EGFR-TKIs resistance

EGFR - TKIs 耐药
  • 文章类型: Journal Article
    对EGFR酪氨酸激酶抑制剂(EGFR-TKIs)的获得性耐药是非小细胞肺癌(NSCLC)患者治疗失败的主要原因。最近发现趋化因子(C-C基序)配体2(CCL2)在确定抗癌治疗反应中起关键作用。然而,CCL2在EGFR-TKIs耐药中的作用和机制尚未完全阐明.在本研究中,我们关注CCL2在NSCLC细胞EGFR-TKIs获得性耐药发展中的作用。我们的结果表明,CCL2在EGFR-TKIs耐药的NSCLC细胞中异常上调,CCL2过表达显着降低了对EGFR-TKIs的敏感性。相反,CCL2合成抑制剂抑制CCL2,bindarit,或CCL2敲低可以逆转这种阻力。CCL2上调还可导致EGFR-TKI耐药NSCLC细胞的迁移增强和上皮间质转化(EMT)标志物表达增加,也可以通过CCL2敲低或抑制来挽救。此外,我们的研究结果表明,CCL2依赖性EGFR-TKIs耐药涉及AKT-EMT信号通路;抑制该通路可有效减弱CCL2诱导的细胞迁移和EMT标志物表达.总之,CCL2促进获得性EGFR-TKIs耐药性和EMT的发展,同时激活NSCLC中的AKT信号传导。这些见解为开发CCL2靶向疗法提供了一个有希望的途径,该疗法可预防NSCLC中的EGFR-TKIs耐药。
    Acquired resistance to EGFR tyrosine kinase inhibitors (EGFR-TKIs) represents a primary cause of treatment failure in non-small cell lung cancer (NSCLC) patients. Chemokine (C-C motif) ligand 2 (CCL2) is recently found to play a pivotal role in determining anti-cancer treatment response. However, the role and mechanism of CCL2 in the development of EGFR-TKIs resistance have not been fully elucidated. In the present study, we focus on the function of CCL2 in the development of acquired resistance to EGFR-TKIs in NSCLC cells. Our results show that CCL2 is aberrantly upregulated in EGFR-TKIs-resistant NSCLC cells and that CCL2 overexpression significantly diminishes sensitivity to EGFR-TKIs. Conversely, CCL2 suppression by CCL2 synthesis inhibitor, bindarit, or CCL2 knockdown can reverse this resistance. CCL2 upregulation can also lead to enhanced migration and increased expressions of epithelial-mesenchymal transition (EMT) markers in EGFR-TKI-resistant NSCLC cells, which could also be rescued by CCL2 knockdown or inhibition. Furthermore, our findings suggest that CCL2-dependent EGFR-TKIs resistance involves the AKT-EMT signaling pathway; inhibition of this pathway effectively attenuates CCL2-induced cell migration and EMT marker expression. In summary, CCL2 promotes the development of acquired EGFR-TKIs resistance and EMT while activating AKT signaling in NSCLC. These insights suggest a promising avenue for the development of CCL2-targeted therapies that prevent EGFR-TKIs resistance in NSCLC.
    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: Letter
    暂无摘要。
    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
    Treatment with epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) has brought significant benefits to non-small cell lung cancer (NSCLC) patients with EGFR mutations. However, most patients eventually develop acquired resistance after treatment. This study investigated the epigenetic effects of mucin 17 (MUC17) in acquired drug-resistant cells of EGFR-TKIs. We found that GR/OR (gefitinib/osimertinib-resistance) cells enhance genome-wide DNA hypermethylation, mainly in 5-UTR associated with multiple oncogenic pathways, in which GR/OR cells exerted a pro-oncogenic effect by downregulating mucin 17 (MUC17) expression in a dose- and time-dependent manner. Gefitinib/osimertinib acquired resistance mediated down-regulation of MUC17 by promoting DNMT1/UHRF1 complex-dependent promoter methylation, thereby activating NF-κB activity. MUC17 increased the generation of IκB-α and inhibit NF-κB activity by promoting the expression of MZF1. In vivo results also showed that DNMT1 inhibitor (5-Aza) in combination with gefitinib/osimertinib restored sensitivity to OR/GR cells. Acquired drug resistance of gefitinib/osimertinib promoted UHRF1/DNMT1 complex to inhibit the expression of MUC17. MUC17 in GR/OR cells may act as an epigenetic sensor for biomonitoring the resistance to EGFR-TKIs.
    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
    丙酮酸激酶2(PKM2)和丙酮酸脱氢酶激酶1(PDK1)是肿瘤糖代谢途径中的两个关键酶,不仅通过加速有氧糖酵解促进肿瘤的生长和增殖,但也有助于非小细胞肺癌(NSCLC)的耐药性。考虑到仅靶向PKM2或PDK1似乎不足以重塑异常的葡萄糖代谢以实现显著的抗肿瘤活性,我们提出了同步调节PKM2和PDK1的“两步法”。首先,我们发现ML265(PKM2激活剂)和AZD7545(PDK1抑制剂)的组合可以协同抑制H1299细胞的增殖并诱导凋亡。基于此,我们设计了一系列新型紫草素(SK)硫醚衍生物作为PKM2/PDK1双靶点药物,其中以苯环上2-甲基取代为特征的最有效的化合物E5对EGFR突变NSCLC细胞H1975的抑制活性显着增加(IC50=1.51μmol/L),其活性比先导化合物SK(IC50=4.56μmol/L)和阳性对照吉非替尼(IC50=25.56μmol/L)高3倍和17倍,分别。此外,E5在异种移植小鼠模型中也显示出良好的抗肿瘤活性,毒性副作用明显低于SK。此外,E5还抑制PKM2进入细胞核以调节癌基因的转录激活,从而恢复H1975细胞对吉非替尼的敏感性。总的来说,这些数据表明,PKM2/PDK1的双重抑制剂E5可能是吉非替尼治疗EGFR-TKIs耐药NSCLC的有希望的辅助药物,值得进一步调查。
    Pyruvate kinase 2 (PKM2) and pyruvate dehydrogenase kinase 1 (PDK1) are two key enzymes in tumor glucose metabolism pathway that not only promote tumor growth and proliferation through accelerating aerobic glycolysis, but also contribute to drug resistance of non-small cell lung cancer (NSCLC). Considering that targeting PKM2 or PDK1 alone seems insufficient to remodel abnormal glucose metabolism to achieve significant antitumor activity, we proposed a \"two-step approach\" that regulates PKM2 and PDK1 synchronously. Firstly, we found that the combination of ML265 (PKM2 activator) and AZD7545 (PDK1 inhibitor) could synergistically inhibit proliferation and induce apoptosis in H1299 cells. Base on this, we designed a series of novel shikonin (SK) thioether derivatives as PKM2/PDK1 dual-target agents, among which the most potent compound E5 featuring a 2-methyl substitution on the benzene ring exerted significantly increased inhibitory activity toward EGFR mutant NSCLC cell H1975 (IC50 = 1.51 μmol/L), which was 3 and 17-fold more active than the lead compound SK (IC50 = 4.56 μmol/L) and the positive control gefitinib (IC50 = 25.56 μmol/L), respectively. Additionally, E5 also showed good anti-tumor activity in xenografted mouse models, with significantly lower toxicity side effects than SK. Moreover, E5 also inhibited the entry of PKM2 into nucleus to regulate the transcriptional activation of oncogenes, thus restoring the sensitivity of H1975 cell to gefitinib. Collectively, these data demonstrate that E5, a dual inhibitor of PKM2/PDK1, may be a promising adjunct to gefitinib in the treatment of EGFR-TKIs resistant NSCLC, deserving further investigation.
    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
    有利的临床证据表明,晚期非小细胞肺癌(NSCLC)新疗法的下一个趋势将是联合疗法。然而,不可避免的表皮生长因子受体-酪氨酸激酶抑制剂(EGFR-TKI)耐药极大地限制了携带EGFR激活突变体的患者的临床疗效。在这项研究中,我们发现一名临床奥希替尼耐药患者在奥希替尼联合安洛替尼治疗后恢复了阳性反应.构建了两种奥希替尼耐药细胞系,和AXL在NSCLC细胞系中赋予奥希替尼耐药性。在两种奥希替尼耐药的NSCLC细胞系和异种移植物中,安洛替尼和奥希替尼的联合作用恢复了对奥希替尼的敏感性。此外,安洛替尼抑制两种抗性细胞系中AXL的磷酸化。机械上,我们证实MYC与AXL的启动子结合以促进其在NSCLC细胞中的转录,我们证明,安洛替尼联合奥希替尼治疗可通过使c-MET/MYC/AXL轴失活来逆转NSCLC中的奥希替尼耐药,从而增强抗肿瘤作用.总之,我们的研究结果有力地支持了这种联合治疗可以有效提高晚期NSCLC患者的治疗效果.
    Favorable clinical evidence suggests that the next trend in new treatments for advanced non-small cell lung cancer (NSCLC) will be combination therapies. However, inevitable epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) resistance greatly limits the clinical efficacy of patients carrying EGFR-activating mutants. In this study, we found a patient with clinical osimertinib resistance who regained a positive response after osimertinib plus anlotinib treatment. Two osimertinib-resistant cell lines were constructed, and AXL conferred resistance to osimertinib in NSCLC cell lines. The combined effects of anlotinib and osimertinib restored sensitivity to osimertinib in two osimertinib-resistant NSCLC cell lines and in xenografts. Moreover, anlotinib inhibits the phosphorylation of AXL in both resistant cell lines. Mechanistically, we confirmed that MYC binds to the promoter of AXL to promote its transcription in NSCLC cells, and we demonstrated that anlotinib combined with osimertinib treatment enhances the anti-tumor effect by inactivating the c-MET/MYC/AXL axis to reverse osimertinib resistance in NSCLC. In conclusion, our results provide strong support that this combination therapy may be effective in enhancing the efficacy of treatments in patients with advanced NSCLC.
    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:我们研究了影响TP53突变的因素对EGFR-酪氨酸激酶抑制剂疗效和潜在治疗策略的影响。
    方法:收集肿瘤样本,通过下一代测序筛选基因突变,以及患者的基线特征。基于每个随访时间点的间期计算机断层扫描来评估对TKI治疗的总体反应。Fisher精确检验和对数秩检验用于确定本研究中的统计学差异。
    结果:共收集了1134例NSCLC患者的临床样本,在644例中发现了TP53mut,在622例中发现了EGFRmut。TP53mut的低频率或EGFR共突变率超过50%与TKI治疗患者的预后相关。此外,DB域以外区域的TP53mut与TKI抗性的相关性最强,而DB域中的各种类型的突变仅对PFS有影响。一项基于EGFR-TKI治疗的分组研究显示,EGFR-TKIs联合化疗与TP53mut预后患者的生存获益更显著相关,而EGFR-TKI治疗对TP53wt患者有利。此外,TP53mut可以缩短患者术后复发的时间,他们也可能对化疗后的EGFR-TKIs反应良好。
    结论:TP53mut的各种特征不同程度地影响TKI治疗患者的预后。EGFR-TKIs联合化疗对预后TP53mut患者的生存有益,为EGFRmut患者的治疗管理提供重要参考。
    We investigated the impact of factors that influence TP53 mutations on the efficacy of EGFR-tyrosine kinase inhibitors and potential treatment strategies.
    Tumor samples were collected to screen gene mutations by next-generation sequencing, as well as the patients\' baseline characteristics. The overall response to treatment with TKIs was evaluated based on interval computed tomography scans at each follow-up time point. A Fisher\'s exact test and log-rank test were used to determine the statistical differences in this study.
    A total of 1134 clinical samples were collected from NSCLC patients, and TP53mut was identified in 644 cases and EGFRmut in 622 cases. A low frequency of TP53mut or more than 50% EGFR co-mutation rate were related to the prognosis of TKI-treated patients. In addition, TP53mut in the region outside of the DB domain had the strongest correlation with TKI resistance, whereas various types of mutations in the DB domain only had an impact on PFS. A grouping study of EGFR-TKI-based treatment revealed that EGFR-TKIs with chemotherapy were associated with more significant survival benefits for patients with prognostic TP53mut, whereas EGFR-TKI therapy was favorable for TP53wt patients. Furthermore, TP53mut could shorten the time to the relapse of postoperative patients, who will also likely respond well to EGFR-TKIs with chemotherapy.
    Various characteristics of TP53mut affect the prognosis of TKI-treated patients to varying degrees. EGFR-TKIs with chemotherapy were benefit for patients\' survival with prognostic TP53mut, which provides an important reference for treatment management of EGFRmut patients.
    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
    The follow-up treatment of patients with advanced non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) mutation after drug resistance to EGFR-tyrosine kinase inhibitors (TKIs) have become a hotspot and difficulty at present. Immune checkpoint inhibitors (ICIs) therapy is a new and important choice for these patients, but many studies have shown unsatisfactory efficacy. However, some domestic and foreign studies have shown that ICIs combination therapy is still effective in some patients with positive driver genes and drug resistance after targeted therapy. So, in the era of immunotherapy, what are the differences in the efficacy of different combination immunotherapy strategies for different patients? What are the factors that affect efficacy? What are the interrelationships between these factors and other immunotherapy efficacy prediction biomarkers? All these problems have broad and important research value.
.
    【中文题目:EGFR突变NSCLC患者TKIs耐药后
ICIs治疗进展】 【中文摘要:表皮生长因子受体酪氨酸激酶抑制剂(epidermal growth factor receptor-tyrosine kinase inhibitors, EGFR-TKIs)耐药后的EGFR突变晚期非小细胞肺癌(non-small cell lung cancer, NSCLC)患者的后续治疗已成为目前的热点和难点。免疫检查点抑制剂(immune checkpoint inhibitors, ICIs)治疗对于这部分患者来说是一个崭新而重要的选择,然而众多研究却显示疗效差强人意。但同时,国内外也有研究表明部分驱动基因阳性且靶向治疗耐药后的患者接受ICIs联合治疗仍显效。那么,在免疫治疗时代,不同的患者接受不同的ICIs联合策略的疗效有何差异?影响疗效的因素有哪些?这些因素与其他免疫治疗疗效预测标志物的内在联系有哪些?这些问题都有着广阔而重要的研究价值。
】 【中文关键词:EGFR突变;EGFR-TKIs耐药;肺肿瘤;免疫检查点抑制剂;免疫治疗】.
    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
    表皮生长因子受体酪氨酸激酶抑制剂(EGFR-TKIs)的使用为具有EGFR突变的晚期NSCLC患者的生存带来了显着的益处。不幸的是,获得性耐药似乎是不可避免的,限制了EGFR-TKIs在临床实践中的应用。这项研究报道了维持耐药性的共同分子机制和克服EGFR-TKIs耐药性的潜在治疗选择。
    建立抗EGFR-TKIs的NSCLC细胞,并通过MTT测定证实。在体内检测胆固醇含量并确定胆固醇对NSCLC生长的促进作用。然后,我们确定ERRα表达是胆固醇介导的耐药的下游因子.剖析监管机制,我们做了实验,包括免疫荧光,免疫共沉淀,荧光素酶报告基因测定和染色质免疫沉淀测定。
    长期暴露于EGFR-TKIs会产生耐药性,其特征是胆固醇在脂筏中积累,促进EGFR和Src相互作用并引导EGFR/Src/Erk信号再激活介导的SP1核易位和ERRα再表达。进一步的研究确定ERRα为SP1的靶基因。功能上,ERRα的再表达通过调节ROS解毒过程来维持细胞增殖。洛伐他汀,一种用来降低胆固醇水平的药物,和ERRα的反向激动剂XCT790,在体外和体内克服吉非替尼和奥希替尼耐药。
    我们的研究表明,胆固醇/EGFR/Src/Erk/SP1轴诱导的ERRα再表达可促进吉非替尼和奥希替尼耐药癌细胞的存活。此外,我们证明了降低胆固醇和下调ERRα作为NSCLC有效辅助治疗的潜力.
    The use of epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) brings remarkable benefits for the survival of patients with advanced NSCLC harboring EGFR mutations. Unfortunately, acquired resistance seems to be inevitable and limits the application of EGFR-TKIs in clinical practice. This study reported a common molecular mechanism sustaining resistance and potential treatment options to overcome EGFR-TKIs resistance.
    EGFR-TKIs resistant NSCLC cells were established and confirmed by MTT assay. Cholesterol content was detected and the promotional function of cholesterol on NSCLC growth was determined in vivo. Then, we identified ERRα expression as the downstream factor of cholesterol-mediated drug resistance. To dissect the regulatory mechanism, we conducted experiments, including immunofluorescence, co-immunoprecipitation, luciferase reporter assay and chromatin immunoprecipitation assay.
    Long-term exposure to EGFR-TKIs generate drug resistance with the characteristic of cholesterol accumulation in lipid rafts, which promotes EGFR and Src to interact and lead EGFR/Src/Erk signaling reactivation-mediated SP1 nuclear translocation and ERRα re-expression. Further investigation identifies ERRα as a target gene of SP1. Functionally, re-expression of ERRα sustains cell proliferation by regulating ROS detoxification process. Lovastatin, a drug used to decrease cholesterol level, and XCT790, an inverse agonist of ERRα, overcome gefitinib and osimertinib resistance both in vitro and in vivo.
    Our study indicates that cholesterol/EGFR/Src/Erk/SP1 axis-induced ERRα re-expression promotes survival of gefitinib and osimertinib-resistant cancer cells. Besides, we demonstrate the potential of lowing cholesterol and downregulation of ERRα as effective adjuvant treatment of NSCLC.
    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
    Exosomes are messengers for intercellular communication and signal transduction. Circular RNA (circRNA) abnormal expression and regulation are involved in the occurrence and development of a variety of tumors. In the present study, exosomes in the serum of five patients with non-small cell lung cancer (NSCLC) were isolated before and after EGFR-TKIs resistance, and the circRNA expression profile was screened using a circRNA microarray. The effects of the exosome circRNA_102481 on cell proliferation and apoptosis were analyzed. The interaction between miR-30a-5p and circRNA_102481 or ROR1 was predicted by starBase software, and was confirmed by RNA pull-down and dual-luciferase reporter assays. The results showed that exosomes containing circRNA_102481 were significantly up-regulated in NSCLC with EGFR-TKIs resistance (p<0.05), and that circRNA_102481 was mainly secreted by EGFR-TKIs resistance cell via exosomes (p<0.05). Both circRNA_102481 silencing and si-circRNA_102481 transported by exosomes could inhibit EGFR-TKIs resistance cell proliferation and promote cell apoptosis and circRNA_102481 overexpression could promote EGFR-TKIs sensitive cell proliferation and inhibit cell apoptosis in vitro (p<0.05). CircRNA_102481 served as a miR-30a-5p sponge to regulate ROR1 expression (p<0.05). Furthermore, the expression of circRNA_102481 in exosomes was associated with TNM stage, tumor differentiation status, brain metastasis, and PFS and OS duration. Therefore, it was concluded that tumor-derived exosomal circRNA_ 102481 could contribute to EGFR-TKIs resistance via the microRNA-30a-5p/ROR1 axis in NSCLC. Exosomal circRNA_102481 may serve as a novel diagnostic biomarker and a therapeutic target for EGFR-TKIs resistance in NSCLC.
    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
    表皮生长因子受体-酪氨酸激酶抑制剂(EGFR-TKIs)在非小细胞肺癌(NSCLC)治疗中取得了满意的临床疗效,但获得性耐药性限制了其临床应用。已显示NRF2增强对放疗和某些化疗诱导的细胞凋亡的抗性。在这项研究中,我们研究了NRF2在EGFR-TKIs耐药中的作用.我们表明,由于NRF2蛋白的缓慢降解,一组EGFR-TKI抗性NSCLC细胞系中NRF2蛋白水平显着增加。NRF2敲除克服了HCC827ER和HCC827GR细胞对EGFR-TKIs的抗性。此外,我们证明NRF2通过上调GPX4和SOD2在HCC827细胞中赋予EGFR-TKIs耐药性,而抑制GPX4和SOD2逆转了EGFR-TKIs耐药性.因此,我们得出结论,靶向NRF2-GPX4/SOD2途径是克服EGFR-TKIs耐药的潜在策略.
    Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) have achieved satisfactory clinical effects in the therapy of non-small cell lung cancer (NSCLC), but acquired resistance limits their clinical application. NRF2 has been shown to enhance the resistance to apoptosis induced by radiotherapy and some chemotherapy. In this study, we investigated the role of NRF2 in resistance to EGFR-TKIs. We showed that NRF2 protein levels were markedly increased in a panel of EGFR-TKI-resistant NSCLC cell lines due to slow degradation of NRF2 protein. NRF2 knockdown overcame the resistance to EGFR-TKIs in HCC827ER and HCC827GR cells. Furthermore, we demonstrated that NRF2 imparted EGFR-TKIs resistance in HCC827 cells via upregulation of GPX4 and SOD2, and suppression of GPX4 and SOD2 reversed resistance to EGFR-TKIs. Thus, we conclude that targeting NRF2-GPX4/SOD2 pathway is a potential strategy for overcoming resistance to EGFR-TKIs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号