Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Pharmacol ; 64(1): 94-102, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37566903

RESUMO

Lasmiditan is an in vitro inhibitor of P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) efflux transporters. We aimed to confirm predictions from physiologically based pharmacokinetic models of lasmiditan, and assess the safety and tolerability of rosuvastatin and dabigatran co-administered with lasmiditan. In this open-label, post-marketing drug-drug interaction, phase 1 clinical trial, eligible participants were adults aged 21-70 years with a body mass index of 18.5-35.0 kg/m2 . Part 1 (P-gp, 150 mg dabigatran etexilate with 200 mg lasmiditan) and part 2 (BCRP, 10 mg rosuvastatin with 200 mg lasmiditan) employed similar designs: a single dose of probe substrate administered on day -2 with pharmacokinetic evaluation; 1-week washout; lasmiditan administered on days 8 and 9 alone; lasmiditan co-administered with a single dose of probe substrate on day 10, with pharmacokinetic evaluation of probe substrate and lasmiditan. Sixty-six participants were included in part 1 and 30 participants were included in part 2. Following dabigatran co-administration with lasmiditan, versus dabigatran alone, 90% confidence intervals for geometric least-squares (LS) mean ratios of area under the plasma concentration-time curve from time 0 extrapolated to infinity (AUC0-∞ ) and maximum observed drug concentration (Cmax ) were not contained within the non-effect boundaries (0.80 to 1.25). Dabigatran AUC0-∞ increased by 25% and Cmax increased by 22%. The median time of maximum observed drug concentration (tmax ) for dabigatran was 2.0 to 3.0 hours. Following rosuvastatin co-administration with lasmiditan, versus rosuvastatin alone, 90%CIs for geometric LS mean ratios of AUC0-∞ and Cmax were contained within non-effect boundaries (0.80-1.25). Rosuvastatin AUC0-∞ increased by 15% and Cmax increased by 7%. The median tmax for rosuvastatin was 4.0 hours. Results suggest that lasmiditan has a weak effect on P-gp substrates and no clinically relevant effect on BCRP substrates.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP , Neoplasias da Mama , Adulto , Feminino , Humanos , Área Sob a Curva , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Estudos Cross-Over , Dabigatrana , Interações Medicamentosas , Proteínas de Neoplasias , Rosuvastatina Cálcica/farmacocinética , Adulto Jovem , Pessoa de Meia-Idade , Idoso
2.
3.
Pharm Res ; 40(11): 2499-2511, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36635486

RESUMO

PURPOSE: The objective of this work was to demonstrate that clinical OAT1-mediated DDIs can be predicted using physiologically based pharmacokinetic (PBPK) modeling. METHODS: LY404039 is a metabotropic glutamate receptor 2/3 agonist and the active moiety of the prodrug pomaglumetad methionil (LY2140023). After oral administration, pomaglumetad methionil is rapidly taken up by enterocytes via PEPT1 and once absorbed, converted to LY404039 via membrane dehydropeptidase 1 (DPEP1). LY404039 is renally excreted by both glomerular filtration and active secretion and in vitro studies showed that the active secretion of LY404039 was mediated by the organic anion transporter 1 (OAT1). Both clinical and in vitro data were used to build a PBPK model to predict OAT1-mediated DDIs. RESULTS: In vitro inhibitory potencies (IC50) of the known OAT inhibitors, probenecid and ibuprofen, were determined to be 4.00 and 2.63 µM, respectively. Subsequently, clinical drug-drug interaction (DDI) study showed probenecid reduced the renal clearance of LY404039 by 30 to 40%. The PBPK bottom-up model, predicted a renal clearance that was approximately 20% lower than the observed one. The middle-out model, using an OAT1 relative activity factor (RAF) of 3, accurately reproduced the renal clearance of LY404039 and pharmacokinetic (PK) changes of LY404039 in the presence of probenecid. CONCLUSIONS: OAT1- mediated DDIs can be predicted using in vitro measured IC50 and PBPK modeling. The effect of ibuprofen was predicted to be minimal (AUC ratio of 1.15) and not clinically relevant.


Assuntos
Aminoácidos , Compostos Bicíclicos Heterocíclicos com Pontes , Óxidos S-Cíclicos , Interações Medicamentosas , Aminoácidos/metabolismo , Óxidos S-Cíclicos/sangue , Óxidos S-Cíclicos/farmacocinética , Compostos Bicíclicos Heterocíclicos com Pontes/sangue , Compostos Bicíclicos Heterocíclicos com Pontes/farmacocinética , Modelos Biológicos , Pró-Fármacos/metabolismo , Pró-Fármacos/farmacocinética , Humanos , Masculino , Feminino , Adulto , Pessoa de Meia-Idade
4.
CPT Pharmacometrics Syst Pharmacol ; 9(9): 523-533, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32683787

RESUMO

Abemaciclib is an oral anticancer drug that inhibits cyclin dependent kinases 4 and 6 and is metabolized by cytochrome P450 3A in the intestines and liver to active metabolites. The objectives were (1) to develop a mechanistic model to characterize the pharmacokinetics (PK) of the active moieties and investigate the effect of patient factors and (2) apply the model to dat from two phase III breast cancer trials of abemaciclib in combination with endocrine therapy. To develop the model, data from seven phase I studies and two phase II studies including 421 patients with cancer and 65 healthy individuals were pooled for nonlinear mixed effects modeling. The PK was similar between patients and healthy subjects, and the effects of diarrhea, formulation, race, and patient covariates on exposure were negligible. Application of the model confirmed its predictive performance and that abemaciclib PK did not change when coadministered with endocrine therapy.


Assuntos
Aminopiridinas/farmacocinética , Benzimidazóis/farmacocinética , Neoplasias da Mama/tratamento farmacológico , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacocinética , Adulto , Idoso , Idoso de 80 Anos ou mais , Aminopiridinas/administração & dosagem , Aminopiridinas/uso terapêutico , Antineoplásicos Hormonais/administração & dosagem , Antineoplásicos Hormonais/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica , Benzimidazóis/administração & dosagem , Benzimidazóis/uso terapêutico , Estudos de Casos e Controles , Citocromo P-450 CYP3A/metabolismo , Relação Dose-Resposta a Droga , Composição de Medicamentos/métodos , Desenvolvimento de Medicamentos/métodos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Dinâmica não Linear , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/uso terapêutico
5.
J Clin Pharmacol ; 60(7): 915-930, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32080863

RESUMO

Abemaciclib, a selective inhibitor of cyclin-dependent kinases 4 and 6, is metabolized mainly by cytochrome P450 (CYP)3A4. Clinical studies were performed to assess the impact of strong inhibitor (clarithromycin) and inducer (rifampin) on the exposure of abemaciclib and active metabolites. A physiologically based pharmacokinetic (PBPK) model incorporating the metabolites was developed to predict the effect of other strong and moderate CYP3A4 inhibitors and inducers. Clarithromycin increased the area under the plasma concentration-time curve (AUC) of abemaciclib and potency-adjusted unbound active species 3.4-fold and 2.5-fold, respectively. Rifampin decreased corresponding exposures 95% and 77%, respectively. These changes influenced the fraction metabolized via CYP3A4 in the model. An absolute bioavailability study informed the hepatic and gastric availability. In vitro data and a human radiolabel study determined the fraction and rate of formation of the active metabolites as well as absorption-related parameters. The predicted AUC ratios of potency-adjusted unbound active species with rifampin and clarithromycin were within 0.7- and 1.25-fold of those observed. The PBPK model predicted 3.78- and 7.15-fold increases in the AUC of the potency-adjusted unbound active species with strong CYP3A4 inhibitors itraconazole and ketoconazole, respectively; and 1.62- and 2.37-fold increases with the concomitant use of moderate CYP3A4 inhibitors verapamil and diltiazem, respectively. The model predicted modafinil, bosentan, and efavirenz would decrease the AUC of the potency-adjusted unbound active species by 29%, 42%, and 52%, respectively. The current PBPK model, which considers changes in unbound potency-adjusted active species, can be used to inform dosing recommendations when abemaciclib is coadministered with CYP3A4 perpetrators.


Assuntos
Aminopiridinas/metabolismo , Aminopiridinas/farmacocinética , Benzimidazóis/metabolismo , Benzimidazóis/farmacocinética , Quinases Ciclina-Dependentes/metabolismo , Quinases Ciclina-Dependentes/farmacocinética , Indutores do Citocromo P-450 CYP3A/farmacocinética , Inibidores do Citocromo P-450 CYP3A/farmacocinética , Administração Oral , Adulto , Idoso , Alcinos/farmacocinética , Aminopiridinas/administração & dosagem , Aminopiridinas/sangue , Área Sob a Curva , Benzimidazóis/administração & dosagem , Benzimidazóis/sangue , Benzoxazinas/farmacocinética , Bosentana/farmacocinética , Claritromicina/administração & dosagem , Claritromicina/farmacocinética , Simulação por Computador , Quinases Ciclina-Dependentes/administração & dosagem , Quinases Ciclina-Dependentes/sangue , Ciclopropanos/farmacocinética , Indutores do Citocromo P-450 CYP3A/administração & dosagem , Inibidores do Citocromo P-450 CYP3A/administração & dosagem , Diltiazem/farmacocinética , Interações Medicamentosas , Feminino , Voluntários Saudáveis , Humanos , Itraconazol/farmacocinética , Cetoconazol/farmacocinética , Masculino , Pessoa de Meia-Idade , Modafinila/farmacocinética , Modelos Biológicos , Rifampina/administração & dosagem , Rifampina/farmacocinética , Verapamil/farmacocinética
6.
Clin Pharmacol Ther ; 107(5): 1082-1115, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31628859

RESUMO

Physiologically-based pharmacokinetic (PBPK) modeling has been extensively used to quantitatively translate in vitro data and evaluate temporal effects from drug-drug interactions (DDIs), arising due to reversible enzyme and transporter inhibition, irreversible time-dependent inhibition, enzyme induction, and/or suppression. PBPK modeling has now gained reasonable acceptance with the regulatory authorities for the cytochrome-P450-mediated DDIs and is routinely used. However, the application of PBPK for transporter-mediated DDIs (tDDI) in drug development is relatively uncommon. Because the predictive performance of PBPK models for tDDI is not well established, here, we represent and discuss examples of PBPK analyses included in regulatory submission (the US Food and Drug Administration (FDA), the European Medicines Agency (EMA), and the Pharmaceuticals and Medical Devices Agency (PMDA)) across various tDDIs. The goal of this collaborative effort (involving scientists representing 17 pharmaceutical companies in the Consortium and from academia) is to reflect on the use of current databases and models to address tDDIs. This challenges the common perceptions on applications of PBPK for tDDIs and further delves into the requirements to improve such PBPK predictions. This review provides a reflection on the current trends in PBPK modeling for tDDIs and provides a framework to promote continuous use, verification, and improvement in industrialization of the transporter PBPK modeling.


Assuntos
Interações Medicamentosas , Proteínas de Membrana Transportadoras/metabolismo , Modelos Biológicos , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Preparações Farmacêuticas/administração & dosagem , Preparações Farmacêuticas/metabolismo , Farmacocinética
7.
Pharm Res ; 36(12): 170, 2019 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-31654151

RESUMO

PURPOSE: Many bioactive molecules show a type of solution phase behavior, termed promiscuous aggregation, whereby at micromolar concentrations, colloidal drug-rich aggregates are formed in aqueous solution. These aggregates are known to be a major cause of false positives and false negatives in select enzymatic high-throughput screening assays. The goal of this study was to investigate the impact of drug-rich aggregates on in vitro drug screening metabolism assays. METHODS: Cilnidipine was selected as an aggregate former and its impact on drug metabolism was evaluated against rCYP2D6, rCYP1A2, rCYP2C9 and human liver microsomes. RESULTS: The cilnidipine aggregates were shown to non-specifically inhibit multiple cytochrome P450 enzymes with an IC50 comparable with the IC50 of potent model inhibitors. CONCLUSIONS: This newly demonstrated mode of "promiscuous inhibition" is of great importance as it can lead to false positives during drug metabolism evaluations and thus it needs to be considered in the future to better predict in vivo drug-drug interactions.


Assuntos
Sistema Enzimático do Citocromo P-450/química , Di-Hidropiridinas/química , Microssomos Hepáticos/metabolismo , Proteínas Recombinantes/química , Carvedilol/química , Carvedilol/metabolismo , Coloides/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Diclofenaco/química , Diclofenaco/metabolismo , Di-Hidropiridinas/metabolismo , Interações Medicamentosas , Ensaios de Triagem em Larga Escala/métodos , Humanos , Concentração Inibidora 50 , Cinética , Taxa de Depuração Metabólica/efeitos dos fármacos , Fenacetina/química , Fenacetina/metabolismo , Proteínas Recombinantes/metabolismo , Solventes/química , Tamoxifeno/química , Tamoxifeno/metabolismo
8.
CPT Pharmacometrics Syst Pharmacol ; 8(9): 664-675, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31250974

RESUMO

The drug-drug interaction profile of atorvastatin confirms that disposition is determined by cytochrome P450 (CYP) 3A4 and organic anion transporting polypeptides (OATPs). Drugs that affect gastric emptying, including dulaglutide, also affect atorvastatin pharmacokinetics (PK). Atorvastatin is a carboxylic acid that exists in equilibrium with a lactone form in vivo. The purpose of this work was to assess gastric acid-mediated lactone equilibration of atorvastatin and incorporate this into a physiologically-based PK (PBPK) model to describe atorvastatin acid, lactone, and their major metabolites. In vitro acid-to-lactone conversion was assessed in simulated gastric fluid and included in the model. The PBPK model was verified with in vivo data including CYP3A4 and OATP inhibition studies. Altering the gastric acid-lactone equilibrium reproduced the change in atorvastatin PK observed with dulaglutide. The model emphasizes the need to include gastric acid-lactone conversion and all major atorvastatin-related species for the prediction of atorvastatin PK.


Assuntos
Atorvastatina/farmacocinética , Gastroparesia/complicações , Peptídeos Semelhantes ao Glucagon/análogos & derivados , Lactonas/química , Proteínas Recombinantes de Fusão/farmacocinética , Atorvastatina/administração & dosagem , Células Cultivadas , Citocromo P-450 CYP3A , Relação Dose-Resposta a Droga , Interações Medicamentosas , Ácido Gástrico/metabolismo , Peptídeos Semelhantes ao Glucagon/administração & dosagem , Peptídeos Semelhantes ao Glucagon/farmacocinética , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Modelos Biológicos , Transportadores de Ânions Orgânicos , Proteínas Recombinantes de Fusão/administração & dosagem
9.
Int J Pharm ; 543(1-2): 29-37, 2018 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-29572154

RESUMO

Amorphous solid dispersions (ASDs) are a promising formulation strategy to increase both the apparent aqueous solubility and bioavailability of poorly water-soluble drugs. Upon dissolution under nonsink conditions, ASDs can generate highly supersaturated drug solutions which can undergo liquid-liquid phase separation (LLPS) and/or crystallization. In this study, the phase behavior of supersaturated solutions generated by antisolvent addition and upon the dissolution of ASDs was evaluated using fluorescence lifetime measurements and several other orthogonal techniques, including steady-state fluorescence spectroscopy, ultraviolet (UV) extinction and concentration profiles, ultracentrifuge measurements and nanoparticle tracking analysis. Ritonavir and lopinavir were chosen as poorly water-soluble model drugs, and the polymer, Kollidon VA64, was selected to form the dispersions. The fluorescence lifetime of the environment-sensitive fluoroprobe, PRODAN, was monitored to determine the occurrence of LLPS and crystallization. It was found that only the 10% w/w drug loading ASDs dissolved to a concentration in solution higher than the LLPS concentration and this led to an increase in the lifetime of PRODAN due to partitioning of the fluoroprobe into the drug-rich phase. In contrast, the 50% w/w drug loading ASDs did not reach the amorphous solubility, pointing to a dissolution behavior controlled by the low water solubility and high hydrophobicity of the drug. Fluorescence lifetime measurements were demonstrated to be extremely useful for the characterization of the phase behavior of supersaturated solutions of poorly water-soluble drugs.


Assuntos
Soluções Farmacêuticas/química , 2-Naftilamina/análogos & derivados , 2-Naftilamina/química , Corantes Fluorescentes/química , Inibidores da Protease de HIV/química , Lopinavir/química , Nanopartículas/química , Pirrolidinas/química , Ritonavir/química , Solubilidade , Espectrometria de Fluorescência , Compostos de Vinila/química , Água/química
10.
Clin Transl Sci ; 10(6): 509-519, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28749581

RESUMO

Baricitinib, an oral selective Janus kinase 1 and 2 inhibitor, undergoes active renal tubular secretion. Baricitinib was not predicted to inhibit hepatic and renal uptake and efflux drug transporters, based on the ratio of the unbound maximum eliminating-organ inlet concentration and the in vitro half-maximal inhibitory concentrations (IC50 ). In vitro, baricitinib was a substrate for organic anion transporter (OAT)3, multidrug and toxin extrusion protein (MATE)2-K, P-glycoprotein (P-gp), and breast cancer resistance protein (BCRP). Probenecid, a strong OAT3 inhibitor, increased the area under the concentration-time curve from time zero to infinity (AUC[0-∞] ) of baricitinib by twofold and decreased renal clearance to 69% of control in healthy subjects. Physiologically based pharmacokinetic (PBPK) modeling reproduced the renal clearance of baricitinib and the inhibitory effect of probenecid using the in vitro IC50 value of 4.4 µM. Using ibuprofen and diclofenac in vitro IC50 values of 4.4 and 3.8 µM toward OAT3, 1.2 and 1.0 AUC(0-∞) ratios of baricitinib were predicted. These predictions suggest clinically relevant drug-drug interactions (DDIs) with ibuprofen and diclofenac are unlikely.


Assuntos
Azetidinas/farmacologia , Proteínas de Membrana Transportadoras/metabolismo , Sulfonamidas/farmacologia , Adulto , Área Sob a Curva , Azetidinas/sangue , Azetidinas/farmacocinética , Interações Medicamentosas , Células HEK293 , Humanos , Masculino , Pessoa de Meia-Idade , Purinas , Pirazóis , Sulfonamidas/sangue , Sulfonamidas/farmacocinética , Fatores de Tempo , Adulto Jovem
11.
Drug Metab Dispos ; 43(3): 325-34, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25504564

RESUMO

Pemetrexed, an anionic anticancer drug with a narrow therapeutic index, is eliminated mainly by active renal tubular secretion. The in vitro to in vivo extrapolation approach used in this work was developed to predict possible drug-drug interactions (DDIs) that may occur after coadministration of pemetrexed and nonsteroidal anti-inflammatory drugs (NSAIDs), and it included in vitro assays, risk assessment models, and physiologically based pharmacokinetic (PBPK) models. The pemetrexed transport and its inhibition parameters by several NSAIDs were quantified using HEK-PEAK cells expressing organic anion transporter (OAT) 3 or OAT4. The NSAIDs were ranked according to their DDI index, calculated as the ratio of their maximum unbound concentration in plasma over the concentration inhibiting 50% (IC50) of active pemetrexed transport. A PBPK model for ibuprofen, the NSAID with the highest DDI index, was built incorporating active renal secretion in Simcyp Simulator. The bottom-up model for pemetrexed underpredicted the clearance by 2-fold. The model we built using a scaling factor of 5.3 for the maximal uptake rate (Vmax) of OAT3, which estimated using plasma concentration profiles from patients given a 10-minute infusion of 500 mg/m(2) of pemetrexed supplemented with folic acid and vitamin B12, recovered the clinical data adequately. The observed/predicted increases in Cmax and the area under the plasma-concentration time curve (AUC0-inf) of pemetrexed when ibuprofen was coadministered were 1.1 and 1.0, respectively. The coadministration of all other NSAIDs was predicted to have no significant impact on the AUC0-inf based on their DDI indexes. The PBPK model reasonably reproduced pemetrexed concentration time profiles in cancer patients and its interaction with ibuprofen.


Assuntos
Transporte Biológico/fisiologia , Interações Medicamentosas/fisiologia , Glutamatos/metabolismo , Glutamatos/farmacocinética , Guanina/análogos & derivados , Rim/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Anti-Inflamatórios não Esteroides/metabolismo , Área Sob a Curva , Linhagem Celular Tumoral , Feminino , Guanina/metabolismo , Guanina/farmacocinética , Células HeLa , Humanos , Ibuprofeno/metabolismo , Ibuprofeno/farmacocinética , Masculino , Proteínas de Membrana Transportadoras/metabolismo , Pessoa de Meia-Idade , Modelos Biológicos , Pemetrexede
12.
Pharm Res ; 30(11): 2931-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23959853

RESUMO

PURPOSE: To determine the effect of PepT1 on the absorption and disposition of cefadroxil, including the potential for saturable intestinal uptake, after escalating oral doses of drug. METHODS: The absorption and disposition kinetics of [3H]cefadroxil were determined in wild-type and PepT1 knockout mice after 44.5, 89.1, 178, and 356 nmol/g oral doses of drug. The pharmacokinetics of [3H]cefadroxil were also determined in both genotypes after 44.5 nmol/g intravenous bolus doses. RESULTS: PepT1 deletion reduced the area under the plasma concentration-time profile (AUC0-120) of cefadroxil by 10-fold, the maximum plasma concentration (Cmax) by 17.5-fold, and increased the time to reach a maximum plasma concentration (Tmax) by 3-fold. There was no evidence of nonlinear intestinal absorption since AUC0-120 and Cmax values changed in a dose-proportional manner. Moreover, the pharmacokinetics of cefadroxil were not different between genotypes after intravenous bolus doses, indicating that PepT1 did not affect drug disposition. Finally, no differences were observed in the peripheral tissue distribution of cefadroxil (i.e., outside gastrointestinal tract) once these tissues were corrected for differences in perfusing blood concentrations. CONCLUSIONS: The findings demonstrate convincingly the critical role of intestinal PepT1 in both the rate and extent of oral administration for cefadroxil and potentially other aminocephalosporin drugs.


Assuntos
Antibacterianos/administração & dosagem , Antibacterianos/farmacologia , Cefadroxila/administração & dosagem , Cefadroxila/farmacocinética , Simportadores/genética , Administração Oral , Animais , Antibacterianos/sangue , Cefadroxila/sangue , Deleção de Genes , Absorção Intestinal , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Knockout , Transportador 1 de Peptídeos , Simportadores/metabolismo
13.
Pharm Res ; 30(4): 1017-25, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23224978

RESUMO

PURPOSE: To determine the contribution of intestinal PepT1 on the permeability and oral absorption of the ß-lactam antibiotic drug cefadroxil. METHODS: The effective permeability (P eff ) of cefadroxil was evaluated in wild-type and PepT1 knockout mice following in situ single-pass intestinal perfusions. The plasma concentration-time profiles of cefadroxil were also examined after oral gavage. RESULTS: The P eff (cm/s) of cefadroxil in wild-type mice was 0.49 × 10(-4) in duodenum, 0.80 × 10(-4) in jejunum, 0.88 × 10(-4) in ileum and 0.064 × 10(-4) in colon. The P eff (cm/s) in PepT1 knockout mice was significantly reduced in small intestine, but not in colon, as shown by values of 0.003 × 10(-4), 0.090 × 10(-4), 0.042 × 10(-4) and 0.032 × 10(-4), respectively. Jejunal uptake of cefadroxil was saturable (Km = 2-4 mM) and significantly attenuated by the sodium-proton exchange inhibitor 5-(N,N-dimethyl)amiloride. Jejunal permeability of cefadroxil was not affected by L-histidine, glycine, cephalothin, p-aminohippurate or N-methylnicotinamide. In contrast, cefadroxil permeability was significantly reduced by glycylproline, glycylsarcosine, or cephalexin. Finally, PepT1 ablation resulted in 23-fold reductions in peak plasma concentrations and 14-fold reductions in systemic exposure of cefadroxil after oral dosing. CONCLUSIONS: The findings are definitive in demonstrating that PepT1 is the major transporter responsible for the small intestinal permeability of cefadroxil as well as its enhanced oral drug performance.


Assuntos
Antibacterianos/farmacocinética , Cefadroxila/farmacocinética , Técnicas de Inativação de Genes , Mucosa Intestinal/metabolismo , Simportadores/genética , Administração Oral , Animais , Antibacterianos/metabolismo , Cefadroxila/metabolismo , Feminino , Masculino , Camundongos , Camundongos Knockout , Transportador 1 de Peptídeos , Permeabilidade , Simportadores/metabolismo
14.
Pharm Res ; 24(11): 2156-67, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17668300

RESUMO

PURPOSE: Following extracellular drug clearance, we analyzed the rate of doxorubicin efflux from the nucleus of three human leukemic cells (K562, Molt4 and CCRF-CEM) and related it to their differential sensitivity to this drug, after a short drug pulse. RESULTS: For many pulse-chase regimes, K562 cell viability was least affected by doxorubicin. In K562 cells, nuclear drug accumulation was greatest, but nuclear drug egress was also greatest. P-glycoprotein over-expression in a doxorubicin-resistant, K562/DOX sub-line did not facilitate doxorubicin efflux from the nucleus. In K562 cells, doxorubicin accumulated in multivesicular bodies (MVBs) through a pH-dependent mechanism. Inhibiting drug sequestration in MVBs did not affect nuclear efflux. The rates of doxorubicin efflux from the nuclei of live and digitonin-permeabilized K562 cells were similar. However, extracting cytoplasmic membranes with Triton X-100 significantly inhibited nuclear drug efflux following extracellular drug clearance. CONCLUSION: Our results are consistent with drug efflux from the nucleus being primarily mediated by an ATP-independent, passive diffusion mechanism. The effect of membrane extraction suggests that nonspecific drug absorption to cytoplasmic membranes plays a role in facilitating nuclear efflux in K562 cells, perhaps by lowering the concentration of free doxorubicin from a perinuclear diffusion boundary layer.


Assuntos
Antibióticos Antineoplásicos/metabolismo , Núcleo Celular/metabolismo , Doxorrubicina/metabolismo , Transporte Ativo do Núcleo Celular , Trifosfato de Adenosina/fisiologia , Cloreto de Amônio/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citoplasma/metabolismo , Resistencia a Medicamentos Antineoplásicos , Endocitose , Humanos , Células K562 , Fusão de Membrana , Taxa de Depuração Metabólica
15.
Artigo em Inglês | MEDLINE | ID: mdl-23482717

RESUMO

High-throughput microscopic screening instruments can generate huge collections of images of live cells incubated with combinatorial libraries of fluorescent molecules. Organizing and visualizing these images to discern biologically important patterns that link back to chemical structure is a challenge. We present an analysis and visualization methodology - Cheminformatic Assisted Image Array (CAIA) - that greatly facilitates data mining efforts. For illustration, we considered a collection of microscopic images acquired from cells incubated with each member of a combinatorial library of styryl molecules being screened for candidate bioimaging probes. By sorting CAIAs based on quantitative image features, the relative contribution of each combinatorial building block on probe intracellular distribution could be visually discerned. The results revealed trends hidden in the dataset: most interestingly, the building blocks of the styryl molecules appeared to behave as chemical address tags, additively and independently encoding spatial patterns of intracellular fluorescence. Translated into practice, CAIA facilitated discovery of several outstanding styryl molecules for live cell nuclear imaging applications.

16.
Pharm Res ; 23(8): 1687-95, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16841193

RESUMO

PURPOSE: This study investigates the subcellular pharmacokinetics of drug efflux in cancer cells and explores the role of the multivesicular body (MVB) in facilitating efflux of doxorubicin, a widely used DNA-targeting anticancer agent, from the nucleus. METHODS: Human erythroleukemic K562 cells were pulsed with doxorubicin and then chased in drug-free media to allow for efflux. Microscopy and biochemical techniques were used to visualize the subcellular localization of the drug and measure drug content and distribution during the efflux period. To explore the role of the MVB in doxorubicin efflux, K562 cells were transfected with dominant negative mutant forms of VPS4a-GFP chimeras. RESULTS: Although the intracellular concentration of drug exceeds the extracellular concentration, nuclear efflux of doxorubicin occurs in living cells at a faster rate than doxorubicin unbinding from isolated nuclei into drug-free buffer. In cells expressing dominant negative VPS4a, doxorubicin accumulates in VPS4a-positive vesicles and drug sequestration is inhibited, directly implicating the MVB pathway in the egress route of doxorubicin in this cell type. CONCLUSIONS: Cellular membranes are a component of the doxorubicin efflux mechanism in K562 cells. Dominant-negative GFP chimeric mutants can be used to elucidate the role of specific membrane trafficking pathways in subcellular drug transport routes.


Assuntos
Adenosina Trifosfatases/fisiologia , Antineoplásicos/farmacocinética , DNA/metabolismo , Proteínas Repressoras/fisiologia , Transdução de Sinais/fisiologia , ATPases Associadas a Diversas Atividades Celulares , Antibióticos Antineoplásicos/farmacocinética , Antineoplásicos/metabolismo , Núcleo Celular , Citoplasma/metabolismo , Doxorrubicina/farmacocinética , Complexos Endossomais de Distribuição Requeridos para Transporte , Humanos , Células K562 , Plasmídeos/genética , Frações Subcelulares/metabolismo , Transfecção , Vesículas Transportadoras/metabolismo , ATPases Vacuolares Próton-Translocadoras , Proteínas de Transporte Vesicular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...