en
×

分享给微信好友或者朋友圈

使用微信“扫一扫”功能。
通讯作者:

施敏骅,E-mail:shiminhua@163.com

中图分类号:R734.2

文献标识码:A

文章编号:1007-4368(2022)09-1246-07

DOI:10.7655/NYDXBNS20220908

参考文献 1
SIEGEL R L,MILLER K D,FUCHS H E,et al.Cancer Statistics,2021[J].CA Cancer J Clin,2021,71(1):7-33
参考文献 2
MARTINS D,MENDES F,SCHMITT F.Microbiome:a supportive or a leading actor in lung cancer?[J].Pathobi⁃ ology,2021,88(2):198-207
参考文献 3
严晓娣,史国振,毛旭华.高通量测序技术检测非小细胞肺癌相关驱动基因的突变[J].南京医科大学学报(自然科学版),2021,41(2):193-197
参考文献 4
ZHENG S B,WANG X D,FU Y,et al.Targeted next⁃gen⁃ eration sequencing for cancer ⁃ associated gene mutation and copy number detection in 206 patients with non⁃small⁃ cell lung cancer[J].Bioengineered,2021,12(1):791-802
参考文献 5
TAN A C,LAI G G Y,TAN G S,et al.Utility of incorpo⁃ rating next⁃generation sequencing(NGS)in an Asian non⁃ small cell lung cancer(NSCLC)population:incremental yield of actionable alterations and cost⁃effectiveness analy⁃ sis[J].Lung Cancer Amsterdam Neth,2020,139:207-215
参考文献 6
TSUBATA Y,TANINO R,ISOBE T.Current therapeutic strategies and prospects for EGFR mutation⁃positive lung cancer based on the mechanisms underlying drug resis⁃ tance[J].Cell,2021,10(11):3192
参考文献 7
CHANG S C,LAI Y C,CHANG C Y,et al.Concomitant genetic alterations are associated with worse clinical out⁃ come in EGFR mutant NSCLC patients treated with tyro⁃ sine kinase inhibitors[J].Transl Oncol,2019,12(11):1425-1431
参考文献 8
WANG Z J,CHENG Y,AN T T,et al.Detection of EGFR mutations in plasma circulating tumour DNA as a selec⁃ tion criterion for first ⁃line gefitinib treatment in patients with advanced lung adenocarcinoma(BENEFIT):a phase 2,single⁃arm,multicentre clinical trial[J].Lancet Respir Med,2018,6(9):681-690
参考文献 9
ZHAO Z,WAN J,GUO M,et al.Expression and prognos⁃ tic significance of m6A⁃related genes in TP53⁃mutant non ⁃ small ⁃ cell lung cancer[J].J Clin Lab Anal,2022,36(1):e24118
参考文献 10
SAHIN I,SAAT H,AKSOY S,et al.Liquid biopsy:novel perspectives on the importance and spectrum of PIK3CA,PTEN and RET mutations in solid tumors[J].Mol Clin Oncol,2022,16(1):1
参考文献 11
SONG X M,CHEN Q,WANG J F,et al.Clinical and prog⁃ nostic implications of an immune ⁃ related risk model based on TP53 status in lung adenocarcinoma[J].J Cell Mol Med,2022,26(2):436-448
参考文献 12
LI W,LIU J,HOU L,et al.Liquid biopsy in lung cancer:significance in diagnostics,prediction,and treatment mon⁃ itoring[J].Mol Cancer,2022,21(1):25
参考文献 13
HEITZER E,VAN DEN BROEK D,DENIS M G,et al.Recommendations for a practical implementation of circu⁃ lating tumor DNA mutation testing in metastatic non⁃small ⁃cell lung cancer[J].ESMO Open,2022,7(2):100399
目录contents

    摘要

    目的:探索肺癌患者伴随基因突变/扩增频数的变化,并分析其临床意义。方法:回顾性分析苏州大学附属第二医院2015年12月—2020年7月经新一代基因测序(next-generation sequencing technology,NGS)检测的71例共132份肺癌患者标本基因检测数据(其中包括血液标本67份、肿瘤组织48份、胸腔积液15份、脑脊液2份),采用GraphPad Prism7.0统计分析不同临床标本伴随基因的突变/扩增状态。结果:所有检测标本除表皮生长因子受体(epidermal growth factor receptor,EGFR)突变之外,另可检测到前5位突变/扩增基因分别为肿瘤蛋白p53(tumor suppressor protein p53,TP53)、鼠类肉瘤病毒癌基因(Kirsten rat sarcoma viral oncogene,KRAS)、表皮生长因子受体2(human epithelial growth factor receptor 2,ERBB2)、视网膜母细胞瘤基因 1(retinoblastoma1,RB1)、肝细胞生长因子酪氨酸激酶受体(hepatocyte growth factor receptor,MET)。初诊肺癌患者血液标本共入组50例,EGFR突变阳性率34%(17/50),除EGFR突变之外,所检测标本还可检测到79种基因突变/扩增,所有标本共出现 123次伴随基因突变/扩增,平均2.46次/例,血液标本伴随基因突变/扩增频数与患者临床分期、吸烟、年龄、性别、病理类型均无关(均P >0.05)。初诊肺癌患者肿瘤组织共入组46例,EGFR突变阳性率50%(23/46),除EGFR突变之外,所检测标本还可检测到160种基因突变/扩增,所有标本共出现285次伴随基因突变/扩增,平均6.20次/例,肿瘤组织伴随基因突变/扩增频数与患者临床分期、年龄均无关(均 P >0.05),与吸烟、性别、病理类型均有关(U =74.000,P <0.001;U =130.5,P =0.003;F =8.968,P = 0.011)。亚组分析发现,血液标本及肿瘤组织中TP53是否突变对患者生存率影响差异无统计学意义(血液标本:χ2 =0.321,P = 0.571;肿瘤组织:χ2 =0.309,P =0.579)。对同时进行肿瘤组织和血液标本配对检测的患者,初诊肿瘤组织伴随基因突变/扩增频数[3(1,8)]高于同一患者的血液标本[3(0,1)],差异有统计学意义(W =-150,P =0.001)。动态监测抗肿瘤治疗前后的17例患者,抗肿瘤治疗前及抗肿瘤治疗后两组伴随基因突变/扩增频数相比[1(0,2)vs.1(0,3)],差异无统计学意义(W =-3,P = 0.916)。结论:肺癌患者抗肿瘤治疗前即存在多种伴随基因突变/扩增,有吸烟史、男性患者具有较高水平的伴随基因突变/扩增频数,伴随基因突变/扩增频数的改变不能提示患者病情进展,TP53突变与否对患者生存率无显著影响。

    Abstract

    Objective:This study aims to observe the changes of the concomitant gene mutation/amplification in patients with lung cancer based on next generation sequencing(NGS)technology and analyze its clinical significance. Methods:Retrospective analysis was performed on 71 lung cancer patients(132 samples)detected by NGS technology in the Second Affiliated Hospital of Soochow University from December 2015 to July 2020,including67 blood samples,48 tumor tissues,15 pleural effusion and 2 cerebrospinal fluid. GraphPad prism7.0 statistical software was used for statistical analysis of concomitant gene mutation/amplification in different clinical samples. Results:In addition to EGFR mutation,mutations/amplification of the first 5 genes were:tumor suppressor protein p53(TP53),Kirsten rat sarcoma viral oncogene(KRAS),human epithelial growth factor receptor 2(ERBB2),retinoblastoma1(RB1)and hepatocyte growth factor receptor(MET). A total of 50 newly diagnosed patients’blood samples were detected by NGS technology. The results showed that EGFR positive rate was 34%(17/50). Except EGFR mutation,there were79 kinds of gene mutations/ amplifications detected,with an average of 2.46 times/example,but without statistical difference in concomitant gene mutation/ amplification among clinical stage,smoking status,age,gender and pathological classification groups in blood samples. A total of 46 newly diagnosed patients’tissue samples were detected by NGS technology,of which EGFR positive rate was 50%(23/46). Except EGFR mutation,there were160 kinds of gene mutations/amplifications detected,with an average of 6.20 times/example. There was no statistical difference in concomitant gene mutation/amplification between clinical stage and age groups in tissue samples(P >0.05). But in terms of smoking status,gender and pathological classification groups,there were statistically differences(U =74.000,P <0.001; U =130.5,P =0.003;F =8.968,P =0.011). At the same time,TP53 mutation or not had no statistical significance on survival rate(the blood group:χ2 =0.321,P =0.571;the tissue group:χ2 =0.309,P =0.579). For the same patients,the frequency of gene mutation/ amplification in the tissue group([3(1,8)])was higher than that in the blood group[3(0,1)],and the differences were statistically significant(W =-150,P =0.001). In the17 patients with dynamic monitoring,there was no statistically significant difference in the frequency of concomitant gene mutation/amplification between the two groups before and after anti-tumor treatment(W =-3,P =0.916). Conclusion:There may be multiple parallel gene mutations/amplifications before the first - line treatment for lung cancer. Previous smoking and male patients are associated with higher levels of mutation/amplification status. Changes in the frequency of gene mutation/amplification cannot indicate disease progression,and TP53 mutation has no significant effect on the survival rate of patients.

    关键词

    肺肿瘤基因检测基因扩增突变

  • 2021年美国癌症协会数据报告,肺癌仍是目前人类死亡率最高(男性22%,女性22%)的肿瘤[1]。肺癌的恶性程度较高,病情进展较快,超过70%的患者在确诊时已出现局部或远隔器官的转移,失去了手术治疗的最佳时机[2]。表皮生长因子受体⁃酪氨酸激酶抑制剂(epidermal growth factor receptor ⁃ tyrosine kinase inhibitor,EGFR⁃TKI)已成为表皮生长因子受体(epidermal growth factor receptor,EGFR)基因敏感突变患者的一线标准治疗,建议初诊非小细胞肺癌患者完善新一代基因测序(next ⁃generation sequencing technology,NGS)检测[3]。NGS检测除了可为患者选择合适的靶向治疗外,还可以预测疗效,发现耐药机制,同时能发现新的基因突变类型,为靶向药物研发提供依据[4]。本研究回顾性分析经NGS检测的71例肺癌患者的基因检测数据,并进一步分析其临床意义。

  • 1 对象和方法

  • 1.1 对象

  • 纳入苏州大学附属第二医院2015年12月— 2020年7月入组的肺癌患者,依据气管镜、电子计算机断层扫描(computed tomography,CT)引导下经皮肺穿刺、胸腔积液病理结果明确诊断。患者或家属均已签署书面知情同意书,本研究已经苏州大学附属第二医院伦理委员会审批[(2013)伦审科研第 (K11)号]通过。

  • 纳入标准:①组织学或细胞学确诊的肺癌患者;②年龄≥18岁。排除标准:①同时有其他恶性肿瘤;②存在可能影响随访和短期生存的其他严重疾病;③既往服用过EGFR⁃TKI等靶向药物、免疫药物;④既往接受过化疗、放疗或其他治疗。共纳入71例患者(132份标本)。

  • 1.2 标本收集及患者随访

  • 1.2.1 检测标本

  • 肿瘤组织来源为气管镜下活检组织标本、肺穿刺活检组织标本,循环肿瘤DNA(circulating tumor deoxyribo nucleic acid,ctDNA)检测标本为外周血液、胸腔积液、脑脊液。

  • 1.2.2 标本收集

  • 血液、胸腔积液、脑脊液标本:收集患者用药前和耐药后全血(8mL)或胸腔积液(8mL)或脑脊液 (3mL),保存于streck管,并于管壁标记患者姓名及样品号,15~35℃条件下运输,72h内运送至实验室。 4℃条件下,1 590 g离心10min,收集上清至15mL离心管,4℃条件下16 000 g离心10min,再次收集上清至新的15mL离心管。封口膜封口后干冰运输。

  • 石蜡组织标本:收集患者肿瘤组织切片,要求恶性肿瘤细胞占比 ≥10%,坏死组织区域 ≤ 50%,标本编号标记规范清晰,石蜡组织标本切片厚度5 μm(防脱玻片),切片时摊片即可,无需烤片处理,取6周以内的石蜡切片,每组切片15张,常温或2~8℃运输。

  • 1.2.3 检测方法

  • 采用NGS检测技术(广州燃石医学检验所有限公司:Illumina公司检测平台,美国)捕获与肿瘤发生发展高度相关基因的全部外显子,包括腺瘤性结肠息肉(adenomatous polyposis coli,APC)、乳腺癌易感基因1(breast cancer susceptibility gene1, BRCA1)、细胞周期依赖性激酶抑制基因2A(cyclin⁃ dependent kinase inhibitor 2A gene,CDKN2A)等,及与肺癌个性化治疗相关或高频突变基因的重要外显子及部分内含子区域,包括间变性淋巴瘤激酶 (anaplastic lymphoma kinase,ALK)、鼠类肉瘤病毒癌基因同源物B1(v ⁃ raf murine sarcoma viral onco⁃ gene homolog B1,BRAF)、细胞周期蛋白E1(cyclin E1,CCNE1)等,进行1 000×高深度测序,以测量上述基因中出现的低频及超低频的来自肿瘤的DNA突变、重排、拷贝数增加等。

  • 1.2.4 生存随访

  • 通过门诊或电话随访,于2016年1月开始,每个月随访1次,直至患者死亡或研究结束。随访截至2020年11月30日,患者在随访截止时间仍未死亡,则按截尾数据处理。中位随访时间28.5个月。随访率为95%,3例失访。总生存期(overall survival,OS) 定义为患者治疗开始至患者死亡或末次随访的时间,中位OS定义为生存率为50%时所对应的生存时间。

  • 1.3 统计学方法

  • 采用GraphPad Prism7.0统计软件进行统计分析。定性资料采用例数和率描述;符合正态分布的定量资料,采用均数±标准差(x-±s)表示;非正态分布资料,采用中位数(四分位数)[MP25P75)]表示,两组间比较采用Mann⁃Whitney U检验,多组间比较采用Kruskal⁃Wallis检验,配对设计资料采用Wil⁃ coxon符号秩和检验。采用Kaplan⁃Meier法绘制生存曲线图,比较采用Log⁃rank检验,P< 0.05为差异有统计学意义。

  • 2 结果

  • 2.1 一般临床资料

  • 本研究共入组71例患者,共132份标本,检测标本类型包括血液(67份)、肿瘤组织(48份)、胸腔积液(15份)、脑脊液(2份)。71例患者均为初诊患者,其中男性32例、女性39例;年龄(65±11)岁,年龄范围36~85岁;其中Ⅰ期患者2例,Ⅱ期患者3例, Ⅲ期患者3例,Ⅳ期患者63例;吸烟患者23例;腺癌患者62例,鳞状细胞癌患者7例,小细胞肺癌患者2例。所有检测标本EGFR突变阳性率39.4%(52/132)。所有检测标本除EGFR突变之外,另外可检测到前5位基因突变/扩增分别是肿瘤蛋白p53 (tumor suppressor protein p53,TP53)、鼠类肉瘤病毒癌基因(Kirsten rat sarcoma viral oncogene,KRAS)、表皮生长因子受体2(human epithelial growth factor receptor 2,ERBB2)、视网膜母细胞瘤基因1(retino⁃ blastoma1,RB1)、肝细胞生长因子酪氨酸激酶受体 (hepatocyte growth factor receptor,MET)。

  • 2.2 初诊肺癌患者血液伴随基因状态

  • 共入组初诊肺癌患者血液标本50例,基于血浆ctDNA NGS检测结果显示,EGFR突变阳性率34%(17/50),除EGFR突变之外,所检测标本还可检测到79种基因突变/扩增,共出现123次伴随基因突变/扩增,人均2.46次/例(123次/50例),伴随基因突变/扩增频数≥2次的见图1,伴随基因突变/扩增频数为1次的共有60种基因,伴随基因突变/扩增(除EGFR外)与患者临床分期、吸烟、年龄、性别、病理类型均无关(均P >0.05,表1)。

  • 除EGFR突变之外,初诊肺癌患者血液标本中伴随基因TP53突变频数最高,因此针对此基因做进一步生存分析。入组Ⅳ期肺癌患者血液标本40例, TP53阳性患者中位OS为66个月,TP53阴性患者中位OS为47个月,两组生存曲线差异无统计学意义 (χ2=0.321,P=0.571,图2)。

  • 2.3 初诊肺癌患者肿瘤组织伴随基因状态

  • 共入组初诊肺癌患者肿瘤组织46例,EGFR突变阳性率50%(23/46),除EGFR突变之外,所检测标本还可检测到160种基因突变/扩增,所有标本共出现285次伴随基因突变/扩增,人均6.20次/例 (285次/46例),伴随基因突变/扩增频数≥2次的见图3,伴随基因突变/扩增频数为1次的共有107种,伴随基因突变/扩增(除EGFR外)与患者临床分期、年龄均无关(均 P >0.05),与吸烟状态、性别、病理类型均有关(均P <0.05,表2)。

  • 除EGFR突变之外,初诊肺癌患者肿瘤组织标本中伴随基因TP53出现突变频数最高,因此针对此基因做进一步生存分析,入组Ⅳ期肺癌患者肿瘤组织标本25例,TP53阳性患者中位OS未达到,TP53阴性患者中位OS为38.5个月,两组生存曲线间差异无统计学意义(χ2=0.309,P=0.579,图4)。

  • 2.4 同时进行肿瘤组织和血液标本配对检测的患者伴随基因状态

  • 共入组25例,初诊肿瘤组织中基因突变/扩增频数[3(1,8)]高于同一患者的血液组[3(0,1)],两者之间差异有统计学意义(W=-150,P=0.001);动态监测抗肿瘤治疗前后的17例患者,其中使用EGFR⁃ TKI治疗9例,间变性淋巴瘤激酶抑制剂(anaplastic lymphoma kinase inhibitor,ALK⁃TKI)治疗3例,化疗5例,抗肿瘤治疗前后两组伴随基因突变/扩增频数相比[1(0,2)vs.1(0,3)],差异无统计学意义(W=-3, P=0.916)。

  • 图1 50例初诊肺癌患者血液标本伴随基因突变/扩增频数分布图

  • Fig.1 Frequency distribution of gene mutation/amplification in blood samples from 50newly diagnosed lung cancer patients

  • 表1 50例初诊肺癌患者血液标本伴随基因突变/扩增频数与临床病理特征的关系

  • Table1 Correlation between frequency of associated gene mutation/amplification and clinicopatho⁃ logical features in blood samples of 50newly diagnosed lung cancer patients

  • 图2 肺癌患者血液标本伴随基因TP53阳性和阴性患者生存曲线的比较

  • Fig.2 Comparison of survival curves between patients with TP53positive and negative concomitant gene in blood samples of lung cancer patients

  • 3 讨论

  • 全面的分子生物学检测信息可为肺癌患者方案选择、预后判断以及临床试验入组提供依据。准确获取患者基因的突变状态,有助于筛选更加合适的治疗人群,精准地指导临床个体化治疗,为肿瘤诊断、治疗方案选择、预后监控等提供重要信息[5]。以EGFR为例,与标准化疗方案相比,EGFR敏感突变型晚期非小细胞肺癌患者使用小分子靶向药物EGFR⁃TKI治疗,极大地提高了该患者群的临床疗效(客观缓解率、无进展生存期、总生存时间)及生命质量[6]。但仍有20%~30%的EGFR敏感基因突变的患者出现原发性耐药或快速进展,可能是由于EGFR突变合并其他伴随基因突变,导致下游或旁路信号通路激活[7]。BENEFIT研究是一项单臂、多中心、前瞻性、大型Ⅱ期临床试验(CTONG1405; NCT02282267),2014年12月—2016年1月,共纳入中国15个中心的426例Ⅳ期肺腺癌患者,其中391例具有组织和血液样本,188例患者血液EGFR突变阳性并接受吉非替尼治疗(250mg/d),在180例具有基线NGS数据的EGFR突变患者中,仅存在EGFR敏感突变患者,无进展生存期可达13.2个月,EGFR敏感突变合并TP53突变患者,无进展生存期为9.3个月,伴随其他基因突变亚组患者,无进展生存期仅为5.5个月[8]

  • 图3 46例初诊肺癌患者肿瘤组织标本伴随基因突变/扩增频数分布图

  • Fig.3 Frequency distribution of gene mutation/amplification in tumor tissue samples from 46newly diagnosed lung can⁃ cer patients

  • 正常情况下TP53可以控制细胞循环周期、调节转录、DNA复制和诱导细胞程序性死亡及抗血管生成。TP53突变几乎在所有人类癌症中均有发生,是最常见的突变基因之一,在肺鳞状细胞癌患者中甚至高达80%[9-10]。TP53突变可激活T细胞介导的免疫反应,可能成为免疫治疗疗效的潜在预测因子[11]。本研究发现,伴随基因TP53突变与否与生存率无关,可能与本研究入组患者多为使用靶向药物治疗患者,且免疫治疗患者相对较少有关,后续有待进一步增大样本量进行统计学分析。在肺癌患者组织标本中行生存分析时,TP53阳性患者中位OS未达到,考虑可能与随访时间仍较短,且样本量不足有关,后续有待进一步增大样本量行统计学分析。

  • 本研究共入组初诊肺癌患者血液标本50例,除EGFR突变之外,所检测标本还可检测到79种基因突变/扩增,共出现123次伴随基因突变/扩增;本研究入组初诊肺癌患者肿瘤组织46例,除EGFR突变之外,所检测标本还可检测到160种基因突变/扩增,所有标本共出现285次伴随基因突变/扩增,同一患者肿瘤组织标本中伴随基因突变/扩增频数高于血液标本,原因可能是肿瘤患者体内ctDNA的含量低、半衰期短、需要高灵敏度的检测技术进行基因检测[12]。另外,需考虑肿瘤存在异质性,肿瘤组织检测反映肿瘤局部基因水平状态,基于ctDNA的基因检测更能反映患者的整体状态,并允许实时监测,可作为肿瘤组织基因检测的补充[13]

  • 表2 46例初诊肺癌患者肿瘤组织标本伴随基因突变/扩增频数与临床病理特征的关系

  • Table2 Correlation between frequency of associated gene mutation/amplification and clinicopatho⁃ logical features in tumor tissue samples of 46newly diagnosed lung cancer patients

  • 图4 肺癌患者肿瘤组织标本伴随基因TP53阳性和阴性患者生存曲线的比较

  • Fig.4 Comparison of survival curves between patients with TP53positive and negative concomitant gene in tumor tissue samples of lung cancer patients

  • 本研究针对抗肿瘤治疗前后的17例患者进行基因检测结果分析,发现抗肿瘤治疗前和抗肿瘤治疗后两组伴随基因突变/扩增频数相比,差异无统计学意义,原因可能为入组病例较少,且各组患者一线治疗方案有所差异,不同的治疗方案可能导致基因突变/扩增发生改变,后续将进一步扩大样本量,同时针对不同临床标本及不同治疗方案的患者分别进行统计分析。另外,本研究入组胸腔积液、脑脊液病例以及病理类型为鳞状细胞癌、小细胞癌病例均较少,后续应尽可能扩大样本量。本研究为单中心研究,期待有更多的随机对照研究进一步探索伴随基因异常对患者预后的影响。

  • 肺癌患者抗肿瘤治疗前即存在多种伴随基因突变/扩增,有吸烟史、男性患者具有较高水平的伴随基因突变/扩增频数,伴随基因突变/扩增频数的改变不能提示患者病情进展,随着靶向治疗和免疫治疗逐渐在肿瘤治疗中占据主导地位,基因测序技术具有极大的发展前景。肺癌是一个多基因参与的复杂病变过程,采用多标本、多基因的平行、动态检测有助于反映肺癌驱动基因全貌,更全面评估肿瘤的耐药机制,对临床具有指导意义。

  • 参考文献

    • [1] SIEGEL R L,MILLER K D,FUCHS H E,et al.Cancer Statistics,2021[J].CA Cancer J Clin,2021,71(1):7-33

    • [2] MARTINS D,MENDES F,SCHMITT F.Microbiome:a supportive or a leading actor in lung cancer?[J].Pathobi⁃ ology,2021,88(2):198-207

    • [3] 严晓娣,史国振,毛旭华.高通量测序技术检测非小细胞肺癌相关驱动基因的突变[J].南京医科大学学报(自然科学版),2021,41(2):193-197

    • [4] ZHENG S B,WANG X D,FU Y,et al.Targeted next⁃gen⁃ eration sequencing for cancer ⁃ associated gene mutation and copy number detection in 206 patients with non⁃small⁃ cell lung cancer[J].Bioengineered,2021,12(1):791-802

    • [5] TAN A C,LAI G G Y,TAN G S,et al.Utility of incorpo⁃ rating next⁃generation sequencing(NGS)in an Asian non⁃ small cell lung cancer(NSCLC)population:incremental yield of actionable alterations and cost⁃effectiveness analy⁃ sis[J].Lung Cancer Amsterdam Neth,2020,139:207-215

    • [6] TSUBATA Y,TANINO R,ISOBE T.Current therapeutic strategies and prospects for EGFR mutation⁃positive lung cancer based on the mechanisms underlying drug resis⁃ tance[J].Cell,2021,10(11):3192

    • [7] CHANG S C,LAI Y C,CHANG C Y,et al.Concomitant genetic alterations are associated with worse clinical out⁃ come in EGFR mutant NSCLC patients treated with tyro⁃ sine kinase inhibitors[J].Transl Oncol,2019,12(11):1425-1431

    • [8] WANG Z J,CHENG Y,AN T T,et al.Detection of EGFR mutations in plasma circulating tumour DNA as a selec⁃ tion criterion for first ⁃line gefitinib treatment in patients with advanced lung adenocarcinoma(BENEFIT):a phase 2,single⁃arm,multicentre clinical trial[J].Lancet Respir Med,2018,6(9):681-690

    • [9] ZHAO Z,WAN J,GUO M,et al.Expression and prognos⁃ tic significance of m6A⁃related genes in TP53⁃mutant non ⁃ small ⁃ cell lung cancer[J].J Clin Lab Anal,2022,36(1):e24118

    • [10] SAHIN I,SAAT H,AKSOY S,et al.Liquid biopsy:novel perspectives on the importance and spectrum of PIK3CA,PTEN and RET mutations in solid tumors[J].Mol Clin Oncol,2022,16(1):1

    • [11] SONG X M,CHEN Q,WANG J F,et al.Clinical and prog⁃ nostic implications of an immune ⁃ related risk model based on TP53 status in lung adenocarcinoma[J].J Cell Mol Med,2022,26(2):436-448

    • [12] LI W,LIU J,HOU L,et al.Liquid biopsy in lung cancer:significance in diagnostics,prediction,and treatment mon⁃ itoring[J].Mol Cancer,2022,21(1):25

    • [13] HEITZER E,VAN DEN BROEK D,DENIS M G,et al.Recommendations for a practical implementation of circu⁃ lating tumor DNA mutation testing in metastatic non⁃small ⁃cell lung cancer[J].ESMO Open,2022,7(2):100399

  • 参考文献

    • [1] SIEGEL R L,MILLER K D,FUCHS H E,et al.Cancer Statistics,2021[J].CA Cancer J Clin,2021,71(1):7-33

    • [2] MARTINS D,MENDES F,SCHMITT F.Microbiome:a supportive or a leading actor in lung cancer?[J].Pathobi⁃ ology,2021,88(2):198-207

    • [3] 严晓娣,史国振,毛旭华.高通量测序技术检测非小细胞肺癌相关驱动基因的突变[J].南京医科大学学报(自然科学版),2021,41(2):193-197

    • [4] ZHENG S B,WANG X D,FU Y,et al.Targeted next⁃gen⁃ eration sequencing for cancer ⁃ associated gene mutation and copy number detection in 206 patients with non⁃small⁃ cell lung cancer[J].Bioengineered,2021,12(1):791-802

    • [5] TAN A C,LAI G G Y,TAN G S,et al.Utility of incorpo⁃ rating next⁃generation sequencing(NGS)in an Asian non⁃ small cell lung cancer(NSCLC)population:incremental yield of actionable alterations and cost⁃effectiveness analy⁃ sis[J].Lung Cancer Amsterdam Neth,2020,139:207-215

    • [6] TSUBATA Y,TANINO R,ISOBE T.Current therapeutic strategies and prospects for EGFR mutation⁃positive lung cancer based on the mechanisms underlying drug resis⁃ tance[J].Cell,2021,10(11):3192

    • [7] CHANG S C,LAI Y C,CHANG C Y,et al.Concomitant genetic alterations are associated with worse clinical out⁃ come in EGFR mutant NSCLC patients treated with tyro⁃ sine kinase inhibitors[J].Transl Oncol,2019,12(11):1425-1431

    • [8] WANG Z J,CHENG Y,AN T T,et al.Detection of EGFR mutations in plasma circulating tumour DNA as a selec⁃ tion criterion for first ⁃line gefitinib treatment in patients with advanced lung adenocarcinoma(BENEFIT):a phase 2,single⁃arm,multicentre clinical trial[J].Lancet Respir Med,2018,6(9):681-690

    • [9] ZHAO Z,WAN J,GUO M,et al.Expression and prognos⁃ tic significance of m6A⁃related genes in TP53⁃mutant non ⁃ small ⁃ cell lung cancer[J].J Clin Lab Anal,2022,36(1):e24118

    • [10] SAHIN I,SAAT H,AKSOY S,et al.Liquid biopsy:novel perspectives on the importance and spectrum of PIK3CA,PTEN and RET mutations in solid tumors[J].Mol Clin Oncol,2022,16(1):1

    • [11] SONG X M,CHEN Q,WANG J F,et al.Clinical and prog⁃ nostic implications of an immune ⁃ related risk model based on TP53 status in lung adenocarcinoma[J].J Cell Mol Med,2022,26(2):436-448

    • [12] LI W,LIU J,HOU L,et al.Liquid biopsy in lung cancer:significance in diagnostics,prediction,and treatment mon⁃ itoring[J].Mol Cancer,2022,21(1):25

    • [13] HEITZER E,VAN DEN BROEK D,DENIS M G,et al.Recommendations for a practical implementation of circu⁃ lating tumor DNA mutation testing in metastatic non⁃small ⁃cell lung cancer[J].ESMO Open,2022,7(2):100399