Effects of genetic polymorphisms on the pharmacokinetics and pharmacodynamics of proton pump inhibitors

Effects of genetic polymorphisms on the pharmacokinetics and pharmacodynamics of proton pump inhibitors

Journal Pre-proof Effects of Genetic Polymorphisms on the Pharmacokinetics and Pharmacodynamics of Proton Pump Inhibitors He-Jian Zhang, Xue-Hui Zhang...

4MB Sizes 0 Downloads 18 Views

Journal Pre-proof Effects of Genetic Polymorphisms on the Pharmacokinetics and Pharmacodynamics of Proton Pump Inhibitors He-Jian Zhang, Xue-Hui Zhang, Jie Liu, Lu-Ning Sun, Yi-Wen Shen, Chen Zhou, Hong-Wen Zhang, Li-Jun Xie, Juan Chen, Yun Liu, Yong-Qing Wang

PII:

S1043-6618(19)31318-0

DOI:

https://doi.org/10.1016/j.phrs.2019.104606

Reference:

YPHRS 104606

To appear in:

Pharmacological Research

Received Date:

11 July 2019

Revised Date:

13 December 2019

Accepted Date:

13 December 2019

Please cite this article as: Zhang H-Jian, Zhang X-Hui, Liu J, Sun L-Ning, Shen Y-Wen, Zhou C, Zhang H-Wen, Xie L-Jun, Chen J, Liu Y, Wang Y-Qing, Effects of Genetic Polymorphisms on the Pharmacokinetics and Pharmacodynamics of Proton Pump Inhibitors, Pharmacological Research (2019), doi: https://doi.org/10.1016/j.phrs.2019.104606

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 Published by Elsevier.

Effects of Genetic Polymorphisms on the Pharmacokinetics and Pharmacodynamics of Proton Pump Inhibitors He-Jian Zhang1,2, Xue-Hui Zhang3*, Jie Liu4, Lu-Ning Sun1, Yi-Wen Shen1, Chen Zhou1, Hong-Wen Zhang1, Li-Jun Xie1, Juan Chen1, Yun Liu5#, Yong-Qing Wang1,3# 1

Research Division of Clinical Pharmacology, the First Affiliated Hospital of Nanjing

Medical University & Jiangsu Province Hospital, Nanjing, China Xuzhou Medical University, Xuzhou, China

3

Department of Pharmacy, Jiangsu Shengze Hospital, Nanjing Medical University,

ro of

2

4

-p

Suzhou, China

Pukou Branch, the First Affiliated Hospital of Nanjing Medical University, Nanjing,

Department of Geriatrics endocrinology, the First Affiliated Hospital of Nanjing

lP

5

re

China

na

Medical University & Jiangsu Province Hospital, Nanjing, China

*

Co-first author: Xue-Hui Zhang and He-Jian Zhang contributed equally to this work

#

ur

as co-first authors.

Co-correspondence authors.

Jo

Corresponding author:

Yong-Qing Wang, PhD, Research Division of Clinical Pharmacology, the First Affiliated Hospital of Nanjing Medical University & Jiangsu Province Hospital, 300 Guangzhou Road, Nanjing 210029, China. E-mail address: [email protected]. Yun Liu, PhD, Department of Geriatrics endocrinology, the First Affiliated Hospital of 1

Nanjing Medical University & Jiangsu Province Hospital, E-mail address: [email protected].

re

-p

ro of

Graphical abstract

Abstract

lP

Proton pump inhibitors (PPIs) are used widely for the treatment of acid-related

na

disorders. Despite their excellent efficacy and tolerance, the pharmacodynamics and pharmacokinetics of PPIs are affected by each patient’s CYP2C19 and gastric

ur

H+,K+-ATPase genotype. The aim of this review was to analyze the effect of genetic polymorphisms on the pharmacodynamic and pharmacokinetic properties of PPIs.

Jo

The gastric acid-suppressive effect of PPIs is affected by both gastric H+,K+-ATPase and CYP2C19 polymorphisms, although gastric H+,K+-ATPase polymorphisms may have larger effects. Ilaprazole and rabeprazole show relatively small differences in the pharmacokinetic and pharmacodynamic properties and clinical efficacy among the different CYP2C19 genotypes. Compared with oral administration, the intravenous 2

infusion of PPIs is less affected by CYP2C19 polymorphism. At the same dose, each enantiomer has less variation among different CYP2C19 genotypes than a racemate mixture. Keywords: Proton pump inhibitors (PPIs), CYP2C19, gastric H+,K+-ATPase, Polymorphism, Pharmacokinetics, Pharmacodynamics

ro of

Chemical compounds

Omeprazole (PubChem CID: 4594); Esomeprazole (PubChem CID: 9568614);

Lansoprazole (PubChem CID: 3883); Dexlansoprazole (PubChem CID: 9578005);

-p

Pantoprazole (PubChem CID: 4679); Rabeprazole (PubChem CID: 5029); Ilaprazole

re

(PubChem CID: 214351).

lP

Abbreviations

na

PPIs: proton pump inhibitors; H. pylori: Helicobacter pylori; H+,K+-ATPase: hydrogen-potassium adenosine triphosphatase; CYP450: cytochrome P450; PK:

ur

pharmacokinetic; PD: pharmacodynamic; UMs: ultra-rapid metabolizers; homEMs: homozygous extensive metabolizers; hetEMs: heterozygous extensive metabolizers;

Jo

PMs: poor metabolizers; AUC: area under the concentration versus time curve; Cmax: maximum plasma concentration; t1/2: elimination half-life; TpH>4 (%): percentage time of intragastric pH>4.0; MR: metabolic rate; H2RAs: histamine 2 receptor antagonists; SNPs: single nucleotide polymorphisms.

3

Introduction Proton pump inhibitors (PPIs) are a class of acid-suppressing drugs that is used widely for the treatment of acid-related disorders, such as peptic ulcer disease, gastroesophageal reflux disease, and Zollinger-Ellison syndrome[1-3]. In addition to the general therapy for acid-related diseases, PPIs can also be used to treat other diseases, such as Helicobacter pylori (H. pylori) infection, cancer, and viral

ro of

infections[4].

Gastric hydrogen-potassium adenosine triphosphatase (H+,K+-ATPase, proton

pump), an α, β-heterodimeric enzyme, has a key role in the secretion of gastric acid.

-p

PPIs are benzimidazole-based compounds that suppress gastric acid secretion by

re

irreversibly blocking the gastric H+,K+-ATPase after the drug is converted into its active form in gastric parietal cells[5], as the active sulfonamide formulation can

lP

covalently bind to H+,K+-ATPase. Not all PPIs bind to the same cysteines: rabeprazole

na

binds to cysteines 813, 892, 822, and 321; lansoprazole binds to cysteines 813, 892, and 321; omeprazole binds to cysteines 813 and 892; and pantoprazole binds to

ur

cysteines 813 and 822. In some cases, the affinity of PPIs decreases, which may lead to a reduction in acid inhibition efficacy[6, 7].

Jo

Cytochrome P450 (CYP450) enzymes are responsible for the metabolism of a

majority of therapeutic drugs and have important roles in bioavailability, elimination, and drug-drug interactions. PPIs are mainly metabolized by CYP450 enzymes; CYP2C19 and CYP3A4/5 are the two main isoenzymes of the CYP superfamily that mediate the hydroxylation and sulfoxidation of PPIs[8]. These enzymes are expressed 4

in humans and may influence both the pharmacokinetic (PK) and pharmacodynamic (PD) profiles of PPIs[9]. Most clinical research has reported CYP2C19 genotypes by using star (*) allele nomenclature; the main alleles and their respective influences on the activity of CYP2C19 enzymes are shown in Figure 1. The genotypes of CYP2C19 are classified as ultra-rapid, homozygous extensive, heterozygous extensive or poor metabolizers, depending on the genotypic combination of an individual. Individuals

ro of

with the higher activity allele (*17) are classified as ultra-rapid metabolizers (UMs). CYP2C19*17 is a gain-of-function allele, which is thought to increase the clearance and reduce plasma concentrations of PPIs compared with the normal allele.

-p

Individuals with two normal (non-mutant, *1/*1) alleles are classified as homozygous

re

extensive metabolizers (homEMs), and are able to produce an abundance of enzymes and to metabolize PPIs at a faster rate, which may lower the anti-secretory efficacy of

lP

PPIs. Individuals with one normal function allele and one no-function allele (taure 1)

na

are classified as heterozygous extensive metabolizers (hetEMs), whereas individuals with two copies of the CYP2C19 no-function allele are classified as poor metabolizers

ur

(PMs). CYP2C19 PMs have a much slower rate of metabolism, conferring greater bioavailability and subsequently increasing the anti-secretory efficacy of PPIs[10-12].

Jo

There are marked geographic differences in the frequency distribution of these genotypes[11-15]. The frequency of PMs is much higher in Asian populations, such as Japanese and Chinese populations, than in American, white European, or African populations (Table 1)[12]. Previous studies have reported significant relationships between CYP2C19 polymorphisms and PK/PD parameters after the administration of 5

omeprazole[16], rabeprazole[17, 18], lansoprazole[19, 20], and pantoprazole[21]. This review has focused on the differences in PK/PD parameters and clinical efficacy of different PPIs among the different genotypes of CYP2C19 and gastric H+,K+-ATPase. In addition, we evaluated the literature related to administration methods to determine the underlying patterns of how genetic polymorphisms affect

ur

na

lP

re

-p

ro of

the PK/PD of PPIs.

Figure 1. Allelic variants and genotypes of CYP2C19. UMs: Ultra-metabolisers; HomEMs:

Jo

homozygous extensive metabolizers; HetEMs: heterozygous extensive metabolizers; PMs: poor metabolizers. See http://www.cypalleles.ki.se/cyp2c19.htm for updates on CYP2C19 allelic variants and nomenclature.

6

Table 1. Frequencies of CYP2C19 genotypes among different races[11-15]. Phenotype

Ratio

Metabolism of PPIs

Asian

Caucasian

African

UMs

2%-17%

26%-34%

9%-28%

At highest rates

HomEMs

23%-53%

41%-70%

29%-42%

At higher rates

HetEMs

35%-55%

22%-30%

32%-49%

At moderate rates

PMs

10%-23%

1%-5%

3%-4%

At lowest rates

UMs: Ultra-metabolisers; HomEMs: homozygous extensive metabolizers; HetEMs: heterozygous

ro of

extensive metabolizers; PMs: poor metabolizers.

Omeprazole

Omeprazole, a first-generation PPI, is the most extensively studied PPI for the

-p

treatment of acid-related diseases[22]. Omeprazole is mainly metabolized by CYP2C19 to hydroxyomeprazole, and partially metabolized by CYP3A4 to

re

omeprazole sulfone, which is subsequently metabolized by CYP2C19 to

lP

hydroxyomeprazole sulfone[23] (Figure 2). The success rate of H. pylori eradication may be impacted by the CYP2C19 genotype; the eradication rates of the five main

na

PPIs are summarized in Table 2. In addition, the PK/PD parameters and clinical efficacy of omeprazole are significantly impacted by the genetic polymorphism of

ur

CYP2C19. The PK parameters (AUC, Cmax, t1/2, and clearance) and PD properties

Jo

(TpH>4 (%) and 24-h median intragastric pH) were significantly different between CYP2C19 PMs and homEMs after a single dose of oral omeprazole (Table 3)[24, 25]. The mean area under the concentration versus time curve (AUC) of omeprazole after a single oral dose in homEMs was much lower than that in PMs; the ratios of maximum plasma concentration (Cmax) and elimination half-life (t1/2) value in PMs vs. homEMs for omeprazole were consistent with the observations in AUC; in addition, 7

homEMs exhibited significantly higher clearance than PMs (Table 3). The difference in mean AUC after a single intravenous dose was relatively small among CYP2C19 genotypes, although the mean AUC in PMs was still higher than that in EMs[16, 26]. The difference in relative AUC ratios after oral dosing among different CYP2C19 genotypes is greater than that after intravenous dosing. One possible explanation may be the absence of inter-formulation and inter-individual differences in the absorption

ro of

of omeprazole when using an intravenous dosage form. PPIs (such as omeprazole)

reach maximal plasma concentrations within 2 h after oral dosing and are eliminated from systemic circulation quickly. The PK/PD parameters after single oral, single

-p

intravenous, repeated oral, and repeated intravenous doses of omeprazole in different

re

genotypes are summarized in Table 3. After repeated doses, similar differences in 24-h median pH and the percentage time of intragastric pH>4.0 (TpH>4 (%)) were observed

lP

among different CYP2C19 genotypes[15, 16, 24, 27-32]. There is a greater gastric

na

acid-suppression effect in PMs than in EMs, which is in accordance with the rate of H. pylori eradication shown in Table 2. A significant increase in 24-h median pH in EMs

ur

was observed during the progression from single dose to repeated doses, and the PK parameters of omeprazole were also different between single dose and repeated doses,

Jo

with a significantly higher Cmax and AUC after repeated omeprazole dosing in EMs, but not in PMs[16]. The current PPIs on the market have a relatively short t1/2 of 1–2 h, which means they are cleared quickly from the blood. However, PK parameters (AUC, Cmax, t1/2, and clearance) reveal a significant accumulation in EMs after repeated doses of PPIs. 8

This may be because PPIs are inhibitors of hepatic microsomal enzymes, which leads to the inhibition of CYP2C19 enzymes after repeated doses. In addition, only the pumps that are active can be inhibited on the first day of once-a-day dosing, and not all pumps are active. On the second day, more newly activated pumps may be inhibited by PPIs. Generally, at steady-state, 70% of the pump population is inhibited by the third day[16, 33].

ro of

Owing to the specific binding between drugs and molecules (e.g., transporters, receptors, enzymes, and DNA), the metabolic process and clinical efficacy of PPIs may differ between individual stereoisomers and racemic mixtures, even if the

-p

different stereoisomers have equal pharmacological activity[22, 34-36]. Esomeprazole,

re

the S-isomer of omeprazole, is a more potent acid inhibitor than omeprazole and was first marketed in America in 2001[22, 37]. It is also metabolized by CYP2C19, but the

lP

clearance is lower than that of omeprazole. In a comparison of the acid-inhibitory

na

effect between esomeprazole and omeprazole, CYP2C19 genotype-dependent differences were found to be smaller after the oral administration of esomeprazole (20

ur

mg twice daily), although there was no significant difference between esomeprazole and omeprazole in TpH>4 (%). In addition, in HomEMs, the median 24-h intragastric

Jo

pH on Day 7 with esomeprazole was significantly higher than that with omeprazole[30]. No significant difference in the acid-suppression effect was observed between homEMs and hetEMs after either a single dose or repeated dosing (5 days) of esomeprazole. The plasma levels were higher after repeated intravenous dosing than after single dose administration as the clearance was decreased by 29%; thus, the 9

increased AUC of esomeprazole following repeated doses is probably due to a combination of a decreased systemic clearance and a decrease in first-pass

lP

re

-p

ro of

elimination[38].

na

Figure 2. Metabolic pathways of omeprazole. The thicker the arrow, the larger the contribution of

Jo

ur

the CYP isoforms to the metabolic pathway.

10

Table 2. H. pylori eradication rates in different treatment regimens according to CYP2C19 Genetic Polymorphisms Compound

HomEMs

HetEMs

PMs

Omeprazole[39-42]

61%-85%

83%-90%

100%

Lansoprazole[43, 44]

74%-80%

81%-98%

85%-100%

Pantoprazole[45-47]

69%-87%

77%-84%

82%-100%

Rabeprazole[40, 44, 45, 48, 49]

60%-89%

62%-97%

72%-100%

Esomeprazole[49]

73%-88%

91%-95%

90%-100%

ro of

HomEMs: homozygous extensive metabolizers; HetEMs: heterozygous extensive metabolizers;

Jo

ur

na

lP

re

-p

PMs: poor metabolizers.

11

f oo

Table 3. Differences in pharmacokinetic and pharmacodynamic parameters for PPIs according to CYP2C19 polymorphisms.

Pantoprazole[65-68]

24-h Median pH

TpH> 4(%)

HomEMs:HetEMs:PMs

HomEMs:HetEMs:PMs

HomEMs:HetEMs:PMs

HomEMs:HetEMs:PMs

HomEMs:HetEMs:PMs

HomEMs:HetEMs:PMs

1.0:(1.1-3.7):(1.4-20.0) 1.0:(1.5-1.7):(1.5-4.3)) 1.0:(1.3-5.3):(2.0-13.1) 1.0:(1.0-1.3):(1.4-4.8) 1.0:(1.4-2.1):(3.7-8.5) 1.0:(1.4-1.8):(2.8-4.0) 1.0:1.7:3.9 1.0:(4.2-7.4)* 1.0:(1.2-2.9):(1.4-5.1) 1.0:(2.3-3.2):(4.1-4.9) 1.0:(1.0-2.3):(1.2-4.3) 1.0:(1.1-3.0):(1.7-5.3) -

1.0:(1.1-2.6):(1.8-7.6) 1.0:1.1:1.2 1.0:(1.3-2.5):(1.5-4.3) 1.0:1.2:1.4 1.0:(1.1-1.7):(1.6-2.8) 1.0:(1.2-1.5):(1.1-1.4) 1.0:1.3:2.0 1.0:(1.4-1.8) * 1.0:1.2:1.2 1.0:3.0:3.8 1.0:(1.1-1.7):(1.8-3.6) 1.0:(1.5-2.3):(2.2-4.1) -

1.0:(0.7-2.7):(1.2-3.8) 1.0:(1.0-1.3):(1.3-2.7) 1.0:(1.0-1.9):(1.0-3.5) 1.0:1.2:1.2 1.0:(1.0-2.1):(1.9-5.6) 1.0:(1.2-1.4):(3.1-3.5) 1.0:1.3:3.3 1.0:(2.2-3.5) * 1.0:1.3:5.7 1.0:(1.3-3.4) * 1.0:(1.1-2.3):(1.3-3.8) 1.0:(1.0-1.9):(1.2-3.2) -

(3.6-20.7):(3.2-5.9):1.0 (1.4-6.4):(1.4-4.0):1.0 (4.2-13.5):(2.5-2.7):1.0 1.1:1.1:1.0 (3.4-8.5):(2.4-4.3):1.0 (2.9-3.2):(1.5-2.2):1.0 3.2:2.0:1.0 4.7:2.9:1.0 5.8:4.0:1.0 (1.6-4.2):1.0* (1.6-5.2):(1.3-2.8):1.0 3.8:3.2:1.0 -

(0.9-4.4):(1.1-6.8):(1.3-6.7) (2.6-4.7):(4.3-6.3):(5.3-7.4) (1.5-5.9):(1.6-6.6):(4.0-7.6) (2.6-6.1):(4.3-6.8):(5.3-7.5) (3.6–4.8):(4.4–5.8):(5.0–6.4) (4.2-7.6):(6.2-7.8):(5.5-7.9) (3.5-5.5):(3.4-6.9):(4.7-7.3) (3.8–4.1):(3.4–5.0):(5.0–6.1) (2.7-6.9):(2.9-6.7):(6.6-7.2) 4.1:(6.1-6.2):(4.9-5.6) (4.2-8.0):(5.6-7.4):(6.0-6.6) (2.1-5.2):(2.0-6.0):(3.1-6.8) (2.1-6.4):(2.9-6.5):(4.1-7.4) -

(14-32):(27-53):(19-78) 59:66:79 (23-62):(24-75):(48-95) (58-78):(80-86):(93-94) 20:16# 73:86:85 (26-61):(37-69):(72-93) 49:75:90 32:48:44 64:66:100 44:54:60 84:93:99 (45-53):(51-68):(68-89) (36-75):(53-78):(53-91) -

pr

clearance

Jo ur

Rabeprazole[18, 69-79]

t1/2

e-

Lansoprazole[7, 19, 20, 57-64]

Single oral dose Single intravenous dose Repeated oral dose Repeated intravenous dose Single oral dose Single intravenous dose Repeated oral dose Repeated intravenous dose Single oral dose Single intravenous dose Repeated oral dose Repeated intravenous dose Single oral dose Single intravenous dose Repeated oral dose Repeated intravenous dose

Cmax

na l

Omeprazole[10, 14-17, 20, 24-32, 50-56]

AUC

Pr

compound

AUC: Area under the concentration versus time curve; Cmax: Maximum plasma concentration; t1/2: elimination half-life; Median pH: median pH value; TpH> 4(%): percent of time that intragastric pH is above 4.0. HomEMs: homozygous extensive metabolizers; HetEMs: heterozygous extensive metabolizers; PMs, poor metabolizers. Pharmacokinetic data is standardized with homEM (Cmax, AUC, t1/2) or PM (Clearance) in each study to visualize the effects of polymorphisms on pharmacokinetic parameters of PPIs. * Relative ratios between EMs and PMs. # Relative ratios between HomEMs and HetEMs.

12

Lansoprazole Lansoprazole, another first-generation PPI, is also a benzimidazole derivative. It has been used widely for the treatment of H. pylori infection (in combination with amoxicillin and clarithromycin), gastroesophageal reflux, and other acid-related disorders[80]. There are significant inter-individual differences in the PK of lansoprazole and its metabolites, which are related to genetic polymorphisms of

ro of

CYP450; a summary of PK/PD parameters of lansoprazole is shown in Table 3.

Lansoprazole is predominantly metabolized to 5'-hydroxy lansoprazole by CYP2C19

through hydroxylation, lansoprazole sulfone by CYP3A4/5 through sulfoxidation, and

-p

then further hydroxylated to its 5'-hydroxy metabolite by CYP2C19 (Figure 3). When

re

administered orally, the hydroxylation of lansoprazole was significantly influenced by CYP2C19 polymorphisms, and PMs exhibited a greater AUC and longer t1/2 than

lP

homEMs and hetEMs[19, 54, 61]. In terms of the main metabolites (5'-hydroxy

na

lansoprazole and lansoprazole sulfone), the metabolic rate (MR) of the conversion of lansoprazole to 5'-hydroxy lansoprazole is lower in PMs, whereas the MR of

ur

lansoprazole to lansoprazole sulfone is markedly higher. The metabolic ratio of lansoprazole to lansoprazole sulfone of PMs was approximately 12.6-fold higher than

Jo

that of EMs[19, 57, 62]. The Tmax of lansoprazole sulfone in CYP2C19 PMs was approximately 4 h, but approximately 0.76 h in EMs. In particular, the AUC and Cmax of lansoprazole sulfone in CYP2C19 PMs were approximately 75.1- and 8.7-fold higher than those in EMs, which means that CYP2C19 polymorphisms have a significant effect on the metabolic process of lansoprazole in vivo[81]. Compared 13

with homEMs, the PK parameters (AUC, Cmax, and t1/2) of lansoprazole are markedly higher in PMs after single oral administration; these results are summarized in Table 3. There was no significant difference between homEMs and hetEMs, whereas the clearance of lansoprazole was significantly lower in PMs[19, 60, 61, 63]. The PK parameters indicated relatively small differences after repeated oral doses among different CYP2C19 genotypes, although the mean AUC in PMs was still higher than

ro of

in EMs. The values of AUC and t1/2 in CYP2C19 PMs were higher than those in EMs after a single intravenous administration of lansoprazole[81]. In addition, intravenous formulations of lansoprazole were observed to exert single- and multiple-dose effects

-p

on PK/PD properties, similar to those observed after administration with the same

re

dose of intact capsules in healthy volunteers[82]. Intravenous lansoprazole is approved in the USA for short-term use (up to 7 days) in patients with erosive

lP

esophagitis who are temporarily unable to take oral formulations and is still the first

na

choice for acute non-variceal gastrointestinal bleeding[83]. In addition, some patients requiring acid-suppression therapy may be unable to take oral medications owing to

ur

dysphagia. Therefore, lansoprazole injection cannot be replaced by its oral formulation in many cases.

Jo

Lansoprazole is a racemic mixture of its S- and R-enantiomers; dexlansoprazole

is the R-enantiomer of lansoprazole. As with lansoprazole[30, 61], the PK/PD parameters of dexlansoprazole[7] are also significantly affected by CYP2C19 polymorphism. Our previous study demonstrated that the mean AUC had ratios of 1.00, 1.85, and 3.95 in homEMs, hetEMs, and PMs, respectively, after a single 14

intravenous administration (30 mg) of dexlansoprazole; the t1/2 value in PMs was approximately 3.47-fold higher than that in homEM, while PD parameters divided by CYP2C19 genotypes had relatively small differences between homEMs and PMs[7]. In addition, gastric H+,K+-ATPase polymorphisms have a significant influence on the acid-suppression effect of dexlansoprazole. Significant differences between gastric H+,K+-ATPase homozygote and wildtype were found in the Δ (delta)-values (values at pH-t

ro of

baseline subtracted from values after administration) for the mean pH, AUCτ 1-τ 2 , TpH≥3 (%), TpH≥4 (%), and TpH≥6 (%) during 0–4, 14–24, and 0–24 h intervals, whereas PK parameters were not affected by the gastric H+,K+-ATPase genotype.

-p

Further analysis to compare the PD properties between CYP2C19 and gastric

re

H+,K+-ATPase genotypes was undertaken to reveal which genotype had a stronger gastric acid suppression effect. There was no significant difference in acid inhibition

lP

effect among CYP2C19 homEMs, hetEMs, and PMs during the first 4 h after

na

intravenous administration of dexlansoprazole, whereas H+,K+-ATPase homozygotes pH-t

had significantly greater gastric acid-suppression effects for ΔAUCτ 1-τ 2 , ΔTpH≥3(%),

ur

and ΔTpH≥4(%) than heterozygotes or wildtypes. Even in CYP2C19 EMs, the same results were observed among each group of H+,K+-ATPase genotypes which indicated

Jo

that the gastric H+,K+-ATPase polymorphisms may be one of the major causes of inter-individual differences in efficacy. This provides a partial explanation for why some patients cannot achieve a satisfactory gastric acid-suppression effect after a conventional clinical dose of PPIs[7].

15

ro of -p

lP

the CYP isoforms to the metabolic pathway.

re

Figure 3. Metabolic pathways of lansoprazole. The thicker the arrow, the larger the contribution of

Pantoprazole

na

Pantoprazole, as a traditional PPI and benzimidazole derivative, is used widely in clinical practice for acid inhibition and the treatment of gastroesophageal reflux disease,

ur

peptic ulcer diseases, and infection with H. pylori[58, 84]. It is mainly metabolized by

Jo

different metabolic enzymes, as shown in Figure 4 (CYP2C19, CYP3A4). The blood concentration of pantoprazole at 3 h post-oral dose was significantly lower in carriers of the CYP2C19*17/*17 genotype than the CYP2C19 wild-type after a single oral dose of pantoprazole (40 mg). Volunteers with the CYP2C19*2/*2 allele showed a 506% increase in AUC and a 572% prolongation in t1/2. The lowest clearance was observed in the carriers of the *2/*2 genotype (3.68 L/h) and the highest clearance in 16

subjects with the *17/*17 genotype (31.13 L/h)[67]. In addition, the 1-year cumulative failure rate of on-demand therapy was significantly higher in homEMs than in hetEMs or PMs after a continuous 40 mg daily oral dose of pantoprazole tablets for patients with severe reflux esophagitis (Los Angeles grade C or D). The mean monthly number of tablets for pantoprazole was higher in homEMs and hetEMs than in PMs (18.6 vs 16.3 vs 11.5, P < 0.05). Sheu et al[85] proposed that the effective shift to on-demand

ro of

therapy was mainly dependent on the CYP2C19 genotype of patients; with reflux

esophagitis, patients with Los Angeles grade C or D showed perfect recovery after

repeated oral dosing of pantoprazole. It has been reported that Cmax and AUC values

-p

are much higher in PMs than those in homEMs and hetEMs after oral administration

re

of pantoprazole[67, 68], although the differences were smaller than those after administration of omeprazole or lansoprazole. Deshpande et al.[68] proposed that

lP

PMs had the highest AUC, whereas UMs had the lowest AUC on the fifth day after

na

oral administration of pantoprazole in Asian individuals. These data were in accordance with the observations made in Caucasians after a single administration of

ur

pantoprazole[67]. The PK/PD parameters following the intravenous administration of pantoprazole are summarized for different genotypes in Table 3. The 24-h median pH

Jo

was different among various genotypes after a single intravenous dose of pantoprazole, whereas the differences after repeated dosing were relatively small among the different CYP2C19 genotypes[66]. Several previous studies that evaluated whether oral or intravenous pantoprazole was a consistent and long-lasting inhibitor of gastric acid secretion have reported satisfactory results in patients with acid-related 17

diseases[86]. Numerous studies and meta-analyses have confirmed the efficacy of intravenous pantoprazole for the treatment of peptic ulcer bleeding and stress ulcer bleeding[87]. Thus, pantoprazole is now one of the most commonly administered

lP

re

-p

ro of

intravenous PPIs for the treatment of non-variceal gastrointestinal bleeding.

na

Figure 4. Metabolic pathways of pantoprazole. The thicker the arrow, the larger the contribution of the CYP isoforms to the metabolic pathway.

ur

Rabeprazole

Jo

Rabeprazole, a second-generation PPI, is also a substituted benzimidazole. It provides reliable control of gastric acid secretion, with more powerful antisecretory activity and rapid increases in intragastric pH than first-generation PPIs such as omeprazole and lansoprazole[31, 88, 89]. Rabeprazole has a better acid control and significantly longer duration of intragastric pH>4 in the 24 h post-dose than lansoprazole, omeprazole, or pantoprazole[90]. In addition, night-time pH 18

measurements made on the first dosing day have revealed a superior effect on intragastric pH than that achieved by standard doses of pantoprazole or omeprazole[90]. In our previous research, the AUC ratios of the low- (10 mg), middle- (20 mg), and high-dose (40 mg) groups were 1:1.9:3.6 and 1:1.8:3.4 respectively, after a single intravenous injection of rabeprazole (on Day 1) or after repeated dosing for 5 days[91]. In addition, the high-dose group had the longest pH>4

ro of

and pH>6 holding times, whereas the low-dose group had the shortest holding time. These results indicated that the PK/PD of rabeprazole is concentration-dependent,

with higher 24-h intragastric pH values obtained with higher AUC and Cmax values,

-p

which was similar to that for other PPIs[7, 92-94]. Rabeprazole has been reported to

re

be mainly metabolized (approximately 85%) via a non-enzymatic pathway to thioether-rabeprazole, with only minor involvement of CYP2C19 and CYP3A4[71,

lP

91]. The thioether-rabeprazole is then further stereoselectively re-oxidized, mainly to

na

(R)-rabeprazole. Therefore, the acid-suppressive effect of rabeprazole is considered to be less affected by CYP2C19 genotype-phenotype or genotype status compared with

ur

other PPIs (Figure 5). In healthy volunteers, the AUC ratio (PMs vs. homEMs) of rabeprazole (20 mg) was reported to be 3.3 after administration of a single oral dose,

Jo

whereas that of omeprazole (20 mg) was 10.7[24]. The AUC ratio (PMs vs. homEMs) was found to be 1.2 after approximately 2 months of oral administration of rabeprazole (20 mg) in patients with gastroesophageal reflux disease[54]. Shimatani et al.[95] reported that the potency of acid inhibition by rabeprazole was slightly influenced by CYP2C19 polymorphisms, whereas Hu et al.[71] demonstrated that the 19

PK properties of rabeprazole were partially dependent on the CYP2C19 genotype and that the acid-suppression efficacy was not significantly influenced by CYP2C19 polymorphisms. These results indicated that the differences in PK/PD parameters were relatively small among homEMs, hetEMs, and PMs after the administration of rabeprazole in healthy volunteers or patients with gastroesophageal reflux disease. In terms of PK parameters, rabeprazole is more applicable for patients with different

ro of

CYP2C19 genotypes. It would therefore be expected that patients treated with

rabeprazole show a more consistent healing efficacy than those receiving other PPIs. In particular, H. pylori eradication is also affected by CYP2C19 polymorphism in

-p

patients undergoing rabeprazole-based therapies (Table 2). The ratio of H. pylori

re

eradication rate (PMs vs. homEMs) was reported to be 1, 1, 0.9, 1.2 and 1.7 in patients receiving rabeprazole-based therapies[40, 44, 45, 48, 49], which was smaller

lP

than for omeprazole- (1.2, 1.6, 1.4 and 1.4)[39-42], lansoprazole- (1.2 and 1.2)[43, 44]

na

or pantoprazole- based therapies (1, 1.4 and 1.4)[45-47]. This may be because rabeprazole is mostly metabolized via a nonenzymatic process, with only minor

Jo

ur

participation of CYP2C19.

20

ro of -p re lP

Figure 5. Metabolic pathways of rabeprazole. The thicker the arrow, the larger the contribution of

Ilaprazole

na

the CYP isoforms to the metabolic pathway.

ur

Ilaprazole[88] (also called as IY81149), a newly marketed PPI, belongs to the class of substituted benzimidazole molecules, and is chemically related to

Jo

omeprazole[96, 97]. Ilaprazole can elicit a marked and dose-dependent gastric acid suppression effect, together with a longer plasma half-life compared with omeprazole[98]. Shin et al.[99] demonstrated that ilaprazole achieved better intragastric pH control than esomeprazole for the entire 24-h period, whereas esomeprazole provided better intragastric pH control during the initial 4 h. Although 21

most PPIs are mainly metabolized by CYP2C19, CYP3A may have a more critical role in the clearance of ilaprazole than CYP2C19 because ilaprazole sulfone (the major product) and hydroxyl ilaprazole (the minor product) are the two main circulating metabolites in plasma (Figure 6)[100, 101]. In addition, inhibition of CYP3A4 had a significant effect on ilaprazole concentration, whereas the inhibition of CYP2C19 had no significant effect, which also indicated that the metabolism of

ro of

ilaprazole in vivo was mainly related to CYP3A4[100]. In addition, there was no

significant difference in Css, max and AUC0-24 of ilaprazole or ilaprazole sulfone among

different CYP2C19 genotypes after daily oral administration of 10 mg ilaprazole for 7

-p

days[102]. The mean 24-h intragastric pH, TpH>4 (%), and the AUC0-24 of serum

re

gastrin were not significantly different among CYP2C19 genotypes. No serious adverse events were observed during the short-term use (5 days) of ilaprazole[103,

lP

104], however, research on this topic, especially on the long-term safety, remains

Jo

ur

na

insufficient.

22

ro of -p

the CYP isoforms to the metabolic pathway.

lP

DISCUSSION

re

Figure 6. Metabolic pathways of ilaprazole. The thicker the arrow, the larger the contribution of

na

Since the first PPI (omeprazole) was marketed, PPIs have gradually replaced histamine 2 receptor antagonists (H2RAs) for the treatment of gastroesophageal reflux

ur

disease and other acid-related disorders[88]. Several studies have demonstrated an association between polymorphisms of CYP2C19/CYP3A4 and inter-individual

Jo

variability in the disposition and pharmacologic responses of PPIs[105-107]. Among the main PPIs, the various influences of the CYP2C19 polymorphisms on metabolic parameters are different (omeprazole > lansoprazole > pantoprazole > rabeprazole > ilaprazole). The greatest impact was on omeprazole, which showed the largest differences in PK/PD properties and clinical efficacy among different CYP2C19 23

genotypes, whereas the S-isomer of omeprazole (esomeprazole) was less sensitive to CYP2C19 polymorphism, resulting in more stable plasma concentrations among the different CYP2C19 genotypes[108]. These data were in accordance with the observations made for lansopraozle. PPIs are unstable in an acidic environment; thus, oral dosage forms are usually formulated as enteric preparations to prevent their early conversion in stomach[109].

ro of

The absorption of PPIs after oral administration is affected by formulation type, diet, gastrointestinal physiology, and various other factors, which will influence the PK parameters of PPIs[110]. In contrast, intravenous preparations are less affected by

-p

these factors[7], and have relatively small individual differences in PK/PD values.

re

The approved dose of omeprazole in Japan is 20 mg or lower, but 40 mg in most western countries. Asian populations (e.g., Japanese and Chinese populations) have a

lP

higher proportion of PMs relative to Caucasian and African populations[12, 13], and

na

strong gastric acid-suppression can be obtained even with low doses of PPIs. As a non-enzymatic metabolized PPI, rabeprazole has smaller differences in PK/PD

ur

parameters and acid-suppression effect among the different CYP2C19 genotypes[31, 54]. However, few studies have investigated the effect of CYP2C19 polymorphism on

Jo

rabeprazole injection. Thus, it is difficult to compare the effect of CYP2C19 polymorphism on the PK/PD parameters of rabeprazole between oral and intravenous preparations. In addition, the disposition of PPIs is known to be time- and dose-dependent, with the PK parameters following repeated dosing different from those following a single dose[111]. In particular, single nucleotide polymorphisms 24

(SNPs) of the gastric H+,K+-ATPase α-subunit may be correlated with the configuration of the gastric H+,K+-ATPase, and may be related to the differences in individual gastric acid inhibition by PPIs[6]. However, few studies on this topic have been published and clinical studies are needed for confirmation. In contrast, even within the the same genotype, it is necessary to choose the appropriate PPI and dosage. Owing to the effect of the CYP2C19 genotype on H.

ro of

pylori eradication, it is important to identify and optimize an appropriate treatment

strategy, as the standard dosage may achieve poor eradication efficacy in homEMs or UMs[112]. Although a high dose of PPIs can slightly increase the therapeutic

-p

effect[113], selection of the correct drug appears to be a more reliable approach, as

re

double doses of rabeprazole or esomeprazole may increase the eradication rate of H. pylori by 8%–12% compared with first-generation PPIs[114]. This may be due to the

lP

small inter-individual variation of rabeprazole or esomeprazole metabolism[115]. In

na

particular, the metabolism of PPIs in vivo is not only dependent on genotype, but also closely related to the phenoconversion of CYP2C19 enzyme activity, which has a

ur

direct effect on the clinical efficacy of PPIs[116, 117]. Klieber et al.[116] found that the genotype-phenotype discordance increased by 28% after the administration of

Jo

omeprazole/esomeprazole (EM subjects converted to the PM phenotype), which is in accordance with the accumulation effect in PK parameters after repeated dosing of omeprazole observed in our previous research[16]. These results indicated that the identification of phenoconversion is also important for personalization of therapy after the administration of PPIs. Although few published studies evaluated the 25

phenoconversion of CYP2C19 after PPI administration, this may be an important direction for precision medicine. Based on these findings, genotype detection (gastric H+,K+-ATPase rs2733743 and CYP2C19) and phenoconversion identification could be helpful in determining the optimal dose of PPIs to avoid treatment failure and PPI-related side effects in patients This review has considered many studies of the effects of CYP2C19 and gastric

ro of

H+,K+-ATPase polymorphisms on the PK, PD, and clinical efficacy of PPIs,

encompassing both racemates and individual enantiomers. The main conclusions can

be summarized as follows: (1) Among the main PPIs, rabeprazole and ilaprazole have

-p

relatively small differences in PK/PD properties and clinical efficacy among different

re

CYP2C19 genotypes; (2) enantiomers may have less variation among different CYP2C19 genotypes than racemates after administration at the same dose; (3)

lP

compared with oral administration, intravenous doses have relatively smaller

na

differences among different CYP2C19 genotypes; and (4) gastric H+,K+-ATPase rs2733743 polymorphisms may have a greater effect than CYP2C19 polymorphisms

ur

on gastric acid-suppression efficiency (as has been confirmed with dexlansoprazole injection). It is necessary for physicians to personalize PPI regimens according to

Jo

each patient’s CYP2C19 and gastric H+,K+-ATPase genotype. Esomeprazole and rabeprazole are preferred in CYP2C19 homEMs and UMs, and the first dose should be doubled while follow-up treatment (regular dose) only needs to ensure an inhibitory effect on the subsequent activation of new pumps. A conventional dose of PPIs can achieve satisfactory inhibition in CYP2C19 hetEMs and PMs. A 26

dose-adjustment of regimens based on the gastric H+,K+-ATPase genotype may also increase the success rate of acid suppression therapy. However, research in this area is relatively rare and further clinical studies are required to determine the correct dose-adjustment protocol.

Confilct of Interest Statement

-p

conflict of interest in connection with the work submitted.

ro of

We declare that we have no commercial or associative interest that represents a

re

Acknowledgements This project was supported by the grants from the National

lP

Natural Sciences Foundation of China (81673515, 81870436, 81503160), Natural Science Foundation of Jiangsu Province (BK20161591), Six Talent Peaks Project in

na

Jiangsu Province (2014-YY-001), Jiangsu Provincial Medical Youth Talent (QNRC2016215), Suzhou science and education Youth Project (KJXW2016067),

ur

Suzhou industrial technology innovation (SYSD2016046), National key Research &

Jo

Development plan of Ministry of Science and Technology of the People’s Republic of China (2018YFC1314900, 2018YFC1314901), the 2016 industry prospecting and common key technology key projects of Jiangsu Province Science and Technology Department (BE2016002-4), the 2017 projects of Jiangsu Provincial Department of Finance (2150510), and the 2016 projects of Nanjing Science Bureau (201608003).

27

Reference [1] J.L. Birch, S.J. Newell, Gastrooesophageal reflux disease in preterm infants: current management and diagnostic dilemmas, Arch. Dis. Child. Fetal Neonatal Ed. 94 (2009) F379-383. [2] R. Badillo, D. Francis, Diagnosis and treatment of gastroesophageal reflux disease, World J. Gastrointest. Pharmacol. Ther. 5 (2014) 105-112. [3] K.W. Sylvester, J.W. Cheng, M.R. Mehra, Esomeprazole and aspirin fixed combination for the prevention of cardiovascular events, Vasc Health Risk Manag. 9 (2013) 245-254. [4] L.Y. Yu, L.N. Sun, X.H. Zhang, Y.Q. Li, L. Yu, Z.Q. Yuan, L. Meng, H.W. Zhang, Y.Q. Wang, A Review of the Novel Application and Potential Adverse Effects of Proton Pump Inhibitors, Adv. Ther. 34 (2017) 1070-1086. [5] L.S. Welage, Pharmacologic properties of proton pump inhibitors, Pharmacotherapy. 23 (2003) 74s-80s.

ro of

[6] H. Li, L. Meng, F. Liu, J.F. Wei, Y.Q. Wang, H+/K+-ATPase inhibitors: a patent review, Expert Opin. Ther. Pat. 23 (2013) 99-111.

[7] L.N. Sun, Y. Cao, Y.Q. Li, Y.Q. Fang, H.W. Zhang, M.F. Wang, L.J. Xie, J. Chen, Z.C. Yang, M.L. Bian, H. Li, P.P. Zhang, J.F. Wei, L. Meng, X.H. Zhang, P. Zhao, Y.Q. Wang, Impact of Gastric H(+)/K(+)-ATPase rs2733743 on the Intragastric pH-Values of Dexlansoprazole Injection in Chinese Subjects, Front.

-p

Pharmacol. 8 (2017) 670.

[8] Sapone, A., The clinical role of cytochrome p450 genotypes in Helicobacter pylori management, The American Journal of Gastroenterology. 98 (2003) 1010-1015.

re

[9] M. Robinson, J. Horn, Clinical pharmacology of proton pump inhibitors: what the practising physician needs to know, Drugs. 63 (2003) 2739-2754.

[10] T. Furuta, K. Ohashi, K. Kosuge, X.J. Zhao, M. Takashima, M. Kimura, M. Nishimoto, H. Hanai, E.

lP

Kaneko, T. Ishizaki, CYP2C19 genotype status and effect of omeprazole on intragastric pH in humans, Clin. Pharmacol. Ther. 65 (1999) 552-561.

[11] C.H. Kuo, C.Y. Lu, H.Y. Shih, C.J. Liu, M.C. Wu, H.M. Hu, W.H. Hsu, F.J. Yu, D.C. Wu, F.C. Kuo, CYP2C19 16029-16036.

na

polymorphism influences Helicobacter pylori eradication, World J. Gastroenterol. 20 (2014) [12] N. El Rouby, J.J. Lima, J.A. Johnson, Proton pump inhibitors: from CYP2C19 pharmacogenetics to precision medicine, Expert Opin. Drug Metab. Toxicol. 14 (2018) 447-460.

ur

[13] B.I. Drogemoller, G.E. Wright, D.J. Niehaus, L. Koen, S. Malan, D.M. Da Silva, R. Hillermann-Rebello, A.M. La Grange, M. Venter, L. Warnich, Characterization of the genetic profile of CYP2C19 in two South African populations, Pharmacogenomics. 11 (2010) 1095-1103.

Jo

[14] M. Roman, D. Ochoa, S.D. Sanchez-Rojas, M. Talegon, R. Prieto-Perez, A. Rivas, F. Abad-Santos, T. Cabaleiro, Evaluation of the relationship between polymorphisms in CYP2C19 and the pharmacokinetics of omeprazole, pantoprazole and rabeprazole, Pharmacogenomics. 15 (2014) 1893-1901.

[15] N.G. Hunfeld, R.A. Mathot, D.J. Touw, R.H. van Schaik, P.G. Mulder, P.F. Franck, E.J. Kuipers, W.P. Geus, Effect of CYP2C19*2 and *17 mutations on pharmacodynamics and kinetics of proton pump inhibitors in Caucasians, Br. J. Clin. Pharmacol. 65 (2008) 752-760. [16] Y. Wang, H. Zhang, L. Meng, M. Wang, H. Yuan, N. Ou, H. Zhang, Z. Li, R. Shi, Influence of CYP2C19 on the relationship between pharmacokinetics and intragastric pH of omeprazole administered by

28

successive intravenous infusions in Chinese healthy volunteers, Eur J Clin Pharmacol. 66 (2010) 563-569. [17] H.L. Qiao, Y.R. Hu, X. Tian, L.J. Jia, N. Gao, L.R. Zhang, Y.Z. Guo, Pharmacokinetics of three proton pump inhibitors in Chinese subjects in relation to the CYP2C19 genotype, Eur. J. Clin. Pharmacol. 62 (2006) 107-112. [18] S. Hayato, S. Hasegawa, S. Hojo, H. Okawa, H. Abe, N. Sugisaki, M. Munesue, Y. Horai, A. Ohnishi, Dose-response relationships of rabeprazole 5, 10, 20, and 40 mg once daily on suppression of gastric acid secretion through the night in healthy Japanese individuals with different CYP2C19 genotypes, Eur. J. Clin. Pharmacol. 68 (2012) 579-588. [19] H.R. Xu, W.L. Chen, X.N. Li, N.N. Chu, The effect of CYP2C19 activity on pharmacokinetics of lansoprazole and its active metabolites in healthy subjects, Pharm. Biol. 48 (2010) 947-952. [20] T. Furuta, N. Shirai, M. Kodaira, M. Sugimoto, A. Nogaki, S. Kuriyama, M. Iwaizumi, M. Yamade, I. Terakawa, K. Ohashi, T. Ishizaki, A. Hishida, Pharmacogenomics-based tailored versus standard

ro of

therapeutic regimen for eradication of H. pylori, Clin. Pharmacol. Ther. 81 (2007) 521-528.

[21] R.M. Ward, B. Tammara, S.E. Sullivan, D.L. Stewart, N. Rath, X. Meng, M.K. Maguire, G.M. Comer, Single-dose, multiple-dose, and population pharmacokinetics of pantoprazole in neonates and

preterm infants with a clinical diagnosis of gastroesophageal reflux disease (GERD), Eur. J. Clin. Pharmacol. 66 (2010) 555-561.

-p

[22] Z.C. Yang, F. Yu, Y.Q. Wang, J.F. Wei, Stereoselective Pharmacodynamics and Pharmacokinetics of Proton Pump Inhibitors, Curr Drug Metab. 17 (2016) 692-702. Pharmacokinet. 44 (2005) 441-466.

re

[23] C. Litalien, Y. Theoret, C. Faure, Pharmacokinetics of proton pump inhibitors in children, Clin. [24] N. Shirai, T. Furuta, Y. Moriyama, H. Okochi, K. Kobayashi, M. Takashima, F. Xiao, K. Kosuge, K. Nakagawa, H. Hanai, K. Chiba, K. Ohashi, T. Ishizaki, Effects of CYP2C19 genotypic differences in the

lP

metabolism of omeprazole and rabeprazole on intragastric pH, Aliment. Pharmacol. Ther. 15 (2001) 1929-1937.

[25] X.P. Hu, J.M. Xu, Y.M. Hu, Q. Mei, X.H. Xu, Effects of CYP2C19 genetic polymorphism on the (2007) 517-524.

na

pharmacokinetics and pharmacodynamics of omeprazole in Chinese people, J. Clin. Pharm. Ther. 32 [26] T. Uno, T. Niioka, M. Hayakari, N. Yasui-Furukori, K. Sugawara, T. Tateishi, Absolute bioavailability and metabolism of omeprazole in relation to CYP2C19 genotypes following single intravenous and oral

ur

administrations, Eur. J. Clin. Pharmacol. 63 (2007) 143-149. [27] M. Sagar, G. Tybring, M.L. Dahl, L. Bertilsson, R. Seensalu, Effects of omeprazole on intragastric pH and plasma gastrin are dependent on the CYP2C19 polymorphism, Gastroenterology. 119 (2000)

Jo

670-676.

[28] T. Shimatani, M. Inoue, T. Kuroiwa, Y. Horikawa, H. Mieno, M. Nakamura, Effect of omeprazole 10 mg on intragastric pH in three different CYP2C19 genotypes, compared with omeprazole 20 mg and lafutidine 20 mg, a new H2-receptor antagonist, Aliment. Pharmacol. Ther. 18 (2003) 1149-1157. [29] Y.H. Sohn, W.S. Lee, C.H. Park, Y.E. Joo, H.S. Kim, S.K. Choi, J.S. Rew, S.J. Kim, [Influence of CYP2C19 polymorphism and Helicobacter pylori status on the antisecretory effect of omeprazole in gastroesophageal reflux disease], Korean J Gastroenterol. 48 (2006) 162-171. [30] S. Sahara, M. Sugimoto, T. Uotani, H. Ichikawa, M. Yamade, M. Iwaizumi, T. Yamada, S. Osawa, K. Sugimoto, K. Umemura, H. Miyajima, T. Furuta, Twice-daily dosing of esomeprazole effectively inhibits

29

acid secretion in CYP2C19 rapid metabolisers compared with twice-daily omeprazole, rabeprazole or lansoprazole, Aliment. Pharmacol. Ther. 38 (2013) 1129-1137. [31] M. Sugimoto, N. Shirai, M. Nishino, C. Kodaira, T. Uotani, S. Sahara, H. Ichikawa, T. Kagami, K. Sugimoto, T. Furuta, Comparison of acid inhibition with standard dosages of proton pump inhibitors in relation to CYP2C19 genotype in Japanese, Eur. J. Clin. Pharmacol. 70 (2014) 1073-1078. [32] M. Sugimoto, T. Furuta, N. Shirai, M. Ikuma, A. Hishida, T. Ishizaki, Initial 48-hour acid inhibition by intravenous infusion of omeprazole, famotidine, or both in relation to cytochrome P450 2C19 genotype status, Clin. Pharmacol. Ther. 80 (2006) 539-548. [33] G. Sachs, J.M. Shin, O. Vagin, N. Lambrecht, I. Yakubov, K. Munson, The gastric H,K ATPase as a drug target: past, present, and future, J. Clin. Gastroenterol. 41 Suppl 2 (2007) S226-242. [34] S.W. Smith, Chiral toxicology: it's the same thing...only different, Toxicol. Sci. 110 (2009) 4-30. [35] Q. Shen, L. Wang, H. Zhou, H.D. Jiang, L.S. Yu, S. Zeng, Stereoselective binding of chiral drugs to plasma proteins, Acta Pharmacol. Sin. 34 (2013) 998-1006.

ro of

[36] H. Nagaya, N. Inatomi, A. Nohara, H. Satoh, Effects of the enantiomers of lansoprazole (AG-1749) on (H+ + K+)-ATPase activity in canine gastric microsomes and acid formation in isolated canine parietal cells, Biochem. Pharmacol. 42 (1991) 1875-1878.

[37] B.S. Sheu, A.W. Kao, H.C. Cheng, S.F. Hunag, T.W. Chen, C.C. Lu, J.J. Wu, Esomeprazole 40 mg twice daily in triple therapy and the efficacy of Helicobacter pylori eradication related to CYP2C19

-p

metabolism, Aliment. Pharmacol. Ther. 21 (2005) 283-288.

[38] T. Andersson, M. Hassan-Alin, G. Hasselgren, K. Rohss, L. Weidolf, Pharmacokinetic studies with esomeprazole, the (S)-isomer of omeprazole, Clin. Pharmacokinet. 40 (2001) 411-426.

re

[39] D.D. Nabinger, L.E. Mazzoleni, G.B. Sander, F. Mazzoleni, M.C. Osorio, M.G. Klein, T.F. Rech, L. Basso da Silva, G.S. de Moraes, R.A. Cristovam, E.F. Nardelli, C.F. Francesconi, D. Simon, Influence of CYP2C19 on Helicobacter pylori eradication in Brazilian patients with functional dyspepsia, Genet. Mol.

lP

Res. 15 (2016).

[40] Y.-A. Lin, H. Wang, Z.-J. Gu, W.-J. Wang, X.-Y. Zeng, Y.-L. Du, S.-S. Ying, B.-H. Zhang, Effect of CYP2C19 Gene Polymorphisms on Proton Pump Inhibitor, Amoxicillin, and Levofloxacin Triple Therapy for Eradication of Helicobacter Pylori, Med. Sci. Monit. 23 (2017) 2701-2707.

na

[41] O. Sezgin, I.O. Barlas, E. Ucbilek, E. Yengel, E. Altintas, Modified sequential Helicobacter pylori eradication therapy using high dose omeprazole and amoxicillin in the initial phase in the extensive metaboliser Turkish patients for CYP2C19 polymorphism is ineffective, Acta Gastroenterol. Belg. 77

ur

(2014) 3-7.

[42] J.C. Yang, H.L. Wang, H.D. Chern, C.T. Shun, B.R. Lin, C.J. Lin, T.H. Wang, Role of omeprazole dosage and cytochrome P450 2C19 genotype in patients receiving omeprazole-amoxicillin dual

Jo

therapy for Helicobacter pylori eradication, Pharmacotherapy. 31 (2011) 227-238. [43] M. Schwab, E. Schaeffeler, U. Klotz, G. Treiber, CYP2C19 polymorphism is a major predictor of treatment failure in white patients by use of lansoprazole-based quadruple therapy for eradication of Helicobacter pylori, Clin. Pharmacol. Ther. 76 (2004) 201-209. [44] J.H. Lee, H.Y. Jung, K.D. Choi, H.J. Song, G.H. Lee, J.H. Kim, The Influence of CYP2C19 Polymorphism on Eradication of Helicobacter pylori: A Prospective Randomized Study of Lansoprazole and Rabeprazole, Gut Liver. 4 (2010) 201-206. [45] A. Ormeci, Z. Emrence, B. Baran, S. Gokturk, O.M. Soyer, S. Evirgen, F. Akyuz, C. Karaca, F. Besisik, S. Kaymakoglu, D. Ustek, K. Demir, Effect of cytochrome P450 2C19 polymorphisms on the

30

Helicobacter pylori eradication rate following two-week triple therapy with pantoprazole or rabeprazole, Eur. Rev. Med. Pharmacol. Sci. 20 (2016) 879-885. [46] B. Gawronska-Szklarz, A. Siuda, M. Kurzawski, D. Bielicki, W. Marlicz, M. Drozdzik, Effects of CYP2C19, MDR1, and interleukin 1-B gene variants on the eradication rate of Helicobacter pylori infection by triple therapy with pantoprazole, amoxicillin, and metronidazole, Eur. J. Clin. Pharmacol. 66 (2010) 681-687. [47] J.H. Oh, M.S. Dong, M.G. Choi, H.W. Yoo, S.B. Lee, Y.I. Park, I.S. Chung, Effects of CYP2C19 and MDR1 genotype on the eradication rate of Helicobacter pylori infection by triple therapy with pantoprazole, amoxycillin and clarithromycin, J. Gastroenterol. Hepatol. 24 (2009) 294-298. [48] T.J. Lin, H.C. Lee, C.L. Lin, C.K. Wang, K.Y. Chen, D.C. Wu, CYP2C19 polymorphism has no influence on rabeprazole-based hybrid therapy for Helicobacter pylori eradication, World J Clin Cases. 6 (2018) 514-520. [49] X. Pan, Y. Li, Y. Qiu, Q. Tang, B. Qian, L. Yao, R. Shi, G. Zhang, Efficacy and tolerability of first-line

ro of

triple therapy with levofloxacin and amoxicillin plus esomeprazole or rabeprazole for the eradication

of Helicobacter pylori infection and the effect of CYP2C19 genotype: a 1-week, randomized, open-label study in Chinese adults, Clin. Ther. 32 (2010) 2003-2011.

[50] Y. Ishizawa, N. Yasui-Furukori, T. Takahata, M. Sasaki, T. Tateishi, The effect of aging on the relationship between the cytochrome P450 2C19 genotype and omeprazole pharmacokinetics, Clin.

-p

Pharmacokinet. 44 (2005) 1179-1189.

[51] O.Q. Yin, B. Tomlinson, A.H. Chow, M.M. Waye, M.S. Chow, Omeprazole as a CYP2C19 marker in Chinese subjects: assessment of its gene-dose effect and intrasubject variability, J. Clin. Pharmacol. 44

re

(2004) 582-589.

[52] G.L. Kearns, T. Andersson, L.P. James, A. Gaedigk, R.A. Kraynak, S.M. Abdel-Rahman, K. Ramabadran, J.N. van den Anker, Omeprazole Disposition in Children following Single-Dose

lP

Administration, The Journal of Clinical Pharmacology. 43 (2003) 840-848. [53] M. Chang, G. Tybring, M.L. Dahl, E. Gotharson, M. Sagar, R. Seensalu, L. Bertilsson, Interphenotype differences in disposition and effect on gastrin levels of omeprazole--suitability of omeprazole as a probe for CYP2C19, Br. J. Clin. Pharmacol. 39 (1995) 511-518.

na

[54] T. Sakai, N. Aoyama, T. Kita, T. Sakaeda, K. Nishiguchi, Y. Nishitora, T. Hohda, D. Sirasaka, T. Tamura, Y. Tanigawara, M. Kasuga, K. Okumura, CYP2C19 genotype and pharmacokinetics of three proton pump inhibitors in healthy subjects, Pharm. Res. 18 (2001) 721-727.

ur

[55] J.H. Tu, D.L. Hu, L.L. Dai, Y. Sun, L. Fan, M. Zhang, Z.R. Tan, Y. Chen, Z. Li, H.H. Zhou, Effect of glycyrrhizin on CYP2C19 and CYP3A4 activity in healthy volunteers with different CYP2C19 genotypes, Xenobiotica. 40 (2010) 393-399.

Jo

[56] S. Shilbayeh, M.F. Tutunji, Possible interethnic differences in omeprazole pharmacokinetics : comparison of Jordanian Arabs with other populations, Clin. Pharmacokinet. 45 (2006) 593-610. [57] D. Zhang, Y. Zhang, M. Liu, X. Wang, M. Yang, J. Han, H. Liu, Pharmacokinetics of lansoprazole and its main metabolites after single and multiple intravenous doses in healthy Chinese subjects, Eur. J. Drug Metab. Pharmacokinet. 38 (2013) 209-215. [58] T. Furuta, C. Kodaira, M. Nishino, M. Yamade, M. Sugimoto, M. Ikuma, A. Hishida, H. Watanabe, K. Umemura, [13C]-pantoprazole breath test to predict CYP2C19 phenotype and efficacy of a proton pump inhibitor, lansoprazole, Aliment. Pharmacol. Ther. 30 (2009) 294-300.

31

[59] M. Saito, N. Yasui-Furukori, T. Uno, T. Takahata, K. Sugawara, A. Munakata, T. Tateishi, Effects of clarithromycin on lansoprazole pharmacokinetics between CYP2C19 genotypes, Br. J. Clin. Pharmacol. 59 (2005) 302-309. [60] N. Yasui-Furukori, M. Saito, T. Uno, T. Takahata, K. Sugawara, T. Tateishi, Effects of fluvoxamine on lansoprazole pharmacokinetics in relation to CYP2C19 genotypes, J. Clin. Pharmacol. 44 (2004) 1223-1229. [61] I. Ieiri, Y. Kishimoto, H. Okochi, K. Momiyama, T. Morita, M. Kitano, T. Morisawa, Y. Fukushima, K. Nakagawa, J. Hasegawa, K. Otsubo, T. Ishizaki, Comparison of the kinetic disposition of and serum gastrin change by lansoprazole versus rabeprazole during an 8-day dosing scheme in relation to CYP2C19 polymorphism, Eur. J. Clin. Pharmacol. 57 (2001) 485-492. [62] D. Zhang, X. Wang, M. Yang, G. Wang, H. Liu, Effects of CYP2C19 polymorphism on the pharmacokinetics of lansoprazole and its main metabolites in healthy Chinese subjects, Xenobiotica. 41 (2011) 511-517.

ro of

[63] Y. Sakurai, M. Hirayama, M. Hashimoto, T. Tanaka, S. Hasegawa, S. Irie, K. Ashida, Y. Kayano, M. Taguchi, Y. Hashimoto, Population pharmacokinetics and proton pump inhibitory effects of intravenous lansoprazole in healthy Japanese males, Biol. Pharm. Bull. 30 (2007) 2238-2243.

[64] K. Adachi, T. Katsube, A. Kawamura, T. Takashima, M. Yuki, K. Amano, S. Ishihara, R. Fukuda, M. Watanabe, Y. Kinoshita, CYP2C19 genotype status and intragastric pH during dosing with lansoprazole

-p

or rabeprazole, Aliment. Pharmacol. Ther. 14 (2000) 1259-1266.

[65] M. Tanaka, T. Ohkubo, K. Otani, A. Suzuki, S. Kaneko, K. Sugawara, Y. Ryokawa, T. Ishizaki, Stereoselective pharmacokinetics of pantoprazole, a proton pump inhibitor, in extensive and poor

re

metabolizers of S-mephenytoin, Clin. Pharmacol. Ther. 69 (2001) 108-113.

[66] J.H. Oh, M.G. Choi, M.S. Dong, J.M. Park, C.N. Paik, Y.K. Cho, J.J. Jeong, I.S. Lee, S.W. Kim, S.W. Han, K.Y. Choi, I.S. Chung, Low-dose intravenous pantoprazole for optimal inhibition of gastric acid in Korean

lP

patients, J. Gastroenterol. Hepatol. 22 (2007) 1429-1434.

[67] B. Gawronska-Szklarz, U. Adamiak-Giera, E. Wyska, M. Kurzawski, W. Gornik, M. Kaldonska, M. Drozdzik, CYP2C19 polymorphism affects single-dose pharmacokinetics of oral pantoprazole in healthy volunteers, Eur. J. Clin. Pharmacol. 68 (2012) 1267-1274.

na

[68] N. Deshpande, S. V, V.R. V, V.V.M. H, S. M, R. Banerjee, M. Tandan, N.R. D, Rapid and ultra-rapid metabolizers with CYP2C19*17 polymorphism do not respond to standard therapy with proton pump inhibitors, Meta Gene. 9 (2016) 159-164.

ur

[69] Y. Horai, M. Kimura, H. Furuie, K. Matsuguma, S. Irie, Y. Koga, T. Nagahama, M. Murakami, T. Matsui, T. Yao, A. Urae, T. Ishizaki, Pharmacodynamic effects and kinetic disposition of rabeprazole in relation to CYP2C19 genotypes, Aliment. Pharmacol. Ther. 15 (2001) 793-803.

Jo

[70] M. Sugimoto, T. Furuta, N. Shirai, M. Kajimura, A. Hishida, M. Sakurai, K. Ohashi, T. Ishizaki, Different dosage regimens of rabeprazole for nocturnal gastric acid inhibition in relation to cytochrome P450 2C19 genotype status, Clin. Pharmacol. Ther. 76 (2004) 290-301. [71] Y.M. Hu, J.M. Xu, Q. Mei, X.H. Xu, S.Y. Xu, Pharmacodynamic effects and kinetic disposition of rabeprazole in relation to CYP2C19 genotype in healthy Chinese subjects, Acta Pharmacol. Sin. 26 (2005) 384-388. [72] M. Sugimoto, T. Furuta, N. Shirai, A. Nakamura, M. Kajimura, A. Hishida, K. Ohashi, T. Ishizaki, Comparison of an increased dosage regimen of rabeprazole versus a concomitant dosage regimen of famotidine with rabeprazole for nocturnal gastric acid inhibition in relation to cytochrome P450 2C19 genotypes, Clin. Pharmacol. Ther. 77 (2005) 302-311. 32

[73] Y.M. Hu, Q. Mei, X.H. Xu, X.P. Hu, N.Z. Hu, J.M. Xu, Pharmacodynamic and kinetic effect of rabeprazole on serum gastrin level in relation to CYP2C19 polymorphism in Chinese Hans, World J. Gastroenterol. 12 (2006) 4750-4753. [74] T. Niioka, T. Uno, N. Yasui-Furukori, M. Shimizu, K. Sugawara, T. Tateishi, Identification of the time-point which gives a plasma rabeprazole concentration that adequately reflects the area under the concentration-time curve, Eur. J. Clin. Pharmacol. 62 (2006) 855-861. [75] M. Shimizu, T. Uno, N. Yasui-Furukori, K. Sugawara, T. Tateishi, Effects of clarithromycin and verapamil on rabeprazole pharmacokinetics between CYP2C19 genotypes, Eur. J. Clin. Pharmacol. 62 (2006) 597-603. [76] T. Uno, M. Shimizu, N. Yasui-Furukori, K. Sugawara, T. Tateishi, Different effects of fluvoxamine on rabeprazole pharmacokinetics in relation to CYP2C19 genotype status, Br. J. Clin. Pharmacol. 61 (2006) 309-314. [77] Z.S. Li, X.B. Zhan, G.M. Xu, N.N. Cheng, Z. Liao, Effect of esomeprazole and rabeprazole on

ro of

intragastric pH in healthy Chinese: an open, randomized crossover trial, J. Gastroenterol. Hepatol. 22 (2007) 815-820.

[78] H.O. Yamano, H.O. Matsushita, S. Yanagiwara, Plasma concentration of rabeprazole after 8-week administration in gastroesophageal reflux disease patients and intragastric pH elevation, J. Gastroenterol. Hepatol. 23 (2008) 534-540.

-p

[79] Y.C. Sheng, K. Wang, Y.C. He, J. Yang, Q.S. Zheng, Effect of CYP2C19 genotypes on the

pharmacokinetic/pharmacodynamic relationship of rabeprazole after a single oral dose in healthy Chinese volunteers, Eur. J. Clin. Pharmacol. 66 (2010) 1165-1169.

re

[80] Y. Bian, M. Zhong, Investigation of the bioequivalence of two lansoprazole formulations in healthy Chinese volunteers after a single oral administration, Artif Cells Nanomed Biotechnol. 45 (2017) 1425-1430.

lP

[81] D. Zhang, M. Yang, M. Liu, Y. Zhang, X. Wang, X. Xiao, H. Liu, Pharmacokinetics of lansoprazole and its main metabolites after single intravenous doses in healthy Chinese subjects, Xenobiotica. 42 (2012) 1156-1162.

[82] K.M. Olsen, J.W. Devlin, Comparison of the enteral and intravenous lansoprazole

na

pharmacodynamic responses in critically ill patients, Aliment. Pharmacol. Ther. 28 (2008) 326-333. [83] T.M. Dando, G.L. Plosker, Intravenous lansoprazole: in erosive oesophagitis, Drugs. 64 (2004) 2085-2089; discussion 2090.

ur

[84] S. Shi, U. Klotz, Proton pump inhibitors: an update of their clinical use and pharmacokinetics, Eur. J. Clin. Pharmacol. 64 (2008) 935-951. [85] B.S. Sheu, H.C. Cheng, Y.C. Yeh, W.L. Chang, CYP2C19 genotypes determine the efficacy of

Jo

on-demand therapy of pantoprazole for reflux esophagitis as Los-Angeles grades C and D, J. Gastroenterol. Hepatol. 27 (2012) 104-109. [86] L. Moreira Dias, Pantoprazole: a proton pump inhibitor, Clin. Drug Investig. 29 Suppl 2 (2009) 3-12.

[87] T.O. Kovacs, D.M. Jensen, The short-term medical management of non-variceal upper gastrointestinal bleeding, Drugs. 68 (2008) 2105-2111. [88] E. Savarino, A. Ottonello, I. Martinucci, P. Dulbecco, V. Savarino, Ilaprazole for the treatment of gastro-esophageal reflux, Expert Opin. Pharmacother. 17 (2016) 2107-2113. [89] V. Savarino, F. Di Mario, C. Scarpignato, Proton pump inhibitors in GORD An overview of their pharmacology, efficacy and safety, Pharmacol. Res. 59 (2009) 135-153. 33

[90] D. Pantoflickova, G. Dorta, M. Ravic, P. Jornod, A.L. Blum, Acid inhibition on the first day of dosing: comparison of four proton pump inhibitors, Aliment. Pharmacol. Ther. 17 (2003) 1507-1514. [91] Y. Wang, Y. Yuan, L. Meng, H. Fan, J. Xu, H. Zhang, M. Wang, H. Yuan, N. Ou, H. Zhang, Y. Chao, R. Shi, Study of the pharmacokinetics and intragastric pH of rabeprazole given as successive intravenous infusion to healthy Chinese volunteers, Eur. J. Clin. Pharmacol. 67 (2011) 25-31. [92] T. Kita, T. Sakaeda, N. Aoyama, T. Sakai, Y. Kawahara, M. Kasuga, K. Okumura, Optimal dose of omeprazole for CYP2C19 extensive metabolizers in anti-Helicobacter pylori therapy: pharmacokinetic considerations, Biol. Pharm. Bull. 25 (2002) 923-927. [93] B.K. Tammara, J.E. Sullivan, K.G. Adcock, J. Kierkus, J. Giblin, N. Rath, X. Meng, M.K. Maguire, G.M. Comer, R.M. Ward, Randomized, open-label, multicentre pharmacokinetic studies of two dose levels of pantoprazole granules in infants and children aged 1 month through <6 years with gastro-oesophageal reflux disease, Clin. Pharmacokinet. 50 (2011) 541-550. [94] V. Savarino, G.S. Mela, P. Zentilin, G. Bisso, M. Pivari, S. Vigneri, R. Termini, S. Fiorucci, P. Usai, A.

ro of

Malesci, G. Celle, Comparison of 24-h control of gastric acidity by three different dosages of pantoprazole in patients with duodenal ulcer, Aliment. Pharmacol. Ther. 12 (1998) 1241-1247.

[95] T. Shimatani, M. Inoue, T. Kuroiwa, Y. Horikawa, Rabeprazole 10 mg twice daily is superior to 20 mg once daily for night-time gastric acid suppression, Aliment. Pharmacol. Ther. 19 (2004) 113-122.

[96] L. Wang, L. Zhou, S. Lin, H. Hu, J. Xia, A new PPI, ilaprazole compared with omeprazole in the

-p

treatment of duodenal ulcer: a randomized double-blind multicenter trial, J. Clin. Gastroenterol. 45 (2011) 322-329.

[97] H. Wang, N. Ou, L. Lang, R. Shi, P. Hu, J. Jiang, Pharmacokinetics and pharmacodynamics of

re

intravenous ilaprazole in healthy subjects after single ascending doses, Xenobiotica. 46 (2016) 1133-1141.

[98] N. de Bortoli, I. Martinucci, M. Giacchino, C. Blandizzi, S. Marchi, V. Savarino, E. Savarino, The Toxicol. 9 (2013) 1361-1369.

lP

pharmacokinetics of ilaprazole for gastro-esophageal reflux treatment, Expert Opin. Drug Metab. [99] J.S. Shin, J.Y. Lee, K.H. Cho, H.L. Park, M. Kukulka, J.T. Wu, D.Y. Kim, S.H. Park, The pharmacokinetics, pharmacodynamics and safety of oral doses of ilaprazole 10, 20 and 40 mg and esomeprazole 40 mg in

na

healthy subjects: a randomised, open-label crossover study, Aliment. Pharmacol. Ther. 40 (2014) 548-561.

[100] Y. Li, W. Zhang, D. Guo, G. Zhou, H. Zhou, Z. Xiao, Pharmacokinetics of the new proton pump

ur

inhibitor ilaprazole in Chinese healthy subjects in relation to CYP3A5 and CYP2C19 genotypes, Clin. Chim. Acta. 391 (2008) 60-67.

[101] J. Pu, F. Wang, W. Tang, M. Zhu, Biotransformation of Ilaprazole in Human Liver Microsomes and

Jo

Human: Role of CYP3A4 in Ilaprazole Clearance and Drug-Drug Interaction, Drug Metab. Dispos. 46 (2018) 1453-1461. [102] H. Cho, M.K. Choi, D.Y. Cho, C.W. Yeo, H.E. Jeong, J.H. Shon, J.Y. Lee, J.S. Shin, M. Cho, D.Y. Kim, J.G. Shin, Effect of CYP2C19 genetic polymorphism on pharmacokinetics and pharmacodynamics of a new proton pump inhibitor, ilaprazole, J. Clin. Pharmacol. 52 (2012) 976-984. [103] A.P. Periclou, R. Goldwater, S.M. Lee, D.W. Park, D.Y. Kim, K.D. Cho, F. Boileau, W.T. Jung, A comparative

pharmacodynamic

study

of

IY-81149

versus

omeprazole

gastroesophageal reflux disease, Clin. Pharmacol. Ther. 68 (2000) 304-311.

34

in

patients

with

[104] Y.Q. Du, W.Y. Guo, D.W. Zou, X.B. Zhan, Z. Li, J.H. Hu, Y.F. Gong, J. He, J.P. Lu, Z.S. Li, Acid inhibition effect of ilaprazole on Helicobacter pylori-negative healthy volunteers: an open randomized cross-over study, J. Dig. Dis. 13 (2012) 113-119. [105] H.C. Cheng, B.S. Sheu, Intravenous proton pump inhibitors for peptic ulcer bleeding: Clinical benefits and limits, World J Gastrointest Endosc. 3 (2011) 49-56. [106] R.M. Ward, G.L. Kearns, Proton pump inhibitors in pediatrics : mechanism of action, pharmacokinetics, pharmacogenetics, and pharmacodynamics, Paediatr. Drugs. 15 (2013) 119-131. [107] S. Sahara, M. Sugimoto, T. Uotani, H. Ichikawa, M. Yamade, T. Kagami, Y. Hamaya, M. Iwaizumi, S. Osawa, K. Sugimoto, H. Miyajima, T. Furuta, Potent Gastric Acid Inhibition Over 24 Hours by 4-Times Daily Dosing of Esomeprazole 20 mg, Digestion. 91 (2015) 277-285. [108] N.G. Hunfeld, D.J. Touw, R.A. Mathot, P.G. Mulder, V.A.N.S. RH, E.J. Kuipers, J.C. Kooiman, W.P. Geus, A comparison of the acid-inhibitory effects of esomeprazole and pantoprazole in relation to pharmacokinetics and CYP2C19 polymorphism, Aliment. Pharmacol. Ther. 31 (2010) 150-159.

ro of

[109] J.R. Horn, C.W. Howden, Review article: similarities and differences among delayed-release proton-pump inhibitor formulations, Aliment. Pharmacol. Ther. 22 Suppl 3 (2005) 20-24.

[110] B.T. Vanderhoff, R.M. Tahboub, Proton pump inhibitors: an update, Am. Fam. Physician. 66 (2002) 273-280.

[111] T. Andersson, K. Andren, C. Cederberg, P.O. Lagerstrom, P. Lundborg, I. Skanberg, healthy subjects, Br. J. Clin. Pharmacol. 29 (1990) 557-563.

-p

Pharmacokinetics and bioavailability of omeprazole after single and repeated oral administration in

[112] S.D. Georgopoulos, V. Papastergiou, S. Karatapanis, Treatment of Helicobacter Pylori infection:

re

optimization strategies in a high resistance era, Expert Opin. Pharmacother. 16 (2015) 2307-2317. [113] M. Vallve, M. Vergara, J.P. Gisbert, X. Calvet, Single vs. double dose of a proton pump inhibitor in triple therapy for Helicobacter pylori eradication: a meta-analysis, Aliment. Pharmacol. Ther. 16 (2002)

lP

1149-1156.

[114] P. Malfertheiner, F. Megraud, C.A. O'Morain, J. Atherton, A.T. Axon, F. Bazzoli, G.F. Gensini, J.P. Gisbert, D.Y. Graham, T. Rokkas, E.M. El-Omar, E.J. Kuipers, Management of Helicobacter pylori infection--the Maastricht IV/ Florence Consensus Report, Gut. 61 (2012) 646-664.

na

[115] V.W. Lee, T.S. Chau, A.K. Chan, K.K. Lee, M.M. Waye, T.K. Ling, F.K. Chan, Pharmacogenetics of esomeprazole or rabeprazole-based triple therapy in Helicobacter pylori eradication in Hong Kong non-ulcer dyspepsia Chinese subjects, J. Clin. Pharm. Ther. 35 (2010) 343-350.

ur

[116] M. Klieber, H. Oberacher, S. Hofstaetter, B. Beer, M. Neururer, A. Amann, H. Alber, A. Modak, CYP2C19 Phenoconversion by Routinely Prescribed Proton Pump Inhibitors Omeprazole and Esomeprazole: Clinical Implications for Personalized Medicine, J. Pharmacol. Exp. Ther. 354 (2015)

Jo

426-430.

[117] R.R. Shah, R.L. Smith, Addressing phenoconversion: the Achilles' heel of personalized medicine, Br. J. Clin. Pharmacol. 79 (2015) 222-240.

35