You are on page 1of 11

REVIEW ARTICLE

Screening and surveillance for gastric cancer in the United


States: Is it needed?
Gwang Ha Kim, MD, PhD,1 Peter S. Liang, MD,2 Sung Jo Bang, MD,3 Joo Ha Hwang, MD, PhD2
Busan, Ulsan, Korea; Seattle, Washington, USA

Background and Aims: Although the incidence of gastric cancer in the United States is relatively low, the
incidence of gastric cancer is higher than for esophageal cancer, for which clear guidelines for screening and sur-
veillance exist. With the increasing availability of endoscopic therapy, such as endoscopic submucosal dissection,
for treating advanced dysplasia and early gastric cancer, establishing guidelines for screening and surveillance of
patients who are at high risk of developing gastric cancer has the potential to diagnose and treat gastric cancer at
an earlier stage and improve mortality from gastric cancer. The aims of this article were to review the data
regarding the risk factors for developing gastric cancer, methods for gastric cancer screening, and results of
national screening programs.
Methods: A review of the existing literature related to the aims was performed.
Results: Risk factors for gastric cancer that were identified include race/ethnicity (East Asian, Russian, or South
American), first-degree relative diagnosed with gastric cancer, positive Helicobacter pylori status, and presence of
atrophic gastritis or intestinal metaplasia. Endoscopy has the highest rate of detecting gastric cancer compared
with other gastric cancer screening methods. The national screening program in Japan has demonstrated a
mortality reduction from gastric cancer based on cohort data.
Conclusions: Gastric cancer screening with endoscopy should be considered in individuals who are immigrants
from regions associated with a high risk of gastric cancer (East Asia, Russia, or South America) or who have a
family history of gastric cancer. Those with findings of atrophic gastritis or intestinal metaplasia on screening
endoscopy should undergo surveillance endoscopy every 1 to 2 years. Large prospective multicenter studies
are needed to further identify additional risk factors for developing gastric cancer and to assess whether gastric
cancer screening programs for high-risk populations in the United States would result in improved mortality.
(Gastrointest Endosc 2016;84:18-28.)

Gastric cancer is one of the world’s leading causes of cancers) occurred globally in 2012, making it the fifth most-
morbidity and mortality from malignant disease. An estimated common malignancy in the world, after lung, breast, colo-
1 million cases of gastric cancer (952,000 cases; 6.8% of all rectal, and prostate cancers.1,2 More than 70% of gastric
cancer cases occur in the developing world, and approxi-
Abbreviations: AG, atrophic gastritis; CI, confidence interval; EGC, early
mately 50% occur in East Asia.3 Gastric cancer is less
gastric cancer; IM, intestinal metaplasia; OR, odds ratio; PG, pepsin- common in the United States, with the incidence of gastric
ogen; UGI, upper GI. cancer among males and females in the United States at
DISCLOSURE: All authors disclosed no financial relationships relevant 12.3 and 6.0 per 100,000/year, respectively; however, there
to this publication. is a disproportionally higher incidence in Asians.4 The
estimated number of new cases of gastric cancer in the
United States in 2015 was 24,590 (1.5% of all cancers) and
Use your mobile device to scan this 10,720 deaths (1.8% of all cancer deaths).5 It should be
QR code and watch the author in- noted that the estimated number of new cases of
terview. Download a free QR code esophageal cancer in the United States for 2015 was 16,980
scanner by searching “QR Scanner” (1.0% of all cancers), which is less than the number of new
in your mobile device’s app store. cases of gastric cancer. Guidelines for esophageal cancer
Copyright ª 2016 by the American Society for Gastrointestinal Endoscopy
screening and surveillance of Barrett’s esophagus exist;
0016-5107/$36.00 however, guidelines for gastric cancer screening and
http://dx.doi.org/10.1016/j.gie.2016.02.028 surveillance of gastric intestinal metaplasia (IM) are lacking.

18 GASTROINTESTINAL ENDOSCOPY Volume 84, No. 1 : 2016 www.giejournal.org


Kim et al Screening and surveillance for gastric cancer

TABLE 1. Main risk factors for gastric cancer according to the tumor location

Risk estimates (95% CI)


Noncardia cancer Cardia cancer Reference

H pylori infection RR 2.97 (2.34-3.77) RR .99 (.72-1.35) Helicobacter and Cancer Collaborative Group23
Cigarette smoking RR 1.60 (1.41-1.80) RR 1.87 (1.31-2.67) Ladeiras-Lopes et al97
Alcohol RR 1.07 (.91-1.26) RR .94 (.78-1.13) Tramacere et al98
Obesity (BMI > 30) RR 1.26 (.89-1.78) RR 2.06 (1.63-2.61) Yang et al99
Vegetables RR .75 (.59-.95) RR .63 (.50-.79) Lunet et al.100
Fruit RR .61 (.44-.84) RR .58 (.38-.89) Lunet et al.100
High salt intake OR 2.05 (1.60-2.62) Ge et al.101
Family history of gastric cancer OR 2.82 (1.83-4.46) Shin et al.27
Intestinal metaplasia RR 6.4 (2.6-16.1)* Uemura et al.37
CI, Confidence interval; RR, relative risk; BMI, body mass index; OR, odds ratio.
*In H pylori–positive individuals.

Since the 1970s there have been notable improvements region or in the distal noncardia region. Advanced age,
in the 5-year relative survival rates for gastric cancer in the male sex, smoking, and family history are common risk
United States, from 15% in 1975 to 29% in 2009.6 However, factors for both cardia and noncardia cancers. In terms of
these low survival rates suggest that most cases (over 65%) race/ethnicity, whites tend to develop cardia cancer,
are still diagnosed at an advanced stage.5 The overall 5-year whereas Hispanics and Asians tend to develop noncardia
relative survival rate is about 20% in most parts of the cancer. Helicobacter pylori infection and dietary factors,
world, but in Japan and Korea 5-year survival rates above such as high intake of salt, increase the risk of noncardia
70% for stage I and II gastric cancer have been re- cancer. On the other hand, obesity and GERD are mainly
ported.7-9 A multicenter retrospective study of 2191 associated with cancers arising from the cardia. The
patients with gastric cancer undergoing surgical resection estimates of risk factors for gastric cancer are summarized
showed that early gastric cancer (EGC) comprised approx- in Table 1.
imately 20% of all surgically resected cancers in North
America compared with 50% of resected cancers in Age
Japan.10 Although these differences can be explained by The incidence of gastric cancer increases with age. Be-
multiple factors, one of most plausible reasons is the tween 2007 and 2011 only 6% of cases occurred in individ-
implementation of a screening program for detection of uals younger than age 45 in the United States, whereas
EGC in Japan and Korea. approximately 70% of cases were diagnosed in individuals
Identification of risk factors involved in carcinogenesis aged 55 to 84.
and interventions to address these risk factors may
reduce the incidence of gastric cancer. Reducing gastric
Sex
cancer mortality also requires early identification of
Compared with women, men have a higher risk of both
patients who are at high risk for gastric cancer and
cardia (5-fold) and noncardia (2-fold) gastric cancer.11 The
management strategies to slow or prevent progression
reason for this difference is unclear, but environmental or
of gastric cancer. It is likely to be more cost-effective to
occupational exposures may play a role. Men have
detect and treat early-stage gastric cancer with endo-
historically tended to smoke more than women, whereas
scopic resection rather than surgical resection. In this
estrogens may protect against the development of gastric
review we discuss risk factors for gastric cancer, define
cancer.12 Delayed menopause and increased fertility may
high-risk groups, and summarize current guidelines for
lower the risk of gastric cancer, whereas antiestrogen
screening and surveillance strategies for these high-risk
drugs such as tamoxifen may increase the rates of gastric
groups. Finally, we propose an algorithm for screening
cancer.13,14 These hormones may protect against gastric
and surveillance of individuals who are at high risk for
cancer among women of childbearing age, but their effect
gastric cancer in the United States.
is diminished after menopause. Consequently, women
develop gastric cancer in a manner similar to men, albeit
RISK FACTORS FOR GASTRIC CANCER with a 10- to 15-year lag period.15

Gastric cancer is a multifactorial disease involving both Race/ethnicity


genetic and environmental risk factors. Its risk factors differ There is significant variability in the gastric cancer inci-
depending on whether cancers arise in the proximal cardia dence across different ethnicities in the United States.

www.giejournal.org Volume 84, No. 1 : 2016 GASTROINTESTINAL ENDOSCOPY 19


Screening and surveillance for gastric cancer Kim et al

450

400

350
Incidence rates (per 100,000/year)

300
White
250 African American
Asian/Pacific Islander
American Indian/Alaska Native
200 Hispanic
Native Korean
Native Japanese
150

100

50

0
15-19
5-19220-24
0-24225-29 30-34 35-39
5-39440-44
0-44445-49 50-54
0-54555-59
5-59660-64
0-64665-69
5-69770-74
0-74775-79
5-79880-84 >85
Age (years)

Figure 1. Incidence of gastric cancers according to age and race in the United States, Japan, and Korea.5,41,42

Data from the 2002 to 2006 Surveillance, Epidemiology, Helicobacter pylori


and End Results registries show that the incidence of H pylori infection triggers a series of inflammatory
gastric cancer among whites (men, 10.7 per 100,000/year, reactions, which are considered an important cause of
and women, 5.0 per 100,000/year) is approximately chronic gastritis. Progression from chronic gastritis to
half that of Asians/Pacific Islanders (men, 20.8 per gastric atrophy and IM is an early step of mucosal changes
100,000/year, and women, 11.7 per 100,000/year), African in the stomach leading to dysplasia and ultimately cancer.19
Americans (men, 18.4 per 100,000/year, and women, 9.2 H pylori has been classified as a World Health Organization
per 100,000/year), and Hispanics (men, 17.1 per 100,000/ Class I carcinogen since 1994 because several studies have
year, and women, 10.0 per 100,000/year) for both men demonstrated an association between H pylori infection
and women (Fig. 1).4,5 Among Asian American subgroups, and development of gastric cancer, especially intestinal-
Korean and Japanese Americans have an especially high type noncardia gastric cancer.20 Gastric cancer develops
incidence rate (Fig. 2).16 In a study evaluating the effect in approximately 1% of H pylori–infected subjects21;
of immigration on the incidence of gastric cancer among conversely, more than 90% of patients with gastric cancer
Japanese in Hawaii, first-generation participants had high have had current or past H pylori infection.22 In a
rates of gastric cancer17; however, after 2 generations, pooled analysis of 12 prospective studies, which included
gastric cancer rates among Japanese Americans had 762 patients and 2250 control subjects, the odds ratio
decreased to a level that was similar to those of (OR) of H pylori infection for noncardia cancer was 2.97
Americans of European ancestry. (95% confidence interval [CI], 2.34-3.77).23 On the other
By anatomic site, noncardia gastric cancer is approxi- hand, the OR of H pylori infection for cardia cancer was
mately half as common in whites compared with other .99 (95% CI, .40-1.77) in an analysis of 274 patients and
ethnic groups,18 whereas cardia gastric cancer is 827 control subjects.23 When only cases occurring at least
approximately twice as common.11 The risk of noncardia 10 years after the blood draw used for H pylori diagnosis
gastric cancer in the United States is highest among were included in the pooled analysis, the OR increased
Asians/Pacific Islanders, African Americans, and Hispanics to 5.93 (95% CI, 3.41-10.3) for noncardia cancer but
and is least common in whites.6 decreased to .46 (95% CI, .23-.90) for cardia cancer.

20 GASTROINTESTINAL ENDOSCOPY Volume 84, No. 1 : 2016 www.giejournal.org


Kim et al Screening and surveillance for gastric cancer

60

50
Incidence rates (per 100,000/year)

40

30 Male
Female

20

10

0
All races Non-Hispanic African Hispanic Asian/Pacific Japanese Korean
White American Islander American American

Figure 2. Age-adjusted incidence rate of gastric cancer by race/ethnicity in the United States.4,16

Family history of gastric cancer TABLE 2. Hereditary syndromes associated with increased risk for
Having a first-degree relative with gastric cancer is asso- gastric cancer
ciated with an OR of 2 to 10 for developing gastric cancer,
Gastric
depending on race.24 Two Western studies showed that a Hereditary syndromes Genes cancer risk
first-degree family history of gastric cancer increased the
risk by 2.6- to 3.5-fold, with a calculated attributable risk Hereditary diffuse gastric CGH1 >80%
cancer
of 8%.25,26 A Korean study examining subjects with first-
degree relatives with gastric cancer found a comparable Lynch syndrome MLH1, MSH2, MSH6, 6%-13%
PMS2, Epcam
adjusted OR of 2.85 (95% CI, 1.83-4.46).27 In addition, in
a meta-analysis studying H pylori infection and gastric Hereditary breast/ovarian BRCA1/2 2.6%-5.5%
cancer syndrome
histology in the first-degree relatives of gastric cancer pa-
tients, individuals with a family history of gastric cancer Li-Fraumeni syndrome p53 2.8%
were approximately twice as likely to have H pylori infec- Familial adenomatous APC .5%-2%
tion, atrophic gastritis (AG), and IM.28 polyposis syndrome
Up to 1% to 3% of gastric cancer cases are thought to be Juvenile polyposis syndrome SMAD4, BMPR1A 21%
related to hereditary syndromes (Table 2).29 Hereditary Peutz-Jeghers syndrome STK11 29%
diffuse gastric cancer is a rare, autosomal dominant,
genetic syndrome characterized by the early onset of
diffuse gastric adenocarcinoma (ie, before age 40), an as hereditary breast and ovarian cancer, Li-Fraumeni, famil-
increased risk of lobular breast cancer and signet-ring cell ial adenomatous polyposis, Peutz-Jeghers, and juvenile
colorectal cancer, and a poor prognosis.30,31 This syndrome polyposis are also associated with an increased risk for
is caused by a germline mutation in the CDH1 gene on chro- gastric cancer.33
mosome 16q22, which encodes for E-cadherin. In hereditary
diffuse gastric cancer families, carriage of the abnormal Atrophic gastritis and intestinal metaplasia
E-cadherin gene confers a greater than 80% lifetime risk of AG and IM are considered precursor conditions of
developing gastric cancer; therefore, carriers of the CDH1 gastric cancer, and both are strongly associated with H
mutation are recommended to undergo prophylactic total pylori infection.19 A nationwide cohort study in the
gastrectomy or endoscopic screening.31 Netherlands reported that the risk of gastric cancer
Gastric cancer risk is also increased in patients with increased in a stepwise manner according to the severity
Lynch syndrome, with affected individuals carrying a 10% of premalignant gastric lesions. Within 5 years of
lifetime risk.29 Most gastric cancers (90%) that develop in diagnosis, the annual incidence of gastric cancer was
patients with Lynch syndrome are intestinal-type cancers .1% for patients with AG, .25% for IM, .6% for mild-to-
and have the same natural history as sporadic intestinal- moderate dysplasia, and 6% for severe dysplasia.34
type gastric cancer.32 Other hereditary syndromes such Although the risk of gastric cancer in individuals with AG

www.giejournal.org Volume 84, No. 1 : 2016 GASTROINTESTINAL ENDOSCOPY 21


Screening and surveillance for gastric cancer Kim et al

depends on the severity of AG, the adjusted relative risk of of patients with risk factors among practicing gastroenter-
gastric cancer in patients with severe AG was 5.76.35 ologists, particularly in Western countries.43 Because of
Gastric cancer incidence in IM patients was reported to the low incidence of gastric cancer in the United States,
range from 0% to 10% in a systematic review,36 which may endoscopic screening is not currently recommended.
be because of the variation in sample size and follow-up However, screening and surveillance strategies are well
period in the included studies. A nationwide, histology- established in the United States for Barrett’s esophagus,
based Dutch study including 61,707 patients with IM which likely has a lower rate of progression to
showed that the cumulative 10-year incidence of gastric adenocarcinoma than do the aforementioned risk factors
cancer was 1.8% (.18% yearly),34 and a large Japanese for progression to gastric cancer.43 Because endoscopic
study of 1246 H pylori–infected individuals with IM resection techniques such as EMR and endoscopic
with a mean follow-up period of 7.8 years showed that submucosal dissection are becoming increasingly
the relative risk of progression to gastric cancer was 6.4 available in the United States, many EGCs can be
(95% CI, 2.6-16.1).37 The progression from gastric IM to endoscopically resected without the need for surgery.
gastric cancer is highly associated with the histologic Given these developments, a change in our approach to
subtype of IM. IM can be subclassified as complete type managing individuals at high risk for developing gastric
or incomplete type. Complete-type IM is characterized cancer is needed. Because a high-risk patient population
by the presence of a small intestinal-type mucosal pheno- exists and endoscopic screening can further risk-stratify
type with goblet cells containing sialomucin interspersed patients, the establishment of a screening and surveillance
among absorptive cells and with a well-defined brush protocol for high-risk individuals is warranted.
border, whereas incomplete-type IM is characterized by a The optimal age to start screening for gastric cancer
colonic-type mucosal phenotype with tortuous crypts lined is not clear. It has been reported that it would take
by tall columnar cells containing abundant sulfomucin. 44 months for early-stage gastric cancer to develop to
Incomplete type is considered to be a more advanced advanced-staged disease.44 Because the incidence of
stage of IM and has a higher risk of progressing to gastric gastric cancer increases sharply after age 40, it has been
cancer.38 In a pooled analysis, 13 of 14 cross-sectional suggested that individuals older than 40 may undergo
studies and 6 of 10 cohort studies reported significantly screening in high-incidence countries such as Japan and Ko-
higher gastric cancer prevalence in the patients with rea.45 However, based on evolving data demonstrating a
incomplete-type IM than complete-type IM, with a relative decreasing incidence of gastric cancer in individuals
risk of 4 to 11.37,39 Therefore, patients found to have between ages 40 and 49 years in Japan, the new national
incomplete-type IM should undergo surveillance for gastric guidelines in Japan now recommend that screening be
cancer. started at age 50. Other authors outside Korea also
recommend screening commencement after age 50.46
Other risk factors Given the incidence of gastric cancer by age and the
Other factors such as cigarette smoking, alcohol, incidence threshold used to recommend colorectal cancer
obesity, fruit/vegetable consumption, and salt intake have screening in the United States (w50 per 100,000),
also been investigated. Their effects appear to be modest endoscopic screening for gastric cancer in asymptomatic
compared with the risk factors discussed. Risk estimates adults from high-risk populations should be considered
are summarized in Table 1. starting at age 50. A study evaluating the cost-effectiveness
of screening the general population for upper GI (UGI) can-
Screening for gastric cancer: who and when cers including EGC in the United States by performing an
Screening can be performed in the general population upper endoscopy at the time of screening colonoscopy
(mass screening) or only for individuals identified to showed that the incremental cost-effectiveness ratio for
have an increased risk for developing gastric cancer. this intervention was $95,559 per quality-adjusted life year
Although the effectiveness of mass screening for gastric saved; this is comparable with published incremental cost-
cancer still remains controversial,40 it has been effectiveness ratios for other cancer screening interventions
undertaken in Korea and Japan, where there is a high that are commonly performed in the United States.47
incidence of the disease.41,42 On the other hand, in Therefore, a screening program targeting a smaller high-
countries with a low incidence of gastric cancer, such as risk population should be substantially more cost-effective.
the United States, mass screening would not be cost-
effective, and only individuals identified to be at high risk METHODS FOR GASTRIC CANCER SCREENING
for gastric cancer should be considered for screening.
Abundant evidence shows that H pylori infection, family Screening for gastric cancer generally involves 4
history of gastric cancer, and AG/IM are associated with an methods: UGI series, serum pepsinogen (PG) testing, H
increased risk of gastric cancer, and therefore individuals pylori serology, and endoscopy. The ideal screening test
with these risk factors could be considered high risk. How- should be simple, safe, validated, and tolerable to patients;
ever, there is significant confusion regarding management however, there is no single method that meets all these

22 GASTROINTESTINAL ENDOSCOPY Volume 84, No. 1 : 2016 www.giejournal.org


Kim et al Screening and surveillance for gastric cancer

TABLE 3. Pros and cons of each screening method for gastric cancer

Pros Cons

H pylori serology Noninvasive Very low sensitivity


Does not detect premalignant lesions
Serum pepsinogen testing Noninvasive Optimal cut-off values can be affected by
Acceptable sensitivity and specificity several factors (age, sex, race)
Predicts premalignant lesions Requires endoscopy for confirmation
Upper gastrointestinal series Noninvasive Exposure to radiation
Moderate evidence Requires endoscopy for confirmation
Endoscopy Most accurate Invasive and expensive
Biopsy sampling can be performed Low evidence

criteria. Pros and cons of each screening method are dis- however, the evidence was not considered to be strong
cussed below and summarized in Table 3. enough to incorporate serum PG testing into the
national screening program in Japan.58 The limitations of
using serum PG include variable proposed cut-off values
H pylori serology
for PG I and the PG I/II ratio, lack of sensitivity and speci-
H pylori serology as a screening test for gastric cancer
ficity, variability of PG I and the PG I/II ratio (depending on
is limited by low sensitivity and a failure to detect prema-
age, sex, and race), and scant and inconclusive data outside
lignant lesions. It is often negative in the presence of long-
of Japan.
standing AG/IM. Even though H pylori virulence factors
Combining H pylori serology and serum PG testing may
such as Cag A, Vac A, and Bab A may increase its sensitivity
provide additional information for predicting the develop-
for gastric cancer detection, their sensitivity is still low.48
ment of gastric cancer. It has been reported that a combi-
Therefore, H pylori serology is not useful as a stand-
nation of low PG I or PG I/II ratio with negative H pylori
alone screening test.
serology antibodies suggests the highest risk for gastric
cancer,53 because it may indicate extensive atrophy
Serum PG testing where the H pylori burden in the stomach is reduced.
PG is a precursor of pepsin that is produced in the
gastric mucosa. PG is classified biochemically and immuno-
logically into 2 different isozymes, PG I and PG II. PG I is
UGI series
The barium meal indirect radiograph examination was
produced by chief cells in the gastric fundus and body,
introduced as a mass screening program for gastric cancer
and PG II is produced by the cells throughout the entire
in the 1960s in Japan.59 In a meta-analysis including 5
stomach.49,50 A decrease in serum PG I level and PG I/II
case-control studies and 2 cohort studies, gastric cancer
ratio is associated with the extent of gastric atrophy and
screening using UGI series was reported to be useful,60
reduction in gastric acid secretion ability. Stepwise
but the evidence is weak.61 The sensitivity of UGI series
decrease in the PG I/II ratio is closely correlated with the
ranged from 60% to 80%, whereas the specificity and true
progression of gastric atrophy.51,52 Therefore, serum PG
positive rate were 90% and .7% to 2.0%, respectively.62
level has been suggested to be a useful marker of AG,
Case-control studies conducted in Japan showed that
which in turn is a precursor of intestinal-type adenocarci-
screening by UGI series resulted in a 40% to 60% reduction
noma in the stomach.
in gastric cancer mortality, which is the primary reason
To detect gastric cancer, serum PG testing, followed by
this remains an accepted method for the national gastric
endoscopic examination, has been introduced in mass
cancer screening programs in Japan and Korea.63,64
screenings of high-risk patients for gastric cancer. PG I 
70 ng/L and PG I/II ratio  3.0 are associated with an
increased risk for gastric cancer.53-55 In a pooled meta- Endoscopy
analysis assessing approximately 300,000 people, the sensi- Endoscopy is the only method available for direct visual
tivity and specificity of serum PG testing for gastric cancer examination of the gastric mucosa, and it allows for biopsy
screening were 77% and 73%, respectively.56 In a case- sampling so that histologic evaluation can be performed.
control study using serum PG level for gastric cancer Endoscopy is the criterion standard test for diagnosing
screening, the ORs for death from gastric cancer among gastric cancer because of its high detection rate. Although
control subjects screened within 1 and 2 years were .24 UGI series was the initial tool for mass screening of gastric
(95% CI, .06-.93) and .38 (95% CI, .16-.91), respectively.57 cancer, a cohort analysis demonstrated that endoscopy
These results suggest that gastric cancer screening using resulted in a 3- to 5-fold higher detection rate for
serum PG level may decrease gastric cancer mortality; EGC compared with UGI series and was also more

www.giejournal.org Volume 84, No. 1 : 2016 GASTROINTESTINAL ENDOSCOPY 23


Screening and surveillance for gastric cancer Kim et al

cost-effective.65,66 The sensitivity of endoscopy for identi- TABLE 4. Suggested intervals of surveillance endoscopy in individuals
fying gastric cancer is reported to be 78% to 84%.67,68 In at high risk for gastric cancer
Japan and Korea, endoscopy has become the primary
Country/ Indication for Surveillance Reference and
method for gastric cancer screening given its superior
region surveillance interval (y) year
test characteristics, availability, and affordability (EGD costs
approximately U.S.$40 in Korea).66 However, the use of Australia IM 1-3 Busuttil et al,38
2009
endoscopy for gastric cancer screening in the United
States does have several potential limitations, such as United Extensive AG/IM* 3 Correa et al,92
States 2010
the need for additional trained endoscopists to meet
the increased demand, potential adverse events of Italy High-risk IMy 1 Zullo et al,90
2012
endoscopy, patient acceptance, and cost.62
Several studies suggest that advanced endoscopic imag- Low-risk IM 2-3
ing modalities such as chromoendoscopy or narrow-band Europe Extensive AG/IM 1 Dinis-Ribeiro
imaging can increase accuracy for diagnosing gastric et al,91 2012
neoplasia compared with standard white light endoscopy. Korea AG/IM or family history of 1 Yoon et al,82
Chromoendoscopy using indigo carmine and acetic acid gastric cancer 2015
has been demonstrated to clarify subtle mucosal irregular- IM, Intestinal metaplasia; AG, atrophic gastritis.
*Definition of extensive AG: serum PG I level < 70 mg/L and a PG I/PG II ratio < 3;
ities and to delineate the lateral extent of EGCs.69,70 Meth- definition of extensive IM: (1) IM present in at least 2 gastric locations or (2) moderate
ylene blue magnification chromoendoscopy was shown or marked IM in at least 2 biopsy specimens.
to identify IM and intestinal dysplasia with sensitivities of y(1) IM extension > 20%, (2) the presence of incomplete type IM, (3) first-degree
relative of gastric cancer patients, and (4) smokers.
76% and 97% and specificities of 87% and 81%, respec-
tively, and good interobserver agreement (k Z .74).71
Narrow-band imaging and digital-based image enhance- years is now recommended. The proportion of EGC in
ment technologies such as computed virtual chromoendo- Japan is more than 50% of all gastric cancers diagnosed,
scopy have been reported to increase the diagnostic yield whereas in Europe EGC only comprises 15% of all cases.82
and accuracy for the detection of gastric neoplasia; howev- Similarly, in Korea the National Cancer Screening Program
er, imaging criteria are inconsistent between studies and for gastric cancer screening that began in 1999
comparative or validation studies have yet to be per- recommends endoscopy or UGI series for individuals aged
formed.72-77 Furthermore, because of the rapid develop- 40 years and older every 2 years.83 As a result
ment of imaging technologies (including the resolution approximately 46% to 67% of gastric cancers are detected
of white-light endoscopy), it is difficult to make any defin- at an early stage by screening,84 and the 5-year survival
itive conclusions regarding any of the advanced imaging rate has increased from 43% in 1993 to 1995 to 69% in
technologies. A consensus on criteria for characterizing 2006 to 2011; however, reduction in mortality from gastric
mucosal patterns is needed to further study the role of cancer as a result of screening has yet to be demonstrated.41
mucosal enhancement imaging technologies to obtain an These data from Japan and Korea support the effectiveness
accurate assessment of their validity and performance. of screening for gastric cancer in high-risk populations.
Further studies are required to assess the ability of new
imaging technologies to identify gastric IM and EGCs. Screening intervals
Based on the existing evidence and ease of use, it is Few studies have addressed the optimal interval for
reasonable to apply technologies such as narrow-band endoscopic screening for gastric cancer, and no guidelines
imaging or digital-based image enhancement technologies exist. In a prospective study in which population-based
in addition to high-definition white-light endoscopy to screening using endoscopy was performed twice over a
examine the gastric mucosa and target biopsy sampling. 5-year interval in China, which also has a high incidence
of gastric cancer, no reduction in mortality from gastric
cancer was observed.85 On the other hand, in a Japanese
RESULTS OF NATIONAL SCREENING study the 5-year survival rate for patients who had under-
PROGRAMS gone endoscopy within 2 years before the detection of
gastric cancer was significantly higher than that for patients
In Japan, annual gastric cancer screening for all resi- who had not undergone endoscopy within 2 years.86
dents aged 40 years and older with UGI series was initiated However, the survival rates were not different between
in 1983.60 In 2015 the Japanese guidelines were updated to the patients who had undergone endoscopy within 1
allow screening to be performed with either UGI series or year before the detection of gastric cancer and the
endoscopy based on studies from Japan demonstrating a patients who had undergone endoscopy between 1 and
decrease in mortality from gastric cancer when screening 2 years. These results suggest that every 2 years may
is performed with either method.58,78-81 In addition, be an optimal interval for endoscopic screening for
screening starting at age 50 and performed every 2 to 3 gastric cancer. Another Korean study also showed that

24 GASTROINTESTINAL ENDOSCOPY Volume 84, No. 1 : 2016 www.giejournal.org


Kim et al Screening and surveillance for gastric cancer

1st or 2nd generation immigrant from high-incidence region†, No


or No screening
family history of gastric cancer

Yes

EGD at age 50*

HP(-) HP(+)
AG/IM(+) OR
AG/IM(-) AG/IM(-)
FHx(+)
FHx(-) FHx(-)

Eradicate HP If HP(+), eradicate

AG/IM(-) AG/IM(+)
No follow-up EGD in 3-5 yrs EGD every 1-2 yrs

Figure 3. Suggested screening algorithm for gastric cancer in the United States. yEast Asia, Russia, and South America. *If there is a first-degree relative
with gastric cancer, start screening 10 years before the age at diagnosis in the first-degree relative or age 50 (whichever is earlier).
HP, H pylori; FHx, family history of gastric cancer; AG, atrophic gastritis; IM, intestinal metaplasia.

endoscopic screening every 2 years decreased the locations or moderate or marked IM in at least 2 biopsy
incidence of gastric cancer and that endoscopic resection specimens) or incomplete-type IM found on index endos-
could be applied to more patients who underwent EGD copy undergo surveillance endoscopy with mapping or
screening within 2 years.87 serum PG testing at 1 year and then repeat surveillance
Appropriateness of the above-mentioned interval for endoscopy every 3 years if extensive AG/IM or
surveillance endoscopy is not well documented in Western incomplete-type IM persists.
countries. However, 2 cohort studies in England88 and At the present time there are no well-defined guidelines
Italy89 showed that only 36% and 38% of detected gastric regarding screening for gastric cancer or surveillance of
cancers were in an early stage when the endoscopic gastric intestinal metaplasia in the United States. The
surveillance interval was 1 or 2 years, respectively. Based 2015 American Society for Gastrointestinal Endoscopy
on these results, it seems that a 3-year interval follow-up guidelines suggest that surveillance endoscopy be per-
is not appropriate in clinical practice. Therefore, a Euro- formed in patients with gastric intestinal metaplasia who
pean review article proposed that annual endoscopic are at an increased risk of gastric cancer because of ethnic
surveillance would appear justified in all patients with IM background or family history and that surveillance intervals
having at least 1 of following additional risk factors: IM should be individualized.93 Table 4 summarizes previously
extension > 20%, presence of incomplete-type IM, first- proposed intervals for endoscopic surveillance in
degree relative with gastric cancer, and smokers.90 In the individuals at high risk for gastric cancer.
remaining IM patients, surveillance endoscopy with a 2-
to 3-year interval is recommended. According to the
consensus guidelines from the European Gastroenterology PROPOSED ALGORITHM FOR SCREENING AND
Societies, it is recommended that patients with extensive SURVEILLANCE OF GASTRIC CANCER IN THE
AG and/or IM should be offered surveillance endoscopy UNITED STATES
once every 3 years.91
Correa et al92 proposed that U.S. patients with extensive Based on the aforementioned evidence and guidelines,
gastric IM (defined as IM present in at least 2 gastric we propose the following simplified strategy for gastric

www.giejournal.org Volume 84, No. 1 : 2016 GASTROINTESTINAL ENDOSCOPY 25


Screening and surveillance for gastric cancer Kim et al

cancer screening and surveillance of high-risk individuals evaluate the effectiveness of screening and surveillance
(excluding those with hereditary diseases) according to strategies to detect gastric cancer, it would be reasonable
race, H pylori infection status, family history of gastric can- to begin screening individuals who are at high risk for
cer, and AG/IM status (Fig. 3). We recommend endoscopic developing gastric cancer and then to perform surveillance
screening for gastric cancer starting at age 50 for endoscopy at 1- or 2-year intervals if IM is identified on
individuals who are first- or second-generation immigrants screening endoscopy or if the patient has a family history
from high-incidence regions (East Asia, Russia, and South of gastric cancer. Gastric cancer screening in the appro-
America). Individuals with a family history of gastric priate population will likely lead to an increase in the
cancer are recommended to begin endoscopic screening detection of EGCs, which may improve the likelihood of
10 years before diagnosis in the affected relative, similar being able to intervene with endoscopic therapy, such as
to recommendations for colorectal cancer.94 At the time endoscopic submucosal dissection, and reduce mortality
of screening endoscopy, at least 5 nontargeted biopsy from gastric cancer.
specimens should be obtained according to the updated
Sydney System, which includes 2 biopsy samples from REFERENCES
the antrum (lesser and greater curvature), 1 biopsy
sample from the incisura angularis, and 2 biopsy samples 1. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mor-
from the body (lesser and greater curvature).95 Biopsy tality worldwide: sources, methods and major patterns in GLOBOCAN
specimens should be submitted for pathologic evaluation 2012. Int J Cancer 2015;136:E359-86.
2. Ferlay J, Soerjomataram I, Ervik M, et al. GLOBOCAN 2012 v1.0, Cancer
in separate vials labeled by region of the stomach incidence and mortality worldwide: IARC CancerBase No. 11. Lyon,
(antrum, incisura angularis, and body). This biopsy France: International Agency for Research on Cancer; 2013. Available
protocol has been demonstrated to be both sensitive for at: http://globocan.iarc.fr. Accessed March 8, 2015.
the identification of H pylori infection and AG/IM when 3. Ferlay J, Shin HR, Bray F, et al. Estimates of worldwide burden of can-
performed in a population considered to be at high risk cer in 2008: GLOBOCAN 2008. Int J Cancer 2010;127:2893-917.
4. Lui FH, Tuan B, Swenson SL, et al. Ethnic disparities in gastric cancer
for gastric preneoplastic lesions.96 Additional targeted incidence and survival in the USA: an updated analysis of 1992-2009
biopsy samples of visible lesions should also be obtained SEER data. Dig Dis Sci 2014;59:3027-34.
and submitted in separate vials.91 5. Surveillance, Epidemiology, and End Results Program. Cancer query
After screening EGD, those with no H pylori infection, system: SEER Incidence Statistics (2000-2012). Available at: http://
no family history of gastric cancer, and no AG/IM identified seer.cancer.gov/. Accessed March 3, 2015.
6. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin
on screening endoscopy require no further follow-up. 2015;65:5-29.
Those with H pylori infection alone should be treated for 7. Isobe Y, Nashimoto A, Akazawa K, et al. Gastric cancer treatment in
H pylori eradication therapy with confirmation of eradica- Japan: 2008 annual report of the JGCA nationwide registry. Gastric
tion and then undergo endoscopic examination in 3 to 5 Cancer 2011;14:301-16.
years to evaluate for progression to AG/IM. If there is no 8. Jang JS, Shin DG, Cho HM, et al. Differences in the survival of gastric
cancer patients after gastrectomy according to the medical insurance
evidence of progression to AG/IM, then surveillance can status. J Gastric Cancer 2013;13:247-54.
be discontinued because the risk of gastric cancer is low 9. Bollschweiler E, Boettcher K, Hoelscher AH, et al. Is the prognosis for
in the absence of AG/IM. Additionally, those at high risk Japanese and German patients with gastric cancer really different?
(family history of gastric cancer or presence of AG/IM) Cancer 1993;71:2918-25.
should undergo endoscopy every 1 to 2 years with eradica- 10. Noguchi Y, Yoshikawa T, Tsuburaya A, et al. Is gastric carcinoma
different between Japan and the United States? Cancer 2000;89:
tion therapy in patients with H pylori infection. Prospec- 2237-46.
tive studies are needed to assess whether this screening 11. Brown LM, Devesa SS. Epidemiologic trends in esophageal and
and surveillance protocol for gastric cancer in the United gastric cancer in the United States. Surg Oncol Clin North Am
States will produce a survival benefit similar to that 2002;11:235-56.
observed in Korea and Japan. 12. Karimi P, Islami F, Anandasabapathy S, et al. Gastric cancer: descrip-
tive epidemiology, risk factors, screening, and prevention. Cancer
Epidemiol Biomarkers Prev 2014;23:700-13.
13. Derakhshan MH, Liptrot S, Paul J, et al. Oesophageal and gastric
CONCLUSIONS intestinal-type adenocarcinomas show the same male predomi-
nance due to a 17 year delayed development in females. Gut
2009;58:16-23.
Comprehensive U.S. guidelines for gastric cancer
14. Sheh A, Ge Z, Parry NM, et al. 17Beta-estradiol and tamoxifen prevent
screening and surveillance of high-risk individuals are war- gastric cancer by modulating leukocyte recruitment and oncogenic
ranted. The optimal gastric cancer prevention program will pathways in Helicobacter pylori-infected INS-GAS male mice. Cancer
combine risk stratification for screening and surveillance Prev Res (Phila) 2011;4:1426-35.
for high-risk groups. Because race, H pylori infection, fam- 15. Camargo MC, Goto Y, Zabaleta J, et al. Sex hormones, hormonal inter-
ily history of gastric cancer, and AG/IM are significant risk ventions, and gastric cancer risk: a meta-analysis. Cancer Epidemiol
Biomarkers Prev 2012;21:20-38.
factors for gastric cancer, the initial approach should be 16. Miller BA, Chu KC, Hankey BF, et al. Cancer incidence and mortality
to identify individuals with these risk factors. Although patterns among specific Asian and Pacific Islander populations in
large, prospective, multicenter studies are needed to the U.S. Cancer Causes Control 2008;19:227-56.

26 GASTROINTESTINAL ENDOSCOPY Volume 84, No. 1 : 2016 www.giejournal.org


Kim et al Screening and surveillance for gastric cancer

17. Maskarinec G, Noh JJ. The effect of migration on cancer incidence 43. Gomez JM, Wang AY. Gastric intestinal metaplasia and early gastric
among Japanese in Hawaii. Ethn Dis 2004;14:431-9. cancer in the west: a changing paradigm. Gastroenterol Hepatol
18. El-Serag HB, Mason AC, Petersen N, et al. Epidemiological differences 2014;10:369-78.
between adenocarcinoma of the oesophagus and adenocarcinoma 44. You WC, Brown LM, Zhang L, et al. Randomized double-blind factorial
of the gastric cardia in the USA. Gut 2002;50:368-72. trial of three treatments to reduce the prevalence of precancerous
19. Correa P. Human gastric carcinogenesis: a multistep and multifacto- gastric lesions. J Natl Cancer Inst 2006;98:974-83.
rial processdFirst American Cancer Society Award Lecture on Cancer 45. Lin JT. Screening of gastric cancer: who, when, and how. Clin Gastro-
Epidemiology and Prevention. Cancer Res 1992;52:6735-40. enterol Hepatol 2014;12:135-8.
20. Pounder RE, Ng D. The prevalence of Helicobacter pylori infection in 46. Areia M, Carvalho R, Cadime AT, et al. Screening for gastric cancer
different countries. Aliment Pharmacol Ther 1995;9(suppl 2):33-9. and surveillance of premalignant lesions: a systematic review of
21. de Vries AC, Kuipers EJ. Epidemiology of premalignant gastric lesions: cost-effectiveness studies. Helicobacter 2013;18:325-37.
implications for the development of screening and surveillance stra- 47. Gupta N, Bansal A, Wani SB, et al. Endoscopy for upper GI cancer
tegies. Helicobacter 2007;12(suppl 2):22-31. screening in the general population: a cost-utility analysis. Gastroint-
22. Kato S, Matsukura N, Tsukada K, et al. Helicobacter pylori infection- est Endosc 2011;74:610-24.e2.
negative gastric cancer in Japanese hospital patients: incidence and 48. Ley C, Mohar A, Guarner J, et al. Screening markers for chronic atro-
pathological characteristics. Cancer Sci 2007;98:790-4. phic gastritis in Chiapas, Mexico. Cancer Epidemiol Biomarkers Prev
23. Helicobacter and Cancer Collaborative Group. Gastric cancer and 2001;10:107-12.
Helicobacter pylori: a combined analysis of 12 case control studies 49. Samloff IM. Cellular localization of group I pepsinogens in human
nested within prospective cohorts. Gut 2001;49:347-53. gastric mucosa by immunofluorescence. Gastroenterology 1971;61:
24. Yaghoobi M, Bijarchi R, Narod SA. Family history and the risk of 185-8.
gastric cancer. Br J Cancer 2010;102:237-42. 50. Samloff IM, Liebman WM. Cellular localization of the group II pepsin-
25. Palli D, Galli M, Caporaso NE, et al. Family history and risk of stomach ogens in human stomach and duodenum by immunofluorescence.
cancer in Italy. Cancer Epidemiol Biomarkers Prev 1994;3:15-8. Gastroenterology 1973;65:36-42.
26. La Vecchia C, Negri E, Franceschi S, et al. Family history and the risk of 51. Borch K, Axelsson CK, Halgreen H, et al. The ratio of pepsinogen A to
stomach and colorectal cancer. Cancer 1992;70:50-5. pepsinogen C: a sensitive test for atrophic gastritis. Scand J Gastroen-
27. Shin CM, Kim N, Yang HJ, et al. Stomach cancer risk in gastric cancer terol 1989;24:870-6.
relatives: interaction between Helicobacter pylori infection and family 52. Asaka M, Kato M, Kudo M, et al. Relationship between Helicobacter
history of gastric cancer for the risk of stomach cancer. J Clin Gastro- pylori infection, atrophic gastritis and gastric carcinoma in a Japanese
enterol 2010;44:e34-9. population. Eur J Gastroenterol Hepatol 1995;7(suppl 1):S7-10.
28. Rokkas T, Sechopoulos P, Pistiolas D, et al. Helicobacter pylori infec- 53. Watabe H, Mitsushima T, Yamaji Y, et al. Predicting the development
tion and gastric histology in first-degree relatives of gastric cancer of gastric cancer from combining Helicobacter pylori antibodies and
patients: a meta-analysis. Eur J Gastroenterol Hepatol 2010;22: serum pepsinogen status: a prospective endoscopic cohort study.
1128-33. Gut 2005;54:764-8.
29. Lynch HT, Grady W, Suriano G, et al. Gastric cancer: new genetic 54. Yanaoka K, Oka M, Mukoubayashi C, et al. Cancer high-risk subjects
developments. J Surg Oncol 2005;90:114-33; discussion 133. identified by serum pepsinogen tests: outcomes after 10-year
30. Corso G, Marrelli D, Roviello F. Familial gastric cancer and germline follow-up in asymptomatic middle-aged males. Cancer Epidemiol Bio-
mutations of E-cadherin. Ann Ital Chir 2012;83:177-82. markers Prev 2008;17:838-45.
31. Fitzgerald RC, Hardwick R, Huntsman D, et al. Hereditary diffuse 55. Oishi Y, Kiyohara Y, Kubo M, et al. The serum pepsinogen test as a
gastric cancer: updated consensus guidelines for clinical manage- predictor of gastric cancer: the Hisayama study. Am J Epidemiol
ment and directions for future research. J Med Genet 2010;47:436-44. 2006;163:629-37.
32. Aarnio M, Salovaara R, Aaltonen LA, et al. Features of gastric cancer in 56. Dinis-Ribeiro M, da Costa-Pereira A, Lopes C, et al. Validity of serum
hereditary non-polyposis colorectal cancer syndrome. Int J Cancer pepsinogen I/II ratio for the diagnosis of gastric epithelial dysplasia
1997;74:551-5. and intestinal metaplasia during the follow-up of patients at risk
33. Chun N, Ford JM. Genetic testing by cancer site: stomach. Cancer J for intestinal-type gastric adenocarcinoma. Neoplasia 2004;6:449-56.
2012;18:355-63. 57. Yoshihara M, Hiyama T, Yoshida S, et al. Reduction in gastric cancer
34. de Vries AC, van Grieken NC, Looman CW, et al. Gastric cancer risk in mortality by screening based on serum pepsinogen concentration:
patients with premalignant gastric lesions: a nationwide cohort study a case-control study. Scand J Gastroenterol 2007;42:760-4.
in the Netherlands. Gastroenterology 2008;134:945-52. 58. Sugano K. Screening of gastric cancer in Asia. Best Pract Res Clin Gas-
35. Tatsuta M, Iishi H, Nakaizumi A, et al. Fundal atrophic gastritis as a risk troenterol 2015;29:895-905.
factor for gastric cancer. Int J Cancer 1993;53:70-4. 59. Hisamichi S, Sugawara N. Mass screening for gastric cancer by X-ray
36. Rugge M, Correa P, Dixon MF, et al. Gastric dysplasia: the Padova in- examination. Jpn J Clin Oncol 1984;14:211-23.
ternational classification. Am J Surg Pathol 2000;24:167-76. 60. Hamashima C, Shibuya D, Yamazaki H, et al. The Japanese guidelines
37. Uemura N, Okamoto S, Yamamoto S, et al. Helicobacter pylori infec- for gastric cancer screening. Jpn J Clin Oncol 2008;38:259-67.
tion and the development of gastric cancer. N Engl J Med 61. Asaka M, Kato M, Takahashi S, et al. Guidelines for the management
2001;345:784-9. of Helicobacter pylori infection in Japan: 2009 revised edition. Helico-
38. Busuttil RA, Boussioutas A. Intestinal metaplasia: a premalignant bacter 2010;15:1-20.
lesion involved in gastric carcinogenesis. J Gastroenterol Hepatol 62. Kato M, Asaka M. Recent development of gastric cancer prevention.
2009;24:193-201. Jpn J Clin Oncol 2012;42:987-94.
39. Gonzalez CA, Sanz-Anquela JM, Gisbert JP, et al. Utility of subtyping 63. Fukao A, Tsubono Y, Tsuji I, et al. The evaluation of screening for
intestinal metaplasia as marker of gastric cancer risk. A review of gastric cancer in Miyagi Prefecture, Japan: a population-based case-
the evidence. Int J Cancer 2013;133:1023-32. control study. Int J Cancer 1995;60:45-8.
40. Leung WK, Wu MS, Kakugawa Y, et al. Screening for gastric cancer in 64. Inaba S, Hirayama H, Nagata C, et al. Evaluation of a screening pro-
Asia: current evidence and practice. Lancet Oncol 2008;9:279-87. gram on reduction of gastric cancer mortality in Japan: preliminary
41. Korean Statistical Information Service. Cancer registration statistics in results from a cohort study. Prev Med 1999;29:102-6.
2012. Available at: http://kosis.kr/. Accessed July 15, 2015. 65. Tashiro A, Sano M, Kinameri K, et al. Comparing mass screening tech-
42. Cancer Information Service. Cancer statistics in Japan ’13. Available at: niques for gastric cancer in Japan. World J Gastroenterol 2006;12:
http://ganjoho.jp/. Accessed August 6, 2015. 4873-4.

www.giejournal.org Volume 84, No. 1 : 2016 GASTROINTESTINAL ENDOSCOPY 27


Screening and surveillance for gastric cancer Kim et al

66. Cho E, Kang MH, Choi KS, et al. Cost-effectiveness outcomes of the 88. Whiting JL, Sigurdsson A, Rowlands DC, et al. The long term results of
national gastric cancer screening program in South Korea. Asian endoscopic surveillance of premalignant gastric lesions. Gut 2002;50:
Pac J Cancer Prev 2013;14:2533-40. 378-81.
67. Hosokawa O, Hattori M, Takeda T, et al. Accuracy of endoscopy in de- 89. Tava F, Luinetti O, Ghigna MR, et al. Type or extension of intestinal
tecting gastric cancer. J Gastroenterol Mass Surv 2004;42:33-9. metaplasia and immature/atypical “indefinite-for-dysplasia” le-
68. Otsuji M, Kouno Y, Otsuji A, et al. Assessment of small diameter pan- sions as predictors of gastric neoplasia. Hum Pathol 2006;37:
endoscopy for diagnosis of gastric cancer: comparative study with 1489-97.
follow-up survey date. Stomach Intest 1989;24:1291-7. 90. Zullo A, Hassan C, Romiti A, et al. Follow-up of intestinal metaplasia in
69. Lee BE, Kim GH, Park do Y, et al. Acetic acid-indigo carmine chro- the stomach: when, how and why. World J Gastrointest Oncol 2012;4:
moendoscopy for delineating early gastric cancers: its usefulness ac- 30-6.
cording to histological type. BMC Gastroenterol 2010;10:97. 91. Dinis-Ribeiro M, Areia M, de Vries AC, et al. Management of precan-
70. Eleftheriadis N, Inoue H, Ikeda H, et al. Acetic acid spray enhances cerous conditions and lesions in the stomach (MAPS): guideline
accuracy of narrow-band imaging magnifying endoscopy for endo- from the European Society of Gastrointestinal Endoscopy (ESGE),
scopic tissue characterization of early gastric cancer. Gastrointest European Helicobacter Study Group (EHSG), European Society of
Endosc 2014;79:712. Pathology (ESP), and the Sociedade Portuguesa de Endoscopia Diges-
71. Areia M, Amaro P, Dinis-Ribeiro M, et al. External validation of a clas- tiva (SPED). Endoscopy 2012;44:74-94.
sification for methylene blue magnification chromoendoscopy in 92. Correa P, Piazuelo MB, Wilson KT. Pathology of gastric intestinal
premalignant gastric lesions. Gastrointest Endosc 2008;67:1011-8. metaplasia: clinical implications. Am J Gastroenterol 2010;105:
72. Kaise M. Advanced endoscopic imaging for early gastric cancer. Best 493-8.
Pract Res Clin Gastroenterol 2015;29:575-87. 93. ASGE Standards of Practice Committee; Evans JA, Chandrasekhara V,
73. Li HY, Dai J, Xue HB, et al. Application of magnifying endoscopy with Chathadi KV, et al. The role of endoscopy in the management of pre-
narrow-band imaging in diagnosing gastric lesions: a prospective malignant and malignant conditions of the stomach. Gastrointest
study. Gastrointest Endosc 2012;76:1124-32. Endosc 2015;82:1-8.
74. Li CQ, Li Y, Zuo XL, et al. Magnified and enhanced computed virtual 94. Samadder NJ, Jasperson K, Burt RW. Hereditary and common familial
chromoendoscopy in gastric neoplasia: a feasibility study. World J colorectal cancer: evidence for colorectal screening. Dig Dis Sci
Gastroenterol 2013;19:4221-7. 2015;60:734-47.
75. Alaboudy AA, Elbahrawy A, Matsumoto S, et al. Conventional narrow- 95. Dixon MF, Genta RM, Yardley JH, et al. Classification and grading of
band imaging has good correlation with histopathological severity of gastritis. The updated Sydney System. International Workshop on
Helicobacter pylori gastritis. Dig Dis Sci 2011;56:1127-30. the Histopathology of Gastritis, Houston 1994. Am J Surg Pathol
76. Capelle LG, Haringsma J, de Vries AC, et al. Narrow band imaging for 1996;20:1161-81.
the detection of gastric intestinal metaplasia and dysplasia during 96. Guarner J, Herrera-Goepfert R, Mohar A, et al. Diagnostic yield of
surveillance endoscopy. Dig Dis Sci 2010;55:3442-8. gastric biopsy specimens when screening for preneoplastic lesions.
77. Pohl J, May A, Rabenstein T, et al. Computed virtual chromoendo- Hum Pathol 2003;34:28-31.
scopy: a new tool for enhancing tissue surface structures. Endoscopy 97. Ladeiras-Lopes R, Pereira AK, Nogueira A, et al. Smoking and gastric
2007;39:80-3. cancer: systematic review and meta-analysis of cohort studies. Cancer
78. Hamashima C, Ogoshi K, Okamoto M, et al. A community-based, case- Causes Control 2008;19:689-701.
control study evaluating mortality reduction from gastric cancer by 98. Tramacere I, Negri E, Pelucchi C, et al. A meta-analysis on alcohol
endoscopic screening in Japan. PLoS One 2013;8:e79088. drinking and gastric cancer risk. Ann Oncol 2012;23:28-36.
79. Hosokawa O, Miyanaga T, Kaizaki Y, et al. Decreased death from 99. Yang P, Zhou Y, Chen B, et al. Overweight, obesity and gastric cancer
gastric cancer by endoscopic screening: association with a risk: results from a meta-analysis of cohort studies. Eur J Cancer
population-based cancer registry. Scand J Gastroenterol 2008;43: 2009;45:2867-73.
1112-5. 100. Lunet N, Valbuena C, Vieira AL, et al. Fruit and vegetable consump-
80. Lee KJ, Inoue M, Otani T, et al. Gastric cancer screening and subse- tion and gastric cancer by location and histological type: case-
quent risk of gastric cancer: a large-scale population-based cohort control and meta-analysis. Eur J Cancer Prev 2007;16:312-27.
study, with a 13-year follow-up in Japan. Int J Cancer 2006;118: 101. Ge S, Feng X, Shen L, et al. Association between habitual dietary salt
2315-21. intake and risk of gastric cancer: a systematic review of observational
81. Mizoue T, Yoshimura T, Tokui N, et al. Prospective study of screening studies. Gastroenterol Res Pract 2012;2012:808120.
for stomach cancer in Japan. Int J Cancer 2003;106:103-7.
82. Yoon H, Kim N. Diagnosis and management of high risk group for
gastric cancer. Gut Liver 2015;9:5-17.
83. Choi IJ. Gastric cancer screening and diagnosis. Korean J Gastroenter-
ol 2009;54:67-76. [In Korean].
84. Choi KS, Kwak MS, Lee HY, et al. Screening for gastric cancer in Korea: Received November 24, 2015. Accepted February 19, 2016.
population-based preferences for endoscopy versus upper gastroin-
testinal series. Cancer Epidemiol Biomarkers Prev 2009;18:1390-8. Current affiliations: Department of Internal Medicine, Pusan National
85. Riecken B, Pfeiffer R, Ma JL, et al. No impact of repeated endoscopic University School of Medicine and Biomedical Research Institute, Pusan
screens on gastric cancer mortality in a prospectively followed Chi- National University Hospital, Busan, Korea (1), Division of Gastroenter-
nese population at high risk. Prev Med 2002;34:22-8. ology, Department of Medicine, University of Washington, Seattle, Wash-
86. Mori Y, Arita T, Shimoda K, et al. Effect of periodic endoscopy for ington, USA, (2), Department of Internal Medicine, Ulsan University
gastric cancer on early detection and improvement of survival. Hospital, University of Ulsan College of Medicine, Ulsan, Korea (3).
Gastric Cancer 2001;4:132-6. Reprint requests: Joo Ha Hwang, MD, PhD, Division of Gastroenterology,
87. Nam SY, Choi IJ, Park KW, et al. Effect of repeated endoscopic Department of Medicine, University of Washington, 325 Ninth Avenue,
screening on the incidence and treatment of gastric cancer in health Seattle, WA 98104.
screenees. Eur J Gastroenterol Hepatol 2009;21:855-60.

28 GASTROINTESTINAL ENDOSCOPY Volume 84, No. 1 : 2016 www.giejournal.org

You might also like