Download World Journal of Surgical, Medical and Radiation Oncology

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
Transcript
World Journal of Surgical, Medical
and Radiation Oncology
Review
Open Access
Hereditary Gastric Cancer – Diagnosis and Clinical Implications
1Banasiewicz
Tomasz, 2,3 Stojcev Zoran, 4Pławski Andrzej
Department of General Surgery, Oncologic Gastroenterological and Plastic Surgery, Poznań
University of Medical Sciences, Poland
2 Regional Hospital, Słupsk, Department of General, Vascular and Oncologic Surgery, Słupsk, Poland
3 Department of Oncologic Surgery, Gdańsk Medical University, Gdańsk, Poland 4 Institute of Human
Genetics, Polish Academy of Sciences Poznan, Poland
1
This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution, and reproduction in any
medium, provided the original work is properly cited
Abstract
Gastric cancer is still one of the world's leading causes of cancer mortality. Approximately 10% of
gastric cancers appear to have a familial predisposition. In this this group, 1-3% are considered as
hereditary syndromes, with a clear genetic pathway, associated with the most important
mechanisms of the CDH1 germline mutations. This mutation leads to the hereditary diffuse gastric
cancer syndrome (HDGC). Other syndromes of familial prevalence of the gastric cancer, correlated
with genetic mutations, are the hereditary nonpolyposis colorectal cancer, Li-Fraumeni syndrome,
Peutz-Jeghers syndrome, Familial Adenomatous Polyposis, Cowden disease. Families HDGC can be
identified and tested for causative mutations in CDH1; in other syndromes screening
gastroduodenoscopy is indicated.
Key words gastric cancer; CDH1 mutations; gastrectomy, HDGC
Hereditary Gastric Cancer
3% are considered as hereditary syndromes,
with a clear genetic pathway, associated with
Gastric cancer is one of the world's leading
the most important mechanisms of the CDH1
causes of cancer mortality [1], with an average
germline mutations leaded to the hereditary
700,000 deaths a year [2, 3]. There are two
diffuse gastric cancer syndrome (HDGC). Other
main histological types, diffuse and intestinal.
syndromes, correlated with genetic mutations
Intestinal type usually follows multifocal
are the hereditary nonpolyposis colorectal
atrophic gastritis and accompanied by
cancer, Li-Fraumeni syndrome, Peutz-Jeghers
intestinal metaplasia is more frequently
syndrome, Familial Adenomatous Polyposis,
observed in older patients. Diffuse cancer,
Cowden disease, gastric adenocarcinoma and
observed more frequently on young patients,
proximal polyposis of the stomach (GAPPS) and
can be multifocal and can be hereditary [4].
others [5].
Approximately 10% of gastric cancers appear
to have a familial predisposition, and only 10%
shows a familial cluster. In this this group, 1Address for correspondence and reprint requests to:
Department
of
General
Surgery,
Oncologic
Gastroenterological and Plastic Surgery, Poznań University of
Medical Sciences, Poland Email [email protected]
© 2014 Tomasz B et al. Licensee Narain Publishers Pvt. Ltd.
(NPPL)
Submitted Friday, June 14, 2013Accepted Tuesday, August
06, 2013Published: Wednesday, Sunday, January 26, 2014
1
Gastric carcinoma involves numerous genetic
and epigenetic alterations. The most common,
sporadic gastric cancer is developed through
multistep processes, including the infectious
cases as Helicobacter pylori or Epstein-Barr
virus. Those types of the two infectionassociated gastric cancers are characterized by
global CpG island methylation in the promoter
region of cancer-related genes [6]. A
significantly smaller part of gastric cancers can
http://www.npplweb.com/wjsmro/content/3/1
World J Surg Med Radiat Oncol 2014;3:1-9
be caused by a specific germ-line mutation of
the E-cadherin gene (CDH1).
Germline mutations in the E-cadherin/CDH1
gene
The existence of a familial form of gastric
cancer was described in the Napoleon
Bonaparte family, with multiple cases of the
gastric cancers in this family [7]. During long
years individuals from these families lived with
the uncertainty of developing lethal gastric
cancer, but now families with this type of
gastric cancer can be identified and tested for
causative mutations in CDH1 [8]. The
cumulative lifetime risk of developing gastric
cancer in CDH1 mutation carriers is up to 80%.
Women from these families have an increased
risk for developing lobular breast cancer [9].
Approximately 1–3% of gastric cancer can be
caused by a germline mutation in the CDH1
gene, encoding E-cadherin. The data from these
families suggest that the penetrance of CDH1
gene mutations is high, ranging between 70 and
80% [10]. In families with HDGC CDH1
germline mutation is observed in 30-40%, the
remaining 60–70% are genetically unexplained
and may be caused by alterations in other
genes [11]. Those mutations were initially
identified in 1998 in New Zeeland, in Maori
families that were predisposed to diffuse
gastric cancer [12].
E-cadherin plays an import role in the
processes of development, cell differentiation,
and maintenance of epithelial architecture [13].
Analysis of genetic abnormalities in CDH1found
in HDGC shows that the most common are
inactivating mutations (splice site, frameshift,
and nonsense) rather than missense mutations.
Furthermore, CDH1 germline mutations are
evenly distributed along the E-cadherin gene. In
families with HDGC recently frequent deletions
of CDH1 have been recognized [14]. The
clustering in exons 7–9 is observed in sporadic
diffuse gastric cancer C [15].
Loss of
heterozygosity as the “second hit” does not
appear to be frequent in HDGC. As well as Ecadherin dysregulation, overexpression of
2
Tomasz B et al.
epidermal growth factor receptor (EGFR) is
among the most frequent genetic alterations
associated with diffuse-type gastric carcinoma.
There are some evidence suggests a functional
relationship between E-cadherin and EGFR that
regulates both proteins [16].
Germline alleles of E-cadherin (CDH1) can
explain approximately 30-40% of HDGC. The
factors driving susceptibility for the remaining
families remain unknown. Some of them can be
explain for example by the germline truncating
allele of alpha-E-catenin (CTNNA1). It can call
attention to the broader signaling network
surrounding these proteins in HDGC, because
alpha-E-catenin functions in the same complex
as E-cadherin [17].
Microsatellite Instability
Genetic instability at the level of microsatellite
instability (MSI) occurs in many sporadic
human cancers. The levels of MSI found in
gastric carcinomas from both Western and
Eastern populations are probably in the region
of up to 15–20% [18, 19]. In some studies
frameshift mutations in the Wnt pathway genes
AXIN2 and TCF7L2 have been found in GCs with
high microsatellite instability [20]. Alternative
lengthening of telomeres frequently occurs in
mismatch repair system-deficient gastric
carcinoma [21]. Several authors demonstrated
that the subset of sporadic GC with highfrequency MSI (MSI-H) is characterized by the
distinct clinicopathologic and genetic profile,
when compare with those with a low frequency
(MSI-L) or microsatellite stable (MSS) genotype
[22, 23, and 24]. However, whereas the role of
microsatellite instability and DNA mismatch
repair gene defects in HNPCC is unquestionable
and well established, the relevance of this
phenomenon in GC has limited clinical value
and is currently still don’t recognized [25].
Others Hereditary Syndromes
There are
risk of the
associated
mismatch
multiple syndromes with elevated
gastric cancer, as: Lynch syndrome
with germline mutations in DNA
repair genes and microsatellite
http://www.npplweb.com/wjsmro/content/3/1
World J Surg Med Radiat Oncol 2014;3:1-9
instability, in hereditary breast and ovarian
cancer syndrome due to germline BRCA1 and
BRCA2 mutations, in familial adenomatous
polyposis caused by germline APC mutations, in
Li-Fraumeni syndrome due to germline p53
mutations,
in
Peutz-Jeghers
syndrome
associated with germline STK11 mutations, in
juvenile polyposis syndrome associated with
germline mutations in the SMAD4 and BMPR1A
genes and in Cowden disease associated with
mutations in PTEN gene [26].
FAP: The incidence of gastric cancer in Western
FAP patients is not significantly different from
the general population.Asian patients, on the
other hand, have increased prevalence of
gastric cancer in FAP patients (2, 1 - 4.2%) [27].
In most cases gastric cancer arises from
adenomatous
polyps
via
the
adenomacarcinoma sequence, but several
reports
have
shown
that
gastric
adenocarcinoma can be developed from fundic
gland polyp in FAP patients [28,29,30].
Fundic gland polyps are the most prevalent
gastric lesions, followed by adenomatous
polyps. Those polyps occurred in about 50% of
FAP patients and are in this group the most
common type of the gastric polyps. In some
recent study the prevalence of fundic gland
polyp was about 60%, with the prevalence of
gastric adenoma as 6-14% [31]. Fundic gland
polyps are common in familial adenomatous
polyposis and attenuated familial adenomatous
polyposis and, if voluminous, may interfere
with effective endoscopic gastric cancer
surveillance. This family is believed to be the
first of its type reported with focus upon
education and genetic counseling in the setting
of a family information service [32].
Gastric adenocarcinoma and proximal
polyposis of the stomach (GAPPS) is the new,
recent describe autosomal dominant gastric
polyposis syndrome. GAPPS is a unique gastric
polyposis syndrome with a significant risk of
gastric adenocarcinoma. It is characterised by
the autosomal dominant transmission of fundic
gland polyposis, including areas of dysplasia or
intestinal-type
gastric
adenocarcinoma,
restricted to the proximal stomach, and with no
3
Hereditary Gastric Cancer
evidence of colorectal or duodenal polyposis or
other
heritable
gastrointestinal
cancer
syndromes. This syndrome was recognized in
three families from Australia, the USA and
Canada. The affected families were identified
through referral to centralised clinical genetics
centers [33].
In a nationwide epidemiologic study in Sweden
Hemminki and Jiang [34] found that the
population-attributable proportion of familial
gastric carcinoma was much lower than that
cited in the literature. Patterns of multiple
carcinomas suggested that immunologic factors
modulate susceptibility to gastric carcinoma.
The authors concluded that environmental
factors, perhaps H. pylori infections, were the
main reason for familial clustering of gastric
carcinoma.
Gastric cancer arises also from the
accumulation of genetic and epigenetic
alterations. Genetic variants of the genes IL-10,
IL-17, MUC1, MUC6, DNMT3B, SMAD4, and
SERPINE1 have been reported to modify the
risk of developing GC. Several genes have been
newly associated with gastric carcinogenesis,
both through oncogenic activation (GSK3β,
CD133, DSC2, P-Cadherin, CDH17, CD168,
CD44, metalloproteinases MMP7 and MMP11,
and a subset of miRNAs) and through tumor
suppressor gene inactivation mechanisms
(TFF1, PDX1, BCL2L10, XRCC, psiTPTE-HERV,
HAI-2, GRIK2, and RUNX3). The another
potential mechanism involved in the gastric
carcinogenesis is the inflammatory mediator
cyclooxygenase-2 (COX-2), discussed also as a
potential molecular target for therapy [35].
Lynch syndrome: The patients with colorectal
cancer and MMR gene mutations are at
increased risk of a greater range of cancers
than the recognized spectrum of Lynch
syndrome cancers, including breast, prostate
and gastric cancer. The risk for stomach cancer
is in this patients elevated compared with the
general population (SIR = 5.65, 95% CI = 2.32
to 9.69) for both sexes [36]. In patients with
Lynch syndromes the MLH1 methylation may
lead to the increased risk colorectal cancer as
http://www.npplweb.com/wjsmro/content/3/1
World J Surg Med Radiat Oncol 2014;3:1-9
well as stomach cancer and possibly ovarian
and liver cancer [37]. Somatic mutations of
mismatch repair (MMR) genes such as hMLH1
or hMSH2 are extremely rare in sporadic GCs,
with only one mutation found, in hMSH2 and
two cases of a germline frameshift mutation in
hMLH1 [36]. More recently, 29 sporadic GCs
with high level of MSI were screened for
somatic mutations in MLH1, MSH2, MSH6,
MLH3, and MBD4, and only five truncating
mutations (3 in MSH6, 1 in MLH3, and 1 in
MBD4) and one missense mutation (MLH1)
were identified. All truncating mutations were
found in the coding poly-A tracts, thus
suggesting that they result from the MSI
phenotype rather than causing it [38].
However, MSI positive tumours can still lack
hMLH1 protein expression and many studies
suggest that hypermethylation of the hMLH1
promoter region may be the principal
mechanism of gene inactivation in sporadic
gastric carcinomas with a high frequency of MSI
[39,40]. There is well-known fact that MSI may
be found in sporadic carcinomas that are
characteristic of hereditary nonpolyposis
colorectal cancer (HNPCC) [41], a syndrome
where germline mutations of the mismatch
repair genes are present. Genome-wide
expression profiles of sporadic GCs with and
without microsatellite instability reveal that the
immune and apoptotic gene networks
efficiently discriminated these two cancer types
[42].
Li-Fraumeni syndrome: hereditary, as well as
sporadic, gastric cancer can also be caused by
germline mutations of the TP53tumour
suppressor gene. This mutation occurs in the
Li–Fraumeni syndrome [43] and new germ line
mutations in this gene continue to be
discovered [44]. Mutations of tumor protein
p53 (TP53) and β-catenin (CTNNB1) genes are
detected early in the development of GC.
Furthermore, significant numbers of GCs show
loss of Runx3 due to hemizygous deletion and
hypermethylation of the promoter region.
Aberrant Cdx2 expression has been shown in
precancerous lesions as well as GC. However, it
4
Tomasz B et al.
remains unclear whether Cdx2 plays an
oncogenic role in gastric carcinogenesis [45].
BRCA1 and BRCA2 gene mutations: Those
mutations increased the risk of the breast
cancer, but the risk of gastric, ovarian,
pancreatic, and prostate cancers is also higher
than in normal population [46].
Clinical implications in
hereditary gastric cancer
families
with
The recent advances in the molecular genetics
lead to the recognition of the hereditary diffuse
gastric cancer that inherited in a dominant
autosomal manner with incomplete penetrance.
About 25-30% of families fulfilling the criteria
have germline mutation of the CDH1 gene
coding the calcium-dependent E-cadherin
protein. In the families with the occurrence of
the HDGC testing for CDH1 mutation is
recommended [47,48]. HDGC families can be
identified, tested for causative mutations in
CDH1, and for those families where a
pathogenic mutation can be identified,
prophylactic total gastrectomy can be
implemented in asymptomatic mutation
carriers who elect to virtually eliminate their
risk of developing this lethal disease [49].
Prophylactic gastrectomies are recommended
in unaffected CDH1 mutation carriers, because
screening endoscopic examinations and blind
biopsies have proven inadequate for
surveillance [50] The total gastrectomy is
suggested in the early adolescent age [51]. In
families with HDGC there is also an increased
frequency of cancers occurring at other sites
such as the breast, colorectum, and prostate in
these mutation carriers [52]. However,
inclusion of associated cancers into the
definition of HDGC is not yet recommended
[53].
There is a lack of recommendation in the other
genetic syndromes with the increased risk of
the gastric cancer. In families with polyposis
syndromes esophagogastroduodenoscopy by
age 25 years or prior to colon surgery is
recommended [54]. Endoscopy should be
repeated every 1-2 years. The screening
http://www.npplweb.com/wjsmro/content/3/1
World J Surg Med Radiat Oncol 2014;3:1-9
esophagogastroduodenoscopy should be the
most accepted procedure in families with the
aggregation of the gastric cancers.
Prevention and Treatment in Hereditary
Gastric Cancer
Hereditary Gastric Cancer
The interesting data analyzed the influence of
the selenium on the chromosome breakage in
BRCA1 carriers suggest that this treatment may
be effective also in other genetic syndromes
correlated with the hereditary gastric cancer
[62].
The prevalence of the gastric cancer, both
sporadic as the hereditary forms, is possible
mainly through the elimination of the life style
and environmental risk factors. The cigarette
smoking and H. pylori infection are classically
associated with gastric cancer [55], and diet is a
known etiological factor, especially for
intestinal-type adenocarcinoma whereby an
adequate intake of fruit and vegetables appears
to lower the risk with ascorbic acid,
carotenoids, folates and tocopherols acting as
antioxidants [56]. There are some data
suggested, that cereal fiber intake may reduce
the risk of adenocarcinoma, particularly diffuse
type [57]. The interplay of diet on genomic
stability has been recognized, by showing that
substances such as green tea can affect
methylation status of genes [58].
Conclusions
Different studies confirm the role of selenium in
carcinogenesis.
The
development
and
progression of gastric carcinoma can be
associated with decreasing levels of serum Se.
This concept is greatly supported by the
antioxidant action of Se [59]. Supplementation
of the selenium may protect against the
development of esophageal squamous cell
carcinoma (ESCC), esophageal adenocarcinoma
(EAC), and gastric cardia adenocarcinoma
(GCA) [60]
In others hereditary syndromes with familial
prevalence of the gastric cancer, as HNPCC, LiFraumeni syndrome, Peutz-Jeghers syndrome,
Familial Adenomatous Polyposis, Cowden
disease
the
screening
esophagogastroduodenoscopy should be the
most accepted procedure
The number of genetic therapies in gastric
cancer is still very limited. Targeted therapy
against GC with ERBB2 amplification recently
improved the prognosis of patients with
advanced GC. In addition, epigenetic changes in
GC could be attractive targets for cancer
treatment with modulators. A genome-wide
search has been undertaken to identify novel
methylation-silenced genes in GC, which will
help us understand the overall molecular
features of GC and further provide novel
opportunities in the treatment of GC [61].
5
Identification of the mutation and test for
causative mutation in CDH1 is indicated in
hereditary diffuse gastric cancer syndrome. For
families, where a pathogenic mutation can be
identified, prophylactic total gastrectomy can
be implemented in asymptomatic mutation
carriers who elect to eliminate their risk of
developing this lethal disease. Prophylactic
gastrectomies are recommended in unaffected
CDH1 mutation carriers, because screening
endoscopic examinations and blind biopsies
have proven inadequate for surveillance. In
families with HDGC there is also an increased
frequency of cancers occurring at other sites
such as the breast, colorectum, and prostate in
these mutation carriers.
Competing interests
The authors declare that they have no
competing interests.
Authors' contributions
TB conceived of the study, and participated in
its design and coordination and helped to draft
the manuscript.
ZS participated in the design of the publication,
review of the literature and data analysis, final
review and draft preparation
http://www.npplweb.com/wjsmro/content/3/1
World J Surg Med Radiat Oncol 2014;3:1-9
PK participated in the design of the publication,
review of the literature and data analysis
AP participated in the design of the publication,
review of the literature and data analysis
All authors read and approved the final
manuscript.
References
1.
2.
3.
4.
5.
6.
7.
8.
9.
6
Bakkelund KE, Nordrum IS, Fossmark R,
Waldum HL.: Gastric carcinomas localized
to the cardia. Gastroenterol Res Pract. 2012;
2012:457831. [pubmed]
Corso G, Marrelli D, Roviello F.: Familial
gastric cancer and germline mutations of Ecadherin. Ann Ital Chir. 2012; 83(3): 17782.[pubmed]
Parkin DM, Bray F, Ferlay J, Pisani P.: Global
cancer statistics, 2002. CA Cancer J Clin.
2005; 55(2): 74-108.[pubmed]
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(2): 128-36.[pubmed]
Corso G, Marrelli D, Pascale V, Vindigni C,
Roviello F.: Frequency of CDH1 germline
mutations in gastric carcinoma coming from
high- and low-risk areas: metanalysis and
systematic review of the literature. BMC
Cancer. 2012; 12: 8. [pubmed]
Jang BG, Kim WH.: Molecular pathology of
gastric carcinoma. Pathobiology. 2011;
78(6): 302-10.[pubmed]
SokoloV B.: Predisposition to cancer in the
Bonaparte family. Am J Surg 1938: 40: 637–
638?
Schrader K, Huntsman D.: Hereditary
diffuse gastric cancer. Cancer Treat Res.
2010; 155: 33-63.[pubmed]
Park SH, Song CW, Kim YB, Kim YS, Chun
HR, Lee JH, Seol WJ, Yoon HS, Lee MK, Lee
JH, Bhang CS, Park JH, Park YH, Do BH, Park
YD, Yoon SJ, Park CW, Kim JP, Choi JH, Shin
KC,
Park
SM.:
Clinicopathologic
characteristics of superficial gastric cancer
diagnosed at primary health care
institutions in 2011. Korean J Gastroenterol.
2012; 60(5): 285-91. [pubmed]
Tomasz B et al.
10. Pharoah PD, Guilford P, Caldas C;
International Gastric Cancer Linkage
Consortium.: Incidence of gastric cancer and
breast cancer in CDH1 (E-cadherin)
mutation carriers from hereditary diffuse
gastric cancer families. Gastroenterology.
2001; 121(6): 1348-53.
11. Oliveira MJ, Costa AC, Costa AM, Henriques
L, Suriano G, Atherton JC, Machado JC,
Carneiro F, Seruca R, Mareel M, Leroy A,
Figueiredo C.: Helicobacter pylori induces
gastric epithelial cell invasion in a c-Met and
type IV secretion system-dependent
manner. J Biol Chem. 2006; 281(46): 3488896. [pubmed]
12. Guilford P, Hopkins J, Harraway J, McLeod
M, McLeod N, Harawira P, Taite H, Scoular
R, Miller A, Reeve AE.: E-cadherin germline
mutations in familial gastric cancer. Nature.
1998; 392(6674): 402-5. [pubmed]
13. Grunwald GB.: The structural and functional
analysis of cadherin calcium-dependent cell
adhesion molecules. Curr Opin Cell Biol.
1993; 5(5): 797-805. [pubmed]
14. Oliveira C, Seruca R, Carneiro F.: Hereditary
gastric cancer. Best Pract Res Clin
Gastroenterol. 2009; 23(2): 147-57.
[pubmed]
15. Berx G, Nollet F, van Roy F.: Dysregulation
of the E-cadherin/catenin complex by
irreversible
mutations
in
human
carcinomas. Cell Adhes Commun. 1998; 6(23): 171-84. [pubmed]
16. Bremm A, Walch A, Fuchs M, Mages J,
Duyster J, Keller G, Hermannstädter C,
Becker KF, Rauser S, Langer R, von Weyhern
CH, Höfler H, Luber B.: Enhanced activation
of epidermal growth factor receptor caused
by tumor-derived E-cadherin mutations.
Cancer Res. 2008; 68(3): 707-14.
17. Majewski IJ, Kluijt I, Cats A, Scerri TS, de
Jong D, Kluin RJ, Hansford S, Hogervorst FB,
Bosma AJ, Hofland I, Winter M, Huntsman D,
Jonkers J, Bahlo M, Bernards R.: An alpha-Ecatenin (CTNNA1) mutation in hereditary
diffuse gastric cancer. J Pathol. 2012. doi:
10.1002/path.4152. [Epub ahead of print]
[pubmed]
18. Hayden JD, Cawkwell L, Quirke P, Dixon MF,
Goldstone AR, Sue-Ling H, Johnston D,
Martin IG.: Prognostic significance of
microsatellite instability in patients with
gastric carcinoma. Eur J Cancer. 1997;
33(14): 2342-6.
http://www.npplweb.com/wjsmro/content/3/1
World J Surg Med Radiat Oncol 2014;3:1-9
19. Carneiro F.: Hereditary gastric cancer.
Pathologe. 2012; 33 Suppl 2: 231-4.
20. Kim JH, Shin HS, Lee SH, Lee I, Lee YS, Park
JC, Kim YJ, Chung JB, Lee YC.: Contrasting
activity of Hedgehog and Wnt pathways
according
to
gastric
cancer
cell
differentiation: relevance of crosstalk
mechanisms. Cancer Sci. 2010; 101(2): 32835.[pubmed]
21. Omori Y, Nakayama F, Li D, Kanemitsu K,
Semba S, Ito A, Yokozaki H.: Alternative
lengthening of telomeres frequently occurs
in mismatch repair system-deficient gastric
carcinoma. Cancer Sci. 2009; 100(3): 413-8.
[pubmed]
22. Wu MS, Chang MC, Huang SP, Tseng CC,
Sheu JC, Lin YW, Shun CT, Lin MT, Lin JT.:
Correlation of histologic subtypes and
replication
error
phenotype
with
comparative genomic hybridization in
gastric cancer. Genes Chromosomes Cancer.
2001; 30(1): 80-6.[pubmed]
23. Falchetti M, Saieva C, Lupi R, Masala G,
Rizzolo P, Zanna I, Ceccarelli K, Sera F,
Mariani-Costantini R, Nesi G, Palli D, Ottini
L.: Gastric cancer with high-level
microsatellite instability: target gene
mutations, clinicopathologic features, and
long-term survival. Hum Pathol. 2008;
39(6): 925-32.
24. dos Santos NR, Seruca R, Constância M,
Seixas
M,
Sobrinho-Simões
M.:
Microsatellite instability at multiple loci in
gastric
carcinoma:
clinicopathologic
implications
and
prognosis.
Gastroenterology. 1996; 110(1): 38-44.
25. Hayden JD, Martin IG, Cawkwell L, Quirke P.:
The role of microsatellite instability in
gastric carcinoma. Gut. 1998; 42(2): 3003.[pubmed]
26. Chun N, Ford JM.: Genetic testing by cancer
site: stomach. Cancer J. 2012; 18(4): 35563[pubmed]
27. Spigelman AD, Williams CB, Talbot IC,
Domizio
P,
Phillips
RK.
Upper
gastrointestinal cancer in patients with
familial adenomatous polyposis. Lancet
1989; 2: 783-785.
28. Zwick A, Munir M, Ryan CK, et al. Gastric
adenocarcinoma and dysplasia in fundic
gland polyps of a patient with attenuated
adenomatous
polyposis
coli.
Gastroenterology
1997;
113:
659663.[pubmed]
7
Hereditary Gastric Cancer
29. Hofgärtner WT, Thorp M, Ramus MW, et al.
Gastric adenocarcinoma associated with
fundic gland polyps in a patient with
attenuated familial adenomatous polyposis.
Am J Gastroenterol 1999; 94: 22752281.[pubmed]
30. Garrean S, Hering J, Saied A, Jani J, Espat NJ.
Gastric adenocarcinoma arising from fundic
gland polyps in a patient with familial
adenomatous polyposis syndrome. Am Surg
2008; 74: 79-83.[pubmed]
31. Park SY, Ryu JK, Park JH, Yoon H, Kim JY,
Yoon YB, Park JG, Lee SH, Kang SB, Park JW,
Oh JH.: Prevalence of gastric and duodenal
polyps and risk factors for duodenal
neoplasm in korean patients with familial
adenomatous polyposis. Gut Liver. 2011;
5(1): 46-51.[pubmed]
32. Lynch HT, Snyder C, Davies JM, Lanspa S,
Lynch J, Gatalica Z, Graeve V, Foster J.: FAP,
gastric cancer, and genetic counseling
featuring children and young adults: a
family study and review. Fam Cancer. 2010;
9(4): 581-8.[pubmed]
33. Worthley DL, Phillips KD, Wayte N,
Schrader KA, Healey S, Kaurah P, Shulkes A,
Grimpen F, Clouston A, Moore D, Cullen D,
Ormonde D, Mounkley D, Wen X, Lindor N,
Carneiro F, Huntsman DG, Chenevix-Trench
G, Suthers GK. Gastric adenocarcinoma and
proximal polyposis of the stomach (GAPPS):
a new autosomal dominant syndrome. Gut.
2012; 61(5): 774-9.
34. Hemminki K, Jiang Y.: Familial myeloid
leukemias from the Swedish Family-Cancer
Database. Leuk Res. 2002; 26(6): 6113.[pubmed]
35. Resende C, Thiel A, Machado JC, Ristimäki
A.:
Gastric
cancer:
basic
aspects.
Helicobacter. 2011; 16 Suppl 1: 3844.[pubmed]
36. Win AK, Lindor NM, Young JP, Macrae FA,
Young GP, Williamson E, Parry S, Goldblatt J,
Lipton L, Winship I, Leggett B, Tucker KM,
Giles GG, Buchanan DD, Clendenning M,
Rosty C, Arnold J, Levine AJ, Haile RW,
Gallinger S, Le Marchand L, Newcomb PA,
Hopper JL, Jenkins MA.: Risks of primary
extracolonic cancers following colorectal
cancer in lynch syndrome. J Natl Cancer
Inst. 2012; 104(18): 1363-72.
37. Levine AJ, Win AK, Buchanan DD, Jenkins
MA, Baron JA, Young JP, Long TI,
Weisenberger DJ, Laird PW, McCall RL,
http://www.npplweb.com/wjsmro/content/3/1
World J Surg Med Radiat Oncol 2014;3:1-9
38.
39.
40.
41.
42.
43.
44.
8
Duggan DJ, Haile RW.Cancer risks for the
relatives of colorectal cancer cases with a
methylated MLH1 promoter region: data
from the Colorectal Cancer Family Registry.
Cancer Prev Res (Phila). 2012; 5(2): 32835.[pubmed]
Pinto M, Wu Y, Mensink RG, Cirnes L, Seruca
R, Hofstra RM.: Somatic mutations in
mismatch repair genes in sporadic gastric
carcinomas are not a cause but a
consequence of the mutator phenotype.
Cancer Genet Cytogenet. 2008; 180(2): 1104.
Fleisher AS, Esteller M, Wang S, Tamura G,
Suzuki H, Yin J, Zou TT, Abraham JM, Kong
D, Smolinski KN, Shi YQ, Rhyu MG, Powell
SM, James SP, Wilson KT, Herman JG,
Meltzer SJ.: Hypermethylation of the hMLH1
gene promoter in human gastric cancers
with microsatellite instability. Cancer Res.
1999; 59(5): 1090-5. [pubmed]
Leung SY, Yuen ST, Chung LP, Chu KM, Chan
AS, Ho JC.: hMLH1 promoter methylation
and lack of hMLH1 expression in sporadic
gastric carcinomas with high-frequency
microsatellite instability. Cancer Res. 1999;
59(1): 159-64. [pubmed]
Peltomäki P, Lothe RA, Aaltonen LA,
Pylkkänen L, Nyström-Lahti M, Seruca R,
David L, Holm R, Ryberg D, Haugen A:
Microsatellite instability is associated with
tumors that characterize the hereditary
non-polyposis
colorectal
carcinoma
syndrome. Cancer Res. 1993; 53(24): 58535. [pubmed]
D'Errico M, de Rinaldis E, Blasi MF, Viti V,
Falchetti M, Calcagnile A, Sera F, Saieva C,
Ottini L, Palli D, Palombo F, Giuliani A,
Dogliotti E.: Genome-wide expression
profile of sporadic gastric cancers with
microsatellite instability. Eur J Cancer.
2009; 45(3): 461-9. [pubmed]
Olivier M, Goldgar DE, Sodha N, Ohgaki H,
Kleihues P, Hainaut P, Eeles RA.: LiFraumeni
and
related
syndromes:
correlation between tumor type, family
structure, and TP53 genotype. Cancer Res.
2003; 63(20): 6643-50. [pubmed]
Yamada H, Shinmura K, Okudela K, Goto M,
Suzuki M, Kuriki K, Tsuneyoshi T, Sugimura
H.: Identification and characterization of a
novel germ line p53 mutation in familial
gastric cancer in the Japanese population.
Tomasz B et al.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
Carcinogenesis. 2007; 28(9): 2013-8.
[pubmed]
Jang BG, Kim WH.: Molecular pathology of
gastric carcinoma. Pathobiology. 2011;
78(6): 302-10.[pubmed]
Jakubowska A, Nej K, Huzarski T, Scott RJ,
Lubiński J.: BRCA2 gene mutations in
families with aggregations of breast and
stomach cancers. Br J Cancer. 2002 7; 87(8):
888-91. [pubmed]
Seevaratnam R, Coburn N, Cardoso R, Dixon
M, Bocicariu A, Helyer L.: A systematic
review of the indications for genetic testing
and prophylactic gastrectomy among
patients with hereditary diffuse gastric
cancer. Gastric Cancer. 2012; 15 Suppl 1:
S153-63. [pubmed]
Onitilo AA, Aryal G, Engel JM.: Hereditary
Diffuse Gastric Cancer: A Family Diagnosis
and Treatment. Clin Med Res. 2012 [Epub
ahead of print][pubmed]
Schrader K, Huntsman D.: Hereditary
diffuse gastric cancer. Cancer Treat Res.
2010; 155: 33-63.[pubmed]
Chun N, Ford JM.: Genetic testing by cancer
site: stomach. Cancer J. 2012; 18(4): 355-63.
[pubmed]
Vasas P, Bijendra P.: Genetic background
and clinical features of hereditary diffuse
gastric cancer. Orv Hetil. 2011; 152(28):
1105-9.[pubmed]
Pharoah PD, Guilford P, Caldas C;
International Gastric Cancer Linkage
Consortium.: Incidence of gastric cancer and
breast cancer in CDH1 (E-cadherin)
mutation carriers from hereditary diffuse
gastric cancer families. Gastroenterology.
2001; 121(6): 1348-53.
Caldas C, Carneiro F, Lynch HT, Yokota J,
Wiesner GL, Powell SM, Lewis FR,
Huntsman DG, Pharoah PD, Jankowski JA,
MacLeod P, Vogelsang H, Keller G, Park KG,
Richards FM, Maher ER, Gayther SA,
Oliveira C, Grehan N, Wight D, Seruca R,
Roviello F, Ponder BA, Jackson CE.: Familial
gastric cancer: overview and guidelines for
management. J Med Genet. 1999; 36(12):
873-80. [pubmed]
APC-Associated
Polyposis
Conditions.
Authors: Jasperson KW, Burt RW. Source:
GeneReviews™ [Internet]. Seattle (WA):
University of Washington, Seattle; 1993-.
1998 Dec 18
http://www.npplweb.com/wjsmro/content/3/1
World J Surg Med Radiat Oncol 2014;3:1-9
55. Shikata K, Doi Y, Yonemoto K, Arima H,
Ninomiya T, Kubo M, Tanizaki Y, Matsumoto
T, Iida M, Kiyohara Y.: Population-based
prospective study of the combined influence
of cigarette smoking and Helicobacter
pylori infection on gastric cancer incidence:
the Hisayama Study. Am J Epidemiol. 2008;
168(12): 1409-15.
56. Jenab M, Riboli E, Ferrari P, Friesen M,
Sabate J, Norat T, Slimani N, Tjønneland A,
Olsen A, Overvad K, Boutron-Ruault MC,
Clavel-Chapelon F, Boeing H, Schulz M,
Linseisen J, Nagel G, Trichopoulou A, Naska
A, Oikonomou E, Berrino F, Panico S, Palli D,
Sacerdote C, Tumino R, Peeters PH, Numans
ME, Bueno-de-Mesquita HB, Büchner FL,
Lund E, Pera G, Chirlaque MD, Sánchez MJ,
Arriola L, Barricarte A, Quirós JR, Johansson
I, Johansson A, Berglund G, Bingham S,
Khaw KT, Allen N, Key T, Carneiro F, Save V,
Del Giudice G, Plebani M, Kaaks R, Gonzalez
CA.: Plasma and dietary carotenoid, retinol
and tocopherol levels and the risk of gastric
adenocarcinomas
in
the
European
prospective investigation into cancer and
nutrition. Br J Cancer. 2006; 95(3): 406-15.
57. Mendez AM: Cereal Wber intake may reduce
risk of gastric adenocarcinomas: the EPICEURGAST study. Int J Cancer 2007; 121(7):
1618–23.[pubmed]
Hereditary Gastric Cancer
58. Yuasa Y, Nagasaki H, Akiyama Y, Hashimoto
Y, Takizawa T, Kojima K, Kawano T,
Sugihara K, Imai K, Nakachi K.: DNA
methylation status is inversely correlated
with green tea intake and physical activity
in gastric cancer patients. Int J Cancer.
2009; 124(11): 2677-82.[pubmed]
59. Charalabopoulos K, Kotsalos A, Batistatou A,
Charalabopoulos A, Peschos D, Vezyraki P,
Kalfakakou V, Metsios A, Charalampopoulos
A, Macheras A, Agnantis N, Evangelou A.:
Serum and tissue selenium levels in gastric
cancer patients and correlation with CEA.
Anticancer Res. 2009; 29(8): 34657.[pubmed]
60. Steevens J, van den Brandt PA, Goldbohm
RA, Schouten LJ.: Selenium status and the
risk of esophageal and gastric cancer
subtypes: the Netherlands cohort study.
Gastroenterology. 2010; 138(5): 1704-13.
61. Jang BG, Kim WH.: Molecular pathology of
gastric carcinoma. Pathobiology. 2011;
78(6): 302-10.[pubmed]
62. Kowalska E, Narod SA, Huzarski T, Zajaczek
S, Huzarska J, Gorski B, Lubinski J.:
Increased rates of chromosome breakage in
BRCA1 carriers are normalized by oral
selenium
supplementation.
Cancer
Epidemiol
Biomarkers
Prev.
2005;
14(5):1302-6.[pubmed]
World Journal of Surgical, Medical
and Radiation Oncology
Published by Narain Publishers Pvt. Ltd. (NPPL)
The Open Access publishers of peer reviewed
journals. All articles are immediately published
online on acceptance.
All articles published by NPPL are available
free online
Authors retain the copyright under the
Creative commons attribution license.
The license permits unrestricted use,
distribution, and reproduction in any medium,
provided the original work is properly cited
9
http://www.npplweb.com/wjsmro/content/3/1