EGFR amplification

EGFR 扩增
  • 文章类型: Journal Article
    神经胶质瘤中的EGFR扩增通常定义为EGFR/CEP7比值≥2。在主要参考实验室进行的测试中,一小部分患者有≥5个EGFR和CEP7拷贝,但未通过EGFR/CEP7比值扩增,并被指定为高度多体病例.为了确定这些肿瘤是否与传统定义的EGFR扩增或非扩增的神经胶质瘤更密切相关,一项回顾性研究发现,在1143例(1.9%)神经胶质瘤中,EGFR和CEP7的平均拷贝/细胞≥5个,EGFR/CEP7比值<2,表现出高多体.在这些案件中,4名患者的临床病理数据不足以纳入额外分析,15个是胶质母细胞瘤,2个是IDH突变型星形细胞瘤,1是高级别神经胶质肿瘤,NOS.可用于3例的下一代测序显示一个具有TERT启动子突变,TP53突变在所有情况下,没有EGFR突变或扩增,最接近非扩增病例。放大后的中位总生存时间为42.86、66.07和41.14周,高度多生体,和非扩增,分别,并且没有显着差异(p=0.3410)。高7号染色体多体神经胶质瘤很少见,但我们的数据表明它们可能与非扩增性神经胶质瘤在生物学上相似。
    EGFR amplification in gliomas is commonly defined by an EGFR/CEP7 ratio of ≥2. In testing performed at a major reference laboratory, a small subset of patients had ≥5 copies of both EGFR and CEP7 yet were not amplified by the EGFR/CEP7 ratio and were designated high polysomy cases. To determine whether these tumors are more closely related to traditionally defined EGFR-amplified or nonamplified gliomas, a retrospective search identified 22 out of 1143 (1.9%) gliomas with an average of ≥5 copies/cell of EGFR and CEP7 with an EGFR/CEP7 ratio of <2 displaying high polysomy. Of these cases, 4 had insufficient clinicopathologic data to include in additional analysis, 15 were glioblastomas, 2 were IDH-mutant astrocytomas, and 1 was a high-grade glial neoplasm, NOS. Next-generation sequencing available on 3 cases demonstrated one with a TERT promoter mutation, TP53 mutations in all cases, and no EGFR mutations or amplifications, which most closely matched the nonamplified cases. The median overall survival times were 42.86, 66.07, and 41.14 weeks for amplified, highly polysomic, and nonamplified, respectively, and were not significantly different (p =  0.3410). High chromosome 7 polysomic gliomas are rare but our data suggest that they may be biologically similar to nonamplified gliomas.
    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
    2021年世界卫生组织中枢神经系统肿瘤分类更新了神经胶质瘤亚型和分级系统,并将EGFR扩增(Amp)作为胶质母细胞瘤(GBM)的诊断标志物之一。
    本研究旨在描述频率,基于最新分类的EGFRAmp在弥漫性胶质瘤中的临床价值和分子相关性。
    我们回顾了2011年至2022年在我们医院的神经胶质瘤患者,其中包括187名成人神经胶质瘤患者,有可用的肿瘤组织用于检测EGFRAmp和其他59个感兴趣的分子标志物。临床,根据EGFRAmp在不同胶质瘤亚型中的状态分析影像学和病理学资料。
    163个胶质瘤被分类为成人型弥漫性胶质瘤,星形细胞瘤的数量,少突胶质细胞瘤和GBM分离为41、46和76。EGFRAmp在IDH野生型弥漫性神经胶质瘤(66.0%)和GBM(85.5%)中比IDH突变型弥漫性神经胶质瘤(32.2%)及其亚型(星形细胞瘤,29.3%;少突胶质细胞瘤,34.8%)。EGFRAmp未对IDH突变型弥漫性神经胶质瘤和星形细胞瘤的总生存期(OS)进行分层,虽然与IDH野生型弥漫性神经胶质瘤的OS较差显著相关,组织学2级和3级IDH-野生型弥漫性星形胶质细胞瘤和GBM。
    我们的研究验证了EGFRAmp是GBM的诊断标志物,并且仍然是该组中OS缩短的有用预测因子。
    UNASSIGNED: The 2021 World Health Organization Classification of Central Nervous System Tumors updates glioma subtyping and grading system, and incorporates EGFR amplification (Amp) as one of diagnostic markers for glioblastoma (GBM).
    UNASSIGNED: This study aimed to describe the frequency, clinical value and molecular correlation of EGFR Amp in diffuse gliomas based on the latest classification.
    UNASSIGNED: We reviewed glioma patients between 2011 and 2022 at our hospital, and included 187 adult glioma patients with available tumor tissue for detection of EGFR Amp and other 59 molecular markers of interest. Clinical, radiological and pathological data was analyzed based on the status of EGFR Amp in different glioma subtypes.
    UNASSIGNED: 163 gliomas were classified as adult-type diffuse gliomas, and the number of astrocytoma, oligodendroglioma and GBM was 41, 46, and 76. EGFR Amp was more common in IDH-wildtype diffuse gliomas (66.0%) and GBM (85.5%) than IDH-mutant diffuse gliomas (32.2%) and its subtypes (astrocytoma, 29.3%; oligodendroglioma, 34.8%). EGFR Amp did not stratify overall survival (OS) in IDH-mutant diffuse gliomas and astrocytoma, while was significantly associated with poorer OS in IDH-wildtype diffuse gliomas, histologic grade 2 and 3 IDH-wildtype diffuse astrocytic gliomas and GBM.
    UNASSIGNED: Our study validated EGFR Amp as a diagnostic marker for GBM and still a useful predictor for shortened OS in this group.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    鉴于它们良好的抗肿瘤作用,表皮生长因子受体(EGFR)酪氨酸激酶抑制剂(TKIs)是EGFR敏感突变的标准一线治疗,包括外显子19缺失和外显子21L858R突变。EGFR融合突变和EGFR扩增在非小细胞肺癌(NSCLC)中非常罕见。我们描述了2例EGFR融合突变(EGFR-MACF1和EGFR-GNAT3)合并EGFR扩增的NSCLC患者。两名患者均接受EGFR-TKI治疗,其中1例显示抗肿瘤反应。
    Given their good antitumor effects, epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) are standard first-line therapy for EGFR-sensitive mutations, including exon 19 deletions and exon 21 L858R mutations. EGFR fusion mutations and EGFR amplification are very rare in non-small cell lung cancer (NSCLC). We describe 2 patients with NSCLC harboring EGFR fusion mutations (EGFR-MACF1 and EGFR-GNAT3) combined with EGFR amplification. Both patients received EGFR-TKI treatment, and 1 of them showed an antitumor response.
    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
    目的:2021年,WHO中枢神经系统(CNS)肿瘤分类标准增加了弥漫性星形胶质细胞瘤的诊断,IDH野生型,具有胶质母细胞瘤的分子特征,世卫组织4级(DAG-G)。尽管组织学分级较低,但DAG-G可能表现出胶质母细胞瘤(GBM)的侵袭性和恶性。因此,精确的术前诊断可以帮助神经外科医生制定更精细的个性化治疗计划。本研究旨在建立基于术前MRI影像组学的DAG-G无创鉴别预测模型。
    方法:华山医院经病理证实的脑胶质瘤患者,复旦大学,对2019年9月至2021年7月间的数据进行回顾性分析。此外,利用武汉协和医院和徐州市肿瘤医院的两个外部验证数据集来验证预测模型的可靠性和准确性。使用半自动工具ITK-SNAP(4.0.0版)在患者的术前MRI图像上描绘了两个感兴趣区域(ROI),分别命名为最大异常区域(ROI1)和肿瘤区域(ROI2),应用Pyradiomics3.0进行特征提取。使用最小绝对收缩和选择算子(LASSO)滤波器和Spearman相关系数进行特征选择。六个分类器,包括高斯朴素贝叶斯(GNB),K-最近邻(KNN),随机森林(RF),自适应提升(AB),以及具有线性核和多层感知器(MLP)的支持向量机(SVM),用于构建预测模型,并通过五次交叉验证评估了六个分类器的预测性能。此外,使用曲线下面积(AUC)评估预测模型的性能,精度(PRE),和其他指标。
    结果:根据纳入和排除标准,172例2-3级星形细胞瘤患者最终纳入研究,共有44例患者符合DAG-G的诊断。在DAG-G的预测任务中,GNB分类器的平均AUC为0.74±0.07,KNN分类器为0.89±0.04,RF分类器为0.96±0.03,AB分类器为0.97±0.02,SVM分类器为0.88±0.05,MLP分类器为0.91±0.03,其中,AB分类器取得了最好的预测性能。此外,AB分类器在武汉协和医院和徐州肿瘤医院获得的两个外部验证数据集中获得的AUC分别为0.91和0.89,分别。
    结论:本研究建立的基于术前MRI影像组学的预测模型基本可以实现前瞻性,非侵入性,和DAG-G的准确诊断,这对进一步优化此类患者的治疗计划具有重要意义,包括扩大手术范围和积极进行放射治疗,靶向治疗,或手术后的其他治疗,从根本上最大限度地提高患者的预后。
    OBJECTIVE: In 2021, the WHO central nervous system (CNS) tumor classification criteria added the diagnosis of diffuse astrocytic glioma, IDH wild-type, with molecular features of glioblastoma, WHO grade 4 (DAG-G). DAG-G may exhibit the aggressiveness and malignancy of glioblastoma (GBM) despite the lower histological grade, and thus a precise preoperative diagnosis can help neurosurgeons develop more refined individualized treatment plans. This study aimed to establish a predictive model for the non-invasive identification of DAG-G based on preoperative MRI radiomics.
    METHODS: Patients with pathologically confirmed glioma in Huashan Hospital, Fudan University, between September 2019 and July 2021 were retrospectively analyzed. Furthermore, two external validation datasets from Wuhan Union Hospital and Xuzhou Cancer Hospital were also utilized to verify the reliability and accuracy of the prediction model. Two regions of interest (ROI) were delineated on the preoperative MRI images of the patients using the semi-automatic tool ITK-SNAP (version 4.0.0), which were named the maximum anomaly region (ROI1) and the tumor region (ROI2), and Pyradiomics 3.0 was applied for feature extraction. Feature selection was performed using a least absolute shrinkage and selection operator (LASSO) filter and a Spearman correlation coefficient. Six classifiers, including Gauss naive Bayes (GNB), K-nearest neighbors (KNN), Random forest (RF), Adaptive boosting (AB), and Support vector machine (SVM) with linear kernel and multilayer perceptron (MLP), were used to build the prediction models, and the prediction performance of the six classifiers was evaluated by fivefold cross-validation. Moreover, the performance of prediction models was evaluated using area under the curve (AUC), precision (PRE), and other metrics.
    RESULTS: According to the inclusion and exclusion criteria, 172 patients with grade 2-3 astrocytoma were finally included in the study, and a total of 44 patients met the diagnosis of DAG-G. In the prediction task of DAG-G, the average AUC of GNB classifier was 0.74 ± 0.07, that of KNN classifier was 0.89 ± 0.04, that of RF classifier was 0.96 ± 0.03, that of AB classifier was 0.97 ± 0.02, that of SVM classifier was 0.88 ± 0.05, and that of MLP classifier was 0.91 ± 0.03, among which, AB classifier achieved the best prediction performance. In addition, the AB classifier achieved AUCs of 0.91 and 0.89 in two external validation datasets obtained from Wuhan Union Hospital and Xuzhou Cancer Hospital, respectively.
    CONCLUSIONS: The prediction model constructed based on preoperative MRI radiomics established in this study can basically realize the prospective, non-invasive, and accurate diagnosis of DAG-G, which is of great significance to help further optimize treatment plans for such patients, including expanding the extent of surgery and actively administering radiotherapy, targeted therapy, or other treatments after surgery, to fundamentally maximize the prognosis of 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
    目的:虽然免疫检查点抑制剂(ICI)已成功治疗各种恶性肿瘤,其对脑癌的疗效尚不清楚.使用PD-1抑制剂治疗复发性胶质母细胞瘤(GBM)的回顾性和前瞻性研究尚未确定生存益处。这项研究评估了ICI是否对某些复发性GBM患者有效。
    方法:这是一项单中心回顾性研究,对诊断为首次复发的GBM并接受派姆单抗或纳武单抗联合或不联合贝伐单抗的成年患者进行研究。将存档组织用于免疫组织化学(IHC)和靶向DNA下一代测序(NGS)分析。
    结果:从初始诊断开始的中位总生存期(mOS)为24.5个月(范围10-42)。ICI发病后的mOS为10个月(范围1-31),75%存活>6个月,46%存活>12个月。对来自8名患者的肿瘤的另外的IHC分析显示,对于具有升高的PD-L1表达的那些,ICI后的存活更长的趋势。来自15名患者的样本的NGS在初始诊断时和在任何时间点鉴定EGFR扩增与ICI后较差的生存相关(HR12.2,95%CI1.37-108,p=0.025和HR3.92,95%CI1.03-14.9,p=0.045,分别)。通过原位杂交,先前测试的EGFR扩增证实了这种意义。
    结论:ICI不能延长复发性GBM的总生存期。然而,分子测序确定EGFR扩增与较差的生存率相关。前瞻性研究可以验证EGFR扩增是否是ICI抗性的生物标志物,并确定其使用是否可以将应答者与非应答者分层。
    OBJECTIVE: While immune checkpoint inhibitors (ICI) have had success with various malignancies, their efficacy in brain cancer is still unclear. Retrospective and prospective studies using PD-1 inhibitors for recurrent glioblastoma (GBM) have not established survival benefit. This study evaluated if ICI may be effective for select patients with recurrent GBM.
    METHODS: This was a single-center retrospective study of adult patients diagnosed with first recurrence GBM and received pembrolizumab or nivolumab with or without concurrent bevacizumab. Archival tissue was used for immunohistochemistry (IHC) and targeted DNA next-generation sequencing (NGS) analysis.
    RESULTS: Median overall survival (mOS) from initial diagnosis was 24.5 months (range 10-42). mOS from onset of ICI was 10 months (range 1-31) with 75% surviving > 6 months and 46% > 12 months. Additional IHC analysis on tumors from eight patients demonstrated a trend of longer survival after ICI for those with elevated PD-L1 expression. NGS of samples from 15 patients identified EGFR amplification at initial diagnosis and at any time point to be associated with worse survival after ICI (HR 12.2, 95% CI 1.37-108, p = 0.025 and HR 3.92, 95% CI 1.03-14.9, p = 0.045, respectively). This significance was corroborated with previously tested EGFR amplification via in situ hybridization.
    CONCLUSIONS: ICI did not extend overall survival for recurrent GBM. However, molecular sequencing identified EGFR amplification as associated with worse survival. Prospective studies can validate if EGFR amplification is a biomarker of ICI resistance and determine if its use can stratify responders from non-responders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    EGFR突变是肺腺癌基因改变的最重要驱动因素,对EGFR-TKIs敏感。然而,EGFR-TKIs耐药在大多数EGFR突变的肺癌患者中是不可避免的.迄今为止,已经揭示了许多抗性机制,更多的人仍在调查中。由于选择的压力,耐药后可能存在肿瘤内异质性,尤其是在多行治疗后的患者。对那些病人来说,重要的是选择针对肿瘤主干/主要克隆的治疗方法,以实现最佳的临床获益.这里,我们将报道一个EGFR突变的肺腺癌患者,其耐药机制包括EGFR扩增的异质性,RB1的大片段缺失,以及靶向治疗后的组织学转变。在我们的案例中,根据液体活检监测和组织活检的下一代测序(NGS)结果,EGFR扩增似乎是耐药机制的主要克隆。考虑到高EGFR扩增水平,患者接受EGFR-TKI联合尼妥珠单抗联合治疗,抗EGFR单克隆抗体(mAb),取得了一定的临床效益。我们的案例揭示了EGFR突变患者EGFR扩增的治疗,并表明EGFR-TKI与抗EGFRmAb的组合可能是基于基因测试的可能治疗选择之一。此外,治疗方法的决定应关注肿瘤的主要克隆,并应根据治疗过程中遗传特征的动态变化进行及时调整。
    EGFR mutations are the most important drivers of gene alterations in lung adenocarcinomas and are sensitive to EGFR-TKIs. However, resistance to EGFR-TKIs is inevitable in the majority of EGFR-mutated lung cancer patients. Numerous resistant mechanisms have been revealed to date, and more are still under investigation. Owing to the selective pressure, intratumoral heterogeneity may exist after resistance, especially in patients after multiple lines of treatment. For those patients, it is important to choose therapies focused on the trunk/major clone of the tumor in order to achieve optimal clinical benefit. Here, we will report an EGFR-mutated lung adenocarcinoma patient with heterogeneity of resistant mechanisms including EGFR amplification, large fragment deletion of RB1, and histological transformations after targeted treatments. In our case, EGFR amplification seemed to be the major clone of the resistant mechanism according to the next-generation sequencing (NGS) results of both liquid biopsy monitoring and tissue biopsies. In consideration of the high EGFR amplification level, the patient was administered by combination treatment with EGFR-TKI plus nimotuzumab, an anti-EGFR monoclonal antibody (mAb), and achieved a certain degree of clinical benefit. Our case sheds light on the treatment of EGFR-mutant patients with EGFR amplification and indicates that a combination of EGFR-TKI with anti-EGFR mAb might be one of the possible treatment options based on genetic tests. Moreover, the decision on therapeutic approaches should focus on the major clone of the tumor and should make timely adjustments according to the dynamic changes of genetic characteristics during treatment.
    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
    特定分子畸变的鉴定指导2级星形细胞瘤的预后分层和处理。在大多数成人弥漫性低度神经胶质瘤(DLGG)中发现的异柠檬酸脱氢酶(IDH)1和2的突变,与IDH野生型(IDH-wt)对应物相比,似乎与良好的预后有关。此外,IDH-wt组可以发展额外的分子改变,恶化预后,例如表皮生长因子受体扩增(EGFR-amp)和端粒酶逆转录酶启动子(pTERT-mut)的突变。这篇综述分析了成年LGG遗传改变的预后影响和治疗意义。
    The identification of specific molecular aberrations guides the prognostic stratification and management of grade 2 astrocytomas. Mutations in isocitrate dehydrogenase (IDH) 1 and 2, found in the majority of adult diffuse low-grade glioma (DLGG), seem to relate to a favorable prognosis compared to IDH wild-type (IDH-wt) counterparts. Moreover, the IDH-wt group can develop additional molecular alterations worsening the prognosis, such as epidermal growth factor receptor amplification (EGFR-amp) and mutation of the promoter of telomerase reverse transcriptase (pTERT-mut). This review analyzes the prognostic impact and therapeutic implications of genetic alterations in adult LGG.
    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
    胶质母细胞瘤(GBM)是最常见的原发性脑肿瘤之一。有限的治疗选择和高复发率导致预后不良。一个频繁的,推定的驱动突变是致癌受体酪氨酸激酶EGFR的基因组扩增.通常伴有EGFRvIII等变体,异源表达和不依赖配体的信号使这种肿瘤亚型更加难以治疗,因为EGFR定向疗法最多只能显示微弱的效果。所以EGFR扩增的GBM被认为预后更差,因此,我们迫切需要更深入地了解新的治疗策略的分子机制和潜在靶点的检测.在这项研究中,我们观察了microRNAs(miRs)的水平,小的非编码RNA经常在癌症中失调,既充当癌基因又充当肿瘤抑制因子。有和没有EGFR扩增的GBM的比较分析应该深入了解miR的表达谱。它们既被认为是定向治疗的潜在靶标,也被认为是治疗试剂。EGFR扩增和EGFR正常GBM的miR谱的比较揭示了miR-183/96/182簇的上调,这与几种肿瘤实体的致癌特性有关。该miR簇的一个突出靶标是促凋亡因子FOXO1。通过观察FOXO1在EGFR扩增肿瘤中的下调,我们可以看到EGFR扩增的显著相关性,miR-183/96/182簇上调,以及对FOXO1的镇压。虽然在总生存率上没有显著差异,这些数据可能有助于对该肿瘤亚型的分子理解,并为基于miR的治疗提供潜在靶点.
    Glioblastoma (GBM) is one of the most frequent primary brain tumors. Limited therapeutic options and high recurrency rates lead to a dismal prognosis. One frequent, putative driver mutation is the genomic amplification of the oncogenic receptor tyrosine kinase EGFR. Often accompanied by variants like EGFRvIII, heterogenous expression and ligand independent signaling render this tumor subtype even more difficult to treat, as EGFR-directed therapeutics show only weak effects at best. So EGFR-amplified GBM is considered to have an even worse prognosis, and therefore, deeper understanding of molecular mechanisms and detection of potential targets for novel therapeutic strategies is urgently needed. In this study, we looked at the level of microRNAs (miRs), small non-coding RNAs frequently deregulated in cancer, both acting as oncogenes and tumor suppressors. Comparative analysis of GBM with and without EGFR amplification should give insight into the expression profiles of miRs, which are considered both as potential targets for directed therapies or as therapeutic reagents. Comparison of miR profiles of EGFR-amplified and EGFR-normal GBM revealed an upregulation of the miR-183/96/182 cluster, which is associated with oncogenic properties in several tumor entities. One prominent target of this miR cluster is FOXO1, a pro-apoptotic factor. By observing FOXO1 downregulation in EGFR-amplified tumors, we can see a significant correlation of EGFR amplification, miR-183/96/182 cluster upregulation, and repression of FOXO1. Although no significant difference in overall survival is shown, these data may contribute to the molecular understanding of this tumor subtype and offer potential targets for miR-based therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Case Reports
    表皮生长因子受体(EGFR)酪氨酸激酶抑制剂(TKIs)的开发彻底改变了晚期非小细胞肺癌(NSCLC)的药理学管理范式。发展对靶向治疗的抗性是不可避免的。大多数情况下,对获得EGFR-TKI治疗耐药后的单分子改变的治疗方法进行了很好的研究。然而,复杂耐药机制的治疗策略证据有限.本文介绍的是EGFR突变的NSCLC患者,其在一线EGFR-TKI后出现了复杂的耐药谱:T790M点突变和EGFR扩增。该患者使用奥希替尼和埃克替尼的组合安全治疗,并获得了显着的临床反应和明确的分子反应。在这里,我们提供了奥希替尼联合埃克替尼治疗EGFR经典突变以及T790M点突变和EGFR扩增的耐药突变的临床证据。
    The paradigm for the pharmacological management of advanced non-small cell lung cancer (NSCLC) has been revolutionized by the development of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs). Developing resistance to target therapy is unavoidable. Mostly, treatments for single molecular alteration after acquiring EGFR-TKI treatment resistance are well studied. However, there is limited evidence of treatment strategies for complex resistance mechanisms. Presented here is a case of an EGFR-mutated NSCLC patient who developed a complex resistance profile: T790M point mutation and EGFR amplification after first-line EGFR-TKI. This patient was safely treated with a combination of osimertinib and icotinib and achieved a significant clinical response and clear molecular response. Here we present the clinical evidence of the efficacy of osimertinib combined with icotinib in the treatment of EGFR classical mutation along with resistant mutation of T790M point mutation and EGFR amplification.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • DOI:
    文章类型: Journal Article
    胶质母细胞瘤(GBM)是成人最常见的恶性原发性脑肿瘤。这种癌症表现得很快,高度渗透生长,单独或成群侵入周围组织。GBM的侵袭性肿瘤生物学具有破坏性后果,中位生存期为15个月。GBM通常具有表皮生长因子受体(EGFR)异常。尽管GBM肿瘤生物学的研究取得了新进展,目前尚不清楚GBM中的突变是否与EGFR扩增和相关表型如肿瘤浸润相关.本研究旨在在30个人类GBM样品中进行全外显子组测序分析,以鉴定与EGFR扩增和浸润模式相关的突变肖像。我们的结果表明,EGFR扩增的肿瘤比未扩增的EGFR具有更高的突变率。2029个候选基因中的6个基因显示与EGFR扩增状态相关的突变。这些GBM基因的突变是新的,以前没有在GBM中报道过,并且在TCGA数据库中很少存在。GPR179、USP48和BLK仅在EGFR扩增病例中显示突变,所有受影响的病例都表现出弥漫性渗透模式。另一方面,ADGB中的突变,Ehhadh,和PTPN13仅存在于无EGFR扩增组,具有更多样化的浸润表型.总的来说,我们的工作确定了GBM的不同突变特征,这些突变特征与EGFR扩增和肿瘤浸润相关.
    Glioblastoma (GBM) is the most common malignant primary brain tumor in adults. This cancer shows rapid, highly infiltrative growth, that invades individually or in small groups the surrounding tissue. The aggressive tumor biology of GBM has devastating consequences with a median survival of 15 months. GBM often has Epidermal Growth Factor Receptor (EGFR) abnormalities. Despite recent advances in the study of GBM tumor biology, it is unclear whether mutations in GBM are related to EGFR amplification and relevant phenotypes like tumor infiltration. This study aimed to perform whole-exome sequencing analysis in 30 human GBM samples for identifying mutational portraits associated with EGFR amplification and infiltrative patterns. Our results show that EGFR-amplified tumors have overall higher mutation rates than EGFR-no-amplified. Six genes out of 2029 candidate genes show mutations associated with EGFR amplification status. Mutations in these genes for GBM are novel, not previously reported in GBM, and with little presence in the TCGA database. GPR179, USP48, and BLK show mutation only in EGFR-amplified cases, and all the affected cases exhibit diffuse infiltrative patterns. On the other hand, mutations in ADGB, EHHADH, and PTPN13, were present only in the EGFR-no-amplified group with a more diverse infiltrative phenotype. Overall, our work identified different mutational portraits of GBM related to well-established features like EGFR amplification and tumor infiltration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号