Zaanan Aziz, Bouché Olivier, de la Fouchardière Christelle, Le Malicot Karine, Pernot Simon, Louvet Christophe, Artru Pascal, Le Brun Ly Valérie, Aldabbagh Kais, Khemissa-Akouz Faiza, Lecomte Thierry, Castanie Hélène, Laly Margot, Botsen Damien, Roth Gael, Samalin Emmanuelle, Muller Marie, Breysacher Gilles, Manfredi Sylvain, Phelip Jean-Marc, Taieb Julien
Department of Gastroenterology and Digestive Oncology, European Georges Pompidou Hospital, Assistance Publique Hôpitaux de Paris, University of Paris Cité, Paris, France.
Department of Digestive Oncology, CHU Reims, Université Reims Champagne Ardenne, Reims, France.
Lancet Oncol. 2025 Jun;26(6):732-744. doi: 10.1016/S1470-2045(25)00130-5. Epub 2025 Apr 23.
Perioperative FLOT (fluorouracil, oxaliplatin, and docetaxel) triplet chemotherapy is the standard of care for localised and resectable gastric and gastro-oesophageal junction adenocarcinoma. We aimed to compare a modified FLOT regimen (also known as TFOX) with FOLFOX as first-line treatment for patients with HER2-negative advanced gastric and gastro-oesophageal junction adenocarcinoma.
PRODIGE 51-FFCD-GASTFOX is an open-label, multicentre, randomised, phase 3 trial conducted at 96 medical centres in France. Eligible individuals were aged 18 years or older, had histologically confirmed, HER2-negative adenocarcinoma of the stomach or gastro-oesophageal junction that was locally advanced unresectable or metastatic and previously untreated, measurable disease per Response Evaluation Criteria in Solid Tumours, an Eastern Cooperative Oncology Group performance status of 0 or 1, and adequate organ function. Patients were randomly assigned (1:1), using the minimisation method, to receive FOLFOX (folinic acid 400 mg/m, oxaliplatin 85 mg/m, and 5-fluorouracil bolus 400 mg/m then 5-fluorouracil 2400 mg/m as a continuous 46 h infusion every 2 weeks) or TFOX (docetaxel 50 mg/m, folinic acid 400 mg/m, and oxaliplatin 85 mg/m then 5-fluorouracil 2400 mg/m as a continuous 46 h infusion every 2 weeks). Randomisation was stratified by centre, ECOG performance status, (neo)adjuvant chemotherapy or chemoradiotherapy, tumour stage, tumour location, and pathological histological subtype. The primary endpoint was progression-free survival (assessed in the intention-to-treat population), defined as time from randomisation to the first radiological or clinical progression (or both), or death due to any cause, whichever occurred first. Secondary endpoints included overall survival (defined as time from randomisation to death due to any cause) and objective response rate (defined as the proportion of patients with a best overall complete or partial response). Hazard ratio and 95% CIs were estimated using an unstratified Cox proportional hazards model. When the proportional hazards assumption was violated, the restricted mean survival time was used to estimate the treatment effect size. This study is registered with ClinicalTrials.gov, NCT03006432, and EudraCT, 2016-002331-16.
Between Dec 19, 2016, and Dec 26, 2022, 507 patients were randomly assigned (254 to the TFOX group and 253 to the FOLFOX group [intention-to-treat population]). The median age was 64·2 years (IQR 56·7-70·8), and 399 (79%) participants were male and 108 (21%) were female. At median follow-up of 42·8 months (25·8-49·9), the median progression-free survival was 7·59 months (95% CI 7·06-7·95) in the TFOX group versus 5·98 months (5·65-6·97) in the FOLFOX group. The assumption of proportional hazards was violated (p=0·013); therefore, the 12-month restricted mean progression-free survival was calculated: 7·52 months (7·06-7·97) in the TFOX group versus 6·62 months (6·16-7·09) in the FOLFOX group (p=0·0072). The median overall survival was 15·08 months (13·70-16·72) in the TFOX group versus 12·65 months (10·94-14·00) in the FOLFOX group (proportional hazards assumption was confirmed; HR 0·82 [0·68-0·99]; p=0·048) and the objective response rate was 62·3% (56·0-68·3) versus 53·4% (47·0-59·8; p=0·045). The most common grade 3 and 4 treatment-emergent adverse events were diarrhoea (37 [15%] in the TFOX group vs 18 [7%] in the FOLFOX group), peripheral neuropathy (80 [32%] vs 49 [20%]), neutropenia (67 [27%] vs 44 [18%]), and fatigue (40 [16%] vs 20 [8%]). Serious treatment-related adverse events occurred in 66 (27%) participants in the TFOX group and 33 (13%) in the FOLFOX group. There were two (<1%) treatment-related deaths in the TFOX group (one due to septic shock and one due to gastrointestinal perforation) and one (<1%) in the FOLFOX group (due to septic shock).
The modified FLOT/TFOX regimen significantly improved progression-free survival, overall survival, and objective response rate compared with FOLFOX in previously untreated patients with advanced HER2-negative gastric and gastro-oesophageal junction adenocarcinoma. The modified FLOT/TFOX regimen might represent a new first-line treatment option for patients eligible for this docetaxel triplet chemotherapy.
Fédération Francophone de Cancérologie Digestive.
围手术期氟尿嘧啶、奥沙利铂和多西他赛三联化疗是局部可切除的胃及胃食管交界腺癌的标准治疗方案。我们旨在比较改良的FLOT方案(也称为TFOX)与FOLFOX方案作为人表皮生长因子受体2(HER2)阴性的晚期胃及胃食管交界腺癌患者的一线治疗方案。
PRODIGE 51-FFCD-GASTFOX是一项在法国96个医疗中心进行的开放标签、多中心、随机、3期试验。符合条件的个体年龄在18岁及以上,组织学确诊为胃或胃食管交界的HER2阴性腺癌,局部晚期不可切除或转移性且既往未接受过治疗,根据实体瘤疗效评价标准有可测量的疾病,东部肿瘤协作组体能状态为0或1,且器官功能良好。患者采用最小化法随机分配(1:1),接受FOLFOX方案(亚叶酸钙400mg/m²、奥沙利铂85mg/m²、5-氟尿嘧啶推注400mg/m²,然后5-氟尿嘧啶2400mg/m²持续46小时静脉滴注,每2周一次)或TFOX方案(多西他赛50mg/m²、亚叶酸钙400mg/m²、奥沙利铂85mg/m²,然后5-氟尿嘧啶2400mg/m²持续46小时静脉滴注,每2周一次)。随机分组按中心、东部肿瘤协作组体能状态、(新)辅助化疗或放化疗、肿瘤分期、肿瘤位置和病理组织学亚型进行分层。主要终点是无进展生存期(在意向性治疗人群中评估),定义为从随机分组到首次影像学或临床进展(或两者)或任何原因导致的死亡的时间,以先发生者为准。次要终点包括总生存期(定义为从随机分组到任何原因导致的死亡的时间)和客观缓解率(定义为最佳总体完全缓解或部分缓解的患者比例)。使用未分层的Cox比例风险模型估计风险比和95%置信区间。当比例风险假设被违反时,使用受限平均生存时间来估计治疗效果大小。本研究已在ClinicalTrials.gov注册,注册号为NCT03006432,在欧盟临床试验数据库注册,注册号为2016-002331-16。
2016年12月19日至2022年12月26日期间,507例患者被随机分配(254例至TFOX组,253例至FOLFOX组[意向性治疗人群])。中位年龄为64.2岁(四分位间距56.7 - 70.8),399例(79%)参与者为男性,108例(21%)为女性。中位随访42.8个月(25.8 - 49.9),TFOX组的中位无进展生存期为7.59个月(95%置信区间7.06 - 7.95),而FOLFOX组为5.98个月(5.65 - 6.97)。比例风险假设被违反(p = 0.013);因此,计算了12个月的受限平均无进展生存期:TFOX组为7.52个月(7.06 - 7.97),FOLFOX组为6.62个月(6.16 - 7.09)(p = 0.0072)。TFOX组的中位总生存期为15.08个月(13.70 - 16.72),而FOLFOX组为12.65个月(10.94 - 14.00)(比例风险假设得到证实;风险比0.82[0.68 - 0.99];p = 0.048),客观缓解率为62.3%(56.0 - 68.3),而FOLFOX组为53.4%(47.0 - 59.8;p = 0.045)。最常见的3级和4级治疗中出现的不良事件为腹泻(TFOX组37例[15%],FOLFOX组18例[7%])、周围神经病变(80例[32%]对49例[20%])、中性粒细胞减少(67例[27%]对44例[18%])和疲劳(40例[16%]对20例[8%])。TFOX组66例(27%)参与者和FOLFOX组33例(13%)发生了严重的治疗相关不良事件。TFOX组有2例(<1%)治疗相关死亡(1例因感染性休克,1例因胃肠道穿孔),FOLFOX组有1例(<1%)(因感染性休克)。
与FOLFOX方案相比,改良的FLOT/TFOX方案在既往未接受过治疗的HER2阴性晚期胃及胃食管交界腺癌患者中显著改善了无进展生存期、总生存期和客观缓解率。改良的FLOT/TFOX方案可能是适合接受这种多西他赛三联化疗的患者的一种新的一线治疗选择。
法语国家消化肿瘤联合会。