Driver oncogene

驱动癌基因
  • 文章类型: Journal Article
    背景:本研究旨在评估非小细胞肺癌(NSCLC)一线全身治疗进展的现实世界影响。关注驱动基因突变和程序性死亡配体1(PD-L1)表达水平的作用。
    方法:在日本八家医疗机构进行,这个多中心,回顾性观察性研究纳入了2015年1月至2022年12月间诊断为NSCLC并接受治疗的863例患者.根据接受的全身治疗类型对患者进行分类:细胞毒性药物,分子靶向剂,免疫检查点抑制剂,和组合疗法。进行了全面的分子和免疫组织化学分析,并进行统计评估。
    结果:中位总生存期(OS)显示治疗组之间存在显着差异,靶向治疗显示最长的OS。这项研究还表明,在使用免疫检查点抑制剂治疗的组中,高PD-L1表达是常见的。多因素分析用于确定抗癌药物的类型和诊断时PD-L1的表达作为影响5年OS的影响变量。
    结论:本研究强调了靶向治疗的有效性以及综合分子诊断和PD-L1表达在影响NSCLC患者OS中的关键作用。倡导将其融入常规临床实践。
    BACKGROUND: This study aims to assess the real-world impact of advancements in first-line systemic therapies for non-small-cell lung cancer (NSCLC), focusing on the role of driver gene mutations and programmed death-ligand 1 (PD-L1) expression levels.
    METHODS: Conducted across eight medical facilities in Japan, this multicenter, retrospective observational research included 863 patients diagnosed with NSCLC and treated between January 2015 and December 2022. The patients were categorized based on the type of systemic therapy received: cytotoxic agents, molecular targeting agents, immune checkpoint inhibitors, and combination therapies. Comprehensive molecular and immunohistochemical analyses were conducted, and statistical evaluations were performed.
    RESULTS: The median overall survival (OS) shows significant variations among treatment groups, with targeted therapies demonstrating the longest OS. This study also revealed that high PD-L1 expression was common in the group treated with immune checkpoint inhibitors. Multivariate analysis was used to identify the type of anticancer drug and the expression of PD-L1 at diagnosis as the impactful variables affecting 5-year OS.
    CONCLUSIONS: This study underscores the efficacy of targeted therapies and the critical role of comprehensive molecular diagnostics and PD-L1 expression in affecting OS in NSCLC patients, advocating for their integration into routine clinical practice.
    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
    KRAS是晚期最常见的突变癌基因,非鳞状,西方国家的非小细胞肺癌(NSCLC)。在各种KRAS突变体中,KRASG12C是最常见的变体(约40%),占晚期非鳞状细胞肺癌的10-13%。最近监管批准的KRASG12C选择性抑制剂sotorasib和adagrasib用于携带KRASG12C的晚期或转移性NSCLC患者,已将KRAS转化为可药用靶标。在这次审查中,我们探讨了KRAS在晚期NSCLC中从预后到预测性生物标志物的演变作用,讨论KRASG12C生物学,真实世界的患病率,共突变的临床相关性,和分子测试方法。真实世界的证据表明,KRASG12C患病率存在显著的地理差异(美国为8.9%-19.5%,欧洲为9.3-18.4%,拉丁美洲为6.9-9.0%,亚洲为1.4-4.3%)在晚期非小细胞肺癌中。此外,与STK11,KEAP1和TP53等KRASG12C共突变相关的临床数据正在增加.在现实世界的证据中,KRASG12C突变NSCLC与STK11,KEAP1和TP53共突变相关,占10.3-28.0%,6.3-23.0%,17.8-50.0%的患者,分别。虽然sotorasib和adagrasib目前已被批准用于二线治疗及以上的晚期/转移性NSCLC患者,KRASG12C变异体的检测和报告应包括在一线治疗前的常规生物标志物检测中.KRASG12C测试结果应清楚记录在患者的健康记录中,说明进展时的可操作性。如有,建议使用下一代测序,以促进在单次运行中同时检测潜在可行的生物标志物,从而保护组织.分子检测结果应告知治疗KRASG12C突变的晚期NSCLC患者的临床决策。
    KRAS is the most commonly mutated oncogene in advanced, non-squamous, non-small cell lung cancer (NSCLC) in Western countries. Of the various KRAS mutants, KRAS G12C is the most common variant (~40%), representing 10-13% of advanced non-squamous NSCLC. Recent regulatory approvals of the KRASG12C-selective inhibitors sotorasib and adagrasib for patients with advanced or metastatic NSCLC harboring KRASG12C have transformed KRAS into a druggable target. In this review, we explore the evolving role of KRAS from a prognostic to a predictive biomarker in advanced NSCLC, discussing KRAS G12C biology, real-world prevalence, clinical relevance of co-mutations, and approaches to molecular testing. Real-world evidence demonstrates significant geographic differences in KRAS G12C prevalence (8.9-19.5% in the US, 9.3-18.4% in Europe, 6.9-9.0% in Latin America, and 1.4-4.3% in Asia) in advanced NSCLC. Additionally, the body of clinical data pertaining to KRAS G12C co-mutations such as STK11, KEAP1, and TP53 is increasing. In real-world evidence, KRAS G12C-mutant NSCLC was associated with STK11, KEAP1, and TP53 co-mutations in 10.3-28.0%, 6.3-23.0%, and 17.8-50.0% of patients, respectively. Whilst sotorasib and adagrasib are currently approved for use in the second-line setting and beyond for patients with advanced/metastatic NSCLC, testing and reporting of the KRAS G12C variant should be included in routine biomarker testing prior to first-line therapy. KRAS G12C test results should be clearly documented in patients\' health records for actionability at progression. Where available, next-generation sequencing is recommended to facilitate simultaneous testing of potentially actionable biomarkers in a single run to conserve tissue. Results from molecular testing should inform clinical decisions in treating patients with KRAS G12C-mutated 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: Observational Study
    背景:尽管由于患者的呼吸或一般状况,有时难以进行经支气管的诊断程序,内镜超声与支气管镜引导细针穿刺(EUS-B-FNA),已知的经食道诊断程序,可能对这种情况有用。我们进行了这项前瞻性三中心观察性研究,以评估EUS-B-FNA在呼吸或一般状况较差的疑似肺癌患者中的安全性和有效性。
    方法:疑似肺癌伴呼吸衰竭患者,东部肿瘤协作组表现为2或更高,或严重的呼吸道症状,已注册。主要终点是肺癌的诊断率及其安全性,次要终点是分子和程序性死亡配体1(PD-L1)分析的成功率,肺癌患者的6个月生存率。
    结果:我们招募了30名患者,其中29人被纳入分析。其中,26人最终诊断为肺癌。肺癌的诊断率为100%(26/26)。没有与需要停止手术的EUS-B-FNA相关的不良事件。EGFR分子分析的成功率,ALK,ROS-1和BRAF为100%(14/14),100%(11/11)100%(9/9)和75%(6/8),分别。PD-L1分析的成功率为100%(15/15)。肺癌患者的6个月生存率为53.8%(95%可信区间[CI]:33.4-76.4),中位总生存期(OS)为196天(95%CI:142-446)。
    结论:EUS-B-FNA是一种安全有效的诊断方法,即使是在怀疑患有呼吸不良或一般状况的肺癌患者中。
    背景:该临床试验在https://www注册。乌明。AC.jp/ctr/index。htm(UMIN000041235,2020年7月28日批准)。
    BACKGROUND: Although transbronchial diagnostic procedures are sometimes difficult to perform because of the patient\'s respiratory or general conditions, endoscopic ultrasound with bronchoscope-guided fine-needle aspiration (EUS-B-FNA), a known transesophageal diagnostic procedure, might be useful for such cases. We conducted this prospective three-center observational study to evaluate the safety and efficacy of EUS-B-FNA in suspected lung cancer patients with poor respiratory or general conditions.
    METHODS: Patients with suspected lung cancer with respiratory failure, Eastern Cooperative Oncology Group performance status of 2 or higher, or severe respiratory symptoms, were enrolled. The primary endpoints were the diagnostic yield of lung cancer and its safety, and the secondary endpoints were the success rate of molecular and programmed death ligand 1 (PD-L1) analyses, and the 6-month survival rate in patients with lung cancer.
    RESULTS: We enrolled 30 patients, of which 29 were included in the analysis. Among them, 26 were eventually diagnosed with lung cancer. The diagnostic yield for lung cancer was 100% (26/26). There were no adverse events associated with EUS-B-FNA requiring procedure discontinuation. The success rates of molecular analysis for EGFR, ALK, ROS-1, and BRAF were 100% (14/14), 100% (11/11), 100% (9/9), and 75% (6/8), respectively. The success rate of the PD-L1 analysis was 100% (15/15). The 6-month survival rate in patients with lung cancer was 53.8% (95% confidence interval [CI]: 33.4-76.4), and the median overall survival (OS) was 196 days (95% CI: 142-446).
    CONCLUSIONS: EUS-B-FNA is a safe and effective diagnostic method, even in patients with suspected lung cancer with poor respiratory or general conditions.
    BACKGROUND: This clinical trial was registered at https://www.umin.ac.jp/ctr/index.htm (UMIN000041235, approved on 28/07/2020).
    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
    程序性死亡配体1(PD-L1)表达是晚期肺腺癌中针对程序性死亡1/PD-L1的免疫疗法反应的预测性生物标志物。这项研究的目的是探讨PD-L1表达与临床病理特征之间的关系。预后,以及肺腺癌患者的驱动癌基因改变。
    我们评估了1,005例手术切除的肺腺癌标本中的PD-L1表达,通过免疫组织化学使用22C3抗体。PD-L1阳性根据组织微阵列上染色的肿瘤细胞(TPS)的比例定义:<1%(阴性),1-49%(弱正),≥50%(强阳性)。PD-L1表达与临床病理特征的相关性,预后,和驱动癌基因(EGFR,KRAS,ALK,分析了ROS1和RET)在肺腺癌中的改变。
    在1,005例肿瘤的726例(72%)中,PD-L1表达为阴性,161位(16%)弱阳性,和强积极的118(12%)。男性,吸烟,血清癌胚抗原水平升高,晚期病理阶段,高级别肿瘤,主要是实体瘤,有淋巴渗透或血管或胸膜浸润的肿瘤,无EGFR突变的肿瘤,与PD-L1阴性肿瘤患者(TPS<1%)相比,具有KRAS突变的肿瘤在PD-L1阳性肿瘤患者中更为常见.PD-L1阳性与ALK无关,ROS1或RET融合状态。尽管PD-L1阳性与所有患者的总体生存率低和无复发生存率低相关,在多变量分析中校正预后因素后,该结果无统计学意义.在根据驱动癌基因改变的亚组分析中,PD-L1阳性仅在EGFR突变肿瘤患者中与无复发生存不良相关。
    手术切除的PD-L1表达增加的肺腺癌是经常发生在男性吸烟者中的生物学侵袭性肿瘤。PD-L1表达及其预后意义因驱动癌基因改变而异。
    Programmed death-ligand 1 (PD-L1) expression is a predictive biomarker of response to immunotherapies targeting programmed death-1/PD-L1 in advanced-stage lung adenocarcinoma. The aim of this study was to investigate the associations between PD-L1 expression and clinicopathological features, prognosis, and driver oncogene alterations in patients with lung adenocarcinoma.
    We evaluated PD-L1 expression in 1,005 surgically resected lung adenocarcinoma specimens, by immunohistochemistry using the 22C3 antibody. PD-L1 positivity was defined based on the proportion of stained tumor cells (TPS) on tissue microarrays: <1% (negative), 1-49% (weakly positive), and ≥ 50% (strongly positive). Correlations between PD-L1 expression and clinicopathological features, prognosis, and driver oncogene (EGFR, KRAS, ALK, ROS1, and RET) alterations in lung adenocarcinoma were analyzed.
    PD-L1 expression was negative in 726 (72%) of 1,005 tumors, weakly positive in 161 (16%), and strongly positive in 118 (12%). Male sex, smoking, elevated serum carcinoembryonic antigen levels, advanced pathological stages, high-grade tumors, predominantly solid tumors, tumors with lymphatic permeation or vascular or pleural invasion, tumors without EGFR mutations, and tumors with KRAS mutations were more common in patients with PD-L1-positive tumors (TPS ≥ 1%) than in those with PD-L1-negative tumors (TPS < 1%). PD-L1 positivity was not associated with ALK, ROS1, or RET fusion status. Although PD-L1 positivity was associated with poor overall survival and poor relapse-free survival in all patients, this was not statistically significant after adjusting for prognostic factors in the multivariate analysis. In the subgroup analysis according to driver oncogene alterations, PD-L1 positivity was associated with poor relapse-free survival only in patients with EGFR-mutated tumors.
    Surgically resected lung adenocarcinomas with increased PD-L1 expression were biologically aggressive tumors that frequently occurred in male smokers. PD-L1 expression and its prognostic significance differed according to driver oncogene alterations.
    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
    由于呼吸道状况较差的肺癌患者很难通过气道观察获得肿瘤组织,内镜超声与支气管镜引导细针穿刺(EUS-B-FNA),经食管手术,对这类患者有效。我们在此报告了三名因肺癌相关症状而被送往急诊科的驱动癌基因患者。由于患者呼吸状况不佳,因此进行了EUS-B-FNA以检测驱动癌基因。一般情况有所改善,患者使用酪氨酸激酶抑制剂获得了长期生存。我们的发现表明,尽管急诊科的呼吸状况较差,但应考虑EUS-B-FNA检测肺癌患者的驱动癌基因。
    Since it is difficult to obtain tumor tissue via airway observation for lung cancer patients with a poor respiratory condition, endoscopic ultrasound with bronchoscope-guided fine-needle-aspiration (EUS-B-FNA), a transesophageal procedure, is effective for such patients. We herein report three patients with driver oncogenes taken to the emergency department because of lung cancer-related symptoms. EUS-B-FNA was performed because of the patients\' poor respiratory conditions to detect driver oncogenes. The general conditions improved, and the patients achieved a long-term survival with tyrosine kinase inhibitors. Our findings suggest that EUS-B-FNA should be considered to detect driver oncogenes in lung cancer patients despite poor respiratory conditions in emergency departments.
    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:已在癌症和该基因座的基因中观察到基因座8p11.23的扩增,包括ZNF703(锌指蛋白703),NSD3(核受体结合SET结构域蛋白3)和FGFR1(成纤维细胞生长因子受体1),已被提出作为显性癌基因,在具有扩增功能的癌症中赋予病理生理益处。然而,对于它们中的每一个或扩增子的任何其他基因的重要性没有共识,或者甚至对于哪些基因是扩增子的一部分没有共识。
    方法:使用公开数据来表征在8p11.23扩增的基因座,并获得关于每个基因和作为癌基因的作用的信息。在cBioportal平台中检查了基因座中扩增的频率,扩增病例中扩增子基因的表达水平来自平台中报道的基因组研究。使用K-M绘图仪检查基因座每个基因的mRNA表达对乳腺癌无复发生存的影响。
    结果:8p11.23扩增子在肺鳞癌中的出现频率较高,乳腺癌和膀胱癌,在其他癌症中很少观察到。扩增子内最频繁扩增的基因在不同类型的癌症之间变化。在乳腺癌中,放大病例最常见的是腔B型。扩增的基因并不总是过表达,并且在具有基因之间变异的扩增子基因中扩增与过表达的相关性低。扩增子的存在不影响乳腺癌的非整倍性评分或肿瘤突变负担。关于预后,mRNA高表达预示乳腺癌无复发不良生存的扩增子的两个基因是EIF4EBP1(真核转录起始因子4E结合蛋白1)和LSM1(LSM1同源物,mRNA降解相关)。
    结论:除了先前提出的在8p11.23癌症扩增基因座中作为主要癌基因发挥作用的基因,其他基因在乳腺癌中也可能是重要的,因为它们的扩增和mRNA表达的高度相关性以及过度表达导致的不良预后。与致癌作用一致。
    BACKGROUND: Amplification of the locus 8p11.23 has been observed in cancer and genes of this locus, including ZNF703 (Zinc finger protein 703), NSD3 (Nuclear receptor binding SET domain protein 3) and FGFR1 (Fibroblast growth factor receptor 1), have been put forward as dominant oncogenes conferring pathophysiologic benefit in cancers with amplifications. However, there is no consensus on the importance of each of them or any other genes of the amplicon or even a consensus on which genes are part of the amplicon.
    METHODS: Publicly available data were used to characterize the locus amplified at 8p11.23 and derive information on each of the genes and roles as oncogenes. The frequency of the amplifications in the locus was examined in the cBioportal platform, and expression levels of the amplicon genes in amplified cases were derived from genomic studies reported in the platform. Examination of the influence of mRNA expressions of each gene of the locus for Recurrence-free survival in breast cancer was performed using K-M plotter.
    RESULTS: The 8p11.23 amplicon is present in higher frequency in squamous cell lung carcinomas, breast cancers and bladder carcinomas and is only rarely observed in other cancers. The most frequently amplified genes within the amplicon vary between different types of cancers. In breast cancer, amplified cases are most commonly of the luminal B type. Amplified genes are not always over-expressed and there is a low correlation of amplification with over-expression in amplicon genes with variation between genes. The presence of the amplicon does not influence the aneuploidy score or the tumor mutation burden of breast cancers. Regarding prognosis, the two genes of the amplicon whose mRNA hyper-expression portends adverse relapse-free survival in breast cancer are EIF4EBP1 (Eukaryotic transcription initiation factor 4E binding protein 1) and LSM1 (LSM1 homolog, mRNA degradation associated).
    CONCLUSIONS: Besides the previously proposed genes to play a role as dominant oncogenes in the 8p11.23 cancer amplified locus, other genes may also be important in breast cancer based on the high correlation of their amplification and mRNA expression and adverse prognosis conferred by over-expression, consistent with an oncogenic role.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    有效的MYC癌蛋白在许多人类癌症中失调,包括乳腺癌,并与侵袭性疾病相关。为了了解MYC驱动的乳腺癌的机制和脆弱性,我们已经建立了一个体内模型,该模型模拟了响应MYC失调的人类疾病。在磷酸肌醇3激酶途径(PIK3CAH1047R)中异位表达常见乳腺癌突变的MCF10A细胞导致小鼠腺泡结构的发展。表达PIK3CAH1047R和解除调节的MYC导致浸润性导管癌的发展。因此,在这种情况下,MYC表达的失调产生了MYC依赖性的正常-肿瘤转换,其可以在体内测量。这些MYC驱动的肿瘤在病理和分子水平上都表现出人类乳腺癌的经典标志。此外,肿瘤生长依赖于持续的MYC表达失调,进一步证明了对这种有效的癌基因和基因转录调节因子的成瘾。因此,我们提供了一个依赖MYC的乳腺癌模型,可用于测定体内肿瘤信号通路,从正常乳腺腺泡向浸润性乳腺癌的增殖和转化。我们预计,这种新的MYC驱动的转化模型将是一个有用的研究工具,以更好地了解MYC的致癌功能和治疗漏洞的识别。
    The potent MYC oncoprotein is deregulated in many human cancers, including breast carcinoma, and is associated with aggressive disease. To understand the mechanisms and vulnerabilities of MYC-driven breast cancer, we have generated an in vivo model that mimics human disease in response to MYC deregulation. MCF10A cells ectopically expressing a common breast cancer mutation in the phosphoinositide 3 kinase pathway (PIK3CAH1047R) led to the development of organised acinar structures in mice. Expressing both PIK3CAH1047R and deregulated MYC led to the development of invasive ductal carcinoma. Therefore, the deregulation of MYC expression in this setting creates a MYC-dependent normal-to-tumour switch that can be measured in vivo These MYC-driven tumours exhibit classic hallmarks of human breast cancer at both the pathological and molecular level. Moreover, tumour growth is dependent upon sustained deregulated MYC expression, further demonstrating addiction to this potent oncogene and regulator of gene transcription. We therefore provide a MYC-dependent model of breast cancer, which can be used to assay in vivo tumour signalling pathways, proliferation and transformation from normal breast acini to invasive breast carcinoma. We anticipate that this novel MYC-driven transformation model will be a useful research tool to better understand the oncogenic function of MYC and for the identification of therapeutic vulnerabilities.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Radon gas is the leading cause of lung cancer in the nonsmoking population. The World Health Organization (WHO) recommends indoor concentrations of < 100 Bq/m³. Several molecular alterations have been described in non-small-cell lung cancer (NSCLC), mainly in nonsmokers, with no risk factors identified. We studied the role of indoor radon in NSCLC patients harboring specific driver alterations.
    We assessed the radon concentration from EGFR-, BRAF-mutated (m), and ALK-rearranged (r) NSCLC patients measured by an alpha-track detector placed in their homes between September 2014 and August 2015. Clinical characteristics were collected prospectively, and pathologic samples were reviewed retrospectively.
    Forty-eight patients were included (36 EGFRm, 10 ALKr, 2 BRAFm). Median radon concentration was 104 Bq/m³ (IQR 69-160) overall, and was 96 Bq/m³ (42-915) for EGFRm, 116 (64-852) for ALKr, and 125 for BRAFm, with no significant differences. Twenty-seven patients (56%) had indoor radon above WHO recommendations, 8 (80%) of 10 ALKr, 2 (100%) of 2 BRAFm, and 17 (47%) of 36 EGFRm.
    The median indoor radon concentration was above the WHO recommendations, with no differences between EGFR, ALK, and BRAF patients. Concentrations above the WHO recommendations were most common with ALKr and BRAFm. These findings should be validated in larger studies.
    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
    背景:对年轻非小细胞肺癌(NSCLC)患者的亚组了解甚少。我们回顾性研究了临床特征,基因突变,非小细胞肺癌患者(年龄≤40岁)的结局。
    结果:2001年2月至2016年10月诊断的7494例肺癌患者中,252例年龄≤40岁。我们根据组织学结果将他们的病例分为非鳞状细胞癌和鳞状细胞癌组。在252例年轻NSCLC患者中,173例(69%)患者有IIIB期或IV期,196人(78%)从未吸烟。最常见的四种转移是肺内病变,胸膜,骨头,和大脑。在腺癌患者中,29(40%,n=73)有表皮生长因子受体(EGFR)突变,25(34%,n=74)包含间变性淋巴瘤激酶(ALK)翻译,和1(14%,n=7)包含ROS原癌基因1受体酪氨酸激酶(ROS1)翻译。接受化疗的患者的中位无进展生存期(PFS)和总生存期(OS)分别为3.3和27.6个月(n=65)。接受EGFR酪氨酸激酶抑制剂(TKIs)的患者为12.1和33.6个月(n=13),分别。接受克唑替尼的患者的中位PFS时间为21.9个月(n=8)。
    结论:年轻患者与基因突变的可能性增加相关,当携带基因突变的患者接受EGFR-TKIs或ALK抑制剂治疗时,可以获得更好的预后。
    BACKGROUND: The subgroup of young patients with non-small-cell lung cancer (NSCLC) is poorly understood. We retrospectively studied the clinical characteristics, gene mutations, and outcomes of patients with NSCLC (aged ≤ 40 years).
    RESULTS: Of the 7494 patients with lung cancer diagnosed from February 2001 to October 2016, 252 aged ≤ 40 years showed NSCLC. We divided their cases into non-squamous cell carcinoma and squamous cell carcinoma groups according to their histology results. Of the 252 young NSCLC patients, 173 (69%) patients had stage IIIB or IV, and 196 (78%) had never smoked. The four most common metastases were intrapulmonary lesions, pleura, bone, and brain. Among patients with adenocarcinoma, 29 (40%, n = 73) harbored epidermal growth factor receptor (EGFR) mutations, 25 (34%, n = 74) harbored anaplastic lymphoma kinase (ALK) translations, and 1 (14%, n = 7) harbored ROS proto-oncogene 1 receptor tyrosine kinase (ROS1) translations. The median progression-free survival (PFS) and overall survival (OS) were 3.3 and 27.6 months for patients receiving chemotherapy (n = 65), and 12.1 and 33.6 months for patients receiving EGFR tyrosine kinase inhibitors (TKIs) (n = 13), respectively. Patients receiving crizotinib had a median PFS time of 21.9 months (n = 8).
    CONCLUSIONS: Young patients are associated with an increased likelihood of gene mutations and can receive a better prognosis when patients harboring gene mutations are treated with EGFR-TKIs or ALK inhibitors.
    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
    Nivolumab是一种免疫检查点抑制剂,具有针对几种恶性肿瘤的功效。EGFR和ALK等驱动癌基因的改变是nivolumab治疗非小细胞肺癌(NSCLC)的不良预后因素。而吸烟史是众所周知的,有利的预后因素。然而,nivolumab治疗多原发恶性肿瘤(MPMTs)的疗效尚未见报道,其对吸烟者驱动癌基因阳性NSCLC的疗效尚不清楚。在这里,我们报道了一例有大量吸烟史的患者,该患者发展为ALK阳性NSCLC和胃癌,对nivolumab治疗有疗效.一名76岁的重度吸烟者出现声音嘶哑和吞咽困难的症状到我们医院就诊。他最终被诊断为ALK阳性晚期NSCLC。使用ALK抑制剂(阿来替尼),肺癌病变有所改善。阿来替尼治疗持续5个月。此后,左肺下叶的病变表现为再生。在同一时期,患者出现上腹痛。胃镜检查提示胃癌。他接受了nivolumab治疗肺癌和胃癌。两个月后,肺部病变和胃部病变的大小均减小.Nivolumab疗法可能是重度吸烟者同步MPMT和NSCLC的有效疗法,即使肺癌有驱动癌基因突变。
    Nivolumab is an immune checkpoint inhibitor with demonstrated efficacy against several malignant tumors. Alterations in driver oncogenes such as EGFR and ALK are a poor prognostic factor in nivolumab therapy for non-small cell lung cancer (NSCLC), whereas a smoking history is a well-known, favorable prognostic factor. However, an efficacy of nivolumab therapy for multiple primary malignant tumors (MPMTs) has not been reported, and its efficacy for driver oncogene-positive NSCLC in smokers is unclear. Herein, we report the case of a patient with a history of heavy smoking who developed synchronous ALK-positive NSCLC and gastric cancer that responded to nivolumab therapy. A 76-year-old man who was a heavy smoker presented to our hospital with symptoms of hoarseness and dysphagia. He was ultimately diagnosed with ALK-positive advanced NSCLC. An ALK inhibitor (alectinib) was administered, and the lung cancer lesions showed improvement. The alectinib therapy was continued for 5 months. Thereafter, the lesions in the left lower lobe of the lung showed regrowth. During the same period, the patient experienced epigastric pain. Gastrointestinal endoscopy examination revealed gastric cancer. He was administered nivolumab to treat both the lung cancer and the gastric cancer. Two months later, both the lung lesions and the gastric lesions had diminished in size. Nivolumab therapy might be an effective therapy for synchronous MPMTs and NSCLC in heavy smokers, even if the lung cancer possesses driver oncogene mutations.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号