Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Clin Pharmacol Ther ; 112(3): 605-614, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35355249

RESUMEN

PF-06835919 is a first-in-class ketohexokinase inhibitor (KHKi), recently under development for the treatment of metabolic and fatty liver diseases, which inhibited organic anion transporting polypeptide (OATP)1B1 in vitro and presented drug-drug interaction (DDI) risk. This study aims to investigate the dose-dependent effect of KHKi on OATP1B in vivo activity. We performed an open-label study comparing pharmacokinetics of atorvastatin (OATP1B probe) dosed alone (20 mg single dose) and coadministered with two dose strengths of KHKi (50 and 280 mg once daily) in 12 healthy participants. Additionally, changes in exposure of coproporphyrin-I (CP-I), an endogenous biomarker for OATP1B, were assessed in the atorvastatin study (1.12-fold and 1.49-fold increase in area under the plasma concentration-time profile (AUC) with once-daily 50 and 280 mg, respectively), and a separate single oral dose study of KHKi alone (100-600 mg, n = 6 healthy participants; up to a 1.80-fold increase in AUC). Geometric mean ratios (90% confidence interval) of atorvastatin AUC following 50 and 280 mg KHKi were 1.14 (1.00-1.30) and 1.54 (1.37-1.74), respectively. Physiologically-based pharmacokinetic modeling of CP-I plasma exposure following a single dose of KHKi predicted in vivo OATP1B inhibition from about 13% to 70% over the 100 to 600 mg dose range, while using the in vitro inhibition potency (1.9 µM). Model-based analysis correctly predicted "no-effect" (AUC ratio < 1.25) at the low dose range and "weak" effect (AUC ratio < 2) on atorvastatin pharmacokinetics at the high dose range of KHKi. This study exemplified the utility of biomarker-informed model-based approach in discerning even small effects on OATP1B activity in vivo, and to project DDI risk at the clinically relevant doses.


Asunto(s)
Fructoquinasas , Atorvastatina , Biomarcadores , Interacciones Farmacológicas , Fructoquinasas/metabolismo , Humanos , Transportador 1 de Anión Orgánico Específico del Hígado/metabolismo , Medición de Riesgo
2.
Curr Drug Metab ; 22(14): 1103-1113, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34915831

RESUMEN

BACKGROUND: Herbal medicine represents a significant component of disease prevention and therapy in most African countries. Herb-drug interactions (HDI) can arise from the co-administration of herbal and orthodox medicines. OBJECTIVE: This study assessed the potential for HDI of V. amygdalina, O. gratissimum, M. oleifera, A. indica, and P. nitida extracts using in vitro assays. Little is known about these medicinal plants' potential for drug interaction despite their extensive use in Nigeria for several disease conditions. METHOD: The medicinal plant crude extracts were evaluated for Cytochrome P450 (CYP) enzyme induction using cryopreserved human hepatocytes. Enzyme activity was determined by quantifying probe substrate metabolism and metabolite formation using liquid chromatography-mass spectrometry/mass spectrometry. The extracts were evaluated for the potential to inhibit P-glycoprotein (P-gp) activity using human embryonic kidney membrane vesicles over-expressing human P-gp. The herbal extracts in vivo drug interaction potential was predicted based on the USFDA drug interaction guidance. RESULT: O. gratissimum and P. nitida methanol extracts induced CYP1A2 enzyme activity by greater than 3-fold. P. nitida methanol extracts showed over 2-fold induction of CYP1A2 mRNA expression. O. gratissimum methanol extract induced CYP2B6 mRNA expression over 2-fold. P. nitida and A. indica methanol extracts showed potent inhibition of P-gp activity (IC50: 3.8 and 5.4 µg/mL), respectively, while V. amygdalina and M. oleifera methanol extracts showed moderate P-gp inhibition (IC50: 12.1 and 37.2 µg/mL, respectively). CONCLUSION: Our studies suggested that the medicinal plants' extracts can modulate CYP enzymes and P-gp activity with the potential to cause herb-drug interaction in vivo.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Inductores de las Enzimas del Citocromo P-450/farmacología , Extractos Vegetales/farmacología , Plantas Medicinales/química , Células Cultivadas , Cromatografía Liquida/métodos , Inductores de las Enzimas del Citocromo P-450/aislamiento & purificación , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Interacciones de Hierba-Droga , Humanos , Concentración 50 Inhibidora , Riñón/efectos de los fármacos , Riñón/metabolismo , Medicinas Tradicionales Africanas , Nigeria , Extractos Vegetales/administración & dosificación , Espectrometría de Masas en Tándem/métodos
3.
Eur J Drug Metab Pharmacokinet ; 46(3): 437-450, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33844145

RESUMEN

BACKGROUND AND OBJECTIVE: The use of herbal medicines is common in Africa, and patients often use a combination of herbs and drugs. Concurrent herbal and pharmaceuticals treatments can cause adverse effects through herb-drug interactions (HDI). This study evaluated the potential risk of HDI for five medicinal plants, Vernonia amygdalina, Ocimum gratissimum, Moringa oleifera, Azadirachta indica, and Picralima nitida, using in vitro assays. Patients with diabetes and some other disease conditions commonly use these medicinal plants in Nigeria, and little is known regarding their potential for drug interaction, despite their enormous use. METHODS: Crude extracts of the medicinal plants were evaluated for reversible and time-dependent inhibition (TDI) activity of six cytochrome P450 (CYP) enzymes using pooled human liver microsomes and cocktail probe-based assays. Enzyme activity was determined by quantifying marker metabolites' formation using liquid chromatography-mass spectrometry/mass spectrometry. The drug interaction potential was predicted for each herbal extract using the in vitro half-maximal inhibitory concentration (IC50) values and the percentage yield. RESULTS: O. gratissimum methanol extracts reversibly inhibited CYP 1A2, 2C8, 2C9 and 2C19 enzymes (IC50: 6.21 µg/ml, 2.96 µg/ml, 3.33 µg/ml and 1.37 µg/ml, respectively). Additionally, V. amygdalina methanol extract inhibited CYP2C8 activity (IC50: 5.71 µg/ml); P. nitida methanol and aqueous extracts inhibited CYP2D6 activity (IC50: 1.99 µg/ml and 2.36 µg/ml, respectively) while A. indica methanol extract inhibited CYP 3A4/5, 2C8 and 2C9 activity (IC50: 7.31 µg/ml, 9.97 µg/ml and 9.20 µg/ml, respectively). The extracts showed a potential for TDI of the enzymes when incubated at 200 µg/ml; V. amygdalina and A. indica methanol extracts exhibited TDI potential for all the major CYPs. CONCLUSIONS: The medicinal plants inhibited CYP activity in vitro, with the potential to cause in vivo HDI. Clinical risk assessment and proactive monitoring are recommended for patients who use these medicinal plants concurrently with drugs that are cleared through CYP metabolism.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/farmacología , Hipoglucemiantes/farmacología , Extractos Vegetales/farmacología , Plantas Medicinales/química , Cromatografía Liquida , Inhibidores Enzimáticos del Citocromo P-450/administración & dosificación , Inhibidores Enzimáticos del Citocromo P-450/aislamiento & purificación , Diabetes Mellitus/tratamiento farmacológico , Femenino , Interacciones de Hierba-Droga , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/aislamiento & purificación , Técnicas In Vitro , Concentración 50 Inhibidora , Masculino , Microsomas Hepáticos/metabolismo , Nigeria , Extractos Vegetales/administración & dosificación , Espectrometría de Masas en Tándem
4.
Drug Metab Dispos ; 49(6): 470-478, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-33824168

RESUMEN

About 30% of approved drugs are cleared predominantly by renal clearance (CLr). Of these, many are secreted by transporters. For these drugs, in vitro-to-in vivo extrapolation of transporter-mediated renal secretory clearance (CLsec,plasma) is important to prospectively predict their renal clearance and to assess the impact of drug-drug interactions and pharmacogenetics on their pharmacokinetics. Here we compared the ability of the relative expression factor (REF) and the relative activity factor (RAF) approaches to quantitatively predict the in vivo CLsec,plasma of 26 organic anion transporter (OAT) substrates assuming that OAT-mediated uptake is the rate-determining step in the CLsec,plasma of the drugs. The REF approach requires protein quantification of each transporter in the tissue (e.g., kidney) and transporter-expressing cells, whereas the RAF approach requires the use of a transporter-selective probe substrate (both in vitro and in vivo) for each transporter of interest. For the REF approach, 50% and 69% of the CLsec,plasma predictions were within 2- and 3-fold of the observed values, respectively; the corresponding values for the RAF approach were 65% and 81%. We found no significant difference between the two approaches in their predictive capability (as measured by accuracy and bias) of the CLsec,plasma or CLr of OAT drugs. We recommend that the REF and RAF approaches can be used interchangeably to predict OAT-mediated CLsec,plasma Further research is warranted to evaluate the ability of the REF or RAF approach to predict CLsec,plasma of drugs when uptake is not the rate-determining step. SIGNIFICANCE STATEMENT: This is the first direct comparison of the relative expression factor (REF) and relative activity factor (RAF) approaches to predict transporter-mediated renal clearance (CLr). The RAF, but not REF, approach requires transporter-selective probes and that the basolateral uptake is the rate-determining step in the CLr of drugs. Given that there is no difference in predictive capability of the REF and RAF approach for organic anion transporter-mediated CLr, the REF approach should be explored further to assess its ability to predict CLr when basolateral uptake is not the sole rate-determining step.


Asunto(s)
Vías de Eliminación de Fármacos/fisiología , Interacciones Farmacológicas , Transportadores de Anión Orgánico , Eliminación Renal/efectos de los fármacos , Transporte Biológico/fisiología , Desarrollo de Medicamentos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Transportadores de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico/farmacocinética , Preparaciones Farmacéuticas/metabolismo , Farmacocinética , Valor Predictivo de las Pruebas
5.
J Clin Pharmacol ; 56 Suppl 7: S110-21, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27385169

RESUMEN

With numerous drugs cleared renally, inhibition of uptake transporters localized on the basolateral membrane of renal proximal tubule cells, eg, organic anion transporters (OATs) and organic cation transporters (OCTs), may lead to clinically meaningful drug-drug interactions (DDIs). Additionally, clinical evidence for the possible involvement of efflux transporters, such as P-glycoprotein (P-gp) and multidrug and toxin extrusion protein 1/2-K (MATE1/2-K), in the renal DDIs is emerging. Herein, we review recent progress regarding mechanistic understanding of transporter-mediated renal DDIs as well as the quantitative predictability of renal DDIs using static and physiologically based pharmacokinetic (PBPK) models. Generally, clinical DDI data suggest that the magnitude of plasma exposure changes attributable to renal DDIs is less than 2-fold, unlike the DDIs associated with inhibition of cytochrome P-450s and/or hepatic uptake transporters. It is concluded that although there is a need for risk assessment early in drug development, current available data imply that safety concerns related to the renal DDIs are generally low. Nevertheless, consideration must be given to the therapeutic index of the victim drug and potential risk in a specific patient population (eg, renal impairment). Finally, in vitro transporter data and clinical pharmacokinetic parameters obtained from the first-in-human studies have proven useful in support of quantitative prediction of DDIs associated with inhibition of renal secretory transporters, OATs or OCTs.


Asunto(s)
Interacciones Farmacológicas/fisiología , Riñón/metabolismo , Transportadores de Anión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Preparaciones Farmacéuticas/metabolismo , Animales , Evaluación Preclínica de Medicamentos/métodos , Predicción , Humanos , Riñón/efectos de los fármacos , Tasa de Depuración Metabólica/efectos de los fármacos , Tasa de Depuración Metabólica/fisiología , Preparaciones Farmacéuticas/administración & dosificación
6.
Drug Metab Dispos ; 44(2): 209-19, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26608081

RESUMEN

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.


Asunto(s)
Acetamidas/farmacocinética , Pirimidinonas/farmacocinética , Tiouracilo/farmacocinética , Administración Oral , Animales , Disponibilidad Biológica , Células CACO-2 , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Perros , Evaluación Preclínica de Medicamentos/métodos , Femenino , Células HEK293 , Haplorrinos , Hepatocitos/metabolismo , Humanos , Absorción Intestinal/fisiología , Masculino , Ratones , Microsomas Hepáticos/metabolismo , Peroxidasa/metabolismo , Unión Proteica , Ratas , Ratas Wistar
7.
Expert Opin Drug Metab Toxicol ; 9(4): 459-72, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23331046

RESUMEN

INTRODUCTION: Membrane transporters have been recognized to play a key role in determining the absorption, distribution and elimination processes of drugs. The organic anion-transporting polypeptide (OATP)1B1 and OATP1B3 isoforms are selectively expressed in the human liver and are known to cause significant drug-drug interactions (DDIs), as observed with an increasing number of drugs. It is evident that DDIs involving hepatic transporters are capable of altering systemic, as well as tissue-specific, exposure of drug substrates resulting in marked differences in drug safety and/or efficacy. It is therefore essential to quantitatively predict such interactions early in the drug development to mitigate clinical risks. AREAS COVERED: The role of hepatic uptake transporters in drug disposition and clinical DDIs has been reviewed with an emphasis on the current state of the models applicable for quantitative predictions. The readers will also gain insight into the in vitro experimental tools available to characterize transport kinetics, while appreciating the knowledge gaps in the in vitro-in vivo extrapolation (IVIVE), which warrant further investigation. EXPERT OPINION: Static and dynamic models can be convincingly applied to quantitatively predict drug interactions, early in drug discovery, to mitigate clinical risks as well as to avoid unnecessary clinical studies. Compared to basic models, which focus on individual processes, mechanistic models provide the ability to assess DDI potential for compounds with systemic disposition determined by both transporters and metabolic enzymes. However, complexities in the experimental tools and an apparent disconnect in the IVIVE of transport kinetics have limited the physiologically based pharmacokinetic modeling strategies. Emerging data on the expression of transporter proteins and tissue drug concentrations are expected to help bridge these gaps. In addition, detailed characterization of substrate kinetics can facilitate building comprehensive mechanistic models.


Asunto(s)
Hígado/citología , Hígado/efectos de los fármacos , Proteínas de Transporte de Membrana/efectos de los fármacos , Línea Celular , Fenómenos Químicos , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Fluorobencenos/farmacocinética , Fluorobencenos/uso terapéutico , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Cinética , Hígado/metabolismo , Modelos Teóricos , Pravastatina/farmacocinética , Pravastatina/uso terapéutico , Pirimidinas/farmacocinética , Pirimidinas/uso terapéutico , Rosuvastatina Cálcica , Sulfonamidas/farmacocinética , Sulfonamidas/uso terapéutico
8.
Expert Opin Drug Metab Toxicol ; 6(8): 939-52, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20433402

RESUMEN

IMPORTANCE OF THE FIELD: Kidney plays a key role in the elimination of xenobiotics and metabolic products from the body, where renal clearance is determined by glomerular filtration, tubular secretion and reabsorption processes. The proximal tubule of the nephron is equipped with multi-specificity uptake and efflux transporters for the secretion of a broad range of xenobiotics, while the compound physicochemical space drives the tubular reabsorption. Due to involvement of transporters, renal clearance is possibly associated with renal drug-drug interactions (DDIs) in clinical situations. Nevertheless, renal insufficiency in diseased population is associated with altered transporter activity and evidently affects the pharmacokinetics of both renally and non-renally cleared compounds. Thus, early information on renal clearance is critical for successful development of compounds in certain chemical space. AREAS COVERED IN THIS REVIEW: This review provides updated information on the influence of physicochemical properties and the relevance of transporters in renal clearance and the associated drug interactions. In silico tools to predict the renal clearance at the discovery stage along with the potential alterations in drug disposition in the renal disease state are discussed with preclinical and clinical examples. WHAT THE READER WILL GAIN: The review provides comprehensive knowledge with recent examples to moderate renal disposition concerns in the drug discovery and development settings. TAKE HOME MESSAGE: Consideration of clearance pathways early in the discovery process has become critical for successful development of compounds. Although significant progress has been made in elucidating the physicochemical drivers and biochemical processes for this pathway, the predictive ability remains a challenge. Furthermore, development of renally cleared compounds should progress with a clear understanding of possible issues including transporter-mediated DDI and disease state.


Asunto(s)
Diseño de Fármacos , Riñón/metabolismo , Preparaciones Farmacéuticas/metabolismo , Animales , Transporte Biológico , Ensayos Clínicos como Asunto , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Humanos
9.
Pharmacol Res ; 48(4): 347-59, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12902205

RESUMEN

Drug efflux pumps like P-glycoprotein (P-gp) and multidrug resistance (MDR) proteins were recognized to possess functional role in determining the pharmacokinetics of drugs administered by peroral as well as parenteral route. Advancements in molecular biology, to some extent, had revealed the structure, localization and functional role of P-glycoprotein and its mechanism of drug efflux. Broad substrate recognition by this protein and clinical implications of its inhibition has revolutionized cancer chemotherapy leading to design and development of novel P-glycoprotein inhibitors. In the recent times, the application of these inhibitors in improving peroral drug delivery has gained special interest. Inhibition of P-glycoprotein improves intestinal absorption and tissue distribution while reducing the substrate metabolism and its elimination. Eventually, various screening methodologies have been developed for determining the activity of P-glycoprotein, kinetics of drug transport and identification of substrates and inhibitors. In the present review, techniques used for screening P-glycoprotein inhibitors and the scope of these inhibitors in optimizing peroral drug absorption and pharmacokinetics are discussed along with a brief introduction to P-glycoprotein, its physiological function and active role in extrusion of drugs.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/farmacocinética , Evaluación Preclínica de Medicamentos/métodos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/química , Animales , Disponibilidad Biológica , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/fisiología , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA