Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Drug Metab Dispos ; 49(7): 540-547, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33863817

RESUMEN

Clinical induction liability is assessed with human hepatocytes. However, underpredictions in the magnitude of clinical induction have been reported. Unfortunately, in vivo studies in animals do not provide additional insight because of species differences in drug metabolizing enzymes and their regulatory pathways. To circumvent this limitation, transgenic animals expressing human orthologs were developed. The aim of this work was to investigate the utility of mouse models expressing human orthologs of pregnane X receptor, constitutive androstane receptor, and CYP3A4/7 (Tg-Composite) in evaluating clinical induction. Rifampin, efavirenz, and pioglitazone, which were employed to represent strong, moderate, and weak inducers, were administered at multiple doses to Tg-Composite animals. In vivo CYP3A activity was monitored by measuring changes in the exposure of the CYP3A probe substrate triazolam. After the in vivo studies, microsomes were prepared from their livers to measure changes of in vitro CYP3A4 activity. In both in vivo and in vitro, distinction of clinic induction was recapitulated as rifampin yielded the greatest inductive effect followed by efavirenz and pioglitazone. Interestingly, with rifampin, in vivo CYP3A activity was approximately 4-fold higher than in vitro activity. Conversely, there was no difference between in vivo and in vitro CYP3A activity with efavirenz. These findings are consistent with the report that, although rifampin exhibits differential inductive effects between the intestines and liver, efavirenz does not. These data highlight the promise of transgenic models, such as Tg-Composite, to complement human hepatocytes to enhance the translatability of clinical induction as well as become a powerful tool to further study mechanisms of drug disposition. SIGNIFICANCE STATEMENT: Underprediction of the magnitude of clinical induction when using human hepatocytes has been reported, and transgenic models may improve clinical translatability. The work presented here showcases the human orthologs of pregnane X receptor, constitutive androstane receptor, and CYP3A4/7 model, which was able to recapitulate the magnitude of clinical induction and to differentiate tissue-dependent induction observed with rifampin but not with efavirenz. These results not only foreshadow the potential application of such transgenic models in assessing clinical induction but also in further investigation of the mechanism of drug disposition.


Asunto(s)
Inductores del Citocromo P-450 CYP3A/farmacocinética , Alquinos/administración & dosificación , Alquinos/farmacocinética , Animales , Benzoxazinas/administración & dosificación , Benzoxazinas/farmacocinética , Receptor de Androstano Constitutivo/genética , Receptor de Androstano Constitutivo/metabolismo , Ciclopropanos/administración & dosificación , Ciclopropanos/farmacocinética , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Inductores del Citocromo P-450 CYP3A/administración & dosificación , Evaluación Preclínica de Medicamentos/métodos , Interacciones Farmacológicas , Estudios de Factibilidad , Femenino , Humanos , Ratones , Ratones Transgénicos , Microsomas Hepáticos , Pioglitazona/administración & dosificación , Pioglitazona/farmacocinética , Receptor X de Pregnano/genética , Receptor X de Pregnano/metabolismo , Rifampin/administración & dosificación , Rifampin/farmacocinética , Especificidad de la Especie , Triazolam/administración & dosificación , Triazolam/farmacocinética
2.
Xenobiotica ; 49(5): 602-610, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-29768081

RESUMEN

1. Oatp inhibitors have been shown to significantly increase the plasma exposure of statins. However, understanding alterations of liver concentration is also important. While modeling has simulated liver concentration changes, availability of experimental data is limited, especially when concerning drug-drug interactions (DDI). The objective of this work was to determine blood and liver concentrations of fluvastatin, lovastatin and pitavastatin, when blocking uptake transporters. 2. In wild-type mouse, rifampin pre-treatment decreased the unbound liver-to-plasma ratio (Kp,uu) of fluvastatin by 4.2-fold to 2.2, lovastatin by 4.9-fold to 0.81 and pitavastatin by 10-fold to 0.21. Changes in Kp,uu were driven by increases in systemic exposures as liver concentrations were not greatly altered. 3. In Oatp1a/1b knockout mouse (KO), rifampin exerted no additional effect on fluvastatin and lovastatin. Contrarily, rifampin further decreased pitavastatin Kp,uu by 3.4-fold, suggesting that the KO is inadequate to completely block liver uptake of pitavastatin as there are additional rifampin-sensitive uptake mechanism(s) not captured in the KO model. 4. This work provides experimental data showing that the plasma compartment is more sensitive to Oatp modulation than the liver compartment, even for rifampin-mediated DDI. Consistent with previous simulations, inhibiting or targeting Oatps may change Kp,uu, but exhibit only a minimal effect on absolute liver concentrations.


Asunto(s)
Fluvastatina , Hígado/metabolismo , Lovastatina , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Quinolinas , Animales , Fluvastatina/farmacocinética , Fluvastatina/farmacología , Lovastatina/farmacocinética , Lovastatina/farmacología , Ratones , Ratones Noqueados , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Quinolinas/farmacocinética , Quinolinas/farmacología
3.
Mol Pharm ; 14(5): 1754-1759, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28345929

RESUMEN

Species differences in the expression, activity, regulation, and substrate specificity of metabolizing enzymes preclude the use of animal models to predict clinical drug-drug interactions (DDIs). The objective of this work is to determine if the transgenic (Tg) Cyp3a-/-Tg-3A4Hep/Int and Nr1i2/Nr1i3-/--Cyp3a-/-Tg-PXR-CAR-3A4/3A7Hep/Int (PXR-CAR-CYP3A4/3A7) mouse models could be used to predict in vivo DDI of 10 drugs; alprazolam, bosutinib, crizotinib, dasatinib, gefitinib, ibrutinib, regorafenib, sorafenib, triazolam, and vandetinib (as victims); with varying magnitudes of reported CYP3A4 clinical DDI. As an assessment of the effect of CYP3A4 inhibition, these drugs were coadministered to Cyp3a-/-Tg-3A4Hep/Int mice with the CYP3A inhibitor, itraconazole. For crizotinib, regorafenib, sorafenib, and vandetanib, there was no significant increase of AUC observed; with alprazolam, bosutinib, ibrutinib, dasatinib, and triazolam, pretreatment with itraconazole resulted in a 2-, 4-, 17-, 7-, and 15-fold increase in AUC, respectively. With the exception of gefinitib for which the DDI effect was overpredicted (12-fold in Tg-mice vs 2-fold in the clinic), the magnitude of AUC increase observed in this study was consistent (within 2-fold) with the clinical DDI observed following administration with itraconazole/ketoconazole. As an assessment of CYP3A4 induction, following rifampin pretreatment to PXR-CAR-3A4/3A7Hep/Int mice, an 8% decrease in vandetanib mean AUC was observed; 39-52% reduction in AUC were observed for dasatinib, ibrutinib, regorafenib, and sorafenib compared to vehicle treated mice. The greatest effect of rifampin induction was observed with alprazolam, bosutinib, crizotinib, gefitinib, and triazolam where 72-91% decrease in AUC were observed. With the exception of vandetanib for which rifampin induction was under-predicted, the magnitude of induction observed in this study was consistent (within 2-fold) with clinical observations. These data sets suggest that, with two exceptions, these transgenic mice models were able to exclude or capture the magnitude of CYP3A4 clinical inhibition and induction. Data generated in transgenic mice may be used to gain confidence and complement in vitro and in silico methods for assessing DDI potential/liability.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Interacciones Farmacológicas , Alprazolam/metabolismo , Compuestos de Anilina/metabolismo , Animales , Cromatografía Liquida , Receptor de Androstano Constitutivo , Crizotinib , Dasatinib/metabolismo , Femenino , Humanos , Itraconazol/metabolismo , Cetoconazol/metabolismo , Ratones , Ratones Transgénicos , Nitrilos/metabolismo , Piperidinas/metabolismo , Pirazoles/metabolismo , Piridinas/metabolismo , Quinazolinas/metabolismo , Quinolinas/metabolismo , Rifampin/metabolismo , Espectrometría de Masas en Tándem , Triazolam/metabolismo
4.
Drug Metab Dispos ; 44(11): 1736-1741, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27538915

RESUMEN

Animals are not commonly used to assess drug-drug interactions due to poor clinical translatability arising from species differences that may exist in drug-metabolizing enzymes and transporters, and their regulation pathways. In this study, a transgenic mouse model expressing human pregnane X receptor (PXR), constitutive androstane receptor (CAR), CYP3A4/CYP3A7, and CYP2D6 (Tg-composite) was used to investigate the effect of induction mediated by rifampin on the pharmacokinetics of tamoxifen and its metabolites. In humans, tamoxifen is metabolized primarily by CYP3A4 and CYP2D6, and multiple-day treatment with rifampin decreased tamoxifen exposure by 6.2-fold. Interestingly, exposure of tamoxifen metabolites 4-hydroxytamoxifen (4OHT), N-desmethyltamoxifen (NDM), and endoxifen also decreased. In the Tg-composite model, pretreatment with rifampin decreased tamoxifen area under the time-concentration curve between 0 and 8 hours (AUC0-8) from 0.82 to 0.20 µM*h, whereas AUC0-8 of 4OHT, NDM, and endoxifen decreased by 3.4-, 4.7-, and 1.3-fold, respectively, mirroring the clinic observations. In the humanized PXR-CAR (hPXR-CAR) model, rifampin decreased AUC0-8 of tamoxifen and its metabolites by approximately 2-fold. In contrast, no significant modulation by rifampin was observed in the nonhumanized C57BL/6 (wild-type) animals. In vitro kinetics determined in microsomes prepared from livers of the Tg-composite animals showed that, although Km values were not different between vehicle- and rifampin-treated groups, rifampin increased the Vmax for the CYP3A4-mediated pathways. These data demonstrate that, although the hPXR-CAR model is responsive to rifampin, the extent of the clinical rifampin-tamoxifen interaction is better represented by the Tg-composite model. Consequently, the Tg-composite model may be a suitable tool to examine the extent of rifampin-mediated induction for other compounds whose metabolism is mediated by CYP3A4 and/or CYP2D6.


Asunto(s)
Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP3A/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Esteroides/metabolismo , Rifampin/farmacología , Tamoxifeno/metabolismo , Animales , Receptor de Androstano Constitutivo , Femenino , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/metabolismo , Receptor X de Pregnano , Tamoxifeno/análogos & derivados
5.
Drug Metab Lett ; 10(2): 91-100, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27063863

RESUMEN

BACKGROUND: Significant under-prediction of in vivo clearance in rat was observed for a potent p21-activated kinase (PAK1) inhibitor, GNE1. OBJECTIVE: Rate-determining (rapid uptake) and rate-limiting (slow excretion) steps in systemic clearance and elimination of GNE1, respectively, were evaluated to better understand the cause of the in vitro-in vivo (IVIV) disconnect. METHODS: A series of in vivo, ex vivo, and in vitro experiments were carried out: 1) the role of organic cation transporters (Oct or Slc22a) was investigated in transporter knock-out and wild-type animals with or without 1-aminobenzotriazole (ABT) pretreatment; 2) the concentration-dependent hepatic extraction ratio was determined in isolated perfused rat liver; and 3) excreta were collected from both bile duct cannulated and non-cannulated rats after intravenous injection. RESULTS: After intravenous dosing, the rate-determining step in clearance was found to be mediated by the active uptake transporter, Oct1. In cannulated rats, biliary and renal clearance of GNE1 accounted for only approximately 14 and 16% of the total clearance, respectively. N-acetylation, an important metabolic pathway, accounted for only about 10% of the total dose. In non-cannulated rats, the majority of the dose was recovered in feces as unchanged parent (up to 91%) overnight following intravenous administration. CONCLUSION: Because the clearance of GNE1 is mediated through uptake transporters rather than metabolism, the extrahepatic expression of Oct1 in kidney and intestine in rat likely plays an important role in the IVIV disconnect in hepatic clearance prediction. The slow process of intestinal secretion is the rate-limiting step for in vivo clearance of GNE1.


Asunto(s)
Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/metabolismo , Hígado/metabolismo , Inhibidores de Proteínas Quinasas/farmacocinética , Pirazoles/farmacocinética , Pirimidinas/farmacocinética , Quinasas p21 Activadas/antagonistas & inhibidores , Animales , Proteínas de Transporte de Catecolaminas en la Membrana Plasmática/genética , Femenino , Masculino , Ratones , Ratones Noqueados , Proteínas de Transporte de Catión Orgánico/metabolismo , Ratas , Ratas Sprague-Dawley , Triazoles/farmacología
6.
ILAR J ; 57(2): 157-165, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-28053069

RESUMEN

Improved small molecule bioanalytical sensitivity and concomitant decreased sample volume requirements provide an opportunity to reconsider how toxicokinetic (TK) data are collected in rat toxicity studies. Often, satellite groups of rats are designated to separate procedural effects of TK blood collection from the primary toxicity evaluation. Blood microsampling (i.e., ≤50 µL) decreases the blood volume collected such that TK samples can be collected from toxicity groups without impacting toxicity assessment. Small plasma sampling uses slightly higher blood volumes (i.e., 200 µL) with comparable technical feasibility and, importantly, allows multiple analyses with no negative impact on study interpretation. Our "base case" study designs utilize sparse TK sampling from sample toxicity group rats (1-2 samples/rat). Alternate designs with satellite animals may still be warranted based on study objectives (e.g., biomarkers), intolerability, or smaller rat strains; however, we propose these as exceptions rather than standard practice and with a focus to use the fewest animals possible. We review the state of knowledge in bioanalytical and blood sampling techniques and support the paradigm whereby TK sampling of main study animals significantly decreases the overall number of rats required for toxicity assessments and refines study interpretation with additional data options. These efforts maintain a commitment to the 3Rs (replacement, reduction, and refinement) while maintaining high-quality TK evaluations on toxicity studies.


Asunto(s)
Recolección de Muestras de Sangre/métodos , Toxicocinética , Animales , Biomarcadores/sangre , Recolección de Muestras de Sangre/instrumentación , Ratas
7.
Drug Metab Dispos ; 43(6): 864-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25813936

RESUMEN

Data from the clinical absolute bioavailability (F) study with cobimetinib suggested that F was lower than predicted based on its low hepatic extraction and good absorption. The CYP3A4 transgenic (Tg) mouse model with differential expression of CYP3A4 in the liver (Cyp3a(-/-)Tg-3A4Hep) or intestine (Cyp3a(-/-)Tg-3A4Int) and both liver and intestine (Cyp3a(-/-)Tg-3A4Hep/Int) were used to study the contribution of intestinal metabolism to the F of cobimetinib. In addition, the effect of CYP3A4 inhibition and induction on cobimetinib exposures was tested in the Cyp3a(-/-)Tg-3A4Hep/Int and PXR-CAR-CYP3A4/CYP3A7 mouse models, respectively. After i.v. administration of 1 mg/kg cobimetinib to wild-type [(WT) FVB], Cyp3a(-/-)Tg-3A4Hep, Cyp3a(-/-)Tg-3A4Int, or Cyp3a(-/-)Tg-3A4Hep/Int mice, clearance (CL) (26-35 ml/min/kg) was similar in the CYP3A4 transgenic and WT mice. After oral administration of 5 mg/kg cobimetinib, the area under the curve (AUC) values of cobimetinib in WT, Cyp3a(-/-)Tg-3A4Hep, Cyp3a(-/-)Tg-3A4Int, or Cyp3a(-/-)Tg-3A4Hep/Int mice were 1.35, 3.39, 1.04, and 0.701 µM⋅h, respectively. The approximately 80% lower AUC of cobimetinib in transgenic mice when intestinal CYP3A4 was present suggested that the intestinal first pass contributed to the oral CL of cobimetinib. Oxidative metabolites observed in human circulation were also observed in the transgenic mice. In drug-drug interaction (DDI) studies using Cyp3a(-/-)Tg-3A4Hep/Int mice, 8- and 4-fold increases in oral and i.v. cobimetinib exposure, respectively, were observed with itraconazole co-administration. In PXR-CAR-CYP3A4/CYP3A7 mice, rifampin induction decreased cobimetinib oral exposure by approximately 80%. Collectively, these data support the conclusion that CYP3A4 intestinal metabolism contributes to the oral disposition of cobimetinib and suggest that under certain circumstances the transgenic model may be useful in predicting clinical DDIs.


Asunto(s)
Antineoplásicos/farmacocinética , Azetidinas/farmacocinética , Citocromo P-450 CYP3A/metabolismo , Mucosa Intestinal/enzimología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Microsomas Hepáticos/enzimología , Piperidinas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacocinética , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/sangre , Hidrocarburo de Aril Hidroxilasas/genética , Hidrocarburo de Aril Hidroxilasas/metabolismo , Azetidinas/administración & dosificación , Azetidinas/sangre , Disponibilidad Biológica , Citocromo P-450 CYP3A/química , Citocromo P-450 CYP3A/genética , Inductores del Citocromo P-450 CYP3A/efectos adversos , Inhibidores del Citocromo P-450 CYP3A/efectos adversos , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Femenino , Semivida , Humanos , Inyecciones Intravenosas , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Tasa de Depuración Metabólica , Ratones Noqueados , Ratones Transgénicos , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Piperidinas/administración & dosificación , Piperidinas/sangre , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/sangre , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
8.
J Pharm Sci ; 104(4): 1508-21, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25615572

RESUMEN

Advanced tissue composition-based models can predict the tissue-plasma partition coefficient (Kp ) values of drugs under in vivo conditions on the basis of in vitro and physiological input data. These models, however, focus on healthy tissues and do not incorporate data from tumors. The objective of this study was to apply a tissue composition-based model to six marketed antineoplastic drugs (docetaxel, DOC; doxorubicin, DOX; gemcitabine, GEM; methotrexate, MTX; topotecan, TOP; and fluorouracil, 5-FU) to predict their Kp values in three human tumor xenografts (HCT-116, H2122, and PC3) as well as in healthy tissues (brain, muscle, lung, and liver) under steady-state in vivo conditions in female NCR nude mice. The mechanisms considered in the tissue/tumor composition-based model are the binding to lipids and to plasma proteins, but the transporter effect was also investigated. The method consisted of analyzing tissue composition, performing the pharmacokinetics studies in mice, and calculating the corresponding in vivo Kp values. Analyses of tumor composition indicated that the tumor xenografts contained no or low amounts of common transporters by contrast to lipids. The predicted Kp values were within twofold and threefold of the measured values in 77% and 93% of cases, respectively. However, predictions for brain for each drug, for liver for MTX, and for each tumor xenograft for GEM were disparate from the observed values, and, therefore, not well served by the model. Overall, this study is the first step toward the mechanism-based prediction of Kp values of small molecules in healthy and tumor tissues in mouse when no transporter and permeation limitation effect is evident. This approach will be useful in selecting compounds based on their abilities to penetrate human cancer xenografts with a physiologically based pharmacokinetic (PBPK) model, thereby increasing therapeutic index for chemotherapy in oncology study.


Asunto(s)
Antineoplásicos/farmacocinética , Neoplasias del Colon/metabolismo , Neoplasias Pulmonares/metabolismo , Modelos Biológicos , Neoplasias de la Próstata/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/sangre , Antineoplásicos/química , Neoplasias del Colon/sangre , Neoplasias del Colon/patología , Perros , Femenino , Células HCT116 , Xenoinjertos , Humanos , Infusiones Intravenosas , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/patología , Células de Riñón Canino Madin Darby , Masculino , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones Desnudos , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/patología , Distribución Tisular , Transfección
9.
Mol Pharm ; 11(11): 4199-207, 2014 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-25243894

RESUMEN

Cobimetinib is a MEK inhibitor currently in clinical trials as an anticancer agent. The objectives of this study were to determine in vitro and in vivo if cobimetinib is a substrate of P-glycoprotein (P-gp) and/or breast cancer resistance protein (Bcrp1) and to assess the implications of efflux on cobimetinib pharmacokinetics (PK), brain penetration, and target modulation. Cell lines transfected with P-gp or Bcrp1 established that cobimetinib was a substrate of P-gp but not a substrate of Bcrp1. In vivo, after intravenous and oral administration of cobimetinib to FVB (wild-type; WT), Mdr1a/b(-/-), Bcrp1 (-/-), and Mdr1a/b(-/-)/Bcrp(-/-) knockout (KO) mice, clearance was similar in WT (35.5 ± 16.7 mL/min/kg) and KO animals (22.0 ± 3.6 to 27.6 ± 5.2 mL/min/kg); oral exposure was also similar between WT and KO animals. After an oral 10 mg/kg dose of cobimetinib, the mean total brain to plasma ratio (Kp) at 6 h postdose was 0.3 and 0.2 in WT and Bcrp1(-/-) mice, respectively. In Mdr1a/b(-/-) and Mdr1a/1b/Bcrp1(-/-) KO mice and WT mice treated with elacridar (a P-gp and BCRP inhibitor), Kp increased to 11, 6, and 7, respectively. Increased brain exposure in Mdr1a/b(-/-) and Mdr1a/1b/Bcrp1(-/-) KO and elacridar treated mice was accompanied by up to ∼65% suppression of the target (pErk) in brain tissue, compared to WT mice. By MALDI imaging, the cobimetinib signal intensity was relatively high and was dispersed throughout the brain of Mdr1a/1b/Bcrp1(-/-) KO mice compared to low/undetectable signal intensity in WT mice. The efflux of cobimetinib by P-gp may have implications for the treatment of patients with brain tumors/metastases.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/fisiología , Transportadoras de Casetes de Unión a ATP/fisiología , Azetidinas/farmacocinética , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/metabolismo , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Piperidinas/farmacocinética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Animales , Azetidinas/farmacología , Transporte Biológico , Encéfalo/efectos de los fármacos , Cromatografía Liquida , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Femenino , Ratones , Ratones Noqueados , Piperidinas/farmacología , Espectrometría de Masas en Tándem , Distribución Tisular , Miembro 4 de la Subfamilia B de Casete de Unión a ATP
10.
Bioanalysis ; 6(11): 1445-57, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25046046

RESUMEN

BACKGROUND: NAD(+) is an endogenous analyte and is unstable during blood sample collection, both of which present obstacles for quantitation. Moreover, current procedures for NAD(+) sample collection require onsite treatment with strong acid to stabilize the NAD(+) in mouse blood cells. RESULTS: NAD(+) can be stabilized by addition of acid before the frozen mouse blood sample was thawed. A simple sample collection procedure was proposed to facilitate the analysis of NAD(+) in mouse blood and tissue samples. A LC-MS/MS method was developed for quantifying NAD(+) in mouse blood and various tissue samples. The described method was used to measure endogenous NAD(+) levels in mouse blood following oral administration of the nicotinamide phosphoribosyltransferase inhibitor GNE-617. CONCLUSION: This study presents a suitable assay and sample collection procedure for high throughput screening of NAD(+) samples in preclinical discovery studies.


Asunto(s)
Cromatografía Liquida/métodos , Espectrometría de Masas/métodos , NAD/sangre , Animales , Recolección de Muestras de Sangre , Ratones
11.
Drug Metab Dispos ; 42(6): 1067-73, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24671957

RESUMEN

Atorvastatin is eliminated by CYP3A4 which follows carrier-mediated uptake into hepatocytes by OATP1B1, OATP1B3, and OATP2B1. Multiple clinical studies demonstrated that OATP inhibition by rifampin had a greater impact on atorvastatin systemic concentration than itraconazole-mediated CYP3A4 inhibition. If it is assumed that the blood and hepatocyte compartments are differentiated by the concentration gradient that is established by OATPs, and if the rate of uptake into the hepatocyte is rate-determining to the elimination of atorvastatin from the body, then it is hypothesized that blood concentrations may not necessarily reflect liver concentrations. In wild-type mice, rifampin had a greater effect on systemic exposure of atorvastatin than ketoconazole, as the blood area under the blood concentration-time curve increased 7- and 2-fold, respectively. In contrast, liver concentrations were affected more by ketoconazole than by rifampin, as liver levels increased 21- and 4-fold, respectively. Similarly, in Cyp3a knockout animals, 39-fold increases in liver concentrations were observed despite insignificant changes in the blood area under the blood concentration-time curve. Interestingly, blood and liver levels in Oatp1a/b knockout animals were similar to wild types, suggesting that Oatp1a/b knockout may be necessary but not sufficient to completely describe atorvastatin uptake in mice. Data presented in this work indicate that there is a substantial drug interaction when blocking atorvastatin metabolism, but the effects of this interaction are predominantly manifested in the liver and may not be captured when monitoring changes in the systemic circulation. Consequently, there may be a disconnect when trying to relate blood exposure to instances of hepatotoxicity because a pharmacokinetic-toxicity relationship may not be obvious from blood concentrations.


Asunto(s)
Sistema Enzimático del Citocromo P-450/deficiencia , Ácidos Heptanoicos/sangre , Cetoconazol/farmacocinética , Hígado/metabolismo , Proteína 1 de Transporte de Anión Orgánico/deficiencia , Pirroles/sangre , Rifampin/farmacocinética , Animales , Atorvastatina , Citocromo P-450 CYP3A , Interacciones Farmacológicas/fisiología , Femenino , Células HEK293 , Ácidos Heptanoicos/metabolismo , Humanos , Hígado/efectos de los fármacos , Ratones , Ratones Noqueados , Pirroles/metabolismo
12.
Mol Pharm ; 11(3): 1062-8, 2014 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-24450768

RESUMEN

CYP Oxidoreductase (Por) is the essential electron donor for all CYP enzymes and is responsible for the activation of CYP. The Taconic Hepatic CYP Reductase Null (HRN) mouse model possesses a targeted mutation that results in liver-specific deletion of the Por gene thereby resulting in a disruption of CYP metabolism in the liver. The objectives of these studies were to further characterize the HRN mouse using probe drugs metabolized by CYP. In addition, tumor exposure in xenograft tumor bearing HRN immune-compromised (nude) mice was also determined. In HRN mice following intravenous (iv) administration of midazolam, clearance (CL) was reduced by ∼ 80% compared to wild-type mice (WT). After oral administration, the AUC of midazolam was increased by ∼ 20-fold in HRN mice compared to WT mice; this greater effect suggests that hepatic first pass plays a role in the oral CL of midazolam. A 50% and an 80% decrease in CL were also observed in HRN mice following iv administration of docetaxel and theophylline, respectively, compared to WT mice. In addition, a 2- to 3-fold increase in tumor concentrations of G4222, a tool compound, were observed in tumor bearing HRN nude mice compared to tumor bearing nude WT mice. The observations from these experiments demonstrate that, for compounds that are extensively metabolized by hepatic CYP, the HRN mouse model could potentially be valuable for evaluating in vivo efficacy of tool compounds in drug discovery where high hepatic CL and low exposure may prevent in vivo evaluation of a new chemical entity.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Descubrimiento de Drogas , Fibrosarcoma/metabolismo , Midazolam/farmacocinética , NADPH-Ferrihemoproteína Reductasa/fisiología , Animales , Antineoplásicos/farmacocinética , Docetaxel , Femenino , Fibrosarcoma/tratamiento farmacológico , Hipnóticos y Sedantes/farmacocinética , Hígado/enzimología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Taxoides/farmacocinética , Teofilina/farmacocinética , Vasodilatadores/farmacocinética
13.
Drug Metab Dispos ; 42(3): 343-51, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24389420

RESUMEN

Vismodegib (Erivedge, GDC-0449) is a first-in-class, orally administered small-molecule Hedgehog pathway inhibitor that is approved for the treatment of advanced basal cell carcinoma. Previously, we reported results from preclinical and clinical radiolabeled mass balance studies in which we determined that metabolism is the main route of vismodegib elimination. The metabolites of vismodegib are primarily the result of oxidation followed by glucuronidation. The focus of the current work is to probe the mechanisms of formation of three pyridine ring-cleaved metabolites of vismodegib, mainly M9, M13, and M18, using in vitro, ex vivo liver perfusion and in vivo rat studies. The use of stable-labeled ((13)C2,(15)N)vismodegib on the pyridine ring exhibited that the loss of carbon observed in both M9 and M13 was from the C-6 position of pyridine. Interestingly, the source of the nitrogen atom in the amide of M9 was from the pyridine. Evidence for the formation of aldehyde intermediates was observed using trapping agents as well as (18)O-water. Finally, we conclude that cytochrome P450 is involved in the formation of M9, M13, and M18 and that M3 (the major mono-oxidative metabolite) is not the precursor for the formation of these cleaved products; rather, M18 is the primary cleaved metabolite.


Asunto(s)
Anilidas/metabolismo , Piridinas/metabolismo , Anilidas/química , Animales , Biotransformación , Cromatografía Líquida de Alta Presión , Perros , Proteínas Hedgehog/antagonistas & inhibidores , Humanos , Técnicas In Vitro , Hígado/efectos de los fármacos , Hígado/enzimología , Macaca fascicularis , Masculino , Espectrometría de Masas , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Estructura Molecular , Oxidación-Reducción , Perfusión , Piridinas/química , Ratas , Ratas Sprague-Dawley
14.
Xenobiotica ; 44(3): 235-41, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24138296

RESUMEN

1. Modern high-throughput small molecule drug discovery requires rapid screening of the pharmacokinetic parameters of multiple candidate molecules in parallel. The mouse is often used for such screening, as are solvent-based intravenous formulations. Despite this, the intravenous toxicity of many commonly used solvents is unknown. The purpose of this investigation is to establish recommended no-observed-effect level (NOEL) and maximum tolerated dose (MTD) for several commonly used intravenous solvents in the CD-1 mouse. 2. The acute tolerability of polyethylene glycol 400, N-methylpyrrolidone, dimethyl sulfoxide, ethanol, dimethylacetamide and propylene glycol was established, along with combinations of polyethylene glycol 400 and/or ethanol and DMSO. Based on these data, an acute NOEL and recommended MTD is reported for each solvent or solvent combination. 3. These data can guide the use of these solvents to support single-dose intravenous pharmacokinetic studies in mice. By establishing a defined dose tolerability range for the most commonly used intravenous solvents, undue pain and distress in animals can be avoided while maximizing the generation of critical pharmacokinetic data for project teams.


Asunto(s)
Relación Dosis-Respuesta a Droga , Ensayos Analíticos de Alto Rendimiento/métodos , Farmacocinética , Solventes/efectos adversos , Acetamidas/administración & dosificación , Acetamidas/efectos adversos , Administración Intravenosa , Animales , Dimetilsulfóxido/administración & dosificación , Dimetilsulfóxido/efectos adversos , Etanol/administración & dosificación , Etanol/efectos adversos , Femenino , Dosis Máxima Tolerada , Ratones , Nivel sin Efectos Adversos Observados , Polietilenglicoles/administración & dosificación , Polietilenglicoles/efectos adversos , Propilenglicol/administración & dosificación , Propilenglicol/efectos adversos , Pirrolidinonas/administración & dosificación , Pirrolidinonas/efectos adversos , Solventes/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...