New Carbothioamide and Carboxamide Derivatives of 3-Phenoxybenzoic Acid as Potent VEGFR-2 Inhibitors: Synthesis, Molecular Docking, and Cytotoxicity Assessment.

IF 2.3 4区 医学 Q3 ONCOLOGY Current cancer drug targets Pub Date : 2024-05-13 DOI:10.2174/0115680096307334240429050730
Mohammad Hamza Heriz, Ammar A Razzak Mahmood, Lubna H Tahtamouni, Mai F AlSakhen, Sana I Kanaan, Khaled M Saleh, Salem R Yasin
{"title":"New Carbothioamide and Carboxamide Derivatives of 3-Phenoxybenzoic Acid as Potent VEGFR-2 Inhibitors: Synthesis, Molecular Docking, and Cytotoxicity Assessment.","authors":"Mohammad Hamza Heriz, Ammar A Razzak Mahmood, Lubna H Tahtamouni, Mai F AlSakhen, Sana I Kanaan, Khaled M Saleh, Salem R Yasin","doi":"10.2174/0115680096307334240429050730","DOIUrl":null,"url":null,"abstract":"<p><strong>Introduction/background: </strong>Because of the well-established link between angiogenesis and tumor development, the use of antiangiogenic therapeutics, such as those targeting VEGFR-2, presents a promising approach to cancer treatment. In the current study, a set of five hydrazine-1-- carbothioamide (compounds 3a-e) and three hydrazine-1-carboxamide derivatives (compounds 4a-c) were successfully synthesized from 3-phenoxybenzoic acid. These compounds were specially created as antiproliferative agents with the goal of targeting cancer cells by inhibiting VEGFR-2 tyrosine kinase.</p><p><strong>Materials and methods: </strong>The new derivatives were synthesized by conventional organic methods, and their structure was versified by IR, 1HNMR, 13CNMR, and mass spectroscopy. In silico investigation was carried out to identify the compounds' target, molecular similarity, ADMET, and toxicity profile. The cytotoxic activity of the prepared compounds was evaluated in vitro against three human cancer cell lines (DLD1 colorectal adenocarcinoma, HeLa cervical cancer, and HepG2 hepatocellular carcinoma). The effects of the leading compound on cell cycle progression and apoptosis induction were investigated by flow cytometry, and the specific apoptotic pathway triggered by the treatment was evaluated by RT-PCR and immunoblotting. Finally, the inhibitory activities of the new compounds against VEGFR-2 was measured.</p><p><strong>Results: </strong>The designed derivatives exhibited comparable binding positions and interactions to the VEGFR-2 binding site to that of sorafenib (a standard VEGFR-2 tyrosine kinase inhibitor), as determined by molecular docking analysis. Compound 4b was the most cytotoxic compound, achieving the lowest IC50 against HeLa cells. Compound 4b, a strong representative of the synthesized series, induced cell cycle arrest at the G2/M phase, increased the proportion of necrotic and apoptotic HeLa cells, and activated caspase 3. The EC50 value of compound 4b against VEGFR-2 kinase activity was comparable to sorafenib's.</p><p><strong>Conclusion: </strong>Overall, the findings suggest that compound 4b has a promising future as a starting point for the development of new anticancer drugs.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3000,"publicationDate":"2024-05-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Current cancer drug targets","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.2174/0115680096307334240429050730","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Introduction/background: Because of the well-established link between angiogenesis and tumor development, the use of antiangiogenic therapeutics, such as those targeting VEGFR-2, presents a promising approach to cancer treatment. In the current study, a set of five hydrazine-1-- carbothioamide (compounds 3a-e) and three hydrazine-1-carboxamide derivatives (compounds 4a-c) were successfully synthesized from 3-phenoxybenzoic acid. These compounds were specially created as antiproliferative agents with the goal of targeting cancer cells by inhibiting VEGFR-2 tyrosine kinase.

Materials and methods: The new derivatives were synthesized by conventional organic methods, and their structure was versified by IR, 1HNMR, 13CNMR, and mass spectroscopy. In silico investigation was carried out to identify the compounds' target, molecular similarity, ADMET, and toxicity profile. The cytotoxic activity of the prepared compounds was evaluated in vitro against three human cancer cell lines (DLD1 colorectal adenocarcinoma, HeLa cervical cancer, and HepG2 hepatocellular carcinoma). The effects of the leading compound on cell cycle progression and apoptosis induction were investigated by flow cytometry, and the specific apoptotic pathway triggered by the treatment was evaluated by RT-PCR and immunoblotting. Finally, the inhibitory activities of the new compounds against VEGFR-2 was measured.

Results: The designed derivatives exhibited comparable binding positions and interactions to the VEGFR-2 binding site to that of sorafenib (a standard VEGFR-2 tyrosine kinase inhibitor), as determined by molecular docking analysis. Compound 4b was the most cytotoxic compound, achieving the lowest IC50 against HeLa cells. Compound 4b, a strong representative of the synthesized series, induced cell cycle arrest at the G2/M phase, increased the proportion of necrotic and apoptotic HeLa cells, and activated caspase 3. The EC50 value of compound 4b against VEGFR-2 kinase activity was comparable to sorafenib's.

Conclusion: Overall, the findings suggest that compound 4b has a promising future as a starting point for the development of new anticancer drugs.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
作为强效 VEGFR-2 抑制剂的 3-苯氧基苯甲酸的新硫代甲酰胺和甲酰胺衍生物:合成、分子对接和细胞毒性评估。
导言/背景:由于血管生成与肿瘤发生之间的联系已得到证实,因此使用抗血管生成治疗药物(如靶向 VEGFR-2 的药物)是一种很有前景的癌症治疗方法。本研究以 3-苯氧基苯甲酸为原料,成功合成了五种肼-1-硫代甲酰胺(化合物 3a-e)和三种肼-1-甲酰胺衍生物(化合物 4a-c)。这些化合物专门用作抗增殖剂,目的是通过抑制血管内皮生长因子受体-2(VEGFR-2)酪氨酸激酶来靶向治疗癌细胞:采用传统有机方法合成了新的衍生物,并通过红外光谱、1HNMR、13CNMR 和质谱分析了它们的结构。为了确定化合物的靶标、分子相似性、ADMET 和毒性特征,进行了硅学研究。在体外评估了所制备化合物对三种人癌细胞株(DLD1 大肠腺癌、HeLa 宫颈癌和 HepG2 肝癌)的细胞毒活性。流式细胞术研究了前导化合物对细胞周期进展和凋亡诱导的影响,RT-PCR 和免疫印迹法评估了治疗引发的特异性凋亡途径。最后,测定了新化合物对 VEGFR-2 的抑制活性:分子对接分析表明,所设计的衍生物与索拉非尼(一种标准的血管内皮生长因子受体-2酪氨酸激酶抑制剂)在血管内皮生长因子受体-2结合位点的结合位置和相互作用相似。化合物 4b 是细胞毒性最强的化合物,对 HeLa 细胞的 IC50 值最低。化合物 4b 是合成系列中的强势代表,能诱导细胞周期停滞在 G2/M 期,增加 HeLa 细胞坏死和凋亡的比例,并激活 caspase 3。化合物 4b 对 VEGFR-2 激酶活性的 EC50 值与索拉非尼相当:总之,研究结果表明,化合物 4b 作为开发新型抗癌药物的起点前景广阔。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
Current cancer drug targets
Current cancer drug targets 医学-肿瘤学
CiteScore
5.40
自引率
0.00%
发文量
105
审稿时长
1 months
期刊介绍: Current Cancer Drug Targets aims to cover all the latest and outstanding developments on the medicinal chemistry, pharmacology, molecular biology, genomics and biochemistry of contemporary molecular drug targets involved in cancer, e.g. disease specific proteins, receptors, enzymes and genes. Current Cancer Drug Targets publishes original research articles, letters, reviews / mini-reviews, drug clinical trial studies and guest edited thematic issues written by leaders in the field covering a range of current topics on drug targets involved in cancer. As the discovery, identification, characterization and validation of novel human drug targets for anti-cancer drug discovery continues to grow; this journal has become essential reading for all pharmaceutical scientists involved in drug discovery and development.
期刊最新文献
Screening miRNAs to Hinder the Tumorigenesis of Renal Clear Cell Carcinoma Associated with KDR Expression Dendrobine Suppresses Tumor Growth by Regulating the PD-1/PD-L1 Checkpoint Pathway in Lung Cancer. Magnesium as a Co-Factor: A Vital Cation with Pro- and Anti-Tumor Effects. Progressive Dynamics of Cancer Stem Cells in Oral Squamous Cell Carcinoma. SELENBP1 Inhibits the Warburg Effect and Tumor Growth by Reducing the HIF1α Expression in Colorectal Cancer.
×
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
现在去查看 取消
×
提示
确定
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
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
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