MET amplification

MET 扩增
  • 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: Case Reports
    胆管癌(CCA)是一种侵袭性疾病,治疗选择有限。尽管努力探索更好的治疗方案,以吉西他滨为基础的化疗几十年来一直是标准的一线治疗方法.随着精准医学领域的不断发展,生物标志物指导的治疗越来越受欢迎。MET改变在各种癌症类型中经常发生,使它成为一个有希望的目标。
    一名53岁的男子主诉上腹痛到我院就诊。根据转移性淋巴结的活检和免疫组织化学诊断晚期CCA。下一代测序显示MET扩增。由于患者对传统化疗不耐受,给予savolitinib(c-MET抑制剂).取得了部分反应,治疗耐受性良好。一年后,病人发展为进行性疾病,表皮生长因子受体扩增的出现可能对此有贡献。
    我们的研究验证了c-MET抑制剂在晚期CCA-harordingMET扩增中的治疗价值,并为对化疗不耐受的患者提供了替代策略。
    UNASSIGNED: Cholangiocarcinoma (CCA) is an aggressive disease with limited treatment options. Despite substantial efforts to explore better regimens, gemcitabine-based chemotherapy has been the standard first-line treatment for decades. With the growing field of precision medicine, biomarker-guided treatments are gaining popularity. MET alteration is a frequent occurrence in various cancer types, making it a promising target.
    UNASSIGNED: A 53-year-old man visited our hospital with a complaint of upper abdominal pain. Advanced CCA was diagnosed based on the biopsy of the metastatic lymph nodes and immunohistochemistry. Next-generation sequencing revealed MET amplification. As the patient was intolerant to traditional chemotherapy, savolitinib (a c-MET inhibitor) was administered. Partial response was achieved, and the treatment was well tolerated. After 1 year, the patient developed progressive disease, to which the emergence of epidermal growth factor receptor amplification may have contributed.
    UNASSIGNED: Our study verified the therapeutic value of a c-MET inhibitor in advanced CCA-harboring MET amplification and provides an alternative strategy for patients who are intolerant to chemotherapy.
    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
    利用针对激活突变的靶向治疗为晚期非小细胞肺癌(NSCLC)患者开辟了治疗范式的新时代。对于表皮生长因子(EGFR)突变的癌症患者,EGFR抑制剂,包括第三代酪氨酸激酶抑制剂(TKI)奥希替尼,显著延长无进展生存期和总生存期,是目前的护理标准。然而,EGFR抑制后总是发生进展,进一步的研究有助于阐明抗性机制。间质上皮转化(MET)致癌途径的异常被认为是进展后的常见改变,MET扩增是最常见的机制之一。对MET具有抑制活性的多种药物,包括TKIs,抗体,和抗体-药物缀合物,已在晚期NSCLC中进行了开发和研究。对于发现具有MET驱动的耐药机制的患者,将MET和EGFR结合是一种有希望的治疗策略。TKI联合治疗和EGFR-MET双特异性抗体在早期临床试验中显示出有希望的抗肿瘤活性。未来的研究,包括正在进行的大规模EGFR-MET联合抑制试验,将有助于澄清针对EGFR耐药背后的这种机制是否对晚期EGFR突变的NSCLC患者具有有意义的临床益处。
    Utilizing targeted therapy against activating mutations has opened a new era of treatment paradigms for patients with advanced non-small cell lung cancer (NSCLC). For patients with epidermal growth factor (EGFR)-mutated cancers, EGFR inhibitors, including the third-generation tyrosine kinase inhibitor (TKI) osimertinib, significantly prolong progression-free survival and overall survival, and are the current standard of care. However, progression after EGFR inhibition invariably occurs, and further study has helped elucidate mechanisms of resistance. Abnormalities in the mesenchymal-epithelial transition (MET) oncogenic pathway have been implicated as common alterations after progression, with MET amplification as one of the most frequent mechanisms. Multiple drugs with inhibitory activity against MET, including TKIs, antibodies, and antibody-drug conjugates, have been developed and studied in advanced NSCLC. Combining MET and EGFR is a promising treatment strategy for patients found to have a MET-driven resistance mechanism. Combination TKI therapy and EGFR-MET bispecific antibodies have shown promising anti-tumor activity in early clinical trials. Future study including ongoing large-scale trials of combination EGFR-MET inhibition will help clarify if targeting this mechanism behind EGFR resistance will have meaningful clinical benefit for patients with advanced EGFR-mutated 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: Case Reports
    临床前病例表明,EGFR酪氨酸激酶抑制剂(TKIs)加METTKIs是治疗具有MET扩增获得性耐药的非经典EGFR突变肺癌的潜在疗法。在这里,我们首次报道了EGFRG719X/S768I/L861Q患者新型联合治疗方案的有效性.直到最后一次后续评估,两名患者在改用阿法替尼联合萨沃利替尼(PFS:10个月)和呋喃替尼联合克唑替尼(PFS:6个月)后,生存率得到改善,分别,未观察到不良事件的发生率和严重程度增加。根据本研究的结果和文献综述,在EGFR突变和MET扩增不常见的NSCLC患者中,观察到联合治疗的各种缓解.此外,下一代测序(NGS)导致发现不常见的EGFR,并揭示NSCLC中的共突变。
    Preclinical cases suggest that EGFR tyrosine kinase inhibitors (TKIs) plus MET TKIs are a potential therapy for non-classical EGFR mutant lung cancers with MET amplification acquired resistance. Herein, we report for the first time the effectiveness of novel combination treatment regimens for patients with EGFR G719X/S768I/L861Q. Until the last follow-up assessment, two patients demonstrated improved survival after they switched to afatinib combined with savolitinib (PFS: 10 months) and furmonertinib combined with crizotinib (PFS: 6 months), respectively, that did not observed increased incidence and severity of adverse events. According to the findings of this study and literature review, various responses were observed from the combined therapy in NSCLC patients who harbored uncommon EGFR mutations and MET amplification. Furthermore, Next generation sequencing (NGS) leads to the discovery of uncommon of EGFR and reveals the co-mutations 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: Case Reports
    胆管癌是一种高度病态的胃肠道恶性肿瘤,可用的治疗方法有限。护理标准包括细胞毒性化疗,如吉西他滨,铂剂,nab-紫杉醇,和氟嘧啶类似物.然而,这些方案的耐受性各不相同,不耐受化疗的患者的靶向治疗和免疫治疗选择有限.在胆管癌中,间充质上皮转化因子(MET)扩增可能为靶向治疗方法提供额外的机会,特别是考虑到非小细胞肺癌的新数据。在这种情况下,我们介绍了一名具有高水平MET基因扩增的转移性胆管癌患者,一种具有抗c-MET活性的酪氨酸激酶抑制剂,在化疗停止后提供了部分反应。
    Cholangiocarcinoma is a highly morbid gastrointestinal malignancy for which available therapies are limited. Standard of care includes cytotoxic chemotherapies such as gemcitabine, platinum agents, nab-paclitaxel, and fluoropyrimidine analogues. However, tolerability of these regimens varies, and patients who do not tolerate chemotherapy have limited targeted therapies and immunotherapy options. In cholangiocarcinoma, mesenchymal-epithelial transition factor (MET) amplification may present an additional opportunity for a targeted therapeutic approach, especially considering emerging data in non-small cell lung cancer. In this case, we present a metastatic cholangiocarcinoma patient with high-level MET gene amplification for whom capmatinib, a tyrosine kinase inhibitor with activity against c-MET, provided a partial response after cessation of chemotherapy.
    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
    副肿瘤性嗜酸性粒细胞增多是在1%实体瘤病例中观察到的罕见并发症,并且似乎具有肿瘤类型依赖性预后影响。其中嗜酸性粒细胞计数增加通常与不良预后相关。在英国文学中,超过20例患者在诊断时被报道与原发性非小细胞肺癌(NSCLC)相关的嗜酸性粒细胞增多,所有这些人都接受了两种手术,化疗,或对症治疗。在这里,我们描述了一个有副肿瘤嗜酸性粒细胞增多和白细胞增多并接受靶向治疗的IV期NSCLC患者的临床过程.一名64岁的男性前吸烟者被诊断为具有EGFRL858R突变和MET扩增的肺腺癌。血液嗜酸性粒细胞增多在诊断时表现出来,并通过消除其他潜在原因证实为副肿瘤。该疾病在使用EGFR抑制剂埃克替尼的一个月内迅速进展,然后在第一个月内快速反应后,在三个月内使用埃克替尼加克唑替尼。加入多靶点激酶抑制剂安洛替尼,尽管最初自我报告为无症状的高性能状态,但疾病在一个月后进展。患者失去了随后的随访。疾病控制或进展的影像学评估与嗜酸性粒细胞增多的各自明显缓解或恶化相吻合。与以前关于血液嗜酸性粒细胞增多相关的不良临床结局的报道一致,我们的结果提示EGFR-/MET改变的NSCLC对预后有负面影响.这个案子是,据我们所知,首次提供血液嗜酸性粒细胞增多与EGFR和MET改变的肺腺癌在靶向治疗下的疾病进展平行的证据,这表明血液嗜酸性粒细胞增多对驾驶员阳性NSCLC的预后有负面影响。
    Paraneoplastic eosinophilia is a rare complication observed in 1% solid tumor cases and appears to have tumor type-dependent prognostic impact, in which the increased eosinophil count was generally associated with unfavorable prognosis. In the English literature, more than 20 patients have been reported of eosinophilia associated with primary non-small cell lung cancer (NSCLC) at diagnosis, all of whom underwent either surgery, chemotherapy, or symptomatic therapy. Herein, we describe clinical course a stage IV NSCLC patient with paraneoplastic eosinophilia and leukocytosis and receiving targeted therapy. A 64-year-old male former smoker was diagnosed with lung adenocarcinoma harboring EGFR L858R mutation and MET amplification. Blood eosinophilia was manifested at diagnosis and confirmed to be paraneoplastic by eliminating other potential causes. The disease progressed rapidly within a month on EGFR inhibitor icotinib and then within three months on icotinib plus crizotinib after rapid response within the first month. A multi-target kinase inhibitor anlotinib was added, and the disease progressed one month later despite initial self-reported asymptomatic high-performance status. The patient was lost to subsequent follow-ups. Radiographic evaluation of disease control or progression coincided with respective distinct alleviation or worsening of eosinophilia. Consistent with previous reports of poor clinical outcome associated with blood eosinophilia, our results suggested a negative prognostic impact in EGFR-/MET-altered NSCLC. This case is, to the best of our knowledge, the first to provide evidence for blood eosinophilia paralleling disease progression in an EGFR- and MET-altered lung adenocarcinoma under targeted therapy, which suggested negative prognostic impact of blood eosinophilia in driver-positive NSCLC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号