首页 > 最新文献

Anti-cancer agents in medicinal chemistry最新文献

英文 中文
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 Q3 CHEMISTRY, MEDICINAL 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

Introduction: 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.

Method: 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.

Results: 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.

Conclusion: Our results suggest that pranlukast inhibits the PD-1/PD-L1 axis, meriting its repurposing as an antitumor drug.

简介可以通过调节患者的免疫反应来靶向治疗肿瘤。程序性细胞死亡蛋白 1(PD-1)和程序性细胞死亡配体 1(PD-L1)是癌症生物学中关键的免疫检查点。某些癌症免疫疗法的疗效是通过使用单克隆抗体靶向这些分子实现的:小分子药物也被开发为 PD-1/PD-L1 轴的抑制剂,其作用机制与抗体不同:它们能诱导 PD-L1 同源二聚体的形成,使其稳定、内化并随后降解。药物再利用是一种为已批准的药物寻找新用途的策略,根据最新研究结果加快药物的临床转化。在这项研究中,我们根据已报道的以 PD-L1 为靶点的小分子药物生成了一个药理模型,并用它在 FDA 批准的药物中识别出潜在的 PD-L1 抑制剂:结果:我们发现了 12 种药理匹配化合物,但只有 4 种在对接实验中重现了参考抑制剂的结合模式。分子动力学的进一步表征表明,用于治疗哮喘的白三烯受体拮抗剂普仑司特能与 PD-L1 同源二聚体产生稳定且能量有利的相互作用,并诱导重组 PD-L1 同源二聚化:我们的研究结果表明,普萘卡斯特能抑制 PD-1/PD-L1 轴,值得将其重新用作抗肿瘤药物。
{"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 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-10-10 DOI: 10.2174/0118715206322603241002064435
Michał Antoszczak, Magdalena Mielczarek-Puta, Marta Struga, Adam Huczyński

Background: 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.

Objective: Thus, a very interesting research direction is the chemical modification of SAL structure, which could give more biologically active agents.

Methods: 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.

Results: 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.

Conclusion: 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.

背景:自从发现一种名为盐霉素(SAL)的天然聚醚离子态化合物可选择性地抑制人类癌细胞以来,科学界一直对这种化合物给予特别关注。对它的研究已有近 15 年的历史:因此,一个非常有趣的研究方向是对 SAL 结构进行化学修饰,从而获得更具生物活性的制剂:我们评估了 C20-表氨基水杨霉素(化合物 3b)的(硫)脲类似物类的抗癌活性。研究内容包括活性氧(ROS)的生成、促凋亡活性、细胞毒性活性以及体外脂质过氧化反应:结果:硫脲类化合物 5a-5d 对特定人类结肠癌细胞株的抗增殖活性高于化学未修饰的 SAL,对结肠癌转移变异细胞(SW620)的效力高出 2~10 倍。从机理上讲,SAL 衍生物在原发性结肠癌细胞中显示出促凋亡活性,并诱导这些细胞产生活性氧(ROS)。在 SW620 细胞中,SAL 衍生物增加了脂质过氧化反应,但对细胞凋亡的影响较弱,ROS 的形成较少,细胞毒性作用之后是细胞抑制作用,这表明这些化合物对原发性和转移性结肠癌细胞具有不同的作用模式:本研究结果表明,SAL 的脲和硫脲衍生物为合理开发新的抗癌活性制剂提供了很好的线索。
{"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 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-10-09 DOI: 10.2174/0118715206327654240823074318
Noha S Basuony, Tarek M Mohamed, Doha M Beltagy, Ahmed A Massoud, Mona M Elwan

Objective: 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).

Methods: 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.

Results: 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.

Conclusion: 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.

目的:藏红花中的主要抗氧化剂藏红花苷(CRO)因其抗癌特性而闻名。然而,由于生物利用度低、吸收性差和理化稳定性低,其局部治疗效果有限。本研究旨在制备藏红花苷纳米颗粒(CRO-NPs),以提高其药效,并评估藏红花苷纳米颗粒与多柔比星(DOX)化疗对两种细胞系:人肝癌细胞(HepG2)和非癌细胞(WI38)的协同作用:采用乳液扩散技术制备了 CRO-NPs,并通过透射电子显微镜(TEM)、扫描电子显微镜(SEM)、Zeta 电位和傅立叶变换红外光谱(FT-IR)对其进行了表征。采用 MTT 法评估 DOX、CRO、CRO-NPs 和 DOX+CRO-NPs 对细胞增殖的抑制作用。流式细胞术评估了细胞凋亡和细胞周期,实时聚合酶链反应分析了凋亡基因(P53)和自噬基因(ATG5 和 LC3)的表达变化:TEM和SEM显示,CRO-NPs呈相对球形,平均尺寸为9.3 nm,zeta电位分析表明与原生CRO相比,CRO-NPs具有更好的稳定性。与原生 CRO(IC50 = 6.1 毫克/毫升和 3.2 毫克/毫升)相比,CRO-NPs 在 24 小时和 48 小时后对 HepG2 细胞的抗肿瘤效果显著更高(IC50 = 1.1 毫克/毫升和 0.57 毫克/毫升)。对 HepG2 细胞进行的 Annexin-V 检测表明,所有处理的细胞凋亡率都有所上升,其中 CRO-NPs 的凋亡率最高,同时细胞周期停滞在 G2/M 阶段。此外,基因表达分析表明,与单独处理相比,在 DOX/CRO-NPs 联合处理中,P53、ATG5 和 LC3 基因上调。相比之下,WI38 细胞对 DOX 的毒性更敏感,但对 CRONPs 没有不良反应:尽管还需要在动物模型中进行更多的体内研究来证实这些结果,但我们的研究结果表明,CRO-NPs 可作为一种潜在的肝细胞癌抗癌新药。此外,CRO-NPs 与 DOX 对 HepG2 细胞有协同作用,并能减轻 DOX 对正常 WI38 细胞的毒性。
{"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 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-10-03 DOI: 10.2174/0118715206329892240927081033
Xueshuai Ye, Yongqiang Wu, Haiqiang Zhang

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.

claudin18.2(CLDN18.2)是一种膜蛋白,对胃黏膜分化上皮细胞的紧密连接至关重要,在屏障功能中发挥着重要作用,但由于其极性模式,很难被免疫细胞识别。随着胃肿瘤细胞极性的改变,claudin18.2暴露于细胞表面,从而被免疫系统识别,成为理想的靶点。在这篇综述中,我们总结了claudin18.2在正常细胞和恶性肿瘤细胞中的表达调控机制。此外,我们还分析了claudin18.2阳性胃癌和claudin18.2靶向治疗的现有临床结果和未来研究的潜在领域。总之,claudin18.2是胃癌治疗的理想靶点,claudin18.2靶向治疗改变了胃癌的治疗模式。
{"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 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-10-01 DOI: 10.2174/0118715206343550240919055701
Zhanhu Zhang, Yiqian Ding

Background: 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.

Objective: 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.

Methods: 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.

Results: 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.

Conclusion: 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.

背景:肿瘤细胞对顺铂的耐药性是子宫内膜癌化疗中常见的难题,其产生机制多种多样。使用蛋白酶体抑制剂(如 MG132)的靶向疗法已被研究用于增强顺铂的敏感性,有可能提供一种新的治疗方法:方法:将人子宫内膜癌 RL95-2 细胞暴露于 MG132,以剂量依赖的方式评估细胞活力。研究评估了 MG132 对顺铂诱导的增殖抑制和细胞凋亡的影响,这与 Caspase-3 激活和活性氧(ROS)上调有关。此外,我们还检测了 MG132 和顺铂联合给药期间泛素-蛋白酶体系统的抑制作用以及促炎细胞因子 IL-1β、IL-6、IL-8 和 IL-13 的表达:结果:暴露于 MG132 会以剂量依赖的方式明显降低细胞活力。它增强了顺铂诱导的增殖抑制和细胞凋亡,与 Caspase-3 激活和 ROS 上调相关。分子分析表明,MG132 对泛素-蛋白酶体系统有很强的抑制作用。MG132 还能明显增加顺铂诱导的促炎细胞因子的表达,表明炎症从慢性向急性过渡:结论:MG132通过抑制泛素-蛋白酶体途径、降低细胞活力、增强细胞凋亡和改变炎症反应,增强了顺铂对人类EC细胞的疗效。这些发现凸显了 MG132 作为子宫内膜癌化疗辅助药物的潜力。还需要进一步的研究来探索这种联合疗法的详细机制和临床应用。
{"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 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-10-01 DOI: 10.2174/0118715206315415240830052608
Venkata Sowjanya Thanneeru, Naresh Panigrahi

Background: 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.

Objective: This study presents the synthesis, characterization, and evaluation of novel quinoline nitrate derivatives as potential anticancer agents.

Methods: 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.

Results: 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.

Conclusion: 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.

背景:一氧化氮(NO)近来已被公认为是癌症治疗领域一种前景广阔的方法。喹啉支架在癌症药物研究中举足轻重,以其多功能性和作用机制多样性而著称:本研究介绍了新型硝酸喹啉衍生物作为潜在抗癌药物的合成、表征和评估:这些化合物是通过多步骤合成的,包括制备取代的 1-(2-氨基苯基)乙-1-酮,然后合成取代的 2-(氯甲基)-3,4-二甲基喹啉,最后形成取代的(3,4-二甲基喹啉-2-基)甲基硝酸酯衍生物。利用各种光谱技术对合成的化合物进行了表征。进行了分子对接研究,以评估化合物与表皮生长因子受体酪氨酸激酶结构域的结合亲和力:对接得分显示了不同程度的结合亲和力,其中化合物 6k 的得分最高。结果表明,分子对接得分与抗癌活性之间存在相关性。进一步的评估包括 MTT 试验,以确定化合物对非小细胞肺癌(A-549)和胰腺癌(PANC-1)细胞系的细胞毒性。带有电子供能基团的化合物具有显著的抗癌潜力,一氧化氮的释放与抗癌活性之间存在相关性。研究还调查了化合物的一氧化氮释放情况,结果显示化合物 6g 的一氧化氮释放量最高:结论:合成的硝酸喹啉衍生物显示出良好的抗癌活性,其中化合物 6g 是一个潜在的先导化合物。分子对接、一氧化氮释放和抗癌活性之间的相关性表明,特定的结构特征在设计有效抗癌剂方面具有重要意义。
{"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 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-09-24 DOI: 10.2174/0118715206333399240912071555
Mohammed Merae Alshahrani

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.

癌症是一种复杂而可怕的疾病,它的表现形式多种多样,病情发展无情,不断挑战着医学科学。在寻求新型治疗策略的过程中,有机杂环化合物因其多变的化学结构以及与生物系统复杂的相互作用而成为前景广阔的候选化合物。其中,吡嗪衍生物的特点是具有一个六元芳香环,其中四个碳原子和两个氮原子呈 1,4 取向分布。这些化合物因其作为抗癌剂的潜力而备受关注。本综述详细分析了 2010 年至 2024 年这一时期的研究进展,包括吡嗪衍生物的化学多样性、它们在细胞水平的作用机制以及结构-活性关系。通过研究吡嗪衍生物的治疗潜力、挑战和未来前景,本综述为了解吡嗪衍生物作为抗癌利器的发展状况提供了宝贵的见解。
{"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 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-09-24 DOI: 10.2174/0118715206330232240913100744
Preeti Kumari, Rakhi Mishra, Rupa Mazumder, Avijit Mazumder

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.

酰基脲化合物在癌症治疗中备受关注,尤其是对主要影响女性的癌症(如乳腺癌和卵巢癌)的潜在疗效。本文报告了酰基脲化合物的研究情况,据说涉及到合成化学、生物检测和计算建模等多方面的方法。本文从不同来源收集了大量有关酰基脲及其对女性癌症的所谓作用的信息,并将其浓缩,以便读者广泛了解酰基脲在抗癌方面的作用。酰基脲通过选择性抑制与癌症进展相关的关键分子靶点,如表皮生长因子受体、ALK、HER2 和 Wnt/β-catenin 信号通路,显示出良好的效果。具体来说,以酰基脲为靶点可阻碍肿瘤增殖和转移,同时最大限度地减少对健康组织的伤害,提供了一种与传统化疗相比副作用更小的靶向治疗方法。为了优化酰基脲类疗法的疗效和安全性,扩大其在各种癌症类型中的适用性,继续开展研究和临床试验势在必行。酰基脲化合物是治疗女性癌症的一类前景广阔的疗法,特别是因为它们能够选择性地抑制参与肿瘤生长和恶化的关键分子靶点。通过合成优化、生物评估和计算建模的结合,我们发现了几种具有显著抗癌潜力的先导化合物。本摘要探讨了酰基脲类化合物在抗击这些恶性肿瘤方面的治疗机制和靶向途径,这将有助于今后的研究。
{"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
Pioneering the Battle Against Breast Cancer: The Promise of New Bcl-2 Family. 抗击乳腺癌的先锋:新 Bcl-2 家族的希望。
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-09-20 DOI: 10.2174/0118715206320224240910054728
Ali Farhang Boroujeni, Zeynep Ates-Alagoz

Currently, breast cancer is the most common cancer type, accounting for 1 in every 4 cancer cases. Leading both in mortality and incidence, breast cancer causes 1 in 4 cancer deaths. To decrease the burden of breast cancer, novel therapeutic agents which target the key hallmarks of cancer, are being explored. The Bcl-2 family of proteins has a crucial role in governing cell death, making them an attractive target for cancer therapy. As cancer chemotherapies lead to oncogenic stress, cancer cells upregulate the Bcl-2 family to overcome apoptosis, leading to failure of treatment. To fix this issue, Bcl-2 family inhibitors, which can cause cell death, have been introduced as novel therapeutic agents. Members of this group have shown promising results in in-vitro studies, and some are currently in clinical trials. In this review, we will investigate Bcl-2 family inhibitors, which are already in trials as monotherapy or combination therapy for breast cancer, and we will also highlight the result of in vitro studies of novel Bcl-2 family inhibitors on breast cancer cells. The findings of these studies have yielded encouraging outcomes regarding the identification of novel Bcl-2 family inhibitors. These compounds hold significant potential as efficacious agents for employment in both monotherapy and combination therapy settings.

目前,乳腺癌是最常见的癌症类型,每 4 个癌症病例中就有 1 个是乳腺癌。乳腺癌的死亡率和发病率均居首位,每 4 例癌症死亡病例中就有 1 例死于乳腺癌。为了减轻乳腺癌的负担,人们正在探索针对癌症关键特征的新型治疗药物。Bcl-2 蛋白家族在控制细胞死亡方面起着至关重要的作用,因此成为癌症治疗的一个有吸引力的靶点。由于癌症化疗会导致致癌压力,癌细胞会上调 Bcl-2 家族以克服细胞凋亡,从而导致治疗失败。为了解决这个问题,Bcl-2 家族抑制剂作为新型治疗药物问世,可导致细胞死亡。这类抑制剂在体外研究中显示出良好的效果,其中一些目前正在进行临床试验。在这篇综述中,我们将探讨已经作为乳腺癌单一疗法或联合疗法进行试验的 Bcl-2 家族抑制剂,并重点介绍新型 Bcl-2 家族抑制剂对乳腺癌细胞进行体外研究的结果。这些研究结果为确定新型 Bcl-2 家族抑制剂提供了令人鼓舞的成果。这些化合物极有可能成为单药治疗和联合治疗的有效药物。
{"title":"Pioneering the Battle Against Breast Cancer: The Promise of New Bcl-2 Family.","authors":"Ali Farhang Boroujeni, Zeynep Ates-Alagoz","doi":"10.2174/0118715206320224240910054728","DOIUrl":"https://doi.org/10.2174/0118715206320224240910054728","url":null,"abstract":"<p><p>Currently, breast cancer is the most common cancer type, accounting for 1 in every 4 cancer cases. Leading both in mortality and incidence, breast cancer causes 1 in 4 cancer deaths. To decrease the burden of breast cancer, novel therapeutic agents which target the key hallmarks of cancer, are being explored. The Bcl-2 family of proteins has a crucial role in governing cell death, making them an attractive target for cancer therapy. As cancer chemotherapies lead to oncogenic stress, cancer cells upregulate the Bcl-2 family to overcome apoptosis, leading to failure of treatment. To fix this issue, Bcl-2 family inhibitors, which can cause cell death, have been introduced as novel therapeutic agents. Members of this group have shown promising results in in-vitro studies, and some are currently in clinical trials. In this review, we will investigate Bcl-2 family inhibitors, which are already in trials as monotherapy or combination therapy for breast cancer, and we will also highlight the result of in vitro studies of novel Bcl-2 family inhibitors on breast cancer cells. The findings of these studies have yielded encouraging outcomes regarding the identification of novel Bcl-2 family inhibitors. These compounds hold significant potential as efficacious agents for employment in both monotherapy and combination therapy settings.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2024-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142306980","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
Alterations of Krüppel-like Factor Signaling and Potential Targeted Therapy for Hepatocellular Carcinoma. 克鲁珀尔样因子信号的改变与肝细胞癌的潜在靶向疗法
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-09-20 DOI: 10.2174/0118715206301453240910044913
Rongfei Fang, Chunxiu Sha, Qun Xie, Min Yao, Dengfu Yao

Krüppel-like factors (KLFs, total 18 members) from the zinc finger protein (ZFP) super-family have a wide range of biological functions in hepatocellular carcinoma (HCC). This paper reviews the recent some progresses of aberrant KLFs with their potential values for diagnosis, prognosis, and targeted therapy in HCC. The recent advances of oncogenic KLFs in the diagnosis, prognosis, and targeted therapy of HCC were reviewed based on the related literature on PUBMED and clinical investigation. Based on the recent literature, KLFs, according to biological functions in HCC, are divided into 4 subgroups: promoting (KLF5, 7, 8, 13), inhibiting (KLF3, 4, 9~12, 14,17), dual (KLF2,6), and unknown functions (KLF1, 15, 16, or 18 ?). HCC-related KLFs regulate downstream gene transcription during hepatocyte malignant transformation, participating in cell proliferation, apoptosis, invasion, and metastasis. Some KLFs have diagnostic or prognostic value, and other KLFs with inhibiting promoting function or over-expressing inhibiting roles might be molecular targets for HCC therapy. These data have suggested that Abnormal expressions of KLFs were associated with HCC progression. Among them, some KLFs have revealed the clinical values of diagnosis or prognosis, and other KLFs with the biological functions of promotion or inhibition might be as effectively molecular targets for HCC therapy.

锌指蛋白(ZFP)超家族中的Krüppel样因子(KLFs,共有18个成员)在肝细胞癌(HCC)中具有广泛的生物学功能。本文回顾了异常 KLFs 的最新研究进展及其在 HCC 诊断、预后和靶向治疗中的潜在价值。根据 PUBMED 上的相关文献和临床研究,综述了致癌 KLFs 在 HCC 诊断、预后和靶向治疗方面的最新进展。根据最新文献,KLFs 在 HCC 中的生物学功能可分为 4 个亚组:促进(KLF5、7、8、13)、抑制(KLF3、4、9~12、14、17)、双重(KLF2、6)和未知功能(KLF1、15、16 或 18?)与 HCC 相关的 KLFs 在肝细胞恶性转化过程中调节下游基因转录,参与细胞增殖、凋亡、侵袭和转移。一些 KLFs 具有诊断或预后价值,其他具有抑制促进功能或过度表达抑制作用的 KLFs 可能是治疗 HCC 的分子靶点。这些数据表明,KLFs 的异常表达与 HCC 的进展有关。其中,一些KLFs具有诊断或预后的临床价值,而其他具有促进或抑制生物学功能的KLFs则可能成为治疗HCC的有效分子靶点。
{"title":"Alterations of Krüppel-like Factor Signaling and Potential Targeted Therapy for Hepatocellular Carcinoma.","authors":"Rongfei Fang, Chunxiu Sha, Qun Xie, Min Yao, Dengfu Yao","doi":"10.2174/0118715206301453240910044913","DOIUrl":"https://doi.org/10.2174/0118715206301453240910044913","url":null,"abstract":"<p><p>Krüppel-like factors (KLFs, total 18 members) from the zinc finger protein (ZFP) super-family have a wide range of biological functions in hepatocellular carcinoma (HCC). This paper reviews the recent some progresses of aberrant KLFs with their potential values for diagnosis, prognosis, and targeted therapy in HCC. The recent advances of oncogenic KLFs in the diagnosis, prognosis, and targeted therapy of HCC were reviewed based on the related literature on PUBMED and clinical investigation. Based on the recent literature, KLFs, according to biological functions in HCC, are divided into 4 subgroups: promoting (KLF5, 7, 8, 13), inhibiting (KLF3, 4, 9~12, 14,17), dual (KLF2,6), and unknown functions (KLF1, 15, 16, or 18 ?). HCC-related KLFs regulate downstream gene transcription during hepatocyte malignant transformation, participating in cell proliferation, apoptosis, invasion, and metastasis. Some KLFs have diagnostic or prognostic value, and other KLFs with inhibiting promoting function or over-expressing inhibiting roles might be molecular targets for HCC therapy. These data have suggested that Abnormal expressions of KLFs were associated with HCC progression. Among them, some KLFs have revealed the clinical values of diagnosis or prognosis, and other KLFs with the biological functions of promotion or inhibition might be as effectively molecular targets for HCC therapy.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2024-09-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142306979","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
期刊
Anti-cancer agents in medicinal chemistry
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