You are on page 1of 12

Pain Physician 2009; 12:269-280 • ISSN 1533-3159

Back to Basics

Potential Analgesic Mechanisms of


Acetaminophen

Howard S. Smith, MD

From: Albany Medical College,


Albany, NY.
Despite nearing the end of the decade of pain research, the analgesic mechanisms
Dr. Smith is Associate Professor of one of the most widely used and popular analgesics remains uncertain.
and Academic Director of Acetaminophen (APAP) (paracetamol [PARA]) has been used clinically for over a
Pain Management for Albany half of a century and although clinicians seem to be comfortable with its benefits,
Medical College Department of risks, and limitations, they still remain in the dark as to precisely what is providing its
Anesthesiology, Albany, NY. pain relief. What does seem clearer is that the predominant mechanisms of APAP’s
analgesic effects are in the central nervous system (CNS).
Address correspondence:
Howard S. Smith, MD
Albany Medical College Although, which central effects are largely responsible for APAP’s effects on pain
Department of Anesthesiology continue to be uncertain. Perhaps, the most accepted theory is that of APAP’s
47 New Scotland Avenue; MC-131 positive effects on the serotonergic descending inhibitory pathways. However,
Albany, New York 12208 interactions with opioidergic systems, eicosanoid systems, and/or nitric oxide
E-mail: smithh@mail.amc.edu
containing pathways may be involved as well.
Disclaimer: There was no external
funding in the preparation of this Furthermore, endocannabinoid signaling may play a role in APAP’s activation of the
manuscript. serotonergic descending inhibitory pathways. A greater understanding of APAP’s
Conflict of interest: None. analgesic mechanisms may promote optimal utilization of analgesic polypharmacy.
Manuscript received: 12/10/2008
Key words: Acetaminophen (APAP), paracetamol (PARA), pain, analgesia,
Accepted for publication:
12/31/2008 mechanisms of action, serotonin, opioids, endocannabinoids

Free full manuscript: Pain Physician 2009; 12:1:269-280


www.painphysicianjournal.com

A cetaminophen (APAP) continues to be one


of the most popular analgesics available
in the United States despite that the
mechanisms of antinociception elicited by APAP are
not yet elucidated with certainty.
Mering in 1887 (2). However, during the period when
phenacetin was popular, its use was dormant. The
studies of Brodie and Axelrod (3) led to its “rediscov-
ery” and marketing in the 1950s in the United States
as an analgesic replacement for phenacetin, which
Although paracetamol (PARA) appears to be the was abandoned due to its nephrotoxicity. APAP be-
recommended international nonproprietary name of came available in the United States in 1955 and in the
APAP, acetaminophen (or APAP) will be used in this United Kingdom the following year. Unfounded con-
article since the term “paracetamol” is not widely rec- cerns about APAP’s safety delayed its widespread ac-
ognized in the United States. APAP was synthesized ceptance until the 1970s (4). APAP remains the most
in 1878 by Morse (1) and first used clinically by von popular analgesic/antipyretic used in children.

www.painphysicianjournal.com
Pain Physician: January/February 2009:12:269-280

Potential Mechanisms of APAP- as COX-1 and COX-2 (8). However, the nomenclature
Induced Analgesia
PGHS is preferred because there are 2 active sites
Although suspected for many years, it is now be- on this enzyme: a COX site and a POX site. The ac-
coming clearer that the mechanisms, which are largely tivity of the COX enzyme relies on its being in the
responsible for APAP’s analgesic effects are largely oxidized form and it is suggested that APAP reduces
central in origin. the amount of the oxidized form by an action on the
Crawley et al (5) studied the efficacy of systemic POX site (9). An alternative suggestion is that a PGHS
(oral) and intrathecal (IT) applications of APAP in pre- variant (COX-3) exists in the central nervous system
venting the development of hyperalgesia induced (CNS), and that this variant is exquisitely sensitive to
through the direct activation of pro-algesic spinal APAP (10).
receptors. Spinal administration of substance P (SP, Rezende et al (11) concluded that although both
30 nmol, IT) in rats produced a decreased thermal APAP and dipyrone are inhibitors of COX isoforms and
threshold, indicating centrally mediated hyperalge- thus of prostaglandin biosynthesis and were analgesic
sia. Pretreatment of rats with oral APAP (300 mg/ in our model, their analgesic actions were functionally
kg), but not vehicle, significantly attenuated IT SP- and mechanistically different.
induced hyperalgesia. APAP given IT also produced The involvement of prostaglandins (PGs) in the
a dose-dependent (10 – 200 µg) antinociceptive ef- analgesic mechanisms of action of APAP has been
fect. In addition, oral APAP suppressed spinal PGE2 proposed, taking into account the inhibition of the
release evoked by IT SP in an in vivo IT dialysis model. central cyclo-oxygenases (COX-1, COX-2, and COX-3)
The ability of IT as well as oral APAP to reverse this exerted by this drug (10,12-15), although the results
spinally initiated hyperalgesia emphasizes the likely are controversial in this regard (16).
central action and bioavailability of the systemically Despite limitations with the belief that COX-3 may
delivered drug (5). be the site of APAP action, it has been suggested that
For the systemic route of delivery, the observation there may be varied products from the 2 distinct COX
that APAP reversed that centrally mediated hyperal- proteins with overlapping contributions to prostanoid
gesia is consistent with its known ability to penetrate production throughout the body (17).
into the brain (6) at a dose which failed to alter the Much investigation has centered on APAP’s inhibi-
acute thermal threshold (5). This emphasizes that the tion of the COX enzyme because its analgesic and an-
site of systemic drug action was within the neuraxis tipyretic effects are similar to those of aspirin, the ar-
via mechanisms that mediate spinal sensitization (5). chetype non-steroidal anti-inflammatory drug (NSAID)
Similarly, the oral dose of APAP required to produce (18). However, APAP does not have significant anti-in-
a central antihyperalgesic effect was 1,000 times the flammatory activity nor does it inhibit production of
dose required when administered intrathecally. It is the pro-clotting TXAs. It seems reasonable to assume
therefore unlikely that the spinal effect of the IT drug that although there may be some APAP effect on COX
effect was due to redistribution of the drug into the enzymes, this effect may be different from that seen
periphery (5). Multiple mechanisms may contribute to with the NSAIDs (18).
the analgesia provided by APAP. The conversion of arachidonic acid to PGG2 is
The results of Seo et al (7) suggest that LPS-in- dependent on the tyrosine-385 radical (Tyr385*) at
duced hyperalgesia in the formalin second phase may the COX site (19). However, generation of this radi-
be involved in the SP-sensitive neuronal pathways, in cal from Tyr385 is reliant on generation of a ferryl
which the hyperalgesic response elicited by LPS is at- protoporphyrin IX radical cation (Fe4+ =OPP*+) at the
tenuated by APAP with supraspinal pain modulatory POX site. APAP interferes with this process by acting
mechanisms. as a reducing cosubstrate in a reaction that partially
reduces Fe4+ = OPP*+ so that less Fe4+ = OPP*+ is avail-
APAP and the Eicosanoid System able to be transferred to the COX site. Consequently,
Prostaglandin H2 synthetase (PGHS) is the en- less Tyr385* is available to stimulate conversion of ar-
zyme responsible for metabolism of arachidonic acid achidonic acid to PGG2 (9,20). APAP acts with greater
to the unstable PGH2. The 2 major forms of this en- efficacy in environments with low peroxide tone and
zyme are the constitutive PGHS-1 and the inducible low arachidonic acid levels, such as what exists within
PGHS-2. These 2 enzymes are commonly referred to the CNS (13).

270 www.painphysicianjournal.com
Potential Analgesic Mechanisms of Acetaminophen

Studies have shown that therapeutic concentra- actions of APAP is still a matter of debate (16,33,34), it
tions of APAP have inhibited PG synthesis in brain ho- seems that there is at least some contribution from the
mogenates (12) and ram seminal vesicle microsomes opioidergic system (which may be indirect).
(21). However, these results have not been confirmed
in other studies (22,23).
APAP and the Opioidergic System
Graham and Scott (13) have suggested that there It seems likely that at least one mechanism con-
may be 2 important consequences of the metabolism tributing to the analgesic mechanisms of APAP, di-
of APAP by the peroxidase function of the COX-1 and rectly or indirectly, is its effects on opioid containing
COX-2. First, the metabolism utilizes reduced glutathi- pathways. Pretreatment with APAP or dipyrone (60–
one and there may be a local depletion of the glu- 360 mg kg(-1)) reversed hyperalgesia induced by intra-
tathione. Reduced glutathione is a cofactor of many plantar injection of 250 microg lamda-carrageenan CG
enzymes and, in particular, is a cofactor of membrane- and increased nociceptive threshold in the inflamed
associated PGE synthase. Consequently, the local de- paw above the basal level (hypoalgesia) (11). APAP,
pletion of reduced glutathione may lead to decreased but not dipyrone, also raised nociceptive thresholds in
production of PGE2. The second possible consequence the non-inflamed paw (11). Subcutaneous, but not lo-
of the metabolism of APAP is that the 2 reactive me- cal, administration of naltrexone, a specific opioid an-
tabolites may combine directly with enzymes involved tagonist, reversed the hypoalgesia induced by APAP,
in PG synthesis and inhibit them (13). Furthermore, but similar naltrexone treatment had no effect on di-
APAP appears to be a weak competitive inhibitor of pyrone-induced analgesia (11).
cyclooxygenase. This is indicated by the observation Ruggieri et al (35) evaluated the possible role of
that APAP can competitively inhibit the cyclooxygen- the opioidergic system in the antinociceptive effect
ase function of COX-1 under conditions in which the of the APAP metabolite AM404, since μ and κ recep-
peroxidase function is inactive (23). The general find- tors are strongly implicated in supraspinal and spinal
ing at least in suboptimal conditions, is that the effect opioid induced analgesia. A functional relationship
of APAP on COX-2 is weak inhibition (24-26). between the opioidergic and serotonergic systems in
It is conceivable that APAP may affect COX activ- the pain control pathways has been suggested on the
ity. Lee et al (27) using repeated dosing of APAP found basis of behavioral studies (36). The 2 pathways are
that COX-1 gene expression was significantly down- closely interconnected and can interact to modulate
regulated at 24 hours by ketorolac, rofecoxib, and and produce many behavioral changes, including no-
APAP. APAP suppression of PGE2 without inhibiting ciception (35,37).
TXB2 release, when COX-2 gene expression is up-regu- Ruggieri and colleagues (35) assessed the possible
lated, suggests that APAP is a selective CPX-2 inhibi- implication of 5-hydroxytryptamine (5-HT) [serotonin]
tor in vivo (27). The up-regulation of COX-2 gene and in the antinociceptive effect of AM404, by evaluating
down-regulation of COX-1 gene expression suggests the influence of 5-HT1A (NAN-190), 5-HT2 (ketanse-
that APAP may result in changes in COX-derived pros- rin), and 5-HT3 (ondansetron) receptor antagonists
tanoids with reported doses (27). and studying the possible changes in 5-HT and 5-hy-
Interaction with multiple other neurotransmitter sys- droxyindoleacetic acid (5-HIAA) concentrations in the
tems has been proposed to explain the analgesic effects frontal cortex and the pons of the rat.
of APAP, including the serotonergic system, opioidergic, Comparing the %MPE values with those obtained
noradrenergic, cholinergic, and nitric acid (NO)-synthase with APAP, it was observed that the effect of AM404
systems (16,28,29). APAP may interfere with nocicep- was significantly lower (about 50%) in either test (35).
tion associated with spinal NMDA receptor activation. These data might suggest that APAP produced its ef-
This effect may involve an inhibitory action on spinal NO fect only partially through its metabolite AM404. This
mechanisms (28). APAP-induced antinociception seems finding is supported by the observation that either
to derive from the synergy between peripheral, spinal, APAP (400 mg/kg) or AM404 (10 mg/kg) produced sim-
and supraspinal sites (16,30). The supraspinal compo- ilar plasma AM404 concentrations (35).
nents contributing to the analgesic mechanisms of APAP Ruggieri et al (35) demonstrated the involvement
include opioid-like and serotonergic (31,32), with the of the opioid system in the antinociceptive effects of
latter (28) of much more significance (16). Although the these drugs. μ1 and κ opioid receptors seem to be im-
involvement of μ-opioid receptors in the antinociceptive plicated, since the activity of AM404 was significantly

www.painphysicianjournal.com 271
Pain Physician: January/February 2009:12:269-280

decreased by naloxonazine (a selective μ1 receptor an- malin test. It has been postulated that APAP-induced
tagonist) and MR2266 (a selective κ antagonist). Simi- stimulation of 5-HT1A receptors may lead to reinforce-
larly, the antinociceptive activity of partially APAP was ment of bulbospinal 5-HT descending inhibitory path-
inhibited by these antagonists, suggesting that APAP ways at the supraspinal level (46). However, analgesic
also works at least in part, through the μ and κ opioid mechanisms of APAP may depend on the impact of
receptors as previously observed (16). APAP upon both COX activities and the 5-HT system.
APAP metabolites (e.g., AM404) seem to behave APAP-elicited stimulation of spinal 5-HT1A recep-
like the parent compound, activating, at least in part, tors, thought to be due to the reinforcement of 5-HT
the opioidergic and cannabinoid systems. The interac- descending pathways, modulates pain transmission
tion between CB1 and opioid receptors may modulate in a complex manner. Among the cellular events in-
many neurotransmitters, including 5-HT, glutamate, volved, Bonnefont and colleagues (47) described that
and γ-aminobutyric acid (35,38). APAP was up-regulating the expression of GH and IGF-
Serotonergic neurons are part of pathways which 1 receptors, which may participate in its antinocicep-
interact with and may be intertwined or blended with tive activity more or less robustly.
opioid mediated pain modulatory circuit. The interac- The antinociceptive effects of APAP have been
tion of opioid and serotonin systems in contributing correlated with a decrease in the maximal number
to analgesia has been suggested to occur (39,40) via of cortical 5-HT2A receptors (31). Both APAP and mor-
regulation of central μ and κ receptors (41). Ruggieri phine induced an antinociceptive effect in rats on
et al (35) have shown that the block of the opioidergic day one but only APAP maintained this effect for 7
system (through μ and κ receptor antagonists) is able days while morphine did not (15). The number of μ-
to revert the changes in 5-HT levels and serotonergic opioid receptors decreased on days 1, 3, and 7 by a
turnover induced by APAP. Therefore, the implication similar percentage after APAP administration (by 29,
of both serotonergic and opioidergic systems in the 31, and 34%, respectively), while morphine produced
activity of APAP could occur through a mechanism in a progressive decrease in comparison with controls
which the activation of one pathway will influence (by 37, 49, and 60%, respectively) and κ-opioid recep-
the other (35). tors were unaffected. Both drugs similarly decreased
Marek (42) demonstrated a physiological interac- the 5-HT2 receptor number on all days of treatment
tion between 5-HT2 and μ-opioid receptors in some (by about 30 %) (16). Sandrini et al (16) concluded
cortical areas. Thus, APAP may act through both opi- that the opioidergic and serotonergic systems are in-
oidergic and serotonergic systems in a synergistic volved in different ways in the induction and main-
manner. tenance of antinociception after APAP or morphine
treatment.
APAP and the Serotonergic System The involvement of PGs in the mechanism of ac-
The analgesic mechanism of action of APAP may tion of APAP has been proposed, taking into account
involve 5-HT playing a key contributing factor as sug- the inhibition of the central cyclo-oxygenases (COX-
gested by multiple authors (31,35,43): an increase in 5- 1, COX-2 and COX-3) exerted by this drug (10,12-15),
HT levels in various CNS structures has been detected although the results are controversial in this regard.
after APAP treatment (29). Ruggieri and colleagues Moreover, interaction with many neurotransmitter
(35) demonstrated a differential contribution of systems, in particular with the serotonergic system,
AM404 and APAP on the serotonergic system in their has been proposed to explain the effect of APAP: opi-
antinociceptive effects. Investigations also examined oidergic, noradrenergic, serotonergic, cholinergic, and
the roles of serotonergic (5-HT1A, 5-HT2, and 5-HT3) re- NO-synthase systems have been studied in this regard
ceptor subtypes in the mechanisms of APAP-induced (28,29). Indeed, APAP-induced antinociception seems
analgesia (44). to derive from the synergy between peripheral, spinal,
Some type of interaction with the 5-HT system and supraspinal sites (30). The supraspinal components
likely contributes to APAP-induced antinociception identified in the mechanism of APAP are opioid-like
(29). In agreement with this, Bonnefont and colleagues and serotonergic (31,32), the former contributing less
(45) have shown that blocking spinal 5-HT1A receptors than the latter (28), although the involvement of μ-
inhibits the antinociceptive action of APAP in the for- opioid receptors in the antinociceptive action of APAP

272 www.painphysicianjournal.com
Potential Analgesic Mechanisms of Acetaminophen

is still a matter of debate (33,34). We had previously (15). However, these and other theories have not
detected an increase in serotonin concentration in explained how the action of APAP initiates stimu-
the cerebral cortex and in the pons after acute APAP lation of serotonergic descending inhibitory path-
treatment. ways to ameliorate pain.
The central effect of APAP is blocked by some se-
rotonin antagonists, although the receptors have not APAP and the Cannabinoid System
been identified precisely (33,48-50). The depletion The discovery of involvement of endocannabi-
of brain serotonin also blocks the analgesic effect of noids on pain modulation has opened new mecha-
APAP (31,51). The interaction between APAP and se- nistic perspectives (54,55). Anandamide and 2-arachi-
rotonin appears to be indirect because no binding of donoylglycerol, 2 endogenous ligands of CB1 and CB2
APAP has been detected with a variety of serotoner- receptors, mainly metabolized by the fatty acid amide
gic receptors or on the uptake of serotonin (33,52). hydrolase (FAAH), and the monoacylglycerol lipase,
No significant binding or interaction with uptake sites respectively, induce antinociceptive effects (56,57).
has been found with other neurotransmitters. One Similarly, activation of this system by exogenous li-
possible indirect link is that APAP may interact with gands for cannabinoid (particularly CB1) receptors
serotonergic pathways through a decrease in PG syn- induces antinociception in various acute pain tests in
thesis. Perhaps by altering CNS levels of prostanoids, rodents (56,58,59) but also in several animal models
APAP may affect several monoamine neuron types in of chronic pain (60). Several studies reported that ce-
the brain containing the EP3 receptor, a major recep- rebral injection of cannabinoids in the periaqueductal
tor for PGE2 (13). gray (PAG) or the rostroventral medulla (RVM) elicits
The reversal of the antinociceptive effect of sys- antinociception, suggesting the modulation of de-
temically administered APAP by IT administration of scending pathways to inhibit pain processing at the
the potent 5-HT(3) receptor antagonist tropisetron has spinal level (59,61,62).
been reported in rats subjected to the paw pressure Högestätt et al (63) demonstrated that APAP un-
test, suggesting that APAP action is mediated through dergoes a two-step metabolic transformation in the
spinal 5-HT(3) receptors (53). Unlike tropisetron, other brain to form N-arachidonoyl-phenolamine (AM404),
5-HT(3) receptor antagonists, such as ondansetron which is an agonist of transient receptor potential
and granisetron, injected intrathecally were unable to vanilloid type 1, a ligand at selective cannabinoid
reverse the antinociceptive effect of APAP (53). More- subtype 1 (CB1) receptors and an inhibitor of cellu-
over, pretreatment with spinal 5-HT(3) receptors anti- lar anandamide uptake, whose inhibition leads to
sense oligodeoxynucleotides (AODNs) did not reverse an increase in endogenous cannabinoids (64). Can-
the APAP-induced antinociceptive effects (53). These nabinoids produce antinociceptive effects that are
results suggest that APAP-induced antinociceptive ac- mediated chiefly by CB1 receptors (65). Cannabinoid-
tion involves a spinal tropisetron-sensitive receptor induced antinociception also seems to depend, to
that is not the 5-HT(3) receptor and that remains to some extent, on the release of opioid peptides into
be identified (53). the brain (66).
A major hypothesis of APAP’s analgesic mecha- APAP could be metabolized in the brain into
nisms is APAP leading to promotion of the inhibito- AM404, and then inhibit the reuptake of anandamide
ry activity of the descending inhibitory serotonergic (67), with subsequent stimulation of CB1 receptors via
pathways on spinal nociceptive processing, a the- FAAH (63). Thus, the antinociceptive activity of APAP
ory which has been supported by different groups may rely on an interaction with the endocannabinoid
(29,31,51). Lesioning of the bulbospinal descending system (68). Mallet et al (69) hypothesized that the in-
serotonergic pathways abolishes the antinocicep- teraction of APAP with the endocannabinoid system
tive action of APAP (51). The laboratory of Alain could be on the basis of the reinforcement of the se-
Eschalier demonstrated that different spinal 5-HT rotonergic system.
receptor subtypes are involved in the antinocicep- Zygmunt et al (70) started from the observation
tive effect of APAP in rats (31,45) and other inves- of the striking structural similarity between APAP and
tigations supported this serotonergic mechanism as the fatty acid amide N-arachidonoyl-phenolamine
contributing to APAP-induced analgesia in humans (AM404). Zygmunt and colleagues (70) have shown

www.painphysicianjournal.com 273
Pain Physician: January/February 2009:12:269-280

that APAP, following deacetylation to its primary neuropathic pain (sciatic nerve ligation in the rat),
amine (p-aminophenol) is conjugated with arachidon- and inflammatory pain (complete Freund’s adju-
ic acid in the brain and spinal cord to form AM404 (via vant injection in the rat). In the formalin model,
FAAH), which also catalyzes the hydrolysis of anan- administration of AM404 (1-10 mg/kg i.p.) elicited
damide and which can also act in the reverse direc- dose-dependent antinociceptive effects, which were
tion and catalyze the synthesis of anandamide from prevented by the CB1 cannabinoid receptor antago-
ethanolamine and arachidonic acid. Zygmunt and col- nist rimonabant (SR141716A; 1 mg/kg i.p.) but not
leagues (70) have shown that FAAH can indeed syn- by the CB2 antagonist SR144528 (1 mg/kg i.p.) or the
thesize AM404 from p-aminophenol and arachidonic vanilloid antagonist capsazepine (30 mg/kg i.p.) (73).
acid in vitro, and that, in addition, no formation of Comparable effects were observed with UCM707
AM404 is observed in vitro or in vivo in brain tissue [N-(3-furylmethyl)-eicosa-5,8,11,14-tetraenamide],
from FAAH gene in knockout mice. Bertolini and col- another anandamide transport inhibitor. In both
leagues (71) produced experimental data suggesting the chronic constriction injury and complete Freud’s
that the analgesic activity of APAP involves potentia- adjuvant model, daily treatment with AM404 (1-10
tion of the cannabinoid/vanilloid tone in the brain mg/kg s.c.) for 14 days produced a dose-dependent
and in dorsal root ganglia. reduction in nocifensive responses to thermal and
APAP does not bind to CB1 receptors but rather mechanical stimuli, which was prevented by a single
activates CB1 receptors via an indirect pathway rely- administration of rimonabant (1 mg/kg i.p.) and was
ing on FAAH-dependent AM404 formation and subse- accompanied by decreased expression of cyclooxy-
quent AM404 effects on anandamide transport. genase-2 and inducible nitric-oxide synthase in the
Mallet et al (69) assessed the influence of FAAH sciatic nerve (73).
inhibitors on the antinociceptive activity of APAP. Caballero et al (74) evaluated the effect of AM404
Phenylmethylsulphonyl fluoride (PMSF), an FAAH in- in human T cells, discovering that AM404 is a potent
hibitor, (10 mg/kg, s.c.), used at a dose shown to be inhibitor of T-cell receptor (TCR)-mediated T-cell acti-
able to abolish the FAAH-dependent metabolism of vation. They found that AM404 specifically inhibited
APAP (63), significantly inhibited the antinociceptive both IL-2 and TNF-α gene transcription and TNF-α
effect of APAP in both the paw pressure and the for- synthesis in CD3/CD28-stimulated Jurkat T cells in a
malin tests (69). In addition, URB597 (0.15 mg/kg, i.p.), FAAH independent way (74). AM404 inhibited both
an irreversible brain-penetrating FAAH inhibitor, also the binding to DNA and the transcriptional activity of
reversed the antinociception elicited by APAP (69). endogenous NFAT and the transcription activity driven
Jayamanne et al (72) reported that systemic ad- by the over expressed fusion protein Gal4-NFAT. How-
ministration of a selective FAAH inhibitor URB597 ever, AM404 did not affect early steps in NFAT signal-
(0.3 mg kg(-1)) reduced the mechanical allodynia and ing such as CD3-induced calcium mobilization and
thermal hyperalgesia in the complete Freund’s adju- NFAT1 dephosphorylation (74).
vant (CFA) model of inflammatory pain. The effects of Different studies reported the implication of the
URB597 in the CFA model were dose dependent and endocannabinoid system in pain modulation either
were reduced by coadministration with the cannabi- at spinal or supraspinal levels (37). Notably, the stim-
noid CB1 antagonist AM251 (1 mg kg(-1)), or the CB2 ulation of CB1 receptors in PAG and RVM reduced
and SR144528 (1 mg kg(-1)). Coadministration with GABA release from the presynaptic boutons of local
AM251 plus SR144528 completely reversed the ef- interneurons which can reduce the GABAergic nega-
fects of URB597. These findings suggest that the FAAH tive influence on the inhibitory descending pathways
inhibitor URB597 produces cannabinoid CB1 and CB2 (75,76). The antinociceptive activity of APAP has been
receptor-mediated analgesia in inflammatory pain shown to depend on spinal serotonergic receptors
states (72). (33,43,45,77) and that the source of spinal 5-HT comes
La Rana et al (73) investigated the effects of exclusively from supraspinal centers and mainly from
the endocannabinoid transport inhibitor AM404 [N- RVM (37). Mallet and colleagues (69) suspected that
(4-hydroxyphenyl)-eicosa-5,8,11,14-tetraenamide] the participation of the endocannabinoid system in
on rodent models of acute and persistent nocicep- the effect of APAP would occur through the rein-
tion (intraplantar formalin injection in the mouse), forcement of the activity of the bulbospinal seroto-

274 www.painphysicianjournal.com
Potential Analgesic Mechanisms of Acetaminophen

nergic pathways. Mallet et al (69) demonstrated that a multi-step process. Firstly, the drug would need to
the antinociceptive effect induced by a CB1 recep- be metabolized in 2 steps to form cerebral FAAH de-
tor agonist arachidonyl-2’-chloroethylamide (ACEA) pendent AM404. This metabolite could reinforce the
needed intact descending bulbospinal serotonergic activity of the endocannabinoid system through CB1
pathways. Furthermore, ACEA-dependent recruit- receptors, which would in turn reinforce the activity
ment of the serotonergic system involved similar spi- of the bulbospinal serotonergic inhibitory pathways
nal 5-HT receptors to those involved in the antino- (Fig. 1). Finally, antinociception would occur at the spi-
ciceptive action of APAP and 5-HT (43,45,46,48,50), nal level where activation of stimulus-dependent 5-HT
i.e., the 5-HT1A receptor in the formalin test and the receptors would block the transmission of nociceptive
5-HT3/4 receptor in the paw pressure test. This simi- influx (69).
lar 5-HT-dependent mechanism and the inhibition Still another theory has been advanced to help
of the effect of APAP by inactivation of CB1 recep- partially explain the analgesic effects of APAP. Smith
tors suggest that the endocannabinoid system is an (81) has proposed that it is conceivable that increased
important link between APAP and 5-HT to produce levels of AM404 (resulting from increased APAP’s con-
antinociception (69). version to AM404 via FAAH) may compete with other
NSAIDs inhibit fatty-acid amide hydrolase FAAH, substrates for FAAH and thus may lead to increased
the enzyme responsible for the metabolism of anan- levels of PEA which exhibits antinociceptive activity
damide, an endocannabinoid (78). Guindon et al (78) likely via agonist effects at the peroxisome prolifera-
showed that the analgesic effect of the combination tors-activated receptor-α. Smith (81) has postulated
of NSAIDs and anandamide was synergistic. Guindon that increased anandamide may increase PEA levels
and colleagues (78) also found that paw tissue levels in tissues by competing with PEA for FAAH-medi-
of anandamide, oleoylethanolamide, and palmitoyle- ated hydrolysis (if most of the FAAH is being used
thanolamide (PEA) were significantly higher when to synthesize AM404 from APAP and/or metabolize
anandamide was combined with NSAIDs and that this anandamide to arachidonic acid and ethanolamine,
effect was greater with rofecoxib. then it is conceivable that there may be functionally
It also remains possible that AM404 mediates less FAAH to metabolize PEA, and thus, effectively
the antinociceptive activity of APAP through other resulting in higher levels of PEA). Smith (81) then
mechanisms (69). For instance, AM404 could ac- reasoned that some of APAP’s analgesic action (al-
tivate the TRPV1 receptor, whose stimulation has beit a minor problem) may derive from the increased
been shown to elicit antinociception in the PAG (61) levels of PEA, a compound which may exhibit analge-
and to be active in the tetrad tests (79). In addition, sic activity at least in part by activating the nuclear
AM404 has been shown to inhibit COX activities in receptor peroxisome proliferator-activated receptor
vitro, with a potency close to those of NSAIDs (63). — α (PPAR-α). LoVerme et al (82) demonstrated the
The involvement of a central COX inhibition during PPAR-α mediates the anti-inflammatory actions of
the APAP-induced antinociceptive and/or antipyret- PEA, a compound which Calignano et al (83,84) sug-
ic activities will have to be confirmed in vivo (69). gested may function as an endogenous regulator of
The fact that AM404 is not found in blood after ad- nociception.
ministration of APAP (63) might explain why APAP Broad-spectrum analgesia by PEA has been docu-
does not exert any significant peripheral anti-in- mented in a variety of pain models. PEA reduces pain
flammatory effect (69). This could therefore further behaviors elicited by formalin (83,85), magnesium sul-
explain why APAP does not exert significant clinical fate (84), carrageenan (86,87), nerve growth factor
anti-inflammatory effect. Other studies stated that (88), and turpentine (85,89). Moreover, PEA was found
the antinociceptive activity of APAP may rely on a to inhibit hyperalgesia after sciatic nerve ligation, a
decrease in spinal NO (e.g., 4), which could also be a model of neuropathic pain (90). Because PEA-induced
consequence of the reinforcement of the endocan- analgesia is rapid and precedes the compound’s anti-
nabinoid system (69,80). inflammatory actions, it has been suggested that PEA
The data of Mallet et al (69) together with results may function as an endogenous regulator of nocicep-
published by Högestätt et al (63) suggest that the tion (83).
activity of APAP, orally administered, would require

www.painphysicianjournal.com 275
Pain Physician: January/February 2009:12:269-280

Fig. 1. Potential analgesic mechanisms of APAP.

276 www.painphysicianjournal.com
Potential Analgesic Mechanisms of Acetaminophen

Conclusion
The precise analgesic actions of APAP remain terlinked) which may contribute to APAP’s analgesic
uncertain. It is becoming clear that the predominant activity. A greater understanding of how APAP pro-
mechanisms largely responsible for APAP’s analgesic vides pain relief may lead to optimal analgesia as well
activity are located in the CNS. Also, it appears that as improved utilization of analgesic polypharmacy.
there may be multiple mechanisms (which may be in-

References
1. Morse HN. Ueber eine neue Darstel- phen and other analgesic ⁄ antipyret- ase-2 inhibition involving interactions
lungsmethode der Acetylamidophe- ic drugs: Cloning, structure, and ex- with Ser-530 and Tyr-385. J Biol Chem
nole. Ber Deutscher Chem Ges 1878; pression. Proc Natl Acad Sci USA 2002; 2003; 278:45763-45769.
11:232-233. 99:13926-13931. 20. Smith WL, DeWitt DL, Garavito RM. Cy-
2. von Mering J. Beitrage zur Kenntniss 11. Rezende RM, França DS, Menezes GB, clooxygenases: Structural, cellular,
der Antipyretica. Ther Monatsch 1893; dos Reis WG, Bakhle YS, Francischi JN. and molecular biology. Annu Rev Bio-
7:577-587. Different mechanisms underlie the an- chem 2000; 69:145-182.
3. Brodie BB, Axelrod J. The fate of acet- algesic actions of paracetamol and di- 21. Hsuanyu Y, Dunford HB. Prostaglan-
anilide in man. J Pharmacol Exp Ther pyrone in a rat model of inflammatory din H synthase kinetics. The effect of
1948; 94:29-38. pain. Br J Pharmacol 2008; 153:760- substituted phenols on cyclooxygen-
4. Bertolini A, Ottani A, Sandrini M. Se- 768. ase activity and the substituent effect
lective COX-2 inhibitors and dual act- 12. Flower RJ, Vane JR. Inhibition of pros- on phenolic peroxidatic activity. J Biol
ing anti-inflammatory drugs: Critical re- taglandin synthetase in brain explains Chem 1992; 267:17649-17657.
marks. Curr Med Chem 2002; 9:1033- the anti-pyretic activity of paracetamol 22. Ferrari RA, Ward SJ, Zobre CM, Van
1043. (4-acetamidophenol). Nature 1972; Liew DK, Perrone MH, Connell MJ. Esti-
5. Crawley B, Saito O, Malkmus S, Fitzsim- 240:410-411. mation of the in vivo effect of cyclooxy-
mons B, Hua XY, Yaksh TL. Acetamino- 13. Graham GG, Scott KF. Mechanism of genase inhibitors on prostaglandin E2
phen prevents hyperalgesia in cen- action of paracetamol. Am J Ther 2005; levels in mouse brain. Eur J Pharmacol
tral pain cascade. Neurosci Lett 2008; 12:46-55. 1990; 179:25-34.
442:50-53. 14. Kis B, Snipes JA, Busija DW. Acetamin- 23. Thompson D, Eling T. Mechanism of in-
6. Courad JP, Besse D, Delchambre C, ophen and ciclooxygenase-3 puzzle: hibition of prostaglandin H synthase
Hanoun N, Hamon M, Eschalier A, Sorting out facts, fictions, and uncer- by eugenol and other phenolic peroxi-
Caussade F, Cloarec A. Acetaminophen tainties. J Pharmacol Exp Ther 2005; dase substrates. Mol Pharmacol 1989;
distribution in the rat central nervous 315: 1-7. 36:809-817.
system. Life Sci 2001; 69:1455-1464. 15. Pickering G, Loriot MA, Libert F, Eschal- 24. Swierkosz TA, Jordan L, McBride M,
7. Seo YJ, Kwon MS, Choi HW, Choi SM, ier A, Beaune P, Dubray C. Analgesic McGough K, Devlin J, Botting RM. Ac-
Nam JS, Lee JK, Jung JS, Park SH, Suh effect of acetaminophen in humans: tions of paracetamol on cyclooxygen-
HW. The differential effects of acet- First evidence of a central serotoner- ases in tissue and cell homogenates
aminophen on lipopolysaccharide in- gic mechanism. Clin Pharmacol Ther of mouse and rabbit. Med Sci Monit
duced hyperalgesia in carious mouse 2006; 79:371-378. 2002; 8:BR496-BR503.
pain models. Pharmacol Biochem Be- 16. Sandrini M, Vitale G, Ruggieri V, Pini 25. Boutaud O, Aronoff DM, Richardson
hav 2008; 91:121-127. LA. Effect of acute and repeated ad- JH, Marnett LJ, Oates JA. Determinants
8. Chandrasekharan NV, Simmons DL. The ministration of paracetamol on opi- of the cellular specificity of acetamino-
cyclooxygenases. Genome Biol 2004; oidergic and serotonergic systems in phen as an inhibitor of prostaglandin
5:241. rats. Inflamm Res 2007; 56:139-142. H(2) synthases. Proc Natl Acad Sci USA
17. Warner TD, Mitchell JA. Cyclooxygen- 2002; 99:7130-7135.
9. Aronoff DM, Oates JA, Boutaud O. New
insights into the mechanism of action ase-3 (COX-3): Filling in the gaps to- 26. Ouellet M, Percival MD. Mechanism of
of acetaminophen: Its clinical phar- ward a COX continuum? Proc Natl Acad acetaminophen inhibition of cycloox-
macologic characteristics reflect its in- Sci USA 2002; 99:13371-13373. ygenase isoforms. Arch Biochem Bio-
hibition of the two prostaglandin H2 18. Anderson BJ. Paracetamol (acetamino- phys 2001; 387:273-280.
synthases. Clin Pharmacol Ther 2006; phen): Mechanisms of action. Paediatr 27. Lee YS, Kim H, Brahim JS, Rowan J, Lee
79:9-19. Anaesth 2008; 18:915-921. G, Dionne RA. Acetaminphen selec-
10. Chandrasekharan NV, Dai H, Roos 19. Rowlinson SW, Kiefer JR, Prusakiewicz tively suppresses peripheral prosta-
KL, Evanson NK, Tomsik J, Elton TS, JJ, Pawlitz JL, Kozak KR, Kalgutkar AS, glandin E2 release and increases COX-
Simmons DL. COX-3, a cyclooxygen- Stallings WC, Kurumbail RG, Marnett 2 gene expression in a clinical mod-
ase-1 variant inhibited by acetamino- LJ. A novel mechanism of cyclooxygen- el of acute inflammation. Pain 2007;
129:279-286.

www.painphysicianjournal.com 277
Pain Physician: January/February 2009:12:269-280

28. Björkman R. Central antinociceptive 781. 50. Bardin L, Lavarenne J, Eschalier A. Se-
effects of nonsteroidal antiinflamma- 39. Watkins LR, Mayer DJ. Multiple endog- rotonin receptor subtypes involved in
tory drugs and paracetamol. Experi- enous opiate and nonopiate analgesia the spinal antinociceptive effect of 5-
mental studies in the rat. Acta Anaes- systems: Evidence of their existence HT in rats. Pain 2000; 86:11-18.
thesiol Scand Suppl 1995; 103:1-44. and clinical implications. Ann NY Acad 51. Tjolsen A, Lund A, Hole K. Antinoci-
29. Bonnefont J, Courade JP, Alloui A, Es- Sci 1986; 467:273-299. ceptive effect of paracetamol in rats
chalier A. Mécanisme de l’action an- 40. Lipp J. Possible mechanisms of mor- is partly dependent on spinal seroto-
tinociceptive du paracetamol. Drugs phine analgesia. Clin Neuropharmacol nergic systems. Eur J Pharmacol 1991;
2003; 63:1-4. 1991; 14:131-147. 193:193-201.
30. Raffa SB, Stone DJ, Tallarida RJ. Discov- 41. Fields H. State-dependent opioid con- 52. Raffa RB, Codd EE. Lack of binding of
ery of “self-synergistic” spinal/supra- trol of pain. Nat Rev Neurosci 2004; acetaminophen to 5-HT receptor or up-
spinal antinociception produced by ac- 5:565-575. take sites (or eleven other binding/up-
etaminophen (paracetamol). J Pharma- take assays). Life Sci 1996; 59:L37-
42. Marek GJ. Behavioral evidence of mu-
col Exp Ther 2000; 295:291-294. L40.
opioid and 5-HT2A receptor interac-
31. Pini LA, Sandrini M, Vitale G. The an- tions. Eur J Pharmacol 2003; 474:77- 53. Libert F, Bonnefont J, Bourinet E,
tinociceptive action of paracetamol is 83. Doucet E, Alloui A, Hamon M, Nargeot
associated with changes in the sero- J, Eschalier A. Acetaminophen: A cen-
43. Pelissier T, Halloui A, Paeile C, Eschalier
tonergic system in the rat brain. Eur J tral analgesic drug that involves a spi-
A. Evidence in central antinociceptive
Pharmacol 1996; 308:31-40. nal tropisetron-sensitive, non-5-HT(3)
effect of paracetamol involving spinal
32. Pini LA, Vitale G, Ottani A, Sandrini M. receptor-mediated effect. Mol Phar-
5- HT3 receptors. Neuroreport 1995;
Naloxone-reversible antinociception macol 2004; 66:728-734.
6:983-993.
by paracetamol in the rat. J Pharmacol 54. Di Marzo V, Bifulco M, De Petrocellis L.
44. Sufka KJ, Shomburg FM, Giordano J. Re-
Exp Ther 1997; 280:934-940. The endocannabinoid system and its
ceptor mediation of 5-HT-induced in-
33. Pelissier T, Alloui A, Caussade F, Es- therapeutic exploitation. Nat Rev Drug
flammation and nociception in rats.
chalier A. Paracetamol exerts a spinal Discov 2004; 3:771-784.
Pharmacol Biochem Behav 1992; 41:53-
antinociceptive effect involving an in- 56. 55. Piomelli D, Giuffrida A, Calignano A,
direct interaction with 5-hydroxytryp- Rodriguez de Fonseca F. The endocan-
45. Bonnefont J, Chapuy E, Clottes E, Alloui
tamine3 receptors: In vivo and in vitro nabinoid system as a target for ther-
A, Eschalier A. Spinal 5-HT1A receptors
evidence. J Pharmacol Exp Ther 1996; apeutic drugs. Trends Pharmacol Sci
differentially influence nociceptive pro-
287:8-14. 2000; 21:218-224.
cessing according to the nature of the
34. Carlsson KH, Monzel W, Jurna I. De- noxious stimulus in rats: Effect of WAY- 56. Hohmann AG, Suplita Jr RL. Endocan-
pression by morphine and the non- 100635 on the antinociceptive activi- nabinoid mechanisms of pain modula-
opioid analgesic agents, metamizol ties of paracetamol, venlafaxine and 5- tion. Aaps J 2006; 8:E693-E708.
(dipyrone), lysine acetylsalicylate and HT. Pain 2005; 114:482-490. 57. Walker JM, Huang SM. Endocannabi-
paracetamol, of activity in rat thala- noids in pain modulation. Prostaglan-
46. Bonnefont J, Alloui A, Chapuy E, Clottes
mus neurones evoked by electrical dins Leukot Essent Fatty Acids 2002;
E, Eschalier A. Orally administered
stimulation of nociceptive afferents. 66:235-242.
paracetamol does not act locally in the
Pain 1988; 32:313-326.
rat formalin test: Evidence for a supra- 58. Fox A, Bevan S. Therapeutic potential
35. Ruggieri V, Vitale G, Pini LA, Sandri- spinal, serotonin-dependent antino- of cannabinoid receptor agonists as
ni M. Differential involvement of opi- ciceptive mechanism. Anesthesiology analgesic agents. Expert Opin Investig
oidergic and serotonergic systems in 2003; 99:976-981. Drugs 2005; 14:695-703.
the antinociceptive activity of N-ara-
47. Bonnefont J, Daulhac L, Etienne M, 59. Meng ID, Manning BH, Martin WJ,
chidonoyl-phenolamine (AM404) in
Chapuy E, Mallet C, Ouchchane L, De- Fields HL. An analgesia circuit acti-
the rat: Comparison with paracetamol.
val C, Courade JP, Ferrara M, Eschalier vated by cannabinoids. Nature 1998;
Naunyn-Schmiedeberg’s Arch Pharma-
A, Clottes E. Acetaminophen recruits 395:381-383.
col 2008; 377:219-229
spinal p42/p44 MAPKs and GH/IGF-1 60. Fox A, Kesingland A, Gentry C, McNair
36. Duman EN, Kesim M, Kadioglu M, Yaris receptors to produce analgesia via the K, Patel S, Laszlo U, Iain J. The role of
E, Kalyoncu I, Erciyes N. Possible in- serotonergic system. Mol Pharmacol central and peripheral Cannabinoid1
volvement of opioidergic and seroto- 2007; 71:407-415. receptors in the antihyperalgesic activ-
nergic mechanisms in antinociceptive
48. Alloui A, Chassaing C, Schmidt J, Ardid ity of cannabinoids in a model of neu-
effect of paroxetine in acute pain. J
D, Dubray C. Paracetamol exerts a spi- ropathic pain. Pain 2001; 92:91-100.
Pharmacol Sci 2004; 94:161-165.
nal, tropisetron-reversible, antinoci- 61. Maione S, Bisogno T, de Novellis V,
37. Millan MJ. Descending control of pain. ceptive effect in an inflammatory pain Palazzo E, Cristino L, Valenti M, Petro-
Progr Neurobiol 2002; 66:355-474. model in rats. Eur J Pharmacol 2002; sino S, Gugliemotti V, Rossi F, Di Mar-
38. Schoffelmeer ANM, Hogenboom F, 443:71-77. zo V. Elevation of endocannabinoid
Wardeh G, De Vries TJ. Interaction be- 49. Chen C, Bazan NG. Acetaminophen levels in the ventrolateral periaque-
tween CB1 and μ opioid receptors me- modifies hippocampal synaptic plas- ductal grey through inhibition of fatty
diating inhibition of neurotransmitter ticity via a presynaptic 5-HT2 receptor. acid amide hydrolase affects descend-
release in rat nucleus accumbens core. Neuroreport 2003; 14:743-747. ing nociceptive pathways via both can-
Neuropharmacology 2006: 51:773-

278 www.painphysicianjournal.com
Potential Analgesic Mechanisms of Acetaminophen

nabinoid receptor type 1 and transient ni S, Tacchi R, Leone S. Paracetamol: 80. Costa B, Siniscalco D, Trovato AE, Co-
receptor potential vanilloid type-1 re- New vistas of an old drug. CNS Drug melli F, Sotgiu ML, Colleoni M, Maione
ceptors. J Pharmacol Exp Ther 2006; Reviews 2006; 12:250-275. S, Rossi F, Giagnoni G. AM404, an in-
316:969-982. 72. Jayamanne A, Greenwood R, Mitchell hibitor of anandamide uptake, pre-
62. Meng ID, Johansen JP. Antinociception VA, Asian S, Piomelli D, Vaughan CW. vents pain behaviour and modulates
and modulation of rostral ventrome- Actions of the FAAH inhibitor URB597 cytokine and apoptotic pathways in
dial medulla neuronal activity by lo- in neuropathic and inflammatory a rat model of neuropathic pain. Br J
cal microinfusion of a cannabinoid re- chronic pain models. Br J Pharmacol Pharmacol 2006; 148:1022-1032.
ceptor agonist. Neuroscience 2004; 2006; 147:281-288. 81. Smith HS. A novel hypothesis of a po-
124:685-693. 73. La Rana G, Russo R, Campolongo P, tential antinociceptive mechanism of
63. Högestätt ED, Jönsson BA, Ermund A, Bortolato M, Mamgieri RA, Cuomo V, acetaminophen. Presented at the Cap-
Andersson DA, Björk H, Alexander JP, Iacono A, Mattace Raso G, Meli R, Pi- itol District Pain Meeting. October,
Cravatt BF, Basbaum AI, Zygmunt PM. omelli D, Calignano A. Modulation of 2008. Albany, NY.
Conversion of acetaminophen to bio- neuropathic and inflammatory pain 82. LoVerme JL, Fu J, Astarita G, La Rana G,
active N-acylphenolamine AM404 via by the endocannabinoid transport in- Russo R, Calignano A, Piomelli D. The
fatty acid amide hydrolase-depen- hibitor AM404 9 (N-[4-Hydroxyphenyl]- nuclear receptor peroxisome prolifer-
dent arachidonic acid conjugation in eicosa-5,8,11,14-tetraenamide). J Phar- ators-activated receptor-{alpha} medi-
the nervous system. J Biol Chem 2005; macol Exp Ther 2006; 317:1365-1371. ates the anti-inflammatory actions of
280:31405-31412. 74. Caballero FJ, Navarrete CM, Hess S, palmitoylethanolamide. Mol Pharma-
64. Flegley D, Kathuria S, Mercier R, Li C, Fiebich BL, Appendino G, Macho A, col 2005; 67:15-19.
Goutopoulus A, Makriannis A, Piomel- Muñoz E, Sancho R. The acetamino- 83. Calignano A, La Rana G, Giuffrida A,
li D. Anandamide transport is indepen- phen-derived bioactive N-acylphenol- Piomelli D. Control of pain initiation
dent of fatty acid hydrolase activity amine AM404 inhibits NFAT by tar- by endogenous cannabinoids. Nature
and is blocked by the hydrolysis-resis- geting nuclear regulatory events. Bio- 1998; 394:277-281.
tant inhibitor AM1172. Proc Natl Acad chem Pharmacol 2007; 73:1013-1023. 84. Calignano A, La Rana G, Piomelli D.
Sci USA 2004; 101:8756-8761. 75. Vaughan CW, Connor M, Bagley EE, Antinociceptive activity of the endog-
65. Palazzo E, de Novellis V, Petrosino S, Christie MJ. Actions of cannabinoids enous fatty acid amide, palmityle-
Marabese I, Vita D, Giordano C, Di Mar- on membrane properties and synap- thanolamide. Eur J Pharmacol 2001;
zo V, Mangoni GS, Rossi F, Maione S. tic transmission in rat periaqueductal 419:191-198.
Neuropathic pain and the endocan- gray neurons in vitro. Mol Pharmacol 85. Jaggar SI, Hasnie FS, Sellaturay S, Rice
nabinoid system in the dorsal raphe: 2000; 57:288-295. AS. The anti-hyperalgesic actions of
Pharmacological treatment and inter- 76. Vaughan CW, McGregor IS, Christie MJ. the cannabinoid anandamide and the
actions with the serotonergic system. Cannabinoid receptor activation inhib- putative CB2 receptor agonist palmi-
Eur J Neurosci 2006; 24:2011-2020. its GABAergic neurotransmission in toylethanolamide in visceral and so-
66. Cichewicz DL. Synergistic interactions rostral ventromedial medulla neurons matic inflammatory pain. Pain 1998;
between cannabinoid and opioid anal- in vitro. Br J Pharmacol 1999; 127:935- 76:189-199.
gesics. Life Sci 2004; 74:1317-1324. 940. 86. Conti S, Costa B, Colleoni M, Parolaro
67. Di Marzo V, Deutsch DG. Biochemistry 77. Courade JP, Chassaing C, Bardin L, Al- D, Giagnoni G. Antiinflammatory ac-
of the endogenous ligands of canna- loui A, Eschalier A. 5-HT receptor sub- tion of endocannabinoid palmitoyleth-
binoid receptors. Neurobiol Dis 1998; types involved in the spinal antino- anolamide and the synthetic cannabi-
5:386-404. ciceptive effect of acetaminophen in noid nabilone in a model of acute in-
rats. Eur J Pharmacol 2001; 432:1-7. flammation in the rat. Br J Pharmacol
68. Ottani A, Leone S, Sandrini M, Ferrari
A, Bertolini A. The analgesic activity of 78. Guindon J, LoVerme J, De Léan A, Pio- 2002; 135:181-187.
paracetamol is prevented by the block- melli D, Beaulieu P. Synergistic anti- 87. Mazzari S, Canella R, Petrelli L, Mar-
ade of cannabinoid CB1 receptors. Eur nociceptive effects of anandamide, an colongo G, Leon A. N-(2-hydroxyethyl)
J Pharmacol 2006; 531:280-281. endocannabinoid, and nonsteroidal hexadecanamide is orally active in re-
anti-inflammatory drugs in peripheral ducing edema formation and inflam-
69. Mallet C, Daulhac L, Bonnefort J, Le-
tissues: A role for endogenous fatty- matory hyperalgesia by down-modu-
dent C, Etienne M, Chapuy E, Libert F,
acid ethanolamides. Eur J Pharmacol lating mast cell activation. Eur J Phar-
Eschalier A. Endocannabinoid and se-
2006; 550:68-77. macol 1996; 300:227-236.
rotonergic systems are needed for ac-
etaminophen-induced analgesia. Pain 79. Di Marzo V, Bisogno T, De Petrocellis L, 88. Farquhar-Smith WP, Rice AS. A novel
2008; 139:190-200. Brandi I, Jefferson RG, Winckler RL, Da- neuroimmune mechanism in canna-
vis JB, Dasse O, Mahadevan A, Razdan binoid-mediated attenuation of nerve
70. Zygmunt PM, Chuang H, Movahed
RK, Martin BR. Highly selective CB(1) growth factor-induced hyperalgesia.
P, Julius D, Hogestatt ED. The anan-
cannabinoid receptor ligands and nov- Anesthesiology 2003; 99:1391-1401.
damide transport inhibitor AM404 ac-
el CB(1)/VR(1) vanilloid receptor ‘‘hy- 89. Farquhar-Smith WP, Rice AS. Adminis-
tivates vanilloid receptors. Eur J Phar-
brid” ligands. Biochem Biophys Res tration of endocannabinoids prevents
macol 2000; 396:39-42.
Commun 2001; 281:444-451. a referred hyperalgesia associated
71. Bertolini A, Ferrari A, Ottani A, Guerzo-
with inflammation of the urinary blad-

www.painphysicianjournal.com 279
Pain Physician: January/February 2009:12:269-280

der. Anesthesiology 2001; 94:507-513. induced sensory neuropeptide release


90. Helyes Z, Nemeth J, Than M, Bolcskei in vivo and neuropathic hyperalgesia
K, Pinter E, Szolcsanyi J. Inhibitory ef- in the rat. Life Sciences 2003; 73:2345-
fect of anandamide on resiniferatoxin- 2353.

280 www.painphysicianjournal.com

You might also like