新型查尔酮类似物作为去势抵抗性前列腺癌潜在多靶点治疗的进展

O. Hussein, F. Alali, Ala‐Eddin Al Mustafa, Ashraf Khalil
{"title":"新型查尔酮类似物作为去势抵抗性前列腺癌潜在多靶点治疗的进展","authors":"O. Hussein, F. Alali, Ala‐Eddin Al Mustafa, Ashraf Khalil","doi":"10.29117/quarfe.2021.0114","DOIUrl":null,"url":null,"abstract":"Prostate cancer (PCa) is the second most frequently diagnosed malignancy, as well as a leading cause of cancer-related mortality in men globally. Despite the initial response to hormonal targeted therapy, the majority of patients ultimately progress to a lethal form of the disease, castration-resistant prostate cancer (CRPC). Therefore, the objective of this study was to discover and develop novel treatment modalities for CRPC. Chalcones are among the highly attractive scaffolds being investigated for their antitumor activities. A library of 26 chalcone analogs were designed, synthesized and evaluated as potential therapies for CRPC. The design was guided by in-silico ADMET prediction in which analogs with favorable drug-likeness properties were prioritized. The new compounds were synthesized, purified and characterized by extensive structural elucidation studies. The compounds in vitro cytotoxicity was evaluated against two androgen receptor (AR)-negative prostate cancer cell lines (PC3 and DU145). Among the tested compounds, pyridine containing analogs (13, 15 and 16) showed potent antiproliferative activities with IC50 values ranging between 4.32-6.47 µM against PC3 and DU145 cell lines. Detailed biological studies of the lead molecule 16 revealed that it can significantly induce apoptosis through upregulation of Bax and downregulation of Bcl-2. In addition, compound 16 potently inhibited colony formation and reduced cell migration of AR-negative PCa cell lines (PC3 and DU145). The molecular pathway analysis showed that the anticancer activity of compound 16 is associated with blocking of ERK1/2 and Akt activities. Furthermore, compound 16 inhibited angiogenesis in the chick chorioallantoic membrane (CAM) model as compared to control. Structure-activity relationship study revealed that the cytotoxicity could dramatically improve via changing the methoxylation pattern by more than 2-folds (IC50 << 2.5 μM). These results indicate that pyridine-based chalcones could serve as promising lead molecules for the treatment of CRPC; thus, further in vitro and in vivo studies are warranted.","PeriodicalId":9295,"journal":{"name":"Building Resilience at Universities: Role of Innovation and Entrepreneurship","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2021-10-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":"{\"title\":\"Development of Novel Chalcone Analogs as Potential Multi-Targeted Therapies for Castration-Resistant Prostate Cancer\",\"authors\":\"O. Hussein, F. Alali, Ala‐Eddin Al Mustafa, Ashraf Khalil\",\"doi\":\"10.29117/quarfe.2021.0114\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"Prostate cancer (PCa) is the second most frequently diagnosed malignancy, as well as a leading cause of cancer-related mortality in men globally. Despite the initial response to hormonal targeted therapy, the majority of patients ultimately progress to a lethal form of the disease, castration-resistant prostate cancer (CRPC). Therefore, the objective of this study was to discover and develop novel treatment modalities for CRPC. Chalcones are among the highly attractive scaffolds being investigated for their antitumor activities. A library of 26 chalcone analogs were designed, synthesized and evaluated as potential therapies for CRPC. The design was guided by in-silico ADMET prediction in which analogs with favorable drug-likeness properties were prioritized. The new compounds were synthesized, purified and characterized by extensive structural elucidation studies. The compounds in vitro cytotoxicity was evaluated against two androgen receptor (AR)-negative prostate cancer cell lines (PC3 and DU145). Among the tested compounds, pyridine containing analogs (13, 15 and 16) showed potent antiproliferative activities with IC50 values ranging between 4.32-6.47 µM against PC3 and DU145 cell lines. Detailed biological studies of the lead molecule 16 revealed that it can significantly induce apoptosis through upregulation of Bax and downregulation of Bcl-2. In addition, compound 16 potently inhibited colony formation and reduced cell migration of AR-negative PCa cell lines (PC3 and DU145). The molecular pathway analysis showed that the anticancer activity of compound 16 is associated with blocking of ERK1/2 and Akt activities. Furthermore, compound 16 inhibited angiogenesis in the chick chorioallantoic membrane (CAM) model as compared to control. Structure-activity relationship study revealed that the cytotoxicity could dramatically improve via changing the methoxylation pattern by more than 2-folds (IC50 << 2.5 μM). These results indicate that pyridine-based chalcones could serve as promising lead molecules for the treatment of CRPC; thus, further in vitro and in vivo studies are warranted.\",\"PeriodicalId\":9295,\"journal\":{\"name\":\"Building Resilience at Universities: Role of Innovation and Entrepreneurship\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":0.0000,\"publicationDate\":\"2021-10-20\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Building Resilience at Universities: Role of Innovation and Entrepreneurship\",\"FirstCategoryId\":\"1085\",\"ListUrlMain\":\"https://doi.org/10.29117/quarfe.2021.0114\",\"RegionNum\":0,\"RegionCategory\":null,\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"\",\"JCRName\":\"\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Building Resilience at Universities: Role of Innovation and Entrepreneurship","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.29117/quarfe.2021.0114","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

摘要

前列腺癌(PCa)是第二常见的恶性肿瘤,也是全球男性癌症相关死亡的主要原因。尽管最初对激素靶向治疗有反应,但大多数患者最终进展为一种致命的疾病,去势抵抗性前列腺癌(CRPC)。因此,本研究的目的是发现和发展CRPC的新治疗方式。查尔酮因其抗肿瘤活性而成为备受关注的支架材料之一。设计、合成了26个查尔酮类似物,并对其作为CRPC的潜在治疗药物进行了评价。该设计以计算机ADMET预测为指导,其中具有良好药物相似特性的类似物被优先考虑。这些新化合物经过合成、纯化并进行了广泛的结构解析研究。对两种雄激素受体(AR)阴性的前列腺癌细胞株PC3和DU145进行了体外细胞毒性评价。在这些化合物中,含有吡啶的类似物(13、15和16)对PC3和DU145细胞株具有较强的抗增殖活性,IC50值在4.32 ~ 6.47µM之间。对铅分子的详细生物学研究表明,其可通过上调Bax和下调Bcl-2显著诱导细胞凋亡。此外,化合物16还能有效抑制ar阴性PCa细胞株(PC3和DU145)的集落形成和细胞迁移。分子通路分析表明,化合物16的抗癌活性与阻断ERK1/2和Akt活性有关。此外,与对照组相比,化合物16抑制了鸡绒毛膜尿囊膜(CAM)模型的血管生成。构效关系研究表明,通过改变甲氧基化模式,细胞毒性可显著提高2倍以上(IC50 << 2.5 μM)。上述结果表明,吡啶类查尔酮可作为治疗CRPC的铅分子;因此,进一步的体外和体内研究是必要的。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
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
Development of Novel Chalcone Analogs as Potential Multi-Targeted Therapies for Castration-Resistant Prostate Cancer
Prostate cancer (PCa) is the second most frequently diagnosed malignancy, as well as a leading cause of cancer-related mortality in men globally. Despite the initial response to hormonal targeted therapy, the majority of patients ultimately progress to a lethal form of the disease, castration-resistant prostate cancer (CRPC). Therefore, the objective of this study was to discover and develop novel treatment modalities for CRPC. Chalcones are among the highly attractive scaffolds being investigated for their antitumor activities. A library of 26 chalcone analogs were designed, synthesized and evaluated as potential therapies for CRPC. The design was guided by in-silico ADMET prediction in which analogs with favorable drug-likeness properties were prioritized. The new compounds were synthesized, purified and characterized by extensive structural elucidation studies. The compounds in vitro cytotoxicity was evaluated against two androgen receptor (AR)-negative prostate cancer cell lines (PC3 and DU145). Among the tested compounds, pyridine containing analogs (13, 15 and 16) showed potent antiproliferative activities with IC50 values ranging between 4.32-6.47 µM against PC3 and DU145 cell lines. Detailed biological studies of the lead molecule 16 revealed that it can significantly induce apoptosis through upregulation of Bax and downregulation of Bcl-2. In addition, compound 16 potently inhibited colony formation and reduced cell migration of AR-negative PCa cell lines (PC3 and DU145). The molecular pathway analysis showed that the anticancer activity of compound 16 is associated with blocking of ERK1/2 and Akt activities. Furthermore, compound 16 inhibited angiogenesis in the chick chorioallantoic membrane (CAM) model as compared to control. Structure-activity relationship study revealed that the cytotoxicity could dramatically improve via changing the methoxylation pattern by more than 2-folds (IC50 << 2.5 μM). These results indicate that pyridine-based chalcones could serve as promising lead molecules for the treatment of CRPC; thus, further in vitro and in vivo studies are warranted.
求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
自引率
0.00%
发文量
0
期刊最新文献
Investigating the Concomitant Removal of Hydrocarbons and Heavy Metals by highly adapted Bacillus and Pseudomonas strains Exploring QU Health Students' Experiences of Burnout, Anxiety, and Empathy during the COVID-19 Pandemic: A Mixed Method Study Dietary Patterns and Risk of Inflammatory Bowel Disease: Findings from a Case-Control Study Understanding COVID-19-related Burnout in Qatar’s Community Pharmacists using the Job Demands-Resources Theory Experimental Investigations of Gas Kick for Single and Two-Phase Gas-liquid Flow in near Horizontal Wells
×
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