首页 > 最新文献

Anti-cancer agents in medicinal chemistry最新文献

英文 中文
Drug Repositioning for Ovarian Cancer Treatment: An Update 卵巢癌治疗药物的重新定位:最新进展
IF 2.8 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-02-16 DOI: 10.2174/0118715206282904240122063914
Maria Maddalena Cavalluzzi, Maurizio Viale, Natalie Paola Rotondo, Valeria Ferraro, Giovanni Lentini
: Ovarian cancer (OC) is one of the most prevalent malignancies in female reproductive organs, and its 5-year survival is below 45%. Despite the advances in surgical and chemotherapeutic options, OC treatment is still a challenge, and new anticancer agents are urgently needed. Drug repositioning has gained significant attention in drug discovery, representing a smart way to identify new clinical applications for drugs whose human safety and pharmacokinetics have already been established, with great time and cost savings in pharmaceutical development endeavors. This review offers an update on the most promising drugs repurposable for OC treatment and/or prevention.
:卵巢癌(OC)是女性生殖器官中最常见的恶性肿瘤之一,其 5 年生存率低于 45%。尽管在手术和化疗方面取得了进展,但卵巢癌的治疗仍然是一项挑战,迫切需要新的抗癌药物。药物重新定位在新药研发中备受关注,它是为人体安全性和药代动力学已经确定的药物寻找新的临床应用领域的明智之举,大大节省了药物研发的时间和成本。本综述介绍了可重新用于治疗和/或预防 OC 的最有前途药物的最新情况。
{"title":"Drug Repositioning for Ovarian Cancer Treatment: An Update","authors":"Maria Maddalena Cavalluzzi, Maurizio Viale, Natalie Paola Rotondo, Valeria Ferraro, Giovanni Lentini","doi":"10.2174/0118715206282904240122063914","DOIUrl":"https://doi.org/10.2174/0118715206282904240122063914","url":null,"abstract":": Ovarian cancer (OC) is one of the most prevalent malignancies in female reproductive organs, and its 5-year survival is below 45%. Despite the advances in surgical and chemotherapeutic options, OC treatment is still a challenge, and new anticancer agents are urgently needed. Drug repositioning has gained significant attention in drug discovery, representing a smart way to identify new clinical applications for drugs whose human safety and pharmacokinetics have already been established, with great time and cost savings in pharmaceutical development endeavors. This review offers an update on the most promising drugs repurposable for OC treatment and/or prevention.","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":"24 1","pages":""},"PeriodicalIF":2.8,"publicationDate":"2024-02-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139772302","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
Targeting Breast Cancer with N-Acetyl-D-Glucosamine: Integrating Machine Learning and Cellular Assays for Promising Results 用 N-乙酰-D-氨基葡萄糖靶向乳腺癌:整合机器学习和细胞检测,取得可喜成果
IF 2.8 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-02-02 DOI: 10.2174/0118715206270568231129054853
Ömür Baysal, Deniz Genç, Ragıp Soner Silme, Kevser Kübra Kırboğa, Dilek Çoban, Naeem Abdul Ghafoor, Leyla Tekin, Osman Bulut
Background:: Breast cancer is a common cancer with high mortality rates. Early diagnosis is crucial for reducing the prognosis and mortality rates. Therefore, the development of alternative treatment options is necessary. Objective:: This study aimed to investigate the inhibitory effect of N-acetyl-D-glucosamine (D-GlcNAc) on breast cancer using a machine learning method. The findings were further confirmed through assays on breast cancer cell lines. Methods:: MCF-7 and 4T1 cell lines (ATCC) were cultured in the presence and absence of varying concentrations of D-GlcNAc (0.5 mM, 1 mM, 2 mM, and 4 mM) for 72 hours. A xenograft mouse model for breast cancer was established by injecting 4T1 cells into mammary glands. D-GlcNAc (2 mM) was administered intraperitoneally to mice daily for 28 days, and histopathological effects were evaluated at pre-tumoral and post-tumoral stages. Results:: Treatment with 2 mM and 4 mM D-GlcNAc significantly decreased cell proliferation rates in MCF-7 and 4T1 cell lines and increased Fas expression. The number of apoptotic cells was significantly higher than untreated cell cultures (p < 0.01 - p < 0.0001). D-GlcNAc administration also considerably reduced tumour size, mitosis, and angiogenesis in the post-treatment group compared to the control breast cancer group (p < 0.01 - p < 0.0001). Additionally, molecular docking/dynamic analysis revealed a high binding affinity of D-GlcNAc to the marker protein HER2, which is involved in tumour progression and cell signalling. Conclusion:: Our study demonstrated the positive effect of D-GlcNAc administration on breast cancer cells, leading to increased apoptosis and Fas expression in the malignant phenotype. The binding affinity of D-GlcNAc to HER2 suggests a potential mechanism of action. These findings contribute to understanding D-GlcNAc as a potential anti-tumour agent for breast cancer treatment.
背景乳腺癌是一种死亡率很高的常见癌症。早期诊断是降低预后和死亡率的关键。因此,有必要开发替代治疗方案。目的本研究旨在利用机器学习方法研究 N-乙酰-D-葡萄糖胺(D-GlcNAc)对乳腺癌的抑制作用。通过对乳腺癌细胞系的检测进一步证实了研究结果。方法::将 MCF-7 和 4T1 细胞系(ATCC)在不同浓度的 D-GlcNAc(0.5 mM、1 mM、2 mM 和 4 mM)存在和不存在的情况下培养 72 小时。将 4T1 细胞注射到乳腺中,建立了乳腺癌异种移植小鼠模型。每天给小鼠腹腔注射 D-GlcNAc(2 毫摩尔),连续 28 天,并评估瘤前和瘤后阶段的组织病理学效应。结果显示2 mM 和 4 mM D-GlcNAc 能显著降低 MCF-7 和 4T1 细胞系的细胞增殖率,并增加 Fas 的表达。凋亡细胞的数量明显高于未处理的细胞培养物(p < 0.01 - p < 0.0001)。与对照乳腺癌组相比,D-GlcNAc 给药也大大减少了治疗后乳腺癌组的肿瘤大小、有丝分裂和血管生成(p < 0.01 - p < 0.0001)。此外,分子对接/动态分析显示,D-GlcNAc 与标志蛋白 HER2 有很高的结合亲和力,而 HER2 参与肿瘤进展和细胞信号传导。结论我们的研究表明,服用 D-GlcNAc 对乳腺癌细胞有积极影响,可导致细胞凋亡和恶性表型中 Fas 表达的增加。D-GlcNAc 与 HER2 的结合亲和力提示了一种潜在的作用机制。这些发现有助于人们了解 D-GlcNAc 作为一种潜在的抗肿瘤药物对乳腺癌治疗的作用。
{"title":"Targeting Breast Cancer with N-Acetyl-D-Glucosamine: Integrating Machine Learning and Cellular Assays for Promising Results","authors":"Ömür Baysal, Deniz Genç, Ragıp Soner Silme, Kevser Kübra Kırboğa, Dilek Çoban, Naeem Abdul Ghafoor, Leyla Tekin, Osman Bulut","doi":"10.2174/0118715206270568231129054853","DOIUrl":"https://doi.org/10.2174/0118715206270568231129054853","url":null,"abstract":"Background:: Breast cancer is a common cancer with high mortality rates. Early diagnosis is crucial for reducing the prognosis and mortality rates. Therefore, the development of alternative treatment options is necessary. Objective:: This study aimed to investigate the inhibitory effect of N-acetyl-D-glucosamine (D-GlcNAc) on breast cancer using a machine learning method. The findings were further confirmed through assays on breast cancer cell lines. Methods:: MCF-7 and 4T1 cell lines (ATCC) were cultured in the presence and absence of varying concentrations of D-GlcNAc (0.5 mM, 1 mM, 2 mM, and 4 mM) for 72 hours. A xenograft mouse model for breast cancer was established by injecting 4T1 cells into mammary glands. D-GlcNAc (2 mM) was administered intraperitoneally to mice daily for 28 days, and histopathological effects were evaluated at pre-tumoral and post-tumoral stages. Results:: Treatment with 2 mM and 4 mM D-GlcNAc significantly decreased cell proliferation rates in MCF-7 and 4T1 cell lines and increased Fas expression. The number of apoptotic cells was significantly higher than untreated cell cultures (p &lt; 0.01 - p &lt; 0.0001). D-GlcNAc administration also considerably reduced tumour size, mitosis, and angiogenesis in the post-treatment group compared to the control breast cancer group (p &lt; 0.01 - p &lt; 0.0001). Additionally, molecular docking/dynamic analysis revealed a high binding affinity of D-GlcNAc to the marker protein HER2, which is involved in tumour progression and cell signalling. Conclusion:: Our study demonstrated the positive effect of D-GlcNAc administration on breast cancer cells, leading to increased apoptosis and Fas expression in the malignant phenotype. The binding affinity of D-GlcNAc to HER2 suggests a potential mechanism of action. These findings contribute to understanding D-GlcNAc as a potential anti-tumour agent for breast cancer treatment.","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":"180 1","pages":""},"PeriodicalIF":2.8,"publicationDate":"2024-02-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139664824","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
Cryptolepine Analog exhibits Antitumor Activity against Ehrlich Ascites Carcinoma Cells in Mice via Targeting Cell Growth, Oxidative Stress, and PTEN/Akt/mTOR Signaling Pathway 隐托拉平类似物通过靶向细胞生长、氧化应激和 PTEN/Akt/mTOR 信号通路对小鼠艾氏腹水癌细胞表现出抗肿瘤活性
IF 2.8 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-02-02 DOI: 10.2174/0118715206274318231128072821
Bishoy El-Aarag, Eman S. Shalaan, Abdullah A.S. Ahmed, Ibrahim El Tantawy El Sayed, Wafaa M. Ibrahim
Background: The efficacy of chemotherapy continues to be limited due to associated toxicity and chemoresistance. Thus, synthesizing and investigating novel agents for cancer treatment that could potentially eliminate such limitations is imperative. Objective: The current study aims to explore the anticancer potency of cryptolepine (CPE) analog on Ehrlich ascites carcinoma cells (EACs) in mice. Methods: The effect of a CPE analog on EAC cell viability and ascites volume, as well as malonaldehyde, total antioxidant capacity, and catalase, were estimated. The concentration of caspase-8 and mTOR in EACs was also measured, and the expression levels of PTEN and Akt were determined. Results: Results revealed that CPE analog exerts a cytotoxic effect on EAC cell viability and reduces the ascites volume. Moreover, this analog induces oxidative stress in EACs by increasing the level of malonaldehyde and decreasing the level of total antioxidant capacity and catalase activity. It also induces apoptosis by elevating the concentration of caspase-8 in EACs. Furthermore, it decreases the concentration of mTOR in EACs. Moreover, it upregulates the expression of PTEN and downregulates the expression of Akt in EACs. result: Results revealed that CPE analog exerts a cytotoxic effect on EAC cell viability and reduces the ascites volume. Moreover, this analog induces oxidative stress in EACs by increasing the level of malonaldehyde and decreasing the level of total antioxidant capacity and catalase activity. It also induces apoptosis by elevating the concentration of caspase-8 in EACs. Furthermore, it decreases the concentration of mTOR in EACs. Moreover, it upregulates the expression of PTEN and downregulates the expression of Akt in EACs. Conclusion: Our findings showed the anticancer activity of CPE analog against EACs in mice mediated by regulation of the PTEN/Akt/mTOR signaling pathway. conclusion: Our findings showed the anticancer activity of newly CPE analog against EACs in mice mediated by regulation of PTEN/Akt/mTOR signaling pathway.
背景:由于相关毒性和化疗耐药性,化疗的疗效仍然有限。因此,合成和研究可能消除这些限制的新型癌症治疗药物势在必行。研究目的本研究旨在探讨隐托拉平(CPE)类似物对小鼠艾氏腹水癌细胞(EACs)的抗癌效力。研究方法评估 CPE 类似物对 EAC 细胞活力和腹水体积的影响,以及对丙二醛、总抗氧化能力和过氧化氢酶的影响。还测量了 EAC 细胞中 Caspase-8 和 mTOR 的浓度,并测定了 PTEN 和 Akt 的表达水平。结果显示结果表明,CPE 类似物对 EAC 细胞活力具有细胞毒性作用,并能减少腹水体积。此外,该类似物通过增加丙二醛水平、降低总抗氧化能力和过氧化氢酶活性水平诱导 EAC 细胞氧化应激。它还通过提高 EAC 中 caspase-8 的浓度诱导细胞凋亡。此外,它还会降低 EAC 中 mTOR 的浓度。此外,它还能上调 EAC 中 PTEN 的表达,下调 Akt 的表达:结果表明,CPE 类似物对 EAC 细胞活力具有细胞毒性作用,并能减少腹水体积。此外,该类似物通过增加丙二醛水平、降低总抗氧化能力和过氧化氢酶活性水平,诱导 EAC 细胞氧化应激。它还通过提高 EAC 中 caspase-8 的浓度诱导细胞凋亡。此外,它还会降低 EAC 中 mTOR 的浓度。此外,它还能上调 EAC 中 PTEN 的表达,下调 Akt 的表达。结论我们的研究结果表明,CPE 类似物对小鼠 EACs 的抗癌活性是通过调节 PTEN/Akt/mTOR 信号通路介导的:我们的研究结果表明,新CPE类似物对小鼠EACs的抗癌活性是通过调节PTEN/Akt/mTOR信号通路介导的。
{"title":"Cryptolepine Analog exhibits Antitumor Activity against Ehrlich Ascites Carcinoma Cells in Mice via Targeting Cell Growth, Oxidative Stress, and PTEN/Akt/mTOR Signaling Pathway","authors":"Bishoy El-Aarag, Eman S. Shalaan, Abdullah A.S. Ahmed, Ibrahim El Tantawy El Sayed, Wafaa M. Ibrahim","doi":"10.2174/0118715206274318231128072821","DOIUrl":"https://doi.org/10.2174/0118715206274318231128072821","url":null,"abstract":"Background: The efficacy of chemotherapy continues to be limited due to associated toxicity and chemoresistance. Thus, synthesizing and investigating novel agents for cancer treatment that could potentially eliminate such limitations is imperative. Objective: The current study aims to explore the anticancer potency of cryptolepine (CPE) analog on Ehrlich ascites carcinoma cells (EACs) in mice. Methods: The effect of a CPE analog on EAC cell viability and ascites volume, as well as malonaldehyde, total antioxidant capacity, and catalase, were estimated. The concentration of caspase-8 and mTOR in EACs was also measured, and the expression levels of PTEN and Akt were determined. Results: Results revealed that CPE analog exerts a cytotoxic effect on EAC cell viability and reduces the ascites volume. Moreover, this analog induces oxidative stress in EACs by increasing the level of malonaldehyde and decreasing the level of total antioxidant capacity and catalase activity. It also induces apoptosis by elevating the concentration of caspase-8 in EACs. Furthermore, it decreases the concentration of mTOR in EACs. Moreover, it upregulates the expression of PTEN and downregulates the expression of Akt in EACs. result: Results revealed that CPE analog exerts a cytotoxic effect on EAC cell viability and reduces the ascites volume. Moreover, this analog induces oxidative stress in EACs by increasing the level of malonaldehyde and decreasing the level of total antioxidant capacity and catalase activity. It also induces apoptosis by elevating the concentration of caspase-8 in EACs. Furthermore, it decreases the concentration of mTOR in EACs. Moreover, it upregulates the expression of PTEN and downregulates the expression of Akt in EACs. Conclusion: Our findings showed the anticancer activity of CPE analog against EACs in mice mediated by regulation of the PTEN/Akt/mTOR signaling pathway. conclusion: Our findings showed the anticancer activity of newly CPE analog against EACs in mice mediated by regulation of PTEN/Akt/mTOR signaling pathway.","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":"36 1","pages":""},"PeriodicalIF":2.8,"publicationDate":"2024-02-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139664589","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
Synthesis of a Novel Gold(I) Complex and Evaluation of Its Anticancer Properties in Breast Cancer Cells 一种新型金(I)配合物的合成及其在乳腺癌细胞中的抗癌性能评估
IF 2.8 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-02-02 DOI: 10.2174/0118715206281182231127113608
Haseeb A. Khan, Anvarhusein A. Isab, Abdullah S. Alhomida, Mansour K. Gatasheh, Ali Al-Hoshani, Bashayr A. Aldhafeeri, N Rajendra Prasad
Background: Platinum complexes are commonly used for cancer chemotherapy; however, they are not only highly-priced but also have various side effects. It is, therefore, important to design affordable anticancer drugs with minimal side effects. background: Platinum complexes are commonly used for cancer chemotherapy however they are not only highly priced but also have various side effects. It is therefore important to design affordable anticancer drugs with minimal side effects. Methods: We synthesized a new gold(I) complex, PF6{(BDPEA)(TPPMS) digold(I)} (abbreviated as PBTDG) and tested its cytotoxicity of MCF-7 breast cancer cells. We also evaluated the effects of PBTDG on mitochondrial membrane potential, generation of reactive oxygen species (ROS) and apoptosis in breast cancer cells. objective: To synthesize a novel gold(I) complex and test its cytotoxicity and apoptotic potential in MCF-7 breast cancer cells. Results: The IC50 values for PBTDG and sorafenib were found to be 1.48 μM and 4.45 μM, respectively. Exposure to PBTDG caused significant and concentration-dependent depletion of ATP and disruption of mitochondrial membrane potential. PBTDG induced 2.6, 3.6, and 5.7-fold apoptosis for 1 μM, 3 μM, and 10 μM concentrations, respectively. The induction of apoptosis by the same concentrations of sorafenib was 1.2, 1.3, and 1.6-fold, respectively. The low concentration of PBTDG (1 μM) induced the generation of ROS by 99.83%, which was significantly higher than the ROS generation caused by the same concentration of sorafenib (73.76%). The ROS induction caused by higher concentrations (5 μM) of PBTDG and sorafenib were 104.95% and 122.11%, respectively. method: We synthesized a new gold(I) complex, PF6{(BDPEA)(TPPMS)digold(I)} (abbreviated as PBTDG) and tested the cytotoxicity in MCF-7 breast cancer cells using MTT assay. We used spectrophotometry for ATP analysis and flow cytometry for mitochondrial potential, apoptosis and ROS analyses. Conclusion: The lower concentration of PBTDG produced similar cytotoxicity and apoptotic effects that were caused by a comparatively higher concentration of known anticancer drug (sorafenib). The anticancer effects of PBTDG are attributed to its tendency to disrupt mitochondrial membrane potential, induction of apoptosis and generation of ROS. Further studies are warranted to test the anticancer effects of PBTDG in animal models of cancer. result: The IC50 values for PBTDG and sorafenib were found to be 1.48 μM and 4.45 μM, respectively. Exposure to PBTDG caused significant and concentration-dependent depletion of ATP and disruption of mitochondrial membrane potential. PBTDG induced 2.6, 3.6, and 5.7-folds apoptosis for 1 µM, 3 µM, and 10 µM concentrations, respectively. The induction of apoptosis by same concentrations of sorafenib was 1.2, 1.3, and 1.6-folds, respectively. The low concentration of PBTDG (1 µM) induced the generation of reactive oxygen species (ROS) by 99.83% which was significantly higher
背景:铂络合物常用于癌症化疗;然而,它们不仅价格昂贵,而且有各种副作用。因此,设计副作用最小、价格合理的抗癌药物非常重要:铂络合物常用于癌症化疗,但它们不仅价格昂贵,而且有各种副作用。因此,设计价格合理、副作用最小的抗癌药物非常重要。方法:我们合成了一种新的金(I)复合物 PF6{(BDPEA)(TPPMS) digold(I)}(简称 PBTDG),并测试了它对 MCF-7 乳腺癌细胞的细胞毒性。我们还评估了 PBTDG 对线粒体膜电位、活性氧(ROS)生成和乳腺癌细胞凋亡的影响:合成一种新型金(I)复合物,并测试其对 MCF-7 乳腺癌细胞的细胞毒性和凋亡潜能。结果发现 PBTDG 和索拉非尼的 IC50 值分别为 1.48 μM 和 4.45 μM。暴露于 PBTDG 会导致 ATP 的显著消耗和线粒体膜电位的破坏,且呈浓度依赖性。浓度分别为 1 μM、3 μM 和 10 μM 的 PBTDG 可诱导 2.6 倍、3.6 倍和 5.7 倍的细胞凋亡。相同浓度的索拉非尼对细胞凋亡的诱导分别为 1.2 倍、1.3 倍和 1.6 倍。低浓度 PBTDG(1 μM)诱导产生的 ROS 为 99.83%,明显高于相同浓度索拉非尼诱导产生的 ROS(73.76%)。更高浓度(5 μM)的PBTDG和索拉非尼引起的ROS诱导率分别为104.95%和122.11%:我们合成了一种新的金(I)复合物--PF6{(BDPEA)(TPPMS)digold(I)}(缩写为 PBTDG),并用 MTT 法检测了其对 MCF-7 乳腺癌细胞的细胞毒性。我们使用分光光度法进行 ATP 分析,使用流式细胞仪进行线粒体电位、细胞凋亡和 ROS 分析。结论较低浓度的 PBTDG 产生的细胞毒性和凋亡效应与较高浓度的已知抗癌药物(索拉非尼)相似。PBTDG 的抗癌作用归因于其破坏线粒体膜电位、诱导细胞凋亡和产生 ROS 的倾向。有必要进一步研究 PBTDG 在癌症动物模型中的抗癌效果:发现 PBTDG 和索拉非尼的 IC50 值分别为 1.48 μM 和 4.45 μM。暴露于 PBTDG 会导致 ATP 的显著消耗和线粒体膜电位的破坏,且呈浓度依赖性。浓度为 1 µM、3 µM 和 10 µM 的 PBTDG 可分别诱导 2.6 倍、3.6 倍和 5.7 倍的细胞凋亡。相同浓度的索拉非尼诱导的细胞凋亡分别为 1.2、1.3 和 1.6 倍。低浓度的 PBTDG(1 µM)诱导产生的活性氧(ROS)为 99.83%,明显高于相同浓度的索拉非尼诱导产生的 ROS(73.76%)。更高浓度(5 µM)的 PBTDG 和索拉非尼引起的 ROS 诱导率分别为 104.95% 和 122.11%:有必要进一步研究 PBTDG 在癌症动物模型中的抗癌效果。
{"title":"Synthesis of a Novel Gold(I) Complex and Evaluation of Its Anticancer Properties in Breast Cancer Cells","authors":"Haseeb A. Khan, Anvarhusein A. Isab, Abdullah S. Alhomida, Mansour K. Gatasheh, Ali Al-Hoshani, Bashayr A. Aldhafeeri, N Rajendra Prasad","doi":"10.2174/0118715206281182231127113608","DOIUrl":"https://doi.org/10.2174/0118715206281182231127113608","url":null,"abstract":"Background: Platinum complexes are commonly used for cancer chemotherapy; however, they are not only highly-priced but also have various side effects. It is, therefore, important to design affordable anticancer drugs with minimal side effects. background: Platinum complexes are commonly used for cancer chemotherapy however they are not only highly priced but also have various side effects. It is therefore important to design affordable anticancer drugs with minimal side effects. Methods: We synthesized a new gold(I) complex, PF6{(BDPEA)(TPPMS) digold(I)} (abbreviated as PBTDG) and tested its cytotoxicity of MCF-7 breast cancer cells. We also evaluated the effects of PBTDG on mitochondrial membrane potential, generation of reactive oxygen species (ROS) and apoptosis in breast cancer cells. objective: To synthesize a novel gold(I) complex and test its cytotoxicity and apoptotic potential in MCF-7 breast cancer cells. Results: The IC50 values for PBTDG and sorafenib were found to be 1.48 μM and 4.45 μM, respectively. Exposure to PBTDG caused significant and concentration-dependent depletion of ATP and disruption of mitochondrial membrane potential. PBTDG induced 2.6, 3.6, and 5.7-fold apoptosis for 1 μM, 3 μM, and 10 μM concentrations, respectively. The induction of apoptosis by the same concentrations of sorafenib was 1.2, 1.3, and 1.6-fold, respectively. The low concentration of PBTDG (1 μM) induced the generation of ROS by 99.83%, which was significantly higher than the ROS generation caused by the same concentration of sorafenib (73.76%). The ROS induction caused by higher concentrations (5 μM) of PBTDG and sorafenib were 104.95% and 122.11%, respectively. method: We synthesized a new gold(I) complex, PF6{(BDPEA)(TPPMS)digold(I)} (abbreviated as PBTDG) and tested the cytotoxicity in MCF-7 breast cancer cells using MTT assay. We used spectrophotometry for ATP analysis and flow cytometry for mitochondrial potential, apoptosis and ROS analyses. Conclusion: The lower concentration of PBTDG produced similar cytotoxicity and apoptotic effects that were caused by a comparatively higher concentration of known anticancer drug (sorafenib). The anticancer effects of PBTDG are attributed to its tendency to disrupt mitochondrial membrane potential, induction of apoptosis and generation of ROS. Further studies are warranted to test the anticancer effects of PBTDG in animal models of cancer. result: The IC50 values for PBTDG and sorafenib were found to be 1.48 μM and 4.45 μM, respectively. Exposure to PBTDG caused significant and concentration-dependent depletion of ATP and disruption of mitochondrial membrane potential. PBTDG induced 2.6, 3.6, and 5.7-folds apoptosis for 1 µM, 3 µM, and 10 µM concentrations, respectively. The induction of apoptosis by same concentrations of sorafenib was 1.2, 1.3, and 1.6-folds, respectively. The low concentration of PBTDG (1 µM) induced the generation of reactive oxygen species (ROS) by 99.83% which was significantly higher ","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":"306 1","pages":""},"PeriodicalIF":2.8,"publicationDate":"2024-02-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139664828","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
Hyperoside Inhibits RNF8-mediated Nuclear Translocation of β-catenin to Repress PD-L1 Expression and Prostate Cancer 金丝桃苷抑制 RNF8 介导的 β-catenin 核转运,从而抑制 PD-L1 的表达和前列腺癌的发生
IF 2.8 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-02-02 DOI: 10.2174/0118715206289246240110044931
Jie Chen, Yi Zhao, Xiaoli Wang, Long Zang, Dengke Yin, Song Tan
Background: Hyperoside is a flavonol glycoside isolated from Hypericum perforatum L. that has inhibitory effects on cancer cells; however, its effects on prostate cancer (PCa) remain unclear. Therefore, we studied the anti-PCa effects of hyperoside and its underlying mechanisms in vitro and in vivo. Aim: This study aimed to explore the mechanism of hyperoside in anti-PCa. Methods: 3-(4,5-Dimethyl-2-Thiazolyl)-2,5-Diphenyl Tetrazolium Bromide (MTT), transwell, and flow cytometry assays were used to detect PCa cell growth, invasion, and cell apoptosis. Immunoblot analysis, immunofluorescence, immunoprecipitation, and quantitative real-time PCR (qRT-PCR) were used to analyze the antitumor mechanism of hyperoside. Results: Hyperoside inhibited PCa cell growth, invasion, and cell cycle and induced cell apoptosis. Furthermore, RING finger protein 8 (RNF8), an E3 ligase that assembles K63 polyubiquitination chains, was predicted to be a direct target of hyperoside and was downregulated by hyperoside. Downregulation of RNF8 by hyperoside impeded the nuclear translocation of β-catenin and disrupted the Wnt/β-catenin pathway, which reduced the expression of the target genes c-myc, cyclin D1, and programmed death ligand 1 (PD-L1). Decreased PD-L1 levels contributed to induced immunity in Jurkat cells in vitro. Finally, in vivo studies demonstrated that hyperoside significantly reduced tumor size, inhibited PD-L1 and RNF8 expression, and induced apoptosis in tumor tissues of a subcutaneous mouse model. Conclusion: Hyperoside exerts its anti-PCa effect by reducing RNF8 protein, inhibiting nuclear translocation of β-catenin, and disrupting the Wnt/β-catenin pathway, in turn reducing the expression of PD-L1 and improving Jurkat cell immunity.
背景:金丝桃苷是从贯叶连翘中分离出来的一种黄酮醇苷,对癌细胞有抑制作用;但它对前列腺癌(PCa)的作用仍不清楚。因此,我们研究了金丝桃苷在体外和体内的抗 PCa 作用及其内在机制。目的:本研究旨在探索金丝桃苷抗 PCa 的机制。方法:采用 3-(4,5-二甲基-2-噻唑基)-2,5-二苯基溴化四氮唑(MTT)、透孔法和流式细胞术检测 PCa 细胞的生长、侵袭和凋亡。免疫印迹分析、免疫荧光、免疫沉淀和定量实时 PCR(qRT-PCR)用于分析金丝桃苷的抗肿瘤机制。结果金丝桃苷抑制了PCa细胞的生长、侵袭和细胞周期,并诱导细胞凋亡。此外,RING 手指蛋白 8(RNF8)是一种组装 K63 多泛素化链的 E3 连接酶,被预测为金丝桃苷的直接靶标,并被金丝桃苷下调。高甙对RNF8的下调阻碍了β-catenin的核转位,破坏了Wnt/β-catenin通路,从而降低了靶基因c-myc、细胞周期蛋白D1和程序性死亡配体1(PD-L1)的表达。PD-L1水平的降低有助于诱导体外Jurkat细胞产生免疫力。最后,体内研究表明,金丝桃苷能显著缩小肿瘤大小,抑制 PD-L1 和 RNF8 的表达,并诱导皮下小鼠模型肿瘤组织中的细胞凋亡。结论金丝桃苷通过减少RNF8蛋白、抑制β-catenin的核转位和破坏Wnt/β-catenin通路,进而减少PD-L1的表达和提高Jurkat细胞的免疫力来发挥抗PCa作用。
{"title":"Hyperoside Inhibits RNF8-mediated Nuclear Translocation of β-catenin to Repress PD-L1 Expression and Prostate Cancer","authors":"Jie Chen, Yi Zhao, Xiaoli Wang, Long Zang, Dengke Yin, Song Tan","doi":"10.2174/0118715206289246240110044931","DOIUrl":"https://doi.org/10.2174/0118715206289246240110044931","url":null,"abstract":"Background: Hyperoside is a flavonol glycoside isolated from Hypericum perforatum L. that has inhibitory effects on cancer cells; however, its effects on prostate cancer (PCa) remain unclear. Therefore, we studied the anti-PCa effects of hyperoside and its underlying mechanisms in vitro and in vivo. Aim: This study aimed to explore the mechanism of hyperoside in anti-PCa. Methods: 3-(4,5-Dimethyl-2-Thiazolyl)-2,5-Diphenyl Tetrazolium Bromide (MTT), transwell, and flow cytometry assays were used to detect PCa cell growth, invasion, and cell apoptosis. Immunoblot analysis, immunofluorescence, immunoprecipitation, and quantitative real-time PCR (qRT-PCR) were used to analyze the antitumor mechanism of hyperoside. Results: Hyperoside inhibited PCa cell growth, invasion, and cell cycle and induced cell apoptosis. Furthermore, RING finger protein 8 (RNF8), an E3 ligase that assembles K63 polyubiquitination chains, was predicted to be a direct target of hyperoside and was downregulated by hyperoside. Downregulation of RNF8 by hyperoside impeded the nuclear translocation of β-catenin and disrupted the Wnt/β-catenin pathway, which reduced the expression of the target genes c-myc, cyclin D1, and programmed death ligand 1 (PD-L1). Decreased PD-L1 levels contributed to induced immunity in Jurkat cells in vitro. Finally, in vivo studies demonstrated that hyperoside significantly reduced tumor size, inhibited PD-L1 and RNF8 expression, and induced apoptosis in tumor tissues of a subcutaneous mouse model. Conclusion: Hyperoside exerts its anti-PCa effect by reducing RNF8 protein, inhibiting nuclear translocation of β-catenin, and disrupting the Wnt/β-catenin pathway, in turn reducing the expression of PD-L1 and improving Jurkat cell immunity.","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":"4 1","pages":""},"PeriodicalIF":2.8,"publicationDate":"2024-02-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139665066","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
Cordyceps militaris: A Comprehensive Study on Laboratory Cultivation and Anticancer Potential in Dalton's Ascites Lymphoma Tumor Model 冬虫夏草:实验室培养和道尔顿腹水淋巴瘤模型抗癌潜力的综合研究
IF 2.8 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-02-02 DOI: 10.2174/0118715206282174240115082518
Diksha Dutta, Namram Sushindrajit Singh, Rohit Aggarwal, Akalesh Kumar Verma
Background: Cancer, a predominant cause of mortality, poses a formidable challenge in our pursuit of elevating life expectancy. Throughout history, individuals have sought natural remedies with minimal side effects as an appealing substitute for chemotherapeutic drugs. One such remedy is Cordyceps militaris, a renowned medicinal mushroom deeply entrenched in Asian ethnomedicine. Revered for its rejuvenating and curative attributes, it relied upon for ages. Objective: The mushroom’s soaring demand outpaced natural availability, necessitating controlled laboratory cultivation as the core focus and exploring the potential of methanolic extracts from harvested Cordyceps militaris fruiting bodies against Dalton's Lymphoma Ascites (DLA) cells in vitro, with a specific emphasis on its anticancer traits. Methods: For cultivation, we employed a diverse range of rice substrates, among which bora rice showed promising growth of C. militaris fruiting bodies. To assess DLA cell cytotoxicity, several assays, including trypan blue exclusion assay, MTT assay, and LDH assay, were employed at different time points (24-96 h), which provided valuable insights on DLA cell viability and proliferation, shedding light on its therapeutic potential against cancer. Results: Our studies unveiled that methanolic extract prompts apoptosis in DLA cells via AO/EB dual staining, manifesting consistent apoptosis indicators such as membrane blebbing, chromatin condensation, nuclei fragmentation, and cellular shrinkage at 48-96 h of treatment. Furthermore, these striking repercussions of apoptosis were comprehended by an in silico approach having molecular docking simulation against antiapoptotic proteins like BCL-2, BCL-XL, MCL-1, BFL-1 & HSP100. Conclusion: Methanolic C. militaris extracts exhibited cytotoxicity and apoptotic alterations in DLA cells
背景:癌症是导致死亡的主要原因之一,对我们提高预期寿命提出了严峻的挑战。纵观历史,人们一直在寻找副作用最小的天然疗法来替代化疗药物。冬虫夏草就是这样一种疗法,它是一种在亚洲民族医学中根深蒂固的著名药用蘑菇。冬虫夏草具有返老还童和治疗疾病的功效,历来受到人们的推崇。目标:这种蘑菇的需求量急剧上升,超过了自然供应量,因此有必要将受控实验室栽培作为核心重点,并在体外探索从收获的冬虫夏草子实体中提取甲醇提取物对抗道尔顿淋巴瘤腹水(DLA)细胞的潜力,特别强调其抗癌特性。方法:在培养过程中,我们采用了多种水稻基质,其中波拉水稻对米氏蘑菇子实体的生长具有良好的促进作用。为了评估 DLA 细胞的细胞毒性,我们在不同的时间点(24-96 小时)采用了几种检测方法,包括胰蓝排除法、MTT 法和 LDH 法。研究结果我们的研究揭示了甲醇提取物通过 AO/EB 双重染色促使 DLA 细胞凋亡,在处理 48-96 h 时表现出一致的凋亡指标,如膜出血、染色质凝结、细胞核破碎和细胞萎缩。此外,这些令人震惊的细胞凋亡反响是通过针对抗凋亡蛋白(如 BCL-2、BCL-XL、MCL-1、BFL-1 & HSP100)进行分子对接模拟的硅学方法来理解的。结论甲醇 C. militaris 提取物在 DLA 细胞中表现出细胞毒性和凋亡改变
{"title":"Cordyceps militaris: A Comprehensive Study on Laboratory Cultivation and Anticancer Potential in Dalton's Ascites Lymphoma Tumor Model","authors":"Diksha Dutta, Namram Sushindrajit Singh, Rohit Aggarwal, Akalesh Kumar Verma","doi":"10.2174/0118715206282174240115082518","DOIUrl":"https://doi.org/10.2174/0118715206282174240115082518","url":null,"abstract":"Background: Cancer, a predominant cause of mortality, poses a formidable challenge in our pursuit of elevating life expectancy. Throughout history, individuals have sought natural remedies with minimal side effects as an appealing substitute for chemotherapeutic drugs. One such remedy is Cordyceps militaris, a renowned medicinal mushroom deeply entrenched in Asian ethnomedicine. Revered for its rejuvenating and curative attributes, it relied upon for ages. Objective: The mushroom’s soaring demand outpaced natural availability, necessitating controlled laboratory cultivation as the core focus and exploring the potential of methanolic extracts from harvested Cordyceps militaris fruiting bodies against Dalton's Lymphoma Ascites (DLA) cells in vitro, with a specific emphasis on its anticancer traits. Methods: For cultivation, we employed a diverse range of rice substrates, among which bora rice showed promising growth of C. militaris fruiting bodies. To assess DLA cell cytotoxicity, several assays, including trypan blue exclusion assay, MTT assay, and LDH assay, were employed at different time points (24-96 h), which provided valuable insights on DLA cell viability and proliferation, shedding light on its therapeutic potential against cancer. Results: Our studies unveiled that methanolic extract prompts apoptosis in DLA cells via AO/EB dual staining, manifesting consistent apoptosis indicators such as membrane blebbing, chromatin condensation, nuclei fragmentation, and cellular shrinkage at 48-96 h of treatment. Furthermore, these striking repercussions of apoptosis were comprehended by an in silico approach having molecular docking simulation against antiapoptotic proteins like BCL-2, BCL-XL, MCL-1, BFL-1 &amp; HSP100. Conclusion: Methanolic C. militaris extracts exhibited cytotoxicity and apoptotic alterations in DLA cells","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":"15 1","pages":""},"PeriodicalIF":2.8,"publicationDate":"2024-02-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139664717","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
Chalcones as Potential Cyclooxygenase-2 Inhibitors: A Review. 查尔酮作为环氧合酶-2抑制剂的研究进展
IF 2.8 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-01-01 DOI: 10.2174/0118715206267309231103053808
Mohammad Mahboubi-Rabbani, Rosa Zarei, Mehdi Baradaran, Maryam Bayanati, Afshin Zarghi

Cyclooxygenases (COXs) play a pivotal role in inflammation, a complex phenomenon required in human defense, but also involved in the emergence of insidious human disorders. Currently-used COX-1 inhibitors (Non-Steroidal Anti-Inflammatory Drugs-NSAIDs), as the most frequent choices for the treatment of chronic inflammatory diseases, have been identified to be associated with a variety of adverse drug reactions, especially dyspepsia, as well as peptic ulcer, which lead to diminished output. Moreover, the structural similarities of COX- 1 and -2, along with the availability of comprehensive information about the three-dimensional structure of COX- 2, co-crystallized with various inhibitors, search selective COX-2 inhibitors a formidable challenge. COX-2 inhibitors were shown to minimize the incidence of metastasis in cancer patients when administered preoperatively. Developing selective COX-2 inhibitors to tackle both cancer and chronic inflammatory illnesses has been identified as a promising research direction in recent decades. Identifying innovative scaffolds to integrate as the major component of future COX-2 inhibitors is critical in this regard. The presence of a central, α, β-unsaturated carbonyl- containing scaffold, as a characteristic structural pattern in many selective COX-2 inhibitors, along with a huge count of chalcone-based anticancer agents representing the basic idea of this review; providing a survey of the most recently published literature concerning development of chalcone analogs as novel COX-2 inhibitors until 2022 with efficient anticancer activity. A brief overview of the most recent developments concerning structure- activity relationship insights and mechanisms is also reported, helping pave the road for additional investigation.

环氧合酶(cox)在炎症中起着关键作用,炎症是人体防御所需的复杂现象,但也涉及潜在人类疾病的出现。目前使用的COX-1抑制剂(非甾体抗炎药- nsaids)作为治疗慢性炎症性疾病的最常用选择,已被确定与各种药物不良反应有关,特别是消化不良,以及消化性溃疡,导致输出减少。此外,COX- 1和COX-2结构的相似性,以及COX-2与各种抑制剂共晶的三维结构的全面信息的可用性,给寻找选择性COX-2抑制剂带来了艰巨的挑战。术前给予COX-2抑制剂可将癌症患者转移的发生率降至最低。近几十年来,开发选择性COX-2抑制剂来治疗癌症和慢性炎症疾病已被确定为一个有前途的研究方向。在这方面,确定创新的支架作为未来COX-2抑制剂的主要成分是至关重要的。中心β-不饱和羰基支架的存在,作为许多选择性COX-2抑制剂的特征结构模式,以及大量基于查尔酮的抗癌药物代表了本综述的基本思想;提供了最近发表的关于查尔酮类似物作为新型COX-2抑制剂发展的文献综述,直到2022年具有有效的抗癌活性。简要概述了有关结构-活性关系的见解和机制的最新发展,有助于为进一步的研究铺平道路。
{"title":"Chalcones as Potential Cyclooxygenase-2 Inhibitors: A Review.","authors":"Mohammad Mahboubi-Rabbani, Rosa Zarei, Mehdi Baradaran, Maryam Bayanati, Afshin Zarghi","doi":"10.2174/0118715206267309231103053808","DOIUrl":"10.2174/0118715206267309231103053808","url":null,"abstract":"<p><p>Cyclooxygenases (COXs) play a pivotal role in inflammation, a complex phenomenon required in human defense, but also involved in the emergence of insidious human disorders. Currently-used COX-1 inhibitors (Non-Steroidal Anti-Inflammatory Drugs-NSAIDs), as the most frequent choices for the treatment of chronic inflammatory diseases, have been identified to be associated with a variety of adverse drug reactions, especially dyspepsia, as well as peptic ulcer, which lead to diminished output. Moreover, the structural similarities of COX- 1 and -2, along with the availability of comprehensive information about the three-dimensional structure of COX- 2, co-crystallized with various inhibitors, search selective COX-2 inhibitors a formidable challenge. COX-2 inhibitors were shown to minimize the incidence of metastasis in cancer patients when administered preoperatively. Developing selective COX-2 inhibitors to tackle both cancer and chronic inflammatory illnesses has been identified as a promising research direction in recent decades. Identifying innovative scaffolds to integrate as the major component of future COX-2 inhibitors is critical in this regard. The presence of a central, α, β-unsaturated carbonyl- containing scaffold, as a characteristic structural pattern in many selective COX-2 inhibitors, along with a huge count of chalcone-based anticancer agents representing the basic idea of this review; providing a survey of the most recently published literature concerning development of chalcone analogs as novel COX-2 inhibitors until 2022 with efficient anticancer activity. A brief overview of the most recent developments concerning structure- activity relationship insights and mechanisms is also reported, helping pave the road for additional investigation.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":"77-95"},"PeriodicalIF":2.8,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"92152292","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Anticancer Activity of Sargassum fluitans Extracts in Different Cancer Cells. 马尾藻提取物对不同癌细胞的抗癌活性
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-01-01 DOI: 10.2174/0118715206282983240215050314
José Arnold González-Garrido, Javier Alejandro Gómez-García, Oswaldo Ignacio Hernández-Abreu, Ivonne María Olivares-Corichi, Fernando Pereyra-Vergara, José Rubén García-Sánchez

Background: The arrival of large quantities of Sargassum in the Mexican Caribbean Sea has generated major environmental, health and economic problems. Although Sargassum has been used in the generation of some commercial products, few studies have described its possible applications as a source of compounds with anticancer activity.

Objective: This study aimed to evaluate the antiproliferative effects of different Sargassum extracts on various cancer cell lines. Furthermore, LC/QTOF-MS was used to identify the compounds related to the antiproliferative effect.

Methods: First, determination of the seaweed was performed, and dichloromethane, chloroform and methanol extracts were obtained. The extracts were evaluated for their antiproliferative effects by MTT in breast (MDAMB- 231 and MCF-7), prostate (DU-145), lung (A549) and cervical (SiHa) cancer cell lines. Finally, LC/QTOFMS identified the compounds related to the antiproliferative effect.

Results: The authentication showed Sargassum fluitans as the predominant species. The extracts of dichloromethane and chloroform showed an antiproliferative effect. Interestingly, the fractionation of the chloroform extract showed two fractions (FC1 and FC2) with antiproliferative activity in MDA-MB-231, SiHa and A549 cancer cell lines. On the other hand, three fractions of dichloromethane extract (FD1, FD4 and FD5) also showed antiproliferative effects in the MDA-MB-231, MCF-7, SiHa and DU-145 cancer cell lines. Furthermore, LC/QTOF-MS revealed the presence of eight major compounds in FC2. Three compounds with evidence of anticancer activity were identified (D-linalool-3-glucoside, (3R,4S,6E,10Z)-3,4,7,11-tetramethyl-6,10-tridecadienal and alpha-tocotrienol).

Conclusion: These findings showed that Sargassum fluitans extracts are a possible source of therapeutic agents against cancer and could act as scaffolds for new drug discovery.

背景:大量马尾藻进入墨西哥加勒比海,引发了重大的环境、健康和经济问题。虽然马尾藻已被用于生产一些商业产品,但很少有研究描述其作为具有抗癌活性的化合物来源的可能应用:本研究旨在评估不同马尾藻提取物对各种癌细胞株的抗增殖作用。此外,还采用 LC/QTOF-MS 方法鉴定与抗增殖作用相关的化合物:方法:首先对海藻进行测定,得到二氯甲烷、氯仿和甲醇提取物。用 MTT 法评估提取物对乳腺癌(MDAMB- 231 和 MCF-7)、前列腺癌(DU-145)、肺癌(A549)和宫颈癌(SiHa)细胞株的抗增殖作用。最后,LC/QTOFMS鉴定了与抗增殖作用相关的化合物:鉴定结果表明,马尾藻是主要的物种。二氯甲烷和氯仿提取物具有抗增殖作用。有趣的是,氯仿提取物的分馏结果显示,两个馏分(FC1 和 FC2)对 MDA-MB-231、SiHa 和 A549 癌细胞株具有抗增殖活性。另一方面,二氯甲烷提取物的三个馏分(FD1、FD4 和 FD5)也在 MDA-MB-231、MCF-7、SiHa 和 DU-145 癌细胞系中显示出抗增殖作用。此外,LC/QTOF-MS 发现 FC2 中含有八种主要化合物。结论:这些研究结果表明,马尾藻具有抗癌活性:这些研究结果表明,马尾藻萃取物是一种可能的癌症治疗药物来源,可作为新药研发的支架。
{"title":"Anticancer Activity of <i>Sargassum fluitans</i> Extracts in Different Cancer Cells.","authors":"José Arnold González-Garrido, Javier Alejandro Gómez-García, Oswaldo Ignacio Hernández-Abreu, Ivonne María Olivares-Corichi, Fernando Pereyra-Vergara, José Rubén García-Sánchez","doi":"10.2174/0118715206282983240215050314","DOIUrl":"10.2174/0118715206282983240215050314","url":null,"abstract":"<p><strong>Background: </strong>The arrival of large quantities of <i>Sargassum</i> in the Mexican Caribbean Sea has generated major environmental, health and economic problems. Although <i>Sargassum</i> has been used in the generation of some commercial products, few studies have described its possible applications as a source of compounds with anticancer activity.</p><p><strong>Objective: </strong>This study aimed to evaluate the antiproliferative effects of different <i>Sargassum</i> extracts on various cancer cell lines. Furthermore, LC/QTOF-MS was used to identify the compounds related to the antiproliferative effect.</p><p><strong>Methods: </strong>First, determination of the seaweed was performed, and dichloromethane, chloroform and methanol extracts were obtained. The extracts were evaluated for their antiproliferative effects by MTT in breast (MDAMB- 231 and MCF-7), prostate (DU-145), lung (A549) and cervical (SiHa) cancer cell lines. Finally, LC/QTOFMS identified the compounds related to the antiproliferative effect.</p><p><strong>Results: </strong>The authentication showed <i>Sargassum fluitans</i> as the predominant species. The extracts of dichloromethane and chloroform showed an antiproliferative effect. Interestingly, the fractionation of the chloroform extract showed two fractions (FC1 and FC2) with antiproliferative activity in MDA-MB-231, SiHa and A549 cancer cell lines. On the other hand, three fractions of dichloromethane extract (FD1, FD4 and FD5) also showed antiproliferative effects in the MDA-MB-231, MCF-7, SiHa and DU-145 cancer cell lines. Furthermore, LC/QTOF-MS revealed the presence of eight major compounds in FC2. Three compounds with evidence of anticancer activity were identified (D-linalool-3-glucoside, (3R,4S,6E,10Z)-3,4,7,11-tetramethyl-6,10-tridecadienal and alpha-tocotrienol).</p><p><strong>Conclusion: </strong>These findings showed that <i>Sargassum fluitans</i> extracts are a possible source of therapeutic agents against cancer and could act as scaffolds for new drug discovery.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":"745-754"},"PeriodicalIF":2.6,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139929654","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
Phytol and α-Bisabolol Synergy Induces Autophagy and Apoptosis in A549 Cells and Additional Molecular Insights through Comprehensive Proteome Analysis via Nano LC-MS/MS. 植物醇和α-双羟基苯酚协同作用诱导 A549 细胞自噬和凋亡以及通过纳米 LC-MS/MS 进行蛋白质组综合分析获得的其他分子见解
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-01-01 DOI: 10.2174/0118715206289038240214102951
Chandramohan Kiruthiga, Kambati Niharika, Kasi Pandima Devi
<p><strong>Background: </strong>Non-Small Cell Lung Cancer (NSCLC) is a malignancy with a significant prevalence and aggressive nature, posing a considerable challenge in terms of therapeutic interventions. Autophagy and apoptosis, two intricate cellular processes, are integral to NSCLC pathophysiology, each affecting the other through shared signaling pathways. Phytol (Phy) and α-bisabolol (Bis) have shown promise as potential anticancer agents individually, but their combined effects in NSCLC have not been extensively investigated.</p><p><strong>Objective: </strong>The present study was to examine the synergistic impact of Phy and Bis on NSCLC cells, particularly in the context of autophagy modulation, and to elucidate the resulting differential protein expression using LCMS/ MS analysis.</p><p><strong>Methods: </strong>The A549 cell lines were subjected to the patented effective concentration of Phy and Bis, and subsequently, the viability of the cells was evaluated utilizing the MTT assay. The present study utilized real-time PCR analysis to assess the expression levels of crucial apoptotic genes, specifically Bcl-2, Bax, and Caspase-9, as well as autophagy-related genes, including Beclin-1, SQSTM1, Ulk1, and LC3B. The confirmation of autophagy marker expression (Beclin-1, LC3B) and the autophagy-regulating protein SQSTM1 was achieved through the utilization of Western blot analysis. Differentially expressed proteins were found using LC-MS/MS analysis.</p><p><strong>Results: </strong>The combination of Phy and Bis demonstrated significant inhibition of NSCLC cell growth, indicating their synergistic effect. Real-time PCR analysis revealed a shift towards apoptosis, with downregulation of Bcl-2 and upregulation of Bax and Caspase-9, suggesting a shift towards apoptosis. Genes associated with autophagy regulation, including Beclin-1, SQSTM1 (p62), Ulk1, and LC3B, showed significant upregulation, indicating potential induction of autophagy. Western blot analysis confirmed increased expression of autophagy markers, such as Beclin-1 and LC3B, while the autophagy-regulating protein SQSTM1 exhibited a significant decrease. LC-MS/MS analysis revealed differential expression of 861 proteins, reflecting the modulation of cellular processes. Protein-protein interaction network analysis highlighted key proteins involved in apoptotic and autophagic pathways, including STOML2, YWHAB, POX2, B2M, CDA, CAPN2, TXN, ECHS1, PEBP1, PFN1, CDC42, TUBB1, HSPB1, PXN, FGF2, and BAG3, emphasizing their crucial roles. Additionally, PANTHER pathway analysis uncovered enriched pathways associated with the differentially expressed proteins, revealing their involvement in a diverse range of biological processes, encompassing cell signaling, metabolism, and cellular stress responses.</p><p><strong>Conclusion: </strong>The combined treatment of Phy and Bis exerts a synergistic inhibitory effect on NSCLC cell growth, mediated through the interplay of apoptosis and autophagy. T
背景:非小细胞肺癌(NSCLC非小细胞肺癌(NSCLC)是一种发病率高、侵袭性强的恶性肿瘤,给治疗干预带来了巨大挑战。自噬和细胞凋亡是两个错综复杂的细胞过程,是 NSCLC 病理生理学不可或缺的组成部分,两者通过共享的信号通路相互影响。植物醇(Phy)和α -双羟基苯酚(Bis)已显示出作为潜在抗癌剂的前景,但它们在NSCLC中的联合作用尚未得到广泛研究:本研究旨在考察 Phy 和 Bis 对 NSCLC 细胞的协同作用,尤其是在自噬调节方面,并利用 LCMS/ MS 分析法阐明由此产生的不同蛋白质表达:方法:将 A549 细胞系置于专利有效浓度的 Phy 和 Bis 中,然后利用 MTT 检测法评估细胞的存活率。本研究利用实时 PCR 分析评估了关键凋亡基因(特别是 Bcl-2、Bax 和 Caspase-9)以及自噬相关基因(包括 Beclin-1、SQSTM1、Ulk1 和 LC3B)的表达水平。自噬标志物(Beclin-1、LC3B)和自噬调节蛋白 SQSTM1 的表达是通过 Western 印迹分析确认的。通过 LC-MS/MS 分析发现了不同表达的蛋白质:结果:Phy 和 Bis 的组合能显著抑制 NSCLC 细胞的生长,表明它们具有协同作用。实时 PCR 分析显示,Bcl-2 下调,Bax 和 Caspase-9 上调,表明细胞向凋亡方向转变。与自噬调控相关的基因,包括 Beclin-1、SQSTM1 (p62)、Ulk1 和 LC3B,都出现了显著的上调,表明可能会诱导自噬。Western 印迹分析证实 Beclin-1 和 LC3B 等自噬标记物的表达增加,而自噬调节蛋白 SQSTM1 的表达则明显减少。LC-MS/MS 分析显示了 861 种蛋白质的不同表达,反映了细胞过程的调节。蛋白-蛋白相互作用网络分析突出了参与凋亡和自噬通路的关键蛋白,包括 STOML2、YWHAB、POX2、B2M、CDA、CAPN2、TXN、ECHS1、PEBP1、PFN1、CDC42、TUBB1、HSPB1、PXN、FGF2 和 BAG3,强调了它们的关键作用。此外,PANTHER通路分析发现了与差异表达蛋白相关的富集通路,揭示了它们参与了多种生物过程,包括细胞信号传导、新陈代谢和细胞应激反应:结论:Phy和Bis联合治疗可通过细胞凋亡和自噬的相互作用对NSCLC细胞的生长产生协同抑制作用。观察到的蛋白质表达差异以及鉴定出的蛋白质和丰富的通路为了解潜在的分子机制提供了宝贵的信息。这些发现为进一步探索 Phy 和 Bis 在治疗 NSCLC 方面的治疗潜力奠定了基础。
{"title":"Phytol and α-Bisabolol Synergy Induces Autophagy and Apoptosis in A549 Cells and Additional Molecular Insights through Comprehensive Proteome Analysis <i>via</i> Nano LC-MS/MS.","authors":"Chandramohan Kiruthiga, Kambati Niharika, Kasi Pandima Devi","doi":"10.2174/0118715206289038240214102951","DOIUrl":"10.2174/0118715206289038240214102951","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Non-Small Cell Lung Cancer (NSCLC) is a malignancy with a significant prevalence and aggressive nature, posing a considerable challenge in terms of therapeutic interventions. Autophagy and apoptosis, two intricate cellular processes, are integral to NSCLC pathophysiology, each affecting the other through shared signaling pathways. Phytol (Phy) and α-bisabolol (Bis) have shown promise as potential anticancer agents individually, but their combined effects in NSCLC have not been extensively investigated.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Objective: &lt;/strong&gt;The present study was to examine the synergistic impact of Phy and Bis on NSCLC cells, particularly in the context of autophagy modulation, and to elucidate the resulting differential protein expression using LCMS/ MS analysis.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;The A549 cell lines were subjected to the patented effective concentration of Phy and Bis, and subsequently, the viability of the cells was evaluated utilizing the MTT assay. The present study utilized real-time PCR analysis to assess the expression levels of crucial apoptotic genes, specifically Bcl-2, Bax, and Caspase-9, as well as autophagy-related genes, including Beclin-1, SQSTM1, Ulk1, and LC3B. The confirmation of autophagy marker expression (Beclin-1, LC3B) and the autophagy-regulating protein SQSTM1 was achieved through the utilization of Western blot analysis. Differentially expressed proteins were found using LC-MS/MS analysis.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;The combination of Phy and Bis demonstrated significant inhibition of NSCLC cell growth, indicating their synergistic effect. Real-time PCR analysis revealed a shift towards apoptosis, with downregulation of Bcl-2 and upregulation of Bax and Caspase-9, suggesting a shift towards apoptosis. Genes associated with autophagy regulation, including Beclin-1, SQSTM1 (p62), Ulk1, and LC3B, showed significant upregulation, indicating potential induction of autophagy. Western blot analysis confirmed increased expression of autophagy markers, such as Beclin-1 and LC3B, while the autophagy-regulating protein SQSTM1 exhibited a significant decrease. LC-MS/MS analysis revealed differential expression of 861 proteins, reflecting the modulation of cellular processes. Protein-protein interaction network analysis highlighted key proteins involved in apoptotic and autophagic pathways, including STOML2, YWHAB, POX2, B2M, CDA, CAPN2, TXN, ECHS1, PEBP1, PFN1, CDC42, TUBB1, HSPB1, PXN, FGF2, and BAG3, emphasizing their crucial roles. Additionally, PANTHER pathway analysis uncovered enriched pathways associated with the differentially expressed proteins, revealing their involvement in a diverse range of biological processes, encompassing cell signaling, metabolism, and cellular stress responses.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusion: &lt;/strong&gt;The combined treatment of Phy and Bis exerts a synergistic inhibitory effect on NSCLC cell growth, mediated through the interplay of apoptosis and autophagy. T","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":"773-788"},"PeriodicalIF":2.6,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139982201","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
An Insight into the Effect of Schiff Base and their d and f Block Metal Complexes on Various Cancer Cell Lines as Anticancer Agents: A Review. 洞察希夫碱及其 d 和 f 嵌段金属配合物作为抗癌剂对各种癌症细胞系的影响:综述。
IF 2.8 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2024-01-01 DOI: 10.2174/0118715206280314231201111358
Presenjit, Shubhra Chaturvedi, Akanksha Singh, Divya Gautam, Kaman Singh, Anil Kumar Mishra

Over the last few decades, an alarming rise in the percentage of individuals with cancer and those with multi-resistant illnesses has forced researchers to explore possibilities for novel therapeutic approaches. Numerous medications currently exist to treat various disorders, and the development of small molecules as anticancer agents has considerable potential. However, the widespread prevalence of resistance to multiple drugs in cancer indicates that it is necessary to discover novel and promising compounds with ideal characteristics that could overcome the multidrug resistance issue. The utilisation of metallo-drugs has served as a productive anticancer chemotherapeutic method, and this approach may be implemented for combating multi-resistant tumours more successfully. Schiff bases have been receiving a lot of attention as a group of compounds due to their adaptable metal chelating abilities, innate biologic properties, and versatility to tweak the structure to optimise it for a specific biological purpose. The biological relevance of Schiff base and related complexes, notably their anticancer effects, has increased in their popularity as bio-inorganic chemistry has progressed. As a result of learning about Schiff bases antitumor efficacy against multiple cancer cell lines and their complexes, researchers are motivated to develop novel, side-effect-free anticancer treatments. According to study reports from the past ten years, we are still seeking a powerful anticancer contender. This study highlights the potential of Schiff bases, a broad class of chemical molecules, as potent anticancer agents. In combination with other anticancer strategies, they enhance the efficacy of treatment by elevating the cytotoxicity of chemotherapy, surmounting drug resistance, and promoting targeted therapy. Schiff bases also cause cancer cell DNA repair, improve immunotherapy, prevent angiogenesis, cause apoptosis, and lessen the side effects of chemotherapy. The present review explores the development of potential Schiff base and their d and f block metal complexes as anticancer agents against various cancer cell lines.

在过去几十年里,癌症患者和多重耐药性患者的比例出现了惊人的增长,这迫使研究人员探索新型治疗方法的可能性。目前有许多治疗各种疾病的药物,开发小分子抗癌剂也具有相当大的潜力。然而,癌症患者普遍对多种药物产生抗药性,这表明有必要发现具有理想特性的新型化合物,以克服多种药物的抗药性问题。利用金属药物是一种有效的抗癌化疗方法,这种方法可以更成功地对抗多重耐药性肿瘤。希夫碱作为一类化合物受到了广泛的关注,这是因为它们具有适应性强的金属螯合能力、与生俱来的生物特性,以及为特定生物目的而调整结构以达到最佳效果的多功能性。随着生物无机化学的发展,希夫碱和相关配合物的生物相关性,特别是其抗癌作用,越来越受到人们的青睐。由于了解到希夫碱及其复合物对多种癌细胞株的抗肿瘤功效,研究人员有动力开发新型、无副作用的抗癌疗法。根据过去十年的研究报告,我们仍在寻找强有力的抗癌竞争者。这项研究强调了希夫碱这一大类化学分子作为强效抗癌剂的潜力。与其他抗癌策略相结合,它们可以提高化疗的细胞毒性,克服耐药性,促进靶向治疗,从而增强治疗效果。希夫碱还能促进癌细胞 DNA 修复,改善免疫疗法,防止血管生成,导致细胞凋亡,减轻化疗的副作用。本综述探讨了开发潜在的希夫碱及其 d 和 f 嵌段金属配合物作为抗癌剂来对付各种癌细胞系的问题。
{"title":"An Insight into the Effect of Schiff Base and their d and f Block Metal Complexes on Various Cancer Cell Lines as Anticancer Agents: A Review.","authors":"Presenjit, Shubhra Chaturvedi, Akanksha Singh, Divya Gautam, Kaman Singh, Anil Kumar Mishra","doi":"10.2174/0118715206280314231201111358","DOIUrl":"10.2174/0118715206280314231201111358","url":null,"abstract":"<p><p>Over the last few decades, an alarming rise in the percentage of individuals with cancer and those with multi-resistant illnesses has forced researchers to explore possibilities for novel therapeutic approaches. Numerous medications currently exist to treat various disorders, and the development of small molecules as anticancer agents has considerable potential. However, the widespread prevalence of resistance to multiple drugs in cancer indicates that it is necessary to discover novel and promising compounds with ideal characteristics that could overcome the multidrug resistance issue. The utilisation of metallo-drugs has served as a productive anticancer chemotherapeutic method, and this approach may be implemented for combating multi-resistant tumours more successfully. Schiff bases have been receiving a lot of attention as a group of compounds due to their adaptable metal chelating abilities, innate biologic properties, and versatility to tweak the structure to optimise it for a specific biological purpose. The biological relevance of Schiff base and related complexes, notably their anticancer effects, has increased in their popularity as bio-inorganic chemistry has progressed. As a result of learning about Schiff bases antitumor efficacy against multiple cancer cell lines and their complexes, researchers are motivated to develop novel, side-effect-free anticancer treatments. According to study reports from the past ten years, we are still seeking a powerful anticancer contender. This study highlights the potential of Schiff bases, a broad class of chemical molecules, as potent anticancer agents. In combination with other anticancer strategies, they enhance the efficacy of treatment by elevating the cytotoxicity of chemotherapy, surmounting drug resistance, and promoting targeted therapy. Schiff bases also cause cancer cell DNA repair, improve immunotherapy, prevent angiogenesis, cause apoptosis, and lessen the side effects of chemotherapy. The present review explores the development of potential Schiff base and their d and f block metal complexes as anticancer agents against various cancer cell lines.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":"488-503"},"PeriodicalIF":2.8,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139566177","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