You are on page 1of 10

EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY

2022, VOL. 18, NO. 5, 337–346


https://doi.org/10.1080/17425255.2022.2098107

REVIEW

An update on drug–drug interactions associated with proton pump inhibitors


a,b a a,c
Inès Ben Ghezala , Maxime Luu and Marc Bardou
a
Centre d’Investigations Cliniques INSERM 1432 (CIC1432), Dijon Bourgogne University Hospital, Dijon, France; bOphthalmology Department, Dijon
Bourgogne University Hospital, Dijon, France; cGastroenterology and Liver Department, Dijon Bourgogne University Hospital, Dijon, France

ABSTRACT ARTICLE HISTORY


Introduction: Proton pump inhibitors (PPIs) block the gastric H/K-ATPase, therefore inhibiting acid Received 14 February 2022
gastric secretion, leading to an increased pH (>4). They account for an extremely high number of Accepted 1 July 2022
prescriptions worldwide. Numerous drug–drug interactions have been described with PPIs, but all the
KEYWORDS
described interactions do not have clinical significance.
CYP2C19; peptic ulcer;
Areas covered: This review will discuss the latest updates on drug–drug interactions with PPIs, focusing gastroesophageal reflux;
on the last 10-year publications in the following areas: anti-infective agents, anticancer drugs, anti­ Helicobacter; antifungal;
platelet agents and anticoagulants, and antidiabetics. antibiotics; antiplatelet
Expert opinion: Although pharmacokinetic interactions of PPIs have been described with many drugs, agents; antidiabetics;
their clinical relevance remains controversial. However, given the extremely high number of people oncology drugs
being treated with PPIs, clinicians should remain vigilant for interactions that may be clinically
significant and require dose adjustment or therapeutic monitoring. Interestingly, not all PPIs have the
same pharmacokinetic and pharmacodynamic profile, with some having a strong potential to inhibit
CYP2C19, such as omeprazole, esomeprazole, and lansoprazole, while others, pantoprazole, rabepra­
zole, and dexlansoprazole, are weak CYP2C19 inhibitors. These may be preferred depending on co-
prescribed treatments.
In addition, new formulations have been developed to prevent some of the gastric pH-dependent
drug interactions and should be evaluated in further large-scale prospective comparative studies.

1. Introduction years publications in the following areas: anti-infective agents,


anticancer drugs, antiplatelet agents and anticoagulants, and
Proton pump inhibitors (PPIs) effectively block gastric acid
antidiabetics.
secretion, therefore inhibiting gastric fluid secretion, and par­
ticularly acid secretion in the gastric parietal cells, by irrever­
sibly binding to and inhibiting the hydrogen–potassium 2. Anti-infective drugs
ATPase (H/K-ATPase) pump that resides on the luminal surface
2.1. Antifungals
of the parietal cell membrane. PPIs thus increase pH (>4) [1].
PPIs are weak bases (pKa between 3.8 and 4.9) which accu­ PPIs can reduce the bioavailability of systemic antifungals due
mulate in the acidic space of the secretory canaliculus of to their gastric acid suppressing effect [9]. For example, itra­
parietal cells [2]. Through acid gastric secretion, PPIs are then conazole is a broad-spectrum antifungal treatment with pH-
converted from prodrugs to the active sulfenamide or sulfenic dependent solubility: its dissolution and absorption requires
acid secretion. PPIs are indicated in multiple diseases, notably an acidic gastric pH [10]. Omeprazole alters the dissolution
peptic ulcers and particularly bleeding ulcers, gastroesopha­ and the absorption of itraconazole (Sporanox®, Janssen
geal reflux disease, Zollinger–Ellisvon syndrome and Pharmaceuticals, Inc.), reducing its total and peak plasma
Helicobacter pylori eradication therapy [3,4]. Numerous drug– exposure [11,12]. It is therefore recommended to administer
drug interactions have been described with PPIs [5,6]. By itraconazole at least 2 h before or 2 h after any PPI adminis­
increasing gastric pH, PPIs use may modify the absorption tration [13]. Super bioavailability (SUBA ®) itraconazole has
and bioavailability of other drugs. They may also modify been developed to enhance its bioavailability thanks to
their distribution and metabolism, especially because of meta­ a polymeric matrix. Coadministration of omeprazole 40 mg
bolic enzyme competition with cytochromes, mostly cyto­ induced a 22% increase in the total plasma exposure (area
chrome P450 2C19 (CYP2C19) and also but to a lesser extent, under the concentration–time curve) and a 31% increase in
cytochrome P450 3A4 (CYP3A4) [7,8]. the peak plasma exposure of SUBA-itraconazole, which may
All described drug–drug interactions do not have a clinical be confer a therapeutic benefit [14]. Controversial findings
significance. We aimed to highlight the latest updates on were reported in a retrospective cohort study, which sug­
drug–drug interactions with PPIs, focusing on the last 10 gested lower serum trough concentrations in patients

CONTACT Marc Bardou marc.bardou@u-bourgogne.fr Clinical Investigation Center, Plurithematic Unit, Dijon Bourgogne University, Hospital 14, Rue
Gaffarel, BP77908, Dijon Cedex 21079, France
© 2022 Informa UK Limited, trading as Taylor & Francis Group
338 I. BEN GHEZALA ET AL.

pantoprazole or ilaprazole, in countries where it is already


Article highlights accepted, should be prescribed preferentially in combination
● Numerous drug–drug interactions have been reported with PPIs, the
with voriconazole to avoid overdose, but particular attention
most frequent mechanism being the increase in gastric pH, which should still be paid to the risk of subtherapeutic voriconazole
alters the absorption and bioavailability of numerous treatments with concentrations, and therapeutic drug monitoring is recom­
pH-dependent solubility.
● Some formulations have been developed to prevent these gastric pH-
mended [22,25].
dependent drug–drug interactions. Their efficacy should be further No impact of PPI co-prescription on fluconazole bioavail­
demonstrated large-scale comparative prospective studies. ability has been described. However, Lu et al. [26] have
● Another frequent mechanism is the competitive inhibition of cyto­
chromes P450, notably CYP2C19 and CYP3A4
shown synergistic action of PPIs and fluconazole: PPIs
● Some PPIs, omeprazole, esomeprazole, and lansoprazole, have high could significantly decrease fluconazole minimal inhibitory
CYP2C19 inhibitory potential. The others, pantoprazole, rabeprazole, concentrations in vitro, leading to an increased sensitivity
and dexlansoprazole, have a low CYP2C19 inhibitory potential, and
may be preferred according to the co-prescribed treatments.
of resistant C. albicans to fluconazole. In vivo, PPIs plus
● All described drug–drug interactions do not have significant clinical fluconazole prolonged the survival rate of infected Galleria
impact, but clinicians should be careful with the potentially clinically mellonella larvae by two-fold compared with that for the
significant ones. They must also understand that because of the
retrospective nature of most of the studies assessing clinical rele­
fluconazole monotherapy group [26]. This synergistic action
vance, the level of evidence is quite low. can be explained by the PPI-induced suppression of efflux
● According to the co-prescribed drugs, dose adjustment or therapeutic pump activity, which is one of the resistance mechanisms
drug monitoring may be required.
of C. albicans [26].

2.2. Antibiotics
receiving PPIs (95% CI -0.32 to -0.05, p = 0.007) [15].
Nevertheless, in several studies, SUBA-itraconazole seems to PPIs are frequently co-administered with antibiotics, particularly
be overall an efficient formulation to reach therapeutic itraco­ for the treatment of Helicobacter pylori infection. Macrolides,
nazole concentrations, in adults and in children as well, and its such as clarithromycin, are potent inhibitors of CYP3A4 [27].
prescription should be considered, especially with coprescrip­ Concomitant use of omeprazole and clarithromycin can increase
tion of PPIs. Therapeutic drug monitoring should still be per­ both clarithromycin and omeprazole plasmatic concentrations
formed [15,16]. [28]. Controversial interaction between PPI and macrolide have
Voriconazole is another broad-spectrum antifungal agent been published. For example, whereas Saito and colleagues [29]
with significant pharmacokinetic interpatient variability, have reported that the bioavailability of lansoprazole might, to
notably because of its hepatic metabolism by CYP2C19, some extent, be increased through inhibition of P-glycoprotein
which converts it into an inactive metabolite [17]. CYP2C19 during clarithromycin treatment, whereas Cao and colleagues
has significant genetic polymorphism and can be inhibited [30] found the mean Cmax was decreased by 24.9% when ila­
by PPIs [18]. Lansoprazole and omeprazole interact with prazole was given in combination compared to its administration
voriconazole via CYP2C19 and CYP3A4 to increase voricona­ as a single agent. However, this interaction, between ilaprazole
zole plasma concentrations [19,20]. In the Blanco Dorado’s and clarithromycin and amoxicillin, was not any longer observed
study [20], no significant differences were found in plasma in a more recent randomized crossover study investigating the
voriconazole concentration between patients without PPIs coadministration of ilaprazole 10 mg once or twice daily, clari­
and those receiving omeprazole, pantoprazole, or esomepra­ thromycin and amoxicillin in 32 volunteers [31]. In practice, the
zole. Nevertheless, voriconazole plasma concentration was combination of PPI and clarithromycin has long be in the back­
lower in patients treated with pantoprazole than in those bone therapy for H. pylori eradication without any particular
treated with omeprazole (1.44 ± 1.22 μg/mL vs usage precaution [32]. It has been suggested that rabeprazole
2.67 ± 1.88 μg/mL, p = 0.013), and 44% of the patients should be preferred to omeprazole, in combination with amox­
treated with pantoprazole, vs 22% in those receiving ome­ icillin and levofloxacin, as Helicobacter pylori eradication rate
prazole, had a subtherapeutic voriconazole trough concen­ could be higher (85% vs 76.3%, p = 0.16) especially in CYP2C19
tration [20]. These results could be explained by the greater extensive metabolizers (84.6% vs 60.7%, p = 0.03) [33].
cytochrome P450 inhibition capacity of omeprazole com­ PPI-induced pH increase can not only directly affect absorp­
pared to pantoprazole [21]. In an in vitro study in human tion and bioavailability but also change the performance of
liver microsomes, all tested PPIs (omeprazole, lansoprazole, pH-dependent coating strategies. A common strategy to
pantoprazole, esomeprazole, and ilaprazole) showed inhibi­ achieve specificity of pH-dependent drug delivery systems is
tory effects on the voriconazole metabolism [22]. However, coating SDFs with polymers with a pH-dependent solubility.
only lansoprazole, omeprazole, and esomeprazole signifi­ Soares et al. [34], showed that the PPI-induced pH increase
cantly increased voriconazole plasma concentrations in vivo interferes with the release processes of tablets coated with
[22]. Similar results were found in patients with malignant E100 and leads to an increased lag time, a delayed disintegra­
hematological diseases [23]. Coadministration of voricona­ tion process, and a lower area under curve (AUC). In more than
zole with omeprazole has also been reported to lead to a half of the volunteers, metronidazole was not detected in
a twofold increase in maximal plasmatic concentration of plasma in the 5 h after administration [34]. More generally, pH-
omeprazole, and it is therefore recommended to reduce dependent coating strategies should be avoided with coadmi­
omeprazole dose by one-half [24]. It may be suggested that nistration of PPIs.
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 339

2.3. Antivirals the impact of concomitant PPI use on efficacy and pharmaco­
kinetics of glecaprevir/pibrentasvir in 263 patients taking PPIs
Doravirine is a novel non-nucleoside reverse transcriptase
[42]. Rates of SVR12 were 97.0% among patients who used
inhibitor for the treatment of patients with human immuno­
acid-reducing agents and 97.5% among those not using acid-
deficiency virus infection, on the market since 2018 [35,36]. In
reducing agents (P = 0.6), and was not different in those
a three-period open-label trial conducted in healthy volun­
taking low dose (97.4%) vs high dose (96.3%, daily dose
teers, coadministration of pantoprazole lead to a 12%
greater than 20 mg omeprazole or equivalent) of PPIs [42].
decrease in doravirine maximum plasma concentration,
Of note, high-dose PPI use significantly decreased glecaprevir
which was not clinically meaningful [37]. These results support
bioavailability (P < 0.001) with a mean 41% decrease in AUC
the use of acid-reducing agents with doravirine [37].
[42]. No dose adjustment is recommended when PPIs are co-
Ritonavir is an HIV protease inhibitor often used in combi­
prescribed with glecaprevir/pribrentasvir.
nation with other protease inhibitors, marketed since 1999. It
is a lipophilic base (pKa 2.6) with pH-dependent solubility,
formulated as a solid dispersion [7]. Van den Abeele et al. 2.4. Antiparasitics
[38] showed in a crossover study that pretreatment with PPIs
Hydroxychloroquine is used in the treatment of malaria and
lead to a drop in ritonavir gastric solubility, with a tenfold
inflammatory rheumatic diseases. It was also considered
lower median drug solubility and persistence of solid drug
a potentially effective medication for coronavirus disease
material in the stomach. However, duodenal concentrations
(COVID-19) at the beginning of the pandemic in 2020.
remained unaffected. In another crossover study, de Waal
Hydroxychloroquine is a weak base with high solubility in an
et al. [7] showed that, in healthy volunteers, a three-day pre-
acidic environment. Significant inter-individual variability in
treatment with the PPI esomeprazole reduced ritonavir plasma
relative bioavailability has been reported [43]. A randomized,
concentrations by 40%. The gastric residual volume and gas­
parallel drug–drug interaction trial in healthy volunteers found
tric fluid volume decreased by 41% and 44%, respectively,
that hydroxychloroquine AUC0-72h and Cmax did not differ
while the duodenal fluid volume was reduced by 33%, and
between groups without and with pantoprazole (arithmetic
authors suggest it may limit the amount of ritonavir that can
mean; AUC0-72h, 7649 ng/ml • h, and 8429 ng/ml • h, P = .50;
be absorbed [7]. Further studies are needed to investigate this
Cmax, 448 and 451.5 ng/mL, P = .96, respectively) [44]. These
potential interaction and its clinical implications.
results are consistent with those of a randomized double-blind
The treatment of hepatitis C has been dramatically chan­
placebo-controlled trial on patients with systemic lupus trea­
ged by the arrival in 2016 of new treatments combining
ted with hydroxychloroquine, which described that PPI use
molecules that act directly on viral replication. These include
had no significant impact on blood hydroxychloroquine con­
elbasvir/grazoprevir, a combined therapy recently approved to
centrations [45]. Therefore, PPIs and hydroxychloroquine can
treat patients infected with genotype 1 or 4 viruses.
be co-prescribed if needed, without any dose adjustment.
Grazoprevir is a potent once-daily nonstructural protein 3/4A
protease inhibitor, and elbasvir a potent once-daily nonstruc­
tural protein 5A protein inhibitor. Elbasvir is a basic com­ 3. Antiplatelet agents and anticoagulants
pound, and increasing gastric pH may decrease its solubility.
3.1. Antiplatelet agents
Some studies showed a slightly increased plasmatic concen­
tration of elbasvir and grazoprevir when co-administered with Clopidogrel is an antiplatelet agent. CYP2C19 has to convert
PPIs [39]. However, a pooled analysis of six phase 3 studies clopidogrel to an active metabolite for clopidogrel to be
reported that, in 162 patients who were co-prescribed PPIs effective [46]. Some studies have shown that PPIs could
and elbasvir/grazoprevir 12-week sustained viral response reduce the platelet inhibitory effects of clopidogrel, tested
(SVR12) was 96%, not statistically different from the 97% in by vasodilator-stimulated phosphoprotein phosphorylation,
the 1160 patients not self-reporting PPI use [40]. Area under through a competitive effect on CYP2C19 [47,48]. Hence,
the plasma concentration–time curve within 24 h and max­ this reduced anti-aggregation effect could increase the risk
imum plasma concentrations for elbasvir were similar between of cardiovascular ischemic events. Ho et al. [49] reported, in
patients with and without reported PPI use [40]. Elbasvir/gra­ a retrospective cohort study on patients who had an acute
zoprevir summary of pharmacological characteristics (SPC) coronary syndrome (ACS), that use of clopidogrel plus PPI
states that PPI co-prescription does not require any dose was associated with a 25% increased risk of death or rehos­
adjustment. pitalization for ACS compared with use of clopidogrel with­
Glecaprevir is a potent pangenotypic nonstructural protein out PPI (adjusted odds ratio [AOR], 1.25; 95% confidence
3/4A protease inhibitor and pibrentasvir a potent pangenoty­ interval [CI], 1.11–1.41). Same findings were reported in
pic NS5A inhibitor. Glecaprevir/pibrentasvir has been a multi-ethnic Asian population study where clopidogrel
approved for the treatment of chronic genotypes 1–6 HCV with omeprazole was found to be associated with an
infections. Omeprazole has been shown to reduce the max­ increased risk of MI (16% vs 3.8%; AHR 2.03: 95%CI 1.70–
imum plasma concentration and the AUC of glecaprevir from 2.44), but not mortality or stroke [50]. The inconsistency of
22% to 64% [41]. An integrated analysis of nine phase 2 and 3 the increase in risk (from 25% to 200%) is noteworthy, as it
studies, including 2369 patients infected with HCV genotypes highlights the lack of robustness of these pharmaco-
1–6 and compensated liver disease treated with an all-oral epidemiological findings. However, clopidogrel SPC states
regimen of glecaprevir/pibrentasvir for 8–16 weeks, evaluated that administration of omeprazole 80 mg once daily, either
340 I. BEN GHEZALA ET AL.

at the same time as clopidogrel or 12 h apart, decreased the 3.2. Anticoagulants


exposure to the active metabolite by 45% (loading dose) and
PPIs are often co-prescribed with anticoagulants to reduce the
40% (at the maintenance dose). Inhibition of platelet aggre­
risk of gastrointestinal (GI) bleeding [61]. Warfarin is commonly
gation was also reduced, by 39% (loading dose) and 21%
used but requires a careful monitoring of the International
(maintenance dose). A similar interaction is expected with
Normalized Ratio [62], as it has a narrow therapeutic index
esomeprazole and, as a precaution, the association of ome­
and numerous potential drug-drug interactions, especially via
prazole or esomeprazole with clopidogrel is not advised
cytochromes P450. Coprescription of warfarin with PPIs has
[51,52].
been shown to be very frequent, especially in the elderly, and
Controversy exists regarding this reported interaction,
an Australian study reported a 43% rate of coprescription of
which has mainly been described in retrospective studies,
PPI in patients under warfarin for atrial fibrillation [63].
whereas the results from randomized controlled trials evaluat­
A recently published meta-analysis of 72 studies reporting
ing omeprazole compared with placebo showed no difference
on 3,735,775 patients including 11 randomized clinical trials
in ischemic outcomes, despite a reduction in upper gastroin­
and 61 observational studies found a protective effect of PPIs
testinal bleeding with omeprazole [53]. This was later con­
against warfarin-related GI bleeding (OR = 0.69; 95% CI 0.64–
firmed in two meta-analyses of randomized controlled trials
0.73), without significant effect on thromboembolic events or
or propensity scores matched studies [54,55].
mortality [64]. Controversy exists regarding the possible clini­
In a retrospective study on Asian patients receiving clopidogrel,
cally significant interaction between warfarin and PPIs.
platelet inhibition levels did not differ between patients taking or
A retrospective Australian study on 4494 patients receiving
not PPIs, except for dexlansoprazole which induced a decreased
warfarin reported that patients taking warfarin plus PPI had
level of platelet inhibition (25.7% ± 24.3% vs 14.0% ± 21.6%,
a lower mean percentage time in therapeutic range (TTR) than
P = .0297) [56]. In a retrospective study of more than 300,000
patients without PPIs (78.5 ± 9.7% vs 81.7 ± 10.2%, respec­
concomitant PPIs and clopidogrel users, esomeprazole, lansopra­
tively, p < 0.0001) [65]. Patients with PPIs also had a higher
zole, omeprazole, and rabeprazole were not associated with an
incidence of minor bleeds vs patients without PPIs (32.4% vs
increased risk of stroke compared to pantoprazole, which, not
26.1%, respectively, P = 0.0487), but not for major bleeds,
being a strong inhibitor of CYP2C19, is not considered for drug–
questioning the clinical relevance of the statistically significant
drug interactions with clopidogrel [57]. A meta-analysis of 28
difference in TTR [65]. In patients under warfarin, the PPI
studies focusing on major adverse cardiovascular endpoints
prescription should be done after cautiously evaluating the
(MACE), myocardial infarction (MI), cardiovascular death, and gas­
risk–benefit balance and should lead to a careful and regular
trointestinal bleeding and totaling 131,412 patients, found that
INR monitoring.
the pooled HR of adjusted events for MACEs showed that the
Drug–drug interactions between PPIs and factor Xa inhibitors
increased risk of MACEs was similar for 4 classes of PPIs but not for
have also been described. Co-treatment with PPIs was associated
rabeprazole (HR: 1.32; 95% CI 0.69–2.53, P = .40). This was consis­
with a very significant decrease in dabigatran trough and peak
tent with the findings by Shi et al. [58], who reported a twofold
levels (through: 83 ± 42.3 vs 55.5 ± 24.6 ng/mL; and peak:
increased risk of stroke during the 2-year follow-up (HR 2.1,
184.1 ± 107.7 vs 124 ± 59.2 ng/mL, without and with PPI, respec­
p < 0.001) and a 1.6-fold increased risk of myocardial infarction
tively; P < 0.003), probably because of a decrease of its solubility
after 6 months (HR 1.6, p = 0.01). But the same group, working on
due to raised gastric pH [66,67]. A study assessing the bioequiva­
the same database, the China Acute Myocardial Infarction (CAMI)
lence of a newly developed dabigatran tablet showed that at high
registry, found that PPIs in combination with clopidogrel was
gastric pH, a mean gastric pH of 5.3, bioavailability of the tablet was
associated with decreased risk for MACCE in AMI patients [59].
reduced by approximately 70%, which may lead to reduced effi­
In a retrospective cohort study, Muthiah et al. [50] found
cacy [68]. This effect being reversible 2 weeks after PPI withdrawal
that the concomitant use of clopidogrel with omeprazole was
[69]. Further studies investigating clinical potential consequences
associated with an increased risk of myocardial infarction,
of this interaction are required. Of interest, patients treated with
especially in Chinese and Malay patients, but not mortality
anticoagulants are at increased risk of gastrointestinal bleeding,
or stroke. A retrospective cohort study on the Korean
which can be severe and life-threatening. Patients benefitting from
National Health Insurance Database found an increased risk
a coprescription of anticoagulants and PPIs have a 34–47% lower
of major thrombotic events in patients under clopidogrel and
incidence of hospitalization for upper gastrointestinal tract bleed­
PPIs compared with clopidogrel only (HR 1.27, 95% CI 1.12–
ing, so this coprescription should not be discouraged [70,71].
1.45) [60]. The study also reported that the risk was higher
with PPIs with high CYP2C19 inhibitory potential (omeprazole,
esomeprazole, and lansoprazole, HR 1.28, 95% CI 1.02–1.61)
4. Antidiabetics
than PPI of low CYP2C19 inhibitory potential (pantoprazole,
rabeprazole, and dexlansoprazole) [60]. Metformin is a first-line treatment for patients with type 2
These recent data tend to question the existence of diabetes. In vitro studies have shown that PPIs were potent
a clinically significant interaction between PPI and clopidogrel. organic cation transporters inhibitors, therefore limiting active
As a precaution PPIs should still be used cautiously in associa­ transport of metformin into its target cells [72]. A randomized
tion with clopidogrel. Low CYP2C19-inhibitory potential PPIs crossover study found that PPIs induced a slight increase in
such as pantoprazole or rabeprazole may be preferred in metformin concentration (+15% and +22% with pantoprazole
association with clopidogrel. and rabeprazole, respectively) [73]. A retrospective cohort
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 341

study, using the Health Improvement Network database in the impairing their efficacy, and consequently the patient’s
United Kingdom, reported a minimally, and not clinically rele­ prognosis.
vant, better glycemic control exhibited by a − 0.06% decrease
in HbA1C 3–9 months after the start of metformin in those 5.1.1. EGFR inhibitors
also taking PPIs (−1.69% vs −1.63%, respectively, p = 0.01) [74]. Erlotinib is a TKI targeting specifically the epidermal growth
A low-quality (small sample-size, bias not discussed) case– factor receptor (EGFR). The interaction between PPI and erlo­
control study found that the prescription of PPIs in addition tinib in non-small cell lung cancer (NSCLC) has been demon­
to metformin was associated with better glycemic control as strated in several studies [82,84,85]. PPI intake significantly
expressed by mean HbA1C, which was 8.15% for patients lowers the plasma concentration-to-dose ratio of erlotinib by
treated with metformin only and 6.01% with patients treated 23% (in µg·mL·mg·kg: 0.39; 0.08–0.76 vs 0.51; 0.28–1.28 in PPI
with metformin and PPIs (p = 0.002) [75]. A meta-analysis of users and non-users respectively, p < 0.05], and its oral clear­
seven studies (n = 342) for glycemic control and 5 studies ance by 28% (in L/h: 5.55; 3.36–14.52, vs 3.95; 2.01–10.44 in PPI
(n = 244 439) for risk of incident diabetes, found that com­ users and non-users, respectively, p < 0.05) [85].
pared with standard therapy, add-on PPI was associated with Same drug–drug interaction has been described gefitinib,
a significant decrease in HbA1c (−0.36%; −0.68 to −0.05; another EGFR-TKI, with a significant decrease of AUC0-24 (med­
P = 0.025) and Fasting Blood glucose (−10.0 mg/dL; −19.4 to ian AUC0-24 values: 8542 vs 13,103 ng.h/mL, in PPI users and
−0.6; P = 0.037). PPI use did not reduce the risk of incident non-users respectively; P = 0.005) [86].
diabetes [76]. This could be explained by a PPI-induced If publications are quite consistent on the PK consequences of
increase in gastrin levels [75,76]. Further large-scale prospec­ PPI-TKI drug–drug interaction, that is much less the case for the
tive studies are required to investigate this hypothesis. As of clinical relevance of this interaction. A meta-analysis published in
now, no clear clinically significant interaction between PPIs 2021, including 45,626 patients from 7 RCTs and 18 observational
and metformin has been demonstrated, and PPIs and metfor­ studies, with esophageal/gastric, colorectal, pancreatic, lung,
min may be co-prescribed if needed. breast, prostate, kidney, and other cancers, and found
An oral glucagon-like peptide-1 analog, semaglutide a significantly worse prognosis (worse PFS, HR 1.64; 1.14–2.37,
(Rybelsus®, Novo Nordisk A/S), has been approved by health and OS, HR 1.13; 1.05–1.21) only for patients with lung cancer
authorities in 2019–2020 [77,78]. Oral semaglutide has been [87]. No association was found with any of the other cancer
formulated with an absorption enhancer, sodium assessed [87]. Data from a cohort of 12,538 patients with cancer,
N-(8-[2-hydroxylbenzoyl] amino) caprylate (SNAC), which from the Medicare database, showed that prevalence of TKI-PPI
increases gastric pH. However, in a randomized open-label use was 22.7% and that TKI-PPI use decreased survival (HR = 1.16;
trial, the coprescription of omeprazole with oral semaglutide 95% CI = 1.05, 1.28) at 90 days and 1 year (median survival time
was associated with a non-statistically significant, and not 260 days vs 306 days, HR = 1.10; 95% CI = 1.04,1.18) [88]. In a recent
clinically relevant, increase in semaglutide exposure meta-analysis on 1,114 patients from 14 studies treated with EGFR-
(AUC0-24h,semaglutide,Day10, estimated treatment ratio 1.13; TKIs for a non-small cell lung cancer, Sim et al. [89] showed that TKI
0.88–1.45, and Cmax,semaglutide,Day10, estimated treatment ratio users alone had more favorable survival outcomes than TKI-PPIs
1.16; 90% CI 0.90, 1.49) [79]. Semaglutide and PPIs can be co- users, with a better overall survival (OS) rate at 1.98 (95% CI: 1.33–
prescribed without any dose adjustment. 2.94, P = 0.0007), and progression-free survival (PFS) rate
(HR = 3.39, 95% CI: 2.18–5.26, P < 0.00001). Nevertheless, it remains
unclear whether this negative impact on survival is exclusively due
5. Oncology drugs to the drug–drug interactions or if the pro-carcinogenic effect of
long-term PPI use can play a role.
PPIs are widely used in cancer patients to manage anticancer
drug-related gastrointestinal symptoms. At least a fifth of
patients treated for a cancer are taking PPIs at normal dose, 5.1.2. VEGF inhibitors
but this estimation is likely to be largely underestimated in The interaction between molecules targeting the vascular
countries where the over-the-counter use may be substantial endothelial growth factor (VEGF) and PPI has now been stu­
[80]. Additionally, the recent development of oral cancer died now for a decade, but findings are still contradictory. In
therapies has increased their potential indirect drug interac­ a pooled analysis of 10 phase II and III trials, totaling 2,188
tion with PPIs, mainly due to the absorption alteration. patients, treated with sunitinib (n = 952), axitinib (n = 626) or
sorafenib (n = 610) for metastatic renal cell carcinoma, the PPI
users (n = 120) showed similar OS, PFS and objective response
rates than non-PPI users [90]. But the number of patients
5.1. Tyrosine kinase inhibitors (TKI)
receiving PPI may be too small to find a statistical difference.
There are numerous publications on the interactions between On the contrary, in a retrospective review on 231 patients
TKI and PPI, and we do not present here a systematic review of treated by sunitinib for metastatic renal cell carcinoma (45
all available evidences. Interactions between PPI and Tyrosine with concomitant PPI use), median PFS and OS were shorter
Kinase Inhibitors are mainly of pharmacokinetic nature. TKIs in patients who received continuous acid suppression (mPFS
are weak bases that require a low pH in order to be fully 18.9 weeks, 11.0–23.7 vs 23.6 weeks, 19.0–31.9 weeks and
solubilized for an optimal absorption [81–83]. PPIs by their mOS 40.9 weeks, 26.1–74.4 vs 62.4 weeks, 42.0–82.7 weeks
strong anti-acid effect decrease absorption of TKIs, potentially for PPI users and non-users respectively) [91]. This
342 I. BEN GHEZALA ET AL.

Pazopanib targets VEGFR1, VEGFR12, VEGFR3, PDGFRα and prescribers should remain cautious and be aware that these
β, and c-KIT. Two trials conducted in patients with soft-tissue recommendations are mainly based on physiologically based
sarcoma – the single-arm, phase 2, EORTC 62043 trial and the pharmacokinetic (PBPK) models. Clinical data are currently
placebo-controlled, phase 3, EORTC 62072 (PALETTE) trial – inexistent or sparse for the newest TKIs.
were pooled in a post hoc analysis to evaluate the impact of
concomitant acid-suppressive (AS) treatment, mainly PPI (93/
5.2. Immunotherapies
117; 79.5%), on OS and PFS [92]. At a median follow-up of
27.6 months (IQR 22.9–35.4), patient with concomitant AS Immune checkpoint inhibitors (ICPI) got a renewed interest in
treatment had a significant worse median PFS of 2.8 months the last years, but few data exist yet on their interaction with
versus 4.6 months, respectively (HR 1.49; 1.11–1.99; p = 0.008). PPI in case of co-medication. The main hypothesis is an indir­
Median OS was also shorter in patients receiving AS, ect interaction throughout the gut microbiome modification.
8.0 months versus 12.6 months (1.81, 1.31–2.49; p < 0.001) Several preclinical works have shown an impaired immune
[93]. Unfortunately, no PPI-only subgroup analysis was per­ response when the microbiome is altered [100,101]. PPIs are
formed to corroborate these results. Nonetheless, in a recent known to alter the microbiome and are regularly accused of
monocentric retrospective study, no negative impact of PPI on increasing the risk of Clostrium difficile infection, but the level
clinical outcome was observed [94]. of evidence is very low [102,103]. In a retrospective study on
It is worth noticing that even with a 1-h delay interval the Japanese adverse Drug Event Report (JADER) database,
between pazopanib and PPI intake, pazopanib trough concen­ concomitant use of PPI with ICPI was strongly associated with
tration (Cmin) remains significantly lowered, whether pazopa­ an increased risk of nephritis, but only for male patients
nib is taken fasted at 800 mg or with food at 600 mg. This treated with ipilimumab (OR, 3.88; 1.63–9.04, p = 0.001)
reduction of pazopanib exposure seems to be stronger with [104]. However, considering the overall very low number of
omeprazole (−20% fasted, p = 0.038; −31.6% with food, cases of nephritis reported with PPI use in this study, and the
p = 0.003), than with pantoprazole (−5.6% fasted; p = 0.8, non-capture of other possible confounding factors (antibio­
−11.1% with food, p = 0.5) [95]. tics), this observed association needs to be further confirmed.
Once again, data come mostly from retrospective or post In a pooled post-hoc analysis of the phase II POPLAR and
hoc analysis and the strength of the evidences is very low. phase III OAK trials, patients with NSCLC were randomized to
receive atezolizumab (n = 757) or docetaxel (n = 755) [105].
Within the atezolizumab population, PFS and OS were signifi­
5.1.3. Other TKIs cantly shorter in patients who received PPI (1.9 vs 2.8 months,
Bcr-Abl (Bcr [Breakpoint Cluster Region]-Abl) TKI is indicated HR 1.30, 95% CI 1.10–1.53, P = 0.001, and 9.6 vs 14.5 months,
for chronic myeloid leukemia in chronic phase (CP-CML) and HR 1.45, 95% CI 1.20–1.75, P = 0.0001 respectively). No asso­
does not seem to be affected by the concomitant use of PPIs. ciation between antibiotics and PPI was observed, which
In a retrospective analysis on 748 patients with CP-CML (with strongly support independence of the PPI effect. In brief, the
256 imatinib-resistant or – intolerant patients), the molecular link between PPI and immunotherapy is still uncertain, and in
response at 12 months of nilotinib, was similar AS users com­ the hypothesis of a gut microbiome involvement, future stu­
pared to those without AS treatment (49.6% vs 41.0%, dies must take in account antibiotics co-medication in their
p = 0.13) [96]. Interestingly, in a more recent study on dasati­ analyses. Table 1 is a brief summary of the main drug-to-drug
nib, another Bcr-Abl TKI, in 73 patients with CP-CML, AS users interactions discussed above with their clinical relevance and
had a better compliance to treatment, with no interruption at potential preventive measures.
18 months, while nearly half of non-AS users (18/25, 43.9%)
had to stop because of adverse events [97]. Here also no
specific PPI user group analysis was done.
Apart from the molecules detailed upper, there is no signal 6. Conclusion
of concern regarding PPI interaction with the other TKI. Many drug–drug interactions have been described with PPIs,
Regorafenib, a multikinase inhibitor (VEGFR, KIT, B-Raf, involving different mechanisms, the first one being the
PDGFR, and FGRF) proved in a randomized crossover pharma­ increase in gastric pH, i.e. The principal pharmacodynamic
cokinetic trial to be unaffected by concomitant or subsequent action of PPIs. The absorption and bioavailability of treatments
(3 h later) omeprazole intake [93]. Co-administration of PPI with ph-dependent solubility are often decreased by PPIs
with lenvatinib, sorafenib, vandetanib for example is consid­ concomitant PPI usefor example, with itraconazole. When
ered safe by the FDA and EMA [98]. PPIs are co-administered, clinicians should pay particular
A retrospective review of a prospectively collected electro­ attention to any treatments involving ph-dependent coating
nic database identified 99 patients treated with cabozantinib, strategies, as described for example with metronidazole-
including 43 patients being PPI users, found that, after coated magnetic tablets. Second, many interactions involve
a median follow-up of 30.3 months, PPI users showed similar cytochromes P450, notably CYP2C19 and CYP3A4, via compe­
progression-free survival and overall survival outcomes com­ titive inhibition, as for example with clopidogrel.
pared with PPI nonusers [99]. In the independent pharmaco­ Finally, drug interactions with ppis may involve some less
kinetic cohort, of whom 21 received PPI concomitantly, known mechanismsfor example, via inhibition of organic
Ctrough was similar between the two groups of patients cation transporters, which reduces the uptake of metformin
with mRCC who received cabozantinib [99]. Nevertheless, by its target cells.
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 343

Table 1. drug-drug interactions associated with proton pump inhibitors.


Class of
medications Effect of the drug-to-drug interaction Clinical consequence. Clinical relevance/action
Anti-infectives, antifungeal
-Itraconazole Decreased total and peak plasma exposure Decreased efficacy Uncertain/no simultaneous intake
Increased plasma concentration. Increased antifungeal activity Uncertain/Reduce PPI dose
-Voriconazole No impact /suppression of efflux pump Uncertain/none
-Fluconasole activity
Antibiotics
-Macrolides Increased blood levels Synergistic effect Uncertain/none
Antivirals
-Doravirine 12% decreased blood levels Reduced efficacy No/None
-Ritonavir 40% decreased blood levels Reduced efficacy Possible/ avoid co-prescription
-elbasvir/ Increased blood level None No/No dose adjustment
grazoprevir 22-64% decreased blood level None No/No dose adjustment
-Glecaprevir
Antiplatelet agents
-clopidogrel Reduced conversion into the active Reduced antiplatelet activity/increased Uncertain/prefer other antiplatelet agents or use
metabolite risk for MI rabeprazole
Anticoagulants
-warfarin Reduced proportion of time in therapeutic Controversial effect on bleeding Uncertain/INR monitoring at PPI initiation or
range No clear signal cessation
-Factors Xa decrease in trough and peak levels No/None
inhibitors
Antidiabetics
-Metformin Reduced transport to target cells Better glycaemic control Uncertain/No adaptation
-Semaglutide Increase in blood levels Better glycaemic control Uncertain/No adaptation
Increased plasmatic exposure
Oncology medications
-TKI
-Immunotherapies Decreased absorption, reduced AUC Reduced PFS & OS Possible/ no simultaneous intake
Interaction with gut microbiome Reduced PFS & OS Uncertain/none

7. Expert opinion prescribed or self-medicated. Attention should be paid to the


net effect of PPIs on cancer treatment, as the deleterious effect
PPIs are one of the most prescribed drug classes in the world, due
on plasma exposure may be offset, at least in part, by
to their high efficacy and safety profile perceived as very favorable.
improved compliance because preventive effect on che­
However, many potential adverse effects of PPIs have been
motherapy-induced side effects.
reported in the literature, particularly through drug–drug
In addition, some formulations of cancer treatments have
interactions. Not all of the drug–drug interactions described
been developed to limit gastric pH-dependent drug interac­
have a significant clinical impact, and many remain controver­
tions. The impact of interaction with PPIs may be less with
sial, nevertheless clinicians should routinely question the risk
these new formulations. This still needs to be convincingly
of interactions, which may require dose adjustment or specific
demonstrated by experimental and real-life data.
therapeutic monitoring.
Clinicians should systematically, and carefully, assess
the benefit/risk ratio before introducing a new treatment Funding
with a potential interaction with PPIs. This paper was not funded.
While the clinical response profile induced by different PPIs,
at equivalent doses, can be considered equivalent, this is not
exactly the same for the pharmacokinetic profile. Declaration of interest
Knowledge of the pharmacokinetic profile may be of The authors have no relevant affiliations or financial involvement with any
particular interest when a potentially problematic interac­ organization or entity with a financial interest in or financial conflict with
tion is due to competitive inhibition of hepatic cyto­ the subject matter or materials discussed in the manuscript. This includes
chrome P450. Indeed, some PPIs have a high potential employment, consultancies, honoraria, stock ownership or options, expert
testimony, grants or patents received or pending, or royalties.
for CYP2C19 inhibition, namely omeprazole, esomeprazole,
and lansoprazole, whereas the others, pantoprazole, rabe­
prazole, and dexlansoprazole, have a much lower potential
Reviewer disclosures
for CYP2C19 inhibition. These may be preferred depending
on co-prescribed treatments. Peer reviewers on this manuscript have no relevant financial or other
relationships to disclose.
Assessment of the interaction of PPIs with cancer treat­
ments remains too fragmented, mostly based on low-quality
studies, which is problematic given its potential negative ORCID
impact on disease outcomes. As a substantial proportion of
Inès Ben Ghezala http://orcid.org/0000-0002-4002-7414
patients receive PPIs, it is very important that future clinical Maxime Luu http://orcid.org/0000-0002-9024-293X
trials rigorously and systematically collect PPI use, whether Marc Bardou http://orcid.org/0000-0003-0028-1837
344 I. BEN GHEZALA ET AL.

References pantoprazole-(13)C breath test in GERD patients: clinical relevance


for personalized medicine. J Breath Res. 2016 Dec 17;10(4):046017.
Papers of special note have been highlighted as either of interest (•) or of 19. Wood N, Tan K, Purkins L, et al., Effect of omeprazole on the
considerable interest (••) to readers. steady-state pharmacokinetics of voriconazole. Br J Clin
1. Shin JM, Sachs G. Pharmacology of proton pump inhibitors. Curr Pharmacol. 2003;56(1): 56–61.
Gastroenterol Rep. 2008 Dec;10(6):528–534. • A clear explanation PPI and antifugeal drug-to-drug intera
2. Shin JM, Kim N. Pharmacokinetics and pharmacodynamics of the ction.
proton pump inhibitors. J Neurogastroenterol Motil. 2013 Jan;19 20. Blanco Dorado S, Maronas Amigo O, Latorre-Pellicer A, et al.
(1):25–35. A multicentre prospective study evaluating the impact of
• A very clear description of PPIs’ pharmacology. proton-pump inhibitors omeprazole and pantoprazole on vorico­
3. Scarpignato C, Gatta L, Zullo A, et al. Effective and safe proton nazole plasma concentrations. Br J Clin Pharmacol. 2020 Aug;86
pump inhibitor therapy in acid-related diseases - a position paper (8):1661–1666.
addressing benefits and potential harms of acid suppression. BMC •• This is a prospective study.
Med. 2016 Nov 9;14(1):179. 21. Zvyaga T, Chang SY, Chen C, et al. Evaluation of six proton pump
4. Barkun AN, Almadi M, Kuipers EJ, et al. Management of nonvariceal inhibitors as inhibitors of various human cytochromes P450: focus
upper gastrointestinal bleeding: guideline recommendations from on cytochrome P450 2C19. Drug Metab Dispos. 2012 Sep;40
the international consensus group. Ann Intern Med. 2019 Dec (9):1698–1711.
3;171(11):805–822. 22. Yan M, Wu ZF, Tang D, et al. The impact of proton pump inhibitors
•• These are one of the most complete guidelines on the manage­ on the pharmacokinetics of voriconazole in vitro and in vivo.
ment of gastrointestinal bleeding issued by a group of world­ Biomed Pharmacother. 2018 Dec;108:60–64.
wide experts. 23. Huang Q, Liu Q, Yin T, et al. Effect of proton pump inhibitors on
5. Johnson DA, Katz PO, Armstrong D, et al. The safety of appropriate voriconazole concentrations in Chinese patients with malignant
use of over-the-counter proton pump inhibitors: an hematological diseases. Eur J Clin Pharmacol. 2020 Jun;76
evidence-based review and Delphi consensus. Drugs. 2017 Apr;77 (6):833–842.
(5):547–561. 24. Marsot A, Goirand F, Milesi N, et al. Interaction of thiopental with
6. Patel D, Bertz R, Ren S, et al. A systematic review of gastric esomeprazole in critically ill patients. Eur J Clin Pharmacol. 2013
acid-reducing agent-mediated drug-drug interactions with orally Sep;69(9):1667–1672.
administered medications. Clin Pharmacokinet. 2020 Apr;59 25. Ashbee HR, Barnes RA, Johnson EM, et al. Therapeutic drug mon­
(4):447–462. itoring (TDM) of antifungal agents: guidelines from the British
7. de Waal T, Rubbens J, Grimm M, et al. Exploring the effect of Society for Medical Mycology. J Antimicrob Chemother. 2014
esomeprazole on gastric and duodenal fluid volumes and absorp­ May;69(5):1162–1176.
tion of ritonavir. Pharmaceutics. 2020 Jul 17; 12(7):670 26. Lu M, Yan H, Yu C, et al. Proton pump inhibitors act synergistically
8. El Rouby N, Lima JJ, Johnson JA. Proton pump inhibitors: from with fluconazole against resistant Candida albicans. Sci Rep. 2020
CYP2C19 pharmacogenetics to precision medicine. Expert Opin Jan 16;10(1):498.
Drug Metab Toxicol. 2018 Apr;14(4):447–460. 27. Kapetas AJ, Abuhelwa AY, Sorich MJ, et al. Evidence-based guide­
9. Albengres E, Le Louët H, Tillement JP. Systemic antifungal agents. lines for drug interaction studies: model-informed time course of
drug interactions of clinical significance. Drug Saf. 1998 Feb;18 intestinal and hepatic CYP3A4 inhibition by clarithromycin. AAPS J.
(2):83–97. 2021 Aug 31;23(5):104.
10. Fotaki N, Klein S. Mechanistic understanding of the effect of PPIs 28. Calabresi L, Pazzucconi F, Ferrara S, et al. Pharmacokinetic interac­
and acidic carbonated beverages on the oral absorption of itraco­ tions between omeprazole/pantoprazole and clarithromycin in
nazole based on absorption modeling with appropriate in vitro health volunteers. Pharmacol Res. 2004 May;49(5):493–499.
data. Mol Pharm. 2013 Nov 4;10(11):4016–4023. 29. Saito M, Yasui-Furukori N, Uno T, et al. Effects of clarithromycin on
11. Jaruratanasirikul S, Sriwiriyajan S. Effect of omeprazole on the lansoprazole pharmacokinetics between CYP2C19 genotypes. Br
pharmacokinetics of itraconazole. Eur J Clin Pharmacol. 1998 J Clin Pharmacol. 2005 Mar;59(3):302–309.
Apr;54(2):159–161. 30. Cao S, Zhou G, Ou-Yang DS, et al. Pharmacokinetic interactions
12. Abuhelwa AY, Mudge S, Upton RN, et al. Mechanistic assessment of between ilaprazole and clarithromycin following ilaprazole, clari­
the effect of omeprazole on the in vivo pharmacokinetics of itra­ thromycin and amoxicillin triple therapy. Acta Pharmacol Sin. 2012
conazole in healthy volunteers. Eur J Drug Metab Pharmacokinet. Aug;33(8):1095–1100.
2019 Apr;44(2):201–215. 31. Jin BH, Yoo BW, Park J, et al. Pharmacokinetic drug interaction and
13. Hilton JF, Tu D, Seymour L, et al. An evaluation of the possible safety after coadministration of clarithromycin, amoxicillin, and
interaction of gastric acid suppressing medication and the EGFR ilaprazole: a randomised, open-label, one-way crossover, two par­
tyrosine kinase inhibitor erlotinib. Lung Cancer. 2013 Oct;82 allel sequences study. Eur J Clin Pharmacol. 2018 Sep;74
(1):136–142. (9):1149–1157.
14. Lindsay J, Mudge S, and Thompson GR 3rd. Effects of food and 32. Georgopoulos S, Papastergiou V. An update on current and advan­
omeprazole on a novel formulation of super bioavailability itraco­ cing pharmacotherapy options for the treatment of H. pylori
nazole in healthy subjects. Antimicrob Agents Chemother. 2018;62 infection. Expert Opin Pharmacother. 2021 Apr;22(6):729–741.
(12):e01723–18. 33. Lin YA, Wang H, Gu ZJ, et al. Effect of CYP2C19 gene polymorph­
15. Nield B, Larsen SR, van Hal SJ. Clinical experience with new for­ isms on proton pump inhibitor, amoxicillin, and levofloxacin triple
mulation SUBA(R)-itraconazole for prophylaxis in patients under­ therapy for eradication of Helicobacter pylori. Med Sci Monit. 2017
going stem cell transplantation or treatment for haematological Jun 3;23:2701–2707.
malignancies. J Antimicrob Chemother. 2019 Oct 1;74 34. Soares GA, Pires DW, Pinto LA, et al. The influence of omeprazole
(10):3049–3055. on the dissolution processes of pH-dependent magnetic tablets
16. Abbotsford J, Foley DA, Goff Z, et al. Clinical experience with assessed by pharmacomagnetography. Pharmaceutics. 2021 Aug
SUBA-itraconazole at a tertiary paediatric hospital. J Antimicrob 17; 13(8):1274.
Chemother. 2021 Jan 1;76(1):249–252. 35. Offman E, Schobelock MJ, Brickl R, et al. Pharmacokinetics and
17. Zonios D, Yamazaki H, Murayama N, et al. Voriconazole metabo­ pharmacodynamics of the antiplatelet combination aspirin
lism, toxicity, and the effect of cytochrome P450 2C19 genotype. (acetylsalicylic acid) plus extended-release dipyridamole are
J Infect Dis. 2014 Jun 15;209(12):1941–1948. not altered by coadministration with the potent CYP2C19 inhi­
18. Modak AS, Klyarytska I, Kriviy V, et al. The effect of proton pump bitor omeprazole. Am J Cardiovasc Drugs. 2013 Apr;13
inhibitors on the CYP2C19 enzyme activity evaluated by the (2):113–120.
EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 345

36. Colombier MA, Molina JM. Doravirine: a review. Curr Opin HIV AIDS. coadministered with dual antiplatelet therapy: systematic review.
2018 Jul;13(4):308–314. Circ Cardiovasc Qual Outcomes. 2015 Jan;8(1):47–55.
37. Khalilieh SG, Yee KL, Sanchez RI, et al. A study to evaluate doravir­ 54. Demcsák A, Lantos T, Bálint ER, et al. PPIs are not responsible for
ine pharmacokinetics when coadministered with acid-reducing elevating cardiovascular risk in patients on clopidogrel-a systematic
agents. J Clin Pharmacol. 2019 Aug;59(8):1093–1098. review and meta-analysis. Front Physiol. 2018;9:1550.
38. Van Den Abeele J, Kostantini C, Barker R, et al. The effect of 55. Farhat N, Fortin Y, Haddad N, et al. Systematic review and
reduced gastric acid secretion on the gastrointestinal disposition meta-analysis of adverse cardiovascular events associated with
of a ritonavir amorphous solid dispersion in fasted healthy volun­ proton pump inhibitors used alone or in combination with anti­
teers: an in vivo - in vitro investigation. Eur J Pharm Sci. 2020 Aug;1 platelet agents. Crit Rev Toxicol. 2019 Mar;49(3):215–261.
(151):105377. 56. Lin SF, Lin PC, Chang CC, et al. Investigation of the interaction
39. Pohl O, Osterloh I, Lecomte V, et al. Changes in gastric pH and in between proton pump inhibitors and clopidogrel using VerifyNow
pharmacokinetics of ulipristal acetate - a drug-drug interaction P2Y12 assay. Medicine (Baltimore). 2020 Dec 11;99(50):e23695.
study using the proton pump inhibitor esomeprazole. Int J Clin 57. Leonard CE, Bilker WB, Brensinger CM, et al. Comparative risk of
Pharmacol Ther. 2013 Jan;51(1):26–33. ischemic stroke among users of clopidogrel together with indivi­
40. Reau N, Robertson MN, Feng HP, et al. Concomitant proton pump dual proton pump inhibitors. Stroke. 2015 Mar;46(3):722–731.
inhibitor use does not reduce the efficacy of elbasvir/grazoprevir: 58. Shi W, Ni L, Yang J, et al. The clinical impact of proton pump
a pooled analysis of 1,322 patients with hepatitis C infection. inhibitors when co-administered with dual antiplatelet therapy
Hepatol Commun. 2017 Oct 1;1(8):757–764. in patients having acute myocardial infarction with low risk of
•• Clinical assessment of the clinical consequences of drug-to- gastrointestinal bleeding: insights from the China acute myo­
drug interaction. cardial infarction registry. Front Cardiovasc Med.
41. Trifiro G, Corrao S, Alacqua M, et al. Interaction risk with proton 2021;8:685072.
pump inhibitors in general practice: significant disagreement 59. Shi WC, Gao SD, Yang JG, et al. Impact of proton pump inhibitors
between different drug-related information sources. Br J Clin on clinical outcomes in patients after acute myocardial infarction:
Pharmacol. 2006 Nov;62(5):582–590. a propensity score analysis from China Acute Myocardial Infarction
42. Flamm S, Reddy KR, Zadeikis N, et al. Efficacy and pharmacokinetics (CAMI) registry. J Geriatr Cardiol. 2020 Nov 28;17(11):659–665.
of glecaprevir and pibrentasvir with concurrent use of 60. Kim MS, Song HJ, Lee J, et al. Effectiveness and safety of clopido­
acid-reducing agents in patients with chronic HCV infection. Clin grel co-administered with statins and proton pump inhibitors:
Gastroenterol Hepatol. 2019 Feb;17(3):527–535 e6. a Korean national health insurance database study. Clin
43. Tett S, Day R, Cutler D. Hydroxychloroquine relative bioavailability: Pharmacol Ther. 2019 Jul;106(1):182–194.
within subject reproducibility. Br J Clin Pharmacol. 1996 Mar;41 61. Bang CS, Joo MK, Kim BW, et al. The role of acid suppressants in the
(3):244–246. prevention of anticoagulant-related gastrointestinal bleeding:
44. Stoll F, Blank A, Mikus G, et al. Effect of pantoprazole on the a systematic review and meta-analysis. Gut Liver. 2020 Jan 15;14
absorption of hydroxychloroquinea a randomized drug-drug inter­ (1):57–66.
action trial in healthy adults. Clin Pharmacol Drug Dev. 2022 Feb;11 62. Cole JA, Taylor JS, Hangartner TN, et al. Reducing selection bias in
(2):285–290. case-control studies from rare disease registries. Orphanet J Rare
•• A randomised PK study. Dis. 2011 Sep;12(6):61.
45. Jallouli M, Galicier L, Zahr N, et al. Determinants of hydroxy­ 63. Gadzhanova SRE. Co-prescribing of warfarin with statins and pro­
chloroquine blood concentration variations in systemic lupus ton pump inhibitors in elderly Australians. Adv Pharmacoepidemiol
erythematosus. Arthritis Rheumatol. 2015 May;67(8):2176–2184. Drug Saf. 2014;3:3.
46. Dean L. Clopidogrel therapy and CYP2C19 genotype. In: Pratt VM, 64. Wang M, Zeraatkar D, Obeda M, et al. Drug-drug interactions with
Scott SA, Pirmohamed M, et al., editors. Medical genetics summa­ warfarin: a systematic review and meta-analysis. Br J Clin
ries. Bethesda (MD): National Center for Biotechnology Information Pharmacol. 2021 Nov;87(11):4051–4100.
(US); 2012. 65. Bertram V, Yeo K, Anoopkumar-Dukie S, et al. Proton pump inhibi­
47. Gilard M, Arnaud B, Le Gal G, et al. Influence of omeprazol on the tors co-prescribed with warfarin reduce warfarin control as mea­
antiplatelet action of clopidogrel associated to aspirin. J Thromb sured by time in therapeutic range. Int J Clin Pract. 2019 Nov;73
Haemost. 2006 Nov;4(11):2508–2509. (11):e13382.
48. Gilard M, Arnaud B, Cornily JC, et al. Influence of omeprazole on 66. Kuwayama T, Osanai H, Ajioka M, et al. Influence of proton pump
the antiplatelet action of clopidogrel associated with aspirin: the inhibitors on blood dabigatran concentrations in Japanese patients
randomized, double-blind OCLA (Omeprazole clopidogrel aspirin) with non-valvular atrial fibrillation. J Arrhythm. 2017 Dec;33
study. J Am Coll Cardiol. 2008 Jan 22;51(3):256–260. (6):619–623.
• A well-conduced randomized clinical trial. Unfortunately, the 67. Bolek T, Samos M, Stanciakova L, et al. The impact of proton pump
primary outcome is not a clinical event. inhibition on dabigatran levels in patients with atrial fibrillation.
49. Ho PM, Maddox TM, and Wang L, et al. Risk of adverse outcomes Am J Ther. 2019 May;26(3):e308–e313.
associated with concomitant use of clopidogrel and proton pump 68. Harada A, Ikushima I, Haranaka M, et al. Bioequivalence of a newly
inhibitors following acute coronary syndrome. JAMA. 2009 Mar developed dabigatran etexilate tablet versus the commercial cap­
4;301(9):937–944. sule and impact of rabeprazole-induced elevated gastric ph on
•• One of the leading studies on the topic. exposure in healthy subjects. Am J Cardiovasc Drugs. 2020 Jun;20
50. Muthiah MD, Zheng H, Chew NWS, et al. Outcomes of a (3):249–258.
multi-ethnic Asian population on combined treatment with clopi­ 69. Schnierer M, Samos M, Bolek T, et al. The effect of proton pump
dogrel and omeprazole in 12,440 patients. J Thromb Thrombolysis. inhibitor withdrawal on dabigatran etexilate plasma levels in
2021 Oct;52(3):925–933. patients with atrial fibrillation: a washout study. J Cardiovasc
51. Lau WC, Gurbel PA. The drug-drug interaction between proton Pharmacol. 2020 Apr;75(4):333–335.
pump inhibitors and clopidogrel. Cmaj. 2009 Mar 31;180 70. Ray WA, Chung CP, Murray KT, et al. Association of oral antic­
(7):699–700. oagulants and proton pump inhibitor cotherapy with hospitaliza­
52. Moreira Dias L. Pantoprazole: a proton pump inhibitor. Clin Drug tion for upper gastrointestinal tract bleeding. Jama. 2018 Dec 4;320
Investig. 2009;29(2):3–12. (21):2221–2230.
53. Melloni C, Washam JB, Jones WS, et al. Conflicting results between 71. Chan EW, Lau WC, Leung WK, et al. Prevention of dabigatran-related
randomized trials and observational studies on the impact of pro­ gastrointestinal bleeding with gastroprotective agents: a
ton pump inhibitors on cardiovascular events when population-based study. Gastroenterology. 2015 Sep;149(3):586–95.e3.
346 I. BEN GHEZALA ET AL.

72. Nies AT, Hofmann U, Resch C, et al. Proton pump inhibitors inhibit 89. Sim W, Jain SR, Lim WH, et al. Interactions between epidermal
metformin uptake by organic cation transporters (OCTs). PLoS One. growth factor receptor tyrosine kinase inhibitors and
2011;6(7):e22163. proton-pump inhibitors/histamine type-2 receptor antagonists in
73. Kim A, Chung I, Yoon SH, et al. Effects of proton pump inhibitors on non-small cell lung cancer: a systematic review and meta-analysis.
metformin pharmacokinetics and pharmacodynamics. Drug Metab Transl Lung Cancer Res. 2021 Aug;10(8):3567–3581.
Dispos. 2014 Jul;42(7):1174–1179. 90. Lalani AA, McKay RR, Lin X, et al. Proton pump inhibitors and
74. Flory J, Haynes K, Leonard CE, et al. Proton pump inhibitors do not survival outcomes in patients with metastatic renal cell
impair the effectiveness of metformin in patients with diabetes. Br carcinoma. Clin Genitourin Cancer. 2017 Dec;15(6):724–732.
J Clin Pharmacol. 2015 Feb;79(2):330–336. 91. Ha VH, Ngo M, Chu MP, et al. Does gastric acid suppression
75. Al-Bachaji IN, Al-Buhadiliy AK, Al-Kuraishy HM, et al. Proton pump affect sunitinib efficacy in patients with advanced or meta­
inhibitors regulate metabolic profile and glycaemic indices in static renal cell cancer? J Oncol Pharm Pract. 2015 Jun;21
patients with type 2 diabetes mellitus: a rising dawn of a new (3):194–200.
therapeutic concept. J Pak Med Assoc. 2019 Aug;69(8):S31–S35. 92. Mir O, Touati N, Lia M, et al. Impact of concomitant adminis­
76. Peng CC, Tu YK, Lee GY, et al. Effects of proton pump inhibitors on tration of gastric acid-suppressive agents and pazopanib on
glycemic control and incident diabetes: a systematic review and outcomes in soft-tissue sarcoma patients treated within the
meta-analysis. J Clin Endocrinol Metab. 2021 Oct 21;106 EORTC 62043/62072 trials. Clin Cancer Res. 2019 Mar 1;25
(11):3354–3366. (5):1479–1485.
• A well-conducted meta-analysis. 93. de Man FM, Hussaarts K, de With M, et al. Influence of the proton
77. Ginanneschi F, Volpi N, Giannini F, et al. Rhabdomyolysis in an pump inhibitor esomeprazole on the bioavailability of regorafenib:
elderly multitreated patient: multiple drug interactions after statin a randomized crossover pharmacokinetic study. Clin Pharmacol
withdrawal. J Neurol Sci. 2014 Jan 15;336(1–2):284–287. Ther. 2019 Jun;105(6):1456–1461.
78. Kashour T, Al-Tannir M, Bahamid R. Changing prescription pattern 94. Van De Sijpe G, Beuselinck B, Van Nieuwenhuyse T, et al. Impact of
of omeprazole among patients receiving clopidogrel. Int Heart J. concomitant acid suppressive therapy on pazopanib efficacy and
2014;55(2):93–95. dose reductions in patients with metastatic renal cell carcinoma.
79. Baekdal TA, Breitschaft A, Navarria A, et al. A randomized study Eur J Clin Pharmacol. 2020 Sep;76(9):1273–1280.
investigating the effect of omeprazole on the pharmacokinetics of 95. Krens SD, Lubberman FJE, van Egmond M, et al. The impact of a
oral semaglutide. Expert Opin Drug Metab Toxicol. 2018 Aug;14 1-hour time interval between pazopanib and subsequent intake of
(8):869–877. gastric acid suppressants on pazopanib exposure. Int J Cancer.
80. Raoul JL, Guerin-Charbonnel C, Edeline J, et al. Prevalence of pro­ 2021 Jun 1;148(11):2799–2806.
ton pump inhibitor use among patients with cancer. JAMA 96. Yin OQ, Giles FJ, Baccarani M, et al. Concurrent use of proton pump
Network Open. 2021 Jun 1;4(6):e2113739. inhibitors or H2 blockers did not adversely affect nilotinib efficacy
81. Budha NR, Frymoyer A, Smelick GS, et al. Drug absorption interac­ in patients with chronic myeloid leukemia. Cancer Chemother
tions between oral targeted anticancer agents and PPIs: is Pharmacol. 2012 Aug;70(2):345–350.
pH-dependent solubility the Achilles heel of targeted therapy? 97. Koutake Y, Taniguchi J, Yasumori N, et al. Influence of proton pump
Clin Pharmacol Ther. 2012 Aug;92(2):203–213. inhibitors and H2-receptor antagonists on the efficacy and safety of
82. Peters S, Zimmermann S, Adjei AA. Oral epidermal growth factor dasatinib in chronic myeloid leukemia patients. Int J Hematol. 2020
receptor tyrosine kinase inhibitors for the treatment of non-small Jun;111(6):826–832.
cell lung cancer: comparative pharmacokinetics and drug-drug 98. Hussaarts K, Veerman GDM, Jansman FGA, et al. Clinically relevant
interactions. Cancer Treat Rev. 2014 Sep;40(8):917–926. drug interactions with multikinase inhibitors: a review. Ther Adv
83. van Leeuwen RW, van Gelder T, Mathijssen RH, et al. Drug-drug Med Oncol. 2019;11:1758835918818347.
interactions with tyrosine-kinase inhibitors: a clinical perspective. 99. Rassy E, Cerbone L, Auclin E, et al. The effect of concomitant proton
Lancet Oncol. 2014 Jul;15(8):e315–26. pump inhibitor and cabozantinib on the outcomes of patients with
•• A comprehensive overview of known or suspected drug–drug metastatic renal cell carcinoma. Oncologist. 2021 May;26
interactions between tyrosine-kinase inhibitors and other (5):389–396.
drugs. 100. Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome mod­
84. Veerman GDM, Hussaarts K, Peric R, et al. Influence of cow’s milk ulates response to anti-PD-1 immunotherapy in melanoma
and esomeprazole on the absorption of erlotinib: a randomized, patients. Science. 2018 Jan 5;359(6371):97–103.
crossover pharmacokinetic study in lung cancer patients. Clin 101. Viaud S, Saccheri F, Mignot G, et al. The intestinal microbiota
Pharmacokinet. 2021 Jan;60(1):69–77. modulates the anticancer immune effects of cyclophosphamide.
85. Ohgami M, Kaburagi T, Kurosawa A, et al. Effects of proton pump Science. 2013 Nov 22;342(6161):971–976.
inhibitor coadministration on the plasma concentration of erlotinib 102. Trifan A, Stanciu C, Girleanu I, et al. Proton pump inhibitors therapy
in patients with non-small cell lung cancer. Ther Drug Monit. 2018 and risk of Clostridium difficile infection: systematic review and
Dec;40(6):699–704. meta-analysis. World J Gastroenterol. 2017 Sep 21;23
86. Yokota H, Sato K, Okuda Y, et al. Effects of histamine 2-receptor (35):6500–6515.
antagonists and proton pump inhibitors on the pharmacokinetics 103. Weersma RK, Zhernakova A, Fu J. Interaction between drugs and
of gefitinib in patients with non-small-cell lung cancer. Clin Lung the gut microbiome. Gut. 2020 Aug;69(8):1510–1519.
Cancer. 2017 Nov;18(6):e433–e439. 104. Kato K, Mizuno T, Koseki T, et al. Concomitant proton pump
87. Song HJ, Rhew K, Lee YJ, et al. Acid-suppressive agents and survival inhibitors and immune checkpoint inhibitors increase nephritis
outcomes in patients with cancer: a systematic review and frequency. Vivo. 2021 Sep–Oct;35(5):2831–2840.
meta-analysis. Int J Clin Oncol. 2021 Jan;26(1):34–50. 105. Chalabi M, Cardona A, Nagarkar DR, et al. Efficacy of chemotherapy
88. Sharma M, Holmes HM, Mehta HB, et al. The concomitant use of and atezolizumab in patients with non-small-cell lung cancer
tyrosine kinase inhibitors and proton pump inhibitors: prevalence, receiving antibiotics and proton pump inhibitors: pooled post
predictors, and impact on survival and discontinuation of therapy hoc analyses of the OAK and POPLAR trials. Ann Oncol. 2020
in older adults with cancer. Cancer. 2019 Apr 1;125(7):1155–1162. Apr;31(4):525–531.

View publication stats

You might also like