首页 > 最新文献

Anti-cancer agents in medicinal chemistry最新文献

英文 中文
Effects of Citrus-derived Diosmetin on Melanoma: Induction of Apoptosis and Autophagy Mediated by PI3K/Akt/mTOR Pathway Inhibition.
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-01-21 DOI: 10.2174/0118715206360266250115065234
Jie Li, Mingyuan Xu, Nanhui Wu, Fei Wu, Jiashe Chen, Xiaoxiang Xu, Fei Tan, Yeqiang Liu

Background: Diosmetin (DIOS) is a naturally abundant flavonoid and possesses various biological activities that hold promise as an anti-cancer agent. However, the anti-cancer activities and underlying mechanism of DIOS on cutaneous melanoma remain unclear.

Objective: This study seeks to explore the anti-tumor effect and mechanism of DIOS in cutaneous melanoma.

Methods: Here, a variety of in vitro and in vivo experiments, combined with RNA sequencing (RNA-seq), were employed to ascertain the potential anti-cutaneous melanoma capacity and mechanism of DIOS.

Results: The results demonstrated that DIOS considerably impeded cell proliferation and triggered cell apoptosis in a dose- and time-dependent manner. Concurrently, DIOS markedly elevated the expression of pro-apoptotic proteins (Cleaved caspase-3, Bax, Cleaved PARP, and Cleaved caspase-9) and downregulated the expression of Bcl-2. Additionally, DIOS markedly upregulated the protein expressions of LC3B-II and Atg5, while downregulating p62 protein expression. Notably, pre-treatment with an autophagy inhibitor significantly inhibited DIOSinduced cell apoptosis and autophagy. Mechanistically, DIOS was identified to repress the PI3K/Akt/mTOR signaling pathway by western blot analyses and RNA-seq. Finally, in vivo experiments using a syngeneic mouse model confirmed the anti-tumor effect of DIOS, which exhibited high levels of apoptosis and autophagy.

Conclusion: These findings propose that DIOS acts as a potential melanoma therapy that exerts its anti-tumor effects by triggering apoptosis and autophagy via inhibition of the PI3K/Akt/mTOR pathway.

{"title":"Effects of Citrus-derived Diosmetin on Melanoma: Induction of Apoptosis and Autophagy Mediated by PI3K/Akt/mTOR Pathway Inhibition.","authors":"Jie Li, Mingyuan Xu, Nanhui Wu, Fei Wu, Jiashe Chen, Xiaoxiang Xu, Fei Tan, Yeqiang Liu","doi":"10.2174/0118715206360266250115065234","DOIUrl":"https://doi.org/10.2174/0118715206360266250115065234","url":null,"abstract":"<p><strong>Background: </strong>Diosmetin (DIOS) is a naturally abundant flavonoid and possesses various biological activities that hold promise as an anti-cancer agent. However, the anti-cancer activities and underlying mechanism of DIOS on cutaneous melanoma remain unclear.</p><p><strong>Objective: </strong>This study seeks to explore the anti-tumor effect and mechanism of DIOS in cutaneous melanoma.</p><p><strong>Methods: </strong>Here, a variety of in vitro and in vivo experiments, combined with RNA sequencing (RNA-seq), were employed to ascertain the potential anti-cutaneous melanoma capacity and mechanism of DIOS.</p><p><strong>Results: </strong>The results demonstrated that DIOS considerably impeded cell proliferation and triggered cell apoptosis in a dose- and time-dependent manner. Concurrently, DIOS markedly elevated the expression of pro-apoptotic proteins (Cleaved caspase-3, Bax, Cleaved PARP, and Cleaved caspase-9) and downregulated the expression of Bcl-2. Additionally, DIOS markedly upregulated the protein expressions of LC3B-II and Atg5, while downregulating p62 protein expression. Notably, pre-treatment with an autophagy inhibitor significantly inhibited DIOSinduced cell apoptosis and autophagy. Mechanistically, DIOS was identified to repress the PI3K/Akt/mTOR signaling pathway by western blot analyses and RNA-seq. Finally, in vivo experiments using a syngeneic mouse model confirmed the anti-tumor effect of DIOS, which exhibited high levels of apoptosis and autophagy.</p><p><strong>Conclusion: </strong>These findings propose that DIOS acts as a potential melanoma therapy that exerts its anti-tumor effects by triggering apoptosis and autophagy via inhibition of the PI3K/Akt/mTOR pathway.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143021816","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
Optimized Rutin-incorporating PEGylated Nanoliposomes as a Model with Remarkable Selectivity Against PANC1 and MCF7 Cell Lines
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-01-21 DOI: 10.2174/0118715206231749241209073759
Ali Al-Samydai, Moath Al Qaraleh, Lidia K Al-Halaseh, Maha N Abu Hajleh, Simone Carradori, Maryam Abdulmaged, Rand Kareem, Hasanain Alzaidi, Mohamad Ak Mousa, Yusuf Al-Hiari, Hamdi Nsairat, Walhan Alshaer

Background: This study aims to enhance the delivery of polyphenols using nanotechnology.

Objective: To develop and evaluate liposomal formulations for improved delivery and stability of polyphenols, specifically focusing on Rutin.

Methods: Liposomal formulations were meticulously prepared via the Thin-Film Hydration method. Comprehensive physical characterization was conducted, including stability assessments using Dynamic Light Scattering (DLS) and Thermogravimetric Analysis (TGA). The free radical scavenging activity was measured using the DPPH• assay, and MTT cell viability assays were performed to assess cytotoxicity.

Results: The results demonstrated a significant reduction in nanoparticle size from 123 nm to 116 nm and an increase in charge from -14 to -22 with rising Rutin concentrations. The formulation achieved enhanced homogeneity at a Rutin concentration of 2.0 mg/mL and showed higher stability. Incorporating Rutin improved the formulation's stability over 30 days, as evidenced by a decrease in the Differential Scanning Calorimetry peak temperature from 58.65 °C to 54.42 °C. Rutin-loaded and co-loaded nanoliposomes exhibited remarkable selectivity against PANK1 and MCF7 cell lines, with IC50 values of 2.13±0.35 μg/mL and 4.75±0.19 μg/mL, respectively.

Conclusion: PEGylated Rutin-loaded nanoliposomes offer a promising platform for biodegradable and biocompatible drug delivery systems, enhancing the bioavailability, solubility, and stability of the polyphenols.

{"title":"Optimized Rutin-incorporating PEGylated Nanoliposomes as a Model with Remarkable Selectivity Against PANC1 and MCF7 Cell Lines","authors":"Ali Al-Samydai, Moath Al Qaraleh, Lidia K Al-Halaseh, Maha N Abu Hajleh, Simone Carradori, Maryam Abdulmaged, Rand Kareem, Hasanain Alzaidi, Mohamad Ak Mousa, Yusuf Al-Hiari, Hamdi Nsairat, Walhan Alshaer","doi":"10.2174/0118715206231749241209073759","DOIUrl":"10.2174/0118715206231749241209073759","url":null,"abstract":"<p><strong>Background: </strong>This study aims to enhance the delivery of polyphenols using nanotechnology.</p><p><strong>Objective: </strong>To develop and evaluate liposomal formulations for improved delivery and stability of polyphenols, specifically focusing on Rutin.</p><p><strong>Methods: </strong>Liposomal formulations were meticulously prepared via the Thin-Film Hydration method. Comprehensive physical characterization was conducted, including stability assessments using Dynamic Light Scattering (DLS) and Thermogravimetric Analysis (TGA). The free radical scavenging activity was measured using the DPPH• assay, and MTT cell viability assays were performed to assess cytotoxicity.</p><p><strong>Results: </strong>The results demonstrated a significant reduction in nanoparticle size from 123 nm to 116 nm and an increase in charge from -14 to -22 with rising Rutin concentrations. The formulation achieved enhanced homogeneity at a Rutin concentration of 2.0 mg/mL and showed higher stability. Incorporating Rutin improved the formulation's stability over 30 days, as evidenced by a decrease in the Differential Scanning Calorimetry peak temperature from 58.65 °C to 54.42 °C. Rutin-loaded and co-loaded nanoliposomes exhibited remarkable selectivity against PANK1 and MCF7 cell lines, with IC50 values of 2.13±0.35 μg/mL and 4.75±0.19 μg/mL, respectively.</p><p><strong>Conclusion: </strong>PEGylated Rutin-loaded nanoliposomes offer a promising platform for biodegradable and biocompatible drug delivery systems, enhancing the bioavailability, solubility, and stability of the polyphenols.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143021820","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 Properties of Phenylboronic Acid in Androgen-Dependent (LNCaP) and Androgen-Independent (PC3) Prostate Cancer Cells via MAP Kinases by 2D and 3D Culture Methods. 苯基硼酸通过MAP激酶在雄激素依赖性(LNCaP)和雄激素非依赖性(PC3)前列腺癌细胞中的抗癌作用
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-01-20 DOI: 10.2174/0118715206352302241227031015
Duygu Gürsoy Gürgen, Arzu Güneş, Oğuzhan Köse, Arife Ahsen Kaplan, Seda Karabulut, M Başak Tunalı, İlknur Keskin

Objective: This study utilized three cell lines: normal prostate epithelial RWPE-1, androgen-dependent LNCaP, and androgen-independent PC3. We investigated the inhibitory effects of phenylboronic acid (PBA)'s inhibitory effect on cellular proliferation due to its ability to disrupt microtubule formation in prostate cancer cell lines. Additionally, this study aimed to assess the cytotoxic effects of PBA on prostate cancer cells using twodimensional (2D) and three-dimensional (3D) cell culture models.

Methods: The IC50 values of PBA and colchicine were determined through viability assays in 2D and 3D models. Colony formation, proliferation, and migration assays were conducted. Immunofluorescence intensity analysis of MAPKKK proteins (ERK, JNK, p38) was performed to explore the mechanism of cellular response to PBA.

Results: The IC50 values were determined for each treatment group. After 48-hour of PBA treatment, migration was inhibited more effectively than with colchicine in both cancer cell lines. After 24-hour, PBA reduced colony formation and proliferation. PBA treatment for 24-hour decreased JNK expression in PC3 and LNCaP cells in 2D models. Both PBA and colchicine increased p38 expression in PC3 spheroids. PBA's effects on cell deformation were visualized in semi-thin sections, marking the first ultrastructural observation of PBA-induced morphological defects in cancer cells.

Conclusion: PBA exerts antimitotic effects by inhibiting proliferation and migration and triggers diverse metabolic responses across different cell lines. Furthermore the low toxicity of PBA's low toxicity on RWPE-1 cells suggests its potential as a promising chemotherapeutic agent for future studies.

目的:本研究利用三种细胞系:正常前列腺上皮细胞RWPE-1、雄激素依赖性LNCaP和雄激素非依赖性PC3。我们研究了苯硼酸(PBA)对前列腺癌细胞增殖的抑制作用,这是由于它能够破坏微管的形成。此外,本研究旨在通过二维(2D)和三维(3D)细胞培养模型评估PBA对前列腺癌细胞的细胞毒性作用。方法:采用2D和3D模型,采用活力测定法测定PBA和秋水仙碱的IC50值。进行了菌落形成、增殖和迁移试验。对MAPKKK蛋白(ERK、JNK、p38)进行免疫荧光强度分析,探讨细胞对PBA反应的机制。结果:测定各治疗组的IC50值。PBA处理48小时后,两种癌细胞的迁移都比秋水仙碱更有效地受到抑制。24小时后,PBA减少菌落形成和增殖。在二维模型中,PBA处理24小时可降低PC3和LNCaP细胞中JNK的表达。PBA和秋水仙碱均可增加PC3球体中p38的表达。PBA对细胞变形的影响在半薄切片上可见,标志着PBA诱导的癌细胞形态缺陷的首次超微结构观察。结论:PBA通过抑制细胞增殖和迁移发挥抗有丝分裂作用,并在不同细胞系中引发不同的代谢反应。此外,PBA对RWPE-1细胞的低毒性表明其作为一种有前景的化疗药物在未来的研究中具有潜力。
{"title":"Anticancer Properties of Phenylboronic Acid in Androgen-Dependent (LNCaP) and Androgen-Independent (PC3) Prostate Cancer Cells via MAP Kinases by 2D and 3D Culture Methods.","authors":"Duygu Gürsoy Gürgen, Arzu Güneş, Oğuzhan Köse, Arife Ahsen Kaplan, Seda Karabulut, M Başak Tunalı, İlknur Keskin","doi":"10.2174/0118715206352302241227031015","DOIUrl":"https://doi.org/10.2174/0118715206352302241227031015","url":null,"abstract":"<p><strong>Objective: </strong>This study utilized three cell lines: normal prostate epithelial RWPE-1, androgen-dependent LNCaP, and androgen-independent PC3. We investigated the inhibitory effects of phenylboronic acid (PBA)'s inhibitory effect on cellular proliferation due to its ability to disrupt microtubule formation in prostate cancer cell lines. Additionally, this study aimed to assess the cytotoxic effects of PBA on prostate cancer cells using twodimensional (2D) and three-dimensional (3D) cell culture models.</p><p><strong>Methods: </strong>The IC50 values of PBA and colchicine were determined through viability assays in 2D and 3D models. Colony formation, proliferation, and migration assays were conducted. Immunofluorescence intensity analysis of MAPKKK proteins (ERK, JNK, p38) was performed to explore the mechanism of cellular response to PBA.</p><p><strong>Results: </strong>The IC50 values were determined for each treatment group. After 48-hour of PBA treatment, migration was inhibited more effectively than with colchicine in both cancer cell lines. After 24-hour, PBA reduced colony formation and proliferation. PBA treatment for 24-hour decreased JNK expression in PC3 and LNCaP cells in 2D models. Both PBA and colchicine increased p38 expression in PC3 spheroids. PBA's effects on cell deformation were visualized in semi-thin sections, marking the first ultrastructural observation of PBA-induced morphological defects in cancer cells.</p><p><strong>Conclusion: </strong>PBA exerts antimitotic effects by inhibiting proliferation and migration and triggers diverse metabolic responses across different cell lines. Furthermore the low toxicity of PBA's low toxicity on RWPE-1 cells suggests its potential as a promising chemotherapeutic agent for future studies.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142998834","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
Uses of Cyclohexan-1,3-diones to Synthesis Xanthenes Derivatives with Anti-proliferative Activity Against Cancer Cell Lines and their Inhibitions Toward Tyrosine Kinases. 利用环己烷-1,3-二酮合成具有抗癌活性的杂蒽衍生物及其对酪氨酸激酶的抑制作用。
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-01-16 DOI: 10.2174/0118715206350037241206062610
Rafat Milad Mohareb, Nadia Y Megally Abdo, Marwa Shokry Ibrahim

Background: Xanthene derivatives are a notable class of heterocyclic compounds widely studied for their significant biological impact. These molecules, found in both natural and synthetic forms, have attracted substantial scientific interest due to their broad spectrum of biological activities. The xanthene nucleus, in particular, is associated with a range of potential pharmaceutical properties, including antibacterial, antiviral, antiinflammatory, anticancer, and antioxidant effects. Their structural flexibility allows for modifications that can enhance specific biological functions, making them valuable candidates in medicinal chemistry and drug development.

Objective: Multi-component reactions involving two equivalents of 5,5-dimethylcyclohexane-1,3-dione with aromatic aldehydes yield xanthene derivatives that are known for their biological activity. Additionally, fused xanthene derivatives are formed through subsequent heterocyclization reactions, resulting in compounds with a broad range of biological properties.

Methods: Various xanthene derivatives incorporating thiophene and thiazole moieties were synthesized. Compounds 3a-c were further subjected to heterocyclization reactions to produce fused xanthene derivatives with additional heterocyclic components, enhancing their biological activity. The cytotoxic effects of the synthesized compounds were assessed across six cancer cell lines. Inhibition studies on c-Met kinase and the PC-3 cell line were conducted.

Result: Additionally, the compounds' inhibitory activity against tyrosine kinases was evaluated, and morphological changes in the A549 cell line were observed with the two most potent compounds.

Conclusion: The synthesized heterocyclic compounds, derived from 5,5-dimethylcyclohexane-1,3-dione and related cyclohexanone derivatives, exhibited significant inhibitory effects across various cancer cell lines. Specifically, compounds 3b, 5c, 5d, 7b, 7c, 7d, 9a, 9b, 10b, 10c, 12c, 15b, 15c, 16b, 16c, 17c, 17d, 17e, and 17f demonstrated high levels of inhibition, indicating potential for further exploration of xanthene-based heterocyclic compounds to enhance anticancer properties.

背景:杂环衍生物是一类重要的杂环化合物,因其具有重要的生物学作用而被广泛研究。这些分子以自然和合成的形式存在,由于其广泛的生物活性,已经引起了大量的科学兴趣。特别是,杂蒽核具有一系列潜在的药物特性,包括抗菌、抗病毒、抗炎、抗癌和抗氧化作用。它们的结构灵活性允许修改,可以增强特定的生物功能,使它们在药物化学和药物开发中有价值的候选者。目的:涉及两种当量的5,5-二甲基环己烷-1,3-二酮与芳香醛的多组分反应产生以其生物活性而闻名的杂蒽衍生物。此外,通过随后的杂环化反应形成融合的杂蒽衍生物,从而产生具有广泛生物特性的化合物。方法:合成含噻吩和噻唑基团的多种杂蒽衍生物。化合物3a-c进一步进行杂环化反应,生成含有额外杂环成分的融合杂蒽衍生物,提高其生物活性。在六种癌细胞系中评估了合成化合物的细胞毒性作用。对c-Met激酶和PC-3细胞株进行了抑制研究。结果:测定了两种化合物对酪氨酸激酶的抑制活性,并观察了两种最有效化合物对A549细胞株的形态学改变。结论:合成的由5,5-二甲基环己烷-1,3-二酮及其相关环己酮衍生物衍生的杂环化合物对多种癌细胞具有明显的抑制作用。具体而言,化合物3b、5c、5d、7b、7c、7d、9a、9b、10b、10c、12c、15b、15c、16b、16c、17c、17d、17e和17f显示出高水平的抑制作用,表明进一步探索以杂环烷为基础的化合物以增强抗癌特性的潜力。
{"title":"Uses of Cyclohexan-1,3-diones to Synthesis Xanthenes Derivatives with Anti-proliferative Activity Against Cancer Cell Lines and their Inhibitions Toward Tyrosine Kinases.","authors":"Rafat Milad Mohareb, Nadia Y Megally Abdo, Marwa Shokry Ibrahim","doi":"10.2174/0118715206350037241206062610","DOIUrl":"https://doi.org/10.2174/0118715206350037241206062610","url":null,"abstract":"<p><strong>Background: </strong>Xanthene derivatives are a notable class of heterocyclic compounds widely studied for their significant biological impact. These molecules, found in both natural and synthetic forms, have attracted substantial scientific interest due to their broad spectrum of biological activities. The xanthene nucleus, in particular, is associated with a range of potential pharmaceutical properties, including antibacterial, antiviral, antiinflammatory, anticancer, and antioxidant effects. Their structural flexibility allows for modifications that can enhance specific biological functions, making them valuable candidates in medicinal chemistry and drug development.</p><p><strong>Objective: </strong>Multi-component reactions involving two equivalents of 5,5-dimethylcyclohexane-1,3-dione with aromatic aldehydes yield xanthene derivatives that are known for their biological activity. Additionally, fused xanthene derivatives are formed through subsequent heterocyclization reactions, resulting in compounds with a broad range of biological properties.</p><p><strong>Methods: </strong>Various xanthene derivatives incorporating thiophene and thiazole moieties were synthesized. Compounds 3a-c were further subjected to heterocyclization reactions to produce fused xanthene derivatives with additional heterocyclic components, enhancing their biological activity. The cytotoxic effects of the synthesized compounds were assessed across six cancer cell lines. Inhibition studies on c-Met kinase and the PC-3 cell line were conducted.</p><p><strong>Result: </strong>Additionally, the compounds' inhibitory activity against tyrosine kinases was evaluated, and morphological changes in the A549 cell line were observed with the two most potent compounds.</p><p><strong>Conclusion: </strong>The synthesized heterocyclic compounds, derived from 5,5-dimethylcyclohexane-1,3-dione and related cyclohexanone derivatives, exhibited significant inhibitory effects across various cancer cell lines. Specifically, compounds 3b, 5c, 5d, 7b, 7c, 7d, 9a, 9b, 10b, 10c, 12c, 15b, 15c, 16b, 16c, 17c, 17d, 17e, and 17f demonstrated high levels of inhibition, indicating potential for further exploration of xanthene-based heterocyclic compounds to enhance anticancer properties.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142998751","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
Safety and Efficacy of Anlotinib-based Regimen in Patients with Unresectable or Metastatic Bone and Soft-tissue Sarcomas: A Retrospective Institutional Study. 以安洛替尼为基础的方案治疗不可切除或转移性骨和软组织肉瘤的安全性和有效性:一项回顾性机构研究。
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-01-16 DOI: 10.2174/0118715206336884241216070930
Lina Pang, Shengli Zhang, Liye Wang, Shuai Gong, Wei He

Background: Anlotinib has demonstrated durable clinical benefits in patients with unresectable or metastatic bone and soft-tissue sarcomas.

Methods: 92 patients treated with chemotherapy combined with or without anlotinib were collected and analyzed. The objective response rate (ORR) and disease control rate (DCR) were analyzed. Long-term survival was assessed using the Kaplan-Meier method, including median progression-free survival (mPFS) and overall survival (mOS).

Results: Liposarcoma, synovial sarcoma, and rhabdomyosarcoma were the primary pathological subtypes of the 92 patients. The median age was 46 (range, 11-75) years. The ORR and DCR of the anlotinib-chemotherapy combination used as first-line therapy were 31.9% and 85.1%, respectively. However, the ORR and DCR were only 6.7% and 57.8% in the chemotherapy alone, respectively. Compared with the chemotherapy group, improvements were observed in the mPFS and mOS with anlotinib-based regimen (mPFS, 8.3 vs. 3.0 months; mOS, 59.0 vs. 22.0 months). Anlotinib-associated adverse events were well tolerated and mainly occurred in grades I and II. New anlotinib-related adverse reactions were not noted.

Conclusion: Anlotinib-based regimen as a first-line therapy showed a positive effect on the treatment of unresectable or metastatic BSTSs. The anlotinib-associated adverse events were minor and well tolerated.

背景:安洛替尼在不可切除或转移性骨和软组织肉瘤患者中显示出持久的临床疗效。方法:收集和分析92例联合或不联合安洛替尼化疗的患者。分析客观有效率(ORR)和疾病控制率(DCR)。采用Kaplan-Meier法评估长期生存期,包括中位无进展生存期(mPFS)和总生存期(mOS)。结果:92例患者的主要病理亚型为脂肪肉瘤、滑膜肉瘤和横纹肌肉瘤。中位年龄为46岁(范围11-75岁)。安洛替尼联合化疗作为一线治疗的ORR和DCR分别为31.9%和85.1%。而单纯化疗的ORR和DCR分别仅为6.7%和57.8%。与化疗组相比,基于anlotinib方案的mPFS和mOS均有改善(mPFS, 8.3个月vs. 3.0个月;平均寿命59.0个月vs. 22.0个月)。anlotinib相关不良事件耐受性良好,主要发生在I级和II级。未发现新的anlotinib相关不良反应。结论:以安洛替尼为基础的一线治疗方案对不可切除或转移性BSTSs有积极的疗效。anlotinib相关不良事件轻微且耐受性良好。
{"title":"Safety and Efficacy of Anlotinib-based Regimen in Patients with Unresectable or Metastatic Bone and Soft-tissue Sarcomas: A Retrospective Institutional Study.","authors":"Lina Pang, Shengli Zhang, Liye Wang, Shuai Gong, Wei He","doi":"10.2174/0118715206336884241216070930","DOIUrl":"https://doi.org/10.2174/0118715206336884241216070930","url":null,"abstract":"<p><strong>Background: </strong>Anlotinib has demonstrated durable clinical benefits in patients with unresectable or metastatic bone and soft-tissue sarcomas.</p><p><strong>Methods: </strong>92 patients treated with chemotherapy combined with or without anlotinib were collected and analyzed. The objective response rate (ORR) and disease control rate (DCR) were analyzed. Long-term survival was assessed using the Kaplan-Meier method, including median progression-free survival (mPFS) and overall survival (mOS).</p><p><strong>Results: </strong>Liposarcoma, synovial sarcoma, and rhabdomyosarcoma were the primary pathological subtypes of the 92 patients. The median age was 46 (range, 11-75) years. The ORR and DCR of the anlotinib-chemotherapy combination used as first-line therapy were 31.9% and 85.1%, respectively. However, the ORR and DCR were only 6.7% and 57.8% in the chemotherapy alone, respectively. Compared with the chemotherapy group, improvements were observed in the mPFS and mOS with anlotinib-based regimen (mPFS, 8.3 vs. 3.0 months; mOS, 59.0 vs. 22.0 months). Anlotinib-associated adverse events were well tolerated and mainly occurred in grades I and II. New anlotinib-related adverse reactions were not noted.</p><p><strong>Conclusion: </strong>Anlotinib-based regimen as a first-line therapy showed a positive effect on the treatment of unresectable or metastatic BSTSs. The anlotinib-associated adverse events were minor and well tolerated.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142998750","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
Irisquinone's Anti-cancer Potential: Targeting TrxR to Trigger ROS-mediated Apoptosis and Pyroptosis. 鸢尾醌的抗癌潜力:靶向TrxR触发ros介导的细胞凋亡和焦亡。
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-01-15 DOI: 10.2174/0118715206339230241202062826
Qifeng Zhang, Xinyan Wang, Gegen Tana, Guodong Liang, Yuheng Ma, Ren Bu, Lu Ga

Background: Irisquinone, an important compound extracted from Semen Irisis, has been used clinically as a radiotherapy sensitizer for lung, oesophageal, head and neck, breast and leukemia cancers. However, the mechanism by which it acts against cancer is still unclear.

Objective: The present study aims to investigate the anti-tumor activity and mechanism of Irisquinone.

Methods: The effect of Irisquinone on cell viability and proliferation was evaluated using the CCK-8 assay. Fluorescence probe (Fast-TRFS) and DTNB assay were used to observe the inhibitory effect of Irisquinone on both intracellular and extracellular thioredoxin reductase (TrxR). The level of reactive oxygen species (ROS) in tumour cells was assessed using the DCFH-DA probe. Annexin V-FITC/PI, staining and microscopy experiments, were used to examine the apoptosis and pyroptosis. Western blotting analyses confirmed that Irisquinone induced apoptosis and pyroptosis in cancer cells by inhibiting TrxR to increase ROS generation.

Results: Our research has shown that Irisquinone has anti-proliferative effects on several cancer cell lines while having low toxicity to normal cells. The amount of ROS induced by inhibition of TrxR activated the BAX (proapoptotic protein) and caspase-1(the pro-pyroptotic protein) to induce apoptosis and pyroptosis.

Conclusion: Irisquinone showed anticancer activity through inhibiting TrxR. These results suggested that Irisquinone will be developed to be an anti-tumor drug possibility.

背景:鸢尾醌是鸢尾中提取的一种重要化合物,临床上已被用作肺癌、食管癌、头颈癌、乳腺癌和白血病的放疗增敏剂。然而,它对抗癌症的机制仍不清楚。目的:探讨鸢尾醌的抗肿瘤活性及其作用机制。方法:采用CCK-8法观察鸢尾醌对细胞活力和增殖的影响。采用荧光探针(Fast-TRFS)和DTNB法观察鸢尾醌对细胞内和细胞外硫氧还蛋白还原酶(TrxR)的抑制作用。使用DCFH-DA探针评估肿瘤细胞中的活性氧(ROS)水平。采用Annexin V-FITC/PI染色和显微镜观察凋亡和焦亡情况。Western blotting分析证实,鸢尾醌通过抑制TrxR增加ROS生成,诱导癌细胞凋亡和焦亡。结果:我们的研究表明,鸢尾醌对几种癌细胞系具有抗增殖作用,而对正常细胞的毒性较低。抑制TrxR诱导的ROS量激活BAX(促凋亡蛋白)和caspase-1(促焦亡蛋白),诱导细胞凋亡和焦亡。结论:鸢尾醌通过抑制TrxR具有抗癌活性。这些结果提示鸢尾醌将有可能成为一种抗肿瘤药物。
{"title":"Irisquinone's Anti-cancer Potential: Targeting TrxR to Trigger ROS-mediated Apoptosis and Pyroptosis.","authors":"Qifeng Zhang, Xinyan Wang, Gegen Tana, Guodong Liang, Yuheng Ma, Ren Bu, Lu Ga","doi":"10.2174/0118715206339230241202062826","DOIUrl":"https://doi.org/10.2174/0118715206339230241202062826","url":null,"abstract":"<p><strong>Background: </strong>Irisquinone, an important compound extracted from Semen Irisis, has been used clinically as a radiotherapy sensitizer for lung, oesophageal, head and neck, breast and leukemia cancers. However, the mechanism by which it acts against cancer is still unclear.</p><p><strong>Objective: </strong>The present study aims to investigate the anti-tumor activity and mechanism of Irisquinone.</p><p><strong>Methods: </strong>The effect of Irisquinone on cell viability and proliferation was evaluated using the CCK-8 assay. Fluorescence probe (Fast-TRFS) and DTNB assay were used to observe the inhibitory effect of Irisquinone on both intracellular and extracellular thioredoxin reductase (TrxR). The level of reactive oxygen species (ROS) in tumour cells was assessed using the DCFH-DA probe. Annexin V-FITC/PI, staining and microscopy experiments, were used to examine the apoptosis and pyroptosis. Western blotting analyses confirmed that Irisquinone induced apoptosis and pyroptosis in cancer cells by inhibiting TrxR to increase ROS generation.</p><p><strong>Results: </strong>Our research has shown that Irisquinone has anti-proliferative effects on several cancer cell lines while having low toxicity to normal cells. The amount of ROS induced by inhibition of TrxR activated the BAX (proapoptotic protein) and caspase-1(the pro-pyroptotic protein) to induce apoptosis and pyroptosis.</p><p><strong>Conclusion: </strong>Irisquinone showed anticancer activity through inhibiting TrxR. These results suggested that Irisquinone will be developed to be an anti-tumor drug possibility.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142998748","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
KW2478 and Cisplatin Synergistically Anti-colorectal Cancer by Targeting PI3K/AKT/mTOR Pathway. 靶向PI3K/AKT/mTOR通路的KW2478与顺铂协同抗结直肠癌
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-01-14 DOI: 10.2174/0118715206356311241128075924
Jianping Wang, Jun An, Lixuan Tian, Yuzi Jin, Yalei Li, Peijian Ding, Wenjing Yun, Yunpeng Zhang, Shuang Zhao
<p><strong>Objective: </strong>The objective of this study is to examine the impact of KW-2478 combined with DDP on colorectal cancer cells both in vitro and in vivo and to elucidate the molecular mechanism of KW-2478 in colorectal cancer.</p><p><strong>Methods: </strong>qRT-PCR and Western blot were employed to assess HSP90 mRNA and protein expression in normal intestinal epithelial and colorectal cancer cells. DLD-1 and HCT116 were selected for the experiment. CCK-8 was used to detect cytotoxicity; apoptosis rate was measured using flow cytometry; Western blot was employed to measure the expression levels of apoptotic and PI3K/AKT/mTOR pathway proteins. HCT116 was used to construct a subcutaneous tumor model in nude mice. After treatment with KW-2478 and DDP, the growth rate, volume, and weight of the tumor were observed. The expression of Ki67 was detected by immunohistochemistry. Apoptosis of tumor cells was detected using TUNEL. Western blot was employed to measure the expression levels of apoptotic and PI3K/AKT/mTOR pathway proteins.</p><p><strong>Results: </strong>HSP90 mRNA and protein levels were elevated in colorectal cancer cells compared to normal colorectal epithelial cells. HSP90 mRNA and protein expression levels were also significantly elevated in HCT116 and DLD-1 cells compared to other colorectal cancer cells. In DLD-1 and HCT116 cells, KW2478 and DDP inhibited cell viability. The combination of KW2478 and DDP exhibited a significantly higher inhibitory effect compared to either KW2478 or DDP alone. DDP markedly triggered apoptosis in HCT116 and DLD-1. KW2478 at 3 μg/ml and 6 μg/ml induced apoptosis in HCT116 cells but not in DLD-1 cells. The combination of KW2478 and DDP induced a significantly higher apoptosis rate as compared to either KW2478 or DDP alone. Treatment of HCT116 and DLD-1 with KW2478 or DDP alone increased Bax, Caspase9, and Caspase3 protein expression, while decreasing BCL-2. The KW2478+DDP combined treatment group exhibited more significant changes. Phosphorylation of PI3k, AKT, and mTOR decreased in the KW2478 or DDP treatment groups, with more significant changes observed in the KW2478 + DDP combination group. The growth rate, volume, and weight of subcutaneous tumors in the KW2478 or DDP treatment groups were significantly lower than control, and the KW2478+DDP combination group was more affected. Ki67 expression in subcutaneous tumors was reduced in the KW2478 or DDP treatment groups compared to the vehicle control group, with the lowest expression observed in the KW2478 + DDP combination group. The fluorescence intensity of subcutaneous tumors was higher in both the KW2478 and DDP treatment groups compared to the vehicle control group, and the KW2478 + DDP combination group exhibited the strongest fluorescence intensity among them.</p><p><strong>Conclusion: </strong>The combination of KW2478 and cisplatin inhibits colorectal cancer cell proliferation and induces apoptosis by regulating the PI3K/AKT/mTOR p
目的:本研究的目的是在体外和体内研究KW-2478联合DDP对结直肠癌细胞的影响,并阐明KW-2478在结直肠癌中的分子机制。方法:采用qRT-PCR和Western blot检测正常肠上皮细胞和结直肠癌细胞中HSP90 mRNA和蛋白的表达。实验选用DLD-1和HCT116。CCK-8检测细胞毒性;流式细胞术检测细胞凋亡率;Western blot检测凋亡和PI3K/AKT/mTOR通路蛋白的表达水平。采用HCT116构建裸鼠皮下肿瘤模型。经KW-2478和DDP治疗后,观察肿瘤的生长速度、体积和重量。免疫组织化学检测Ki67的表达。TUNEL法检测肿瘤细胞凋亡。Western blot检测凋亡和PI3K/AKT/mTOR通路蛋白的表达水平。结果:与正常结肠上皮细胞相比,结直肠癌细胞中HSP90 mRNA和蛋白水平升高。与其他结直肠癌细胞相比,HCT116和DLD-1细胞中HSP90 mRNA和蛋白表达水平也显著升高。在DLD-1和HCT116细胞中,KW2478和DDP抑制细胞活力。与单独使用KW2478或DDP相比,KW2478与DDP联合使用的抑制效果明显更高。DDP显著刺激HCT116和DLD-1细胞凋亡。3 μg/ml和6 μg/ml的KW2478诱导HCT116细胞凋亡,而对DLD-1细胞无诱导作用。与单独使用KW2478或DDP相比,KW2478和DDP联合使用可显著提高细胞的凋亡率。KW2478或DDP单独处理HCT116和DLD-1后,Bax、Caspase9和Caspase3蛋白表达增加,BCL-2表达降低。KW2478+DDP联合治疗组变化更为显著。在KW2478或DDP治疗组中,PI3k、AKT和mTOR的磷酸化水平下降,其中KW2478 + DDP联合治疗组的变化更为显著。KW2478和DDP治疗组皮下肿瘤的生长速度、体积和重量均显著低于对照组,且KW2478+DDP联合治疗组受影响更大。与对照药组相比,KW2478或DDP治疗组皮下肿瘤中Ki67的表达降低,其中KW2478 + DDP联合治疗组表达最低。KW2478和DDP治疗组皮下肿瘤的荧光强度均高于对照,其中KW2478 + DDP联合治疗组荧光强度最强。结论:KW2478与顺铂联用可通过调节PI3K/AKT/mTOR通路抑制结直肠癌细胞增殖,诱导凋亡。
{"title":"KW2478 and Cisplatin Synergistically Anti-colorectal Cancer by Targeting PI3K/AKT/mTOR Pathway.","authors":"Jianping Wang, Jun An, Lixuan Tian, Yuzi Jin, Yalei Li, Peijian Ding, Wenjing Yun, Yunpeng Zhang, Shuang Zhao","doi":"10.2174/0118715206356311241128075924","DOIUrl":"https://doi.org/10.2174/0118715206356311241128075924","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Objective: &lt;/strong&gt;The objective of this study is to examine the impact of KW-2478 combined with DDP on colorectal cancer cells both in vitro and in vivo and to elucidate the molecular mechanism of KW-2478 in colorectal cancer.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;qRT-PCR and Western blot were employed to assess HSP90 mRNA and protein expression in normal intestinal epithelial and colorectal cancer cells. DLD-1 and HCT116 were selected for the experiment. CCK-8 was used to detect cytotoxicity; apoptosis rate was measured using flow cytometry; Western blot was employed to measure the expression levels of apoptotic and PI3K/AKT/mTOR pathway proteins. HCT116 was used to construct a subcutaneous tumor model in nude mice. After treatment with KW-2478 and DDP, the growth rate, volume, and weight of the tumor were observed. The expression of Ki67 was detected by immunohistochemistry. Apoptosis of tumor cells was detected using TUNEL. Western blot was employed to measure the expression levels of apoptotic and PI3K/AKT/mTOR pathway proteins.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;HSP90 mRNA and protein levels were elevated in colorectal cancer cells compared to normal colorectal epithelial cells. HSP90 mRNA and protein expression levels were also significantly elevated in HCT116 and DLD-1 cells compared to other colorectal cancer cells. In DLD-1 and HCT116 cells, KW2478 and DDP inhibited cell viability. The combination of KW2478 and DDP exhibited a significantly higher inhibitory effect compared to either KW2478 or DDP alone. DDP markedly triggered apoptosis in HCT116 and DLD-1. KW2478 at 3 μg/ml and 6 μg/ml induced apoptosis in HCT116 cells but not in DLD-1 cells. The combination of KW2478 and DDP induced a significantly higher apoptosis rate as compared to either KW2478 or DDP alone. Treatment of HCT116 and DLD-1 with KW2478 or DDP alone increased Bax, Caspase9, and Caspase3 protein expression, while decreasing BCL-2. The KW2478+DDP combined treatment group exhibited more significant changes. Phosphorylation of PI3k, AKT, and mTOR decreased in the KW2478 or DDP treatment groups, with more significant changes observed in the KW2478 + DDP combination group. The growth rate, volume, and weight of subcutaneous tumors in the KW2478 or DDP treatment groups were significantly lower than control, and the KW2478+DDP combination group was more affected. Ki67 expression in subcutaneous tumors was reduced in the KW2478 or DDP treatment groups compared to the vehicle control group, with the lowest expression observed in the KW2478 + DDP combination group. The fluorescence intensity of subcutaneous tumors was higher in both the KW2478 and DDP treatment groups compared to the vehicle control group, and the KW2478 + DDP combination group exhibited the strongest fluorescence intensity among them.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusion: &lt;/strong&gt;The combination of KW2478 and cisplatin inhibits colorectal cancer cell proliferation and induces apoptosis by regulating the PI3K/AKT/mTOR p","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142998749","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
Investigation of the Anticarcinogenic Effects of Hypericum perforatum Extract on Human Thyroid Cancer. 贯叶连翘提取物对人甲状腺癌的抗癌作用研究。
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-01-14 DOI: 10.2174/0118715206340411241120051020
Cigdem Gokcek-Sarac, Gizem Altunkaya, Serdar Karakurt

Introduction/objective: Plants and their bioactive compounds play a crucial role in the pharmaceutical industry for treating cancer. To date, the cytotoxic and antiproliferative effects of Hypericum perforatum methanol extract on human thyroid cancer cell lines have not been thoroughly explored. The present study aimed to assess the potential anti-cancer effects of HPME on human thyroid cancer and investigate its potential therapeutic benefits.

Methods: HPME was prepared using the maceration method, and its antioxidant activity was examined. Cytotoxicity studies were then carried out, followed by an investigation of the possible effects of HPME on metastasis and colony-forming capacities of human thyroid cancer cells. Afterward, qRT-PCR, western blotting, and apoptosis assays were performed.

Results: Cytotoxicity studies revealed notable cytotoxicity of HPME against the TT cell line. Moreover, HPME significantly curtailed metastasis and invasion of TT cells in an in vitro wound healing assay. Analyses of gene expressions demonstrated an elevation in caspase-12, caspase-3, and Bax, coupled with a reduction in BcL-2, APOE, and CLU expression. Following HPME treatment, there was an increase in the protein expression levels of Bax and Caspase-12, while a decrease in the BcL-2, APOE, and CLU protein expression. Furthermore, apoptotic studies indicated an increase in early apoptosis.

Conclusion: Overall results revealed that HPME demonstrates a notable antioxidant capacity in human thyroid cancer. It exerts an influence on crucial biological processes associated with cancer, indicating its potential to hinder the proliferation of human thyroid cancer cells by enhancing apoptosis through the upregulation of gene and protein expression, particularly involving caspases.

简介/目的:植物及其生物活性化合物在治疗癌症的制药工业中起着至关重要的作用。迄今为止,贯叶连翘甲醇提取物对人甲状腺癌细胞的细胞毒性和抗增殖作用尚未得到充分的研究。本研究旨在评估HPME对人甲状腺癌的潜在抗癌作用,并探讨其潜在的治疗效益。方法:采用浸渍法制备HPME,考察其抗氧化活性。随后进行了细胞毒性研究,随后调查了HPME对人甲状腺癌细胞转移和集落形成能力的可能影响。随后进行qRT-PCR、western blotting和细胞凋亡检测。结果:细胞毒性研究显示HPME对TT细胞系具有明显的细胞毒性。此外,在体外伤口愈合实验中,HPME显著减少TT细胞的转移和侵袭。基因表达分析显示caspase-12、caspase-3和Bax表达升高,同时BcL-2、APOE和CLU表达降低。HPME处理后,Bax和Caspase-12蛋白表达水平升高,BcL-2、APOE和CLU蛋白表达降低。此外,凋亡研究表明早期凋亡增加。结论:HPME对人甲状腺癌具有显著的抗氧化作用。它对与癌症相关的关键生物学过程产生影响,表明它有可能通过上调基因和蛋白质表达(特别是涉及半胱天冬酶)来增强细胞凋亡,从而阻碍人类甲状腺癌细胞的增殖。
{"title":"Investigation of the Anticarcinogenic Effects of Hypericum perforatum Extract on Human Thyroid Cancer.","authors":"Cigdem Gokcek-Sarac, Gizem Altunkaya, Serdar Karakurt","doi":"10.2174/0118715206340411241120051020","DOIUrl":"https://doi.org/10.2174/0118715206340411241120051020","url":null,"abstract":"<p><strong>Introduction/objective: </strong>Plants and their bioactive compounds play a crucial role in the pharmaceutical industry for treating cancer. To date, the cytotoxic and antiproliferative effects of Hypericum perforatum methanol extract on human thyroid cancer cell lines have not been thoroughly explored. The present study aimed to assess the potential anti-cancer effects of HPME on human thyroid cancer and investigate its potential therapeutic benefits.</p><p><strong>Methods: </strong>HPME was prepared using the maceration method, and its antioxidant activity was examined. Cytotoxicity studies were then carried out, followed by an investigation of the possible effects of HPME on metastasis and colony-forming capacities of human thyroid cancer cells. Afterward, qRT-PCR, western blotting, and apoptosis assays were performed.</p><p><strong>Results: </strong>Cytotoxicity studies revealed notable cytotoxicity of HPME against the TT cell line. Moreover, HPME significantly curtailed metastasis and invasion of TT cells in an in vitro wound healing assay. Analyses of gene expressions demonstrated an elevation in caspase-12, caspase-3, and Bax, coupled with a reduction in BcL-2, APOE, and CLU expression. Following HPME treatment, there was an increase in the protein expression levels of Bax and Caspase-12, while a decrease in the BcL-2, APOE, and CLU protein expression. Furthermore, apoptotic studies indicated an increase in early apoptosis.</p><p><strong>Conclusion: </strong>Overall results revealed that HPME demonstrates a notable antioxidant capacity in human thyroid cancer. It exerts an influence on crucial biological processes associated with cancer, indicating its potential to hinder the proliferation of human thyroid cancer cells by enhancing apoptosis through the upregulation of gene and protein expression, particularly involving caspases.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142998835","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
Bioactive Products Targeting C-Met As Potential Antitumour Drugs. 靶向C-Met的生物活性产品作为潜在的抗肿瘤药物。
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-01-13 DOI: 10.2174/0118715206346207241217064022
Liying Zhao, Chunmei Qian, Xiaoqi Ma, Xiaoyu Wang

Mesenchymal‒epithelial transition factor (c-Met), a receptortyrosine kinase (RTK), plays a vital role in cell proliferation, migration and invasion, and tumour metastasis.

Objective: With increasing duration of treatment, many tumours gradually develop drug resistance. Therefore, novel antitumour drugs need to be developed to treat patients with tumours. Targeting c-met inhibitors may be an effective treatment strategy.

Methods: Scientific databases such as ScienceDirect, PubMed, the Wiley Online Library, and Social Sciences Citation Index were used to collect information. All the relevant literature was reviewed, and the available literature was screened. The upstream and downstream pathways of c-Met and their relevance to antitumour effects were searched based on the articles' title, abstract, and full text. The c-Met-targeting drugs with antitumour effects are summarized below. A "citation within a citation" or snowballing approach was used in this screening process to identify additional papers that may have been missed in the initial literature screening process. High-quality studies published in peer-reviewed journals were summarized and prioritized for citation in the review.

Results: In recent years, research on small-molecule targeted drugs has developed rapidly. Many results have also been achieved in the synthesis and isolation of c-Met inhibitors from natural compounds and traditional Chinese medicines.

Conclusion: This article summarizes the developments in anti-c-Met drugs, which are synthesized and isolated from natural compounds and traditional Chinese medicine (TCM). This study provides primary resources for the development of c-Met inhibitors.

间充质上皮转化因子(c-Met)是一种受体酪氨酸激酶(RTK),在细胞增殖、迁移侵袭和肿瘤转移中起着至关重要的作用。目的:随着治疗时间的延长,许多肿瘤逐渐产生耐药性。因此,需要开发新的抗肿瘤药物来治疗肿瘤患者。靶向c-met抑制剂可能是一种有效的治疗策略。方法:利用ScienceDirect、PubMed、Wiley Online Library、Social Sciences Citation Index等科学数据库收集信息。对所有相关文献进行综述,并对现有文献进行筛选。根据文章标题、摘要和全文检索c-Met的上游和下游途径及其与抗肿瘤作用的相关性。现将具有抗肿瘤作用的c- met靶向药物总结如下。在筛选过程中使用了“引文中的引文”或滚雪球方法,以确定在最初的文献筛选过程中可能遗漏的其他论文。本综述对发表在同行评议期刊上的高质量研究进行了总结和优先引用。结果:近年来,小分子靶向药物的研究发展迅速。从天然化合物和中药中合成和分离c-Met抑制剂也取得了许多成果。结论:本文综述了从天然化合物和中药中合成和分离的抗c- met药物的研究进展。本研究为开发c-Met抑制剂提供了主要资源。
{"title":"Bioactive Products Targeting C-Met As Potential Antitumour Drugs.","authors":"Liying Zhao, Chunmei Qian, Xiaoqi Ma, Xiaoyu Wang","doi":"10.2174/0118715206346207241217064022","DOIUrl":"https://doi.org/10.2174/0118715206346207241217064022","url":null,"abstract":"<p><p>Mesenchymal‒epithelial transition factor (c-Met), a receptortyrosine kinase (RTK), plays a vital role in cell proliferation, migration and invasion, and tumour metastasis.</p><p><strong>Objective: </strong>With increasing duration of treatment, many tumours gradually develop drug resistance. Therefore, novel antitumour drugs need to be developed to treat patients with tumours. Targeting c-met inhibitors may be an effective treatment strategy.</p><p><strong>Methods: </strong>Scientific databases such as ScienceDirect, PubMed, the Wiley Online Library, and Social Sciences Citation Index were used to collect information. All the relevant literature was reviewed, and the available literature was screened. The upstream and downstream pathways of c-Met and their relevance to antitumour effects were searched based on the articles' title, abstract, and full text. The c-Met-targeting drugs with antitumour effects are summarized below. A \"citation within a citation\" or snowballing approach was used in this screening process to identify additional papers that may have been missed in the initial literature screening process. High-quality studies published in peer-reviewed journals were summarized and prioritized for citation in the review.</p><p><strong>Results: </strong>In recent years, research on small-molecule targeted drugs has developed rapidly. Many results have also been achieved in the synthesis and isolation of c-Met inhibitors from natural compounds and traditional Chinese medicines.</p><p><strong>Conclusion: </strong>This article summarizes the developments in anti-c-Met drugs, which are synthesized and isolated from natural compounds and traditional Chinese medicine (TCM). This study provides primary resources for the development of c-Met inhibitors.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142982529","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
Rhizopogon luteolus and Ganoderma adspersum Extracts Inhibit Invasion through the Crosstalk between Anti-oxidant Activity and Apoptosis Induced by pAKT/Rb. 毛根连根和灵芝提取物通过抗氧化活性与pAKT/Rb诱导的细胞凋亡之间的串扰抑制入侵。
IF 2.6 4区 医学 Q3 CHEMISTRY, MEDICINAL Pub Date : 2025-01-13 DOI: 10.2174/0118715206349668241118104255
Aydın Demiray, Ege Rıza Karagür, Gülsen Tel-Cayan, Onur Tokgün, Hakan Akça, Mehmet Emin Duru

Objective: Lung cancer is the primary cause of cancer-related deaths globally. Protein kinase B (AKT) protein is associated with many pathways in non-small cell lung cancer (NSCLC), such as proliferation, migration, invasion, and apoptosis. Mushrooms have a long history of being used in traditional medicine to treat various diseases. Scientists have been exploring the potential of mushrooms for their antioxidant and anticancer properties. In our study, the anti-oxidant, invasion, and apoptosis effects of mushroom extracts were investigated in NSCLC.

Materials and methods: Non-Small Cell Lung Cancer cell lines H1299, PC-3, and PC-14 were used in our study. After obtaining the extracts of Rhizopogon luteolus and Ganoderma adspersum, IC50 value was calculated as 25.04-11.73-16.54 ng/ul for R. luteolus and 2.97-1.53-1.01ug/ul for G. adspersum, respectively, in H1299, PC3 and PC14 cell lines. Afterward, proliferative and invasion effects, as well as apoptosis and anti-oxidant effects, were investigated using the IC50 dose. Western blotting was performed to investigate the pathways of these effects.

Results: According to the results of our study, Rhizopogon luteolus and Ganoderma adspersum extracts have anti-proliferative and anti-invasive effects on non-small lung cancer cell lines and induced apoptosis, which has been found to increase the anti-oxidant effect. It was found that this effect was due to cross-talk between antioxidant activity and the AKT-Rb pathway.

Conclusion: We anticipate that Rhizopogon luteolus and Ganoderma adspersum extracts will be effective in cancer treatment by suppressing lung cancer cells via p-Akt and Rb.

目的:肺癌是全球癌症相关死亡的主要原因。蛋白激酶B (AKT)蛋白与非小细胞肺癌(NSCLC)的增殖、迁移、侵袭和凋亡等多种途径相关。蘑菇在传统医学中用于治疗各种疾病有着悠久的历史。科学家们一直在探索蘑菇的抗氧化和抗癌特性。本研究探讨了香菇提取物在非小细胞肺癌中的抗氧化、侵袭和凋亡作用。材料与方法:本研究采用非小细胞肺癌细胞系H1299、PC-3、PC-14。在H1299、PC3和PC14细胞系中,得到毛根芝和仙芝提取物,计算毛根芝和仙芝的IC50值分别为25.04 ~ 11.73 ~ 16.54 ng/ul和2.97 ~ 1.53 ~ 1.01ug/ul。然后,使用IC50剂量研究细胞的增殖和侵袭作用,以及细胞凋亡和抗氧化作用。Western blotting研究这些作用的途径。结果:根据我们的研究结果,黄连和灵芝提取物对非小细胞肺癌细胞系具有抗增殖和抗侵袭作用,诱导细胞凋亡,增强抗氧化作用。发现这种作用是由于抗氧化活性和AKT-Rb途径之间的交叉作用。结论:我们预测黄连和灵芝提取物可能通过p-Akt和Rb抑制肺癌细胞,从而起到治疗肿瘤的作用。
{"title":"Rhizopogon luteolus and Ganoderma adspersum Extracts Inhibit Invasion through the Crosstalk between Anti-oxidant Activity and Apoptosis Induced by pAKT/Rb.","authors":"Aydın Demiray, Ege Rıza Karagür, Gülsen Tel-Cayan, Onur Tokgün, Hakan Akça, Mehmet Emin Duru","doi":"10.2174/0118715206349668241118104255","DOIUrl":"https://doi.org/10.2174/0118715206349668241118104255","url":null,"abstract":"<p><strong>Objective: </strong>Lung cancer is the primary cause of cancer-related deaths globally. Protein kinase B (AKT) protein is associated with many pathways in non-small cell lung cancer (NSCLC), such as proliferation, migration, invasion, and apoptosis. Mushrooms have a long history of being used in traditional medicine to treat various diseases. Scientists have been exploring the potential of mushrooms for their antioxidant and anticancer properties. In our study, the anti-oxidant, invasion, and apoptosis effects of mushroom extracts were investigated in NSCLC.</p><p><strong>Materials and methods: </strong>Non-Small Cell Lung Cancer cell lines H1299, PC-3, and PC-14 were used in our study. After obtaining the extracts of Rhizopogon luteolus and Ganoderma adspersum, IC50 value was calculated as 25.04-11.73-16.54 ng/ul for R. luteolus and 2.97-1.53-1.01ug/ul for G. adspersum, respectively, in H1299, PC3 and PC14 cell lines. Afterward, proliferative and invasion effects, as well as apoptosis and anti-oxidant effects, were investigated using the IC50 dose. Western blotting was performed to investigate the pathways of these effects.</p><p><strong>Results: </strong>According to the results of our study, Rhizopogon luteolus and Ganoderma adspersum extracts have anti-proliferative and anti-invasive effects on non-small lung cancer cell lines and induced apoptosis, which has been found to increase the anti-oxidant effect. It was found that this effect was due to cross-talk between antioxidant activity and the AKT-Rb pathway.</p><p><strong>Conclusion: </strong>We anticipate that Rhizopogon luteolus and Ganoderma adspersum extracts will be effective in cancer treatment by suppressing lung cancer cells via p-Akt and Rb.</p>","PeriodicalId":7934,"journal":{"name":"Anti-cancer agents in medicinal chemistry","volume":" ","pages":""},"PeriodicalIF":2.6,"publicationDate":"2025-01-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142982531","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