Você está na página 1de 68

Inflammopharmacol (2009) 17:275–342

DOI 10.1007/s10787-009-0016-x Inflammopharmacology


REVIEW

Ibuprofen: pharmacology, efficacy and safety


K. D. Rainsford

Received: 17 July 2009 / Accepted: 4 September 2009 / Published online: 21 November 2009
 Birkhäuser Verlag, Basel/Switzerland 2009

Abstract comparator studies, as well as in epidemiological studies,


Objectives This review attempts to bring together infor- shows that ibuprofen has relatively low risks for gastro-
mation from a large number of recent studies on the intestinal (GI), hepato-renal and other, rarer, ADRs com-
clinical uses, safety and pharmacological properties of pared with other NSAIDs and coxibs. A slightly higher risk
ibuprofen. Ibuprofen is widely used in many countries for of cardiovascular (CV) events has been reported in some,
the relief of symptoms of pain, inflammation and fever. but not all studies, but the risks are in general lower than
The evidence for modes of action of ibuprofen are con- with some coxibs and diclofenac. The possibility that ibu-
sidered in relation to its actions in controlling profen may interfere with the anti-platelet effects of aspirin,
inflammation, pain and fever, as well as the adverse effects though arguably of low grade or significance, has given rise
of the drug. to caution on its use in patients that are at risk for CV
Summary of outcomes At low doses (800–1,200 mg conditions that take aspirin for preventing these conditions.
day-1) which in many countries are approved for non-pre- Paediatric use of ibuprofen is reviewed and the main results
scription (over-the-counter) sale ibuprofen has a good are that the drug is relatively safe and effective as a treat-
safety profile comparable with paracetamol. Its analgesic ment of acute pain and fever. It is probably more effective
activity is linked to its anti-inflammatory effects and is than paracetamol as an antipyretic.
related to reduction in the ex vivo production in blood of Conclusions This assessment of the safety and benefits of
cyclo-oxygenase (COX)-1 and COX-2 derived prostanoids. ibuprofen can be summarized thus: (1) Ibuprofen at OTC
Higher prescription doses (circa 1,800–2,400 mg day-1) doses has low possibilities of serious GI events, and little
are employed long-term for the treatment of rheumatic and prospect of developing renal and associated CV events.
other more severe musculo-skeletal conditions. Recent Ibuprofen OTC does not represent a risk for developing
evidence from large-scale clinical trials with the newer liver injury especially the irreversible liver damage
coxibs, where ibuprofen was as a comparator, have con- observed with paracetamol and the occasional liver reac-
firmed earlier studies which have shown that ibuprofen has tions from aspirin. (2) The pharmacokinetic properties of
comparable therapeutic benefits with coxibs and other ibuprofen, especially the short plasma half-life of elimi-
NSAIDs. For long-term usage (6? months) there are greater nation, lack of development of pathologically related
numbers of drop-outs due to reduced effectiveness of metabolites (e.g. covalent modification of liver proteins by
therapy, a feature which is common with NSAIDs. Spon- the quinine–imine metabolite of paracetamol or irreversible
taneous reports of adverse events and adverse drug acetylation of biomolecules by aspirin) are support for the
reactions (ADRs) in clinical trails from long-term coxib view that these pharmacokinetic and notably metabolic
effects of ibuprofen favour its low toxic potential. (3) The
multiple actions of ibuprofen in controlling inflammation
K. D. Rainsford (&) combine with moderate inhibition of COX-1 and COX-2
Emeritus Professor of Biomedical Sciences,
and low residence time of the drug in the body may account
Biomedical Research Centre, Sheffield Hallam University,
Howard Street, Sheffield S1 1WB, UK for the low GI, CV and renal risks from ibuprofen, espe-
e-mail: k.d.rainsford@shu.ac.uk cially at OTC doses.
276 K. D. Rainsford

Keywords Ibuprofen  Arthritis  Pain  accidental or deliberate ingestion or with serious adverse
Anti-inflammatory  Analgesia  Antipyresis  reactions. Indeed, it has been described as ‘‘the mildest
Prostaglandins  Non-prostaglandin mechanisms  NSAID with the fewest side effects which has been in
Adverse reactions clinical use for a long time’’ (General Practice Notebook,
http://www.gpnotebook.co.uk, accessed 12/11/07).
Abbreviations Ibuprofen was initially introduced in the UK in 1969 and
ADME Absorption, distribution, metabolism and afterwards during the 1970s worldwide as a prescription-
elimination only medication, where it was recommended to be pre-
AEs Adverse events scribed at up to 2,400 mg day-1 (or higher dose in the
AUC Area-under-the curve (plasma USA) for the treatment of musculo-skeletal pain and
concentration) inflammation as well as other painful conditions (Rainsford
CV Cardiovascular 1999a). In the 1970s it was often prescribed either as a first
CoA Coenzyme A line NSAID or in place of aspirin, indomethacin or phe-
COX-1 Cyclo-oxygenase-1 nylbutazone for treatment of arthritic conditions where it
COX-2 Cyclo-oxygenase-2 had a reputation for good efficacy and lower gastrointes-
Coxibs COX-2 selective NSAIDs tinal adverse effects. Initially the drug was used in low
(a sub-group of NSAIDs) doses ranging from 400 to 1,200 mg day-1 and with lati-
Cl/F Clearance (fractional) tude and experience by physicians’ cautious dose-
Cmax Maximal plasma concentration escalation proceeded to the current recommended dosage
CNS Central nervous system of 2,400 mg day-1. The emphasis on cautious use of ibu-
Cp Plasma concentration profen was one of the hallmarks of its early success and the
CYP Cytochrome P450 increasing confidence that it was safe (Rainsford 1999a).
GI Gastrointestinal Over the years there have been many challenges to ibu-
Kel Elimination rate profen, some from concerns about safety including the
NOS Nitric oxide synthase (iNOS, inducible occurrence of some very rare but serious adverse reactions
eNOS, endothelial, nNOS, neuronal) [e.g. Stevens-Johnson and Lyell’s (toxic epidermal necrol-
NSAIDs Non-steroidal anti-inflammatory drugs ysis) syndromes, renal or hepatic failure, necrotising
NS-NSAIDs Non-selective NSAIDs, i.e. those with fasciitis] as well as some that are more common to the class
approximately equivalent COX-1 and of NSAIDs (Khan and Styrt 1997; Zerr et al. 1999; Rainsford
COX-2 inhibitory effects 2003). Most recent of these have been cardiovascular (CV)
OR Odds ratios conditions that were highlighted by the occurrence of myo-
OTC Over-the-counter, non-prescription cardial infarction and cardio-renal symptoms in patients
PMNs Polymorphonuclear leucocyte receiving the newer class of NSAIDs, the coxibs [rofecoxib,
Rac- Racemic valdecoxib and to some extent celecoxib (Ostor and Hazl-
tNSAIDs traditional NSAIDs, e.g. aspirin, ibuprofen, eman 2005; Psaty and Furberg 2005; Rainsford 2005a; Topol
naproxen 2004, 2005)]. This had the effect of regulatory agencies
t1/2 Half-time of elimination worldwide examining the potential of all NSAIDs, to cause
Tmax Time of maximal concentration CV and cardio-renal symptoms, an aspect that is still of
UDP Uridine diphosphate concern for some of the coxibs and some other NSAIDs.
VD Volume of distribution There have also been many challenges from newer
NSAIDs, particularly the wave of some 20–30 new NSA-
IDs introduced in the period of 1970s–1980s and the much
publicised introduction of the selective cyclooxygenase-2
Introduction (COX-2) inhibitors (coxibs) that appeared in 1999 fol-
lowing the discovery of COX-2 as the main prostaglandin
Ibuprofen is one of the most widely used analgesic–anti- synthesizing enzyme expressed in inflammation and pain
pyretic–anti-inflammatory drugs today. It probably ranks pathways (Rainsford 2007). Surprisingly, over half of the
after aspirin and paracetamol in non-prescription over-the- NSAIDs introduced to the clinic since the 1970s have been
counter (OTC) use for the relief of symptoms of acute pain, withdrawn mostly due to unacceptable and unpredictable
inflammation and fever, although the patterns of use of toxicities. In a sense, ibuprofen has survived these chal-
these analgesics vary considerably from country to country. lenges both from the point of view of competition from the
Of these three analgesics, OTC ibuprofen is probably the newer drugs and the inevitable negative impact of the
least toxic, being rarely associated with deaths from failures of other drugs and associated safety issues (e.g. the
Ibuprofen 277

CV risks raised by the coxibs). Mostly, these issues have Naproxen and ketoprofen have been competitors with
concerned prescription-only NSAIDs although those sold ibuprofen, in those countries (e.g. USA, UK and Australia)
OTC like ibuprofen may also have been affected by these where they are marketed for OTC use. Both these drugs are
issues. more potent as anti-inflammatory agents and prostaglandin
Another major competitor has been paracetamol (acet- inhibitors than ibuprofen and are associated with higher
aminophen) especially in the OTC field (Peterson 2005) risk of upper GI adverse reactions at prescription doses
but also, as discussed later, in the therapy of osteoarthritis. (Hersh et al. 2000a, 2007; Milsom et al. 2002). Naproxen
In non-prescription OTC, paediatric use both ibuprofen and tends to be used as a second-line drug for treatment of
paracetamol are equally effective in controlling fever but primary dysmenorrhoea where aspirin, ibuprofen and par-
there are recent data to suggest that combination of these acetamol are found less effective. Ketoprofen is favoured
two drugs may be particularly useful in severe febrile or by some for more severe joint pain in arthritic disease.
painful conditions (Hay et al. 2006, 2009). Arguably, Overall, ibuprofen has withstood competition and chal-
ibuprofen and paracetamol have differing modes of action lenges over the 4 decades since its introduction as a
(see later sections) and so it is possible that they may have prescription drug and over 2 decades since it was intro-
additive or even synergistic effects. duced for OTC sale.
Claims by those advocating paracetamol is that its use is
associated with lower gastrointestinal (GI) and renal
adverse reactions than observed with ibuprofen. For OTC Oral pharmacokinetics
use these differences are minimal or nonexistent (Rainsford
et al. 1997; Bjarnason 2007; Moore 2007). At higher doses Ibuprofen (R/S) in the form that is present in most OTC
used in arthritis therapy the consensus is that the differ- ibuprofen-containing preparations as well as the generic
ences in GI adverse reactions are marginal, while renal preparations sold by prescription (for daily doses up to
adverse reactions may be more prevalent in patients taking 2,400 mg) and non-prescription or OTC sale (for daily doses
ibuprofen. A major issue with paracetamol is hepatotox- up to 1,200 mg) exists as a diastereoisomeric mixture. This
icity especially when taken in the range of 3–4 g daily comprises half as the S(?) enantiomer which is pharmaco-
long-term and with alcohol. The situation about con- logically active as a prostaglandin (PG) synthesis inhibitor
sumption of alcohol with paracetamol and the use of this and the other half mass as R(-) ibuprofen which is less
drug in patients with alcoholic liver disease or signs of active as a PG synthesis inhibitor but which may have some
alcohol abuse is quite serious. These aspects have been pharmacological properties relevant to the anti-inflamma-
considered extensively elsewhere (Novak and Lewis 2003; tory actions of ibuprofen (Brocks and Jamali 1999;
Larson et al. 2005; Myers et al. 2007; Suzuki et al. 2009). Rainsford 1999b, 2003; Graham and Williams 2004). About

Fig. 1 Conversion of R(-)-


ibuprofen to its S(?)
enantiomer via catalytic activity
of fatty acyl coenzyme
thioesterase. From Graham and
Williams (2004) reproduced
with permission of the
publishers, Taylor & Francis,
London
278 K. D. Rainsford

40–60% of the R(-)-form of ibuprofen is metabolically CYP-2C8 (Fig. 2, Graham and Williams 2004). There
converted to the S(?) form (Fig. 1) (Rudy et al. 1991; appears to be differential involvement of these cytochrome
Brocks and Jamali 1999) in the intestinal tract and liver after isoforms on the metabolism of the enantiomers with CYP-
oral absorption (Jeffrey et al. 1991; Jamali et al. 1992). 2C9 favouring formation of S(?)-2 and S(?)-2 hydroxy-
The first step involves the activation of R(-)-ibuprofen ibuprofen and CYP-2C8 favouring R(-)-2-hydroxyibupro-
with ATP Mg to form the AMP-derivative which is then fen formation (Hamman et al. 1997). Inhibition of CYP-2C8
esterified with coenzyme A by the action of acyl-CoA by administration of gemfibrozil to humans increases the
synthetase. The R-ibuprofen-CoA undergoes epimerization plasma concentrations of R(-)-ibuprofen by about one third
via the actions of epimerase to form S-ibuprofen-CoA along with prolonging the elimination half lives of R(-) and
which is then hydrolysed by a hydrolase to form S-ibu- S(?) by 54 and 34%, respectively, and increase of AUC
profen (Brocks and Jamali 1999) (Fig. 1). values by about 20% (Tornio et al. 2007). This suggests that
CYP-2C8 plays a major role in oxidative metabolism of the
Pharmacokinetics in adults ibuprofen enantiomers. Differences in genotypes of CYP-
2C9 have been associated with marked variations in drugs
Ibuprofen is rapidly absorbed from the upper GI tract with that are metabolized by this isoform (Kirchheiner and
peak values of the R(-) and S(?) drug in the plasma or Brockmöller 2005). Thus, compared with CYP-2C9*1/*1
serum at approximately 1–2 h (Brocks and Jamali 1999; individuals with either *1/*2, *2/*2 *1/*3 or *3/*3 variants
Graham and Williams, 2004; Table 1) with some variations have 88, 78, 72 or 55% reduction in the clearance of the drug,
according to pharmaceutical formulation (Jamali et al. respectively (Kirchheiner and Brockmöller 2005). In Span-
1988; Aiba et al. 1999; Halsas et al. 1999; Higton 1999; ish populations the occurrence of varying allelic frequencies
Scott et al. 1999; Trocóniz et al. 2000; Ding et al. 2007). in CYP-2C*8 and the CYP2C*3 allele have been shown to
The plasma S/R ratio can vary according to the time-release result in decreased metabolism of ibuprofen leading to
characteristics of the formulation with higher ratios being increased AUC0?? and reduced clearance (López-Rodri-
obtained with sustained-release (SR) compared with guez et al. 2008). For other NSAIDs (e.g. celecoxib,
immediate-release (IR) formulations, respectively (Ding diclofenac) there is either increased or decreased clearance
et al. 2007). A liquid or liquigel formulation of ibuprofen in individuals with these isoforms. So there is marked vari-
has proven to be popular for rapid analgesia which is ation in the pharmacokinetics of ibuprofen and other
related to fast absorption (Laska et al. 1986; Seymour et al. NSAIDs according to the CYP-2C9 and CYP2C8 status.
1991; Hersh et al. 2000b). Differential metabolism of S(?) and R(-)-ibuprofen
Table 1 summarizes the pharmacokinetic properties of occurs by the CYP-2C9 and CYP-2C8 with these being
R/S-ibuprofen, the data for which has been derived from referred to as S(?) ibuprofen and R(-)-ibuprofen
various studies in volunteers and patients with arthritic hydroxylase activities, respectively (Kirchheiner et al.
conditions. 2002). The allelic frequencies of these CYP isoenzymes the
As can be seen from the data in Table 1 the mean (±SD) 3 ascribed to CYP-2C9 comprise the wild type CYP-2C9*1
values for many of these parameters in adults show which is characterized by an arginine at codon 359 on the
remarkable consistency from the different studies and gene. In the variant CYP-2C9*2 this arginine is replace by
indicate that ibuprofen has, in general, predictable and cysteine, and in the variant CYP-2C9*3 the isoleucine-359
reliable kinetic properties. Furthermore, there are dose- is replaced by leucine. In vitro studies and human PK
related plasma concentration, Cp, and to some extent AUC studies have shown that CYP-2C9*2 has only slightly less
values but the kinetic constants reflected by t1/2 (or the activity than that of the wild type CYP2C9*1 whereas that
inverse, Kel) suggest that there is little variation with dos- of CYP-2C9*3 is 10–30% less so (Kirchheiner et al. 2002).
age. There is also little variation of these kinetic parameters In comparisons of the pharmacokinetics of the S(?)
with repeated dosage. enantiomer the rates of clearance were found to parallel the
The pathways of oxidative metabolism of ibuprofen are enzymic activity with subjects having the CYP-2C9*1/*2
shown in Fig. 2. These principally involve cytochrome and *3/*3 variants having 27 and 53% less clearance than
P450 2C9 (CYP-2C9), CYP-2C8 and 2C19 participating in those with the wild type *1/*1 genotype (Kirchheiner et al.
the oxidation of the alkyl side chain to hydroxyl and car- 2002).
boxyl derivatives. Other studies have examined the role of CYP-2C9 and
Phase 1 Metabolism of R(-) and S(?) ibuprofen involves CYP-2C19 polymorphisms for associations with drug-
hydroxylation of the isobutyl chains to 2 or 3-hydroxy- induced idiosyncratic reactions (Pachkoria et al. 2007).
derivatives and subsequent oxidation to 3-carboxy-ibupro- While arguably liver reactions from NSAIDs may be
fen and p-carboxy-2-propionate; these oxidative reactions associated with abnormalities of phase 1 and phase 2
being catalyzed by cytochromes P450 2C9 (CYP-2C9) and metabolism, the studies by Pachkoria et al. (2007) have
Ibuprofen 279

Fig. 2 Oxidative metabolism of


ibuprofen. (From Graham and
Williams, 2004; reproduced
with permission of the
publishers, Taylor & Francis,
London)

failed to establish if polymorphisms of CYP-2C9 or CYP- ibuprofen and higher S(?)/R(-) ratios have been observed
2C19 are associated with liver disease. in patients with a variety of chronic inflammatory diseases
There do not appear to be any differences in R(-)/S(?) who have impaired renal function (Chen and Chen 1995).
pharmacokinetics with sex in adults (Knights et al. 1995; Surgical removal of wisdom teeth has been found to
Walker and Carmody 1998) with the exception of the cause substantial reduction in the serum concentrations of
volume of distribution VD/F being about twice that in adult both R(-) and S(?)-ibuprofen (by 2.6 and 3.5-fold,
females compared with males (Walker and Carmody respectively) with prolonged Tmax from approximately 1 h
1998). As noted later there are age differences in plasma to 4–6 h depending on the dose (Jamali and Kunz-Dober
levels and kinetics of ibuprofen; elderly subjects have 1999). It was suggested that a number of stress-related
slightly prolonged values of elimination half life for the factors influencing GI functions could contribute to
total drug concentrations (Albert and Gernaat 1984) or the reduced gastric absorption of ibuprofen (Jamali and Kunz-
AUC for S(?) ibuprofen (Chen and Chen 1995). Dober 1999). These observations are of particular thera-
Compromised liver metabolism in patients with mod- peutic importance for they argue in favour of higher doses
erate to severe cirrhosis leads to prolongation of the t1/2 to of the drug for pre-emptive or post-operative surgery.
3.1 h and 3.4 h for R(-) and S(?) ibuprofen with evidence Phase II metabolism involves formation of phenolic and
of reduced metabolic inversion of the R(-) to S(?) acyl glucuronides (Rudy et al. 1991; Kepp et al. 1997;
enantiomer (Li et al. 1993). Alcoholic liver disease also Brocks and Jamali 1999; Graham and Hicks 2004) and a
prolongs the Tmax for the total Cp as well as the t1/2 (Albert minor route of conjugation with taurine which is stereo-
and Gernaat 1984). Elevation of the AUC for S(?) specific to the S(?) enantiomer because of formation from
280

Table 1 Pharmacokinetic properties of oral ibuprofen in adults


Dose of R(-)-ibuprofen S(?)-ibuprofen Reference
Ibuprofen
Cmax Tmax t1/2 AUC CI/F VD Cmax Tmax t1/2 AUC CI/F VD
(lg mL-1) (h) (h) (lg h mL-1) (L h) (L) (lg mL-1) (h) (h) (lg h mL-1) (L h) (L)

400 mg 15.4 1.0 1.6 44.5 16.2 1.0 2.4 67.4 Hynninen et al. (2006),
Cheng (1994)
400 mg 1.74 3.52 7.8 23.5 3.0 1.7 100.8 3.4 4.6 Ding et al. (2007)
600 mg (IR) 20.8 2.96 1.2 65.9 5.0 6.45 23.9 2.4 90.8 Jamali et al. (1988)
600 mg III 20.3 2.0 56.9 26.7 2.2 100.2
600 mg IV 17.3 2.2 51.5 15.6 2.0 2.6 63.9 Tornio et al. (2007)
400 mg 16.4 1.5 2.9 50.1 19.0 1.64 2.18 75.0 Suri et al. (1997)
3.3 [286.8] Chen and Chen (1995)
2
400 mg 17.8 1.59 1.33 52.2 4.0 7.46 26.0 1.3 2.0 93 1.0 Cox et al. (1991)
800 mg (Boots) 3.1 23.9 2.6 11.0 16.8 1.9 2.2 32.4 Geisslinger et al. (1989)
800 mg (Motrin) 24 1.1 1.8 67 1.52 16.4 1.8 2.3 91.7 Geisslinger et al. (1990)
(Arthritic pts.)
600 mg 14.8 1.7 1.8 57.2 5.37 17.8 20.3 2.4 2.1 86.2 13.3 Geisslinger et al. (1993)
600 mg 14.3 1.5 2.3 54.3 5.66 18.7 11.3 1.13 1.9 43.7 Evans et al. (1990)
600 mg (arthritic pts.) 2.3 2.0 67.6 14.0 16.9 1.87 3.4 75.4 Evans et al. (1990)
200 mg 10 1.1 1.9 23.9 4.19 11.2 31.6 1.89 2.0 141 Evans et al. (1990)
400 mg 14.1 1.4 3.2 41.8 4.78 22.3 47.7 2.19 2.0 216 Evans et al. (1990)
800 mg 24.4 1.6 2.1 73.6 5.44 16.7 6.1 1.0 19.5 Jamali and Kunz-Dober
(1999)
1,200 mg 29.5 1.6 4.2 86.1 6.97 42.6 14.5 1.0 45.5
200 mg (pre-surgery) 6.1 1.0 17.7 8.7 1.1 31.6
600 mg 14.8 1.0 35.3 16.9 1.63 2.65 70.4
Mean (SD) 13.1 2.02 2.15 78.2
200 mg 8.1 1.1 20.8 28.8 1.6 2.4 117
400 mg 15.4 1.3 2.7 47.2 4.1 12.5
600 mg 17.1 1.9 1.7 55.5 4.0 14.3
800 mg 24.2 1.4 2.3 70.0 3.2 13.9
K. D. Rainsford
Ibuprofen 281

the thioester CoA which participates in the R(-) to S(?) fluid concentrations of the enantiomers vary less than those
conversion (Shirley et al. 1994). Biliary excretion in humans in plasma and plasma values after about 5.5 h; it being
of unchanged drug and active phase II metabolites accounts inferred from pharmacokinetic analysis that the accumu-
for about 1% of the drug, which compares with 50% of the lation is primarily of protein-bound drug (Day et al. 1988).
urinary excretion (Schneider et al. 1990). The 15 known Non-bound (to albumin) NSAID concentrations (i.e.
UDP-gluronyltransferases that catalyze formation of glu- free) are generally considered to be those which are phar-
curonides in human liver have been shown to be controlled macologically relevant to the actions of these drugs as well
by 5 UGTIA and 5 UGT2B genes and the development of as being of relevance to the untoward effects of drug–drug
these proceeds from birth to 6 months of age (Strassburg interactions where toxic effects of NSAIDs or other drugs
et al. 2002). Hepatic glucuronidation of ibuprofen has been relate to displacement of one or other following binding to
found to be 24-fold lower in children aged 13–24 months albumin or other plasma proteins. As with many NSAIDs
than in adults (Strassburg et al. 2002). This low level of most of which bind to plasma proteins to around 99%,
detoxification of ibuprofen is clearly an important consid- ibuprofen also is strongly bound to albumin (Brocks and
eration for safe dosage of the drug to infants. Jamali 1999; Graham and Hicks 2004).
Stereospecific disposition of ibuprofen enantiomers The binding of ibuprofen in plasma compared with
occurs into the synovial fluids of arthritic patients, many of synovial fluids depends on the concentrations of albumin in
whom have synovitis or inflammation of their knees. There these compartments (Wanwimolruk et al. 1983). There are
is appreciable accumulation of R/S-ibuprofen in synovial differences in the affinity of S(?) compared with R(-)
fluids with broad peaks occurring over a period of 2–6 h ibuprofen for binding on human serum albumin; there being
which follows the peak plasma or serum concentrations two binding sites for both ibuprofen enantiomers of varying
(Glass and Swannell 1978; Mäkelä et al. 1981; Albert and affinity (Hage et al. 1995; Itoh et al. 1997) with some
Gernaat 1984; Gallo et al. 1986). The ratios of total ibu- evidence of allosteric cooperative binding at high concen-
profen concentrations in the synovial fluid to those in trations of S(?) ibuprofen (Hage et al. 1995). There may be
plasma is about 1.24 at 7 h following single dose of greater binding of S(?) ibuprofen to the site II (diazepam)
600 mg of the drug and 0.52–1.46 at 3–12 h after 3 daily binding site of albumin (Cheruvallath et al. 1997).
doses of ibuprofen 1.8 g day-1 (Gallo et al. 1986). The Ibuprofen accumulates in the cerebrospinal fluid (CSF) of
mean free total ibuprofen in synovial fluid ranges from 1.81 patients (undergoing lumbar puncture for treatment of nerve
to 2.91% compared with that in plasma which is 1.54– root compression) with the AUC’s of the R and S enantio-
2.53%. Thus, there is appreciable total and free R/S-ibu- mers in the CFS being 0.9 and 1.5% those in plasma,
profen that accumulates in synovial fluids of arthritic respectively (Bannwarth et al. 1995). The estimated t1/2’s
patients and clearly this will have therapeutic significance were 3.9 and 7.9 h for the R(-) and S(?) enantiomers
in relation to the local anti-inflammatory and analgesic (compared with 1.7 and 2.5 h in plasma). The pathological
effects of the drug in pain control. In studies of the dis- condition being treated in these patients involves neuro-
position of the individual enantiomers it has been found inflammatory reactions with associated drug accumulation
that the concentrations of the S(?) isomer as well as values in the central nervous system, so these observations may
of AUC S(?) always exceed those of the R(-) enantiomer have relevance to central analgesic actions of ibuprofen.
(Day et al. 1988; Cox et al. 1991; Geisslinger et al. 1993; Two other routes of metabolism of ibuprofen which
Seideman et al. 1994) with similar selective accumulation though they do not appear to have clear pharmacological
being shown in experimentally induced skin suction blis- significance have been considered to be of theoretical
ters (Seideman et al. 1994). The patterns of synovial fluid significance. Thus, the stereoselective uptake of R(-)
accumulation of the enantiomers follows that of the peak ibuprofen forming mixed hybrid triglycerides as a result of
plasma levels with broad peaks of R(-) and S(?) ibu- thioester-CoA formation and subsequent fatty acid metab-
profen at about 2–4 h and extending to about 12–15 h olism (Williams et al. 1986) has been considered important
(Seideman et al. 1994) thus showing persistence of the though wanting for evidence for its significance either
enantiomers in synovial fluids well past those of the peak toxicologically or pharmacologically.
plasma concentrations of these enantiomers. The acylation of proteins to form adducts from reaction
As with many other NSAIDs inter-subject variability is of acyl glucuronides of ibuprofen (Castillo et al. 1995;
a common feature of the pharmacokinetics of ibuprofen Vandenhoeven et al. 2006) may be of relevance for adverse
(Wagener and Vögtle-Junkert 1996) which does not relate reactions in the liver analogous to those occurring with
to variation in the rates of inversion of R(-) to S(?) ibu- some other NSAIDs though evidence for this is lacking.
profen (Geisslinger et al. 1993). The kinetics of the In summary, the pharmacokinetic properties of ibupro-
enantiomers appears similar after multiple compared with fen (Brocks and Jamali 1999; Graham and Williams 2004)
single dosage of the racemate (Cox et al. 1991). Synovial can be summarized thus:
282 K. D. Rainsford

(1) There are relatively fast rates of absorption of the neonates, infants and children has considerable impact on
drug with subsequent ‘‘first pass’’ liver phase 1 and the absorption, distribution, metabolism and elimination
phase 2 metabolism to well-characterized (a) phenolic (ADME) of drugs (Kearns and Reed 1989). Among the
and carboxylic acid derivatives via CYP-2C8, CYP- notable differences in drug disposition between infants and
2C9 and possibly CYP-2C19 activities, and (b) children compared with neonates and young adults are the
conjugates with glucuronic acid and taurine (a minor decline in total body water (TBW), increase in intracellular
metabolite). water (ICW) and reduced extracellular water (ECW) that
(2) The biodisposition of ibuprofen reflects high plasma occurs post-natally up to about 1–2 years with subsequent
protein binding and low volume of distribution but variable increases in TBW and ICW up to about 25 years
with the capacity to be accumulated in appreciable where upon those in males are greater than in females
quantities in inflamed compartments where there is (Kearns and Reed 1989). Likewise, fat content increases
need for anti-inflammatory/analgesic activity (syno- post-natally up to about 1 year, plateaus to mid-teens then
vial fluids, CSF). declining until mid 20s where upon differences occur
(3) Ibuprofen has a relatively short plasma elimination between the sexes with females having greater body fat
half-life and although prolonged in liver and renal than males (Kearns and Reed 1989). These changes in body
diseases this is not so appreciable as to be a factor composition along with plasma protein concentrations will
accounting for higher frequency of adverse events in have marked effects on the volume of distribution of drugs.
patients with these conditions compared with those Protein binding of drugs is influenced by a variety of fac-
with relatively normal hepato-renal functions. Indeed, tors and the total concentration of plasma proteins is
the short plasma half-life (t1/2) has been suggested as decreased in the neonate and infant compared with that in
a factor accounting for relatively low incidence of children (Kearns and Reed 1989).
serious GI events compared with traditional NSAIDs Age-related changes in hepatic biotransformation are
(bleeding, peptic ulcers) (Henry et al. 1996, 1998). evident from the neonatal period where they are not
(4) Ibuprofen exhibits approximately linear kinetics to developed and approximate adult values by about
within 1,200 mg dosage or near compliance with 6 months (Kearns and Reed 1989). Thus, Phase I hepatic
expected kinetics. Thus, when taken up to 1,200 mg transformation progressively increases to near adult values
repeated doses of ibuprofen the elimination is not by 6 months of age (Kearns and Reed 1989; Kearns
saturated. 1993). Glucuronidation increases from ±10 days to
(5) Chronic disease states (arthritis) have relatively little 2 months while conjugation with amino acids increases
impact on the overall kinetics of ibuprofen. However, from ±10 days to 3 months (Kearns and Reed 1989;
acute surgical pain reduces the plasma concentrations Strassburg et al. 2002). These physiological factors have
of R(-) and S(?)-ibuprofen which may arise from considerable significance in the age-related pharmacoki-
the stressful conditions of the surgery. This has been netics of paracetamol and NSAIDs in children (Walson
suggested as evidence to necessitate considering and Mortensen 1989; Jacqz-Aigrain and Anderson 2006).
dosage adjustment in the therapy of acute surgical Changes (usually as increased values) of t1/2, VD, and C of
pain on the basis of allowance for increasing dosage these drugs occur progressively from the neonatal period
to meet adequate pain control. to 1–3 years, whereupon they often (but not always)
(6) The t1/2, AUC, VD and clearance kinetics of conven- assume adult values. Of particular relevance is the pos-
tional ibuprofen tablets suggest that the usual dosage sibility that the febrile state which is often treated with
regime of either 400 mg t.i.d. for OTC use or 400– analgesics/NSAIDs can influence drug metabolism and
800 mg t.i.d. or q.i.d. as appropriate for prescription biodisposition as a consequence of release of cytokines
use to 2,400 mg daily. Extended-release formulations [e.g. interleukin-1 (IL-1), tumour necrosis factor-a (TNF-
that have been developed could enable twice a)] during infections which are pyrogenic and alter drug
daily dosage to limits of 1,200 mg day-1 OTC or metabolism.
2,400 mg day-1 prescription requirements. The pharmacokinetics and what is known of the phar-
macodynamic properties of ibuprofen in \12 year children
can be considered to be similar to that of young-middle
Pharmacokinetics in children aged adults in whom most investigations have been per-
formed. A possible caveat to this application of adult
Before considering some of the factors affecting the PK of pharmacokinetics to that in children/infants (\12 year)
ibuprofen in young adults and children it is useful to might be related to differences in growth rates thus
consider some of the differences in drug metabolism and affecting body mass and gender affecting hormonal regu-
disposition in these groups. Physiological development in lation of drug metabolizing enzymes.
Ibuprofen 283

Table 2 Pharmacokinetics of Ibuprofen in children Given these provisos it should be possible to describe
Oral absorption t1/2: 0.3–0.9 h
the pharmacokinetic (PK) and pharmacodynamic (PD)
Tmax: 1–2 h
properties of ibuprofen in\12 year olds as in general being
related to the information that is available in adults. A few
10 mg kg-1 ? Cmax: 44 mg L-1
PK studies have been performed in children that overlap
Protein binding 99%
the 12–18 year age group suffice to conclude that, in
Active isomer S(?)
general, the PK properties are similar to those in adults.
Plasma concentration S(?) children \ adults
Less is known about PD properties in the young (12–
Metabolism CYP450 2C9 and 2C8
18 year) except that dose-related pain relief is similar in
t1/2 0.9–2.3 h
young adults to that in younger children and also in older
From Autret-Leca (2003) adults. This suggests that the PD properties of ibuprofen
shown in studies in adult populations, and supported by in
With the exception of the conversion of R(-)-ibuprofen vitro as well as animal model investigations are likely to be
to its S(?) enantiomer the pharmacokinetic parameters of similar in all age groups.
ibuprofen in children are comparable with those in adults Some pharmacokinetic parameters for ibuprofen in chil-
(Table 1 c.f. Table 2). It appears that the rate of conversion dren of various ages are shown in Table 3. In essence, the
of R(-) ibuprofen is lower in children than it adults major overall features that vary in children compared with
(Table 2). In a study in 11 infants (6–18 months) the young to mid-aged adults are (1) the greater proportion of the
plasma levels of the S(?) enantiomer of ibuprofen were S(?)-enantiomer compared with the R(-)-form in plasma,
lower than in adults while the values for t1/2 for R(-) and and (2) the greater variation in t1/2 (Autret-Leca 2003; Jacqz-
S(?) ibuprofen are within the range of those expected in Aigrain and Anderson 2006). The VD for S(?) ibuprofen in
older children or adults (Kauffman and Nelson 1992; see children is greater than in adults (Kelley et al. 1992) and may
also Jacqz-Aigrain and Anderson 2006). It is suggested that reflect a higher unbound fraction in plasma compared with
the relatively low levels of S(?) ibuprofen argue for a that of R(-) ibuprofen (Brocks and Jamali 1999).
higher dosage of ibuprofen in infants. Inspection of Table 3 shows that the t1/2 and VD of
Other appreciable changes in paediatric populations ibuprofen in patients receiving i.v. drugs is about 25-fold
have been observed in young children aged less than higher than from orally-administered ibuprofen yet there is
5 years where the clearance (CL/F) and volume of distri- the same order of elimination and distribution of oral
bution (VD/F) may be less than that in adults or older ibuprofen from an early age of about 0.5 year; thereafter
children and the plasma half-life of elimination (t1/2) pro- the t1/2 and VD are within the range of that in adults. The
longed to about twice that in adults or older children rates of clearance are, however, greater in young children
(Jacqz-Aigrain and Anderson 2006) (Table 3). up to about 5 years and decline in higher age groups and

Table 3 Pharmacokinetic parameter estimates for NSAIDs in paediatric patients modified from Jacqz-Aigrain and Anderson (2006)
Age Formulation CL/F (ml h-1 kg-1) V/F (L kg-1) t1/2 (h)

Ibuprofen
22–31 weeksa iv 2.06 (0.33) 0.062 (0.004) 30.5
28.6 (1.9) weeksa iv 9.49 (6.82) 0.357 (0.121) 43.1 (26.1)
0.5–1.5 years Suspension 110 (40) 0.20 (0.09) 1.6 (0.4)
11 months–11 years Suspension 57.6 0.164 1.97
3 months–12 years Suspension 80 (10) SE 0.16 (0.02) SE 1.44 (0.15)
3 months–12 years Suspension 110 (10) SE 0.22 (0.02) SE 1.37 (0.09)
5.2 (1.7) years Suspension 140 (32) 0.27 (0.11) 1.4 (0.5)
5.2 (2.5) years Tablet 114 (26) 0.26 (0.1) 1.6 (0.4)
4–16 yearsb Suspension/granules 71 (CV 24%) Vc 0.06, Vp 0.1 –
(4.05 L h-1 9 70 kg)-1 (CV 65%)
Variability presented as SD in parentheses, range (x–y) or SE
CL/F, apparent drug plasma clearance; iv, intravenous; t1/2, elimination half-life; V/F, apparent volume of distribution; Vss volume of distribution
at steady state; Vc, initial volume of distribution; Vp, apparent volume of distribution of peripheral compartment
a
Age is gestation age (GA, weeks)
b
Data reported using allometric model. Estimate presented for a 30 kg individual
284 K. D. Rainsford

are appreciably lower in i.v. administered infants (Table 3). the solubilization of drug in body fluids (Florence and
Ibuprofen has lower glomerular filtration in premature Attwood 1998).
infants and this may be a factor accounting for higher t1/2 NSAIDs and paracetamol vary considerably in their
and VD in this group compared with that in adults. rates of absorption when administered rectally (Van Hoo-
gdalem et al. 1991). The formulations of these drugs
clearly are a major factor in influencing their absorption.
Rectal pharmacokinetics For example addition of increasing amounts of lecithin can
delay the rectal absorption of diclofenac (van Hoogdalem
Fundamental physiological and pharmaceutical et al. 1991). The physico-chemical properties of NSAIDs
considerations in drug administration by the rectal route can influence their absorption. Thus, ketoprofen is well-
absorbed with bioavailability approaching that of the orally
Drugs in suppositories administered by the rectal route are administered drug (van Hoogdalem et al. 1991).
placed in intimate contact with the rectal mucosa which is
pH 7.2–7.4 and has a lipoidal barrier (Florence and Attwood Basic studies with rectal ibuprofen
1998). Suppositories are in contact with the mucous mem-
brane of the rectal ampulla which comprises a layer of Amongst the early studies were those performed by Eller
epithelial cells without villi (Florence and Attwood 1998). et al. (1989) in which eight healthy volunteers received
The main blood supply to the rectum is in the superior rectal solution or suspension formulations of ibuprofen for
or haemorrhoidal artery while drug absorption takes place administration rectally in a randomized fashion. The bio-
through the venous network of the submucous plexus which availability for these forms was compared with that of the
then becomes the inferior, middle with superior rectal veins. orally administered drug. In essence, the results showed
The latter two veins connect to the portal veins and thus that both rectal formulations showed similar extent of
transport drugs direct to the liver. The inferior veins enter the bioavailability being about 60% of the oral formulation; the
inferior vena cava and thus by-pass the liver. The proportion Cmax values being 62–67% and the Tmax was longer than
of drug that is absorbed by these two venous routes depends with the oral formulation.
on the extent to which the suppository migrates in its original In their studies, Eller et al. (1989) compared the bio-
or molten form up the intestinal tract. Thus, this can be availability of rectally or orally administered sodium or
variable and so drugs administered rectally may not bypass aluminium salts of ibuprofen as aqueous buffered solutions
the liver (Florence and Attwood 1998). (pH 7.8) or suspensions (pH 5.2) in eight normal healthy,
The factors influencing rectal absorption of drugs non-obese, male subjects using a Latin square design. The
include (a) the melting point and liquefaction properties of rectal solutions were administered as retention enemas
the suppository, and (b) solubility properties of the drug following prior treatment the night before with Phospho-
that initially influence contact of the drug with the mucosa. Soda then 1 h before a Fleet enema. The orally
Aqueous solubility and pKa of the drug influence absorp- administered preparations were taken following an over-
tion from ‘‘fat’’ based or liposoluble drugs. Viscosity of the night fast. Both the rectally administered preparations had
base and excipients or dispersants added to disperse the fat significantly less bioavailable as shown by the AUC values
can influence absorption. The rate-limiting step in drug (Table 4) and were relatively high as shown by the Cmax
absorption for suppositories made from a fatty base is the values compared with the respective oral solutions/sus-
partitioning of the dissolved drug from the molten base, not pensions. However, as expected, the Tmax values were

Table 4 Bioavailability of rectal compared with oral solutions/suspensions of ibuprofen in eight normal, non-obese male human volunteers
Parameter Treatment A Treatment B Treatment C Treatment D
Oral solution Oral suspension Rectal solution Rectal suspension

Peak concentration (lg mL-1) 80.7 (6) 28.7 (28) 50.3 (36) 19.2 (63)
AUC (lg mL-1 h)
0–12 197.8 (12) 164.2 (21) 172.5 (36) 97.6 (73)
0–? 200.3 (12) 179.1 (30) 175.5 (36) 102.9 (74)
Peak time (h) 0.33 (30) 2.12 (28) 1.14 (36) 2.44 (45)
Mean residence time (h) 2.60 (12) 5.99 (23) 3.19 (6) 4.49 (24)
Terminal elimination rate constant (h-1) 0.351 (11) 0.211 (19) 0.344 (6) 0.367 (22)
From Eller et al. (1989)
Ibuprofen 285

longer for the rectally administered preparation than those AUC were greater after administering ibuprofen acid sup-
taken orally (Table 4). The t1/2 were almost identical for positories compared with the lysine salt even though the
the oral and rectal solutions and about 1/3 lower with the latter were more rapidly absorbed.
oral suspension compared with the former or the rectally
administered suspension. Studies with suppository formulations
The rectal solution had greater bioavailability than the
suspension and achieved higher serum Cmax values than the Kyllönen et al. (2005) investigated the R(-) and S(?)
suspension (Table 4). Likewise, the mean residence time pharmacokinetics of Burana (Orion Pharma, Espoo, Fin-
(MRT) was shorter for the rectal solution than the sus- land) suppositories of ibuprofen in 9 full-term infants aged
pension. These basic results showed that the sodium 1–7weeks, 8 infants aged 8–25weeks, 7 infants aged
solution was the preferred salt to be used in any funda- 26–52weeks and 7 adults aged 20–40 years following
mental considerations of suppository formulations. administration of approximately 19–20 mg kg-1 ibuprofen
Glowka (2000) studied the enantiomeric pharmacoki- suppository after induction of anaesthesia for minor gen-
netics in rabbits of suppositories of ibuprofen acid and the eral or orthopaedic surgery in infants or lumbar disc
lysine salt prepared in the lipophilic base, Witepsol H-15 surgery in adults.
and showed that there was no presystemic inversion of R(-) The results summarized in Table 5 show that in all age
to the S(?) enantiomers. The S:R ratios only increased groups ibuprofen was rapidly absorbed from the suppository
after about 1.5 h following administration of both formu- formulation with the Tmax in infants for R/S ibuprofen
lations and were greater with the lysine salt. The values of being 1.6–3.3 h and the t1/2 for absorption (the so-called

Table 5 Pharmacokinetic
(S)-(?)-ibuprofen (R)-(-)-ibuprofen (R, S)-(±)-ibuprofen AUC ratio
variables of the R(-), S(?) and
R/S ibuprofen in infants, Group 1 (n = 5) suppository retained
children and adults that received
suppositories of 20 mg kg-1 Cmax (mg L-1) 29.3 ± 16.2 23.8 ± 9.4 49.2 ± 20.7
racemic ibuprofen Tmax (h) 2.2 ± 1.0  1.8 ± 1.3  1.9 ± 1.2 
perioperatively Chronological t1/2 (h) 2.9 ± 1.8 3.2 ± 2.7 4.6 ± 5.1
   
Physiological t1/2 (h) 5.8 ± 3.5 6.6 ± 5.4 8.9 ± 10.1
AUC (mg L-1 h) 159 ± 81* 112 ± 54 299 ± 69* 1.7 ± 1.1
Group 1 (n = 4) suppository expelled
Cmax (mg L-1) 12.4 ± 6.4 13.4 ± 8.1 25.7 ± 14.2
Cmax (h) 1.9 ± 0.9 1.9 ± 0.9 1.9 ± 0.9
Chronological t1/2 (h) 3.8 ± 2.9 3.1 ± 2.4 2.9 ± 2.1
Physiological t1/2 (h) 7.8 ± 5.8 6.3 ± 5.1 6.0 ± 4.4
AUC (mg L-1 h) 66 ± 40 54 ± 48 108 ± 83 1.6 ± 1.4
Group 2 (n = 8)
Cmax (mg L-1) 38.5 ± 20.7 40.0 ± 21.8 75.6 ± 44.6
   
Tmax (h) 1.6 ± 0.7 1.4 ± 0.8 1.6 ± 0.7 
From Kyllönen et al. (2005),
Chronological t1/2 (h) 1.7 ± 0.4 2.2 ± 0.7 1.9 ± 0.5
reproduced with permission
Physiological t1/2 (h) 3.1 ± 0.9 3.9 ± 1.4 3.4 ± 1.0
Values are mean ± SD. Only
those patients in group 1 in AUC (mg L-1 h) 131 ± 79 124 ± 67 248 ± 153 1.1 ± 0.2
whom the suppository was Group 3 (n = 7)
retained were included in the Cmax (mg L-1) 42.7 ± 16.0 49.7 ± 23.3 87.9 ± 36.6
comparisons between the    
groups 1 and 4 Tmax (h) 1.7 ± 0.3 1.6 ± 0.7 1.6 ± 0.3 
AUC ratio is the ratio of Chronological t1/2 (h) 2.8 ± 1.3 1.8 ± 0.4 2.1 ± 0.7
(S)-(?)-ibuprofen AUC to Physiological t1/2 (h) 4.6 ± 2.3 2.9 ± 0.7 3.6 ± 1.3
that of (R)-(-)-ibuprofen AUC (mg L-1 h) 180 ± 98 167 ± 56 339 ± 136 1.1 ± 0.4
* Significantly (P \ 0.05) Group 4 (n = 7)
different from the
Cmax (mg L-1) 30.1 ± 12.5 30.1 ± 9.9 63.8 ± 20.4
corresponding value in group 1
where the suppository was Tmax (h) 3.5 ± 0.8 2.9 ± 1.0 3.3 ± 0.8
expelled Chronological t1/2 (h) 2.1 ± 0.3 2.5 ± 0.7 2.2 ± 0.4
 
Significantly (P \ 0.05) Physiological t1/2 (h) 2.1 ± 0.3 2.5 ± 0.6 2.2 ± 0.4
different from the AUC (mg L-1 h) 160 ± 65 177 ± 59 334 ± 123 0.9 ± 0.1
corresponding value in group 4
286 K. D. Rainsford

‘‘physiological’’ half-life) being 1.9–2.9 h. In four of the Table 6 Summary of pharmacokinetic aspects of ibuprofen in rela-
youngest group of infants (1–7weeks, Group 1) the Tmax was tion to safety in adults
similar to that in those where the suppository was retained Short t1/2
even though the Cmax values were about 40% less than in the Healthy normal subjects
non-retained suppository group. The only differences in R = 1.6–4.2 h
Tmax for R/S ibuprofen were observed in the adults (group 4) S = 1.9–3.4 h
where this was 3.3 h and so was greater than in all the other OA patients (Dose dependent)
groups (infants) which ranged from 1.6 to 1.9 h. R = 1.7–2.9 h
Surprisingly, the AUC values for the R/S, R(-) and S = 2.0–3.0 h
S(?) isomers were similar in all the groups except, as Renal clearance R/S
expected in the youngest infant group who had expelled Variably affected by arthritic state but to minor extent
suppositories. Increased by 50% at[70 years but age does not otherwise affect PK
The values of Tmax for R(-) ibuprofen ranged from 1.4 With insufficiency increase in AUC (S?) with age and hypertension
to 2.9 h, with the higher value (2.9 h) in adults in contrast
Variability with pain response—increased renal clearance
to the range of values in infants (1.4–1.8 h) and in which
Liver disease
there was no significant difference between the infant
t1/2 increased; AUC (S?) increased; glucuronides decreased
groups but there was between the two older infant groups
Possible small effects of gender
and the adult group. Only in adults was the Tmax greater for
the S(?) isomer (3.5 h) than for the R(-) enantiomers
(1.6–2.2 h). Since the R(-)/S(?) ratios of AUC values
The Boots Company had already experienced problems
were greater in the youngest infant group (1.7 retained and
with ibufenac in causing hepatic reactions in patients with
1.6 expelled suppository groups) compared with those in
rheumatoid arthritis during early stage clinical trials. Fur-
all the other groups (0.9–1.1) this suggests that there is a
thermore, there was a major objective in the pre-clinical
greater rate of conversion of R(-) to S(?) ibuprofen from
programme to discover a drug which was safer to the
suppositories in adults and which is similar to that observed
gastrointestinal tract than was evident with aspirin and
following oral administration of the drug. The t1/2 of
some other NSAIDs at the time. Thus, to obviate the
plasma elimination (so-called chronological half-life) of
possibility that ibuprofen might cause hepatic reactions as
both the racemic ibuprofen as well as the R(-) and S(?)
previously seen with ibufenac evidence was obtained from
enantiomers was greater in the youngest of the infant
radiobelled-drug studies to show that there was a lower rate
groups compared with those in others and adults indicating
of accumulation in the liver with ibuprofen than with
slower rates of elimination in young infants in whom drug
ibufenac (Rainsford 1999b).
metabolising enzymes are not fully developed.
In considering the pharmacokinetic profile of ibuprofen
These results show that rectal administration of ibu-
(Tables 1 c.f. 6) there are several important features that
profen is an easy and effective way of achieving
should be noted;
therapeutic plasma concentrations especially in the peri-
operative and post-operative periods. Except for delayed 1. The drug has a relatively short plasma elimination
absorption of ibuprofen in adults (which may have arisen half-life (t1/2) a feature which has been identified in
because of the stress of the more extensive disc hernia comparative studies of gastrointestinal gastro-ulcerog-
surgery) (e.g. as in dental surgery, see Jamali and Kunz- enicity (Henry et al. 1996) which is probably a key
Dober 1999) and higher physiological and chronological safety feature. The plasma half-life of the drug
half-life in infants aged 1–7weeks there are no major dif- averages between 2 and 3 h with there being some
ferences in pharmacokinetics between infants and adults. inter subject and intra subject variability but not such
Aside from these investigations there do not appear to that this vastly influences half-life values (Brocks and
have been any other published studies on the pharmaco- Jamali 1999; Graham and Williams 2004). Differences
kinetics of rectal ibuprofen in children or infants. have also been observed in the bioconversion of the
inactive (R-) an enantiomer to (S?) enantiomers and
in their clearance under conditions of acute surgical
Pharmacokinetic aspects of importance in the actions pain.
and safety of ibuprofen 2. There is no evidence of accumulation in elderly or
retention in specific body compartments. There is no
During the discovery of ibuprofen pharmacokinetic issues evidence of formation of bio-reactive metabolites
were of particular importance especially in the pre-clinical sufficient to cause covalent modification of liver or
evaluation of the toxicity of the drug (Rainsford 1999a). other proteins that might contribute to toxicity of the
Ibuprofen 287

kind seen in the case of paracetamol-induced irrevers- doses, respectively. The t1/2s were 1.6 h in both groups
ible hepatic injury (Graham and Hicks 2004). and the rates of oral clearance 1.2 ± 0.4 and
Glucuronide conjugates of ibuprofen represent the 1.4 ± 0.5 mL min-1 kg-1 with the two dose levels,
major metabolites of the drug and it has been respectively, showing that the serum PK are unaffected by
speculated that these conjugates might lead to forma- dose. An earlier study by Walson et al. (1989) using liquid
tion of adducts such as seen with other phenyl ibuprofen in febrile children showed that the values of the
propionic and benzoic acid NSAIDs (Castillo et al. racemic Cmax were slightly lower at the 5 mg kg-1 dose than
1995). Whether these conjugates contribute to covalent observed in the study by Nahata et al. (1991) but were within
modification of proteins that leads to toxicity is not the same range.
known. There does not appear to be any indication of In a later study the same group performed a randomized,
any appreciable accumulation of ibuprofen in the liver double-blind, parallel-group placebo-controlled study in 56
and other organs such as might result from such infants and children (0.5–12 year) who were primarily
covalent modification. It is very likely that there is a investigated for antipyretic effects (Nahata et al. 1992).
considerable degree of spontaneous hydrolysis both of They were given 5 and 10 mg kg-1 ibuprofen suspension or
the glucuronides and the ibuprofenyl-derivatives of placebo in separate groups but blood samples for PK assay of
proteins. the plasma concentrations of the racemate in only 17
3. There is little variation in the main PK properties of patients, who received the drug alone. The mean maximal
ibuprofen in different arthritic states. Cmax of the racemate were 28.4 and 43.6 lg mL-1 at 1.0 and
4. Mild/moderate renal impairment does not appear to 1.5 h for the 5 and 10 mg kg-1 dosage groups, respectively.
cause any elongation of the plasma elimination half- These plasma values (Nahata et al. 1992) correspond closely
life and there is little evidence of alterations in plasma with those in serum which were obtained in the earlier study
pharmacokinetics in patients with mild hepatic disease. (Nahata et al. 1991) showing consistency both in plasma c.f.
Clearly, patients with considerable renal impairment or serum and between the studies.
liver malfunctions should not be taking ibuprofen as Another study in febrile patients was performed by
there would be an expected increase in risk of systemic Kauffman and Nelson (1992) in 49 infants and children
accumulation although this risk is probably of a low aged 3 month to 10.4 year, the primary purpose being to
order in comparison with the short plasma half-life of investigate the relationship between plasma concentration
the drug. of the racemic form of the drug and antipyretic effects.
Fever was diagnosed as arising from a variety of conditions
Paediatric pharmacokinetics in the febrile state including pneumonia, otitis media, upper respiratory tract
infection, tonsillo-pharyngitis and various other conditions.
Among the most frequent indications for use of ibuprofen The dose of ibuprofen was 8 mg kg-1 which is between
in children is for the treatment of fever. Since febrile that of 5 and 10 mg kg-1 used in the earlier studies. Fur-
conditions lead to elevation of febrile-inducing pro- ther discussion about the therapeutic effects of this and
inflammatory cytokines (especially IL-1b, TNFa, IL-6) and other studies reported in this section will be considered in
these can lead to alterations in the activities of drug- the next section. However, it was found that there was a
metabolizing enzymes it is important to understand if the delay in peak concentration of ibuprofen and maximal
pharmacokinetics of antipyretic agents is altered in febrile decrease in temperature, highlighting that the therapeutic
conditions in children. Earlier reviews (e.g. Walson and benefit follows the peak or optimal plasma concentrations
Mortensen 1989) emphasized the lack of PK data in chil- of the drug.
dren; a situation that has been addressed more extensively The kinetic analysis showed that there was no affect of
in recent years, although there are still some gaps in age on the pharmacokinetic properties of the drug in a final
knowledge of PKs of antipyretic/analgesic drugs especially group of 38 patients. The results showed that oral ibuprofen
in infants. suspension was rapidly absorbed with a Cmax of 35.8 ±
Nahata et al. (1991) studied the PK of ibuprofen in 17 16.7 lg mL-1 (mean ± SD) at 0.7 ± 0.5 h (mean ± SD).
patients (aged 3–10 year) with fever from streptococcal The absorption was faster than that in earlier studies and
pharyngitis and otitis media who received 5 and 10 mg kg-1 similarly the half-life of absorption was fast (t1/2abs
liquid formulation of the drug (mean ± SD ages for this 0.3 ± 0.3 h). The plasma elimination t1/2 was 1.6 ± 0.7
group being 6.7 ± 2.5 and 6.2 ± 2.1 year.) The peak (mean ± SD) h which was within the range observed in
(mean ± SD) serum concentrations of the total of the race- other studies and in adults.
mate [i.e. R(-) and S(?)-ibuprofen] in these two Brown et al. (1992) investigated the disposition of 5 or
groups were 28.4 ± 7.5 and 43.6 ± 18.6 lg mL-1 which 10 mg kg-1 ibuprofen and 12.5 mg kg-1 paracetamol in
were evident at 1.1 and 1.2 h, with the 5 and 10 mg kg-1 153 febrile children. The Cmax occurred about 2‘ h earlier
288 K. D. Rainsford

than the maximal antipyresis with both drugs thus being in expected to have profound consequences for drug metab-
agreement with the study of Kauffman and Nelson (1992). olism and biodisposition in these patients.
The plasma AUC0–00 was lower for the high dose of ibu- From a pharmacokinetic viewpoint there are indications
profen than the lower, an observation which is at variance that alterations in serum/plasma albumin concentrations,
with that obtained in other studies. especially if subnormal in severe disease may affect the
In a investigation of the pharmacokinetics of the R(-) and percentage free concentrations of NSAIDs in the circula-
S(?) enantiomers of ibuprofen in febrile children, Kelley tion, which might have some toxicological or therapeutic
et al. (1992) employed a randomized, open-label parallel consequences (Skeith and Jamali 1991; Furst 1992;
study design in which 39 patients (11 month–11.5 year) Litalien and Jacqz-Aigrain 2001).
received 6 mg kg-1 ibuprofen suspension or 5–10 mg kg-1 Before the introduction of ibuprofen, aspirin was the
paracetamol. For some unexplained reason only values of most widely and successfully used drug for treating pain
Cmax being 33.5 ± 14.7 (mean ± SD) lg mL-1 and Tmax and joint symptoms in juvenile arthritis (Ansell 1973,
being 60 ± 19.7 min were recorded but not the values for 1983).
the individual enantiomers. Adverse effects are, however, more frequent with aspi-
rin as well as some of the other NSAIDs (indomethacin,
Pharmacokinetics during analgesia meclofenamic acid, naproxen; Skeith and Jamali 1991)
whereas these appear fewer with ibuprofen (Ansell 1983;
The disposition of the enantiomers was studied in 11 Furst 1992).
infants (6–18 months), who were anaesthetized for minor The dose of ibuprofen employed in juvenile arthritis
genitor-urinary surgery and given 7.6 ± 0.3 mg kg-1 (30–40 mg kg-1 day-1) is much higher than generally
ibuprofen oral suspension post-operatively (Re et al. 1994). employed in infants and children for the treatment of fever
The values of racemic, S(?) and R(-) were 24.4 ± 6.6, and painful conditions (5–10 mg kg-1 day-1) and is more
9.7 ± 2.9 and 11.8 ± 4.4 lg mL-1 at Tmax approximately in line with that employed in cystic fibrosis. Reference to
2–4 h, respectively. It was apparent from these studies that the extensively studied PK properties of ibuprofen in CF
the peak plasma concentrations were much longer than may, therefore, be instructive and complement those in
those observed in the previous studies in febrile infants and juvenile arthritis.
children suggesting that either the surgical-anaesthetic Among the earlier published reports of the PK of ibu-
procedure delayed GI absorption of the drug or the age of profen in juvenile arthritis were two studies by Mäkelä
the infants influenced the PK of ibuprofen. The lower S/R et al. (1979, 1981). These authors determined the concen-
ratio obtained is in contrast to that of other investigators in trations of racemic drug in serum and synovial fluids in 17
infants where this was higher. patients with juvenile arthritis (aged 1.5–15 years) who
There are three paediatric groups where ibuprofen has received approximately 40 mg kg-1 day-1 ibuprofen. A
been investigated for therapeutic benefits in relation to striking feature of this study was that although there was a
pharmacokinetic properties. These are for relief of pain and high degree of variability in the serum and synovial fluid
joint symptoms in juvenile idiopathic (chronic) or juvenile concentrations that the ratio of ibuprofen in the synovial
rheumatoid arthritis (JIA, JRA, respectively), the i.v. fluids was relatively high compared with that in the serum
administration in patent ductus arteriosus (PDA) and in high (Fig. 3). The absorption of oral ibuprofen is rapid and
doses in cystic fibrosis (CF). While both these treatments may comparable to that in adults (Mäkelä et al. 1979, 1981). In
be considered outside the norm none-the-less they are 33 patients (1.5–15 years) that received approximately
potentially important uses of the drug in therapy. Moreover, 40 mg kg-1 day-1 t.i.d., peak serum concentrations Cmax
these studies provide important therapeutic data on the were 31 lg mL-1 at 1.0–2.0 h. The t1/2 was 2.3 h which is
pharmacokinetic properties of ibuprofen in extremes of dos- comparable with that in adults.
age and administration which, with safety data, are important The relationship of serum/plasma concentrations to
for giving outside values for indications for the drug. therapeutic efficacy appears to be best understood by
comparing the concentrations of the pharmacologically
Pharmacokinetics in patients with juvenile rheumatoid active S(?) enantiomers (Skeith and Jamali 1991).
(or idiopathic) arthritis
Cystic fibrosis
Limited studies have been undertaken to investigate the
pharmacokinetics of oral ibuprofen in patients with juve- Though not a recognised indication for the drug it has been
nile rheumatoid arthritis (JRA; juvenile idiopathic (or explored for therapy of cystic fibrosis and these studies are
chronic) arthritis, JIA). The production of pro-inflamma- useful for consideration of safety and efficacy of ibuprofen.
tory cytokines and other inflammatory reactions would be In particular, data on the PK of ibuprofen in cystic fibrosis
Ibuprofen 289

Fig. 3 Reproduced with


permission of the publishers of
Clinical Pharmacokinetics of
oral ibuprofen in juvenile
rheumatoid arthritis. Redrawn
from Skeith and Jamali (1991)

(CF) are quite extensive and are instructive as they give Tmax values were 68, 128, 109 min indicating that at the
information on drug disposition at high dosages, in a highest dose there was some limitation to gastric absorp-
condition where there is considerable pulmonary (often tion. Indeed, there are indications of absorption effects and
with accompanying Pseudomonas or other bacterial a wide scattering of Cmax data in relation to dose (mg kg-1)
infections) and gastrointestinal inflammation as well as of ibuprofen suggesting that some of the GI effects of the
systemic manifestations of disease some of the respiratory disease (hyper mucus secretion) may influence absorption
inflammatory symptoms of which are markedly amelio- of the drug. Compared with PK in normal adults or those
rated by ibuprofen. Thus, there is considerable interest in with arthritic diseases (Tables 4, 5) the values of Cmax and
these data from safety aspects. Tmax are higher by a twofold factor or greater. The values
Konstan et al. (1995) have pioneered use of ibuprofen of AUC (5.8, 6.3 and 10.8 mg min-1 mL-1) for the three
for treating CF. In a randomized, double-blind, placebo- doses also appear higher than in adults with the rates of
controlled, dose-escalating study in 19 children (6– clearance (1.8, 2.1, 1.9 ml min kg-1) being relatively low.
12 years) in Ohio (USA) Konstan et al. (1991) compared The t1/2 was approximately 68, 128 and 109 hr for each
the plasma PK of racemic (R/S) ibuprofen based on its dosage level reflecting extension of residence time of the
enantiomers (assayed by HPLC) following administration drug in the body. Thus, these investigations show that there
of 300 mg o.d. of the drug for the first month, 400 mg o.d. are marked differences in the PK of ibuprofen in CF
in the second month and 600 mg o.d. in the third month. patients compared with young or mid-aged adults.
The dose of ibuprofen was increased if the peak plasma In the detailed second part of this investigation the
level was B50 lg mL-1 and if placebo then this was also plasma concentrations and AUC values in 13 CF patients
increased. The PK of ibuprofen was also investigated in 13 (6.1 ± 1.7; mean ± SD mg min-1 mL-1) were signifi-
children who received 13.4 ± 4.1 mg kg-1 (mean ± SD) cantly lower than in controls (11.3 ± 3.4, mean ± SD,
compared with that in 4 normal children who received mg min-1 mL-1) with reduction in clearance by about 1/3
similar doses of the drug. and an approximate equivalent increase in VD. The possible
In the former investigation 11 patients in the ibuprofen reasons for these substantial alterations in PK include
and 5 in the placebo group who completed the study decreased bioavailability (possibly from GI absorption),
underwent 24 PK studies. In the dose escalation study increased metabolic clearance and increased unbound
values of Cmax were 38, 29 and 65 lg mL-1 for the 3 fraction in plasma leading to increased clearance through
dosages 300, 400 and 600 mg day-1, respectively. The the kidneys (Brocks and Jamali 1999).
290 K. D. Rainsford

In another study in North Carolina, USA in 38 children of inflammatory prostanoids (mainly PGE2) (Rainsford 1999b;
both sexes age ranges 2–13 years with CF the enantiomer Burian and Geisslinger 2005; Hinz and Brune 2006). Pain
PKs were investigated in a single-dose, open-label investi- relief is attributed to peripheral (anti-inflammatory) and
gation following 20 mg kg-1 racemic ibuprofen (Dong et al. central nervous system (CNS) effects of S(?) ibuprofen on
2000). The S:R enantiomeric ratio of the plasma AUC was the inducible COX-2 and iNOS/cNOS present in inflamed or
2.09:1 and the free and conjugated ibuprofen in urine was inflammatory cells of the peripherally affected regions as
13.9:1 (S:R) which indicated there were no differences in well as in the dorsal horn and higher spino-thalamic tracts
these parameters compared with those in normal children. mediating pain transmission. There may be some contribu-
While there were no differences observed in other PK tion of inhibition of COX-1 in the CNS to the analgesic
parameters there was an inverse relationship between the CI/ actions of ibuprofen (Martı́nez et al. 2002). Recently, much
F for R(-) ibuprofen with age in CF patients. There was no interest has been shown in the possibility that ibuprofen may
significant difference in PK parameters with sex or formu- enhance the synthesis of endogenous cannabinoids and so
lations (suspensions, tablets) of ibuprofen. contribute to the central analgesia via cannabinoid receptor
The dose of ibuprofen employed by Doyle et al. (2002) activation (Fowler et al. 2009) as well as acting on NMDA
was 20 mg kg-1 and was greater than that in the second receptors (Björkman et al. 1996). There is possibly a con-
PK study by Konstan et al. (1991) (13.4 mg kg-1 in CF tribution of R(-) ibuprofen to the effects on both the
and 13.9 in controls) so the differences in PKs between leucocyte activation neural activity and spinal transmission
these studies might be explained, in part, by differences in influencing the effects of ibuprofen in inflammatory pain. In
dosages even though the actual values for the R(-) and analgesic studies the pharmacokinetics of ibuprofen has
S(?) enantiomers were not clear from the study by Kon- been found to relate to the cortical evoked potentials and
stan and co-workers. subjective pain response following tooth pulp stimulation in
Arranz et al. (2003) investigated the population PK of human volunteers (Suri et al. 1997). These observations
serum ibuprofen in 59 CF patients (2–18 year) in order to indicate that the plasma profile of the active S(?) enantio-
identify the factors accounting for inter individual variability. mer which is the potent PG synthesis inhibitor parallels the
The PK analysis revealed that the inter-individual variability analgesic response and the ex vivo inhibition of PG synthesis
was such that the absorption constant (Ka) could not be in the blood of volunteers (Suri et al. 1997).
estimated accurately. Dose-dependent kinetics was demon- Antipyretic effects of ibuprofen are principally due to the
strated affecting clearance and VD. As would be expected, the effects of S(?) ibuprofen on the synthesis of PGE2, the main
fasting status and formulation (acid or lysine salt) of the signalling mediator of pyresis synthesized in the hypotha-
affected the bioavailability and clearance of ibuprofen while lamic—preoptic region at the base of the brain; pyrogens
slower absorption of the free acid was evident compared with such as bacterial lipopolysaccharides and the cytokines and
that of the lysine salt of ibuprofen. It should be noted that cell breakdown (lytic) products produced from activated
these authors did not assay for the enantiomers of the drug leucocytes being the source of pyrogens that activate
and so the ratio of S:R could be another factor accounting for receptors in the hypothalamic-preoptic area causing pro-
population variability (Re et al. 1994). duction of COX-2 derived PGE2 (Rainsford 1999b). Anti-
inflammatory effects of both R(-) and S(?) ibuprofen on
activated leucocytes at inflamed sites could reduce the
Pharmacodynamic activities release of pyrogenic molecules and so contribute to the anti-
pyretic effects of the drug.
Ibuprofen has multiple actions on different inflammatory Ibuprofen is frequently used in control of menstrual pain
pathways and cellular systems involved in acute and in young adult women (Milsom et al. 2002; Grimes et al.
chronic inflammation (Rainsford, 1999b, 2003). A sum- 2006). The major therapeutic effects of the drug in this
mary of the principle inflammatory reactions that are condition are attributed to the inhibition of PGF2a and
affected by ibuprofen is shown in Table 7. This summary is PGE2 that are responsible for the uterine smooth muscle
essentially based on various experimental observations. In spasm and local inflammatory pain in this condition (Da-
some cases there are paradoxical actions (e.g. on the syn- wood and Khan-Dawood 2007).
thesis of nitric oxide (NO) which are system-dependent and Ibuprofen is also widely used in dental pain, including that
do not always show uniform effects. What is presented here from impacted third molars and in their surgical removal;
is a consensus view of the modes of action of ibuprofen. these being painful conditions frequently experienced by
The principle pharmacodynamic (PD) actions of ibu- young adults (Dionne 1998; Dionne and Cooper 1999;
profen, like that of other NSAIDs, that are involved in Beaver 2003; Derry et al. 2009). Extensive clinical investi-
control of acute pain, fever and acute inflammatory reactions gations have shown the effectiveness of OTC dosages (circa
are the inhibition of COX-1 and COX-2 derived pro- 800–1,200 mg) of ibuprofen in pain relief in the surgical
Ibuprofen 291

Table 7 Summary of modes of action of ibuprofen and its enantiomers


Effect Reference

Inhibition of prostaglandin synthesis


Rac–Ibu: is an equi-potent (unselective) inhibitor of COX-1 and COX-2 Mitchell et al. (1994), Warner et al. (1999), Kawai et al.
(1998), Kargman et al. (1996), Boneberg et al. (1996)
S(?) more potent inhibitor than R(-) on COX-1 and COX-2; no time- Boneberg et al. (1996), Gierse et al. (1999)
dependent but reversible inhibition
R(-) competes with S(?) at active site on COX-1 Rainsford (1999b, 2003)
S(?) more potent inhibitor of COX-1 than COX-2 Boneberg et al. (1996)
Rac-Ibu more potent inhibitor of peroxidase activity of COX-2, than Gierse et al. (1999)
COX-1
Thoester of R(-)-Ibu and S(?) competitive inhibitors of COX-1 and Neupert et al. (1997)
COX-2
Inhibition of leukotriene production
Ca??-ionophore-stimulated LTB4 and 5-lipoxygenase by PMN’s reduced Villanueva et al. (1993)
equally by Rac-, S(?) and R(-) Ibuprofen
Inhibition of leucocyte functions
Reduced migration of PMN’s, expression of ICAM-1, E-Selectin and Hofbauer et al. (2000), Menzel et al. (1999), Zhang
vascular adhesion molecules et al. (2006)
Reduced oxyradical production by fMLP stimulated PMN’s and xanthine Chan et al. (2008), Perez et al. (2007)
oxidase
Reduced release of b-glucuronidase from fMLP-stimulated PMNs Villanueva et al. (1993)
Nitric oxide production and actions
Reduced exhaled NO and urinary nitrite/nitrate in human volunteers and Vandivier et al. (1999), Akbulut et al. (2005)
NO in lungs of rats after endotoxin
Transient increase, followed by sustained decrease of NO by PMNs after Vural et al. (2002)
oral S(?) ibuprofen 400 mg
Decreased NOS protein & mRNA in LPS and cIFN-stimulated Menzel & Kolarz (1997), Stratman et al. (1997),
macrophages or glial cells, but increased iNOS and NO in endothelial cells Aeberhard et al. (1995), Miyamoto et al. (2007)
Decreased NO-mediated vasodilation Hardy et al. (1998)
Decreased NO, peroxynitrite and oxyradical production by human PMNs Costa et al. (2006)
Increased iNOS in stomach after ibu-arginate Tegeder et al. (2001)
Inhibition of production of transcription factors, MAPkinase, nuclear
receptors, heat shock proteins
Decreased NFjB, IjB, IKK, Erk 1/2, pGORSK, NAG-1, HSF-1, Hsp70, Lagunas et al. (2004), Gomez et al. (2005)
PPARa, PPARr
Inhibition of IL-1b-induced cAMP levels Vij et al. (2008)
Inhibition of cytokine production
Reduced production of IL-1b and IL-6 in microdialysates in forearm of skin Angst et al. (2008)
after adjacent sunburn-inflammation by 800 mg but not 400 mg Ibuprofen
in volunteers, coincident with pain reduction
Increased production of c-reactive protein and pro-Inflammatory cytokines Nieman et al. (2006)
in blood of ultramarathon runners following racing
S(?) and R(-) ibuprofen equally active as inhibitors of monocyte IL-1 & Hofbauer et al. (2000)
TNFa
Inhibition of antibody (IgG, IgM) synthesis
Reduced in human peripheral monocytes Bancos et al. (2009)
Apoptosis
Reduction of apoptosis and TNFa induced by Fas signalling in vivo Cazanave et al. (2008)
Reduces NO donor effects on cell viability & apoptosis Asanuma et al. (2001)
Anandamide production
Inhibits adadamide hydrolase in nervous system causing increased Fowler et al. (2009)
endogenous cannabinoid (anandamide)
292 K. D. Rainsford

removal of third molars that is superior to placebo and in ibuprofen enantiomer (present as half the mass of ingested
most studies that of paracetamol where it is equal to or more drug) to the prostaglandin synthesis inhibitory or active
effective than this frequently used analgesic (Dionne and S(?) isomer, and (b) these isomers each have pharmaco-
Cooper 1999; Beaver 2003; Derry et al. 2009). Recently, logical effects of importance to the therapeutic actions of
ibuprofen has been used as a reference drug for comparing the drug, thus highlighting the interrelationships between
effects of coxibs in dental pain where it has been found to be its metabolism and pharmacodynamic effects (Fig. 4)
about equally effective in pain relief (Morse et al. 2006; (modified from Rainsford 1999b using information in
Bannwarth and Bérenbaum 2007; Cheung et al. 2007; Geu- Table 7).
sens and Lems 2008). The analgesic effects of ibuprofen in Like many conventional (or traditional) tNSAIDs, ibu-
dental surgery have been attributed to (a) inhibition of the profen inhibits both the constitutive cyclo-oxygenase-1
local (dental) production of PGE2 by S(?) ibuprofen as well (COX-1) which is responsible for production of prosta-
as the anti-oedemic activity of the parent drug (Dionne and noids (PGs and thromboxane A2, TxA2) that control a
McCullagh 1998; Dionne and Cooper 1999), and (b) increase range of physiological or ‘‘housekeeping’’ functions (vas-
in plasma b-endorphins (Troullos et al. 1997); paradoxically, cular, blood flow, gastric and renal functions), and the
however, results from the same group have shown the inducible COX-2 whose synthesis is increased leading to
opposite effects with S(?)-ibuprofen which produced rapid amplified production of PGE2 in inflammation and pain
analgesia (Dionne and McCullagh 1998). Studies in rats (Cryer and Feldman 1998; Rainsford 1999b, 2004a, b;
suggest that ibuprofen (as well as rofecoxib) may reduce the Warner et al. 1999; Vane and Botting 2001). COX-1
expression of COX-2 in dental pulp (Holt et al. 2005). inhibition has been considered a factor underlying the
The effectiveness of ibuprofen in headache and migraine possibility of NSAIDs to cause some adverse effects (GI
has been demonstrated in a number of studies in children and ulcers, bleeding, renal abnormalities) although there are
young adults (Stewart and Lipton 1998; Suthisisang et al. other biochemical and cellular actions of NSAIDs that
2007; Silver et al. 2008). Less is known about the mecha- contribute to their untoward effects (Rainsford 2004a;
nisms of action in these conditions but it is probably due in Bjarnason et al. 2007). The ratio of inhibition of COX-1 to
part to S(?) ibuprofen affecting platelet activation and COX-2 has been considered to relate to likelihood of
thromboxane A2 production and local vascular effects in the developing upper GI and possibly renal and other reactions
affected regions of brain vessels. It is significant that ibu- by NSAIDs in relation to their anti-inflammatory activities
profen can penetrate into the CNS so that this may contribute (Cryer and Feldman 1998; Warner et al. 1999; Vane and
to the central analgesic effects including that in headache and Botting 2001). The newer class of highly selective COX-2
migraine as a consequence of local accumulation of the drug inhibitors, the coxibs, were developed in attempts to reduce
(Bannwarth et al. 1995; McCrory and Fitzgerald 2004; the risks of serious upper GI and other reactions (Rainsford
Kokki et al. 2007). Several experimental studies suggest that 2004b).
S(?) ibuprofen administered intrathecally (i.a.) into the CNS It has been suggested that one the reasons for the low
has direct analgesic effects which are greater than the R(-) gastro-ulcerogenicity of ibuprofen may relate to the
form (Malmberg and Yaksh 1992; Björkman et al. 1996). competition of the COX-1 inactive R(-)-isomer with the
Ibuprofen at OTC doses is extensively used in sports and active S(?)-enantiomer for the active site of COX-1 so
other acute minor injuries where it has proven effectiveness effectively diminishing the potential for inhibition of PG
over placebo and equivalent or superior effects to some other synthesis by the drug (Rainsford 1999b, 2003). Thus,
NSAIDs and non-narcotic analgesics. In this and other acute presence of the R(-)-ibuprofen in the ingested drug may
pain conditions the OTC dosages (*1,200 mg day-1) suf- mask the interaction of the S(?)-isomer with the active
fice to relieve pain although occasionally higher anti- site of COX-1 in the stomach and platelets circulating
arthritic doses (up to 2,400 mg day-1) may be employed. through the gastric circulation so reducing the inhibitory
Most pain studies show placebo effects amounting to potential of the latter isomer on production of gastric
approximately 30% reduction in VAS scores or other indices prostaglandins (Rainsford 1999b). The short plasma
of pain measurement and the effects of ibuprofen are at least elimination half-life of the drug may also be a feature
double this or greater and more prolonged ([6 h) than accounting for low risks of upper GI injury form the drug
placebo (Kean et al. 1999; Rainsford 1999a, b; Moore 2007). (Henry et al. 1996).
To establish what could be regarded as ‘‘clinically-sig-
nificant’’ effects of NSAIDs/coxibs on COX-1 and COX-2
Modes of action in relation to pharmacokinetics activities in humans requires use of either ex vivo whole
blood or in vivo blood or tissue sampling techniques that
What has emerged since ibuprofen was initially discovered are now well-established (Rainsford et al. 1993; Warner
is that (a) the drug undergoes metabolism of inactive R(-)- et al. 1999; Brooks et al. 1999; Shah et al. 2001). Using the
Ibuprofen 293

Fig. 4 Principal metabolic routes of R(-) and S(+)-ibuprofen and actions of metabolites. Modified from Rainsford (1999b) with permission of
the publishers, Taylor & Francis and updated using information from Table 7 and references therein

whole blood assay Blain et al. (2002) compared the effects inhibition ex vivo it was evident that although there was
of ibuprofen, diclofenac and meloxicam on in vitro activ- wide scatter of about 10% of the latter data most fell within
ities of COX isoenzymes using blood from 24 healthy male about 80% inhibition of COX-1 and COX-2 observed in
volunteers and the ex vivo production of COX-1-derived vitro. In comparison after oral intake diclofenac inhibited
TxB2 during clotting and COX-2-derived PGE2 upon COX-1 by 70% and COX-2 by 95 and 97%. COX-1 inhi-
stimulation with endotoxin after the same volunteers took bition from meloxicam was 30 and 55% and COX-2 was 63
single and multiple (3 days for ibuprofen and diclofenac and 83%, respectively after single and repeated doses.
and 5 days with meloxicam) doses of 400 mg ibuprofen A study relating pharmacokinetics of ibuprofen to anal-
(BrufenTM), 75 mg diclofenac SR (VoltarenTM) or 7.5 mg gesic properties from recordings of somatosensory evoked
meloxicam (MobicTM). Plasma concentrations of the drugs potentials (EP) and subjective pain ratings (PR) following
and in the case of ibuprofen the R/S isomers were deter- tooth pulp stimulation was performed using a randomized,
mined and used to relate these as free and unbound double-blind, placebo-controlled 6-way crossover study
concentrations to in vitro inhibition profiles. using ibuprofen 400 mg or flurbiprofen 100 mg each taken
Ibuprofen taken as a single dose or repeatedly inhibited by six healthy volunteers on two separate occasions com-
production of TxB2 by COX-1 by 96 and 90%, respectively. pared with placebo (Suri et al. 1997). A comprehensive
COX-2 production of PGE2 was inhibited by 84 and 76%, analysis of the R(-) and S(?) enantiomers of these drugs
respectively after single and multiple doses. The plasma was also undertaken and the data used to model the PK/PD
concentrations of R(-) and S(?)-ibuprofen at these times properties of these drugs. Both EP and PR data correlated to
were 116.2 (SD, ±38.7) and 70.4 (±1.28) lM, respectively. show that peak analgesia was evident at 2.5 h for ibuprofen;
Thus, almost complete inhibition of both COX-1 and COX-2 the EP data showed prolonged effects of ibuprofen up to 6 h
was achieved under in vivo conditions and this was paral- while the PR data were less so but still extended to the same
leled by the modelling of in vitro inhibition profiles. Near period (Fig. 5). While no data were analyzed in relation to
complete COX-2 inhibition in vitro was achieved at free the enantiomers both these showed peak values of R(-) at
concentrations of the racemate as well as the S(?) enan- 1.33 h and S(?) at 2.18 h inferring that the latter isomer is
tiomer which almost completely inhibited both COX-1 and probably the active form in analgesia in accordance with its
COX-2 in vitro. When these data were modelled with the COX inhibitory properties.
294 K. D. Rainsford

is slightly superior to that of paracetamol, especially if there


is a pronounced inflammatory component to the condition
being treated (McQuay and Moore 1998; 2006).
In children, ibuprofen is principally used for control of
febrile reactions accompanying infections and minor pain
states. Extensive controlled clinical trials and observational
studies have been performed in these conditions. Over-
all, ibuprofen has comparable or greater antipyretic and
analgesic effects in children compared with that of
paracetamol.

Treatment of pain in osteoarthritis, rheumatoid arthritis


and related conditions

Fig. 5 Evoked potentials (EP) and time-dependent development of


There are indications that ibuprofen is used quite exten-
analgesia to ibuprofen. The peak trough effects are coincident with
peak concentrations of R(-) and S(?)-ibuprofen. From Suri et al. sively in some countries where it is available for OTC use
1997; redrawn and reproduced with permission of the publishers of for occasional treatment of osteoarthritis (OA) and other
Int J Clin Pharmacol Ther musculoskeletal conditions. Here ibuprofen finds particular
value in being more effective than paracetamol and having
The data obtained in these studies gives a clear basis for less GI symptoms than aspirin. Evidence in support of
relating the in vivo effects of ibuprofen on prostanoids from ibuprofen being effective in relieving pain and joint
COX-1 and COX-2 activities with the anti-inflammatory symptoms at OTC dosage in OA has come from various
and analgesic effects of the drug in various clinical models clinical trials; the earlier studies showing effectiveness in
and during therapy. Since the doses of ibuprofen used in this condition were reviewed by Kean et al. (1999).
these studies (1.2 g day-1) correspond to those used OTC Among these studies, Bradley et al. (1991) found that
and these doses produce effective analgesic activity in var- 1 month’s treatment with ibuprofen 1,200 mg day-1 (refer-
ious pain models, notably also inflammatory states such as red to as an analgesic dose) had superior rest and walking
dental pain involving extraction of molars or throat pain pain scores and comparable Health Assessment Question-
where there is local inflammation it can be concluded that naire (HAQ—Stanford University) pain and walking scores
inhibition of COX-2 as well as COX-1 (Martı́nez et al. 2002) with paracetamol (acetaminophen) 4,000 mg day-1 in
underlies the therapeutic effects of ibuprofen in these con- patients ([70% females; total number of 182 subjects) with
ditions (Boureau 1998). It could be argued that the studies of OA of the knee. Adverse events were minor symptomatic and
Blain et al. (2002) were in normal volunteers and during comparable with both drugs. The higher prescription-level
inflammatory pain there is likely to be more COX-2 activity. dose of 2,400 mg days-1 ibuprofen (referred to as an anti-
However, the in vitro inhibition profiles modelled against inflammatory dose) resulting in decreased pain and walking
plasma profiles of R(-) and S(?) ibuprofen determined scores. A later re-analysis of this study by the same group
before and after surgery suggest that these concentrations as (Bradley et al. 1992) attempted to resolve the issue of whe-
well as free concentrations of the drug would be sufficient to ther the drug effects on joint tenderness and swelling,
achieve about 70–80% inhibition. reflecting synovitis, being affected by ‘‘anti-inflammatory’’
Thus, this evidence shows that there is a direct relation- doses of ibuprofen without any indications of any joint
ship between inhibition of the synthesis of inflammatory symptoms reflecting anti-inflammatory effects. A later re-
prostanoids with the dose range of ibuprofen that corre- evaluation of the earlier study by the same group (Bradley
sponds to that used in OTC conditions for the relief of minor et al. 2001) showed that baseline pain could influence the
aches and pain; a central tenet to the therapeutic actions of anti-inflammatory/analgesic effects of ibuprofen with better
ibuprofen in the context of the OTC dosage of the drug. anti-inflammatory response being observed when there was
higher levels of baseline pain. This is relevant since there
were indications that the original study population (Bradley
Efficacy et al. 1991) comprised patients with mild-moderate joint
conditions. Thus, the effectiveness of ibuprofen is greater in
Ibuprofen has been studied extensively in adults and chil- patients with more pronounced joint symptoms.
dren in a variety of painful and inflammatory conditions The semantic definition of analgesic effects at OTC
where its therapeutic effects are comparable to other NSA- dosage of ibuprofen (1,200 mg) being separate from anti-
IDs and with that of paracetamol; in some studies ibuprofen inflammatory effects of this drug (Bradley et al. 1991) is
Ibuprofen 295

probably not justified since there are several studies in and improvements in functional status (Towheed et al.
chronic arthritic conditions showing that joint symptoms 2006).
are significantly improved with 800–1,200 mg day-1 ibu- Thus, of the available OTC treatments, ibuprofen would
profen (Kean et al. 1999). Moreover, there is an important appear to have particular advantage for self-treatment of
study performed by Deodhar et al. (1973) on the effects of joint pain and symptoms in OA in being superior in effi-
ibuprofen 1,200 mg day-1 for 1 week on joint inflamma- cacy to paracetamol and preferable to aspirin because of a
tion in RA patients in which they investigated knee lower incidence of GI symptoms (see later section on
inflammation following i.v. injection of radioactive per- adverse reactions).
technate (99mTc). The uptake of 99mTc into knee joints was
significantly reduced by ibuprofen compared with placebo. Outcomes from large-scale clinical trials with coxibs
A correlation was observed between inflammatory indices
of knee functions and 99mTc uptake. This is direct evidence The studies with the coxibs conducted during the past
for local anti-inflammatory effects of ibuprofen under decade were undertaken with large numbers of patients
conditions where there is effective relief of joint inflam- under modern standards of clinical investigation and with
matory symptoms and pain in arthritic diseases. Such relief demanding requirements to establish safety in the GI, CV
of joint inflammatory pain is not evident with high doses of and other organ systems where serious adverse events with
paracetamol. the NSAIDs often occur at low frequencies. Ibuprofen was
In another short-term study designed to measure pain used in a number of these studies as a ‘‘bench standard’’ in
relief (measured using a 100 mm Visual Analogue Scale, recognition of it being accepted as amongst the safest of all
VAS) over the 8-h period following the first and sixth dose NSAIDs that is still widely used in rheumatological and
(total 1,200 mg day-1) of ibuprofen 200 mg tablets were other musculo-skeletal conditions. These studies have
compared with placebo or ibuprofen ? codeine (30 mg) to afforded a useful and high quality source of data for
29 patients with knee OA (Quiding et al. 1992). Relatively assessment of the adverse reaction and general safety
rapid pain relief was evident with ibuprofen (as well as profile of ibuprofen from studies performed with rigorous
ibuprofen ? codeine) and this was sustained throughout the quality controls and in a setting where the drug is being
8-h period of evaluation. These results attest to the specific critically evaluated against competitors.
time-course effects of ibuprofen in relief of pain in OA. The individual adverse reactions in GI, CV and other
Schiff and Minic (2004) compared the effects of organ systems are reviewed in detail in later sections. Here,
1,200 mg day-1 ibuprofen with naproxen sodium 660 mg the adverse reaction profiles for ibuprofen and comparator
day-1 at an OTC dose (another NSAID that is available OTC drugs are viewed in a global sense employing outcome
in some countries) and placebo for 7 days in 2 multicentre measures that are considered good indicators of overall
parallel group studies in 440 patients with knee OA. They patient and physician acceptability for safety and efficacy.
found that both NSAIDs reduced joint symptoms with It is important to note that withdrawal of an NSAID from
naproxen being slightly more effective than ibuprofen. use can reflect occurrence of serous adverse events as well
However, ibuprofen was more effective in relief of day pain. as lack of efficacy.
Another large multi-centre study (known as Ibuprofen, In an evaluation of the tolerability in adverse events in
Paracetamol Study in Osteoarthritis or IPSO) by Boureau clinical trials conducted with the objective of assessing
et al. (2004) in 222 patients with OA of the hip (30% celecoxib (Celebrex, Pfizer) in osteoarthritis and rheuma-
patients) or knee (70% patients) ibuprofen 400 mg taken as toid arthritis Moore et al. (2005) used data from the
single or multiple (1,200 mg day-1) doses was compared manufacturer’s database (Pfizer) of clinical trials for com-
with paracetamol 1,000 mg single dose or 4,000 mg day-1 paring the occurrence of adverse events, clinical responses
for relief of pain and joint symptoms, WOMAC (Western and discontinuations in the treatment of arthritis from lack of
Ontario McMaster University) scores over 2 weeks. Sig- efficacy or side effects of celecoxib with those from ibu-
nificant reduction was observed in Pain Intensity Scores profen, diclofenac, naproxen, paracetamol and rofecoxib.
over the first 6-h period and then was progressively Although there are limited data available on ibuprofen the
reduced over the 2 week period with ibuprofen compared data reveals that discontinuation due to adverse events with
with that of paracetamol. This study shows that ibuprofen ibuprofen following 12 or 24 plus weeks of treatment were
is superior to paracetamol at OTC doses in relief of joint similar to those from diclofenac or celecoxib when either the
symptoms in both knee and hip OA. This conclusion is number of events or the percentage of discontinuation is
supported by a more general Cochrane Review of ran- compared (Table 8). These data should be evaluated in
domised and placebo-controlled trials in which NSAIDs relationship to the 95% confidence interval ranges which
(including ibuprofen) were superior to paracetamol in notably overlap. The lack discontinuation due to lack of
achieving reduction in pain, global efficacy assessments efficacy over 12 weeks was lowest with rofecoxib and
296 K. D. Rainsford

Table 8 Rates of discontinuations in treatment of arthritis in the celecoxib manufacturer’s trials because of lack of efficacy, or from adverse
events
Duration Treatment Dose Number of Total Discontinuations, % Number of Total Discontinuations %
(weeks) (mg day-1) events number (95% CI) events number (95% CI)

12 Pla 521 1,135 45.9 (43.0–48.8) 70 1,135 6.2 (4.8–7.6)


Ibu 2,400 145 692 21 (18.1–23.9) 52 692 7.5 (5.5–9.5)
Dic 100/150 571 6,094 9.4 (8.6–10.2) 488 6,094 8.0 (7.4–8.6)
Nap 1,000 492 6,166 8.0 (7.4–8.6) 590 6,166 9.6 (8.8–10.4)
Cel 100 128 435 29.4 (25.1–33.7) 28 435 6.4 (4.0–8.8)
Cel 200 1 132 0.8 (0.0–2.4) 13 132 9.8 (4.7–14.9)
Cel 400 374 2,399 15.6 (14.2–17.0) 316 2,399 13.2 (11.8–14.6)
Cel 800 120 4,311 2.8 (2.2–3.4) 338 4,311 7.8 (7.0–8.6)
Rof 25 14 345 4.1 (1.9–6.3) 37 345 10.7 (7.4–14)
24 Ibu 2,400 456 1,985 23.0 (21.2–24.8) 456 1,985 23.0 (21.2–24.8)
Dic 100/150 331 2,325 14.2 (12.8–15.6) 593 2,325 25.5 (23.7–27.3)
Cel 400 26 326 8.0 (5.1–10.9) 34 326 10.4 (7.1–13.7)
Cel 800 691 3,987 17.3 (16.1–18.5) 892 3,987 22.4 (21–23.8)
Pla placebo, Ibu ibuprofen, Dic diclofenac sodium, Cel celecoxib
From Moore et al. (2005)

diclofenac, followed by ibuprofen which had a low rate of that the longer plasma half-lives of drugs such as naproxen
discontinuation at prescription level of the drug, then cele- and celecoxib may contribute to these drugs having more
coxib at doses of 100–400 mg day-1. There was a high rate sustained analgesic and anti-inflammatory activity com-
of discontinuation when the drugs had been taken for 24 plus pared with that of ibuprofen thus indicating that it may be a
weeks (Table 8). These results suggest that ibuprofen has a question of the duration in circulation of these drugs that
relatively low rate of adverse event discontinuation com- accounts for any differences in their therapeutic effects.
pared with the other NSAIDs or coxibs and that this is not There is little available evidence to support these concepts
impacted by lack of efficacy. and therefore they can only be considered hypothetical.
Similar data available from a large scale randomized Another study with a coxib, lumiracoxib 400 mg
trial of the efficacy and tolerability of rofecoxib versus (Prexige, Novartis) was compared with ibuprofen
ibuprofen in patients with osteoarthritis by Day et al. 2,400 mg and naproxen 1,000 mg taken for 52 weeks in
(2000) showed that patients that received ibuprofen 18,325 patients randomized for the treatment of osteoar-
2,400 mg day-1 for 6 weeks had rates of discontinuation thritis (Farkouh et al. 2004; Schnitzer et al. 2004). There
through adverse events of approximately 12% and through were two major studies performed one addressing the CV
lack of efficacy of approximately 3% compared with those events (Farkouh et al. 2004) and the other GI safety
of rofecoxib where the discontinuation from adverse events (Schnitzer et al. 2004), with each of these having two sub-
were approximately half these values from ibuprofen but studies, one a comparison of lumiracoxib with ibuprofen
lack of efficacy was comparable. This is an important and the other comparing the former with naproxen. Only
observation since it has often been argued that the lower the data from the ibuprofen sub-study is reviewed here
rates of ADRs and toxicity of ibuprofen (including that in although it is interesting that aside from CV events ibu-
the GI tract) may be a consequence of the drug being less profen had lower or comparable safety with naproxen in
potent or that it may have differing patterns of prescribing the occurrence of other ADRs.
compared with that of other NSAIDs. The evidence is, Here, the numbers of discontinuations and the reasons
however, that the anti-inflammatory and analgesic effects for withdrawal are considered (Table 9). It is apparent from
of ibuprofen are comparable to that of other NSAIDs when this data that there were similar rates of discontinuation
given at recommended prescription levels (Kean et al. from ibuprofen compared with that from lumiracoxib; the
1999). It its true that drugs such as diclofenac and keto- same was also evident with naproxen. The losses were
profen are more potent prostaglandin synthesis inhibitors mainly due to adverse events, these being relatively low
than ibuprofen and that the selective COX-2 activities of compared with what would be expected in a study lasting
the coxibs such as celecoxib and etoricoxib may reflect 1 year. Likewise, the losses due to unsatisfactory thera-
greater efficacy of these drugs. However, it is more likely peutic effects are low and comparable with one another.
Ibuprofen 297

Table 9 Discontinuations in the TARGET study: comparisons of in adults and in children (Hersh et al. 2000b; Eccles 2006)
Ibuprofen with Lumiracoxib. Redrawn and produced with permission and is a preferred drug among the OTC analgesics for
of Lancet
treatment of tension-type headache (Verhagen et al. 2006).
Lumiracoxib vs It has wide applications in the treatment of viral respiratory
ibuprofen sub-study infections where there is an appreciable inflammatory
Lumiracoxib Ibuprofen component (Winter and Mygynd 2003). There are, how-
(n = 4,399) (n = 4,415) ever, some key points about the efficacy of ibuprofen which
Discontinued 1,751 (40%) 1,941 (44%)
need to be emphasized in the context of the OTC use of this
drug and comparisons with other analgesics, as follows:
Reason for discontinuation
Adverse events 699 (16%) 789 (18%) 1. Ibuprofen has rapid onset of analgesia and this is
Abnormal laboratory values 42 (1%) 33 (1%) maintained in parallel with the plasma elimination half
Abnormal test procedure results 11 (\1%) 13 (\1%) life of ibuprofen which for both the active (S?) and
Unsatisfactory therapeutic effect 393 (9%) 429 (10%) inactive (R-) enantiomers is approximately 2 h (Gra-
Patient’s condition no longer requires 11 (\1%) 16 (\1%) ham and Williams 2004); the analgesia extending to
study drug approximately 6 h as evidenced from a number of
Protocol violation 187 (4%) 192 (4%) analgesia models (e.g. third molar dental extraction
Patient withdraw consent 363 (8%) 419 (9%) pain model; Dionne and Cooper 1999).
Administrative problems 21 (\1%) 22 (1%) As mentioned previously in acute pain there are
Lost to follow-up 15 (\1%) 17 (\1%) alterations in the pharmacokinetics of ibuprofen
Death 9 (\1%) 11 (\1%) resulting in decreased serum levels of the enantiomers
From Schnitzer et al. (2004) after dental surgery (Jamali and Kunz-Dober 1999).
Gender differences have been observed in response to
These studies from large scale clinical trails attest to the acute pain in the dental pain model with ibuprofen
comparable rates for withdrawals from trials with ibupro- being more effective in men than in women (Walker
fen and the coxibs. They show that the newer coxibs are and Carmody 1998). Other studies using a similar third
neither more effective nor less likely to produce adverse molar extraction dental pain model have not revealed
reactions leading to withdrawals from studies compared any gender differences in response to analgesia with
with that of ibuprofen. ibuprofen (Averbuch and Katzper 2000). Aside from
these factors it appears therefore that variability in
Relief of pain, inflammation and fever response to analgesia with ibuprofen may relate to
acute pain and altering the pharmacokinetics of the
It is well established that ibuprofen at both OTC and pre- drug and possibly gender differences.
scription level dosages is effective in controlling pain and 2. Ibuprofen has been found to be effective in chronic
inflammation in a variety of inflammatory and painful arthritic pain with associated improvement in joint
conditions. Among these are rheumatic and other musculo- inflammatory symptoms even at low (800–
skeletal conditions (Boardman et al. 1967; Owen-Smith and 1,200 mg day-1) doses (Kean et al. 1999). Under
Burry 1972; Altman 1984; Dieppe et al. 1989; Haase and these conditions paracetamol is less effective. Indeed
Fischer 1991; Bliddel et al. 2000; Singer et al. 2000; several studies have shown that paracetamol has little
Buchanan and Kean 2002), dental pain and surgery, or no effects in controlling chronic inflammatory pain
dysmenorrhoea, upper respiratory tract conditions (colds, in rheumatic diseases at the recommended OTC
influenza), headaches, accidental sports injuries and surgi- dosages of 1,000 mg day-1 (Kean et al. 1999).
cal conditions (Nørholt et al. 1998; Kean et al. 1999; Dionne 3. There is unequivocal evidence for a dose–response
and Cooper 1999a, b; Rainsford 1999c; Hersh et al. 2000a, effect with ibuprofen in acute pain conditions
b; Steen Law et al. 2000; Doyle et al. 2002; Beaver 2003; (McQuay and Moore 2006; Wan Po 2006). Thus, a
Dalton and Schweinle 2006; Sachs 2005; McQuay and meta-analysis of McQuay and Moore (2006) only
Moore 2006; Eccles 2006; Verhagen et al. 2006; Huber and showed that 400 mg day-1 ibuprofen was superior to
Terezhalmi 2006). It is not the purpose here to review the 200 mg. More extensive analysis of studies by Schou
evidence in extenso about the clinical efficacy in various et al. (1998), showed that there was a much greater
painful states since this is well established from over range of dose–response that extended from 100 to
40 years of research and applications of ibuprofen in the 400 mg. Using the number needed to treat (NNT)
treatment of these conditions (Kean et al. 1999; Rainsford analysis for ibuprofen was in the range of 6–23 for the
1999c). Ibuprofen is very effective in controlling fever both high to low dose comparisons compared with that of
298 K. D. Rainsford

paracetamol 1,000–500 mg which was 6–20; the general physicians as well as pharmacists have a role in the
inference being that there is no difference in the application or prescription of ibuprofen for the treatment of
NNT between these two treatments. However, a fever. While ibuprofen has second place in the treatment of
consensus view is that in certain inflammatory pain fever over the past 3–4 decades, paracetamol has found
conditions (e.g. dental surgery) ibuprofen is superior at wide popular application and is considered to be a safe and
its recommended OTC dosage of 200–400 mg per effective treatment for most febrile conditions. However,
single treatment compared with that of paracetamol there are indications from some clinical trials that ibupro-
500–1,000 mg (Dionne and Cooper 1999; Hargreaves fen, especially in the long-term, may be more effective
and Keiser 2002; McQuay and Moore 1998). than paracetamol (van den Anker 2007). There are also
4. In comparison with other NSAIDs, including the newer indications that alternating treatment with paracetamol and
coxibs, ibuprofen has been shown in a variety of studies then ibuprofen or combinations of these two is becoming
to be at least as effective as these drugs with the increasingly popular especially amongst paediatricians and
possible exception that longer half-life drugs such as those involved in the treatment of children under emer-
rofecoxib exhibit a longer duration of action (Dionne gency or outpatient conditions (Sarrell et al. 2006). The
and Cooper 1999; Huber and Terezhalmi 2006; application of combinations or alternating treatments with
Hargreaves and Keiser 2002; Edwards et al. 2004) paracetamol and ibuprofen is highly controversial and is
and as well, ibuprofen as been shown to be effective in regarded by most as having potential risks. Indeed this
a variety of acute pain conditions (Sachs 2005). author believes that there is a major issue concerning
potential toxicity in certain organs for example in the liver
A key pharmacokinetic feature about the analgesic
and kidney. Paediatric patients with very high febrile states
actions of ibuprofen is that the drug has the ability to
that lead to cytokine activation of liver reactions, may be at
penetrate the CNS and is present in free (i.e. non-protein
risk following paracetamol administration. Monotherapy is
bound) concentrations in the CSF (Brocks and Jamali 1999;
generally preferred and ibuprofen has a key place in
Graham and Williams 2004). As well ibuprofen accumu-
treatment of fever in infants and children.
lates and is retained in inflamed joints of arthritic patients
Despite these apparent benefits the application of anti-
(Brocks and Jamali 1999; Graham and Williams 2004).
pyretic agents to treat fever in infants and children has not
Thus the drug is present in sites where analgesic and anti-
been without its critics, furthermore parents and care-pro-
inflammatory effects are required.
viders have been considered to have numerous
Recent reviews of the choice of analgesics for pain
misconceptions about what fever is and how it should be
management have highlighted that although paracetamol
treated (Crocetti et al. 2001; Stagnara et al. 2005). One
may be useful and perhaps a choice initially for pain
leading US physician, Barton-Schmitt MD, some 20 years
control that the safest and most effective of all NSAIDs is
ago found that parents had many misconceptions about what
ibuprofen at doses of 400 mg for acute non-specific pain
fever really is in terms of temperature and he coined the
(Sachs 2005) and is highly effective in tension-type head-
term ‘‘Fever phobia’’ (Crocetti et al. 2001). In a survey of
ache (Verhagen 2006). A particularly challenging opinion
340 care-providers in two urban based paediatric clinics in
provided by a recent analytical review by Arora et al.
the Baltimore region, Maryland USA, considerable varia-
(2007) suggested that oral ibuprofen is as effective in
tion was evident in potential harm that fever cause their
analgesia as parenteral ketolorac, a drug which is used in
children, or even what temperature range actually consti-
postoperative surgical pain control as well as in acute
tutes fever and in the application of sponging and other
traumatic musculo-skeletal pain conditions. Since ketol-
treatments to control fever.
orac is amongst the NSAIDs with the highest risks for
In an editorial in the British Medical Journal, Hay et al.
showing upper gastrointestinal bleeding and ulcers, it
(2006) reviewed some of the recent studies on single and
appears that substitution of oral ibuprofen in acute surgery
combination antipyretic therapies and highlighted that the
and traumatic conditions may represent a valid alternative
safety of combinations is limited. These authors noted the
with much lower risk than parenteral ketolorac.
occurrence of renal failure or renal tubular necrosis from
ibuprofen and the potential for nephrotoxic metabolite for-
Acute fever in children mation from paracetamol (quinine–imine paracetamol) in
producing both nephrotoxicity and hepatic reactions. They
General aspects concerning treatment of fever in children also pointed out that the definition of clinically useful dif-
ference in temperature after treatment is still debatable. To
Ibuprofen is widely used in the treatment of fever and in achieve better understanding continuous thermometry
the treatment of this condition care-providers can have a should be employed. Hay et al. (2009) have recently reported
considerable involvement. Likewise, paediatricians and cost benefit analysis and a trial in a UK National Health
Ibuprofen 299

System (NHS) setting on the application of ibuprofen alone juvenile rheumatoid arthritis or juvenile arthritis application
or in combination with paracetamol for treatment of fever in of ibuprofen is recommended at the dose of 10 mg kg-1 for
children. On the basis of this investigation Hay et al. (2009) the 1 month to 18-year-old group 3–4 times daily or up to 6
recommended in unwell patients with fever that ibuprofen be times daily in systemic juvenile arthritis.
used first, but that the combination may be employed where In Martindale (2002) ‘‘The Complete Drug Reference’’
longer duration of control of fever is required but the risks of ibuprofen is not recommended for children below 7 kg
toxicity should be considered. Both ibuprofen alone or the bodyweight in the same way as with the previous authors
combination were superior to paracetamol. dosage on a bodyweight basis but is recommended in the
Dlugosz et al. (2006) have reviewed the appropriate use range of 20–30 mg kg-1 day-1 in divided doses or alter-
of non-prescription antipyretics in paediatric patients. They natively in the 6–12 m age group 150 mg day-1, 1–2 year
referred to the ongoing debate about whether and when to 150–200 mg day-1, 3–7 year 300–400 mg day-1 and 8–
treat fever, but clinicians agree that antipyretic therapy is 12 year of age 600–800 mg day-1. These authorities
important for febrile children who have (a) chronic cardio- clearly differ in the recommendations in for treating fever
pulmonary disease, metabolic disorders or neurological and pain in children on a dosage basis. Arguably, however,
conditions, and (b) are at risk for febrile seizures. They dose recommendations are probably rather similar and it is
point to the lack of guidelines on the use of antipyretic a question of the application of information that is given to
agents in other categories of fever in children. Thus, patient the care giver.
comfort is cited most often as the deciding factor. More- The UK National Institute for Health and Clinical
over, there is little support for administering antipyretic Excellence (NIHCE) has prepared recommendations for the
agents when the temperature is less than 38.3C (101F) assessment and management of children younger than
unless the child is uncomfortable. None-the-less they 5 years in their report ‘‘Feverish Illness in Children’’ (2007)
regard paracetamol and ibuprofen as effective agents for (see http://www.guidance.nice.org.uk/CG47/NiceGuidance/
reducing fever and this is supported by evidence from pdf/English, accessed 16 Nov 2009). They have prepared
meta-analyses and other studies. They also point to risks of two reference guides ‘‘Understanding NICE Guidance’’ one
paracetamol hepatotoxicity especially in children with of which is a quick reference guide (N1247) and the other is
diabetes, those with concomitant viral infections, patients ‘‘Understanding NICE Guidance’’ (N1248). In the NICE
with a family history of hepatotoxic reactions, obese chil- clinical guideline No. 47 emphasis is given on the detection
dren and chronically malnourished individuals. Dlugosz of fever and the clinical assessment of the child with fever as
et al. (2006) also emphasized the precise dosing of paedi- well as the relative roles of the non-paediatric practitioner
atric patients with either ibuprofen or paracetamol (based and paediatric specialist.
on recommendations of the American Academy of Pedi- Surprisingly the NICE recommendations state that
atrics) and in the case of a patient less than 6 months with a antipyretics do not prevent febrile convulsions and should
temperature C37.9C (100.2F) recommend immediate not be used specifically for this purpose. This is in con-
consultation of a physician or paediatrician. traindication to the published evidence which show that
The application of ibuprofen and other antipyretics to antipyretic agents do have a role in the control of febrile
prevent the development of febrile seizures is now a well- convulsions. Their recommendations also give a consid-
established treatment for this condition (van Stuijvenberg erable number of clinical diagnostic indices for fever of
et al. 1998a, b, 1999). various origins. Some of these recommendations are
The question of precise dosage of antipyretics for treat- complex in themselves. Among them ‘‘A Traffic Light
ment of fever and pain has been addressed by a number of System’’ for identifying risks of serious illness involving
experts and professional organisations. Among these the colour coding of green for low risk, amber for intermediate
Royal College of Paediatrics and Child Health with the risk and red for high risk with appropriate diagnostic and
Neonatal and Paediatric Pharmacists Group in their mono- investigative procedures for identifying the origin of
graph ‘‘Medicines for Children’’ (2003) (see http://www. fevers.
rcpch.ac.uk/Publications/bnfc; accessed 16 Nov 2009) rec- The NICE recommendation for antipyretic interventions
ommend for pyrexia and mild to moderate pain that state that tepid sponging is not recommended for the
ibuprofen is given by the oral route and that the dosage treatment of fever. This is in contrast with recommenda-
should be in relation to body weight (5 mg kg-1) 3–4 times tions of other authorities. On the question of the application
daily when treating infants, and children from 1 month to of antipyretic agents these should be considered in children
12 years of age. Dosage by age is recommended above with fever who appear distressed or unwell. Antipyretic
1 year and for 1–2 years 50 mg, 3–7 years 100 mg, and agents should not routinely be used with the sole aim for
8–12 years 200 mg of ibuprofen. In 12–18 year age group reducing temperature in children with fever who are
200–600 mg ibuprofen is recommended 3–4 times daily. In otherwise well. The views and wishes of parents and care
300 K. D. Rainsford

givers should be taken into consideration; this would in any upper respiratory tract infection or systemic viral infections.
physician’s eyes be regarded as a statement of the obvious! There was a difference in the initial temperatures of patients
NICE recommendations are that either paracetamol or that were treated for upper respiratory tract infection in that
ibuprofen can be used to reduce temperature in children the mean initial temperature was 39.9C in the ibuprofen
with fever but that they should not be given at the same group and 40.81C in the paracetamol group. Despite this
time or alternatively. The only case for alternative drug difference both ibuprofen and paracetamol produced sta-
treatment would be considered for a child that does not tistically significant reduction in rectal temperatures
respond to the first agent. following administration of 7 mg kg-1 of bodyweight of
The guideline development group (GDG) has made a ibuprofen or 8 mg kg-1 bodyweight of paracetamol; the
number of recommendations for research based on the application of these been given in a random order. Initial
review of evidence to improve NICE guidance for patient reduction in temperatures of patients with upper respiratory
care in the future for example predictive values of heart tract infections incurred at 0.5–1 h with the maximum at 4 h
rate, remote assessment and a number of issues concerning after administration of both drugs. The level of antipyretic
diagnosis. They also recommend investigation of the activity was evident up to 8 h with patients having tem-
application of antipyretics in primary and secondary set- peratures in the range of 37.5C. In the group of children
tings in relationship to the degree of illness. with fever due to viral infections both the mean tempera-
tures were comparable (40.51–40.75) and similar results
Evidence for antipyretic efficacy of ibuprofen were observed as in the patients with upper respiratory tract
and comparisons with other antipyretic agents infections. The exception is that by 8 h the temperatures had
risen to 38.34–38.77C which is somewhat higher than that
The original developer of ibuprofen (Boots Ltd., Notting- observed in the patients with upper respiratory tract infec-
ham) undertook a number of clinical trials in children with tions and probably reflects ongoing viral activities.
various febrile conditions in various countries during the A single blind, parallel group investigation comparing the
1980s which involved investigating the effects of ibuprofen antipyretic properties of ibuprofen syrup versus aspirin
in comparison with aspirin or paracetamol for the treatment syrup in 78 febrile children aged 6 months to 10 years was
of fever in children. These studies were done under con- undertaken in two centres in Belgium by Heremans and co-
ditions that were regarded as being quite reasonable for the workers (1988). Using doses of ibuprofen syrup (6 mg kg-1
time but would now probably not be to the high standards body weight) or aspirin 10 mg kg-1 bodyweight, significant
of GCP today. None-the-less these studies have given an reduction in rectal temperature were observed with both
important basis for showing the effectiveness of ibuprofen treatments; there being no statistically significance between
as well as giving some information on safety. It is not the two. These patients had a greater variety of clinical
proposed to review these early studies at length although history although most were being treated for upper respira-
some key points will be considered in this section. More tory tract infections in some cases with antibiotics being co-
detailed consideration will be provided to more recent administered. Significant reductions in temperatures were
published information which is giving some insight into the observed by 0.5–1 h with both treatments and maximum
relative efficacy of ibuprofen in various febrile conditions reduction in rectal temperature being observed with both
as well as its safety. drugs at 4 h and being maintained 6 h after administration.
Among the early studies was that by Kandoth et al. (1983) A summary of more recent data from various studies
of the KEM Hospital, Bombay, India. This study was an reviewed by Autret-Leca (2003) is shown in Table 10.
open-label comparative investigation of ibuprofen suspen- These studies have been performed with modern method-
sion with that of aspirin in a cross-over design in 28 children ologies and in some cases under GCP conditions. Dose-
suffering from fever due to upper respiratory tract infections ranging studies (Table 11) show that ibuprofen is effective
or other causes and whose rectal temperature was about in the recommended dosages that are over a wide range.
38C and having been admitted to the hospital. In this small The results show that ibuprofen is equal to or in some cases
study, rectal temperatures of those patients given a daily slightly more effective than paracetamol in relief of febrile
dose of ibuprofen 20 mg kg-1 or aspirin 45 mg kg-1 symptoms in a variety of age groups in children.
bodyweight were found to be approximately equivalent with
the reduction in mean body temperature being observed at Pain relief in children
1 h with the maximum fall at 3 h after administration. There
did not appear to be any record of adverse reactions. Acute painful or inflammatory conditions
Another study by Amdekar and Desai (1985), the anti-
pyretic activity of ibuprofen was compared with that of Ibuprofen has been successfully employed in a variety of
paracetamol in 25 children suffering from fever due to acute pain models in children (Autret-Leca 2003; Perrott
Ibuprofen 301

Table 10 Comparative studies of ibuprofen and paracetamol in the treatment of acute fever
Dose Age No of Dose of ibuprofen Dose of paracetamol Outcome
frequency (years) children (mg kg-1) (mg kg-1)

Sidler et al. (1990) Multiple 1.25–13 90 7 or 10 10 Ibu 7 [ Para


Ibu 10 [ Para
Wilson et al. (1991) Single 0.25–12 178 5 or 10 12.5 Ibu 10 [ Para
Autret et al. (1994) Multiple 3 days 0.5–5 154 7.5 10 Ibu = Para
Van Esch et al. (1995) Multiple 3 days 0.25–4 70 7.5 10 Ibu [ Para
Vauzelle-Kerroëdan et al. (1997) Single 4 ± 0.6 116 10 10 Ibu = Para
Autret et al. (1997) Single 0.5–2 351 7.5 10 Ibu [ Para
From Autret-Leca (2003)

Table 11 Dose-ranging studies


Author (year) No of children Age range (years) Dose (mg kg-1) Efficacy (dose)
of ibuprofen for the treatment of
acute fever Kotob et al. (1985) 44 2–12 5, 7.5 7.5 [ 5
Walson et al. (1989) 127 2–11 5, 10 10 [ 5
Sidler et al. (1990) 90 0.4–13 7, 10 10 [ 7
Wilson et al. (1991) 178 0.25–12 5, 10 10 [ 5
Marriott et al. (1991) 93 Mean 2.5 0.625, 1.25, 2.5, 5 –
Kauffman and Nelson (1992) 37 2–12 7.5, 10 7.5 = 10
Nahata et al. (1992) 56 0.4–12 5, 10 10 [ 5
Walson et al. (1992) 64 0.5–11 2.5, 5, 10 –
From Autret-Leca (2003)

Table 12 Comparative studies


Indication No of Age Dose (mg kg-1) Results
with ibuprofen in the treatment
patients (years)
of acute pain
Dental pain 45 5–12 Ibuprofen 200 mg (6.5 mg kg-1) Ibu = Para
Paracetamol 300 mg (10 mg kg-1)
Tonsillopharyngitis 231 6–12 Ibuprofen 10 Ibu = Para
Paracetamol 10
Migraine 88 4–16 Ibuprofen 10 Ibu [ Para
Paracetamol 15
Juvenile idiopathic arthritis 86 2–15 Ibuprofen 30–40 day-1 Ibu = Asp
Aspirin 60–80 day-1
From Autret-Leca (2003)

et al. 2004; Table 12). These include pain from post-dental migraine. They concluded that of the treatments available
extraction (Moore et al. 1985), tonsillo-pharyngitis (Bertin in [6 year olds, ibuprofen 7.5 mg kg-1 (4–16 year or 6–
et al. 1991), otitis media (Bertin et al. 1996), migraine 12 year, respectively) is effective and paracetamol
(Hämäläinen et al. 1997; Annequin and Tourniaire 2005; 15 mg kg-1 is probably effective for the treatment of acute
Géraud et al. 2004; Cuvellier et al. 2005), orthodontic migraine with both being recommended. The evidence for
separator placement or orthodontic pain (Bernhardt et al. ibuprofen being most effective being from two double-
2001; Bird et al. 2007; Bradley et al. 2007) in which ibu- blind, placebo, Class 1 trials, and was considered a Level A
profen has equivalent or greater efficacy than paracetamol treatment above that of paracetamol (Level B). Triptans are
(Table 12). However, a meta-analysis by Perrott et al. also recommended (Class A) especially in their role as
(2004) concluded that ibuprofen was superior as an anti- preventative therapies. Ibuprofen for migraine is also rec-
pyretic compared with paracetamol. ommended in children B6 months by the ‘‘French
Lewis et al. (2004) in a report of the American Academy Recommendations for Clinical Practice; Diagnosis and
of Neurology Quality Standards Sub-Committee and the Therapy of Migraine’’ (Géraud et al. 2004). A systematic
Practice Committee of the Child Neurology Society review of medication trials in children by Damen et al.
reviewed the pharmacological treatment of childhood (2005) considered the evidence showed that ibuprofen and
302 K. D. Rainsford

paracetamol with or without triptans was effective in 30 mg kg-1 day-1 and 47 aspirin 200 mg tablets or 300 mg
treating childhood migraine. caplets according to body weight (60 mg kg-1 day-1) for
Ibuprofen has also been found to have beneficial effects in 12 weeks. This was followed by an open-label study in 84
children with musculo-skeletal conditions and post-opera- patients (aged 1–15 years, mean 5.3 years; av. body weight
tive pain (Kokki et al. 1994), transient synovitis of the hip 19.9 kg [range 10.0–58.0 kg]). Ten patients failed to com-
(Kermond et al. 2002), ankle sprains (Dalton and Schweinle plete the double-blind study, nine of whom had received
2006) and musculo-skeletal trauma (Clark et al. 2007). aspirin and ibuprofen; while a further six patients were
excluded from the aspirin group due to variation in diagnosis
Juvenile idiopathic (chronic) or juvenile rheumatoid or disease condition. All the patients on ibuprofen showed
arthritis reduction in all five joint parameters, while those that
received aspirin showed significantly and clinically fewer
This condition presents with a varying spectrum of clinical reductions in joint inflammation and pain on motion
manifestations that include differing joint involvement: although the reduction in morning stiffness was the same in
including pauciarticular, B4 joints; polyarticular B5 joints both groups.
with juvenile rheumatoid arthritis (JRA, Still’s Disease) as In the open-label study, 3 dropped out and 16 out of 84
a subgroup resembling the adult disease (Dieppe et al. failed to complete the 24 weeks of the study. Time-
1985; Klippel 1997). Pyrexia is common (50%) while dependent improvement in overall efficacy scores was
lymphadenopathy, splenomegaly, pericarditis and rashes observed in all 65 patients that completed the study who
may occur (Dieppe et al. 1985). High doses of the NSAIDs, received 30–50 mg kg-1 day-1 ibuprofen. One or more
especially aspirin and other salicylates, have been widely ADR’s were observed in 55% patients that were classified
used in the treatment of juvenile arthritis and more recently as possibly, probably or definitely related to the drug.
the coxibs (Ansell 1983; Hollingworth 1993; Klippel 1997; Upper GI disturbances were recorded in 31 and 27% in the
Eustace and Hare 2007). lower tract, with dose-related effects on the former group.
Amongst the earlier studies on the effects of ibuprofen Of these three patients had GI bleeding which resolved
in JRA was that by Ansell (1973). She undertook an open- after discontinuing the drug. Increased serum alkaline
label investigation in eight patients (aged 7–14 years; 5 phosphatase and bilirubin occurred in two patients that had
female, 3 male) most of them were treated because they 40 mg kg-1 day-1 ibuprofen.
were unable to tolerate aspirin and had a prior history of Steans et al. (1990) examined the safety, efficacy and
dyspepsia (5) or GI bleeding (2) or in one case where there acceptability of 10 (initially)–40 (maximum) mg kg day-1
was poor control. These patients received various doses of ibuprofen syrup in 46 children with JRA (aged 18 months–
ibuprofen (13–32 mg kg-1). Initially they received 200– 13 years; mean 6.8 years) in a multicentre, open-label study
300 mg day-1 in those with body weight of 20–30 kg and that extended on average for 8 months (range 8 weeks–2?
400 mg for those [30 kg. Later all but one received years). Six patients failed to complete the study, two of
600 mg day-1 and one 1,200 mg day-1 for what appears which had suspected side-effects. Assessments of active
to have been long periods of time (12–24 months). Satis- joints and disease activity at monthly intervals over the first
factory control of pain and stiffness were observed in six of 3 months showed that statistically significant reduction in
eight cases, although in two of these the dose had to be numbers of swollen and/or tender joint at B2 months of
increased before this was achieved. Occult blood which therapy which progressed to 6 months, while the physician’s
had been observed in those patients who were on aspirin VAS was reduced by 1 month and showed significant
became negative with ibuprofen. In six patients, liver improvement thereafter which was sustained at 4–6 months.
function tests were performed and none showed increased Side effects included gastritis (1 patient), abdominal pain (1
SGOT, SGPT or alkaline phosphatise and some showed patient), and taste disturbance and nausea (1 patient). Of the
decrease in these values. This is important since plasma/ 39 children that completed the trial, 28 showed improve-
serum levels of elevated liver enzymes have been fre- ment on therapy, 7 were worse and 4 remained unchanged.
quently observed in patients with JRA that have received
aspirin and ADR’s in the GI tract and other systems are Effects of suppositories
frequently observed with the salicylates and other NSAIDs
in these conditions (Hollingworth 1993; Kean et al. 2004). A study performed by Stagnara et al. (2005) in Lyon,
Giannini et al. (1990) undertook two studies—one being a France was an open-label, multi-centre, non-comparative
multi-centre, randomized, double-blind study in 92 children study designed to evaluate the antipyretic effects and
(76 girls, 16 boys) mean age 7.7 years [range 1.8– acceptability of 60 or 125 mg ibuprofen suppositories over
15.1 years], mean body weight 26.4 (range 11.5–58.7) kg a 5 day period. The study was performed (in a planned
with JRA. Of these 45 received ibuprofen suspension number of 80 patients but involved 45 patients who were
Ibuprofen 303

analyzed) aged 3 months to 6 years. Assessments were Assessments in this section with supporting data and
undertaken by both parents and practitioners and involved information show (a) the overall safety profile of ibuprofen
categorical scale determinations of acceptability and tem- at the current recommended prescription dosage 1,800–
perature measurements. Temperatures declined within 1/ 2,400 mg day-1 for the short and long-term treatment of
2 h after administration of the suppositories and were acute and chronic moderate to severe inflammatory pain
lowered for up to 8 h afterwards. Similar reductions in conditions including rheumatoid- and osteoarthritis,
temperatures were evident in 2 year olds compared with spondylo-arthropathies and other rheumatic conditions; (b)
6 year olds showing there was no age dependence in the safety of ibuprofen B1,200 mg day-1 for a maximum
antipyretic effects of the drug. The efficacy of drug treat- dosage period of 7–14 days which in a number of countries
ment was found to be favourable with 88.4% of parents and worldwide is sold as a non-prescription over the counter sale
86% of physicians reporting that the treatment was ‘‘Effi- (OTC) analgesic for the relief of mild to moderate painful
cacious’’ or ‘‘Very Efficacious’’. The treatment was judged conditions many of which have a moderate acute inflam-
to be ‘‘Well Adapted’’ or ‘‘Perfectly Adapted’’ in the matory component; (c) the efficacy and therapeutic activities
clinical situation by 86% of the investigators. Eight of ibuprofen principally at OTC dosage; (d) assessment of
patients had adverse effects which were non-serious. Rectal the risks/benefits of ibuprofen compared with other anal-
examinations by the investigators showed that there were gesics (paracetamol) and OTC NSAIDs (ketoprofen,
no local reactions but parents reported abnormalities in naproxen) that are sold as non-prescription (OTC) analge-
four patients that were minor in which redness was sics in some countries; and (e) a review of the
observed but this did not lead to stoppage of treatment. recommendations and guidelines for the use and safety
issues derived from the countries where ibuprofen is a drug
sold in considerable quantities for OTC use or General Sales
General safety profile List (GSL) (e.g. USA, UK, Canada, Australia and other
Commonwealth countries) with EMEA recommendations
There were indications of a favourable safety profile for and requirements and guidelines on the safety of NSAIDs
ibuprofen from the post-marketing data during the 15 year and analgesics. The conclusion from this assessment of the
period after approval in the USA (Royer et al. 1984). This safety and benefits of ibuprofen can be summarized thus:
information was important in decisions made by the FDA
• Ibuprofen at OTC doses has a low possibilities of
in granting approval for OTC use in the USA (Rainsford
causing serious GI events, and little prospect of
1999c).
developing renal and associated CV events.
Since then the safety profile of ibuprofen has been
• Ibuprofen OTC does not pose a risk for developing
compared with a range of other new and established
liver injury especially the irreversible liver damage
NSAIDs on the basis of being a recognized ‘‘bench mark’’
observed with paracetamol and the occasional liver
for safety and efficacy comparisons (Kean et al. 1999). In
reactions from aspirin.
particular, ibuprofen has been used as a comparator drug in
• The pharmacokinetic properties of ibuprofen especially
several trials with the newer coxib class of NSAIDs; an
short plasma half-life of elimination, lack of develop-
aspect that will be considered later in this section and
ment of pathologically-related metabolites (e.g.
subsequent sections on the adverse events and toxicity in
covalent modification of liver proteins by the quinine-
individual organ systems. Data from epidemiological
imine metabolite of paracetamol or irreversible acety-
sources and from a large number of controlled clinical
lation of biomolecules by aspirin) support the view that
trials performed in comparison with the newer coxib class
these pharmacokinetic and notably metabolic effects of
of NSAIDs has shown that ibuprofen is amongst the
ibuprofen favour its low toxic potential.
tNSAIDs with the lowest risks of serious GI events (i.e.
• Moderate inhibition of COX-1 and COX-2 combined
peptic ulcer bleeds, PUBs etc.) (Rainsford et al. 2008). In
with low retention as shown by the mean residence time
comparison with coxibs ibuprofen has shown slightly
of the drug in the body may account for the low GI, CV
higher risks when given short term (1–3 months) yet after
and renal risks from ibuprofen, especially at OTC doses.
long-term (6? months) ibuprofen has comparable and low
risks of serious GI events. Symptomatic GI events (dys- There are clearly low risks from OTC ibuprofen based
pepsia, nausea, heartburn, diarrhoea, constipation, on the safety profile of the drug and the experience in
vomiting) from ibuprofen are, in general, comparable with regulatory domains worldwide. The benefits to the public
those from the coxibs. Thus, ibuprofen can be considered are also clear in providing superior pain-relief in a variety
to have consistently shown relatively low risks of serious of acute and chronic inflammatory pain conditions with
GI events in the recent extensive studies in large scale lower risks of toxicity compared with that of the other two
controlled clinical trials. most commonly used analgesics, aspirin and paracetamol.
304 K. D. Rainsford

ADRs and safety in prescription level doses in adults epidemiological investigations (e.g. sufficient numbers of
study subjects or validity of databases).
The overall pattern of adverse events from ibuprofen at the Among the more reliable of these was a study by Freis
prescription dosage-level has been principally derived from et al. (1991) who performed an investigation of the relative
studies in adult populations principally those with rheu- toxicity of a range of NSAIDs used in the treatment of
matic and other acute and chronic painful conditions rheumatoid arthritis in the USA with data recorded in the
(Rainsford 1999c). These probably conform to that of all Arthritis, Rheumatism, and Aging Medical Information
NSAIDs (Rainsford 2001). The diagrammatic representa- System (ARAMIS). It should be noted that this study was
tion shown in Fig. 6 indicates the spectrum of adverse undertaken in what can be described as the ‘‘pre-coxib’’
reactions that is in general observed with the NSAIDs. era, i.e. before the introduction of coxibs during late 1999.
This has given rise to the concept that there are This has significance since the coxibs had an appreciable if
NSAID adverse events that can be considered to be variable share of the NSAID market world-wide and thus
class-related (i.e. common to all NSAIDs). Within this influenced the overall patterns of use of the NSAIDs in
concept it is clear that NSAIDs vary considerably in their rheumatic and other musculo-skeletal conditions. A sum-
frequency or occurrence of individual side effects. Some mary of the Standardized Toxicity Index from the 11 most-
of these are mechanism-related that is to say related to frequently prescribed NSAIDs including ibuprofen in RA
the effects on prostaglandin production via COX-1 inhi- patients adjusted for weightings, demographic factors etc.,
bition for example in the GI tract and kidneys. However, as part of a sensitivity analyses, is shown in Table 13.
it has been argued that in relationship to some of these An epidemiological safety investigation known as the
effects that there are important interactions between Safety Profile of Antirheumatics in Long-term Administra-
inhibition of COX-1 and COX-2, nitric oxide synthase tion (SPALA) was undertaken during the late 1980s to 1990
and physico-chemical factors which are of significance in involving 30,000 rheumatic patients in participating centres
the development of these effects (Bjarnason et al. 2007) in West Germany (N = 9), Switzerland (N = 3) and Austria
so that they all can not be considered to be mechanism- (N = 4) (Brune et al. 1992). Of the 10 most-frequently
related. prescribed NSAIDs (N = 36,147 prescriptions) ibuprofen
was the second most-frequently prescribed drug after dic-
Epidemiological studies lofenac, ranking it fourth in the overall total number of
ADRs among the ten drugs. As shown in Table 14 ibuprofen
A number of studies have been performed examining the was associated with the least number of reactions in the GI,
relative safety and adverse events attributed to ibuprofen liver and biliary, and ‘‘body as a whole systems’’.
compared with other NSAIDs. Many of these studies have These two studies show that ibuprofen at prescription-
involved examination of the occurrence of adverse events level doses given to rheumatic patients is amongst the
e.g. in specific system organ classes (SOC) or individual lowest toxicity ratings of frequently prescribed NSAIDs.
reactions in organ systems (e.g. gastro-intestinal ulcers and
bleeding). These studies are reviewed in subsequent sec- Outcomes from large-scale clinical trials
tions of this review. There are relatively few studies where
overall ‘‘toxicity’’ of NSAIDs has been determined under The studies with the coxibs conducted during the past
conditions for meeting adequate standards for such decade were undertaken with large numbers of patients

Fig. 6 Patterns of adverse Adverse Reactions from NSAIDS


reactions from the NSAIDs
• Stomach/intestinal ulcers & bleeding
(Bjarnason et al. 2005). The (GI-symptoms (nausea, gastric pain diarrhoeaetc.) Most prevalent
occurrence and severity of these Skin rashes, eruptions, itching, reddening
reactions differs with each drug. • Abnormal kidney & liver functions
Reproduced with permission of Confusion, visual & abnormalities
Birkhäuser Verlag • Asthma & Respiratory reactions
• Immune reactions/hypersensitivity to „self“
molecule
• Joint destruction (in osteoarthritis)
• Kidney failure
• Liver injury and failure
Bleeding & small vessel damage
• Severe (bullous) skin reactions
Least prevalent-rare
Severe reactions shown in bold and underlined; others of low severity but higher frequency
Cardiovascular risks unclear at present
Ibuprofen 305

Table 13 Toxicity indices of 11 most-frequently prescribed NSAIDs, including ibuprofen in patients with rheumatoid arthritis in data derived
from five centres in USA and Canada analyzed from the ARAMIS database (Freis et al. 1991)
Drug All patients Drug starts only
No. of courses Standardized toxicity index No. of courses Standardized toxicity index
score, ±SEM (rank) score, ±SEM (rank)

Aspirin 1,669 1.19 ± 0.10 (1) 410 1.37 ± 0.35 (1)


Salsalate 121 1.28 ± 0.34 (2) 107 1.30 ± 0.30 (2)
Ibuprofen 503 1.94 ± 0.43 (3) 238 2.34 ± 0.55 (3)
Naproxen 939 2.17 ± 0.23 (4) 327 3.43 ± 0.58 (4)
Sulindac 511 2.24 ± 0.39 (5) 220 2.89 ± 0.45 (5)
Piroxicam 790 2.52 ± 0.23 (6) 291 3.33 ± 0.46 (6)
Fenoprofen 161 2.95 ± 0.77 (7) 71 3.09 ± 0.65 (7)
Ketoprofen 190 3.45 ± 1.07 (8) 147 3.44 ± 0.78 (8)
Meclofenamate 157 3.86 ± 0.66 (9) 84 4.43 ± 0.84 (9)
Tolmetin 215 3.96 ± 0.74 (10) 120 4.83 ± 0.78 (10)
Indomethacin 386 3.99 ± 058 (11) 159 4.32 ± 0.60 (11)
These data comprise toxicities from all patients in the database and those that are considered ‘‘drug starts’’. The results obtained with these
differing periods of drug exposure were essentially similar. Ibuprofen was in a group with the two other salicylates, aspirin and salsalate) having
the lowest toxicity ratings

Table 14 Frequencies of
Frequencies of adverse events (no. AEs/no. prescriptions) and percentage individual AEs
adverse events (AE) in relation
to numbers (no.) of four most- Organ system classes Diclofenac Ibuprofen Indomethacin Acemetacin
frequently prescribed NSAIDs
in the SPALA study in No. of prescriptions 14,477 4,037 3,896 3,633
rheumatic patients in West Gastrointestinal system (%) 14.10 11.20 15.90 19.10
Germany, Austria and
Switzerland Skin and appendages (%) 3.50 3.30 3.50 4.50
Central and peripheral NS (%) 2.50 3.00 7.90 4.80
Liver and Biliary system (%) 2.20 0.70 1.80 1.50
Body as a whole—general (%) 2.70 2.20 3.10 4.10
From Brune et al. (1992)

under modern standards of clinical investigation and with As previously mentioned the tolerability in adverse
demanding requirements to establish safety in the GI, events in clinical trials conducted with the objective of
CV and other organ systems where serious adverse assessing celecoxib in osteoarthritis and rheumatoid
events with the NSAIDs often occur at low frequencies. arthritis Moore et al. (2005) in clinical trials for comparing
Ibuprofen was used in a number of these studies as a the occurrence of responses and adverse reactions from
‘‘bench standard’’ in recognition of it being accepted as celecoxib with that of ibuprofen, diclofenac, naproxen,
amongst the safest of all NSAIDs that is still widely paracetamol and rofecoxib (Table 8). The data showed that
used in rheumatologic and other musculo-skeletal con- adverse event discontinuation with ibuprofen following 12
ditions. These studies have afforded a useful and high or 24? weeks of treatment were similar to those from
quality source of data for assessment of the adverse diclofenac or celecoxib when either the number of events
reaction and general safety profile of ibuprofen from or the percentage of discontinuation is compared (Table 8).
studies performed with rigorous quality controls and in a In the TARGET study, lumiracoxib 400 mg was compared
setting where the drug is being critically evaluated with ibuprofen 2,400 mg and naproxen 1,000 mg taken for
against competitors. 52 weeks (Farkouh et al. 2004; Schnitzer et al. 2004).
The individual adverse reactions in GI, CV and other There were two major studies performed one addressing
organ systems are reviewed in detail in later sections. Here, the CV events (Farkouh et al. 2004) and the data from
the overall adverse reaction profiles for ibuprofen and ibuprofen sub-study is reviewed here in which this drug
comparator drugs are viewed in global sense employing had lower or comparable safety with naproxen.
outcome measures that are considered good indicators of These studies from large scale clinical trails attest to the
overall patient and physician acceptability for safety and comparable rates for withdrawals from trials with ibupro-
efficacy. fen and the coxibs. They show that the newer coxibs are
306 K. D. Rainsford

neither more effective or less likely to produce adverse AEs in different patient groups, although the number of
reactions leading to withdrawals from studies compared patients in each of the groups was probably not sufficient to
with that of ibuprofen. meet statistical requirements for being assessable. As par-
acetamol may be considered a ‘benchmark’ drug with a
Adverse events at non-prescription (OTC) dosages low propensity to cause serious GI events this study sug-
gests as there was no differences in GI AEs between
A considerable number of studies have been reported com- ibuprofen and paracetamol at OTC dosages, ibuprofen can
paring the adverse reactions from non-prescription (OTC) be considered to have low risk of GI reactions comparable
doses of ibuprofen with placebo, paracetamol (acetamino- with paracetamol.
phen) or other analgesics. These studies have been performed Kellstein et al. (1999) performed a meta-analysis of
using a variety of methodologies and study designs, some of reports of randomized, double-blind, placebo-controlled
which may have been critical to the outcomes. parallel-group studies having initially reviewed published
Earlier reviews of published literature showed that OTC literature and established that only eight studies, all of
ibuprofen has comparable reports of adverse events (AEs) which were unpublished but claimed as independent stud-
with paracetamol or placebo (Furey et al. 1993; DeArmond ies performed under the auspices of Whitehall–Robbins
et al. 1995; Moore et al. 1996). Healthcare met the criteria as specified above according to
A systematic analytical review of published studies GCP conditions (Table 17). AEs were codified according
compared OTC ibuprofen with paracetamol where the to the conventional Coding Symbol Thesaurus for Adverse
drugs were taken as single doses or daily dosages up to Reaction Terms (COSTART) with the exception of
10 days (Rainsford et al. 1997; Tables 15, 16). abdominal pain, which was ‘‘conservatively’’ assigned to
The subjects in these studies were either healthy vol- the ‘‘body as a whole’’ digestive system. This may in fact
unteers, or those who had experienced various types of have disguised the importance of this AE since it is a
acute pain or chronic inflammatory conditions. Some relatively frequent event in trials with NSAIDs and
studies involved comparisons with other analgesics/NSA- paracetamol.
IDs or placebo. Thus, there were a wide range of conditions The eight selected studies were in mixed patient groups
in which the treatments were compared. The results comprising three in OA pain, two in Delayed Onset Muscle
showed that there were no significant differences between Soreness (DOMS), and one each in sore throat pain, dental
ibuprofen and paracetamol in occurrence of AEs after pain and a study of Maximal Use Safety and Tolerability
single or multiple daily doses taken for up to 10 days (MUST) of non-prescription ibuprofen. The study dosages
(Table 15) although there was a trend to increased GI AEs ranged from 400 mg b.i.d. (800 mg day-1, 2 studies) and
in both groups with increased duration of drug intake. 400 mg t.i.d. (1,200 mg day-1, 6 studies) with the duration
There did not appear to be any discernable differences in of intake being 1–10 days. The primary purpose was to

Table 15 Overall adverse event rates and exposure grouped by duration of dosing
Days dosed Drug No. of Exposurea Total number Overall percent Total number with Total number of
groups of patients with adverse events adverse eventsb adverse eventsc

\1 Paracetamol 27 0 4,644 10 444 479


\1 Ibuprofen 25 0 2,312 6 148 172
1 Paracetamol 11 420 420 10 43 49
1 Ibuprofen 5 215 215 8 18 22
2–7 Paracetamol 15 2,882 687 8 57 64
2–7 Ibuprofen 9 1,015 227 9 20 29
8–30 Paracetamol 6 5,496 207 19 39 39
8–30 Ibuprofen 9 5,960 272 19 52 52
31–90 Ibuprofen 5 6,504 85 29 25 29
Total Paracetamol 59 8,798 5,958 10 583 631
Total Ibuprofen 53 13,694 3,111 8 263 304
a
Number of patient days
b
Adverse events grouped as the total number of patients having these events
c
Adverse events grouped as the total of all recorded adverse events
From Rainsford et al. (1997)
Ibuprofen 307

Table 16 Statistical comparison of adverse events from data in (d) Overall tests for homogeneity among the study groups
Table 17 of studies where ibuprofen and paracetamol were compared using the Breslow–Day statistical test showed no
directly (n C 40, treatment C 7 days)
significant differences between the occurrences of all
Adverse events F P Significance individual AEs over all the study groups.
(e) Serious AEs over all categories were fewer in the
Gastrointestinal
ibuprofen compared with placebo in both the single
Constipation 1.000 0.341 N/S
and multiple dosage groups. Urinary tract infections
Discomfort 1.225 0.297 N/S
while rare were more frequent in ibuprofen than
Vomiting 1.227 0.297 N/S
placebo groups.
Nausea 0.006 0.941 N/S
Heartburn 0.001 0.975 N/S The reason for higher rates of AEs from placebo in ‘‘all
General 1.060 0.328 N/S body systems’’ and ‘‘body-as-a-whole’’ compared with
Gastrointestinal total 0.639 0.443 N/S ibuprofen is attributed to a larger number of patients in the
CNS total 1.013 0.338 N/S placebo group reporting headaches, neck pain and malaise.
Other total 1.138 0.311 N/S The lower rates of these reactions in the ibuprofen groups
Total 0.733 0.414 N/S are consistent with its analgesic activity.
While the studies employed in this meta-analysis are
N/S, Not significant; analysis of variance single factor, level of sig-
nificance set at 0.05
from unpublished investigations that have not been sub-
From Rainsford et al. (1997)
jected to peer-review they are none-the-less from
investigations that were performed according to GCP
requirements and would have been in the company data-
base that is subject to scrutiny by the US FDA.
compare the effects of single-dose with multiple daily
Another study from the same company involved a pro-
doses of ibuprofen with placebo. The subjects covered a
spective investigation of GI tolerability of the maximum
wide range of ages (12–97 years) and racial groups of both
daily OTC dose of 1,200 mg ibuprofen compared with
genders in a total of 1,094 ibuprofen and 1,093 placebo-
placebo taken for 10 days in 1,246 healthy volunteers
treated subjects.
(Doyle et al. 1999). A total of 19% of ibuprofen-treated
Table 17 summarizes the serious AEs from this study.
subjects (67 of 413) and 16% of placebo treated (161 of 833)
The principle outcomes can be summarized thus:
individuals experienced GI AEs, there being no significant
(a) The overall number of AEs, those in body-as-a-whole differences between the two groups. The GI adverse reac-
and the digestive system were greater after multiple doses tions were dyspepsia, abdominal pain, nausea, diarrhoea,
compared with single doses of ibuprofen and placebo. flatulence and constipation. Occult blood tests were positive
(b) There were no differences in AEs in all body systems in 1.4% of all subjects; there being no differences between
and in the digestive system after single doses of the two treatments in the occurrence of these reactions. The
ibuprofen compared with placebo or in the digestive results in this prospective study confirmed the data from
system and body-as-a-whole after multiple doses. previous retrospective studies and showed that ibuprofen at
(c) In an analysis of individual AEs by COSTART, OTC dosages has comparable GI reactions to placebo.
dizziness was identified among the central nervous In a pharmaco-epidemiological investigation involving
system reactions to be significantly increased after questionnaires distributed to 40 pharmacies in the Camp-
multiple doses in the ibuprofen (2.5%) compared with ania region of Southern Italy, Motola et al. (2001) observed
placebo (1.4%); there being no differences after single that GI AEs were the most frequent with OTC and pre-
doses of the treatments. scription NSAIDs with the incidence being 5.5% overall.

Table 17 Number (N) and percentage (%) of subjects


Pool studies All body systems Digestive system Body-as-a-whole system
Placebo N (%) Ibuprofen N (%) Placebo N (%) Ibuprofen N (%) Placebo N (%) Ibuprofen N (%)

Single-day studies 7/318 (2.2) 1/319 (0.3) 2/318 (0.6) 1/319 (0.3) 5/318 (1.6) 0/319 (0.0)
Multiple-day studies 59/775 (7.6) 38/775 (4.9) 21/775 (2.7) 14/775 (1.8) 36/775 (4.6) 21/775 (2.7)
All studies 66/1,093 (6.0) 39/1,094 (3.6) 23/1,093 (2.1) 15/1,094 (1.4) 41/1,093 (3.8) 21/1,094 (1.9)
Experiencing a severe adverse reaction over all body systems, the digestive system, and the body-as-a-whole system
From Kellstein et al. (1999)
308 K. D. Rainsford

In a large scale general practice (known as the PAIN Table 18 Most frequent significant adverse events by COSTART
Study) based investigation in 4,291 patients in France, Le body systems and terms
Parc et al. (2002) compared the tolerability of 7 days treat- Systems/terms Ibuprofen (%) Aspirin (%) Paracetamol (%)
ment of ibuprofen (up to 1.2 g day-1) with aspirin (up to
Body as a whole 5.4 7.4 5.7
3 g day-1) for relief of musculoskeletal conditions. So-
called ‘‘significant’’ AEs were reported in 15.0% of patients Digestive system 3.6 4.7 4.3
who took ibuprofen, 17% on paracetamol and 20.5% on Nervous system 1.1 2.2 1.1
aspirin; the difference between the ibuprofen and paraceta- Respiratory system 1.2 1.5 1.3
mol groups being not statistically significant but significantly Abdominal pain 2.0 5.1 2.7
different from the aspirin group (Table 18). GI AEs were Nausea 1.6 1.8 1.3
fewer in the ibuprofen group (4.4%) than in the paracetamol Dyspepsia 0.9 1.9 1.3
(6.5%) or aspirin (8.6%) group, the differences in all groups Headache 1.2 1.1 1.6
being statistically significant from one another. In the non- From Moore et al. (2002)
musculoskeletal group there were similar trends although
there was no occurrence of serious digestive AEs.
Using the data acquired in the abovementioned PAIN ‘‘suspect’’ risk because of indicative symptoms requiring
Study, Moore et al. (2004) performed an assessment of risk investigation) or patients with rheumatic diseases. The focus
factors that accounted for the development or association of these studies has been to identify the risks of serious AEs
with AEs. By employing multivariant logistic regression in the GI tract from intake of OTC analgesics. The rheumatic
analysis of 8,633 evaluable patients they identified the patients may have increased susceptibility to GI events from
following risk factors (a) indication (e.g. musculo-skeletal intake of NSAIDs as a consequence of their disease, con-
pain, sore throat, colds and flu, menstrual pain, headache) current disease (e.g. diabetes, CHD), concomitant
(b) concomitant medications, (c) history of previous GI medications (either anti-rheumatic e.g. steroids, or other
disorders, and (d) female gender. Age was not a risk factor. agents to control diabetes, hypertension, CV disorders or
There were fewer clinically significant risk factors for GI subnormal renal function), as well as socio-psychologic
AEs in the ibuprofen compared with paracetamol groups. stress or Helicobacter pylori infection. Since many patients
The overall conclusion was that the main risk factor was with rheumatic disorders take OTC analgesic medications
concomitant medications. on a self-administered p.r.n. basis and as a reflection on costs
A meta-analysis undertaken by Ashraf et al. (2001) in of prescription NSAIDs which for the elderly or members of
elderly ([65 years) osteoarthritic patients in which the lower socio-economic classes could be a major problem, use
incidence of adverse events (COSTART coded) from ibu- of OTC analgesics in these patients can be regarded as one of
profen 1,200 mg daily was compared with those that the ‘‘real-world’’ uses of these drugs.
received placebo. Following an initial assessment of the Among the reports in GI suspect risk patients Blot and
quality of papers, three independent clinical trials that had McLaughlin (2000) reported investigations conducted by a
been performed by Whitehall Robins (USA) were selected mail survey of members of the American College of Gas-
in which the drug treatments were for B10 days. The troenterology (ACG) designed to identify risks of GI
pooled overall incidence of adverse events was 29.4% with bleeding associated with intake of analgesics at OTC
the ibuprofen group (N = 197 patients) and 29.0% in the dosages within the previous week of intake. The method-
placebo group (N = 210 patients), with the three studies ology involved data collected from the ACG Registry
individually showing no statistically significant differ- (N = 627 patients) and ‘‘procedure-matched’’ endoscopy
ences. The percentages of adverse events in the organ controls. Suspect factors (e.g. tobacco, alcohol intake etc.)
systems were for (a) ‘body as a whole’—12.7% on ibu- were also identified. These hospitalized patients had a
profen c.f. 9.5% on placebo; (b) digestive system—12.2% variety of upper or lower GI conditions that led to bleeding
on ibuprofen c.f. 13.3% on placebo; and (c) nervous sys- in the OTC analgesic group but with no bleeding in the
tem—10.2% on ibuprofen c.f. 8.1% on placebo; the control group. The number of patients in these groups
differences being not statistically significant. might be considered relatively small and questions can be
This study is important in showing that ibuprofen at raised about the statistical validity of the sub group analysis
OTC doses is safe in the elderly OA patient, a group who of risk factors. The cases tended to be older subjects (mean
frequently self-administer the drug. 60 years) compared with controls (55 years) with 45%
Several investigations have been performed in what could cases being over 65 years compared with 33% controls,
be regarded as ‘‘at risk’’ patients (based on prior clinical and they were more often male (63%) cases compared with
history) either those admitted to hospitals for clinical controls (49%). The balance of races was comparable with
investigation (and who could be regarded as being at about two thirds being non-hispanic whites. GI risk,
Ibuprofen 309

especially in the upper tract was greater in those that had In their practitioner-based population study of 2,015
consumed alcoholic beverages, this being increased in primary care physicians throughout continental United
smokers, but cigarette smoking was unrelated to GI risks. States of America, Lesko and Mitchell (1995) undertook a
Of the major analgesics reported intake of drugs was randomised, double-blind, office-based paracetamol (acet-
associated with GI bleeding in 9.5% aspirin-takers, 4.2% aminophen)-controlled trial of a total of 84,192 patients
ibuprofen takers, and 5.4% paracetamol users. A consider- aged 6 months to 12 years of age who were randomly
able number of patients had taken mixtures of two analgesics assigned to receive ibuprofen 5 or 10 mg kg-1 suspensions
or prescribed NSAIDs. It should be emphasized that the (Children’s Motrin, McNeill), or 12 mg kg-1 paraceta-
numbers of patients were relatively small among the single mol suspension (Calpol, Burroughs Wellcome) for the
analgesic-users (56 aspirin, 25 ibuprofen, 32 paracetamol) so treatment of acute febrile illness. The study provided for a
it is questionable to ascribe causality to individual drugs. 4 week follow-up period to determine the occurrence of
Non-the-less these data are instructive at least for assessment side-effects. The primary outcome measures were hospi-
of potential GI bleeding in at risk patients. It is interesting that talizations for acute GI bleeding, acute renal failure and
paracetamol was associated with GI bleeding as it is normally anaphylaxis. The occurrence of Reye’s syndrome was also
considered a low risk GI ‘‘safe’’ drug. In this complex group monitored. Secondary outcomes included identification of
of patients with evident underlying disease it is clear that previously unrecognized serious reactions. Two patients
ibuprofen is somewhat safer than the other two analgesics. died one in a road accident who had received paracetamol
In conclusion: (a) the studies at prescription-level doses and the other who had ibuprofen, died from bacterial
show that ibuprofen has amongst the lowest risks for meningitis; both these fatalities could be considered to be
adverse events, (b) this drug is as good in safety and effi- unrelated to the drugs. A total of 1% of the patients was
cacy as any of the newer coxibs (which were designed to admitted to hospital in the 4 weeks following enrolment.
have lower incidence of adverse reactions), with serious Four children were hospitalized for acute GI bleeding that
events being rare, and (c) at OTC doses ibuprofen has low was due to ibuprofen (2 from 10 mg kg-1 and 2 from
or at least amongst the lowest rating of risks for developing 5 mg kg-1 of the drug) giving a risk of GI bleeding as 7.2
adverse reactions compared with other analgesics. per 100,000 (95% CI 2–38 per 1000,000) with the risk from
paracetamol being zero; the difference being not statisti-
Adverse events and safety in paediatric populations cally significant. Gastritis or vomiting was observed in 20
patients that had received ibuprofen giving a risk of 36 per
The safety profile of ibuprofen has been extensively eval- 100,000 (95% CI 22–55) and in 6 patients on paracetamol,
uated in traditional paediatric clinical trials of fever and/or the risk being 21 per 100,000 (95% CI 7.9–46). There were
pain have been evaluated in a number of trials or in critical 24 patients who had received paracetamol who had asthma
reviews (Walson et al. 1989; Czaykowski et al. 1994; (RR = 85, 95% CI 55–150) and 44 on ibuprofen (RR = 80
Rainsford et al. 1997, Rainsford 1998, 1999, 2001; Diez- per 100,000; 95% CI 57–110), thus showing there was no
Domingo et al. 1998; van den Anker 2007). All have difference in risks between the two drugs. There was no
shown the low incidence of serious and non-serious risk from Reye’s syndrome, acute renal failure or ana-
adverse events (AEs) with ibuprofen. While these data are phylaxis among 55,785 children that received ibuprofen.
useful it is only in large scale population based studies that Low white blood cell count was observed in eight children
it is possible to obtain sufficient data to obtain a sound that had received ibuprofen (but the causality could not be
basis for safety evaluation. established) and none in the paracetamol group. The
In a series of papers, Lesko and Mitchell (1995; 1997; authors considered that the risks from outcomes of low
Lesko et al. 2002) have compared the safety of ibuprofen incidence could not be ascertained because of the power of
and paracetamol with a focus in particular on ibuprofen, in the study. This study attests to the low risks for serious GI,
practitioner-based clinical trials, the methodologies of renal of anaphylactic events form ibuprofen, and a lack of
which were reviewed by Mitchell and Lesko (1995). These association with severe renal or asthmatic events.
studies have been supported by both leading companies In what is probably the largest study designed to
producing the antipyretics in the USA as well as the US investigate the safety of analgesics in children B2years old,
FDA, US NIH and other pharmaceutical companies sup- Lesko and Mitchell (1999) used data from the Boston
porting the Sloane Epidemiology Unit of Boston University Collaborative Fever Study in a total of 27,065 febrile
School of Medicine (Brookline, MA, USA) where these children who were randomized to receive 5 or 10 mg kg-1
studies have been based. Among these studies the use of ibuprofen of 12 mg kg-1 paracetamol suspensions. The
Advisory Groups has been employed which helps retain the study was double-blind and practitioner-based with chil-
abilities to critically assess data and ensure proper conduct dren being eligible if, in the opinion of the attending
of trials. physician, their illnesses warranted treatment with an
310 K. D. Rainsford

antipyretic; duration and height of fever were not a criteria ADR’s in children B2 years and especially those B6 months
for participation. Follow-up was achieved by mailed which receive antipyretic therapy for febrile illness.
questionnaire or telephone interviews. The most common Another large investigation into the overall safety of
cause of fever in children was otitis media (45%), upper ibuprofen in paediatric populations was performed by
respiratory tract infection (40%), pharyngitis (15%), lower Ashraf et al. (1999). This study known as the Children’s
respiratory tract infection (7.4%) and gastro-intestinal Analgesic Medicines Project (CAMP) was a prospective,
infection (2.2%). Data from the two doses of ibuprofen multicentre, all-comers, multi-dose, open-randomized and
were combined because there were no discernible differ- open label study designed to compare the safety of ibu-
ences between the groups; thus the size of the ibuprofen profen (Children’s Advil) with that of paracetamol
group is about twice that of the paracetamol group. (Children’s Tylenol) given for relief of pain and/or fever.
The risk of hospitalization for any reason in the 4-weeks A total of 41,810 children aged 1–18 years were enrolled in
after enrolment (N = 385 patients in total) was the same in a so-called naturalistic outpatient paediatric setting
the ibuprofen group [Relative Risk, RR = 1.1 (0.9–1.3) (PEGASUS Research Inc., Salt Lake City, UT, USA)
compared with that of the paracetamol group as a reference involving 68 clinics in the USA. Among 30,144 children who
RR = 1.0]. The absolute risks were 1.5% (1.3–1.6, 95% CI) took one dose of either ibuprofen or paracetamol, 14,281
for the ibuprofen group and 1.4% (1.1–1.6%) for the para- were ‘‘younger’’ aged B2 years and 15,863 were ‘‘older’’
cetamol group. None of the study participants was and aged 2–12 years. There were no serious AEs in C1% of
hospitalized for acute renal failure, anaphylaxis or Reye’s patients in either group. There were no cases of Reye’s
syndrome. Three children who received ibuprofen were syndrome, gastric bleeding and/or ulceration, renal failure,
hospitalized for GI bleeding; which was non-serious and was necrotizing fasciitis, Steven’s Johnson or Lyell’s syndromes,
resolved with conservative management. The risk of hospi- anaphylaxis or any other serious condition that are known to
talization from GI bleeding was estimated to be 11 per be associated with either drug in any population.
100,000 (95% CI 2.2–32 per 100,000) for antipyretic Statistically significant but small and clinically in-sig-
assignment and 17 per 100,000 (95% CI, 3.5–49 per nificant differences were observed in AEs in both age
100,000) in those children B2 years who received ibuprofen. groups that received ibuprofen compared with those that
Among children \6 months of age there was no observed had paracetamol being 17.6% c.f. 15%, respectively in the
risk for hospitalization for any of the primary outcomes. younger and 11.9 and 10.7% in the older groups. The
The risks with hospitalization for asthma/bronchiolitis increased incidence of AEs in the ibuprofen groups was
for ibuprofen were 0.9 (95% CI, 0.5–1.4) compared with related to the greater disease severity in those groups. Four
paracetamol; a total of 65 children being hospitalized for deaths were recorded (herpes encephalitis, sepsis due to
these group of conditions (they were grouped together Staphyloccocus pneumoniae, medulloblastoma and sudden
because of frequent misdiagnosis of these two conditions). death syndrome) and were unrelated to the study medica-
Of nine children hospitalized for vomiting or gastritis the tions. Those AEs related to the special senses occurred
risk did not vary according to antipyretic assignment. occasionally followed by the digestive and respiratory
Of the 385 who were hospitalized, those in whom creat- systems and skin (all in 3–4% approximately in the
inine levels were available (29%) and were considered to be younger and slightly lower in the older group).
only of borderline statistical significance between the ibu-
profen and paracetamol groups. There was no significant
differences between these two treatment groups when age, Specific organ toxicity
weight, sex or admission diagnosis of dehydration were
compared. When alternate cut-off points were used to define Gastro-intestinal (GI) toxicity
an elevated creatinine level (44 or 53 lmol L-1) there were
no significant differences between the antipyretic groups. Serious GI ADRs (upper GI bleeding and ulcers) are a
While this was the largest controlled study ever under- major cause of concern and in the past 3–4 decades have
taken of antipyretic use in children B6 months of age the aroused much interest among clinicians, experimentalists
authors admitted that the power to detect serious adverse and regulators alike (Voutilainen et al. 1998; Wolfe et al.
events is limited (especially those that occur at low fre- 1999; Lewis et al., 2005; Schaffer et al. 2006; Arroyo and
quency). Some clinical and demographic information Lanas 2006; Lanas et al. 2006; Laine et al. 2006). The
suggested that the study participants probably reflected a problems are particularly apparent in rheumatic patients
wide spread of febrile illnesses in the view of the authors (Singh et al. 1996; Singh 2000) and the elderly (Griffin
even though socioeconomic data were limited. 1998; Beyth and Schorr 1999; Seinelä and Ahvenainen
These data are important in showing that (a) there is a 2000; Mamdani et al. 2002; Kean et al. 2008). Definitions
remarkable low incidence of serious and even non-serious vary on what constitutes the elderly but most agree [65, a
Ibuprofen 311

time that seems to have derived over a century ago from Table 19 Risk factors for the development of NSAID-associated
Otto von Bismark who required Prussian officers to retire gastro-duodenal ulcers
at this age) (Kean and Buchanan 1987; Buchanan 1990). Established risk factor Possible risk factor
Early studies indicated that ibuprofen was well-tolerated in
Advancing age High Alcohol consumption
elderly patients (Buckler et al. 1975).
A range of factors influence the development of NSAID- High dose NSAID or paracetamol Cigarette smoking
associated GI ulcerations and bleeding (Table 19). This Use of more than 2 NSAIDs Helicobacter pylori Infection
makes it difficult to ascribe a quantitative component of the Concurrent paracetamol
NSAID to the occurrence of serious GI events. Concurrent anti-coagulants
A number of studies have reported that prescription Concurrent aspirin (even low dose)
dosage level ibuprofen produces time and dose-dependent Prior history of peptic ulcer disease
blood loss (assessed using the radiochromium blood loss Rheumatoid arthritis
technique) from the GI tract of volunteers or patients From Wolfe et al. (1999) and Laine (2001); modified and with
(Warrington et al. 1982; Aabakken et al. 1989; Hunt et al. additional information from Rainsford (1984; 2004a, 2005b)
2000) and mild-moderate endoscopic changes in fasted
human volunteers (Lanza et al. 1979, 1981, 1987; Fried-
man et al. 1990; Bergmann et al. 1992; Roth et al. 1993; was able to show that the relative risks of these events from
Müller et al. 1995; Gallego-Sandin et al. 2004) or those different NSAIDs ranged considerably (Fig. 7). They
with rheumatic diseases (Teixeira et al. 1997). The extent found that ibuprofen had the lowest risks for developing GI
of the loss of blood maybe overestimated using the radio- complications (Fig. 7).
chromium technique as a consequence of biliary excretion Henry et al. (1996, 1998) also observed (a) dose-related
of the radiolabelled chromium (Schneider et al. 1984; occurrence of GI complications with ibuprofen, naproxen
Rainsford 2004a). The extent of the mucosal changes and indomethacin (Fig. 7), and (b) the ranking of GI
(lesions, ulcers) and blood loss from ibuprofen is low complications was directly related to the plasma elimina-
compared with other NSAIDs but is above that of placebo tion half-life (t1/2) of the individual NSAIDs (Tables 20, 21
and paracetamol (Strom et al. 1997; Rainsford 1999c). and Fig. 8). Again, as with the overall analysis ibuprofen
had the lowest rates of occurrence of GI complications
Epidemiological studies of GI risks which is attributed to its short t1/2 (*2 h). Thus, there is
good pharmacokinetic rationale to account for the low GI
A considerable number of population studies have been ADRs with ibuprofen.
reported over the past 2–3 decades comparing the occur- Among the most comprehensive studies that have been
rence of serious GI events from ibuprofen and other performed to evaluate overall adverse drug reactions in
NSAIDs, at prescription dosage level dosages, with some European populations has been the study by Lugardon et al.
studies being dose-ranging (Kaufman et al. 1993; Lang- (2004). These authors undertook an analysis of spontane-
man et al. 1994; Henry et al. 1993, 1996; MacDonald ous reports to the French Pharmacovigilance network
et al. 1997; Henry and McGettigan 2003; Hippisley-Cox which is probably one of the most extensive and compre-
et al. 2005; Thomsen et al. 2006; see also Table 20). The hensive pharmacovigilance systems in Europe. A summary
study designs, outcomes measures and variables (dosage of the data shown in the Table 22 compares the reporting
and duration) vary considerably among these studies. odds ratios for GI events of heteroarylacetic acids that
Some measures having included the occurrence of peptic include ibuprofen, diclofenac, naproxen and ketoprofen
ulcer bleeds (PUBs), upper GI bleeding, ulcers viewed at with that of the two principle coxibs rofecoxib and cele-
endoscopy (usually investigated as a consequence of coxib as well as the oxicams, principally meloxicam,
clinical symptoms or as part of a planned investigation) or piroxicam and tenoxicam. The unadjusted and adjusted
the more general grouping of ‘‘serious events’’. While odds ratios for ibuprofen ADRs are amongst the lowest all
these studies vary considerably they are useful in com- the drugs that were studied by Lugardon et al. (2004).
paring the risks of serious GI events attributed to A similar conclusion can be drawn from the case–con-
ibuprofen with that of a range of other NSAIDs with trol study of Laporte et al. (2004) as shown in Table 23.
known ulcerogenicity. Case–control investigations by Garcia-Rodriguez and
A summary of some of the population studies reported Hernandez-Diaz (2001) using data from the UK General
in the 1990s in the period before the introduction of the Practice Database also show the low risks of GI events with
newer class of coxib NSAIDs is shown in Table 20. ibuprofen in contrast with those from various doses and
In a meta-analysis of published studies comparing the periods of taking NSAIDs with or without concurrent
GI ADRs for various NSAIDs, Henry et al. (1996, 1998) aspirin (Table 24). These data are also instructive in
312 K. D. Rainsford

from random-digit phone dialling in the same region. Use


of OTC doses of non-aspirin NSAIDs C4 days in the past
week was associated with an adjusted odds ratio of 1.83
(95% CI 1.14–2.95); the risks from ibuprofen being much
lower. Risks were increased with higher doses of the drugs
confirming what has been well-known about the dose–
response relationships among most NSAIDs being associ-
ated with serious GI AEs (Henry et al. 1996, 1998).

GI effects of ibuprofen in coxib studies at prescription


doses

Data on the GI peptic ulcer bleeds (PUB), bleeding ulcers or


Fig. 7 From Henry et al. (1998). Reproduced with permission of endoscopically observed upper GI injury have been obtained
Kluwer Academic Publishers (now part of Springer Verlag) from a considerable number of large scale clinical trials
comparing the effects of coxibs with ibuprofen and other
NSAIDs at prescription doses. These data are useful for
giving and indication under controlled clinical trial conditions
in arthritic populations of the relative risks of developing
serious gastro-duodenal injury at high doses of these drugs.
Among these studies the data on ulcer complications in
the CLASS study observed at 6 months showed there were
differences between celecoxib and NSAIDs (Silverstein
et al. 2000). However, as pointed out by Jüni et al. (2002)
these differences were not apparent at 12 months
(Table 25) suggesting there are time-dependent factors that
are significant in considering ulcer incidence of both coxibs
and NSAIDs. The clinical significance of these data like
that from other long term studies is that when coxibs are
Fig. 8 From Henry et al. (1998). Reproduced with permission of taken for relatively short periods of time (2–4 weeks) they
Kluwer Academic Publishers (now part of Springer Verlag)
are less likely to cause ulcer complications than NSAIDs
such as naproxen and diclofenac especially if they are
highlighting the fact that high dose paracetamol (hitherto taken for several months or longer. There is also the issue
regarded as a GI safe drug) when taken at doses of of what has been described as ‘‘channelling’’ where
[2 g day-1 alone or in combination with NSAIDs is patients with a history of GI complaints or GI ulcer disease
associated with relative risks [2 (alone) or [6 (combina- may be prescribed coxibs in the belief they will be ‘‘gas-
tion with NSAIDs) of haemorrhage. tric-safe’’, this may be such that benefits for using these
In rheumatic patients, Singh (2000) has produced data dugs may be less apparent and the patients may require
from the ARAMIS (a US rheumatic disease patient data- anti-ulcer co-therapy (e.g. with H2 receptor antagonists or
base) showing relatively high risks of GI bleeding (or proton pump inhibitors [PPIs]). The cost-benefits of coxibs
peptic ulcer bleeds) from all NSAIDs, with little difference therapy may prove less favourable as not only are these
between individual NSAIDs (including ibuprofen). Lower drugs notably more expensive than conventional NSAIDs
risks were associated with paracetamol. While there have but if PPIs or other anti-ulcer therapies have to be
been claims made by this author that the GI risks are employed they may as well be given with cheaper NSAIDs,
similar to those from OTC dosages of these analgesics especially those with a lower propensity to cause CV
there is little information available on their duration of use, complications (e.g. naproxen) or combinations with aspirin
concomitant medications and other risk factors. for cardioprotection.
More insight into the GI risks associated with OTC These calculations (Table 26) show that although the
analgesics/NSAIDs has been provided by Lewis et al. percentage of relative risk reduction (RRR) for celecoxib
(2005) in a case–control study of hospitalized patients c.f. NSAIDs (ibuprofen, diclofenac) is 61% and for ro-
recruited from 28 hospitals. The cases (N = 359) had fecoxib c.f. naproxen is 60% the values for absolute risk
upper GI bleeding, benign gastric outlet obstructions or reduction (ARR) are relatively small being 0.7 and 0.8%,
perforations, while controls (N = 1,889) were obtained respectively (Shoenfeld 2001). The latter values represent
Ibuprofen 313

Table 20 Serious outcome gastro-intestinal toxicity ranking of tNSAIDs


Drug Kaufman et al. Henry et al. Langman et al. Rodrigues and Jick Henry et al. MacDonald et al.
(1993) (1993) (1994) (1994) (1996) (1997)

Aspirin 10
Azapropazone 1 1 2
Diclofenac 6 7 6 7 9 4
Diflunisal 1 8 7
Fenbufen 11
Fenprofen 4 5 11 1
Ibuprofen 7 8 7 9 12 8
Indomethacin 5 3 4 4 5 9
Ketoprofen 1 2 2 2 2 5
Nabumetone 10
Naproxen 3 4 5 3 6 6
Mefenamic acid 7
Piroxicam 2 5 3 1 3 3
Sulindac 6 6 8
Tolmetin 4
Toxicity rankings of NSAIDs with those associated with the greatest risk of ulcer complication is given the number 1. The studies used different
methodologies

Table 21 Ranking of GI complications from NSAIDs with plasma Table 22 Adverse drug GI reaction reporting odds ratio (OR) (with
elimination Half-Life (t1/2) of the drugs their 95% confidence interval) according to main classes of non-
steroidal anti-inflammatory drugs (NSAIDs)
G-I safety, dose and plasma half-life of NSAIDS
Ranking of RR of ulcers c.f. t1/2 (h) Drugs Adjusted ORa (95% CI) Adjusted ORb (95% CI)
Ibuprofen (2.5) \ Diclofenac (1.5–5) \ Diflunisal Coxibs 4.6 (3.3–6.5)* 14.9 (9.3–23.7)*
(10.8) \ Fenprofen (2.2) \ Aspirin (0.5–4.5) \ Sulindac
(14.0) \ Naproxen (14.0) \ Indomethacin (3.8) \ Piroxicam Rofecoxib 5.2 (3.1–8.7)* 21.0 (10.6–41.6)*
(48.0) \ Ketoprofen (8.5) \ Tometin(6.8) \ Azapropazone (22.0) Celecoxib 3.7 (2.4–5.8)* 11.7 (6.6–20.9)*
Dose-relationships—low c.f. high dose: Oxicams 12.2 (6.7–22.2)* 25.3 (11.9–53.6)*
Ibuprofen RR 1.8–4.0 Heteroaryl acetic acids
Naproxen RR 3.8–6.0 Ibuprofen 4.5 (2.3–8.8)* 7.3 (3.2–16.6)*
Indomethacin RR 2.3–6.5 Diclofenac 3.9 (2.1–7.2)* 9.2 (3.8–22.2)*
Naproxen 10.6 (4.7–23.7)* 17.9 (6.7–47.6)*
From Henry et al. (1998)
Ketoprofen 8.6 (5.3–13.9)* 19.9 (10.7–37.0)*
a
the fact that the percentage incidence of GI complications Adjustment for matching factors (age, sex, period of occurrence)
b
for the NSAID as well as for the two coxibs is in the low Adjustment for matching factors (age, sex, period of occurrence)
and confounding factors (regional pharmacovigilance centre, work
end range of 0.4–1.14% which are very low percentages.
place of health professional and drug-exposure (anticoagulants, anti-
Thus, with such small differences calculations of RRR are platelet drugs, aspirin, gastroprotective agents, and other NSAIDs)
meaningless and give a false impression of improved From Lugardon et al. (2004)
benefit to the GI tract of the coxibs. This approach of using * P \ 0.0001
RRR percentage benefits has been extensively exploited in
published data on coxib trials and must, therefore, be 150 mg day-1 taken for 12 weeks. This study was of
regarded as suspect statistical treatment of data which has shorter duration than the other coxib studies reviewed
little relevance clinically. Indeed clinical significance in previously. There is a trend noted by Jüni et al. (2002) for
many coxib trials has rarely been considered in contrast to differences in GI events between celecoxib and the NSA-
statistical significance. IDs to become smaller with time. Thus at the shorter time
In another large scale coxib study Sikes et al. (2002) period it might have been expected that if valdecoxib had a
compared the incidence of gastric and duodenal ulcers favourable GI profile that it would show lower incidence of
from two dose levels of valdecoxib 10 and 20 mg day-1 GI events compared with the two comparator NSAIDs. The
with that from ibuprofen 2,400 mg day-1 and diclofenac data in Table 27 shows that there was a somewhat higher
314 K. D. Rainsford

Table 23 Gastro-intestinal
Drug Cases Controls Odds ratio Population
bleeding from NSAIDs in a
(no (%)) (no (%)) (95% CI) attributable
multicentre case–control study
risk (%)
in Spain and Italy
NSAIDs
Aceclofenac 15 (0.5) 30 (0.4) 1.4 (0.6, 3.3) –
Aspirin (acetylsalicylic acid) 591 (21.1) 403 (5.7) 8.0 (6.7, 9.6) 18.5
Dexketoprofen 16 (0.6) 8 (0.1) 4.9 (1.7, 13.9) 0.5
Diclofenac 100 (3.6) 98 (1.4) 3.7 (2.6, 5.4) 2.6
Ibuprofen 60 (2.1) 58 (0.8) 3.1 (2.0, 4.9) 1.5
Indomethacin 29 (1.0) 16 (0.2) 10.0 (4.4, 22.6) 0.9
Ketoprofen 16 (0.6) 9 (0.1) 10.0 (3.9, 25.8) 0.5
Ketorolac 33 (1.2) 6 (0.1) 24.7 (8.0, 77.0) 1.1
Meloxicam 14 (0.5) 11 (0.2) 5.7 (2.2, 15.0) 0.4
Naproxen 52 (1.9) 27 (0.4) 10.0 (5.7, 17.6) 1.7
Nimesulide 48 (1.7) 46 (0.6) 3.2 (1.9, 5.6) 1.2
Piroxicam 119 (4.3) 40 (0.6) 15.5 (10.0, 24.2) 4
Rofecoxib 10 (0.4) 10 (0.1) 7.2 (2.3, 23.0) 0.3
Other NSAIDs 34 (1.2) 33 (0.5) 3.6 (2.0, 6.8) 0.9
NSAIDs ? antiplatelet drugs 140 (5.0) 54 (0.8) 16.6 (11.3, 24.2) 4.7
Analgesics
Lysine Clonixinate 26 (0.9) 47 (0.7) 1.3 (0.7, 2.6) –
Metamizole 117 (4.2) 155 (2.2) 1.9 (1.4, 2.6) 2
Paracetamol (acetaminophen) 376 (13.4) 612 (8.6) 1.2 (1.0, 1.5) –
Propyphenazone 17 (0.6) 38 (0.5) 1.3 (0.6, 2.8) –
From Laporte et al. (2004)

incidence of gastric and duodenal ulcers and all GI ulcers conventional NSAIDs, among which etodolac, ibuprofen,
from ibuprofen compared with valdecoxib, but these were nabumetone and possibly diclofenac (although the intesti-
fewer than seen with diclofenac. nal ulceration and hepatotoxicity reduce any favourable a
This study is of importance in that there were separate favourable safety profile it might have had).
analysis of ulcer incidence in (a) H. pylori negative and H. An interesting and possibly important point that should
pylori positive subjects, and (b) between those who were be considered is the arthritic condition in the CLASS as
aspirin takers (for CV prophylaxis) and non-takers. well as other studies with the coxib. Patients in the VIGOR
The data in Table 28 shows that H. pylori status made only had RA half of whom were receiving corticosteroids
little if any difference to the ulcer incidence in subjects but were not allowed aspirin at cardioprotective doses
that received any of the drugs, but taking of aspirin did (Silverstein et al. 2000) whereas those in the CLASS study
increase the incidence of ulcers in the ibuprofen and had both RA and OA and were allowed aspirin (Bombar-
naproxen groups and to a lesser extent in the valdecoxib dier et al. 2000). It has been claimed that there were no
groups. differences in ulcer complications in patients with OA c.f.
From the point of view of GI safety, there may have RA which is surprising in view of the differing medications
been pathological consequences of hepato-renal ADRs and allowed in these studies. This is in one sense surprising
hypertension that contributed to the vascular aetiology of since as noted earlier it has been speculated that patients
upper GI ulcer disease as well as the consequences of the with RA may be more susceptible to NSAIDs than those
diuretics and anti hypertensive drugs (which as noted ear- with OA. It could be that the selection criteria for patients
lier increase the risk for developing ulcers) as well as entered in the CLASS Study were such that RA as well as
NSAID-drug interactions that patients with hepato-renal OA patients were relatively ‘‘fit’’ and without complicating
conditions and hypertension received for treatment of these chronic conditions that inevitably occur in older more in-
conditions (Rainsford et al. 2008). In the end, what has firmed patients, especially those with RA (Kean et al.
emerged in the safety analysis of the coxibs is summarized 2008).
in Table 26 it is clear that the benefits of what now are Indeed the incidence of ulcers in the CLASS study
classed as ‘‘first generation’’ coxibs (celecoxib, rofecoxib, (Tables 25, 26) as well as in the meta analysis of celecoxib
valdecoxib) may have been marginal compared with some trials by Moore et al. (2005) reveal a remarkably low
Ibuprofen 315

Table 24 Epidemiological data from general practice database (UK) Table 26 Estimation of serious gastrointestinal reactions from the
on peptic ulcer bleeding risks from aspirin and other NSAIDs and CLASS trial of celecoxib compared with NSAIDs
paracetamol and other sources
In the CLASS Trial (non-aspirin-using patients only)
Drug Usage/factor Relative Percentage of patients with serious NSAID-associated
risk gastrointestinal complications was:
Ibuprofen Lowest risk *1.0–2.0 Celecoxib = 0.44% diclofenac = 0.48% (no statistically significant
(dose-dependent) difference between Diclofenac and celecoxib)
Aspirin Overall use Users 2.0 Celecoxib = 0.44% Ibuprofen = 1.14%
cf Non-users 1.0 ARR = 1.14 - 0.44% = 0.7%
Recent users 1.5 NNT = 1/0.7% = 1/0.007 = 143
Past users 1.1 RRR = 1.14% - 0.44%/1.14% = 61%
Dose 75–300 mg day -1
2.1 Results are reported as serious NSAID-associated gastrointestinal
complications (i.e. gastrointestinal bleeds, perforations and
[400 mg day-1 3.1 obstructions) per 100 patient-years
\50 mg day-1 0.7 ARR = absolute risk reduction; RRR = relative risk reduction
Period of use 1–60 day 4.5 NNT = number needed to treat
61–180 day 2.7
From Schoenfeld (2001)
181–730 day 1.0
[730 day 1.6
incidence in placebo and NSAID groups. This gives sup-
Paracetamol \1 g day-1 1.0
-1 port to the view that patients selected for inclusion in these
1–2 g day 0.9
-1 studies may have been of relatively better health. Another
2–4 g day 3.4
issue is that if there were any real differences in data
[4 g day-1 6.5
concerning ulcer complications in say a proportion of
2 g with NSAID 4.2
patients with RA c.f. those with OA these may have been
[2 g with NSAID 13.5
disguised in the grouping of data together such as in the
cf NSAID alone 3.5
CLASS results.
NSAIDs Low dose 2.5
In conclusion, the epidemiological and large-scale
High dose 5.0
clinical trials show that ibuprofen has amongst the lowest
Duration 1–30 days 4.3
risk of NSAIDs for serious GI events. The concomitant
[730 days 3.5
ingestion of aspirin may raise the risk of GI complications
Formulation Plasma t1/2 in a similar way to that seen with celecoxib and rofecoxib
\12 h (high dose) 4.2 (Strand 2007).
C12 h 5.4
(slow release 6.2 GI symptomatic adverse reactions
\12 h (low dose) 2.4
C12 h 2.8 GI symptoms (nausea, heartburn, epigastric distress, vom-
Data from Garcia-Rodriguez and Hernandez-Diaz (2001) iting, diarrhoea) are among the major reasons for
withdrawal by patients from therapy with NSAIDs. Much
effort has been undertaken to establish if there is a rela-
tionship between symptoms and gastric injury but in
general the results have been inconclusive. Yet often
Table 25 Summary of adverse events in the CLASS study symptoms are a cause for referral for endoscopic or other
Event % Celecoxib Diclofenac Ibuprofen clinical investigations.
Meta-analysis of the tolerability and adverse events
GI 45.6 55.0 46.2
from a range of trials of celecoxib compared with NS-
Withdrawal 12.2 16.6* 13.4
NSAIDs, paracetamol and placebo (using data from the
Renal 6.8 6.7 10.3*
published and unpublished trials from Pfizer) (Moore et al.
Withdrawal 1.0 0.6 1.3
2005) revealed some interesting features and trends con-
CV non-ASA 1.6 1.2 0.4
cerning the occurrence of GI symptoms notably nausea,
Hepatic 1.8 6.9* 1.9
dyspepsia, diarrhoea, abdominal pain, vomiting. These
Withdrawal 0.3 3.5* 0.3 constitute main reasons (other than ulcer/bleeds or other
Based on data published by Silverstein et al. (2000) serious ADRs) for withdrawal from therapy, and indeed the
* Significantly different compared with celecoxib data by Moore et al. confirmed this pattern.
316 K. D. Rainsford

Table 27 Incidence rate of upper-gastrointestinal ulcers from ibuprofen compared with diclofenac and valdecoxib
Placebo Valdecoxib Valdecoxib Ibuprofen Diclofenac
10 mg daily 20 mg daily 800 mg t.i.d 75 mg b.i.d.

12-week cohort (n (%))


N 123 142 157 149 145
Gastroduodenal 8 (7) 7 (5) 7 (4) 24 (16)a,b,c 25 (17)a,b,c
a,b,c
Gastric 6 (5) 5 (4) 6 (4) 20 (14) 20 (14)a,b,c
Duodenal 3 (2) 2 (1) 1 (1) 6 (4) 7 (5)c
ITT cohort (n (%))
N 178 189 198 184 187
Gastroduodenal 8 (4) 7 (4) 7 (4) 25 (14)a,b,c 25 (13)a,b,c
Gastric 6 (3) 5 (3) 6 (3) 21 (11)a,b,c 20 (11)a,b,c
Duodenal 3 (2) 2 (1) 1 (1) 6 (3) 7 (5)c
12-week cohort includes patients who took the study medication for the entire 12-week period. ITT cohort includes all patients who had a post-
treatment endoscopy irrespective of whether they completed 12 weeks of treatment
From Sikes et al. (2002)
a
Significantly different from placebo at P [ 0.05
b
Significantly different from valdecoxib 10 mg daily at P [ 0.05
c
Significantly different from valdecoxib 20 mg daily at P [ 0.05

Table 28 Week 12 gastroduodenal ulcer incidence (%) by aspirin use, age or Helicobacter pylori status
Group Placebo Valdecoxib Valdecoxib Ibuprofen Diclofenac
10 mg daily 20 mg daily 800 mg t.i.d 75 mg b.i.d.

H. pylori-positive 4/46 (9)a,b 3/40 (8)a,b 2/49 (4)a,b 9/45 (20) 11/54 (20)
H. pylori-negative 4/119 (3)a,b 3/134 (2)a,b 5/136 (4)a,b 15/123 (12) 14/122 (12)
H. pylori status unknown 0/2 (0) 1/3 (33) 0/6 (0) 0/7 (0) 0/5 (0)
Taking aspirin 0/26 (0)a,b 3/16 (19)a,b,c 2/29 (7)a,b 10/31 (32)c 10/33 (30)c
Not taking aspirin 8/141 (6)a,b 4/161 (3)a,b 5/162 (3)a,b 14/144 (10) 15/148 (10)
a,b a,b,d
Age C 65 years 4/65 (6) 6/56 (11) 3/67 (5)a,b 15/71 (21)d 14/78 (18)
Age [ 65 years 4/102 (4)a,b 1/121 (1)a,b 4/124 (3)a,b 9/104 (9) 11/103 (11)
From Sikes et al. (2002)
a
P B 0.014 vs. ibuprofen
b
P B 0.008 vs. diclofenac
c
P B 0.017 vs. not taking aspirin
d
P B 0.025 vs. age \ 65 years

Data on the GI symptoms from NSAIDs and coxibs paracetamol or rofecoxib, but there were more patients on
summarised in the report by Moore et al. (2005) highlight NS-NSAIDs. Celecoxib was responsible for abdominal
that (1) the occurrence and relative risks of most GI pain in about 5% patients there being no difference c.f.
symptoms in patients receiving celecoxib, rofecoxib or placebo or paracetamol but more patients on NS-NSAIDs
paracetamol being greater than that of placebo, (2) while and rofecoxib experienced this adverse effect. Other GI
there are trends for a lower incidence of some symptoms symptomatic effects as well as overall GI tolerability there
with low dose celecoxib the differences are less distinct were trends in favour of celecoxib in comparison with the
with higher dose celecoxib, and (3) the data on confidence other treatments but the 95% confidence intervals for rel-
intervals in relative risks with most comparisons often ative risk often overlapped those of comparator drugs. This
overlaps to the extent that it is doubtful if any differences, makes extrapolation of these findings difficult from what
especially those favouring celecoxib, have any meaning. are relatively small values for incidence and percentage
The authors of this study noted that the proportion of differences of symptomatic GI ADRs like that of clinical
patients having dyspepsia was about 7% and that there ulcers and bleeds in this meta analysis (Moore et al. 2005)
were no differences in comparison with placebo, is probably of limited value.
Ibuprofen 317

GI events at OTC dosages compared with 0.1% that had paracetamol, while in the older
group the incidence was 0.6 and 0.2% for ibuprofen and
GI symptoms (nausea, epigastric or abdominal pain, dys- paracetamol, respectively.
pepsia, diarrhoea, flatulence and constipation) are among These results attest to the relative gastric safety in
more frequent reactions observed with OTC use of ibuprofen children of ibuprofen in comparison with paracetamol
as well as with paracetamol and aspirin, and generally the accord with earlier investigations (Walson et al. 1989;
symptoms are of the same order as in subjects who have Czaykowski et al. 1994; Rainsford et al. 1997; Diez-Do-
received placebo (Rainsford et al. 1997, 2001; Doyle et al. mingo et al. 1998). They confirm the safety of both drugs
1999; Kellstein et al. 1999; Ashraf et al., 2001; Le Parc et al. being comparable and relatively low in the open clinical
2002; Boureau et al. 2004; Biskupiak et al. 2006). The paediatric study.
occurrence of GI symptoms with ibuprofen has often been
found to be lower than with aspirin and comparable with Cardiovascular safety
those from paracetamol (Rainsford et al. 1997; Moore et al.
1999; Le Parc et al. 2002; Boureau et al. 2004). Serious GI There are three main issues concerning cardio-vascular
reactions are rare and have not been reported in significant (CV) safety of ibuprofen. The first of these concerns the
numbers in trials with OTC ibuprofen (Doyle et al. 1999; possible risks of triggering serious CV conditions such as
Kellstein et al. 1999; Ashraf et al. 2001; Le Parc et al. 2002; congestive heart failure (CHF) and myocardial infarction
Boureau et al. 2004). Thus, it may be concluded that GI (MI); a situation which has arisen as a consequence of the
events are essentially non-serious with OTC ibuprofen, are re-evaluation of risks of MI from all NSAIDs following the
probably reversible upon cessation of the drug (an action identification of risks of this condition with rofecoxib and
likely to be taken by most subjects), and are no different other coxibs (Purcell 2007; Strand 2007; Layton et al.
from those with paracetamol and less than with aspirin. 2008; Solomon et al., 2008; Sørensen et al. 2008; van der
Linden et al. 2008). The second issue concerns the effects
GI safety in paediatric populations of NSAIDs, including ibuprofen, on blood pressure in
hypertensive individuals; elevation of blood pressure being
As noted earlier in the discussion of the large-scale pae- regarded as a potential marker for risks of MI or stroke
diatric study by Lesko and Mitchell (1995) four children (Topol 2004, 2005; White 2007). This situation is com-
were hospitalized for acute GI bleeding that was due to plicated because most NSAIDs reduce the effectiveness of
ibuprofen (2 from 10 mg kg-1 and 2 from 5 mg kg-1 of anti-hypertensive drugs and diuretics as a consequence of
the drug) which gives a risk of GI bleeding as 7.2 per blunting by NSAIDs of these drugs on their prostaglandin-
100,000 (95% CI 2–38 per 1000,000) with the risk from mediated actions (Hersh et al. 2007; Ishiguro et al. 2008;
paracetamol being zero; the difference was not statistically White 2008). Linked to the effects of NSAIDs in elevating
significant. Gastritis/vomiting was observed in 20 patients blood pressure are their effects on renal functions which
that had received ibuprofen with a risk of 36 per 100,000 can contribute to their hypertensive potential as a conse-
(95% CI 22–55) and in 6 patients on paracetamol with a quence of inhibition of renal prostaglandin production
risk of 21 per 100,000 (95% CI 7.9–46). (White 2007). This has given rise to the so-called ‘‘cardio-
In their later study in B2 year olds, Lesko and Mitchell renal’’ syndrome of NSAIDs and again has come from
(1995) observed that three children who received ibuprofen recognition of the pronounced renal effects of coxibs as a
were hospitalized for GI bleeding. The risk of hospitaliza- consequence of inhibition of COX-2 in the macula densa
tion from GI bleeding was estimated to be 11 per 100,000 (Harris et al. 1994; Haas et al. 1998; Khan et al. 1998;
(95% CI 2.2–32 per 100,000) for antipyretic assignment and Inoue et al. 1998; Ichihara et al. 1999; Wolf et al. 1999;
17 per 100,000 (95% CI, 3.5–49 per 100,000) in those Roig et al. 2002); aspects of this are discussed under
children B2 years who received ibuprofen. ‘‘Renal Toxicity’’. The third issue is the possibility that
As noted earlier in the discussion of the large-scale pae- ibuprofen might reduce the anti-platelet effects of aspirin
diatric study by Ashraf et al. (1999) no occurrences of gastric and thus reduce the anti-thrombotic effectiveness of the
bleeding or ulcers were observed with either ibuprofen or latter (Purcell 2007).
paracetamol. The incidence of adverse events (AEs) in the
digestive system was 3.0 and 2.1% in the younger group Serious CV conditions
(B2 years) that received ibuprofen or paracetamol and 2.1
and 1.2% for these drugs in the older group (2–12 years); the The available evidence reviewed here shows that ibuprofen
statistical tests showed the former being non-significant but has low CV risks although there may be effects on blood
in the latter this was statistically significant. Abdominal pain pressure and on the actions of drugs used to control blood
occurred in 0.6% of the younger patients that had ibuprofen pressure.
318 K. D. Rainsford

Serious CV events principally ischaemic heart condi- which the risks of MI or other serious CV accidents have
tions were initially highlighted by long term studies with been examined. Among these Garcia-Rodriguez et al.
the coxibs (Topol 2004, 2005; Kanna et al. 2005; Ostor and (2004) employed data in the UK General Practice Research
Hazleman 2005; Rainsford 2005a; White 2007). The CV Database (GPRD) which records reports from GP’s sent
events included myocardial infarction and hypertension anonymously to the UK MHRA. This database records
and were noted particularly with rofecoxib (Vioxx) in the demographic and patient data and over 90% of GP referrals
VIGOR study as well as in a number of other studies. They along with prescription details. The associations of MI with
were of sufficient concern for the company producing this various patient and risk factors were discriminated in this
drug, Merck Sharp & Dohme, to withdraw it from the study. The odds ratios (ORs) for development of MI after
market on 30th September 2004 (Topol 2004, 2005; multi-variant adjustment with current use of NSAIDs were
American College of Rheumatology, Hotline, 2005; Psaty found to have an OR = 1.06 (0.87–1.29; 95% CI values)
and Furberg 2005). In the wake of the issues surrounding for ibuprofen, contrasted with those at the upper end of risk
withdrawal of rofecoxib the FDA determined that valdec- with piroxicam with an OR of 1.25 (0.69–2.25). Prior
oxib (Bextra, Pfizer Inc.) had similar CV risks and this as history of CHD or concomitant intake of aspirin did not
well as the skin reactions that emerged with this drug lead increase the risk of MI from ibuprofen.
to its withdrawal in 2005. Jick et al. (2006) performed a case–control analysis of
The FDA and other agencies worldwide were alerted and data from the UK GPRD did not find any increased risk of
alarmed about the CV ADRs with rofecoxib such that acute MI with either ibuprofen or naproxen, but did find
extensive reviews were undertaken by these agencies increased risks with diclofenac, rofecoxib and celecoxib.
world-wide of both coxibs and NSAIDs based on the Kimmel et al. (2004) performed a study of hospitalized
somewhat unfounded premise that inhibition of COX-2 patients for MI in a five-county region around Philadelphia
which occurs with all these drugs might well underlie the (USA). They observed reductions in the risks of MI among
increased risks of MI and elevation of blood pressure. It is non-aspirin NSAID users. The reduction was also observed
important to note that the VIGOR study investigating the with ibuprofen with the adjusted OR being 0.52 (0.39–
long-term GI effects of rofecoxib was performed in patients 0.69; 95% CI) compared with that of 0.53 (0.42–0.67) for
with rheumatoid arthritis (RA). RA patients are known to aspirin and 0.48 (0.28–0.82) for naproxen, a drug which
have a markedly higher risk of developing MI and other has often been found to have low CV risk and in fact may
serious CV events (Nurmohamed et al. 2002; Assous et al. have some cardio-protective actions (Topol 2004, 2005;
2007) and this is not a feature generally recognized in the Kanna et al. 2005; Kean et al. 2008).
assessment of CV risks of coxibs and NSAIDs. Further- In a nested case–control study of data from a leading US
more, there are indications from the studies performed with health maintenance organization, Kaiser Permanente,
the coxibs in long-term preventative studies in cancer or Graham et al. (2005) examined 8,143 cases of serious
Alzheimer’s disease that high doses of these drugs were coronary disease (from 2,302,029 patient-years follow-up).
employed and these patients were clearly very sick. They found that the adjusted OR for ibuprofen for current
Etoricoxib (Arcoxia) also developed by Merck Sharp use was 1.26 (1.00–1.60), compared with that of naproxen
and Dohme is probably the most selective inhibitor of OR = 1.36 (1.06–1.75) and rofecoxib low dose
COX-2 of those drugs that have been developed to date. (\ 25 mg day-1) OR = 1.47 (0.99–2.17) and high dose
While long-term investigations are awaited there are indi- ([25 mg g-1) OR = 3.58 (1.27–10.11). For remote use
cations that GI and CV events from etoricoxib may be the OR for ibuprofen was 1.06 (0.96–1.17) contrasted with
lower than with NSAIDs and celexocib. that of rofecoxib (high dose,[25 mg day-1) with an OR of
Lumiracoxib (Prexige; Novartis) is not chemically like 3.0 (1.09–8.31).
that of other coxibs as it is a derivative of diclofenac. It also In a combined study of CV and GI events in 49, 711 US
does not have the high COX-2 selectivity of etoricoxib or Medicare beneficiaries ([65 years of age) Schneeweiss
rofecoxib. Novartis embarked on large scale studies to et al. (2006) found that the risks of acute MI was 1.20 with
determine the CV safety with lumiracoxib which was ibuprofen, compared with 1.01 with naproxen, 1.54 with
compared with ibuprofen and naproxen all at prescription diclofenac, 1.58 with celecoxib and 1.56 with rofecoxib.
level dosages (Schnitzer et al. 2004; Matchaba et al. 2005; Another nested case–control study using clinical
Farkouh et al. 2009) (see Tables 30, 31). records of the UK general practice database (known as
QRESERCH) Hippisley-Cox and Coupland (2005) found
Epidemiological studies that recent use (\3 months) of ibuprofen was associated
with an adjusted OR of 1.24 (1.11–1.39), compared with
The awareness of CV risks from coxibs and NSAIDs has that of diclofenac which had an OR of 1.55 (1.39–1.72),
led to a substantial number of studies have been reported in naproxen 1.09 (0.96–1.24), celecoxib 1.14 (0.93–1.40)
Ibuprofen 319

and rofecoxib 1.05 (0.89–1.24). The lack of any signals lowest overall risk of all NSAIDs for developing CV
with rofecoxib and to some extent with celecoxib is odd events. For ibuprofen the relative risks range from 1.07 for
but may reflect more limited use of this drug according to all CV, mostly MI, to all vascular events 1.51. It should be
the guidelines by the UK National Institute for Clinical noted, however, that the confidence intervals for these risks
Excellence (NICE). overlap considerably and are approximately unity in com-
A retrospective study of hospitalization records of C65- parison with placebo.
year-old patients admitted for acute MI (as well as GI A review in the form of a memorandum from Dr. David J.
events) in Québéc (Canada) by Rahme and Nedjar (2007) Graham of the Office of Drug Safety of the FDA (addressed
showed that the adjusted hazard ratios for ibuprofen were to Dr. Paul Siegelman, acting Director of Office of Drug
1.05 (0.74–2.41), 1.69 (1.35–2.10) for diclofenac, 1.59 Safety) entitled ‘‘Risk of Acute Myocardial Infarction and
(1.31–1.93) for naproxen, 1.34 (1.19–1.52) for celecoxib, Sudden Cardiac Death in Patients treated with COX-2
1.27 (1.13–1.42) for rofecoxib, and 1.29 (1.17–1.42) for selective and non-selective NSAIDs’’, dated 30th Sep-
paracetamol. tember 2004 (see http://www.fda.gov/.../Drugs/DrugSafety/
Other recent studies have essentially confirmed the view PostmarketDrugSafetyInformationforPatientsandProviders/
that tNSAIDs (including ibuprofen) have risks for CV ucm1068; accessed 16 Nov 2009), showed that ibuprofen
events similar to those of some of the coxibs but the data had a risk of acute MI of 1.09 (CI 95% 0.9–1.21) compared
on risks are quite variable (Strand 2007; Layton et al. 2008; with that of naproxen relative risk 1.18 (CI 95% 1.04–1.35)
Solomon et al. 2008; Sørensen et al. 2008; van der Linden whereas that of rofecoxib ranged from 3.15 to 1.29 in a dose-
et al. 2009). related manner.
Overall, therefore, these epidemiological investigations In the large multicentre trial intended to establish the
highlight (with admittedly some variability of ORs) the risks of CV events from lumiracoxib with those from
relatively low-moderate risk of ibuprofen being associated ibuprofen or naproxen in some 14,000 patients it was found
with serious CV conditions such as MI. These observations that the number of confirmed or probable myocardial
contrast with the higher risks with diclofenac, the coxibs, infarctions and ischaemic events in the ibuprofen group
and in one study with paracetamol, and variable risks with was comparable with that from lumiracoxib as well as the
naproxen. naproxen treatment group (Fig. 9; Tables 30, 31; Farkouh
et al. 2004, 2009).
Clinical trials and meta-analyses The combination of the risks of CV and GI events has
been considered a major element in determining the overall
The CLASS study (Table 25) showed that ibuprofen had a safety of coxibs and NSAIDs (Antman et al. 2007). To
relatively low incidence of cardiovascular events and this examine this Schnitzer et al. (2004) undertook an analysis
has been confirmed in a number of other studies with of the combined risks of these drugs. A summary of their
coxibs. These large scale studies are valuable for high- data showing the combined GI and CV events is shown in
lighting under control conditions in clinical trials in Table 31 (Schnitzer et al. 2004). These data show that the
patients with rheumatic disease that ibuprofen has a low hazard ratio is significantly lower for lumiracoxib than the
risk of developing cardiovascular effects. two NS-NSAIDs although there is considerable overlap of
The most recent evaluation of the cardiovascular risks the values of the 95% confidence intervals. These data do
with coxibs and NSAIDs undertaken by Antman et al. show, however, that the combined risks of serious CV and
(2007) which was a study under the auspices of the GI events with ibuprofen are relatively low.
American Heart Association has been instructive in criti- Overall, therefore, these studies show that ibuprofen has
cally evaluating data and offering a clear statement of low risks for developing cardiovascular events, principally
cardiovascular risks of all NSAIDs including coxibs. As serious condition such as myocardial infarction although
shown in Table 29 (which is a summary of the cardio- vascular events might be slightly increased in risk.
vascular risk reported in placebo-controlled clinical trials
with the non-selective NSAIDs) the various outcome Interaction of ibuprofen with the anti-platelet effects
measures and nature of assessments from either random- of aspirin
ized controlled trials observational studies or registry data
showed that there is some variability in cardiovascular risk The possibility that ibuprofen may interfere with the anti-
with the different NSAIDs. Overall, ibuprofen has a platelet effects of aspirin was highlighted by the article by
slightly lower relative risk than diclofenac but naproxen Catella-Lawson et al. (2001) in the New England Journal of
has a notably lower risk of cardiovascular events. Indeed, Medicine. They undertook a study which aspirin (81 mg)
in the VIGOR study and several other studies that were was taken 2 h before ibuprofen (400 mg) each morning for
reviewed back in 2004 it was found that naproxen had the 6 days. The order of taking the two drugs was reversed and
320 K. D. Rainsford

Table 29 Cardiovascular risks


Type of study Outcome RR 95% CI
of ibuprofen and other NSAIDs
from meta-analysis of placebo- Versus placebo or no treatment
controlled clinical trials,
observation studies and registry Naproxen
data Meta-analysis of RCTs Vascular events 0.02 0.67–1.26
Meta-analysis of OSs CV events, mostly MI 0.07 0.87–1.07
Ibuprofen
Meta-analysis of RCTs Vascular events 1.51 0.06–2.37
Meta-analysis of OSs CV events, mostly MI 1.07 0.07–1.16
Registry Recurrent MI 1.25 1.07–1.46
Registry 1.50 1.36–1.67
Diclofenac
Meta-analysis of RCTs Vascular events 1.63 1.12–2.37
Meta-analysis of OSs CV events, mostly MI 1.40 1.16–1.70
Registry Recurrent MI 1.54 1.23–1.43
Registry 2.40 2.00–2.80
Versus selective COX-2 inhibitor
Naproxen
Meta-analysis of any non-naproxen NSAID Vascular events 0.64 0.49–0.83
From Antman et al. (2007) (primarily diclofenac or ibuprofen)
OSs observational studies, CV Meta-analysis of RCTs indicates Vascular events 1.14 0.80–1.46
cardiovascular, MI myocardial randomized, controlled trails
infarction

a similar design was incorporated with paracetamol aggregation. This impairment of platelet aggregation and
1,000 mg. Serum thromboxane B2 levels as an indicator of thromboxane production by ibuprofen was not evident with
COX-1 activity in platelets and platelet aggregation were paracetamol, diclofenac or rofecoxib.
measured and were found to be significantly inhibited by The consequence of these studies was that there were a
aspirin; the maximum inhibition being evident on day 6 considerable number of pharmaco-epidemiological inves-
when the drug was taken alone. The authors found that tigations to establish if NSAIDs would in general impair
when aspirin was given followed by ibuprofen as well as the anti-thrombotic potential of aspirin and its prevention
before taking aspirin then there was complete inhibition of of myocardial infarction. Thus, MacDonald and Wei
the effect of aspirin on serum thromboxane and platelet (2003) analysed data from the Scottish Administrative

Fig. 9 Incidence of confirmed


or probable myocardial
infarctions (clinical and silent),
from ibuprofen compared with
lumiracoxib and naproxen by
sub-study and aspirin use. From
Farkouh et al. 2004)
Ibuprofen 321

Table 30 Incidence of confirmed or probable ischaemic events including myocardial infarctions (clinical and silent), from ibuprofen compared
with lumiracoxib and naproxen by sub-study and aspirin use
Both sub-studies Lumiracoxib versus Lumiracoxib versus
ibuprofen sub-study naproxen sub-study
Lumira-coxib NSAIDs Lumira-coxib Ibuprofen Lumira-coxib Naproxen

Number of patients in non-aspirin population 6,950 6,968 3,401 3,431 3,549 3,537
Patients with confirmed or probable ischaemic events (%) 34 (0.49) 27 (0.39) 13 (0.38) 12 (0.35) 21 (0.59) 15 (0.42)
All myocardial infections (%) 14 (0.20) 9 (0.13) 4 (0.12) 5 (0.15) 10 (0.28) 4 (0.11)
Clinical (%) 14 (0.20) 5 (0.07) 4 (0.12) 3 (0.09) 10 (0.28) 2 (0.06)
Silent (%) 0 4 (0.06) 0 2 (0.06) 0 2 (0.06)
Ischaemic stroke (%) 12 (0.17) 8 (0.11) 6 (0.18) 2 (0.06) 6 (0.17) 6 (0.17)
Unstable angina (%) 5 (0.07) 5 (0.07) 1 (0.03) 4 (0.12) 4 (0.11) 1 (0.03)
Transient ischaemic attack (%) 3 (0.04) 5 (0.07) 2 (0.06) 1 (0.03) 1 (0.03) 4 (0.11)
Number of patients in aspirin population 2,167 2,159 975 966 1,192 1,193
Patients with confirmed or probable (%) 29 (1.34) 24 (1.11) 9 (0.92) 9 (0.93) 20 (1.68) 15 (1.25)
Ischaemic events
All myocardial infarctions (%) 9 (0.42) 8 (0.37) 1 (0.10) 2 (0.21) 8 (0.67) 6 (0.50)
Clinical (%) 6 (0.28) 7 (0.32) 1 (0.10) 2 (0.21) 5 (0.42) 5 (0.42)
Silent (%) 3 (0.14) 1 (0.05) 0 0 3 (0.25) 1 (0.08)
Ischaemic stroke (%) 11 (0.51) 9 (0.42) 2 (0.21) 4 (0.41) 9 (0.76) 5 (0.42)
Unstable angina (%) 5 (0.23) 6 (0.28) 3 (0.31) 3 (0.31) 2 (0.17) 3 (0.25)
Transient ischaemic attack (%) 4 (0.18) 1 (0.05) 3 (0.31) 0 1 (0.08) 1 (0.08)
From Farkouh et al. (2004)

Table 31 Combined incidence of gastrointestinal and cardiovascular events from lumiracoxib, compared with ibuprofen and naproxen, by sub-
study (safety population (Schnitzer et al. 2004, 2009)
Both substudiesa Number of patients with Hazard ratio (95% CI) P*
events/number at risk (%)

Lumiracoxib 89/9,117 (98%) 0.65 (0.49–0.84) 0.0014


Non-steroidal anti-inflammatory drugs 133/9,127 (1.46%)
Lumiracoxib vs ibuprofen substudyb
Lumiracoxib 30/4,376 (0.69%) 0.50 (0.32–0.79) 0.0025
Ibuprofen 56/4,397 (1.27%)
Lumiracoxib versus naproxen substudyb
Lumiracoxib 59/4,741 (1.24%) 0.75 (0.53–1.05) 0.0961
Naproxen 77/4,730 (1.63%)
Cox proportional-hazards models include, in addition to treatment group, the factors
a
Sub study, low-dose aspirin, and age
b
Low-dose aspirin and age
* Based on Wald v2 statistic for treatment group comparison

Pharmacy Database and found that patients with cardiac which was a randomized trial in which subjects received
disease had been prescribed combinations of ibuprofen and aspirin and NSAIDs and who were at increased risk of
aspirin had an increase in cardiovascular mortality com- cardiovascular events compared with patients who did not
pared with that of patients who had taken aspirin alone. use NSAIDs. The increased risk of NSAIDs causing pos-
This effect was not evident when diclofenac was taken with sibility of adverse events when given with aspirin while
aspirin. dose-dependent was relatively small and required the drugs
About the same time Kurth et al. (2003) published data to be used for long periods. The study by Kimmel et al.
from patients enrolled in the Physicians Health Study (2004) which has already been mentioned was interesting
322 K. D. Rainsford

because this managed to put a completely different slant on Since this was already in a group who had greater that
the whole story. In patients with no history of coronary 98% mean inhibition of thromboxane production this
artery disease the use of aspirin was associated with the could not be regarded as clinically significant (Cryer et al.
lower risk of myocardial infarction as expected but this 2005). These results show that prior treatment for 8 days
benefit was not seen in patients who took any NSAIDs in with aspirin is not affected by subsequent ibuprofen
addition to aspirin. Patients, who had established coronary treatment in terms of platelet thromboxane production. A
disease, who used aspirin with NSAIDs were at similar risk similar study has been performed by Pongbhaesaj et al.
of developing myocardial infarction compared with that of (2003) and published in abstract form which showed
patients who had taken aspirin alone. Thus, there is an almost identical results.
important issue relating to whether patients have coronary However, more recent investigations involving ex vivo
disease or not in the effect of the NSAIDs. It should be production of COX-1 activity, measurement of platelet
noted that the earlier study of Catella-Lawson et al. (2001) aggregation and modelling of data have indicated that
had been undertaken in normal subjects. ibuprofen at a single dose of 400 mg can cause a transient
In a study in elderly patients, who had already experi- interference with the anti-platelet effects of aspirin but that
enced a myocardial infarction the mortality of those who with complete recovery after 6 h (Hong et al. 2008).
had received aspirin and a non-steroidal drug was similar to Thus, it may be concluded that the timing of aspirin and
that of patients who been prescribed alone (Ko et al. 2002; ibuprofen intake may have considerable bearing on the
Curtis et al. 2003; Kean et al. 2008). No apparent differ- interaction of ibuprofen with aspirin on platelets. The
ences were observed in the mortality and analysis of clinical significance of this in terms of the prevention of
patients who had been prescribed aspirin and ibuprofen cardiovascular disease in patients especially those taking
compared with those prescribed aspirin alone (Curtis et al. OTC ibuprofen, who are at risk of developing these con-
2003). ditions clearly is of minor importance when viewed in
A consensus view suggests that the mode of action of context of the study by Kimmel et al. (2004).
ibuprofen in impairing the inhibition by aspirin of platelet Another important aspect arising from these studies is that
function is due to competition between ibuprofen and the ibuprofen itself inhibits platelet aggregation or functions
active site of COX-1 which is irreversibly inhibited by (Brooks et al. 1973; McIntyre et al., 1978; Barclay 2005).
covalent modification by the acetyl group of aspirin at or The mechanisms of the inhibition of platelet aggregation by
near the active site (Curtis and Krumholz 2004; Gaziano ibuprofen are, however, different from those of aspirin. Thus,
and Gibson 2006). Brooks et al. (1973) observed that 4 weeks treatment of male
A pharmacodynamic study by Cryer et al. (2005) volunteers with ibuprofen 1,800 mg day-1 reduced aggre-
investigated the effects of ibuprofen on aspirin-induced gation induced by collagen and ADP but not in re-
thromboxane B2 production, which was intended as a calcificated prior-citrated blood (a thrombin-induced reac-
follow-up to the paper by Catella-Lawson et al. (2001). tion that is inhibited by aspirin). Forty minutes after 7 days
This study was undertaken in 51 volunteers in a double- treatment with ibuprofen platelet aggregation was inhibited
blind randomized parallel placebo-controlled study. The but this returned to normal after 24 h; a situation where it
objections to the Catella-Lawson study were that it did not would normally be expected that aspirin would have pro-
feature a placebo group and there were issues about the duced [90% inhibition of aggregation and prolongation of
study population. The basis of the use of single mea- bleeding time. Moreover, ibuprofen does not cause inhibi-
surement of thromboxane production is that this correlates tion of coagulation in re-calcificated prior-citrated blood or
to a high degree to the inhibition of platelet aggregation prothrombin times (Brooks et al. 1973).
when aspirin is taken. Thromboxane production was Notwithstanding the obviously differing basis of the
measured over 10 days at 1, 3 and 7 days (in the period aspirin–ibuprofen interaction the US FDA pronounced a
prior to randomization to treatment with ibuprofen or warning on the concomitant use of aspirin and ibuprofen
placebo) during 8 days treatment with 81 mg aspirin once in patients, who may be taking aspirin for the preven-
daily in the morning. This resulted in greater than 90% tion of coronary vascular disease (Ellisson et al. 2007).
thromboxane inhibition. On the ninth day and subse- Indeed, the FDA has published on its MedWatch website
quently for 10 days the subjects were randomly assigned (http://www.fda.gov/medwatch/report.htm2007) informa-
to receive ibuprofen or placebo and their thromboxane B2 tion for health care professionals and drug facts concerning
levels were measured on day 0, 1, 3, 7 and 10. In both ibuprofen warning of the concomitant use of ibuprofen and
groups, there was greater than or equal to 98% inhibition aspirin. In the information for healthcare professionals it is
of thromboxane B2 production although there was a small stated that with occasional use of aspirin there is likely to
but clinically in-significant difference between the two be a minimal risk from any attenuation of the anti-platelet
treatment groups of thromboxane inhibition on day 7. effects of low-dose aspirin because of the long-lasting
Ibuprofen 323

effect of aspirin on platelets. Moreover, they state that placebo (Radack et al. 1987). Similarly, increased blood
patients who use immediate release aspirin (not enteric- pressure was noted in a placebo-controlled clinical trial in
coated) and take a single dose of ibuprofen 400 mg should patients receiving hydrochlorthiazide and 1,800 mg day-1
take the dose of ibuprofen at least 30 min or longer after ibuprofen (Gurwitz et al. 1996). However, in a study in stage
the aspirin to avoid attenuation of the effect of aspirin on 1 and 2 hypertensive patients on low and high sodium diets
platelets. They state that recommendations about the tim- receiving the angiotensin-converting enzyme (ACE) inhib-
ing of concomitant use of ibuprofen and enteric-coated itor, enalapril, ibuprofen 1,200 mg day-1 did not affect
low-dose aspirin cannot be made on the base on available systolic or diastolic blood pressure although in a related
data. Thus, on the basis of information of the FDA and the study indomethacin reduced the effects of capropril (Velo
available published literature it is clear that separation of et al. 1987). Other NSAIDs are well-known to interfere with
the dose of aspirin from that of ibuprofen is a practical the actions of ACE inhibitors (Badin et al. 1997). Con-
means of being able to avoid the potential for impairment versely, inhibition of the renin-angiotensin system
of the anti-platelet effect of aspirin by ibuprofen. upregulates COX-2 (Wolf et al. 1999) and thus may exac-
It should be noted that an earlier study in patients with erbate NSAID related renal functions. Calcium channel
rheumatoid arthritis by Grennan et al. (1979) showed that blockers do not appear to be affected by ibuprofen and other
high-dose aspirin (3.6 g day-1) but not a lower dose of NSAIDs in hypertensive patients (Miwa and Jones 1999).
2.4 g day-1 in combination with high- or low-dose ibu-
profen showed that there was a weak clinical additive Congestive heart failure and cardio-renal effects
effect on indices of articular function and pain and this
appeared to be related to an increase in serum ibuprofen by Several studies have indicated that use of NSAIDs in
aspirin but ibuprofen administration did not affect serum patients with a history of heart disease may cause an
salicylate levels. Thus, high doses of aspirin (not those increased risk of congestive heart failure (McGettigan and
usually used for anti-thrombotic effects) may have some Henry 2000). It appears that this effect of NSAID intake
impact on the clinical efficacy of ibuprofen in a positive maybe a class effect and confined to patients that have been
sense but this is related to effects on ibuprofen concen- taking the normal anti-arthritic doses of these drugs. The
tration in the plasma. risk of increased occurrence of coronary heart failure is
overall has an odds ratio of 2.8 but those with a history of
Effects of ibuprofen in hypertension heart disease this may be increased to 10.5. It appears that
plasma half-life of elimination plays a role in the risk of
Elevation of blood pressure is regarded as an indicator or coronary heart failure inasmuch as this risk seems to be
surrogate for CV risk especially in patients that are at risk of doubled in long half-life versus short half-life drugs
CV events. Studies with the coxibs, especially rofecoxib, (McGettigan and Henry 2000).
indicated that they could increase blood pressure and pro- Since inevitably the interference by NSAIDs by pros-
duce oedema in patients with rheumatic conditions (Topol taglandin-dependent processes including haemostasis,
2004, 2005; Kanna et al. 2005; Ostor and Hazleman 2005; vasodilatation, vasoconstrictor balance and renal functions
Rainsford 2005a, b; Antman et al. 2007) NSAIDs, including (including electrolyte balance) influences the potential for
ibuprofen, cause little or no increase in blood pressure in cardiac toxicity via renal effects this class effect with
normotensive individuals (Pope et al. 1993; Johnson et al. NSAIDs usually seen at high doses of NSAIDs with long
1994; Miwa and Jones 1999; Nurmohamed et al. (2002). half-lives maybe a significant feature in the increase in
This has been confirmed in extensive meta-analyses of hypertension and subsequent risk of cardiovascular disease
various clinical trials (Johnson et al. 1994). The issue is, (McGettigan et al. 2000).
however, that NSAIDs interfere with the actions of b- In paediatric studies there have been no records of
blockers (Johnson et al. 1994) and other drugs (Fendrick serious or non-serious CV or cardio-renal effects of ibu-
et al. 2008). These effects are due to the influence of NSA- profen (Ashraf et al. 1999).
IDs, including iburprofen, on production of prostaglandins
and nitric oxide which can affect the actions of anti-hyper- Renal toxicity
tensive agents (Murray and Brater, 1999; Rodriguez et al.
2001; Roig et al. 2002). In a controlled clinical trial in Renal effects of ibuprofen are common to all those syn-
patients with mild to moderate hypertension receiving anti- dromes that are known to be produced by NSAIDs (Dunn
hypertensive medications (b-blockers ? diuretics) 3 weeks et al. 1984; Brater 1998; Breyer 1999; Murray and Brater
treatment with ibuprofen 1,200 mg day-1 caused an 1999; Mounier et al. 2006). The four main primary types of
increase in supine blood pressure by 5.3 mmHg and in sit- renal impairments observed with the NSAIDs include (a)
ting mean arterial pressure of 5.8 mm Hg compared with acute ischaemic renal insufficiency, (b) effects on sodium
324 K. D. Rainsford

potassium and water homeostasis with interference with reactions it is been noted that analgesic nephropathy is not
the effects of diuretics and anti-hypertensive therapy and, been widely recognized or reported effect of renal OTC
(c) acute interstitial nephritis and renal capillary necrosis. ibuprofen (Mann et al. 1993) and certainly this is only
The association of ibuprofen intake with the development infrequently reported in ADR reports made to the UK CSM
of adverse renal effects is probably due to its widespread (Prescott and Martin 1992). In the analysis of risks of renal
use rather than to any particular characteristic of the drug side effects of ibuprofen by Mann et al. (1993), it was
per se since irreversible effects are rare (Murray and Brater found that the renal effects are dose-dependent and these
1999). Renal dysfunction may be more pronounced in effects are almost exclusively in elderly subjects with low
patients that have known risk factors including prior renal intravascular volume and low cardiac output. Furey et al.
disease or impaired renal function (for example changes in (1993) observed that renal-vascular effects of OTC ibu-
creatinine clearance) (Chen et al. 1994; Bennett 1997; profen in elderly patients with mild thiazide-treated
Castellani et al. 1997; Galzin et al. 1997; Schwartz et al. hypertension and renal insufficiency does not appear to be
2002). The issue is of probably greater concern in elderly a risk factor for the development of renal compromise or
subjects because of the higher prevalence of arthritic dis- hypertension even though renal function declines with age.
ease among them and the greater need for NSAID therapy Farquhar et al. 1999; Farquhar and Kenney 1999, have
(Murray and Brater 1999; Kean et al. 2008). Rarely serious shown that OTC dose of ibuprofen (1.2 g day-1) in normal
renal pathology has been observed with ibuprofen subjects subjected to the heat-stress, low-sodium diet or
(Carmichael and Shankel 1985; Radford et al. 1996; Cook dehydration may cause impairment of renal blood flow,
et al. 1997; Silvarajan and Wasse 1997; Brater 1998; glomerular filtration and electrolyte excretion which is
Murray and Brater 1999) but these reactions have not been related to inhibition of renal prostaglandin production.
observed in trials with OTC ibuprofen (Whelton et al. Studies in rabbits suggest that those with pre-existing renal
1990; Whelton 1995). The evidence from literature surveys failure receiving ibuprofen may have alterations in the
and clinical trials suggests OTC use of ibuprofen does not pharmacokinetics of the two enantiomers of the drug (Chen
cause significant renal injury (Rainsford et al. 1997, 2001; et al. 1994) with the clearance of the active (S?) isomer
Doyle et al. 1999; Kellstein et al. 1999; Hersh et al. 2000a; being significantly impaired in this model of renal dys-
Ashraf et al. 2001; Le Parc et al. 2002; Boureau et al. function. Thus, there may be increased prostaglandin
2004). inhibition in the renal tubular systems in individuals with
Griffin et al. (2000) using Tennessee (USA) Medicaid renal impairment.
US Federal State Program Database with patients at greater Overall, these studies suggest that OTC ibuprofen is a
than 65 years of age, undertook an analysis of the effects of low risk factor for developing acute or chronic renal con-
NSAIDs on the development of acute renal failure in these ditions but that as with other NSAIDs there is increasing
elderly patients (see also Kean et al. 2008). Their analysis risk particularly in elderly individuals or those with com-
included consideration for conventional population vari- promised renal function where the drug is taken at high
ables as well as the concomitant intake of prescription prescription anti-arthritic doses (Kean et al. 2008).
drugs and aspirin. In their study they identified 1,799
persons aged greater that 65 years of age with a ‘‘com- Renal effects in children
munity-acquired’’ pre-renal failure or intrinsic renal failure
that required hospitalization for varying periods of time. A number of nephrologists, paediatricians and other phy-
Patients with acute renal failure are different from controls sicians have expressed concerns over the years about the
in a variety of population-related and drug-related factors. risks of serious analgesic-related renal reactions in children
NSAIDs featured in the increasing risk for pre-renal failure (van den Anker 2007).
among those without any underlying renal insufficiency. In the two large-scale practitioner-based studies as part
The odds-ratios were estimated (along with the 95% con- of the Boston University Fever Study, Lesko and Mitchell
fidence intervals) for the risk of association between use of (1995, 1999) noted in particular that there were no cases of
individual NSAIDs and hospitalization for acute renal acute renal failure observed in patients that received either
failure among this population in a case–control compari- ibuprofen (5 or 10 mg kg-1) or paracetamol (12 mg kg-1).
son. Ibuprofen had an odds ratio of 1.63 (1.23–2.08 95% In a subset of 288 of the 795 infants or children that were
CI) and was exceeded only by piroxicam, fenoprofen and hospitalized patients from the first study Lesko and
several other single or multiple-use NSAIDs. The slightly Mitchell (1997) blood urea nitrogen (BUN) and creatinine
higher risk associated with ibuprofen intake may be a levels were evaluated as markers for renal effects of the
reflection of its wide-spread unsupervised use. drugs. Mean BUN levels were *4 mmol L-1 and creati-
In relation to OTC use of ibuprofen and its possible nine levels were *42 mmol L-1 in each of the three drug
association with the development of nephrotoxicity adverse treatment groups. The prevelalence of BUN levels above
Ibuprofen 325

6.4 mmol L-1 and creatinine levels above 62 mmol L-1 group in Barcelona (Spain) attempted to give proportional
was slightly higher in all the hospitalized patients where estimates of the occurrence of different liver reactions
there was evidence of concomitant dehydration. attributed to individual NSAIDs. Tables 33 and 34 sum-
As noted earlier in the discussion of the large-scale marize data from Sanchez-Matienzo et al. Unfortunately,
paediatric study by Ashraf et al. (1999) there were no there are a number of critical issues about this data among
occurrences of renal failure or other serious renal condi- them (a) there are no assessments of the likelihood of the
tions observed with either ibuprofen or paracetamol in the event being associated with intake of a specific drug, (b)
groups totalling 31,144 of younger or older children that there is no information on confounding patient, disease or
were analysed. drug-related factors, (c) there is probably considerable
Six cases of acute renal failure in children have been double counting between the FDA and WHO data, (d) there
reported to be associated with ibuprofen and others with is no information on the intake of drugs in DDD/100,000
NSAIDs (Ulinksi et al. 2004). All patients recovered with patients, and (e) these data are in no sense quantitative and
normalization of serum creatinine levels after 308 days the WHO cautions especially on the use of the data from
following cessation of the drug. what are spontaneous reports. At best, these data only give
Thus, it would appear that although renal effects are signals. Thus, the data only show that ibuprofen has been
known for ibuprofen as with the other NSAIDs there is a reported to produce liver reactions with concomitant use of
low risk of these adverse events occurring in children. No hepatotoxic drugs being implicated in a considerable pro-
doubt dehydration (Leroy et al. 2007) and other factors can portion of cases.
play an important role in the occurrence of renal effects
form all NSAIDs in view of their concentrating in the renal
Table 32 Patterns of hepatopathy observed with various hepatotoxic
tubular systems (Murray and Brater 1999). agents
Drug Exposed cases
Hepatotoxicity
Total Cholestatic Hepatocellular Mixed
Hepatic reactions have been of concern because of serious
Anti-bacterial agents
liver injury being reported with some NSAIDs and coxibs
Clavulanic acid 17 9 3 5
e.g. diclofenac, sulindac (in the USA), celecoxib, lumi-
Isoziazid ? 8 1 7 –
racoxib (O’Brien and Bagby 1985; Stricker 1992; Cameron rifampicin ?
et al. 1996; Tolman 1998; Zimmerman 2000; Lacroix et al. pyrazinamide
2004; Bannwarth and Berenbaum 2005; Chang and Schi- Isoniazid 4 – 3 1
ano 2007) as well as paracetamol even at usual OTC doses Erythromycin 3 – 1 2
(Watkins et al. 2006; Heard et al. 2007). The problem with Analgesics
attributing a particular drug whether it be an NSAID or Paracetamol 17 4 11 2
otherwise, is that there are so many commonly used drugs Musculo-skeletal system
that are hepatotoxic especially those drugs used by rheu- Acetylsalicylic acid 10 2 7 1
matic patients e.g., antibiotiocs, anti-hypertensives, statins Diclofenac 5 1 4 –
etc. (Stricker and 1992; Cameron et al. 1996; Zimmerman Allopurinol 3 2 – 1
2000). Moreover, the pattern of hepatopathies varies con- Droxicam 3 1 1 1
siderably among the different drugs may be taken with Nimesulide 3 – 3 –
ibuprofen (or other NSAIDs) and that are associated with Aceclofenac 2 1 – 1
hepatoxicity (Table 32). Indomethacin 2 – 2 –
To obtain some indication of the incidence of hepatic
Alimentary tract and metabolism
reactions in patients taking NSAIDs Traversa et al. (2003)
Ranitidine 8 4 3 1
investigated the occurrence of hepatotoxicity in a cohort
Glibenclamide 5 3 2 –
study in Umbria (Northern Italy) in subjects that had
Omeprazole 5 1 2 2
recently taken NSAIDs. A total of two events were recor-
Ebrotididne 4 – 4 –
ded as ‘‘all hepatopathies’’ (of 122 cases that had NSAIDs)
Metoclopramide 4 1 3 –
and two with liver injury associated (of 126 cases that had
Cardiovascular system
NSAIDs) with recent use of ibuprofen. Considering the
Ticlopidine 7 7 – –
extensive use of ibuprofen this is a low incidence.
Atenolol 5 1 2 2
In a ‘‘case/non-case’’ compilation of reports extracted
Amiodarone 2 1 1 –
from the FDA and WHO (NIMBUS) databases Sanchez-
Matienzo et al. (2006) of the Pfizer Global Epidemiology From Sabate et al. (2007)
326 K. D. Rainsford

Overall, the data suggest that hepatic reactions are of intolerance to these drugs ranges from severe broncho-
probably rarely associated with ibuprofen. Since there have spasm that is often associated with nasal polyposis, rhino-
been no specific indications of reports of hepatic reactions conjunctivitis, urticaria, cervico-facial erythema, angio-
with OTC use of ibuprofen from trials (Doyle et al. 1999; oedema, hypotension and digestive disturbances (Arnaud
Kellstein et al. 1999; Boureau et al. 2004) or in literature 1995f). These symptoms may occur individually or in any
analyses (Whelton 1995; Rainsford et al. 1997) it is likely combination (Arnaud 1995; de Weck et al. 2006). The
that hepatotoxicity is not a significant risk factor at OTC symptoms may be manifest within a few minutes to several
dosages. hours after ingestion of the drug.
These reactions have been frequently reported with
Hepatic reactions in children aspirin and the terms ‘‘aspirin-associated asthma’’ or
‘‘aspirin-sensitive asthma’’ (ASA) have been employed to
Hepatic reactions do not appear to have been reported in describe the association of symptoms of asthma with
any of the large scale hospitalization practitioner based aspirin (Arnaud 1995; Rainsford 2004a: de Weck et al.
studies in children (Lesko and Mitchell, 1995, 1999; Ashraf 2006; Quiralte et al. 2007). In patients with ASA small
et al., 1999) or in critical reviews of clinical trails doses of aspirin can lead to severe attacks. Frequently,
(Rainsford et al. 1997, 1999, 2001). Hepatitis has been sensitivity to NSAIDs overlaps that with aspirin, as well as
frequently reported in trials of NSAIDs including ibuprofen with ibuprofen (Jenkins et al. 2004). This has given rise to
and aspirin in JRA or JIA (Giannini et al. 1990) but in the the concept of sensitization and the use of desensitizing
small long-term study Ansell (1973) found that liver procedures to treat this condition (Rainsford 2004a, b;
function tests were unaltered in these patients. The risks of Jenkins et al. 2004; de Weck et al. 2006; Quiralte et al.
liver reactions especially in JRA or JIA would appear to be 2007). Paracetamol has been found to be tolerated in some
low except where concomitant hepatotoxic drugs are taken patients with NSAID intolerance but has in recent years
(e.g. paracetamol, methotrexate) (Furst 1992; Hollingworth been reported to be associated with asthma and hypersen-
1993). sivity reactions (Arnaud 1995).
The incidence of NSAID intolerance has been variously
Hypersensitivity reactions and asthma estimated depending on the method of evaluation, study
design and population. Overall the incidence can be 0.6–
NSAIDs are associated with the development of a range of 2.5% of the general population (de Weck et al. 2006). The
hypersensitivity reactions including asthma. The symptoms incidence is 4% when asthmatic patients are interviewed

Table 33 Proportion of reports (PRs) of various hepatic case definitions among cyclooxygenase (COX)-2 selective inhibitors and NSAIDs in the
World Health Organization Uppsala Monitoring Centre data source, updated to the end of quarter 3 of 2003
Drug Overall hepatic Abnormal hepatic Jaundice Hepato-cellular Non-infectious Hepatic Total no. Rank
disorders function damage hepatitis failure of reports

Bromfenac 20.7 10.8 3.2 3.5 4.3 2.2 2,057 14


Celecoxib 2.1 1.3 0.4 0.2 0.5 0.2 17,748 12
Diclofenac 4.7 3.2 1.0 0.2 1.4 0.1 21,082 5
Etodolac 3.6 2.5 1.0 0.4 1.2 0.3 3,553 9
Ibuprofen 1.8 1.1 0.4 0.2 0.5 0.1 32,973 13
Indomethacin 1.8 1.0 0.5 0.1 0.5 0.1 14,576 7
Ketorolac 0.6 0.4 0.1 0.1 0.1 0.2 1,867 6
Meloxicam 0.8 0.4 0.1 0.0 0.4 0.0 3,042 11
Multiple NSAIDsa 5.0 3.1 1.2 0.4 1.4 0.4 33,660 4
Naproxen 1.6 0.8 0.3 0.2 0.3 0.1 13,646 1
Nimesulide 14.4 7.2 2.0 1.0 5.7 0.4 1,057 3
Proxicam 2.0 1.2 0.4 0.1 0.6 0.1 13,973 8
Rofecoxib 1.5 0.8 0.2 0.1 0.4 0.1 20,429 2
Sulindac 9.9 5.2 3.2 0.5 3.1 0.2 5,777 10
From Sanchez-Matienzo et al. (2006). Values are percentages
PRs of concomitant use of other hepatotoxic drugs excluded
a
Includes reports involving [ 1 COX-2 selective inhibitor and/or NSAID
Ibuprofen 327

Table 34 Frequencies of potential confounders in the US Food and et al. 2005; Mascia et al. 2005; Debley et al. 2005;
Drug Administration (under Freedom of Information) (FDA/FOI) and Kanabar 2007; Ponvert and Scheinmann 2007). Two large
World Health Organization Uppsala Monitoring Centre (WHO/UMC)
scale studies in febrile asthmatic children (McIntyre and
data sources
Hull 1996; Lesko et al. 2002) found that ibuprofen far
Drug Concomitant use of Age C65 years from being associated with increased risk of asthma
hepatotoxic drugs
compared with paracetamol actually showed a slightly
FDA/FOI WHO/UMC FDA/FOI WHO/UMC reduced risk. A randomised, double-blind, placebo-con-
Nimesulide 46.8 13.2 27.8 20.3
trolled, crossover bronchoprovocation challenge study in
100 prescreened school-aged children (6–18 years) found
Celecoxib 35.0 17.4 35.3 34.2
that ibuprofen induced bronchospasm was evidence in 2%
Sulindac 31.0 16.7 33.4 33.2
of asthmatic subjects (Debley et al. 2005). Another 2%
Meloxicam 30.1 5.9 38.0 13.5
had clinically relevant decreases in spirometric measure-
Diclofenac 29.4 11.0 34.0 17.6
ments after ibuprofen administration but these did not
Etodolac 28.0 13.5 29.8 21.7
meet the authors a priori criteria for a positive challenge
Indomethacin 27.9 17.1 25.0 18.0
test. These authors considered that ibuprofen-sensitive
Ibuprofen 25.0 18.1 17.6 13.8
asthma has a low prevalence but none the less ibuprofen-
Rofecoxib 20.9 5.8 38.4 21.4
induced bronchospasm should be considered as a risk in
Piroxicam 19.4 7.5 29.4 17.9
childhood asthma. A recent review also concludes that the
Naproxen 18.7 15.3 21.6 16.7
risks of asthma are relatively low in children (Kanabar
Ketorolac 17.6 69.6 22.0 19.1
2007).
Bromfenac 15.5 8.7 8.7 9.1
a
Multiple NSAIDs 48.1 15.0 34.8 22.5
Cutaneous reactions
From Sanchez-Matienzo et al. (2006). Values are in percentages
a
Includes reports involving [1 cyclooxygenase-2 selective inhibitor Minor or ‘‘non-serious’’ skin reactions are among the more
and/or non-selective NSAID frequent reactions observed with NSAIDs including ibu-
profen (Bigby and Stern 1985; O’Brien 1987; Ponvert and
and can range up to 10–29% in adult patients with asthma Scheinmann 2007). The risks of various skin reactions
or those that have been challenged (Arnaud 1995; de Weck occurring with ‘‘ibuprofen containing medications’’ has
et al. 2006). Hypersensivity to NSAIDs usualIy appears in been highlighted by Sanchez-Borges (2005). Their review
the 2nd or 3rd decade and occurs in atopic subjects over the mentioned the different types of skin reactions and the lack
age of 40 years; this being more common in females than of quantitative information on the associations with
males (Arnaud 1995; de Weck et al. 2006). Aspirin or ibuprofen.
NSAID intolerance is occurs infrequently in children (de A case–control study performed in Denmark of the
Weck et al. 2006). Aspirin does not normally involve occurrence of angio-oedema among NSAID and coxib
sensitization through IgE (Arnaud 1995; de Weck et al. users in hospital admissions by Downing et al. (2006)
2006). showed that the relative risks for this condition were higher
The mechanisms of NSAID sensitivity have been in coxib users than those taking traditional NSAIDs. There
debated over the years but there is some consensus that it is were 25 cases out of a total of 377 patients.
due to COX inhibition in susceptible individuals leading to Data of reports on serious and non-serious cutaneous
over production of peptidoleukotries and accompanying reactions for NSAIDs reported in Italy as part of an overall
symptoms of bronchoconstriction and other asthmatic programme of drug surveillance by Naldi et al. (1999) are
symptoms (Arnaud et al. 1995; Rainsford 2005d). Other shown in Fig. 10.
hypotheses suggest that there may be genetic influences These show that ibuprofen ranked in the mid-range of
relating to variability in leukotriene or prostanoid receptors reports. Thus, ibuprofen like other NSAIDs is associated
(de Weck et al. 2006). with the occurrence of skin reactions many of which can be
rated as mild. Serious ADRs in the skin are rare. There
Asthma and hypersensitivity reactions in children have been occasional reports of Stevens-Johnson and
Lyell’s syndromes as well as severe bullous reactions
Asthma and hypersensitivity reactions have long been a (Bigby and Stern 1985; Miwa and Jones 1999; Sanchez
cause for concern in children and the cross-reactions of Borges et al. 2005). However, these serious conditions
ibuprofen with aspirin-sensitive asthma have been high- have not been reported in controlled trials or literature on
lighted by several authors (Body and Potier 2004; Kidon OTC events from ibuprofen (Rainsford et al. 1997, 2001;
328 K. D. Rainsford

Doyle et al. 1999; Kellstein et al., 1999; Hersh et al. 2000a; The possibility of fractures being associated with
Ashraf et al. 2001; Le Parc et al. 2002; Boureau et al. NSAID use has been identified in patients with rheumatoid
2004). and osteoarthritis (Vestergaard et al. 2006). In these epi-
demiological studies adjustments were made for stratifying
Skin reactions in children two cumulative daily dosages (defined daily dose DDD)
and other confounders. There was an odd disease associa-
In the large scale Boston University Fever studies Lesko tion that was observed in these studies in that osteoarthritis
and Mitchell (1995, 1999) did not observe any hospital- was associated with a decreased risk of any fracture and
izations for anaphylaxis. However, three cases of erythema rheumatoid arthritis was associated with increased devel-
multiforme occurred in patients that had received ibuprofen opment of fractures. These studies highlighted that high
and one that had received paracetamol, thus suggesting that dosage intake of aspirin, paracetamol, diclofenac, meloxi-
the risks of these events very low (Lesko and Mitchell, cam and some other NSAIDs but not coxibs was associated
1995). Ashraf et al. (1999) in their large scale trial also with an increased risk of fractures. Ibuprofen showed an
noted there were no cases of Stevens-Johnson syndrome odd inverse dose-related effect in as much as the adjusted
among their patients. odds ratios (less than or 20–74 DDDs approx 1.8–1.82)
were higher than those where the drug was taken in greater
Other adverse reactions quantities (1.42). Thus, hip fractures and other bone
fractures are a risk factor in elderly rheumatic patients
Rare adverse events that have been reported at prescrip- taking NSAIDs for long periods of time maybe more a
tion-level doses with ibuprofen and less frequently with class indication as distinct from a specific risk factor
OTC doses are common to those seen with all NSAIDs. associated (i.e. common to NSAIDs as a group) with
Among these thrombocytopaenia, agranulocytosis, anae- anyone drug.
mia) aseptic meningitis and anaphylactoid reactions, Using the UK general practice data base Van Staa et al.
interactions with the immune, endocrine and metabolic (2000) examined the factor risk exposed to NSAIDs using a
systems, central nervous system and ocular effects (Hoff- case control approach. Regular NSAID intake was associ-
man and Gray 1982; O’Brien and Bagby 1985; Haupt et al. ated with an increase of risk compared with control of 1.47
1991; Miwa and Jones 1999; Jackson et al. 2006; Layton (1.42–1.52 95% CI) of non-vertebral fractures while the
et al. 2006; Neuman and Nicar 2007). Most but not all risk of hip fractures was relatively low being 1.08 (0.9–
these adverse events are rare with the exception of allergies 1.19 95% CI) it appeared from this study that ibuprofen had
including aspirin-sensitive asthma, especially with OTC the lowest risk of non-vertebral fractures as it was used as
dosages of ibuprofen. the reference but there was a larger number of cases of
ibuprofen compared with other NSAIDs probably reflect-
ing a wider-spread use. In contrast to these observations on
the development of fractures studies by Persson et al.
(2005) suggest that long-term treatment of patients who
have undergone revision of hip arthroplasty.
In children some of the abovementioned rare ADRs
have not been reported. During the 1990s there was con-
cern about the possibility of ibuprofen being associated
with the development of necrotizing fasciitis in previously
healthy children who had primary varicella (Khan and Styrt
1997; Neutel and Pless 1997). This was reinforced by a
case–control study that hospitalizations for this condition
had an OR of 11.5 (95% CI 1.4–96.6) (Zerr et al. 1999).
The high range of CI values makes this study of dubious
value for ascertainment of risks.
Lesko et al. (2001) undertook study involving 25 tertiary
care hospitals from across the USA in which a total of 97
Fig. 10 Reporting rates of serious (black bars) and non-serious children were identified with possible group A strepto-
cutaneous reactions to NSAIDs and analgesics compared with reports/ coccal infections with primary varicella infections. They
consumption in DDD’s/1,000 inhabitants day-1 in 4 regions in Italy.
Drug consumption data was derived from pharmacy sales data or
found that the risk of necrotizing fasciitis was not associ-
hospital pharmacies. Numbers of reports for each drug are shown in ated with use of ibuprofen. Moreover, in their large scale
brackets. Reproduced from Naldi et al. (1999), with permission studies Lesko and Mitchell (1995, 1999) and Ashraf et al.
Ibuprofen 329

(1999) have failed to find evidence of ibuprofen being outcomes including deaths (Hawton et al. 2001; Morgan
associated with necrotozing fasciitis. et al. 2007).
Hypothermia is rarely reported to be associated with Recent statistics from the UK National Poisonings
ibuprofen but other NSAIDs are also associated with this Information Service (Anonymous 2006) show that over
AE (Desai and Sriskandan 2003). 115,000 enquiries have been received about paracetamol
annually that comprised 99,000 visits to paracetamol
Cases of poisoning in UK poisoning information on TOXBASE—the NPIS’s online
information database—and about 16,000 telephone calls.
The numbers of reports to the National Poisons Advisory In comparison 42,000 enquiries were received by the NPIS
Service (NPIS; the main referral centre in the UK for en- about ibuprofen and 25,000 about aspirin.
quiries and reports of poisonings) those from ibuprofen are
now second to those of paracetamol (Volans and Fitzpa-
trick 1999). Cases of poisoning from ibuprofen considered General safety assessment
by Volans and Fitzpatrick (1999) were reviewed princi-
pally in the period when the drug was available Assessment of safety in children (0.3–12 year old)
prescription in the UK. It is clear from a large number of
studies (Rainsford 1999c, 2004a) that ibuprofen has the The question of assessing the safety of NSAIDs in the
lowest toxicity of all analgesics. Mechanistically, there is group of 0.3–-12 year old children has been the subject of
no evidence of irreversible toxic actions that could be debate as to whether studies in ‘‘Adults’’ aged [18 years
attributed to the covalent modification of endogenous and the ‘‘Elderly’’ variously aged B65 years can be
biomolecules analogous to that observed in the liver tox- extrapolated to children and is so upon what criteria? It is
icity from quinine–imine metabolite of paracetamol arguable that hormonal and developmental influences can
(Graham and Hicks 2004) or the GI ulceration and bleeding make for marked variations in the enzymes and receptors
arising from irreversible acetylation of platelets and other involved in the expression and control of inflammatory
cyclo-oxygenases or biomolecules from the acetyl-moiety states thus influencing the pharmacodynamics and toxico-
of acetylsalicylic acid (aspirin) (Rainsford et al. 1981, logical potential of NSAIDs. Furthermore, variations in
1984; Rainsford 2004a; Graham et al. 2004). More liver and extra-hepatic metabolism and elimination of
recently, the assessment by Moore (2007) of the risks of NSAIDs could affect their pharmacokinetics in young
serious events from overdose has concluded that ibuprofen adults compared with that in adults. Given these theoretical
is essentially benign. issues it is of relevance to ask how this relates to ibuprofen
having appreciable variations in PK and PD in children
Impact of limitations on analgesic pack sizes in UK [1–2 years (Table 3) c.f. adults (Tables 1, 2) and the
degree to which in the absence of information on these
During the 1990s, there was growing concern in the UK parameters that studies in adults can be extrapolated from
about the toxicity to the liver from paracetamol. This drug the extensive data in adult populations.
is known to cause irreversible liver damage often with fatal First, it is clear that dose-adjustment for age in the
outcome in poisoning. There is also evidence that this drug children and infants is a basic factor which as long as the
may cause severe liver injury in certain conditions in doses recommended dosages of ibuprofen are taken effectively
C4 g day-1. controls drug exposure in this age group. Second, it is clear
As a consequence of these concerns the UK Medicines that the pharmacokinetics of ibuprofen and especially the
and Healthcare Products Authority (MHRA) reviewed the drug metabolizing systems differ little if at all with age in
safety of all analgesics since there have been ongoing children c.f. adults (Tables 1 c.f. 2, 3).
concerns about gastro-intestinal and renal effects of aspirin, From the point of view of risks for children who receive
in particular, and to a lesser extent from ibuprofen. the oral formulation of the drug at what is in general much
A consequence of this was that legislation was intro- lower doses than the prescription level in adults or in
duced in September 1998 to limit the general sales and severe conditions (e.g. arthritis) it is reasonable to conclude
sales by pharmacies of paracetamol and aspirin by that risks from exposure to ibuprofen especially when it is
restricting the pack size (Morgan et al. 2007) Ibuprofen largely given for short periods of time in moderate to low
with or without codeine had already been sold in small doses for acute conditions that the occurrence of serious
pack sizes consistent with its OTC labelling. The limita- ADRs would be relatively low and acceptable from a
tion of pack sizes of paracetamol or aspirin has had a health and safety viewpoint. The evidence of relatively low
significant impact in reducing cases of poisoning, acci- occurrence of serious ADRs or rare events from large scale
dental or deliberate, with a trend in reducing in serious practitioner or centre based studies (Lesko and Mitchell,
330 K. D. Rainsford

Fig. 11 Risks of GI bleed or


heart attack from NSAIDs.
From Moore et al. (2008),
reproduced with permission.
• = GI risks. m = risks of
heart attack. Reproduced on
basis of Creative Commons
Attribution Licence (http://
creativecommons.org/licenses/
by/2.0)

1995, 1999; Lesko et al., 2001, 2002; Ashraf, 1999) and very effective drug for use in a range of painful, acute and
critical reviews of clinical trials (Rainsford et al. 1997, chronic inflammatory conditions.
1999, 2001). The relatively low intrinsic toxicity of the
drug also gives support to the view that it has a high degree
of tolerance from the point of view of human toxicity.
References
Overall safety assessment
Aabakken L, Dybdahl JH, Eidsaunet W, Haaland A, Larsen S, Osnes
Overall, the data reviewed here shows that in comparison M (1989) Optimal assessment of gastrointestinal side effects
with other NSAIDs it appears that ibuprofen has relatively induced by non-steroidal anti-inflammatory drugs. Endoscopic
lesions, faecal blood loss, and symptoms not necessarily
low risks for GI and CV ADRs, especially in the more correlated, as observed after naproxen and oxindanac in healthy
serious category. Some attempts to rate risks of ADRs from volunteers. Scand J Gastroenterol 24:1007–1013
NSAIDs against known life events has recently been con- Aeberhard EE, Henderson SA, Arabolos NS, Griscavage JM, Castro
sidered by Moore et al. (2008). They point out that people FE, Barrett CT, Ignarro LJ (1995) Nonsteroidal anti-inflamma-
tory drugs inhibit expression of the inducible nitric oxide
tend to underestimate common risks and overestimate rare synthase gene. Biochem Biophys Res Commun 208:1053–1059
risks. Also people primarily respond emotionally to risk Aiba T, Tse MM, Lin ET, Koizumi T (1999) Effect of dosage form on
and are risk-averse when confronted with medical inter- stereoisomeric inversion of ibuprofen in volunteers. Biol Pharm
ventions. Taking these and other factors into account these Bull 22:616–622
Akbulut H, Celik I, Ayar A, Vural P, Canbaz M (2005) Ibuprofen
authors have devised a rating of (amongst other ADRs) reduces plasma nitrite/nitrate levels in a rabbit model of
serious GI and CV risks against known life events endotoxin shock. Neuro Endocrinol Lett 26:407–412
(Fig. 11). The risks of these two serious events occurring Albert KS, Gernaat CM (1984) Pharmacokinetics of ibuprofen. Am J
with ibuprofen is low although as the graph shows the Med 77(1A):40–46
Altman RD (1984) Review of ibuprofen for osteoarthritis. Am J Med
points for each ADR with a scattered group it is hard to 77(1A):10–18
distinguish differences among the NSAIDs. It should be Amdekar YK, Desai RZ (1985) Antipyretic activity of ibuprofen and
pointed out that there risks are from the large scale trials paracetamol in children with pyrexia. Br J Clin Pract 39:140–143
with prescription levels NSAIDs performed long-term. American College of Rheumatology, Hotline (2005) The Safety of
COX-2 Inhibitors. Deliberations from the February 16–18, 2005,
Thus, these estimates represent risks at what could be FDA Meeting. www.rheumatology.org/publications/hotline/
regarded as the ‘‘high end’’ of drug exposure. 0305COX2.asp?aud+mem [accessed 16/01/2007]
While there have only been limited investigations into Angst MS, Clark JD, Carvalho B, Tingle M, Schmelz M, Yeomans DC
the effects of application of the suppository formulation in (2008) Cytokine profile in human skin in response to experi-
mental inflammation, noxious stimulation, and administration of
children these and the ADR data show that it is an a COX-inhibitor: a microdialysis study. Pain 139:15–27
acceptable formulation that can be applied to children. Annequin D, Tourniaire B (2005) Migraine and headache in
Further preclinical and clinical investigations, as well as childhood. Arch Pediatr 2:624–629
epidemiological studies should be undertaken to establish Anonymous (2006) Pharmaceutical J 277:321
Ansell BM (1973) Ibuprofen in the management of Still’s disease.
further the safety and efficacy of ibuprofen suppositories. Practitioner 211:659–663
In conclusion, there is substantial evidence of safety and Ansell BM (1983) The medical management of chronic arthritis in
efficacy in support of the view that ibuprofen is a safe and childhood. Ann Acad Med Singapore 12:168–173
Ibuprofen 331

Antman EM, Bennett JS, Daugherty A, Furberg C, Roberts H, ketoprofen, ibuprofen and aspirin on the gastroduodenal mucosa.
Taubert KA, American Heart Association (2007) Use of Eur J Clin Pharmacol 42:685–688
nonsteroidal antiinflammatory drugs: an update for clinicians: a Bernhardt MK, Southard KA, Batterson KD, Logan HL, Baker KA,
scientific statement from the American Heart Association. Jakobsen JR (2001) The effect of preemptive and/or postoper-
Circulation 115:1634–1642 ative ibuprofen therapy for orthodontic pain. Am J Orthod
Arnaud A (1995) Allergy and intolerance to nonsteroidal anti- Dentofacial Orthop 120:20–27
inflammatory agents. Clin Rev Allergy Immunol 13:245–251 Bertin L, Pons G, d’Athis P, Lasfargues G, Maudelonde C, Duhamel
Arora S, Wagner JG, Herbert M (2007) Myth: parenteral ketorolac JF, Olive G (1991) Randomized, double-blind, multicenter,
provides more effective analgesia than oral ibuprofen. CJEM controlled trial of ibuprofen versus acetaminophen (paracetamol)
9:30–32 and placebo for treatment of symptoms of tonsillitis and
Arranz I, Martin-Suarez A, Lanao JM, Mora F, Vazquez C, Escribano pharyngitis in children. J Pediatr 119:811–814
A, Juste M, Mercader J, Ripoll E (2003) Population pharmaco- Bertin L, Pons G, d’Athis P, Duhamel JF, Maudelonde C, Lasfargues
kinetics of high dose ibuprofen in cystic fibrosis. Arch Dis Child G, Guillot M, Marsac A, Debregeas B, Olive G (1996) A
88:1128–1130 randomized, double-blind, multicentre controlled trial of ibu-
Arroyo M, Lanas A (2006) NSAID-induced gastrointestinal damage. profen versus acetaminophen and placebo for symptoms of acute
Minerva Gastroenterol Dietol 52:249–259 otitis media in children. Fundam Clin Pharmacol 10:387–392
Asanuma M, Nishibayashi-Asanuma S, Miyazaki I, Kohno M, Ogawa Bigby M, Stern R (1985) Cutaneous reactions to nonsteroidal anti-
N (2001) Neuroprotective effects of non-steroidal anti-inflam- inflammatory drugs. A review. J Am Acad Dermatol 12:866–876
matory drugs by direct scavenging of nitric oxide radicals. J Bird SE, Williams K, Kula K (2007) Preoperative acetaminophen vs
Neurochem 76:1895–1904 ibuprofen for control of pain after orthodontic separator place-
Ashraf E, Cooper S, Kellstein D, Jayawardena S (2001) Safety profile ment. Am J Orthod Dentofacial Orthop 132(4):504–510
of non-prescription ibuprofen in the elderly osteoarthritic Biskupiak JE, Brixner DI, Howard K, Oderda GM (2006) Gastroin-
patient: a meta-analysis. Inflammopharmacology 9:35–41 testinal complications of over-the-counter nonsteroidal anti-
Ashraf E, Ford L, Geetha R, Cooper S (1999) Safety profile of inflammatory drugs. J Pain Palliat Care Pharmacother 20:7–14
ibuprofen suspension in young children. Inflammopharmacology Bjarnason I (2007) Ibuprofen and gastrointestinal safety: a dose-
7:219–225 duration-dependent phenomena. J R Soc Med 100(Suppl 48):10–13
Autret-Leca E (2003) A general overview of the use of ibuprofen in Bjarnason I, Bissoli F, Conforti A, Maiden L, Moore N, Moretti U,
paediatrics. Int J Clin Pract Suppl (135):9–12 Rainsford KD, Takeuchi K, Velo GP (2005) Adverse reactions
Autret E, Breart G, Jonville AP, Courcier S, Lassale C, Goehrs JM and their mechanisms from nimesulide. In: Rainsford KD (ed)
(1994) Comparative efficacy and tolerance of ibuprofen syrup Nimesulide. Actions and uses. Birkhäuser, Basel, pp 315–415
and acetaminophen syrup in children with pyrexia associated Bjarnason I, Scarpignato C, Takeuchi K, Rainsford KD (2007)
with infectious diseases and treated with antibiotics. Eur J Clin Determinants of the short-term gastric damage caused by
Pharmacol 46:197–201 NSAIDs in man. Aliment Pharmacol Ther 26:95–106
Autret E, Reboul-Marty J, Henry-Launois B, Laborde C, Courcier S, Björkman R, Hallman KM, Hedner J, Hedner T, Henning M (1996)
Goehrs JM, Languillat G, Launois R (1997) Evaluation of Nonsteroidal antiinflammatory drug modulation of behavioral
ibuprofen versus aspirin and paracetamol on efficacy and responses to intrathecal N-methyl-D-aspartate, but not to sub-
comfort in children with fever. Eur J Clin Pharmacol 51:367– stance P and amino-methyl-isoxazole-propionic acid in the rat. J
371 Clin Pharmacol 36(Suppl 12):20S–26S
Averbuch M, Katzper M (2000) A search for sex differences in Blain H, Boileau C, Lapicque F, Nédélec E, Loeuille D, Guillaume C,
response to analgesia. Arch Intern Med 160:3424–3428 Gaucher A, Jeandel C, Netter P, Jouzeau JY (2002) Limitation of
Badin C, Chambrier C, Anouifi A, Boucaud C, Bouletreau P (1997) the in vitro whole blood assay for predicting the COX selectivity
Non-steroidal anti-inflammatory agents and angiotensin convert- of NSAIDs in clinical use. Br J Clin Pharmacol 53:255–265
ing enzyme inhibitor: a dangerous combination during Bliddal H, Rosetzsky A, Schlichting P, Weidner MS, Andersen LA,
postoperative period. Ann Fr Anesth Reanim 16:55–57 Ibfelt HH, Christensen K, Jensen ON, Barslev J (2000) A
Bancos S, Bernard MP, Topham DJ, Phipps RP (2009) Ibuprofen and randomized, placebo-controlled, cross-over study of ginger
other widely used non-steroidal anti-inflammatory drugs inhibit extracts and ibuprofen in osteoarthritis. Osteoarthr Cartil 8:9–12
antibody production in human cells. J Cell Immunol 258:18–28 Blot WJ, McLaughlin JK (2000) Over the counter non-steroidal anti-
Bannwarth B, Berenbaum F (2005) Clinical pharmacology of inflammatory drugs and risk of gastrointestinal bleeding. J
lumiracoxib, a second-generation cyclooxygenase 2 selective Epidemiol Biostat 5:137–142
inhibitor. Expert Opin Investig Drugs 14:521–533 Boardman PL, Nuki G, Hart FD (1967) Ibuprofen in the treatment of
Bannwarth B, Bérenbaum F (2007) Lumiracoxib in the management rheumatoid arthritis and osteo-arthritis. Ann Rheum Dis 26:560–561
of osteoarthritis and acute pain. Expert Opin Pharmacother Body R, Potier K (2004) Best evidence topic report. Non-steroidal
8:1551–1564 anti-inflammatory drugs and exacerbations of asthma in children.
Bannwarth B, Lapicque F, Pehourcq F, Gillet P, Schaeverbeke T, Emerg Med J 21:713–714
Laborde C, Dehais J, Gaucher A, Netter P (1995) Stereoselective Bombardier C, Laine L, Reicin A, Shapiro D, Burgos-Vargas R,
disposition of ibuprofen enantiomers in human cerebrospinal Davis B, Day R, Ferraz MB, Hawkey CJ, Hochberg MC, Kvien
fluid. Br J Clin Pharmacol 40:266–269 TK, Schnitzer TJ, VIGOR Study Group (2000) Comparison of
Barclay L (2005) Platelet function may normalize by 24 hours after upper gastrointestinal toxicity of rofecoxib and naproxen in
last ibuprofen dose. Medscape Medical News. www.med- patients with rheumatoid arthritis. VIGOR study group. N Engl J
scape.com/viewarticle/502566?src=mp [accessed 15/04/2005] Med 343:1520–1528
Beaver W (2003) Review of the analgesic efficacy of ibuprofen. Intl J Boneberg EM, Zou M-H, Ullrich V (1996) Inhibition of cyclooxy-
Clin Pract S135:13–17 genase-1 and -2 by R(-) and S(+)-ibuprofen. J Clin Pharmacol
Bennett WM (1997) Drug interactions and consequences of sodium 36:16S–19S
restriction. Am J Clin Nutr 65:678S–681S Boureau F (1998) Multicentre study of the efficacy of ibuprofen
Bergmann JF, Chassany O, Geneve J, Abiteboul M, Caulin C, compared with paracetamol in throat pain associated with
Segrestaa JM (1992) Enodoscopic evaluation of the effect of tonsillitis. In: Rainsford KD, Powanda MC (eds) Safety and
332 K. D. Rainsford

efficacy of non-prescription (OTC) analgesics and NSAIDs. Carmichael J, Shankel SW (1985) Effects of non-steroidal anti-
Kluwer, Dordrecht, pp 119–121 inflammatory drugs on prostaglandins and renal function. Am J
Boureau F, Schneid H, Zeghari N, Wall R, Bourgeois P (2004) The Med 78:992–1000
IPSO study: ibuprofen, paracetamol study in osteoarthritis. A Castellani S, Ungar A, LaCava G, Cantini C, Stefanile C, Camaiti A,
randomised comparative clinical study comparing the efficacy Messeri G, Coppo M, Vallotti B, DiSerio C, Brocchi A, Masotti
and safety of ibuprofen and paracetamol analgesic treatment G (1997) Renal adaptation to stress: a possible role of endothelin
of osteoarthritis of the knee or hip. Ann Rheum Dis 63:1028– release and prostaglandin modulation in human subjects. J Lab
1034 Clin Med 129:462–469
Bradley JD, Brandt KD, Katz BP, Kalasinski LA, Ryan SI (1991) Castillo M, Lam YW, Dooley MA, Stahl E, Smith PC (1995)
Comparison of an anti-inflammatory dose of ibuprofen, an Disposition and covalent binding of ibuprofen and its acyl
analgesic dose of ibuprofen and acetaminophen in the treatment glucuronide in the elderly. Clin Pharmacol Ther 57:636–644
of patients with osteoarthritis of the knee. N Engl J Med 325:87–91 Catella-Lawson F, Reilly MP, Kapoor SC, Cucchiara AJ, DeMarco S,
Bradley JD, Brandt KD, Katz BP, Kalasinski LA, Ryan SI (1992) Tournier B, Vyas SN, FitzGerald GA (2001) Cyclooxygenase
Treatment of knee osteoarthritis: relationship of clinical features inhibitors and the antiplatelet effects of aspirin. N Engl J Med
of joint inflammation to the response to a nonsteroidal anti- 345:1809–1817
inflammatory drug or pure analgesic. J Rheumatol 19:1950–1954 Cazanave S, Vadrot N, Tinel M, Berson A, Lettéron P, Larosche I,
Bradley JD, Katz BP, Brandt KD (2001) Severity of knee pain does Descatoire V, Feldmann G, Robin MA, Pessayre D (2008)
not predict a better response to an anti-inflammatory dose of Ibuprofen administration attenuates serum TNF-alpha levels,
ibuprofen than to analgesic therapy in patients with osteoarthri- hepatic glutathione depletion, hepatic apoptosis and mouse mor-
tis. J Rheumatol 28:1073–1076 tality after Fas stimulation. Toxicol Appl Pharmacol 231:336–343
Bradley RL, Ellis PE, Thomas P, Bellis H, Ireland AJ, Sandy JR Chan SCH, Shum DKY, Tipoe GL, Mak JCW, Leung ETM, Ip MSM
(2007) A randomized clinical trial comparing the efficacy of (2008) Upregulation of ICAM-1 expression in bronchial epithe-
ibuprofen and paracetamol in the control of orthodontic pain. lial cells by airway secretions in bronchiectasis. Resp Med
Am J Orthod Dentofacial Orthop 132:511–517 102:287–298
Brater DC (1998) Renal safety of ibuprofen: pharmacokinetic aspects. Chang CY, Schiano TD (2007) Review article: drug hepatotoxicity.
In: Rainsford KD, Powanda MC (eds) Safety and efficacy of Aliment Pharmacol Ther 25:1135–1151
non-prescription (OTC) analgesics and NSAIDs. Kluwer, Dordr- Chen CY, Chen CS (1995) Stereoselective disposition of ibuprofen in
echt, pp 73–76 patients with compromised renal haemodynamics. Br J Clin
Breyer M (1999) Editorial. COX2 selective NSAIDs and renal Pharmacol 40:67–72
function: gain without pain? Kidney Int 55:738–739 Chen CY, Pang VF, Chen SS (1994) Assessment of ibuprofen-
Beyth RJ, Shorr RI (1999) Epidemiology of adverse drug reactions in asociated nephrotoxicity in renal dysfunction. J Pharmacol Exp
the elderly by drug class. Drugs Aging 14:231–239 Ther 270:1307
Brocks D, Jamali F (1999) The pharmacokinetics of ibuprofen in Cheng H, Rogers JD, Demetriades JL, Holland SD, Seibold JR,
humans and animals. In: Rainsford KD (ed) Ibuprofen. A critical Depuy E (1994) Pharmacokinetics and bioinversion of ibuprofen
bibliographic review. Taylor & Francis, London, pp 89–142 enantiomers in humans. Pharm Res 11:824–830.
Brooks CD, Schlagel CA, Sekhar NC, Sobota JT (1973) Tolerance Cheruvallath VK, Riley CM, Narayanan SR, Lindenbaum S, Perrin
and pharmacology of ibuprofen. Curr Ther Res Clin Exp JH (1997) A quantitative circular dichroic investigation of the
15:180–190 binding of the enantiomers of ibuprofen and naproxen to human
Brooks P, Emery P, Evans JF, Fenner H, Hawkey CJ, Patrono C, serum albumin. J Pharm Biomed Anal 15:1719–1724
Smolen J, Breedveld F, Day R, Dougados M, Ehrich EW, Gijon- Cheung R, Krishnaswami S, Kowalski K (2007) Analgesic efficacy of
Baños J, Kvien TK, Van Rijswijk MH, Warner T, Zeidler H celecoxib in postoperative oral surgery pain: a single-dose, two-
(1999) Interpreting the clinical significance of the differential center, randomized, double-blind, active- and placebo-controlled
inhibition of cyclooxygenase-1 and cyclooxygenase-2. Rheuma- study. Clin Ther 29(Suppl):2498–2510
tology (Oxford) 38:779–788 Clark E, Plint AC, Correll R, Gaboury I, Passi B (2007) A randomized,
Brown RD, Wilson JT, Kearns GL, Eichler VF, Johnson VA, controlled trial of acetaminophen, ibuprofen, and codeine for acute
Bertrand KM (1992) Single-dose pharmacokinetics of ibuprofen pain relief in children with musculoskeletal trauma. Pediatrics
and acetaminophen in febrile children. J Clin Pharmacol 119:460–467 Erratum in: Pediatrics 119(6):1271
32(3):231–241 Cook ME, Wallin JD, Thakur VD, Kadowitz PJ, McNamara DB,
Brune K, Fenner H, Kurowski M, Lanz R, Members of the SPALA Garcia MM, Lipani JA, Poland M (1997) Comparative effects of
Group (1992) Adverse reactions to NSAIDs: consecutive nabumetone, sulindac, and ibuprofen on renal function. J
evaluation of 30, 000 patients in rheumatology. In: Rainsford Rheumatol 24:1137–1144
KD, Velo GP (eds) Side-effects of anti-inflammatory drugs 3. Costa D, Moutinho L, Lima JLFC, Fernandes E (2006) Antioxidant
Kluwer, Dordrecht, pp 33–42 activity and inhibition of human neutrophil oxidative burst by
Buchanan WW (1990) Implications of NSAID therapy in elderly arylpropionic acid non-steroidal anti-inflammatory drugs. Biol
patients. J Rheumatol 17:29–32 Pharmaceut Bull 29:1659–1670
Buchanan WW, Kean WP (2002) Osteoarthritis: clinical therapeutic Cox SR, Gall EP, Forbes KK, Gresham M, Goris G (1991)
trials and treatment. Inflammopharmacology 10:79–155 Pharmacokinetics of the R(-) and S(+) enantiomers of ibuprofen
Buckler JW, Hall JE, Rees JA, Sheldrake FE, Miller AC (1975) The in the serum and synovial fluid of arthritis patients. J Clin
tolerance and acceptability of ibuprofen (‘Brufen’) in the elderly Pharmacol 31:88–94
patient. Curr Med Res Opin 3:558 Crocetti M, Moghbeli N, Serwint J (2001) Fever phobia revisited:
Burian M, Geisslinger G (2005) COX-dependent mechanisms have parental misconceptions about fever changed in 20 years?
involved in the antinociceptive action of NSAIDs at central Pediatrics 107:1241–1246
and peripheral sites. Pharmacol Therap 107:139–154 Cryer B, Berlin RG, Cooper SA, Hsu C, Wason S (2005) Double-
Cameron RG, Feuer G, de la Iglesia FA (eds) (1996) Drug-induced blind, randomized, parallel, placebo-controlled study of ibupro-
hepatotoxicity. Handbook of experimental pharmacology, vol fen effects on thromboxane B2 concentrations in aspirin-treated
121. Springer, Berlin healthy adult volunteers. Clin Ther 27:185–191
Ibuprofen 333

Curtis JP, Krumholz HM (2004) The case for an adverse interaction rate on pharmacokinetics of ibuprofen in relation to chiral
between aspirin and non-steroidal anti-inflammatory drugs: is it inversion in humans. J Pharm Pharmacol 59:1509–1513
time to believe the hype? J Am Coll Cardiol 43:991–993 Dionne RA (1998) Evaluation and analgesic mechanisms and NSAIDs
Curtis JP, Wang Y, Portnay EL, Masoudi FA, Havranek EP, for acute pain using the oral surgery model. In: Rainsford KD,
Krumholz HM (2003) Aspirin, ibuprofen, and mortality after Powanda MC (eds) Safety and efficacy of non-prescription (OTC)
myocardial infarction: retrospective cohort study. Br Med J analgesics and NSAIDs. Kluwer, Dordrecht, pp 105–117
327:1322–1323 Dionne RA, Cooper SA (1999) Use of ibuprofen in dentistry. In:
Cryer B, Feldman M (1998) Cyclooxygenase-1 and cyclooxygenase-2 Rainsford KD (ed) Ibuprofen. A Critical bibliographic review.
selectivity of widely used nonsteroidal anti-inflammatory drugs. Taylor & Francis, London, pp 409–430
Am J Med 104:413–421 Dionne RA, McCullagh L (1998) Enhanced analgesia and suppression
Cuvellier JC, Joriot S, Auvin S, Vallée L (2005) Traitement of plasma b-endorphin by the S(+)-isomer of ibuprofen. Clin
médicamenteux de l’accès migraineux chez l’enfant. [Pharma- Pharmacol Ther 63:694–701
cologic treatment of acute migraine attack in children]. Arch Długosz CK, Chater RW, Engle JP (2006) Appropriate use of
Pediatr 12(3):316–325 nonprescription analgesics in pediatric patients. J Pediatr Health
Czaykowski D, Fratarcangelo P, Rosefsky J (1994) Evaluation of the Care 20:316–325
antipyretic efficacy of single-dose ibuprofen suspension cam- Dong JQ, Ni L, Scott CS, Retsch-Bogart GZ, Smith PC (2000)
pared to acetaminophen elixir in febrile children. Pediatric Res Pharmacokinetics of ibuprofen enantiomers in children with
35:A141 cystic fibrosis. J Clin Pharmacol 40(8):861–868
Dalton JD Jr, Schweinle JE (2006) Randomized controlled noninfe- Downing A, Jacobsen J, Sorensen HT, McLaughlin JK, Johnsen SP
riority trial to compare extended release acetaminophen and (2006) Risk of hospitalization for angio-oedema among users of
ibuprofen for the treatment of ankle sprains. Ann Emerg Med newer COX-2 selective inhibitors and other nonsteroidal anti-
48:615–623 inflammatory drugs. Br J Clin Pharmacol 62:496–501
Damen L, Bruijn JK, Verhagen AP, Berger MY, Passchier J, Koes Doyle G, Furey S, Berlin R, Cooper S, Jayawardena S, Ashraf E,
BW (2005) Symptomatic treatment of migraine in children: a Baird L (1999) Gastrointestinal safety and tolerance of ibuprofen
systematic review of medication trials. Pediatrics 116:e295–e302 at maximum over-the-counter dose. Aliment Pharmacol Ther
Dawood MY, Khan-Dawood FS (2007) Clinical efficacy and 13:897–906
differential inhibition of menstrual fluid prostaglandin F2alpha Doyle G, Jayawardena S, Ashraf E, Cooper SA (2002) Efficacy and
in a randomized, double-blind, crossover treatment with placebo, tolerability of nonprescription ibuprofen versus celecoxib for
acetaminophen, and ibuprofen in primary dysmenorrhea. Am J dental pain. J Clin Pharmacol 42:912–919
Obstet Gynecol 196:35.e1–35.e5 Dunn MJ, Scharschmidt L, Zambraski E (1984) Mechanisms of
Day RO, Graham GG, Williams KM (1988) Pharmacokinetics of non- nephrotoxicity if non-steroidal anti-inflammatory drugs. Arch
steroidal anti-inflammatory drugs. Baillieres Clin Rheumatol Toxicol 7:328–337
2(2):363–393 Eccles R (2006) Efficacy and safety of over-the-counter analgesics in
Day R, Morrison B, Luza A, Castaneda O, Strusberg A, Nahir M, the treatment of common cold and flu. J Clin Pharm Ther
Helgetveit KB, Kress B, Daniels B, Bolognese J, Krupa D, 31:309–319
Seidenberg B, Ehrich E (2000) A randomized trial of the efficacy Edwards JE, Moore RA, McQuay HJ (2004) Individual patient meta-
and tolerability of the COX-2 inhibitor rofecoxib vs ibuprofen in analysis of single-dose rofecoxib in postoperative pain. BMC
patients with osteoarthritis. Arch Intern Med 160:1781–1787 Anesthesiol 4:1–10
DeArmond B, Francisco CA, Lin JS, Huang FY, Halladay S, Bartziek Eller MG, Wright C, Della-Coletta AA III (1989) Absorption kinetics
RD, Skare KL (1995) Safety profile of over-the-counter of rectally and orally administered ibuprofen. Biopharm Drug
naproxen sodium. Clin Ther 17:587–601 Disp 10:269–278
Debley JS, Carter ER, Gibson RL, Rosenfeld M, Redding GJ (2005) Ellison J, Dager W (2007) Recent FDA warning of the concomitant
The prevalence of ibuprofen-sensitive asthma in children: a use of aspirin and ibuprofen and the effects on platelet
randomized controlled bronchoprovocation challenge study. J aggregation. Prev Cardiol 10:61–63
Pediatr 147:233–238 Eustace N, Hare B (2007) Use of nonsteroidal anti-inflammatory
Deodhar SD, Dick WC, Hodgkinson R, Buchanan WW (1973) drugs in infants. A survey of members of the Association of
Measurement of clinical response to anti-inflammatory drug Paediatric Anaesthetists of Great Britain and Ireland. Paediatr
therapy in rheumatoid arthritis. Q J Med 166:387–401 New Anaesth 17:464–469
Series Evans AM, Nation RL, Sansom LN, Bochner F, Somogyi AA (1990)
Derry C, Derry S, Moore RA, McQuay HJ (2009) Single dose oral The relationship between the pharmacokinetics of ibuprofen
ibuprofen for acutepostoperative pain in adults. Cochrane enantiomers and the dose of racemic ibuprofen in humans.
Database Syst Rev 8(3):CD001548 Biopharm Drug Dispos 11:507–518
Desai PR, Sriskandan S (2003) Hypothermia in a child secondary to Farkouh ME, Kirshner H, Harrington RA, Ruland S, Verheugt FWA,
ibuprofen. Arch Dis Child 88:87–88 Schnitzer TJ, Burmester GR, Mysler E, Hochberg MC, Doherty
de Weck AL, Gamboa PM, Esparza R, Sanz ML (2006) Hypersen- M, Ehrsam E, Gitton X, Krammer G, Mellein B, Gimona A,
sitivity to aspirin and other nonsteroidal anti-inflammatory drugs Matchaba P, Hawkey CJ, Chesebro JH (2004) Comparison of
(NSAIDs). Curr Pharm Des 12:3347–3358 lumiracoxib with naproxen and ibuprofen in the therapeutic
Dieppe PA, Harkness JAL, Higgs ER (1989) Osteoarthritis. In: Wall arthritis research and gastrointestinal event trial (TARGET),
PD, Melzack R (eds) Textbook of Pain, 2nd edn. Churchill- cardiovascular outcomes: randomised controlled trial. Lancet
Livingstone, Edinburgh, pp 306–326 364:675–684
Dı́ez-Domingo J, Planelles MV, Baldo JM, Ballester A, Nunez F, Farkouh ME, Greenberg JD, Jeger RV, Ramanathan K, Verheugt
Jubert A, Domı́nguez Granados R (1998) Ibuprofen prophylaxis FWA, Chesebro JH, Kirshner H, Hochman JS, Lay CL, Mellein
fro adverse reactions to diphtheria-tetanus-pertussis vaccination: B, Matchaba PT, Fuster V, Abramson SB (2009) Cardiovascular
A randomised trial. Curr Ther Res – Clin Expt 59:588–597 outcomes in high risk patients with osteoarthritis treated with
Ding G, Liu Y, Sun J, Takeuchi Y, Toda T, Hayakawa T, Fukushima ibuprofen, naproxen or lumiracoxib. Ann Rheum Dis 66:764–
S, Kishimoto S, Lin W, Inotsume N (2007) Effect of absorption 770
334 K. D. Rainsford

Farquhar WB, Kenney WL (1999) Age and renal prostaglandin Geusens P, Lems W (2008) Efficacy and tolerability of lumiracoxib, a
inhibition during exercise and heat stress. J Appl Physiol highly selective cyclo-oxygenase-2 (COX2) inhibitor, in the
86:1936–1943 management of pain and osteoarthritis. Ther Clin Risk Manag
Farquhar WB, Morgan AL, Zambraski EJ, Kenney WL (1999) Effects 4:337–344
of acetaminophen and ibuprofen on renal function in the stressed Giannini EH, Brewer EJ, Miller ML, Gibbas D, Passo MH, Hoyeraal
kidney. J Appl Physiol 86:598–604 HM, Bernstein B, Person DA, Fink CW (1990) Ibuprofen
Fendrick AM, Pan DE, Johnson GE (2008) OTC analgesics and drug suspension in the treatment of juvenile rheumatoid arthritis.
interactions:clinical implications. Osteopath Med Prim Care 2:2–6 Pediatric rheumatology collaborative study group. J Pediatr
Florence AT, Attwood D (1998) Physicochemical principles in 117:645–652
pharmacy, 3rd edn. Macmillan Press, Houndmills Basingstoke, Gierse JK, Koboldt CM, Walker MC, Seibert K, Isakson PC (1999)
pp 441–446 Kinetic basis for selective inhibition of cyclo-oxygenases.
Fowler CJ, Naidu PS, Lichtman A, Onnis V (2009) The case for the Biochem J 339:607–614
development of novel analgesic agents targeting both fatty acid Glass RC, Swannell AJ (1978) Concentrations of ibuprofen in serum
amide hydrolase and either cyclooxygenase or TRPV1. Br J and synovial fluid from patients with arthritis. Br J Clin
Pharmacol 156:412–419 Pharmacol 6:453P–454P
Freis JF, Williams CA, Bloch DA (1991) The relative toxicity of Glowka FK (2000) Stereoselective pharmacokinetics of ibuprofen and
nonsteroidal antiinflammatory drugs. Arth Rheum 34:1353–1360 its lysinate from suppositories in rabbits. Int J Pharm 199:159–
Friedman H, Seckman C, Lanza F, Royer G, Perry K, Francom S 166
(1990) Clinical pharmacology of predisintegrated ibuprofen Gomez PF, Pillinger MH, Attur M, Marjanovic N, Dave M, Park J,
800 mg tablets: an endoscopic and pharmacokinetic study. J Clin Bingham CO III, Al-Mussair H, Abramson SB (2005) Resolution
Pharmacol 30:57–63 of inflammation: Prostaglandin E2 dissociates nuclear trafficking
Furey SA, Vargas R, McMahon FG (1993) Renovascular effects of of individual NF-jB subunits (p65, p50) in stimulated rheuma-
non-prescription ibuprofen in elderly hypertensive patients with toid synovial fibroblasts. J Immunol 175:6924–6930
mild renal impairment. Pharmacotherapy 13:143–148 Graham GG, Hicks M (2004) Pharmacokinetics and metabolism of
Furst DE (1992) Toxicity of antirheumatic medications in children paracetamol (acetaminophen). In: Rainsford KD (ed) Aspirin
with juvenile arthritis. J Rheumatol Suppl 33:11–15 and related drugs. Taylor & Francis, London, pp 182–213
Gallo JM, Gall EP, Gillespie WR, Albert KS, Perrier D (1986) Graham GG, Williams KM (2004) Metabolism and pharmacokinetics
Ibuprofen kinetics inplasma and synovial fluid of arthritic of ibuprofen. In: Rainsford KD (ed) Aspirin and related drugs.
patients. J Clin Pharmacol 26(1):65–70 Taylor & Francis, London, pp 157–180
Gallego-Sandin S, Novalbos J, Rosado A, Gisbert JP, Galvez-Mugica Graham GG, Roberts MS, Day RO, Rainsford KD (2004) Pharma-
M-A, Garcia AG, Pajares JM, Abad-Santos F (2004) Effect of cokinetics and metabolism of the salicylates. In: Rainsford KD
ibuprofen on cyclooxygenase and nitric oxide synthase of gastric (ed) Aspirin and related drugs. Taylor & Francis, London, pp
mucosa: correlation with endoscopic lesions and adverse reac- 97–155
tions. Dig Dis Sci 49:1538–1544 Graham DJ, Campen D, Hui R, Spence M, Cheetham C, Levy G,
Galzin M, Brunet P, Burtey S, Dussol B, Berland Y (1997) Tubular Shoor S, Ray WA (2005) Risk of acute myocardial infarction
necrosis after non-steroidal anti-inflammatory agents and acute and sudden cardiac death in patients treated with cyclo-
alcoholic intoxication. Nephrologie 18:113–115 oxygenase 2 selective and non-selective non-steroidal anti-
Garcia-Rodriguez LA, Hernandez-Diaz S (2001) The risk of upper inflammatory drugs: nested case-control study. Lancet 365:475–
gastrointestinal complications associated with nonsteroidal anti- 481
inflammatory drugs, glucocorticoids, acetaminophen, and com- Grennan DM, Ferry DG, Ashworth ME, Kenny RE, Mackinnon M
binations of these agents. Arthritis Res 3:98–101 (1979) The aspirin-ibuprofen interaction in rheumatoid arthritis.
Garcia Rodriguez LA, Varas-Lorenzo C, Maguire A, Gonzalez-Perez Br J Clin Pharmacol 8:497–503
A (2004) Nonsteroidal antiinflammatory drugs and the risk of Griffin MR (1998) Epidemiology of nonsteroidal anti-inflammatory
myocardial infarction in the general population. Circulation drug-associated gastrointestinal injury. Am J Med 104(3A):23S–
109:3000–3006 29S
Gaziano JM, Gibson CM (2006) Potential for drug-drug interactions Griffin MR, Yared A, Ray WA (2000) Nonsteroidal anti-inflamma-
in patients taking analgesics for mild-to-moderate pain and low- tory drugs and acute renal failure in elderly persons. Am J
dose aspirin for cardioprotection. Am J Cardiol 97(9A):23–29 Epidemiol 15:488–496
Geisslinger G, Dietzel K, Bezler H, Nuernberg B, Brune K Grimes DA, Hubacher D, Lopez LM, Schulz KF (2006) Non-steroidal
(1989) Therapeutically relevant differences in the pharmacoki- anti-inflammatory drugs for heavy bleeding or pain associated
netical and pharmaceutical behavior of ibuprofen lysinate as with intrauterine-device use. Cochrane Database Syst Rev
compared to ibuprofen acid. Int J Clin Pharmacol Ther Toxicol 18(4):CD006034
27:324–328 Gurwitz JH, Everitt DE, Monane M, Glynn RJ, Choodnovskiy I,
Geisslinger G, Schuster O, Stock KP, Loew D, Bach GL, Brune K Beaudet MP, Avorn J (1996) The impact of ibuprofen on the
(1990) Pharmacokinetics of S(+)- and R(-)-ibuprofen in volun- efficacy of antihypertensive treatment with hydrochlorothiazide
teers and first clinical experience of S(+)-ibuprofen in in elderly persons. J Gerontol A Biol Sci Med Sci 51:M74–M79
rheumatoid arthritis. Eur J Clin Pharmacol 38:493–497 Haas M, Moolenaar F, Meijer DK, Jong PE, de Zeeuw D (1998)
Geisslinger G, Stock KP, Loew D, Bach GL, Brune K (1993) Renal targeting of a non-steroidal anti-inflammatory drug:
Variability in the stereoselective disposition of ibuprofen in effects on renal prostaglandin synthesis in the rat. Clin Sci
patients with rheumatoid arthritis. Br J Clin Pharmacol 35:603– 95:603–609
607 Haase W, Fischer M (1991) Statistical meta-analysis of multicenter
Géraud G, Lantéri-Minet M, Lucas C, Valade D, French Society for clinical studies of ibuprofen with regard to cohort size. Z
the Study of Migraine Headache (SFEMC) (2004) French Rheumatol 50(Suppl 1):77–83
guidelines for the diagnosis and management of migraine in Hage DS, Noctor TA, Wainer IW (1995) Characterization of the
adults and children. Clin Ther. 26:1305–1318 protein binding of chiral drugs by high-performance affinity
Ibuprofen 335

chromatography. Interactions of R- and S-ibuprofen with human Hersh EV, Pinto A, Moore PA (2007) Adverse drug interactions
serum albumin. J Chromatogr A 693:23–32 involving common prescription and over-the-counter analgesic
Halsas M, Hietala J, Veski P, Jürjenson H, Marvola M (1999) Morning agents. Clin Ther 29(Suppl):2477–2497
versus evening dosing of ibuprofen using conventional and time- Higton F (1999) The pharmaceutics of ibuprofen. In: Ibuprofen. A
controlled release formulations. Int J Pharm 189:179–185 critical bibliographic review. Taylor & Francis, London, pp 53–
Hämäläinen ML, Hoppu K, Valkeila E, Santavuori P (1997) 86
Ibuprofen or acetaminophen for the acute treatment of migraine Hinz B, Brune K (2006) Antipyretic analgesics: nonsteroidal anti-
in children: a double-blind, randomized, placebo-controlled, inflammatory drugs, selective COX-2 inhibitors, paracetamol
crossover study. Neurology 48(1):103–107 and pyrazolinones. In: Starke K (ed) Handbook of experimental
Hamman MA, Thompson GA, Hall SD (1997) Regioselective and pharmacology. Springer, Heidelberg, pp 65–93
stereoselective metabolism of ibuprofen by human cytochrome Hippisley-Cox J, Coupland C (2005) Risk of myocardial infarction in
P450 2C. Biochem Pharmacol 54:33–41 patients taking cyclo-oxygenase-2 inhibitors or conventional
Hardy P, Abran D, Hou X, Lahaie I, Peri KG, Asselin P, Varma DR, non-steroidal anti-inflammatory drugs: population based nested
Chemtob S (1998) A major role for prostacyclin in nitric oxide- case-control analysis. Br Med J 330:1366
induced ocular vasorelaxation in the piglet. Circ Res 83:721–729 Hippisley-Cox J, Coupland C, Logan R (2005) Risk of adverse
Hargreaves KM, Keiser K (2002) Development of new pain gastrointestinal outcomes in patients taking cyclo-oxygenase-2
management strategies. J Dent Educ 66:113–121 inhibitors or conventional non-steroidal anti-inflammatory drugs:
Harris RC, McKanna JA, Akai Y, Jacobson HR, Duboois RN, Breyer population based nested case–control analysis. Br Med J
MD (1994) Cyclooxygenase-2 is associated with the macula 331:1310–1316
densa of rat kidney and increases with salt restriction. J Clin Hofbauer R, Kaye AD, Moser D, Gmeiner B, Rasse M, Evans AM
Invest 94:2504–2510 (2000) Frass M (2000) Stereoselective effects of ibuprofen
Haupt MT, Jastrmski MS, Clemmer TP, Metz CA, Goris GB (1991) anantiomers on leukocyte transmigration through endothelial
Effect of ibuprofen in patients with severe sepsis: a randomized, cell monolayers. Inflammopharm 8:326–327
double-blind, multicentre study. The ibuprofen study group. Crit Hoffman M, Gray RG (1982) Ibuprofen-induced meningitis in mixed
Care Med 19:1339–1347 connective tissue disease. Clin Rheumatol 1:128–130
Hawton K, Townsend E, Deeks J, Appleby L, Gunnell D, Bennewith Hollingworth P (1993) The use of non-steroidal anti-inflammatory
O, Cooper J (2001) Effects of legislation restricting pack sizes of drugs in paediatric rheumatic diseases. Br J Rheumatol
paracetamol and salicylate on self poisoning in the United 32(1):73–77
Kingdom: before and after study. Br Med J 322:1203–1207 Holt CI, Hutchins MO, Pileggi R (2005) A real time quantitative PCR
Hay AD, Redmond N, Fletcher M (2006) Antipyretic drugs for analysis and correlation of COX-1 and COX-2 enzymes in
children. BMJ 333:4–5 inflamed dental pulps following administration of three different
Hay AD, Redmond NM, Costelloe C, Montgomery AA, Fletcher M, NSAIDs. J Endod 31:799–804
Hollinghurst S, Peters TJ (2009). Paracetamol and ibuprofen for Hong Y, Gengo FM, Rainka MM, Bates VE, Mager DE (2008)
the treatment of fever in children: the PITCH randomised Population pharmacodynamic modelling of aspirin- and ibupor-
controlled trial. Health Technol Assess. 13(27):iii–iv, ix–x, 1–163 fen-induced inhibition of platelet aggregation in healthy subjects.
Heard K, Green JL, Bailey JE, Bogden GM, Dart RC (2007) A Clin Pharmacokinet 47:129–137
randomized trial to determine the change in alanine aminotrans- Huber MA, Terezhalmi GT (2006) The use of COX-2 inhibitors for
ferase during 10 days of paracetamol (acetaminophen) in acute dental pain. J Am Dent Assoc 137:480–487
subjects who consume moderate amount of alcohol. Aliment Hunt RH, Bowen B, Mortensen ER, Simon TJ, James C, Cagliola A,
Pharmacol Ther 26:283–290 Quan H, Bolognese JA (2000) A randomized trial measuring
Henry D, McGettigan P (2003) Epidemiology overview of gastroin- foecal blood loss after treatment with rofecoxib, ibuprofen, or
testinal and renal toxicity of NSAIDs. Intl J Clin Pract S135:43– placebo in healthy subjects. Am J Med 109:201–206
49 Hynninen VV, Olkkola KT, Leino K, Lundgren S, Neuvonen PJ,
Henry DA, Dobson A, Turner C (1993) Variability in the risk of Rane A, Valtonen M, Vyyryläinen H, Laine K (2006) Effects of
major gastrointestinal complications from non-aspirin nonste- the antifungals voriconazole and fluconazole on the pharmaco-
roidal anti-inflammatory drugs. Gastroenterology 105:1078– kinetics of S-(+)- and R-(-)-Ibuprofen. Antimicrob Agents
1088 Chemother 50:1967–1972
Henry D, Lim LL-Y, Garcia Rodriguez LA, Perez Gutthann S, Carson Ichihara A, Imig JD, Navar LG (1999) Cyclooxygenase-2 modulates
JL, Griffin M, Savage R, Logan R, Moride Y, Hawkey C et al afferent arteriolar responses to increases in pressure. Hyperten-
(1996) Variability in risk of gastrointestinal complications with sion 34:843–847
individual non-steroidal anti-inflammatory drugs: results of a Inoue T, Mi Z, Gillesoie DG, Jackson EK (1998) Cyclooxygenase
collaborative meta-analysis. Br Med J 312:1563–1566 inhibition reveals synergistic action of vasoconstrictors on
Henry D, Drew A, Beuzeville S (1998) Adverse drug reactions in the mesangial cell growth. Eur J Pharmacol 361:285–291
gastrointestinal system attributed to ibuprofen. In: Rainsford KD, Ishiguro C, Fujita T, Omori T, Fujii Y, Mayama T, Sato T (2008)
Powanda MC (eds) Safety and efficacy of non-prescription Assessing the effects of non-steroidal anti-inflammatory drugs
(OTC) analgesics and NSAIDs. Kluwer, Dordrecht, pp 19–45 on antihypertensive drug therapy using post-marketing surveil-
Heremans G, Dehaen F, Rom N, Ramet J, Verboven M, Loeb H lance database. J Epidemiol 18:119–124
(1988) A single-blind parallel group study investigating the Itoh T, Saura Y, Tsuda Y, Yamada H (1997) Stereoselectivity and
antipyretic properties of ibuprofen syrup versus acetylsalicylic enantiomer-enantiomer interactions in the binding of ibuprofen
acid syrup in febrile children. Br J Clin Pract 42:245–247 to human serum albumin. Chirality 9(7):643–649
Hersh EV, Moore PA, Ross GL (2000a) Over-the-counter analgesics Jackson LA, Dunstan M, Starkovich P, Dunn J, Yu O, Nelson JC,
and antipyretics: a critical assessment. Clin Ther 22:500–548 Rees T, Zavitkovsky A (2006) Prophylaxis with acetaminophen
Hersh EV, Levin LM, Cooper SA, Doyle G, Waksman J, Wedell D, or ibuprofen for prevention of local reaction to the fifth
Hong D, Secreto SA (2000b) Ibuprofen liquigel for oral surgery diphtheria-tetanus toxoids-acellular pertussis vaccination: a
pain. Clin Ther 22:1306–1318 randomized, controlled trial. Pediatrics 117:620–625
336 K. D. Rainsford

Jacqz-Aigrain E, Anderson BJ (2006) Pain control: non-steroidal anti- Kean WF, Buchanan WW, Rainsford KD (1999) Therapeutics of
inflammatory agents. Semin Fetal Neonatal Med 11(4):251–259 ibuprofen in rheumatic and other chronic and painful diseases.
Jamali F, Kunz-Dober CM (1999) Pain-mediated altered absorption In: Rainsford KD (ed) Ibuprofen. A critical bibliographic review.
and metabolism of ibuprofen: an explanation for decreases Taylor & Francis, London, pp 279–353
serum enantiomer concentration after dental surgery. Br J Clin Kean WF, Kean R, Buchanan WW (2004) Osteoarthritis: symptoms,
Pharmacol 47:391–396 signs and source of pain. Inflammopharmacology 12:3–31
Jamali F, Singh NN, Pasutto FM, Russell AS, Coutts RT (1988) Kean WF, Rainsford KD, Kean IRL (2008) Management of chronic
Pharmacokinetics of ibuprofen enantiomers in humans following musculoskeletal pain in the elderly: opinions on oral medication
oral administration of tablets with different absorption rates. use. Inflammopharmacology 16:53–75
Pharm Res 5:40–43 Kearns GL (1993) Hepatic drug metabolism in cystic fibrosis: recent
Jamali F, Mehvar R, Russell AS, Sattari S, Yakimets WW, Koo J developments and future directions. Ann Pharmacother
(1992) Human pharmacokinetics of ibuprofen enantiomers 27(1):74–79
following different doses and formulations: intestinal chiral Kearns GL, Reed MD (1989) Clinical pharmacokinetics in infants and
inversion. J Pharm Sci 81:221–225 children. A reappraisal. Clin Pharmacokinet 17(Suppl 1):29–67
Jeffrey P, Tucker GT, Bye A, Crewe HK, Wright PA (1991) The site Kellstein DE, Waksman JA, Furey SA, Binstok G, Cooper SA (1999)
of inversion of R(-)-ibuprofen: studies using rat in situ isolated The safety profile of nonprescription ibuprofen in multiple-dose
perfused intestine/liver preparations. J Pharm Pharmacol use: a meta-analysis. J Clin Pharmacol 39:520–532
43:715–720 Kelley MT, Walson PD, Edge JH, Cox S, Mortensen ME (1992)
Jenkins C, Costello J, Hodge L (2004) Systematic review of Pharmacokinetics and pharmacodynamics of ibuprofen isomers
prevalence of aspirin induced asthma and its implications for and acetaminophen in febrile children. Clin Pharmacol Ther
clinical practice. Br Med J 328:434 52(2):181–189
Jick H, Kaye JA, Russmann S, Jick SS (2006) Nonsteroidal antiinflam- Kepp DR, Sidelmann UG, Hansen SH (1997) Isolation and charac-
matory drugs and acute myocardial infarction in patients with no terization of major phase I and II metabolites of ibuprofen.
major risk factors. Pharmacotherapy 26:1379–1387 Pharm Res 14:676–680
Johnson AG, Nguyen TV, Day RO (1994) Do nonsteroidal anti- Kermond S, Fink M, Graham K, Carlin JB, Barnett P (2002) A
inflammatory drugs affect blood pressure? A meta-analysis. Ann randomized clinical trial: should the child with transient
Intern Med 121:289–300 synovitis of the hip be treated with nonsteroidal anti-inflamma-
Jüni P, Rutjes AWS, Dieppe PA (2002) Are selective COX-2 tory drugs? Ann Emerg Med 40:294–299
inhibitors superior to traditional nonsteroidal anti-inflammatory Khan KN, Venturini CM, Bunch RT, Brassard JA, Koki AT, Morris
drugs. Br Med J 324:1287–1288 DL, Trump BF, Maziasz TJ, Alden CL (1998) Interspecies
Kahn LH, Styrt BA (1997) Necrotizing soft tissue infections reported differences in renal localization of cyclooxygenase isoforms:
with nonsteroidal antiinflammatory drugs. Ann Pharmacother implications in nonsteroidal anti-inflammatory drug-related
31:1034–1039 nephrotoxicity. Toxicol Pathol 26:612–620
Kandoth PW, Joshi MK, Joshi VR, Satoskar RS (1984) Comparative Kidon MI, Kang LW, Chin CW, Hoon LS, See Y, Goh A, Lin JT,
evaluation of antipyretic activity of ibuprofen and aspirin in Chay OM (2005) Early presentation with angioedema and
children with pyrexia of varied aetiology. J Int Med Res 12:292– urticaria in cross-reactive hypersensitivity to nonsteroidal anti-
297 inflammatory drugs among young, Asian, atopic children.
Kanna D, Khanna PP, Durst DE (2005) COX-2 controversy: where Pediatrics 116:e675–e680
are we and where do we go from here? Inflammopharmacology Kimmel SE, Berlin JA, Reilly M, Jaskowiak J, Kishel L, Chittams J,
13:395–402 Strom BL (2004) The effects of nonselective non-aspirin non-
Kanabar D (2007) Ibuprofen and asthma in paediatric populations: steroidal anti-inflammatory medications on the risk of nonfatal
clinical concern or clinical reality? J R Soc Med 100(Suppl myocardial infarction and their interaction with aspirin. J Am
48):15–17 Coll Cardiol 43:985–990
Kargman S, Wong E, Greig GM, Falgueyret J-P, Cromlish W, Ethier Kirchheiner J, Brockmöller J (2005) Clinical consequences of
D, Yergey JA, Riendeau D, Evans JF, Kennedy B, Tagari P, cytochrome P450 2C9 polymorphisms. Clin Pharmacol Ther
Francis DA, O’Neill GP (1996) Mechanism of selective 77:1–16
inhibition of human prostaglandin G/H synthase-1 and -2 in Kirchheiner J, Meineke I, Freytag G, Meisel C, Roots I, Brock-
intact cells. Biochem Pharmacol 52:1113–1125 möller J (2002) Enantiospecific effects of cytochrome P450
Kauffman RE, Nelson MV (1992) Effect of age on ibuprofen 2C9 amino acid variants on ibuprofen pharmacokinetics and on
pharmacokinetics and antipyretic response. J Pediatr the inhibition of cyclooxygenases 1 and 2. Clin Pharmacol Ther
121(6):969–973 72:62–75
Kauffman RE, Sawyer LA, Scheinbaum ML (1992) Antipyretic Klippel JH (ed) (1997) Primer on the rheumatic diseases. The
efficacy of ibuprofen vs acetaminophen. Am J Dis Child Arthritis Foundation, Atlanta, Georgia, pp 316–318
146:622–625 Knights KM, McLean CF, Tonkin AL, Miners JO (1995) Lack of
Kaufman DW, Kelly JP, Sheehan JE, Laszlo A, Wiholm BE, effect of gender and oral contraceptive steroids on the pharma-
Alfredsson L, Koft RS, Shapiro S (1993) Nonsteroidal anti- cokinetics of (R)-ibuprofen in humans. Br J Clin Pharmacol
inflammatory drug use in relation to major gastrointestinal 40:153–161
bleeding. Clin Pharmacol Ther 53:485–494 Ko D, Wang Y, Berger AK, Radford MJ, Krumholz HM (2002)
Kawai S, Nishida S, Kato M, Furumaya Y, Okamoto R, Koshino T, Nonsteroidal anti-inflammatory drugs after acute myocardial
Mizushima Y (1998) Comparison of cyclooxygenase-1 and -2 infarction. Am Heart J 143:475–481
inhibitory activities of various nonsteroidal anti-inflammatory Kokki H, Hendolin H, Maunuksela EL, Vainio J, Nuutinen L (1994)
drugs using human platelets and synovial cells. Eur J Pharmacol Ibuprofen in the treatment of postoperative pain in small
347:87–94 children. A randomized double-blind-placebo controlled parallel
Kean WF, Buchanan WW (1987) Antirheumatic drug therapy in the group study. Acta Anaesthesiol Scand 38:467–472
elderly: a case of failure to identify the correct issues? J Am Kokki H, Kumpulainen E, Lehtonen M, Laisalmi M, Heikkinen M,
Geriatr Soc 35:363–364 Savolainen J, Rautio J (2007) Cerebrospinal fluid distribution of
Ibuprofen 337

ibuprofen after intravenous administration in children. Pediatrics Layton D, Marshall V, Boshier A, Friedmann P, Shakir SA (2006)
120:e1002–e1008 Serious skin reactions and selective COX-2 inhibitors: a case
Konstan MW, Hoppel CL, Chai BL, Davis PB (1991) Ibuprofen in series from prescription-event monitoring in England. Drug Saf
children with cystic fibrosis: pharmacokinetics and adverse 29:687–696
effects. J Pediatr 118(6):956–964 Layton D, Souverein PC, Heerdink ER, Shakir SA, Egberts AC
Konstan MW, Byard PJ, Hoppel CL, Davis PB (1995) Effect of high- (2008) Evaluation of risk profiles for gastrointestinal and
dose ibuprofen in patients with cystic fibrosis. N Engl J Med cardiovascular adverse effects in nonselective NSAID and
332(13):848–854 COX-2 inhibitor users: a cohort study using pharmacy dispens-
Kotob A (1985) A comparative study of two dosage levels of ing data in The Netherlands. Drug Saf 31:143–158
ibuprofen syrup in children with pyrexia. J Int Med Res 13:122– Leroy S, Mosca A, Landre-Peigne C, Cosson MA, Pons G (2007)
126 Ibuprofen in childhood: evidence-based review of efficacy and
Kurth T, Glynn RJ, Walker AM, Chan KA, Buring JE, Hennekens safety. Arch Pediatr 14:477–484
CH, Gaziano JM (2003) Inhibition of clinical benefits of aspirin Lesko SM (2003) The safety of ibuprofen suspension in children. Int J
on first myocardial infarction by nonsteroidal antiinflammatory Clin Pract 50–53, 2003
drugs. Circulation 108:1191–1195 Lesko SM, Mitchell AA (1995) An assessment of the safety of
Kyllönen M, Olkkola KT, Seppälä T, Ryhänen P (2005) Perioperative pediatric ibuprofen. A practitioner-based randomized clinical
pharmacokinetics of ibuprofen enantiomers after rectal admin- trial. JAMA 273:929–933
istration. Paediatr Anaesth 15(7):566–573 Lesko SM, Mitchell AA (1997) Renal function after short-term
Lacroix I, Lapeyre-Mestre M, Bagheri H, Pathak A, Montastruc JL ibuprofen use in infants and children. Pediatrics 100:954–957
(2004) Nonsteroidal anti-inflammatory drug-induced liver Lesko SM, Mitchell AA (1999) The safety of acetaminophen and
injury: a case–control study in primary care. Fundam Clin ibuprofen among children younger than two years old. Pediatrics
Pharmacol 18:201–206 104(4):e39
Lagunas L, Bradbury CM, Laszlo A, Hunt CR, Gius D (2004) Lesko SM, Louik C, Vezina RM, Mitchell AA (2002) Asthma
Indomethacin and ibuprofen induce Hsc70 nuclear localization morbidity after the short-term use of ibuprofen in children.
and activation of the heat shock response in HeLa cells. Biochem Pediatrics 109(2):E20
Biophys Res Commun 313:863–870 Lesko SM, O’Brien KL, Schwartz B, Vezina R, Mitchell AA (2001)
Laine L (2001) Approaches to nonsteroidal anti-inflammatory drug Invasive group A streptococcal infection and nonsteroidal
use in the high-risk patient. Gastroenterology 120:594–606 antiinflammatory drug use among children with primary vari-
Laine L, Smith R, Min K, Chen C, Dubois RW (2006) Systematic cella. Pediatrics 107:1108–1115
review: the lower gastrointestinal adverse effects of non-steroidal Lewis D, Ashwal S, Hershey A, Hirtz D, Yonker M, Silberstein S,
anti-inflammatory drugs. Aliment Pharmacol Ther 24:751–767 American Academy of Neurology Quality Standards Subcom-
Lanas A, Garcia-Rodriguez LA, Arroyo MT, Gomollon F, Feu F, mittee, Practice Committee of the Child Neurology Society
Gonzalez-Perez A, Zapata E, Bastida G, Rodrigo L, Santolaria S, (2004) Practice parameter: pharmacological treatment of
Güell M, de Argila CM, Quintero E, Borda F, Pique JM (2006) migraine headache in children and adolescents: report of the
Risk of upper gastrointestinal ulcer bleeding associated with American Academy of Neurology Quality Standards Subcom-
selective cyclo-oxygenase-2 inhibitors, traditional non-aspirin mittee and the Practice Committee of the Child Neurology
non-steroidal anti-inflammatory drugs, aspirin and combinations. Society. Neurology 63:2215–2224
Gut 55:1731–1738 Lewis JD, Kimmel SE, Localio AR, Metz DC, Farrar JT, Nessel L,
Langman MJS, Weil J, Wainwright P, Lawson DH, Rawlins MD, Brensinger C, McGibney K, Strom BL (2005) Risk of serious
Logan RFA, Murphy M, Vessey MP, Colin-Jones DG (1994) upper gastrointestinal toxicity with over-the-counter nonaspirin
Risk of bleeding peptic ulcers associated with individual non- nonsteroidal anti-inflammatory drugs. Gastroenterology
steroidal anti-inflammatory drugs. Lancet 343:1075–1078 129:1865–1874
Lanza FL, Royer GL, Nelson RS, Chen TT, Seckman CE, Rack MF Le Parc JM, Van Ganse E, Moore N, Wall R, Schneid H, Verrière F
(1979) The effects of ibuprofen, indomethacin, aspirin, (2002) Comparative tolerability of paracetamol, aspirin and
naproxen, and placebo on the gastric mucosa of normal ibuprofen for short-term analgesia in patients with musculoskel-
volunteers. A gastroscopic and photographic study. Dig Dis etal conditions: results in 4291 patients. Clin Rheumatol 21:28–
Sci 24:823–828 31
Lanza FL, Royer GL, Nelson RS, Chen TT, Seckman CE, Rack MF Li G, Treiber G, Maier K, Walker S, Klotz U (1993) Disposition of
(1981) A comparative endoscopic evaluation of the damaging ibuprofen in patients with liver cirrhosis. Stereochemical con-
effects of nonsteroidal anti-inflammatory agents on the gastric siderations. Clin Pharmacokinet 25:154–163
and duodenal mucosa. Am J Gastroenterol 75:17–21 Litalien C, Jacqz-Aigrain E (2001) Risks and benefits of nonsteroidal
Lanza F, Rack MF, Lynn M, Wolf J, Sanda M (1987) An endoscopic anti-inflammatory drugs in children: a comparison with paracet-
comparison of the effects of etodolac, indomethacin, ibuprofen, amol. Paediatr Drugs 3(11):817–858
naproxen, and placebo on the gastrointestinal mucosa. J Rheu- López-Rodriguez R, Novalbos J, Gallego-Sandin S, Román-Martinez
matol 14:338–341 M, Torrado J, Gisbert JP, Abad-Santos F (2008) Inflluence of
Laporte J-R, Ibanez L, Vidal X, Vendrell L, Leone R (2004) Upper CYP2C8 and CYP2C9 polymorphisms on pharmacokinetic and
gastrointestinal bleeding associated with the use of NSAIDs. pharmacodynamic parameters of racemic and enantiomeric form
Newer versus older agents. Drug Saf 27:411–420 of ibuprofen in healthy volunteers. Pharmacol Res 58:77–84
Larson AM, Polson J, Fontana RJ, Davern TJ, Lalani E, Hynan LS, Lugardon S, Lapeyre-Mestre M, Montastruc JL (2004) Upper
Reisch JS, Schiødt FV, Ostapowicz G, Shakil AO, Lee WM, gastrointestinal adverse drug reactions and cyclo-oxygenase-2
Acute Liver Failure Study Group (2005) Acetaminophen- inhibitors (celecoxib and rofecoxib): a case/non-case study from
induced acute liver failure: results of a United States multicenter, the French Pharmacovigilance Database Eur. J Clin Pharmacol
prospective study. Hepatology 42:1364–1372 60:673–677
Laska EM, Sunshine A, Marrero I, Olson N, Siegel C, McCormick N MacDonald TM, Wei L (2006) Is there an interaction between the
(1986) The correlation between blood levels of ibuprofen and cardiovascular protective effects of low-dose aspirin and
clinical analgesic response. Clin Pharmacol Ther 40:1–7 ibuprofen? Basic Clin Pharmacol Toxicol 98:275–280
338 K. D. Rainsford

MacDonald TM, Morant SV, Robinson GC, Shield MJ, McGilchrist ibuprofen, acetaminophen, and placebo in the treatment of
MM, Murray FE, McDevitt DG (1997) Association of upper primary dysmenorrhea: a pooled analysis of five studies. Clin
gastrointestinal toxicity of non-steroidal anti-inflammatory drugs Ther 24:1384–1400
with continued exposure: cohort study. Br Med J 315:1333–1337 Mitchell AA, Lesko SM (1995) When a randomised controlled trial is
Mäkela AL, Yrjänä T, Mattila M (1979) Dosage of salicylates for needed to assess drug safety. The case of paediatric ibuprofen.
children with juvenile rheumatoid arthritis. A prospective Drug Saf 13:15–24
clinical trial with three different preparations of acetylsalicylic Mitchell JA, Akasereenont P, Thiemermann C, Flower RJ, Vane JR
acid. Acta Paediatr Scand 68:423–430 (1994) Selectivity of nonsteroidal anti-inflammatory drugs as
Mäkelä AL, Lempiainen M, Ylijoki H (1981) Ibuprofen levels in inhibitors of constituitive and inducible cyclooxygenase. Proc
serum and synovial fluid. Scand J Rheumatol Suppl 39:15–17 Natl Acad Sci USA 90:11693–11697
Malmberg AB, Yaksh TL (1992) Antinociceptive actions of spinal Miwa LJ, Jones JK (1999) Adverse drug reactions attributed to
nonsteroidal anti-inflammatory agents on the formalin test in the ibuprofen: effects other that gastrointestinal. In: Rainsford KD
rat. J Pharmacol Exp Ther 263:136–146 (ed) Ibuprofen: a critical bibliographic review. Taylor & Francis,
Mamdani M, Rochon PA, Juurlink DN, Kopp A, Anderson GM, London, pp 499–538
Naglie G, Austin PC, Laupacis A (2002) Observational study of Miyamoto A, Hashiguchi Y, Obi T, Ishiguro S, Nishio A (2007)
upper gastrointestinal haemorrhage in elderly patients given Ibuprofen or ozagrel increases NO release and l-nitro arginine
selective cyclo-oxygenase-2 inhibitors or conventional non- induces TXA(2) release from cultured porcine basilar arterial
steroidal anti-inflammatory drugs. Br Med J 325:624–629 endothelial cells. Vascul Pharmacol 46:85–90
Mann JF, Goerig M, Brune K, Luft FC (1993) Ibuprofen as an over- Moore N (2007) Ibuprofen: journey form prescription to over-the-
the-counter drug: is there a risk for renal injury? Clin Nephrol counter use. J R Soc Med 100(Suppl 48):2–6
39:1–6 Moore PA, Acs G, Hargreaves JA (1985) Postextraction pain relief in
Marriott SC, Stephenson TJ, Hull D, Pownall R, Smith CM, Butler A children: a clinical trial of liquid analgesics. Int J Clin Pharmacol
(1991) A dose ranging study of ibuprofen suspension as an Ther Toxicol 23:573–577
antipyretic. Arch Dis Child 66:1037–1041 Moore N, Noblet C, Breemeersch C (1996) Focus on the safety of
Martindale R (2002) The complete drug reference. Pharmaceutical ibuprofen at the analgesic-antipyretic dose. Therapy 51:458–463
Press, London Moore N, Charlesworth A, Van Ganse E, LeParc JM, Jones JK, Wall
Martı́nez RV, Reval ID, Campos MD, Terrón JA, Domı́nguez R, Schneid H, Verrière F (2003) Risk factors for adverse events
Ramirez AM, López-Muňoz FJ (2002) Involvment of peripheral in analgesic drug users: results from the PAIN study. Pharma-
cyclooxygenase-1 and cyclooxygenase-2 in inflammatory pain. J coepidemiol Drug Saf 12:601–610
Pharm Pharmacol 54:405–412 Moore N, Le Parc JM, van Ganse E, Wall R, Schneid H, Cairns R
Mascia K, Haselkorn T, Deniz YM, Miller DP, Bleecker ER, Borish (2002) Tolerability of ibuprofen, aspirin and paracetamol for the
L, TENOR Study Group (2005) Aspirin sensitivity and severity treatment of cold and flu symptoms and sore throat pain. Int J
of asthma: evidence for irreversible airway obstruction in Clin Pract 56:732–734
patients with severe or difficult-to-treat asthma. J Allergy Clin Moore A, Derry S, Makinson GT, McQuay HJ (2005) Tolerability
Immunol 116:970–975 and adverse events in clinical trials of celecoxib in osteoarthritis
Matchaba P, Gitton X, Krammer G, Ehrsam E, Schoor-Sloan V, and rheumatoid arthritis: systematic review and meta-analysis of
Olson M, Mellein B, Hoexter G, Orloff J, Garaud J-J (2005) information from company clinical trials reports. Arthritis Res
Cardiovascular safety of lumiracoxib: A meta-analysis of all Ther 7:R644–R665
randomized controlled trial B1 week and up to one year in Morgan O, Hawkins L, Edwards N, Dargan P (2007) Paracetamol
duration of patients with osteoarthritis and rheumatoid arthritis. (acetaminophen) pack size restrictions and poisoning severity:
Clin Ther 27:1196–1214 time trends in enquiries to a UK poisons centre. J Clin Pharm
McCrory C, Fitzgerald D (2004) Spinal prostaglandin formation and Ther 32:449–455
pain perception following thoracotomy: a role for cyclooxygen- Motola G, Russo F, Mazzeo F, Rinaldi B, Capuano A, Rossi F,
ase-2. Chest 125:1321–1327 Filippelli A (2001) Over-the-counter oral nonsteroidal anti-
McGettigan P, Platona A, Henry DA (2000) Renal and cardiovascular inflammatory drugs: a pharmacoepidemiologic study in southern
toxicity of non-steroidal anti-inflammatory drugs. Inflammo- Italy. Adv Ther 18:216–222
pharmacology 8:1–18 Morse Z, Tump A, Kevelham E (2006) Ibuprofen as a pre-emptive
McIntyre BA, Philp RB, Inwood MJ (1978) Effect of ibuprofen on analgesic is as effective as rofecoxib for mandibular third molar
platelet function in normal subjects and haemophiliac patients. surgery. Odontology 94:59–63
Clin Pharmacol Ther 24:616–621 Moore RA, Derry S, McQuay HJ, Paling J (2008) What do we know
McIntyre J, Hull D (1996) Comparing efficacy and tolerability of about communicating risk? A brief review and suggestion for
ibuprofen and paracetamol in fever. Arch Dis Child 74:164–167 contextualising serious, but rare, risk, and the example of cox-2
McQuay H, Moore A (eds) (1998) An evidence-based resource for selective and non-selective NSAIDs. Arthritis Res Ther 10(1):R20
pain relief. Oxford University Press, Oxford Mounier G, Guy C, Berthoux F, Beynes MN, Ratrema M, Ollagnier
McQuay HJ, Moore RA (2006) Dose–response in direct comparisons M (2006) Severe renal adverse events with arylcarboxylic non-
of different doses of aspirin, ibuprofen and paracetamol (acet- steroidal anti-inflammatory drugs: results on an eight-year
aminophen) in analgesic studies. B J Clin Pharmacol 63:271–278 French national survey. Therapie 61:255–266
Menzel JE, Kolarz G (1997) Modulation of nitric oxide synthase Müller P, Koch EMW, Simon B (1995) Schutzwirkung von Ranitidin
activity by ibuprofen. Inflammation 21:451–461 bei Ibuprofen-Gastroduodenopathie. Arzneim Forsch/Drug Res
Menzel EJ, Burtscher H, Kolarz G (1999) Inhibition of cytokine 45:601–603
production and adhesion molecule expression by ibuprofen is Murray MD, Brater DC (1999) Renal effects of ibuprofen. In:
without effect on transendothelial migration of monocytes. Rainsford KD (ed) Ibuprofen. A critical bibliographic review.
Inflammation 23(3):275–286 Taylor & Francis, London, p 459
Milsom I, Minic M, Dawood MY, Akin MD, Spann J, Niland NF, Myers RP, Li B, Shaheen AA (2007) Emergency department visits for
Squire RA (2002) Comparison of the efficacy and safety of acetaminophen overdose: a Canadian population-based epide-
nonprescription doses of naproxen and naproxen sodium with miologic study (1997–2002). CJEM 9(4):267–274
Ibuprofen 339

Nahata MC, Durrell DE, Powell DA, Gupta N (1991) Pharmacoki- Pope JE, Anderson JJ, Felson DT (1993) Meta-analysis of the effects
netics of ibuprofen in febrile children. Eur J Clin Pharmacol of non-steroidal anti-inflammatory drugs on blood pressure. Arch
40(4):427–428 Intern Med 22:477–484
Nahata MC, Powell DA, Durrell DE, Miller MA, Gupta N (1992) Prescott LF, Martin U (1992) Current status of nephrotoxicity caused
Efficacy of ibuprofen in pediatric patients with fever. Int J Clin by non-steroidal anti-inflammatory drugs. In: Rainsford KD,
Pharmacol Ther Toxicol 30(3):94–96 Velo GP (eds) Side-effects of anti-inflammatory drugs 3.
Naldi L, Conforti A, Venegoni M, Troncon MG, Caputi A, Ghiotto E, Kluwer, Dordrecht
Cocci A, Moretti U, Velo G, Leone R (1999) Cutaneous Psaty BM, Furberg CD (2005) COX-2 inhibitors-lessons in drug
reactions to drugs. An analysis of spontaneous reports in four safety. N Engl J Med 352:1133–1135
Italian regions. Br J Clin Pharmacol 48:839–846 Purcell H (2007) Ibuprofen and cardiovascular safety: where do we
Neuman M, Nicar M (2007) Apoptosis in ibuprofen-induced Steven- stand? J R Soc Med 100(Suppl 48):7–10
Johnson syndrome. Transl Res 149:254–259 Quiding H, Grimstad J, Rusten K, Stubhaug A, Bremnes J, Breivik H
Neupert W, Brugger R, Euchenhofer C, Brune K, Geisslinger G (1992) Ibuprofen plus codeine, ibuprofen, and placebo in a
(1997) Effects of ibuprofen enantiomers and its coenzyme A single- and multidose cross-over comparison for coxarthrosis
thioesters on human prostaglandin endoperoxide synthases. Br J pain. Pain 50:303–307
Pharmacol 122:487–492 Quiralte J, Blanco C, Delgado J, Ortega N, Alcntára M, Castillo R,
Neutel CI, Pless RP (1997) Ibuprofen use in children: a benefit or a Anguita JL, Sáenz de San Pedro B, Carrillo T (2007) Challenge-
risk? Ann Epidemiol 7:437–439 based clinical patterns of 223 Spanish patients with nonsteroidal
Nieman DC, Henson DA, Dumke CL, Oley K, McAnulty SR, Davis anti-inflammatory-drug-induced-reactions. J Investig Allergol
JM, Murphy EA, Utter AC, Lind RH, McAnulty LS, Morrow JD Clin Immunol 17:182–188
(2006) Ibuprofen use, endotoxaemia, inflammation, and plasma Radack KL, Deck CC, Bloomfield SS (1987) Ibuprofen interferes
cytokines during ultramarathon competition. Brain Behav Im- with the efficacy of antihypertensive drugs. A randomized,
mun 20:578–584 double-blind, placebo-controlled trial of ibuprofen compared
Nørholt SE, Aagaard E, Svensson P, Sindet-Pedersen S (1998) with acetaminophen. Ann Intern Med 107:628–635
Evaluation of trismus, bite force, and pressure algometry after Radford MG, Holley KE, Grande JP, Larson TS, Wagoner RD,
third molar surgery: a placebo-controlled study of ibuprofen. J Donadio JV, McCarthy JT (1996) Reversible membranous
Oral Maxillofac Surg 56:420–427 nephropathy associated with the use of nonsteroidal anti-
Novak D, Lewis JH (2003) Drug-induced liver disease. Curr Opin inflammatory drugs. JAMA 276:466–469
Gastroenterol 19:203–215 Rahme E, Nedjar H (2007) Risks and benefits of COX-2 inhibitors vs
Nurmohamed MT, van Halm VP, Dijkmans BA (2002) Cardiovas- non-selective NSAIDs: does their cardiovascular risk exceed
cular risk profile of antirheumatic agents in patients with their gastrointestinal benefit? A retrospective cohort study.
osteoarthritis and rheumatoid arthritis. Drugs 62:1599–1609 Rheumatology 46:435–438
O’Brien WM (1987) Rare adverse reactions to non-steroidal anti- Rainsford KD, Schweitzer A, Brune K (1981) Autoradiographic
inflammatory drugs. In: Rainsford KD, Velo GP (eds) Side-effects and biochemical observations on the distribution of non-
of anti-inflammatory drugs 3. Kluwer, Dordrecht, pp 73–98 steroid anti-inflammatory drugs. Arch Int Pharmacodyn Ther
O’Brien W, Bagby GF (1985) Rare adverse reactions to nonsteroidal 250:180–194
anti-inflammatory drugs. J Rheumatol 12:1–24 Rainsford KD (ed.) (1984) Aspirin and the salicylates. Butterworth,
Ostor AJK, Hazleman BL (2005) The murky waters of the coxibs: a London, Boston
review of the current state of play. Inflammopharmacology Rainsford KD (1999a) History and development of ibuprofen. In:
13:371–380 Rainsford KD (ed) Ibuprofen. A critical bibliographic review.
Owen-Smith BD, Burry HC (1972) Ibuprofen in the management of Taylor & Francis, London, pp 3–24
osteoarthrosis of the hip. Rheumatol Phys Med 11:281–286 Rainsford KD (1999b) Pharmacology and toxicology of ibuprofen. In:
Pachkoria K, Lucena MI, Molokhia M, Cueto R, Carballo AS, Rainsford KD (ed) Ibuprofen. A critical bibliographic review.
Carvajal A, Andrade RJ (2007) Genetic and molecular factors in Taylor & Francis, London, pp 145–275
drug-induced liver injury: a review. Curr Drug Saf 2:97–112 Rainsford KD (1999c) Safety and efficacy of non-prescription (OTC)
Perez C, Sanchez J, Marmol F, Puig-parellada P, Pouplana R (2007) ibuprofen. In: Rainsford, KD (ed) Ibuprofen. A critical biblio-
Reactivity of biologically important NSAID compounds with graphic review. Taylor & Francis, London, pp 357–405
superoxide (O2-) nitric oxide (NO) and cyclooxygenase inhibi- Rainsford KD (2001) The ever-emerging anti-inflammatories. Have
tion. QSAR Comb Sci 26:368–377 there been any real advances? J Physiol (Paris) 95:11–19
Perrott DA, Piira T, Goodenough B, Champion GD (2004) Efficacy Rainsford KD (2003) Discovery, mechanisms of action and safety of
and safety of acetaminophen vs ibuprofen for treating children’s ibuprofen. Intl J Clin Pract S135:3–8
pain or fever: a meta-analysis. Arch Pediatr Adolesc Med Rainsford KD (2004a) Side effects and toxicology of the salicylates.
158:521–526 In: Rainsford KD (ed) Aspirin and related drugs. Taylor &
Peterson GM (2005) Selecting non-prescription analgesics. Am J Francis, London, pp 367–554
Therap 12:67–79 Rainsford KD (2004b) Inhibitors of eicosanoids. In: Curtis-Prior P
Pongbhaesaj P, Yamwong P, Chattanamatta P et al. (2003) Effects of (ed) The eicosanoids. Wiley, Chichester, pp 189–210
ibuprofen and different doses of aspirin on platelet aggregation. Rainsford KD (2005a) Introduction—the coxib controversies. Inflam-
Artheroscl Suppl. 4 : 247 Abstract # 3P-0841 mopharmacology 13:331–341
Ponvert C, Scheinmann P (2007) Les reactions allergiques et Rainsford KD (ed) (2005b) Nimesulide. Actions and uses. Birkhäuser,
pseudoallergiques aux antalgiques, antipyrétics et anti-inflam- Basel
matoires non stéroı̈diens. Allergic and pseudoallergic reactions Rainsford KD (1998) Review of published clinical trial data on the
to antalgics, antipyretics and non-steroidal anti-inflammatory adverse reactions from ibuprofen and paracetamol/acetamino-
drugsAllergic and pseudoallergic reactions to antalgics, antipy- phen at OTC dosages. In: Rainsford KD, Powanda MC (eds)
retics and non-steroidal anti-inflammatory drugs. Archives Safety and Efficacy of Non-Prescription (OTC) Analgesics and
Pédaitr 14:507–512 NSAIDs. Kluwer Academic Publishers, Dordrecht, pp 11–18
340 K. D. Rainsford

Rainsford KD (2007) Anti-inflammatory drugs in the 21st century. In: gastrointestinal events during long-term exposure: a systematic
Harris R (ed) Inflammation in the pathogenesis of chronic diseases. review. MJA 185:501–506
The COX-2 Controversy. Springer, Heidelberg, pp 3–27 Schiff M, Minic M (2004) Comparison of the analgesic efficacy and
Rainsford KD, Roberts SC, Brown S (1997) Ibuprofen and paracet- safety of non-prescription doses of naproxen sodium and
amol: relative safety in non-prescription dosages. J Pharm ibuprofen in the treatment of osteoarthritis of the knee. J
Pharmacol 49:345–376 Rheumatol 31:1373–1383
Rainsford KD, Adesioye J, Dawson S (2001) Relative safety of Schneeweiss S, Solomon DH, Wang PS, Rassen J, Brookhart MA
NSAIDs and analgesics for non-prescription use or in equivalent (2006) Simultaneous assessment of short-term gastrointestinal
doses. Inflammopharmacology 8:351–359 benefits and cardiovascular risks of selective cyclooxygenase 2
Rainsford KD, Kean IRL, Kean WF (2008) Gastro-intestinal inhibitors and nonselective nonsteroidal antiinflammatory drugs:
complications of anti-rheumatic drugs. In: Font J, Ramos-Casals an instrumental variable analysis. Arthritis Rheum 54:3390–3398
M, Rhodes J (eds) Handbook of systemic autoimmune diseases, Schneider HT, Nernberg B, Dietzel K, Brune K (1990) Biliary
vol 8, Chap 18. Elsevier, Amsterdam, pp 243–275 elimination on non-steroidal anti-inflammatory drugs in patients.
Rainsford KD, James C, Johnson DM, Stetsko PI, Hill RE, Salena BJ, Br J Clin Pharmacol 29:127–131
Hunt RH (1993) Effects of chronic NSAIDs on gastric mucosal Schnitzer TJ, Burmester GR, Mysker E, Hochberg MC, Doherty M,
injury related to mucosal prostanoids, and plasma drug concen- Ehrsam E, Gitton X, Krammer G, Meillein B, Matchaba P,
trations in human volunteers. Agents Actions 39 (Special Gimona A, Hawkey CJ (2004) Comparison of lumiracoxib
Issue):C21–C23 with naproxen and ibuprofen in the Therapeutic Arthritis
Re E, Pariente-Khayat A, Gouyet L, Vauzelle-Kervroëdan F, Pons G, Research and Gastrointestinal Event Trial (TARGET), reduc-
D’Athis Ph, Dubois MC, Murat I, Lassale C, Goehrs M, Sainst- tion in ulcer complications: randomised controlled trial. Lancet
Maurice Cl, Olive G (1994) Stereoselective disposition of 364:665–674
ibuprofen enantiomers in infants. Br J Clin Pharmacol 38:378– Schoenfeld P (2001) An evidence-based approach to the gastrointes-
375 tinal safety profile of COX-2 selective anti-inflammatories.
Roth SH, Tindall EA, Jain AK, McMahon FG, April PA, Bockow BI, Gastroent Clin N Am 30:1027–1044
Cohen SB, Fleischmann RM (1993) A controlled study com- Schou S, Nielsen H, Nattestad A, Hillerup S, Ritzau M, Branebjerg
paring the effects of nabumetone, ibuprofen, and ibuprofen plus PE, Bugge C, Skoglund LA (1998) Analgesic dose–response
misoprostol on the upper gastrointestinal tract mucosa. Arch relationship of ibuprofen 50, 100, 200, and 400 mg after surgical
Intern Med 153:2565–2571 removal of third molars: a single-dose, randomized, placebo-
Rodrigues LAG, Jick H (1994) Risk of upper gastrointestinal bleeding controlled, and double-blind study of 304 patients. J Clin
and perforation associated with individual non-steroidal anti- Pharmacol 38:447–454
inflammatory drugs. Lancet 343:769–772 Schwartz JI, Vandormael K, Malice MP, Kalyani RN, Lasseter KC,
Rodriguez F, Llinas MT, Moreno C, Salazar FJ (2001) Role of Holmes GB, Gertz BJ, Gottesdiener KM, Laurenzi M, Redfern
cyclooxygenase-2-derived metabolites and NO in renal response K-J, Brune K (2002) Comparison of rofecoxib, celecoxib, and
to bradykinin. Hypertens 37:129–134 naproxen on renal function in elderly subjects receiving normal-
Roig F, Llinas MT, Lopez R, Salazar FJ (2002) Role of cyclooxy- salt diet. Clin Pharmacol Ther 72:50–61
genase-2 in the prolonged regulation of renal function. Scott CS, Retsch-Bogart GZ, Kustra RP, Graham KM, Glasscock BJ,
Hypertension 40:721–728 Smith PC (1999) The pharmacokinetics of ibuprofen suspension,
Royal College of Paediatrics and Child Health with the Neonatal and chewable tablets, and tablets in children with cystic fibrosis. J
Paediatric Pharmacists Group (2003) Medicines for Children Pediatr 134:58–63
Royer GL, Seckman CE, Schwartz JH, Bennett K (1985) Effects of Seideman P, Lohrer F, Graham GG, Duncan MW, Williams KM, Day
ibuprofen on normal subjects: clinical and routine and special RO (1994) The stereospecific disposition of the enantiomers of
laboratory assessments. Curr Ther Res 37:412–426 ibuprofen in blood, blister and synovial fluid. Br J Clin
Rudy AC, Knight PM, Brater DC, Hall SD (1991) Stereoselective Pharmacol 38:211–227
metabolism of ibuprofen in humans: administration of R-, S- and Seinelä L, Ahvenainen J (2000) Peptic ulcer in the very old patients.
racemic ibuprofen. J Pharmacol Exp Ther 259:1133–1139 Gerontology 46:271–275
Erratum in: J Pharmacol Exp Ther 1992; 260(3):1456 Seymour RA, Hawkesford JE, Weldon M, Brewster D (1991) An
Sabaté M, Ibáñez L, Pérez E, Vidal X, Buti M, Xiol X, Mas A, evaluation of different ibuprofen preparations in the control of
Guarner C, Forné M, Solà R, Castellote J, Rigau J, Laporte JR postoperative pain after third molar surgery. Br J Clin Pharmacol
(2007) Risk of acute liver injury associated with the use of drugs: 31:83–87
a multicentre population survey. Aliment Pharmacol Ther Shah AA, Thjodleifsson B, Murray FE, Kay E, Barry M, Sigthorsson
25(12):1401–1409 G, Gudjonsson H, Oddsson E, Price AB, Fitzgerald DJ,
Sachs CJ (2005) Oral analgesics for acute non-specific pain. Am Bjarnason I (2001) Selective inhibition of COX-2 in humans is
Family Phys 71:913 associated with less gastrointestinal injury: a comparison of
Sánchez-Borges M, Capriles-Hulett A, Caballero-Fonseca F (2005) nimesulide and naproxen. Gut 48:339–346
Risk of skin reactions when using ibuprofen-based medicines. Shirley MA, Guan X, Kaiser DG, Halstead GW, Baillie TA (1994)
Expert Opin Drug Saf 4:837–848 Taurine conjugation of ibuprofen in humans and in rat liver in
Sanchez-Matienzo D, Arana A, Castellsague J, Perez-Gutthann S vitro. Relationship to metabolic chiral inversion. J Pharmacol
(2006) Hepatic disorders in patients treated with COX-2 Exp Ther 269:1166–1175
selective inhibitors or nonselective NSAIDs: a case/noncase Sidler J, Frey B, Baerlocher K (1990) A double-blind comparison of
analysis of spontaneous reports. Clin Ther 28:1123–1132 ibuprofen and paracetamol in juvenile pyrexia. Br J Clin Pract
Sarrell EM, Wielunsky E, Cohen HA (2006) Antipyretic treatment in Suppl 70:22–25
young children with fever: acetaminophen, ibuprofen, or both Sikes DH, Agrawal NM, Zhao WW, Kent JD, Recker DP, Verburg
alternating in a randomized, double-blind study. Arch Pediatr KM (2002) Incidence of gastroduodenal ulcers associated with
Adolesc Med 160(2):197–202 valdecoxib compared with that of ibuprofen and diclofenac in
Schaffer D, Florin T, Eagle C, Marschner I, Singh G, Grobler M, Fenn patients with osteoarthritis. Eur J Gastroenterol Hepatol
C, Schou M, Curnow KM (2006) Risk of serious NSAID-related 14:1101–1111
Ibuprofen 341

Silvarajan M, Wasse L (1997) Perioperative renal failure associated Strom BL, Schinnar R, Bilker WB, Feldman H, Farrar JT, Carson JL
with preoperative intake of ibuprofen. Am Soc Anestisiol (1997) Gastrointestinal tract bleeding associated with naproxen
86:1390–1392 sodium vs ibuprofen. Arch Intern Med 157:2626–2631
Silver S, Gano D, Gerretsen P (2008) Acute treatment of paediatric Suri A, Grundy BL, Derendorf H (1997) Pharmacokinetics and
migraine: a meta-analysis of efficacy. J Paediatr Child Health pharmacodynamics of enantiomers of ibuprofen and flurbiprofen
44:3–9 after oral administration. Int J Clin Pharmacol Ther 35:1–8
Silverstein FE, Faich G, Goldstein JL, Simon LS, Pincus T, Whelton Suthisisang C, Poolsup N, Kittikulsuth W, Pudchakan P, Wiwatpa-
A, Makuch R, Eisen G, Agrawal NM, Stenson WF, Burr AM, nich P (2007) Efficacy of low-dose ibuprofen in acute migraine
Zhao WW, Kent JD, Lefkowith JB, Verburg KM, Geis GS treatment: systematic review and meta-analysis. Ann Pharmac-
(2000) Gastrointestinal toxicity with celecoxib vs nonsteroidal other 41:1782–1791
anti-inflammatory drugs for osteoarthritis and rheumatoid arthri- Suzuki A, Yuen N, Walsh J, Papay J, Hunt CM, Diehl AM (2009) Co-
tis. JAMA 284:1247–1255 medications that modulate liver injury and repair influence
Singer F, Mayrhofer F, Klein G, Hawel R, Kollenz CJ (2000) clinical outcome of acetaminophen-associated liver injury. Clin
Evaluation of the efficacy and dose–response relatioinship of Gastroenterol Hepatol 7:882–888
dexibuprofen (S(+)-ibuprofen) in patients with osteoarthritis of the Tegeder I, Pfeilschifter J, Geisslinger G (2001) Cyclooxygenase-
hip and comparison with racemic ibuprofen using the WOMAC independent actions of cyclooxygenase inhibitors. FASEB J
osteoarthritis index. Int J Clin Pharmacol Ther 38:15–24 15:2057–2072
Singh G (2000) Gastrointesstinal complications of prescription and Teixeira AVasconcelos, Abrunhosa R, Poças L (1977) Observations
over-the-counter nonsteroidal anti-inflammatory drugs: a view on the gastric mucosa of rheumatic patients before and after
from the ARAMIS database. Arthritis, Rheumatism, and Aging ibuprofen administration as studied by the pentagastrin test,
Medical Information System. Am J Ther 7:115–121 endoscopy, and light and electron microscopy. Int J Med Res
Singh G, Ramey DR, Morfeld D, Shi H, Hatoum HT, Fries JF 5:243–252
(1996) Gastrointestinal tract complications of nonsteroidal Thomsen RW, Riis A, Munk EM, Nørgaard M, Christensen S,
anti-inflammatory drug treatment in rheumatoid arthritis. A Sørensen HT (2006) 30-day mortality after peptic ulcer perfo-
prospective observational cohort study. Arch Intern Med ration among users of newer selective COX-2 inhibitors and
156:1530–1536 traditional NSAIDs: a population-based study. Am J Gastroen-
Skeith KJ, Jamali F (1991) Clinical pharmacokinetics of drugs used in terol 101:2704–2710
juvenile arthritis. Clin Pharmacokinet 21:129–149 Tolman KG (1998) Hepatotoxicity of non-narcotic analgesics. Am J
Solomon DH, Glynn RJ, Rothman KJ, Schneeweiss S, Setoguchi S, Med 105:13S–19S
Mogun H, Avorn J, Stürmer T (2008) Subgroup analyses to Topol EJ (2004) Failing the public health—rofecoxib, Merck, and the
determine cardiovascular risk associated with nonsteroidal FDA. N Engl J Med 351:1707–1709
antiinflammatory drugs and coxibs in specific patient groups. Topol EJ (2005) Arthritis medicines and cardiovascular events—
Arthritis Rheum 59:1097–1104 ‘‘house of coxibs’’. J Am Med Assn 293(3):366–368
Sørensen R, Abildstrom SZ, Torp-Pedersen C, Gislason GH (2008) Tornio A, Niemi M, Neuvonen PJ, Backman JT (2007) Stereoselec-
Use of selective cyclooxygenase-2 inhibitors and nonselective tive interaction between the CYP2C8 inhibitor gemfibrozil and
nonsteroidal antiinflammatory drugs in high doses increases racemic ibuprofen. Eur J Clin Pharmacol 63:463–469
mortality and risk of reinfarction in patients with prior myocar- Towheed TE, Maxwell L, Judd MG, Catton M, Hochberg MC, Wells
dial infarction. J Cardiovasc Nurs 23:14–19 G. (2006) Acetaminophen for osteoarthritis. Cochrane Database
Stagnara J, Vermont J, Dürr F, Ferradji K, Mege L, Duquesne A, Syst Rev 25(1):CD004257
Ferley JP, Kassaı̈ B (2005). Parents’ attitudes towards childhood Traversa G, Bianchi C, Da Cas R, Abraha I, Menniti-Ippolito F,
fever. A cross-sectional survey in the Lyon metropolitan area Venegoni M (2003) Cohort study of hepatotoxicity associated
(202 cases). Presse Med 34:1129–1136 with nimesulide and other non-steroidal anti-inflammatory
Steans A, Manners PJ, Robinson IG (1990) A multicentre, long-term drugs. Br Med J 327:18–22
evaluation of the safety and efficacy of ibuprofen syrup in Trocóniz IF, Armenteros S, Planelles MV, Benı́tez J, Calvo R,
children with juvenile chronic arthritis. Br J Clin Pract 44:172– Domı́nguez R (2000) Pharmacokinetic-Pharmacodynamic Mod-
175 elling of the antipyretic effect of two oral formulations of
Steen Law SL, Southard KA, Law AS, Logan HL, Jakobsen JR ibuprofen. Clin Pharmacokinet 38:505–518
(2000) An evaluation of preoperative ibuprofen for treatment of Troullos E, Hargreaves KM, Dionne RA (1997) Ibuprofen elevates
pain associated with orthodontic separator placement. Am J immunoreactive b-endorphin levels in humans during surgical
Orthod Dentofacial Orthop 118:629–635 stress. Clin Pharmacol Ther 62:74–81
Stewart WF, Lipton RB (1998) Ibuprofen plus caffeine in the Ulinski T, Guigonis V, Dunan O, Bensman A (2004) Acute renal
treatement of Migraine. In: Rainsford KD, Powanda MC (eds) failure after treatment with non-steriodal anti-inflammatory
Safety and efficacy of non-prescription (OTC) analgesics and drugs. Eur J Paediatr 163:148–150
NSAIDs. Kluwer Academic Publishers, Dordrecht, pp 123–124 van den Anker JN (2007) Use of ibuprofen in paediatric populations. J
Strand V (2007) Are COX-2 inhibitors preferable to non-selective R Soc Med 100(Suppl 48):18–22
non-steroidal anti-inflammatory drugs in patients with risk of van der Linden MW, van der Bij S, Welsing P, Kuipers EJ, Herings
cardiovascular events taking low-dose aspirin? Lancet RM (2009) The balance between severe cardiovascular and
370:2138–2151 gastrointestinal events among users of selective and non-
Strassburg CP, Strassburg A, Kneip S, Barut A, Tukey RH, Rodeck B, selective non-steroidal anti-inflammatory drugs. Ann Rheum
Manns MP (2002) Developmental aspects of human hepatic drug Dis 68:668–673
glucuronidation in young adults and children. Gut 50:259–265 Van Esch A, Van Steensel-Moll HA, Steyerberg EW, Offringa M,
Stratman NC, Carter DB, Sethy VH (1997) Ibuprofen: effect on Habbema JD, Derksen-Lubsen G (1995) Antipyretic efficacy of
inducible nitric oxide synthase. Brain Res Mol Brain Res ibuprofen and acetaminophen in children with febrile seizures.
50:107–112 Arch Pediatr Adolesc Med 149:632–637
Stricker BH Ch. (1992) Drug-induced hepatic injury. Elsevier, Van Hoogdalem EJ, de Boer AG, Breimer DD (1991) Pharmacoki-
Amsterdam netics of rectal drug administration, Part II. Clinical
342 K. D. Rainsford

appplications of peripherally acting drugs, and conclusions. Clin Walson PD, Galletta G, Chomilo F, Braden NJ, Sawyer LA,
Pharmacokinet 21:110–128 Scheinbaum ML (1992) Comparison of multidose ibuprofen
Van Staa TP, Leufkens HGM, Cooper C (2000) Use of non-steroidal and acetaminophen therapy in febrile children. Am J Dis Child
anti-inflammatory drugs and risk of fractures. Bone 27:563–568 146(5):626–632
van Stuijvenberg M, Derksen-Lubsen G, Steyerberg EW, Habbema Wanwimolruk S, Brooks PM, Birkett DJ (1983) Protein binding of
JD, Moll HA (1998a) Randomized, controlled trial of ibuprofen non-steroidal anti-inflammatory drugs in plasma and synovial
syrup administered during febrile illnesses to prevent febrile fluid of arthritic patients. Br J Clin Pharmacol 15:91–94
seizure recurrences. Pediatrics 102(5):E51 Wan Po A (2006) Editorial commentary. Dose–response of minor
van Stuijvenberg M, Steyerberg EW, Derksen-Lubsen G, Moll HA analgesics in acute pain. B J Clin Pharmacol 63:268–270
(1998b) Temperature, age, and recurrence of febrile seizure. Wagener HH, Vögtle-Junkert U (1996) Intrasubject variability in
Arch Pediatr Adolesc Med 152(12):1170–1175 bioequivalence studies illustrated by the example of ibuprofen.
van Stuijvenberg M, de Vos S, Tjiang GC, Steyerberg EW, Derksen- Int J Clin Pharmacol Ther 34:21–31
Lubsen G, Moll HA (1999) Parents’ fear regarding fever and Warner TD, Giuliano F, Vojnovic I, Bukasa A, Mitchell JA, Vane JR
febrile seizures. Acta Paediatr 88(6):618–622 (1999) Nonsteroid drug selectivities for cyclo-oxygenase-1
Vanderhoeven SJ, Lindon JC, Troke J, Nicholson JK, Wilson ID rather than cyclo-oxygenase-2 are associated with human
(2006) NMR spectroscopic studies of the transacylation reactiv- gastrointestinal toxicity: a full in vitro analysis. Proc Natl Acad
ity of ibuprofen 1-beta-)-acyl-glucuronide. J Pharm Biomed Sci USA 96:7563–7568
Anal 41:1002–1006 Warrington SJ, Halsey A, O’Donnell L (1982) A comparison of
Vandivier RW, Eidsath A, Banks SM, Preas HL III, Leighton SB, gastrointestinal bleeding in healthy volunteers treated with
Godin PJ, Suffredini AF, Danner RL (1999) Down-regulation of tiaprofenic acid, aspirin or ibuprofen. Rheumatology 7:107–110
nitric oxide production by ibuprofen in human volunteers. J Watkins PB, Kaplowitz N, Slattery JT, Colonese CR, Colucci SV,
Pharm Exptl Ther 289:1398–1403 Stewart PW, Harris SC (2006) Aminotransferase elevations in
Vauzelle-Kervroëdan F, d’Athis P, Pariente-Khayat A, Debregeas S, healthy adults receiving 4 grams of acetaminophen daily: a
Olive G, Pons G (1997) Equivalent antipyretic activity of ibuprofen randomized controlled trial. J Am Med Assn 296:87–93
and paracetamol in febrile children. J Pediatr 131:683–687 Whelton A (1995) Renal effects of over-the-counter analgesics. J Clin
Vane JR, Botting RM (Eds) (2001) Therapeutic roles of selective Pharmacol 35:454–463
COX-2 inhibitors. William Harvey Press, London Whelton A, Stout RL, Spilman PS, Klassen DK (1990) Renal effects
Verhagen AP, Damen L, Berger MY, Passchier J, Merlijn V, Koes of ibuprofen, piroxicam, and sullindac in patients with asymp-
BW (2006) Is any one analgesic superior for episodic tension- tomatic renal failure. A prospective, randomized, crossover
type headache? J Fam Pract 55:1064–1072 comparison. Ann Intern Med 113:481–482
Vestergaard P, Rejnmark L, Mosekilde L (2006) Fracture risk White WB (2007) Cardiovascular effects of the selective cyclooxy-
associated with use of nonsteroidal anti-inflammatory drugs, genase-2 inhibitors. Subcell Biochem 42:145–158
acetylsalicylic acid, and acetaminophen and the effects of White WB (2008) Defining the problem of treating the patient with
rheumatoid arthritis and osteoarthritis. Calcif Tissue Int 79:84–94 hypertension and arthritis pain. Am J Med 122(5 Suppl):S3–
Velo GP, Minuz P, Arosio E, Capuzzo MG, Covi G, Lechi A (1987) S9
Interaction between non-steroidal anti-inflammatory drugs and Wilson JT, Brown RD, Kearns GL, Eichler VF, Johnson VA,
angiotensin-converting enzyme inhibitors in man. In: Rainsford Bertrand KM, Lowe BA (1991) Single-dose, placebo-controlled
KD, Velo GP (eds) Side-Effects of Anti-inflammatory Drugs. comparative study of ibuprofen and acetaminophen antipyresis
Part 1. Clinical and Epidemiological Aspects. MTP Press, in children. J Pediatr 119:803–811
Lancaster (UK), pp 195–201 Williams K, Day R, Knihinicki R, Duffield A (1986) The stereose-
Vij N, Amoako MO, Mazur S, Zeitlin PL (2008) CHOP transcription lective uptake of ibuprofen enantiomers into adipose tissue.
factor mediates IL-8 signaling in cystic fibrosis bronchial Biochem Pharmacol 35(19):3403–3405
epithelial cells. Am J Respir Cell Mol Biol 38:176–184 Winter B, Mygynd N (2003) Potential benefits of ibuprofen in the
Villanueva M, Heckenberger R, Strobach H, Palmer M, Schrör K treatment of viral respiratory infections. Inflammopharmacol
(1993) Equipotent inhibition by R(-)-, S(+)- and racemic 11:445–453
ibuprofen of human polymorphonuclear cell function in vitro. Wolf K, Castrop H, Hartner A, Goppelt-Strübe M, Hilgers K, Kurtz A
Br J Pharmacol 35:235–242 (1999) Inhibition of the rennin-angiotensin system upregulates
Volans G, Fitzpatrick R (1999) Human toxicity of ibuprofen. In: cyclooxygenase-2 expression in the macula densa. Hypertension
Rainsford KD (ed) Ibuprofen. A critical bibliographic review. 34:503–507
Taylor & Francis, London, pp 541–559 Wolfe MM, Lichtenstein DR, Singh G (1999) Gastrointestinal
Voutilainen M, Sokka T, Juhola M, Färkkilä M, Hannonen P (1998) toxicity of nonsteroidal anti-inflammatory drugs. N Engl J Med
Scand J Gastroenterol 33:811–816 340:1888–1899
Vural P, Canbaz M, Akbulut A, Celik I (2002) Effects of ibuprofen on Zerr DM, Alexander ER, Duchin JS, Koutsky LA, Rubens CE (1999)
plasma endothelin levels and some vital parameters during A case-control study of necrotizing fasciitis during primary
endotoxin shock in rabbits. Clin Chim Acta 316:101–108 varicella. Pediatrics 103:783–790
Walker JS, Carmody JJ (1998) Experimental pain in healthy human Zhang DH, Marconi A, Xu ML, Yang CX, Sun GW, Feng XL, Ling
subjects: gender differences in nociception and in response to CQ, Qin WZ, Uzan G, d’Alessio P (2006) Tripterine inhibits the
ibuprofen. Anesth Analg 86:1257–1262 expression of adhesion molecules in activated endothelial cells. J
Walson PD, Mortensen ME (1989) Pharmacokinetics of common Leukoc Biol 80:309–319
analgesics, anti-inflammatories and antipyretics in children. Clin Zimmerman HJ (2000) Hepatotoxicity. The adverse effects of drugs
Pharmacokinet 17(Suppl 1):116–137 and other chemicals on the liver, 2nd edn. Lippincott, Williams
Walson PD, Galletta G, Braden NJ, Alexander L (1989) Ibuprofen, & Wilkins, Philadelphia
acetaminophen, and placebo treatment of febrile children. Clin
Pharmacol Ther 46:9–17

Você também pode gostar