Sunteți pe pagina 1din 7

GASTROENTEROLOGY 1998;115:642–648

Helicobacter pylori Infection Induces Gastric Cancer


in Mongolian Gerbils

TAKESHI WATANABE,* MAYUMI TADA,‡ HIROFUMI NAGAI,*


SATOSHI SASAKI,* and MASAFUMI NAKAO‡
*Drug Safety Research Laboratories and ‡Pharmacology Laboratories, Takeda Chemical Industries, Ltd., Osaka, Japan

actually associated with gastric carcinogenesis. Gastric


See editorial on page 780.
cancer is a multifactorial disease10; therefore, genetic
factors and environmental factors other than H. pylori can
Background & Aims: Although epidemiological studies complicate the elucidation of the association between
have indicated that Helicobacter pylori infection plays H. pylori infection and gastric cancer. We believe that
a crucial role in gastric carcinogenesis in humans, studies using animal models of H. pylori infection,
there is no direct proof that H. pylori is actually especially those following the effects of long-term infec-
associated with gastric carcinogenesis. The purpose of
tion on the gastric mucosa, should be able to shed further
this study was to elucidate the relationship between
light on this unresolved issue. The Mongolian gerbil
H. pylori infection and gastric carcinogenesis using an
animal model of long-term H. pylori infection. Methods:
(Meriones unguiculatus) is an appropriate animal in which
Mongolian gerbils were orally inoculated with H. pylori, various gastrointestinal diseases such as gastritis and
and the sequential morphological changes in the stom- ulcers that mimic human H. pylori infection can be
ach were examined for up to 62 weeks. Results: studied.11,12 In our current research, we therefore exam-
H. pylori was constantly detected in all infected ani- ined the pathological consequences of long-term H. pylori
mals throughout the study. At the 26th week, severe infection using specific pathogen–free Mongolian gerbils
active chronic gastritis, ulcers, and intestinal metapla- with the aim of clarifying the association of H. pylori and
sia could be observed in infected animals. By the end gastric carcinogenesis.
of the study, adenocarcinoma had developed in the
Materials and Methods
pyloric region of 37% of the infected animals. All
tumors consisted of well-differentiated intestinal-type Five-week-old male specific pathogen–free Mongolian
epithelium, and their development seemed to be closely gerbils (MGS/Sea) were purchased from Seiwa Experimental
related to intestinal metaplasia. Conclusions: We have Animals (Fukuoka, Japan) and maintained under standard
successfully demonstrated that long-term infection laboratory conditions (room temperature, 23°C 6 2°C; rela-
with H. pylori induces adenocarcinoma in Mongolian tive humidity, 55% 6 5%; 12/12-hour light/dark cycle) with
gerbils. The observations are thus highly suggestive of free access to a commercial rodent diet (CE-2; Clea Japan,
the involvement of H. pylori infection in gastric carcino- Tokyo, Japan) and tap water. H. pylori TN2GF4 used in this
genesis in humans. study had been isolated from a patient with gastric ulcer and
was motile and urease, catalase, and oxidase positive. The strain
produced vacuolating cytotoxin and contained the cytotoxin-
lthough the overall incidence of gastric cancer has
A steadily declined over the past 50 years, it is still a
major health problem and remains the second most
associated gene (cagA). H. pylori for the experimental inocula-
tion was grown in brucella broth supplemented with 2.5%
heat-inactivated fetal bovine serum in GasPak jars (BBL,
common cancer in the world.1 Since the discovery of Cockeysville, MD) containing CampyPak (BBL) at 37°C for
Helicobacter pylori in 1983,2 this microorganism has been about 24 hours. Sterilized glycerol was added to the cultures at
a final concentration of 15%, and cultures were kept at 280°C
implicated in gastric carcinogenesis on the basis of
until use. Fifty-five animals were inoculated with 1 mL of
various epidemiological studies.3–7 A Working Group of
broth culture containing 107.54 colony-forming units (CFU) of
the World Health Organization International Agency for H. pylori by intragastric gavage after fasting for 24 hours.
Research on Cancer concluded in 1994 that H. pylori is a
group 1 carcinogen in humans.8 However, the prevalence
of H. pylori in patients with gastric carcinoma still Abbreviation used in this paper: AB-HID, alcian blue with high-iron
diamine.
remains considerably variable among the studies,9 and r 1998 by the American Gastroenterological Association
there has been no direct demonstration that H. pylori is 0016-5085/98/$3.00
September 1998 HELICOBACTER PYLORI INDUCES GASTRIC CANCER 643

Thirty animals served as uninfected controls. Eight of the ture); and (8) carcinoid (diagnosed by neuroendocrine nests in
infected animals died between 40 weeks after inoculation and excess of three glands in diameter). In addition, immunohisto-
the end of the experiment. Data from these animals were chemical staining to detect expression of p53 protein was
excluded from analysis because of autolytic changes in the performed. Formalin-fixed, paraffin-embedded sections from
stomach. Six, 26, 39, and 52 weeks after inoculation, 5 infected animals diagnosed with adenocarcinoma and from 5 uninfected
animals were killed, and, in addition, all surviving animals, 27 animals killed 62 weeks after inoculation were pretreated by
infected and 30 uninfected animals, were killed 62 weeks after autoclaving (105°C, 10 minutes) in a target unmasking fluid
inoculation. At each time point, one half of the stomach from 5 (Monosan; Uden, The Netherlands). After routine inhibition of
infected animals was used for the quantitative determination of endogeneous peroxidase activity with 0.3% H2O2 in methanol,
the bacterium. The other half of the stomach from these 5 nonspecific proteins were blocked with normal goat serum.
animals and the entire stomach from the other animals, killed Specimens were incubated with a monoclonal antibody reactive
62 weeks after inoculation, were examined for sequential to wild-type and mutant-type p53 protein (Dako Japan Corp.,
morphological changes. After the stomach was opened along Kyoto, Japan). Immunolocalization was demonstrated using
the greater curvature, the longitudinal half of the stomach was the Envision method (Dako Japan Corp.) with diaminobenzi-
homogenized with physiological saline. An aliquot of dilutions dine as the substrate. Only distinctly increased nuclear staining
was inoculated onto modified Skirrow’s agar and incubated at was regarded as positive, and when staining was observed in all
37°C for 4 days under microaerobic conditions. The density of or nearly all tumor cells, tumors were considered positive.
infection was estimated by counting the number of colonies per Negative control tissues were prepared in exactly the same way
plate and expressed as log CFU per gastric wall. For histologi- as described above, except the primary antibody was omitted.
cal examination, the stomach was stapled onto paper and fixed All experimental protocols described were conducted in
in 10% neutral buffered formalin. Fixed tissue was cut into accordance with the Guidelines for the Care and Use of
longitudinal strips, about 3 mm wide, for histopathologic Laboratory Animals in Pharmaceutical Research Division,
examination of the entire gastric mucosa. After processing for Takeda Chemical Industries, Ltd., and approved by the Ethical
histology by routine methods, paraffin-embedded sections were Committee for the animal experiments of our division.
cut and stained with H&E; for mucus clarification or for
demonstration of neuroendocrine cells, when necessary, alcian
blue (pH 2.5) with high-iron diamine (AB-HID) stain or Results
Grimelius’ and Sevier–Mungar stains were applied. Histopatho- H. pylori was detected in all infected animals
logic lesions of the glandular stomach were categorized as throughout the study for up to 62 weeks. The bacterial
follows: (1) active chronic gastritis (characterized by severe count reached 105.17 CFU/gastric wall at 6 weeks and
inflammatory cell infiltration and multiple lymphoid follicle
then remained constant until the end of the experiment
formation throughout the pyloric region and part of the fundic
region); (2) ulcer; (3) regenerative hyperplasia; (4) invagination
(Table 1). Sequential histopathologic changes are summa-
of glands into the submucosa; (5) hyperplastic polyp; (6) rized in Table 1. By the 6th week, severe active chronic
intestinal metaplasia (diagnosed by the presence of goblet cells gastritis, characterized by dense neutrophil and mono-
that were positive for AB-HID staining); (7) adenocarcinoma nuclear cell infiltration, and multiple lymphoid follicle
(diagnosed by the presence of atypical glands that locally formation in both the mucosa and submucosa could be
invaded the muscle layer and destroyed the original architec- observed in all infected animals (Figure 1). The gastritis

Table 1. Sequential Microbiological and Histopathologic Changes in the Glandular Stomach of Mongolian Gerbils
Infected With H. pylori
Duration after HP inoculation (wk) 6 26 39 52 62
No. of animals examined 5 5 5 5 27 a
Microbiology
Bacterial count (log CFU/gastric wall) 5.17 6 0.41b 5.33 6 0.98 5.89 6 0.51 4.59 6 1.45 5.40 6 0.95
Histopathology
Active chronic gastritis 5 c (100) 5 (100) 5 (100) 5 (100) 27 (100)
Ulcer 0 (0) 5 (100) 5 (100) 2 (40) 16 (59)
Regenerative hyperplasia 5 (100) 5 (100) 5 (100) 5 (100) 27 (100)
Invagination of glands 0 (0) 5 (100) 5 (100) 5 (100) 27 (100)
Hyperplastic polyp 0 (0) 0 (0) 0 (0) 3 (60) 4 (15)
Intestinal metaplasia 0 (0) 3 (60) 4 (80) 5 (100) 23 (85)
Adenocarcinoma 0 (0) 0 (0) 0 (0) 0 (0) 10 (37)
Carcinoid 0 (0) 0 (0) 0 (0) 0 (0) 3 (11)

HP, H. pylori.
a Microbiological examination was performed on 5 animals.
b Mean 6 SD.
c Incidence with percentage in parentheses.
644 WATANABE ET AL. GASTROENTEROLOGY Vol. 115, No. 3

Figure 1. (A ) Active chronic gastritis observed in the pyloric region of


Figure 2. (A) Intestinal metaplasia observed near the ulcer in Mongolian
Mongolian gerbils 6 weeks after H. pylori inoculation. (B ) The gastritis
gerbils 39 weeks after H. pylori inoculation. (B) The metaplastic glands are
is characterized by severe neutrophil and mononuclear cell infiltration
composed of columnar cells and a large number of goblet cells producing
in both the mucosa and submucosa. The normal mucosal architecture
sialomucins (blue) and occasionally sulfomucins (gray) (H&E [A] and
is almost completely lost and replaced by regenerative hyperplastic
AB-HID [B]; original magnification 653 [A] and 1303 [B]).
epithelium. The thickness of the mucosa increased remarkably
compared with that of an uninfected animal (B) (H&E; original
magnification 653). submucosal cystic glands. These metaplastic glands ap-
peared more atypical than the surrounding nonmetaplas-
originated in the pyloric antrum, extended to the fundic tic, hyperplastic glands (Figure 3). Budding of glands,
region, and was observed continuously throughout the enlarged and vesicular nuclei with prominent nucleoli,
study. The normal mucosal architecture was almost and stratification of nuclei were frequently observed. By
completely lost and was replaced by hyperplastic epithe- the end of the study, adenocarcinoma had developed in 10
lium. The thickness of the mucosa increased remarkably, of the remaining 27 (37%) infected animals. Grossly,
and pseudopyloric gland formation and invagination into almost all tumors were located in the pyloric region and
the submucosa of hyperplastic glands were noted at the detected as crater-like areas with irregular elevated
fundus-pylorus border. After the 26th week, ulcers, ridges. Gastric wall thickening was frequently observed
extending to the muscular layer, developed in the region in these regions. The histomorphological features of these
of the fundus-pylorus border of all infected animals. tumors were quite similar (Figure 4). The tumors arose in
Epithelial hyperplasia progressed, and intestinal metapla- the areas of intestinal metaplasia in deep mucosa or
sia was observed in the mucosa of the pyloric region, submucosa and grew downward. They penetrated into
especially in the area close to the ulcer. These metaplastic the muscle layer and occasionally spread into the serosa.
glands were composed of columnar cells containing little Metastasis or vascular invasion, however, was not ob-
or no mucus and a moderate number of goblet cells
producing sialomucins and occasionally sulfomucins (Fig-
ure 2). In addition, paneth cells were occasionally
observed in some of the metaplastic glands. Cystic
dilatation of the invaginated glands in the submucosa was
a common finding during this period. These cystic
submucosal glands were mainly lined by low, columnar,
mucous cells with little cellular atypia. From the 39th to
52nd week, the structural irregularity of the hyperplastic
glands was more prominent, and complex branching or
cystic dilatation of these glands was frequently observed.
Hyperplastic polyps, characterized by hyperplasia of the
tall columnar surface epithelium and prominent edema,
were also noted in the fundus. Despite such structural Figure 3. (A ) Intestinal metaplasia observed in the pyloric region of
irregularities, little cellular atypia could be observed in Mongolian gerbils 52 weeks after H. pylori inoculation. (B ) The
metaplastic glands show complex branching and budding, and en-
the hyperplastic epithelium. The frequency of intestinal
larged and vesicular nuclei with prominent nucleoli and stratification
metaplasia increased during this period and could be of nuclei are frequently recognized (H&E; original magnifications 503
observed not only in the mucosa but also in the [A ] and 1303 [B ]).
September 1998 HELICOBACTER PYLORI INDUCES GASTRIC CANCER 645

Figure 4. (A ) An adenocarcinoma observed in the pyloric region of a Mongolian gerbil 62 weeks after H. pylori inoculation. (B ) The tumor has
arisen in the area of intestinal metaplasia in the deep mucosa, and the direction of growth is downward. Penetration into the lamina muscularis
and destruction of the original architecture can be observed. (C ) Spreading into the serosa of the neoplastic glands is also noted, and these
glands show nuclear pleomorphism. (D ) The neoplastic glands are composed of intestinal-type epithelium, which contains goblet cells producing
predominantly sialomucins (blue). Cystic dilatation of the neoplastic glands with an accumulation of mucus and extracellular mucus pools are
frequently seen in the submucosa. (E ) Immunohistochemically, almost all tumor cells express p53 protein (H&E; original magnifications 303 [A ],
653 [B ], 1303 [C ]; AB-HID, 1303 [D ]; p53, 1303 [E ]).
646 WATANABE ET AL. GASTROENTEROLOGY Vol. 115, No. 3

served in any case. The tumors retained a well preserved H. pylori infection in humans and Mongolian gerbils, the
glandular structure and consisted of well-differentiated topographic location was similar.6,14–17 The histological
columnar intestinal-type epithelium, which contained a subtype of cancer is difficult to assess accurately because
moderate to large number of goblet cells producing of the existing controversy about the difference between
predominantly sialomucins. Irregular budding or branch- intestinal and diffuse types of gastric cancer in their
ing of glands and nuclear pleomorphism were frequently association with H. pylori infection in humans.5–7,14–17
observed. Copious mucous production was a characteris- The reason for these conflicting observations is consid-
tic feature, and cystic dilatation of neoplastic glands with ered to be mainly the different methods used for the
an accumulation of mucus and extracellular mucus pools detection of H. pylori.15 In studies based on histological
were commonly seen in the submucosa. The stroma of detection of H. pylori, the prevalence has generally been
these tumors showed various degrees of inflammation and higher in the intestinal rather than in the diffuse cancer
fibrosis, and osseous metaplastic foci were occasionally type.7,18,19 This difference has been less distinct or
noted. Immunohistochemically, intranuclear overexpres- nonexistent in most serological studies,5,6,14,17 although
sion of p53 protein was observed in 4 of the 10 (40%) there are some exceptions.15,16 However, in focusing on
adenocarcinomas. In these cases, p53 protein–expressing early gastric cancer to eliminate any possible complicat-
cells were densely distributed in almost all tumor cells. ing effects of tumor progression on colonization with
The relationship between p53 protein expression and H. pylori, the prevalence of H. pylori infection is signifi-
tumor histology, including the invasive pattern, was not cantly higher in intestinal-type gastric cancer regardless
clear. There was no increase in immunoreactivity in the of the method used to detect H. pylori infection.16,20,21 In
uninfected animals, and negative controls stained as addition, several reports have shown that H. pylori
expected. In addition to the adenocarcinoma, carcinoids infection and intestinal-type gastric carcinoma share
derived from fundic mucosa were also detected in 3 some similar epidemiological characteristics.15 The patho-
infected animals. Tumor cells were moderately pleomor- genesis of gastric cancer is certainly multifactorial, and
phic, with pale or eosinophilic cytoplasm and argyro- genetic host and/or environmental factors, other than H.
philic, and were arranged in solid sheets or small nests in pylori, may complicate the resolution of this issue in
lamina propria. One of three carcinoids spread into the humans. It seems to be important, therefore, that tumors
submucosa. No other hyperplastic changes in neuroendo- induced in the H. pylori–infected Mongolian gerbils, in
crine cells were observed in any infected animal. In the which other environmental factors could be minimized,
uninfected animals, no significant changes had been were all intestinal-type adenocarcinoma.
observed by the end of the study. Epidemiological surveys,22,23 histopathologic examina-
tion,24 and biochemical25 and genetic26 analyses have
shown that intestinal metaplasia, especially the sulfomu-
Discussion cin-secreting type, may be a possible precancerous lesion
We successfully showed that long-term infection leading to intestinal-type adenocarcinoma in humans.27
with H. pylori induces gastric adenocarcinoma in Mongo- In the H. pylori–infected Mongolian gerbils, intestinal
lian gerbils. The tumor incidence was 37% (10 of 27 metaplasia occurred in the mucosa or submucosal invagi-
animals) 62 weeks after H. pylori inoculation. Although nated glands in the pyloric region before the cancer
H. pylori infection has been categorized as a risk factor for developed. Although these metaplastic glands produced
gastric cancer based on the results of various epidemiologi- sialomucins predominantly, the metaplastic epithelium
cal studies,3–7 no direct evidence of this relationship has was more dysplastic than the surrounding nonmetaplastic
been presented to date. In animal studies, Fox et al.13 hyperplastic epithelium, and the tumors arose in associa-
reported that Helicobacter mustelae, a relative of H. pylori, is tion with metaplastic epithelium. Furthermore, all tu-
associated with gastric carcinogenesis in naturally in- mors found in the present study consisted solely of
fected ferrets. To the best of our knowledge, our study is intestinal-type epithelial cells. All these results indicate a
the first showing a direct relationship between H. pylori close relationship between intestinal metaplasia and
infection and gastric carcinogenesis. gastric carcinogenesis, and it can be concluded that
All tumors induced in the H. pylori–infected Mongo- intestinal metaplasia is a precancerous lesion leading to
lian gerbils showed similar histological features. They gastric cancer in this model.
were located in the pyloric region and consisted of Intestinal metaplasia has been widely assumed to be
well-differentiated intestinal-type epithelium, correspond- linked to chronic gastritis with mucosal atrophy.28
ing to intestinal-type adenocarcinoma in humans. In Although the pathogenesis of intestinal metaplasia has
comparing the histology of the cancer associated with not been clarified as yet, Scott et al.29 suggested that a
September 1998 HELICOBACTER PYLORI INDUCES GASTRIC CANCER 647

hyperproliferative state in the inflamed gastric mucosa response to cellular damage or inflammatory products
may promote the progression from normal mucosa to stimulate the proliferation of gastric epithelial cells and
metaplastic epithelium. Moreover, Recavarren-Arce et may cause an increased opportunity for mutation.37 An
al.30 reported that an altered gastric microflora, subse- additional explanation is the effect of the formation of
quent to hypochlorhydria due to atrophy, increases nitric oxide, a putative endogenous mutagen in the
N-nitroso compounds in the gastric lumen, and these presence of oxide. It has been reported that a soluble
substances may induce intestinal metaplasia.30 In our protein from H. pylori enhances the expression of the NO
study, so-called mucosal atrophy, characterized by a synthase gene38 and NO production in macrophage cell
decrease in mucosal height and gland density, was not lines.39 In addition, high concentrations of NO induce
evident; however, the original glands, including oxyntic wild-type p53 protein accumulation,40 and the NO-
glands, were almost completely lost and were replaced by related deamination of DNA has been reported to cause
hyperplastic epithelium. This mucosal state would cause GC-AT transitions, which are frequently found in p53
the generation of metaplastic epithelium. Metaplastic mutations in gastric cancer.39 The overexpression of p53
epithelium was frequently noted around the ulcers; protein in gastric cancer observed in the present model,
therefore, ulcer formation might have contributed to may therefore suggest the participation of NO in the
intestinal metaplasia in this model, possibly by enhance- gastric carcinogenesis in H. pylori–infected Mongolian
ment of the mucosal proliferative activity in its regenera- gerbils.
tive process. We have successfully shown that long-term H. pylori
No significant changes were noted in uninfected infection induces gastric cancer in Mongolian gerbils. All
animals in this study. Also, severe gastritis in of the induced tumors were located in the pyloric region
H. pylori–infected Mongolian gerbils can be reversed by and consisted of well-differentiated intestinal-type epithe-
clearance of the organism.12 This indicates that H. pylori lium. The development of these tumors seemed to be
infection alone caused severe inflammatory changes includ- closely related to intestinal metaplasia formed in the
ing intestinal metaplasia, and may also induce gastric chronically inflamed mucosa. Although careful interpre-
cancer. Several animal models for H. pylori produce severe tation is required in extrapolating the data from such an
gastritis, including gnotobiotic piglets, monkeys, and animal model to humans, our present findings are highly
mice.31–33 However, induction of ulceration or intestinal suggestive of the involvement of H. pylori infection in
metaplasia has never been reported in these models, gastric carcinogenesis in humans.
although higher levels of colonization were noted in some References
of them.33 Possibly host factors such as differences in
1. Correa P, Haenszel W. Epidemiology of gastric cancer. In: Correa
mast cell subtype or cytokine production are responsible P, Haenszel W, eds. Epidemiology of cancer of the digestive tract.
for the susceptibility of Mongolian gerbils to H. pylori.12 Hague: Martinus-Nijhoff, 1982:58–84.
Although the pathogenesis of the tumor is unknown, 2. Warren JR, Marshall BJ. Unidentified curved bacilli on gastric
epithelium in active chronic gastritis. Lancet 1983;1:1273–
there are several possible mechanisms by which H. pylori 1275.
infection may play a part in the mutagenesis of the gastric 3. The Eurogast Study Group. An international association between
epithelium. One explanation is increased epithelial turn- Helicobacter pylori infection and gastric cancer. Lancet 1993;341:
1359–1362.
over in the inflamed mucosa. Enhanced cell replication 4. Nomura A, Stemmermann GN, Chyou P-H, Kato I, Perez-Perez GI,
increases the frequency of mutation by errors in gene Blaser MJ. Helicobacter pylori infection and gastric carcinoma
replication, by conversion of endogenous or exogenous among Japanese Americans in Hawaii. N Engl J Med 1991;325:
DNA adducts to mutation, or by diminishing the time 1132–1136.
5. Parsonnet J, Friedman GD, Vandersteen DP, Chang Y, Vogelman
for the DNA repair processes.34 Cahill et al.35 have stated JH, Orentreich N, Sibley RK. Helicobacter pylori infection and the
that gastric mucosal cell proliferation is significantly risk of gastric carcinoma. N Engl J Med 1991;325:1127–1131.
higher in patients with H. pylori–positive gastritis than in 6. Talley NJ, Zinsmeister AR, Weaver A, DiMagno EP, Carpenter HA,
Perez-Perez GI, Blaser MJ. Gastric adenocarcinoma and Helicobac-
those with H. pylori–negative gastritis. Recently, a novel ter pylori infection. J Natl Cancer Inst 1991;83:1734–1739.
cell proliferation gene ( pag) has been shown to share 7. Parsonnet J, Vandersteen D, Goates J, Sibley RK, Pritikin J, Chang
approximately 50% homology with a 26-kilodalton Y. Helicobacter pylori infection in intestinal- and diffuse-type
gastric adenocarcinomas. J Natl Cancer Inst 1991;83:640–643.
antigen of H. pylori.36 In the Mongolian gerbil model, 8. International Agency for Research on Cancer. IARC Monographs
prominent regenerative hyperplasia was a characteristic on the evaluation of carcinogenic risks to humans, Volume 61,
feature and constantly observed throughout the study. Schistosomes, liver flukes and Helicobacter pylori. Lyon, France:
This was suggestive of persistently and highly enhanced 1994.
9. Sipponen P, Kosunen TU, Valle J, Riihelä M, Seppälä K. Helicobac-
cellular proliferation in the gastric mucosa in this model. ter pylori infection and chronic gastritis in gastric cancer. J Clin
Therefore, H. pylori itself and/or an indirect hyperplastic Pathol 1992;45:319–323.
648 WATANABE ET AL. GASTROENTEROLOGY Vol. 115, No. 3

10. Correa P. Human gastric carcinogenesis: a multistep and multifac- evolution of chronic atrophic gastritis and gastric ulcer. A follow
torial process. Cancer Res 1992;52:6735–6740. up study. Gut 1990;31:1097–1104.
11. Hirayama F, Takagi S, Kusuhara H, Iwao E, Yokoyama Y, Ikeda Y. 28. Correa P, Haenszel W, Cuello C, Zavala D, Fontham E, Zarama G,
Induction of gastric ulcer and intestinal metaplasia in Mongolian Tannenbaum S, Collazos T, Ruiz B. Gastric precancerous process
gerbils infected with Helicobacter pylori. J Gastroenterol 1996;31: in a high risk population: cross-sectional studies. Cancer Res
755–757. 1990;50:4731–4736.
12. Matsumoto S, Washizuka Y, Matsumoto Y, Tawara S, Ikeda F, 29. Scott N, Lansdown M, Diament R, Rathbone B, Murday V, Wyatt
Yokota Y, Karita M. Induction of ulceration and severe gastritis in JI, McMahon M, Dixon MF, Quirke P. Helicobacter gastritis and
Mongolian gerbil by Helicobacter pylori infection. J Med Microbiol intestinal metaplasia in a gastric cancer family (letter). Lancet
1997;46:391–397. 1990;1:728.
13. Fox JG, Dangler CA, Sager W, Borkowski R, Gliatto JM. Helicobac- 30. Recavarren-Arce S, León-Barúa R, Cok J, Berendson R, Gilman
ter mustelae–associated gastric adenocarcinoma in ferrets (Mus- RH, Ramı́rez-Ramos A, Rodrı́guez C, Spira WM. Helicobacter pylori
tela putorius furo). Vet Pathol 1997;34:225–229. and progressive gastric pathology that predisposes to gastric
14. Hansson L-E, Engstrand L, Nyrén O, Lindgren A. Prevalence of cancer. Scand J Gastroenterol 1991;26(suppl 181):51–57.
Helicobacter pylori infection in subtypes of gastric cancer. Gastro- 31. Krakowka S, Morgan DR, Kraft WG, Leunk RD. Establishment of
enterology 1995;109:885–888. gastric Campylobacter pylori infection in the neonatal gnotobiotic
15. Martin-De-Argila C, Boixeda D, Redondo C, Alvarez I, Gisbert JP, piglet. Infect Immun 1987;55:2789–2796.
Garcia Plaza A, Canton R. Relation between histologic subtypes 32. Shuto R, Fujioka T, Kubota T, Nasu M. Experimental gastritis
and location of gastric cancer and Helicobacter pylori. Scand J induced by Helicobacter pylori in Japanese monkeys. Infect
Gastroenterol 1997;32:303–307. Immun 1993;61:933–939.
16. Wu M-S, Chen S-Y, Shun C-T, Lee W-J, Wang H-P, Wang T-H, Chen 33. Lee A, O’Rourke J, Corazon De Ungria M, Robertson B, Daskalo-
C-J, Lin J-T. Increased prevalence of Helicobacter pylori infection poulos G, Dixon MF. A standardized mouse model of Helicobacter
among patients affected with intestinal-type gastric cancer at pylori infection: introducing the Sydney strain. Gastroenterology
non-cardiac locations. J Gastroenterol Hepatol 1997;12:425– 1997;112:1386–1397.
428. 34. Butterworth BE, Goldsworthy TL. The role of cell proliferation in
17. Hansson L-E, Engstrand L, Nyrén O, Evans DJ, Lindgren A, multistage carcinogenesis. Proc Soc Exp Biol Med 1991;198:683–
Bergström R, Andersson B, Athlin L, Bendtsen O, Tracz P. 687.
Helicobacter pylori infection: Independent risk indicator of gastric 35. Cahill RJ, Sant S, Beattie S, Hamilton H, O’Morain C. Helicobacter
adenocarcinoma. Gastroenterology 1993;105:1098–1103. pylori and increased epithelial cell proliferation: a risk factor for
18. Buruk F, Berberoglu U, Pak I, Aksaz E, Celen O. Gastric cancer and cancer. Eur J Gastroenterol Hepatol 1994;6:1123–1127.
Helicobacter pylori infection. Br J Surg 1993;80:378–379. 36. Prospéri M-T, Ferbus D, Karczinski I, Goubin G. A human cDNA
19. Loffeld RJLF, Willems I, Flendrig JA, Arends JW. Helicobacter pylori corresponding to a gene overexpressed during cell proliferation
and gastric carcinoma. Histopathology 1990;17:537–541. encodes a product sharing homology with amoebic and bacterial
20. Endo S, Ohkusa T, Saito Y, Fujiki K, Okayasu I, Sato C. Detection proteins. J Biol Chem 1993;268:11050–11056.
of Helicobacter pylori infection in early stage gastric cancer: a 37. Goldstone AR, Quirke P, Dixon MF. Helicobacter pylori infection
comparison between intestinal- and diffuse-type gastric adenocar- and gastric cancer. J Pathol 1996;179:129–137.
cinomas. Cancer 1995;75:2203–2208. 38. Chi DS, Shen D, Li C, Ha T, Ferguson D, Thomas E. TNF-a and
21. Tatsuta M, Iishi H, Okuda S, Taniguchi H, Yokota Y. The associa- nitric oxide (NO) synthase gene expressions in cultured macro-
tion of Helicobacter pylori with differentiated-type early gastric phages enhanced by soluble proteins from Helicobacter pylori
cancer. Cancer 1993;72:1841–1845. (abstr). FASEB J 1994;8:A253.
22. Correa P, Cuello C, Duque E. Carcinoma and intestinal metaplasia 39. Tsuji S, Kawano S, Tsujii M, Takei Y, Tanaka M, Sawaoka H,
of the stomach in Colombian migrants. J Natl Cancer Inst Nagano K, Fusamoto H, Kamada T. Helicobacter pylori extract
1970;44:297–306. stimulates inflammatory nitric oxide production. Cancer Lett
23. Correa P, Cuello C, Duque E, Burbano LC, Garcia FT, Bolanos O, 1996;108:195–200.
Brown C, Haenszel W. Gastric cancer in Colombia. III. Natural 40. Ambs S, Hussain SP, Harris CC. Interactive effects of nitric oxide
history of precursor lesions. J Natl Cancer Inst 1976;57:1027– and the p53 tumor suppressor gene in carcinogenesis and tumor
1035. progression. FASEB J 1997;11:443–448.
24. Nakamura K, Sugano H, Takagi K. Carcinoma of the stomach in
incipient phase: its histogenesis and histological appearances.
Gann 1968;59:251–258. Received October 28, 1997. Accepted May 5, 1998.
25. Kawachi T, Kurisu M, Numanyu N, Sasajima K, Sano T, Sugimura Address requests for reprints to: Takeshi Watanabe, M.D., Drug
T. Precancerous changes in the stomach. Cancer Res 1976;36: Safety Research Laboratories, Takeda Chemical Industries, Ltd.,
2673–2677. 17-85, Jusohonmachi 2-chome, Yodogawa-ku, Osaka 532-8686,
26. Tahara E, Kuniyasu H, Yasui W, Yokozaki H. Gene alterations in Japan. Fax (81) 6-300-6918.
intestinal metaplasia and gastric cancer. Eur J Gastroenterol The authors thank Jeffrey A. Hogan, Leslie I. Brezak, Toshimasa
Hepatol 1994;6(suppl 1):S97–S101. Kyutoku, Yasuharu Yamamoto, Naomi Inui, and Yuri Ozaki for their
27. Silva S, Filipe MI, Pinho A. Variants of intestinal metaplasia in the helpful assistance.

S-ar putea să vă placă și