You are on page 1of 18

Review

European Journal of Preventive


Cardiology

Non-steroidal anti-inflammatory 2020, Vol. 27(8) 850–867


! The European Society of
Cardiology 2019
drug effects on renal and Article reuse guidelines:
sagepub.com/journals-permissions
cardiovascular function: from DOI: 10.1177/2047487319848105
journals.sagepub.com/home/cpr
physiology to clinical practice

Aderville Cabassi1, Stefano Tedeschi1,2, Stefano Perlini3,


Ignazio Verzicco1, Riccardo Volpi1, Gianluca Gonzi4 and

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


Stefano Del Canale5

Abstract
Excessive or inappropriate use of non-steroidal anti-inflammatory drugs can affect cardiovascular and renal function.
Non-steroidal anti-inflammatory drugs, both non-selective and selective cyclooxygenase 2 inhibitors, are among the most
widely used drugs, especially in the elderly, with multiple comorbidities. Exposition to a polypharmacy burden represents
a favourable substrate for the onset of non-steroidal anti-inflammatory drug-induced deleterious effects. Cardiovascular
and renal issues concerning the occurrence of myocardial infarction, atrial fibrillation, heart failure and arterial hyper-
tension, as well as acute or chronic kidney damage, become critical for clinicians in their daily practice. We discuss
current available knowledge regarding prostanoid physiology in vascular, cardiac and renal systems, pointing out potential
negative non-steroidal anti-inflammatory drug-related issues in clinical practice.

Keywords
Non-steroidal anti-inflammatory drugs, acute and chronic renal disease, atrial fibrillation, myocardial infarction, heart
failure, arterial hypertension
Received 22 December 2018; accepted 11 April 2019

Epidemiology of non-steroidal country NSAID sales of 175 m Euros (i.e. 2.89 Euros
anti-inflammatory drugs use and per inhabitant); the number of sold defined daily doses/
1000 inhabitants/day in 2017 was 19.2 (slightly declin-
potential risks ing from 2013), much lower than those reported for
Non-steroidal anti-inflammatory drugs (NSAIDs) are Northern European countries in 2016, where the
among the most used drugs in the world to treat a var- values were 74.3 in Iceland, 73.9 in Finland, 54.4 in
iety of conditions including pain, rheumatoid arthritis, Sweden, 43.8 in Norway and 31.8 in Denmark.4,5
osteoarthritis, musculoskeletal disorders. NSAID use is Moreover, according to the same Italian OSMED
reported to increase with age as many chronic pain report, NSAID consumption reached a prevalence of
conditions, such as osteoarthritis, become more preva-
lent; overall, an estimated 10–40% of people aged over 1
Cardiorenal Research Unit, University of Parma, Parma, Italy
65 years use prescribed or over-the-counter NSAIDs 2
Cardiology Unit, Ospedale Vaio, Vaio-Fidenza, Parma, Italy
daily.1 In the USA, NSAIDs reached 98 m prescriptions 3
Unità di Medicina Interna, Università di Pavia, Vaio-Fidenza, Parma, Italy
in 2012 with more than 29 m adults being regular 4
Cardiology Unit, Azienda Ospedaliera-Universitaria di Parma, Italy
users.2 In Sweden, NSAIDs are the most commonly 5
Parma Health Authority, Parma, Italy
prescribed oral analgesics for musculoskeletal pain,
accounting for almost 80% of prescriptions over a Corresponding author:
Aderville Cabassi, Cardiorenal Research Unit, Clinica e Terapia Medica,
five-year period.3 In Italy, the 2017 OSMED report4 Department of Medicine and Surgery, DIMEC University of Parma, Via
(which includes both doctors’ prescriptions and over- Gramsci 14, 43126 Parma, Italy.
the-counter NSAID consumption) refers to total Email: aderville.cabassi@unipr.it
Cabassi et al. 851

33% for people > 65 years.4 Because chronic pain often (COX-1)-selective ketorolac caused the highest risk of
calls for prolonged use of NSAIDs, elderly people with hospitalization among inappropriately prescribed
multiple comorbidities are exposed to a worrisome drug medications.9 The excessive or inappropriate use of
burden (a phenomenon known as polypharmacy). The NSAIDs can affect cardiovascular and renal function
latter, coupled with age-related progressive decline in because of their specific effects on prostanoid synthe-
renal function makes the elderly more vulnerable to the sis.10 Issues concerning cardiovascular, renal and
noxious effects of many drugs, especially NSAIDs.6 gastrointestinal safety are therefore critical for clin-
Their potential toxicity is further aggravated by the icians. Consequently, we decided first to review the
fact that their use is often inappropriate, and without prostanoid physiology of vascular, cardiac and renal
real control by doctors. Patients themselves, motivated systems to explain the effects of NSAIDs and their
by the efficacy and availability of these drugs, increase potential deleterious mechanisms, and then to translate
the doses and prolong their use. Uncontrolled NSAID the information to clinical conditions.

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


use, mainly fuelled by the over-the-counter market and
widespread advertising, is a worldwide problem exacer- Physiology of prostanoids: how do they
bated by varying prescription rules which can under- interfere with cardiovascular and renal
rate true NSAID consumption. NSAID-related adverse
effects are among the leading causes of hospital admis-
function?
sion, especially in the elderly, and rank among the top Prostanoids, including prostaglandins (PGs) and
drugs causing adverse effects.7 We recently reported, in thromboxanes (TXs), are derivative product of C20
a study conducted in a large population of elderly unsaturated fatty acids metabolism, mainly arachidonic
patients (n ¼ 80,229) in Emilia Romagna, a Northern acid (C20:4(-6)), the most abundant precursor in
region of Italy, that prolonged use of NSAIDs (more (C20:4(o-6)) humans with 5 cis- and 13 trans- double-
than 15 days) was the most common inappropriately bond prostanoids relevant in human species (Figure
prescribed class of medications. Specific interventions 1).10 Arachidonic acid is released from cell membrane
focused on the appropriateness of prescriptions in the phospholipids after ester bond cleavage is performed by
elderly by general practitioners halved the inappropri- the enzymes of the phospholipase A2 family. Once the
ate use of NSAIDs.8 After 10 years (2003–2013) of fol- ester bond between arachidonic acid and the membrane
lowing a cohort of elderly patients who resided in is hydrolysed, arachidonic acid is metabolised by pros-
Northern Italy, we found that cyclooxygenase-1 taglandin G/H synthase (PGHS), the key enzyme

Membrane phospholipids

Phospholipase A 2
H3C

Arachidonic Acid
NSAIDs or X HO
Cyclooxygenase 1 and 2
COX 2 inhibitor PGH2
O

PGG 2
synthase

PGH2
PGD2 PGE2 PGF2 PGI2 TxA 2
synthase isomerases synthase synthase synthase
PGD2 PGE2 PGF2 PGI2 TxA2

DP1 DP2 EP1 EP2 EP3 EP4 FP IP TP


Gαs Gαi Gαq Gαs Gαi/s/q Gαs Gαq Gαs Gαi/s/q

cAMP cAMP [Ca++] cAMP cAMP cAMP [Ca++] cAMP cAMP


[Ca++] [Ca++] [Ca++]

PGs and Tx receptors: green boxes indicate «relaxant», red boxes «constrictive» receptors.

Figure 1. Pathways in biosynthesis of prostanoids from arachidonic acid to prostaglandins (PGs) and thromboxane (TX) generation
and receptor signalling. cAMP: cyclic adenosin monophosphate; COX: cycloxygenase; DP, EP, FP, IP, TP are PGs and Tx receptors; Gai/
s/q are G protein coupled receptor subunits; NSAID: non-steroidal anti-inflammatory drug.
852 European Journal of Preventive Cardiology 27(8)

involved in PG and TX synthesis and commonly endothelial cells.18 Further studies are needed to clarify
known as COX. COX has both cyclooxygenase and their specific physiological role in PGI2 generation.
hydroperoxidase activity, with PGH2 as the reaction
end-product. PGH2 is further converted by specific
Vascular system
PG synthases to specific and biologically active PGs
or to TXA2 through TX synthase. As many prostanoids The effects of PGs and TXs on vascular tone and func-
(PGI2 – usually referred to as prostacyclin, PGH2 and tion, including platelet activation and aggregation, are
TXA2) are chemically unstable molecules that are probably the most studied in prostanoid research. Given
degraded to inactive products within a few minutes, the premise that most of the seminal information and
they are expected to act locally and play a paracrine scientific advances on cardiovascular and renal effects of
role. Prostanoids exert their effects through five main prostanoids derive from experiments in mice that repre-
subtypes of receptors: EP1, EP2, EP3 and EP4 for PGE2, sent the animal model most used in knockout gene

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


DP1 and DP2 for PGD2, and FP, IP and TP receptors investigation, the observation of their characteristics,
for PGF2a, PGI2 and TXA2 respectively (Figure 1). In although not directly transferable to humans, clearly
humans, alternative splicing leads to an additional eight contributes to the understanding of human physiology.
EP3 receptor isoforms, two TP isoforms (TPa, TPb), Protein coding gene regions in humans and mice are
and some other FP isoforms.11 Human PG receptors 85% identical with a range between 60–99% homology.
are dependent on G-protein activation and are grouped Amino acid homology between human and mouse PGs
in G-protein-dependent cAMP and/or calcium signal- and TXs receptors ranges between 71–88%, with IP and
ling receptors. In fact, they can be coupled to either TP receptor sharing 79 and 74% of the sequence,
Gas (EP2, EP4, IP and DP1) that elevates intracellular respectively.11 PGI2 and TXA2 have opposing effects
levels of cAMP, or Gaq (EP1, EP3 and FP) that on both platelets and blood vessels, with the former
increases calcium intracellular levels, or Gas-proteins being an endogenous anticoagulant agent as shown in
(EP3, DP2 and TP) that can both increase or decrease knock-out mice (IP –/–) for its receptor, and the latter
levels of cAMP and calcium.10,11 PG receptor signalling having vasoconstrictive and pro-thrombotic effects. IP
involves both a cAMP-protein kinase A dependent –/– mice have normal development and show throm-
mechanism and independent mechanisms; the latter botic diathesis when early endothelial damage
are represented by the exchange proteins directly acti- occurs.19 TP knock-out mice were prone to bleed due
vated by cAMP (Epac1 and 2), which participate in to a reduced ability to activate platelets in the case of
cardiovascular functions, regulating calcium handling balloon catheter-induced injury.20 A possible explan-
and vascular tone.12 In the PG and TX biosynthetic ation for the PGI2 anticoagulant effect came from the
cascade, two COX isoforms have been described in observation of its ability to stimulate thombomodulin in
humans, with a 60% homology in amino acid sequence; endothelial cells, eventually promoting C-protein activ-
COX-1 (PGHS-1) is typically considered the constitu- ity. However, in this study, some of the pro-thrombotic
tive, housekeeping and almost ubiquitously expressed and vascular effects of IP deletion were completely abro-
isoform, responsible for prostanoid production in most gated by TP deletion, suggesting that PGI2 acts mainly
tissues.13 COX-1 is the major source of TXA2 in both as a modulator of the pro-thrombotic and vasocon-
endothelial and activated platelets and represents a strictive action of TXA2.21 PGE2 is also involved in
target for acetylation and inhibition by low dose acet- platelet aggregation, facilitating activation through the
ylsalicylate. COX-2 (PGHS-2) has long been believed to EP3 receptor.22 Aside from a modulatory effect on plate-
be the inducible isoform, expressed at the site of inflam- let activity, prostanoids are also implicated in athero-
mation, infection or cancer.14 However, recent gene sclerosis development. Indeed, apoE-deficient mice, a
expression studies in mice demonstrated a constitutive model of atherogenesis, crossbred with IP –/– or TP
expression of COX-2 in tissues such as brain, gut, –/– mice showing distinctive vascular patterns. ApoE
thymus and kidney.15 The constitutive expression of –/– TP –/– and apoE –/– IP –/– mice show, respectively,
COX-2 in kidneys is regulated by transcriptional fac- a delay or an acceleration in atherogenesis. ApoE –/– IP
tors unrelated to inflammation, supporting a physio- –/– mice have a more vulnerable plaque profile (due to
logical role for this enzyme. In human pathological partial endothelial disruption) and a greater expression
conditions, such as arterial hypertension, diabetes or of adhesion molecules like ICAM-1, promoting macro-
heart failure (HF), COX-2 expression increases in phage migration.23 Prostanoids play an important role
both renal cortex and medulla.16 COX-2 is responsible in endothelial-mediated vascular tone: in this regard
for PGI2 generation in humans.17 Yet, other reports COX-2 seems to be of major relevance, as COX-2 –/–
based on immunohistochemistry and bioluminescent mice, albeit showing normal constitutive PG synthesis,
imaging point to COX-1 as a principal source of evidence both a hypertensive pattern and an enhanced
PGI2 in physiological conditions in human aortic pressor effect after angiotensin II infusion.24 In human
Cabassi et al. 853

coronary arteries from diabetic patients, COX-2 over- with a specific loss of EP4 receptor only in vascular
expression has been proposed as a relevant compensa- smooth muscle, finding an attenuation of the acute
tory mechanism to preserve coronary flow under PGE2-mediated vasodilation. They also reported the
conditions of impaired nitric oxide-dependent vascular ability to attenuate a rise in blood pressure after high-
control through increased PGI2 production.25 PGI2 and salt feeding or long-term angiotensin II infusion in mice
TXA2 show opposite effects on vascular tone regulation, where the loss of EP4 receptors occurred in all tissues
determining vasodilator and vasoconstrictive action but not in those where the loss of EP4 was limited only
respectively.10 NSAIDs inhibit COX isoforms differ- to vascular smooth muscle cells. The concept of a cen-
ently and reduce PG and TX synthesis (Figure 1).16,26 tral and dominant role played by EP4 receptors in vas-
An increased cardiovascular risk associated with cular tone control and arterial hypertension prevention
NSAIDs derives from the degree of COX-1 and COX- is emerging.29
2 inhibition which differently reduce both the antith-

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


rombotic PGI2 and the pro-aggregatory and vasocon-
Heart
strictive TXA2. NSAID-induced PGI2 lowering leads to
increased platelet aggregation which might be counter- Several reports indicate a direct and specific effect of
balanced by a simultaneous inhibition of platelet COX- PGs on cardiac tissue; most of this information derives
1 activity resulting in reduced TXA2 levels. However, from studies in transgenic animal models that evaluated
except for low-dose acetylsalicylate (75–150 mg) and the effects of prostanoids on cardiac fibrosis, ischaemia
high-dose naproxen (1000 mg), both nonselective and and arrhythmias. PGE2 stimulates cardiac myocyte
COX-2 selective NSAIDs do not modify platelet hypertrophy, acting through EP4 receptors and seems
COX-1 activity to the degree necessary (>95%) to inhi- to have a protective effect after ischaemia/reperfusion
bit TXA2 pro-aggregatory effects. Thus the net effect injury.30,31 A similar effect on cardiomyocyte preserva-
will be an impairment of the balance between TXA2 tion after ischaemia/reperfusion injury had been
and PGI2 levels, thereby fostering atherosclerosis, reported for PGI2 in a IP –/– model of mice. PGI2
thrombosis and other cardiovascular complications.10,26 showed favourable action on ventricular remodelling,
Apart from the latter effects mediated via the inhibition with increased cardiac fibrosis (perivascular and inter-
of the COX pathway, both non selective and COX-2 stitial) in IP –/– mice following different kinds of vas-
selective NSAIDs may induce cell apoptosis and cular injuries, namely MI and aortic banding.32 Tissue
increase the generation of reactive oxygen species by specific gene-deletion of COX-2 in cardiomyocytes sug-
altering specific cell signalling pathways thus contribut- gested a role for PGs in preserving normal ventricular
ing, through different mechanisms, to the genesis of car- function that otherwise appeared to be depressed and
diovascular complications such as thrombosis, with an increased susceptibility to ventricular
myocardial infarction (MI), HF and arrhythmia.27 arrhythmias.33
PGE2, the most abundant prostaglandin in humans,
shows a diversified effect on vascular tone based on
Kidney
the district studied.21 This effect is dependent on binding
to the four membrane receptors for EP1, EP2, EP3 and COX-1 is expressed in the kidney and regulates physio-
EP4, coupled to different signalling pathways. logical renal vascular and tubular functions as well as
Vasodilation and hypotension obtained after PGE2 hormonal secretion; COX-2 is constitutively expressed
infusion is mediated through EP2/EP4 receptors, both in the kidney.16 Both are located at the endothelial and
coupled to adenylyl cyclase activation, whereas vaso- tubular levels as well as expressed in the interstitial
constriction (in cerebral circulation) is related to stimu- medullary cells. PG synthesis is not uniformly distribu-
lation of EP1 receptors that are coupled to Gaq proteins, ted along the nephron.34 The medullary synthesis of PG
resulting in increased phosphatidylinositol hydrolysis exceeds that of the cortex.34 In the renal medulla most
and elevation of intra-cellular calcium levels. The EP2 PG synthesis comes from interstitial medullary cells and
receptor plays a greater role in modulating vascular tone collecting tubules, whereas in the cortex the interlobular
and blood pressure regulation as observed in knock-out arteries, the afferent arterioles and the glomeruli are the
mice, inducing a rise in blood pressure levels associated major biosynthetic sites. Reduced COX activity is
with impaired sodium handling.10,28 EP2 and EP4 are observed in proximal tubules and the loops of Henle.
generally considered the relaxant receptors mainly PGE2 is the major prostanoid secreted by tubules,
because of their effect on vascular smooth muscle while at the glomerular levels both PGE2 and PGI2 are
cells. A complex role for the EP4 receptor in the control present in a similar amount. Renal PGE2 and PGI2 are
of vascular tone and pathogenesis of arterial hyperten- responsible for renin secretion from the macula densa,
sion has recently been suggested.29 Using conditional even if other arachidonic acid metabolites have been
gene targeting techniques, the authors generated mice suggested to participate directly in renin release
854 European Journal of Preventive Cardiology 27(8)

regulation.35 A specific role for COX-2-generated PGs in release from glomerular mesangial and epithelial cells
the macula densa has been demonstrated in acute and and renal medullary interstitial cells, which is blunted
chronic regulation of renin release.36,37 COX-2 specific by COX-1 and COX-2 inhibition leading to an increase
antagonism or COX-2-deficient mice showed reduced in arteriolar constriction (Figure 2(a) and (b)).
levels of renin expression and activity.35,38 Conversely, Therefore, when true hypovolaemia due to gastro-
increased COX-2 expression has been associated with intestinal or renal losses (as occurs with diuretic ther-
high-renin conditions such as in volume-depleted long- apy) or an effective volume depletion due to HF or
term furosemide-treated rats and in patients with Bartter cirrhosis or nephrotic syndrome occurs, PG generation
syndrome, linking COX-2-related PG synthesis to NaCl becomes clinically relevant in order to antagonise the
uptake through NKCC2 in the loop of Henle.39 PGs are renal effects of vasoconstrictors and the reduction in
also able to antagonise the antidiuretic effect of vaso- renal blood flow and glomerular filtration rate. Thus
pressin and are involved in the natriuretic effect of dopa- an increased risk of ischaemia-related acute tubular

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


mine and natriuretic peptides.16,40 Renal function, necrosis exists when there is a reduction of peritubular
including vascular perfusion and resistance, glomerular flow following NSAID inhibition of PG-related affer-
filtration and sodium excretion in normal subjects is ent arteriole vasodilation.41 The risk of tubular
marginally dependent on PG secretion. In fact, damage related to nephrotoxic drugs or contrast
NSAID administration does not affect renal function media can be facilitated by concomitant NSAID
when other regulatory mechanisms, such as adrenergic intake.41 In addition, in human and experimental
tone, the renin-angiotensin aldosterone system, the glomerulonephritis the increased generation of PGs
endothelin system or the adenosine and dopamine sys- and TXA2 by the glomeruli are involved in the severe
tems, are not activated.34 When hypoperfusion or a reduction of glomerular filtration rate with relative
hypovolaemic setting occurs, and a concurrent higher renal blood flow sparing. NSAIDs can differently
circulating release of vasoconstrictors (angiotensin II, affect not only afferent arteriolar and glomerular
norepinephrine, vasopressin and endothelin) is elicited, haemodynamics, but also glomerular capillary ultrafil-
PG generation becomes fundamental to maintain renal tration coefficients.41,42 Hence, NSAIDs can be
perfusion and limit ischaemia. Norepinephrine, angio- responsible for kidney dysfunction and for altered
tensin and vasopressin stimulate PGE2 and PGI2 electrolyte and water balance.16,41

(a) (b)
PG COX-1 and COX-2
PG ANG II ANG II
inhibition
INTERLOBULAR ARTERY
INTERLOBULAR ARTERY

AFFERENT ARTERIOLE
AFFERENT ARTERIOLE

EFFERENT ARTERIOLE
EFFERENT ARTERIOLE

RBF normal RBF reduced


GFR normal GFR reduced

Figure 2. (a) Prostaglandin (PG)-mediated vasodilation of afferent arterioles prevents renal vasoconstriction and reduction of
glomerular filtration rate (GFR) in clinical conditions characterised by low effective volume such as heart failure. (b) Non-steroidal
anti-inflammatory drugs impair afferent arteriole vasodilation leading to renal hypoperfusion (renal blood flow (RBF)) and reduced
GFR. ANG II: angiotensin II; COX: cycloxygenase.
Cabassi et al. 855

Pharmacology of NSAIDs and dose. Because NSAIDs (except for acetylsalicylate)


NSAIDs derive from different chemical structures and tightly bind to protein, they can increase the risk of
are classified, based on their half-lives, into short and bleeding in patients treated with vitamin K antagonists
long acting drugs (Table 1). This is an important issue through warfarin displacement from plasma proteins.44
to deal with, because the evaluation of efficacy and The level of COX isoform inhibition is responsible for
toxicity must include the time it takes for an NSAID NSAIDs efficacy and toxicity.14,43 As specified above,
to reach a plasma steady-state level, which is about COX possesses both cyclooxygenase and hydroperoxi-
3–5 times its half-life. Most NSAIDs, after oral admin- dase catalysis properties: NSAIDs and acetylsalicylate
istration, are well absorbed, show minimal first-pass are able to inhibit both reactions whereas acetamino-
hepatic metabolism and bind tightly to serum pro- phen acts by blocking only hydroperoxidase catalysis.45
teins.43 When plasma albumin concentration falls, Such a specific effect explains why acetaminophen
because of reduced hepatic synthesis, as occurs in cir- does not display anti-inflammatory or antithrombotic

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


rhosis, during the active phase of inflammatory diseases effects as NSAIDs do, but rather antipyretic and
or because of urinary albumin loss in nephrotic syn- analgesic properties. NSAIDs efficacy and toxicity
drome, higher levels of free protein-unbound active are also dependent on non-PG-mediated NSAID
NSAIDs are circulating. The risk of different events effects, involving a modulatory effect on specific tran-
varies depending upon the clinical context, medication scription factors such as nuclear factor kappa B and

Table 1. Non-steroidal anti-inflammatory drug (NSAID) classification based on duration of action, chemical structure and
cyclooxygenase (COX) isoform selectivity.
Short acting (< 6 hours) Long acting (> 6 hours)

Diclofenac Naproxen
Ketoprofen Piroxicam
Acetaminophen Nabumetone
Acetylsalycilate (Aspirin) Valdecoxib
Indomethacin Celecoxib
Ibuprofen Etoricoxib
Ketorolac Sulindac

Chemical Structure

Salycilic Acid: Acetylsalycilate Diarylheterocyclic: Celecoxib, Valdecoxib,


Etoricoxib
Acetic Acid: Ketorolac, Indomethacin,
Nabumetone, Sulindac Enolic acid: Piroxicam

Phenylacetic Acid: Diclofenac Aniline derivative: Acetaminophen

Propionic Acid: Naproxen, Ketoprofen,


Ibuprofen, Fenoprofen

COX 1 selectivity COX 1 COX 2 COX 2 selectivity

Ketorolac
Ketoprofen
Indomethacin
Acetylsalycilate (Aspirin)
Nabumetone
Sulindac
Naprossen
Piroxicam
Ibuprofen
Acetaminophen
Diclofenac
Celecoxib
Valdecoxib
Etoricoxib
856 European Journal of Preventive Cardiology 27(8)

activator proteins on neutrophil-endothelial adhesion 1 g/24 h) can suggest a NSAID-induced glomerular


molecules and on nitric oxide synthase enzymes.46 lesion (minimal change disease or membranous nephro-
The interactions between NSAIDs and other drugs pathy) often associated with hyaline casts.16 NSAID-
can clearly increase the risk of toxicity, with both related AKI could be also due to acute tubular necrosis:
renal (haemodynamic) and cardiovascular (haemor- in this case urinary sediment contains renal tubular epi-
rhagic risk) adverse effects. Several NSAIDs can thelial cell casts, renal tubular epithelial cells or granu-
increase the plasma levels of drugs by partially inhibit- lar casts. Furthermore, NSAIDs can lead to AIN
ing CYP-2C9 (indomethacin, ibuprofen and diclofenac) showing in this case an increase in white blood cells
or CYP-2C8/2D6 (celecoxib).47 A careful clinical and and white blood cell casts in the urinary sediment.
pharmacological anamnesis is a fundamental step of NSAIDs as a class (in particular propionic acid deriva-
NSAID therapy. Multiple NSAID combinations as tives such as fenoprofen) (Table 1), including COX-2
well as concurrent NSAID use with acetaminophen at selective drugs, can induce both AIN and a nephrotic

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


high doses should be avoided because of the possibility syndrome associated with minimal change or mem-
of a higher risk of gastrointestinal complications and branous nephropathy.52,53 A 30% rise of NSAID-
no greater efficacy of their combination.48 mediated AIN cases is reported especially in elderly
people, mainly because of polypharmacy. Nephrotic
Acute and chronic renal damage by non- syndrome can occur in NSAID-mediated AIN but
is rare (less than 1%).53 AIN occurrence is not
selective and COX-2 selective NSAIDs dose-dependent and can recur if the same drug is re-
Around 3% of all patients using NSAIDs can experi- administered.54 There may be a period of variable
ence adverse renal effects translating into more than latency (from a few days to several months) between
2.5 m patients experiencing one or more nephrotoxic the onset of NSAIDs treatment and the appearance of
events annually.49 Both non-selective and COX-2 AIN. Patients with AIN show an acute rise in creatin-
selective NSAIDs can induce an acute kidney injury ine levels associated with both oliguria or non-oliguria.
(AKI).16,41 NSAIDs can induce several forms of Fever, rash or blood eosinophilia and urine eosinophils
kidney damage including haemodynamically mediated can sometimes (less than 10% in combination) accom-
AKI, oedema, electrolyte and acid-base disorders, pany the AIN clinical pattern. Discontinuation of
acute tubular necrosis and acute interstitial nephritis NSAIDs allows recovery from AIN within several
(AIN), which may be accompanied by the nephrotic weeks to several months. The exact mechanism of
syndrome and papillary necrosis.41 Both non-selective NSAID-mediated AIN is not clear, even if T-helper
and COX-2 selective NSAIDs were able to reduce lymphocyte activation by leukotrienes (whose gener-
glomerular filtration rate to a similar degree in an ation seems facilitated by NSAID-mediated COX
elderly population.50 The presence of only one of the inhibition), has been suggested.55 In addition to AKI,
following risk factors is necessary to generate NSAID- an excessive use of NSAIDs, in particular analgesic
induced AKI: absolute volume depletion and dehydra- mixtures containing phenacetin, was found to be a rele-
tion, reduced effective arterial volume or severe hyper- vant cause of progressive kidney disease, through direct
calcaemia.16 Chronic kidney disease and old age are renal papillary ischaemia up to necrosis or chronic
also major risk factors leading to NSAID-induced interstitial nephritis. Up to the late 1980s, at least
AKI. Diuretics, angiotensin-converting enzyme inhibi- 10% of end-stage renal disease in Switzerland, and in
tors (ACEis), angiotensin II receptor 1 blockers, also other countries such as Belgium and Australia, was due
increase the risk of AKI. Patients with HF and reduced to analgesic-mediated nephropathy: only reduced use of
effective arterial volume doubled the risk of developing analgesic mixtures containing phenacetin allowed a
AKI when treated with COX-2 selective and non- marked lowering of its incidence.56 The renal medulla
selective NSAIDs by blunting PG-mediated vasodila- is the major site of ischaemic damage associated with
tion of afferent arterioles, thus favouring renal hypo- vasa recta capillary sclerosis and consequent papillary
perfusion and reducing glomerular filtration rate tip necrosis. Both low PG generation and glutathione
(GFR) (Figure 2(a) and (b)).16,34 Special attention depletion contribute to the toxic medullary effect of
should be paid even in chronic kidney disease patients NSAIDs. The laboratory pattern of analgesic nephro-
with mild reductions of GFR when chronic treatment pathy is not specific and includes a long-period, slow-
with NSAIDs is started.51 In haemodynamically- rising increase in plasma creatinine levels associated
mediated AKI following recent NSAID use, the labora- with normal urine sediment, or only mild proteinuria.
tory pattern includes increased plasma creatinine levels, If the analgesic mixture is discontinued, the progression
the absence of significant proteinuria (proteinuria less of chronic renal disease can be stopped, and glomerular
than 1 g/24 h), haematuria and a bland urine sediment. filtration can even improve, except in those with previ-
The presence of higher levels of proteinuria (more than ous severe decline of renal function when high doses
Cabassi et al. 857

were used.56,57 Even acetaminophen (paracetamol), the arm.65 High doses of acetylsalicylate can affect blood
main metabolite of phenacetin and a widely prescribed pressure levels, whereas a low dose (75 mg/day) does
antipyretic and analgesic drug, may be associated with not.66 Long-term oral administration of acetaminophen
a progressive loss of renal function in patients with can increase both systolic and diastolic blood pressure
advanced renal failure, especially when the lifetime levels,67 even if several studies showed a neutral effect in
accumulated dose is elevated. In patients whose renal hypertensive patients.68 On the contrary, acute intra-
function is only mildly reduced, no risk of progression venous acetaminophen administration lowers blood
was observed with less frequent drug consumption.58 pressure.69 Therefore, because NSAIDs exert their
Yet, as recently reported, if acetaminophen therapeutic effects in a dose-dependent way, they should be used
doses are used in association with low-moderate alco- at the lowest effective dose and for the shortest duration
hol consumption, a higher percentage of patients with of time in hypertensive patients. It is also known that
progressive loss of renal function occurs.59 Chronic NSAIDs use can cause alterations in electrolyte and

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


therapeutic use of low doses of acetylsalycilate does water balance. These alterations include hyperkalaemia,
not seem to be related to progressive risk of renal fail- hyponatraemia and oedema. Hyperkalaemia can be the
ure in the majority of studies, but an increased risk can result of both reduced renin and aldosterone secretion
occur when they are given in combination with acet- by lowering PG generation in the macula densa and
aminophen and other NSAIDs.58,60 There is consider- angiotensin II sensitivity in the adrenal cortex respect-
able evidence that high doses of both COX-2-selective ively. Hyperkalaemia can also be the result of NSAID-
and non-selective NSAIDs should be avoided in related kidney damage, and the concurrent use of drugs
patients with chronic renal insufficiency.16,41 The which can raise potassium plasma levels (ACEis, angio-
recent Prospective Randomized Evaluation of tensin II receptor 1 blockers, direct renin inhibitors and
Celecoxib Integrated Safety Versus Ibuprofen or potassium sparing diuretics). The magnitude of the rise
Naproxen (PRECISION) trial, which included osteo- in plasma potassium ranges from less than half of a
arthritis and rheumatoid arthritis patients with at millimole to more than a millimole per litre in elderly
least one cardiovascular risk factor, clearly showed people.70 No rise or a smaller increase can be observed
that the COX-2 selective drug celecoxib, when admin- in healthy subjects. Less common is NSAID-induced
istered at moderate doses, may be associated with fewer hypokalaemia. A few case reports have shown that ibu-
renal events than ibuprofen (–39%). A similar trend profen, at high doses, can induce renal tubular acidosis
was also observed when celecoxib was compared with with severe hypokalaemia and that its discontinuation
naproxen.61 allows rapid recovery from hypokalaemia and hyper-
chloraemic acidosis.71 Both non-selective and COX-2
NSAID effects on blood pressure, selective NSAIDs can also affect free water excretion
by facilitating antidiuretic hormone (ADH) activity
electrolytes and water balance leading to hyponatraemia.16 When effective arterial
COX-2 selective and non-selective NSAIDs can induce volume is reduced, such as in severe HF patients,
a rise of blood pressure in both normotensive and ADH levels are increased. Administering NSAIDs can
hypertensive patients. An estimate of NSAID-related further increase ADH secretion and activity, and deter-
blood pressure increase is around 1–2 mm Hg in normo- mine or worsen hyponatraemia in these patients.
tensive subjects and can reach 14 mm Hg in patients Hyponatraemia can also be induced in patients with
with hypertension.62,63 These drugs can impair the effi- cirrhosis and nephrotic syndrome or in those affected
cacy of antihypertensive drugs in already treated hyper- by a syndrome of inappropriate ADH secretion.16,41
tensive patients; the negative impact of NSAIDs (COX- Hyponatraemia is a well-recognised complication of
2 selective included) on blood pressure control is not thiazide diuretic treatment through mechanisms inter-
observed when calcium channel blockers are used, and fering with maximal urinary dilution and stimulation of
is minimal for beta adrenergic blockers.64 Renal COX-2 ADH release. Concurrent therapy with NSAIDs can
inhibition appears to be one of the mechanisms leading facilitate its appearance.72 Both nonselective and
to a lower natriuresis and increased intravascular COX-2 selective NSAIDs can induce sodium retention
volume, with a subsequent significant rise in blood pres- leading to oedema formation and diuretic resistance in
sure. Recently the results of the PRECISION-ABPM HF patients.16,34 They can also decompensate chronic
trial, where moderate doses of a COX-2 selective inhibi- HF patients by diminishing diuretic efficacy, inducing
tor were compared to two non-selective NSAIDs on sodium and water retention: This leads to weight gain
24-hour ambulatory blood pressure in patients affected and increased vascular resistance. All NSAIDs
by osteoarthritis and rheumatoid arthritis, indicate a bring about these effects; sulindac and low-dose acetyl-
neutral effect on mean systolic blood pressure of cele- salicylate induce a partial and transient inhibition of
coxib, but a significant increase in the ibuprofen-treated renal PG synthesis, differently from the irreversible
858 European Journal of Preventive Cardiology 27(8)

inhibition of platelets by COX-1, and therefore no consumption and cardiac arrest. A direct association
or much less frequent kidney damage occurs with between ibuprofen and diclofenac use and the risk of
these drugs.73 cardiac arrest has been observed in a case-time-control
study based on the Danish Cardiac Arrest Registry. In
patients, who experienced an out-of-hospital cardiac
Non-selective and COX-2 selective arrest, NSAID exposure up to 30 days before cardiac
NSAID effects on cardiac function arrest was identified as a major risk factor. Ibuprofen
increased the risk of cardiac arrest by 31% and
Atrial fibrillation and cardiac arrest diclofenac by 50%, whereas no significant association
Case-controls, randomised and observational cohort was found with COX-2 selective inhibitors nor with
studies and a meta-analysis have evaluated the effect naproxen.81 These findings suggest that both non-
of COX-1 and COX-2 inhibition on atrial fibrillation selective and COX-2 selective NSAID prescription

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


(AF) development and recurrence (Table 2).74–79 In a may represent a potential risk factor for AF
case-control study of general practitioners in the UK a occurrence.
significant increase in the risk of chronic but not par-
oxysmal AF, was reported in patients taking NSAIDs.
This risk was greater in long-term users, when treat-
Myocardial infarction
ment duration was longer than one year and gradually Following the development of COX-2 selective inhibi-
disappeared after NSAIDs withdrawal.74 A Danish tors, several concerns regarding cardiovascular safety,
study showed that both non-selective NSAIDs and in particular atherothrombotic vascular events, have
COX-2 selective inhibitors users were respectively been raised. The first observation came from the
associated with a 33% and a 50% higher risk of a Vioxx gastrointestinal outcomes research (VIGOR)
first episode of AF or atrial flutter as compared to study where rheumatoid arthritis patients taking rofe-
non-users. A higher risk was identified in the elderly coxib, despite presenting less upper gastrointestinal
and in patients with chronic kidney disease or rheuma- bleeding, experienced an excess of vascular events
toid arthritis, particularly when they started treatment when compared to the group of patients treated with
with COX-2 inhibitors.75 In another study, new users naproxen.82 However, very few events were reported,
of non-selective NSAIDs were at increased risk of AF; and the study was clearly too underpowered to permit
in particular, the risk doubled in HF patients. Instead, statistical evaluation of cardiovascular safety.82 After
patients exposed to selective COX-2 inhibitor did not this observation, various randomised controlled trials
show a higher risk (except for those with chronic (RCTs) and observational studies concerned the cardio-
kidney or pulmonary diseases).76 In contrast, a large vascular safety of COX-2 selective and COX-1 COX-2
cohort population study from Sweden showed a stron- non-selective NSAID drugs (Table 3). In 2004, rofe-
ger association between selective COX-2 inhibitor use coxib was withdrawn from the market because of con-
and the incidence of AF, although a significant risk cerns over an increased risk in MI and strokes
was also observed for non-selective NSAIDs.77 The associated with a long-term (beginning after 18
incidence of AF was also more common in a nation- months of continuous therapy in the Adenomatous
wide cohort study from Denmark where patients with Polyp PRevention On Vioxx (APPROVe) study), and
MI were prescribed NSAIDs for long period but also a high-dose usage.83 Other evidence indicated an earlier
for short-term treatment (0–14 days).78 In a meta-ana- rise in MI risk, occurring within a week after the first
lysis of five observational studies with 400,000 cases of prescription of rofecoxib in patients with a previous his-
AF, NSAID users had a 12% higher risk of AF, rising tory of MI.84,85 Interestingly, in contrast with other
to 53% for new users. COX-2 inhibitors, including NSAIDs, where the risk for MI was present only
diclofenac, were associated with major risk as com- during treatment, rofecoxib-related MI risk may have
pared to non-selective NSAIDs.79 The mechanisms continued for an undefined period after drug with-
linking AF development and NSAID use are unclear. drawal.85,86 The first meta-analysis of data derived
It has been hypothesised that renal COX-2 inhibition from RCTs reported an increased incidence of vascular
can trigger a cascade of events including fluid reten- events for coxibs versus placebo (42% proportional
tion, increased blood pressure, increased end-diastolic increased risk), caused mainly by increased MI occur-
pressure, all leading to AF. A role for atrial fibrosis is rence. No differences arose between coxibs vs NSAIDs
also suggested but still under investigation.80 Serum (high dose ibuprofen or diclofenac), but high doses of
potassium level changes as well as the pro-arrhythmic naproxen evidenced a safer profile.87 Another meta-
effects of concomitant drug use in polypharmacy-trea- analysis of RCTs indicating comparable risk of vascular
ted subjects might be additional and contributing events with coxibs and diclofenac or high dose ibupro-
mechanisms. A recent report linked NSAID fen confirmed the results.88 Interestingly, although the
Table 2. Association between non-steroidal anti-inflammatory drug (NSAID) use and atrial fibrillation in clinical studies.

First author and publi- Anti-inflammatory


cation year Study type drugs Number of patients Endpoint analysed Risk modification Comments
Cabassi et al.

74
De Caterina et al., Nested case control NSAIDs and SAIDs 1570 case patients Atrial fibrillation RR of 1.44 (95% CI No effect of previously
2010 study 1.08–1.91) for diagnosis of HF in
chronic atrial fibril- modifying RR for
lation atrial fibrillation in
RR was highest for the NSAIDs users.
use of NSAIDs for
more than 1 year
(RR 1.80; 95% CI
1.20–2.72).
75
Schmidt et al., 2011 Population-based con- NSAIDs and COX-2 2925 case patients Atrial fibrillation in Adjusted incident rate Higher risk for elderly
trol-study inhibitors current or past ratio for current and CKD patients.
users NSAID users 1.17
(95% CI 1.10–1.24)
Adjusted incident rate
ratio for current
COX-2 users 1.27
(1.20–1.34).
76
Chao et al., 2012 Nationwide case-con- NSAIDs and COX-2 7280 case patients Atrial fibrillation OR 1.651 for new COX-2 use increased
trol study inhibitors users atrial fibrillation risk
OR 1.920 for new only in CKD and
users HF patients pulmonary disease.
Non-significant risk
increase for COX-2
exposure.
77
Back et al., 2012 Nation-wide popula- COX-2 inhibitors 6,991,465 patients Cardiovascular events Adjusted HR 1.16 Borderline significance
tion-based study analysis (95% CI 1.046– for increase stroke
1.289 for atrial fib- risk (p ¼ 0.08) in
rillation in COX-2 Coxib exposed
inhibitors exposed patients.
patients.
78
Schjerning Olsen Nationwide adminis- NSAIDs and COX-2 7831 case patients Atrial fibrillation in Adjusted HR 1.29 Risk greater in short
et al., 2015 trative registry inhibitors post-myocardial (95% CI 1.14–1.45) term users (0 to 14
infarction patients for NSAIDs; HR days).
1.21 (95% CI 1.00–
1.55) for COX-2
inhibitor exposed
patients.
(continued)
859

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


860 European Journal of Preventive Cardiology 27(8)

(high dose increased


new users (RR 1.53,

users (more than 30


95% CI 1.37–1.70).
relative risk of vascular events seemed to be similar irre-

risk); no increased
effect was evident
Highest risk among
spective of baseline patient characteristics, after excess

risk for remote


Dose dependency
risk calculation, the authors reported that in patients

days back).
with a low basal cardiovascular risk, the predicted abso-
Comments

lute risk of major vascular events was small, irrespective


of the drug used. In contrast, in patients with a baseline
at higher vascular risk, the absolute risk of developing a

CI: confidence interval; COX-2: cyclooxygenase-2; CV: cardiovascular; HF: heart failure; HR: hazard ratio; OR: odds ratio; RR: risk ratio; SAID: steroidal anti-inflammatory drug.
MI was higher when coxibs or high dose ibuprofen or
RR 1.12 (95% CI 1.06–

2.88) in 15–30 days


diclofenac were used.88 The differences in MI risk
1.76 (95% CI 1.07–
1.18) for atrial fib-

geneity among the

Multiple adjusted HR
moderate hetero-
rillation risk with

among NSAIDs were generally attributed to their


Risk modification

pharmacodynamics, in particular to the specific


COX-2 inhibition potency. A strict correlation between
studies.

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


COX-2 activity inhibition in whole blood and relative
use.

risk of MI was found in NSAID users. When the drugs


used were grouped in high COX-2 inhibition (at least
90% – rofecoxib, diclofenac, indomethacin and piroxi-
cam) and low COX-2 inhibition (<90% – ibuprofen,
meloxicam, celecoxib and etoricoxib) a significant dif-
Endpoint analysed

Atrial fibrillation

Atrial fibrillation

ference in MI risk was found (60% increase in the


former vs 18% in the latter).89 Similarly, the MI risk
for ibuprofen was dose-dependent and related to the
degree of COX-2 inhibition. This study, confirmed
that naproxen is associated with a safer profile showing
no differences when compared to a non at-risk general
population. Overall, among COX-2 selective and non-
5 studies, more than
50 thousand case
Number of patients

selective NSAID users, an increased relative risk of MI


ranging from 30–40% should be taken into account,
8243 patients

although some distinctions need to be made. The


patient

higher the dose, the higher the risk, especially in patients


with pre-existent vascular or cardiac disease; in low car-
diovascular risk patients, the additional risk of low dose
NSAIDs is reasonably marginal and their use relatively
safer.89 Because patients commonly receive both acetyl-
salicylate and ACEis, concerns had already been raised
Anti-inflammatory

about 20 years ago about a putative counteraction


between these two drugs, reducing their beneficial
effects.90 In fact, ACEis increase bradykinin PGI2 and
NSAIDs

NSAIDs
drugs

PGE2, while acetylsalicylate and NSAIDs affect and


blunt relaxant PGs generation. An analysis of this
issue, which combined the data from large clinical
trials on ischemic heart disease patients, did not confirm
this interaction, that is patients benefitted from ACEi
follow-up study
Population-based

effects irrespective of concomitant acetylsalicylate ther-


apy.91 Interestingly too, a retrospective analysis of the
Metanalysis
Study type

Survival of Myocardial Infarction Long-term


Evaluation (SMILE-4) study showed that in hyperten-
sive patients, but not in normotensive ones with acute
MI complicated by left ventricular dysfunction, the
First author and publi-

combination of zofenopril and acetylsalicylate was


Table 2. Continued

Krijthe et al., 2014

superior in terms of preventing major cardiovascular


Liu et al., 2015

outcomes to that of ramipril and acetylsalicylate.92


This observation can reopen the debate about the con-
cation year

cept of class effect. This could suggest that zofenopril, a


sulfhydryl-containing ACEi, might have protective car-
diovascular effects through mechanisms that involve
79

80
Table 3. Association between non-steroidal anti-inflammatory drug (NSAID) use and myocardial infarction in clinical studies.

First author and publi- Anti-inflammatory


cation year Study type drugs Number of patients Endopoint analysed Risk modification Comments
Cabassi et al.

83
Bresalier et al., 2005 Randomised placebo Rofecoxib 2586 patients CV thrombotic Total thombotic events Small number of CV
controlled trial vents HR 1.92 (95% CI events, single
1.19–3.11) in rofecoxib molecule tested.
group. Cardiac events
HR 2.8 (95% CI
1.44–5.45).
84
Levesque et al., 2006 Population-based COX-2 inhibitors 113,927 patients 66 Myocardial Within first 5.8 days of Risk of myocardial
cohort study years or older infarction exposure to rofecoxib infarction decreased
HR 1.7 (95% CI over time for
1.26–2.31). No con- rofecoxib.
sistent data for
celecoxib.
85
Schjerning Olsen Nationwide study in NSAIDs 102,138 patients Death or re- Increased risk of death or Naproxen and short
et al., 2011 myocardial infarction re-infarction in NSAIDs term ibuprofen had
infarction exposed patients (HR the lower risk.
1.45 (95% CI
1.29–1.62) in the first 7
days. Higher risk in
7–14 days of treatment
(HR 1.68 95% CI
1.5–1.88).
86
Ross et al., 2010 Long-term follow-up Rofecoxib 1451 patients Retrospective with RR 2.97 (95% CI 1.2–7.32) Very low number of
of study 078 (cogni- intention to for cardiovascular events and not pre-
tive impairment) treat analysis thromboembolic specified endpoint.
events in rofecoxib
patients.
87
Kearney et al., 2006 Metanalysis 138 Randomised trials Vascular events and HR 1.86 (95% CI Insufficient data for
available on COX-2 myocardial 1.33–2.59) for myocar- determining a dose
inhibitors and infarction dial infarction in coxib- dependency.
NSAIDs exposed patients
versus placebo.
88
Bhala et al., 2013 Metanalysis 280 trials on COX-2 Major coronary Coxib associated to RR Naproxen lower risk
inhibitors and events (fatal and 1.76 (95% CI 1.31– for vascular events
NSAIDs non-fatal myo- 2.37) for myocardial compared to other
cardial infarction . Diclofenac NSAIDs and coxib.
infarction) RR 1.70 (1.19–2.41) for
myocardial infarction.
Ibuprofen RR 2.22
861

(1.10–4.48).
(continued)
Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024
862 European Journal of Preventive Cardiology 27(8)

2 inhibition in vitro.
whole blood COX-
less PGs generation than does ramipril. Appropriate

Relationship between

diclofenac, ibupro-
Increased risk for

fen and rofecoxib


compared to low
relative risk and
and scaled studies are needed to define potential differ-

higher doses of
ences in cardiovascular protective effects within the
same class of ACEis.
Comments

doses.
Heart failure
Non-selective and COX-2 selective NSAIDs lead to
NSAIDS (RR 1.35 95%
MI for current user of

impaired sodium and water excretion, and increased


Overall increased risk of

vascular tone that contribute to an increased risk of


HF. Haemodynamic and renal effects of NSAIDs and
CI 1.23– 1.48).
Risk modification

coxibs can induce the development of new cases,

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


worsen congestion and contribute to rehospitalization
of patients with pre-existing HF. Their continuous use
can also increase the risk of mortality.88 Treatment with
NSAIDs for one week doubles the probability of hos-
pital admission, particularly in the elderly. Higher risk
Endopoint analysed

was observed in patients with a previous HF diagnosis.


Non-fatal myocar-
dial infarction

Risk was greater with increasing doses and use of


long vs short half-life NSAIDs.93 In an interesting
meta-analysis of randomised control trial data invol-
ving subjects not affected by rheumatic disease, an asso-
ciation between NSAID use and HF was found without
differences between non-selective and COX-2 selective
drugs.94 In a nested case-control study including sub-
Myocardial infarction
Number of patients

jects from health-care databases from four European


countries (Germany, Italy, Netherland and UK) it
CI: confidence interval; COX-2: cyclooxygenase-2; CV: cardiovascular; HR: hazard ratio; RR: risk ratio.
case 8852

was confirmed that NSAID users (in the preceding 14


days) had a 19% increased relative risk of a first HF-
related hospital admission compared to past users, and
that this risk was directly proportional to the NSAID
dosage.95 In the same study, patients with or without a
previous history of cardiac disease showed a similar
NSAIDs and COX-2

excessive risk of HF development as age increased.95


Anti-inflammatory

From the pathophysiological point of view, NSAID


inhibitors

use may antagonise the positive effects of renin-angio-


tensin blockade on vascular tone (ACE-I, angiotensin
drugs

II receptor 1 antagonists), minimising the effects of


diuretics on volume congestion in HF patients. In par-
ticular, renal COX-2 inhibition seems to be of major
nested case-control

importance in inducing vasoconstriction, antidiuresis


and antinatriuresis in both animal models of HF and
Data from THIN

in humans.16 Ahmetaj-Shala et al. reported that COX-2


gene deletion in mice, as well as treatment with cele-
Study type

coxib or naproxen in humans, resulted in increased cir-


study

culating levels of asymmetric dimethyl arginine


(ADMA), an endogenous competitive inhibitor of
endothelial nitric oxide synthase, which could explain
First author and publi-

reduced nitric oxide generation and contribution to


Table 3. Continued

Garcı́a Rodrı́guez

increased peripheral vascular resistance, salt and


water retention.96 In a large observational study of
et al., 2008

pre-existing HF patients, a dose-dependent rise in mor-


cation year

tality (ranging from 70 to 108% compared to untreated


patients) was reported for COX-2 selective drugs (such
as celecoxib, rofecoxib and diclofenac) whereas only
89
Cabassi et al. 863

high, but not low doses, of naproxen and ibuprofen Portugal, Spain and Hungary) would appear to be
were associated with a significant increase risk of less toxic than NSAIDs, although a few countries
death (from 22% to 31% vs untreated patients).93 have banned the drug because of a risk of agranulo-
However, in the PRECISION trial, celecoxib, at mod- cytosis.104,105 In 2013, the European League Against
erate doses (200 mg per day for most patients), was not Rheumatism (EULAR) recommended that all patients
associated with a higher cardiovascular risk (including with osteoarticular degenerative conditions should be
hospitalisation for HF) when compared to non-selec- offered an individualised management plan to maintain
tive NSAIDs, naproxen and ibuprofen.61 Even in the physical activity which includes an exercise program
subgroup of patients with pre-existing cardiac disease with aerobic and muscle-strengthening exercises,
treated with low-dose acetylsalicylate, there were no advice on weight loss if necessary, and physiotherapy
statistically significant differences among moderate programmes.106
doses of celecoxib and the non-selective NSAIDs.97

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


Overall, the evidence available indicates that COX-2
Conclusions
selective as well as non-selective NSAIDs should be
avoided in patients with an elevated risk of cardiovas- The widespread use of prescribed and self-prescribed
cular disease and in those with established cardiovascu- NSAIDs may lead to deleterious renal and cardiovas-
lar disease. In all cases, short periods of treatment and cular effects. Through multiple mechanisms related to
low doses should be used. PG inhibition, NSAIDs may increase blood pressure
and blunt the effects of anti-hypertensive drugs,
Pharmacological and non-pharmacological namely ACEis, angiotensin II receptor 1 blockers and
thiazide diuretics.62,63,107 Due to their potential cardio-
alternative therapies to NSAIDs
vascular toxicity, patients with pre-existing vascular or
The weak COX-2 inhibitor acetaminophen is recom- cardiac disease should avoid NSAID use, especially 3–6
mended as initial and useful therapy for osteoarthritic months after an acute event. Hepatic and renal diseases
pain even though long-term therapy with high doses frequently encountered in elderly patients can alter
(up to 4 g in the general population or 3 g in elderly NSAID pharmacokinetics and pharmacodynamics,
patients) may lead to hepatic toxicity. Moreover, thus predisposing them to an increased risk of toxic
high-dose acetaminophen should not be used in com- effects. In particular, elderly patients with prior
bination with acetylsalicylate which, through glutathi- impaired renal function are at greater risk of develop-
one depletion in the kidney cortex and papilla, may ing a wide spectrum of renal dysfunctions. Overall,
favour acetaminophen-induced nephrotoxicity.98 these previous conditions point to NSAIDs as a leading
Some patients report improvement in pain with dietary cause of hospitalization, especially in the elderly.9 The
supplements such as glucosamine/chondroitin, but in need to use the lowest effective dose for a short period
controlled trials glucosamine treatment did not confirm of time has been emphasised by both the US Food and
significant improvement in pain or function.99 Intra- Drug Administration and the European Medicines
articular corticosteroid and hyaluronic acid treatment Agency with regard to NSAID-related thrombotic
proved to be superior to NSAID treatment in knee risk.108,109 We are aware that successful restriction of
osteoarthritis.100 Acupuncture, therapeutic ultrasound their use is arduous. Lifestyle modifications are of over-
and manual manipulation seem to exert moderate pain whelming importance in patients affected by osteoarth-
reduction especially in osteoarthritis; topical diclofenac ritis and rheumatic disease to maintain cardiovascular
or ketoprophen gel and capsaicin (by reducing nocicep- health and reduce cardiovascular risk.110 These changes
tive pain signals to the central nervous system) have not only limit NSAID use but also provide a holistic
shown relative efficacy in several placebo-controlled approach, enabling patients to manage cartilage degen-
trials.101 A number of medicinal plants such as Arnica eration pain, reduce inflammation and prevent the dele-
montana, Boswella serrata, Curcuma longa and terious effects of a sedentary lifestyle. However, when
Harpagophytum procumbens have been shown to signifi- NSAIDs use remains unavoidable, therapy with the
cantly improve pain and functionality in subjects with least toxic drugs (naproxen when there is no concern
knee and hip osteoarthritis.102 Upgrade of treatment to about gastro-duodenal toxicity, as well as moderate
weak opioids (tramadol and codeine) alone or in com- doses of celecoxib or, alternatively, acetaminophen up
bination with acetaminophen or even more powerful to 3 g/day or low-dose ibuprofen) should be
opioids such as fentanyl or hydrocodone may be an chosen.6,89,111 Careful identification of frail at-risk indi-
effective option although risk of addiction has become viduals (e.g. older, HF and chronic renal failure
a worldwide problem in western countries.103 patients) together with close monitoring of side effects
Metamizole/dipyrone (available in Germany, Italy, (namely cardiovascular and renal) are mandatory.
864 European Journal of Preventive Cardiology 27(8)

Declaration of conflicting interests 12. Lezoualc’h F, Fazal L, Laudette M, et al. Cyclic AMP
The author(s) declared no potential conflicts of interest with sensor EPAC proteins and their role in cardiovascular
respect to the research, authorship and/or publication of this function and disease. Circ Res 2016; 118: 881–897.
article. 13. Grosser T, Fries S and FitzGerald GA. Biological basis
for the cardiovascular consequences of COX-2 inhibition:
Therapeutic challenges and opportunities. J Clin Invest
Funding 2006; 116: 4–15.
The author(s) received no financial support for the research, 14. Vane JR, Bakhle YS and Botting RM. Cyclooxygenases 1
authorship and/or publication of this article. and 2. Annu Rev Pharmacol Toxicol 1998; 38: 97–120.
15. Kirkby NS, Chan MV, Zaiss AK, et al. Systematic study
of constitutive cyclooxygenase-2 expression: Role of NF-
References kB and NFAT transcriptional pathways. Proc Natl Acad
1. Wehling M. Non-steroidal anti-inflammatory drug use in Sci U S A 2016; 113: 434–439.

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


chronic pain conditions with special emphasis on the eld- 16. Hörl WH. Nonsteroidal anti-inflammatory drugs and the
erly and patients with relevant comorbidities: kidney. Pharmaceuticals (Basel) 2010; 3: 2291–2321.
Management and mitigation of risks and adverse effects. 17. Ricciotti E, Yu Y, Grosser T, et al. COX-2, the dominant
Eur J Clin Pharmacol 2014; 70: 1159–1172. source of prostacyclin. Proc Natl Acad Sci U S A 2013;
2. Zhou Y, Boudreau DM and Freedman AN. Trends in the 110: E183.
use of aspirin and nonsteroidal anti-inflammatory drugs 18. Kirkby NS, Zaiss AK, Urquhart P, et al. LC-MS/MS
in the general U.S. population. Pharmacoepidemiol Drug confirms that COX-1 drives vascular prostacyclin whilst
Saf 2014; 23: 43–50. gene expression pattern reveals non-vascular sites of
3. Brattwall M, Turan I and Jakobsson J. Musculoskeletal COX-2 expression. PLoS One 2013; 8: e69524.
pain: Prescription of NSAID and weak opioid by primary 19. Murata T, Ushikubi F, Matsuoka T, et al. Altered pain
health care physicians in Sweden 2004–2008 – a retro- perception and inflammatory response in mice lacking
spective patient record review. J Pain Res 2010; 3: prostacyclin receptor. Nature 1997; 388: 678–682.
131–135. 20. Cheng Y, Austin SC, Rocca B, et al. Role of prostacyclin
4. Osmed. The Medicines Utilisation Monitoring Centre. in the cardiovascular response to thromboxane A2.
National report on medicines use in Italy. 2017. Rome: Science 2002; 296: 539–541.
Italian Medicines Agency, 2018. 21. Félétou M, Huang Y and Vanhoutte PM. Endothelium-
5. Kristensen KB, Karlstad O, Martikainen JE, et al. mediated control of vascular tone: COX-1 and COX-2
Nonaspirin nonsteroidal antiinflammatory drug use in products. Br J Pharmacol 2011; 164: 894–912.
the Nordic countries from a cardiovascular risk perspec- 22. Fabre JE, Nguyen M, Athirakul K, et al. Activation of
tive, 2000–2016: A drug utilization study. the murine EP3 receptor for PGE2 inhibits cAMP pro-
Pharmacotherapy 2019; 39: 150–160. duction and promotes platelet aggregation. J Clin Invest
6. Solomon DH, Husni ME, Wolski KE, et al. PRECISION 2001; 107: 603–610.
Trial Investigators. Differences in safety of nonsteroidal 23. Kobayashi T, Tahara Y, Matsumoto M, et al. Roles of
antiinflammatory drugs in patients with osteoarthritis thromboxane A(2) and prostacyclin in the development
and patients with rheumatoid arthritis: A randomized of atherosclerosis in apoE-deficient mice. J Clin Invest
clinical trial. Arthritis Rheumatol 2018; 70: 537–546. 2004; 114: 784–794.
7. Franceschi M, Scarcelli C, Niro V, et al. Prevalence, clin- 24. Langenbach R, Morham SG, Tiano HF, et al.
ical features and avoidability of adverse drug reactions as Prostaglandin synthase 1 gene disruption in mice reduces
cause of admission to a geriatric unit: A prospective study arachidonic acid-induced inflammation and indometha-
of 1756 patients. Drug Saf 2008; 31: 545–556. cin-induced gastric ulceration. Cell 1995; 83: 483–492.
8. Keith SW, Maio V, Dudash K, et al. A physician-focused 25. Szerafin T, Erdei N, Fülöp T, et al. Increased cyclooxy-
intervention to reduce potentially inappropriate medica- genase-2 expression and prostaglandin-mediated dilation
tion prescribing in older people: A 3-year, Italian, pro- in coronary arterioles of patients with diabetes mellitus.
spective, proof-of-concept study. Drugs Aging 2013; 30: Circ Res 2006; 99: e12–e17.
119–127. 26. Fanelli A, Ghisi D, Aprile PL, et al. Cardiovascular and
9. Varga S, Alcusky M, Keith SW, et al. Hospitalization cerebrovascular risk with nonsteroidal anti-inflammatory
rates during potentially inappropriate medication use in drugs and cyclooxygenase 2 inhibitors: Latest evidence
a large population-based cohort of older adults. Br J Clin and clinical implications. Ther Adv Drug Saf 2017; 8:
Pharmacol 2017; 83: 2572–2580. 173–182.
10. Narumiya S and Fitzgerald GA. Genetic and pharmaco- 27. Ghosh R, Alajbegovic A and Gomes AV. NSAIDs and
logical analysis of prostanoid receptor function. J Clin cardiovascular diseases: Role of reactive oxygen species.
Invest 2001; 108: 25–30. Oxid Med Cell Longev 2015; 2015: 536962.
11. Woodward DF, Jones RL and Narumiya S. International 28. Kennedy CR, Zhang Y, Brandon S, et al. Salt-
Union of Basic and Clinical Pharmacology. LXXXIII: sensitive hypertension and reduced fertility in mice lack-
Classification of prostanoid receptors, updating 15 ing the prostaglandin EP2 receptor. Nat Med 1999; 5:
years of progress. Pharmacol Rev 2011; 63: 471–538. 217–220.
Cabassi et al. 865

29. Herrera M, Yang T, Sparks MA, et al. Complex role for 47. Rodrigues AD. Impact of CYP2C9 genotype on pharma-
E-prostanoid 4 receptors in hypertension. J Am Heart cokinetics: Are all cyclooxygenase inhibitors the same?
Assoc 2019; 8: e010745. Drug Metab Dispos 2005; 33: 1567–1575.
30. Mendez M and LaPointe MC. PGE2-induced hypertro- 48. McAlindon TE, Bannuru RR, Sullivan MC, et al.
phy of cardiac myocytes involves EP4 receptor-dependent OARSI guidelines for the non-surgical management of
activation of p42/44 MAPK and EGFR transactivation. knee osteoarthritis. Osteoarthritis Cartilage 2014; 22:
Am J Physiol Heart Circ Physiol 2005; 288: 363–388.
H2111–H2117. 49. Whelton A. Renal aspects of treatment with conventional
31. Xiao CY, Yuhki K, Hara A, et al. PGE2 protects the nonsteroidal anti-inflammatory drugs versus cyclooxy-
heart from ischemia-reperfusion injury via its receptor genase-2-specific inhibitors. Am J Med 2001; 110:
subtype EP4. Circulation 2004; 109: 2462–2468. 33S–42S.
32. Hara A, Yuhki K, Fujino T, et al. Augmented cardiac 50. Zhang X, Donnan PT, Bell S, et al. Non-steroidal anti-
hypertrophy in response to pressure overload in mice inflammatory drug induced acute kidney injury in the

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


lacking the prostaglandin I2 receptor. Circulation 2005; community dwelling general population and people with
112: 84–92. chronic kidney disease: Systematic review and meta-ana-
33. Wang D, Patel VV, Ricciotti E, et al. Cardiomyocyte lysis. BMC Nephrol 2017; 18: 256.
cyclooxygenase-2 influences cardiac rhythm and func- 51. Plantinga L, Grubbs V, Sarkar U, et al. Nonsteroidal
tion. Proc Natl Acad Sci USA 2009; 106: 7548–7552. anti-inflammatory drug use among persons with chronic
34. Patrono C and Dunn MJ. The clinical significance of kidney disease in the United States. Ann Fam Med 2011;
inhibition of renal prostaglandin synthesis. Kidney Int 9: 423.
1987; 32: 1–12. 52. Vega J, Goecke H, Méndez GP, et al. Nephrotic syn-
35. Schnermann J and Briggs JP. Tubular control of renin drome and acute tubular necrosis due to meloxicam
synthesis and secretion. Pflugers Arch 2013; 465: use. Ren Fail 2012; 34: 1344–1347.
39–51. 53. Markowitz GS, Bomback AS and Perazella MA. Drug-
36. Theilig F, Campean V, Paliege A, et al. Epithelial COX-2 induced glomerular disease: Direct cellular injury. Clin J
expression is not regulated by nitric oxide in rodent renal Am Soc Nephrol 2015; 10: 1291–1299.
cortex. Hypertension 2002; 39: 848–853. 54. Mohammed EP and Stevens JM. Recurrence of
37. Harris RC, McKanna JA, Akai Y, et al. Cyclooxygenase- Arthrotec-associated nephrotic syndrome with re-chal-
2 is associated with the macula densa of rat kidney and lenge. Clin Nephrol 2000; 53: 483–485.
increases with salt restriction. J Clin Invest 1994; 94: 55. Praga M, Sevillano A, Auñón P, et al. Changes in the
2504–2510. aetiology, clinical presentation and management of acute
38. Yang T, Endo Y, Huang YG, et al. Renin expression in interstitial nephritis, an increasingly common cause of
COX-2 knockout mice on normal or low-salt diets. Am J acute kidney injury. Nephrol Dial Transplant 2015; 30:
Physiol Renal Physiol 2000; 279: F819–F825. 1472–1479.
39. Komhoff M, Jeck ND, Seyberth HW, et al. 56. De Broe ME and Elseviers MM. Over-the-counter anal-
Cyclooxygenase-2 expression is associated with the gesic use. J Am Soc Nephrol 2009; 20: 2098–2103.
renal macula densa of patients with Bartter-like syn- 57. Nderitu P, Doos L, Jones PW, et al. Non-steroidal anti-
drome. Kidney Int 2000; 58: 2420–2424. inflammatory drugs and chronic kidney disease progres-
40. Olesen ET and Fenton RA. Is there a role for PGE2 in sion: A systematic review. Fam Pract 2013; 30: 247–255.
urinary concentration? J Am Soc Nephrol 2013; 24: 58. Perneger TV, Whelton PK and Klag MJ. Risk of kidney
169–178. failure associated with the use of acetaminophen, aspirin,
41. Perazella MA. Renal vulnerability to drug toxicity. Clin J and nonsteroidal antiinflammatory drugs. N Engl J Med
Am Soc Nephrol 2009; 4: 1275–1283. 1994; 331: 1675–1679.
42. Takahashi K, Schreiner GF, Yamashita K, et al. 59. Ndetan H, Evans Jr MW, Singal AK, et al. Light to
Predominant functional roles for thromboxane A2 and moderate drinking and therapeutic doses of acetamino-
prostaglandin E2 during late nephrotoxic serum glomer- phen: An assessment of risks for renal dysfunction. Prev
ulonephritis in the rat. J Clin Invest 1990; 85: 1974–1982. Med Rep 2018; 12: 253–258.
43. Brune K and Patrignani P. New insights into the use of 60. Agodoa LY, Francis ME and Eggers PW. Association of
currently available non-steroidal anti-inflammatory analgesic use with prevalence of albuminuria and reduced
drugs. J Pain Res 2015; 8: 105–118. GFR in US adults. Am J Kidney Dis 2008; 51: 573–583.
44. Chan TY. Adverse interactions between warfarin and 61. Nissen SE, Yeomans ND, Solomon DH, et al.
nonsteroidal antiinflammatory drugs: Mechanisms, clin- PRECISION Trial Investigators. Cardiovascular safety
ical significance, and avoidance. Ann Pharmacother 1995; of celecoxib, naproxen, or ibuprofen for arthritis.
29: 1274–1283. N Engl J Med 2016; 375: 2519–2529.
45. Ouellet M and Percival MD. Mechanism of acetamino- 62. Walker C and Biasucci LM. Cardiovascular safety of
phen inhibition of cyclooxygenase isoforms. Arch non-steroidal anti-inflammatory drugs revisited.
Biochem Biophys 2001 15; 387: 273–280. Postgrad Med 2018; 130: 55–71.
46. Sorokin A. Nitric oxide synthase and cyclooxygenase 63. Snowden S and Nelson R. The effects of nonsteroidal
pathways: A complex interplay in cellular signaling. anti-inflammatory drugs on blood pressure in hyperten-
Curr Med Chem 2016; 23: 2559–2578. sive patients. Cardiol Rev 2011; 19: 184–191.
866 European Journal of Preventive Cardiology 27(8)

64. Ishiguro C, Fujita T, Omori T, et al. Assessing the effects 79. Liu G, Yan Y-P, Zheng X-X, et al. Meta-analysis of
of non-steroidal anti-inflammatory drugs on antihyper- nonsteroidal anti-inflammatory drug use and risk of
tensive drug therapy using post-marketing surveillance atrial fibrillation. Am J Cardiol 2014; 114: 1523–1529.
database. J Epidemiol 2008; 18: 119–124. 80. Krijthe BP, Heeringa J, Hofman A, et al. Non-steroidal
65. Ruschitzka F, Borer JS, Krum H, et al. Differential blood anti-inflammatory drugs and the risk of atrial fibrillation:
pressure effects of ibuprofen, naproxen, and celecoxib in A population-based follow-up study. BMJ Open 2014; 4:
patients with arthritis: The PRECISION-ABPM e004059.
(Prospective Randomized Evaluation of Celecoxib 81. Sondergaard KB and Gislason G. NSAIDs and cardiac
Integrated Safety Versus Ibuprofen or Naproxen arrest: Non-steroidal anti-inflammatory drug use is asso-
Ambulatory Blood Pressure Measurement) Trial. Eur ciated with increased risk of out-of-hospital cardiac
Heart J 2017; 38: 3282–3292. arrest: A nationwide case-time-control study. Eur Heart
66. Zanchetti A, Hansson L, Leonetti G, et al. Low-dose J 2017; 38: 1788–1789.
aspirin does not interfere with the blood pressure-lower- 82. Bombardier C, Laine L, Reicin A, et al. VIGOR Study

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


ing effects of antihypertensive therapy. J Hypertens 2002; Group. Comparison of upper gastrointestinal toxicity of
20: 1015–1022. rofecoxib and naproxen in patients with rheumatoid arth-
67. Sudano I, Flammer AJ, Périat D, et al. Acetaminophen ritis. VIGOR Study Group. N Engl J Med 2000; 343:
increases blood pressure in patients with coronary artery 1520–1528.
disease. Circulation 2010; 122: 1789–1796. 83. Bresalier RS, Sandler RS, Quan H, et al. Adenomatous
68. Dawson J, Fulton R, McInnes GT, et al. Acetaminophen Polyp Prevention on Vioxx (APPROVe) Trial
use and change in blood pressure in a hypertensive popu- Investigators. Cardiovascular events associated with rofe-
lation. J Hypertens 2013; 31: 1485–1490. coxib in a colorectal adenoma chemoprevention trial. N
69. Chiam E, Weinberg L, Bailey M, et al. The haemo- Engl J Med 2005; 352: 1092–1102.
dynamic effects of intravenous paracetamol (acetamino- 84. Lévesque LE, Brophy JM and Zhang B. Time variations
phen) in healthy volunteers: A double-blind, randomized, in the risk of myocardial infarction among elderly users
triple crossover trial. Br J Clin Pharmacol 2016; 81: of COX-2 inhibitors. CMAJ 2006; 174: 1563–1569.
605–612. 85. Schjerning Olsen AM, Fosbøl EL, Lindhardsen J, et al.
70. Lafrance JP and Miller DR. Dispensed selective and non- Duration of treatment with nonsteroidal anti-inflamma-
selective nonsteroidal anti-inflammatory drugs and the tory drugs and impact on risk of death and recurrent
risk of moderate to severe hyperkalemia: A nested case- myocardial infarction in patients with prior myocardial
control study. Am J Kidney Dis 2012; 60: 82–89. infarction: A nationwide cohort study. Circulation 2011;
71. Ng JL, Morgan DJ, Loh NK, et al. Life-threatening 123: 2226–2235.
hypokalaemia associated with ibuprofen-induced renal 86. Ross JS, Madigan D, Konstam MA, et al. Persistence of
tubular acidosis. Med J Aust 2011; 194: 313–316. cardiovascular risk after rofecoxib discontinuation. Arch
72. Clark BA, Shannon RP, Rosa RM, et al. Increased sus- Intern Med 2010; 170: 2035–2036.
ceptibility to thiazide-induced hyponatremia in the eld- 87. Kearney PM, Baigent C, Godwin J, et al. Do selective
erly. J Am Soc Nephrol 1994; 5: 1106–1111. cyclo-oxygenase-2 inhibitors and traditional non-steroi-
73. Patrono C, Patrignani P and Garcı́a Rodrı́guez LA. dal anti-inflammatory drugs increase the risk of athero-
Cyclooxygenase-selective inhibition of prostanoid forma- thrombosis? Meta-analysis of randomised trials. BMJ
tion: Transducing biochemical selectivity into clinical 2006; 332: 1302–1308.
read-outs. J Clin Invest 2001; 108: 7–13. 88. Bhala N, Emberson J, Merhi A, et al. Coxib and trad-
74. De Caterina R, Ruigomez A and Garcia Rodriguez LA. itional NSAID Trialists’ (CNT) Collaboration. Vascular
Long-term use of anti-inflammatory drugs and risk of and upper gastrointestinal effects of non-steroidal anti-
atrial fibrillation. Arch Intern Med 2010; 170: 1450–1455. inflammatory drugs: Meta-analyses of individual partici-
75. Schmidt M, Christiansen CF, Mehnert F, et al. Non-ster- pant data from randomised trials. Lancet 2013; 382:
oidal anti-inflammatory drug use and risk of atrial fibril- 769–779.
lation or flutter: Population based case-control study. 89. Garcı́a Rodrı́guez LA, Tacconelli S and Patrignani P.
BMJ 2011; 343: d3450. Role of dose potency in the prediction of risk of myocar-
76. Chao T-F, Liu C-J, Chen S-J, et al. The association dial infarction associated with nonsteroidal anti-inflam-
between the use of non-steroidal anti-inflammatory matory drugs in the general population. J Am Coll
drugs and atrial fibrillation: A nationwide case-control Cardiol 2008; 52: 1628–1636.
study. Int J Cardiol 2013; 168: 312–316. 90. Stys T, Lawson WE, Smaldone GC, et al. Does aspirin
77. Bäck M, Yin L and Ingelsson E. Cyclooxygenase-2 attenuate the beneficial effects of angiotensin-converting
inhibitors and cardiovascular risk in a nation-wide enzyme inhibition in heart failure. Arch Intern Med 2000;
cohort study after the withdrawal of rofecoxib. Eur 160: 1409–1413.
Heart J 2012; 33: 1928–1933. 91. Latini R, Tognoni G, Maggioni AP, et al. Clinical effects
78. Schjerning Olsen AM, Fosbøl EL, Pallisgaard J, et al. of early angiotensin-converting enzyme inhibitor treat-
NSAIDs are associated with increased risk of atrial fib- ment for acute myocardial infarction are similar in the
rillation in patients with prior myocardial infarction: A presence and absence of aspirin: Systematic overview of
nationwide study. Eur Heart J Cardiovasc Pharmacother individual data from 96,712 randomized patients.
2015; 1: 107–114. Angiotensin-converting Enzyme Inhibitor Myocardial
Cabassi et al. 867

Infarction Collaborative Group. J Am Coll Cardiol 101. Dadabo J, Fram J and Jayabalan P. Noninterventional
2000; 35: 1801–1807. therapies for the management of knee osteoarthritis.
92. Borghi C, Ambrosioni E, Novo S, et al. SMILE-4 J Knee Surg 2019; 32: 46–54.
Working Party. Comparison between zofenopril and 102. Dragos D, Gilca M, Gaman L, et al. Phytomedicine in
ramipril in combination with acetylsalicylic acid in joint disorders. Nutrients 2017; 9: e70.
patients with left ventricular systolic dysfunction after 103. Jones P, Dalziel SR, Lamdin R, et al. Oral non-steroidal
acute myocardial infarction: Results of a randomized, anti-inflammatory drugs versus other oral analgesic
double-blind, parallel-group, multicenter, European agents for acute soft tissue injury. Cochrane Database
study (SMILE-4). Clin Cardiol 2012; 35: 416–423. Syst Rev 2015; 7: CD007789.
93. Gislason GH, Rasmussen JN, Abildstrom SZ, et al. 104. Kötter T, da Costa BR, Fässler M, et al. Metamizole-
Increased mortality and cardiovascular morbidity asso- associated adverse events: A systematic review and
ciated with use of nonsteroidal anti-inflammatory drugs meta-analysis. PLoS One 2015; 10: e0122918.
in chronic heart failure. Arch Intern Med 2009; 169: 105. Huber M, Andersohn F, Sarganas G, et al. Metamizole-

Downloaded from https://academic.oup.com/eurjpc/article/27/8/850/5924934 by guest on 09 March 2024


141–149. induced agranulocytosis revisited: Results from the pro-
94. Scott PA, Kingsley GH and Scott DL. Non-steroidal spective Berlin Case-Control Surveillance Study. Eur J
anti-inflammatory drugs and cardiac failure: Meta-ana- Clin Pharmacol 2015; 71: 219–227.
lyses of observational studies and randomised controlled 106. Fernandes L, Hagen KB, Bijlsma JW, et al. EULAR
trials. Eur J Heart Fail 2008; 10: 1102–1107. recommendations for the nonpharmacological core
95. Arfè A, Scotti L, Varas-Lorenzo C, et al. Safety of Non- management of hip and knee osteoarthritis. Ann
steroidal Anti-inflammatory Drugs (SOS) Project Rheum Dis 2013; 72: 1125–1135.
Consortium. Non-steroidal anti-inflammatory drugs 107. Floor-Schreudering A, De Smet PA, Buurma H, et al.
and risk of heart failure in four European countries: NSAID-antihypertensive drug interactions: Which out-
Nested case-control study. BMJ 2016; 354: i4857. patients are at risk for a rise in systolic blood pressure?.
96. Ahmetaj-Shala B, Kirkby NS, Knowles R, et al. Eur J Prev Cardiol 2015; 22: 91–99.
Evidence that links loss of cyclooxygenase-2 with 108. US Food and Drug Administration. FDA Drug Safety
increased asymmetric dimethylarginine: Novel explan- Communication: FDA strengthens warning that non-
ation of cardiovascular side effects associated with aspirin nonsteroidal anti-inflammatory drugs
anti-inflammatory drugs. Circulation 2015; 131: (NSAIDs) can cause heart attacks or strokes, http://
633–642. www.fda.gov/Drugs/DrugSafety/ucm451800.htm (2015,
97. Reed GW, Abdallah MS, Shao M, et al. Effect of aspirin accessed 10 July 2018).
coadministration on the safety of celecoxib, naproxen, 109. European Medicines Agency. Assessment report for
or ibuprofen. J Am Coll Cardiol 2018; 71: 1741–1751. non-steroidal anti-inflammatory drugs (NSAIDs) and
98. Sampathkumar K, Rajiv A and Sampathkumar D. cardiovascular risk, www.ema.europa.eu/docs/en_GB/
Analgesic nephropathy–a painful progression. Clin document_library/Report/2012/11/WC500134717.pdf
Med Insights Urol 2016; 9: 7–10. (2012, accessed 10 July 2018).
99. Ogata T, Ideno Y, Akai M, et al. Effects of glucosamine 110. Piepoli MF and Villani GQ. Lifestyle modification in
in patients with osteoarthritis of the knee: A systematic secondary prevention. Eur J Prev Cardiol 2017; 24:
review and meta-analysis. Clin Rheumatol 2018; 37: 101–107.
2479–2487. 111. Schmidt M, Lamberts M, Olsen AM, et al.
100. Bannuru RR, Schmid CH, Kent DM, et al. Cardiovascular safety of non-aspirin non-steroidal
Comparative effectiveness of pharmacologic interven- anti-inflammatory drugs: Review and position paper
tions for knee osteoarthritis: A systematic review and by the working group for Cardiovascular
network meta-analysis. Ann Intern Med 2015; 162: Pharmacotherapy of the European Society of
46–54. Cardiology. Eur Heart J 2016; 37: 1015–1023.

You might also like