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:

尹媛,E-mail:yinyuan@njmu.edu.cn

中图分类号:R734.2

文献标识码:A

文章编号:1007-4368(2023)08-1133-07

DOI:10.7655/NYDXBNS20230814

参考文献 1
RECK M,RODRÍGUEZ⁃ABREU D,ROBINSON A G,et al.Pembrolizumab versus chemotherapy for PD ⁃L1⁃posi⁃ tive non⁃small⁃cell lung cancer[J].N Engl J Med,2016,375(19):1823-1833
参考文献 2
BORGHAEI H,PAZ⁃ARES L,HORN L,et al.Nivolumab versus docetaxel in advanced nonsquamous non⁃small⁃cell lung cancer[J].N Engl J Med,2015,373(17):1627-1639
参考文献 3
HONG L,NEGRAO M V,DIBAJ S S,et al.Programmed death ⁃ ligand 1 heterogeneity and its impact on benefit from immune checkpoint inhibitors in NSCLC[J].J Tho⁃ rac Oncol,2020,15(9):1449-1459
参考文献 4
SCHOENFELD A J,RIZVI H,BANDLAMUDI C,et al.Clinical and molecular correlates of PD ⁃L1 expression in patients with lung adenocarcinomas[J].Ann Oncol,2020,31(5):599-608
参考文献 5
BRUECKL W M,FICKER J H,ZEITLER G.Clinically relevant prognostic and predictive markers for immune ⁃ checkpoint ⁃inhibitor(ICI)therapy in non ⁃ small cell lung cancer(NSCLC)[J].BMC Cancer,2020,20(1):1185-1200
参考文献 6
BAGCHI S,YUAN R,ENGLEMAN E G.Immune check⁃ point inhibitors for the treatment of cancer:clinical im⁃ pact and mechanisms of response and resistance[J].An⁃ nu Rev Pathol,2021,16:223-249
参考文献 7
PENG L,WANG Y,LIU F,et al.Peripheral blood mark⁃ ers predictive of outcome and immune ⁃ related adverse events in advanced non ⁃ small cell lung cancer treated with PD ⁃ 1 inhibitors[J].Cancer Immunol Immunother,2020,69(9):1813-1822
参考文献 8
RUIZ ⁃ BAÑOBRE J,ARESES ⁃ MANRIQUE M C,MOS⁃ QUERA ⁃MARTÍNEZ J,et al.Evaluation of the lung im⁃ mune prognostic index in advanced non ⁃ small cell lung cancer patients under nivolumab monotherapy[J].Transl Lung Cancer Res,2019,8(6):1078-1085
参考文献 9
ZHUO M,CHEN H,ZHANG T,et al.The potential pre⁃ dictive value of circulating immune cell ratio and tumor marker in atezolizumab treated advanced non ⁃ small cell lung cancer patients[J].Cancer Biomark,2018,22(3):467-476
参考文献 10
GANDINI S,MASSI D,MANDALÀ M.PD⁃L1 expression in cancer patients receiving anti PD⁃1/PD⁃L1 antibodies:a systematic review and meta⁃analysis[J].Crit Rev Oncol Hematol,2016,100:88-98
参考文献 11
MOK T,WU Y L,KUDABA I,et al.Pembrolizumab ver⁃ sus chemotherapy for previously untreated,PD ⁃ L1 ⁃ ex⁃ pressing,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
参考文献 12
HELLMANN M D,CIULEANU T E,PLUZANSKI A,et al.Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden[J].N Engl J Med,2018,378(22):2093-2104
参考文献 13
MARABELLE A,FAKIH M,LOPEZ J,et al.Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab:prospective biomarker analysis of the multicohort,open ⁃ label,phase 2 KEYNOTE ⁃ 158 study[J].Lancet Oncol,2020,21(10):1353-1365
参考文献 14
ZHAO P,LI L,JIANG X,et al.Mismatch repair deficien⁃ cy/microsatellite instability ⁃ high as a predictor for anti ⁃ PD⁃1/PD ⁃L1 immunotherapy efficacy[J].J Hematol On⁃ col,2019,12(1):54-67
参考文献 15
SCOGNAMIGLIO G,De CHIARA A,Di BONITO M,et al.Variability in immunohistochemical detection of pro⁃ grammed death ligand 1(PD ⁃ L1)in cancer tissue types [J].Int J Mol Sci,2016,17(5):790-800
参考文献 16
HO W J,YARCHOAN M,HOPKINS A,et al.Association between pretreatment lymphocyte count and response to PD1 inhibitors in head and neck squamous cell carcino⁃ mas[J].J Immunother Cancer,2018,6(1):84-91
参考文献 17
AMERATUNGA M,CHÉNARD ⁃POIRIER M,MORENO C I,et al.Neutrophil⁃lymphocyte ratio kinetics in patients with advanced solid tumours on phase I trials of PD⁃1/PD⁃ L1 inhibitors[J].Eur J Cancer,2018,89:56-63
参考文献 18
HUEMER F,LANG D,WESTPHAL T,et al.Baseline ab⁃ solute lymphocyte count and ECOG performance score are associated with survival in advanced non ⁃ small cell lung cancer undergoing PD⁃1/PD⁃L1 blockade[J].J Clin Med,2019,8(7):1014-1030
参考文献 19
DIEM S,SCHMID S,KRAPF M,et al.Neutrophil⁃to⁃lym⁃ phocyte ratio(NLR)and platelet ⁃ to ⁃ lymphocyte ratio(PLR)as prognostic markers in patients with non ⁃ small cell lung cancer(NSCLC)treated with nivolumab[J].Lung Cancer,2017,111:176-181
参考文献 20
BUDER⁃BAKHAYA K,HASSEL J C.Biomarkers for clin⁃ ical benefit of immune checkpoint inhibitor treatment ⁃ a review from the melanoma perspective and beyond[J].Front Immunol,2018,9:1474-1490
参考文献 21
PAN Y,YU Y,WANG X,et al.Tumor⁃associated macro⁃ phages in tumor immunity[J].Front Immunol,2020,11:583-591
参考文献 22
LIN Y,XU J,LAN H.Tumor ⁃associated macrophages in tumor metastasis:biological roles and clinical therapeu⁃ tic applications[J].J Hematol Oncol,2019,12(1):76-91
参考文献 23
CARRETERO R,SEKTIOGLU I M,GARBI N,et al.Eo⁃ sinophils orchestrate cancer rejection by normalizing tu⁃ mor vessels and enhancing infiltration of CD8(+)T cells [J].Nat Immunol,2015,16(6):609-617
参考文献 24
GATAULT S,DELBEKE M,DRISS V,et al.IL⁃18 is in⁃ volved in eosinophil⁃mediated tumoricidal activity against a colon carcinoma cell line by upregulating LFA ⁃ 1 and ICAM⁃1[J].J Immunol,2015,195(5):2483-2492
参考文献 25
HOLUB K,BIETE A.Impact of systemic inflammation biomarkers on the survival outcomes of cervical cancer patients[J].Clin Transl Oncol,2019,21(7):836-844
参考文献 26
ROSNER S,KWONG E,SHOUSHTARI A N,et al.Pe⁃ ripheral blood clinical laboratory variables associated with outcomes following combination nivolumab and ipili⁃ mumab immunotherapy in melanoma[J].Cancer Med,2018,7(3):690-697
参考文献 27
REINA ⁃CAMPOS M,SCHARPING N E,GOLDRATH A W.CD8(+)T cell metabolism in infection and cancer[J].Nat Rev Immunol,2021,21(11):718-738
参考文献 28
LI F,LI C,CAI X,et al.The association between CD8+ tu⁃ mor ⁃infiltrating lymphocytes and the clinical outcome of cancer immunotherapy:a systematic review and meta ⁃ analysis[J].EClinicalMedicine,2021,41:2589-2600
参考文献 29
MIAO K,ZHANG X,WANG H,et al.Peripheral blood lymphocyte subsets predict the efficacy of immune check⁃ point inhibitors in non ⁃ small cell lung cancer[J].Front Immunol,2022,13:912-921
目录contents

    摘要

    目的:研究基线外周血细胞参数对非小细胞肺癌(non-small cell lung cancer,NSCLC)患者一线使用免疫检查点抑制剂联合化疗疗效的预测价值,探索外周血中的预测性生物标志物。方法:回顾性分析2020年1月—2022年9月于南京医科大学第一附属医院一线使用程序性死亡蛋白1(programmed cell death protein1,PD-1)单抗治疗联合化疗的84例不能手术的Ⅲ期或Ⅳ期NSCLC患者的临床资料、血细胞分类、淋巴细胞亚群以及炎症指标包括C反应蛋白(C-reactive protein,CRP)、降钙素原 (procalcitonin,PCT)及白介素-6(interleukin,IL-6),根据RECIST1.1标准评价疗效并随访无进展生存期(progression-free surviv- al,PFS),使用COX比例风险回归模型进行单因素和多因素生存分析,筛选治疗前的基线参数中与疗效相关的指标。采用Ka- plan-Meier法绘制生存曲线,用Log-rank检验比较组间生存率。结果:中位随访时间为9.93(5.68~15.38)个月,中位无进展生存期(median progress free survival,mPFS)为9.42(5.63~12.88)个月。COX单因素回归分析和Kaplan-Meier生存曲线结果显示,PD -L1表达是免疫疗效的独立预测因素(HR=0.299,P=0.003);高单核细胞(monocyte,MO)百分比组相比低MO%组患者PFS更长 (HR=0.313,95%CI:0.137~0.711,P=0.006,mPFS 14.37个月vs. 7.60个月);高嗜酸性粒细胞(eosinophil,EOS)百分比组相比低 EOS%组也有更长的PFS(HR=0.296,95%CI:0.092~0.958,P=0.042,mPFS 13.87个月vs. 7.93个月)。对以上2个参数进行COX 多因素回归分析,P均<0.05。淋巴细胞亚群、炎症指标(CRP、PCT及IL-6)与mPFS均无显著相关性(P>0.05)。结论:对于 NSCLC患者,高水平的MO%和EOS%与较长的控制时间相关,可作为预测一线使用化疗联合免疫治疗疗效的潜在生物标志物,为临床选择获益人群提供一定参考。

    Abstract

    Objective:The current study aims to investigate the correlation between baseline peripheral blood biomarkers and outcomes in patients with non - small cell lung cancer(NSCLC)receiving immune checkpoint inhibitors(ICIs). Methods:A retrospective analysis was conducted of 84 patients with stage Ⅲ or Ⅳ NSCLC,who received PD - 1 inhibitors combined with chemotherapy as first-line treatment in the First Affiliated Hospital of Nanjing Medical University betwee January 2020 and September 2022. The efficacy was evaluated according to RECIST1.1 criteria and the patients were followed up of progression-free survival(PFS). COX regression models were used in the univariate and multivariate survival analysis to assess prognostic effect of baseline peripheral blood parameters before treatments. Kaplan - Meier method as used for survival analysis. Log - rank tests were used to analyze the survival rates between groups. Results:The median follow - up time was 9.93(5.68~15.38)months,and the median progression -free survival(mPFS)was 9.42(5.63~12.88)months. COX univariate regression analysis and Kaplan-Meier survival curves showed that PD- L1 was an independent predictor of immune efficacy(HR=0.299,P=0.003),patients with a high monocyte percentage(≥8.15%)had a better PFS than those with low monocyte percentage(HR=0.313,95% CI:0.137~0.711,P=0.006,mPFS 14.37 months vs. 7.60 months). Patients with a high eosinophil percentage(≥1.90%)also had a better PFS than those with low eosinophil percentage(HR= 0.296,95%CI:0.092~0.958,P=0.042,mPFS 13.87 months vs. 7.93 months). COX multivariate regression analysis showed that both high monocyte percentage and high monocyte percentage parameters were independently associated with better PFS. However, peripheral blood lymphocyte subsets and inflammatory parameters had no predictive role for ICI efficacy. Conclusion:For patients with NSCLC,the percentage of monocytes and eosinophils may be useful predictive markers of response to chemotherapy combined with immunotherapy as first-line therapy,and higher percentages of moncytes and eosinophils were associated with longer progression- free survival time.

  • 由于免疫治疗在癌症应用的突破性进展,对于局部晚期或转移性非小细胞肺癌(non ⁃ small cell lung cancer,NSCLC)患者,指南已纳入包含免疫检查点抑制剂(immune checkpoint inhibitor,ICI)的一线、二线治疗方案[1]。ICI可单独治疗或与化疗、抗血管生成药物以及其他形式的 ICI 联合治疗,部分 NSCLC 患者从 ICI 中获益明显,具有良好的疗效和更久的生存时间[2]。然而还有些患者在使用的初始阶段增加死亡风险,这可能与原发性耐药、肿瘤超进展生长或免疫相关不良事件有关,因此评估患者使用 ICI 是否获益至关重要。目前临床应用成熟的预测性标志物主要有程序性死亡配体 1 (programmed death⁃ligand 1,PD⁃L1)、肿瘤突变负荷 (tumor mutation burden,TMB)和微卫星不稳定性 (microsatellite instability,MSI)等,这些生物标志物的检测已广泛开展,但存在组织异质性[3]、基因检测的规范化、价格昂贵、检测时间长等问题[4],使得临床应用受到一定限制。目前免疫药物在一线方案有越来越广泛的适应证,对于NSCLC,探索可以简易获取的生物标志物有助于综合性评估选择免疫治疗的受益人群,对临床制定个体化策略有一定指导价值。

  • 据报道除PD⁃L1等肿瘤的生物特征性参数外,临床诊疗中的常规血液检查指标可作为ICI治疗和预后相关的血液生物标志物[5]。炎症反应被认为是肿瘤患者免疫抵抗的一种机制,机体炎症状态已被证明与多种癌症类型的不良结局有关。在头颈部鳞癌、黑色素瘤或肺癌等多种实体肿瘤中,一些血液炎症指标被发现与免疫治疗疗效及结局有关,如淋巴细胞计数、中性粒细胞与淋巴细胞比值(neutro⁃ phil to lymphocyte ration,NLR)、中性粒细胞(/ 白细胞-中性粒细胞)比值(dNLR)和血小板与淋巴细胞比值(platelet to lymphocyte ratio,PLR)等[6]。对于单用 PD⁃1 抑制剂免疫治疗的 NSCLC,高水平 NLR 是疗效差、预后不良的危险因素[7-8]。此外,免疫治疗的关键机制是淋巴细胞的激活,近期有临床研究发现,淋巴细胞亚群与ICI的疗效有关[9]。对于不可手术的NSCLC患者,2021年中国临床肿瘤学会(CSCO) 已将免疫治疗联合化疗作为一线治疗方案纳入指南,目前尚不清楚接受联合治疗的NSCLC患者外周血中的各类细胞分类水平(包括血常规中各类血细胞水平、淋巴细胞亚群等)是否与治疗疗效相关。本研究旨在寻找外周血中有助于预测不可手术NSCLC患者一线使用ICI联合化疗疗效的标志物。

  • 1 对象和方法

  • 1.1 对象

  • 纳入 84 例 2020 年 1 月—2022 年 9 月于南京医科大学第一附属医院接受治疗的 NSCLC 患者。纳入标准:①病理学明确诊断为 NSCLC;②初诊时临床分期为不可手术Ⅲ期或Ⅳ期;③一线治疗采用含铂双药化疗联合抗PD⁃1单抗免疫治疗(包括帕博利珠单抗、信迪利单抗、卡瑞利珠单抗及替雷利珠单抗);④免疫治疗过程中病历资料、血常规报告、淋巴细胞亚群、炎症标志物、用于评估疗效的影像学资料完整。排除标准:①接受治疗时合并过敏性疾病、血液系统疾病、自身免疫性疾病或免疫抑制状态;②ECOG PS(Eastern Cooperative Oncology Group⁃ performance status)评分≥2分;③治疗前1个月合并发热或感染;④治疗前1个月使用类固醇激素全身治疗;⑤存在已知的表皮生长因子受体(epidermal growth factor receptor,EGFR)敏感突变、间变性淋巴瘤激酶(anaplastic lymphoma kinase,ALK)、ROS 原癌基因 1(ROS proto⁃oncogene1,ROS1)或转染原癌基因时重排(rearranged during transfection proto⁃on⁃ cogene,RET)融合等《2021年CSCO非小细胞肺癌诊疗指南》包括的驱动基因阳性的情况。

  • 1.2 方法

  • 1.2.1 临床资料和治疗方案

  • 以首次使用 ICI 的时间为观察起点,根据实体瘤疗效评估标准(RECIST1.1)评价疗效,通过计算机断层扫描(CT)每8~12周评估1次药物疗效,直至出现肿瘤进展或死亡为观察终点。无进展生存期 (progression⁃free survival,PFS)定义为从治疗开始到临床或影像学进展或死亡的时间,随访时间截至 2022年9月。

  • 免疫治疗包括:帕博利珠单抗、信迪利单抗、替雷利珠单抗、卡瑞利珠单抗,剂量及频次均按照药物说明书推荐使用。联合化疗为采用《2021年CSCO非小细胞肺癌诊疗指南》推荐的含铂双药化疗,药物根据肿瘤组织学确定,包括培美曲塞、紫杉醇脂质体/白蛋白紫杉醇、吉西他滨等。

  • 1.2.2 观察指标

  • 通过电子病历系统收集的临床指标包括:年龄、性别、体重指数(body mass index,BMI)、病理类型、TNM 分期、吸烟史、ECOG PS 评分、驱动基因突变状态、TMB、组织PD⁃L1表达水平、既往方案、治疗线数等。鉴于《2021年CSCO 非小细胞肺癌诊疗指南》已将帕博利珠单抗单药一线治疗作为Ⅰ级推荐用于PD⁃L1肿瘤细胞阳性比例评分(TPS)≥1%的Ⅳ期驱动基因阴性的NSCLC患者,本研究选择TPS≥1% 作为PD⁃L1表达阳性的标准。

  • 基线血常规为在接受 PD⁃1抑制剂治疗前检测的血常规,观察指标包括各类血细胞参数和相关比值。通过血常规结果计算的比值参数包括:NLR、 dNLR、PLR 以及淋巴细胞/单核细胞比值(LMR)。基线淋巴细胞亚群为在接受 PD⁃1抑制剂治疗前通过流式细胞术检测的淋巴细胞亚群相对计数,观察指标包括 CD3 + 细胞、CD3 + CD4 + 细胞(辅助性 T 细胞)、CD3+ CD8+ 细胞(抑制性T细胞)、CD16+ CD56+ 细胞(NK 细胞)、CD19+ 细胞(B 细胞)及调节性 T 细胞 (Treg)细胞,细胞计数单位均为109 个/L。

  • 1.3 统计学方法

  • 应用SPSS 26.0统计学软件进行数据分析。正态分布的计量资料用均数±标准差(x-±s)表示,时间以中位数(四分位数)[MP25P75)]表示,根据各参数的中位数患者被分组为低水平和高水平组。使用 Kaplan⁃Meier 法进行生存分析,并使用对数秩检验对生存曲线进行组间比较。应用COX 比例风险回归模型筛选与PFS相关的变量,结果通过风险比 (hazard ratio,HR)和P值来描述。将单因素分析中差异具有统计学意义的变量纳入多因素分析。在整个分析过程中,P <0.05为差异有统计学意义。

  • 2 结果

  • 2.1 基线资料

  • 本研究共纳入 84 例患者,所有患者均为一线使用免疫治疗联合化疗,所有患者ECOGPS评分为0 或 1 分;其中,男 68 例(81%),女 16 例(19%);年龄 44~79 岁,平均(63.05±8.26)岁。40 例(47.6%)腺癌,44 例(52.4%)鳞癌;28 例(33.3%)Ⅲ期,56 例 (66.7%)Ⅳ期;49例患者进行基因检测,37例检测出基因突变,12例未发现基因突变,35例基因检测情况不详;53 例(63.1%)有吸烟史;使用帕博利珠单抗、卡瑞利珠单抗、信迪利单抗、替雷利珠单抗治疗的患者分别为14例、15例、25例、30例。进行TMB 检测24例,60例TMB检测情况不详。检测PD⁃L1表达的患者有44例,其中TPS≥1%有33例,另外40例 PD⁃L1检测情况不详。

  • 2.2 生存分析

  • 总人群的中位无进展生存期(mPFS)为 9.42 (5.63,12.88)个月。对各项临床特征分组,纳入 COX 单因素回归分析,以探究能与疗效相关的因素。分析显示PD⁃L1表达阳性(TPS≥1%)是免疫疗效的独立预测因素(HR=0.299,P=0.003)。此外,女性患者、有吸烟史等临床因素可能与更长的PFS相关,但本研究差异无统计学意义(表1)。

  • 对血液检验中的变量均以中位数作为截断值,将所有变量分为高和低两组,进行COX比例风险回归模型分析所有参数与PFS的相关性(表2)。结果显示,高单核细胞百分比(MO%≥8.15%)是延长患者 PFS 的因素(HR=0.313,95%CI:0.137~0.711,P= 0.006,mPFS 14.37个月vs.7.60个月),另外高嗜酸性粒细胞百分比(EOS%≥1.9%)是延长患者PFS的另一因素(HR=0.296,95%CI:0.092~0.958,P=0.042),高比例组均能降低进展风险。将两者纳入多因素回归分析,差异仍具有统计学意义(分别为 P=0.030 和 0.022)。将基线淋巴细胞亚群、C反应蛋白、降钙素原及白介素⁃6纳入单因素COX回归分析,结果显示各类免疫细胞亚群及炎症指标与mPFS均无相关性(表3)。

  • 对两因素分别绘制相应的生存曲线(图1、2)。高 MO%(≥8.15%)组患者的 PFS 显著长于低 MO% 组的患者(P=0.004),mPFS 分别为 14.37 个月(95% CI:9.77~18.97)和 7.60 个月(95% CI:5.23~9.97)。高 EOS%(≥1.9%)组患者相较于低EOS%组也同样有更长的 PFS(mPFS:13.87 个月 vs.7.93 个月,P= 0.003)。结果显示血 MO%和 EOS%是 NSCLC 患者免疫治疗疗效的有效预测因素。

  • 表1 84例非小细胞肺癌患者临床资料回归分析

  • Table1 Clinical characteristics and COX regression analysis of 84 patients with NSCLC

  • 表2 NSCLC患者血常规参数与PFS单因素和多因素COX回归分析

  • Table2 Univariate and multivariate analysis of PFS with peripheral blood counts

  • 3 讨论

  • 目前主要的免疫治疗预测标志物包括PD⁃L1、 TMB和MSI等,已在临床广泛应用于筛选免疫治疗中可能获益的患者,对肿瘤免疫治疗方案的选择具有重要指导价值。多项针对肺癌免疫药物治疗的临床研究表明,PD⁃L1高表达预示着患者具有更好的疗效和更长的控制时间,2021年CSCO将帕博利珠单抗单药一线治疗作为Ⅰ级推荐,用于 PD ⁃L1 TPS≥1%的Ⅳ期驱动基因阴性的 NSCLC 患者[10-11]。 TMB 是另一种备受关注的疗效评估的预测性生物标志物,TMB 高的患者对免疫药物的响应率更高、临床获益更多[12-13],FDA已批准帕博利珠单抗用于 TMB⁃H、既往治疗后疾病进展的不可手术或转移性的实体瘤患者的治疗。错配修复途径在细胞的 DNA 复制过程中发挥识别和修复错配碱基的作用。错配修复缺陷(different mismatch repair,dMMR)会导致MSI的高突变表型,由于基因突变负荷的积累和新抗原的产生,刺激宿主的免疫反应。多项临床研究表明,微卫星不稳定性高与免疫治疗的良好预后显著相关。然而,只有极少数肺癌患者具有MSI⁃H 特征,一项基于 18 个癌种 5 930 个癌症外显子的 MSI 位点的研究分析发现,MSI⁃H 在肺腺癌和肺鳞癌中的比例均不到1%[14]。TMB、MSI和PD⁃L1的交集在各个癌种中有所不同,一项泛癌种的研究发现只有 0.6%的肿瘤同时为MSI⁃H、高TMB且PD⁃L1阳性,这个比例在肺癌中更低。此外,以上讨论的指标,主要受到肿瘤异质性[15]、样本类型、检测技术和标准化的影响,因此,临床上如果能发现更多方便易得的标志物,则有望进一步构建涵盖多因素的个体化预测模型,为肺癌的精准免疫治疗提供方向。

  • 表3 NSCLC患者T细胞亚群和炎症指标与PFS单因素COX回归分析

  • Table3 Univariate Cox regression of PFS with lymphocyte subsets and inflammatory parameters

  • 图1 高MO%组与低MO%组NSCLC患者PFS生存曲线

  • Figure1 Kaplan⁃Meier survival analysis for PFS accord⁃ ing to MO⁃ratio

  • 图2 高EOS%组与低EOS%组NSCLC患者PFS生存曲线

  • Figure2 Kaplan⁃Meier survival analysis for PFS accord⁃ ing to EOS⁃ratio

  • 近年来外周血标志物作为一项无创性、方便易行的检测项目逐渐受到了研究者的关注。此类检测指标具有廉价、便捷及可重复性等优点,近年来关于外周血标志物的研究越来越多。目前多项研究已证实其与多种实体瘤如头颈部鳞癌、黑色素瘤、支气管肺癌等免疫治疗的预后相关[16-18]。然而对于免疫联合化疗的NSCLC患者,外周血参数是否与患者预后存在相关性目前尚不清楚。炎症状态与癌症的预后密切相关,它有利于微环境中癌细胞生长和扩散以及致癌信号通路的激活。目前已报道一些与炎症相关的外周血参数的预后价值,包括淋巴细胞水平、dNLR、NLR、LMR、PLR[519]。已有研究表明 NLR与接受免疫治疗的黑色素瘤患者的预后有关[20]。同样,治疗前高水平NLR被认为是NSCLC的不良预后指标[5]。本研究中未能发现以上指标与 PFS有明确相关性,造成这种差异的一个可能因素是研究对象和治疗方案存在差异。本研究的研究对象为免疫联合化疗患者,NLR相关的研究多集中于单药免疫治疗人群,不同癌种、不同方案的预测指标有待扩充病例数后进一步对比验证。

  • 本研究发现治疗前单核细胞百分比可影响患者的PFS,单核细胞百分比高的患者PFS更长,表明单核细胞百分比可能是选择ICI联合化学治疗时评估是否受益的一个有用参数。单核细胞是先天免疫系统的关键组成部分,在维持组织稳态中起着至关重要的作用。在肿瘤微环境中,外周血单核细胞可被循环肿瘤细胞(circulating tumor cell,CTC)产生的趋化因子募集至肿瘤微环境(tumor microenviron⁃ ment,TME)中,转变为肿瘤相关巨噬细胞(tumor⁃as⁃ sociated macrophage,TAM),是肿瘤基质中主要的免疫细胞之一[21]。根据激活状态的不同分为M1和M2 两种功能相反的亚型,前者通常发挥抗肿瘤作用,包括直接介导细胞毒性和抗体依赖性细胞介导的细胞毒性(ADCC)杀死肿瘤细胞;后者可促进肿瘤细胞的发生和转移。M1和M2巨噬细胞都具有高度的可塑性,因此可以在肿瘤微环境变化或治疗干预时相互转化[22]。本文猜测,高单核细胞百分比的患者之所以可以提高联合治疗的疗效,其潜在机制可能和单核细胞向巨噬细胞的不同亚型分化有关,但需要进一步的基础研究证据来支持。

  • 有研究发现,嗜酸性粒细胞对肿瘤微环境中的其他免疫细胞具有调节功能。活化的嗜酸性粒细胞可通过分泌免疫介质促进 CD8+ T 淋巴细胞浸润、促进巨噬细胞极化和抑制肿瘤血管生成来抑制肿瘤生长[23]。此外,嗜酸性粒细胞还可对肿瘤细胞直接产生细胞毒功能,从而发挥直接或间接抗肿瘤作用[24]。曾有研究发现宫颈癌患者中高水平嗜酸性粒细胞的患者生存时间更长[25]。一项回顾性研究纳入607例接受帕博利珠单抗治疗的黑色素瘤患者,发现高水平的 EOS%(≥1.5%)与良好的总生存期相关[26]。这个结果与本研究发现的高EOS%是延长肺癌患者PFS的独立预测因素的结论一致。

  • 本研究还初步探讨了外周血淋巴细胞亚群对 ICI 疗效的预测作用。T 细胞是参与免疫应答的主要效应细胞,一般可分为细胞毒性T细胞(CD8+)和调节性T细胞(CD4+)。CD8+ T细胞具有细胞毒性和从外周血迁移到肿瘤组织的能力[27]。Li 等[28] 研究表明,肿瘤浸润CD8+ T细胞的数量与PD⁃1抑制剂的抗肿瘤疗效密切相关,然而外周血循环CD8+ T细胞与 NSCLC 的获益无关。本研究结果同样认为基线CD8+ T细胞与ICI联合治疗的疗效没有相关性。与 CD8+ T细胞直接杀伤肿瘤的作用不同,CD4+ T细胞更多地发挥免疫调节和旁分泌作用。最近临床研究表明,CD4+ CD45RA- T 细胞与 NSCLC 患者 ICI 治疗的预后相关[29],而CD45RA-是记忆性T细胞的特征,本研究未进一步进行该记忆亚群分类,因此尚未得出阳性结果。

  • 由于本研究的随访时间相对较短,还不能对患者的总体生存时间(overall survival,OS)相关性进行研究。因此,后期需要探索上述指标对OS的预测价值。考虑本研究为单中心研究且样本量偏小,证据级别有限,有待扩大样本量进一步研究证实。

  • 综上所述,外周血细胞中的单核细胞百分比和嗜酸性粒细胞百分比水平可作为 NSCLC 患者一线使用免疫联合化疗预测疗效的潜在临床标志物,为今后的临床工作提供更快捷有效的参考价值。

  • 参考文献

    • [1] RECK M,RODRÍGUEZ⁃ABREU D,ROBINSON A G,et al.Pembrolizumab versus chemotherapy for PD ⁃L1⁃posi⁃ tive non⁃small⁃cell lung cancer[J].N Engl J Med,2016,375(19):1823-1833

    • [2] BORGHAEI H,PAZ⁃ARES L,HORN L,et al.Nivolumab versus docetaxel in advanced nonsquamous non⁃small⁃cell lung cancer[J].N Engl J Med,2015,373(17):1627-1639

    • [3] HONG L,NEGRAO M V,DIBAJ S S,et al.Programmed death ⁃ ligand 1 heterogeneity and its impact on benefit from immune checkpoint inhibitors in NSCLC[J].J Tho⁃ rac Oncol,2020,15(9):1449-1459

    • [4] SCHOENFELD A J,RIZVI H,BANDLAMUDI C,et al.Clinical and molecular correlates of PD ⁃L1 expression in patients with lung adenocarcinomas[J].Ann Oncol,2020,31(5):599-608

    • [5] BRUECKL W M,FICKER J H,ZEITLER G.Clinically relevant prognostic and predictive markers for immune ⁃ checkpoint ⁃inhibitor(ICI)therapy in non ⁃ small cell lung cancer(NSCLC)[J].BMC Cancer,2020,20(1):1185-1200

    • [6] BAGCHI S,YUAN R,ENGLEMAN E G.Immune check⁃ point inhibitors for the treatment of cancer:clinical im⁃ pact and mechanisms of response and resistance[J].An⁃ nu Rev Pathol,2021,16:223-249

    • [7] PENG L,WANG Y,LIU F,et al.Peripheral blood mark⁃ ers predictive of outcome and immune ⁃ related adverse events in advanced non ⁃ small cell lung cancer treated with PD ⁃ 1 inhibitors[J].Cancer Immunol Immunother,2020,69(9):1813-1822

    • [8] RUIZ ⁃ BAÑOBRE J,ARESES ⁃ MANRIQUE M C,MOS⁃ QUERA ⁃MARTÍNEZ J,et al.Evaluation of the lung im⁃ mune prognostic index in advanced non ⁃ small cell lung cancer patients under nivolumab monotherapy[J].Transl Lung Cancer Res,2019,8(6):1078-1085

    • [9] ZHUO M,CHEN H,ZHANG T,et al.The potential pre⁃ dictive value of circulating immune cell ratio and tumor marker in atezolizumab treated advanced non ⁃ small cell lung cancer patients[J].Cancer Biomark,2018,22(3):467-476

    • [10] GANDINI S,MASSI D,MANDALÀ M.PD⁃L1 expression in cancer patients receiving anti PD⁃1/PD⁃L1 antibodies:a systematic review and meta⁃analysis[J].Crit Rev Oncol Hematol,2016,100:88-98

    • [11] MOK T,WU Y L,KUDABA I,et al.Pembrolizumab ver⁃ sus chemotherapy for previously untreated,PD ⁃ L1 ⁃ ex⁃ pressing,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

    • [12] HELLMANN M D,CIULEANU T E,PLUZANSKI A,et al.Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden[J].N Engl J Med,2018,378(22):2093-2104

    • [13] MARABELLE A,FAKIH M,LOPEZ J,et al.Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab:prospective biomarker analysis of the multicohort,open ⁃ label,phase 2 KEYNOTE ⁃ 158 study[J].Lancet Oncol,2020,21(10):1353-1365

    • [14] ZHAO P,LI L,JIANG X,et al.Mismatch repair deficien⁃ cy/microsatellite instability ⁃ high as a predictor for anti ⁃ PD⁃1/PD ⁃L1 immunotherapy efficacy[J].J Hematol On⁃ col,2019,12(1):54-67

    • [15] SCOGNAMIGLIO G,De CHIARA A,Di BONITO M,et al.Variability in immunohistochemical detection of pro⁃ grammed death ligand 1(PD ⁃ L1)in cancer tissue types [J].Int J Mol Sci,2016,17(5):790-800

    • [16] HO W J,YARCHOAN M,HOPKINS A,et al.Association between pretreatment lymphocyte count and response to PD1 inhibitors in head and neck squamous cell carcino⁃ mas[J].J Immunother Cancer,2018,6(1):84-91

    • [17] AMERATUNGA M,CHÉNARD ⁃POIRIER M,MORENO C I,et al.Neutrophil⁃lymphocyte ratio kinetics in patients with advanced solid tumours on phase I trials of PD⁃1/PD⁃ L1 inhibitors[J].Eur J Cancer,2018,89:56-63

    • [18] HUEMER F,LANG D,WESTPHAL T,et al.Baseline ab⁃ solute lymphocyte count and ECOG performance score are associated with survival in advanced non ⁃ small cell lung cancer undergoing PD⁃1/PD⁃L1 blockade[J].J Clin Med,2019,8(7):1014-1030

    • [19] DIEM S,SCHMID S,KRAPF M,et al.Neutrophil⁃to⁃lym⁃ phocyte ratio(NLR)and platelet ⁃ to ⁃ lymphocyte ratio(PLR)as prognostic markers in patients with non ⁃ small cell lung cancer(NSCLC)treated with nivolumab[J].Lung Cancer,2017,111:176-181

    • [20] BUDER⁃BAKHAYA K,HASSEL J C.Biomarkers for clin⁃ ical benefit of immune checkpoint inhibitor treatment ⁃ a review from the melanoma perspective and beyond[J].Front Immunol,2018,9:1474-1490

    • [21] PAN Y,YU Y,WANG X,et al.Tumor⁃associated macro⁃ phages in tumor immunity[J].Front Immunol,2020,11:583-591

    • [22] LIN Y,XU J,LAN H.Tumor ⁃associated macrophages in tumor metastasis:biological roles and clinical therapeu⁃ tic applications[J].J Hematol Oncol,2019,12(1):76-91

    • [23] CARRETERO R,SEKTIOGLU I M,GARBI N,et al.Eo⁃ sinophils orchestrate cancer rejection by normalizing tu⁃ mor vessels and enhancing infiltration of CD8(+)T cells [J].Nat Immunol,2015,16(6):609-617

    • [24] GATAULT S,DELBEKE M,DRISS V,et al.IL⁃18 is in⁃ volved in eosinophil⁃mediated tumoricidal activity against a colon carcinoma cell line by upregulating LFA ⁃ 1 and ICAM⁃1[J].J Immunol,2015,195(5):2483-2492

    • [25] HOLUB K,BIETE A.Impact of systemic inflammation biomarkers on the survival outcomes of cervical cancer patients[J].Clin Transl Oncol,2019,21(7):836-844

    • [26] ROSNER S,KWONG E,SHOUSHTARI A N,et al.Pe⁃ ripheral blood clinical laboratory variables associated with outcomes following combination nivolumab and ipili⁃ mumab immunotherapy in melanoma[J].Cancer Med,2018,7(3):690-697

    • [27] REINA ⁃CAMPOS M,SCHARPING N E,GOLDRATH A W.CD8(+)T cell metabolism in infection and cancer[J].Nat Rev Immunol,2021,21(11):718-738

    • [28] LI F,LI C,CAI X,et al.The association between CD8+ tu⁃ mor ⁃infiltrating lymphocytes and the clinical outcome of cancer immunotherapy:a systematic review and meta ⁃ analysis[J].EClinicalMedicine,2021,41:2589-2600

    • [29] MIAO K,ZHANG X,WANG H,et al.Peripheral blood lymphocyte subsets predict the efficacy of immune check⁃ point inhibitors in non ⁃ small cell lung cancer[J].Front Immunol,2022,13:912-921

  • 参考文献

    • [1] RECK M,RODRÍGUEZ⁃ABREU D,ROBINSON A G,et al.Pembrolizumab versus chemotherapy for PD ⁃L1⁃posi⁃ tive non⁃small⁃cell lung cancer[J].N Engl J Med,2016,375(19):1823-1833

    • [2] BORGHAEI H,PAZ⁃ARES L,HORN L,et al.Nivolumab versus docetaxel in advanced nonsquamous non⁃small⁃cell lung cancer[J].N Engl J Med,2015,373(17):1627-1639

    • [3] HONG L,NEGRAO M V,DIBAJ S S,et al.Programmed death ⁃ ligand 1 heterogeneity and its impact on benefit from immune checkpoint inhibitors in NSCLC[J].J Tho⁃ rac Oncol,2020,15(9):1449-1459

    • [4] SCHOENFELD A J,RIZVI H,BANDLAMUDI C,et al.Clinical and molecular correlates of PD ⁃L1 expression in patients with lung adenocarcinomas[J].Ann Oncol,2020,31(5):599-608

    • [5] BRUECKL W M,FICKER J H,ZEITLER G.Clinically relevant prognostic and predictive markers for immune ⁃ checkpoint ⁃inhibitor(ICI)therapy in non ⁃ small cell lung cancer(NSCLC)[J].BMC Cancer,2020,20(1):1185-1200

    • [6] BAGCHI S,YUAN R,ENGLEMAN E G.Immune check⁃ point inhibitors for the treatment of cancer:clinical im⁃ pact and mechanisms of response and resistance[J].An⁃ nu Rev Pathol,2021,16:223-249

    • [7] PENG L,WANG Y,LIU F,et al.Peripheral blood mark⁃ ers predictive of outcome and immune ⁃ related adverse events in advanced non ⁃ small cell lung cancer treated with PD ⁃ 1 inhibitors[J].Cancer Immunol Immunother,2020,69(9):1813-1822

    • [8] RUIZ ⁃ BAÑOBRE J,ARESES ⁃ MANRIQUE M C,MOS⁃ QUERA ⁃MARTÍNEZ J,et al.Evaluation of the lung im⁃ mune prognostic index in advanced non ⁃ small cell lung cancer patients under nivolumab monotherapy[J].Transl Lung Cancer Res,2019,8(6):1078-1085

    • [9] ZHUO M,CHEN H,ZHANG T,et al.The potential pre⁃ dictive value of circulating immune cell ratio and tumor marker in atezolizumab treated advanced non ⁃ small cell lung cancer patients[J].Cancer Biomark,2018,22(3):467-476

    • [10] GANDINI S,MASSI D,MANDALÀ M.PD⁃L1 expression in cancer patients receiving anti PD⁃1/PD⁃L1 antibodies:a systematic review and meta⁃analysis[J].Crit Rev Oncol Hematol,2016,100:88-98

    • [11] MOK T,WU Y L,KUDABA I,et al.Pembrolizumab ver⁃ sus chemotherapy for previously untreated,PD ⁃ L1 ⁃ ex⁃ pressing,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

    • [12] HELLMANN M D,CIULEANU T E,PLUZANSKI A,et al.Nivolumab plus ipilimumab in lung cancer with a high tumor mutational burden[J].N Engl J Med,2018,378(22):2093-2104

    • [13] MARABELLE A,FAKIH M,LOPEZ J,et al.Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab:prospective biomarker analysis of the multicohort,open ⁃ label,phase 2 KEYNOTE ⁃ 158 study[J].Lancet Oncol,2020,21(10):1353-1365

    • [14] ZHAO P,LI L,JIANG X,et al.Mismatch repair deficien⁃ cy/microsatellite instability ⁃ high as a predictor for anti ⁃ PD⁃1/PD ⁃L1 immunotherapy efficacy[J].J Hematol On⁃ col,2019,12(1):54-67

    • [15] SCOGNAMIGLIO G,De CHIARA A,Di BONITO M,et al.Variability in immunohistochemical detection of pro⁃ grammed death ligand 1(PD ⁃ L1)in cancer tissue types [J].Int J Mol Sci,2016,17(5):790-800

    • [16] HO W J,YARCHOAN M,HOPKINS A,et al.Association between pretreatment lymphocyte count and response to PD1 inhibitors in head and neck squamous cell carcino⁃ mas[J].J Immunother Cancer,2018,6(1):84-91

    • [17] AMERATUNGA M,CHÉNARD ⁃POIRIER M,MORENO C I,et al.Neutrophil⁃lymphocyte ratio kinetics in patients with advanced solid tumours on phase I trials of PD⁃1/PD⁃ L1 inhibitors[J].Eur J Cancer,2018,89:56-63

    • [18] HUEMER F,LANG D,WESTPHAL T,et al.Baseline ab⁃ solute lymphocyte count and ECOG performance score are associated with survival in advanced non ⁃ small cell lung cancer undergoing PD⁃1/PD⁃L1 blockade[J].J Clin Med,2019,8(7):1014-1030

    • [19] DIEM S,SCHMID S,KRAPF M,et al.Neutrophil⁃to⁃lym⁃ phocyte ratio(NLR)and platelet ⁃ to ⁃ lymphocyte ratio(PLR)as prognostic markers in patients with non ⁃ small cell lung cancer(NSCLC)treated with nivolumab[J].Lung Cancer,2017,111:176-181

    • [20] BUDER⁃BAKHAYA K,HASSEL J C.Biomarkers for clin⁃ ical benefit of immune checkpoint inhibitor treatment ⁃ a review from the melanoma perspective and beyond[J].Front Immunol,2018,9:1474-1490

    • [21] PAN Y,YU Y,WANG X,et al.Tumor⁃associated macro⁃ phages in tumor immunity[J].Front Immunol,2020,11:583-591

    • [22] LIN Y,XU J,LAN H.Tumor ⁃associated macrophages in tumor metastasis:biological roles and clinical therapeu⁃ tic applications[J].J Hematol Oncol,2019,12(1):76-91

    • [23] CARRETERO R,SEKTIOGLU I M,GARBI N,et al.Eo⁃ sinophils orchestrate cancer rejection by normalizing tu⁃ mor vessels and enhancing infiltration of CD8(+)T cells [J].Nat Immunol,2015,16(6):609-617

    • [24] GATAULT S,DELBEKE M,DRISS V,et al.IL⁃18 is in⁃ volved in eosinophil⁃mediated tumoricidal activity against a colon carcinoma cell line by upregulating LFA ⁃ 1 and ICAM⁃1[J].J Immunol,2015,195(5):2483-2492

    • [25] HOLUB K,BIETE A.Impact of systemic inflammation biomarkers on the survival outcomes of cervical cancer patients[J].Clin Transl Oncol,2019,21(7):836-844

    • [26] ROSNER S,KWONG E,SHOUSHTARI A N,et al.Pe⁃ ripheral blood clinical laboratory variables associated with outcomes following combination nivolumab and ipili⁃ mumab immunotherapy in melanoma[J].Cancer Med,2018,7(3):690-697

    • [27] REINA ⁃CAMPOS M,SCHARPING N E,GOLDRATH A W.CD8(+)T cell metabolism in infection and cancer[J].Nat Rev Immunol,2021,21(11):718-738

    • [28] LI F,LI C,CAI X,et al.The association between CD8+ tu⁃ mor ⁃infiltrating lymphocytes and the clinical outcome of cancer immunotherapy:a systematic review and meta ⁃ analysis[J].EClinicalMedicine,2021,41:2589-2600

    • [29] MIAO K,ZHANG X,WANG H,et al.Peripheral blood lymphocyte subsets predict the efficacy of immune check⁃ point inhibitors in non ⁃ small cell lung cancer[J].Front Immunol,2022,13:912-921

  • 通知关闭
    郑重声明