Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Eur J Drug Metab Pharmacokinet ; 47(1): 91-103, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34716565

ABSTRACT

BACKGROUND AND OBJECTIVE: Increased glycine availability at the synaptic cleft may enhance N-methyl-D-aspartate receptor signalling and provide a promising therapeutic strategy for cognitive impairment associated with schizophrenia. These studies aimed to assess the pharmacokinetics of BI 425809, a potent glycine-transporter-1 inhibitor, when co-administered with a strong cytochrome P450 3A4 (CYP3A4) inhibitor (itraconazole) and inducer (rifampicin). METHODS: In vitro studies using recombinant CYPs, human liver microsomes, and human hepatocytes were conducted to determine the CYP isoforms responsible for BI 425809 metabolism. In addition, two open-label, fixed-treatment period, phase I studies in healthy male volunteers are described. Period 1: participants received oral BI 425809 25 mg (single dose) on day 1; period 2: participants received multiple doses, across 10 days, of oral itraconazole or rifampicin combined with a single dose of oral BI 425809 25 mg on day 4/7 of the itraconazole/rifampicin treatment, respectively. Pharmacokinetic and safety endpoints were assessed in the absence/presence of itraconazole/rifampicin and included area under the concentration-time curve (AUC) over the time interval 0-167 h (AUC0‒167; itraconazole), 0-168 h (AUC0‒168; rifampicin), or 0-infinity (AUC0-∞; rifampicin and itraconazole), maximum measured concentration (Cmax) of BI 425809, and adverse events. RESULTS: In vitro results suggested that CYP3A4 accounted for ≥ 90% of the metabolism of BI 425809. BI 425809 exposure (adjusted geometric mean ratio [%]) was higher in the presence of itraconazole (AUC0‒167: 265.3; AUC0-∞: 597.0; Cmax: 116.1) and lower in the presence of rifampicin (AUC0‒168: 10.3; AUC0-∞: 9.8; Cmax: 37.4) compared with BI 425809 alone. Investigational treatments were well tolerated. CONCLUSIONS: Systemic exposure of BI 425809 was altered in the presence of strong CYP3A4 modulators, corroborating in vitro results that CYP3A4 mediates a major metabolic pathway for BI 425809. TRIAL REGISTRATION NUMBER: NCT02342717 (registered on 15 January 2015) and NCT03082183 (registered on 10 March 2017).


Subject(s)
Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Itraconazole/pharmacokinetics , Nootropic Agents/pharmacokinetics , Organic Chemicals/pharmacokinetics , Rifampin/pharmacokinetics , Schizophrenia/drug therapy , Adolescent , Adult , Area Under Curve , Cell Line , Cytochrome P-450 CYP3A Inhibitors/blood , Drug Synergism , Glycine Plasma Membrane Transport Proteins/metabolism , Healthy Volunteers , Humans , Itraconazole/administration & dosage , Itraconazole/blood , Male , Middle Aged , Nootropic Agents/administration & dosage , Nootropic Agents/blood , Organic Chemicals/administration & dosage , Organic Chemicals/blood , Rifampin/administration & dosage , Rifampin/blood , Young Adult
2.
J Antimicrob Chemother ; 75(9): 2657-2660, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32688374

ABSTRACT

BACKGROUND: The combination lopinavir/ritonavir is recommended to treat HIV-infected patients at the dose regimen of 400/100 mg q12h, oral route. The usual lopinavir trough plasma concentrations are 3000-8000 ng/mL. A trend towards a 28 day mortality reduction was observed in COVID-19-infected patients treated with lopinavir/ritonavir. OBJECTIVES: To assess the plasma concentrations of lopinavir and ritonavir in patients with severe COVID-19 infection and receiving lopinavir/ritonavir. PATIENTS AND METHODS: Mechanically ventilated patients with COVID-19 infection included in the French COVID-19 cohort and treated with lopinavir/ritonavir were included. Lopinavir/ritonavir combination was administered using the usual adult HIV dose regimen (400/100 mg q12h, oral solution through a nasogastric tube). A half-dose reduction to 400/100 mg q24h was proposed if lopinavir Ctrough was >8000 ng/mL, the upper limit considered as toxic and reported in HIV-infected patients. Lopinavir and ritonavir pharmacokinetic parameters were determined after an intensive pharmacokinetic analysis. Biological markers of inflammation and liver/kidney function were monitored. RESULTS: Plasma concentrations of lopinavir and ritonavir were first assessed in eight patients treated with lopinavir/ritonavir. Median (IQR) lopinavir Ctrough reached 27 908 ng/mL (15 928-32 627). After the dose reduction to 400/100 mg q24h, lopinavir/ritonavir pharmacokinetic parameters were assessed in nine patients. Lopinavir Ctrough decreased to 22 974 ng/mL (21 394-32 735). CONCLUSIONS: In mechanically ventilated patients with severe COVID-19 infections, the oral administration of lopinavir/ritonavir elicited plasma exposure of lopinavir more than 6-fold the upper usual expected range. However, it remains difficult to safely recommend its dose reduction without compromising the benefit of the antiviral strategy, and careful pharmacokinetic and toxicity monitoring are needed.


Subject(s)
Betacoronavirus , Coronavirus Infections/blood , Intensive Care Units/trends , Lopinavir/blood , Pneumonia, Viral/blood , Respiration, Artificial/trends , Ritonavir/blood , Administration, Oral , COVID-19 , Coronavirus Infections/drug therapy , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/blood , Drug Therapy, Combination , Female , Humans , Lopinavir/administration & dosage , Male , Middle Aged , Pandemics , Pharmaceutical Solutions/administration & dosage , Pharmaceutical Solutions/pharmacokinetics , Pneumonia, Viral/drug therapy , Prospective Studies , Ritonavir/administration & dosage , SARS-CoV-2
3.
Ann Hematol ; 99(9): 1989-2007, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32683457

ABSTRACT

Several small molecule inhibitors (SMIs) have been recently approved for AML patients. These targeted therapies could be more tolerable than classical antineoplastics, but potential drug-drug interactions (DDI) are relatively frequent. Underestimation or lack of appropriate awareness and management of DDIs with SMIs can jeopardize therapeutic success in AML patients, which often require multiple concomitant medications in the context of prior comorbidities or for the prevention and treatment of infectious and other complications. In this systematic review, we analyze DDIs of glasdegib, venetoclax, midostaurin, quizartinib, gilteritinib, enasidenib, and ivosidenib. CYP3A4 is the main enzyme responsible for SMIs metabolism, and strong CYP3A4 inhibitors, such azoles, could increase drug exposure and toxicity; therefore dose adjustments (venetoclax, quizartinib, and ivosidenib) or alternative therapies or close monitoring (glasdegib, midostaurin, and gilteritinib) are recommended. Besides, coadministration of strong CYP3A4 inducers with SMIs should be avoided due to potential decrease of efficacy. Regarding tolerability, QTc prolongation is frequently observed for most of approved SMIs, and drugs with a potential to prolong the QTc interval and CYP3A4 inhibitors should be avoided and replaced by alternative treatments. In this study, we critically assess the DDIs of SMIs, and we summarize best management options for these new drugs and concomitant medications.


Subject(s)
Antineoplastic Agents/blood , Cytochrome P-450 CYP3A Inhibitors/blood , Drug Approval , Drug Interactions/physiology , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/drug therapy , Antineoplastic Agents/adverse effects , Benzimidazoles/adverse effects , Benzimidazoles/blood , Bridged Bicyclo Compounds, Heterocyclic/adverse effects , Bridged Bicyclo Compounds, Heterocyclic/blood , Cytochrome P-450 CYP3A Inhibitors/adverse effects , Drugs, Investigational/adverse effects , Drugs, Investigational/metabolism , Humans , Long QT Syndrome/blood , Long QT Syndrome/chemically induced , Phenylurea Compounds/adverse effects , Phenylurea Compounds/blood , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/blood , Staurosporine/adverse effects , Staurosporine/analogs & derivatives , Staurosporine/blood , Sulfonamides/adverse effects , Sulfonamides/blood
5.
J Clin Pharmacol ; 60(7): 931-941, 2020 07.
Article in English | MEDLINE | ID: mdl-32035014

ABSTRACT

ACT-539313 is a potent and selective orexin-1 receptor antagonist. CYP3A is the major cytochrome P450 (CYP) enzyme involved in the metabolism and clearance of ACT-539313 in man. The main objective of this study was to investigate the effect of ACT-539313 on the pharmacokinetics of orally administered midazolam. Thereby, this single-center, open-label, fixed-sequence study investigated the CYP3A interaction potential of ACT-539313 following single- (on day 2) and repeated-dose (on day 11) twice-daily administration of 200 mg ACT-539313. Exposure to midazolam was higher during concomitant administration of single as well as after repeated doses of ACT-539313 over 10 days compared to midazolam alone (day 1). In the presence of ACT-539313, the geometric mean ratio of the maximum plasma concentration and the area under the plasma concentration-time curve from time 0 to 24 hours increased by 1.18- and 1.79-fold on day 2, and by 2.13- and 4.54-fold on day 11, respectively. A similar outcome was also shown in the additionally evaluated urinary 6ß-hydroxycortisol/cortisol ratio (6ß-CR), as the geometric mean ratio of the 6ß-CR showed a decrease to 0.78 on day 2 and to 0.61 on day 11. The most commonly reported adverse events (AEs) included somnolence and headache. All AEs were transient and of mild intensity. No treatment-related effects on vital signs, clinical laboratory, and electrocardiogram were observed. In summary, the observed corresponding decrease of both the validated, exogenous (midazolam/1-hydroxymidazolam ratio) and a frequently used endogenous (6ß-CR) marker of CYP3A activity is indicative of CYP3A inhibition occurring after ACT-539313 treatment.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Midazolam/pharmacokinetics , Orexin Receptor Antagonists/pharmacokinetics , Orexin Receptors/metabolism , Adult , Area Under Curve , Biomarkers/blood , Cytochrome P-450 CYP3A/drug effects , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/adverse effects , Cytochrome P-450 CYP3A Inhibitors/blood , Drug Administration Schedule , Drug Interactions , Healthy Volunteers , Humans , Hydrocortisone/analogs & derivatives , Hydrocortisone/urine , Male , Midazolam/administration & dosage , Midazolam/adverse effects , Midazolam/analogs & derivatives , Midazolam/blood , Middle Aged , Orexin Receptor Antagonists/administration & dosage , Orexin Receptor Antagonists/adverse effects , Orexin Receptor Antagonists/blood , Orexin Receptors/drug effects , Young Adult
6.
Biopharm Drug Dispos ; 40(5-6): 176-187, 2019 May.
Article in English | MEDLINE | ID: mdl-30985942

ABSTRACT

We previously verified a physiologically based pharmacokinetic (PBPK) model for mirabegron in healthy subjects using the Simcyp Simulator by incorporating data on the inhibitory effect on cytochrome P450 (CYP) 2D6 and a multi-elimination pathway mediated by CYP3A4, uridine 5'-diphosphate-glucuronosyltransferase (UGT) 2B7 and butyrylcholinesterase (BChE). The aim of this study was to use this PBPK model to assess the magnitude of drug-drug interactions (DDIs) in an elderly population with severe renal impairment (sRI), which has not been evaluated in clinical trials. We first determined the system parameters, and meta-analyses of literature data suggested that the abundance of UGT2B7 and the BChE activity in an elderly population with sRI was almost equivalent to and 20% lower than that in healthy young subjects, respectively. Other parameters, such as the CYP3A4 abundance, for an sRI population were used according to those built into the Simcyp Simulator. Second, we confirmed that the PBPK model reproduced the plasma concentration-time profile for mirabegron in an sRI population (simulated area under the plasma concentration-time curve (AUC) was within 1.5-times that of the observed value). Finally, we applied the PBPK model to simulate DDIs in an sRI population. The PBPK model predicted that the AUC for mirabegron with itraconazole (a CYP3A4 inhibitor) was 4.12-times that in healthy elderly subjects administered mirabegron alone, and predicted that the proportional change in AUC for desipramine (a CYP2D6 substrate) with mirabegron was greater than that in healthy subjects. In conclusion, the PBPK model was verified for the purpose of DDI assessment in an elderly population with sRI.


Subject(s)
Acetanilides/pharmacokinetics , Adrenergic beta-3 Receptor Agonists/pharmacokinetics , Models, Biological , Renal Insufficiency/metabolism , Thiazoles/pharmacokinetics , Acetanilides/blood , Adolescent , Adrenergic beta-3 Receptor Agonists/blood , Adult , Aged , Aging/metabolism , Butyrylcholinesterase/metabolism , Cytochrome P-450 CYP2D6 Inhibitors/blood , Cytochrome P-450 CYP2D6 Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/blood , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Desipramine/blood , Desipramine/pharmacokinetics , Drug Interactions , Female , Gemfibrozil/blood , Gemfibrozil/pharmacokinetics , Glucuronosyltransferase/metabolism , Humans , Itraconazole/blood , Itraconazole/pharmacokinetics , Lorazepam/blood , Lorazepam/pharmacokinetics , Male , Middle Aged , Renal Insufficiency/blood , Thiazoles/blood , Young Adult , Zidovudine/blood , Zidovudine/pharmacokinetics
7.
Article in English | MEDLINE | ID: mdl-30851617

ABSTRACT

A continuous effort has been given to find out a new drug that is effective against tuberculosis (TB) from both susceptible and resistant strains of Mycobacterium tuberculosis. Bedaquiline represents a recently approved anti-TB drug, which has a unique mechanism of action to fight against multi drug resistance (MDR). Some severe side effects and drug-drug interactions are associated with the treatment of bedaquiline. Moreover, World Health Organisation (WHO) has also been provided guidelines in the year of 2013 for the use of bedaquiline and encourages additional investigation into it. Hence, the pharmacokinetics of bedaquiline upon coadministration with the drug has to be explored in the preclinical model and for which a liquid chromatography tandem mass spectrometry (LC-MS/MS) based bioanalytical method for quantitation of bedaquiline will be useful. A simple, sensitive and rapid LC-MS/MS method was developed, validated and successfully applied to drug interactions of bedaquiline upon coadministration with cytochrome P450 3A4 (CYP3A4) inducers/inhibitors orally in Wistar rats. Results reveal that ciprofloxacin and fluconazole have marked effect to hinder the pharmacokinetics of bedaquiline but isoniazid, verapamil and carbamazepine have no significant effect on bedaquiline pharmacokinetics. Overall, this new bioanalytical method for estimation of bedaquiline in rat plasma was found to be helpful to assess the pharmacokinetics of bedaquiline and very much useful for evaluation of preclinical drug-drug interaction before considering costly and perilous clinical exploration.


Subject(s)
Antitubercular Agents/pharmacokinetics , Chromatography, Liquid/methods , Diarylquinolines/pharmacokinetics , Tandem Mass Spectrometry/methods , Animals , Antitubercular Agents/blood , Antitubercular Agents/chemistry , Cytochrome P-450 CYP3A Inhibitors/blood , Cytochrome P-450 CYP3A Inhibitors/chemistry , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Diarylquinolines/blood , Diarylquinolines/chemistry , Drug Interactions , Female , Linear Models , Rats , Rats, Wistar , Reproducibility of Results , Sensitivity and Specificity
8.
Clin Pharmacol Ther ; 105(1): 219-228, 2019 01.
Article in English | MEDLINE | ID: mdl-29696643

ABSTRACT

Dasabuvir is mainly metabolized by cytochrome P450 (CYP) 2C8 and is predominantly used in a regimen containing ritonavir. Ritonavir and clopidogrel are inhibitors of CYP3A4 and CYP2C8, respectively. In a randomized, crossover study in 12 healthy subjects, we examined the impact of clinical doses of ritonavir (for 5 days), clopidogrel (for 3 days), and their combination on dasabuvir pharmacokinetics, and the effect of ritonavir on clopidogrel. Clopidogrel, but not ritonavir, increased the geometric mean AUC0-∞ of dasabuvir 4.7-fold; range 2.0-10.1-fold (P = 8·10-7 ), compared with placebo. Clopidogrel and ritonavir combination increased dasabuvir AUC0-∞ 3.9-fold; range 2.1-7.9-fold (P = 2·10-6 ), compared with ritonavir alone. Ritonavir decreased the AUC0-4h of clopidogrel active metabolite by 51% (P = 0.0001), and average platelet inhibition from 51% without ritonavir to 31% with ritonavir (P = 0.0007). In conclusion, clopidogrel markedly elevates dasabuvir concentrations, and patients receiving ritonavir are at risk for diminished clopidogrel response.


Subject(s)
Clopidogrel/blood , Cytochrome P-450 CYP3A Inhibitors/blood , Platelet Aggregation Inhibitors/blood , Ritonavir/blood , Sulfonamides/blood , Uracil/analogs & derivatives , 2-Naphthylamine , Adult , Clopidogrel/administration & dosage , Cross-Over Studies , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Drug Synergism , Drug Therapy, Combination , Female , Humans , Male , Platelet Aggregation Inhibitors/administration & dosage , Ritonavir/administration & dosage , Sulfonamides/administration & dosage , Uracil/administration & dosage , Uracil/blood , Young Adult
9.
Transpl Infect Dis ; 21(1): e13007, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30295407

ABSTRACT

INTRODUCTION: Isavuconazole, a triazole antifungal, is an inhibitor of cytochrome P450 3A4, which also metabolizes tacrolimus and sirolimus. In previous studies, isavuconazole administration increased tacrolimus and sirolimus area under the curve values by 2.3-fold and 1.8-fold, respectively, in healthy adults and tacrolimus concentration/dose (C/D) ratio by 1.3-fold in solid organ transplant patients. We aimed to determine the magnitude of effect of isavuconazole administration on tacrolimus and sirolimus C/D ratios in allogeneic hematopoietic stem cell transplant (alloHSCT) patients. METHODS: A retrospective, single-center, single-arm study in adult alloHSCT patients who received at least 10 days of combination therapy with isavuconazole and tacrolimus and/or sirolimus as inpatients or outpatients was conducted. Tacrolimus and sirolimus trough serum concentrations were measured up to twice weekly for up to 4 weeks. RESULTS: Twenty-two patients receiving tacrolimus and twenty patients receiving sirolimus met the inclusion criteria. The mean C/D ratio increased from baseline by 1.42-fold for tacrolimus during week 1 (P = 0.002) and up to 1.56-fold for sirolimus during week 2 (P = 0.02). For the remaining timepoints, tacrolimus and sirolimus C/D ratios were not statistically significantly different from baseline. CONCLUSION: In alloHSCT patients, modest increases in tacrolimus and sirolimus C/D ratios from baseline were observed within the first 2 weeks after initiation of isavuconazole.


Subject(s)
Antifungal Agents/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , Immunosuppressive Agents/pharmacology , Adult , Aged , Antifungal Agents/therapeutic use , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors/blood , Cytochrome P-450 CYP3A Inhibitors/therapeutic use , Drug Interactions , Drug Therapy, Combination/adverse effects , Drug Therapy, Combination/methods , Female , Humans , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Nitriles/blood , Nitriles/pharmacology , Nitriles/therapeutic use , Pyridines/blood , Pyridines/pharmacology , Pyridines/therapeutic use , Retrospective Studies , Sirolimus/blood , Sirolimus/metabolism , Sirolimus/pharmacology , Sirolimus/therapeutic use , Tacrolimus/blood , Tacrolimus/metabolism , Tacrolimus/pharmacology , Tacrolimus/therapeutic use , Transplantation, Homologous/adverse effects , Triazoles/blood , Triazoles/pharmacology , Triazoles/therapeutic use , Young Adult
10.
Clin Pharmacol Drug Dev ; 7(8): 844-859, 2018 11.
Article in English | MEDLINE | ID: mdl-30044899

ABSTRACT

Amenamevir (formerly ASP2151) is a helicase-primase inhibitor being developed for the treatment of herpesvirus infection. Amenamevir is both a substrate and inducer of cytochrome P450 (CYP) 3A4. Three studies were done in healthy volunteers to investigate potential CYP3A pharmacokinetic interactions with the following drugs: (1) Midazolam (probe substrate for CYP3A): After 10 days' pretreatment with amenamevir 400 mg daily, geometric mean maximum concentration of drug in blood plasma (Cmax ) and area under the plasma drug concentration-time curve from time zero to infinity (AUC0-∞ ) of midazolam 7.5 mg were about 68% and 51%, respectively, of those after midazolam alone. (2) Cyclosporine (substrate and inhibitor of CYP3A): After 5 days' pretreatment with cyclosporine 100 mg twice daily, geometric mean Cmax of amenamevir after 400-mg and 1200-mg single doses was, respectively, about 66% and 69%, and AUC0-∞ about 82% and 79%, of those after amenamevir alone. (3) Ritonavir (inhibitor of CYP3A): When given with single doses of ritonavir 600 mg, geometric mean Cmax of amenamevir after 400-mg and 1200-mg single doses was, respectively, about 1.4 and 1.6 times higher, and geometric mean AUC0-∞ about 2.6 and 3.3 times higher, than after amenamevir alone. Amenamevir has the potential to be involved in CYP3A-mediated pharmacokinetic interactions in clinical practice.


Subject(s)
Cyclosporine/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Midazolam/pharmacokinetics , Oxadiazoles/pharmacokinetics , Ritonavir/pharmacokinetics , Adolescent , Adult , Cyclosporine/blood , Cyclosporine/pharmacology , Cytochrome P-450 CYP3A Inducers/blood , Cytochrome P-450 CYP3A Inducers/pharmacokinetics , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inhibitors/blood , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Drug Interactions , Healthy Volunteers , Humans , Male , Midazolam/blood , Midazolam/pharmacology , Middle Aged , Oxadiazoles/blood , Oxadiazoles/pharmacology , Ritonavir/blood , Ritonavir/pharmacology , Young Adult
11.
Pak J Pharm Sci ; 31(3): 801-806, 2018 May.
Article in English | MEDLINE | ID: mdl-29716858

ABSTRACT

Cardiac patients with weak immune system are susceptible to bacterial infections. Their prescriptions frequently contain simvastatin and clarithromycin together. The objective of present project was to assess the potential interaction between simvastatin and clarithromycin by evaluating the clarithromycin effects on the pharmacokinetics of simvastatin in healthy adult male subjects. The study design comprised of two phases, used at interval of one week. In first phase simvastatin 20 mg alone was administered to each volunteer. In second phase, co-administration of simvastatin 20 mg with clarithromycin 250 mg was made under similar specified conditions. Blood samples were collected at specified time intervals. Simvastatin plasma concentrations were analyzed through High Performance Liquid Chromatography with UV detector at 238 nm wavelength. Using one compartment open model, MW/PHARM version 3.02 software program was used by F. Rombut for pharmacokinetic parameters calculation. Clarithromycin co-treatment resulted in 2.3 fold increase in maximum plasma concentration Cmax (from 2.47±0.34 ng.mL-1 to 5.66±1.18 ng.mL-1; p<0.05) and 3.9 fold increase in area under time versus concentration curve from 0 to 10 hours AUC0-10 (from 15.10±3.73 ng.hr.mL-1 to 58.49±15.73 ng.hr.mL-1; p<0.05) of simvastatin. These results suggest that co-prescription of simvastatin and clarithromycin should be avoided to minimize the adverse events resulting from high simvastatin concentration, without sacrificing therapeutic worth of simvastatin.


Subject(s)
Clarithromycin/blood , Cytochrome P-450 CYP3A Inhibitors/blood , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Simvastatin/blood , Administration, Oral , Adult , Clarithromycin/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Drug Interactions/physiology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Male , Simvastatin/administration & dosage , Young Adult
12.
Eur J Clin Pharmacol ; 74(7): 913-920, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29572563

ABSTRACT

PURPOSE: Cytochrome P450 (CYP) 3A4 is responsible for the metabolism of more than 30% of clinically used drugs. Inherent between subject variability in clearance of CYP3A4 substrates is substantial; by way of example, midazolam clearance varies by > 10-fold between individuals before considering the impact of extrinsic factors. Relatively little is known about inter-racial variability in the activity of this enzyme. METHODS: This study assessed inter-racial variability in midazolam exposure in a cohort (n = 30) of CYP3A genotyped, age-matched healthy males of Caucasian and South Asian ancestries. Midazolam exposure was assessed at baseline, following 7 days of rifampicin and following 3 days of clarithromycin. RESULTS: The geometric mean baseline midazolam area under the plasma concentration curve (AUC0-6) in Caucasians (1057 µg/L/min) was 27% greater than South Asians (768 µg/L/min). Similarly, the post-induction midazolam AUC0-6 in Caucasians (308 µg/L/min) was 50% greater than South Asians (154 µg/L/min), while the post-inhibition midazolam AUC0-6 in Caucasians (1834 µg/L/min) was 41% greater than South Asians (1079 µg/L/min). The difference in baseline AUC0-6 between Caucasians and South Asians was statistically significant (p ≤ 0.05), and a trend toward significance (p = 0.067) was observed for the post-induction AUC0-6 ratio, in both unadjusted and genotype adjusted analyses. CONCLUSIONS: Significantly higher midazolam clearance was observed in healthy age-matched males of South Asian compared to Caucasian ancestry that was not explained by differences in the frequency of CYP3A genotypes.


Subject(s)
Asian People , Cytochrome P-450 CYP3A/metabolism , Midazolam/pharmacokinetics , White People , Adult , Area Under Curve , Asian People/genetics , Clarithromycin/blood , Clarithromycin/pharmacokinetics , Clarithromycin/pharmacology , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A Inducers/blood , Cytochrome P-450 CYP3A Inducers/pharmacokinetics , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inhibitors/blood , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Enzyme Induction , Genotype , Humans , Male , Midazolam/blood , Racial Groups , Rifampin/blood , Rifampin/pharmacokinetics , Rifampin/pharmacology , White People/genetics , Young Adult
13.
Basic Clin Pharmacol Toxicol ; 122(3): 331-340, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28945011

ABSTRACT

Schisantherin A and schisandrin A, the most abundant active ingredients of Wuzhi capsule, are known to inhibit tacrolimus metabolism by inhibiting CYP3A4/5. We aimed to predict the contribution of schisantherin A and schisandrin A to drug-drug interaction (DDI) between Wuzhi capsule and tacrolimus using physiologically-based pharmacokinetic (PBPK) modelling. Firstly, the inhibition mechanism of schisantherin A and schisandrin A on CYP3A4/5 was investigated. Thereafter, PBPK models of schisantherin A, schisandrin A and tacrolimus were established. Finally, tacrolimus pharmacokinetics were evaluated after the combined use with schisantherin A or schisandrin A. The blood area under the curve (AUC) of tacrolimus increased 1.77- and 2.61-fold after a single dose and multiple doses of schisantherin A, respectively. Meanwhile, schisandrin A inhibited tacrolimus metabolism to a smaller extent. Also, it showed that mechanism-based inhibition (MBI) played a more important role in DDI than reversible inhibition after long-term administration, while reversible inhibition was comparable to MBI after single-dose administration. In conclusion, we utilized PBPK modelling to quantify the contribution of schisantherin A and schisandrin A to DDI between tacrolimus and Wuzhi capsule. This may provide more insights for the rational use of this drug combination.


Subject(s)
Cyclooctanes/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Dioxoles/pharmacokinetics , Immunosuppressive Agents/pharmacokinetics , Lignans/pharmacokinetics , Models, Biological , Polycyclic Compounds/pharmacokinetics , Protective Agents/pharmacokinetics , Tacrolimus/pharmacokinetics , Area Under Curve , Biotransformation/drug effects , China , Computational Biology , Computer Simulation , Cyclooctanes/administration & dosage , Cyclooctanes/blood , Cytochrome P-450 CYP3A Inhibitors/blood , Dioxoles/administration & dosage , Dioxoles/blood , Dose-Response Relationship, Drug , Drug Interactions , Drugs, Chinese Herbal/analysis , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacokinetics , Expert Systems , Female , Humans , Immunosuppressive Agents/blood , Lignans/administration & dosage , Lignans/blood , Male , Polycyclic Compounds/administration & dosage , Polycyclic Compounds/blood , Protective Agents/administration & dosage , Protective Agents/analysis , Software , Tacrolimus/blood
14.
Xenobiotica ; 48(10): 1028-1036, 2018 Oct.
Article in English | MEDLINE | ID: mdl-28990837

ABSTRACT

1. Quercetin is a dietary flavonoid has extremely low water solubility and found to possess CYP3A inhibitory activity. The purpose of the present study was to evaluate the effect of quercetin and quercetin nanoparticles (NQC) on the pharmacokinetics of bromocriptine (BRO) in rats. 2. NQC prepared by antisolvent precipitation method and characterized by SEM and dissolution test. The following methods were used in this study i.e. in vitro liver and intestinal CYP3A microsomal activity and in vitro non-everted sac method. To confirm these findings, an in vivo pharmacokinetic study was also performed. 3. The results indicate that quercetin significantly (p < 0.05) inhibited the CYP3A activity in liver and intestinal microsomes. In non-everted sac study, the intestinal transport and Papp of BRO were significantly increased in NQC and quercetin groups. Furthermore, in vivo study revealed that the increased levels of Cmax and AUC were comparatively high in NQC pretreated group than quercetin group. In addition, pretreatment with quercetin and NQC significantly (p < 0.05) decreased the mean CL/F and Vd/F of BRO. 4. NQC pretreatment might be result in higher plasma levels of quercetin that could inhibit the CYP3A enzyme and enhanced the bioavailability of BRO.


Subject(s)
Bromocriptine/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Intestines/enzymology , Liver/enzymology , Nanoparticles/chemistry , Quercetin/pharmacology , Administration, Oral , Animals , Biological Transport/drug effects , Bromocriptine/administration & dosage , Bromocriptine/blood , Bromocriptine/pharmacology , Calibration , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/blood , Male , Microsomes/drug effects , Microsomes/enzymology , Nanoparticles/ultrastructure , Permeability , Rats, Wistar
15.
Eur J Pharm Sci ; 114: 55-63, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29223619

ABSTRACT

The tyrosine kinase inhibitors sorafenib and imatinib are important in the treatment of a range of cancers but adverse effects in some patients necessitate dosage modifications. CYP3A4 has a major role in the oxidation of sorafenib to its N-oxide and N-hydroxymethyl metabolites and also acts in concert with CYP2C8 to mediate imatinib N-demethylation. CYP3A4 expression and function are impaired in patients with advanced liver disease, whereas the functions of CYP2C enzymes are relatively preserved. We evaluated the biotransformation of sorafenib and imatinib in well-characterized microsomal fractions from 17 control subjects and 19 individuals with hepatic cirrhosis of varying severity. The principal findings were that liver disease impaired the microsomal oxidation of sorafenib to its major metabolites to 40-44% of control (P<0.01), whereas the N-demethylation of imatinib was relatively unimpaired. The impairments in sorafenib biotransformation were correlated with decreased serum albumin concentrations and increased serum bilirubin concentrations in patients with liver disease, but not with the overall grade of liver disease according to the Child-Pugh system. In contrast, there was no relationship between imatinib N-demethylation and clinicopathologic factors in liver disease patients. These findings were accounted for in terms of the differential roles of CYPs 3A4 and 2C8 in the intrinsic clearance of the drugs. CYP3A4 has the major role in the intrinsic clearance of sorafenib but plays a secondary role to CYP2C8 in the intrinsic clearance of imatinib. In agreement with these findings CYP2C protein expression and CYP2C8-mediated paclitaxel 6α-hydroxylation were unimpaired in cirrhotic livers. This information could be adapted in individualized approaches such as in vivo CYP3A4 phenotyping to optimize sorafenib safety and efficacy in cancer patients with liver dysfunction.


Subject(s)
Cytochrome P-450 Enzyme System/blood , Imatinib Mesylate/blood , Liver Cirrhosis/blood , Metabolic Clearance Rate/physiology , Microsomes, Liver/enzymology , Niacinamide/analogs & derivatives , Phenylurea Compounds/blood , Adolescent , Adult , Aged , Child , Child, Preschool , Cytochrome P-450 CYP3A Inhibitors/blood , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A Inhibitors/therapeutic use , Female , Humans , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Infant , Infant, Newborn , Liver Cirrhosis/drug therapy , Male , Metabolic Clearance Rate/drug effects , Microsomes, Liver/drug effects , Middle Aged , Niacinamide/blood , Niacinamide/pharmacology , Niacinamide/therapeutic use , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Sorafenib , Young Adult
16.
J Clin Pharmacol ; 58(4): 533-540, 2018 04.
Article in English | MEDLINE | ID: mdl-29194698

ABSTRACT

Pharmacokinetics and antithrombotic effects of the Factor Xa inhibitor rivaroxaban were studied in subjects with mild renal insufficiency concurrently taking the P-glycoprotein and moderate CYP3A inhibitor verapamil, a drug commonly administered to patients with hypertension, ischemic heart disease, or atrial fibrillation. Age-matched controls with normal renal function were studied concurrently. Subjects' overall mean age was 59 years. Mean creatinine clearance values in the 2 groups were 105 and 71 mL/min. After single 20-mg oral doses, rivaroxaban area under the curve (AUC) was increased by a factor of 1.11 (ratio of geometric means [RGM]) in mild renal insufficiency compared to controls. Verapamil coadministration independently increased AUC to the same extent in both the mild renal insufficiency and control groups (RGM, 1.39 and 1.43). Concurrent mild renal insufficiency and verapamil produced additive inhibition compared to controls without verapamil (RGM, 1.58). Prothrombin time (PT) prolongation and Factor Xa inhibition tracked plasma rivaroxaban, and were enhanced by verapamil. Concentration-response relationships for PT (linear) and Factor Xa inhibition (hyperbolic) were unaffected by renal function or verapamil. The absolute and relative increases in rivaroxaban AUC caused by verapamil in mild renal insufficiency subjects are potentially associated with an increased bleeding risk. Modification of recommended dosage may be required in this combination of circumstances to reduce risk to patients.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Factor Xa Inhibitors/administration & dosage , Renal Insufficiency/metabolism , Rivaroxaban/administration & dosage , Verapamil/administration & dosage , Adult , Aged , Cytochrome P-450 CYP3A Inhibitors/blood , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Drug Interactions , Factor Xa Inhibitors/blood , Factor Xa Inhibitors/pharmacokinetics , Female , Hemorrhage/chemically induced , Humans , Male , Middle Aged , Prothrombin Time , Rivaroxaban/blood , Rivaroxaban/pharmacokinetics , Verapamil/blood , Verapamil/pharmacokinetics
17.
Ther Drug Monit ; 39(4): 305-307, 2017 08.
Article in English | MEDLINE | ID: mdl-28700519

ABSTRACT

No drug-drug interaction study has been conducted to date for the combination of ombitasvir, paritaprevir/ritonavir, dasabuvir (3D), and mycophenolic acid (MPA). We here report the case of a hepatitis C virus-infected patient treated with 3D and MPA for vasculitis. In light of the threat of drug-drug interaction, the concentration of MPA was measured before, during, and 15 days after the end of the 3D treatment. Similar values were found at all 3 time points, thus indicating that there is probably no need to adapt MPA dosage to 3D.


Subject(s)
Anilides/blood , Carbamates/blood , Hepatitis C/blood , Macrocyclic Compounds/blood , Mycophenolic Acid/blood , Ritonavir/blood , Sulfonamides/blood , Uracil/analogs & derivatives , 2-Naphthylamine , Aged , Anilides/administration & dosage , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/blood , Antiviral Agents/administration & dosage , Antiviral Agents/blood , Carbamates/administration & dosage , Cyclopropanes , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/blood , Disease Management , Drug Interactions/physiology , Drug Monitoring/methods , Drug Therapy, Combination , Hepatitis C/diagnosis , Hepatitis C/drug therapy , Humans , Lactams, Macrocyclic , Macrocyclic Compounds/administration & dosage , Male , Mycophenolic Acid/administration & dosage , Proline/analogs & derivatives , Ritonavir/administration & dosage , Sulfonamides/administration & dosage , Uracil/administration & dosage , Uracil/blood , Valine
18.
Br J Clin Pharmacol ; 83(9): 1954-1965, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28370390

ABSTRACT

AIMS: The pharmacokinetics of voriconazole show a nonlinear dose-exposure relationship caused by inhibition of its own CYP3A-dependent metabolism. Because the magnitude of autoinhibition also depends on voriconazole concentrations, infusion rate might modulate voriconazole exposure. The impact of four different infusion rates on voriconazole pharmacokinetics was investigated. METHODS: Twelve healthy participants received 100 mg voriconazole intravenous over 4 h, 400 mg over 6 h, 4 h, and 2 h in a crossover design. Oral midazolam (3 µg) was given at the end of infusion. Blood and urine samples were collected up to 48 h. Voriconazole and its N-oxide metabolite were quantified using high-performance liquid chromatography coupled to tandem mass spectrometry. Midazolam estimated metabolic clearance (eCLmet) was calculated using a limited sampling strategy. Voriconazole-N-oxide inhibition of cytochrome P450 (CYP) isoforms 2C19 and 3A4 were assessed with the P450-Glo luminescence assay. RESULTS: Area under the concentration-time curve for 400 mg intravenous voriconazole was 16% (90% confidence interval: 12-20%) lower when administered over 6 h compared to 2 h infusion. Dose-corrected area under the concentration-time curve for 100 mg over 4 h was 34% lower compared to 400 mg over 4 h. Midazolam eCLmet was 516 ml min-1 (420-640) following 100 mg 4 h-1 voriconazole, 152 ml min-1 (139-166) for 400 mg 6 h-1 , 192 ml min-1 (167-220) for 400 mg 4 h-1 , and 202 ml min-1 (189-217) for 400 mg 2 h-1 . Concentration giving 50% CYP inhibition of voriconazole N-oxide was 146 ± 23 µmol l-1 for CYP3A4, and 40.2 ± 4.2 µmol l-1 for CYP2C19. CONCLUSIONS: Voriconazole pharmacokinetics is modulated by infusion rate, an autoinhibitory contribution voriconazole metabolism by CYP3A and 2C19 and to a lesser extent its main N-oxide metabolite for CYP2C19. To avoid reduced exposure, the infusion rate should be 2 h.


Subject(s)
Drug Administration Schedule , Voriconazole/pharmacology , Voriconazole/pharmacokinetics , Adult , Cytochrome P-450 CYP2C19/drug effects , Cytochrome P-450 CYP3A/drug effects , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/blood , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Female , Humans , Infusions, Intravenous , Male , Metabolic Clearance Rate , Midazolam/blood , Midazolam/pharmacokinetics , Midazolam/urine , Middle Aged , Voriconazole/administration & dosage , Voriconazole/analogs & derivatives , Young Adult
19.
J Pharm Pract ; 30(4): 459-463, 2017 Aug.
Article in English | MEDLINE | ID: mdl-27147607

ABSTRACT

Strong cytochrome P450 (CYP) 3A4 inhibitors may induce Cushing syndrome and subsequent adrenal insufficiency when administered concurrently with corticosteroids. This drug-drug interaction has been well described with HIV protease inhibitors. A similar drug-drug interaction with corticosteroids and other strong CYP 3A4 inhibitors (antifungals [imidazoles]) has recently been reported but remains less well known. To our knowledge, we report the first case of probable drug-induced Cushing syndrome with subsequent adrenal insufficiency as a result of concomitant oral voriconazole with intranasal mometasone and inhaled fluticasone administration as well as a review of the current literature supporting this drug-drug interaction.


Subject(s)
Adrenal Cortex Hormones/adverse effects , Adrenal Insufficiency/chemically induced , Adrenal Insufficiency/diagnosis , Antifungal Agents/adverse effects , Voriconazole/adverse effects , Administration, Inhalation , Adrenal Cortex Hormones/administration & dosage , Adrenal Cortex Hormones/blood , Adrenal Insufficiency/blood , Antifungal Agents/administration & dosage , Antifungal Agents/blood , Cushing Syndrome/blood , Cushing Syndrome/chemically induced , Cushing Syndrome/diagnosis , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/adverse effects , Cytochrome P-450 CYP3A Inhibitors/blood , Drug Interactions/physiology , Female , Humans , Middle Aged , Voriconazole/administration & dosage , Voriconazole/blood
20.
AAPS J ; 18(5): 1254-1261, 2016 09.
Article in English | MEDLINE | ID: mdl-27317471

ABSTRACT

Endogenous metabolites of cytochrome P450 (CYP3A) are useful in predicting drug-drug interactions between in vivo CYP3A inhibitors and inducers for clinical applications of CYP3A substrate drugs. This study aimed to develop predictable markers of the magnitude of hepatic CYP3A induction and inhibition in healthy female subjects using pharmacometabolomics. Twelve female subjects received midazolam during three study phases: 1 mg midazolam (control phase), 1 mg midazolam after pretreatment with 400 mg ketoconazole once daily for 4 days (CYP3A inhibition phase), and 2.5 mg midazolam after pretreatment with 600 mg rifampicin once daily for 10 days (CYP3A induction phase). Throughout the study, blood samples were collected 24 h after midazolam administration and urine samples at 12-h intervals during the 24 h before and after midazolam administration for the analysis of endogenous steroid metabolites. A statistical model was generated to predict midazolam clearance using measurements of endogenous metabolites associated with the inhibition and induction of CYP3A. Mean midazolam clearance decreased to ∼20% of control levels during the inhibition phase and increased more than 2-fold during the induction phase. Of the urine and plasma metabolites measured, the 6ß-hydroxycortisol/cortisol ratio was most significantly correlated with midazolam clearance during hepatic CYP3A inhibition and induction. Our results suggest that the urinary 6ß-hydroxycortisol/cortisol ratio is the best predictor of hepatic CYP3A activity under both maximal inhibition and maximal induction. Furthermore, the predictive model including 6ß-hydroxycortisol/cortisol as a covariate could be applied to predict the magnitude of CYP3A-mediated drug interactions.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Hydrocortisone/analogs & derivatives , Hydrocortisone/urine , Liver/metabolism , Metabolomics/methods , Midazolam/administration & dosage , Administration, Intravenous , Administration, Oral , Adult , Biomarkers/urine , Cytochrome P-450 CYP3A Inhibitors/blood , Drug Interactions/physiology , Enzyme Induction/drug effects , Enzyme Induction/physiology , Female , Humans , Ketoconazole/administration & dosage , Ketoconazole/blood , Liver/drug effects , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Midazolam/blood , Middle Aged , Rifampin/administration & dosage , Rifampin/blood , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...