Vous êtes sur la page 1sur 10

Carcinogenesis vol.30 no.3 pp.

377386, 2009
doi:10.1093/carcin/bgp014
Advance Access publication January 9, 2009

REVIEW
The COX-2/PGE2 pathway: key roles in the hallmarks of cancer and adaptation to the
tumour microenvironment

Alexander Greenhough, Helena J.M.Smartt, Amy The genetic changes that occur in colorectal cancer appear to more
E.Moore, Heather R.Roberts, Ann C.Williams, Christos or less follow the histological progression from small, pre-malignant
Paraskeva and Abderrahmane Kaidi1 adenomas, to advanced metastatic tumours, with both early and late
stages of the disease associated with the number of sequentially ob-
Cancer Research UK Colorectal Tumour Biology Group, Department of tained genetic alterations (reviewed in ref. 4). This implies that each
Cellular and Molecular Medicine, University of Bristol, University Walk, successive genetic hit confers an advantageous characteristic upon
Clifton, Bristol BS8 1TD, UK and 1The Wellcome Trust/Cancer Research UK the expanding tumour mass. While it may be true that numerous
Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge different genes can become altered during the development of a given
CB2 1QN, UK
tumour, it has also recently been proposed that all cancers arise and

To whom correspondence should be addressed. Tel: 44 117 331 2072; are maintained by the deregulation of a relatively small number of
Fax: 44 117 928 7896; signalling pathways (1,5). This is an attractive premise from a thera-
Email: c.paraskeva@bristol.ac.uk peutic standpoint, given that there are far fewer cancer-related path-
It is widely accepted that alterations to cyclooxygenase-2 (COX-2) ways than cancer-related genes (1).
expression and the abundance of its enzymatic product prosta- While perturbation of the WNT-signalling pathway is believed to
glandin E2 (PGE2) have key roles in influencing the development account for the initiation of colorectal tumours (6), the aberrant ex-
of colorectal cancer. Deregulation of the COX-2/PGE2 pathway pression of cyclooxygenase-2 (COX-2) that occurs in the majority of
appears to affect colorectal tumorigenesis via a number of distinct colorectal tumours is thought to play a crucial role during colorectal
mechanisms: promoting tumour maintenance and progression, cancer development (7). Deregulation of COX-2 expression leads to
encouraging metastatic spread, and perhaps even participating an increased abundance of its principal metabolic product, prostaglan-
in tumour initiation. Here, we review the role of COX-2/PGE2 din E2 (PGE2), the pleiotropic effects of which appear to affect most,
signalling in colorectal tumorigenesis and highlight its ability to if not all, of the hallmarks of cancer. This ability of the COX-2/PGE2
influence the hallmarks of cancerattributes defined by Hana- pathway to affect multiple aspects of cell physiology required for
han and Weinberg as being requisite for tumorigenesis. In addi- tumour development and maintenance may also offer an explanation
tion, we consider components of the COXprostaglandin pathway for the effectiveness of COX inhibitors and non-steroidal anti-
emerging as important regulators of tumorigenesis; namely, the inflammatory drugs (NSAIDs) at reducing both the incidence and
prostanoid (EP) receptors, 15-hydroxyprostaglandin dehydroge- progression of intestinal tumours in animal models of cancer, and
nase and the prostaglandin transporter. Finally, based on recent more importantly in human cancer patients (reviewed in ref. 8).
findings, we propose a model for the cellular adaptation to the In this review, we discuss the impact of the COX-2/PGE2 pathway
hypoxic tumour microenvironment that encompasses the inter- on colorectal cancer development and highlight its influence on each
play between COX-2, hypoxia-inducible factor 1 and dynamic of the hallmarks of cancer. Furthermore, we consider the emerging
switches in b-catenin function that fine-tune signalling networks regulators of COXprostaglandin signalling and novel roles for
to meet the ever-changing demands of a tumour. COX-2/PGE2 signalling in the tumour microenvironment.

Cyclooxygenases and prostaglandins


A plethora of data indicates that human cancer is a multistage genetic NSAIDs such as aspirin have been used traditionally as analgesics and
and epigenetic disease. Like all cancers, colorectal cancers are anti-inflammatories for centuries and are some of the worlds most
thought to originate from a single replication-competent cell (stem widely used drugs. The seminal work by Sir John Vane delineated the
cell or proliferative progenitor cell) (1). In colorectal tumours, the mechanism by which aspirin exerts its anti-inflammatory, analgesic
initiating event is thought to be the acquisition of a genetic alteration and antipyretic actions. In the early 1970s, the enzyme prostaglandin
that deregulates the gatekeeping pathwaythe adenomatous poly- G/H synthase, now more often referred to as COX, was elucidated as
posis coli (APC)/b-catenin pathway (also known as the WNT-signal- the target for inhibition by aspirin and other NSAIDs (9). Since this
ling pathway)with tumour progression occurring through clonal discovery, there has been a great deal of interest in identifying the
selection via the protracted acquisition of multiple genetic lesions components of the COX pathway as well as its functions in both
(1,2). These alterations, while having the potential to produce a wide physiological and pathological conditions. COX enzymes play key
array of different cancer cell genotypes, have been proposed to affect roles in the biosynthesis of prostaglandins from arachidonic acid fol-
six principal aspects of cell physiology that are obligate for cancer lowing its release from the plasma membrane by the action of phos-
development. These are termed the hallmarks of cancer (3): ac- pholipase-A2 (reviewed in ref. 10). In the initial cyclooxygenase
quired capabilities that represent breaches to the normal regulatory reaction, the COX enzymes catalyse the formation of the unstable
mechanisms controlling cell survival, proliferation, migration, inva- intermediate prostaglandin G2 from arachidonic acid, which is then
sion and the interactions with neighbouring cells and stroma. converted into prostaglandin H2 by the peroxidase activity of COX
(11). Prostaglandin H2 is the precursor for several structurally related
prostaglandins, which are formed by the action of specialized prosta-
glandin synthases (reviewed in ref. 12). The prostaglandins synthe-
Abbreviations: APC, adenomatous polyposis coli; COX, cyclooxygenase; sized by this pathway include the aforementioned PGE2, as well as
EGFR, epidermal growth factor receptor; ERK, extracellular signal-regulated pro- prostaglandin D2, prostaglandin F2a (PGF2a), prostaglandin I2 (also
tein kinase; GSK3b, glycogen synthase kinase-3 beta; HIF-1, hypoxia-inducible
known as prostacyclin) and thromboxane-A2. The actions of these
factor-1; MAPK, mitogen-activated protein kinase; NSAID, non-steroidal anti-
inflammatory drug; PGE2, prostaglandin E2; PGES, prostaglandin E synthase;
prostanoid ligands are mediated by their engagement of specific
PGF2a, prostaglandin F2a; PGT, prostaglandin transporter; PI3K, phosphatidyli- cell-surface G-protein-coupled receptors designated EP14 for
nositol-3-OH kinase; PPAR, peroxisome proliferator-activated receptor; PGE2 and prostaglandin F2a receptor, thromboxane A2 receptor,
15-PGDH, 15-hydroxyprostaglandin dehydrogenase; TCF, T-cell factor; TGFb, prostaglandin I2 receptor and thromboxane A2 receptor for PGF2a,
transforming growth factor-beta; VEGF, vascular endothelial growth factor. prostaglandin D2, prostaglandin I2 and thromboxane-A2, respectively.

The Author 2009. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oxfordjournals.org 377
A.Greenhough et al.

These prostaglandins are important for a large number of normal body in mice has been demonstrated sufficient to retard the growth of
physiological processes in a broad range of tissues. These include transplantable tumours in vivo (39). In addition, PGE2 has been found
the modulation of immune responses, protection of the gastrointes- to result in augmented incidence and multiplicity of carcinogen-
tinal mucosa, maintenance of renal homeostasis and the regulation induced colon tumours in rats (40) and enhances intestinal adenoma
of blood clotting. Furthermore, prostaglandins also function in path- growth in ApcMin/ mice (41). Accumulating evidence from in vitro
ological conditions where they can promote inflammation, swelling, studies has further substantiated the in vivo evidence and also pro-
pain and fever (reviewed in ref. 10). vided insights into the mechanisms underlying the oncogenic role of
Later work on the biology of COX enzymes led to the distinction PGE2 (reviewed in ref. 42). Nevertheless, it is worth noting that while
between two COX enzymes: COX-1 (13) and COX-2 (14). A third good evidence exists suggesting that COX-2/PGE2 pathway inhibition
form (COX-3) has also been reported (15), although recent studies may be useful in the prevention of colorectal tumours, this may not be
indicate that this represents a splice variant of COX-1 that encodes the case under all circumstances. Indeed, COX-2/PGE2 signalling is
a truncated protein lacking enzymatic activity (16). In humans, COX- likely to act in concert with other important signalling pathways that
1 is found constitutively expressed in a wide range of tissues including become deregulated in cancer, meaning that such pathways would
the kidney, lung, stomach, small intestine and colon. Thus, COX-1 is also need to be targeted in conjunction with the COX-2/PGE2 path-
considered a housekeeping enzyme responsible for maintaining basal way for efficient prevention (or treatment) of colorectal cancer.
prostaglandin levels important for tissue homeostasis. In contrast, While there is a large body of data indicating an important tumour-
most tissues do not normally express COX-2 constitutively, notable promoting role for COX-2 during tumorigenesis, it is important to note
exceptions including the central nervous system (17), kidneys (18) that some reports imply that the role of COX-2 and/or prostaglandins in
and seminal vesicles (19). However, the stimulation of COX-2 expres- cancer might not be as straightforward as initially proposed. For exam-
sion in Src-transformed fibroblasts (14), endothelial cells and mono- ple, some NSAIDs produce antitumoral effects in vitro independently
cytes treated with the tumour promoter tetradecanoyl-phorbol-acetate of their ability to inhibit COX-2 or to reduce PGE2 synthesis (4345),
(20) or lipopolysaccharide (21) led to the notion that COX-2 is an and furthermore, some NSAIDs actually induce COX-2 expression
inducible enzyme that produces prostaglandins during inflammatory (46,47). In addition, the COX-2/PGE2 pathway may even elicit what
and tumorigenic settings. Because of this, there has been a fervent might be considered counterintuitive effects under certain circumstan-
interest in studying the biology of COX-2 in relation to tumorigenesis, ces, by acting in a tumour-suppressive manner (4851). In one study,
particularly with regard to colorectal tumorigenesis where its actions the authors investigated the effects of exogenous administration of the
appear to have a major impact on tumour development. stable synthetic PGE2 analogue 16,16-dimethyl-PGE2 on tumour de-
velopment in ApcMin/ mice; unexpectedly, the authors observed reduc-
Cyclooxygenases and colorectal cancer tions in both the number and size of intestinal tumours (48).
The first indication that COX enzymes might play a role in colorectal Furthermore, transgenic mice engineered to over-express COX-2 via
tumorigenesis came from the observation that patients with Gardners the keratin 14 promoter (resulting in COX-2 over-expression and ele-
syndrome (which causes multiple intestinal polyposis) treated with vated PGE2 levels in the skin) were found to be more resistant, rather
NSAIDs displayed a reduction in adenoma number (22). This was the than more sensitive, to the development of skin tumours induced by an
first clinical evidence that NSAIDs could be useful in the prevention initiation/promotion protocol (49). These findings contrast with other
of cancer. Subsequently, in 1991, Thun et al. (23) undertook an epi- similar studies reporting tumour-promoting effects of COX-2/PGE2
demiological study, which reported that regular aspirin intake at low signalling in mice (41,52,53), adding further layers of complexity
doses reduces the risk of colorectal cancer, and suggested the link to the role of the COX-2/PGE2 pathway during intestinal tumour
between COX enzymes and cancer development. A large body of development.
evidence now indicates that elevated levels of COX-2 are present in It is not clear what accounts for the apparent disparities between
the majority of colorectal carcinomas (2428) and in a subset of these studies, but a number of possibilities can be considered. For
adenomas (24,25). The observation that COX-2 expression is up- instance, it could be speculated that the precise level of PGE2 cells
regulated in both pre-malignant as well as malignant colorectal tissue are exposed to may result in different outcomes in a given experimen-
is regarded as particularly significanthaving potential implications tal system. Indeed, we have previously reported that human colorectal
for both the prevention and treatment of colorectal cancer (29). adenoma cells are growth stimulated by low concentrations of PGE2,
Several lines of evidence now support a critical role for COX-2 but growth inhibited by high PGE2 concentrations (54). Conversely,
during colorectal tumorigenesis. Most convincingly, in a randomized colorectal carcinoma cells are growth stimulated by both low and high
double-blind placebo-controlled trial, administration of the COX-2- concentrations of PGE2 (54). We hypothesize that during adenoma
selective NSAID celecoxib significantly decreased the occurrence of progression, further eventssuch as over-expression of the EP4
sporadic colorectal adenomas (30). Notably, in this scenario, celecox- receptormay be necessary for tumours to become growth stimulated
ib might potentially act not only by suppressing the growth of existing by high concentrations of PGE2. Therefore, it is possible that the use
adenomas but also by preventing the formation of new adenomas (30). of stable PGE2 (16,16-dimethyl-PGE2) (48) led to high levels and/or
Similarly, in a study of patients with familial adenomatous polyposis, accumulation of PGE2, resulting in the inhibition of adenoma growth.
administration of celecoxib was associated with regression of Another interesting possibility, as noted earlier, is whether there could
adenomas of both the colon and the rectum (31). By the same token, perhaps be an important role for other genetic lesions occurring in
studies have revealed that genetic disruption or pharmacological in- parallel to deregulation of the COX-2/PGE2 pathway; that is, addi-
hibition of COX-2 results in a substantial decrease in adenoma size tional changes might be required for COX-2 to promote tumorigenesis
and number in murine models of intestinal tumorigenesis [(32,33), efficiently. Thus, although much of data suggest an important role for
respectively]. COX-2/PGE2 signalling in the promotion of tumorigenesis, further
The pro-tumorigenic effects of COX-2 in the colorectum are largely research is still required in order to elucidate its precise roles in
thought to be attributed to its role in producing PGE2. Indeed, in- specific cellular and experimental contexts.
creased levels of PGE2 have been reported both in human colorectal
adenomas as well as carcinomas (34,35). PGE2 levels have also been
reported to increase in a size-dependent manner in the adenomas of COX-2, prostaglandins and their contribution to the hallmarks of
familial adenomatous polyposis patients (36) as well as in the adeno- cancer
mas of ApcMin/ mice (37). Furthermore, in vivo studies have revealed As mentioned previously, COX-2 is frequently over-expressed in co-
that prevention of adenoma development in familial adenomatous lorectal cancer, and PGE2 has been identified as the principal prosta-
polyposis patients is more effective when tissue prostaglandin levels noid promoting cell growth and survival in colorectal tumours. PGE2
are reduced through NSAID treatment (38), and more specifically, is able to exert pleiotropic effects in colorectal tumours, promoting
sequestration of PGE2 by administration of a PGE2 monoclonal anti- proliferation, survival, angiogenesis, migration and invasion. Here,

378
The COX-2/PGE2 pathway

we discuss the influence of the COX-2/PGE2 pathway on the hall- velopment, there remains an interesting debate as to the precise role
marks of cancer (3). PPARd plays during intestinal tumorigenesis (74). It is also interesting
to note that in addition to the apoptosis-suppressive effects of COX-2-
derived PGE2, deregulated expression of the COX-2 protein itself
Evasion of apoptosis might also alter the susceptibility of cells to undergo apoptosis by
Apoptosis, the process of programmed cell death (55), is a critical reducing the cellular pool of its substrate arachidonic acid, which can
mechanism by which metazoan organisms control cell number, where stimulate apoptosis by stimulating ceramide production (45).
selective cell suicide enables the efficient removal of superfluous, While the exact mechanism by which the COX-2/PGE2 pathway
damaged or infected cells (for reviews, see refs 56,57). Apoptosis suppresses apoptosis remains to be fully elucidated, it is clear that the
plays an essential role during embryonic development and is required influence of the COX-2/PGE2 pathway has on this hallmark of cancer
for maintaining tissue homeostasis throughout the lifespan of meta- plays an important role in aiding tumour progression. For instance,
zoan organisms. Disturbances to the apoptotic machinery that result in under conditions of hypoxiaa situation conducive to the induction
excessive cell survival or cell death underlie a number of pathological of cell deathPGE2 appears to promote cell survival in colorectal
conditions, but most pertinent here, the failure to initiate cell death in tumour cells by the stimulation of Ras-MAPK signalling (75). As well
response to apoptotic stimuli is thought to play a central role in as promoting the survival of cancer cells under harsh microenviron-
tumorigenesis (58). Indeed, deregulated cell proliferation, coupled mental conditions, PGE2 might also play a crucial role in encouraging
with an acquired resistance to apoptosis, has been proposed to con- the transition from adenoma to carcinoma. We have previously re-
stitute a platform both necessary and sufficient for tumour growth and ported that increased EP4 receptor expression occurs during the co-
malignant progression (59,60). lorectal adenomacarcinoma sequence (54) and that EP4 receptor
While the acquired ability of tumour cells to evade apoptosis can over-expression confers an anchorage-independent phenotype to oth-
arise via a variety of mechanisms, the majority of such changes result erwise anchorage-dependent human colorectal adenoma cells (54).
in an impaired ability of a cell to engage the intrinsic cell death This suggests an important role for a COX-2/PGE2/EP4-axis in the
machinery (i.e. the mitochondrial pathway of apoptosis). This path- suppression of apoptosis; indeed, in a recent complementary study,
way is governed by the ratio of pro-apoptotic and pro-survival BCL-2 Hull and colleagues (76) reported that EP4 receptor over-expression
family members that set a threshold for activation of the apoptotic results in the suppression of apoptosis and enhanced tumorigenic
caspase cascade upstream of the mitochondria (61). Establishing the behaviour of the human colon cancer cell line HT29 (76).
connection between the receipt of extracellular signals and the acti- Finally, it is important to note that while the ability of the COX-2/
vation of pathways such as the phosphatidylinositol-3-OH kinase PGE2 pathway to suppress intrinsic cell death mechanisms might
(PI3K)/AKT (also known as protein kinase B) and Ras-mitogen- facilitate cancer development, it may also confer a decreased sensi-
activated protein kinase (MAPK)/extracellular signal-regulated pro- tivity of colorectal tumour cells to cytotoxic cancer treatments. For
tein kinase (ERK) cascades to the suppression of apoptosis via the example, it has previously been reported that PGE2 attenuates radia-
intrinsic cell death machinery was a crucial finding of recent years tion-induced apoptosis in colorectal cancer cells via activation of
(62,63). Indeed, this is likely to represent the mechanism by which the EGFR/AKT signalling and a mechanism that prevents the transloca-
COX-2/PGE2 pathway regulates apoptosis, via the autocrine and/or tion of pro-apoptotic Bax to the mitochondria (68). Observations such
paracrine effects of PGE2 acting at one or more of the four cell- as these merit further investigation into the potential of NSAIDs and/
surface EP receptors. or COX-2-selective inhibitors as adjuvants to current chemotherapy/
Although still not entirely clear, several mechanisms for the sup- radiation regimens for patients with colorectal cancer.
pression of apoptosis by the COX-2/PGE2 pathway have been sug-
gested, and engagement of one or more of the signalling pathways Self-sufficiency in growth signals
downstream of EP receptor activation may be responsible for this pro- The behaviour of normal cells in multicellular organisms is controlled
survival response. Moreover, the ability of the COX-2/PGE2 pathway by the coordinated regulation of complex signalling pathways, which
to control an apoptosis evasion programme in tumour cells may differ transduce signals from growth factors and cytokines into decisions
between cell types and depend on factors such as the tumour micro- that direct cell fate. It is thought that the deregulation of these signal-
environment (for example, in hypoxic conditions). The initial indica- ling networks underlies tumour initiation and progression, causing
tion that COX-2 might modulate the intrinsic pathway of apoptosis cells to grow uncontrollably and unregulated by environmental cues.
came when its forced over-expression led to elevated levels of the pro- Two major pathways frequently mutated in many human cancers,
survival protein BCL-2 and conferred increased resistance to the di- including colorectal, are the Ras-MAPK- and the PI3K/AKT-signalling
etary fibre fermentation product butyrate-induced apoptosis in rat pathways; activation of which stimulates cell growth, proliferation
intestinal epithelial cells (64). Interestingly, the first suggestion that and survival (reviewed in ref. 77,78). Deregulation of these signalling
chemopreventive agents might produce their effects via the induction pathways can occur by a variety of mechanisms, including alterations
of tumour cell apoptosis arose from studies with butyrate in colorectal to both upstream and downstream components of the pathways. Most
tumour cells (65); thus, the ability of COX-2 expression to suppress commonly, constitutive activation of the Ras-MAPK pathway occurs
butyrate-induced apoptosis suggests that the chemopreventive action via mutations to KRAS [in 50% of colorectal tumours (79)] or BRAF
of COX-inhibiting NSAIDs may act in part by re-sensitizing cells to [in 1020% of colorectal tumours (80)] genes, whereas deregulation
apoptosis-inducing luminal factors. Further studies indicated that the of the PI3K/AKT pathway occurs most commonly by PIK3CA mu-
mechanism by which COX-2/PGE2 might suppress apoptosis and in- tation [in 32% of colorectal tumours (81)], phosphatase and tensin
crease the expression of BCL-2 was via activation of the Ras-MAPK/ homolog mutations [in up to 20% of colorectal tumours (82)] or
ERK pathway (66). More recently, several studies indicate that the mutations in the pleckstrin homology domain of AKT [in 6% of co-
COX-2/PGE2 pathway might alter apoptotic thresholds by engaging lorectal tumours (83)].
a number of different signalling pathways. Indeed, PGE2 has been As already mentioned, COX-2-derived PGE2 is able to signal via
reported to activate pro-survival pathways including the PI3K/AKT the PI3K/AKT- and Ras-MAPK/ERK-signalling pathways to enhance
pathway (67,68), ERK signalling (69), cyclic adenosine monophos- cell survival. An interesting possibility arising from such studies is the
phate (cAMP)/protein kinase A signalling (70) and activation of prospect that the COX-2/PGE2 pathway represents an alternative
epidermal growth factor receptor (EGFR) signalling (71,72). PGE2 mechanism by which tumours acquire growth factor autonomyin
has more recently been shown to promote cell survival in murine the absence of PI3K pathway- and MAPK pathway-activating muta-
intestinal adenomas by indirectly transactivating the nuclear peroxi- tions. In other words, aberrant activation of the COX-2/PGE2 pathway
some proliferator-activated receptor (PPAR)-delta via a PI3K/AKT- might phenocopy-activating mutations in the PI3K/AKT and/or Ras-
dependent mechanism (52). However, while this and other evidence MAPK pathways, which could play an important role in promoting
(73) point to an important role for PPARd in intestinal tumour de- tumour progression. This supports the notion that while the cohort of

379
A.Greenhough et al.

alterations to cancer-related genes and proteins might differ between cycle arrest, become differentiated and are eventually shed in to the
individual tumours, alterations to seemingly disparate genes may in lumen. However, in colorectal tumours, aberrant activation of b-catenin
reality be functionally equivalent; bestowing one or more of the hall- (i.e. the WNT pathway) serves to block this normal differentiation
marks of cancer on cells by deregulating the same molecular pathway programme and maintains cells in a progenitor-like state (88). Recent
(1). Indeed, deregulation of the COX-2/PGE2 pathway has been shown evidence indicates that the COX-2/PGE2 pathway can activate the
to behave in a similar manner to constitutively active Ras in murine APC/b-catenin pathway (89). Thus, in the context of colorectal can-
intestinal adenomas, resulting in a positive feedback loop that boosts cer, where mutations in the APC/b-catenin pathway are common, the
COX-2 expression and further stimulation of tumour growth (41). This COX-2/PGE2 pathway might enhance the activation of the APC/
mechanism might also operate during hypoxic (as well as normoxic) b-catenin pathway to keep cells in a progenitor-like state. Further-
conditions. In fact, we have previously reported that during hypoxia, more, another attractive hypothesis would be that inappropriate acti-
the transcription factor hypoxia-inducible factor-1 (HIF-1) directly up- vation of the COX-2/PGE2 pathway discourages differentiation in the
regulates COX-2 expression and increases PGE2 production (75). This absence of APC/b-catenin mutations; in this scenario, the activation
leads to activation of the Ras-MAPK pathway, which may act in a pos- of the APC/b-catenin pathway by PGE2 might even be able to con-
itive feedback loop to maintain an active pro-survival COX-2/PGE2 tribute to tumour initiation. In addition, the ability of PGE2 to switch
pathway during hostile microenvironmental conditions (75). on b-catenin signalling might also contribute to the acquisition of an
immortalized phenotype, as discussed below.

Insensitivity to anti-growth signals Limitless replicative potential


As noted earlier, disturbances to normal tissue homeostasis that shift The intrinsic capacities for self-renewal and limitless replicative po-
the balance from a state of equilibrium towards increased cell growth tential are characteristics thought to be shared by stem cells and
will invariably lead to the appearance of a neoplastic population of cancer cells (90). Because of these apparent similarities, it has been
cells (84). Under normal physiological conditions, in addition to the proposed that cancer arises from the deregulation of pathways that
strict control of apoptosis and growth-stimulatory signals, a further maintain the stem/progenitor cell phenotype in a given tissue (6,91).
crucial mechanism for maintaining tissue homeostasis is via the re- As mentioned earlier, colorectal tumours are thought to originate from
straint of cell growth through the action of anti-proliferative signals such replication-competent cells (i.e. a proliferative progenitor cell or
(3). These include soluble growth inhibitors, as well as membrane- stem cell) initiated by activating mutations in the APC/b-catenin
bound ligands, both of which act on cell-surface receptors that couple pathway. Perturbations to the APC/b-catenin pathway are detectable
to intracellular pathways to repress cell growth. Hanahan and Weinberg in the earliest lesions of the colorectal epithelium (aberrant crypt foci)
(3) proposed two discrete mechanisms by which anti-growth signals and are thought to occur in virtually all colorectal tumours. This most
can block proliferation to restrain inappropriate cell growth and main- commonly arises by loss of the APC tumour suppressor gene, occur-
tain tissue homeostasis. Importantly, both of these mechanisms are ring in 80% of all colorectal tumours (92). The tumour-suppressive
frequently hijacked in colorectal cancers. Furthermore, deregulation action of APC is thought to be mediated in large part by its ability to
of the COX-2/PGE2 pathway may provide an additional and/or sec- regulate the levels of intracellular b-catenin, which transduces signal-
ondary mechanism by which tumour cells can attain this hallmark. ling by the WNT ligand. The mutations in either APC or indeed other
The first mechanism by which anti-growth signals can block pro- components of the WNT pathway observed in colorectal tumours lead
liferation is by preventing cells from advancing through the G1 phase to the formation of a constitutively active b-cateninT-cell factor
of the cell cycle, which serves to maintain cells in a quiescent (G0) (TCF) complex that mimics active WNT signalling. Evidence sug-
state. The archetypal example of such an anti-proliferative signal is gests that the proliferative compartments (stem cells and progenitor
the soluble signalling factor transforming growth factor-beta (TGFb), cells) in intestinal crypts are maintained through activation of the
which blocks progression through the G1 phase of the cell cycle via WNT pathway (88). Indeed, disruption of the WNT pathway by de-
the suppression of c-Myc and activation of cyclin-dependent kinase letion of Tcf4 in mice results in loss of the stem cell and proliferative
inhibitors such as p15Ink4B and p21Cip1 (reviewed in ref. 85). Often, compartments in the small intestine (93). This suggests that the WNT
cancer cells acquire the inability to respond to the growth-suppressive pathway maintains the crypt stem/progenitor cell phenotype and that
effect of TGFb, which can occur as a result of inactivating mutations the same pathway is active in both colorectal cancer cells and the
in the downstream signalling effectors of TGFb signalling (e.g. loss of stem/progenitor cells of the intestinal crypt.
SMAD function) or mutations to the receptors themselves (reviewed The recently discovered connection between the WNT pathway
in ref. 85). The latter mechanism is particularly relevant in colorectal and the COX-2/PGE2 pathway (89) indicates that PGE2 signalling
tumours with microsatellite instability, where TGFb type II receptor may be able to contribute to the crypt progenitor phenotype by acti-
mutations occur at a high frequency (86). However, not all cancer vating b-catenin/TCF signalling in colorectal cancer cells. Gutkind
cells exhibit mutational inactivation of the TGFb type II receptor, and colleagues (89) reported that upon engagement of the EP2 re-
and a further mechanism by which cancer cells lose TGFb respon- ceptor, PGE2 stimulates a dual-signalling cascade involving activation
siveness is via the down-regulation of the TGFb receptors. Indeed, of the PI3K/AKT pathway by the EP2-associated G-protein bc sub-
this could represent a mechanism by which the COX-2/PGE2 pathway units and the association of the G-protein as subunit with Axin (89).
prevents the receipt of anti-growth signals since over-expression of This leads to the activation of b-catenin by a dual mechanism: first,
COX-2 has been reported to cause down-regulation of the TGFb type the activation of AKT inhibits glycogen synthase kinase-3 beta
II receptor (64). (GSK3b), which reduces the inhibitory effect of GSK3b-mediated
A second mechanism by which cells can be instructed to curb their phosphorylation on b-catenin; and second, the complex between the
proliferative potential involves the implementation of differentiation EP2-associated G-protein as subunit and Axin promotes the release of
programmes that instigate an irreversible post-mitotic (or terminally b-catenin from the AxinGSK3b complex, freeing b-catenin and
differentiated) state (3). An example of such an anti-growth pathway facilitating its nuclear accumulation (89). Thus, by activating the
operates in the colorectal epithelium and is often deregulated in neo- b-catenin/TCF pathway, it is tempting to speculate that COX-2/
plasia. Under normal physiological conditions in the colonic crypt, PGE2 signalling might contribute to limitless replicative potential
the long-lived stem cells continually give rise to rapidly proliferating by promoting the acquisition of a progenitor or stem cell-like pheno-
progenitor cells (also known as transit-amplifying cells), which com- type; although, at present, whether COX-2/PGE2 signalling is able to
prise the proliferative region in the lower two-thirds of the crypt. promote such a phenotype is unknown.
These progenitor cells are able to differentiate into the cell lineages In addition, it is also possible that the COX-2/PGE2 pathway, by
that occupy the upper third of the crypt: absorptive enterocytes, enter- enhancing cell survival and growth, serves to prime cells for the
oendocrine cells and mucin-secreting goblet cells (87). Upon reaching acquisition of further cellular alterations that contribute to immortal-
the top third of the crypt, committed progenitor cells undergo cell ization and the progression towards the full malignant phenotype.

380
The COX-2/PGE2 pathway

Lastly, it is of interest to note that recent studies suggest a role for metastatic potential of colorectal tumours in both humans (106) and
COX-2/PGE2 signalling in the maintenance of haematopoietic stem mice (107), over-expression of COX-2 in intestinal cells can modulate
cell homeostasis (94) and in the suppression of embryonic stem cell their adhesive properties (64) and increase matrix metalloproteinase
apoptosis (95). Whether the COX-2/PGE2 pathway influences the activity to promote invasion (108). Further studies have demonstrated
behaviour of stem cells of the intestinal and colonic epithelium awaits that PGE2 promotes cytoskeletal reorganization and increases colorec-
investigation. tal cancer cell migration and invasion via PI3K signalling (67). The
stimulation of invasion and motility by PGE2 is dependent on the
intracellular Src-mediated transactivation of the EGFR (72). Further-
Sustained angiogenesis more, the hepatocyte growth factor receptor, c-Met, is also transacti-
In the course of solid tumour development, it is well recognized that the vated by PGE2 in an EGFR-dependent fashion in colorectal cancer cells
avascular tumour mass becomes dependent on angiogenesis for main- (109). Hepatocyte growth factor/c-Met signalling is classically associ-
tenance and progression, leading to the concept known as the angio- ated with loss of cellcell contact (or scattering) and invasive growth
genic switch (96). The initiation of this morphogenic process is (110). Transactivation of c-Met by PGE2 leads to increased nuclear
controlled by the relative balance of pro-angiogenic and anti-angio- b-catenin accumulation, increased urokinase-type plasminogen activa-
genic factors in a given tissue, such that a tipping of the balance towards tor receptor expression and invasion through matrigel (109). Further-
pro-angiogenic factors results in the stimulation of blood vessel forma- more, COX-2, c-Met and b-catenin are co-expressed at the invasive
tion (96). In the case of colorectal cancer, over-expression of COX-2 in front of colorectal tumour specimens (109). Although PGE2 is the most
colon cancer cells induces the production of angiogenic factors such as abundant prostaglandin found in colorectal cancer tissue (35), PGF2a
vascular endothelial growth factor (VEGF) and basic fibroblast growth has also been detected in colorectal tumours, and PGF2a can stimulate
factor, which are instrumental in stimulating the formation of new motility in both colorectal carcinoma and adenoma cells and invasion
blood vesselsa requirement for tumours should they wish to develop of colorectal carcinoma cells (111).
beyond a few millimetres in size. These pro-angiogenic effects of COX- More recently, the significance of COX-2not only as a critical player
2 can be inhibited by NSAIDs, resulting in the inhibition of endothelial in tumour development but also as being necessary for dissemination of
cell proliferation, migration and vascular tube formation in vitro (97). cancer cells to other organswas demonstrated during an elegant series
The inhibitory effect of NSAIDs on angiogenesis in this context can be of experiments in an in vivo model of breast cancer cell metastasis to the
rescued by adding exogenous PGE2 (98), suggesting that COX-2- lungs (112). In this study, using both genetic and pharmacological
derived PGE2 is at least part responsible for the pro-angiogenic effects approaches, the authors identified COX-2 as one of four key metastasis
of COX-2 over-expression; although it is important to note that pros- progression genes, which collectively synergize to mediate both tu-
taglandin-independent mechanisms of NSAID action on angiogenesis mour development and metastasis to other organs. Notably, the tumour
were also reported (98). Consistent with this, PGE2 has been reported to maintenance and progression functions of COX-2 were mediated solely
stimulate VEGF expression in colon cancer cells through the activation by the COX-2 expressed by the tumour cells themselvesas opposed to
of HIF-1, a key regulator of VEGF expression (99). Furthermore, in COX-2 expression and PGE2 production by other cells (i.e. stromal
vivo studies have also demonstrated an important role for the COX-2/ cells) in the tumour microenvironment (100,113). Therefore, these find-
PGE2 pathway in angiogenesis. In mice, homozygous deletion of COX- ings contribute to the ongoing discussion concerning the roles of tu-
2 (but not COX-1) results in impeded growth of tumour xenografts and mour-derived and stroma-derived COX-2 during the different stages of
is associated with a reduced tumour vascular density (100). Such ob- tumorigenesis and within specific cellular contexts (114).
servations are likely to be at least in part mediated by the resultant
reduction in PGE2, since the intestinal adenomas present in APCD716/
mice that are null for the PGE2 receptor EP2 are defective in VEGF Evasion of the antitumour immune response and its regulation by the
induction (101). One mechanism by which COX-2 might promote COX-2/PGE2 pathway
tumour vascularization is via the production of PGE2 and prostaglan- In addition to the six hallmarks of cancer originally proposed by
din I2, which have been shown to participate in inducing endothelial Hanahan and Weinberg, additional characteristics of tumours have
cell spreading and migration by integrin aVb3-mediated activation of recently been proposed, one of which is the ability of tumours to
the small guanosine 5#-triphosphatases Cdc42 and Rac (102). More evade attack by the immune system (reviewed in ref. 115). Tumour-
recently, PGE2 has been shown to regulate angiogenesis though the specific antigens are capable of eliciting an antitumour immune re-
modulation of chemokine receptor signalling: PGE2 can enhance sponse involving cytotoxic CD8 T cells, resulting in tumour cell
VEGF and basic fibroblast growth factor-induced chemokine recep- lysis. However, in cancer patients, these antitumour immune re-
tor-4 that is important for microvessel assembly in vivo (103). In sponses are generally ineffective and increasing evidence suggests
addition, PGE2 can induce the expression of the pro-angiogenic che- that tumours evolve several strategies to escape tumour-specific im-
mokine CXCL-1 in vivo (104). PGE2 may also work concomitantly munity (116,117). In light of the known ability of PGE2 to modulate
with the hypoxic tumour microenvironment to orchestrate the process multiple aspects of the immune response (118), this additional cancer
of angiogenesis, as discussed later (75). hallmark may therefore be influenced by inappropriate expression of
COX-2 and PGE2 production that occurs commonly in colorectal
tumours. Furthermore, COX-2 over-expression and the resulting in-
Tissue invasion and metastasis crease in PGE2 levels may represent a strategy adopted by tumours
It is well established that the primary cause of cancer mortality is the that contributes to the evasion of tumour-specific immune response
formation of distant metastases, making the capacity of tumour cells to (119). For example, PGE2 has been reported to shift the production of
invade and metastasize one of the most pertinent hallmarks of cancer cytokines by antigen-presenting dendritic cells, away from a Th1
from a therapeutic perspective. In order to achieve metastases, cancer (type 1 T cell) profile, leading to a reduced activation of antitumour
cells must exhibit a more motile, invasive phenotype, dissociate from cytotoxic CD8 T cells (118120). This ability of PGE2 to suppress
neighbouring cells within the tumour, invade through extracellular ma- these immune responses may allow tumour cells to escape immuno-
trix components and intravasate into local blood or lymphatics (re- surveillance, adding to the already countless roles of the COX-2/PGE2
viewed in ref. 105). Having made their escape from the primary pathway during tumour development.
tumour, cancer cells must then extravasate from the blood or lym-
phatics into the surrounding tissue in order to colonize distant sites.
Several lines of evidence indicate that COX-2 and the prostaglandins Emerging regulators of the COX-2/PGE2 pathway: PGE2 synthesis and
play important roles in aiding these processesmore specifically, signalling as alternative chemopreventive/therapeutic targets
PGE2 is thought to promote a more metastatic phenotype in colorectal Until relatively recently, COX expression was believed to be the
tumour cells. While inhibition of COX-2 in vivo can attenuate the major determinant of PGE2 levels, but recent data indicate that

381
A.Greenhough et al.

multiple levels of control exist for the regulation of PGE2 production Prostaglandin synthesis and degradation are believed to be com-
and turnover. In addition to regulation via COX enzymes, PGE2 syn- partmentalized within the cell. This implies that following synthesis
thesis can be regulated via expression of specific prostaglandin and release, prostaglandins must be re-imported into the cell for deg-
E synthases (PGES), which act on the COX product prostaglandin radation by the cytoplasmic-localized 15-PGDH. Prostaglandins are
H2 to produce PGE2 (reviewed in ref. 42,121). Moreover, in addition believed to traverse membranes poorly and their cellular uptake is
to regulated synthesis, PGE2 levels are also determined by the rate of thought to occur mostly via PGT (also known as solute carrier organic
its degradation, which can be regulated via the expression of 15- ion transporter family, member 2A1 or SLCO2A1) (reviewed in ref.
hydroxyprostaglandin dehydrogenase (15-PGDH) and the prostaglan- 132). Indeed, in HeLa cells, co-expression of both 15-PGDH and PGT
din transporter (PGT) (122). The cellular response to PGE2 can also is required for metabolism of exogenously added PGE2 (122). Simi-
be regulated via the differential expression of the four PGE2 receptors larly to 15-PGDH, PGT was recently shown to be down-regulated in
(EP14), which activate distinct downstream signalling responses (re- colorectal neoplasia in both humans and ApcMin/ mice (138), sug-
viewed in ref. 42,123). An understanding of these further levels of gesting that PGE2 signalling can be further controlled via regulated
regulation controlling levels of (and response to) PGE2 may reveal uptake.
novel strategies for prevention and/or treatment of colorectal cancer. It would be therapeutically desirable to increase prostaglandin deg-
There are currently three characterized PGES enzymes: cytosolic radation in colorectal tumours but at present relatively little is known
PGES, microsomal PGES-1 and microsomal PGES-2 (reviewed about the potential for therapeutic re-expression of 15-PGDH and
in ref. 42). Microsomal PGES-1 is, like COX-2, induced by pro- PGT. However, PGT expression can be induced by the histone deace-
inflammatory stimuli and up-regulated in colorectal tumours (124). tylase inhibitor trichostatin A and the histone demethylating agent
Moreover, loss of microsomal PGES-1 expression is reported to sup- 5-azacytidine; two classes of drug relevant to cancer therapy (138).
press intestinal neoplasia in APC-mutant mice (125), although the re- Moreover, histone deacetylase inhibitors and 5-azacytidine can also
verse has also been observed (126), possibly reflecting the knock-on up-regulate 15-PGDH (139). Future advancements in our understand-
enhancement of synthesis of other prostaglandins with pro-tumorigenic ing of prostaglandin catabolism in the intestine should allow insight
effects (such as PGF2a). Further work will be required to ascertain the into the potential for targeting 15-PGDH and PGT in colorectal cancer
potential utility of PGES inhibition as an anticancer strategy in the colon. prevention and therapy.
As noted earlier, the four characterized receptors for PGE2 (EP14)
each show distinct downstream signalling effects. For example,
whereas activation of EP2 and EP4 receptors can both stimulate Regulation of COX-2 expression by the tumour microenvironment
cAMP production, activation of EP3 results in the inhibition of cAMP COX-2 up-regulation has been reported in a number of different
production (reviewed in ref. 42). Recent evidence suggests a role for tumour types (reviewed in ref. 140), and accordingly there has been
the EP receptors in colorectal neoplasia. As previously discussed, we a great deal of interest in attempting to achieve a greater understand-
have recently showed that up-regulation of EP4 occurs during human ing of the regulatory networks that control COX-2 expression.
colorectal tumorigenesis in vivo (54) and others have shown that Whereas activating mutations in the PTGS2 gene (the gene encoding
genetic or pharmacological inactivation of EP4 inhibits tumour COX-2 in humans) have not yet been described, there are several
growth in a mouse model of intestinal neoplasia (127). Other PGE2 known mechanisms underlying enhanced COX-2 expression in tu-
receptors are also probably play a role in colorectal neoplasia; for mour cells. Principally, these mechanisms include deregulated
example, homozygous deletion of the gene encoding the EP2 receptor growth factor signalling and oncogene activation. Examples of these
reduces adenoma size and number in ApcD716/ mice (101). In addi- mechanisms include activation of the WNT pathway (141,142) and
tion, EP3 has been shown to stimulate angiogenesis and growth of the Ras-MAPK pathway (41,143) signalling via growth factor re-
tumours arising from implanted sarcoma cells in mice (128). ceptors including EGFR (144), TGFb receptors (145), c-Met (146)
However, down-regulation of EP3 has been observed in colorectal and gastrin receptors (147). More recently, our laboratory has shown
neoplasia and azoxymethane-induced tumorigenesis is enhanced in that COX-2 can be induced by the hypoxic microenvironment in
EP3-null mice, suggesting a tumour-suppressive role for EP3 in the colorectal tumour cells derived from both adenomas and carcinomas
intestine (129). It will be of interest to determine the contribution of (75). This up-regulation of COX-2 is transcriptional and is mediated
different PGE2 receptors to colorectal neoplasia and determine the by the master regulator of transcription during hypoxia, HIF-1.
potential utility of specific EP antagonists in anticancer therapy in COX-2 has also been reported to be up-regulated in lung cancer cells
the intestine (130,131). via a mechanism that is dependent on HIF-1 (148). COX-2 up-
Recent evidence suggests that deregulated catabolism of PGE2 also regulation during hypoxia has also been described previously in
plays a significant role in colorectal neoplasia. Prostaglandins are endothelial cells (149), monocytes (150) and corneal epithelial
essentially local-acting hormones with diverse effects in numerous cells (151); whereas in endothelial cells, COX-2 up-regulation ap-
different tissues. Therefore, it is crucial that their catabolism is strictly pears to be mediated by nuclear factor-kappa B (149), in corneal
regulated in order to prevent them from reaching distant organs in the epithelial cells COX-2 over-expression was linked to PPARa/b
bloodstream (reviewed in ref. 132). 15-PGDH is a cytosolic enzyme activation (151).
that inactivates prostaglandins by oxidation to their 15-keto form
(reviewed in ref. 133) and was recently shown to act as a colorectal
tumour suppressor (134,135). In the normal human colon, 15-PGDH The role of COX-2 and PGE2 during hypoxia
is expressed in the non-proliferative epithelium at the top of the crypts As noted above, the HIF-1-dependent COX-2 up-regulation during
and loss of 15-PGDH expression and activity is seen in human co- hypoxia is associated with an increase in PGE2 levels (75). Given
lorectal tumours (134,135). Several pathways deregulated in colorec- the pivotal role of PGE2 in colorectal tumorigenesis, these findings
tal neoplasia may contribute to this down-regulation as 15-PGDH is build on the already known roles of PGE2 by demonstrating a role for
negatively regulated by growth-stimulatory epidermal growth factor PGE2 in the promotion of cell survival under hypoxic conditionsa
signalling (up-regulated in colorectal neoplasia) and positively regu- property that tumour cells must acquire in order to propagate and
lated by TGFb (which colorectal cancer cells become resistant to) progress in vivo (152). Although the mechanism for this pro-survival
(134,136). Functional evidence for the tumour suppressor activity of attribute of PGE2 is not yet defined, it is likely to involve the alteration
15-PGDH in the intestine has been obtained using mouse models. of apoptotic thresholds by modulating the intrinsic cell death machin-
Loss of the gene encoding 15-PGDH in mice results in a doubling ery; for example, by increasing BCL-2 expression, which has been
of colonic mucosal PGE2 levels and promotes colorectal tumour previously demonstrated to promote cell survival under hypoxic con-
growth in ApcMin/ mice (137). Furthermore, restoring 15-PGDH ditions (153). The ability of hypoxia (154) and PGE2 (155) to activate
expression in colon cancer cells inhibits their ability to form tumours nuclear factor-kappa B may also be important in enhancing cell sur-
in immune-deficient mice (134). vival under hypoxic conditions. PGE2 may also confer alternative

382
The COX-2/PGE2 pathway

survival strategies to cells via the up-regulation of pro-survival factors whereas hampering the pro-proliferative effect of PGE2. The net ef-
such as inhibitor of apoptosis-2 (156), which has been shown to be up- fect of this selective strategy is enhanced tumour cell survival due to
regulated by hypoxia (157). a cell cycle arrest that enables the mitigation of hypoxia-induced
In addition to promoting cell survival under hypoxic conditions, the cellular stresses, which would otherwise compromise cell survival if
amplification of the COX-2/PGE2 pathway in hypoxic colorectal can- proliferation was allowed to continue during hypoxic conditions. This
cer cells may have important implications for stimulating angiogen- scenario may represent a spatialtemporal switching in PGE2 signal-
esis, a process that is considered critical for solid tumour growth and ling and function during colorectal tumorigenesis. This model is sum-
progression (96). While bearing in mind the important separate roles marized in Figure 1.
of both hypoxia and COX-2/PGE2 signalling in the promotion of The role of PGE2 in the activation of b-catenin/TCF signalling also
tumour angiogenesis, we suggest that during hypoxia PGE2 signalling suggests that PGE2 might serve an important function at early stages
may be integrated to act concordantly in the regulation of angiogen- of colorectal tumorigenesis. This highlights the possibility that basal
esis. Indeed, increased levels of PGE2 appear to enhance HIF-1 tran- PGE2 levels may switch on the b-catenin-signalling cascade, which in
scriptional activity and VEGF production in hypoxic colorectal turn would promote cell proliferation that may contribute to tumour
cancer cells (75). Another possible level for the integration between initiation. Given that COX-2 is not induced at the very early stages of
PGE2 and hypoxia signalling in the modulation of angiogenesis could colorectal tumorigenesis, COX-1 might be the source of PGE2 during
occur at the level of Src; a key factor in hypoxia-induced VEGF and tumour initiation, perhaps enhancing b-catenin/TCF-4 signalling
PGE2-mediated transactivation of EGFR that has implications for upon the loss of APC. This possibility is consistent with the findings
colorectal tumour progression (72). Furthermore, as mentioned pre- that loss of either COX-1 or COX-2 decreases tumour burden in
viously, PGE2 has been shown to enhance the expression of chemokine mouse models of intestinal tumorigenesis (53). This may also explain
receptor-4 (103), whose expression has been shown to be regulated by why inhibition of both COX-1 and COX-2 enzymes (for example with
HIF-1/hypoxia (158,159). The ability of PGE2 to enhance HIF-1 tran- aspirin or sulindac) is as effectiveif not potentially more soas
scriptional activity stands to be a potentially important observation. selective COX-2 inhibitors as chemopreventive agents for colorectal
Accordingly, PGE2 may promote the expression of several HIF-1 tar- cancer.
gets (in addition to VEGF) that are crucially involved in tumorigenesis.
If it turns out to be the case, PGE2 may then be regarded as a key Concluding remarks
positive modulator of the transcriptional response to hypoxia, which Compelling evidence gained from mechanistic studies with cancer
is believed to be critical for the progression of solid tumours including cell lines, mouse models of intestinal tumorigenesis and a number
colorectal cancer (99). The HIF-1-mediated transcriptional response of clinical trials with both non-selective and COX-2-selective
provides both short- and long-term adaptive strategies, including cell NSAIDs support an important role for the COXprostaglandin path-
survival, metabolic shift, angiogenesis, invasion and metastasis. There- way in the development of colorectal tumours. Furthermore, these
fore, further investigations into cross talk between HIF-1 and PGE2 studies have validated the COX-2/PGE2 pathway as a bona fide target
signalling may provide greater insight into the mechanisms that pro- for cancer chemoprevention and therapy. Evidence suggests that the
mote adaptation to hypoxia and hence tumour progression. effectiveness of suppressing the COX-2/PGE2 pathway in cancer pre-
vention and therapy stems from its role in driving the hallmarks of
Interplay between the COX-2/PGE2 pathway, HIF-1 and b-catenin
cancer, without which tumours cannot sustain their growth and
signalling
development.
One of the mechanisms by which COX-2/PGE2 signalling influences Recent clinical trials involving highly selective COX-2 inhibitors as
the hallmarks of cancer is via the ability of PGE2 to simulate the chemopreventive agents for colorectal cancer have cast doubt on the
activation of b-catenin, which leads to the stimulation of cell growth suitability of such drugs for long-term use due to increased risks of
and proliferation via its interaction with TCF-4 in the nucleus (89). adverse cardiovascular events (30,161). Notwithstanding, the COX-2
However, while in one study we found that COX-2/PGE2 levels are
increased in response to hypoxia (75), we also found that hypoxia
results in a transient cell cycle arrest that involves the inhibition of
b-catenin/TCF-4 activity (160). In the latter study, we found that the
cell cycle arrest induced by hypoxia in colorectal cancer cells results
from the direct binding of HIF-1a to b-catenin; this results in the
suppression of b-catenin/TCF-4 activity and a subsequent shutting
down of cell proliferation via the c-Myc-p21-axis (160). Furthermore,
we also revealed a novel role for b-catenin in the enhancement of HIF-
1 transcriptional activity, resulting in cell survival and adaptation to
the hypoxic microenvironment. Hypoxia, therefore, appears to shift
the balance from b-catenin signalling and co-activation of TCF-4 to
b-catenin interacting with and enhancing of HIF-1 activitythereby
favouring cell quiescence and survival over proliferation. This sce-
nario illustrates an intriguing dynamicity in b-catenin function in re-
sponse to conditions of low oxygen tension. The ability of b-catenin to Fig. 1. Under normoxic conditions, COX-2 expression can be induced
interact with and enhance HIF-1 transcriptional activity represents through the activation of the oncogenic pathways such as the Ras-MAPK
a novel function for b-catenin in colorectal tumorigenesis that is pathway. Furthermore, COX-2-derived PGE2 promotes cell proliferation at
applicable to hypoxic conditions and may have implications for other least in part via the stimulation of b-catenin/TCF-4 activity, as well as via
aspects of carcinogenesis including metabolic adaptation, invasion and enhancement of Ras-MAPK signalling. During hypoxia, b-catenin is
metastasis. displaced from TCF-4 (which results in TCF-4 repression) via its interaction
Together, our findings (75), with those of Castellone et al. (89), with HIF-1, and b-catenin enhances HIF-1 transcriptional activity. In
suggest on the one hand, a selective role for PGE2 in the stimulation of addition, COX-2 is up-regulated during hypoxia via HIF-1 activation,
cell proliferation that is restricted to normoxic conditions. On the resulting in increased levels of PGE2. Both enhanced HIF-1 transcriptional
activity and increased PGE2 levels promote cell survival leading to
other hand, during hypoxia, the proliferative effect of PGE2 may be adaptation to hypoxia. Whereas b-catenin acts as essential activator of TCF-4
overridden as a result of HIF-1 competing with TCF-4 for b-catenin, in normoxia, b-catenin results in the enhancement of HIF-1 transcriptional
causing the inhibition of b-catenin/TCF-4 activity and cell cycle arrest activity during hypoxia. These effects are likely to be transient and reversible
(160). Therefore, we propose a model in which hypoxia favours the (dynamic) depending on the tumour microenvironment and fluctuations in
pro-survival (anti-apoptotic) and pro-angiogenic functions of PGE2, oxygen levels.

383
A.Greenhough et al.

selective inhibitor celecoxib remains a clinically relevant agent for 20. Kujubu,D.A. et al. (1991) TIS10, a phorbol ester tumor promoter-induc-
colorectal cancer prevention and therapy, and further studies will be ible mRNA from Swiss 3T3 cells, encodes a novel prostaglandin synthase/
required to establish the usefulness of NSAIDs in chemoprevention cyclooxygenase homologue. J. Biol. Chem., 266, 1286612872.
particularly in patients at a high risk from colorectal cancer. Given the 21. Hla,T. et al. (1992) Human cyclooxygenase-2 cDNA. Proc. Natl Acad.
Sci. USA, 89, 73847388.
importance of COX-2/PGE2 in influencing the hallmarks of cancer, it
22. Waddell,W.R. et al. (1983) Sulindac for polyposis of the colon. J. Surg.
will also be important to determine the applicability of NSAIDs as Oncol., 24, 8387.
adjuvants to current chemotherapy/radiation regimens, not only for 23. Thun,M.J. et al. (1991) Aspirin use and reduced risk of fatal colon cancer.
colorectal cancer patients but also for patients with other tumour types N. Engl. J. Med., 325, 15931596.
displaying deregulated COX-2 expression. 24. Eberhart,C.E. et al. (1994) Up-regulation of cyclooxygenase 2 gene ex-
Future studies into the emerging players within the COX-2/PGE2 pression in human colorectal adenomas and adenocarcinomas. Gastroen-
pathway may reveal novel approaches for more safely targeting this terology, 107, 11831188.
pathway for both cancer chemoprevention and therapy. 25. Elder,D.J. et al. (2002) Human colorectal adenomas demonstrate a size-
dependent increase in epithelial cyclooxygenase-2 expression. J. Pathol.,
198, 428434.
Funding 26. Kargman,S.L. et al. (1995) Expression of prostaglandin G/H synthase-1
Cancer Research UK programme; The Citrina Foundation; The John and -2 protein in human colon cancer. Cancer Res., 55, 25562559.
27. Sano,H. et al. (1995) Expression of cyclooxygenase-1 and -2 in human
James Bristol Foundation.
colorectal cancer. Cancer Res., 55, 37853789.
28. Sheehan,K.M. et al. (1999) The relationship between cyclooxygenase-2
Acknowledgements expression and colorectal cancer. JAMA, 282, 12541257.
29. Elder,D.J. et al. (1998) COX-2 inhibitors for colorectal cancer. Nat. Med.,
Conflict of Interest Statement: None declared. 4, 392393.
30. Arber,N. et al. (2006) Celecoxib for the prevention of colorectal adeno-
matous polyps. N. Engl. J. Med., 355, 885895.
References 31. Steinbach,G. et al. (2000) The effect of celecoxib, a cyclooxygenase-2
inhibitor, in familial adenomatous polyposis. N. Engl. J. Med., 342,
1. Vogelstein,B. et al. (2004) Cancer genes and the pathways they control. 19461952.
Nat. Med., 10, 789799. 32. Oshima,M. et al. (1996) Suppression of intestinal polyposis in Apc
2. Fearon,E.R. et al. (1990) A genetic model for colorectal tumorigenesis. delta716 knockout mice by inhibition of cyclooxygenase 2 (COX-2). Cell,
Cell, 61, 759767. 87, 803809.
3. Hanahan,D. and Weinberg,R.A. (2000) The hallmarks of cancer. Cell, 33. Jacoby,R.F. et al. (2000) The cyclooxygenase-2 inhibitor celecoxib is
100, 5770. a potent preventive and therapeutic agent in the min mouse model of
4. Fodde,R. (2002) The APC gene in colorectal cancer. Eur. J. Cancer, 38, adenomatous polyposis. Cancer Res., 60, 50405044.
867871. 34. Pugh,S. et al. (1994) Patients with adenomatous polyps and carcinomas
5. Wood,L.D. et al. (2007) The genomic landscapes of human breast and have increased colonic mucosal prostaglandin E2. Gut, 35, 675678.
colorectal cancers. Science, 318, 11081113. 35. Rigas,B. et al. (1993) Altered eicosanoid levels in human colon cancer.
6. Reya,T. et al. (2005) Wnt signalling in stem cells and cancer. Nature, 434, J. Lab. Clin. Med., 122, 518523.
36. Yang,V.W. et al. (1998) Size-dependent increase in prostanoid levels in
843850.
adenomas of patients with familial adenomatous polyposis. Cancer Res.,
7. Sinicrope,F.A. et al. (2004) Role of cyclooxygenase-2 in colorectal can-
58, 17501753.
cer. Cancer Metastasis Rev., 23, 6375.
37. Kettunen,H.L. et al. (2003) Intestinal immune responses in wild-type and
8. Brown,J.R. et al. (2005) COX-2: a molecular target for colorectal cancer
Apcmin/ mouse, a model for colon cancer. Cancer Res., 63, 51365142.
prevention. J. Clin. Oncol., 23, 28402855. 38. Giardiello,F.M. et al. (2004) Prostanoids, ornithine decarboxylase, and
9. Vane,J.R. (1971) Inhibition of prostaglandin synthesis as a mechanism of polyamines in primary chemoprevention of familial adenomatous poly-
action for aspirin-like drugs. Nat. New Biol., 231, 232235. posis. Gastroenterology, 126, 425431.
10. Funk,C.D. (2001) Prostaglandins and leukotrienes: advances in eicosa- 39. Stolina,M. et al. (2000) Specific inhibition of cyclooxygenase 2 restores
noid biology. Science, 294, 18711875. antitumor reactivity by altering the balance of IL-10 and IL-12 synthesis.
11. Smith,W.L. et al. (2000) Cyclooxygenases: structural, cellular, and mo- J. Immunol., 164, 361370.
lecular biology. Annu. Rev. Biochem., 69, 145182. 40. Kawamori,T. et al. (2003) Enhancement of colon carcinogenesis by pros-
12. Cha,Y.I. et al. (2006) Fishing for prostanoids: deciphering the develop- taglandin E2 administration. Carcinogenesis, 24, 985990.
mental functions of cyclooxygenase-derived prostaglandins. Dev. Biol., 41. Wang,D. et al. (2005) Prostaglandin E2 enhances intestinal adenoma
289, 263272. growth via activation of the Ras-mitogen-activated protein kinase cascade.
13. Yokoyama,C. et al. (1988) Primary structure of sheep prostaglandin en- Cancer Res., 65, 18221829.
doperoxide synthase deduced from cDNA sequence. FEBS Lett., 231, 42. Chell,S. et al. (2006) Mediators of PGE2 synthesis and signalling down-
347351. stream of COX-2 represent potential targets for the prevention/treatment
14. Xie,W.L. et al. (1991) Expression of a mitogen-responsive gene encoding of colorectal cancer. Biochim. Biophys. Acta, 1766, 104119.
prostaglandin synthase is regulated by mRNA splicing. Proc. Natl Acad. 43. Elder,D.J. et al. (1997) Induction of apoptotic cell death in human co-
Sci. USA, 88, 26922696. lorectal carcinoma cell lines by a cyclooxygenase-2 (COX-2)-selective
15. Chandrasekharan,N.V. et al. (2002) COX-3, a cyclooxygenase-1 variant nonsteroidal anti-inflammatory drug: independence from COX-2 protein
inhibited by acetaminophen and other analgesic/antipyretic drugs: clon- expression. Clin. Cancer Res., 3, 16791683.
ing, structure, and expression. Proc. Natl Acad. Sci. USA, 99, 44. Hanif,R. et al. (1996) Effects of nonsteroidal anti-inflammatory drugs on
proliferation and on induction of apoptosis in colon cancer cells by a pros-
1392613931.
16. Snipes,J.A. et al. (2005) Cloning and characterization of cyclooxygenase- taglandin-independent pathway. Biochem. Pharmacol., 52, 237245.
45. Chan,T.A. et al. (1998) Mechanisms underlying nonsteroidal antiinflam-
1b (putative cyclooxygenase-3) in rat. J. Pharmacol. Exp. Ther., 313,
matory drug-mediated apoptosis. Proc. Natl Acad. Sci. USA, 95, 681686.
668676. 46. Paik,J.H. et al. (2000) Two opposing effects of non-steroidal anti-inflam-
17. Svensson,C.I. et al. (2002) The spinal phospholipase-cyclooxygenase- matory drugs on the expression of the inducible cyclooxygenase. Media-
prostanoid cascade in nociceptive processing. Annu. Rev. Pharmacol. tion through different signaling pathways. J. Biol. Chem., 275,
Toxicol., 42, 553583. 2817328179.
18. Adegboyega,P.A. et al. (2004) Immunohistochemical expression of cyclo- 47. Pang,L. et al. (2003) Cyclooxygenase-2 expression by nonsteroidal anti-
oxygenase-2 in normal kidneys. Appl. Immunohistochem. Mol. Morphol., inflammatory drugs in human airway smooth muscle cells: role of perox-
12, 7174. isome proliferator-activated receptors. J. Immunol., 170, 10431051.
19. Kirschenbaum,A. et al. (2000) Immunohistochemical localization of cy- 48. Wilson,J.W. et al. (2000) The effect of exogenous prostaglandin admin-
clooxygenase-1 and cyclooxygenase-2 in the human fetal and adult male istration on tumor size and yield in Min/ mice. Cancer Res., 60,
reproductive tracts. J. Clin. Endocrinol. Metab., 85, 34363441. 46454653.

384
The COX-2/PGE2 pathway

49. Bol,D.K. et al. (2002) Cyclooxygenase-2 overexpression in the skin of 77. Downward,J. (2003) Targeting RAS signalling pathways in cancer ther-
transgenic mice results in suppression of tumor development. Cancer apy. Nat. Rev. Cancer, 3, 1122.
Res., 62, 25162521. 78. Vivanco,I. et al. (2002) The phosphatidylinositol 3-Kinase AKT pathway
50. Yegnasubramanian,S. et al. (2004) Hypermethylation of CpG islands in in human cancer. Nat. Rev. Cancer, 2, 489501.
primary and metastatic human prostate cancer. Cancer Res., 64, 79. Bos,J.L. et al. (1987) Prevalence of ras gene mutations in human colorec-
19751986. tal cancers. Nature, 327, 293297.
51. Murata,H. et al. (2004) Promoter hypermethylation silences cyclooxyge- 80. Davies,H. et al. (2002) Mutations of the BRAF gene in human cancer.
nase-2 (Cox-2) and regulates growth of human hepatocellular carcinoma Nature, 417, 949954.
cells. Lab. Invest., 84, 10501059. 81. Samuels,Y. et al. (2004) High frequency of mutations of the PIK3CA gene
52. Wang,D. et al. (2004) Prostaglandin E(2) promotes colorectal adenoma in human cancers. Science, 304, 554.
growth via transactivation of the nuclear peroxisome proliferator-activated 82. Nassif,N.T. et al. (2004) PTEN mutations are common in sporadic micro-
receptor delta. Cancer Cell, 6, 285295. satellite stable colorectal cancer. Oncogene, 23, 617628.
53. Chulada,P.C. et al. (2000) Genetic disruption of Ptgs-1, as well as Ptgs-2, 83. Carpten,J.D. et al. (2007) A transforming mutation in the pleckstrin ho-
reduces intestinal tumorigenesis in Min mice. Cancer Res., 60, mology domain of AKT1 in cancer. Nature, 448, 439444.
47054708. 84. Siegel,P.M. et al. (2003) Cytostatic and apoptotic actions of TGF-beta in
54. Chell,S.D. et al. (2006) Increased EP4 receptor expression in colorectal homeostasis and cancer. Nat. Rev. Cancer, 3, 807821.
cancer progression promotes cell growth and anchorage independence. 85. Massague,J. (2008) TGFbeta in Cancer. Cell, 134, 215230.
Cancer Res., 66, 31063113. 86. Markowitz,S. et al. (1995) Inactivation of the type II TGF-beta receptor in
55. Kerr,J.F. et al. (1972) Apoptosis: a basic biological phenomenon with colon cancer cells with microsatellite instability. Science, 268,
wide-ranging implications in tissue kinetics. Br. J. Cancer, 26, 239257. 13361338.
56. Adams,J.M. (2003) Ways of dying: multiple pathways to apoptosis. Genes 87. Radtke,F. et al. (2005) Self-renewal and cancer of the gut: two sides of
Dev., 17, 24812495. a coin. Science, 307, 19041909.
57. Green,D.R. (1998) Apoptotic pathways: the roads to ruin. Cell, 94, 88. van de Wetering,M. et al. (2002) The beta-catenin/TCF-4 complex impo-
695698. ses a crypt progenitor phenotype on colorectal cancer cells. Cell, 111,
58. Thompson,C.B. (1995) Apoptosis in the pathogenesis and treatment of 241250.
disease. Science, 267, 14561462. 89. Castellone,M.D. et al. (2005) Prostaglandin E2 promotes colon cancer cell
59. Green,D.R. et al. (2002) A matter of life and death. Cancer Cell, 1, 1930. growth through a Gs-axin-beta-catenin signaling axis. Science, 310,
60. Pelengaris,S. et al. (2002) Suppression of Myc-induced apoptosis in beta 15041510.
cells exposes multiple oncogenic properties of Myc and triggers carcino- 90. Reya,T. et al. (2001) Stem cells, cancer, and cancer stem cells. Nature,
genic progression. Cell, 109, 321334. 414, 105111.
61. Danial,N.N. et al. (2004) Cell death: critical control points. Cell, 116, 91. Feinberg,A.P. et al. (2006) The epigenetic progenitor origin of human
205219. cancer. Nat. Rev. Genet, 7, 2133.
62. Datta,S.R. et al. (1997) Akt phosphorylation of BAD couples survival 92. Kinzler,K.W. et al. (1996) Lessons from hereditary colorectal cancer. Cell,
signals to the cell-intrinsic death machinery. Cell, 91, 231241. 87, 159170.
63. Bonni,A. et al. (1999) Cell survival promoted by the Ras-MAPK signaling 93. Korinek,V. et al. (1998) Depletion of epithelial stem-cell compartments in
pathway by transcription-dependent and -independent mechanisms. Sci- the small intestine of mice lacking Tcf-4. Nat. Genet., 19, 379383.
ence, 286, 13581362. 94. North,T.E. et al. (2007) Prostaglandin E2 regulates vertebrate haemato-
64. Tsujii,M. et al. (1995) Alterations in cellular adhesion and apoptosis in poietic stem cell homeostasis. Nature, 447, 10071011.
epithelial cells overexpressing prostaglandin endoperoxide synthase 2. 95. Liou,J.Y. et al. (2007) Cyclooxygenase-2-derived prostaglandin e2 pro-
Cell, 83, 493501. tects mouse embryonic stem cells from apoptosis. Stem Cells, 25,
65. Hague,A. et al. (1993) Sodium butyrate induces apoptosis in human co- 10961103.
lonic tumour cell lines in a p53-independent pathway: implications for the 96. Hanahan,D. et al. (1996) Patterns and emerging mechanisms of the an-
possible role of dietary fibre in the prevention of large-bowel cancer. Int. J. giogenic switch during tumorigenesis. Cell, 86, 353364.
Cancer, 55, 498505. 97. Tsujii,M. et al. (1998) Cyclooxygenase regulates angiogenesis induced by
66. Sheng,H. et al. (1998) Modulation of apoptosis and Bcl-2 expression by colon cancer cells. Cell, 93, 705716.
prostaglandin E2 in human colon cancer cells. Cancer Res., 58, 362366. 98. Jones,M.K. et al. (1999) Inhibition of angiogenesis by nonsteroidal anti-
67. Sheng,H. et al. (2001) Prostaglandin E2 increases growth and motility of inflammatory drugs: insight into mechanisms and implications for cancer
colorectal carcinoma cells. J. Biol. Chem., 276, 1807518081. growth and ulcer healing. Nat. Med., 5, 14181423.
68. Tessner,T.G. et al. (2004) Prostaglandin E2 reduces radiation-induced 99. Fukuda,R. et al. (2003) Vascular endothelial growth factor gene expres-
epithelial apoptosis through a mechanism involving AKT activation and sion in colon cancer cells exposed to prostaglandin E2 is mediated by
bax translocation. J. Clin. Invest., 114, 16761685. hypoxia-inducible factor 1. Cancer Res., 63, 23302334.
69. Pozzi,A. et al. (2004) Colon carcinoma cell growth is associated with 100. Williams,C.S. et al. (2000) Host cyclooxygenase-2 modulates carcinoma
prostaglandin E2/EP4 receptor-evoked ERK activation. J. Biol. Chem., growth. J. Clin. Invest., 105, 15891594.
279, 2979729804. 101. Sonoshita,M. et al. (2001) Acceleration of intestinal polyposis through
70. Leone,V. et al. (2007) PGE2 inhibits apoptosis in human adenocarcinoma prostaglandin receptor EP2 in Apc(Delta 716) knockout mice. Nat. Med.,
Caco-2 cell line through Ras-PI3K association and cAMP-dependent ki- 7, 10481051.
nase A activation. Am. J. Physiol. Gastrointest. Liver Physiol., 293, 102. Dormond,O. et al. (2001) NSAIDs inhibit alpha V beta 3 integrin-medi-
G673G681. ated and Cdc42/Rac-dependent endothelial-cell spreading, migration and
71. Pai,R. et al. (2002) Prostaglandin E2 transactivates EGF receptor: a novel angiogenesis. Nat. Med., 7, 10411047.
mechanism for promoting colon cancer growth and gastrointestinal hy- 103. Salcedo,R. et al. (2003) Angiogenic effects of prostaglandin E2 are me-
pertrophy. Nat. Med., 8, 289293. diated by up-regulation of CXCR4 on human microvascular endothelial
72. Buchanan,F.G. et al. (2003) Prostaglandin E2 regulates cell migration via cells. Blood, 102, 19661977.
the intracellular activation of the epidermal growth factor receptor. J. Biol. 104. Wang,D. et al. (2006) CXCL1 induced by prostaglandin E2 promotes
Chem., 278, 3545135457. angiogenesis in colorectal cancer. J. Exp. Med., 203, 941951.
73. He,T.C. et al. (1999) PPARdelta is an APC-regulated target of nonsteroi- 105. Weinberg,R.A. (2008) Mechanisms of malignant progression. Carcino-
dal anti-inflammatory drugs. Cell, 99, 335345. genesis, 29, 10921095.
74. Reed,K.R. et al. (2004) PPARdelta status and Apc-mediated tumourigen- 106. Fenwick,S.W. et al. (2003) The effect of the selective cyclooxygenase-2
esis in the mouse intestine. Oncogene, 23, 89928996. inhibitor rofecoxib on human colorectal cancer liver metastases. Gastro-
75. Kaidi,A. et al. (2006) Direct transcriptional up-regulation of cyclooxyge- enterology, 125, 716729.
nase-2 by hypoxia-inducible factor (HIF)-1 promotes colorectal tumor 107. Yao,M. et al. (2003) Inhibition of cyclooxygenase-2 by rofecoxib attenu-
cell survival and enhances HIF-1 transcriptional activity during hypoxia. ates the growth and metastatic potential of colorectal carcinoma in mice.
Cancer Res., 66, 66836691. Cancer Res., 63, 586592.
76. Hawcroft,G. et al. (2007) Prostaglandin E2-EP4 receptor signalling pro- 108. Tsujii,M. et al. (1997) Cyclooxygenase-2 expression in human colon
motes tumorigenic behaviour of HT-29 human colorectal cancer cells. cancer cells increases metastatic potential. Proc. Natl Acad. Sci. USA,
Oncogene, 26, 30063019. 94, 33363340.

385
A.Greenhough et al.

109. Pai,R. et al. (2003) Prostaglandins promote colon cancer cell invasion; 138. Holla,V. et al. (2008) Regulation of prostaglandin transporters in colorec-
signaling by cross-talk between two distinct growth factor receptors. tal neoplasia. Cancer Prev. Res., 1, 9399.
FASEB J., 17, 16401647. 139. Backlund,M.G. et al. (2008) Repression of 15-hydroxyprostaglandin de-
110. Birchmeier,C. et al. (2003) Met, metastasis, motility and more. Nat. Rev. hydrogenase involves histone deacetylase 2 and snail in colorectal cancer.
Mol. Cell Biol., 4, 915925. Cancer Res., 68, 93319337.
111. Qualtrough,D. et al. (2007) Prostaglandin F(2alpha) stimulates motility 140. Subbaramaiah,K. et al. (2003) Cyclooxygenase 2: a molecular target for
and invasion in colorectal tumor cells. Int. J. Cancer, 121, 734740. cancer prevention and treatment. Trends Pharmacol. Sci., 24, 96102.
112. Gupta,G.P. et al. (2007) Mediators of vascular remodelling co-opted for 141. Howe,L.R. et al. (2001) PEA3 is up-regulated in response to Wnt1 and
sequential steps in lung metastasis. Nature, 446, 765770. activates the expression of cyclooxygenase-2. J. Biol. Chem., 276,
113. Hull,M.A. et al. (2006) Regulation of stromal cell cyclooxygenase-2 in the 2010820115.
ApcMin/ mouse model of intestinal tumorigenesis. Carcinogenesis, 27, 142. Howe,L.R. et al. (1999) Transcriptional activation of cyclooxygenase-2 in
382391. Wnt-1-transformed mouse mammary epithelial cells. Cancer Res., 59,
114. Christofori,G. (2007) Cancer: division of labour. Nature, 446, 735736. 15721577.
115. Kroemer,G. et al. (2008) Tumor cell metabolism: cancers Achilles heel. 143. Araki,Y. et al. (2003) Regulation of cyclooxygenase-2 expression by the
Cancer Cell, 13, 472482. Wnt and ras pathways. Cancer Res., 63, 728734.
116. Palucka,A.K. et al. (2007) Taming cancer by inducing immunity via den- 144. Coffey,R.J. et al. (1997) Epidermal growth factor receptor activation in-
dritic cells. Immunol. Rev., 220, 129150. duces nuclear targeting of cyclooxygenase-2, basolateral release of pros-
117. Zou,W. (2005) Immunosuppressive networks in the tumour environment taglandins, and mitogenesis in polarizing colon cancer cells. Proc. Natl
and their therapeutic relevance. Nat. Rev. Cancer, 5, 263274. Acad. Sci. USA, 94, 657662.
118. Harris,S.G. et al. (2002) Prostaglandins as modulators of immunity. 145. Sheng,H. et al. (1997) Cyclooxygenase-2 induction and transforming
Trends Immunol., 23, 144150. growth factor beta growth inhibition in rat intestinal epithelial cells. Cell
119. Harizi,H. et al. (2005) The impact of eicosanoids on the crosstalk between Growth Differ., 8, 463470.
innate and adaptive immunity: the key roles of dendritic cells. Tissue 146. Jones,M.K. et al. (1999) HGF triggers activation of the COX-2 gene in rat
Antigens, 65, 507514. gastric epithelial cells: action mediated through the ERK2 signaling path-
120. Ahmadi,M. et al. (2008) Prevention of both direct and cross-priming of way. FASEB J., 13, 21862194.
antitumor CD8 T-cell responses following overproduction of prostaglan- 147. Guo,Y.S. et al. (2002) Gastrin stimulates cyclooxygenase-2 expression in
din E2 by tumor cells in vivo. Cancer Res., 68, 75207529. intestinal epithelial cells through multiple signaling pathways. Evidence
121. Park,J.Y. et al. (2006) Prostaglandin E2 synthesis and secretion: the role of for involvement of ERK5 kinase and transactivation of the epidermal
PGE2 synthases. Clin. Immunol., 119, 229240.
growth factor receptor. J. Biol. Chem., 277, 4875548763.
122. Nomura,T. et al. (2004) The two-step model of prostaglandin signal ter-
148. Csiki,I. et al. (2006) Thioredoxin-1 modulates transcription of cycloox-
mination: in vitro reconstitution with the prostaglandin transporter and
ygenase-2 via hypoxia-inducible factor-1alpha in non-small cell lung can-
prostaglandin 15 dehydrogenase. Mol. Pharmacol., 65, 973978.
cer. Cancer Res., 66, 143150.
123. Hull,M.A. et al. (2004) Prostaglandin EP receptors: targets for treatment
149. Schmedtje,J.F. Jr, et al. (1997) Hypoxia induces cyclooxygenase-2 via the
and prevention of colorectal cancer? Mol. Cancer Ther., 3, 10311039.
NF-kappaB p65 transcription factor in human vascular endothelial cells.
124. Yoshimatsu,K. et al. (2001) Inducible microsomal prostaglandin E syn-
thase is overexpressed in colorectal adenomas and cancer. Clin. Cancer J. Biol. Chem., 272, 601608.
Res., 7, 39713976. 150. Demasi,M. et al. (2003) Effects of hypoxia on monocyte inflammatory
125. Nakanishi,M. et al. (2008) Genetic deletion of mPGES-1 suppresses in- mediator production: dissociation between changes in cyclooxygenase-2
testinal tumorigenesis. Cancer Res., 68, 32513259. expression and eicosanoid synthesis. J. Biol. Chem., 278, 3860738616.
126. Elander,N. et al. (2008) Genetic deletion of mPGES-1 accelerates intes- 151. Bonazzi,A. et al. (2000) Regulation of cyclooxygenase-2 by hypoxia and
tinal tumorigenesis in APC(Min/) mice. Biochem. Biophys. Res. Com- peroxisome proliferators in the corneal epithelium. J. Biol. Chem., 275,
mun., 372, 249253. 28372844.
127. Mutoh,M. et al. (2002) Involvement of prostaglandin E receptor subtype 152. Vaupel,P. et al. (2004) Tumor hypoxia and malignant progression. Meth-
EP(4) in colon carcinogenesis. Cancer Res., 62, 2832. ods Enzymol., 381, 335354.
128. Amano,H. et al. (2003) Host prostaglandin E(2)-EP3 signaling regulates 153. Shimizu,S. et al. (1995) Prevention of hypoxia-induced cell death by Bcl-2
tumor-associated angiogenesis and tumor growth. J. Exp. Med., 197, and Bcl-xL. Nature, 374, 811813.
221232. 154. Koong,A.C. et al. (1994) Hypoxic activation of nuclear factor-kappa B is
129. Shoji,Y. et al. (2004) Downregulation of prostaglandin E receptor subtype mediated by a Ras and Raf signaling pathway and does not involve MAP
EP3 during colon cancer development. Gut, 53, 11511158. kinase (ERK1 or ERK2). Cancer Res., 54, 52735279.
130. Fulton,A.M. et al. (2006) Targeting prostaglandin E EP receptors to in- 155. Poligone,B. et al. (2001) Positive and negative regulation of NF-kappaB by
hibit metastasis. Cancer Res., 66, 97949797. COX-2: roles of different prostaglandins. J. Biol. Chem., 276, 3865838664.
131. Yang,L. et al. (2006) Host and direct antitumor effects and profound re- 156. Liston,P. et al. (1996) Suppression of apoptosis in mammalian cells by
duction in tumor metastasis with selective EP4 receptor antagonism. Can- NAIP and a related family of IAP genes. Nature, 379, 349353.
cer Res., 66, 96659672. 157. Dong,Z. et al. (2001) Up-regulation of apoptosis inhibitory protein IAP-2
132. Schuster,V.L. (2002) Prostaglandin transport. Prostaglandins Other Lipid by hypoxia. Hif-1-independent mechanisms. J. Biol. Chem., 276,
Mediat., 6869, 633647. 1870218709.
133. Ensor,C.M. et al. (1995) 15-Hydroxyprostaglandin dehydrogenase. 158. Staller,P. et al. (2003) Chemokine receptor CXCR4 downregulated by von
J. Lipid Mediat. Cell Signal., 12, 313319. Hippel-Lindau tumour suppressor pVHL. Nature, 425, 307311.
134. Yan,M. et al. (2004) 15-Hydroxyprostaglandin dehydrogenase, a COX-2 159. Ding,M. et al. (2006) Loss of the tumor suppressor Vhlh leads to upregu-
oncogene antagonist, is a TGF-beta-induced suppressor of human gastro- lation of Cxcr4 and rapidly progressive glomerulonephritis in mice. Nat.
intestinal cancers. Proc. Natl Acad. Sci. USA, 101, 1746817473. Med., 12, 10811087.
135. Backlund,M.G. et al. (2005) 15-Hydroxyprostaglandin dehydrogenase is 160. Kaidi,A. et al. (2007) Interaction between beta-catenin and HIF-1 pro-
down-regulated in colorectal cancer. J. Biol. Chem., 280, 32173223. motes cellular adaptation to hypoxia. Nat. Cell Biol., 9, 210217.
136. Mann,J.R. et al. (2006) Repression of prostaglandin dehydrogenase by 161. Bresalier,R.S. et al. (2005) Cardiovascular events associated with rofe-
epidermal growth factor and snail increases prostaglandin E2 and pro- coxib in a colorectal adenoma chemoprevention trial. N. Engl. J. Med.,
motes cancer progression. Cancer Res., 66, 66496656. 352, 10921102.
137. Myung,S.J. et al. (2006) 15-Hydroxyprostaglandin dehydrogenase is an
in vivo suppressor of colon tumorigenesis. Proc. Natl Acad. Sci. USA, 103, Received September 8, 2008; revised December 4, 2008;
1209812102. accepted January 4, 2009

386

Vous aimerez peut-être aussi