Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Drug Metab Dispos ; 49(8): 601-609, 2021 08.
Article in English | MEDLINE | ID: mdl-34011531

ABSTRACT

Ozanimod, recently approved for treating relapsing multiple sclerosis, produced a disproportionate, active, MAO B-catalyzed metabolite (CC112273) that showed remarkable interspecies differences and led to challenges in safety testing. This study explored the kinetics of CC112273 formation from its precursor RP101075. Incubations with human liver mitochondrial fractions revealed K Mapp, V max, and intrinsic clearance (Clint) for CC112273 formation to be 4.8 µM, 50.3 pmol/min/mg protein, and 12 µl/min/mg, respectively, whereas Michaelis-Menten constant (K M) with human recombinant MAO B was 1.1 µM. Studies with liver mitochondrial fractions from preclinical species led to K Mapp, V max, and Clint estimates of 3.0, 35, and 33 µM, 80.6, 114, 37.3 pmol/min/mg, and 27.2, 3.25, and 1.14 µl/min/mg in monkey, rat, and mouse, respectively, and revealed marked differences between rodents and primates, primarily attributable to differences in the K M Comparison of Clint estimates revealed monkey to be ∼2-fold more efficient and the mouse and rat to be 11- and 4-fold less efficient than humans in CC112273 formation. The influence of stereochemistry on MAO B-mediated oxidation was also investigated using the R-isomer of RP101075 (RP101074). This showed marked selectivity toward catalysis of the S-isomer (RP101075) only. Docking into MAO B crystal structure suggested that although both the isomers occupied its active site, only the orientation of RP101075 presented the C-H on the α-carbon that was ideal for the C-H bond cleavage, which is a requisite for oxidative deamination. These studies explain the basis for the observed interspecies differences in the metabolism of ozanimod as well as the substrate stereospecificity for formation of CC112273. SIGNIFICANCE STATEMENT: This study evaluates the enzymology and the species differences of the major circulating metabolite of ozanimod, CC112273. Additionally, the study also explores the influence of stereochemistry on MAO B-catalyzed reactions. The study is of significance to the DMD readers given that this oxidation is catalyzed by a non-cytochrome P450 enzyme, and that marked species difference and notable stereospecificity was observed in MAO B-catalyzed biotransformation when the indaneamine enantiomers were used as substrates.


Subject(s)
Indans/pharmacokinetics , Monoamine Oxidase/metabolism , Oxadiazoles/pharmacokinetics , Animals , Biotransformation , Deamination , Drug Evaluation, Preclinical , Haplorhini , Humans , Indans/blood , Metabolic Clearance Rate , Mice , Mitochondria, Liver/metabolism , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Oxadiazoles/blood , Oxidation-Reduction , Rats , Species Specificity , Sphingosine 1 Phosphate Receptor Modulators/blood , Sphingosine 1 Phosphate Receptor Modulators/pharmacokinetics , Stereoisomerism
2.
Clin Pharmacol Drug Dev ; 10(11): 1316-1324, 2021 11.
Article in English | MEDLINE | ID: mdl-33864709

ABSTRACT

Opicapone (OPC) is a third-generation catechol-O-methyltransferase inhibitor developed to treat Parkinson disease and motor fluctuations. This open-label, single-center, phase 1 study aimed to evaluate the pharmacokinetics (PK) of OPC and its metabolites when administered as single and multiple doses in healthy White and Chinese subjects. The study enrolled a total of 30 White and Chinese healthy subjects, equally balanced among groups. The first dose of OPC was administered orally as a single dose of 50 mg on day 1, followed by a 10-day once-daily treatment from day 5 to day 14. Plasma concentrations of OPC and its metabolites were measured at 0 to 72 and 0 to 144 hours after dosing for single dose and multiple dose, respectively. Moreover, urine concentrations of OPC and its metabolite were measured 0 to 24 hours after dosing. PK parameters were derived from noncompartmental analysis. Geometric mean ratios and 90% confidence intervals for the main PK parameters were conducted to evaluate the ethnic difference between White and Chinese subjects. The plasma and urine exposure of OPC and its metabolites in Chinese subjects were similar to those in White subjects. These results indicated that ethnicity had no significant impact on PK of OPC between White and Chinese subjects.


Subject(s)
Asian People , Catechol O-Methyltransferase Inhibitors/pharmacokinetics , Oxadiazoles/pharmacokinetics , White People , Adult , Catechol O-Methyltransferase Inhibitors/blood , Catechol O-Methyltransferase Inhibitors/urine , China , Female , Healthy Volunteers , Humans , Male , Oxadiazoles/blood , Oxadiazoles/urine
3.
Bioanalysis ; 12(21): 1545-1555, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33064028

ABSTRACT

Background: This paper describes for the first-time analytical procedures established to resolve the challenges associated with simultaneous and direct quantification of ataluren and ataluren-O-1ß-acyl glucuronide (AAG) by LC-MS/MS in human plasma and urine matrices. Methodology/results: The plasma quantification method was validated for calibration range of 12.5-12500 ng/ml for ataluren and 6.25-2500 ng/ml for AAG. The urine quantification method was validated for calibration range of 0.01-10 and 1-1000 µg/ml for ataluren and AAG, respectively. Plasma and urine samples were stabilized upon collection and through storage to prevent hydrolysis and acyl migration of AAG. Conclusion: Methods described in this paper enabled successful completion of ataluren clinical pharmacology studies for simultaneous pharmacokinetic assessment of ataluren and AAG.


Subject(s)
Chromatography, Liquid/methods , Oxadiazoles/blood , Oxadiazoles/urine , Tandem Mass Spectrometry/methods , Humans , Oxadiazoles/pharmacology
4.
Adv Ther ; 37(7): 3234-3245, 2020 07.
Article in English | MEDLINE | ID: mdl-32440976

ABSTRACT

INTRODUCTION: Amenamevir (ASP2151), a herpesvirus helicase-primase inhibitor, is currently used for the treatment of herpes zoster in Japan. Amenamevir is mainly metabolized in the liver, and urinary excretion of amenamevir is approximately 10% in healthy adults. The increase of systemic exposure in non-dialysis patients with severe renal impairment was much less than that associated with nucleoside antiviral agents. The aim of this study was to evaluate the pharmacokinetics and dialyzability of a single oral dose (400 mg) of amenamevir in hemodialysis patients. METHODS: This was a single-arm, open-label, multicenter clinical pharmacology study. Nine patients aged 20-80 years with end-stage kidney disease and undergoing maintenance hemodialysis three times weekly were enrolled. Pharmacokinetics and dialyzability were investigated by serial collection of blood samples until 48 h post-dose during the study. RESULTS: The maximum plasma concentration and time to reach maximum plasma concentration during 24 h post-dose were 1585 ng/mL and 6.2 h, respectively. The area under the plasma concentration-time curve (AUC) from time zero to 24 h was 23,890 ng h/mL. The median terminal elimination half-life within 24 h before, during, and after hemodialysis was 14.7, 15.2, and 12.4 h, respectively. The AUC in hemodialysis patients was approximately double that in healthy adults. This increase in AUC was much less than that reported in nucleoside antiviral agents. The hemodialysis clearance, elimination fraction percentage, and amount of amenamevir removed were 37.8 mL/min, 28.1%, and 132.0 µg, respectively. The amount of amenamevir removed by hemodialysis was minimal. None of the hemodialysis parameters were associated with serum albumin. This study revealed no clinically relevant safety concerns. CONCLUSION: There were no clinically relevant safety concerns when 400 mg of amenamevir was administered as a single dose to hemodialysis patients without dose adjustment and/or modification of the dosing schedule. TRIAL REGISTRATION: JapicCTI-184242.


Subject(s)
Antiviral Agents/adverse effects , Antiviral Agents/therapeutic use , Herpes Zoster/drug therapy , Oxadiazoles/blood , Oxadiazoles/pharmacokinetics , Oxadiazoles/therapeutic use , Renal Insufficiency/therapy , Administration, Oral , Adult , Aged , Aged, 80 and over , Female , Humans , Japan , Male , Middle Aged , Renal Dialysis , Young Adult
5.
Invest New Drugs ; 38(4): 1067-1076, 2020 08.
Article in English | MEDLINE | ID: mdl-31471863

ABSTRACT

Purpose BAL101553, the prodrug of the microtubule-destabilizer BAL27862, previously showed signs of antitumor activity when administered as a 2-h infusion, but its use was limited by vascular toxicity. We investigated an alternative dosing strategy aimed at improving the safety profile of BAL101553. Methods This multicenter, open-label, Phase 1 dose-escalation study used a 3 + 3 design to determine the maximum tolerated dose (MTD), dose-limiting toxicities (DLTs), pharmacokinetics, and antitumor activity of BAL101553 administered as a 48-h IV infusion on Days 1, 8, and 15 of a 28-day cycle. Patients received oral BAL101553 on Days 15-21 of cycle 2 to assess oral bioavailability. Results BAL101553 was well tolerated at doses up to ≤70 mg/m2. Three grade 3 DLTs occurred: hypotension (70 mg/m2), hyponatremia and neutropenia (both 90 mg/m2). The MTD for 48-h IV BAL101553 was 70 mg/m2. At this dose level, the AUC for BAL27862 was 8580 ng.h/mL and the Cmax was 144 ng/mL. No apparent dose-related effects on blood pressure were observed with 48-h BAL101553 IV infusion. BAL27862 oral bioavailability was >80%. Conclusions Continuous 48-h IV BAL101553 infusion achieved higher exposure of the BAL27862 active metabolite than a 2-h infusion at the RP2D and did not cause vascular toxicity. Clinicaltrials.gov registration: NCT02895360.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzimidazoles/therapeutic use , Neoplasms/drug therapy , Oxadiazoles/therapeutic use , Prodrugs/therapeutic use , Administration, Oral , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Benzimidazoles/adverse effects , Benzimidazoles/blood , Benzimidazoles/pharmacokinetics , Female , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Microtubules , Middle Aged , Neoplasms/blood , Neoplasms/metabolism , Oxadiazoles/adverse effects , Oxadiazoles/blood , Oxadiazoles/pharmacokinetics , Prodrugs/adverse effects , Prodrugs/pharmacokinetics , Treatment Outcome
6.
Drug Test Anal ; 12(1): 101-108, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31486294

ABSTRACT

The small molecule, stemazole, has significant therapeutic effects on neurodegenerative diseases, such as Alzheimer's disease (AD), due to its neuroprotective effects and remarkable survival-promoting activity in stem cells. However, pharmacokinetic properties of stemazole were unclear. In this study, a rapid and effective ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method was developed to detect stemazole. The detector was operated in the positive-ion mode with an electrospray ionization (ESI) interface in multiple reaction monitoring (MRM) mode. Chromatographic separation was performed on an Acquity UPLC® BEH C18 column with gradient elution. Stemazole was extracted from plasma following a one-step protein precipitation method. The method was fully validated for its selectivity, specificity, and sensitivity. The calibration curve range of 5-1125 ng/mL showed good linearity for stemazole. Intra-day and inter-day precision rates were less than 10%, and accuracy ranged from 95.87% to 105.23%. The pharmacokinetic profiles were illustrated through the newly developed method for the first time. The absolute oral bioavailability of stemazole is 32.10%. Therefore, it is feasible as an oral medication, which greatly facilitates its broad application. The biological distribution of tritium-labeled stemazole in mice was studied, and the results showed that stemazole was absorbed rapidly and distributed widely, mainly in the liver and kidneys. A specific amount was also detected in the brain, which provides a prerequisite for the use of stemazole to treat neurodegenerative diseases. This work represents first description of the pharmacokinetics, bioavailability, and tissue distribution of stemazole and will lay the foundation for further investigation and drug development.


Subject(s)
Hydrazines/pharmacokinetics , Oxadiazoles/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Chromatography, High Pressure Liquid , Hydrazines/administration & dosage , Hydrazines/blood , Male , Mice , Oxadiazoles/administration & dosage , Oxadiazoles/blood , Rats, Sprague-Dawley , Tandem Mass Spectrometry , Tissue Distribution , Tritium/analysis
7.
Xenobiotica ; 49(8): 961-969, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30124356

ABSTRACT

A 1,2,4-oxadiazole ring-containing compound DS-8500a was developed as a novel G protein-coupled receptor 119 agonist. In vivo metabolic fates of [14C]DS-8500a differently radiolabeled in the benzene ring or benzamide side carbon in rats were investigated. Differences in mass balances were observed, primarily because after the oxadiazole ring-opening and subsequent ring-cleavage small-molecule metabolites containing the benzene side were excreted in the urine, while those containing the benzamide side were excreted in the bile. DS-8500a was detected at trace levels in urine and bile, demonstrating extensive metabolism prior to urinary/biliary excretion. At least 16 metabolite structures were proposed in plasma, urine, and bile samples from rats treated with [14C]DS-8500a. Formation of a ring-opened metabolite (reduced DS-8500a) in hepatocytes of humans, monkeys, and rats was confirmed; however, it was not affected by typical inhibitors of cytochrome P450s, aldehyde oxidases, or carboxylesterases in human hepatocytes. Extensive formation of the ring-opened metabolite was observed in human liver microsomes fortified with an NADPH-generating system under anaerobic conditions. These results suggest an in vivo unique reductive metabolism of DS-8500a is mediated by human non-cytochrome P450 enzymes.


Subject(s)
Benzamides/metabolism , Cyclopropanes/metabolism , Metabolic Networks and Pathways , Oxadiazoles/metabolism , Receptors, G-Protein-Coupled/agonists , Administration, Oral , Anaerobiosis , Animals , Benzamides/administration & dosage , Benzamides/blood , Benzamides/pharmacokinetics , Carbon Radioisotopes/chemistry , Cyclopropanes/administration & dosage , Cyclopropanes/blood , Cyclopropanes/pharmacokinetics , Humans , Macaca fascicularis , Male , Oxadiazoles/administration & dosage , Oxadiazoles/blood , Oxadiazoles/pharmacokinetics , Oxidation-Reduction , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/metabolism
8.
Clin Pharmacol Drug Dev ; 8(5): 595-602, 2019 07.
Article in English | MEDLINE | ID: mdl-30412362

ABSTRACT

Amenamevir is an inhibitor of the helicase-primase enzyme complex developed for the treatment of varicella zoster virus. This mass balance study investigated the absorption, metabolism, and excretion of a single dose (200 mg) of 14 C-labeled amenamevir in healthy male volunteers. Blood, urine, and feces samples were collected for up to 8 days after the dose. Safety and tolerability were assessed through voluntary reporting of adverse events, physical examination, and clinical laboratory testing. Amenamevir was rapidly absorbed, with a median time to peak drug concentration of 1.0 to 1.5 hours and a plasma half-life of 8 to 9 hours. Overall, 95.3% of the administered dose was recovered, with the majority of radiolabeled drug excreted in feces (74.6%) followed by urine (20.6%). The major route of elimination was fecal, with around 70% of the dose excreted as metabolites and <0.1% as the unchanged drug. Metabolic profiling revealed that predominantly radiolabeled amenamevir (80%) and its hydroxyl metabolite R5 (up to 7.1%) were present in plasma. Single-dose amenamevir was well tolerated; 3 transient and mild adverse events were reported in 3 subjects. Overall, >95% of a single 200-mg dose of amenamevir was eliminated by 168 hours after the dose, with the major route of elimination being fecal.


Subject(s)
Antiviral Agents/pharmacokinetics , Oxadiazoles/pharmacokinetics , Adult , Antiviral Agents/adverse effects , Antiviral Agents/blood , Antiviral Agents/urine , Carbon Radioisotopes , Feces/chemistry , Half-Life , Humans , Male , Middle Aged , Oxadiazoles/adverse effects , Oxadiazoles/blood , Oxadiazoles/urine , Young Adult
9.
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
10.
Clin Pharmacol Drug Dev ; 7(8): 860-870, 2018 11.
Article in English | MEDLINE | ID: mdl-29870591

ABSTRACT

Amenamevir (formerly ASP2151) induces cytochrome P450 (CYP)2B6 and CYP3A4 and inhibits CYP2C8.  We conducted 2 studies, 1 using montelukast as a probe to assess CYP2C8 and the other bupropion to assess CYP2B6.  The montelukast study examined the effect of amenamevir on the pharmacokinetics of montelukast in 24 healthy men: each subject received montelukast 10 mg alone, followed by montelukast 10 mg with amenamevir 400 mg, or vice versa after a washout period.  In the bupropion study, 24 subjects received a single dose of 150 mg bupropion on days 1, 15, 22, and 29, and repeated once-daily doses of 400 mg amenamevir on days 6-15.  Amenamevir increased peak concentration and area under the concentration-time curve of montelukast by about 22% (ratio 121.7%, 90%CI [114.8, 129.1]; 121% [116.2, 128.4], respectively) with a similar increase in hydroxymontelukast (ratio 121.4%, 90%CI [106.4, 138.5]; 125.6 % [111.3, 141.7]).  Amenamevir reduced peak concentration and area under the concentration-time curve of bupropion by 16% (84.29%, 90%CI [78.00, 91.10]; 84.07%, 90%CI [78.85, 89.63]), with recovery after 1 week; the pharmacokinetics of the primary metabolite hydroxybupropion was unaffected.  Thus, amenamevir increased plasma concentrations of montelukast and decreased those of bupropion, but it did not do so enough to require dose adjustment of coadministered substrates of either CYP2C8 or CYP2B6.


Subject(s)
Acetates/pharmacokinetics , Bupropion/pharmacokinetics , Cytochrome P-450 CYP2B6/metabolism , Cytochrome P-450 CYP2C8/metabolism , Oxadiazoles/pharmacokinetics , Quinolines/pharmacokinetics , Acetates/blood , Adolescent , Adult , Bupropion/blood , Cyclopropanes , Cytochrome P-450 CYP2B6/biosynthesis , Cytochrome P-450 CYP2B6 Inducers/blood , Cytochrome P-450 CYP2B6 Inducers/pharmacokinetics , Cytochrome P-450 CYP2B6 Inducers/pharmacology , Cytochrome P-450 CYP2C8 Inhibitors/blood , Cytochrome P-450 CYP2C8 Inhibitors/pharmacokinetics , Cytochrome P-450 CYP2C8 Inhibitors/pharmacology , Drug Interactions , Healthy Volunteers , Hepatocytes/metabolism , Humans , Male , Middle Aged , Oxadiazoles/blood , Oxadiazoles/pharmacology , Quinolines/blood , Sulfides , Young Adult
11.
Eur J Drug Metab Pharmacokinet ; 43(6): 693-706, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29748821

ABSTRACT

BACKGROUND AND OBJECTIVES: The helicase-primase inhibitor amenamevir (ASP2151) is a novel therapeutic agent which has been approved for the treatment of herpes zoster. The present study examined the pharmacokinetic profile of amenamevir in rodents and compared it with data from the literature of past and current established therapies (acyclovir and valaciclovir) to provide additional data to facilitate drug discovery and proper drug use. METHODS: In situ absorption, blood and plasma radioactivity concentrations, tissue distribution, and excretion were determined using liquid scintillation counting. Plasma amenamevir concentrations were measured using a validated chromatographic method. Chemical structures of in vivo metabolites were investigated using liquid chromatography-mass spectrometry and nuclear magnetic resonance spectroscopy. RESULTS: Amenamevir, after single intravenous administration to mice, had an elimination half-life of 2 h. Bioavailability was 40% after single oral administration. In situ absorption data indicated that amenamevir is mainly absorbed in the small intestine. The main component in mouse plasma was amenamevir, accounting for 87.9% of amenamevir-derived components. Our results suggest that the main elimination pathway in mice is oxidative metabolism at a methyl group and a 1,2,3-trisubstituted benzene ring followed by biliary and fecal excretion. Following oral administration of 14C-amenamevir to mice, 100.63% of the dose (10.06% in urine and 90.46% in feces) was excreted by 96 h post-dose. CONCLUSIONS: The underlying mechanism of the improved pharmacokinetic profile of amenamevir was linked to an improved absorption ratio (not hepatic availability) compared to acyclovir, and qualitative differences in elimination (slow metabolism of amenamevir vs rapid urinary excretion of acyclovir/valaciclovir).


Subject(s)
Acyclovir/pharmacokinetics , Oxadiazoles/pharmacokinetics , Valacyclovir/pharmacokinetics , Acyclovir/blood , Animals , Biological Availability , Carbon Radioisotopes/pharmacokinetics , Male , Mice , Oxadiazoles/blood , Rats , Tissue Distribution , Valacyclovir/blood
12.
Clin Pharmacol Drug Dev ; 7(6): 634-640, 2018 08.
Article in English | MEDLINE | ID: mdl-29125718

ABSTRACT

Ozanimod (RPC1063) is an oral selective modulator of the sphingosine-1-phosphate 1 and 5 receptors under development for the treatment of relapsing multiple sclerosis and inflammatory bowel disease. The effects of high-fat and low-fat meals on the pharmacokinetics (PK) of a single oral dose of ozanimod were evaluated in 24 healthy volunteers in a randomized, open-label crossover trial. Each subject received a 1-mg dose of ozanimod hydrochloride under 3 meal conditions (fasted, high-fat, and low-fat), each separated by 7 days. Mean plasma concentration-time profiles for ozanimod and its active metabolites (RP101988 [major], RP101075 [minor]) were similar under all 3 conditions. Moreover, all PK parameters for ozanimod, RP101988, and RP101075 were similar under the 3 meal conditions. The 90% confidence intervals (CIs) for the ratios of geometric least-squares mean (fed/fasted) were within the equivalence limits of 0.80 to 1.25 for area under the concentration-time curve from time 0 to infinity (AUC0-∞ ) and maximum plasma concentration (Cmax ) for ozanimod, RP101988, and RP101075, except for the high-fat effect on RP101075 Cmax (90%CI, 0.76-0.88). Given this lack of a food effect on the exposure of ozanimod and its active metabolites, ozanimod can be taken without regard to meals.


Subject(s)
Diet, Fat-Restricted , Diet, High-Fat , Drug-Related Side Effects and Adverse Reactions/etiology , Food-Drug Interactions , Indans/pharmacokinetics , Oxadiazoles/pharmacokinetics , Receptors, Lysosphingolipid/metabolism , Administration, Oral , Adolescent , Adult , Cross-Over Studies , Female , Healthy Volunteers , Humans , Indans/administration & dosage , Indans/blood , Male , Middle Aged , Oxadiazoles/administration & dosage , Oxadiazoles/blood , Young Adult
13.
Neuropharmacology ; 125: 146-155, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28734868

ABSTRACT

Opicapone is a novel catechol-O-methyltransferase (COMT) inhibitor that emerged to fulfil the need of a safer and more efficacious COMT inhibitor. The present study was carried out in order to assess and compare the pharmacokinetics and pharmacodynamics (COMT inhibition) of opicapone after single and multiple oral administrations (30 mg/kg) to Wistar rats. For this purpose, at predefined time points up to 72 h post-dosing, blood, liver and kidneys were collected and, then, the concentrations of opicapone and its active metabolite (BIA 9-1079) were determined in plasma and in liver and kidney tissues, as well as the erythrocyte, liver and kidney COMT activity. No systemic, renal or hepatic accumulation of opicapone was observed following repeated administration. Furthermore, the tissue-systemic exposure relationships to opicapone suggested a low drug exposure in the liver and kidneys. After single-dosing, COMT inhibition profiles were reasonably comparable in all the studied matrices; although similar results were found after multiple-dosing, a higher degree of inhibition was observed, indicating a continuous peripheral COMT inhibition when opicapone is administered once-daily. Despite having a short elimination half-life (≤2.94 h), opicapone showed a strong and long-lasting COMT inhibition in both studies, since more than 50% of the COMT activity was still inhibited at 24 h post-dosing.


Subject(s)
Catechol O-Methyltransferase Inhibitors/pharmacology , Catechol O-Methyltransferase Inhibitors/pharmacokinetics , Oxadiazoles/pharmacology , Oxadiazoles/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Catechol O-Methyltransferase/metabolism , Catechol O-Methyltransferase Inhibitors/blood , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Erythrocytes/drug effects , Erythrocytes/enzymology , Kidney/drug effects , Kidney/enzymology , Liver/drug effects , Liver/enzymology , Male , Oxadiazoles/blood , Rats, Wistar
14.
J Clin Pharmacol ; 57(8): 988-996, 2017 08.
Article in English | MEDLINE | ID: mdl-28398597

ABSTRACT

The sphingosine-1-phosphate 1 receptor (S1P1R ) is expressed by lymphocytes, dendritic cells, and vascular endothelial cells and plays a role in the regulation of chronic inflammation and lymphocyte egress from peripheral lymphoid organs. Ozanimod is an oral selective modulator of S1P1R and S1P5R receptors in clinical development for the treatment of chronic immune-mediated, inflammatory diseases. This first-in-human study characterized the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of ozanimod in 88 healthy volunteers using a range of single and multiple doses (7 and 28 days) and a dose-escalation regimen. Ozanimod was generally well tolerated up to a maximum single dose of 3 mg and multiple doses of 2 mg/d, with no severe adverse events (AEs) and no dose-limiting toxicities. The most common ozanimod-related AEs included headache, somnolence, dizziness, nausea, and fatigue. Ozanimod exhibited linear PK, high steady-state volume of distribution (73-101 L/kg), moderate oral clearance (204-227 L/h), and an elimination half-life of approximately 17 to 21 hours. Ozanimod produced a robust dose-dependent reduction in total peripheral lymphocytes, with a median decrease of 65% to 68% observed after 28 days of dosing at 1 and 1.5 mg/d, respectively. Ozanimod selectivity affected lymphocyte subtypes, causing marked decreases in cells expressing CCR7 and variable decreases in subsets lacking CCR7. A dose-dependent negative chronotropic effect was observed following the first dose, with the dose-escalation regimen attenuating the first-dose negative chronotropic effect. Ozanimod safety, PK, and PD properties support the once-daily regimens under clinical investigation.


Subject(s)
Indans , Oxadiazoles , Adult , Double-Blind Method , Fasting/metabolism , Female , Healthy Volunteers , Humans , Indans/adverse effects , Indans/blood , Indans/pharmacokinetics , Indans/pharmacology , Lymphocyte Count , Lymphocytes/drug effects , Male , Middle Aged , Oxadiazoles/adverse effects , Oxadiazoles/blood , Oxadiazoles/pharmacokinetics , Oxadiazoles/pharmacology , Receptors, Lysosphingolipid/metabolism , Young Adult
15.
Toxicol Appl Pharmacol ; 323: 9-15, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28322896

ABSTRACT

Opicapone is a novel potent, reversible and purely peripheral catechol-O-methyltransferase inhibitor that has been developed to be used as an adjunct to levodopa/aromatic L-amino acid decarboxylase inhibitor therapy for Parkinson's disease. Thus, this study aimed to compare the plasma pharmacokinetics of opicapone and its active metabolite (BIA 9-1079) after the administration of single and multiple oral doses to rats. Wistar rats (n=8 per group) were orally treated with single (30, 60 or 90mg/kg) or multiple (30mg/kg once-daily for seven consecutive days) oral doses of opicapone. Blood samples were collected up to 24h post-dosing through a cannula introduced in the tail vein of rats. After quantifying opicapone and BIA 9-1079 in plasma, a non-compartmental pharmacokinetic analysis was performed. Opicapone was quickly absorbed (time to reach the maximum plasma concentration≤2h) in both dosage regimens and the extent of systemic exposure to opicapone increased approximately in a dose-proportional manner after single-dosing within the studied dose range (30-90mg/kg). Opicapone and BIA 9-1079 showed a relatively short plasma elimination half-life (1.58-4.50h) and a small systemic accumulation after multiple-dosing. Hence, no pharmacokinetic concerns are expected when opicapone is administered with a once-daily dosing regimen.


Subject(s)
Catechol O-Methyltransferase Inhibitors/administration & dosage , Catechol O-Methyltransferase Inhibitors/pharmacokinetics , Oxadiazoles/administration & dosage , Oxadiazoles/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Catechol O-Methyltransferase Inhibitors/blood , Catechol O-Methyltransferase Inhibitors/toxicity , Drug Administration Schedule , Gastrointestinal Absorption , Half-Life , Male , Metabolic Clearance Rate , Models, Biological , Oxadiazoles/blood , Oxadiazoles/toxicity , Rats, Wistar , Risk Assessment
16.
Br J Clin Pharmacol ; 83(3): 540-553, 2017 03.
Article in English | MEDLINE | ID: mdl-27763682

ABSTRACT

AIMS: To compare the levodopa/carbidopa (LC) and levodopa/benserazide (LB) pharmacokinetic profiles following repeated doses of opicapone (OPC) administered apart from levodopa. METHODS: Two randomized, double blind, sex-balanced, placebo-controlled studies in four groups of 12 or 18 healthy subjects each. In each group, enrolled subjects received a once-daily morning (5, 15 and 30 mg) or evening (5, 15 and 50 mg) administration of OPC or placebo for up to 28 days. On the morning of Day 11, 12 h after the OPC or placebo evening dose, or the morning of Day 21, 1 h after the OPC or placebo dose, a single dose of immediate-release 100/25 mg LC was administered. Similarly, on Day 18 morning, 12 h after the OPC or placebo evening dose, or Day 28 morning, 1 h after the OPC or placebo dose, a single dose of immediate-release 100/25 mg LB was administered. RESULTS: All OPC treatments, in relation to the placebo group, presented a higher extent of exposure (AUC) to levodopa following either LC or LB doses. A relevant but not dose-dependent increase in the levodopa AUC occurred with all OPC dose groups in relation to placebo. All active treatments significantly inhibited both peak (Emax ) and extent (AUEC) of the catechol-O-methyltransferase activity in relation to placebo. The tolerability profile was favourable. CONCLUSION: Opicapone, as once-daily oral evening regimen and/or 1 h apart from levodopa therapy, increases the bioavailability of levodopa associated with its pronounced, long-lasting and sustained catechol-O-methyltransferase inhibition. The tolerability profile was favourable and similar between OPC and placebo.


Subject(s)
Benserazide/pharmacokinetics , Levodopa/pharmacokinetics , Oxadiazoles/pharmacology , Oxadiazoles/pharmacokinetics , Adult , Antiparkinson Agents/pharmacokinetics , Benserazide/adverse effects , Benserazide/blood , Benserazide/pharmacology , Biological Availability , Carbidopa/adverse effects , Carbidopa/pharmacology , Catechol O-Methyltransferase Inhibitors/adverse effects , Catechol O-Methyltransferase Inhibitors/blood , Catechol O-Methyltransferase Inhibitors/pharmacokinetics , Catechol O-Methyltransferase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Double-Blind Method , Drug Combinations , Female , Humans , Levodopa/adverse effects , Levodopa/blood , Levodopa/pharmacology , Male , Middle Aged , Oxadiazoles/adverse effects , Oxadiazoles/blood
17.
Schizophr Res ; 172(1-3): 152-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26922656

ABSTRACT

INTRODUCTION: Activation of metabotropic glutamate (mGluR2/3) receptors has been proposed as an alternative mechanism to dopaminergic-based antipsychotics to correct glutamatergic deficits hypothesized to underlie schizophrenia symptoms. This study investigates the efficacy and safety of AZD8529, a selective positive allosteric modulator (PAM) at the mGlu2 receptor, in symptomatic patients with schizophrenia. METHODS: Patients were randomized to receive AZD8529 40 mg, risperidone 4 mg, or placebo as monotherapy. Treatment lasted for 28 days, and clinical efficacy was assessed using Positive and Negative Syndrome Scale (PANSS) and Clinical Global Impression (CGI) scores. RESULTS: There were no significant differences between patients treated with AZD8529 versus placebo in change from baseline to endpoint in PANSS total, negative and positive symptom subscale, or CGI-S scores. In contrast, risperidone demonstrated significant efficacy relative to placebo. CONCLUSION: These results do not support a role for the mGluR-2 PAM AZD8529 as an antipsychotic and indicate that positive modulation of mGluR type 2 receptors alone is not sufficient for antipsychotic effects in acutely ill schizophrenia patients.


Subject(s)
Antipsychotic Agents/therapeutic use , Indoles/therapeutic use , Oxadiazoles/therapeutic use , Receptors, Metabotropic Glutamate/metabolism , Schizophrenia/drug therapy , Adolescent , Adult , Allosteric Regulation , Antipsychotic Agents/adverse effects , Antipsychotic Agents/blood , Double-Blind Method , Female , Humans , Indoles/adverse effects , Indoles/blood , Male , Middle Aged , Oxadiazoles/adverse effects , Oxadiazoles/blood , Psychiatric Status Rating Scales , Risperidone/adverse effects , Risperidone/blood , Risperidone/therapeutic use , Schizophrenia/metabolism , Treatment Outcome , Young Adult
18.
J Pediatric Infect Dis Soc ; 5(1): 53-62, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26407253

ABSTRACT

BACKGROUND: Neonatal enterovirus sepsis has high mortality. Antiviral therapy is not available. METHODS: Neonates with suspected enterovirus sepsis (hepatitis, coagulopathy, and/or myocarditis) with onset at ≤15 days of life were randomized 2:1 to receive oral pleconaril or placebo for 7 days. Serial virologic (oropharynx, rectum, urine, serum), clinical, pharmacokinetic, and safety evaluations were performed. RESULTS: Sixty-one subjects were enrolled (43 treatment, 18 placebo), of whom 43 were confirmed enterovirus infected (31 treatment, 12 placebo). There was no difference in day 5 oropharyngeal culture positivity (primary endpoint; 0% in both groups). However, enterovirus-infected subjects in the treatment group became culture negative from all anatomic sites combined faster than placebo group subjects (median 4.0 versus 7.0 days, P = .08), and fewer subjects in the treatment group remained polymerase chain reaction (PCR)-positive from the oropharynx when last sampled (23% versus 58%, P = .02; median, 14.0 days). By intent to treat, 10/43 (23%) subjects in the treatment group and 8/18 (44%) in the placebo group died (P = .02 for 2-month survival difference); among enterovirus-confirmed subjects, 7/31 (23%) in the treatment group died versus 5/12 (42%) in the placebo group (P = .26). All pleconaril recipients attained concentrations greater than the IC90 after the first study day, but 38% were less than the IC90 during the first day of treatment. One subject in the treatment group and three in the placebo group had treatment-related adverse events. CONCLUSIONS: Shorter times to culture and PCR negativity and greater survival among pleconaril recipients support potential efficacy and warrant further evaluation.


Subject(s)
Antiviral Agents/therapeutic use , Enterovirus Infections/complications , Enterovirus Infections/drug therapy , Enterovirus/drug effects , Neonatal Sepsis/drug therapy , Neonatal Sepsis/virology , Oxadiazoles/therapeutic use , Antiviral Agents/blood , Antiviral Agents/pharmacokinetics , Antiviral Agents/urine , Double-Blind Method , Enterovirus/genetics , Enterovirus/isolation & purification , Enterovirus Infections/blood , Enterovirus Infections/urine , Female , Humans , Infant , Infant, Newborn , Male , Neonatal Sepsis/blood , Neonatal Sepsis/urine , Oropharynx/virology , Oxadiazoles/blood , Oxadiazoles/pharmacokinetics , Oxadiazoles/urine , Oxazoles , Rectum/virology
19.
Clin Pharmacokinet ; 55(5): 595-604, 2016 May.
Article in English | MEDLINE | ID: mdl-26507722

ABSTRACT

BACKGROUND AND OBJECTIVE: Azilsartan medoxomil (AZL-M) is an angiotensin II receptor blocker approved to treat hypertension. After oral dosing, AZL-M is quickly hydrolyzed to azilsartan (AZL). The aims of this study were to assess the effects of age, sex, and race on the pharmacokinetics of AZL-M in healthy subjects, as well as safety and tolerability. METHODS: Sixty-one healthy adults were enrolled in this phase I, single-blind, randomized placebo-controlled study (placebo control was for assessment of safety/tolerability only). Subjects were stratified by age (18-45 vs. 65-85 years), sex, and race (black vs. white) and given oral AZL-M 60 mg (3 × 20 mg capsules) or placebo as a single dose (Day 1) and consecutive daily doses (Days 4-8) (6:2 ratio for AZL-M:placebo per group). Pharmacokinetics were evaluated (AZL-M patients only) on Days 1-3 and 8-9 and safety/tolerability was monitored. RESULTS: Age, sex, and race had no clinically meaningful effect on AZL exposures after single or multiple dosing. Pharmacokinetic parameters remained similar between Days 1 and 8 for each age, sex, and race subgroup. The frequency of adverse events was similar for AZL-M (32%) and placebo (29%). No discontinuations or serious adverse events occurred. CONCLUSIONS: Based on these pharmacokinetic and safety/tolerability findings, no AZL-M dose adjustments are required based on age, sex, or race (black/white).


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Benzimidazoles/pharmacokinetics , Oxadiazoles/pharmacokinetics , Adolescent , Adult , Aged , Aged, 80 and over , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin II Type 1 Receptor Blockers/adverse effects , Angiotensin II Type 1 Receptor Blockers/blood , Benzimidazoles/administration & dosage , Benzimidazoles/adverse effects , Benzimidazoles/blood , Female , Healthy Volunteers , Humans , Male , Middle Aged , Oxadiazoles/administration & dosage , Oxadiazoles/adverse effects , Oxadiazoles/blood , Racial Groups , Single-Blind Method , Young Adult
20.
J Clin Pharmacol ; 56(8): 988-98, 2016 08.
Article in English | MEDLINE | ID: mdl-26632101

ABSTRACT

Population pharmacokinetic and exposure-response models for azilsartan medoxomil (AZL-M) and chlorthalidone (CLD) were developed using data from an 8-week placebo-controlled phase 3, factorial study of 20, 40, and 80 mg AZL-M every day (QD) and 12.5 and 25 mg CLD QD in fixed-dose combination (FDC) in subjects with moderate to severe essential hypertension. A 2-compartment model with first-order absorption and elimination was developed to describe pharmacokinetics. An Emax model for exposure-response analysis evaluated AZL-M/CLD effects on ambulatory systolic blood pressure (SBP). Estimated oral clearance and apparent volume of distribution (central compartment) were 1.47 L/h and 3.98 L for AZL, and 4.13 L/h and 62.1 L for CLD. Age as a covariate had the largest effect on AZL and CLD exposure (±20% change). Predicted maximal SBP responses (Emax ) were -15.6 and -23.9 mm Hg for AZL and CLD. Subgroup analysis identified statistically significant Emax differences for black vs nonblack subjects, whereby the reduced AZL response in black subjects was offset by greater response to CLD. The estimated Emax for AZL and CLD was generally greater in subjects with higher baseline BP. In conclusion, no dose adjustments to AZL-M or CLD are warranted based on identified covariates, and antihypertensive efficacy of AZL-M/CLD combination therapy is comparable in black and nonblack subjects.


Subject(s)
Benzimidazoles/administration & dosage , Benzimidazoles/blood , Chlorthalidone/administration & dosage , Chlorthalidone/blood , Hypertension/blood , Hypertension/drug therapy , Oxadiazoles/administration & dosage , Oxadiazoles/blood , Aged , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin II Type 1 Receptor Blockers/blood , Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/blood , Antihypertensive Agents/pharmacokinetics , Benzimidazoles/pharmacokinetics , Chlorthalidone/pharmacokinetics , Double-Blind Method , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Oxadiazoles/pharmacokinetics , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...