国际肿瘤学杂志››2024,Vol. 51››Issue (3): 175-180.doi:10.3760/cma.j.cn371439-20231109-00028
收稿日期:
2023-11-09修回日期:
2023-12-04出版日期:
2024-03-08发布日期:
2024-04-10通讯作者:
沙丹,Email:
基金资助:
Liu Yulan1,2, Jing Haiyan3, Sun Jing1, Song Wei2, Sha Dan2()
Received:
2023-11-09Revised:
2023-12-04Online:
2024-03-08Published:
2024-04-10Contact:
Sha Dan, Email:
Supported by:
摘要:
由于胃癌具有较高的肿瘤内及肿瘤间异质性,导致患者在免疫治疗疗效和预后方面差异极大。程序性死亡受体配体1、人表皮生长因子受体2、肿瘤微环境特征、外周血炎性标志物及Claudin18.2等多种生物标志物均显示出对免疫治疗疗效和患者预后的预测价值,有助于临床医生筛选免疫治疗潜在获益人群,实现精准化治疗。
刘玉兰, 井海燕, 孙静, 宋伟, 沙丹. 胃癌免疫治疗疗效预测及预后标志物的研究进展[J]. 国际肿瘤学杂志, 2024, 51(3): 175-180.
Liu Yulan, Jing Haiyan, Sun Jing, Song Wei, Sha Dan. Advances in predicting efficacy and prognostic markers of immunotherapy for gastric cancer[J]. Journal of International Oncology, 2024, 51(3): 175-180.
[1] | Cao W, Chen HD, Yu YW, et al. Changing profiles of cancer burden worldwide and in China: a secondary analysis of the global cancer statistics 2020[J].Chin Med J (Engl),2021,134(7): 783-791. DOI:10.1097/CM9.0000000000001474. |
[2] | Chen M, Li C, Sun M, et al. Recent developments in PD-1/PD-L1 blockade research for gastroesophageal malignancies[J].Front Immunol,2022,13: 1043517. DOI:10.3389/fimmu.2022.1043517. |
[3] | Janjigian YY, Shitara K, Moehler M, et al. First-line nivolumab plus chemotherapy versus chemotherapy alone for advanced gastric, gastro-oesophageal junction, and oesophageal adenocarcinoma (CheckMate 649): a randomised, open-label, phase 3 trial[J].Lancet,2021,398(10294): 27-40. DOI:10.1016/S0140-6736(21)00797-2. pmid:34102137 |
[4] | Xu JM, Jiang H, Pan Y, et al. LBA53 Sintilimab plus chemotherapy (chemo) versus chemo as first-line treatment for advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma (ORIENT-16): first results of a randomized, double-blind, phase Ⅲ study[J].Ann Oncol,2021,32: S1331. DOI:10.1016/j.annonc.2021.08.2133. |
[5] | Gao Y, Li S, Xu D, et al. Prognostic value of programmed death-1, programmed death-ligand 1, programmed death-ligand 2 expression, and CD8(+) T cell density in primary tumors and metastatic lymph nodes from patients with stage T1-4N+M0gastric adenocarcinoma[J].Chin J Cancer,2017,36(1): 61. DOI:10.1186/s40880-017-0226-3. |
[6] | Park Y, Koh J, Na HY, et al. PD-L1 testing in gastric cancer by the combined positive score of the 22C3 PharmDx and SP263 assay with clinically relevant cut-offs[J].Cancer Res Treat,2020,52(3): 661-670. DOI:10.4143/crt.2019.718. pmid:32019283 |
[7] | Van Cutsem E, Bang YJ, Feng-Yi F, et al. HER2 screening data from ToGA: targeting HER2 in gastric and gastroesophageal junction cancer[J].Gastric Cancer,2015,18(3): 476-484. DOI:10.1007/s10120-014-0402-y. pmid:25038874 |
[8] | Janjigian YY, Kawazoe A, Bai Y, et al. Pembrolizumab plus trastuzumab and chemotherapy for HER2-positive gastric or gastro-oesophageal junction adenocarcinoma: interim analyses from the phase 3 KEYNOTE-811 randomised placebo-controlled trial[J].Lancet,2023,402(10418): 2197-2208. DOI:10.1016/S0140-6736(23)02033-0. pmid:37871604 |
[9] | Lian J, Zhang G, Zhang Y, et al. PD-L1 and HER2 expression in gastric adenocarcinoma and their prognostic significance[J].Dig Liver Dis,2022,54(10): 1419-1427. DOI:10.1016/j.dld.2022.01.128. |
[10] | Catenacci DVT, Kang YK, Park H, et al. Margetuximab plus pembrolizumab in patients with previously treated, HER2-positive gastro-oesophageal adenocarcinoma (CP-MGAH22-05): a single-arm, phase 1b-2 trial[J].Lancet Oncol,2020, 21(8): 1066-1076. DOI:10.1016/S1470-2045(20)30326-0. |
[11] | Triulzi T, Forte L, Regondi V, et al. HER2 signaling regulates the tumor immune microenvironment and trastuzumab efficacy[J].Oncoimmunology,2018,8(1): e1512942. DOI:10.1080/2162402X.2018.1512942. |
[12] | Yagi S, Wakatsuki T, Yamamoto N, et al. Clinical significance of intratumoral HER2 heterogeneity on trastuzumab efficacy using endoscopic biopsy specimens in patients with advanced HER2 positive gastric cancer[J].Gastric Cancer,2019,22(3): 518-525. DOI:10.1007/s10120-018-0887-x. pmid:30328533 |
[13] | Park SR, Park YS, Ryu MH, et al. Extra-gain of HER2-positive cases through HER2 reassessment in primary and metastatic sites in advanced gastric cancer with initially HER2-negative primary tumours: results of GASTric cancer HER2 reassessment study 1 (GASTHER1)[J].Eur J Cancer,2016,53: 42-50. DOI:10.1016/j.ejca.2015.09.018. pmid:26693898 |
[14] | Seo S, Ryu MH, Park YS, et al. Loss of HER2 positivity after anti-HER2 chemotherapy in HER2-positive gastric cancer patients: results of the GASTric cancer HER2 reassessment study 3 (GASTHER3)[J].Gastric Cancer,2019,22(3): 527-535. DOI:10.1007/s10120-018-0891-1. pmid:30386954 |
[15] | Choi S, Park S, Kim H, et al. Gastric cancer: mechanisms, biomarkers, and therapeutic approaches[J].Biomedicines,2022,10(3): 543. DOI:10.3390/biomedicines10030543. |
[16] | Hinshaw DC, Shevde LA. The tumor microenvironment innately modulates cancer progression[J].Cancer Res,2019,79(18): 4557-4566. DOI:10.1158/0008-5472.CAN-18-3962. pmid:31350295 |
[17] | Tian C, Jing H, Wang C, et al. Prognostic role of tumour-infiltrating lymphocytes assessed by H&E-stained section in gastric cancer: a systematic review and meta-analysis[J].BMJ Open,2021,11(1): e044163. DOI:10.1136/bmjopen-2020-044163. |
[18] | 吴杨, 陈雨琪, 石通国, 等. 胃癌患者免疫微环境中表型特征及其与临床病理参数和预后的关系[J].中华医学杂志,2023,103(35): 2786-2794. DOI:10.3760/cma.j.cn112137-20230314-00394. |
[19] | Mori T, Tanaka H, Suzuki S, et al. Tertiary lymphoid structures show infiltration of effective tumor-resident T cells in gastric cancer[J].Cancer Sci,2021,112(5): 1746-1757. DOI:10.1111/cas.14888. |
[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. DOI:10.3389/fonc.2020.00980. pmid:32733793 |
[21] | Yin YX, Ling YH, Wei XL, et al. Impact of mature tertiary lymphoid structures on prognosis and therapeutic response of Epstein-Barr virus-associated gastric cancer patients[J].Front Immunol,2022,13: 973085. DOI:10.3389/fimmu.2022.973085. |
[22] | Jiang Q, Tian C, Wu H, et al. Tertiary lymphoid structure patterns predicted anti-PD1 therapeutic responses in gastric cancer[J].Chin J Cancer Res,2022,34(4): 365-382. DOI:10.21147/j.issn.1000-9604.2022.04.05. |
[23] | Lugli A, Zlobec I, Berger MD, et al. Tumour budding in solid cancers[J].Nat Rev Clin Oncol,2021,18(2): 101-115. DOI:10.1038/s41571-020-0422-y. |
[24] | Xue C, Du Y, Li Y, et al. Tumor budding as a predictor for prognosis and therapeutic response in gastric cancer: a mini review[J].Front Oncol,2022,12: 1003959. DOI:10.3389/fonc.2022.1003959. |
[25] | Zhang N, Wang D, Duan Y, et al. The special immune microenvironment of tumor budding and its impact on prognosis in gastric adenocarcinoma[J].Pathol Res Pract,2020,216(6): 152926. DOI:10.1016/j.prp.2020.152926. |
[26] | Ulase D, Heckl S, Behrens HM, et al. Prognostic significance of tumour budding assessed in gastric carcinoma according to the criteria of the international tumour budding consensus conference[J].Histopathology,2020,76(3): 433-446. DOI:10.1111/his.13997. pmid:31538348 |
[27] | Szalai L, Jakab Á, Kocsmár I, et al. Prognostic ability of tumor budding outperforms poorly differentiated clusters in gastric cancer[J].Cancers (Basel),2022,14(19): 4731. DOI:10.3390/cancers14194731. |
[28] | Piersma B, Hayward MK, Weaver VM. Fibrosis and cancer: a strained relationship[J].Biochim Biophys Acta Rev Cancer,2020,1873(2):188356. DOI:10.1016/j.bbcan.2020.188356. |
[29] | Aurello P, Berardi G, Giulitti D, et al. Tumor-stroma ratio is an independent predictor for overall survival and disease free survival in gastric cancer patients[J].Surgeon,2017,15(6): 329-335. DOI:10.1016/j.surge.2017.05.007. |
[30] | Kim EY, Abdul-Ghafar J, Chong Y, et al. Calculated tumor-associated neutrophils are associated with the tumor-stroma ratio and predict a poor prognosis in advanced gastric cancer[J].Biomedicines,2022,10(3): 708. DOI:10.3390/biomedicines10030708. |
[31] | Qu Z, Wang Q, Wang H, et al. The effect of inflammatory markers on the survival of advanced gastric cancer patients who underwent anti-programmed death 1 therapy[J].Front Oncol,2022,12: 783197. DOI:10.3389/fonc.2022.783197. |
[32] | Chen Y, Zhang C, Peng Z, et al. Association of lymphocyte-to-monocyte ratio with survival in advanced gastric cancer patients treated with immune checkpoint inhibitor[J].Front Oncol,2021,11: 589022. DOI:10.3389/fonc.2021.589022. |
[33] | Shitara K, Lordick F, Bang YJ, et al. Zolbetuximab plus mFOLFOX6 in patients with CLDN18.2-positive, HER2-negative, untreated, locally advanced unresectable or metastatic gastric or gastro-oesophageal junction adenocarcinoma (SPOTLIGHT): a multicentre, randomised, double-blind, phase 3 trial[J].Lancet,2023,401(10389): 1655-1668. DOI:10.1016/S0140-6736(23)00620-7. pmid:37068504 |
[34] | Rohde C, Yamaguchi R, Mukhina S, et al. Comparison of claudin 18.2 expression in primary tumors and lymph node metastases in Japanese patients with gastric adenocarcinoma[J].Jpn J Clin Oncol,2019,49(9): 870-876. DOI:10.1093/jjco/hyz068. pmid:31087075 |
[35] | Sahin U, Türeci Ö, Manikhas G, et al. FAST: a randomised phase Ⅱ study of zolbetuximab (IMAB362) plus EOX versus EOX alone for first-line treatment of advanced CLDN18.2-positive gastric and gastro-oesophageal adenocarcinoma[J].Ann Oncol,2021,32(5): 609-619. DOI:10.1016/j.annonc.2021.02.005. pmid:33610734 |
[36] | Gao J, Wang Z, Jiang W, et al. CLDN18.2 and 4-1BB bispecific antibody givastomig exerts antitumor activity through CLDN18.2-expressing tumor-directed T-cell activation[J].J Immunother Cancer,2023,11(6): e006704. DOI:10.1136/jitc-2023-006704. |
[1] | 刘娜, 寇介丽, 杨枫, 刘桃桃, 李丹萍, 韩君蕊, 杨立洲.血清miR-106b-5p、miR-760联合低剂量螺旋CT诊断早期肺癌的临床价值[J]. 国际肿瘤学杂志, 2024, 51(6): 321-325. |
[2] | 钱晓涛, 石子宜, 胡格, 吴晓维.Ⅲ~ⅣA期食管鳞状细胞癌放化疗后行巩固化疗的疗效:一项真实世界临床研究[J]. 国际肿瘤学杂志, 2024, 51(6): 326-331. |
[3] | 杨蜜, 别俊, 张加勇, 邓佳秀, 唐组阁, 卢俊.局部晚期可切除食管癌新辅助治疗疗效及预后分析[J]. 国际肿瘤学杂志, 2024, 51(6): 332-337. |
[4] | 袁健, 黄燕华.Hp-IgG抗体联合血清DKK1、sB7-H3对早期胃癌的诊断价值[J]. 国际肿瘤学杂志, 2024, 51(6): 338-343. |
[5] | 陈红健, 张素青.血清miR-24-3p、H2AFX与肝癌患者临床病理特征及术后复发的关系研究[J]. 国际肿瘤学杂志, 2024, 51(6): 344-349. |
[6] | 郭泽浩, 张俊旺.PFDN及其亚基在肿瘤发生发展中的作用[J]. 国际肿瘤学杂志, 2024, 51(6): 350-353. |
[7] | 张百红, 岳红云.新作用机制的抗肿瘤药物进展[J]. 国际肿瘤学杂志, 2024, 51(6): 354-358. |
[8] | 许凤琳, 吴刚.EBV在鼻咽癌肿瘤免疫微环境和免疫治疗中的研究进展[J]. 国际肿瘤学杂志, 2024, 51(6): 359-363. |
[9] | 王盈, 刘楠, 郭兵.抗体药物偶联物在转移性乳腺癌治疗中的研究进展[J]. 国际肿瘤学杂志, 2024, 51(6): 364-369. |
[10] | 张蕊, 褚衍六.基于FIT与肠道菌群的结直肠癌风险评估模型的研究进展[J]. 国际肿瘤学杂志, 2024, 51(6): 370-375. |
[11] | 高凡, 王萍, 杜超, 褚衍六.肠道菌群与结直肠癌非手术治疗的相关研究进展[J]. 国际肿瘤学杂志, 2024, 51(6): 376-381. |
[12] | 王丽, 刘志华, 杨伟洪, 蒋凤莲, 李全泳, 宋浩杰, 鞠文东.ROS1突变肺腺鳞癌合并脑梗死为主要表现的Trousseau综合征1例[J]. 国际肿瘤学杂志, 2024, 51(6): 382-384. |
[13] | 范志鹏, 余静, 胡静, 廖正凯, 徐禹, 欧阳雯, 谢丛华.炎症标志物的变化趋势对一线接受免疫联合化疗的晚期非小细胞肺癌患者预后的预测价值[J]. 国际肿瘤学杂志, 2024, 51(5): 257-266. |
[14] | 刘静, 刘芹, 黄梅.基于SMOTE算法的食管癌放化疗患者肺部感染的预后模型构建[J]. 国际肿瘤学杂志, 2024, 51(5): 267-273. |
[15] | 杨琳, 路宁, 温华, 张明鑫, 朱琳.炎症负荷指数与胃癌临床关系研究[J]. 国际肿瘤学杂志, 2024, 51(5): 274-279. |
阅读次数 | ||||||
全文 |
|
|||||
摘要 |
|
|||||