首页 > 最新文献

Archiv der Pharmazie最新文献

英文 中文
Unraveling the Anticancer Activity of Sunitinib Derivatives Through Modifications in Solvent-Exposed Regions: Synthesis, In Vitro Evaluation, and Computational Studies. 通过溶剂暴露区域的修饰揭示舒尼替尼衍生物的抗癌活性:合成,体外评价和计算研究。
IF 3.6 3区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-12-01 DOI: 10.1002/ardp.70168
Robby Gus Mahardika, Ade Danova, Chanat Aonbangkhen, Jaruwan Chatwichien, Sutthida Wongsuwan, Warinthorn Chavasiri, Elvira Hermawati, Anita Alni

This study modified the solvent-exposed region of sunitinib by replacing its diethylaminoethyl tail with linear and heterocyclic amines, guided by lipophilicity, steric, and electronic considerations to enhance the anticancer activity and selectivity. Sunitinib and its 20 derivatives, including 14 new compounds (4a, 4c, 4d, 4g-4i, 5a, 5b, 7a-7e, and 8) and 6 known compounds (4b, 4e-4f, 5c-5d, and 6), were successfully synthesized. The cytotoxic effects of sunitinib and its derivatives were evaluated against three human cancer cell lines (HeLa, SH-SY5Y, and HepG2) and one normal cell line (L929) using the MTT assay. Sunitinib exhibited the strongest cytotoxicity toward SH-SY5Y neuroblastoma cells, with an IC₅₀ of 3.88 µM. Among the derivatives, compound 5b showed the highest potency with an IC₅₀ of 4.28 µM against SH-SY5Y. Compound 6 displayed the highest selectivity index (SI ≥ 5) for HeLa, indicating good selectivity toward cancer over normal cells. Overall, these results suggest that targeted modification of the solvent-exposed region in sunitinib can improve anticancer activity without toxicity against normal cell lines, with compound 5b emerging as a promising lead for further development, particularly against neuroblastoma. The solvent-exposed region is strategic for drug development, allowing modifications that improve affinity, selectivity, solubility, and pharmacokinetics without disrupting ligand-protein interactions.

本研究利用亲脂性、位阻性和电子学方面的考虑,对舒尼替尼的溶剂暴露区进行修饰,用线性胺和杂环胺取代其二乙胺乙基尾部,以提高其抗癌活性和选择性。成功合成了舒尼替尼及其20个衍生物,包括14个新化合物(4a、4c、4d、4g-4i、5a、5b、7a-7e、8)和6个已知化合物(4b、4e-4f、5c-5d、6)。采用MTT法评价舒尼替尼及其衍生物对三种人类癌细胞系(HeLa、SH-SY5Y和HepG2)和一种正常细胞系(L929)的细胞毒作用。舒尼替尼对SH-SY5Y神经母细胞瘤细胞表现出最强的细胞毒性,IC₅0为3.88µM。在这些衍生物中,化合物5b对SH-SY5Y的IC₅0为4.28µM,显示出最高的效力。化合物6对HeLa的选择性指数最高(SI≥5),表明其对肿瘤的选择性优于正常细胞。总的来说,这些结果表明,靶向修饰舒尼替尼的溶剂暴露区域可以提高抗肿瘤活性,而对正常细胞系没有毒性,化合物5b成为进一步开发的有希望的线索,特别是针对神经母细胞瘤。溶剂暴露区域对药物开发具有战略意义,允许在不破坏配体-蛋白质相互作用的情况下进行修饰,提高亲和力、选择性、溶解度和药代动力学。
{"title":"Unraveling the Anticancer Activity of Sunitinib Derivatives Through Modifications in Solvent-Exposed Regions: Synthesis, In Vitro Evaluation, and Computational Studies.","authors":"Robby Gus Mahardika, Ade Danova, Chanat Aonbangkhen, Jaruwan Chatwichien, Sutthida Wongsuwan, Warinthorn Chavasiri, Elvira Hermawati, Anita Alni","doi":"10.1002/ardp.70168","DOIUrl":"https://doi.org/10.1002/ardp.70168","url":null,"abstract":"<p><p>This study modified the solvent-exposed region of sunitinib by replacing its diethylaminoethyl tail with linear and heterocyclic amines, guided by lipophilicity, steric, and electronic considerations to enhance the anticancer activity and selectivity. Sunitinib and its 20 derivatives, including 14 new compounds (4a, 4c, 4d, 4g-4i, 5a, 5b, 7a-7e, and 8) and 6 known compounds (4b, 4e-4f, 5c-5d, and 6), were successfully synthesized. The cytotoxic effects of sunitinib and its derivatives were evaluated against three human cancer cell lines (HeLa, SH-SY5Y, and HepG2) and one normal cell line (L929) using the MTT assay. Sunitinib exhibited the strongest cytotoxicity toward SH-SY5Y neuroblastoma cells, with an IC₅₀ of 3.88 µM. Among the derivatives, compound 5b showed the highest potency with an IC₅₀ of 4.28 µM against SH-SY5Y. Compound 6 displayed the highest selectivity index (SI ≥ 5) for HeLa, indicating good selectivity toward cancer over normal cells. Overall, these results suggest that targeted modification of the solvent-exposed region in sunitinib can improve anticancer activity without toxicity against normal cell lines, with compound 5b emerging as a promising lead for further development, particularly against neuroblastoma. The solvent-exposed region is strategic for drug development, allowing modifications that improve affinity, selectivity, solubility, and pharmacokinetics without disrupting ligand-protein interactions.</p>","PeriodicalId":128,"journal":{"name":"Archiv der Pharmazie","volume":"358 12","pages":"e70168"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145792629","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Design, Synthesis, and Biological Evaluation of Novel Benzimidazole/Schiff Base Hybrid Derivatives With Potential Biological Activities. 具有潜在生物活性的新型苯并咪唑/希夫碱杂化衍生物的设计、合成及生物学评价。
IF 3.6 3区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-12-01 DOI: 10.1002/ardp.70165
Mohamed Y Abdel-Hady, Martha M Morcoss, Abdullah Yahya Abdullah Alzahrani, Bahaa G M Youssif, El Shimaa M N Abdelhafez, Mohamed Abdel-Aziz

A novel series of benzimidazole-based derivatives (5a-g), (6a-b), and (7a-b) were designed, synthesized, and evaluated for their potential as dual inhibitors of EGFR and HER-2. The synthesized compounds were subjected to in vitro screening against a panel of selected human cancer cell lines. Additionally, their cytotoxicity was assessed using normal human mammary epithelial cells (MCF-10A) to evaluate their safety profile. Among the tested derivatives, compounds 5b, 5f, and 6a demonstrated the most pronounced antiproliferative activity, exhibiting IC₅₀ values of 6, 8, and 5 µM, respectively. These values reflect a potency at least fourfold greater than that of the reference drug Doxorubicin (IC₅₀ = 33 µM). EGFR and HER-2 enzyme inhibition assays were conducted to explore the potential molecular targets responsible for the observed anticancer effects. Notably, compound 6a (R₁ = phenyl, thiosemicarbazide) exhibited superior efficacy against the MCF-7 breast cancer cell line, with an IC₅₀ of 5 µM, approximately six times more potent than Doxorubicin. Conversely, compound 7b, with an IC₅₀ value of 85 µM against MCF-7 cells, was the least active, underscoring the critical role of the phenyl moiety in antiproliferative activity. Furthermore, a molecular docking study was conducted to investigate the binding interactions of 6a within the active sites of EGFR and HER-2, providing insight into its potential mechanism of action.

设计、合成了一系列新的苯并咪唑衍生物(5a-g)、(6a-b)和(7a-b),并评估了它们作为EGFR和HER-2双重抑制剂的潜力。合成的化合物在体外对一组选定的人类癌细胞进行筛选。此外,使用正常人乳腺上皮细胞(MCF-10A)评估其细胞毒性以评估其安全性。在所测试的衍生物中,化合物5b、5f和6a表现出最明显的抗增殖活性,其IC₅0值分别为6、8和5µM。这些值反映的效力至少是参比药物阿霉素(IC₅₀= 33 μ M)的四倍。进行了EGFR和HER-2酶抑制试验,以探索观察到的抗癌作用的潜在分子靶点。值得注意的是,化合物6a (R₁= phenyl, thiosemicarbazide)对MCF-7乳腺癌细胞系表现出卓越的功效,IC₅₀为5 μ M,效力约为阿霉素的6倍。相反,化合物7b对MCF-7细胞的IC₅0值为85µM,活性最低,强调了苯基部分在抗增殖活性中的关键作用。此外,我们还进行了分子对接研究,研究了6a在EGFR和HER-2活性位点的结合相互作用,从而深入了解其潜在的作用机制。
{"title":"Design, Synthesis, and Biological Evaluation of Novel Benzimidazole/Schiff Base Hybrid Derivatives With Potential Biological Activities.","authors":"Mohamed Y Abdel-Hady, Martha M Morcoss, Abdullah Yahya Abdullah Alzahrani, Bahaa G M Youssif, El Shimaa M N Abdelhafez, Mohamed Abdel-Aziz","doi":"10.1002/ardp.70165","DOIUrl":"https://doi.org/10.1002/ardp.70165","url":null,"abstract":"<p><p>A novel series of benzimidazole-based derivatives (5a-g), (6a-b), and (7a-b) were designed, synthesized, and evaluated for their potential as dual inhibitors of EGFR and HER-2. The synthesized compounds were subjected to in vitro screening against a panel of selected human cancer cell lines. Additionally, their cytotoxicity was assessed using normal human mammary epithelial cells (MCF-10A) to evaluate their safety profile. Among the tested derivatives, compounds 5b, 5f, and 6a demonstrated the most pronounced antiproliferative activity, exhibiting IC₅₀ values of 6, 8, and 5 µM, respectively. These values reflect a potency at least fourfold greater than that of the reference drug Doxorubicin (IC₅₀ = 33 µM). EGFR and HER-2 enzyme inhibition assays were conducted to explore the potential molecular targets responsible for the observed anticancer effects. Notably, compound 6a (R₁ = phenyl, thiosemicarbazide) exhibited superior efficacy against the MCF-7 breast cancer cell line, with an IC₅₀ of 5 µM, approximately six times more potent than Doxorubicin. Conversely, compound 7b, with an IC₅₀ value of 85 µM against MCF-7 cells, was the least active, underscoring the critical role of the phenyl moiety in antiproliferative activity. Furthermore, a molecular docking study was conducted to investigate the binding interactions of 6a within the active sites of EGFR and HER-2, providing insight into its potential mechanism of action.</p>","PeriodicalId":128,"journal":{"name":"Archiv der Pharmazie","volume":"358 12","pages":"e70165"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145761703","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Alantolactone as a Bioactive Sesquiterpene Lactone: Molecular Mechanisms and Anticancer Potential for Pharmaceutical Development. Alantolactone是一种具有生物活性的倍半萜内酯:分子机制和药物开发的抗癌潜力。
IF 3.6 3区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-12-01 DOI: 10.1002/ardp.70170
Alice Njolke Mafe, Lipana Dorcas Bappa, Javad Sharifi-Rad, William N Setzer, Daniela Calina

Cancer remains a major global health challenge, responsible for millions of deaths each year. The limitations of current therapies, including adverse effects and drug resistance, have intensified the search for safer, more effective alternatives, particularly from natural sources. Alantolactone, a sesquiterpene lactone extracted from Inula helenium and related medicinal plants, has emerged as a promising anticancer candidate due to its diverse pharmacological actions. Although notable anticancer properties have been reported, its full therapeutic potential, mechanisms of action, and translational relevance remain insufficiently explored and scattered across the literature. This review provides a comprehensive synthesis of current evidence on alantolactone's anticancer effects. It examines its sources, phytochemical features, structure-activity relationships, bioavailability challenges, mechanistic pathways, and preclinical findings, alongside perspectives for future clinical application. Evidence shows that alantolactone modulates multiple molecular targets and signaling cascades, including NF-κB, STAT3, MAPK, and apoptotic regulators. It demonstrates strong cytotoxic activity across diverse cancer cell lines and tumor-bearing animal models. However, barriers such as low systemic bioavailability, limited pharmacokinetic profiling, and the absence of human clinical data impede its therapeutic development. Alantolactone nonetheless holds significant promise as a multi-targeted anticancer compound with encouraging preclinical outcomes. Further research is required to address pharmacological limitations and establish safety and efficacy in clinical contexts. This review highlights alantolactone's potential contribution to future cancer therapy and emphasizes the need for interdisciplinary research to support its clinical translation and formulation advancement.

癌症仍然是一个重大的全球健康挑战,每年造成数百万人死亡。目前治疗方法的局限性,包括副作用和耐药性,促使人们加紧寻找更安全、更有效的替代方法,特别是天然来源的替代方法。Alantolactone是从菊属及相关药用植物中提取的倍半萜内酯,由于其多种药理作用而成为一种很有前景的抗癌候选药物。尽管已报道了显著的抗癌特性,但其全部治疗潜力、作用机制和翻译相关性仍未得到充分探索,并分散在文献中。本文综述了目前有关阿兰妥内酯抗癌作用的证据。它考察了其来源、植物化学特征、结构-活性关系、生物利用度挑战、机制途径和临床前发现,以及未来临床应用的前景。有证据表明,alantolactone调节多种分子靶点和信号级联,包括NF-κB、STAT3、MAPK和凋亡调节因子。它在不同的癌细胞系和荷瘤动物模型中显示出很强的细胞毒活性。然而,诸如低系统生物利用度、有限的药代动力学分析和缺乏人类临床数据等障碍阻碍了其治疗发展。尽管如此,作为一种多靶点抗癌化合物,阿兰妥内酯仍具有令人鼓舞的临床前结果。需要进一步的研究来解决药理学限制,并在临床环境中建立安全性和有效性。这篇综述强调了阿兰妥内酯对未来癌症治疗的潜在贡献,并强调了跨学科研究的必要性,以支持其临床转化和配方的进步。
{"title":"Alantolactone as a Bioactive Sesquiterpene Lactone: Molecular Mechanisms and Anticancer Potential for Pharmaceutical Development.","authors":"Alice Njolke Mafe, Lipana Dorcas Bappa, Javad Sharifi-Rad, William N Setzer, Daniela Calina","doi":"10.1002/ardp.70170","DOIUrl":"https://doi.org/10.1002/ardp.70170","url":null,"abstract":"<p><p>Cancer remains a major global health challenge, responsible for millions of deaths each year. The limitations of current therapies, including adverse effects and drug resistance, have intensified the search for safer, more effective alternatives, particularly from natural sources. Alantolactone, a sesquiterpene lactone extracted from Inula helenium and related medicinal plants, has emerged as a promising anticancer candidate due to its diverse pharmacological actions. Although notable anticancer properties have been reported, its full therapeutic potential, mechanisms of action, and translational relevance remain insufficiently explored and scattered across the literature. This review provides a comprehensive synthesis of current evidence on alantolactone's anticancer effects. It examines its sources, phytochemical features, structure-activity relationships, bioavailability challenges, mechanistic pathways, and preclinical findings, alongside perspectives for future clinical application. Evidence shows that alantolactone modulates multiple molecular targets and signaling cascades, including NF-κB, STAT3, MAPK, and apoptotic regulators. It demonstrates strong cytotoxic activity across diverse cancer cell lines and tumor-bearing animal models. However, barriers such as low systemic bioavailability, limited pharmacokinetic profiling, and the absence of human clinical data impede its therapeutic development. Alantolactone nonetheless holds significant promise as a multi-targeted anticancer compound with encouraging preclinical outcomes. Further research is required to address pharmacological limitations and establish safety and efficacy in clinical contexts. This review highlights alantolactone's potential contribution to future cancer therapy and emphasizes the need for interdisciplinary research to support its clinical translation and formulation advancement.</p>","PeriodicalId":128,"journal":{"name":"Archiv der Pharmazie","volume":"358 12","pages":"e70170"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145754957","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Quinoline/Pyrido-Pyrimidine Derivatives as Tubulin Polymerization Inhibitors: Design, Synthesis, Computational, and Anticancer Evaluation. 喹啉/吡啶嘧啶衍生物作为微管蛋白聚合抑制剂:设计、合成、计算和抗癌评价。
IF 3.6 3区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-12-01 DOI: 10.1002/ardp.70172
Divyakshi Arya, Gulshan Aara Khan, Shweta Singh, Anjali Upadhyay, Bharat Prasad Sharma, Rajesh Maharjan, Motee Lal Sharma, Munna P Gupt, Som Shankar Dubey, Prateek Pathak, Ashish Ranjan Dwivedi, Sarvesh Kumar Pandey

Cancer prevails a substantial health threat, with breast and colon cancers being the second and third most recurrent worldwide. In recent decades, quinoline/pyrido[2,3-d]pyrimidin-4(3H)-one derivatives have procured attention as propitious anticancer agents. This study acquaint a series of such compounds synthesized and characterized using spectroscopic (1H NMR, 13C NMR, IR, MS) and computational (DFT) methods. Their tubulin polymerization inhibitory and antiproliferative activities were assessed against cancer cell lines MCF-7, MDA-MB-231, and HCT-116, accompanying cytotoxicity screening against normal HEK-293 cells, divulging selective anticancer potential. SAR study accentuated the role of methoxy-substituted phenyl and cycloheptane rings in escalating activity. Peculiarly, compound 4g (IC50 = 3.02 ± 0.63 μM against cell line MCF-7) exhibited profound tubulin inhibition and was additionally substantiated via molecular docking and dynamics simulations, ratifying its drug-like behavior.

癌症是普遍存在的重大健康威胁,乳腺癌和结肠癌是全世界第二和第三大复发癌症。近几十年来,喹啉/吡啶[2,3-d]嘧啶-4(3H)- 1衍生物作为良好的抗癌药物引起了人们的关注。本研究采用波谱(1H NMR, 13C NMR, IR, MS)和计算(DFT)方法合成并表征了一系列此类化合物。对MCF-7、MDA-MB-231和HCT-116细胞系进行了微管蛋白聚合抑制和抗增殖活性评估,并对正常HEK-293细胞进行了细胞毒性筛选,揭示了选择性抗癌潜力。SAR研究强调了甲氧基取代苯基环和环庚烷环在提高活性中的作用。其中,化合物4g(对MCF-7的IC50 = 3.02±0.63 μM)具有较强的微管蛋白抑制作用,并通过分子对接和动力学模拟得到证实,证实了其药物样行为。
{"title":"Quinoline/Pyrido-Pyrimidine Derivatives as Tubulin Polymerization Inhibitors: Design, Synthesis, Computational, and Anticancer Evaluation.","authors":"Divyakshi Arya, Gulshan Aara Khan, Shweta Singh, Anjali Upadhyay, Bharat Prasad Sharma, Rajesh Maharjan, Motee Lal Sharma, Munna P Gupt, Som Shankar Dubey, Prateek Pathak, Ashish Ranjan Dwivedi, Sarvesh Kumar Pandey","doi":"10.1002/ardp.70172","DOIUrl":"https://doi.org/10.1002/ardp.70172","url":null,"abstract":"<p><p>Cancer prevails a substantial health threat, with breast and colon cancers being the second and third most recurrent worldwide. In recent decades, quinoline/pyrido[2,3-d]pyrimidin-4(3H)-one derivatives have procured attention as propitious anticancer agents. This study acquaint a series of such compounds synthesized and characterized using spectroscopic (1H NMR, 13C NMR, IR, MS) and computational (DFT) methods. Their tubulin polymerization inhibitory and antiproliferative activities were assessed against cancer cell lines MCF-7, MDA-MB-231, and HCT-116, accompanying cytotoxicity screening against normal HEK-293 cells, divulging selective anticancer potential. SAR study accentuated the role of methoxy-substituted phenyl and cycloheptane rings in escalating activity. Peculiarly, compound 4g (IC<sub>50</sub> = 3.02 ± 0.63 μM against cell line MCF-7) exhibited profound tubulin inhibition and was additionally substantiated via molecular docking and dynamics simulations, ratifying its drug-like behavior.</p>","PeriodicalId":128,"journal":{"name":"Archiv der Pharmazie","volume":"358 12","pages":"e70172"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145792646","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Design, Synthesis, and Integrated In Silico-In Vitro Evaluation of Triazole-Linked Benz/Imidazole-2-Thione/Selone Derivatives as Selective CDK1 Inhibitors for Breast Cancer Therapy. 设计,合成和集成在硅-体外评价三唑连接的奔驰/咪唑-2-硫酮/Selone衍生物作为选择性CDK1抑制剂用于乳腺癌治疗。
IF 3.6 3区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-12-01 DOI: 10.1002/ardp.70173
Alameer Ezat Abdulkareem, Ahmed Hassoon Mageed

Benz/imidazole-2-thione/selone-based triazoles, particularly their thione and selone analogs, are gaining attention for anticancer drug development due to their structural diversity and biological activity. However, their potential as targeted inhibitors of cancer-related proteins remains underexplored. This study reports the design, synthesis, and evaluation of novel benz/imidazole-2-thione/selone-based triazoles, focusing on cyclin-dependent kinase 1 (CDK1), a key regulator of cancer cell proliferation. The compounds were synthesized via a multistep approach involving imidazolium salt intermediates, followed by sulfur or selenium incorporation. Structural confirmation was achieved using FT-IR, NMR, and mass spectrometry. Molecular docking against CDK1, TERT, and VEGFR2 revealed strong binding affinities (-9.7 to -7.3 kcal/mol), with CDK1 selected for further in vitro study using MCF-7 breast cancer cells. Molecular dynamics (MD) simulations confirmed stable CDK1 binding for Compounds 2, 4, and 9, although Compound 9 showed conformational instability after 60 ns. ADMET profiling indicated favorable drug-likeness and permeability but highlighted metabolic liabilities and hERG inhibition risks, particularly for Compounds 4 and 9. The target prediction and pathway enrichment analyses predict that benz/imidazole-2-thione/selone-based triazoles exert their pharmacological effects primarily through the regulation of GPCR signaling pathways, likely via direct interaction with key regulators such as RGS8 and RGS4. In vitro assays demonstrated dose-dependent cytotoxicity, with Compound 4 showing the highest potency (IC50 = 106.12 ± 1.03 µg/mL), followed by 9 and 2. These findings suggest that benz/imidazole-2-thione/selone-based triazoles are promising CDK1 inhibitors and support their further optimization as targeted breast cancer therapies.

基于本茨/咪唑-2-硫酮/自酮的三唑类化合物,尤其是它们的硫酮和自酮类似物,由于其结构的多样性和生物活性,在抗癌药物开发中越来越受到关注。然而,它们作为癌症相关蛋白靶向抑制剂的潜力仍未得到充分开发。本研究报道了基于苯并/咪唑-2-硫酮/自酮的新型三唑的设计、合成和评价,重点关注细胞周期蛋白依赖性激酶1 (CDK1),这是癌细胞增殖的关键调节因子。这些化合物是通过咪唑盐中间体,然后加入硫或硒的多步骤方法合成的。使用FT-IR, NMR和质谱法进行结构确认。与CDK1、TERT和VEGFR2的分子对接显示出很强的结合亲和力(-9.7至-7.3 kcal/mol), CDK1被选择用于进一步使用MCF-7乳腺癌细胞的体外研究。分子动力学(MD)模拟证实了化合物2、4和9与CDK1的稳定结合,尽管化合物9在60 ns后表现出构象不稳定性。ADMET分析显示了良好的药物相似性和渗透性,但强调了代谢负荷和hERG抑制风险,特别是化合物4和9。靶标预测和途径富集分析预测,苯并/咪唑-2-硫酮/自酮基三唑主要通过调控GPCR信号通路发挥药理作用,可能与RGS8和RGS4等关键调控因子直接相互作用。体外实验显示出剂量依赖性的细胞毒性,化合物4的效价最高(IC50 = 106.12±1.03µg/mL),其次是化合物9和化合物2。这些发现表明,以苯并咪唑-2-硫酮/自酮为基础的三唑类药物是很有前景的CDK1抑制剂,并支持其作为乳腺癌靶向治疗的进一步优化。
{"title":"Design, Synthesis, and Integrated In Silico-In Vitro Evaluation of Triazole-Linked Benz/Imidazole-2-Thione/Selone Derivatives as Selective CDK1 Inhibitors for Breast Cancer Therapy.","authors":"Alameer Ezat Abdulkareem, Ahmed Hassoon Mageed","doi":"10.1002/ardp.70173","DOIUrl":"https://doi.org/10.1002/ardp.70173","url":null,"abstract":"<p><p>Benz/imidazole-2-thione/selone-based triazoles, particularly their thione and selone analogs, are gaining attention for anticancer drug development due to their structural diversity and biological activity. However, their potential as targeted inhibitors of cancer-related proteins remains underexplored. This study reports the design, synthesis, and evaluation of novel benz/imidazole-2-thione/selone-based triazoles, focusing on cyclin-dependent kinase 1 (CDK1), a key regulator of cancer cell proliferation. The compounds were synthesized via a multistep approach involving imidazolium salt intermediates, followed by sulfur or selenium incorporation. Structural confirmation was achieved using FT-IR, NMR, and mass spectrometry. Molecular docking against CDK1, TERT, and VEGFR2 revealed strong binding affinities (-9.7 to -7.3 kcal/mol), with CDK1 selected for further in vitro study using MCF-7 breast cancer cells. Molecular dynamics (MD) simulations confirmed stable CDK1 binding for Compounds 2, 4, and 9, although Compound 9 showed conformational instability after 60 ns. ADMET profiling indicated favorable drug-likeness and permeability but highlighted metabolic liabilities and hERG inhibition risks, particularly for Compounds 4 and 9. The target prediction and pathway enrichment analyses predict that benz/imidazole-2-thione/selone-based triazoles exert their pharmacological effects primarily through the regulation of GPCR signaling pathways, likely via direct interaction with key regulators such as RGS8 and RGS4. In vitro assays demonstrated dose-dependent cytotoxicity, with Compound 4 showing the highest potency (IC<sub>50</sub> = 106.12 ± 1.03 µg/mL), followed by 9 and 2. These findings suggest that benz/imidazole-2-thione/selone-based triazoles are promising CDK1 inhibitors and support their further optimization as targeted breast cancer therapies.</p>","PeriodicalId":128,"journal":{"name":"Archiv der Pharmazie","volume":"358 12","pages":"e70173"},"PeriodicalIF":3.6,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145792677","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exploring Nirmatrelvir Derivatives Through P2 Substituent Modifications and Warhead Innovations Targeting the Main Protease of SARS-CoV-2 通过P2取代基修饰和弹头创新探索针对SARS-CoV-2主要蛋白酶的Nirmatrelvir衍生物
IF 3.6 3区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-11-29 DOI: 10.1002/ardp.70158
Felipe Cardoso Prado Martins, Johannes Lang, Fernanda dos Reis Rocho, Xianxian Wang, Vinícius Bonatto, Jerônimo Lameira, Christian Klein, Carlos Alberto Montanari

The COVID-19 pandemic underscored the urgent need for effective antiviral agents, particularly against coronaviruses, which pose a continuing threat of future outbreaks. Targeting the main protease (Mpro), a key enzyme in viral replication, represents a promising therapeutic strategy. This study investigates structural modifications to known Mpro inhibitors, focusing on substitutions at the P2 position to explore alterations in both inhibitory potency and metabolic stability. Computational modeling and biochemical assays revealed that incorporating large, hydrophobic, and π-rich groups, such as the 4-phenylproline, significantly enhances binding affinity. Additionally, we evaluated warheads that have not yet been explored in the context of SARS-CoV-2 Mpro inhibition. Among these, fluoro-vinylsulfone and nitrile groups demonstrated superior inhibitory activity. A fragment-merging strategy combining an optimized P2 substituent with the nitrile warhead yielded a hybrid molecule with binding affinity comparable to nirmatrelvir. However, other analogs incorporating individual warhead optimizations displayed similar potency. These findings generate valuable insights into the design of robust Mpro inhibitors and support their potential development as broad-spectrum antiviral agents.

2019冠状病毒病大流行凸显了迫切需要有效的抗病毒药物,特别是针对冠状病毒的抗病毒药物,因为冠状病毒对未来疫情构成持续威胁。靶向主要蛋白酶(Mpro)是病毒复制的关键酶,是一种很有前景的治疗策略。本研究研究了已知Mpro抑制剂的结构修饰,重点关注P2位置的取代,以探索抑制效力和代谢稳定性的变化。计算模型和生化分析表明,加入大的、疏水的、富π的基团,如4-苯基脯氨酸,显著提高了结合亲和力。此外,我们还评估了尚未在SARS-CoV-2 Mpro抑制背景下探索的弹头。其中,氟乙烯基砜和腈基表现出较好的抑制活性。将优化的P2取代基与腈战斗部结合的片段合并策略产生了具有与nirmatrelvir相当的结合亲和力的杂化分子。然而,其他包含单个弹头优化的类似物显示出类似的效力。这些发现为设计强大的Mpro抑制剂提供了有价值的见解,并支持其作为广谱抗病毒药物的潜在发展。
{"title":"Exploring Nirmatrelvir Derivatives Through P2 Substituent Modifications and Warhead Innovations Targeting the Main Protease of SARS-CoV-2","authors":"Felipe Cardoso Prado Martins,&nbsp;Johannes Lang,&nbsp;Fernanda dos Reis Rocho,&nbsp;Xianxian Wang,&nbsp;Vinícius Bonatto,&nbsp;Jerônimo Lameira,&nbsp;Christian Klein,&nbsp;Carlos Alberto Montanari","doi":"10.1002/ardp.70158","DOIUrl":"https://doi.org/10.1002/ardp.70158","url":null,"abstract":"<p>The COVID-19 pandemic underscored the urgent need for effective antiviral agents, particularly against coronaviruses, which pose a continuing threat of future outbreaks. Targeting the main protease (M<sup>pro</sup>), a key enzyme in viral replication, represents a promising therapeutic strategy. This study investigates structural modifications to known M<sup>pro</sup> inhibitors, focusing on substitutions at the P2 position to explore alterations in both inhibitory potency and metabolic stability. Computational modeling and biochemical assays revealed that incorporating large, hydrophobic, and π-rich groups, such as the 4-phenylproline, significantly enhances binding affinity. Additionally, we evaluated warheads that have not yet been explored in the context of SARS-CoV-2 M<sup>pro</sup> inhibition. Among these, fluoro-vinylsulfone and nitrile groups demonstrated superior inhibitory activity. A fragment-merging strategy combining an optimized P2 substituent with the nitrile warhead yielded a hybrid molecule with binding affinity comparable to nirmatrelvir. However, other analogs incorporating individual warhead optimizations displayed similar potency. These findings generate valuable insights into the design of robust M<sup>pro</sup> inhibitors and support their potential development as broad-spectrum antiviral agents.</p>","PeriodicalId":128,"journal":{"name":"Archiv der Pharmazie","volume":"358 11","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/ardp.70158","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145619285","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
C-5-Arylidene-Thiazolidine-2,4-Dione Analogs Against Aldose Reductase: Design, Synthesis, Biological Evaluation, and Computational Insights 抗醛糖还原酶的c -5-芳基烷-噻唑烷-2,4-二酮类似物:设计、合成、生物学评价和计算见解
IF 3.6 3区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-11-29 DOI: 10.1002/ardp.70159
Rajiv Patel, Sanjeev Ranjan,  Nandini, Anwesha Das, Arijit Nandi, Sant Kumar Verma

Diabetic complications are major health problems that happen to people with diabetes when their blood sugar levels stay high for a long time and damage various body organs and systems. Inhibition of aldose reductase has emerged as a promising strategy for drug development and the management of diabetes-associated complications. In the research article, a novel series of 12 compounds, “C−5-arylidene thiazolidine-2,4-dione derivatives” was constructed, synthesized, and characterized using both spectral and non-spectral techniques. The confirmed synthesized compounds were subsequently screened for in vitro aldose reductase inhibitory activity. Among them, compounds 9h and 9a exhibited superior potency against aldose reductase protein with IC50 values of 0.98 and 1.05 µM, respectively, as compared with the reference drug epalrestat (IC50 value of 1.20 µM). Additionally, compounds 9l and 9i showed moderate aldose reductase inhibitory activity with an IC50 value of 1.36 and 2.47 µM, respectively. Furthermore, the top four best active compounds were selected for in vivo anti-hyperglycaemic activity. Results revealed that all the tested compounds significantly decreased blood glucose levels at all time intervals. Computational analysis revealed that all synthesized candidates bound firmly in the protein cavity of aldose reductase via hydrogen bonds and steric interaction with the range of docking score −10.21 to −11.81 kcal/mol. Predicted in silico toxicity data suggested that the synthesized compounds were inactive and nontoxic against peroxisome proliferator-activated receptor-γ protein. These finding results support the potential of thiazolidine-2,4-dione derivatives as promising AR inhibitors for managing diabetic complications.

糖尿病并发症是糖尿病患者的主要健康问题,当他们的血糖水平长期处于高位时,会损害身体的各个器官和系统。醛糖还原酶的抑制已成为药物开发和糖尿病相关并发症管理的一种有前途的策略。本文构建、合成了12个新的化合物“C−5-芳基噻唑烷-2,4-二酮衍生物”,并利用光谱和非光谱技术对其进行了表征。确定的合成化合物随后筛选体外醛糖还原酶抑制活性。其中化合物9h和9a对醛糖还原酶蛋白的IC50值分别为0.98和1.05µM,优于对照药依帕司他(IC50值为1.20µM)。化合物91和9i具有中等醛糖还原酶抑制活性,IC50值分别为1.36和2.47µM。此外,我们还筛选出了4种具有较强体内抗高血糖活性的活性化合物。结果显示,所有测试的化合物在所有时间间隔显著降低血糖水平。计算分析表明,所有合成的候选物通过氢键和空间相互作用牢固地结合在醛糖还原酶的蛋白腔中,对接分数范围为−10.21 ~−11.81 kcal/mol。硅毒性预测数据表明,合成的化合物对过氧化物酶体增殖体激活受体-γ蛋白无活性且无毒。这些发现结果支持噻唑烷-2,4-二酮衍生物作为治疗糖尿病并发症的有希望的AR抑制剂的潜力。
{"title":"C-5-Arylidene-Thiazolidine-2,4-Dione Analogs Against Aldose Reductase: Design, Synthesis, Biological Evaluation, and Computational Insights","authors":"Rajiv Patel,&nbsp;Sanjeev Ranjan,&nbsp; Nandini,&nbsp;Anwesha Das,&nbsp;Arijit Nandi,&nbsp;Sant Kumar Verma","doi":"10.1002/ardp.70159","DOIUrl":"https://doi.org/10.1002/ardp.70159","url":null,"abstract":"<div>\u0000 \u0000 <p>Diabetic complications are major health problems that happen to people with diabetes when their blood sugar levels stay high for a long time and damage various body organs and systems. Inhibition of aldose reductase has emerged as a promising strategy for drug development and the management of diabetes-associated complications. In the research article, a novel series of 12 compounds, “<i>C</i>−5-arylidene thiazolidine-2,4-dione derivatives” was constructed, synthesized, and characterized using both spectral and non-spectral techniques. The confirmed synthesized compounds were subsequently screened for in vitro aldose reductase inhibitory activity. Among them, compounds <b>9h</b> and <b>9a</b> exhibited superior potency against aldose reductase protein with IC<sub>50</sub> values of 0.98 and 1.05 µM, respectively, as compared with the reference drug epalrestat (IC<sub>50</sub> value of 1.20 µM). Additionally, compounds <b>9l</b> and <b>9i</b> showed moderate aldose reductase inhibitory activity with an IC<sub>50</sub> value of 1.36 and 2.47 µM, respectively. Furthermore, the top four best active compounds were selected for in vivo anti-hyperglycaemic activity. Results revealed that all the tested compounds significantly decreased blood glucose levels at all time intervals. Computational analysis revealed that all synthesized candidates bound firmly in the protein cavity of aldose reductase via hydrogen bonds and steric interaction with the range of docking score −10.21 to −11.81 kcal/mol. Predicted in silico toxicity data suggested that the synthesized compounds were inactive and nontoxic against peroxisome proliferator-activated receptor-γ protein. These finding results support the potential of thiazolidine-2,4-dione derivatives as promising AR inhibitors for managing diabetic complications.</p></div>","PeriodicalId":128,"journal":{"name":"Archiv der Pharmazie","volume":"358 11","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145618930","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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 Kojic Acid-Thiadiazole-Urea Hybrids: Potent Tyrosinase Inhibitors With Enhanced Anti-Melanogenic Activity 新型曲酸-噻二唑-尿素复合物:具有增强抗黑色素活性的强效酪氨酸酶抑制剂
IF 3.6 3区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-11-29 DOI: 10.1002/ardp.70147
Ayyub Mojaddami, Mojtaba Hamzi, Mehdi Khoshneviszadeh, Maryam Kabiri, Mahshid Attarroshan, Alireza Foroumadi, Mahsa Toolabi

A new class of kojic acid–derived tyrosinase inhibitors was rationally designed through the incorporation of a thiadiazole-urea framework. Accordingly, a series of 1-(aryl)-3-(5-{[(5-hydroxy-4-oxo-4H-pyran-2-yl)methyl]thio}-1,3,4-thiadiazol-2-yl)urea derivatives was synthesized starting from thiosemicarbazide. Through this approach, 13 new compounds were prepared with good yields via a straightforward procedure. The strategy involved substituting the hydroxymethyl group with a thiadiazol-urea pharmacophore, leading to the discovery of compounds that exhibited significantly enhanced inhibitory activity (IC50 range: 0.02–0.96 µM) compared to kojic acid (IC50 = 64.32 µM) against tyrosinase. Further kinetic, antioxidant, and docking studies, along with melanin content assays, were conducted to elucidate the mechanism of action for the most active compounds.

通过引入噻二唑-尿素结构,合理设计了一类新的曲酸酪氨酸酶抑制剂。据此,以硫代氨基脲为起始原料,合成了一系列1-(芳基)-3-(5-{(5-羟基-4-氧- 4h -吡喃-2-基)甲基]硫代}- 3,4-噻二唑-2-基)脲衍生物。通过这种方法,通过简单的程序,以良好的收率制备了13种新化合物。该策略涉及用噻二唑-尿素药效团取代羟甲基,导致发现与曲酸(IC50 = 64.32µM)相比,化合物对酪氨酸酶的抑制活性显著增强(IC50范围:0.02-0.96µM)。进一步进行了动力学、抗氧化和对接研究,以及黑色素含量测定,以阐明大多数活性化合物的作用机制。
{"title":"Novel Kojic Acid-Thiadiazole-Urea Hybrids: Potent Tyrosinase Inhibitors With Enhanced Anti-Melanogenic Activity","authors":"Ayyub Mojaddami,&nbsp;Mojtaba Hamzi,&nbsp;Mehdi Khoshneviszadeh,&nbsp;Maryam Kabiri,&nbsp;Mahshid Attarroshan,&nbsp;Alireza Foroumadi,&nbsp;Mahsa Toolabi","doi":"10.1002/ardp.70147","DOIUrl":"https://doi.org/10.1002/ardp.70147","url":null,"abstract":"<div>\u0000 \u0000 <p>A new class of kojic acid–derived tyrosinase inhibitors was rationally designed through the incorporation of a thiadiazole-urea framework. Accordingly, a series of 1-(aryl)-3-(5-{[(5-hydroxy-4-oxo-4<i>H</i>-pyran-2-yl)methyl]thio}-1,3,4-thiadiazol-2-yl)urea derivatives was synthesized starting from thiosemicarbazide. Through this approach, 13 new compounds were prepared with good yields via a straightforward procedure. The strategy involved substituting the hydroxymethyl group with a thiadiazol-urea pharmacophore, leading to the discovery of compounds that exhibited significantly enhanced inhibitory activity (IC<sub>50</sub> range: 0.02–0.96 µM) compared to kojic acid (IC<sub>50</sub> = 64.32 µM) against tyrosinase. Further kinetic, antioxidant, and docking studies, along with melanin content assays, were conducted to elucidate the mechanism of action for the most active compounds.</p></div>","PeriodicalId":128,"journal":{"name":"Archiv der Pharmazie","volume":"358 11","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145619350","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Reevaluating 1,3-Thiazolidinone Reports: Deciphering the Authentic 1,3-Thiazolidinone Frameworks via Synthetic and Spectroscopic Insights 重新评估1,3-噻唑烷酮报告:通过合成和光谱见解解读真实的1,3-噻唑烷酮框架
IF 3.6 3区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-11-29 DOI: 10.1002/ardp.70157
Roghayeh Hosseininia, Farough Nasiri

Accurate structural determination of heterocyclic scaffolds is essential for medicinal chemistry and drug discovery. In this study, products obtained from three-component reactions of aryl isothiocyanates, dialkyl acetylenedicarboxylates, and hydrazine hydrate, as well as four-component reactions incorporating aldehydes, were carefully re-examined. Previous studies had described these compounds as thiazine derivatives; however, our comprehensive synthetic and spectroscopic investigations demonstrate that they actually correspond to thiazolidinone frameworks. Key evidence was provided by gradient-selected heteronuclear multiple bond correlation (HMBC) and gated 13C NMR spectroscopy, which enabled the determination of regioselectivity and stereochemical features. Comparative analysis with reported X-ray crystallographic data further supported our revised assignments. These results resolve inconsistencies in earlier reports and highlight the critical importance of rigorous characterization in multicomponent heterocyclic synthesis. The findings establish a reliable framework for the identification of related heterocycles, thereby offering valuable guidance for future applications in drug-oriented synthesis and the design of bioactive molecules.

杂环支架的精确结构测定对药物化学和药物发现至关重要。在本研究中,对芳基异硫氰酸酯、二烷基乙炔二羧酸酯和水合肼的三组分反应以及含醛的四组分反应的产物进行了仔细的重新检查。以前的研究将这些化合物描述为噻嗪衍生物;然而,我们的综合合成和光谱研究表明,它们实际上对应于噻唑烷酮框架。梯度选择异核多键相关(HMBC)和门控13C核磁共振波谱提供了关键证据,可以确定区域选择性和立体化学特征。与已报道的x射线晶体学数据的比较分析进一步支持了我们修改后的作业。这些结果解决了早期报告中的不一致,并强调了在多组分杂环合成中严格表征的重要性。研究结果为相关杂环化合物的鉴定建立了可靠的框架,从而为未来在药物导向合成和生物活性分子设计中的应用提供了有价值的指导。
{"title":"Reevaluating 1,3-Thiazolidinone Reports: Deciphering the Authentic 1,3-Thiazolidinone Frameworks via Synthetic and Spectroscopic Insights","authors":"Roghayeh Hosseininia,&nbsp;Farough Nasiri","doi":"10.1002/ardp.70157","DOIUrl":"https://doi.org/10.1002/ardp.70157","url":null,"abstract":"<div>\u0000 \u0000 <p>Accurate structural determination of heterocyclic scaffolds is essential for medicinal chemistry and drug discovery. In this study, products obtained from three-component reactions of aryl isothiocyanates, dialkyl acetylenedicarboxylates, and hydrazine hydrate, as well as four-component reactions incorporating aldehydes, were carefully re-examined. Previous studies had described these compounds as thiazine derivatives; however, our comprehensive synthetic and spectroscopic investigations demonstrate that they actually correspond to thiazolidinone frameworks. Key evidence was provided by gradient-selected heteronuclear multiple bond correlation (HMBC) and gated <sup>13</sup>C NMR spectroscopy, which enabled the determination of regioselectivity and stereochemical features. Comparative analysis with reported X-ray crystallographic data further supported our revised assignments. These results resolve inconsistencies in earlier reports and highlight the critical importance of rigorous characterization in multicomponent heterocyclic synthesis. The findings establish a reliable framework for the identification of related heterocycles, thereby offering valuable guidance for future applications in drug-oriented synthesis and the design of bioactive molecules.</p></div>","PeriodicalId":128,"journal":{"name":"Archiv der Pharmazie","volume":"358 11","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145626413","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Mechanistic Insights into the Anticancer Potential of Echinacoside: Therapeutic Applications and Future Directions 紫锥菊苷抗癌潜力的机制研究:治疗应用及未来发展方向
IF 3.6 3区 医学 Q2 CHEMISTRY, MEDICINAL Pub Date : 2025-11-29 DOI: 10.1002/ardp.70153
Anju Sheokand, Dolly Koli, Karan Wadhwa, Gulshan Sharma, Rakesh Pahwa, Hardeep Singh Tuli

Echinacoside, a naturally occurring phenylethanoid glycoside, primarily derived from medicinal plants such as Cistanche tubulosa and Echinacea angustifolia, has long been valued in traditional medicine for its diverse pharmacological activities, including antidiabetic, anti-inflammatory, antifatigue, anti-allergic, antiaging, wound healing, and aphrodisiac effects. Recent research has steadily highlighted echinacoside's promising role as a multi-targeted oncotherapeutic agent due to its potent anticancer properties. Its antineoplastic efficacy is mediated through multiple mechanisms, including induction of apoptosis, inhibition of metastasis and angiogenesis, and modulation of key oncogenic signaling pathways such as PI3K/Akt/mTOR and MAPK/ERK. Additionally, echinacoside has significant synergistic potential with traditional chemotherapeutics, enhancing cytotoxicity while mitigating systemic toxicity and overcoming drug resistance. Despite compelling evidence for its multi-targeted anticancer mechanisms, its clinical translation is restricted by various pharmacokinetic challenges, including limited absorption and fast metabolic clearance. Although few reviews have addressed the general pharmacological activities of echinacoside, this article provides a novel and forward-looking perspective by critically bridging the gap between its multi-targeted anticancer mechanisms and its translational potential. This manuscript also comprehensively integrates the latest preclinical findings, particularly highlighting emerging nanotechnological and synergistic strategies designed to overcome its bioavailability and stability barrier, thereby uniquely outlining a strategic roadmap for future research to facilitate the clinical application of echinacoside into precision oncology.

紫锥菊苷是一种天然存在的苯乙醇类糖苷,主要来源于肉苁蓉和紫锥菊等药用植物,因其多种药理活性,包括抗糖尿病、抗炎、抗疲劳、抗过敏、抗衰老、伤口愈合和壮阳作用,长期以来在传统医学中受到重视。由于其有效的抗癌特性,最近的研究逐渐强调了紫锥菊苷作为一种多靶向肿瘤治疗剂的有希望的作用。其抗肿瘤作用是通过多种机制介导的,包括诱导细胞凋亡、抑制转移和血管生成,以及调节PI3K/Akt/mTOR和MAPK/ERK等关键的致癌信号通路。此外,紫锥菊苷与传统化疗药物具有显著的协同作用潜力,增强细胞毒性,减轻全身毒性,克服耐药。尽管有令人信服的证据表明其多靶点抗癌机制,但其临床转化受到各种药代动力学挑战的限制,包括有限的吸收和快速的代谢清除。尽管关于紫锥菊苷的一般药理活性的综述很少,但本文通过批判性地弥合其多靶点抗癌机制与其翻译潜力之间的差距,提供了一个新颖且具有前瞻性的视角。本文还全面整合了最新的临床前研究成果,特别强调了新兴的纳米技术和协同策略,旨在克服其生物利用度和稳定性障碍,从而独特地概述了未来研究的战略路线图,以促进紫锥菊苷在精准肿瘤学中的临床应用。
{"title":"Mechanistic Insights into the Anticancer Potential of Echinacoside: Therapeutic Applications and Future Directions","authors":"Anju Sheokand,&nbsp;Dolly Koli,&nbsp;Karan Wadhwa,&nbsp;Gulshan Sharma,&nbsp;Rakesh Pahwa,&nbsp;Hardeep Singh Tuli","doi":"10.1002/ardp.70153","DOIUrl":"https://doi.org/10.1002/ardp.70153","url":null,"abstract":"<div>\u0000 \u0000 <p>Echinacoside, a naturally occurring phenylethanoid glycoside, primarily derived from medicinal plants such as <i>Cistanche tubulosa</i> and <i>Echinacea angustifolia</i>, has long been valued in traditional medicine for its diverse pharmacological activities, including antidiabetic, anti-inflammatory, antifatigue, anti-allergic, antiaging, wound healing, and aphrodisiac effects. Recent research has steadily highlighted echinacoside's promising role as a multi-targeted oncotherapeutic agent due to its potent anticancer properties. Its antineoplastic efficacy is mediated through multiple mechanisms, including induction of apoptosis, inhibition of metastasis and angiogenesis, and modulation of key oncogenic signaling pathways such as PI3K/Akt/mTOR and MAPK/ERK. Additionally, echinacoside has significant synergistic potential with traditional chemotherapeutics, enhancing cytotoxicity while mitigating systemic toxicity and overcoming drug resistance. Despite compelling evidence for its multi-targeted anticancer mechanisms, its clinical translation is restricted by various pharmacokinetic challenges, including limited absorption and fast metabolic clearance. Although few reviews have addressed the general pharmacological activities of echinacoside, this article provides a novel and forward-looking perspective by critically bridging the gap between its multi-targeted anticancer mechanisms and its translational potential. This manuscript also comprehensively integrates the latest preclinical findings, particularly highlighting emerging nanotechnological and synergistic strategies designed to overcome its bioavailability and stability barrier, thereby uniquely outlining a strategic roadmap for future research to facilitate the clinical application of echinacoside into precision oncology.</p></div>","PeriodicalId":128,"journal":{"name":"Archiv der Pharmazie","volume":"358 11","pages":""},"PeriodicalIF":3.6,"publicationDate":"2025-11-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145626466","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"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
Archiv der Pharmazie
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