Advances in Clinical Medicine
Vol. 13  No. 05 ( 2023 ), Article ID: 64985 , 7 pages
10.12677/ACM.2023.135993

PD-1/PD-L1抑制剂在晚期胃癌治疗中的 研究进展

李雨婷,刘定定,黄子静,马雨琪,杨卫卫*

延安大学附属医院肿瘤科,陕西 延安

收稿日期:2023年4月7日;录用日期:2023年4月29日;发布日期:2023年5月8日

摘要

胃癌是我国常见的恶性肿瘤,传统化疗对晚期胃癌患者往往疗效有限。PD-1及其配体PD-L1是目前免疫治疗的研究热点。在肿瘤微环境中,肿瘤细胞表达大量PD-L1,PD-l与其配体结合后可抑制T细胞的功能,而PD-1/PD-L1抑制剂可重新激活效应T细胞,阻止肿瘤细胞逃逸。免疫单药治疗晚期胃癌已在临床中显示出显著疗效,但其联合用药模式是否将更好地改善患者预后尚未明确。期待未来深入研究PD-1/PD-L1抑制剂联合用药治疗晚期胃癌可使患者获益更多,包括化疗药物、抗血管生成药物及靶向药物等。

关键词

PD-1,PD-L1,晚期胃癌,免疫检查点抑制剂

Research Progress of PD-1/PD-L1 Inhibitors in the Treatment of Advanced Gastric Carcinoma

Yuting Li, Dingding Liu, Zijing Huang, Yuqi Ma, Weiwei Yang*

Department of Oncology, Affiliated Hospital of Yan’an University, Yan’an Shaanxi

Received: Apr. 7th, 2023; accepted: Apr. 29th, 2023; published: May 8th, 2023

ABSTRACT

The gastric carcinoma is a common malignant tumor in our country, and there is usually limited therapeutic effect on advanced gastric carcinoma treated by traditional chemotherapy. PD-1 and its ligand, PD-L1, have been the focus of immunotherapy research. In the tumor microenvironment, tumor cells express a large amount of PD-L1, and PD-l can inhibit the function of T cells after binding with its ligand, while PD-1/PD-L1 inhibitors can reactivate effector T cells and prevent the escape of tumor cells. Immunomonotherapy has shown significant clinical efficacy in the treatment of advanced gastric carcinoma, but it is not clear whether its combination mode will better improve the prognosis of patients. It is expected that further studies on the combination of PD-1/PD-L1 inhibitors in the treatment of advanced gastric carcinomacan bring more benefits to patients, including chemotherapy drugs, anti-angiogenic drugs and targeted drugs.

Keywords:PD-1, PD-L1, Advanced Gastric Carcinoma, Immune Checkpoint Inhibitors

Copyright © 2023 by author(s) and Hans Publishers Inc.

This work is licensed under the Creative Commons Attribution International License (CC BY 4.0).

http://creativecommons.org/licenses/by/4.0/

1. 引言

近几年,胃癌的发病率与致死率在恶性肿瘤中一直居高不下 [1] 。尤其是晚期胃癌患者5年生存率低,传统化疗方案疗效不佳。可喜的是,曲妥珠单抗(Trastuzumab)在人表皮生长因子受体2 (Human epidermal growth factor receptor 2, HER2)阳性的晚期胃癌患者中显示出显著疗效。然而在我国的晚期胃癌患者中,HER2阳性率仅12%左右,曲妥珠单抗对患者的获益也较为局限 [2] 。故研究新的靶向药物,或可为改善晚期胃癌患者的预后带来希望。

根据癌症基因组图谱(The Cancer Genome Atlas, TCGA)的研究成果,2014年美国国家癌症研究所(NationalCancerInstitute, NCI)提出了4种胃腺癌分子亚型:EB病毒感染型(Epstein-Barr virus, EBV)、微卫星不稳定型(Microsatellite Instability, MSI)、染色体不稳定型(Chromosomal instability, CIN)及基因组稳定型(Genomically Stable, GS)。其中,EBV型和MSI型免疫治疗可能获益,而CIN型和GS型不推荐免疫治疗 [3] 。2015年亚洲癌症研究组织(Asian Cancer Research Group, ACRG)根据MSI状态、TP53基因功能和上皮–间充质转化(Epithelial-to-mesenchymal transition, EMT)将胃癌分为4个亚型:高度微卫星不稳定(High-frequency MSI, MSI-H)、微卫星稳定(Microsatellite stability, MSS)/TP53+、MSS/TP53−和MSS/EMT [4] 。

活化的肿瘤浸润淋巴细胞(Tumor infiltrating lymphocytes, TILs)参与抗肿瘤免疫反应,包括CD8+T细胞、CD4+T细胞、B细胞和固有淋巴细胞等 [5] 。DNA错配修复基因MLH1、MSH2、MSH6和PMS2参与DNA的修复过程,基因功能缺失时将导致高度微卫星不稳定/错配修复缺陷(MSI-H/Deficient mismatch repair, MSI-H/dMMR)的发生。当肿瘤细胞发生MSI-H/dMMR时,细胞内肿瘤突变负荷(Tumor mutation burden, TMB)增加,进而形成了相关新抗原,使大量淋巴细胞浸润,有助于免疫检查点抑制剂(Immune checkpoint inhibitors, ICIs)发挥治疗效果 [6] 。

程序性死亡分子-1 (Programmed death-1,PD-l)及其配体(Programmed death-ligand 1, PD-L1)是目前免疫治疗的研究热点。PD-l作为一个表达于免疫细胞的负调控分子,与其配体PD-L1和PD-L2结合后可抑制T细胞的功能。在肿瘤微环境中,肿瘤细胞通过大量表达PD-L1,诱导负性免疫应答,从而得以逃逸。研究PD-1/PD-L1抑制剂可重新激活效应T细胞,阻止肿瘤细胞逃逸,达到抗肿瘤的作用 [7] 。现就PD-1及PD-L1抑制剂在晚期胃癌治疗中的研究进展予以综述。

2. PD-1抑制剂在晚期胃癌治疗中的应用现状

2.1. 纳武利尤单抗(Nivolumab)

在针对亚洲人群的ATTRACTION-2III期临床试验中,评估纳武利尤单抗对比安慰剂用于晚期胃/胃食管交界处腺癌(Gastric carcinoma/Gastro esophageal junction carcinoma, GC/GEJC)患者的疗效。结果显示,与安慰剂组相比,纳武利尤单抗组的客观缓解率(Objective response rate, ORR)为11%,中位无进展生存期(Median Progression free survival, mPFS)延长0.16个月(1.61个月vs 1.45个月),中位总生存期(Median Overall survival, mOS)延长1.12个月(5.26个月vs 4.14个月),1年生存率提高15.3% (26.2% vs 10.9%)。最常见的治疗相关不良反应(Treatment-related adverse events, TRAEs)为瘙痒、腹泻、皮疹、乏力、食欲减退和恶心等 [8] 。基于此研究结果,2017年纳武利尤单抗被日本批准用于晚期GC/GEJC的治疗,成为全球首个被批准用于治疗胃癌的ICIs。

在针对亚洲人群的ATTRACTION-4III期临床研究中,评估HER2阴性晚期GC/GEJC患者使用纳武利尤单抗联合SOX/XELOX (奥沙利铂 + 替吉奥/卡培他滨)的疗效及安全性。结果显示,纳武利尤单抗联合化疗组的患者显著改善了PFS (10.45个月vs 8.34个月),ORR提高了9.7% (57.5% vs 47.8%),缓解持续时间(Duration of response, DoR)延长了4.24个月(12.91个月vs 8.67个月),但未改善OS (17.45个月vs 17.15个月)。最常见的3/4级TRAEs是中性粒细胞计数下降和血小板计数下降 [9] 。而在CheckMate-649III期临床试验中,评估相比于单独化疗,纳武利尤单抗联合XELOX/FOLFOX (奥沙利铂 + 5-氟尿嘧啶 + 亚叶酸钙)作为一线方案治疗HER2阴性、既往未治疗过的晚期GC/GEJC患者的疗效。结果显示,在PD-L1阳性且联合阳性评分(Combined positive score, CPS) ≥ 5分的患者中,纳武利尤单抗联合化疗组患者的mPFS延长了1.7个月(7.7个月vs 6.0个月),mOS延长了3.3个月(14.4个月vs 11.1个月)。纳武利尤单抗联合细胞毒T淋巴细胞相关抗原4 (Cytotoxic T lymphocyte-associated antigen-4, CTLA-4)抑制剂依匹木单抗(Ipilimumab)组对比化疗组,表明MSI-H人群的OS和ORR均优于MSS人群。针对MSI-H/dMMR人群,纳武利尤单抗联合化疗组相比于单纯化疗组OS显著延长(38.7个月vs 12.3个月),ORR也大幅提高(55% vs 39%)。且在所有人群中,纳武利尤单抗联合化疗组的患者也实现了PFS和OS双双获益 [10] [11] 。相比于全球人群,中国亚组人群在纳武利尤单抗联合化疗一线治疗晚期GC/GEJC中获益更显著 [12] 。研究表明,纳武利尤单抗联合化疗方案可成为晚期GC/GEJC患者一线治疗的新选择。基于此研究结果,纳武利尤单抗成为中国第一个获批晚期胃癌一线治疗全人群适应症的PD-1抑制剂。

2.2. 帕博利珠单抗(Pembrolizumab)

在KEYNOTE-059II期临床试验中,评估PD-L1CPS ≥ 1的晚期GC/GEJC患者三线应用帕博利珠单抗单药治疗的安全性及有效性。结果显示:帕博利珠单抗的ORR为12%,mPFS为2个月,mOS为6个月。最常见的TRAEs为乏力、瘙痒、皮疹、食欲减退、贫血、恶心和腹泻等,3/4级TRAEs发生率为17.8%,该研究提示帕博利珠单抗在晚期GC/GEJC中具有较好的疗效和安全性 [13] 。基于此研究结果,2017年美国食品药品监督管理局(Food and Drug Administration, FDA)批准帕博利珠单抗用于PD-L1CPS ≥ 1的局部晚期、复发性或转移性GC/GEJC患者的三线治疗。

在KEYNOTE-062Ⅲ期临床试验中,评估帕博利珠单抗联合化疗对比单纯化疗一线治疗PD-L1CPS ≥ 1、既往未治疗过的晚期GC/GEJC的安全性及有效性。结果表明:与单纯化疗相比,帕博利珠单抗联合化疗组并不能显著延长患者mOS (12.5个月vs 11.1个月)和mPFS (6.9个月vs 6.4个月) [14] 。基于此研究结果,帕博利珠单抗联合化疗一线治疗胃癌效果还不明朗,仍需要大规模的临床试验来进一步探索。有学者对KEYNOTE-062中的MSI-H/dMMR晚期胃癌患者进行回顾性分析,结果表明:帕博利珠单抗单药或联合化疗疗效均显著优于化疗,ORR分别提高20.3%和27.9%,提示帕博利珠单抗在MSI-H/dMMR的晚期胃癌治疗中可取得较好的疗效且联合化疗可能优于单药治疗 [15] 。除此之外,PANTHERA研究 [16] 及KEYNOTE-811试验 [17] 均显示出,帕博利珠单抗联合曲妥珠单抗和化疗也显著改善了HER2阳性晚期胃癌患者的临床疗效。

2.3. 特瑞普利单抗(Toripalimab)

特瑞普利单抗是首款我国自主研发并在中国上市的PD-1单抗,也是NMPA批准的用于临床试验首批PD-1单克隆抗体之一 [18] 。在NCT02915432Ib/II期临床试验中,评估特瑞普利单抗单药治疗晚期胃癌的疗效。结果显示:ORR为12.1%,疾病控制率(Disease control rate, DCR)为39.7%,mPFS为1.9个月,mOS为4.8个月。最常见的TRAEs为贫血、瘙痒、蛋白尿、食欲减退、腹泻、恶心呕吐、发热和腹痛等。该研究表明特瑞普利单抗在晚期胃癌中显示出良好的疗效及安全性 [19] 。目前特瑞普利单抗已被批准用于治疗无法切除或转移性难治性黑色素瘤 [20] ,相信随着不断开展的临床研究,有望获批更多治疗适应症。

2.4. 信迪利单抗(Sintilimab)

信迪利单抗是我国自主研发并上市的第二款PD-1人源型IgG4单克隆抗体。在NCT02937116Ib期临床研究中,评估信迪利单抗联合XELOX一线治疗晚期GC/GEJC的疗效及安全性。结果显示:信迪利单抗联合化疗的ORR为85.0%,mPFS为7.5个月,1年生存率为68.0%。最常见的TRAEs为血象下降、甲状腺功能减退、皮疹、恶心呕吐、血钾降低、发热和蛋白尿等。该研究显示出信迪利单抗良好的疗效及安全性 [21] 。在ORIENT-16研究中,评估信迪利单抗联合XELOX一线治疗中国晚期胃癌患者的疗效及安全性。结果显示,总体人群ORR为58.2%,在PD-L1 CPS ≥ 5的患者中,ORR为72.8%,mPFS和OS分别为7.7个月和18.4个月,均显著优于单纯化疗。该实验数据提示,无论PD-L1的表达水平如何,信迪利单抗联合化疗方案均显示出良好的生存获益,证实了其在胃癌一线治疗中的疗效 [22] 。因此,信迪利单抗于2022年6月获得NMPA批准联合含氟尿嘧啶类和铂类药物化疗用于不可切除的局部晚期、复发或转移性GC/GEJC的一线治疗。同年信迪利单抗联合化疗被纳入CSCO胃癌诊疗指南2022中晚期胃癌一线全人群推荐。

2.5. 卡瑞利珠单抗(Camrelizumab)

卡瑞利珠单抗也是我国自主研发的PD-1抑制剂。在NCT02742935I期临床试验中,评估晚期GC/GEJC患者应用卡瑞利珠单抗治疗的有效性及安全性。结果显示:卡瑞利珠单抗的ORR为23.3%,DCR为43.3%,mPFS为8.0周。最常见的TRAEs为反应性毛细血管瘤、瘙痒、乏力、贫血、蛋白尿、恶心、发热和腹胀等。该研究数据提示,卡瑞利珠单抗在我国晚期GC/GEJC患者中显示出令人振奋的疗效 [23] 。2019年,NMPA批准卡瑞利珠单抗用于经典型霍奇金淋巴瘤患者的治疗,越来越多的临床研究积极开展,不断探索其单药和联合治疗的疗效及安全性。

2.6. 替雷利珠单抗(Tislelizumab)

替雷利珠单抗同为我国自主研发的抗PD-1单克隆抗体。在NCT03469557II期临床试验中,评估了替雷利珠单抗联合化疗治疗局部晚期/转移性GC/EGJC患者的安全性和有效性。结果表明,患者的ORR和DCR分别为46.7%和80.0%,mPFS为6.1个月,有7例患者获得部分缓解(Partial Remission, PR),3例患者获得稳定(Stable Disease, SD)。最常见的TRAEs为贫血、食欲减退、恶心和乏力等。该研究显示,无论PD-L1表达水平如何,替雷利珠单抗联合化疗治疗GC/EGJC均可取得长期的临床疗效,且具有良好的安全性 [24] 。此外,一项Ⅲ期临床研究正在进行中(NCT03777657),替雷利珠单抗联合化疗对比安慰剂联合化疗一线治疗局部晚期不可切除或转移性的GC/GEJC,期待早日传来好消息。目前替雷利珠单抗已获得NMPA附条件批准用于尿路上皮癌、经典型霍奇金淋巴瘤、肝细胞癌等多个瘤种的治疗。

3. PD-L1抑制剂在晚期胃癌治疗中的应用现状

3.1. 阿维单抗(Avelumab)

在JAVELIN Solid TumorIb期临床试验中,评估阿维单抗治疗晚期GC/GEJC的疗效及安全性,试验分为一线化疗后序贯阿维单抗治疗和阿维单抗二线治疗两个组。结果显示:两组的ORR均为6.7%,DCR分别为56.7%和28.3%,mPFS分别为2.8个月和1.4个月,mOS分别为11.1个月和6.6个月。最常见的TRAEs为输液反应、乏力、食欲下降、寒战、瘙痒、贫血、血小板计数减少和腹痛等。该研究表明阿维单抗良好的疗效及安全性 [25] 。而另一项JAVELIN Gastric300III期临床试验,评估阿维单抗与化疗分别作为三线治疗晚期GC/GEJC患者的安全性及有效性。结果显示,与化疗组相比,阿维单抗没有改善ORR (2.2% vs 4.3%),mOS (4.6个月vs 5.0个月)和mPFS (1.4个月vs 2.7个月),但阿维单抗治疗的安全性更高。最常见的TRAEs为恶心呕吐、腹泻、贫血、食欲减退、输液反应和乏力等 [26] 。同样在JAVELIN Gastric100III期临床试验中,比较阿维单抗和化疗作为一线维持治疗HER2阴性晚期GC/GEJC患者的疗效。结果表明:阿维单抗并没有显示出比持续化疗更好的Mos (10.4个月vs 10.9个月) [27] 。因此,阿维单抗在晚期GC/GEJC的疗效仍需要大规模的临床试验来进一步探索。

3.2. 度伐利尤单抗(Durvalumab)

在NCT02572687Ia/b期临床试验中,评估血管内皮生长因子受体2 (Vascular endothelial growth factor receptor 2, VEGFR2)拮抗剂雷莫卢单抗(Ramucirumab)联合度伐利尤单抗治疗晚期GC/GEJC患者的疗效。结果显示,患者的ORR为21%,DCR为55%,mPFS为2.6个月,mOS为2.4个月。与PD-L1低表达组相比,PD-L1高表达组显著改善ORR (36% vs 0%),mPFS (5.5个月vs 1.5个月)及mOS (14.8个月vs 5.5个月) [28] 。另一项Ib/II期临床试验中,评估CTLA-4抑制剂替西木单抗(Tremelimumab)和度伐利尤单抗单药或联合治疗晚期GC/GEJC患者的有效性及安全性。结果表明,患者的ORR为(8.3% vs 0% vs 7.4%),DCR为(25.0% vs 12.5% vs 25.9%),mPFS为(1.7个月vs 1.6个月vs 1.8个月),mOS为(7.7个月vs 3.4个月vs 9.2个月),1年生存率为(22.9% vs 4.6% vs 37.0%)。最常见的TRAEs为食欲下降、乏力、腹泻和恶心呕吐等。无论何种疗法都没有使患者显著受益 [29] 。

3.3. 恩沃利单抗(Envafolimab)

恩沃利单抗是一种重组人源化PD-L1单域抗体,是首款皮下注射PD-L1抑制剂。在NCT03667170II期临床研究中,评估恩沃利单抗单药治疗既往标准治疗失败的晚期MSI-H/dMMR实体肿瘤的疗效及安全性。结果显示,65例结直肠癌患者的ORR为43.1%,18例胃癌患者的ORR为44.4%,20例其他实体瘤患者的ORR为40.0%。总体人群的ORR为42.7%,mPFS为11.1个月,12个月DoR率为92.2%,1年生存率为74.6%。3/4级TRAEs发生率为16%,未发生TRAEs导致死亡。3/4级免疫相关不良反应(Immune-related adverse events, irAEs)发生率为8%,未发生免疫相关性肺炎、结肠炎、肾炎和神经系统毒性等。9%的患者发生注射部位反应,但均为1/2级。该试验显示出恩沃利单抗良好的疗效及安全性 [30] 。基于此研究数据,2021年恩沃利单抗得到NMPA批准用于治疗既往标准治疗失败的晚期MSI-H/dMMR实体瘤。今后需要更多的大规模临床研究来进一步证实恩沃利单抗联合用药的有效性和安全性。

4. 小结

PD-l表达于免疫细胞,与其配体PD-L1和PD-L2结合后可抑制T细胞的功能。肿瘤细胞通过大量表达PD-L1,诱导负性免疫应答,从而得以逃逸 [7] 。因而研究PD-1/PD-L1抑制剂可达到抗肿瘤的作用。尤其是晚期胃癌患者5年生存率低,传统化疗方案疗效不佳,免疫治疗或可为改善晚期胃癌患者预后带来希望。随着PD-1/PD-L1抑制剂单药治疗晚期GC/GEJC的临床试验越来越多,其疗效和安全性已得到充分证实。但PD-1/PD-L1抑制剂与其他药物联合治疗的疗效及安全性,仍需更多的临床研究进一步明确。

文章引用

李雨婷,刘定定,黄子静,马雨琪,杨卫卫. PD-1/PD-L1抑制剂在晚期胃癌治疗中的研究进展
Research Progress of PD-1/PD-L1 Inhibitors in the Treatment of Advanced Gastric Carcinoma[J]. 临床医学进展, 2023, 13(05): 7108-7114. https://doi.org/10.12677/ACM.2023.135993

参考文献

  1. 1. Sung, H., Ferlay, J., Siegel, R.L., et al. (2021) Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer Journal for Clinicians, 71, 209-249. https://doi.org/10.3322/caac.21660

  2. 2. 张俊, 秦叔逵. HER2阳性晚期胃癌分子靶向治疗的中国专家共识(2016版) [J]. 临床肿瘤学杂志, 2016, 21(9): 831-839.

  3. 3. Cancer Genome Atlas Research Network (2014) Com-prehensive Molecular Characterization of Gastric Adenocarcinoma. Nature, 513, 202-209. https://doi.org/10.1038/nature13480

  4. 4. Cristescu, R., Lee, J., Nebozhyn, M., et al. (2015) Molecular Analysis of Gastric Cancer Identifies Subtypes Associated with Distinct Clinical Outcomes. Nature Medicine, 21, 449-456. https://doi.org/10.1038/nm.3850

  5. 5. Paijens, S.T., Vledder, A., de Bruyn, M., et al. (2021) Tumor-Infiltrating Lymphocytes in the Immunotherapy Era. Cellular & Molecular Immunology, 18, 842-859. https://doi.org/10.1038/s41423-020-00565-9

  6. 6. Shum, B., Larkin, J. and Turajlic, S. (2022) Predictive Bi-omarkers for Response to Immune Checkpoint Inhibition. Seminars in Cancer Biology, 79, 4-17. https://doi.org/10.1016/j.semcancer.2021.03.036

  7. 7. McDermott, D.F. and Atkins, M.B. (2013) PD-1 as a Po-tential Target in Cancer Therapy. Cancer Medicine, 2, 662-673. https://doi.org/10.1002/cam4.106

  8. 8. Kang, Y.-K., Boku, N., Satoh, T., et al. (2017) Nivolumab in Patients with Advanced Gastric or Gastro-Oesophageal Junction Cancer Refractory to, or Intolerant of, at Least Two Previous Chem-otherapy Regimens (ONO-4538-12, ATTRACTION-2): A Randomised, Double-Blind, Placebo-Controlled, Phase 3 Trial. The Lancet, 390, 2461-2471.

  9. 9. Kang, Y.K., Chen, L.T., Ryu, M.H., et al. (2022) Nivolumab plus Chemo-therapyversus Placebo plus Chemotherapy in Patients with HER2-Negative, Untreated, Unresectable Advanced or Re-current Gastric or Gastroesophageal Junction Cancer (ATTRACTION-4): A Randomised, Multicentre, Double-Blind, Placebo-Controlled, Phase 3 Trial. The Lancet Oncology, 23, 234-247. https://doi.org/10.1016/S1470-2045(21)00692-6

  10. 10. Janjigian, Y.Y., Shitara, K., Moehler, M., et al. (2021) First-Line Nivolumab plus Chemotherapy versus Chemotherapy Alone for advanced Gastric, Gastro-Oesophageal Junc-tion, and Oesophageal Adenocarcinoma (CheckMate 649): A Randomised, Open-Label, Phase 3 Trial. The Lancet, 398, 27-40. https://doi.org/10.1016/S0140-6736(21)00797-2

  11. 11. Janjigian, Y.Y., Ajani, J.A., Moehler, M., et al. (2021) LBA7 Nivolumab (NIVO) plus Chemotherapy (Chemo) or Ipilimumab (IPI) vs Chemo as First-Line (1L) Treat-ment for Advanced Gastric Cancer/Gastroesophageal Junction Cancer/Esophageal Adenocarcinoma (GC/GEJC/EAC): CheckMate 649 Study. Annals of Oncology, 32, S1329-S1330. https://doi.org/10.1016/j.annonc.2021.08.2131

  12. 12. Shen, L., Bai, Y.X., Lin, X.Y., et al. (2021) First-Line (1L) Nivolumab (NIVO) plus Chemotherapy (Chemo) versus Chemo in Patients (pts) with Advanced Gastric Can-cer/Gastroesophageal Junction Cancer/Esophageal Adenocarcinoma (GC/GEJC/EAC): CheckMate 649 Chinese Sub-group Analysis. Cancer Research, 81, CT184. https://doi.org/10.1158/1538-7445.AM2021-CT184

  13. 13. Fuchs, C.S., Doi, T., Jang, R.W., et al. (2018) Safety and Efficacy of Pembrolizumab Monotherapy in Patients with Previously Treated Advanced Gastric and Gastroesophageal Junction Cancer: Phase 2 Clinical KEYNOTE-059 Trial. JAMA Oncology, 4, e180013.

  14. 14. Shitara, K., Van Cutsem, E., Bang, Y.J., et al. (2020) Efficacy and Safety of Pembrolizumab or Pembrolizumab plus Chemotherapy vs Chemo-therapy Alone for Patients with First-line, Advanced Gastric Cancer: The KEYNOTE-062 Phase 3 Randomized Clinical Trial. JAMA Oncology, 6, 1571-1580. https://doi.org/10.1001/jamaoncol.2020.3370

  15. 15. Pietrantonio, F., Randon, G., Di Bartolomeo, M., et al. (2021) Predictive Role of Microsatellite Instability for PD-1 Blockade in Patients with Ad-vanced Gastric Cancer: A Meta-Analysis of Randomized Clinical Trials. ESMO Open, 6, Article ID: 100036. https://doi.org/10.1016/j.esmoop.2020.100036

  16. 16. Rha, S., Lee, C.K., Kim, H.S., et al. (2021) A Mul-ti-Institutional Phase Ib/II Trial of First-Line Triplet Regimen (Pembrolizumab, Trastuzumab, Chemotherapy) for HER2-Positive Advanced Gastric and Gastroesophageal Junction Cancer (PANTHERA Trial): Molecular Profiling and Clinical Update. Journal of Clinical Oncology, 39, 218. https://doi.org/10.1200/JCO.2021.39.3_suppl.218

  17. 17. Janjigian, Y.Y., Kawazoe, A., Yanez, P., et al. (2021) The KEYNOTE-811 Trial of Dual PD-1 and HER2 Blockade in HER2-Positive Gastric Cancer. Nature, 600, 727-730. https://doi.org/10.1038/s41586-021-04161-3

  18. 18. Fu, J., Wang, F., Dong, L.-H., et al. (2017) Preclinical Evalua-tion of the Efficacy, Pharmacokinetics and Immunogenicity of JS-001, a Programmed Cell Death Protein-1 (PD-1) Mon-oclonal Antibody. Acta Pharmacologica Sinica, 38, 710-718. https://doi.org/10.1038/aps.2016.161

  19. 19. Wang, F., Wei, X.L., Wang, F.H., et al. (2019) Safety, Efficacy and Tumor Mutational Burden as a Biomarker of Overall Survival Benefit in Chemo-Refractory Gastric Cancer Treated with Toripalimab, a PD-1 Antibody in Phase Ib/II Clinical Trial NCT02915432. Annals of Oncology, 30, 1479-1486. https://doi.org/10.1093/annonc/mdz197

  20. 20. Keam, S.J. (2019) Toripalimab: First Global Approval. Drugs, 79, 573-578. https://doi.org/10.1007/s40265-019-01076-2

  21. 21. Jiang, H.P., Zheng, Y.L., Qian, J., et al. (2020) Safety and Ef-ficacy of Sintilimab Combined with Oxaliplatin/Capecitabine as First-Line Treatment in Patients with Locally Advanced or Metastatic Gastric/Gastroesophageal Junction Adenocarcinoma in a Phase Ib Clinical Trial. BMC Cancer, 20, Article No. 760. https://doi.org/10.1186/s12885-020-07251-z

  22. 22. Xu, J.M., Jiang, H.P., Pan, Y.Y., et al. (2021) LBA53 Sintili-mab 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. An-nals of Oncology, 32, S1331. https://doi.org/10.1016/j.annonc.2021.08.2133

  23. 23. Huang, J., Mo, H.N., Zhang, W.L., et al. (2018) Promising Ef-ficacy of SHR‐1210, a Novel Anti-Programmed Cell Death 1 Antibody, in Patients with Advanced Gastric and Gas-troesophageal Junction Cancer in China. Cancer, 125, 742-749. https://doi.org/10.1002/cncr.31855

  24. 24. Xu, J.M., Bai, Y.X., Xu, N., et al. (2020) Tislelizumab plus Chemotherapy as First-line Treatment for Advanced Esophageal Squamous Cell Carcinoma and Gastric/Gastroesophageal Junction Adenocarcinoma. Clinical Cancer Research, 26, 4542-4550. https://doi.org/10.1158/1078-0432.CCR-19-3561

  25. 25. Chung, H.C., Arkenau, H.T., Lee, J., et al. (2019) Avelumab (Anti-PD-L1) as First-Line Switch-Maintenance or Second-Line Therapy in Patients with Advanced Gastric or Gastroesophageal Junction Cancer: Phase 1b Results from the JAVELIN Solid Tumor Trial. Journal for Im-munotherapy of Cancer, 7, 30. https://doi.org/10.1186/s40425-019-0508-1

  26. 26. Bang, Y.J., Ruiz, E.Y., Van Cutsem, E., et al. (2018) Phase III, Randomised Trial of Avelumab versus Physician’s Choice of Chemotherapy as Third-Line Treatment of Patients with Advanced Gastric or Gastro-Oesophageal Junction Cancer: Primary Analysis of JAVELIN Gastric 300. Annals of On-cology, 29, 2052-2060. https://doi.org/10.1093/annonc/mdy264

  27. 27. Moehler, M., Dvorkin, M., Boku, N., et al. (2021) Phase III Trial of Avelumab Maintenance after First-Line Induction Chemotherapy versus Continuation of Chemotherapy in Patients with Gastric Cancers: Results from JAVELIN Gastric 100. Journal of Clinical Oncology, 39, 966-977. https://doi.org/10.1200/JCO.20.00892

  28. 28. Bang, Y.J., Golan, T., Dahan, L., et al. (2020) Ramucirumab and Durvalumab for Previously Treated, Advanced Non-Small-Cell Lung Cancer, Gastric/Gastro-Oesophageal Junction Ad-enocarcinoma, or Hepatocellular Carcinoma: An Open-Label, Phase Ia/b Study (JVDJ). European Journal of Cancer, 137, 272-284. https://doi.org/10.1016/j.ejca.2020.06.007

  29. 29. Kelly, R.J., Lee, J., Bang, Y.J., et al. (2020) Safety and Efficacy of Durvalumab and Tremelimumab Alone or in Combination in Patients with Advanced Gastric and Gastroesophageal Junc-tion Adenocarcinoma. Clinical Cancer Research, 26, 846-854. https://doi.org/10.1158/1078-0432.CCR-19-2443

  30. 30. Shen, L., Li, J., Deng, Y.H., et al. (2020) Envaolimab (KN035) in Advanced Tumors with Mismatch-Repair Deficiency. Journal of Clinical Oncology, 38, S3021. https://doi.org/10.1200/JCO.2020.38.15_suppl.3021

期刊菜单