selumetinib

司美替尼
  • 文章类型: Journal Article
    整合药物警戒和网络毒理学方法,以探索司美替尼的潜在不良药物事件(ADE)和毒性机制,为快速了解新上市药品的安全性和毒理机制提供参考。
    以司美替尼为例,本研究整合了基于现实数据的药物警戒方法和网络毒理学方法,分析了其ADE及其潜在的毒理学机制。首先,司美替尼的ADE报告摘自美国食品和药物管理局(FDA)不良事件报告系统(FAERS),ADE信号通过报告比值比(ROR)和英国药品和医疗保健产品监管机构(MHRA)方法检测。根据源自调节活动医学词典(MedDRA)的优选术语(PT)和系统器官类别(SOC)对ADE信号进行分类和描述。网络毒理学方法用于分析感兴趣的SOCs的毒理学机理。具体步骤包括使用TOXRIC预测司美替尼的潜在靶标,STITCH,ChEMBL,CTD,SwissTargetPreditcion,和Super-PRED数据库,使用GeneCards数据库收集SOC的目标,通过STRING数据库进行蛋白质-蛋白质相互作用(PPI)分析,通过DAVID数据库进行基因本体论(GO)和京都基因和基因组百科全书(KEGG)分析,并使用AutoDock软件测试分子亲和力。
    共提取了1388份与司美替尼相关的ADE报告,通过ROR和MHRA方法检测到53个阳性信号,其中20个信号未在包装说明书中提及,包括钉子在内,高磷酸盐血症,心脏瓣膜疾病,血尿,中性粒细胞减少症,等。对6种SOCs参与ADE阳性信号的毒理机制分析显示,关键靶点包括EGFR,STAT3、AKT1、IL6、BCL2等。,关键通路包括PI3K/Akt通路,凋亡,ErbB信号通路,和EGFR酪氨酸激酶抑制剂耐药,等。分子对接试验显示司美替尼与这些途径中的关键靶标自发结合。
    药物警戒分析确定了一些新的潜在的司美替尼的ADE,网络毒理学分析表明,司美替尼的毒性作用可能与PI3K/Akt通路有关,凋亡,ErbB信号通路,EGFR酪氨酸激酶抑制剂耐药等通路。
    UNASSIGNED: To integrate pharmacovigilance and network toxicology methods to explore the potential adverse drug events (ADEs) and toxic mechanisms of selumetinib, and to provide a reference for quickly understanding the safety and toxicological mechanisms of newly marketed drugs.
    UNASSIGNED: Taking selumetinib as an example, this study integrated pharmacovigilance methods based on real-world data and network toxicology methods to analyze its ADE and its potential toxicological mechanism. First, the ADE reports of selumetinib were extracted from the US Food and Drug Administration (FDA) adverse event reporting system (FAERS), and the ADE signals were detected by reporting odds ratio (ROR) and UK medicines and healthcare products regulatory agency (MHRA) methods. The ADE signals were classified and described according to the preferred terms (PTs) and system organ class (SOC) derived from the Medical Dictionary for Regulatory Activities (MedDRA). The network toxicology method was used to analyze the toxicological mechanism of the interested SOCs. The specific steps included predicting the potential targets of selumetinib using TOXRIC, STITCH, ChEMBL, CTD, SwissTargetPreditcion, and Super-PRED databases, collecting the targets of SOC using GeneCards database, conducting protein-protein interaction (PPI) analysis through STRING database, conducting gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) analysis through DAVID database, and testing the molecular affinity using AutoDock software.
    UNASSIGNED: A total of 1388 ADE reports related to selumetinib were extracted, and 53 positive signals were detected by ROR and MHRA methods, of which 20 signals were not mentioned in the package insert, including ingrowing nail, hyperphosphatemia, cardiac valve disease, hematuria, neutropenia, etc. Analysis of the toxicological mechanism of six SOCs involved in positive ADE signals revealed that the key targets included EGFR, STAT3, AKT1, IL6, BCL2, etc., and the key pathways included PI3K/Akt pathway, apoptosis, ErbB signaling pathway, and EGFR tyrosine kinase inhibitor resistance, etc. Molecular docking assays showed spontaneous binding of selumetinib to key targets in these pathways.
    UNASSIGNED: The pharmacovigilance analysis identified some new potential ADEs of selumetinib, and the network toxicology analysis showed that the toxic effects of selumetinib may be related to PI3K/Akt pathway, apoptosis, ErbB signaling pathway, EGFR tyrosine kinase inhibitor resistance and other pathways.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:结直肠癌(CRC)是世界上第三大恶性肿瘤。基于5-氟尿嘧啶(5-FU)的化疗是CRC的一线化疗方案,而获得性耐药对治疗CRC患者构成巨大障碍,其机制仍不清楚。因此,鉴定与5-FU化疗相关的基因并寻求二线治疗是改善CRC患者生存和预后的必要手段。
    方法:使用癌症治疗反应门户(CTRP)数据库和癌症药物敏感性基因组学(GDSC)数据库来鉴定CRC相关基因和5-FU耐药CRC的潜在二线治疗。从GEO数据集获得CRC组织的单细胞RNA测序数据。在体外和体内模型中研究了ITGA2和5-FU抗性之间的关系。
    结果:ACOX1和ITGA2被确定为与5-FU耐药相关的风险生物标志物。我们开发了一个风险签名,由ACOX1和ITGA2组成,能够很好地区分5-FU抗性和5-FU敏感性。单细胞测序数据显示,ITGA2主要富集在恶性细胞中。ITGA2与大多数小分子抑制剂的IC50值呈负相关,其中司米替尼的负相关性最高。最后,敲低ITGA2可以使5-FU耐药CRC细胞对5-FU敏感,联合司美替尼可以提高5-FU耐药细胞的治疗效果。
    结论:总之,我们的研究结果证明了ITGA2在增强CRC细胞化疗耐药中的关键作用,并提示司美替尼可以通过抑制ITGA2表达恢复化疗耐药CRC细胞对5-FU的敏感性.
    BACKGROUND: Colorectal cancer (CRC) is the third most malignant tumor in the world. 5-fluorouracil (5‑FU) -based chemotherapy is the first-line chemotherapy scheme for CRC, whereas acquired drug resistance poses a huge obstacle to curing CRC patients and the mechanism is still obscure. Therefore, identification of genes associated with 5‑FU chemotherapy and seeking second-line treatment are necessary means to improve survival and prognosis of patients with CRC.
    METHODS: The Cancer Therapeutics Response Portal (CTRP) database and Genomics of Drug Sensitivity in Cancer (GDSC) database were used to identify CRC-related genes and potential second-line therapies for 5-FU-resistant CRC. The single-cell RNA sequencing data for CRC tissues were obtained from a GEO dataset. The relationship between ITGA2 and 5-FU-resistant was investigated in vitro and in vivo models.
    RESULTS: ACOX1 and ITGA2 were identified as risk biomarkers associated with 5-FU-resistance. We developed a risk signature, consisting of ACOX1 and ITGA2, that was able to distinguish well between 5-FU-resistance and 5-FU-sensitive. The single-cell sequencing data showed that ITGA2 was mainly enriched in malignant cells. ITGA2 was negatively correlated with IC50 values of most small molecule inhibitors, of which selumetinib had the highest negative correlation. Finally, knocking down ITGA2 can make 5-FU-resistant CRC cells sensitive to 5-FU and combining with selumetinib can improve the therapeutic effect of 5-FU resistant cells.
    CONCLUSIONS: In summary, our findings demonstrated the critical role of ITGA2 in enhancing chemotherapy resistance in CRC cells and suggested that selumetinib can restore the sensitivity of chemotherapy-resistant CRC cells to 5-FU by inhibiting ITGA2 expression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Selumetinib是一种口服,有效,和靶向丝裂原活化蛋白激酶1和2(MEK1/2)的选择性酪氨酸激酶抑制剂,在多种肿瘤类型中临床活跃,如1型神经纤维瘤病(NF1),黑色素瘤,胶质瘤和非小细胞肺癌(NSCLC)。本文的目的是评估司美替尼对12种人UDP-葡萄糖基转移酶(UGT)活性的影响,包括UGT1A1、1A3、1A4、1A6、1A7、1A8、1A9、1A10、2B4、2B7、2B15和2B17,以及其诱导临床药物-药物相互作用(DDIs)的潜力。结果表明,司米替尼通过混合抑制机制有效抑制UGT2B7的活性,抑制常数为5.79±0.65μM。此外,当患者每天两次服用司美替尼75mg时,作为共同给药药物的UGT2B7底物的血浆浓度预计将增加至少84%,提示诱导临床DDI的高潜力。塞美替尼对其他人类UGTs表现出弱抑制作用,不太可能触发UGTs介导的DDI,除了UGT2B7。因此,司美替尼与UGT2B7底物药物联合使用需要额外注意,以避免临床治疗中的不良事件.
    Selumetinib is an oral, effective, and selective tyrosine kinase inhibitor targeting mitogen-activated protein kinase 1 and 2 (MEK1/2), which is clinically active in multiple tumor types, such as neurofibromatosis type 1 (NF1), melanoma, gliomas and non-small cell lung cancer (NSCLC). The purpose of this article was to assess the effects of selumetinib on the activities of twelve human UDP-glucosyltransferases (UGTs) including UGT1A1, 1A3, 1A4, 1A6, 1A7, 1A8, 1A9, 1A10, 2B4, 2B7, 2B15, and 2B17, and its potential for inducing clinical drug-drug interactions (DDIs). The results demonstrated that selumetinib potently inhibited the activity of UGT2B7 through the mechanism of mixed inhibition with the inhibition constant value of 5.79 ± 0.65 μM. Furthermore, the plasma concentration of UGT2B7 substrate as the co-administered drug was predicted to be increased by at least 84 % when patients took selumetinib 75 mg twice daily, suggesting a high potential to induce clinical DDIs. Selumetinib exhibited weak inhibitory effects on other human UGTs and was unlikely to trigger off UGTs-mediated DDIs except for UGT2B7. Therefore, the combination of selumetinib with the substrate drug of UGT2B7 requires additional attention to avoid adverse events in clinical treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:在1型神经纤维瘤病(NF1)和无法手术的丛状神经纤维瘤(PN)患者中批准司美替尼重塑了该疾病的临床管理格局,需要进一步综合评价该药的疗效和安全性。
    方法:在Pubmed数据库中全面搜索了报告埃米替尼在NF1患者中的疗效和安全性的原始文章,Embase数据库,科克伦图书馆,和WebofScience数据库,并筛选符合标准的研究。我们汇总了客观反应率(ORR),疾病控制率(DCR),疾病进展率(DPR),和使用meta分析的PN相关并发症的改善率。对药物相关不良事件的发生率进行统计学分析。
    结果:本研究包括10项临床试验,涉及268名患者。合并的ORR为68.0%(95%CI58.0-77.3%),DCR为96.8%(95%CI90.8~99.7%),DPR仅为1.4%(95%CI0~4.3%).疼痛的合并改善率为75.3%(95%CI56.2-90.9%),运动障碍的合并改善率为77.8%(95%CI63.1-92.5%)。大多数不良事件是轻微的,最常见的是胃肠道反应(腹泻:62.5%;呕吐:54.5%)。
    结论:我们的研究表明,司美替尼对NF1和PN患者有效,显着改善与PN相关的严重并发症,并具有可耐受的毒性。我们的发现有助于提高临床医生应用司美替尼的信心,并促进新药的临床采用和获益。
    BACKGROUND: The approval of selumetinib in patients with neurofibromatosis type 1(NF1) and inoperable plexiform neurofibromas (PN) has reshaped the landscape of clinical management of the disease, and further comprehensive evaluation of the drug\'s efficacy and safety is needed.
    METHODS: Original articles reporting on the efficacy and safety of elumetinib in patients with NF1 were comprehensively searched in the Pubmed database, Embase database, Cochrane Library, and Web of Science database and screened for inclusion of studies that met the criteria. We pooled the objective response rate (ORR), disease control rate (DCR), disease progression rate (DPR), and the rate of improvement in PN-related complications using meta-analysis. The incidence of drug-related adverse events was also statistically analyzed.
    RESULTS: This study included 10 clinical trials involving 268 patients. The pooled ORR was 68.0% (95% CI 58.0-77.3%), the DCR was 96.8% (95% CI 90.8-99.7%) and the DPR was only 1.4% (95% CI 0-4.3%). The pooled improvement rate was 75.3% (95% CI 56.2-90.9%) for pain and 77.8% (95% CI 63.1-92.5%) for motor disorders. Most adverse events were mild, with the most common being gastrointestinal reactions (diarrhea: 62.5%; vomiting: 54.5%).
    CONCLUSIONS: Our study demonstrates that selumetinib is effective in patients with NF1 and PN, significantly improving the serious complications associated with PN as well as having tolerable toxicities. Our findings help to increase clinicians\' confidence in applying selumetinib and promote the clinical adoption and benefit of the new drug.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    吉非替尼,作为第一代表皮生长因子受体酪氨酸激酶抑制剂(EGFR-TKI),在非小细胞肺癌(NSCLC)的治疗方面取得了长足的进步,但是耐药性不可避免地会发生。因此,探索吉非替尼的耐药机制和开发新的联合治疗策略具有重要意义.在我们的研究中,结果显示,曲美替尼(AZD6244)与吉非替尼协同抑制NSCLC的增殖。塞美替尼还增强吉非替尼诱导的吉非替尼耐药肺癌细胞系的凋亡和迁移抑制能力。随后,观察到MIG6和STAT3之间的负调节,并通过STRING数据库和蛋白质印迹分析进行验证.在吉非替尼耐药细胞中与司美替尼共同治疗时,STAT3的持续激活显着下调。然而,曲美替尼和吉非替尼联合使用导致的p-STAT3下调被MIG6缺失所抵消,提示曲美替尼通过调节NSCLC中的MIG6/STAT3增强吉非替尼敏感性.相比之下,当MIG6过表达时,用吉非替尼和司美替尼治疗后,p-STAT3被进一步抑制。此外,司美替尼和吉非替尼联合用药可有效促进肺癌移植瘤体内对吉非替尼的敏感性,抑瘤率达到81.49%,而吉非替尼单药组的肿瘤抑制率仅为31.95%。总的来说,MIG6/STAT3负调控在STAT3的持续激活和对EGFR-TKIs的耐药中起重要作用。我们的研究还表明,EGFR-TKIs联合MEK1/2抑制剂,如司美替尼,可能对那些对EGFR-TKIs产生原发性或获得性耐药的NSCLC患者有益,为TKIs与司美替尼联合应用于临床肿瘤治疗提供理论支持。
    Gefitinib, as the first-generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), has achieved great advances in the treatment of non-small cell lung cancer (NSCLC), but drug resistance will inevitably occur. Therefore, exploring the resistance mechanism of gefitinib and developing new combination treatment strategies are of great importance. In our study, the results showed that selumetinib (AZD6244) synergistically inhibited the proliferation of NSCLC with gefitinib. Selumetinib also enhanced gefitinib-induced apoptosis and migration inhibition ability in gefitinib-resistant lung cancer cell lines. Subsequently, the negative regulation between MIG6 and STAT3 was observed and verified through the STRING database and western blotting assays. Sustained activation of STAT3 was significantly downregulated when co-treatment with selumetinib in gefitinib-resistant cells. However, the downregulation of p-STAT3, resulting from the combination of selumetinib and gefitinib was counteracted by the deletion of MIG6, suggesting that selumetinib enhanced gefitinib sensitivity by regulating MIG6/STAT3 in NSCLC. In contrast, p-STAT3 was further inhibited after treatment with gefitinib and selumetinib when MIG6 was overexpressed. Furthermore, the combined administration of selumetinib and gefitinib effectively promoted the sensitivity of lung cancer xenografts to gefitinib in vivo, and the tumor inhibition rate reached 81.49%, while the tumor inhibition rate of the gefitinib monotherapy group was only 31.95%. Overall, MIG6/STAT3 negative regulation plays an important role in the sustained activation of STAT3 and the resistance to EGFR-TKIs. Our study also suggests that EGFR-TKIs combined with MEK1/2 inhibitors, such as selumetinib, may be beneficial to those NSCLC patients who develop a primary or acquired resistance to EGFR-TKIs, providing theoretical support for combining TKIs and selumetinib in clinical cancer treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:针对MEK分子的1型神经纤维瘤病(NF1)相关丛状神经纤维瘤(pNF)的靶向治疗尚未显示出完全抑制疾病的令人信服的结果。可能是由于其他信号通路串扰。我们先前的研究表明,NF1相关pNF中YAP分子的核易位增加。在这里,我们决定进一步研究MEK治疗期间YAP干扰对NF1相关pNF的治疗关系。方法:通过司美替尼(MEK抑制剂),首先进行RNA测序以鉴定pNF雪旺细胞中信号通路的变化,这可能与YAP监管有关。进行核-细胞质分级分离和蛋白质印迹以显示在司美替尼治疗下的细胞内YAP变化。第三,进行了一系列的体外试验,包括流式细胞术,CCK-8,以及在司美替尼和维替泊芬(YAP抑制剂)双重治疗下的集落/球形成。此外,采用Chou-Talalay方法评价此类药物组合的协同抑制作用。异种移植研究也用于检测体内的组合效应。结果:RNA测序显示,司美替尼治疗可能与NF1相关pNF肿瘤细胞中Hippo通路的不良激活有关,这可能会降低其药效。接下来,核-细胞质分级分离和进一步的研究表明,司美替尼可以在体外促进YAP的核易位和转录激活,这可能会导致上述对司美替尼治疗的耐药性。此外,当基于维替泊芬和司美替尼进行联合治疗时,在体外和体内异种移植模型中观察到对NF1相关pNF肿瘤细胞的细胞毒性的协同作用。结论:抑制YAP可有效提高NF1相关pNF肿瘤细胞对司美替尼的敏感性。YAP和MEK的双重靶向可能是治疗NF1相关pNF的有希望的治疗策略。
    Background: Targeted therapy of Neurofibromatosis type 1 (NF1) related plexiform neurofibroma (pNF) aiming at MEK molecule has not demonstrated a convincing result for complete disease inhibition, probably due to other signal pathways crosstalk. Our previous study revealed an increased nuclear translocation of YAP molecule in NF1 related pNF. Herein, we decided to further investigate the therapeutic relations of YAP interference during the MEK treatment against NF1 related pNF. Methods: By means of selumetinib (MEK-inhibitor), RNA-sequencing was firstly performed to identify the changes of signal pathways in pNF Schwann cells, which was probably related to YAP regulation. Nuclear-cytoplasmic fractionation and western blotting were performed to show the intracellular YAP changes under selumetinib treatment. Thirdly, a series of in vitro assays were performed including flow cytometry, CCK-8, and colony/sphere formation under dual treatment of selumetinib and verteporfin (YAP-inhibitor). In addition, Chou-Talalay method was adopted to evaluate the synergistic inhibiting effects of such drug combination. Xenograft study was also used to detect the combining effects in vivo. Results: RNA-sequencing revealed that selumetinib treatment might be associated with the undesirable activation of Hippo pathway in NF1 related pNF tumor cells, which might reduce its pharmaceutic effects. Next, nuclear-cytoplasmic fractionation and further studies demonstrated that selumetinib could promote the nuclear translocation and transcriptional activation of YAP in vitro, which might cause the aforementioned resistance to selumetinib treatment. Additionally, when combined treatments were performed based on verteporfin and selumetinib, synergistic effects were observed on cytotoxicity of NF1 related pNF tumor cells in vitro and in vivo xenograft models. Conclusion: YAP inhibition can effectively sensitize NF1 related pNF tumor cells to selumetinib. Dual targeting of YAP and MEK might be a promising therapeutic strategy for treating NF1 related pNF.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    非小细胞肺癌(NSCLC)是所有肺癌中最常见的亚型,KRAS是该人群中最常见的突变。不幸的是,由于缺乏经批准的靶向治疗,这个亚组仍然"不可用药".在几个试验中,塞美替尼已作为二级治疗进行了研究,并与各种药物方案进行了比较。因此,我们进行了系统评价和网络荟萃分析,以确定该药物与其他药物在晚期和恶性NSCLC患者中的疗效比较.
    截至2020年7月1日,9个数据库(PubMed,Scopus,WebofScience,mRCT,ICTRP,clinicaltrials.gov,VHL,SIGLE,和GoogleScholar)搜索了遵循PICOS框架的研究:随机试验报告了司美替尼与其他治疗相比在晚期/转移性NSCLC患者中的疗效(疾病进展/无缓解率)。使用修订的Cochrane偏倚风险工具评估检索到的研究的质量。进行了频繁网络荟萃分析,以评估与其他疗法和/或安慰剂相比,司米替尼的疗效。
    在主要搜索产生的163篇文章中,9项研究(1,195例患者)最终纳入我们的系统评价。大多数临床病例的表现状态(PS)为0-2,平均年龄为62岁。司美替尼的总体疗效为71.77%(95%CI:63.24-81.45%),与联合治疗相比,单独使用司美替尼具有更好的疗效(65.20%vs.74.08%)。在网络分析中,与化疗或免疫治疗相比,司美替尼的疗效更高,但不是很明显。司美替尼的总体SAE率为42.96%(95%CI:34.74-53.13%),与联合治疗相比,司米替尼具有明显更好的安全性(10.49%vs.47.38%)。在网络分析中,安慰剂的安全性最好,其次是司米替尼和化疗和免疫治疗.五项研究有很高的偏倚风险,2有一些顾虑,和2有低风险的偏倚。
    与联合治疗NSCLC相比,司美替尼的疗效并不优越,但确实具有更好的安全性。目前的证据仍然有限,和更强大的试验仍然需要。
    UNASSIGNED: Non-small cell lung cancer (NSCLC) is the most common subtype of all lung cancers, and KRAS is the most common mutation in this population. Unfortunately, this subgroup remains \"undruggable\" with the lack of an approved targeted therapy. Selumetinib has been investigated as a secondary therapy in several trials and compared to various drug regimens. Therefore, we conducted this systematic review and network meta-analysis to determine the comparative effectiveness of this drug as compared to others in patients with late-stage and malignant NSCLC.
    UNASSIGNED: Up to July 1, 2020, 9 databases (PubMed, Scopus, Web of Science, mRCT, ICTRP, clinicaltrials.gov, VHL, SIGLE, and Google Scholar) were searched for studies following the PICOS framework: randomized trials reporting the efficacy (rate of disease progression/lack of response) of selumetinib compared to other therapies in patients with late-stage/metastatic NSCLC. The quality of retrieved studies were assessed with the revised Cochrane risk-of-bias tool. Frequentist network meta-analysis was conducted to estimate the efficacy of selumetinib as compared to other therapies and/or placebo.
    UNASSIGNED: Out of the 163 articles yielded from the primary search, 9 studies (1,195 patients) were finally included in our systematic review. The majority of clinical cases had a performance status (PS) of 0-2, and the mean age was 62 years. The overall efficacy of selumetinib was 71.77% (95% CI: 63.24-81.45%), with selumetinib administered alone having better efficacy compared to combined therapy (65.20% vs. 74.08%). In the network analysis, selumetinib had higher efficacy compared to chemo- or immune therapy, but not significantly so. The overall SAE rate of selumetinib was 42.96% (95% CI: 34.74-53.13%), with selumetinib having a significantly better safety profile compared to combined therapy (10.49% vs. 47.38%). In the network analysis, the placebo had the best safety profile followed by selumetinib and chemo- and immune therapy. Five studies had high risk of bias, 2 had some concerns, and 2 had low risk of bias.
    UNASSIGNED: The efficacy of selumetinib is not superior compared to combined therapy for treating NSCLC but does have a better safety profile. Current evidence is still limited, and more robust trials are still required.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    基于分子对接和分子动力学模拟,寻找MEK抑制剂塞美替尼治疗低级别胶质瘤(LGG)的新靶点和机制,为其临床用药提供理论指导。通过化合物靶标预测数据库捕获所有可能的Selumetinib靶标。发现了司马替尼治疗LGG的新靶点,并通过分子对接评估了其机制。基因差异分析,分子动力学模拟,和蛋白质亚细胞定位预测。本研究共筛选了100种赛美替尼靶标和85种LGG相关靶标。在两者的交汇处有7个活动目标。通过蛋白质相互作用(PPI),基因富集分析,和基因差异分析,最终筛选出一个有效的西米替尼靶点,CDK2主要存在于细胞质中,内质网,和质膜;该靶标通过抑制PI3KAkt的信号通路,参与肽氨基酸修饰等生物学过程,在LGG的治疗中发挥作用,细胞内信号转导的调节,和细胞代谢的正向调节。CDK2可能是司马替尼在临床治疗LGG的新方向。
    Based on molecular docking and molecular dynamics simulation, to find a new target and mechanism of MEK inhibitor Selumetinib in the treatment of low-grade glioma (LGG), and to provide theoretical guidance for its clinical medication. All possible targets of Selumetinib were fished through the compound target prediction database. New targets of Selumetinib in the treatment of LGG were found and its mechanism was evaluated employing molecular docking, gene difference analysis, molecular dynamics simulation, and protein subcellular localization prediction. A total of 100 Selumetinib targets and 85 LGG-related targets were screened in this study. There were 7 active targets at the intersection of the two. Through protein interaction (PPI), gene enrichment analysis, and gene difference analysis, one effective target of Selumetinib was finally screened, CDK2 mainly existing in the cytoplasm, endoplasmic reticulum, and plasma membrane; the target plays a role in the treatment of LGG by inhibiting the signal pathways of PI3K Akt and participating in biological processes such as peptide amino acid modification, regulation of intracellular signal transduction, and positive regulation of cell metabolism. CDK2 may be a new direction of Selumetinib in the clinical treatment of LGG.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    本研究旨在研究木犀草素对结直肠癌(CRC)的影响,并探讨其作用机制。HCT-116和HT-29细胞用木犀草素处理,顺铂,或司美替尼。细胞存活,细胞增殖,凋亡和细胞周期分布,和DNA损伤检测使用细胞计数试剂盒-8,集落形成,流式细胞术,和免疫荧光染色分析,分别。免疫印迹法检测细胞凋亡相关蛋白的表达,周期相关,DNA损伤相关,丝裂原活化蛋白激酶(MAPK)通路相关蛋白。木犀草素通过减少细胞存活和增殖而对细胞生长表现出抑制作用,诱导细胞凋亡和DNA损伤,并在HCT-116和HT-29细胞中以浓度依赖性方式抑制细胞周期。同时,木犀草素增加促凋亡蛋白的表达,p-CHK1(诱导细胞周期停滞的核心),和DNA切除修复蛋白和减少的抗凋亡蛋白,G2-M期相关蛋白,和DNA修复蛋白。与单独使用顺铂相比,顺铂和木犀草素的组合显着降低了HCT-116和HT-29细胞的细胞存活率并增加了凋亡率。使用比较毒性基因组学数据库和STITCH和MalaCards数据库的生物信息学分析显示,MAPK途径涉及木犀草素的药理学。此外,Westernblotting表明木犀草素通过抑制CRC中的MAPK信号通路发挥抑制作用,与司美替尼联合使用时会增强。木犀草素还可以在体内预防CRC中的肿瘤发生。总之,木犀草素抑制细胞增殖,阻断了细胞周期,并通过介导MAPK通路诱导CRC细胞DNA损伤和凋亡进程。
    This study aimed to investigate the effects of luteolin on colorectal cancer (CRC) and explore its underlying mechanism. HCT-116 and HT-29 cells were treated with luteolin, cisplatin, or selumetinib. The cell survival, cell proliferation, apoptosis and cell cycle distribution, and DNA damage were detected using Cell Counting Kit-8, colony formation, flow cytometry, and immunofluorescence staining analysis, respectively. Western blotting was used to detect the expression of apoptosis-related, cycle-related, DNA-damage-related, and mitogen-activated protein kinase (MAPK) pathway-related proteins. Luteolin showed inhibitory effects on cellular growth by reducing cell survival and proliferation, inducing apoptosis and DNA damage, and arresting the cell cycle in a concentration-dependent manner in HCT-116 and HT-29 cells. Meanwhile, luteolin increased the expression of pro-apoptotic proteins, p-CHK1 (central to the induction of cell cycle arrest), and DNA excision repair protein and decreased anti-apoptotic proteins, G2-M phase-related proteins, and DNA repair proteins. The combination of cisplatin and luteolin significantly decreased cell survival and increased the apoptosis rate of HCT-116 and HT-29 cells compared with cisplatin alone. Bioinformatic analysis using the Comparative Toxicogenomics Database and STITCH and MalaCards databases showed that the MAPK pathway is involved in the pharmacology of luteolin. Furthermore, western blotting demonstrated that luteolin plays an inhibitory role by suppressing the MAPK signaling pathway in CRC, which is enhanced when combined with selumetinib. Luteolin can also prevent tumourigenesis in CRC in vivo. In conclusion, luteolin suppressed cell proliferation, blocked the cell cycle, and induced DNA damage and apoptosis progression in CRC cells by mediating the MAPK pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:肝细胞癌(HCC)是全球最常见的恶性肿瘤之一,预后不良。索拉非尼,肝癌的唯一靶向治疗剂,是一种多激酶抑制剂,其靶标包括RAF和VEGFR-2/3,显示出非常有限的能力,可以延长晚期转移性HCC患者的生存期约三个月。在临床试验中,包括曲美替尼和司米替尼在内的MEK抑制剂与索拉非尼联合使用已显示出有希望的疗效。然而,这些药物联合作用的机制尚不清楚.
    方法:实验中使用两种HCC细胞系(Bel7402和SMMC7721)。通过蛋白质印迹定量HCC细胞系的蛋白质表达。通过细胞计数试剂盒-8和集落形成测定检查细胞活力。通过联合指数(CI)值确定索拉非尼与曲美替尼/司美替尼之间的药物相互作用。
    结果:在这项研究中,我们发现短期索拉非尼治疗可以抑制Bel7402和SMMC7721细胞的下游RAF效应子磷酸化(p)-MEK和p-ERK,而长期索拉非尼治疗可以诱导这两种人肝癌细胞系中p-MEK和p-ERK表达的反弹。然后,我们测试了索拉非尼与两种不同的FDA批准的MEK抑制剂联合使用的效果。曲美替尼和司美替尼,在两个细胞系中。Westernblot分析显示曲美替尼/司美替尼可以消除长期索拉非尼治疗引起的ERK活化。细胞计数试剂盒-8和集落形成试验表明,索拉非尼或曲美替尼/司美替尼单独使用对这些HCC细胞系的增殖产生较小的影响,而联合疗法诱导强烈的生长抑制。使用CompuSyn软件的CI测定表明,组合疗法可以在这两种细胞系中产生协同作用。此外,机制研究表明,联合疗法可以协同降低增殖相关蛋白的表达,包括细胞周期蛋白D1和c-Myc。
    结论:综合来看,我们的研究表明,长期索拉非尼治疗引起的p-ERK反弹可能会限制索拉非尼单药治疗的益处,MEK抑制剂曲美替尼和司美替尼可以增强索拉非尼在肝癌细胞中的疗效。
    BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most common malignant tumours worldwide and has a poor prognosis. Sorafenib, the only targeted therapeutic agent for HCC, is a multiple kinase inhibitor with targets including RAF and VEGFR-2/3 that display a very limited ability to extend the survival of patients with advanced metastatic HCC for approximately three months. MEK inhibitors including trametinib and selumetinib have shown promising efficacy in combination with sorafenib in clinical trials. However, the mechanisms about the combined effect of these drugs remain unclear.
    METHODS: Two HCC cell lines (Bel7402 and SMMC7721) were used in the experiments. The protein expression of HCC cell lines was quantified via western blotting. Cell viability was examined by cell counting kit-8 and colony formation assays. Drug interactions between sorafenib and trametinib/selumetinib were determined by the combination index (CI) value.
    RESULTS: In this study, we found that short-term sorafenib treatment could inhibit the downstream RAF effectors phosphorylated (p)-MEK and p-ERK in Bel7402 and SMMC7721 cells, while long-term sorafenib treatment could induce a rebound of p-MEK and p-ERK expression in these two human HCC cell lines. We then tested the effect of sorafenib combined with two different FDA-approved MEK inhibitors, trametinib and selumetinib, in the two cell lines. Western blot analysis showed that trametinib/selumetinib could abolish the ERK activation caused by long-term sorafenib treatment. Cell counting kit-8 and colony formation assays indicated that the use of sorafenib or trametinib/selumetinib alone produced a minor effect on the proliferation of these HCC cell lines, while the combination therapies induced strong growth inhibition. CI assays using CompuSyn software indicated that the combined therapies could produce a synergistic effect in these two cell lines. In addition, mechanistic studies revealed that the combination therapies could synergistically reduce the expression of proliferation-related proteins, including cyclin D1 and c-Myc.
    CONCLUSIONS: Taken together, our study showed that the rebound of p-ERK induced by long-term sorafenib treatment might limit the benefit of sorafenib monotherapy, and the MEK inhibitors trametinib and selumetinib could enhance the efficacy of sorafenib in HCC cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号