You are on page 1of 11

REVIEWS

Gastrin — active participant or


bystander in gastric carcinogenesis?
Susan A. Watson, Anna M. Grabowska, Mohamad El-Zaatari and Arjun Takhar
Abstract | Gastrin is a pro-proliferative, anti-apoptotic hormone with a central role in
acid secretion in the gastric mucosa and a long-standing association with malignant
progression in transgenic mouse models. However, its exact role in human gastric
malignancy requires further validation. Gastrin expression is tightly regulated by two
closely associated hormones, somatostatin and gastrin-releasing peptide, and aspects of
their interaction may be deregulated during progression to gastric adenocarcinoma.
Furthermore, agonists and antagonists of the receptors for all three hormones have
shown modest clinical efficacy against gastric adenocarcinoma, which might provide
useful information on the future combined use of these agents.

Proton pump inhibitors


The polypeptide hormone gastrin induces gastric acid would therefore represent a therapeutic modality for
Agents that block acid secretion during the passage of food in the stomach, gastrin-sensitive malignancies. However, we now know
secretion in the stomach and thereby maintaining the appropriate acidic pH for the that gastrin might also have a role in cell differentiation
are used for the treatment of function of gastric digestive enzymes. However, since and carcinogenesis, within both proximal and distal loca-
stomach ulcers.
the mid-1980s, several research groups have focused on the tions of the GI tract. Gastrin is a diverse transcriptional
Enterochromaffin-like cell proposal that gastrin also increases gastrointestinal (GI) activator, mediating gene expression that is associated
A neuroendocrine cell, found carcinogenesis. Gastrin expression is commonly increased with cell division, invasion, angiogenesis and anti-apop-
in the gastric mucosa, that through two factors: infection with Helicobacter pylori and totic activity, which are all pivotal in the gain of malig-
secretes histamine following administration of proton pump inhibitors (PPIs), the former nant potential. Furthermore, the source of gastrin can
hormonal stimulation.
being associated with gastric adenocarcinoma. However, be either endocrine, through gastric mucosal secretion,
Antro-pyloric mucosa it is still unclear whether gastrin is a central player or a or of tumour origin, through de novo activation of the
The lower region of the secondary phenomenon in the development of gastric gastrin gene following early mutational events, including
stomach spanning the antrum adenocarcinoma. Nevertheless, gastrin can be regarded adenomatosis polyposis coli (APC) gene mutations, as
and pylorus.
as a pioneer in investigations to determine the multifunc- gastrin is a target of β-catenin-mediated transcription.
tional role of gut hormones in both proximal and distal Importantly, gastrin expression is normally tightly regu-
GI carcinogenesis. The role of gastrin in human gastric lated by the positive regulator gastrin-releasing peptide
carcinogenesis will ultimately be determined in clinical (GRP, the mammalian homologue of bombesin) and the
trials, but the field has been sustained by a progression family of related hormones, acting together with a nega-
of new concepts over the past 20 years that have emerged tive regulator, somatostatin. All have been independently
mainly from mouse models. examined for their role in GI carcinogenesis, resulting in
Hypergastrinaemia has been shown to promote the generation of potential therapeutic candidates.
development of gastric adenocarcinoma in mice and This Review explores the emerging role of these hor-
yet clinical conditions that are associated with hyper- mones in gastric carcinogenesis, with a focus on gastrin,
gastrinaemia in humans, such as the Zollinger–Ellison as, to date, information on how GRP and somatostatin
syndrome, result in the development of hyperplasia of contribute to gastric malignancy is more limited. Where
Academic Unit of Cancer enterochromaffin-like (ECL) cells and carcinoid tumours. possible, however, any findings have been included.
Studies, University of Here this discrepancy will be discussed by contrasting
Nottingham, Nottingham, pathological changes in transgenic mouse models with Normal roles of gastrin, somatostatin and GRP
NG7 2UH, UK. those encountered in humans. The main site of gastrin synthesis is the G cell within the
Correspondence to S.A.W.
In the early 1980s the concept was simple: serum- antro-pyloric mucosa (FIG. 1a), a classic gut endocrine cell with
e-mail: sue.watson@
nottingham.ac.uk associated gastrin binds a cell-surface receptor and microvilli on the luminal surface that detect food within
doi:10.1038/nrc2014 induces a mitogenic stimulus. Blocking this receptor the stomach. Gastrin is translated as a 101-amino-acid

936 | DECEMBER 2006 | VOLUME 6 www.nature.com/reviews/cancer


© 2006 Nature Publishing Group
REVIEWS

At a glance epidermal growth factor (EGF)18,19, but does not affect


basal transcription. In the fundus, the release of soma-
• Coordinated activity between somatostatin and gastrin-releasing peptide (GRP) tostatin by D cells in response to neurohumoral agents
regulates secretion of gastrin from G cells in a negative and positive manner, mediates direct inhibition of gastric acid secretion by the
respectively, to facilitate the release of gastric acid from parietal cells.
parietal cell20 and indirect inhibition by reducing ECL-
• In transgenic mouse models, hypergastrinaemia contributes to the development of cell histamine-release (FIG. 1b) through the somatostatin
atrophy in the proximal gastric mucosa through the induction of parietal cell receptor subtype 2 (SSTR2) (REFS 21,22).
apoptosis, leading to the development of gastric adenocarcinoma. This phenomenon
Two forms of GRP (a 27-amino-acid peptide and a
has not been confirmed in human gastric cancer.
carboxy-10-amino-acid peptide fragment23), released
• Infection with Helicobacter pylori is associated with increased gastrin levels in serum from the neuronal fibres of the stomach, facilitate gas-
and components of the bacterium might activate the gastrin promoter directly and
trin secretion by acting as secretagogues (FIG. 1a). Gastrin
indirectly. Conversely, infection with H. pylori reduces somatostatin activity.
Furthermore, GRP, somatostatin and gastrin seem to modulate the inflammatory release is also mediated by a GRP homologue, bombesin24.
response to the bacterium. However, in the fundic mucosa, GRP has a negative
• Gastrin and GRP have well-documented pro-carcinogenic roles, particularly in animal
effect, mediating somatostatin release by D cells25 and
models, that affect proliferation, angiogenesis and apoptosis, whereas the potential reducing acid secretion.
pro-apoptotic signal of somatostatin is lost during carcinogenesis owing to weak Each of the hormones bind to G-protein-coupled
expression of somatostatin receptor isoforms. receptors, for which there are various functional isoforms
• Clinical agonists and antagonists of the three hormones have shown modest activity expressed within the gastric mucosa (TABLE 1).
in patients with gastrointestinal cancer, and rational combinations of these agents
might prove to be more clinically beneficial. Gastric cancer
Gastric cancer can be divided into two main categories,
diffuse and intestinal, which are based on the Lauren
Oxyntic mucosa
precursor1 and undergoes sequential processing to the classification. The diffuse, or signet ring, type, which is
The region within the amidated end-products gastrin 17 and gastrin 34 (REFS 2,3). more common in Western populations, is associated with
gastric mucosa that contains It is exocytosed from secretory granules along the the proximal area of the stomach, and tumours tend to
acid-secreting glands. baso-lateral border as a result of endocrine, paracrine, be poorly differentiated. The intestinal type, which typi-
neurocrine and local luminal factors4 and is transported cally involves the distal stomach, is common in Asian and
Parietal cells
Cells of the gastric mucosa to its site of action, the oxyntic mucosa (FIG. 1a), in an endo- elderly populations and is associated with the develop-
that produce gastric acid in crine manner. There, it interacts with the cholecystokinin 2 ment of intestinal-like glands26–28. Infection with H. pylori
response to histamine, (CCK2) receptors on ECL cells, potentiating the release of is associated with 72% of antral gastric cancers and it
gastrin or stimulation by histamine, which induces gastric acid secretion by parietal leads to a ninefold increase in gastric cancer risk29.
the vagal nerve.
cells5–8 (FIG. 1b). There have been some reports that gastrin The gastric epithelium undergoes continuous renewal
Fundus can directly stimulate acid release from parietal cells, which of stem cells in the isthmus niche, which differentiate
The dome-shaped upper part are based on the observation that histamine antagonists are into specialized cell types. The development of gastric
of the stomach. unable to completely block acid secretion in patients, but cancer is associated with deregulation of these prolif-
this is controversial9–11. The most convincing evidence for eration30 and differentiation processes31,32 that regulate
Secretagogue
A molecule that stimulates the expression of CCK2 receptors in parietal cells comes the renewal and specification of gastric stem cells into
the secretion of gastric or from histological studies in the human stomach using specialized gastric epithelial cell types.
pancreatic peptides. immunohistochemistry and in situ reverse transcription
PCR. However, other studies have been unable to dem- Roles of the hormones in cell differentiation
Lauren classification
A histological method for
onstrate the expression of CCK2 receptors in parietal cells The glandular structure of the main body of the gastric
classifying gastric tumours into using a fluorescence-labelled CCK, which is similar to mucosa (fundus and corpus) consists of stem cells that
intestinal or diffuse type. The gastrin, or a gastrin analogue. Expression of CCK2 recep- reside in the isthmus region, which migrate bidirec-
intestinal type is characterized tors has been described in isolated parietal cells12–14 and tionally to give rise to several differentiated cell types,
by the formation of cohesive
gastrin potentiated the action of histamine in these cells15 including mucous-secreting pit and neck cells, pepsino-
gland-like tubular structures,
whereas the diffuse type but, although these cell preparations were enriched for gen-secreting base cells, histamine-releasing ECL cells,
involves infiltration and parietal cells, they might also have included ECL cells. In and acid-secreting parietal cells.
thickening of the stomach other studies, gastrin only stimulated acid release when the A potential mechanism by which gastrin might influ-
wall by individual cells. release of histamine was also stimulated16, and in histamine ence the gastric stem-cell niche is through the induction
Signet ring
receptor H2 (Hrh2) knockout mice neither histamine nor of regenerating islet-derived 1α (REG1α), which is
A poorly differentiated diffuse gastrin was able to induce gastric acid secretion17. upregulated in conditions of hypergastrinaemia and
type of gastric cancer in which However, the caveat to this is that canine parietal in gastric adenocarcinoma 33, and seems to induce
the nuclei of glandular cells cells were used, indicating that there might be species stem-cell proliferation34,35. A transgenic mouse model
are pushed aside by mucin
differences in the receptor expression profiles5. overexpressing Reg1 had significantly increased gas-
vacuoles within the cell, giving
them the appearance of a Gastrin release is controlled by somatostatin and tric mucosal thickness and proliferation. Increased Reg1
signet ring. GRP in a negative and positive manner, respectively expression also led to a roughly twofold increase in parietal
(FIG. 1a). Somatostatin, secreted by D cells in the antrum and zymogenic cell numbers. Therefore, Reg1 might have
Isthmus in response to luminal acid, inhibits gastrin release by a role in promoting proliferation of precursor cells and
The region of the gastric
gland between the pit and
G cells; somatostatin is also able to inhibit upregulation of differentiation into parietal and ECL cells36, and seems
neck that contains immature gastrin gene expression by G cells in response to external to be important in mediating the effects of gastrin on
progenitor cells. mediators, such as dibutyryl-cyclic-AMP, carbachol and the proliferation and differentiation of ECL cells.

NATURE REVIEWS | CANCER VOLUME 6 | DECEMBER 2006 | 937


© 2006 Nature Publishing Group
REVIEWS

a Oesophagus GRP receptor + Ach


GRP nerve Vagus
Luminal D cell GRP

Pit cells
nerve
Cardia Fundus acid
Pit cell H+
Gastrin Capillary
Stomach receptors Stem cell Somatostatin bed

Isthmus
Preganglionic
Antrum Corpus CCK2R cholinergic
SSTR2 nicotinic
Pylorus nerves
Amino G cell
ECL cell
Base
acids, Gastrin
peptides
G cells Gastrin G cell
D cell GRP
Duodenum GRP nerve
Mucous cell GRP receptor + Ach

Postganglionic cholinergic muscarinic nerves


b Oesophagus
–Ach + Ach
Vagus
D cell
Pit cells

GRP nerve nerve


+ Ach
Cardia Fundus GRP
Pit cell Ach
Gastrin
receptors ECL cell +
Stomach Stem cell Somatostatin
Isthmus

Preganglionic
Antrum Corpus Parietal cell cholinergic
SSTR2 nicotinic
Neck

Pylorus nerves
Histamine
H+
Mucous cell H+ Parietal cell H+ HRH2
Base

G cells Gastrin ECL cell CCK2R Gastrin


D cell
Duodenum + Ach + Ach
Base cell

Postganglionic cholinergic muscarinic nerves


Figure 1 | The macroscopic and microscopic cellular interactions of the human gastric antro-pyloric and corpic/
fundic mucosa. a | The main site of gastrin synthesis is the gastrin-containing G cell within the antro-pyloric mucosa. A
representative gland is shown in the middle panel. The G cell is a classic gut endocrine cell with microvilli on the
luminal surface that mediate the detection of food within the stomach. Gastrin is released into the circulation from
secretory granules along the baso-lateral border (right panel), in close proximity to mucosal blood vessels4. Gastrin is
released by explosive exocytosis, an activity modulated by endocrine, paracrine, neurocrine and local luminal factors4.
Somatostatin, which is secreted by D cells, controls gastrin secretion by the G cell through modification of gastrin gene
transcription. Furthermore, gastrin-releasing peptide (GRP), present in the neuronal fibres innervating the antral
mucosa, facilitates gastrin secretion by acting as a secretagogue. b | Following exocytosis from the G cell, gastrin is
transported to its site of action, the oxyntic mucosa of the stomach, in an endocrine manner. Once there, it interacts
with the CCK2 receptor (CCK2R) on enterochromaffin-like (ECL) cells, potentiating the release of histamine, which
then interacts with the parietal cell to induce gastric acid secretion (right panel)5–8. Facilitation of this process is further
mediated by gastrin regulation of the expression and activity of histidine decarboxylase, which increases histamine
production159. D cells are also present within the oxyntic mucosa and somatostatin release can inhibit gastric acid
secretion by local diffusion through the intercellular space or the local mucosal circulation4. Fundic somatostatin works
in two ways, by inhibiting gastric acid secretion and by modulating the gastric acid response to gastrin4,160. GRP can
also have a negative regulatory role in the fundic mucosa by directly mediating the release of somatostatin by the D
cell25. Ach, acetylcholine; HRH2, histamine receptor H2; SSTR2, somatostatin receptor subtype 2. Right panels of a and
b are adapted with permission from REF. 4 © (1994) Balliere Tindall.

A second factor, sonic hedgehog (SHH), is required the kinase PKB (also known as AKT) regulates gastric
for maintaining gastric mucosal development by control- acid secretion42 and is essential for SHH signalling43,44.
Corpus ling the processes of proliferation and apoptosis through Gastrin, somatostatin and GRP therefore have the poten-
The main body of the stomach.
activation of the Gli family of transcription factors37–39. tial to regulate SHH transcription through gastric acid
Zymogenic cell Deregulation might have a role in early pre-malignant secretion45, which warrants further investigation.
Also known as chief cells, these change. Gastrin has been shown to indirectly induce SHH
secrete pepsinogen and are mRNA expression (M.E.-Z., A.M.G. and S.A.W., unpub- Gastrin and somatostatin mouse models
characterized by spherical lished observations) and the proteolytic processing and Perturbation of the gastrin–somatostatin axis has been
zymogen granules. They
differentiate as they migrate
activation of SHH protein through acid secretion40. A rela- achieved in several transgenic mouse systems. The
downwards from the isthmus to tionship between gastric acid secretion and SHH expres- transgenic mouse that overexpresses human gastrin
the base of the gastric gland. sion has been confirmed in parietal cells41. For example, (InsGas mouse) develops corpus atrophy, intestinal-like

938 | DECEMBER 2006 | VOLUME 6 www.nature.com/reviews/cancer


© 2006 Nature Publishing Group
REVIEWS

Table 1 | CCK2 receptor, somatostatin and GRP isoforms associated with the gastric mucosa
Receptor isoform Characteristics or normal location Receptor function References
CCK2 receptor
Classical Expressed by ECL cells, parietal cells, Gastric acid secretion, 161–165
gastric stem cells and neuroendocrine histamine release and parietal
cells cell differentiation
Intron IV splice variant Gastric mucosa adjacent to gastric Not known in gastric setting 166
tumour
Truncated in N-terminal Expression with classical receptor in Not known 167
domain normal gastric mucosa
Truncated in extracellular Only in gastric and colorectal cancer Not known 168
and transmembrane domains (sporadic and hereditary)
Somatostatin receptors
SSTR2A G cells, ECL cells and nerve fibres Histamine release, 156,169–172
anti-proliferative
SSTR2B Parietal cells Pro-proliferative 172
SSTR3 Gastric mucosa Pro-apoptotic 115
GRP receptors
BB2 Gastric antrum G-cell secretagogue 114,173
BB2, bombesin; CCK2, cholecystokinin 2; ECL, enterochromaffin-like; GRP, gastrin-releasing peptide; SSTR, somatostatin receptor.

metaplastic lesions, dysplasia and ultimately gastric eration and survival of cells within the gastric isthmus,
adenocarcinoma within 20 months46. Conversely, in a which seem to be sensitive to EGF receptor ligands52,53.
gastrin-deficient mouse model, parietal cells are lost Interferon-γ production also suppresses somatostatin
and antral inflammation is increased, resulting in antral expression levels and this affects the production of the
atrophy, intestinal metaplasia and antral tumours47. T helper cell type 2 (T H 2) cytokine IL4, which is
Gastrin overexpression facilitates cancer development, stimulated by somatostatin54 and has a role in reversing
which is based on an amplified mitogenic effect, whereas H. pylori-induced gastritis.
gastrin loss potentially leads to loss of gastric acid secretion Somatostatin has also been shown to inhibit dendritic
and bacterial overgrowth in the stomach, which can cell (DC) responsiveness to H. pylori55, reducing the
also result in cancer development. Somatostatin- severity of gastritis through the stimulation of IL4
deficient mice have hyperplastic fundi and increased production. Preliminary work by our group supports
parietal cell numbers but do not develop tumours47. a hormonal influence of gastrin on DC maturation56.
These transgenic models confirm that deregulation of Recently, Makarenkova et al.57 described how GRP and
gastrin is a crucial factor in mouse gastric carcinogenesis bombesin lead to dose-dependent inhibition of DC
and, where there is development of corpus atrophy, maturation. Based on the above, coordinated action of
G cells are maintained but parietal cells are lost. The the three hormones might help to regulate DC function,
resulting hypergastrinaemia can contribute to malig- and deregulation of hormone expression levels might
nant progression through several downstream signalling affect DC-mediated immune surveillance.
pathways that will be described in later sections. In addition, H. pylori has been shown to activate the
gastrin promoter through a mechanism involving the type
Infection with H. pylori IV secretory system (T4SS) (REF. 58) that, following
Explosive exocytosis
The rapid release of the Helicobacter pylori (BOX 1) is an important risk factor adherence of H. pylori, translocates the pathogenicity
contents of cellular vesicles at for cancer of the distal stomach 48. Infection with factor protein CagA into epithelial cells where it under-
the cell surface. H. pylori downregulates SHH expression, resulting in goes tyrosine phosphorylation and affects the phospho-
the loss of morphogenic differentiation, the disruption rylation of host proteins2,59–62 In a human gastric cancer
Corpus atrophy
Loss of glandular structure
of glandular structure and the gain of a more intestinal cell line, MKN45, gastrin and CCK2 receptor expression
within the corpus mucosa phenotype by upregulation of intestine-related genes, were increased following incubation with a pathogenic
which is possibly accompanied such as CDX2, MUC2 and villin47. The development of form of H. pylori to a greater extent than with a less
by fibrosis, thinning of the pre-malignant lesions such as intestinal metaplasia pre- pathogenic strain63,64. The synergy of gastrin expression
lamina propria and
dispose to gastric cancer. and Helicobacter infection in the progression of gastric
replacement of gastric
epithelium by intestinal During H. pylori-induced inflammation, cytokines adenocarcinoma was further demonstrated by the infec-
metaplastic epithelium. such as IL8, IL1β and TNFα have been implicated in tion of InsGas transgenic mice with Helicobacter felis,
increasing gastrin production from G cells49,50. Gastrin which decreased the development time of the malignant
Helicobacter felis and IL1β have been shown to synergistically increase phenotype from 20 to 9 months46. Alongside inflamma-
A strain of Helicobacter that
was originally isolated from a
the expression of the heparin-binding EGF-like growth tory changes, mouse gastrin levels were increased46,
cat and is also able to colonize factor (HB-EGF) and amphiregulin from parietal cells51. owing in part to the loss of parietal cells and the
mice and dogs. This effect might be important in modifying the prolif- acid-secretory activity of the gastric mucosa.

NATURE REVIEWS | CANCER VOLUME 6 | DECEMBER 2006 | 939


© 2006 Nature Publishing Group
REVIEWS

In human conditions with sustained tenfold elevations


Box 1 | Helicobacter pylori
in hypergastrinaemia, such as Zollinger–Ellison syn-
Helicobacter pylori is a spiral shaped, Gram-negative bacteria that colonizes the mucous drome or pernicious anaemia, ECL cell hyperplasia
pit layer of the gastric mucosa. It is acquired during childhood and persists throughout might develop but rarely leads to carcinoids. There have
life if not treated with antibiotics. Infection is thought to be associated with cancer been various explanations for these differing phenomena
development, as it slowly induces gastric atrophy, which is known to be a precursor
including increased ECL cell numbers in the rat76,77 and
lesion to intestinal metaplasia.
Gastric atrophy in response to infection with H. pylori develops slowly over a long
a net increase in glycine-extended gastrin levels in the
period of 20 to 40 years158. In 1993, The World Health Organization (WHO) and the mouse when compared with rats and humans 78–81;
International Agency for Research on Cancer (IARC) performed an extensive literature the significance of the latter being that glycine-extended
review and concluded that there is a clear gastrin might help to prevent the loss of parietal cells82.
association between H. pylori and the However, the key issue is the role of hypergastrinaemia
development of gastric cancer. Therefore, that is secondary to H. pylori infection, particularly in
H. pylori was designated as a group 1 infected patients that are treated with PPIs.
carcinogen. Helicobacter pylori infects 40% In patients with H. pylori infection who are treated
of the population in developed countries with PPIs there seems to be a shift in the location of
by the age of 50, with a much higher
associated gastritis from the antrum to the corpus83,84,
prevalence in developing countries.
However, only ~0.4% of the infected
which is significant in that the development of corpus-
population develop gastric cancer. predominant gastritis is associated with an increased
risk for gastric adenocarcinoma85. This has led to the
Image courtesy of David Kelly, University of Sheffield. recommendation by some clinicians that H. pylori
should be eradicated before treatment with PPIs86.
Infection with H. pylori in Mongolian gerbils is asso- Intriguingly, although patients with pernicious anae-
ciated with the loss of somatostatin receptor expression mia rarely develop carcinoma, those with pre-existing
and reduced somatostatin levels in the gastric mucosa65. corpus-associated gastric atrophy or gastritis have an
Furthermore, lipopolysaccharide from H. pylori inhibits increased risk of developing gastric cancer87–89. Studies
the binding of somatostatin to its receptor in the gastric of patients with Zollinger–Ellison are also difficult to
mucosa66. The receptor subtype has not been described, interpret as H. pylori infection in this patient group has
but if SSTR2A was involved, this might potentially been found to be relatively low, as is the development
reverse the inhibition mediated by SSTR2B on parietal of gastric atrophy90–94.
cells. Furthermore, as SSTR2A has been shown to induce Helicobacter pylori might increase serum gastrin
apoptosis67, inhibition might have profound effects on two to threefold, but this does not always lead to malig-
the architecture and cellular composition of the gastric nancy as it is dependent on a variety of factors, including
unit. Additional factors released following infection expression of pathogenicity elements, such as CagA, and
with H. pylori, such as platelet-activating factor, increase associated inflammatory events. Therefore, in humans,
GRP-induced gastrin release68, although the mechanism gastrin might not lead to cancer development in its own
for this activity is not clearly delineated. right, but we speculate that it is an important cofactor
Recent data have shown that acute Helicobacter once pre-malignant changes are triggered. However, the
infection resulted in an influx of bone-marrow-derived contribution made by hypergastrinaemia to the loca-
stem cells (BMDCs) in C57/Bl mice69. By 30 weeks, tion shift of atrophy in patients infected with H. pylori
entire glands were repopulated by BMDCs, which, and to the clinical outcomes from these pre-malignant
after 12 months, gave rise to intraepithelial dysplasia phenotypes is currently unknown71.
and invasive neoplastic glands. This finding indicates a
new mechanism for the development of pre-malignant Roles of the hormones in pre-malignant changes
gastric lesions and has implications for potential che- Infection with H. pylori is associated with 40% of gastric
moprophylactic modalities. The role of gastrin in this atrophy cases and results in a reduction of G cells and
process is unknown, but as bacterial infection might extension of antral type glands (pseudo-pyloric gland
have created a hypergastrinaemic environment, it would metaplasia) into the corpus95. However, pseudo-pyloric
be interesting to evaluate whether CCK2 receptors are glandular ducts that arise within the fundic area are still
expressed in the BMDCs that repopulated the gastric associated with gastrin-expressing G cells (1.03% of total
glands, as seen in the proliferative gastric component cell types within each duct) in 31.63% of cases, leading
of a hypergastrinaemic mouse70. to maintained expression of gastrin96. A larger subset of
individuals infected with H. pylori have corpus mucosal
Hypergastrinaemia and gastric carcinogenesis atrophy with loss of parietal cells97. In corpus atrophy,
This is a controversial area with many confounding fac- G cells within the pyloric glands of the antrum remain,
tors to address, making interpretation of the literature and therefore gastrin is overexpressed in response to pari-
Pseudo-pyloric gland difficult. However, recent reviews have helped to clarify etal cell loss in either intestinal metaplasia or in a mixed
metaplasia certain conflicting issues71,72. gastric and intestinal metaplastic phenotype96.
Gastric metaplasia in which Hypergastrinaemia in the InsGas mouse results in CCK2 receptor expression has been shown to be
gastric glands disappear and
are replaced by tubules that
gastric adenocarcinoma and is not accompanied by ECL maintained in early pre-malignant changes within
closely resemble normal cell hyperplasia46, whereas in the rat hypergastrinaemia the gastric mucosa. For example, in atrophic gastritis,
pyloric glands. results in the development of ECL cell carcinoids73–75. despite the loss of parietal and ECL cells, CCK2 receptors

940 | DECEMBER 2006 | VOLUME 6 www.nature.com/reviews/cancer


© 2006 Nature Publishing Group
REVIEWS

are still expressed at low levels (around 4% of the cell gene transcription through enhancing SP1 levels within
population)98. CCK2 receptor expression increased to the cell102 or by increasing the phosphorylation of SP1,
8.5% of the cell population in intestinal metaplasia. In thereby maximizing the affinity of SP1 for the gastrin
dysplasic gastric mucosa there was the greatest change promoter105. A protein–protein interaction between
in CCK2 receptor expression between moderate dys- SP1 and JUN further mediates EGF activation of the
plasia (12%) and severe dysplasia (29%) with 33.5% gastrin gene and might explain the inhibitory effect of
of adenocarcinoma cells expressing the receptor98. somatostatin on this process as somatostatin inhibits
Expression levels of the CCK2 receptor were not related JUN transcriptional activation 106. Furthermore, a
to serum gastrin levels, but it is possible that increased helix–loop–helix cis-regulatory element on the gas-
receptor occupancy could have been responsible trin promoter mediates somatostatin’s inhibition of
for the upregulation. Expression did not seem to be EGF-stimulated gastrin gene transcription107.
restricted to classical CCK2 receptor-expressing cells, Further transacting factors that interact with the
although co-staining would be needed to confirm this98. gastrin promoter and might affect gastric carcinogen-
Interestingly in the InsGas mouse model, work within esis include the zinc-finger transcription factor, ZBP89
our laboratory has confirmed a sequential upregulation (REF. 108), which represses the induction of gastrin gene
of CCK2 receptors in samples of increasing malignant expression109. ZBP89 might link gastrin gene transcrip-
potential (M.E.-Z., unpublished observations). tion to TP53 mutations as ZBP89 also binds to and sta-
In a study on human samples, D-cell numbers were bilizes p53 (REFS 110,111). Gastrin transcription is also
reduced, potentially reducing somatostatin levels, linked to the β-catenin pathway by a binding site for the
within mixed gastric and intestinal metaplasia of the transcription factor TCF4 in the gastrin promoter112.
fundus and the antrum. D cells are almost absent in full Expression of somatostatin in non-endocrine cells of
intestinal metaplasia but they are retained in pseudo- gastric adenocarcinomas seems undefined, with just one
pyloric gland metaplasia within the fundus96. A recent report describing mRNA expression in poorly differenti-
study examining the gastric phenotype in a somatosta- ated gastric carcinomas113. Somatostatin expression lev-
tin-deficient transgenic mouse model suggested that els in endocrine cells within antral and fundic atrophy,
parietal cell numbers were increased47. This is interest- however, seem to follow the same pattern as gastrin96,
ing based on the recent observation that in the InsGas and D-cell production of somatostatin might still inter-
mouse model corpus atrophy is due to gastrin-induced act with cell types within gastric adenocarcinoma if they
apoptosis of parietal cells31. Furthermore, in the InsGas express the relevant SSTR isoforms.
model, a histamine receptor H2 antagonist reversed GRP does not seem to be expressed in gastric adeno-
parietal cell loss and atrophy development, leading carcinomas. However, GRP expressed by normal cells
the authors to suggest that gastrin-induced histamine within the gastric mucosa might affect gastrin produc-
overexpression might have contributed to parietal cell tion by the epithelial cells of gastric adenocarcinoma
apoptosis. Somatostatin expression was not examined through appropriate receptor isoform expression by the
in this model but if the findings in the somatostatin cancer cells114.
knockout mice were extrapolated to these observations, CCK2 receptor deregulation in gastric adeno-
it might be speculated that overexpression of somato- carcinoma has been reported in several studies with
statin in response to hypergastrinaemia might have also contrasting results. Some data indicate that receptor
played a part. expression might correlate with differentiation sta-
tus and is higher in intestinal compared with diffuse
Deregulation of hormone expression type gastric cancer101. However, the data in different
The gastrin gene is expressed de novo in non-endocrine studies are highly variable, probably owing to low
epithelial cells within gastric adenocarcinoma, unlike levels of receptor expression and to the use of different
in normal mucosa in which the expression is restricted methodologies.
to G cells, and tumour-associated gastrin peptides Somatostatin receptors are weakly expressed in
can participate in autocrine–paracrine pathways99. gastric adenocarcinoma. For example, SSTR3 showed
Furthermore, the gastrin gene has been confirmed as reduced expression both in gastric adenocarcinoma,
a target of β-catenin-mediated transcription100. Placing compared with normal mucosa, and in poorly differen-
these findings in a clinical context, co-expression tiated cancers, compared with more well-differentiated
of gastrin and the CCK2 receptor is prognostic in terms of cancers. Interestingly, when SSTR3 was expressed in a
survival in diffuse types of gastric adenocarcinoma101. gastric cancer cell line, administration of the somatosta-
EGF receptor (EGFR) ligands induce gastrin gene tin analogue octreotide induced apoptosis115, indicating
transcription in a human gastric cancer cell line, that downregulation of the receptor in tumours could
through an EGF response element (gERE) that has contribute to the anti-apoptotic phenotype. In the rat,
been mapped to the gastrin promoter102. In the gastric SSTR2A expressed on ECL cells was anti-proliferative
mucosa, sources of EGF family ligands include the whereas SSTR2B was pro-proliferative. We speculate that
parietal cell46,103,and the related ligand, transforming SSTR2B might be preferentially expressed in tumours
growth factor-α (TGFα), is secreted by inflammatory because these receptors can induce pro-proliferative
cells, such as macrophages104. The SP1 and SP3 tran- signalling pathways.
scription factors are the predominant factors that bind GRP receptors have been found to be expressed
to the gERE element, and EGF might increase gastrin on around 50% of gastric adenocarcinomas 116. In a

NATURE REVIEWS | CANCER VOLUME 6 | DECEMBER 2006 | 941


© 2006 Nature Publishing Group
REVIEWS

study that specifically evaluated non-antral gastric Proliferation. Gastrin increases proliferation of gastric
adenocarcinomas, 8 out of 20 cancers expressed the GRP mucosal cells through transcriptional activation of a
receptor117. Some expressed mutated forms of the receptor, series of genes. One well-characterized pro-proliferative
which were either non-functional or showed constitu- gene, the expression of which is induced by gastrin stim-
tive activity. However, functional receptor expression ulation of ECL cells as previously described, is REG1α,
was not related to prognosis, which might be due to which affects the proliferation status of the stem-cell
the low number of cases investigated and unknown niche and is also overexpressed in gastric cancer127.
co-expression of other factors. It is possible that co- Reg1 gene expression is upregulated through
expression of gastrin might also be necessary for a H. pylori-induced increases in serum gastrin levels in the
prognostic relationship to be attained. Mongolian gerbil model of gastric neoplasia128.
Gastrin also promotes proliferation by activating the
Hormone functions in gastric cancer transcription of cyclin D1 in gastric cancer cells through
Gastrin, GRP and somatostatin can influence carcino- a mechanism involving both β-catenin and the tran-
genesis through several pathways (FIG. 2). The origin scription factor CREB pathways129, which correlates with
of gastrin for these effects might be either serum- the finding that gastrin, as well as being a downstream
associated endocrine gastrin or autocrine gastrin that is target of β-catenin, also stabilizes the expression of
endogenously produced by tumour cells. β-catenin and activates β-catenin-dependent transcrip-
tion130. Interestingly, infection with H. pylori, possibly
Apoptosis. Gastrin induces the expression and function through CagA, also increases the nuclear expression of
of anti-apoptotic proteins, which seems to be poten- β-catenin131. In colorectal cancer, gastrin increases the
tiated by GRP, whereas somatostatin induces pro- expression of cyclins D1, E, A and B1, whereas soma-
apoptotic proteins. An initial report confirmed that tostatin expression is inversely related to the expression
gastrin inhibits apoptosis of a rat pancreatic tumour of these cell-cycle proteins132, which further confirms
cell line, AR42J, by activating PKB/AKT42 through the opposing activities of these hormones, although
a phosphatidylinositol 3-kinase (PI3K)-dependent, somatostatin activities, as always, are dependent on
mitogen-activated protein kinase (MAPK)-independent appropriate receptor expression. The GRP homologue,
pathway. AKT activation promotes survival by phos- bombesin, has been shown to have a direct effect on
phorylating the pro-apoptotic protein BAD and cyclin D1 expression through the upregulation of the
inhibiting the transcription of forkhead transcription early growth response protein 1 (EGR1) (REF. 133) and
factors118. Furthermore, gastrin mediates the activa- so it could interact in an additive manner with gastrin,
tion of AKT through degradation of IκBα in a protein increasing transit through the cell cycle.
kinase C (PKC)-dependent fashion in human gastric Gastrin increases the expression of the EGFR ligands
cancer and oesophageal cell lines119,120. Gastrin has amphiregulin and EGF, and is linked to increased TGFα
also been linked to an increase in expression of the expression134,135. The activation of EGFR has an impor-
anti-apoptotic protein X-linked inhibitor of apoptosis tant role in increasing cell proliferation, angiogenesis and
(XIAP) in a gastro-oesophageal cell line121. metastasis, and reducing apoptosis24, and the presence of
In the large intestine there is reciprocal activity of EGFR ligands in human gastric cancer correlates with
somatostatin and gastrin on pro- and anti-apoptotic the degree of invasion and lymph node metastasis and a
proteins, respectively, with high gastrin and low worse 5-year survival rate136. Many of these growth fac-
somatostatin expression being related to increased tors remain tethered to the cell membrane in an inactive
expression of the anti-apoptotic protein BCL2 and, form and, interestingly, GRP receptor activation results
conversely, high somatostatin and low gastrin expres- in EGF ligand release137, which might act synergistically
sion being related to increased expression of the with gastrin to generate active EGF ligands and maxi-
pro-apoptotic protein BAX122. Somatostatin receptor mize EGF ligand-mediated gastrin gene transcription
isoforms SSTR1, SSTR4 and SSTR5 are associated through the gERE.
with cell-cycle arrest, whereas SSTR3 and SSTR2 seem
mostly linked to triggering apoptosis, although this Angiogenesis. Gastrin induces tubule formation of the
is dependent on the cell type involved123. Activation human endothelial cell line HUVEC through the tran-
of SSTR3 causes a conformational change in p53 and scriptional activation of HB-EGF138. The clinical sig-
induces BAX expression, both of which result in the nificance of this effect was determined by showing that
onset of apoptosis, whereas SSTR2-induced apoptosis in GI cancer patients with hypergastrinaemia, micro-
is p53-independent124. Interestingly, in a pituitary cell vessel density was increased at the tumour margin138.
line somatostatin has been shown to induce the expres- Furthermore, the angiogenic activity of gastrin might be
sion of the tumour suppressor ZAC1, which induces indirectly mediated through its ability to transcription-
cell-cycle arrest and apoptosis, possibly through ally upregulate cyclooxygenase 2 (COX2) (REFS 139–141),
dephosphorylation of PKB/AKT125, however, it is not particularly as COX2 expression in gastric cancer speci-
clear whether this mechanism is relevant in gastric mens correlates with microvessel density and is related to
tumours. GRP might further potentiate gastrin’s effect tumour invasion and lymph node metastasis142.
on PKB/AKT as it can downregulate the expression of SSTR2 has been shown to be upregulated during the
the tumour suppressor protein PTEN, which blocks angiogenic switch from resting to proliferating endothe-
PKB/AKT signalling126. lium143, whereas SSTR3 has been proposed to show

942 | DECEMBER 2006 | VOLUME 6 www.nature.com/reviews/cancer


© 2006 Nature Publishing Group
REVIEWS

Somatostatin pathway, which involved the phosphorylation of focal


Gastrin
GRP GRP
adhesion kinase (FAK)148. Gastrin also induced branch-
ing morphogenesis in a human gastric cancer cell line
Serum Autocrine that overexpressed the CCK2 receptor149. In terms of
hypergastrinaemia gastrin cell invasion, a human gastric epithelial cell line was
CCK2R shown to express matrix metalloproteinase 9 (MMP9) in
Helicobacter pylori Signalling pathways
Proton pump inhibitors • PKB/AKT
response to stimulation with gastrin through a PKC–Raf
• ERK/MAPK pathway 150, which increased invasion through an artifi-
• PKC cial basement membrane. This finding might be clini-
Somatostatin Somatostatin cally relevant as patients with serum hypergastrinaemia
GRP have increased MMP9 in their stomach150.

Hormone-targeted therapies
Cell proliferation Promotion of
• Cyclins angiogenesis Several agents targeting gastrin have been evaluated
• HB-EGF clinically, including CCK2 receptor antagonists and a
Circumvention • COX 2
Increased invasive of apoptosis gastrin vaccine. The CCK2 receptor antagonist gastra-
GRP enzyme expression • PKB/AKT zole (JB5008) was evaluated in a randomized, blind trial
Cell migration/
• MMPs • NF-κB Somatostatin in patients with advanced pancreatic cancer. Gastrazole
adhesion
• Integrins significantly increased median survival when compared
Somatostatin • FAK GRP
with a placebo (7.9 versus 4.5 months median survival,
P = 0.02) with a 1-year survival of 33% versus 11%
Figure 2 | Gastrin-mediated carcinogenic pathways. Gastrin is a diverse (P = 0.02) (REF. 151). In a second trial, no significant dif-
transcriptional activator, mediating gene expression that is associated with cell division, ference was observed when gastrazole was compared with
invasion, angiogenesis and anti-apoptotic activity, which are all pivotal in the gain of 5-fluorouracil (median survival 3.6 versus 4.2 months)151.
malignant potential. Furthermore, the source of gastrin can be either endocrine in nature No toxicity was shown for gastrazole, so it could
through gastric mucosal secretion, or of tumour origin through de novo activation of the potentially be used in combination with cytotoxic drugs.
gastrin gene following early mutational events (for example, mutation of the
A gastrin vaccine, G17DT, has been evaluated in
adenomatosis polyposis coli (APC) gene in GI carcinogenesis). Importantly, gastrin
functions are regulated by the positive regulator, gastrin-releasing peptide (GRP; the
several clinical trials. The immunogen induces anti-
mammalian homologue of bombesin) and the family of related hormones, and a negative gastrin antibodies that neutralize the amidated and
regulator, somatostatin. CCK2R, CCK2 receptor; COX2, cyclooxygenase 2; ERK, glycine-extended forms of gastrin 17 (REF. 152). In a
extracellular signal-regulated kinase; FAK, focal adhesion kinase; HB-EGF, heparin- Phase II study in patients with gastric cancer, G17DT
binding EGF-like growth factor; MAPK, mitogen-activated protein kinase; MMPs, matrix was well tolerated and antibodies were functional in that
metalloproteinases; NF-κB, nuclear factor κB; PKB, protein kinase B. they prevented gastrin from binding to CCK2 receptors
in a competitive ex vivo ligand binding assay 153. In a
larger, open-label study in advanced gastric and gastro-
anti-angiogenic activity144. Interestingly, SSTR2 has been oesophageal cancer, G17DT vaccination combined with
detected in the peri-tumoral vessels of gastric tumours145. 5-fluorouracil and leucovorin correlated with longer
The function of such receptors might be associated with time to progression and increased median survival154.
vasoconstriction, resulting in tumour hypoxia and In addition, anti-gastrin response was an independent
necrosis145. Furthermore, in hypoxic conditions, gastrin prognostic factor for overall survival154.
might upregulate the expression of the transcription The GRP receptor antagonist RC-3095 was used in a
factor hypoxia-inducible factor-1α (HIF1α), a phenom- 25-patient Phase I trial in advanced solid malignancies,
enon recently shown in our laboratory (E. Royal, A.M.G. including gastric cancer155. There was no objective tumour
and S.A.W., unpublished observations). The gastrin- response as the dose escalation procedure could not be
mediated upregulation of HIF1α might increase undertaken owing to local toxicity. It was therefore proposed
angiogenesis partially by inducing vascular endothelial that new formulations would be required to progress to
growth factor (VEGF), and perhaps by counteracting Phase II, and improved formulations with more favourable
somatostatin vasoconstriction. pharmacokinetics are currently being investigated.
The GRP homologue bombesin has been shown to One trial of 107 patients with advanced GI cancer,
increase VEGF transcription through an NF-κB-mediated including 15 patients with advanced gastric cancer, has
pathway in prostate cancer cells146. However, this finding been performed with the somatostatin analogue
has not been confirmed in gastric cancer. octreotide. Octreotide extended survival in the treated
patients (median of 20 versus 11 weeks, P < 0.0001).
Motility and invasion. Gastrin can modify the cell–cell adhe- Although this result was encouraging, this therapeutic
sion molecules p120, α- and β-catenins and E-cadherin, approach has not been pursued for gastric adenocar-
resulting in loss of adhesion and increased cell motility cinomas, perhaps owing to low somatostatin receptor
in intestinal epithelial-cell-culture systems147. A glycine- expression in this cancer type115,156.
extended version of gastrin, known to mediate autocrine Based on the SSTR and GRP expression levels in
growth pathways in GI cancer cells99, was shown to gastric cancer and their interrelationship with gastrin and
stimulate the migration of a mouse gastric epithelial cell CCK2 receptor expression, combined therapies might be
line through a Rho and ROCK (Rho kinase)-dependent synergistic. GRP antagonists could be used to prevent

NATURE REVIEWS | CANCER VOLUME 6 | DECEMBER 2006 | 943


© 2006 Nature Publishing Group
REVIEWS

constitutive secretion of precursor forms of gastrin from Conclusion


malignant epithelial cells, which have biological activity but The interwoven network between gastrin, somatostatin
no defined receptor157. This approach might complement and GRP, which exists in the normal gastric environment,
G17DT therapy, which blocks amidated and glycine- has been exploited in gastric carcinogenesis and data pre-
extended gastrin, and the use of CCK2 receptor antagonists, sented in this Review reinforces the complexity and vari-
which are likely to be more efficacious against serum- ability in pathological outcome of pre-malignant gastric
associated amidated gastrin forms. Additionally, if gastrin lesions arising within different mucosal areas within the
suppresses SSTR expression in tumour cells, these receptors stomach. This in itself is a simplistic scenario consider-
might be re-expressed when gastrin expression is reduced. ing the vast array of additional GI hormones that have an
This indicates that the efficacy of a somatostatin analogue effect on these pathways. However, it has highlighted areas
could be increased if used as an adjunct to anti-gastrin treat- ripe for future investigation, and further analysis of the
ment. Also, following eradication of H. pylori, SSTR might inter-relationships of these hormones in gastric tumours
reappear on pre-malignant lesions, which could be targeted will enable investigators to define the optimal combination
with ligands such as octreotide. of clinical candidates to target this hormone trio.

1. Wiborg, O. et al. Structure of a human gastrin gene. 20. Park, J., Chiba, T. & Yamada, T. Mechanisms for This study confirmed that the hedgehog pathway is
Proc. Natl Acad. Sci. USA 81, 1067–1069 (1984). direct inhibition of canine gastric parietal cells by essential for tumour growth through the induction
2. Dickinson, C. J., Sawada, M., Guo, Y. J., Finniss, S. & somatostatin. J. Biol. Chem. 262, 14190–14196 of ligand expression and that this proliferation
Yamada, T. Specificity of prohormone convertase (1987). could be suppressed by the hedgehog pathway
endoproteolysis of progastrin in AtT-20 cells. J. Clin. 21. Prinz, C., Sachs, G., Walsh, J. H., Coy, D. H. & antagonist cyclopamine.
Invest. 96, 1425–1431 (1995). Wu, S. V. The somatostatin receptor subtype on rat 38. Osawa, H. et al. Sonic hedgehog stimulates the
3. Marino, L. R., Takeuchi, T., Dickinson, C. J. & Yamada, T. enterochromaffinlike cells. Gastroenterology 107, proliferation of rat gastric mucosal cells through ERK
Expression and post-translational processing of 1067–1074 (1994). activation by elevating intracellular calcium
gastrin in heterologous endocrine cells. J. Biol. Chem. 22. Allen, J. P. et al. Identification of cells expressing concentration. Biochem. Biophys. Res. Commun. 344,
266, 6133–6136 (1991). somatostatin receptor 2 in the gastrointestinal tract of 680–687 (2006).
4. Lloyd, K. C. Gut hormones in gastric function. Baillieres Sstr2 knockout/lacZ knockin mice. J. Comp. Neurol. 39. Ma, X. et al. Frequent activation of the hedgehog
Clin. Endocrinol. Metab. 8, 111–136 (1994). 454, 329–340 (2002). pathway in advanced gastric adenocarcinomas.
This review provides a detailed overview of the 23. Reeve, J. R. Jr et al. Amino acid sequences of three Carcinogenesis 26, 1698–1705 (2005).
interactions between gastrin, somatostatin and bombesin-like peptides from canine intestine extracts. 40. Zavros, Y. et al. Gastrin-induced pepsinogen regulates
GRP on the maintenance of stomach homeostasis. J. Biol. Chem. 258, 5582–5588 (1983). sonic hedgehog processing and prevents gastric
5. Soll, A. H. & Grossman, M. I. The interaction of 24. Sugano, K., Park, J., Soll, A. H. & Yamada, T. atrophy. Gastroenterology 130, A17 (2006).
stimulants on the function of isolated canine parietal Stimulation of gastrin release by bombesin and canine 41. Todisco, A. et al. Molecular mechanisms for the
cells. Philos. Trans. R. Soc. Lond. B 296, 5–15 (1981). gastrin-releasing peptides. Studies with isolated antiapoptotic action of gastrin. Am. J. Physiol.
6. Obrink, K. J. Histamine and gastric acid secretion. canine G cells in primary culture. J. Clin. Invest. 79, Gastrointest. Liver Physiol. 280, G298–G307
A review. Scand. J. Gastroenterol. Suppl. 180, 4–8 935–942 (1987). (2001).
(1991). 25. Schubert, M. L., Hightower, J., Coy, D. H. & Makhlouf, This paper overviews the signalling pathways
7. Walsh, J. H. Physiology and pathophysiology of G. M. Regulation of acid secretion by bombesin/GRP through which gastrin can mediate anti-apoptotic
gastrin. Mt Sinai J. Med. 59, 117–124 (1992). neurons of the gastric fundus. Am. J. Physiol. 260, activity in cancer.
8. Chuang, C. N., Tanner, M., Chen, M. C., Davidson, S. & G156–G160 (1991). 42. Todisco, A. et al. Functional role of protein kinase
Soll, A. H. Gastrin induction of histamine release from 26. Hohenberger, P. & Gretschel, S. Gastric cancer. Lancet B/AKT in gastric acid secretion. J. Biol. Chem. 276,
primary cultures of canine oxyntic mucosal cells. Am. 362, 305–315 (2003). 46436–46444 (2001).
J. Physiol. 263, G460–G465 (1992). 27. La Vecchia, C., Negri, E., Franceschi, S. & Gentile, A. 43. Riobo, N. A., Lu, K., Ai, X., Haines, G. M. & Emerson,
9. Waldum, H. L. et al. The cellular localization of the Family history and the risk of stomach and colorectal C. P. Jr. Phosphoinositide 3-kinase and AKT are
cholecystokinin 2 (gastrin) receptor in the stomach. cancer. Cancer 70, 50–55 (1992). essential for sonic hedgehog signaling. Proc. Natl
Pharmacol. Toxicol. 91, 359–362 (2002). 28. Lauren, P. The two histological main types of Acad. Sci. USA 103, 4505–4510 (2006).
10. Lindstrom, E., Chen, D., Norlen, P., Andersson, K. & gastric carcinoma: diffuse and so-called intestinal- 44. Stepan, V. et al. Regulation and function of the sonic
Hakanson, R. Control of gastric acid secretion:the type carcinoma. An attempt at a histo-clinical hedgehog signal transduction pathway in isolated
gastrin–ECL cell-parietal cell axis. Comp. Biochem. classification. Acta Pathol. Microbiol. Scand. 64, gastric parietal cells. J. Biol. Chem. 280,
Physiol. 128, 505–514 (2001). 31–49 (1965). 15700–15708 (2005).
11. Komazawa, Y. et al. Tolerance to famotidine and 29. Miyaji, H. et al. Helicobacter pylori infection occurs 45. Dimmler, A. et al. Transcription of sonic hedgehog, a
ranitidine treatment after 14 days of administration in via close contact with infected individuals in early potential factor for gastric morphogenesis and gastric
healthy subjects without Helicobacter pylori infection. childhood. J. Gastroenterol. Hepatol. 15, 257–262 mucosa maintenance, is up-regulated in acidic
J. Gastroenterol. Hepatol. 18, 678–682 (2003). (2000). conditions. Lab. Invest. 83, 1829–1837 (2003).
12. Soll, A. H., Amirian, D. A., Thomas, L. P., Reedy, T. J. 30. Wang, L. D. et al. Changes in p53 and cyclin D1 46. Wang, T. C. et al. Synergistic interaction between
& Elashoff, J. D. Gastrin receptors on isolated canine protein levels and cell proliferation in different stages hypergastrinemia and Helicobacter infection in a
parietal cells. J. Clin. Invest. 73, 1434–1447 (1984). of human esophageal and gastric-cardia mouse model of gastric cancer. Gastroenterology 118,
13. Helander, H. F., Wong, H., Poorkhalkali, N. & carcinogenesis. Int. J. Cancer 59, 514–519 (1994). 36–47 (2000).
Walsh, J. H. Immunohistochemical localization of 31. Takaishi, S. et al. Synergistic inhibitory effects of This is a milestone publication highlighting that
gastrin/CCK-B receptors in the dog and guinea-pig gastrin and histamine receptor antagonists on Helicobacter infection and hypergastrinaemia
stomach. Acta Physiol. Scand. 159, 313–320 (1997). Helicobacter-induced gastric cancer. Gastroenterology interact during the development of gastric cancer.
14. Kopin, A. S. et al. Expression cloning and 128, 1965–1983 (2005). 47. Zavros, Y. et al. Chronic gastritis in the hypochlorhydric
characterization of the canine parietal cell gastrin 32. Delle Fave, G., Capurso, G., Milione, M. & Panzuto, F. gastrin-deficient mouse progresses to adenocarcinoma.
receptor. Proc. Natl Acad. Sci. USA 89, 3605–3609 Endocrine tumours of the stomach. Best Pract. Res. Oncogene 24, 2354–2366 (2005).
(1992). Clin. Gastroenterol. 19, 659–673 (2005). 48. Helicobacter & Cancer Group. Gastric cancer and
15. Cabero, J. L., Li, Z. Q. & Mardh, S. Gastrin potentiates 33. Sekikawa, A. et al. REG Iα protein may function as a Helicobacter pylori: a combined analysis of 12 case
histamine-stimulated aminopyrine accumulation in trophic and/or anti-apoptotic factor in the control studies nested within prospective cohorts.
isolated rat parietal cells. Am. J. Physiol. 261, development of gastric cancer. Gastroenterology 128, Gut 49, 347–353 (2001).
G621–G627 (1991). 642–653 (2005). 49. Beales, I. et al. Effect of Helicobacter pylori products
16. Brenna, E. & Waldum, H. L. Studies of isolated 34. Kazumori, H., Ishihara, S., Fukuda, R. & Kinoshita, Y. and recombinant cytokines on gastrin release from
parietal and enterochromaffin-like cells from the rat. Localization of REG receptor in rat fundic mucosa. J. cultured canine G cells. Gastroenterology 113,
Scand. J. Gastroenterol. 26, 1295–1306 (1991). Lab. Clin. Med. 139, 101–108 (2002). 465–471 (1997).
17. Kobayashi, T. et al. Abnormal functional and 35. Fukui, H. et al. Regenerating gene protein may This study was the first to show that H. pylori
morphological regulation of the gastric mucosa in mediate gastric mucosal proliferation induced by products might contribute indirectly to hypergastri-
histamine H2 receptor-deficient mice. J. Clin. Invest. hypergastrinemia in rats. Gastroenterology 115, naemia through the induction of cytokines that go
105, 1741–1749 (2000). 1483–1493 (1998). on to potentiate gastrin release from the G cell.
18. Karnik, P. S. & Wolfe, M. M. Somatostatin stimulates 36. Miyaoka, Y. et al. Transgenic overexpression of REG 50. Suzuki, T. et al. TNF-α and interleukin 1 activate
gastrin mRNA turnover in dog antral mucosa. J. Biol. protein caused gastric cell proliferation and gastrin gene expression via MAPK- and PKC-
Chem. 265, 2550–2555 (1990). differentiation along parietal cell and chief cell dependent mechanisms. Am. J. Physiol. Gastrointest.
19. Godley, J. M. & Brand, S. J. Regulation of the lineages. Oncogene 23, 3572–3579 (2004). Liver Physiol. 281, G1405–G1412 (2001).
gastrin promoter by epidermal growth factor and 37. Berman, D. M. et al. Widespread requirement for 51. Beales, I. L. Gastrin and interleukin-1β stimulate
neuropeptides. Proc. Natl Acad. Sci. USA 86, hedgehog ligand stimulation in growth of digestive growth factor secretion from cultured rabbit gastric
3036–3040 (1989). tract tumours. Nature 425, 846–851 (2003). parietal cells. Life Sci. 75, 2983–2995 (2004).

944 | DECEMBER 2006 | VOLUME 6 www.nature.com/reviews/cancer


© 2006 Nature Publishing Group
REVIEWS

52. Dockray, G. J., Varro, A., Dimaline, R. & Wang, T. The 74. Havu, N. Enterochromaffin-like cell carcinoids of 96. Otsuka, T. et al. Coexistence of gastric- and intestinal-
gastrins: their production and biological activities. gastric mucosa in rats after life-long inhibition type endocrine cells in gastric and intestinal mixed
Annu. Rev. Physiol. 63, 119–139 (2001). of gastric secretion. Digestion 35, 42–55 (1986). intestinal metaplasia of the human stomach. Pathol.
53. Kinoshita, Y. & Ishihara, S. Mechanism of gastric 75. Havu, N., Mattsson, H., Ekman, L. & Carlsson, E. Int. 55, 170–179 (2005).
mucosal proliferation induced by gastrin. Enterochromaffin-like cell carcinoids in the rat gastric 97. Correa, P. Chronic gastritis: a clinico-pathological
J. Gastroenterol. Hepatol. 15, D7–D11 (2000). mucosa following long-term administration of classification. Am. J. Gastroenterol. 83, 504–509
54. Zavros, Y. et al. Treatment of Helicobacter gastritis ranitidine. Digestion 45, 189–195 (1990). (1988).
with IL-4 requires somatostatin. Proc. Natl Acad. Sci. 76. Hakanson, R. et al. Effects of antrectomy or 98. Henwood, M., Clarke, P. A., Smith, A. M. & Watson, S. A.
USA 100, 12944–12949 (2003). portacaval shunting on the histamine-storing Expression of gastrin in developing gastric
55. Kao, J. Y. et al. Somatostatin inhibits dendritic cell endocrine-like cells in oxyntic mucosa of rat stomach. adenocarcinoma. Br. J. Surg. 88, 564–568 (2001).
responsiveness to Helicobacter pylori. Regul. Pept. A fluorescence histochemical, electron microscopic 99. Watson, S. A., Morris, T. M., Varro, A., Michaeli, D. &
134, 23–29 (2006). and chemical study. J. Physiol. 259, 785–800 Smith, A. M. A comparison of the therapeutic
56. Takhar, A., Eremin, O. & Watson, S. A. The role of (1976). effectiveness of gastrin neutralisation in two human
gastrin in dendritic cell function. Br. J. Surg. 93 77. Simonsson, M. et al. Endocrine cells in the human gastric cancer models: relation to endocrine and
(Suppl. 3), A15 (2006). oxyntic mucosa. A histochemical study. Scand. J. autocrine/paracrine gastrin mediated growth. Gut 45,
57. Makarenkova, V. P. et al. Lung cancer-derived Gastroenterol. 23, 1089–1099 (1988). 812–817 (1999).
bombesin-like peptides down-regulate the 78. Varro, A. et al. Processing of the gastrin precursor. 100. Koh, T. J. & Chen, D. Gastrin as a growth factor in the
generation and function of human dendritic cells. Modulation of phosphorylated, sulfated, and gastrointestinal tract. Regul. Pept. 93, 37–44
J. Neuroimmunol. 145, 55–67 (2003). amidated products. J. Biol. Chem. 265, (2000).
58. Rieder, G., Merchant, J. L. & Haas, R. Helicobacter 21476–21481 (1990). 101. Hur, K. et al. Expression of gastrin and its receptor in
pylori cag-type IV secretion system facilitates corpus 79. Azuma, T., Taggart, R. T. & Walsh, J. H. Effects of human gastric cancer tissues. J. Cancer Res. Clin.
colonization to induce precancerous conditions bombesin on the release of glycine-extended Oncol. 132, 85–91 (2006).
in Mongolian gerbils. Gastroenterology 128, progastrin (gastrin G) in rat antral tissue culture. This is the most comprehensive study to date
1229–12242 (2005). Gastroenterology 93, 322–329 (1987). examining co-expression of gastrin and the CCK2
This is a milestone paper defining a potential 80. Huebner, V. D. et al. Purification and structural receptor on human gastric cancer tissues, including
mechanism through which H. pylori can directly characterization of progastrin-derived peptides from evaluation of their relationship to grade, sex and
activate gastrin gene transcription by the G cell a human gastrinoma. J. Biol. Chem. 266, prognosis.
and also might indicate a way through which non- 12223–12227 (1991). 102. Merchant, J. L., Du, M. & Todisco, A. SP1
endocrine malignant cells could gain an activated 81. Azuma, T. et al. Carboxyl terminal glycine extended phosphorylation by ERK 2 stimulates DNA binding.
gastrin gene. progastrin (gastrin-G) in gastric antral mucosa of Biochem. Biophys. Res. Commun. 254, 454–461
59. Walter, P., Gilmore, R. & Blobel, G. Protein patients with gastric or duodenal ulcer and in (1999).
translocation across the endoplasmic reticulum. gastrinomas. J. Gastroenterol. Hepatol. 5, 525–529 103. Murayama, Y. et al. Localization of heparin-binding
Cell 38, 5–8 (1984). (1990). epidermal growth factor-like growth factor in human
60. Burgess, T. L. & Kelly, R. B. Constitutive and regulated 82. Chen, D. et al. Glycine-extended gastrin synergizes gastric mucosa. Gastroenterology 109, 1051–1059
secretion of proteins. Annu. Rev. Cell Biol. 3, 243–293 with gastrin-17 to stimulate acid secretion in gastrin (1995).
(1987). deficient mice. Gastroenterology 116, 756–765 104. Madtes, D. K., Malden, L. T., Raines, E. W. & Ross, R.
61. Calam, J., Dockray, G. J., Walker, R., Tracy, H. J. & (1999). Induction of transcription and secretion of TGF-α
Owens, D. Molecular forms of gastrin in peptic ulcer: 83. Kuipers, E. J. et al. Cure of Helicobacter pylori by activated human monocytes. Chest 99, S79
comparison of serum and tissue concentrations of infection in patients with reflux oesophagitis (1991).
G17 and G34 in gastric and duodenal ulcer subjects. treated with long term omeprazole reverses 105. Mortensen, E. R., Marks, P. A., Shiotani, A. &
Eur. J. Clin. Invest. 10, 241–247 (1980). gastritis without exacerbation of reflux disease: Merchant, J. L. Epidermal growth factor and okadaic
62. Dickinson, C. J., Daugherty, D., Guo, Y. J., Hughes, P. results of a randomised controlled trial. Gut 53, acid stimulate SP1 proteolysis. J. Biol. Chem. 272,
& Yamada, T. Molecular analysis of dibasic 12–20 (2004). 16540–16547 (1997).
endoproteolytic cleavage signals. J. Biol. Chem. 267, 84. Eissele, R., Brunner, G., Simon, B., Solcia, E. & Arnold, R. 106. Todisco, A., Seva, C., Takeuchi, Y., Dickinson, C. J. &
21795–21801 (1992). Gastric mucosa during treatment with lansoprazole: Yamada, T. Somatostatin inhibits AP-1 function via
63. Dickson, J., Grabowska, A., Atherton, J. & Watson, S. A. Helicobacter pylori is a risk factor for argyrophil cell multiple protein phosphatases. Am. J. Physiol. 269,
Role of gastrin in Helicobacter pylori induced HB-EGF hyperplasia. Gastroenterology 112, 707–717 G160–G166 (1995).
gene up-regulation and ectodomain shedding. (1997). 107. Bachwich, D., Merchant, J. & Brand, S. J. Identification
Gastroenterology 126, A59 (2004). 85. Uemura, N. et al. Helicobacter pylori infection and the of a cis-regulatory element mediating somatostatin
64. Dickson, J. H., Atherton, J. & Watson, S. A. development of gastric cancer. N. Engl. J. Med. 345, inhibition of epidermal growth factor-stimulated gastrin
Helicobacter pylori may up-regulate gastrin/CCK-2 784–789 (2001). gene transcription. Mol. Endocrinol. 6, 1175–1184
receptor expression in exposed cells and human gastric 86. Gillen, D. & McColl, K. E. Problems associated with (1992).
mucosa. Gastroenterology 128, W1341 (2005). the clinical use of proton pump inhibitors. Pharmacol. 108. Merchant, J. L. et al. ZBP-89, a Kruppel-like zinc
65. Brzozowski, T. et al. Effect of probiotics and triple Toxicol. 89, 281–286 (2001). finger protein, inhibits epidermal growth factor
eradication therapy on the cyclooxygenase (COX)-2 87. Borch, K., Renvall, H., Liedberg, G. & Andersen, B. N. induction of the gastrin promoter. Mol. Cell. Biol. 16,
expression, apoptosis, and functional gastric mucosal Relations between circulating gastrin and endocrine 6644–6653 (1996).
impairment in Helicobacter pylori-infected Mongolian cell proliferation in the atrophic gastric fundic mucosa. 109. Law, G. L. et al. Transcription factor ZBP-89 regulates
gerbils. Helicobacter 11, 10–20 (2006). Scand. J. Gastroenterol. 21, 357–363 (1986). the activity of the ornithine decarboxylase promoter.
66. Piotrowski, J., Skrodzka, D., Slomiany, A. & Slomiany, 88. Solcia, E., Fiocca, R., Villani, L., Luinetti, O. & J. Biol. Chem. 273, 19955–19964 (1998).
B. L. Reversal of gastric somatostatin receptor Capella, C. Hyperplastic, dysplastic, and neoplastic 110. Merchant, J. L., Tucker, T. P. & Zavros, Y. in Gastrin in
inhibition by Helicobacter pylori lipopolysaccharide enterochromaffin-like-cell proliferations of the the New Millennium (eds Merchant, J. L., Buchan, A.
with ebrotidine and sulglycotide. Gen. Pharmacol. 28, gastric mucosa. Classification and histogenesis. M. J. & Wang, T. C.) 55–70 (CURE Foundation,
705–708 (1997). Am. J. Surg. Pathol. 19, S1–S7 (1995). 2004).
67. Guillermet, J. et al. Somatostatin receptor subtype 2 89. Delle Fave, G. et al. Hypergastrinemia and 111. Bai, L. & Merchant, J. L. ZBP-89 promotes growth
sensitizes human pancreatic cancer cells to death enterochromaffin-like cell hyperplasia. Yale J. Biol. arrest through stabilization of p53. Mol. Cell. Biol. 21,
ligand-induced apoptosis. Proc. Natl Acad. Sci. USA Med. 71, 291–301 (1998). 4670–4683 (2001).
100, 155–160 (2003). 90. Fich, A., Talley, N. J., Shorter, R. G. & Phillips, S. F. 112. Koh, T. J. et al. Gastrin is a target of the β-catenin/TCF-4
68. Beales, I. L. Effect of platelet-activating factor on Zollinger–Ellison syndrome. Relation to Helicobacter growth-signaling pathway in a model of intestinal
gastrin release from cultured rabbit G-cells. Dig. Dis. pylori-associated chronic gastritis and gastric acid polyposis. J. Clin. Invest. 106, 533–539 (2000).
Sci. 46, 301–306 (2001). secretion. Dig. Dis. Sci. 36, 10–14 (1991). 113. Zhang, Q. X., Dou, Y. L., Shi, X. Y. & Ding, Y. Expression
69. Li, H. C., Stoicov, C., Rogers, A. B. & Houghton, J. 91. Saeed, Z. A. et al. Helicobacter pylori and Zollinger– of somatostatin mRNA in various differentiated types of
Stem cells and cancer: evidence for bone marrow stem Ellison syndrome. Dig. Dis. Sci. 36, 15–18 (1991). gastric carcinoma. World J. Gastroenterol. 4, 48–51
cells in epithelial cancers. World J. Gastroenterol. 12, 92. Weber, H. C., Venzon, D. J., Jensen, R. T. & Metz, D. C. (1998).
363–371 (2006). Studies on the interrelation between Zollinger–Ellison 114. Patel, O., Shulkes, A. & Baldwin, G. S. Gastrin-
70. Kanda, N. et al. Involvement of cyclooxygenase-2 in syndrome, Helicobacter pylori, and proton pump releasing peptide and cancer. Biochim. Biophys. Acta
gastric mucosal hypertrophy in gastrin transgenic inhibitor therapy. Gastroenterology 112, 84–91 1766, 23–41 (2006).
mice. Am. J. Physiol. Gastrointest. Liver Physiol. 290, (1997). 115. Hu, C. et al. The effect of somatostatin and SSTR3 on
G519–G527 (2006). 93. Hirschowitz, B. I. & Haber, M. M. Helicobacter pylori proliferation and apoptosis of gastric cancer cells.
71. Jensen, R. T. Consequences of long-term proton pump effects on gastritis, gastrin and enterochromaffin-like Cancer Biol. Ther. 3, 726–730 (2004).
blockade: insights from studies of patients with gastri- cells in Zollinger–Ellison syndrome and non-Zollinger– 116. Preston, S. R., Woodhouse, L. F., Jones-Blackett, S.,
nomas. Basic Clin. Pharmacol. Toxicol. 98, 4–19 Ellison syndrome acid hypersecretors treated long- Wyatt, J. I. & Primrose, J. N. High affinity binding sites
(2006). term with lansoprazole. Aliment. Pharmacol. Ther. 15, for gastrin releasing peptide on human gastric cancer
72. Laine, L., Ahnen, D., McClain, C., Solcia, E. & 87–103 (2001). and Menetrier’s mucosa. Cancer Res. 53, 5090–5092
Walsh, J. H. Review article: potential gastrointestinal 94. Peghini, P. L. et al. Effect of chronic hypergastrinemia (1993).
effects of long-term acid suppression with proton pump on human enterochromaffin-like cells: insights from 117. Carroll, R. E., Carroll, R. & Benya, R. V. Characterization
inhibi-tors. Aliment. Pharmacol. Ther. 14, 651–668 patients with sporadic gastrinomas. Gastroenterology of gastrin-releasing peptide receptors aberrantly
(2000). 123, 68–85 (2002). expressed by non-antral gastric adenocarcinomas.
73. Mattsson, H. et al. Partial gastric corpectomy results 95. El-Zimaity, H. M., Ota, H., Graham, D. Y., Akamatsu, T. Peptides 20, 229–237 (1999).
in hypergastrinemia and development of gastric & Katsuyama, T. Patterns of gastric atrophy in intestinal This paper shows that mutated isoforms of the
enterochromaffinlike-cell carcinoids in the rat. type gastric carcinoma. Cancer 94, 1428–1436 GRP receptor are expressed by non-antral gastric
Gastroenterology 100, 311–319 (1991). (2002). adenocarcinoma.

NATURE REVIEWS | CANCER VOLUME 6 | DECEMBER 2006 | 945


© 2006 Nature Publishing Group
REVIEWS

118. Ramamoorthy, S., Stepan, V. & Todisco, A. 138. Clarke, P. A., Dickson, J. H., Harris, J. C., Grabowska, A. 158. Asaka, M., Kudo, M., Kato, M., Sugiyama, T. &
Intracellular mechanisms mediating the anti-apoptotic & Watson, S. A. Gastrin enhances the angiogenic Takeda, H. Review article: long-term Helicobacter
action of gastrin. Biochem. Biophys. Res. Commun. potential of endothelial cells via modulation of heparin pylori infection--from gastritis to gastric cancer.
323, 44–48 (2004). binding epidermal-like growth factor. Cancer Res. 66, Aliment. Pharmacol. Ther. 12, 9–15 (1998).
119. Ogasa, M. et al. Gastrin activates nuclear factor κB 3504–3512 (2006). 159. Dimaline, R. & Sandvik, A. K. Histidine decarboxylase
(NFκB) through a protein kinase C dependent pathway 139. Guo, Y. S. et al. Gastrin stimulates cyclooxygenase-2 gene expression in rat fundus is regulated by gastrin.
involving NFκB inducing kinase, inhibitor κB (IκB) expression in intestinal epithelial cells through multiple FEBS Lett. 281, 20–22 (1991).
kinase, and tumour necrosis factor receptor associated signaling pathways. Evidence for involvement of ERK5 160. Fykse, V., Coy, D. H., Waldum, H. L. & Sandvik, A. K.
factor 6 (TRAF6) in MKN-28 cells transfected with kinase and transactivation of the epidermal growth Somatostatin-receptor 2 (sst2)-mediated effects of
gastrin receptor. Gut 52, 813–819 (2003). factor receptor. J. Biol. Chem. 277, 48755–48763 endogenous somatostatin on exocrine and endocrine
120. Harris, J. C., Clarke, P. A., Awan, A., Jankowski, J. & (2002). secretion of the rat stomach. Br. J. Pharmacol. 144,
Watson, S. A. An antiapoptotic role for gastrin and the 140. Abdalla, S. I. et al. Gastrin-induced cyclooxygenase-2 416–421 (2005).
gastrin/CCK-2 receptor in Barrett’s esophagus. Cancer expression in Barrett’s carcinogenesis. Clin. Cancer 161. Bakke, I., Qvigstad, G., Sandvik, A. K. & Waldrum, H. L.
Res. 64, 1915–1919 (2004). Res. 10, 4784–4792 (2004). The CCK-2 receptor is located on the ECL cell, but not
121. Harris, J. C., Clarke, P. & Watson, S. A. Gastrin 141. Colucci, R. et al. Gastrin promotes human colon on the parietal cell. Scand. J. Gastroenterol. 11,
mediated XIAP up-regulation and associated cisplatin cancer cell growth via CCK-2 receptor-mediated 1128–1133 (2001).
resistance in the pre-malignant and malignant cyclooxygenase-2 induction and prostaglandin E2 162. Nakajima, T. et al. Gastrin stimulates the growth of
oesophagus. Gastroenterology 128 (Suppl. 3), A42 production. Br. J. Pharmacol. 144, 338–348 (2005). gastric pit cell precursors by inducing its own receptors.
(2005). 142. Sun, W. H. et al. Expression of cyclooxygenase-2 and Am. J. Physiol. Gastrointest. Liver Physiol. 282,
122. Mao, J. D. et al. Correlation between expression of matrix metalloproteinase-9 in gastric carcinoma and G359–G366 (2002).
gastrin, somatostatin and cell apoptosis regulation its correlation with angiogenesis. Jpn J. Clin. Oncol. 163. Schmidt, W. E. & Schmitz, F. Cellular localization of
gene BCL-2/BAX in large intestine carcinoma. World J. 35, 707–713 (2005). cholecystokinin receptors as the molecular basis of the
Gastroenterol. 11, 721–725 (2005). 143. Woltering, E. A. Development of targeted somatostatin- periperal regulation of acid secretion. Pharmacol.
123. Ferjoux, G. et al. Signal transduction of somatostatin based antiangiogenic therapy: a review and future Toxicol. 91, 351–358 (2002).
receptors negatively controlling cell proliferation. perspectives. Cancer Biother. Radiopharm. 18, 164. Schmitz, F. et al. CCK-B/gastrin receptors in human colo-
J. Physiol. Paris 94, 205–210 (2000). 601–609 (2003). rectal cancer. Eur. J. Clin. Invest. 31, 812–820 (2001).
This study defines how somatostatin receptor 144. Florio, T. et al. Somatostatin inhibits tumor 165. Ito, M. et al. Functional characterization of two
subtypes can mediate distinct signal-transduction angiogenesis and growth via somatostatin receptor-3- cholecystokinin-B/gastrin receptor isoforms: a
pathways that inhibit proliferation, induce cell-cycle mediated regulation of endothelial nitric oxide synthase preferential splice donor site in the human receptor
arrest and promote angiogenesis. and mitogen-activated protein kinase activities. gene. Cell Growth Differ. 5, 1127–1135 (1994).
124. Sharma, K., Patel, Y. C. & Srikant, C. B. Subtype- Endocrinology 144, 1574–1584 (2003). 166. Zhou, J. et al. Human gastric tissues coexpress two
selective induction of wild-type p53 and apoptosis, 145. Reubi, J. C. Peptide receptors as molecular targets for different splicing cholecystokinin-B/gastrin receptors.
but not cell cycle arrest, by human somatostatin cancer diagnosis and therapy. Endocr. Rev. 24, Sheng Wu Yi Xue Gong Cheng Xue Za Zhi 21,
receptor 3. Mol. Endocrinol. 10, 1688–1696 389–427 (2003). 440–443 (2004).
(1996). 146. Levine, L. et al. Bombesin stimulates nuclear factor κB 167. Miyake, A. A truncated isoform of human CCK-B/
125. Theodoropoulou, M. et al. Octreotide, a somatostatin activation and expression of proangiogenic factors in gastrin receptor generated by alternative usage of a
analogue, mediates its antiproliferative action in prostate cancer cells. Cancer Res. 63, 3495–3502 novel exon. Biochem. Biophys. Res. Commun. 208,
pituitary tumor cells by altering phosphatidylinositol (2003). 230–237 (1995).
3-kinase signaling and inducing Zac1 expression. 147. Ferrand, A. et al. Involvement of JAK2 upstream of 168. Laghi, L. et al. Frameshift mutations of human gastrin
Cancer Res. 66, 1576–1582 (2006). the PI 3-kinase in cell-cell adhesion regulation by receptor gene (hGARE) in gastrointestinal cancers with
126. Qiao, J., Kang, J., Cree, J., Evers, B. M. & Chung, D. H. gastrin. Exp. Cell Res. 301, 128–138 (2004). microsatellite instability. Lab. Invest. 82, 265–271
Gastrin-releasing peptide-induced down-regulation of 148. He, H., Pannequin, J., Tantiongco, J. P., Shulkes, A. & (2002).
tumor suppressor protein PTEN (phosphatase and Baldwin, G. S. Glycine-extended gastrin stimulates cell 169. Gugger, M., Waser, B., Kappeler, A., Schonbrunn, A. &
tensin homolog deleted on chromosome ten) in proliferation and migration through a Rho- and ROCK- Reubi, J. C. Cellular detection of sst2A receptors in
neuro-blastomas. Ann. Surg. 241, 684–691; dependent pathway, not a Rac/Cdc42-dependent human gastrointestinal tissue. Gut 53, 1431–1436
discussion 691–692 (2005). pathway. Am. J. Physiol. Gastrointest. Liver Physiol. (2004).
127. Kinoshita, Y., Ishihara, S., Kadowaki, Y., Fukui, H. & 289, G478–G488 (2005). 170. van der Hoek, J., Hofland, L. J. & Lamberts, S. W.
Chiba, T. REG protein is a unique growth factor of 149. Pagliocca, A. et al. Stimulation of the gastrin- Novel subtype specific and universal somatostatin
gastric mucosal cells. J. Gastroenterol. 39, 507–513 cholecystokininB receptor promotes branching analogues: clinical potential and pitfalls. Curr. Pharm.
(2004). morphogenesis in gastric AGS cells. Am. J. Physiol. Des. 11, 1573–1592 (2005).
128. Fukui, H. et al. Effects of Helicobacter pylori infection Gastrointest. Liver Physiol. 283, G292–G299 (2002). 171. Panteris, V. & Karamanolis, D. G. The puzzle of
on the link between regenerating gene expression and 150. Wroblewski, L. E., Pritchard, D. M., Carter, S. & Varro, somatostatin: action, receptors, analogues and therapy.
serum gastrin levels in Mongolian gerbils. Lab. Invest. A. Gastrin-stimulated gastric epithelial cell invasion: Hepatogastroenterology 52, 1771–1781 (2005).
83, 1777–1786 (2003). the role and mechanism of increased matrix 172. Alderton, F., Fan, T. P., Schindler, M. & Humphrey, P. P.
129. Pradeep, A. et al. Gastrin-mediated activation of cyclin metalloproteinase 9 expression. Biochem. J. 365, Rat somatostatin sst2(a) and sst2(b) receptor isoforms
D1 transcription involves β-catenin and CREB pathways 873–879 (2002). mediate opposite effects on cell proliferation. Br. J.
in gastric cancer cells. Oncogene 23, 3689–3699 151. Chau, I. et al. Gastrazole (JB95008), a novel Pharmacol. 125, 1630–1633 (1998).
(2004). CCK2/gastrin receptor antagonist, in the treatment 173. Ohki-Hamazaki, H., Iwabuchi, M. & Maekawa, F.
130. Song, D. H. et al. Gastrin stabilises β-catenin protein of advanced pancreatic cancer: results from two Development and function of bombesin-like peptides
in mouse colorectal cancer cells. Br. J. Cancer 92, randomised controlled trials. Br. J. Cancer 94, and their receptors. Int. J. Dev. Biol. 49, 293–300
1581–1587 (2005). 1107–11015 (2006). (2005).
131. Franco, A. T. et al. Activation of β-catenin by carcino- 152. Watson, S. A. et al. Gastrimmune raises antibodies
genic Helicobacter pylori. Proc. Natl Acad. Sci. USA that neutralize amidated and glycine-extended Acknowledgements
102, 10646–10651 (2005). gastrin-17 and inhibit the growth of colon cancer. The authors would like to acknowledge J. McClelland from
132. Wu, P. et al. Correlation between the expressions of Cancer Res. 56, 880–885 (1996). the Academic Unit of Cancer Studies for her input into the
gastrin, somatostatin and cyclin and cyclin-depend 153. Gilliam, A. D. et al. A phase II study of G17DT in diagrams within this article and E. Royal for the personal
kinase in colorectal cancer. World J. Gastroenterol. gastric carcinoma. Eur. J. Sur. Oncol. 30, 536–543 communication regarding her ongoing studies.
11, 7211–7217 (2005). (2004).
133. Xiao, D., Chinnappan, D., Pestell, R., Albanese, C. & 154. Ajani, J. A. et al. An open-label, multinational, Competing interests statement
Weber, H. C. Bombesin regulates cyclin D1 expression multicenter study of G17DT vaccination combined with The authors declare competing finantial interests: see Web
through the early growth response protein EGR-1 in cisplatin and 5-fluorouracil in patients with untreated, version for details.
prostate cancer cells. Cancer Res. 65, 9934–9942 advanced gastric or gastroesophageal cancer: the GC4
(2005). study. Cancer 106, 1908–1916 (2006).
DATABASES
134. Tsutsui, S. et al. Induction of heparin binding epidermal 155. Schwartsmann, G. et al. A phase I trial of the
The following terms in this article are linked online to:
growth factor-like growth factor and amphiregulin bombesin/gastrin-releasing peptide (BN/GRP)
Entrez Gene: http://www.ncbi.nlm.nih.gov/entrez/query.
mRNAs by gastrin in the rat stomach. Biochem. antagonist RC3095 in patients with advanced solid
fcgi?db=gene
Biophys. Res. Commun. 235, 520–523 (1997). malignancies. Invest. New Drugs 24, 403–412
APC | β-catenin | CCK2 | CDX2 | COX2 | EGF | EGFR | EGR1 |
135. Miyazaki, Y. et al. Gastrin induces heparin-binding (2006).
Hrh2 | HB-EGF | MUC2 | PKB | REG1α | SHH | SSTR2 | TGFα |
epidermal growth factor-like growth factor in rat 156. Reubi, J. C., Waser, B., Schaer, J. C. & Laissue, J. A.
TP53 | ZBP89
gastric epithelial cells transfected with gastrin Somatostatin receptor sst1–sst5 expression in normal
Entrez Protein: http://www.ncbi.nlm.nih.gov/entrez/query.
receptor. Gastroenterology 116, 78–89 (1999). and neoplastic human tissues using receptor
fcgi?db=protein
136. Tokunaga, A. et al. Clinical significance of epidermal autoradiography with subtype-selective ligands. Eur. J.
gastrin | GRP | somatostatin
growth factor (EGF), EGF receptor, and c-erbB-2 in Nucl. Med. 28, 836–846 (2001).
human gastric cancer. Cancer 75, 1418–1425 (1995). This remains the most comprehensive assessment FURTHER INFORMATION
137. Thomas, S. M. et al. Gastrin-releasing peptide of ‘functional’ somatostatin isoforms expressed by Wolfson Digestive Diseases Centre:
receptor mediates activation of the epidermal growth human gastric adenocarcinoma specimens. http://www.nottingham.ac.uk/wddc
factor receptor in lung cancer cells. Neoplasia 7, 157. Ferrand, A. & Wang, T. C. Gastrin and cancer: a review. Access to this interactive links box is free online.
426–431 (2005). Cancer Lett. 238, 15–29 (2005).

946 | DECEMBER 2006 | VOLUME 6 www.nature.com/reviews/cancer


© 2006 Nature Publishing Group

You might also like