Anti-cancer agents in medicinal chemistry最新文献

筛选
英文 中文
Targeting Ferroptosis: Small-molecule Inducers as Novel Anticancer Agents. 靶向铁突变:作为新型抗癌药物的小分子诱导剂
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2024-10-15 DOI: 10.2174/0118715206342278241008081126
Shihao Jin, Huannan Wang, Zhen Zhang, Maocai Yan
{"title":"Targeting Ferroptosis: Small-molecule Inducers as Novel Anticancer Agents.","authors":"Shihao Jin, Huannan Wang, Zhen Zhang, Maocai Yan","doi":"10.2174/0118715206342278241008081126","DOIUrl":"https://doi.org/10.2174/0118715206342278241008081126","url":null,"abstract":"<p><p>Ferroptosis, a distinct form of regulated cell death characterized by iron-dependent lipid peroxidation and reactive oxygen species (ROS) accumulation, is increasingly recognized for its role in cancer development and as a potential therapeutic target. This review consolidates insights into the molecular mechanisms underpinning ferroptosis and evaluates the therapeutic potential of small-molecule inducers, such as erastin, RSL3, sulfasalazine, and sorafenib, which selectively trigger ferroptosis in cancer cells. It highlights the distinct morphological and molecular signatures of ferroptosis, its complex interplay with iron, lipid, and amino acid metabolic pathways, and the resultant implications for cancer treatment strategies. Strategic manipulation of the ferroptosis pathway offers a groundbreaking approach to cancer treatment, potentially circumventing the resistance that cancers develop against traditional apoptosis-inducing agents. Furthermore, it also emphasizes the necessity of refining these small molecules for clinical application and exploring their synergistic potential when combined with current therapies to augment overall treatment efficacy and improve patient outcomes. Ferroptosis thus emerges as a promising avenue in the realm of cancer therapy. Moving forward, research endeavors should focus on a more nuanced understanding of the interconnections between ferroptosis and other cell death modalities. Additionally, comprehensive evaluations of the long-term safety and therapeutic indices of the involved compounds are imperative. Such investigations are poised to herald a transformative shift in the paradigm of oncology, paving the way for innovative and targeted interventions.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142456293","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
Anticancer Properties Against Select Cancer Cell Lines and Metabolomics Analysis of Tender Coconut Water. 嫩椰子水对部分癌症细胞株的抗癌特性及代谢组学分析
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2024-10-15 DOI: 10.2174/0118715206327789241008162423
Jaganathan Lakshmanan, Vaitheesh L Jaganathan, Boachun Zhang, Grace Werner, Tyler S Allen, David J Schultz, Carolyn M Klinge, Brian G Harbrecht
{"title":"Anticancer Properties Against Select Cancer Cell Lines and Metabolomics Analysis of Tender Coconut Water.","authors":"Jaganathan Lakshmanan, Vaitheesh L Jaganathan, Boachun Zhang, Grace Werner, Tyler S Allen, David J Schultz, Carolyn M Klinge, Brian G Harbrecht","doi":"10.2174/0118715206327789241008162423","DOIUrl":"https://doi.org/10.2174/0118715206327789241008162423","url":null,"abstract":"<p><strong>Background: </strong>Tender Coconut Water (TCW) is a nutrient-rich dietary supplement that contains in bioactive secondary metabolites and phytohormones with anti-oxidative and anti-inflammatory properties. Studies on TCW's anti-cancer properties are limited and the mechanism of its anti-cancer effects have not been defined.</p><p><strong>Objective: </strong>In the present study, we investigate TCW for its anti-cancer properties and, using untargeted metabolomics, we identify components form TCW with potential anti-cancer activity.</p><p><strong>Methodology: </strong>Cell viability assay, BrdU incorporation assay, soft-agar assay, flow-cytometery, and Western blotting were used to analyze TCW's anticancer properties and to identify mechanism of action. Liquid chromatography- Tandem Mass Spectroscopy (LC-MS/MS) was used to identify TCW components.</p><p><strong>Results: </strong>TCW decreased the viability and anchorage-independent growth of HepG2 hepatocellular carcinoma (HCC) cells and caused S-phase cell cycle arrest. TCW inhibited AKT and ERK phosphorylation leading to reduced ZEB1 protein, increased E-cadherin, and reduced N-cadherin protein expression in HepG2 cells, thus reversing the 'epithelial-to-mesenchymal' (EMT) transition. TCW also decreased the viability of Hep3B hepatoma, HCT-15 colon, MCF-7 and T47D luminal A breast cancer (BC) and MDA-MB-231 and MDA-MB-468 triplenegative BC cells. Importantly, TCW did not inhibit the viability of MCF-10A normal breast epithelial cells. Untargeted metabolomics analysis of TCW identified 271 metabolites, primarily lipids and lipid-like molecules, phenylpropanoids and polyketides, and organic oxygen compounds. We demonstrate that three components from TCW: 3-hydroxy-1-(4-hydroxyphenyl)propan-1-one, iondole-3-carbox aldehyde and caffeic acid inhibit the growth of cancer cells.</p><p><strong>Conclusion: </strong>TCW and its components exhibit anti-cancer effects. TCW inhibits the viability of HepG2 hepatocellular carcinoma cells by reversing the EMT process through inhibition of AKT and ERK signalling.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2024-10-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142456330","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 Prediction of Pranlukast as a Stabilizer of PD-L1 Homodimers. 对 Pranlukast 作为 PD-L1 同源二聚体稳定剂的硅学预测
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2024-10-14 DOI: 10.2174/0118715206303675241009104647
Luis Córdova-Bahena, Carlos Landero-Marín, Xcaret Flores-Hernández, Leonardo Daniel Alvarez-Coronel, Alexis Paulina Jiménez-Uribe, Nohemí Salinas-Jazmín, Zhiqiang An, Marco Velasco-Velázquez
{"title":"In silico Prediction of Pranlukast as a Stabilizer of PD-L1 Homodimers.","authors":"Luis Córdova-Bahena, Carlos Landero-Marín, Xcaret Flores-Hernández, Leonardo Daniel Alvarez-Coronel, Alexis Paulina Jiménez-Uribe, Nohemí Salinas-Jazmín, Zhiqiang An, Marco Velasco-Velázquez","doi":"10.2174/0118715206303675241009104647","DOIUrl":"https://doi.org/10.2174/0118715206303675241009104647","url":null,"abstract":"<p><strong>Introduction: </strong>Tumors can be targeted by modulating the immune response of the patient. Programmed cell death protein 1 (PD-1) and programmed cell death ligand 1 (PD-L1) are critical immune checkpoints in cancer biology. The efficacy of certain cancer immunotherapies has been achieved by targeting these molecules using monoclonal antibodies.</p><p><strong>Method: </strong>Small-molecule drugs have also been developed as inhibitors of the PD-1/PD-L1 axis, with a mechanism of action that is distinct from that of antibodies: they induce the formation of PD-L1 homodimers, causing their stabilization, internalization, and subsequent degradation. Drug repurposing is a strategy in which new uses are sought after for approved drugs, expediting their clinical translation based on updated findings. In this study, we generated a pharmacophore model that was based on reported small molecules that targeted PD-L1 and used it to identify potential PD-L1 inhibitors among FDA-approved drugs.</p><p><strong>Results: </strong>We identified 12 pharmacophore-matching compounds, but only 4 reproduced the binding mode of the reference inhibitors in docking experiments. Further characterization by molecular dynamics showed that pranlukast, an antagonist of leukotriene receptors that is used to treat asthma, generated stable and energyfavorable interactions with PD-L1 homodimers and induced homodimerization of recombinant PD-L1.</p><p><strong>Conclusion: </strong>Our results suggest that pranlukast inhibits the PD-1/PD-L1 axis, meriting its repurposing as an antitumor drug.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2024-10-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142456332","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
Urea and Thiourea Derivatives of Salinomycin as Agents Targeting Malignant Colon Cancer Cells. 盐霉素的尿素和硫脲衍生物作为靶向恶性结肠癌细胞的药物
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2024-10-10 DOI: 10.2174/0118715206322603241002064435
Michał Antoszczak, Magdalena Mielczarek-Puta, Marta Struga, Adam Huczyński
{"title":"Urea and Thiourea Derivatives of Salinomycin as Agents Targeting Malignant Colon Cancer Cells.","authors":"Michał Antoszczak, Magdalena Mielczarek-Puta, Marta Struga, Adam Huczyński","doi":"10.2174/0118715206322603241002064435","DOIUrl":"https://doi.org/10.2174/0118715206322603241002064435","url":null,"abstract":"<p><strong>Background: </strong>Since it was discovered that a natural polyether ionophore called salinomycin (SAL) selectively inhibits human cancer cells, the scientific world has been paying special attention to this compound. It has been studied for nearly 15 years.</p><p><strong>Objective: </strong>Thus, a very interesting research direction is the chemical modification of SAL structure, which could give more biologically active agents.</p><p><strong>Methods: </strong>We evaluated the anticancer activity of (thio)urea analogues class of C20-epi-aminosalinomycin (compound 3b). The studies covered the generation of reactive oxygen species (ROS), proapoptotic activity, cytotoxic activity, and lipid peroxidation in vitro.</p><p><strong>Results: </strong>Thioureas 5a‒5d showed antiproliferative activity against selected human colon cancer cell lines greater than that of chemically unmodified SAL, with a 2~10-fold higher potency towards a metastatic variant of colon cancer cells (SW620). Mechanistically, SAL derivatives showed pro-apoptotic activity in primary colon cancer cells and induced the production of reactive oxygen species (ROS) in these cells. In SW620 cells, SAL derivatives increased lipid peroxidation with a weak effect on apoptosis and low ROS formation with cytotoxic effects followed by cytostatic ones, suggesting different modes of action of the compounds against primary and metastatic colon cancer cells.</p><p><strong>Conclusion: </strong>The results of this study suggested that urea and thiourea derivatives of SAL provide promising leads for the rational development of new anticancer active agents.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142399180","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
Therapeutic Effects of Crocin Nanoparticles Alone or in Combination with Doxorubicin against Hepatocellular Carcinoma In vitro. Crocin 纳米粒子单独或与多柔比星联合使用对肝细胞癌的体外治疗效果。
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2024-10-09 DOI: 10.2174/0118715206327654240823074318
Noha S Basuony, Tarek M Mohamed, Doha M Beltagy, Ahmed A Massoud, Mona M Elwan
{"title":"Therapeutic Effects of Crocin Nanoparticles Alone or in Combination with Doxorubicin against Hepatocellular Carcinoma In vitro.","authors":"Noha S Basuony, Tarek M Mohamed, Doha M Beltagy, Ahmed A Massoud, Mona M Elwan","doi":"10.2174/0118715206327654240823074318","DOIUrl":"https://doi.org/10.2174/0118715206327654240823074318","url":null,"abstract":"<p><strong>Objective: </strong>Crocin (CRO), the primary antioxidant in saffron, is known for its anticancer properties. However, its effectiveness in topical therapy is limited due to low bioavailability, poor absorption, and low physicochemical stability. This study aimed to prepare crocin nanoparticles (CRO-NPs) to enhance their pharmaceutical efficacy and evaluate the synergistic effects of Cro-NPs with doxorubicin (DOX) chemotherapy on two cell lines: human hepatocellular carcinoma cells (HepG2) and non-cancerous cells (WI38).</p><p><strong>Methods: </strong>CRO-NPs were prepared using the emulsion diffusion technique and characterized by transmission electron microscopy (TEM), scanning electron microscopy (SEM), Zeta potential, and Fourier transform infrared spectroscopy (FT-IR). Cell proliferation inhibition was assessed using the MTT assay for DOX, CRO, CRO-NPs, and DOX+CRO-NPs. Apoptosis and cell cycle were evaluated by flow cytometry, and changes in the expression of apoptotic gene (P53) and autophagic genes (ATG5 & LC3) were analyzed using real-time polymerase chain reaction.</p><p><strong>Results: </strong>TEM and SEM revealed that CRO-NPs exhibited a relatively spherical shape with an average size of 9.3 nm, and zeta potential analysis indicated better stability of CRO-NPs compared to native CRO. Significantly higher antitumor effects of CRO-NPs were observed against HepG2 cells (IC50 = 1.1 mg/ml and 0.57 mg/ml) compared to native CRO (IC50 = 6.1 mg/ml and 3.2 mg/ml) after 24 and 48 hours, respectively. Annexin-V assay on HepG2 cells indicated increased apoptotic rates across all treatments, with the highest percentage observed in CRO-NPs, accompanied by cell cycle arrest at the G2/M phase. Furthermore, gene expression analysis showed upregulation of P53, ATG5, and LC3 genes in DOX/CRO-NPs co-treatment compared to individual treatments. In contrast, WI38 cells exhibited greater sensitivity to DOX toxicity but showed no adverse response to CRONPs.</p><p><strong>Conclusion: </strong>Although more in vivo studies in animal models are required to corroborate these results, our findings suggest that CRO-NPs can be a potential new anticancer agent for hepatocellular carcinoma. Moreover, they have a synergistic effect with DOX against HepG2 cells and mitigate the toxicity of DOX on normal WI38 cells.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142456294","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
Emerging Claudin18.2-targeting Therapy for Systemic Treatment of Gastric Cancer: Seeking Nobility Amidst Danger. 用于胃癌全身治疗的新兴 Claudin18.2 靶向疗法:险中求胜。
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2024-10-03 DOI: 10.2174/0118715206329892240927081033
Xueshuai Ye, Yongqiang Wu, Haiqiang Zhang
{"title":"Emerging Claudin18.2-targeting Therapy for Systemic Treatment of Gastric Cancer: Seeking Nobility Amidst Danger.","authors":"Xueshuai Ye, Yongqiang Wu, Haiqiang Zhang","doi":"10.2174/0118715206329892240927081033","DOIUrl":"https://doi.org/10.2174/0118715206329892240927081033","url":null,"abstract":"<p><p>Gastric cancer in advanced stages lacked effective treatment options. claudin18.2 (CLDN18.2) is a membrane protein that is crucial for close junctions in the differentiated epithelial cells of the gastric mucosa, playing a vital role in barrier function, and can be hardly recognized by immune cells due to its polarity pattern. As the polarity of gastric tumor cells changes, claudin18.2 is exposed on the cell surface, resulting in immune system recognition, and making it an ideal target. In this review, we summarized the expression regulation mechanism of claudin18.2 both in normal cells and malignant tumor cells. Besides, we analyzed the available clinical results and potential areas for future research on claudin18.2-positive gastric cancer and claudin18.2-targeting therapy. In conclusion, claudin18.2 is an ideal target for gastric cancer treatment, and the claudin18.2-targeting therapy has changed the treatment pattern of gastric cancer.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142370794","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
MG132-mediated Suppression of the Ubiquitin-proteasome Pathway Enhances the Sensitivity of Endometrial Cancer Cells to Cisplatin. MG132- 介导的泛素-蛋白酶体通路抑制增强了子宫内膜癌细胞对顺铂的敏感性
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2024-10-01 DOI: 10.2174/0118715206343550240919055701
Zhanhu Zhang, Yiqian Ding
{"title":"MG132-mediated Suppression of the Ubiquitin-proteasome Pathway Enhances the Sensitivity of Endometrial Cancer Cells to Cisplatin.","authors":"Zhanhu Zhang, Yiqian Ding","doi":"10.2174/0118715206343550240919055701","DOIUrl":"https://doi.org/10.2174/0118715206343550240919055701","url":null,"abstract":"<p><strong>Background: </strong>Tumor cell resistance to cisplatin is a common challenge in endometrial cancer chemotherapy, stemming from various mechanisms. Targeted therapies using proteasome inhibitors, such as MG132, have been investigated to enhance cisplatin sensitivity, potentially offering a novel treatment approach.</p><p><strong>Objective: </strong>The aim of this study was to investigate the effects of MG132 on cisplatin sensitivity in the human endometrial cancer (EC) cell line RL95-2, focusing on cell proliferation, apoptosis, and cell signaling.</p><p><strong>Methods: </strong>Human endometrial cancer RL95-2 cells were exposed to MG132, and cell viability was assessed in a dose-dependent manner. The study evaluated the effect of MG132 on cisplatin-induced proliferation inhibition and apoptosis, correlating with caspase-3 activation and reactive oxygen species (ROS) upregulation. Additionally, we examined the inhibition of the ubiquitin-proteasome system and the expression of pro-inflammatory cytokines IL-1β, IL-6, IL-8, and IL-13 during MG132 and cisplatin co-administration.</p><p><strong>Results: </strong>MG132 exposure significantly reduced cell viability in a dose-dependent manner. It augmented cisplatin- induced proliferation inhibition and enhanced apoptosis, correlating with caspase-3 activation and ROS upregulation. Molecular analysis revealed a profound inhibition of the ubiquitin-proteasome system. MG132 also significantly increased the expression of cisplatin-induced pro-inflammatory cytokines, suggesting a transition from chronic to acute inflammation.</p><p><strong>Conclusion: </strong>MG132 enhances the therapeutic efficacy of cisplatin in human EC cells by suppressing the ubiquitin- proteasome pathway, reducing cell viability, enhancing apoptosis, and shifting the inflammatory response. These findings highlighted the potential of MG132 as an adjuvant in endometrial cancer chemotherapy. Further research is needed to explore detailed mechanisms and clinical applications of this combination therapy.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142360951","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
Novel Quinoline Nitrate Derivatives: Synthesis, Characterization, and Evaluation of their Anticancer Activity with a Focus on Molecular Docking and NO Release. 新型硝酸喹啉衍生物:合成、表征及其抗癌活性评估,重点关注分子对接和 NO 释放。
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2024-10-01 DOI: 10.2174/0118715206315415240830052608
Venkata Sowjanya Thanneeru, Naresh Panigrahi
{"title":"Novel Quinoline Nitrate Derivatives: Synthesis, Characterization, and Evaluation of their Anticancer Activity with a Focus on Molecular Docking and NO Release.","authors":"Venkata Sowjanya Thanneeru, Naresh Panigrahi","doi":"10.2174/0118715206315415240830052608","DOIUrl":"https://doi.org/10.2174/0118715206315415240830052608","url":null,"abstract":"<p><strong>Background: </strong>Nitric Oxide (NO) has recently gained recognition as a promising approach in the field of cancer therapy. The quinoline scaffold is pivotal in cancer drug research and is known for its versatility and diverse mechanisms of action.</p><p><strong>Objective: </strong>This study presents the synthesis, characterization, and evaluation of novel quinoline nitrate derivatives as potential anticancer agents.</p><p><strong>Methods: </strong>The compounds were synthesized through a multi-step process involving the preparation of substituted 1-(2-aminophenyl) ethan-1-one, followed by the synthesis of substituted 2- (chloromethyl)-3,4-dimethylquinolines, and finally, the formation of substituted (3,4- dimethylquinolin-2-yl) methyl nitrate derivatives. The synthesized compounds were characterized using various spectroscopic techniques. Molecular docking studies were conducted to assess the binding affinity of the compounds to the EGFR tyrosine kinase domain.</p><p><strong>Results: </strong>The docking scores revealed varying degrees of binding affinity, with compound 6k exhibiting the highest score. The results suggested a correlation between molecular docking scores and anticancer activity. Further evaluations included MTT assays to determine the cytotoxicity of the compounds against Non-Small Cell Lung Cancer (A-549) and pancreatic cancer (PANC-1) cell lines. Compounds with electron-donating groups displayed notable anticancer potential, and there was a correlation between NO release and anticancer activity. The study also investigated nitric oxide release from the compounds, revealing compound 6g as the highest NO releaser.</p><p><strong>Conclusion: </strong>The synthesized quinoline nitrate derivatives showed promising anticancer activity, with compound 6g standing out as a potential lead compound. The correlation between molecular docking, NO release, and anticancer activity suggests the importance of specific structural features in the design of effective anticancer agents.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142360952","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
Advancements in Pyrazine Derivatives as Anticancer Agents: A Comprehensive Review (2010-2024). 作为抗癌剂的吡嗪衍生物的研究进展:全面回顾(2010-2024 年)》。
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2024-09-24 DOI: 10.2174/0118715206333399240912071555
Mohammed Merae Alshahrani
{"title":"Advancements in Pyrazine Derivatives as Anticancer Agents: A Comprehensive Review (2010-2024).","authors":"Mohammed Merae Alshahrani","doi":"10.2174/0118715206333399240912071555","DOIUrl":"https://doi.org/10.2174/0118715206333399240912071555","url":null,"abstract":"<p><p>Cancer, an intricate and formidable disease, continues to challenge Medical Science with its diverse manifestations and relentless progression. In the pursuit of novel therapeutic strategies, organic heterocyclic compounds have emerged as promising candidates due to their versatile chemical structures and intricate interactions with biological systems. Among these, pyrazine derivatives are characterized by a six-membered aromatic ring containing four carbon and two nitrogen atoms situated in a 1,4-orientation. These compounds garnered significant attention for their potential as anticancer agents. This comprehensive review provides a detailed analysis of the advancements made during this timeframe, encompassing the chemical diversity of pyrazine derivatives, their mechanisms of action at the cellular level, and structure-activity relationships, spanning the years 2010 to 2024. By examining their therapeutic potential, challenges, and future prospects, this review offers valuable insights into the evolving landscape of pyrazine derivatives as potent tools in the fight against cancer.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2024-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142339474","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
Acyl Urea Compounds Therapeutics and its Inhibition for Cancers in Women: A Review. 酰基尿素化合物疗法及其对女性癌症的抑制作用:综述。
IF 2.6 4区 医学
Anti-cancer agents in medicinal chemistry Pub Date : 2024-09-24 DOI: 10.2174/0118715206330232240913100744
Preeti Kumari, Rakhi Mishra, Rupa Mazumder, Avijit Mazumder
{"title":"Acyl Urea Compounds Therapeutics and its Inhibition for Cancers in Women: A Review.","authors":"Preeti Kumari, Rakhi Mishra, Rupa Mazumder, Avijit Mazumder","doi":"10.2174/0118715206330232240913100744","DOIUrl":"https://doi.org/10.2174/0118715206330232240913100744","url":null,"abstract":"<p><p>Acyl urea compounds have garnered significant attention in cancer therapeutics, particularly for their potential effectiveness against cancers that predominantly affect women, such as breast and ovarian cancers. The paper presents a report on the investigation of acyl urea compounds that are reported to involve a multi-faceted approach, including synthetic chemistry, biological assays, and computational modeling. A wealth of information on acyl urea and its purported effects on cancer affecting women has been gathered from different sources and condensed to provide readers with a broad understanding of the role of acyl urea in combating cancer. Acylureas demonstrate promising results by selectively inhibiting key molecular targets associated with cancer progressions, such as EGFR, ALK, HER2, and the Wnt/β-catenin signaling pathway. Specifically, targeting acyl ureas impedes tumor proliferation and metastasis while minimizing harm to healthy tissues, offering a targeted therapeutic approach with reduced side effects compared to conventional chemotherapy. Continued research and clinical trials are imperative to optimize the efficacy and safety profiles of acylurea-based therapies and broaden their applicability across various cancer types. Acyl urea compounds represent a promising class of therapeutics for the treatment of cancers in women, particularly due to their ability to selectively inhibit key molecular targets involved in tumor growth and progression. The combination of synthetic optimization, biological evaluation, and computational modeling has facilitated the identification of several lead compounds with significant anticancer potential. This abstract explores the therapeutic mechanisms and targeted pathways of acyl ureas in combating these malignancies, which will be useful for future studies.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2024-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142339473","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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信