You are on page 1of 15

Current Pharmaceutical Design, 2003, 9, 2177-2190 2177

Cyclooxygenase-2 Biology

Joan Clària*

DNA Unit, Institut d´Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de
Barcelona, Barcelona 08036, Spain

Abstract: In mammalian cells, eicosanoid biosynthesis is usually initiated by the activation of phospholipase A 2
and the release of arachidonic acid from membrane phospholipids in response to the interaction of a stimulus with a
receptor on the cell surface. Arachidonic acid is subsequently transformed by the enzyme cyclooxygenase (COX) to
prostaglandins (PGs) and thromboxane (TX). The COX pathway is of particular clinical relevance because it is the
major target for non-steroidal anti-inflammatory drugs, which are commonly used for relieving inflammation, pain
and fever. In 1991, it was disclosed that COX exists in two distinct isozymes (COX-1 and COX-2), one of which,
COX-2, is primarily responsible for inflammation but apparently not for gastrointestinal integrity or platelet
aggregation. For this reason, in recent years, novel compounds that are selective for this isozyme, the so-called
selective COX-2 inhibitors or COXIBs, which retain anti-inflammatory activity but minimize the risk of
gastrointestinal toxicity and bleeding, have been developed. This review article provides an overview and an
update on the progress achieved in the area of COX-2 and PG biosynthesis and describes the role of COX-2 in health
and disease. It also discusses some unresolved issues related to the use of selective COX-2 inhibitors as a safe and
promising therapeutic option not only for the treatment of inflammatory states but also for cancer.
Key Words: Eicosanoids, prostaglandins, cyclooxygenase-2, non-steroidal anti-inflammatory drugs, cyclooxygenase-2
inhibitors, gastrointestinal tract.

PROSTAGLANDINS aggregatory and vasoconstrictor molecule, thromboxane (TX)


A2, whose structure and biosynthesis was elucidated soon
The history of prostaglandins (PGs) date back in the early after the discovery of PGs [4].
1930´s when Goldblatt and Von Euler who independently
discovered a fatty acid in human seminal fluid with potent PROSTAGLANDIN BIOSYNTHESIS
vasoactive properties in rabbits and guinea pigs [1, 2]. von
Euler named this compound prostaglandin because he In 1971, Vane [5], Ferreira et al. [6] and Smith et al. [7]
assumed it was originated in the prostate gland (it is now published their seminal observations proposing that the
known to be originated in the seminal vesicles) and suggested ability of aspirin-like drugs to suppress inflammation rests
that prostaglandins played a role in sperm transport [2]. primarily on their ability to inhibit the production of PGs.
Thirty years later, Bergstrom and Samuelsson established the This work fostered further research with focus on the
structure of the first two PGs which were termed PGE and elucidation of the pathway leading to the biosynthesis of
PGF because of their respective partitions into ether and PGs in animal cells, which concluded that the synthesis of
phosphate buffer (fosfat in Swedish) and demonstrated that PGs from arachidonic acid is initiated by the enzyme
they were produced from the essential fatty acid arachidonic cyclooxygenase (COX). Although in human cells there are at
acid [3]. Today the nomenclature of PGs describes ten least two different COX isozymes designated COX-1 and
specific molecular groups, designated by the letters A COX-2 (as discussed below a third COX isoform termed
through J, that are unique by their variation in the functional COX-3 has recently been identified), the basic catalytic
groups attached to positions 9 and 11 of the cyclopentane activities of these isozymes are similar enough to treat them
ring [4]. Each group of PGs consists of three series design- as if they were biochemically identical. Accordingly, the
ated by the subscript numbers 1, 2 and 3, depending on the biochemistry of these COX isozymes is herewith discussed
substrate originated: di-homo-γ -linoleic acid, arachidonic as one. Moreover, although other names for COX are PG
acid, and eicosapentaenoic acid, respectively. Among the endoperoxide synthase, PG G/H synthase and PGH synthase,
PGs, the most widely distributed and best characterized are in this review article this enzyme is referred to as COX.
those derived from arachidonic acid (i.e., PGE2, PGD 2, PGI 2 COX is a membrane-bound byfunctional enzyme that
and PGF 2α). For PGF, the additional subscript α or β catalyzes the first two committed steps in the pathway
denotes the spatial configuration of the carbon 9 hydroxyl leading to the formation of PGs and TX, namely
group. Special mention should be given to the platelet pro- cyclooxygenation and peroxidation. In the first step, COX
cyclizes and adds two molecules of O2 to arachidonic acid to
*Address correspondence to this author at the DNA Unit, Hospital Clínic, form the cyclic hydroperoxide PGG2. Subsequently, COX
Villarroel 170, Barcelona 08036, Spain; Tel: 34-93-2275400 ext. 2814; reduces PGG2 to PGH 2 [8, 9]. PGH 2 is a highly unstable
Fax: 34-93-2275454; E-mail: jclaria@clinic.ub.es endoperoxide, which functions as an intermediate substrate

1381-6128/03 $41.00+.00 © 2003 Bentham Science Publishers Ltd.


2178 Current Pharmaceutical Design, 2003, Vol. 9, No. 27 Joan Clària

for the biosynthesis, by specific synthases and isomerases, of COX is an integral membrane protein found mainly in
PGs of the E2, F 2 and D2 series and also of PGI2 microsomal membranes. Though confocal fluorescence imag-
(prostacyclin) and TXA2 [4]. Interestingly, the coupling of ing microscopy and histofluorescence staining techniques
PGH2 synthesis to its transformation to PGs and TX by reveal that COX-1 and COX-2 are located in the endoplas-
downstream enzymes is intricately orchestrated in a cell- mic reticulum and nuclear envelope, COX-2 is more highly
specific fashion. That is, any given prostanoid-forming cell concentrated in the nuclear envelope [15]. It is unclear why
tends to form only one of these compounds as its major COX is associated with several different membrane systems
product. Thus, for example, in brain and mast cells, PGH2 is but one possibility may be that PG synthesis at different
converted to PGD2 by cytosolic enzyme PGD synthase. subcellular sites is elicited by different stimuli. In any case,
PGH2 can alternatively be converted to PGF 2α by PGF the aspirin acetylation site, the site of trypsin cleavage and
synthase, which is mainly expressed in the uterus. Vascular the major antigenic determinants of COX are on the cyto-
endothelial cells produce PGI2 or prostacyclin from PGH2 by plasmic side of the endoplasmic reticulum indicating that
means of the PGI or prostacyclin synthase, and platelets PGH2 is generated intracellularly rather than extracellularly.
release TXA2 from the same precursor (PGH 2) as the PGs Most of the synthases, which further metabolize PGH2 also
through the action of the enzyme TX synthase. Both PGI2 appear to be localized on the endoplasmic reticulum.
and TXA2, have a very short half-life (30 seconds and 3
minutes, respectively) and are rapidly hydrolyzed to the PROSTAGLANDIN RECEPTORS
inactive compounds TXB2 and 6-keto-PGF1α, respectively. Similar to what occurs with the PGH2 downstream
Finally, PGE2 is formed in many cell types by the enzyme metabolizing enzymes (i.e., PG synthases and isomerases),
PGE synthase. A schematic representation of these pathways prostanoid receptors are also cell and/or tissue specific. There
is shown in Fig. (1). It is important to note that PGE are at least 9 known PG receptors as well as several of their
synthase is also present in an inducible form (mPGE splice variants that belong to a subfamily of the G protein-
synthase), which is membrane-bound, glutathione-dependent coupled receptor (GPCR) superfamily of seven transmem-
and whose expression is induced by proinflammatory brane spanning proteins [16]. Four of the receptor subtypes
compounds, such as interleukin-1β [10, 11]. The coordinate bind PGE 2 (EP 1-EP4), two bind PGD2 (DP 1 and DP 2) and the
induction of multiple enzymes of the prostanoid pathway, rest are single receptors for PGF2α, PGI 2 and TXA2 (FP, IP
such as COX-2 and mPGE synthase, during the inflam- and TP, respectively) [16-18]. IP, DP1, EP2 and EP 4
matory response is a concept that is currently evolving. receptors are coupled with the activation of Gs proteins and
Both cyclooxygenase and peroxidase activities of COX linked to increases in intracellular cAMP, whereas EP1, FP
are associated with a single protein molecule and use a single and TP signal through Gq-mediated increases in intracellular
heme moiety within the enzyme [9]. However, the active calcium concentration. Exceptionally, the EP 3 receptor is
sites for the cyclooxygenase and peroxidase activities of coupled to Gi and decreases cAMP formation. Interestingly,
COX have been shown to be distinct and independent in in addition to signaling through G-protein linked receptors,
pharmacological and in vitro mutagenesis studies [9]. Hence, PGs generated from COX-2 located in the nuclear envelope
the two active sites for cyclooxygenase and peroxidase may mediate signals preferentially through a nuclear
probably overlap [9]. The cyclooxygenase active site is pathway, whereas COX-1 derived PGs may mediate signals
proposed to be a long hydrophobic channel, which extends solely through cell surface receptors [16]. This emphasizes
from the membrane binding domain to near the heme group. the importance of COX-2 and nuclear receptors (i.e.,
This hydrophobic channel has been shown to bind peroxisome proliferator-activating receptors, PPARs) in cell
arachidonate and also other fatty acids as substrates. Thus, in growth and survival.
addition to the 20-carbon arachidonate (20:4 ω 6), COX also
uses di-homo-γ -linolenate (20:3 ω 6), 5,8,11,14,17-eicosa- COX ISOZYMES: COX-1, COX-2 AND NOW COX-3
pentanoic acid (EPA), γ -linolenic acid, α-linolenic acid and
The understanding that NSAIDs block prostanoid
linolenic acid. EPA is subsequently converted to PGH3
formation via inhibition of COX explained the anti-
whereas the 18-carbon fatty acids are converted into mono-
inflammatory effects of these drugs. Similarly, prostanoids,
hydroxy acids. Although COX-1 and COX-2 oxygenate
such as PGE2 and prostacyclin, were found to be protective
arachidonate with almost identical kinetics, in general, COX-
to the stomach and, therefore, inhibition of their formation
2 is much more efficient with alternative substrates. For
provided an explanation for the gastrointestinal toxicity
instance, COX-2 utilizes both fatty acids and endocanna-
associated with prolonged use of NSAIDs (see later in this
binoids, including 2-archidonylglycerol and anandamide, as
review). On the other hand, inhibition of COX in platelets
substrates. COX-2 action on 2-archidonylglycerol and
explained the ability of aspirin to reduce platelet aggregation
anandamide generates endocannabinoid-derived prostanoids,
and blood clotting. But still a number of questions remained
among them PG glycerol esters and ethanolamides [12]. In
unanswered; such as why the toxicity of NSAIDs differs in
addition, aspirin-acetylated COX-2 converts arachidonic acid
the gastrointestinal tract when used at similar anti-
to 15R-hydroxyeicosatetraenoic acid (15R-HETE), which is
inflammatory doses? The answer was provided in the early
further transformed into 15-epi-lipoxins [13]. Moreover,
1990´s when it was established that there are at least two
when exposed to aspirin, COX-2-expressing cells enzymati-
COX isozymes present in human cells: COX-1, which is
cally transform omega-3 docosaexaenoic acid (DHA) to
constitutively expressed; and COX-2, which is inducible
previously unrecognized compounds, i.e., a novel 17R series
[19-21].
of hydroxy-DHAs [14]. Both 15-epi-lipoxins and hydroxy-
DHAs bear potent anti-inflammatory properties and play a COX-1 and COX-2 are the products of two distinct
key role in the resolution of inflammation. genes, which in humans are localized on chromosomes 9 and
Cyclooxygenase-2 Biology Current Pharmaceutical Design, 2003, Vol. 9, No. 27 2179

Phospholipids

COOH

Arachidonic Acid

O COOH
O

COX-1 OOH
PGG2
COX-2

COX-3

O COOH
O

OH
ynth ase PGH2
TX S PGI S
yntha
se
O
COOH
O
O
PGD

OH
TXA2 HO
Syn

OH
e
has

thas

PGI2
ynt

OH
e

COOH
ES

PGF Synthase

O
PG

COOH
HO
HO O
OH
TXB2 HO
HO OH
O COOH 6-Keto-PGF1α
COOH

HO OH O OH
PGE2
PGD2
HO

COOH

HO OH
PGF2α
Fig. (1). Enzymatic pathway of prostaglandin (PG) formation from arachidonic acid. Cyclooxygenase (COX), which is present in three
different isozymes COX-1, 2 and 3, oxygenates arachidonic acid to form PGG 2 which is further reduced to PGH 2. PGH2 is a highly
unstable endoperoxide that is rapidly converted by specific synthases to PGs of the E2, F2 and D2 series and also to PGI2
(prostacyclin) and thromboxane (TX) A2. Both PGI 2 and TXA2 have a very short half-life (30 seconds and 3 minutes, respectively) and
are rapidly hydrolyzed to the inactive compounds 6-keto-PGF1α and TXB2, respectively.

1, respectively [22, 23]. The COX-1 gene is about 22 kb in has revealed several potential transcription regulatory
length with 11 exons and is transcribed as a 2.8 kb mRNA elements, including a TATA box, a NF-IL-6 motif, two AP-
[22, 24]. The COX-2 gene is about 8 kb long with 10 exons 2 sites, three Sp1 sites, two NF-kB sites a CRE motif and
and it is transcribed as 4.6, 4.0 and 2.8 kb mRNAs variants an E-box [28].
[25-27]. Sequence analysis of the COX-2 5´-flanking region
2180 Current Pharmaceutical Design, 2003, Vol. 9, No. 27 Joan Clària

The amino acid sequences of COX-1 and COX-2 from a immediate-early genes, it can be rapidly upregulated in
single species are about 60% identical. COX-1 is a response to growth factors and cytokines [28]. This has led
glycoprotein that in its native, processed form has 576 to the supposition that the inducible COX-2 isoform is
amino acids with an apparent molecular mass of 70 kDa [22, responsible for the synthesis of PGs involved in
24]. The cDNA for COX-2 encodes a polypeptide that, inflammatory response, whereas COX-1-derived PGs are
before cleavage of the signal sequence, contains 604 amino involved in preserving the physiological functions of these
acids with an apparent molecular mass of 70 kDa [25, 26]. prostanoids. However, this distinction is not entirely
The main difference is that the COX-1 protein contains a 17 accurate, since COX-1 can be induced or upregulated under
amino acid sequence near its amino terminus that is not certain conditions and COX-2 has been consistently shown
present in COX-2. In contrast, COX-2 contains a 18 amino to be constitutively expressed in organs, such as the brain
acid sequence near its carboxyl terminus that is not present and the kidneys (see later in this review).
in COX-1 [22, 24-26]. However, and as already mentioned, Although the discovery of COX-2 was a great
the two COX isoforms catalyze identical reactions and advancement in our understanding of COX biology, it did
exhibit the same kinetic constants for the conversion of not appear to explain everything. In particular, the
arachidonic acid to prostanoids. mechanism of action of acetaminophen remained a mystery.
The most striking distinctions between COX-1 and Hugely popular and with a history almost as venerable as
COX-2 are the differential regulation of their expression and that of aspirin, this drug exerts antipyretic and analgesic
their tissue distribution (Fig. 2). COX-1 is ubiquitous and is effects without displaying anti-inflammatory activity.
constitutively expressed throughout the gastrointestinal Moreover, at therapeutic concentrations, acetaminophen only
system, the kidneys, the vascular smooth muscle and weakly inhibits COX-1 and COX-2. Willoughby first
platelets [28]. COX-1 is presumably involved in the suggested that this is because of the presence of another
housekeeping functions of PGs, such as the cytoprotective COX isozyme, COX-3 [29]. In fact, Dan Simmons´s group
effects in the gastric mucosa, the integrity of platelet has recently demonstrated the existence of a new COX that is
function and the maintenance of renal perfusion. Conversely, especially sensitive to acetaminophen and related compounds
COX-2 is undetectable in most tissues, but its expression (Fig. 2) [30]. This novel COX, termed COX-3, is a splice
can be induced by a variety of stimuli related to variant of COX-1 and in human tissues its mRNA is
inflammatory response. COX-2 is, therefore, commonly expressed as an 5.2-kb transcript, which is most abundant in
referred to as the inducible COX isoform because, like other cerebral cortex and heart [30].

Fig. (2). Proposed separate functions for prostaglandins (PGs) derived from cyclooxygenase (COX)-1, -2 and -3. While COX-1 is
found in most tissues and is believed to participate in physiologic homeostasis in the stomach, kidneys and platelets, COX-2 is
preferentially expressed in cells that mediate inflammation and in the central nervous system. Therefore, inhibition of COX-2 is
supposed to be responsible for the beneficial actions of non-steroidal anti-inflammatory drugs (NSAIDs), whereas inhibition of COX-
1 accounts for the unwanted side-effects of these drugs. A splice variant of COX-1, termed COX-3, that is especially sensitive to
acetaminophen and may explain the antipyretic and analgesic effects of this drug, has been recently identified.
Cyclooxygenase-2 Biology Current Pharmaceutical Design, 2003, Vol. 9, No. 27 2181

INHIBITION OF THE COX PATHWAY: NSAIDS The discovery of the COX-2 isozyme and the
AND COXIBS characterization of its role in inflammation fostered the
development of a new class of compounds that selectively
COX is the main pharmacological target for NSAIDs. inhibit COX-2, without affecting the COX-1-dependent PG
The observation that NSAIDs reduce or prevent the biosynthesis necessary for physiological functions [38-41].
production of PGs by direct inhibition of COX enzymes was This new generation of anti-inflammatory drugs has been
first reported by Vane, Ferreira et al. and Smith et al. in proven in vitro to selectively inhibit COX-2 activity and to be
1971 [5-7]. At present, NSAIDs are among the most widely as efficacious as standard NSAIDs in a number of in vivo
prescribed class of pharmaceutical agents worldwide, having models of inflammation (rat carrageenan-induced foot pad
broad clinical utility in treating pain, fever and inflammation edema and rat adjuvant-induced arthritis) and hyperalgesia (rat
[31, 32]. The most popular NSAID, aspirin, is also sought carrageenan-induced hyperalgesia) [40, 41]. Two COXIBS
as a potentially viable option in the prevention of sporadic (selective COX-2 inhibitors), celecoxib (Celebrex) and
colon cancer and neurodegenerative disorders [33, 34]. rofecoxib (Vioxx) have been marketed in the past three years,
Unfortunately and apart from the beneficial anti- and in clinical trials have proven to provide significant relief of
inflammatory, antipyretic and analgesic effects of NSAIDs, the signs and symptoms of osteoarthritis and rheumatoid
COX inhibition also results in unwanted side effects, arthritis and in alleviating pain following dental extraction,
particularly in the gastrointestinal tract [35]. Gastroduodenal while reducing the incidence of gastrointestinal ulcers and
ulceration is the best-characterized serious adverse event of erosions seen with standard NSAID therapy [42-47].
NSAID therapy and is the consequence of inhibiting PGs, Moreover, these eagerly awaited highly selective COX-2
which are the most important gastric cytoprotective agents inhibitors are of great interest because they may represent an
[36]. On the other hand although the incidence of renal side alternative therapeutic option for the treatment of inflamma-
effects in healthy subjects is not significant, adverse renal tion in diseases, such as in cirrhosis with ascites, in which
events are frequent in those patients with impaired effective renal function is critically dependent on PGs [48, 49]. A
arterial blood volume in which renal function is critically second-generation of selective COX-2 inhibitors (i.e.,
dependent on PGs, such as decompensated liver cirrhosis valdecoxib and etoricoxib) with a higher COX-1 to COX-2
[37]. Since COX-1-derived PGs are presumably involved in selectivity ratio than celecoxib and rofecoxib is currently
housekeeping functions, such as gastrointestinal cytoprotec- under evaluation in patients with osteoarthritis and rheumatoid
tion, and COX-2-derived PGs are implicated in inflammation, arthritis. Furthermore, the analgesic efficacy and tolerability of
NSAID gastrotoxicity is considered to be the consequence of parecoxib, an injectable prodrug of valdecoxib, is currently
the inhibition of both COX-1 and COX-2 isozymes by being tested in postoperative laparotomy and orthopedic knee
traditional NSAIDs (i.e., aspirin, indomethacin, ibuprofen, surgery patients [50, 51]. A list of currently available
meclofenamate, …). That is, at the concentrations required to NSAIDs and COXIBs is provided in Table 1.
inhibit PG biosynthesis at sites of inflammation (COX-2
activity), they also elicit a marked suppression of PG COX-2 IN HEALTH AND DISEASE
production in the gastrointestinal and renal systems (COX-1
Activation of prostanoid receptors triggers an astonishing
activity) jeopardizing the integrity of the gastric mucosa and
array of biological effects (Table 2). The most important
renal and platelet function.
targets of PGs include the gastrointestinal tract, the

Table 1. Classification of Non-Steroidal Anti-Inflammatory Drugs According to their Selectivity Towards COX-2. Brand
Names are Given in Brackets

Traditional NSAIDs Preferential COX-2 inhibitors Selective COX-2 inhibitors

Aspirin Etodolac ( Lodine) Celecoxib ( Celebrex)

Diclofenac (Arthrotec, Cataflam, Voltaren) Meloxicam ( Mobic) Etoricoxib (Arcoxia)

Flurbiprofen (Ansaid) Nabumetone (6-MNA) (Relafen) Parecoxib (Dynastat)

Ibuprofen (Advil, Motrin, Nuprin, Others) Nimesulide (Mesulid, Nexen) Rofecoxib (Vioxx)

Indomethacin (Indocin, Inacid, Others) Valdecoxib (Bextra)

Ketoprofen (Orudis, Oruvail)

Ketorolac (Acular, Toradol)

Naproxen (Aleve, Naprelan, Naprosyn )

Piroxicam (Feldene)

Sulindac (Clinoril)

Tolmetin (Tolectin)
2182 Current Pharmaceutical Design, 2003, Vol. 9, No. 27 Joan Clària

Table 2. Biological Effects of Cyclooxygenase Products

Tissue/Organ Mediators Effects

Female Reproductive Organs PGE2 , PGF2 α Uterine Contraction, Oxytocic Action

Male Reproductive Organs PGE2 , PGF2 α Fertility

Cardiovascular System TXA 2 , PGI2 Thrombosis, Platelet Aggregation


TXA 2 Vascular Permeability
PGE2 , PGI2 Arterial Vasodilation
TXA 2 , PGF2 α Venous vasoconstriction
PGE2 , PGI2 Patency of the Fetal Ductus Arteriosus

Respiratory System PGE2 Bronchodilation


PGF2 α , TXA 2 Bronchoconstriction

Renal System PGE2 , PGI2 Regulation Renal Blood Flow and Glomerular Filtration Rate
PGE2 , PGI2 Renin Release
PGE2 Inhibition Hydroosmotic Effect of ADH

Gastrointestinal System PGE2 , PGI2 Cytoprotection

Immune System PGE2 , PGI2 Inhibition T and B lymphocyte activation and proliferation

Central Nervous System PGE2 Fever


PGD2 Sleep
PGE2 , PGI2 Pain

cardiovascular system, the central and peripheral nervous Recently, COX-2 has been reported to be induced in
systems, the renal system and the reproductive organs. A human gastric mucosa infected with Helicobacter pylori [58,
detailed discussion of PG actions and the role of COX-2 59]. Moreover, the production of PGE2 in gastric mucosa is
under physiologic and pathologic conditions is herewith increased in patients with gastritis and infection with H.
provided. pylori stimulates synthesis of PGE2 in gastric mucosal cells
in vitro [58, 60]. While H. pylori infection is considered to
COX-2 and the Gastrointestinal Tract
increase acid secretion in duodenal ulcer patients, in severe
Gastric Secretion gastritis, acid secretion is generally decreased and basal
In human gastric mucosa, PGE 2 and PGF 2α have been gastrin levels are assumed to be stimulated. In particular,
reported as the most abundant COX-derived products, with acid secretion was significantly reduced in an experimental
low production of PGD 2 and PGI2 [52]. In 1967, initial group of mice with H. pylori infection, an effect that was
observations reported PGE2 to be anti-secretory and anti- restored to the same level as in the control mice following
ulcerogenic [53]. Subsequently, it was demonstrated that COX-2 inhibition [61].
intravenous administration of PGE1 or PGE 2 inhibits acid
Gastrointestinal Motility
and pepsin secretion induced by secretagogues, such as
pentagrastin and histamine [54]. Moreover, PGE2 has been Essential to digestion, gastrointestinal motility is a
shown to inhibit gastric parietal cells and to increase complex process regulated by both neural and humoral
bicarbonate secretion, resulting in decreased acidity of gastric components. Paracrine modulation of normal intestinal
juice [55]. It must be taken into account that these motility by eicosanoids such as PGs is still a matter of
observations on gastric acid secretion were obtained by debate, but pharmacological doses of certain prostanoids
administering PGs at pharmacological doses and that have been shown to alter motility both in vivo and in vitro
establishing the role of endogenously produced PGs in [62]. Vascular reactivity studies in isolated intestinal muscle
gastric acid secretion has proven to be more challenging. In show that PGE2 induces contraction of longitudinal muscle
this regard, blockade of PG biosynthesis with COX and relaxation of circular muscle, whereas PGF 2α induces
inhibitors has yielded disparate and non-conclusive results contraction of both muscles [63]. Isolated colonic
[56]. Moreover, although studies in experimental models longitudinal and circular muscle layers respond to PGE2 and
have shown that animals actively or passively immunized PGF 2α in the same manner as the small intestine, although
with PG-protein conjugates develop gastrointestinal tract the results obtained preclude any generalization. For
erosions and ulcerations primarily in the stomach and example, low doses of PGE2 can induce activity in the distal
occasionally in the small bowel, in these experiments basal colon and be inhibitory in the proximal colon [64]. On the
and stimulated acid secretion did not differ in the treated other hand, in most animal studies, PGs of the E series relax
versus non-treated animals [57]. Therefore, the role of the lower esophageal sphincter whereas PGF2α constricts it
endogenous COX-derived products in gastric secretory [55]. In any case, when administered in vivo both PGs
physiology still remains controversial. decrease transit time and produce diarrhea through increased
Cyclooxygenase-2 Biology Current Pharmaceutical Design, 2003, Vol. 9, No. 27 2183

fluid and electrolyte secretion and/or altered gastrointestinal of a selective COX-2 inhibitor [80]. Therefore, the use of
motility [65, 66]. COX-2 selective inhibitors in patients with IBD should be
viewed with the same caution as the use of traditional
Little is known about the roles that COX-1 and COX-2
NSAIDs.
may play in the regulation of gastrointestinal motility.
Although several studies have shown that the non-selective COX-2 and Colon Cancer
NSAIDs indomethacin and aspirin impair intestinal
The most solid, unequivocal and well-established role of
peristalsis, which is the most relevant clinical motor pattern
COX-2 in the gastrointestinal system is its participation in
of the gut, recent findings suggest that this effect is unrelated
colon cancer [81-84].
to COX inhibition and have different targets, such as nuclear
factor-κ B and/or peroxisome proliferator-activated receptors Early observations in this field were obtained in studies
γ [67]. In fact, the selective COX-2 inhibitor NS-398 has comparing the levels of PGs in colon tumors with those of
been shown not to affect gastric motility in rats with normal colon tissue. The concentration of PGs, in particular
gastroduodenal ulcerogenic responses induced by of PGE 2, was found increased in human colorectal cancer
hypothermic stress [68]. Nevertheless, a role for prostanoids tissue when compared with normal colonic mucosa [85-87].
derived from COX-2 in postoperative bowel motility has Similar findings were reported in experimental models of
been proposed. In this regard, selective inhibition of COX-2 colonic carcinogenesis [88]. These early investigations also
significantly increases postoperative small bowel transit in established which PG series is involved in colon cancer.
experimental rodent models of postoperative ileus [69, 70]. Rigas et al. compared the levels of different PGs in colon
tumors with those of normal colon tissue and found that
Gastrointestinal Inflammation
PGE2 was elevated about two-fold in the cancer samples [86].
Eicosanoids along with many other soluble factors, such In contrast, no significant changes were found in the levels
as cytokines, reactive oxygen species, bacterial products and of PGF 2α and TXA 2, and, even, PGI2 was reduced about
lipid mediators, play a critical role in gastrointestinal three-fold in tumors [86]. Therefore, because both human and
inflammation. Several eicosanoids, including products from experimental colonic tumors produce increased amounts of
COX (PGE2 and TXB 2) and lipoxygenase (leukotriene (LT) this particular PG, PGE2 appears to be the eicosanoid
B 4 and HETEs), are increased in inflamed tissues compared involved in colon carcinogenesis.
with normal mucosa [71]. A positive correlation exists Since PGE2 levels are increased in tumors, COX is
between the increase in these eicosanoids and the activity of suspected to play a key role in the progression of colon
the disease, returning the levels during remission to values cancer. The relative levels of COX-1 and COX-2 expression
similar to those from normal colorectal mucosa [55]. An in colorectal cancers were evaluated by a number of
increasing body of evidence, especially in animal models of independent laboratories. All the investigations reported
inflammatory bowel disease (IBD), indicate that PGs increased expression of COX-2, but not COX-1, in colorectal
counteract the pro-inflammatory activity of LTB4. Indeed, carcinomas [89-91]. Published reports indicate that roughly
the administration of NSAIDs to either patients or animal 85% of adenocarcinomas exhibit a two- to fifty-fold increase
models with IBD does not have a salutary effect but rather is in COX-2 expression at both mRNA and protein levels
associated with flare-ups and colitis [72, 73]. This compared with matched, macroscopically normal, colonic
phenomenon is interpreted as NSAIDs suppressing PGE2 mucosa from the same patient [89-92]. Both carcinogen-
formation thus allowing LTB4 synthesis to proceed toward induced (azoxymethane-induced colonic tumors in rats) and
pro-inflammation. genetic (Min mice with multiple intestinal neoplasia) animal
In experiments using animal models of inflammation, models of colon cancer have confirmed the existence of an
such as carrageenan injection into the footpad and the murine increased expression of COX-2 in tumors [92-94].
air pouch model, COX-2 expression and activity is markedly Nevertheless, although COX-2 is differentially expressed in
upregulated [38, 74]. In these experimental models of acute various regions of the colon in human colorectal cancer, with
inflammation, selective COX-2 inhibition by inhibiting the COX-2 protein being greatly over-expressed in tumors
edema at the inflammatory site and being analgesic is located in the rectum [89-92], at present its cellular origin is
beneficial [38, 74, 75]. Therefore, because there is increased still debated. Cancer cells, nontransformed epithelial cells,
expression of COX-2 in both animal models of colitis and in stromal cells, vascular endothelial cells and inflammatory
human IBD [76, 77], in theory, selective COX-2 inhibition infiltrates are constituents of colon tissue and all of these cell
would also likely be beneficial in this disease. Although types are reported to have elevated levels of COX-2
preliminary results suggest that COX-2 inhibitors may be compared with the normal colon tissues [95]. On the
safe and beneficial in most patients with IBD [78], other contrary, subsequent studies have demonstrated that the
investigators do not agree because they have enough evidence increased expression of COX-2 in colon cancer originates
to support that PGs derived from COX-2 are important in mainly from the interstitial cells (i.e., macrophages), whereas
promoting the healing of mucosal injury, in protecting little COX-2 expression is found in epithelial cancer cells
against bacterial invasion, and in down-regulating the [96, 97].
mucosal immune system [79]. Indeed, in experimental Several mechanisms have been suggested for the
models, suppression of COX-2 in a setting of enhanced COX-2 gene expression in colon cancer, including
gastrointestinal inflammation and ulceration has been shown mutations of APC and ras, activation of EGF receptor and
to result in the impairment of healing and exacerbation on IGF-I receptor pathways and heregulin/HER-2 receptor
inflammation-mediated injury [76, 79]. In humans, clear pathway, direct induction by the Epstein-Barr virus
cases of exacerbation of IBD have been reported with the use oncoprotein and latent membrane protein 1 [98-100]. For
2184 Current Pharmaceutical Design, 2003, Vol. 9, No. 27 Joan Clària

instance, Oshima et al. assessed the development of apoptosis and suppress growth in many carcinomas,
intestinal adenomas in APC∆ 716 knockout mice (a model in including cultured human cancers of the stomach, esophagus,
which a targeted truncation deletion in the tumor suppressor tongue, brain, lung and pancreas [116].
gene APC causes intestinal polyposis) in a wild type and Recent studies indicate that angiogenesis, which is the
homozygous null COX-2 genetic background. The number development of new blood vessels and an essential step in
and size of polyps were dramatically reduced (six to eight- tumor growth, requires COX-2 [84]. In fact, COX-2
fold) in the COX-2 null mice compared with COX-2 wild- overexpressing cells produce large amounts of vascular
type mice [101]. endothelial growth factor (VEGF), a key proangiogenic
In vitro, the functional consequences of COX-2 over- factor, which stimulates both endothelial migration and in
expression have been analyzed in the rat intestinal cell line vitro angiogenesis [102]. These effects may be blocked by
(RIE cells), the gastrocolonic cell lines HT-29 and HCA-7 selective COX-2 inhibitors, highlighting the potential
and in the human colon cell line Caco-2 transfected with application of these compounds in blocking developing
COX-2. Overall, these experiments have revealed that cells tumors [113]. On the other hand, mice lacking the COX-2
over-expressing COX-2 undergo phenotypic changes that gene have deficient VEGF expression, reduced tumor
could enhance their tumorigenic potential, such as exhibition angiogenesis and decreased tumor growth [117]. Moreover,
of an increased adhesion to extracellular matrix proteins and selective inhibition of COX-2, but not COX-1, suppresses
resistance to apoptosis [102, 103]. The proliferative activity the growth of corneal capillary blood vessels in rats exposed
of COX-2 is believed to be primarily mediated by PGs. to basic fibroblast growth factor and inhibits the growth of
Along these lines, the human gastrocolonic cancer cell line, several human tumors implanted in mice [117, 118]. Taken
HT-29, shows increased proliferation in the presence of PGs together, these results support the notion of the benefit of
in the culture medium [104]. Moreover, in vivo, colonocyte selective COX-2 inhibition as an anti-angiogenic therapy.
proliferation is significantly stimulated by the injection of In addition to the well-documented implication of COX-
the stable derivative of PGE2, dimethyl PGE 2, into normal 2 in colon cancer, several investigations have recently
mice [104]. A conclusive proof of the role of COX-2 in cell examined its potential role in other epithelial cancers. These
growth is provided by the use of selective COX-2 inhibitors. studies have provided strong evidence that COX-2 may also
The effects of the highly selective COX-2 inhibitor, SC- be implicated in breast, head and neck, lung, pancreatic and
58125, was tested in two different cell lines, only one of gastric cancers and suggest that the beneficial effects of
which had a high level of COX-2 expression and activity. It COX-2 inhibitors may extend to these other cancers [119-
was observed that SC-58125 decreased cell growth only in 122]. Indeed, celecoxib consistently and dose-dependently
the COX-2 expressing cell line [105]. inhibits tumor growth and the number of lung metastasis in
In vivo, in human and experimental animals, a body of the syngenic Lewis lung carcinoma model [116].
evidence support the concept that inhibition of COX, and, in Interestingly, in animal studies, celecoxib has been shown to
particular COX-2, protects against colon cancer. potentiate the antitumor activity of conventional
Epidemiological studies have demonstrated the association chemotherapy and radiation [123, 124].
between regular long-term consumption of NSAIDs, in
particular aspirin, and reduced incidence of colon cancer [33, COX-2 and the Kidney
106-108]. Aspirin and sulindac have also been shown to
PGs play multiple roles in the adult kidney. PGE 2 and
reduce the number and size of its nonmalignant precursor,
PGI2 are powerful renal vasodilators that modulate intrarenal
the adenomatous colonic polyp, in patients with familial
vascular tone and influence glomerular hemodynamics and
adenomatous polyposis (FAP) [109, 110]. Moreover, a
renal perfusion [125, 126]. Moreover, PGE2 modulates renal
recent paper provides evidence of a protective association
sodium through direct inhibition of sodium reabsorption in
between aspirin and NSAIDs and esophageal cancer [111].
tubular epithelial cells [125, 126]. PGE2 also modulates
These epidemiological data are considered robust, because
renal water homeostasis by antagonizing the hydroosmotic
separate studies differing in locale, design and population
effect of antidiuretic hormone in the collecting duct [125,
have consistently yielded similar results. Parallel studies in
126]. Finally, PGs are involved in the renal responses to
animal models of colon carcinogenesis have also proven that
loop diuretics and in the regulation of renin secretion [126,
aspirin, as well as other traditional NSAIDs, such as
127]. Since PG synthesis is initiated by COX-1 and COX-2,
piroxicam, indomethacin, sulindac, ibuprofen and
and both COX isoforms are constitutively expressed in the
ketoprofen, inhibit chemically-induced colon cancer in rats
kidneys [128, 129], it was of interest to establish which
and mice [33]. The mechanism by which NSAIDs reduce the
COX isoform is involved in the maintenance of renal
risk of cancer is likely related to the inhibition of COX-2. In
function under normal conditions. The distribution of COX-
fact, a randomized clinical trial has shown that the selective
1 in the renal area differs significantly from that of COX-2.
COX-2 inhibitor, celecoxib, effectively inhibits the growth
In this regard, COX-1 is preferentially expressed in the renal
of adenomatous polyps and causes regression of existing
vasculature and in the medullary and papillary collecting
polyps in patients with hereditary FAP [112]. Studies in
ducts in humans, monkeys, dogs, rabbits and rats, whereas
rodents have also demonstrated that selective
COX-2 expression is consistently focal, and limited to the
pharmacological inhibition of COX-2 activity prevents
macula densa of the juxtaglomerular apparatus, epithelial
chemically-induced carcinogenesis and intestinal polyp
cells of the thick ascending limb and papillary interstitial
formation in an experimental model of FAP [113-115]. In
cells of rats, rabbits and dogs [128, 129]. In the adult human
Min mice and rats exposed to chemical carcinogens, selective
kidney, expression of COX-2 protein has been observed in
COX-2 inhibitors induce apoptosis as well as stimulate
Cyclooxygenase-2 Biology Current Pharmaceutical Design, 2003, Vol. 9, No. 27 2185

endothelial and smooth muscle cells of arteries and veins, appearance of fever [138]. Moreover, selective inhibitors of
and intra-glomerularly in podocytes, but not in macula densa COX-2 are potent antipyretic agents [139].
cells [130].
PGs are also now recognized as mediators of
At present, it is hypothesized that only PGs derived from inflammatory reactions in neural tissue and more recently of
COX-1 are involved in normal renal function. The marketed brain function. Constitutive COX-2 immunoreactivity and
selective COX-2 inhibitors, rofecoxib and celecoxib, have COX-2 mRNA expression have been detected in neurons and
been evaluated in randomized controlled trials in terms of specially in the forebrain [140]. The expression of COX-2 in
their effects on renal PGs, renal function and occurrence of the brain is particularly high in neonates [141]. Since brains
adverse renal events. Most of the studies have reported that of neonates have higher levels of cerebral PGs than those of
selective COX-2 inhibition does not significantly impair adults [141], and since these PGs are involved in the
renal function in healthy subjects [131]. Nevertheless, it is regulation of blood flow in the newborn [142], this
worthy to note that in healthy elderly patients, a population phenomenon suggests that COX-2 is likely to be important
more susceptible to renal NSAID-damaging effects, both in the modulation of blood flow at this stage of life.
celecoxib and rofecoxib induce a reduction in urinary sodium Furthermore, COX-2 is thought to play a key role in the
excretion similar to that of indomethacin and naproxen [132, final stages of development and brain modeling when COX-
133]. Although the role of COX-2-derived PGs in the kidney 2 becomes active in a manner that coincides with the
is still under debate, the finding that mice lacking the COX- imprinting of environmental influences [143].
2 gene develop mild to severe nephropathy during the first
weeks after birth [134, 135], supports the eventuality that Intense nerve stimulation, leading to seizures, induces
COX-2-derived PGs may play a physiological role in the COX-2 mRNA in discrete neurons of the hippocampus,
kidney other than maintenance of circulatory and excretory whereas acute stress raises levels in the cerebral cortex [140,
functions. 144]. The actual role of COX-2 and PGs in these sites is not
understood, but associations between COX-2 induction and
The incidence of renal side effects secondary to NSAID neural degeneration after glutamate stimulation, seizures and
treatment is low, and the role of PGs in renal homeostasis is spreading depression waves suggest that COX-2 may play
hardly noticeable in healthy subjects. In contrast, more of a role in the selective loss of neural connections than
administration of NSAIDs to patients with unbalanced in their formation [145]. Finally, current studies show that
effective arterial blood volume represents a major clinical COX-2 potentiates brain parenchymal amyloid plaque
problem since renal function in these patients is critically formation leading to Alzheimer’s disease, thus supporting a
dependent on PGs [37]. In these circumstances, which therapeutic potential for NSAIDs and COXIBs in the
include low sodium diet, hemorrhagic hypovolemia and treatment of this neurological disease [146].
edematous conditions associated with congestive heart
failure, nephrotic syndrome and decompensated liver COX-2 and the Reproductive Function
cirrhosis, NSAIDs induce a pronounced impairment in renal
plasma flow, glomerular filtration rate, urine volume and PGs are important for inducing uterine contractions
urinary sodium excretion. Since selective COX-2 inhibitors during labor. NSAIDs, such as indomethacin, delay
have been developed to effectively inhibit COX-2 while premature labor by inhibiting the production of PGs [147].
sparing physiologic PG production from COX-1, these novel However, the inhibition of PGs with indomethacin led to a
compounds could potentially be the anti-inflammatory of high mortality among infants because of premature closure of
choice in these patients. In this regard, in recent studies in the ductus arteriosus, a circulatory shunt maintained by PGs
rats with carbon tetrachloride-induced cirrhosis and ascites, an that allows the output of the left ventricle to bypass the fetal
experimental model that closely reproduces human liver lungs [147, 148]. PGs originating from COX-2 may play a
disease, selective COX-2 inhibition with celecoxib did not role in the birth process because COX-2 mRNA in the
seriously compromise renal function [48, 49]. Interestingly, amnion and placenta increases markedly immediately before
in these animals COX-1 protein expression was found to be and after the initiation of labor [149]. Theoretically, selective
unchanged whereas COX-2 protein levels were consistently inhibitors of COX-2 are able to reduce PG synthesis in fetal
upregulated [49]. Therefore, these results indicate that despite membranes and could be useful in delaying premature labor
abundant renal COX-2 protein expression, the maintenance of without the unwanted side effects of traditional NSAIDs.
renal function in altered effective arterial blood volume However, COX-2 also plays a role in ovulation, the
conditions, such as liver cirrhosis is mainly dependent on successful rupture of the follicle and in the implantation of
COX-1-derived PGs. the embryo in the uterine endometrium [150]. Indeed, COX-
2 null mice show multiple failures in reproductive function,
COX-2 and the Brain including ovulation, fertilization, implantation and
decidualization [151].
PGs have long been recognized as mediators of fever.
Fever is produced by PGE 2, acting on temperature-sensing COX-2 and Inflammation and Arthritis
neurons in the preoptic area [136]. The endogenous fever-
producing PGE2 is thought to originate from COX-2 induced The most important action of PGs is their role in
by lipopolysaccharide or interleukin-1 in endothelial cells inflammation [4]. In response to an inflammatory insult, the
lining the cerebral blood vessels [137]. In fact, release of PGs, and more importantly PGE 2, constitutes a
intraperitoneal injection of lipopolysaccharide to rats induces key event in the development of the three cardinal signs of
COX-2 in brain endothelial cells in parallel with the inflammation: swelling/redness, pain and fever. Given its
2186 Current Pharmaceutical Design, 2003, Vol. 9, No. 27 Joan Clària

potent vasodilatory properties, PGE2 increases tissue blood ABBREVIATIONS


flow, which results in the characteristic erythema (redness) of
COX = Cyclooxygenase
inflammation [4]. On the other hand, together with other
soluble factors including bradykinin, histamine and COXIBs = Selective COX-2 inhibitors
leukotrienes, PGE2 increases vascular permeability FAP = Familial adenomatous polyposis
contributing to fluid extravasation and the appearance of
edema (swelling) [4]. PGE2 also sensitizes afferent nerve LT = Leukotrienes
fibers acting both at peripheral sensory neurons and at central NSAIDs = Non-steroidal anti-inflammatory drugs
sites within the spinal cord and brain to evoke hyperalgesia
(pain) [4]. Finally PGE2 is a potent pyretic mediator PGs = Prostaglandins
involved in the appearance of fever [136]. Moreover, PGs TX = Thromboxane
also contribute to the amplification of the inflammatory
response by enhancing and prolonging signals produced by VEGF = Vascular endothelial growth factor
pro-inflammatory agents, such as bradykinin, histamine,
neurokinins and complement. REFERENCES
Although the importance of COX and PGs in References 157-159 are related articles recently published in
inflammation has been known for years, the inducibility of Current Pharmaceutical Design.
COX and PG formation has only fully been appreciated [1] Goldblatt MW. A depressor substance in seminal fluid. J Soc
recently with the uncovering of the properties of COX-2. Chem Ind (London) 1993; 52: 1056-1057 .
COX-2 was originally discovered as a transcript that was [2] von Euler US. A depressor substance in the vesicular gland. J
Physiology 1935; 84: 21P.
upregulated during inflammation and cellular transformation [3] Bergström S, Danielsson H, Samuelsson B. The enzymatic
[19-21]. Animal models of inflammatory arthritis provided formation of prostaglandin E2 from arachidonic acid. Biochim
the first available information regarding expression of COX- Biophys Acta 1964; 90: 207-210.
2 in acute and chronic inflammation, indicating that [4] Hecker M, Foegh ML, Ramwell PW. The eicosanoids:
increased expression of COX-2 is responsible for the Prostaglandins, Thromboxanes, Leukotrienes and related
compounds. In Basic and Clinical Pharmacology. Katzung, B.G.
increased PG production seen in inflamed joint tissues [75]. Eds; Appleton & Lange, Norwalk, CT, 1995; pp 290-304.
COX-2 induction was observed in both human osteoarthritis- [5] Vane JR. Inhibition of prostaglandin synthesis as a mechanism of
affected cartilage as well as in synovial tissue taken from action for aspirin-like drugs. Nat New Biology 1991; 231: 232-
patients with rheumatoid arthritis [152, 153]. The critical 235.
[6] Ferreira SH, Moncada S, Vane JR. Indomethacin and aspirin
role of COX-2 in inflammation led to the rational design of abolish prostaglandin release from the spleen. Nat New Biology
the first clinical trials for selective COX-2 inhibitors in 1971; 231: 237-239.
patients with osteoarthritis and rheumatoid arthritis [42-45]. [7] Smith JB, Willis AL. Aspirin selectively inhibits prostaglandin
The results of these drug-screening studies are sufficient to production in human platelets. Nat New Biology 1971; 231: 235-
guarantee that COX-2 inhibitors achieve the same anti- 237.
[8] Ohki S, Ogino N, Yamamoto S, Hayaishi O. Prostaglandin
inflammatory efficacy as traditional NSAIDs [42-45]. hydroperoxidase, an integral part of prostaglandin endoperoxide
synthetase from bovine vesicular gland microsomes. J Biol Chem
COX-2 and the Cardiovascular System 1979; 254: 829-836.
[9] Smith WL, Song I. The enzymology of prostaglandin
The major effects of PGs on the cardiovascular system are endoperoxide H synthases-1 and -2. Prostaglandins Other Lipid
on the smooth muscle cells and platelets where prostacyclin Mediat 2002; 68-69: 115-28.
(PGI2) has opposite biologic properties to those of TXA 2. [10] Jakobsson PJ, Thoren S, Morgenstern R, Samuelsson B.
Identification of human prostaglandin E synthase: a microsomal,
Thus, on the vasculature and especially on veins, PGI2 glutathione-dependent, inducible enzyme, constituting a potential
mainly promotes vasorelaxation while TXA2 acts as a novel drug target. Proc Natl Acad Sci USA 1999; 96: 7220-7225.
vasoconstrictor [4]. It was classically considered that PGI2 [11] Mancini JA, Blood K, Guay J, Gordon R, Claveau D, Chan CC, et
produced by vascular endothelial and smooth muscle cells al. Cloning, expression, and up-regulation of inducible rat
prostaglandin synthase during lipopolysaccharide-induced pyresis
was formed via COX-1. However, recent work has and adjuvant-induced arthritis. J Biol Chem 2001; 276: 4469-4475 .
demonstrated that PGI2 in vascular cells is produced by [12] Kozak KR, Rowlinson SW, Marnett LJ. Oxygenation of the
COX-2 as well as COX-1 [28]. Treatment of volunteers with endocannabinoid, 2-arachidonylglycerol, to glyceryl prostaglandins
a selective COX-2 inhibitor decreased urinary excretion of by cyclooxygenase-2. J Biol Chem 2000; 275: 3744-33749.
PGI2 without affecting TXA2 [132]. These results raised the [13] Clària J, Serhan CN. Aspirin triggers previously undescribed
bioactive eicosanoids by human endothelial cell-leukocyte
possibility of an increased risk of cardiovascular events interactions. Proc Natl Acad Sci USA 1995; 92: 9475-9479.
associated with COXIBs. However, two major randomized [14] Serhan CN, Hong S, Gronert K, Colgan SP, Devchand PR, Mirick
trials have recently shown that the relative risk of a G, et al. A family of bioactive products of omega-3 fatty acid
cardiovascular event with selective COX-2 inhibitors is transformation circuits initiated by aspirin treatment that counter
proinflammation signals. J Exp Med 2002; 196: 1025-103.
similar to that of traditional NSAIDs [154, 155]. On the [15] Morita I, Schindler M, Regier MK, Otto JC, Hori T, DeWitt DL, et
other hand, in platelets, TXA2 induces platelet aggregation al. Different intracellular locations for prostaglandin
whereas PGI2 strongly inhibits aggregation. It is widely endoperoxide H synthase-1 and -2. J Biol Chem 1995; 270: 10902-
recognized that platelets express only COX-1. Nevertheless, 10908.
in certain stages of megakaryogenesis, COX-2, as well as [16] Funk CD. Prostaglandins and leukotrienes: advances in eicosanoid
biology. Science. 2001; 294 : 1871-1875.
COX-1 are detected [156]. The biological significance of this [17] Hirai H, Tanaka K, Yoshie O, Ogawa K, Kenmotsu K, Takamori
phenomenon is, at present, unclear. Y, et al. Prostaglandin D2 selectively induces chemotaxis in T
Cyclooxygenase-2 Biology Current Pharmaceutical Design, 2003, Vol. 9, No. 27 2187

helper type 2 cells, eosinophils, and basophils via seven- of cyclooxygenase 2 in inflammation and pain. Proc Natl Acad
transmembrane receptor CRTH2. J Exp Med 2001; 193: 255-261. Sci USA 1994; 91: 12013-1201.
[18] Monneret G, Gravel S, Diamond M, Rokach J, Powell WS. [39] Gilroy DW, Tomlinson A, Willoughby DA. Differential effects of
Prostaglandin D2 is a potent chemoattractant for human inhibitors of cyclooxygenase (cyclooxygenase 1 and
eosinophils that acts via a novel DP receptor. Blood 2001; 98: cyclooxygenase 2) in acute inflammation. Eur J Pharmacology
1942-1948. 1998; 355: 211-217.
[19] Kujubu DA, Fletcher BS, Varnum BC, Lim RW, Herschman HR. [40] Smith CJ, Zhang Y, Koboldt CM, Muhammad J, Zweifel BS,
TIS10, a phorbol ester tumor promoter-inducible mRNA from Shaffer A, et al. Pharmacological analysis of cyclooxygenase-1 in
Swiss 3T3 cells, encodes a novel prostaglandin inflammation. Proc Natl Acad Sci USA 1998; 95: 13313-13318.
synthase/cyclooxygenase homologue. J Biol Chem 1991; 266: [41] Warner TD, Giuliano F, Vojnovic I, Bukasa A, Mitchell JA, Vane
12866-12872. JR. Nonsteroid drug selectivities for cyclo-oxygenase-1 rather
[20] Xie W, Chipman JG, Robertson DL, Erikson RL, Simmons DL. than cyclo-oxygenase-2 are associated with human
Expression of a mitogen-responsive gene encoding prostaglandin gastrointestinal toxicity: a full in vitro analysis. Proc Natl Acad Sci
synthase is regulated by mRNA splicing. Proc Natl Acad Sci USA. USA 1999; 967563-756.
1991; 88: 2692-2696. [42] Simon LS, Lanza FL, Lipsky PE, Hubbard RC, Talwalker S,
[21] O'Banion MK, Sadowski HB, Winn V, Young DA. A serum- and Schwartz BD, et al. Preliminary study of the safety and efficacy
glucocorticoid-regulated 4-kilobase mRNA encodes a of SC-58635, a novel cyclooxygenase 2 inhibitor: efficacy and
cyclooxygenase-related protein. J Biol Chem 1991; 266: 23261- safety in two placebo-controlled trials in osteoarthritis and
23267. rheumatoid arthritis, and studies of gastrointestinal and platelet
[22] Funk CD, Funk LB, Kennedy ME, Pong AS, Fitzgerald GA. effects. Arthritis Rheum. 1998; 41: 1591-1602.
Human platelet/erythroleukemia cell prostaglandin G/H synthase: [43] Simon LS, Weaver AL, Graham DY, Kivitz AJ, Lipsky PE,
cDNA cloning, expression, and gene chromosomal assignment. Hubbard RC, et al. Anti-inflammatory and upper gastrointestinal
FASEB J. 1991; 5 : 2304-2312. effects of celecoxib in rheumatoid arthritis: a randomized
[23] Yokoyama C, Tanabe T. Cloning of human gene encoding controlled trial. JAMA 1999; 282: 1921-1928.
prostaglandin endoperoxide synthase and primary structure of the [44] Emery P, Zeidler H, Kvien TK, Guslandi M, Naudin R, Stead H, et
enzyme. Biochem Biophys Res Commun 1989; 165 : 888-8. al. Celecoxib versus diclofenac in long-term management of
[24] Jones DA, Carlton DP, McIntyre TM, Zimmerman GA, Prescott rheumatoid arthritis: randomised double-blind comparison. Lancet
SM. Molecular cloning of human prostaglandin endoperoxide 1999 ; 354 : 2106-2111.
synthase type II and demonstration of expression in response to [45] Feldman M, McMahon AT. Do cyclooxygenase-2 inhibitors
cytokines. J Biol Chem 1993; 268: 9049-9054. provide benefits similar to those of traditional nonsteroidal anti-
[25] Tay A, Squire JA, Goldberg H, Skorecki K. Assignment of the inflammatory drugs, with less gastrointestinal toxicity? Ann Intern
human prostaglandin-endoperoxide synthase 2 (PTGS2) gene to Med 2000; 132: 134-143.
1q25 by fluorescence in situ hybridization. Genomics. 1994; 23: [46] Malmstrom K, Daniels S, Kotey P, Seidenberg BC, Desjardins PJ.
718-719. Comparison of rofecoxib and celecoxib, two cyclooxygenase-2
[26] Tanabe T, Tohnai N. Cyclooxygenase isozymes and their gene inhibitors, in postoperative dental pain: a randomized, placebo-
structures and expression. Prostaglandins Other Lipid Mediat and active-comparator-controlled clinical trial. Clin Ther 1999;
2002; 68-69: 95-114. 21: 1653-1663.
[27] Hla T, Neilson K. Human cyclooxygenase-2 cDNA. Proc Natl [47] Lanas A. Clinical experience with cyclooxygenase-2 inhibitors.
Acad Sci USA 1992; 89: 7384-7388. Rheumatology 2002; Supp 1, 16-22.
[28] Morita I. Distinct functions of COX-1 and COX-2. Prostaglandins [48] Bosch-Marcé M, Clària J, Titos E, Masferrer JL, Altuna R, Poo
Other Lipid Mediat 2002; 68-69: 165-175. JL, et al. Selective inhibition of cyclooxygenase-2 spares renal
[29] Willoughby DA, Moore AR, Colville-Nash PR. COX-1, COX-2, function and prostaglandin synthesis in cirrhotic rats with ascites.
and COX-3 and the future treatment of chronic inflammatory Gastroenterology 1999; 116: 1167-1175.
disease. Lancet 2000; 355: 646-648. [49] López-Parra M, Clària J, Planagumà A, Titos E, Masferrer JL,
[30] Chandrasekharan NV, Dai H, Roos KL, Evanson NK, Tomsik J, Koki A, et al. Cyclooxygenase-1 derived prostaglandins are
Elton TS, et al. COX-3, a cyclooxygenase-1 variant inhibited by involved in the maintenance of renal function in rats with cirrhosis
acetaminophen and other analgesic/antipyretic drugs: cloning, and ascites. Brit J Pharm 2002; 135: 891-900.
structure, and expression. Proc Natl Acad Sci USA 2002; 99: [50] Barton SF, Langeland FF, Snabes MC, LeComte D, Kuss ME,
13926-13931. Dhadda SS, et al. Efficacy and safety of intravenous parecoxib
[31] Flower RJ, Moncada S, Vane JR, Gillman AG, Goodman LS, Rall sodium in relieving acute postoperative pain following
TW, et al. Analgesic-antipyretics and anti-inflammatory agents: gynecologic laparotomy surgery. Anesthesiology 2002; 97: 306-
drugs employed in the treatment of gout. In The Pharmacological 314.
Basis of Therapeutics., editors. New York: Macmillan, 1992; pp [51] Rasmussen GL, Steckner K, Hogue C, Torri S, Hubbard RC.
674. Intravenous parecoxib sodium foracute pain after orthopedic
[32] Payan DG, Katzung BG. Nonsteroidal anti-inflammatory drugs; knee surgery. Am J Orthop 2002; 31: 336-343.
nonopioid analgesics; drugs used in gout. In Basic and Clinical [52] Redfern JS, Lee E, Feldman M. Effect of indomethacin on gastric
Pharmacology Katzung, B.G., Ed. Norwalk: Appleton & Lange, mucosal prostaglandins in humans. Correlation with mucosal
1995; pp 536-559. damage. Gastroenterology 1987; 92: 969-977.
[33] Thun MJ, Henley SJ, Patrono C. Nonsteroidal anti-inflammatory [53] Robert A, Nezamis JE, Phillips JP. Inhibition of gastric secretion
drugs as anticancer agents: mechanistic, pharmacologic, and by prostaglandins. Am J Dig Dis 1967; 12: 1073-1076.
clinical issues. J Natl Cancer Inst 2002; 94: 252-266. [54] Banerjee AK, Phillips J, Winning WM. E-type prostaglandins and
[34] Bazan NG, Colangelo V, Lukiw WJ. Prostaglandins and other lipid gastric acid secretion in the rat. Nat New Biol 1972; 238: 177-179.
mediators in Alzheimer's disease. Prostaglandins Other Lipid [55] Eberhart CE, Dubois RN. Eicosanoids and the gastrointestinal
Mediat 2002; 68-69: 197-210. tract. Gastroenterology 1995; 109: 285-301.
[35] Lanas A. Cyclo-oxygenase-1/cyclo-oxygenase-2 non selective [56] Redfern JS, Feldman M. Role of endogenous prostaglandins in
non-steroidal anti-inflammatory drugs: epidemiology of preventing gastrointestinal ulceration: induction of ulcers by
gastrointestinal events. Dig Liver Dis 2001; 33 Suppl 2, S29-34. antibodies to prostaglandins. Gastroenterology 1989; 96: 596-605.
[36] Giercksky KE, Huseby G, Rugstad HE. Epidemiology of NSAID- [57] Olson GA, Leffler CW, Fletcher AM. Gastroduodenal ulceration
related gastrointestinal side effects. Scand. J. Gastroenterol. Suppl in rabbits producing antibodies to prostaglandins. Prostaglandins
1989; 163: 3-8. 1985; 29: 475-480.
[37] Arroyo V, Ginés P, Rimola A, Gaya J. Renal function [58] Sawaoka H, Kawano S, Tsuji S, Tsuji M, Sun W, Gunawan ES, et
abnormalities, prostaglandins, and effects of nonsteroidal anti- al. Helicobacter pylori infection induces cyclooxygenase-2
inflammatory drugs in cirrhosis with ascites. An overview with expression in human gastric mucosa. Prostaglandins Leukot Essent
emphasis on pathogenesis. Am. J. Med. 1986; 81: 104-122. Fatty Acids. 1998; 59: 313-316.
[38] Seibert K, Zhang Y, Leahy K, Hauser S, Masferrer J, Perkins W, [59] McCarthy CJ, Crofford LJ, Greenson J, Scheiman JM.
et al. Pharmacological and biochemical demonstration of the role Cyclooxygenase-2 expression in gastric antral mucosa before and
2188 Current Pharmaceutical Design, 2003, Vol. 9, No. 27 Joan Clària

after eradication of Helicobacter pylori infection. Am J [82] Levy GN. Prostaglandin H synthases, nonsteroidal anti-
Gastroenterology 1999; 94: 1218-1223. inflammatory drugs, and colon cancer. FASEB J 1997; 11: 234-
[60] Romano M, Ricci V, Memoli A, Tuccillo C, Di Popolo A, Sommi P, 247.
et al. Helicobacter pylori up-regulates cyclooxygenase-2 mRNA [83] Kawai N, Tsujii M, Tsuji S. Cyclooxygenases and colon cancer.
expression and prostaglandin E2 synthesis in MKN 28 gastric Prostaglandins Other Lipid Mediat 2002; 68-69: 187-96 .
mucosal cells in vitro. J Biol Chem 1998; 273: 28560-28563. [84] Prescott SM. Is cyclooxygenase-2 the alpha and the omega in
[61] Xiao F, Furuta T, Takashima M, Shirai N, Hanai H. Effects of cancer? J Clin Invest 2000 ; 105 : 1511-1513.
cyclooxygenase-2 inhibitor on gastric acid secretion in [85] Bennett A, Civier A, Hensby CN, Melhuish PB, Stamford IF.
Helicobacter pylori-infected C57BL/6 mice. Scand J Measurement of arachidonate and its metabolites extracted from
Gastroenterology 2001; 36: 577-583. human normal and malignant gastrointestinal tissues. Gut 1987; 28:
[62] Burakoff R, Percy WH. Studies in vivo and in vitro on effects of 315-318.
PGE2 on colonic motility in rabbits. Am J Physiology 1992; 262: [86] Rigas B, Goldman IS, Levine L. Altered eicosanoid levels in
G23-G29. human colon cancer. J Lab Clin Med 1993; 122: 518-523.
[63] Moore PK. Eicosanoids and gastrointestinal motility. In [87] Pugh S, Thomas GA. Patients with adenomatous polyps and
Eicosanoids and the gastrointestinal tract. Hillier, K., Ed. M.T.P, carcinomas have increased colonic mucosal prostaglandin E2.
Boston, 1998; pp 145-162. Gut 1994; 35: 675-678.
[64] Bennett A, Eley KG, Stockley HL. The effects of prostaglandins [88] Rao CV, Simi B, Wynn TT, Garr K, Reddy BS. Modulating effect
on guinea-pig isolated intestine and their possible contribution to of amount and types of dietary fat on colonic mucosal
muscle activity and tone. Br J Pharmacology 1975; 54: 197-204. phospholipase A2, phosphatidylinositol-specific phospholipase C
[65] Soffer EE, Launspach J. Effect of misoprostol on postprandial activities, and cyclooxygenase metabolite formation during
intestinal motility and orocecal transit time in humans. Dig Dis Sci different stages of colon tumor promotion in male F344 rats.
1993; 38: 851-855. Cancer Res 1986; 56: 532-537.
[66] Milton-Thompson GJ, Cummings JH, Newman A, Billings JA, [89] Eberhart CE, Coffey RJ, Radhika A, Giardiello FM, Ferrenbach S,
Misiewicz JJ. Colonic and small intestinal response to intravenous Dubois RN. Up-regulation of cyclooxygenase 2 gene expression
prostaglandin F2 alpha and E2 in man. Gut 1995; 16: 42-46. in human colorectal adenomas and adenocarcinomas.
[67] Shahbazian A, Schuligoi R, Heinemann A, Peskar BA, Holzer P. Gastroenterology 1994; 107; 1183-1188.
Disturbance of peristalsis in the guinea-pig isolated small intestine [90] Kargman SL, O'Neill GP, Vickers PJ, Evans JF, Mancini JA, Jothy
by indomethacin, but not cyclo-oxygenase isoform-selective S. Expression of prostaglandin G/H synthase-1 and -2 protein in
inhibitors. Br J Pharmacology 2001; 132: 1299-13097. human colon cancer. Cancer Res 1995; 55: 2556-2559.
[68] Takeuchi K, Suzuki K, Araki H, Mizoguchi H, Sugamoto S, [91] Kutchera W, Jones DA, Matsunami N, Groden J, McIntyre TM,
Umdeda M. Roles of endogenous prostaglandins and nitric oxide Zimmerman GA, et al. Prostaglandin H synthase 2 is expressed
in gastroduodenal ulcerogenic responses induced in rats by abnormally in human colon cancer: evidence for a transcriptional
hypothermic stress. J Physiology Paris 1999 ; 93 : 423-431. effect. Proc Natl Acad Sci USA 1996; 93: 4816-4820.
[69] Josephs MD, Cheng G, Ksontini R, Moldawer LL, Hocking MP. [92] Gustafson-Svard C, Lilja I, Hallbook O, Sjodahl R.
Products of cyclooxygenase-2 catalysis regulate postoperative Cyclooxygenase-1 and cyclooxygenase-2 gene expression in
bowel motility. J Surg Res 1999; 86: 50-54. human colorectal adenocarcinomas and in azoxymethane induced
[70] Schwarz NT, Kalff JC, Turler A, Engel BM, Watkins SC, Billiar colonic tumours in rats. Gut 1996; 38: 79-84.
TR, et al. Prostanoid production via COX-2 as a causative [93] Dubois RN, Radhika A, Reddy BS, Entingh AJ. Increased
mechanism of rodent postoperative ileus. Gastroenterology 2001; cyclooxygenase-2 levels in carcinogen-induced rat colonic
121: 1354-1371. tumors. Gastroenterology 1996; 110: 1259-1262.
[71] Donowitz M. Arachidonic acid metabolites and their role in [94] Williams CS, Luongo C, Radhika A, Zhang T, Lamps LW, Nanney
inflammatory bowel disease. An update requiring addition of a LB, et al. Elevated cyclooxygenase-2 levels in Min mouse
pathway. Gastroenterology 1985; 88: 580-587. adenomas. Gastroenterology 1986; 111: 1134-1140.
[72] Kaufmann HJ, Taubin HL. Nonsteroidal anti-inflammatory drugs [95] Muller-Decker K, Albert C, Lukanov T, Winde G, Marks F,
activate quiescent inflammatory bowel disease. Ann Intern Med Furstenberger G. Cellular localization of cyclo-oxygenase
1987; 107: 513-516. isozymes in Crohn's disease and colorectal cancer. Int J
[73] Wallace JL, Keenan CM, Cucala M, Mugridge KG, Parente L. Colorectal Dis 1999; 14: 212-218.
Mechanisms underlying the protective effects of interleukin 1 in [96] Shattuck-Brandt RL, Varilek GW, Radhika A, Yang F,
experimental nonsteroidal anti-inflammatory drug gastropathy. Washington MK, Dubois RN. Cyclooxygenase 2 expression is
Gastroenterology 1992; 102: 1176-1185. increased in the stroma of colon carcinomas from IL-10(-/-)
[74] Masferrer JL, Zweifel BS, Manning P,T, Hauser SD, Leahy KM, mice. Gastroenterology 2000; 118: 337-345.
Smith WG, et al. Selective inhibition of inducible cyclooxygenase [97] Bamba H, Ota S, Kato A, Adachi A, Itoyama S, Matsuzaki F. High
2 in vivo is antiinflammatory and nonulcerogenic. Proc Natl Acad expression of cyclooxygenase-2 in macrophages of human
Sci USA 1994; 91: 3228-3232. colonic adenoma. Int J Cancer 1999; 83: 470-475.
[75] Anderson GD, Hauser SD, McGarity KL Bremer ME, Isakson PC, [98] Di Popolo A, Memoli A, Apicella A, Tuccillo C, di Palma A,
Gregory SA. Selective inhibition of cyclooxygenase (COX)-2 Ricchi P, et al. IGF-II/IGF-I receptor pathway up-regulates COX-
reverses inflammation and expression of COX-2 and interleukin 6 2 mRNA expression and PGE2 synthesis in Caco-2 human colon
in rat adjuvant arthritis. J Clin Invest 1996; 97: 2672-2679. carcinoma cells. Oncogene 2000; 19: 5517-5524.
[76] Reuter BK, Asfaha S, Buret A, Sharkey KA, Wallace JL. [99] Vadlamudi R, Mandal M, Adam L, Steinbach G, Mendelsohn J,
Exacerbation of inflammation-associated colonic injury in rat Kumar R. Regulation of cyclooxygenase-2 pathway by HER2
through inhibition of cyclooxygenase-2. J Clin Invest 1996; 98 : receptor. Oncogene 1999; 18: 305-314.
2076-2085. [100] Murono S, Inoue H, Tanabe T, Joab I, Yoshizaki T, Furukawa M,
[77] Hendel J, Nielsen OH. Expression of cyclooxygenase-2 mRNA in et al. Induction of cyclooxygenase-2 by Epstein-Barr virus latent
active inflammatory bowel disease. Am J Gastroenterology 1997; membrane protein 1 is involved in vascular endothelial growth
92: 1170-1173. factor production in nasopharyngeal carcinoma cells. Proc Natl
[78] Mahadevan U, Loftus EV Jr, Tremaine WJ, Sandborn WJ. Safety Acad. Sci USA 2001; 98: 6905-6910.
of selective cyclooxygenase-2 inhibitors in inflammatory bowel [101] Oshima M, Dinchuk JE, Kargman SL, Oshima H, Hancock B,
disease. Am J Gastroenterology 2002; 97: 910-914. Kwong E, et al. Suppression of intestinal polyposis in Apc
[79] Wallace JL. Prostaglandin biology in inflammatory bowel disease. delta716 knockout mice by inhibition of cyclooxygenase 2 (COX-
Gastroenterology Clin North Am 2001; 30: 971-980 . 2). Cell 1996; 87: 803-809.
[80] Bonner GF. Exacerbation of inflammatory bowel disease [102] Tsujii M, Dubois RN. Alterations in cellular adhesion and
associated with use of celecoxib. Am J Gastroenterology 2001; apoptosis in epithelial cells overexpressing prostaglandin
96: 1306-1308. endoperoxide synthase 2. Cell 1995; 83: 493-501.
[81] Dubois RN. Nonsteroidal anti-inflammatory drug use and sporadic [103] Dubois RN, Shao J, Tsujii M, Sheng H, Beauchamp RD. G1 delay
colorectal adenomas. Gastroenterology 1995; 108: 1310-1314. in cells overexpressing prostaglandin endoperoxide synthase-2.
Cancer Res 1996; 56: 733-737.
Cyclooxygenase-2 Biology Current Pharmaceutical Design, 2003, Vol. 9, No. 27 2189

[104] Qiao L, Kozoni V, Tsioulias GJ, Koutsos MI, Hanif R, Shiff SJ, et [126] Mattix HJ, Badr KF. Arachidonic acid metabolites and the kidney.
al. Selected eicosanoids increase the proliferation rate of human In The Kidney, Brenner, B.M. and Rector, F.C.; Eds.; Saunders:
colon carcinoma cell lines and mouse colonocytes in vivo. Philadelphia, 2000; pp 756.
Biochim Biophys Acta 1995; 1258: 215-2. [127] Katyama S, Attallah AA, Stahl RA, Bloch DL, Lee JB. Mechanism
[105] Sheng H, Shao J, Kirkland SC, Isakson P, Coffey RJ, Morrow J, et of furosemide-induced natriuresis by direct stimulation of renal
al. Inhibition of human colon cancer cell growth by selective prostaglandin E 2 . Am J Physiology 1984; 247: F555-561.
inhibition of cyclooxygenase-2. J. Clin. Invest 1997; 99 : 2254- [128] O´Neill GP, Ford-Hutchinson AW. Expression of mRNA for
2259. cyclooxygenase-1 and cyclooxygenase-2 in human tissues. FEBS
[106] Thun MJ, Namboodiri MM, Calle EE, Flanders WD, Heath CWJr. Lett 1993; 330: 156-160.
Aspirin use and risk of fatal cancer. Cancer Res 1993; 53: 1322- [129] Khan KN, Venturini CM, Bunch RT, Brassard JA, Koki AT,
1327. Morris DL, et al. Interspecies differences in renal localization of
[107] Giovannucci E, Egan KM, Hunter DJ, Stampfer MJ, Colditz GA, cyclooxygenase isoforms: implications in nonsteroidal
Willett WC, et al. Aspirin and the risk of colorectal cancer in antiinflammatory drug-related nephrotoxicity. Toxicology
women. N Engl J Med 1995; 333: 609-614. Pathology 1998; 26: 612-620.
[108] Schreinemachers DM, Everson RB. Aspirin use and lung, colon, [130] Kömhoff M, Gröne HJ, Klein T, Seyberth HW, Nüssing RM.
and breast cancer incidence in a prospective study. Epidemiology Localization of cyclooxygenase-1 and -2 in adult and fetal human
1994; 5: 138-146. kidney: implication for renal function. Am J Physiology 1997; 272:
[109] Rigau J, Pique JM, Rubio E, Planas R, Tarrech JM, Bordas JM. F460-F468.
Effects of long-term sulindac therapy on colonic polyposis. Ann [131] Whelton A, Maurath CJ, Verburg KM, Geis GS. Renal safety and
Intern Med 1991; 115: 952-954. tolerability of celecoxib, a novel cyclooxygenase-2 inhibitor. Am
[110] Suh O, Mettlin C, Petrelli NJ. Aspirin use, cancer, and polyps of J Ther 2000; 7: 159-17.
the large bowel. Cancer 1993; 72: 1171-1177. [132] Catella-Lawson F, McAdam B, Morrison BW, Kapoor S, Kujubu
[111] Corley DA, Kerlikowske K, Verma R, Buffler P. Protective D, Antes L, et al. Effects of specific inhibition on yclooxygenase-
association of aspirin/NSAIDs and esophageal cancer: a 2 on sodium balance, hemodynamics and vasoactive eicosanoids.
systematic review and meta-analysis. Gastroenterology 2003; 124: J Pharm Exp Ther 1999; 289: 735-741.
47-56. [133] Whelton A, Schulman G, Wallemark C, Drower EJ, Isakson PC,
[112] Steinbach G, Lynch PM, Phillips RK, Wallace MH, Hawk E, Verburg KM, et al. Effects of celecoxib and naproxen on renal
Gordon GB, et al. The effect of celecoxib, a cyclooxygenase-2 function in the elderly. Arch Intern Med 2000; 160: 1465-1470.
inhibitor, in familial adenomatous polyposis. N Engl J Med 2000; [134] Morham SG, Langenbach R, Loftin CD, Tiano HF, Vouloumanos
342: 1946-1952. N, Jennette JC, et al. Prostaglandin synthase 2 gene disruption
[113] Kawamori T, Rao CV, Seibert K, Reddy BS. Chemopreventive causes severe renal pathology in the mouse. Cell 1995; 83: 473-
activity of celecoxib, a specific cyclooxygenase-2 inhibitor, 482.
against colon carcinogenesis. Cancer Res 1998; 58: 409-412. [135] Dinchuk JE, Car BD, Focht RJ, Johnston JJ, Jaffee BD, Covington
[114] Jacoby RF, Seibert K, Cole CE, Kelloff G, Lubet RA. The MB, et al. Renal abnormalities and an altered inflammatory
cyclooxygenase-2 inhibitor celecoxib is a potent preventive and response in mice lacking cyclooxygenase II. Nature 1995; 378:
therapeutic agent in the min mouse model of adenomatous 406-409.
polyposis. Cancer Res 2000; 60: 5040-5044. [136] Milton AS. Antipyretic actions of aspirin. In Aspirin and other
[115] Oshima M, Murai N, Kargman S, Arguello M, Luk P, Kwong E, et salicylates, Vane, J.R. and Botting, R.M.; Eds; Chapman&Hall,
al. Chemoprevention of intestinal polyposis in the Apcdelta716 London, 1992; pp 213-244.
mouse by rofecoxib, a specific cyclooxygenase-2 inhibitor. [137] Cao C, Matsumara K, Yamagata K, Watanabe Y. Endotelial cells
Cancer Res 2001; 61: 1733-1740. of the brain vasculature express cyclooxygenase-2 mRNA in
[116] Koki AT, Khan NK, Woerner BM, Seibert K, Harmon JL, response to systemic interleukin-1b: a posible site of prostaglandin
Dannenberg AJ, et al. Characterization of cyclooxygenase-2 synthesis responsible for fever. Brain Res 1996; 733: 263-272.
(COX-2) during tumorigenesis in human epithelial cancers: [138] Cao C, Matsumara K, Yamagata K, Watanabe Y. Involvement of
evidence for potential clinical utility of COX-2 inhibitors in cyclooxygenase-2 in LPS-induced fever and regulation of its
epithelial cancers. Prostaglandins Leukot Essent Fatty Acids 2002; mRNA by LPS in the rat brain. Am J Physiology 1997; 271:
66: 13-18. R1712-R1725.
[117] Williams CS, Tsujii M, Reese J, Dey SK, Dubois RN. Host [139] Futaki N, Yoshikawa K, Hamasaka Y. NS-398, a novel non-
cyclooxygenase-2 modulates carcinoma growth. J Clin Invest steroidal anti-inflammatory drug with potent analgesic and
2000 ; 105 : 1589-1594. antipyretic effects which causes minimal stomach lesions. Gen
[118] Masferrer JL, Leahy KM, Koki AT, Zweifel BS, Settle SL, Pharmacology 1993; 24: 105-110.
Woerner BM, et al. Antiangiogenic and antitumor activities of [140] Yamagata K, Andreasson KI, Kaufman WE, Barnes CA.
cyclooxygenase-2 inhibitors. Cancer Res 2000; 60: 1306-1311. Expression of a mitogen-inducible cyclooxygenase in brain
[119] Subbaramaiah K, Telang N, Ramonetti, JT, Araki, R, DeVit, B, neurons: regulation by synaptic activity and glucocorticoids.
Weksler BB, et al. Transcription of cyclooxygenase-2 is Neuron 1993; 11: 371-386 .
enhanced in transformed mammary epithelial cells. Cancer Res [141] Peri KG, Hardy P, Li DY, Varma DR, Chemtob S. Prostaglandin
1996; 56: 4424-4429. G/H synthase-2 is a major contributor of brain prostaglandins in
[120] Chan G, Boyle JO, Yang EK, Zhang F, Sacks PG, Shah JP, et al. the newborn. J Biol Chem 1995; 270: 24615-20.
Cyclooxygenase-2 expression is up-regulated in squamous cell [142] Chemtob S, Beharry K, Rex J, Varma DR, Aranda JV. Prostanoids
carcinoma of the head and neck. Cancer Res 1999; 59: 991-994. determine the range of cerebral blood flow autoregulation of
[121] Tucker ON, Dannenberg AJ, Yang EK, Zhang F, Teng L, Daly newborn piglets. Stroke 1990; 21: 777-84.
JM, et al. Cyclooxygenase-2 expression is up-regulated in human [143] Kaufmann WE, Worley PF, Pegg J, Bremer M, Isakson P.
pancreatic cancer. Cancer Res 1999; 59: 987-990. Cyclooxygenase-2 expression during rat neocortical development
[122] Ristimaki A, Honkanen N, Jankala H, Sipponen P, Harkonen M. and in Rett syndrome. Brain Dev 1997; 19: 25-34.
Expression of cyclooxygenase-2 in human gastric carcinoma. [144] Marcheselli VL, Bazan NG. Sustained induction of prostaglandin
Cancer Res 1997; 57: 1276-1280. endoperoxide synthase-2 by seizures in hippocampus. J Biol Chem
[123] Milas L, Kishi K, Hunter N, Mason K, Masferrer JL, Tofilon PJ. 1996; 271: 24794-9.
Enhancement of tumor response to gamma-radiation by an [145] Miettinen S, Fusco FR, Yrjanheikki J, Keinanen R, Hirvonen T,
inhibitor of cyclooxygenase-2 enzyme. J. Natl. Cancer Inst 1999; Roivainen R, et al. Spreading depression and focal brain ischemia
91: 1501-1504. induce cyclooxygenase-2 in cortical neurons through N-methyl-
[124] Trifan OC, Durham WF, Salazar VS, Horton J, Levine BD, D-aspartic acid-recptors and phospholipase A2. Proc Natl Acad
Zweifel BS, et al. Cyclooxygenase-2 inhibition with celecoxib Sci USA 1997; 94: 6500-6505.
enhances antitumor efficacy and reduces diarrhea side effect of [146] Stewart WF, Kawas C, Corrada M, Metter EJ. Risk of
CPT-11. Cancer Res 2002; 62: 5778-5784 . Alzheimer´s disease and duration of NSAID use. Neurology
[125] Schlondorff D, Ardaillou R. Prostaglandins and other arachidonic 1997; 48: 626-632.
acid metabolites in the kidney. Kidney Int 1986; 29: 108-119.
2190 Current Pharmaceutical Design, 2003, Vol. 9, No. 27 Joan Clària

[147] Zuckerman H, Reiss U, Rubinstein I. Inhibition of human [154] Bombardier C, Laine L, Reicin A, Shapiro D, Burgos-Vargas R,
premature labor by 94 indomethacin Obstet Gynecology 1974 ; Davis B, et al. Comparison of upper gastrointestinal toxicity of
44 : 787-92. rofecoxib and naproxen in patients with rheumatoid arthritis.
[148] Hammerman C. Patent ductus arteriosus. Clinical relevance of VIGOR Study Group. N Engl J Med 2000; 343: 1520-8, 2.
prostaglandins and prostaglandin inhibitors in PDA [155] Silverstein FE, Faich G, Goldstein JL, Simon LS, Pincus T,
pathophysiology and treatment. Clin Perinatology 1995; 22: 457- Whelton A, et al. Gastrointestinal toxicity with celecoxib vs
79. nonsteroidal anti-inflammatory drugs for osteoarthritis and
[149] Gibb W, Sun M. Localization of prostaglandin H synthase type 2 rheumatoid arthritis: the CLASS study: A randomized controlled
protein and mRNA in term human fetal membranes and decidua. J trial. Celecoxib Long-term Arthritis Safety Study. JAMA 2000;
Endocrinology 1996; 150: 97-503. 284: 1247-1255.
[150] Dubois RN, Abramson SB, Crofford L, Gupta RA, Simon LS, Van [156] Rocca B, Secchiero P, Ciabattoni G, Ranelletti FO, Catani L,
De Putte LB, et al. Cyclooxygenase in biology and disease. Guidotti L, et al. Cyclooxygenase-2 expression is induced during
FASEB J 1998; 12: 1063-1073. human megakaryopoiesis and characterizes newly formed
[151] Lim H, Paria BC, Das SK, Dinchuk JE, Langenbach R, Trzaskos platelets. Proc Natl Acad Sci USA 2002; 99: 7634-7639.
JM, et al. Multiple female reproductive failures in [157] Oshima M, Taketo MM. COX selectivity and animal models for
cyclooxygenase 2-deficient mice. Cell 1997; 91: 197-208. colon cancer. Curr Pharm Design 2002; 8(12): 1021-34.
[152] Amin AR, Attur M, Patel RN, Thakker GD, Marshall PJ, Rediske [158] Rodriguez-Tellez M, Arguelles F, Herrerias JM Jr, Ledro D,
J, et al. Superinduction of cyclooxygenase-2 activity in human Esteban J, Herrerias JM. Antiinflamatory agents less dangerous
osteoarthritis-affected cartilage. Influence of nitric oxide. J Clin for gastrointestinal tract. Curr Pharm Design 2001; 7(10): 951-76.
Invest 1997; 99: 1231-1237. [159] Takeuchi K, Tanaka A, Suzuki K, Mizoguchi H. Gastrointestinal
[153] Kang RY, Freire-Moar J, Sigal E, Chu CQ. Expression of sparing anti-inflammatory drugs--effects on ulcerogenic and
cyclooxygenase-2 in human and an animal model of rheumatoid healing responses. Curr Pharm Design 2001; 7(1): 49-69.
arthritis. Br J Rheumatology 1996; 35: 711-718.
.

You might also like