切换至 "中华医学电子期刊资源库"

中华胸部外科电子杂志 ›› 2025, Vol. 12 ›› Issue (03) : 176 -184. doi: 10.3877/cma.j.issn.2095-8773.2025.03.07

综述

全程新辅助治疗策略在食管癌及食管胃交界部癌中的研究进展
康晓征, 张瑞祥, 王镇, 陈先凯, 尚晓滨, 李勇, 秦建军, 李印()   
  1. 100021 北京,国家癌症中心/国家肿瘤临床医学研究中心/中国医学科学院北京协和医学院肿瘤医院胸外科
  • 收稿日期:2025-07-29 修回日期:2025-08-18 接受日期:2025-08-26 出版日期:2025-08-28
  • 通信作者: 李印
  • 基金资助:
    科技基础资源调查专项(2019FY101101); 北京市自然科学基金资助项目(L258061); 中央高水平医院临床科研业务费/中国医学科学院肿瘤医院合作基金(CFA202501004); 希思科-默沙东肿瘤研究基金项目(Y-MSDPU2022-0309)

Advances in total neoadjuvant therapy strategies for esophageal and esophagogastric junction cancers

Xiaozheng Kang, Ruixiang Zhang, Zhen Wang, Xiankai Chen, Xiaobin Shang, Yong Li, Jianjun Qin, Yin Li()   

  1. Section of Esophageal and Mediastinal Oncology, Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
  • Received:2025-07-29 Revised:2025-08-18 Accepted:2025-08-26 Published:2025-08-28
  • Corresponding author: Yin Li
引用本文:

康晓征, 张瑞祥, 王镇, 陈先凯, 尚晓滨, 李勇, 秦建军, 李印. 全程新辅助治疗策略在食管癌及食管胃交界部癌中的研究进展[J/OL]. 中华胸部外科电子杂志, 2025, 12(03): 176-184.

Xiaozheng Kang, Ruixiang Zhang, Zhen Wang, Xiankai Chen, Xiaobin Shang, Yong Li, Jianjun Qin, Yin Li. Advances in total neoadjuvant therapy strategies for esophageal and esophagogastric junction cancers[J/OL]. Chinese Journal of Thoracic Surgery(Electronic Edition), 2025, 12(03): 176-184.

食管癌及食管胃交界部癌是我国高发的恶性肿瘤,确诊时多为中晚期。新辅助治疗已成为局部进展期患者的标准方案。然而,传统的新辅助同步放化疗(nCRT)虽可提高手术切除率,但远处转移率仍居高不下,提示治疗策略仍需进一步优化。全程新辅助治疗(TNT)通过早期强化全身治疗、多药序贯联合及动态适应性调整,从时空维度克服肿瘤异质性导致的耐药问题,在食管腺癌和鳞癌中均展现出良好前景。现有研究表明,TNT策略在食管腺癌中可提高R0切除率[如FLOT(多西他赛+奥沙利铂+亚叶酸钙+5-氟尿嘧啶)方案病理完全缓解率(pCR)达20%],而nCRT联合免疫治疗(如阿替利珠单抗)可进一步改善生存[PERFECT试验2年总生存(OS)率达92%]。在食管鳞癌中,免疫联合nCRT显著提升pCR率(PALACE-1研究55.6%,CRIS研究63.2%),且特定生物标志物(如TCF-1+ T细胞)可能预测疗效。然而,TNT的毒性管理(如淋巴细胞减少)及最佳治疗时序仍需进一步优化。未来研究方向包括:开发ctDNA动态监测体系以指导个体化治疗;探索双免疫阻断、抗体-药物偶联物等创新组合;通过Ⅲ期临床试验(如KEYNOTE-975、EA2174)验证长期生存获益。多学科协作与精准免疫治疗将推动食管癌新辅助治疗的革新。

Esophageal cancer and esophagogastric junction cancer are highly prevalent malignant tumors in China, and most patients are diagnosed at an advanced stage. Neoadjuvant therapy has become the standard treatment for locally advanced patients. However, although traditional neoadjuvant chemoradiotherapy (nCRT) can improve the surgical resection rate, the distant metastasis rate remains high, indicating that the treatment strategy needs further optimization. Total neoadjuvant therapy (TNT) overcomes the drug resistance caused by tumor heterogeneity from the spatiotemporal dimension through early intensive systemic therapy, multidrug sequential combination, and dynamic adaptive adjustment, showing good prospects in both esophageal adenocarcinoma and squamous cell carcinoma. Existing studies have shown that the TNT strategy can improve the R0 resection rate in esophageal adenocarcinoma [for example, the pathological complete response rate (pCR) of the FLOT regimen (docetaxel + oxaliplatin + leucovorin + 5-fluorouracil) reaches 20%], and nCRT combined with immunotherapy (such as atezolizumab) can further improve survival [the 2-year overall survival (OS) rate in the PERFECT trial reaches 92%]. In esophageal squamous cell carcinoma, immunotherapy combined with nCRT significantly increases the pCR rate (55.6% in the PALACE-1 study and 63.2% in the CRIS study), and specific biomarkers (such as TCF-1+ T cells) may predict therapeutic efficacy. However, the toxicity management of TNT (such as lymphopenia) and the optimal treatment sequence still need further optimization. Future research directions include: developing a dynamic monitoring system for ctDNA to guide individualized treatment; exploring innovative combinations such as dual immunotherapy blockade and antibody-drug conjugates; and verifying long-term survival benefits through phase Ⅲ clinical trials (such as KEYNOTE-975 and EA2174). Multidisciplinary collaboration and precision immunotherapy will drive the innovation of neoadjuvant therapy for esophageal cancer.

表1 食管/胃食管结合部腺癌TNT治疗方案临床研究结果汇总
研究/作者 研究类型 病例数 治疗方案 生存结果 其他结局指标
NeoFLOT[12] 前瞻性单臂研究 50 FLOT方案(6周期) 中位DFS:32.9个月;1年OS率:79.3% R0切除率:86%;pCR率:20%;TRG 1a/b级:40%;发热性中性粒细胞减少:1.7%;≥3级中性粒细胞减少:29.3%;剂量调整:43.1%
Wo等[13] 前瞻性单臂研究 25 nFOLFIRINOX(8周期)+新辅助放化疗 2年PFS率:55%;2年OS率:72% TNT完成率:88%;手术切除率:80%;pCR率:35%;≥4级毒性:80%(淋巴细胞减少)
Dunne等[14] 前瞻性单臂研究 41 FOLFOX(3周期)+CROSS方案 中位RFS:3.1年;2年RFS率:71.5% 95%完成新辅助化疗;98%完成放化疗;R0切除率:97%;pCR率:22%
Shi等[15] 前瞻性单臂研究 28 放化疗(45 Gy)+SOX方案(6周期) 3年OS率:63%(手术组) R0切除率:100%;pCR率:35.7%
Ho等[16] 回顾性对照研究 580 诱导化疗+新辅助放化疗比单纯新辅助放化疗 中位OS:3.38年比2.45年(P=0.001) 淋巴结阴性率:21.2%比16.9%(无显著差异)
Carr等[17] 回顾性对照研究 451 诱导FOLFOX+新辅助放化疗比单纯新辅助放化疗 2年DFS率:68%比44%(P<0.001) pCR率:33%比22%(无显著差异);术后并发症无显著差异
PERFECT[18] 前瞻性单臂Ⅱ期临床研究 40 阿替利珠单抗+CROSS方案 2年OS率:92%(IFN-γ高表达亚组) pCR率:25%;IFN-γ特征基因可预测疗效
Uboha研究[19] 前瞻性单臂Ⅰ/Ⅱ期临床研究 22 阿维鲁单抗+同步放化疗 1年RFS率:71%;1年OS率:81% pCR率:26%;鳞癌67%比腺癌19%;versican蛋白水解状态可预测疗效
Ko等[20] 前瞻性单臂Ⅱ期临床研究 33 Sotigalimab(CD40激动剂)+放化疗 未报道 pCR率:37.9%;食管鳞癌pCR率达60%;增强树突细胞/T细胞免疫应答
表2 食管鳞癌TNT治疗方案临床研究结果汇总
1
Sung HFerlay JSiegel RL,et al.Global Cancer Statistics 2020:GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries[J].CA Cancer J Clin202171(3):209-249.
2
Morgan ESoerjomataram IRumgay H,et al.The Global Landscape of Esophageal Squamous Cell Carcinoma and Esophageal Adenocarcinoma Incidence and Mortality in 2020 and Projections to 2040:New Estimates From GLOBOCAN 2020[J].Gastroenterology2022163(3):649-658.e2.
3
Chen RZheng RZhang S,et al.Patterns and trends in esophageal cancer incidence and mortality in China:An analysis based on cancer registry data[J].J Natl Cancer Cent20233(1):21-27.
4
Ajani JAD’Amico TABentrem DJ,et al.Esophageal and Esophagogastric Junction Cancers,Version 2.2023,NCCN Clinical Practice Guidelines in Oncology[J].J Natl Compr Canc Netw202321(4):393-422.
5
Obermannová RAlsina MCervantes A,et al.Oesophageal cancer:ESMO Clinical Practice Guideline for diagnosis,treatment and follow-up[J].Ann Oncol202233(10):992-1004.
6
Eyck BMvan Lanschot JJBHulshof MCCM,et al.Ten-Year Outcome of Neoadjuvant Chemoradiotherapy Plus Surgery for Esophageal Cancer:The Randomized Controlled CROSS Trial[J].J Clin Oncol202139(18):1995-2004.
7
Kato KMachida RIto Y,et al.Doublet chemotherapy,triplet chemotherapy,or doublet chemotherapy combined with radiotherapy as neoadjuvant treatment for locally advanced oesophageal cancer(JCOG1109 NExT):a randomised,controlled,open-label,phase 3 trial[J].Lancet2024404(10447):55-66.
8
Hoeppner JBrunner TSchmoor C,et al.Perioperative Chemotherapy or Preoperative Chemoradiotherapy in Esophageal Cancer[J].N Engl J Med2025392(4):323-335.
9
Yoo RNKim HJ.Total neoadjuvant therapy in locally advanced rectal cancer:Role of systemic chemotherapy[J].Ann Gastroenterol Surg20193(4):356-367.
10
Ganesh KMassagué J.Targeting metastatic cancer[J].Nat Med202127(1):34-44.
11
Goldie JHColdman AJ.A mathematic model for relating the drug sensitivity of tumors to their spontaneous mutation rate[J].Cancer Treat Rep197963(11/12):1727-1733.
12
Schulz CKullmann FKunzmann V,et al.NeoFLOT:Multicenter phase Ⅱ study of perioperative chemotherapy in resectable adenocarcinoma of the gastroesophageal junction or gastric adenocarcinoma-Very good response predominantly in patients with intestinal type tumors[J].Int J Cancer2015137(3):678-685.
13
Wo JYClark JWEyler CE,et al.Results and Molecular Correlates from a Pilot Study of Neoadjuvant Induction FOLFIRINOX Followed by Chemoradiation and Surgery for Gastroesophageal Adenocarcinomas[J].Clin Cancer Res202127(23):6343-6353.
14
Dunne RFHsu MHarrington CA,et al.Induction FOLFOX prior to CROSS chemoradiotherapy and surgery in patients with locally-advanced esophageal and gastroesophageal junction cancer:a phase Ⅱ study[J].J Clin Oncol202240(4_suppl):327.
15
Shi JMLi NJiang LM,et al.A prospective phase II clinical trial of total neoadjuvant therapy for locally advanced gastric cancer and gastroesophageal junction adenocarcinoma[J].Sci Rep202414(1):7522.
16
Ho FTorphy RJFriedman C,et al.Induction Chemotherapy Plus Neoadjuvant Chemoradiation for Esophageal and Gastroesophageal Junction Adenocarcinoma[J].Ann Surg Oncol202128(12):7208-7218.
17
Carr RAHsu MHarrington CA,et al.Induction FOLFOX and PET-Directed Chemoradiation for Locally Advanced Esophageal Adenocarcinoma[J].Ann Surg2023277(3):e538-e544.
18
van den Ende Tde Clercq NCvan Berge Henegouwen MI,et al.Neoadjuvant Chemoradiotherapy Combined with Atezolizumab for Resectable Esophageal Adenocarcinoma:A Single-arm Phase Ⅱ Feasibility Trial (PERFECT)[J].Clin Cancer Res202127(12):3351-3359.
19
Uboha NVBasree MMEickhoff JC,et al.Phase I/II Trial of Perioperative Avelumab in Combination With Chemoradiation in the Treatment of Stage Ⅱ/Ⅲ Resectable Esophageal and Gastroesophageal Junction Cancer[J].J Surg Oncol2025131(7):1293-1301.
20
Ko AHChao JNoel MS,et al.A Phase 2 Study of Sotigalimab,a CD40 Agonist Antibody,plus Concurrent Chemoradiation as Neoadjuvant Therapy for Esophageal and Gastroesophageal Junction Cancers[J].Cancer Res Commun20255(2):349-357.
21
Yang HWang FHallemeier CL,et al.Oesophageal cancer[J].Lancet2024404(10466):1991-2005.
22
Peng LZhang JLi Y,et al.Sequential chemo-immunotherapy as a novel bridging strategy for non-complete responders after neoadjuvant chemoradiotherapy in esophageal cancer:first prospective phase 2 trial challenging the immediate-surgery paradigm[J].J Clin Oncol202543(16_suppl):4066.
23
Jiang YWang LChen X,et al.Neoadjuvant chemoradiotherapy (nCRT) combined with sequential tislelizumab for resectable esophageal squamous cell carcinoma (ESCC):preliminary results of a single-arm,open-label phase Ib trial (CRIS)[J].J Clin Oncol202442(16_suppl):e16116.
24
Xu XSun ZLiu Q,et al.Neoadjuvant chemoradiotherapy combined with sequential perioperative toripalimab in locally advanced esophageal squamous cell cancer[J].J Immunother Cancer202412(3):e008631.
25
Li CZhao SZheng Y,et al.Preoperative pembrolizumab combined with chemoradiotherapy for oesophageal squamous cell carcinoma (PALACE-1)[J].Eur J Cancer2021144:232-241.
26
Chen RLiu QLi Q,et al.A phase Ⅱ clinical trial of toripalimab combined with neoadjuvant chemoradiotherapy in locally advanced esophageal squamous cell carcinoma (NEOCRTEC1901)[J].EClinicalMedicine202362:102118.
27
Yang HLiu HChen Y,et al.Neoadjuvant Chemoradiotherapy Followed by Surgery Versus Surgery Alone for Locally Advanced Squamous Cell Carcinoma of the Esophagus (NEOCRTEC5010):A Phase Ⅲ Multicenter,Randomized,Open-Label Clinical Trial[J].J Clin Oncol201836(27):2796-2803.
28
Huang TCGuo JCLin CC,et al.Exploration of potential biomarkers associated with treatment outcomes in locally advanced esophageal squamous cell carcinoma patients receiving neoadjuvant nivolumab plus paclitaxel and cisplatin chemoradiotherapy[A].Cancer Res202585(8_Suppl_1):Abstract 744.
29
Pantel KAlix-Panabières C.Minimal residual disease as a target for liquid biopsy in patients with solid tumours[J].Nat Rev Clin Oncol202522(1):65-77.
30
Amin APlimack ERErnstoff MS,et al.Safety and efficacy of nivolumab in combination with sunitinib or pazopanib in advanced or metastatic renal cell carcinoma:the CheckMate 016 study[J].J Immunother Cancer20186(1):109.Erratum in:J Immunother Cancer,2019,7(1):73.
31
Shitara KBang YJIwasa S,et al.Trastuzumab deruxtecan in HER2-positive advanced gastric cancer:exploratory biomarker analysis of the randomized,phase 2 DESTINY-Gastric01 trial[J].Nat Med202430(7):1933-1942.
32
Shah MABennouna JDoi T,et al.KEYNOTE-975 study design:a Phase Ⅲ study of definitive chemoradiotherapy plus pembrolizumab in patients with esophageal carcinoma[J].Future Oncol202117(10):1143-1153.
33
Eads JRGibson MKLoConte NK,et al.A phase Ⅱ/Ⅲ study of peri-operative nivolumab and ipilimumab in patients with locoregional esophageal and gastroesophageal junction adenocarcinoma:Results of the neoadjuvant pathologic complete response rate(ECOG-ACRIN EA2174)[J].J Clin Oncol202442(16_suppl):4000.
[1] 罗兵, 董凤群, 牛艺臻, 王锟, 程志华, 刘宏强. 胎儿超声心动图在单纯性肺动脉瓣狭窄及预后评估中的价值[J/OL]. 中华医学超声杂志(电子版), 2025, 22(08): 740-747.
[2] 王达, 朱建敏. 血小板/淋巴细胞计数比值对乳腺癌新辅助化疗疗效的预测效能[J/OL]. 中华普外科手术学杂志(电子版), 2025, 19(06): 650-653.
[3] 贺雅莉, 黄丽, 杨培娟. 功能保留手术在低位直肠癌治疗中的研究进展[J/OL]. 中华普外科手术学杂志(电子版), 2025, 19(06): 701-704.
[4] 杨志, 夏雪峰, 管文贤. DeepSurv深度学习模型辅助胃癌术后精准化疗策略研究[J/OL]. 中华普外科手术学杂志(电子版), 2025, 19(05): 501-505.
[5] 徐其银, 韩尚志. 术前结合术后营养支持对直肠癌患者康复的影响[J/OL]. 中华普外科手术学杂志(电子版), 2025, 19(05): 543-546.
[6] 张聪, 李成. 胰头区恶性肿瘤外科手术预后现状及相关因素的研究进展[J/OL]. 中华普外科手术学杂志(电子版), 2025, 19(05): 574-578.
[7] 陈育纯, 王倩倩, 彭天明, 李勇, 田凯文, 刘志烨, 吴坤林, 蒲小勇, 刘久敏. 基于GEO数据库探究前列腺癌淋巴结转移和内脏转移中基因差异及预后[J/OL]. 中华腔镜泌尿外科杂志(电子版), 2025, 19(05): 579-585.
[8] 腾鹏, 田景昌, 鄂春翔, 向阳, 田伯宇. 肌层浸润性膀胱癌患者根治性膀胱切除术预后列线图模型的构建及验证[J/OL]. 中华腔镜泌尿外科杂志(电子版), 2025, 19(05): 645-652.
[9] 张燕, 许丁伟, 胡满琴, 黄昊扬, 宋光娜, 黄洁. 术前免疫炎症指标对肝癌肝切除术患者生存预后的预测价值[J/OL]. 中华肝脏外科手术学电子杂志, 2025, 14(05): 707-715.
[10] 方兴保, 庞国莲, 李月宏, 蔡艳. 基于多组学分析MCAM在肝癌中表达及其与生存预后和免疫细胞浸润的关系[J/OL]. 中华肝脏外科手术学电子杂志, 2025, 14(05): 716-724.
[11] 陈佳乐, 余安海, 袁文康, 张超, 张冲. 肝切除术围手术期监测及处理[J/OL]. 中华肝脏外科手术学电子杂志, 2025, 14(05): 785-788.
[12] 王淼, 贺佳佳, 潘颖威, 王小兰, 姚袆, 刘明宝, 陆兮, 苏丽洁. 远端胆管癌术后生存预后及其影响因素分析[J/OL]. 中华肝脏外科手术学电子杂志, 2025, 14(04): 582-588.
[13] 刘晓萍, 汪嵘嵘, 吴佳慧, 吴紫云, 周伯宣. 多组学分析HAPLN1与肝癌预后及免疫细胞浸润关系[J/OL]. 中华肝脏外科手术学电子杂志, 2025, 14(04): 609-618.
[14] 陈亚磊, 卢年芳, 刘安琪, 刘虎南, 赵培宏, 陈健文. 终末期肾病合并脓毒症患者临床特征及预后影响因素分析[J/OL]. 中华肾病研究电子杂志, 2025, 14(04): 196-203.
[15] 丘燕娇, 陈莹, 吴振梅, 钟秋杰, 韩小妍, 李又佳, 罗宋宝, 韩倩倩. 脑心健康管理师介入的营养管理对老年急性缺血性脑卒中患者预后的影响[J/OL]. 中华脑血管病杂志(电子版), 2025, 19(04): 312-317.
阅读次数
全文


摘要


AI


AI小编
你好!我是《中华医学电子期刊资源库》AI小编,有什么可以帮您的吗?