You are on page 1of 11

SEMINAR

Seminar

Gastric cancer

Peter Hohenberger, Stephan Gretschel

The past decade has seen many advances in knowledge about gastric cancer. Notably, tumour biology and lymphatic
spread are now better understood, and treatment by surgical and medical oncologists has become more standardised.
Since refrigerators have replaced other methods of food conservation, Helicobacter pylori has become a factor in the
cause of gastric cancer. Cancers that arise at the oesophagogastric junction might be further examples of wealth-
associated disease. To tailor treatment better, the western hemisphere needs to borrow from the East by establishing
screening programmes for early diagnosis, through careful surgical resection, and through detailed analysis of tumour
spread. In Europe and the USA, most patients reach treatment with cancers already at an advanced stage. For these
patients, three important randomised trials are underway that evaluate combined therapy. Cytostatic drugs, especially
angiogenesis inhibitors have proved disappointing; however, basic research efforts to detect familial gastric cancers
and to assess minimally residual disease look more hopeful.

Cancer of the stomach is a disease for which treatment treatment. Incidence of tumours in the proximal stomach
and attitudes vary in different regions of the world. That or cardia has increased during the past two decades,
an organ cancer should show varying causative factors in whereas the incidence of distal tumours is actually
different parts of the world is not unusual; however, with dropping.2,7 A strong association between gastric cancer
gastric cancer, it is not only the incidence of the disease, and age exists, irrespective of ethnic group and sex
but also the approach to early diagnosis and treatment (figure 2).2,8 Improvements in survival have been most
that varies greatly between the western and the eastern striking in Japan and Korea, where up to 40% of tumours
hemispheres. Information from Asia about the disease has are detected early by screening programmes.
been used in Europe for teaching purposes and study Studies on the development of gastric cancer suggest
guidance. that genetic predisposition, infection, and diet are part of
Our understanding of the causes of this cancer of the a complex interaction. While assessment of classic risk
stomach, knowledge about tumour spread, and the ability factors such as smoking, alcohol intake, or drinking green
to detect it have greatly improved. Management tea show inconsistent results.9 Regular users of aspirin and
guidelines have been published.1 Surgical treatment has other non-steroidal anti-inflammatory drugs (NSAIDs)
been standardised, and treatment programmes tailored to were at a decreased risk of non-cardia gastric
the individual patient have resulted from new technology adenocarcinoma (odds ratio [OR] 0·46; 95% CI
and a molecular understanding of tumour dissemination. 0·31–0·68), but not of gastric cardia adenocarcinoma
This review summarises these advances, with an emphasis (0·80) when compared with never users, although
on publications since 1995, outlines remaining areas of whether the association is causal remains unclear.
controversy, and provides pointers to what might soon be In 1965, Laurén10 described two histological subtypes of
achievable. gastric cancer, diffuse and intestinal (figure 3). Since then,
differentiation between intestinal and “glandular or
Epidemiology gastric” types has become more distinct in recent years.
Worldwide, gastric cancer is the second largest cause of The intestinal type usually arises in the distal part of the
cancer-related death. The disease is most common in stomach and is found in elderly patients. Its development
Japan and China; in Europe the annual incidence is 12–15 was assumed to follow from chronic atrophic gastritis via
per 100 000, with Portugal at the top end of this range.2,3 metaplasia of the glands induced by exogenous factors.
In the USA, the incidence is low and the death rate for Population-based case-control studies from Spain11 and
gastric cancer is 5·2 deaths per 100 000 compared with 90 Sweden12 ascribed a cancer-promoting effect to nitrates
per 100 000 in Japan.4 During the past 50 years, incidence and nitrose compounds. However, there is no proof from
of and mortality from gastric cancer have decreased prospective studies that the more common use of
worldwide, especially in developed countries (figure 1).2,5,6 refrigerators instead of salting, smoking, and pickling for
These improvements are explained by better living food conservation explains the drop in the incidence of
conditions and increased consumption of fresh fruit, gastric cancer of intestinal type. The World Cancer
vitamins, and vegetables, rather than by improvements in Research Fund notes convincing evidence for an inverse
relation between fruit and vegetable intake and gastric
Lancet 2003; 362: 305–15 cancer.13 However, evidence from interventional studies
that increased intake of vitamins and tracer elements can
Division of Surgery and Surgical Oncology, Robert Roessle Hospital prevent cancer is weak. Results of a Swedish population-
at the Max Delbrück Center for Molecular Medicine, Charité,
Humboldt University at Berlin, Germany (Prof P Hohenberger MD, Search strategy
S Gretschel MD)
We searched the MEDLINE database (years 1995 to July,
Correspondence to: Prof Peter Hohenberger, Division of Surgery and 2002) with the keywords: gastric cancer, stomach cancer,
Surgical Oncology, Robert Roessle Hospital and Tumour Institute at epidemiology, Helicobacter pylori, D2-lymphadenectomy,
the Max Delbrück Center for Molecular Medicine, Charité, Campus
adjuvant, neoadjuvant, randomized trial, minimal residual
Berlin-Buch, Humboldt University, D-13125 Berlin, Germany
disease.
(e-mail: hohenberger@rrk-berlin.de)

THE LANCET • Vol 362 • July 26, 2003 • www.thelancet.com 305

For personal use. Only reproduce with permission from The Lancet.
SEMINAR

35
White men
32·5
White women
30
Black men
27·5 Black women

25

22·5
Rate per 100 000

20

17·5

15

12·5

10

7·5

2·5

0
73
74
75
76
77
78
79
80
81
82
83
84
85
86
87
88
89
90
91
92
93
94
95
96
97
98
99
19
19
19
19
19
19
19
19
19
19
19
19
19
19
19
19
19
19
19
19
19
19
19
19
19
19
19
Year of diagnosis
Figure 1: Incidence of gastric cancer in the USA from 1973 to 1999
Data from SEER Cancer Statistics Review.5

based case-control study showed that there was no Examination of very small early carcinomas also raises
reduction in risk of cancer of the gastric cardia, and that doubts about the importance of dysplasia as a
more than 25 000 individuals would need to increase their precancerous lesion, because at the periphery of
food and vegetable consumption to prevent one carcinomas that were less than 5 mm in size no precursor
oesophageal cancer per year.14 dysplasias were noted.15
Cancer of intestinal type can also arise without By comparison with the intestinal type, the diffuse type of
metaplasia, and the sequence of atrophic gastritis to gastric cancer is more common in young patients, and
intestinal metaplasia to dysplasia to carcinoma has been people with blood group A are predisposed to the disease.
challenged by the postulated role for Helicobacter pylori. The incidence of genetically associated diffuse cancers is
assumed to be 5–10%.16 Germline mutations of the CDH1
180 gene that encodes E-cadherin, an epithelial cell adhesion
White men molecule, were identified in few families with hereditary
160
White women diffuse gastric carcinoma in a pattern suggestive of
140 Black men autosomal dominant inheritance with incomplete
Rate per 100 000

Black women penetrance. Prophylactic gastrectomy revealed


120
intramucosal signet-ring cell adenocarcinoma in regions of
100 the stomach that had not been detected with endoscopy.
These findings have opened up a debate about the merits of
80
prophylactic total gastrectomy for gene mutation carriers.
60 In diffuse-type cancer, minute signet-ring tumours develop
40
de novo from stem cells in the proliferative cell zone at the
neck region of gastric glands.17 Intestinal metaplasia,
20 dysplasia, and adenocarcinoma arise coincidentally,18 which
0 implies that no precursor is present for any of these
malignant diseases and development of the intestinal type
00–04
05–09
10–14
15–19
20–24
25–29
30–34
35–39
40–44
45–49
50–54
55–59
60–64
65–69
70–74
75–79
80–84
85

from signet-cell cancer might be a time-dependent event.19

Age at diagnosis (years) Role of H pylori


In 1983, Marshall and Warren20 detected a gram-negative
Figure 2: Age dependent incidence of gastric cancer in the bacillus, now called H pylori, which has since been
USA associated with gastric ulcer disease. Infection is most
Data from SEER Cancer Statistics Review.5 likely to take place during childhood via the faeco-oral

306 THE LANCET • Vol 362 • July 26, 2003 • www.thelancet.com

For personal use. Only reproduce with permission from The Lancet.
SEMINAR

carcinoma,30,31 whereas individuals with extensive corpus


gastritis develop hypochlorhydria and gastric atrophy.
Host factors seem to play a crucial part since interleukin-1
(IL-1) gene cluster polymorphisms suspected of
enhancing production of IL-1 are associated with an
increased risk of both hypochlorhydria induced by
H pylori and gastric cancer. IL-1 is a proinflammatory
cytokine and a powerful inhibitor of gastric acid secretion.
Host genetic factors that affect IL-1 might determine
why some individuals infected with H pylori develop
gastric cancer and others do not.
IL-1 is not the only molecular factor that might be
relevant to H pylori infection. For example, the adherence
of H pylori to the gastric mucosa involves specific bacterial
adhesins and host receptors. The Lewis (b) blood-group
antigen mediates H pylori attachment to gastric mucosa.
H pylori infection has been shown to induce formation of

Reproduced with permission from Armed Forces Institute of Pathology


sialyl-Lewis X antigens in gastric epithelium in humans
and in rhesus monkeys.32 The ability of many H pylori
strains to adhere to sialylated glycoconjugates expressed
during chronic inflammation might thus contribute to the
virulence and extraordinary chronicity of H pylori
infection and account for the existence of H pylori strains
of differing virulence, especially cagA-positive strains.33
CagA positive strains are able to colonise both the gastric
and duodenal mucosa. With PCR, H pylori virulence
genes were analysed and it could be shown that strains
coexpressing cagA, s1m1 vacA, and babA2 genes worsened
inflammation significantly.34 Soluble glycoproteins
presenting the Lewis (b) antigen or antibodies to it inhibit
bacterial binding. Gastric tissues that do not express
Lewis (b) do not bind H pylori, and the bacteria do not
Figure 3: Histological subtypes of gastric cancer bind to this antigen when it is substituted with a terminal
(A) Gastric carcinoma of intestinal type. Normal mucosa is replaced by GalNAc--1-3 residue, the blood group A determinant.
infiltrating tubular profiles. Haematoxylin and eosin, 200. (B) Signet-ring This finding suggests that the availability of H pylori
cell gastric carcinoma of the diffuse type. There is diffuse infiltration of
the mucosa by signet ring cells. The gastric pits appear normal and there receptors might be reduced in individuals of blood group
is no evidence of gastritis or intestinal metaplasia. Haematoxylin and A and B phenotypes, compared with blood group O
eosin, 200. individuals.35
H pylori substantially impairs the bioavailability of
route, and transmission is from person to person.21–24 vitamin C, which together with the reduced vitamin C
Prevalence of the bacteria is high in Asia and Eastern intake of H pylori-positive people, greatly reduces
Europe and low in Western and Northern Europe as well concentrations of vitamin C in plasma. The reduced
as in North America. In a population-based case-control circulating concentrations of vitamin C in people infected
study, 72% of 190 patients with gastric adenocarcinomas with H pylori may be a causative factor in gastric cancer, as
were seropositive by IgG ELISA and 91% by immunoblot well as other diseases associated with antioxidant
assay, versus 55% in 144 controls. 71% of non-cardia deficiency.36 Successful eradication of H pylori improves
adenocarcinomas were attributable to H pylori. Results secretion of vitamin C into gastric juice, which might
from Japan and the UK suggest that in patients younger increase protection against gastric cancer.37 Results of a
than 40 years, 45–55% of all gastric cancers might be randomised, placebo-controlled trial in a Colombian
associated with this infection. Serological investigations population of people at high risk of gastric cancer showed
might be misleading: elderly patients with mucosal that vitamin C, beta-carotene, and H pylori eradication
atrophy develop seroreversion. Initial analysis showed that were statistically significant in promoting regression of
89% of histological sections of patients with stomach precursor lesions.38
cancer of intestinal type contained H pylori in non- Despite recent advances in knowledge about differences
cancerous tissue compared with 32% in diffuse type in carcinogenesis of gastric cancer subtypes, genetic
cancer;25 however, results of serological analysis showed alterations between H pylori positive and non-H pylori
an association between H pylori and both types of gastric associated cancers could not be verified.15,39
cancer.26,27 The association between cancer of the gastric
cardia and H pylori infection is ambiguous. Progress in diagnosis
H pylori infection is associated with various clinical Patients in eastern countries are mainly those with early
outcomes, including gastric cancer and duodenal ulcer cancers, whereas western patients are usually treated
disease. There is also evidence that infection in children when the cancer is at an advanced stage. This difference
and adults leads to different pathology.28 This variation in might have contributed to confusion in histopathological
outcome seems to be related to the severity and the classification of the disease (panel 1 and 2), and made
anatomical distribution of the gastritis attributable to comparison of treatment results from the two regions
H pylori.29 Patients who have duodenal ulcer with antrum- difficult.40 In the west, inflammatory and regenerative
dominant H pylori gastritis and only very low-grade corpus changes of the mucosa of the stomach were classified as
H pylori gastritis retain normal (or even high) acid dysplasia, as were well-differentiated adenocarcinomas. In
secretion, and they very rarely develop gastric follow-up studies, people who had gastric mucosa biopsy

THE LANCET • Vol 362 • July 26, 2003 • www.thelancet.com 307

For personal use. Only reproduce with permission from The Lancet.
SEMINAR

Panel 1: TNM staging system and appearance of Panel 3: Vienna classification of epithelial
gastric cancer (5th edn, 1997) neoplasia of the gastrointestinal tract36
Primary tumour (T) Category comments
Tis Carcinoma in situ, intraepithelial tumour (see 1 Negative for neoplasia
Vienna classification) 2 Indefinitive for neoplasia
T1 Tumour invades lamina propria (T1a) or 3 Non-invasive low-grade neoplasia
submucosa (T1b) 4 Non-invasive high-grade neoplasia
T2 Tumour invades muscularis propria or 4·1 High-grade adenoma
subserosa; extension into omenta or 4·2 Non-invasive carcinoma
gastrohepatic ligaments is classified T2, until 4·3 Suspicious for invasive carcinoma
perforation of the visceral peritoneum has 5 Invasive neoplasia
occurred 5·1 Intramucosal carcinoma
T3 Tumour penetrates serosa 5·2 Submucosal carcinoma or beyond
T4 Tumour invades adjacent structures
judged. Now, endoscopic ultrasonography is accepted as
Lymph-node metastases (N)
superior to conventional CT. However, CT with gastric
Histological examination of a regional lymphadectomy
distention of 600–800 mL of water (hydro-CT) might be
specimen will ordinarily include 15 or more nodes
a complementary investigation and should be noted
N0 No regional lymph-node metastases
(table 1). Endoluminal ultrasonography is used with
NX Less than 15 investigated lymph nodes
rotating or sectorial scanners of 7·5 MHz or 12 MHz.
N1 1–6 regional lymph-node metastases
Rotating scanners improve orientation, while the sectorial
N2 7–15 regional lymph-node metastases
probes allow biopsies to be taken. The gastric fundus is a
N3 More than 15 regional lymph-node metastases
difficult area to assess as is the lesser gastric curvature
M1 Metastases in retropancreatic, mesenteric, or
where the scanner cannot be positioned exactly. In early
para-aortic nodes
gastric cancer, rotating miniprobes of higher resolution
Distant metastases (M) (20 MHz) can be used to separate mucosal from
M0 No distant metastases submucosal invasion.43 Endoluminal ultrasonography
M1 Distant metastases cannot reliably discriminate a T2 from a T3 lesion,
especially if the carcinoma invades the greater or lesser
specimens classified as high-grade dysplasia, up to 75% of omentum; however, even high-resolution CT cannot
the patients developed a cancer within a median time of differentiate between T1 and T2 lesions.
8 months. There were no similar reports from Japan, Routine detection of early gastric cancer is a rare event.
Korea, or Asia. As a consequence of this finding, reported Therefore, workers have investigated indigo-carmine
by the British Society of Gastroenterology first in 1990 staining of the mucosa applied locally via the endoscope,
with confirmative reports later on,41 the Vienna or fluorescein given intravenously (chromoendoscopy).
classification of epithelial neoplasia of the gastrointestinal Results of this work might allow detection of early cancer
tract (oesophagus, stomach, colorectum) was introduced even if the lesion does not belong to either the protruded
in 1998.42 Differences between western and Japanese or the depressed type.44
pathologists’ diagnostic classification of gastrointestinal
epithelial neoplastic lesions could largely be resolved by Limited resection in early gastric cancer
the adoption of a terminology based on cytological and In early gastric cancer, 5 year survival can be better than
architectural severity and invasion status (panel 3). Here, 90% if treated in experienced hands. In Japan, screening
high-grade dysplasia, non-invasive carcinoma, and endoscopy results in the detection of up to 40% of gastric
suspected invasive carcinoma are integrated into one cancers at an early stage; in Europe the proportion is less
term. Although it might be advantageous to use these than 15%. In early cancers, lymphnode metastasis is
categories to initiate adequate follow-up, local unusual; up to 4% for the mucosal type T1a and about
endoscopical, or surgical treatment it is local staging 23% for the submucosal type T1b. If morphological
about the depth of invasion that is the essential factors (size, growth type, ulceration, grading) are
prerequisite for any treatment decision. considered, T1 tumours with a very low risk of lymph-
node metastases can be selected.45 Results from a study in
Endoscopical ultrasound for local staging Japan46 of more than 5000 patients with early gastric
Flexible endoscopy is the method of choice to establish cancer who had gastrectomy with lymph-node dissection,
the diagnosis of gastric cancer, and biopsies are needed for showed that none out of 1230 well-differentiated
confirmation. Early gastric cancer (ie, confined to the intramucosal cancers of less than 30 mm diameter was
mucosa [T1a]) might be treated locally in some cases, and associated with metastases. None of the 929 lesions
thus the depth of infiltration to the gastric wall without ulceration was associated with nodal metastases
(T-category) is of utmost importance, because it allows irrespective of tumour size. In submucosal cancers, there
the risk of lymphatic dissemination of the tumour to be was a correlation between tumour size greater than
30 mm and lymphatic-vascular involvement and increased
Panel 2: TNM staging system stage classification CT EUS Hydro-CT Lap
N0 N1 N2 N3 T category 25–66% 71–92% 51% 47%
M0 T1 Ia Ib II IV N category 25–68% 55–87% 51% 60–90%
T2 Ib II IIIa IV M category 65–72% ·· 79% 80–90%
T3 II IIIa IIIb IV EUS=endogastric ultrasonography. Lap=laparoscopy (with laparoscopic
T4 IIIa IV IV IV ultrasonography).
M1 IV IV IV IV Table 1: Accuracy of four diagnostic methods to assess TNM
features in patients with gastric carcinoma

308 THE LANCET • Vol 362 • July 26, 2003 • www.thelancet.com

For personal use. Only reproduce with permission from The Lancet.
SEMINAR

risk of nodal metastases. In these patients with smaller


Panel 4: Laparoscopic staging of gastric cancer54
tumours, total gastrectomy with clearance of the
lymphatic drainage area is not recommended. In Japan, After inflation of the pneumo-peritoneum, a visual turnaround
surgeons favour endoscopic mucosal resection, which is of the abdominal cavity is done and peritoneal or liver surface
thought to have high curative potential and to avoid the is inspected for small tumour deposits. Assessment of
need for further radical surgery; however, such an serosal invasion (T3) can be based on direct view and biopsy.
approach should only be done if very accurate local Thereafter, the lesser sac is opened to explore local
staging has been achieved.47 Furthermore, the risk of resectability in tumours of the dorsal part of the stomach.
missing lymph-node metastases is about 2·5%. Lavage of the bursa is done; thereafter, laparoscopic
Early endoscopic mucosal resection techniques used ultrasonography under direct view allows a thorough
norepinephrine to raise lesions, which were then resected examination of subdiaphragmal liver segments not
with an electrical sling: R0-resection rates ranged from assessable to transcutaneous ultrasonography. Perigastric
45–65%. Techniques evolved to use a panendoscope to lymph nodes, the coeliac axis and nodes along the
obtain a specimen with a large 20 mm circular cap under hepatoduodenal ligament are examined. Most important,
full suction.48 After resection of 167 specimens from 97 suspicious lymph nodes can be excised or biopsied.
lesions in 85 patients, 93% of elevated lesions were
resected completely; however, in flat or depressed lesions by opening the lesser sac via the minor omentum or the
only 53% of the tumours could be removed by a single gastrocolic ligament. Laparoscopic staging done by a
approach with clear margins. Endoscopic mucosal skilled surgeon immediately before treatment surgery
resection is often impossible for lesions located at the seems to be the best diagnostic tool to guide decisions
gastric cardia or lesser curvature, but these difficulties about surgical resection or palliative measures.55,56 If the
might be overcome by combining endoscopic and staging laparoscopy is done as a separate procedure,
laparoscopic mucosal resection. The area of the removed lavage of the omental sac or the abdominal cavity can be
specimen could be as large as 5050 mm. 5-year follow- used for peritoneal washing. Intraperitoneal free cancer
up results with laparoscopic resection in the lesion-lifting cells imply a very poor prognosis in gastric carcinoma;
method (ie, laparascopic resection of the tumour-bearing however, there is no consensus on how these patients
segment with a linear cutter after the gastric wall has been should be managed.
lifted with a T-shaped metal stab inserted beneath the
tumour while it is under endoscopical control), showed Cancer of the oesphagogastric junction
that only one patient needed gastrectomy for tumour By contrast with the diminishing frequency of gastric
invasion to the submucosal layer, and two developed cancer, the rate of carcinoma at the oesphagogastric
intragastric recurrence near the staple line.49 The Japan junction is rising, especially in industrialised countries. In
Gastric Cancer Association issued treatment guidlelines in Europe, increased incidence of this type of cancer has
March, 2001, for doctors, patients, and their families.50 been reported in Denmark, Italy, and the UK.2,7,57,58 The
frequency of adenocarcinoma of the gastric cardia
Assessment of abdominal spread in advanced increased 29·1% to 52·2% from 1984 to 1993
cancers (p<0·0001). This tumour type is distinct from cancer of
Lymph-node metastases arise in up to 44% of patients the lower oesophagus or Barrett’s carcinoma. Only a small
with T2 tumours, and 64% in those with T3 cancers. subgroup of these cancers is promoted by
Surgical clearance of the lymphatics is essential if a gastroesophageal reflux, so antireflux surgery will not
curative approach is to be taken, or if patients are to have prevent them. In a population-based retrospective cohort
preoperative chemotherapy. study in Sweden, 67 000 patients with gastro-oesophageal
Knowledge of the presence and extent of nodal spread reflux disease were compared with 11 000 who underwent
is helpful in planning treatment. A CT scan, however, anti-reflux surgery. The standardised incidence ratio of
does not provide much information, and only the size of gastric cardia adenocarcinoma was 2·4 ( 95% CI 1·7–3·3)
nodes can be used as a criterion. Endoluminal in men who did not have surgery and 5·3 (95% CI
ultrasonography can assess lymph nodes close to the 3·0–8·7) in those who had undergone surgery.59 The
gastric wall, and features such as a rounded shape, cancer risk pattern in women was similar, but the number
hypoechoic patterns, and a size larger than 1 cm suggest of cases was very much smaller. Associations have also
metastasis. Rarely, analysis of transgastric biopsy samples been reported between cancer of the gastric cardia and
can prove tumour infiltration, and within this context, H pylori infection and Epstein-Barr virus.60,61
laparoscopy has had a new lease of life. Laparoscopy had Irrespective of the cause, cancer of the gastric cardia is
been used for around four decades for the staging of characterised by poor prognosis, which might be partly
gastrointestinal cancer,51 but its use diminished with the due to the fact that tumours are located at the border of
arrival of CT and MRI in the mid sixties. However, since three areas of lymphatic drainage—mediastinal,
1990, laparoscopy has been used again because of new abdominal, and retroperitoneal. Furthermore, there is no
techniques, such as combining the procedure with peritoneal cover in the dorsal part of the cardia. Lymph-
pneumoperitoneum, the use of trocars, and the node metastases might develop early and at locations such
development of specialised instruments, 30 viewing as the left renal vein and para-aortic nodes—regions not
optics, and laparoscopic ultrasound probes.52,53 The routinely cleared during surgery for gastric cancer.62,63 A
sequence of staging laparascopy is shown in panel 4. delay in diagnosis might occur with cancers in oesophago-
Laparoscopic staging can achieve a 92% rate of detection gastric junction because the subcardial area cannot be
for advanced gastric cancer, compared with only 58% easily viewed by endoscopy, and because symptoms
with CT and 63% with endoscopic ultrasonography caused by tumour stenosis occur only at a late stage in the
(table 1). Workers in several prospective series52–56 have disease. Carcinomas of the gastro-oesophageal junction
reported that up to a third of patients had a tumour at a might arise from the distal oesophagus, the true cardia or
more advanced stage than had been expected, and the the subcardial region.
treatment plan had to be changed. Even if the tumour is at Siewert and Stein64 have proposed a classification based
the dorsal part of the stomach, it can be assessed directly on the different origins of oesophago-gastric tumours.

THE LANCET • Vol 362 • July 26, 2003 • www.thelancet.com 309

For personal use. Only reproduce with permission from The Lancet.
SEMINAR

Number Regimen R0- Survival In a Dutch randomised trial, 56 patients were allocated
of resection time either up to four cycles of 5-fluorouracil, adriamycin, and
patients rate (%) (months) methotrexate (FAMTX) before surgery or to surgery only.
Ajani73 56 Preop FAMTX, postop 61 15* 12 patients (44%) could not complete chemotheraphy
5-FU/CDDP because of tumour progression toxicity, or both. This
48 Preop and postop EAP 77 16† small trial revealed no benefit for chemotherapy, and the
Fink74 49 Preop PLF 76 36*
Dutch investigators concluded future trials should use
Kang75 107 Surgery vs PEF and 61 vs 78 30 vs 42*
surgery (p=0·049) p=0·11 more active regimens.76 Although some workers have
Songun76 56 Surgery vs preop 62 vs 56, 13·1 vs reported success in the downstaging locally advanced
FAMTX and surgery (ns) 12·8 (ns) cancer and also long survival rates in patients with
77
Skoropad 78 Surgery vs preop RT Data not 9 vs 21 (ns) otherwise unresectable cancers, surgical staging was not
20 Gy IORT+surgery shown done to confirm the unresectability of lesions.79,80
Zhang78 370 Surgery vs 40 Gy and 79 vs 89 20 vs 30
surgery (p=0·01) (10 yr)
Three large phase 3 trials are underway to assess the
p=0·0094 usefulness of neoadjuvant treatment. The MAGIC trial in
Preop=preoperative. Postop=postoperative. FAMTX=5-fluorouracil, doxorubicin,
the UK compared surgery alone with ECF (epirubicin,
methotrexate. CDDP=cisplatin. EAP=etoposide, doxorubicin, cisplatin. cisplatin, and 5-fluorouracil). Most recently, preliminary
PEF=cisplatin, etoposide, 5-fluorouracil. PLF=cisplatin, leucovorin, fluorouracil. data of this trial involving 503 patients were released.81
RT=radiotherapy. ns=not significant. IORT=intraoperative radiotherapy. *Median
survival time. †Mean survival time.
The authors reported an increased R0 resection rate of
79% after combined modality therapy compared with 69%
Table 2: Results of neaodjuvant treatment concepts for locally
after resection alone (p<0·02). Pathological examination of
advanced gastric cancer
the resection specimens suggested a downsizing of the
tumour. Perioperative complication rates seemed not to be
Type I represents adenocarcinoma of the distal raised. Whereas progression-free survival was improved
oesophagus with the centre of the tumour lying 2·5 cm significantly, there was no advantage in overall survival
above the anatomical cardia. Such tumours should be after a minimum of 1 year follow-up in all patients. The
treated by subtotal resection of the oesophagus including Swiss study, SAKK 43/99, compares surgery plus
the cardia and gastric fundus together with removal of the preoperative TCF (taxotere, cisplatin, and 5-fluorouracil)
lymph nodes to the coeliac axis. The type II tumour is true with surgery followed by the same chemotherapy regimen.
carcinoma of the cardia with centres in the region 1 cm A multinational European trial (EORTC 40954) compares
above and 2 cm below the anatomical cardia. surgery alone with surgery after neoadjuvant PLF
Gastrectomy, splenectomy, and resection of the distal part (cisplatin, leucovorin, and 5-fluorouracil).
of the oesophagus is recommended for type 2. Type 3 To identify patients who should receive palliative care
cancer is a gastric carcinoma with its centre 2–5 cm below rather than neoadjuvant treatment, staging laparoscopy
the anatomical cardia and should be treated by total with inspection of the bursa omentalis should be done to
gastrectomy including splenectomy and removal of a short identify peritoneal spread.
part of the distal oesophagus. Lymphatic clearance is
recommended caudally to the coeliac axis, hilus of the Gastric resection and lymphatic clearance
spleen, and para-aortic nodes. This classification system Whether the optimum surgery for gastric cancer in the
has been endorsed by the International Society for distal half of the stomach is subtotal or total gastrectomy is
Diseases of the Oesophagus and the International Gastric not clear. The Italian Gastrointestinal Tumour Study
Cancer Association. Group randomly allocated 624 patients with cancer in the
It is essential that the type of lesion is known before distal half of the stomach to subtotal gastrectomy (n=320)
planning a patient’s treatment. Because gastric cardia or total gastrectomy (n=304). Both procedures included
cancer is a three-compartment tumour, it is not surprising a second-level lymphadenectomy. Inclusion criteria were a
that incomplete resections, either for ascending intramural tumour with a proximal edge at least 6 cm from the cardia,
spread to the oesophagus or transmural spread to the and no intraperitoneal or distant spread. At 5 years, the
pericardiac soft tissues affects prognosis.65,66 survival probability was 65·3% for patients with subtotal
and 62·4% for those with total gastrectomy. Subtotal
Neoadjuvant treatment options gastrectomy with lymphadenectomy of compartments one
In western countries, when patients with cancer of the and two has been associated with a better nutritional status
stomach are referred for treatment, they are likely to have and quality of life, and it should be the procedure of
locally advanced disesase: 44% of patients in the UK- choice.82
lymphadenectomy study had pT3 tumours,67 59% in a Lymph-node metastasis decisively affects prognosis in
German study had stage III or IV disease,68 as did 67% of stomach cancer. Operative clearance of the lymphatics is of
18 365 patients in the American College of Surgeons’ utmost importance and requires a thorough surgical
study.69 In the presence of advanced disease, complete training if it is to be done effectively and safely. Removal of
resection of the tumour and its lymphatic spread can be the perigastric lymph nodes only is called D1 resection. D2-
achieved in fewer than 50% of patients. Furthermore, lymphadenectomy adds removal of the lymphatic chains
median survival rates at 5 years are, at best, around 30% along the coeliac axis, the common hepatic and splenic
because of locoregional recurrence and systemic artery, and at the hilus of the spleen, and this procedure has
metastases.70–72 Consequently, several trials have sought to been recommended by the Japanese Society for Research
assess whether patients benefit from preoperative in Gastric Cancer, the European Society of Surgical
chemotherapy to downstage the tumour, and eliminate Oncology, the International Gastric Cancer Association,
micrometastases and intraoperative spillage of tumour and the US National Comprehensive Cancer Network.
cells. Unfortunately, encouraging results of phase 2 trials The D2 procedure is done to achieve: accurate staging and
have not been reported in full, while data from randomised regional disease control, and because of potential benefit to
trials do not lend support to the neoadjuvant approach, a subgroup of patients with occult disease in D2 nodes. D2
even if chemotherapy is combined with preoperative or lymphadenectomy is safe if done by a skilled surgeon, and if
intraoperative radiation treatment (table 2). splenectomy and pancreatic resection are avoided.

310 THE LANCET • Vol 362 • July 26, 2003 • www.thelancet.com

For personal use. Only reproduce with permission from The Lancet.
SEMINAR

Patients Morbidity (%) Mortality (%) 5-year


(D1/D2) (D1/D2) (D1/D2) survival (%)
(D1/D2)
Bonenkamp85,86 380/331 25/43 4/10 45/47
Cuschieri67,71 200/200 28/46 6·5/13 35/33
Dent87 22/21 15/30 0/0 78/76
(3 years)
Table 3: Comparison of morbidity, mortality, and 5-year survival
in randomised trials of D1 versus D2 lymphadenectomy

Rights were not granted to include this


Individualised assessment of lymphatic spread
image in electronic media. Please To avoid unnecessary removal of lymph nodes that are
refer to the printed journal. not at risk of tumour, two approaches have been pursued.
The first technique uses a computerised database of
information to convert a large amount of information and
experience to a treatment decision for an individual
patient. The second approach uses information derived
from dissection of the sentinel node.
Maruyama and his co-workers developed a computer
program (Kampschoer and colleagues)90 to calculate the
probability of nodal spread to each individual lymph-node
station, using histopathological features of the primary
tumour. At first, the program used information from about
Figure 4: Multidirectional lymphatic network of the stomach 2000 patients from the National Cancer Centre of Tokyo;
and its relation to the pancreas, liver, mediastinum, and however, the program is continuously updated and it
retroperitoneum now includes data from more than 8000 patients. Depth of
Figure from Inoue83 infiltration, tumour size, tumour location, grading, typing,
and macroscopic appearance are used to predict the
Lymphatic drainage of the stomach is multidirectional probability of nodal metastases. Application of this program
and involves a wide net of lymphatic channels. Detailed in Germany and Italy resulted in a diagnostic accuracy
descriptions of these pathways go back to the 1930s of 74% and 94%, respectively.91,92 However, the point at
(figure 4).83 Skip metastases can occur in up to 20% of which lymph-node dissection should be done is not clear.
patients and the aim of clearing one lymph-node station Maruyama and colleagues initially recommended a cutpoint
beyond the last node occupied by cancer led to the of 18%; however, if this recommendation was used, up to a
development of D2 lymphadenectomy. fifth of patients would have incomplete operations.
Results of eight prospective, non-randomised trials In the 1950s, Weinberg93 was the first to identify
showed substantial survival advantages for D2 over D1 perigastric lymphatic vessels and nodes by injecting blue
lymphadenectomy, especially in patients with stage dye (figure 4). Later, activated carbon was favoured
2 and 3a disease.84 However, morbidity and mortality are for lymphatic mapping. In a prospective randomised
associated with D2 lymphadenectomy, most often evaluation of preoperative endoscopic vital staining with
explained by lesions to the pancreatic tail or because the CH40 before D2 lymph-node dissection, the rate of
pancreas needs to be resected to achieve complete metastatic nodes was substantially higher in patients who
removal of the lymph nodes at the splenic hilus (table 3). had staining versus those who did not.94 The success of
Three randomised, controlled trials have been done isolated drainage of CH40 led to the use of this approach
in the past years to assess whether D2 lymphadenectomy for endolymphatic chemotherapeutic treatment of nodal
in combination with total gastrectomy really yields a metastases via absorption of mitomycin C.95 None of these
substantial survival advantage. A Dutch multicentre methods was able to detect the first draining lymph-
trial85 used instructors from Japan and strict quality node—ie, the so-called sentinel node, which has been
control to ensure that D2 lymphadenectomy was done shown to be a marker of lymph-node metastases in
accurately. However, all three trials showed increased melanoma and breast cancer by several studies from the
morbidity with D2 lymphadenectomy and improved USA and Europe. Because the stomach has four main
survival rates were not evident. Pancreatic fistula and lymphatic drainage directions (coeliac axis, liver,
pancreatitis resulting from splenectomy and resection mediastinum, and retroperitoneum) more prospective data
of the pancreatic tail were the main contributors to are needed before drainage directions revealed by blue dye
morbidity. In a subgroup of patients without splenectomy or 99Tc colloid can be allowed to affect decisions about
or resection of the pancreatic tail, D2 lymphadenectomy removal or non-removal of lymph nodes in patients with
was associated with a drop in local recurrence rates in cancer of the stomach. If non-stained or non-radiolucent
the Dutch trial,85 and the best 5-year survival rates in areas are not removed, nodes occupied by cancer could be
a UK trial.71 Splenectomy is an especially adverse left behind. Table 4 shows that the detection rate of the
prognostic factor of its own and suppression of initial draining (ie, sentinel) node ranges from 95% to
T-cell function is thought to be one of the linking 100%. However, the sensitivity of detecting nodes involved
factors.88 Therefore, splenectomy should be done only by cancer as controlled by full D2 lymphadenectomy is in
in cases of locally advanced tumour of the upper third the range of 90% depending on the stage of tumours
of the stomach, tumours of the greater curvature, recruited. Although this approach seems promising, it is
and those of the gastric cardia. The incidence of lymph- yet to be fully tested. Future procedures might see patients
node metastases is between 10% and 25%, and if with T1 or small T2 lesions undergoing lymphadenectomy
splenectomy is advised, the pancreas should be limited to the area indicated by the sentinel node
preserved.89 procedure.

THE LANCET • Vol 362 • July 26, 2003 • www.thelancet.com 311

For personal use. Only reproduce with permission from The Lancet.
SEMINAR

Number of Method Detection Sensitivity Node concluded that the quality of surgical treatment was
patients rate (%) (%) positive poor.108 A substantial proportion of patients had had D0
and T patients resections—ie, no clearance of the lymphatics. A
category (%)
comparison of 5-year survival rates in patients with
Kitagawa96 127 T1 m99Tc 95% 92% 17% different stage cancers showed that surgery and
18 T2 Sn-colloid postoperative radiochemotherapy had the same rate of
Ichikura97 62 T1/2 dye 100% 87% 24%
ICG
success as those achieved in the randomised trials for D1
Hiratsuka 98
44 T1 dye 99% 90% 14% or D2 lymphadenectomy alone.109 Only 10% of patients
30 T2 ICG had received a D2 lymphadenectomy, and as a result
ICG=indocyan green 64% of controls had recurrence, which involved regional
Table 4: Results of sentinel node detection gastric cancer
sites in 72%, local sites in 29%, and distant sites in 18%.
Recently, surgical information about treatment of various
lymph node stations was re-analysed. The Maruyama
Reconstruction after total gastrectomy program to estimate the likelihood of disease in
Replacement of the resected stomach should prevent undissected regional node stations was applied,84 and
reflux from the intestine to the oesophagus and thus survival probability was calculated by Cox’s multivariate
prevent oesophagitis. Furthermore, measures should be regression. As a result, 54% of patients had undergone a
taken to avoid postoperative malabsorption and D0 lymphadenectomy and the index of unresected
maldigestion, because total gastrectomy leads to impaired disease proved to be an independent prognostic factor,
nutritional status and loss of more than 10% of the even after adjustment for linked variables such as T stage
preoperative weight. Reconstruction of the alimentary or the number of positive nodes.108 In this trial, surgical
canal after gastric resection has long been a point of undertreatment undermined survival estimates—the
debate and prospective randomised trials have been done results could be interpreted as has having treated residual
to assess whether a J-shaped pouch reservoir constructed disease by radiochemotherapy in a significant subgroup
from the proximal jejunum is needed to replace the of patients. After R0 resection of cancer of the stomach
stomach.99,100 Furthermore, investigators also sought to and adequate lymphadenectomy, adjuvant treatment
determine whether any reconstruction should be cannot be recommended outside of prospective
positioned as a Roux-en-Y oesophago-jejunostomy or randomised trials.
whether the duodenal passage should be maintained.
Because it is a technically simple procedure, Roux-en-Y Assessment of minimal residual disease as a
oesophago-jejunostomy is the preferred type of prognostic factor
anastomosis. A comparison of a J-pouch replacement for Apart from macrometastases in lymph nodes and other
the stomach with straight anastomosis shows that distant sites, micrometastases and single disseminated
functional results seem to be superior in the pouch group tumour cells are detected in bone marrow, blood, and
during the first year after surgery.99,100 However, patients lymph nodes in patients with gastric cancer. Tumour cell
who had this more sophisticated reconstruction procedure clusters of less than 2 mm with stromal reaction from
did not avoid weight loss or show an improved quality of surrounding tissue are called micrometastases. Minimal
life. Results of a randomised comparison of J-pouch residual disease is represented by single disseminated
reconstruction with or without maintainence of the tumour cells detected by cytological examination,
duodenal passage in 120 patients showed that there are no immunohistochemical analysis, or RT-PCR. Cytology
significant differences between the procedures with detects one epithelial cell per 100 cells, immuno-
respect to anastomotic dehiscence rates, general histochemical techniques detect one cell per 105 cells;
complications, mortality, operation time, or bodyweight, however, RT-PCR allows detection of one epithelial cell
Visick scoring,101 or Spitzer index.102,103 Differences per 107 cells.110 The higher the sensitivity, the less
between the procedures seemed to represent only an early specificity should be expected. There is much debate
adaptive process, and advantages could not be seen about whether these cells represent true metastatic cells
during longer follow-up. and precursors of overt metastases, or whether they have a
limited life span and are merely a sign of shedded tumour
Adjuvant treatment cells. Recently, the tumorigenic potential of epithelial cells
Many studies have investigated the use of postoperative has been proven in animal experiments in studies with
adjuvant systemic chemotherapy. Results of a meta- cells derived from oesophageal nodal metastases and
analysis104 showed that there is no evidence of a survival prostate cancer.111,112 Characterisation of those cells
advantage with this treatment. However, some data from showed less expression of proliferation markers (p120 and
non-randomised trials and one small randomised trial CK67) and HLA class 1 receptors are downregulated and
suggest that postoperative intraperitoneal chemotherapy might contribute to less activation of T-cell mediated
remains worthy of investigation.105,106 destruction.113
Recently, workers from a US multi-institutional In gastric cancer, disseminated epithelial cells can be
randomised trial of 556 patients claimed that adjuvant detected by antibodies against cytokeratin-18 and
radiochemotherapy improved postoperative median urokinase-type plasminogen activator (uPA).114 The
overall survival from 27 months to 36 months (p<0·005, detection rate of epithelial cells in bone marrow or lymph
hazard ratio 1·35) as well as relapse-free survival (1·52) nodes that stain negative in conventional haematoxylin
after a medium follow-up of 3 years.107 Improvement in and eosin (pN0) depends on the stage of disease;
survival rates could be shown through all stages of disease however, it was also reported for early cancer of
and was attributed to the 45 Gy of radiotherapy and three submucosal type.115 Carcinoembryonic antigen-mRNA
cycles of 5-fluorocacil (425 mg/m2) and leucovorin positive cells were detected in an increased range after
(20 mg/m2) that were given in addition to resection of surgical manipulation.116 The presence of disseminated
non-metastatic gastric cancer with curative intent. tumour cells in bone marrow is indicative of systemic
Initially, the investigators considered this regimen a disease even in early stages of cancer of the stomach. The
standard of care; however, during the study they extent of tumour-cell detection in bone marrow correlates

312 THE LANCET • Vol 362 • July 26, 2003 • www.thelancet.com

For personal use. Only reproduce with permission from The Lancet.
SEMINAR

12 Hansson LE, Nyren O, Bergstrom R, et al. Nutrients and gastric


Panel 5: Potential advances in the treatment and cancer risk: a population-based casecontrol study in Sweden.
diagnosis of gastric cancer in the next 5–10 years Int J Cancer 194; 57: 638–44.
13 Ogimoto I, Shibata A, Fukuda K. World Cancer Research Fund and
● increase to 50% the proportion of gastric cancers in the
American Institute of Cancer Research 1997 recommendations:
western hemisphere that are diagnosed early by improved applicability to digestive tract cancer in Japan. Cancer Causes Control
endoscopic methods 2000; 11: 9–23.
● explore the incidence of inherited gastric cancer and 14 Terry P, Lagergren J, Hansen H, Wolk A, Nyren O. Fruit and
locate the genomic defect vegetable consumption in the prevention of oesophageal and cardia
cancers. Eur J Cancer Prev 2001; 10: 365–69.
● eradication of H pylori, thus eliminating one causal agent
15 Meining A, Morgner A, Miehlke S, Bayerdörffer E, Stolte M.
● development of a vaccine against highly-invasive, tumour- Atrophy-meta-plasia-dysplasia-carcinoma sequence in the stomach: a
inducing strains of H pylori reality or merely a hypothesis? Best practice and research clinical
● clarification of the mechanisms of individualised reaction gastroenterology 2001; 15: 983–98
to H pylori infection (interleukins, vascular endothelial 16 La Vecchia C, Negri E, Franceschi S, Gentile A. Family history
growth factor) and the risk of stomach and colorectal cancer. Cancer 1992; 70:
50–55.
● ‘proof of principle’ that eradication of H pylori prevents
17 Hattori, T. Development of adenocarcinomas in the stomach. Cancer
development of gastric cancer 1986; 57: 1528–34.
● identification of the lymphatic drainage basin by combined 18 Kirchner T, Muller S, Hattori T, et al. Metaplasia, intraepithelial
endoscopic and laparoscopic sentinel node procedure in neoplasia and early cancer of the stomach are related to
T1b and T2 tumours, leading to limited lymphadenectomy dedifferentiated epithelial cells defined by cytokeratin-7 expression in
in most patients gastritis. Virchows Arch 2001; 439: 512–22.
19 Bamba M, Sugihara H, Kushima R, et al. Time-dependent
● establish a clear indication for neoadjuvant treatment and
expression of intestinal phenotype in signet ring cell carcinomas of
select patients by expected reponse to treatment the human stomach. Virchows Arch 2001; 438: 49–56.
● develop a method to fight peritoneal carcinosis (intra- 20 Marshall BJ, Warren JR. Unidentified curved bacilli in the stomach
abdominal antibodies, migration factor inhibitors, anti- of patients with gastritis and peptic ulceration. Lancet 1984; 1:
proliferative substances) 1311–15.
● characterisation of single tumour cells in bone marrow as 21 Miyaji H, Azuma T, Ito S, et al. Helicobacter pylori infection occurs
via close contact with infected individuals in early childhood.
a basis for directed adjuvant therapy J Gastroenterol Hepatol 2000; 15: 257–62.
22 Kikuchi S, Kurosawa M, Sakiyama T. Helicobacter pylori risk
with prognosis in patients who undergo resection of associated with sibship size and family history of gastric diseases in
Japanese adults. Jpn J Cancer Res 1998; 89: 1109–12.
curative intent. Therefore, epithelial cells in bone marrow 23 Kurosawa M, Kikuchi S, Inaba Y, Ishibashi T, Kobayashi F.
could be a selection criteria for adjuvant treatment if there Helicobacter pylori infection among Japanese children.
were active drugs or efficient strategies.117 However, by no J Gastroenterol Hepatol 2000; 15: 1382–85.
means should the detection of these cells direct a patient 24 Krumbiegel P, Herbarth O, Fritz G, et al. Helicobacter pylori
to treatment of purely palliative intent. At present, the prevalence in Leipzig’s 1998 school entries: methodology and first
results. Int J Hyg Environ Health 2000; 203: 11–16.
detection of disseminated epithelial cells in patients with 25 Parsonnet J, Vandersteen D, Goates J, Sibley RK, Pritikin J,
gastric cancer is an area of intense research but does not Chang Y. Helicobacter pylori infection in intestinal- and diffuse-type
yet have clinical consequences. gastric adenocarcinomas. J Natl Cancer Inst 1991; 83: 640–43.
Panel 5 shows the possibilities for future developments 26 Nomura A, Stemmermann GN, Chyou PH, Kato I,
in treatment of gastric cancer. Perez-Perez GI, Blaser MJ. Helicobacter pylori infection and gastric
carcinoma among Japanese Americans in Hawaii. N Engl J Med
1991; 325: 1132–36.
Conflict of interest statement 27 Parsonnet J, Friedman GD, Vandersteen DP, et al. Helicobacter pylori
None declared.
infection and the risk of gastric carcinoma. N Engl J Med 1991; 325:
1127–31.
28 Gallo N, Zambon C-F, Navaglia F, et al. Helicobacter pylori infection
References in children and adults: a single pathogen but a different pathology.
1 Allum WH, Griffin SM, Watson A, Colin-Jones D. Guidelines for the Helicobacter 2003; 8: 21–28.
management of oesophageal and gastric cancer. Gut 2002; 29 Uemura N, Okamoto S, Yamamoto S, et al. Helicobacter pylori
50 (suppl 5): v1–23. infection and the development of gastric cancer. N Engl J Med 2001;
2 Wayman J, Forman D, Griffin SM. Monitoring the changing pattern 345: 784–89.
of esophago-gastric cancer: data from a UK regional cancer registry. 30 Hansson LE, Nyren O, Hsing AW, et al. The risk of stomach cancer
Cancer Causes Control 2001; 12: 943–49. in patients with gastric or duodenal ulcer disease. N Engl J Med 1996;
3 Terry MB, Gaudet MM, Gammon MD. The epidemiology of gastric 335: 242–49.
cancer. Semin Radiat Oncol 2002; 12: 111–27. 31 El-Omar EM, Carrington M, Chow W-H, et al. Interleukin-I
4 WHO. WHO World Health Report, Geneva: World Health polymorphisms associated with increased risk of gastric cancer.
Organization, 1997. Nature 2002; ??: 398–402.
5 National Cancer Institute. Surveillance, epidemiology, and end 32 Boren T, Falk P, Roth KA, Larson G, Normark S. Attachment of
results: incidence, stomach cancer. http://www.seer.cancer.gov/ Helicobacter pylori to human gastric epithelium mediated by blood
faststats/html/inc_stomach.html (accessed July 12, 2002). group antigens. Science 1993; 262: 1892–95.
6 Parker SL, Tong T, Bolden S, Wingo PA. Cancer statistics, 1997. 33 Chow WH, Blaser MJ, Blot WJ, et al. An inverse relation between
CA Cancer J Clin 1997; 47: 5–27. cagA+ strains of Helicobacter pylori infection and risk of oesophageal
7 Dolan K, Sutton R, Walker S J, Morris A I, Campbell F, and gastric cardia adenocarcinoma. Cancer Res 1998; 58: 588–90.
Williams EM. New classification of of oesophageal and gastric 34 Zambon C-F, Navaglia F, Basso D, Rugge M, Plebani M.
carcinomas derived from changing patterns in epidemiology. Helicobacter pylori babA2, cagA, and s1 vacA genes work
Br J Cancer 1999; 80: 834–42. synergistically in causing intestinal metaplasia. J Clin Pathol 2003, 56:
8 Kranenbarg EK, van de Velde CJ. Gastric cancer in the elderly. 287–91.
Eur J Surg Oncol 1998; 24: 384–90. 35 Mahdavi J, Sonden B, Hurtig M, et al. Helicobacter pylori SabA
9 Anonymus. IARC Handbooks of Cancer Prevention, Vol 1 Geneva: adhesin in persistent infection and chronic inflammation. Science
International Agency for Research on Cancer, WHO; 1997. 2002; 297: 573–78.
10 Laurén P. The two histological main types of gastric carcinoma: 36 Woodward M, Tunstall-Pedoe H, McColl K. Helicobacter pylori
Diffuse and so-called intestinal-type carcinoma. infection reduces systemic availability of dietary vitamin C.
Acta Path Microbiol Imunol Scand 1965; 64: 31–49. Eur J Gastroenterol Hepatol 2001; 13: 233–37.
11 La Vacchia C, Ferraroni M, D’Avanzo B, Decarli A, Franceschi S. 37 Sobala GM, Schorah CJ, Shires S, et al. Effect of eradication of
Selected micronutrient intake and the risk of gastric cncer. Helicobacter pylori on gastric juice ascorbic acid concentrations. Gut
Cancer Epidemiol Biomarkers 1994; 3: 393–98. 1993; 34: 1038–41.

THE LANCET • Vol 362 • July 26, 2003 • www.thelancet.com 313

For personal use. Only reproduce with permission from The Lancet.
SEMINAR

38 Correa P, Fontham ET, Bravo JC, et al. Chemoprevention of gastric 65 Stein HJ, Feith M, Siewert JR. Cancer of the esophagogastric
dysplasia: randomised trial of antioxidant supplements and anti- junction. Surg Oncol 2000; 9: 35–41.
helicobacter pylori therapy. J Natl Cancer Inst 2000; 92: 1881–88. 66 de Manzoni G, Pedrazzani C, Pasini F, et al. Results of surgical
39 Wu MS, Shun CT, Wang HP, et al. Genetic alterations in gastric treatment of adenocarcinoma of the gastric cardia. Ann Thorac Surg
cancer: relation to histological subtypes, tumor stage and Helicobacter 2002; 73: 1035–40.
pylori infection. Gastroenterology 1997; 112: 1457–65. 67 Cuschieri A, Fayers P, Fielding J, et al, for the Surgical
40 Schlemper R J, Itabashi M, and Kato Y. Differences and diagnostic Cooperative Group. Postoperative morbidity and mortality after
criteria for gastric carcinoma between Japanese and western D1 and D2 resections for gastric cancer: preliminary results of
pathologists. Lancet 1997; 349: 1725–29. the MRC randomised controlled surgical trial. Lancet 1996; 347:
41 de Dombal FT, Price AB, Thompson H, et al. The British Society of 995–99.
Gastroenterology early gastric cancer/dysplasia survey: an interim 68 Bottcher K, Siewert JR, Roder JD, Busch R, Hermanek P, Meyer HJ.
report. Gut 1990; 31: 115–20. [Risk of surgical therapy of stomach cancer in Germany. Results of
42 Schlemper RJ, Riddell RH, Kato Y. The Vienna classification of the German 1992 Stomach Cancer Study. German Stomach Cancer
gastrointestinal epithelial neoplasia. Gut 2000; 47: 251–55. Study Group (‘92)] in german. Chirurg 1994; 65: 298–306.
43 Hunerbein M, Ghadimi BM, Haensch W, Schlag PM. 69 Wanebo HJ, Kennedy BJ, Chmiel J, Steele G, Winchester D,
Transendoscopic ultrasound of esophageal and gastric cancer using Osteen R. Cancer of the stomach. A patient care study by the
miniaturized ultrasound catheter probes. Gastrointest Endosc 1998; American College of Surgeons. Ann Surg 1993; 218: 583–92.
48: 371–75. 70 Akoh JA, Macintyre IM. Improving survival in gastric cancer: review
44 Bhunchet E, Hatakawa H, Sakai Y, Shibata T. Fluorescein electronic of 5-year survival rates in English language publications from 1970.
endoscopy: a novel method for detection of early stage gastric cancer Br J Surg 1992; 79: 293–99.
not evident to routine endoscopy. Gastrointest Endosc 2002; 55: 71 Cuschieri A, Weeden S, Fielding J, et al. Patient survival after D1
562–71. and D2 resections for gastric cancer: long-term results of the MRC
45 Shimoyama S, Yasuda H, Mafune K, Kaminishi M. Indications of a randomized surgical trial. Surgical Co-operative Group. Br J Cancer
minimized scope of lymphadenectomy for submucosal gastric cancer. 1999; 79: 1522–30.
Ann Surg Oncol 2002; 9: 625–31. 72 Siewert JR, Bottcher K, Stein HJ, Roder JD. Relevant prognostic
46 Gotoda T, Yanagisawa A, Sasako M, et al. Incidence of lymph node factors in gastric cancer: ten-year results of the German Gastric
metastasis from early gastric cancer: estimation with a large number Cancer Study. Ann Surg 1998; 228: 449–61.
of cases at two large centers. Gastric.Cancer 2000; 3: 219–25. 73 Ajani JA, Mansfield PF, Lynch PM, et al. Enhanced staging and all
47 Hiki Y, Sakakibara Y, Mieno H, Shimao H, Kobayashi N, Katada N. chemotherapy preoperatively in patients with potentially resectable
Endoscopic treatment of gastric cancer. Surg Endosc 1991; 5: 11–13. gastric carcinoma. J Clin Oncol 1999; 17: 2403–11.
48 Tani M, Takeshita K, Inoue H, Iwai T. Adequate endoscopic 74 Fink U, Ott K, Dittler HJ, et al. Neoadjuvant cisplatinum, leucovorin
mucosal resection for early gastric cancer obtained from the and fluorouracil (PLF) in adequately staged patients with locally
dissecting microscopic features of the resected specimens. advanced gastric carcinoma. Proc Am Soc Clin Oncol 1999; 18: 272.
Gastric Cancer 2001; 4: 122–31. 75 Kang Y, Choi D, Im Y, et al. A phase III randomized comparison of
49 Ohgami M, Otani Y, Kumai K, Kubota T, Kim YI, Kitajima M. neoadjuvant chemotherapy followed by surgery versus surgery for
Curative laparoscopic surgery for early gastric cancer: five years locally advanced stomach cancer. Proc Am Soc Clin Oncol 1996;.
experience. World J Surg 1999; 23: 187–92. 15: 215.
50 Nakajima T. Gastric cancer treatment guidelines in Japan. 76 Songun I, Keizer HJ, Hermans J, et al. Chemotherapy for operable
Gastric Cancer 2002; 5: 1–5. gastric cancer: results of the Dutch randomised FAMTX trial. The
Dutch Gastric Cancer Group (DGCG). Eur J Cancer 1999; 35:
51 Berndt H, Gütz HJ. Laparoskopie bei Magenkrebs. Z Gastroenterol
558–62.
1965; 3: 317–20.
77 Skoropad VY, Berdov BA, Mardynski YS, Titova LN. A prospective,
52 Burke EC, Karpeh MS, Conlon KC, Brennan MF. Laparoscopy in
randomized trial of pre-operative and intraoperative radiotherapy
the management of gastric adenocarcinoma. Ann Surg 1997; 225:
versus surgery alone in resectable gastric cancer. Eur J Surg Oncol
262–77.
2000; 26: 773–79.
53 Hunerbein M, Rau B, Hohenberger P, Schlag PM. The role of
78 Zhang ZX, Gu XZ, Yin WB, Huang GJ, Zhang DW, Zhang RG.
staging laparoscopy for multimodal therapy of gastrointestinal cancer.
Randomized clinical trial on the combination of preoperative
Surg Endosc 1998; 12: 921–25.
irradiation and surgery in the treatment of adenocarcinoma of gastric
54 Feussner H, Kraemer SJM, Siewert JR. Extended diagnostic cardia (AGC)—report on 370 patients. Int J Radiat Oncol Biol Phys
laparoscopy in gastric cancer. In: Hohenberger P, Conlon KC, eds. 1998; 42: 929–34.
Staging laparoscopy. Berlin: Springer, 2002. 83–95. 79 Melcher AA, Mort D, Maughan TS. Epirubicin, cisplatin and
55 Stell DA, Carter CR, Stewart I, Anderson JR. Prospective comparison continuous infusion 5-fluorouracil (ECF) as neoadjuvant
of laparoscopy, ultrasonography and computed tomography in the chemotherapy in gastro-oesophageal cancer. Br J Cancer 1996; 74:
staging of gastric cancer. Br J Surg 1996; 83: 1260–62. 1651–54.
56 D’Ugo DM, Persiani R, Caracciolo F, Ronconi P, Coco C, 80 Cascinu S, Labianca R, Alessandroni P, et al. Intensive weekly
Picciocchi A. Selection of locally advanced gastric carcinoma by chemotherapy for advanced gastric cancer using fluorouracil,
preoperative staging laparoscopy. Surg Endosc 1997; 11: 1159–62. cisplatin, epi-doxorubicin, 6S-leucovorin, glutathione, and filgrastim:
57 Kocher HM, Linklater K, Patel S, Ellul JP. Epidemiological study of a report from the Italian Group for the Study of Digestive Tract
oesophageal and gastric cancer in southeast England. Br J Surg 2001; Cancer. J Clin Oncol 1997; 15: 3313–19.
88: 1249–57. 81 Allum W, Cunningham D, Weeden S, for the UK NCRI Upper GI
58 Corley DA, Buffler PA. Oesophageal and gastric cardia Clinical Studies Group. Perioperative chemotherapy in operable
adenocarcinomas: analysis of regional variation using the Cancer gastric and lower oesophageal cancer: randomsied controlled trial
Incidence in Five Continents database. Int J Epidemiol 2001; 30: (the MAGIC trial). Proc Am Soc Clin Oncol 2003; 22: A998.
1415–25. 82 Bozzetti F, Marubini E, Bonfanti G, et al. Total versus subtotal
59 Ye W, Chow WH, Lagergren J, Yin L, Nyren O. Risk of gastrectomy: surgical morbidity and mortality rates in a multicenter
adenocarcinomas of the esophagus and gastric cardia in patients with Italian randomized trial. The Italian Gastrointestinal Tumor Study
gastroesophageal reflux diseases and after antireflux surgery. Group. Ann Surg 1997; 226: 613–20.
Gastroenterology 2001; 121: 1286–93. 83 Inoue Y. About the lymphatic system of the stomach, duodenum,
60 Corvalan A, Koriyama C, Akiba S, et al. Epstein-Barr virus in gastric pancreas and diaphragm (in Japanese). Acta Anat Nipponica 1936;
carcinoma is associated with location in the cardia and with a diffuse 35–117.
histology: a study in one area of Chile. Int J Cancer 2001; 94: 527–30. 84 Siewert JR, Kestlmeier R, Busch R, et al. Benefits of D2 lymph
61 Fukayama M, Chong JM, Uozaki H. Pathology and molecular node dissection for patients with gastric cancer and pN0 and pN1
pathology of Epstein-Barr virus-associated gastric carcinoma. lymph node metastases [see comments]. Br J Surg 1996; 83:
Curr Top Microbiol Immunol 2001; 258: 91–102. 1144–47.
62 Aikou T, Natugoe S, Tenabe G, Baba M, Shimazu H. Lymph 85 Bonenkamp JJ, Hermans J, Sasako M, van-de-Velde CJ. Extended
drainage originating from the lower esophagus and gastric cardia as lymph-node dissection for gastric cancer. Dutch Gastric Cancer
measured by radioisotope uptake in the regional lymph nodes Group. N Engl J Med 1999; 340: 908–14.
following lymphoscintigraphy. Lymphology 1987; 20: 145–51. 86 Bonenkamp JJ, Songun I, Hermans J, et al. Randomised comparison
63 de Manzoni G, Pedrazzani C, Di Leo A, et al. Metastases to the para- of morbidity after D1 and D2 dissection for gastric cancer in 996
aortic lymph nodes in adenocarcinoma of the cardia. Eur J Surg Dutch patients [see comments]. Lancet 1995; 345: 745–48.
2001; 167: 413–18. 87 Dent DM, Madden MV, Price SK. Randomized comparison of R1
64 Siewert JR, Stein HJ. Classification of adenocarcinoma of the and R2 gastrectomy for gastric carcinoma. Br J Surg 1988; 75:
oesophagogastric junction. Br J Surg 1998; 85: 1457–59. 110–12.

314 THE LANCET • Vol 362 • July 26, 2003 • www.thelancet.com

For personal use. Only reproduce with permission from The Lancet.
SEMINAR

88 Wolf HM, Eibl MM, Georgi E, et al. Long-term decrease of 104 Earle CC, Maroun JA. Adjuvant chemotherapy after curative
CD4+CD45RA+ T cells and impaired primary immune response resection for gastric cancer in non-Asian patients: revisiting a
after post-traumatic splenectomy. Br J Haematol 1999; 107: 55–68. meta-analysis of randomised trials. Eur J Cancer 1999; 35: 1059–64.
89 Kitamura K, Nishida S, Ichikawa D, et al. No survival benefit from 105 Rosen HR, Jatzko G, Repse S, et al. Adjuvant intraperitoneal
combined pancreaticosplenectomy and total gastrectomy for gastric chemotherapy with carbon-adsorbed mitomycin in patients with
cancer. Br J Surg 1999; 86: 119–22. gastric cancer: results of a randomized multicenter trial of the
90 Kampschoer GH, Maruyama K, van de Velde CJ, Sasako M, Austrian Working Group for Surgical Oncology. J Clin Oncol 1998;
Kinoshita T, Okabayashi K. Computer analysis in making 16: 2733–38.
preoperative decisions: a rational approach to lymph node dissection 106 Wansik Y, Whang I, Suh I, et al. Prospective rndomized trial of early
in gastric cancer patients. Br J Surg 1989; 76: 905–08. postoperative intraperitoneal chemotherapy as an adjuvant to
91 Bollschweiler E, Boettcher K, Hoelscher AH, et al. Preoperative resectable gastric cancer. Ann Surg 1998; 228: 347–54.
assessment of lymph node metastases in patients with gastric cancer: 107 MacDonald JS, Smalley SR, Benedetti J, Hundahl SA, Estes NC,
evaluation of the Maruyama computer program. Br J Surg 1992; 79: Stemmermann GN, Haller DG, Ajani JA, Gunderson LL, Jessup JM,
156–60. Martenson JA. Chemoradiotherapy after surgery compared with
92 Guadagni S, de-Manzoni G, Catarci M, et al. Evaluation of the surgery alone for adenocarcinoma of the stomach or gastroesophageal
Maruyama computer program accuracy for preoperative estimation of junction. N Engl J Med 2001; 10: 725–30.
lymph node metastases from gastric cancer. World J Surg 2000; 24: 108 Hundahl SA, Macdonald JS, Benedetti J, Fitzsimmons T. Surgical
1550–58. treatment variation in a prospective, randomized trial of
93 Weinberg JGEM. Identification of regional lymph nodes by means chemoradiotherapy in gastric cancer: the effect of undertreatment.
of a vital staining dye during surgery of gastric cancer. Ann Surg Oncol 2002; 9: 278–86.
Surg Gynecol Obstet 1950; 90: 561–67. 109 Schwartz RE. Postoperative adjuvant chemoradiation therapy for
94 Catarci M, Guadagni S, Zaraca F, et al. Prospective randomized patients with resected gastric cancer: intergroup 116. J Clin Oncol
evaluation of preoperative endoscopic vital staining using CH-40 for 2001; 19: 1879–84.
lymph node dissection in gastric cancer [see comments]. 110 Mattano LA Jr, Moss TJ, Emerson SG. Sensitive detection
Ann Surg Oncol 1998; 5: 580–84. of rare circulating neuroblastoma cells by the reverse
95 Takahashi T, Sawai K, Hagiwara A, Takahashi S, Seiki K, transcriptase-polymerase chain reaction. Cancer Res 1992; 52:
Tokuda H. Type-oriented therapy for gastric cancer effective for 4701–05.
lymph node metastasis: management of lymph node metastasis using 111 Hosch S, Kraus J, Scheunemann P, et al. Malignant potential and
activated carbon particles adsorbing an anticancer agent. cytogenetic characteristics of occult disseminated tumor cells in
Semin Surg Oncol 1991; 7: 378–83. esophageal cancer. Cancer Res 2000; 60: 6836–40.
96 Kitagawa Y, Fujii H, Mukai M, Kubota T, Otani Y, Kitajima M. 112 Molino A, Colombatti M, Bonetti F, et al. A comparative analysis of
Radio-guided sentinel node detection for gastric cancer. Br J Surg three different techniques for the detection of breast cancer cells in
2002; 89: 604–08. bone marrow. Cancer 1991; 67: 1033–36.
97 Ichikura T, Morita D, Uchida T, et al. Sentinel node concept in 113 Putz E, Witter K, Offner S, et al. Phenotypic characteristics
gastric carcinoma. World J Surg 2002; 26: 318–22. of cell lines derived from disseminated cancer cells in bone
98 Hiratsuka M, Miyashiro I, Ishikawa O, et al. Application of marrow of patients with solid epithelial tumors: establishment of
sentinel node biopsy to gastric cancer surgery. Surgery 2001; 129: working models for human micrometastases. Cancer Res 1999; 59:
335–40. 241–48.
99 Troidl H, Kusche J, Vestweber KH, Eypasch E, Maul U. Pouch 114 Allgayer H, Heiss MM, Riesenberg R, et al. Urokinase plasminogen
versus esophagojejunostomy after total gastrectomy: a randomized activator receptor (uPA-R): one potential characteristic of metastatic
clinical trial. World J Surg 1987; 11: 699–712. phenotypes in minimal residual tumor disease. Cancer Res 1997; 57:
100 Schwarz A, Buchler M, Usinger K, et al. Importance of the duodenal 1394–99.
passage and pouch volume after total gastrectomy and reconstruction 115 Choi HJ, Kim YK, Kim YH, Kim SS, Hong SH. Occurrence and
with the Ulm pouch: prospective randomized clinical study. prognostic implications of micrometastases in lymph nodes from
World J Surg 1996; 20: 60–66. patients with submucosal gastric carcinoma. Ann Surg Oncol 2002; 9:
101 Visick A H. Measured radical gastrectomy. Lancet 1948; 2: 505. 13–19.
102 Spitzer W O, Dobson A J, Hall J, et al. Measuring the quality of olife 116 Miyazono F, Natsugoe S, Takao S, et al. Surgical maneuvers
of cancer patients: a concise QL-index for use by physicians. enhance molecular detection of circulating tumor cells during gastric
J Chronic Dis 1981; 34: 585. cancer surgery. Ann Surg 2001; 233: 189–94.
103 Fuchs KH, Thiede A, Engemann R, Deltz E, Stremme O, 117 Jauch KW, Heiss MM, Gruetzner U, et al. Prognostic significance of
Hamelmann H. Reconstruction of the food passage after total bone marrow micrometastases in patients with gastric cancer.
gastrectomy: randomized trial. World J Surg 1995; 19: 698–705. J Clin Oncol 1996; 14: 1810–17.

THE LANCET • Vol 362 • July 26, 2003 • www.thelancet.com 315

For personal use. Only reproduce with permission from The Lancet.

You might also like