首页 > 最新文献

MedChemComm最新文献

英文 中文
Introduction to the themed collection on ‘AI in Medicinal Chemistry’ 药物化学中的人工智能 "主题文集简介
IF 3.597 Q2 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-10-09 DOI: 10.1039/D4MD90035A
Jian Zhang, Ola Engkvist and Gerhard Hessler

A graphical abstract is available for this content

本内容有图解摘要
{"title":"Introduction to the themed collection on ‘AI in Medicinal Chemistry’","authors":"Jian Zhang, Ola Engkvist and Gerhard Hessler","doi":"10.1039/D4MD90035A","DOIUrl":"https://doi.org/10.1039/D4MD90035A","url":null,"abstract":"<p >A graphical abstract is available for this content</p>","PeriodicalId":88,"journal":{"name":"MedChemComm","volume":null,"pages":null},"PeriodicalIF":3.597,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142443210","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Rationally modified SNX-class Hsp90 inhibitors disrupt extracellular fibronectin assembly without intracellular Hsp90 activity† 经过合理改良的 SNX 类 Hsp90 抑制剂能破坏细胞外纤维粘连蛋白的组装,但不影响细胞内 Hsp90 的活性
IF 3.597 Q2 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-09-02 DOI: 10.1039/D4MD00501E
Gciniwe S. Mathenjwa, Abir Chakraborty, Abantika Chakraborty, Ronel Muller, Mathew P. Akerman, Moira L. Bode, Adrienne L. Edkins and Clinton G. L. Veale

Despite Hsp90's well documented promise as a target for developing cancer chemotherapeutics, its inhibitors have struggled to progress through clinical trials. This is, in part, attributed to the cytoprotective compensatory heat shock response (HSR) stimulated through intracellular Hsp90 inhibition. Beyond its intracellular role, secreted extracellular Hsp90 (eHsp90) interacts with numerous pro-oncogenic extracellular clients. This includes fibronectin, which in the tumour microenvironment enhances cell invasiveness and metastasis. Through the rational modification of known Hsp90 inhibitors (SNX2112 and SNX25a) we developed four Hsp90 inhibitory compounds, whose alterations restricted their interaction with intracellular Hsp90 and did not stimulate the HSR. Two of the modified cohort (compounds 10 and 11) were able to disrupt the assembly of the extracellular fibronectin network at non-cytotoxic concentrations, and thus represent promising new tool compounds for studying the druggability of eHsp90 as a target for inhibition of tumour invasiveness and metastasis.

尽管 Hsp90 作为开发癌症化疗药物的靶点前景广阔,但其抑制剂在临床试验中一直举步维艰。这部分归因于细胞内 Hsp90 抑制所激发的细胞保护代偿性热休克反应(HSR)。除了细胞内作用外,分泌的细胞外 Hsp90(eHsp90)还与许多促癌细胞外客户相互作用。其中包括纤维粘连蛋白,它在肿瘤微环境中会增强细胞的侵袭性和转移性。通过对已知的 Hsp90 抑制剂(SNX2112 和 SNX25a)进行合理改造,我们开发出了四种 Hsp90 抑制化合物,其改造限制了它们与细胞内 Hsp90 的相互作用,并且不会刺激 HSR。其中两种经过修饰的化合物(化合物 10 和 11)能够以非细胞毒性浓度破坏细胞外纤维连接蛋白网络的组装,因此是研究 eHsp90 作为抑制肿瘤侵袭性和转移性靶点的可药用性的有前途的新工具化合物。
{"title":"Rationally modified SNX-class Hsp90 inhibitors disrupt extracellular fibronectin assembly without intracellular Hsp90 activity†","authors":"Gciniwe S. Mathenjwa, Abir Chakraborty, Abantika Chakraborty, Ronel Muller, Mathew P. Akerman, Moira L. Bode, Adrienne L. Edkins and Clinton G. L. Veale","doi":"10.1039/D4MD00501E","DOIUrl":"10.1039/D4MD00501E","url":null,"abstract":"<p >Despite Hsp90's well documented promise as a target for developing cancer chemotherapeutics, its inhibitors have struggled to progress through clinical trials. This is, in part, attributed to the cytoprotective compensatory heat shock response (HSR) stimulated through intracellular Hsp90 inhibition. Beyond its intracellular role, secreted extracellular Hsp90 (eHsp90) interacts with numerous pro-oncogenic extracellular clients. This includes fibronectin, which in the tumour microenvironment enhances cell invasiveness and metastasis. Through the rational modification of known Hsp90 inhibitors (SNX2112 and SNX25a) we developed four Hsp90 inhibitory compounds, whose alterations restricted their interaction with intracellular Hsp90 and did not stimulate the HSR. Two of the modified cohort (compounds <strong>10</strong> and <strong>11</strong>) were able to disrupt the assembly of the extracellular fibronectin network at non-cytotoxic concentrations, and thus represent promising new tool compounds for studying the druggability of eHsp90 as a target for inhibition of tumour invasiveness and metastasis.</p>","PeriodicalId":88,"journal":{"name":"MedChemComm","volume":null,"pages":null},"PeriodicalIF":3.597,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.rsc.org/en/content/articlepdf/2024/md/d4md00501e?page=search","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142252006","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Design, synthesis and biological evaluation of a novel PSMA–PI3K small molecule drug conjugate† 新型 PSMA-PI3K 小分子药物共轭物的设计、合成和生物学评价。
IF 3.597 Q2 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-08-24 DOI: 10.1039/D4MD00246F
Shouguo Peng, Haixia Li, Weilu Cui, Tianning Xiong, Jiaqi Hu, Haixiang Qi, Songwen Lin, Deyu Wu, Ming Ji and Heng Xu

Small molecule drug conjugates are an emerging targeted therapy for cancer treatment. Building upon the overexpressed prostate-specific membrane antigen (PSMA) in prostate cancer, we herein report the design and synthesis of a novel PSMA–PI3K small molecule drug conjugate 1. Conjugate 1 demonstrates potent inhibition against PI3K with an IC50 value of 0.40 nM and simultaneously targets PSMA, giving rise to selective growth inhibition activity for PSMA-positive cancer cells. Conjugate 1 also potently inhibits the phosphorylation of PI3K main downstream effectors and arrests the cell cycle in the G0/G1 phase in PSMA-positive 22Rv1 prostate cancer cells. Further in vivo evaluation shows that conjugate 1 has favorable efficacy and tolerability in a 22Rv1 xenograft model, demonstrating its potential application in targeted cancer therapy.

小分子药物共轭物是一种新兴的癌症靶向疗法。基于前列腺癌中过表达的前列腺特异性膜抗原(PSMA),我们在此报告了新型 PSMA-PI3K 小分子药物共轭物 1 的设计与合成。共轭物 1 对 PI3K 具有强效抑制作用,IC50 值为 0.40 nM,同时还能靶向 PSMA,从而对 PSMA 阳性癌细胞产生选择性生长抑制活性。在 PSMA 阳性的 22Rv1 前列腺癌细胞中,共轭物 1 还能有效抑制 PI3K 主要下游效应物的磷酸化,并使细胞周期停滞在 G0/G1 期。进一步的体内评估显示,共轭物 1 在 22Rv1 异种移植模型中具有良好的疗效和耐受性,证明了它在癌症靶向治疗中的潜在应用。
{"title":"Design, synthesis and biological evaluation of a novel PSMA–PI3K small molecule drug conjugate†","authors":"Shouguo Peng, Haixia Li, Weilu Cui, Tianning Xiong, Jiaqi Hu, Haixiang Qi, Songwen Lin, Deyu Wu, Ming Ji and Heng Xu","doi":"10.1039/D4MD00246F","DOIUrl":"10.1039/D4MD00246F","url":null,"abstract":"<p >Small molecule drug conjugates are an emerging targeted therapy for cancer treatment. Building upon the overexpressed prostate-specific membrane antigen (PSMA) in prostate cancer, we herein report the design and synthesis of a novel PSMA–PI3K small molecule drug conjugate <strong>1</strong>. Conjugate <strong>1</strong> demonstrates potent inhibition against PI3K with an IC<small><sub>50</sub></small> value of 0.40 nM and simultaneously targets PSMA, giving rise to selective growth inhibition activity for PSMA-positive cancer cells. Conjugate <strong>1</strong> also potently inhibits the phosphorylation of PI3K main downstream effectors and arrests the cell cycle in the G0/G1 phase in PSMA-positive 22Rv1 prostate cancer cells. Further <em>in vivo</em> evaluation shows that conjugate <strong>1</strong> has favorable efficacy and tolerability in a 22Rv1 xenograft model, demonstrating its potential application in targeted cancer therapy.</p>","PeriodicalId":88,"journal":{"name":"MedChemComm","volume":null,"pages":null},"PeriodicalIF":3.597,"publicationDate":"2024-08-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142154861","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Pyridazinone-based derivatives as anticancer agents endowed with anti-microbial activity: molecular design, synthesis, and biological investigation† 哒嗪酮类衍生物作为具有抗微生物活性的抗癌剂:分子设计、合成和生物学研究。
IF 3.597 Q2 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-08-16 DOI: 10.1039/D4MD00481G
Mohamed K. S. El-Nagar, Mai I. Shahin, Mohammed F. El-Behairy, Ehab S. Taher, Mohamed F. El-Badawy, Marwa Sharaky, Dalal A. Abou El Ella, Khaled A. M. Abouzid and Mai Adel

Cancer patients undergoing chemotherapy are highly susceptible to infections owing to their compromised immune system, which also promotes cancer progression through inflammation. Thus, this study aimed to develop novel chemotherapeutic agents with both anticancer and antimicrobial properties. A series of diarylurea derivatives based on pyridazinone scaffolds were designed, synthesized, and characterized as surrogates for sorafenib. The synthesized compounds were tested for their antimicrobial activity and screened against 60 cancer cell lines at the National Cancer Institute (NCI). Compound 10h exhibited potent antibacterial activity against Staphylococcus aureus (MIC = 16 μg mL−1), whereas compound 8g showed significant antifungal activity against Candida albicans (MIC = 16 μg mL−1). Additionally, ten compounds were further evaluated for VEGFR-2 inhibition, with compound 17a showing the best inhibitory activity. Compounds 8f, 10l, and 17a demonstrated significant anticancer activity against melanoma, NSCLC, prostate cancer, and colon cancer, with growth inhibition percentages (GI%) ranging from 62.21% to 100.14%. Compounds 10l and 17a were selected for five-dose screening, displaying GI50 values of 1.66–100 μM. Compound 10l induced G0–G1 phase cell cycle arrest in the A549/ATCC cell line, increasing the cell population from 85.41% to 90.86%. Gene expression analysis showed that compound 10l upregulated pro-apoptotic genes p53 and Bax and downregulated the anti-apoptotic gene Bcl-2. Molecular docking studies provided insights into the binding modes of the compounds to the VEGFR-2 enzyme. In conclusion, the pyridazinone-based diarylurea derivatives developed in this study show promise as dual-function antimicrobial and anticancer agents, warranting further investigation.

接受化疗的癌症患者由于免疫系统受损,极易受到感染,而免疫系统受损又会通过炎症促进癌症进展。因此,本研究旨在开发兼具抗癌和抗菌特性的新型化疗药物。研究人员设计、合成了一系列基于哒嗪酮支架的二芳基脲衍生物,并将其表征为索拉非尼的替代物。美国国家癌症研究所(NCI)对合成的化合物进行了抗菌活性测试,并针对 60 种癌细胞系进行了筛选。化合物 10h 对金黄色葡萄球菌具有强效抗菌活性(MIC = 16 μg mL-1),而化合物 8g 对白色念珠菌具有显著的抗真菌活性(MIC = 16 μg mL-1)。此外,还进一步评估了 10 个化合物对 VEGFR-2 的抑制作用,其中化合物 17a 显示出最佳的抑制活性。化合物 8f、10l 和 17a 对黑色素瘤、NSCLC、前列腺癌和结肠癌具有显著的抗癌活性,生长抑制率(GI%)从 62.21% 到 100.14% 不等。化合物 10l 和 17a 被选中进行五剂量筛选,其 GI50 值为 1.66-100 μM。化合物 10l 可诱导 A549/ATCC 细胞系的 G0-G1 期细胞周期停滞,使细胞数量从 85.41% 增加到 90.86%。基因表达分析表明,化合物 10l 上调了促凋亡基因 p53 和 Bax,下调了抗凋亡基因 Bcl-2。分子对接研究揭示了化合物与 VEGFR-2 酶的结合模式。总之,本研究开发的基于哒嗪酮的二元脲衍生物有望成为抗菌和抗癌的双重功能药物,值得进一步研究。
{"title":"Pyridazinone-based derivatives as anticancer agents endowed with anti-microbial activity: molecular design, synthesis, and biological investigation†","authors":"Mohamed K. S. El-Nagar, Mai I. Shahin, Mohammed F. El-Behairy, Ehab S. Taher, Mohamed F. El-Badawy, Marwa Sharaky, Dalal A. Abou El Ella, Khaled A. M. Abouzid and Mai Adel","doi":"10.1039/D4MD00481G","DOIUrl":"10.1039/D4MD00481G","url":null,"abstract":"<p >Cancer patients undergoing chemotherapy are highly susceptible to infections owing to their compromised immune system, which also promotes cancer progression through inflammation. Thus, this study aimed to develop novel chemotherapeutic agents with both anticancer and antimicrobial properties. A series of diarylurea derivatives based on pyridazinone scaffolds were designed, synthesized, and characterized as surrogates for sorafenib. The synthesized compounds were tested for their antimicrobial activity and screened against 60 cancer cell lines at the National Cancer Institute (NCI). Compound <strong>10h</strong> exhibited potent antibacterial activity against <em>Staphylococcus aureus</em> (MIC = 16 μg mL<small><sup>−1</sup></small>), whereas compound <strong>8g</strong> showed significant antifungal activity against <em>Candida albicans</em> (MIC = 16 μg mL<small><sup>−1</sup></small>). Additionally, ten compounds were further evaluated for VEGFR-2 inhibition, with compound <strong>17a</strong> showing the best inhibitory activity. Compounds <strong>8f</strong>, <strong>10l</strong>, and <strong>17a</strong> demonstrated significant anticancer activity against melanoma, NSCLC, prostate cancer, and colon cancer, with growth inhibition percentages (GI%) ranging from 62.21% to 100.14%. Compounds <strong>10l</strong> and <strong>17a</strong> were selected for five-dose screening, displaying GI<small><sub>50</sub></small> values of 1.66–100 μM. Compound <strong>10l</strong> induced G0–G1 phase cell cycle arrest in the A549/ATCC cell line, increasing the cell population from 85.41% to 90.86%. Gene expression analysis showed that compound <strong>10l</strong> upregulated pro-apoptotic genes p53 and Bax and downregulated the anti-apoptotic gene Bcl-2. Molecular docking studies provided insights into the binding modes of the compounds to the VEGFR-2 enzyme. In conclusion, the pyridazinone-based diarylurea derivatives developed in this study show promise as dual-function antimicrobial and anticancer agents, warranting further investigation.</p>","PeriodicalId":88,"journal":{"name":"MedChemComm","volume":null,"pages":null},"PeriodicalIF":3.597,"publicationDate":"2024-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142154866","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Synthesis of a celastrol derivative as a cancer stem cell inhibitor through regulation of the STAT3 pathway for treatment of ovarian cancer† 通过调节 STAT3 通路合成一种作为癌症干细胞抑制剂的青霉烯醇衍生物,用于治疗卵巢癌。
IF 3.597 Q2 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-08-10 DOI: 10.1039/D4MD00468J
Meijuan Liu, Na Li, Zhaoxue Wang, Shuo Wang, Shaoda Ren and Xiaojing Li

Accumulating evidence suggests that the root of drug chemoresistance in ovarian cancer is tightly associated with subpopulations of cancer stem cells (CSCs), whose activation is largely associated with signal transducer and activator of transcription 3 (STAT3) signaling. Recently, celastrol has shown a significant anti-cancer effect on ovarian cancer, but its clinical translation is very challenging due to its oral bioavailability and high organ toxicity. In this study, a celastrol derivative (Cel-N) was synthesized to augment the overall efficacy, and its underlying mechanisms were also explored. Different ovarian cancer cells, SKOV3 and A2780, were used to evaluate and compare the anticancer effects. Cel-N displayed potent activities against all the tested ovarian cancer cells, with the lowest IC50 value of 0.14–0.25 μM. Further studies showed that Cel-N effectively suppressed the colony formation and sphere formation ability, decreased the percentage of CD44+CD24 and ALDH+ cells, and induced ROS production. Furthermore, western blot analysis indicated that Cel-N significantly inhibited both Tyr705 and Ser727 phosphorylation and reduced the protein expression of STAT3. In addition, Cel-N could dramatically induce apoptosis and cell cycle arrest, and inhibit migration and invasion. Importantly, Cel-N showed a potent antitumor efficacy with no or limited systemic toxicity in mice xenograft models. The anticancer effect of Cel-N is stronger than celastrol. Cel-N attenuates cancer cell stemness, inhibits the STAT3 pathway, and exerts anti-ovarian cancer effects in cell and mouse models. Our data support that Cel-N is a potent drug candidate for ovarian cancer.

越来越多的证据表明,卵巢癌化疗耐药性的根源与癌症干细胞亚群密切相关,而癌症干细胞的活化主要与信号转导和激活转录3(STAT3)信号转导有关。最近,塞拉斯托(celastrol)对卵巢癌有显著的抗癌作用,但由于其口服生物利用度和高器官毒性,其临床转化非常具有挑战性。本研究合成了一种青霉烷醇衍生物(Cel-N),以增强其整体疗效,并探索其潜在机制。研究人员使用不同的卵巢癌细胞(SKOV3 和 A2780)来评估和比较其抗癌效果。Cel-N 对所有测试的卵巢癌细胞都显示出强大的活性,最低 IC50 值为 0.14-0.25 μM。进一步的研究表明,Cel-N能有效抑制细胞的集落形成和球形成能力,降低CD44+CD24-和ALDH+细胞的比例,并诱导ROS的产生。此外,Western 印迹分析表明,Cel-N 能显著抑制 Tyr705 和 Ser727 的磷酸化,并降低 STAT3 的蛋白表达。此外,Cel-N 还能显著诱导细胞凋亡和细胞周期停滞,并抑制细胞的迁移和侵袭。重要的是,在小鼠异种移植模型中,Cel-N显示出了强大的抗肿瘤功效,并且没有或仅有有限的全身毒性。Cel-N 的抗癌效果强于 celastrol。Cel-N 可减轻癌细胞干性,抑制 STAT3 通路,并在细胞和小鼠模型中发挥抗卵巢癌作用。我们的数据支持 Cel-N 是一种治疗卵巢癌的有效候选药物。
{"title":"Synthesis of a celastrol derivative as a cancer stem cell inhibitor through regulation of the STAT3 pathway for treatment of ovarian cancer†","authors":"Meijuan Liu, Na Li, Zhaoxue Wang, Shuo Wang, Shaoda Ren and Xiaojing Li","doi":"10.1039/D4MD00468J","DOIUrl":"10.1039/D4MD00468J","url":null,"abstract":"<p >Accumulating evidence suggests that the root of drug chemoresistance in ovarian cancer is tightly associated with subpopulations of cancer stem cells (CSCs), whose activation is largely associated with signal transducer and activator of transcription 3 (STAT3) signaling. Recently, celastrol has shown a significant anti-cancer effect on ovarian cancer, but its clinical translation is very challenging due to its oral bioavailability and high organ toxicity. In this study, a celastrol derivative (<strong>Cel-N</strong>) was synthesized to augment the overall efficacy, and its underlying mechanisms were also explored. Different ovarian cancer cells, SKOV3 and A2780, were used to evaluate and compare the anticancer effects. <strong>Cel-N</strong> displayed potent activities against all the tested ovarian cancer cells, with the lowest IC<small><sub>50</sub></small> value of 0.14–0.25 μM. Further studies showed that <strong>Cel-N</strong> effectively suppressed the colony formation and sphere formation ability, decreased the percentage of CD44<small><sup>+</sup></small>CD24<small><sup>−</sup></small> and ALDH<small><sup>+</sup></small> cells, and induced ROS production. Furthermore, western blot analysis indicated that <strong>Cel-N</strong> significantly inhibited both Tyr705 and Ser727 phosphorylation and reduced the protein expression of STAT3. In addition, <strong>Cel-N</strong> could dramatically induce apoptosis and cell cycle arrest, and inhibit migration and invasion. Importantly, <strong>Cel-N</strong> showed a potent antitumor efficacy with no or limited systemic toxicity in mice xenograft models. The anticancer effect of <strong>Cel-N</strong> is stronger than celastrol. <strong>Cel-N</strong> attenuates cancer cell stemness, inhibits the STAT3 pathway, and exerts anti-ovarian cancer effects in cell and mouse models. Our data support that <strong>Cel-N</strong> is a potent drug candidate for ovarian cancer.</p>","PeriodicalId":88,"journal":{"name":"MedChemComm","volume":null,"pages":null},"PeriodicalIF":3.597,"publicationDate":"2024-08-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142154878","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A comprehensive apoptotic assessment of niloticin in cervical cancer cells: a tirucallane-type triterpenoid from Aphanamixis polystachya (Wall.) Parker† 对宫颈癌细胞中尼洛替丁(niloticin)凋亡作用的全面评估:一种来自 Aphanamixis polystachya (Wall.) Parker 的 tirucallane 型三萜类化合物。
IF 3.597 Q2 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-08-08 DOI: 10.1039/D4MD00318G
Anuja Gracy Joseph, Mohanan Biji, Vishnu Priya Murali, Daisy R. Sherin, Alisha Valsan, Vimalkumar P. Sukumaran, Kokkuvayil Vasu Radhakrishnan and Kaustabh Kumar Maiti

Pharmacologically active small organic molecules derived from natural resources are prominent drug candidates due to their inherent structural diversity. Herein, we explored one such bioactive molecule, niloticin, which is a tirucallane-type triterpenoid isolated from the stem barks of Aphanamixis polystachya (Wall.) Parker. After initial screening with other isolated compounds from the same plant, niloticin demonstrated selective cytotoxicity against cervical cancer cells (HeLa) with an IC50 value of 11.64 μM. Whereas the compound exhibited minimal cytotoxicity in normal epithelial cell line MCF-10A, with an IC50 value of 83.31 μM. Subsequently, in silico molecular docking studies of niloticin based on key apoptotic proteins such as p53, Fas, FasL, and TNF β revealed striking binding affinity, reflecting docking scores of −7.2, −7.1, −6.8, and −7.2. Thus, the binding stability was evaluated through molecular dynamic simulation. In a downstream process, the apoptotic capability of niloticin was effectively validated through in vitro fluorimetric assays, encompassing nuclear fragmentation. Additionally, an insightful approach involving surface-enhanced Raman spectroscopy (SERS) re-establishes the occurrence of DNA cleavage during cellular apoptosis. Furthermore, niloticin was observed to induce apoptosis through both intrinsic and extrinsic pathways. This was evidenced by the upregulation of upstream regulatory molecules such as CD40 and TNF, which facilitate the activation of caspase 8. Concurrently, niloticin-induced p53 activation augmented the expression of proapoptotic proteins Bax and Bcl-2 and downregulation of IAPs, leading to the release of cytochrome C and subsequent activation of caspase 9. Therefore, the reflection of mitochondrial-mediated apoptosis is in good agreement with molecular docking studies. Furthermore, the anti-metastatic potential was evidenced by wound area closure and Ki67 expression patterns. This pivotal in vitro assessment confirms the possibility of niloticin being a potent anti-cancer drug candidate, and to the best of our knowledge, this is the first comprehensive anticancer assessment of niloticin in HeLa cells.

从自然资源中提取的具有药理活性的有机小分子因其固有的结构多样性而成为重要的候选药物。在本文中,我们探索了这样一种生物活性分子--niloticin,它是从 Aphanamixis polystachya (Wall.) Parker 的茎皮中分离出来的一种桐木烷型三萜类化合物。在与从同一种植物中分离出来的其他化合物进行初步筛选后,尼罗替丁对宫颈癌细胞(HeLa)具有选择性细胞毒性,IC50 值为 11.64 μM。而该化合物对正常上皮细胞系 MCF-10A 的细胞毒性很小,IC50 值为 83.31 μM。随后,基于 p53、Fas、FasL 和 TNF β 等关键凋亡蛋白对尼洛替星进行的硅学分子对接研究显示,该化合物与这些蛋白的结合亲和力惊人,对接得分分别为 -7.2、-7.1、-6.8 和 -7.2。因此,通过分子动力学模拟对其结合稳定性进行了评估。在下游过程中,通过体外荧光测定(包括核破碎)有效地验证了尼罗替丁的凋亡能力。此外,一种涉及表面增强拉曼光谱(SERS)的具有洞察力的方法再次证实了细胞凋亡过程中 DNA 断裂的发生。此外,还观察到尼洛替星通过内在和外在途径诱导细胞凋亡。CD40和TNF等上游调控分子的上调证明了这一点,它们促进了caspase 8的活化。同时,尼罗替丁诱导的 p53 激活增加了促凋亡蛋白 Bax 和 Bcl-2 的表达,并下调了 IAPs,导致细胞色素 C 的释放和随后的 caspase 9 激活。因此,线粒体介导的细胞凋亡反映与分子对接研究非常吻合。此外,伤口面积闭合和 Ki67 表达模式也证明了抗转移潜力。据我们所知,这是首次在 HeLa 细胞中对尼洛替星进行全面的抗癌评估。
{"title":"A comprehensive apoptotic assessment of niloticin in cervical cancer cells: a tirucallane-type triterpenoid from Aphanamixis polystachya (Wall.) Parker†","authors":"Anuja Gracy Joseph, Mohanan Biji, Vishnu Priya Murali, Daisy R. Sherin, Alisha Valsan, Vimalkumar P. Sukumaran, Kokkuvayil Vasu Radhakrishnan and Kaustabh Kumar Maiti","doi":"10.1039/D4MD00318G","DOIUrl":"10.1039/D4MD00318G","url":null,"abstract":"<p >Pharmacologically active small organic molecules derived from natural resources are prominent drug candidates due to their inherent structural diversity. Herein, we explored one such bioactive molecule, niloticin, which is a tirucallane-type triterpenoid isolated from the stem barks of <em>Aphanamixis polystachya</em> (Wall.) Parker. After initial screening with other isolated compounds from the same plant, niloticin demonstrated selective cytotoxicity against cervical cancer cells (HeLa) with an IC<small><sub>50</sub></small> value of 11.64 μM. Whereas the compound exhibited minimal cytotoxicity in normal epithelial cell line MCF-10A, with an IC<small><sub>50</sub></small> value of 83.31 μM. Subsequently, <em>in silico</em> molecular docking studies of niloticin based on key apoptotic proteins such as p53, Fas, FasL, and TNF β revealed striking binding affinity, reflecting docking scores of −7.2, −7.1, −6.8, and −7.2. Thus, the binding stability was evaluated through molecular dynamic simulation. In a downstream process, the apoptotic capability of niloticin was effectively validated through <em>in vitro</em> fluorimetric assays, encompassing nuclear fragmentation. Additionally, an insightful approach involving surface-enhanced Raman spectroscopy (SERS) re-establishes the occurrence of DNA cleavage during cellular apoptosis. Furthermore, niloticin was observed to induce apoptosis through both intrinsic and extrinsic pathways. This was evidenced by the upregulation of upstream regulatory molecules such as CD40 and TNF, which facilitate the activation of caspase 8. Concurrently, niloticin-induced p53 activation augmented the expression of proapoptotic proteins Bax and Bcl-2 and downregulation of IAPs, leading to the release of cytochrome C and subsequent activation of caspase 9. Therefore, the reflection of mitochondrial-mediated apoptosis is in good agreement with molecular docking studies. Furthermore, the anti-metastatic potential was evidenced by wound area closure and Ki67 expression patterns. This pivotal <em>in vitro</em> assessment confirms the possibility of niloticin being a potent anti-cancer drug candidate, and to the best of our knowledge, this is the first comprehensive anticancer assessment of niloticin in HeLa cells.</p>","PeriodicalId":88,"journal":{"name":"MedChemComm","volume":null,"pages":null},"PeriodicalIF":3.597,"publicationDate":"2024-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142154859","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Synthesis and cytotoxic activity of madecassic acid–silybin conjugate compounds in liver cancer cells† 马黛茶酸-水飞蓟宾共轭化合物的合成及其对肝癌细胞的细胞毒性活性
IF 3.597 Q2 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-08-02 DOI: 10.1039/D4MD00170B
Chien Van Tran, Thao Thi Phuong Tran, Anh The Nguyen, Loc Van Tran, Ninh Thi Pham, Luu Thi Nguyen, Dung Thi Nguyen, Michelle D. Garrett, Nga Thi Nguyen, Thao Thi Do, Christopher J. Serpell and Sung Van Tran

A series of 14 conjugates of 2α,3β,23-triacetyl-madecassic acid and silybin were designed and synthesized. The madecassic acid unit was linked to silybin either directly at position C-7 or C-3; or through an amino acid linker (glycine, β-alanine, or 11-aminoundecanoic acid) at position C-3. The conjugates were tested in vitro for their cytotoxic effect on HepG2 cells using the MTT assay. The results confirmed that the conjugated compounds demonstrated a stronger cytotoxic effect compared to the parent compounds. Of these compounds, the most promising conjugate, compound 8, was evaluated for cytotoxic activity in the additional Hep3B, Huh7, and Huh7R human hepatocellular carcinoma cell lines and also for cell cycle changes and induction of apoptosis in HepG2 cells. This compound caused a rapid and significant induction of caspase 3 activity and induced cell cycle arrest in the S phase – effects distinct from the activity of madecassic acid. This is the first study on the synthesis and cytotoxicity of madecassic acid–silybin conjugates, and of their testing against liver cancer cell lines and provides evidence for a distinct biological profile versus madecassic acid alone.

我们设计并合成了一系列 14 种 2α,3β,23-三乙酰基棕榈酸与水飞蓟宾的共轭物。水飞蓟酸单元与水飞蓟宾的连接方式有两种,一种是在 C-7 位或 C-3 位直接连接,另一种是在 C-3 位通过氨基酸连接体(甘氨酸、β-丙氨酸或 11-氨基十一烷酸)连接。采用 MTT 法体外测试了共轭物对 HepG2 细胞的细胞毒性作用。结果证实,与母体化合物相比,共轭化合物具有更强的细胞毒性作用。在这些化合物中,最有前景的共轭化合物化合物 8 在其他 Hep3B、Huh7 和 Huh7R 人肝癌细胞系中进行了细胞毒性活性评估,并在 HepG2 细胞中进行了细胞周期变化和诱导细胞凋亡评估。该化合物可快速、显著地诱导 Caspase 3 的活性,并诱导细胞周期停滞在 S 期,其效果与马来酸的活性截然不同。这是第一项关于疯草酸-水飞蓟宾共轭物的合成和细胞毒性的研究,也是第一项针对肝癌细胞系的测试研究,为其与单独的疯草酸相比具有不同的生物特性提供了证据。
{"title":"Synthesis and cytotoxic activity of madecassic acid–silybin conjugate compounds in liver cancer cells†","authors":"Chien Van Tran, Thao Thi Phuong Tran, Anh The Nguyen, Loc Van Tran, Ninh Thi Pham, Luu Thi Nguyen, Dung Thi Nguyen, Michelle D. Garrett, Nga Thi Nguyen, Thao Thi Do, Christopher J. Serpell and Sung Van Tran","doi":"10.1039/D4MD00170B","DOIUrl":"10.1039/D4MD00170B","url":null,"abstract":"<p >A series of 14 conjugates of 2α,3β,23-triacetyl-madecassic acid and silybin were designed and synthesized. The madecassic acid unit was linked to silybin either directly at position C-7 or C-3; or through an amino acid linker (glycine, β-alanine, or 11-aminoundecanoic acid) at position C-3. The conjugates were tested <em>in vitro</em> for their cytotoxic effect on HepG2 cells using the MTT assay. The results confirmed that the conjugated compounds demonstrated a stronger cytotoxic effect compared to the parent compounds. Of these compounds, the most promising conjugate, compound <strong>8</strong>, was evaluated for cytotoxic activity in the additional Hep3B, Huh7, and Huh7R human hepatocellular carcinoma cell lines and also for cell cycle changes and induction of apoptosis in HepG2 cells. This compound caused a rapid and significant induction of caspase 3 activity and induced cell cycle arrest in the S phase – effects distinct from the activity of madecassic acid. This is the first study on the synthesis and cytotoxicity of madecassic acid–silybin conjugates, and of their testing against liver cancer cell lines and provides evidence for a distinct biological profile <em>versus</em> madecassic acid alone.</p>","PeriodicalId":88,"journal":{"name":"MedChemComm","volume":null,"pages":null},"PeriodicalIF":3.597,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11343037/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142056380","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The synthesis and bioactivities of ROCK2 inhibitors with 1,2-dithiolan-3-yl motif† 具有 1,2-二硫环戊-3-基基团的 ROCK2 抑制剂的合成及其生物活性
IF 3.597 Q2 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-08-01 DOI: 10.1039/D4MD00438H
Ruolin Cao, Fangyu Du, Zhiqiang Liu, Pengcheng Cai, Minggang Qi, Wei Xiao, Xuefei Bao and Guoliang Chen

Rho-associated coiled-coil containing kinase (ROCK) plays an important role in inflammation. Herein, a series of compounds were designed and synthesized as ROCK inhibitors based on the structure-based drug design (SBDD) strategy and were evaluated for cytotoxicity, antioxidant activity and anti-inflammatory activity. Among them, compound DC24 was identified as the optimal hit in enzymatic screening with an IC50 value of 0.124 μM against ROCK2 and 50-fold selectivity over ROCK1. DC24 has a novel lipid amide scaffold with a bis(4-fluorophenyl)methyl substituent, and DC24 is the first ROCK2 inhibitor interacting with the hinge region of ROCK2 via the 1,2-dithiolan-3-yl motif, which has been confirmed by the binding model of DC24 with ROCK2. In a complete Freund's adjuvant (CFA) induced acute inflammation model, DC24 at a dose of 5 mg kg−1 exhibited an anti-inflammatory effect better than that of belumosudil. Furthermore, DC24 exhibits good safety in vivo.

Rho相关含盘卷激酶(ROCK)在炎症中发挥着重要作用。本文基于基于结构的药物设计(SBDD)策略,设计合成了一系列化合物作为ROCK抑制剂,并对其细胞毒性、抗氧化活性和抗炎活性进行了评价。其中,化合物 DC24 在酶筛选中被确定为最佳靶点,其对 ROCK2 的 IC50 值为 0.124 μM,选择性是 ROCK1 的 50 倍。DC24 具有双(4-氟苯基)甲基取代基的新型脂质酰胺支架,是首个通过 1,2-二硫环戊-3-基基团与 ROCK2 铰链区相互作用的 ROCK2 抑制剂,DC24 与 ROCK2 的结合模型证实了这一点。在完全弗氏佐剂(CFA)诱导的急性炎症模型中,剂量为 5 mg kg-1 的 DC24 的抗炎效果优于贝卢莫司地。此外,DC24 在体内表现出良好的安全性。
{"title":"The synthesis and bioactivities of ROCK2 inhibitors with 1,2-dithiolan-3-yl motif†","authors":"Ruolin Cao, Fangyu Du, Zhiqiang Liu, Pengcheng Cai, Minggang Qi, Wei Xiao, Xuefei Bao and Guoliang Chen","doi":"10.1039/D4MD00438H","DOIUrl":"10.1039/D4MD00438H","url":null,"abstract":"<p >Rho-associated coiled-coil containing kinase (ROCK) plays an important role in inflammation. Herein, a series of compounds were designed and synthesized as ROCK inhibitors based on the structure-based drug design (SBDD) strategy and were evaluated for cytotoxicity, antioxidant activity and anti-inflammatory activity. Among them, compound <strong>DC24</strong> was identified as the optimal hit in enzymatic screening with an IC<small><sub>50</sub></small> value of 0.124 μM against ROCK2 and 50-fold selectivity over ROCK1. <strong>DC24</strong> has a novel lipid amide scaffold with a bis(4-fluorophenyl)methyl substituent, and <strong>DC24</strong> is the first ROCK2 inhibitor interacting with the hinge region of ROCK2 <em>via</em> the 1,2-dithiolan-3-yl motif, which has been confirmed by the binding model of <strong>DC24</strong> with ROCK2. In a complete Freund's adjuvant (CFA) induced acute inflammation model, <strong>DC24</strong> at a dose of 5 mg kg<small><sup>−1</sup></small> exhibited an anti-inflammatory effect better than that of belumosudil. Furthermore, <strong>DC24</strong> exhibits good safety <em>in vivo</em>.</p>","PeriodicalId":88,"journal":{"name":"MedChemComm","volume":null,"pages":null},"PeriodicalIF":3.597,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142222000","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Gold(i) and gold(iii) carbene complexes from the marine betaine norzooanemonin: inhibition of thioredoxin reductase, antiproliferative and antimicrobial activity† 来自海洋甜菜碱 norzooanemonin 的金(i)和金(iii)碳烯配合物:抑制硫氧还原酶、抗增殖和抗菌活性。
IF 3.597 Q2 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-07-31 DOI: 10.1039/D4MD00358F
Seyedeh Mahbobeh Mahdavi, Dirk Bockfeld, Igor V. Esarev, Petra Lippmann, René Frank, Mark Brönstrup, Ingo Ott and Matthias Tamm

The natural marine betaine norzooanemonin (1,3-dimethylimidazolim-4-carboxylate) and its methyl and ethyl esters were used as ligand precursors to prepare a systematic series (12 members) of neutral monocarbene gold(I/III) and cationic dicarbene gold(I/III) complexes. The complexes were evaluated as inhibitors of bacterial thioredoxin reductase and for their antiproliferative and antimicrobial activities. While gold complexes with the parent norzooanemonin scaffold resulted in overall poor performance, the more lipophilic esters proved to be highly bioactive agents, related to their higher cellular uptake. The monocarbene gold(I/III) complexes showed significant potency as inhibitors of bacterial thioredoxin reductase. In most assays, the efficacy of both gold(I) and gold(III) analogues was found to be comparable. The cytotoxicity of dicarbene gold(I/III) complexes against cancer cells was strong, in some cases exceeding that of the standard reference auranofin.

研究人员以天然海洋甜菜碱 Norzooanemonin(1,3-二甲基咪唑啉-4-羧酸酯)及其甲酯和乙酯为配体前体,制备了一系列(12 个成员)中性单碳烯金(i/iii)和阳离子二碳烯金(i/iii)配合物。这些配合物被评估为细菌硫氧还蛋白还原酶的抑制剂,并具有抗增殖和抗菌活性。亲脂性更强的酯类被证明具有很高的生物活性,这与它们较高的细胞吸收率有关。单碳烯金(i/iii)复合物作为细菌硫代还原酶的抑制剂显示出显著的效力。在大多数试验中,金(i)和金(iii)类似物的功效相当。二碳烯金(i/iii)复合物对癌细胞的细胞毒性很强,在某些情况下甚至超过了标准参考物金诺芬。
{"title":"Gold(i) and gold(iii) carbene complexes from the marine betaine norzooanemonin: inhibition of thioredoxin reductase, antiproliferative and antimicrobial activity†","authors":"Seyedeh Mahbobeh Mahdavi, Dirk Bockfeld, Igor V. Esarev, Petra Lippmann, René Frank, Mark Brönstrup, Ingo Ott and Matthias Tamm","doi":"10.1039/D4MD00358F","DOIUrl":"10.1039/D4MD00358F","url":null,"abstract":"<p >The natural marine betaine norzooanemonin (1,3-dimethylimidazolim-4-carboxylate) and its methyl and ethyl esters were used as ligand precursors to prepare a systematic series (12 members) of neutral monocarbene gold(<small>I</small>/<small>III</small>) and cationic dicarbene gold(<small>I</small>/<small>III</small>) complexes. The complexes were evaluated as inhibitors of bacterial thioredoxin reductase and for their antiproliferative and antimicrobial activities. While gold complexes with the parent norzooanemonin scaffold resulted in overall poor performance, the more lipophilic esters proved to be highly bioactive agents, related to their higher cellular uptake. The monocarbene gold(<small>I</small>/<small>III</small>) complexes showed significant potency as inhibitors of bacterial thioredoxin reductase. In most assays, the efficacy of both gold(<small>I</small>) and gold(<small>III</small>) analogues was found to be comparable. The cytotoxicity of dicarbene gold(<small>I</small>/<small>III</small>) complexes against cancer cells was strong, in some cases exceeding that of the standard reference auranofin.</p>","PeriodicalId":88,"journal":{"name":"MedChemComm","volume":null,"pages":null},"PeriodicalIF":3.597,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11342128/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142056378","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Synthesis and antiproliferative potency of 1,3,4-thiadiazole and 1,3-thiazolidine-4-one based new binary heterocyclic molecules: in vitro cell-based anticancer studies† 基于 1,3,4-噻二唑和 1,3-噻唑烷-4-酮的新二元杂环分子的合成和抗增殖效力:体外细胞抗癌研究
IF 3.597 Q2 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2024-07-31 DOI: 10.1039/D4MD00279B
Avik Maji, Ambati Himaja, Sripathi Nikhitha, Soumitra Rana, Abhik Paul, Ajeya Samanta, Uday Shee, Chhanda Mukhopadhyay, Balaram Ghosh and Tapan Kumar Maity

Herein, we report the synthesis and anticancer properties of 21 new 1,3,4-thiadiazole-2-yl-imino-thiazolidine-4-one containing binary heterocyclic molecules. Cytotoxicity of the synthesized molecules was evaluated on various in vitro cancer cell lines (MCF-7, PC3, 4T1, MDA-MB-231, and MOC2) and normal human embryonic cell lines (HEK-293) via MTT assay. The cytotoxicity data of developed compounds was compared with the reference anticancer molecule BG45, a selective inhibitor of the HDAC3 enzyme. All compounds showed a significant cytotoxic effect higher than BG45 on tested cancer cell lines. Moreover, the compounds exhibited better selectivity on cancer cells than on normal cells. Among the molecules, compound 6e is the most potent in cytotoxic activity on MCF-7 cell lines (IC50 value of 3.85 μM). Additional mechanistic investigation revealed that compound 6e promotes apoptosis (25.3%) and G0/G1 phase cell cycle arrest of MCF-7 cells. Also, compound 6e induces intracellular ROS accumulation and subsequent nuclear fragmentation. Hence, this research finds new hybrid molecules active against in vitro cancer cells.

在此,我们报告了 21 种新的 1,3,4-噻二唑-2-基-亚氨基噻唑烷-4-酮二元杂环分子的合成和抗癌特性。通过 MTT 试验评估了合成分子对各种体外癌细胞株(MCF-7、PC3、4T1、MDA-MB-231 和 MOC2)和正常人胚胎细胞株(HEK-293)的细胞毒性。所开发化合物的细胞毒性数据与参考抗癌分子 BG45(一种 HDAC3 酶的选择性抑制剂)进行了比较。在测试的癌细胞系中,所有化合物的细胞毒性效果均明显高于 BG45。此外,这些化合物对癌细胞的选择性优于正常细胞。在这些分子中,化合物 6e 对 MCF-7 细胞株的细胞毒性最强(IC50 值为 3.85 μM)。其他机理研究表明,化合物 6e 能促进 MCF-7 细胞凋亡(25.3%)和 G0/G1 期细胞周期停滞。此外,化合物 6e 还能诱导细胞内 ROS 的积累和随后的核破碎。因此,这项研究发现了对体外癌细胞具有活性的新混合分子。
{"title":"Synthesis and antiproliferative potency of 1,3,4-thiadiazole and 1,3-thiazolidine-4-one based new binary heterocyclic molecules: in vitro cell-based anticancer studies†","authors":"Avik Maji, Ambati Himaja, Sripathi Nikhitha, Soumitra Rana, Abhik Paul, Ajeya Samanta, Uday Shee, Chhanda Mukhopadhyay, Balaram Ghosh and Tapan Kumar Maity","doi":"10.1039/D4MD00279B","DOIUrl":"10.1039/D4MD00279B","url":null,"abstract":"<p >Herein, we report the synthesis and anticancer properties of 21 new 1,3,4-thiadiazole-2-yl-imino-thiazolidine-4-one containing binary heterocyclic molecules. Cytotoxicity of the synthesized molecules was evaluated on various <em>in vitro</em> cancer cell lines (MCF-7, PC3, 4T1, MDA-MB-231, and MOC2) and normal human embryonic cell lines (HEK-293) <em>via</em> MTT assay. The cytotoxicity data of developed compounds was compared with the reference anticancer molecule <strong>BG45</strong>, a selective inhibitor of the HDAC3 enzyme. All compounds showed a significant cytotoxic effect higher than <strong>BG45</strong> on tested cancer cell lines. Moreover, the compounds exhibited better selectivity on cancer cells than on normal cells. Among the molecules, compound <strong>6e</strong> is the most potent in cytotoxic activity on MCF-7 cell lines (IC<small><sub>50</sub></small> value of 3.85 μM). Additional mechanistic investigation revealed that compound <strong>6e</strong> promotes apoptosis (25.3%) and G0/G1 phase cell cycle arrest of MCF-7 cells. Also, compound <strong>6e</strong> induces intracellular ROS accumulation and subsequent nuclear fragmentation. Hence, this research finds new hybrid molecules active against <em>in vitro</em> cancer cells.</p>","PeriodicalId":88,"journal":{"name":"MedChemComm","volume":null,"pages":null},"PeriodicalIF":3.597,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141872883","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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
MedChemComm
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: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