Perspective of Precision Therapy on Lung Cancer
-
摘要:
目前,肺癌仍然是我国发病率和死亡率较高的恶性肿瘤。传统放化疗疗效有限,肺癌治疗新药物和新方案的探索一直在进行中。过去20多年,随着分子靶向和免疫治疗的飞速发展,临床疗效不断刷新,越来越多的癌症患者从中获益。肺癌治疗方法日新月异,是实体肿瘤精准治疗领域的“排头兵”。本文梳理了肺癌精准靶向和免疫治疗的发展历程,并对存在的问题及未来发展方向进行探讨。
Abstract:Lung cancer remains to have the highest morbidity and mortality rates in China among known malignant tumors. Novel drugs and regimens have been sought because of the limited efficiency of traditional chemotherapy and radiotherapy in lung cancer treatment. In the last 20 years, rapid developments in molecular targeted therapy and immunotherapy have increased clinical efficacy and benefitted patients with cancer. Treatments for lung cancer are the most rapidly developed among treatments for solid tumors, pioneering tumor precision medicine. This manuscript reviews the evolution and development of targeted therapy and immunotherapy and discusses existing problems and future directions in the precision therapy of lung cancer.
-
Key words:
- Lung cancer /
- Precision medicine /
- Molecular targeted therapy /
- Immunotherapy
-
0 引言
目前,乳腺癌严重威胁着女性的健康。在乳腺癌的转移机制中,上皮间质转化(epithelial-mesenchymal transition, EMT)是其中一个核心的生物学进程,与乳腺癌的侵袭和转移密切相关。JMJD3又称赖氨酸去甲基化酶6B(lysine demethylase 6B, KDM6B)作为一种Lys27位点三甲基化组蛋白H3(histone H3 methylated Lys27,H3K27me3)去甲基化酶[1],其异常表达与多种肿瘤的发生、发展密切相关[2-3]。JMJD3在肿瘤中是促癌基因还是抑癌基因研究报道尚不一致,有关JMJD3在乳腺癌中的作用,以及JMJD3与上皮间质转化的相关性尚未见类似报道。本研究收集120例乳腺浸润性导管癌组织标本,采用免疫组织化学法检测JMJD3和上皮间质转化相关蛋白E-cadherin、Vimentin和N-cadherin的表达,并分析其相关性及临床病理意义,以拓展和丰富人们对乳腺癌发病机制的认识。
1 资料与方法
1.1 病例资料
收集郑州人民医院2014年1月—2015年12月间收治的120例乳腺原发浸润性导管癌及相应的癌旁病理活检标本和相关临床资料。患者术前均未接受放疗或化疗。按照2012年WHO乳腺癌组织病理分级分类法对其进行分组。所有标本均经10%的甲醛固定,石蜡包埋,并经郑州人民医院病理科确诊。
1.2 主要试剂
JMJD3一抗兔抗人多克隆抗体购自英国Abcam公司,E-cadherin、Vimentin和N-cadherin一抗兔抗人单克隆抗体(即用型)、二抗及二氨基联苯胺(DAB)均为福州迈新公司产品。
1.3 免疫组织化学染色
主要步骤如下:(1)石蜡组织切片、烤片、脱蜡并水化;(2)EDTA抗原修复;(3)兔抗人单克隆抗体一抗37℃孵育2 h;(4)二抗37℃孵育20~30 min;(5)DAB显色5~10 min;(6)脱水、透明和封片。用已知JMJD3、E-cadherin、Vimentin和N-cadherin阳性的乳腺癌组织和PBS分别代替一抗作为阳性和空白对照。
1.4 免疫组织化学结果判断
JMJD3阳性信号呈棕褐色,表达于细胞核;E-cadherin和N-cadherin阳性信号为棕黄色,表达于细胞膜;Vimentin阳性信号为棕黄色,表达于细胞质。按样本中JMJD3、E-cadherin、Vimentin及N-cadherin阳性细胞占总细胞的比例分为4个等级[4]: < 5%为阴性、5%~ < 25%为弱阳性、25%~50%为中等阳性、≥50%为强阳性。其中阴性和弱阳性判断为低表达,中等阳性和强阳性判断为高表达,阳性表达率=(中等阳性+强阳性样本量)/总样本量,免疫组织化学的判读由两位高年资病理医师采用双盲法进行。
1.5 统计学方法
采用SPSS17.0统计软件进行数据处理与统计分析,JMJD3、E-cadherin、Vimentin和N-cadherin阳性表达率和病理因素的相关性采用χ2检验,蛋白间的相关性采用Spearman相关分析,以P < 0.05为差异有统计学意义。
2 结果
2.1 乳腺浸润性导管癌中JMJD3、E-cadherin、Vimentin和N-cadherin的表达
JMJD3主要定位于细胞核,在乳腺浸润性导管癌及癌旁组织中呈棕褐色,在导管癌中的阳性表达率明显低于癌旁组织,差异有统计学意义(χ2=7.326, P=0.007),见图 1、表 1。JMJD3表达较高者,组织学分化程度高,细胞黏附性强,极性较好,JMJD3表达较低者,组织学分化程度差,细胞黏附性下降,失去极性,细胞梭形更为明显,见图 2。
表 1 乳腺癌和癌旁组织中JMJD3、E-cadherin、Vimentin及N-cadherin的阳性表达(n(%))Table 1 Expression of JMJD3, E-cadherin, Vimentin and N-cadherin in breast cancer tissues and adjacent tissues (n(%))E-cadherin在乳腺浸润性导管癌及癌旁组织中主要定位于细胞膜,为粗细相对均匀的棕褐色颗粒,乳腺浸润性导管癌中E-cadherin阳性表达率明显低于癌旁组织,差异有统计学意义(χ2=10.256, P=0.001)。Vimentin主要表达于上皮周围的间质细胞中,在癌及癌旁组织中主要定位于细胞质,为粗细不等的棕褐色颗粒,乳腺浸润性导管癌中Vimentin阳性表达率明显高于癌旁组织,差异有统计学意义(χ2=24.033, P=0.000)。N-cadherin在癌及癌旁组织中主要定位于细胞膜,为粗细相对均匀的棕褐色颗粒,乳腺浸润性导管癌中N-cadherin阳性表达率为明显高于癌旁组织,差异有统计学意义(χ2=35.281, P=0.000),见图 1、表 1。
2.2 JMJD3、E-cadherin、Vimentin和N-cadherin与乳腺浸润性导管癌临床病理特征的关系
在乳腺浸润性导管癌中,JMJD3与肿瘤直径(χ2=9.697, P=0.008)、组织学分级(χ2=27.313, P=0.000)、淋巴结转移(χ2=12.587, P=0.000)TNM分期有关(χ2=12.385, P=0.000),与乳腺癌患者年龄和肿瘤位置均无关(均P > 0.05)。E-cadherin在高分化、中分化和低分化乳腺癌中表达率依次降低,差异具有统计学意义(χ2=14.152, P=0.001);其在淋巴结有转移组中低于无淋巴结转移组,差异有统计学意义(χ2=9.903, P=0.002);其在TNM Ⅲ期、Ⅳ期组别低于Ⅰ期、Ⅱ期组别,差异具有统计学意义(χ2=20.771, P=0.000)。E-cadherin与乳腺癌患者年龄、肿瘤位置和肿瘤直径均无关(均P > 0.05)。Vimentin在高分化、中分化和低分化乳腺癌中表达率依次升高,差异具有统计学意义(χ2=28.410, P=0.000);其在淋巴结有转移组中高于无淋巴结转移组(χ2=14.090, P=0.000),TNMⅢ期、Ⅳ期组高于Ⅰ期、Ⅱ期组(χ2=13.218, P=0.000),与年龄、肿瘤位置和肿瘤直径均无关(均P > 0.05)。N-cadherin在高分化、中分化和低分化中表达率依次升高(χ2=28.410, P=0.000);其在淋巴结有转移组中高于无淋巴结转移组(χ2=17.678, P=0.000),在TNM Ⅲ期、Ⅳ期组高于Ⅰ期、Ⅱ期组(χ2=30.035, P=0.000)。N-cadherin与乳腺癌患者年龄、肿瘤位置及肿瘤直径均无关(均P > 0.05),见表 2。
表 2 乳腺癌中JMJD3、E-cadherin、Vimentin及N-cadherin的表达与临床病理特征的关系(n(%))Table 2 Correlation of JMJD3, E-cadherin, Vimentin and N-cadherin expression with clinicopathological characteristics of breast cancer patients (n(%))2.3 乳腺浸润性导管癌中JMJD3与E-cadherin、Vimentin和N-cadherin相关性分析
采用Spearman相关分析,结果显示JMJD3和E-cadherin表达呈正相关(r=0.204, P=0.025)。JMJD3和Vimentin表达呈负相关(r=-0.467, P=0.000)。JMJD3和N-cadherin表达呈负相关(r=-0.500, P=0.000),见表 3。
表 3 乳腺癌中JMJD3与E-cadherin, Vimentin和N-cadherin的Spearman相关性分析Table 3 Spearman correlation analysis of JMJD3 expression with E-cadherin, Vimentin and N-cadherin expression in breast cancer tissues3 讨论
JMJD3编码基因位于人类第17号染色体上,其异常表达与肿瘤的发生发展有密切关系。Tokunaga等[5]对151例结肠癌患者的术后生存分析结果表明,JMJD3低表达者术后生存期普遍较短,且回归分析表明JMJD3的低表达是患者不良预后的独立危险因素之一。国内有研究发现JMJD3过表达对胃癌细胞的增殖具有抑制作用,对细胞凋亡具有促进作用[6]。胡贺芳等[7]研究发现JMJD3表达降低可能促进肺癌的发生发展。李彦伟等[8]发现胶质瘤细胞中JMJD3较瘤旁组织表达降低,其过表达可增强胶质瘤细胞迁移能力,该机制通过影响SNAIL基因调控区H3K27me3水平而调节胶质瘤侵袭过程[9]。
JMJD3是促癌基因还是抑癌基因研究报道尚不一致,本研究采用免疫组织化学方法发现JMJD3在乳腺浸润性导管癌组织表达率较癌旁组织低,其在组织学分化差、有淋巴结转移及较高TNM分期的组织中表达率减低,提示JMJD3在乳腺癌的浸润转移中扮演了抑癌基因的角色。此外本研究结果还显示JMJD3低表达与肿瘤直径相关,肿瘤直径的大小反映细胞增殖能力,我们推测JMJD3对乳腺癌细胞增殖具有抑制作用,这与以往其在胃癌研究报道观点一致[6]。
在乳腺癌的转移机制中,EMT是其中一个核心的生物学进程。EMT的过程中,上皮细胞标志性蛋白E-cadherin减少,而间质细胞标志性蛋白Vimentin和N-cadherin表达量上升[10-12]。E-cadherin主要分布于胚胎组织和成熟组织的上皮细胞中,除具有调节胚胎组织的发育、组织形成、参与细胞与细胞间信息传递交流等作用外,对促进细胞的黏附聚集、维持上皮形态结构完整性、极性和参与分化调节也至关重要。Vimentin主要表达于间叶组织和淋巴造血细胞,在上皮中几乎不表达,当细胞出现上皮间质转化时可有表达。N-cadherin表达于神经外胚层和中胚层来源的组织,正常上皮组织不表达或低表达。N-cadherin在上皮组织中的异位表达可以影响上皮细胞的形态和行为,诱导其向间质转化,即产生EMT。
本研究结果显示从乳腺癌旁组织到乳腺癌组织JMJD3和E-cadherin阳性率逐渐减少,而Vimentin和N-cadherin阳性率逐渐增高;同时它们均与乳腺癌分化程度、TNM分期和淋巴结转移相关,表明JMJD3和EMT相关蛋白共同参与乳腺癌的发生、发展,其可能机制是通过降低肿瘤细胞间的黏附力,使乳腺癌更容易发生浸润和转移。本研究中E-cadherin、Vimentin和N-cadherin的表达情况与以往文献报道的结果基本一致[13-14]。
通过查阅国内外文献,我们发现JMJD3表达对上皮间质转化的影响在胶质瘤细胞[15]、肝癌细胞[16-17]、肾癌细胞[18]和弥漫大B细胞淋巴瘤[19]已有研究报道,其在乳腺癌中尚未见研究报道。上述研究均表明JMJD3过表达能够促进上皮间质转化过程进而促进肿瘤的浸润转移。本研究相关分析结果发现JMJD3与E-cadherin表达呈正相关,与Vimentin和N-cadherin表达呈负相关,表明JMJD3低表达可能通过下调E-cadherin及上调Vimentin和N-cadherin的表达使乳腺癌上皮细胞获得间质细胞的特性,进而促进EMT的发生。提示JMJD3阳性率较低的乳腺癌患者更易发生淋巴结转移和较差的组织学分化,而组织学分化较差的乳腺癌细胞在形态学上细胞极性消失,细胞梭形明显,黏附性下降,从形态学上表明JMJD3可能通过上皮间质转化途径参与乳腺癌的发生发展。本研究从蛋白水平对乳腺癌中JMJD3、E-cadherin和Vimentin表达进行分析,尚缺少分子生物学调节实验,还需进一步深入研究。
综上所述,乳腺浸润性导管癌中组蛋白去甲基化酶JMJD3与E-cadherin呈正相关,与Vimentin和N-cadherin呈负相关,JMJD3可能通过调节乳腺癌细胞上皮间质转化,调控乳腺癌侵袭转移,其具体机制有待后续深入研究。
Competing interests: The authors declare that they have no competing interests.利益冲突声明:所有作者均声明不存在利益冲突。作者贡献:任胜祥:总体策划、论文构思、文稿撰写、修改与审校邱天羽:文献收集、论文构思、文稿撰写 -
[1] Zheng R, Zhang S, Zeng H, et al. Cancer incidence and mortality in China, 2016[J]. J Natl Cancer Cent, 2022, 2(1): 1-9. doi: 10.1016/j.jncc.2022.02.002
[2] Yuan M, Huang L, Chen J, et al. The emerging treatment landscape of targeted therapy in non-small-cell lung cancer[J]. Signal Transduct Target Ther, 2019, 4: 61. doi: 10.1038/s41392-019-0099-9
[3] Papadimitrakopoulou VA, Mok TS, Han JY, et al. Osimertinib versus platinum-pemetrexed for patients with EGFR T790M advanced NSCLC and progression on a prior EGFR-tyrosine kinase inhibitor: AURA3 overall survival analysis[J]. Ann Oncol, 2020, 31(11): 1536-1544. doi: 10.1016/j.annonc.2020.08.2100
[4] Lu S, Wang QM, Zhang GJ, et al. Efficacy of Aumolertinib (HS-10296) in Patients With Advanced EGFR T790M+ NSCLC: Updated Post-National Medical Products Administration Approval Results From the APOLLO Registrational Trial[J]. J Thorac Oncol, 2022, 17(3): 411-422. doi: 10.1016/j.jtho.2021.10.024
[5] Shi YK, Hu XS, Zhang SC, et al. Efficacy, safety, and genetic analysis of furmonertinib (AST2818) in patients with EGFR T790M mutated non-small-cell lung cancer: a phase 2b, multicentre[J]. Lancet Respir Med, 2021, 9(8): 829-839. doi: 10.1016/S2213-2600(20)30455-0
[6] Soria JC, Ohe Y, Vansteenkiste J, et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer[J]. N Engl J Med, 2018, 378(2): 113-125. doi: 10.1056/NEJMoa1713137
[7] Ramalingam SS, Gray JE, Ohe Y, et al. Osimertinib vs comparator EGFR-TKI as first-line treatment for EGFRm advanced NSCLC (FLAURA): Final overall survival analysis[J]. Ann Oncol, 2019, 30(Suppl_5): V851-V934. http://www.sciencedirect.com/science/article/pii/s0923753419604369
[8] Cho BC, Chewaskulyong B, Lee KH, et al. Osimertinib versus Standard of Care EGFR TKI as First-Line Treatment in Patients with EGFRm Advanced NSCLC: FLAURA Asian Subset[J]. J Thorac Oncol, 2019, 14(1): 99-106. doi: 10.1016/j.jtho.2018.09.004
[9] Shi Y, Chen G, Wang X, et al. Furmonertinib (AST2818) versus gefitinib as first-line therapy for Chinese patients with locally advanced or metastatic EGFR mutation-positive non-small-cell lung cancer (FURLONG): a multicentre, double-blind, randomised phase 3 study[J]. Lancet Respir Med, 2022, 10(11): 1019-1028. doi: 10.1016/S2213-2600(22)00168-0
[10] Lu S, Dong X, Jian H, et al. AENEAS: A Randomized Phase ⅢTrial of Aumolertinib Versus Gefitinib as First-Line Therapy for Locally Advanced or Metastatic Non-Small-Cell Lung Cancer With EGFR Exon 19 Deletion or L858R Mutations[J]. J Clin Oncol, 2022, 40(27): 3162-3171. doi: 10.1200/JCO.21.02641
[11] Park K, Haura EB, Leighl NB, et al. Amivantamab in EGFR Exon 20 Insertion-Mutated Non-Small-Cell Lung Cancer Progressing on Platinum Chemotherapy: Initial Results From the CHRYSALIS PhaseⅠ Study[J]. J Clin Oncol, 2021, 39(30): 3391-3402. doi: 10.1200/JCO.21.00662
[12] Han H, Li S, Chen T, et al. Targeting HER2 Exon 20 Insertion-Mutant Lung Adenocarcinoma with a Novel Tyrosine Kinase Inhibitor Mobocertinib[J]. Cancer Res, 2021, 81(20): 5311-5324. doi: 10.1158/0008-5472.CAN-21-1526
[13] Zhong WZ, Wang Q, Mao WM, et al. Gefitinib Versus Vinorelbine Plus Cisplatin as Adjuvant Treatment for Stage Ⅱ-ⅢA (N1-N2) EGFR-Mutant NSCLC: Final Overall Survival Analysis of CTONG1104 PhaseⅢ Trial[J]. J Clin Oncol, 2021, 39(7): 713-722. doi: 10.1200/JCO.20.01820
[14] Yue DS, Xu SD, Wang Q, et al. Erlotinib versus vinorelbine plus cisplatin as adjuvant therapy in Chinese patients with stage ⅢA EGFR mutation-positive non-small-cell lung cancer (EVAN): a randomised, open-label, phase 2 trial[J]. Lancet Respir Med, 2018, 6(11): 863-873. doi: 10.1016/S2213-2600(18)30277-7
[15] Zhou C, He J, Su C, et al. Icotinib versus Chemotherapy as Adjuvant Treatment for Stage Ⅱ-ⅢA EGFR-Mutant NSCLC (EVIDENCE): A Randomized, Open-Label, Phase 3 Study[J]. J Thorac Oncol, 2021, 16(3): S232-S232.
[16] Wu Y, John T, Grohe C, et al. Postoperative Chemotherapy Use and Outcomes from ADAURA: Osimertinib as Adjuvant Therapy for Resected EGFR-Mutated NSCLC[J]. J Thorac Oncol, 2022, 17(3): 423-433. doi: 10.1016/j.jtho.2021.10.014
[17] Zhong WZ, Wu YL, Chen KN, et al. LBA48_PRCTONG 1103: Erlotinib versus gemcitabine plus cisplatin as neo-adjuvant treatment for stage ⅢA-N2 EGFR-mutation non-small cell lung cancer (EMERGING): A randomised study[J]. Ann Oncol, 2018, 29(suppl_8): 738-738. http://www.sciencedirect.com/science/article/pii/S0923753419504495
[18] Lyu C, Fang W, Jiao W, et al. Osimertinib as neoadjuvant therapy in patients with EGFR mutated resectable stage Ⅱ-ⅢB lung adenocarcinoma (NEOS): Updated results[J]. Ann Oncol, 2022, 33(suppl 2): S71-S72.
[19] Zhou CC, Kim SW, Reungwetwattana T, et al. Alectinib versus crizotinib in untreated Asian patients with anaplastic lymphoma kinase-positive non-small-cell lung cancer (ALESIA): a randomised phase 3 study[J]. Lancet Respir Med, 2019, 7(5): 437-446. doi: 10.1016/S2213-2600(19)30053-0
[20] Mok T, Kim DW, Wu YL, et al. First-line crizotinib versus pemetrexed-cisplatin or pemetrexed-carboplatin in patients (pts) with advanced ALK-positive non-squamous non-small cell lung cancer (NSCLC): results of a phaseⅢ study (PROFILE 1014)[J]. J Clin Oncol, 2014, 32(15_Suppl): 8002. doi: 10.1200/jco.2014.32.15_suppl.8002
[21] Mok T, Camidge DR, Gadgeel SM, et al. Updated overall survival and final progression-free survival data for patients with treatment-naive advanced ALK-positive non-small-cell lung cancer in the ALEX study[J]. Ann Oncol, 2020, 31(8): 1056-1064. doi: 10.1016/j.annonc.2020.04.478
[22] Cai DJ, Hu CP, Li L, et al. The prevalence and prognostic value of KRAS co-mutation subtypes in Chinese advanced non-small cell lung cancer patients[J]. Cancer Med, 2020, 9(1): 84-93. doi: 10.1002/cam4.2682
[23] Tan DS, Geater S, Yu CJ, et al. First-line ceritinib versus chemotherapy in patients (pts) with advanced ALK rearranged (ALK+) non-small cell lung cancer (NSCLC): ASCEND-4 Asian subgroup analysis[J]. Ann Oncol, 2019, 30(suppl 5): 599.
[24] Camidge DR, Kim HR, Ahn MJ, et al. Brigatinib Versus Crizotinib in Advanced ALK Inhibitor-Naive ALK-Positive Non-Small Cell Lung Cancer: Second Interim Analysis of the PhaseⅢ ALTA-1L Trial[J]. J Clin Oncol, 2020, 38(31): 3592-3603. doi: 10.1200/JCO.20.00505
[25] Horn L, Wang ZP, Wu G, et al. Ensartinib vs. Crizotinib for Patients With Anaplastic Lymphoma Kinase-Positive Non-Small Cell Lung Cancer A Randomized Clinical Trial[J]. JAMA Oncol, 2021, 7(11): 1617-1625. doi: 10.1001/jamaoncol.2021.3523
[26] Shaw AT, Bauer TM, de Marinis F, et al. First-Line Lorlatinib or Crizotinib in Advanced ALK-Positive Lung Cancer[J]. N Engl J Med, 2020, 383(21): 2018-2029. doi: 10.1056/NEJMoa2027187
[27] Bergethon K, Shaw AT, Ou SHI, et al. ROS1 Rearrangements Define a Unique Molecular Class of Lung Cancers[J]. J Clin Oncol, 2012, 30(8): 863-870. doi: 10.1200/JCO.2011.35.6345
[28] Wu YL, Yang JCH, Kim DW, et al. PhaseⅡ Study of Crizotinib in East Asian Patients With ROS1-Positive Advanced Non-Small-Cell Lung Cancer[J]. J Clin Oncol, 2018, 36(14): 1405-1411. doi: 10.1200/JCO.2017.75.5587
[29] D'Angelo A, Sobhani N, Chapman R, et al. Focus on ROS1-Positive Non-Small Cell Lung Cancer (NSCLC): Crizotinib, Resistance Mechanisms and the Newer Generation of Targeted Therapies[J]. Cancers (Basel), 2020, 12(11): 3293. doi: 10.3390/cancers12113293
[30] Shaw AT, Solomon BJ, Chiari R, et al. Lorlatinib in advanced ROS1-positive non-small-cell lung cancer: a multicentre, open-label, single-arm, phase 1-2 trial[J]. Lancet Oncol, 2019, 20(12): 1691-1701. doi: 10.1016/S1470-2045(19)30655-2
[31] Jaenne PA, Riely GJ, Gadgeel SM, et al. Adagrasib in Non-Small-Cell Lung Cancer Harboring a KRASG12C Mutation[J]. N Engl J Med, 2022, 387(2): 120-131. doi: 10.1056/NEJMoa2204619
[32] Mazieres J, Peters S, Lepage B, et al. Lung Cancer That Harbors an HER2 Mutation: Epidemiologic Characteristics and Therapeutic Perspectives[J]. J Clin Oncol, 2013, 31(16): 1997-2003. doi: 10.1200/JCO.2012.45.6095
[33] Song ZB, Lv DQ, Chen SQ, et al. Pyrotinib in Patients with HER2-Amplified Advanced Non-Small Cell Lung Cancer: A Prospective, Multicenter, Single-Arm Trial[J]. Clin Cancer Res, 2022, 28(3): 461-467. doi: 10.1158/1078-0432.CCR-21-2936
[34] Elamin YY, Robichaux JP, Carter BW, et al. Poziotinib for Patients With HER2 Exon 20 Mutant Non-Small-Cell Lung Cancer: Results From a PhaseⅡ Trial[J]. J Clin Oncol, 2022, 40(7): 702-709. doi: 10.1200/JCO.21.01113
[35] Iwama E, Zenke Y, Sugawara S, et al. Trastuzumab emtansine for patients with non-small cell lung cancer positive for human epidermal growth factor receptor 2 exon-20 insertion mutations[J]. Eur J Cancer, 2022, 162: 99-106. doi: 10.1016/j.ejca.2021.11.021
[36] Li BT, Smit EF, Goto Y, et al. Trastuzumab Deruxtecan in HER2-Mutant Non-Small-Cell Lung Cancer[J]. N Engl J Med, 2022, 386(3): 241-251. doi: 10.1056/NEJMoa2112431
[37] Han BH, Li K, Wang QM, et al. Effect of Anlotinib as a Third-Line or Further Treatment on Overall Survival of Patients With Advanced Non-Small Cell Lung Cancer The ALTER 0303 Phase 3 Randomized Clinical Trial[J]. JAMA Oncol, 2018, 4(11): 1569-1575. doi: 10.1001/jamaoncol.2018.3039
[38] Cheng Y, Wang QM, Li K, et al. Anlotinib vs. placebo as third- or further-line treatment for patients with small cell lung cancer: a randomised, double-blind, placebo-controlled Phase 2 study[J]. Brit J Cancer, 2021, 125(3): 366-371. doi: 10.1038/s41416-021-01356-3
[39] Reck M, Rodriguez-Abreu D, Robinson AG, et al. Five-Year Outcomes With Pembrolizumab Versus Chemotherapy for Metastatic Non-Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score≥50%[J]. J Clin Oncol, 2021, 39(21): 2339-2349. doi: 10.1200/JCO.21.00174
[40] Mok TSK, Wu YL, Kudaba I, et al. Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial[J]. Lancet, 2019, 393(10183): 1819-1830. doi: 10.1016/S0140-6736(18)32409-7
[41] Jassem J, de Marinis F, Giaccone G, et al. Updated Overall Survival Analysis From IMpower110: Atezolizumab Versus Platinum-Based Chemotherapy in Treatment-Naive Programmed Death-Ligand 1-Selected NSCLC[J]. J Thorac Oncol, 2021, 16(11): 1872-1882. doi: 10.1016/j.jtho.2021.06.019
[42] Sezer A, Kilickap S, Gumus M, et al. Cemiplimab monotherapy for first-line treatment of advanced non-small-cell lung cancer with PD-L1 of at least 50%: a multicentre, open-label, global, phase 3, randomised, controlled trial[J]. Lancet, 2021, 397(10274): 592-604. doi: 10.1016/S0140-6736(21)00228-2
[43] Peters S, Creelan B, Hellmann MD, et al. Abstract CT082; Impact of tumor mutation burden on the efficacy of first-line nivolumab in stage iv or recurrent non-small cell lung cancer: An exploratory analysis of CheckMate 026[J]. Cancer Res, 2017, 77(13 Suppl): CT082-CT082. http://cancerres.aacrjournals.org/content/77/13_Supplement/CT082
[44] Judd J, Borghaei H. Combining Immunotherapy and Chemotherapy for Non-Small Cell Lung Cancer[J]. Thorac Surg Clin, 2020, 30(2): 199-206. doi: 10.1016/j.thorsurg.2020.01.006
[45] Cheng Y, Zhang L, Hu J, et al. Pembrolizumab Plus Chemotherapy for Chinese Patients With Metastatic Squamous NSCLC in KEYNOTE-407[J]. JTO Clin Res Rep, 2021, 2(10): 100225. http://www.sciencedirect.com/science/article/pii/S2666364321000849
[46] Wang J, Lu S, Yu XM, et al. Tislelizumab Plus Chemotherapy vs. Chemotherapy Alone as First-line Treatment for Advanced Squamous Non-Small-Cell Lung Cancer A Phase 3 Randomized Clinical Trial[J]. JAMA Oncol, 2021, 7(5): 709-717. doi: 10.1001/jamaoncol.2021.0366
[47] Lu S, Wang J, Yu Y, et al. Tislelizumab Plus Chemotherapy as First-Line Treatment for Locally Advanced or Metastatic Nonsquamous NSCLC (RATIONALE 304): A Randomized Phase 3 Trial[J]. J Thorac Oncol, 2021, 16(9): 1512-1522. doi: 10.1016/j.jtho.2021.05.005
[48] Yang Y, Wang Z, Fang J, et al. Final overall survival (OS) data of sintilimab plus pemetrexed (SPP) and platinum as first-line (1L) treatment for locally advanced or metastatic nonsquamous NSCLC (AMnsqNSCLC) in the phaseⅢ ORIENT-11 study[J]. Ann Oncol, 2022, 33: S28-S28.
[49] Zhou CC, Wu L, Fan Y, et al. Sintilimab Plus Platinum and Gemcitabine as First-Line Treatment for Advanced or Metastatic Squamous NSCLC: Results From a Randomized, Double-Blind, Phase 3 Trial (ORIENT-12)[J]. J Thorac Oncol, 2021, 16(9): 1501-1511. doi: 10.1016/j.jtho.2021.04.011
[50] Zhou CC, Chen GY, Huang YC, et al. Camrelizumab plus carboplatin and pemetrexed versus chemotherapy alone in chemotherapy-naive patients with advanced non-squamous non-small-cell lung cancer (CameL): a randomised, open-label, multicentre, phase 3 trial[J]. Lancet Respir Med, 2021, 9(3): 305-314. doi: 10.1016/S2213-2600(20)30365-9
[51] Zhou C, Cheng Y, Chen J, et al. First-line camrelizumab plus carboplatin and paclitaxel for advanced squamous non-small cell lung cancer: Updated overall survival results from the phase Ⅲ CameL-sq trial[J]. Ann Oncol, 2022, 33(suppl 2): S28.
[52] Wang J, Wang Z, Wu L, et al. CHOICE-01: A Phase 3 Study of Toripalimab Versus Placebo in Combination With First-Line Chemotherapy for Advanced NSCLC[J]. J Thorac Oncol, 2021, 16(10): S927-S928. doi: 10.1016/j.jtho.2021.08.181
[53] Lu S, Fang J, Wang ZP, et al. Results from the IMpower132 China cohort: Atezolizumab plus platinum-based chemotherapy in advanced non-small cell lung cancer[J]. Cancer Med, 2023, 12(3): 2666-2667. doi: 10.1002/cam4.5144
[54] Horn L, Mansfield AS, Szczesna A, et al. First-Line Atezolizumab plus Chemotherapy in Extensive-Stage Small-Cell Lung Cancer[J]. N Engl J Med, 2018, 379(23): 2220-2229. doi: 10.1056/NEJMoa1809064
[55] Paz-Ares L, Chen Y, Reinmuth N, et al. Durvalumab +/- tremelimumab plus platinum-etoposide in first-line extensive-stage SCLC (ES-SCLC): 3-year overall survival update from the phaseⅢ CASPIAN study[J]. Ann Oncol, 2021, 32(suppl 5): S1338-S1338.
[56] Paz-Ares LG, Ciuleanu TE, Lee JS, et al. Nivolumab (NIVO) plus ipilimumab (IPI) versus chemotherapy (chemo) as first-line (1L) treatment for advanced non-small cell lung cancer (NSCLC): 4-year update from CheckMate 227[J]. J Clin Oncol, 2021, 39(15 suppl): 9016. http://www.researchgate.net/publication/352077636_Nivolumab_NIVO_plus_ipilimumab_IPI_versus_chemotherapy_chemo_as_first-line_1L_treatment_for_advanced_non-small_cell_lung_cancer_NSCLC_4-year_update_from_CheckMate_227
[57] Reck M, Ciuleanu TE, Cobo M, et al. First-line nivolumab (NIVO) plus ipilimumab (IPI) plus two cycles of chemotherapy (chemo) versus chemo alone (4 cycles) in patients with advanced non-small cell lung cancer (NSCLC): Two-year update from CheckMate 9LA[J]. J Clin Oncol, 2021, 39(15_suppl): 9000. doi: 10.1200/JCO.2021.39.15_suppl.9000
[58] Reinmuth N, Johnson M, Cho BC, et al. Durvalumab +/- Tremelimumab plus Chemotherapy as First-line Treatment for mNSCLC: Results from the Phase 3 POSEIDON Study[J]. J Thorac Oncol, 2021, 16(10_supplement): S844. http://www.sciencedirect.com/science/article/pii/S1556086421024527
[59] Rizvi NA, Cho BC, Reinmuth N. Durvalumab With or Without Tremelimumab vs. Standard Chemotherapy in First-line Treatment of Metastatic Non-Small Cell Lung Cancer: The MYSTIC Phase 3 Randomized Clinical Trial[J]. JAMA Oncol, 2020, 6(5): 661-674. doi: 10.1001/jamaoncol.2020.0237
[60] Boyer M, Sendur MAN, Rodriguez-Abreu D, et al. Pembrolizumab Plus Ipilimumab or Placebo for Metastatic Non-Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score≥50%: Randomized, Double-Blind PhaseⅢ KEYNOTE-598 Study[J]. J Clin Oncol, 2021, 39(21): 2327-2338. doi: 10.1200/JCO.20.03579
[61] Ciciola P, Cascetta P, Bianco C, et al. Combining Immune Checkpoint Inhibitors with Anti-Angiogenic Agents[J]. J Clin Med, 2020, 9(3): 675. doi: 10.3390/jcm9030675
[62] Socinski MA, Nishio M, Jotte RM, et al. IMpower150 Final Overall Survival Analyses for Atezolizumab Plus Bevacizumab and Chemotherapy in First-Line Metastatic Nonsquamous NSCLC[J]. J Thorac Oncol, 2021, 16(11): 1909-1924. doi: 10.1016/j.jtho.2021.07.009
[63] Lu S, Wu L, Jian H, et al. Sintilimab plus bevacizumab biosimilar IBI305 and chemotherapy for patients with EGFR-mutated non-squamous non-small-cell lung cancer who progressed on EGFR tyrosine-kinase inhibitor therapy (ORIENT-31): first interim results from a randomised, double-blind, multicentre, phase 3 trial[J]. Lancet Oncol, 2022, 23(9): 1167-1179. doi: 10.1016/S1470-2045(22)00382-5
[64] Fan Y, Zhao J, Wang QM, et al. Camrelizumab Plus Apatinib in Extensive-Stage SCLC (PASSION): A Multicenter, Two-Stage, Phase 2 Trial[J]. J Thorac Oncol, 2021, 16(2): 299-309. doi: 10.1016/j.jtho.2020.10.002
[65] Yang JCH, Luft A, Jimenez ED, et al. Pembrolizumab (Pembro) with or without lenvatinib (Lenva) in first-line metastatic NSCLC with PD-L1 TPS≥1% (LEAP-007): A phaseⅢ, randomized, double-blind study[J]. Ann Oncol, 2021, 32: S1429-S1430.
[66] Spigel DR, Faivre-Finn C, Gray JE, et al. Five-Year Survival Outcomes From the PACIFIC Trial: Durvalumab After Chemoradiotherapy in StageⅢ Non-Small-Cell Lung Cancer[J]. J Clin Oncol, 2022, 40(12): 1301-1311. doi: 10.1200/JCO.21.01308
[67] Forde PM, Spicer J, Lu S, et al. Neoadjuvant Nivolumab plus Chemotherapy in Resectable Lung Cancer[J]. N Engl J Med, 2022, 386(21): 1973-1985. doi: 10.1056/NEJMoa2202170
[68] Zhou N, Sepesi B, Leung CH, et al. Impact of genomic aberrations and additional therapies on survival outcomes of patients with operable non-small cell lung cancer (NSCLC) from the NEOSTAR study[J]. J Clin Oncol, 2021, 39(15_suppl): 8542. doi: 10.1200/JCO.2021.39.15_suppl.8542
[69] Felip E, Altorki N, Zhou C, et al. Adjuvant atezolizumab after adjuvant chemotherapy in resected stageⅠB-ⅢA non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label, phase 3 trial[J]. Lancet, 2021, 398(10308): 1344-1357. doi: 10.1016/S0140-6736(21)02098-5
[70] Wei J, Liu L, Chen X, et al. Improved sensitivity of integrative DNA and RNA NGS assay on fusion detection[J]. Ann Oncol, 2020, 31: S1105. http://www.sciencedirect.com/science/article/pii/S0923753420413547
[71] Cohen D, Hondelink LM, Solleveld-Westerink N, et al. Optimizing Mutation and Fusion Detection in NSCLC by Sequential DNA and RNA Sequencing[J]. J Thorac Oncol, 2020, 15(6): 1000-1014. doi: 10.1016/j.jtho.2020.01.019
[72] Kurata J, Price K, Banks K, et al. Multiomic, Plasma-Only ctDNA NGS Assay Developed for Minimal Residual Disease (MRD) Detection in Early-Stage NSCLC[J]. J Thorac Oncol, 2021, 16(10): S952-S953. http://www.sciencedirect.com/science/article/pii/S1556086421026435
[73] Liu SY, Wu YL. Biomarker for personalized immunotherapy[J]. Transl Lung Cancer Res, 2019, 8(Suppl 3): S308-S317.
[74] Wang MN, Herbst RS, Boshoff C. Toward personalized treatment approaches for non-small-cell lung cancer[J]. Nat Med, 2021, 27(8): 1345-1356. doi: 10.1038/s41591-021-01450-2
[75] Lim JU. Overcoming Osimertinib Resistance in Advanced Non-small Cell Lung Cancer[J]. Clin Oncol, 2021, 33(10): 619-626. doi: 10.1016/j.clon.2021.07.015
[76] Lim SM, Park CW, Zhang Z, et al. BLU-945, a fourth-generation, potent and highly selective epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) with intracranial activity, demonstrates robust in vivo antitumor activity in models of osimertinib-resistant non-small cell lung cancer (NSCLC)[J]. Cancer Res, 2021, 81(13_Supplement): 1467. doi: 10.1158/1538-7445.AM2021-1467
-
期刊类型引用(2)
1. 苗晓红,袁志英,李媛,韩秀青,李光. 宫颈癌组织含Jumonji结构域的蛋白2B、造血相关PBX相互作用蛋白的表达与预后关系的分析. 中国性科学. 2023(01): 60-65 . 百度学术
2. 徐小艳,王建君,闫琛,李川,刘微,杨金花. ALKBH5促进乳腺浸润性导管癌细胞上皮间质转化和血管生成并增强其侵袭和转移. 细胞与分子免疫学杂志. 2021(04): 355-361 . 百度学术
其他类型引用(0)