Elsevier

Pharmacological Reports

Volume 62, Issue 3, May–June 2010, Pages 530-535
Pharmacological Reports

NSAIDs and cardiovascular disease: transducing human pharmacology results into clinical read-outs in the general population

https://doi.org/10.1016/S1734-1140(10)70310-8Get rights and content

Abstract

Traditional (t) non-steroidal anti-inflammatory drugs (NSAIDs) and selective cyclooxygenase (COX)-2 inhibitors (coxibs) are important and efficacious drugs for the management of musculoskeletal symptoms. These drugs have both beneficial and adverse effects due to the inhibition of prostanoids. Although the tNSAID and coxib inhibition of COX-2-dependent prostaglandin (PG)E2 production is effective in ameliorating symptoms of inflammation and pain, a small but consistent increased risk of myocardial infarction has been detected in association with their use. Convincing evidence suggests that cardiovascular toxicity associated with the administration of these compounds occurs through a common mechanism involving inhibition of COX-2-dependent prostacyclin. The development of biomarkers that predict the impact of NSAIDs on COX-1 and COX-2 activities in vitro, ex vivo and in vivo has been essential to read-out the clinical consequences of the varying degrees of inhibition of the two COX-isozymes in humans. Whole blood assays for COX-1 and COX-2 might be candidates as surrogate end-points of toxicity and efficacy of NSAIDs. Using a biomarker strategy, we have shown that the degree of inhibition of COX-2 and the functional selectivity with which it is achieved are relevant to the level of cardiovascular hazard from NSAIDs and relate to drug potency (exposure). We propose that the assessment of COX-2 in whole blood ex vivo, either alone or in combination with urinary levels of 2,3–dmor-6–keto-PGF1a a biomarker of prostacyclin biosynthesis in vivo, may represent a valid surrogate end-point to predict cardiovascular risk for functionally selective COX-2 inhibitors.

Introduction

Non-steroidal anti-inflammatory drugs (NSAIDs) are an important and efficacious class of drugs for the management of musculoskeletal symptoms. These compounds cause both beneficial and adverse effects due to the inhibition of prostanoids [12, 37]. Prostanoids are biologically active derivatives of arachidonic acid (AA) released from membrane phospholipids by phospholipases [14]. AA is transformed into prostaglandin (PG)H2, through the activity of cyclooxygenase (COX) enzymes (i.e., COX-1 and COX-2). PGH2 is subsequently metabolized by terminal synthases into the biologically active prostanoids, such as prostacyclin (PGI2), PGD2, PGF, PGE2 and thromboxane (TX)A2 [14, 17]. In particular, the isomeriza-tion of PGH2 to PGE2 is catalyzed by three different isomerases: a cytosolic PGE synthase (cPGES) and two membrane-bound PGESs, mPGES-1 and mPGES-2 [22]. Of these isomerases, cPGES and mPGES-2 are constitutive enzymes whereas mPGES-1 is mainly an induced isoform.

COX-1 and COX-2 have the same catalytic activities: cyclooxygenase and peroxidase [34, 38]. However, the two isoforms of COX are the products of different genes. The COX-1 gene is considered a “housekeeping gene” by virtue of the constitutively low levels of expression in most cell types. In contrast, the gene for COX-2 is a primary response gene with multiple regulatory sites. COX-2 expression can be rapidly induced by bacterial endotoxin (LPS), cytokines (such as interleukin (IL)-1β and tumor necrosis factor (TNF)- α), growth factors, and the tumor promoter phorbol myristate acetate (PMA) [19]. COX-1-dependent prostanoids play an essential homeostatic role in gastrointestinal (GI) cytoprotection [20, 30, 39], while COX-2-dependent prostanoids play dominant roles in pathophysiologic processes, such as inflammation [6, 9]. Several lines of evidence suggest that the main mechanism of action for traditional (t)NSAIDs and NSAIDs selective for COX-2 (named coxibs), used for the treatment of pain and inflammatory joint disease, is the inhibition of COX-2-dependent PGE2 [11]. Inhibition of constitutively expressed COX-1 in the GI tract and presumably in platelets by tNSAIDs seems to play a role in the increased risk of upper GI bleeding/perforation [30]. Proof of concept of this hypothesis came from the finding that reduced incidence of serious GI adverse effects has been shown for two COX-2 inhibitors, such as rofecoxib and lumiracoxib, when compared to tNSAIDs in large randomized clinical trials (RCTs) [3, 33].

Although the inhibition of COX-2-dependent PGE2 production by tNSAIDs and coxibs is effective in ameliorating symptoms of inflammation and pain, a small but consistent increased risk of myocardial infarction (MI) has been detected in NSAID users [15, 16, 26]. Increased incidences of thrombotic events have been detected in placebo-controlled trials involving the COX-2 inhibitors celecoxib, rofecoxib and valdecoxib [4, 24, 25, 35]. However, the results of observational studies and a meta-analysis of data derived from trials with coxibs have shown that the cardiovascular hazard is also related to some tNSAIDs, such as diclofenac [15, 18, 21]. Convincing evidence suggests that cardiovascular toxicity associated with the administration of NSAIDs selective for COX-2 and some tNSAIDs occurs through a common mechanism involving the inhibition of COX-2-dependent prostacyclin.

Section snippets

Pharmacology of NSAIDs

The biochemical selectivity of COX inhibitors for COX-2 has been assessed in the past using different in vitro assays. These experiments yielded variable results depending on the assay used. In fact, several variables may affect the heterogeneity of the results, including the concentrations of exogenous AA, the lack of plasma proteins, and different types of isolated enzyme [5]. To overcome the variability associated with these assays, the human whole blood assays were developed. They are

Cardiovascular hazard of NSAIDs

It has recently been shown that patients taking NSAIDs, both coxibs and tNSAIDs, had a 35% increased risk of non-fatal MI [relative risk 1.35, (95% CI), 1.23 to 1.48] [15]. This elevation of risk increased with increasing treatment duration and daily dose. Using a translational medicine approach, from proof of concept in cells and experimental animals to studies in humans, it was possible to show that the inhibition of COX-2-dependent prostacyclin is the most plausible mechanism for the

Perspectives

The development of biomarkers capable of predicting the impact of NSAIDs on COX-1 and COX-2 activities in vitro, ex vivo and in vivo has been essential to understanding the clinical consequences of varying degree of inhibition of COX-isozymes in humans.

Whole blood assays for COX-1 and COX-2 might be candidates for monitoring levels of toxicity and efficacy of NSAIDs. Using a biomarker strategy, we have shown that, within the group of functionally COX-2 selective NSAIDs with respect to platelets

Acknowledgments

We would like to thank Dr. Licia Totani, Dr. Virgilio Evangelista, Dr. Luigia Di Francesco and Dr. Annalisa Bruno for their invaluable contributions to the results discussed in this review.

References (39)

  • Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial infarction, and stroke in high risk patients

    BMJ

    (2002)
  • E. Arehart et al.

    Acceleration of cardiovascular disease by a dysfunctional prostacyclin receptor mutation: potential implications for cyclooxygenase-2 inhibition

    Circ Res

    (2008)
  • C. Bombardier et al.

    Comparison of upper gastrointestinal toxicity of rofecoxib and naproxen in patients with rheumatoid arthritis. VIGOR Study Group

    N Engl J Med

    (2000)
  • R.S. Bresalier et al.

    Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial

    N Engl J Med

    (2005)
  • P. Brooks et al.

    Interpreting the clinical significance of the differential inhibition of cyclooxygenase-1 and cyclooxygenase-2

    Rheumatology

    (1999)
  • Y. Cao et al.

    Many actions of cyclooxygenase-2 in cellular dynamics and in cancer

    J Cell Physiol

    (2002)
  • M.L. Capone et al.

    Clinical pharmacology of platelet, monocyte, and vascular cyclooxygenase inhibition by naproxen and low-dose aspirin in healthy subjects

    Circulation

    (2004)
  • Y. Cheng et al.

    Cyclooxygenases, microsomal prostaglandin E synthase-1, and cardiovascular function

    J Clin Invest

    (2006)
  • L.G. Crofford

    COX-1 and COX-2 tissue expression: implications and predictions

    J Rheumatol

    (1997)
  • Cited by (37)

    • Investigation of the anti-inflammatory and analgesic activities of promising pyrazole derivative

      2022, European Journal of Pharmaceutical Sciences
      Citation Excerpt :

      Gastrointestinal erosions, ulcerations and bleeding have, for long, been the most commonly reported ADRs, with a 15–30% incidence rate of peptic ulcers among NSAID users (Butt et al., 1988; SE Burke and FitzGerald, 2006). Furthermore, cardiovascular complications of NSAIDS were reported to be correlated with doses used and potency of cyclooxygenase 2 (COX-2) inhibition of individual agents (García Rodríguez et al., 2008; Capone et al., 2010). Attempts of preserving the analgesic and anti-inflammatory effects of NSAIDs while eliminating ADRs have led to the development of selective COX-2 inhibitors.

    • Dual cyclooxygenase-fatty acid amide hydrolase inhibitor exploits novel binding interactions in the cyclooxygenase active site

      2018, Journal of Biological Chemistry
      Citation Excerpt :

      This problem was addressed by the development of COX-2–selective inhibitors that preserved COX-1–dependent gastrointestinal PGE2 production. These drugs exhibit good anti-inflammatory activity with reduced gastrointestinal side effects; however, subsequent clinical trials uncovered adverse cardiovascular side effects associated with inhibition of COX-2 (10, 11). An alternative approach to NSAID-mediated gastrointestinal toxicity is to increase the levels of AEA and other fatty acyl ethanolamides (FAEs), such as palmitoylethanolamide, in individuals receiving NSAID therapy.

    • Modulation by NADPH oxidase of the chronic cardiovascular and autonomic interaction between cyclosporine and NSAIDs in female rats

      2017, European Journal of Pharmacology
      Citation Excerpt :

      A review of the available safety data demonstrated that diclofenac use was associated with the highest cardiovascular risk among nonselective NSAIDs (Farkouh and Greenberg, 2009). The inhibition of COX2-mediated prostacyclin synthesis is proposed as the cause for the poor cardiovascular outcomes (Capone et al., 2010). In addition, prostaglandin F-2α, a product of COX activity, contributes to myocardial fibrosis in a manner that is dependent on ROCK activity (Ding et al., 2012) offering a potential for crosstalk between CSA- and NSAID/COX inhibition-mediated cardiotoxicity.

    View all citing articles on Scopus
    View full text