Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 587
Filter
1.
J Pediatr Gastroenterol Nutr ; 78(6): 1342-1354, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38644678

ABSTRACT

BACKGROUND: The safety and efficacy of sofosbuvir-velpatasvir in children aged 3-17 years with chronic hepatitis C virus (HCV) infection of any genotype were evaluated. METHODS: In this Phase 2, multicenter, open-label study, patients received once daily for 12 weeks either sofosbuvir-velpatasvir 400/100 mg tablet (12-17 years), 200/50 mg low dose tablet or oral granules (3-11 years and ≥17 kg), or 150/37.5 mg oral granules (3-5 years and <17 kg). The efficacy endpoint was sustained virologic response 12 weeks after therapy (SVR12). Dose appropriateness was confirmed by intensive pharmacokinetics in each age group. FINDINGS: Among 216 patients treated, 76% had HCV genotype 1% and 12% had genotype 3. Rates of SVR12 were 83% (34/41) among 3-5-year-olds, 93% (68/73) among 6-11-year-olds, and 95% (97/102) among 12-17-year-olds. Only two patients experienced virologic failure. The most common adverse events were headache, fatigue, and nausea in 12-17-year-olds; vomiting, cough, and headache in 6-11-year-olds; and vomiting in 3-5-year-olds. Three patients discontinued treatment because of adverse events. Four patients had serious adverse events; all except auditory hallucination (n = 1) were considered unrelated to study drug. Exposures of sofosbuvir, its metabolite GS-331007, and velpatasvir were comparable to those in adults in prior Phase 2/3 studies. Population pharmacokinetic simulations supported weight-based dosing for children in this age range. INTERPRETATION: The pangenotypic regimen of sofosbuvir-velpatasvir is highly effective and safe in treating children 3-17 years with chronic HCV infection.


Subject(s)
Antiviral Agents , Carbamates , Drug Combinations , Hepatitis C, Chronic , Heterocyclic Compounds, 4 or More Rings , Sofosbuvir , Humans , Sofosbuvir/therapeutic use , Sofosbuvir/pharmacokinetics , Sofosbuvir/administration & dosage , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Heterocyclic Compounds, 4 or More Rings/adverse effects , Child , Carbamates/therapeutic use , Carbamates/pharmacokinetics , Carbamates/adverse effects , Carbamates/administration & dosage , Male , Child, Preschool , Female , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacokinetics , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Adolescent , Hepatitis C, Chronic/drug therapy , Treatment Outcome , Hepacivirus/genetics , Hepacivirus/drug effects , Sustained Virologic Response , Genotype , Benzimidazoles , Benzopyrans
2.
AAPS PharmSciTech ; 25(6): 161, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38992175

ABSTRACT

Drug solubility and dissolution remain a significant challenge in pharmaceutical formulations. This study aimed to formulate and evaluate repanglinide (RPG) nanosuspension-based buccal fast-dissolving films (BDFs) for dissolution enhancement. RPG nanosuspension was prepared by the antisolvent-precipitation method using multiple hydrophilic polymers, including soluplus®, polyvinyl alcohol, polyvinyl pyrrolidine, poloxamers, and hydroxyl propyl methyl cellulose. The nanosuspension was then directly loaded into BDFs using the solvent casting technique. Twelve formulas were prepared with a particle size range of 81.6-1389 nm and PDI 0.002-1 for the different polymers. Nanosuspensions prepared with soluplus showed a favored mean particle size of 82.6 ± 3.2 nm. The particles were spherical and non-aggregating, as demonstrated by SEM imaging. FTIR showed no interaction between soluplus and RPG. Faster dissolution occurred for the nanosuspension in comparison with pure RPG (complete release vs 60% within 30 min). The nanosuspension was successfully incorporated into BDFs. The optimum film formula showed 28 s disintegration time, and 97.3% RPG released within 10 min. Ex-vivo permeation profiles revealed improved RPG nanosuspension permeation with the cumulative amount of RPG permeated is103.4% ± 10.1 and a flux of 0.00275 mg/cm2/min compared to 39.3% ± 9.57 and a flux of 0.001058 mg/cm2/min for pure RPG. RPG was successfully formulated into nanosuspension that boosted drug dissolution and permeation. The selection of the ultimate NP formula was driven by optimal particle size, distribution, and drug content. Soluplus NPs were shown to be the successful formulations, which were further incorporated into a buccal film. The film was evaluated for ex-vivo permeation, confirming successful RPG formulation with improved performance compared to pure drugs.


Subject(s)
Carbamates , Nanoparticles , Particle Size , Piperidines , Solubility , Suspensions , Nanoparticles/chemistry , Piperidines/chemistry , Piperidines/administration & dosage , Piperidines/pharmacokinetics , Carbamates/chemistry , Carbamates/administration & dosage , Carbamates/pharmacokinetics , Animals , Chemistry, Pharmaceutical/methods , Drug Liberation , Polyvinyls/chemistry , Polymers/chemistry , Administration, Buccal , Polyethylene Glycols/chemistry , Drug Compounding/methods
3.
Br J Clin Pharmacol ; 89(2): 874-886, 2023 02.
Article in English | MEDLINE | ID: mdl-36098470

ABSTRACT

To evaluate the pharmacokinetic effects of SHR3680 on repaglinide and bupropion and its metabolite hydroxybupropion. METHODS: A single-centre, open-label, single-arm, fixed-sequence clinical trial in 18 patients with prostate cancer. RESULTS: After a single oral dose of 0.5 mg repaglinide and SHR3680, geometric mean peak plasma concentration (Cmax ) of plasma repaglinide was 14.240 and 5.887 ng/mL, geometric mean area under the concentration-time curve (AUC0-t )was 20.577 and 7.320 h ng/mL, geometric mean AUC0-∞ was 20.949 and 7.451 h ng/mL, mean half-life (t1/2 ) was 1.629 and 1.195 hours, and geometric mean oral clearance (CL/F) was 23.867 and 67.107 L/h, respectively. After a single oral administration of 150 mg bupropion and SHR3680, geometric mean Cmax of plasma bupropion was 85.430 and 33.747 ng/mL, geometric mean AUC0-t was 1003.896 and 380.158 h ng/mL, geometric mean AUC0-∞ was 1038.054 and 401.387 h ng/mL, mean t1/2 was 22.533 and 17.733 hours, and geometric mean CL/F was 144.501 and 373.705 L/h, respectively. The plasma geometric mean Cmax of its main active metabolic hydroxybupropion was 268.113 and 177.318 ng/mL, geometric mean AUC0-t was 14 283.087 and 5420.219 h ng/mL, geometric mean AUC0-∞ was 15 218.158 and 5364.625 h ng/mL, mean t1/2 were 36.069 and 16.688 hours, and geometric mean CL/F was 8.623 L/h and 27.961 L/h, respectively. CONCLUSION: Coadministration of SHR3680 with repaglinide or bupropion significantly shortened the elimination half-lives, significantly increased the apparent clearance rate, and significantly decreased the in vivo exposure of repaglinide, bupropion and hydroxybupropion compared with single administration of repaglinide or bupropion.


Subject(s)
Bupropion , Prostatic Neoplasms , Humans , Male , Area Under Curve , Carbamates/pharmacokinetics , Cross-Over Studies
4.
Molecules ; 28(1)2022 Dec 22.
Article in English | MEDLINE | ID: mdl-36615272

ABSTRACT

The combination regimen targeting BRAF and MEK inhibition, for instance, encorafenib (Braftovi™, ENF) plus binimetinib (Mektovi®, BNB), are now recommended as first-line treatment in patients with unresectable or metastatic melanoma with a BRAF V600-activating mutation. Patients treated with combination therapy of ENF and BNB demonstrated a delay in resistance development, increases in antitumor activity, and attenuation of toxicities compared with the activity of either agent alone. However, the pharmacokinetic profile of the FDA-approved ENF and BNB is still unclear. In this study, a rapid and sensitive LC-MS/MS bioanalytical method for simultaneous quantification of ENF and BNB in rat plasma was developed and validated. Chromatography was performed on an Agilent Eclipse plus C18 column (50 mm × 2.1 mm, 1.8 µm), with an isocratic mobile phase composed of 0.1% formic acid in water/acetonitrile (67:33, v/v, pH 3.2) at a flow rate of 0.35 mL/min. A positive multiple reaction monitoring (MRM) mode was chosen for detection and the process of analysis was run for 2 min. Plasma samples were pre-treated using protein precipitation with acetonitrile containing spebrutinib as the internal standard (IS). Method validation was assessed as per the FDA guidelines for the determination of ENF and BNB over concentration ranges of 0.5-3000 ng/mL (r2 ≥ 0.997) for each drug (plasma). The lower limits of detection (LLOD) for both drugs were 0.2 ng/mL. The mean relative standard deviation (RSD) of the results for accuracy and precision was ≤ 7.52%, and the overall recoveries of ENF and BNB from rat plasma were in the range of 92.88-102.28%. The newly developed approach is the first LC-MS/MS bioanalytical method that can perform simultaneous quantification of ENF and BNB in rat plasma and its application to a pharmacokinetic study. The mean result for Cmax for BNB and ENF was found to be 3.43 ± 0.46 and 16.42 ± 1.47 µg/mL achieved at 1.0 h for both drugs, respectively. The AUC0-∞ for BNB and ENF was found to be 18.16 ± 1.31 and 36.52 ± 3.92 µg/mL.h, respectively. On the other hand, the elimination half-life (t1/2kel) parameters for BNB and ENF in the rat plasma were found to be 3.39 ± 0.43 h and 2.48 ± 0.24 h, and these results are consistent with previously reported values.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Benzimidazoles , Carbamates , Melanoma , Sulfonamides , Tandem Mass Spectrometry , Animals , Rats , Chromatography, Liquid/methods , Proto-Oncogene Proteins B-raf/metabolism , Reproducibility of Results , Tandem Mass Spectrometry/methods , Carbamates/blood , Carbamates/pharmacokinetics , Sulfonamides/blood , Sulfonamides/pharmacokinetics , Benzimidazoles/blood , Benzimidazoles/pharmacokinetics , Melanoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/blood , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics
5.
J Pharmacol Exp Ther ; 376(2): 306-321, 2021 02.
Article in English | MEDLINE | ID: mdl-33203660

ABSTRACT

Integrated in silico chemical clustering and melatonin receptor molecular modeling combined with in vitro 2-[125I]-iodomelatonin competition binding were used to identify carbamate insecticides with affinity for human melatonin receptor 1 (hMT1) and human melatonin receptor 2 (hMT2). Saturation and kinetic binding studies with 2-[125I]-iodomelatonin revealed lead carbamates (carbaryl, fenobucarb, bendiocarb, carbofuran) to be orthosteric ligands with antagonist apparent efficacy at hMT1 and agonist apparent efficacy at hMT2 Furthermore, using quantitative receptor autoradiography in coronal brain slices from C3H/HeN mice, carbaryl, fenobucarb, and bendiocarb competed for 2-[125I]-iodomelatonin binding in the suprachiasmatic nucleus (SCN), paraventricular nucleus of the thalamus (PVT), and pars tuberalis (PT) with affinities similar to those determined for the hMT1 receptor. Carbaryl (10 mg/kg i.p.) administered in vivo also competed ex vivo for 2-[125I]-iodomelatonin binding to the SCN, PVT, and PT, demonstrating the ability to reach brain melatonin receptors in C3H/HeN mice. Furthermore, the same dose of carbaryl given to C3H/HeN mice in constant dark for three consecutive days at subjective dusk (circadian time 10) phase-advanced circadian activity rhythms (mean = 0.91 hours) similar to melatonin (mean = 1.12 hours) when compared with vehicle (mean = 0.04 hours). Carbaryl-mediated phase shift of overt circadian activity rhythm onset is likely mediated via interactions with SCN melatonin receptors. Based on the pharmacological actions of carbaryl and other carbamate insecticides at melatonin receptors, exposure may modulate time-of-day information conveyed to the master biologic clock relevant to adverse health outcomes. SIGNIFICANCE STATEMENT: In silico chemical clustering and molecular modeling in conjunction with in vitro bioassays identified several carbamate insecticides (i.e., carbaryl, carbofuran, fenobucarb, bendiocarb) as pharmacologically active orthosteric melatonin receptor 1 and 2 ligands. This work further demonstrated that carbaryl competes for melatonin receptor binding in the master biological clock (suprachiasmatic nucleus) and phase-advances overt circadian activity rhythms in C3H/HeN mice, supporting the relevance of circadian effects when interpreting toxicological findings related to carbamate insecticide exposure.


Subject(s)
Carbamates/pharmacology , Insecticides/pharmacology , Receptors, Melatonin/metabolism , Animals , Binding Sites , Brain/drug effects , Brain/metabolism , CHO Cells , Carbamates/pharmacokinetics , Circadian Rhythm/drug effects , Cricetinae , Cricetulus , Humans , Insecticides/pharmacokinetics , Male , Mice , Molecular Docking Simulation , Protein Binding , Receptors, Melatonin/chemistry
6.
Drug Metab Dispos ; 49(7): 530-539, 2021 07.
Article in English | MEDLINE | ID: mdl-33958385

ABSTRACT

Accurately predicting the pharmacokinetics of compounds that are transporter substrates has been notoriously challenging using traditional in vitro systems and physiologically based pharmacokinetic (PBPK) modeling. The objective of this study was to use PBPK modeling to understand the translational accuracy of data generated with human embryonic kidney 293 (HEK293) cells overexpressing the hepatic uptake transporters organic anion transporting polypeptide (OATP) 1B1/3 with and without plasma while accounting for transporter expression. Models of four OATP substrates, two with low protein binding (pravastatin and rosuvastatin) and two with high protein binding (repaglinide and pitavastatin) were explored, and the OATP in vitro data generated in plasma incubations were used for a plasma model, and in buffer incubations for a buffer model. The pharmacokinetic parameters and concentration-time profiles of pravastatin and rosuvastatin were similar and well predicted (within 2-fold of observed values) using the plasma and buffer models without needing an empirical scaling factor, whereas the dispositions of the highly protein bound repaglinide and pitavastatin were more accurately simulated with the plasma models than the buffer models. This work suggests that data from HEK293 overexpressing transporter cells corrected for transporter expression represent a valid approach to improve bottom-up PBPK modeling for highly protein bound OATP substrates with plasma incubations and low protein binding OATP substrates with or without plasma incubations. SIGNIFICANCE STATEMENT: This work demonstrates the bottom-up approach of using in vitro data directly without employing empirical scaling factors to predict the intravenous pharmacokinetic (PK) profiles reasonably well for four organic anion transporting polypeptide (OATP) substrates. Based on these results, using HEK293 overexpressing cells, examining the impact of plasma for highly bound compounds, and incorporating transporter quantitation for the lot in which the in vitro data were generated represents a valid approach to achieve more accurate prospective PK predictions for OATP substrates.


Subject(s)
Drug Discovery/methods , Liver-Specific Organic Anion Transporter 1/metabolism , Models, Biological , Plasma/metabolism , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , Administration, Intravenous , Carbamates/administration & dosage , Carbamates/pharmacokinetics , HEK293 Cells , Humans , Piperidines/administration & dosage , Piperidines/pharmacokinetics , Pravastatin/administration & dosage , Pravastatin/pharmacokinetics , Quinolines/administration & dosage , Quinolines/pharmacokinetics , Rosuvastatin Calcium/administration & dosage , Rosuvastatin Calcium/pharmacokinetics
7.
J Sep Sci ; 44(19): 3691-3699, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34347375

ABSTRACT

Two valine carbamate prodrugs of daidzein were designed to improve its bioavailability. To compare the pharmacokinetic behavior of these prodrugs with different protected phenolic hydroxyl groups of daidzein, a rapid and sensitive method for simultaneous quantification of daidzein, its valine carbamate prodrug, and daidzein-7-O-glucuronide in rat plasma was developed and validated in this study. The samples were processed using a fast one-step protein precipitation method with methanol added to 50 µL of plasma and were analyzed by ultra-high performance liquid chromatography with tandem mass spectrometry. To improve the selectivity, peak shape, and peak elution, several key factors, especially stationary phase and the composition of the mobile phase, were tested, and the analysis was performed using the Kinetex® C18 column (100 × 2.1 mm, 2.6 µm) within only 2.6 min under optimal conditions. The established method exhibited good linearity over the concentration range of 2.0-1000 ng/mL for daidzein, and 8.0-4000 ng/mL for the prodrug and daidzein-7-O-glucuronide. The accuracy of the quality control samples was between 95.5 and 110.2% with satisfactory intra- and interday precision (relative standard deviation values < 10.85%), respectively. This sensitive, rapid, low-cost, and high-throughput method was successfully applied to compare the pharmacokinetic behavior of different daidzein carbamate prodrugs.


Subject(s)
Chromatography, High Pressure Liquid/methods , Glucuronides/blood , Isoflavones/blood , Prodrugs/analysis , Tandem Mass Spectrometry/methods , Animals , Carbamates/blood , Carbamates/chemistry , Carbamates/pharmacokinetics , Glucuronides/chemistry , Glucuronides/pharmacokinetics , Isoflavones/chemistry , Isoflavones/pharmacokinetics , Linear Models , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Rats , Reproducibility of Results , Sensitivity and Specificity , Valine/blood , Valine/chemistry , Valine/pharmacokinetics
8.
Biomed Chromatogr ; 35(8): e5119, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33749889

ABSTRACT

A valine carbamate prodrug of naringenin (NAR) called 4'V was synthesized to enhance its oral bioavailability because of low water solubility and poor membrane permeability of NAR. This study developed and fully validated a sensitive, rapid, and robust HPLC-MS/MS method for the simultaneous determination of NAR and 4'V in plasma. The analytes were treated using liquid-liquid extraction, separated on a Phenomenex Kinetex XB-C18 column, and detected using a triple-quadrupole tandem mass spectrometer equipped with an electrospray ionization interface. The analytes were eluted within only 4 min by gradient procedure. The excellent linear correlations were validated over the range of 4-400 ng/mL (r = 0.9990) for NAR and 2-2000 ng/mL (r = 0.9951) for 4'V, with lower limits of quantification of 4 and 2 ng/mL, respectively. For all quality control samples, the intra-day and inter-day precision and accuracy were within ±15%. The validated method was economical, high throughput, and reliable and was first successfully applied to a pharmacokinetic study of NAR and 4'V after oral administration to Sprague-Dawley rats. The results of the pharmacokinetic study demonstrated that the idea of amino acid carbamate prodrug is a promising strategy to improve the bioavailability of NAR.


Subject(s)
Carbamates/blood , Chromatography, High Pressure Liquid/methods , Flavanones/blood , Tandem Mass Spectrometry/methods , Valine/blood , Animals , Biological Availability , Carbamates/chemistry , Carbamates/pharmacokinetics , Flavanones/chemistry , Flavanones/pharmacokinetics , Linear Models , Male , Prodrugs , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity , Valine/chemistry , Valine/pharmacokinetics
9.
Int J Mol Sci ; 22(7)2021 Mar 25.
Article in English | MEDLINE | ID: mdl-33806023

ABSTRACT

We recently reported a new class of carbamate derivatives as anticonvulsants. Among these, 3-methylpentyl(4-sulfamoylphenyl)carbamate (MSPC) stood out as the most potent compound with ED50 values of 13 mg/kg (i.p.) and 28 mg/kg (p.o.) in the rat maximal electroshock test (MES). 3-Methylpropyl(4-sulfamoylphenyl)carbamate (MBPC), reported and characterized here, is an MSPC analogous compound with two less aliphatic carbon atoms in its structure. As both MSPC and MBPC are chiral compounds, here, we studied the carbonic anhydrase inhibitory and anticonvulsant action of both MBPC enantiomers in comparison to those of MSPC as well as their pharmacokinetic properties. Racemic-MBPC and its enantiomers showed anticonvulsant activity in the rat maximal electroshock (MES) test with ED50 values in the range of 19-39 mg/kg. (R)-MBPC had a 65% higher clearance than its enantiomer and, consequently, a lower plasma exposure (AUC) than (S)-MSBC and racemic-MSBC. Nevertheless, (S)-MBPC had a slightly better brain permeability than (R)-MBPC with a brain-to-plasma (AUC) ratio of 1.32 (S-enantiomer), 1.49 (racemate), and 1.27 (R-enantiomer). This may contribute to its better anticonvulsant-ED50 value. The clearance of MBPC enantiomers was more enantioselective than the brain permeability and MES-ED50 values, suggesting that their anticonvulsant activity might be due to multiple mechanisms of action.


Subject(s)
Carbamates/chemistry , Central Nervous System/drug effects , Animals , Anticonvulsants/pharmacology , Area Under Curve , Brain/drug effects , Carbamates/pharmacokinetics , Carbonic Anhydrases/chemistry , Electroshock , Male , Protein Isoforms , Rats , Rats, Sprague-Dawley , Seizures/drug therapy , Solvents , Stereoisomerism
10.
Article in English | MEDLINE | ID: mdl-32015036

ABSTRACT

The purpose of this study was to evaluate the pharmacokinetics of ritonavir-boosted fosamprenavir during pregnancy and postpartum. Amprenavir (the active moiety of fosamprenavir) and ritonavir intensive pharmacokinetic evaluations were performed at steady state during the second and third trimesters of pregnancy and postpartum. Plasma concentrations of amprenavir and ritonavir were measured using high-performance liquid chromatography. The target amprenavir area under the concentration-versus-time curve (AUC) was higher than the 10th percentile (27.7 µg · h/ml) of the median area under the curve for ritonavir-boosted fosamprenavir in adults receiving twice-daily fosamprenavir-ritonavir at 700 mg/100 mg. Twenty-nine women were included in the analysis. The amprenavir AUC from time zero to 12 h (AUC0-12) was lower (geometric mean ratio [GMR], 0.60 [confidence interval {CI}, 0.49 to 0.72] [P < 0.001]) while its apparent oral clearance was higher (GMR, 1.68 [CI, 1.38 to 2.03] [P < 0.001]) in the third trimester than postpartum. Similarly, the ritonavir AUC0-12 was lower in the second (GMR, 0.51 [CI, 0.28 to 0.91] [P = 0.09]) and third (GMR, 0.72 [CI, 0.55 to 0.95] [P = 0.005]) trimesters than postpartum, while its apparent oral clearance was higher in the second (GMR, 1.98 [CI, 1.10 to 3.56] [P = 0.06]) and third (GMR, 1.38 [CI, 1.05 to 1.82] [P = 0.009]) trimesters than postpartum. The amprenavir area under the curve exceeded the target for 6/8 (75%) women in the 2nd trimester, 18/28 (64%) in the 3rd trimester, and 19/22 (86.4%) postpartum, and the trough concentrations (Cmin) of amprenavir were 4- to 16-fold above the mean amprenavir-protein-adjusted 50% inhibitory concentration (IC50) of 0.146 µg/ml. Although amprenavir plasma concentrations in women receiving ritonavir-boosted fosamprenavir were lower during pregnancy than postpartum, the reduced amprenavir concentrations were still above the exposures needed for viral suppression.


Subject(s)
Carbamates/pharmacokinetics , Furans/pharmacokinetics , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacokinetics , Pregnancy Complications, Infectious/drug therapy , Ritonavir/pharmacokinetics , Sulfonamides/pharmacokinetics , Adult , Area Under Curve , Carbamates/adverse effects , Female , Furans/adverse effects , HIV Protease Inhibitors/adverse effects , Humans , Maternal Age , Pregnancy , Pregnancy Trimesters , RNA, Viral/blood , Ritonavir/adverse effects , Sulfonamides/adverse effects , Viral Load
11.
Drug Metab Dispos ; 48(12): 1312-1320, 2020 12.
Article in English | MEDLINE | ID: mdl-32978223

ABSTRACT

Revefenacin inhalation solution is an anticholinergic indicated for the maintenance treatment of patients with chronic obstructive pulmonary disease. Mass balance, pharmacokinetics, and metabolism of revefenacin were evaluated after intravenous and oral administration of [14C]-revefenacin in healthy subjects. Pharmacological activity of the major revefenacin metabolite was also assessed. Adult males (n = 9) received 20 µg intravenously of approximately 1 µCi [14C]-revefenacin and/or a single 200-µg oral solution of approximately 10 µCi [14C]-revefenacin. Mean recovery of radioactive material was 81.4% after intravenous administration (54.4% in feces; 27.1% in urine) and 92.7% after oral dosing (88.0% in feces, 4.7% in urine). Mean absolute bioavailability of oral revefenacin was low (2.8%). Intact revefenacin accounted for approximately 52.1% and 13.1% of the total radioactivity in plasma after intravenous and oral administration, respectively. Two main circulating metabolites were observed in plasma. After an intravenous dose, a hydrolysis product, THRX-195518 (M2) was observed that circulated in plasma at 14.3% of total radioactivity. After an oral dose, both THRX-195518 and THRX-697795 (M10, N-dealkylation and reduction of the parent compound) were observed at 12.5% of total circulating radioactivity. THRX-195518 was the major metabolite excreted in feces and comprised 18.8% and 9.4% of the administered intravenous and oral dose, respectively. The major metabolic pathway for revefenacin was hydrolysis to THRX-195518. In vitro pharmacological evaluation of THRX-195518 indicated that it had a 10-fold lower binding affinity for the M3 receptor relative to revefenacin. Receptor occupancy analysis suggested that THRX-195518 has minimal contribution to systemic pharmacology relative to revefenacin after inhaled administration. SIGNIFICANCE STATEMENT: The major metabolic pathway for revefenacin was hydrolysis to the metabolite THRX-195518 (M2), and both revefenacin and THRX-195518 underwent hepatic-biliary and fecal elimination after oral or intravenous administration with negligible renal excretion. Pharmacological evaluation of THRX-195518 indicated that it had a 10-fold lower binding affinity for the M3 muscarinic receptor relative to revefenacin and that THRX-195518 has minimal contribution to systemic pharmacology after inhaled administration.


Subject(s)
Benzamides/pharmacokinetics , Bronchodilator Agents/pharmacokinetics , Carbamates/pharmacokinetics , Muscarinic Antagonists/pharmacokinetics , Administration, Inhalation , Administration, Oral , Adult , Benzamides/administration & dosage , Benzamides/analysis , Biological Availability , Bronchodilator Agents/administration & dosage , Carbamates/administration & dosage , Carbamates/analysis , Feces/chemistry , Healthy Volunteers , Hepatobiliary Elimination , Humans , Infusions, Intravenous , Male , Middle Aged , Muscarinic Antagonists/administration & dosage , Pulmonary Disease, Chronic Obstructive/drug therapy , Solutions , Young Adult
12.
Drug Metab Dispos ; 48(10): 934-943, 2020 10.
Article in English | MEDLINE | ID: mdl-32665417

ABSTRACT

The PXB-mouse is potentially a useful in vivo model to predict human hepatic metabolism and clearance. Four model compounds, [14C]desloratadine, [3H]mianserin, cyproheptadine, and [3H]carbazeran, all reported with disproportionate human metabolites, were orally administered to PXB- or control SCID mice to elucidate the biotransformation of each of them. For [14C]desloratadine in PXB-mice, O-glucuronide of 3-hydroxydesloratadine was observed as the predominant metabolite in both the plasma and urine. Both 3-hydroxydesloratadine and its O-glucuronide were detected as major drug-related materials in the bile, whereas only 3-hydroxydesloratadine was detected in the feces, suggesting that a fraction of 3-hydroxydesloratadine in feces was derived from deconjugation of its O-glucuronide by gut microflora. This information can help understand the biliary clearance mechanism of a drug and may fill the gap in a human absorption, distribution, metabolism, and excretion study, in which the bile samples are typically not available. The metabolic profiles in PXB-mice were qualitatively similar to those reported in humans in a clinical study in which 3-hydroxydesloratadine and its O-glucuronide were major and disproportionate metabolites compared with rat, mouse, and monkey. In the control SCID mice, neither of the metabolites was detected in any matrix. Similarly, for the other three compounds, all human specific or disproportionate metabolites were detected at a high level in PXB-mice, but they were either minimally observed or not observed in the control mice. Data from these four compounds indicate that studies in PXB-mice can help predict the potential for the presence of human disproportionate metabolites (relative to preclinical species) prior to conducting clinical studies and understand the biliary clearance mechanism of a drug. SIGNIFICANCE STATEMENT: Studies in PXB-mice have successfully predicted the human major and disproportionate metabolites compared with preclinical safety species for desloratadine, mianserin, cyproheptadine, and carbazeran. In addition, biliary excretion data from PXB-mice can help illustrate the human biliary clearance mechanism of a drug.


Subject(s)
Hepatobiliary Elimination , Liver/metabolism , Animals , Bile/metabolism , Biotransformation , Carbamates/administration & dosage , Carbamates/pharmacokinetics , Cyproheptadine/administration & dosage , Cyproheptadine/pharmacokinetics , Drug Evaluation, Preclinical/methods , Hepatocytes/metabolism , Hepatocytes/transplantation , Humans , Liver/cytology , Loratadine/administration & dosage , Loratadine/analogs & derivatives , Loratadine/pharmacokinetics , Male , Mianserin/administration & dosage , Mianserin/pharmacokinetics , Mice , Transplantation Chimera/metabolism
13.
Drug Metab Dispos ; 48(7): 580-586, 2020 07.
Article in English | MEDLINE | ID: mdl-32357972

ABSTRACT

Carbazeran is a potent phosphodiesterase inhibitor with species-dependent metabolic profiles in rats, dogs, and humans. In this study, we investigated the aldehyde oxidase (AOX)-mediated oxidation of carbazeran to 4-oxo derivatives in chimeric NOD/Shi-scid IL2 receptor gamma-null mice expressing a herpes simplex virus type 1 thymidine kinase transgene with humanized livers (humanized-liver mice). Liver cytosolic fractions from humanized-liver mouse effectively catalyzed carbazeran 4-oxidation with high affinity for the substrate, similar to those of the human liver cytosolic fractions and recombinant human AOX1 protein. Furthermore, hepatocytes prepared from humanized-liver mice and humans also exhibited substantial metabolism via carbazeran 4-oxidation. After a single oral administration of carbazeran (10 mg/kg), plasma levels of 4-oxo-carbazeran, N-desethyl-4-oxo-carbazeran, and 6,7-dimethoxy-1-[4-(hydroxy)-piperidino]-4-phthalazinone (three human metabolites formed via 4-oxidation) were higher in humanized-liver mice than in the control mice. In contrast, plasma levels of O-desmethylcarbazeran (a major metabolite in dogs) in control mice were higher than those in the humanized-liver mice. Relative excreted amounts of the three 4-oxidation-derived human-specific metabolites in the urine and feces were greater for humanized-liver mice than control mice, whereas the relative excreted amounts of O-desmethylcarbazeran were predominant in the urine and feces of control mice. Thus, the production of carbazeran 4-oxo derivatives was elevated in humanized-liver mice compared with control mice, in agreement with our in vitro enzyme-mediated oxidation data. These results suggest that hepatic human AOX1 functions in humanized-liver mice at the in vivo level and that humanized-liver mice may be useful for predicting drug metabolism in humans, at least with regard to human AOX1-dependent metabolism. SIGNIFICANCE STATEMENT: We found that the production of carbazeran 4-oxo derivatives was higher in humanized-liver mice than in control mice. These results were supported by the fact that carbazeran was rapidly metabolized to 4-oxo-carbazeran in humanized-liver mouse hepatocytes expressing human aldehyde oxidase 1. These results suggest that human aldehyde oxidase 1 is functional in humanized-liver mice in vivo and that chimeric NOD/Shi-scid IL2 receptor gamma-null mice expressing a herpes simplex virus type 1 thymidine kinase transgene transplanted with human hepatocytes may be a suitable model animal for predicting aldehyde oxidase-dependent biotransformation of drugs in humans.


Subject(s)
Aldehyde Oxidase/metabolism , Carbamates/pharmacokinetics , Administration, Oral , Adolescent , Adult , Aged , Animals , Biotransformation , Carbamates/administration & dosage , Cells, Cultured , Child , Child, Preschool , Dogs , Drug Evaluation, Preclinical/methods , Enzyme Assays , Feasibility Studies , Female , Guinea Pigs , Hepatocytes/metabolism , Humans , Interleukin Receptor Common gamma Subunit/genetics , Macaca fascicularis , Male , Mice , Mice, Knockout , Middle Aged , Oxidation-Reduction , Rabbits , Rats , Recombinant Proteins/metabolism , Swine , Swine, Miniature , Transplantation Chimera/metabolism , Young Adult
14.
Ther Drug Monit ; 42(2): 163-164, 2020 04.
Article in English | MEDLINE | ID: mdl-32195986
15.
Ann Pharmacother ; 54(10): 1016-1020, 2020 10.
Article in English | MEDLINE | ID: mdl-32270686

ABSTRACT

OBJECTIVE: The purpose of this article is to review the available clinical trial data that led to the Food and Drug Administration (FDA) approval of solriamfetol as well as its role in clinical practice. DATA SOURCES: A MEDLINE/PubMed search was conducted (January 2000 to February 2020) using the keyword solriamfetol to discover appropriate clinical trials. STUDY SELECTION AND DATA EXTRACTION: Articles were included that were published in the English language and related to the FDA approval of solriamfetol or provided novel information regarding this drug entity. DATA SYNTHESIS: The findings of the review show that solriamfetol may be a safe and effective option for the treatment of excessive sleepiness (ES) related to narcolepsy and obstructive sleep apnea (OSA). RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE: Solriamfetol is distinguished from other stimulants in that it has lower binding affinity to dopamine and norepinephrine transporters and does not have the monoamine-releasing effects of amphetamines at usual therapeutic doses. Because of solriamfetol's unique mechanism of action, there may be a reduction in abuse potential compared with the other currently FDA-approved options. CONCLUSIONS: In clinical trials, solriamfetol has shown dose-dependent improvement in wakefulness over placebo and adds another option for clinicians when treating ES in narcolepsy and OSA.


Subject(s)
Carbamates/therapeutic use , Disorders of Excessive Somnolence/drug therapy , Narcolepsy/complications , Phenylalanine/analogs & derivatives , Sleep Apnea, Obstructive/complications , Wakefulness-Promoting Agents/therapeutic use , Carbamates/administration & dosage , Carbamates/pharmacokinetics , Clinical Trials as Topic , Disorders of Excessive Somnolence/etiology , Humans , Narcolepsy/drug therapy , Phenylalanine/administration & dosage , Phenylalanine/pharmacokinetics , Phenylalanine/therapeutic use , Sleep Apnea, Obstructive/drug therapy , Sleepiness , Treatment Outcome , United States , United States Food and Drug Administration , Wakefulness-Promoting Agents/administration & dosage , Wakefulness-Promoting Agents/pharmacokinetics
16.
Blood Purif ; 49(6): 692-699, 2020.
Article in English | MEDLINE | ID: mdl-32289799

ABSTRACT

BACKGROUND/AIMS: Direct-acting antivirals (DAAs) play a key role in the eradication of hepatitis C virus (HCV) infection. However, limited data are available on DAA for treating HCV infection in hemodialysis (HD) patients. This study was to evaluate the pharmacokinetic characteristics and effectiveness of daclatasvir/sofosbuvir (DAC/SOF) and ledipasvir/SOF (LDV/SOF) in HD patients. METHODS: Seven patients were given SOF coadministered with DAC or LDV once daily for 12 weeks. The plasma concentrations of SOF007, DAC, and LDV were determined by high-performance liquid chromatography-electrospray ionization tandem mass spectrometry. RESULTS: A sustained virologic response in week 12 (SVR12) was achieved in 6 (100%) patients, except for 1 patient dying due to severe cerebral hemorrhage not related to antiviral therapy. The extraction ratio of SOF007 was 66.67%, and the estimated HD clearance of SOF007 was 5.65 L/h. CONCLUSION: The combination of SOF with either DAC or LDV is well tolerated and offers high SVR12 in HD patients.


Subject(s)
Benzimidazoles/pharmacokinetics , Carbamates/pharmacokinetics , Fluorenes/pharmacokinetics , Hepatitis C, Chronic/complications , Imidazoles/pharmacokinetics , Kidney Failure, Chronic/complications , Pyrrolidines/pharmacokinetics , Renal Dialysis , Sofosbuvir/pharmacokinetics , Valine/analogs & derivatives , Adult , Aged , Benzimidazoles/therapeutic use , Carbamates/therapeutic use , Clinical Trials as Topic , Drug Monitoring , Drug Therapy, Combination , Female , Fluorenes/therapeutic use , Hepatitis C, Chronic/drug therapy , Humans , Imidazoles/therapeutic use , Kidney Failure, Chronic/therapy , Male , Middle Aged , Pyrrolidines/therapeutic use , Renal Dialysis/adverse effects , Renal Dialysis/methods , Sofosbuvir/therapeutic use , Treatment Outcome , Valine/pharmacokinetics , Valine/therapeutic use
17.
J Formos Med Assoc ; 119(5): 933-940, 2020 May.
Article in English | MEDLINE | ID: mdl-31594667

ABSTRACT

BACKGROUND/PURPOSE: The assessment of drug-drug interaction (DDI) is important not only for safety but also for maintaining the efficacy of direct acting antivirals in chronic hepatitis C (CHC). This study aims to evaluate DDI before and during elbasvir/grazoprevir (EBR/GZR) treatment. METHODS: CHC patients who treated with EBR/GZR in five hospitals were enrolled. The patients' demographic data, comorbidities, concomitant medications taken before and during EBR/GZR were recorded. DDI was evaluated using a tool from the HEP Drug Interactions (www.hep-druginteractions.org) website. In addition to the evaluation of DDI for EBR/GZR, the virtual DDI of ledipasvir/sofosbuvir (LDV/SOF), sofosbuvir/velpatasvir (SOF/VEL) and glecaprevir/pibrentasvir (GLE/PIB) were evaluated. Degrees of DDI were classified as "do not co-administer", "potential interaction", and "potentially weak interaction". RESULTS: A total of 460 patients were enrolled. At baseline, 80.1% of patients had one or more comorbidities and 72.8% took one or more medications. Cardiovascular diseases (43.9%), gastrointestinal diseases (37.4%), and metabolic diseases (36.7%) were the three most common comorbidities. The prevalence of DDI before EBR/GZR treatment was 12.8% (59 patients). Among the same population, the prevalence of virtual DDI of SOF/VEL, GLE/PIB, and LDV/SOF were 38.5% (179 patients), 48.8% (220 patients), and 57.0% (262 patients), respectively. During EBR/GZR treatment, 167 patients (36.3%) took newly prescribed medications. One patient (0.2%) and seven patients (1/5%) exhibited do-not-co-administer and potential interaction with EBR/GZR, respectively. CONCLUSION: DDI was limited in treatment with EBR/GZR. DDI can occur upon the administering of a new medication during antiviral treatment and attention should be paid to it. TRIAL REGISTRATION NUMBER: NCT03706222.


Subject(s)
Amides , Antiviral Agents , Carbamates , Cyclopropanes , Hepatitis C, Chronic , Quinoxalines , Sulfonamides , Amides/pharmacokinetics , Antiviral Agents/therapeutic use , Benzofurans/pharmacokinetics , Carbamates/pharmacokinetics , Cyclopropanes/pharmacokinetics , Drug Interactions , Hepacivirus/genetics , Hepatitis C, Chronic/drug therapy , Humans , Imidazoles/pharmacokinetics , Quinoxalines/pharmacokinetics , Sulfonamides/pharmacokinetics
18.
Drug Dev Res ; 81(1): 62-69, 2020 02.
Article in English | MEDLINE | ID: mdl-31593323

ABSTRACT

The role of theranostics in cancer management is growing so is the selection of vectors used to deliver these modalities to cancer cells. We describe biological evaluation of a novel theranostic agent targeted to microtubules. Methyl N-[5-(3'-[131 I]iodobenzoyl)-1H-benzimidazol-2-yl]carbamate (1) and methyl N-[5-(3'-[125 I]iodobenzoyl)-1H-benzimidazol-2-yl]carbamate (2) were synthesized from a common precursor 3'-stannylated derivative (4). Antiproliferative effects and radiotoxicity of 131 I-labeled ß-particle emitting 1 were examined in vitro in human neuroblastoma and glioblastoma cells lines. The therapeutic potential of 1 was also examined in a subcutaneous mouse model of human glioblastoma U-87 MG. Compound 1 at the extracellular radioactive concentration of 0.35 MBq/mL, easily achievable in vivo, kills >90% of neuroblastoma cells and >60% glioblastoma cells as measured in a clonogenic assay. D10 doses established for 1 indicate that as few as 3,000 decays are sufficient to kill 90% of BE(2)-C cells. Even U-87 MG cells, the least sensitive of the tested cell lines, require <20,000 decays of intracellular 131 I to reduce number of clonogenic cells by 90%. Biodistribution studies of 2 delivered either intratumorally or intraperitoneally show a similar tissue distribution for both routes of the drug administration. The whole body clearance half-lives were on average 6 hr. Intratumor administration of 1 produces significant tumor growth delay. After a single dose of 8.4 ± 0.3 MBq of compound 1, the tumor doubling times were 3.2 ± 0.1 and 7.9 ± 0.6 days in control and treated mice, respectively. Methyl N-[5-(3'-radiohalobenzoyl)-1H-benzimidazol-2-yl]carbamates have properties compatible with a theranostic approach to cancer management.


Subject(s)
Brain Neoplasms/radiotherapy , Carbamates/administration & dosage , Glioblastoma/radiotherapy , Iodine Radioisotopes/chemistry , Animals , Carbamates/chemistry , Carbamates/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Humans , Injections, Intraperitoneal , Mice , Theranostic Nanomedicine , Tissue Distribution , Treatment Outcome , Xenograft Model Antitumor Assays
19.
Molecules ; 25(21)2020 Nov 02.
Article in English | MEDLINE | ID: mdl-33147810

ABSTRACT

The review presents data from the last few years on bioanalytical methods used in therapeutic drug monitoring (TDM) of the 1st-3rd generation and the newest antiepileptic drug (AEDs) cenobamate in patients with various forms of seizures. Chemical classification, structure, mechanism of action, pharmacokinetic data and therapeutic ranges for total and free fractions and interactions were collected. The primary data on bioanalytical methods for AEDs determination included biological matrices, sample preparation, dried blood spot (DBS) analysis, column resolution, detection method, validation parameters, and clinical utility. In conclusion, the most frequently described method used in AED analysis is the LC-based technique (HPLC, UHPLC, USLC) combined with highly sensitive mass detection or fluorescence detection. However, less sensitive UV is also used. Capillary electrophoresis and gas chromatography have been rarely applied. Besides the precipitation of proteins or LLE, an automatic SPE is often a sample preparation method. Derivatization was also indicated to improve sensitivity and automate the analysis. The usefulness of the methods for TDM was also highlighted.


Subject(s)
Anticonvulsants , Carbamates , Chlorophenols , Drug Monitoring , Seizures , Tetrazoles , Anticonvulsants/pharmacokinetics , Anticonvulsants/therapeutic use , Carbamates/chemistry , Carbamates/pharmacokinetics , Chlorophenols/chemistry , Chlorophenols/pharmacokinetics , Humans , Seizures/drug therapy , Seizures/metabolism , Tetrazoles/chemistry , Tetrazoles/pharmacokinetics
20.
Hepatology ; 68(6): 2145-2157, 2018 12.
Article in English | MEDLINE | ID: mdl-30070722

ABSTRACT

This open-label, phase IIa study assessed the safety, pharmacokinetics, and efficacy of direct-acting antiviral agent (DAA) regimens in patients with chronic hepatitis C virus (HCV) infection. Multiple 6-12-week oral regimens of 400-800 mg once daily (QD) AL-335 + 50 mg QD/every other day odalasvir ± 75-150 mg QD simeprevir were evaluated in treatment-naïve, HCV genotype (GT)1/3-infected patients without cirrhosis. Safety/pharmacokinetic parameters, HCV-RNA, and sequencing data were assessed. Treatment regimens for later study cohorts were adjusted based on emerging data. In total, 112 patients were enrolled. Three serious treatment-emergent adverse events occurred, one of which (a Mobitz type 1 second-degree atrioventricular block [Wenckebach]) was possibly related to high odalasvir exposure and resulted in premature discontinuation of study drugs. No other clinically significant safety findings were identified. GT1-infected patients receiving 3-DAA for 6-8 weeks achieved 100% sustained virologic response 12 weeks and 24 weeks after the end of treatment (sustained virologic response [SVR12/24]). GT1-infected patients receiving 2-DAA or GT3-infected patients receiving 3-DAA had SVR12/24 less than 90%, whether treated for 8 weeks or 12 weeks. Virologic failure was associated with the emergence of generally persistent NS5A and/or transient NS5B resistance-associated substitutions in most patients. Pharmacokinetic characteristics of the three drugs were also elucidated. Conclusions: In treatment-naïve subjects without cirrhosis, AL-335 + odalasvir + simeprevir for 6-8 weeks was generally safe and highly efficacious against HCV GT1. However, inadequate efficacy was observed for the 2-DAA regimen in GT1-infected subjects and the 3-DAA regimen in GT3-infected subjects.


Subject(s)
Antiviral Agents/administration & dosage , Benzimidazoles/administration & dosage , Carbamates/administration & dosage , Hepacivirus/genetics , Hepatitis C/drug therapy , Indoles/administration & dosage , Simeprevir/administration & dosage , Adult , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Benzimidazoles/adverse effects , Benzimidazoles/pharmacokinetics , Carbamates/adverse effects , Carbamates/pharmacokinetics , Drug Resistance, Viral/genetics , Drug Therapy, Combination , Female , Hepatitis C/virology , Humans , Indoles/adverse effects , Indoles/pharmacokinetics , Male , Middle Aged , Simeprevir/adverse effects , Simeprevir/pharmacokinetics , Viral Nonstructural Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL