• Refine Query
  • Source
  • Publication year
  • to
  • Language
  • 18
  • 9
  • 9
  • 2
  • 2
  • 2
  • 1
  • Tagged with
  • 45
  • 45
  • 12
  • 10
  • 9
  • 9
  • 9
  • 9
  • 8
  • 8
  • 7
  • 7
  • 7
  • 7
  • 6
  • About
  • The Global ETD Search service is a free service for researchers to find electronic theses and dissertations. This service is provided by the Networked Digital Library of Theses and Dissertations.
    Our metadata is collected from universities around the world. If you manage a university/consortium/country archive and want to be added, details can be found on the NDLTD website.
11

Influência do EPP-AF® na atividade da glicoproteína P e do citocromo P450 em voluntários sadios usando coquetel de marcadores / Effect of EPP-AF® on cytochrome P450 and P-glycoprotein activity in healthy subjects using the cocktail approach

Cusinato, Diego Alberto Ciscato 24 August 2017 (has links)
O EPP-AF® é um extrato padronizado de própolis quimicamente caracterizado e com eficácia e segurança pré-clínica estabelecidas. O objetivo principal deste trabalho foi realizar um ensaio clínico de segurança para avaliar a influência do EPP-AF® na atividade da P-gp e das principais isoformas CYP, através de um teste in vivo tipo coquetel de fármacos marcadores administrados em doses subterapêuticas. Foram investigados 16 voluntários adultos sadios antes e após a exposição a 375 mg de EPP-AF® por via oral durante 15 dias. As amostras seriadas de sangue foram colhidas até 12 h após a administração do coquetel contendo midazolam (0,2 mg), cafeína (10 mg), omeprazol (2 mg), metoprolol (10 mg), losartana (2 mg) e fexofenadina (10 mg). Foram desenvolvidos e validados três métodos analíticos empregando LC-MS/MS para quantificar as concentrações plasmáticas de fexofenadina, losartana, E-3174 (método 1), omeprazol, 5-OH-omeprazol, midazolam, metoprolol, ?-OHmetoprolol (método 2) e cafeína (método 3). Os métodos não apresentaram efeito matriz ou efeito residual e mostraram-se lineares para os analitos nos intervalos de 0,05-20 ng/mL (fexofenadina); 0,03 - 5 ng/mL (losartana e E31-74); 0,1 - 50 ng/mL (omeprazol), 0,3 - 50 ng/mL (5-OH-omeprazol), 0,01 - 10 ng/mL (midazolam), 0,05 - 50 ng/mL (metoprolol e ?- OH-metoprolol) e 5 - 1000 ng/mL (cafeína). Os parâmetros farmacocinéticos dos compostos foram calculados com base nas curvas de concentração plasmática versus tempo (AUC) empregando o programa Phoenix® WinNonlin®. Os valores das razões das AUC0-t e Cmax após e antes da exposição ao EPP-AF®, apresentados como média geométrica (IC90%) foram de 0,74 (0,62 - 0,89) e 0,90 (0,76 - 1,07) para fexofenadina; 0,88 (0,80 - 0,97) e 0,86 (0,76 - 0,98) para losartana; 0,96 (0,83 - 1,11) e 0,91 (0,79 - 1,04) para E-3174; 1,18 (0,91 - 1,54) e 1,21 (0,87- 1,70) para omeprazol; 1,12 (0,95 - 1,31) e 1,22 (0,95 - 1,67) para 5-OHomeprazol; 1,14 (1,03 - 1,28) e 1,21 (1,00 - 1,46) para o midazolam; 1,04 (0,92 - 1,18) e 0,94 (0,80 - 1,12) para o metoprolol; 1,05 (0,99 - 1,12) e 0,99 (0,88 - 1,12) para ?-OH-metoprolol; 0,97 (0,77 - 1,21) e 0,87 (0,69 - 1,11) para a cafeína. Quando observadas as razões metabólicas das AUC0-t E3174/losartana, 5-OH-omeprazol/omeprazol e ?-OHmetoprolol/ metoprolol encontramos, respectivamente, 1,11 (0,98 - 1,25); 0,94 (0,81 - 1,10) e 1,01 (0,88 - 1,16), indicando que, com exceção do CYP2D6, a administração de EPP-AF® nas condições estudadas apresenta potencial para inibição das isoformas CYP2C19 e CYP3A4 e indução das enzimas CYP1A2, CYP2C9 e do transportador de efluxo P-gp, embora as suas magnitudes encontram-se abaixo dos limites definidos pelos órgãos reguladores e portanto não apresentam relevância clínica / EPP-AF® is a standardized extract of propolis chemically characterized and with established pre-clinical efficacy and safety. The main objective of this work was to perform a clinical trial to evaluate the effect of EPP-AF® on P-gp and the major CYP isoforms activity, through an in vivo assay using the cocktail approach with sub-therapeutic doses. Sixteen healthy adult volunteers were investigated before and after exposure to orally administered 375 mg/day of EPP-AF® for 15 days. Serum blood samples were collected up to 12 h after the administration of midazolam (0.2 mg), caffeine (10 mg), omeprazole (2 mg), metoprolol (10 mg), losartan (2 mg) and fexofenadine (10 mg). Three analytical methods were developed and validated applying LC-MS/MS to quantify plasma concentrations of fexofenadine, losartan, E-3174 (method 1), omeprazole, 5-OH-omeprazole, midazolam, metoprolol, ?-OH-metoprolol (method 2), and caffeine (Method 3). Neither matrix effect nor carryover effect were observed. The methods were linear for the analytes in the ranges of 0.05 - 20 ng/mL (fexofenadine); 0.03 - 5 ng/ml (losartan and E-3174); 0.1 - 50 ng/mL (omeprazole), 0.3 - 50 ng/mL (5-OH-omeprazole), 0.01 - 10 ng/mL (midazolam), 0.05 - 50 ng/mL (metoprolol and ?-OH-metoprolol) and 5 - 1000 ng/mL (caffeine). The pharmacokinetic parameters of the compounds were calculated based on plasma concentration versus time (AUC) curves applying Phoenix® WinNonlin® software. AUC0-t and Cmax ratios after and before the EPPAF ® exposure, presented as geometric mean (CI 90%) were 0.74 (0.62 - 0.89) and 0.90 (0.76 - 1.07) for fexofenadine, 0.88 (0.80 - 0.97) and 0.86 (0.76 - 0.98) for losartan, 0.96 (0.83 - 1.11) and 0.91 (0.79 - 1.04) for E-3174, 1.18 (0.91 - 1.54) and 1.21 (0.87 - 1.70) for omeprazole; 1.12 (0.95 - 1.31) and 1.22 (0.95 - 1.67) for 5-OH-omeprazole, 1.14 (1.03 - 1.28) and 1.21 (1.00 - 1.46) for midazolam, 1.04 (0.92 - 1.18) and 0.94 (0.80 - 1.12) for metoprolol, 1.05 (0.99 - 1.12) and 0.99 (0.88 - 1.12) for ?-OH-metoprolol, 0.97 (0.77 - 1.21) and 0.87 (0.69 - 1.11) for caffeine. AUC0-t metabolic ratios of E3174/losartan, 5-OH-omeprazole/omeprazole and ?-OH-metoprolol/metoprolol we found to be, respectively, 1.11 (0.98 - 1.25), 0.94 (0.81 - 1.10 ) and 1.01 (0.88 - 1.16), indicating that, with the exception of CYP2D6, the administration of EPP-AF® under the conditions studied shows potential for CYP2C19 and CYP3A4 inhibition and CYP1A2, CYP2C9 and P-gp induction, although their magnitudes are below the limits defined by the regulatory agencies and therefore exhibit no clinical relevance
12

Influência do verapamil na farmacocinética e na perfusão cerebral da oxcarbazepina e dos enantiômeros do metabólito 10-hidroxicarbazepina em voluntários sadios / Influence of verapamil on the pharmacokinetics and cerebral perfusion of oxcarbazepine and the enantiomers of its metabolite 10- hydroxycarbazepine in healthy volunteers

Antunes, Natalicia de Jesus 25 November 2014 (has links)
A oxcarbazepina (OXC) é indicada como terapia adjuvante ou monoterapia no tratamento de crises epilépticas parciais ou crises tônico-clônicas generalizadas em adultos e crianças. A OXC sofre rápida eliminação pré-sistêmica com formação do metabólito ativo 10-hidroxicarbazepina (MHD), o qual possui como enantiômeros o R-(-)- e o S-(+)-MHD. A OXC e o MHD são substratos da glicoproteína-P (P-gp), que pode ser inibida pelo verapamil. O presente estudo avalia a influência do verapamil na farmacocinética e perfusão cerebral da OXC e dos enantiômeros do MHD em voluntários sadios. Os voluntários sadios (n=12) receberam em uma ocasião doses de 300 mg/12h de OXC e em outra ocasião doses de 300 mg/12h de OXC associadas com 80 mg/8h de verapamil. As amostras de sangue foram coletadas no estado de equilíbrio durante 12 horas e a avaliação da perfusão cerebral realizada utilizando a tomografia computadorizada por emissão de fóton único (SPECT) antes do início do tratamento e nos tempos 4, 6 ou 12h após a administração da OXC. As concentrações plasmáticas total e livre da OXC e dos enantiômeros do MHD foram avaliadas por LC-MS/MS. A análise farmacocinética não compartimental foi realizada com o programa WinNonlin e a farmacocinética populacional foi desenvolvida utilizando a modelagem não-linear de efeitos mistos com o programa NONMEM. Os limites de quantificação obtidos foram de 12,5 ng OXC/mL de plasma e 31,25 ng de cada enantiômero MHD/mL de plasma para a análise da concentração total, enquanto foi de 4,0 ng de OXC/mL de plasma e de 20,0 ng de cada enantiômero do MHD/mL de plasma para a determinação da concentração livre. Os coeficientes de variação obtidos nos estudos de precisão e a porcentagem de inexatidão inter e intra-ensaios foram inferiores a 15%, assegurando a reprodutibilidade e repetibilidade dos resultados. A análise farmacocinética não compartimental da OXC em monoterapia resultou nos seguintes parâmetros: concentração plasmática máxima (Cmax) de 1,35 ?g/mL como valor total e 0,32 ?g/mL como concentração livre em 1,0 h, área sob a curva concentração plasmática versus tempo (AUC0-12) de 3,98 ?g.h/mL e meia-vida de eliminação de 2,45 h, volume de distribuição aparente (Vss/F) de 352,17 L e clearance aparente (CLss/F) de 75,58 L/h. A disposição cinética do MHD é enantiosseletiva, com observação de maior proporção para o enantiômero S-(+)-MHD em relação ao R-(-)-MHD (razão AUC0-12 S-(+)/R-(-) de 4,26). A fração livre avaliada no tmax da OXC foi 0,26 para a OXC, 0,42 para o R-(-)-MHD e 0,38 para o S- (+)-MHD, mostrando enantiosseletividade na ligação às proteínas plasmáticas do MHD. O tratamento com o verapamil reduziu o tempo médio de residência (MRT) (4,71 vs 3,83 h) e Cmax como concentração livre (0,32 vs 0,53 ?g/mL) da OXC e aumentou os valores para ambos os enantiômeros do MHD de Cmax como valor total (2,60 vs 3,27 ?g/mL para o R-(-)- e 11,05 vs 11,94 ?g/mL para o S-(+)-MHD), Cmax como concentração livre (3,11 vs 4,14 ?g/mL para o S-(+)-MHD), Cmédia (2,11 vs 2,42 ?g/mL para o R-(-)- e 8,10 vs 9,07 ?g/mL para o S-(+)-MHD) e AUC0-12 (25,36 vs 29,06 ?g.h/mL para o R-(-)- e 97,19 vs 111,37 ?g.h/mL para o S-(+)-MHD). A ii farmacocinética populacional da OXC foi melhor descrita por modelo de dois compartimentos com eliminação de primeira ordem e com um conjunto de três compartimentos de trânsito para descrever o perfil de absorção da OXC. A disposição de ambos os enantiômeros do MHD foi caracterizada por modelo de um compartimento. Os valores de CLss/F estimados na monoterapia foram de 84,9 L/h para a OXC e de 2,0 L/h para ambos enantiômeros do MHD, enquanto os valores de Vss/F foram de 587 L para a OXC, 23,6 L para o R-(-)-MHD e 31,7 L para o S-(+)- MHD. Concluindo, a associação do verapamil aumentou a biodisponibilidade da OXC em 12% (farmacocinética populacional) e aumentou os valores de AUC de ambos os enantiômeros do metabólito MHD (farmacocinética não compartimental), o que está provavelmente relacionado com a inibição da P-gp no trato intestinal. A associação do verapamil aumentou as concentrações cerebrais preditas de ambos os enantiômeros do MHD em maior extensão do que aquelas observadas no plasma. As mudanças no fluxo sanguíneo cerebral (SPECTs realizados 6h após a administração da OXC) associadas à coadministração de verapamil provavelmente foram causadas pelo aumento dos níveis cerebrais de ambos os enantiômeros do MHD. A confirmação dessa observação requer um braço experimental adicional com SPECTs realizados também após a administração do verapamil em monoterapia. / Oxcarbazepine (OXC) is indicated as adjunctive therapy or monotherapy for the treatment of partial or generalized tonic-clonic seizures in adults and children. OXC undergoes rapid pre-systemic reduction with formation of the active metabolite 10- hydroxycarbazepine (MHD), which has the enantiomers R-(-)- and S-(+)-MHD. OXC and MHD are substrates of P-glycoprotein (P-gp), which can be inhibited by verapamil. The present study evaluates the influence of verapamil on the pharmacokinetics and cerebral perfusion of OXC and the MHD enantiomers in healthy volunteers. The healthy volunteers (n=12) received on one occasion doses of 300 mg/12h OXC and on another occasion they received doses of 300 mg/12h OXC associated with 80 mg/8h of verapamil. Blood samples were collected at steady state for 12 hours and the assessment of cerebral perfusion was performed using a single-photon emission computed tomography (SPECT) before the beginning of treatment and at times 4, 6 or 12 hours after OXC administration. The total and free plasma concentrations of OXC and MHD enantiomers were assessed by LC-MS/MS. The non-compartmental pharmacokinetics analysis was performed using the WinNonlin program, and population pharmacokinetics was developed using nonlinear mixed effects modelling with NONMEM.The limits of quantification obtained were 12.5 ng/mL plasma for OXC and 31.25 ng of each MHD enantiomer/mL plasma for total concentration analysis, while it was 4.0 ng OXC/mL plasma and 20.0 ng of each MHD enantiomer/mL plasma for the free concentration determination. The coefficients of variation obtained in studies of accuracy and the percentage of inaccuracy inter and intra-assay were less than 15%, ensuring the result reproducibility and repeatability. The non-compartmental pharmacokinetic analysis of OXC in monotherapy treatment, resulted in the following parameters: maximum plasma concentration (Cmax) of 1.35 ?g/mL as total concentration and 0.32 mg/mL as free concentration in 1.0 h, area under the plasma concentration vs time curve (AUC0-12) was 3.98 ?g.h/mL, half-life of 2.45 h, apparent volume of distribution (Vss/F) of 352.17 L and the apparent clearance (CLSS/F) of 75.58 L/h. The MHD kinetic disposition is enantioselective, with observation of a greater proportion of the S-(+)-MHD enantiomer compared to R-(-)-MHD (ratio AUC0-12 S-(+)/R-(-) of 4.26). The free fraction measured in the tmax of OXC was 0.26 for OXC, 0.42 for R-(-)-MHD and 0.38 for S-(+)-MHD, showing enantioselectivity in the plasma protein binding of MHD. Verapamil treatment reduced the mean residence time (MRT) (4.71 vs 3.83 h) and Cmax (0.26 vs 0.31 ?g/mL) as free concentration for OXC and increased the both MHD enantiomers values of Cmax (2.60 vs 3.27 ?g/mL for R-(-)- and 11.94 vs 11.05 ?g/mL for S-(+)-MHD) as total concentration, Cmax (3.11 vs 4,14 ?g/mL for S- (+)-MHD) as free concentration, Cavg (2.11 vs 2.42 ?g/mL for R-(-)- and 8.10 vs 9.07 ?g/mL for S-(+)-MHD) and AUC0-12 (25.36 vs 29.06 ?g.h/mL for R-(-)- and 97.19 vs 111.37 ?g.h/mL for S-(+)-MHD). The population pharmacokinetics of oxcarbazepine was best described by a two-compartment model with first-order elimination and a iv set of three transit compartments to describe the absorption profile of the parent compound. The disposition of both MHD enantiomers was characterised by onecompartment model. The CLss/F estimates in monotherapy were 84.9 L/h for OXC and 2.0 L/h for both MHD enantiomers, whereas the values of Vss/F were 587 L for OXC, 23.6 L for R-(-)-MHD and 31.7 L for S-(+)-MHD. In conclusion, verapamil coadministration increased the OXC bioavailability in 12% (population pharmacokinetics) and increased the AUC of both metabolite MHD enantiomers (non-compartmental pharmacokinetics), which is probably related to the inhibition of P-gp in the intestinal tract. Verapamil co-administration increased the predicted brain concentrations of both MHD enantiomers in a greater extent than those observed in plasma. Changes in cerebral blood flow (SPECTs performed 6h after administration of OXC) associated with co-administration of verapamil were probably caused by an increase in brain levels of both MHD enantiomers. Confirmation of this observation requires additional experimental arm with SPECTs also performed after administration of verapamil in monotherapy.
13

Influência do verapamil na farmacocinética e na perfusão cerebral da oxcarbazepina e dos enantiômeros do metabólito 10-hidroxicarbazepina em voluntários sadios / Influence of verapamil on the pharmacokinetics and cerebral perfusion of oxcarbazepine and the enantiomers of its metabolite 10- hydroxycarbazepine in healthy volunteers

Natalicia de Jesus Antunes 25 November 2014 (has links)
A oxcarbazepina (OXC) é indicada como terapia adjuvante ou monoterapia no tratamento de crises epilépticas parciais ou crises tônico-clônicas generalizadas em adultos e crianças. A OXC sofre rápida eliminação pré-sistêmica com formação do metabólito ativo 10-hidroxicarbazepina (MHD), o qual possui como enantiômeros o R-(-)- e o S-(+)-MHD. A OXC e o MHD são substratos da glicoproteína-P (P-gp), que pode ser inibida pelo verapamil. O presente estudo avalia a influência do verapamil na farmacocinética e perfusão cerebral da OXC e dos enantiômeros do MHD em voluntários sadios. Os voluntários sadios (n=12) receberam em uma ocasião doses de 300 mg/12h de OXC e em outra ocasião doses de 300 mg/12h de OXC associadas com 80 mg/8h de verapamil. As amostras de sangue foram coletadas no estado de equilíbrio durante 12 horas e a avaliação da perfusão cerebral realizada utilizando a tomografia computadorizada por emissão de fóton único (SPECT) antes do início do tratamento e nos tempos 4, 6 ou 12h após a administração da OXC. As concentrações plasmáticas total e livre da OXC e dos enantiômeros do MHD foram avaliadas por LC-MS/MS. A análise farmacocinética não compartimental foi realizada com o programa WinNonlin e a farmacocinética populacional foi desenvolvida utilizando a modelagem não-linear de efeitos mistos com o programa NONMEM. Os limites de quantificação obtidos foram de 12,5 ng OXC/mL de plasma e 31,25 ng de cada enantiômero MHD/mL de plasma para a análise da concentração total, enquanto foi de 4,0 ng de OXC/mL de plasma e de 20,0 ng de cada enantiômero do MHD/mL de plasma para a determinação da concentração livre. Os coeficientes de variação obtidos nos estudos de precisão e a porcentagem de inexatidão inter e intra-ensaios foram inferiores a 15%, assegurando a reprodutibilidade e repetibilidade dos resultados. A análise farmacocinética não compartimental da OXC em monoterapia resultou nos seguintes parâmetros: concentração plasmática máxima (Cmax) de 1,35 ?g/mL como valor total e 0,32 ?g/mL como concentração livre em 1,0 h, área sob a curva concentração plasmática versus tempo (AUC0-12) de 3,98 ?g.h/mL e meia-vida de eliminação de 2,45 h, volume de distribuição aparente (Vss/F) de 352,17 L e clearance aparente (CLss/F) de 75,58 L/h. A disposição cinética do MHD é enantiosseletiva, com observação de maior proporção para o enantiômero S-(+)-MHD em relação ao R-(-)-MHD (razão AUC0-12 S-(+)/R-(-) de 4,26). A fração livre avaliada no tmax da OXC foi 0,26 para a OXC, 0,42 para o R-(-)-MHD e 0,38 para o S- (+)-MHD, mostrando enantiosseletividade na ligação às proteínas plasmáticas do MHD. O tratamento com o verapamil reduziu o tempo médio de residência (MRT) (4,71 vs 3,83 h) e Cmax como concentração livre (0,32 vs 0,53 ?g/mL) da OXC e aumentou os valores para ambos os enantiômeros do MHD de Cmax como valor total (2,60 vs 3,27 ?g/mL para o R-(-)- e 11,05 vs 11,94 ?g/mL para o S-(+)-MHD), Cmax como concentração livre (3,11 vs 4,14 ?g/mL para o S-(+)-MHD), Cmédia (2,11 vs 2,42 ?g/mL para o R-(-)- e 8,10 vs 9,07 ?g/mL para o S-(+)-MHD) e AUC0-12 (25,36 vs 29,06 ?g.h/mL para o R-(-)- e 97,19 vs 111,37 ?g.h/mL para o S-(+)-MHD). A ii farmacocinética populacional da OXC foi melhor descrita por modelo de dois compartimentos com eliminação de primeira ordem e com um conjunto de três compartimentos de trânsito para descrever o perfil de absorção da OXC. A disposição de ambos os enantiômeros do MHD foi caracterizada por modelo de um compartimento. Os valores de CLss/F estimados na monoterapia foram de 84,9 L/h para a OXC e de 2,0 L/h para ambos enantiômeros do MHD, enquanto os valores de Vss/F foram de 587 L para a OXC, 23,6 L para o R-(-)-MHD e 31,7 L para o S-(+)- MHD. Concluindo, a associação do verapamil aumentou a biodisponibilidade da OXC em 12% (farmacocinética populacional) e aumentou os valores de AUC de ambos os enantiômeros do metabólito MHD (farmacocinética não compartimental), o que está provavelmente relacionado com a inibição da P-gp no trato intestinal. A associação do verapamil aumentou as concentrações cerebrais preditas de ambos os enantiômeros do MHD em maior extensão do que aquelas observadas no plasma. As mudanças no fluxo sanguíneo cerebral (SPECTs realizados 6h após a administração da OXC) associadas à coadministração de verapamil provavelmente foram causadas pelo aumento dos níveis cerebrais de ambos os enantiômeros do MHD. A confirmação dessa observação requer um braço experimental adicional com SPECTs realizados também após a administração do verapamil em monoterapia. / Oxcarbazepine (OXC) is indicated as adjunctive therapy or monotherapy for the treatment of partial or generalized tonic-clonic seizures in adults and children. OXC undergoes rapid pre-systemic reduction with formation of the active metabolite 10- hydroxycarbazepine (MHD), which has the enantiomers R-(-)- and S-(+)-MHD. OXC and MHD are substrates of P-glycoprotein (P-gp), which can be inhibited by verapamil. The present study evaluates the influence of verapamil on the pharmacokinetics and cerebral perfusion of OXC and the MHD enantiomers in healthy volunteers. The healthy volunteers (n=12) received on one occasion doses of 300 mg/12h OXC and on another occasion they received doses of 300 mg/12h OXC associated with 80 mg/8h of verapamil. Blood samples were collected at steady state for 12 hours and the assessment of cerebral perfusion was performed using a single-photon emission computed tomography (SPECT) before the beginning of treatment and at times 4, 6 or 12 hours after OXC administration. The total and free plasma concentrations of OXC and MHD enantiomers were assessed by LC-MS/MS. The non-compartmental pharmacokinetics analysis was performed using the WinNonlin program, and population pharmacokinetics was developed using nonlinear mixed effects modelling with NONMEM.The limits of quantification obtained were 12.5 ng/mL plasma for OXC and 31.25 ng of each MHD enantiomer/mL plasma for total concentration analysis, while it was 4.0 ng OXC/mL plasma and 20.0 ng of each MHD enantiomer/mL plasma for the free concentration determination. The coefficients of variation obtained in studies of accuracy and the percentage of inaccuracy inter and intra-assay were less than 15%, ensuring the result reproducibility and repeatability. The non-compartmental pharmacokinetic analysis of OXC in monotherapy treatment, resulted in the following parameters: maximum plasma concentration (Cmax) of 1.35 ?g/mL as total concentration and 0.32 mg/mL as free concentration in 1.0 h, area under the plasma concentration vs time curve (AUC0-12) was 3.98 ?g.h/mL, half-life of 2.45 h, apparent volume of distribution (Vss/F) of 352.17 L and the apparent clearance (CLSS/F) of 75.58 L/h. The MHD kinetic disposition is enantioselective, with observation of a greater proportion of the S-(+)-MHD enantiomer compared to R-(-)-MHD (ratio AUC0-12 S-(+)/R-(-) of 4.26). The free fraction measured in the tmax of OXC was 0.26 for OXC, 0.42 for R-(-)-MHD and 0.38 for S-(+)-MHD, showing enantioselectivity in the plasma protein binding of MHD. Verapamil treatment reduced the mean residence time (MRT) (4.71 vs 3.83 h) and Cmax (0.26 vs 0.31 ?g/mL) as free concentration for OXC and increased the both MHD enantiomers values of Cmax (2.60 vs 3.27 ?g/mL for R-(-)- and 11.94 vs 11.05 ?g/mL for S-(+)-MHD) as total concentration, Cmax (3.11 vs 4,14 ?g/mL for S- (+)-MHD) as free concentration, Cavg (2.11 vs 2.42 ?g/mL for R-(-)- and 8.10 vs 9.07 ?g/mL for S-(+)-MHD) and AUC0-12 (25.36 vs 29.06 ?g.h/mL for R-(-)- and 97.19 vs 111.37 ?g.h/mL for S-(+)-MHD). The population pharmacokinetics of oxcarbazepine was best described by a two-compartment model with first-order elimination and a iv set of three transit compartments to describe the absorption profile of the parent compound. The disposition of both MHD enantiomers was characterised by onecompartment model. The CLss/F estimates in monotherapy were 84.9 L/h for OXC and 2.0 L/h for both MHD enantiomers, whereas the values of Vss/F were 587 L for OXC, 23.6 L for R-(-)-MHD and 31.7 L for S-(+)-MHD. In conclusion, verapamil coadministration increased the OXC bioavailability in 12% (population pharmacokinetics) and increased the AUC of both metabolite MHD enantiomers (non-compartmental pharmacokinetics), which is probably related to the inhibition of P-gp in the intestinal tract. Verapamil co-administration increased the predicted brain concentrations of both MHD enantiomers in a greater extent than those observed in plasma. Changes in cerebral blood flow (SPECTs performed 6h after administration of OXC) associated with co-administration of verapamil were probably caused by an increase in brain levels of both MHD enantiomers. Confirmation of this observation requires additional experimental arm with SPECTs also performed after administration of verapamil in monotherapy.
14

Peripheral Inflammatory Pain and P-Glycoprotein in a Model of Chronic Opioid Exposure

Schaefer, Charles, Schaefer, Charles January 2017 (has links)
The rates of opioid prescription and use have continued to increase over the last few decades. In turn, a greater number of patients suffer from opioid tolerance. Treatment of acute pain is a clinical challenge for these patients. Acute pain can arise from common occurrences like surgical pain and pain resulting from the injury. P-glycoprotein (p-gp) is a transporter at the blood-brain barrier (BBB) associated with a decrease in the analgesic efficacy of morphine. Peripheral inflammatory pain (PIP) is a pain state known to cause a change in p-gp trafficking at the BBB. P-gp traffics from the nucleus to the luminal surface of endothelial cells making up the BBB. This surface where circulating blood interfaces with the endothelial cell is where p-gp will efflux morphine back into circulation. Osmotic minipumps were used as a long-term delivery method in this model of opioid tolerance in female rats. PIP induced p-gp trafficking away from nuclear stores showed a 2-fold increase when animals were exposed to opioids for 6 days. This observation presents a possible relationship between p-gp trafficking and the challenges of treating post-surgical pain in opioid tolerant patients. This could reveal potential strategies for improving pain management in these patients.
15

Transcytose à travers la barrière hémato-encéphalique : étude in vitro du transport des lipoproteines de basse densité et du peptide ß-amyloïde / Transcytosis through the blood brain barrier : in vitro studies of the transport of low density lipoprotein and ß-amyloid peptide

Candela, Pietra 03 December 2010 (has links)
La Barrière Hémato-Encéphalique (BHE) est une interface localisée au niveau des cellules endothéliales des capillaires cérébraux (CECs). Elle présente des caractéristiques structurales et métaboliques spécifiques restreignant considérablement les échanges entre le sang et le cerveau dans le but de maintenir l’homéostasie du système nerveux central (SNC). La présence de transporteurs et récepteurs au niveau de CECs permet l’apport de nutriments essentiels au fonctionnement cérébral. L’expression de ces propriétés est induite par l’environnement cérébral et notamment par la population astrocytaire. Dans notre laboratoire, la BHE est reconstituée in vitro en co-cultivant des CECs bovines et des cellules gliales primaires de rat reproduisant les principales caractéristiques de la BHE in vivo. Son utilisation a permis de mettre en évidence une voie originale de transcytose (récepteur-dépendante) assurant le transport des lipoprotéines de basse densité (LDL) vers le parenchyme cérébral. Nous avons caractérisé les premières étapes de ce processus et démontré que celles-ci impliquent des structures cellulaires spécialisées comme les cavéoles et les cavéosomes, permettant aux lipoprotéines d’être relarguées intactes vers le compartiment cérébral. Récemment, de nombreux travaux ont placé la BHE au centre des échanges des peptides β-amyloïde (Aβ) entre le sang et le cerveau. Un défaut des mécanismes de transport est suspecté d’être à l’origine de l’accumulation cérébrale de ces peptides responsables de la maladie d’Alzheimer (MA). En utilisant notre modèle in vitro de BHE, nous avons étudié l’implication des CECs dans les échanges de ces peptides. Nous avons démontré l’implication du récepteur « receptor for advanced glycation end-products » (RAGE) dans l’entrée vers le compartiment cérébral des peptides Aβ. Ce transport est spécifique et implique la voie des cavéoles. L’implication de pompes d’efflux telles que la P-glycoprotéine (P-gp) et la « breast cancer resistance protein » (BCRP) dans la restriction de l’influx des peptides Aβ a également été mise en évidence. D’autre part, des résultats préliminaires suggèrent que le récepteur « low density lipoprotein receptor-related 1 » (LRP1) n’est pas impliqué dans l’efflux des peptides Aβ. Ces résultats contribuent à apporter une meilleure compréhension du rôle de la BHE dans la MA et permettent d’envisager de nouvelles approches thérapeutiques. / The blood-brain barrier (BBB) is a dynamic interface located at the brain capillary endothelial cells (BCECs) level. This barrier possesses some morphological and enzymatic properties whose aim is to maintain homeostasis of central nervous system (CNS) by firmly reducing the passages between blood and brain. BCECs express carrier-mediated transporters and receptors allowing the income of nutrients that are essential to brain function. These BBB properties come from brain environment and especially from the astrocytic population. In our laboratory, we have developed an in vitro BBB model consisting of a co-culture of bovine BCECs and new-born rat glial cells that closely mimics the in vivo situation. In this in vitro model, an original transcytosis pathway (receptor-dependent) that ensures low density lipoproteins (LDL) transport into the brain parenchyma has been discovered. We have characterized the first steps of this transport and we have shown that involve specialized cellular structures such as caveolae and caveosomes, allowing intact lipoproteins to be released into the brain compartment. Recently, many studies have considered the BBB like a very suitable site for the exchanges of amyloid β (Aβ) peptides between blood and brain. An alteration in the mechanisms of transport is suspected to result in cerebral accumulation of these peptides which are responsible for Alzheimer's Disease (AD). Using our in vitro model, we investigated the involvement of the BCECs in the exchanges of these peptides. Our works show an asymmetrical transport across the BBB suggesting the involvement of specific receptors and transporters. We have demonstrated the involvement of « receptor for advanced glycation end products » (RAGE) in the entry into the brain compartment. This transport is specific and involves the caveolae’s pathway. The involvement of P-glycoprotein (P-gp) and « breast cancer resistance protein » (BCRP) efflux pumps in restricting the influx of Aβ peptides was also highlighted. Moreover, preliminary results suggest that « low density lipoprotein receptor related-1 » (LRP1) is not involved in the efflux of Aβ peptides. These studies help to provide a better understanding of the role of the BBB in AD and allow to consider new therapeutic approaches.
16

Synthèse et étude biologique d'analogues di- et tripeptidiques de réversines susceptibles de moduler l'activité de deux protéines de transport, la glycoprotéine R et BCRP

Koubeissi, Ali 10 December 2007 (has links) (PDF)
Cette thèse s'inscrit dans un ensemble de travaux menés au laboratoire de Chimie Thérapeutique de Lyon sur la mise au point d'analogues de di- ou tripeptides hydrophobes appelés réversines susceptibles de moduler la chimiorésistance des cellules tumorales liée à l'expression de protéines de transport appartenant à la famille « ATP-Binding Cassette » (ABC). Des analogues de ces réversines ont été synthétisés afin d'augmenter leur activité et leur biodisponibilité et de préciser les requis structuraux nécessaires à cette activité vis-à-vis de deux protéines de transport, la glycoprotéine P (P-gp) et la « Breast Cancer Resistance Protein » (BCRP). Dans un premier temps, afin d'évaluer l'influence de la liaison peptidique, nous avons synthétisé des analogues aminométhyléniques des réversines les plus actives ainsi que des dérivés cétométhyléniques d'une réversine de référence. Dans un second temps, nous avons entrepris la synthèse d'autres dérivés modifiés au niveau de la chaîne latérale du côté N-terminal d'une réversine de référence ainsi que des dérivés contraints de cette chaîne latérale afin d'explorer l'espace conformationnel dans cette région de liaison aux protéines de type ABC. Les activités biologiques des analogues synthétisés ont été évaluées sur les deux protéines de transport de type ABC, P-gp et BCRP. Les premiers résultats ont montré une sélectivité des produits vis-à-vis de la P-gp par rapport à BCRP ainsi que l'interaction de la chaîne latérale du côté N-terminal avec le site de liaison de la P-gp. Cette démarche a permis d'aboutir à des analogues plus actifs que les réversines de référence sur P-gp notamment ceux des dérivés contraints.
17

The Regulation of Multidrug Resistance Phosphoglycoprotein (MDR1/P-gp) and Breast Cancer Resistance Protein (BCRP) in the Human Placenta

Rainey, Jenna 04 May 2011 (has links)
Multidrug resistance phosphoglycoprotein (MDR1/P-gp) and breast cancer resistance protein (BCRP) were first isolated in chemoresistant cancer cells and have since been found in a variety of normal tissue, including the placenta. The potential function of MDR1/P-gp and BCRP in the human placenta is to protect the fetus from maternally circulating endogenous steroids and hormones, therapeutic drugs and toxins. The objective of this study was to examine the role of maternal steroids in the regulation of MDR1/P-gp and BCRP in the human placenta. Trophoblast cells were isolated from term placenta tissues and immunohistochemistry, western blot analysis and transport studies were used to determine the effect of maternal steroids on MDR1/P-gp and BCRP regulation. Maternal steroids, present at high concentrations in maternal serum, did not have an effect on BCRP in human syncytiotrophoblast. Estrogen and progesterone did not alter MDR1/P-gp levels in human syncytiotrophoblast, but cortisol significantly decreased MDR1/P-gp levels.
18

The Regulation of Multidrug Resistance Phosphoglycoprotein (MDR1/P-gp) and Breast Cancer Resistance Protein (BCRP) in the Human Placenta

Rainey, Jenna 04 May 2011 (has links)
Multidrug resistance phosphoglycoprotein (MDR1/P-gp) and breast cancer resistance protein (BCRP) were first isolated in chemoresistant cancer cells and have since been found in a variety of normal tissue, including the placenta. The potential function of MDR1/P-gp and BCRP in the human placenta is to protect the fetus from maternally circulating endogenous steroids and hormones, therapeutic drugs and toxins. The objective of this study was to examine the role of maternal steroids in the regulation of MDR1/P-gp and BCRP in the human placenta. Trophoblast cells were isolated from term placenta tissues and immunohistochemistry, western blot analysis and transport studies were used to determine the effect of maternal steroids on MDR1/P-gp and BCRP regulation. Maternal steroids, present at high concentrations in maternal serum, did not have an effect on BCRP in human syncytiotrophoblast. Estrogen and progesterone did not alter MDR1/P-gp levels in human syncytiotrophoblast, but cortisol significantly decreased MDR1/P-gp levels.
19

LACTONE-CARBOXYLATE INTERCONVERSION AS A DETERMINANT OF THE CLEARANCE AND ORAL BIOAVAILABILTY OF THE LIPOPHILIC CAMPTOTHECIN ANALOG AR-67

Adane, Eyob Debebe 01 January 2010 (has links)
The third generation camptothecin analog, AR-67, is undergoing early phase clinical trials as a chemotherapeutic agent. Like all camptothecins it undergoes pH dependent reversible hydrolysis between the lipophilic lactone and the hydrophilic carboxylate. The physicochemical differences between the lactone and carboxylate could potentially give rise to differences in transport across and/or entry into cells. In vitro studies indicated reduced intracellular accumulation and/or apical to basolateral transport of AR-67 lactone in P-gp and/or BCRP overexpressing MDCKII cells and increased cellular uptake of carboxylate in OATP1B1 and OATP1B3 overexpressing HeLa-pIRESneo cells. Pharmacokinetic studies were conducted in rats to study the disposition and oral bioavailability of the lactone and carboxylate and to evaluate the extent of the interaction with uptake and efflux transporters. A pharmacokinetic model accounting for interconversion in the plasma was developed and its performance evaluated through simulations and in vivo transporter inhibition studies using GF120918 and rifampin. The model predicted well the likely scenarios to be encountered clinically from pharmacogenetic differences in transporter proteins, drug-drug interactions and organ function alterations. Oral bioavailability studies showed similarity following lactone and carboxylate administration and indicated the significant role ABC transporters play in limiting the oral bioavailability.
20

The Regulation of Multidrug Resistance Phosphoglycoprotein (MDR1/P-gp) and Breast Cancer Resistance Protein (BCRP) in the Human Placenta

Rainey, Jenna 04 May 2011 (has links)
Multidrug resistance phosphoglycoprotein (MDR1/P-gp) and breast cancer resistance protein (BCRP) were first isolated in chemoresistant cancer cells and have since been found in a variety of normal tissue, including the placenta. The potential function of MDR1/P-gp and BCRP in the human placenta is to protect the fetus from maternally circulating endogenous steroids and hormones, therapeutic drugs and toxins. The objective of this study was to examine the role of maternal steroids in the regulation of MDR1/P-gp and BCRP in the human placenta. Trophoblast cells were isolated from term placenta tissues and immunohistochemistry, western blot analysis and transport studies were used to determine the effect of maternal steroids on MDR1/P-gp and BCRP regulation. Maternal steroids, present at high concentrations in maternal serum, did not have an effect on BCRP in human syncytiotrophoblast. Estrogen and progesterone did not alter MDR1/P-gp levels in human syncytiotrophoblast, but cortisol significantly decreased MDR1/P-gp levels.

Page generated in 0.0349 seconds