Sie sind auf Seite 1von 9

British Journal of Rheumatology 1996; 35(suppl.

1):4—12

PHARMACOLOGY OF MELOXICAM, A NEW NON-STEROIDAL


ANTI-INFLAMMATORY DRUG WITH AN IMPROVED SAFETY PROFILE
THROUGH PREFERENTIAL INfflBITION OF COX-2
G.ENGELHARDT
Dr Karl Thomae GmbH, Department of BiologicalResearch.D-88400 BiberachlRiss, Germany

SUMMARY
Thisreviewfocuses on key pharmacological findings with a new NSAID, meloxicam. Unlike established NSAIDs, it preferentially
inhibits inducible COX-2 in guinea-pig peritoneal macrophages and human COX-2 in COS cells. Compared with other NSAIDs,
meloxicam is the most potent inhibitor of prostaglandin biosynthesis in pleural and peritoneal exudate, but only a weak inhibitor in
the gastric tract and kidney. Ulcerogenicity in the rat stomach is weak in relation to anti-inflammatory potency,resultingin a high
therapeutic index. Meloxicam's high anti-inflammatory potency combined with good tolerability can be explained by its
preferential inhibition of COX-2. In adjuvant arthritis rats, meloxicam inhibits not only paw swelling, but also bone and cartilage
destruction and systemic signs of disease. It inhibits leucocyte migration, but has no effect on leucotriene B4 or C4. Meloxicam
shows a long-lasting anti-inflammatory and analgesic effect on inflammatory pain and reduces pyrogen-induced fever, but has no
central nervous system effects. The pharmacokinetic profile of meloxicam in the rat is similar to that in man. Metabolites are
inactive.
KEY WORDS: Meloxicam, Anti-inflammatory, Cyclooxygenase, Pharmacology, Prostaglandins.

MELOXICAM (Fig. 1) is a new non-steroidal anti- inflammatory effects of NSAIDs are thought to be
inflammatory drug (NSAID), registered in France, mediated via the inhibition of the other isoform,
Meloxicam shows a novel pharmacodynamic profile. COX-2 [10]. The induction of COX-2 by inflammatory
Since 1971 [1] it has been generally accepted that the stimuli, cytokines or lipopolysaccharides has been
mechanism for both the therapeutic anti-inflammatory, demonstrated not only in macrophages [4, 11-15] but
analgesic and antipyretic actions and the common also in endothelial cells [13, 16, 17] and synoviocytes
deleterious effects [2] of aspirin-like drugs is mediated [18-21]. The pharmacology of COX-1 is also different
through their inhibition of cyclooxygenase (COX), the from that of COX-2, such that several NSAIDs have
rate-limiting enzyme in the synthesis of prostaglandins. been shown to display differential inhibitory activity
This action fails to explain why NSAIDs, at equipotent against COX-2 and COX-1 [22, 23]. Indomethacin,
doses, cause different degrees of gastrointestinal (GI) acetylsalicylic acid and piroxicam are more active
adverse effects [3]. against COX-1 than against COX-2. This differential
We now know that COX exists in two isoforms [4-6], inhibitory activity is thought to explain the differing
known as COX-1 and COX-2. The two isoforms have side-effect profiles of current NSAIDs such that those
different structures and functions [7-9]. The constitutive with the highest selectivity for COX-1 tend to provoke
COX isoform, COX-1, is involved in processes such as most adverse events. Moreover, in principle, a NSAID
the production of thromboxane A.2 (TXA2), the which displays preferential COX-2 inhibition would be
production of prostaglandin E2 (PGE2) in the kidneys expected to have potent anti-inflammatory effects whilst
and the production of prostacyclin, which is both sparing the patient from treatment-limiting effects, on
antithrombogenic and, in the gastric mucosa, cyto- the gastric mucosa for example. Meloxicam exhibits
protective. The undesirable side-effects of NSAIDs on preferential inhibition of COX-2 over COX-1. A
the stomach and kidneys are thought to be due to number of other potential selective COX-2 inhibitors
the inhibition of COX-1 [10]. The beneficial anti- are also in the early phases of development. These
include: flosulide (CGP-28238), SC-58125, NS-398,
L-745,337 and DUP-697.
OH This review presents the key pharmacological
CONH
findings for meloxicam, focusing on those which have
led to the characterization of meloxicam as a NSAID
with an improved safety profile over current treatments
through the preferential inhibition of COX-2.
EFFECTS OF MELOXICAM
Fio. 1.—Structure of meloxicam. Influence on arachidonic acid metabolism
The in vitro and in vivo activities of meloxicam and
Correspondence to: G. Engelhardt, Department of Biological other NSAIDs against COX-1 and COX-2 have been
Research, Dr Karl Thomae GmbH, D-88400 Biberach/Riss, Germany. compared in several models.

O 1996 British Society for Rheumatology


ENGELHARDT: PHARMACOLOGY OF MELOXICAM

TABLE I
Influence on COX-1 and COX-2 activity of guinea-pig peritoneal 0-1 -i
macrophages during 6 h of incubation
COX-1 COX-2 Ratio
NSAID IC» (nmol/1) ICJO (nmol/I) COX-2/COX-1

Meloxkam 5.8 1.9 0.33


(4.6-7J) (1.4-2.7)
Piroxicam 5.3 175 33
(3.6-7.4) (149-202)
Tenoxicam 20 322 16
(5.8-179) (207-489)
Tenidap 393 47 800 122
(299-519) (39 200-59 700) 1000
Indometnacin 0.21 6.4 30
(0.13-0.31) (5.0-8.0)
Diclofenac 0.86 1.9 13.
(0.58-1.17) (1.5-2.4)
Flurbiprofen 15 4760 317 Fio. 2.—Relation of inhibitory activity against COX-1 and COX-2 in
(8.6-28.2) (1270-12 700) guinea-pig peritoneal macrophages in vitro. Ratio of IC50 against
COX-2/ICJO against COX-1.
* Figures in parentheses are 95% confidence limits.

acetylsalicylic acid, naproxen and ibuprofen were either


non-selective inhibitors of COX-1 and COX-2 or
Investigations have been conducted in models where selective inhibitors of COX-1 [26].
COX-1 and COX-2 expression are induced in animal The preferential inhibition of COX-2 by meloxicam is
intact cell systems of guinea-pig macrophages or highly dependent on the structure of the drug.
isolated from cell-free preparations from bovine seminal Modification of meloxicam to the 4'-isomer (the methyl
vesicles, bovine brain or sheep placenta for in vitro substitution on the thiazole group is located at position
studies. In addition, effects have been established using 4 rather than position 5) resulted in a significant loss of
human COX-1 and COX-2 transfected into cultured selectivity against COX-2. The 4'-isomer showed
COS-2 cells. selectivity for COX-1 [27].
Meloxicam shows weak activity against COX-1 in a The influence of meloxicam and some other NSAIDs
cell-free enzyme preparation from bovine seminal on PGE2 synthesis in various non-inflamed and
vesicles [24, 25], whilst indomethacin is 18 times, inflamed tissues has been investigated in vivo. Their
diclofenac 29 times and flurbiprofen 45 times more differential abilities to inhibit the expression of COX-2
active than meloxicam. in inflamed areas (pleurisy of the rat, peritonitis of
In the intact cell system, indomethacin was the most mice) and to influence the activity of constitutive
potent inhibitor of COX-1 activity in non-stimulated COX-1 in non-inflamed areas such as the stomach,
macrophages. Meloxicam and piroxicam were more kidney and brain were examined. These investigations
active than flurbiprofen, tenoxicam and tenidap in this are of clinical interest because the acute, noxious effects
cell system (Table I). Lipopolysaccharide (LPS) is used of NSAIDs, such as decrease in renal blood flow
to induce COX-2 and stimulate PGE2 production in and electrolyte and water retention, are mediated by
guinea-pig peritoneal macrophages. The differences inhibition of" this intrarenal PGE2 synthesis [28]. Ex-
between the activity of the NSAIDs tested against these cretion of intact, non-metabolized PGE2 is a measure
two cell models of COX-2 and COX-1 are demon- for COX-1-induced production of intrarenal PGE2 [29].
strated in Table I. In the gastric mucosa, COX-1 mediates the formation
It can be seen from the COX-2/COX-1 inhibitory of cytoprotective prostaglandins. Specifically, the
ratios that there are clear differences between the inhibition of PGI2 and PGE2 synthesis is implicated in
different NSAIDs in terms of preferential inhibition of the pathogenesis of NSAID-induced gastric ulcers [30].
one COX isozyme over the other (Table I and Fig. 2). Pleurisy of the rat was used as a model of the
Of all the NSAIDs tested, only meloxicam prefer- inflammatory process. In this carrageenan-induced
entially inhibited COX-2-induced in LPS-stimulated model of inflammation, COX-2 is responsible for the
cells over COX-1 present in non-stimulated cells. production of prostaglandins [31].
Piroxicam, tenoxicam, tenidap, indomethacin and flur- All NSAIDs tested lowered the PGE2 content of the
biprofen inhibited the COX activity in non-stimulated pleuritic exudate from the rat in a dose-dependent
macrophages more potently than in LPS-stimulated manner. Meloxicam was twice as potent as tenoxicam,
macrophages. Diclofenac displayed similar activity three times as potent as flurbiprofen and eight times as
against COX-1 and COX-2 in these cell models. potent as diclofenac as an inhibitor of COX-2-induced
These results were confirmed in human COX-1 and PGE2 production in this model. Tenidap displayed only
COX-2 stably transfected into cultured COS-2 cells. weak inhibitory activity against COX-2 [32].
Meloxicam is a selective inhibitor of human COX-2 in Intrarenal PGE2 synthesis, mediated by COX-1,
this assay system. In contrast, piroxicam, indomethacin, was inhibited by all of the NSAIDs tested in a
STUDIES ON MELOXICAM (MOBIQ

TABLED higher than for diclofenac and 50 times higher than for
Influence on PGE2 content of pleuritic exudate and urine of rats tenidap (Table II). Similarly, the ratio of the ID50
Pleuritic exudate Urine IDJO Ratio for inhibition of PGE2 production in the gastric
NSAID I D M (mg/kg/day) (mg/kg) urine/pleurisy mucosa to the ID50 for the inhibition of PGE2
production in the pleuritic exudate gives an indication
Meloxicam 0.65 1.85 2.8
(0.54-0.78)' (1.05-2.78)
of the gastric tolerance of an agent (Table III). For
Piroxkam 0.85 0.24 0.28 meloxicam the ratio was 40 times higher than that of
(0.60-1.09) (0.10-0.41) diclofenac and naproxen and 100 times greater than
Tenoxicam 1.32 0.62 0.47 that of flurbiprofen.
(0.14-1.52) (0.40-0.94)
Tenidap 12.8 0.64 0.05
Further investigations have reviewed the effects of
(9.62-18.0) (0.23-1.50) various NSAIDs on COX activity in vivo in other tissues
Diclofenac 5.06 1.86 0.37 such as the brain and serum, and also on inflammatory
(3.71-4.62) (1.23-2.55) products of lipooxygenase (another enzyme, distinct
Flurbiprofen 2.18 0.26 0.12 from COX, which is active within the arachidonic acid
(1.78-2.75) (0.11-0.58)
cascade).
* Figures in parentheses are 95% confidence limits. In rats and mice, under physiological conditions, a
small amount of PGE2 is present in brain tissue.
TABLE IU Following administration of a convulsant dose of
Influence on PGE2 content of pleuritic exudate and gastric juice of pentetrazole to rats and mice, a rapid rise in brain PGE2
rats
can be observed. In this model the glucocorticosteroid
Pleuritic exudate Gastric juice Ratio gastric dexamethasone does not inhibit the pentetrazole-
NSATD ID50 (mg/kg/day) ID30 (mg/kg) juice/pleurisy induced increase in PGE2. This indicates that the
Meloxicam 0.65 8.99 13.8 stimulated PGE2 synthesis is due to constitutive COX-1
(0.54-0.78) (7.23-10.3) activity. PGE2 production was dose-dependently in-
Diclofenac 5.06 1.64 0.32 hibited by all of the NSAIDs tested in this model.
(3.71-6.62) (1.44-Z05)
Naproxen 12.7 3.56 0.28
Meloxicam had the weakest ability to suppress rat brain
(9.74-16.6) (2.29-4.23) COX-1, followed by tenoxicam which was three times as
Flurbiprofen 2.18 0.14 0.064 potent, indomethacin which was five times as potent
(1.78-2.75) (0.10-O.28) and piroxicam which was 10timesas potent. Diclofenac
'Figures in parentheses are 95% confidence limits. displayed the greatest inhibitory activity against COX-1
and was 20 times as potent an inhibitor as meloxicam
[32].
dose-dependent manner. Meloxicam had a similar In the serum, COX-1 is the isozyme responsible for
potency to diclofenac in suppressing PGE2 excretion. formation of thromboxane (TXA2), which is implicated
Piroxicam and flurbiprofen displayed the greatest in platelet aggregation. The effects of each NSAID on
potency of all agents investigated and were eight times the TXB2 content of rat serum (TXB2 is the stable
as potent as meloxicam [32] (Table II). metabolite of TXA2) were once again dose-dependent.
PGE2 content of the rat gastric juice was lowered by Meloxicam, tenidap and indomethacin were weak
all NSAIDs tested in a dose-dependent manner [32]. inhibitors of TXB2 production compared with
Meloxicam is only a weak inhibitor of PGE2 synthesis tenoxicam and piroxicam. Interestingly, under the same
in the rat gastric mucosa. Diclofenac caused the same experimental conditions, acetylsalicylic acid, an
effects at one-fifth the dosage, whilst flurbiprofen was irreversible inhibitor of platelet COX-1, was still only 15
the most potent with 64 times the inhibitory activity of times less potent than meloxicam [32].
meloxicam in this model (Table III). The effect of NSAIDs on the formation of products
Consequently, NSAID effects on COX-1 mediated resulting from the actions of lipooxygenase on
PG production in the gastric mucosa and kidney have arachidonic acid have also been studied. These products
been compared with their influence on PG synthesis are known as leucotrienes, and we specifically looked at
induced by COX-2 in the rat pleural exudate (Tables II the effects of NSAIDs on the production of two types,
and III) [32]. Such a comparison can be used to LTC4 and LTB4. Notably, meloxicam did not affect
characterize each NSAID with respect to renal and levels of either LTB4 in the pleural exudate or LTC4 in
gastric tolerability at therapeutic doses. The ratio of the the mouse peritoneal exudate at concentrations which
ED50 for inhibition of PGE2 production in the urine to had previously reduced PGE2 formation in each
the ID50 for inhibition in the pleuritic exudate gives an inflammatory model. In contrast, indomethacin and
indication of the relative selectivity of a NSAID for tenidap increased the LTB4 content of pleuritic exudate
inhibiting COX-2 activity over COX-1 in the kidney. Of and tenidap increased the LTC4 content of peritoneal
all the NSAIDs tested, meloxicam showed the greatest exudate in a dose-dependent manner, at doses known to
difference between the doses sufficient to inhibit PGE2 inhibit PGE2 synthesis [32]. Leucotriene C4 is
synthesis in the pleuritic exudate and those necessary to responsible for mediating some of the symptoms of
inhibit urinary PGE2 excretion. For meloxicam this asthma and thus an agent which raises levels could
ratio was 10 times higher than for piroxicam, eight times further exacerbate a pre-existing asthmatic condition.
ENGELHARDT: PHARMACOLOGY OF MELOXICAM

Caution is commonly expressed against the use of Meloxicam's anti-inflammatory activities have been
NSAIDs in asthmatic patients. compared with other, established NSAIDs in a rat
model of progressive and destructive joint disease. In
Anti-inflammatory effects adjuvant-induced arthritis of the rat the acute
Standard animal models of inflammation, including symptoms are related to COX-2 expression [18] and
carrageenan- or kaolin-induced rat paw oedema, inflammation is also immunologically mediated.
granuloma formation following implantation of cotton Meloxicam exhibits greater anti-inflammatory potency
pellets in the raj, carrageenan-induced rat pleurisy and than other compounds tested in this model (Table IV
rat adjuvant-induced arthritis, have been used to and Fig. 3) [33, 34]. At low doses meloxicam prevented
establish the anti-inflammatory effects of meloxicam. not only oedema but also bone and cartilage
In all models meloxicam was able to suppress the destruction. In comparison, piroxicam showed similar
inflammation with a single dose producing a prolonged activity at higher doses whilst diclofenac and tenidap
effect [33]. were only weakly active in preventing bone and
cartilage destruction at doses which suppressed swelling
[34].
TABLE IV As a consequence of nnmunological reaction in rat
Anti-inflammatory potency against adjuvant arthritis (inhibition of adjuvant arthritis, both spleen weight and erythrocyte
swelling provoked by the secondary reaction) and ukerogenic sedimentation rate (ESR) are increased. Meloxicam was
potency in the rat able to diminish the observed increase in spleen weight
Adjuvant Stomach and reduce elevated ESR dose dependently. Piroxicam
arthritis ID»- ukeration ED» Ratio was only effective at higher doses, with both diclofenac
NSAID (mg/kg/day) (mg/kg/day) ulcus/arthritis and tenidap showing no activity at doses sufficient to
Meloxicam 0.12 2.42 20 reduce swelling [34]. Consequently, only meloxicam,
(0.09-0.14)* (1.64-3.56) at low doses, was able to antagonize immunologically
Diclofenac 1.23 2.71 2.2 mediated effects
(0.84-2.76 (2.38-3.09) Meloxicam shows anti-exudative effects which are
Piroxicam 0.77 1.09 1.4 characteristic of all cyclooxygenase inhibitors tested in
(0.46-1.71) (0.26-1.41)
Naproxen 11.8 11.2 0.95
rat paw oedema models. Meloxicam displays anti-
(8.1-14.9) (8.1-15.4 exudative activity in carrageenan-induced oedema with
Fhirbrprofen 0.97 071 072 a potency, at a single oral dose of 1 mg/kg, exceeding
(0.55-2.16) (0.15-0.30) that of piroxicam, indomethacin, diclofenac, naproxen
Acetlysaticylic 198 32.4 0.16 and acetylsalicylic acid (Table V). In addition, meloxi-
acid (169-245) (21.1-49.7) cam has dose-dependent effects against kaolin-induced
•Figures in parentheses are 95% confidence limits oedema which are of a similar potency to piroxicam,

100-

90-

80

8 70-

60-
c
S 60
z ONtoloxtcani
• Pfrexfcam
40-
A Diclofenac
30- A Indomethadr
ONaproxan
20- • AcatytoaJlcydkjacW
10- I I I III nri|
.01 .1 10 100 1000
Dally oral dose [mg/kg]

Fio. 3.—Inhibition of adjuvant-induced specific secondary reaction (oedema of the contralateral hind paw) in the rat after dairy oral administration
for 21 days.
STUDIES ON MELOXICAM (MOBIQ

indomethacin and diclofenac and higher than those and the ulcerogenic dose. For currently established
of tenoxicam, tenidap and naproxen (Table VI). NSAIDs this therapeutic margin is quite narrow such
Meloxicam's anti-inflammatory activity is still apparent that anti-inflammatory potency does not strictly
in adrenalectomized rats, thus indicating that this correlate with gastrointestinal tolerance. For example,
effect is not mediated by endogenous corticosteroids. at therapeutic doses, indomethacin and piroxicam are
However, meloxicam, like other NSAIDs, has no effect associated with a higher risk of gastrointestinal toxicity
on egg white-induced oedema which is mediated by than other NSAIDs [3, 40, 41].
histamine [33]. It is apparent that the complex pathogenesis of
In another inflammatory model of carrageenan- stomach ulcerations accompanying NSAID therapy is
induced pleurisy of the rat, meloxicam was able to directly related to the inhibition of the biosynthesis of
inhibit both exudate formation and polymorphonuclear cytoprotective prostaglandins in the gastric mucosa.
leucocyte immigration, once again in a dose-dependent Specifically, PGE2 and PGI2 protect the mucous
fashion. Piroxicam only showed similar effects at doses membrane and inhibit acid secretion in the stomach.
which were four times greater than equivalent doses of As we have seen, meloxicam is a weak inhibitor of
meloxicam [34]. PGE2 production in the rat stomach (Table III).
Furthermore, meloxicam is a much less potent
Analgesic effects stimulant of acid secretion in the rat stomach than, for
Meloxicam, in accordance with findings for diclo- example, piroxicam and indomethacin [42].
fenac, indomethacin, piroxicam and naproxen, has no Meloxicam shows weak gastric ulcerogenicity in the
effect on either heat-induced (the hot-plate technique) rat stomach, in contrast to its potent anti-inflammatory
[35] or mechanically-induced (the tail clamp test) [36] efficacy (Table IV). These characteristics are parti-
pain in the mouse or on the visceral painreflexin the rat cularly striking when viewed in the context of ratios
[37]. Therefore, it can be assumed that meloxicam has obtained for other NSAIDs in the same model.
no central analgesic effects [33]. Specifically, the therapeutic range displayed by mel-
Against inflammatory pain, measured according to oxicam in the rat is 10-90 times greater than that of
Randall and Selitto [38] in the rat, meloxicam showed other commonly used NSAIDs [24].
a very prolonged effect. Following a single oral Thus, it is tempting to suggest that based on the
administration the analgesic effect of meloxicam is not unusual parity of meloxicam's pharmacokinetic data in
reduced by 50% until 18 hours after administration. rat and man, that pharmacodynamic and toxicological
Meloxicam has a markedly longer duration of action similarities may also exist [43]. However, such
than piroxicam, diclofenac and indomethacin. judgements can only be made following the results of
clinical investigation.
Antipyretic effects
Unlike paracetamol and phenazone derivatives, Effects on renal function
meloxicam and all other NSAIDs have no influence on In a standard model used to assess the effect of
the body temperature of a normothermic mammal, NSAIDs on renal function, meloxicam, at doses of up
because NSAIDs have no direct effect on the calorific to 16 mg/kg, has no influence on water and electrolyte
centre. NSAIDs are only effective against pyrogen- excretion. In contrast, phenylbutazone displays a potent
induced fever. Meloxicam shows a lower potency inhibitory effect in the same model using water and
against yeast-induced pyrexia than diclofenac and electrolyte-loaded rats [44].
piroxicam [33]. At a dose of 0.1 mg/kg meloxicam
reduces endotoxin-induced fever in the cat [39]. Effects on cartilage metabolism
It has been suggested that treatment with NSAIDs
Gastric tolerance can contribute to cartilage deterioration [45], but there
It is widely accepted that gastric ulcerogenicity is the is no evidence that meloxicam causes this problem. In
dose-limiting side-effect common to all established long-term studies in rats and mice meloxicam has been
NSAIDs. Therefore, the ability to achieve a good
therapeutic response is determined by the difference
between the dose required to obtain the desired effects TABLE VI
Inhibition of hind paw oedema of rats induced by kaolin 5.5 h after
oral administration
TABLE V
Inhibition of carragecnan-induced oedema in the rat hind paw: Confidence limits Regression
measurement of the effect of single oral doses by AUC Drug ID35 (mg/kg) (95%) coefficient

NSATD AUC (% inhibition x h)/ing/kg Meloxicam 3.35 2.93-3.91 34.5


Piroxicam 2.71 2.30-3.25 30.8
Meloxkam 254 Tenoxicam 8.35 6.87-10.7 29.0
Piroxicam 144 Tenidap 6.06 4.37-13.1 44.5
Indomethacin 84 Indomethacin 3.42 2.39-4.49 31.4
Diclofenac 59 Diclofenac 4.03 3.01-5.08 20.8
Naproxen 44 Naproxen 6.25 5.46-7.11 34.1
Acetylsalkylic acid 1.6 Acetylsalkylic acid 318 251-542 29.5
ENGELHARDT: PHARMACOLOGY OF MELOXICAM

TABLE VH
Mean pharmacokisetic parameters of meloxicam in rat and man
CatxM (mg/ml) Cl (ml/min/kg) Plasma-protein
Species Dose (mg/kg/day) MVISJJ. MVISJX MViiD. binding (%) Ref.
Rat* 11 x 1.0 6.3 ± 0.76 0.1110.04 15.516.2 99.5-99.7 [43]
Man 7x0.11 0.88 ± 0.20 0.1110.03 20.416.4 99.5-99.7 [57]
•Male rat.

established as chondroneutral towards arthrotic ero- vascular side-effects in a number of species [42]. Even at
sions [46]. Meloxicam neither increased nor inhibited high oral doses, meloxicam had no significant effect on
the development of spontaneously occurring osteo- systolic pressure of the conscious rat. When adminis-
arthrotic changes in rats and mice. These Findings tered intraduodenally to anaesthetized cats, very high
correlate with the results of in vitro studies [47, 48] dose levels of meloxicam (100 and 200 mg/kg) caused a
which showed that meloxicam, in contrast to indo- slight, but not significant, reduction in mean blood
mcthacin and acetylsalicylic acid, does not influence the pressure together with a slight decrease in heart rate.
synthesis and degradation of proteoglycans by human Even after high doses, meloxicam had no effect on
chondrocytes. respiratory minute volume. When administered intra-
venously to anaesthetized cats, meloxicam had no effect
on systolic pressure, diastolic pressure, carotid artery
Uricosuric effects flow, heart rate, electrocardiogram or respiratory
Many NSAIDs are able to increase uric acid volume at doses of up to 4 mg/kg. Oral doses of
excretion independently of their anti-inflammatory 2-8 mg/kg had no effect on mean arterial pressure in
activity [49]. This uricosuric effect has been demon- conscious dogs [56].
strated by piroxicam in patients [50, 51]. In rats, where
the uric acid level had been raised through treatment In conscious rats, oral doses of meloxicam up to
with oxonic acid, meloxicam showed a weaker 32 mg/kg had no significant effect on intestinal transit
uricosuric effect than piroxicam [33]. time [44]. Meloxicam remains free of any effect on
stomach emptying in conscious rats at doses up to an
oral dose of 32 mg/kg [44].
Influence on bronchial muscle Meloxicam does not affect carbohydrate metabolism.
NSAIDs are effective inhibitors of bradykinin- There were no changes in blood glucose levels in rabbits
induced bronchospasm in guinea-pigs [52]. The when meloxicam was administered orally at doses up to
bronchospastic effect of bradykinin is an indirect effect, 4 mg/kg [56].
mediated by TXA2 liberated in the lung [53]. After Meloxicam had no influence on thromboplastin time
intraduodenal administration, doses of meloxicam in the rat when given orally at doses up to 8 mg/kg, on
which were effective against bradykinin-induced bron- two consecutive days [56].
chospasm in guinea-pigs were in the same range as In in vitro studies carried out on isolated organs of
those shown by indomethacin [33]. guinea-pigs and rats, meloxicam was found to have no
Unlike piroxicam and diclofenac, even at very low anticholinergic, papaverine-like, Hi-antagonist, angio-
intravenous doses, meloxicam had an effect on PAF- tensin II-antagonist, PGE2-antagonist, serotonin-
induced bronchospasm in the guinea-pig. PAF-induced antagonist or bradykinin-antagonist properties at
bronchospasm is mediated by TXA2 [54] and LTC4 [55]. concentrations up to 10 ug/ml in a protein-free medium
Even at high doses, meloxicam has no broncholytic or [53].
bronchoconstrictive effect on acetylcholine-induced
bronchospasm in the guinea-pig [33]. In summary, the general pharmacology studies did
not reveal any pharmacodynamic effects of meloxicam
which would restrict its therapeutic use as an NSAID.
General pharmacology
A number of studies have been carried out in mice to
examine the effect of meloxicam on CNS functions. Pharmacodynamic interactions
Oral doses of meloxicam of up to 25 mg/kg did not The pharmacokinetic profile of meloxicam in the rat
affect reflexes or sensory functions and had no effect on (especially in male rats, commonly used in phar-
spontaneous motility; nor did it affect hexobarbital- macological tests) is very similar to its pharmacokinetic
induced sleeping time. No muscle-relaxant activity was profile in man (Table VII) [43, 57]. Consequently, most
seen at pharmacologically relevant doses. Meloxicam of the studies designed to detect interactions between
also has no anticonvulsant properties. It did not affect meloxicam and other drugs have been carried out in the
pentetrazole-induced shock or maximal electric shock rat.
at doses up to 50 mg/kg, nor did it enhance or reduce Concomitant administration with paracetamol
the anticonvulsant effect of phenobarbitone on mice enhances the effect of meloxicam on inflammatory pain
subjected to electric shock [44]. in the rat The doses of paracetamol used had only very
Meloxicam has been tested for possible cardio- weak analgesic activity [56].
10 STUDIES ON MELOXICAM (MOBIQ

Similarly, paracetamol enhances the acute anti- meloxicam has a stronger and much more sustained
exudative effect of meloxicam in carrageenan-induced anti-inflammatory effect than piroxicam, diclofenac
paw oedema in the rat [56]. and indomethacin. More importantly, meloxicam has a
Concomitant administration of pirenzepine with much greater potency in the rat model of progressive
meloxicam minimizes the ulcerogenic effect of meloxi- and destructive joint disease, adjuvant arthritis,
cam in the stomach of the rat in a dose-dependent preventing not only oedema but also bone and cartilage
manner [56]. destruction. Meloxicam's superior antiarthritic activity
Meloxicam does not reduce the increase in water and may be due to both its high potency against COX-2 [18]
electrolyte excretion which is seen in rats after and its accumulation in inflamed tissue [60].
administration of chlortalidone [56]. In contrast to conventional NSAIDs, anti-
At very high doses (4 mg/kg and above), meloxicam inflammatory doses of meloxicam inhibit leucocyte
augments the increase in prothrombin time which is migration. Like other NSAIDs, meloxicam does not
caused by phenprocoumon in the rat [56]. show true central analgesic effects. However, this is not
Meloxicam does not influence the effect of tol- relevant with regard to its effect on inflammatory pain.
butamide on blood glucose levels in the rabbit [56]. Meloxicam has no direct influence on the caloric
centre and only weak effects against pyrogen fever. It
Pharmacodynamic effects of metabolites of meloxicam has a mild uricosuric effect. Thus meloxicam is not an
Four principal metabolites of meloxicam have been alternative to conventional antipyretic or uricosuric
identified in rats and humans. These metabolites are drugs.
rapidly excreted in the urine and are, therefore, not Meloxicam, in contrast to indomethacin and acetyl-
detectable in the plasma [58]. salicylic acid, is chondroneutral.
The metabolites did not show any inhibitory effects As meloxicam does not affect water and electrolyte
on COX or display any anti-inflammatory or analgesic excretion and has only a weak influence on intrarenal
activities in the rat following oral administration [59]. PGE2 biosynthesis, it would not be expected to
Doses up to 10 times higher than active doses of influence kidney function at therapeutic doses. The
meloxicam were administered in these studies. ulcerogenic potency of meloxicam on the stomach in
relation to the anti-inflammatory potency is weak.
Pharmacological and toxicological findings suggest that
CONCLUSIONS meloxicam is not associated with any side-effect un-
Using models of chronic inflammation in related to its mechanism of cyclooxygenase inhibition.
conjunction with a model of gastric damage, meloxicam Only the native compound has a pharmacodynamic
has been identified as a NSAID with greater anti- activity; the metabolites of meloxicam are inert.
inflammatory activity than existing drugs and low The pharmacokinetic behaviour of meloxicam in the
toxicity in the stomach. The compound has now been rat [43] is very similar to that observed in man [61].
shown to be a preferential inhibitor of COX-2.
Therefore, pharmacodynamic and toxicological find-
This review has focused on presenting the pharmaco- ings with meloxicam established in the rat [37] can be
dynamic activity of meloxicam and has concentrated on extrapolated, with confidence, to man.
its effects on the classical mediators of inflammation
which are the common site of action for all NSAIDs. In clinical study, analysis of the safety data from over
Furthermore, meloxicam's action has been compared 4000 patients suggests that meloxicam is fulfilling its
with that of well established conventional NSAIDs. early promise from pre-clinical studies [62]. In
This has enabled us to show a clear differentiation particular, analysis of upper Gl perforations,
between meloxicam's anti-inflammatory profile and ulccrations and bleeding indicates a much lower
that of other NSAIDs in current clinical use. These likelihood of occurence in those patients treated with
effects have been demonstrated in a variety of cell types meloxicam 7.5 mg and 15 mg once daily than with
and tissues. therapeutically equivalent doses of piroxicam,
The basis of meloxicam's superior risk-benefit profile diclofenac and naproxen.
over existing NSAIDs (e.g. piroxicam, diclofenac, For the new compounds described to be selective
naproxen, flurbiprofen) can be explained by its selective COX-2 inhibitors (DUP-697 [63], SC-58125 [64],
inhibition of COX-2 in preference to COX-1. However, NS-398 [65], L-745,337 [66]) unfortunately clinical data
there are other differences in the pharmacodynamic are not available. An improved gastric tolerance of
profile of meloxicam in comparison with other flosulide has been confirmed in humans [67]. The
NSAIDs. Anti-inflammatory doses of meloxicam do claimed selectivity of nimesulide for COX-2 [68] is not
not influence lipoxygenase activity. Meloxicam does not proved by the cited [69] data.
increase LTC4 content of tissue and has no influence on In summary, in thisreviewwe show that meloxicam is
bronchial tone. That means that the risk of a NSAID which has the potential to redefine our
bronchoconstriction may be lower with meloxicam than expectations of risk-benefit ratio associated with
with known NSAIDs. current NSAID treatment of inflammatory disease. The
Like other NSAIDs, meloxicam displays classical explanation of the exceptional pharmacological profile
anti-inflammatory, antipyretic and analgesic properties. of such an agent has been enabled through new insights
However, in the model of an acute oedema in the rat, into the mode of action of NSAIDs.
ENGELHARDT: PHARMACOLOGY OF MELOXICAM 11

REFERENCES M. Induction of cyclooxygenase by interleukin-1: com-


Vase JR. Inhibition of prostaglandin synthesis as a parative study between human synovial cells and
mechanism of action for the aspirin-like drugs. Nature chondrocytes. J Rheumatol 1994^1:462-6.
New 5to/1971^31|232-5. 20. Angel J, Berenbaum F, Le Deumat C, Nevalainen T,
Vane JR. In: Robinson HJ, Vane JR, eds. Prostaglandin Masliah J, Fournier C. Interleukin-1-induced pro-
synthetase inhibitors. New York: Raven Press, 1974. staglandin E2 biosynthesis in human synovial cells involves
T-angman FJS, Weil J, Wainwright P et al. Risk of bleeding the activation of cytosolic phospholipase A2 and cyclooxy-
peptic nicer associated with individual non-steroidal anti- genase-2. EurJBiochem 1994^26:125-31.
inflammatory drugs. Lancet 1994^43:1075-8. 21. Crofford LJ, Wilder RL, Ristimaki AP et al. Cyclooxy-
Fu J-Y, Masferrer JL, Seibert K, Raz A, Necdleman P. The genase-1 and -2 expression in rheumatoid synovial tissue.
induction and suppression of prostaglandin H2 synthase Effects of interleukin-1 p, phorbol ester and corucoids.
(cyclooxygenase) in human monocytes. J Biol Chem / Clin Invest 1994^3:1095-101.
1990^65:16737-40. 22. Meade EA, Smith WL, DeWitt DC. Differential inhibition
Masferrer JL, Zweifel BS, Seibert K, Needleman P. of prostaglandin endoperoxide synthase (cyclooxygenase)
Selective regulation of cellular cyclooxygenase by dex- isozymes by aspirin and other non-steroidal anti-inflam-
amethasone and endotoxin in mice. J Clin Invest matory drugs. JBiol Chem 1993;268.-6610-4.
1990;86:1375-9. 23. Mitchell JA, Akarasereemont P, Thiemermann C, Flower
Masferrer JL, Seibert K, Zweifel B, Needleman P. Endo- RJ, Vane JR. Selectivity of nonsteroidal anti-mflamma-
genous glucocorticoids regulate inducible cyclooxygenase tory drugs as inhibitors of constitutive and inducible
enzyme. Proc NatlAcadSci USA 1992;89:3917-21. cyclooxygenase. Proc Natl Acad Sci USA 1993;9O:
Wong WYL, Richards JS. Evidence for two antigenicaUy 11693-7.
district molecular weight variants of prostaglandin H syn- 24. Engelhardt G. Meloxicam inhibits preferentially COX-2.
thase in the rat ovary. Mol Endocrinol 1991^:1269-79. Eur J Clin Pharmacol 1994;47:A98.
Xie W, Chipman JG, Robertson DL, Erikson RC, Sim- 25. Engelhardt G, Bogel R, Schnitzler C, Utzmann R. Meloxi-
mons DL. Expression of a mitogen-responsive gene cam: influence on arachidonic acid metabolism. Part I. In
encoding prostaglandin synthase is regulated by RNA vitrofindings.Biochem Pharmacol 1996£l:21-8.
splicing. Proc NatlAcadSci USA 1991;8&2692-8. 26. Churchill L, Graham A, Farina P, Grab P. Inhibition of
Loll PJ, Garavito RM. The isofonns of cyclooxygenase: human cyclooxygenase-2 (COX-2) by meloxicam. XJUth
structure and function. Exp Opin Invest Drugs 1994;3: European Congress of Rheumatology, Amsterdam, June
1171-80. 18-23,1995.
10. 27. Pairet M, Engelhardt G. The preferential inhibition of
11. Lee SH, Soyoola E, Chanmugam P et al. Selective expres- COX-2 by meloxicam is highly dependent on the structure
sion of mitogen-indutible cyclooxygenase in macrophages of the drug. Rheumatol Eur 1995;24(suppl. 3):272.
stimulated with lipopolysaccharide. J Biol Chem 28. Murray MD, Brater DC. Renal toxicity of non-steroidal
1992^67^5934-8. anti-inflammatory drugs. Anna Rev Pharmacol Tox
12. O'Sulhvan MG, Chilton FH, Huggjns EM, McCall CE. 199332:435-65.
Lipopolysaccharide priming of alveolar macrophages for 29. Rosenkranz B, Fischer C, Meese CO, FrSlich JC. Effects
enhanced synthesis of prostanoids involves induction of a of salicylic and acetylsalicylic acid alone and in combina-
novel prostaglandin H synthetase. / Biol Chem 1992; tion on platelet-aggregation and prostanoid synthesis in
267:14547-50. man. Br J Clin Pharmacol 1986^1:309-17.
13. Akarasereenont P, Mitchell JA, Bakhle YS, Thiemermann 30. Whittle BJR, Higgs GA, Rakins KE, Moncada S, Vane
C, Vane JR. Comparison of the induction of cyclooxy- JR. Selective inhibition of prostaglandin production in
genase and nitric oxide synthase by endotoxin in inflammatory exudates and gastric mucosa. Nature
endothelial cells and macrophages. Eur J Pharmacol 1980^84^71-3.
1995^73:121-8. 31. Harada Y, Hatawaka K, Kawamura M et al. Possible
14. Masferrer JL, Reddy ST, Zweifel BS, Seibert K, Needle- involvement of inducible cyclooxygenase (COX) in
man P. In vivo glucocorticoids regulate cyclooxygenase-2 carrageenan-induced pleurisy. World Congress of Inflam-
but not cyclooxygenase-1 in peritoneal macrophages. J mation 1993, Vienna, October 10-15, 1993;abstract no.
Pharmacol Exp Ther 1994^70:1340-^. P153.
15. Vane JR, Mitchell JA, Appleton J et al. Inducible isofonns 32. Engelhardt G, Bogel R, Schnitzler C, Utzmann R. Meloxi-
of cyclooxygenase and nitric synthase in inflammation. cam: influence on arachidonic acid metabolism. Part II. In
Proc Natl Acad Sci USA 1994^1^046-50. vivofindings.Biochem Pharmacol 1996^1:29-38.
16. Habib A, Creminon C, Frobert Y, Grassi J, PradeDes P, 33. Engelhardt G, Homma D, Schlegel K, Utzmann R,
Maclouf J. Demonstration of an inducible cyclooxygenase Schnitzler C. Anti-inflammatory, analgesic, antipyretic
in human endothelial cells using antibodies raised against and related properties of meloxicam, a new non-steroidal
the carboxyl-terminal region of the cyclooxygenase-2. J anti-inflammatory agent with favourable gastrointestinal
Biol Chem 1993;26fc23448-54. tolerance. Inflamm Res 1995;44:423-33.
17. Szcepanski A, Moatter T, Carley WW, Gerritsen ME. 34. Engelhardt G, Homma D, Schnitzler C. Meloxicam: a
Induction of cyclooxygenase II in human synovial potent inhibitor of adjuvant arthritis in the Lewis rat. In-
microvessel endothelial cells by interleukin-1. Arthritis flamm Res 1995;44:548-55.
Rheum 1994^7:495-503. 35. Chen JYP, Beckman H. A satisfactory apparatus for study
18. Sano H, Hla T, Maier JAM et al. In vivo cyclooxygenase of analgesia in mice. Science 1951;113:631.
expression in synovial tissues of patients with rheumatoid 36. Haffner F. Experimentelle Priifung schmerzstillender
arthritis and osteoarthritis and rats with adjuvant and Mittel. Dtsch Med Wochenschr 1929^4:731-3.
streptococcal cell wall arthritis. / Clin Invest 1992; 37. Lembeck F, Skofitsch G. Visceral pain reflex after
8937-108. pretreatment with capsaicin and morphine. Naunyn-
19. Knott I, Dieu M, Burton M, Houbion A, Remade J, Raes Schmiedeberg's Arch Pharmakol 1982;312;116-22.
12 STUDIES ON MELOXICAM (MOBIQ

38. Randall LO, Selitto JJ. A method for measurement of pig lungs involves mechanisms distinct from those for leuk-
analgesic activity or inflammation tissue. Arch Int Pharma- otriene. Br J Pharmacol 1984;82:565-75.
codyn 1957;111:409-19. 55. Voclkel NF, Worthen S, Reeves JT, Henson PM, Murphy
39. Justus C, Quirke JF. Dose response relationship for the RC. Non-immunological production of leukotrienes
antipyretic effect of meloxicam in an endotoxin model in induced by platelet-activating factor. Science
cats. Veterin Res Commun 1995;19321-30. 198231&286-8.
40. Rodriguez LAG, Jick H. Risk of upper gastrointestinal 56. Engelhardt G, Homma D, Schlegel K, Schnitzler Chi,
bleeding and perforation associated with individual non- Utzmann R. General pharmacology of meloxicam. Part.
steroidal anti-inflammatory drugs. Lancet 1994;343: 2. Gen Pharmacol 1995;in press.
769-72. 57. Turck D, Busch U, Heinzel G, Greischel A, Nehmiz G,
41. Kaufman DW, Kelly JP, Sheeman JE et al. Non-steroidal Narjes H. The basic cUnical pharmacokinetics of meloxi-
anti-inflammatory drug use in relation to major upper gas- cam, a new NSAID. Scand J Rheumatol 1994;Suppl
trointestinal bleeding. Clin Pharmacol Ther 1993^3: 98:A120.
485-94. 58. Schmid J, Busch U, Heinzel G, Bozler G, Kaschke S,
42. Engelhardt G, Bogel R, Krug I, Walcher I. Meloxicam: Kummer M. Meloxicam: pharmacokinetics and metabolic
lack of correlation between inhibition of PG-biosynthesis pattern after intravenous infusion and oral administration
in the inflammatory field and in the stomach. Magy to healthy subjects. Drug Metab Dispos 1995^3:1206-13.
Rheumatol 1991;32(suppl.):354. 59. Engelhardt G, Trummlitz G. Biological activity of the
43. Busch U. The pharmacokinetics of meloxicam in animals. main metabolites of meloxicam. Drugs Exp Clin Res
Scand J Rheumatol 1994;Soppl9&A119. 1990;16:53-6.
44. Engelhardt G, Homma D, Schlegel K, Schnitzler C, 60. Busch U, Engelhardt G. Distribution of [14C]meloxicam in
Utzmann R. General pharmacology of meloxicam. Part 1. joints of rats with adjuvant arthritis. Drugs Exp Clin Res
Gen Pharmacol 1996:51:29-38. 1990,17:49-52.
45. Ghosh P, Brooks P. Chondroprotection—exploring the 61. Busch U, Schmid J. Pharmacokinetic properties of meloxi-
concept. J Rheumatol 1991;18:161-6. cam: a new NSAID in different species. Clin Exp
46. Mohr W, Lehmann H. Chondroneutrality of meloxic.'~:-. Rheumatol 1987;5(suppl. 2): 145.
spontaneously occuring osteoarthrosis in rats and mice. 62. Distel M, Mueller C, Bluhmki E, Fries J. Safety of meloxi-
Congress of Degenerative and Inflammatory Processes in cam: a global analysis of clinical trials. Br J Rheumatol
the Joints, Bari, Italy, September 29-October 1,1994. 1996;35(suppl. 1): 68-77.
47. Rainsford K, Ying C, Smith F. Comparative effects of 63. Copeland RA, Williams JM, Giannaras J et al. Mecha-
meloxicam on cartilage-synovial components of joint nism of selective inhibition of the inducible isofonn of
injury in vitro. 2nd International Congress of the prostaglandin G/H synthase. Proc Natl Acad Sci USA
Osteoarthritis Research Society, Orlando, Florida, 1994,-91:11202-6.
December 9-11,1994.
48. Bassleer C, Franchimont P. Effects of meloxicam 64. Seibert K, Zhang Y, Leahy K et al. Pharmacological and
compared to acetyl salicylic acid in human articular biochemical demonstration of the role of cyclooxygenase
chondrocytes. XOTth European Congress of Rheumatol- 2 in inflammation and pain. Proc Natl Acad Sci USA
ogy, Amsterdam, June 18-23,1995:abstractno. E46. 1994:91:12013-7.
49. Swingle KF. Evaluation of anti-inflammatory activity. In: 65. Gierse JK, HauseT SD, Creely DP et al. Expression and
Scherer RA, Whitehouse MW, eds. Anti-inflammatory selective inhibition of the constitutive and inducible forms
agents. New York: Academic Press, 1974. of human cyclo-oxygenase. Biochem J 1995^05:479-84.
50. Tausch G, Eberl R. Efficacy, tolerance and safety of pi- 66. Chan CC, Gordon R, Brideau C et al. In vivo pharmacol-
roxicam in the treatment of acute gout. Eur J Rheumatol ogy of L-745,337: a novel non-steroidal anti-inflammatory
Inflamm 1978;l:365-8. agent (NSAID) with an ulcerogenk sparing effect in rat
51. Murphy JE. Piroxicam in the treatment of acute gout. J and monkey stomach. Can J Physiol Pharmacol 1994;
Intern Med Res 1979;7:507-10. 72(suppl. 1)^66.
52. Collier HOJ, Hogate JA, Schachter M, Shoriey PG. An ap- 67. Bjamason I, Hayllar J, Parker J, Schupp J, MacPherson A.
parent bronchoconstrictor action of bradykinin and its A randomised, double blind, crossover comparative
suppression by some anti-inflammatory drugs. / Physiol endoscopy study on the gastroduodenal tolerability of
1959;149:54P-5P. flusolide and naproxen. Gastroenterology 1994;106(suppl.
53. Hamberg M, Svensson J, Samuelsson B. Thromboxanes: a 4):A53.
new group of biologically active compounds derived from 68. Bennett A, Tavares IA. NSAIDs, COX-2 inhibitors, and
prostaglandin peroxides. Proc Natl Acad Sci USA the gut. Lancet 1995;346:1105.
1975;72:2994-8. 69. Tavares IA, Bishai PM, Bennett A. Activity of nimesuh'de
54. Lefort J, Rotilio D, Vergaftig BB. The platelet-independent on constitutive and inducible cyclooxygenases. Arzneimit-
release of thromboxane A2 by PAF-acether from guinea- telForsch 1995;45:1093-5.

Das könnte Ihnen auch gefallen