Review Open Access
Copyright ©The Author(s) 2016. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Sep 7, 2016; 22(33): 7478-7485
Published online Sep 7, 2016. doi: 10.3748/wjg.v22.i33.7478
Regional but fatal: Intraperitoneal metastasis in gastric cancer
Jia Wei, Nan-Die Wu, Bao-Rui Liu, Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, Nanjing 210008, Jiangsu Province, China
Author contributions: Wei J and Wu ND contributed to literature review; Liu BR revised the article and all authors have read and approved the final version to be published.
Supported by National Natural Science Foundation of China, No. 81220108023, No. 81370064 and No. 81572329; Fundamental Research Funds for the Central Universities, No. 20620140729; Jiangsu Provincial Program of Medical Sciences, No. BL2012001; and Distinguished Young Investigator Project of Nanjing, No. JQX12002.
Conflict-of-interest statement: The authors have no conflict of interest related to the manuscript.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Bao-Rui Liu, MD, PhD, Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University, Clinical Cancer Institute of Nanjing University, 321 Zhongshan Road, Nanjing 210008, Jiangsu Province, China. baoruiliu@nju.edu.cn
Telephone: +86-25-83107081 Fax: +86-25-83317016
Received: April 18, 2016
Peer-review started: April 19, 2016
First decision: May 12, 2016
Revised: May 15, 2016
Accepted: June 15, 2016
Article in press: June 15, 2016
Published online: September 7, 2016

Abstract

Peritoneal carcinomatosis appears to be the most common pattern of metastasis or recurrence and is associated with poor prognosis in gastric cancer patients. Many efforts have been made to improve the survival in patients with peritoneal metastasis. Hyperthermic intraperitoneal chemotherapy remains a widely accepted strategy in the treatment of peritoneal dissemination. Several phase II-III studies confirmed that the combined cytoreducitve surgery and hyperthermic intraperitoneal chemotherapy resulted in longer survival in patients with peritoneal carcinomatosis. In addition, proper selection and effective regional treatment in patients with high risk of peritoneal recurrence after resection will further improve prognosis in local advanced gastric cancer patients.

Key Words: Gastric cancer, Intraperitoneal metastasis, Regional metastasis, Cytoreductive surgery, Hyperthermic intraperitoneal chemotherapy

Core tip: The recurrence rate of gastric cancer after surgery within 2 years remains at 79%. Gastric cancer patients with peritoneal metastases have a median survival of only 3.1 mo. Understanding the influence of peritoneal metastasis on survival in gastric cancer patients, the potential molecular mechanism of peritoneal metastasis, and individualized treatment of patients with high risk of peritoneal metastasis is essential for selecting effective treatment strategies in advanced gastric cancer. In this review, we summarized translational and clinical researches on peritoneal carcinomatosis, providing comprehensive information to better understand the fatal role of peritoneal metastasis in gastric cancer.



INTRODUCTION

Gastric adenocarcinoma is the fourth most common cancer and the second leading cause of cancer-related death worldwide[1]. Apart from countries with national screening programs such as Japan and South Korea, most gastric cancer patients present with advanced disease because early-stage tumors are usually asymptomatic and often develop metastatic recurrences even after curative resection. Despite improvements in the surgical treatment of gastric adenocarcinoma, a high recurrence rate persists, with a 5-year overall survival rate for all diagnosed patients of only 24.5% in Europe[2] and 40%-60% in Asia[3,4].The most frequent cause of treatment failure following surgery for gastric cancer is peritoneal dissemination, mainly caused by the seeding of free cancer cells from the primary gastric cancer, which is the most common type of spread. Gastric cancer patients with evidence of macroscopic peritoneal carcinomatosis have very poor prognoses, with a median overall survival of 3-6 mo[5,6].

In this review, we aim to summarize the influence of peritoneal metastasis on survival in gastric cancer patients, the potential molecular mechanism of peritoneal metastasis, and individualized treatment of patients who have high risk of peritoneal metastasis.

PERITONEAL METASTASIS IS THE MOST IMPORTANT FACTOR FOR PROGNOSIS IN GASTRIC CANCER

The Recurrence rate of gastric cancer remains high, particularly in patients with advanced stage disease. Among patients receiving R0 resection, 79% have documented recurrences within 2 years, and the median time to death from the time of recurrence is 6 mo[7]. Many patients, especially those with stage III disease, develop locoreginal recurrence, peritoneal recurrence, or distant metastasis[8]. Many investigators have analyzed recurrence patterns of gastric cancer after curative surgery, but the data have shown variable incidences of these patterns. Schwarz et al[9] found that the most common pattern was distant metastasis while Eom et al[10] found that hematogenous metastasis was most common among patients with early recurrence and locoregional and peritoneal recurrence among patients with late recurrence using a cutoff time of one year after curative resection for patient subgroups. This disagreement was attributed to differences in patient cohorts undergoing evaluation, the cutoff at which recurrence was determined, and the methods for determining recurrence patterns. In addition, autopsy studies revealed only end-stage disease, but not early recurrence patterns, and re-operation series probably reflect early locoregional and peritoneal recurrence. Laparoscopy and peritoneal cytology have been shown to detect occult metastatic disease not seen on conventional imaging[11].

A recent study of 1178 patients with metastatic or recurrent gastric cancer showed that about 46% of patients had peritoneal metastases and about 30% had liver metastases[12]. Several other clinical studies have reported recurrence patterns in a population of patients with early stage to advanced disease[7,9,13-15], showing that 30%-54% of patients had peritoneal recurrence alone or in combination.

Our unpublished data showed in a total number of 349 patients with stage III and IV gastric cancer, peritoneal metastasis was detected in 62.8% of the patients. And 81.1% of the patients developed metastasis in peritoneal cavity (peritoneal and liver) at the time of recurrence or diagnosis. Furthermore, peritoneal cancer involvement is associated with poor prognosis and quality of life compared with metastasis to other organs. Our research showed that stage IV patients with peritoneal metastasis had shorter survival (7.5 mo vs 14 mo) and a higher risk of mortality (HR = 2.026, P = 0.004).

MOLECULAR MECHANISMS OF PERITONEAL METASTASIS

Cancer cells are thought to undergo the following sequential steps to form peritoneal metastases: (1) penetration of cancerous tissues into the visceral serosa; (2) exfoliation of the cancer cells from the primary tumor; (3) dissemination and survival of the cancer cells within the abdominal cavity; (4) adhesion of cancer cells to the peritoneum; (5) invasion of cancer cells through the peritoneal membrane; and (6) formation of the peritoneal metastasis[16]. However, the mechanisms governing the formation of peritoneal metastasis remain poorly understood. A global expression profile of 21168 genes was analyzed in a gastric cancer cell line established from a primary main tumor and other cell lines established from the metastasis to the peritoneal cavity. They found that 24 genes of cell adhesion, epithelial markers, drug metabolism and signal transduction were up-regulated and 17 genes of immune response, cell cycle and adhesion were down-regulated[17] (Table 1). Loss of hypoxia inducible factor-1α may accelerate the development of peritoneal dissemination via the upregulation of matrix metalloproteinases (MMP) -1 in gastric cancer cells, which was manifested in a mouse model[18]. MMP-7-positive gastric cancer patients have significantly poorer overall survival and die more frequently of peritoneal recurrence than those patients with MMP-7-negative tumors in a Japanese cohort[19]. Another comparative analysis between the parental cell line GC9811 and its highly metastatic peritoneal counterpart, cell line GC9811-P revealed and confirmed that recombinant human S100 calcium binding protein A4 (S100A4) and cadherin-associated protein beta 1 (CTNNB1) were upregulated and phosphatase and tensin homolog deleted on chromosome ten was downregulated in GC9811-P cells. Identification of these differentially expressed genes could disclose the molecular mechanisms involved and provide new targets for therapeutic intervention to avoid peritoneal dissemination of gastric adenocarcinoma[20]. A recent study revealed that intraoperative hemorrhages were strongly correlated with peritoneal recurrence, probably due to an increased ability of cancer cells and mesothelial cells to adhere to each other in the presence of factors in plasma[16]. Zinc protoporphyrin IX[21] androquoishomeobox protein 1 (IRX1)[22] was reported to inhibit peritoneal metastasis via neovascularization. Identification of these differentially expressed genes could disclose the molecular mechanisms involved and provide new targets for therapeutic intervention to avoid peritoneal dissemination of gastric adenocarcinoma. At present, chemokinereceptor 5 antagonism can reduce the potential for gastric cancer cell dissemination[23]. P38- mitogen-activated protein kinase inhibition by targeted small molecule inhibitor was demonstrated to be beneficial in preventing the peritoneal dissemination in poorly differentiated gastric cancer[24]. Nevertheless, the molecular mechanisms of peritoneal dissemination need to be further clarified to provide more information for peritoneal dissemination therapy.

Table 1 Twenty-four up-regulated and 17 down-regulated genes in gastric cancer cells from malignant ascites compared with those from primary lesions.
Gene expression levelGene nameGene function
Down-regulated
Nucleobinding 2Signaling (apoptosis)
Acyl-Coenzyme A dehydrogenaseSignaling
Chaperonin containing TCP1Signaling
FKBP54Signaling
Histone deacetylase 3Signaling
p27kipSignaling
PAK-interacting exchange factor alphaSignaling
CD4Immune response
IL4 statImmune response
L2 receptor gammaImmune response
IGFBP2Growth and metabolism
RAD51 homologue CChromosome stability
Heterogenous nuclear ribonucleoproteinCell adhesion
Integrin β4Cell adhesion
Tubulin beta-1 chainCell adhesion
Death associated proteinApoptosis
H2A histone family member LApoptosis
Up-regulated
Dopa decarboxylaseSignaling or progression
Caveolin-3Signaling (modification)
CD9Signaling
Dystroglycan1Signaling
Inositol triphosphate receptorSignaling
LMO 7Signaling
Sodium/hydrogen exchanger, isoform 1Signaling
Cystein protease (legumain)Invasion
Myosin 6Intracellular organelle transport
Destrin (actin depolymerising factor)Interaction with extracellular matrix
Renal tumor antigen RAGE1Immune response
Aldehyde dehydrogenaseDrug metabolism
Aldo-keto reductase family 1Drug metabolism
Keratin 14Cell adhesion, invasion
Keratin 7Cell adhesion, invasion
Keratin 8Cell adhesion, invasion
CD44Cell adhesion
Desmoplakin (DPI, DPII)Cell adhesion
Galectin 3 (lectin)Cell adhesion
Integrin alpha3Cell adhesion
OccludinCell adhesion
S100 A10 (ligand of Annexin II)Cell adhesion
Leukocyte elastase inhibitorApoptosis
TGFb-induced anti-apoptotic factorApoptosis
EFFECTIVE TREATMENTS FOR PATIENTS WITH PERITONEAL METASTASIS

Patients with peritoneal carcinomatosis of gastric origin have an extremely bad prognosis. Systemic chemotherapy improves median survival in metastatic gastric cancer to 7-10 mo, but in patients with peritoneal carcinomatosis from gastric cancer, the same improvement has not been reported[25]. And 20%-50% of patients treated with radical surgery will develop postoperative peritoneal recurrence[26], and intraperitoneal spread of tumor cells was observed in 54% of patients who died of recurrence after surgery for advanced gastric cancer[27].

At present, hyperthermic intraperitoneal chemotherapy (HIPEC) is the most widely accepted strategy among the treatment options for peritoneal dissemination which is the most frequent metastatic pattern in gastric cancer[28]. The theoretical advantage of the HIPEC is to add the direct cytotoxic effects of heat to a high local concentration of used cytostatic drug[29,30]. In addition to the mechanical washing effect, HIPEC also has the theoretical advantage of delivering a higher anticancer drug concentration into abdominal lavage with reduced systemic toxicity. There are many molecular explanations for the effect of HIPEC. For example, induction of apoptosis, alterations of cell membrane property, changes in intracellular proteins and in their synthesis and heat inhibition of DNA repair enhanced by inhibitors of the cellular heat-shock response[31,32].

In gastric cancer patients with peritoneal carcinomatosis, surgical treatments aiming at removing the primary lesion of peritoneal dissemination is palliative. The combination of cytoreductive surgery (CRS) and HIPEC was first described in 1980 by Spratt et al[33]. In the following years, Sugarbaker and his colleagues applied and introduced this innovative technique for peritoneal carcinomatosis[34]. Phase II-III studies revealed that patients who received CRS plus HIPEC obtained better survival results only if completeness of cytoreduction (CCR-0) resection was achieved. However, the survival benefit of HIPEC remains extremely low if cytoreductive surgery can not accomplish sufficient down-staging of the carcinomatosis burden[35-37]. The largest experience published so far was a retrospective French study involving 159 patients which confirmed this combination advantage in a selected CCR0 group of patients[35]. The dismal effect of HIPEC in patients with extensive peritoneal carcinomatosis not amenable to downstaging to CCR-0 may be explained by limited drug penetration leading to no anti-tumor effect on the deeply invasive microfoci[38]. Thus, drug delivery system with high permeability has the potential perspective role in the treatment of extensive peritoneal carcinomatosis cases[39].

OPTIONAL AGENTS FOR INTRAPERITONEAL TREATMENT

Even though multimodal treatment strategies have been used to improve the prognosis of gastric cancer patients with peritoneal recurrence, the results remain unsatisfactory[40]. The oral anticancer drug S1 is a fluoropyrimidine derivative, combining tegafur with two modulators. A recent meta-analysis showed that the use of S1 monotherapy was associated with a significant survival benefit (HR = 0.48, 95%CI: 0.32-0.70, P = 0.0002)[41]. The advantage of S1 over other chemotherapeutic agents is its ability to attain higher concentrations intraperitoneally, due to the higher concentrations of 5-FU and CDHP achieved in peritoneal tumors than in plasma[42,43].

In addition to S1, paclitaxel and docetaxel, which binds to tubulin, leading to microtubule stabilization, and mitotic arrest, also have high sensitivity against diffuse-type adenocarcinoma, which is a common type of peritoneal tumor. And some of these compounds are transported into the peritoneal cavity when administered intravenously[44].

There have been numerous studies evaluating intraperitoneal drug delivery in gastric cancer. Intraperitoneal administration of anticancer drugs enables an extremely high concentration of drugs to directly contact the target cancer lesions in the peritoneal cavity. However, intraperitoneal administration of mitomycin C or cisplatin yielded no apparent therapeutic effects against peritoneal metastasis of gastric cancer due to immediate absorption through the peritoneum[45]. In contrast to these drugs, intraperitoneal administration of paclitaxel was developed to enhance antitumor activity against peritoneal metastasis by maintaining a high concentration of the drug in the peritoneal cavity over a long period, and its clinical effects have been verified by a number of convincing clinical trials in ovarian cancer with peritoneal metastasis.These superior results were due to the pharmacokinetic advantage of taxanes after regional delivery[46]. Taxanes are absorbed through the openings of lymphatic system, such as the milky spots and the stomata which are important sites for the formation of peritoneal dissemination[47], due to their large molecular weight and fat solubility[48]. A phase I/II study of intraperitoneal docetaxel plus S1 for the gastric cancer patients with peritoneal carcinomatosis demonstrated a superior 1-year overall survival rate of 70%, and peritoneal cytology turned negative in 81% of the patients[49]. Fujiwara et al[50] also reported a median survival of 24.6 mo ingastric cancer with peritoneal carcinomatosis treated with intraperitoneal docetaxel combined with S1.

Although intraperitoneal paclitaxel showed a profound pharmacokinetic advantage 1000 times higher than systemic administration, the main problem of intraperitoneal chemotherapy is the limited depth of penetration of anticancer drugs directly into the tumor. Accordingly, optimum use of paclitaxel may consist of intraperitoneal and intravenous administration, because intraperitoneal paclitaxel reaches the systemic circulation in only a small amount[51]. Actually, Ishigami et al[48] established intraperitoneal paclitaxel with S1 plus intravenous paclitaxel as systemic chemotherapy. The phase II study showed an overall response rate of 56% of patients with target lesions and decrease or disappearance of malignant ascites in 62% of the patients.

Another recent phase II trial in serosa-positive gastric cancer patients showed a higher similar response rate of 71.4%, and the 3- and 5-year OS rates of 78.0% and 74.9%, respectively[52].

In addition, the efficacy of intraperitoneal irinotecan has been demonstrated in several animal studies. The AUC ratio of SN-38 varied between 3.7 and 14.8 depending on the concentration of administered irinotecan[53]. Moreover, pemetrexed has been proven to be an option when used intraperitoneally in a phase I trial in ovarian cancer[54].

Except for chemotherapeutic agents, catumaxomab, a rat-mouse hybrid monoclonal antibody, was registered for the treatment of malignant ascites of various epithelial cell adhesion molecule (EpCAM) positive malignancies, including ovarian, gastric, breast and colorectal cancer. Two studies[55,56] demonstrate that this drug seems to improve progression-free survival in patients with gastric cancer (median 71 d vs 44 d, P = 0.03) and that it seems to improve the survival of patients with gastrointestinal anti-EpCAM positive tumors in intraperitoneal use.

SELECTED POPULATION FOR INTRAPERITONEAL CHEMOTHERAPY

Positive peritoneal cytology was classified as metastatic disease (M1) in the 7th edition of the American Joint Committee on Cancer tumor node metastasis staging system for gastric cancer[57]. Intraperitoneal free cancer cells isolated during peritoneal washing in patients with gastric cancer have been demonstrated to be significantly and independently related to the prognosis, influencing both recurrence and survival. It is important to prevent peritoneal recurrence after curative surgery to improve the prognosis of gastric cancer patients. However, to apply this modality, selection of patients who are at high risk for peritoneal recurrence is crucial. And, the recent trend in treatment is the administration of adjuvant intraperitoneal chemotherapy immediately after resection in patients who are at high risk of peritoneal recurrence[58,59].

Although the precise mechanism driving peritoneal recurrence remains unclear, the presence of malignant cells in the peritoneum at the time of surgery can lead to peritoneal recurrence[60,61]. Therefore, examination of peritoneal fluids has emerged as an option for identifying patients who are at high risk for peritoneal recurrence after curative resection.

Although conventional peritoneal cytology is the standard and reliable method for detecting free cancer cells in the peritoneal wash and for predicting peritoneal metastasis, in large-sample studies, approximately 4%-11% of patients will have cytology positive and therefore it is not practical or cost-effective to perform it in all patients[62]. Furthermore, it lacks sensitivity for the detection of residual cancer cells and prediction of peritoneal spread[63,64]. A recent prospective clinical study demonstrated that conventional cytology is not beneficial for predicting peritoneal recurrence after curative surgery for gastric cancer, because peritoneal washing cytology was not able to predict peritoneal recurrence or survival in gastric cancer patients[65].

A study including 655 patients indicated that intraoperatively assessed macroscopic serosal changes confer a poor prognosis and increased peritoneal recurrence for patients with curatively resected gastric cancer. Macroscopic serosal changes were defined as changes in color or nodular texture of the serosal surface on inspection and palpation. Macroscopic assessment of serosal changes may be a useful indicator that allows better risk stratification of patients with resected gastric cancer in terms of prognosis and peritoneal recurrence[66].

Recently, genetic detection using reverse transcriptase polymerase chain reaction analysis has been found more sensitive than conventional cytology. The target genes of carcinoembryonic antigen (CEA), heparanase, matrix metalloproteinase-7, cytokeratin 20, telomerase, zinc-finger E-box binding homeobox 1 and melanoma-associated gene in single or in combination were used as potent molecular markers[67-69].

However, the amplified mRNA may be derived from dead cells or phagocytes that have engulfed tumor cells, and can be released from hematopoietic cells in an inflammatory context[70]. Therefore, the clinical issue of false-positive cases remains to be addressed. Using DNA methylation or flow cytometry to identify intraperitoneal tumor cells is another valuable alteration for selecting patients who might have a high risk of peritoneal metastasis[71,72].

CONCLUSION

Gastric cancer is the second leading cause of cancer death worldwide and more than half of the gastric cancer patients show disease progression and die of peritoneal carcinomatosis. Proper selection of intraperitoneal chemotherapy in patients with peritoneal metastasis or patients with potential risk of peritoneal recurrence may be a promising approach to improve the prognosis of advanced gastric cancer patients. Administration of chemotherapeutic agents with a maintaining high concentration and a high permeability in the peritoneal cavity is an ideal choice for intraperitoneal chemotherapy. Moreover, the study of potential biomarkers from peritoneal washing could provide valuable information for a better selection of subsequent treatment combinations.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report classification

Grade A (Excellent): A

Grade B (Very good): B

Grade C (Good): C, C

Grade D (Fair): 0

Grade E (Poor): 0

P- Reviewer: Beltran MA, Caboclo JLF, Marrelli D, Nagahara H S- Editor: Ma YJ L- Editor: Ma JY E- Editor: Wang CH

References
1.  Leake PA, Cardoso R, Seevaratnam R, Lourenco L, Helyer L, Mahar A, Law C, Coburn NG. A systematic review of the accuracy and indications for diagnostic laparoscopy prior to curative-intent resection of gastric cancer. Gastric Cancer. 2012;15 Suppl 1:S38-S47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 93]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
2.  Sant M, Allemani C, Santaquilani M, Knijn A, Marchesi F, Capocaccia R. EUROCARE-4. Survival of cancer patients diagnosed in 1995-1999. Results and commentary. Eur J Cancer. 2009;45:931-991.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 597]  [Cited by in F6Publishing: 608]  [Article Influence: 40.5]  [Reference Citation Analysis (0)]
3.  Zeng WJ, Hu WQ, Wang LW, Yan SG, Li JD, Zhao HL, Peng CW, Yang GF, Li Y. Long term follow up and retrospective study on 533 gastric cancer cases. BMC Surg. 2014;14:29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 15]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
4.  Matsuda T, Saika K. The 5-year relative survival rate of stomach cancer in the USA, Europe and Japan. Jpn J Clin Oncol. 2013;43:1157-1158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
5.  Yonemura Y, Endou Y, Sasaki T, Hirano M, Mizumoto A, Matsuda T, Takao N, Ichinose M, Miura M, Li Y. Surgical treatment for peritoneal carcinomatosis from gastric cancer. Eur J Surg Oncol. 2010;36:1131-1138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 88]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
6.  Yonemura Y, Elnemr A, Endou Y, Hirano M, Mizumoto A, Takao N, Ichinose M, Miura M, Li Y. Multidisciplinary therapy for treatment of patients with peritoneal carcinomatosis from gastric cancer. World J Gastrointest Oncol. 2010;2:85-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 92]  [Cited by in F6Publishing: 76]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
7.  D’Angelica M, Gonen M, Brennan MF, Turnbull AD, Bains M, Karpeh MS. Patterns of initial recurrence in completely resected gastric adenocarcinoma. Ann Surg. 2004;240:808-816.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Chang JS, Lim JS, Noh SH, Hyung WJ, An JY, Lee YC, Rha SY, Lee CG, Koom WS. Patterns of regional recurrence after curative D2 resection for stage III (N3) gastric cancer: implications for postoperative radiotherapy. Radiother Oncol. 2012;104:367-373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 46]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
9.  Schwarz RE, Zagala-Nevarez K. Recurrence patterns after radical gastrectomy for gastric cancer: prognostic factors and implications for postoperative adjuvant therapy. Ann Surg Oncol. 2002;9:394-400.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Eom BW, Yoon H, Ryu KW, Lee JH, Cho SJ, Lee JY, Kim CG, Choi IJ, Lee JS, Kook MC. Predictors of timing and patterns of recurrence after curative resection for gastric cancer. Dig Surg. 2010;27:481-486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 43]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
11.  Leake PA, Cardoso R, Seevaratnam R, Lourenco L, Helyer L, Mahar A, Rowsell C, Coburn NG. A systematic review of the accuracy and utility of peritoneal cytology in patients with gastric cancer. Gastric Cancer. 2012;15 Suppl 1:S27-S37.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 76]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
12.  Jo JC, Ryu MH, Koo DH, Ryoo BY, Kim HJ, Kim TW, Choi KD, Lee GH, Jung HY, Yook JH. Serum CA 19-9 as a prognostic factor in patients with metastatic gastric cancer. Asia Pac J Clin Oncol. 2013;9:324-330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 24]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
13.  Deng J, Liang H, Wang D, Sun D, Pan Y, Liu Y. Investigation of the recurrence patterns of gastric cancer following a curative resection. Surg Today. 2011;41:210-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 71]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
14.  Yoo CH, Noh SH, Shin DW, Choi SH, Min JS. Recurrence following curative resection for gastric carcinoma. Br J Surg. 2000;87:236-242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 523]  [Cited by in F6Publishing: 531]  [Article Influence: 22.1]  [Reference Citation Analysis (0)]
15.  Youn HG, An JY, Choi MG, Noh JH, Sohn TS, Kim S. Recurrence after curative resection of early gastric cancer. Ann Surg Oncol. 2010;17:448-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 87]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
16.  Arita T, Ichikawa D, Konishi H, Komatsu S, Shiozaki A, Hiramoto H, Hamada J, Shoda K, Kawaguchi T, Hirajima S. Increase in peritoneal recurrence induced by intraoperative hemorrhage in gastrectomy. Ann Surg Oncol. 2015;22:758-764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
17.  Sakakura C, Hagiwara A, Nakanishi M, Shimomura K, Takagi T, Yasuoka R, Fujita Y, Abe T, Ichikawa Y, Takahashi S. Differential gene expression profiles of gastric cancer cells established from primary tumour and malignant ascites. Br J Cancer. 2002;87:1153-1161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 69]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
18.  Hiraki M, Kitajima Y, Kai K, Nakamura J, Hashiguchi K, Noshiro H, Miyazaki K. Knockdown of hypoxia-inducible factor-1α accelerates peritoneal dissemination via the upregulation of MMP-1 expression in gastric cancer cell lines. Exp Ther Med. 2012;4:355-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
19.  Yonemura Y, Endou Y, Fujita H, Fushida S, Bandou E, Taniguchi K, Miwa K, Sugiyama K, Sasaki T. Role of MMP-7 in the formation of peritoneal dissemination in gastric cancer. Gastric Cancer. 2000;3:63-70.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Bai FH, Wang NJ, Wang J, Yang L, Zhang FM, Yin F, Liang J, Wu KC, Fan DM. Screening and identification of peritoneal metastasis-related genes of gastric adenocarcinoma using a cDNA microarray. Genet Mol Res. 2012;11:1682-1689.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
21.  Shang FT, Hui LL, An XS, Zhang XC, Guo SG, Kui Z. ZnPPIX inhibits peritoneal metastasis of gastric cancer via its antiangiogenic activity. Biomed Pharmacother. 2015;71:240-246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
22.  Jiang J, Liu W, Guo X, Zhang R, Zhi Q, Ji J, Zhang J, Chen X, Li J, Zhang J. IRX1 influences peritoneal spreading and metastasis via inhibiting BDKRB2-dependent neovascularization on gastric cancer. Oncogene. 2011;30:4498-4508.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 50]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
23.  Mencarelli A, Graziosi L, Renga B, Cipriani S, D’Amore C, Francisci D, Bruno A, Baldelli F, Donini A, Fiorucci S. CCR5 Antagonism by Maraviroc Reduces the Potential for Gastric Cancer Cell Dissemination. Transl Oncol. 2013;6:784-793.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Graziosi L, Mencarelli A, Santorelli C, Renga B, Cipriani S, Cavazzoni E, Palladino G, Laufer S, Burnet M, Donini A. Mechanistic role of p38 MAPK in gastric cancer dissemination in a rodent model peritoneal metastasis. Eur J Pharmacol. 2012;674:143-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
25.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 50]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
26.  Roviello F, Marrelli D, Neri A, Cerretani D, de Manzoni G, Pedrazzani C, Cioppa T, Nastri G, Giorgi G, Pinto E. Treatment of peritoneal carcinomatosis by cytoreductive surgery and intraperitoneal hyperthermic chemoperfusion (IHCP): postoperative outcome and risk factors for morbidity. World J Surg. 2006;30:2033-240; discussion 2033-240;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 69]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
27.  Fujimoto S, Takahashi M, Mutou T, Kobayashi K, Toyosawa T. Successful intraperitoneal hyperthermic chemoperfusion for the prevention of postoperative peritoneal recurrence in patients with advanced gastric carcinoma. Cancer. 1999;85:529-534.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Di Vita M, Cappellani A, Piccolo G, Zanghì A, Cavallaro A, Bertola G, Bolognese A, Facchini G, D’Aniello C, Di Francia R. The role of HIPEC in the treatment of peritoneal carcinomatosis from gastric cancer: between lights and shadows. Anticancer Drugs. 2015;26:123-138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
29.  Desai AD, Hawksworth GM. Cryopreservation of rat hepatocytes with high attachment efficiency and mixed function oxidase activity post thawing. Biochem Soc Trans. 1990;18:1214.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Brücher BL, Piso P, Verwaal V, Esquivel J, Derraco M, Yonemura Y, Gonzalez-Moreno S, Pelz J, Königsrainer A, Ströhlein M. Peritoneal carcinomatosis: cytoreductive surgery and HIPEC--overview and basics. Cancer Invest. 2012;30:209-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 73]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
31.  Hildebrandt B, Wust P, Ahlers O, Dieing A, Sreenivasa G, Kerner T, Felix R, Riess H. The cellular and molecular basis of hyperthermia. Crit Rev Oncol Hematol. 2002;43:33-56.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Eppink B, Krawczyk PM, Stap J, Kanaar R. Hyperthermia-induced DNA repair deficiency suggests novel therapeutic anti-cancer strategies. Int J Hyperthermia. 2012;28:509-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 76]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
33.  Spratt JS, Adcock RA, Sherrill W, Travathen S. Hyperthermic peritoneal perfusion system in canines. Cancer Res. 1980;40:253-255.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Sugarbaker PH, Stuart OA, Yoo D. Strategies for management of the peritoneal surface component of cancer: cytoreductive surgery plus perioperative intraperitoneal chemotherapy. J Oncol Pharm Pract. 2005;11:111-119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
35.  Glehen O, Gilly FN, Arvieux C, Cotte E, Boutitie F, Mansvelt B, Bereder JM, Lorimier G, Quenet F, Elias D. Peritoneal carcinomatosis from gastric cancer: a multi-institutional study of 159 patients treated by cytoreductive surgery combined with perioperative intraperitoneal chemotherapy. Ann Surg Oncol. 2010;17:2370-2377.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 320]  [Cited by in F6Publishing: 303]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
36.  Bozzetti F, Yu W, Baratti D, Kusamura S, Deraco M. Locoregional treatment of peritoneal carcinomatosis from gastric cancer. J Surg Oncol. 2008;98:273-276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 74]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
37.  Graziosi L, Marino E, Donini A. Role of CRS plus HIPEC in gastric cancer peritoneal carcinomatosis. J Surg Oncol. 2015;111:248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
38.  Fujimoto S, Takahashi M, Kobayashi K, Kure M, Mutou T, Masaoka H, Ohkubo H. Relation between clinical and histologic outcome of intraperitoneal hyperthermic perfusion for patients with gastric cancer and peritoneal metastasis. Oncology. 1993;50:338-343.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Sha H, Zou Z, Xin K, Bian X, Cai X, Lu W, Chen J, Chen G, Huang L, Blair AM. Tumor-penetrating peptide fused EGFR single-domain antibody enhances cancer drug penetration into 3D multicellular spheroids and facilitates effective gastric cancer therapy. J Control Release. 2015;200:188-200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 75]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
40.  Glockzin G, Piso P. Current status and future directions in gastric cancer with peritoneal dissemination. Surg Oncol Clin N Am. 2012;21:625-633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 43]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
41.  Cabalag CS, Chan ST, Kaneko Y, Duong CP. A systematic review and meta-analysis of gastric cancer treatment in patients with positive peritoneal cytology. Gastric Cancer. 2015;18:11-22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
42.  Shitara K, Mizota A, Matsuo K, Sato Y, Kondo C, Takahari D, Ura T, Tajika M, Muro K. Fluoropyrimidine plus cisplatin for patients with advanced or recurrent gastric cancer with peritoneal metastasis. Gastric Cancer. 2013;16:48-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
43.  Oshima T, Yamada R, Hatori S, Kunisaki C, Imada T. Pharmacokinetics of S-1 in patients with peritoneal dissemination of gastric cancer. Oncol Rep. 2006;16:361-366.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Naitoh H, Kawaguch A, Yamamoto H, Mekata E, Tan T, Morii H, Chiba M. [Measurement of docetaxel concentration in blood and ascites after drip infusion into each vessel and intraperitoneal cavity of gastric cancer]. Gan To Kagaku Ryoho. 2004;31:2031-2034.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Sautner T, Hofbauer F, Depisch D, Schiessel R, Jakesz R. Adjuvant intraperitoneal cisplatin chemotherapy does not improve long-term survival after surgery for advanced gastric cancer. J Clin Oncol. 1994;12:970-974.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Morgan RJ, Doroshow JH, Synold T, Lim D, Shibata S, Margolin K, Schwarz R, Leong L, Somlo G, Twardowski P. Phase I trial of intraperitoneal docetaxel in the treatment of advanced malignancies primarily confined to the peritoneal cavity: dose-limiting toxicity and pharmacokinetics. Clin Cancer Res. 2003;9:5896-5901.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Tsujimoto H, Hagiwara A, Shimotsuma M, Sakakura C, Osaki K, Sasaki S, Ohyama T, Ohgaki M, Imanishi T, Yamazaki J. Role of milky spots as selective implantation sites for malignant cells in peritoneal dissemination in mice. J Cancer Res Clin Oncol. 1996;122:590-595.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Ishigami H, Kitayama J, Kaisaki S, Hidemura A, Kato M, Otani K, Kamei T, Soma D, Miyato H, Yamashita H. Phase II study of weekly intravenous and intraperitoneal paclitaxel combined with S-1 for advanced gastric cancer with peritoneal metastasis. Ann Oncol. 2010;21:67-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 168]  [Cited by in F6Publishing: 183]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
49.  Fushida S, Kinoshita J, Kaji M, Hirono Y, Goda F, Yagi Y, Oyama K, Sudo Y, Watanabe Y, Fujimura T. Phase I/II study of intraperitoneal docetaxel plus S-1 for the gastric cancer patients with peritoneal carcinomatosis. Cancer Chemother Pharmacol. 2013;71:1265-1272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
50.  Fujiwara Y, Takiguchi S, Nakajima K, Miyata H, Yamasaki M, Kurokawa Y, Mori M, Doki Y. Intraperitoneal docetaxel combined with S-1 for advanced gastric cancer with peritoneal dissemination. J Surg Oncol. 2012;105:38-42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 54]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
51.  Markman M. Intraperitoneal antineoplastic drug delivery: rationale and results. Lancet Oncol. 2003;4:277-283.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Peng YF, Imano M, Itoh T, Satoh T, Chiba Y, Imamoto H, Tsubaki M, Nishida S, Yasuda T, Furukawa H. A phase II trial of perioperative chemotherapy involving a single intraperitoneal administration of paclitaxel followed by sequential S-1 plus intravenous paclitaxel for serosa-positive gastric cancer. J Surg Oncol. 2015;111:1041-1046.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
53.  Turcotte S, Sideris L, Younan R, Drolet P, Dubé P. Pharmacokinetics of intraperitoneal irinotecan in a pig model. J Surg Oncol. 2010;101:637-642.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
54.  Chambers SK, Chow HH, Janicek MF, Cragun JM, Hatch KD, Cui H, Laughren C, Clouser MC, Cohen JL, Wright HM. Phase I trial of intraperitoneal pemetrexed, cisplatin, and paclitaxel in optimally debulked ovarian cancer. Clin Cancer Res. 2012;18:2668-2678.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 29]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
55.  Heiss MM, Murawa P, Koralewski P, Kutarska E, Kolesnik OO, Ivanchenko VV, Dudnichenko AS, Aleknaviciene B, Razbadauskas A, Gore M. The trifunctional antibody catumaxomab for the treatment of malignant ascites due to epithelial cancer: Results of a prospective randomized phase II/III trial. Int J Cancer. 2010;127:2209-2221.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 380]  [Cited by in F6Publishing: 352]  [Article Influence: 25.1]  [Reference Citation Analysis (0)]
56.  Ströhlein MA, Lordick F, Rüttinger D, Grützner KU, Schemanski OC, Jäger M, Lindhofer H, Hennig M, Jauch KW, Peschel C. Immunotherapy of peritoneal carcinomatosis with the antibody catumaxomab in colon, gastric, or pancreatic cancer: an open-label, multicenter, phase I/II trial. Onkologie. 2011;34:101-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 46]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
57.  Washington K. 7th edition of the AJCC cancer staging manual: stomach. Ann Surg Oncol. 2010;17:3077-3079.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 702]  [Cited by in F6Publishing: 801]  [Article Influence: 61.6]  [Reference Citation Analysis (0)]
58.  Matharu G, Tucker O, Alderson D. Systematic review of intraperitoneal chemotherapy for gastric cancer. Br J Surg. 2011;98:1225-1235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
59.  Yan TD, Black D, Sugarbaker PH, Zhu J, Yonemura Y, Petrou G, Morris DL. A systematic review and meta-analysis of the randomized controlled trials on adjuvant intraperitoneal chemotherapy for resectable gastric cancer. Ann Surg Oncol. 2007;14:2702-2713.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 260]  [Cited by in F6Publishing: 285]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
60.  Kojima N, Kunieda K, Matsui K, Kato H, Saji S. Evaluation of carcinoembryonic antigen mRNA in living, necrotic, and apoptotic gastric cancer cells by reverse transcriptase-polymerase chain reaction. Surg Today. 2003;33:839-846.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
61.  To EM, Chan WY, Chow C, Ng EK, Chung SC. Gastric cancer cell detection in peritoneal washing: cytology versus RT-PCR for CEA transcripts. Diagn Mol Pathol. 2003;12:88-95.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  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;110:291-297.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 46]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
63.  Bentrem D, Wilton A, Mazumdar M, Brennan M, Coit D. The value of peritoneal cytology as a preoperative predictor in patients with gastric carcinoma undergoing a curative resection. Ann Surg Oncol. 2005;12:347-353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 172]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
64.  Kodera Y, Nakanishi H, Ito S, Mochizuki Y, Ohashi N, Yamamura Y, Fujiwara M, Koike M, Tatematsu M, Nakao A. Prognostic significance of intraperitoneal cancer cells in gastric carcinoma: analysis of real time reverse transcriptase-polymerase chain reaction after 5 years of followup. J Am Coll Surg. 2006;202:231-236.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 37]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
65.  Kang KK, Hur H, Byun CS, Kim YB, Han SU, Cho YK. Conventional cytology is not beneficial for predicting peritoneal recurrence after curative surgery for gastric cancer: results of a prospective clinical study. J Gastric Cancer. 2014;14:23-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
66.  Yoo C, Ryu MH, Park YS, Yoo MW, Park SR, Ryoo BY, Jang SJ, Yook JH, Kim BS, Kang YK. Intraoperatively assessed macroscopic serosal changes in patients with curatively resected advanced gastric cancer: clinical implications for prognosis and peritoneal recurrence. Ann Surg Oncol. 2015;22:2940-2947.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
67.  Fujiwara Y, Doki Y, Taniguchi H, Sohma I, Takiguchi S, Miyata H, Yamasaki M, Monden M. Genetic detection of free cancer cells in the peritoneal cavity of the patient with gastric cancer: present status and future perspectives. Gastric Cancer. 2007;10:197-204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 52]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
68.  Yabusaki N, Yamada S, Murai T, Kanda M, Kobayashi D, Tanaka C, Fujii T, Nakayama G, Sugimoto H, Koike M. Clinical significance of zinc-finger E-box binding homeobox 1 mRNA levels in peritoneal washing for gastric cancer. Mol Clin Oncol. 2015;3:435-441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
69.  Jeon CH, Kim IH, Chae HD. Prognostic value of genetic detection using CEA and MAGE in peritoneal washes with gastric carcinoma after curative resection: result of a 3-year follow-up. Medicine (Baltimore). 2014;93:e83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
70.  Kowalewska M, Chechlinska M, Nowak R. Carcinoembryonic antigen and cytokeratin 20 in peritoneal cells of cancer patients: are we aware of what we are detecting by mRNA examination? Br J Cancer. 2008;98:512-513; author reply 514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
71.  Yu JL, Lv P, Han J, Zhu X, Hong LL, Zhu WY, Wang XB, Wu YC, Li P, Ling ZQ. Methylated TIMP-3 DNA in body fluids is an independent prognostic factor for gastric cancer. Arch Pathol Lab Med. 2014;138:1466-1473.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
72.  Kitayama J, Emoto S, Yamaguchi H, Ishigami H, Onoyama H, Yamashita H, Seto Y, Matsuzaki K, Watanabe T. Flow Cytometric Quantification of Intraperitoneal Free Tumor Cells is a Useful Biomarker in Gastric Cancer Patients with Peritoneal Metastasis. Ann Surg Oncol. 2015;22:2336-2342.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 18]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]