首页 > 最新文献

Current cancer drug targets最新文献

英文 中文
A Phase II Clinical Study on Apatinib Plus Vinorelbine in Refractory HER2-Negative Breast Cancer and its Metabolic Implications of Drug Resistance. 阿帕替尼联合长春瑞滨治疗难治性HER2阴性乳腺癌的II期临床研究及其耐药性对代谢的影响
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-10-01 DOI: 10.2174/0115680096303785240822155217
Jing Wu, Pan Deng, La Zou, Xiaoyu Liu, Xianjun Tang, Xiaohua Zeng, Shengchun Liu

Background: Apatinib, a tyrosine-kinase inhibitor that targets the vascular endothelial growth factor receptor 2, contributes to the inhibition of angiogenesis. Vinorelbine, a semisyn-thetic vinca alkaloid, primarily inhibits metaphase mitosis of cancer cells through its interactions with tubulin. This study aimed to evaluate whether apatinib combined with vinorelbine was ef-fective and safe for refractory human epidermal growth factor receptor 2 (HER2)-negative breast cancer patients who failed taxanes and/or anthracycline and analyze the possible mechanism of drug resistance through metabolomic analysis.

Methods: Eligible patients were HER2-negative, inoperable, locally advanced, or metastatic breast cancer patients who progressed after at least one chemotherapy regimen in this present prospective phase II study. Patients took oral apatinib (250-500 mg/day) plus intravenous infusion of vinorelbine (25 mg/m2 on day 1, day 8 at 3-week intervals). Objective response rate (ORR) was our primary endpoint, while disease control rate (DCR), overall survival (OS), progression-free survival (PFS), and toxicity were our secondary endpoints. The exploratory purpose was to identify biomarkers or drug resistance mechanisms through metabolomics changes before and after the combination therapy.

Results: Between September, 2019 and June, 2022, a total of 34 patients were included. ORR and DCR were 32.4% (11/34) and 85.3% (29/34), respectively. The median PFS was 5.0 months (95% CI, 3.766-6.234), while the median OS was 13.0 months (95% CI, 8.714-17.286). Side effects included hematologic toxicity, gastrointestinal reaction, and sinus tachycardia, which were mild to moderate. The mainly disturbed metabolic pathways were the cAMP signaling pathway, the alanine/aspartate/glutamate metabolism, the central carbon metabolism in cancer, the beta-alanine metabolism, the butanoate metabolism, and the glyoxylate and dicarboxylate metabolism, which may lead to the resistance of patients to this combination therapy.

Conclusion: Apatinib combined with vinorelbine is effective and safe in patients with locally advanced or metastatic refractory HER2-negative breast cancer. The findings of this study con-tribute to a better understanding of the metabolic effect of apatinib and vinorelbine therapy.

背景:阿帕替尼是一种靶向血管内皮生长因子受体2的酪氨酸激酶抑制剂,有助于抑制血管生成。长春瑞滨是一种半合成长春花生物碱,主要通过与微管蛋白的相互作用抑制癌细胞的有丝分裂。本研究旨在评估阿帕替尼联合长春瑞滨对紫杉类药物和/或蒽环类药物治疗失败的难治性人表皮生长因子受体2(HER2)阴性乳腺癌患者是否有效和安全,并通过代谢组学分析可能的耐药机制:符合条件的患者为HER2阴性、无法手术、局部晚期或转移性乳腺癌患者,这些患者在本前瞻性II期研究中至少接受过一次化疗后病情出现进展。患者口服阿帕替尼(250-500毫克/天),同时静脉输注长春瑞滨(25毫克/平方米,第1天和第8天各一次,间隔3周)。客观反应率(ORR)是我们的主要终点,疾病控制率(DCR)、总生存期(OS)、无进展生存期(PFS)和毒性是我们的次要终点。探索性目的是通过联合疗法前后代谢组学的变化来确定生物标志物或耐药机制:2019年9月至2022年6月,共纳入34例患者。ORR和DCR分别为32.4%(11/34)和85.3%(29/34)。中位PFS为5.0个月(95% CI,3.766-6.234),中位OS为13.0个月(95% CI,8.714-17.286)。副作用包括血液学毒性、胃肠道反应和窦性心动过速,均为轻度至中度。受干扰的代谢途径主要是cAMP信号通路、丙氨酸/天冬氨酸/谷氨酸代谢、癌症中枢碳代谢、β-丙氨酸代谢、丁酸代谢、乙醛酸和二羧酸代谢,这可能导致患者对该联合疗法产生耐药性:阿帕替尼联合长春瑞滨对局部晚期或转移性难治性HER2阴性乳腺癌患者有效且安全。本研究结果有助于更好地理解阿帕替尼和长春瑞滨治疗的代谢效应。
{"title":"A Phase II Clinical Study on Apatinib Plus Vinorelbine in Refractory HER2-Negative Breast Cancer and its Metabolic Implications of Drug Resistance.","authors":"Jing Wu, Pan Deng, La Zou, Xiaoyu Liu, Xianjun Tang, Xiaohua Zeng, Shengchun Liu","doi":"10.2174/0115680096303785240822155217","DOIUrl":"https://doi.org/10.2174/0115680096303785240822155217","url":null,"abstract":"<p><strong>Background: </strong>Apatinib, a tyrosine-kinase inhibitor that targets the vascular endothelial growth factor receptor 2, contributes to the inhibition of angiogenesis. Vinorelbine, a semisyn-thetic vinca alkaloid, primarily inhibits metaphase mitosis of cancer cells through its interactions with tubulin. This study aimed to evaluate whether apatinib combined with vinorelbine was ef-fective and safe for refractory human epidermal growth factor receptor 2 (HER2)-negative breast cancer patients who failed taxanes and/or anthracycline and analyze the possible mechanism of drug resistance through metabolomic analysis.</p><p><strong>Methods: </strong>Eligible patients were HER2-negative, inoperable, locally advanced, or metastatic breast cancer patients who progressed after at least one chemotherapy regimen in this present prospective phase II study. Patients took oral apatinib (250-500 mg/day) plus intravenous infusion of vinorelbine (25 mg/m2 on day 1, day 8 at 3-week intervals). Objective response rate (ORR) was our primary endpoint, while disease control rate (DCR), overall survival (OS), progression-free survival (PFS), and toxicity were our secondary endpoints. The exploratory purpose was to identify biomarkers or drug resistance mechanisms through metabolomics changes before and after the combination therapy.</p><p><strong>Results: </strong>Between September, 2019 and June, 2022, a total of 34 patients were included. ORR and DCR were 32.4% (11/34) and 85.3% (29/34), respectively. The median PFS was 5.0 months (95% CI, 3.766-6.234), while the median OS was 13.0 months (95% CI, 8.714-17.286). Side effects included hematologic toxicity, gastrointestinal reaction, and sinus tachycardia, which were mild to moderate. The mainly disturbed metabolic pathways were the cAMP signaling pathway, the alanine/aspartate/glutamate metabolism, the central carbon metabolism in cancer, the beta-alanine metabolism, the butanoate metabolism, and the glyoxylate and dicarboxylate metabolism, which may lead to the resistance of patients to this combination therapy.</p><p><strong>Conclusion: </strong>Apatinib combined with vinorelbine is effective and safe in patients with locally advanced or metastatic refractory HER2-negative breast cancer. The findings of this study con-tribute to a better understanding of the metabolic effect of apatinib and vinorelbine therapy.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142361279","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
MMP7, Regulated by c-Jun, is Involved in Oral Squamous Cell Carcinoma and Associated with Cancer-Related Fibroblasts Infiltration. 受c-Jun调控的MMP7参与口腔鳞状细胞癌并与癌相关成纤维细胞浸润有关
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-10-01 DOI: 10.2174/0115680096309161240821171847
Jian Wei, Xiaoxi Jiang, Yiwen Xu, Minhai Nie, Sen Yang, Xiao Chen, Lijuan Huang, Xuqian Liu

Objective: This study aimed to analyze the expression of Matrix Metalloproteinase 7 (MMP7) and molecular mechanism at the Transcription Factor (TF) level in Oral Squamous Cell Carcinoma (OSCC).

Methods: MMP7 expression was preliminarily explored in Head and Neck Squamous Cell Car-cinoma (HNSCC) in the online database, followed by functional analysis and prediction of TF of MMP7. IHC was employed to detect MMP7 levels in OSCC samples. SCC9 and 293T cells were used to explore the transcriptional and regulatory effects of predicted TF on MMP7 by reporter double luciferase assay, RT-qPCR, western blotting, and cellular immunofluorescence. Transwell and TUNEL were employed to detect the migration and apoptosis.

Results: MMP7 was significantly up-regulated in HNSCC and OSCC tissues. Moreover, MMP7 was positively correlated with CAFs and significantly enriched in the signaling pathway of RNA degradation. The c-Jun pathway was also up-regulated in OSCC tissues, and predicted to be optimal TF of MMP7 with positive regulatory relationship. In OSCC, silencing and over-expression of c-Jun significantly decreased and increased the level of MMP7. Meanwhile, c-Jun affected the behavior of SCC9 cells, which showed that after c-Jun gene silencing, the ability of cell migration was weakened, while apoptosis was enhanced. When c-Jun gene was overexpressed, the migration ability was enhanced, but apoptosis was not significantly affected.

Conclusion: MMP7 has been proven to be a key protein in the development of OSCC, and has the potential to become a biological marker and therapeutic target. It has been found that c-Jun could bind to the MMP7 promoter region, and the silencing or overexpression of c-Jun can positively regulate the expression of MMP7.

研究目的本研究旨在分析基质金属蛋白酶7(MMP7)在口腔鳞状细胞癌(OSCC)中的表达及转录因子(TF)水平的分子机制:方法:从在线数据库中初步探究头颈部鳞状细胞癌(HNSCC)中MMP7的表达情况,然后对MMP7的转录因子进行功能分析和预测。采用 IHC 检测 OSCC 样本中的 MMP7 水平。利用 SCC9 和 293T 细胞,通过双荧光素酶检测、RT-qPCR、Western 印迹和细胞免疫荧光,探讨预测的 TF 对 MMP7 的转录和调控作用。采用Transwell和TUNEL检测迁移和凋亡:结果:MMP7在HNSCC和OSCC组织中明显上调。此外,MMP7 与 CAFs 呈正相关,并在 RNA 降解信号通路中明显富集。在 OSCC 组织中,c-Jun 通路也被上调,并被预测为 MMP7 的最佳 TF,具有正向调控关系。在 OSCC 中,c-Jun 的沉默和过度表达会显著降低和提高 MMP7 的水平。同时,c-Jun对SCC9细胞的行为也有影响,沉默c-Jun基因后,细胞迁移能力减弱,凋亡能力增强。当c-Jun基因过表达时,细胞迁移能力增强,但细胞凋亡未受明显影响:结论:MMP7 已被证实是 OSCC 发病过程中的一个关键蛋白,并有可能成为一种生物学标志物和治疗靶点。研究发现,c-Jun能与MMP7启动子区域结合,沉默或过表达c-Jun能正向调节MMP7的表达。
{"title":"MMP7, Regulated by c-Jun, is Involved in Oral Squamous Cell Carcinoma and Associated with Cancer-Related Fibroblasts Infiltration.","authors":"Jian Wei, Xiaoxi Jiang, Yiwen Xu, Minhai Nie, Sen Yang, Xiao Chen, Lijuan Huang, Xuqian Liu","doi":"10.2174/0115680096309161240821171847","DOIUrl":"https://doi.org/10.2174/0115680096309161240821171847","url":null,"abstract":"<p><strong>Objective: </strong>This study aimed to analyze the expression of Matrix Metalloproteinase 7 (MMP7) and molecular mechanism at the Transcription Factor (TF) level in Oral Squamous Cell Carcinoma (OSCC).</p><p><strong>Methods: </strong>MMP7 expression was preliminarily explored in Head and Neck Squamous Cell Car-cinoma (HNSCC) in the online database, followed by functional analysis and prediction of TF of MMP7. IHC was employed to detect MMP7 levels in OSCC samples. SCC9 and 293T cells were used to explore the transcriptional and regulatory effects of predicted TF on MMP7 by reporter double luciferase assay, RT-qPCR, western blotting, and cellular immunofluorescence. Transwell and TUNEL were employed to detect the migration and apoptosis.</p><p><strong>Results: </strong>MMP7 was significantly up-regulated in HNSCC and OSCC tissues. Moreover, MMP7 was positively correlated with CAFs and significantly enriched in the signaling pathway of RNA degradation. The c-Jun pathway was also up-regulated in OSCC tissues, and predicted to be optimal TF of MMP7 with positive regulatory relationship. In OSCC, silencing and over-expression of c-Jun significantly decreased and increased the level of MMP7. Meanwhile, c-Jun affected the behavior of SCC9 cells, which showed that after c-Jun gene silencing, the ability of cell migration was weakened, while apoptosis was enhanced. When c-Jun gene was overexpressed, the migration ability was enhanced, but apoptosis was not significantly affected.</p><p><strong>Conclusion: </strong>MMP7 has been proven to be a key protein in the development of OSCC, and has the potential to become a biological marker and therapeutic target. It has been found that c-Jun could bind to the MMP7 promoter region, and the silencing or overexpression of c-Jun can positively regulate the expression of MMP7.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142361281","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
The Therapeutic Potential of Targeting Tumor Microenvironment and Modulation of Immunotherapy in Gastrointestinal Cancer. 胃肠癌中靶向肿瘤微环境和免疫疗法调节的治疗潜力。
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-09-30 DOI: 10.2174/0115680096319027240820055043
Saeideh Khorshid Sokhangouy, Hamid Jamialahmadi, Mahdieh Sadat Mortazavi Sani, Ghazaleh Khalili-Tanha, Arian Karimi Rouzbehani, Golnaz Mahmoudvand, Zahra Goudarzi, Arshia Fakouri, Simin Farokhi, Majid Khazaei, Seyed Mahdi Hassanian, Gordon A Ferns, Elham Nazari, Amir Avan

Immunotherapy, as a novel treatment approach for various disorders, including cancers, is designed to either stimulate or suppress the immune system with high speci-ficity. The recent achievements of this therapy in clinical trials are set to transform tradi-tional treatment methods. Furthermore, it holds promise for enhancing the survival rates of patients suffering from both metastatic cancers and primary stages. Gastrointestinal Cancers (GI) account for 26% of global incidence and 35% of worldwide deaths. Treat-ment can be carried out using targeted immunotherapy in these cancers. If the tiers are superior, improvement could require more enterprise. On account that the function of immunotherapy in GI has been so promising, solely in sufferers with severe metastatic levels, within the literature, the immune checkpoint inhibitors in cancer immunotherapy of GI cancers, chimeric antigen receptor T-cell (vehicle-T), modulators of the tumor mi-croenvironment, and drug resistance mechanisms in immunotherapy as an effective treatment approach to GI cancers along with colon, pancreas, gastric, and esophageal cancers have been addressed. This review provides an overview of FDA-approved im-munotherapy drugs and ongoing preclinical developments. Additionally, we offer in-sights into the future of immunotherapy for GI cancer patients, addressing the associated challenges.

免疫疗法是一种治疗包括癌症在内的各种疾病的新方法,其目的是高度特异性地刺激或抑制免疫系统。这种疗法最近在临床试验中取得的成就将改变传统的治疗方法。此外,它还有望提高转移性癌症和原发性癌症患者的生存率。胃肠道癌症(GI)占全球发病率的 26%,占全球死亡人数的 35%。对这些癌症可以采用靶向免疫疗法进行治疗。如果层级较高,改善可能需要更多的企业。鉴于免疫疗法在消化道癌症中的作用前景广阔,但仅在转移程度严重的患者中使用,因此在文献中论述了免疫检查点抑制剂在消化道癌症免疫疗法中的应用、嵌合抗原受体T细胞(carrier-T)、肿瘤mi-croenvironment调节剂以及免疫疗法中的耐药机制,以此作为结肠癌、胰腺癌、胃癌和食管癌等消化道癌症的有效治疗方法。本综述概述了 FDA 批准的免疫疗法药物和正在进行的临床前研究进展。此外,我们还将展望消化道癌症患者免疫疗法的未来,并探讨相关的挑战。
{"title":"The Therapeutic Potential of Targeting Tumor Microenvironment and Modulation of Immunotherapy in Gastrointestinal Cancer.","authors":"Saeideh Khorshid Sokhangouy, Hamid Jamialahmadi, Mahdieh Sadat Mortazavi Sani, Ghazaleh Khalili-Tanha, Arian Karimi Rouzbehani, Golnaz Mahmoudvand, Zahra Goudarzi, Arshia Fakouri, Simin Farokhi, Majid Khazaei, Seyed Mahdi Hassanian, Gordon A Ferns, Elham Nazari, Amir Avan","doi":"10.2174/0115680096319027240820055043","DOIUrl":"https://doi.org/10.2174/0115680096319027240820055043","url":null,"abstract":"<p><p>Immunotherapy, as a novel treatment approach for various disorders, including cancers, is designed to either stimulate or suppress the immune system with high speci-ficity. The recent achievements of this therapy in clinical trials are set to transform tradi-tional treatment methods. Furthermore, it holds promise for enhancing the survival rates of patients suffering from both metastatic cancers and primary stages. Gastrointestinal Cancers (GI) account for 26% of global incidence and 35% of worldwide deaths. Treat-ment can be carried out using targeted immunotherapy in these cancers. If the tiers are superior, improvement could require more enterprise. On account that the function of immunotherapy in GI has been so promising, solely in sufferers with severe metastatic levels, within the literature, the immune checkpoint inhibitors in cancer immunotherapy of GI cancers, chimeric antigen receptor T-cell (vehicle-T), modulators of the tumor mi-croenvironment, and drug resistance mechanisms in immunotherapy as an effective treatment approach to GI cancers along with colon, pancreas, gastric, and esophageal cancers have been addressed. This review provides an overview of FDA-approved im-munotherapy drugs and ongoing preclinical developments. Additionally, we offer in-sights into the future of immunotherapy for GI cancer patients, addressing the associated challenges.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142343166","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting MGST1 Makes Non-Small Cell Lung Cancer Cells Sensitive to Radiotherapy by Epigenetically Enhancing ALOX15-Mediated Ferroptosis. 靶向 MGST1 通过表观遗传学增强 ALOX15 介导的铁突变,使非小细胞肺癌细胞对放疗敏感
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-09-27 DOI: 10.2174/0115680096317925240820053934
Yechen Ma, Yuping Peng, Shulin Cheng, Long Jin

Background: Ferroptosis is closely related to radiotherapy resistance in multiple can-cers. Herein, the role of microsomal glutathione S-transferase 1 (MGST1) in regulating ferropto-sis and radiotherapy resistance in non-small cell lung cancer (NSCLC) was investigated.

Methods: Radiation-resistant NSCLC cells (NCI-1299-IR and HCC827-IR cells) were estab-lished. After exposure to X-ray, cell proliferation and survival were assessed by colony formation assay and CCK-8 assay, and lipid ROS level was examined by the fluorophore BODIPY™ 581/591 C11. MDA, GSH, and Fe2+ levels were measured by ELISA kits. The molecular interac-tion was analyzed using ChIP and MSP assays.

Results: Our results showed that RSL3 treatment greatly enhanced the radiotherapy sensitivity of NCI-1299-IR and HCC827-IR cells. It was subsequently revealed that MGST1 was highly ex-pressed in NCI-1299-IR and HCC827-IR cells than its parent cells, and silencing of MGST1 re-duced radioresistance of NCI-1299-IR and HCC827-IR cells by facilitating ferroptosis. Mechanis-tically, MGST1 knockdown greatly reduced HO-1 and DNMT1/3A protein levels, leading to re-duced DNA methylation on the ALOX15 promoter region, thereby epigenetically upregulating ALOX15 expression. As expected, the promoting effects of MGST1 silencing on radiosensitivity and ferroptosis in radiation-resistant NSCLC cells were strikingly eliminated by ALOX15 knock-down.

Conclusion: MGST1 knockdown epigenetically enhanced radiotherapy sensitivity of NCSLC cells by promoting ALOX15-mediated ferroptosis through regulating the HO-1/DNMT1 pathway.

背景:铁蛋白沉积与多种癌症的放疗耐药性密切相关。本文研究了微粒体谷胱甘肽 S 转移酶 1(MGST1)在非小细胞肺癌(NSCLC)中调控铁蛋白沉积和放疗耐药性的作用:方法:建立抗放射治疗的NSCLC细胞(NCI-1299-IR和HCC827-IR细胞)。暴露于 X 射线后,细胞增殖和存活通过集落形成试验和 CCK-8 试验进行评估,脂质 ROS 水平通过荧光团 BODIPY™ 581/591 C11 进行检测。MDA、GSH 和 Fe2+ 含量通过 ELISA 试剂盒进行测定。使用 ChIP 和 MSP 检测分析了分子间的相互作用:结果:我们的研究结果表明,RSL3能大大提高NCI-1299-IR和HCC827-IR细胞的放疗敏感性。随后的研究发现,MGST1在NCI-1299-IR和HCC827-IR细胞中的外显率高于其母细胞,而沉默MGST1可通过促进铁凋亡重新降低NCI-1299-IR和HCC827-IR细胞的放射抗性。从机理上讲,MGST1的敲除大大降低了HO-1和DNMT1/3A蛋白的水平,导致ALOX15启动子区域的DNA甲基化重新降低,从而在表观遗传学上上调了ALOX15的表达。正如预期的那样,MGST1沉默对耐辐射NSCLC细胞放射敏感性和铁变态反应的促进作用被ALOX15敲除所显著消除:结论:MGST1基因敲除通过调控HO-1/DNMT1通路促进ALOX15介导的铁蜕变,从而从表观遗传学角度提高了NSCLC细胞的放疗敏感性。
{"title":"Targeting MGST1 Makes Non-Small Cell Lung Cancer Cells Sensitive to Radiotherapy by Epigenetically Enhancing ALOX15-Mediated Ferroptosis.","authors":"Yechen Ma, Yuping Peng, Shulin Cheng, Long Jin","doi":"10.2174/0115680096317925240820053934","DOIUrl":"https://doi.org/10.2174/0115680096317925240820053934","url":null,"abstract":"<p><strong>Background: </strong>Ferroptosis is closely related to radiotherapy resistance in multiple can-cers. Herein, the role of microsomal glutathione S-transferase 1 (MGST1) in regulating ferropto-sis and radiotherapy resistance in non-small cell lung cancer (NSCLC) was investigated.</p><p><strong>Methods: </strong>Radiation-resistant NSCLC cells (NCI-1299-IR and HCC827-IR cells) were estab-lished. After exposure to X-ray, cell proliferation and survival were assessed by colony formation assay and CCK-8 assay, and lipid ROS level was examined by the fluorophore BODIPY™ 581/591 C11. MDA, GSH, and Fe2+ levels were measured by ELISA kits. The molecular interac-tion was analyzed using ChIP and MSP assays.</p><p><strong>Results: </strong>Our results showed that RSL3 treatment greatly enhanced the radiotherapy sensitivity of NCI-1299-IR and HCC827-IR cells. It was subsequently revealed that MGST1 was highly ex-pressed in NCI-1299-IR and HCC827-IR cells than its parent cells, and silencing of MGST1 re-duced radioresistance of NCI-1299-IR and HCC827-IR cells by facilitating ferroptosis. Mechanis-tically, MGST1 knockdown greatly reduced HO-1 and DNMT1/3A protein levels, leading to re-duced DNA methylation on the ALOX15 promoter region, thereby epigenetically upregulating ALOX15 expression. As expected, the promoting effects of MGST1 silencing on radiosensitivity and ferroptosis in radiation-resistant NSCLC cells were strikingly eliminated by ALOX15 knock-down.</p><p><strong>Conclusion: </strong>MGST1 knockdown epigenetically enhanced radiotherapy sensitivity of NCSLC cells by promoting ALOX15-mediated ferroptosis through regulating the HO-1/DNMT1 pathway.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142343165","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
Phase I Clinical Trial of CD19 CAR-T Cells Expressing CXCR5 Protein for the Treatment of Relapsed or Refractory B-cell Lymphoma. 表达 CXCR5 蛋白的 CD19 CAR-T 细胞治疗复发性或难治性 B 细胞淋巴瘤的 I 期临床试验。
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-09-27 DOI: 10.2174/0115680096304530240816111936
Jiaxi Wang, Yirong Jiang, Min Luo, Wenyi Lu, Jixiang He, Meng Zhang, Zhuoxin Yao, Xin Jin, Xia Xiao, Jianhang Chen, Guangchao Li, Wen Ding, Jie Zhou, Zhiyin Zhang, Mingfeng Zhao

Background: It is difficult for CD19 CAR-T cells to enter solid tumors, which is one reason for their poor efficacy in lymphoma treatment. The chemokine CXCL13 secreted by stro-mal cells of the lymph nodes induces the homing of B and T lymphocytes, which express its receptor CXCR5. Preclinical trials have shown that the expression of CXCR5 on CD19 CAR-T cells can increase their migration to the tumor microenvironment and enhance their antitumor function.

Methods: We engineered the CD19 CAR-T cells to express a second receptor, CXCR5. Then, we conducted a phase I clinical trial to evaluate the safety and efficacy of CXCR5 CD19 CAR-T cells in the treatment of relapsed or refractory (R/R) B-cell lymphoma.

Results: We recruited 10 patients with R/R B-cell lymphoma undergoing CXCR5 CD19 CAR-T cell therapy. The objective response rate was 80%, and the complete response rate was 50%. The median follow-up time was 15.48 months (3.4-22.3 months), and the median Progression-Free Survival (PFS) time was 8.15 months (1.5-22.33 months). One patient received ASCT at 1.5 months (at PR) after infusion of CAR-T cells. The incidence of grade 1 and grade 2 Cytokine Release Syndrome (CRS) was 70% and 20%, respectively. No patient experienced grade 3 or higher levels of CRS, neurotoxicity, or infusion-related dose toxicity.

Conclusion: The results obtained in this study suggest that CXCR5 CD19 CAR-T cells should be investigated in a trial with broader patient populations.

Trial registration: The trials were registered at www.chictr.org.cn as ChiCTR2100052677 and ChiCTR1900028692.

背景:CD19 CAR-T 细胞很难进入实体瘤,这也是其治疗淋巴瘤疗效不佳的原因之一。淋巴结中胚层细胞分泌的趋化因子 CXCL13 可诱导 B 淋巴细胞和 T 淋巴细胞归巢,而 B 淋巴细胞和 T 淋巴细胞均表达其受体 CXCR5。临床前试验表明,CD19 CAR-T细胞表达CXCR5可增加其向肿瘤微环境的迁移,并增强其抗肿瘤功能:方法:我们设计了表达第二种受体CXCR5的CD19 CAR-T细胞。然后,我们进行了一项I期临床试验,评估CXCR5 CD19 CAR-T细胞治疗复发或难治性(R/R)B细胞淋巴瘤的安全性和有效性:我们招募了10名接受CXCR5 CD19 CAR-T细胞治疗的R/R B细胞淋巴瘤患者。客观反应率为80%,完全反应率为50%。中位随访时间为15.48个月(3.4-22.3个月),中位无进展生存期(PFS)为8.15个月(1.5-22.33个月)。一名患者在输注 CAR-T 细胞后 1.5 个月(PR 时)接受了 ASCT。1级和2级细胞因子释放综合征(CRS)的发生率分别为70%和20%。没有患者出现3级或3级以上的CRS、神经毒性或输注相关剂量毒性:结论:本研究的结果表明,CXCR5 CD19 CAR-T细胞应在更广泛的患者群体中进行试验研究:试验注册于 www.chictr.org.cn,注册号分别为 ChiCTR2100052677 和 ChiCTR1900028692。
{"title":"Phase I Clinical Trial of CD19 CAR-T Cells Expressing CXCR5 Protein for the Treatment of Relapsed or Refractory B-cell Lymphoma.","authors":"Jiaxi Wang, Yirong Jiang, Min Luo, Wenyi Lu, Jixiang He, Meng Zhang, Zhuoxin Yao, Xin Jin, Xia Xiao, Jianhang Chen, Guangchao Li, Wen Ding, Jie Zhou, Zhiyin Zhang, Mingfeng Zhao","doi":"10.2174/0115680096304530240816111936","DOIUrl":"https://doi.org/10.2174/0115680096304530240816111936","url":null,"abstract":"<p><strong>Background: </strong>It is difficult for CD19 CAR-T cells to enter solid tumors, which is one reason for their poor efficacy in lymphoma treatment. The chemokine CXCL13 secreted by stro-mal cells of the lymph nodes induces the homing of B and T lymphocytes, which express its receptor CXCR5. Preclinical trials have shown that the expression of CXCR5 on CD19 CAR-T cells can increase their migration to the tumor microenvironment and enhance their antitumor function.</p><p><strong>Methods: </strong>We engineered the CD19 CAR-T cells to express a second receptor, CXCR5. Then, we conducted a phase I clinical trial to evaluate the safety and efficacy of CXCR5 CD19 CAR-T cells in the treatment of relapsed or refractory (R/R) B-cell lymphoma.</p><p><strong>Results: </strong>We recruited 10 patients with R/R B-cell lymphoma undergoing CXCR5 CD19 CAR-T cell therapy. The objective response rate was 80%, and the complete response rate was 50%. The median follow-up time was 15.48 months (3.4-22.3 months), and the median Progression-Free Survival (PFS) time was 8.15 months (1.5-22.33 months). One patient received ASCT at 1.5 months (at PR) after infusion of CAR-T cells. The incidence of grade 1 and grade 2 Cytokine Release Syndrome (CRS) was 70% and 20%, respectively. No patient experienced grade 3 or higher levels of CRS, neurotoxicity, or infusion-related dose toxicity.</p><p><strong>Conclusion: </strong>The results obtained in this study suggest that CXCR5 CD19 CAR-T cells should be investigated in a trial with broader patient populations.</p><p><strong>Trial registration: </strong>The trials were registered at www.chictr.org.cn as ChiCTR2100052677 and ChiCTR1900028692.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142343164","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
Disulfiram-Copper Potentiates Anticancer Efficacy of Standard Chemo-therapy Drugs in Bladder Cancer Animal Model through ROS-Autophagy-Ferroptosis Signalling Cascade. 双硫仑-铜通过 ROS-Autophagy-Ferroptosis 信号级联增强标准化疗药物在膀胱癌动物模型中的抗癌效果
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-09-25 DOI: 10.2174/0115680096325879240815105227
Neeru Sharma, Sumit Dey, Sheetal Singh, Sandeep Kumar, Monidipa Konar, Priyanka Naithani, Ritika Panwar, Pulkit Rastogi, Ashish Kakkar, Ravimohan S Mavuduru, Smita Pattanaik
<p><strong>Background: </strong>Cost-effective management of Urinary Bladder Cancer (UBC) is an unmet need.</p><p><strong>Aims: </strong>Our study aims to demonstrate the efficacy of a drug repurposing strategy by using disulfiram (DSF) and copper gluconate (Cu) as an add-on treatment combination to traditional GC-based chemother-apy against N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN)-induced UBC mice (C57J) model.</p><p><strong>Methods: </strong>Male C57BL/6J mice were given 0.05% BBN in drinking water ad libitum, and tumour for-mation was verified by histological and physical evaluation. Animals were subsequently divided into eight groups and received treatment with different drug combinations. Control animals received only ve-hicle (DMSO). At the end of the treatment schedule, the bladder tumour was excised and further used to check the expression (mRNA and protein) of ALDH1 isoenzymes using qRT-PCR, western blot, and IHC methods. Autophagy induction was assessed by quantifying the expression of LC3B and SQSTM1/p62 proteins through IHC. Biochemical analysis of superoxide dismutase (SOD), reduced glutathione (GSH), and lipid peroxidation levels in the freshly isolated tumours was performed to check the alterations in the antioxidant system caused by combination treatment.</p><p><strong>Results: </strong>We observed significant induction of an invasive form of bladder cancer in the mice after nine-teen weeks of BBN exposure. The animals began exhibiting early indications of inflammatory alterations as early as the sixth week following BBN treatment. Furthermore, the wet bladder weight and overall tu-mour burden were significantly decreased (p< 0.0001) by DSF-Cu co-treatment in addition to the GC-based chemotherapy. Real-time PCR analysis revealed that treatment with disulfiram and copper glu-conate significantly decreased (p<0.0001) the mRNA expression of ALDH1 isoenzymes. Comparing the triple drug combination group (GC+DSF-Cu) to the untreated mice, a significant rise in LC3B puncta (p<0.0001) and a decrease in P62/SQSTM1 (p=0.0002) were noted, indicating the induction of autophagy flux in the add-on group. When GC+DSF-Cu treated mice were compared to the untreated tumour group, a substantial decrease in ALDH1/2 protein expression was observed (p= 0.0029 in IHC and p<0.0001 in western blot). Lipid peroxidation was significantly higher (p<0.0001) in the triple drug combination group than in untreated mice. There was a simultaneous decrease in reduced glutathione (GSH) and en-zyme superoxide dismutase (SOD) levels (p<0.0001), which strongly suggests the generation of reactive oxygen species and induction of ferroptotic cell death in the add-on therapy group. Additionally, in both IHC and western blot assays, ALDH1A3 expression was found to be significantly increased (p=0.0033, <0.0001 respectively) in GC+DSF-Cu treated mice relative to the untreated group, suggesting a potential connection between the ferroptosis path
背景:具有成本效益的膀胱癌(UBC)治疗是一项尚未满足的需求:目的:我们的研究旨在证明一种药物再利用策略的疗效,即使用双硫仑(DSF)和葡萄糖酸铜(Cu)作为传统的基于GC的化疗方案的附加治疗组合,来对抗N-丁基-N-(4-羟基丁基)亚硝胺(BBN)诱导的UBC小鼠(C57J)模型:雄性 C57BL/6J 小鼠自由饮用含 0.05% BBN 的饮用水,并通过组织学和物理评估验证肿瘤的形成。动物随后被分为八组,接受不同药物组合的治疗。对照组动物只接受静脉注射(DMSO)。治疗结束后,切除膀胱肿瘤,并进一步使用 qRT-PCR、Western 印迹和 IHC 方法检测 ALDH1 同工酶的表达(mRNA 和蛋白质)。自噬诱导是通过 IHC 定量 LC3B 和 SQSTM1/p62 蛋白的表达来评估的。对新鲜分离的肿瘤中的超氧化物歧化酶(SOD)、还原型谷胱甘肽(GSH)和脂质过氧化水平进行了生化分析,以检查联合治疗对抗氧化系统造成的改变:结果:我们观察到,小鼠在接触 BBN 9-14 周后,浸润性膀胱癌的诱导作用明显。早在 BBN 治疗后的第六周,动物就开始出现炎症改变的早期迹象。此外,除了基于 GC 的化疗外,DSF-Cu 联合治疗也显著降低了湿膀胱重量和膀胱癌的总体负担(p< 0.0001)。实时 PCR 分析显示,双硫仑和葡糖酸铜治疗可显著降低(p<0.0001)ALDH1 同工酶的 mRNA 表达。三药联合组(GC+DSF-Cu)与未处理小鼠相比,LC3B点位明显上升(p<0.0001),P62/SQSTM1下降(p=0.0002),表明加药组诱导了自噬通量。当 GC+DSF-Cu 治疗组小鼠与未治疗肿瘤组相比时,观察到 ALDH1/2 蛋白表达大幅下降(IHC 中 p= 0.0029,Western 印迹中 p<0.0001)。三药联合组小鼠的脂质过氧化反应明显高于未处理组小鼠(p<0.0001)。还原型谷胱甘肽(GSH)和超氧化物歧化酶(SOD)水平同时下降(p<0.0001),这有力地表明了附加疗法组中活性氧的生成和诱导铁变态反应细胞死亡。此外,在IHC和Western印迹检测中发现,GC+DSF-Cu治疗组小鼠的ALDH1A3表达相对于未治疗组显著增加(分别为p=0.0033和<0.0001),这表明铁变态反应途径与ALDH1A3过表达之间存在潜在联系:结论:研究发现,双硫仑加铜治疗可通过诱导铁氧化还原反应抑制膀胱肿瘤的生长,并进一步激活自噬过程。我们的研究结果证明,DSF-铜可作为标准化疗药物的有效辅助疗法,用于治疗膀胱癌。
{"title":"Disulfiram-Copper Potentiates Anticancer Efficacy of Standard Chemo-therapy Drugs in Bladder Cancer Animal Model through ROS-Autophagy-Ferroptosis Signalling Cascade.","authors":"Neeru Sharma, Sumit Dey, Sheetal Singh, Sandeep Kumar, Monidipa Konar, Priyanka Naithani, Ritika Panwar, Pulkit Rastogi, Ashish Kakkar, Ravimohan S Mavuduru, Smita Pattanaik","doi":"10.2174/0115680096325879240815105227","DOIUrl":"https://doi.org/10.2174/0115680096325879240815105227","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Cost-effective management of Urinary Bladder Cancer (UBC) is an unmet need.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Aims: &lt;/strong&gt;Our study aims to demonstrate the efficacy of a drug repurposing strategy by using disulfiram (DSF) and copper gluconate (Cu) as an add-on treatment combination to traditional GC-based chemother-apy against N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN)-induced UBC mice (C57J) model.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;Male C57BL/6J mice were given 0.05% BBN in drinking water ad libitum, and tumour for-mation was verified by histological and physical evaluation. Animals were subsequently divided into eight groups and received treatment with different drug combinations. Control animals received only ve-hicle (DMSO). At the end of the treatment schedule, the bladder tumour was excised and further used to check the expression (mRNA and protein) of ALDH1 isoenzymes using qRT-PCR, western blot, and IHC methods. Autophagy induction was assessed by quantifying the expression of LC3B and SQSTM1/p62 proteins through IHC. Biochemical analysis of superoxide dismutase (SOD), reduced glutathione (GSH), and lipid peroxidation levels in the freshly isolated tumours was performed to check the alterations in the antioxidant system caused by combination treatment.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;We observed significant induction of an invasive form of bladder cancer in the mice after nine-teen weeks of BBN exposure. The animals began exhibiting early indications of inflammatory alterations as early as the sixth week following BBN treatment. Furthermore, the wet bladder weight and overall tu-mour burden were significantly decreased (p&#60; 0.0001) by DSF-Cu co-treatment in addition to the GC-based chemotherapy. Real-time PCR analysis revealed that treatment with disulfiram and copper glu-conate significantly decreased (p&#60;0.0001) the mRNA expression of ALDH1 isoenzymes. Comparing the triple drug combination group (GC+DSF-Cu) to the untreated mice, a significant rise in LC3B puncta (p&#60;0.0001) and a decrease in P62/SQSTM1 (p=0.0002) were noted, indicating the induction of autophagy flux in the add-on group. When GC+DSF-Cu treated mice were compared to the untreated tumour group, a substantial decrease in ALDH1/2 protein expression was observed (p= 0.0029 in IHC and p&#60;0.0001 in western blot). Lipid peroxidation was significantly higher (p&#60;0.0001) in the triple drug combination group than in untreated mice. There was a simultaneous decrease in reduced glutathione (GSH) and en-zyme superoxide dismutase (SOD) levels (p&#60;0.0001), which strongly suggests the generation of reactive oxygen species and induction of ferroptotic cell death in the add-on therapy group. Additionally, in both IHC and western blot assays, ALDH1A3 expression was found to be significantly increased (p=0.0033, &#60;0.0001 respectively) in GC+DSF-Cu treated mice relative to the untreated group, suggesting a potential connection between the ferroptosis path","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142343163","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
Deoxypodophyllotoxin Mediates Autophagy Death through Inhibition of GRP78 in Human Osteosarcoma. 脱氧鬼臼毒素通过抑制人骨肉瘤中的GRP78介导自噬死亡
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-09-23 DOI: 10.2174/0115680096312622240812093046
Xiao-Jun Tang, Ling-Li Luo, Wen-Chao Zhou, Gang-Qing Shi, Xiao-Xu Wang, Tian Zeng, Xiong-Jin Tan, Wei-Ming Guo, An-Bo Gao, Yukun Li, Juan Zou

Background: Glucose-regulated protein 78 (GRP78), as a chaperone protein, can protect the endoplasmic reticulum of cells and is expressed to influence chemoresistance and prognosis in cancer. Deoxypodophyllotoxin (DPT) is a compound with antitumor effects on cancers. DPT inhibits the proliferation of osteosarcoma by inducing apoptosis, necrosis, or cell cycle arrest.

Object: This study was performed to demonstrate the molecular mechanism by which DPT attenuates osteosarcoma progression through GRP78.

Methods: Natural compound libraries and western blot (WB) were used to screen the inhibitors of osteosarcoma GRP78. The expression of mitochondria-related genes in cancer cells of the treatment group was detected by quantitative real-time PCR (qPCR) and WB. 3-(4,5)- Dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT) and 5-ethynyl-2'- deoxyuridine (EDU) were used to discover the activity and proliferation of osteosarcoma cells treated with DPT. We constructed an in vivo mouse model of DPT drug therapy and carried out immunohistochemical detection of xenografts. The treated osteosarcoma cells were analyzed using bioinformatics and electron microscopy. The data were analyzed finally.

Results: DPT inhibited osteosarcoma cell survival and the growth of tumor xenografts. It promoted up-regulation of BCL2-associated X protein (Bax) and B-cell CLL/lymphoma 2 (Bcl-2), which serves to mediate and attenuate, respectively, the killing activities of DPT through mitochondria dysfunction. The effect of DPT against cancer cells could be attenuated by the overexpression of GRP78, characterized by the inactivation of the caspase cascade. The loss of GRP78 in osteosarcoma cells negatively mediated the basal level of autophagyassociated genes. DPT stimulated autophagy via the phosphoinositide 3-kinase (PI3K)-v-akt murine thymoma viral oncogene homolog (AKT), a mechanistic target of rapamycin (mTOR) axis. The autophagy caused by DPT played an active role in the osteosarcoma of humans and blocked the apoptotic cascade.

Conclusion: Combination treatment with the GRP78 inhibitor DPT and pharmacological autophagy inhibitors will be a meaningful method of obviating osteosarcoma cells.

背景:葡萄糖调节蛋白78(GRP78)是一种伴侣蛋白,可以保护细胞的内质网,其表达可影响癌症的化疗抗性和预后。脱氧鬼臼毒素(DPT)是一种对癌症具有抗肿瘤作用的化合物。DPT 通过诱导细胞凋亡、坏死或细胞周期停滞来抑制骨肉瘤的增殖:本研究旨在证明 DPT 通过 GRP78 抑制骨肉瘤进展的分子机制:方法:采用天然化合物库和免疫印迹(WB)筛选骨肉瘤 GRP78 抑制剂。通过实时定量 PCR(qPCR)和 WB 检测治疗组癌细胞中线粒体相关基因的表达。用 3-(4,5)- Dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT) 和 5-ethynyl-2'- deoxyuridine (EDU) 来检测经 DPT 处理的骨肉瘤细胞的活性和增殖情况。我们构建了 DPT 药物治疗小鼠体内模型,并对异种移植进行了免疫组化检测。利用生物信息学和电子显微镜对处理过的骨肉瘤细胞进行分析。结果结果:DPT抑制了骨肉瘤细胞的存活和肿瘤异种移植的生长。它促进了BCL2相关X蛋白(Bax)和B细胞CLL/淋巴瘤2(Bcl-2)的上调,分别通过线粒体功能障碍介导和削弱了DPT的杀伤活性。DPT对癌细胞的作用可通过过量表达GRP78而减弱,GRP78的特点是使caspase cascade失活。骨肉瘤细胞中 GRP78 的缺失会对自噬相关基因的基础水平产生负向介导作用。DPT通过磷酸肌醇3-激酶(PI3K)-v-akt小鼠胸腺瘤病毒癌基因同源物(AKT)--雷帕霉素机制靶标(mTOR)轴刺激自噬。DPT引起的自噬在人类骨肉瘤中发挥了积极作用,并阻断了凋亡级联:结论:GRP78抑制剂DPT与药理自噬抑制剂的联合治疗将是一种有效的消除骨肉瘤细胞的方法。
{"title":"Deoxypodophyllotoxin Mediates Autophagy Death through Inhibition of GRP78 in Human Osteosarcoma.","authors":"Xiao-Jun Tang, Ling-Li Luo, Wen-Chao Zhou, Gang-Qing Shi, Xiao-Xu Wang, Tian Zeng, Xiong-Jin Tan, Wei-Ming Guo, An-Bo Gao, Yukun Li, Juan Zou","doi":"10.2174/0115680096312622240812093046","DOIUrl":"https://doi.org/10.2174/0115680096312622240812093046","url":null,"abstract":"<p><strong>Background: </strong>Glucose-regulated protein 78 (GRP78), as a chaperone protein, can protect the endoplasmic reticulum of cells and is expressed to influence chemoresistance and prognosis in cancer. Deoxypodophyllotoxin (DPT) is a compound with antitumor effects on cancers. DPT inhibits the proliferation of osteosarcoma by inducing apoptosis, necrosis, or cell cycle arrest.</p><p><strong>Object: </strong>This study was performed to demonstrate the molecular mechanism by which DPT attenuates osteosarcoma progression through GRP78.</p><p><strong>Methods: </strong>Natural compound libraries and western blot (WB) were used to screen the inhibitors of osteosarcoma GRP78. The expression of mitochondria-related genes in cancer cells of the treatment group was detected by quantitative real-time PCR (qPCR) and WB. 3-(4,5)- Dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT) and 5-ethynyl-2'- deoxyuridine (EDU) were used to discover the activity and proliferation of osteosarcoma cells treated with DPT. We constructed an in vivo mouse model of DPT drug therapy and carried out immunohistochemical detection of xenografts. The treated osteosarcoma cells were analyzed using bioinformatics and electron microscopy. The data were analyzed finally.</p><p><strong>Results: </strong>DPT inhibited osteosarcoma cell survival and the growth of tumor xenografts. It promoted up-regulation of BCL2-associated X protein (Bax) and B-cell CLL/lymphoma 2 (Bcl-2), which serves to mediate and attenuate, respectively, the killing activities of DPT through mitochondria dysfunction. The effect of DPT against cancer cells could be attenuated by the overexpression of GRP78, characterized by the inactivation of the caspase cascade. The loss of GRP78 in osteosarcoma cells negatively mediated the basal level of autophagyassociated genes. DPT stimulated autophagy via the phosphoinositide 3-kinase (PI3K)-v-akt murine thymoma viral oncogene homolog (AKT), a mechanistic target of rapamycin (mTOR) axis. The autophagy caused by DPT played an active role in the osteosarcoma of humans and blocked the apoptotic cascade.</p><p><strong>Conclusion: </strong>Combination treatment with the GRP78 inhibitor DPT and pharmacological autophagy inhibitors will be a meaningful method of obviating osteosarcoma cells.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142343162","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Advancements in Phytosomal Therapies for Liver Cancer: A Comprehensive Review. 肝癌植物疗法的进展:全面回顾。
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-09-23 DOI: 10.2174/0115680096319007240703072650
Sachin Kothawade, Manjusha Bhange, Vishal Vijay Pande

Phytosomes, innovative lipid-compatible complexes formed by combining natural phospholipids with water-soluble phytoconstituents, represent a groundbreaking technology in herbal medicine. This review examines the novel applications of phytosomes in liver cancer treatment. Phytosome technology overcomes traditional obstacles in utilizing herbal potential for modern medicine, such as issues with potency, solubility, permeability, and stability, which has led to increased interest in herbal drug sources. By enhancing the bioavailability and bioefficacy of polyphenolic phytoconstituents, particularly those with anti-angiogenic properties critical for tumor growth and embryonic nourishment, phytosome technology addresses these challenges. The complexity of liver cancer, including both cholangiocarcinoma and hepatocellular carcinoma, demands comprehensive medical management. Although natural compounds like resveratrol, curcumin, and silymarin show promise with their anticancer effects, their full efficacy in human trials is not yet confirmed. Phytosomal preparations, which incorporate these compounds into lipid complexes, offer a potential solution for improved bioavailability and absorption. This review thoroughly explores the application of phytosome technology in herbal medicine, highlighting its potential role in tackling the complexities of liver cancer treatment. As research advances, phytosomes are expected to be a valuable addition to the evolving field of natural product-based therapeutic formulations.

植物体是由天然磷脂与水溶性植物成分结合形成的创新型脂质兼容复合物,是草药领域的一项突破性技术。本综述探讨了植物载体在肝癌治疗中的新应用。植物载体技术克服了在现代医学中利用草药潜力的传统障碍,如药效、溶解性、渗透性和稳定性等问题,从而提高了人们对草药来源的兴趣。通过提高多酚类植物成分的生物利用度和生物有效性,特别是那些对肿瘤生长和胚胎营养至关重要的抗血管生成特性,植物体技术解决了这些难题。肝癌(包括胆管癌和肝细胞癌)病情复杂,需要综合治疗。虽然白藜芦醇、姜黄素和水飞蓟素等天然化合物的抗癌效果值得期待,但它们在人体试验中的全部功效尚未得到证实。将这些化合物纳入脂质复合物的植物载体制剂为提高生物利用度和吸收率提供了一种潜在的解决方案。这篇综述深入探讨了植物体技术在中草药中的应用,强调了它在解决复杂的肝癌治疗中的潜在作用。随着研究的深入,植物载体有望成为不断发展的天然产品治疗配方领域的重要补充。
{"title":"Advancements in Phytosomal Therapies for Liver Cancer: A Comprehensive Review.","authors":"Sachin Kothawade, Manjusha Bhange, Vishal Vijay Pande","doi":"10.2174/0115680096319007240703072650","DOIUrl":"https://doi.org/10.2174/0115680096319007240703072650","url":null,"abstract":"<p><p>Phytosomes, innovative lipid-compatible complexes formed by combining natural phospholipids with water-soluble phytoconstituents, represent a groundbreaking technology in herbal medicine. This review examines the novel applications of phytosomes in liver cancer treatment. Phytosome technology overcomes traditional obstacles in utilizing herbal potential for modern medicine, such as issues with potency, solubility, permeability, and stability, which has led to increased interest in herbal drug sources. By enhancing the bioavailability and bioefficacy of polyphenolic phytoconstituents, particularly those with anti-angiogenic properties critical for tumor growth and embryonic nourishment, phytosome technology addresses these challenges. The complexity of liver cancer, including both cholangiocarcinoma and hepatocellular carcinoma, demands comprehensive medical management. Although natural compounds like resveratrol, curcumin, and silymarin show promise with their anticancer effects, their full efficacy in human trials is not yet confirmed. Phytosomal preparations, which incorporate these compounds into lipid complexes, offer a potential solution for improved bioavailability and absorption. This review thoroughly explores the application of phytosome technology in herbal medicine, highlighting its potential role in tackling the complexities of liver cancer treatment. As research advances, phytosomes are expected to be a valuable addition to the evolving field of natural product-based therapeutic formulations.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-09-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142343152","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 miRNAs to Hinder the Tumorigenesis of Renal Clear Cell Carcinoma Associated with KDR Expression 筛选与 KDR 表达相关的 miRNA 以阻碍肾透明细胞癌的肿瘤发生
IF 3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-09-18 DOI: 10.2174/0115680096321287240826065718
Kaviyaprabha R, Miji TV, Suseela R, Muthusami S, Thangaleela S, Almoallim HS, Priyadarshini S, Bharathi M
Introduction: This study delved into the role of Kinase Insert Domain Receptor (KDR) and its associated miRNAs in renal cell carcinoma through an extensive computational analysis. The potential of our findings to guide future research in this area is significant. Methods: Our methods, which included the use of UALCAN and GEPIA2 databases, as well as miRDB, MirDIP, miRNet v2.0, miRTargetLink, MiEAA v2.1, TarBase v8.0, INTERNET, and miRTarBass, were instrumental in identifying the regulation of miRNA associated with KDR expression. The predicted miRNA was validated with the TCGA-KIRC patients’ samples by implementing CancerMIRNome. The TargetScanHuman v8.0 was implemented to identify the associations between human miRNAs and KDR. A Patch Dock server analyzed the interactions between hsa-miR-200b-3p-KDR and hsa-miR-200b-3p with KDR. Results: The KDR expression rate was investigated in the Kidney Renal Cell Carcinoma (KIRC) samples, and adjacent normal tissues revealed that the expression rate was significantly higher than the normal samples, which was evident from the strong statistical significance (P = 1.63e-12). Likely, the KDR ex-pression rate was estimated as high at tumor grade 1 and gradually decreased till the metastasis grade, reducing the survival rate of the KIRC patients. To identify these signals early, we predicted a miRNA that could trigger the expression of KDR. Furthermore, we uncovered the potential associations between miR-200c-3p expressions by regulating KDR towards the progression of KIRC. Discussion: Upon examining the outcome, it became evident that miR-200c-3p was significantly down-regulated in KIRC compared to the normal samples. Moreover, the negative correlation was obtained for hsa-miR-200c-3p (R = - 0.276) along with the KDR expression describing that the increased rate of hsa-miR-200c-3p might reduce the KDR expression rate, which may suppress the KIRC initiation or progres-sion. Conclusion: The in-silico analysis indicated that the significant increase in KDR expression during the initiation of KIRC could serve as an early diagnostic marker. Moreover, KDR could be utilized to identify advancements in KIRC stages. Additionally, hsa-miR-200c-3p was identified as a potential regulator capable of downregulating and upregulating KDR expression among the 24 miRNAs screened. This find-ing holds promise for future research endeavors. Concurrent administration of the FDA-approved 5-fluor-ouracil with KIRC drugs, such as sorafenib, zidovudine, and everolimus, may have the potential to en-hance the therapeutic efficacy in downregulating hsa-miR-200c-3p. However, further in vitro studies are imperative to validate these findings and gain a comprehensive understanding of the intricate regulatory interplay involving hsa-miR-200c-3p, KDR, 5-fluorouracil, and other FDA-approved drugs for the treat-ment of KIRC. This will facilitate the identification of KIRC stage progression and its underlying pre-ventative mechanisms.
导言:本研究通过广泛的计算分析,深入研究了激酶插入域受体(KDR)及其相关 miRNA 在肾细胞癌中的作用。我们的研究结果对该领域未来的研究具有重要的指导意义。方法:我们的方法包括使用 UALCAN 和 GEPIA2 数据库以及 miRDB、MirDIP、miRNet v2.0、miRTargetLink、MiEAA v2.1、TarBase v8.0、INTERNET 和 miRTarBass,这些方法有助于确定与 KDR 表达相关的 miRNA 的调控。通过使用 CancerMIRNome,预测的 miRNA 与 TCGA-KIRC 患者样本进行了验证。TargetScanHuman v8.0 用于识别人类 miRNA 与 KDR 之间的关联。Patch Dock 服务器分析了 hsa-miR-200b-3p-KDR 和 hsa-miR-200b-3p 与 KDR 之间的相互作用。结果:对肾肾细胞癌(KIRC)样本和邻近正常组织的 KDR 表达率进行了调查,结果显示,KDR 的表达率明显高于正常样本,这在统计学上有很强的显著性(P = 1.63e-12)。据估计,KDR的表达率在肿瘤1级时较高,到转移级时逐渐降低,从而降低了KIRC患者的生存率。为了及早发现这些信号,我们预测了一种能触发 KDR 表达的 miRNA。此外,我们还发现了调控 KDR 的 miR-200c-3p 表达与 KIRC 进展之间的潜在关联。讨论研究结果表明,与正常样本相比,miR-200c-3p 在 KIRC 中明显下调。此外,hsa-miR-200c-3p(R = - 0.276)与 KDR 的表达呈负相关,说明 hsa-miR-200c-3p 的增加可能会降低 KDR 的表达率,从而抑制 KIRC 的发生或发展。结论室内分析表明,在 KIRC 启动过程中,KDR 表达的显著增加可作为早期诊断标志物。此外,KDR 还可用于识别 KIRC 的进展阶段。此外,在筛选出的 24 个 miRNA 中,hsa-miR-200c-3p 被确定为能够下调和上调 KDR 表达的潜在调控因子。这一发现为未来的研究工作带来了希望。在服用索拉非尼、齐多夫定和依维莫司等 KIRC 药物的同时服用美国 FDA 批准的 5-氟-尿嘧啶,可能会增强下调 hsa-miR-200c-3p 的疗效。然而,要验证这些发现,并全面了解涉及 hsa-miR-200c-3p、KDR、5-氟尿嘧啶和其他经 FDA 批准用于治疗 KIRC 的药物的错综复杂的调控相互作用,进一步的体外研究势在必行。这将有助于确定 KIRC 的阶段性进展及其潜在的前期机制。
{"title":"Screening miRNAs to Hinder the Tumorigenesis of Renal Clear Cell Carcinoma Associated with KDR Expression","authors":"Kaviyaprabha R, Miji TV, Suseela R, Muthusami S, Thangaleela S, Almoallim HS, Priyadarshini S, Bharathi M","doi":"10.2174/0115680096321287240826065718","DOIUrl":"https://doi.org/10.2174/0115680096321287240826065718","url":null,"abstract":"Introduction: This study delved into the role of Kinase Insert Domain Receptor (KDR) and its associated miRNAs in renal cell carcinoma through an extensive computational analysis. The potential of our findings to guide future research in this area is significant. Methods: Our methods, which included the use of UALCAN and GEPIA2 databases, as well as miRDB, MirDIP, miRNet v2.0, miRTargetLink, MiEAA v2.1, TarBase v8.0, INTERNET, and miRTarBass, were instrumental in identifying the regulation of miRNA associated with KDR expression. The predicted miRNA was validated with the TCGA-KIRC patients’ samples by implementing CancerMIRNome. The TargetScanHuman v8.0 was implemented to identify the associations between human miRNAs and KDR. A Patch Dock server analyzed the interactions between hsa-miR-200b-3p-KDR and hsa-miR-200b-3p with KDR. Results: The KDR expression rate was investigated in the Kidney Renal Cell Carcinoma (KIRC) samples, and adjacent normal tissues revealed that the expression rate was significantly higher than the normal samples, which was evident from the strong statistical significance (P = 1.63e-12). Likely, the KDR ex-pression rate was estimated as high at tumor grade 1 and gradually decreased till the metastasis grade, reducing the survival rate of the KIRC patients. To identify these signals early, we predicted a miRNA that could trigger the expression of KDR. Furthermore, we uncovered the potential associations between miR-200c-3p expressions by regulating KDR towards the progression of KIRC. Discussion: Upon examining the outcome, it became evident that miR-200c-3p was significantly down-regulated in KIRC compared to the normal samples. Moreover, the negative correlation was obtained for hsa-miR-200c-3p (R = - 0.276) along with the KDR expression describing that the increased rate of hsa-miR-200c-3p might reduce the KDR expression rate, which may suppress the KIRC initiation or progres-sion. Conclusion: The in-silico analysis indicated that the significant increase in KDR expression during the initiation of KIRC could serve as an early diagnostic marker. Moreover, KDR could be utilized to identify advancements in KIRC stages. Additionally, hsa-miR-200c-3p was identified as a potential regulator capable of downregulating and upregulating KDR expression among the 24 miRNAs screened. This find-ing holds promise for future research endeavors. Concurrent administration of the FDA-approved 5-fluor-ouracil with KIRC drugs, such as sorafenib, zidovudine, and everolimus, may have the potential to en-hance the therapeutic efficacy in downregulating hsa-miR-200c-3p. However, further in vitro studies are imperative to validate these findings and gain a comprehensive understanding of the intricate regulatory interplay involving hsa-miR-200c-3p, KDR, 5-fluorouracil, and other FDA-approved drugs for the treat-ment of KIRC. This will facilitate the identification of KIRC stage progression and its underlying pre-ventative mechanisms.","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":3.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142251399","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
Dendrobine Suppresses Tumor Growth by Regulating the PD-1/PD-L1 Checkpoint Pathway in Lung Cancer. 石斛碱通过调节肺癌中的 PD-1/PD-L1 检查点通路抑制肿瘤生长
IF 2.3 4区 医学 Q3 ONCOLOGY Pub Date : 2024-09-18 DOI: 10.2174/0115680096305416240820040314
Linmao Li, Jiejin Nong, Jingui Li, Lini Fang, Meini Pan, Haixian Qiu, Shiqing Huang, Yepeng Li, Meijuan Wei, Haiying Yin

Background: Dendrobine is a bioactive alkaloid isolated from Dendrobium nobile. Studies have evaluated the anti-tumor effect of dendrobine in cancers, including lung cancer. However, the mechanism of dendrobine inhibiting tumors requires further study.

Methods: Bioinformatics was performed to screen the potential targets of dendrobine. The in-tersection of dendrobine and lung cancer targets was performed for KEGG analysis. CCK-8 was used to detect cell viability after dendrobine treatment. A xenograft mouse model was es-tablished to explore the effect of dendrobine on lung cancer. The percentages of PD-L1+, CD4+, CD8+, CD11b+, CD25+FOXP3+ cells, the expression of Ki-67 and caspase-3, the ex-pression of pathway-related proteins, the levels of IL-2, IFN-γ, and TGF-β, and the changes of indicators of liver and renal function were measured.

Results: Dendrobine regulated the PD1/PD-L1 checkpoint signaling pathway and affected the occurrence and development of lung cancer. Dendrobine decreased the cell viability of lung cancer. Dendrobine and anti-PD-L1 decreased tumor growth, increased caspase-3 expression, and reduced Ki-67 expression in tumor tissues. Dendrobine and anti-PD-L1 suppressed pro-tein expression of PD-L1, p-JAK1/JAK1, and p-JAK2/JAK2 in tumor tissues. Greatly, den-drobine and anti-PD-L1 decreased the percentages of PD-L1+, CD11b+, and CD25+FOXP3+ cells, increased the percentages of CD4+ and CD8+cells, and enhanced the levels of IL-2, IFN-γ, and TGF-β in tumor tissues. Dendrobine demonstrated no hepatorenal toxicity to the tumor mice. The combination of dendrobine and anti-PD-L1 greatly strengthened the effects of dendrobine on tumors.

Conclusion: Dendrobine inhibited tumor immune escape by suppressing the PD-1/PD-L1 checkpoint pathway, thus restricting tumor growth of lung cancer.

背景:石斛碱是从金钗石斛中分离出来的一种生物活性生物碱。研究评估了石斛碱对癌症(包括肺癌)的抗肿瘤作用。然而,石斛碱抑制肿瘤的机制还需要进一步研究:方法:采用生物信息学方法筛选石斛碱的潜在靶点。方法:采用生物信息学方法筛选石斛碱的潜在靶点,并对石斛碱与肺癌靶点的内切关系进行 KEGG 分析。用CCK-8检测石斛碱处理后的细胞活力。建立异种移植小鼠模型,探讨石斛碱对肺癌的影响。结果表明:石斛碱对PD-L1+、CD4+、CD8+、CD11b+、CD25+FOXP3+细胞的百分比、Ki-67和caspase-3的表达、通路相关蛋白的表达、IL-2、IFN-γ和TGF-β的水平以及肝肾功能指标的变化均有调节作用:结果:石斛碱调节了PD1/PD-L1检查点信号通路,影响了肺癌的发生和发展。结果:石斛碱调控PD1/PD-L1检查点信号通路,影响肺癌的发生和发展。石斛碱和抗PD-L1能降低肿瘤组织中的肿瘤生长、增加caspase-3的表达和减少Ki-67的表达。石斛碱和抗 PD-L1 可抑制肿瘤组织中 PD-L1、p-JAK1/JAK1 和 p-JAK2/JAK2 的蛋白表达。地屈孕酮和抗-PD-L1可显著降低肿瘤组织中PD-L1+、CD11b+和CD25+FOXP3+细胞的百分比,增加CD4+和CD8+细胞的百分比,并提高IL-2、IFN-γ和TGF-β的水平。石斛碱对肿瘤小鼠没有肝肾毒性。石斛碱与抗PD-L1联用可大大增强石斛碱对肿瘤的作用:结论:石斛碱通过抑制PD-1/PD-L1检查点通路抑制肿瘤免疫逃逸,从而限制肺癌肿瘤的生长。
{"title":"Dendrobine Suppresses Tumor Growth by Regulating the PD-1/PD-L1 Checkpoint Pathway in Lung Cancer.","authors":"Linmao Li, Jiejin Nong, Jingui Li, Lini Fang, Meini Pan, Haixian Qiu, Shiqing Huang, Yepeng Li, Meijuan Wei, Haiying Yin","doi":"10.2174/0115680096305416240820040314","DOIUrl":"https://doi.org/10.2174/0115680096305416240820040314","url":null,"abstract":"<p><strong>Background: </strong>Dendrobine is a bioactive alkaloid isolated from Dendrobium nobile. Studies have evaluated the anti-tumor effect of dendrobine in cancers, including lung cancer. However, the mechanism of dendrobine inhibiting tumors requires further study.</p><p><strong>Methods: </strong>Bioinformatics was performed to screen the potential targets of dendrobine. The in-tersection of dendrobine and lung cancer targets was performed for KEGG analysis. CCK-8 was used to detect cell viability after dendrobine treatment. A xenograft mouse model was es-tablished to explore the effect of dendrobine on lung cancer. The percentages of PD-L1+, CD4+, CD8+, CD11b+, CD25+FOXP3+ cells, the expression of Ki-67 and caspase-3, the ex-pression of pathway-related proteins, the levels of IL-2, IFN-γ, and TGF-β, and the changes of indicators of liver and renal function were measured.</p><p><strong>Results: </strong>Dendrobine regulated the PD1/PD-L1 checkpoint signaling pathway and affected the occurrence and development of lung cancer. Dendrobine decreased the cell viability of lung cancer. Dendrobine and anti-PD-L1 decreased tumor growth, increased caspase-3 expression, and reduced Ki-67 expression in tumor tissues. Dendrobine and anti-PD-L1 suppressed pro-tein expression of PD-L1, p-JAK1/JAK1, and p-JAK2/JAK2 in tumor tissues. Greatly, den-drobine and anti-PD-L1 decreased the percentages of PD-L1+, CD11b+, and CD25+FOXP3+ cells, increased the percentages of CD4+ and CD8+cells, and enhanced the levels of IL-2, IFN-γ, and TGF-β in tumor tissues. Dendrobine demonstrated no hepatorenal toxicity to the tumor mice. The combination of dendrobine and anti-PD-L1 greatly strengthened the effects of dendrobine on tumors.</p><p><strong>Conclusion: </strong>Dendrobine inhibited tumor immune escape by suppressing the PD-1/PD-L1 checkpoint pathway, thus restricting tumor growth of lung cancer.</p>","PeriodicalId":10816,"journal":{"name":"Current cancer drug targets","volume":null,"pages":null},"PeriodicalIF":2.3,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142281668","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
Current cancer drug targets
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