Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Clin Pharmacol Ther ; 115(6): 1428-1440, 2024 Jun.
Article En | MEDLINE | ID: mdl-38493369

In a genome-wide association study of atorvastatin pharmacokinetics in 158 healthy volunteers, the SLCO1B1 c.521T>C (rs4149056) variant associated with increased area under the plasma concentration-time curve from time zero to infinity (AUC0-∞) of atorvastatin (P = 1.2 × 10-10), 2-hydroxy atorvastatin (P = 4.0 × 10-8), and 4-hydroxy atorvastatin (P = 2.9 × 10-8). An intronic LPP variant, rs1975991, associated with reduced atorvastatin lactone AUC0-∞ (P = 3.8 × 10-8). Three UGT1A variants linked with UGT1A3*2 associated with increased 2-hydroxy atorvastatin lactone AUC0-∞ (P = 3.9 × 10-8). Furthermore, a candidate gene analysis including 243 participants suggested that increased function SLCO1B1 variants and decreased activity CYP3A4 variants affect atorvastatin pharmacokinetics. Compared with individuals with normal function SLCO1B1 genotype, atorvastatin AUC0-∞ was 145% (90% confidence interval: 98-203%; P = 5.6 × 10-11) larger in individuals with poor function, 24% (9-41%; P = 0.0053) larger in those with decreased function, and 41% (16-59%; P = 0.016) smaller in those with highly increased function SLCO1B1 genotype. Individuals with intermediate metabolizer CYP3A4 genotype (CYP3A4*2 or CYP3A4*22 heterozygotes) had 33% (14-55%; P = 0.022) larger atorvastatin AUC0-∞ than those with normal metabolizer genotype. UGT1A3*2 heterozygotes had 16% (5-25%; P = 0.017) smaller and LPP rs1975991 homozygotes had 34% (22-44%; P = 4.8 × 10-5) smaller atorvastatin AUC0-∞ than noncarriers. These data demonstrate that genetic variation in SLCO1B1, UGT1A3, LPP, and CYP3A4 affects atorvastatin pharmacokinetics. This is the first study to suggest that LPP rs1975991 may reduce atorvastatin exposure. [Correction added on 6 April, after first online publication: An incomplete sentence ("= 0.017) smaller in heterozygotes for UGT1A3*2 and 34% (22%, 44%; P × 10-5) smaller in homozygotes for LPP noncarriers.") has been corrected in this version.].


Area Under Curve , Atorvastatin , Cytochrome P-450 CYP3A , Genome-Wide Association Study , Glucuronosyltransferase , Liver-Specific Organic Anion Transporter 1 , Polymorphism, Single Nucleotide , Humans , Atorvastatin/pharmacokinetics , Atorvastatin/blood , Liver-Specific Organic Anion Transporter 1/genetics , Glucuronosyltransferase/genetics , Male , Female , Adult , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Young Adult , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacokinetics , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Middle Aged , Genotype , Healthy Volunteers , Pharmacogenomic Variants
2.
Br J Clin Pharmacol ; 90(2): 557-567, 2024 02.
Article En | MEDLINE | ID: mdl-37872104

AIMS: Ibrutinib is used in the treatment of certain B-cell malignancies. Due to its CYP3A4-mediated metabolism and highly variable pharmacokinetics, it is prone to potentially harmful drug-drug interactions. METHODS: In a randomized, placebo-controlled, three-phase crossover study, we examined the effect of the CYP3A4-inhibiting antifungal posaconazole on ibrutinib pharmacokinetics. Eleven healthy participants ingested repeated doses of 300 mg of posaconazole either in the morning or in the evening, or placebo. A single dose of ibrutinib (30, 70 or 140 mg, respectively) was administered at 9 AM, 1 or 12 h after the preceding posaconazole/placebo dose. RESULTS: On average, morning posaconazole increased the dose-adjusted geometric mean area under the plasma concentration-time curve from zero to infinity (AUC0-∞ ) and peak plasma concentration (Cmax ) of ibrutinib 9.5-fold (90% confidence interval [CI] 6.3-14.3, P < 0.001) and 8.5-fold (90% CI 5.7-12.8, P < 0.001), respectively, while evening posaconazole increased those 10.3-fold (90% CI 6.7-16.0, P < 0.001) and 8.2-fold (90% CI 5.2-13.2, P < 0.001), respectively. Posaconazole had no significant effect on the half-life of ibrutinib, but substantially reduced the metabolite PCI-45227 to ibrutinib AUC0-∞ ratio. There were no significant differences in ibrutinib pharmacokinetics between morning and evening posaconazole phases. CONCLUSIONS: Posaconazole increases ibrutinib exposure substantially, by about 10-fold. This interaction cannot be avoided by dosing the drugs 12 h apart. In general, a 70-mg daily dose of ibrutinib should not be exceeded during posaconazole treatment to avoid potentially toxic systemic ibrutinib concentrations.


Adenine/analogs & derivatives , Cytochrome P-450 CYP3A , Percutaneous Coronary Intervention , Piperidines , Triazoles , Humans , Cross-Over Studies , Area Under Curve
3.
Br J Clin Pharmacol ; 89(1): 242-252, 2023 01.
Article En | MEDLINE | ID: mdl-35942816

AIMS: The aim was to comprehensively investigate the effects of genetic variability on the pharmacokinetics of rosuvastatin. METHODS: We conducted a genome-wide association study and candidate gene analyses of single dose rosuvastatin pharmacokinetics in a prospective study (n = 159) and a cohort of previously published studies (n = 88). RESULTS: In a genome-wide association meta-analysis of the prospective study and the cohort of previously published studies, the SLCO1B1 c.521 T > C (rs4149056) single nucleotide variation (SNV) associated with increased area under the plasma concentration-time curve (AUC) and peak plasma concentration of rosuvastatin (P = 1.8 × 10-12 and P = 3.2 × 10-15 ). The candidate gene analysis suggested that the ABCG2 c.421C > A (rs2231142) SNV associates with increased rosuvastatin AUC (P = .0079), while the SLCO1B1 c.388A > G (rs2306283) and SLCO2B1 c.1457C > T (rs2306168) SNVs associate with decreased rosuvastatin AUC (P = .0041 and P = .0076). Based on SLCO1B1 genotypes, we stratified the participants into poor, decreased, normal, increased and highly increased organic anion transporting polypeptide (OATP) 1B1 function groups. The OATP1B1 poor function phenotype associated with 2.1-fold (90% confidence interval 1.6-2.8, P = 4.69 × 10-5 ) increased AUC of rosuvastatin, whereas the OATP1B1 highly increased function phenotype associated with a 44% (16-62%; P = .019) decreased rosuvastatin AUC. The ABCG2 c.421A/A genotype associated with 2.2-fold (1.5-3.0; P = 2.6 × 10-4 ) increased AUC of rosuvastatin. The SLCO2B1 c.1457C/T genotype associated with 28% decreased rosuvastatin AUC (11-42%; P = .01). CONCLUSION: These data suggest roles for SLCO1B1, ABCG2 and SLCO2B1 in rosuvastatin pharmacokinetics. Poor SLCO1B1 or ABCG2 function genotypes may increase the risk of rosuvastatin-induced myotoxicity. Reduced doses of rosuvastatin are advisable for patients with these genotypes.


Genome-Wide Association Study , Organic Anion Transporters , Rosuvastatin Calcium/pharmacokinetics , Pharmacogenomic Testing , Prospective Studies , Polymorphism, Single Nucleotide , Genotype , Organic Anion Transporters/genetics
4.
Clin Pharmacol Ther ; 112(3): 676-686, 2022 09.
Article En | MEDLINE | ID: mdl-35652242

We investigated genetic determinants of single-dose simvastatin pharmacokinetics in a prospective study of 170 subjects and a retrospective cohort of 59 healthy volunteers. In a microarray-based genomewide association study with the prospective data, the SLCO1B1 c.521T>C (p.Val174Ala, rs4149056) single nucleotide variation showed the strongest, genomewide significant association with the area under the plasma simvastatin acid concentration-time curve (AUC; P = 6.0 × 10-10 ). Meta-analysis with the retrospective cohort strengthened the association (P = 1.6 × 10-17 ). In a stepwise linear regression candidate gene analysis among all 229 participants, SLCO1B1 c.521T>C (P = 1.9 × 10-13 ) and CYP3A4 c.664T>C (p.Ser222Pro, rs55785340, CYP3A4*2, P = 0.023) were associated with increased simvastatin acid AUC. Moreover, the SLCO1B1 c.463C>A (p.Pro155Thr, rs11045819, P = 7.2 × 10-6 ) and c.1929A>C (p.Leu643Phe, rs34671512, P = 5.3 × 10-4 ) variants associated with decreased simvastatin acid AUC. Based on these results and the literature, we classified the volunteers into genotype-predicted OATP1B1 and CYP3A4 phenotype groups. Compared with the normal OATP1B1 function group, simvastatin acid AUC was 273% larger in the poor (90% confidence interval (CI), 137%, 488%; P = 3.1 × 10-6 ), 40% larger in the decreased (90% CI, 8%, 83%; P = 0.036), and 67% smaller in the highly increased function group (90% CI, 46%, 80%; P = 2.4 × 10-4 ). Intermediate CYP3A4 metabolizers (i.e., heterozygous carriers of either CYP3A4*2 or CYP3A4*22 (rs35599367)), had 87% (90% CI, 39%, 152%, P = 6.4 × 10-4 ) larger simvastatin acid AUC than normal metabolizers. These data suggest that in addition to no function SLCO1B1 variants, increased function SLCO1B1 variants and reduced function CYP3A4 variants may affect the pharmacokinetics, efficacy, and safety of simvastatin. Care is warranted if simvastatin is prescribed to patients carrying decreased function SLCO1B1 or CYP3A4 alleles.


Organic Anion Transporters , Simvastatin , Cytochrome P-450 CYP3A/genetics , Genotype , Humans , Liver-Specific Organic Anion Transporter 1/genetics , Organic Anion Transporters/genetics , Polymorphism, Single Nucleotide , Prospective Studies , Retrospective Studies , Simvastatin/pharmacokinetics
5.
Clin Transl Sci ; 15(2): 409-421, 2022 02.
Article En | MEDLINE | ID: mdl-34585840

The aim of this study was to search for associations of genetic variants with celiprolol pharmacokinetics in a large set of pharmacokinetic genes, and, more specifically, in a set of previously identified candidate genes ABCB1, SLCO1A2, and SLCO2B1. To this end, we determined celiprolol single-dose (200 mg) pharmacokinetics and sequenced 379 pharmacokinetic genes in 195 healthy volunteers. Analysis with 46,064 common sequence variants in the 379 genes did not identify any novel genes associated with celiprolol exposure. The candidate gene analysis showed that the ABCB1 c.3435T>C and c.2677T/G>A, and the SLCO1A2 c.516A>C variants were associated with reduced celiprolol area under the plasma concentration-time curve (AUC0-∞ ). An alternative analysis with ABCB1 haplotypes showed that, in addition to SLCO1A2 c.516A>C, three ABCB1 haplotypes were associated with reduced celiprolol AUC0-∞ . A genotype scoring system was developed based on these variants and applied to stratify the participants to low and high celiprolol exposure genotype groups. The mean AUC0-∞ of celiprolol in the low exposure genotype group was 55% of the mean AUC0-∞ in the high exposure group (p = 1.08 × 10-11 ). In addition, the results showed gene-gene interactions in the effects of SLCO1A2 and ABCB1 variants on celiprolol AUC0-∞ (p < 5 × 10-6 ) suggesting an interplay between organic anion transporting polypeptide 1A2 and P-glycoprotein in celiprolol absorption. Taken together, these data indicate that P-glycoprotein and organic anion transporting polypeptide 1A2 play a role in celiprolol pharmacokinetics. Furthermore, patients with ABCB1 and SLCO1A2 genotypes associated with low celiprolol exposure may have an increased risk of poor blood-pressure lowering response to celiprolol.


Celiprolol , Organic Anion Transporters , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Celiprolol/pharmacokinetics , Genotype , Humans , Organic Anion Transporters/metabolism , Pharmacogenetics , Polymorphism, Single Nucleotide
6.
Clin Pharmacol Ther ; 108(4): 885-895, 2020 10.
Article En | MEDLINE | ID: mdl-32498119

To investigate how variability in multiple pharmacokinetic genes associates with telmisartan exposure, we determined telmisartan single-dose (40 mg) pharmacokinetics and sequenced 379 genes in 188 healthy volunteers. Intronic UGT1A variants showed the strongest associations with the area under the plasma concentration-time curve from zero hours to infinity (AUC0-∞ ) and peak plasma concentration (Cmax ) of telmisartan. These variants were strongly linked with the increased function UGT1A3*2 allele, suggesting that it is the causative allele underlying these associations. In addition, telmisartan plasma concentrations were lower in men than in women. The UGT1A3*2 was associated with a 64% and 63% reduced AUC0-∞ of telmisartan in UGT1A3*2 heterozygous and homozygous men, respectively (P = 1.21 × 10-16 and 5.21 × 10-8 ). In women, UGT1A3*2 heterozygosity and homozygosity were associated with 57% (P = 1.54 × 10-11 ) and 72% (P = 3.31 × 10-15 ) reduced AUC0-∞ , respectively. Furthermore, a candidate gene analysis suggested an association of UGT1A3*3 and the SLCO1B3 c.767G>C missense variant with telmisartan pharmacokinetics. A genotype score, which reflects the effects of sex and genetic variants on telmisartan AUC0-∞ , associated with the effect of telmisartan on diastolic blood pressure. These data indicate that sex and UGT1A3 are major determinants and suggest a role for OATP1B3 in telmisartan pharmacokinetics.


Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Blood Pressure/drug effects , Glucuronosyltransferase/genetics , Pharmacogenomic Variants , Polymorphism, Single Nucleotide , Telmisartan/pharmacokinetics , Adult , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin II Type 1 Receptor Blockers/blood , Female , Glucuronosyltransferase/metabolism , Healthy Volunteers , Humans , Male , Mutation, Missense , Pharmacogenetics , Sex Factors , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Telmisartan/administration & dosage , Telmisartan/blood , Young Adult
7.
Clin Pharmacol Ther ; 108(6): 1254-1264, 2020 12.
Article En | MEDLINE | ID: mdl-32558923

In vitro, esomeprazole is a time-dependent inhibitor of CYP2C19. Additionally, racemic omeprazole induces CYP1A2 and omeprazole and its metabolites inhibit CYP3A4 in vitro. In this 5-phase study, 10 healthy volunteers ingested 20 mg pantoprazole, 0.5 mg midazolam, and 50 mg caffeine as respective index substrates for CYP2C19, 3A4, and 1A2 before and 1, 25, 49 (pantoprazole only), and 73 hours after an 8-day pretreatment with 80 mg esomeprazole twice daily. The area under the plasma concentration-time curve (AUC) of R-pantoprazole increased 4.92-fold (90% confidence interval (CI) 3.55-6.82), 2.31-fold (90% CI 1.85-2.88), and 1.33-fold (90% CI 1.06-1.68) at the 1-hour, 25-hour, and 73-hour phases, respectively, consistent with a substantial and persistent inhibition of CYP2C19. The AUC of midazolam increased up to 1.44-fold (90% CI 1.22-1.72) and the paraxanthine/caffeine metabolic ratio up to 1.19-fold (90% CI 1.04-1.36), when the index substrates were taken 1 hour after esomeprazole. Based on the recovery of R-pantoprazole oral clearance, the turnover half-life of CYP2C19 was estimated to average 53 hours. Pharmacokinetic simulation based on the observed concentrations of esomeprazole and its metabolites as well as their published CYP2C19 inhibitory constants was well in line with the observed changes in R-pantoprazole pharmacokinetics during the course of the study. Extrapolations assuming linear pharmacokinetics of esomeprazole suggested weak to moderate inhibition at 20 and 40 mg twice daily dosing. In conclusion, high-dose esomeprazole can cause strong inhibition of CYP2C19, but only weakly inhibits CYP3A4 and leads to minor induction of CYP1A2. The enzymatic activity of CYP2C19 recovers gradually in ~ 3-4 days after discontinuation of esomeprazole treatment.


Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2C19 Inhibitors/pharmacology , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP3A/metabolism , Esomeprazole/pharmacology , Administration, Oral , Caffeine/pharmacokinetics , Cross-Over Studies , Cytochrome P-450 CYP1A2 Inducers/pharmacology , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C19 Inhibitors/administration & dosage , Cytochrome P-450 CYP2C19 Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Esomeprazole/administration & dosage , Esomeprazole/pharmacokinetics , Female , Healthy Volunteers , Humans , Male , Midazolam/pharmacokinetics , Models, Biological , Pantoprazole/pharmacokinetics , Pharmacogenomic Variants
8.
Clin Transl Sci ; 13(2): 345-351, 2020 03.
Article En | MEDLINE | ID: mdl-31664782

The oral bioavailability of ibrutinib is low and variable, mainly due to extensive first-pass metabolism by cytochrome P450 (CYP) 3A4. The unpredictable exposure can compromise its safe and effective dosing. We examined the impact of itraconazole on ibrutinib pharmacokinetics. In a randomized crossover study, 11 healthy subjects were administered itraconazole 200 mg or placebo twice on day 1, and once on days 2-4. On day 3, 1 hour after itraconazole (placebo) and breakfast, ibrutinib (140 mg during placebo; 15 mg during itraconazole) was administered. Itraconazole increased the dose-adjusted geometric mean area under the concentration-time curve from zero to infinity (AUC0-∞ ) of ibrutinib 10.0-fold (90% confidence interval (CI) 7.2-13.9; P < 0.001) and peak plasma concentration (Cmax ) 8.8-fold (90% CI 6.3-12.1; P < 0.001). During itraconazole, the intersubject variation for the AUC0-∞ (55%) and Cmax (53%) was around half of that during placebo (104%; 99%). In conclusion, itraconazole markedly increases ibrutinib bioavailability and decreases its interindividual variability, offering a possibility to improved dosing accuracy and cost savings.


Adenine/analogs & derivatives , Biological Variation, Population/drug effects , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Itraconazole/pharmacokinetics , Piperidines/pharmacokinetics , Adenine/administration & dosage , Adenine/pharmacokinetics , Administration, Oral , Adult , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Area Under Curve , Biological Availability , Cross-Over Studies , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Drug Interactions , Healthy Volunteers , Humans , Itraconazole/administration & dosage , Male , Piperidines/administration & dosage , Young Adult
9.
Clin Pharmacol Ther ; 106(3): 668-680, 2019 09.
Article En | MEDLINE | ID: mdl-30989645

The aim of this study was to investigate how variability in multiple genes related to pharmacokinetics affects fluvastatin exposure. We determined fluvastatin enantiomer pharmacokinetics and sequenced 379 pharmacokinetic genes in 200 healthy volunteers. CYP2C9*3 associated with significantly increased area under the plasma concentration-time curve (AUC) of both 3R,5S-fluvastatin and 3S,5R-fluvastatin (by 67% and 94% per variant allele copy, P = 3.77 × 10-9 and P = 3.19 × 10-12 ). In contrast, SLCO1B1 c.521T>C associated with increased AUC of active 3R,5S-fluvastatin only (by 34% per variant allele copy; P = 8.15 × 10-8 ). A candidate gene analysis suggested that CYP2C9*2 also affects the AUC of both fluvastatin enantiomers and that SLCO2B1 single-nucleotide variations may affect the AUC of 3S,5R-fluvastatin. Thus, SLCO transporters have enantiospecific effects on fluvastatin pharmacokinetics in humans. Genotyping of both CYP2C9 and SLCO1B1 may be useful in predicting fluvastatin efficacy and myotoxicity.


Anticholesteremic Agents/chemistry , Anticholesteremic Agents/pharmacokinetics , Cytochrome P-450 CYP2C9/genetics , Fluvastatin/chemistry , Fluvastatin/pharmacokinetics , Liver-Specific Organic Anion Transporter 1/genetics , Area Under Curve , Half-Life , Humans , Pharmacogenetics , Polymorphism, Single Nucleotide
10.
Clin Pharmacol Ther ; 104(1): 158-168, 2018 07.
Article En | MEDLINE | ID: mdl-28940478

To identify the genetic basis of interindividual variability in montelukast exposure, we determined its pharmacokinetics and sequenced 379 pharmacokinetic genes in 191 healthy volunteers. An intronic single nucleotide variation (SNV), strongly linked with UGT1A3*2, associated with reduced area under the plasma concentration-time curve (AUC0-∞ ) of montelukast (by 18% per copy of the minor allele; P = 1.83 × 10-10 ). UGT1A3*2 was associated with increased AUC0-∞ of montelukast acyl-glucuronide M1 and decreased AUC0-∞ of hydroxymetabolites M5R, M5S, and M6 (P < 10-9 ). Furthermore, SNVs in SLCO1B1 and ABCC9 were associated with the AUC0-∞ of M1 and M5R, respectively. In addition, a candidate gene analysis suggested that CYP2C8 and ABCC9 SNVs also affect the AUC0-∞ of montelukast. The found UGT1A3 and ABCC9 variants associated with increased expression of the respective genes in human liver samples. Montelukast and its hydroxymetabolites were glucuronidated by UGT1A3 in vitro. These results indicate that UGT1A3 plays an important role in montelukast pharmacokinetics, especially in UGT1A3*2 carriers.


Acetates/pharmacokinetics , Cytochrome P-450 CYP1A2 Inducers/pharmacokinetics , Glucuronosyltransferase/genetics , Quinolines/pharmacokinetics , Acetates/metabolism , Adult , Area Under Curve , Cyclopropanes , Cytochrome P-450 CYP1A2 Inducers/metabolism , Cytochrome P-450 CYP2C8/genetics , Cytochrome P-450 CYP2C8/metabolism , Female , Glucuronosyltransferase/metabolism , Humans , In Vitro Techniques , Liver-Specific Organic Anion Transporter 1/genetics , Liver-Specific Organic Anion Transporter 1/metabolism , Male , Pharmacogenomic Testing , Polymorphism, Single Nucleotide , Quinolines/metabolism , Sulfides , Sulfonylurea Receptors/genetics , Sulfonylurea Receptors/metabolism , Young Adult
11.
Basic Clin Pharmacol Toxicol ; 122(3): 341-345, 2018 Mar.
Article En | MEDLINE | ID: mdl-28990360

Several single nucleotide variations (SNVs) affect carboxylesterase 1 (CES1) activity, but the effects of genetic variants on CES1 gene expression have not been systematically investigated. Therefore, our aim was to investigate effects of genetic variants on CES1 gene expression in two independent whole blood sample cohorts of 192 (discovery) and 88 (replication) healthy volunteers and in a liver sample cohort of 177 patients. Furthermore, we investigated possible effects of the found variants on clopidogrel pharmacokinetics (n = 106) and pharmacodynamics (n = 46) in healthy volunteers, who had ingested a single 300 mg or 600 mg dose of clopidogrel. Using massively parallel sequencing, we discovered two CES1 SNVs, rs12443580 and rs8192935, to be strongly and independently associated with a 39% (p = 4.0 × 10-13 ) and 31% (p = 2.5 × 10-8 ) reduction in CES1 whole blood expression per copy of the minor allele. These findings were replicated in the replication cohort. However, these SNVs did not affect CES1 liver expression, or clopidogrel pharmacokinetics or pharmacodynamics. Conversely, the CES1 c.428G>A missense SNV (rs71647871) impaired the hydrolysis of clopidogrel, increased exposure to clopidogrel active metabolite and enhanced its antiplatelet effects. In conclusion, the rs12443580 and rs8192935 variants reduce CES1 expression in whole blood but not in the liver. These tissue-specific effects may result in substrate-dependent effects of the two SNVs on CES1-mediated drug metabolism.


Carboxylic Ester Hydrolases/genetics , Gene Expression Regulation, Enzymologic , Pharmacogenomic Variants , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Aggregation/drug effects , Polymorphism, Single Nucleotide , Ticlopidine/analogs & derivatives , Biopsy , Carboxylic Ester Hydrolases/blood , Carboxylic Ester Hydrolases/chemistry , Carboxylic Ester Hydrolases/metabolism , Clopidogrel , Cohort Studies , DNA Mutational Analysis , Dose-Response Relationship, Drug , Female , Finland , Gastric Bypass , Humans , Hydrolysis , Introns , Liver/enzymology , Liver/metabolism , Liver/pathology , Male , Mutation, Missense , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacology , RNA, Messenger/metabolism , Reproducibility of Results , Ticlopidine/administration & dosage , Ticlopidine/blood , Ticlopidine/pharmacokinetics , Ticlopidine/pharmacology
12.
Pharmacogenet Genomics ; 23(1): 19-24, 2013 Jan.
Article En | MEDLINE | ID: mdl-23151832

OBJECTIVE: A nonsynonymous single nucleotide polymorphism (SNP) in the SLCO2B1 gene encoding organic anion transporting polypeptide 2B1 (OATP2B1), c.935G>A (p.R312Q; rs12422149), has been associated with reduced plasma concentrations of montelukast in patients with asthma. Our aim was to examine the possible effects of the SLCO2B1 c.935G>A SNP on the single-dose pharmacokinetics of the suggested OATP2B1 substrates montelukast and aliskiren. METHODS: Sixteen healthy volunteers with the SLCO2B1 c.935GG genotype, 12 with the c.935GA genotype, and five with the c.935AA genotype ingested a single 10 mg dose of montelukast or a 150 mg dose of aliskiren, with a washout period of 1 week. Plasma montelukast concentrations were measured up to 24 h. Plasma and urine aliskiren concentrations were measured up to 72 and 12 h, respectively, and plasma renin activity up to 24 h after aliskiren intake. RESULTS: The SLCO2B1 genotypes had no significant effect on the pharmacokinetics of montelukast or aliskiren. The geometric mean ratios with 90% confidence intervals of montelukast area under the plasma concentration-time curve from 0 h to infinity (AUC(0-∞)) in participants with the c.935GA or the c.935AA genotype to those with the c.935GG genotype were 1.02 (0.87, 1.21) or 0.88 (0.71, 1.10), respectively (P=0.557). The geometric mean ratios (90% confidence interval) of aliskiren AUC(0-∞) in participants with the c.935GA or the c.935AA genotype to those with the c.935GG genotype were 0.98 (0.74, 1.30) or 1.24 (0.85, 1.80), respectively (P=0.576). CONCLUSION: These data do not support the suggested functional significance of the SLCO2B1 c.935G>A SNP on OATP2B1 activity in vivo.


Acetates/pharmacokinetics , Amides/pharmacokinetics , Fumarates/pharmacokinetics , Organic Anion Transporters/genetics , Polymorphism, Single Nucleotide/genetics , Quinolines/pharmacokinetics , Acetates/pharmacology , Adult , Amides/pharmacology , Antihypertensive Agents/pharmacokinetics , Antihypertensive Agents/pharmacology , Chromatography, Liquid , Cross-Over Studies , Cyclopropanes , Female , Fumarates/pharmacology , Genotype , Humans , Leukotriene Antagonists/pharmacokinetics , Leukotriene Antagonists/pharmacology , Male , Middle Aged , Quinolines/pharmacology , Radioimmunoassay , Renin/antagonists & inhibitors , Renin/metabolism , Sulfides , Tandem Mass Spectrometry , Tissue Distribution , Young Adult
13.
Br J Clin Pharmacol ; 71(5): 718-26, 2011 May.
Article En | MEDLINE | ID: mdl-21204914

AIM: The aim of this study was to investigate the effects of orange juice and apple juice on the pharmacokinetics and pharmacodynamics of aliskiren. METHODS: In a randomized crossover study, 12 healthy volunteers ingested 200 ml of orange juice, apple juice or water three times daily for 5 days. On day 3, they ingested a single 150-mg dose of aliskiren. Plasma aliskiren concentrations were measured up to 72 h, its excretion into urine up to 12 h and plasma renin activity up to 24 h. RESULTS: Orange and apple juice reduced aliskiren peak plasma concentrations by 80% (95% CI 63%, 89%, P < 0.001) and 84% (95% CI 72%, 91%, P < 0.001), and the area under the plasma aliskiren concentration-time curve (AUC) by 62% (95% CI 47%, 72%, P < 0.001) and 63% (95% CI 46%, 74%, P < 0.001), respectively, but had no significant effect on its elimination half-life or renal clearance. The decreases in aliskiren AUC by orange and apple juice correlated with aliskiren AUC during the water phase (r= 0.98, P < 0.001). Plasma renin activity was 87% and 67% higher at 24 h after aliskiren during the orange juice and apple juice phases, respectively, than during the water phase (P < 0.05). CONCLUSIONS: Orange juice and apple juice greatly reduce the plasma concentrations and renin-inhibiting effect of aliskiren, probably by inhibiting its OATP2B1-mediated influx in the small intestine. Concomitant intake of aliskiren with orange or apple juice is best avoided.


Amides/blood , Antihypertensive Agents/blood , Beverages , Citrus sinensis , Food-Drug Interactions , Fumarates/blood , Malus , Adult , Female , Humans , Male , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/physiology , Renin/blood , Young Adult
14.
J Clin Pharmacol ; 51(3): 359-67, 2011 Mar.
Article En | MEDLINE | ID: mdl-20400651

In a randomized crossover study, 11 healthy volunteers took 100 mg (first dose 200 mg) of the antifungal drug itraconazole, a P-glycoprotein and CYP3A4 inhibitor, or placebo twice daily for 5 days. On day 3, they ingested a single 150-mg dose of aliskiren, a renin inhibitor used in the treatment of hypertension. Itraconazole raised the peak plasma aliskiren concentration 5.8-fold (range, 1.1- to 24.3-fold; P < .001) and the area under the plasma aliskiren concentration-time curve 6.5-fold (range, 2.6- to 20.5-fold; P < .001) but had no significant effect on aliskiren elimination half-life. Itraconazole increased the amount of aliskiren excreted into the urine during 12 hours 8.0-fold (P < .001) and its renal clearance 1.2-fold (P = .042). Plasma renin activity 24 hours after aliskiren intake was 68% lower during the itraconazole phase than during the placebo phase (P = .011). In conclusion, itraconazole markedly raises the plasma concentrations and enhances the renin-inhibiting effect of aliskiren. The interaction is probably mainly explained by inhibition of the P-glycoprotein-mediated efflux of aliskiren in the small intestine, with a minor contribution from inhibition of CYP3A4. Concomitant use of aliskiren and itraconazole is best avoided.


ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Amides/pharmacokinetics , Antifungal Agents/pharmacology , Antihypertensive Agents/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors , Fumarates/pharmacokinetics , Itraconazole/pharmacology , Renin/antagonists & inhibitors , Adult , Amides/blood , Amides/pharmacology , Amides/urine , Antihypertensive Agents/blood , Antihypertensive Agents/pharmacology , Antihypertensive Agents/urine , Biotransformation/drug effects , Cross-Over Studies , Cytochrome P-450 CYP3A , Drug Interactions , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/urine , Female , Fumarates/blood , Fumarates/pharmacology , Fumarates/urine , Half-Life , Humans , Itraconazole/analogs & derivatives , Itraconazole/blood , Male , Metabolic Clearance Rate , Renin/blood , Young Adult
15.
Eur J Clin Pharmacol ; 66(9): 865-70, 2010 Sep.
Article En | MEDLINE | ID: mdl-20496145

PURPOSE: This study aimed to investigate the possible effects of ABCB1 haplotypes on the pharmacokinetics and renin-inhibiting effect of aliskiren. METHODS: Eleven healthy volunteers homozygous for the ABCB1 c.1236C-c.2677G-c.3435C (CGC) haplotype and 11 homozygous for the c.1236T-c.2677T-c.3435T (TTT) haplotype ingested a single 150-mg dose of aliskiren. Plasma aliskiren concentrations were measured up to 72 h, its excretion into urine up to 12 h, and plasma renin activity up to 24 h. RESULTS: The ABCB1 haplotypes had no significant effect on the pharmacokinetics or renin-inhibiting effect of aliskiren. The geometric mean ratio (95% confidence interval) of aliskiren peak plasma concentration and area under the plasma concentration-time curve from 0 h to infinity in participants homozygous for the TTT haplotype to those in participants homozygous for the CGC haplotype were 1.14 (0.66, 1.96; P = 0.631) and 1.01 (0.58, 1.76; P = 0.960) respectively. CONCLUSIONS: These data suggest that the ABCB1 c.1236C-c.2677G-c.3435C and c.1236T-c.2677T-c.3435T haplotypes have no clinically meaningful effect on the pharmacokinetics of aliskiren.


ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Amides/pharmacology , Antihypertensive Agents/pharmacology , Fumarates/pharmacology , Renin/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Amides/blood , Amides/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Area Under Curve , Female , Fumarates/blood , Fumarates/pharmacokinetics , Haplotypes , Humans , Male
16.
Eur J Clin Pharmacol ; 66(5): 497-502, 2010 May.
Article En | MEDLINE | ID: mdl-20179914

PURPOSE: This study aimed to investigate the effect of rifampicin, an inducer of CYP3A4 and P-glycoprotein, on the pharmacokinetics and pharmacodynamics of aliskiren, a renin inhibitor used in the treatment of hypertension. METHODS: In a randomized crossover study, 12 healthy volunteers took 600 mg rifampicin or placebo once daily for 5 days. On day 6, they ingested a single 150-mg dose of aliskiren. Plasma aliskiren concentrations were measured up to 72 h and urine concentrations up to 12 h; pharmacodynamic variables were measured up to 24 h. RESULTS: Rifampicin reduced the peak plasma aliskiren concentration (C(max)) by 39% (95% confidence interval 0.41, 0.90; P = 0.017) and the area under the plasma aliskiren concentration-time curve AUC(0-infinity) by 56% (95% confidence interval 0.35, 0.56; P < 0.001). Rifampicin had no significant effect on aliskiren elimination half-life (t(1/2)) or its renal clearance (Cl(renal)). Plasma renin activity 24 h after aliskiren intake was 61% higher during the rifampicin phase than during the placebo phase (P = 0.008). CONCLUSIONS: Rifampicin considerably reduces the plasma concentrations and the renin-inhibiting effect of aliskiren by decreasing its oral bioavailability.


Amides/blood , Amides/pharmacology , Antihypertensive Agents/blood , Antihypertensive Agents/pharmacology , Enzyme Inhibitors/adverse effects , Fumarates/blood , Fumarates/pharmacology , Renin/antagonists & inhibitors , Rifampin/adverse effects , Adult , Amides/urine , Antihypertensive Agents/urine , Blood Pressure/drug effects , Cross-Over Studies , Drug Interactions , Female , Fumarates/urine , Heart Rate/drug effects , Humans , Male , Renin/blood
...