Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
AAPS J ; 26(1): 17, 2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38267790

ABSTRACT

Ritlecitinib, an orally available Janus kinase 3 and tyrosine kinase inhibitor being developed for the treatment of alopecia areata (AA), is highly soluble across the physiological pH range at the therapeutic dose. As such, it is expected to dissolve rapidly in any in vitro dissolution conditions. However, in vitro dissolution data showed slower dissolution for 100-mg capsules, used for the clinical bioequivalence (BE) study, compared with proposed commercial 50-mg capsules. Hence, a biowaiver for the lower 50-mg strength using comparable multimedia dissolution based on the f2 similarity factor was not possible. The in vivo relevance of this observed in vitro dissolution profile was evaluated with a physiologically based pharmacokinetic (PBPK) model. This report describes the development, verification, and application of the ritlecitinib PBPK model to translate observed in vitro dissolution data to an in vivo PK profile for ritlecitinib capsule formulations. Virtual BE (VBE) trials were conducted using the Simcyp VBE module, including the model-predicted within-subject variability or intra-subject coefficient of variation (ICV). The results showed the predicted ICV was predicted to be smaller than observed clinical ICV, resulting in a more optimistic BE risk assessment. Additional VBE assessment was conducted by incorporating clinically observed ICV. The VBE trial results including clinically observed ICV demonstrated that proposed commercial 50-mg capsules vs clinical 100-mg capsules were bioequivalent, with > 90% probability of success. This study demonstrates a PBPK model-based biowaiver for a clinical BE study while introducing a novel method to integrate clinically observed ICV into VBE trials with PBPK models. Trial registration: NCT02309827, NCT02684760, NCT04004663, NCT04390776, NCT05040295, NCT05128058.


Subject(s)
Alopecia Areata , Humans , Therapeutic Equivalency , Probability , Protein Kinase Inhibitors , Risk Assessment
3.
J Pharm Sci ; 113(1): 64-71, 2024 01.
Article in English | MEDLINE | ID: mdl-37805075

ABSTRACT

PAXLOVID™ is a combination medicine of nirmatrelvir tablets co-packaged with ritonavir tablets. Nirmatrelvir is a peptidomimetic inhibitor of SARS-CoV2 main protease (Mpro), developed for the treatment of COVID-19. Ritonavir is co-administered as a pharmacokinetics (PK) enhancer to inhibit CYP3A mediated metabolism increasing exposures of nirmatrelvir. In the solid form, nirmatrelvir exists in a stable single conformational state (ANTI form). However, nirmatrelvir exhibits atropisomerism in solution whereby upon dissolution the ANTI rotational isomer reversibly converts to another conformation state (SYN form). Nirmatrelvir rotamer conversion follows pseudo first order kinetics with a conversion half-life of approximately 15 min in aqueous solutions, which is on a similar time scale of diffusion mediated dissolution from the solid form. In vitro dissolution studies further indicated that rotamer conversion is one of the processes controlling nirmatrelvir dissolution. It was hypothesized that rotamer conversion kinetics would affect oral absorption of nirmatrelvir in vivo. Consequently, a physiologically based pharmacokinetic (PBPK) model for Paxlovid was developed in Simcyp™ using the advanced dissolution, absorption, and metabolism model (ADAM) by incorporating rotamer conversion kinetics to achieve a more mechanistic description of nirmatrelvir oral absorption. The results demonstrate that the established absorption model with rotamer kinetics adequately described observed clinical data from various nirmatrelvir doses, dosage forms, and dosing regimens. The predicted vs. observed AUCinf and Cmax ratios were within 2-fold. The model has been internally used to inform clinical studies and dose recommendations for pediatrics.


Subject(s)
RNA, Viral , Ritonavir , Humans , Child , Solubility , Antiviral Agents
4.
Pharm Res ; 40(8): 1927-1938, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37231296

ABSTRACT

PURPOSE: PAXLOVID™ is nirmatrelvir tablets co-packaged with ritonavir tablets. Ritonavir is used as a pharmacokinetics (PK) enhancer to reduce metabolism and increase exposure of nirmatrelvir. This is the first disclosure of Paxlovid physiologically-based pharmacokinetic (PBPK) model. METHODS: Nirmatrelvir PBPK model with first-order absorption kinetics was developed using in vitro, preclinical, and clinical data of nirmatrelvir in the presence and absence of ritonavir. Clearance and volume of distribution were derived from nirmatrelvir PK obtained using a spray-dried dispersion (SDD) formulation where it is considered to be dosed as an oral solution, and absorption is near complete. The fraction of nirmatrelvir metabolized by CYP3A was estimated based on in vitro and clinical ritonavir drug-drug interaction (DDI) data. First-order absorption parameters were established for both SDD and tablet formulation using clinical data. Nirmatrelvir PBPK model was verified with both single and multiple dose human PK data, as well as DDI studies. Simcyp® first-order ritonavir compound file was also verified with additional clinical data. RESULTS: The nirmatrelvir PBPK model described the observed PK profiles of nirmatrelvir well with predicted AUC and Cmax values within ± 20% of the observed. The ritonavir model performed well resulting in predicted values within twofold of observed. CONCLUSIONS: Paxlovid PBPK model developed in this study can be applied to predict PK changes in special populations, as well as model the effect of victim and perpetrator DDI. PBPK modeling continues to play a critical role in accelerating drug discovery and development of potential treatments for devastating diseases such as COVID-19. NCT05263895, NCT05129475, NCT05032950 and NCT05064800.


Subject(s)
COVID-19 , Ritonavir , Humans , Ritonavir/pharmacokinetics , Computer Simulation , Kinetics , Drug Interactions , Models, Biological
5.
Drug Metab Dispos ; 44(2): 209-19, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26608081

ABSTRACT

The thiouracil derivative PF-06282999 [2-(6-(5-chloro-2-methoxyphenyl)-4-oxo-2-thioxo-3,4-dihydropyrimidin-1(2H)-yl)acetamide] is an irreversible inactivator of myeloperoxidase and is currently in clinical trials for the potential treatment of cardiovascular diseases. Concerns over idiosyncratic toxicity arising from bioactivation of the thiouracil motif to reactive species in the liver have been largely mitigated through the physicochemical (molecular weight, lipophilicity, and topological polar surface area) characteristics of PF-06282999, which generally favor elimination via nonmetabolic routes. To test this hypothesis, pharmacokinetics and disposition studies were initiated with PF-06282999 using animals and in vitro assays, with the ultimate goal of predicting human pharmacokinetics and elimination mechanisms. Consistent with its physicochemical properties, PF-06282999 was resistant to metabolic turnover from liver microsomes and hepatocytes from animals and humans and was devoid of cytochrome P450 inhibition. In vitro transport studies suggested moderate intestinal permeability and minimal transporter-mediated hepatobiliary disposition. PF-06282999 demonstrated moderate plasma protein binding across all of the species. Pharmacokinetics in preclinical species characterized by low to moderate plasma clearances, good oral bioavailability at 3- to 5-mg/kg doses, and renal clearance as the projected major clearance mechanism in humans. Human pharmacokinetic predictions using single-species scaling of dog and/or monkey pharmacokinetics were consistent with the parameters observed in the first-in-human study, conducted in healthy volunteers at a dose range of 20-200 mg PF-06282999. In summary, disposition characteristics of PF-06282999 were relatively similar across preclinical species and humans, with renal excretion of the unchanged parent emerging as the principal clearance mechanism in humans, which was anticipated based on its physicochemical properties and supported by preclinical studies.


Subject(s)
Acetamides/pharmacokinetics , Pyrimidinones/pharmacokinetics , Thiouracil/pharmacokinetics , Administration, Oral , Animals , Biological Availability , Caco-2 Cells , Cytochrome P-450 Enzyme Inhibitors/metabolism , Dogs , Drug Evaluation, Preclinical/methods , Female , HEK293 Cells , Haplorhini , Hepatocytes/metabolism , Humans , Intestinal Absorption/physiology , Male , Mice , Microsomes, Liver/metabolism , Peroxidase/metabolism , Protein Binding , Rats , Rats, Wistar
6.
Pharm Res ; 33(1): 167-76, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26286187

ABSTRACT

PURPOSE: Currently, the FDA allows biowaivers for Class I (high solubility and high permeability) and Class III (high solubility and low permeability) compounds of the Biopharmaceutics Classification System (BCS). Scientific evidence should be provided to support biowaivers for BCS Class I and Class III (high solubility and low permeability) compounds. METHODS: Data on the effects of excipients on drug permeability are needed to demonstrate that commonly used excipients do not affect the permeability of BCS Class III compounds, which would support the application of biowaivers to Class III compounds. This study was designed to generate such data by assessing the permeability of four BCS Class III compounds and one Class I compound in the presence and absence of five commonly used excipients. RESULTS: The permeability of each of the compounds was assessed, at three to five concentrations, with each excipient in two different models: Caco-2 cell monolayers, and in situ rat intestinal perfusion. No substantial increases in the permeability of any of the compounds were observed in the presence of any of the tested excipients in either of the models, with the exception of disruption of Caco-2 cell monolayer integrity by sodium lauryl sulfate at 0.1 mg/ml and higher. CONCLUSION: The results suggest that the absorption of these four BCS Class III compounds would not be greatly affected by the tested excipients. This may have implications in supporting biowaivers for BCS Class III compounds in general.


Subject(s)
Biopharmaceutics/classification , Biopharmaceutics/standards , Excipients/chemistry , Algorithms , Animals , Caco-2 Cells , Humans , Intestinal Absorption , Jejunum/metabolism , Permeability , Rats , Rats, Sprague-Dawley , Sodium Dodecyl Sulfate/chemistry , Surface-Active Agents/chemistry , Therapeutic Equivalency , United States , United States Food and Drug Administration
7.
Int J Pharm ; 428(1-2): 8-17, 2012 May 30.
Article in English | MEDLINE | ID: mdl-22349051

ABSTRACT

Ziprasidone, commercially available as Geodon capsules, is an atypical antipsychotic used in the treatment of schizophrenia and bipolar disorder. It is a BCS Class II drug that shows up to a 2-fold increase in absorption in the presence of food. Because compliance is a major issue in this patient population, we developed and characterized solubilized formulations of ziprasidone in an effort to improve absorption in the fasted state, thereby resulting in a reduced food effect. Three formulations utilizing solubilization technologies were studied: (1) an amorphous inclusion complex of ziprasidone mesylate and a cyclodextrin, (2) a nanosuspension of crystalline ziprasidone free base, and (3) jet-milled ziprasidone HCl coated crystals made by spray drying (CCSD) the drug with hypromellose acetate succinate. The formulations were characterized by in vitro methods appropriate to each particular solubilization technology. These studies confirmed that ziprasidone mesylate - cyclodextrin was an amorphous inclusion complex with enhanced dissolution rates. The ziprasidone free base crystalline nanosuspension showed a mean particle size of 274 nm and a monomodal particle size distribution. In a membrane permeation test, the CCSD showed a 1.5-fold higher initial flux compared to crystalline ziprasidone HCl. The three formulations were administered to fasted beagle dogs and their pharmacokinetics compared to Geodon capsules administered in the fed state. The amorphous complex and the nanosuspension showed increased absorption in the fasted state, indicating that solubilized formulations of ziprasidone have the potential to reduce the food effect in humans.


Subject(s)
Nanoparticles/chemistry , Piperazines/chemistry , Thiazoles/chemistry , Absorption , Acetates/chemistry , Animals , Antipsychotic Agents/chemistry , Antipsychotic Agents/pharmacokinetics , Chemistry, Pharmaceutical/methods , Cyclodextrins/chemistry , Dogs , Drug Stability , Fasting/metabolism , Hypromellose Derivatives , Male , Methylcellulose/analogs & derivatives , Methylcellulose/chemistry , Particle Size , Piperazines/pharmacokinetics , Solubility , Succinic Acid/chemistry , Suspensions/chemistry , Thiazoles/pharmacokinetics
8.
J Pharm Sci ; 99(1): 430-41, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19499569

ABSTRACT

The purpose of this study was to investigate the in vitro and in vivo interactions between flavonoids and P-glycoprotein (P-gp) substrates. The inhibitory effects of flavonoids on P-gp were determined by accumulation studies in P-gp-overexpressing MCF-7/ADR cells using daunomycin (DNM) as a model substrate. Morin, phloretin, biochanin A, chalcone, and silymarin significantly increased DNM accumulation by greater than 2.5-fold, suggesting they are P-gp inhibitors. To explore potential in vivo interactions of flavonoids with P-gp, the effect of biochanin A on the pharmacokinetics of the P-gp substrates doxorubicin, cyclosporine A, and paclitaxel was investigated. In contrast to the in vitro results, intraperitoneal or oral administration of biochanin A did not significantly change the pharmacokinetics of doxorubicin and cyclosporine A. Moderate interaction was observed between biochanin A and paclitaxel, resulting in lower AUC values after both i.v. and oral administration of paclitaxel. The disconnect between the in vitro and in vivo data suggests that P-gp interactions mediated by biochanin A may be limited due to its poor bioavailability and rapid clearance. It is also possible that other transporters or metabolizing enzymes are more important in the in vivo disposition of doxorubicin, cyclosporine A, and paclitaxel than P-gp.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Genistein/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Chromatography, Liquid , Daunorubicin/pharmacokinetics , Daunorubicin/pharmacology , Drug Interactions , Female , Humans , Male , Rats , Rats, Sprague-Dawley , Substrate Specificity , Tandem Mass Spectrometry
9.
J Pharm Sci ; 98(7): 2494-500, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19177514

ABSTRACT

The gastric pH values are controversial in the literature. Some suggest the dog gastric pH is higher than human and dog gastric pH after fed with particular diet is uncertain. Gastric pH in 16 male beagle dogs was measured using Bravo pH telemetry system. For the fed study, the dogs received 10 or 200 g of dog dry food (5L18) 15 min before dosing the Bravo pH capsule, followed by a 50 mL of water to aid in swallowing. It was surprising to find a small, but statistically significantly lower pH in the fed compared to the fasted stomach. The average gastric pH in fasted dogs was 2.05 and 1.08 and 1.26 for 10 and 200 g fed dogs. The average gastric emptying time of the capsule was 1.4, 9.4 and 20 h for fasted, 10 g fed and 200 g fed dogs, respectively. The inter-individual variability was higher in fasted dogs than in fed dogs. The results showed the gastric pH in each colony of dogs can be different from reported values in the literature. It emphasizes that the importance of measuring the pH in each colony when dogs are used to evaluate pharmacokinetics of pH sensitive drugs or formulations.


Subject(s)
Dogs/physiology , Hydrogen-Ion Concentration , Stomach/chemistry , Stomach/physiology , Telemetry/methods , Animals , Calibration , Diet , Fasting , Gastric Emptying , Gastrointestinal Contents , Humans , Time Factors
10.
AAPS J ; 8(3): E433-42, 2006 Jul 07.
Article in English | MEDLINE | ID: mdl-17025260

ABSTRACT

Biochanin A (BCA) is a dietary isoflavone present in legumes, most notably red clover, and in many herbal dietary supplements. BCA has been reported to have chemopreventive properties and is metabolized to the isoflavone genistein (GEN), BCA conjugates, and GEN conjugates. The metabolites may contribute to the chemopreventive effects of BCA. The absorption, metabolism, and disposition of BCA have not been determined in rats. Our objective was to evaluate the pharmacokinetics and metabolism of BCA in rats. Male Sprague-Dawley rats were administered BCA by intravenous injection (1 and 5 mg/kg), by intraperitoneal injection (5 and 50 mg/kg), and orally (5 and 50 mg/kg). Plasma and bile samples were enzymatically hydrolyzed in vitro to determine conjugate concentrations for BCA and GEN. Equilibrium dialysis was used to determine protein binding. The BCA and GEN concentrations in plasma, urine, and bile were determined by liquid chromatography-tandem mass spectrometry (LC/MS/MS). The pharmacokinetic parameters of BCA were analyzed by noncompartmental analysis. Significant levels of BCA conjugates and GEN conjugates were detected in plasma and bile. Both BCA and GEN were found to have a high clearance and a large apparent volume of distribution; the bioavailability of both was poor (<4%). Reentry peaks were evident after oral administration of both BCA and GEN, suggesting enterohepatic cycling. The free fraction of BCA in rat plasma was 1.5%. A 2-compartment model that included both linear and nonlinear clearance terms and enterohepatic recirculation best described the plasma data. This represents the first evaluation of the dose-dependent pharmacokinetics and metabolism of BCA in rats.


Subject(s)
Genistein/blood , Trifolium/chemistry , Animals , Biological Availability , Biotransformation , Genistein/analysis , Genistein/metabolism , Genistein/pharmacokinetics , Male , Rats , Rats, Sprague-Dawley , Glycine max/chemistry
11.
Drug Metab Dispos ; 33(3): 341-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15608138

ABSTRACT

Breast cancer resistance protein (BCRP) is a recently identified ATP-binding cassette transporter, important in drug disposition and in the development of multidrug resistance in cancer. Flavonoids, a major class of natural compounds widely present in foods and herbal products, have been shown to be human BCRP inhibitors. The objective of the present study was to evaluate the potential for in vivo pharmacokinetic interactions by comparing the pharmacokinetics of topotecan (a model BCRP substrate) after oral administration of 2 mg/kg topotecan with and without different doses of the flavonoids chrysin or 7,8-benzoflavone (BF) in rats and mdr1a/1b (-/-) mice. Coadministration of 50 mg/kg GF120918 [N-(4-[2-(1,2,3,4-tetrahydro-6,7-dimethoxy-2-isoquinolinyl)ethyl]-phenyl)-9, 10-dihydro-5-methoxy-9-oxo-4-acridine carboxamide] with 2 mg/kg topotecan significantly increased the area under the plasma concentration-time curve and bioavailability of topotecan by more than 4-fold in these animals, indicating the importance of BCRP in the bioavailability and disposition of topotecan in rats. Chrysin (50 microM) and BF (5 microM) significantly inhibited the BCRP-mediated transport of topotecan in BCRP-overexpressing MCF-7 MX100 cells (MCF-7 cells selected with mitoxantrone) to a level comparable to that observed with 10 microM fumitremorgin C (a potent BCRP inhibitor). However, neither chrysin nor BF significantly altered topotecan pharmacokinetics in rats or in mdr1a/1b (-/-) mice after oral coadministration of doses up to 50 mg/kg. The reason(s) for this lack of in vitro-in vivo association may be the lack of potent inhibition activity of the flavonoids against mouse or rat BCRP, as evidenced by our observation that these flavonoids have only weak, if any, inhibition activity against mouse Bcrp1-mediated transport of topotecan in MDCK-Bcrp1 cells.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Antineoplastic Agents/pharmacokinetics , Benzoflavones/pharmacology , Flavonoids/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Topotecan/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Acridines/pharmacology , Animals , Antineoplastic Agents/antagonists & inhibitors , Antineoplastic Agents/blood , Area Under Curve , Biological Availability , Cell Line, Tumor , Drug Interactions , Female , Humans , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Tetrahydroisoquinolines/pharmacology , Topotecan/antagonists & inhibitors , Topotecan/blood , ATP-Binding Cassette Sub-Family B Member 4
12.
Am J Physiol Heart Circ Physiol ; 282(3): H1118-26, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11834511

ABSTRACT

This study investigated the effects of cardiac glycosides on single-channel activity of the cardiac sarcoplasmic reticulum (SR) Ca2+ release channels or ryanodine receptor (RyR2) channels and how this action might contribute to their inotropic and/or toxic actions. Heavy SR vesicles isolated from canine left ventricle were fused with artificial planar lipid bilayers to measure single RyR2 channel activity. Digoxin and actodigin increased single-channel activity at low concentrations normally associated with therapeutic plasma levels, yielding a 50% of maximal effect of approximately 0.2 nM for each agent. Channel activation by glycosides did not require MgATP and occurred only when digoxin was applied to the cytoplasmic side of the channel. Similar results were obtained in human RyR2 channels; however, neither the crude skeletal nor the purified cardiac channel was activated by glycosides. Channel activation was dependent on [Ca2+] on the luminal side of the bilayer with maximal stimulation occurring between 0.3 and 10 mM. Rat RyR2 channels were activated by digoxin only at 1 microM, consistent with the lower sensitivity to glycosides in rat heart. These results suggest a model in which RyR2 channel activation by digoxin occurs only when luminal [Ca2+] was increased above 300 microM (in the physiological range). Consequently, increasing SR load (by Na+ pump inhibition) serves to amplify SR release by promoting direct RyR2 channel activation via a luminal Ca2+-sensitive mechanism. This high-affinity effect of glycosides could contribute to increased SR Ca2+ release and might play a role in the inotropic and/or toxic actions of glycosides in vivo.


Subject(s)
Cardiac Glycosides/pharmacology , Digoxin/pharmacology , Heart/physiology , Ryanodine Receptor Calcium Release Channel/drug effects , Animals , Cardenolides/pharmacology , Dogs , Heart/drug effects , Lipid Bilayers , Muscle, Skeletal/physiology , Rats , Ryanodine Receptor Calcium Release Channel/physiology , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/physiology , Sodium-Potassium-Exchanging ATPase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...