免疫检查点抑制剂治疗晚期胃癌的研究进展
Research Progress of Immune Checkpoint Inhibitors in the Treatment of Advanced Gastric Cancer
DOI: 10.12677/ACM.2023.13102264, PDF, HTML, XML, 下载: 173  浏览: 224 
作者: 王静如*, 张恩慧, 曲颜丽#:新疆医科大学附属肿瘤医院,新疆 乌鲁木齐
关键词: 胃癌晚期胃癌免疫检查点抑制剂生物标志物Gastric Cancer Advanced Gastric Cancer Immune Checkpoint Inhibitors Biomarkers
摘要: 胃癌(gastric cancer, GC)是世界范围内癌症相关死亡原因第三位,其发病率及死亡率均位居全球恶性肿瘤的前五位。多数胃癌患者确诊时往往已进展至中晚期,缺乏有效的治疗手段,预后极差。尽管化疗和靶向治疗在内的内科治疗是目前晚期胃癌(advanced gastric cancer, AGC)的主要治疗手段,但因为胃癌的异质性,化疗和靶向治疗的疗效似乎已经达到平台期。近年来,免疫检查点抑制剂(immune checkpoint inhibitors, ICIs)治疗在部分恶性肿瘤的治疗中取得重大进展,在AGC中,ICIs的疗效也被多项大规模研究进一步证实。本综述探讨了不同ICIs在晚期胃癌中的临床研究进展及近期研究数据。
Abstract: Gastric cancer is the third leading cause of cancer-related deaths worldwide, and its incidence and mortality rates are among the top five malignant tumors in the world. Most patients with gastric cancer have already progressed to the middle and late stages when diagnosed, and lack of effective treatments, resulting in a very poor prognosis. Although medical treatment, including chemother-apy and targeted therapy, is currently the main treatment for advanced gastric cancer, the efficacy of chemotherapy and targeted therapy seems to have reached a plateau because of the heterogene-ity of gastric cancer. In recent years, immune checkpoint inhibitors (ICIs) therapy has made signifi-cant progress in the treatment of some malignant tumors, and the efficacy of ICIs has been further confirmed by several large-scale studies in AGC. This review discusses the progress of clinical stud-ies and recent research data of different ICIs in advanced gastric cancer.
文章引用:王静如, 张恩慧, 曲颜丽. 免疫检查点抑制剂治疗晚期胃癌的研究进展[J]. 临床医学进展, 2023, 13(10): 16191-16197. https://doi.org/10.12677/ACM.2023.13102264

1. 背景

胃癌(gastric cancer, GC)是世界范围内一大待解的难题,全球每年约有1万的新发病例,其发病率及死亡率均位居全球恶性肿瘤的前五位 [1] 。90%以上的胃癌为胃腺癌,早期症状不典型,确诊时往往已进展至中晚期,5年生存率仅为20% [2] 。晚期胃癌(advanced gastric cancer, AGC)患者目前主要的治疗手段包括化疗和靶向治疗在内的内科治疗,缺乏有效的治疗手段,但预后仍较差 [3] 。AGC的主要一线化疗包括铂类药物(通常为奥沙利铂)和细胞毒性化合物(如5FU),主要是FOLFOX或CAPOX方案,联合或不联合曲妥珠单抗(如果HER2过度表达) [4] 。二线治疗方案为紫杉醇、多西他赛、伊立替康等单药化疗,或根据一线治疗方案 [5] [6] 的药物选择采用两药联合化疗方案。然而,由于GC的异质性,晚期胃癌常规化疗的中位总生存期(Overall Survival, OS)仅为8个月 [7] [8] [9] [10] [11] 。化疗药物的疗效似乎已经到达了瓶颈,而传统的靶向治疗药物进展缓慢,近期并无重大突破,尽管针对GC靶向药物的研究较多,但目前临床上仅有曲妥珠单抗等抗HER2药物和阿帕替尼等抗血管生成通路药物,AGC患者在分子水平上仍缺乏其他有效的靶向药物 [12] 。

免疫治疗作为一种近年来兴起的治疗手段,在晚期胃癌治疗中取得了令人鼓舞的进展,有多项相关的临床研究已开展或正在开展中,希望能进一步提高晚期胃癌的疗效及延长晚期胃癌患者的生存时间。ICIs在许多癌症类型中取得的重大进步以及在临床治疗上的成功应用促使我们探索ICIs在AGC中的效用。

随着免疫治疗的发展,多种免疫检查点被纳入实验研究中,并取得了不错的进展。同时,研究中的进步也为恶性胃癌患者的临床治疗提供了多种选择,譬如程序性死亡蛋白1 (programmed death-1, PD-1)、程序性死亡配体1 (programmed death-ligand 1, PD-L1)及细胞毒性T淋巴细胞相关抗原4 (anti-CTLA4 antibodies, CTLA-4 )等免疫检查点相关抑制剂目前已为较多临床工作者熟知。

2. 抗PD-1/PD-L1单克隆抗体

程序性死亡蛋白1 (programmed death-1, PD-1)主要表达于活化的T细胞、B细胞、树突状细胞、自然杀伤细胞上,它与表达于肿瘤细胞表面的程序性死亡配体1 (programmed death-ligand 1, PD-L1)相结合可使免疫细胞失活,并发生免疫抑制作用 [13] 。免疫检查点抑制剂,通过与PD-1结合,阻断与受体的结合,激活T细胞的功能,增强T细胞对肿瘤的免疫监视和杀伤能力,产生肿瘤免疫应答,恢复机体正常的抗肿瘤功能,从而实现肿瘤控制和肿瘤细胞清除 [14] 。近年来,PD-1抑制剂在多种晚期实体肿瘤(包括恶性黑色素瘤、非小细胞肺癌、头颈部鳞癌)中展现出较好疗效 [15] ,也在晚期胃癌治疗中展现出效果( [16] [17] , p. 2)。一些研究报道AGC患者对抗PD-1/PD-L1单克隆抗体表现出良好的反应率 [18] 。

2.1. 单药治疗

帕博利珠单抗是一种选择性、人源化、高亲和力的IgG4-κ单克隆抗体,旨在与PD-1结合,从而阻断PD-1与其配体之间的相互作用。

2016年开展的KEYNOTE-012 (NCT01848834)研究正式开启了晚期胃癌免疫治疗的探索之路。KEYNOTE-012,是一项多中心、开放性、1b期试验,旨在评估抗PD-1抗体帕博利珠单抗在PD-L1阳性的复发或转移性胃或胃食管结合部腺癌患者中的安全性和活性。帕博利珠单抗获得的11.4个月(95%CI 5.7~未达到)的中位OS长于单药化疗的4~8个月,与二线方案中联合方案获得的9~10个月OS相当 [16] 。

基于KEYNOTE-012中帕博利珠单抗取得的可观的抗肿瘤活性及可控的相关毒副作用,随即设计开展了II期临床试验——KEYNOTE-059,其中,帕博利珠单抗组的客观缓解率(Objective Response rate, ORR)为11.6% (95%CI,8.0%~16.1%;259例患者中30例),中位无进展生存期(Progression-Free Survival, PFS)为2.0个月(95%CI为2.0~2.1),中位OS为5.6个月(95%CI, 4.3~6.9),其结果表明,帕博利珠单抗单药治疗在既往至少接受2线治疗的晚期胃或胃食管结合部癌患者中显示出不错的活性和可管理的安全性 [19] 。

随后,一项随机、双盲、安慰剂对照、3期试验——ATTRACTION-2在日本开展。其中,纳武单抗组中位OS为5.26个月(95%CI 4.60~6.37),较安慰剂组中位生存期4.14个月(3.42~4.86)明显延长 [17] 。

基于KEYNOTE-012及ATTRACTION-2的研究结果,2017年9月,美国食品药品监督管理局(FDA)和中国国家药品监督管理局(NMPA)分别批准了帕博利珠单抗和纳武单抗作为复发局部晚期或转移性胃癌或胃食管结合部癌患者的三线及后线治疗的适应症 [20] 。

以上充满希望的结果,似乎说明PD-1/PD-L1抑制剂在晚期GC的治疗中显示出临床活性,然而,同期一项III期临床试验——KEYNOTE-061却显示,帕博利珠单抗治疗组对比化疗组,其患者在ORR及PFS上并未出现明显获益,这结果为研究者们带来了困惑。

同时,在JAVELIN 100研究中,阿维鲁单抗作单药治疗组作为晚期GC/GEJC一线治疗(奥沙利铂/氟尿嘧啶)后的维持治疗,在所有入组患者或PD-L1 TPS ≥ 1%的患者中,与继续化疗相比,未能改善OS ( [21] , p. 100)。此外,一项III期随机试——JAVELIN 300中显示,与化疗相比,在三线情况下使用单药阿维尤单抗治疗GC/GEJC患者并未改善OS或PFS [21] 。

综上,PD-1/PD-L1抑制剂单药治疗的获益明显受限,探讨PD-1/PD-L1抑制剂联合其他方案治疗似乎是一条新出路。目前针对PD-1/PD-L1抑制剂联合治疗的研究已有多项取得不错的结果。其传统的常见免疫治疗联合化疗,也有联合靶向治疗和放射治疗,或双免治疗,为晚期胃癌的治疗提供了不错的依据。

2.2. 联合治疗

PD-1/PD-L1抑制剂与标准化疗联合可能通过调节免疫系统和重塑肿瘤微环境发挥协同抗肿瘤活性,从而改善多种癌症的生存 [22] [23] [24] 。研究表明化疗药物和放疗调节肿瘤微环境的免疫状态,促进肿瘤抗原的释放 [25] [26] [27] 。这种方法可能与PD-1/PD-L1抑制剂在胃癌中产生了协同作用。而抗血管生成药物可以改变肿瘤免疫微环境,增强免疫治疗的疗效 [28] [29] [30] 。

在KEYNOTE-059实验中,队列二(帕博利珠单抗联合顺铂和5-FU或卡培他滨后)的结果提示,给予帕博利珠单抗联合化疗后,其总ORR为60%,客观缓解率为60.0% [95%CI, 38.7~78.9] (联合治疗)和25.8% (95%CI, 11.9~44.6) (单药治疗),中位PFS为6.6个月(95%CI 5.9~10.6),中位OS为13.8个月(95%CI 8.6~不可估计) [31] 。

基于KEYNOTE-059队列二的结果,一项Ⅲ期研究——KEYNOTE-062,报道了在PD-L1 CPS ≥ 1及CPS ≥ 10的AGC患者中比较一线帕博利珠单抗单药或帕博利珠单抗联合化疗与化疗的研究。最终分析,对于CPS ≥ 1或更高的患者中,帕博利珠单抗的OS不劣于化疗(mOS,10.6 vs 11.1个月,HR = 0.91 [99.2%CI, 0.69~1.18])。与化疗对比,在CPS ≥ 10或更高的患者中(mOS,17.4 vs 10.8个月,HR = 0.69 [95%CI, 0.49~0.97]),帕博利珠单抗延长了OS,但这种差异未经统计学检验。而在CPS ≥ 1 (mOS,12.5个月vs 11.1个月,HR = 0.85 [0.70~1.03],P = 0.046)和CPS ≥ 10 (12.3个月vs 10.8个月,HR = 0.85 [0.62~1.17],P = 0.158)人群中,化疗联合帕博利珠单抗组对OS的延长并不优于单纯化疗,尽管帕博利珠单抗联合化疗(49%,CPS ≥ 1时为37%)的ORR更高。在PD-L1 CPS ≥ 1或更高的人群中,帕博利珠单抗加化疗组(6.9个月,95%CI,5.7~7.3)对比单纯化疗组(6.4个月,95%CI,5.7~7.0)的中位PFS并不占优势 [32] 。

针对晚期胃癌一线免疫治疗探索的步伐并未停止,ATTRACTION-4是基于ATTRACTION-2对晚期胃癌三线治疗所取得的成功,探讨一线纳武单抗联合SOX方案及联合XELOX化疗的II/III期研究。纳武单抗 + SOX方案的ORR为57.1% (95%CI, 34.0~78.2),纳武单抗 + XELOX方案的ORR为76.5% (95%CI, 50.1~93.2),中位无进展生存期分别为9.7个月(5.8~NR)和10.6个月(5.6~12.5) [33] 。其三期部分结果表明,纳武利尤单抗联合化疗组在mOS (17.5个月vs 17.2个月,95%CI,0.75~1.08)并统计学无差异。其mPFS (10.5个月vs 8.3个月,95%CI,0.51~0.90)则显著改善 [34] 。

ATTRACTION-4是首个定位于亚洲人群的针对晚期胃癌的免疫治疗的大样本临床研究,并选择了普遍适用于亚洲人群的化疗方案。

2017年,一项多中心、随机、开放标签的3期试验——CheckMate-649进行开展,纳武单抗联合化疗显示出更好的OS,与单独化疗相比,PD-L1 CPS为≥5的患者中位OS (14.4个月[95%CI 13.1~16.2] vs 11.1个月[10.0~12.1])改善了3.3个月。在PD-L1 CPS为5个及以上≥5的患者中,纳武单抗联合化疗也提供了更好的PFS,与单纯化疗相比,纳武单抗联合化疗组中位PFS为7.7个月(95%CI 7.0~9.2),单纯化疗组中位PFS为6.0个月(5.6~6.9)。纳武单抗是第一个在既往未治疗的晚期胃、胃食管结合部或食管腺癌患者中,联合化疗与单独化疗相比,显示出更好的OS、PFS效益和可接受的安全性的PD-1抑制剂 [35] 。

随后,一项旨在评估帕博利珠单抗联合曲妥珠单抗 + 化疗一线治疗晚期HER2阳性胃腺癌或GEJ腺癌患者的疗效和安全性的III期试验——KEYNOTE-811报道了其第一次中期分析的可喜结果,在疗效人群中,研究中观察到,帕博利珠单抗组的ORR为74.4% (95%CI, 66.2~81.6),安慰剂组为51.9% (95%CI, 43.0~60.7)。这导致帕博利珠单抗组的客观缓解率提高了22.7%,具有统计学意义(95%CI, 11.2~33.7; P = 0.00006) [36] 。

基于该研究结果及之前对帕博利珠单抗和曲妥珠单抗加化疗的研究的发现,美国食品药品监督管理局批准帕博利珠单抗联合曲妥珠单抗和含氟嘧啶和铂类化疗,用于局部晚期或不可切除或转移性HER2阳性胃或胃食管交界腺癌的一线治疗。

2022年6月,基于ORIENT-16的研究结果,信迪利单抗被中国国家药监局(NMPA)批准用于一线治疗不可切除的局部晚期、复发性或转移性胃或胃食管交界处癌。

ORIENT-16是一项随机、双盲、III期临床研究,在2021年ESMO年会上,研究者对其结果进行口头报道,在所有患者中,信迪利单抗联合化疗组与单纯化疗组的OS和PFS分别为15.2 vs 12.3个月(HR 0.766; 95%CI: 0.626~0.936; P < 0.0090);7.1 vs 5.7个月(HR 0.636; 95%CI: 0.525~0.771; P < 0.0001)。在CPS评分 ≥ 5的患者中,信迪利单抗联合化疗组与单纯化疗组的OS和PFS分别为18.4 vs 12.9个月(HR 0.660; 95%CI: 0.525~0.771; P < 0.0001);7.7 vs 5.8个月(HR 0.628; 95%CI: 0.489~0.805; P = 0.0002) [37] 。

PD-1抑制剂联合治疗已有多种组合被推荐为晚期胃癌的一线治疗方案,但其联合方案的搭配仍有优化的空间,或许会对其治疗效果产生影响。目前有多项相关研究正在进行中,如帕博利珠单抗联合化疗的III期临床研究——KEYNOTE-859,替雷利珠单抗联合化疗的RA-TIONALE 305等,这些研究的结果值得我们期待。

3. 其他免疫检查点抑制剂

细胞毒性T淋巴细胞抗原-4 (CTL-associated protein 4, CTLA-4)是一种在T细胞表面表达的小分子。CTLA-4可与B7-1或B7-2结合来抑制T细胞表面的CD28与其配体B7-1或APC或肿瘤细胞上的B7-2结合提供的共刺激信号诱导T细胞活化的过程,该反应也称为共抑制过程 [38] 。伊匹木单抗是CTLA-4抑制剂,已经FDA批准用于治疗晚期黑色素瘤患者。另一项II期临床研究发现曲美木单抗在治疗晚期GC和GEJ腺癌中使得一名患者获得部分缓解。人源化CTLA-4单克隆抗体在恶性肿瘤中的获益还待更多样本量的研究来进行探索。

吲哚胺2,3-双加氧酶(Indoleamine 2,3-dioxygenase, IDO)涉及T细胞抑制。TIM3是一种选择性表达在辅助性T细胞1 (Th1)上的分子,可抑制Th1细胞与其配体半乳糖凝集素-9结合时的活化 [39] 。这些潜在的免疫检查点都需要更进一步的研究探索。

4. 结论

近年来,与ICIs有关的临床研究多次证实了其在恶性肿瘤中存在不小的获益,并具有一定的安全性。但由于晚期胃癌的高异质性,ICIs在不同分型的胃癌中显示出不同的效用。在本文中,我们回顾了ICIs近年来的研究进展及部分获得临床批准的PD-1/PD-L1单克隆抗体,并对ICIs获益相关的部分因素进行罗列,这使得我们对晚期胃癌中ICIs未来的发展充满信心。在将来的研究中,应建立更好的生物标志物,以选择最佳适配的晚期胃癌患者并取得最好的疗效,对于ICIs治疗的进一步研究和了解在未来将有助于优化晚期胃癌患者的治疗策略。

NOTES

*第一作者。

#通讯作者。

参考文献

[1] Sung, H., Ferlay, J., Siegel, R.L., Laversanne, M., Soerjomataram, I. and Jemal, A. (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] Kole, C., Charalampakis, N., Tsakatikas, S., Kouris, N., Papaxoinis, G. and Michalis, V.K. (2022) Immunotherapy for Gastric Cancer: A 2021 Update. Immunother-apy, 14, 41-64.
https://doi.org/10.2217/imt-2021-0103
[3] Kawazoe, A., Shitara, K., Boku, N., Yoshikawa, T. and Terashima, M. (2021) Current Status of Immunotherapy for Advanced Gastric Cancer. Japanese Journal of Clinical Oncology, 51, 20-27.
https://doi.org/10.1093/jjco/hyaa202
[4] Zhang, Y., Yang, Y., Chen, Y.R., Lin, W., Chen, X.L. and Liu, J. (2022) PD-L1: Biological Mechanism, Function, and Immunotherapy in Gastric Cancer. Frontiers in Immunology, 13, Article ID: 1060497.
https://doi.org/10.3389/fimmu.2022.1060497
[5] Eliza, H., Alicia, F.C.O., Demetris, P., Rao, S., Ashley, S. and Charalambous, H. (2011) Docetaxel and Irinotecan as Second-Line Therapy for Advanced Oesophagogastriccancer. Eu-ropean Journal of Cancer (Oxford, England : 1990), 47, 1146-1151.
https://doi.org/10.1016/j.ejca.2010.12.021
[6] Hironaka, S., Ueda, S., Yasui, H., Nishina, T., Tsuda, M. and Tsumura, T. (2013) Randomized, Open-Label, Phase III Study Comparing Irinotecan with Paclitaxel in Patients with Advanced Gastric Cancer without Severe Peritoneal Metastasis after Failure of Prior Combination Chemotherapy Using Fluoropyrimidine plus Platinum: WJOG 4007 Trial. Journal of Clinical Oncology: Official Journal of the American Soci-ety of Clinical Oncology, 31, 4438-4444.
https://doi.org/10.1200/JCO.2012.48.5805
[7] Zhao, Q., Lian, C.H., Huo, Z.B., Li, M., Liu, Y. and Fan, L.Q. (2020) The Efficacy and Safety of Neoadjuvant Chemotherapy on Patients with Advanced Gastric Cancer: A Multicenter Randomized Clinical Trial. Cancer Medicine, 9, 5731-5745.
https://doi.org/10.1002/cam4.3224
[8] Tian, Y., Zhao, Q., Li, Y., Fan, L.Q., Zhang, Z.D. and Zhao, X.F. (2021) Efficacy of Neoadjuvant Chemotherapy DOX and XELOX Regimens for Patients with Resectable Gastric or Gastroesophageal Junction Adenocarcinoma. Gastroenterology Re-search and Practice, 2021, Article ID: 5590626.
https://doi.org/10.1155/2021/5590626
[9] Yu, Y.Y., Fang, Y., Shen, Z.B., Wang, Y., Yan, M. and Cao, H. (2019) Oxaliplatin plus Capecitabine in the Perioperative Treatment of Lo-cally Advanced Gastric Adenocarcinoma in Combination with D2 Gastrectomy: NEO-CLASSIC Study. The Oncologist, 24, 1311-e989.
https://doi.org/10.1634/theoncologist.2019-0416
[10] Xue, K., Ying, X.J., Bu, Z.D., Wu, A., Li, Z.W. and Tang, L. (2018) Oxaliplatin plus S-1 or Capecitabine as Neoadjuvant or Adjuvant Chemotherapy for Locally Advanced Gastric Cancer with D2 Lymphadenectomy: 5-Year Follow-Up Results of a Phase II-III Randomized Trial. Chinese Journal of Cancer Research, 30, 516-525.
https://doi.org/10.21147/j.issn.1000-9604.2018.05.05
[11] Leowattana, W., Leowattana, P. and Leowattana, T. (2023) Immunotherapy for Advanced Gastric Cancer. World Journal of Methodology, 13, 79-97.
https://doi.org/10.5662/wjm.v13.i3.79
[12] Wang, J.Z., He, Y.D., Zhang, B.W., Lv, H.F., Nie, C.Y. and Chen, B.B. (2022) The Efficacy and Safety of Sintilimab Combined with Nab-Paclitaxel as a Second-Line Treatment for Advanced or Metastatic Gastric Cancer and Gastroesophageal Junction Cancer. Frontiers in Oncology, 12, Article ID: 924149.
https://doi.org/10.3389/fonc.2022.924149
[13] Jin, X., Liu, Z.R., Yang, D.X., Yin, K. and Chang, X.S. (2022) Recent Progress and Future Perspectives of Immunotherapy in Advanced Gastric Cancer. Frontiers in Immunology, 13, Article ID: 948647.
https://doi.org/10.3389/fimmu.2022.948647
[14] Zhang, L., Mai, W.Q., Jiang, W.Y. and Geng, Q. (2020) Sintili-mab: A Promising Anti-Tumor PD-1 Antibody. Frontiers in Oncology, 10, Article ID: 594558.
https://doi.org/10.3389/fonc.2020.594558
[15] Sun, L.T., Zhang, L.Y., Yu, J.R., Zhang, Y., Pang, X. and Ma, C.H. (2020) Clinical Efficacy and Safety of Anti-PD-1/ PD-L1 Inhibitors for the Treatment of Advanced or Metastatic Cancer: A Systematic Review and Meta-Analysis. Scientific Reports, 10, Article No. 2083.
https://doi.org/10.1038/s41598-020-58674-4
[16] Muro, K., Chung, H.C., Shankaran, V., Geva, R., Catenacci, D. and Gupta, S. (2016) Pembrolizumab for Patients with PD-L1-Positive Advanced Gastric Cancer (KEYNOTE-012): A Multicentre, Open-Label, Phase 1b Trial. The Lancet Oncology, 17, 717-726.
https://doi.org/10.1016/S1470-2045(16)00175-3
[17] Kang, Y.-K., Boku, N., Satoh, T., Ryu, M.-H., Chao, Y. and Kato, K. (2017) Nivolumab in Patients with Advanced Gastric or Gastro-Oesophageal Junction Cancer Refractory to, or Intolerant of, at Least Two Previous Chemotherapy Regimens (ONO-4538-12, ATTRACTION-2): A Randomised, Double-Blind, Placebo-Controlled, Phase 3 Trial. The Lancet (London, England), 390, 2461-2471.
https://doi.org/10.1016/S0140-6736(17)31827-5
[18] Zhang, Z., Liu, N. and Sun, M. (2023) Research Progress of Immunotherapy for Gastric Cancer. Technology in Cancer Research & Treatment, 22.
https://doi.org/10.1177/15330338221150555
[19] Fuchs, C.S., Doi, T., Jang, R.W., Muro, K., Satoh, T. and Ma-chado, M. (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.
https://doi.org/10.1001/jamaoncol.2018.0013
[20] Kexin, L., Zhang, A., Li, X., Zhang, H.T. and Zhao, L.M. (2021) Advances in Clinical Immunotherapy for Gastric Cancer. Biochimica et Biophysica Acta (BBA)—Reviews on Cancer, 1876, Article ID: 188615.
https://doi.org/10.1016/j.bbcan.2021.188615
[21] Bang, Y.-J., Ruiz, E.Y., Cutsem, E.V., Lee, K.-W., Wyrwicz, L. and Schenker, M. (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 Oncology: Official Journal of the European Society for Medical Oncology, 29, 2052-2060.
https://doi.org/10.1093/annonc/mdy264
[22] Kon, E. and Benhar, I. (2019) Immune Checkpoint Inhib-itor Combinations: Current Efforts and Important Aspects for Success. Drug Resistance Updates, 45, 13-29.
[23] Davern, M. and Lysaght, J. (2020) Cooperation between Chemotherapy and Immunotherapy in Gas-troesophageal Cancers. Cancer Letters, 495, 89-99.
https://doi.org/10.1016/j.canlet.2020.09.014
[24] Burtness, B., Harrington, K.J., Greil, R., Soulières, D., Tahara, M. and Castro Jr., G.D. (2019) Pembrolizumab Alone or with Chemo-therapy versus Cetuximab with Chemotherapy for Recurrent or Metastatic Squamous Cell Carcinoma of the Head and Neck (KEYNOTE-048): A Randomised, Open-Label, Phase 3 Study. The Lancet (London, England), 394, 1915-1928.
https://doi.org/10.1016/S0140-6736(19)32591-7
[25] Hodge, J.W., Garnett, C.T., Farsaci, B., Palena, C., Tsang, K.-Y. and Ferrone, S. (2013) Chemotherapy-Induced Immunogenic Modulation of Tumor Cells Enhances Killing by Cytotoxic T Lymphocytes and Is Distinct from Immunogenic Cell Death. International Journal of Cancer, 133, 624-636.
https://doi.org/10.1002/ijc.28070
[26] Li, J.-Y., Chen, Y.-P., Li, Y.-Q., Liu, N. and Ma, J. (2021) Chemotherapeu-tic and Targeted Agents Can Modulate the Tumor Microenvironment and Increase the Efficacy of Immune Checkpoint Blockades. Molecular Cancer, 20, Article No. 27.
https://doi.org/10.1186/s12943-021-01317-7
[27] Seliger, B. (2019) Combinatorial Approaches with Checkpoint Inhibitors to Enhance Anti-Tumor Immunity. Frontiers in Immunol-ogy, 10, Article No. 999.
https://doi.org/10.3389/fimmu.2019.00999
[28] Leong, A. and Kim, M. (2020) The An-giopoietin-2 and TIE Pathway as a Therapeutic Target for Enhancing Antiangiogenic Therapy and Immunotherapy in Pa-tients with Advanced Cancer. International Journal of Molecular Sciences, 21, Article No. 8689.
https://doi.org/10.3390/ijms21228689
[29] Zhou, S. and Zhang, H. (2020) Synergies of Targeting Angiogenesis and Immune Checkpoints in Cancer: From Mechanism to Clinical Applications. Anti-Cancer Agents in Medicinal Chem-istry, 20, 768-776.
https://doi.org/10.2174/1871520620666200207091653
[30] Zhu, N., Weng, S., Wang, J., Chen, J.Q., Yu, L.Z. and Fang, X.F. (2019) Preclinical Rationale and Clinical Efficacy of Antiangiogenic Therapy and Immune Checkpoint Blockade Combination Therapy in Urogenital Tumors. Journal of Cancer Research and Clinical Oncology, 145, 3021-3036.
https://doi.org/10.1007/s00432-019-03044-5
[31] Bang, Y.-J., Kang, Y.-K., Catenacci, D.V., Muro, K., Fuchs, C.S. and Geva, R. (2019) Pembrolizumab Alone or in Combination with Chemotherapy as First-Line Therapy for Patients with Advanced Gastric or Gastroesophageal Junction Adenocarcinoma: Results from the Phase II Nonrandom-ized KEYNOTE-059 Study. Gastric Cancer, 22, 828-837.
https://doi.org/10.1007/s10120-018-00909-5
[32] Shitara, K., Cutsem, E.V., Bang, Y.-J., Fuchs, C., Wyrwicz, L. and Lee, K.-W. (2020) Efficacy and Safety of Pembrolizumab or Pembrolizumab plus Chemotherapy vs Chemotherapy 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
[33] Boku, N., Ryu, M.-H., Kato, K., Chung, H.C., Minashi, K. and Lee, K.-W. (2019) Safety and Efficacy of Nivolumab in Combination with S-1/Capecitabine plus Oxaliplatin in Patients with Previously Untreated, Unresectable, Advanced, or Recurrent Gastric/Gastroesophageal Junction Cancer: Interim Results of a Randomized, Phase II Trial (ATTRACTION- 4). Annals of Oncology, 30, 250-258.
https://doi.org/10.1093/annonc/mdy540
[34] Kang, Y.-K., Chen, L.-T., Ryu, M.-H., Oh, D.-Y., Oh, S.C. and Chung, H.C. (2022) Nivolumab plus Chemotherapy versus Placebo plus Chemotherapy in Patients with HER2-Negative, Untreated, Unresectableadvanced or Recurrent Gastric or Gastro-Oesophageal 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
[35] Janjigian, Y.Y., Shitara, K., Moehler, M., Garrido, M., Sal-man, P. and Shen, L. (2021) First-Line Nivolumab plus Chemotherapy versus Chemotherapy Alone for Advanced Gas-tric, Gastro-Oesophageal Junction, and Oesophageal Adenocarcinoma (CheckMate 649): A Randomised, Open-Label, Phase 3 Trial. The Lancet (London, England), 398, 27-40.
https://doi.org/10.1016/S0140-6736(21)00797-2
[36] Chung, H.C., Bang, Y.-J., Fuchs, C.S., Qin, S.-K., Satoh, T. and Shitara, K. (2021) First-Line Pembrolizumab/Placebo plus Trastuzumab and Chemotherapy in HER2-Positive Ad-vanced Gastric Cancer: KEYNOTE-811. Future Oncology (London, England), 17, 491-501.
https://doi.org/10.2217/fon-2020-0737
[37] Xu, J., Jiang, H., Pan, Y., Gu, K., Cang, S. and Han, L. (2021) LBA53 Sintilimab plus Chemotherapy (Chemo) versus Chemo as First-Line Treatment for Advanced Gastric or Gastroesophage-al Junction (G/GEJ) Adenocarcinoma (ORIENT- 16): First Results of a Randomized, Double-Blind, Phase III Study. Annals of Oncology, 32, S1331.
https://doi.org/10.1016/j.annonc.2021.08.2133
[38] Grierson, P., Lim, K.-H. and Amin, M. (2017) Immunotherapy in Gastrointestinal Cancers. Journal of Gastrointestinal Oncology, 8, 474-484.
https://doi.org/10.21037/jgo.2017.05.01
[39] Karlsson, M., Marits, P., Dahl, K., Dagöö, T., Enerbäck, S. and Thörn, M. (2010) Pilot Study of Sentinel-Node-Based Adoptive Immunotherapy in Advanced Colorectal Cancer. Annals of Surgical Oncology, 17, 1747-1757.
https://doi.org/10.1245/s10434-010-0920-8