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) ›› 2021, Vol. 15 ›› Issue (09) : 712 -715. doi: 10.3877/cma.j.issn.1674-0785.2021.09.013

综述

结直肠癌免疫检查点抑制剂研究进展
胡冬至1, 孔大陆1,()   
  1. 1. 300060 天津,天津医科大学肿瘤医院结直肠肿瘤科 国家肿瘤临床医学研究中心 天津市“肿瘤防治”重点实验室 天津市恶性肿瘤临床医学研究中心
  • 收稿日期:2021-07-18 出版日期:2021-09-15
  • 通信作者: 孔大陆

Research progress of immune checkpoint inhibitors for colorectal cancer

Dongzhi Hu1, Dalu Kong1,()   

  1. 1. Department of Colorectal Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
  • Received:2021-07-18 Published:2021-09-15
  • Corresponding author: Dalu Kong
引用本文:

胡冬至, 孔大陆. 结直肠癌免疫检查点抑制剂研究进展[J]. 中华临床医师杂志(电子版), 2021, 15(09): 712-715.

Dongzhi Hu, Dalu Kong. Research progress of immune checkpoint inhibitors for colorectal cancer[J]. Chinese Journal of Clinicians(Electronic Edition), 2021, 15(09): 712-715.

近十年来,结直肠癌免疫治疗取得了长足的进展,尤其对于错配修复缺陷(dMMR)/高微卫星不稳定性(MSI-H)表型的结直肠癌,免疫治疗具有持续的治疗反应,并取得了显著的临床疗效。但是大多数转移性结直肠癌为非错配修复缺陷(pMMR)或微卫星稳定(MSS)状态,这部分患者从免疫检查点抑制剂(ICIs)中获益较少。随着ICIs在结直肠癌中临床试验的广泛开展,免疫治疗因确定的疗效,逐步扩大了适应证,对于dMMR/MSI-H的晚期结直肠癌患者,已成为标准一线治疗方案。而对于pMMR/MSS患者,ICIs通过联合化疗及靶向治疗,也显示出了潜在的治疗效果。本文主要就ICIs在结直肠癌中的应用展开讨论,分析肿瘤与免疫系统之间的相互关系,并概述结直肠癌的免疫学特征及分类以及ICIs在结直肠癌治疗中的最新进展。

In recent ten years, immunotherapy for colorectal cancer has made great progress. Especially for colorectal cancer (CRC) with deficient mismatch repair (dMMR) / microsatellite instability high (MSI-H) phenotype, immunotherapy has sustained response and achieved significant clinical efficacy. However, most of the metastatic colorectal cancer patients are in the state of mismatch repair defect (pMMR) or microsatellite stable (MSS). These patients benefit less from immune checkpoint inhibitors (ICIs). With the extensive development of ICIS in colorectal cancer clinical trials, immunotherapy has gradually expanded the indications because of its definite efficacy. For patients with advanced colorectal cancer with dMMR/MSI-H, it has become the standard first-line treatment. For pMMR/MSS patients, ICIs also showed potential therapeutic effect through combined chemotherapy and targeted therapy. In this review, we mainly discuss the application of ICIs in colorectal cancer, explain the relationship between tumor and immune system, and summarize the immunological characteristics and classification of CRC, as well as the latest progress of ICIs in the treatment of CRC.

1
Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries [J].CA Cancer J Clin, 2018, 68(6): 394-424.
2
Le DT, Kim TW, Van Cutsem E, et al. Phase Ⅱ Open-Label Study of Pembrolizumab in treatment-refractory, microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: KEYNOTE-164 [J]. J Clin Oncol, 2020, 38(1): 11-19.
3
FDA. FDA approves first cancer treatment for any solid tumor with a specific genetic feature [EB/OL]. 2017-05-23.

URL    
4
Overman MJ, McDermott R, Leach JL, et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study [J]. Lancet Oncol, 2017, 18(9): 1182-1191.
5
FDA. FDA grants accelerated approval to ipilimumab for MSI-H or dMMR metastatic colorectal cancer[EB/OL]. 2018-7-11.

URL    
6
FDA. FDA grants nivolumab accelerated approval for MSI-H or dMMR colorectal cancer [EB/OL]. 2017-08-01.

URL    
7
Andre T, Shiu KK, Kim TW, et al. Pembrolizumab in microsatellite-instability-high advanced colorectal cancer [J]. N Engl J Med, 2020, 383(23): 2207-2218.
8
FDA. FDA approves pembrolizumab for first-line treatment of MSI-H/dMMR colorectal cancer [EB/OL]. 2020-6-30.

URL    
9
Andre T, Shiu K-K, Kim TW, et al. Final overall survival for the phase Ⅲ KN177 study: Pembrolizumab versus chemotherapy in microsatellite instability-high/mismatch repair deficient (MSI-H/dMMR) metastatic colorectal cancer (mCRC) [J].J Clin Oncol, 2021, 39(15_suppl): 3500.
10
Brunner MC, Chambers CA, Chan FK, et al. CTLA-4-Mediated inhibition of early events of T cell proliferation [J]. J Immunol, 1999, 162(10): 5813-5820.
11
Linsley PS, Greene JL, Brady W, et al. Human B7-1 (CD80) and B7-2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors [J]. Immunity, 1994, 1(9): 793-801.
12
Bachmann MF, Köhler G, Ecabert B, et al. Cutting edge: lymphoproliferative disease in the absence of CTLA-4 is not T cell autonomous [J]. J Immunol, 1999, 163(3): 1128-1131.
13
Matheu MP, Othy S, Greenberg ML, et al. Imaging regulatory T cell dynamics and CTLA4-mediated suppression of T cell priming [J]. Nat Commun, 2015, 6: 6219.
14
Jain N, Nguyen H, Chambers C, et al. Dual function of CTLA-4 in regulatory T cells and conventional T cells to prevent multiorgan autoimmunity [J]. Proc Natl Acad Sci U S A, 2010, 107(4): 1524-1528.
15
Herbst RS, Soria JC, Kowanetz M, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients [J]. Nature, 2014, 515(7528): 563-567.
16
Yarchoan M, Hopkins A, Jaffee EM. Tumor mutational burden and response rate to PD-1 inhibition [J]. N Engl J Med, 2017, 377(25): 2500-2501.
17
Guinney J, Dienstmann R, Wang X, et al. The consensus molecular subtypes of colorectal cancer [J]. Nat Med, 2015, 21(11): 1350-1356.
18
Ganesh K, Stadler ZK, Cercek A, et al. Immunotherapy in colorectal cancer: rationale, challenges and potential [J]. Nat Rev Gastroenterol Hepatol, 2019, 16(6): 361-375.
19
Samstein RM, Lee CH, Shoushtari AN, et al. Tumor mutational load predicts survival after immunotherapy across multiple cancer types [J]. Nat Genet, 2019, 51(2): 202-206.
20
Chan TA, Yarchoan M, Jaffee E, et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic [J]. Ann Oncol, 2019, 30(1): 44-56.
21
Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade [J]. Science, 2017, 357(6349): 409-413.
22
Roth MT, Das S. Pembrolizumab in unresectable or metastatic MSI-high colorectal cancer: safety and efficacy [J]. Expert Rev Anticancer Ther, 2021, 21(2): 229-238.
23
Wang QX, Qu CH, Gao YH, et al. The degree of microsatellite instability predicts response to PD-1 blockade immunotherapy in mismatch repair-deficient/microsatellite instability-high colorectal cancers [J]. Exp Hematol Oncol, 2021, 10(1): 2.
24
Chalabi M, Fanchi LF, Dijkstra KK, et al. Neoadjuvant immunotherapy leads to pathological responses in MMR-proficient and MMR-deficient early-stage colon cancers [J]. Nat Med, 2020, 26(4): 566-576.
25
Yuki S, Bando H, Tsukada Y, et al. Short-term results of VOLTAGE-A: Nivolumab monotherapy and subsequent radical surgery following spanoperative chemoradiotherapy in patients with microsatellite stable and microsatellite instability-high locally advanced rectal cancer [J]. J Clin Oncol 2020, 38(15_suppl): 4100.
26
Chen EX, Jonker DJ, Loree JM, et al. Effect of combined immune checkpoint inhibition vs best supportive care alone in patients with advanced colorectal cancer: The Canadian Cancer Trials Group CO.26 Study [J].JAMA Oncol,2020, 6(6): 831-838.
27
Wang L, Hui H, Agrawal K, et al. m(6) A RNA methyltransferases METTL3/14 regulate immune responses to anti-PD-1 therapy [J]. Embo J, 2020, 39(20): e104514.
28
Segal NH, Cercek A, Ku G, et al. Phase II single-arm study of Durvalumab and Tremelimumab with concurrent radiotherapy in patients with mismatch repair-proficient metastatic colorectal cancer [J]. Clin Cancer Res, 2021, 27(8): 2200-2208.
29
Monjazeb AM, Giobbie-Hurder A, Lako A, et al. A randomized trial of combined PD-L1 and CTLA-4 inhibition with targeted low-dose or hypofractionated radiation for patients with metastatic colorectal cancer [J]. Clin Cancer Res, 2021, 27(9): 2470-2480.
30
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.
31
Bergqvist V, Hertervig E, Gedeon P, et al. Vedolizumab treatment for immune checkpoint inhibitor-induced enterocolitis [J]. Cancer Immunol Immunother, 2017, 66(5): 581-592.
32
Brahmer JR, Lacchetti C, Schneider BJ, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline [J]. J Clin Oncol, 2018, 36(17): 1714-1768.
33
Lee JH, Long GV, Menzies AM, et al. Association between circulating tumor DNA and pseudoprogression in patients with metastatic melanoma treated with anti-programmed cell death 1 antibodies [J]. JAMA Oncol, 2018, 4(5): 717-721.
[1] 康夏, 田浩, 钱进, 高源, 缪洪明, 齐晓伟. 骨织素抑制破骨细胞分化改善肿瘤骨转移中骨溶解的机制研究[J]. 中华乳腺病杂志(电子版), 2023, 17(06): 329-339.
[2] 代莉, 邓恢伟, 郭华静, 黄芙蓉. 术中持续输注艾司氯胺酮对腹腔镜结直肠癌手术患者术后睡眠质量的影响[J]. 中华普通外科学文献(电子版), 2023, 17(06): 408-412.
[3] 李越洲, 张孔玺, 李小红, 商中华. 基于生物信息学分析胃癌中PUM的预后意义[J]. 中华普通外科学文献(电子版), 2023, 17(06): 426-432.
[4] 王得晨, 杨康, 杨自杰, 归明彬, 屈莲平, 张小凤, 高峰. 结直肠癌微卫星稳定状态和程序性死亡、吲哚胺2,3-双加氧酶关系的研究进展[J]. 中华普通外科学文献(电子版), 2023, 17(06): 462-465.
[5] 唐旭, 韩冰, 刘威, 陈茹星. 结直肠癌根治术后隐匿性肝转移危险因素分析及预测模型构建[J]. 中华普外科手术学杂志(电子版), 2024, 18(01): 16-20.
[6] 张生军, 赵阿静, 李守博, 郝祥宏, 刘敏丽. 高糖通过HGF/c-met通路促进结直肠癌侵袭和迁移的实验研究[J]. 中华普外科手术学杂志(电子版), 2024, 18(01): 21-24.
[7] 李婷, 张琳. 血清脂肪酸代谢物及维生素D水平与结直肠癌发生的关系研究[J]. 中华普外科手术学杂志(电子版), 2023, 17(06): 661-665.
[8] 魏小勇. 原发性肝癌转化治疗焦点问题探讨[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 602-607.
[9] 陈安, 冯娟, 杨振宇, 杜锡林, 柏强善, 阴继凯, 臧莉, 鲁建国. 基于生物信息学分析CCN4在肝细胞癌中表达及其临床意义[J]. 中华肝脏外科手术学电子杂志, 2023, 12(06): 702-707.
[10] 倪文凯, 齐翀, 许小丹, 周燮程, 殷庆章, 蔡元坤. 结直肠癌患者术后发生延迟性肠麻痹的影响因素分析[J]. 中华结直肠疾病电子杂志, 2023, 12(06): 484-489.
[11] 范小彧, 孙司正, 鄂一民, 喻春钊. 梗阻性左半结肠癌不同手术治疗方案的选择应用[J]. 中华结直肠疾病电子杂志, 2023, 12(06): 500-504.
[12] 关旭, 王锡山. 基于外科与免疫视角思考结直肠癌区域淋巴结处理的功与过[J]. 中华结直肠疾病电子杂志, 2023, 12(06): 448-452.
[13] 顾睿祈, 方洪生, 蔡国响. 循环肿瘤DNA检测在结直肠癌诊治中的应用与进展[J]. 中华结直肠疾病电子杂志, 2023, 12(06): 453-459.
[14] 吴蓉菊, 向平超. COPD频繁急性加重表型与炎性因子相关性研究[J]. 中华临床医师杂志(电子版), 2023, 17(9): 939-947.
[15] 符梅沙, 周玉华, 李慧, 薛春颜. 淋巴细胞免疫治疗对复发性流产患者外周血T淋巴细胞亚群分布与PD1/PD-L1表达的影响及意义[J]. 中华临床医师杂志(电子版), 2023, 17(06): 726-730.
阅读次数
全文


摘要