Skip to main content

MINI REVIEW article

Front. Pharmacol., 21 June 2021
Sec. Pharmacogenetics and Pharmacogenomics
This article is part of the Research Topic NSAIDs Pharmacogenomics View all 9 articles

Pharmacogenomics of NSAID-Induced Upper Gastrointestinal Toxicity

  • Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, United Kingdom

Non-steroidal anti-inflammatory drugs (NSAIDs) are a group of drugs which are widely used globally for the treatment of pain and inflammation, and in the case of aspirin, for secondary prevention of cardiovascular disease. Chronic non-steroidal anti-inflammatory drug use is associated with potentially serious upper gastrointestinal adverse drug reactions (ADRs) including peptic ulcer disease and gastrointestinal bleeding. A few clinical and genetic predisposing factors have been identified; however, genetic data are contradictory. Further research is needed to identify clinically relevant genetic and non-genetic markers predisposing to NSAID-induced peptic ulceration.

Introduction

NSAIDs comprise a heterogeneous group of non-opioid drugs with effective analgesic, anti-inflammatory and antipyretic properties (Calatayud and Esplugues, 2016). They are employed in the treatment of acute and chronic pain conditions characterized by inflammation. While aspirin was used as an analgesic in the past, its main use nowadays is as an antithrombotic particularly at low doses, and for cancer prevention (Shaheen et al., 2002; Sandler et al., 2003; Bashir et al., 2019). Overall, NSAIDs are well tolerated, especially when used short-term (Ong et al., 2007), but because of the enormous usage globally, they are often implicated in adverse drug reactions.

NSAIDs, including low dose aspirin (LDA), are one of the most commonly prescribed classes of medication, accounting for approximately 5–10% of prescriptions globally (Onder et al., 2004). Two decades ago, >30 million people were estimated to take NSAIDs daily (Singh and Triadafilopoulos, 1999). Pharmacoepidemiological studies indicate that NSAID use is increasing. A 2010 National Health Interview Survey (CDC, United States) reported increases of 57 and 41% in aspirin and NSAID use, respectively, over 5 years (Zhou et al., 2014), but this may still be an underestimate given the wide availability of aspirin and ibuprofen as over-the-counter (OTC) formulations. Telephone surveys of United States OTC NSAID users found that drugs were often used/taken inappropriately with 26% of respondents exceeding recommended doses (Goldstein and Lefkowith, 1998; Wilcox et al., 2005; Goldstein and Cryer, 2015). In Germany, analgesic use significantly increased from 19.2% in 1998 to 21.4% in 2008–2011. This rise was found to be attributed exclusively to the use of OTC formulations increasing from 10.0 to 12.2% (prescribed analgesic use remained constant at 7.9%). Ibuprofen was most commonly used, followed by aspirin (Sarganas and et al., 2015). Higher frequencies of ibuprofen use have also been documented in Denmark (Olsen and et al., 2011) and Spain (Gómez-Acebo and et al., 2018). In contrast, however, diclofenac was reported as the most frequently used NSAID (followed by ibuprofen) in a study across 15 countries (Australia, Bangladesh, Canada, China, China (Hong Kong), England, Indonesia, Malaysia, New Zealand, Pakistan, Philippines, Singapore, Taiwan, Thailand, and Vietnam) (McGettigan and Henry, 2013). The trend for increasing OTC analgesic use has also been echoed in Australia. Between 2001 and 2009, there was a 15% increase in the use of ibuprofen, naproxen and diclofenac (Stosic et al., 2011). In general practice, NSAID use by the elderly is prolific, reported at 96% (96% in males, 96.7% in females) in patients >65 years (Pilotto et al., 2003).

NSAIDs are responsible for ∼30% of ADR hospitalisations (Pirmohamed et al., 2004); cardiovascular, gastrointestinal (GI) and renal complications are associated with their use (Bhala et al., 2013; Szeto et al., 2020). Estimates suggest that 5,000–16,500 deaths in the United States and 400–1,000 deaths in the United Kingdom are a direct consequence of NSAID-induced upper GI ulceration and bleeding annually (Wolfe et al., 1999; Langman, 2001; Hawkey and Langman, 2003).

Genetic factors predisposing to NSAID-induced upper GI toxicity have been described, yet findings have been inconsistent and contradictory. This mini review discusses current literature and seeks to identify areas to focus collaborative efforts in the field.

NSAID-Induced Upper Gastrointestinal Toxicity

The first serious NSAID-induced adverse event to be identified was upper GI injury (Walt et al., 1986; Langman, 1988). It is also recognized as one of the most predominant ADRs in the United States (Butt et al., 1988; Fries, 1996). NSAIDs and aspirin have now overtaken Helicobacter pylori as the principal cause of GI toxicity in western countries (Musumba et al., 2009; Bjarnason, 2013). “Gastrointestinal toxicity” refers to a collection of heterogeneous pathologies affecting various tissues and organs of the GI tract.

Clinical Problem and Disease Burden

NSAID-related ADRs range from mild to severe and can result in death. Cryer (2004) conveniently stratified typical manifestations of NSAID-induced GI injury in to three tiers: dyspepsia; asymptomatic ulceration; and more serious complications (GI bleeding, perforation, obstruction, symptomatic ulceration) (Cryer, 2004). Although upper GI events are more common, the entirety of the GI tract may be affected (Maiden et al., 2005). Lower GI complications are not as well defined (Langman et al., 1985). In the large intestine, non-specific colitis, increased gut permeability, malabsorption and bleeding have been reported. Inflammatory bowel disease (IBD) (Bjorkman, 1998; Faucheron, 1999; Forrest et al., 1999) and diverticular disease (Campbell and Steele, 1991) may also be exacerbated by NSAIDs. In this article, we only focus on upper GI events.

Patient characteristics increasing the risk of NSAID-induced GI events have been identified (summarized in Table 1). These include: advanced age (Fries et al., 1991; Hernández-Díaz and Rodríguez, 2000; Russell, 2001; Sostres et al., 2013; Chi et al., 2018); H. pylori infection (Leontiadis and et al., 2007; Sostres et al., 2010); multimorbidity/comorbidity (Chi et al., 2018; Jankovic et al., 2009; Weil and et al., 2000; Kim, 2015); polypharmacy (Davies and Wallace, 1996) and concomitant medications (de Abajo et al., 1999; Silverstein et al., 2000; Sorensen et al., 2000; Garcia Rodriguez and Hernández-Díaz, 2001; Johnsen et al., 2001; Lazzaroni and Bianchi Porro, 2001; Laine et al., 2002; de Jong et al., 2003; Helin-Salmivaara et al., 2007; Lanas et al., 2007; Åhsberg et al., 2010; Masclee et al., 2013; Sostres et al., 2013; Anglin et al., 2014; Olsen et al., 2020). Studies have shown that the risk of NSAID-induced GI complications is dose-dependent (Silverstein and et al., 1995; Bombardier et al., 2000; Laporte et al., 2004; González-Pérez and et al., 2014; Figueiras and et al., 2016) and remains linear over time (Silverstein and et al., 1995; Bombardier et al., 2000; Rostom et al., 2007; Goldstein et al., 2011).

TABLE 1
www.frontiersin.org

TABLE 1. Reported risk factors for developing NSAID-induced upper GI toxicity.

Increased/prolonged exposure (habitual in chronic pain conditions through high-dose and multiple-NSAID use) elevates the risk and/or severity of toxicity (Garcia Rodriguez and Hernández-Díaz, 2001; Bhala et al., 2013; Lanas et al., 2015). Treatment guidelines recommend the lowest effective dose for the shortest period of time. Whilst it is recommended that long-term NSAID use should be avoided (Bhatt et al., 2008; Lanza et al., 2009), this is difficult in practice for many patients. Individuals with a past history of GI injury (including uncomplicated or complicated ulcers) are considered at the highest risk of complications (Sostres et al., 2013; Chi et al., 2018; Silverstein and et al., 1995; Van Hecken et al., 2000; Laine, 2001; Laine, 2006). COX-2 inhibitors (Coxibs) and gastroprotective therapies such as proton pump inhibitors (effective anti-secretory agents) help to mitigate the risk of GI injury (Gargallo et al., 2014; Melcarne et al., 2016).

Mild upper GI symptoms are reported by up to 40% of NSAID users (Larkai et al., 1989; Hirschowitz, 1994). The most clinically significant upper GI NSAID-induced ADRs are symptomatic and/or complicated peptic ulcer. Symptomatic peptic ulcer incidence rates in chronic NSAID users are between 2–4% annually (1–3% for serious ulcer/upper GI complications) (Cryer, 2004; Silverstein et al., 2000; Silverstein and et al., 1995; Bombardier et al., 2000; Blower and et al., 1997; Paulus, 1988), a threefold to fivefold increase compared to non-users (García Rodríguez and Jick, 1994; Gutthann et al., 1997; Hernández-Díaz and Rodríguez, 2000; Cryer, 2004; Gevers et al., 2014).

It is important to note that symptoms do not necessarily correlate with the severity of peptic ulcer disease and/or its complications (Sostres et al., 2013)—50–60% of patients can be asymptomatic (including having unremarkable endoscopies (McCarthy, 1989)) prior to developing potentially fatal NSAID-induced complicated ulcers (Armstrong and Blower, 1987). A prospective observational study found this figure to be as high as 80% (Singh and et al., 1996). In NSAID users who do develop lesions (endoscopically confirmed subepithelial hemorrhages, erosions, and ulcers), healing often occurs before symptoms manifest—“silent ulceration”—because of efficient gastroprotective mechanisms (Larkai et al., 1987; Lanas and Hunt, 2006; Sostres et al., 2013; Carr et al., 2017). Notwithstanding, peptic ulcer prevalence ranges from 15–40% in chronic NSAID users (McCarthy, 1989; Geis et al., 1991; Dajani and Agrawal, 1995; McCarthy, 1998). Gastric ulcer risk is slightly higher than duodenal ulcer risk (McCarthy, 1989; Larkai et al., 1987; McCarthy, 1998; Henry et al., 1993; Gabriel et al., 1991; Griffin and et al., 1991). Endoscopy remains the gold standard for PUD diagnosis (Dunlap and Patterson, 2019).

LDA is extensively used as prophylactic therapy against cardiovascular thrombotic events in high-risk individuals (Collaboration., 2002; Patrono et al., 2004). Throughout Europe and the United States, approximately 23% of the population use aspirin at least once per week (Larson et al., 2005). A prospective analysis of ADRs requiring hospitalization found that aspirin was the most commonly implicated, responsible for 18% of admissions; 74% of these patients were taking LDA. GI bleeding was the most common adverse event, observed in 72% of aspirin-related admissions (Pirmohamed et al., 2004). This is most likely to be due to aspirin-associated peptic ulceration (Niv et al., 2005). Generally, individuals on LDA therapy are older and multimorbid, increasing the likelihood of polypharmacy; some patients take both aspirin and NSAID(s) (Murray and et al., 1998) which is known to further raise the risk of GI toxicity and bleeding (Silverstein et al., 2000; Sorensen et al., 2000; Garcia Rodriguez and Hernández-Díaz, 2001). GI symptoms in chronic LDA therapy are challenging and are a reason for discontinuation of treatment and non/poor compliance (Cayla et al., 2012). Adverse events have been cited as responsible for LDA-discontinuation in almost 50% of patients (Herlitz et al., 2010). Discontinuation is associated with increased risk of new CV events (Biondi-Zoccai et al., 2006; Sostres and Lanas, 2011) and should only be advocated after sound clinical assessment when the risk of GI bleeding outweighs that of CV events.

NSAIDs differ in their propensity to cause upper GI injury based on their COX-1/COX-2 selectivity. Due to selective COX-2 inhibition, coxibs offer greater GI safety and are associated with a lower risk of upper GI injury and clinically significant ulcer complications compared to traditional NSAIDs (tNSAIDs) (Rostom et al., 2007; Chan et al., 2010). A meta-analysis by Castellsague et al. (2012) calculated the relative risk (RR) of upper GI complications as follows:

• RR <2 aceclofenac, celecoxib, ibuprofen.

• RR 2 to <4 diclofenac, ketoprofen, meloxicam, nimesulide, rofecoxib, sulindac.

• RR 4—5 diflunisal, indomethacin, naproxen, tenoxicam.

• RR >5 azapropazone, ketorolac, piroxicam (Castellsague et al., 2012).

Celecoxib has been shown to be associated with a lower risk of upper-GI events (Silverstein et al., 2000; García-Rayado et al., 2018) whilst celecoxib plus proton pump inhibitor is preferred over tNSAID (naproxen) plus proton pump inhibitor to reduce the risk of recurrent upper GI bleeding in individuals requiring concomitant aspirin and NSAID (Chan et al., 2017).

Mechanisms of NSAID-Induced Gastrointestinal Toxicity

NSAID pharmacology, first described by Vane. (1971), is well documented (Vane, 1971). The cyclo-oxygenase (COX) pathway is responsible for the biosynthesis of prostanoids, including prostaglandins (PGs) and thromboxane (TX), from arachidonic acid (AA). Two clinically significant COX isoenzymes exist, COX-1 and COX-2, encoded by the genes PTGS1 and PTGS2, respectively (Plaza-Serón et al., 2018). Prostanoids are pro-inflammatory mediators: PGs produced by COX-1 (and to a lesser degree, COX-2 (Wallace, 2008)) are essential in maintaining/regulating gastroprotection (Dickman and Ellershaw, 2004). Fundamentally, PG generation is forestalled by NSAIDs via reversible inhibition (except aspirin, an irreversible inhibitor (Funk et al., 1991; Catella-Lawson et al., 2001)) of COX isoenzymes, thereby decreasing inflammation.

NSAIDs are sub-divided based on COX-selectivity: tNSAIDs are non-selective, inhibiting both isoenzymes. COX-2 selective inhibitors (coxibs) are isoform-specific, designed to help mitigate the GI adverse events of tNSAIDs whilst retaining anti-inflammatory and analgesic activity. Inhibition of COX-2 is the desired therapeutic aim of NSAIDs. Upper GI toxicity is more common with tNSAIDs than with coxibs (Pilotto et al., 2005).

Under normal conditions, gut homeostasis is maintained. However, imbalance(s) in the gastroduodenal mucosal lining may result in GI injury. Protective factors comprise the gastric epithelial cells and hydrophobic mucus-bicarbonate bilayer, cellular repair, remodeling, restitution, and adequate blood supply–all of these are PG-regulated. Microvascular damage reduces gastric mucosal blood flow, an initial and crucial event in ulcer pathogenesis (Musumba et al., 2009; Bjarnason et al., 2018). The locale of greatest localized ischemia correlates with the site of most NSAID-induced ulceration, the gastric antrum (Musumba et al., 2009). Gastroprotective strategies can mitigate the risk of PUD, but will not be discussed further in this review: Wallace. (2008), Musumba et al. (2009), Bjarnason et al. (2018) provide excellent analyses (Wallace, 2008; Musumba et al., 2009; Bjarnason et al., 2018).

The pathogenesis of NSAID-induced GI injury is complex and multifactorial and can be divided into two broad areas: 1) Topical mechanisms (direct damage to GI mucosa). 2) Systemic mechanisms (COX-inhibition mediated).

1) Topical effects—NSAIDs can cause disturbances in the gastric mucosal epithelium initially through the development of erosions. NSAID toxicity is dependent on their physiochemical properties as lipid-soluble weak organic acids (pKa 3–5) (Brune et al., 1976; Brune and Graf, 1978; Rainsford and Whitehouse, 1980; Musumba et al., 2009; Bjarnason et al., 2018). Detergent properties allow interaction with surface membrane phospholipids, disrupting the mucosal barrier and provoking superficial injury (Lichtenberger, 2001). This permits NSAID to move from the lumen (low gastric pH measuring 1.5–3.5) into epithelial cells (pH neutral, 6.5–7.0), kickstarting disruption of the cellular metabolic pathways culminating in dysfunction, cytotoxic events and apoptotic pathway activation (Musumba et al., 2009; Handa et al., 2014).

2) Systemic effects—Robust gastric mucosal defence/repair is heavily dependent upon perpetual synthesis of COX-derived prostanoids. COX-1 is expressed in most tissues performing “housekeeping duties,” maintaining and regulating various physiological functions. In the GI tract, COX-1 is abundant, producing PGs and TXs involved in cytoprotection. COX-2 is expressed at low levels in the intact stomach, but rapidly induced by COX-1 inhibition or inflammatory stimuli/injury, and produced in vast quantities by cytokines and hormones (Rainsford, 2007; Ricciotti and FitzGerald, 2011; Wongrakpanich et al., 2018).

Classically, GI complications were primarily attributed to COX inhibition: decreased PG levels resulting in increased gastric acid secretion, suppressed mucus and bicarbonate secretion, decreased mucosal blood flow and decreased cell proliferation (Cohen, 1987; Wallace, 1992; Wallace, 2008) resulting in compromised mucosal defense and delayed healing. Selective COX-1 inhibition leads to suppression of mucosal PG production by 95–98% without observable inflammation or ulceration (Ligumsky et al., 1983; Ligumsky et al., 1990; Langenbach et al., 1995; Sigthorsson et al., 2002). Inhibition of COX-2 has yielded similar results (Morham et al., 1995; Sigthorsson et al., 2002). However dual inhibition of both isoenzymes by NSAIDs induces severe gastric lesions (Wallace et al., 2000; Gretzer et al., 2001; Tanaka and et al., 2001). Both COX isoenzymes are also important in ulcer healing (Tanaka et al., 2002; Schmassmann et al., 2006; To et al., 2001; Bhandari et al., 2005; Starodub et al., 2008; Chan and et al., 2005).

In isolation, topical mechanisms are unlikely to induce notable GI toxicity. In combination with systemic factors, however, NSAIDs provoke an imbalance that incites mucosal injury. Cellular damage translates into tissue damage. COX-1 inhibition dramatically suppresses gastroprotection leading to microvascular damage, ischemia, and decreased mucus production. A complex interplay between factors ignites a cascade, involving many mechanisms that disrupt gastroprotection, engendering ulcerogenic conditions.

Genetic Risk Factors

It is well acknowledged that interindividual variability exists in response to drugs, including NSAIDs, which may at least be genetic in origin. Using conventional dosing regimens in analgesia, for example, some individuals will have inadequate pain control while others will encounter toxicity from the same dose (Kapur et al., 2014). Genetic variants affecting treatment outcomes can be categorized into two broad types: 1) genes affecting drug pharmacokinetics (PK), and 2) genes affecting drug pharmacodynamics (PD) (Stamer et al., 2010; Kapur et al., 2014). It should however be noted that given the different mechanisms of action and metabolic pathways, the PK and PD genetic variability cannot be generalized across all NSAID classes. Table 2 summarizes notable genetic risk factors for NSAID-induced upper GI toxicity.

TABLE 2
www.frontiersin.org

TABLE 2. Notable genetic associations in NSAID-induced upper GI toxicity.

Pharmacokinetic-Related Associations

Cytochrome P450 (CYP) gene polymorphisms are strongly associated with adverse drug reactions in general (Johansson and Ingelman-Sundberg, 2011; Sim et al., 2013). Collectively, CYP isoenzymes mediate ∼80% of phase I metabolism of clinically relevant drugs (Evans and Relling, 1999; Eichelbaum et al., 2006; Petrović et al., 2020), including NSAIDs (Blanco et al., 2005). Phase I oxidative metabolism precedes phase II conjugative metabolism (glucuronidation) driven by uridine 5′-diphosphate glucuronosyltransferases (UDP-glucuronosyltransferase, UGTs) (Kuehl et al., 2005; Monrad et al., 2014; Sandson, 2015), supplemented by sulfate conjugation (sulfotransferases) (Ulrich et al., 2006; Loetsch and Oertel, 2013). NSAID metabolism varies and this can be due to CYP polymorphisms, which have been implicated in NSAID-induced ADRs including PUD and UGIB. Studies have also demonstrated that GI damage is dose-dependent (Laporte et al., 2004; Figueiras and et al., 2016).

NSAID metabolism is specifically associated with the CYP2C subfamily (Ingelman-Sundberg et al., 2007; Musumba et al., 2013) (including the highly polymorphic CYP2C8, CYP2C9, and CYP2C19 isoforms), which make up 20% of hepatic CYP450 content and metabolize 25–30% of clinically used drugs. Polymorphisms in these genes may result in modified expression or functionality, correlating with altered metabolism and clearance which may affect bioavailability (Dai et al., 2001; Kirchheiner et al., 2002; Musumba et al., 2013; Krasniqi et al., 2016; Guengerich, 2020). Celecoxib, ibuprofen, lornoxicam and piroxicam are extensively (>90%) metabolized by CYP2C enzymes (Agúndez et al., 2009).

Estimates suggest that CYP2C9 is responsible for biotransformation and metabolic clearance in 15–20% of all phase I metabolized drugs (Goldstein and de Morais, 1994; Schwarz, 2003; Van Booven et al., 2010; Niinuma et al., 2014). CYP2C9 is involved as the main or secondary enzyme in the metabolism of most NSAIDs, including aceclofenac, aspirin, celecoxib, diclofenac, flurbiprofen, indomethacin, lornoxicam, meloxicam, naproxen, piroxicam, and tenoxicam (Davies et al., 2000; Rodrigues, 2005; Agúndez et al., 2009; Samer et al., 2013; Theken et al., 2020). With celecoxib, lornoxicam and piroxicam, CYP2C9 is the predominant enzyme, responsible for 90% of drug metabolism (Agúndez et al., 2009).

At least 62 variant alleles and multiple sub-alleles have been reported for CYP2C9 (PharmVar (https://www.pharmvar.org/gene/CYP2C9) (PharmVar, 2017; Gaedigk et al., 2018; Theken et al., 2020) which vary in frequency ethnically, geographically and racially (Theken et al., 2020; Theken et al., 2020; Gene‐specific Information Tables for CYP2C9, 2020). The allelic variants CYP2C9*2 (rs1799853) and CYP2C9*3 (rs1057910) show high allele frequencies in human populations (García-Martín et al., 2006); the estimated prevalence in European populations is 14% for CYP2C9*2 and 8% for CYP2C9*3 (Lee et al., 2002; Xie et al., 2002; Sánchez-Diz et al., 2009). These variants, most notably CYP2C9*3, have been extensively studied and have been associated with decreased NSAID metabolism (Visser et al., 2005; García-Martín et al., 2006; Agúndez et al., 2009; Wadelius et al., 2009; Wang et al., 2011).

Carriers of low activity CYP2C9 alleles could be at greater risk of GI toxicity due to increased NSAID exposure. Frustratingly, case-control studies investigating associations between CYP2C9*2 and *3 variants and NSAID-induced ADRs have often been contradictory. Several studies have reported no associations between CYP2C9 variants and NSAID-induced PUD and/or UGIB (Martin et al., 2001; Van Oijen et al., 2005; Vonkeman et al., 2006; Lopezrodriguez et al., 2008; Ma et al., 2008; Musumba et al., 2013; Ishihara et al., 2014), while others have reported that CYP2C9 variants predispose to PUD (Martínez et al., 2004; Pilotto et al., 2007), including the presence of a gene-dose effect (Figueiras and et al., 2016), and that there may be a combined effect of CYP2C8/CYP2C9 variation (Blanco et al., 2005; Blanco et al., 2008).

Efforts to provide PGx-based guidance on NSAIDs recently culminated in the Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for CYP2C9 and NSAIDs (Theken et al., 2020). The guideline categorizes CYP2C9 alleles as follows: Normal function (wild type) CYP2C9*1, decreased function CYP2C9*2, *5, *8, *11, and no function CYP2C9*3, *6, *13 (Theken et al., 2020; Theken et al., 2020; Gene‐specific Information Tables for CYP2C9, 2020). In vitro and clinical studies have suggested that the “decreased-function” and “no-function” CYP2C9 alleles are substrate-dependent. A meta-analysis of CYPC9 variant alleles on NSAID exposure, based on predicted metaboliser phenotypes (poor metabolisers, intermediate metabolisers and normal metabolisers), showed that CYP2C9 poor metabolisers had decreased metabolic clearance, a prolonged plasma elimination half-life and increased plasma concentrations. Since NSAID-induced GI toxicity is known to be dose-dependent, the risk and severity of toxicity is likely to be increased (Bhala et al., 2013; Laporte et al., 2004; Figueiras and et al., 2016; Lanas et al., 2015). Hence, to mitigate the risk of PUD, dose-reduction, careful monitoring for toxicity/ADRs and alternative therapies, (e.g. non-CYP2C9-metabolized NSAIDs (such as naproxen)) should be used (Theken et al., 2020).

A subsequent systematic review and meta-analysis by Macías et al. (2020) also addressed discrepancies in previous studies. In an exhaustive study, they concluded: “There is a clear and consistent association of the development of GI adverse events with the CYP2C9 genotype, and the association is slightly stronger in patients with GI bleeding.” Consistent with this, Theken et al. (2020), found that the associations were stronger in poor metabolisers (again based on metaboliser phenotypes) with a significant gene-dose effect. Furthermore, allele-specific analyses showed that CYP2C9*2 was a poor risk predictor (marginal effects) in contrast to CYP2C9*3, which clearly showed a highly significant association with increased risk of upper GI adverse events and GI bleeding (Macías et al., 2020). This supports the findings of Figueiras and et al. (2016) who demonstrated dose-dependency in NSAID-induced GI damage and the indication that the CYP2C9*3 allele can be used as a predictive UGIB risk marker for CYP2C9-metabolised NSAIDS (Figueiras and et al., 2016).

CYP2C8 plays an accessory metabolic role for certain NSAIDs including ibuprofen and diclofenac (Agúndez et al., 2009; Garciamartin and et al., 2004). CYP2C9*2 is in strong linkage disequilibrium (LD) with CYP2C8*3 (Speed et al., 2009). Findings however have again been conflicting: no association between CYP2C8*3 and UGIB was reported by Musumba et al. (Musumba et al., 2013) while others have reported a positive association with GI bleeding (Agúndez et al., 2009), including when combined with CYP2C9*2 (Blanco et al., 2005; Blanco et al., 2008).

CYP2C19 plays a minor role in NSAID metabolism. Interestingly, we showed that the CYP2C19*17 “gain of function” polymorphism was associated with PUD (Musumba et al., 2013), irrespective of the etiology of PUD. This may be related to the fact that CYP2C19 is involved in the metabolism of AA, and more extensive metabolism of AA may impair gastric mucosal defences. This is consistent with the fact that disrupted AA metabolism is involved in PUD pathogenesis (Musumba et al., 2009).

Thus, the role of the of the CYP2C gene locus in predisposing to PUD is complex and likely to vary with the NSAID given, its dose and the complement of SNPs the patient has at the CYP2C gene locus, including at CYP2C9 (where low activity variants will increase exposure to NSAIDs) and the gain of function polymorphism at CYP2C19 which increases the metabolism of gastro-protective AA.

Pharmacodynamic-Related Associations

COX enzymes produce PGs from AA, playing vital roles in gastric defence. COX enzymes are the primary target of NSAIDs. There is potential for peptic ulcer pathogenesis to be influenced by functional polymorphisms in the COX-encoding genes (Agúndez et al., 2015). Individuals carrying reduced function COX-enzymes may be potentially susceptible to NSAID-induced peptic ulceration (Musumba et al., 2009).

COX-2 single nucleotide polymorphism (SNPs) have been observed to affect responsiveness to celecoxib (Lee et al., 2017), but no association with NSAID-induced GI injury was reported. Studies of COX-1 genetic polymorphisms and GI injury have also yielded contradictory findings (van Oijen et al., 2006; Arisawa et al., 2007). As such, there is insufficient evidence currently to determine whether the COX gene polymorphisms predispose to NSAID-induced PUD.

A recent preliminary study by Mallah et al. identified polymorphisms in platelet activity, angiogenesis and inflammatory response genes were associated with aspirin-related UGIB (Mallah and et al., 2020), building on the findings of previous investigations (Shiotani et al., 2010; Shiotani and et al., 2013; Shiotani et al., 2014; Wu et al., 2016; Cho and et al., 2016; Milanowski et al., 2017). The group identified “positive markers,” that indicated an increased risk of aspirin related UGIB and protective “negative markers” which decreased the risk (Mallah and et al., 2020). Of particular interest, rs689466 T > C, a SNP in the COX-2 gene was associated with an increased risk of UGIB (Mallah and et al., 2020). However, these data are preliminary and need to be replicated in other cohorts.

There is significant literature describing associations between NSAID-induced immune-mediated ADRs and HLA alleles. These associations are with type I (immediate) urticarial and anaphylactic reactions (HLA-DR11 and aspirin (Quiralte et al., 1999) and also type IV (delayed) reactions such as Stevens-Johnson syndrome/toxic epidermal necrolysis (HLA-B*73:01 and oxicam (Lonjou et al., 2008)). To the best of our knowledge, there are no reports of HLA associations with NSAID-induced GI toxicity which is reflective of the non-immune mediated nature of the adverse event.

Based on the pharmacogenomic data presented herein, it is reasonable to suggest that evidence for genetic predisposition to NSAID-induced upper GI toxicity is somewhat contradictory. This reflects the complex nature of the etiology of this ADR but also the significant inter-individual variability in non-genetic risk factors. There are a number of factors which may influence inter-individual pre-disposition to NSAID GI and need to be integrated with pharmacogenomics to further our understanding of individual risk. These include, but not limited to: 1) the impact of altered GI physiology and subsequent drug disposition in special groups and the impact of disease physiology (Stillhart et al., 2020), 2) variability in physiological regulation via gastric enteroendocrine cells (Mace et al., 2015) and 3) Helicobacter pylori infection which is a significant risk factor for peptic ulceration independent of NSAID use (Huang et al., 2002).

Beyond the Human Genome

Spurred on by the completion of the Human Microbiome Project (2012), growing evidence supports the role of the microbiome in health and disease. Detrimental changes in gut microbiota composition (intestinal microbiota dysbiosis) are suggested as markers of GI pathogenesis in Crohn’s disease (Gevers et al., 2014), UC (Shen et al., 2018), NSAID-induced enteropathy (Montalto et al., 2013; Syer and Wallace, 2014; Rekatsina et al., 2020) and ulcer healing (Blackler et al., 2015). Microbiome heterogeneity may also influence drug response (Structure, function and diversity of the healthy human microbiome, 2012; Forslund et al., 2015; Wu et al., 2017; Ma et al., 2019; Sharma et al., 2019; Doestzada et al., 2018; Zimmermann et al., 2019; Yip and Chan, 2015; Kashyap et al., 2017).

NSAID disposition, therapeutic efficacy and toxicity are influenced by dynamic and complex host-gut microbiota interactions. Gut microbiota may directly modify NSAID chemistry or manipulate host metabolic processes affecting drug pharmacokinetics and pharmacodynamics. NSAIDs may modify gut microbiota composition resulting in dysbiosis (Maseda and Ricciotti, 2020). Rogers and Aronoff (2016) reported differences in gut microbiota profiles between non-users and NSAID-users. Gut microbiome composition also varied with the type of NSAID ingested (Rogers and Aronoff, 2016).

The capacity to intentionally manipulate the microbiota through diet (Dzutsev et al., 2017; Thiele et al., 2017; Ma et al., 2019), antibiotics/antimicrobials (Lanas and Scarpignato, 2006), fecal microbiota transplant (FMT) (Garber, 2015) or administration of selected probiotic strains (Gionchetti et al., 2000; Ulisse et al., 2001; Gionchetti et al., 2003; Montalto et al., 2010; Montalto et al., 2013; Suzuki et al., 2017; Mortensen et al., 2019; Rekatsina et al., 2020) to enhance efficacy and preserve functional mucosal integrity provides potential therapeutic value for various GI diseases. Recent developments in metaproteomics-based assays offer novel insights into microbiome absolute abundance and functional responses to drugs (Li and et al., 2020). Deployment of next generation sequencing technologies to identify predictive, diagnostic, and prognostic biomarkers need to be further investigated to determine their role in NSAID-induced GI pathology.

Future Opportunities and Clinical Implementation

Further research into genetic risk factors predisposing individuals to NSAID-induced upper GI toxicity is justified. Knowledge gaps remain. Currently, there is a shortfall of genome-wide studies, with candidate gene approaches dominating. Lack of consensus and scarcity of independent replication to validate findings has been problematic in published studies. Given the burden and prevalence of NSAID-induced GI ADRs, this is perplexing. A genome-wide approach would allow for unbiased identification of common and rare variants in both PK and PD related genes but would require a large sample size with detailed phenotyping of cases and controls. Indeed, a variability in phenotype definitions is a further challenge in identifying tractable genetic associations. Thus, focused collaborative efforts to standardize phenotype definitions, as shown with other ADRs (Carr et al., 2017; Pirmohamed et al., 2011; Alfirevic et al., 2014; Nicoletti and et al., 2020; Aithal et al., 2011; Behr et al., 2012; Pirmohamed et al., 2011) should be encouraged, drawing inspiration from an adapted ‘consensus approach’ (Pirmohamed et al., 2011).

Conclusion

The pathogenesis of NSAID-induced GI toxicity is complex. PGx is a useful tool to improve pharmacotherapy, and aims to shift the paradigm of dosing regimens being extrapolated to entire populations (Jaccard et al., 2020). Interrogation of genetic factors which predispose individuals to NSAID-induced upper GI toxicity offers unquestionable benefit: pre-emptive testing minimizes the risks of ADRs, informing clinical practice guidelines and therapeutic recommendations to enhance pharmacotherapy (Macías et al., 2020; Theken et al., 2020). To date, the CYP2C locus has been shown to be important in some but not all studies. Taken together with the fact that some NSAIDs are only partially metabolized by CYP2C9, but still cause PUD, it can be concluded that CYP2C genetic variants are neither necessary nor sufficient to predispose to NSAID-induced PUD, but the presence of low activity CYP2C9 alleles in a patient given a CYP2C9-metabolised NSAID may increase the risk of PUD. Further work however is also required to identify novel genetic predisposing factors using genome-wide approaches but this will require larger numbers of well phenotyped patients.

Another factor to consider is that as our population continues to age, the prevalence of multimorbidity will increase (Uijen and van de Lisdonk, 2008), which will be accompanied by polypharmacy. Multimorbidity and polypharmacy increase the likelihood of drug- and gene-based interactions (Turner and et al., 2020). The elderly are liable to be prescribed NSAIDs, including LDA, because of the high prevalence of cardiovascular disease and arthritis. The risk of drug-drug interactions with concomitantly administered drugs is likely to be increased in the elderly: these may occur at both pharmacokinetic, (e.g. inhibition of CYP2C9 metabolism) and pharmacodynamic, (e.g. use of NSAIDs and corticosteroids) levels, and may thus contribute to the increased risk of upper GI toxicity. Thus with prolific NSAID use in the over 65 year olds, combinations of risk factors will be common and cumulative, increasing the risks of NSAID-induced ADRs (Laine et al., 2002). It is likely that these ADRs will increase in prevalence despite the use of gastroprotective therapies such as proton pump inhibitors.

In summary therefore, there is a continuing need to define genetic predisposing factors for NSAID-induced PUD because NSAIDs are very widely used, the occurrence of PUD is associated with a high degree of morbidity and mortality, and it is likely with the change in our age demographics, the problems of NSAID-induced PUD are likely to increase rather than decrease.

Author Contributions

DC, LM, and MP all contributed to the concept, content, writing and editing of this manuscript.

Funding

The work of MP and DC was part-funded by the Medical Research Council grant for the Center for Drug Safety Science, University of Liverpool (Grant Number: MR/L006758/1).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Abbreviations

AA, Arachidonic acid; ADR, Adverse drug reaction; ASA, (Acetylsalicylic acid) Aspirin; COX, cyclo-oxygenase; Coxibs, COX-2 inhibitors; CPIC, Clinical Pharmacogenetics Implementation Consortium; CV, Cardiovascular; CYP, Cytochrome P450; FMT, Fecal microbiota transplant; GI, Gastrointestinal; GWAS, Genome wide association study; IBD, Inflammatory bowel disease; LD, Linkage disequilibrium; LDA, Low dose aspirin; NSAID, Non-steroidal anti-inflammatory drug; OTC, Over-the-counter; PG, Prostaglandin; PGx, Pharmacogenomics; PhV, Pharmacovigilance; PUD, Peptic ulcer disease; SNP, Single nucleotide polymorphism; TX, Thromboxane; tNSAID, Traditional NSAID; UC, Ulcerative colitis; UGIB, Upper gastrointestinal bleeding; UGT, Uridine 5′-diphosphate glucuronosyltransferases (UDP-glucuronosyltransferase).

References

Agúndez, J. A., Blanca, M., Cornejo-García, J. A., and García-Martín, E. (2015). Pharmacogenomics of Cyclooxygenases. Pharmacogenomics 16 (5), 501–522. doi:10.2217/pgs.15.6

PubMed Abstract | CrossRef Full Text | Google Scholar

Agúndez, J. A., García-Martín, E., and Martínez, C. (2009). Genetically Based Impairment in CYP2C8- and CYP2C9-dependent NSAID Metabolism as a Risk Factor for Gastrointestinal Bleeding: Is a Combination of Pharmacogenomics and Metabolomics Required to Improve Personalized Medicine? Expert Opin. Drug Metab. Toxicol. 5 (6), 607–620. doi:10.1517/17425250902970998

PubMed Abstract | CrossRef Full Text | Google Scholar

Åhsberg, K., Höglund, P., Kim, W.-H., and von Holstein, C. S. (2010). Impact of Aspirin, NSAIDs, Warfarin, Corticosteroids and SSRIs on the Site and Outcome of Non-variceal Upper and Lower Gastrointestinal Bleeding. Scand. J. Gastroenterol. 45 (12), 1404–1415. doi:10.3109/00365521.2010.510567

PubMed Abstract | CrossRef Full Text | Google Scholar

Aithal, G. P., Watkins, P. B., Andrade, R. J., Larrey, D., Molokhia, M., Takikawa, H., et al. (2011). Case Definition and Phenotype Standardization in Drug-Induced Liver Injury. Clin. Pharmacol. Ther. 89 (6), 806–815. doi:10.1038/clpt.2011.58

PubMed Abstract | CrossRef Full Text | Google Scholar

Alfirevic, A., Neely, D., Armitage, J., Chinoy, H., Cooper, R. G., Laaksonen, R., et al. (2014). Phenotype Standardization for Statin-Induced Myotoxicity. Clin. Pharmacol. Ther. 96 (4), 470–476. doi:10.1038/clpt.2014.121

PubMed Abstract | CrossRef Full Text | Google Scholar

Anglin, R., Yuan, Y., Moayyedi, P., Tse, F., Armstrong, D., and Leontiadis, G. I. (2014). Risk of Upper Gastrointestinal Bleeding with Selective Serotonin Reuptake Inhibitors with or without Concurrent Nonsteroidal Anti-inflammatory Use: a Systematic Review and Meta-Analysis. Am. J. Gastroenterol. 109 (6), 811–819. doi:10.1038/ajg.2014.82

PubMed Abstract | CrossRef Full Text | Google Scholar

Arisawa, T, Tahara, T, Shibata, T, Nagasaka, M, Nakamura, M, Kamiya, Y, et al. (2007). Association between Genetic Polymorphisms in the Cyclooxygenase-1 Gene Promoter and Peptic Ulcers in Japan. Int. J. Mol. Med. 20 (3), 373–8.

PubMed AbstractGoogle Scholar

Armstrong, C. P., and Blower, A. L. (1987). Non-steroidal Anti-inflammatory Drugs and Life Threatening Complications of Peptic Ulceration. Gut 28 (5), 527–532. doi:10.1136/gut.28.5.527

PubMed Abstract | CrossRef Full Text | Google Scholar

Bashir, A. I. J., Kankipati, C. S., Jones, S., Newman, R. M., Safrany, S. T., Perry, C. J., et al. (2019). A Novel Mechanism for the Anticancer Activity of Aspirin and Salicylates. Int. J. Oncol. 54 (4), 1256–1270. doi:10.3892/ijo.2019.4701

PubMed Abstract | CrossRef Full Text | Google Scholar

Behr, E. R., January, C., Schulze-Bahr, E., Grace, A. A., Kääb, S., Fiszman, M., et al. (2012). The International Serious Adverse Events Consortium (iSAEC) phenotype standardization project for drug-induced torsades de pointes. Eur. Heart J. 34 (26), 1958–1963. doi:10.1093/eurheartj/ehs172

PubMed Abstract | CrossRef Full Text | Google Scholar

Bhala, N., Bhala, N., Emberson, J., Merhi, A., Abramson, S., Arber, N., et al. (2013). Vascular and Upper Gastrointestinal Effects of Non-steroidal Anti-inflammatory Drugs: Meta-Analyses of Individual Participant Data from Randomised Trials. Lancet 382 (9894), 769–779. doi:10.1016/S0140-6736(13)60900-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Bhandari, P., Bateman, A. C., Mehta, R. L., and Patel, P. (2005). Mucosal Expression of Cyclooxygenase Isoforms 1 and 2 Is Increased with Worsening Damage to the Gastric Mucosa. Histopathology 46 (3), 280–286. doi:10.1111/j.1365-2559.2005.02053.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Bhatt, D. L., Scheiman, J., Abraham, N. S., Antman, E. M., Chan, F. K., Furberg, C. D., et al. (2008). ACCF/ACG/AHA 2008 Expert Consensus Document on Reducing the Gastrointestinal Risks of Antiplatelet Therapy and NSAID Use: a Report of the American College of Cardiology Foundation Task Force on Clinical Expert Consensus Documents. J. Am. Coll. Cardiol. 52 (18), 1502–1517. doi:10.1016/j.jacc.2008.08.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Biondi-Zoccai, G. G. L., Lotrionte, M., Agostoni, P., Abbate, A., Fusaro, M., Burzotta, F., et al. (2006). A Systematic Review and Meta-Analysis on the Hazards of Discontinuing or Not Adhering to Aspirin Among 50 279 Patients at Risk for Coronary Artery Disease. Eur. Heart J. 27 (22), 2667–2674. doi:10.1093/eurheartj/ehl334

PubMed Abstract | CrossRef Full Text | Google Scholar

Bjarnason, I. (2013). Gastrointestinal Safety of NSAIDs and Over-the-counter Analgesics. Int. J. Clin. Pract. 67 (s178), 37–42. doi:10.1111/ijcp.12048

PubMed Abstract | CrossRef Full Text | Google Scholar

Bjarnason, I., Scarpignato, C., Holmgren, E., Olszewski, M., Rainsford, K. D., and Lanas, A. (2018). Mechanisms of Damage to the Gastrointestinal Tract from Nonsteroidal Anti-inflammatory Drugs. Gastroenterology 154 (3), 500–514. doi:10.1053/j.gastro.2017.10.049

PubMed Abstract | CrossRef Full Text | Google Scholar

Bjorkman, D. (1998). Nonsteroidal Anti-inflammatory Drug-Associated Toxicity of the Liver, Lower Gastrointestinal Tract, and Esophagus. Am. J. Med. 105 (5Suppl. 1), 17S–21S. doi:10.1016/s0002-9343(98)00276-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Blackler, R. W., Motta, J.-P., Manko, A., Workentine, M., Bercik, P., Surette, M. G., et al. (2015). Hydrogen Sulphide Protects against NSAID-Enteropathy through Modulation of Bile and the Microbiota. Br. J. Pharmacol. 172 (4), 992–1004. doi:10.1111/bph.12961

PubMed Abstract | CrossRef Full Text | Google Scholar

Blanco, G., Martínez, C., García-Martín, E., and Agúndez, J. A. G. (2005). Cytochrome P450 Gene Polymorphisms and Variability in Response to NSAIDs. Clin. Res. Regul. Aff. 22 (2), 57–81. doi:10.1080/10601330500214559

CrossRef Full Text | Google Scholar

Blanco, G., Martínez, C., Ladero, J. M., Garcia-Martin, E., Taxonera, C., Gamito, F. G., et al. (2008). Interaction of CYP2C8 and CYP2C9 Genotypes Modifies the Risk for Nonsteroidal Anti-inflammatory Drugs-Related Acute Gastrointestinal Bleeding. Pharmacogenet Genomics 18 (1), 37–43. doi:10.1097/fpc.0b013e3282f305a9

PubMed Abstract | CrossRef Full Text | Google Scholar

Blower, A. L., Brooks, A., Fenn, GC., Hill, A., Pearce, MY., Morant, S., et al. (1997). Emergency Admissions for Upper Gastrointestinal Disease and Their Relation to NSAID Use. Aliment. Pharmacol. Ther. 11 (2). doi:10.1046/j.1365-2036.1997.d01-604.x

CrossRef Full Text | Google Scholar

Bombardier, C., Laine, L., Reicin, A., Shapiro, D., Burgos-Vargas, R., Davis, B., et al. (2000). Comparison of Upper Gastrointestinal Toxicity of Rofecoxib and Naproxen in Patients with Rheumatoid Arthritis. N. Engl. J. Med. 343 (21), 1520–1528. doi:10.1056/nejm200011233432103

PubMed Abstract | CrossRef Full Text | Google Scholar

Brune, K., Glatt, M., and Graf, P. (1976). Mechanisms of Action of Anti-inflammatory Drugs. Gen. Pharmacol. Vasc. Syst. 7 (1), 27–IN1. doi:10.1016/0306-3623(76)90028-8

CrossRef Full Text | Google Scholar

Brune, K., and Graf, P. (1978). Non-steroid Anti-inflammatory Drugs: Influence of Extra-cellular pH on Biodistribution and Pharmacological Effects. Biochem. Pharmacol. 27 (4), 525–530. doi:10.1016/0006-2952(78)90388-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Butt, J. H., Barthel, J. S., and Moore, R. A. (1988). Clinical Spectrum of the Upper Gastrointestinal Effects of Nonsteroidal Anti-inflammatory Drugs. Am. J. Med. 84 (2a), 5–14. doi:10.1016/0002-9343(88)90248-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Calatayud, S., and Esplugues, J. V. (2016). “Chemistry, Pharmacodynamics, and Pharmacokinetics of NSAIDs,” in NSAIDs and Aspirin: Recent Advances and Implications for Clinical Management. Editor A. Lanas (Cham: Springer International Publishing), 3–16. doi:10.1007/978-3-319-33889-7_1

CrossRef Full Text | Google Scholar

Campbell, K., and Steele, R. J. (1991). Non-steroidal Anti-inflammatory Drugs and Complicated Diverticular Disease: a Case-Control Study. Br. J. Surg. 78 (2), 190–191. doi:10.1002/bjs.1800780218

PubMed Abstract | CrossRef Full Text | Google Scholar

Carbonell, N., Verstuyft, C., Massard, J., Letierce, A., Cellier, C., Deforges, L., et al. (2010). CYP2C9*3 Loss-Of-Function Allele Is Associated with Acute Upper Gastrointestinal Bleeding Related to the Use of NSAIDs Other Than Aspirin. Clin. Pharmacol. Ther. 87 (6), 693–698. doi:10.1038/clpt.2010.33

PubMed Abstract | CrossRef Full Text | Google Scholar

Carr, D. F., Ayehunie, S., Davies, A., Duckworth, C. A., French, S., Hall, N., et al. (2017). Towards Better Models and Mechanistic Biomarkers for Drug-Induced Gastrointestinal Injury. Pharmacol. Ther. 172, 181–194. doi:10.1016/j.pharmthera.2017.01.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Castellsague, J., Riera-Guardia, N., Calingaert, B., Varas-Lorenzo, C., Fourrier-Reglat, A., Nicotra, F., et al. (2012). Individual NSAIDs and Upper Gastrointestinal Complications. Drug Saf. 35 (12), 1127–1146. doi:10.1007/bf03261999

PubMed Abstract | CrossRef Full Text | Google Scholar

Catella-Lawson, F., Reilly, M. P., Kapoor, S. C., Cucchiara, A. J., DeMarco, S., Tournier, B., et al. (2001). Cyclooxygenase Inhibitors and the Antiplatelet Effects of Aspirin. N. Engl. J. Med. 345 (25), 1809–1817. doi:10.1056/nejmoa003199

PubMed Abstract | CrossRef Full Text | Google Scholar

Cayla, G., Collet, J.-P., Silvain, J., Thiefin, G., Woimant, F., and Montalescot, G. (2012). Prevalence and Clinical Impact of Upper Gastrointestinal Symptoms in Subjects Treated with Low Dose Aspirin: The UGLA Survey. Int. J. Cardiol. 156 (1), 69–75. doi:10.1016/j.ijcard.2010.10.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Chan, F. K., Lanas, A., Scheiman, J., Berger, M. F., Nguyen, H., and Goldstein, J. L. (2010). Celecoxib versus Omeprazole and Diclofenac in Patients with Osteoarthritis and Rheumatoid Arthritis (CONDOR): a Randomised Trial. The Lancet 376 (9736), 173–179. doi:10.1016/s0140-6736(10)60673-3

CrossRef Full Text | Google Scholar

Chan, F. K. L., Ching, J. Y. L., Tse, Y. K., Lam, K., Wong, G. L. H., Ng, S. C., et al. (2017). Gastrointestinal Safety of Celecoxib versus Naproxen in Patients with Cardiothrombotic Diseases and Arthritis after Upper Gastrointestinal Bleeding (CONCERN): an Industry-independent, Double-Blind, Double-Dummy, Randomised Trial. The Lancet 389 (10087), 2375–2382. doi:10.1016/s0140-6736(17)30981-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Chan, F. K. L., Hung, L. C. T., Suen, B. Y., Wong, V. W. S., Hui, A. J., Wu, J. C. Y., et al. (2005). Effect of Celecoxib on the Healing of Complicated Gastric Ulcers: A Prospective, Double Blinded, Randomized Trial, A24–A25.

Chi, T.-Y., Zhu, H.-M., and Zhang, M. (2018). Risk Factors Associated with Nonsteroidal Anti-inflammatory Drugs (NSAIDs)-Induced Gastrointestinal Bleeding Resulting on People over 60 Years Old in Beijing. Medicine 97 (18), e0665. doi:10.1097/md.0000000000010665

PubMed Abstract | CrossRef Full Text | Google Scholar

Cho, J. H., Choi, J. S., Chun, S. W., Lee, S., Han, K. J., Kim, H, M., et al. (2016). The IL-1B Genetic Polymorphism Is Associated with Aspirin-Induced PepticUlcers in a Korean Ethnic Group. Gut Liver 10 (3), 362–368. doi:10.5009/gnl15129

PubMed Abstract | CrossRef Full Text | Google Scholar

Cohen, M.M. (1987). Role of Endogenous Prostaglandins in Gastric Secretion and Mucosal Defense. Clin. Invest. Med. 10 (3), 226–31.

PubMed AbstractGoogle Scholar

Collaboration, A. T.Antithrombotic Trialists' Collaboration (2002). Collaborative Meta-Analysis of Randomised Trials of Antiplatelet Therapy for Prevention of Death, Myocardial Infarction, and Stroke in High Risk Patients. Bmj 324 (7329), 71–86. doi:10.1136/bmj.324.7329.71

PubMed Abstract | CrossRef Full Text | Google Scholar

Cryer, B. (2004). COX-2-specific Inhibitor or Proton Pump Inhibitor Plus Traditional NSAID: Is Either Approach Sufficient for Patients at Highest Risk of NSAID-Induced Ulcers? Gastroenterology 127 (4), 1256–1258. doi:10.1053/j.gastro.2004.08.029

PubMed Abstract | CrossRef Full Text | Google Scholar

Dai, D., Zeldin, D. C., Blaisdell, J. A., Chanas, B., Coulter, S. J., Ghanayem, B. I., et al. (2001). Polymorphisms in Human CYP2C8 Decrease Metabolism of the Anticancer Drug Paclitaxel and Arachidonic Acid. Pharmacogenetics 11 (7), 597–607. doi:10.1097/00008571-200110000-00006

PubMed Abstract | CrossRef Full Text | Google Scholar

Dajani, E. Z., and Agrawal, N. M. (1995). Prevention of Nonsteroidal Anti-inflammatory Drug-Induced Gastroduodenal Ulcers: Role of Mucosal Protective and Gastric Antisecretory Drugs. Dig. Dis. 13 (Suppl. 1), 48–61. doi:10.1159/000171526

PubMed Abstract | CrossRef Full Text | Google Scholar

Davies, N. M., McLachlan, A. J., Day, R. O., and Williams, K. M. (2000). Clinical Pharmacokinetics and Pharmacodynamics of Celecoxib. Clin. Pharmacokinet. 38 (3), 225–242. doi:10.2165/00003088-200038030-00003

PubMed Abstract | CrossRef Full Text | Google Scholar

Davies, N. M., and Wallace, J. L. (1996). Selective Inhibitors of Cyclooxygenase-2. Drugs and Aging 9 (6), 406–417. doi:10.2165/00002512-199609060-00004

PubMed Abstract | CrossRef Full Text | Google Scholar

de Abajo, F. J., Rodríguez, L. A. G., and Montero, D. (1999). Association between Selective Serotonin Reuptake Inhibitors and Upper Gastrointestinal Bleeding: Population Based Case-Control Study. Bmj 319 (7217), 1106–1109. doi:10.1136/bmj.319.7217.1106

PubMed Abstract | CrossRef Full Text | Google Scholar

de Jong, J. C. F., Van Den Berg, P. B., Tobi, H., and De Jong Van Den Berg, L. T. W. (2003). Combined Use of SSRIs and NSAIDs Increases the Risk of Gastrointestinal Adverse Effects. Br. J. Clin. Pharmacol. 55 (6), 591–595. doi:10.1046/j.0306-5251.2002.01770.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Dickman, A., and Ellershaw, J. (2004). For Discussion NSAIDs: Gastroprotection or Selective COX-2 Inhibitor? Palliat. Med. 18 (4), 275–286. doi:10.1191/0269216304pm894fd

PubMed Abstract | CrossRef Full Text | Google Scholar

Doestzada, M., Vila, A. V., Zhernakova, A., Koonen, D. P. Y., Weersma, R. K., Touw, D. J., et al. (2018). Pharmacomicrobiomics: a Novel Route towards Personalized Medicine? Protein Cell 9 (5), 432–445. doi:10.1007/s13238-018-0547-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Dunlap, J. J., and Patterson, S. (2019). Peptic Ulcer Disease. Gastroenterol. Nurs. 42 (5), 451–454. doi:10.1097/sga.0000000000000478

PubMed Abstract | CrossRef Full Text | Google Scholar

Dzutsev, A., Badger, J. H., Perez-Chanona, E., Roy, S., Salcedo, R., Smith, C. K., et al. (2017). Microbes and Cancer. Annu. Rev. Immunol. 35, 199–228. doi:10.1146/annurev-immunol-051116-052133

PubMed Abstract | CrossRef Full Text | Google Scholar

Eichelbaum, M., Ingelman-Sundberg, M., and Evans, W. E. (2006). Pharmacogenomics and Individualized Drug Therapy. Annu. Rev. Med. 57, 119–137. doi:10.1146/annurev.med.56.082103.104724

PubMed Abstract | CrossRef Full Text | Google Scholar

Evans, W. E., and Relling, M. V. (1999). Pharmacogenomics: Translating Functional Genomics into Rational Therapeutics. Science 286 (5439), 487–491. doi:10.1126/science.286.5439.487

PubMed Abstract | CrossRef Full Text | Google Scholar

Faucheron, J.-L. (1999). Toxicity of Non-steroidal Anti-inflammatory Drugs in the Large Bowel. Eur. J. Gastroenterol. Hepatol. 11 (4), 389–392. doi:10.1097/00042737-199904000-00005

PubMed Abstract | CrossRef Full Text | Google Scholar

Figueiras, A., Estany-Gestal, A., Aguirre, C., Ruiz, B., Vidal, X., Carvajal, A., et al. (2016). CYP2C9 Variants as a Risk Modifier of NSAID-Related Gastrointestinal Bleeding: a Case–Control Study. Pharmacogenetics and Genomics 26 (2). doi:10.1097/fpc.0000000000000186

CrossRef Full Text | Google Scholar

Forrest, E. H., Russell, R. I., Orchard, T. R., and Jewell, D. P. (1999). Peripheral Arthropathies in Inflammatory Bowel Disease Reply. Gut 44 (3), 439. doi:10.1136/gut.44.3.439

PubMed Abstract | CrossRef Full Text | Google Scholar

Forslund, K., au, fnm., Hildebrand, F., Nielsen, T., Falony, G., Le Chatelier, E., et al. (2015). Disentangling Type 2 Diabetes and Metformin Treatment Signatures in the Human Gut Microbiota. Nature 528 (7581), 262–266. doi:10.1038/nature15766

PubMed Abstract | CrossRef Full Text | Google Scholar

Fries, J. F., Williams, C. A., Bloch, D. A., and Michel, B. A. (1991). Nonsteroidal Anti-inflammatory Drug-Associated Gastropathy: Incidence and Risk Factor Models. Am. J. Med. 91 (3), 213–222. doi:10.1016/0002-9343(91)90118-h

PubMed Abstract | CrossRef Full Text | Google Scholar

Fries, J. (1996). Toward an Understanding of NSAID-Related Adverse Events: the Contribution of Longitudinal Data. Scand. J. Rheumatol. 25, 3–8. doi:10.3109/03009749609097225

CrossRef Full Text | Google Scholar

Funk, C. D., Funk, L. B., Kennedy, M. E., Pong, A. S., and Fitzgerald, G. A. (1991). Human Platelet/erythroleukemia Cell Prostaglandin G/H Synthase: cDNA Cloning, Expression, and Gene Chromosomal Assignment. FASEB j. 5 (9), 2304–2312. doi:10.1096/fasebj.5.9.1907252

PubMed Abstract | CrossRef Full Text | Google Scholar

Gabriel, S. E., Jaakkimainen, L., and Bombardier, C. (1991). Risk for Serious Gastrointestinal Complications Related to Use of Nonsteroidal Anti-inflammatory Drugs. Ann. Intern. Med. 115 (10), 787–796. doi:10.7326/0003-4819-115-10-787

PubMed Abstract | CrossRef Full Text | Google Scholar

Gaedigk, A., Ingelman-Sundberg, M., Miller, N. A., Leeder, J. S., Whirl-Carrillo, M., and Klein, T. E. (2018). The Pharmacogene Variation (PharmVar) Consortium: Incorporation of the Human Cytochrome P450 (CYP ) Allele Nomenclature Database. Clin. Pharmacol. Ther. 103 (3), 399–401. doi:10.1002/cpt.910

PubMed Abstract | CrossRef Full Text | Google Scholar

Garber, K. (2015). Drugging the Gut Microbiome. Nat. Biotechnol. 33 (3), 228–231. doi:10.1038/nbt.3161

PubMed Abstract | CrossRef Full Text | Google Scholar

Garcia Rodriguez, L., and Hernández-Díaz, S. (2001). The Risk of Upper Gastrointestinal Complications Associated with Nonsteroidal Anti-inflammatory Drugs, Glucocorticoids, Acetaminophen, and Combinations of These Agents. Arthritis Res. 3, 98–101. doi:10.1186/ar146

PubMed Abstract | CrossRef Full Text | Google Scholar

García Rodríguez, L., and Jick, H. (1994). Risk of Upper Gastrointestinal Bleeding and Perforation Associated with Individual Non-steroidal Anti-inflammatory Drugs. The Lancet 343 (8900), 769–772. doi:10.1016/s0140-6736(94)91843-0

CrossRef Full Text | Google Scholar

García-Martín, E., Martínez, C., Ladero, J. M., and Agúndez, J. A. G. (2006). Interethnic and Intraethnic Variability of CYP2C8 and CYP2C9 Polymorphisms in Healthy Individuals. Mol. Diag. Ther. 10 (1), 29–40. doi:10.1007/bf03256440

CrossRef Full Text | Google Scholar

García-Rayado, G., Navarro, M., and Lanas, A. (2018). NSAID Induced Gastrointestinal Damage and Designing GI-Sparing NSAIDs. Expert Rev. Clin. Pharmacol. 11 (10), 1031–1043. doi:10.1080/17512433.2018.1516143

PubMed Abstract | CrossRef Full Text | Google Scholar

García-Martín, E., Martínez, C., Tabarés, B., Frías, J., and Agúndez, J. A. G. (2004). Interindividual Variability in Ibuprofen Pharmacokinetics Is Related to Interaction of Cytochrome P450 2C8 and 2C9 Amino Acid Polymorphisms*1. Clin. Pharmacol. Ther. 76 (2), 119–127. doi:10.1016/j.clpt.2004.04.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Gargallo, C. J., Sostres, C., and Lanas, A. (2014). Prevention and Treatment of NSAID Gastropathy. Curr. Treat. Options. Gastro. 12 (4), 398–413. doi:10.1007/s11938-014-0029-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Geis, GS, Stead, H, Wallemark, CB, and Nicholson, PA (1991). Prevalence of Mucosal Lesions in the Stomach and Duodenum Due to Chronic Use of NSAID in Patients with Rheumatoid Arthritis or Osteoarthritis, and Interim Report on Prevention by Misoprostol of Diclofenac Associated Lesions. J. Rheumatol. Suppl. 28, 11–4.

PubMed AbstractGoogle Scholar

Gevers, D., Kugathasan, S., Denson, L. A., Vázquez-Baeza, Y., Van Treuren, W., Ren, B., et al. (2014). The Treatment-Naive Microbiome in New-Onset Crohn's Disease. Cell Host and Microbe 15 (3), 382–392. doi:10.1016/j.chom.2014.02.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Gionchetti, P., Rizzello, F., Helwig, U., Venturi, A., Lammers, K. M., Brigidi, P., et al. (2003). Prophylaxis of Pouchitis Onset with Probiotic Therapy: a Double-Blind, Placebo-Controlled Trial. Gastroenterology 124 (5), 1202–1209. doi:10.1016/s0016-5085(03)00171-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Gionchetti, P., Rizzello, F., Venturi, A., Brigidi, P., Matteuzzi, D., Bazzocchi, G., et al. (2000). Oral Bacteriotherapy as Maintenance Treatment in Patients with Chronic Pouchitis: a Double-Blind, Placebo-Controlled Trial. Gastroenterology 119 (2), 305–309. doi:10.1053/gast.2000.9370

PubMed Abstract | CrossRef Full Text | Google Scholar

Goldstein, J. A., and de Morais, S. M. F. (1994). Biochemistry and Molecular Biology of the Human CYP2C Subfamily. Pharmacogenetics 4 (6), 285–300. doi:10.1097/00008571-199412000-00001

PubMed Abstract | CrossRef Full Text | Google Scholar

Goldstein, J., and Cryer, B. (2015). Gastrointestinal Injury Associated with NSAID Use: a Case Study and Review of Risk Factors and Preventative Strategies. Dhps 7, 31–41. doi:10.2147/dhps.s71976

CrossRef Full Text | Google Scholar

Goldstein, J. L., Chan, F. K. L., Lanas, A., Wilcox, C. M., Peura, D., Sands, G. H., et al. (2011). Haemoglobin Decreases in NSAID Users over Time: an Analysis of Two Large Outcome Trials. Aliment. Pharmacol. Ther. 34 (7), 808–816. doi:10.1111/j.1365-2036.2011.04790.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Goldstein, J. L., and Lefkowith, J. B. (1998). Public Misunderstanding of Nonsteroidal Antiinflammatory Drug (NSAID)-mediated Gastrointestinal (GI) Toxicity: a Serious Potential Health Threat. Gastroenterology 114, A136. doi:10.1016/s0016-5085(98)80552-0

CrossRef Full Text | Google Scholar

Gómez-Acebo, I., Dierssen-Sotos, T., de Pedro, M., Pérez-Gómez, B., Castaño-Vinyals, G., Fernández-Villa, F., et al. (2018). Epidemiology of Non-steroidal Anti-inflammatory Drugs Consumption in Spain. The MCC-Spain Study. BMC Public Health 18 (1), 1134. doi:10.1186/s12889-018-6019-z

PubMed Abstract | CrossRef Full Text | Google Scholar

González-Pérez, A., Sáez, M. E., Johansson, S., Nagy, P., and García Rodríguez, L. A (2014). Risk Factors Associated with Uncomplicated Peptic Ulcer and Changes in Medication Use after Diagnosis. PloS one 9 (7), e101768. doi:10.1371/journal.pone.0101768

PubMed Abstract | CrossRef Full Text | Google Scholar

Gretzer, B., Maricic, N., Respondek, M., Schuligoi, R., and Peskar, B. M. (2001). Effects of Specific Inhibition of Cyclo-Oxygenase-1 and Cyclo-Oxygenase-2 in the Rat Stomach with normal Mucosa and after Acid challenge. Br. J. Pharmacol. 132 (7), 1565–1573. doi:10.1038/sj.bjp.0703955

PubMed Abstract | CrossRef Full Text | Google Scholar

Griffin, M. R., Piper, J. M., Daugherty, J. R., Snowden, M., and Ray, W. A. (1991). Nonsteroidal Anti-inflammatory Drug Use and Increased Risk for Peptic Ulcer Disease in Elderly Persons. Ann. Intern. Med. 114 (4), 257–263. doi:10.7326/0003-4819-114-4-257

PubMed Abstract | CrossRef Full Text | Google Scholar

Guengerich, F. P. (2020). A history of the roles of cytochrome P450 enzymes in the toxicity of drugs. Toxicol. Res. 37, 1–23. doi:10.1007/s43188-020-00056-z

Google Scholar

Gutthann, S. P., GarcíaRodríguez, L. A., and Raiford, D. S. (1997). Individual Nonsteroidal Antiinflammatory Drugs and Other Risk Factors for Upper Gastrointestinal Bleeding and Perforation. Epidemiology 8 (1), 18–24. doi:10.1097/00001648-199701000-00003

PubMed Abstract | CrossRef Full Text | Google Scholar

Halushka, M., Walker, L. P., and Halushka, P. V. (2003). Genetic Variation in Cyclooxygenase 1: Effects on Response to Aspirin. Clin. Pharmacol. Ther. 73 (1), 122–130. doi:10.1067/mcp.2003.1

PubMed Abstract | CrossRef Full Text | Google Scholar

Handa, O., Majima, A., Onozawa, Y., Horie, H., Uehara, Y., Fukui, A., et al. (2014). The Role of Mitochondria-Derived Reactive Oxygen Species in the Pathogenesis of Non-steroidal Anti-inflammatory Drug-Induced Small Intestinal Injury. Free Radic. Res. 48 (9), 1095–1099. doi:10.3109/10715762.2014.928411

PubMed Abstract | CrossRef Full Text | Google Scholar

Hawkey, C. J., and Langman, M. J. (2003). Non-steroidal Anti-inflammatory Drugs: Overall Risks and Management. Complementary Roles for COX-2 Inhibitors and Proton Pump Inhibitors. Gut 52 (4), 600–608. doi:10.1136/gut.52.4.600

PubMed Abstract | CrossRef Full Text | Google Scholar

Helin-Salmivaara, A., Huttunen, T., Grönroos, J. M., Klaukka, T., and Huupponen, R. (2007). Risk of Serious Upper Gastrointestinal Events with Concurrent Use of NSAIDs and SSRIs: a Case-Control Study in the General Population. Eur. J. Clin. Pharmacol. 63 (4), 403–408. doi:10.1007/s00228-007-0263-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Henry, D., Dobson, A., and Turner, C. (1993). Variability in the Risk of Major Gastrointestinal Complications from Nonaspirin Nonsteroidal Anti-inflammatory Drugs. Gastroenterology 105 (4), 1078–1088. doi:10.1016/0016-5085(93)90952-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Herlitz, J., Tóth, P. P., and Næsdal, J. (2010). Low-Dose Aspirin Therapy for Cardiovascular Prevention. Am. J. Cardiovasc. Drugs 10 (2), 125–141. doi:10.2165/11318440-000000000-00000

PubMed Abstract | CrossRef Full Text | Google Scholar

Hernández-Díaz, S., and Rodríguez, L. A. G. (2000). Association between Nonsteroidal Anti-inflammatory Drugs and Upper Gastrointestinal Tract Bleeding/Perforation. Arch. Intern. Med. 160 (14), 2093–2099. doi:10.1001/archinte.160.14.2093

PubMed Abstract | CrossRef Full Text | Google Scholar

Hirschowitz, BI (1994). Nonsteroidal Antiinflammatory Drugs and the Gastrointestinal Tract. Gastroenterologist 2 (3), 207–23.

PubMed AbstractGoogle Scholar

Huang, J.-Q., Sridhar, S., and Hunt, R. H. (2002). Role of Helicobacter pylori Infection and Non-steroidal Anti-inflammatory Drugs in Peptic-Ulcer Disease: a Meta-Analysis. The Lancet 359 (9300), 14–22. doi:10.1016/s0140-6736(02)07273-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Human Microbiome Project Consortium (2012). Structure, Function and Diversity of the Healthy Human Microbiome. Nature 486 (7402), 207–214. doi:10.1038/nature11234

PubMed Abstract | CrossRef Full Text | Google Scholar

Ingelman-Sundberg, M., Sim, S. C., Gomez, A., and Rodriguez-Antona, C. (2007). Influence of Cytochrome P450 Polymorphisms on Drug Therapies: Pharmacogenetic, Pharmacoepigenetic and Clinical Aspects. Pharmacol. Ther. 116 (3), 496–526. doi:10.1016/j.pharmthera.2007.09.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Ishihara, M., Ohmiya, N., Nakamura, M., Funasaka, K., Miyahara, R., Ohno, E., et al. (2014). Risk Factors of Symptomatic NSAID-Induced Small Intestinal Injury and Diaphragm Disease. Aliment. Pharmacol. Ther. 40 (5), 538–547. doi:10.1111/apt.12858

PubMed Abstract | CrossRef Full Text | Google Scholar

Jaccard, E, Redin, C, Girardin, F, Waeber, G, Fellay, J, and Vollenweider, P (2020). Pharmacogenomics : a Toolbox to Improve Drug Prescription. Rev. Med. Suisse 16 (716), 2259–2263.

PubMed AbstractGoogle Scholar

Jankovic, SM, Aleksic, J, Rakovic, S, Aleksic, A, Stevanovic, I, Stefanovic-Stoimenov, N, et al. (2009). Nonsteroidal Antiinflammatory Drugs and Risk of Gastrointestinal Bleeding Among Patients on Hemodialysis. J. Nephrol. 22 (4), 502–7.

PubMed AbstractGoogle Scholar

Johansson, I., and Ingelman-Sundberg, M. (2011). Genetic Polymorphism and Toxicology-With Emphasis on Cytochrome P450. Toxicol. Sci. 120 (1), 1–13. doi:10.1093/toxsci/kfq374

PubMed Abstract | CrossRef Full Text | Google Scholar

Johnsen, SP, Sørensen, HT, Mellemkjoer, L, Blot, WJ, Nielsen, GL, McLaughlin, JK, et al. (2001). Hospitalisation for Upper Gastrointestinal Bleeding Associated with Use of Oral Anticoagulants. Thromb. Haemost. 86 (2), 563–8.

PubMed AbstractGoogle Scholar

Kapur, B. M., Lala, P. K., and Shaw, J. L. (2014). Pharmacogenetics of Chronic Pain Management. Clin. Biochem. 47 (13-14), 1169–1187. doi:10.1016/j.clinbiochem.2014.05.065

PubMed Abstract | CrossRef Full Text | Google Scholar

Kashyap, P. C., Chia, N., Nelson, H., Segal, E., and Elinav, E. (2017). Microbiome at the Frontier of Personalized Medicine. Mayo Clinic Proc. 92 (12), 1855–1864. doi:10.1016/j.mayocp.2017.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, J., Lee, J., Shin, C. M., Lee, D. H., and Park, B.-J. (2015). Risk of Gastrointestinal Bleeding and Cardiovascular Events Due to NSAIDs in the Diabetic Elderly Population. BMJ Open Diab. Res. Care, 3(1): p. e000133. doi:10.1136/bmjdrc-2015-000133

PubMed Abstract | CrossRef Full Text | Google Scholar

Kirchheiner, J., Meineke, I., Freytag, G., Meisel, C., Roots, I., and Brockmöller, J. (2002). Enantiospecific Effects of Cytochrome P450 2C9 Amino Acid Variants on Ibuprofen Pharmacokinetics and on the Inhibition of Cyclooxygenases 1 and 2*. Clin. Pharmacol. Ther. 72 (1), 62–75. doi:10.1067/mcp.2002.125726

PubMed Abstract | CrossRef Full Text | Google Scholar

Krasniqi, V., Dimovski, A., Domjanović, I. K., Bilić, I., and Božina, N. (2016). How Polymorphisms of the Cytochrome P450 Genes Affect Ibuprofen and Diclofenac Metabolism and Toxicity/Kako Polimorfizmi Gena Citokroma P450 Utječu Na Metabolizam I Toksičnost Ibuprofena I Diklofenaka. Arhiv za higijenu rada i toksikologiju 67 (1), 1–8. doi:10.1515/aiht-2016-67-2754

PubMed Abstract | CrossRef Full Text | Google Scholar

Kuehl, G. E., Lampe, J. W., Potter, J. D., and Bigler, J. (2005). Glucuronidation of Nonsteroidal Anti-inflammatory Drugs: Identifying the Enzymes Responsible in Human Liver Microsomes. Drug Metab. Dispos. 33 (7), 1027–1035. doi:10.1124/dmd.104.002527

PubMed Abstract | CrossRef Full Text | Google Scholar

Laine, L. (2001). Approaches to Nonsteroidal Anti-inflammatory Drug Use in the High-Risk Patient. Gastroenterology 120 (3), 594–606. doi:10.1053/gast.2001.21907

PubMed Abstract | CrossRef Full Text | Google Scholar

Laine, L., Bombardier, C., Hawkey, C. J., Davis, B., Shapiro, D., Brett, C., et al. (2002). Stratifying the Risk of NSAID-Related Upper Gastrointestinal Clinical Events: Results of a Double-Blind Outcomes Study in Patients with Rheumatoid Arthritis. Gastroenterology 123 (4), 1006–1012. doi:10.1053/gast.2002.36013

PubMed Abstract | CrossRef Full Text | Google Scholar

Laine, L. (2006). GI Risk and Risk Factors of NSAIDs. J. Cardiovasc. Pharmacol. 47, S60–S66. doi:10.1097/00005344-200605001-00011

PubMed Abstract | CrossRef Full Text | Google Scholar

Lanas, A., García-Rodríguez, L. A., Arroyo, M. T., Bujanda, L., Gomollón, F., Forné, M., et al. (2007). Effect of Antisecretory Drugs and Nitrates on the Risk of Ulcer Bleeding Associated with Nonsteroidal Anti-inflammatory Drugs, Antiplatelet Agents, and Anticoagulants. Am. J. Gastroenterol. 102, 507–515. doi:10.1111/j.1572-0241.2006.01062.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Lanas, A., and Hunt, R. (2006). Prevention of Anti‐inflammatory Drug‐induced Gastrointestinal Damage: Benefits and Risks of Therapeutic Strategies. Ann. Med. 38 (6), 415–428. doi:10.1080/07853890600925843

PubMed Abstract | CrossRef Full Text | Google Scholar

Lanas, A., and Scarpignato, C. (2006). Microbial flora in NSAID-Induced Intestinal Damage: a Role for Antibiotics? Digestion 73 (Suppl. 1), 136–150. doi:10.1159/000089789

PubMed Abstract | CrossRef Full Text | Google Scholar

Lanas, Á., Carrera-Lasfuentes, P., Arguedas, Y., García, S., Bujanda, L., Calvet, X., et al. (2015). Risk of Upper and Lower Gastrointestinal Bleeding in Patients Taking Nonsteroidal Anti-inflammatory Drugs, Antiplatelet Agents, or Anticoagulants. Clin. Gastroenterol. Hepatol. 13 (5), 906–912. doi:10.1016/j.cgh.2014.11.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Langenbach, R., Morham, S. G., Tiano, H. F., Loftin, C. D., Ghanayem, B. I., Chulada, P. C., et al. (1995). Prostaglandin Synthase 1 Gene Disruption in Mice Reduces Arachidonic Acid-Induced Inflammation and Indomethacin-Induced Gastric Ulceration. Cell 83 (3), 483–492. doi:10.1016/0092-8674(95)90126-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Langman, M. J., Morgan, L., and Worrall, A. (1985). Use of Anti-inflammatory Drugs by Patients Admitted with Small or Large Bowel Perforations and Haemorrhage. Bmj 290 (6465), 347–349. doi:10.1136/bmj.290.6465.347

PubMed Abstract | CrossRef Full Text | Google Scholar

Langman, M. J. S. (1988). Ulcer Complications and Nonsteroidal Anti-inflammatory Drugs. Am. J. Med. 84 (2a), 15–19. doi:10.1016/0002-9343(88)90249-5

CrossRef Full Text | Google Scholar

Langman, M. J. S. (2001). Ulcer Complications Associated with Anti-inflammatory Drug Use. What Is the Extent of the Disease burden? Pharmacoepidem. Drug Safe. 10 (1), 13–19. doi:10.1002/pds.561

PubMed Abstract | CrossRef Full Text | Google Scholar

Lanza, F. L., Chan, F. K. L., and Quigley, E. M. M. (2009). Guidelines for Prevention of NSAID-Related Ulcer Complications. Am. J. Gastroenterol. 104 (3), 728–738. doi:10.14309/00000434-200903000-00035

PubMed Abstract | CrossRef Full Text | Google Scholar

Laporte, J.-R., Ib????ez, L., Vidal, X., Vendrell, L., and Leone, R. (2004). Upper Gastrointestinal Bleeding Associated with the Use of NSAIDs. Drug Saf. 27 (6), 411–420. doi:10.2165/00002018-200427060-00005

PubMed Abstract | CrossRef Full Text | Google Scholar

Larkai, EN, Smith, JL, Lidsky, MD, and Graham, DY (1987). Gastroduodenal Mucosa and Dyspeptic Symptoms in Arthritic Patients during Chronic Nonsteroidal Anti-inflammatory Drug Use. Am. J. Gastroenterol. 82 (11), 1153–8.

PubMed AbstractGoogle Scholar

Larkai, E. N., Smith, J. L., Lidsky, M. D., Sessoms, S. L., and Graham, D. Y. (1989). Dyspepsia in NSAID Users. J. Clin. Gastroenterol. 11 (2), 158–162. doi:10.1097/00004836-198904000-00009

PubMed Abstract | CrossRef Full Text | Google Scholar

Larson, A. M., Polson, J., Fontana, R. J., Davern, T. J., Lalani, E., Hynan, L. S., et al. (2005). Acetaminophen-induced Acute Liver Failure: Results of a United States Multicenter, Prospective Study. Hepatology 42 (6), 1364–1372. doi:10.1002/hep.20948

PubMed Abstract | CrossRef Full Text | Google Scholar

Lazzaroni, M., and Bianchi Porro, G. (2001). Prophylaxis and Treatment of Non-steroidal Anti-inflammatory Drug-Induced Upper Gastrointestinal Side-Effects. Dig. Liver Dis. 33 (Suppl. 2), S44–S58. doi:10.1016/s1590-8658(01)80158-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, C. R., Goldstein, J. A., and Pieper, J. A. (2002). Cytochrome P450 2C9 Polymorphisms: a Comprehensive Review of the In-Vitro and Human Data. Pharmacogenetics 12 (3), 251–263. doi:10.1097/00008571-200204000-00010

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, S. J., Park, M. K., Shin, D., and Chun, M. H. (2017). Variability of the Drug Response to Nonsteroidal Anti-inflammatory Drugs According to Cyclooxygenase-2 Genetic Polymorphism. Dddt 11, 2727–2736. doi:10.2147/dddt.s143807

PubMed Abstract | CrossRef Full Text | Google Scholar

Leontiadis, G. I., Sreedharan, A., Dorward, S., Barton, P., Delaney, B., Howden, C W., et al. (2007). Systematic Reviews of the Clinical Effectiveness and Cost-Effectiveness of Proton Pump Inhibitors in Acute Upper Gastrointestinal Bleeding. Health Technol. Assess. 11 (51), 1–164. doi:10.3310/hta11510

CrossRef Full Text | Google Scholar

Li, L., Ning, Z., Zhang, X., Mayne, J., Cheng, K., Stintzi, A., et al. (2020). RapidAIM: a Culture- and Metaproteomics-Based Rapid Assay of Individual Microbiome Responses to Drugs. Microbiome 8 (1), 33. doi:10.1186/s40168-020-00806-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Lichtenberger, L. M. (2001). Where Is the Evidence that Cyclooxygenase Inhibition Is the Primary Cause of Nonsteroidal Anti-inflammatory Drug (NSAID)-induced Gastrointestinal Injury? Biochem. Pharmacol. 61 (6), 631–637. doi:10.1016/s0006-2952(00)00576-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Ligumsky, M., Golanska, E. M., Hansen, D. G., and Kauffman, G. L. (1983). Aspirin Can Inhibit Gastric Mucosal Cyclo-Oxygenase without Causing Lesions in Rat. Gastroenterology 84 (4), 756–761. doi:10.1016/0016-5085(83)90143-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Ligumsky, M., Sestieri, M., Karmeli, F., Zimmerman, J., Okon, E., and Rachmilewitz, D. (1990). Rectal Administration of Nonsteroidal Andinflammatory Drugs. Gastroenterology 98 (5), 1245–1249. doi:10.1016/0016-5085(90)90340-7, Part 1

PubMed Abstract | CrossRef Full Text | Google Scholar

Loetsch, J., and Oertel, B. (2013). “NSAIDs, Pharmacokinetics,” in Encyclopedia of Pain. Editors G. F. Gebhart, and R. F. Schmidt (Berlin, Heidelberg: Springer Berlin Heidelberg), 2350–2361. doi:10.1007/978-3-642-28753-4_2853

CrossRef Full Text | Google Scholar

Lonjou, C., Borot, N., Sekula, P., Ledger, N., Thomas, L., Halevy, S., et al. (2008). A European Study of HLA-B in Stevens-Johnson Syndrome and Toxic Epidermal Necrolysis Related to Five High-Risk Drugs. Pharmacogenet Genomics 18 (2), 99–107. doi:10.1097/fpc.0b013e3282f3ef9c

PubMed Abstract | CrossRef Full Text | Google Scholar

Lopezrodriguez, R., Novalbos, J., Gallegosandin, S., Romanmartinez, M., Torrado, J., Gisbert, J., et al. (2008). Influence of CYP2C8 and CYP2C9 Polymorphisms on Pharmacokinetic and Pharmacodynamic Parameters of Racemic and Enantiomeric Forms of Ibuprofen in Healthy Volunteers. Pharmacol. Res. 58 (1), 77–84. doi:10.1016/j.phrs.2008.07.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, J., Yang, X. Y., Qiao, L., Liang, L. Q., and Chen, M. H. (2008). CYP2C9 Polymorphism in Non-steroidal Anti-inflammatory Drugs-Induced Gastropathy. J. Dig. Dis. 9 (2), 79–83. doi:10.1111/j.1751-2980.2008.00326.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, W., Mao, Q., Xia, W., Dong, G., Yu, C., and Jiang, F. (2019). Gut Microbiota Shapes the Efficiency of Cancer Therapy. Front. Microbiol. 10 (1050). doi:10.3389/fmicb.2019.01050

PubMed Abstract | CrossRef Full Text | Google Scholar

Mace, O. J., Tehan, B., and Marshall, F. (2015). Pharmacology and Physiology of Gastrointestinal Enteroendocrine Cells. Pharmacol. Res. Perspect. 3 (4), e00155. doi:10.1002/prp2.155

PubMed Abstract | CrossRef Full Text | Google Scholar

Macías, Y., Gómez Tabales, J., García-Martín, E., and Agúndez, J. A. G. (2020). An Update on the Pharmacogenomics of NSAID Metabolism and the Risk of Gastrointestinal Bleeding. Expert Opin. Drug Metab. Toxicol. 16 (4), 319–332. doi:10.1080/17425255.2020.1744563

PubMed Abstract | CrossRef Full Text | Google Scholar

Maiden, L., Thjodleifsson, B., Theodors, A., Gonzalez, J., and Bjarnason, I. (2005). A Quantitative Analysis of NSAID-Induced Small Bowel Pathology by Capsule Enteroscopy. Gastroenterology 128 (5), 1172–1178. doi:10.1053/j.gastro.2005.03.020

PubMed Abstract | CrossRef Full Text | Google Scholar

Mallah, N., Zapata-Cachafeiro, M., Aguirre, C., Ibarra-García, E., Palacios-Zabalza, I., Macías-García, F., et al. (2020). Influence of Polymorphisms Involved in Platelet Activation and Inflammatory Response on Aspirin-Related Upper Gastrointestinal Bleeding: A Case-Control Study. Front. Pharmacol. 11, 860. doi:10.3389/fphar.2020.01072

PubMed Abstract | CrossRef Full Text | Google Scholar

Martin, J. H., Begg, E. J., Kennedy, M. A., Roberts, R., and Barclay, M. L. (2001). Is Cytochrome P450 2C9 Genotype Associated with NSAID Gastric Ulceration? Br. J. Clin. Pharmacol. 51 (6), 627–630. doi:10.1046/j.0306-5251.2001.01398.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Martínez, C., Blanco, G., Ladero, J. M., García-Martín, E., Taxonera, C., Gamito, F. G., et al. (2004). Genetic Predisposition to Acute Gastrointestinal Bleeding after NSAIDs Use. Br. J. Pharmacol. 141 (2), 205–208. doi:10.1038/sj.bjp.0705623

PubMed Abstract | CrossRef Full Text | Google Scholar

Masclee, G. M. C., Valkhoff, V. E., van Soest, E. M., Schade, R., Mazzaglia, G., Molokhia, M., et al. (2013). Cyclo-oxygenase-2 Inhibitors or Nonselective NSAIDs Plus Gastroprotective Agents: what to Prescribe in Daily Clinical Practice? Aliment. Pharmacol. Ther. 38 (2), 178–189. doi:10.1111/apt.12348

PubMed Abstract | CrossRef Full Text | Google Scholar

Maseda, D., and Ricciotti, E. (2020). NSAID-gut Microbiota Interactions. Front. Pharmacol. 11, 1153. doi:10.3389/fphar.2020.01153

PubMed Abstract | CrossRef Full Text | Google Scholar

McCarthy, D. M. (1989). Nonsteroidal Antiinflammatory Drug-Induced Ulcers: Management by Traditional Therapies. Gastroenterology 96 (2 Pt 2 Suppl. l), 662–674. doi:10.1016/s0016-5085(89)80063-0

PubMed Abstract | CrossRef Full Text | Google Scholar

McCarthy, D. (1998). Nonsteroidal Anti-inflammatory Drug-Related Gastrointestinal Toxicity: Definitions and Epidemiology. Am. J. Med. 105 (5a), 3s–9s. doi:10.1016/s0002-9343(98)00274-5

PubMed Abstract | CrossRef Full Text | Google Scholar

McGettigan, P., and Henry, D. (2013). Use of Non-steroidal Anti-inflammatory Drugs that Elevate Cardiovascular Risk: an Examination of Sales and Essential Medicines Lists in Low-, Middle-, and High-Income Countries. Plos Med. 10 (2), e1001388. doi:10.1371/journal.pmed.1001388

PubMed Abstract | CrossRef Full Text | Google Scholar

Melcarne, L., García-Iglesias, P., and Calvet, X. (2016). Management of NSAID-Associated Peptic Ulcer Disease. Expert Rev. Gastroenterol. Hepatol. 10 (6), 723–733. doi:10.1586/17474124.2016.1142872

PubMed Abstract | CrossRef Full Text | Google Scholar

Milanowski, L., Pordzik, J., Janicki, P. K., Kaplon-Cieslicka, A., Rosiak, M., Peller, M., et al. (2017). New Single-Nucleotide Polymorphisms Associated with Differences in Platelet Reactivity and Their Influence on Survival in Patients with Type 2 Diabetes Treated with Acetylsalicylic Acid: an Observational Study. Acta Diabetol. 54 (4), 343–351. doi:10.1007/s00592-016-0945-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Monrad, R. N., Errey, J. C., Barry, C. S., Iqbal, M., Meng, X., Iddon, L., et al. (2014). Dissecting the Reaction of Phase II Metabolites of Ibuprofen and Other NSAIDS with Human Plasma Protein. Chem. Sci. 5 (10), 3789–3794. doi:10.1039/c4sc01329h

CrossRef Full Text | Google Scholar

Montalto, M., Gallo, A., Curigliano, V., D’Onofrio, F., Santoro, L., Covino, M., et al. (2010). Clinical Trial: the Effects of a Probiotic Mixture on Non-steroidal Anti-inflammatory Drug Enteropathy—a Randomized, Double-Blind, Cross-Over, Placebo-Controlled Study. Aliment. Pharmacol. Ther. 32 (2), 209–214. doi:10.1111/j.1365-2036.2010.04324.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Montalto, M., Gallo, A., Gasbarrini, A., and Landolfi, R. (2013). NSAID Enteropathy: Could Probiotics Prevent it? J. Gastroenterol. 48 (6), 689–697. doi:10.1007/s00535-012-0648-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Morham, S. G., Langenbach, R., Loftin, C. D., Tiano, H. F., Vouloumanos, N., Jennette, J. C., et al. (1995). Prostaglandin Synthase 2 Gene Disruption Causes Severe Renal Pathology in the Mouse. Cell 83 (3), 473–482. doi:10.1016/0092-8674(95)90125-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Mortensen, B., Murphy, C., O’Grady, J., Lucey, M., Elsafi, G., Barry, L., et al. (2019). Bifidobacterium Breve Bif195 Protects against Small-Intestinal Damage Caused by Acetylsalicylic Acid in Healthy Volunteers. Gastroenterology 157 (3), 637–646. doi:10.1053/j.gastro.2019.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Murray, F., O'Donovan, D G., Sheehan, K. M., and Murray, F. E. (1998). Prospective Evaluation of the Utilization of Aspirin and Non-steroidal Anti-inflammatory Drugs in Acute Medical Admissions. Headache 1, 3–7.

Google Scholar

Musumba, C. O., Jorgensen, A., Sutton, L., Van Eker, D., Zhang, E., O’Hara, N., et al. (2013). CYP2C19*17 Gain-Of-Function Polymorphism Is Associated with Peptic Ulcer Disease. Clin. Pharmacol. Ther. 93 (2), 195–203. doi:10.1038/clpt.2012.215

PubMed Abstract | CrossRef Full Text | Google Scholar

Musumba, C., Pritchard, D. M., and Pirmohamed, M. (2009). Review Article: Cellular and Molecular Mechanisms of NSAID-Induced Peptic Ulcers. Aliment. Pharmacol. Ther. 30 (6), 517–531. doi:10.1111/j.1365-2036.2009.04086.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Nicoletti, P., Carr, D. F., Barrett, S., McEvoy, L., Friedmann, P. S., Shear, N. H., et al. (2020). Beta-lactam-induced Immediate Hypersensitivity Reactions: A Genome-wide Association Study of a Deeply Phenotyped Cohort. J. Allergy Clin. Immunol. 147 (5), 1830.e15–1837.e15. doi:10.1016/j.jaci.2020.10.004

Google Scholar

Niinuma, Y., Saito, T., Takahashi, M., Tsukada, C., Ito, M., Hirasawa, N., et al. (2014). Functional Characterization of 32 CYP2C9 Allelic Variants. Pharmacogenomics J. 14 (2), 107–114. doi:10.1038/tpj.2013.22

PubMed Abstract | CrossRef Full Text | Google Scholar

Niv, Y., Battler, A., Abuksis, G., Gal, E., Sapoznikov, B., and Vilkin, A. (2005). Endoscopy in Asymptomatic Minidose Aspirin Consumers. Dig. Dis. Sci. 50 (1), 78–80. doi:10.1007/s10620-005-1281-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Olsen, A.-M. S., McGettigan, P., Gerds, T. A., Fosbøl, E. L., Olesen, J. B., Sindet-Pedersen, C., et al. (2011). Duration of Treatment with Nonsteroidal Anti-inflammatory Drugs and Impact on Risk of Death and Recurrent Myocardial Infarction in Patients with Prior Myocardial Infarction. Circulation 123 (20), 2226–2235.

PubMed AbstractGoogle Scholar

Olsen, A.-M. S., McGettigan, P., Gerds, T. A., Fosbøl, E. L., Olesen, J. B., Sindet-Pedersen, C., et al. (2020). Risk of Gastrointestinal Bleeding Associated with Oral Anticoagulation and Non-steroidal Anti-inflammatory Drugs in Patients with Atrial Fibrillation: a Nationwide Study. Eur. Heart J. Cardiovasc. Pharmacother. 6 (5), 292–300. doi:10.1093/ehjcvp/pvz069

PubMed Abstract | CrossRef Full Text | Google Scholar

Onder, G., Pellicciotti, F., Gambassi, G., and Bernabei, R. (2004). NSAID-related Psychiatric Adverse Events. Drugs 64 (23), 2619–2627. doi:10.2165/00003495-200464230-00001

PubMed Abstract | CrossRef Full Text | Google Scholar

Ong, C. K. S., Lirk, P., Tan, C. H., and Seymour, R. A. (2007). An Evidence-Based Update on Nonsteroidal Anti-inflammatory Drugs. Clinical Medicine and Research 5 (1), 19–34. doi:10.3121/cmr.2007.698

PubMed Abstract | CrossRef Full Text | Google Scholar

Patrono, C., Coller, B., FitzGerald, G. A., Hirsh, J., and Roth, G. (2004). Platelet-Active Drugs: The Relationships Among Dose, Effectiveness, and Side Effects. Chest 126 (3), 234S–264S. doi:10.1378/chest.126.3_suppl.234s

PubMed Abstract | CrossRef Full Text | Google Scholar

Paulus, H. E. (1988). FDA Arthritis Advisory Committee Meeting: Serious Gastrointestinal Toxicity of Nonsteroidal Antiinflammatory Drugs; Drug-Containing Renal and Biliary Stones; Diclofenac and Carprofen Approved. Arthritis Rheum. 31 (11), 1450–1451. doi:10.1002/art.1780311118

CrossRef Full Text | Google Scholar

Petrović, J., Pešić, V., and Lauschke, V. M. (2020). Frequencies of Clinically Important CYP2C19 and CYP2D6 Alleles Are Graded across Europe. Eur. J. Hum. Genet. 28 (1), 88–94. doi:10.1038/s41431-019-0480-8

PubMed Abstract | CrossRef Full Text | Google Scholar

PharmGKB, Pharmacogenomics Knowledgebase (PharmGKB) (2020). Gene‐specific Information Tables for CYP2C9. https://www.pharmgkb.org/page/cyp2c9RefMaterials.

PharmVar (2017). Pharmacogene Variation Consortium. https://www.pharmvar.org/.

Pilotto, A., Franceschi, M., Leandro, G., and Di Mario, F. (2003). NSAID and Aspirin Use by the Elderly in General Practice. Drugs and Aging 20 (9), 701–710. doi:10.2165/00002512-200320090-00006

PubMed Abstract | CrossRef Full Text | Google Scholar

Pilotto, A., Franceschi, M., Vitale, D. F., Zaninelli, A., Masotti, G., and Rengo, F. (2005). Upper Gastrointestinal Symptoms and Therapies in Elderly Out-Patients, Users of Non-selective NSAIDs or Coxibs. Aliment. Pharmacol. Ther. 22 (2), 147–155. doi:10.1111/j.1365-2036.2005.02537.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Pilotto, A., Seripa, D., Franceschi, M., Scarcelli, C., Colaizzo, D., Grandone, E., et al. (2007). Genetic Susceptibility to Nonsteroidal Anti-inflammatory Drug-Related Gastroduodenal Bleeding: Role of Cytochrome P450 2C9 Polymorphisms. Gastroenterology 133 (2), 465–471. doi:10.1053/j.gastro.2007.05.025

PubMed Abstract | CrossRef Full Text | Google Scholar

Pirmohamed, M., Aithal, G. P., Behr, E., Daly, A., and Roden, D. (2011). The Phenotype Standardization Project: Improving Pharmacogenetic Studies of Serious Adverse Drug Reactions. Clin. Pharmacol. Ther. 89 (6), 784–785. doi:10.1038/clpt.2011.30

PubMed Abstract | CrossRef Full Text | Google Scholar

Pirmohamed, M., Friedmann, P. S., Molokhia, M., Loke, Y. K., Smith, C., Phillips, E., et al. (2011). Phenotype Standardization for Immune-Mediated Drug-Induced Skin Injury. Clin. Pharmacol. Ther. 89 (6), 896–901. doi:10.1038/clpt.2011.79

PubMed Abstract | CrossRef Full Text | Google Scholar

Pirmohamed, M., James, S., Meakin, S., Green, C., Scott, A. K., Walley, T. J., et al. (2004). Adverse Drug Reactions as Cause of Admission to Hospital: Prospective Analysis of 18 820 Patients. BMJ 329 (7456), 15–19. doi:10.1136/bmj.329.7456.15

PubMed Abstract | CrossRef Full Text | Google Scholar

Plaza-Serón, M. d. C., García-Martín, E., Agúndez, J. A., and Ayuso, P. (2018). Hypersensitivity Reactions to Nonsteroidal Anti-inflammatory Drugs: an Update on Pharmacogenetics Studies. Pharmacogenomics 19 (13), 1069–1086. doi:10.2217/pgs-2018-0079

PubMed Abstract | CrossRef Full Text | Google Scholar

Quiralte, J., Sánchez-García, F., Torres, M.-J., Blanco, C., Castillo, R., Ortega, N., et al. (1999). Association of HLA-DR11 with the Anaphylactoid Reaction Caused by Nonsteroidal Anti-inflammatory Drugs. J. Allergy Clin. Immunol. 103 (4), 685–689. doi:10.1016/s0091-6749(99)70243-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Rainsford, K. D. (2007). Anti-inflammatory Drugs in the 21st century. Subcell Biochem. 42, 3–27. doi:10.1007/1-4020-5688-5_1

PubMed Abstract | CrossRef Full Text | Google Scholar

Rainsford, K. D., and Whitehouse, M. W. (1980). Anti-inflammatory/anti-pyretic Salicylic Acid Esters with Low Gastric Ulcerogenic Activity. Agents and Actions 10 (5), 451–456. doi:10.1007/bf01968046

PubMed Abstract | CrossRef Full Text | Google Scholar

Rekatsina, M., Paladini, A., Cifone, M. G., Lombardi, F., Pergolizzi, J. V., and Varrassi, G. (2020). Influence of Microbiota on NSAID Enteropathy: A Systematic Review of Current Knowledge and the Role of Probiotics. Adv. Ther. 37 (5), 1933–1945. doi:10.1007/s12325-020-01338-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Ricciotti, E., and FitzGerald, G. A. (2011). Prostaglandins and Inflammation. Arterioscler. Thromb. Vasc. Biol. 31 (5), 986–1000. doi:10.1161/atvbaha.110.207449

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodrigues, A. D. (2005). Impact of Cyp2C9 Genotype on Pharmacokinetics: Are All Cyclooxygenase Inhibitors the Same?: Table 1. Drug Metab. Dispos. 33 (11), 1567–1575. doi:10.1124/dmd.105.006452

PubMed Abstract | CrossRef Full Text | Google Scholar

Rogers, M. A. M., and Aronoff, D. M. (2016). The Influence of Non-steroidal Anti-inflammatory Drugs on the Gut Microbiome. Clin. Microbiol. Infect. 22 (2), 178.e1–178.e9. doi:10.1016/j.cmi.2015.10.003

CrossRef Full Text | Google Scholar

Rostom, A., Muir, K., Dubé, C., Jolicoeur, E., Boucher, M., Joyce, J., et al. (2007). Gastrointestinal Safety of Cyclooxygenase-2 Inhibitors: a Cochrane Collaboration Systematic Review. Clin. Gastroenterol. Hepatol. 5 (7), 818–828. doi:10.1016/j.cgh.2007.03.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Russell, R. I. (2001). Non-steroidal Anti-inflammatory Drugs and Gastrointestinal Damage---problems and solutions. Postgrad. Med. J. 77 (904), 82–88. doi:10.1136/pmj.77.904.82

PubMed Abstract | CrossRef Full Text | Google Scholar

Samer, C. F., Lorenzini, K. I., Rollason, V., Daali, Y., and Desmeules, J. A. (2013). Applications of CYP450 testing in the clinical setting. Mol. Diagn. Ther. 17 (3), 165–184. doi:10.1007/s40291-013-0028-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Sánchez-Diz, P., Estany-Gestal, A., Aguirre, C., Blanco, A., Carracedo, A., Ibáñez, L., et al. (2009). Prevalence of CYP2C9 polymorphisms in the south of Europe. Pharmacogenomics J. 9 (5), 306–310. doi:10.1038/tpj.2009.16

PubMed Abstract | CrossRef Full Text | Google Scholar

Sandler, R. S., Halabi, S., Baron, J. A., Budinger, S., Paskett, E., Keresztes, R., et al. (2003). A randomized trial of aspirin to prevent colorectal adenomas in patients with previous colorectal cancer. N. Engl. J. Med. 348 (10), 883–890. doi:10.1056/nejmoa021633

PubMed Abstract | CrossRef Full Text | Google Scholar

Sandson, N. B. (2015). “Uridine 5′-diphospho-glucuronoslytransferases (UGTs): Conjugating Cousins,” in A Case Approach to Perioperative Drug-Drug Interactions. Editor C. Marcucciet al. (New York, NY: Springer New York), 57–60. doi:10.1007/978-1-4614-7495-1_12

CrossRef Full Text | Google Scholar

Sarganas, G., Buttery, A. K., Zhuang, W., Wolf, I. K., Grams, D., Rosario, A. S., et al. (2015). Prevalence, trends, patterns and associations of analgesic use in Germany. BMC Pharmacol. Toxicol. 16 (1), 28. doi:10.1186/s40360-015-0028-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Schmassmann, A., Zoidl, G., Peskar, B. M., Waser, B., Schmassmann-Suhijar, D., Gebbers, J.-O., et al. (2006). Role of the different isoforms of cyclooxygenase and nitric oxide synthase during gastric ulcer healing in cyclooxygenase-1 and -2 knockout mice. Am. J. Physiology-Gastrointestinal Liver Physiol. 290 (4), G747–G756. doi:10.1152/ajpgi.00416.2005

PubMed Abstract | CrossRef Full Text | Google Scholar

Schwarz, U. I. (2003). Clinical relevance of genetic polymorphisms in the human CYP2C9 gene. Eur. J. Clin. Invest. 33 (Suppl. 2), 23–30. doi:10.1046/j.1365-2362.33.s2.6.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Shaheen, N. J., Straus, W. L., and Sandler, R. S. (2002). Chemoprevention of gastrointestinal malignancies with nonsteroidal antiinflammatory drugs. Cancer 94 (4), 950–963. doi:10.1002/cncr.10333

PubMed Abstract | CrossRef Full Text | Google Scholar

Sharma, A., Buschmann, M. M., and Gilbert, J. A. (2019). Pharmacomicrobiomics: The Holy Grail to Variability in Drug Response? Clin. Pharmacol. Ther. 106 (2), 317–328. doi:10.1002/cpt.1437

PubMed Abstract | CrossRef Full Text | Google Scholar

Shen, Z.-H., Zhu, C.-X., Quan, Y.-S., Yang, Z.-Y., Wu, S., Luo, W.-W., et al. (2018). Relationship between intestinal microbiota and ulcerative colitis: Mechanisms and clinical application of probiotics and fecal microbiota transplantation. Wjg 24 (1), 5–14. doi:10.3748/wjg.v24.i1.5

PubMed Abstract | CrossRef Full Text | Google Scholar

Shiotani, A., Murao, T., Fujita, Y., Fujimura, Y., Sakakibara, T., Nishio, K., et al. (2013). Novel single nucleotide polymorphism markers for low dose aspirin-associated small bowel bleeding. PloS one 8 (12), e84244. doi:10.1371/journal.pone.0084244

PubMed Abstract | CrossRef Full Text | Google Scholar

Shiotani, A., Murao, T., Fujita, Y., Fujimura, Y., Sakakibara, T., Nishio, K., et al. (2014). Single nucleotide polymorphism markers for low-dose aspirin-associated peptic ulcer and ulcer bleeding. J. Gastroenterol. Hepatol. 29, 47–52. doi:10.1111/jgh.12770

PubMed Abstract | CrossRef Full Text | Google Scholar

Shiotani, A., Sakakibara, T., Nomura, M., Yamanaka, Y., Nishi, R., Imamura, H., et al. (2010). Aspirin-induced peptic ulcer and genetic polymorphisms. J. Gastroenterol. Hepatol. 25, S31–S34. doi:10.1111/j.1440-1746.2009.06212.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Sigthorsson, G., Simpson, R. J., Walley, M., Anthony, A., Foster, R., Hotz–Behoftsitz, C., et al. (2002). COX-1 and 2, intestinal integrity, and pathogenesis of nonsteroidal anti-inflammatory drug enteropathy in mice. Gastroenterology 122 (7), 1913–1923. doi:10.1053/gast.2002.33647

PubMed Abstract | CrossRef Full Text | Google Scholar

Silverstein, F. E., Graham, D. Y., Senior, J. R., Davies, H. W., Struthers, B. J., and Bittman, R. M. (1995). Misoprostol Reduces Serious Gastrointestinal Complications in Patients with Rheumatoid Arthritis Receiving Nonsteroidal Anti-Inflammatory Drugs. Ann. Intern. Med. 123 (4), 241–249. doi:10.7326/0003-4819-123-4-199508150-00001

PubMed Abstract | CrossRef Full Text | Google Scholar

Silverstein, F. E., Faich, G., Goldstein, J. L., Simon, L. S., Pincus, T., Whelton, A., et al. (2000). Gastrointestinal Toxicity With Celecoxib vs Nonsteroidal Anti-inflammatory Drugs for Osteoarthritis and Rheumatoid Arthritis. JAMA 284 (10), 1247–1255. doi:10.1001/jama.284.10.1247

PubMed Abstract | CrossRef Full Text | Google Scholar

Sim, S. C., Kacevska, M., and Ingelman-Sundberg, M. (2013). Pharmacogenomics of drug-metabolizing enzymes: a recent update on clinical implications and endogenous effects. Pharmacogenomics J. 13 (1), 1–11. doi:10.1038/tpj.2012.45

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, G, and Triadafilopoulos, G (1999). Epidemiology of NSAID induced gastrointestinal complications. J. Rheumatol. Suppl. 56, 18–24.

PubMed AbstractGoogle Scholar

Singh, G., Ramey, D. R., Morfeld, D., Shi, H., Hatoum, H. T., and Fries, J. F. (1996). Gastrointestinal tract complications of nonsteroidal anti-inflammatory drug treatment in rheumatoid arthritis. A prospective observational cohort study. Arch. Intern. Med. 156 (14), 1530–1536. doi:10.1001/archinte.156.14.1530

PubMed Abstract | CrossRef Full Text | Google Scholar

Sorensen, H. T., Mellemkjaer, L., Blot, W. J., Nielsen, G. L., Steffensen, F. H., McLaughlin, J. K., et al. (2000). Risk of upper gastrointestinal bleeding associated with use of low-dose aspirin. Am. J. Gastroenterol. 95 (9), 2218–2224. doi:10.1111/j.1572-0241.2000.02248.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Sostres, C., Gargallo, C. J., Arroyo, M. T., and Lanas, A. (2010). Adverse effects of non-steroidal anti-inflammatory drugs (NSAIDs, aspirin and coxibs) on upper gastrointestinal tract. Best Pract. Res. Clin. Gastroenterol. 24 (2), 121–132. doi:10.1016/j.bpg.2009.11.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Sostres, C., Gargallo, C. J., and Lanas, A. (2013). Nonsteroidal anti-inflammatory drugs and upper and lower gastrointestinal mucosal damage. Arthritis Res. Ther. 15 (Suppl. 3Suppl 3), S3. doi:10.1186/ar4175

PubMed Abstract | CrossRef Full Text | Google Scholar

Sostres, C., and Lanas, A. (2011). Should Prophylactic Low-dose Aspirin Therapy be Continued in Peptic Ulcer Bleeding? Drugs 71 (1), 1–10. doi:10.2165/11585320-000000000-00000

PubMed Abstract | CrossRef Full Text | Google Scholar

Speed, W. C., Kang, S. P., Tuck, D. P., Harris, L. N., and Kidd, K. K. (2009). Global variation in CYP2C8-CYP2C9 functional haplotypes. Pharmacogenomics J. 9 (4), 283–290. doi:10.1038/tpj.2009.10

PubMed Abstract | CrossRef Full Text | Google Scholar

Stamer, U. M., Zhang, L., and Stüber, F. (2010). Personalized therapy in pain management: where Do we stand? Pharmacogenomics 11 (6), 843–864. doi:10.2217/pgs.10.47

PubMed Abstract | CrossRef Full Text | Google Scholar

Starodub, O. T., Demitrack, E. S., Baumgartner, H. K., and Montrose, M. H. (2008). Disruption of the Cox-1 gene slows repair of microscopic lesions in the mouse gastric epithelium. Am. J. Physiology-Cell Physiol. 294 (1), C223–C232. doi:10.1152/ajpcell.00395.2006

PubMed Abstract | CrossRef Full Text | Google Scholar

Stillhart, C., Vučićević, K., Augustijns, P., Basit, A. W., Batchelor, H., Flanagan, T. R., et al. (2020). Impact of gastrointestinal physiology on drug absorption in special populations--An UNGAP review. Eur. J. Pharm. Sci. 147, 105280. doi:10.1016/j.ejps.2020.105280

PubMed Abstract | CrossRef Full Text | Google Scholar

Stosic, R., Dunagan, F., Palmer, H., Fowler, T., and Adams, I. (2011). Responsible self-medication: perceived risks and benefits of over-the-counter analgesic use. Int. J. Pharm. Pract. 19 (4), 236–245. doi:10.1111/j.2042-7174.2011.00097.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Suzuki, T., Masui, A., Nakamura, J., Shiozawa, H., Aoki, J., Nakae, H., et al. (2017). Yogurt Containing Lactobacillus gasseri Mitigates Aspirin-Induced Small Bowel Injuries: A Prospective, Randomized, Double-Blind, Placebo-Controlled Trial. Digestion 95 (1), 49–54. doi:10.1159/000452361

PubMed Abstract | CrossRef Full Text | Google Scholar

Syer, S. D., and Wallace, J. L. (2014). Environmental and NSAID-Enteropathy: Dysbiosis as a Common Factor. Curr. Gastroenterol. Rep. 16 (3), 377. doi:10.1007/s11894-014-0377-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Szeto, C.-C., Sugano, K., Wang, J.-G., Fujimoto, K., Whittle, S., Modi, G. K., et al. (2020). Non-steroidal anti-inflammatory drug (NSAID) therapy in patients with hypertension, cardiovascular, renal or gastrointestinal comorbidities: joint APAGE/APLAR/APSDE/APSH/APSN/PoA recommendations. Gut 69 (4), 617–629. doi:10.1136/gutjnl-2019-319300

PubMed Abstract | CrossRef Full Text | Google Scholar

Tanaka, A., Araki, H., Hase, S., Komoike, Y., and Takeuchi, K. (2002). Up-regulation of COX-2 by inhibition of COX-1 in the rat: a key to NSAID-induced gastric injury. Aliment. Pharmacol. Ther. 16 (Suppl. 2), 90–101. doi:10.1046/j.1365-2036.16.s2.22.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Tanaka, A., Araki, H., Komoike, Y., Hase, S., and Takeuchi, K. (2001). Inhibition of Both COX-1 and COX-2 is required for development of gastric damage in response to nonsteroidal antiinflammatory drugs. J. Physiol. Paris 95 (1-6), 21–27. doi:10.1016/s0928-4257(01)00005-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Theken, K. N., Lee, C. R., Gong, L., Caudle, K. E., Formea, C. M., Gaedigk, A., et al. (2020). Clinical Pharmacogenetics Implementation Consortium Guideline (CPIC) for CYP2C9 and Nonsteroidal Anti‐Inflammatory Drugs. Clin. Pharmacol. Ther. 108 (2), 191–200. doi:10.1002/cpt.1830

PubMed Abstract | CrossRef Full Text | Google Scholar

Theken, K. N., Lee, C. R., Gong, L., Caudle, K. E., Formea, C. M., Gaedigk, A., et al. (2020). “Supplemental Material Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for CYP2C9 and Nonsteroidal Anti-inflammatory Drugs. CPIC GUIDELINE. Editors.

Google Scholar

Thiele, I., Clancy, C. M., Heinken, A., and Fleming, R. M. T. (2017). Quantitative systems pharmacology and the personalized drug-microbiota-diet axis. Curr. Opin. Syst. Biol. 4, 43–52. doi:10.1016/j.coisb.2017.06.001

PubMed Abstract | CrossRef Full Text | Google Scholar

To, K. F., Chan, F. K. L., Cheng, A. S. L., Lee, T. L., Ng, Y. P., and Sung, J. J. Y. (2001). Up-regulation of cyclooxygenase-1 and -2 in human gastric ulcer. Aliment. Pharmacol. Ther. 15 (1), 25–34. doi:10.1046/j.1365-2036.2001.00889.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Turner, R. M., de Koning, E. M., Fontana1, V., Thompson1, A., and Pirmohamed, M. (2020). Multimorbidity, polypharmacy, and drug-drug-gene interactions following a non-ST elevation acute coronary syndrome: analysis of a multicentre observational study. BMC Med. 18 (1), 367. doi:10.1186/s12916-020-01827-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Uijen, A. A., and van de Lisdonk, E. H. (2008). Multimorbidity in primary care: prevalence and trend over the last 20 years. Eur. J. Gen. Pract. 14 (Suppl. 1), 28–32. doi:10.1080/13814780802436093

PubMed Abstract | CrossRef Full Text | Google Scholar

Ulisse, S., Gionchetti, P., D'Alo, S., Russo, F. P., Pesce, I., Ricci, G., et al. (2001). Expression of cytokines, inducible nitric oxide synthase, and matrix metalloproteinases in pouchitis: effects of probiotic treatment. Am. J. Gastroenterol. 96 (9), 2691–2699. doi:10.1111/j.1572-0241.2001.04139.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Ulrich, C. M., Bigler, J., and Potter, J. D. (2006). Non-steroidal anti-inflammatory drugs for cancer prevention: promise, perils and pharmacogenetics. Nat. Rev. Cancer 6 (2), 130–140. doi:10.1038/nrc1801

PubMed Abstract | CrossRef Full Text | Google Scholar

Van Booven, D., Marsh, S., McLeod, H., Carrillo, M. W., Sangkuhl, K., Klein, T. E., et al. (2010). Cytochrome P450 2C9-CYP2C9. Pharmacogenetics and genomics 20 (4), 277–281. doi:10.1097/fpc.0b013e3283349e84

PubMed Abstract | CrossRef Full Text | Google Scholar

Van Hecken, A, Schwartz, JI, Depré, M, De Lepeleire, I, Dallob, A, Tanaka, W, et al. (2000). Comparative inhibitory activity of rofecoxib, meloxicam, diclofenac, ibuprofen, and naproxen on COX-2 versus COX-1 in healthy volunteers. J. Clin. Pharmacol. 40 (10), 1109–20.

PubMed AbstractGoogle Scholar

van Oijen, M. G. H., Huybers, S., Peters, W. H. M., Drenth, J. P. H., Laheij, R. J. F., Verheugt, F. W. A., et al. (2005). Polymorphisms in genes encoding acetylsalicylic acid metabolizing enzymes are unrelated to upper gastrointestinal health in cardiovascular patients on acetylsalicylic acid. Br. J. Clin. Pharmacol. 60 (6), 623–628. doi:10.1111/j.1365-2125.2005.02495.x

PubMed Abstract | CrossRef Full Text | Google Scholar

van Oijen, M. G. H., Laheij, R. J. F., Koetsier, M., de Kleine, E., te Morsche, R. H. M., van Kerkhoven, L. A. S., et al. (2006). Effect of a specific cyclooxygenase-gene polymorphism (A-842G/C50T) on the occurrence of peptic ulcer hemorrhage. Dig. Dis. Sci. 51 (12), 2348–2352. doi:10.1007/s10620-006-9475-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Vane, J. R. (1971). Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs. Nat. New Biol. 231 (25), 232–235. doi:10.1038/newbio231232a0

PubMed Abstract | CrossRef Full Text | Google Scholar

Visser, L., Vanschaik, R., Vanvliet, M., Trienekens, P., Desmet, P., Vulto, A., et al. (2005). Allelic variants of cytochrome P450 2C9 modify the interaction between nonsteroidal anti-inflammatory drugs and coumarin anticoagulants. Clin. Pharmacol. Ther. 77 (6), 479–485. doi:10.1016/j.clpt.2005.02.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Vonkeman, H. E., van de Laar, M. A. F. J., van der Palen, J., Brouwers, J. R. B. J., and Vermes, I. (2006). Allele variants of the cytochrome P450 2C9 genotype in white subjects from The Netherlands with serious gastroduodenal ulcers attributable to the use of NSAIDs. Clin. Ther. 28 (10), 1670–1676. doi:10.1016/j.clinthera.2006.10.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Wadelius, M., Chen, L. Y., Lindh, J. D., Eriksson, N., Ghori, M. J. R., Bumpstead, S., et al. (2009). The largest prospective warfarin-treated cohort supports genetic forecasting. Blood 113 (4), 784–792. doi:10.1182/blood-2008-04-149070

PubMed Abstract | CrossRef Full Text | Google Scholar

Wallace, J. L., McKnight, W., Reuter, B. K., and Vergnolle, N. (2000). NSAID-induced gastric damage in rats: Requirement for inhibition of Both cyclooxygenase 1 and 2. Gastroenterology 119 (3), 706–714. doi:10.1053/gast.2000.16510

PubMed Abstract | CrossRef Full Text | Google Scholar

Wallace, J. L. (1992). Prostaglandins, NSAIDs, and cytoprotection. Gastroenterol. Clin. North America 21 (3), 631–641. doi:10.1016/s0889-8553(21)00052-2

CrossRef Full Text | Google Scholar

Wallace, J. L. (2008). Prostaglandins, NSAIDs, and gastric mucosal protection: why doesn't the stomach digest itself? Physiol. Rev. 88 (4), 1547–1565. doi:10.1152/physrev.00004.2008

PubMed Abstract | CrossRef Full Text | Google Scholar

Walt, R., Logan, R., Katschinski, B., Ashley, J., and Langman, M. (1986). Rising frequency of ulcer perforation in elderly people in the United Kingdom. The Lancet 327 (8479), 489–492. doi:10.1016/s0140-6736(86)92940-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, L., McLeod, H. L., and Weinshilboum, R. M. (2011). Genomics and drug response. N. Engl. J. Med. 364 (12), 1144–1153. doi:10.1056/nejmra1010600

PubMed Abstract | CrossRef Full Text | Google Scholar

Weil, J., Langmana, M. J. S., Wainwrighta, P., Lawsone, D. H., Rawlinsb, M., Loganc, R. F. A., et al. (2000). Peptic ulcer bleeding: accessory risk factors and interactions with non-steroidal anti-inflammatory drugs. Gut 46 (1), 27–31. doi:10.1136/gut.46.1.27

PubMed Abstract | CrossRef Full Text | Google Scholar

Wilcox, CM, Cryer, B, and Triadafilopoulos, G (2005). Patterns of use and public perception of over-the-counter pain relievers: focus on nonsteroidal antiinflammatory drugs. J. Rheumatol. 32 (11), 2218–24.

PubMed AbstractGoogle Scholar

Wolfe, M. M., Lichtenstein, D. R., and Singh, G. (1999). Gastrointestinal toxicity of nonsteroidal antiinflammatory drugs. N. Engl. J. Med. 340 (24), 1888–1899. doi:10.1056/nejm199906173402407

PubMed Abstract | CrossRef Full Text | Google Scholar

Wongrakpanich, S., Wongrakpanich, A., Melhado, K., and Rangaswami, J. (2018). A Comprehensive Review of Non-Steroidal Anti-Inflammatory Drug Use in The Elderly. AandD 9 (1), 143–150. doi:10.14336/ad.2017.0306

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, H., Esteve, E., Tremaroli, V., Khan, M. T., Caesar, R., Mannerås-Holm, L., et al. (2017). Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat. Med. 23 (7), 850–858. doi:10.1038/nm.4345

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Y., Hu, Y., You, P., Chi, Y.-J., Zhou, J.-H., Zhang, Y.-Y., et al. (2016). Study of Clinical and Genetic Risk Factors for Aspirin-induced Gastric Mucosal Injury. Chin. Med. J. 129 (2), 174–180. doi:10.4103/0366-6999.173480

PubMed Abstract | CrossRef Full Text | Google Scholar

Xie, H.-G., Prasad, H. C., Kim, R. B., and Stein, C. M. (2002). CYP2C9 allelic variants: ethnic distribution and functional significance. Adv. Drug Deliv. Rev. 54 (10), 1257–1270. doi:10.1016/s0169-409x(02)00076-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Yip, L. Y., and Chan, E. C. Y. (2015). Investigation of Host-Gut Microbiota Modulation of Therapeutic Outcome. Drug Metab. Dispos. 43 (10), 1619–1631. doi:10.1124/dmd.115.063750

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Y., Boudreau, D. M., and Freedman, A. N. (2014). Trends in the use of aspirin and nonsteroidal anti-inflammatory drugs in the general U.S. population. Pharmacoepidemiol. Drug Saf. 23 (1), 43–50. doi:10.1002/pds.3463

PubMed Abstract | CrossRef Full Text | Google Scholar

Zimmermann, M., Zimmermann-Kogadeeva, M., Wegmann, R., and Goodman, A. L. (2019). Mapping human microbiome drug metabolism by gut bacteria and their genes. Nature 570 (7762), 462–467. doi:10.1038/s41586-019-1291-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: pharmacogenomic, NSAID (non-steroidal anti-inflammatory drug), gastrointestinal toxicities, aspirin, adverse drug reaction

Citation: McEvoy L, Carr DF and Pirmohamed M (2021) Pharmacogenomics of NSAID-Induced Upper Gastrointestinal Toxicity. Front. Pharmacol. 12:684162. doi: 10.3389/fphar.2021.684162

Received: 22 March 2021; Accepted: 11 May 2021;
Published: 21 June 2021.

Edited by:

José A G Agúndez, University of Extremadura, Spain

Reviewed by:

Ioannis S Vizirianakis, Aristotle University of Thessaloniki, Greece
Volker Martin Lauschke, Karolinska Institutet (KI), Sweden

Copyright © 2021 McEvoy, Carr and Pirmohamed. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: D. F. Carr, D.Carr@liverpool.ac.uk

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.