Editorial Open Access
Copyright ©2010 Baishideng. All rights reserved.
World J Gastrointest Oncol. Feb 15, 2010; 2(2): 59-64
Published online Feb 15, 2010. doi: 10.4251/wjgo.v2.i2.59
Early-onset gastric cancer: Learning lessons from the young
Anya N Milne, Department of Pathology, University Medical Centre Utrecht, Postbus 85500, 3508 GA, Utrecht, The Netherlands
G Johan A Offerhaus, Department of Pathology, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
Author contributions: Milne AN designed the study and wrote the paper; Offerhaus GJA provided significant intellectual content and critically revised the manuscript.
Correspondence to: Anya N Milne, PhD, Department of Pathology, University Medical Centre Utrecht, Postbus 85500, 3508GA, Utrecht, The Netherlands. a.n.a.milne@umcutrecht.nl
Telephone: +31-30-2507663 Fax: +31-30-2544990
Received: June 10, 2009
Revised: July 27, 2009
Accepted: August 3, 2009
Published online: February 15, 2010

Abstract

There is by no means a clear-cut pattern of mutations contributing to gastric cancers, and gastric cancer research can be hampered by the diversity of factors that can induce gastric cancer, such as Helicobacter pylori infection, diet, ageing and other environmental factors. Tumours are unquestionably riddled with genetic changes yet we are faced with an unsolvable puzzle with respect to a temporal relationship. It is postulated that inherited genetic factors may be more important in early-onset gastric cancer (EOGC) than in gastric cancers found in older patients as they have less exposure to environmental carcinogens. EOGC, therefore, could provide a key to unravelling the genetic changes in gastric carcinogenesis. Gastric cancers occurring in young patients provide an ideal background on which to try and uncover the initiating stages of gastric carcinogenesis. This review summarizes the literature regarding EOGC and also presents evidence that these cancers have a unique molecular-genetic phenotype, distinct from conventional gastric cancer.

Key Words: Gastric cancer, Early-onset gastric cancer, Helicobacter pylori



INTRODUCTION

Gastric cancer is the fourth most common malignancy in the world and ranks second in terms of cancer-related death[1]. It is thought that gastric cancer results from a combination of environmental factors and the accumulation of generalized and specific genetic alterations, and consequently affects mainly older patients often after a long period of atrophic gastritis. The most common cause of gastritis is infection by Helicobacter pylori (H. pylori), which is the single most common cause of gastric cancer[2,3] and has been classified by the World Health organization (WHO) as a classIcarcinogen since 1994[4]. The risk of infection varies with age, geographical location and ethnicity, but overall 15%-20% of infected patients develop gastric or duodenal ulcer disease and less than 1% will develop gastric adenocarcinoma[4]. Environmental and other risk factors for gastric cancer are summarised in Table 1 and have been recently reviewed by Milne et al[5,6].

Table 1 Environmental and other risk factors for conventional gastric cancer.
Increased risk of gastric cancerDecreased risk of gastric cancer
CHD1, TP53, BRCA2 germline mutationFruit and vegetables
Hereditary non-polyposis colorectal cancerAscorbic acid
Helicobacter pyloriinfectionCarotenoids
Ebstein barrvirus infectionFolates
Cigarette smokingTocopherols
Smoked or cured meat and fishCereal fibres
Pickled vegetablesNumerous polymorphisms (as mentioned under increased risk)
Chilli peppers
Alcohol
Exposure to nitrosamines + inorganic dust
Obesity
Pharmacological gastric acid suppression
IL-1β-31 polymorphism
1195 COX-2 polymorphism
Other polymorphisms: MTHFR, PSCA, XPA, XPC, ERCC2, GSTT1, SULT1A1, NAT2, EPHX1, Toll-like receptor 4

A pattern of gastritis has also been shown to correlate strongly with the risk of gastric adenocarcinoma. The presence of antral-predominant gastritis, the most common form, confers a higher risk of developing peptic ulcers; whereas corpus predominant gastritis and multifocal atrophic gastritis lead to a higher risk of developing gastric ulcers and subsequent gastric cancer[7,8]. The response to H. pylori infection and the subsequent pattern of gastritis depends on the genotype of the patient and in particular a polymorphism in IL-1β, an inflammatory mediator triggered by H. pylori infection, is known to be of importance[9]. Multifocal atrophic gastritis is usually accompanied by intestinal metaplasia and leads to cancer via dysplasia, and thus intestinal metaplasia is considered to be a dependable morphological marker for gastric cancer risk. Unlike intestinal gastric cancer, the diffuse type typically develops following chronic inflammation without passing through the intermediate steps of atrophic gastritis or intestinal metaplasia.

Several classification systems have been proposed, but the most commonly used are those of the WHO and of Laurén who describes two main histological types, diffuse and intestinal[10]. Intestinal adenocarcinoma predominates in high-risk areas whereas the diffuse adenocarcinoma is more common in low-risk areas[11]. Although classification varies between Japan and the West, attempts have been made recently to standardize systems[12]. Early gastric cancer is a term to describe carcinomas limited to the mucosa or to both the mucosa and submucosa, regardless of nodal status. The prevalence of this lesion is higher in countries such as Japan, where a screening programme is carried out.

There is by no means a clear-cut pattern of mutations in gastric cancers, with no known multi-step pathway, and genetic research can often be hampered by the diversity of changes that are induced by H. pylori infection, diet, ageing and other environmental factors. Tumours are unquestionably riddled with genetic changes, as summarized in Figure 1, yet we are faced with an unsolvable puzzle with respect to a temporal relationship. In order to solve this problem, one approach is to investigate tumours that are less influenced by these environmental factors. Gastric cancers occurring in young patients, known as early-onset gastric cancers (EOGC), provide an ideal background on which to try and uncover the initiating stages in gastric carcinogenesis. In addition, hereditary cancers can often illuminate discrete mutations that can initiate the pathway of gastric carcinogenesis.

Figure 1
Figure 1 This figure summarizes the molecular genetic changes in conventional gastric cancer and emphasizes the lack of a multi-step pathway in gastric cancer, despite extensive research in the field. It highlights the need for a new approach to understanding gastric cancer, such as examining early-onset gastric cancers and hereditary gastric cancers, where we can learn from the young.
EARLY ONSET GASTRIC CANCER

Gastric cancer is rare below the age of 30 thereafter it increases rapidly and steadily to reach the highest rates in the oldest age groups, both in males and females. The intestinal type rises faster with age than the diffuse type and is more frequent in males than in females. EOGC is defined as gastric cancer presenting at the age of 45 or younger. Approximately 10% of gastric cancer patients fall into the EOGC category[13], although rates vary between 2.7%[14] and 15%[15] depending on the population studied. Young patients more frequently develop diffuse lesions, which often arise on the background of histologically “normal” gastric mucosa. It is postulated that genetic factors may be more important in EOGC than in older patients as younger patients have less exposure to environmental carcinogens[5,16], thus these cancers could provide a key to unravelling the genetic changes in gastric carcinogenesis. H. pylori may still play a role in the development of gastric cancer in young patients[17-19], and there is, in fact, no difference in the distribution of gastric cancer predisposing IL1β polymorphisms between young and old patients[20]. However, the role of H. pylori is likely to involve a much smaller percentage of patients than in the older age group. Epstein-Barr virus (EBV), which is observed in 7%-20% of gastric cancers, has been implicated in gastric carcinogenesis and occurs slightly more frequently in diffuse-type gastric cancers[21-23]. However, the levels of EBV infection appear to be much lower (or absent) in EOGC[24].

The clinicopathological features of gastric carcinoma are said to differ between young and elderly patients[25] and it has been claimed that young patients have a poorer prognosis[26]. Others report that tumour staging and prognosis for young patients is similar to older patients and depends on whether the patients undergo a curative resection[13,15,27]. Young patients with gastric cancer in the United States are more likely to be black, Asian or Hispanic[28]. Relative to older patients, young patients have a female preponderance, a more frequent occurrence of diffuse cancer and less intestinal metaplasia[13,28,29]. This predominance of females is considered by some to be due to hormonal factors[30,31]. Cancers in young patients are more often multifocal than in older patients[32] as is also seen in HDGC[33].

Approximately 10% of young gastric cancer patients have a positive family history[13], some of which are accounted for by inherited gastric cancer predisposition syndromes. Although the underlying genetic events are not always known, it can involve CDH1 germline mutations[34-36], encoding an aberrant form of E-cadherin, resulting in hereditary diffuse gastric cancer (HDGC), as recently reviewed by Carneiro et al[36]. In fact, some suggest that when looking at hMLH1 and CDH1 germline mutations, 2%-3% of EOCG cases in North Americans may be due to high-risk genetic mutations[37,38]. The 90% without a family history emphasizes that the occurrence of gastric cancer in young patients remains largely unexplained, and is probably caused by a predisposing genotype that has facilitated cancer development due various environmental triggers[6].

It has been discovered that EOGCs have a different clinicopathological profile than conventional gastric carcinomas. This suggests that they represent a separate entity within gastric carcinogenesis and indeed evidence at a molecular genetic level supports this. The majority of gastric adenocarcinomas, like many other solid tumours, show defects in the maintenance of genome stability, resulting in DNA copy number alteration that can be analysed by (microarray-based) comparative genomic hybridization (array CGH). Hierarchical cluster analysis of array CGH data on 46 gastric cancer patients (including 12 young patients) revealed clusters with genomic profiles that correlated significantly with age[39]. Gains at chromosomes 17q, 19q and 20q have been found in EOGC with comparative genomic hybridization[40] and LOH findings have also shown that losses are infrequent in EOGC[24].

The presence of microsatellite instability (MSI), which usually occurs at a frequency of 15%-20% in older gastric carcinomas, also varies dramatically between gastric cancer in young and old patients, with MSI consistently absent in EOGC[24,29,41,42]. These results have been found despite the analysis of distal tumours (where MSI is usually more common) and inclusion of mixed and intestinal type tumours (diffuse tumours generally have less MSI)[43]. However, it may be that geographical factors play a role[44]. A lack of MSI excludes the mutator phenotype as an important predisposing factor in the development of EOGC. This contrasts with the situation in colorectal cancer where 58% of patients without HNPCC aged under 35 years showed evidence of MSI[45]. EOGC also contrasts with colorectal cancer with respect to the tumor suppressor gene APC, which causes familial adenomatosis polyposis syndrome. The role of APC in EOGC is limited and nuclear expression of β-catenin has not been found to differ between EOGC and conventional gastric cancers[46,47].

Molecular expression profiles of EOGC and conventional gastric cancers have been found to differ and EOGC has a COX-2 Low, TFF-1 expressing phenotype[46]. In light of studies showing the reduced risk of gastric cancer in non-steroidal anti-inflammatory drug users[48,49], these results may have clinical implications, as they suggest that this reduced risk may apply only to gastric cancer in older patients, as COX-2 does not appear to play an important role in EOGC. It also implies that genetic changes typical for conventional tumors more readily induce COX-2 expression than those associated with EOGC. Interestingly, this COX-2 low phenotype cannot be explained by the increased presence of the COX2 -765 G>C polymorphism in EOGC[50]. A higher incidence of aberrant E-cadherin expression in EOGC regardless of histological type[29] has also been reported, although a more recent report that compared EOGC with conventional cancers showed that aberrant expression of E-cadherin correlated significantly with the diffuse type[46].

Deregulation of the cell cycle is known to be a critical event in the onset of tumourigenesis, and thus the finding of low molecular weight isoforms of cyclin E in EOGC, which are reported to be constitutively active in breast cancer[51], are of great interest. The expression of these isoforms differs between EOGC and conventional cancers, being present in 35% of EOGCs, compared to in 8% of conventional gastric cancers and 4% of stump cancers[52]. In addition, these low molecular weight isoforms in EOGC diverge from the classical role of cyclin E as oncogenes and were found to be an independent positive prognostic indicator in EOGC[52] adding to reports where the role of cyclin E conflicted with previous dogma[53,54]. This complexity of molecular wiring in carcinogenesis has also been emphasized in recent literature, with the conclusion that cancer can no longer be viewed purely in terms of a network of oncogenes and tumour suppressor genes[55,56].

Further evidence that EOGCs display molecular characteristics different from conventional carcinomas comes from a study where amplification at 11p12-13 was found in gastric cancer using representational difference analysis and was confirmed by Southern blot analysis. It was found that overexpression of the isoform CD44v6 correlated with this amplification in diffuse type cancer and that this overexpression occurred more commonly in EOGC regardless of histological type[57].

The gene RUNX3 has been a subject of great debate in gastric cancer studies in recent years, following a study where loss of the gene was shown to be associated with stimulated proliferation and suppressed apoptosis of gastric epithelial cells[58]. Conflicting evidence has, however, also been present, as the expression of RUNX3 in the gastric mucosa of mice differed significantly between strains analysed[59]. Furthermore, the gastric hyperplasia observed in the Runx3-/- mice used in Li’s study was not observed in the mouse strain studied by Levanon et al[60]. Recent literature regarding RUNX3 has excluded it as having a tumour suppressor function in EOGC[61], although as some of the cell lines used in this study were from conventional gastric cancers, the implications may be more far-reaching and call the importance of RUNX3 in all gastric cancers into question.

Classic genetics alone cannot explain sporadic EOGC and cancer development in patients with a weak family history. The concept of epigenetics offers a partial explanation and may have important clinical implications for these types of cancer. The best-known epigenetic marker is DNA methylation, which occurs in CpG sites (islands), has critical roles in the control of gene activity, and is influenced by the modifications in histone structure that are commonly disrupted in cancer cells. Gene promoter methylation, a phenomenon that increases with age and may account for the increase in cancer in older age groups, has also been found to occur in EOGC[47]. However, comparison with the conventional group has not yet been carried out.

As supported by the literature, summarised in Table 2, EOGCs differ from conventional gastric cancers, not only at a clinicopathological level, but also at a molecular genetic level. If this is indeed due to the fact that the environment plays a smaller role in triggering the carcinogenic pathway, the investigation of this group of cancers may reveal genetic changes that assist in the task of putting forward a multistep pathway for gastric cancer.

Table 2 Clinico-pathological and molecular-genetic differences between early-onset and conventional gastric cancers.
Conventional gastric cancerEarly-onset gastric cancerRef.
Equally common in male and femalesMore common in females[13,28,30,31]
Intestinal type cancer more commonDiffuse type cancer more common[13,28]
Usually unifocalOften multifocal[32,33]
Often preceded by intestinal metaplasiaNo intestinal metaplasia[13,28]
Microsatellite Instability in 15%-20%Lack of MSI[24,29,41-43]
Commonly find loss of heterozygosityInfrequent loss of heterozygosity[24]
COX2 overexpression in 66%COX2 overexpression in 10%[46]
Loss of TFF1 expression in 73%Loss of TFF1 expression in 39%[46]
Loss of RUNX3 geneNo loss of RUNX3[58-61]
Widespread gains throughout genomeGains at chromosomes 17q, 19q and 20q[40]
Distinct gene clusters on hierarchical analysisDistinct gene clusters on hierarchical analysis[39]
Infrequent LMW isoforms of cyclin EFrequent LMW isoforms of cyclin E[52]
CD44v6 expressionCD44v6 more commonly expressed[57]
Usually no family history10% with a family history[13]
FUTURE PROSPECTIVES

Gastric carcinoma continues to be a cause of premature death, despite progress in detection and treatment and despite advances in our understanding of the molecular basis of cancer. The need to develop efficient and effective cancer-specific drugs is coupled with the importance of accurate prediction of disease outcome for various patient groups, some of whom, due to the biology of their disease, will do better than others and may warrant a different treatment protocol. However, the multi-step pathway of carcinogenesis that occurs in some epithelial cancers and that has allowed accurate clinical and pathologic characterization is not yet elucidated in gastric cancer. Gastric cancers often occur without any consistent mutational abnormality and with considerable variation in pathogenesis ranging from a stepwise progression of changes to tumours arising in the absence of a precursor lesion. As has been highlighted in this article, there is growing evidence to support the hypothesis that young patients develop carcinomas with a different molecular genetic profile from that of sporadic carcinomas occurring at a later age. Further study of hereditary gastric cancers and EOGC as unique subsets of gastric cancer may aid us in the search for a gastric cancer pathway.

Footnotes

Peer reviewer: László Herszényi, MD, PhD, Associate Professor, 2nd Department of Medicine, Semmelweis University, H-1088 Budapest, Szentkiralyi Str 46, Hungary

S- Editor Li LF L- Editor Lutze M E- Editor Lin YP

References
1.  Parkin DM, Bray F, Ferlay J, Pisani P. Estimating the world cancer burden: Globocan 2000. Int J Cancer. 2001;94:153-156.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  Forman D, Newell DG, Fullerton F, Yarnell JW, Stacey AR, Wald N, Sitas F. Association between infection with Helicobacter pylori and risk of gastric cancer: evidence from a prospective investigation. BMJ. 1991;302:1302-1305.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Parsonnet J, Friedman GD, Vandersteen DP, Chang Y, Vogelman JH, Orentreich N, Sibley RK. Helicobacter pylori infection and the risk of gastric carcinoma. N Engl J Med. 1991;325:1127-1131.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Suerbaum S, Michetti P. Helicobacter pylori infection. N Engl J Med. 2002;347:1175-1186.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Milne AN, Sitarz R, Carvalho R, Carneiro F, Offerhaus GJ. Early onset gastric cancer: on the road to unraveling gastric carcinogenesis. Curr Mol Med. 2007;7:15-28.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Milne AN, Carneiro F, O'Morain C, Offerhaus GJ. Nature meets nurture: molecular genetics of gastric cancer. Hum Genet. 2009;126:615-628.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Craanen ME, Dekker W, Blok P, Ferwerda J, Tytgat GN. Intestinal metaplasia and Helicobacter pylori: an endoscopic bioptic study of the gastric antrum. Gut. 1992;33:16-20.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Watanabe T, Tada M, Nagai H, Sasaki S, Nakao M. Helicobacter pylori infection induces gastric cancer in mongolian gerbils. Gastroenterology. 1998;115:642-648.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, Young HA, Herrera J, Lissowska J, Yuan CC, Rothman N. Interleukin-1 polymorphisms associated with increased risk of gastric cancer. Nature. 2000;404:398-402.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Lauren P. The Two Histological Main Types of Gastric Carcinoma: Diffuse and So-Called Intestinal-Type Carcinoma. An Attempt at a Histo-Clinical Classification. Acta Pathol Microbiol Scand. 1965;64:31-49.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Hamilton SR, Aaltonen LA.  World Health Organization Classification of Tumours. Pathology and Genetics of Tumours of the Digestive System. Lyon, France: IARC Press 2000; 204.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Schlemper RJ, Riddell RH, Kato Y, Borchard F, Cooper HS, Dawsey SM, Dixon MF, Fenoglio-Preiser CM, Fléjou JF, Geboes K. The Vienna classification of gastrointestinal epithelial neoplasia. Gut. 2000;47:251-255.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Kokkola A, Sipponen P. Gastric carcinoma in young adults. Hepatogastroenterology. 2001;48:1552-1555.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Umeyama K, Sowa M, Kamino K, Kato Y, Satake K. Gastric carcinoma in young adults in Japan. Anticancer Res. 1982;2:283-286.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Ramos-De la Medina A, Salgado-Nesme N, Torres-Villalobos G, Medina-Franco H. Clinicopathologic characteristics of gastric cancer in a young patient population. J Gastrointest Surg. 2004;8:240-244.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Correa P, Shiao YH. Phenotypic and genotypic events in gastric carcinogenesis. Cancer Res. 1994;54:1941s-1943s.  [PubMed]  [DOI]  [Cited in This Article: ]
17.  Rugge M, Busatto G, Cassaro M, Shiao YH, Russo V, Leandro G, Avellini C, Fabiano A, Sidoni A, Covacci A. Patients younger than 40 years with gastric carcinoma: Helicobacter pylori genotype and associated gastritis phenotype. Cancer. 1999;85:2506-2511.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Koshida Y, Koizumi W, Sasabe M, Katoh Y, Okayasu I. Association of Helicobacter pylori-dependent gastritis with gastric carcinomas in young Japanese patients: histopathological comparison of diffuse and intestinal type cancer cases. Histopathology. 2000;37:124-130.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Haruma K, Komoto K, Kamada T, Ito M, Kitadai Y, Yoshihara M, Sumii K, Kajiyama G. Helicobacter pylori infection is a major risk factor for gastric carcinoma in young patients. Scand J Gastroenterol. 2000;35:255-259.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Sitarz R, de Leng WW, Polak M, Morsink FH, Bakker O, Polkowski WP, Maciejewski R, Offerhaus GJ, Milne AN. IL-1B -31T>C promoter polymorphism is associated with gastric stump cancer but not with early onset or conventional gastric cancers. Virchows Arch. 2008;453:249-255.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Rugge M, Genta RM. Epstein-Barr virus: a possible accomplice in gastric oncogenesis. J Clin Gastroenterol. 1999;29:3-5.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Shibata D, Weiss LM. Epstein-Barr virus-associated gastric adenocarcinoma. Am J Pathol. 1992;140:769-774.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Shibata D, Hawes D, Stemmermann GN, Weiss LM. Epstein-Barr virus-associated gastric adenocarcinoma among Japanese Americans in Hawaii. Cancer Epidemiol Biomarkers Prev. 1993;2:213-217.  [PubMed]  [DOI]  [Cited in This Article: ]
24.  Carvalho R, Milne AN, van Rees BP, Caspers E, Cirnes L, Figueiredo C, Offerhaus GJ, Weterman MA. Early-onset gastric carcinomas display molecular characteristics distinct from gastric carcinomas occurring at a later age. J Pathol. 2004;204:75-83.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Maehara Y, Emi Y, Tomisaki S, Oshiro T, Kakeji Y, Ichiyoshi Y, Sugimachi K. Age-related characteristics of gastric carcinoma in young and elderly patients. Cancer. 1996;77:1774-1780.  [PubMed]  [DOI]  [Cited in This Article: ]
26.  Theuer CP, de Virgilio C, Keese G, French S, Arnell T, Tolmos J, Klein S, Powers W, Oh T, Stabile BE. Gastric adenocarcinoma in patients 40 years of age or younger. Am J Surg. 1996;172:473-476; discussion 476-477.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Medina-Franco H, Heslin MJ, Cortes-Gonzalez R. Clinicopathological characteristics of gastric carcinoma in young and elderly patients: a comparative study. Ann Surg Oncol. 2000;7:515-519.  [PubMed]  [DOI]  [Cited in This Article: ]
28.  Matley PJ, Dent DM, Madden MV, Price SK. Gastric carcinoma in young adults. Ann Surg. 1988;208:593-596.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Lim S, Lee HS, Kim HS, Kim YI, Kim WH. Alteration of E-cadherin-mediated adhesion protein is common, but microsatellite instability is uncommon in young age gastric cancers. Histopathology. 2003;42:128-136.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Maeta M, Yamashiro H, Oka A, Tsujitani S, Ikeguchi M, Kaibara N. Gastric cancer in the young, with special reference to 14 pregnancy-associated cases: analysis based on 2,325 consecutive cases of gastric cancer. J Surg Oncol. 1995;58:191-195.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Derakhshan MH, Liptrot S, Paul J, Brown IL, Morrison D, McColl KE. Oesophageal and gastric intestinal-type adenocarcinomas show the same male predominance due to a 17 year delayed development in females. Gut. 2009;58:16-23.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Furukawa H, Iwanaga T, Imaoka S, Hiratsuka M, Fukuda I, Kabuto T, Ishikawa O, Sasaki Y. Multifocal gastric cancer in patients younger than 50 years of age. Eur Surg Res. 1989;21:313-318.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Carneiro F, Huntsman DG, Smyrk TC, Owen DA, Seruca R, Pharoah P, Caldas C, Sobrinho-Simões M. Model of the early development of diffuse gastric cancer in E-cadherin mutation carriers and its implications for patient screening. J Pathol. 2004;203:681-687.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Suriano G, Oliveira C, Ferreira P, Machado JC, Bordin MC, De Wever O, Bruyneel EA, Moguilevsky N, Grehan N, Porter TR. Identification of CDH1 germline missense mutations associated with functional inactivation of the E-cadherin protein in young gastric cancer probands. Hum Mol Genet. 2003;12:575-582.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Suriano G, Yew S, Ferreira P, Senz J, Kaurah P, Ford JM, Longacre TA, Norton JA, Chun N, Young S. Characterization of a recurrent germ line mutation of the E-cadherin gene: implications for genetic testing and clinical management. Clin Cancer Res. 2005;11:5401-5409.  [PubMed]  [DOI]  [Cited in This Article: ]
36.  Carneiro F, Oliveira C, Suriano G, Seruca R. Molecular pathology of familial gastric cancer, with an emphasis on hereditary diffuse gastric cancer. J Clin Pathol. 2008;61:25-30.  [PubMed]  [DOI]  [Cited in This Article: ]
37.  Bacani JT, Soares M, Zwingerman R, di Nicola N, Senz J, Riddell R, Huntsman DG, Gallinger S. CDH1/E-cadherin germline mutations in early-onset gastric cancer. J Med Genet. 2006;43:867-872.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Bacani J, Zwingerman R, Di Nicola N, Spencer S, Wegrynowski T, Mitchell K, Hay K, Redston M, Holowaty E, Huntsman D. Tumor microsatellite instability in early onset gastric cancer. J Mol Diagn. 2005;7:465-477.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Buffart TE, Carvalho B, Hopmans E, Brehm V, Kranenbarg EK, Schaaij-Visser TB, Eijk PP, van Grieken NC, Ylstra B, van de Velde CJ. Gastric cancers in young and elderly patients show different genomic profiles. J Pathol. 2007;211:45-51.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Varis A, van Rees B, Weterman M, Ristimäki A, Offerhaus J, Knuutila S. DNA copy number changes in young gastric cancer patients with special reference to chromosome 19. Br J Cancer. 2003;88:1914-1919.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Hayden JD, Cawkwell L, Sue-Ling H, Johnston D, Dixon MF, Quirke P, Martin IG. Assessment of microsatellite alterations in young patients with gastric adenocarcinoma. Cancer. 1997;79:684-687.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Carneiro F, Oliveira C, Leite M, Seruca R. Molecular targets and biological modifiers in gastric cancer. Semin Diagn Pathol. 2008;25:274-287.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Seruca R, Sobrinho-Simões M. Assessment of microsatellite alterations in young patients with gastric adenocarcinoma. Cancer. 1997;80:1358-1360.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Hayden JD, Cawkwell L, Dixon MF, Pardal F, Murgatroyd H, Gray S, Quirke P, Martin IG. A comparison of microsatellite instability in early onset gastric carcinomas from relatively low and high incidence European populations. Int J Cancer. 2000;85:189-191.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Liu B, Farrington SM, Petersen GM, Hamilton SR, Parsons R, Papadopoulos N, Fujiwara T, Jen J, Kinzler KW, Wyllie AH. Genetic instability occurs in the majority of young patients with colorectal cancer. Nat Med. 1995;1:348-352.  [PubMed]  [DOI]  [Cited in This Article: ]
46.  Milne AN, Carvalho R, Morsink FM, Musler AR, de Leng WW, Ristimäki A, Offerhaus GJ. Early-onset gastric cancers have a different molecular expression profile than conventional gastric cancers. Mod Pathol. 2006;19:564-572.  [PubMed]  [DOI]  [Cited in This Article: ]
47.  Kim HC, Kim JC, Roh SA, Yu CS, Yook JH, Oh ST, Kim BS, Park KC, Chang R. Aberrant CpG island methylation in early-onset sporadic gastric carcinoma. J Cancer Res Clin Oncol. 2005;131:733-740.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Langman MJ, Cheng KK, Gilman EA, Lancashire RJ. Effect of anti-inflammatory drugs on overall risk of common cancer: case-control study in general practice research database. BMJ. 2000;320:1642-1646.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Akre K, Ekström AM, Signorello LB, Hansson LE, Nyrén O. Aspirin and risk for gastric cancer: a population-based case-control study in Sweden. Br J Cancer. 2001;84:965-968.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Sitarz R, Leguit RJ, de Leng WW, Polak M, Morsink FM, Bakker O, Maciejewski R, Offerhaus GJ, Milne AN. The COX-2 promoter polymorphism -765 G>C is associated with early-onset, conventional and stump gastric cancers. Mod Pathol. 2008;21:685-690.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Keyomarsi K, Conte D Jr, Toyofuku W, Fox MP. Deregulation of cyclin E in breast cancer. Oncogene. 1995;11:941-950.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Milne AN, Carvalho R, Jansen M, Kranenbarg EK, van de Velde CJ, Morsink FM, Musler AR, Weterman MA, Offerhaus GJ. Cyclin E low molecular weight isoforms occur commonly in early-onset gastric cancer and independently predict survival. J Clin Pathol. 2008;61:311-316.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Berglund P, Stighall M, Jirström K, Borgquist S, Sjölander A, Hedenfalk I, Landberg G. Cyclin E overexpression obstructs infiltrative behavior in breast cancer: a novel role reflected in the growth pattern of medullary breast cancers. Cancer Res. 2005;65:9727-9734.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Takano Y, Kato Y, van Diest PJ, Masuda M, Mitomi H, Okayasu I. Cyclin D2 overexpression and lack of p27 correlate positively and cyclin E inversely with a poor prognosis in gastric cancer cases. Am J Pathol. 2000;156:585-594.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Ishikawa Y. Wnt signaling and orthopedic diseases. Am J Pathol. 2005;167:1-3.  [PubMed]  [DOI]  [Cited in This Article: ]
56.  Sharpless NE, DePinho RA. Cancer: crime and punishment. Nature. 2005;436:636-637.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Carvalho R, Milne AN, Polak M, Offerhaus GJ, Weterman MA. A novel region of amplification at 11p12-13 in gastric cancer, revealed by representational difference analysis, is associated with overexpression of CD44v6, especially in early-onset gastric carcinomas. Genes Chromosomes Cancer. 2006;45:967-975.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Li QL, Ito K, Sakakura C, Fukamachi H, Inoue K, Chi XZ, Lee KY, Nomura S, Lee CW, Han SB. Causal relationship between the loss of RUNX3 expression and gastric cancer. Cell. 2002;109:113-124.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Levanon D, Brenner O, Otto F, Groner Y. Runx3 knockouts and stomach cancer. EMBO Rep. 2003;4:560-564.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Levanon D, Bettoun D, Harris-Cerruti C, Woolf E, Negreanu V, Eilam R, Bernstein Y, Goldenberg D, Xiao C, Fliegauf M. The Runx3 transcription factor regulates development and survival of TrkC dorsal root ganglia neurons. EMBO J. 2002;21:3454-3463.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Carvalho R, Milne AN, Polak M, Corver WE, Offerhaus GJ, Weterman MA. Exclusion of RUNX3 as a tumour-suppressor gene in early-onset gastric carcinomas. Oncogene. 2005;24:8252-8258.  [PubMed]  [DOI]  [Cited in This Article: ]