消化道肿瘤免疫抑制性微环境研究进展
doi:
Research Progress in Immunosuppressive Tumor Microenvironment of Gastrointestinal Cancer
-
摘要: 消化道肿瘤(gastrointestinal cancer, GI)是我国常见高发的恶性肿瘤,随着其发病率的日益升高和愈发年轻化的趋势,新的治疗技术也在不断发展和创新,其中免疫疗法正在引领消化道肿瘤治疗新时代。然而,免疫抑制性肿瘤微环境(tumor microenvironment, TME)的复杂性和多样性给消化道实体瘤的免疫治疗增添了重重阻碍。本综述以消化道实体肿瘤为切入点,回顾了影响其免疫抑制性微环境形成的主要因素,概述了靶向免疫抑制微环境治疗的策略,分析了各类免疫联合疗法的协同机制,总结了消化道恶性肿瘤患者免疫治疗的最新进展和未来方向,旨在为消化道实体瘤的免疫治疗提供新思路。
-
关键词:
- 消化道肿瘤 /
- 免疫抑制性肿瘤微环境 /
- 免疫疗法 /
- 肿瘤治疗
Abstract: Gastrointestinal (GI) cancer, a common malignant tumor with a high incidence in China, is showing a trend of rising incidence and is afflicting increasingly younger patients. Meanwhile, there have been constant development and innovations in new therapeutic technologies, among which, immunotherapy is now leading in a new era in the treatment of GI cancer. However, the complexity and diversity of immunosuppressive tumor microenvironment (TME) bring many obstacles to the immunotherapy of solid tumors in the GI tract. In this paper, focusing on solid tumors in the GI tract, we reviewed the main factors affecting the formation of immunosuppressive TME, and summarized strategies for targeted immunosuppressive TME-based therapies. Moreover, we analyzed the synergistic mechanism of various combination immunotherapies and reported on the latest progress in and future direction of immunotherapy for GI cancer, intending to provide new perspectives for treating solid tumors in the GI tract with immumotherapy. -
koko体育app
图 1 消化道肿瘤免疫抑制性微环境的组成模式图
Figure 1. 🥂 A schematic illustration of the immunosuppressive tumor microenvironment in gastrointestinal cancer
MDSCs: Myeloid-derived suppressor cells; VEGF: Vascular endothelial growth factor; CTL: Cytotoxic T lymphocytes; Th1: Effector T helper 1 cells; Th17: Effector T helper 17 cells; Tregs: Regulatory T cells; DCs: Dendritic cells; TAMs: Tumor-associated macrophages; TANs: Tumour-associated neutrophils; CSCs: Cancer stem cells; CTLA4: Cytotoxic T lymphocyte antigen 4; TIM3: T cell immunoglobulin and mucin domain-containin protein 3; PD1: Programmed cell death protein 1; IDO: Indoleamine 2,3-dioxygenase; IL: Interleukin; ROS: Reactive oxygen species; TGF-β: Transforming growth factor-β; CXCL12: C-X-C motif chemokine ligand 12; CXCR4: C-X-C motif chemokine receptor 4.图 2 消化道肿瘤免疫抑制性微环境的腺苷通路图
Figure 2. ♎ Adenosinergic pathway in the immunosuppressive tumor microenvironment in gastrointestinal cancer
FOXP3: Forkhead box P3; LAG3: Lymphocyte activation gene 3; IFN-γ: Interferon-γ; TNF: Tumour necrosis factor; TCR: T cell receptor; ATP: Adenosine triphosphate; PKA: Protein kinase A; cAMP: Cyclic AMP; HIF1A: Hypoxia-inducible factor 1A; NF-κB: Nuclear factor-κB; MHC Ⅱ: Major histocompatibility complex class Ⅱ; TLR4: Toll-like receptor 4.图 3 靶向消化道肿瘤免疫抑制微环境的治疗策略模式图
Figure 3. ๊ Immunosuppressive TME-based therapeutical strategies in gastrointestinal cancer
HDAC: Histone deacetylase; TRAILR2: TNF-related apoptosis-inducing ligand receptor 2; TLR9: Toll like receptor 9. -
[1] SHALAPOUR S, KARIN M. Cruel to be kind: Epithelial, microbial, and immune cell interactions in gastrointestinal cancers. Annu Rev Immunol,2020,38: 649–671. doi: [2] SUNG H, FERLAY J, SIEGEL R L, et al. Global cancer statistics 2020: Globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin,2021,71(3): 209–249. doi: [3] QUANTE M, VARGA J, WANG T C, et al. The gastrointestinal tumor microenvironment. Gastroenterology,2013,145(1): 63–78. doi: [4] JOYCE J A, FEARON D T. T cell exclusion, immune privilege, and the tumor microenvironment. Science,2015,348(6230): 74–80. doi: [5] BARKER H E, PAGET J T, KHAN A A, et al. The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat Rev Cancer,2015,15(7): 409–425. doi: [6] HANAHAN D, WEINBERG R A. Hallmarks of cancer: The next generation. Cell,2011,144(5): 646–674. doi: [7] ZHANG Y, XU J, ZHANG N, et al. Targeting the tumour immune microenvironment for cancer therapy in human gastrointestinal malignancies. Cancer Lett,2019,458: 123–135. doi: [8] DAVERN M, DONLON N E, POWER R, et al. The tumour immune microenvironment in oesophageal cancer. Br J Cancer,2021,125(4): 479–494. doi: [9] BINNEWIES M, ROBERTS E W, KERSTEN K, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med,2018,24(5): 541–550. doi: [10] NAGARSHETH N, WICHA M S, ZOU W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat Rev Immunol,2017,17(9): 559–572. doi: [11] OLLAURI-IBÁÑEZ C, AYUSO-ÍÑIGO B, PERICACHO M. Hot and cold tumors: is endoglin (cd105) a potential target for vessel normalization? Cancers (Basel), 2021, 13(7): 1552[2022-12-20]. . [12] GAO H, MA J, CHENG Y, et al. Exosomal transfer of macrophage-derived miR-223 confers doxorubicin resistance in gastric cancer. Onco Targets Ther,2020,13: 12169–12179. doi: [13] SAHAI E, ASTSATUROV I, CUKIERMAN E, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer,2020,20(3): 174–186. doi: [14] BIFFI G, TUVESON D A. Diversity and biology of cancer-associated fibroblasts. Physiol Rev,2021,101: 147–176. doi: [15] MORTEZAEE K. Enriched cancer stem cells, dense stroma, and cold immunity: Interrelated events in pancreatic cancer. J Biochem Mol Toxicol, 2021, 35(4): e22708[2022-12-20]. . [16] ERIN N, GRAHOVAC J, BROZOVIC A, et al. Tumor microenvironment and epithelial mesenchymal transition as targets to overcome tumor multidrug resistance. Drug Resist Updat, 2020, 53: 100715[2022-12-20]. . [17] LIN C C, LIAO T T, YANG M H. Immune adaptation of colorectal cancer stem cells and their interaction with the tumor microenvironment. Front Oncol, 2020, 10: 588542[2022-12-20]. . [18] LIAO T T, LIN C C, JIANG J K, et al. Harnessing stemness and PD-L1 expression by AT-rich interaction domain-containing protein 3B in colorectal cancer. Theranostics,2020,10(14): 6095–6112. doi: [19] HUI E, CHEUNG J, ZHU J, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science,2017,355(6332): 1428–1433. doi: [20] ZHAO H K, WU L, YAN G F, et al. Inflammation and tumor progression: Signaling pathways and targeted intervention, Signal Transduct Target Ther, 2021, 6(1): 263[2022-12-20]. . [21] RAHMA O E, HODI F S. The intersection between tumor angiogenesis and immune suppression. Clin Cancer Res,2019,25(18): 5449–5457. doi: [22] FLEMING C, MORRISSEY S, CAI Y, et al. γδ T cells: Unexpected regulators of cancer development and progression. Trends Cancer,2017,3(8): 561–570. doi: [23] HUANG T X, TAN X Y, HUANG H S, et al. Targeting cancer-associated fibroblast-secreted WNT2 restores dendritic cell-mediated antitumour immunity. Gut, 2021: gutjnl-2020-322924[2022-12-20]. . [24] GONÇALVES-RIBEIRO S, DÍAZ-MAROTO N G, BERDIEL-ACER M, et al. Carcinoma-associated fibroblasts affect sensitivity to oxaliplatin and 5-FU in colorectal cancer cells. Oncotarget,2016,7(37): 59766–59780. doi: [25] NAKAMURA K, YAGUCHI T, OHMURA G, et al. Involvement of local renin-angiotensin system in immunosuppression of tumor microenvironment. Cancer Sci,2018,109(1): 54–64. doi: [26] HAM I H, OH H J, JIN H, et al. Targeting interleukin-6 as a strategy to overcome stroma-induced resistance to chemotherapy in gastric cancer. Mol Cancer, 2019, 18(1): 68[2022-12-20]. . [27] 赵昆, 时荣臣, 缪洪明. 肿瘤相关巨噬细胞的脂质代谢重编程. koko体育app 学报(医学版),2021,52(1): 45–49. doi: [28] XIANG X N, WANG J G, LU D, et al. Targeting tumor-associated macrophages to synergize tumor immunotherapy. Signal Transduct Target Ther, 2021, 6(1): 75[2022-12-20]. . [29] LI X, YAO W, YUAN Y, et al. Targeting of tumour-infiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy against hepatocellular carcinoma. Gut,2017,66(1): 157–167. doi: [30] ALDINUCCI D, CASAGRANDE N. Inhibition of the CCL5/CCR5 axis against the progression of gastric cancer. Int J Mol Sci, 2018, 19(5): 1477[2022-12-20]. . [31] SUAREZ-CARMONA M, CHAORENTONG P, KATHER J N, et al. CCR5 status and metastatic progression in colorectal cancer. Oncoimmunology, 2019, 8(9): e1626193[2022-12-20]. . [32] OKUGAWA Y, TOIYAMA Y, ICHIKAWA T, et al. Colony-stimulating factor-1 and colony-stimulating factor-1 receptor co-expression is associated with disease progression in gastric cancer. Int J Oncol,2018,53(2): 737–749. doi: [33] HIAM-GALVEZ K J, ALLEN B M, SPITZER M H. Systemic immunity in cancer. Nat Rev Cancer,2021,21(6): 345–359. doi: [34] LIMAGNE E, EUVRARD R, THIBAUDIN M, et al. Accumulation of mdsc and Th17 cells in patients with metastatic colorectal cancer predicts the efficacy of a folfox-bevacizumab drug treatment regimen. Cancer Res,2016,76(18): 5241–5252. doi: [35] ZHANG X, SHEN L, LIU Q, et al. Inhibiting PI3 kinase-γ in both myeloid and plasma cells remodels the suppressive tumor microenvironment in desmoplastic tumors. J Control Release,2019,309: 173–180. doi: [36] ZAPPASODI R, SERGANOVA I, COHEN I J, et al. CTLA-4 blockade drives loss of Treg stability in glycolysis-low tumours. Nature,2021,591(7851): 652–658. doi: [37] KANIKARLA P, HAYMAKER C, PARRA E R, et al. Pilot clinical trial of perioperative durvalumab and tremelimumab in the treatment of resectable colorectal cancer liver metastases. Clin Cancer Res,2021,27(11): 3039–3049. doi: [38] ZHANG L, SUN L, ZHOU Y, et al. Association of survival and immune-related adverse events with anti-PD-1/PD-l1 and anti-CTLA-4 inhibitors, alone or their combination for the treatment of cancer: A systematic review and meta-analysis of 13 clinical trials. Front Oncol, 2021, 11: 575457[2022-12-20]. . [39] CHAUVIN J M, ZAROUR H M. TIGIT in cancer immunotherapy. J Immunother Cancer, 2020, 8(2): e000957[2022-12-20]. . [40] QI Y, CHEN L, LIU Q, et al. Research progress concerning dual blockade of lymphocyte-activation gene 3 and programmed death-1/programmed death-1 ligand-1 blockade in cancer immunotherapy: Preclinical and clinical evidence of this potentially more effective immunotherapy strategy. Front Immunol, 2021, 11: 563258[2022-12-20]. . [41] ABDUL-LATIF M, TOWNSEND K, DEARMAN C, et al. Immunotherapy in gastrointestinal cancer: The current scenario and future perspectives. Cancer Treat Rev, 2020, 88: 102030[2022-12-20]. . [42] CIOFFI M, TRABULO S, HIDALGO M, et al. Inhibition of CD47 effectively targets pancreatic cancer stem cells via dual mechanisms. Clin Cancer Res,2015,21(10): 2325–2337. doi: [43] WANG Y, CHEN M, WU Z, et al. CD133-directed CAR T cells for advanced metastasis malignancies: A phase I trial. Oncoimmunology, 2018, 7(7): e1440169[2022-12-20]. . [44] LIU Z, ZHAO Q, ZHENG Z, et al. Vascular normalization in immunotherapy: a promising mechanisms combined with radiotherapy. Biomed Pharmacother, 2021, 139: 111607[2022-12-20]. . [45] O’HARA M H, O’REILLY E M, ROSEMARIE M, et al. Abstract CT004: A Phase Ib study of CD40 agonistic monoclonal antibody APX005M together with gemcitabine (Gem) and nab-paclitaxel (NP) with or without nivolumab (Nivo) in untreated metastatic ductal pancreatic adenocarci- noma (PDAC) patients. Cancer Res,2019,79(13 Suppl): CT004. doi: [46] FUKUMURA D, KLOEPPER J, AMOOZGAR Z, et al. Enhancing cancer immunotherapy using antiangiogenics: Opportunities and challenges. Nat Rev Clin Oncol,2018,15(5): 325–340. doi: [47] CLARA J A, MONGE C, YANG Y, et al. Targeting signalling pathways and the immune microenvironment of cancer stem cells—A clinical update. Nat Rev Clin Oncol,2020,17(4): 204–232. doi: [48] GANDHI L, RODRÍGUEZ-ABREU D, GADGEEL S, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med,2018,378(22): 2078–2092. doi: -