Sie sind auf Seite 1von 50

Expert Review of Clinical Pharmacology

ISSN: 1751-2433 (Print) 1751-2441 (Online) Journal homepage: http://www.tandfonline.com/loi/ierj20

NSAID induced gastrointestinal damage and


designing GI-sparing NSAIDs

Guillermo García-Rayado, Mercedes Navarro & Angel Lanas

To cite this article: Guillermo García-Rayado, Mercedes Navarro & Angel Lanas (2018): NSAID
induced gastrointestinal damage and designing GI-sparing NSAIDs, Expert Review of Clinical
Pharmacology, DOI: 10.1080/17512433.2018.1516143

To link to this article: https://doi.org/10.1080/17512433.2018.1516143

Accepted author version posted online: 23


Aug 2018.

Submit your article to this journal

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=ierj20
Review

NSAID induced gastrointestinal damage and designing GI-sparing


NSAIDs

Guillermo García-Rayado (1,2), Mercedes Navarro (1,2), Angel Lanas (1,2,3,4)

1. Hospital Clínico Universitario Lozano Blesa. Zaragoza. Spain

t
ip
2. IIS Aragón. Zaragoza. Spain

cr
3. CIBERehd. Madrid. Spain

4. University of Zaragoza. Zaragoza

us
an
Address Correspondence:
M

Guillermo Garcia-Rayado
ed

Servicio de Aparato Digestivo.

Hospital Clínico Universitario Lozano Blesa


pt

C/ San Juan Bosco 15


ce

50009 Zaragoza

Email: ggarciara@salud.aragon.es
Ac
Abstract

NSAIDs are widely used to treat pain and rheumatic conditions, but they induce adverse

events in different body systems, although the major, most frequent events occur in the upper

and lower gastrointestinal (GI) tracts.

Areas covered: This review is focused on damage caused by NSAIDs in the upper and lower

GI tracts, the different mechanisms of damage and the GI-sparing NSAIDs designed to

t
ip
minimize adverse events based on understanding of these mechanisms.

cr
Expert commentary: Among the new NSAIDs, COX-2 selective inhibitors have been

us
extensively investigated, and some were approved for human use. Celecoxib demonstrated its

safety for the entire GI tract, compared to traditional NSAIDs. However, coxibs, like
an
traditional NSAIDs, are toxic to the cardiovascular (CV) system. Other GI-sparing agents

include nitric oxide-NSAIDs and phosphatidylcholine-associated NSAIDs. Testing in animal


M
models and humans they showed some GI advantages over the parent NSAID compounds,

but none obtained regulatory approval or were further investigated. Hydrogen sulfide-
ed

releasing NSAIDs are currently under clinical development, and more data are needed before

clinical use. Alternative therapies, such as modulating gut microbiota, are being explored.
pt

Currently, clinicians must continue prescribing traditional NSAIDs or coxibs, associated


ce

with/without proton pump inhibitor therapy, based on the presence of GI/CV risk factors.
Ac

Key words

NSAIDs, Gastrointestinal, COX-2 selective inhibitors, Nitric oxide, Hydrogen sulfide,

Phosphatidylcholine
1. Introduction

Nonsteroidal anti-inflammatory drugs (NSAIDs) are among the most widely used drugs

worldwide. NSAIDs are largely sold by prescription and over-the-counter (OTC) for treating

fever, acute or chronic pain, and inflammatory conditions, such as rheumatic diseases. Over

30 million people take NSAIDs each day [1]. In Europe, NSAIDs represent more than 7.7%

of all prescriptions [2]. In the United States, over 70 million NSAID prescriptions are written

t
ip
annually, and with OTC use, more than 30 billion NSAID doses are consumed annually [3].

NSAID use increases among older individuals, particularly those affected by arthritis; thus,

cr
use is expected to increase with an ageing population. A survey of people aged 65 years or

us
older showed that 70% used NSAIDs at least once weekly, and 34% used NSAIDs daily [4].

However, like many other drugs, NSAIDs are associated with a broad spectrum of
an
adverse effects, including gastrointestinal (GI) injury, cardiovascular (CV) events, renal

toxicity, congestive heart failure deterioration, and blood pressure elevation, among others.
M

Recently, CV events have caused much concern [5,6], but GI toxicity is probably the most
ed

important adverse effect, due to its frequency and severity.

In this review, we focus on the damage caused by NSAIDs in the upper and lower GI
pt

tracts, the different mechanisms of damage, and the GI-sparing NSAIDs designed to avoid or

minimize GI damage.
ce

2. Mechanisms of GI tract damage


Ac

Traditional NSAIDs are lipid-soluble, weak acids [7], believed to affect the GI tract by

inhibiting the two isoforms of cyclooxygenase (COX), COX-1 and COX-2, and by inducing

topical effects. Topical effects involve detergent-like interactions with phospholipids and the

uncoupling of cellular oxidative phosphorylation [7].


2.1 Inhibition of COX-1 and COX-2

NSAIDs inhibit COXs, key enzymes in the synthesis of prostaglandins derived from

arachidonic acid. Both isoforms, COX-1 and COX-2 are inhibited by traditional NSAIDs,

which include acetylsalicylic acid (ASA) at doses above 300 mg. In contrast, selective

NSAIDs only inhibit COX-2, and low-dose aspirin only inhibits COX-1 [8].

COX-1 is present in most tissues. It is associated with hemostasis regulation, GI and

t
ip
renal tract integrity, platelet function, and macrophage differentiation. COX-1 is involved in

the synthesis of prostaglandins that stimulate the production and secretion of mucus and

cr
bicarbonate, increase mucosal blood flow, and promote epithelial cell proliferation [8].

us
COX-1 induces platelets to produce thromboxane A2, which stimulates platelet aggregation

in the presence of blood vessel damage. Blocking platelet COX-1 with aspirin is irreversible,
an
and it promotes bleeding when blood vessels are damaged [9].

In contrast, COX-2 is an inducible enzyme. In most tissues, it is expressed in


M

inflammatory conditions [8]. COX-2 is involved in the synthesis of prostaglandins that


ed

induce inflammation, pain, and fever, and are involved in cell proliferation, angiogenesis

promotion, and mucosal integrity restoration. Thus, COX-2 is the primary target of anti-
pt

inflammatory drugs. It provides pain relief with less adverse GI events, because it does not

affect COX-1-derived prostaglandins [10].


ce

NSAID inhibition of COXs has important implications in the intestinal


Ac

microcirculation. Regulation and maintenance of intestinal microcirculation involve several

mediators, including prostaglandins (one of the most relevant), leukotrienes, nitric oxide

(NO), and hydrogen sulfide (H2S) [11]. After injury, these molecules increase microvascular

blood flow. NSAID inhibition of COXs decreases this compensatory blood flow [12].

Moreover, NSAIDs induce the expression of neutrophil adhesion molecules within the

endothelium, which could mechanically compromise microvascular blood flow. As reviewed


below, in this setting, NO and H2S can be therapeutic in preventing NSAID-induced GI

damage, because they increase mucosal blood flow and reduce leukocyte adhesion to

endothelial cells [11].

Various studies have shown that the absence or selective inhibition of COX-1 reduced

the level of prostaglandins by 95% or more, without increasing intestinal permeability,

inflammation, or ulcers [13]. Similar effects were observed with short-term selective deletion

t
ip
or inhibition of COX-2 [14]. These effects were observed in animals, but not humans [13,14].

Therefore, it seems that “a priori” both isoenzymes must be inhibited to disrupt the GI

cr
mucosa. However, after this COX1/COX2 hypothesis was launched, the topical effects of

us
NSAIDs (interacting with phospholipids and uncoupling oxidative phosphorylation) and

luminal aggressors were shown to play larger roles than expected [12].
an
Therefore, COX inhibition is one of the most important factors to consider in the

pathogenesis of NSAID-induced GI (both upper and lower) damage, although it is probably


M

not the primary, the sole, or the initiating event in NSAID-induced GI damage. We must also
ed

consider topical effects and luminal aggressive factors, which vary along the tract, from the

stomach to the small bowel [12].


pt

2.2 Topical effects: phospholipid interactions and oxidative phosphorylation uncoupling


ce

Mucus performs different roles in the GI tract. It acts as a lubricant between the surface

epithelium and the luminal content, and it serves as a matrix for phospholipids that maintain
Ac

GI integrity [15]. It protects the epithelium by restricting access to different compounds, such

as digestive enzymes, bacteria, and large hydrophilic molecules. Moreover, mucus acts as a

buffer from luminal acids in the stomach [15].

Interactions between the surface epithelium and luminal aggressors, like Helicobacter

pylori, in the stomach, or bile, in the small bowel, are mediated by multiple factors. These
factors determine the production and secretion of mucus. Mucus properties, such as thickness

and composition, vary along the different GI tract regions [16].

NSAIDs interact with the mucus layer and the phospholipid bilayer of the GI tract.

NSAIDs decrease the hydrophobicity of GI mucosa, the hydrophobic lining, which leads to

mucosal exposure to different luminal aggressors, such as acid and pepsin in the stomach and

bile in the small bowel.

t
ip
On the other hand, NSAIDs uncouple mitochondrial oxidative phosphorylation, even

at millimolar concentrations [17]. This uncoupling leads to a cascade of detrimental effects

cr
that causes cellular ATP depletion and loss of intercellular junction integrity in the GI tract.

us
These effects increase mucosal permeability, and finally, apoptosis and cell death occur.

COX-2-selective agents can also uncouple oxidative phosphorylation, but at lower potency
an
than traditional NSAIDs [18].

Topical effects can initiate GI damage, but the addition of COX-1 inhibition and
M

luminal aggressors causes mucosal erosions and ulcers. Uncoupling leads to increased
ed

intestinal permeability and low-grade inflammation. When combined with reduced mucosal

prostaglandin production and the mucosal aggressors, these effects increase the severity of
pt

inflammatory and ulcerative damage.


ce

2.3 Luminal aggressive factors

Different luminal aggressors are found in the stomach (acid, pepsin, and H. pylori) and the
Ac

small bowel (bile, intestinal enzymes, and commensal bacteria). The importance of gastric

acid damage related to NSAIDs has been clearly proven as the incidence of damage, in short

and long-term users of these drugs, is reduced associating proton pump inhibitors (PPIs) or

high-dose histamine 2-receptor inhibitors [19,20]. Acid damage would depend on the

exposure of the mucosa to back diffusion of acid due to NSAIDs topical effects. On the other

hand, the role of H. pylori infections is unclear in the pathogenesis of NSAID-associated


gastric lesions. According to a recent systematic review and meta-analysis, in 2015, 4.4

billion individuals were infected with H. pylori [21]. These individuals develop different

types of gastritis, including pangastritis and antral predominant gastritis, with different levels

of acid secretion. The type of gastritis depends on the intrinsic virulence factors of the

bacteria and the host immune response, mediated by cytokines, which is genetically

determined. Gastric acid secretion is typically normal or reduced in patients with pangastritis

t
ip
and increased in patients with antral gastritis [22,23]. Gastric damage induced by short-term

NSAID use does not appear to be mediated by H. pylori, but damage induced by long-term

cr
use occurs in addition to the gastritis related to H. pylori [24]. On the other hand, gastric

us
adaptation to the short-term administration of ASA could be affected by H. pylori [25].

Depending on the gastritis induced by H. pylori, we will have different findings: exacerbation
an
of ASA induced gastric damage in patients with an increased acid secretion; or reduction of

the damage in those with an acid hyposecretion [26]. From a clinical perspective, a meta-
M

analysis concluded that long-term NSAID use and H. pylori infections were independent,
ed

additive risk factors for peptic ulcer development, but they acted separately in the ulcer

complication of bleeding [27].


pt

Bile appears to play an important role in the pathogenesis of small bowel lesions. Bile

also contributes to NSAID-induced intestinal and gastric damage, but the biochemical
ce

mechanisms have not been fully established. It was reported that the severity of NSAID
Ac

enteropathy was correlated with the amount of drug excreted in the bile and the rate of

enterohepatic circulation [28]. Indeed, a bile duct ligation almost completely abolished the

small bowel macroscopic damage from NSAIDs [29].

Some studies have suggested that the combination of bile and NSAIDs is more toxic

than either one alone [30]. For example, when bile acids were co-administered with

indomethacin in rats, the incidence and severity of gastric and small bowel damage were
significantly higher than the damage incurred by either alone [31]. On the other hand, bile

induces a biotransformation in different NSAIDs, for example diclofenac is transformed to

acyl glucuronide, which accounts for a significant part of its small bowel toxicity. Although

these conjugates can be harmful in their own way, commensal bacteria also play a role in the

metabolism of NSAID conjugates, by deconjugating them into even more toxic compounds

[32]. The interaction between biliary NSAID excretion and intestinal bacteria deconjugation

t
ip
can explain mild and distal location of NSAID enteropathy. In addition, NSAID-induced

mucosal permeability facilitates the action of bacteria. Moreover, the lipopolysaccharide

cr
components present in gram negative intestinal bacteria can activate the transmembrane toll-

us
like receptor (TLR4) present in intestinal cells. TLR4 promotes mucosal cytokine expression,

which leads to neutrophil recruitment, and finally, the release of reactive oxygen species and
an
proteases that cause mucosal injury [33].
M

3. Upper gastrointestinal damage

Upper GI tract damage was the first clinically recognized adverse effect of NSAIDs [34].
ed

About 30 to 50% of individuals that use NSAIDs exhibit endoscopic lesions, mainly in the
pt

gastric antrum, often without any clinical manifestations [35,36]. In addition, up to 40% of

individuals that use NSAIDs experience upper GI symptoms, including dyspepsia or


ce

gastroesophageal reflux [35]. However, these symptoms are not predictive of mucosal

damage, and approximately 50% of symptomatic patients exhibited normal mucosa [37]. In
Ac

contrast, over 50% of patients with serious peptic ulcer complications had no previous

warning symptoms [38]. About 1-2% of individuals that use NSAIDs experience a serious

complication, like bleeding, perforation, or obstruction. The use of NSAIDs has been

associated with a 3- to 5-fold increase in the risk of upper GI complications [39]. These
observations suggested that prevention therapies should be implemented based on the

presence of risk factors, not on the occurrence of dyspeptic symptoms [40].

Several risk factors are related to NSAID-induced GI complications (Table 1),

including a history of peptic ulcer, age >65 years, severe illness, H. pylori infection, and the

use of concomitant therapy with other potentially damaging gastrotoxic drugs, including

other NSAIDs [41]. Some risk factors can be modified, and should be taken into account

t
ip
before starting NSAID therapies. The different available NSAIDs carry different risks of GI

bleeding. A 2012 systematic review and meta-analysis of observational studies (SOS project)

cr
confirmed the variability in the risk of upper GI complications among NSAIDs used in

us
clinical practice. The pooled relative risks (RRs) were <2 for aceclofenac, celecoxib, and

ibuprofen, 2-4 for rofecoxib, sulindac, diclofenac, meloxicam, nimesulide, and ketoprofen, 4-
an
5 for tenoxicam, naproxen, indomethacin, and diflunisal, and >5 for piroxicam, ketorolac, and

azapropazone [42]. The doses and durations of individual NSAIDs that are linked to upper GI
M

damage should be addressed in future studies.


ed

Table 1. Risk factors for NSAID-related GI complications


pt

Risk factors

Age (>65 years, specially >70 years)


ce

History of peptic ulcer

Severe illness
Ac

Concomitant therapy with antiplatelet agents, anticoagulants, selective serotonin reuptake

inhibitors or corticosteroids

H. pylori infection

Use of two or more NSAIDs at the same time

Use of other gastro-erosive agents


Before prescribing NSAIDs, one should consider the concomitant use of other drugs,

such as antiplatelets, anticoagulants, or selective serotonin reuptake inhibitors (SSRIs). A

recent retrospective cohort study examined the comparative safety of individual NSAIDs

when given concomitantly with clopidogrel. They concluded that the bleeding risks of

individual NSAIDs varied more markedly than thrombotic risks, when used concomitantly

with clopidogrel. Indomethacin, diclofenac, naproxen, and rofecoxib showed statistically

t
ip
significantly higher bleeding risks than ibuprofen, but valdecoxib showed significantly

reduced bleeding risk. Moreover, in the presence of clopidogrel, the bleeding and thrombotic

cr
risks of individual NSAIDs did not appear to be inversely related [43].

us
Also of interest, a 2014 systematic review and meta-analysis of observational studies

evaluated the risk of upper GI bleeding with SSRIs, with or without concurrent NSAID use.
an
They concluded that SSRIs were associated with a modest increase in the risk of upper GI

bleeding (odds ratio [OR]: 1.66, 95% CI: 1.44-1.92), and the risk was significantly higher
M

when SSRIs were combined with NSAIDs (OR: 4.25, 95% CI: 2.82-6.42) [44].
ed

H. pylori infection and NSAID use seem to have synergistic effects on the risk of

upper GI bleeding. A meta-analysis of 16 studies showed that, among patients with H. pylori
pt

infection that used NSAIDs, the OR for peptic ulcer was 61.1 (95% CI: 9.98-373), compared

to individuals without H. pylori that did not use NSAIDs [27]. Similarly, Sostres and
ce

colleagues performed a case control study that included 1332 patients, 666 with peptic ulcer
Ac

bleeding. They concluded that NSAIDs, low-dose ASA use, and H. pylori infections were

independent risk factors for the development of peptic ulcer bleeding. However, they found

different interaction effects. They found that an H. pylori infection did not interact with

low-dose ASA use, but had additive effects with NSAID use (RR: 8, 95% CI: 5.0-12.8) [45].

Of note, the best available evidence regarding NSAID-related GI bleeding is from

randomized controlled trials with patients that used NSAIDs to manage chronic rheumatic
diseases. However, a recent Spanish study analyzed the characteristics of patients

hospitalized for upper GI bleeding in clinical practice and found that the majority reported

short-term NSAID use for reasons other than rheumatic disease. That finding suggested that

current prevention strategies may not reach a substantial population that use NSAIDs for

short-term treatments [46].

t
ip
4. Lower GI damage

Over the last decades, hospitalization rates have changed due to GI complications [47,48]. In

cr
a multicenter study, annual hospitalization rates for NSAID gastropathy increased from 0.6%

us
in 1981, to a peak of 1.5% in 1992, and then decreased to 0.5% in 2000 [47]. The latter

reduction could be explained by the use of lower NSAIDs doses, PPIs, and less toxic
an
NSAIDs, in addition to a decreasing prevalence of H. pylori infection.

Along the same lines, in 2011, Lanas and colleagues conducted a population-based
M

study and found that the incidences (per 100 000 person-years) of upper GI bleeding and

perforations decreased over time, respectively, from 54.6 and 3.9 in 1996, to 25.8 and 2.9 in
ed

2005. In contrast, the incidences (per 100 000 person-years) of colonic diverticular bleeding
pt

and angiodysplasia bleeding increased over time, respectively, from 3.3 and 0.9 in 1996, to

8.0 and 2.6 in 2005. In addition, a small increase in intestinal perforations was reported [48].
ce

Increasing evidence has suggested that NSAIDs also damage the lower GI tract,

although this damage has not been characterized as well as the damage caused in the upper
Ac

GI tract. Many recent studies have focused on the damage induced by NSAIDs in the small

bowel where mechanistic studies (see above) and evidence is now greater than that obtained

for the colon. Video capsule endoscopy (VCE) for evaluation of the small bowel helped to

improve the study of the side effects of NSAIDs in the lower GI tract. One study with

hundred twenty and forty patients on long-term NSAIDs and COX-2 selective agents
respectively and sixty healthy volunteers as controls, showed that 62% of patients on

conventional NSAIDs and 50% of patients on selective COX-2 inhibitors had abnormal VCE

studies which differed significantly (P < 0.001) from controls. The main pathology related

were reddened folds, denuded areas and mucosal breaks [49]. These findings were also found

on short-term diclofenac treatment. Many other studies are consistent with these findings

[50,51]. This NSAID effect in the small bowel might, at least in part, explain the increased

t
ip
rate of hospitalizations due to lower GI bleeding. Indeed, the prevalence of NSAID-

associated adverse effects, including both clinical and subclinical effects, might be higher in

cr
the small bowel than in the upper GI tract. In the lower GI, NSAIDs cause a wide spectrum of

us
lesions, which includes increased gut permeability, inflammation, blood loss, anemia,

malabsorption, protein loss, and mucosal ulceration (Table 2).


an
Table 2. Prevalence of main NSAID-associated lower GI side-effects. Adapted from
M

Lanas et al [52].

Increased gut permeability 44-70%


ed

Gut inflammation 60-70%

Blood loss and anemia 30%


pt

Malabsorption 40-70%
ce

Protein loss 10%

Mucosal ulceration 30-40%


Ac

The development of mucosal inflammation and increased gut permeability are the

most frequent abnormalities associated with NSAID use [53]. Small bowel mucosal

inflammation is present in 60-70% of patients that take NSAIDs, and it can be detected up to

3 years after long-term NSAID treatment has been interrupted. Inflammation is associated

with protein and blood loss, but it is often silent [54]. Increased gut permeability can be
observed 12 h after NSAID ingestion, but is not observed in NSAIDs that do not undergo

enterohepatic circulation [53]. Some studies have suggested that COX-2-selective agents do

not increase intestinal permeability [55]. NSAID use can induce erythema, mucosal

hemorrhage, erosions, and ulceration in the small bowel. Mucosal ulceration was observed in

up to 40% of patients that use NSAIDs [52], and it could be associated with occult bleeding,

major GI bleeding, or perforation [56].

t
ip
When taken long-term, NSAIDs can induce enteropathy associated with continuous,

mild blood loss, which might result in anemia or iron deficiency. The precise burden and

cr
clinical impact of this problem in patients taking NSAIDs or ASA has not been well

us
established. A systematic review of randomized trials that included 1162 subjects found that

most individuals taking NSAIDs and low-dose ASA exhibited a small average increase in
an
fecal blood loss of 1 to 2 ml/day, from about 0.5 ml/day at baseline. Some individuals lost

much more blood than average; 5% of individuals taking NSAIDs had blood losses of 5
M

ml/day or more, and 1% of individuals lost 10 ml/day or more. With daily doses of 1800 mg
ed

ASA or greater, daily blood loss of 5 ml/day and 10 ml/day occurred in 31% and 10% of

individuals, respectively [57].


pt

In 2008, the Multinational Etoricoxib and Diclofenac Arthritis Long-Term (MEDAL)

program was designed to evaluate the incidence of CV events in patients taking either
ce

etoricoxib or diclofenac. That trial also evaluated the occurrence of severe upper and lower
Ac

GI events in 34 701 patients with rheumatoid arthritis or osteoarthritis, aged 50 years or

older, with a mean therapy duration of 18 months. Lower GI complication rates (including

perforation, obstruction, or bleeding) were 0.32 and 0.38 per 100 patients, for etoricoxib and

diclofenac, respectively (HR: 0.84, 95% CI: 0.63-1.13). Bleeding occurred most commonly,

at rates of 0.19 and 0.23 per 100 patient-years, for etoricoxib and diclofenac, respectively.
Finally, a multivariable analysis revealed two significant risk factors: prior lower GI event(s)

(HR: 4.06, 95% CI: 2.93-5.62) and age above 65 years (HR: 1.98, 95% CI: 1.45-2.71) [58].

A 2014 prospective study evaluated the risk of lower GI bleeding associated with

NSAIDs, low-dose ASA, thienopyridine, and other antiplatelet drugs. They concluded that

nonselective or selective NSAID use was associated with lower GI bleeding (OR: 2.3, 95%

CI: 1.6-3.2) [59]. In addition, a recent case-control study associated NSAID use with

t
ip
increased risks of both upper (RR: 2.6, 95% CI: 2.0-3.5) and lower GI bleeding (RR: 1.4,

95% CI: 1.0-1.9) [60].

cr
Complicated colonic diverticular disease is another clinical adverse effect associated

us
with NSAIDs in the lower GI tract. In 2011, a cohort study that included 47 210 men, ages

40-75 years at baseline, identified 939 cases of diverticulitis and 256 cases of diverticular
an
bleeding during a 22-year follow-up. After adjusting for risk factors, individuals that

regularly used NSAIDs had an increased risk of diverticulitis (HR: 1.72, 95% CI: 1.40-2.11)
M

and diverticular bleeding (HR: 1.74, 95% CI: 1.15-2.64), compared to men that denied using
ed

these medications [61]. That finding was confirmed in a 2014 systematic review and meta-

analysis of observational studies; they concluded that ASA and NSAID use were strongly and
pt

consistently associated with an increased risk of colonic diverticular bleeding (RR: 2.48, 95%

CI: 1.86-3.31) [62]. In addition, a prospective study investigated the risk factors associated
ce

with transfusion, further bleeding, and prolonged length of stay in patients with acute
Ac

episodes of diverticular bleeding. Multivariate analyses revealed that NSAID use (OR: 5.9;

p=0.04) and stigmata of bleeding (OR: 11; p <0.01) were significant risk factors for further

bleeding. Moreover, age >70 years (OR: 2.1; p =0.04) and NSAID use (OR: 2.7; p =0.03)

were independent risk factors for prolonged hospitalization [63].


5. Designing GI-sparing NSAIDs

Information gained over the last decades on the biochemical characteristics of NSAIDs and

the mechanism of damage to the GI tract mucosa has initiated a search for NSAIDs with low

or no capacity to induce gastric or intestinal mucosal damage. Multiple studies have

investigated new or complex compounds, but few have passed testing in humans, and much

fewer have been commercialized or used in clinical practice.

t
ip
5.1 COX-2-selective agents

cr
The discovery of the gene encoding the COX-2 iso-enzyme opened the door to the

development of NSAIDs that selectively inhibited COX-2, or preferentially inhibited COX-2

us
over COX-1, at doses commonly used in clinical practice. Selective and preferential COX-2
an
inhibitors induce pain relief, reduce inflammation, and cause fewer GI adverse events than

traditional NSAIDs. Although COX-1 inhibition is not the only mechanism involved in
M
NSAID-related GI toxicity, these data reinforced the hypothesis that COX-1 inhibition is

important in NSAID-induced GI damage.


ed

NSAIDs are classified according to their specific inhibition of COX-2 [64].

“Preferential” COX-2 inhibitors, such as meloxicam, mainly inhibit COX-2, but they show
pt

some inhibition of COX-1 at clinically effective doses. “Selective” COX-2 agents (also called
ce

coxibs) only inhibit COX-2 at clinically effective doses, but they may inhibit COX-1 at high

doses (Table 3). Coxibs on the market include celecoxib and etoricoxib; other coxibs, such as
Ac

rofecoxib or lumiracoxib, were withdrawn from the market due to concerns about increased

CV risk (rofecoxib) or liver damage (lumiracoxib) [65].

Table 3. NSAID selectivity according to COX-1 and COX-2 inhibition

Dual inhibition COX-1 and COX-2 Naproxen, ibuprofen, ketoprofen,

indomethacin, piroxicam, diclofenac


COX-1 Selective inhibition Low-dose ASA

COX-2 Preferential inhibition Nimesulide, meloxicam, nabumetone

COX-2 Selective inhibition Celecoxib, rofecoxib

lumiracoxib, valdecoxib, etoricoxib

Coxib effects on the GI have been characterized in large GI outcome studies over the

t
last 15 years . A systematic review of randomized controlled trials showed that, compared to

ip
non-selective NSAIDs, coxibs produced significantly fewer gastroduodenal ulcers (RR: 0.26,

cr
95% CI: 0.23-0.3), fewer clinically important ulcer complications (RR: 0.39, 95% CI: 0.31-

us
0.5), and better GI tolerability [66]. The CONDOR trial was a multi-center, double-blind

study. They randomly assigned 4484 patients to receive celecoxib or diclofenac slow release
an
plus PPI, and evaluated the risk of GI events in each group. The risk of clinically significant

upper and lower GI events was higher with diclofenac plus PPI than with celecoxib (HR: 4.3,
M

95% CI: 2.6–7.0; p<0.0001). Importantly, patients taking concomitant aspirin were excluded

from the trial [67].


ed

A common clinical scenario is the patient that takes low-dose ASA for primary or,

more commonly, secondary CV prevention and also needs to take NSAIDs for an arthritic
pt

condition. Low-dose ASA acts as a selective COX-1 inhibitor [10] and the combination of
ce

coxib plus low-dose ASA should act as a dual COX inhibitor. This scenario raised the

question of whether low-dose ASA combined with a coxib was better or worse than low-dose
Ac

ASA combined with a traditional NSAID. Evidence from several studies [67-70], suggested

that co-administration of ASA reduced the GI-safety of coxibs. However, a more recent

meta-analysis of randomized controlled trials showed that coxib plus low-dose ASA was

associated with a lower risk of upper GI complications, compared to non-selective NSAIDs

plus low-dose ASA [71]. Nevertheless, those data must be interpreted with caution, because

patients had not been randomly assigned to the different groups, and the risk of bias could not
be ruled out. The PRECISION (Prospective Randomized Evaluation of Celecoxib Integrated

Safety versus Ibuprofen or Naproxen) trial evaluated the global safety (i.e., major adverse CV

events, noncardiovascular death, and GI or renal events) of naproxen, ibuprofen, and

celecoxib when co-administered with low-dose ASA. Without aspirin, celecoxib had a better

overall safety profile than the other drugs. This difference was attenuated by adding ASA, but

celecoxib remained associated with fewer renal events than ibuprofen and fewer GI events

t
ip
than ibuprofen or naproxen [72].

Prior to choosing a NSAID, one should weigh the GI benefits of selective COX-2

cr
agents against the known CV risks, although CV toxicity is not exclusively a property of

us
selective COX-2 NSAIDs. The mechanism of CV damage remains unclear, though it might

be partly explained by the inhibitory effect of traditional NSAIDs and coxibs on the COX-2-
an
mediated endothelial cardioprotective pathway. These drugs counteract the COX-2 inhibition

of PGI2 in the vasculature, which releases the constraints on platelet function, and leads to a
M

prothrombotic situation. Moreover, a recent study showed that NSAIDs potently induced
ed

reactive oxygen species (ROS) in cardiac cells, which could lead to cardiotoxicity through a

ROS-dependent mechanism involving mitochondrial and proteasome dysfunction [73].


pt

Unlike most other NSAIDs, naproxen, particularly at high doses (500 mg), causes an

antiplatelet effect through a persistent, but reversible, inhibition of platelet COX-1 activity.
ce

This naproxen effect might improve its CV tolerability, but also increase the risk of GI
Ac

adverse events [10].

In a 2013 meta-analysis of randomized trials that evaluated the vascular and upper GI

effects of NSAIDs, naproxen did not significantly increase major vascular events (RR: 0.93,

95% CI: 0.69-1.27). However, vascular death was increased significantly by coxibs (RR:

1.58, 99% CI 1.0-2.49; p=0.0103) and diclofenac (RR: 1.65, 0.95-2.85; p=0.0187), only non-
significantly by ibuprofen (RR: 1.9, 0.56-6.41; p=0.17), and naproxen had no effect (RR:

1.08, 0.48-2.47; p=0.8) [5].

The PRECISION trial also compared the CV safety of celecoxib, ibuprofen, and

naproxen in patients that needed NSAIDs for treating rheumatic diseases. That study mainly

aimed to assess the non-inferiority of celecoxib with regard to CV death, nonfatal stroke, or

myocardial infarction. Celecoxib at moderate doses was found to be non-inferior to ibuprofen

t
ip
or naproxen with regard to CV safety (HR for celecoxib vs. naproxen, 0.93, 95% CI: 0.76-

1.13; HR for celecoxib vs. ibuprofen, 0.85, 95% CI: 0.70-1.04; P<0.001 for non-inferiority in

cr
both comparisons) [6]. As a secondary objective, they evaluated GI safety. Patients taking

us
NSAIDs plus PPI had infrequent clinically significant GI events. Celecoxib plus PPI had

better overall GI safety compared to ibuprofen or naproxen, despite the co-administration of


an
corticosteroids or low-dose ASA [74]. However, that trial had several limitations: two-thirds

of the patients interrupted treatment with the assigned drug; almost 30% of patients were lost
M

to follow-up; and patients assigned to naproxen and ibuprofen received the highest doses
ed

within the protocol-specified range. These factors could have led to overestimations of

adverse event rates with these drugs [75]. A post-hoc analysis of this trial reported that
pt

naproxen or ibuprofen experienced a significantly higher risk of major toxicity than those that

used celecoxib [76].


ce

Finally, another recent double-blind randomized trial (CONCERN study) evaluated


Ac

the GI safety of celecoxib versus naproxen in patients at high risk of both CV and GI events.

The 514 patients enrolled received either celecoxib 100 mg twice per day plus PPI or

naproxen 500 mg twice per day plus PPI for 18 months. The cumulative incidence of

recurrent bleeding was significantly higher (p=0.008) with naproxen (12.3%, 95% CI: 8.8-

17.1) than with celecoxib (5.6%, 95% CI: 3.3-9.2). The appearance of serious CV events was

not significantly different between groups, though that was not the endpoint of the study [77].
The major criticism of the trial was the different doses of celecoxib (low) and naproxen

(high).

In summary, the available evidence favors the use of coxibs instead of traditional

NSAIDs, when considering the safety of the entire GI tract. However, the use of coxibs

should be balanced against the CV risk, particularly in patients at high risk of CV disease.

When needed, one should prescribe the shortest duration and lowest effective dose of either a

t
ip
traditional NSAID or a coxib [75].

cr
5.2 Nitric oxide-, carbon monoxide- and hydrogen sulfide-releasing NSAIDs

To counteract the harmful effects on intestinal microcirculation produced by NSAIDs, a

us
traditional NSAID can be coupled with a donor molecule that releases a gaseous mediator,
an
like NO or H2S. Reports of adverse CV effects associated with coxibs motivated the

investigation and development of compounds coupled with NO and H2S, which are
M
cardioprotective agents. NO- and H2S-releasing drugs can prevent endothelial dysfunction

and myocardial ischemia and they provide significant benefits to patients with congestive
ed

heart failure [78,79]. Moreover, H2S has anti-atherosclerotic effects, by inhibiting neointimal

hyperplasia, vascular calcification, and vascular inflammation [80]. These beneficial vascular
pt

effects extend to the GI tract; NO and H2S reduce NSAID-induced GI damage by increasing
ce

mucosal microvascular blood flow. In addition, these compounds increase mucus and

bicarbonate secretion in the GI tract and reduce leukocyte adhesion to endothelial cells [81]
Ac

(Table 4).

Table 4. Biological effects of NO, H2S and CO in the GI tract.

Effect on mucosal blood flow Increased

Effect on bicarbonate and mucus secretion Increased

Effect on acid secretion Minimal effect


Cytoprotection Yes

Effect on leukocytes adhesion to endothelial cells Reduced

Aspirin and several NSAIDs, like naproxen or diclofenac, have been coupled to a

nitroxybutyl or nitrosothiol moiety to generate molecular hybrids that release NO [11]. These

NO-releasing NSAIDs have been investigated in preclinical studies and clinical trials.

t
ip
Fiorucci and colleagues showed, in 40 healthy subjects, that NO-aspirin decreased gastric and

duodenal endoscopic injury after 7 days of treatment [82]. However, the most investigated

cr
NO-releasing NSAID is NO-naproxen (also called naproxcinod).

us
In preclinical studies, NO-naproxen caused less upper GI damage than naproxen. For

example, in a proof-of-concept study with 31 healthy volunteers, NO-naproxen (AZD3582)


an
or naproxen was administered at equimolar doses for 12 days. The mean total number of

gastroduodenal erosions was 11.5 with naproxen and 4.1 with NO-naproxen (p <0.0001), and
M

more than half the NO-naproxen group had no erosions [83]. Despite these promising initial

results, phase II studies could not confirm a lower gastrolesive profile for NO-naproxen. In a
ed

double blind phase II trial [84], 970 patients with hip or knee osteoarthritis were randomized
pt

to 750 mg AZD3582 twice daily, 500 mg naproxen twice daily, or placebo twice daily.

Although AZD3582 was favored for some secondary endpoints, like the endoscopy Lanza
ce

score, the primary end point (6-week endoscopic gastroduodenal ulcer incidence) was not

significantly different between AZD3582 and naproxen. The incidence of gastroduodenal


Ac

ulcers was 9.7% with AZD3582 and 13.7% with naproxen (p = 0.07, not significant).

Because long-term clinical trials did not show a significant difference between these two

drugs, the FDA denied approval of AZD3582 in 2010. In addition, the FDA requested a

broader safety assessment of NO-naproxen [11] that covered other adverse effects, such as

drops in blood pressure, which could have a major impact in older individuals.
H2S can have beneficial effects on different systems damaged by NSAIDs (Figure 1).

In the GI tract plays a key role in maintaining mucosal integrity and it can protect the

stomach from damage [85]. Various NSAIDs have been designed, for example, H2S was

coupled with diclofenac, aspirin, and naproxen. These compounds reduced GI toxicity in

rodent models by preventing reductions in gastric mucosal blood flow [86], inhibiting tumor

necrosis factor-α expression, and inhibiting leukocyte adherence to vascular endothelium

t
ip
[87]. H2S might also confer gastroprotection by upregulating the Nrf-2/HO-1 pathway [88].

H2S-naproxen (ATB-346) was also tested in rats with compromised gastric mucosal defense

cr
systems, which mimicked the use of NSAIDs by humans at high risk. ATB-346 did not

us
induce significant GI damage; in fact, it accelerated the healing of pre-existing gastric ulcers

in these models of impaired mucosal defense [89,90]. In addition to the upper GI tract, H2S-
an
donor treatments reduced NSAID-induced enteropathy in rats [91], promoted the resolution

of colonic inflammation [92], and prevented intestinal polyp formation in an APCMin+mouse


M

model [93].
ed

However, H2S-releasing NSAIDs must be assessed in clinical trials to determine

whether their GI safety might diminish in long-term real life settings. Currently, several
pt

proof-of-concept clinical studies with ATB-346 have been initiated in humans, mainly by the

Antibe Therapeutics company [79,94]. A phase 2A effectiveness study in patients with


ce

osteoarthritis of the knee showed that ATB-346 (250 mg once daily) reduced pain with
Ac

equivalent or better efficacy than that provided by naproxen or celecoxib. In a phase 2B GI

safety study, ATB-346 was tested in 244 healthy volunteers. One group received ATB-346

(250 mg once daily), and the other group received naproxen (500 mg twice daily) for 14 days.

Results were announced by Antibe in March, 2018. The gastric or duodenal ulceration rate

was 2.5% (3/118) with ATB-346 and 42.1% (53/126) with naproxen (p<0.001). Other H2S-
releasing NSAIDs, like ketoprofen for acute pain (ATB-352) and low-dose ASA (ATB-340),

are currently under investigation by the same company in pre-clinical stages [94].

A third gaseous mediator of GI mucosal integrity is carbon monoxide (CO). CO

production occurs via the enzymes, hemoxygenase-1 (HO-1, constitutive) and

hemoxygenase-2 (HO-2, inducible). CO is more biologically stable than NO and H2S. It acts

as a signaling molecule, and it has anti-inflammatory and cytoprotective properties. An

t
ip
important limitation to studying CO effects in vivo has been the lack of suitable CO donors

[81]. The most useful tools are a group of molecules called ‘‘CORMs’’ (CO-releasing

cr
molecules), developed in the laboratory of R. Motterlini [95]. A pharmacological CO donor,

us
known as the tricarbonyldichlororuthenium (II) dimer (CORM-2), protected against gastric

damage and accelerated the healing of pre-existing gastric ulcers in animal models [96,97]. In
an
animal models of ethanol-induced gastric damage and administration of different NSAIDs,

CORM-2 pretreatment reduced the number of gastric lesions and improved gastric blood flow
M

[96]. However, human studies on CO in GI diseases remain limited.


ed

5.3 Phosphatidylcholine-associated NSAIDs

As highlighted above, COX inhibition is not the only mechanism of NSAID-induced GI


pt

damage. Prostaglandin-independent mechanisms are also important; mainly the interaction


ce

between NSAIDs and the phospholipids of GI layers. The most abundant phospholipid in the

GI mucosa is phosphatidylcholine (PC), which plays a key role in preserving the hydrophobic
Ac

GI barrier. When NSAIDs disrupt phospholipid layers, luminal aggressors can gain access to

the epithelium [98]. On the other hand, PC is the major phospholipid in bile and able to

prevent bile injury in the lower GI tract. NSAIDs combined with bile salts increase GI

damage by forming toxic mixed micelles. In addition, NSAIDs may bind to PC, which

increases the amount of unbound toxic bile acid [99].


Extensive studies and associated knowledge gained on this mechanism of topical

damage induced by NSAIDs opened the door to the development of therapeutic strategies

based on PC co-administration. New PC-associated NSAIDs (PC-NSAIDs) were designed as

safer drugs for the upper and lower GI tracts that lacked the potential CV damage associated

with COX-2 selective inhibitors. PC-NSAIDs were tested first in animal models and later in

human pilot clinical trials, mostly conducted by Lichtenberger and colleagues [100-106].

t
ip
In rodent models, oral or parenteral administration of PC-NSAIDs reduced the

NSAID-induced GI injury without affecting therapeutic activity [100,101]. In an

cr
experimental study, rats received acute intravenous or chronic subcutaneous PC-associated

us
indomethacin or indomethacin alone. PC-indomethacin produced less GI bleeding and

intestinal damage than indomethacin alone, but they had similar analgesic and anti-
an
inflammatory activities [101]. These protective effects were also investigated with a coxib

combined with aspirin treatment in rats. The gastric toxicity of aspirin combined with a coxib
M

was obviated with PC-associated aspirin, due to the maintenance of the stomach’s
ed

hydrophobic properties. Moreover, aspirin and PC-aspirin equally inhibited gastric mucosal

PGE2 concentrations [102]. PC also reduced the small bowel damage induced by
pt

indomethacin in rats. Administration of PC-indomethacin resulted in significantly fewer

erosions (>50% reduction) and bleeding (>80% reduction), compared to indomethacin alone.
ce

However, neither omeprazole nor lansoprazole protected against indomethacin-induced small


Ac

bowel injury [103].

Those results in animals motivated evaluations of PC-NSAIDs in humans. PC-aspirin

and PC-ibuprofen have been tested. In a pilot randomized, double-blind study [104], 16

healthy volunteers received aspirin or PC-aspirin for 3 days. Endoscopy on day 4 showed

significantly fewer gastric erosions with PC-aspirin than with aspirin alone (2.9 ± 4.3 vs. 8.7

± 10.7; p <0.025). In a larger, randomized, single blind, active-controlled study, 204 healthy
subjects (ages 50-74 years) received 7-day treatments with once daily 325 mg aspirin or PC-

aspirin [105]. Gastroduodenal erosions and/or ulcers were observed in 42.2% of the aspirin-

treated group and 22.2% of the PC-aspirin-treated group (p=0.0027). Finally, PC-ibuprofen

was also assessed in humans [106]. In this randomized, double-blind trial, 125 patients with

osteoarthritis received 2400 mg/day ibuprofen or an equivalent dose of PC-ibuprofen for 6

weeks. A trend was observed in improved GI safety in the PC- ibuprofen group compared to

t
ip
the ibuprofen group, but the difference was not statistically significant. However, in patients

over age 55 years, PC-ibuprofen treatment provided a significant benefit compared to

cr
ibuprofen in the prevention of NSAID-induced upper GI damage. Therefore, PC-ibuprofen

us
reduced upper GI injury, but only in patients at risk that were over 55 years old. These

satisfactory results with PC-NSAIDs in humans should be confirmed in long-term studies that
an
assess clinical GI events.

5.4 Other GI-sparing NSAIDs


M

In addition to coxibs and combinations of NSAIDs with gaseous mediators and


ed

phosphatidylcholine, other GI-sparing NSAIDs have been studied but most of them are in a

preclinical phase and have only been tested in animal models. Examples of these are
pt

encapsulated NSAIDs and combinations of glucosamine hydrochloride (GS-HCl) and

NSAID.
ce

Various vectors as polymer-based nanoparticles, liposomes and solid lipid


Ac

nanoparticles have been designed to encapsulate NSAIDs and decrease their GI side effects.

Both nanoparticles and liposomes have been studied in rodents or in rabbits. Most common

actives that have been tested in animals include flurbiprofen, diclofenac, indomethacin and

oxicams and the most targeted ways of administration routes were ocular and intravenous.

Some outcomes of these in vivo studies showed a bioavailability enhancement, improvement

of drug accumulation in targeted site and prolonged release with a well security profile [107].
Although it will be necessary to conduct similar studies in humans and specifically assess GI

damage outcomes. On the other hand, GS-HCl in combination with NSAID has been tested in

mice to try decrease GI injury of NSAIDs. Combination of GS-HCl and indomethacin

showed significantly decrease of gastric lesions in comparison with indomethacin alone. This

GI sparing effect could be due to inhibit COX-2 N-glycosylation by GS-HCl. GS-HCl

significantly attenuated TNF-a-induced COX-2 expression and also significantly reduced the

t
ip
expressions of other molecules like ICAM-1, VCAM-1, IL-8 and IL-1b [108].

cr
us
6. Expert commentary

The analgesic and anti-inflammatory efficacies of NSAIDs are well proven. NSAIDs remain
an
one of the most common prescriptions worldwide; millions of people take NSAIDs every

day, and NSAIDs will probably remain in use for years. In addition, NSAIDs have potential
M

anti-neoplastic effects, and they prevent colorectal adenomas. Unfortunately, like most

prescribed medications, their use is associated with adverse effects. This review described
ed

NSAID-induced adverse effects in the GI, CV, hepatic, and renal systems. Short- and long-
pt

term use of NSAIDs can produce upper and lower GI damage, mainly in patients with risk

factors, which include a history of peptic ulcer, concomitant therapy with antiplatelet or
ce

anticoagulant agents, older age, and severe comorbidity. NSAIDs-induced GI damage arises

from COX inhibition and topical effects, in addition to luminal GI aggressor actions. The
Ac

principal luminal aggressors include acid, pepsin, and H. pylori, in the upper GI tract, and

bile and bacteria, in the lower GI tract [12].

Investigations have focused for years on finding compounds to minimize adverse

effects, mainly in the GI tract. Several compounds were designed based on the growing

knowledge of the damage mechanisms (Figure 2). The most important development was the
design and introduction of COX-2-selective inhibitors. Coxibs were shown to be safer than

traditional NSAIDs in both the upper and lower GI tracts, but they were toxic to the CV

system. This discovery led to the finding that essentially all NSAIDs could induce CV events,

at least partly due to their effects on COX-2-dependent prostacyclin endothelial secretion.

After decades of NSAID use, this CV effect was not clearly detected until coxibs were

introduced to the market. This prompted the development of new guidelines that included

t
ip
both GI and CV risk factors for patients that needed NSAIDs. However, the beneficial effect

of coxibs in the GI tract, compared to traditional NSAIDs, was reduced when they were co-

cr
administered with low-dose ASA, a common clinical scenario. Consequently, because coxibs

us
did not fulfill initial expectations, other compounds with the potential to reduce both upper

and lower GI adverse events were developed and clinically tested, after successful preclinical
an
studies (Table 5).
M

Table 5: GI and CV risk associated to different NSAIDs. Evidence in humans with


ed

GI-sparing agents different to coxibs is limited or non-existing. The risk expressed are

estimations based on their parent compounds and/or animal data (*).


pt

CV: Cardiovascular. COX: Cyclooxygenase. GI: Gastrointestinal. H2S: Hydrogen

sulfide. PC: Phosphatidylcholine. NO: Nitric oxide. NSAIDs: Non-steroidal anti-


ce

inflammatory drugs.
Ac

GI Risk CV Risk

Traditional NSAIDs ++ ++
(except naproxen)

Naproxen ++ +

COX-2 selective agents + ++

NO and H2S-NSAIDs + +

PC-NSAIDs + ++
One of those compounds, naproxcinod, reached a significant clinical development,

because it combined the NSAID with the best CV profile with NO, an agent shown to be

beneficial for both the GI and CV systems. Unfortunately, in phase III studies, naproxcinod

did not show sufficient advantage over the current drugs, and both the EMA and FDA raised

safety concerns. Other compounds, like PC-associated NSAIDs or PC-aspirin, were also

t
ip
tested in humans with good results, but longer-term studies that assess clinically relevant GI

events are lacking. No new studies or applications for marketing approval are on the horizon.

cr
Currently, the only of new compounds undergoing clinically testing are H2S-releasing

us
NSAIDs. In a very recent phase 2B GI safety study, H2S-naproxen showed significantly

lower gastric and duodenal ulceration rates than naproxen alone in a 14-day treatment [94].
an
We must await results from clinical studies to ascertain whether these new H2S-releasing

NSAIDs will be given the green light to be marketed as a new drug for patients that need
M

NSAIDs.
ed

Currently, clinicians must choose from the available strategies, although other

options, different from GI-sparing NSAIDs, are being tested. One strategy for reducing
pt

NSAID-induced GI damage is to interfere with luminal aggressors. In the upper GI tract, the

most important aggressor is gastric acid. Indeed, PPIs were shown to be highly effective for
ce

reducing NSAID-related complications in the upper GI tract in all populations tested


Ac

[23,109,110]. However, PPIs do not reduce NSAID-induced lower GI injuries. In fact, they

might exacerbate small intestinal damage by inducing dysbiosis [111]. In addition, some

concern remains about the potential side effects of PPIs in long-term use and in drug

interactions. However, much of the current evidence linking PPI use to serious long-term

adverse consequences is weak and not well substantiated [112].


To avoid or minimize NSAID-induced lower GI damage, new strategies, other than

COX-2 selective agents, are needed. Again, it might be beneficial to interfere with luminal

aggressors; in this case, bile and bacteria play relevant roles in the lower GI tract [12].

Bacterial β-glucuronidase in bile can deconjugate NSAIDs, and metabolites can be toxic to

GI mucosa. Some animal studies have shown that specific inhibitors of bacterial β-

glucuronidase [113,114] could reduce small bowel lesions induced by NSAIDs. These

t
ip
compounds have not been tested in human clinical trials. Other investigators have tested

antibiotics and/or probiotics in animal models and humans. One study in thirteen patients

cr
showed that metronidazole reduced intestinal inflammation and blood loss in NSAID-induced

us
enteropathy [115]. However, extensive clinical use of these drugs is limited by potential

negative effects on intestinal microbiota and the occurrence of extragastrointestinal drug


an
resistance. One recent proof-of-concept study was conducted with rifaximin, a poorly

absorbed, effective drug against enterobacteria [116]. In that placebo-controlled study, 60


M

healthy volunteers (median age 26 years) were given diclofenac plus omeprazole, with either
ed

rifaximin (400 mg) or placebo, twice daily for 14 days. Subjects were assessed with video-

capsule endoscopy. Mucosal breaks were detected in 20% of subjects given rifaximin and
pt

43% of subjects given placebo. None of the subjects in the rifaximin arm developed large

lesions, but 9 volunteers developed large lesions in the placebo arm (p<0.001). To confirm
ce

the clinical usefulness of rifaximin, a phase III study should be conducted in patients given
Ac

long-term NSAID treatments. Finally, other studies have tested probiotics, like VSL-3 and

Lactobacillus casei [117,118], for reducing NSAID-induced enteropathy in patients.

However, larger, more consistent clinical trials with probiotics are necessary to translate their

use into clinical practice.

7. Five-year view
Over the past decades, we have observed an important reduction in NSAIDs-induced peptic

ulcer disease [48, 119], mainly due to the widespread use of anti-secretory drugs, like PPIs,

and a reduction in H. pylori prevalence. However, the occurrence of lower GI damage and

complications associated with NSAIDs has become more apparent, and it presents a clear

clinical problem. From that perspective, we need new compounds and/or new therapeutic

strategies that aim to prevent not only upper but also lower GI tract damage these events.

t
ip
Some of the GI-sparing agents developed in the last decades have fulfilled partially this target

(coxibs) and others failed or have not been approved for human use. In 5 years time we will

cr
know whether the new H2S-releasing NSAIDs may be one of those compounds, and will

us
have a better understanding of the role of the microbiome on the damage or protection of the

gastrointestinal tract in patients taking NSAIDs. More effort and funding are needed to
an
investigate both the mechanisms of damage and to develop novel treatment strategies that

target the whole GI tract.


M
ed

8. Key issues

• NSAIDs are associated with a broad spectrum of adverse effects, but GI toxicity is the
pt

most important, due to its frequency and severity. NSAIDs cause damage in the upper

but also in the lower GI tract.


ce

• COX inhibition is one of the most important factors to consider in the pathogenesis of

NSAID-induced GI damage, although we must also consider topical effects of


Ac

NSAIDs and luminal aggressive factors.

• Coxibs are safer than traditional NSAIDs in both the upper and lower GI tracts, but

they are toxic to the CV system, therefore the use of coxibs should be balanced

against the CV risk.


• NO-releasing NSAIDs and PC-associated NSAIDs showed a better GI profile over

the parent NSAID compounds in animals and humans studies, but they did not show

sufficient advantages and did not obtained regulatory approval or were further

investigated.

• H2S-naproxen is currently under clinical development and has showed significantly

less GI damage in a recent phase 2B GI safety study, although more data are needed

t
ip
before clinical use.

cr
Lower GI damage associated with NSAIDs has become more important, and novel

treatment strategies that target NSAID-induced lower GI tract should be developed.

us
Larger and more consistent clinical trials with antibiotics like rifaximin or probiotics

that modulate gut microbiota are needed.


an
Funding

This paper was not funded.


M

Declaration of Interest
ed

The authors have no relevant affiliations or financial involvement with any organization or

entity with a financial interest in or financial conflict with the subject matter or materials
pt

discussed in the manuscript. This includes employment, consultancies, honoraria, stock

ownership or options, expert testimony, grants or patents received or pending, or royalties.


ce

Reviewer disclosures
Ac

Peer reviewers on this manuscript have no relevant financial or other relationships to

disclose.
References

Papers of special note have been highlighted as:


* of interest
** of considerable interest

1. Singh G, Triadafilopoulos G. Epidemiology of NSAID induced gastrointestinal

complications. J Rheumatol. 1999;56(Suppl):18-24.

2. Jones R. Nonsteroidal anti-inflammatory drug prescribing: past, present and future.

t
ip
Am J Med. 2001;110:4S-7S.

3. Wiegand T, Lawrence R, Vernetti C. Nonsteroidal anti-inflammatory drug (NSAID)

cr
toxicity. 2016. http://emedicine.medscape.com/article/816117-overview (accesed may

us
30, 2018).

4. Talley NJ, Evans JM, Fleming KC, et al. Nonsteroidal antiinflammatory drugs and
an
dyspepsia in the elderly. Dig Dis Sci. 1995;40:1345-50.

5. Coxib and Traditional NSAID Trialists’ (CNT) Collaboration, Bhala N, Emberson J,


M

Merhi A, et al. Vascular and upper gastrointestinal effects of non-steroidal anti-


ed

inflammatory drugs: meta-analyses of individual participant data from randomised

trials. Lancet. 2013;382:769-779.


pt

6. Nissen SE, Yeomans ND, Solomon DH, et al. Cardiovascular safety of celecoxib,

naproxen, or ibuprofen for arthritis. N Engl Med. 2016;375:2519-2529.


ce

** A remarkable trial concerning the CV safety of different NSAIDs.

7. Brune K, Glatt M, Graf P. Mechanisms of action of anti-inflammatory drugs. Gen


Ac

Pharmacol. 1976;7:27-33.

8. Smith WL, DeWitt DL, Garavito RM. Cyclooxygenases: structural, cellular, and

molecular biology. Annu Rev Biochem. 2000, 69:145-82.


9. Patrono C, Patrignani P, García Rodríguez LA. Cyclooxygenase-selective inhibition

of prostanoid formation: transducing biochemical selectivity into clinical read-outs. J

Clin Invest. 2001;108:7-13

10. Tacconelli S, Bruno A, Grande R, et al. Nonsteroidal anti-inflammatory drugs and

cardiovascular safety - translating pharmacological data into clinical readouts. Expert

Opin Drug Saf. 2017;16:791-807

t
ip
11. Fiorucci S, Distrutti E. COXIBs, CINODs and H₂S-releasing NSAIDs: current

perspectives in the development of safer non steroidal anti-inflammatory drugs. Curr

cr
Med Chem. 2011;18:3494-505

us
12. Bjarnason I, Scarpignato C, Holmgren E, et al. Mechanisms of damage to the

gastrointestinal tract from nonsteroidal anti-inflammatory drugs. Gastroenterology.


an
2018;154:500-514.
M
** A complete and recent review about the different mechanisms of damage

induced by NSAIDs in the whole GI tract.

13. Langenbach R, Morham SG, Tiano HF, et al. Prostaglandin synthase 1 gene
ed

disruption in mice reduced arachidonic acid-induced inflammation and indomethacin-


pt

induced gastric ulceration. Cell. 1995;83:483-492.

14. Sigthorsson G, Simpson RJ, Walley M, et al. COX-1 and 2, intestinal integrity, and
ce

pathogenesis of nonsteroidal anti-inflammatory drug enteropathy in mice.

Gastroenterology. 2002;122:1913-1923.
Ac

15. Lichtenberger LM, Wang ZM, Romero JJ, et al. Nonsteroidal anti-inflammatory drugs

(NSAIDs) associate with zwitterionic phospholipids: insight into the mechanism and

reversal of NSAID-induced gastrointestinal injury. Nat Med. 1995;1:154-158.


16. Varum FJ, Veiga F, Sousa FS, et al. An investigation into the role of mucus thickness

on mucoadhesion in the gastrointestinal tract of pig. Eur J Pharm Sci. 2010;40:335-

341.

17. Basivireddy J, Vasudevan A, Jacob M, et al. Indomethacin-induced mitocondrial

dysfunction and oxidative stress in villus enterocytes. Biochem Pharmacol.

2002;64:339-349.

t
ip
18. Krause MM, Brand MD, Krauss S, et al. Nonsteroidal antiinflammatory drugs and a

selective cyclooxigenase 2 inhibitor uncouple mitocondria in intact cells. Arthritis

cr
Rheum. 2003;48:1438-1444.

us
19. Yeomans ND, Tulassay Z, Juhász L, et al. A comparison of omeprazole with

ranitidine for ulcers associated with nonsteroidal antiinflammatory drugs. Acid


an
Suppression Trial: Ranitidine versus Omeprazole for NSAID-associated Ulcer

Treatment (ASTRONAUT) Study Group. N Engl J Med. 1998;338:719-26.


M

20. Taha AS, Hudson N, Hawkey CJ, Swannell AJ et al. Famotidine for the prevention of
ed

gastric and duodenal ulcers caused by nonsteroidal antiinflammatory drugs. N Engl J

Med. 1996;334:1435-9.
pt

21. Hooi JKY, Lai WY, Ng WK, et al. Global Prevalence of Helicobacter pylori

Infection: Systematic Review and Meta-Analysis. Gastroenterology. 2017;153:420-


ce

429
Ac

22. Datta De D, Roychoudhury S. To be or not to be: The host genetic factor and beyond

in Helicobacter pylori mediated gastro-duodenal diseases. World J Gastroenterol.

2015;21:2883-95.

23. Lanas A, Chan FK. Peptic ulcer disease. Lancet. 2017;390:613-624


24. White JR, Winter JA, Robinson K. Differential inflammatory response to

Helicobacter pylori infection: etiology and clinical outcomes. J Inflamm Res.

2015;8:137-47

25. Konturek JW, Dembinski A, Konturek SJ, et al. Infection of Helicobacter pylori in

gastric adaptation to continued administration of aspirin in humans. Gastroenterology.

1998;114:245-55

t
ip
26. Iijima K, Ara N, Abe Y, et al. Biphasic effects of H. pylori infection on low-dose

aspirin-induced gastropathy depending on the gastric acid secretion level. J

cr
Gastroenterol. 2012;47:1291-7

us
27. Huang JQ, Sridhar S, Hunt RH. Role of Helicobacter pylori infection and non-

steroidal anti-inflammatory drug in peptic-ulcer disease: a meta-analysis. Lancet.


an
2002;359:14-22.

28. Duggan DE, Hooke KF, Noll RM, Kwan CK. Enterohepatic circulation of
M

indomethacin and its role in intestinal irritation. Biochem Pharmacol. 1975;24:1749-


ed

54.

29. Wax J, Clinger WA, Varner P, et al. Relationship of the enterohepatic cycle to
pt

ulcerogenesis in the rat small bowel with flufenamic acid. Gastroenterology.

1970;58:772-80.
ce

30. Melrange R, Gentry C, O'Connell C, et al. Antiinflammatory and gastrointestinal


Ac

effects of nabumetone or its active metabolite, 6-methoxy-6-naphthylacetic acid

(6MNA). Digestive Disease and Sciences. 1992;37:1847-52

31. Semple PF, Russell RI. Role of bile acids in the pathogenesis of aspirin-induced

gastric mucosal hemorrhage in rats. Gastroenterology. 1975;68:67-70

32. Boelsterli UA, Ramirez-Alcantara V. NSAID acyl glucuronides and enteropathy. Curr

Drug Metab. 2011;12:245-52


33. Watanabe T, Higuchi K, Kobata A, et al. Non-steroidal anti.inflammatory drug-

induced small intestinal damage is toll-like receptor 4 dependent. Gut. 2008;57:181-7

34. Walt R, Katschinski B, Lgan R, et al. Rising frequency of ulcer perforation in elderly

people in the United Kingdom. Lancet. 1986;1:489-492

35. Brun J, Jones R. Non-steroidal anti-inflammatory drug-associated dyspepsia: the scale

of problem. Am J Med. 2001;110:12S-13S

t
ip
36. Lanas A, Hunt R. Prevention of anti-inflammatory drug induced gastrointestinal

damage: benefits and risks of therapeutic strategies. Ann Med. 2006;38:415-428

cr
37. Larkai EN, Smith JL, Lidsky MD, Graham DY. Gastroduodenal mucosa and

us
dyspeptic symptoms in arthritic patients during chronic steroidal anti-inflammatory

drug use. Am J Gastroenterol. 1987;82:1153-8


an
38. Armstrong CP, Blower AL. Non-steroidal anti-inflammatory drugs and life

treathening complications of peptic ulceration. Gut. 1987;28:527-32


M

39. Hernandez-Diaz S, Rodriguez LA. Association between nonsteroidal anti-


ed

inflammatory drugs and upper gastrointestinal tract bleeding/perforation: an oveview

of epidemiologic studies published in the 1990s. Arch Intern Med. 2000;160:2093-9


pt

40. Sostres C, Gargallo CJ, Lanas A. Nonsteroidal anti-inflammatory drug and upper and

lower gastrointestinal mucosal damage. Arthritis Research & Therapy. 2013,15(suppl


ce

3):S3
Ac

* In this thorough review upper and lower GI damage caused by NSAIDs is

explained in detail.

41. Melcarne L, Garcia-Iglesias P, Calvet X. Management of NSAID-associated peptic

ulcer disease. Expert Rev Gastroenterol Hepatol. 2016;10:723-33


42. Castellsague J, Riera-Guardia N, Calingaert B, et al. Individual NSAIDs and upper

gastrointestinal complications: a systematic review and meta-analysis of observational

studies (the SOS Project). Drug Saf. 2012;35:1127-1146

43. Nam YH, Bresinger CM, Bilker WB, et al. Nonsteroidal anti-inflammatory drug

choice and adverse outcomes in clopidogrel users: a retrospective cohort study. PLoS

One. 2018;13:e0193800

t
ip
44. Anglin R, Yuan Y, Moayyedi MB, et al. Risk of upper gastrointestinal bleeding with

selective serotonin reuptake inhibitors with or without concurrent nonsteroidal anti-

cr
inflammatory use: a systematic review and meta-analysis. Am J Gastroenterol.

us
2014;109:811-9

45. Sostres C, Carrera-Lasfuentes P, Benito R, et al. Peptic ulcer bleeding risk. The role
an
of Helicobacter pylori infection in NSAID/Low-dose aspirin users. Am J

Gastroenterol. 2015; 110:684-9


M

46. Sostres C, Carrera-Lasfuentes P, Lanas A. NSAID-related upper gastrointestinal


ed

bleeding: types of drug use and patient profiles in real clinical practice. Curr Med Res

Opin. 2017;33:1815-1820
pt

47. Fries JF, Murtagh KN, Bennett M, et al. The rise and decline of nonsteroidal

antiinflamatory drug-associated gastrophaty in rheumatoid arthritis. Arthritis Rheum.


ce

2004;50:2433-2440
Ac

48. Lanas A, García-Rodriguez LA, Polo-Tomás M, et al. The changing face of

hospitalisation due to gastronintestinal bleeding and perforation. Aliment Pharmacol

Ther. 2011;33:585-591

49. Maiden L, Thjodleifsson B, Seigal A, et al. Long-term effects of nonsteroidal anti-

inflammatory drugs and cyclooxygenase-2 selective agents on the small bowel: a


cross-sectional capsule enteroscopy study. Clin Gastroenterol Hepatol.

2007;5(9):1040-5

50. Maiden L. Capsule endoscopic diagnosis of nonsteroidal anti-inflammatory drug-

induced enteropathy. J Gastroenterol. 2009;44 Suppl 19:64-71.

51. Ishihara M, Ohmiya N, Nakamura M, et al. Risk factors of symptomatic NSAID-

induced small intestinal injury and diaphragm disease. Aliment Pharmacol Ther.

t
ip
2014;40(5):538-47

52. Lanas A, Sopeña F. Nonsteroidal anti-inflammatory drugs and lower gastrointestinal

cr
complications. Gastroenterol Clin North Am. 2009;38:333-52

us
53. Lanas A, Panes J, Piqué JM. Clinical implications of COX-1 and/or COX-2 inhibition

on the distal gastrointestinal tract. Curr Pharm Des. 2003;9:2253-2266


an
54. Bjarnason I, Zanelli G, Prouse P, et al. Blood and protein loss via small intestinal

inflammation induced by nonsteroidal antiinflammatory drugs. Lancet. 1987;2:711-


M

714
ed

55. Sigthorsson G, Crane R, Simon T, et al. COX-2 inhibition with rofecoxib does not

increase intestinal permeability in healthy subjects: a double blind crossover study


pt

comparing rofecoxib with placebo and indomethacin. Gut. 2000;47:527-532

56. Kurahara K, Matsumoto T, Iida M, et al. Clinical and endoscopic features of


ce

nonsteroidal anti-inflammatory drug induced colonic ulcerations. Am J Gastroenterol.


Ac

2001;96:473-480

57. Moore RA, Derry S, McQuay HJ. Faecal blood loss with aspirin, nonsteroidal anti-

inflammatory drugs and cyclo-oxygenase-2 selective inhibitors: systematic review of

randomized trials using autologous chromium-labelled erythrocytes. Arthritis Res

Ther. 2008; 10: R7


58. Laine L, Curtis SP, Langman M, et al. Lower gastrointestinal events in a double-blind

trial of the cyclo-oxigenase-2 selective inihibitor etoricoxib and the traditional

nonsteroidal anti-inflammatory drug diclofenac. Gastroenterology. 2008;135:1517-25

59. Nagata N, Niikura R, Aoki T, et al. Lower GI bleeding risk of nonsteroidal anti-

inflammatory drugs and antiplatelet drug use alone and the effect of combined

therapy. Gastrointest Endosc. 2014;80:1124-31

t
ip
60. Lanas A, Carrera-Lasfuentes P, Arguedas Y, et al. Risk of upper and lower

gastrointestinal bleeding in patients taking nonsteroidal anti-inflammatory drugs,

cr
antiplatelet agents, or anticoagulants. Clin Gastroenterol Hepatol. 2015;13: 906–12

us
61. Strate LL, Liu YL, Huang ES, et al. Use of aspirin or nonsteroidal anti-inflammatory

drugs increases risk for diverticulitis and diverticular bleeding. Gastroenterology.


an
2011;140:1427-33

62. Yuhara H, Corley DA, Nakahara F, et al. Aspirin and non-aspirin NSAIDs increase
M

risk of colonic diverticular bleeding: a systematic review and meta-analysis. J


ed

gastroenterol. 2014;49:992-1000

63. Nagata N, Niikura R, Aoki T, et al. Risk factors for adverse in-hospital outcomes in
pt

acute colonic diverticular hemorrhage. World J Gastroenterol. 2015;21:10697-10703

64. Knights KM, Mangoni AA, Miners JO. Defining the COX inhibitor selectivity of
ce

NSAIDs: implications for understanding toxicity. Expert Rev Clin Pharmacol.


Ac

2010;3:769-76

65. Bombardier C, Laine L, Reicin A, et al. Comparison of upper gastrointestinal toxicity

of rofecoxib and naproxen in patients with rheumatoid arthritis. VIGOR Study Group.

N Engl J Med. 2000;343:1520-8


66. Rostom A, Muir K, Dubé C, et al. Gastrointestinal safety of cyclooxigenase-2

inihibitors: a Cochrane Collaboration systematic review. Clin Gastroenterol Hepatol.

2007;5:818-828

67. KLC F, Lanas A, Scheiman J, et al. Celecoxib versus omeprazole and diclofenac in

patients with osteoarthritis and rheumatoid arthritis (CONDOR): a randomised trial.

Lancet. 2010; 376: 173–79

t
ip
68. Lanas A, García-Rodríguez LA, Arroyo MT, et al. Risk of upper gastrointestinal ulcer

bleeding associated with selective cyclo-oxigenase-2 inhibitors, traditional non-

cr
aspirin non-steroidal anti-inflammatory drugs, aspirin and combinationds. Gut.

us
2006;55:1731-1738

69. Schnitzer TJ, Bumester GR, Mysler E, et al. TARGET study group: Comparison of
an
lumiracoxib with naproxen and ibuprofen in the Therapeutic Arthritis Research and

Gastrointestinal Event Trial (TARGET), reduction in ulcer complications: randomised


M

controlled trial. Lancet. 2004;364:665-674


ed

70. Singh G, Fort JG, Goldstein JL, et al. Celecoxib versus naproxen and diclofenac in

osteoarthritis patients: SUCCESS-1 study. Am J Med. 2006;119:255-266


pt

71. Rostom A, Muir K, Dube C, et al. Prevention of NSAID-related upper gastrointestinal

toxicity: a meta-analysis of traitional NSAIDs with gastroprotection and COX-2


ce

inhibitors. Drug Health Care Pat Saf. 2009;1:1-25


Ac

72. Reed GW, Abdallah MS, Shao M, et al. Effect of Aspirin Coadministration on the

Safety of Celecoxib, Naproxen, or Ibuprofen. J Am Coll Cardiol. 2018;71:1741-1751.

73. Ghosh R, Alajbegovic A, Gomes AV. NSAIDs and cardiovascular diseases: role of

reactive oxygen species. Oxid Med Cell Longev. 2015;2015:536962


74. Yeomans ND, Graham DY, Husni ME, et al. Randomised clinical trial:

gastrointestinal events in arthritis patients treated with celecoxib, ibuprofen or

naproxen in the PRECISION trial. Aliment Pharmacol Ther. 2018;47:1453-1463

75. Pepine CJ, Gurbel PA. Cardiovascular safety of NSAIDs: Additional insights after

PRECISION and point of view. Clin Cardiol. 2017;40:1352-1356

76. Solomon DH, Husni ME, Libby PA, et al. The Risk of Major NSAID Toxicity with

t
ip
Celecoxib, Ibuprofen, or Naproxen: A Secondary Analysis of the PRECISION Trial.

Am J Med. 2017;130:1415-1422

cr
2017;38:3282-3292

us
77. Chan FKL, Ching JYL, Tse YK, et al. Gastrointestinal safety of celecoxib versus

naproxen in patients with cardiothrombotic diseases and arthritis after upper


an
gastrointestinal bleeding (CONCERN): an industry-independent, double-blind,

double-dummy, randomised trial. Lancet. 2017;389:2375-2382


M

78. Andreadou I, Iliodromitis EK, Rassaf T, et al. The role of gasotransmitters NO, H2S
ed

and CO in myocardial ischaemia/reperfusion injury and cardioprotection by

preconditioning, postconditioning and remote conditioning. Br J Pharmacol.


pt

2015;172(6):1587-606.

79. Wallace JL, Vaughan D, Dicay M,et al. Hydrogen Sulfide-Releasing Therapeutics:
ce

Translation to the Clinic. Antioxid Redox Signal. 2018; 28(16):1533-1540.


Ac

* A very recent review about advances in H2S-releasing drugs, such as SG1002 for

cardiovascular disorders and ATB-346 for arthritis.

80. Liu Z, Han Y, Li L, et al. The hydrogen sulfide donor, GYY4137, exhibits anti-

atherosclerotic activity in high fat fed apolipoprotein E(-/-) mice. Br J Pharmacol.

2013;169(8):1795-809.
81. Wallace JL, Ianaro A, de Nucci G. Gaseous Mediators in Gastrointestinal Mucosal

Defense and Injury. Dig Dis Sci. 2017; 62(9):2223-2230.

82. Fiorucci S, Santucci L, Gresele P, et al. Gastrointestinal safety of NO-aspirin (NCX-

4016) in healthy human volunteers: a proof of concept endoscopic study.

Gastroenterology. 2003; 124(3):600-7.

83. Hawkey CJ, Jones JI, Atherton CT, et al. Gastrointestinal safety of AZD3582, a

t
ip
cyclooxygenase inhibiting nitric oxide donator: proof of concept study in humans.

Gut. 2003; 52(11):1537-42.

cr
84. Lohmander LS, McKeith D, Svensson O, et al. A randomised, placebo controlled,

us
comparative trial of the gastrointestinal safety and efficacy of AZD3582 versus

naproxen in osteoarthritis. Ann Rheum Dis. 2005; 64(3):449-56.


an
* A double blind phase II trial with 970 patients, the primary end point (6-week

endoscopic gastroduodenal ulcer incidence) was not significantly different between


M

AZD3582 (NO-naproxen) and naproxen.


ed

85. Wallace JL, Wang R. Hydrogen sulfide-based therapeutics: exploiting a unique but

ubiquitous gasotransmitter. Nat Rev Drug Discov. 2015; 14(5):329-45.


pt

** A complete review by Wallace and Wang about cellular and molecular effects

of H2S, his role in disease processes and the H2S-based therapeutics.


ce

86. Fiorucci S, Antonelli E, Distrutti E, et al. Inhibition of hydrogen sulfide generation


Ac

contributes to gastric injury caused by anti-inflammatory nonsteroidal drugs.

Gastroenterology. 2005; 129(4):1210-24.

87. Wallace JL, Caliendo G, Santagada V, et al. Gastrointestinal safety and anti-

inflammatory effects of a hydrogen sulfide-releasing diclofenac derivative in the rat.

Gastroenterology. 2007; 132(1):261-71.


88. Magierowski M, Magierowska K, Surmiak M, et al. The effect of hydrogen sulfide-

releasing naproxen (ATB-346) versus naproxen on formation of stress-induced gastric

lesions, the regulation of systemic inflammation, hypoxia and alterations in gastric

microcirculation. J Physiol Pharmacol. 2017; 68(5):749-756.

89. Wallace JL, Caliendo G, Santagada V, et al. Markedly reduced toxicity of a hydrogen

sulphide-releasing derivative of naproxen (ATB-346). Br J Pharmacol. 2010;

t
ip
159(6):1236-46.

90. Blackler R, Syer S, Bolla M, et al. Gastrointestinal-sparing effects of novel NSAIDs

cr
in rats with compromised mucosal defence. PLoS One. 2012; 7(4):e35196.

us
91. Blackler RW, Motta JP, Manko A, et al. Hydrogen sulphide protects against NSAID-

enteropathy through modulation of bile and the microbiota. Br J Pharmacol. 2015;


an
172(4):992-1004.

92. Flannigan KL, Agbor TA, Blackler RW, et al. Impaired hydrogen sulfide synthesis
M

and IL-10 signaling underlie hyperhomocysteinemia-associated exacerbation of


ed

colitis. Proc Natl Acad Sci USA. 2014; 111(37):13559-64.

93. Paul-Clark M, Elsheikh W, Kirkby N, et al. Profound Chemopreventative Effects of a


pt

Hydrogen Sulfide-Releasing NSAID in the APCMin/+ Mouse Model of Intestinal

Tumorigenesis. PLoS One. 2016; 11(2):e0147289.


ce

94. http://www.antibethera.com/pipeline/h2s-platform/
Ac

95. Motterlini R, Foresti R. Biological signaling by carbon monoxide and carbon

monoxide-releasing molecules. Am J Physiol Cell Physiol. 2017; 312:C302–C313.

96. Magierowska K, Magierowski M, Hubalewska-Mazgaj M, et al. Carbon Monoxide

(CO) Released from Tricarbonyldichlororuthenium (II) Dimer (CORM-2) in

Gastroprotection against Experimental Ethanol-Induced Gastric Damage. PLoS One.

2015; 10(10):e0140493.
97. Magierowski M, Magierowska K, Hubalewska-Mazgaj M, et al. Carbon monoxide

released from its pharmacological donor, tricarbonyldichlororuthenium (II) dimer,

accelerates the healing of pre-existing gastric ulcers. Br J Pharmacol. 2017;

174(20):3654-3668.

98. Lim YJ, Dial EJ, Lichtenberger LM. Advent of novel phosphatidylcholine-associated

nonsteroidal anti-inflammatory drugs with improved gastrointestinal safety. Gut

t
ip
Liver. 2013; 7(1):7-15.

99. Barrios JM, Lichtenberger LM. Role of biliary phosphatidylcholine in bile acid

cr
protection and NSAID injury of the ileal mucosa in rats. Gastroenterology. 2000;

us
118(6):1179-86.

100. Lichtenberger LM, Ulloa C, Romero JJ, et al. Nonsteroidal anti-inflammatory


an
drug and phospholipid prodrugs: combination therapy with antisecretory agents in

rats. Gastroenterology. 1996; 111(4):990-5.


M

101. Lichtenberger L, Romero JJ, Dial EJ. Gastrointestinal safety and therapeutic
ed

efficacy of parenterally administered phosphatidylcholine-associated indomethacin in

rodent model systems. Br J Pharmacol. 2009; 157(2):252-7.


pt

102. Lichtenberger LM, Romero JJ, Dial EJ. Surface phospholipids in gastric injury

and protection when a selective cyclooxygenase-2 inhibitor (Coxib) is used in


ce

combination with aspirin. Br J Pharmacol. 2007; 150(7):913-9.


Ac

103. Lim YJ, Phan TM, Dial EJ, et al. In vitro and in vivo protection against

indomethacin-induced small intestinal injury by proton pump inhibitors, acid pump

antagonists, or indomethacin-phosphatidylcholine. Digestion. 2012; 86(2):171-7.

104. Anand BS, Romero JJ, Sanduja SK, et al. Phospholipid association reduces the

gastric mucosal toxicity of aspirin in human subjects. Am J Gastroenterol. 1999;

94(7):1818-22.
105. Cryer B, Bhatt DL, Lanza FL, et al. Low-dose aspirin-induced ulceration is

attenuated by aspirin-phosphatidylcholine: a randomized clinical trial. Am J

Gastroenterol. 2011; 106(2):272-7.

* A randomized, single blind, active-controlled study with 204 healthy subjects,

gastroduodenal erosions and/or ulcers were observed in 42.2% of the aspirin-treated

group and 22.2% of the PC-aspirin-treated group (p=0.0027)

t
ip
106. Lanza FL, Marathi UK, Anand BS, et al. Clinical trial: comparison of

ibuprofen-phosphatidylcholine and ibuprofen on the gastrointestinal safety and

cr
analgesic efficacy in osteoarthritic patients. Aliment Pharmacol Ther. 2008;

us
28(4):431-42.

107. Badri W, Miladi K, Nazari QA, et al. Encapsulation of NSAIDs for


an
inflammation management: Overview, progress,challenges and prospects. Int J

Pharm. 2016;515(1-2):757-773
M

108. Park SH, Hong H, Han YM, et al. Nonsteroidal anti-inflammatory drugs
ed

(NSAID) sparing effects of glucosamine hydrochloride through N-glycosylation

inhibition; strategy to rescue stomach from NSAID damage. J Physiol Pharmacol.


pt

2013;64(2):157-65

109. Bhatt DL, Scheiman J, Abraham NS, et al. American College of Cardiology
ce

Foundation Task Force on clinical expert consensus documents. ACCF/ACG/AHA


Ac

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. Circulation. 2008; 118(18):1894–

1909.
110. García-Rayado G, Sostres C, Lanas A. Aspirin and omeprazole for secondary

prevention of cardiovascular disease in patients at risk for aspirin-associated gastric

ulcers. Expert Rev Clin Pharmacol. 2017; 10(8):875-888.

111. Wallace JL, Syer S, Denou E, et al. Proton pump inhibitors exacerbate

NSAID-induced small intestinal injury by inducing dysbiosis. Gastroenterology.

2011; 141(4):1314-22, 1322.e1-5.

t
ip
112. Vaezi MF, Yang YX, Howden CW. Complications of Proton Pump Inhibitor

Therapy. Gastroenterology. 2017; 153(1):35-48.

cr
113. LoGuidice A, Wallace BD, Bendel L,et al. Pharmacologic targeting of

us
bacterial β-glucuronidase alleviates nonsteroidal anti-inflammatory drug-induced

enteropathy in mice. J Pharmacol Exp Ther. 2012; 341(2):447-54.


an
114. Saitta KS, Zhang C, Lee KK, et al. Bacterial β-glucuronidase inhibition

protects mice against enteropathy induced by indomethacin, ketoprofen or diclofenac:


M

mode of action and pharmacokinetics. Xenobiotica. 2014; 44(1):28-35.


ed

115. Bjarnason I, Hayllar J, Smethurst P, et al. Metronidazole reduces intestinal

inflammation and blood loss in non steroidal anti-inflammatory drug induced


pt

enteropathy. Gut. 1992; 33(9):1204-8.

116. Scarpignato C, Dolak W, Lanas A, et al. Rifaximin Reduces the Number and
ce

Severity of Intestinal Lesions Associated With Use of Nonsteroidal Anti-


Ac

Inflammatory Drugs in Humans. Gastroenterology. 2017; 152(5):980-982.e3.

** A placebo-controlled study including 60 volunteers receive diclofenac plus

omeprazole, with either rifaximin or placebo. Mucosal breaks (P=0.05) and large

lesions (P<0.001) were detected in greater proportion in the placebo arm than in

the rifaximin group.


117. Montalto M, Gallo A, Curigliano V, et al. Clinical trial: the effect of a

probiotic mixture on non-steroidal anti-inflammatory drug enteropathy - a

randomized, double-blind, cross-over, placebo-controlled study. Aliment Pharmacol

Ther. 2010; 32(2):209-14.

118. Endo H, Higurashi T, Hosono K,et al. Efficacy of Lactobacillus casei

treatment on small bowel injury in chronic low-dose aspirin users: a pilot randomized

t
ip
controlled study. J Gastroenterol. 2011; 46(7):894-905.

119. Malmi H, Kautiainen H, Virta LJ, et al. Incidence and complications of peptic

cr
ulcer disease requiring hospitalisation have markedly decreased in Finland. Aliment

us
Pharmacol Ther. 2014; 39: 496−506.
an
M
ed
pt
ce
Ac
Declaration of conflict of interest:
Guillermo Garcia-Rayado, Mercedes Navarro and Angel Lanas declare not to have conflict of
interests

t
ip
cr
us
an
M
ed
pt
ce
Ac
Legends off Figures:

Figure 1: Adverse
A effe
fects of NSA
AIDs in diffferent system
ms and som
me of suggessted

beneficial effects
e of H2S in these systems. *M
Most of data from anim
mal models or

preclinical studies in humans.


h

t
ip
cr
us
an
M
ed
pt
ce
Ac
Figure 2: Mechanisms
M s of pathogeenesis of GII damage by
y NSAIDs aand strategies to

reduce this injury (greeen boxes). C


COX: Cyclo
ooxygenasee. GI: Gastroointestinal.

H2RAs: Hiistamine H2
2 receptor anntagonists. H2S: Hydro
ogen sulfidee. NO: Nitriic oxide.

PPIs: Protoon pump inh


hibitors.

t
ip
cr
us
an
M
ed
pt
ce
Ac

Das könnte Ihnen auch gefallen