Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Sep 7, 2014; 20(33): 11552-11559
Published online Sep 7, 2014. doi: 10.3748/wjg.v20.i33.11552
Metachronous gastric cancer after successful Helicobacter pylori eradication
Akiko Shiotani, Ken Haruma, Department of Internal Medicine, Kawasaki Medical School, Kurashiki 701-0192, Japan
David Y Graham, Department of Medicine, Michael E DeBakey VAMC and Baylor College of Medicine, Houston, TX 77030, United States
Author contributions: Shiotani A mainly wrote the manuscript; Haruma K and Graham DY edited the manuscript.
Correspondence to: Akiko Shiotani, MD, PhD, Department of Internal Medicine, Kawasaki Medical School, 577 Matsushima, Kurashiki 701-0192, Japan. shiotani@med.kawasaki-m.ac.jp
Telephone: +81-86-4621111 Fax: +81-86-4641195
Received: October 25, 2013
Revised: December 30, 2013
Accepted: May 28, 2014
Published online: September 7, 2014

Abstract

The high incidence of gastric cancer in Japan initially resulted in establishment of a country-wide gastric cancer screening program to detect early and treatable cancers. In 2013 countrywide Helicobacter pylori (H. pylori) eradication was approved coupled with endoscopy to assess for the presence of chronic gastritis. Current data support the notion that cure of the infection in those with non-atrophic gastritis will prevent development of gastric cancer. However, while progression to more severe damage is halted in those who have already developed, atrophic gastritis/gastric atrophy remain at risk for subsequent development of gastric cancer. That risk is directly related to the extent and severity of atrophic gastritis. Methods to stratify cancer risk include those based on endoscopic assessment of the atrophic border, histologic grading, and non-invasive methods based on serologic testing of pepsinogen levels. Continued surveillance is required because those with atrophic gastritis/gastric atrophy retain considerable gastric cancer risk even after H. pylori eradication. Those who have already experienced a resectable early gastric cancer are among those at highest risk as metachronous lesions are frequent even after H. pylori eradication. We review the role of H. pylori and effect of H. pylori eradication indicating the incidence and the predictive factors on development of metachronous cancer after endoscopic therapy of early gastric cancer. Studies to refine risk markers to stratify for risk, surveillance methods, intervals, and duration after successful H. pylori eradication, and whether adjuvant therapy would change risk are needed.

Key Words: Atrophic gastritis, Pepsinogen, miRNA, Intestinal metaplasia, Cancer prevention

Core tip: For the patients with a history of endoscopic resection of early gastric cancer, Helicobacter pylori (H. pylori) eradication followed by continued surveillance for gastric cancer is generally required because those with severe gastric atrophy retain considerable gastric cancer risk even after H. pylori eradication. We review the role of H. pylori and effect of H. pylori eradication indicating the incidence and the predictive factors on development of metachronous cancer after endoscopic therapy of early gastric cancer.



INTRODUCTION

Gastric cancer is the fourth most common cancer and second leading cause of cancer deaths worldwide with more than 700000 deaths annually[1]. There are marked geographic differences in gastric cancer incidence both within and between countries and regions with the highest incidence occurring in Japan, Korea, China, Eastern Europe and parts of Central and South America[2]. An attempt to reduce gastric cancer mortality led Japan in the 1950’s to establish secondary cancer prevention programs to detect malignant lesions in an early and potentially treatable stage. Improved detection of early lesions using double contrast roentgenography of the stomach, gastrocamera and most recently endoscopic examination with biopsy of suspected lesions has resulted in improved survival of patients receiving operative therapy[3].

Endoscopic methods to remove premalignant as well as superficial malignant gastric lesions such as endoscopic mucosal resection and endoscopic submucosal dissection have now become the standard of care in Japan and Korea for management of early gastric cancers with no evidence of lymph node metastasis. This program has been highly successful and currently more than half of Japanese gastric cancer cases are diagnosed at an early stage[4]. Because endoscopic submucosal dissection involves removal of both the mucosa and submucosa, large lesions can be resected en bloc yielding an improved histopathological diagnosis compared to endoscopic mucosal resection. However, both methods result in greater post-interventional quality of life compared to surgical resection[5].

While endoscopic removal of an early gastric cancer only solves the problem of that particular lesion it does not affect the overall cancer risk. The stomach in patients with early gastric cancer typically exhibits extensive chronic atrophic changes with multiple areas with preneoplastic changes and often contains microscopic foci of intramucosal cancer[6,7]. Prior studies that examined stomachs of patients with early gastric cancer reported that detailed histologic examination will reveal foci of intramucosal cancer in up to 15%[8-10]. The presence of these lesion is likely at least partially responsible for the fact that the risk of developing a metachronous lesion following endoscopic removal of an early gastric cancer has ranged between 1% and 4% per year[8,11-13]. Because of the high risk of metachronous lesions it is recommended that these patients are enrolled in a life-long endoscopic surveillance program.

The factors that influence the rate of appearance of metachronous lesions remain unclear, however some studies have shown that Helicobacter pylori (H. pylori) eradication results in a lower risk of developing a metachronous lesion[14]. Based on prior studies and the cancer field effect, it should not be surprising that either metachronous lesion remains a risk. For example, H. pylori eradication in patients with H. pylori infection and atrophic gastritis but no evidence of cancer has been shown to reduce but not eliminate that risk suggesting a role for the organism itself, for continuing H. pylori-induced inflammation, or both in relation of cancer risk.

H. pylori and gastric cancer

In 1994, the International Agency for Research on Cancer (IARC) of the World Health Organization classified H. pylori as a definite carcinogen[15]. Two decades then passed before this knowledge was translated into the decision to approve population-wide H. pylori eradication for any population[7]. During this interval a number of misconceptions regarding the attributable risk of H. pylori infection in gastric cancer were corrected and the role of atrophic gastritis as a surrogate for cancer risk was confirmed[16]. It is now accepted that H. pylori infection is responsible for more than 95% of gastric cancers (e.g., one study in Japan demonstrated that H. pylori-negative gastric cancer accounted for less than 3% among all gastric cancers)[17]. H. pylori infection causes progressive damage to the stomach that may eventually result in atrophic gastritis/gastric atrophy with a rapidly increasing risk of gastric cancer. It is this progressive nature of the process that makes it so dangerous and many have been lulled into compliancy when deciding what to do with a patient with mild or non-atrophic gastritis without recognizing that the current histology is actually an early stage of a progressive process and the subsequent changes are largely irreversible. However, progression can be prevented or halted by H. pylori eradication but the cancer risk associated with atrophic damage can at best be only partially reversed. As such, H. pylori infection has been likened to infestation with termites which also cause typically silent put progressive damage. As with termites, the best results are obtained when the problem is discovered before permanent and extensive damage has occurred. The failure to recognize the progressive nature of the process can result in compliancy during which an individual cancer risk progressively increases[18] .

H. pylori induced gastritis is typically acquired in childhood. Initially the inflammation and damage is most severe in the non-acid secreting gastric antrum. Over time the damage progresses into the gastric corpus as an advancing atrophic border which can be recognized endoscopically, and the damage clinically progresses more rapidly along the lesser curve than the greater curve[19,20]. Chronic inflammation related with H. pylori affects differentiation and promotes metaplasia[21-23]. As the damage advances into the corpus along the atrophic border it leaves behind a lawn of pyloric metaplasia (also known as pseudopyloric or mucus metaplasia) now recognized to be similar or identical to spasmolytic polypeptide/trefoil factor family 2 (TFF2)-expressing metaplasia (SPEM) described in animal models of gastric cancer[24,25]. The recognition that pyloric metaplasia could be easily recognized by immunostaining as SPEM rather than the previous cumbersome process of identifying it on the basis of corpus location and pepsinogen I staining allowed many older observations to be rapidly confirmed and extended[16]. It is now believed that intestinal metaplasia arises from SPEM and SPEM may also provide the cell of origin of gastric cancer[25,26]. Intestinal metaplasia is no longer thought to be the precursor of gastric cancer but rather is an easy recognized surrogate for the presence and extent of gastric mucosal atrophy[27-29]. The concept of multifocal atrophic gastritis actually represents scattered areas of intestinal metaplasia arising within a lawn of SPEM-type atrophy damage[30-32]. The ease of diagnosing pyloric metaplasia has allowed development of a new gastric cancer risk stratification system, the corpus-predominant gastritis index, to join the Operative Link for Gastritis Assessment (OLGA) and OLGA-M histology systems of stratifying gastric cancer risk[33].

SERUM MARKERS FOR GASTRIC CANCER RISK ASSESSMENT
Pepsinogen

For population wide testing it is important have efficient and cost effective practical mass screening methods that correlate with the risk of developing gastric cancer (i.e., non-invasive risk stratification). Since H. pylori infection is the necessary but insufficient cause of gastric cancer, identification and eradication of H. pylori is the most important step; eradication H. pylori infections which will ultimately eliminate gastric cancer. There are a number of validated non-invasive methods to identify H. pylori infection ranging from serologic methods, through the urea breath test and stool H. pylori antigen testing[34]. However in populations where gastric cancer is common, H. pylori eradication alone is often insufficient as many individuals will have already have experienced irreversible gastric damage and thus carry an ongoing risk for development of gastric cancer despite H. pylori eradication. In the past when the emphasis was on identifying incidence cases of gastric cancer in high prevalence countries such as Japan, a number of approaches (secondary cancer prevention) were tested. Measurement of serum pepsinogens proved to be a useful non-invasive method of identification of patients at risk and also proved cost effective for enriching the population with gastric cancer in screening studies[35-37]. The concept is based on the fact that pepsinogen I is produced by the chief and mucous neck cells in the fundic glands whereas and pepsinogen II is produced throughout the stomach as well as by Brunner’s glands[38,39]. Damage to the gastric corpus results in a progressive decline in both pepsinogen I levels and the ratio of pepsinogen I to pepsinogen II (pepsinogen I/II). Pepsinogen testing thus allows a non-invasive assessment of the presence and extent of atrophic gastritis and can be used to risk identify patients endoscopic cancer screening programs or for possibly needing endoscopic surveillance after H. pylori eradication[6,29,35,37,40-42]. While this approach has been shown to be useful, the cumulative data have shown some limitations. Probably the most important limitation is that H. pylori eradication can significantly change pepsinogen levels with a decrease of pepsinogen I and pepsinogen II and an increase of pepsinogen I/II even among those at high risk for gastric cancer[43,44]. Thus, at least as currently used, serum pepsinogen testing cannot be used as a reliable marker of atrophy for patients who already have been treated by eradication therapy.

Japan has a large cadre of endoscopists experienced in detection of atrophic gastritis and early gastric cancer and the decision was made that H. pylori eradication therapy should be accompanied by endoscopy to examine the extent and severity of gastritis. In countries where gastric cancer risk is lower and a large number of experienced endoscopists is lacking, it would probably be more prudent and cost effective to use pre-therapy pepsinogen testing to risk stratify patients into those possibly at higher risk for subsequent gastric cancer and those with little or risk post H. pylori eradication. Those in the higher risk category could then undergo endoscopy using a validated risk stratification system to identify those with indications for continued surveillance.

Micro-RNA

Micro-RNAs (miRNAs) are 18-25 nucleotide noncoding RNA sequences that are transcribed but not translated into proteins. Some miRNAs have been shown to possess oncogenic or tumor suppressor activity and relate to apoptosis, proliferation, differentiation, metastasis, angiogenesis, and immune response, which are all potentially involved in cancer initiation, progression and treatment response[45,46]. MiRNAs can also be detected circulating in a cell-free form in blood, most probably in exosomes which protect them against degradation by ribonuclease, and their signatures in blood are similar in men and women, as well as individuals of different age[47,48]. Furthermore, miRNA levels are similar in plasma and serum, and freeze/thaw as well as prolonged storage at room temperature does not affect their levels[48]. Thus, serum miRNAs have the potential of a novel biomarker for many cancers. Lawrie et al[49] first discovered tumor-specific deregulation of circulating miRNAs and subsequently, circulating miRNAs have been suggested great potential as biomarkers for many cancers including gastric cancer[48,50,51]. Moreover, accumulating reports suggest the potential of miRNAs in the early detection of gastric cancer.

We investigated serum miRNAs as markers to individuals at high risk for gastric cancer not only before H. pylori eradication but also after eradication. The serum levels of miR-106b and let-7d before and after H. pylori eradication; miR-21 after eradication were significantly higher in the high-risk group than in controls. H. pylori eradication significantly changed serum pepsinogen levels even in the high-risk group, whereas eradication did not significantly alter miR-106b and let-7 levels in the high-risk group. These results suggest that serum miRNAs may be equivalent or even superior to serum pepsinogen as a biomarker to detect those at high risk for gastric cancer before and after H. pylori eradication[52].

EFFECT OF H. PYLORI ERADICATION ON CANCER INCIDENCE

In the first half of the 20th century it was recognized that gastric cancer risk was related to atrophic gastritis[7,53]. The late 20th century brought new information and identified that gastric cancer was an inflammation-related cancer caused by chronic infection with H. pylori. It was initially unclear whether H. pylori eradication alone would suffice to eliminate or greatly reduce gastric cancer risk or whether some form of surveillance would be still required. The fact that those with atrophic gastritis whose H. pylori had disappeared spontaneously following destruction of the normal gastric niche for their growth still retained a high risk of gastric cancer suggested that H. pylori eradication alone was likely to prove insufficient[6]. Many clinical studies have subsequently examined the effect of H. pylori eradication on the subsequent incidence of gastric cancer. For example, Take et al[54] in a prospective non-randomized eradication study among more than1100 Japanese patients with peptic ulcers showed that H. pylori eradication reduced the risk of subsequently developing gastric cancer. A follow-up for a mean of 3.9 years of these patients found that gastric cancer developed in less frequently among those who had had successful H. pylori eradication compared to those with persistent infection (0.23% vs 0.70% at 1 year, P = 0.04, log-rank test)[54]. While eradication did not completely eliminate the risk, and the risk was related to the extent of atrophic gastritis at the time of eradication therapy[54]. The remained risk of developing gastric cancer was reported to be 0.30% per year[55]. The Shangdong intervention trial failed to find a difference in gastric cancer incidence after 7.3 years but did find a significant fall 14.7 years post H. pylori eradication therapy[7,56]. The latest meta-analysis has confirmed that successful eradication reduced the risk for gastric cancer and included 6 randomized controlled trials including four from China, one from Japan, and one from Colombia. The median follow-up period was 6 years. The pooled analysis yielded a relative risk for gastric cancer of 0.65 (95%CI: 0.43-0.89) following successful eradication therapy[57].

One effect of H. pylori eradication therapy is to stop the progression of damage and thus lock in or reduce the gastric cancer risk present at the time of H. pylori eradication[6]. Thus, those with non-atrophic gastritis would be expected to have negligible risk of subsequently developing gastric cancer whereas those with atrophic gastritis would be expected to have a risk equal to or somewhat lower than others with the same pattern of gastritis but definitely lower than an untreated cohort whose risk would increase yearly as the disease progressed[6]. The available data confirm these expectations[6,41]. However, there are few studies that have followed patients who were matched based on risk stratification. A longitudinal cohort study of 9.3 years in Japan reported significant reduction in cancer incidence after eradication in H. pylori positive patients with mild atrophic gastritis as evaluated by serum pepsinogen testing. The incidence per 100000 person-year in those with persistent infection was 111 compared to 69 among those the infection was eradiated. However the cancer incidence was not significantly different (237 vs 223) among the patients with more severe atrophy[58].

Eradication of the infection stops the inflammatory process, promotes healing of gastritis and resolution of inflammation. Nonetheless, the link between H. pylori and cancer runs through atrophic gastritis and intestinal metaplasia, and eradication cannot reverse the severe atrophic damage and intestinal metaplasia, especially incomplete type or SPEM that has already occurred. For examples, H. pylori eradication prior to development of intestinal metaplasia improves corpus gastritis enhancing sonic hedgehog (SHH) and its downstream regulators and diminishing SHH methylation and aberrant CDX2 expression, which inhibit intestinal development and differentiation and reverse gastric phenotype. However, eradication in patients with high risk such as atrophy with intestinal metaplasia, especially incomplete type or a history of endoscopic treatment for gastric cancer does not result in much if any improvement[22,59,60]. The ability to predict the point of no return for the development of the malignancy is of particular interest and whether the presence of severe atrophy, SPEM, or some intestinal metaplasia subtypes alone or together correspond to this point, still need to be investigated[61].

CUMULATIVE INCIDENCE OF METACHRONOUS CANCER AFTER ERADICATION

The group with the highest risk of gastric cancer includes those who have already had one cancer cured by upper gastrointestinal endoscopy. Several studies have reported incidence of metachronous cancer after successful H. pylori eradication (Table 1).

Table 1 Incidence of metachronous cancer after successful Helicobacter pylori eradication.
Ref.CountrySubject No.Study designMean follow-up periodsIncidence (%)Eradication effect(95%CI)
Uemura et al[62], 1997Japan65/67NR3 yr0 vs 9effective P = 0.011
Fukase et al[14], 2008Japan272/272Multicenter open-label RCT3 yr3.3 vs 8.8effective 0.35 (0.16-0.78) P = 0.009
Shiotani et al[12], 2008Japan80single arm33 mo11.3
Hanaoka et al[66], 2010Japan8255 mo14.6
Maehata et al[63], 2012Japan177/91retro NR3 yr 1.1-11.1 yr8.5 vs 14.3OR = 1.71 (0.72-4.03)
Kato et al[64], 2013Japan263/105Multicenter retro cohort26.8 mo 2-5 yr3.5/YNS
Seo et al[65], 2013South Korea61/13retro cohort27.2 mo9.8 vs 23.1OR = 0.36 (0.08-1.70),
Chon et al[69], 2013South Korea129 85/44Retro NR26 mo 16.5-30 mo4.7 vs 11.4effective HR = 0.143 P = 0.008

Uemura et al[62] were the first to show that H. pylori eradication could reduce the risk of development of gastric cancer in this group of patients when, in a retrospective study, 132 patients with early gastric cancer were followed after endoscopic resection; metachronous gastric cancer developed only in 6 of 67 (9%) patients without eradication over a follow up of 3 years. A later multi-center prospective randomized study in Japanese patients followed for 3 years after endoscopic removal of an early gastric cancer found metachronous gastric cancer in 9 of 272 (3.3%) patients with H. pylori eradication vs 24 of 272 (8.8%) controls[14]. The incidence of metachronous gastric cancer was reduced significantly (OR = 0.35, 95%CI: 0.16-0.78; P = 0.009) consistent with H. pylori eradication having a benefit in delaying the onset of new cancers in the same stomach.

In contrast, a Japanese retrospective study reported that H. pylori eradication did not reduce the incidence of metachronous gastric cancer. Baseline severe mucosal atrophy and a follow-up of more than 5 years were found to be independent risk factors for the development of metachronous gastric cancer[63]. Moreover, a recent Japanese multicenter retrospective cohort study from 12 hospitals detected metachronous multiple cancers in 65 of 1258 (5.2%) during a mean of 26.8 mo. The cumulative incidence of metachronous cancers increased linearly and the mean annual incidence rate was 3.5%. The incidence rate did not differ between patients with or without H. pylori eradication[64]. A recent study from Korea also reported that metachronous gastric cancer showed a decrease in the eradicated group, but this did not reach statistical significance (OR = 0.36, 95%CI: 0.08-1.70, P = 0.189), although metachronous gastric cancer was significantly decreased in the eradicated group (OR = 0.108, 95%CI: 0.016-0.726, P = 0.035) among the subgroup who were followed-up for more than 18 mo[65].

These recent studies have all confirmed that the presence of one early gastric cancer identifies a group of patient at extremely high risk of metachronous cancer consistent with the histologic analysis of the remaining gastric mucosa in patients with early gastric cancer undergoing gastric resection. The risk of a metachronous cancer among those not having H. pylori eradication appears to be in the range of 3%-4% per year (e.g., 3000 to 4000/100000 per year) (Table 1). Overall H. pylori eradication appears to reduce that risk but this is not seen in all studies and studies are needed to identify risk factors that correlate with subsequent risk such as pattern and extent of atrophy as well as better characterization of the mucosa in terms of inflammation, presence and extent of SPEM, different types of intestinal metaplasia, gastric microbiota, etc..

PREDICTIVE FACTORS FOR METACHRONOUS GASTRIC CANCER AFTER H. PYLORI ERADICATION

Previous studies have looked at factors that helped predict development of gastric cancer after H. pylori eradication and those data might provide clues to risk stratification after endoscopic removal of an early gastric cancer and H. pylori eradication. H. pylori eradication will thus produce two populations: those with minimal to no risk and those with some residual risk for cancer. Those with residual risk can likely be assured that their risk will not increase as they age and although it will probably decrease somewhat, some risk remains[7]. Our previous study showed that atrophy in biopsy specimens from the lesser curvature of the corpus was strongly associated with gastric cancer risk[12]. The frequency of severe atrophy assessed by histology (100% vs 53.2%, P = 0.03) was higher and the serum pepsinogen I/II ratio before H. pylori eradication was significantly lower in the group that developed metachronous cancer compared to the group that did not. A pepsinogen I of < 25 ng/mL before eradication was significantly associated with development of a new lesion[12]. Moreover, extensive atrophic gastritis diagnosed by autofluorescence imaging (AFI), which is new endoscopic imaging technology using illumination of different wavelength light through a filter in a light source[21], was a significant predictor for metachronous cancer developed after successful eradication and could possibly be useful to identify patients undergoing endoscopic submucosal dissection who still required intensive surveillance after eradication[66].

For many individuals H. pylori eradication equates with cancer prevention whereas for others it only produces a reduction in risk. This difference in outcome depends on the level of risk when the eradication is performed. For those with history of endoscopic resection of one cancer, H. pylori eradication followed by surveillance for gastric cancer is generally indicated (i.e., a combination of primary and secondary prevention), because the risk of gastric cancer remains high even after H. pylori eradication[6,53]. The previous study indicated that levels of hMLH1 promoter hypermethylation, which is a frequent cause of the microsatellite instability (MSI) -H phenotype, are similar in the surrounding non-cancerous tissue compared to cancer tissue[67]. In addition, another study indicated that MSI and hypermethylation of hMLH1 in cancer lesions were detected more frequently in the patients with multiple gastric cancers than those with solitary gastric cancer[68]. These results indicate that inactivation of hMLH1 through promoter hypermethylation seems to be involved in the development of multiple gastric cancers following the MSI pathway. MSI or hypermethylation of hMLH1 as well as serum miRNA might be potential predictive markers for metachronous cancer. Studies are required to establish appropriate markers irrespective H. pylori eradication for gastric cancer screening.

Footnotes

P- Reviewer: Mizoshita T, Smolka AJ S- Editor: Ma YJ L- Editor: A E- Editor: Wang CH

References
1.  Melton SD, Genta RM, Souza RF. Biomarkers and molecular diagnosis of gastrointestinal and pancreatic neoplasms. Nat Rev Gastroenterol Hepatol. 2010;7:620-628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
2.  Correa P, Houghton J. Carcinogenesis of Helicobacter pylori. Gastroenterology. 2007;133:659-672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 467]  [Cited by in F6Publishing: 478]  [Article Influence: 28.1]  [Reference Citation Analysis (0)]
3.  Fukao A, Tsubono Y, Tsuji I, HIsamichi S, Sugahara N, Takano A. The evaluation of screening for gastric cancer in Miyagi Prefecture, Japan: a population-based case-control study. Int J Cancer. 1995;60:45-48.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Gotoda T, Yamamoto H, Soetikno RM. Endoscopic submucosal dissection of early gastric cancer. J Gastroenterol. 2006;41:929-942.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 485]  [Cited by in F6Publishing: 489]  [Article Influence: 27.2]  [Reference Citation Analysis (0)]
5.  Ma CJ, Sun LC, Chen FM, Lu CY, Shih YL, Tsai HL, Chuang JF, Wang JY. A double-blind randomized study comparing the efficacy and safety of a composite vs a conventional intravenous fat emulsion in postsurgical gastrointestinal tumor patients. Nutr Clin Pract. 2012;27:410-415.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 28]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
6.  Graham DY, Shiotani A. The time to eradicate gastric cancer is now. Gut. 2005;54:735-738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 90]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
7.  Shiotani A, Cen P, Graham DY. Eradication of gastric cancer is now both possible and practical. Semin Cancer Biol. 2013;23:492-501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 65]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
8.  Nasu J, Doi T, Endo H, Nishina T, Hirasaki S, Hyodo I. Characteristics of metachronous multiple early gastric cancers after endoscopic mucosal resection. Endoscopy. 2005;37:990-993.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 124]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
9.  Takeda J, Toyonaga A, Koufuji K, Kodama I, Aoyagi K, Ohta J, Aoyama Y, Hata H. Resected early gastric cancer--clinicopathological studies on 610 cases. Kurume Med J. 1995;42:87-94.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Miyoshi E, Haruma K, Hiyama T, Tanaka S, Yoshihara M, Shimamoto F, Chayama K. Microsatellite instability is a genetic marker for the development of multiple gastric cancers. Int J Cancer. 2001;95:350-353.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Uedo N, Fujishiro M, Goda K, Hirasawa D, Kawahara Y, Lee JH, Miyahara R, Morita Y, Singh R, Takeuchi M. Role of narrow band imaging for diagnosis of early-stage esophagogastric cancer: current consensus of experienced endoscopists in Asia-Pacific region. Dig Endosc. 2011;23 Suppl 1:58-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 69]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
12.  Shiotani A, Uedo N, Iishi H, Yoshiyuki Y, Ishii M, Manabe N, Kamada T, Kusunoki H, Hata J, Haruma K. Predictive factors for metachronous gastric cancer in high-risk patients after successful Helicobacter pylori eradication. Digestion. 2008;78:113-119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 54]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
13.  Isomoto H, Shikuwa S, Yamaguchi N, Fukuda E, Ikeda K, Nishiyama H, Ohnita K, Mizuta Y, Shiozawa J, Kohno S. Endoscopic submucosal dissection for early gastric cancer: a large-scale feasibility study. Gut. 2009;58:331-336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 470]  [Cited by in F6Publishing: 496]  [Article Influence: 33.1]  [Reference Citation Analysis (1)]
14.  Fukase K, Kato M, Kikuchi S, Inoue K, Uemura N, Okamoto S, Terao S, Amagai K, Hayashi S, Asaka M. Effect of eradication of Helicobacter pylori on incidence of metachronous gastric carcinoma after endoscopic resection of early gastric cancer: an open-label, randomised controlled trial. Lancet. 2008;372:392-397.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 876]  [Cited by in F6Publishing: 850]  [Article Influence: 53.1]  [Reference Citation Analysis (0)]
15.  Nyrén O. Is Helicobacter pylori really the cause of gastric cancer? Semin Cancer Biol. 1998;8:275-283.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  El-Zimaity HM, Ota H, Graham DY, Akamatsu T, Katsuyama T. Patterns of gastric atrophy in intestinal type gastric carcinoma. Cancer. 2002;94:1428-1436.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Kato S, Matsukura N, Tsukada K, Matsuda N, Mizoshita T, Tsukamoto T, Tatematsu M, Sugisaki Y, Naito Z, Tajiri T. Helicobacter pylori infection-negative gastric cancer in Japanese hospital patients: incidence and pathological characteristics. Cancer Sci. 2007;98:790-794.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 76]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
18.  Nakayama Y, Graham DY. Helicobacter pylori infection: diagnosis and treatment. Expert Rev Anti Infect Ther. 2004;2:599-610.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Satoh K, Kimura K, Taniguchi Y, Yoshida Y, Kihira K, Takimoto T, Kawata H, Saifuku K, Ido K, Takemoto T. Distribution of inflammation and atrophy in the stomach of Helicobacter pylori-positive and -negative patients with chronic gastritis. Am J Gastroenterol. 1996;91:963-969.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Kimura K. Chronological transition of the fundic-pyloric border determined by stepwise biopsy of the lesser and greater curvatures of the stomach. Gastroenterology. 1972;63:584-592.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Shiotani A, Iishi H, Uedo N, Ishiguro S, Tatsuta M, Nakae Y, Kumamoto M, Merchant JL. Evidence that loss of sonic hedgehog is an indicator of Helicobater pylori-induced atrophic gastritis progressing to gastric cancer. Am J Gastroenterol. 2005;100:581-587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 73]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
22.  Shiotani A, Uedo N, Iishi H, Tatsuta M, Ishiguro S, Nakae Y, Kamada T, Haruma K, Merchant JL. Re-expression of sonic hedgehog and reduction of CDX2 after Helicobacter pylori eradication prior to incomplete intestinal metaplasia. Int J Cancer. 2007;121:1182-1189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
23.  Silberg DG, Sullivan J, Kang E, Swain GP, Moffett J, Sund NJ, Sackett SD, Kaestner KH. Cdx2 ectopic expression induces gastric intestinal metaplasia in transgenic mice. Gastroenterology. 2002;122:689-696.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Nomura S, Baxter T, Yamaguchi H, Leys C, Vartapetian AB, Fox JG, Lee JR, Wang TC, Goldenring JR. Spasmolytic polypeptide expressing metaplasia to preneoplasia in H. felis-infected mice. Gastroenterology. 2004;127:582-594.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Nozaki K, Ogawa M, Williams JA, Lafleur BJ, Ng V, Drapkin RI, Mills JC, Konieczny SF, Nomura S, Goldenring JR. A molecular signature of gastric metaplasia arising in response to acute parietal cell loss. Gastroenterology. 2008;134:511-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 134]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
26.  Weis VG, Goldenring JR. Current understanding of SPEM and its standing in the preneoplastic process. Gastric Cancer. 2009;12:189-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 101]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
27.  Houghton J, Wang TC. Helicobacter pylori and gastric cancer: a new paradigm for inflammation-associated epithelial cancers. Gastroenterology. 2005;128:1567-1578.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Shiotani A, Iishi H, Uedo N, Kumamoto M, Nakae Y, Ishiguro S, Tatsuta M, Graham DY. Histologic and serum risk markers for noncardia early gastric cancer. Int J Cancer. 2005;115:463-469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 62]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
29.  Varon C, Dubus P, Mazurier F, Asencio C, Chambonnier L, Ferrand J, Giese A, Senant-Dugot N, Carlotti M, Mégraud F. Helicobacter pylori infection recruits bone marrow-derived cells that participate in gastric preneoplasia in mice. Gastroenterology. 2012;142:281-291.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 100]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
30.  Weis VG, Sousa JF, LaFleur BJ, Nam KT, Weis JA, Finke PE, Ameen NA, Fox JG, Goldenring JR. Heterogeneity in mouse spasmolytic polypeptide-expressing metaplasia lineages identifies markers of metaplastic progression. Gut. 2013;62:1270-1279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 87]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
31.  Nam KT, O’Neal RL, Coffey RJ, Finke PE, Barker N, Goldenring JR. Spasmolytic polypeptide-expressing metaplasia (SPEM) in the gastric oxyntic mucosa does not arise from Lgr5-expressing cells. Gut. 2012;61:1678-1685.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
32.  Goldenring JR, Nam KT. Oxyntic atrophy, metaplasia, and gastric cancer. Prog Mol Biol Transl Sci. 2010;96:117-131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 22]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
33.  Tsai YC, Hsiao WH, Yang HB, Cheng HC, Chang WL, Lu CC, Sheu BS. The corpus-predominant gastritis index may serve as an early marker of Helicobacter pylori-infected patients at risk of gastric cancer. Aliment Pharmacol Ther. 2013;37:969-978.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 28]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
34.  Malfertheiner P, Megraud F, O’Morain CA, Atherton J, Axon AT, Bazzoli F, Gensini GF, Gisbert JP, Graham DY, Rokkas T. Management of Helicobacter pylori infection--the Maastricht IV/ Florence Consensus Report. Gut. 2012;61:646-664.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1541]  [Cited by in F6Publishing: 1494]  [Article Influence: 124.5]  [Reference Citation Analysis (3)]
35.  Kitahara F, Kobayashi K, Sato T, Kojima Y, Araki T, Fujino MA. Accuracy of screening for gastric cancer using serum pepsinogen concentrations. Gut. 1999;44:693-697.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Kudo T, Kakizaki S, Sohara N, Onozato Y, Okamura S, Inui Y, Mori M. Analysis of ABC (D) stratification for screening patients with gastric cancer. World J Gastroenterol. 2011;17:4793-4798.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 38]  [Cited by in F6Publishing: 32]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
37.  Miki K. Gastric cancer screening by combined assay for serum anti-Helicobacter pylori IgG antibody and serum pepsinogen levels - “ABC method”. Proc Jpn Acad Ser B Phys Biol Sci. 2011;87:405-414.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Samloff IM. Pepsinogens, pepsins, and pepsin inhibitors. Gastroenterology. 1971;60:586-604.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Samloff IM. Cellular localization of group I pepsinogens in human gastric mucosa by immunofluorescence. Gastroenterology. 1971;61:185-188.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Miki K. Gastric cancer screening using the serum pepsinogen test method. Gastric Cancer. 2006;9:245-253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 181]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
41.  Ohata H, Kitauchi S, Yoshimura N, Mugitani K, Iwane M, Nakamura H, Yoshikawa A, Yanaoka K, Arii K, Tamai H. Progression of chronic atrophic gastritis associated with Helicobacter pylori infection increases risk of gastric cancer. Int J Cancer. 2004;109:138-143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 354]  [Cited by in F6Publishing: 358]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
42.  Watabe H, Mitsushima T, Yamaji Y, Okamoto M, Wada R, Kokubo T, Doi H, Yoshida H, Kawabe T, Omata M. Predicting the development of gastric cancer from combining Helicobacter pylori antibodies and serum pepsinogen status: a prospective endoscopic cohort study. Gut. 2005;54:764-768.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 290]  [Cited by in F6Publishing: 312]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
43.  Kawai T, Miki K, Ichinose M, Kenji Y, Miyazaki I, Kawakami K, Kataoka M, Yamagishi T, Sofuni A, Itoi T. Changes in evaluation of the pepsinogen test result following Helicobacter pylori eradication therapy in Japan. Inflammopharmacology. 2007;15:31-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 16]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
44.  Kaise M, Miwa J, Fujimoto A, Tashiro J, Tagami D, Sano H, Ohmoto Y. Influence of Helicobacter pylori status and eradication on the serum levels of trefoil factors and pepsinogen test: serum trefoil factor 3 is a stable biomarker. Gastric Cancer. 2013;16:329-337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 21]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
45.  Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, Sweet-Cordero A, Ebert BL, Mak RH, Ferrando AA. MicroRNA expression profiles classify human cancers. Nature. 2005;435:834-838.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Wang J, Wang Q, Liu H, Hu B, Zhou W, Cheng Y. MicroRNA expression and its implication for the diagnosis and therapeutic strategies of gastric cancer. Cancer Lett. 2010;297:137-143.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Hunter MP, Ismail N, Zhang X, Aguda BD, Lee EJ, Yu L, Xiao T, Schafer J, Lee ML, Schmittgen TD. Detection of microRNA expression in human peripheral blood microvesicles. PLoS One. 2008;3:e3694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1078]  [Cited by in F6Publishing: 1094]  [Article Influence: 68.4]  [Reference Citation Analysis (0)]
48.  Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O’Briant KC, Allen A. Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA. 2008;105:10513-10518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5636]  [Cited by in F6Publishing: 6046]  [Article Influence: 377.9]  [Reference Citation Analysis (0)]
49.  Lawrie CH, Gal S, Dunlop HM, Pushkaran B, Liggins AP, Pulford K, Banham AH, Pezzella F, Boultwood J, Wainscoat JS. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Br J Haematol. 2008;141:672-675.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1258]  [Cited by in F6Publishing: 1278]  [Article Influence: 79.9]  [Reference Citation Analysis (0)]
50.  Chen X, Hu Z, Wang W, Ba Y, Ma L, Zhang C, Wang C, Ren Z, Zhao Y, Wu S. Identification of ten serum microRNAs from a genome-wide serum microRNA expression profile as novel noninvasive biomarkers for nonsmall cell lung cancer diagnosis. Int J Cancer. 2012;130:1620-1628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 216]  [Cited by in F6Publishing: 235]  [Article Influence: 18.1]  [Reference Citation Analysis (0)]
51.  Ng EK, Chong WW, Jin H, Lam EK, Shin VY, Yu J, Poon TC, Ng SS, Sung JJ. Differential expression of microRNAs in plasma of patients with colorectal cancer: a potential marker for colorectal cancer screening. Gut. 2009;58:1375-1381.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 855]  [Cited by in F6Publishing: 938]  [Article Influence: 62.5]  [Reference Citation Analysis (0)]
52.  Shiotani A, Murao T, Kimura Y, Matsumoto H, Kamada T, Kusunoki H, Inoue K, Uedo N, Iishi H, Haruma K. Identification of serum miRNAs as novel non-invasive biomarkers for detection of high risk for early gastric cancer. Br J Cancer. 2013;109:2323-2330.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 68]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
53.  Graham DY, Asaka M. Eradication of gastric cancer and more efficient gastric cancer surveillance in Japan: two peas in a pod. J Gastroenterol. 2010;45:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 65]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
54.  Take S, Mizuno M, Ishiki K, Nagahara Y, Yoshida T, Yokota K, Oguma K. Baseline gastric mucosal atrophy is a risk factor associated with the development of gastric cancer after Helicobacter pylori eradication therapy in patients with peptic ulcer diseases. J Gastroenterol. 2007;42 Suppl 17:21-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 93]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
55.  Take S, Mizuno M, Ishiki K, Yoshida T, Ohara N, Yokota K, Oguma K, Okada H, Yamamoto K. The long-term risk of gastric cancer after the successful eradication of Helicobacter pylori. J Gastroenterol. 2011;46:318-324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 97]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
56.  Ma JL, Zhang L, Brown LM, Li JY, Shen L, Pan KF, Liu WD, Hu Y, Han ZX, Crystal-Mansour S. Fifteen-year effects of Helicobacter pylori, garlic, and vitamin treatments on gastric cancer incidence and mortality. J Natl Cancer Inst. 2012;104:488-492.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 314]  [Cited by in F6Publishing: 325]  [Article Influence: 27.1]  [Reference Citation Analysis (1)]
57.  Fuccio L, Zagari RM, Eusebi LH, Laterza L, Cennamo V, Ceroni L, Grilli D, Bazzoli F. Meta-analysis: can Helicobacter pylori eradication treatment reduce the risk for gastric cancer? Ann Intern Med. 2009;151:121-128.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Yanaoka K, Oka M, Ohata H, Yoshimura N, Deguchi H, Mukoubayashi C, Enomoto S, Inoue I, Iguchi M, Maekita T. Eradication of Helicobacter pylori prevents cancer development in subjects with mild gastric atrophy identified by serum pepsinogen levels. Int J Cancer. 2009;125:2697-2703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 111]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
59.  Shiotani A, Murao T, Uedo N, Iishi H, Yamanaka Y, Kamada T, Kusunoki H, Inoue K, Haruma K. Eradication of H. pylori did not improve abnormal sonic hedgehog expression in the high risk group for gastric cancer. Dig Dis Sci. 2012;57:643-649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
60.  Shiotani A, Uedo N, Iishi H, Murao T, Kanzaki T, Kimura Y, Kamada T, Kusunoki H, Inoue K, Haruma K. H. pylori eradication did not improve dysregulation of specific oncogenic miRNAs in intestinal metaplastic glands. J Gastroenterol. 2012;47:988-998.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 41]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
61.  Leja M, Wex T, Malfertheiner P. Markers for gastric cancer premalignant lesions: where do we go? Dig Dis. 2012;30:268-276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 24]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
62.  Uemura N, Mukai T, Okamoto S, Yamaguchi S, Mashiba H, Taniyama K, Sasaki N, Haruma K, Sumii K, Kajiyama G. Effect of Helicobacter pylori eradication on subsequent development of cancer after endoscopic resection of early gastric cancer. Cancer Epidemiol Biomarkers Prev. 1997;6:639-642.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Maehata Y, Nakamura S, Fujisawa K, Esaki M, Moriyama T, Asano K, Fuyuno Y, Yamaguchi K, Egashira I, Kim H. Long-term effect of Helicobacter pylori eradication on the development of metachronous gastric cancer after endoscopic resection of early gastric cancer. Gastrointest Endosc. 2012;75:39-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 145]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
64.  Kato M, Nishida T, Yamamoto K, Hayashi S, Kitamura S, Yabuta T, Yoshio T, Nakamura T, Komori M, Kawai N. Scheduled endoscopic surveillance controls secondary cancer after curative endoscopic resection for early gastric cancer: a multicentre retrospective cohort study by Osaka University ESD study group. Gut. 2013;62:1425-1432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 181]  [Article Influence: 16.5]  [Reference Citation Analysis (0)]
65.  Seo JY, Lee DH, Cho Y, Lee DH, Oh HS, Jo HJ, Shin CM, Lee SH, Park YS, Hwang JH. Eradication of Helicobacter pylori reduces metachronous gastric cancer after endoscopic resection of early gastric cancer. Hepatogastroenterology. 2013;60:776-780.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 17]  [Reference Citation Analysis (0)]
66.  Hanaoka N, Uedo N, Shiotani A, Inoue T, Takeuchi Y, Higashino K, Ishihara R, Iishi H, Haruma K, Tatsuta M. Autofluorescence imaging for predicting development of metachronous gastric cancer after Helicobacter pylori eradication. J Gastroenterol Hepatol. 2010;25:1844-1849.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 39]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
67.  Sakata K, Tamura G, Endoh Y, Ohmura K, Ogata S, Motoyama T. Hypermethylation of the hMLH1 gene promoter in solitary and multiple gastric cancers with microsatellite instability. Br J Cancer. 2002;86:564-567.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 34]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
68.  Fukuda M, Yokozaki H, Shiba M, Higuchi K, Arakawa T. Genetic and epigenetic markers to identify high risk patients for multiple early gastric cancers after treatment with endoscopic mucosal resection. J Clin Biochem Nutr. 2007;40:203-209.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 9]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
69.  Chon I, Choi C, Shin CM, Park YS, Kim N, Lee DH. Effect of Helicobacter pylori eradication on subsequent dysplasia development after endoscopic resection of gastric dysplasia. Korean J Gastroenterol. 2013;61:307-312.  [PubMed]  [DOI]  [Cited in This Article: ]