Abstract
Several possibilities in treating advanced gastric cancer exist. Radical surgery associated with chemotherapy represents the cornerstone. Which one is more effective among neoadjuvant, adjuvant or perioperative chemotherapy is still a matter of debate. Several innovative results showed the necessity to keep increasingly into consideration the intraperitoneal administration of chemotherapies. Moreover, classical drugs and their ways of administration should be combined with the new ones to improve results. Lastly the prevention of recurrence should be considered: one possibility is to administer intraperitoneal chemotherapy earlier in the therapeutic algorithm. (www.actabiomedica.it)
Keywords: advanced gastric cancer, chemotherapy, hipec, intraperitoneal, surgery, carcinosis, metastasis
Introduction
Several possibilities exist in treating advanced gastric cancer (AGC). Radical surgery associated with chemotherapy (CT) represents the cornerstone. Several innovative results showed the necessity to keep increasingly into consideration the intraperitoneal administration of chemotherapies (IPC). Moreover, and their ways of administration should be combined with the new ones to improve results. Lastly the prevention of recurrence should be considered: one possibility is to administer intraperitoneal chemotherapy earlier in the therapeutic algorithm.
The CT can be administered through different ways and at different time points. The present review aims to give a comprehensive overview of the different possibilities in treating AC.
Neo-adjuvant chemotherapy
The primary aim of neo-adjuvant chemotherapy (NACT) is to reduce the tumoral extension to potentially increase the effects of a radical surgery and to reduce the biological potential of tumor cells with particular attention to subclinical micrometastases. One possible disadvantage of NACT could be to delay the surgical intervention.
The EORTC 40954 trial (1) showed an increased rate of R0 resections in NACT group, more frequent postoperative morbidity and positive hazard ratio in favor to NACT with regards to survival although not significantly. Few randomized studies were closed prematurely with no favorable results. The FAMTX trial (2, 3) gave no survival differences related to NACT. However, several evidences exist about the value of this kind of CT. A recent meta-analysis including 15 randomized controlled trials (RCTs) and involving 2001 patients showed that the NACT does not give any adverse effect during the perioperative period. In fact, it does not increase the risk of complications nor the post-operative mortality rate. Furthermore, the effect on early gastric cancer (EGC) and AGC was positive in term both of survival and recurrence rate (4).
Perioperative chemotherapy
Perioperative chemotherapy consists in combining CT before surgery and post-operative CT with interval surgery. The concept at the base of this combined approach is to obtain the advantages of neoadjuvant schemes in reducing tumor size and facilitating radical surgery associated to the advantages offered by post-operative drug administration. In Europe, this approach is diffused, and several trials have been published.
The MAGIC trial enrolled gastric or distal esophagus adenocarcinoma (5). Preoperative CT improved R0 resection rate; almost half of the patients who received preoperative treatment completed the postoperative CT. Perioperative CT reduced the risk of relapse and improved median overall survival.
The ACCORD07 RCT enrolled patients with gastro and gastro-esophageal junction cancer (6). Perioperative CT resulted in higher rates of R0 resection, in reduction of the risk of relapse and of the risk of death.
A Cochrane single patient data meta-analysis on the perioperative CT in resectable gastric adenocarcinoma (7) included 14 RCTs. The cumulative analysis showed an increase in overall survival (OS), R0 resection and longer disease-free survival (DFS) with no differences in term of mortality and morbidity. Advantages of the perioperative scheme were more pronounced in gastro-esophageal junction cancers. When radiotherapy was added, a better OS was obtained. The best effect was found in younger patients, whereas no survival benefit was demonstrated for elderly patients.
Another British study (8) demonstrated a considerable gain in DFS in neoadjuvant and adjuvant treatment in comparison with those who didn’t receive postoperative CT. OS was not significantly different.
A recent meta-analysis of RCT, involving 1240 patients comparing prognosis and safety between perioperative CT and adjuvant chemotherapy (ACT), showed an improved survival for patients treated with perioperative CT. In addition, combination CT resulted in better survival compared to monotherapy in the NACT regimens (9).
Adjuvant chemotherapy
ACT is the most applied scheme throughout the world. Many colleagues from surgical and oncological department prefer to face cancer primarily with the surgical intervention, as surgery is universally considered the main curative option in gastric cancer.
The single patient data meta-analysis by the GASTRIC group (10) analyzed 17 RCTs (3838 patients). Results showed as ACT improved 5-years survival with similar DFS. No differences were found regarding the several fluoropyrimidine based drug regimens applied (i.e. mono-, poly-chemotherapy). Further studies, the ACTSGC study (11) and the CLASSIC study (12), (13), confirmed the results.
A recent RCT did not find a significant survival benefit to be associated with ACT with fluoropyrimidines in patients with stage IB-IIIA gastric cancer. However, patients with stage II disease and those receiving uracil-tegafur treatment in the adjuvant group showed significantly better prognosis than those in the surgery-alone group (14).
S-1 is an orally active combination of tegafur, a prodrug that is converted by cells to fluorouracil, gimeracil, which inhibits dihydropyrimidine dehydrogenase, and oteracil, which inhibits the phosphorylation of fluorouracil in the gastrointestinal tract, thereby reducing the toxic gastrointestinal effects of fluorouracil (15). In Japan ACT using S-1 has become a standard treatment in patients treated by curative gastrectomy for stage II or stage III gastric cancer on the basis of results from a randomized phase III study comparing surgery plus adjuvant S-1 with surgery alone (ACTS-GC trial) (16, 17).
New agents
Tumor biology and the cellular and molecular mechanisms of malignant proliferation have been studied deeply, leading to the comprehension of part of their pathways. This permitted to develop targeted therapies against specific mechanisms. Target therapies permitted to decrease toxicity of traditional chemotherapy agents and improve survival. In gastric cancer HER-2/neu (ERBB2) has demonstrated to be the principal molecular target where monoclonal antibodies have showed their efficacy. HER2 is over-expressed in 10-40% of gastric cancer. Data from a few meta-analyses defined the prognostic role of HER2 over-expression in gastric cancer. However contrasting results have been published (18-22) depending from the diagnostic technique. Trastuzumab (Herceptin®, Genentech) demonstrated its efficacy against HER2. The ToGA trial (23) reported a reduced relative risk of death by the addiction to the traditional CT scheme of the monoclonal antibody. This result was even more evident in the HER2-enriched population, with 3+ or 2+ immunohistochemistry and FISH-positive. Several countries routinely use this drug as standard treatment in AGC.
Lapatinib is another tyrosinkynase inhibitor against Epithelial Grow Factor Receptor (EGFR), usually applied in the treatment of breast cancer. The phase II trials that tested it for AGC showed no increase in OS (24, 25).
EGFR over-expression in gastric cancer is happens in 30-50% of cases (25, 26) and tests of new drugs against this agent have been done only in metastatic or inoperable cancers. Cetuximab (Erbitux®) and Panitumumab (Vectibix, Amgen) usage brought discordant results but it seems to slightly improve the progression free survival in AGC (26, 27). Several trials are needed to estimate the real benefit and the eventual translation in operable gastric cancer in perioperative settings. Other molecules have demonstrated their ineffectiveness in gastric cancer (Gefitinib (Iressa®, AstraZeneca Pharmaceuticals) and Erlotinib (Tarceva®, Roche-Genetech) (26).
The role of angiogenesis in tumoral growth and survival and metastatic diffusion are well known pathogenetic factors. For this reason vascular endothelial growth factor (VEGF) and its receptors (VEGFR-1 and VEGFR-2) are main molecular targets of some novel drugs. Bevacizumab, a monoclonal antibody against VEGF, was at the beginning applied in colorectal, lung, ovarian, and renal cell cancers. Two randomized phase III trial, the AVAGAST and the AVATAR trials studied its application in advanced gastric cancer (28, 29). Bevacizumab insertion in treatment algorithm of AGC showed no difference in overall survival but improved progression free survival and overall response rate.
Intra-peritoneal chemotherapy
Gastric cancer cells diffuse mainly through lymphatic flow and via cell seeding after serosa invasion. The 53-60% of patients affected by AGC present peritoneal carcinosis (PC) (stage III-IV), and the 40% hepatic metastases (30) (31). Moreover, the main cause of death is PC, despite R0 resections associated to systemic CT and/or radiotherapy (30, 32-34).
A meta-analysis (32) evaluated the effect of intra-peritoneal chemotherapy (IPC) associated to cytoreductive surgery (CRS) compared with surgery alone, in patients with AGC with or without peritoneal, nodal and distant metastasis. This analysis of 20 RCTs (2145 patients) reported an increase in morbidity rate in the IPC group, but also an improvement in OS, in overall recurrence rate, in hematogenous metastasis rate and in peritoneal recurrence rate in the IPC group. No statistically significant difference in lymph nodal recurrence rate was found.
Another meta-analysis (33) reported the effects of IPC and R0 resections on patients with AGC without PC compared with surgery alone. 16 RCTs (1906 pts.) were included. An increase in survival rate at 1, 2, 3, 5, 9 years and a significant reduction in recurrence rate after 2, 3, 5 years were reported in IPC group. No increase in anastomotic leakage, ileus, bowel perforation, myelosuppression, gastrointestinal reaction and hepatic failure were associated to IPC, only an increased the incidence of abdominal pain.
Lastly, another meta-analysis (35) reported an increased OS in IPC group particularly compared to surgery alone in patients with serosal invasion with no macroscopic spread of disease.
From these data results the feasibility of prophylactic IPC associated to neoadjuvant chemotherapy in order to increase the DFS and OS in patients with AGC without PC.
IPC was considered also in a neoadjuvant setting. In 2012 Yonemura et al. (36) (37) proposed a new therapeutic approach called “bidirectional chemotherapy” which consisted in a neoadjuvant intraperitoneal and systemic chemotherapy (NIPS) that can act on PC from the inside of peritoneum and from the subperitoneal blood vessels. He proposed a drug regimen with oral S-1, i.v. taxotere and cisplatinum and intraperitoneal cisplatinum and docetaxel with good result in terms of CC-0 achievement during surgery, DFS and OS.
The role of intra-peritoneal cytology
The finding of free intraperitoneal tumor cells (FITC) has a fundamental importance in defining the prognosis of patients with AGC (38-40). Positive cytology is described in 11 to 27% of patients with gastric cancer (41).
When gastric serosa is involved, PC could be considered practically unavoidable (32). In case of free peritoneal tumor cells in abdominal cavity the natural evolution in PC occur in 80% of cases, with a distant survival near to 0% (42). PC was considered the more important prognostic factor (more than T or N) for advanced disease, early recurrence, and decreased disease-specific survival following curative resection in patients with AGC (38). In the AJCC-NCCN TNM classification, the positive cytology at the staging laparoscopy is considered as M1 disease (43-45).
The main criticism of peritoneal washing cytology remains its low sensitivity (14-70% reported in the literature, but these rates are in heterogeneous cohort of patients and stage of disease) (39). To improve sensitivity, Homma et al. (40) suggested to perform the washing in multiple cavities (in the right and left subphrenic space, inside the omental bursa, and in the Douglas pouch), and not only in the Douglas pouch (41). Furthermore, with the introduction of new molecular techniques, some studies directly compared cytology by Papanicolaou staining with molecular detection by PCR. Detection methods using PCR offer considerably higher sensitivity and a marginally lower specificity (46).
A meta-analysis including 12883 patients revealed FITC to be associated with poor overall survival poor peritoneal recurrence free survival, regardless of the detection method (47).
Then FITC represents an “in fieri ” PC, practically comparing patients with FITC to those with PC in terms of survival. A meta-analysis focusing on the effect of IPC on patients with AGC with FITC and without macroscopic PC showed that 2- and 5-years survival was increased by IPC (RR=1.62, RR=3.10). Two- and 5-years survival was further increased by IPC associated with peritoneal lavage (PL) (RR=2.33, RR=6.19). Furthermore, peritoneal recurrence was reduced by IPC (OR=0.45) and by IPC with PL (OR=0.13) (48).
Conclusions
Gastric cancer is an aggressive disease with a high risk of peritoneal dissemination even at early stages. The surgical therapy of gastric cancer should be based on radical surgery aiming to eradicate all the macroscopic disease and perform adequate lymphadenectomy. As the peritoneal dissemination of gastric cancer is the main cause of long-term failure of the treatment, a peritoneal fluid cytology should always be done. However, the uncertainty of its results suggests preventing peritoneal dissemination and subsequent carcinosis with an early use of the intraperitoneal CT. Moreover, the use of perioperative and bidirectional CT should be considered. AGC with invasion of serosa and/or positive cytology at stadiation laparoscopy should be treated in experienced centers in order to introduce the use of “prophylactic IPC” even in absence of macroscopic peritoneal dissemination associated to perioperative CT regimen.
References
- 1.Schuhmacher C, Gretschel S, Lordick F, et al. Neoadjuvant chemotherapy compared with surgery alone for locally advanced cancer of the stomach and cardia: European Organisation for Research and Treatment of Cancer randomized trial 40954. J Clin Oncol. 2010;28:5210–8. doi: 10.1200/JCO.2009.26.6114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Songun I, Keizer HJ, Hermans J, Klementschitsch P, de Vries JE, Wils JA, van der Bijl J, van Krieken JH, van de Velde CJ. Chemotherapy for operable gastric cancer: results of the Dutch randomised FAMTX trial. The Dutch Gastric Cancer Group (DGCG) Eur J Cancer. 1999;35:558–62. doi: 10.1016/s0959-8049(98)00429-8. [DOI] [PubMed] [Google Scholar]
- 3.Hartgrink HH, van de Velde CJ, Putter H, Songun I, Tesselaar ME, Kranenbarg EK, de Vries JE, Wils JA, van der Bijl J, van Krieken JH. Group., Cooperating Investigators of The Dutch Gastric Cancer. Neo-adjuvant chemotherapy for operable gastric cancer: long term results of the Dutch randomised FAMTX trial. Eur J Surg Oncol. 2004;30:643–9. doi: 10.1016/j.ejso.2004.04.013. [DOI] [PubMed] [Google Scholar]
- 4.Coccolini F, Nardi M, Montori G, Ceresoli M, Celotti A, Cascinu S, Fugazzola P, Tomasoni M, Glehen O, Catena F, Yonemura Y, Ansaloni L. Neoadjuvant chemotherapy in advanced gastric and esophago-gastric cancer. Meta-analysis of randomized trials. Int J Surg. 2018 Mar;51:120–127. doi: 10.1016/j.ijsu.2018.01.008. doi: 10.1016/j.ijsu.2018.01.008. Epub 2018 Feb 20. [DOI] [PubMed] [Google Scholar]
- 5.Cunningham D, Allum WH, Stenning SP, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355:11–20. doi: 10.1056/NEJMoa055531. [DOI] [PubMed] [Google Scholar]
- 6.Ychou M, Boige V, Pignon JP, et al. Perioperative chemotherapy compared with surgery alone for resectable gastroesophageal adenocarcinoma: an FNCLCC and FFCD multicenter phase III trial. J Clin Oncol. 2011;29:1715–1721. doi: 10.1200/JCO.2010.33.0597. [DOI] [PubMed] [Google Scholar]
- 7.Ronellenfitsch U, Schwarzbach M, Hofheinz R, Kienle P, Kieser M, Slanger TE, Jensen K. and Group., GE Adenocarcinoma Meta-analysis. Perioperative chemo(radio)therapy versus primary surgery for resectable adenocarcinoma of the stomach, gastroesophageal junction, and lower esophagus. Cochrane database Syst Rev. 2013 May 31;5:CD008107. doi: 10.1002/14651858.CD008107.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Reim D, Gertler R, Novotny A, Becker K, Zum Büschenfelde C, Ebert M, et al. Adenocarcinomas of the esophagogastric junction are more likely to respond to preoperative chemotherapy than distal gastric cancer. Ann Surg Oncol. 2012;19:2108–2118. doi: 10.1245/s10434-011-2147-8. [DOI] [PubMed] [Google Scholar]
- 9.Zhao JH, Gao P, Song YX, Sun JX, Chen XW, Ma B, Yang YC, Wang ZN. Which is better for gastric cancer patients, perioperative or adjuvant chemotherapy: a meta-analysis. BMC Cancer. 2016 Aug 12;16:631. doi: 10.1186/s12885-016-2667-5. doi: 10.1186/s12885-016-2667-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.GASTRIC (Global Advanced/Adjuvant Stomach Tumor Research International Collaboration) Group. Paoletti X, Oba K, Burzykowski T, et al. Benefit of adjuvant chemotherapy for resectable gastric cancer: a meta-analysis. JAMA. 2010 May 5;303(17):1729–37. doi: 10.1001/jama.2010.534. [DOI] [PubMed] [Google Scholar]
- 11.Sasako M, Sakuramoto S, Katai H, et al. Fiveyear 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. doi: 10.1200/JCO.2011.36.5908. [DOI] [PubMed] [Google Scholar]
- 12.Bang YJ, Kim YW, Yang HK, et al. Adjuvant capecitabine and oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): a phase 3 openlabel, randomised controlled trial. Lancet. 2012;379:315–321. doi: 10.1016/S0140-6736(11)61873-4. [DOI] [PubMed] [Google Scholar]
- 13.Noh SH, Park SR, Yang HK, et al. and investigators., CLASSIC trial. Adjuvant capecitabine plus oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): 5-year follow-up of anopen-label, randomised phase 3 trial. Lancet Oncol. 2014 Nov;15(12):1389–96. doi: 10.1016/S1470-2045(14)70473-5. [DOI] [PubMed] [Google Scholar]
- 14.Moon JH, Fujiwara Y, Hirao M, Imamura H, Kimura Y, Fujitani K, Fujita J, Tamura S, Takiguchi S, Yano M, Mori M, Doki Y. Randomized Controlled Trial of Adjuvant Chemotherapy with Fluoropyrimidines Versus Surgery-alone for Gastric Cancer. Anticancer Res. 2017 Jun;37(6):3061–3067. doi: 10.21873/anticanres.11661. [DOI] [PubMed] [Google Scholar]
- 15.Namikawa T, Maeda H, Kitagawa H, Oba K, Tsuji A, Yoshikawa T, Kobayashi M, Hanazaki K. Treatment using oxaliplatin and S-1 adjuvant chemotherapy for pathological stage III gastric cancer: a multicenter phase II study (TOSA trial) protocol. BMC Cancer. 2018 Feb 13;18(1):186. doi: 10.1186/s12885-018-4109-z. doi: 10.1186/s12885-018-4109-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Sakuramoto S, Sasako M, Yamaguchi T, Kinoshita T, Fujii M, Nashimoto A, Furukawa H, Nakajima T, Ohashi Y, Imamura H, Higashino M, Yamamura Y, Kurita A, Arai K ACTS-GC Group. Adjuvant chemotherapy for gastric cancer with S-1, an oral fluoropyrimidine. N Engl J Med. 2007;357(18):1810–1820. doi: 10.1056/NEJMoa072252. doi: 10.1056/NEJMoa072252. [DOI] [PubMed] [Google Scholar]
- 17.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(33):4387–4393. doi: 10.1200/JCO.2011.36.5908. doi: 10.1200/JCO.2011.36.5908. [DOI] [PubMed] [Google Scholar]
- 18.Liang JW, Zhang JJ, Zhang T, Zheng ZC. Clinicopathological and prognostic significance of HER2 overexpression in gastric cancer: a meta-analysis of the literature. Tumour Biol. 2014 May;35(5):4849–58. doi: 10.1007/s13277-014-1636-3. [DOI] [PubMed] [Google Scholar]
- 19.Gu J, Zheng L, Wang Y, Zhu M, Wang Q, Li X. Prognostic significance of HER2 expression based on trastuzumab for gastric cancer (ToGA) criteria in gastric cancer: an updated meta-analysis. Tumour Biol. 2014 Jun;35(6):5315–21. doi: 10.1007/s13277-014-1693-7. [DOI] [PubMed] [Google Scholar]
- 20.Chen C, Yang JM, Hu TT, Xu TJ, Yan G, Hu SL, Wei W, Xu WP. Prognostic role of human epidermal growth factor receptor in gastric cancer: a systematic review and meta-analysis. Arch Med Res. 2013 Jul;44(5):380–9. doi: 10.1016/j.arcmed.2013.07.001. [DOI] [PubMed] [Google Scholar]
- 21.Chua TC, Merrett ND. Clinicopathologic factors associated with HER2-positive gastric cancer and its impact on survival outcomes--a systematic review. Int J Cancer. 2012 Jun 15;130(12):2845–56. doi: 10.1002/ijc.26292. [DOI] [PubMed] [Google Scholar]
- 22.Kurokawa Y, Matsuura N, et al. Multicenter large-scale study of prognostic impact of HER2 expression in patients with resectable gastric cancer. Gastric Cancer. 2014 Sep 16 doi: 10.1007/s10120-014-0430-7. [DOI] [PubMed] [Google Scholar]
- 23.Bang YJ, Van Cutsem E, Feyereislova A, et al. and Investigators, ToGA Trial. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet. 2010 Aug 28;376(9742):687–97. doi: 10.1016/S0140-6736(10)61121-X. [DOI] [PubMed] [Google Scholar]
- 24.Satoh T, Xu RH, Chung HC, et al. Lapatinib plus paclitaxel versus paclitaxel alone in the second-line treatment of HER2-amplified advanced gastric cancer in Asian populations: TyTAN--a randomized, phase III study. J Clin Oncol. 2014 Jul 1;32(19):2039–49. doi: 10.1200/JCO.2013.53.6136. [DOI] [PubMed] [Google Scholar]
- 25.Kothari N, Almhanna K. Current status of novel agents in advanced gastroesophageal adenocarcinoma. J Gastrointest Oncol. 2015 Feb;6(1):60–74. doi: 10.3978/j.issn.2078-6891.2014.098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Cappetta A, Lonardi S, Pastorelli D, Bergamo F, Lombardi G, Zagonel V. Advanced gastric cancer (GC) and cancer of the gastro-oesophageal junction (GEJ): focus on targeted therapies. Crit Rev Oncol Hematol. 2012 Jan;81(1):38–48. doi: 10.1016/j.critrevonc.2010.12.006. [DOI] [PubMed] [Google Scholar]
- 27.Lordick F, Kang YK, Chung HC, et al. and Investigators., Arbeitsgemeinschaft Internistische Onkologie and EXPAND. Capecitabine and cisplatin with or without cetuximab for patients with previously untreated advanced gastric cancer (EXPAND): a randomised, open-label phase3 trial. Lancet Oncol. 2013 May;14(6):490–9. doi: 10.1016/S1470-2045(13)70102-5. [DOI] [PubMed] [Google Scholar]
- 28.Ohtsu A, Shah MA, Van Cutsem E, et al. Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a randomized, double-blind, placebo-controlled phase III study. J Clin Oncol. 2011 Oct 20;29(30):3968–76. doi: 10.1200/JCO.2011.36.2236. [DOI] [PubMed] [Google Scholar]
- 29.Shen L, Li J, Xu J, et al. Bevacizumab plus capecitabine and cisplatin in Chinese patients with inoperable locally advanced or metastatic gastric or gastroesophageal junction cancer: randomized, double-blind, phase III study (AVATAR study) Gastric Cancer. 2015 Jan;18(1):168–76. doi: 10.1007/s10120-014-0351-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Montori G, Coccolini F, Ceresoli M, Catena F, Colaianni N, Poletti E, Ansaloni L. The treatment of peritoneal carcinomatosis in advanced gastric cancer: state of the art. Int J Surg Oncol. 2014;2014:912418. doi: 10.1155/2014/912418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Coccolini F, Gheza F, Lotti M, Virzì S, Iusco D, Ghermandi C, Melotti R, Baiocchi G, Giulini SM, Ansaloni L, Catena F. Peritoneal carcinomatosis. World J Gastroenterol. 2013 Nov 7;19(41):6979–94. doi: 10.3748/wjg.v19.i41.6979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Coccolini F, Cotte E, Glehen O, Lotti M, Poiasina E, Catena F, Yonemura Y, Ansaloni L. Intraperitoneal chemotherapy in advanced gastric cancer. Meta-analysis of randomized trials. Eur J Surg Oncol. 2014 Jan;40(1):12–26. doi: 10.1016/j.ejso.2013.10.019. [DOI] [PubMed] [Google Scholar]
- 33.Mi DH, Li Z, Yang KH, et al. Surgery combined with intraoperative hyperthermic intraperitoneal chemotherapy (IHIC) for gastric cancer: a systematic review and meta-analysis of randomised controlled trials. Int J Hyperthermia. 2013;29(2):156–67. doi: 10.3109/02656736.2013.768359. [DOI] [PubMed] [Google Scholar]
- 34.Sadeghi B, Arvieux C, Gilly FN, et al. Peritoneal carcinomatosis from non-gynecologic malignancies: results of the EVOCAPE 1 multicentric prospective study. Cancer. 2000 Jan 15;88(2):358–63. doi: 10.1002/(sici)1097-0142(20000115)88:2<358::aid-cncr16>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
- 35.Sun J, Song Y, Wang Z, Gao P, Chen X, Xu Y, Liang J, Xu H. Benefits of hyperthermic intraperitoneal chemotherapy for patients with serosal invasion in gastric cancer: a meta-analysis of the randomized controlled trials. BMC Cancer. 2012 Nov 16;12:526. doi: 10.1186/1471-2407-12-526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Canbay E, Mizumoto A, Ichinose M, et al. Outcome data of patients with peritoneal carcinomatosis from gastric origin treated by a strategy of bidirectional chemotherapy prior to cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in a single etc. Ann Surg Oncol. 2014 Apr;21(4):1147–52. doi: 10.1245/s10434-013-3443-2. [DOI] [PubMed] [Google Scholar]
- 37.Yonemura Y, Elnemr A, Endou Y, et al. Effects of neoadjuvant intraperitoneal/systemic chemotherapy (bidirectional chemotherapy) for the treatment of patients with peritoneal metastasis from gastric cancer. Int J Surg Oncol. 2012;2012:148420. doi: 10.1155/2012/148420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.De Andrade JP, Mezhir JJ. The critical role of peritoneal cytology in the staging of gastric cancer: an evidence-based review. J Surg Oncol. 2014 Sep;110(3):291–7. doi: 10.1002/jso.23632. doi: 10.1002/jso.23632. Epub 2014 May 22. Review. [DOI] [PubMed] [Google Scholar]
- 39.Ang CW, Tan LC. Peritoneal cytology in the staging process of gastric cancer: do or don’t? J Gastroint Dig Syst. 2013;3:5. [Google Scholar]
- 40.Homma Y, Ushida S, Yamada M, Kobayashi H, Suzuki K. Positive peritoneal washing cytology in multiple cavities can predict poor prognosis of advanced gastric cancer patients. Ann Surg Oncol. 2010;17:455–460. doi: 10.1245/s10434-009-0764-2. [DOI] [PubMed] [Google Scholar]
- 41.Kano Y, Kosugi SI, Ishikawa T, Otani T, Muneoka Y, Sato Y, Hanyu T, Hirashima K, Bamba T, Wakai T. Prognostic significance of peritoneal lavage cytology at three cavities in patients with gastric cancer. Surgery. 2015 May 6 doi: 10.1016/j.surg.2015.04.004. [DOI] [PubMed] [Google Scholar]
- 42.Kodera Y, Yamamura Y, Shimizu Y, et al. Peritoneal washing cytology: prognostic value of positive findings in patients with gastric carcinoma undergoing a potentially curative resection. J Surg Oncol. 1999 Oct;72(2):60–4. doi: 10.1002/(sici)1096-9098(199910)72:2<60::aid-jso3>3.0.co;2-1. discussion 64-5. [DOI] [PubMed] [Google Scholar]
- 43.Ajani A, Bentrem D, Besh S, et al. NCCN Clinical Practice Guidelines in Oncology. Gastric Cancer. 2013 Version 2.2013: www.nccn.org . [Google Scholar]
- 44.S Edge. New York: Springer; 2010. Cancer AJCo: AJCC cancer staging manual. [Google Scholar]
- 45.Ajani JA, In H, Sano T, et al. Stomach, Amin MB E. AJCC Cancer Staging Manual. 2017 [Google Scholar]
- 46.Fujiwara Y, Okada K, Hanada H, Tamura S, Kimura Y, Fujita J, Imamura H, Kishi K, Yano M, Miki H, Okada K, Takayama O, Aoki T, Mori M, Doki Y. The clinical importance of a transcription reverse-transcription concerted (TRC) diagnosis using peritoneal lavage fluids in gastric cancer with clinical serosal invasion: a prospective, multicenter study. Surgery. 2014 Mar;155(3):417–23. doi: 10.1016/j.surg.2013.10.004. [DOI] [PubMed] [Google Scholar]
- 47.Pecqueux M, Fritzmann J, Adamu M, Thorlund K, Kahlert C, Reißfelder C, Weitz J, Rahbari NN. Free intraperitoneal tumor cells and outcome in gastric cancer patients: a systematic review and meta-analysis. Oncotarget. 2015 Nov 3;6(34):35564–78. doi: 10.18632/oncotarget.5595. doi: 10.18632/oncotarget.5595. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Coccolini F, Catena F, Glehen O, Yonemura Y, Sugarbaker PH, Piso P, Ceresoli M, Montori G, Ansaloni L. Effect of intraperitoneal chemotherapy and peritoneal lavage in positive peritoneal cytology in gastric cancer. Systematic review and meta-analysis. Eur J Surg Oncol. 2016 Sep;42(9):1261–7. doi: 10.1016/j.ejso.2016.03.035. doi: 10.1016/j.ejso.2016.03.035. Epub 2016 Apr 19. [DOI] [PubMed] [Google Scholar]
- 49.Bang YJ, Kim YW, Yang HK, Chung HC, Park YK, Lee KH, Lee KW, Kim YH, Noh SI, Cho JY, Mok YJ, Kim YH, Ji J, Yeh TS, Button P, Sirzén F, Noh SH. CLASSIC trial investigators. Adjuvant capecitabine and oxaliplatin for gastric cancer after D2 gastrectomy (CLASSIC): a phase 3 open-label, randomised controlled trial. Lancet. 2012;379(9813):315–321. doi: 10.1016/S0140-6736(11)61873-4. doi: 10.1016/S0140-6736(11)61873-4. [DOI] [PubMed] [Google Scholar]