进展期胃癌的治疗现状及进展
Treatment Status and Progress of Advanced Gastric Cancer
DOI: 10.12677/ACM.2023.134768, PDF, HTML, XML, 下载: 209  浏览: 351 
作者: 刘定定, 李雨婷:延安大学附属医院,陕西 延安 ;杨卫卫*:延安大学附属医院肿瘤科,陕西 延安
关键词: 进展期胃癌化疗放疗免疫治疗Advanced Gastric Cancer Chemotherapy Radiotherapy Immunotherapy
摘要: 胃癌作为一个全球性健康问题,因缺乏体检筛查的意识以及胃癌早期发病隐匿性,并且无明显的特异性症状,容易被忽略。故绝大部分患者在确诊时已是进展期胃癌,目前的医学进展,手术仍然是根治胃癌唯一可能的方法,随着对胃癌发病机制及生物学的不断深入研究,目前进展期胃癌患者以手术治疗为主,结合化、放疗、靶向治疗、免疫治疗的综合治疗模式,本文就目前进展期胃癌的治疗进展进行综述,旨在为进展期胃癌的精准治疗提供参考依据。
Abstract: As a global health problem, gastric cancer is easy to be ignored due to the lack of awareness of physical examination and screening, the occult early onset of gastric cancer, and no obvious specific symptoms. Therefore, most patients have advanced gastric cancer when they are diagnosed. With the current medical progress, surgery is still the only possible method for radical treatment of gas-tric cancer. With the continuous in-depth research on the pathogenesis and biology of gastric cancer, surgical treatment is the main treatment for patients with advanced gastric cancer, and the com-prehensive treatment mode of chemotherapy, radiotherapy, targeted therapy and immunotherapy is adopted. This article reviews the current progress in the treatment of advanced gastric cancer, aiming to provide reference for the precise treatment of advanced gastric cancer.
文章引用:刘定定, 李雨婷, 杨卫卫. 进展期胃癌的治疗现状及进展[J]. 临床医学进展, 2023, 13(4): 5419-5426. https://doi.org/10.12677/ACM.2023.134768

1. 引言

胃癌是一个主要的全球健康问题,特别是在东亚国家 [1] [2] 。在全球范围内,2020年100多万病例中76.8万多人死亡,使胃癌成为世界上第五大最常诊断的癌症以及第三大癌症相关死亡的原因 [3] [4] 。全球胃癌发病率的地理差异很大,高发及低发地区之间的差异为15~20倍。胃癌发病率最高的地区是东北亚、中、南美洲和东欧。在日本和韩国,胃癌是男性最常见的癌症,然而在中国,胃癌是癌症相关死亡的主要原因 [1] [2] 。早期胃癌的预后相对较好,通过手术、辅助化疗以及放疗等综合治疗手段,5年的生存率可达95%。但是早期胃癌通常因为缺乏典型的临床表现,因此大约70%的患者初次去往医院就诊时便已经处于进展期。进展期胃癌(Advanced gastric cancer, AGC)是指癌组织浸润深度达到肌层,浆膜层或浆膜外者。进展期胃癌目前的治疗手段仍然是采取手术治疗为主,但是单纯手术难以达到根治目的,因此目前是以手术治疗为主,化疗、放疗、靶向治疗及免疫治疗的综合治疗模式逐渐成为标准治疗模式。本文就进展期胃癌综合治疗的现状及进展进行概述。

2. 进展期胃癌的新辅助治疗

目前新辅助治疗已被中国临床肿瘤学会(CSCO)、美国国立综合癌症网络(NCCN)以及欧洲肿瘤内科学会(ESMO)胃癌诊疗指南推荐为局部晚期胃癌综合治疗的重要一环。新辅助治疗的优点:有助于了解肿瘤对放化疗等治疗的敏感程度,为进一步的治疗提供依据;还可以缩小肿瘤,从而降低胃癌分期,使更多病人能有外科手术治疗的机会,再者也有利于防止耐药细胞株的形成,以减少术后复发和转移 [5] 。

1) 新辅助化疗(Neoadjuvant chemotherapy, NCT)是在术前给予患者的辅助化疗,也称诱导性化疗。2006年的MAGIC试验 [6] 是一项开创性研究,在可手术的胃食管腺癌患者中,对比了新辅助化疗ECF (表柔比星、顺铂和5-FU) + 手术与单纯手术的5年生存率(36% vs 23%)。2019年FLOT4-AIO研究结果显示 [7] ,ECF/ECX方案(表柔比星 + 顺铂 + 5-FU/卡培他滨)与FLOT方案(氟尿嘧啶 + 奥沙利铂 + 多西他赛 + 亚叶酸钙)相对比而言,FLOT方案能够明显提高进展期胃或胃–食管结合部腺癌病理完全缓解率(pathologic complete response, pCR) (16% vs 6%),中位总生存期(median OS, mOS)对比(50个月vs 35个月),5年的总生存(OS)对比(45% vs 36%)。鉴于该结果,FLOT方案已经替代ECF/ECX方案成为了欧洲胃癌新辅助化疗首选方案。JCOG0405研究 [8] 表明存在广泛的淋巴结转移AGC患者,SP (S-1 + 顺铂)方案化疗加根治术联合主动脉旁淋巴结清扫对于部分肿瘤患者是安全有效。国内开展的一项Ⅲ期RESOLVE随机的对照研究,2组XELOX方案(卡培他滨 + 奥沙利铂)和SOX方案(奥沙利铂 + 替吉奥)的辅助化疗以及SOX方案围手术期的化疗疗效及安全性,结果显示,3组的3年DFS率分别为51.1%、56.5%、59.4%。表明SOX方案的新辅助化疗能够显著改善患者的生存,更有利于进展期胃癌的疗效 [9] 。2020年的美国临床肿瘤学会(ASCO)的会议公布,SOX新辅助化疗组中67.6% (261/386)的患者能够实现肿瘤降期,病理有效率、pCR率达到 67.8%及23.6% [10] 。一项来自日本的回顾性研究(JCOG1302A)结果提示,cStage III (cT34N13)型的胃癌患者可以成为新辅助化疗的获益人群 [11] 。

2) 新辅助化疗联合放疗:德国POET研究,术前PLF方案(顺铂 + 亚叶酸钙 + 5-FU)的基础上给予同步放疗,使的pCR率从1.9%提高到14.3% [12] 。MD Anderson的单中心II期研究RTOG9904显示,术前PF方案(顺铂 + 5FU)联合放疗的pCR率可达26% [13] 。

3) HER-2阳性患者:一项II~III期的PETRARCA研究 [14] (NCT02581462),Her2阳性患者,曲妥珠单抗或帕妥珠单抗联合FLOT化疗与FLOT组对比,显著提高了患者的pCR率(35% vs. 12%, P = 0.02)以及ypN0率(68% vs. 39%)。

4) 靶向及免疫的新辅助治疗:一项II期临床研究提示,阿帕替尼联合SOX方案的新辅助治疗对于进展期胃癌的疗效显著,不良反应也是可耐受 [15] 。但因为入组的病例比较少,所以此方案的安全性以及疗效仍需要未来更多的前瞻性研究验证。2021年ASCO的会议上汇报了一项探索卡瑞利珠联合阿帕替尼联合SOX方案用于局部进展期(cT4a/bN+)胃癌的新辅助治疗的II期临床研究结果显示pCR率为16.7%,肿瘤的降期率为79.2% (19/24)。2021年ASCO会议的SHARED研究(ChiCTR1900024428)结果显示,信迪利单抗联合同步放化疗新辅助治疗局部进展期的胃癌pCR为42.1% (8/19) R0切除率为94.7% (18/19)。进展期胃癌的新辅助治疗越来越受到临床医生的重视,逐渐成为综合治疗中的重要环节。但由于东、西方国家肿瘤的生物学的差异性,所以目前仍不确定国外的临床研究成果是否可以用于中国胃癌患者。再者尚无最佳的新辅助治疗的方案共识。仍然需要更多严谨的临床试验,研究新辅助治疗选择性应用的标准,最大限度的发挥新辅助化疗策略优势,避免过度治疗的风险。

3. 手术治疗

日本胃癌治疗指南 [16] 指出,根治性的胃切除术:标准胃切除术、非标准胃切除术。标准胃切除术:远端胃切除术(切除至少2/3的胃,进行D2淋巴结清扫并将近端胃与小肠吻合)或者是食管与小肠吻合的全胃切除术。非标准胃切除术:胃切除和(或)淋巴结切除术的范围需要根据肿瘤分期而改变,包括改良根治术、扩大根治术。改良根治术的胃切除和(或)淋巴结切除范围相对减小(D1、D1+等);扩大根治术胃的切除范围包括:① 胃切除联合邻近受累器官;② 胃切除术伴D2以上扩大淋巴结切除术。非治愈性手术是为了那些被认为是无法治愈的胃癌病人提供的。根据手术目的的不同,分为姑息性手术或者减瘤性手术。姑息性手术:晚期/转移性胃癌患者可能出血或梗阻等严重临床症状。根据原发性肿瘤的可切除性和/或手术风险,可以选择姑息性胃切除术或胃空肠吻合术来缓解症状。据报道,胃分区胃空肠吻合术的功能优于单纯胃空肠吻合术 [17] 。减瘤性手术:对存在不可切除的肝转移、腹膜转移等不可治愈因素,并且无出血、梗阻等肿瘤相关症状的患者进行胃切除术。它的目的是希望通过减少肿瘤的体积从而延长患者生存期或者延迟患者症状的再次出现。然而,一项国际合作的随机对照试验(REGATTA, JCOG0705/KGCA01)未能证明减瘤手术 [18] 的生存效益。

20余年来,随着器械的发展以及外科操作技术的不断进步,尤其是腹腔镜下胃肠道吻合技术的明显进步,腹腔镜胃癌根治术后的消化道重建由腹腔镜辅助治疗向完全性腹腔镜发展,镜下操作可以避免上腹部辅助小切口也更加符合微创的概念与要求,并且能够有效地改善患者的近期疗效,也可以提高患者术后生存质量 [19] 。目前,大量的临床研究证据展现了腹腔镜手术对于治疗早期胃癌的安全性以及可行性,由于其明显微创的优点,即便是中国胃癌患者目前仍以进展期为主,但是我国仍然积极地开展全国性乃至世界性的多中心临床试验研究,期望为腹腔镜手术的临床应用提供了更高级别的循证医学证据 [20] 。

4. 术后、放化疗

历年的研究评估表明了联合放、化疗对比单独放、化疗的生存优势更大 [21] [22] 。在美国,INT0116研究 [23] 显示,术后辅助放化疗被认为是首选的标准治疗,放化疗组与单纯手术组死亡危险比:0.74 (95% CI, 0.60~0.92)。CLASSIC试验 [24] 证实了接受根治性胃切除术伴D2 (扩展)淋巴结清扫后加卡培他滨和奥沙利铂的化疗方案使患者可以从中受益。辅助化疗组3年DFS率为74%,单纯手术组为59%。美国组间试验 [25] SWOG9008/INT-0116,比较了手术加术后化疗(5-FU和叶酸) + 放疗对比单独手术。在随机的3期ACTS-GC试验中 [26] (政府标识NCT00152217)与单独手术相比,辅助治疗S-1,1年后的5年的生存率(OS)可以提高(72% vs 61%),JACCRO GC-07三期试验 [27] (大学医院鉴定网络[UMIN]标识符R000012099),接受治疗性手术和D2淋巴结切除术的病理性3期胃癌患者随机接受辅助治疗(S-1 + 多西他赛或单独使用S1)中期分析时,联合组的3年无复发生存率更好(66% vs 50%)。2018年报道的ARTIST试验比较了完全D2切除的胃癌患者接受卡培他滨 + 顺铂对比同时接受(卡培他滨 + 顺铂) + 放疗 [28] 。总体而言,无病生存率在组间无显著差异。但是,在淋巴结转移患者的亚组中,放化疗组确实显示出统计学上的无病生存期改善。

5. 胃癌的靶向治疗

由于化疗药物在杀伤肿瘤细胞的同时对正常细胞也具有伤害,导致了临床上不可避免的出现严重程度不等的副反应。因此利用分子靶向驱动特点,特异性地消灭肿瘤细胞,从而能够最大程度地降低治疗中出现副作用,进而改善患者的生活质量。

5.1. 抑制肿瘤血管生成

在肿瘤生长的过程中,需要足够的氧气、营养形成新生血管,导致肿瘤的生长、侵袭、转移。因此,在治疗过程中,抑制血管生成(成纤维细胞生长因子(FGF)、血管内皮生长因子(VEGF))可以达到良好的治疗效果 [29] [30] 。胃癌患者的此类代表药物雷莫芦单抗、阿帕替尼、AZD4547。REGARD试验将使用一线化疗后(铂类 + 氟尿嘧啶)病情进展的患者进行随机对照研究,分别使用雷莫卢单抗(8 mg/kg)和安慰剂,结果显示雷莫卢单抗治疗效果较好,患者mOS为5.2个月,而使用安慰剂患者为3.8个月 [31] 。单用紫杉醇对比雷莫卢单抗联合紫杉醇进行晚期胃癌二线治疗的效果,结果显示联合治疗的疗效更好,mOS为9.6个月,单用紫杉醇mOS为7.4个月 [32] 。单药AZD4547治疗患者的中位无进展生存期(PFS)为1.8个月,AZD4547联合紫杉醇治疗患者的中位PFS为3.5个月,且两组患者的不良事件发生情况无明显差异 [33] 。阿帕替尼(apatinib)是我国自主研发的小分子酪氨酸激酶抑制剂(TKI),VEGFR-2为其主要作用靶点。一项III期临床试验 [34] ,显示阿帕替尼组对比安慰剂组PFS (2.6个月vs 1.8个月)和中位OS (6.5个月vs 4.7个月)均明显延长,患者明显获益,并且药物的不良反应也可控。2014年10月,阿帕替尼已被国家药品监督管理(NMPA)已正式批准应用。在我国已被批准为晚期胃癌或胃食管结合部癌的二线用药。

5.2. 抑制肿瘤细胞增殖

HER-2是EGFRs家族的细胞膜蛋白,在跨膜信号传导中有所参与,当HER-2激活或过表达时,可促进肿瘤细胞增殖、侵袭及转移。ToGA试验表明,与单纯化疗相比,曲妥珠单抗 + 卡培他滨 + 顺铂、曲妥珠单抗 + 5-FU + 顺铂作为一线治疗方案患者获益更大,联合治疗患者的mOS为13.8个月,高于单纯化疗患者的11.1个月 [35] 。

6. 免疫治疗

免疫治疗在过去的十年中,免疫检查点封锁已经成为一种令人兴奋的恶性肿瘤治疗策略。包括抑制程序性细胞死亡蛋白1 (PD-1)、PD-L1、细胞毒性t淋巴细胞抗原4 (CTLA-4)。免疫检查点抑制剂(ICIs)被开发用来阻断配体与检查点受体的结合,并重新激活人体细胞免疫反应。对进展期胃癌患者的免疫治疗免疫检查点封锁现在被确定为化疗难治性胃癌(在两次或两次以上化疗后进展的癌症)的治疗方法。

2016年,第一阶段研究 [36] KEYNOTE-012表明36例患者中有8例(22%)表示在使用帕博利珠单抗后有所缓解。在该研究后,KEYNOTE-059队列1显示了使用了帕博利珠单抗后,其客观有效率(ORR)为11.6%,中位无进展生存期(PFS)为2个月,中位总生存期(OS)为5.6个月 [37] 。基于这项研究的结果,美国食品和药物管理局(FDA)于2017年9月批准pembrolizumab作为PD-L1阳性(CPS) ≥ 1的复发的局部晚期或转移性胃癌或胃食管交界癌的治疗。2017年,亚洲的ATTRACTION-2研究 [38] 显示胃癌患者表现出nivolumab组与安慰剂相比,显示mOS为5.26月(95%CI, 4.60~6.37)。2020年ASCO-GI会议报告称nivolumab组对比安慰组的3年OS率(5.6% vs. 1.9%) [39] 。因此,nivolumab是在日本被批准用于胃癌的三线治疗。此外,该项研究发现nivolumab对至少接受过四种不同先前治疗的胃癌患者疗法更有效。在JAVELIN Gastric 100研究中 [40] ,与继续化疗相比,在所有入选患者或PD-L1 TPS为1%的患者中,一线奥沙利铂/氟嘧啶化疗后,单药avelumab作为维持治疗未能改善OS。尽管PD-1/PD-L1抑制剂在治疗晚期胃癌/胃食管交界处癌中表现出临床活性,特别是对晚期一线治疗,但单一疗法的益处相对有限 [41] 。由北大肿瘤医院沈琳教授牵头的国内多中心II期临床试验结果显示,卡瑞利珠单抗联合卡培他滨 + 奥沙利铂(XELOX)序贯卡瑞利珠单抗联合阿帕替尼方案一线治疗48例 HER2 阴性晚期胃/胃食管交界处腺癌的ORR为 58.3%,DCR高达93.8%,中位OS达14.9个月,中位PFS达6.8个月(95%CI:5.6~9.5个月)。具有较好的疗效和安全性 [42] 。NCT02915432研究 [43] 显示,信迪利单抗联合化疗(XELOX)对比单纯化疗,中位生存期延长了2.9个月(15.2个月vs 12.3个月);在PD-L1 CPS ≥ 5的患者中,中位OS延长更多,延长5.5个月(18.4个月vs 12.9个月),死亡风险分别降低23%和34%。多塔利单抗(dostarlimb-gxly)是一种抗PD-1抗体,于2021年8月被FDA批准用于治疗dMMR复发或晚期实体瘤患者 [44] [45] 。目前未在国内上市。

7. 总结及展望

胃癌是一种异质性较强的肿瘤,具有很高的恶性潜能。目前对于进展期胃癌的治疗手段仍然是手术为主,随着综合治疗的开展给进展期胃癌患者带来更好的生存期及生活质量。雷莫芦单抗以及阿帕替尼是目前已被证明能够改善晚期胃癌患者的生存结局,因为胃癌的瘤内、患者内和患者间异质性仍然是靶向治疗药物开发的一个关键障碍,所以很多靶向治疗试验失败甚至提前中止。目前免疫治疗被认为最有潜力改善晚期胃癌患者的生存结局。总而言之,靶向药物以及免疫治疗在进展期胃癌中的治疗的安全性及疗效仍需进一步探索。我们需更多的临床试验,制定个体化治疗。期待未来更多高质量的大型临床试验不断开展,为进展期胃癌患者带来更多的福音。

NOTES

*通讯作者。

参考文献

[1] Torre, L.A., Siegel, R.L., Ward, E.M. and Jemal, A. (2016) Global Cancer Incidence and Mortality Rates and Trends—An Update. Cancer Epidemiology, Biomarkers & Prevention, 25, 16-27.
https://doi.org/10.1158/1055-9965.EPI-15-0578
[2] Etemadi, A., Safiri, S., Sepanlou, S.G., et al. (2020) The Global, Regional, and National Burden of Stomach Cancer in 195 Countries, 1990-2017: A Systematic Analysis for the Global Burden of Disease Study 2017. The Lancet Gastroenterology and Hepatology, 5, 42-54.
[3] Etemadi, A., Safiri, S., Sepanlou, S.G., et al. (2020) The Global, Regional, and National Burden of Stomach Cancer in 195 Countries, 1990-2017: A Systematic Analysis for the Global Burden of Disease Study 2017. The Lancet Gastroenterology and Hepatology, 5, 42-54.
https://doi.org/10.1016/S2468-1253(19)30328-0
[4] Bray, F., Ferlay, J., Soerjomataram, I., et al. (2018) Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Can-cers in 185 Countries. CA: A Cancer Journal for Clinicians, 68, 394-424.
https://doi.org/10.3322/caac.21492
[5] World Health Organization, International Agency for Research on Cancer (2020) GLOBOCAN 2020: Stomach Cancer Fact Sheet.
[6] 留成, 陈建思, 覃宇周, 黄名威, 金钦文. 进展期胃癌新辅助治疗病理完全缓解的研究进展[J]. 中国癌症防治杂志, 2021, 13(5): 559-564.
[7] Cunningham, D., Allum, W.H., Stenning, S.P., et al. (2006) Perioperative Chemotherapy versus Surgery Alone for Resectable Gastroesophageal Cancer. The New England Journal of Medicine, 355, 11-20.
https://doi.org/10.1056/NEJMoa055531
[8] Batran, S.E., Homann, N., Pauligk, C., et al. (2019) Perioperative Chemotherapy with Fluorouracil plus Leucovorin, Oxaliplatin, and Docetaxel versus Fluorouracil or Capecitabine plus Cisplatin and Epirubicin for Locally Advanced, Resectable Gastric or Gastro-Oesophageal Junction Adenocarcinoma (FLOT4): A Randomised, Phase 2/3 Trial. The Lancet, 393, 1948-1957.
https://doi.org/10.1016/S0140-6736(18)32557-1
[9] Tsuburaya, A., et al. (2014) Neoadjuvant Chemotherapy with S-1 and Cisplatin Followed by D2 Gastrectomy with Para-Aortic Lymph Node Dissection for Gastric Cancer with Ex-tensive Lymph Node Metastasis. British Journal of Surgery, 101, 653-660.
https://doi.org/10.1002/bjs.9484
[10] Hang, X.T., Liang, H., Li, Z.Y., et al. (2021) Perioperative or Postoperative Adjuvant Oxaliplatin with S-1 versus Adjuvant Oxaliplatin with Capecitabine in Patients with Locally Advanced Gastric or Gastro-Oesophageal Junction Adenocarcinoma Undergoing D2 Gastrectomy (RESOLVE): An Open-Label, Superior-ity and Non-Inferiority, Phase 3 Randomised Controlled Trial. The Lancet Oncology, 22, 1081-1092.
https://doi.org/10.1016/S1470-2045(21)00297-7
[11] Xie, T.Y., et al. (2020) Early Results of the Randomized, Multicenter, Controlled Evaluation of S-1 and Oxaliplatin as Neoadjuvant Chemotherapy for Chinese Advanced Gastric Cancer Patients (RESONANCE Trial). Journal of Clinical Oncology, 38, 280.
https://doi.org/10.1200/JCO.2020.38.4_suppl.280
[12] Fukagawa, T., Katai, H., Mizusawa, J., et al. (2018) A Prospective Multi-Institutional Validity Study to Evaluate the Accuracy of Clinical Diagnosis of Pathological Stage III Gastric Cancer (JCOG1302A). Gastric Cancer, 21, 68-73.
https://doi.org/10.1007/s10120-017-0701-1
[13] Stahl, M., Walz, M.K., Riera-Knorrenschild J., et al. (2017) Pre-operative Chemotherapy versus Chemoradiotherapy in Locally Advanced Adenocarcinomas of the Oesophagogastric Junction (POET): Long-Term Results of a Controlled Randomised Trial. European Journal of Cancer, 81, 183-190.
https://doi.org/10.1016/j.ejca.2017.04.027
[14] Ajani, J.A., Winter, K., Okawara, G.S., et al. (2006) Phase II Trial of Preoperative Chemoradiation in Patients with Localized Gastric Adenocarcinoma (RTOG 9904): Quality of Combined Modality Therapy and Pathologic Response. Journal of Clinical Oncology, 24, 3953-3958.
https://doi.org/10.1200/JCO.2006.06.4840
[15] Hofheinz, R.D., Haag, G.M., Ettrich, T.J., et al. (2020) Periopera-tive Trastuzumab and Pertuzumab in Combination with FLOT versus FLOT Alone for HER2-Positive Resectable Esophagogastric Adenocarcinoma: Final Results of the PETRARCA Multicenter Randomized Phase II Trial of the AIO. Journal of Clinical Oncology, 38, 4502.
https://doi.org/10.1200/JCO.2020.38.15_suppl.4502
[16] Zheng, Y.N., Yang, X., Yan, C., et al. (2020) Effect of Apatinib plus Neoadjuvant Chemotherapy Followed by Resection on Pathologic Response in Patients with Locally Ad-vanced Gastric Adenocarcinoma: A Single-Arm, Open-Label, Phase II Trial. European Journal of Cancer, 130, 12-19.
https://doi.org/10.1016/j.ejca.2020.02.013
[17] Japanese Gastric Cancer Association (2021) Japanese Gastric Can-cer Treatment Guidelines 2018 (5th Edition). Gastric Cancer, 24, 1-21.
https://doi.org/10.1007/s10120-020-01042-y
[18] Kaminishi, M., Yamaguchi, H., Shimizu, N., et al. (1997) Stom-ach-Partitioning Gastrojejunostomy for Unresectable Gastric Carcinoma. The Archives of Surgery, 132, 184-187.
https://doi.org/10.1001/archsurg.1997.01430260082018
[19] Fujitani K., Yang, H.K., Mizusawa, J., et al. (2016) Gastrectomy plus Chemotherapy versus Hemotherapy Alone for Advanced Gastric Cancer with a Single Non-Curable Factor (REGATTA): A Phase 3, Randomised Controlled Trial. The Lancet Oncology, 17, 309-318.
https://doi.org/10.1016/S1470-2045(15)00553-7
[20] Shen, J., Ma, X., Yang, J., et al. (2020) Digestive Tract Re-construction Options after Laparoscopic Gastrectomy for Gastric Cancer. World Journal of Gastrointestinal Oncology, 12, 21-36.
https://doi.org/10.4251/wjgo.v12.i1.21
[21] 陈起跃, 钟情, 刘治羽, 黄晓波, 阙思瑾, 郑文泽, 李平, 郑朝辉, 黄昌明. 腹腔镜进展期胃癌根治术在中国的进展[J]. 中国肿瘤临床, 2021, 48(3): 110-117.
[22] Moertel, C., Reitemeier, R., Childs, D., Colby, M. and Holbrook, M. (1969) Combined 5-Fluorouracil and Supervoltage Radiation Therapy of Locally Unresectable Gastrointestinal Cancer. The Lancet, 294, 865-867.
https://doi.org/10.1016/S0140-6736(69)92326-5
[23] Gastrointestinal Tumor Study Group (1982) A Comparison of Combination Chemotherapy and Combined Modality Therapy for Locally Advanced Gastric Carcinoma. Cancer, 49, 1771-1777. http://www.ncbi.nlm.nih.gov/pubmed/6176313
https://doi.org/10.1002/1097-0142(19820501)49:9<1771::AID-CNCR2820490907>3.0.CO;2-M
[24] Macdonald, J.S., Smalley, S.R., Benedetti, J., Hundahl, S.A., Estes, N.C., Stemmermann, G.N., et al. (2001) Chemoradiotherapy af-ter Surgery Compared with Surgery Alone for Adenocarcinoma of the Stomach or Gastroesophageal Junction. The New England Journal of Medicine, 345, 725-730.
https://doi.org/10.1056/NEJMoa010187
[25] Bang, Y.J., Kim, Y.W., Yang, H.K., et al. (2012) Adjuvant Capecitabine and Oxaliplatin for Gastric Cancer after D2 Gastrectomy (CLASSIC): A Phase 3 Open-Label, Randomised Controlled Trial. The Lancet, 379, 315-321.
https://doi.org/10.1016/S0140-6736(11)61873-4
[26] Smalley, S.R., Benedetti, J.K., Haller, D.G., Hundahl, S.A., Estes, N.C., Ajani, J.A., et al. (2012) Updated Analysis of SWOG-Directed Intergroup Study 0116: A Phase III Trial of Adjuvant Radiochemotherapy versus Observation after Curative Gastric Cancer Resection. Journal of Clinical Oncology, 30, 2327-2333.
https://doi.org/10.1200/JCO.2011.36.7136
[27] Sasako, M., Sakuramoto, S., Katai, H., et al. (2011) Five-Year Outcomes of a Randomized Phase III Trial Comparing Adjuvant Chemotherapy with S-1 versus Surgery Alone in Stage II or III Gastric Cancer. Journal of Clinical Oncology, 29, 4387-4293.
https://doi.org/10.1200/JCO.2011.36.5908
[28] Yoshida, K., Kodera, Y., Kochi, M., et al. (2019) Addition of Docetaxel to Oral Fluoropyrimidine Improves Efficacy in Patients with Stage III Gastric Cancer: Interim Analysis of JACCRO GC-07, a Randomized Controlled Trial. Journal of Clinical Oncology, 37, 1296-1304.
https://doi.org/10.1200/JCO.18.01138
[29] (2019) Interim Results of a Phase III Trial Involving Adjuvant Chemo-therapy and/or Chemoradiotherapy after D2-Gastrectomy in Stage II/III Gastric Cancer (GC). ASCO Annual Meeting, Chicago, 31 May-4 June 2019, Abstract 4001.
https://doi.org/10.1200/JCO.2019.37.15_suppl.4001
[30] Macedo, F., Ladeira, K., Longatto-Filho, A., et al. (2017) Gastric Cancer and Angiogenesis: Is VEGF a Useful Biomarker to As-sess Progression and Remission? Journal of Gastric Cancer, 17, 1-10.
https://doi.org/10.5230/jgc.2017.17.e1
[31] Mawalla, B., Yuan, X., Luo, X., et al. (2018) Treatment Outcome of Anti-Angiogenesis through VEGF-Pathway in the Management of Gastric Cancer: A Systematic Review of Phase II and III Clinical Trials. BMC Research Notes, 11, 21-22.
https://doi.org/10.1186/s13104-018-3137-8
[32] Chuman, M., Takahashi, Y., Tokito, T., et al. (2019) A Case of Recurrent Gastric Cancer with Clinical Complete Response after Ramucirumab plus Paclitaxel Therapy. Gan to Kagaku Ryoho, 46, 1931-1933.
[33] Kawai, S., Fukuda, N., Yamamoto, S., et al. (2020) Retrospective Observational Study of Salvage Line Ramucirumab Monotherapy for Patients with Ad-vanced Gastric Cancer. BMC Cancer, 20, 338-340.
https://doi.org/10.1186/s12885-020-06865-7
[34] Jang, J., Kim, H.K., Bang, H., et al. (2017) Antitumor Effect of AZD4547 in a Fibroblast Growth Factor Receptor 2-Amplified Gastric Cancer Patient-Derived Cell Model. Translation-al Oncology, 10, 469-475.
https://doi.org/10.1016/j.tranon.2017.03.001
[35] Li, J., Qin, S., Xu, J., et al. (2016) Randomized, Double-Blind, Placebocontrolled Phase III Trial of Apatinib in Patients with Chemotherapy Refractory Advanced or Metastatic Adeno-carcinoma of the Stomach or Gastroesophageal Junction. Journal of Clinical Oncology, 34, 1448-1454.
https://doi.org/10.1200/JCO.2015.63.5995
[36] Aguilar-Mahecha, A., Joseph, S., Cavallone, L., et al. (2019) Pre-cision Medicine Tools to Guide Therapy and Monitor Response to Treatment in a HER-2+ Gastric Cancer Patient: Case Report. Frontiers in Oncology, 9, 698-699.
https://doi.org/10.3389/fonc.2019.00698
[37] Muro, K., Chung, H., Shankaran, V., et al. (2016) Pembrolizumab for Patients with PD-L1-Positive Advanced Gastric Cancer (KEYNOTE-012): A Multicentre, Open-Label, Phase 1b Tri-al. The Lancet Oncology, 17, 717-726.
https://doi.org/10.1016/S1470-2045(16)00175-3
[38] Fuchs, C.S., Doi, T., Jang, R.W., et al. (2018) Safety and Ef-ficacy 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
[39] 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 Pre-vious Chemotherapy Regimens (ONO-4538-12, ATTRACTION-2): A Randomised, Double-Blind, Placebo-Controlled, Phase 3 Trial. The Lancet, 390, 2461-2471.
https://doi.org/10.1016/S0140-6736(17)31827-5
[40] 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 Pa-tients with Gastric Cancers: Results from JAVELIN Gastric 100. Journal of Clinical Oncology, 39, 966-977.
https://doi.org/10.1200/JCO.20.00892
[41] Boku, N., Satoh, T., Ryu, M., Chao, Y., et al. (2021) Nivolumab in Previously Treated Advanced Gastric Cancer (ATTRACTION-2): 3-Year Update and Outcome of Treatment beyond Progression with Nivolumab. Gastric Cancer, 24, 946-958.
https://doi.org/10.1007/s10120-021-01173-w
[42] Peng, Z., Wei, J., Wang, F., et al. (2021) Camrelizumab Com-bined with Chemotherapy Followed by Camrelizumab plus Apatinib as First-Line Therapy for Advanced Gastric or Gas-troesophageal Junction Adenocarcinoma. Clinical Cancer Research, 27, 3069-3078.
https://doi.org/10.1158/1078-0432.CCR-20-4691
[43] Wang, F., Wei, X.L., Wang, F.H., et al. (2019) Safety, Ef-ficacy 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.
[44] Jiang, H., Zheng, Y., Qian, J., et al. (2020) Safety and Efficacy of Sintilimab Combined with Oxali-platin/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, 760.
https://doi.org/10.1186/s12885-020-07251-z
[45] U.S. Food and Drug Administration. (2021) FDA Grants Accel-erated Approval to Dostarlimab-Gxly for dMMR Advanced Solid Tumors.
https://www.fda.gov/drugs/resourcesinformation-approved-drugs/fda-grants-accelerated-approvaldostarlimab-gxly-dmmr-advanced-solid-tumors