Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Pharm Res ; 37(6): 95, 2020 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-32405699

RESUMEN

During non-clinical and clinical development of a new molecular entity (NME), modeling and simulation (M&S) are routinely used to predict the exposure and pharmacokinetics (PK) of the drug compound in humans. The basic methodology and output are generally understood across all functional disciplines. However, this understanding is mostly restricted to traditional methods such as those in simplified kinetic models and void of adequate mechanistic foundation to address questions beyond the observed clinical data. In the past two decades, alternative and more mechanistic methods, particularly for describing absorption, distribution, excretion and metabolism (ADME) of drugs have been developed and applied under the general umbrella of physiologically-based pharmacokinetic (PBPK) methods. Their mechanistic nature gives the ability to ask many other questions which were not traditionally asked and provide some logically and evidenced-based potential answers. Whilst traditional PK methods are mainstream and understood by most scientists, mechanistic absorption models alongside other PBPK approaches are still deemed eclectic, despite making significant strides in the fundamental science as well as regulatory acceptance. On November 3rd, a short course was held at the annual American Association of Pharmaceutical Scientists (AAPS) meeting in San Antonio, Texas. The different talks were tailored to provide a basis or rationale for the subject, introduction to fundamental principles with historical perspective, a critique of the state-of-the-art, examples of successful application of the methods across different phases of the drug development process and the specific standards these mechanistic models should meet to be fully reliable from a regulatory perspective.


Asunto(s)
Modelos Biológicos , Modelos Químicos , Preparaciones Farmacéuticas/química , Administración Oral , Animales , Humanos , Absorción Intestinal , Tasa de Depuración Metabólica , Permeabilidad , Farmacocinética , Solubilidad , Tecnología Farmacéutica , Distribución Tisular
2.
Pharm Res ; 37(12): 245, 2020 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-33215336

RESUMEN

PURPOSE: The purpose of this study is to show how the Ocular Compartmental Absorption & Transit (OCAT™) model in GastroPlus® can be used to characterize ocular drug pharmacokinetic performance in rabbits for ointment formulations. METHODS: A newly OCAT™ model developed for fluorometholone, as well as a previously verified model for dexamethasone, were used to characterize the aqueous humor (AH) concentration following the administration of multiple ointment formulations to rabbit. The model uses the following parameters: application surface area (SA), a fitted application time, and the fitted Higuchi release constant to characterize the rate of passage of the active pharmaceutical ingredient from the ointment formulations into the tears in vivo. RESULTS: Parameter sensitivity analysis was performed to understand the impact of ointment formulation changes on ocular exposure. While application time was found to have a significant impact on the time of maximal concentration in AH, both the application SA and the Higuchi release constant significantly influenced both the maximum concentration and the ocular exposure. CONCLUSIONS: This initial model for ointment ophthalmic formulations is a first step to better understand the interplay between physiological factors and ophthalmic formulation physicochemical properties and their impact on in vivo ocular drug pharmacokinetic performance in rabbits.


Asunto(s)
Dexametasona/farmacocinética , Ojo/metabolismo , Fluorometolona/farmacocinética , Glucocorticoides/farmacocinética , Modelos Biológicos , Absorción Ocular , Administración Oftálmica , Animales , Humor Acuoso/metabolismo , Simulación por Computador , Dexametasona/administración & dosificación , Fluorometolona/administración & dosificación , Glucocorticoides/administración & dosificación , Pomadas , Conejos
3.
Drug Metab Dispos ; 47(8): 818-831, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31101678

RESUMEN

Cytosolic sulfotransferases (SULTs), including SULT1A, SULT1B, SULT1E, and SULT2A isoforms, play noteworthy roles in xenobiotic and endobiotic metabolism. We quantified the protein abundances of SULT1A1, SULT1A3, SULT1B1, and SULT2A1 in human liver cytosol samples (n = 194) by liquid chromatography-tandem mass spectrometry proteomics. The data were analyzed for their associations by age, sex, genotype, and ethnicity of the donors. SULT1A1, SULT1B1, and SULT2A1 showed significant age-dependent protein abundance, whereas SULT1A3 was invariable across 0-70 years. The respective mean abundances of SULT1A1, SULT1B1, and SULT2A1 in neonatal samples was 24%, 19%, and 38% of the adult levels. Interestingly, unlike UDP-glucuronosyltransferases and cytochrome P450 enzymes, SULT1A1 and SULT2A1 showed the highest abundance during early childhood (1 to <6 years), which gradually decreased by approx. 40% in adolescents and adults. SULT1A3 and SULT1B1 abundances were significantly lower in African Americans compared with Caucasians. Multiple linear regression analysis further confirmed the association of SULT abundances by age, ethnicity, and genotype. To demonstrate clinical application of the characteristic SULT ontogeny profiles, we developed and validated a proteomics-informed physiologically based pharmacokinetic model of acetaminophen. The latter confirmed the higher fractional contribution of sulfation over glucuronidation in the metabolism of acetaminophen in children. The study thus highlights that the ontogeny-based age-dependent fractional contribution (fm) of individual drug-metabolizing enzymes has better potential in prediction of drug-drug interactions and the effect of genetic polymorphisms in the pediatric population.


Asunto(s)
Acetaminofén/farmacocinética , Variación Biológica Poblacional/fisiología , Citosol/metabolismo , Hígado/metabolismo , Sulfotransferasas/metabolismo , Adolescente , Adulto , Factores de Edad , Anciano , Área Bajo la Curva , Niño , Preescolar , Cromatografía Líquida de Alta Presión , Interacciones Farmacológicas/fisiología , Femenino , Humanos , Lactante , Recién Nacido , Hígado/citología , Masculino , Persona de Mediana Edad , Modelos Biológicos , Proteómica , Factores Sexuales , Sulfatos/metabolismo , Sulfotransferasas/análisis , Espectrometría de Masas en Tándem , Adulto Joven
4.
Mol Pharm ; 15(3): 831-839, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29337562

RESUMEN

When medicinal chemists need to improve oral bioavailability (%F) during lead optimization, they systematically modify compound properties mainly based on their own experience and general rules of thumb. However, at least a dozen properties can influence %F, and the difficulty of multiparameter optimization for such complex nonlinear processes grows combinatorially with the number of variables. Furthermore, strategies can be in conflict. For example, adding a polar or charged group will generally increase solubility but decrease permeability. Identifying the 2 or 3 properties that most influence %F for a given compound series would make %F optimization much more efficient. We previously reported an adaptation of physiologically based pharmacokinetic (PBPK) simulations to predict %F for lead series from purely computational inputs within a 2-fold average error. Here, we run thousands of such simulations to generate a comprehensive "bioavailability landscape" for each series. A key innovation was recognition that the large and variable number of p Ka's in drug molecules could be replaced by just the two straddling the isoelectric point. Another was use of the ZINC database to cull out chemically inaccessible regions of property space. A quadratic partial least squares regression (PLS) accurately fits a continuous surface to these thousands of bioavailability predictions. The PLS coefficients indicate the globally sensitive compound properties. The PLS surface also displays the %F landscape in these sensitive properties locally around compounds of particular interest. Finally, being quick to calculate, the PLS equation can be combined with models for activity and other properties for multiobjective lead optimization.


Asunto(s)
Química Farmacéutica/métodos , Descubrimiento de Drogas/métodos , Inhibidores Enzimáticos/farmacocinética , Modelos Biológicos , Relación Estructura-Actividad Cuantitativa , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , Administración Oral , Disponibilidad Biológica , Simulación por Computador , Conjuntos de Datos como Asunto , Absorción Intestinal , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Distribución Tisular
5.
Mol Pharm ; 15(3): 821-830, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29337578

RESUMEN

When medicinal chemists need to improve bioavailability (%F) within a chemical series during lead optimization, they synthesize new series members with systematically modified properties mainly by following experience and general rules of thumb. More quantitative models that predict %F of proposed compounds from chemical structure alone have proven elusive. Global empirical %F quantitative structure-property (QSPR) models perform poorly, and projects have too little data to train local %F QSPR models. Mechanistic oral absorption and physiologically based pharmacokinetic (PBPK) models simulate the dissolution, absorption, systemic distribution, and clearance of a drug in preclinical species and humans. Attempts to build global PBPK models based purely on calculated inputs have not achieved the <2-fold average error needed to guide lead optimization. In this work, local GastroPlus PBPK models are instead customized for individual medchem series. The key innovation was building a local QSPR for a numerically fitted effective intrinsic clearance (CLloc). All inputs are subsequently computed from structure alone, so the models can be applied in advance of synthesis. Training CLloc on the first 15-18 rat %F measurements gave adequate predictions, with clear improvements up to about 30 measurements, and incremental improvements beyond that.


Asunto(s)
Química Farmacéutica/métodos , Descubrimiento de Drogas/métodos , Inhibidores Enzimáticos/farmacocinética , Modelos Biológicos , Relación Estructura-Actividad Cuantitativa , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 1/antagonistas & inhibidores , Administración Oral , Animales , Disponibilidad Biológica , Células CACO-2 , Simulación por Computador , Conjuntos de Datos como Asunto , Humanos , Absorción Intestinal , Microsomas Hepáticos , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Ratas , Distribución Tisular
6.
Drug Metab Dispos ; 44(11): 1808-1818, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27538919

RESUMEN

Intestinal CYP3A and P-glycoprotein (P-gp) decrease the intestinal absorption of substrate drugs. Since substrate specificity of CYP3A often overlaps that of P-gp, and estimation of their saturability in the intestine is difficult, dose-dependent FaFg (fraction of the administered drugs that reach the portal blood) of substrate drugs and the relative importance of CYP3A and P-gp have not been clarified in many cases. Thus, we tried to establish the universal methodology for predicting the in vivo absorption of several CYP3A and/or P-gp substrates from in vitro assays. One of the key points is to set up the scaling factor (SF), correcting the difference between the observed in vivo clearance and the predicted clearance from in vitro data. The SFs of Vmax for CYP3A (SFCYP3A) and P-gp (SFP-gp) were simultaneously optimized to explain the FaFg of CYP3A and/or P-gp substrate drugs. The best predictability of FaFg was achieved when considering both SFCYP3A and SFP-gp The simulation also clarified the relative importance of CYP3A and P-gp in determining FaFg In particular, the nonlinear intestinal absorption of verapamil was caused by the saturation of intestinal CYP3A, whereas that of quinidine was governed by the saturation of both CYP3A and P-gp. In addition, the dose-dependent FaFg of selective and dual CYP3A and/or P-gp substrates was well predicted. We therefore propose a methodology for predicting the FaFg of drugs using a mathematical model with optimized SFCYP3A and SFP-gp Our methodology is applicable to in vitro-in vivo extrapolation of intestinal absorption, even if absolute in vivo functions of enzymes/transporters are unclear.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Citocromo P-450 CYP3A/metabolismo , Absorción Intestinal/fisiología , Mucosa Intestinal/metabolismo , Verapamilo/metabolismo , Disponibilidad Biológica , Células CACO-2 , Línea Celular Tumoral , Humanos , Cinética , Modelos Biológicos , Quinidina/metabolismo
7.
Pharmaceutics ; 16(1)2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38258106

RESUMEN

This study aimed to develop a physiologically based pharmacokinetic (PBPK) model that simulates metabolically cleared compounds' pharmacokinetics (PK) in pregnant subjects and fetuses. This model accounts for the differences in tissue sizes, blood flow rates, enzyme expression levels, plasma protein binding, and other physiological factors affecting the drugs' PK in both the pregnant woman and the fetus. The PBPKPlus™ module in GastroPlus® was used to model the PK of metoprolol, midazolam, and metronidazole for both non-pregnant and pregnant groups. For each of the three compounds, the model was first developed and validated against PK data in healthy non-pregnant volunteers and then applied to predict the PK in the pregnant groups. The model accurately described the PK in both the non-pregnant and pregnant groups and explained well the differences in the plasma concentration due to pregnancy. When available, the fetal plasma concentration, placenta, and fetal tissue concentrations were also predicted reasonably well at different stages of pregnancy. The work described the use of a PBPK approach for drug development and demonstrates the ability to predict differences in PK in pregnant subjects and fetal exposure for metabolically cleared compounds.

8.
Mol Pharm ; 9(3): 492-504, 2012 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-22264132

RESUMEN

The aim of this work was to extrapolate in vitro and preclinical animal data to simulate the pharmacokinetic parameters of UK-343,664, a P-glycoprotein (P-gp) and CYP3A4 substrate, in human. In addition, we aimed to develop a simulation model to demonstrate the involvement and the controversial complex interaction of intestinal P-gp and CYP3A4 in its nonlinear absorption, first-pass extraction, and pharmacokinetics using the advanced compartmental absorption and transit (ACAT) model. Finally, we aimed to compare the results predicted from the model to the reported findings in human clinical studies. In situ perfusion, allometric scaling, PBPK Rodger mechanistic approach, in vitro metabolism, and fitting to in vivo data were used to mechanistically explain the absorption, distribution and metabolism, respectively. GastroPlus was used to build the integrated simulation model in human for UK-343,664 to mechanistically explain the observed clinical data at 30, 100, 200, 400, and 800 mg oral doses. The measured in vitro value for CYP3A4 K(m) (465 µM) in rCYPs was converted to units of µg/mL, corrected for assumed microsomal binding (17.8%) and applied to all metabolic processes. The measured in vitro values of V(max) for CYP3A4 (38.9 pmol/min/pmol), 2C8, 2C9, 2C19, and 2D6 were used along with the in vitro CYP3A4 K(m) to simulate liver first pass extraction and systemic clearance. The measured in vitro values of V(max) for CYP3A4 and 2D6 were used along with the in vitro CYP3A4 K(m) to simulate gut first pass extraction. V(max) and K(m) values for P-gp were obtained by fitting to in vivo data and used to simulate gut efflux transport activity. Investigation of the interaction mechanism of P-gp and CYP3A4 in the intestine was achieved by comparing the influence of a virtual knockout of P-gp or gut metabolism on the fraction absorbed, fraction reaching the portal vein, and fraction metabolized in the gut. Comparison between simulation and in vivo results showed that the in silico simulation provided a mechanistic explanation of the observed nonlinear absorption kinetics of UK-343,664 in human following its administration in the range of 30-800 mg as oral solutions. The simulation results of the pharmacokinetic parameters, AUC and C(max), by GastroPlus were comparable with those observed in vivo. This simulation model is one possible mechanistic explanation of the observed in vivo data and suggests that the nonlinear dose dependence could be attributed to saturation of both the efflux transport by P-gp and the intestinal metabolism. However, the concentration ranges for either protein saturation did not overlap and resulted in much greater than dose proportional increases in AUC. At low doses, producing intraenterocyte concentrations below the fitted value of K(m) for P-gp, the influence of P-gp appears to be protective and results in a lower fraction of gut 3A4 metabolism. At higher doses, as P-gp becomes saturated the fraction of gut 3A4 extraction increases, and eventually at the highest doses, where 3A4 becomes saturated, the fraction of gut 3A4 extraction again decreases. Such a complex interpretation of this in vitro-in vivo extrapolation (IVIVE) is another example of the value and insight obtained by physiologically based absorption simulation.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Citocromo P-450 CYP3A/metabolismo , Piperazinas/metabolismo , Piperazinas/farmacocinética , Pirimidinonas/metabolismo , Pirimidinonas/farmacocinética , Absorción , Animales , Humanos , Absorción Intestinal/fisiología , Cinética , Masculino , Ratas , Ratas Sprague-Dawley
9.
Mol Pharm ; 9(4): 815-22, 2012 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-22352942

RESUMEN

The aim of this study was to classify some markers of common herbs used in Western medicine according to the Biopharmaceutical Classification System (BCS). The BCS is a scientific approach to classify drug substances based upon their intestinal permeability and their solubility, at the highest single dose used, within the physiologically relevant pH ranges. Known marker components of twelve herbs were chosen from the USP Dietary Supplement Compendium Monographs. Different BCS parameters such as intestinal permeability (P(eff)) and solubility (C(s)) were predicted using the ADMET Predictor, which is a software program to estimate biopharmaceutical relevant molecular descriptors. The dose number (D0) was calculated when information from the literature was available to identify an upper dose for individual markers. In these cases the herbs were classified according to the traditional BCS parameters using P(eff) and D0. When no upper dose could be determined, then the amount of a marker that is just soluble in 250 mL of water was calculated. This value, M(x), defines when a marker is changing from highly soluble to poorly soluble according to BCS criteria. This biopharmaceutically relevant value can be a useful tool for marker selection. The present study showed that a provisional BCS classification of herbs is possible but some special considerations need to be included into the classification strategy. The BCS classification can be used to choose appropriate quality control tests for products containing these markers. A provisional BCS classification of twelve common herbs and their 35 marker compounds is presented.


Asunto(s)
Biofarmacia/métodos , Preparaciones Farmacéuticas/clasificación , Plantas Medicinales/clasificación , Preparaciones Farmacéuticas/química , Plantas Medicinales/química , Solubilidad
10.
J Control Release ; 352: 833-839, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36334857

RESUMEN

This perspective article draws a distinction between some of the well-known drug classification systems and a "Chemistry Classification System" (CCS). Rather than have drug classification based on some simple properties like solubility and permeability or route of systemic elimination, a CCS results in more than four or five classes and each class has distinct properties that impact formulation development. This perspective provides and outline of 13 classes, but a CCS is a flexible system that introduces a thought process for classification. The number of classes is not rigid, and chemists are encouraged to adapt these methods to their own situations. A CCS utilizes machine-learning models and artificial intelligence (AI) to estimate physicochemical properties that result in unique, frequently observed dissolution, Absorption, Distribution, Metabolism, and Excretion (ADME) properties to guide formulation development.


Asunto(s)
Inteligencia Artificial , Aprendizaje Automático , Solubilidad , Permeabilidad
11.
J Pharm Sci ; 111(1): 262-273, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34678271

RESUMEN

Highly variable disposition after oral ingestion of acyclovir has been reported, although little is known regarding the underlying mechanisms. Different studies using the same reference product (Zovirax ®) showed that Cmax and AUC were respectively 44 and 35% lower in Saudi Arabians than Europeans, consistent with higher frequencies of reduced-activity polymorphs of the organic cation transporter (OCT1) in Europeans. In this study, the contribution of physiology (i.e., OCT1 activity) to the oral disposition of acyclovir immediate release (IR) tablets was hypothesized to be greater than dissolution. The potential role of OCT1 was studied in a validated physiologically-based biopharmaceutics model (PBBM), while dissolution of two Chilean generics (with demonstrated bioequivalence) and the reference product was assessed in vitro. The PBBM suggested that OCT1 activity could partially explain population-related pharmacokinetic differences. Further, dissolution of generics was slower than the regulatory criterion for BCS III IR products. Remarkably, virtual bioequivalence (incorporating in vitro dissolution into the PBBM) correctly and robustly predicted the bioequivalence of these products, showcasing its value in support of failed BCS biowaivers. These findings suggest that very-rapid dissolution for acyclovir IR products may not be critical for BCS biowaiver. They also endorse the relevance of cross-over designs in bioequivalence trials.


Asunto(s)
Aciclovir , Biofarmacia , Solubilidad , Comprimidos , Equivalencia Terapéutica
12.
AAPS J ; 23(4): 89, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34169370

RESUMEN

The purpose of this study was to develop a physiologically based pharmacokinetic (PBPK) model predicting the pharmacokinetics (PK) of different compounds in pregnant subjects. This model considers the differences in tissue sizes, blood flow rates, enzyme expression levels, glomerular filtration rates, plasma protein binding, and other factors affected during pregnancy in both the maternal and fetal models. The PBPKPlus™ module in GastroPlus® was used to model the PK of cefuroxime and cefazolin. For both compounds, the model was first validated against PK data in healthy non-pregnant volunteers and then applied to predict pregnant groups PK. The model accurately described the PK in both non-pregnant and pregnant groups and explained well differences in the plasma concentration due to pregnancy. The fetal plasma and amniotic fluid concentrations were also predicted reasonably well at different stages of pregnancy. This work describes the use of a PBPK approach for drug development and demonstrates the ability to predict differences in PK in pregnant subjects and fetal exposure for compounds excreted renally. The prediction for pregnant groups is also improved when the model is calibrated with postpartum or non-pregnant female group if such data are available.


Asunto(s)
Antibacterianos/farmacocinética , Feto/metabolismo , Modelos Biológicos , Complicaciones Infecciosas del Embarazo/tratamiento farmacológico , Eliminación Renal , Antibacterianos/administración & dosificación , Cefazolina/administración & dosificación , Cefazolina/farmacocinética , Cefuroxima/administración & dosificación , Cefuroxima/farmacocinética , Simulación por Computador , Desarrollo de Medicamentos/métodos , Femenino , Humanos , Riñón/metabolismo , Intercambio Materno-Fetal , Embarazo
13.
Pharmaceutics ; 13(9)2021 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-34575401

RESUMEN

Uridine 5'-diphospho-glucuronosyltransferases (UGTs) are expressed in the small intestines, but prediction of first-pass extraction from the related metabolism is not well studied. This work assesses physiologically based pharmacokinetic (PBPK) modeling as a tool for predicting intestinal metabolism due to UGTs in the human gastrointestinal tract. Available data for intestinal UGT expression levels and in vitro approaches that can be used to predict intestinal metabolism of UGT substrates are reviewed. Human PBPK models for UGT substrates with varying extents of UGT-mediated intestinal metabolism (lorazepam, oxazepam, naloxone, zidovudine, cabotegravir, raltegravir, and dolutegravir) have demonstrated utility for predicting the extent of intestinal metabolism. Drug-drug interactions (DDIs) of UGT1A1 substrates dolutegravir and raltegravir with UGT1A1 inhibitor atazanavir have been simulated, and the role of intestinal metabolism in these clinical DDIs examined. Utility of an in silico tool for predicting substrate specificity for UGTs is discussed. Improved in vitro tools to study metabolism for UGT compounds, such as coculture models for low clearance compounds and better understanding of optimal conditions for in vitro studies, may provide an opportunity for improved in vitro-in vivo extrapolation (IVIVE) and prospective predictions. PBPK modeling shows promise as a useful tool for predicting intestinal metabolism for UGT substrates.

14.
Pharm Res ; 27(12): 2646-56, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20963629

RESUMEN

PURPOSE: There are several endogenous and exogenous species in the gastrointestinal (GI) tract which can act as solubilizing agents and thereby affect drug dissolution. The purpose of this study is to understand food effects on drug dissolution and provide insight into their anticipated overall effect on absorption and bioavailability. METHODS: Dissolution kinetics of 15 drugs of variable logP, charge, and molecular weight were tested in simulated intestinal environment. The ability of a film-equilibrium-based model to predict the influence of a simulated intestinal environment on drug dissolution was examined. RESULTS: The most significant improvement in dissolution kinetics and solubility (up to 6-fold) was evident with highly hydrophobic compounds (logP > 4). Improvement in solubility did not always constitute improvement in dissolution kinetics on a relevant time scale. Comparison of simulation and experimental results indicates that a model considering micelle partitioning as a pseudo-equilibrium process can predict trends in the influence of food-related solubilizing agents on drug dissolution kinetics. CONCLUSIONS: The significance of food-related solubilizing agents to drug dissolution is not always obvious, as it depends on multiple physicochemical parameters; however, simple modeling may provide insight into food effects on dissolution and, ultimately, overall absorption and bioavailability of compounds considered for oral formulation.


Asunto(s)
Alimentos , Contenido Digestivo , Mucosa Intestinal/metabolismo , Modelos Biológicos , Farmacocinética , Cromatografía Líquida de Alta Presión , Micelas , Tamaño de la Partícula , Solubilidad
15.
Eur J Pharm Sci ; 155: 105552, 2020 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-32937212

RESUMEN

The aim of this study was to use a combined in vitro-in silico approach to develop a physiologically based pharmacokinetic model (PBPK) that predicts the bioavailability of albendazole (ABZ), a BCS class II/IV lipophilic weak base, and simulates its main metabolite albendazole sulphoxide (ABZSO) after oral administration of the current marketed dose of 400 mg in the fasted state. In vitro data was collected from solubility and dissolution tests performed with biorelevant media and transfer tests were carried out to evaluate the supersaturation and precipitation characteristics of ABZ upon gastric emptying. These in vitro results were used as biopharmaceutical inputs together with ABZ physicochemical properties including also permeability and in vitro metabolism data and information gathered from different clinical trials reported in the literature, were used to enable PBPK models to be developed using GastroPlus™ (version 9.7). As expected for this weak base with pKa = 3.6, ABZ exhibited a pronounced pH dependent solubility, with the solubility and extent of dissolution being greater at gastric pH and dropping significantly in the intestinal environment suggesting supersaturation and precipitation upon gastric emptying, which was confirmed by the transfer model experiments. PBPK models were set up for heathy volunteers using a full PBPK modeling approach and by implementing dynamic fluid volumes in the ACAT gut physiology in GastroPlus™. When coupling in vitro data (solubility values, dissolution rate and precipitation rate constant, etc.) for ABZ and with fitted values for the Vdss and liver systemic clearance of the sulfoxide metabolite to the PBPK model, the simulated profiles successfully predicated plasma concentrations of ABZ at 400 mg dose and simulated ABZSO at different ABZ dose levels and with different study populations, indicating the usefulness of combing in vitro biorelevant tools with PBPK modeling for the accurate prediction of ABZ bioavailability. The results obtained in this study also helped confirm that ABZ behaves as a BCS class IV compound.


Asunto(s)
Albendazol , Administración Oral , Albendazol/análogos & derivados , Disponibilidad Biológica , Simulación por Computador , Humanos , Solubilidad
16.
Eur J Pharm Biopharm ; 155: 55-68, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32781025

RESUMEN

In vitro dissolution experiments are used to qualitatively assess the impact of formulation composition and process changes on the drug dosage form performance. However, the use of dissolution data to quantitatively predict changes in the absorption profile remains limited. Physiologically-based Pharmacokinetic(s) (PBPK) models facilitate incorporation of in vitro dissolution experiments into mechanistic oral absorption models to predict in vivo oral formulation performance, and verify if the drug product dissolution method is biopredictive or clinically relevant. Nevertheless, a standardized approach for using dissolution data within PBPK models does not yet exist and the introduction of dissolution data in PBPK relies on a case by case approach which accommodates from differences in release mechanism and limitations to drug absorption. As part of the Innovative Medicines Initiative (IMI) Oral Biopharmaceutics Tools (OrBiTo) project a cross-work package was set up to gather a realistic understanding of various approaches used and their areas of applications. This paper presents the approaches shared by academic and industrial scientists through the OrBiTo project to integrate dissolution data within PBPK software to improve the prediction accuracy of oral formulations in vivo. Some general recommendations regarding current use and future improvements are also provided.


Asunto(s)
Simulación por Computador , Desarrollo de Medicamentos/métodos , Modelos Biológicos , Preparaciones Farmacéuticas/metabolismo , Administración Oral , Animales , Biofarmacia/métodos , Biofarmacia/tendencias , Simulación por Computador/tendencias , Desarrollo de Medicamentos/tendencias , Liberación de Fármacos/efectos de los fármacos , Liberación de Fármacos/fisiología , Predicción , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/metabolismo , Humanos , Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/fisiología , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/síntesis química , Solubilidad
17.
Eur J Pharm Biopharm ; 156: 50-63, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32805361

RESUMEN

Oral drug absorption is a complex process depending on many factors, including the physicochemical properties of the drug, formulation characteristics and their interplay with gastrointestinal physiology and biology. Physiological-based pharmacokinetic (PBPK) models integrate all available information on gastro-intestinal system with drug and formulation data to predict oral drug absorption. The latter together with in vitro-in vivo extrapolation and other preclinical data on drug disposition can be used to predict plasma concentration-time profiles in silico. Despite recent successes of PBPK in many areas of drug development, an improvement in their utility for evaluating oral absorption is much needed. Current status of predictive performance, within the confinement of commonly available in vitro data on drugs and formulations alongside systems information, were tested using 3 PBPK software packages (GI-Sim (ver.4.1), Simcyp® Simulator (ver.15.0.86.0), and GastroPlus™ (ver.9.0.00xx)). This was part of the Innovative Medicines Initiative (IMI) Oral Biopharmaceutics Tools (OrBiTo) project. Fifty eight active pharmaceutical ingredients (APIs) were qualified from the OrBiTo database to be part of the investigation based on a priori set criteria on availability of minimum necessary information to allow modelling exercise. The set entailed over 200 human clinical studies with over 700 study arms. These were simulated using input parameters which had been harmonised by a panel of experts across different software packages prior to conduct of any simulation. Overall prediction performance and software packages comparison were evaluated based on performance indicators (Fold error (FE), Average fold error (AFE) and absolute average fold error (AAFE)) of pharmacokinetic (PK) parameters. On average, PK parameters (Area Under the Concentration-time curve (AUC0-tlast), Maximal concentration (Cmax), half-life (t1/2)) were predicted with AFE values between 1.11 and 1.97. Variability in FEs of these PK parameters was relatively high with AAFE values ranging from 2.08 to 2.74. Around half of the simulations were within the 2-fold error for AUC0-tlast and around 90% of the simulations were within 10-fold error for AUC0-tlast. Oral bioavailability (Foral) predictions, which were limited to 19 APIs having intravenous (i.v.) human data, showed AFE and AAFE of values 1.37 and 1.75 respectively. Across different APIs, AFE of AUC0-tlast predictions were between 0.22 and 22.76 with 70% of the APIs showing an AFE > 1. When compared across different formulations and routes of administration, AUC0-tlast for oral controlled release and i.v. administration were better predicted than that for oral immediate release formulations. Average predictive performance did not clearly differ between software packages but some APIs showed a high level of variability in predictive performance across different software packages. This variability could be related to several factors such as compound specific properties, the quality and availability of information, and errors in scaling from in vitro and preclinical in vivo data to human in vivo behaviour which will be explored further. Results were compared with previous similar exercise when the input data selection was carried by the modeller rather than a panel of experts on each in vitro test. Overall, average predictive performance was increased as reflected in smaller AAFE value of 2.8 as compared to AAFE value of 3.8 in case of previous exercise.


Asunto(s)
Biofarmacia/normas , Análisis de Datos , Absorción Intestinal/efectos de los fármacos , Modelos Biológicos , Preparaciones Farmacéuticas/metabolismo , Programas Informáticos/normas , Administración Oral , Biofarmacia/métodos , Ensayos Clínicos como Asunto/métodos , Ensayos Clínicos como Asunto/normas , Bases de Datos Factuales/normas , Predicción , Humanos , Absorción Intestinal/fisiología , Preparaciones Farmacéuticas/administración & dosificación
18.
Pharm Res ; 26(10): 2358-66, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19685173

RESUMEN

PURPOSE: The human dipeptide transporter (hPEPT1) facilitates transport of dipeptides and drugs from the intestine into the circulation. The role of transmembrane domain 10 (TMD10) of hPEPT1 in substrate translocation was investigated using cysteine-scanning mutagenesis with 2-Trimethylammonioethyl methanethiosulfonate (MTSET). METHODS: Each amino acid in TMD10 was mutated individually to cysteine, and transport of [(3)H]Gly-Sar was evaluated with and without MTSET following transfection of each mutant in HEK293 cells. Similar localization and expression levels of wild type (WT) hPEPT1 and all mutants were confirmed by immunostaining and biotinylation followed by western blot analysis. RESULTS: E595C- and G594C-hPEPT1 showed negligible Gly-Sar uptake. E595D-hPEPT1 showed similar uptake to WT-hPEPT1, but E595K- and E595R-hPEPT1 did not transport Gly-Sar. Double mutations E595K/R282E and E595R/R282E did not restore uptake. G594A-hPEPT1 showed similar uptake to WT-hPEPT1, but G594V-hPEPT1 eliminated uptake. Y588C-hPEPT1 showed uptake of 20% that of WT-hPEPT1. MTSET modification supported a model of TMD10 with an amphipathic helix from I585 to V600 and increased solvent accessibility from T601 to F605. CONCLUSIONS: Our results suggest that G594 and E595 in TMD10 of hPEPT1 have key roles in substrate transport and that Y588 may have an important secondary mechanistic role.


Asunto(s)
Sustitución de Aminoácidos/genética , Cisteína/genética , Dipéptidos/genética , Mutagénesis Sitio-Dirigida , Simportadores/genética , Línea Celular , Membrana Celular/química , Membrana Celular/genética , Cisteína/química , Dipéptidos/química , Espacio Extracelular/química , Espacio Extracelular/genética , Ácido Glutámico/química , Ácido Glutámico/genética , Glicina/química , Glicina/genética , Humanos , Transportador de Péptidos 1 , Estructura Terciaria de Proteína/genética , Transporte de Proteínas/genética , Simportadores/química , Tirosina/química , Tirosina/genética
19.
J Pharm Sci ; 108(1): 305-315, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30395828

RESUMEN

The application of preclinical in vitro and in silico models can help formulation scientists to predict the in vivo performance of a drug in an early stage of oral drug product development. An important aspect is that these models should include equations that represent mechanisms that are biorelevant and are sensitive to changes in parameter values. Human gastrointestinal physiology involves many processes that change as a function of time. In this work, a dynamic fluid and pH model was applied in GastroPlus™ to simulate intraluminal and systemic concentrations of the weak base posaconazole in a biorelevant manner. Simulated results were compared with observed data, extracted from a previously reported human in vivo gastrointestinal aspiration study. Three different formulations were explored (i.e., 1 solution [20 mg dose strength] and 2 suspensions [both 40 mg dose strength]). Simulated results were compared and in line with the observed results for different intraluminal (e.g., precipitated fraction) and systemic parameters (e.g., plasma Cmax). The optimization of the advanced compartmental and absorption transit model related to fluid dynamics and dynamic pH in this work creates perspectives to validate this model with other reference data derived from aspiration/magnetic resonance imaging studies.


Asunto(s)
Composición de Medicamentos , Absorción Gastrointestinal/fisiología , Tracto Gastrointestinal/metabolismo , Modelos Biológicos , Triazoles/farmacocinética , Administración Oral , Simulación por Computador , Ayuno , Humanos , Concentración de Iones de Hidrógeno , Inyecciones Intravenosas , Solubilidad , Soluciones , Triazoles/administración & dosificación , Triazoles/sangre , Triazoles/química
20.
J Pharm Sci ; 108(1): 268-278, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30316900

RESUMEN

The purpose of the present study was to develop a physiologically based pharmacokinetic model for dextromethorphan (DEX) and its metabolites in extensive and poor metabolizers. The model was used to study the influence of dissolution rates on the sensitivity of maximum plasma concentration and area under the concentration-time curve for immediate release formulations. Simulation of in vitro cellular transwell permeability was used to confirm lysosomal trapping. GastroPlus™ was used to build a mechanistic absorption and physiologically based pharmacokinetic model of DEX. The model simulations were conducted with and without lysosomal trapping. The simulated results matched well with observed data only when lysosomal trapping was included. The model shows that DEX is rapidly absorbed into the enterocytes, but DEX and its metabolites only appear slowly in the portal vein and plasma, presumably due to lysosomal trapping. For this class of drug, the rate of in vitro and in vivo dissolution is not a sensitive factor in determining bioequivalence. This study shows that dissolution and the rate of absorption into the enterocytes are clinically irrelevant for the performance of DEX immediate release product. An understanding of the entire underlying mechanistic processes of drug disposition is needed to define clinically relevant product specifications for DEX.


Asunto(s)
Dextrometorfano/sangre , Dextrometorfano/química , Lisosomas/metabolismo , Modelos Biológicos , Absorción Fisiológica , Área Bajo la Curva , Células CACO-2 , Simulación por Computador , Citocromo P-450 CYP3A/genética , Enterocitos/metabolismo , Humanos , Tasa de Depuración Metabólica/genética , Permeabilidad , Polimorfismo Genético , Solubilidad , Equivalencia Terapéutica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA