MET amplification

MET 扩增
  • 文章类型: Editorial
    暂无摘要。
    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的扩增是EGFR突变型非小细胞肺癌(NSCLC)对靶向治疗的获得性耐药的主要原因。只能被MET抑制剂的部分疗效暂时抑制。这项研究表明,由于扩增的MET在Wnt/β-catenin信号通路中触发了强大的正反馈回路,因此MET抑制剂的功效出乎意料地有限。即使MET途径再次被抑制,也允许最佳功能。为了检验这一推测并特异性靶向Wnt/β-catenin通路,一种巧妙设计的Wnt缩合前药称为WntSI是使用液液相分离(LLPS)驱动的可逆超分子自组装开发的。该过程涉及MET/pH响应肽(Tyr-Pep)和称为CA的有效Wnt抑制剂。在细胞中过表达的MET识别和磷酸化Tyr-Pep时,它破坏了LLPS倾向,促进了WntSI的解体。因此,这使得它能够抑制β-连环蛋白介导的致癌作用,在细胞系来源和患者来源的肿瘤异种移植(PDX)小鼠模型中,有效克服了由MET扩增引起的对EGFR-TKIs的获得性耐药性,同时保持了出色的生物安全性。这种有效的策略不仅选择性地抑制了Wnt/β-catenin信号通路,但也是通过生物响应性LLPS开发前药的创新范例。
    The amplification of MET is a major cause of acquired resistance to targeted therapy in EGFR-mutant non-small-cell lung cancer (NSCLC), only to be temporarily restrained by the partial efficacy of MET inhibitors. This study reveals that the MET inhibitor has unexpectedly limited efficacy due to amplified MET triggering a strong positive feedback loop in the Wnt/β-catenin signaling pathway, allowing optimal functionality even when the MET pathway is suppressed again. To test this conjecture and specifically target the Wnt/β-catenin pathway, a cleverly designed Wnt condensative pro drug called WntSI is developed using reversible supramolecular self-assembly driven by liquidliquid phase separation (LLPS). This process involves a MET/pH-responsive peptide (Tyr-Pep) and a potent Wnt inhibitor known as CA. Upon recognition and phosphorylation of Tyr-Pep by over expressed MET in cells, it disrupts LLPS propensity and facilitates the disintegration of WntSI. Consequently,this enables it to suppress the carcinogenic effect mediated by β-catenin,effectively overcoming acquired resistance to EGFR-TKIs caused by MET amplification in both cell line-derived and patient-derived tumor xenograft (PDX) mouse models while maintaining exceptional biosecurity. This effective strategy not only suppresses the Wnt/β-catenin signaling pathway selectively, but also serves as an innovative example for pro-drug development through biologically responsive LLPS.
    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)酪氨酸激酶受体的突变或扩增会导致非小细胞肺癌(NSCLC)中受体功能失调并刺激肿瘤生长,最常见的突变是MET外显子14(METex14)。我们试图比较MET改变的NSCLC与MET野生型NSCLC的基因组和免疫前景。
    方法:用TempusxT法对18,047例NSCLC肿瘤进行测序。根据MET外显子14(METex14)突变对肿瘤进行分类;低MET扩增定义为拷贝数增加(CNG)6-9,高MET扩增定义为CNG≥10,以及MET其他类型的突变。在MET改变的和MET野生型组中比较了免疫肿瘤学(IO)生物标志物和其他体细胞基因改变的频率。
    结果:276(1.53%)METex14,138(0.76%)高METamp,63(0.35%)低METamp,27(0.15%)其他MET,并鉴定出17,543(97%)MET野生型。包括METex14在内的任何MET突变的患者年龄较大,而METex14患者更常见的是女性和非吸烟者。MET基因在METamp肿瘤中表达最高。MET改变组的PD-L1阳性率高于MET野生型。METex14表现出最低的肿瘤突变负荷(TMB)和最低的新抗原肿瘤负荷(NTB)。METamp的CD4T细胞比例最低,NK细胞比例最高。METex14和METex14之间的共同改变存在显着差异。
    结论:与非METex14NSCLC肿瘤相比,METex14肿瘤在IO生物标志物和躯体景观方面表现出差异。免疫谱的变化可以影响MET改变的NSCLC中的免疫治疗选择,需要进一步探索。
    BACKGROUND: Mutation or amplification of the mesenchymal-epithelial transition (MET) tyrosine kinase receptor causes dysregulation of receptor function and stimulates tumor growth in non-small cell lung cancer (NSCLC) with the most common mutation being MET exon 14 (METex14). We sought to compare the genomic and immune landscape of MET-altered NSCLC with MET wild-type NSCLC.
    METHODS: 18,047 NSCLC tumors were sequenced with Tempus xT assay. Tumors were categorized based on MET exon 14 (METex14) mutations; low MET amplification defined as a copy number gain (CNG) 6-9, high MET amplification defined as CNG ≥ 10, and MET other type mutations. Immuno-oncology (IO) biomarkers and the frequency of other somatic gene alterations were compared across MET-altered and MET wild-type groups.
    RESULTS: 276 (1.53%) METex14, 138 (0.76%) high METamp, 63 (0.35%) low METamp, 27 (0.15%) MET other, and 17,543 (97%) MET wild-type were identified. Patients with any MET mutation including METex14 were older, while patients with METex14 were more frequently female and nonsmokers. MET gene expression was highest in METamp tumors. PD-L1 positivity rates were higher in MET-altered groups than MET wild-type. METex14 exhibited the lowest tumor mutational burden (TMB) and lowest neoantigen tumor burden (NTB). METamp exhibited the lowest proportion of CD4 T cells and the highest proportion of NK cells. There were significant differences in co-alterations between METamp and METex14.
    CONCLUSIONS: METex14 tumors exhibited differences in IO biomarkers and the somatic landscape compared to non-METex14 NSCLC tumors. Variations in immune profiles can affect immunotherapy selection in MET-altered NSCLC and require further exploration.
    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:3-4%的非小细胞肺癌(NSCLC)患者发生MET外显子14跳跃突变,<1%的患者发生MET高扩增。克唑替尼,一种选择性ATP竞争性小分子c-Met抑制剂,ALK,和ROS1酪氨酸激酶,在具有各种类型的MET激活的癌症模型中显示出活性。
    方法:Co-MET研究是一项单臂2期试验,用于评估克唑替尼在MET抑制剂初治的晚期NSCLC患者中的安全性和有效性MET外显子14跳跃突变(队列1)或高MET基因拷贝数≥7(队列2)。主要终点是队列1中根据RECISTv1.1的客观缓解率(ORR)。关键的次要终点是反应持续时间(DoR),无进展生存期(PFS),总生存期(OS),和安全。
    结果:2018年3月至2020年2月,共纳入28例患者(队列1中23例,队列2中5例)。主要终点符合队列1的ORR(90%置信区间:CI)为38.1%(20.6-58.3)。中位数DoR,PFS,OS(95%CI)为7.6(1.9-NE),5.7(2.1-11.3),9.1(4.0-19.9)个月,分别,在队列1中。队列2的ORR为40.0%(18.9-92.4)。安全性信号通常与克唑替尼的已知安全性特征一致。
    结论:克唑替尼在携带MET外显子14跳跃突变的非小细胞肺癌患者中的临床活性与替替尼和卡马替尼相似。
    背景:UMIN000031623.
    BACKGROUND: MET exon 14 skipping mutations occur in 3-4% and MET high amplifications occur in < 1% of patients with non-small-cell lung cancer (NSCLC). Crizotinib, a selective ATP-competitive small-molecule inhibitor of c-Met, ALK, and ROS1 tyrosine kinases, has shown activity in cancer models with various types of MET activation.
    METHODS: The Co-MET study is a single-arm phase 2 trial to assess the safety and efficacy of crizotinib in MET inhibitor-naïve patients with advanced NSCLC harboring MET exon 14 skipping mutation (cohort 1) or high MET gene copy number of ≥ 7 (cohort 2). The primary endpoint was the objective response rate (ORR) per RECIST v1.1 by independent radiology review in cohort 1. The key secondary endpoints were the duration of response (DoR), progression-free survival (PFS), overall survival (OS), and safety.
    RESULTS: A total of 28 patients (23 in cohort 1 and 5 in cohort 2) were enrolled between March 2018 and February 2020. The primary endpoint was met as the ORR (90% confidence interval: CI) in cohort 1 was 38.1% (20.6-58.3). Median DoR, PFS, and OS (95% CI) were 7.6 (1.9-NE), 5.7 (2.1-11.3), 9.1 (4.0-19.9) months, respectively, in cohort 1. ORR in cohort 2 was 40.0% (18.9-92.4). The safety signals were generally consistent with the known safety profile of crizotinib.
    CONCLUSIONS: Crizotinib showed a clinical activity similar to that of tepotinib and capmatinib in patients with NSCLC harboring MET exon 14 skipping mutations.
    BACKGROUND: UMIN000031623.
    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
    胆囊起源的神经内分泌癌(NEC)特别罕见,仅占原发性胆囊恶性肿瘤的0.38%,标准疗法是有限的。MET基因编码酪氨酸激酶受体,C-Met.MET的致病变异,如MET外显子14跳跃和MET扩增,导致过度的下游信号,促进肿瘤进展。一种MET抑制剂,卡马替尼,阻断c-Met的信号传导,并已被食品和药物管理局批准用于具有MET外显子14跳跃的非小细胞肺癌。据报道,卡马替尼在其他具有MET扩增的癌症中的有效性,但尚未报道具有MET变体的NEC。这里,我们介绍了一个72岁的女性胆囊的NEC与多个肝脏和淋巴结转移,对常规化疗耐药,包括卡铂加依托泊苷作为一线治疗和伊立替康作为二线治疗,但她对卡马替尼有反应.治疗6周后,CT扫描显示部分反应(尺寸减少80%),但13周后,观察到肝转移的再生长。在这里,我们报告了卡马替尼对MET扩增的胆囊源性NEC患者的有意义疗效.
    Neuroendocrine carcinoma (NEC) of the gallbladder origin is particularly rare, accounting for only 0.38% of primary malignancies of the gallbladder, and standard therapies are limited. The MET gene encodes the tyrosine kinase receptor, c-Met. Pathogenic variants of MET, such as MET exon 14 skipping and MET amplification, result in excessive downstream signaling that promotes tumor progression. A MET inhibitor, capmatinib, blocks signaling of c-Met and has been approved by the Food and Drug Administration for non-small cell lung cancer with MET exon 14 skipping. The effectiveness of capmatinib has been reported in other cancers with MET amplification, but NEC with MET variants has not been reported. Here, we present a case of a 72-year-old woman with NEC of the gallbladder with multiple liver and lymph node metastases, who was resistant to conventional chemotherapy including carboplatin plus etoposide as first-line treatment and irinotecan as second-line treatment, but she responded to capmatinib. After 6 weeks of treatment, CT scan showed a partial response (80% reduction in size), but after 13 weeks, regrowth of liver metastasis was observed. Herein, we report a meaningful efficacy of capmatinib to the patient of NEC of the gallbladder origin with MET amplification.
    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
    精准医学已帮助确定了几种肿瘤分子畸变,以进行靶向治疗。这些疗法在具有晚期癌症的特定致癌驱动因素的患者中显示出功效的实质性改善而没有过度毒性。在转移性肺癌中,广泛的分子肿瘤测序平台的实施帮助肿瘤学提供者确定了在预先接受靶向治疗时与更好结局相关的致癌驱动因素.间充质-上皮转化因子(MET)改变存在于高达60%的非小细胞肺癌中,并与不良预后相关。Capmatinib和tepotinib是目前美国食品和药物管理局(FDA)批准的针对MET外显子14跳跃突变患者的两种靶向治疗方法。正在开发几种药物来解决MET改变患者的未满足需求。这些药物中的一些与EGFR靶向治疗组合使用以减轻对EGFR抑制剂的抗性。这些药物有望为这些患者提供新的希望。
    Precision medicine has helped identify several tumor molecular aberrations to be treated with targeted therapies. These therapies showed substantial improvement in efficacy without excessive toxicity in patients with specific oncogenic drivers with advanced cancers. In metastatic lung cancers, the implementation of broad platforms for molecular tumor sequencing has helped oncology providers identify oncogenic drivers linked with better outcomes when treated upfront with targeted therapies. Mesenchymal-epithelial transition factor (MET) alterations are present in up to 60% of non-small cell lung cancer and are associated with a poor prognosis. Capmatinib and tepotinib are currently the only two approved targeted therapies by the U.S. Food and Drug Administration (FDA) for patients with MET exon 14 skipping mutation. Several agents are being developed to tackle an unmet need in patients with MET alterations. Some of these agents are being used in combination with EGFR targeted therapy to mitigate resistance to EGFR inhibitor. These agents are poised to provide new hope for these patients.
    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
    针对表皮生长因子受体(EGFR)和间充质上皮转化(MET)的双靶向治疗(DTT)的一些研究在非小细胞肺癌(NSCLC)中显示出有希望的疗效。因此,DTT抵抗后的患者管理具有重要意义。然而,这些患者的潜在耐药机制和临床结局尚不清楚.
    本研究旨在描述携带EGFR突变并在对DTT产生耐药性后获得MET扩增的NSCLC患者的分子特征和生存结果。
    我们对EGFR突变和获得性MET扩增的NSCLC患者进行了回顾性分析,这些患者表现出对EGFR/METDTT的耐药性。
    在对DTT产生耐药性之前和/或之后,对具有可用组织样本的患者进行了下一代测序(NGS)。根据数据源和随后的抢救处理进行分层分析。采用单变量/多变量Cox回归模型和生存分析来探索潜在的独立预后因素。
    该研究包括77位非小细胞肺癌患者,对19名患者进行了NGS。我们观察到许多抗性机制,包括EGFR依赖性通路(4/19,21.1%),MET依赖性途径(2/19,10.5%),EGFR/MET共同依赖途径(2/19,10.5%),和EGFR/MET非依赖性耐药机制(11/19,57.9%)。接受最佳支持治疗(BSC)的患者的进展后无进展生存期(pPFS)和进展后总生存期(pOS)显着不同,靶向治疗,或化疗(CT),pPFS中位数为1.5、3.9和4.9个月,分别(p=0.003)。中位数pOS分别为2.3、7.7和9.2个月,分别(p<0.001)。DTT耐药后的治疗线数量和东部肿瘤协作组的表现状态成为独立的预后因素。
    这项研究揭示了EGFR/METDTT耐药机制的异质性,与类似的流行率的目标和脱靶机制。靶向治疗或CT,与BSC相比,显示出改善DTT耐药后晚期NSCLC患者生存结局的潜力。
    UNASSIGNED: Some studies of dual-targeted therapy (DTT) targeting epidermal growth factor receptor (EGFR) and mesenchymal-epithelial transition (MET) have shown promising efficacy in non-small-cell lung cancer (NSCLC). Consequently, patient management following DTT resistance has gained significance. However, the underlying resistance mechanisms and clinical outcomes in these patients remain unclear.
    UNASSIGNED: This study aimed to delineate the molecular characteristics and survival outcomes of patients with NSCLC harboring EGFR mutations and acquired MET amplification after developing resistance to DTT.
    UNASSIGNED: We conducted a retrospective analysis of patients with NSCLC with EGFR mutations and acquired MET amplification who exhibited resistance to EGFR/MET DTT.
    UNASSIGNED: Next-generation sequencing (NGS) was performed on patients with available tissue samples before and/or after the development of resistance to DTT. Stratified analyses were carried out based on data sources and subsequent salvage treatments. Univariate/multivariate Cox regression models and survival analyses were employed to explore potential independent prognostic factors.
    UNASSIGNED: The study included 77 NSCLC patients, with NGS conducted on 19 patients. We observed many resistance mechanisms, including EGFR-dependent pathways (4/19, 21.1%), MET-dependent pathways (2/19, 10.5%), EGFR/MET co-dependent pathways (2/19, 10.5%), and EGFR/MET-independent resistance mechanisms (11/19, 57.9%). Post-progression progression-free survival (pPFS) and post-progression overall survival (pOS) significantly varied among patients who received the best supportive care (BSC), targeted therapy, or chemotherapy (CT), with median pPFS of 1.5, 3.9, and 4.9 months, respectively (p = 0.003). Median pOS were 2.3, 7.7, and 9.2 months, respectively (p < 0.001). The number of treatment lines following DTT resistance and the Eastern Cooperative Oncology Group performance status emerged as the independent prognostic factors.
    UNASSIGNED: This study revealed a heterogeneous landscape of resistance mechanisms to EGFR/MET DTT, with a similar prevalence of on- and off-target mechanisms. Targeted therapy or CT, as compared to BSC, exhibited the potential to improve survival outcomes for patients with advanced NSCLC following resistance to DTT.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Clinical Trial, Phase II
    目的:间充质上皮转化因子(MET)的信号调节异常和AXL激活增强与非小细胞肺癌(NSCLC)的发病机制有关。格利沙替尼(MGCD265)是一种研究性的,MET和AXL的口服抑制剂。
    方法:这个开放标签,II期研究调查了晚期患者的格利沙替尼(游离碱悬浮液[FBS]胶囊1050mgBID或喷雾干燥分散体[SDD]片剂750mgBID),根据肿瘤或ctDNA中是否存在MET激活突变或扩增,将之前治疗过的NSCLC分为4个队列.主要终点是客观缓解率(ORR)。
    结果:68例患者入选:n=28和n=8,肿瘤组织和ctDNA中MET外显子14跳跃突变,分别,n=20和n=12,在肿瘤组织和ctDNA中进行MET基因扩增,分别。总的来说,ORR为11.8%,中位无进展生存期为4.0个月,中位总生存期为7.0个月.在MET激活突变的患者中,肿瘤检测的ORR为10.7%,ctDNA检测的ORR为25.0%。对于MET扩增,仅在通过肿瘤检测纳入的患者中观察到应答(ORR15.0%).腹泻(82.4%),恶心(50.0%),丙氨酸转氨酶增加(41.2%),疲劳(38.2%),天冬氨酸转氨酶升高(36.8%)是与研究药物相关的最常见的不良事件.格列沙替尼暴露与SDD片剂和FBS胶囊制剂相似。由于适度的临床活动,赞助商提前终止了该研究。
    结论:格来替尼在晚期患者中具有可接受的安全性,具有MET激活改变的预处理的NSCLC。观察到适度的临床活动,这可能反映了先前报道的I期数据所暗示的次优药物生物利用度,和药效学发现低于预期的循环可溶性脱落MET胞外域(s-MET)的增加。
    OBJECTIVE: Dysregulated signaling by mesenchymal epithelial transition factor (MET) and heightened AXL activation are implicated in the pathogenesis of non-small cell lung cancer (NSCLC). Glesatinib (MGCD265) is an investigational, oral inhibitor of MET and AXL.
    METHODS: This open-label, Phase II study investigated glesatinib (free-base suspension [FBS] capsule 1050 mg BID or spray-dried dispersion [SDD] tablet 750 mg BID) in patients with advanced, previously treated NSCLC across four cohorts grouped according to presence of MET activating mutations or amplification in tumor or ctDNA. The primary endpoint was objective response rate (ORR).
    RESULTS: Sixty-eight patients were enrolled: n = 28 and n = 8 with MET exon 14 skipping mutations in tumor tissue and ctDNA, respectively, and n = 20 and n = 12 with MET gene amplification in tumor tissue and ctDNA, respectively. Overall, ORR was 11.8 %, median progression-free survival was 4.0 months, and median overall survival was 7.0 months. Among patients with MET activating mutations, ORR was 10.7 % with tumor testing and 25.0 % with ctDNA testing. For MET amplification, responses were observed only in patients enrolled by tumor testing (ORR 15.0 %). Diarrhea (82.4 %), nausea (50.0 %), increased alanine aminotransferase (41.2 %), fatigue (38.2 %), and increased aspartate aminotransferase (36.8 %) were the most frequent adverse events assessed as related to study medication. Glesatinib exposure was similar with the SDD tablet and FBS capsule formulations. The study was terminated early by the sponsor due to modest clinical activity.
    CONCLUSIONS: Glesatinib had an acceptable safety profile in patients with advanced, pre-treated NSCLC with MET activating alterations. Modest clinical activity was observed, which likely reflects suboptimal drug bioavailability suggested by previously reported Phase I data, and pharmacodynamic findings of lower than anticipated increases in circulating soluble shed MET ectodomain (s-MET).
    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)

公众号