综述

胰腺导管腺癌病人循环免疫及肿瘤微环境中T细胞亚群异常

  • 苏秉蔚 综述 ,
  • 沈柏用 审校
展开
  • 1.上海交通大学医学院,上海 200025
    2.上海交通大学医学院附属瑞金医院普外科 胰腺疾病诊疗中心,上海交通大学医学院胰腺疾病研究所,癌基因与相关基因国家重点实验室,上海交通大学转化医学研究院,上海 200025
沈柏用,E-mail: shenby@shsmu.edu.cn

收稿日期: 2024-09-05

  网络出版日期: 2025-07-07

Abnormalities in T cell subsets in the circulating immunity and tumor microenvironment in patients with pancreatic ductal adenocarcinoma

  • SU Bingwei ,
  • SHEN Baiyong
Expand
  • 1. Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
    2. Department of General Surgery, Pancreatic Di-sease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Research Institute of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine; State Key Laboratory of Oncogenes and Related Genes (Shanghai); Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai 200025, China

Received date: 2024-09-05

  Online published: 2025-07-07

摘要

胰腺导管腺癌(PDAC)是一种高度恶性的消化系统实体肿瘤,能影响机体免疫系统功能。PDAC病人外周循环和肿瘤微环境中的免疫细胞在分布、百分比、功能、活性等特征上均呈现出异常表现。T细胞是参与细胞免疫和发挥抗肿瘤效应的主要淋巴细胞,根据功能和分化抗原不同,可以分为CD4+、CD8+、CD4+ CD25+等亚群。PDAC病人循环免疫及肿瘤微环境中T细胞亚群水平的异常可能是促进肿瘤发生发展的重要因素。

本文引用格式

苏秉蔚 综述 , 沈柏用 审校 . 胰腺导管腺癌病人循环免疫及肿瘤微环境中T细胞亚群异常[J]. 外科理论与实践, 2025 , 30(2) : 171 -175 . DOI: 10.16139/j.1007-9610.2025.02.13

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant solid tumor of digestive system, which can affect the immune system. The abnormalities of distribution, percentage, function, and activity of immune cells in both peripheral circulation and tumor microenvironment (TME) are detected in patients with PDAC. T cells are the main lymphocytes that participate in cell-mediated immunity and exert anti-tumor effects. According to the function and the cluster of differentiation, T cells can be divided into different subsets, such as CD4+, CD8+, CD4+ CD25+, etc. The abnormalities of T cell subsets in the circulating immunity and TME of patients with PDAC may play an important role in cancer progress.

参考文献

[1] BRAY F, LAVERSANNE M, SUNG H, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries[J]. CA Cancer J Clin, 2024, 74(3):229-263.
[2] 国家卫生健康委办公厅. 胰腺癌诊疗指南(2022年版)[J]. 临床肝胆病杂志, 2022, 38(5):1006-1015.
  General Office of the National Health Commission of China. Standard for diagnosis and treatment of pancreatic cancer (2022 edition)[J]. J Clin Hepatol, 2022, 38(5):1006-1015.
[3] SIEGEL R L, GIAQUINTO A N, JEMAL A. Cancer statistics, 2024[J]. CA Cancer J Clin, 2024, 74(1):12-49.
[4] GORAL V. Pancreatic cancer: pathogenesis and diagnosis[J]. Asian Pac J Cancer Prev, 2015, 16(14):5619-5624.
[5] BALACHANDRAN V P, ?UKSZA M, ZHAO J N, et al. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer[J]. Nature, 2017, 551(7681):512-516.
[6] YING H, YAO W. The prospects of immunotherapy in pancreatic cancer[M]// Pancreat Cancer Multidiscip Approach. Cham:Springer,2022:269-281.
[7] AJINA R, WEINER L M. T-Cell immunity in pancreatic cancer[J]. Pancreas, 2020, 49(8):1014-1023.
[8] DALEY D, ZAMBIRINIS C P, SEIFERT L, et al. γδ T cells support pancreatic oncogenesis by restraining αβ T cell activation[J]. Cell, 2016, 166(6):1485-1499.e15.
[9] LAFONT V, SANCHEZ F, LAPREVOTTE E, et al. Plasticity of γδ T cells: impact on the anti-tumor response[J]. Front Immunol,2014, 5:622
[10] MCALLISTER F, BAILEY J M, ALSINA J, et al. Oncogenic Kras activates a hematopoietic-to-epithelial IL-17 signaling axis in preinvasive pancreatic neoplasia[J]. Cancer cell, 2014, 25(5):621-637.
[11] LI J, MORESCO P, FEARON D T. Intratumoral NKT cell accumulation promotes antitumor immunity in pancreatic cancer[J]. Proc Natl Acad Sci USA, 2024, 121(29):e2403917121.
[12] JANAKIRAM N B, MOHAMMED A, BRYANT T, et al. Loss of natural killer T cells promotes pancreatic cancer in LSL-KrasG12D/+ mice[J]. Immunology, 2017, 152(1):36-51.
[13] XU Y F, LU Y, CHENG H, et al. Abnormal distribution of peripheral lymphocyte subsets induced by PDAC modulates overall survival[J]. Pancreatology, 2014, 14(4):295-301.
[14] LIYANAGE U K, MOORE T T, JOO H G, et al. Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma[J]. J Immunol, 2002, 169(5):2756-2761.
[15] O'NEILL R E, CAO X. Co-stimulatory and co-inhibitory pathways in cancer immunotherapy[J]. Adv Cancer Res, 2019,143:145-194.
[16] LIU C, CHENG H, LUO G, et al. Circulating regulatory T cell subsets predict overall survival of patients with unresectable pancreatic cancer[J]. Int J Oncol, 2017, 51(2):686-694.
[17] YAMAMOTO T, YANAGIMOTO H, SATOI S, et al. Circulating CD4+CD25+ regulatory T cells in patients with pancreatic cancer[J]. Pancreas, 2012, 41(3):409-415.
[18] STROMNES I M, HULBERT A, PIERCE R H, et al. T-cell localization, activation, and clonal expansion in human pancreatic ductal adenocarcinoma[J]. Cancer Immunol Res, 2017, 5(11):978-991.
[19] BAILEY P, CHANG D K, NONES K, et al. Genomic analyses identify molecular subtypes of pancreatic cancer[J]. Nature, 2016, 531(7592):47-52.
[20] PENG J, SUN B F, CHEN C Y, et al. Single-cell RNA-seq highlights intra-tumoral heterogeneity and malignant progression in pancreatic ductal adenocarcinoma[J]. Cell Res, 2019, 29(9):725-738.
[21] BALLI D, RECH A J, STANGER B Z, et al. Immune cytolytic activity stratifies molecular subsets of human pancreatic cancer[J]. Clin Cancer Res, 2017, 23(12):3129-3138.
[22] RYSCHICH E, N?TZEL T, HINZ U, et al. Control of T-cell-mediated immune response by HLA class Ⅰ in human pancreatic carcinoma[J]. Clin Cancer Res, 2005, 11(2 Pt 1):498-504.
[23] TANG Y, XU X, GUO S, et al. An increased abundance of tumor-infiltrating regulatory T cells is correlated with the progression and prognosis of pancreatic ductal adenocarcinoma[J]. PLoS one, 2014, 9(3):e91551.
[24] CARSTENS J L, CORREA DE SAMPAIO P, YANG D, et al. Spatial computation of intratumoral T cells correlates with survival of patients with pancreatic cancer[J]. Nat Commun,2017, 8:15095
[25] YOUSUF S, QIU M, VOITH VON VOITHENBERG L, et al. Spatially resolved multi-omics single-cell analyses inform mechanisms of immune dysfunction in pancreatic cancer[J]. Gastroenterology, 2023, 165(4):891-908.e14.
[26] LIU W, PUTNAM A L, XU-YU Z, et al. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells[J]. J Exp Med, 2006, 203(7):1701-1711.
[27] SAHAI E, ASTSATUROV I, CUKIERMAN E. A framework for advancing our understanding of cancer-associa-ted fibroblasts[J]. Nat Rev Cancer, 2020, 20(3):174-186.
[28] ?HLUND D, HANDLY-SANTANA A, BIFFI G, et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer[J]. J Exp Med, 2017, 214(3):579-596.
[29] JENKINS L, JUNGWIRTH U, AVGUSTINOVA A. Cancer-associated fibroblasts suppress CD8+ T-cell infiltration and confer resistance to immune-checkpoint blockade[J]. Cancer Res, 2022, 82(16):2904-2917.
[30] TAN M C, GOEDEGEBUURE P S, BELT B A, et al. Disruption of CCR5-dependent homing of regulatory T cells inhibits tumor growth in a murine model of pancreatic cancer[J]. J Immunol, 2009, 182(3):1746-1755.
[31] PAN Y, LU F, FEI Q, et al. Single-cell RNA sequencing reveals compartmental remodeling of tumor-infiltrating immune cells induced by anti-CD47 targeting in pancrea-tic cancer[J]. J Hematol Oncol, 2019, 27, 12(1):124.
[32] POSCHKE I, FARYNA M, BERGMANN F, et al. Identification of a tumor-reactive T-cell repertoire in the immune infiltrate of patients with resectable pancreatic ductal adenocarcinoma[J]. Oncoimmunology, 2016, 5(12):e1240859.
[33] ZHANG Y, LAZARUS J, STEELE N G, et al. Regulatory T-cell depletion alters the tumor microenvironment and accelerates pancreatic carcinogenesis[J]. Cancer Discov, 2020, 10(3):422-439.
[34] DAS S, SHAPIRO B. Tumor cell-derived IL1β promotes desmoplasia and immune suppression in pancreatic cancer[J]. Cancer Res, 2020, 80(5):1088-1101.
[35] LEINWAND J, MILLER G. Regulation and modulation of antitumor immunity in pancreatic cancer[J]. Nat Immunol, 2020, 21(10):1152-1159.
[36] FAN J Q, WANG M F, CHEN H L, et al. Current advances and outlooks in immunotherapy for pancreatic ductal adenocarcinoma[J]. Mol Cancer, 2020, 19(1):32.
[37] SHA H, TONG F, NI J, et al. First-line penpulimab (an anti-PD1 antibody) and anlotinib (an angiogenesis inhibitor) with nab-paclitaxel/gemcitabine (PAAG) in metastatic pancreatic cancer: a prospective, multicentre, biomolecular exploratory, phase Ⅱ trial[J]. Signal Transduct Target Ther, 2024, 9(1):143.
[38] PARIKH A R, SZABOLCS A, ALLEN J N, et al. Radiation therapy enhances immunotherapy response in microsatellite stable colorectal and pancreatic adenocarcinoma in a phase Ⅱ trial[J]. Nat Cancer, 2021, 2(11):1124-1135.
[39] ZHANG H, XU W, ZHU H, et al. Overcoming the limitations of immunotherapy in pancreatic ductal adenocarcinoma: combining radiotherapy and metabolic targeting therapy[J]. J Cancer, 2024, 15(7):2003-2023.
[40] PANT S, WAINBERG Z A, WEEKES C D, et al. Lymph-node-targeted, mKRAS-specific amphiphile vaccine in pancreatic and colorectal cancer: the phase 1 AMPLIFY-201 trial[J]. Nat Med, 2024, 30(2):531-542.
[41] ROJAS L A, SETHNA Z, SOARES K C, et al. Persona-lized RNA neoantigen vaccines stimulate T cells in pancreatic cancer[J]. Nature, 2023, 618(7963):144-150.
[42] SETHNA Z, GUASP P, REICHE C, et al. RNA neoantigen vaccines prime long-lived CD8(+) T cells in pancreatic cancer[J]. Nature, 2025, 639(8056):1042-1051.
[43] LOPEZ J, POWLES T, BRAITEH F, et al. Autogene cevumeran with or without atezolizumab in advanced solid tumors: a phase 1 trial[J]. Nat Med, 2025, 31(1):152-164.
[44] MUSHER B L, ROWINSKY E K, SMAGLO B G, et al. LOAd703, an oncolytic virus-based immunostimulatory gene therapy, combined with chemotherapy for unresectable or metastatic pancreatic cancer (LOKON001): results from arm 1 of a non-randomised, single-centre, phase 1/2 study[J]. Lancet Oncol, 2024, 25(4):488-500.
文章导航

/