MET exon 14 skipping

MET 外显子 14 跳跃
  • 文章类型: Journal Article
    目的:MET第14外显子跳跃改变(METex14+)代表了具有独特生物学和基因组特征的非小细胞肺癌(NSCLC)的异质亚组。我们在一个大队列中描述了这种异质性,整合基因组和转录组分析与临床结果,阐明METex14+NSCLC的组织学和分子特征以及生存模式。
    方法:NSCLC组织样本(n=28,739)进行了基于DNA的下一代测序(592个基因,NextSeq)或全外显子组测序(NovaSeq),RNA测序,包括全转录组测序(WTS,NovaSeq),和CarisLifeSciences的PD-L1IHC(Dako22C3)。从大量RNA测序(quanTiseq)估计免疫细胞分数。真实世界生存数据(mOS)是根据保险索赔计算的。统计分析采用卡方,费希尔的精确,或Mann-WhitneyU和对数秩检验,并在适用的情况下进行假设检验。
    结果:共检出711例METex14+。在575例明确的组织学病例中,77(13.6%)为鳞状(Sq),474例(82.3%)为nSq(非鳞状),24例(4.1%)为腺鳞状。富集了POT1和BRCA2的突变,MDM2,HMGA2,CDK4和MET的扩增在METex14肿瘤中很常见。TMB高和TP53突变的肿瘤在METex14+中减少,与组织学无关。KEAP1(2.1vs14.7%)和STK11突变(0.8vs17.1%)仅在METex14+nSq中减少(与METex14+Sq相比,q<0.05)。虽然PD-L1高肿瘤的患病率在METex14+中富集,但与组织学无关,T细胞发炎的肿瘤仅在nSqMETex14+中富集。B细胞和CD8+T细胞(1.07-1.43倍)在nSqMETex14+中富集,和树突细胞(0.32倍)仅在METex14+Sq中减少。与METex14-肿瘤相比,METex14肿瘤的mOS有所改善(mOS=22.9mvs18.6m,HR=0.914,p=0.04)。此外,与未接受免疫疗法(IO)的肿瘤相比,接受免疫疗法(IO)的METex14肿瘤的生存率略有改善(mOS=27.5mvs21.8m;HR=0.803,p=0.03)。与METex14+Sq肿瘤相比,METex14+nSq肿瘤与改善的mOS相关(mOS=27.7vs8.9m,HR=0.493,p<0.0001)。
    结论:METex14+改变是NSCLC的异质性亚组。我们的分析揭示,与METex14+Sq相比,METex14+nSq表现出改善的存活率。组织学上不同的基因组和转录组变异需要临床考虑。
    OBJECTIVE: MET exon 14 skipping alterations (METex14+) represent a heterogeneous subgroup of non-small cell lung cancer (NSCLC) with distinct biological and genomic features. We characterized this heterogeneity in a large cohort, integrating genomic and transcriptomic profiling with clinical outcomes, to elucidate the histologic and molecular traits and survival patterns of METex14+ NSCLC.
    METHODS: NSCLC tissue samples (n = 28,739) underwent DNA-based next-generation sequencing (592 genes, NextSeq) or whole-exome sequencing (NovaSeq), RNA-sequencing including whole transcriptome sequencing (WTS, NovaSeq), and PD-L1 IHC (Dako 22C3) at Caris Life Sciences. Immune cell fractions were estimated from bulk RNA sequencing (quanTIseq). Real-world survival data (mOS) was calculated from insurance claims. Statistical analyses employed Chi-square, Fisher\'s exact, or Mann-Whitney U and log-rank tests and were corrected for hypothesis testing where applicable.
    RESULTS: A total of 711 METex14+ cases were detected. Of 575 cases of defined histology, 77 (13.6 %) were squamous (Sq), 474 (82.3 %) were nSq (non-squamous), and 24 (4.1 %) were adenosquamous. Mutations in POT1 and BRCA2 were enriched, and amplifications in MDM2, HMGA2, CDK4, and MET were common in METex14+ tumors. TMB-high and TP53 mutated tumors were reduced in METex14+ independent of histology. KEAP1 (2.1 vs 14.7 %) and STK11 mutations (0.8 vs 17.1 %) were reduced only in METex14+ nSq (vs METex14+ Sq, q < 0.05). While the prevalence of PD-L1 high tumors was enriched in METex14+ independent of histology, T-cell inflamed tumors were enriched only in nSq METex14+. B-cells and CD8+ T-cells (1.07-1.43-fold) were enriched in nSq METex14+, and dendritic cells (0.32 fold) were reduced only in METex14+ Sq. METex14+ tumors had a modest improvement in mOS compared to METex14- tumors (mOS = 22.9 m vs 18.6 m, HR = 0.914, p = 0.04). Moreover, METex14+ tumors who received immunotherapy (IO) had a modest improvement in survival (mOS = 27.5 m vs 21.8 m; HR = 0.803, p = 0.03) compared to those who did not receive IO. METex14+ nSq tumors were associated with improved mOS compared to METex14+ Sq tumors (mOS = 27.7 vs 8.9 m, HR = 0.493, p < 0.0001).
    CONCLUSIONS: METex14+ alterations are a heterogeneous subgroup of NSCLC. Our analysis reveals that METex14+ nSq exhibit improved survival compared to METex14+ Sq. The distinct genomic and transcriptomic variations across histologies warrant clinical consideration.
    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:间充质上皮转化(MET)外显子14(METex14)跳跃突变是非小细胞肺癌(NSCLC)的一种罕见改变,发生在约3%-4%的病例中。在这里,我们报告疾病和患者特征,来自意大利真实世界注册中心ATLAS的晚期METex14NSCLC患者中MET抑制剂的疗效和耐受性。
    方法:临床病理和分子数据,和治疗疗效/耐受性结局从ATLAS注册中心回顾性收集.
    结果:从2020年7月到2023年7月,共有146名METex14晚期NSCLC患者被纳入27个意大利中心。中位年龄是74岁,大多数患者是男性(52%),东部肿瘤协作组的表现状态<2(72%)和腺癌亚型(83%)。146名患者中有125名(86%)接受了至少一种全身性抗癌疗法。56例(38%)用卡马替尼治疗,34例(23%)用替泊替尼治疗。29%和52%的患者在一线和二线接受了针对性治疗,分别。在接受MET抑制剂治疗的患者队列中,缓解率(RR)为37%(以前接受过治疗的患者为33%,未接受治疗的患者为46%),疾病控制率为62%.中位随访时间为10.8个月,无进展生存期为6.6个月[95%可信区间(CI)4.3~8.3个月],总生存期为10.7个月(95%CI7.2~19.3个月).在可测量的脑转移患者(17例)中,颅内RR为41%。12%的患者发生≥3级治疗相关不良事件(TRAEs),其中7%的患者发生3级周围水肿。一名患者因肺炎发生致命不良反应。在6%和8%的病例中报告了与TRAE相关的剂量减少和停药。分别。
    结论:卡马替尼和替泊替尼是METex14NSCLC患者的有效治疗选择。真实世界的疗效结果比前瞻性临床试验中报道的要差。他们的活动在未接受治疗的人群中更为明显,这表明这是METex14患者管理的正确环境.
    BACKGROUND: Mesenchymal-epithelial transition (MET) exon 14 (METex14) skipping mutation is a rare alteration in non-small-cell lung cancer (NSCLC), occurring in about 3%-4% of cases. Here we report disease and patient characteristics, and efficacy and tolerability of MET inhibitors among advanced METex14 NSCLC patients from the Italian real-world registry ATLAS.
    METHODS: Clinical-pathological and molecular data, and treatment efficacy/tolerability outcomes were retrospectively collected from the ATLAS registry.
    RESULTS: From July 2020 to July 2023 a total of 146 METex14 advanced NSCLC patients were included across 27 Italian centers. Median age was 74 years, and most patients were male (52%), with an Eastern Cooperative Oncology Group performance status < 2 (72%) and adenocarcinoma subtype (83%). One hundred and twenty-five out of 146 (86%) patients received at least one line of systemic anticancer therapy. Fifty-six (38%) were treated with capmatinib and 34 (23%) with tepotinib. 29% and 52% of them received targeted treatment in the first and second line, respectively. In the cohort of patients treated with MET inhibitors, the response rate (RR) was 37% (33% in previously treated patients and 46% in treatment-naïve) with a disease control rate of 62%. With a median follow-up of 10.8 months, progression-free survival was 6.6 months [95% confidence interval (CI) 4.3-8.3 months] and overall survival was 10.7 months (95% CI 7.2-19.3 months). In patients with measurable brain metastases (17 cases), the intracranial RR was 41%. Grade ≥3 treatment-related adverse events (TRAEs) occurred in 12% of patients with grade 3 peripheral edema in 7% of cases. A fatal adverse reaction occurred in one patient due to pneumonitis. TRAEs-related dose reduction and discontinuation were reported in 6% and 8% of cases, respectively.
    CONCLUSIONS: Capmatinib and tepotinib represent an effective treatment option in NSCLC patients with METex14. Real-world efficacy outcomes are worse than those reported in prospective clinical trials. Their activity is more pronounced in the treatment-naïve population, suggesting that this is the right setting in the management of patients with METex14.
    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
    在低分化肿瘤的诊断检查中,T细胞受体(TCR)克隆性一直被认为是T细胞淋巴瘤的证据。MET外显子14跳跃(METex14)是通常在肺腺癌中见到的突变。在这里,我们首次报道了METex14肺腺癌的分离的单克隆TCRγ基因重排。一名69岁的妇女因胸腔积液出现在医院外。胸膜剥脱术显示恶性细胞CD30和CD138阳性,但BerEP4,KRT5和EMA阴性。一个模棱两可的HHV8染色被解释为阳性,导致原发性渗出性淋巴瘤的外部诊断错误。在我们的机构的额外的工作显示缺乏HHV8和T细胞标志物,但TCRγ克隆性的存在,pankeratin,和TTF1表达式。在内部活检中重复进行TCRγ测试,克隆性阴性。下一代测序检测到METex14,确认诊断为肺腺癌。讨论了潜在的诊断陷阱和预后/预测意义。
    In the diagnostic workup of poorly differentiated tumors, T-cell receptor (TCR) clonality has long been considered as evidence of T-cell lymphoma. MET exon 14 skipping (METex14) is a mutation typically seen in lung adenocarcinoma. Herein, we present the first report of METex14 lung adenocarcinoma with isolated monoclonal TCRγ gene rearrangement. A 69-year-old woman presented to an outside hospital with pleural effusions. A pleural decortication demonstrated malignant cells positive for CD30 and CD138 but negative for BerEP4, KRT5, and EMA. An equivocal HHV8 staining was interpreted as positive, leading to the erroneous outside diagnosis of primary effusion lymphoma. Additional workup at our institution revealed a lack of HHV8 and T-cell markers but the presence of TCRγ clonality, pankeratin, and TTF1 expression. Repeat TCRγ testing on the in-house biopsy was negative for clonality. Next-generation sequencing detected METex14, confirming the diagnosis of lung adenocarcinoma. The potential diagnostic pitfall and prognostic/predictive implications are discussed.
    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:最近在临床上使用了全面的癌症基因组谱分析测试来指导最佳治疗。目前批准的测试使用来自组织或血浆样品的DNA来分析几百个基因。RNA面板与DNA面板互补以检测融合和外显子跳跃。
    方法:在2017年4月至2022年3月之间,我们使用TodaiOncoPanel分析了非小细胞肺癌样本,靶向DNA和RNA的匹配的肿瘤/正常配对组。公开可用的基因组数据于2022年11月3日从癌症基因组学和高级治疗学中心数据库下载。
    结果:分析了60例非小细胞肺癌(NSCLC)样品。有了DNA面板,32个样品(53%)具有TP53功能丧失突变。在腺癌中,17人(33%)有EGFR激活突变,和6(12%)有ERBB2激活突变。还检测到一个BRCA1和一个BRCA2致病性种系变异体。用RNA面板,共检测到11个融合基因,都是腺癌.具体来说,EML4-ALK和KIF5B-RET分别从一个样品中检测到,另外9个都是新型融合体,致病性未知。此外,60个(7%)NSCLC样品中的4个具有MET外显子14跳跃。癌症基因组学和高级治疗学中心数据库的分析发现,在1514个NSCLC样本中发现了37个MET外显子14剪接位点突变(2%,p=0.039)。
    结论:用TodaiOncoPanel分析非小细胞肺癌可检测到许多药物靶标。其RNA面板可以高灵敏度地检测MET外显子14跳跃。
    BACKGROUND: Comprehensive cancer genomic profiling tests have recently been used clinically to guide optimal treatment. Currently approved tests use DNA from tissue or plasma samples to analyze a few hundred genes. RNA panels complement DNA panels to detect fusion and exon skipping.
    METHODS: Between April 2017 and March 2022, we analyzed non-small cell lung cancer samples using Todai OncoPanel, a matched tumor/normal pair panel targeting both DNA and RNA. Publicly available genomic data was downloaded from the Center for Cancer Genomics and Advanced Therapeutics database on 2022/11/3.
    RESULTS: Sixty non-small cell lung cancer (NSCLC) samples were analyzed. With the DNA panel, 32 samples (53%) had TP53 loss-of-function mutations. Among adenocarcinoma, 17 (33%) had EGFR activating mutations, and 6 (12%) had ERBB2 activating mutations. One BRCA1 and one BRCA2 pathogenic germline variant were also detected. With the RNA panel, 11 fusion genes were detected, all in adenocarcinoma. Specifically, EML4-ALK and KIF5B-RET were detected from one sample each, and 9 others were all novel fusions with unknown pathogenicity. In addition, 4 of 60 (7%) NSCLC samples harbored MET exon 14 skipping. Analysis of the Center for Cancer Genomics and Advanced Therapeutics database found 37 MET exon 14 splice site mutations in 1514 NSCLC samples (2%, p = 0.039).
    CONCLUSIONS: Analysis of NSCLC with Todai OncoPanel detected many druggable targets. Its RNA panel may detect MET exon 14 skipping with high sensitivity.
    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
    获得MET基因扩增,MET第14外显子跳跃突变,或MET融合可以作为肺癌患者对酪氨酸激酶抑制剂(TKIs)的耐药机制出现。联合使用METTKIs(如克唑替尼,卡马替尼,或替泊替尼)与针对获得性MET抗性的亲本TKIs没有很好地表征。
    多机构回顾性图表回顾确定了83例转移性癌基因驱动的NSCLC患者,将其分为以下两个配对队列:(1)MET队列(n=41)-获得MET耐药的患者继续其父母TKI并添加了METTKI或(2)化疗队列(n=42)-无任何可行的TpreKI并继续其父母铂耐药。临床病理特征,放射学反应(通过实体瘤1.1版的反应评估标准),生存结果,不良事件(AE)(通过不良事件通用术语标准5.0版),并收集基因组数据。使用Kaplan-Meier方法评估生存结果。根据治疗路线调整的多变量建模,脑转移瘤,TP53突变,和寡转移疾病。
    在MET队列中,中位年龄为56岁(范围:36-83岁).大多数患者从不吸烟者(41人中有28人,占68.3%)。基线脑转移是常见的(21/41,51%)。MET队列中最常见的癌基因是EGFR(41个中的30个,73.2%),ALK(41个中的7个,17.1%),和ROS1(41个中的两个,4.9%)。常见的TP53突变(41个中的32个,占78%)。获得的MET改变包括MET基因扩增(41个中的37个,90%),MET外显子14突变(41个中的两个,5%),和MET基因融合(41个中的两个,5%)。经过多变量调整后,MET队列的客观缓解率(ORR)高于化疗队列(ORR:69.2%对20%,p<0.001)。在MET队列中,MET基因拷贝数(≥10对6-10)不影响放射学反应(54.5%对68.4%,p=0.698)。基于使用的METTKI,ORR没有差异(F[2,36]=0.021,p=0.978)。MET和化疗组之间的无进展生存期(5对6个月;风险比=0.64;95%置信区间:0.34-1.23,p=0.18)或总生存期(13对11个月;风险比=0.75;95%置信区间:0.42-1.35,p=0.34)没有差异。在MET队列中,METTKI相关毒性的剂量减少是常见的(41个中的17个,41.4%),但对于父母TKIs的频率较低(41个中的2个,5%).3级AE在克唑替尼之间不显著,卡马替尼,和替泊替尼(p=0.3)。METTKIs的停药率为17%,METTKIs之间没有显着差异(p=0.315)。在MET队列的治疗前和治疗后活检(n=17)中,最常见的下一代测序结果是MET基因扩增丢失(17人中有15人,占88.2%),MET靶突变(17个中的7个,41.2%),新的Ras-Raf-MAPK改变(17个中的三个,17.6%),和EGFR基因扩增(17个中的两个,11.7%)。
    联合使用METTKIs(克唑替尼,卡马替尼,或替泊替尼)与亲本TKIs对获得性MET抗性是有效的。根据所使用的基础METTKI,放射学反应和AE没有显着差异。MET基因扩增丢失,MET靶向突变的发展,Ras-Raf-MAPK改变,和EGFR基因扩增是在双亲和METTKI组合的进展中发现的分子模式。
    UNASSIGNED: Acquired MET gene amplification, MET exon 14 skip mutations, or MET fusions can emerge as resistance mechanisms to tyrosine kinase inhibitors (TKIs) in patients with lung cancer. The efficacy and safety of combining MET TKIs (such as crizotinib, capmatinib, or tepotinib) with parent TKIs to target acquired MET resistance are not well characterized.
    UNASSIGNED: Multi-institutional retrospective chart review identified 83 patients with metastatic oncogene-driven NSCLC that were separated into the following two pairwise matched cohorts: (1) MET cohort (n = 41)-patients with acquired MET resistance continuing their parent TKI with a MET TKI added or (2) Chemotherapy cohort (n = 42)-patients without any actionable resistance continuing their parent TKI with a platinum-pemetrexed added. Clinicopathologic features, radiographic response (by means of Response Evaluation Criteria in Solid Tumors version 1.1), survival outcomes, adverse events (AEs) (by means of Common Terminology Criteria for Adverse Events version 5.0), and genomic data were collected. Survival outcomes were assessed using Kaplan-Meier methods. Multivariate modeling adjusted for lines of therapy, brain metastases, TP53 mutations, and oligometastatic disease.
    UNASSIGNED: Within the MET cohort, median age was 56 years (range: 36-83 y). Most patients were never smokers (28 of 41, 68.3%). Baseline brain metastases were common (21 of 41, 51%). The most common oncogenes in the MET cohort were EGFR (30 of 41, 73.2%), ALK (seven of 41, 17.1%), and ROS1 (two of 41, 4.9%). Co-occurring TP53 mutations (32 of 41, 78%) were frequent. Acquired MET alterations included MET gene amplification (37 of 41, 90%), MET exon 14 mutations (two of 41, 5%), and MET gene fusions (two of 41, 5%). After multivariate adjustment, the objective response rate (ORR) was higher in the MET cohort versus the chemotherapy cohort (ORR: 69.2% versus 20%, p < 0.001). Within the MET cohort, MET gene copy number (≥10 versus 6-10) did not affect radiographic response (54.5% versus 68.4%, p = 0.698). There was no difference in ORR on the basis of MET TKI used (F [2, 36] = 0.021, p = 0.978). There was no difference in progression-free survival (5 versus 6 mo; hazard ratio = 0.64; 95% confidence interval: 0.34-1.23, p = 0.18) or overall survival (13 versus 11 mo; hazard ratio = 0.75; 95% confidence interval: 0.42-1.35, p = 0.34) between the MET and chemotherapy cohorts. In the MET cohort, dose reductions for MET TKI-related toxicities were common (17 of 41, 41.4%) but less frequent for parent TKIs (two of 41, 5%). Grade 3 AEs were not significant between crizotinib, capmatinib, and tepotinib (p = 0.3). The discontinuation rate of MET TKIs was 17% with no significant differences between MET TKIs (p = 0.315). Among pre- and post-treatment biopsies (n = 17) in the MET cohort, the most common next-generation sequencing findings were loss of MET gene amplification (15 of 17, 88.2%), MET on-target mutations (seven of 17, 41.2%), new Ras-Raf-MAPK alterations (three of 17, 17.6%), and EGFR gene amplification (two of 17, 11.7%).
    UNASSIGNED: The efficacy and safety of combining MET TKIs (crizotinib, capmatinib, or tepotinib) with parent TKIs for acquired MET resistance are efficacious. Radiographic response and AEs did not differ significantly on the basis of the underlying MET TKI used. Loss of MET gene amplification, development of MET on-target mutations, Ras-Raf-MAPK alterations, and EGFR gene amplification were molecular patterns found on progression with dual parent and MET TKI combinations.
    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
    脑肿瘤代表一组异质性肿瘤,其特征是高度侵袭性和不良预后。尽管最近的治疗进展,脑肿瘤的治疗,包括胶质母细胞瘤(GBM),侵袭性原发性脑肿瘤与不良预后和对治疗的抵抗力有关,仍然是一个重大挑战。受体酪氨酸激酶(RTK)在发育和成年期至关重要。通过激活突变和基因扩增的RTK的失调有助于许多人类癌症,并为治疗提供有吸引力的治疗靶标。在生理条件下,MetRTK,肝细胞生长因子/散射因子(HGF/SF)受体,促进调节组织修复和胚胎发生中涉及的上皮-间质转化(EMT)的基本信号级联。在癌症中,增加的Met活性通过提供增殖信号促进肿瘤生长和转移,生存,和迁移/入侵。最近的临床基因组研究揭示了MET在GBM中发生遗传改变的多种机制,包括病灶放大,染色体重排产生基因融合,和一个剪接变异突变(外显子14跳跃,METex14del).值得注意的是,MET过表达通过促进肿瘤干细胞样细胞的存活而促进GBM中的化疗抗性。这与独特的Met诱导途径有关,例如DNA修复机制的上调,可以保护肿瘤细胞免受化疗的细胞毒性作用。MET靶向疗法的发展代表了脑肿瘤治疗的重要一步。临床前研究表明,MET靶向治疗(单克隆抗体或小分子抑制剂)可以抑制生长和侵袭,提高常规疗法的疗效。早期临床试验表明,MET靶向治疗在改善复发性GBM患者的总体生存率方面取得了有希望的结果。然而,挑战依然存在,包括患者分层的需要,治疗方案的优化,以及抗性机制的识别。这篇综述旨在强调目前对GBM中MET失调机制的理解。此外,它将重点关注针对GBM中MET失调的治疗的临床前和临床评估.
    Brain tumors represent a heterogeneous group of neoplasms characterized by a high degree of aggressiveness and a poor prognosis. Despite recent therapeutic advances, the treatment of brain tumors, including glioblastoma (GBM), an aggressive primary brain tumor associated with poor prognosis and resistance to therapy, remains a significant challenge. Receptor tyrosine kinases (RTKs) are critical during development and in adulthood. Dysregulation of RTKs through activating mutations and gene amplification contributes to many human cancers and provides attractive therapeutic targets for treatment. Under physiological conditions, the Met RTK, the hepatocyte growth factor/scatter factor (HGF/SF) receptor, promotes fundamental signaling cascades that modulate epithelial-to-mesenchymal transition (EMT) involved in tissue repair and embryogenesis. In cancer, increased Met activity promotes tumor growth and metastasis by providing signals for proliferation, survival, and migration/invasion. Recent clinical genomic studies have unveiled multiple mechanisms by which MET is genetically altered in GBM, including focal amplification, chromosomal rearrangements generating gene fusions, and a splicing variant mutation (exon 14 skipping, METex14del). Notably, MET overexpression contributes to chemotherapy resistance in GBM by promoting the survival of cancer stem-like cells. This is linked to distinctive Met-induced pathways, such as the upregulation of DNA repair mechanisms, which can protect tumor cells from the cytotoxic effects of chemotherapy. The development of MET-targeted therapies represents a major step forward in the treatment of brain tumours. Preclinical studies have shown that MET-targeted therapies (monoclonal antibodies or small molecule inhibitors) can suppress growth and invasion, enhancing the efficacy of conventional therapies. Early-phase clinical trials have demonstrated promising results with MET-targeted therapies in improving overall survival for patients with recurrent GBM. However, challenges remain, including the need for patient stratification, the optimization of treatment regimens, and the identification of mechanisms of resistance. This review aims to highlight the current understanding of mechanisms underlying MET dysregulation in GBM. In addition, it will focus on the ongoing preclinical and clinical assessment of therapies targeting MET dysregulation in GBM.
    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:检查点抑制剂对具有MET外显子14跳跃(META14ex)的非小细胞肺癌(NSCLC)的疗效仍存在争议。
    方法:分析了2016-2022年间在10个德国中心接受一线化疗(CHT)和/或免疫治疗(IO)的110例连续MET14exNSCLC患者。
    结果:35/110(32%)患者接受联合CHT-IO治疗,仅IO到43/110(39%),和CHT到32/110(29%)的前期。与CHT相比,CHT-IO显示更长的无进展生存期(中位PFS6vs.2.5个月,p=0.004),更客观的反应(ORR49%与28%,p=0.086)和数字上更长的总生存期(OS16与10个月,p=0.240)。对于IO单药治疗,操作系统(14与16个月)和反应持续时间(26vs.22个月)与CHT-IO相当。与CHT-IO相比,IO的原发性进行性疾病(PD)更常见(13/43vs.3/35,p=0.018),特别是对于从不吸烟者(p=0.041)。较高的PD-L1TPS与更好的IO结果无关,但是TP53突变的肿瘤显示出数字上改善的ORR(56%与32%,p=0.088)和PFS(6vs.3个月,p=0.160),以及与野生型对应物相比,多变量分析中的OS更长(HR=0.54,p=0.034)。对35/75(47%)的患者进行了任何二线治疗,与克唑替尼相比,卡马替尼或替替尼的生存期更长(PFS10vs.3个月,p=0.013;OS16vs.13个月,p=0.270)。
    结论:CHT-IO优于CHT,和IO单独对MET14exNSCLC也有效,特别是在存在TP53突变且与PD-L1表达无关的情况下,但从不吸烟者患原发性PD的风险较高.
    BACKGROUND: The efficacy of checkpoint inhibitors for non-small cell lung cancer (NSCLC) with MET exon 14 skipping (METΔ14ex) remains controversial.
    METHODS: 110 consecutive METΔ14ex NSCLC patients receiving first-line chemotherapy (CHT) and/or immunotherapy (IO) in 10 German centers between 2016-2022 were analyzed.
    RESULTS: Combined CHT-IO was given to 35/110 (32%) patients, IO alone to 43/110 (39%), and CHT to 32/110 (29%) upfront. Compared to CHT, CHT-IO showed longer progression-free survival (median PFS 6 vs. 2.5 months, p = 0.004), more objective responses (ORR 49% vs. 28%, p = 0.086) and numerically longer overall survival (OS 16 vs. 10 months, p = 0.240). For IO monotherapy, OS (14 vs. 16 months) and duration of response (26 vs. 22 months) were comparable to those of CHT-IO. Primary progressive disease (PD) was more frequent with IO compared to CHT-IO (13/43 vs. 3/35, p = 0.018), particularly for never-smokers (p = 0.041). Higher PD-L1 TPS were not associated with better IO outcomes, but TP53 mutated tumors showed numerically improved ORR (56% vs. 32%, p = 0.088) and PFS (6 vs. 3 months, p = 0.160), as well as longer OS in multivariable analysis (HR=0.54, p = 0.034) compared to their wild-type counterparts. Any second-line treatment was administered to 35/75 (47%) patients, with longer survival for capmatinib or tepotinib compared to crizotinib (PFS 10 vs. 3 months, p = 0.013; OS 16 vs. 13 months, p = 0.270).
    CONCLUSIONS: CHT-IO is superior to CHT, and IO alone also effective for METΔ14ex NSCLC, especially in the presence of TP53 mutations and independent of PD-L1 expression, but never-smokers are at higher risk of primary PD.
    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
    Lung cancer stands as a leading cause of mortality in China, with EGFR mutations frequently identified as pivotal driver genes. Osimertinib, a tyrosine kinase inhibitor targeting EGFR mutations, is typically employed as a first-line treatment for EGFR-sensitive mutations; nevertheless, resistance can emerge. In this case report, we present the case of a 53-year-old non-smoking male diagnosed with stage IV lung adenocarcinoma bearing an EGFR exon 19 deletion. This patient eventually developed resistance to both Erlotinib and Osimertinib after 28 months of treatment. Subsequent genetic testing uncovered the emergence of new MET exon 14 skipping and MET fusion, coexisting with the initial EGFR exon 19 deletion. In light of this complex molecular profile, the patient was administered a combination therapy consisting of Osimertinib and Capmatinib. This novel approach yielded a partial response, and notably, the patient experienced a progression-free survival exceeding 7 months. Vigilant monitoring of the patient\'s progress revealed the disappearance of the MET exon 14 skipping and a notable improvement in the patient\'s symptoms. This case report underscores the potential efficacy of Osimertinib and Capmatinib combination therapy as a viable treatment strategy for patients harboring EGFR-mutated lung cancer who develop resistance to first-line EGFR inhibitors due to MET activation.
    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
    具有MET外显子14跳跃突变(METex14)的非小细胞肺癌与不良预后相关。由于测试和药物获取方面的障碍,新型靶向疗法的整合具有挑战性。我们,因此,试图表征治疗模式,结果,METex14突变非小细胞肺癌患者治疗测序的新问题。
    我们回顾了2014年至2020年间加拿大四个癌症中心的所有METex14改变的NSCLC病例。人口统计,疾病特征,全身治疗,总体反应率(ORR),生存,并对其毒性进行了总结。
    在64例METex14突变非小细胞肺癌患者中,中位总生存期为23.1个月:第1阶段为127.0个月,切除的第2和第3阶段为27.3个月,不可切除的第3或第4阶段为16.6个月。分别。在晚期疾病患者中,22%的患者不适用于全身治疗。MET酪氨酸激酶抑制剂(TKIs)用于28例患者,ORR为33%,中位无进展生存期为2.7个月,和3.8个月的选择性TKIs。25例患者接受了程序性细胞死亡蛋白-1(PD-1)抑制剂治疗,ORR为44%,无进展生存期为10.6个月。初始TKI治疗后,随后的METTKIs未见反应。在接受PD-1抑制剂后接受METTKI的患者中,有64%发生了3级或更高的毒性,而未接受PD-1抑制剂的患者中,有8%发生了毒性。
    许多晚期METex14NSCLC患者身体不适,无法接受治疗。PD-1抑制剂作为初始治疗似乎是有效的,尽管随后的METTKIs具有更大的毒性。因此,及时检测METex14跳越和初始治疗对提高患者生存率至关重要.
    UNASSIGNED: NSCLC with MET exon 14 skipping mutation (METex14) is associated with poor outcomes. Integration of novel targeted therapies is challenging because of barriers in testing and drug access. We, therefore, sought to characterize the treatment patterns, outcomes, and emerging issues of treatment sequencing in patients with METex14-mutant NSCLC.
    UNASSIGNED: We reviewed all NSCLC cases with METex14 alterations between 2014 and 2020 across four Canadian cancer centers. Demographics, disease characteristics, systemic therapy, overall response rates (ORRs), survival, and toxicity were summarized.
    UNASSIGNED: Among 64 patients with METex14-mutant NSCLC, the median overall survival was 23.1 months: 127.0 months in stage 1, 27.3 months in resected stage 2 and 3, and 16.6 months in unresectable stage 3 or 4 disease, respectively. In patients with advanced disease, 22% were too unwell for systemic treatment. MET tyrosine kinase inhibitors (TKIs) were administered to 28 patients with an ORR of 33%, median progression-free survival of 2.7 months, and 3.8 months for selective TKIs. Programmed cell death protein-1 (PD-1) inhibitors were given to 25 patients-the ORR was 44% and progression-free survival was 10.6 months. No responses were seen with subsequent MET TKIs after initial TKI treatment. Grade 3 or higher toxicities occurred in 64% of patients who received MET TKI after PD-1 inhibitors versus 8% in those who did not receive PD-1 inhibitors.
    UNASSIGNED: Many patients with advanced METex14 NSCLC were too unwell to receive treatment. PD-1 inhibitors seem effective as an initial treatment, although greater toxicity was seen with subsequent MET TKIs. Thus, timely testing for METex14 skipping and initial therapy are imperative to improving patient survival.
    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:酪氨酸激酶(TK)改变,如间变性淋巴瘤激酶(ALK)融合,神经营养酪氨酸受体激酶(NTRK)融合,c-ros癌基因1(ROS1)融合和间充质上皮转化因子(MET)外显子14跳跃,已在结直肠癌(CRC)中报道。我们以前在我们的队列中报道过NTRK融合的CRC。然而,其临床病理特征尚未完全阐明。
    结果:对944例患者的951例CRC病变进行了基于组织微阵列(TMA)的免疫组织化学(IHC)。IHC对ALK和ROS1评估为阳性或阴性,对c-MET评估为0至3+。对于ALK和ROS1IHC阳性病例,基于RNA的不平衡基因表达测定,进行ArcherFusionPlex测定和逆转录聚合酶链反应(RT-PCR),然后进行Sanger测序。对于c-METIHC3+病例,进行RT-PCR,然后进行Sanger测序。3例(0.2%)ALKIHC阳性,均显示ALK基因表达失衡。确认了以下ALK融合:EML4(外显子21)::ALK(外显子20),EML4(外显子6)::ALK(外显子19)和HMBOX1(外显子6)::ALK(外显子20)。两个显示微卫星不稳定性高/错配修复(MMR)缺陷,都位于右结肠.在一个病例中,ROS1IHC呈阳性;然而,表达不平衡,ROS1融合为阴性。42例(4.4%)显示c-METIHC3+。在9例病例中证实了MET第14外显子跳跃。所有病例均为微卫星稳定/MMR熟练,八个位于左结肠和直肠。
    结论:这些TK改变的CRC具有明显的临床病理特征。加上我们之前的研究,15例(1.6%)具有可靶向的TK改变(三个NTRK融合,三个ALK融合,9个MET外显子14跳跃)。
    OBJECTIVE: Tyrosine kinase (TK) alterations, such as anaplastic lymphoma kinase (ALK) fusion, neurotrophic tyrosine receptor kinase (NTRK) fusion, c-ros oncogene 1 (ROS1) fusion and mesenchymal-epithelial transition factor (MET) exon 14 skipping, have been reported in colorectal cancers (CRC). We have previously reported CRCs with NTRK fusion among our cohort. However, their clinicopathological features have not been fully elucidated.
    RESULTS: Tissue microarray (TMA)-based immunohistochemistry (IHC) was performed on 951 CRC lesions from 944 patients. IHC was evaluated as positive or negative for ALK and ROS1 and 0 to 3+ for c-MET. For ALK and ROS1 IHC-positive cases, RNA-based imbalanced gene expression assays, Archer FusionPlex assays and reverse transcription-polymerase chain reaction (RT-PCR) followed by Sanger sequencing were performed. For c-MET IHC 3+ cases, RT-PCR followed by Sanger sequencing were performed. ALK IHC was positive in three cases (0.2%) and all showed imbalanced ALK gene expression. The following ALK fusions were confirmed: EML4 (exon 21)::ALK (exon 20), EML4 (exon 6)::ALK (exon 19) and HMBOX1 (exon 6)::ALK (exon 20). Two showed microsatellite instability-high/mismatch repair (MMR)-deficient, and all were located in the right colon. ROS1 IHC was positive in one case; however, imbalanced expression and ROS1 fusion was negative. Forty-two cases (4.4%) showed c-MET IHC3+. MET exon 14 skipping was confirmed in nine cases. All cases were microsatellite stable/MMR-proficient, and eight were located in the left colon and rectum.
    CONCLUSIONS: CRCs with these TK alterations had distinct clinicopathological features. Together with our previous study, 15 cases (1.6%) harboured targetable TK alterations (three NTRK fusion, three ALK fusion, nine MET exon 14 skipping).
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号