Treatment Outcomes of Stage II or III Gastric Cancer Treated with Adjuvant Chemotherapy with TS-1 or XELOX after Radical Surgery

   CME

TCY So, KC Lee, ECY Wong

ORIGINAL ARTICLE    CME
 
Treatment Outcomes of Stage II or III Gastric Cancer Treated with Adjuvant Chemotherapy with TS-1 or XELOX after Radical Surgery
 
TCY So, KC Lee, ECY Wong
Department of Clinical Oncology, Pamela Youde Eastern Hospital, Hong Kong SAR, China
 
Correspondence: Dr TCY So, Department of Clinical Oncology, Pamela Youde Eastern Hospital, Hong Kong SAR, China. Email: scy027@ha.org.hk
 
Submitted: 6 May 2022; Accepted: 2 Dec 2022.
 
Contributors: TCYS and ECYW designed the study. TCYS acquired, analysed the data and drafted the manuscript. KCL and ECYW critically revised the manuscript for important intellectual content. All authors had full access to the data, contributed to the study, approved the final version for publication, and take responsibility for its accuracy and integrity.
 
Conflicts of Interest: All authors have disclosed no conflicts of interest.
 
Funding/Support: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.
 
Data Availability: All data generated or analysed during the present research are available from the corresponding author on reasonable request.
 
Ethics Approval: This research has been approved by the Hong Kong East Cluster Research Ethics Committee of Hospital Authority, Hong Kong (Ref. No.: HKECREC-2022-021) and was conducted in compliance with the Declaration of Helsinki. The requirement for patient consent was waived by the Committee due to the retrospective nature of the study.
 
 
 
 
 
Abstract
 
Introduction
 
Capecitabine plus oxaliplatin (XELOX) and tegafur/gimeracil/oteracil (TS-1, also known as ‘S-1’) are two commonly used adjuvant chemotherapy regimens for gastric cancer in Hong Kong. This study aimed to review the outcomes of patients receiving these two regimens, to investigate important clinical factors that may impact on the risk of disease recurrence, and to explore the roles of neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio (PLR) in prognostication after radical surgery.
 
Methods
 
Patients who received adjuvant treatment (either XELOX or TS-1) for gastric cancer following radical surgical resection from January 2016 to December 2020 at our hospital were included. Patient demographics, overall survival (OS), and disease-free survival (DFS) were analysed.
 
Results
 
A total of 65 patients were included (XELOX: n = 40; TS-1: n = 25). XELOX appeared to have more favourable OS and DFS, although the result was confounded by older and frailer patients in the TS-1 group. An elevated PLR was associated with inferior OS after surgery (p = 0.036). Cox regression analysis showed that Eastern Cooperative Oncology Group (ECOG) performance status score of 2 and nodal stage of N2 to N3 were two independent factors associated with inferior OS. ECOG performance status score of 2, nodal stage of N2 to N3, and chemotherapy dose intensity <70% were significantly associated with a higher risk of relapse.
 
Conclusion
 
Poorer ECOG performance status and more advanced nodal stage are independent factors associated with inferior OS and DFS, and lower chemotherapy dose intensity (<70%) resulted in a higher risk of disease relapse. NLR and PLR is a simple clinical marker that may be further explored as a prognostic marker for gastric cancer after radical surgery.
 
 
Key Words: Blood platelets; Lymphocytes; Neutrophils; Prognosis; Stomach neoplasms
 
 
中文摘要
 
根治性手術後使用TS-1或XELOX輔助化療治療II期或III期胃癌的治療結果
 
蘇駿寅、李建忠、王晉彥
 
簡介
卡培他濱聯合奧沙利鉑(XELOX)及替加氟/吉美嘧啶/氧嗪酸(TS-1,又稱S-1)是香港兩種常用於胃癌的輔助化療方案。本研究旨在回顧接受這兩種方案的病人的結果,調查可能影響疾病復發風險的重要臨床因素,以及研究在根治性手術後嗜中性白血球與淋巴細胞比例(NLR)及血小板與淋巴細胞比例(PLR)在預測方面的角色。
 
方法
本研究包括於2016年1月至2020年12月期間在本院進行根治性手術切除後接受輔助治療(XELOX或TS-1)的胃癌病人,並分析了有關患者的人口特徵、整體存活及無疾病存活。
 
結果
本研究共包括65名患者(XELOX:n = 40;TS-1:n = 25)。雖然XELOX的整體存活及無疾病存活似乎較好,但這些結果受TS-1組別中年紀較大及較虛弱的患者影響。血小板與淋巴細胞比例上升與較差的術後整體存活相關(p = 0.036)。Cox迴歸分析顯示美國東岸癌症臨床研究合作組織(ECOG)身體功能狀態評分為2分及癌症分期為N2至N3,是與較差的整體存活相關的兩個獨立因素。ECOG身體功能狀態評分為2分、癌症分期為N2至N3及化療劑量強度<70%與較高復發風險顯著相關。
 
結論
較差的ECOG身體功能狀態及較晚期的癌症分期是與較差的整體存活及無疾病存活相關的獨立因素,而較低的化療劑量強度<70%造成較高的疾病復發風險。NLR和PLR是簡單的臨床標記,可成為日後的研究方向,以此比例作為根治性手術後胃癌的預後標記。
 
 
 
INTRODUCTION
 
Gastric cancer was the sixth commonest cancer in Hong Kong, accounting for 3.7% of all new cancer cases in 2019.[1] Although the incidence has been gradually declining, compatible with global trends due to efficacious Helicobacter pylori eradication therapy,[2] gastric cancer remains more prevalent in Asian countries than in the West.
 
Clear surgical resection with D2 lymphadenectomy and chemotherapy is considered the standard of care for resectable locoregionally advanced gastric cancer nowadays,[3] and this has been advocated in several international guidelines.[4] [5] Adjuvant chemoradiotherapy (45 Gy over 25 fractions concurrent with 5-fluorouracil and leucovorin) had once been widely adopted, but was later criticised for the inclusion of a high proportion of patients with D1 lymphadenectomy in the study recommending it.[6]
 
The choice of chemotherapy regimen significantly differs among different parts of the world. In European countries, perioperative chemotherapy, such as the combination of epirubicin, cisplatin, and 5-fluorouracil[7] or 5-fluorouracil, leucovorin, oxaliplatin, and docetaxel,[8] is frequently used, whereas in Hong Kong, clinicians tend to use adjuvant chemotherapy as in most Asian countries. The two most commonly used regimens of adjuvant chemotherapy after radical surgery are capecitabine plus oxaliplatin (XELOX) and tegafur/gimeracil/oteracil (TS-1, also known as ‘S-1’). They both demonstrated significant benefits when compared with surgery alone in randomised clinical trials[9] [10] conducted in Asian countries. Despite the two regimens having been widely used, there are no prospective randomised clinical trials directly comparing their efficacy.
 
Regarding the prognostic stratification of patients with resected gastric cancer, several clinical and pathological parameters have long been adopted to predict the recurrence of gastric cancer including age, comorbidities, tumour size, differentiation status, and presence of lymphovascular or perineural invasion.[11] [12] [13] [14] In recent years, the clinical utility of the peripheral neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) as systemic inflammatory markers has been addressed. In relation to cancer prognosis, several meta-analyses showed that elevated NLR and PLR correlated with tumour progression and poor survival in a number of gastrointestinal cancers.[15] [16] However, what the same observation connotes in the adjuvant setting remains uncertain.
 
This retrospective study was conducted with three aims: to compare the efficacy of adjuvant XELOX with TS-1 chemotherapy for patients with stage II or III gastric cancer who received radical surgery in our locality; to investigate important clinical factors that may impact on the risk of disease recurrence; and to explore the prognostic value of NLR and PLR as potentially useful and easily available clinical parameters.
 
METHODS
 
Patients and Data Collection
 
Patients who received adjuvant treatment (XELOX: n = 40; TS-1: n = 25) for gastric cancer following radical surgical resection from January 2016 to December 2020 at the Department of Clinical Oncology, Pamela Youde Eastern Hospital, Hong Kong were included in the study. Patients with metastatic disease at presentation (including small-volume peritoneal metastasis) or double primary cancers were excluded. Patients who received adjuvant radiotherapy were also excluded. Relevant clinical and pathological parameters were captured from clinical notes and the Clinical Management System of Hospital Authority.
 
Treatment
 
XELOX consists of oral capecitabine (1,000 mg/m2 twice daily on days 1-14 of each cycle) plus intravenous oxaliplatin (130 mg/m2 on day 1 of each cycle) up to 8 cycles. TS-1 is oral chemotherapy (daily dose according to body surface area [BSA]: patients with BSA <1.25 m2 received 80 mg daily, those BSA ranging from ≤1.25 m2 to 1.50 m2 received 100 mg daily, and those with BSA ≥1.50 m2 received 120 mg daily) given for 4 weeks followed by 2 weeks of rest for a total of 9 cycles.
 
In practice, patients of an advanced age, borderline Eastern Cooperative Oncology Group (ECOG) performance status and pre-existing neuropathy would be more likely to be given TS-1, as it is a non–self-financed item under the institution.
 
Doses and schedule modifications were conducted based on patients’ ECOG performance status, organ functions, and toxicities by clinicians’ decisions. Dose reduction of chemotherapy was conducted in a stepwise manner (75%-85% of the initial dose for 1st dose reduction, then 60%-70% for the 2nd dose reduction). The relative total dose intensity (RTDI) is the ratio of the delivered actual dose intensity (ATDI) to the standard planned dose intensity (PTDI) for a chemotherapy regimen, which is calculated as follows:
 
 
Follow-up and Assessment
 
Patients were seen by doctors prior to each cycle of chemotherapy, when tolerance of chemotherapy and results of blood tests would be recorded in the Clinical Management System. Patients who had completed the adjuvant chemotherapy would be followed up at an interval of 3 to 6 months. Computed tomography was performed if there was clinical suspicion of disease relapse. Disease relapse was defined as any radiological and/or histological confirmation of recurrence. Elevated tumour markers alone were not considered as relapse without proof of recurrent disease.
 
Statistical Analysis
 
Statistical analysis was performed using SPSS (Windows version 22; IBM Corp, Armonk [NY], United States). Clinical and pathological data were retrospectively reviewed and analysed by descriptive statistics. Pearson’s Chi squared test was used for testing any significant correlations and differences between groups.
 
Treatment outcomes, including disease-free survival (DFS, the time from surgery to disease relapse) and overall survival (OS, the time from diagnosis of disease to death from any cause) were analysed by the Kaplan-Meier method and the difference between groups were tested with the log-rank test. Different clinical parameters were tested for their impact on DFS and OS by Cox regression analysis.
 
In order to have an accurate assessment of the baseline NLR and PLR of our patients, the complete blood counts right before the administration of first cycle of chemotherapy were recorded. This is to minimise the effect due to postoperative inflammation and chemotherapy on peripheral blood counts.
 
Using all-cause mortality as an endpoint for NLR and PLR, the optimal cut-off values were determined by receiver operating curve analysis as shown in Figure 1. The area under the curve of NLR and PLR was 0.653 and 0.575, respectively. The optimal cut-off values determined by the Youden’s index for NLR and PLR were 1.9 and 169, respectively.
 
Figure 1. Receiver operating curve (ROC) analysis for optimal cut-off values of neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR).
 
RESULTS
 
Patient Characteristics
 
Sixty-five patients were identified and included in the analysis. Forty patients received XELOX and 25 received TS-1. The median follow-up time for this study was 33.7 months (range, 6.5-78.9).
 
Patient baseline characteristics are summarised in Table 1. The median age of the entire cohort was 66.0 years. The mean and median age in the XELOX group were 57.7 and 59.0 years respectively, compared to 69.4 and 71.0 years in the TS-1 group. Patients who received TS-1 were significantly older, with 72.0% of them ≥66 years compared to 37.5% in XELOX group (p = 0.007). All included patients had an ECOG performance status score of ≤2. There were significantly more patients with ECOG performance status score ≤1 in the XELOX group (97.5%) than in TS-1 group (60.0%) [p < 0.001].
 
Table 1. Patient baseline characteristics of XELOX and TS-1 groups.
 
Overall and Disease-Free Survival
 
The median OS was 38.9 months for the XELOX group and 22.9 months for TS-1 group. The observed OS and DFS in the XELOX group were significantly longer than those in the TS-1 group (Figure 2) [p = 0.037 and 0.012, respectively]. However, it should be interpreted carefully as the baseline patients’ characteristics suggested a bias towards prescribing TS-1 in the older age-group and less fit patients. These factors likely confound the survival analysis.
 
Figure 2. Kaplan-Meier analysis of (a) overall survival (p = 0.037, log-rank test) and (b) disease-free survival (p = 0.012, log-rank test) in all patients with respect to treatment with TS-1 (tegafur/gimeracil/oteracil) or XELOX (capecitabine plus oxaliplatin).
 
Neutrophil-to-Lymphocyte Ratio and Platelet-to-Lymphocyte Ratio
 
Overall survival analysis showed that patients with high NLR (≥1.9) before adjuvant chemotherapy had shorter OS than those with low NLR (<1.9), although the difference was marginally significant (p = 0.051; Figure 3). The same analysis also demonstrated that patients with high PLR (≥169) before adjuvant chemotherapy had significantly shorter OS than those with low PLR (<169) [p = 0.036; Figure 4].
 
Figure 3. High neutrophil-to-lymphocyte ratio (≥1.9) versus low neutrophil-to lymphocyte-ratio (<1.9) on overall survival (p = 0.051).
 
Figure 4. High platelet-to-lymphocyte ratio (≥169) versus low platelet-to-lymphocyte ratio (<169) on overall survival (p = 0.036).
 
In relation to clinical characteristics, patients with elevated NLR correlated with female gender (borderline p value of 0.049) and elevated PLR was associated with more advanced disease (p = 0.012) [Tables 2 and 3].
 
Table 2. Clinical characteristics of patients with high and low neutrophil-to-lymphocyte ratio.
 
Table 3. Clinical characteristics of patients with high and low platelet-to-lymphocyte ratio.
 
Clinical and Pathological Parameters on Overall Survival and Disease-Free Survival
 
As shown in Table 4, univariate Cox regression analysis showed that ECOG performance status score of 2, nodal stage of N2 to N3, and elevated PLR (≥169) were adverse prognostic factors for OS, while ECOG performance status score of 2, nodal stage of N2 to N3, and RTDI of chemotherapy <70% were adverse factors associated with disease relapse.
 
Table 4. Cox regression analysis on multiple clinical and pathological parameters on overall survival.
 
Multivariable Cox regression analysis demonstrated that ECOG performance status score of 2 and nodal stage of N2 to N3 were the two independent adverse prognostic factors for OS (Table 4). For DFS, ECOG performance status score of 2, nodal stage of N2 to N3, and RTDI of chemotherapy <70% were the three independent factors associated with disease relapse (Table 5).
 
Table 5. Cox regression analysis on multiple clinical and pathological parameters on disease-free survival.
 
DISCUSSION
 
Our study revealed that the XELOX group had more favourable oncological outcomes (both DFS and OS) than the TS-1 group. However, it should be noted that patients included in the TS-1 group in out centre were older (p = 0.007) and of worse ECOG performance status (p < 0.001). This is largely due to the fact that the institutional guideline recommends TS-1 as the treatment of choice for older patients with anticipated poor tolerance to XELOX and that under such circumstances, only the drug costs of TS-1 would be covered by the institution. There has not been any randomised controlled trial comparing the efficacy of the two regimens. Retrospective studies[17] [18] [19] [20] did not demonstrate statistically significant differences in DFS between adjuvant TS-1 and XELOX (Table 6). In the subgroup analysis, one study[17] demonstrated the use of XELOX in stage II disease was associated with better OS while another study[18] suggested the same but in stage IIIB/C disease only. Apart from XELOX, combination chemotherapy with more than three agents has shown superior treatment outcomes in recent years. Combination of TS-1 with oxaliplatin[21] or docetaxel[22] is considered a preferred option for high-risk patients and is increasingly recognised as a new standard of care.
 
Table 6. Summary of selected retrospective studies comparing XELOX and TS-1 regimens.
 
In our cohort, elevated PLR is associated with inferior OS after curative surgery and there was a similar trend for NLR despite not reaching statistical significance (p = 0.051). NLR and PLR are important parameters indicating systemic inflammation. It is observed that a chronic inflammatory state confers unfavourable oncological outcomes.[23] Several meta-analyses revealed that elevated NLR and PLR were associated with tumour progression and poor survival in gastrointestinal cancers.[15] [16] Microscopically, various inflammatory cytokines and growth factors in the tumour microenvironment are known to dampen hosts’ anti-tumour immune response. In tumour models, inflammatory cytokines such as interleukin 6 (IL-6), IL-8 and IL-11 are associated with chemotherapy resistance in gastric cancer through mechanisms such as inhibition of apoptosis pathways, increasing efflux of chemotherapeutic agents, and evasion of DNA damage.[24] [25] [26] We therefore postulated that in an adjuvant setting, the persistent inflammatory state after curative surgery possibly led to tumour evasion from immunosurveillance and enhanced chemoresistance of micrometastases.[27] NLR and PLR are two readily accessible clinical parameters and may serve as simple prognostic tools in addition to performance status, stage, and age.
 
Our study revealed that the RTDI is an independent prognostic factor for disease recurrence. Inadequate chemotherapy dose intensity is either attributed to excessive dose reduction or failure to complete scheduled cycles within the planned time interval. It is noteworthy that severe adverse events of chemotherapy (≥ Grade 3) have been shown to be quite uncommon (≤6%) with TS-1 in a large-scale clinical trial,[28] although these patients were generally frailer and older. For elderly patients who may be more vulnerable to chemotherapy toxicity, proper geriatric assessments (such as comorbidity and frailty indices) are needed, as biological age is not a reliable indicator for chemotherapy dose adjustment, and an adaptive dose optimisation approach is recommended based on patients’ tolerance of each cycle.
 
This study has several limitations. First, it is only a single-centre retrospective study in which the small sample size limits its statistical power. Second, there is imbalance between the baseline characteristics of the two groups of patients. Similar to the Korean studies,[17] [18] [19] [20] patients in the TS-1 group were generally older and had a worse ECOG performance status. There is a tendency for clinicians to prescribe a more conservative chemotherapy dosage in this group of patients, which may explain the lower dose intensity of TS-1 than XELOX. Propensity score matching should be performed in a larger cohort to reduce the bias due to these confounding variables. Third, a much large sample size is needed to further evaluate the prognostic power of NLR and PLR on OS and DFS in the adjuvant setting. In our cohort, high PLR appeared to correlate with patients with more advanced disease (stage III), which is an important confounding factor.
 
CONCLUSION
 
In conclusion, we compared the OS and DFS between adjuvant XELOX and TS-1 in our local gastric cancer patients. Clinical outcomes were statistically better with XELOX- than TS-1–treated patients. However, the results should be viewed with caution because of the limited sample size and obvious imbalance in baseline characteristics. ECOG performance status score of 2 and advanced nodal stage of N2 to N3 are independent adverse prognostic factors associated with poor OS and a higher rate of disease recurrence. NLR and PLR are readily available markers that may be further explored as prognostic markers for gastric cancer after radical surgery. We also speculated that the RTDI of chemotherapy of <70% might affect the risk of disease relapse.
 
REFERENCES
 
1. Centre for Health Protection, Department of Health, Hong Kong SAR Government. Stomach cancer. Available from: https://www.chp.gov.hk/en/healthtopics/content/25/55.html. Accessed 2 May 2022.
 
2. Rawla P, Barsouk A. Epidemiology of gastric cancer: global trends, risk factors and prevention. Prz Gastroenterol. 2019;14:26-38. Crossref
 
3. Randle RW, Swords DS, Levine EA, Fino NF, Squires MH, Poultsides G, et al. Optimal extent of lymphadenectomy for gastric adenocarcinoma: a 7-institution study of the US gastric cancer collaborative. J Surg Oncol. 2016;113:750-5. Crossref
 
4. Smyth EC, Verheij M, Allum W, Cunningham D, Cervantes A, Arnold D, et al. Gastric cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2016;27(suppl 5):v38-49. Crossref
 
5. Hakkenbrak, NA, Jansma EP, van der Wielen N, van der Peet DL, Straatman J. Laparoscopic versus open distal gastrectomy for gastric cancer: a systematic review and meta-analysis. Surgery. 2022;171:1552-61. Crossref
 
6. Macdonald JS, Smalley SR, Benedetti J, Hundahl SA, Estes NC, Stemmermann GN, et al. Chemoradiotherapy after surgery compared with surgery alone for adenocarcinoma of the stomach or gastroesophageal junction. N Engl J Med. 2001;345:725-30. Crossref
 
7. Cunningham D, Allum WH, Stenning SP, Weeden S. Perioperative chemotherapy in operable gastric and lower oesophageal cancer: final results of a randomised, controlled trial (the MAGIC trial, ISRCTN 93793971). J Clin Oncol. 2005;23(16 suppl):4001. Crossref
 
8. Al-Batran SE, Homann N, Schmalenberg H, Kopp HG, Haag GM, Luley KB, et al. Perioperative chemotherapy with docetaxel, oxaliplatin, and fluorouracil/leucovorin (FLOT) versus epirubicin, cisplatin, and fluorouracil or capecitabine (ECF/ECX) for resectable gastric or gastroesophageal junction (GEJ) adenocarcinoma (FLOT4-AIO): a multicenter, randomized phase 3 trial. J Clin Oncol. 2017;35(15 suppl):4004. Crossref
 
9. Bang YJ, Kim YW, Yang HK, Chung HC, Park YK, Lee KH, et al. Adjuvant capecitabine and oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): a phase 3 open-label, randomised controlled trial. Lancet. 2012;379:315-21. Crossref
 
10. Sasako M, Sakuramoto S, Katai H, Kinoshita T, Furukawa H, Yamaguchi T, et al. 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;29:4387-93. Crossref
 
11. Liang YX, Deng JY, Guo HH, Ding XW, Wang XN, Wang BG, et al. Characteristics and prognosis of gastric cancer in patients aged ≥70 years. World J Gastroenterol. 2013;19:6568-78. Crossref
 
12. Stiekema J, Cats A, Kuijpers A, van Coevorden F, Boot H, Jansen EP, et al. Surgical treatment results of intestinal and diffuse type gastric cancer. Implications for a differentiated therapeutic approach? Eur J Surg Oncol. 2013;39:686-93. Crossref
 
13. Feng F, Liu J, Wang F, Zheng G, Wang Q, Liu S, et al. Prognostic value of differentiation status in gastric cancer. BMC Cancer. 2018;18:865. Crossref
 
14. Asplund J, Gottlieb-Vedi E, Leijonmarck W, Mattsson F, Lagergren J. Prognosis after surgery for gastric adenocarcinoma in the Swedish Gastric Cancer Surgery Study (SWEGASS). Acta Oncol. 2021;60:513-20. Crossref
 
15. Yodying H, Matsuda A, Miyashita M, Matsumoto S, Sakurazawa N, Yamada M, et al. Prognostic significance of neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio in oncologic outcomes of esophageal cancer: a systematic review and meta-analysis. Ann Surg Oncol. 2016;23:646-54 Crossref
 
16. Zhang J, Zhang HY, Li J, Shao XY, Zhang CX. The elevated NLR, PLR and PLT may predict the prognosis of patients with colorectal cancer: a systematic review and meta-analysis. Oncotarget. 2017;8:68837-46. Crossref
 
17. Oh SE, An JY, Choi MG, Lee JH, Sohn TS, Bae JM. Comparison of long-term efficacy in S-1 and capecitabine with oxaliplatin as adjuvant chemotherapy for patients with gastric cancer after curative surgery: a retrospective, single-center observational study. Technol Cancer Res Treat. 2021;20:15330338211039679. Crossref
 
18. Kim IH, Park SS, Lee CM, Kim MC, Kwon IK, Min JS, et al. Efficacy of adjuvant S-1 versus XELOX chemotherapy for patients with gastric cancer after D2 lymph node dissection: a retrospective, multi-center observational study. Ann Surg Oncol. 2018;25:1176-83. Crossref
 
19. Cho JH, Lim JY, Cho JY. Comparison of capecitabine and oxaliplatin with S-1 as adjuvant chemotherapy in stage III gastric cancer after D2 gastrectomy. PLoS One. 2017;12:e0186362. Crossref
 
20. Lee CM, Yoo MW, Son YG, Oh SJ, Kim JH, Kim HI, et al. Long-term efficacy of S-1 monotherapy or capecitabine plus oxaliplatin as adjuvant chemotherapy for patients with stage II or III gastric cancer after curative gastrectomy: a propensity score-matched multicenter cohort study. J Gastric Cancer. 2020;20:152-64. Crossref
 
21. Park SH, Lim DH, Sohn TS, Lee J, Zang DY, Kim ST, 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;32:368-74. Crossref
 
22. Kodera Y, Yoshida K, Kochi M, Ichikawa W, Kakeji Y, Sano T, et al. A randomized phase III study comparing S-1 plus docetaxel with S-1 alone as a postoperative adjuvant chemotherapy for curatively resected stage III gastric cancer (JACCRO GC-07 trial). J Clin Oncol. 2018;36(15 suppl):4007. Crossref
 
23. Baniyash M, Sade-Feldman M, Kanterman J. Chronic inflammation and cancer: suppressing the suppressors. Cancer Immunol Immunother. 2014;63:11-20. Crossref
 
24. Ham IH, Oh HJ, Jin H, Bae A, Jeon SM, Choi KS, et al. Targeting interleukin-6 as a strategy to overcome stroma-induced resistance to chemotherapy in gastric cancer. Mol Cancer. 2019;18:68. Crossref
 
25. Kuai WX, Wang Q, Yang XZ, Zhao Y, Yu R, Tang XJ. Interleukin-8 associates with adhesion, migration, invasion and chemosensitivity of human gastric cancer cells. World J Gastroenterol. 2012;18:979-85. Crossref
 
26. Ma J, Song X, Xu X, Mou Y. Cancer-associated fibroblasts promote the chemo-resistance in gastric cancer through secreting IL-11 targeting JAK/STAT3/Bcl2 pathway. Cancer Res Treat. 2019;51:194-210. Crossref
 
27. Olive KP. Fanning the flames of cancer chemoresistance: inflammation and anticancer therapy. J Oncol Pract. 2017;13:181-3. Crossref
 
28. Sakuramoto S, Sasako M, Yamaguchi T, Kinoshita T, Fujii M, Nashimoto A, et al. Adjuvant chemotherapy for gastric cancer with S-1, an oral fluoropyrimidine. N Engl J Med. 2007;357:1810-20. Crossref