epidermal growth factor receptor

表皮生长因子受体
  • 文章类型: Journal Article
    BACKGROUND: EGFR tyrosine kinase inhibitors (TKIs) are the standard therapy for non-small-cell lung cancer (NSCLC) patients with EGFR-activating mutations in the first-line setting. Despite initial efficacy, resistance to EGFR-TKIs often develops, and platinum-based chemotherapy is the predominant subsequent treatment. For this study, we aimed to identify prognostic factors for overall survival (OS) and progression-free survival (PFS) among advanced EGFR-mutant NSCLC patients receiving platinum-pemetrexed after progression on EGFR-TKIs. Our analysis specifically focuses on 1st-line treatments limited to 1st- or 2nd-generation EGFR-TKIs, while not restricting later-line treatments involving osimertinib prior to chemotherapy.
    METHODS: From 2012 to 2017, 363 patients who received first-line treatment with first- or second-generation EGFR-TKIs, including gefitinib, erlotinib, and afatinib were enrolled. Some patients received different EGFR-TKIs, including osimertinib, as later-line treatment before platinum-pemetrexed.
    RESULTS: Median OS from the initiation of platinum-pemetrexed was 22.0 months and median PFS with platinum-pemetrexed was 6.2 months. In the multivariate Cox model, we identified three independent prognostic factors for better OS: postoperative recurrence (HR: 0.34, p = 0.004), first-line EGFR-TKI PFS ≥12 months (HR: 0.54, p = 0.002), and osimertinib treatment after platinum-pemetrexed (HR: 0.56, p = 0.005) while BMI <18.5 indicated poor prognosis (HR:1.76, p = 0.049). No statistically significant independent prognostic factors for PFS were found. Receiving osimertinib before platinum-pemetrexed treatment did not impact PFS with platinum-pemetrexed treatment (HR: 1.11, p = 0.64).
    CONCLUSIONS: Postoperative recurrence, first-line EGFR-TKI PFS ≥12 months and osimertinib treatment after platinum-pemetrexed predicted better OS, while BMI <18.5 predicted worse OS. Osimertinib treatment before platinum-pemetrexed treatment did not affect the efficacy of platinum-pemetrexed.
    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
    基于蛋白质的疗法,包括抗体和抗体样蛋白,由于其与小分子药物相比具有较高的特异性,因此越来越引起人们的关注。Gγ招募系统,用于检测蛋白质-蛋白质相互作用的体内酵母双杂交系统之一,以前是使用酵母信号转导机制开发的。在这项研究中,我们修改了Gγ募集系统,以筛选能有效结合表皮生长因子受体L858R突变体(cytoeEGFRL858R)胞内结构域的蛋白突变体.使用修改后的平台,我们对生长因子受体结合蛋白2(Grb2)及其仅包含Src同源性2(SH2)域的截短变体进行了体内定向进化,成功地鉴定了几种突变体,这些突变体比它们的亲本蛋白质更强烈地结合到cytoEGFRL858R。其中一些含有新的有益突变(F108Y和Q144H),并在体外与重组胞浆磷酸化EGFR特异性结合,突出了进化平台的实用性。
    Protein-based therapeutics, including antibodies and antibody-like-proteins, have increasingly attracted attention due to their high specificity compared to small-molecular drugs. The Gγ recruitment system, one of the in vivo yeast two-hybrid systems for detecting protein-protein interactions, has been previously developed using yeast signal transduction machinery. In this study, we modified the Gγ recruitment system to screen the protein mutants that efficiently bind to the intracellular domain of the epidermal growth factor receptor L858R mutant (cytoEGFRL858R). Using the modified platform, we performed in vivo directed evolution for growth factor receptor-bound protein 2 (Grb2) and its truncated variant containing only the Src-homology 2 (SH2) domain, successfully identifying several mutants that more strongly bound to cytoEGFRL858R than their parental proteins. Some of them contained novel beneficial mutations (F108Y and Q144H) and specifically bound to the recombinant cytosolic phosphorylated EGFR in vitro, highlighting the utility of the evolutionary platform.
    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:一些接受奥希替尼治疗的患者出现进行性疾病。目的是阐明奥希替尼耐药的潜在机制。
    方法:ELUCIDATOR:多中心,prospective,观察性研究包括接受奥希替尼治疗的未接受化疗的晚期非小细胞肺癌患者.癌症相关基因的突变,通过循环肿瘤脱氧核糖核酸样品的超灵敏下一代测序检测,在基线和进行性疾病检测后收集。比较这些配对的血浆样品。
    结果:纳入188例患者(2019年5月至2021年1月),178(119名女性[67%])中位年龄74岁,包括在内。患者,n=95(53%)存在表皮生长因子受体外显子19缺失突变。在115名进行性疾病患者中,分析了85例患者的循环肿瘤脱氧核糖核酸水平。MET扩增(n=4),TP53突变(n=4),PIK3CA突变(n=3),BRINP3突变(n=2),BRAF突变(n=2),APC突变(n=1),RET突变(n=1)和表皮生长因子受体(EGFR)耐药突变,检测到C797S(n=1)。基线TP53突变的患者,MET或EGFR扩增的无进展生存期(PFS)和总生存期较短.PIK3CA突变患者PFS较短。
    结论:MET扩增和PIK3CA突变机制在患者中对奥希替尼耐药不足。基线时共存突变或扩增的患者PFS和总生存期较短。
    BACKGROUND: Several patients treated with osimertinib experience progressive disease. The aim was to clarify the mechanisms underlying resistance to osimertinib.
    METHODS: ELUCIDATOR: A multi-centre, prospective, observational study involved chemotherapy-naive patients with advanced non-small cell lung cancer receiving osimertinib. Mutations in cancer-associated genes, detected via ultrasensitive next-generation sequencing of circulating tumour deoxyribonucleic acid samples, were collected at baseline and after progressive disease detection. These paired plasma samples were compared.
    RESULTS: Of 188 patients enrolled (May 2019-January 2021), 178 (119 females [67 %]) median age 74 years, were included. Patients, n = 95 (53 %) had epidermal growth factor receptor exon 19 deletion mutations. Among 115 patients with progressive disease, circulating tumour deoxyribonucleic acid levels of 85 patients were analysed. MET amplification (n = 4), TP53 mutations (n = 4), PIK3CA mutations (n = 3), BRINP3 mutation (n = 2), BRAF mutation (n = 2), APC mutation (n = 1), RET mutation (n = 1) and epidermal growth factor receptor (EGFR) resistance mutation, and C797S (n = 1) were detected. Patients with baseline TP53 mutations, with MET or EGFR amplification had shorter progression-free (PFS) and overall survival. Patients with PIK3CA mutations tended to shorter PFS.
    CONCLUSIONS: MET amplification and PIK3CA mutation mechanisms underly resistance to osimertinib in patients. Patients with coexisting mutations or amplifications at baseline had shorter PFS and overall survival.
    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
    EGFR突变的非小细胞肺癌(NSCLC)患者对免疫检查点抑制剂(ICIs)的反应较差。据报道,EGFR突变的NSCLC中CD8+T细胞的数量减少。然而,不同CD8+T细胞群体的异质性和效应子功能的程度尚未得到深入研究.此外,目前缺乏研究放疗和ICIs联合治疗是否能提高EGFR突变肺癌ICIs疗效的研究.使用单细胞RNA测序(scRNA-seq)来研究EGFR突变的NSCLC中CD8+T细胞群体的异质性。在EGFR突变和野生型细胞的低分割辐射后探索STING途径。将携带LLC-19del和LLC-EGFR肿瘤的小鼠用放射疗法加抗PD-L1治疗。scRNA-seq数据显示,在EGFR突变的NSCLC中,祖细胞耗尽的CD8+T细胞的百分比较低。此外,EGFR突变的NSCLC中的CD8+T细胞富集氧化磷酸化。在EGFR突变和野生型细胞中,8Gy×3增加募集T细胞并激活cGAS-STING途径的趋化因子的表达。在LLC-19del和LLC-EGFR小鼠模型中,放疗和抗PD-L1联合使用可显著抑制腹腔镜肿瘤的生长。增强的远视效应与全身性CD8T细胞浸润有关。这项研究为EGFR突变的NSCLC中CD8+T细胞的异质性和效应子功能提供了深入的理解。我们表明,大分割辐射和抗PD-L1的组合通过激活小鼠中的CD8T细胞显着增强了EGFR突变和野生型肺癌的远视反应。
    Patients with EGFR-mutated non-small cell lung cancer (NSCLC) respond poorly to immune checkpoint inhibitors (ICIs). It has been reported that the number of CD8+T cells is reduced in EGFR-mutated NSCLC. However, the extent of heterogeneity and effector function of distinct populations of CD8+T cells has not been investigated intensively. In addition, studies investigating whether a combination of radiotherapy and ICIs can improve the efficacy of ICIs in EGFR-mutated lung cancer are lacking. Single-cell RNA sequencing (scRNA-seq) was used to investigate the heterogeneity of CD8+T cell populations in EGFR-mutated NSCLC. The STING pathway was explored after hypofractionated radiation of EGFR-mutated and wild-type cells. Mice bearing LLC-19del and LLC-EGFR tumors were treated with radiotherapy plus anti-PD-L1. The scRNA-seq data showed the percentage of progenitor exhausted CD8+T cells was lower in EGFR-mutated NSCLC. In addition, CD8+T cells in EGFR-mutated NSCLC were enriched in oxidative phosphorylation. In EGFR-mutated and wild-type cells, 8 Gy × 3 increased the expression of chemokines that recruit T cells and activate the cGAS-STING pathway. In the LLC-19del and LLC-EGFR mouse model, the combination of radiation and anti-PD-L1 significantly inhibited the growth of abscopal tumors. The enhanced abscopal effect was associated with systemic CD8+T cell infiltration. This study provided an intensive understanding of the heterogeneity and effector functions of CD8+T cells in EGFR-mutated NSCLC. We showed that the combination of hypofractionated radiation and anti-PD-L1 significantly enhanced the abscopal responses in both EGFR-mutated and wild-type lung cancer by activating CD8+T cells in mice.
    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
    目的:评估基于CT(CT)的影像组学结合临床遗传特征预测III/IV期表皮生长因子受体(EGFR)突变的非小细胞肺癌(NSCLC)患者脑转移的价值。
    方法:本研究纳入了147名符合条件的患者,他们在2018年1月至2021年5月期间在我们机构接受治疗。将患者随机分为两组进行模型训练(n=102)和验证(n=45)。治疗前从胸部CT图像中提取影像组学特征,并使用最小绝对收缩和选择算子回归构建影像组学签名。Kaplan-Meier生存分析用于描述无脑转移生存(BM-FS)风险的差异。使用Cox回归分析建立了临床遗传模型。Radiomics,遗传,并构建了组合预测模型,并通过一致性指数(C指数)评估其预测性能。
    结果:在训练(p<0.0001)和验证(p=0.0016)队列中,低放射组学评分的患者的BM-FS明显长于高放射组学评分的患者。列线图的C指数,结合了影像组学签名和N阶段,整体阶段,第三代酪氨酸激酶抑制剂治疗,和EGFR突变状态,在训练和验证队列中分别为0.886(95%置信区间[CI]0.823-0.949)和0.811(95%CI0.719-0.903),分别。与单一预测模型相比,组合模型具有更高的辨别力和临床实用性。
    结论:联合放射组学-遗传模型可用于预测EGFR突变的III/IV期NSCLC患者的BM-FS。
    OBJECTIVE: To evaluate the value of computed tomography (CT)-based radiomics combined with clinical-genetic features in predicting brain metastasis in patients with stage III/IV epidermal growth factor receptor (EGFR)-mutant non-small-cell lung cancer (NSCLC).
    METHODS: The study included 147 eligible patients treated at our institution between January 2018 and May 2021. Patients were randomly divided into two cohorts for model training (n = 102) and validation (n = 45). Radiomics features were extracted from the chest CT images before treatment, and a radiomics signature was constructed using the Least Absolute Shrinkage and Selection Operator regression. Kaplan-Meier survival analysis was used to describe the differences in brain metastasis-free survival (BM-FS) risk. A clinical-genetic model was developed using Cox regression analysis. Radiomics, genetic, and combined prediction models were constructed, and their predictive performances were evaluated by the concordance index (C-index).
    RESULTS: Patients with a low radiomics score had significantly longer BM-FS than those with a high radiomics score in both the training (p < 0.0001) and the validation (p = 0.0016) cohorts. The C-indices of the nomogram, which combined the radiomics signature and N stage, overall stage, third-generation tyrosine kinase inhibitor treatment, and EGFR mutation status, were 0.886 (95% confidence interval [CI] 0.823-0.949) and 0.811 (95% CI 0.719-0.903) in the training and validation cohorts, respectively. The combined model achieved a higher discrimination and clinical utility than the single prediction models.
    CONCLUSIONS: The combined radiomics-genetic model could be used to predict BM-FS in stage III/IV NSCLC patients with EGFR mutations.
    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
    丛状神经纤维瘤(PNFs)是与NF1相关的常见且严重的表型,其特征是高致畸率和恶性转化的潜力。PNFs的生长和复发归因于Nf1缺陷的施万细胞的异常增殖和迁移。蛋白酪氨酸磷酸酶受体S(PTPRS)被认为通过抑制NF1衍生的恶性周围神经鞘瘤中的EMT过程来调节细胞迁移和侵袭。然而,PTPRS在NF1衍生的PNF中的具体作用仍有待阐明。该研究利用GEO数据库和组织微阵列来说明PNF组织中PTPRS表达的减少,与肿瘤复发有关.此外,PTPRS在Nf1缺陷的雪旺氏细胞系中的下调和过表达导致细胞迁移和EMT过程的变化。此外,RTK法和WB法显示PTPRS敲低可以促进EGFR的表达和磷酸化。通过EGFR敲低和EGFR抑制剂可以实现由雪旺氏细胞中PTPRS水平改变破坏的EMT过程的恢复。此外,EGFR高表达与不良预后显著相关.这些发现强调了PTPRS作为肿瘤抑制因子通过调节EGFR介导的EMT过程在PNF复发中的潜在作用。建议未来临床干预的潜在目标。
    Plexiform neurofibromas (PNFs) are a prevalent and severe phenotype associated with NF1, characterized by a high teratogenic rate and potential for malignant transformation. The growth and recurrence of PNFs are attributed to aberrant proliferation and migration of Nf1-deficient Schwann cells. Protein tyrosine phosphatase receptor S (PTPRS) is believed to modulate cell migration and invasion by inhibiting the EMT process in NF1-derived malignant peripheral nerve sheath tumors. Nevertheless, the specific role of PTPRS in NF1-derived PNFs remains to be elucidated. The study utilized the GEO database and tissue microarray to illustrate a decrease in PTPRS expression in PNF tissues, linked to tumor recurrence. Furthermore, the down- and over-expression of PTPRS in Nf1-deficient Schwann cell lines resulted in the changes of cell migration and EMT processes. Additionally, RTK assay and WB showed that PTPRS knockdown can promote EGFR expression and phosphorylation. The restoration of EMT processes disrupted by alterations in PTPRS levels in Schwann cells can be achieved through EGFR knockdown and EGFR inhibitor. Moreover, high EGFR expression has been significantly correlated with poor prognosis. These findings underscore the potential role of PTPRS as a tumor suppressor in the recurrence of PNF via the regulation of EGFR-mediated EMT processes, suggesting potential targets for future clinical interventions.
    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
    肺癌是癌症相关死亡的主要原因,部分原因是其诊断较晚。表皮生长因子受体(EGFR)在癌细胞中的表达增加与不良预后相关,EGFR酪氨酸激酶抑制剂广泛应用于肿瘤治疗。本研究旨在阐明非小细胞肺癌(NSCLC)患者T细胞EGFR表达与预后的关系。本研究包括40例NSCLC患者和40例健康志愿者。根据其趋化因子受体,用流式细胞仪鉴定外周血CD4辅助T(Th1,Th2,Th9,Th17,Th1Th17,滤泡和外周Th)和细胞毒性T淋巴细胞(CD8滤泡和外周T)亚群。在NSCLC患者中研究了T淋巴细胞上EGFR表达与总生存期(OS)的关系。患者[平均年龄(最小-最大)=64.03(45-83);20期I-III和20期IV]在CD3+T上EGFR表达增加,CD4+Th,与对照相比,Th1、Th2和Th17细胞(p<0.05)。CD3+T的EGFR高表达,CD4+Th,Th1和Th2细胞与不良OS相关。此外,淋巴细胞上的PD-1表达,CD3+T,与对照组相比,NSCLC患者的Th细胞增加。EGFR和PD-1在Th细胞上的高表达,淋巴细胞和Th细胞的百分比降低,特别是在IV期NSCLC患者中,揭示了EGFR活性的增加可能引发Th细胞的凋亡并促进转移的发展,而EGFR在CD3+T上的高表达,CD4+Th,Th1和Th2细胞可能是NSCLC中独立的不良预后标志物。
    Lung cancer is the leading cause of cancer-related deaths, in part due to its late diagnosis. Increased epidermal growth factor receptor (EGFR) expression in cancer cells is associated with a poor prognosis, and EGFR tyrosine kinase inhibitors are widely used in cancer treatment. This study aimed to clarify the relationship between EGFR expression on T cells and cancer prognosis in patients with non-small cell lung cancer (NSCLC). Forty patients with NSCLC and 40 healthy volunteers were included in this study. Peripheral CD4+T helper (Th1, Th2, Th9, Th17, Th1Th17, follicular and peripheral Th) and cytotoxic T lymphocyte (CD8+follicular and peripheral T) subsets were identified with flow cytometry according to their chemokine receptors. EGFR expression on T lymphocytes in relation to overall survival (OS) was investigated in patients with NSCLC. The patients [mean age (min-max) = 64.03 (45-83); 20 stage I-III and 20 stage IV] had increased EGFR expression on CD3+T, CD4+Th, Th1, Th2, and Th17 cells compared to the controls (p < 0.05). High EGFR expression on CD3+T, CD4+Th, Th1, and Th2 cells was associated with poor OS. Also, PD-1 expression on lymphocytes, CD3+T, and Th cells was increased in patients with NSCLC compared to controls. The high expression of EGFR and PD-1 on Th cells and the reduced percentage of lymphocytes and Th cells, especially in stage IV patients with NSCLC, revealed that increased EGFR activity may trigger apoptosis of Th cells and promote the development of metastases, while high EGFR expression on CD3+T, CD4+Th, Th1, and Th2 cells may be an independent poor prognostic marker 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: Case Reports
    背景:表皮生长因子受体(EGFR)突变和c-ros癌基因1(ROS1)重排是非小细胞肺癌(NSCLC)的关键遗传改变和预测性肿瘤标志物,通常被认为是相互排斥的。EGFR/ROS1共突变是一种罕见事件,这种情况的标准治疗方法仍然模棱两可。
    方法:这里,我们报道了一名64岁女性诊断为肺腺癌的病例,伴随EGFRL858R突变和ROS1重排。患者在手术后接受了两个周期的化疗,但是疾病进展了。吉非替尼治疗1个月后,疾病再次进展。然而,改用克唑替尼后,病变变得稳定。目前,克唑替尼已超过53个月,具有显着的治疗效果。
    结论:EGFR酪氨酸激酶抑制剂和克唑替尼在EGFR/ROS1共突变的NSCLC患者中的疗效差异很大。该报告将有助于此类患者的未来治疗。
    BACKGROUND: Epidermal growth factor receptor (EGFR) mutation and c-ros oncogene 1 (ROS1) rearrangement are key genetic alterations and predictive tumor markers for non-small cell lung cancer (NSCLC) and are typically considered to be mutually exclusive. EGFR/ROS1 co-mutation is a rare event, and the standard treatment approach for such cases is still equivocal.
    METHODS: Herein, we report the case of a 64-year-old woman diagnosed with lung adenocarcinoma, with concomitant EGFR L858R mutation and ROS1 rearrangement. The patient received two cycles of chemotherapy after surgery, but the disease progressed. Following 1-month treatment with gefitinib, the disease progressed again. However, after switching to crizotinib, the lesion became stable. Currently, crizotinib has been administered for over 53 months with a remarkable treatment effect.
    CONCLUSIONS: The efficacy of EGFR tyrosine kinase inhibitors and crizotinib was vastly different in this NSCLC patient with EGFR/ROS1 co-mutation. This report will aid future treatment of such patients.
    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(EGFR)是一种跨膜酪氨酸激酶,通常通过翻译后糖基化进行修饰。在癌症中,在很大一部分非小细胞肺癌和乳腺腺癌中检测到EGFR扩增和促进增殖的热点突变如L858R。分子动力学模拟表明,天冬酰胺残基361(N361)处的糖基化促进二聚化和配体结合。我们稳定表达糖基化缺陷突变EGFRN361A,有或没有致癌突变L858R。免疫荧光和流式细胞术证明突变体各自在细胞膜上良好表达。相对于野生型EGFR,N361A降低增殖以及对配体的敏感性降低。测量EGFR与其结合配偶体HER2在细胞中的共定位的邻近连接测定揭示N361A突变增加共定位。N361A,位于EGFR抑制剂necitumumab的结合界面附近,表达致癌EGFRL858R的脱敏细胞对基于抗体的抑制。这些发现强调了翻译后修饰对癌基因功能的关键相关性。
    结论:EGFR将生长因子的信号传导到细胞增殖中,并且在肿瘤中经常被过度激活。N361的EGFR糖基化调节EGFR二聚化,增殖信号的生长因子刺激,和对靶向抑制的敏感性。对EGFR糖基化的见解可能会扩大治疗机会,使癌症患者受益。
    Epidermal growth factor receptor (EGFR) is a transmembrane tyrosine kinase that is frequently modified by glycosylation post-translationally. In cancer, EGFR amplifications and hotspot mutations such as L858R that promote proliferation have been detected in a significant fraction of non-small cell lung carcinomas and breast adenocarcinomas. Molecular dynamic simulations suggested that glycosylation at asparagine residue 361 (N361) promotes dimerization and ligand binding. We stably expressed glycosylation-deficient mutant EGFR N361A, with or without the oncogenic mutation L858R. Immunofluorescence and flow cytometry demonstrated that the mutants were each well expressed at the cell membrane. N361A decreased proliferation relative to wild-type EGFR as well as decreased sensitivity to ligands. Proximity ligation assays measuring co-localization of EGFR with its binding partner HER2 in cells revealed that N361A mutations increased co-localization. N361A, located near the binding interface for the EGFR inhibitor necitumumab, desensitized cells expressing the oncogenic EGFR L858R to antibody-based inhibition. These findings underline the critical relevance of post-translational modifications on oncogene function.
    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)靶向治疗的出现,包括表皮生长因子受体(EGFR)酪氨酸激酶抑制剂,增加了对健壮的伴随诊断测试的需求。如今,通过qPCR和直接DNA测序检测EGFR基因外显子18-21中的可操作变体通常被下一代测序(NGS)取代.在这项研究中,我们评估了靶向NGS在之前通过IVD认证的qPCR检测对DNA参考材料进行分析的临床NSCLC材料中的可药用EGFR变体检测的诊断实用性.我们使用NGS“TruSight肿瘤15”试验(Illumina)和qPCR“cobasEGFR突变测试v2”(罗氏诊断)对59个NSCLC组织和细胞学标本进行了EGFR变异检测。使用具有EGFR变体的已知等位基因频率(VAF)的生物合成和生物DNA参考材料评估靶向NGS测定的灵敏度和特异性。NGS在DNA参考材料中表现出用于诊断应用(VAF<5%)的足够低的检测限;正确地鉴定了所有EGFR变体。NGS在运行之间显示出VAF评估的高可重复性(从0.02到3.98的CV%)。在临床材料中,NGS和qPCR的总体一致性为76.14%(Cohen的Kappa=0.5933)。大多数不一致的结果涉及通过qPCR对EGFR外显子20插入的假阳性检测。在两种测定中,总共59个(15%)临床样品中的9个显示一种或多种EGFR变体的不一致结果。此外,我们观察到TP53是EGFR阳性NSCLC患者中常见的共突变基因.总之,靶向NGS在EGFR变异检测中显示出许多优于qPCR的特征(变异的精确鉴定,等位基因频率的计算,高分析灵敏度),这可能会增强基本诊断报告。
    The emergence of targeted therapies in non-small-cell lung cancer (NSCLC), including inhibitors of epidermal growth factor receptor (EGFR) tyrosine kinase, has increased the need for robust companion diagnostic tests. Nowadays, detection of actionable variants in exons 18-21 of the EGFR gene by qPCR and direct DNA sequencing is often replaced by next-generation sequencing (NGS). In this study, we evaluated the diagnostic usefulness of targeted NGS for druggable EGFR variants testing in clinical NSCLC material previously analyzed by the IVD-certified qPCR test with respect to DNA reference material. We tested 59 NSCLC tissue and cytology specimens for EGFR variants using the NGS \'TruSight Tumor 15\' assay (Illumina) and the qPCR \'cobas EGFR mutation test v2\' (Roche Diagnostics). The sensitivity and specificity of targeted NGS assay were evaluated using the biosynthetic and biological DNA reference material with known allelic frequencies (VAF) of EGFR variants. NGS demonstrated a sufficient lower detection limit for diagnostic applications (VAF < 5%) in DNA reference material; all EGFR variants were correctly identified. NGS showed high repeatability of VAF assessment between runs (CV% from 0.02 to 3.98). In clinical material, the overall concordance between NGS and qPCR was 76.14% (Cohen\'s Kappa = 0.5933). The majority of discordant results concerned false-positive detection of EGFR exon 20 insertions by qPCR. A total of 9 out of 59 (15%) clinical samples showed discordant results for one or more EGFR variants in both assays. Additionally, we observed TP53 to be a frequently co-mutated gene in EGFR-positive NSCLC patients. In conclusion, targeted NGS showed a number of superior features over qPCR in EGFR variant detection (exact identification of variants, calculation of allelic frequency, high analytical sensitivity), which might enhance the basic diagnostic report.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号