MET alterations

  • 文章类型: Journal Article
    胃癌是最常见的癌症之一,也是全球癌症相关死亡的主要原因。因为胃癌是高度异质性的,由具有不同分子和临床特征的不同亚型组成,胃癌的管理需要更好的定义,生物标志物指导,基于分子的治疗策略。MET是介导重要生理过程的受体酪氨酸激酶,比如胚胎发生,组织再生,伤口愈合。然而,越来越多的证据表明,异常的MET通路激活有助于多种癌症类型的肿瘤增殖和转移,包括胃癌,并与不良患者预后相关。因此,MET靶向疗法正在积极开发,并已证明有希望的进展,尤其是MET酪氨酸激酶抑制剂。本综述旨在简要介绍MET改变在胃癌中的作用,并详细总结目前MET酪氨酸激酶抑制剂在该疾病领域的研究进展,重点是萨沃利替尼。tepotinib,卡马替尼,还有克唑替尼.在现有知识的基础上,这篇综述进一步讨论了MET改变测试中存在的挑战,对MET抑制剂的可能耐药机制,以及MET靶向治疗的未来方向。
    Gastric cancer is among the most frequently occurring cancers and a leading cause of cancer-related deaths globally. Because gastric cancer is highly heterogenous and comprised of different subtypes with distinct molecular and clinical characteristics, the management of gastric cancer calls for better-defined, biomarker-guided, molecular-based treatment strategies. MET is a receptor tyrosine kinase mediating important physiologic processes, such as embryogenesis, tissue regeneration, and wound healing. However, mounting evidence suggests that aberrant MET pathway activation contributes to tumour proliferation and metastasis in multiple cancer types, including gastric cancer, and is associated with poor patient outcomes. As such, MET-targeting therapies are being actively developed and promising progress has been demonstrated, especially with MET tyrosine kinase inhibitors. This review aims to briefly introduce the role of MET alterations in gastric cancer and summarize in detail the current progress of MET tyrosine kinase inhibitors in this disease area with a focus on savolitinib, tepotinib, capmatinib, and crizotinib. Building on current knowledge, this review further discusses existing challenges in MET alterations testing, possible resistance mechanisms to MET inhibitors, and future directions of MET-targeting 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: Journal Article
    背景:间充质上皮转化因子(MET)是一种罕见的肿瘤驱动基因,而对携带该驱动基因的非小细胞肺癌(NSCLC)患者的免疫治疗信息有限.在这里,我们评估了不同治疗方案下免疫检查点抑制剂(ICI)对MET改变的NSCLC患者的疗效和安全性。
    方法:从2019年6月至2023年12月,我们评估了ICIs在42例MET改变的NSCLC患者中的疗效和毒性。使用Kaplan-Meier方法绘制生存曲线,并应用Cox比例风险模型进行单变量和多变量分析。我们根据RECISTv1.1评估靶病变的大小,客观反应率(ORR)定义为完全反应(CR)和部分反应(PR)的总和,疾病控制率(DCR)为CR的总和,PR,疾病稳定。
    结果:本回顾性研究共纳入42例MET改变的非小细胞肺癌患者,10是MET14跳跃突变,32是MET扩增。ICI治疗的ORR为30.95%,DCR为71.43%。中位无进展生存期(mPFS)和中位总生存期(OS)分别为4.40和13.97个月,分别。ICI单药治疗和联合ICI治疗的mPFS之间存在统计学差异(2.8vs7.8个月,p=0.022)。药物相关不良反应发生率为47.62%,以骨髓抑制为主(14.28%),免疫相关肺炎(7.14%),肝功能损害(7.14%),6例患者(14.28%)出现3级或以上不良事件。
    结论:MET改变的NSCLC患者可以从免疫治疗中获益,尤其是ICI联合治疗的患者。然而,在使用联合ICI治疗时应特别注意3/4级不良反应的发生。
    BACKGROUND: Mesenchymal epithelial transition factor (MET) is a rare oncologic driver gene, and information on immunotherapy for non-small cell lung cancer (NSCLC) patients with this driver gene is limited. Here we evaluate the efficacy and safety of immune checkpoint inhibitors (ICI) under different therapeutic regimen for NSCLC patients with MET alterations.
    METHODS: From June 2019 to December 2023, we assessed the efficacy and toxicity of ICIs in 42 NSCLC patients with MET alterations. Survival curves were plotted using the Kaplan-Meier method and the Cox proportional hazards model applied for univariate and multivariate analyses. We assessed the size of target lesion according to RECIST v1.1, and objective response rate (ORR) was defined as the sum of complete response (CR) and partial response (PR), disease control rate (DCR) as the sum of CR, PR, and disease stable.
    RESULTS: A total of 42 NSCLC patients with MET alterations were included in this retrospective study, 10 was MET 14 skipping mutation and 32 was MET amplification. The ORR for ICI treatment was 30.95% and the DCR was 71.43%. Median progression-free survival (mPFS) and median overall survival (OS) were 4.40 and 13.97 months, respectively. There exists statistical differences between the mPFS of ICI monotherapy and combine ICI therapy (2.8 vs 7.8 months, p = 0.022). The incidence of drug-related adverse reactions was 47.62%, mainly bone marrow suppression (14.28%), immune-related pneumonia (7.14%), and liver function impairment (7.14%), and six patients (14.28%) experiencing grade 3 or above adverse events.
    CONCLUSIONS: NSCLC patients with MET alterations can benefit from immunotherapy, especially the patients treated by combined ICI therapy. However, special attention should be paid to the occurrence of grade 3/4 adverse reactions while using the combined ICI therapy.
    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
    MET改动,包括MET第14外显子跳跃变体,MET扩增,MET过表达,和MET融合,在原发性肿瘤发生和对靶向治疗的获得性抵抗中发挥关键作用,特别是EGFR酪氨酸激酶抑制剂。它们代表着重要的诊断,预后,和许多实体瘤类型的预测性生物标志物。然而,由于MET改变的复杂性和平台技术的多样性,MET改变的检测具有挑战性.因此,高灵敏度的技术,特异性,和可靠的分子检测精度需要克服这些障碍,并有助于生物标志物指导的治疗。当前的综述强调了MET改变在多种癌症中作为致癌驱动因素的作用,以及它们在靶向治疗抗性发展中的参与。此外,我们的审查提供了有关选择用于检测MET外显子14跳跃变体的各种跨平台技术的概述和建议,MET扩增,MET过表达,和MET融合。此外,讨论了这些常见检测平台背后的挑战和障碍。
    MET alterations, including MET exon 14 skipping variants, MET amplification, MET overexpression, and MET fusion, play pivotal roles in primary tumorigenesis and acquired resistance to targeted therapies, especially EGFR tyrosine kinase inhibitors. They represent important diagnostic, prognostic, and predictive biomarkers in many solid tumor types. However, the detection of MET alterations is challenging due to the complexity of MET alterations and the diversity of platform technologies. Therefore, techniques with high sensitivity, specificity, and reliable molecular detection accuracy are needed to overcome such hindrances and aid in biomarker-guided therapies. The current review emphasizes the role of MET alterations as oncogenic drivers in a variety of cancers and their involvement in the development of resistance to targeted therapies. Moreover, our review provides an overview of and recommendations on the selection of various cross-platform technologies for the detection of MET exon 14 skipping variants, MET amplification, MET overexpression, and MET fusion. Furthermore, challenges and hurdles underlying these common detection platforms are discussed.
    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
    MET原癌基因编码一个关键的酪氨酸激酶受体,结合肝细胞生长因子(HGF,也称为散射因子,SF)和管理基本的生物过程,如器官发生,组织修复,和血管生成。MET的多效性生理功能解释了其在广泛肿瘤的癌症进展中的不同作用;MET的遗传/表观遗传改变驱动肿瘤细胞播散,转移,以及对常规和靶向治疗的获得性抵抗。因此,瞄准MET成为一种有前途的策略,许多努力致力于确定阻碍MET信号的最佳方式。尽管取得了令人鼓舞的结果,然而,MET在肿瘤发生中的功能的复杂性产生了有趣的观察,在我们的理解力上培养谦卑的立场。这篇综述探讨了有关癌症中MET改变的最新发现,阐明它们的生物学影响,讨论治疗途径,并概述了未来的方向。通过对研究问题进行语境化,阐明研究目的,这项工作浏览了MET生物学在癌症中的复杂性,提供全面的视角。
    The MET proto-oncogene encodes a pivotal tyrosine kinase receptor, binding the hepatocyte growth factor (HGF, also known as scatter factor, SF) and governing essential biological processes such as organogenesis, tissue repair, and angiogenesis. The pleiotropic physiological functions of MET explain its diverse role in cancer progression in a broad range of tumors; genetic/epigenetic alterations of MET drive tumor cell dissemination, metastasis, and acquired resistance to conventional and targeted therapies. Therefore, targeting MET emerged as a promising strategy, and many efforts were devoted to identifying the optimal way of hampering MET signaling. Despite encouraging results, however, the complexity of MET\'s functions in oncogenesis yields intriguing observations, fostering a humbler stance on our comprehension. This review explores recent discoveries concerning MET alterations in cancer, elucidating their biological repercussions, discussing therapeutic avenues, and outlining future directions. By contextualizing the research question and articulating the study\'s purpose, this work navigates MET biology\'s intricacies in cancer, offering a comprehensive perspective.
    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
    Targeting the MET pathway in advanced NSCLC has been of particular interest due to its role as both a primary oncogenic driver and secondary oncogenic driver of acquired resistance. Activation of the MET pathway can occur through several mechanisms, which can complicate the diagnostic and treatment approach. Recently, several MET-directed therapies have been developed with promising results. In this narrative review, we summarize the biology and mechanism of MET as a clinically relevant driver mutation, distinct MET alterations including diagnostic challenges, significance in the setting of acquired resistance, and novel treatment strategies in 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:为了帮助肿瘤药物开发,我们调查了在临床实践中接受下一代测序(NGS)的2,239例肿瘤患者的MET原癌基因受体酪氨酸激酶基因异常.材料和方法:从2019年11月到2021年1月,访问肿瘤诊所的2,239名晚期实体瘤患者接受了NGS。NGS面板包括使用档案组织样本的>500个综合NGS测试。1,137(50.8%)和1,761(78.7%)患者可获得程序性死亡配体1(PD-L1)22C3测定结果和关于初始化疗的临床记录,分别进行总生存期(OS)分析。结果:2,239例患者代表37种癌症。NGS小组包括>500个基因,微卫星不稳定状态,肿瘤突变负担,和融合。最常见的癌症类型是结直肠癌(N=702),胃(N=481),和肉瘤(N=180)。在212例患者中检测到MET像差。所有MET扩增的肿瘤都有微卫星稳定状态,和8具有较高的肿瘤突变负担。在46例MET扩增癌症患者中,8例免疫组织化学显示MET阳性蛋白表达(2+和3+)。在10例患者中检测到MET融合。MET融合的伙伴基因包括ST7、TFEC、LRRD1,CFTR,CAV1、PCM1、HLA-DRB1和CAPZA2。在生存分析中,MET基因融合扩增患者的OS和无进展生存期(PFS)比无MET基因融合扩增患者短.因此,MET畸变被确定为化疗反应的一个因素。结论:大约2.1%和0.4%的晚期实体瘤患者出现MET基因扩增和融合,分别,与没有MET基因扩增或融合的患者相比,对化疗的反应更差,OS和PFS明显更短。
    Background: To aid in oncology drug development, we investigated MET proto-oncogene receptor tyrosine kinase gene aberrations in 2,239 oncology patients who underwent next-generation sequencing (NGS) in clinical practice. Materials and methods: From November 2019 to January 2021, 2,239 patientswith advanced solid tumors who visited oncology clinics underwent NGS. The NGS panel included >500 comprehensive NGS tests using archival tissue specimens. Programmed death-ligand 1(PD-L1) 22C3 assay results and clinical records regarding initial chemotherapy were available for 1,137 (50.8%) and 1,761 (78.7%) patients, respectively for overall survival (OS) analysis. Results: The 2,239 patients represented 37 types of cancer. The NGS panel included >500 genes, microsatellite instability status, tumor mutational burden, and fusions. The most common cancer types were colorectal (N = 702), gastric (N = 481), and sarcoma (N = 180). MET aberrations were detected in 212 patients. All MET-amplified tumors had microsatellite stable status, and 8 had a high tumor mutational burden. Of 46 patients with MET-amplified cancers, 8 had MET-positive protein expression by immunohistochemistry (2+ and 3+). MET fusion was detected in 10 patients. Partner genes of MET fusion included ST7, TFEC, LRRD1, CFTR, CAV1, PCM1, HLA-DRB1, and CAPZA2. In survival analysis, patients with amplification of MET gene fusion had shorter OS and progression-free survival (PFS) than those without. Thus, MET aberration was determined to be a factor of response to chemotherapy. Conclusion: Approximately 2.1% and 0.4% of patients with advanced solid tumors demonstrated MET gene amplification and fusion, respectively, and displayed a worse response to chemotherapy and significantly shorter OS and PFS than those without MET gene amplification or fusion.
    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
    MET exon 14 skipping (METex14) is a validated oncogenic driver in lung cancer and MET tyrosine kinase inhibitors are now available as effective clinical treatments. The majority of known METex14 alterations are typical donor/acceptor splicing or ubiquitination site mutations. Herein, two new METex14 variants were detected in two patients with lung adenocarcinoma by targeted next generation sequencing (NGS). Reverse transcription (RT)-based analysis confirmed that these mutations led to MET exon 14 skipping. Our analysis provided evidence for possible targeted therapy options for patients carrying these MET mutations or similar METex14 analogs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    由于突破性的发展和不断进步,晚期和转移性非小细胞肺癌(NSCLC)的治疗已经成为一个令人兴奋的,但越来越具有挑战性的任务。这适用于,特别是,具有致癌驱动改变的非小细胞肺癌亚组。虽然用各种酪氨酸激酶抑制剂(TKIs)治疗表皮生长因子受体(EGFR)突变和间变性淋巴瘤激酶(ALK)重排的NSCLC已经确立,在过去的几年中,已经确定了新的目标,并在临床实践中引入了新的TKIs。即使是KRAS突变,长期以来被认为是“不可定位”的变更,有希望的新药正在出现。抗性机制的检测和深入的分子分析进一步推动了新的治疗策略的发展。这篇综述的目的是对NSCLC中可靶向致癌改变的当前景观进行全面概述。
    Due to groundbreaking developments and continuous progress, the treatment of advanced and metastatic non-small cell lung cancer (NSCLC) has become an exciting, but increasingly challenging task. This applies, in particular, to the subgroup of NSCLC with oncogenic driver alterations. While the treatment of epidermal growth factor receptor (EGFR)-mutated and anaplastic lymphoma kinase (ALK)-rearranged NSCLC with various tyrosine kinase inhibitors (TKIs) is well-established, new targets have been identified in the last few years and new TKIs introduced in clinical practice. Even for KRAS mutations, considered for a long time as an \"un-targetable\" alteration, promising new drugs are emerging. The detection and in-depth molecular analysis of resistance mechanisms has further fueled the development of new therapeutic strategies. The objective of this review is to give a comprehensive overview on the current landscape of targetable oncogenic alterations in NSCLC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号