首页 > 最新文献

Anti-Cancer Drugs最新文献

英文 中文
Preclinical evidence that fibroblast growth factor receptor pathway inhibition by BGJ398 enhances small cell lung cancer response to chemotherapy. 临床前证据表明BGJ398抑制成纤维细胞生长因子受体通路可增强小细胞肺癌对化疗的反应。
IF 1.8 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-17 DOI: 10.1097/CAD.0000000000001683
Yingying Shen, Yan Jiang, Junyao Wu, Chenyu Wang, Jiao Bo Kun Huang, Jie Liu, Sen Chen

Small cell lung cancer (SCLC) is a highly aggressive form of lung cancer with limited therapeutic options and poor prognosis. In this study, we explored the therapeutic potential of BGJ398, a selective fibroblast growth factor receptor (FGFR) inhibitor, alone and in combination with standard chemotherapy (cisplatin and paclitaxel) in SCLC. High-throughput screening of kinase inhibitors was performed on three SCLC cell lines (NCI-H446, NCI-H69, and NCI-H182), identifying BGJ398 as one of the most potent and selective inhibitors. BGJ398 demonstrated significant synergy with cisplatin and paclitaxel in vitro, as indicated by combination index values below 1. In vivo, combination treatments significantly inhibited tumor growth and extended survival in SCLC xenograft models compared to monotherapies. Notably, the combination of BGJ398 with cisplatin exhibited the most pronounced tumor suppression and survival benefits. Immunohistochemistry analysis confirmed that BGJ398 effectively inhibited FGFR signaling pathways, reducing levels of phosphorylated FGFR, protein kinase B, signal transducer and activator of transcription 3, and extracellular signal-regulated kinase. These findings suggest that BGJ398, particularly in combination with chemotherapy, holds significant promise as a treatment strategy for SCLC, providing enhanced anti-tumor efficacy and improved survival outcomes.

小细胞肺癌(SCLC)是一种高度侵袭性的肺癌,治疗选择有限,预后差。在这项研究中,我们探索了BGJ398的治疗潜力,BGJ398是一种选择性成纤维细胞生长因子受体(FGFR)抑制剂,单独或联合标准化疗(顺铂和紫杉醇)治疗SCLC。在三种SCLC细胞系(NCI-H446, NCI-H69和NCI-H182)上进行了激酶抑制剂的高通量筛选,确定BGJ398是最有效和选择性的抑制剂之一。BGJ398与顺铂、紫杉醇体外协同作用显著,联合指数低于1。在体内,与单一治疗相比,联合治疗显著抑制SCLC异种移植模型的肿瘤生长并延长生存期。值得注意的是,BGJ398与顺铂联合使用显示出最明显的肿瘤抑制和生存益处。免疫组织化学分析证实,BGJ398有效抑制FGFR信号通路,降低磷酸化FGFR、蛋白激酶B、信号传导和转录激活因子3以及细胞外信号调节激酶的水平。这些发现表明,BGJ398,特别是联合化疗,作为SCLC的治疗策略具有重要的前景,提供增强的抗肿瘤疗效和改善的生存结果。
{"title":"Preclinical evidence that fibroblast growth factor receptor pathway inhibition by BGJ398 enhances small cell lung cancer response to chemotherapy.","authors":"Yingying Shen, Yan Jiang, Junyao Wu, Chenyu Wang, Jiao Bo Kun Huang, Jie Liu, Sen Chen","doi":"10.1097/CAD.0000000000001683","DOIUrl":"https://doi.org/10.1097/CAD.0000000000001683","url":null,"abstract":"<p><p>Small cell lung cancer (SCLC) is a highly aggressive form of lung cancer with limited therapeutic options and poor prognosis. In this study, we explored the therapeutic potential of BGJ398, a selective fibroblast growth factor receptor (FGFR) inhibitor, alone and in combination with standard chemotherapy (cisplatin and paclitaxel) in SCLC. High-throughput screening of kinase inhibitors was performed on three SCLC cell lines (NCI-H446, NCI-H69, and NCI-H182), identifying BGJ398 as one of the most potent and selective inhibitors. BGJ398 demonstrated significant synergy with cisplatin and paclitaxel in vitro, as indicated by combination index values below 1. In vivo, combination treatments significantly inhibited tumor growth and extended survival in SCLC xenograft models compared to monotherapies. Notably, the combination of BGJ398 with cisplatin exhibited the most pronounced tumor suppression and survival benefits. Immunohistochemistry analysis confirmed that BGJ398 effectively inhibited FGFR signaling pathways, reducing levels of phosphorylated FGFR, protein kinase B, signal transducer and activator of transcription 3, and extracellular signal-regulated kinase. These findings suggest that BGJ398, particularly in combination with chemotherapy, holds significant promise as a treatment strategy for SCLC, providing enhanced anti-tumor efficacy and improved survival outcomes.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142998840","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
Dual inhibition of TYK2 and PD-L1 boosts immune response in triple negative breast cancer. 双重抑制JAK3和PD-L1增强三阴性乳腺癌的免疫应答。
IF 1.8 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-17 DOI: 10.1097/CAD.0000000000001685
Huali Xiang, Binfeng Tu, Xin Feng, Linjing Chen, Yajuan Huang

Recent studies have shown that Janus Kinase inhibitors can enhance the tumor therapeutic effect of immune checkpoint inhibitors. However, it remains to be studied whether TYK2 selective inhibitors can enhance the therapeutic effect of small molecule PD-L1 inhibitors in triple-negative breast cancer (TNBC). We verified the efficacy of the combination of the selective TYK2 inhibitor Deucravacitinib and the small molecule inhibitor of PD-L1, INCB086550, in two TNBC animal models: a syngeneic mouse model (4T1 with humanized PD-L1) and a peripheral blood mononuclear cell (PBMC)-humanized model (MDA-MB-231). Following that, we explored the regulation of immune cell activity in tumors by the combined treatment using flow cytometry. Finally, we validated the expression of genes related to the regulated immune cells through reverse transcription-PCR. Both animal models demonstrated that the addition of a TYK2 inhibitor to a PD-L1 inhibitor significantly enhanced the antitumor capabilities of mice with good safety profiles. The combined therapy significantly elevated the counts of T, B, and natural killer cells while concurrently diminishing myeloid-derived suppressor cells in the syngeneic model. Similarly, in the PBMC-humanized model, this therapy markedly augmented progenitor-like and proliferative precursor-like CD8 T cells, while effectively diminishing exhausted and terminally differentiated CD8 T cell populations. This enhanced antitumor effect is associated with the modulation of antitumor immune-related gene expression by the combined therapy. The combination of TYK2 inhibitors and immune checkpoint inhibitors is a potentially effective strategy for treating TNBC.

最近的研究表明,Janus激酶抑制剂可以增强免疫检查点抑制剂的肿瘤治疗效果。然而,TYK2选择性抑制剂是否能增强小分子PD-L1抑制剂在三阴性乳腺癌(TNBC)中的治疗效果仍有待研究。我们在两种TNBC动物模型中验证了选择性TYK2抑制剂Deucravacitinib与PD-L1小分子抑制剂INCB086550联合治疗的疗效:一种是同基因小鼠模型(4T1与人源化PD-L1),另一种是外周血单核细胞(PBMC)人源化模型(MDA-MB-231)。随后,我们利用流式细胞术探讨了联合治疗对肿瘤免疫细胞活性的调节作用。最后,我们通过逆转录- pcr验证了调控免疫细胞相关基因的表达。两种动物模型均表明,在PD-L1抑制剂中添加TYK2抑制剂可显著增强小鼠的抗肿瘤能力,且具有良好的安全性。在同基因模型中,联合治疗显著提高T、B和自然杀伤细胞的计数,同时减少髓源性抑制细胞。同样,在pmc人源化模型中,该疗法显著增强了祖细胞样和增殖前体细胞样CD8 T细胞,同时有效地减少了耗尽和终末分化的CD8 T细胞群。这种增强的抗肿瘤效果与联合治疗对抗肿瘤免疫相关基因表达的调节有关。TYK2抑制剂和免疫检查点抑制剂联合使用是治疗TNBC的潜在有效策略。
{"title":"Dual inhibition of TYK2 and PD-L1 boosts immune response in triple negative breast cancer.","authors":"Huali Xiang, Binfeng Tu, Xin Feng, Linjing Chen, Yajuan Huang","doi":"10.1097/CAD.0000000000001685","DOIUrl":"10.1097/CAD.0000000000001685","url":null,"abstract":"<p><p>Recent studies have shown that Janus Kinase inhibitors can enhance the tumor therapeutic effect of immune checkpoint inhibitors. However, it remains to be studied whether TYK2 selective inhibitors can enhance the therapeutic effect of small molecule PD-L1 inhibitors in triple-negative breast cancer (TNBC). We verified the efficacy of the combination of the selective TYK2 inhibitor Deucravacitinib and the small molecule inhibitor of PD-L1, INCB086550, in two TNBC animal models: a syngeneic mouse model (4T1 with humanized PD-L1) and a peripheral blood mononuclear cell (PBMC)-humanized model (MDA-MB-231). Following that, we explored the regulation of immune cell activity in tumors by the combined treatment using flow cytometry. Finally, we validated the expression of genes related to the regulated immune cells through reverse transcription-PCR. Both animal models demonstrated that the addition of a TYK2 inhibitor to a PD-L1 inhibitor significantly enhanced the antitumor capabilities of mice with good safety profiles. The combined therapy significantly elevated the counts of T, B, and natural killer cells while concurrently diminishing myeloid-derived suppressor cells in the syngeneic model. Similarly, in the PBMC-humanized model, this therapy markedly augmented progenitor-like and proliferative precursor-like CD8 T cells, while effectively diminishing exhausted and terminally differentiated CD8 T cell populations. This enhanced antitumor effect is associated with the modulation of antitumor immune-related gene expression by the combined therapy. The combination of TYK2 inhibitors and immune checkpoint inhibitors is a potentially effective strategy for treating TNBC.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2025-01-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142943243","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
Berberine inhibits prostate cancer progression by inducing ferroptosis: evidence from network pharmacology. 小檗碱通过诱导铁下垂抑制前列腺癌进展:来自网络药理学的证据。
IF 1.8 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-14 DOI: 10.1097/CAD.0000000000001691
Peiliang Zou, Shenghai Li, Qixiong He, Chixing Zheng

The uncertain ferroptosis-related role of berberine in prostate cancer was explored using network pharmacology methodology. Integration of ferroptosis targets in prostate cancer from the Genecard database and berberine targets from the Traditional Chinese Medicine Systems Pharmacology and SwissTargetPrediction databases revealed 17 common targets. Among these, 10 hub genes, including CCNB1, CDK1, AURKA, AR, CDC42, ICAM1, TYMS, NTRK1, PTGS2, and SCD, were identified. Enrichment analyses yielded 799 Gene Ontology terms and 23 Kyoto Encyclopedia of Genes and Genomes pathways associated with berberine-related targets. Molecular docking simulations indicated berberine's binding capacity to all hub genes. In-vitro studies on LNCaP and PC3 cells demonstrated berberine's inhibition of cell proliferation and significant downregulation of TYMS, CCNB1, AURKA, CDK1, and SCD in both cell lines. Berberine exhibited cell line-specific effects by reducing AR expression in LNCaP cells and suppressing ICAM1 in PC3 cells. Overall, berberine shows promise in inhibiting prostate cancer progression through modulation of ferroptosis-related genes, including TYMS, AR, CCNB1, AURKA, CDK1, ICAM1, NTRK1, SCD, and CDC42.

利用网络药理学方法探讨了小檗碱在前列腺癌中与铁中毒相关的不确定作用。整合来自Genecard数据库的前列腺癌铁下垂靶点和来自中医系统药理学和SwissTargetPrediction数据库的小檗碱靶点,发现了17个共同靶点。其中,共鉴定出CCNB1、CDK1、AURKA、AR、CDC42、ICAM1、TYMS、NTRK1、PTGS2、SCD等10个枢纽基因。富集分析得到799个基因本体术语和23个与小檗碱相关靶点相关的京都基因和基因组百科全书路径。分子对接模拟显示了小檗碱与所有枢纽基因的结合能力。对LNCaP和PC3细胞的体外研究表明,小檗碱对两种细胞系的细胞增殖均有抑制作用,并显著下调TYMS、CCNB1、AURKA、CDK1和SCD。小檗碱通过降低LNCaP细胞中的AR表达和抑制PC3细胞中的ICAM1表现出细胞系特异性作用。总的来说,小檗碱通过调节铁致凋亡相关基因,包括TYMS、AR、CCNB1、AURKA、CDK1、ICAM1、NTRK1、SCD和CDC42,显示出抑制前列腺癌进展的希望。
{"title":"Berberine inhibits prostate cancer progression by inducing ferroptosis: evidence from network pharmacology.","authors":"Peiliang Zou, Shenghai Li, Qixiong He, Chixing Zheng","doi":"10.1097/CAD.0000000000001691","DOIUrl":"https://doi.org/10.1097/CAD.0000000000001691","url":null,"abstract":"<p><p>The uncertain ferroptosis-related role of berberine in prostate cancer was explored using network pharmacology methodology. Integration of ferroptosis targets in prostate cancer from the Genecard database and berberine targets from the Traditional Chinese Medicine Systems Pharmacology and SwissTargetPrediction databases revealed 17 common targets. Among these, 10 hub genes, including CCNB1, CDK1, AURKA, AR, CDC42, ICAM1, TYMS, NTRK1, PTGS2, and SCD, were identified. Enrichment analyses yielded 799 Gene Ontology terms and 23 Kyoto Encyclopedia of Genes and Genomes pathways associated with berberine-related targets. Molecular docking simulations indicated berberine's binding capacity to all hub genes. In-vitro studies on LNCaP and PC3 cells demonstrated berberine's inhibition of cell proliferation and significant downregulation of TYMS, CCNB1, AURKA, CDK1, and SCD in both cell lines. Berberine exhibited cell line-specific effects by reducing AR expression in LNCaP cells and suppressing ICAM1 in PC3 cells. Overall, berberine shows promise in inhibiting prostate cancer progression through modulation of ferroptosis-related genes, including TYMS, AR, CCNB1, AURKA, CDK1, ICAM1, NTRK1, SCD, and CDC42.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2025-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142976983","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
Clinical efficacy of pyrotinib combined with chemotherapy for neoadjuvant treatment in HER2-positive breast cancer: a single-center study. 吡罗替尼联合化疗新辅助治疗her2阳性乳腺癌的临床疗效:一项单中心研究
IF 1.8 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-14 DOI: 10.1097/CAD.0000000000001690
Benkai Wei, Huanhuan Yan, Fan Li, Jun Shen

This study aimed to evaluate the efficacy of pyrotinib, an orally administered small molecule tyrosine kinase inhibitor, combined with neoadjuvant chemotherapy in treating patients with human epidermal growth factor receptor 2 (HER2)-positive breast cancer. Pyrotinib works by inhibiting the HER2 signaling pathway, thereby preventing tumor cell growth. This single-arm clinical trial aimed to assess the total pathological complete response (tpCR; ypT0/is and ypN0) rate as the primary endpoint. A total of 27 patients were enrolled, each receiving 4-8 cycles of pyrotinib in combination with neoadjuvant chemotherapy. Pyrotinib combined with neoadjuvant chemotherapy demonstrated notable antitumor activity in patients with HER2-positive breast cancer. Among 26 patients, the tpCR rate was 26% (7/26), while the breast pathological complete response rate was 30% (8/26), indicating complete inhibition of the primary tumor in some cases. Notably, patients with HR-negative breast cancer demonstrated a higher tpCR rate compared with those with HR-positive breast cancer. The treatment regimen was well-tolerated. Diarrhea was the most common adverse event, occurring in 92.3% of patients, with 46.2% experiencing grade 3 or higher diarrhea. No severe adverse events or treatment-related fatalities were reported.

本研究旨在评估口服小分子酪氨酸激酶抑制剂pyrotinib联合新辅助化疗治疗人表皮生长因子受体2 (HER2)阳性乳腺癌患者的疗效。Pyrotinib通过抑制HER2信号通路起作用,从而阻止肿瘤细胞生长。这项单臂临床试验旨在评估总病理完全缓解(tpCR;ypT0/is和ypN0)率作为主要终点。共有27例患者入组,每位患者接受4-8个周期的吡罗替尼联合新辅助化疗。吡咯替尼联合新辅助化疗在her2阳性乳腺癌患者中显示出显著的抗肿瘤活性。26例患者中,tpCR率为26%(7/26),乳腺病理完全缓解率为30%(8/26),部分患者原发肿瘤得到完全抑制。值得注意的是,hr阴性乳腺癌患者的tpCR率高于hr阳性乳腺癌患者。治疗方案耐受性良好。腹泻是最常见的不良事件,发生在92.3%的患者中,46.2%的患者出现3级或更高级别的腹泻。无严重不良事件或治疗相关死亡报告。
{"title":"Clinical efficacy of pyrotinib combined with chemotherapy for neoadjuvant treatment in HER2-positive breast cancer: a single-center study.","authors":"Benkai Wei, Huanhuan Yan, Fan Li, Jun Shen","doi":"10.1097/CAD.0000000000001690","DOIUrl":"https://doi.org/10.1097/CAD.0000000000001690","url":null,"abstract":"<p><p>This study aimed to evaluate the efficacy of pyrotinib, an orally administered small molecule tyrosine kinase inhibitor, combined with neoadjuvant chemotherapy in treating patients with human epidermal growth factor receptor 2 (HER2)-positive breast cancer. Pyrotinib works by inhibiting the HER2 signaling pathway, thereby preventing tumor cell growth. This single-arm clinical trial aimed to assess the total pathological complete response (tpCR; ypT0/is and ypN0) rate as the primary endpoint. A total of 27 patients were enrolled, each receiving 4-8 cycles of pyrotinib in combination with neoadjuvant chemotherapy. Pyrotinib combined with neoadjuvant chemotherapy demonstrated notable antitumor activity in patients with HER2-positive breast cancer. Among 26 patients, the tpCR rate was 26% (7/26), while the breast pathological complete response rate was 30% (8/26), indicating complete inhibition of the primary tumor in some cases. Notably, patients with HR-negative breast cancer demonstrated a higher tpCR rate compared with those with HR-positive breast cancer. The treatment regimen was well-tolerated. Diarrhea was the most common adverse event, occurring in 92.3% of patients, with 46.2% experiencing grade 3 or higher diarrhea. No severe adverse events or treatment-related fatalities were reported.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2025-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142982537","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
Identification and validation of mitochondria-related LncRNA signatures as a novel prognostic model for glioma. 线粒体相关LncRNA标记作为胶质瘤新预后模型的鉴定和验证。
IF 1.8 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-14 DOI: 10.1097/CAD.0000000000001677
Kaihan Deng, Wei Zhao, Lin Dai, Zixuan Jing, Lixin Ma

A predictive model for long-term survival is needed, and mitochondrial dysfunction is a key feature of cancer metabolism, though its link to glioma is not well understood. The aim of this study was to identify the molecular characteristics associated with glioma prognosis and explore its potential function. We analyzed RNA-seq data from The Cancer Genome Atlas and identified differentially expressed mitochondrial long noncoding RNAs (lncRNAs) using R's 'limma' package. A prognostic model was developed using 10 selected lncRNAs and validated with Cox regression and least absolute shrinkage and selection operator algorithm. The model's efficacy was assessed using Kaplan-Meier and receiver operating characteristic curve analyses, and its correlation with immune cell profiles and drug sensitivity was explored. A 10-mitochondria-related LncRNA signature was generated. The median risk score values are used to classify glioma samples into low-risk and high-risk groups. In breast patients, the signature-based risk score demonstrated a more potent ability to predict survival than conventional clinicopathological features. Furthermore, we noted a substantial disparity in the number of immune cells, including B cells, CD8+T cells, and macrophages, between the two groups. In addition, the high-risk group exhibited lower half-maximal inhibitory concentration values for specific chemotherapy medications, including bortezomib, luminespib, rapamycin, and 5-fluorouracil. Our study elucidates the diagnostic and prognostic value of mitochondria-related-lncRNAs in the promotion, suppression, and treatment of glioma.

需要一个长期生存的预测模型,线粒体功能障碍是癌症代谢的一个关键特征,尽管它与胶质瘤的联系还不清楚。本研究的目的是确定与胶质瘤预后相关的分子特征并探讨其潜在功能。我们分析了来自癌症基因组图谱的RNA-seq数据,并使用R的“limma”软件包鉴定了差异表达的线粒体长链非编码rna (lncRNAs)。使用10个选定的lncrna建立预后模型,并使用Cox回归、最小绝对收缩和选择算子算法进行验证。采用Kaplan-Meier和受者工作特征曲线分析评估模型的疗效,并探讨其与免疫细胞谱和药物敏感性的相关性。产生了10个线粒体相关的LncRNA信号。使用中位风险评分值将胶质瘤样本分为低风险组和高风险组。在乳腺癌患者中,基于特征的风险评分比传统的临床病理特征更能预测患者的生存。此外,我们注意到两组之间免疫细胞数量的巨大差异,包括B细胞、CD8+T细胞和巨噬细胞。此外,高危组对特定化疗药物,包括硼替佐米、鲁米斯匹、雷帕霉素和5-氟尿嘧啶,表现出较低的半最大抑制浓度值。我们的研究阐明了线粒体相关lncrna在促进、抑制和治疗胶质瘤中的诊断和预后价值。
{"title":"Identification and validation of mitochondria-related LncRNA signatures as a novel prognostic model for glioma.","authors":"Kaihan Deng, Wei Zhao, Lin Dai, Zixuan Jing, Lixin Ma","doi":"10.1097/CAD.0000000000001677","DOIUrl":"https://doi.org/10.1097/CAD.0000000000001677","url":null,"abstract":"<p><p>A predictive model for long-term survival is needed, and mitochondrial dysfunction is a key feature of cancer metabolism, though its link to glioma is not well understood. The aim of this study was to identify the molecular characteristics associated with glioma prognosis and explore its potential function. We analyzed RNA-seq data from The Cancer Genome Atlas and identified differentially expressed mitochondrial long noncoding RNAs (lncRNAs) using R's 'limma' package. A prognostic model was developed using 10 selected lncRNAs and validated with Cox regression and least absolute shrinkage and selection operator algorithm. The model's efficacy was assessed using Kaplan-Meier and receiver operating characteristic curve analyses, and its correlation with immune cell profiles and drug sensitivity was explored. A 10-mitochondria-related LncRNA signature was generated. The median risk score values are used to classify glioma samples into low-risk and high-risk groups. In breast patients, the signature-based risk score demonstrated a more potent ability to predict survival than conventional clinicopathological features. Furthermore, we noted a substantial disparity in the number of immune cells, including B cells, CD8+T cells, and macrophages, between the two groups. In addition, the high-risk group exhibited lower half-maximal inhibitory concentration values for specific chemotherapy medications, including bortezomib, luminespib, rapamycin, and 5-fluorouracil. Our study elucidates the diagnostic and prognostic value of mitochondria-related-lncRNAs in the promotion, suppression, and treatment of glioma.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2025-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142982539","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
WDR62 mediates MAPK/ERK pathway to stimulate DNA damage repair and attenuate cisplatin sensitivity in lung adenocarcinoma. WDR62介导MAPK/ERK通路刺激肺腺癌DNA损伤修复,减弱顺铂敏感性。
IF 1.8 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-14 DOI: 10.1097/CAD.0000000000001682
Xu Li, Yingwei Guo, Zecheng Qi, Yi Zheng

Chemotherapy resistance has long stood in the way of therapeutic advancement for lung cancer patients, the malignant tumor with the highest incidence and fatality rate in the world. Patients with lung adenocarcinoma (LUAD) now have a dismal prognosis due to the development of cisplatin (DDP) resistance, forcing them to use more costly second-line therapies. Therefore, overcoming resistance and enhancing patient outcomes can be achieved by comprehending the regulatory mechanisms of DDP resistance in LUAD. WD repeat domain 62 (WDR62) expression in LUAD tissues and in DDP-resistant or sensitive LUAD patients was analyzed bioinformatically, and a K-M plot was utilized to assess survival status. Real-time quantitative PCR was employed for WDR62 expression detection, cell-counting kit-8 assay for half maximal inhibitory concentration determination, flow cytometry for cell apoptosis detection, immunofluorescence for γ-H2AX expression analysis, and western blot for nonhomologous end joining repair and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway-related protein expression analysis. Poor prognosis was linked to WDR62, which was overexpressed in LUAD tissues and cells. Compared to sensitive cells, DDP-resistant cells had increased WDR62 expression. WDR62 knockdown may enhance DDP-induced cell apoptosis while reducing cell proliferation and DNA damage repair. Functional investigations verified that overexpressed WDR62's encouraging impact on DNA damage repair in A549/DDP cells could be reversed by MAPK inhibitors, increasing the cells' susceptibility to DDP. LUAD cells became less sensitive to DDP when WDR62 activated the MAPK/ERK pathway, which promoted DNA damage repair, indicating that DDP resistance might be reversed by treating LUAD with inhibitors of the MAPK pathway.

肺癌是世界上发病率和致死率最高的恶性肿瘤,长期以来,化疗耐药性一直阻碍着肺癌治疗的进步。由于顺铂(DDP)耐药性的发展,肺腺癌(LUAD)患者现在预后不佳,迫使他们使用更昂贵的二线治疗。因此,通过了解LUAD中DDP耐药的调控机制,可以克服耐药性并改善患者预后。从生物信息学上分析了WD重复结构域62 (WDR62)在LUAD组织和ddp耐药或敏感LUAD患者中的表达,并利用K-M图评估生存状况。采用实时荧光定量PCR检测WDR62表达,采用细胞计数试剂盒-8检测一半最大抑制浓度,流式细胞术检测细胞凋亡,免疫荧光检测γ-H2AX表达,western blot检测非同源末端连接修复和丝裂原活化蛋白激酶(MAPK)/细胞外信号调节激酶(ERK)通路相关蛋白表达。不良预后与WDR62有关,WDR62在LUAD组织和细胞中过表达。与敏感细胞相比,ddp耐药细胞的WDR62表达增加。WDR62敲低可增强ddp诱导的细胞凋亡,降低细胞增殖和DNA损伤修复。功能研究证实,在A549/DDP细胞中,过表达WDR62对DNA损伤修复的积极影响可以被MAPK抑制剂逆转,增加细胞对DDP的易感性。当WDR62激活MAPK/ERK通路时,LUAD细胞对DDP的敏感性降低,从而促进DNA损伤修复,这表明用MAPK通路抑制剂治疗LUAD可能逆转DDP抗性。
{"title":"WDR62 mediates MAPK/ERK pathway to stimulate DNA damage repair and attenuate cisplatin sensitivity in lung adenocarcinoma.","authors":"Xu Li, Yingwei Guo, Zecheng Qi, Yi Zheng","doi":"10.1097/CAD.0000000000001682","DOIUrl":"https://doi.org/10.1097/CAD.0000000000001682","url":null,"abstract":"<p><p>Chemotherapy resistance has long stood in the way of therapeutic advancement for lung cancer patients, the malignant tumor with the highest incidence and fatality rate in the world. Patients with lung adenocarcinoma (LUAD) now have a dismal prognosis due to the development of cisplatin (DDP) resistance, forcing them to use more costly second-line therapies. Therefore, overcoming resistance and enhancing patient outcomes can be achieved by comprehending the regulatory mechanisms of DDP resistance in LUAD. WD repeat domain 62 (WDR62) expression in LUAD tissues and in DDP-resistant or sensitive LUAD patients was analyzed bioinformatically, and a K-M plot was utilized to assess survival status. Real-time quantitative PCR was employed for WDR62 expression detection, cell-counting kit-8 assay for half maximal inhibitory concentration determination, flow cytometry for cell apoptosis detection, immunofluorescence for γ-H2AX expression analysis, and western blot for nonhomologous end joining repair and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway-related protein expression analysis. Poor prognosis was linked to WDR62, which was overexpressed in LUAD tissues and cells. Compared to sensitive cells, DDP-resistant cells had increased WDR62 expression. WDR62 knockdown may enhance DDP-induced cell apoptosis while reducing cell proliferation and DNA damage repair. Functional investigations verified that overexpressed WDR62's encouraging impact on DNA damage repair in A549/DDP cells could be reversed by MAPK inhibitors, increasing the cells' susceptibility to DDP. LUAD cells became less sensitive to DDP when WDR62 activated the MAPK/ERK pathway, which promoted DNA damage repair, indicating that DDP resistance might be reversed by treating LUAD with inhibitors of the MAPK pathway.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":""},"PeriodicalIF":1.8,"publicationDate":"2025-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142982540","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
KLF4 activates LATS2 to promote cisplatin sensitivity in ovarian cancer through DNA damage. KLF4 通过 DNA 损伤激活 LATS2,促进卵巢癌对顺铂的敏感性。
IF 1.8 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-01 Epub Date: 2024-09-27 DOI: 10.1097/CAD.0000000000001662
Ling Ma, Xiaoting Zhao, Xiang Lu, Jiahui Shen, Jiankang Huang

We aimed to investigate the role of large tumor suppressor kinase 2 (LATS2) in cisplatin (DDP) sensitivity in ovarian cancer. Bioinformatic analysis explored LATS2 expression, pathways, and regulators. Quantitative reverse transcription-PCR measured LATS2 and KLF4 mRNA levels. Dual-luciferase and chromatin immunoprecipitation assays confirmed their binding relationship. Cell viability, half maximal inhibitory concentration (IC 50 ) values, cell cycle, and DNA damage were assessed using CCK-8, flow cytometry, and comet assays. Western blot analyzed protein expression. The effect of LATS2 on the sensitivity of ovarian cancer to DDP was verified in vivo . LATS2 and KLF4 were downregulated in ovarian cancer, with LATS2 enriched in cell cycle, DNA replication, and mismatch repair pathways. KLF4, an upstream regulator of LATS2, bound to its promoter. Overexpressing LATS2 increased G1-phase cells, reduced cell viability and IC 50 values, and induced DNA damage. Silencing KLF4 alone showed the opposite effect on LATS2 overexpression. Knocking out LATS2 reversed the effects of KLF4 overexpression on cell viability, cell cycle, IC 50 values, and DNA damage in ovarian cancer cells. Inhibiting LATS2 inactivated the Hippo-YAP signaling pathway. In vivo experiments showed that overexpression of LATS2 enhanced the sensitivity of ovarian cancer to DDP. KLF4 activates LATS2 via DNA damage to enhance DDP sensitivity in ovarian cancer, providing a potential target for improving treatment outcomes.

我们旨在研究大肿瘤抑制激酶2(LATS2)在卵巢癌顺铂(DDP)敏感性中的作用。生物信息分析探讨了 LATS2 的表达、通路和调节因子。定量反转录-PCR测定了LATS2和KLF4的mRNA水平。双荧光素酶和染色质免疫沉淀试验证实了它们之间的结合关系。细胞活力、半数最大抑制浓度(IC50)值、细胞周期和DNA损伤通过CCK-8、流式细胞术和彗星试验进行了评估。Western 印迹分析了蛋白质的表达。在体内验证了 LATS2 对卵巢癌对 DDP 敏感性的影响。LATS2和KLF4在卵巢癌中下调,LATS2在细胞周期、DNA复制和错配修复通路中富集。KLF4是LATS2的上游调节因子,与LATS2的启动子结合。过表达 LATS2 会增加 G1 期细胞,降低细胞活力和 IC50 值,并诱导 DNA 损伤。单独抑制 KLF4 对 LATS2 的过度表达产生了相反的效果。敲除 LATS2 可逆转 KLF4 过表达对卵巢癌细胞活力、细胞周期、IC50 值和 DNA 损伤的影响。抑制 LATS2 可使 Hippo-YAP 信号通路失活。体内实验表明,过表达 LATS2 会增强卵巢癌对 DDP 的敏感性。KLF4通过DNA损伤激活LATS2,从而提高卵巢癌对DDP的敏感性,为改善治疗效果提供了一个潜在靶点。
{"title":"KLF4 activates LATS2 to promote cisplatin sensitivity in ovarian cancer through DNA damage.","authors":"Ling Ma, Xiaoting Zhao, Xiang Lu, Jiahui Shen, Jiankang Huang","doi":"10.1097/CAD.0000000000001662","DOIUrl":"10.1097/CAD.0000000000001662","url":null,"abstract":"<p><p>We aimed to investigate the role of large tumor suppressor kinase 2 (LATS2) in cisplatin (DDP) sensitivity in ovarian cancer. Bioinformatic analysis explored LATS2 expression, pathways, and regulators. Quantitative reverse transcription-PCR measured LATS2 and KLF4 mRNA levels. Dual-luciferase and chromatin immunoprecipitation assays confirmed their binding relationship. Cell viability, half maximal inhibitory concentration (IC 50 ) values, cell cycle, and DNA damage were assessed using CCK-8, flow cytometry, and comet assays. Western blot analyzed protein expression. The effect of LATS2 on the sensitivity of ovarian cancer to DDP was verified in vivo . LATS2 and KLF4 were downregulated in ovarian cancer, with LATS2 enriched in cell cycle, DNA replication, and mismatch repair pathways. KLF4, an upstream regulator of LATS2, bound to its promoter. Overexpressing LATS2 increased G1-phase cells, reduced cell viability and IC 50 values, and induced DNA damage. Silencing KLF4 alone showed the opposite effect on LATS2 overexpression. Knocking out LATS2 reversed the effects of KLF4 overexpression on cell viability, cell cycle, IC 50 values, and DNA damage in ovarian cancer cells. Inhibiting LATS2 inactivated the Hippo-YAP signaling pathway. In vivo experiments showed that overexpression of LATS2 enhanced the sensitivity of ovarian cancer to DDP. KLF4 activates LATS2 via DNA damage to enhance DDP sensitivity in ovarian cancer, providing a potential target for improving treatment outcomes.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":"49-61"},"PeriodicalIF":1.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142374909","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
SMAC mimetic BV6 acts in synergy with mTOR inhibitor to increase cisplatin sensitivity in ovarian cancer. SMAC 模拟物 BV6 与 mTOR 抑制剂协同作用,可提高卵巢癌患者对顺铂的敏感性。
IF 1.8 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-01 Epub Date: 2024-10-16 DOI: 10.1097/CAD.0000000000001664
Qi Chen, Hong Zhang

The objective of this study is to observe the antitumor efficacy of the second mitochondria-derived activator of caspases (SMAC) mimetic bivalent smac mimetic (BV6) in combination with target of rapamycin (mTOR) inhibitor on DDP (cisplatin) sensitivity. Ovarian cancer cells were exposed to cisplatin, BV6, DDP + BV6, and DDP + BV6 + mTOR inhibitor Rapamycin. Using proteomics and bioinformatics, protein expression profiles in ovarian cancer were determined. Bagg Albino color nude mice were treated with DDP or BV6 alone or in combination, or BV6 + DDP + Rapamycin. The effects of different treatments on ovarian cancer cells and tumor growth were evaluated in vivo and in vitro . Proteomics and bioinformatics analysis revealed significant changes of protein kinase (AKT)/mTOR pathway. Consistently, western blot data indicated that AKT/mTOR axis was gradually activated in BV6-treated ovarian cancer cells and attenuated the cytotoxic effect of BV6. Functional assays showed that DDP or BV6 treatment alone significantly enhanced the sensitivity and inhibited the migration of ovarian cancer cells, but without any synergistic effects. In addition, combination with BV6 and mTOR inhibitor Rapamycin significantly decreased cell viability and inhibited migration of ovarian cancer cells exposed to DDP. Consistently, the xenograft model showed that co-treatment with Rapamycin with BV6 had significantly suppressed tumor growth and metastasis. Our study demonstrated that SMAC analogue BV6 exhibits a strong anticancer effect on ovarian cancer in vitro and in vivo . Combination with Rapamycin overcomes the activation of mTOR pathway by BV6 and increases the chemosensitivity to DDP. These data suggest a potential application of triple combination with DDP + BV6 + Rapamycin in clinical management of ovarian cancer.

本研究的目的是观察线粒体衍生的第二种活化酶(SMAC)模拟物二价SMAC模拟物(BV6)与雷帕霉素靶点(mTOR)抑制剂联合使用对顺铂(DDP)敏感性的抗肿瘤效果。卵巢癌细胞暴露于顺铂、BV6、DDP + BV6 和 DDP + BV6 + mTOR 抑制剂雷帕霉素。利用蛋白质组学和生物信息学,确定了卵巢癌的蛋白质表达谱。对巴格白化色裸鼠单独或联合使用 DDP 或 BV6,或 BV6 + DDP + 雷帕霉素进行治疗。在体内和体外评估了不同治疗方法对卵巢癌细胞和肿瘤生长的影响。蛋白质组学和生物信息学分析表明,蛋白激酶(AKT)/mTOR通路发生了显著变化。Western印迹数据表明,AKT/mTOR轴在BV6处理的卵巢癌细胞中逐渐被激活,并减弱了BV6的细胞毒性作用。功能试验表明,单独使用 DDP 或 BV6 能显著提高卵巢癌细胞的敏感性并抑制其迁移,但没有任何协同作用。此外,与 BV6 和 mTOR 抑制剂雷帕霉素联用可明显降低暴露于 DDP 的卵巢癌细胞的存活率并抑制其迁移。同样,异种移植模型显示,雷帕霉素与 BV6 联合治疗可明显抑制肿瘤的生长和转移。我们的研究表明,SMAC 类似物 BV6 在体外和体内对卵巢癌都有很强的抗癌作用。与雷帕霉素联用可克服 BV6 对 mTOR 通路的激活,并增加对 DDP 的化疗敏感性。这些数据表明,DDP + BV6 + 雷帕霉素三联疗法有望应用于卵巢癌的临床治疗。
{"title":"SMAC mimetic BV6 acts in synergy with mTOR inhibitor to increase cisplatin sensitivity in ovarian cancer.","authors":"Qi Chen, Hong Zhang","doi":"10.1097/CAD.0000000000001664","DOIUrl":"10.1097/CAD.0000000000001664","url":null,"abstract":"<p><p>The objective of this study is to observe the antitumor efficacy of the second mitochondria-derived activator of caspases (SMAC) mimetic bivalent smac mimetic (BV6) in combination with target of rapamycin (mTOR) inhibitor on DDP (cisplatin) sensitivity. Ovarian cancer cells were exposed to cisplatin, BV6, DDP + BV6, and DDP + BV6 + mTOR inhibitor Rapamycin. Using proteomics and bioinformatics, protein expression profiles in ovarian cancer were determined. Bagg Albino color nude mice were treated with DDP or BV6 alone or in combination, or BV6 + DDP + Rapamycin. The effects of different treatments on ovarian cancer cells and tumor growth were evaluated in vivo and in vitro . Proteomics and bioinformatics analysis revealed significant changes of protein kinase (AKT)/mTOR pathway. Consistently, western blot data indicated that AKT/mTOR axis was gradually activated in BV6-treated ovarian cancer cells and attenuated the cytotoxic effect of BV6. Functional assays showed that DDP or BV6 treatment alone significantly enhanced the sensitivity and inhibited the migration of ovarian cancer cells, but without any synergistic effects. In addition, combination with BV6 and mTOR inhibitor Rapamycin significantly decreased cell viability and inhibited migration of ovarian cancer cells exposed to DDP. Consistently, the xenograft model showed that co-treatment with Rapamycin with BV6 had significantly suppressed tumor growth and metastasis. Our study demonstrated that SMAC analogue BV6 exhibits a strong anticancer effect on ovarian cancer in vitro and in vivo . Combination with Rapamycin overcomes the activation of mTOR pathway by BV6 and increases the chemosensitivity to DDP. These data suggest a potential application of triple combination with DDP + BV6 + Rapamycin in clinical management of ovarian cancer.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":" ","pages":"62-71"},"PeriodicalIF":1.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142456486","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
Screening of a kinase inhibitor library identified novel targetable kinase pathways in triple-negative breast cancer. 激酶抑制剂文库的筛选在三阴性乳腺癌中发现了新的靶向激酶途径。
IF 1.8 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-01 Epub Date: 2024-08-21 DOI: 10.1097/CAD.0000000000001658
Caroline H Rinderle, Christopher V Baker, Courtney B Lagarde, Khoa Nguyen, Sara Al-Ghadban, Margarite D Matossian, Van T Hoang, Elizabeth C Martin, Bridgette M Collins-Burow, Simak Ali, David H Drewry, Matthew E Burow, Bruce A Bunnell

Triple-negative breast cancer (TNBC) is a highly invasive breast cancer subtype that is challenging to treat due to inherent heterogeneity and absence of estrogen, progesterone, and human epidermal growth factor 2 receptors. Kinase signaling networks drive cancer growth and development, and kinase inhibitors are promising anti-cancer strategies in diverse cancer subtypes. Kinase inhibitor screens are an efficient, valuable means of identifying compounds that suppress cancer cell growth in vitro , facilitating the identification of kinase vulnerabilities to target therapeutically. The Kinase Chemogenomic Set is a well-annotated library of 187 kinase inhibitor compounds that indexes 215 kinases of the 518 in the known human kinome representing various kinase networks and signaling pathways, several of which are understudied. Our screen revealed 14 kinase inhibitor compounds effectively inhibited TNBC cell growth and proliferation. Upon further testing, three compounds, THZ531, THZ1, and PFE-PKIS 29, had the most significant and consistent effects across a range of TNBC cell lines. These cyclin-dependent kinase (CDK)12/CDK13, CDK7, and phosphoinositide 3-kinase inhibitors, respectively, decreased metabolic activity in TNBC cell lines and promote a gene expression profile consistent with the reversal of the epithelial-to-mesenchymal transition, indicating these kinase networks potentially mediate metastatic behavior. These data identified novel kinase targets and kinase signaling pathways that drive metastasis in TNBC.

三阴性乳腺癌(TNBC)是一种高度侵袭性的乳腺癌亚型,由于其固有的异质性和缺乏雌激素、黄体酮和人表皮生长因子2受体,治疗具有挑战性。激酶信号网络驱动癌症的生长和发展,激酶抑制剂在不同的癌症亚型中是有希望的抗癌策略。激酶抑制剂筛选是一种有效的、有价值的方法,可以在体外识别抑制癌细胞生长的化合物,促进激酶脆弱性的识别,从而进行靶向治疗。激酶化学基因组集是一个有良好注释的187种激酶抑制剂化合物文库,它索引了已知人类激酶组中518种激酶中的215种激酶,代表了各种激酶网络和信号通路,其中一些尚未得到充分研究。我们的筛选发现14种激酶抑制剂化合物有效地抑制TNBC细胞的生长和增殖。在进一步的测试中,三种化合物THZ531、THZ1和PFE-PKIS 29在一系列TNBC细胞系中具有最显著和一致的作用。这些细胞周期蛋白依赖性激酶(CDK)12/CDK13、CDK7和磷酸肌苷3-激酶抑制剂分别降低TNBC细胞系的代谢活性,并促进与上皮细胞向间质细胞转化逆转一致的基因表达谱,表明这些激酶网络可能介导转移行为。这些数据确定了驱动TNBC转移的新激酶靶点和激酶信号通路。
{"title":"Screening of a kinase inhibitor library identified novel targetable kinase pathways in triple-negative breast cancer.","authors":"Caroline H Rinderle, Christopher V Baker, Courtney B Lagarde, Khoa Nguyen, Sara Al-Ghadban, Margarite D Matossian, Van T Hoang, Elizabeth C Martin, Bridgette M Collins-Burow, Simak Ali, David H Drewry, Matthew E Burow, Bruce A Bunnell","doi":"10.1097/CAD.0000000000001658","DOIUrl":"10.1097/CAD.0000000000001658","url":null,"abstract":"<p><p>Triple-negative breast cancer (TNBC) is a highly invasive breast cancer subtype that is challenging to treat due to inherent heterogeneity and absence of estrogen, progesterone, and human epidermal growth factor 2 receptors. Kinase signaling networks drive cancer growth and development, and kinase inhibitors are promising anti-cancer strategies in diverse cancer subtypes. Kinase inhibitor screens are an efficient, valuable means of identifying compounds that suppress cancer cell growth in vitro , facilitating the identification of kinase vulnerabilities to target therapeutically. The Kinase Chemogenomic Set is a well-annotated library of 187 kinase inhibitor compounds that indexes 215 kinases of the 518 in the known human kinome representing various kinase networks and signaling pathways, several of which are understudied. Our screen revealed 14 kinase inhibitor compounds effectively inhibited TNBC cell growth and proliferation. Upon further testing, three compounds, THZ531, THZ1, and PFE-PKIS 29, had the most significant and consistent effects across a range of TNBC cell lines. These cyclin-dependent kinase (CDK)12/CDK13, CDK7, and phosphoinositide 3-kinase inhibitors, respectively, decreased metabolic activity in TNBC cell lines and promote a gene expression profile consistent with the reversal of the epithelial-to-mesenchymal transition, indicating these kinase networks potentially mediate metastatic behavior. These data identified novel kinase targets and kinase signaling pathways that drive metastasis in TNBC.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":"36 1","pages":"39-48"},"PeriodicalIF":1.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11634125/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142821722","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Combination of anlotinib and sintilimab for the treatment of recurrent or metastatic head and neck squamous cell carcinoma: a single-arm prospective study. anlotinib和sintilmab联合治疗复发或转移性头颈部鳞状细胞癌:单臂前瞻性研究
IF 1.8 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-01 Epub Date: 2024-12-11 DOI: 10.1097/CAD.0000000000001660
Tianxiao Wang, Jiaxin Wang, Yabing Zhang, Yuntao Song, Guohui Xu, Bin Zhang

To investigate whether blocking both programmed cell death protein and vascular endothelial growth factor receptor could offer superior anticancer activity in these patients without compromising safety. In this study, patients were administered oral anlotinib (12 mg/day) on days 1-14 and intravenous sintilimab (200 mg) on day 1 of a 3-weekly cycle. The primary endpoints included the objective response rate and disease control rate. The secondary endpoints included overall survival (OS) and safety. Ten eligible patients were enrolled between June 2019 and May 2022, and eight patients underwent radiographic assessments. The results showed an objective response rate of 50% (partial and complete response in four and zero patients, respectively) and a disease control rate of 100%; four patients demonstrated stable disease for at least 8 weeks. The median OS was 4.37 (in our study, the score was 7), and the OS rate at 12 months was 37.5%. The Kaplan-Meier survival curve showed that the group with high blood glucose levels had a significantly shorter duration of survival than those with normal blood glucose levels. Adverse events of grade 3 and higher occurred in 50% of patients, and the most common severe adverse events included tumor pain (50%), hypertension (37.5%), tumor hemorrhage (25%), and decreased appetite (25%). The combination of anlotinib and sintilimab showed promising efficacy in controlling tumor size. However, the disappointing OS rate suggests that anti-vascular endothelial growth factor receptor agents should be used cautiously after radical radiation therapy. The combination used in this study demonstrated a toxicity profile comparable to that of other agents used in this setting. These findings warrant further investigation into the potential clinical utility of this combination.

研究阻断程序性细胞死亡蛋白和血管内皮生长因子受体是否可以在不影响安全性的情况下为这些患者提供更好的抗癌活性。在这项研究中,患者在第1-14天口服anlotinib (12mg /天),在第1天静脉注射sintilmab (200mg),每个周期为3周。主要终点包括客观缓解率和疾病控制率。次要终点包括总生存期(OS)和安全性。2019年6月至2022年5月期间招募了10名符合条件的患者,其中8名患者接受了放射学评估。结果显示,客观缓解率为50%(4例患者部分缓解,0例患者完全缓解),疾病控制率为100%;4例患者病情稳定至少8周。中位OS为4.37(在我们的研究中,得分为7),12个月的OS率为37.5%。Kaplan-Meier生存曲线显示,高血糖组的生存时间明显短于正常血糖组。50%的患者发生3级及以上不良事件,最常见的严重不良事件包括肿瘤疼痛(50%)、高血压(37.5%)、肿瘤出血(25%)和食欲下降(25%)。安洛替尼联合辛替单抗对控制肿瘤大小有较好的疗效。然而,令人失望的OS率提示抗血管内皮生长因子受体药物在根治性放疗后应谨慎使用。本研究中使用的联合用药显示出与在此环境中使用的其他药物相当的毒性。这些发现为进一步研究这种联合疗法的潜在临床应用提供了依据。
{"title":"Combination of anlotinib and sintilimab for the treatment of recurrent or metastatic head and neck squamous cell carcinoma: a single-arm prospective study.","authors":"Tianxiao Wang, Jiaxin Wang, Yabing Zhang, Yuntao Song, Guohui Xu, Bin Zhang","doi":"10.1097/CAD.0000000000001660","DOIUrl":"10.1097/CAD.0000000000001660","url":null,"abstract":"<p><p>To investigate whether blocking both programmed cell death protein and vascular endothelial growth factor receptor could offer superior anticancer activity in these patients without compromising safety. In this study, patients were administered oral anlotinib (12 mg/day) on days 1-14 and intravenous sintilimab (200 mg) on day 1 of a 3-weekly cycle. The primary endpoints included the objective response rate and disease control rate. The secondary endpoints included overall survival (OS) and safety. Ten eligible patients were enrolled between June 2019 and May 2022, and eight patients underwent radiographic assessments. The results showed an objective response rate of 50% (partial and complete response in four and zero patients, respectively) and a disease control rate of 100%; four patients demonstrated stable disease for at least 8 weeks. The median OS was 4.37 (in our study, the score was 7), and the OS rate at 12 months was 37.5%. The Kaplan-Meier survival curve showed that the group with high blood glucose levels had a significantly shorter duration of survival than those with normal blood glucose levels. Adverse events of grade 3 and higher occurred in 50% of patients, and the most common severe adverse events included tumor pain (50%), hypertension (37.5%), tumor hemorrhage (25%), and decreased appetite (25%). The combination of anlotinib and sintilimab showed promising efficacy in controlling tumor size. However, the disappointing OS rate suggests that anti-vascular endothelial growth factor receptor agents should be used cautiously after radical radiation therapy. The combination used in this study demonstrated a toxicity profile comparable to that of other agents used in this setting. These findings warrant further investigation into the potential clinical utility of this combination.</p>","PeriodicalId":7969,"journal":{"name":"Anti-Cancer Drugs","volume":"36 1","pages":"79-84"},"PeriodicalIF":1.8,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11634082/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142821762","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Anti-Cancer Drugs
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