Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Ann Allergy Asthma Immunol ; 133(3): 310-317.e4, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38697286

RESUMEN

BACKGROUND: Long-term tezepelumab treatment in the DESTINATION study (NCT03706079) resulted in reduced asthma exacerbations, reduced biomarker levels, and improved lung function and symptom control in patients with severe, uncontrolled asthma. OBJECTIVE: To explore the time course of changes in biomarkers and clinical manifestations after treatment cessation after 2 years of tezepelumab treatment. METHODS: DESTINATION was a 2-year, phase 3, multicenter, randomized, placebo-controlled, double-blind study of tezepelumab treatment in patients (12-80 years old) with severe asthma. Patients received their last treatment doses at week 100 and could enroll in an extended follow-up period from weeks 104 to 140. Change over time in key biomarkers and clinical outcomes were assessed in tezepelumab vs placebo recipients for 40 weeks after stopping treatment. RESULTS: Of 569 patients enrolled in the extended follow-up period, 426 were included in the analysis (289 received tezepelumab and 137 placebo). In the 40-week period after the last tezepelumab dose, blood eosinophil counts, fractional exhaled nitric oxide levels, and Asthma Control Questionnaire-6 scores gradually increased from weeks 4 to 10, with a gradual reduction in pre-bronchodilator forced expiratory volume in 1 second such that blood eosinophil counts, fractional exhaled nitric oxide levels, and clinical outcomes returned to placebo levels; however, none of these outcomes returned to baseline levels. Total IgE levels increased later from week 28 and remained well below placebo and baseline levels during the 40-week period after the last tezepelumab dose. CONCLUSION: This analysis reveals the benefits of continued tezepelumab treatment in the management of patients with severe, uncontrolled asthma, compared with stopping treatment after 2 years. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT03706079.


Asunto(s)
Antiasmáticos , Anticuerpos Monoclonales Humanizados , Asma , Biomarcadores , Humanos , Asma/tratamiento farmacológico , Persona de Mediana Edad , Masculino , Femenino , Adulto , Antiasmáticos/uso terapéutico , Antiasmáticos/administración & dosificación , Anciano , Estudios de Seguimiento , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/administración & dosificación , Método Doble Ciego , Resultado del Tratamiento , Adolescente , Anciano de 80 o más Años , Adulto Joven , Niño , Inmunoglobulina E/sangre , Eosinófilos/inmunología , Eosinófilos/efectos de los fármacos
2.
Drug Metab Dispos ; 46(10): 1420-1433, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30068519

RESUMEN

Physiologically based pharmacokinetic (PBPK) modeling for itraconazole using a bottom-up approach is challenging, not only due to complex saturable pharmacokinetics (PK) and the presence of three metabolites exhibiting CYP3A4 inhibition, but also because of discrepancies in reported in vitro data. The overall objective of this study is to provide a comprehensive mechanistic PBPK model for itraconazole in order to increase the confidence in its drug-drug interaction (DDI) predictions. To achieve this, key in vitro and in vivo data for itraconazole and its major metabolites were generated. These data were crucial to developing a novel bottom-up PBPK model in Simcyp (Simcyp Ltd., Certara, Sheffield, United Kingdom) for itraconazole and two of its major metabolites: hydroxy-itraconazole (OH-ITZ) and keto-itraconazole (keto-ITZ). Performance of the model was validated using prespecified acceptance criteria against different dosing regimens, formulations for 29 PK, and DDI studies with midazolam and other CYP3A4 substrates. The main outcome is an accurate PBPK model that simultaneously predicts the PK profiles of itraconazole, OH-ITZ, and keto-ITZ. In addition, itraconazole DDIs with midazolam and other CYP3A4 substrates were successfully predicted within a 2-fold error. Prediction precision and bias of DDI expressed as geometric mean fold error were for the area under the concentration-time curve and peak concentration, 1.06 and 0.96, respectively. To conclude, in this paper a comprehensive data set for itraconazole and its metabolites is provided that enables bottom-up mechanism-based PBPK modeling. The presented model is applicable for studying the contribution from the metabolites and allows improved assessments of itraconazole DDI.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Interacciones Farmacológicas , Itraconazol , Animales , Inhibidores del Citocromo P-450 CYP3A/metabolismo , Inhibidores del Citocromo P-450 CYP3A/farmacocinética , Humanos , Itraconazol/metabolismo , Itraconazol/farmacocinética , Masculino , Midazolam/metabolismo , Midazolam/farmacocinética , Unión Proteica , Ratas , Ratas Wistar
3.
Xenobiotica ; 44(6): 498-510, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24320817

RESUMEN

1. The metabolite profile of the 5α-reductase type II inhibitor finasteride has been studied in pig plasma, urine and bile using high-resolution mass spectrometry. The porcine biotransformation products were compared to those formed by human liver microsomes and to literature data of recently identified human in vivo metabolites. The objective of this study was to gain further evidence for the validity of using pigs for advanced, invasive drug-drug interaction studies that are not possible to perform in humans. 2. The use of high-resolution mass spectrometry with accurate mass measurements enabled identification of the metabolites by calculation of their elemental compositions as well as their fragmentation patterns. 3. There was an excellent match between the porcine and human metabolic profiles, corroborating the pig as a model of human drug metabolism. The glucuronides of the two recently described human hydroxylated metabolites MX and MY and the carboxylated metabolite M3 were identified as the major biotransformation products of finasteride in pig urine and bile. 4. Furthermore, the CYP enzymes involved in the formation of the hydroxylated metabolites were characterized. Human recombinant CYP3A4 could produce the two major hydroxylated metabolites MX and MY, whereas human recombinant CYP2D6 formed MY only.


Asunto(s)
Finasterida/análisis , Finasterida/metabolismo , Espectrometría de Masas/métodos , Fase II de la Desintoxicación Metabólica , Fase I de la Desintoxicación Metabólica , Sus scrofa/sangre , Sus scrofa/orina , Animales , Bilis , Cromatografía Líquida de Alta Presión , Sistema Enzimático del Citocromo P-450/metabolismo , Finasterida/sangre , Finasterida/orina , Humanos , Microsomas Hepáticos/metabolismo , Peso Molecular , Estándares de Referencia
4.
Clin Pharmacol Ther ; 114(4): 825-835, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37376792

RESUMEN

A different drug-drug interaction (DDI) scenario may exist in patients with chronic kidney disease (CKD) compared with healthy volunteers (HVs), depending on the interplay between drug-drug and disease (drug-drug-disease interaction (DDDI)). Physiologically-based pharmacokinetic (PBPK) modeling, in lieu of a clinical trial, is a promising tool for evaluating these complex DDDIs in patients. However, the prediction confidence of PBPK modeling in the severe CKD population is still low when nonrenal pathways are involved. More mechanistic virtual disease population and robust validation cases are needed. To this end, we aimed to: (i) understand the implications of severe CKD on statins (atorvastatin, simvastatin, and rosuvastatin) pharmacokinetics (PK) and DDI; and (ii) predict untested clinical scenarios of statin-roxadustat DDI risks in patients to guide suitable dose regimens. A novel virtual severe CKD population was developed incorporating the disease effect on both renal and nonrenal pathways. Drug and disease PBPK models underwent a four-way validation. The verified PBPK models successfully predicted the altered PKs in patients for substrates and inhibitors and recovered the observed statin-rifampicin DDIs in patients and the statin-roxadustat DDIs in HVs within 1.25- and 2-fold error. Further sensitivity analysis revealed that the severe CKD effect on statins PK is mainly mediated by hepatic BCRP for rosuvastatin and OATP1B1/3 for atorvastatin. The magnitude of statin-roxadustat DDI in patients with severe CKD was predicted to be similar to that in HVs. PBPK-guided suitable dose regimens were identified to minimize the risk of side effects or therapeutic failure of statins when co-administered with roxadustat.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas , Insuficiencia Renal Crónica , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Atorvastatina , Rosuvastatina Cálcica/efectos adversos , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Proteínas de Neoplasias , Interacciones Farmacológicas , Modelos Biológicos , Simulación por Computador
5.
CPT Pharmacometrics Syst Pharmacol ; 11(9): 1194-1209, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35722750

RESUMEN

Physiologically-based pharmacokinetic (PBPK) models have an important role in drug discovery/development and decision making in regulatory submissions. This is facilitated by predefined PBPK platforms with user-friendly graphical interface, such as Simcyp and PK-Sim. However, evaluations of platform differences and the potential implications for disposition-related applications are still lacking. The aim of this study was to assess how PBPK model development, input parameters, and model output are affected by the selection of PBPK platform. This is exemplified via the establishment of simvastatin PBPK models (workflow, final models, and output) in PK-Sim and Simcyp as representatives of established whole-body PBPK platforms. The major finding was that the choice of PBPK platform influenced the model development strategy and the final model input parameters, however, the predictive performance of the simvastatin models was still comparable between the platforms. The main differences between the structure and implementation of Simcyp and PK-Sim were found in the absorption and distribution models. Both platforms predicted equally well the observed simvastatin (lactone and acid) pharmacokinetics (20-80 mg), BCRP and OATP1B1 drug-gene interactions (DGIs), and drug-drug interactions (DDIs) when co-administered with CYP3A4 and OATP1B1 inhibitors/inducers. This study illustrates that in-depth knowledge of established PBPK platforms is needed to enable an assessment of the consequences of PBPK platform selection. Specifically, this work provides insights on software differences and potential implications when bridging PBPK knowledge between Simcyp and PK-Sim users. Finally, it provides a simvastatin model implemented in both platforms for risk assessment of metabolism- and transporter-mediated DGIs and DDIs.


Asunto(s)
Modelos Biológicos , Simvastatina , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Simulación por Computador , Interacciones Farmacológicas , Humanos , Proteínas de Neoplasias , Farmacocinética
6.
Drug Metab Dispos ; 39(5): 847-57, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21317368

RESUMEN

The overall aim of this detailed investigation of the pharmacokinetics (PK) and metabolism of finasteride in pigs was to improve understanding of in vivo PK for this drug and its metabolites. Specific aims were to examine the effects of ketoconazole coadministration on the PK in three plasma compartments (the portal, hepatic, and femoral veins), bile, and urine and to use these data to study in detail the intestinal absorption and the liver extraction ratio and apply a semiphysiological based PK model to the data. The pigs received an intrajejunal dose of finasteride (0.8 mg/kg) either alone (n = 5) or together with ketoconazole (10 mg/kg) (n = 5) or an intravenous dose (0.2 mg/kg) (n = 3). Plasma, bile, and urine (collected from 0 to 6 h) were analyzed with ultraperformance liquid chromatography-tandem mass spectrometry. Ketoconazole increased the bioavailability of finasteride from 0.36 ± 0.23 to 0.91 ± 0.1 (p < 0.05) and the terminal half-life from 1.6 ± 0.4 to 4.0 ± 1.1 h (p < 0.05). From deconvolution, it was found that the absorption rate from the intestine to the portal vein was rapid, and the product of the fraction absorbed and the fraction that escaped gut wall metabolism was high (f(a) · F(G) ∼ 1). Interestingly, the apparent absorption rate constant (k(a)) to the femoral vein was lower than that to the portal vein, probably because of binding and distribution within the liver. The liver extraction ratio was time-dependent and varied with the two routes of administration. After intrajejunal administration, from 1 to 6 h, the liver extraction ratio was significantly (p < 0.05) reduced by ketoconazole treatment from intermediate (0.41 ± 0.21) to low (0.21 ± 0.10).


Asunto(s)
Inhibidores de 14 alfa Desmetilasa/metabolismo , Inhibidores de 5-alfa-Reductasa/metabolismo , Bilis/metabolismo , Finasterida/metabolismo , Absorción Intestinal , Cetoconazol/farmacología , Hígado/metabolismo , Inhibidores de 14 alfa Desmetilasa/farmacología , Inhibidores de 5-alfa-Reductasa/administración & dosificación , Inhibidores de 5-alfa-Reductasa/sangre , Inhibidores de 5-alfa-Reductasa/farmacocinética , Animales , Interacciones Farmacológicas , Finasterida/administración & dosificación , Finasterida/farmacocinética , Finasterida/farmacología , Semivida , Inyecciones Intravenosas , Absorción Intestinal/efectos de los fármacos , Cetoconazol/administración & dosificación , Hígado/efectos de los fármacos , Masculino , Neoplasias de la Próstata/prevención & control , Porcinos
7.
Drug Metab Dispos ; 39(2): 239-46, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20978106

RESUMEN

Ketoconazole has been shown in clinical trials to increase the plasma exposure of the oral anticoagulant prodrug AZD0837 [(2S)-N-{4- [(Z)-amino(methoxyimino)methyl]benzyl}-1-{(2R)-2-[3-chloro-5-(difluoromethoxy)phenyl]-2-hydroxyethanoyl}-azetidine-2-carboxamide] and its active metabolite, AR-H067637 [(2S)-N-{4-[amino(imino)methyl]benzyl}-1-{(2R)-2-[3-chloro-5-(difluoromethoxy)phenyl]-2-hydroxyethanoyl}-azetidine-2-carboxamide]. To investigate the biotransformation of AZD0837 and the effect of ketoconazole on this process, we used an experimental model in pigs that allows repeated sampling from three blood vessels, the bile duct, and a perfused intestinal segment. The pigs received AZD0837 (500 mg) given enterally either alone (n = 5) or together with single-dose ketoconazole (600 mg) (n = 6). The prodrug (n = 2) and its active metabolite (n = 2) were also administered intravenously to provide reference doses. The plasma data revealed considerable interindividual variation in the exposure of the prodrug, intermediate metabolite, and active metabolite. However, AR-H067637 was detected at very high concentrations in the bile with low variability (Ae(bile) = 53 ± 6% of the enteral dose), showing that the compound had indeed been formed in all of the animals and efficiently transported into the bile canaliculi. Concomitant dosing with ketoconazole increased the area under the plasma concentration-time curve for AZD0837 (by 99%) and for AR-H067637 (by 51%). The effect on the prodrug most likely arose from inhibited CYP3A-mediated metabolism. Reduced metabolism also seemed to explain the increased plasma exposure of the active compound because ketoconazole prolonged the terminal half-life with no apparent effect on the extensive biliary excretion and biliary clearance. These in vivo results were supported by in vitro depletion experiments for AR-H067637 in pig liver microsomes with and without the addition of ketoconazole.


Asunto(s)
Amidinas/farmacocinética , Antitrombinas/farmacocinética , Azetidinas/farmacocinética , Bilis/metabolismo , Mucosa Intestinal/metabolismo , Cetoconazol/farmacología , Hígado/metabolismo , Amidinas/administración & dosificación , Amidinas/sangre , Amidinas/farmacología , Animales , Antitrombinas/administración & dosificación , Antitrombinas/sangre , Antitrombinas/farmacología , Azetidinas/administración & dosificación , Azetidinas/sangre , Azetidinas/farmacología , Transporte Biológico/efectos de los fármacos , Biotransformación , Interacciones Farmacológicas , Cetoconazol/administración & dosificación , Masculino , Estructura Molecular , Perfusión , Porcinos
8.
CPT Pharmacometrics Syst Pharmacol ; 9(7): 384-394, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32438492

RESUMEN

Intradermal delivery of AZD8601, an mRNA designed to produce vascular endothelial growth factor A (VEGF-A), has previously been shown to accelerate cutaneous wound healing in a murine diabetic model. Here, we develop population pharmacokinetic and pharmacodynamic models aiming to quantify the effect of AZD8601 injections on the dynamics of wound healing. A dataset of 584 open wound area measurements from 131 mice was integrated from 3 independent studies encompassing different doses, dosing timepoints, and number of doses. Evaluation of several candidate models showed that wound healing acceleration is not likely driven directly by time-dependent VEGF-A concentration. Instead, we found that administration of AZD8601 induced a sustained acceleration of wound healing depending on the accumulated dose, with a dose producing 50% of the maximal effect of 92 µg. Simulations with this model showed that a single dose of 200 µg AZD8601 can reduce the time to reach 50% wound healing by up to 5 days.


Asunto(s)
Diabetes Mellitus Experimental/terapia , ARN Mensajero/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/genética , Cicatrización de Heridas/genética , Animales , Diabetes Mellitus Experimental/complicaciones , Ratones , Modelos Biológicos , ARN Mensajero/genética , Factores de Tiempo
9.
Drug Metab Dispos ; 37(10): 2008-17, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19635781

RESUMEN

The objective of this study was to further investigate the metabolism of the 5alpha-reductase inhibitor, finasteride, and to identify previously unknown phase I and phase II metabolites in vitro and in vivo in human bile and urine. Healthy volunteers were given 5 mg of finasteride, directly to the intestine, and bile and urine were collected for 3 and 24 h, respectively. A single-pass perfusion technique, Loc-I-Gut, was used for drug administration and bile collection from the proximal jejunum, distal to papilla of Vater. Incubations with human liver microsomes/S9 fractions and different cofactors were performed with finasteride and the previously known metabolites, omega-hydroxy finasteride (M1) and finasteride-omega-oic acid (M3). Liquid chromatography coupled to triple quadrupole mass spectrometry (MS) with positive/negative electrospray ionization and ion trap with MS(n) measurements were used for structural investigations and identification of metabolites. Two hydroxy metabolites of finasteride, other than M1, and one intact hydroxy finasteride glucuronide were identified in vitro and in bile and urine. The glucuronide and at least one of the hydroxy metabolites were previously unidentified. M1 and M3 were glucuronidated in vitro by specific human UDP-glucuronosyltransferases, UGT1A4 and UGT1A3, respectively. M1 glucuronide was not identified in vivo, and M3 glucuronide, an acyl glucuronide, was present in low amounts in bile from a few individuals. In conclusion, previously undescribed metabolites were identified, in vitro and in human urine and bile. Bile collection using the Loc-I-Gut technique followed by sensitive mass spectrometry analysis led to the discovery of novel, both phase I and phase II, finasteride metabolites in human bile.


Asunto(s)
Bilis/química , Cromatografía Líquida de Alta Presión/métodos , Finasterida/análisis , Glucurónidos/metabolismo , Espectrometría de Masas en Tándem/métodos , Finasterida/metabolismo , Glucuronosiltransferasa/metabolismo , Humanos , Masculino , Microsomas Hepáticos/metabolismo , Células Tumorales Cultivadas
10.
Drug Metab Dispos ; 37(12): 2349-58, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19773540

RESUMEN

The hepatobiliary transport and local disposition of rosuvastatin in pig were investigated, along with the impact of concomitant dosing with two known multiple transport inhibitors; cyclosporine and gemfibrozil. Rosuvastatin (80 mg) was administered as an intrajejunal bolus dose in treatments I, II, and III (TI, TII, and TIII, respectively; n = 6 per treatment). Cyclosporine (300 mg) and gemfibrozil (600 mg) were administered in addition to the rosuvastatin dose in TII and TIII, respectively. Cyclosporine was administered as a 2-h intravenous infusion and gemfibrozil as an intrajejunal bolus dose. In treatment IV (TIV, n = 4) 5.9 mg of rosuvastatin was administered as an intravenous bolus dose. The study was conducted using a pig model, which enabled plasma sampling from the portal (VP), hepatic (VH), and femoral veins and bile from the common hepatic duct. The biliary recoveries of the administered rosuvastatin dose were 9.0 +/- 3.5 and 35.7 +/- 15.6% in TI and TIV, respectively. Rosuvastatin was highly transported into bile as shown by the median AUC(bile)/AUC(VH) ratio in TI of 1770 (1640-11,300). Gemfibrozil did not have an effect on the plasma pharmacokinetics of rosuvastatin, most likely because the unbound inhibitor concentrations did not exceed the reported IC(50) values. However, cyclosporine significantly reduced the hepatic extraction of rosuvastatin (TI, 0.89 +/- 0.06; TII, 0.46 +/- 0.13) and increased the AUC(VP) and AUC(VH) by 1.6- and 9.1-fold, respectively. In addition, the biliary exposure and f(e, bile) were reduced by approximately 50%. The strong effect of cyclosporine was in accordance with inhibition of sinusoidal uptake transporters, such as members of the organic anion-transporting polypeptide family, rather than canalicular transporters.


Asunto(s)
Bilis/metabolismo , Ciclosporina/farmacología , Circulación Enterohepática/efectos de los fármacos , Fluorobencenos/farmacocinética , Gemfibrozilo/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacocinética , Hígado/efectos de los fármacos , Pirimidinas/farmacocinética , Sulfonamidas/farmacocinética , Administración Oral , Animales , Área Bajo la Curva , Ciclosporina/administración & dosificación , Femenino , Fluorobencenos/administración & dosificación , Fluorobencenos/sangre , Gemfibrozilo/administración & dosificación , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/sangre , Infusiones Intravenosas , Inyecciones Intravenosas , Absorción Intestinal/efectos de los fármacos , Yeyuno/efectos de los fármacos , Yeyuno/metabolismo , Hígado/metabolismo , Masculino , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Modelos Biológicos , Orquiectomía , Pirimidinas/administración & dosificación , Pirimidinas/sangre , Rosuvastatina Cálcica , Sulfonamidas/administración & dosificación , Sulfonamidas/sangre , Porcinos
11.
Br J Pharmacol ; 175(11): 2116-2129, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29574682

RESUMEN

BACKGROUND AND PURPOSE: Drugs metabolically eliminated by several enzymes are less vulnerable to variable compound exposure in patients due to drug-drug interactions (DDI) or if a polymorphic enzyme is involved in their elimination. Therefore, it is vital in drug discovery to accurately and efficiently estimate and optimize the metabolic elimination profile. EXPERIMENTAL APPROACH: CYP3A and/or CYP2D6 substrates with well described variability in vivo in humans due to CYP3A DDI and CYP2D6 polymorphism were selected for assessment of fraction metabolized by each enzyme (fmCYP ) in two in vitro systems: (i) human recombinant P450s (hrP450s) and (ii) human hepatocytes combined with selective P450 inhibitors. Increases in compound exposure in poor versus extensive CYP2D6 metabolizers and by the strong CYP3A inhibitor ketoconazole were mathematically modelled and predicted changes in exposure were compared with in vivo data. KEY RESULTS: Predicted changes in exposure were within twofold of reported in vivo values using fmCYP estimated in human hepatocytes and there was a strong linear correlation between predicted and observed changes in exposure (r2  = 0.83 for CYP3A, r2  = 0.82 for CYP2D6). Predictions using fmCYP in hrP450s were not as accurate (r2  = 0.55 for CYP3A, r2  = 0.20 for CYP2D6). CONCLUSIONS AND IMPLICATIONS: The results suggest that variability in human drug exposure due to DDI and enzyme polymorphism can be accurately predicted using fmCYP from human hepatocytes and CYP-selective inhibitors. This approach can be efficiently applied in drug discovery to aid optimization of candidate drugs with a favourable metabolic elimination profile and limited variability in patients.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Hepatocitos/efectos de los fármacos , Adulto , Anciano , Anciano de 80 o más Años , Inhibidores Enzimáticos del Citocromo P-450/química , Femenino , Humanos , Ligandos , Masculino , Persona de Mediana Edad , Estructura Molecular , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Adulto Joven
12.
Sci Rep ; 8(1): 17509, 2018 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-30504800

RESUMEN

Capable of mediating efficient transfection and protein production without eliciting innate immune responses, chemically modified mRNA holds great potential to produce paracrine factors at a physiologically beneficial level, in a spatiotemporally controlled manner, and with low toxicity. Although highly promising in cardiovascular medicine and wound healing, effects of this emerging therapeutic on the microvasculature and its bioactivity in disease settings remain poorly understood. Here, we longitudinally and comprehensively characterize microvascular responses to AZD8601, a modified mRNA encoding vascular endothelial growth factor A (VEGF-A), in vivo. Using multi-parametric photoacoustic microscopy, we show that intradermal injection of AZD8601 formulated in a biocompatible vehicle results in pronounced, sustained and dose-dependent vasodilation, blood flow upregulation, and neovessel formation, in striking contrast to those induced by recombinant human VEGF-A protein, a non-translatable variant of AZD8601, and citrate/saline vehicle. Moreover, we evaluate the bioactivity of AZD8601 in a mouse model of diabetic wound healing in vivo. Using a boron nanoparticle-based tissue oxygen sensor, we show that sequential dosing of AZD8601 improves vascularization and tissue oxygenation of the wound bed, leading to accelerated re-epithelialization during the early phase of diabetic wound healing.


Asunto(s)
Angiopatías Diabéticas/etiología , Angiopatías Diabéticas/patología , Microvasos/metabolismo , ARN Mensajero/genética , Factor A de Crecimiento Endotelial Vascular/genética , Cicatrización de Heridas/genética , Animales , Angiopatías Diabéticas/diagnóstico por imagen , Modelos Animales de Enfermedad , Humanos , Ratones , Microvasos/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Neovascularización Patológica/diagnóstico por imagen , Neovascularización Patológica/genética , Consumo de Oxígeno , Imagen de Lapso de Tiempo , Cicatrización de Heridas/efectos de los fármacos
13.
Eur J Pharm Sci ; 43(3): 89-98, 2011 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-21447389

RESUMEN

Pigs are becoming increasingly used as a test animal both in pharmacological and toxicological assessment of new drug compounds. For interspecies comparisons and predictions it is important to characterize the expression and function of membrane transport and enzymatic proteins in pigs, particularly at a mechanistic level which will make extrapolation of observation between pig and man to be made with more confidence. The major objective of this report was to increase the integrative knowledge of drug metabolism in pigs and to compare with corresponding data from human liver microsomes. This was done by using human substrates of CYP3A4 (verapamil and testosterone), CYP2C9 (diclofenac) and CYP2D6 (dextromethorphan). In addition, the mRNA expression of important drug metabolizing enzymes and carrier-mediated transporters were assessed in intestine and liver tissues from pigs. It was shown that CYP3A4 activity is quantitatively comparable between the two species but data suggest that qualitative differences may exist. Verapamil showed similar metabolism pattern as in humans and the CYP3A4 inhibitor ketoconazole was able to inhibit the depletion of both R- and S-verapamil. A correlation between individual pig CYP3A mRNA expression and in vivo hepatic extraction ratio was established which indicates that CYP3A is the major determinant factor in both pigs and humans. However, investigations of the metabolism of testosterone resulted in qualitative different metabolite pattern between pigs and humans. The metabolism of diclofenac was very low in pig liver microsomes and did not correlate to corresponding activity in human liver microsomes. In contrast dextromethorphan exhibited a very extensive and rapid metabolism in pig liver microsomes compared to human data. Together with previously determined gene expression data it confirms that CYP2D6 substrates will be very rapidly metabolized in pigs. The mRNA data increased the knowledge of the interindividual variability and the relative expression of different enzymes and transporters in pig intestine and liver. In conclusion, this study has increased the understanding of similarities and differences between pig and human biotransformation of drugs by providing new data for four different model compounds.


Asunto(s)
Citocromo P-450 CYP2D6 , Citocromo P-450 CYP3A , Yeyuno/enzimología , Hígado/enzimología , Testosterona/metabolismo , Verapamilo/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Hidrocarburo de Aril Hidroxilasas/metabolismo , Biotransformación , Citocromo P-450 CYP2C9 , Citocromo P-450 CYP2D6/genética , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Inhibidores del Citocromo P-450 CYP3A , Dextrometorfano/metabolismo , Diclofenaco/metabolismo , Expresión Génica , Glucuronosiltransferasa/metabolismo , Humanos , Cetoconazol/farmacología , Masculino , Microsomas Hepáticos/enzimología , ARN Mensajero/metabolismo , Especificidad de la Especie , Porcinos
14.
Eur J Pharm Sci ; 36(4-5): 433-43, 2009 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-19073252

RESUMEN

The aim of this study was to investigate what the consequences of induced drug metabolism, caused by St. John's wort (SJW, Hypericum perforatum) treatment, would have on the plasma, biliary and urinary pharmacokinetics of finasteride and its two previously identified phase I metabolites (hydroxy-finasteride and carboxy-finasteride). Twelve healthy men were administered 5mg finasteride directly to the intestine via a catheter with a multi-channel tubing system, Loc-I-Gut, before and after 14 days SJW (300mg b.i.d, hyperforin 4%) treatment. Bile samples were withdrawn via the Loc-I-Gut device from the proximal jejunum. LC-ESI-MS/MS was used to analyze finasteride and its metabolites in plasma, bile and urine. HPLC-UV was used to analyze hyperforin in plasma. The herbal treatment significantly reduced the peak plasma concentration (C(max)), the area under the plasma concentration-time curve (AUC(0-24h)) and the elimination half-life (t(1/2)) of finasteride. The geometric mean ratios (90% CI) were 0.42 (0.36-0.49), 0.66 (0.56-0.79) and 0.54 (0.48-0.61), respectively. Finasteride was excreted unchanged to a minor extent into bile and urine. Hydroxy-finasteride was not detected in plasma, bile or urine. Carboxy-finasteride was quantified in all three compartments and its plasma pharmacokinetics was significantly affected by SJW treatment. Hyperforin concentration in plasma was 21+/-7ng/ml approximately 12h after the last dose of the 14 days SJW treatment. In conclusion, SJW treatment for 2 weeks induced the metabolism of finasteride and caused a reduced plasma exposure of the drug. New knowledge was gained about the biliary and urinary excretion or the drug and its metabolites.


Asunto(s)
Sistema Biliar/metabolismo , Inhibidores Enzimáticos/farmacocinética , Finasterida/farmacocinética , Hypericum , Adulto , Compuestos Bicíclicos con Puentes/sangre , Cromatografía Líquida de Alta Presión , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/orina , Finasterida/sangre , Finasterida/orina , Semivida , Humanos , Masculino , Floroglucinol/análogos & derivados , Floroglucinol/sangre , Valores de Referencia , Espectrometría de Masa por Ionización de Electrospray , Espectrofotometría Ultravioleta , Espectrometría de Masas en Tándem , Terpenos/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA