Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-37632598

ABSTRACT

Enfortumab vedotin is an antibody-drug conjugate (ADC) comprised of a Nectin-4-directed antibody and monomethyl auristatin E (MMAE), which is primarily eliminated through P-glycoprotein (P-gp)-mediated excretion and cytochrome P450 3A4 (CYP3A4)-mediated metabolism. A physiologically based pharmacokinetic (PBPK) model was developed to predict effects of combined P-gp with CYP3A4 inhibitor/inducer (ketoconazole/rifampin) on MMAE exposure when coadministered with enfortumab vedotin and study enfortumab vedotin with CYP3A4 (midazolam) and P-gp (digoxin) substrate exposure. A PBPK model was built for enfortumab vedotin and unconjugated MMAE using the PBPK simulator ADC module. A similar model was developed with brentuximab vedotin, an ADC with the same valine-citrulline-MMAE linker as enfortumab vedotin, for MMAE drug-drug interaction (DDI) verification using clinical data. The DDI simulation predicted a less-than-2-fold increase in MMAE exposure with enfortumab vedotin plus ketoconazole (MMAE geometric mean ratio [GMR] for maximum concentration [Cmax], 1.15; GMR for area under the time-concentration curve from time 0 to last quantifiable concentration [AUClast], 1.38). Decreased MMAE exposure above 50% but below 80% was observed with enfortumab vedotin plus rifampin (MMAE GMR Cmax, 0.72; GMR AUClast, 0.47). No effect of enfortumab vedotin on midazolam or digoxin systemic exposure was predicted. Results suggest that combination enfortumab vedotin, P-gp, and a CYP3A4 inhibitor may result in increased MMAE exposure and patients should be monitored for potential adverse effects. Combination P-gp and a CYP3A4 inducer may result in decreased MMAE exposure. No exposure change is expected for CYP3A4 or P-gp substrates when combined with enfortumab vedotin.ClinicalTrials.gov identifier Not applicable.

2.
J Pharmacokinet Pharmacodyn ; 50(5): 365-376, 2023 10.
Article in English | MEDLINE | ID: mdl-37344637

ABSTRACT

Enzalutamide is known to strongly induce cytochrome P450 3A4 (CYP3A4). Furthermore, enzalutamide showed induction and inhibition of P-glycoprotein (P-gp) in in vitro studies. A clinical drug-drug interaction (DDI) study between enzalutamide and digoxin, a typical P-gp substrate, suggested enzalutamide has weak inhibitory effect on P-gp substrates. Direct oral anticoagulants (DOACs), such as apixaban and rivaroxaban, are dual substrates of CYP3A4 and P-gp, and hence it is recommended to avoid co-administration of these DOACs with combined P-gp and strong CYP3A inducers. Enzalutamide's net effect on P-gp and CYP3A for apixaban and rivaroxaban plasma exposures is of interest to physicians who treat patients for venous thromboembolism with prostate cancer. Accordingly, a physiologically-based pharmacokinetic (PBPK) analysis was performed to predict the magnitude of DDI on apixaban and rivaroxaban exposures in the presence of 160 mg once-daily dosing of enzalutamide. The PBPK models of enzalutamide and M2, a major metabolite of enzalutamide which also has potential to induce CYP3A and P-gp and inhibit P-gp, were developed and verified as perpetrators of CYP3A-and P-gp-mediated interaction. Simulation results predicted a 31% decrease in AUC and no change in Cmax for apixaban and a 45% decrease in AUC and a 25% decrease in Cmax for rivaroxaban when 160 mg multiple doses of enzalutamide were co-administered. In summary, enzalutamide is considered to decrease apixaban and rivaroxaban exposure through the combined effects of CYP3A induction and net P-gp inhibition. Concurrent use of these drugs warrants careful monitoring for efficacy and safety.


Subject(s)
Cytochrome P-450 CYP3A , Rivaroxaban , Male , Humans , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Pharmaceutical Preparations/metabolism , Models, Biological
3.
Drug Des Devel Ther ; 16: 1365-1381, 2022.
Article in English | MEDLINE | ID: mdl-35586186

ABSTRACT

Objective: To investigate the pharmacokinetics and safety of peficitinib (Janus kinase inhibitor for the treatment of rheumatoid arthritis) in healthy Chinese subjects following single and multiple doses. Methods: This open-label, randomized study was conducted at one site in China. Subjects received peficitinib 50, 100 or 150 mg as a single dose on Day 1 (fasted) and once daily from Days 8 to 13 in the multiple-dose period (fed). Blood samples were collected before administration each day, and up to 72h post administration. Pharmacokinetic assessments included area under the concentration curve (AUC), half-life (t1/2), maximum concentration (Cmax), and time to maximum concentration (tmax) of peficitinib and its metabolites (H1, H2 and H4). Treatment-emergent adverse events (TEAEs) were evaluated. Results: Thirty-six subjects were enrolled (12 per dose group). After a single dose of peficitinib, median tmax was 1.0-1.5h and mean t1/2 was 7.4-13.0h for all doses. In the multiple-dose period, median tmax was 1.5-2.0h. Dose-proportional increases in Cmax and AUC24h were observed for peficitinib and its metabolites following single and multiple doses, with minimal drug accumulation. The major metabolite was H2, with a systemic exposure of >150% of the parent AUC. Drug-related TEAEs were experienced by 5 (13.9%) and 12 (33.3%) subjects in the single- and multiple-dose periods, respectively. Following multiple doses of peficitinib, TEAEs were more frequent in higher than lower dose groups but were mild in severity with no related discontinuation or death. Conclusion: Following single and multiple doses of peficitinib in healthy Chinese subjects, peficitinib demonstrated rapid absorption and was well tolerated at all doses. Clinicaltrialsgov Identifier: NCT04143477.


Subject(s)
Adamantane , Janus Kinase Inhibitors , Adamantane/analogs & derivatives , Adamantane/pharmacology , Administration, Oral , Area Under Curve , Dose-Response Relationship, Drug , Healthy Volunteers , Humans , Niacinamide/analogs & derivatives , Niacinamide/pharmacology
4.
CPT Pharmacometrics Syst Pharmacol ; 9(11): 659-669, 2020 11.
Article in English | MEDLINE | ID: mdl-33030266

ABSTRACT

Direct oral anticoagulants, such as apixaban and rivaroxaban, are important for the treatment and prophylaxis of venous thromboembolism and to reduce the risk of stroke and systemic embolism in patients with nonvalvular atrial fibrillation. Because apixaban and rivaroxaban are predominantly eliminated by cytochrome P450 (CYP) 3A and P-glycoprotein (P-gp), concomitant use of combined P-gp and strong CYP3A4 inhibitors and inducers should be avoided. Physiologically-based pharmacokinetic models for apixaban and rivaroxaban were developed to estimate the net effect of CYP3A induction, P-gp inhibition, and P-gp induction by rifampicin. The disposition of rivaroxaban is more complex compared with apixaban because both hepatic and renal P-gp is considered to contribute to rivaroxaban elimination. Furthermore, organic anion transporter-3, a renal uptake transporter, may also contribute the elimination of rivaroxaban from systemic circulation. The models were verified with observed clinical drug-drug interactions with CYP3A and P-gp inhibitors. With the developed models, the predicted area under the concentration time curve and maximum concentration ratios were 0.43 and 0.48, respectively, for apixaban, and 0.50-0.52 and 0.72-0.73, respectively, for rivaroxaban when coadministered with 600 mg multiple doses of rifampicin and that were very close to observed data. The impact of each of the elimination pathways was assessed for rivaroxaban, and inhibition of CYP3A led to a larger impact over intestinal and hepatic P-gp. Inhibition of renal organic anion transporter-3 or P-gp led to an overall modest interaction. The developed apixaban and rivaroxaban models can be further applied to the investigation of interactions with other P-gp and/or CYP3A4 inhibitors and inducers.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Atrial Fibrillation/drug therapy , Cytochrome P-450 CYP3A/metabolism , Factor Xa Inhibitors/pharmacokinetics , Biological Transport , Cytochrome P-450 CYP3A Inducers/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Drug Interactions , Embolism/prevention & control , Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/therapeutic use , Humans , Pyrazoles/administration & dosage , Pyrazoles/pharmacokinetics , Pyrazoles/therapeutic use , Pyridones/administration & dosage , Pyridones/pharmacokinetics , Pyridones/therapeutic use , Rifampin/administration & dosage , Rifampin/pharmacokinetics , Rivaroxaban/administration & dosage , Rivaroxaban/pharmacokinetics , Rivaroxaban/therapeutic use , Stroke/prevention & control , Venous Thromboembolism/prevention & control
5.
Biopharm Drug Dispos ; 36(5): 265-74, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25600613

ABSTRACT

The effect of concomitant drugs having a cytochrome P450 (CYP) 3A inhibitory potency on tacrolimus exposure was predicted from in vitro metabolism results. In this study, the IC50 values of concomitant drugs on the formation of M-I, the major metabolite of tacrolimus, were determined, and the effect on oral exposure (AUCp.o.) of tacrolimus was assessed from static models. When an absorbed fraction (Fa ) of 0.97, intestinal wall availability (Fg) of 0.27 and fraction metabolized by CYP3A (fm(CYP3A)) of 0.8 were used, the least bias was observed for the prediction of the AUCp.o. of tacrolimus. The relationship of the IC50 values of 11 inhibitors between tacrolimus and typical CYP3A substrates (midazolam and testosterone) was also analysed. A strong correlation was found between the IC50 values of tacrolimus and typical CYP3A substrates (r(2) ≥ 0.85). The predictability of the effect of inhibitors on tacrolimus AUCp.o. was investigated based on the same static models with the use of published IC50 values for midazolam and testosterone. The bias for the prediction of tacrolimus AUCp.o. was minimal with the use of IC50 values determined using tacrolimus itself as a substrate. These results suggest that tacrolimus itself is still the best choice for predicting the AUCp.o. of tacrolimus, although our findings suggest that midazolam or testosterone may be used instead of tacrolimus to estimate roughly (predicted AUCp.o. within an approximately 2-fold range of observed values) the effect of CYP3A inhibitors on the tacrolimus AUCp.o.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors/pharmacology , Immunosuppressive Agents/pharmacology , Tacrolimus/pharmacology , Area Under Curve , Cytochrome P-450 CYP3A/metabolism , Drug Interactions , Humans , Microsomes, Liver/metabolism , Midazolam/pharmacology , Models, Biological , Testosterone/pharmacology
6.
Bioorg Med Chem ; 21(17): 5261-70, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23845281

ABSTRACT

Type 5 17ß-hydroxysteroid dehydrogenase (17ß-HSD5), also known as aldo-keto reductase 1C3 (AKR1C3), is a member of the aldo-keto reductase superfamily of enzymes and is expressed in the human prostate. One of the main functions of 17ß-HSD5 is to catalyze the conversion of the weak androgen, androstenedione, to the potent androgen, testosterone. The concentration of intraprostatic 5α-dihydrotestosterone (DHT) in patients following chemical or surgical castration has been reported to remain as high as 39% of that of healthy men, with 17ß-HSD5 shown to be involved in this androgen synthesis. Inhibition of 17ß-HSD5 therefore represents a promising target for the treatment of castration-resistant prostate cancer (CRPC). To investigate this, we conducted high-throughput screening (HTS) and identified compound 2, which displayed a structure distinct from known 17ß-HSD5 inhibitors. To optimize the inhibitory activity of compound 2, we first introduced a primary alcohol group. We then converted the primary alcohol group to a tertiary alcohol, which further enhanced the inhibitory activity, improved metabolic stability, and led to the identification of compound 17. Oral administration of compound 17 to castrated nude mice bearing the CWR22R xenograft resulted in the suppression of androstenedione (AD)-induced intratumoral testosterone production. Compound 17 also demonstrated good isoform selectivity, minimal inhibitory activity against either CYP or hERG, and enhanced pharmacokinetic and physicochemical properties.


Subject(s)
3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Hydroxyprostaglandin Dehydrogenases/antagonists & inhibitors , Indoles/chemistry , Piperidines/chemistry , 3-Hydroxysteroid Dehydrogenases/genetics , 3-Hydroxysteroid Dehydrogenases/metabolism , Administration, Oral , Aldo-Keto Reductase Family 1 Member C3 , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/therapeutic use , HEK293 Cells , Half-Life , Humans , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/metabolism , Indoles/pharmacokinetics , Indoles/therapeutic use , Male , Mice , Mice, Nude , Piperidines/pharmacokinetics , Piperidines/therapeutic use , Prostatic Neoplasms/drug therapy , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Testosterone/metabolism , Transplantation, Heterologous
7.
J Sep Sci ; 33(3): 348-58, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20169553

ABSTRACT

The effectiveness of several basic compounds for testing silica-based stationary phases was reviewed by applying them to recent columns for reversed-phase HPLC. Most octadecylsilylated (C18) stationary phases, prepared as a base-deactivated material from high-purity silica gel with endcapping, provided excellent peak shape and column efficiency for the bases including benzylamine and amitriptyline that once caused problems and were subsequently employed for testing silanol activities. However, a cyclic tertiary amine, dextrometorphan, was eluted as an acceptable peak from only a few columns at neutral pH. Such a more sensitive probe is expected to contribute to further improvement of the stationary phase for reversed-phase HPLC.


Subject(s)
Amines/chemistry , Amines/isolation & purification , Chromatography, Reverse-Phase/methods , Silanes/chemistry , Silicon Dioxide/chemistry , Chromatography, High Pressure Liquid/instrumentation , Chromatography, High Pressure Liquid/methods , Chromatography, Reverse-Phase/instrumentation , Ions/chemistry , Ions/isolation & purification
8.
Int J Pharm ; 317(1): 19-25, 2006 Jul 06.
Article in English | MEDLINE | ID: mdl-16621368

ABSTRACT

Human MCT4 (SLC16A3) is responsible for the efflux of L-lactic acid from skeletal muscle cells and is essential for muscle homeostasis. However, the effects of monocarboxylate drugs, such as statins on the MCT4-mediated transport of L-lactic acid have not been elucidated. Inhibition of L-lactic acid transport mediated by MCT4 might to lead to collapse of muscle homeostasis. The aim of this study was to establish an MCT4 transfected cell line and to clarify the transport mechanism of L-lactic acid and the effects of statins on this transport system. Results of Western blot analyses and immunohistochemistry studies indicated that the expression of CD147 and MCT4-FLAG protein were observed and was displayed clear plasma membrane localization in CD147 and MCT4-FLAG co-transfected cell line (cm cells). Uptake of L-lactic acid in cm cells was significantly greater than that in cells transfected with a vector alone. L-lactic acid uptake was concentration-dependent with a K(m) value of 28.43+/-3.87 mM. The results of a previous study showing a K(m) value of 28.5 mM in hMCT4-expressed oocytes. Lipophilic statins significantly inhibited [(14)C] L-lactic acid uptake in a concentration-dependent manner. In contrast, the inhibitory effects of hydrophilic statins were very weak.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/antagonists & inhibitors , Muscle Proteins/antagonists & inhibitors , Animals , Atorvastatin , Basigin/genetics , Basigin/metabolism , Fatty Acids, Monounsaturated/pharmacology , Fluvastatin , Heptanoic Acids/pharmacology , Humans , Indoles/pharmacology , LLC-PK1 Cells , Lovastatin/pharmacology , Monocarboxylic Acid Transporters/genetics , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Pyridines/pharmacology , Pyrroles/pharmacology , Simvastatin/analogs & derivatives , Simvastatin/pharmacology , Swine , Transfection
9.
J Agric Food Chem ; 53(15): 6100-4, 2005 Jul 27.
Article in English | MEDLINE | ID: mdl-16029002

ABSTRACT

In clinical, patients usually take many kinds of drugs at the same time. Thus, drug-drug interactions involving transporters can often directly affect the therapeutic safety and efficacy of many important drugs. However, there have been few studies on food-drug interactions involving transporters. Dietary polyphenols have been widely assumed to be beneficial for human health. Polyphenols are found ubiquitously, and they are commercially prepared and used as functional foods. We have reported that ferulic acid, which is one of the most well-known polyphenols and is used as a functional food, affected the transport of nateglinide, an antidiabetic drug, by Caco-2 cells. In this study, we investigated the effects of other polyphenols on the nateglinide/H(+) transport system. We report here that caffeic acid and p-coumaric acid have a different inhibitory manner on the uptake of nateglinide. The results of this study are useful to identify the substrate specificity of the nateglinide/H(+) cotransporter.


Subject(s)
Cyclohexanes/metabolism , Flavonoids/pharmacology , Hypoglycemic Agents/metabolism , Phenols/pharmacology , Phenylalanine/analogs & derivatives , Protons , Symporters/metabolism , Biological Transport/drug effects , Caco-2 Cells , Caffeic Acids/pharmacology , Coumaric Acids/pharmacology , Humans , Nateglinide , Phenylalanine/metabolism , Polyphenols , Propionates
10.
J Agric Food Chem ; 53(7): 2499-502, 2005 Apr 06.
Article in English | MEDLINE | ID: mdl-15796585

ABSTRACT

In clinical, patients usually take many kinds of drugs at the same time. Thus, drug-drug interactions involving transporters can often directly affect the therapeutic safety and efficacy of many drugs. However, there have been few studies on food-drug interactions involving transporters. Dietary polyphenols have been widely assumed to be beneficial to human health. Polyphenols are commercially prepared and used as functional foods. We report here for the first time that ferulic acid, which is widely used as a functional food, affects the transport of clinical agents. It is important to be aware of the potential of food-drug interactions and to act in order to prevent undesirable and harmful clinical consequences.


Subject(s)
Coumaric Acids/metabolism , Cyclohexanes/metabolism , Fluorescein/metabolism , Food , Monocarboxylic Acid Transporters/metabolism , Phenylalanine/analogs & derivatives , Phenylalanine/metabolism , Caco-2 Cells , Coumaric Acids/pharmacology , Cyclohexanes/pharmacology , Drug Interactions , Humans , Hypoglycemic Agents , Intestinal Mucosa/metabolism , Monocarboxylic Acid Transporters/drug effects , Nateglinide , Phenylalanine/pharmacology
11.
Biochim Biophys Acta ; 1668(2): 190-4, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15737329

ABSTRACT

Nateglinide, a novel oral hypoglycemic agent, rapidly reaches its maximum serum concentration after oral administration, suggesting that it is rapidly absorbed in the intestine. However, nateglinide itself is not transported by MCT1 or PEPT1. The aim of this study was to characterize the transporters on the apical side of the small intestine that are responsible for the rapid absorption of nateglinide. It has been reported that the uptake of fluorescein by Caco-2 cells occurs via an H+-driven transporter and that the intestinal fluorescein transporter is probably not MCT1. We examined the contribution of the fluorescein transporter to the uptake of nateglinide by Caco-2 cells. Fluorescein competitively inhibited H+-dependent nateglinide uptake. All of fluorescein transporter inhibitors examined reduced the uptake of nateglinide. Furthermore, nateglinide inhibited fluorescein uptake. We conclude that the intestinal nateglinide/H+ cotransport system is identical to the intestinal fluorescein/H+ cotransport system.


Subject(s)
Carrier Proteins/metabolism , Cyclohexanes/pharmacokinetics , Fluorescein/metabolism , Intestinal Mucosa/metabolism , Membrane Transport Proteins/metabolism , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacokinetics , Absorption , Biological Transport/physiology , Caco-2 Cells , Humans , Hydrogen-Ion Concentration , Metabolic Clearance Rate , Nateglinide
12.
J Pharmacol Exp Ther ; 312(1): 77-82, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15316092

ABSTRACT

(-)-N-(trans-4-Isopropylcyclohexanecarbonyl)-D-phenylalanine (nateglinide) is a novel oral hypoglycemic agent possessing a carboxyl group and a peptide-type bond in its structure. Although nateglinide quickly reaches the maximal serum concentration after oral administration, nateglinide itself is not transported by PepT1 or MCT1. The aim of this study was to characterize the transporters on the apical side of the small intestine that are responsible for the rapid absorption of nateglinide. The uptake of nateglinide by rat intestinal brush-border membrane vesicles is associated with a proton-coupled transport system. Ceftibuten competitively inhibited H(+)-dependent nateglinide uptake. Glycylsarcosine (Gly-Sar), cephradine, and cephalexin did not significantly inhibit the uptake of nateglinide. The combination of Gly-Sar and nateglinide greatly reduced the uptake of ceftibuten. The effect of the combined treatment was significantly greater than that of Gly-Sar alone. Furthermore, nateglinide competitively inhibited H(+)-driven ceftibuten transporter-mediated ceftibuten uptake. Ceftibuten transport occurs via at least two H(+)-dependent transport systems: one is PepT1, and the other is the ceftibuten/H(+) cotransport system. On the other hand, we demonstrated that nateglinide transport occurs via a single system that is H(+) dependent but is distinct from PepT1 and may be identical to the ceftibuten/H(+) cotransport system.


Subject(s)
Cyclohexanes/pharmacokinetics , Intestine, Small/metabolism , Microvilli/metabolism , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacokinetics , Protons , Animals , Biological Transport/drug effects , Ceftibuten , Cephalosporins/pharmacology , Dose-Response Relationship, Drug , Hypoglycemic Agents/pharmacokinetics , Nateglinide , Rats
13.
Hepatogastroenterology ; 51(57): 652-7, 2004.
Article in English | MEDLINE | ID: mdl-15143885

ABSTRACT

BACKGROUND/AIMS: Use of endoscopic papillary balloon dilation (EPBD) for the treatment of common bile duct stones has increased in recent years, owing to its simplicity and its advantage of preserving sphincter function. It has been reported that EPBD is associated with a lower risk of bleeding, but a higher risk of pancreatitis than endoscopic sphincterotomy. However, there have been few reports on studies of post-EPBD pancreatitis. This report concerns the use of EPBD at our department for the treatment of common bile duct stones and early postoperative complications, with a focus on pancreatitis. METHODOLOGY: The study was conducted in 63 patients with choledocholithiasis, including 4 patients with cirrhosis and 21 patients with periampullary diverticula. The stones were extracted after EPBD conducted with an 8-mm dilatation balloon. RESULTS: Complete removal of stones was achieved in 53 out of 63 patients (84.1%). Pancreatitis meeting the criteria of Cotton et al. occurred in 7 of the 63 patients (11.1%), while 12 patients (19.5%) were affected when milder cases of pancreatitis were included. Severe pancreatitis occurred in 1 patient only. Cholangitis occurred in 3 patients (4.8%) and basket impaction occurred in 1 patient (1.6%), but no serious complications such as bleeding or perforation were encountered. CONCLUSIONS: These results suggest that EPBD is an effective procedure for the treatment of common bile duct stones, with a low risk of serious complications.


Subject(s)
Catheterization , Gallstones/therapy , Adult , Aged , Aged, 80 and over , Ampulla of Vater , Female , Humans , Male , Middle Aged , Postoperative Complications/etiology
SELECTION OF CITATIONS
SEARCH DETAIL