晚期胃癌免疫治疗的现状
The Current Status of Immunotherapy in Advanced Gastric Cancer
DOI: 10.12677/jcpm.2024.32048, PDF, HTML, XML, 下载: 22  浏览: 39 
作者: 任闪闪:青海大学临床医学院,青海 西宁;姬发祥*:青海大学附属医院肿瘤科,青海 西宁
关键词: 胃癌免疫治疗免疫检查点抑制剂过继细胞疗法肿瘤疫苗Gastric Cancer Immunotherapy Immune Checkpoint Inhibitors Adoptive Cell Therapy Tumor Vaccine
摘要: 作为最常见的实体瘤之一,胃癌已被证明是全球第三大死因。胃癌的症状通常不明显,因此很难在早期发现。因此,胃癌一旦在患者中被发现,就已经处于晚期,由于治疗无效和多重耐药,预后较差。最近在了解癌症微环境方面的进展显著促进了晚期胃癌免疫疗法的发展。免疫疗法可以诱导胃癌患者的免疫反应,从而导致癌细胞的破坏。与传统疗法相比,免疫疗法已显示出强大的疗效和可耐受的毒性。因此,这种治疗晚期胃癌的新策略越来越受欢迎。本文综述了免疫治疗晚期胃癌的最新进展,如免疫检查点抑制剂、过继细胞疗法、VEGF抑制剂、癌症疫苗和CAR-T细胞疗法等。
Abstract: As one of the most common solid tumors, gastric cancer has been proven to be the third leading cause of death worldwide. The symptoms of gastric cancer are usually not obvious, so it is difficult to detect them early. Therefore, once gastric cancer is detected in patients, it is already in the advanced stage, and due to ineffective treatment and multiple drug resistance, the prognosis is poor. The recent progress in understanding the cancer microenvironment has significantly promoted the development of immunotherapy for advanced gastric cancer. Immunotherapy can induce an immune response in gastric cancer patients, leading to the destruction of cancer cells. Compared with traditional therapies, immunotherapy has shown strong efficacy and tolerable toxicity. Therefore, this new strategy for treating advanced gastric cancer is becoming increasingly popular. This article reviews the latest advances in immunotherapy for advanced gastric cancer, such as immune checkpoint inhibitors, adoptive cell therapy, VEGF inhibitors, cancer vaccines, and CAR-T cell therapy.
文章引用:任闪闪, 姬发祥. 晚期胃癌免疫治疗的现状[J]. 临床个性化医学, 2024, 3(2): 328-334. https://doi.org/10.12677/jcpm.2024.32048

1. 引言

胃癌是癌症相关死亡的第三大原因 [1] ,由于诊断延迟和缺乏有效治疗,晚期胃癌患者预后不良,寿命短约一年 [2] 。晚期胃癌常用的治疗方法有放疗、化疗和靶向治疗 [3] 。化疗作为晚期胃癌的一种常规治疗,氟尿嘧啶类、铂类和紫衫类药物是晚期胃癌的主要化疗药物。通常一线化疗方案以氟尿嘧啶类药物为基础,联合铂类、和/或紫衫类药物组成两药或三药化疗方案 [4] 。虽然化疗对胃癌患者的疗效逐渐提高,但是其生存期依旧很短。靶向治疗是继传统全身化疗后的治疗方法,目前有曲妥珠单抗、雷莫芦单抗、阿帕替尼被批准用于治疗晚期胃癌。然而,靶向治疗策略提高了临床疗效,但靶向治疗药物筛查受益人群的难度以及耐药倾向 [5] ,使胃癌患者的预后并未得到实质性改善。

近年来,免疫疗法已成为治疗晚期胃癌的新型疗法,并因其惊人的抗肿瘤功效而迅速引起了世界各地研究人员的关注。对肿瘤微环境的更好理解极大地促进了晚期胃癌免疫疗法的发展。针对晚期胃癌应用最广泛的免疫疗法,包括免疫检查点抑制剂(ICI)、过继细胞疗法、癌症疫苗、血管内皮生长因子A (VEGFA)抗体和嵌合抗原受体(CAR) T疗法等 [6] 。研究表明,抗PD-1/PD-L1抗体等ICI可通过激活免疫应答有效杀死癌细胞。ICIs的临床试验已经显示出对癌症患者的疗效和安全性。

在这篇综述中,我们描述了用于治疗晚期胃癌的免疫疗法的最新发展,重点介绍了ICI、过继细胞疗法、癌症疫苗和CAR-T细胞疗法的最新进展。此外,我们还讨论了免疫疗法当前面临的挑战,以及克服这些局限性的潜在策略,例如免疫疗法和靶向疗法的结合。

2. 晚期胃癌的免疫治疗

在过去的几年里,对胃癌免疫机制更好理解极大地促进了新型免疫疗法的发展。ICI可有效中断免疫检查点相互作用,通过激活宿主的免疫系统导致肿瘤细胞的破坏 [7] ,例如过继细胞疗法、VEGF抑制剂、癌症疫苗和CART细胞疗法也显示出有效的抗肿瘤活性 [8] 。免疫治疗的这些成就标志着晚期胃癌治疗的新时代。

2.1. 免疫检查点抑制剂

2011年,ipilimumab成为世界上第一个获批用于治疗黑色素瘤的ICI [9] 。从那时起,免疫疗法彻底改变了晚期胃癌治疗的策略。ICI主要有三种类型:抗PD1/PD-L1和抗CTLA-4抗体。活化的免疫细胞(如T细胞)可以表达PD-1,PD-L1是PD-1的配体,与PD-1结合,从而导致免疫细胞凋亡和免疫抑制。PD-L1在晚期胃癌中过表达,导致肿瘤细胞逃避免疫应答。另一方面,CTLA-4蛋白可高亲和力与B7-1/B7-2相互作用,从而抑制CD28信号转导通路,从而在T细胞活化中发挥关键作用。靶向这些免疫检查点的抑制剂已经在临床前和临床试验中产生和研究。

2.1.1. PD-1抑制剂

PD-1抑制剂纳武利尤单抗是一种单克隆抗体,已于2014年获得FDA批准用于晚期胃肿瘤治疗 [10] 。纳武利尤单抗对晚期胃癌的影响通过在亚洲40多个国家/地区进行的III期临床试验进行了检查。初步结果显示,与安慰剂相比,纳武利尤单抗可以显着提高患者的生存率,纳武利尤单抗治疗胃肿瘤患者的12个月总生存率为26.2%,而安慰剂治疗的生存率为10.9%,这表明对这一预后不良人群有希望治愈。值得注意的是,纳武利尤单抗已被批准用于临床应用,作为治疗晚期和复发性胃癌的新方 [11] 。

帕博利珠单抗是另一种有前途的靶向PD-1的抑制剂。帕博利珠单抗的疗效已在晚期胃癌的II期试验中得到评估,帕博利珠单抗治疗晚期胃癌患者的11个月总生存率为45.8%。除了其高抗肿瘤活性外,帕博利珠单抗还显示出适度的副作用。帕博利珠单抗的这些优势促使其在2017年被批准用于治疗晚期胃肿瘤 [12] 。已在592例不可手术的晚期胃癌患者中进行pembrolizumab的临床试验,以确定pembrolizumab 与紫杉醇相比的疗效。然而,与紫杉醇相比,单独使用帕博利珠单抗并未显示出患者生存率的显着提高,当帕博利珠单抗与紫杉醇联合使用时,检测到增强的抗肿瘤作用和更好的耐受性 [13] 。替雷利珠单抗还因其对晚期胃癌的抗肿瘤作用而被评估,为基于PD-1的免疫疗法在晚期胃肿瘤中的发展提供了希望。

2.1.2. PD-L1抑制剂

PD-L1在各种癌细胞上过表达,并在T细胞抑制中发挥关键作用。众所周知的PD-L1抑制剂包括 avelumab、durvalumab和atezolizumab [14] 。Avelumab是一种抗PD-L1单克隆抗体,在晚期胃癌患者的III期试验中显示出良好的耐受性 [15] 。Avelumab在日本患者中表现出较高的总体缓解率和生存率。此外,avelumab与其他疗法联合使用可增强对晚期胃癌的疗效。然而,一项针对晚期胃癌的I期试验表明,阿替利珠单抗对171例患者中的1例有效 [6] 。该临床试验的反应率与PD-L1表达密切相关,PD-L1抑制剂导致晚期胃癌的机制可能是PD-L1抑制可激活DC细胞、T淋巴细胞和自然杀伤细胞,从而导致胃肿瘤破坏。

2.1.3. CTLA-4抑制剂

CTLA-4在人体免疫系统中起着重要作用。CTLA-4与CD28同源,但它可以与B7-1/B7-2相互作用,亲和力更高。因此,CTLA-4可以调节甚至抑制CD28信号传导。CTLA-4抑制剂tremelimumab和ipilimumab已在晚期胃癌的临床试验中进行了评估 [6] 。在一项针对晚期胃癌患者的II期试验中对ipilimumab进行了评估 [16] 。然而,这项研究被终止,因为与一线靶向药物相比,ipilimumab没有显示出显着提高的生存率。一项针对12名不可手术的晚期胃癌患者的临床研究表明,与同时使用tremelimumab和其他抗癌药物的联合治疗相比,tremelimumab的反应率中等。值得注意的是,靶向CTLA-4和PD-1的联合疗法显示出增强的抗肿瘤免疫力 [17] 。ipilimumab和nivolumab的联合疗法已被批准用于治疗晚期胃癌。然而,CTLA-4抑制剂作为晚期胃癌单药治疗的疗效仍有待进一步研究。

2.1.4. PD-1/CTLA-4双特异性抗体

卡度尼利单抗(AK104)是全球首个进入临床试验的PD-1、CTLA-4双特异性抗体,在早期试验中体现出良好的疗效和安全性 [18] 。2023年更新了AK104联合化疗用于一线治疗胃癌、GEJCⅠb、Ⅱ期临床研究的2年随访数据,联合治疗组全人群中位OS时间达17.41个月,中位PFS时间达9.2个月,12个月OS率为61.4%,ORR达68.2%,疾病控制率(Disease Control Rate, DCR)达92%;在PD-L1 CPS ≥ 5人群中,中位OS时间高达20.24个月。在中国真实世界的晚期胃癌患者中,PD-L1低表达(CPS 1~4)人群或表达阴性(CPS < 1)患者的人群占比分别约为13%和38%,超过全体患者人群的50%以上 [19] 。目前的PD-1抑制剂联合化疗作为一线疗法对这类患者带来的OS和PFS益处仍相对有限,中位OS时间为10~12个月,而该项Ⅱ期研究中AK104联合化疗在PD-L1低表达或阴性患者中仍体现出卓著的疗效。该试验入组的PD-L1表达阴性(CPS < 1)人群占比 > 50%,PD-L1高表达人群(CPS ≥ 5)占比仅15%;亚组分析中,PD-L1 CPS < 1人群中位OS时间达17.64个月,中位PFS时间为8.18个月。这提示双免药物的出现有望为这类患者带来更高效的临床治疗手段,而这样的双免治疗是否优于既往PD-1、PD-L1抑制剂联合化疗一线治疗是特别值得期待的。近期,AK104联合XELOX方案化疗用于一线治疗晚期胃癌、GEJC的Ⅲ期临床试验(AK104-302)也宣布其中期分析达到主要研究终点OS,相关数据有待进一步公开。

2.2. 过继细胞疗法

胃癌细胞可以表达具有高免疫原性的特异性新抗原,从而导致人体免疫系统的激活 [3] 。通过这种方式,癌细胞可以被识别和破坏。然而,癌细胞可产生抑制因子,包括淋巴细胞活化基因3 (LAG-3)、TGF-β、前列腺素E2和IL-10,这些因子会抑制免疫应答,从而逃避免疫系统的检测和清除 [20] 。对于免疫系统无法检测癌细胞并对其做出反应的患者,过继细胞疗法已被证明是治疗晚期胃癌的有效策略 [21] 。过继细胞疗法利用各种免疫细胞,包括肿瘤浸润淋巴细胞(TIL)、淋巴因子激活的杀伤细胞和细胞因子诱导的杀伤(CIK)细胞来诱导清除癌细胞的有效免疫。

TILs免疫疗法已广泛应用于晚期胃癌。特别是,来自胃癌患者的TILs已经暴露于肿瘤特异性抗原,因此在免疫治疗中极为有利。胃癌患者过继细胞疗法的临床试验表明,与单独使用传统疗法相比,基于肿瘤相关淋巴细胞的联合疗法可以将存活率提高到50% [22] 。此外,近年来,扩增的同种异体自然杀伤细胞也被用作治疗晚期胃癌的新型免疫疗法 [23] 。天然杀伤细胞具有高抗肿瘤活性和抗体依赖性细胞毒性。然而,由于缺乏获得大量功能性自然杀伤细胞的策略,天然杀伤细胞在癌症治疗中的临床应用受到严重限制 [24] 。将进行进一步的研究,以研究产生足够的天然杀伤细胞用于癌症免疫治疗的新方法。

2.3. 抗血管生成治疗

血管内皮生长因子A (VEGFA)通过参与新血管的形成(这一过程称为血管生成)在胃癌的发展中发挥重要作用 [25] 。VEGFA在调节癌症免疫应答中发挥作用,这可能导致肿瘤细胞从免疫系统的监视中逃逸。此外,VEGF可以促进Treg细胞向肿瘤部位的转移。在晚期胃癌患者中使用VEGFA抑制剂和免疫检查点抑制剂联合治疗的临床研究显示出有希望的效果,具有增强的抗肿瘤作用和降低毒性。例如,贝伐珠单抗和雷莫西尤单抗可显著防止血管生成 [26] 。在晚期胃癌患者中使用贝伐珠单抗和ICI (如阿替利珠单抗、雷莫西尤单抗、度伐利尤单抗)联合治疗的临床研究显示出良好的疗效 [26] 。这些研究表明,使用VEGFA抑制剂和靶向PD-1或PD-L1的ICI的联合治疗可能为晚期胃癌的有效治疗方法的发展提供启示。

2.4. 癌症疫苗

晚期胃癌的另一种新型免疫疗法是癌症疫苗的应用,它可以激活体内针对肿瘤细胞的免疫反应 [6] 。蛋白质和肽是触发免疫反应的常用抗原。研究最充分的癌症疫苗是mRNA疫苗,它携带抗原的遗传信息,可以快速将其转化为蛋白质以诱导免疫应答,从而导致癌细胞的破坏。研究表明,与传统化疗或靶向治疗相比,mRNA癌症疫苗显示出很强的疗效和适度的副作用 [27] 。此外,在初步临床试验中,癌症疫苗和化疗(如顺铂和5-氟尿嘧啶)的组合对肿瘤细胞的细胞毒性显着增强 [28] 。一项针对HLA-A24和HLA-A2肽的临床研究检查了胃癌患者的外周血单核细胞 [29] 。结果显示,接受癌症疫苗治疗的患者中有50%对接种疫苗的肽的体液和细胞反应增加。

2.5. CAR-T细胞疗法

CAR-T细胞专门设计用于表达合成受体,这些受体可诱导T细胞检测特定的癌症抗原,从而通过宿主的免疫破坏肿瘤细胞 [30] 。claudin 18.2 (CLDN 18.2)、人表皮生长因子受体2 (HER2)、粘蛋白1、自然杀伤受体组2 (NKG2D)、上皮细胞粘附分子(EpCAM)、间皮素(MSLN)和癌胚抗原(CEA)等生物标志物在胃癌的诊断和功能中发挥重要作用 [31] 。研究表明,CAR-T疗法可以有效地靶向上述生物标志物来治疗晚期胃癌 [32] 。

3. 小结

在过去的几十年里,癌症免疫疗法已成为治疗各种癌症的有前途的疗法。ICIs的开发已成为晚期胃癌的突破,并在患者中显示出抗肿瘤作用。然而,免疫检查点抑制的毒性和有效性在很大程度上限制了其广泛的临床应用。其他免疫疗法,包括过继细胞疗法、癌症疫苗和CAR-T细胞疗法,在胃癌患者中也显示出抗肿瘤活性。免疫疗法联合靶向疗法的临床试验显示,与单独使用免疫疗法相比,免疫疗法具有更高的抗肿瘤活性和生存率。尽管免疫疗法在晚期胃癌中具有优势,但中等临床疗效和免疫逃避等挑战阻碍了免疫疗法在晚期胃癌中的广泛应用。克服这些挑战的新策略将涉及CAR-T疗法和ICI的结合,利用免疫调节剂来避免免疫抑制。我们相信,开发新型免疫疗法可能会为晚期胃癌的治疗提供启示。

NOTES

*通讯作者。

参考文献

[1] Ferlay, J., Colombet, M., Soerjomataram, I., et al. (2019) Estimating the Global Cancer Incidence and Mortality in 2018: GLOBOCAN Sources and Methods. International Journal of Cancer, 144, 1941-1953.
https://doi.org/10.1002/ijc.31937
[2] Högner, A. and Moehler, M. (2022) Immunotherapy in Gastric Cancer. Current Oncology (Toronto, Ont.), 29, 1559-1574.
https://doi.org/10.3390/curroncol29030131
[3] Jin, X., Liu, Z., Yang, D., et al. (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
[4] Koizumi, W., et al. (2008) S-1 plus Cisplatin versus S-1 Alone for First-Line Treatment of Advanced Gastric Cancer (SPIRITS Trial): A Phase III Trial. The Lancet Oncology, 9, 215-221.
https://doi.org/10.1016/S1470-2045(08)70035-4
[5] Kawazoe, A., Fukuoka, S., Nakamura, Y., et al. (2020) Lenvatinib plus Pembrolizumab in Patients with Advanced Gastric Cancer in the First-Line or Second-Line Setting (EPOC1706): An Open-Label, Single-Arm, Phase 2 Trial. The Lancet Oncology, 21, 1057-1065.
https://doi.org/10.1016/S1470-2045(20)30271-0
[6] Xie, J., Fu, L. and Jin, L. (2021) Immunotherapy of Gastric Cancer: Past, Future Perspective and Challenges. Pathology, Research and Practice, 218, Article ID: 153322.
https://doi.org/10.1016/j.prp.2020.153322
[7] Postow, M.A., Callahan, M.K. and Wolchok, J.D. (2015) Immune Checkpoint Blockade in Cancer Therapy. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology, 33, 1974-1982.
https://doi.org/10.1200/JCO.2014.59.4358
[8] Kawazoe, A., Shitara, K., Boku, N., et al. (2021) Current Status of Immunotherapy for Advanced Gastric Cancer. Japanese Journal of Clinical Oncology, 51, 20-27.
https://doi.org/10.1093/jjco/hyaa202
[9] Robert, C., Thomas, L., Bondarenko, I., et al. (2011) Ipilimumab plus Dacarbazine for Previously Untreated Metastatic Melanoma. The New England Journal of Medicine, 364, 2517-2526.
https://doi.org/10.1056/NEJMoa1104621
[10] Kato, K., et al. (2019) A Subanalysis of Japanese Patients in a Randomized, Double-Blind, Placebo-Controlled, Phase 3 Trial of Nivolumab for Patients with Advanced Gastric or Gastro-Esophageal Junction Cancer Refractory to, or Intolerant of, at Least Two Previous Chemotherapy Regimens (ONO-4538-12, ATTRACTION-2). Gastric Cancer: Official Journal of the International Gastric Cancer Association and the Japanese Gastric Cancer Association, 22, 344-354.
[11] Doi, T., et al. (2019) Phase 1 Trial of Avelumab (Anti-PD-L1) in Japanese Patients with Advanced Solid Tumors, Including Dose Expansion in Patients with Gastric or Gastroesophageal Junction Cancer: The JAVELIN Solid Tumor JPN Trial. Gastric Cancer: Official Journal of the International Gastric Cancer Association and the Japanese Gastric Cancer Association, 22, 817-827.
https://doi.org/10.1007/s10120-018-0903-1
[12] 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.
https://doi.org/10.1001/jamaoncol.2018.0013
[13] Shitara, K., et al. (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, 1-10.
[14] Doroshow, D.B., Bhalla, S., Beasley, M.B., et al. (2021) PD-L1 as a Biomarker of Response to Immune-Checkpoint Inhibitors. Nature Reviews. Clinical Oncology, 18, 345-362.
https://doi.org/10.1038/s41571-021-00473-5
[15] 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 Oncology: Official Journal of the European Society for Medical Oncology, 29, 2052-2060.
https://doi.org/10.1093/annonc/mdy264
[16] Bang, Y.J., Cho, J.Y., Kim, Y.H., et al. (2017) Efficacy of Sequential Ipilimumab Monotherapy versus Best Supportive Care for Unresectable Locally Advanced/Metastatic Gastric or Gastroesophageal Junction Cancer. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research, 23, 5671-5678.
https://doi.org/10.1158/1078-0432.CCR-17-0025
[17] 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 Junction Adenocarcinoma. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research, 26, 846-854.
https://doi.org/10.1158/1078-0432.CCR-19-2443
[18] Pang, X., et al. (2023) Cadonilimab, a Tetravalent PD-1/CTLA-4 Bispecific Antibody with Trans-Binding and Enhanced Target Binding Avidity. MAbs, 15, Article ID: 2180794.
https://doi.org/10.1080/19420862.2023.2180794
[19] Zhang, L., Wang, Y., Li, Z., et al. (2021) Clinicopathological Features of Tumor Mutation Burden, Epstein-Barr Virus Infection, Microsatellite Instability and PD-L1 Status in Chinese Patients with Gastric Cancer. Diagnostic Pathology, 16, Article No. 38.
https://doi.org/10.1186/s13000-021-01099-y
[20] Gao, X., Mi, Y., Guo, N., et al. (2017) Cytokine-Induced Killer Cells as Pharmacological Tools for Cancer Immunotherapy. Frontiers in Immunology, 8, Article No. 774.
https://doi.org/10.3389/fimmu.2017.00774
[21] Moreno, V., Hernandez, T., De Miguel, M., et al. (2021) Adoptive Cell Therapy for Solid Tumors: Chimeric Antigen Receptor T Cells and Beyond. Current Opinion in Pharmacology, 59, 70-84.
https://doi.org/10.1016/j.coph.2021.05.004
[22] Shen, X., et al. (2007) Persistence of Tumor Infiltrating Lymphocytes in Adoptive Immunotherapy Correlates with Telomere Length. Journal of Immunotherapy (Hagerstown, Md.: 1997), 30, 123-129.
https://doi.org/10.1097/01.cji.0000211321.07654.b8
[23] Cao, B., Liu, M., Huang, J., et al. (2021) Development of Mesothelin-Specific CAR NK-92 Cells for the Treatment of Gastric Cancer. International Journal of Biological Sciences, 17, 3850-3861.
https://doi.org/10.7150/ijbs.64630
[24] Ishikawa, T., Okayama, T., Sakamoto, N., et al. (2018) Phase I Clinical Trial of Adoptive Transfer of Expanded Natural Killer Cells in Combination with IgG1 Antibody in Patients with Gastric or Colorectal Cancer. International Journal of Cancer, 142, 2599-2609.
https://doi.org/10.1002/ijc.31285
[25] Gabrilovich, D.I., Chen, H.L., Girgis, K.R., et al. (1996) Production of Vascular Endothelial Growth Factor by Human Tumors Inhibits the Functional Maturation of Dendritic Cells. Nature Medicine, 2, 1096-1103.
https://doi.org/10.1038/nm1096-1096
[26] Herbst, R.S., Arkenau, H.T., Santana-Davila, R., et al. (2019) Ramucirumab plus Pembrolizumab in Patients with Previously Treated Advanced Non-Small-Cell Lung Cancer, Gastro-Oesophageal Cancer, or Urothelial Carcinomas (JVDF): A Multicohort, Non-Randomised, Open-Label, Phase 1a/B Trial. The Lancet Oncology, 20, 1109-1123.
https://doi.org/10.1016/S1470-2045(19)30458-9
[27] Pardi, N., Hogan, M.J. and Weissman, D. (2020) Recent Advances in MRNA Vaccine Technology. Current Opinion in Immunology, 65, 14-20.
https://doi.org/10.1016/j.coi.2020.01.008
[28] Ajani, J.A., et al. (2006) An Open-Label, Multinational, Multicenter Study of G17DT Vaccination Combined with Cisplatin and 5-Fluorouracil in Patients with Untreated, Advanced Gastric or Gastroesophageal Cancer: The GC4 Study. Cancer, 106, 1908-1916.
https://doi.org/10.1002/cncr.21814
[29] Sato, Y., Shomura, H., Maeda, Y., et al. (2003) Immunological Evaluation of Peptide Vaccination for Patients with Gastric Cancer Based on Pre-Existing Cellular Response to Peptide. Cancer Science, 94, 802-808.
https://doi.org/10.1111/j.1349-7006.2003.tb01522.x
[30] Lv, J., Zhao, R., Wu, D., et al. (2019) Mesothelin Is a Target of Chimeric Antigen Receptor T Cells for Treating Gastric Cancer. Journal of Hematology & Oncology, 12, Article No. 18.
https://doi.org/10.1186/s13045-019-0704-y
[31] Caruso, H.G., Heimberger, A.B. and Cooper, L.J.N. (2019) Steering CAR T Cells to Distinguish Friend from Foe. Oncoimmunology, 8, E1271857.
https://doi.org/10.1080/2162402X.2016.1271857
[32] Bębnowska, D., Grywalska, E., Niedźwiedzka-Rystwej, P., et al. (2020) CAR-T Cell Therapy—An Overview of Targets in Gastric Cancer. Journal of Clinical Medicine, 9, Article No. 1894.
https://doi.org/10.3390/jcm9061894