>\u0000 </div>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"106 6","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145710530","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Correction to “2-Hydroxy-3-Methylanthraquinone Suppresses Hepatocellular Carcinoma Progression by Blocking Annexin A5-Mediated Phosphatidylinositol 3-Kinase/Protein Kinase B Signaling” 更正“2-羟基-3-甲基蒽醌通过阻断膜联蛋白a5介导的磷脂酰肌醇3-激酶/蛋白激酶B信号传导抑制肝癌进展”。
IF 3.3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-09 DOI: 10.1111/cbdd.70216

Luo M, Mo J, Huang C, Mao Y, Wang H, Wang X. 2-Hydroxy-3-Methylanthraquinone Suppresses Hepatocellular Carcinoma Progression by Blocking Annexin A5-Mediated Phosphatidylinositol 3-Kinase/Protein Kinase B Signaling. Chemical Biology and Drug Design. 2025;106(1):e70161.

An important fund, Guike AB24010163, was omitted inadvertently from the article originally published and should have been added to the funder information.

We apologize for this error.

罗敏,莫军,黄晨,毛燕,王慧,王旭。2-羟基-3-甲基蒽醌阻断膜联蛋白a5介导的磷脂酰肌醇3-激酶/蛋白激酶B信号通路抑制肝癌进展。生物化学学报,2015;32(1):391 - 391。一项重要的基金,Guike AB24010163,在最初发表的文章中被无意中遗漏,本应添加到资助者信息中。我们为这个错误道歉。
{"title":"Correction to “2-Hydroxy-3-Methylanthraquinone Suppresses Hepatocellular Carcinoma Progression by Blocking Annexin A5-Mediated Phosphatidylinositol 3-Kinase/Protein Kinase B Signaling”","authors":"","doi":"10.1111/cbdd.70216","DOIUrl":"10.1111/cbdd.70216","url":null,"abstract":"<p>Luo M, Mo J, Huang C, Mao Y, Wang H, Wang X. 2-Hydroxy-3-Methylanthraquinone Suppresses Hepatocellular Carcinoma Progression by Blocking Annexin A5-Mediated Phosphatidylinositol 3-Kinase/Protein Kinase B Signaling. <i>Chemical Biology and Drug Design</i>. 2025;106(1):e70161.</p><p>An important fund, Guike AB24010163, was omitted inadvertently from the article originally published and should have been added to the funder information.</p><p>We apologize for this error.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"106 6","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cbdd.70216","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145710541","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
In Silico Exploration of Potential Inhibitors Targeting SARS-CoV-2 Non-Structural Protein 15 (Nsp15): A Comprehensive Overview of Current Research 针对SARS-CoV-2非结构蛋白15 (Nsp15)的潜在抑制剂的计算机探索:当前研究的综合综述
IF 3.3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-09 DOI: 10.1111/cbdd.70218
Behnaz Yazdani, Hajar Sirous, Farnoosh Farzam, Shayan Ansarizadeh, Mohammadjavad Karimi Taheri, Ghazal Pourmohammad-Hosseini, Maryam Noei, Kosar Daneshipour, Danial Shakeri, Maryam Houshangi, Vincenzo Calderone, Simone Brogi

The SARS-CoV-2 nonstructural protein 15 (Nsp15) is an endoribonuclease that plays a critical role in viral replication and immune evasion through its NendoU domain. The unique enzymatic mechanism of Nsp15 has attracted considerable attention as a potential therapeutic target, and the identification of its inhibitors could facilitate the development of novel antiviral agents against coronaviruses. Although biochemical and structural studies have provided important insights into Nsp15 function, no comprehensive review has yet focused on computational approaches applied to the discovery of Nsp15 inhibitors. Consequently, this study aims to address this gap by summarizing recent in silico research focused on the structure, function, and inhibition of Nsp15. Special attention is given to inhibitors derived from both natural and synthetic sources, as well as their binding interactions and predicted pharmacological potential. By integrating current computational findings, this review highlights novel prospects for the rational design of Nsp15-targeted therapeutics to combat SARS-CoV-2 and other related pathogenic coronaviruses.

SARS-CoV-2非结构蛋白15 (Nsp15)是一种核糖核酸内切酶,通过其NendoU结构域在病毒复制和免疫逃避中发挥关键作用。Nsp15独特的酶促机制作为一种潜在的治疗靶点引起了广泛的关注,其抑制剂的鉴定可以促进新型冠状病毒抗病毒药物的开发。尽管生物化学和结构研究已经为Nsp15的功能提供了重要的见解,但尚未有全面的综述关注应用于发现Nsp15抑制剂的计算方法。因此,本研究旨在通过总结最近关于Nsp15的结构、功能和抑制的硅研究来解决这一空白。特别关注来自天然和合成来源的抑制剂,以及它们的结合相互作用和预测的药理学潜力。通过整合当前的计算结果,本综述强调了合理设计nsp15靶向治疗方法以对抗SARS-CoV-2和其他相关致病性冠状病毒的新前景。
{"title":"In Silico Exploration of Potential Inhibitors Targeting SARS-CoV-2 Non-Structural Protein 15 (Nsp15): A Comprehensive Overview of Current Research","authors":"Behnaz Yazdani,&nbsp;Hajar Sirous,&nbsp;Farnoosh Farzam,&nbsp;Shayan Ansarizadeh,&nbsp;Mohammadjavad Karimi Taheri,&nbsp;Ghazal Pourmohammad-Hosseini,&nbsp;Maryam Noei,&nbsp;Kosar Daneshipour,&nbsp;Danial Shakeri,&nbsp;Maryam Houshangi,&nbsp;Vincenzo Calderone,&nbsp;Simone Brogi","doi":"10.1111/cbdd.70218","DOIUrl":"10.1111/cbdd.70218","url":null,"abstract":"<div>\u0000 \u0000 <p>The SARS-CoV-2 nonstructural protein 15 (Nsp15) is an endoribonuclease that plays a critical role in viral replication and immune evasion through its NendoU domain. The unique enzymatic mechanism of Nsp15 has attracted considerable attention as a potential therapeutic target, and the identification of its inhibitors could facilitate the development of novel antiviral agents against coronaviruses. Although biochemical and structural studies have provided important insights into Nsp15 function, no comprehensive review has yet focused on computational approaches applied to the discovery of Nsp15 inhibitors. Consequently, this study aims to address this gap by summarizing recent in silico research focused on the structure, function, and inhibition of Nsp15. Special attention is given to inhibitors derived from both natural and synthetic sources, as well as their binding interactions and predicted pharmacological potential. By integrating current computational findings, this review highlights novel prospects for the rational design of Nsp15-targeted therapeutics to combat SARS-CoV-2 and other related pathogenic coronaviruses.</p>\u0000 </div>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"106 6","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145710520","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Brusatol Exerts Therapeutic Effects in Ulcerative Colitis by Regulating the UBE3A-LCN2-Mediated Ferroptosis and Inflammation Axis Brusatol通过调节ubea3 - lcn2介导的铁垂和炎症轴对溃疡性结肠炎的治疗作用。
IF 3.3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-08 DOI: 10.1111/cbdd.70201
Xingzhuo Guan, Xiaohui Guan, Zhiping Yang, Yuanshi Wang, Yingze Ma, Yan Cui, Hongjun Xu

Ulcerative colitis (UC), a chronic inflammatory bowel disorder with no clear etiology, causes diverse complications and severely impairs patients' quality of life. Lipocalin-2 (LCN2) is recognized as a potential fecal biomarker for UC. Brusatol (BR), a quassinoid compound isolated from Brucea javanica, has exhibited anti-inflammatory and therapeutic effects in preclinical studies. While BR inhibits UC progression, its specific molecular mechanisms remain to be elucidated. An in vitro cell model of UC was constructed by treating human normal colorectal mucosal cells (FHC) with tumor necrosis factor-alpha (TNF-α). Cell viability and apoptosis were assessed using CCK-8, 5-ethynyl-2′-deoxyuridine (EdU) and flow cytometry. Additionally, inflammatory cytokines and ferroptosis-related factors were analyzed using corresponding kits. Western blot and qRT-PCR were conducted for protein and mRNA detection. Bioinformatics analysis and co-immunoprecipitation (Co-IP) assay were employed to investigate the relationship between LCN2 and ubiquitin protein ligase E3A (UBE3A). Finally, in vivo UC models were established, followed by BR treatment and evaluation of colon pathological changes and target molecule expression. In the TNF-α-induced FHC cell injury model, BR significantly suppressed cell injury and ferroptosis, and this effect was associated with downregulating LCN2 expression. The ubiquitin ligase UBE3A negatively regulated LCN2 expression. The protective effect of UBE3A overexpression against TNF-α-induced damage was abolished by LCN2 upregulation. BR alleviated cell injury by upregulating UBE3A expression, which in turn inhibited LCN2 expression. In the UC mouse model, BR mitigated colonic pathological damage, downregulated LCN2 expression, and upregulated UBE3A levels. BR exerted anti-colitis effects by upregulating the expression of UBE3A and downregulating LCN2 levels, thereby inhibiting cell injury, ferroptosis, and colonic pathological damage.

溃疡性结肠炎(UC)是一种慢性炎症性肠病,病因不明,可引起多种并发症,严重影响患者的生活质量。脂钙素-2 (Lipocalin-2, LCN2)被认为是UC的潜在粪便生物标志物。Brusatol (BR)是一种从鸦茅属植物中分离得到的准藜素类化合物,在临床前研究中具有抗炎和治疗作用。虽然BR抑制UC进展,但其具体的分子机制仍有待阐明。采用肿瘤坏死因子α (tumor necrosis factor -α, TNF-α)处理人正常结肠粘膜细胞(FHC),建立UC体外细胞模型。采用cck - 8,5 -乙基-2'-脱氧尿苷(EdU)和流式细胞术检测细胞活力和凋亡。此外,使用相应的试剂盒分析炎症因子和凋亡相关因子。Western blot和qRT-PCR检测蛋白和mRNA。采用生物信息学分析和共免疫沉淀法(Co-IP)研究LCN2与泛素蛋白连接酶E3A (UBE3A)的关系。最后建立UC体内模型,进行BR治疗,评估结肠病理变化及靶分子表达。在TNF-α-诱导的FHC细胞损伤模型中,BR显著抑制细胞损伤和铁下沉,这种作用与下调LCN2表达有关。泛素连接酶UBE3A负向调控LCN2的表达。UBE3A过表达对TNF-α-诱导的损伤的保护作用被LCN2上调所消除。BR通过上调UBE3A表达,进而抑制LCN2表达,从而减轻细胞损伤。在UC小鼠模型中,BR减轻了结肠病理损伤,下调了LCN2表达,上调了UBE3A水平。BR通过上调UBE3A表达,下调LCN2水平发挥抗结肠炎作用,从而抑制细胞损伤、铁凋亡和结肠病理损伤。
{"title":"Brusatol Exerts Therapeutic Effects in Ulcerative Colitis by Regulating the UBE3A-LCN2-Mediated Ferroptosis and Inflammation Axis","authors":"Xingzhuo Guan,&nbsp;Xiaohui Guan,&nbsp;Zhiping Yang,&nbsp;Yuanshi Wang,&nbsp;Yingze Ma,&nbsp;Yan Cui,&nbsp;Hongjun Xu","doi":"10.1111/cbdd.70201","DOIUrl":"10.1111/cbdd.70201","url":null,"abstract":"<div>\u0000 \u0000 <p>Ulcerative colitis (UC), a chronic inflammatory bowel disorder with no clear etiology, causes diverse complications and severely impairs patients' quality of life. Lipocalin-2 (LCN2) is recognized as a potential fecal biomarker for UC. Brusatol (BR), a quassinoid compound isolated from <i>Brucea javanica</i>, has exhibited anti-inflammatory and therapeutic effects in preclinical studies. While BR inhibits UC progression, its specific molecular mechanisms remain to be elucidated. An in vitro cell model of UC was constructed by treating human normal colorectal mucosal cells (FHC) with tumor necrosis factor-alpha (TNF-α). Cell viability and apoptosis were assessed using CCK-8, 5-ethynyl-2′-deoxyuridine (EdU) and flow cytometry. Additionally, inflammatory cytokines and ferroptosis-related factors were analyzed using corresponding kits. Western blot and qRT-PCR were conducted for protein and mRNA detection. Bioinformatics analysis and co-immunoprecipitation (Co-IP) assay were employed to investigate the relationship between LCN2 and ubiquitin protein ligase E3A (UBE3A). Finally, in vivo UC models were established, followed by BR treatment and evaluation of colon pathological changes and target molecule expression. In the TNF-α-induced FHC cell injury model, BR significantly suppressed cell injury and ferroptosis, and this effect was associated with downregulating LCN2 expression. The ubiquitin ligase UBE3A negatively regulated LCN2 expression. The protective effect of UBE3A overexpression against TNF-α-induced damage was abolished by LCN2 upregulation. BR alleviated cell injury by upregulating UBE3A expression, which in turn inhibited LCN2 expression. In the UC mouse model, BR mitigated colonic pathological damage, downregulated LCN2 expression, and upregulated UBE3A levels. BR exerted anti-colitis effects by upregulating the expression of UBE3A and downregulating LCN2 levels, thereby inhibiting cell injury, ferroptosis, and colonic pathological damage.</p>\u0000 </div>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"106 6","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145703336","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Platinum-Based Chemotherapeutics in the Modern Era: From Classical DNA-Targeting Mechanisms to Next-Generation Innovations in Cancer Therapy 现代以铂为基础的化疗药物:从经典的dna靶向机制到下一代癌症治疗创新。
IF 3.3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-06 DOI: 10.1111/cbdd.70208
Mohammad Bagher Shahsavani, Mahsa Heidari, Reza Yousefi, Ali Akbar Moosavi-Movahedi

Thanks to their DNA-crosslinking methods, platinum-based chemotherapeutics—which were first introduced by the coincidental discovery of cisplatin in 1965—have long been a mainstay of cancer treatment, with notable effectiveness in treating colorectal, ovarian, and testicular cancers. In order to address the enduring problems of toxicity, resistance, and restricted selectivity, this study charts their development from the traditional agents cisplatin, carboplatin, and oxaliplatin to next-generation developments. Classical mechanisms, rooted in aquation and apoptotic induction, are now complemented by emerging targets, including RNA, mitochondria, and protein–protein interactions, alongside novel cell death pathways like ferroptosis. Nonclassical complexes, such as Pt(IV) prodrugs and multinuclear agents, enhance delivery and overcome resistance, while synergistic strategies with immunotherapy (e.g., PD-1 inhibitors), nanoparticle delivery, and radiotherapy amplify efficacy. Precision medicine advances patient stratification via genomic (e.g., TP53 and BRCA) and proteomic biomarkers, liquid biopsies for real-time monitoring, and pharmacogenomics to adapt dosing. Sustainability initiatives, stable formulations, affordable generics, and green synthesis guarantee worldwide access, especially in low-resource environments. Recent trials have validated the use of hypoxia-activated prodrugs, AI-driven predictive models, and DNA repair inhibitors (e.g., NER and PARP) in the fight against resistance. Looking forward, integration with CRISPR, 3D tumor modeling, and epigenetic targeting heralds a new frontier, supported by interdisciplinary collaboration bridging chemistry, biology, and technology. This convergence of foundational principles and cutting-edge innovations positions platinum therapy for a transformative era, promising enhanced precision, efficacy, and equity in cancer care worldwide.

由于他们的dna交联方法,铂基化疗——1965年顺铂的偶然发现首次引入——长期以来一直是癌症治疗的支柱,在治疗结直肠癌、卵巢癌和睾丸癌方面效果显著。为了解决长期存在的毒性、耐药性和限制性选择性问题,本研究描绘了它们从传统药物顺铂、卡铂和奥沙利铂到下一代发展的发展历程。植根于水和凋亡诱导的经典机制现在被新兴靶点补充,包括RNA、线粒体和蛋白质-蛋白质相互作用,以及新的细胞死亡途径,如铁下垂。非经典复合物,如Pt(IV)前药和多核药物,可以增强递送并克服耐药性,而与免疫疗法(如PD-1抑制剂)、纳米颗粒递送和放疗的协同策略可以增强疗效。精准医学通过基因组(如TP53和BRCA)和蛋白质组学生物标志物、用于实时监测的液体活检和用于调整剂量的药物基因组学来推进患者分层。可持续性倡议、稳定的配方、负担得起的仿制药和绿色合成保证了全球范围内的可及性,特别是在资源匮乏的环境中。最近的试验已经验证了缺氧激活前药、人工智能驱动的预测模型和DNA修复抑制剂(例如NER和PARP)在对抗耐药性中的使用。展望未来,与CRISPR、3D肿瘤建模和表观遗传靶向的整合预示着一个新的前沿,在化学、生物学和技术的跨学科合作的支持下。这种基本原理和前沿创新的融合,将铂治疗推向了一个变革时代,有望在全球范围内提高癌症治疗的准确性、有效性和公平性。
{"title":"Platinum-Based Chemotherapeutics in the Modern Era: From Classical DNA-Targeting Mechanisms to Next-Generation Innovations in Cancer Therapy","authors":"Mohammad Bagher Shahsavani,&nbsp;Mahsa Heidari,&nbsp;Reza Yousefi,&nbsp;Ali Akbar Moosavi-Movahedi","doi":"10.1111/cbdd.70208","DOIUrl":"10.1111/cbdd.70208","url":null,"abstract":"<div>\u0000 \u0000 <p>Thanks to their DNA-crosslinking methods, platinum-based chemotherapeutics—which were first introduced by the coincidental discovery of cisplatin in 1965—have long been a mainstay of cancer treatment, with notable effectiveness in treating colorectal, ovarian, and testicular cancers. In order to address the enduring problems of toxicity, resistance, and restricted selectivity, this study charts their development from the traditional agents cisplatin, carboplatin, and oxaliplatin to next-generation developments. Classical mechanisms, rooted in aquation and apoptotic induction, are now complemented by emerging targets, including RNA, mitochondria, and protein–protein interactions, alongside novel cell death pathways like ferroptosis. Nonclassical complexes, such as Pt(IV) prodrugs and multinuclear agents, enhance delivery and overcome resistance, while synergistic strategies with immunotherapy (e.g., PD-1 inhibitors), nanoparticle delivery, and radiotherapy amplify efficacy. Precision medicine advances patient stratification via genomic (e.g., <i>TP53</i> and BRCA) and proteomic biomarkers, liquid biopsies for real-time monitoring, and pharmacogenomics to adapt dosing. Sustainability initiatives, stable formulations, affordable generics, and green synthesis guarantee worldwide access, especially in low-resource environments. Recent trials have validated the use of hypoxia-activated prodrugs, AI-driven predictive models, and DNA repair inhibitors (e.g., NER and PARP) in the fight against resistance. Looking forward, integration with CRISPR, 3D tumor modeling, and epigenetic targeting heralds a new frontier, supported by interdisciplinary collaboration bridging chemistry, biology, and technology. This convergence of foundational principles and cutting-edge innovations positions platinum therapy for a transformative era, promising enhanced precision, efficacy, and equity in cancer care worldwide.</p>\u0000 </div>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"106 6","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145688788","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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, Computational Modeling and Biological Evaluation of Novel N-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)cinnamamides as Potential Covalent Inhibitors for Oncology 新型肿瘤共价抑制剂N-(7H-Pyrrolo[2,3-d]嘧啶-4-yl)肉桂酰胺的设计、合成、计算建模和生物学评价。
IF 3.3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-06 DOI: 10.1111/cbdd.70211
Rameshwar S. Cheke, Prashant S. Kharkar

A novel series of covalent pyrrolo[2,3-d]pyrimidine cinnamamides was synthesized and evaluated for anticancer activity. The compounds were tested against three cancer cell lines—MDA-MB-231 (triple-negative breast cancer), A549 (non-small cell lung cancer), and U-87 MG (glioblastoma), alongside a cytotoxicity assessment on non-tumor NIH/3 T3 mouse embryonic fibroblast cell line. Compound 5k (IC50 = 0.99 ± 0.45 μM) and 5i (IC50 = 1.15 ± 0.14 μM) demonstrated markedly higher potency against MDA-MB-231 cells compared to cisplatin (IC50 = 34.36 ± 0.16 μM), whereas 5e was found active against A549 cells (IC50 = 1.41 ± 1.13 μM). None of the active compounds exhibited significant toxicity against normal NIH/3 T3 cells. Molecular docking studies involving covalent modification at the ATP-binding sites of EGFR (L858R/T790M), HER2, and JAK3 via key interactions with Cys797 (EGFR/HER2) and Cys909 (JAK3) supported their irreversible mechanism of action. Glutathione-binding assay confirmed the covalent adduct formation with a prominent molecular ion peak corresponding to the adduct. Density Functional Theory analyses of the leading compounds revealed low HOMO-LUMO energy gaps, which correlated with elevated electronic reactivity and biological capability. Electrostatic surface potential maps further highlighted distinct electrophilic character around the acrylamide warhead, facilitating potential covalent bond formation with nucleophilic Cys residues in the target proteins. In silico ADMET profiling confirmed favorable drug-likeness and safety attributes. Together, these findings support the discovery of this novel class of covalent pyrrolo[2,3-d] pyrimidine cinnamamides as promising and safer lead candidates for anticancer therapy, particularly against triple-negative breast cancer.

合成了一系列新的共价吡咯[2,3-d]嘧啶肉桂酰胺,并对其抗癌活性进行了评价。化合物对三种癌细胞系mda - mb -231(三阴性乳腺癌),A549(非小细胞肺癌)和U-87 MG(胶质母细胞瘤)进行了测试,并对非肿瘤NIH/3 T3小鼠胚胎成纤维细胞系进行了细胞毒性评估。化合物5k (IC50 = 0.99±0.45 μM)和5i (IC50 = 1.15±0.14 μM)对MDA-MB-231细胞的抑制作用明显高于顺铂(IC50 = 34.36±0.16 μM),而化合物5e对A549细胞的抑制作用明显高于顺铂(IC50 = 1.41±1.13 μM)。活性化合物对正常NIH/3 T3细胞均无明显毒性。通过与Cys797 (EGFR/HER2)和Cys909 (JAK3)的关键相互作用,在EGFR (L858R/T790M)、HER2和JAK3的atp结合位点进行共价修饰的分子对接研究支持了它们的不可逆作用机制。谷胱甘肽结合试验证实了共价加合物的形成,其分子离子峰与加合物相对应。密度泛函理论分析表明,先导化合物具有较低的HOMO-LUMO能隙,这与较高的电子反应性和生物性能有关。静电表面电位图进一步突出了丙烯酰胺战斗部周围明显的亲电性,促进了与靶蛋白中的亲核Cys残基形成潜在的共价键。在计算机ADMET分析证实有利的药物相似性和安全性属性。总之,这些发现支持了这类新型共价吡咯[2,3-d]嘧啶肉桂酰胺作为抗癌治疗,特别是治疗三阴性乳腺癌的有希望和更安全的主要候选药物的发现。
{"title":"Design, Synthesis, Computational Modeling and Biological Evaluation of Novel N-(7H-Pyrrolo[2,3-d]pyrimidin-4-yl)cinnamamides as Potential Covalent Inhibitors for Oncology","authors":"Rameshwar S. Cheke,&nbsp;Prashant S. Kharkar","doi":"10.1111/cbdd.70211","DOIUrl":"10.1111/cbdd.70211","url":null,"abstract":"<div>\u0000 \u0000 <p>A novel series of covalent pyrrolo[2,3-<i>d</i>]pyrimidine cinnamamides was synthesized and evaluated for anticancer activity. The compounds were tested against three cancer cell lines—MDA-MB-231 (triple-negative breast cancer), A549 (non-small cell lung cancer), and U-87 MG (glioblastoma), alongside a cytotoxicity assessment on non-tumor NIH/3 T3 mouse embryonic fibroblast cell line. Compound <b>5k</b> (IC<sub>50</sub> = 0.99 <b>±</b> 0.45 μM) and <b>5i</b> (IC<sub>50</sub> = 1.15 <b>±</b> 0.14 μM) demonstrated markedly higher potency against MDA-MB-231 cells compared to cisplatin (IC<sub>50</sub> = 34.36 ± 0.16 μM), whereas <b>5e</b> was found active against A549 cells (IC<sub>50</sub> = 1.41 ± 1.13 μM). None of the active compounds exhibited significant toxicity against normal NIH/3 T3 cells. Molecular docking studies involving covalent modification at the ATP-binding sites of EGFR (L858R/T790M), HER2, and JAK3 via key interactions with Cys797 (EGFR/HER2) and Cys909 (JAK3) supported their irreversible mechanism of action. Glutathione-binding assay confirmed the covalent adduct formation with a prominent molecular ion peak corresponding to the adduct. Density Functional Theory analyses of the leading compounds revealed low HOMO-LUMO energy gaps, which correlated with elevated electronic reactivity and biological capability. Electrostatic surface potential maps further highlighted distinct electrophilic character around the acrylamide warhead, facilitating potential covalent bond formation with nucleophilic Cys residues in the target proteins. In silico ADMET profiling confirmed favorable drug-likeness and safety attributes. Together, these findings support the discovery of this novel class of covalent pyrrolo[2,3-<i>d</i>] pyrimidine cinnamamides as promising and safer lead candidates for anticancer therapy, particularly against triple-negative breast cancer.</p>\u0000 </div>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"106 6","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145688797","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Correction to “Synthesis, Cytotoxic Activity, and Molecular Docking Study of New Thiazole-Incorporated Indenopyridazine Derivatives as Potential Antiproliferative Drugs” 对“新型噻唑类独立吡嗪衍生物的合成、细胞毒活性及分子对接研究”的更正
IF 3.3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-06 DOI: 10.1111/cbdd.70215

Alaa M. Alqahtani Sultan I. Alkbaysi, Mariam Mojally, Amgad Albohy, Synthesis, Cytotoxic Activity, and Molecular Docking Study of New Thiazole-Incorporated Indenopyridazine Derivatives as Potential Antiproliferative Drugs. Chem Biol Drug Des. 2025;106(5):e70203.

The name of the co-author Sultan I. Alkbaysi has been corrected to Sultan I. Alkubaysi.

We apologize for this error.

Alaa M. Alqahtani . Alkbaysi, Mariam Mojally, Amgad Albohy,新型噻唑-独立吡啶嗪衍生物的合成、细胞毒活性和分子对接研究。化学与药物杂志,2015;106(5):e70203。合著者Sultan I. Alkbaysi的名字已更正为Sultan I. Alkubaysi。我们为这个错误道歉。
{"title":"Correction to “Synthesis, Cytotoxic Activity, and Molecular Docking Study of New Thiazole-Incorporated Indenopyridazine Derivatives as Potential Antiproliferative Drugs”","authors":"","doi":"10.1111/cbdd.70215","DOIUrl":"https://doi.org/10.1111/cbdd.70215","url":null,"abstract":"<p>Alaa M. Alqahtani Sultan I. Alkbaysi, Mariam Mojally, Amgad Albohy, Synthesis, Cytotoxic Activity, and Molecular Docking Study of New Thiazole-Incorporated Indenopyridazine Derivatives as Potential Antiproliferative Drugs. <i>Chem Biol Drug Des</i>. 2025;106(5):e70203.</p><p>The name of the co-author Sultan I. Alkbaysi has been corrected to Sultan I. Alkubaysi.</p><p>We apologize for this error.</p>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"106 6","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/cbdd.70215","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145695028","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Therapeutic Potential of 6-Gingerol in Glioma: Mechanistic Insights and Experimental Validation 6-姜辣素在胶质瘤中的治疗潜力:机制见解和实验验证。
IF 3.3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-06 DOI: 10.1111/cbdd.70217
Shan Miao, Wei Hua, Jin Wang, Long Li, Yang Sun, Xiaopeng Shi, Shanbo Ma

Glioma, a prevalent malignancy globally, presents a serious threat to human health. Recent research has highlighted the significant anti-tumor properties of 6-gingerol (6-G), an active component of the traditional Chinese medicine ginger, though its precise mechanisms in glioma remain to be fully elucidated. This study employed network pharmacology, molecular docking, and dynamic simulation to explore the targets and underlying mechanisms of 6-G's anti-glioma effects, followed by in vitro validation of the key pathways. Drug-related targets were identified via PharmMapper and Swiss target prediction databases, while disease-related targets were sourced from GeneCards, OMIM, and TTD. The intersection of these datasets yielded potential therapeutic targets. Subsequent PPI network analysis using STRING11.5 and core gene screening with Cytoscape 3.9.1 led to the construction of a “drug-target network” and the identification of central genes. GO and KEGG enrichment analyses were conducted on potential therapeutic targets. Molecular docking and dynamic simulations were utilized to analyze the interactions between core genes and 6-G. Ultimately, the primary mechanism underlying 6-G's anti-GC effects was validated through in vitro experiments. A total of 183 potential targets for 6-G treatment of glioma were identified, with PPI analysis and core target screening revealing 10 critical targets. Enrichment analyses underscored the PI3K/AKT pathway as the primary signaling mechanism through which 6-G exerts its therapeutic effects. Molecular docking results showed that AKT1, HSP90AA1, EGFR, and MAPK3 were the key targets of 6-G in the treatment of glioma, and dynamic simulations further verified these findings. In vitro findings demonstrated that 6-G effectively inhibits the proliferation of U87 and U251 glioma cells, induces apoptosis, arrests the G1 phase of the cell cycle, and suppresses migration and invasion. Western blot analysis confirmed a significant downregulation of p-AKT in the PI3K/AKT pathway. In conclusion, 6-G impairs glioma cell proliferation, promotes apoptosis, induces G1 phase arrest, and inhibits migration and invasion, primarily through the suppression of the PI3K-AKT pathway. This finding provides a robust foundation for further research and clinical application.

神经胶质瘤是一种全球普遍存在的恶性肿瘤,对人类健康构成严重威胁。最近的研究强调了6-姜辣素(6-G)的显著抗肿瘤特性,尽管其在胶质瘤中的确切机制仍未完全阐明。本研究采用网络药理学、分子对接、动态模拟等方法,探索6-G抗胶质瘤作用的靶点和机制,并对关键通路进行体外验证。药物相关靶标通过PharmMapper和Swiss靶标预测数据库确定,而疾病相关靶标来自GeneCards、OMIM和TTD。这些数据集的交集产生了潜在的治疗靶点。随后使用STRING11.5进行PPI网络分析,使用Cytoscape 3.9.1进行核心基因筛选,构建了“药物靶点网络”并鉴定了中心基因。对潜在的治疗靶点进行GO和KEGG富集分析。利用分子对接和动态模拟分析了核心基因与6-G的相互作用。最后,通过体外实验验证了6-G抗gc作用的主要机制。通过PPI分析和核心靶点筛选,共发现183个6-G治疗胶质瘤的潜在靶点,揭示了10个关键靶点。富集分析强调PI3K/AKT通路是6-G发挥其治疗作用的主要信号机制。分子对接结果显示,AKT1、HSP90AA1、EGFR、MAPK3是6-G治疗胶质瘤的关键靶点,动态模拟进一步验证了这些发现。体外实验结果表明,6-G能有效抑制U87和U251胶质瘤细胞的增殖,诱导细胞凋亡,阻滞细胞周期G1期,抑制迁移和侵袭。Western blot分析证实了PI3K/AKT通路中p-AKT的显著下调。综上所述,6-G主要通过抑制PI3K-AKT通路,损害胶质瘤细胞增殖,促进细胞凋亡,诱导G1期阻滞,抑制迁移和侵袭。这一发现为进一步的研究和临床应用提供了坚实的基础。
{"title":"Therapeutic Potential of 6-Gingerol in Glioma: Mechanistic Insights and Experimental Validation","authors":"Shan Miao,&nbsp;Wei Hua,&nbsp;Jin Wang,&nbsp;Long Li,&nbsp;Yang Sun,&nbsp;Xiaopeng Shi,&nbsp;Shanbo Ma","doi":"10.1111/cbdd.70217","DOIUrl":"10.1111/cbdd.70217","url":null,"abstract":"<div>\u0000 \u0000 <p>Glioma, a prevalent malignancy globally, presents a serious threat to human health. Recent research has highlighted the significant anti-tumor properties of 6-gingerol (6-G), an active component of the traditional Chinese medicine ginger, though its precise mechanisms in glioma remain to be fully elucidated. This study employed network pharmacology, molecular docking, and dynamic simulation to explore the targets and underlying mechanisms of 6-G's anti-glioma effects, followed by in vitro validation of the key pathways. Drug-related targets were identified via PharmMapper and Swiss target prediction databases, while disease-related targets were sourced from GeneCards, OMIM, and TTD. The intersection of these datasets yielded potential therapeutic targets. Subsequent PPI network analysis using STRING11.5 and core gene screening with Cytoscape 3.9.1 led to the construction of a “drug-target network” and the identification of central genes. GO and KEGG enrichment analyses were conducted on potential therapeutic targets. Molecular docking and dynamic simulations were utilized to analyze the interactions between core genes and 6-G. Ultimately, the primary mechanism underlying 6-G's anti-GC effects was validated through in vitro experiments. A total of 183 potential targets for 6-G treatment of glioma were identified, with PPI analysis and core target screening revealing 10 critical targets. Enrichment analyses underscored the PI3K/AKT pathway as the primary signaling mechanism through which 6-G exerts its therapeutic effects. Molecular docking results showed that AKT1, HSP90AA1, EGFR, and MAPK3 were the key targets of 6-G in the treatment of glioma, and dynamic simulations further verified these findings. In vitro findings demonstrated that 6-G effectively inhibits the proliferation of U87 and U251 glioma cells, induces apoptosis, arrests the G1 phase of the cell cycle, and suppresses migration and invasion. Western blot analysis confirmed a significant downregulation of p-AKT in the PI3K/AKT pathway. In conclusion, 6-G impairs glioma cell proliferation, promotes apoptosis, induces G1 phase arrest, and inhibits migration and invasion, primarily through the suppression of the PI3K-AKT pathway. This finding provides a robust foundation for further research and clinical application.</p>\u0000 </div>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"106 6","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145688806","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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 and Synthesis of Holu-7, a Quinazolinone Derivative With an Active Stereo Geometry and Polar 2,4-Dihydroxyphenyl Group, as a Selective Anticancer Agent 选择性抗癌剂Holu-7的设计与合成。Holu-7是一种具有活性立体几何和极性2,4-二羟基苯基的喹唑啉酮衍生物。
IF 3.3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-05 DOI: 10.1111/cbdd.70209
Kuo-Chu Lai, Hui-Hsia Hsieh, Yi-Liang Lu, Shin-Yi Liu, Hai-Anh Ha, Mann-Jen Hour

Quinazolin-4-one scaffolds offer rich anticancer chemistry but often trade potency for selectivity. Building on a tubulin-targeting lead (MJ-65), 12 new derivatives (2637) were synthesized to dissect geometry–activity relationships and enhance the polarity of C2-substituents. Cytotoxicity was profiled against cancerous (HCT116 and CAL27) and normal (FHC) cells. In silico studies employed CDOCKER against tubulin (PDB 5NJH), GROMACS for molecular dynamics (MD), and ProTox-3.0 for toxicity estimation. The previously synthesized geometry-matched pairs 2125 were active at 10 μM. In contrast, their isomers 2630 were largely inactive, revealing a configuration-dependent binding mechanism. Optimizing polarity yielded Holu-7 (compound 35; 2,4-dihydroxyphenyl at the C2-position), which exhibited sub-50 nM IC50 in HCT116/CAL27 cells and > 10 μM in FHC cells and induced robust G2/M arrest. Docking/MD supported stable vinca-site engagement, with hydrogen bonds to ASP179/ASN206 and π-stacking with TYR224. ProTox-3.0 predicted a higher LD50 and absence of major organ toxicities relative to MJ-65. This data indicates that configuration-aware, polarity-tuned design can deliver selective quinazolinone anticancer candidates; therefore, Holu-7 merits further preclinical evaluation.

喹唑啉-4-酮支架提供了丰富的抗癌化学成分,但往往以选择性换取效力。以微管蛋白靶向先导物(MJ-65)为基础,合成了12个新的衍生物(26-37),以解析几何-活性关系并增强c2取代基的极性。对癌细胞(HCT116和CAL27)和正常细胞(FHC)进行细胞毒性分析。在计算机研究中,CDOCKER针对微管蛋白(PDB 5NJH), GROMACS用于分子动力学(MD), ProTox-3.0用于毒性评估。先前合成的几何匹配对21-25在10 μM范围内具有活性。相比之下,它们的同分异构体26-30大多不活跃,揭示了构型依赖的结合机制。优化极性得到Holu-7(化合物35,2,4-二羟基苯基在c2位置),其在HCT116/CAL27细胞中的IC50低于50 nM,在FHC细胞中的IC50为bb0 10 μM,并诱导了强大的G2/M阻滞。对接/MD通过与ASP179/ASN206的氢键和与TYR224的π叠加,支持稳定的葡萄位点结合。相对于MJ-65, ProTox-3.0预测更高的LD50和无主要器官毒性。这一数据表明,构型感知、极性调谐设计可以提供选择性喹唑啉酮抗癌候选药物;因此,Holu-7值得进一步的临床前评估。
{"title":"Design and Synthesis of Holu-7, a Quinazolinone Derivative With an Active Stereo Geometry and Polar 2,4-Dihydroxyphenyl Group, as a Selective Anticancer Agent","authors":"Kuo-Chu Lai,&nbsp;Hui-Hsia Hsieh,&nbsp;Yi-Liang Lu,&nbsp;Shin-Yi Liu,&nbsp;Hai-Anh Ha,&nbsp;Mann-Jen Hour","doi":"10.1111/cbdd.70209","DOIUrl":"10.1111/cbdd.70209","url":null,"abstract":"<div>\u0000 \u0000 <p>Quinazolin-4-one scaffolds offer rich anticancer chemistry but often trade potency for selectivity. Building on a tubulin-targeting lead (<b>MJ-65</b>), 12 new derivatives (<b>26</b>–<b>37</b>) were synthesized to dissect geometry–activity relationships and enhance the polarity of C2-substituents. Cytotoxicity was profiled against cancerous (HCT116 and CAL27) and normal (FHC) cells. <i>In silico</i> studies employed CDOCKER against tubulin (PDB 5NJH), GROMACS for molecular dynamics (MD), and ProTox-3.0 for toxicity estimation. The previously synthesized geometry-matched pairs <b>21</b>–<b>25</b> were active at 10 μM. In contrast, their isomers <b>26</b>–<b>30</b> were largely inactive, revealing a configuration-dependent binding mechanism. Optimizing polarity yielded <b>Holu-7</b> (compound <b>35</b>; 2,4-dihydroxyphenyl at the C2-position), which exhibited sub-50 nM IC<sub>50</sub> in HCT116/CAL27 cells and &gt; 10 μM in FHC cells and induced robust G<sub>2</sub>/M arrest. Docking/MD supported stable vinca-site engagement, with hydrogen bonds to ASP179/ASN206 and π-stacking with TYR224. ProTox-3.0 predicted a higher LD<sub>50</sub> and absence of major organ toxicities relative to <b>MJ-65</b>. This data indicates that configuration-aware, polarity-tuned design can deliver selective quinazolinone anticancer candidates; therefore, <b>Holu-7</b> merits further preclinical evaluation.</p>\u0000 </div>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"106 6","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145679548","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
In Silico and In Vitro Identification of New Eugenol Derivatives as Inhibitors of the Spike SARS-CoV-2 Protein Interaction With the ACE2 Host Receptor 新型丁香酚衍生物抑制SARS-CoV-2蛋白与ACE2宿主受体相互作用的硅和体外鉴定
IF 3.3 4区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-04 DOI: 10.1111/cbdd.70212
Timoteo Delgado-Maldonado, Alonzo González-González, Adriana Moreno-Rodríguez, Guadalupe Rojas-Verde, Alma D. Paz-Gonzalez, Debasish Bandyopadhyay, Gildardo Rivera

Coronavirus Disease-2019 (COVID-19) caused by SARS-CoV-2 remains a serious health concern worldwide. Inhibitors of the protein–protein interaction (iPPI) between the SARS-CoV-2 spike protein (S) and human Angiotensin-Converting Enzyme-2 (ACE2) are promising as potential antiviral agents. This study aimed to identify and evaluate novel compounds as inhibitors of the S receptor-binding domain (RBD) and ACE2 interaction through Eugenol- and Structure-Based virtual screening approaches. The hit compounds were corroborated as protein–protein interaction inhibitors using an ELISA-based enzyme assay. Molecular docking and molecular dynamics (MD) studies of the selected compounds showed that they maintained a molecular interaction and stability (RMSD fluctuations less than 4 Å) with essential residues of the S protein. The best compound, Eu-1 (IC50 = 16 μM), prevented the interaction between the S protein and ACE2 receptor efficiently with an 83.7% inhibition at 50 μM. In addition, Eu-1 had an adequate value of cytotoxicity (CC50 > 200 μM). Therefore, Eu-1 is a candidate compound for further SARS-CoV-2 preclinical experiments.

由SARS-CoV-2引起的冠状病毒病-2019 (COVID-19)仍然是全球严重的健康问题。SARS-CoV-2刺突蛋白(S)与人血管紧张素转换酶-2 (ACE2)之间的蛋白-蛋白相互作用(iPPI)抑制剂有望成为潜在的抗病毒药物。本研究旨在通过丁香酚和基于结构的虚拟筛选方法,鉴定和评估作为S受体结合域(RBD)和ACE2相互作用抑制剂的新化合物。命中的化合物被证实为蛋白质-蛋白质相互作用抑制剂,使用基于elisa的酶测定。分子对接和分子动力学(MD)研究表明,所选化合物与S蛋白必需残基保持分子相互作用和稳定性(RMSD波动小于4 Å)。最佳化合物Eu-1 (IC50 = 16 μM)在50 μM范围内抑制了S蛋白与ACE2受体的相互作用,抑制率为83.7%。此外,Eu-1具有足够的细胞毒性值(CC50 bb0 200 μM)。因此,Eu-1是进一步开展SARS-CoV-2临床前实验的候选化合物。
{"title":"In Silico and In Vitro Identification of New Eugenol Derivatives as Inhibitors of the Spike SARS-CoV-2 Protein Interaction With the ACE2 Host Receptor","authors":"Timoteo Delgado-Maldonado,&nbsp;Alonzo González-González,&nbsp;Adriana Moreno-Rodríguez,&nbsp;Guadalupe Rojas-Verde,&nbsp;Alma D. Paz-Gonzalez,&nbsp;Debasish Bandyopadhyay,&nbsp;Gildardo Rivera","doi":"10.1111/cbdd.70212","DOIUrl":"10.1111/cbdd.70212","url":null,"abstract":"<div>\u0000 \u0000 <p>Coronavirus Disease-2019 (COVID-19) caused by SARS-CoV-2 remains a serious health concern worldwide. Inhibitors of the protein–protein interaction (iPPI) between the SARS-CoV-2 spike protein (S) and human Angiotensin-Converting Enzyme-2 (ACE2) are promising as potential antiviral agents. This study aimed to identify and evaluate novel compounds as inhibitors of the S receptor-binding domain (RBD) and ACE2 interaction through Eugenol- and Structure-Based virtual screening approaches. The hit compounds were corroborated as protein–protein interaction inhibitors using an ELISA-based enzyme assay. Molecular docking and molecular dynamics (MD) studies of the selected compounds showed that they maintained a molecular interaction and stability (RMSD fluctuations less than 4 Å) with essential residues of the S protein. The best compound, <b>Eu-1</b> (IC<sub>50</sub> = 16 μM), prevented the interaction between the S protein and ACE2 receptor efficiently with an 83.7% inhibition at 50 μM. In addition, <b>Eu-1</b> had an adequate value of cytotoxicity (CC<sub>50</sub> &gt; 200 μM). Therefore, <b>Eu-1</b> is a candidate compound for further SARS-CoV-2 preclinical experiments.</p>\u0000 </div>","PeriodicalId":143,"journal":{"name":"Chemical Biology & Drug Design","volume":"106 6","pages":""},"PeriodicalIF":3.3,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145679564","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Chemical Biology & Drug Design
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