Next Article in Journal
Effectiveness and Safety of an Outpatient Program for Percutaneous Left Atrial Appendage Occlusion
Previous Article in Journal
Cardio-Ankle Vascular Index and Aging: Differences between CAVI and CAVI0
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Updated Adjuvant Chemotherapy for Gastric Cancer

1
Department of Medical Oncology, Tonan Hospital, Sapporo 060-0004, Japan
2
Department of Medical Oncology, Daido Hospital, Nagoya 457-8511, Japan
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(21), 6727; https://doi.org/10.3390/jcm12216727
Submission received: 25 August 2023 / Revised: 3 October 2023 / Accepted: 18 October 2023 / Published: 24 October 2023
(This article belongs to the Section Gastroenterology & Hepatopancreatobiliary Medicine)

Abstract

:
Surgical resection is currently the best curative approach for gastric cancer (GC); however, the prognosis of patients with advanced GC remains poor even with curative resection. For this reason, perioperative chemotherapy has been combined with surgery to reduce the risk of postoperative recurrence. Standard perioperative chemotherapy for resectable advanced GC varies from region to region. Postoperative S-1 therapy was standardized via the ACTS-GC study in East Asia, perioperative ECF (Epirubicin + Cisplatin + Fluorouracil) was standardized via the MAGIC study in Europe, and postoperative chemoradiotherapy was standardized via the US intergroup study in North America. Since then, more intensive regimens have been developed. In recent years, perioperative therapy using novel agents, such as molecular-targeted drugs and immune checkpoint inhibitors (ICIs), has also been tested and evaluated in the three major regions (East Asia, Europe, and North America) with promising results. Perioperative chemotherapy has become an integral part of many treatment strategies and requires continued research and evaluation.

1. Introduction

GC is a highly malignant cancer with high morbidity and mortality rates worldwide. With more than 1 million newly diagnosed cases, GC remains the leading cause of cancer death every year [1].
Surgical resection is currently the most reliable curative approach for GC. Although improvements in surgical techniques and perioperative care have led to decreased surgical morbidity and mortality, the prognosis of patients with advanced GC remains poor even after complete resection [2,3]. The reason is that minimal residual disease (MRD) can remain in the body even after curative resection. Patients with MRD have a high probability of disease recurrence.
Recently, circulating tumor DNA (ctDNA) has been recognized as an important biomarker to assess the recurrence risk of bladder, breast, and colorectal cancers [4,5,6,7]. Postoperative ctDNA can be considered an MRD biomarker offering useful prognostic information regarding recurrence [8]. It has also been reported that ctDNA may be involved in the recurrence of resectable GC [9,10,11]. However, ctDNA assays have not been established in actual clinical practice and need to be further developed and streamlined.
Perioperative chemotherapy has been used worldwide since the 2000s to minimize the risk of recurrence by eliminating MRD.
In recent years, various clinical trials have been conducted in all regions to improve survival by enhancing perioperative chemotherapy.
This article summarizes the perioperative chemotherapy approaches by region.

2. East Asia

2.1. Postoperative Chemotherapy in East Asia (Table 1 and Table 2)

In East Asia, radical gastrectomy with D2 lymph node resection has been the standard surgery for GC. Many studies have investigated postoperative adjuvant chemotherapy, and it is now also considered a standard treatment.
This section summarizes pivotal clinical trials on postoperative chemotherapy.

2.1.1. The ACTS-GC Study

The first convincing evidence from a randomized clinical study for the efficacy of S-1 adjuvant chemotherapy was the phase III Adjuvant Chemotherapy Trial for Gastric Cancer (ACTS-GC) study [12].
In this study, 1059 patients with stage II-III GC who underwent curative gastrectomy with D2 lymph node dissection were enrolled. Patients were randomly assigned to either the S-1 chemotherapy group or the surgery-only group. The primary endpoint was overall survival (OS). The 3-year OS rate was 80.1% in the S-1 group and 70.1% in the surgery-only group. The death hazard ratio (HR) in the S-1 group, as compared with the surgery-only group, was 0.68 (95% confidence interval (CI): 0.52–0.87; p = 0.003). The HR for recurrence-free survival (RFS) at 3 years was 0.62 (95% CI: 0.50–0.77). This analysis was updated after 5 years of follow-up and demonstrated consistent results [13], showing a significant 10.6% improvement in 5-year OS with adjuvant S-1 chemotherapy group compared to the surgery-only group (71.7 vs. 61.1%, HR 0.669, 95% CI: 0.540–0.828), as well as a 12.3% improvement in 5-year RFS compared to the surgery-only group (65.4 vs. 53.1%, HR 0.653, 95% CI: 0.537–0.793). Along with the marked efficacy, low rates of grades 3 and 4 toxicities were observed (6.0% anorexia, 3.7% nausea, and 3.1% diarrhea). Based on this study, S-1 was established as a standard regime of postoperative adjuvant chemotherapy in East Asia for patients with locally advanced GC who underwent radical surgery.
However, it is not clear whether the efficacy of adjuvant S-1, as demonstrated in the ACTS-GC study, is also achievable in Western populations where this drug is poorly tolerated because of CYP2A6 polymorphisms, by which tegafur present in S-1 transforms in the body to 5-FU [14].

2.1.2. The CLASSIC Study

Capecitabine is an oral fluoropyrimidine agent administered worldwide. The Capecitabine and Oxaliplatin Adjuvant Study in Stomach Cancer (CLASSIC) study was a phase III study that was conducted in East Asia. This study reported a significant efficacy regarding disease-free survival (DFS) after 6 months of adjuvant (postoperative) capecitabine plus oxaliplatin (XELOX) following gastrectomy with D2 lymph node dissection compared to surgery alone [15].
In this study, 1035 patients from 37 Asian centers with stages II–IIIB gastric adenocarcinoma who underwent curative gastrectomy with D2 lymph node dissection were randomly assigned to either the surgery-only group or the adjuvant XELOX group. Patients assigned to the XELOX group had significantly improved 3-year DFS (74 vs. 59%, HR 0.56, 95% CI: 0.44–0.72), with only a borderline statistically significant improvement in 3-year OS rate (83 vs. 78%, HR 0.72, 95% CI: 0.52–1.00) at a median follow-up of 34.2 months.
However, with longer follow-up, the improved OS with XELOX became statistically significant (5-year OS 78 vs. 69%, HR 0.66, 95% CI: 0.51–0.85) [16]. The adjuvant XELOX group had a higher rate of grades 3 and 4 toxicities (56 vs. 6% for the surgery-only group), most commonly neutropenia, nausea, vomiting, thrombocytopenia, and anorexia, requiring chemotherapy dose modifications in 90% of patients. Only 67% of patients assigned to the adjuvant XELOX group completed 6 months of chemotherapy compared to 78% in the ACTS-GC study. The CLASSIC study established XELOX as one of the new treatment options for postoperative chemotherapy in GC patients who underwent gastrectomy with D2 lymph node dissection.

2.1.3. The JACCRO GC-07 Study (START-2)

Docetaxel (DTX) has shown efficacy in advanced or recurrent GC not only as a monotherapy but also in combination with fluoropyrimidine plus cisplatin [17,18,19,20,21,22].
The JACCRO GC-07 was a phase III study conducted to prove the superiority of postoperative S-1 plus DTX. This study enrolled patients who underwent curative resection via D2 or more extensive gastrectomy and were diagnosed with stage III GC. They were randomly assigned to either postoperative S-1 plus DTX or postoperative S-1 alone [23].
Chemotherapy was to be started within 6 postoperative weeks. Patients assigned to the S-1 plus docetaxel group were treated with S-1 on days 1 to 14 of a 3-week cycle during the first course. In the second to seventh courses, patients received intravenous infusion of DTX on day 1 of each cycle and S-1 on days 1 to 14 of a 3-week cycle. From the eighth course, treatment with S-1 continued on days 1 to 28 of 6-week cycles for up to 1 year. Patients assigned to the S-1 group were treated with S-1 every 6-week cycle for up to 1 year. The primary endpoint was 3-year RFS.
The interim analysis of RFS proved the superiority of the S-1 plus DTX (HR 0.632; 99.99% CI: 0.400–0.998; p < 0.001), with a 3-year RFS of 66% (95% CI: 59–73%) in the S-1 plus DTX group and 50% (95% CI: 41–58%) in the S-1 group. Median RFS was not reached in the S-1 plus DTX group, whereas in the S-1 group, it was 34.5 months (95% CI: 29.5 months—not reached).
The S-1 plus DTX group had a higher incidence of grades 3 or 4 toxicities, especially neutropenia and leukopenia, than the S-1 group, but all toxicities were manageable.

2.1.4. The ARTIST 2 Study

In North America, the Intergroup-0116 study [24] reported the effectiveness of adjuvant chemoradiotherapy. However, the effectiveness of chemoradiotherapy in patients who underwent gastrectomy with D2 lymph node dissection remains controversial, despite positive data being reported [25]. This study compared three adjuvant regimens: oral S-1 for 1 year, S-1 plus oxaliplatin (SOX) for 6 months, and SOX plus radiotherapy 45 Gy (SOXRT) in a 1:1:1 ratio [26]. The primary endpoint was 3-year DFS; a 33% reduction in the HR for DFS with SOX or SOXRT, when compared with S-1, was considered clinically meaningful.
In this study, 546 patients were randomly assigned to either the S-1, SOX, or SOXRT group. After a median follow-up of 47 months, the 3-year DFS rates were 64.8, 74.3, and 72.8% in the S-1, SOX, and SOXRT groups, respectively. HR for DFS in the control arm (S-1 group) was lower than that in the SOX and SOXRT groups: S-1 versus SOX, 0.692 (p = 0.042); S-1 versus SOXRT, 0.724 (p = 0.074). There was no statistical difference in DFS between SOX and SOXRT (HR 0.971; p = 0.879). The regimens were generally well tolerated, and adverse events were manageable.
This study showed the efficacy of SOX or SOXRT in patients who underwent curative gastrectomy with D2 lymph node dissection, specifically those with pStage II or III node-positive GC. However, this study did not confirm the efficacy of radiotherapy.

2.1.5. Final Notes

Postoperative adjuvant chemotherapy can improve the cure rate after resection and is, thus, recommended for pStage II to III. Although combination therapy is recommended for pStage III, it is not clear what therapy is more effective because there is no direct comparison of the three regimens (S-1/docetaxel, SOX, and XELOX).
Table 1. Pivotal randomized controlled clinical trial for postoperative chemotherapy in East Asia.
Table 1. Pivotal randomized controlled clinical trial for postoperative chemotherapy in East Asia.
Study NPublished YearAreaSettingDisease TypeRecommended ResectionTreatmentSurvivalHRReference
ACTS-GC5302007JapanStage II/IIIgastric cancer 100%D2Surgery alone5-year OS: 61%0.669[12,13]
529AC 100%S-15-year OS: 72%
CLASSIC5152014KoreaStage II/IIIBgastric cancer 98%,GEJ 2%D2Surgery5-year OS: 69%0.66[15,16]
520AC 100%XELOX5-year OS: 78%
JACCRO GC-074592019JapanStage IIIgastric cancer 100%D2S-13-year RFS:50%0.632[23]
454AC 100%S-1 + DTX3-year RFS:66%
ARTIST 21822021KoreaStage II/IIIgastric cancer 100%D2S-13-year DFS:65%-[26]
181SOX3-year DFS:74%0.692
AC 100%
183SOX + RT (45 Gy)3-year DFS:73%0.724
GEJ—gastroesophageal junction; AC—adenocarcinoma; OS—overall survival; RFS—recurrence-free survival; DFS—disease-free survival; XELOX—capecitabine and oxaliplatin; SOX—S-1 and oxaliplatin; RT—radiotherapy; HR—hazard ratio.
Table 2. Postoperative chemotherapy regimens in East Asia.
Table 2. Postoperative chemotherapy regimens in East Asia.
RegimenFrequencyDrugDosePeriod
S-16 weeksS-140–80 mg/body, twice dailydays 1–28every 6 weeks for 1 year
XELOX (CapeOx)3 weeksCapecitabine1000 mg/m2, twice dailydays 1–148 cycles
Oxaliplatine130 mg/m2day 1
S-1 + DTX(1) 3 weeksS-140–80 mg/body, twice dailydays 1–141 cycle
(2) 3 weeksS-140–80 mg/body, twice dailydays 1–146 cycles
Docetaxel40 mg/m2day 1
(3) 6 weeksS-140–80 mg/body, twice dailydays 1–28every 6 weeks for total 1 year
SOX3 weeksS-140–80 mg/body, twice dailydays 1–148 cycles
Oxaliplatine130 mg/m2day 1
SOX + RT(1) 3 weeksS-140–80 mg/body, twice dailydays 1–142 cycles
Oxaliplatine130 mg/m2day 1
(2) 5 weeksS-140–80 mg/body, twice daily5 days a week, over 5 weeks
Radiotherapy45 Gy/25 Fr
(3) weeksS-140–80 mg/body, twice dailydays 1–144 cycles
Oxaliplatine130 mg/m2day 1

2.2. Neoadjuvant Chemotherapy in East Asia (Table 3 and Table 4)

As mentioned above, postoperative chemotherapy is the standard treatment in East Asia.
On the other hand, neoadjuvant chemotherapy (NAC) has also been investigated, and the results are reported below.

2.2.1. JCOG0501

Even with standard perioperative therapy and curative surgery, GCs with linitis plastica (Borrmann type 4) and large ulcero-invasive-type (≥8 cm, Borrmann type 3) have a very poor prognosis. JCOG0501 is a randomized phase III study conducted to evaluate the survival benefit of neoadjuvant (preoperative) chemotherapy of SP (S-1 + CDDP) for postoperative S-1 in resectable type 4 and large type 3 GCs in Japan [27].
Patients assigned to Arm A underwent total or distal gastrectomy with D2 or D3 lymph node dissection. Patients in Arm B received preoperative chemotherapy with SP (S-l administered for the first 3 weeks of a 4-week course, and Cisplatin administered on day 8 of each course). All patients who underwent curative gastrectomy received postoperative chemotherapy with S-1 orally on days 1–28 of each 6-week cycle for 1 year.
A total of 300 patients were enrolled in this study. The 3-year OS rates were 62.4% (95% CI: 54.1–69.6) in Arm A and 60.9% (95% CI: 52.7–68.2) in Arm B. The HR in Arm B, as compared with Arm A, was 0.916 (95% CI: 0.679–1.236).
For Borrmann type 4 or large type 3 GC, the addition of preoperative chemotherapy with SP did not show a survival benefit. The Japanese Gastric Cancer Treatment Guidelines do not recommend neoadjuvant chemotherapy, and still continue to recommend D2 gastrectomy followed by postoperative chemotherapy [28].

2.2.2. PRODIGY

This study was conducted to evaluate whether neoadjuvant (preoperative) docetaxel, oxaliplatin, and S-1 (DOS) followed by surgery and adjuvant (postoperative) S-1 could improve outcomes versus standard treatment (surgery and adjuvant S-1) in patients with resectable GC in Korea [29].
Patients with histologically confirmed primary gastric or gastroesophageal junction adenocarcinoma (clinical TNM staging: T2-3N1 or T4N any) were enrolled. They were randomly assigned to either D2 surgery followed by adjuvant S-1 (SC group) or neoadjuvant DOS before D2 gastrectomy, followed by adjuvant S-1 (CSC group). The primary endpoint was PFS.
In this study, 266 patients were assigned to either the SC group or the CSC group. After a median follow-up of 38.6 (interquartile range, 23.5–62.1) months, PFS was significantly longer in the CSC (HR for PFS adjusted for stratification factors, 0.70; 95% CI: 0.52–0.95; stratified log-rank p = 0.023). The 3-year PFS rates were 66.3% (95% CI: 59.6–72.1) with CSC and 60.2% (95% CI: 53.6–66.3) with SC. Two Grade 5 adverse events (febrile neutropenia and dyspnea) occurred during neoadjuvant chemotherapy. Other than that, treatments were well tolerated.
This study showed the effectiveness of neoadjuvant DOS in Korea.

2.2.3. RESOLVE

This study was conducted to evaluate the efficacy of perioperative (preoperative and postoperative) SOX (S-1 + oxaliplatin) compared with adjuvant (postoperative) CapeOX (Capecitabine + oxaliplatin) in patients with resectable GC undergoing gastrectomy with D2 lymph node dissection in China [30].
This study enrolled patients with histologically confirmed cT4a, N+, M0 or cT4b, N any, M0 gastric or gastroesophageal junction adenocarcinoma who underwent D2 gastrectomy. Patients were randomly assigned (1:1:1) to receive postoperative CapeOX (eight cycles), postoperative SOX (eight cycles), or perioperative SOX (three preoperative cycles and five postoperative cycles followed by three cycles of S-1 monotherapy). The primary endpoint was 3-year DFS to assess the potential superiority of perioperative SOX compared with postoperative SOX and the non-inferiority of postoperative SOX compared with postoperative CapeOX.
A total of 1022 patients were enrolled and randomly assigned to either the postoperative CapeOX group, the postoperative SOX group, or the perioperative SOX group. Three-year DFS was 51.1% (95% CI: 45.5–56.3) in the postoperative CapeOX group, 56.5% (95% CI: 51.0–61.7) in the postoperative SOX group, and 59.4% (95% CI: 53.8–64.6) in the perioperative SOX group. The HR was 0.77 (95% CI: 0.61–0.97; Wald p = 0.028) for the perioperative SOX group compared with the postoperative CapeOX group and 0.86 (95% CI: 0.68–1.07; Wald p = 0.17) for the postoperative SOX group compared with the postoperative CapeOX group.
Perioperative SOX showed clinically meaningful benefits compared with postoperative CapeOX, and postoperative SOX was non-inferior to postoperative CapeOX.
Table 3. Pivotal randomized controlled clinical trial for neoadjuvant chemotherapy in East Asia.
Table 3. Pivotal randomized controlled clinical trial for neoadjuvant chemotherapy in East Asia.
Study NPublished YearAreaSettingDisease TypeRecommended ResectionTreatmentSurvivalHRReference
JCOG 05011492021JapanBorrmann type 3 (≧8 cm)gastric 100%D2/3NAC SP → Adj S-13-year OS: 62%0.916[27]
151Borrmann type 4AC 100% Adj S-13-year OS: 61%
PRODIGY2382021KoreaStage II/IIIgastric 94%,GEJ 6%D2NAC DOS → Adj S-13-year PFS: 66%0.69[29]
246AC 100%Adj S-13-year PFS: 60%
RESOLVE3372021ChinaStage IIIgastric 64%, GEJ 36%D2Perioperative SOX3-year DFS:59%0.77[30]
340Adj SOX3-year DFS: 56%
AC 100%0.86
345Adj Capox3-year DFS:51%
GEJ—gastroesophageal junction; AC—adenocarcinoma; OS—overall survival; PFS—progression-free survival; DFS—disease-free survival; Capox—capecitabine and oxaliplatin; SOX—S-1 and oxaliplatin; HR—hazard ratio.
Table 4. Neoadjuvant chemotherapy regimens in East Asia.
Table 4. Neoadjuvant chemotherapy regimens in East Asia.
StudyRegimenFrequencyDrugDosePeriod
JCOG0501SP4 weeksS-140–80 mg/body, twice dailydays 1–21Preoperative; 2 cycles
Cisplatine60 mg/m2day 8
S-16 weeksS-140–80 mg/body, twice dailydays 1–28Postoperative; every 6 weeks for 1 year
PRODIGYDOS3 weeksDocetaxel50 mg/m2day 1Preoperative; 3 cycles
Oxaliplatine100 mg/m2day 1
S-140–80 mg/body, twice dailydays 1–14
S-16 weeksS-140–80 mg/body, twice dailydays 1–28Postoperative; every 6 weeks for 1 year
RESOLVESOX3 weeksOxaliplatine130 mg/m2day 1Preoperative; 3 cycles
S-140–80 mg/body, twice dailydays 1–14Postoperative; 5 cycles
S-13 weeksS-140–80 mg/body, twice dailydays 1–14After above; 3 cycles

2.2.4. Final Notes

As mentioned above, perioperative chemotherapy is also being evaluated in East Asia.
In China, the RESONANCE trial, a phase III study, using SOX as NAC for clinical stages (cStage) IIA-IIIC, reported that 67.8% of patients receiving NAC showed pathological efficacy, and 23.6% showed complete response without significant differences in short-term surgical outcomes [31].
Treatment strategies may further change depending on the results of other ongoing studies.

3. Europe

3.1. Perioperative Chemotherapy in Europe (Table 5 and Table 6)

The European Society for Medical Oncology (ESMO) clinical practice guidelines suggest gastrectomy with D2 lymph node dissection be performed and recommend perioperative chemotherapy preferentially for patients with stage IB-III GC.
This section summarizes pivotal clinical trials on perioperative chemotherapy in Europe.

3.1.1. MAGIC

A regimen of epirubicin, cisplatin, and infused fluorouracil (ECF) has been reported to improve survival in patients with incurable locally advanced or metastatic GC in Europe [32,33,34]. However, numerous phase III trials at these times did not show the survival benefits of adjuvant chemotherapy.
The Medical Research Council Adjuvant Gastric Infusional Chemotherapy (MAGIC) trial was conducted to prove the efficacy of perioperative ECF for patients with potentially resectable GC [35].
This study enrolled patients with resectable adenocarcinoma of the stomach, esophagogastric junction, or lower esophagus. A total of 503 patients were randomly assigned to either the perioperative ECF group or the surgery-only group. The ECF regimen consisted of a 3-week cycle of intravenous epirubicin and cisplatin on day 1, plus continuous intravenous infusion of fluorouracil on days 1 to 21. Patients assigned to the ECF group received three preoperative and three postoperative cycles of ECF. The primary endpoint was OS.
This study showed a significant improvement in OS in the perioperative ECF group compared to the surgery-only group (HR for death, 0.75; 95% CI: 0.60-0.93; p = 0.009; 5-year survival rate, 36% vs. 23%), as well as an improvement in PFS (HR for progression, 0.66; 95% CI: 0.53–0.81; p < 0.001). The resected tumors were smaller and less advanced in the perioperative ECF group. The adverse effects of ECF were similar to those previously reported among patients with locally advanced and metastatic GC.
Based on the results of this study, ECF has become the standard of perioperative chemotherapy in Europe.

3.1.2. FLOT4

ECF regimens have been considered standard perioperative chemotherapy. However, the overall survival rate is not satisfactory.
Docetaxel has shown efficacy in metastatic GC, both first-line (docetaxel, cisplatin, and fluorouracil (DCF)) [22] and second-line (docetaxel monotherapy) therapy [36]. However, the parental DCF regimen was associated with high toxicity, so a regimen was developed to replace cisplatin with oxaliplatin to reduce toxicities. The combination of fluorouracil, leucovorin, oxaliplatin, and docetaxel (FLOT) was evaluated for locally advanced and metastatic GCs and gastroesophageal junction adenocarcinoma in several phase II studies, and showed that FLOT was better tolerated than DCF and induced stronger tumor response than other regimens, including anthracycline-based triplets [37,38,39,40,41,42].
FLOT4 was conducted to evaluate the efficacy of FLOT as a perioperative chemotherapy for patients with locally advanced, resectable GC [43]. This study enrolled patients with histologically confirmed advanced clinical stage cT2-4 or nodal positive stage (cN+), or both, resectable tumors, with no evidence of distant metastases (cM0). Patients were randomly assigned to either the ECF/ECX group (three preoperative and three postoperative 3-week cycles of epirubicin and cisplatin on day 1 plus either fluorouracil or capecitabine on days 1 to 21) or the FLOT group (four preoperative and four postoperative 2-week cycles of docetaxel, oxaliplatin, leucovorin, and fluorouracil as a 24 h infusion on day 1). The primary endpoint was OS.
A total of 716 patients were randomly assigned to either the ECF/ECX group or the FLOT group. OS improved in the FLOT group compared with the ECF/ECX group (HR 0.77 (95% CI: 0.63–0.94); median OS, 50 months (38.33 to not reached) vs. 35 months (27.35 to 46.26)). The number of patients with related serious adverse effects was similar in the two groups (27% in the ECF/ECX group vs. 27% in the FLOT group). Hospitalization for toxicities was necessary in 26% of ECF/ECX patients and 25% of FLOT patients.
FLOT improved OS without increasing perioperative complications compared with perioperative ECF/ECX in resectable gastric or gastroesophageal junction adenocarcinoma.
Table 5. Pivotal randomized controlled clinical trial for perioperative therapy in Europe.
Table 5. Pivotal randomized controlled clinical trial for perioperative therapy in Europe.
Study NPublished YearAreaSettingDisease TypeRecommended ResectionTreatmentSurvivalHRReference
MAGIC2502006UKNo evidence of distant metastases, or locally advanced inoperable diseasegastric 74%, Lower esophageal/GEJ 26%undefinedsurgery5-year OS: 23%0.75[35]
253AC 100%ECF5-year OS: 36%
FLOT 43602011FrancecT2 or higher nodal positive stage (cN+), or bothgastric 44%, GEJ 56%D2ECFmedian OS: 35 months0.77[43]
356AC 100%FLOTmedian OS: 50 months
GEJ—gastroesophageal junction; AC—adenocarcinoma; ECF—epirubicin, cisplatin and 5-fluorouracil; FLOT—fluorouracil, leucovorin, oxaliplatin and docetaxel; OS—overall survival; HR—hazard ratio.
Table 6. Perioperative chemotherapy regimens in Europe.
Table 6. Perioperative chemotherapy regimens in Europe.
RegimenFrequencyDrugDosePeriod
ECF3 weeksEpirubicin50 mg/m2day 1Preoperative; 3 cycles
Cisplatine60 mg/m2
Fluorouracil200 mg/m2/daydays 1–21Postoperative; 3 cycles
ECX3 weeksEpirubicin50 mg/m2day 1Preoperative; 3 cycles
Cisplatine60 mg/m2
Capecitabine1250 mg/m2days 1–21Postoperative; 3 cycles
FLOT2 weeksDocetaxel50 mg/m2day 1Preoperative; 4 cycles
Oxaliplatine85 mg/m2
Leucovorin200 mg/m2Postoperative; 4 cycles
Fluorouracil2600 mg/m2, 24-h infusion

3.1.3. Final Notes

Perioperative chemotherapy using triplet regimens (fluorinated pyrimidines, platinum, and taxanes) is the standard treatment in Europe. However, ESMO guidelines recommend postoperative chemotherapy or chemoradiotherapy for patients who undergo gastrectomy without preoperative therapy [33]. Postoperative chemotherapy has been adopted in Europe based on the results of the ACTS-GC and CLASSIC studies. Although chemoradiotherapy has been established in North America, the ESMO guidelines indicate it is only for suboptimal surgery with less lymphadenectomy and suspicious micrometastases [44,45].

4. North America

4.1. Chemoradiotherapy in North America (Table 7 and Table 8)

Postoperative chemotherapy in East Asia and preoperative chemotherapy in Europe have been considered the standard treatments, but postoperative chemoradiation therapy has been considered the standard in North America.
The National Comprehensive Cancer Network (NCCN) guidelines agree that gastrectomy with D2 lymph node dissection is associated with low mortality and reasonable survival benefits. However, the NCCN guidelines recommend gastrectomy with D1 or modified D2 lymph node dissection and also emphasize that D2 lymph node dissection should be performed only by experienced surgeons because of its technical difficulty [46,47].
Therefore, different perioperative treatments are recommended in North America compared to other regions. This section summarizes the situation with a focus on postoperative chemoradiotherapy.
Table 7. Pivotal randomized controlled clinical trial for postoperative chemoradiotherapy in North America.
Table 7. Pivotal randomized controlled clinical trial for postoperative chemoradiotherapy in North America.
Study NPublished YearAreaSettingDisease TypeRecommended ResectionTreatmentSurvivalHRReference
INT-01162752001USStage IB–IV M0gastric 80%, GEJ 20%D2 (D0 54%, D1 36%, D2 10%)surgerymedian OS: 27 months1.35[24]
281AC 100%Fluorouracil + leucovorin + Radiation (45 Gy)median OS: 36 months
GEJ—gastroesophageal junction; AC—adenocarcinoma; OS—overall survival; HR—hazard ratio.
Table 8. Chemoradiotherapy regimen in North America.
Table 8. Chemoradiotherapy regimen in North America.
RegimenFrequencyDrugDosePeriod
INT-01164 weeks 5-FU425 mg/m2days 1–5Preradiotherapy; 1 cycle
Leucovorin20 mg/m2
5 weeksRadiotherapy45 Gy/25 Fr5 days a week, over 5 weeks
5-FU400 mg/m2the first 4 days and the last 3 days of radiotherapy
Leucovorin20 mg/m2
4 weeks 5-FU425 mg/m2days 1–5Postradiotherapy; 2 cycles
Leucovorin20 mg/m2

4.1.1. SWOG9008/INT-0116

The curative treatment for GC is resection, but most patients were not successfully treated via surgery alone in the 1990s in North America. This study was designed to prove the efficacy of postoperative chemoradiotherapy for patients with resectable adenocarcinoma [24].
A total of 556 patients with resected adenocarcinoma of the stomach or gastroesophageal junction were randomized to the surgery plus postoperative chemoradiotherapy group or the surgery-only group. The adjuvant treatment consisted of fluorouracil plus 20 mg/m2/day of leucovorin, for five days, followed by 4500 cGy of radiation at 180 cGy/day, administered five days per week for five weeks, with modified doses of fluorouracil and leucovorin on the first four and last three days of radiotherapy.
One month after the completion of radiotherapy, two five-day cycles of fluorouracil plus leucovorin were administered one month apart.
The median OS in the surgery-only group was 27 months compared with 36 months in the chemoradiotherapy group (death HR was 1.35 (95% CI: 1.09–1.66; p = 0.005)). The HR for relapse was 1.52 (95% CI: 1.23–1.86; p < 0.001). Three patients (1%) died due to chemoradiotherapy toxicities; grade 3 toxicities occurred in 41% of the patients in the chemoradiotherapy group, and grade 4 toxicities occurred in 32% of patients.
In a later updated analysis, this study demonstrated strong persistent benefits from postoperative chemoradiotherapy [48]. This study established postoperative chemoradiotherapy in North America. The NCCN guidelines now recommend it only for patients who undergo gastrectomy with less than a D2 lymph node dissection [49].

4.1.2. Final Notes

Postoperative chemoradiotherapy was established in North America and is still recommended in certain cases. Perioperative chemotherapy is also recommended, according to the results of the FLOT4 study [43]. Regarding postoperative chemotherapy, the NCCN guidelines state that it is difficult to apply the results of the ACTS-GC and CLASSIC studies because gastrectomy with D2 lymph node dissection is rarely performed in North American hospitals [50]. Postoperative chemotherapy with XELOX is included only as an option for patients who undergo D2 or modified D2 lymph node dissection [51]. In North America, perioperative treatment is selected based on the circumstances described above.

5. New Agents for Adjuvant Chemotherapy

In recent years, new drugs such as molecular-targeted drugs and ICIs have improved the prognosis of GC [52,53,54,55]. Next, we summarize the trials of perioperative treatment using these agents. Many of these trials were designed around existing regimens containing fluoropyrimidines, platinum, and taxanes. Also, unlike previous trials, these were conducted across multiple regions.

5.1. Molecular-Targeted Drugs (Table 9)

The RAMSES-AIO study examined the utility of a trial treatment adding ramucirumab to perioperative FLOT therapy [56]. The results suggested that the addition of ramucirumab may increase the R0 resection rate.
The PETRARCA-AIO study examined the addition of trastuzumab to conventional FLOT for HER2-positive cancer, and the results suggested an increase in pCR and nodal negativity rates [57].
EORTC also conducted the INNOVATION study to verify the efficacy of perioperative chemotherapy, comparing XP/FP only to experimental treatments with the addition of trastuzumab or trastuzumab and pertuzumab for HER2-positive GC [58].
Table 9. New agents for adjuvant chemotherapy/Molecular-targeted drugs.
Table 9. New agents for adjuvant chemotherapy/Molecular-targeted drugs.
StudyRegimenFrequencyDrugDosePeriod
RAMSESFLOT2 weeksDocetaxel50 mg/m2day 1Preoperative; 4 cycles
Oxaliplatine85 mg/m2
Leucovorin200 mg/m2
Fluorouracil2600 mg/m2, 24 h infusionPostoperative; 4 cycles
Ramcirumab8 mg/kgday 1
Ramcirumab2 weeks8 mg/kgday 1After above; 16 cycles
PETRARCAFLOT2 weeksDocetaxel50 mg/m2day 1Preoperative; 4 cycles
Oxaliplatine85 mg/m2
Leucovorin200 mg/m2Postoperative; 4 cycles
Fluorouracil2600 mg/m2, 24 h infusion
Trastuzumab3 weeks[Initial dose] 8 mg/kgday 1Preoperative; 3 cycles
[Maintenance dose] 6 mg/kgPostoperative; 3 cycles
Perutuzumab840 mgAfter above; 9 cycles
INNOVATIONXELOX (CapOx)3 weeksCapecitabine1000 mg/m2, twice dailydays 1–14Preoperative; 3 cycles
Oxaliplatine130 mg/m2day 1
XPCapecitabine1000 mg/m2, twice dailydays 1–14
Cisplatine80 mg/m2day 1Postoperative; 3 cycles
FPFluorouracil800 mg/m2days 1–5
Cisplatine80 mg/m2day 1
FLOT2 weeksDocetaxel50 mg/m2day 1Preoperative; 4 cycles
Oxaliplatine85 mg/m2
Leucovorin200 mg/m2
Fluorouracil2600 mg/m2, 24 h infusion
FOLFOXOxaliplatine85 mg/m2day 1Postoperative; 4 cycles
Leucovorin400 mg/m2
Fluorouracil[bolus] 400 mg/m2
[continuous infusion] 2400 mg/m2, 46–48 h
Any of the above regimens combined with any of the following regimens
Trastuzumab3 weeks[Initial dose] 8 mg/kgday 1
[Maintenance dose] 6 mg/kgPreoperative; 3 cycles
Perutuzumab840 mgPostoperative; 3 cycles
Trastuzumab[Initial dose] 8 mg/kgday 1After above; 9 cycles
[Maintenance dose] 6 mg/kg

5.2. Immune Checkpoint Inhibitors (Table 10)

The usefulness of immune checkpoint inhibitors has been tested in perioperative treatment as well as in palliative chemotherapy.
The KEYNOTE 585 study evaluated the addition of pembrolizumab to perioperative XP/FP/FLOT [59], and the MATTERHORN study evaluated the addition of durvalumab to perioperative FLOT [60].
In addition, the DANTE study examined the benefits of adding atezolizumab to perioperative FLOT [61].
The results of these clinical trials are expected to change future treatment strategies for GC.
Table 10. New agents for adjuvant chemotherapy/Immune checkpoint inhibitors.
Table 10. New agents for adjuvant chemotherapy/Immune checkpoint inhibitors.
StudyRegimenFrequencyDrugDosePeriod
KEYNOTE-585FLOT2 weeksDocetaxel50 mg/m2day 1Preoperative; 4 cycles
Oxaliplatine85 mg/m2
Leucovorin200 mg/m2Postoperative; 4 cycles
Fluorouracil2600 mg/m2, 24 h infusion
XP3 weeksCapecitabine1000 mg/m2, twice dailyday 1–14Preoperative; 3 cycles
Cisplatine80 mg/m2day 1
FPFluorouracil800 mg/m2day 1–5Postoperative; 3 cycles
Cisplatine80 mg/m2day 1
Any of the above regimens combined with the following regimen
Pembrolizumab3 weeks200 mgday 1Preoperative; 3 cycles
Postoperative; 3 cycles
After above; 11 additional cycles
MATTERHORNFLOT4 weeksDocetaxel50 mg/m2day 1, 15Preoperative; 2 cycles
Oxaliplatine85 mg/m2
Leucovorin200 mg/m2
Fluorouracil2600 mg/m2, 24 h infusionPostoperative; 2 cycles
Durvalumab1500 mgday 1
Durvalumab4 weeks1500 mgday 1After above; 10 additional cycles
DANTEFLOT2 weeksDocetaxel50 mg/m2day 1Preoperative; 4 cycles
Oxaliplatine85 mg/m2
Leucovorin200 mg/m2
Fluorouracil2600 mg/m2, 24 h infusion
FLOT2 weeksDocetaxel50 mg/m2day 1Postoperative; 4 cycles
Oxaliplatine85 mg/m2
Leucovorin200 mg/m2
Fluorouracil2600 mg/m2, 24 h infusion
Atezolizumab840 mgday 1
Atezolizumab3 weeks1200 mgday 1After above; 8 additional cycles

6. Conclusions

The perioperative treatment of GC has undergone significant development in various regions since the 2000s. Perioperative regimens were initially established by drawing from the treatment protocols used for metastatic GC. Consequently, treatment advancements began with S-1 in East Asia and ECF in Europe. Variations in surgical techniques may have also influenced the intensity of chemotherapy, including whether doublet or triplet regimens were employed, as well as the incorporation of radiation therapy. Particularly, in East Asia, more trials have been conducted than in other regions due to the large number of GCs. In this way, perioperative chemotherapy has evolved independently in each region. Now, perioperative chemotherapy based on fluorinated pyrimidines with added platinum and taxanes has been established in all regions with improved outcomes. Perioperative therapy using novel agents has also been tested and evaluated in the three major regions with promising results. Perioperative chemotherapy has become an integral part of many treatment strategies and requires continued research and evaluation.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Lin, J.-X.; Tang, Y.-H.; Lin, G.-J.; Ma, Y.-B.; Desiderio, J.; Li, P.; Xie, J.-W.; Wang, J.-B.; Lu, J.; Chen, Q.-Y.; et al. Association of Adjuvant Chemotherapy With Overall Survival Among Patients With Locally Advanced Gastric Cancer After Neoadjuvant Chemotherapy. JAMA Netw. Open 2022, 5, e225557. [Google Scholar] [CrossRef] [PubMed]
  3. Tegels, J.J.; De Maat, M.F.; Hulsewé, K.W.; Hoofwijk, A.G.; Stoot, J.H. Improving the outcomes in gastric cancer surgery. World J. Gastroenterol. 2014, 20, 13692–13704. [Google Scholar] [CrossRef]
  4. Magbanua, M.; Swigart, L.; Wu, H.-T.; Hirst, G.; Yau, C.; Wolf, D.; Tin, A.; Salari, R.; Shchegrova, S.; Pawar, H.; et al. Circulating tumor DNA in neoadjuvant-treated breast cancer reflects response and survival. Ann. Oncol. 2021, 32, 229–239. [Google Scholar] [CrossRef] [PubMed]
  5. Powles, T.; Assaf, Z.J.; Davarpanah, N.; Banchereau, R.; Szabados, B.E.; Yuen, K.C.; Grivas, P.; Hussain, M.; Oudard, S.; Gschwend, J.E.; et al. ctDNA guiding adjuvant immunotherapy in urothelial carcinoma. Nature 2021, 595, 432–437. [Google Scholar] [CrossRef] [PubMed]
  6. Loupakis, F.; Sharma, S.; Derouazi, M.; Murgioni, S.; Biason, P.; Rizzato, M.D.; Rasola, C.; Renner, D.; Shchegrova, S.; Malashevich, A.K.; et al. Detection of Molecular Residual Disease Using Personalized Circulating Tumor DNA Assay in Patients With Colorectal Cancer Undergoing Resection of Metastases. JCO Precis. Oncol. 2021, 5, 1166–1177. [Google Scholar] [CrossRef]
  7. Reinert, T.; Henriksen, T.V.; Christensen, E.; Sharma, S.; Salari, R.; Sethi, H.; Knudsen, M.; Nordentoft, I.K.; Wu, H.-T.; Tin, A.S.; et al. Analysis of Plasma Cell-Free DNA by Ultradeep Sequencing in Patients With Stages I to III Colorectal Cancer. JAMA Oncol. 2019, 5, 1124–1131. [Google Scholar] [CrossRef] [PubMed]
  8. Mencel, J.; Slater, S.; Cartwright, E.; Starling, N. The Role of ctDNA in Gastric Cancer. Cancers 2022, 14, 5105. [Google Scholar] [CrossRef]
  9. Huffman, B.M.; Aushev, V.N.; Budde, G.L.; Chao, J.; Dayyani, F.; Hanna, D.; Botta, G.P.; Catenacci, D.V.; Maron, S.B.; Krinshpun, S.; et al. Analysis of Circulating Tumor DNA to Predict Risk of Recurrence in Patients With Esophageal and Gastric Cancers. JCO Precis. Oncol. 2022, 6, e2200420. [Google Scholar] [CrossRef]
  10. Kim, Y.-W.; Song, Y.; Kim, H.-S.; Sim, H.W.; Poojan, S.; Eom, B.W.; Kook, M.-C.; Joo, J.; Hong, K.-M. Monitoring circulating tumor DNA by analyzing personalized cancer-specific rearrangements to detect recurrence in gastric cancer. Exp. Mol. Med. 2019, 51, 1–10. [Google Scholar] [CrossRef]
  11. Leal, A.; van Grieken, N.C.T.; Palsgrove, D.N.; Phallen, J.; Medina, J.E.; Hruban, C.; Broeckaert, M.A.M.; Anagnostou, V.; Adleff, V.; Bruhm, D.C.; et al. White blood cell and cell-free DNA analyses for detection of residual disease in gastric cancer. Nat. Commun. 2020, 11, 525. [Google Scholar] [CrossRef] [PubMed]
  12. Sakuramoto, S.; Sasako, M.; Yamaguchi, T.; Kinoshita, T.; Fujii, M.; Nashimoto, A.; Furukawa, H.; Nakajima, T.; Ohashi, Y.; Imamura, H.; et al. Adjuvant Chemotherapy for Gastric Cancer with S-1, an Oral Fluoropyrimidine. New Engl. J. Med. 2007, 357, 1810–1820. [Google Scholar] [CrossRef] [PubMed]
  13. Sasako, M.; Sakuramoto, S.; Katai, H.; Kinoshita, T.; Furukawa, H.; Yamaguchi, T.; Nashimoto, A.; Fujii, M.; Nakajima, T.; Ohashi, Y. 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. J. Clin. Oncol. 2011, 29, 4387–4393. [Google Scholar] [CrossRef] [PubMed]
  14. Tsuji, Y. Adjuvant Chemotherapy: Current Practices, Efficacy and Future Directions. In Cancer Etiology, Diagnosis and Treatments; Nova Medicine & Health: New York, NY, USA, 2018; 327p. [Google Scholar]
  15. Bang, Y.-J.; Kim, Y.-W.; Yang, H.-K.; Chung, H.C.; Park, Y.-K.; Lee, K.H.; Lee, K.-W.; Kim, Y.H.; Noh, S.-I.; Cho, J.Y.; et al. Adjuvant capecitabine and oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): A phase 3 open-label, randomised controlled trial. Lancet 2012, 379, 315–321. [Google Scholar] [CrossRef] [PubMed]
  16. Noh, S.H.; Park, S.R.; Yang, H.-K.; Chung, H.C.; Chung, I.-J.; Kim, S.-W.; Kim, H.-H.; Choi, J.-H.; Kim, H.-K.; Yu, W.; et al. Adjuvant capecitabine plus oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): 5-year follow-up of an open-label, randomised phase 3 trial. Lancet Oncol. 2014, 15, 1389–1396. [Google Scholar] [CrossRef] [PubMed]
  17. Taguchi, T.; Sakata, Y.; Kanamaru, R.; Kurihara, M.; Suminaga, M.; Ota, J.; Hirabayashi, N. Late phase II clinical study of RP56976 (docetaxel) in patients with advanced/recurrent gastric cancer: A Japanese Cooperative Study Group trial (group A). Gan To Kagaku Ryoho 1998, 25, 1915–1924. [Google Scholar] [PubMed]
  18. Yoshida, K.; Hirabayashi, N.; Takiyama, W.; Ninomiya, M.; Takakura, N.; Sakamoto, J.; Nishiyama, M.; Toge, T. Phase I study of combination therapy with S-1 and docetaxel (TXT) for advanced or recurrent gastric cancer. Anticancer Res. 2004, 24, 1843–1851. [Google Scholar] [PubMed]
  19. Yoshida, K.; Ninomiya, M.; Takakura, N.; Hirabayashi, N.; Takiyama, W.; Sato, Y.; Todo, S.; Terashima, M.; Gotoh, M.; Sakamoto, J.; et al. Phase II Study of Docetaxel and S-1 Combination Therapy for Advanced or Recurrent Gastric Cancer. Clin. Cancer Res. 2006, 12, 3402–3407. [Google Scholar] [CrossRef]
  20. Koizumi, W.; The JACCRO and KCSG Study Group; Kim, Y.H.; Fujii, M.; Kim, H.K.; Imamura, H.; Lee, K.H.; Hara, T.; Chung, H.C.; Satoh, T.; et al. Addition of docetaxel to S-1 without platinum prolongs survival of patients with advanced gastric cancer: A randomized study (START). J. Cancer Res. Clin. Oncol. 2014, 140, 319–328. [Google Scholar] [CrossRef]
  21. Wada, Y.; Yoshida, K.; Suzuki, T.; Mizuiri, H.; Konishi, K.; Ukon, K.; Tanabe, K.; Sakata, Y.; Fukushima, M. Synergistic effects of docetaxel and S-1 by modulating the expression of metabolic enzymes of 5-fluorouracil in human gastric cancer cell lines. Int. J. Cancer 2006, 119, 783–791. [Google Scholar] [CrossRef]
  22. Van Cutsem, E.; Moiseyenko, V.M.; Tjulandin, S.; Majlis, A.; Constenla, M.; Boni, C.; Rodrigues, A.; Fodor, M.; Chao, Y.; Voznyi, E.; et al. Phase III Study of Docetaxel and Cisplatin Plus Fluorouracil Compared With Cisplatin and Fluorouracil As First-Line Therapy for Advanced Gastric Cancer: A Report of the V325 Study Group. J. Clin. Oncol. 2006, 24, 4991–4997. [Google Scholar] [CrossRef]
  23. Yoshida, K.; Kodera, Y.; Kochi, M.; Ichikawa, W.; Kakeji, Y.; Sano, T.; Nagao, N.; Takahashi, M.; Takagane, A.; Watanabe, T.; et al. 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. J. Clin. Oncol. 2019, 37, 1296–1304. [Google Scholar] [CrossRef] [PubMed]
  24. Macdonald, J.S.; Smalley, S.R.; Benedetti, J.; Hundahl, S.A.; Estes, N.C.; Stemmermann, G.N.; Haller, D.G.; Ajani, J.A.; Gunderson, L.L.; Jessup, J.M.; et al. Chemoradiotherapy after Surgery Compared with Surgery Alone for Adenocarcinoma of the Stomach or Gastroesophageal Junction. N. Engl. J. Med. 2001, 345, 725–730. [Google Scholar] [CrossRef]
  25. Kim, S.; Lim, D.H.; Lee, J.; Kang, W.K.; MacDonald, J.S.; Park, C.H.; Park, S.H.; Lee, S.-H.; Kim, K.; Park, J.O.; et al. An observational study suggesting clinical benefit for adjuvant postoperative chemoradiation in a population of over 500 cases after gastric resection with D2 nodal dissection for adenocarcinoma of the stomach. Int. J. Radiat. Oncol. 2005, 63, 1279–1285. [Google Scholar] [CrossRef] [PubMed]
  26. Park, S.H.; Lim, D.H.; Sohn, T.S.; Lee, J.; Zang, D.Y.; Kim, S.T.; Kang, J.H.; Oh, S.Y.; Hwang, I.G.; Ji, J.H.; et al. A randomized phase III trial comparing adjuvant single-agent S1, S-1 with oxaliplatin, and postoperative chemoradiation with S-1 and oxaliplatin in patients with node-positive gastric cancer after D2 resection: The ARTIST 2 trial. Ann. Oncol. 2021, 32, 368–374. [Google Scholar] [CrossRef] [PubMed]
  27. Iwasaki, Y.; Terashima, M.; Mizusawa, J.; Katayama, H.; Nakamura, K.; Katai, H.; Yoshikawa, T.; Ito, S.; Kaji, M.; Kimura, Y.; et al. Gastrectomy with or without neoadjuvant S-1 plus cisplatin for type 4 or large type 3 gastric cancer (JCOG0501): An open-label, phase 3, randomized controlled trial. Gastric Cancer 2021, 24, 492–502. [Google Scholar] [CrossRef] [PubMed]
  28. Association, J.G.C. Japanese gastric cancer treatment guidelines 2021 (6th edition). Gastric Cancer 2021, 24, 1–25. [Google Scholar]
  29. Kang, Y.-K.; Yook, J.H.; Park, Y.-K.; Lee, J.S.; Kim, Y.-W.; Kim, J.Y.; Ryu, M.-H.; Rha, S.Y.; Chung, I.J.; Kim, I.-H.; et al. PRODIGY: A Phase III Study of Neoadjuvant Docetaxel, Oxaliplatin, and S-1 Plus Surgery and Adjuvant S-1 Versus Surgery and Adjuvant S-1 for Resectable Advanced Gastric Cancer. J. Clin. Oncol. 2021, 39, 2903–2913. [Google Scholar] [CrossRef]
  30. Zhang, X.; Liang, H.; Li, Z.; Xue, Y.; Wang, Y.; Zhou, Z.; Yu, J.; Bu, Z.; Chen, L.; Du, Y.; et al. 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, superiority and non-inferiority, phase 3 randomised controlled trial. Lancet Oncol. 2021, 22, 1081–1092. [Google Scholar] [CrossRef]
  31. Wang, X.; Li, S.; Xie, T.; Lu, Y.; Guo, X.; Lin, C. Early results of the randomized, multicenter, controlled evaluation of S-1 and oxaliplatin as neoadjuvant chemotherapy for Chinese advanced gastric cancer patients (RESONANCE Trial). J. Clin. Oncol. 2020, 38 (Suppl. S4), 280. [Google Scholar] [CrossRef]
  32. Findlay, M.; Cunningham, D.; Norman, A.; Mansi, J.; Nicolson, M.; Hickish, T.; Nicolson, V.; Nash, A.; Sacks, N.; Ford, H.; et al. A phase II study in advanced gastro-esophageal cancer using epirubicin and cisplatin in combination with continuous infusion 5-fluorouracil (ECF). Ann. Oncol. 1994, 5, 609–616. [Google Scholar] [CrossRef] [PubMed]
  33. Webb, A.; Cunningham, D.; Scarffe, J.H.; Harper, P.; Norman, A.; Joffe, J.K.; Hughes, M.; Mansi, J.; Findlay, M.; Hill, A.; et al. Randomized trial comparing epirubicin, cisplatin, and fluorouracil versus fluorouracil, doxorubicin, and methotrexate in advanced esophagogastric cancer. J. Clin. Oncol. 1997, 15, 261–267. [Google Scholar] [CrossRef] [PubMed]
  34. Ross, P.; Nicolson, M.; Cunningham, D.; Valle, J.; Seymour, M.; Harper, P.; Price, T.; Anderson, H.; Iveson, T.; Hickish, T.; et al. Prospective Randomized Trial Comparing Mitomycin, Cisplatin, and Protracted Venous-Infusion Fluorouracil (PVI 5-FU) With Epirubicin, Cisplatin, and PVI 5-FU in Advanced Esophagogastric Cancer. J. Clin. Oncol. 2002, 20, 1996–2004. [Google Scholar] [CrossRef] [PubMed]
  35. Cunningham, D.; Allum, W.H.; Stenning, S.P.; Thompson, J.N.; Van de Velde, C.J.; Nicolson, M.; Scarffe, J.H.; Lofts, F.J.; Falk, S.J.; Iveson, T.J.; et al. Perioperative Chemotherapy versus Surgery Alone for Resectable Gastroesophageal Cancer. New Engl. J. Med. 2006, 355, 11–20. [Google Scholar] [CrossRef] [PubMed]
  36. Ford, H.E.R.; Marshall, A.; Bridgewater, A.J.; Janowitz, T.; Coxon, F.Y.; Wadsley, J.; Mansoor, W.; Fyfe, D.; Madhusudan, S.; Middleton, G.W.; et al. Docetaxel versus active symptom control for refractory oesophagogastric adenocarcinoma (COUGAR-02): An open-label, phase 3 randomised controlled trial. Lancet Oncol. 2014, 15, 78–86. [Google Scholar] [CrossRef] [PubMed]
  37. Al-Batran, S.-E.; Hartmann, J.T.; Hofheinz, R.; Homann, N.; Rethwisch, V.; Probst, S.; Stoehlmacher, J.; Clemens, M.R.; Mahlberg, R.; Fritz, M.; et al. Biweekly fluorouracil, leucovorin, oxaliplatin, and docetaxel (FLOT) for patients with metastatic adenocarcinoma of the stomach or esophagogastric junction: A phase II trial of the Arbeitsgemeinschaft Internistische Onkologie. Ann. Oncol. 2008, 19, 1882–1887. [Google Scholar] [CrossRef] [PubMed]
  38. Al-Batran, S.-E.; Pauligk, C.; Homann, N.; Hartmann, J.T.; Moehler, M.; Probst, S.; Rethwisch, V.; Stoehlmacher-Williams, J.; Prasnikar, N.; Hollerbach, S.; et al. The feasibility of triple-drug chemotherapy combination in older adult patients with oesophagogastric cancer: A randomised trial of the Arbeitsgemeinschaft Internistische Onkologie (FLOT65+). Eur. J. Cancer 2013, 49, 835–842. [Google Scholar] [CrossRef] [PubMed]
  39. Al-Batran, S.E.; Homann, N.; Pauligk, C.; Illerhaus, G.; Martens, U.M.; Stoehlmacher, J.; Schmalenberg, H.; Luley, K.B.; Prasnikar, N.; Egger, M.; et al. Effect of Neoadjuvant Chemotherapy Followed by Surgical Resection on Survival in Patients With Limited Metastatic Gastric or Gastroesophageal Junction Cancer: The AIO-FLOT3 Trial. JAMA Oncol. 2017, 3, 1237–1244. [Google Scholar] [CrossRef]
  40. Homann, N.; Pauligk, C.; Luley, K.; Kraus, T.W.; Bruch, H.-P.; Atmaca, A.; Noack, F.; Altmannsberger, H.-M.; Jäger, E.; Al-Batran, S.-E. Pathological complete remission in patients with oesophagogastric cancer receiving preoperative 5-fluorouracil, oxaliplatin and docetaxel. Int. J. Cancer 2012, 130, 1706–1713. [Google Scholar] [CrossRef]
  41. Schulz, C.; Kullmann, F.; Kunzmann, V.; Fuchs, M.; Geissler, M.; Vehling-Kaiser, U.; Stauder, H.; Wein, A.; Al-Batran, S.-E.; Kubin, T.; 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. Int. J. Cancer 2015, 137, 678–685. [Google Scholar] [CrossRef]
  42. Becker, K.; Mueller, J.D.; Schulmacher, C.; Ott, K.; Fink, U.; Busch, R.; Bottcher, K.; Siewert, J.R.; Hofler, H. Histomorphology and grading of regression in gastric carcinoma treated with neoadjuvant chemotherapy. Cancer 2003, 98, 1521–1530. [Google Scholar] [CrossRef] [PubMed]
  43. Al-Batran, S.-E.; Homann, N.; Pauligk, C.; Goetze, T.O.; Meiler, J.; Kasper, S.; Kopp, H.-G.; Mayer, F.; Haag, G.M.; Luley, K.; et al. 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. Lancet 2019, 393, 1948–1957. [Google Scholar] [CrossRef] [PubMed]
  44. Kim, I.-H. Current status of adjuvant chemotherapy for gastric cancer. World J. Gastrointest. Oncol. 2019, 11, 679–687. [Google Scholar] [CrossRef] [PubMed]
  45. Dikken, J.L.; Jansen, E.P.; Cats, A.; Bakker, B.; Hartgrink, H.H.; Kranenbarg, E.M.-K.; Boot, H.; Putter, H.; Peeters, K.C.; van de Velde, C.J.; et al. Impact of the Extent of Surgery and Postoperative Chemoradiotherapy on Recurrence Patterns in Gastric Cancer. J. Clin. Oncol. 2010, 28, 2430–2436. [Google Scholar] [CrossRef] [PubMed]
  46. Stiekema, J.; Trip, A.K.; Jansen, E.P.M.; Boot, H.; Cats, A.; Ponz, O.B.; Verheij, M.; van Sandick, J.W. The Prognostic Significance of an R1 Resection in Gastric Cancer Patients Treated with Adjuvant Chemoradiotherapy. Ann. Surg. Oncol. 2014, 21, 1107–1114. [Google Scholar] [CrossRef]
  47. Schwarz, R.E.; Smith, D.D. Clinical impact of lymphadenectomy extent in resectable gastric cancer of advanced stage. Ann. Surg. Oncol. 2007, 14, 317–328. [Google Scholar] [CrossRef] [PubMed]
  48. Smalley, S.R.; Benedetti, J.K.; Haller, D.G.; Hundahl, S.A.; Estes, N.C.; Ajani, J.A.; Gunderson, L.L.; Goldman, B.; Martenson, J.A.; Jessup, J.M.; et al. Updated analysis of SWOG-directed intergroup study 0116: A phase III trial of adjuvant radiochemotherapy versus observation after curative gastric cancer resection. J. Clin. Oncol. 2012, 30, 2327–2333. [Google Scholar] [CrossRef] [PubMed]
  49. National Comprehensive Cancer Network (NCCN): NCCN Clinical Practice Guidelines in Oncology: Gastric Cancer Version 1. 2023. Available online: https://www.nccn.org/professionals/physician_gls/pdf/gastric.pdf (accessed on 15 June 2023).
  50. Songun, I.; Putter, H.; Kranenbarg, E.M.-K.; Sasako, M.; van de Velde, C.J. Surgical treatment of gastric cancer: 15-year follow-up results of the randomised nationwide Dutch D1D2 trial. Lancet Oncol. 2010, 11, 439–449. [Google Scholar] [CrossRef]
  51. Lee, J.; Lim, D.H.; Kim, S.; Park, S.H.; Park, J.O.; Park, Y.S.; Lim, H.Y.; Choi, M.G.; Sohn, T.S.; Noh, J.H.; et al. Phase III Trial Comparing Capecitabine Plus Cisplatin Versus Capecitabine Plus Cisplatin With Concurrent Capecitabine Radiotherapy in Completely Resected Gastric Cancer With D2 Lymph Node Dissection: The ARTIST Trial. J. Clin. Oncol. 2012, 30, 268–273. [Google Scholar] [CrossRef]
  52. Shitara, K.; Van Cutsem, E.; Bang, Y.-J.; Fuchs, C.; Wyrwicz, L.; Lee, K.-W.; Kudaba, I.; Garrido, M.; Chung, H.C.; Lee, J.; et al. 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 Oncol. 2020, 6, 1571–1580. [Google Scholar] [CrossRef]
  53. Moehler, M.; Shitara, K.; Garrido, M.; Salman, P.; Shen, L.; Wyrwicz, L.; Yamaguchi, K.; Skoczylas, T.; Bragagnoli, A.C.; Liu, T.; et al. LBA6_PR Nivolumab (nivo) plus chemotherapy (chemo) versus chemo as first-line (1L) treatment for advanced gastric cancer/gastroesophageal junction cancer (GC/GEJC)/esophageal adenocarcinoma (EAC): First results of the CheckMate 649 study. Ann. Oncol. 2020, 31, S1191. [Google Scholar] [CrossRef]
  54. Shitara, K.; Bang, Y.-J.; Iwasa, S.; Sugimoto, N.; Ryu, M.-H.; Sakai, D.; Chung, H.-C.; Kawakami, H.; Yabusaki, H.; Lee, J.; et al. Trastuzumab Deruxtecan in Previously Treated HER2-Positive Gastric Cancer. N. Engl. J. Med. 2020, 382, 2419–2430. [Google Scholar] [CrossRef] [PubMed]
  55. Boku, N.; Ryu, M.; Oh, D.-Y.; Oh, S.; Chung, H.; Lee, K.-W.; Omori, T.; Shitara, K.; Sakuramoto, S.; Chung, I.; et al. LBA7_PR Nivolumab plus chemotherapy versus chemotherapy alone in patients with previously untreated advanced or recurrent gastric/gastroesophageal junction (G/GEJ) cancer: ATTRACTION-4 (ONO-4538-37) study. Ann. Oncol. 2020, 31, S1192. [Google Scholar] [CrossRef]
  56. Al-Batran, S.-E.; Hofheinz, R.; Schmalenberg, H.; Strumberg, D.; Goekkurt, E.; Angermeier, S.; Zander, T.; Potenberg, J.; Kopp, H.-G.; Pink, D.; et al. 1424MO Perioperative FLOT plus ramucirumab versus FLOT alone for resectable esophagogastric adenocarcinoma—Updated results and subgroup analyses of the randomized phase II/III trial RAMSES/FLOT7 of the German AIO and Italian GOIM. Ann. Oncol. 2020, 31, S901. [Google Scholar] [CrossRef]
  57. Al-Batran, S.-E.; Haag, G.; Ettrich, T.; Borchert, K.; Kretzschmar, A.; Teschendorf, C.; Siegler, G.; Ebert, M.; Goekkurt, E.; Welslau, M.; et al. 1421MO Final results and subgroup analysis of the PETRARCA randomized phase II AIO trial: Perioperative trastuzumab and pertuzumab in combination with FLOT versus FLOT alone for HER2 positive resectable esophagogastric adenocarcinoma. Ann. Oncol. 2020, 31, S899. [Google Scholar] [CrossRef]
  58. Wagner, A.D.; Grabsch, H.I.; Mauer, M.; Marreaud, S.; Caballero, C.; Thuss-Patience, P.; Mueller, L.; Elme, A.; Moehler, M.H.; Martens, U.; et al. EORTC-1203-GITCG—The “INNOVATION”-trial: Effect of chemotherapy alone versus chemotherapy plus trastuzumab, versus chemotherapy plus trastuzumab plus pertuzumab, in the perioperative treatment of HER2 positive, gastric and gastroesophageal junction adenocarcinoma on pathologic response rate: A randomized phase II-intergroup trial of the EORTC-Gastrointestinal Tract Cancer Group, Korean Cancer Study Group and Dutch Upper GI-Cancer group. BMC Cancer 2019, 19, 494. [Google Scholar]
  59. Bang, Y.-J.; Van Cutsem, E.; Fuchs, C.S.; Ohtsu, A.; Tabernero, J.; Ilson, D.H.; Hyung, W.J.; Strong, E.V.; Goetze, T.O.; Yoshikawa, T.; et al. KEYNOTE-585: Phase III study of perioperative chemotherapy with or without pembrolizumab for gastric cancer. Futur. Oncol. 2019, 15, 943–952. [Google Scholar] [CrossRef] [PubMed]
  60. Janjigian, Y.Y.; Van Cutsem, E.; Muro, K.; Wainberg, Z.; Al-Batran, S.-E.; Hyung, W.J.; Molena, D.; Marcovitz, M.; Ruscica, D.; Robbins, S.H.; et al. MATTERHORN: Phase III study of durvalumab plus FLOT chemotherapy in resectable gastric/gastroesophageal junction cancer. Futur. Oncol. 2022, 18, 2465–2473. [Google Scholar] [CrossRef]
  61. Homann, N.; Lorenzen, S.; Schenk, M.; Thuss-Patience, P.C.; Goekkurt, E.; Hofheinz, R.D.; Kretzschmar, A.; Bolling, C.; Angermeier, S.; Wicki, A.; et al. Interim safety analysis of the DANTE trial: Perioperative atezolizumab in combination with FLOT versus FLOT alone in patients with resectable esophagogastric adenocarcinoma—A randomized, open-label phase II trial of the German Gastric Group at the AIO and SAKK. J. Clin. Oncol. 2020, 38 (Suppl. S15), 4549. [Google Scholar]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Takayama, T.; Tsuji, Y. Updated Adjuvant Chemotherapy for Gastric Cancer. J. Clin. Med. 2023, 12, 6727. https://doi.org/10.3390/jcm12216727

AMA Style

Takayama T, Tsuji Y. Updated Adjuvant Chemotherapy for Gastric Cancer. Journal of Clinical Medicine. 2023; 12(21):6727. https://doi.org/10.3390/jcm12216727

Chicago/Turabian Style

Takayama, Toshizo, and Yasushi Tsuji. 2023. "Updated Adjuvant Chemotherapy for Gastric Cancer" Journal of Clinical Medicine 12, no. 21: 6727. https://doi.org/10.3390/jcm12216727

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop