切换至 "中华医学电子期刊资源库"

中华临床医师杂志(电子版) ›› 2024, Vol. 18 ›› Issue (05) : 455 -461. doi: 10.3877/cma.j.issn.1674-0785.2024.05.004

临床研究

胃淋巴上皮瘤样癌三级淋巴结构特征及其与预后的相关性
闫战涛1, 王辉1, 周梓迪1, 史勇强1, 陈铜兵1,()   
  1. 1. 213000 江苏常州,常州市第一人民医院病理科
  • 收稿日期:2024-03-07 出版日期:2024-05-15
  • 通信作者: 陈铜兵
  • 基金资助:
    常州市应用基础研究计划(CJ20220202); 常州市卫健委青年项目(QN202304)

Tertiary lymphatic structure characteristics and their correlation with prognosis in patients with lymphoepithelioma-like gastric carcinoma

Zhantao Yan1, Hui Wang1, Zidi Zhou1, Yongqiang Shi1, Tongbing Chen1,()   

  1. 1. Department of Pathology, The First People's Hospital of Changzhou, Changzhou 213003, China
  • Received:2024-03-07 Published:2024-05-15
  • Corresponding author: Tongbing Chen
引用本文:

闫战涛, 王辉, 周梓迪, 史勇强, 陈铜兵. 胃淋巴上皮瘤样癌三级淋巴结构特征及其与预后的相关性[J]. 中华临床医师杂志(电子版), 2024, 18(05): 455-461.

Zhantao Yan, Hui Wang, Zidi Zhou, Yongqiang Shi, Tongbing Chen. Tertiary lymphatic structure characteristics and their correlation with prognosis in patients with lymphoepithelioma-like gastric carcinoma[J]. Chinese Journal of Clinicians(Electronic Edition), 2024, 18(05): 455-461.

目的

分析胃淋巴上皮瘤样癌(lymphoepithelioma-like gastric carcinoma,LELGC)中三级淋巴结构(tertiary lymphoid structure,TLS)特征及其与临床病理参数和预后的相关性。

方法

收集常州市第一人民医院2009年2月~2020年12月经病理确诊为LELGC的114例手术标本资料;然后手术标本切片进行病理组织学复核、TLS成熟度和密度分析;使用免疫组织化学染色方法检测肿瘤组织中淋巴细胞的CD3、CD4、CD8、CD20、CD21、PD-L1表达情况;使用Kaplan-Meirer和COX回归分析TLS对患者预后的影响。

结果

TLS主要分布在肿瘤间质内,集中在肿瘤侵袭性边缘区域;具有mTLS的肿瘤组织内以T淋巴细胞分布为优势,具有非mTLS的肿瘤组织内以B淋巴细胞分布为优势。mTLS组肿瘤平均直径(t=2.727,P=0.007)、脉管及神经侵犯阳性率(χ2=4.672,P=0.031)、淋巴结转移率(χ2=8.920,P=0.003)、临床分期Ⅲ/Ⅳ期发生率(χ2=15.360,P<0.001)均显著低于非mTLS组。TLS高密度组病理T分期(浸润深度)T3/T4发生率为80.0%(44/55),显著高于TLS低密度组的53.3%(35/59),比较差异具有统计学意义(χ2=5.721,P=0.017)。单因素COX回归分析显示,肿瘤直径(HR=1.177,P=0.011)、淋巴结(HR=11.873,P=0.016)、脉管神经侵犯(HR=4.430,P=0.007)、病理T分期(浸润深度,HR=13.706,P=0.011)、临床分期(HR=6.387,P<0.001)、TLS成熟度(HR=0.225,P=0.001)均与患者的预后显著相关;多因素COX回归分析显示,T分期(浸润深度,HR=9.765,P=0.028)、临床分期(HR=3.523,P=0.014)和TLS成熟度(HR=0.228,P=0.001)与患者的预后显著相关。Kaplan-Meirer生存分析显示,mTLS组患者平均生存期为(72.0±4.7)个月,显著高于非mTLS组的(33.7±5.5)个月,mTLS对总生存期有积极影响(χ2=14.063,P<0.001);TLS高密度组患者平均生存期为(65.4±5.0)个月,显著高于TLS低密度组的(51.8±6.5)个月,TLS高密度对总生存期有积极影响(χ2=5.482,P=0.019)。

结论

LELGC中TLS主要分布在肿瘤侵袭性边缘区域;具有mTLS的肿瘤组织以T淋巴细胞分布为优势,而具有非mTLS的肿瘤组织以B淋巴细胞分布为优势;LELGC中mTLS和高密度TLS可能会对患者的预后有积极影响,为患者的预后和治疗提供新的预测指标和靶点。

Objective

To investigate the characteristics of tertiary lymphoid structure (TLS) and their correlation with clinicopathological parameters and prognosis in patients with lymphoepithelioma-like gastric carcinoma (LELGC).

Methods

A total of 114 patients with LELGC were selected from Changzhou First People's Hospital from February 2009 to December 2020. Histopathological review and TLS maturity and density analysis were performed on 114 surgical specimens. Then, the expression of CD3, CD4, CD8, CD20, CD21, and PD-L1 in lymphocytes in tumor tissues was detected by immunohistochemical staining. Finally, Kaplan-Meirer survival analysis and COX regression analysis were used to examine the impact of TLS on patient prognosis.

Results

TLS was mainly distributed in the tumor stroma and concentrated in the invasive edge area of the tumor. T lymphocyte distribution was dominant in tumor tissues with mature TLS (mTLS), and B lymphocyte distribution was dominant in tumor tissues with non-mTLS. The average tumor diameter (t=2.727, P=0.007), positive rate of vascular and nerve invasion (χ2=4.672, P=0.031), lymph node metastasis rate (χ2=8.920, P=0.003), and incidence of clinical stage Ⅲ/Ⅳ (χ2=15.360, P<0.001) in the mTLS group were significantly lower than those of the non-mTLS group. The incidence of pathological T3/T4 stage (depth of invasion) in the TLS high-density group was 80.0% (44/55), which was significantly higher than that of the TLS low-density group (53.3%, 35/59) (χ2=5.721, P=0.017). Univariate COX regression analysis showed that tumor diameter (hazard ratio [HR]=1.177, P=0.011), lymph node metastasis (HR=11.873, P=0.016), vascular nerve invasion (HR=4.430, P=0.007), pathological T-stage (depth of invasion, HR=13.706, P=0.011), clinical stage (HR=6.387, P<0.001), and TLS maturity (HR=0.225, P=0.001) were significantly correlated with the prognosis of patients. Multivariate COX regression analysis showed that T stage (depth of invasion, HR=9.765, P=0.028), clinical stage (HR=3.523, P=0.014), and TLS maturity (HR=0.228, P=0.001) were significantly correlated with the prognosis of patients. Kaplan-Meirer survival analysis showed that the average survival time of patients in the mTLS group was (72.0±4.7) months, which was significantly longer than that of the non-mTLS group (33.7±5.5 months) (χ2=14.063, P<0.001). The average survival time of patients in the TLS high-density group was (65.4±5.0) months, which was significantly longer than that of the non-mTLS group (51.8±6.5 months) (χ2=5.482, P=0.019). mTLS and high density TLS had a positive effect on overall survival.

Conclusion

TLS in LELGC is mainly distributed in the invasive edge region of tumors. Tumor tissues with mTLS are dominated by T lymphocyte distribution, whereas tumor tissues with non-mTLs are dominated by B lymphocyte distribution. High-density and mature TLS in LELGC may have a positive impact on the prognosis of patients, providing new predictors and targets for the prognosis and treatment of patients.

图1 LELGC中TLS分布特征(200×)。图a为mTLS组织结构,HE染色;图b为CD20+ B淋巴细胞,免疫组化染色;图c为CD3+T淋巴细胞,免疫组化染色;图d为非mTLS组织结构,HE染色;图e为CD20+ B淋巴细胞,免疫组化染色;图f为CD3+T淋巴细胞,免疫组化染色
表1 每400倍视野CD20+细胞数和CD3+细胞数比较(
表2 2组TLS成熟度和密度与临床病理特征的相关性
表3 TLS与患者预后的关系
图2 患者的Kaplan-Meier 生存曲线。图a为mTLS组和非mTLS组;图b为TLS高密度组和TLS组低密度组
1
Sautès-Fridman C, Petitprez F, Calderaro J,et al. Tertiary lymphoid structures in the era of cancer immunotherapy [J]. Nat Rev Cancer, 2019, 19(6): 307-325.
2
Vanhersecke L, Brunet M, Guégan JP, et al. Mature tertiary lymphoid structures predict immune checkpoint inhibitor efficacy in solid tumors independently of PD-L1 expression [J]. Nat Cancer, 2021, 2(8): 794-802.
3
胡春芳,林华,陈艳佳,等. EB病毒相关胃癌地域分布特点及临床病理特征分析 [J]. 中华肿瘤杂志,2020,42(11): 943-948.
4
Pyo JS, Kim NY, Son BK, et al. Clinicopathological features and prognostic implication of gastric carcinoma with lymphoid stroma [J]. Gastroenterol Res Pract, 2020, 2020: 6628412.
5
Barmpoutis P, Di Capite M, Kayhanian H, et al. Tertiary lymphoid structures (TLS) identification and density assessment on H&E-stained digital slides of lung cance [J]. PLoS One, 2021, 16(9): e0256907.
6
N J, J T, Sl N, et al. Tertiary lymphoid structures and B lymphocytes in cancer prognosis and response to immunotherapies [J]. Oncoimmunology, 2021, 10(1): 1900508.
7
Salgado R, Denkert C, Demaria S, et al. The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer:recommendations by an International TILs Working Group 2014 [J]. Ann Onco, 2015, 26(2): 259-271.
8
Hissong E, Ramrattan G, Zhang P, et al. Gastric carcinomas with lymphoid stroma: an evaluation of the histopathologic and molecular features [J]. Am J Surg Pathol, 2018, 42(4): 453-462.
9
Zhao H, Wu L, Yan G, et al. Inflammation and tumor progression: signaling pathways and targeted intervention [J]. Signal Transduct Target Ther, 2021, 6(1): 263.
10
Meylan M, Petitprez F, Becht E, et al. Tertiary lymphoid structures generate and propagate anti-tumor antibody-producing plasma cells in renal cell cancer [J]. Immunity, 2022, 55(3): 527-541.e5.
11
Fridman WH, Meylan M, Petitprez F, et al. B cells and tertiary lymphoid structures as determinants of tumour immune contexture and clinical outcome [J]. Nat Rev Clin Oncol, 2022, 19(7): 441-457.
12
Schumacher TN, Thommen DS. Tertiary lymphoid structures in cancer [J]. Science, 2022, 375(6576): eabf9419.
13
Romero D. B cells and TLSs facilitate a response to ICI [J]. Nat Rev Clin Oncol, 2020, 17(4): 195.
14
Posch F, Silina K, Leibl S, et al. Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer [J].Oncoimmunology, 2017, 7(2): e1378844.
15
Calderaro J, Petitprez F, Becht E, et al. Intra-tumoral tertiary lymphoid structures are associated with a low risk of early recurrence of hepatocellular carcinoma [J]. J Hepatol, 2019, 70(1): 58-65.
16
Lauss M, Donia M, Svane IM, et al. B cells and tertiary lymphoid structures: friends or foes in cancer immunotherapy? [J]. Clin Cancer Res, 2022, 28(9): 1751-1758.
17
Calderaro J, Petitprez F, Becht E, et al.Intra-tumoral tertiary lymphoid structures are associated with a low risk of early recurrence of hepatocellular carcinoma [J]. J Hepatol, 2019, 70(1): 58-65.
18
Posch F, Silina K, Leibl S, et al. Maturation of tertiary lymphoid structures and recurrence of stage II and III colorectal cancer [J]. Oncoimmunology, 2017, 7(2): e1378844.
19
黄东东, 邬万新, 陈彩萍, 等. 乳腺导管原位癌三级淋巴结构的临床病理特征分析及其与肿瘤进展的关系 [J]. 临床与实验病理学杂志, 2022, 38(6): 738-741, 745.
20
He W, Zhang D, Liu H, et al. The high level of tertiary lymphoid structure is correlated with superior survival in patients with advanced gastric cancer [J]. Front Oncol, 2020, 10: 980.
21
Dong C. Cytokine regulation and function in T cells [J]. Annu Rev Immunol, 2021, 39: 51-76.
[1] 牛海刚, 郭文科. 三阴性乳腺癌组织中双特异性磷酸酶14与核受体相互作用蛋白1的表达及预后价值[J]. 中华乳腺病杂志(电子版), 2024, 18(04): 199-205.
[2] 丁丁, 杨云川, 马翔, 马中正, 霍俊一, 周磊. 术前C-反应蛋白-白蛋白-淋巴细胞比值在肝细胞癌预后中的价值评估[J]. 中华普通外科学文献(电子版), 2024, 18(04): 261-265.
[3] 李怡泉, 谢宇斌, 胡宏, 张燕茹, 陈图锋. 基于生物信息学分析HDAC8在结肠癌中的临床意义及其与免疫浸润的关系[J]. 中华普通外科学文献(电子版), 2024, 18(04): 275-281.
[4] 孙建娜, 孔令军, 任崇禧, 穆坤, 王晓蕊. 266例首诊Ⅳ期乳腺癌手术患者预后分析[J]. 中华普外科手术学杂志(电子版), 2024, 18(05): 502-505.
[5] 袁庆港, 刘理想, 张亮, 周世振, 高波, 丁超, 管文贤. 尿素-肌酐比值(UCR)可预测结直肠癌患者术后的长期预后[J]. 中华普外科手术学杂志(电子版), 2024, 18(05): 506-509.
[6] 黄福, 王黔, 金相任, 唐云川. VEGFR2、miR-27a-5p在胃癌组织中的表达与临床病理参数及预后的关系研究[J]. 中华普外科手术学杂志(电子版), 2024, 18(05): 558-561.
[7] 蔡大明, 陆晓峰, 王行舟, 王萌, 刘颂, 夏雪峰, 沈晓菲, 杜峻峰, 管文贤. 三级淋巴结构在胃神经内分泌瘤中的预后价值及预后预测模型构建[J]. 中华普外科手术学杂志(电子版), 2024, 18(04): 401-405.
[8] 达热拜·热达提, 刘林, 赵为民, 孟涛, 雷程, 金博, 毕建军, 李新宇, 王海江. 中低位直肠癌新辅助放化疗后侧方淋巴结清扫术的临床观察[J]. 中华普外科手术学杂志(电子版), 2024, 18(04): 415-418.
[9] 李勇, 彭天明, 王倩倩, 陈育纯, 蒲小勇, 刘久敏. 基于失巢凋亡相关基因的膀胱癌预后模型构建及分析[J]. 中华腔镜泌尿外科杂志(电子版), 2024, 18(04): 331-339.
[10] 钱承博, 殷虎明, 邱峰, 侯建全, 黄玉华, 魏雪栋. 高龄患者行腹腔镜膀胱根治W形回肠新膀胱术的临床价值与风险评估[J]. 中华腔镜泌尿外科杂志(电子版), 2024, 18(04): 346-352.
[11] 张斌, 孙代宇, 胡昕, 韩菲, 李久明, 李功雨, 吴伟力, 冯宝富, 彭国辉. 评分系统预测不同经验手术者输尿管软镜术后结石清除率准确性的比较研究[J]. 中华腔镜泌尿外科杂志(电子版), 2024, 18(04): 353-360.
[12] 刘中文, 刘畅, 高洋, 刘东, 林世庆, 杨建华, 赵福义. 尿液microRNA-326与腹腔镜根治性膀胱切除术治疗膀胱癌患者预后的相关性研究[J]. 中华腔镜泌尿外科杂志(电子版), 2024, 18(04): 386-391.
[13] 杜鑫, 刘霞霞, 张恬波, 张夏林, 杨林花, 张睿娟. AHNAK基因高表达与老年急性髓系白血病患者预后不良相关[J]. 中华细胞与干细胞杂志(电子版), 2024, 14(04): 204-211.
[14] 杨秀君, 崔梦莹, 刘水, 盛基尧, 张丹. 基于SEER数据库胰头部胰腺神经内分泌癌患者预后列线图构建与验证[J]. 中华肝脏外科手术学电子杂志, 2024, 13(04): 520-525.
[15] 曾谣, 谢琴, 陈显育, 王平根, 毛玲秋, 何丹玲, 杜飞, 郑希彦, 何函樨. CDC42EP2基因与肝癌预后、免疫细胞浸润关系及其对细胞迁移侵袭的影响[J]. 中华肝脏外科手术学电子杂志, 2024, 13(03): 363-369.
阅读次数
全文


摘要