Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Eur J Pharm Sci ; 179: 106309, 2022 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-36265816

RESUMO

This work aimed to develop a physiologically based pharmacokinetic (PBPK) model for raltegravir accounting for UDP-glucuronosyltransferase (UGT) metabolism to assess the effect of UGT gene polymorphisms. Raltegravir elimination was evaluated using Km and Vmax values from human recombinant systems and UGT tissue scalar considering liver, kidney, and intestine. The predicted/observed ratios for raltegravir PK parameters were within a 2-fold error range in UGT1A1 poor and normal metabolizers, except in Asian UGT1A1 poor metabolizers. This PBPK modeling approach suggests that UGT1A3 is the main contributor to raltegravir's metabolism. UGT1A3 and UGT1A1 gene polymorphisms might have an additive effect on raltegravir's drug disposition and response. The final model accounting for hepatic, renal, and intestinal UGT metabolism, biliary clearance, and renal excretion improved model predictions compared with the previously published models. This PBPK model with the quantitative characterization of raltegravir elimination pathways can support dose adjustments in different clinical scenarios.


Assuntos
Glucuronosiltransferase , Microssomos Hepáticos , Humanos , Raltegravir Potássico/metabolismo , Microssomos Hepáticos/metabolismo , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Cinética , Isoformas de Proteínas/metabolismo
2.
Life Sci ; 308: 120948, 2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-36096241

RESUMO

AIMS: To assess the potential direct effects of the integrase strand-transfer inhibitors (INsTIs) dolutegravir, bictegravir, and raltegravir, drugs used as treatment for people living with human immunodeficiency virus (PLWH), on human adipose cells. MAIN METHODS: Drugs were added to the differentiation medium of human Simpson-Golabi-Behmel syndrome (SGBS) adipose cells and morphological adipogenesis was monitored for 10 days. Also, adipocytes were exposed to drugs following differentiation (day 14). The gene expression levels of selected adipogenesis markers, adipocyte metabolism markers, adipokines, and cytokines were determined by quantitative-reverse transcription polymerase-chain reaction. The release of adiponectin and leptin into the culture medium was measured using specific enzyme-linked immunosorbent assay, and release of interleukin-6 and chemokine (CC motif) ligand-2 using Multiplex assays. KEY FINDINGS: Overall morphological adipogenesis was unaltered by INsTIs. The expression of adipogenesis marker genes (peroxisome proliferator-activated receptor-Ɣ and lipoprotein lipase) was slightly reduced in dolutegravir-treated differentiating adipocytes. Bictegravir repressed gene expression and the release of pro-inflammatory cytokines in differentiating adipocytes. Dolutegravir and raltegravir increased interleukin-6 gene expression, but only dolutegravir increased interleukin-6 release. Dolutegravir repressed adiponectin expression and release in differentiating adipocytes and had a similar but milder effect on leptin. Drug treatment of mature adipocytes reduced adiponectin gene expression in response to dolutegravir. SIGNIFICANCE: The INsTIs studied do not have a significant effect on human adipose cell differentiation but exert distinct effects on gene expression and secretion of adipokines and cytokines. These findings will help understand and manage the effects of INsTI-containing treatments on body weight and metabolic dysregulation in PLWH.


Assuntos
Adipocinas , Leptina , Adipócitos/metabolismo , Adipocinas/metabolismo , Adiponectina/metabolismo , Amidas , Citocinas/metabolismo , Compostos Heterocíclicos com 3 Anéis , Humanos , Inflamação/metabolismo , Integrases/metabolismo , Integrases/farmacologia , Interleucina-6/metabolismo , Leptina/metabolismo , Ligantes , Lipase Lipoproteica , Oxazinas , Receptores Ativados por Proliferador de Peroxissomo , Piperazinas , Piridonas , Raltegravir Potássico/metabolismo , Raltegravir Potássico/farmacologia
3.
Viruses ; 12(9)2020 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-32858802

RESUMO

The process of viral integration into the host genome is an essential step of the HIV-1 life cycle. The viral integrase (IN) enzyme catalyzes integration. IN is an ideal therapeutic enzyme targeted by several drugs; raltegravir (RAL), elvitegravir (EVG), dolutegravir (DTG), and bictegravir (BIC) having been approved by the USA Food and Drug Administration (FDA). Due to high HIV-1 diversity, it is not well understood how specific naturally occurring polymorphisms (NOPs) in IN may affect the structure/function and binding affinity of integrase strand transfer inhibitors (INSTIs). We applied computational methods of molecular modelling and docking to analyze the effect of NOPs on the full-length IN structure and INSTI binding. We identified 13 NOPs within the Cameroonian-derived CRF02_AG IN sequences and further identified 17 NOPs within HIV-1C South African sequences. The NOPs in the IN structures did not show any differences in INSTI binding affinity. However, linear regression analysis revealed a positive correlation between the Ki and EC50 values for DTG and BIC as strong inhibitors of HIV-1 IN subtypes. All INSTIs are clinically effective against diverse HIV-1 strains from INSTI treatment-naïve populations. This study supports the use of second-generation INSTIs such as DTG and BIC as part of first-line combination antiretroviral therapy (cART) regimens, due to a stronger genetic barrier to the emergence of drug resistance.


Assuntos
Inibidores de Integrase de HIV/metabolismo , Integrase de HIV/genética , Integrase de HIV/metabolismo , HIV-1/genética , Amidas , Sítios de Ligação , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , Integrase de HIV/química , Inibidores de Integrase de HIV/farmacologia , HIV-1/efeitos dos fármacos , HIV-1/enzimologia , Compostos Heterocíclicos com 3 Anéis/metabolismo , Compostos Heterocíclicos com 3 Anéis/farmacologia , Compostos Heterocíclicos de 4 ou mais Anéis/metabolismo , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Modelos Lineares , Modelos Moleculares , Simulação de Acoplamento Molecular , Oxazinas/metabolismo , Oxazinas/farmacologia , Piperazinas/metabolismo , Piperazinas/farmacologia , Piridonas/metabolismo , Piridonas/farmacologia , Quinolonas/metabolismo , Quinolonas/farmacologia , Raltegravir Potássico/metabolismo , Raltegravir Potássico/farmacologia , Alinhamento de Sequência
4.
J Pharm Pharmacol ; 72(10): 1361-1369, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32657432

RESUMO

OBJECTIVES: Raltegravir (RAL) that can form chelates with multivalent metal cations shows lateral interactions with multivalent metal cation and polycationic polymer. We investigated the interactions of RAL with multivalent metal cation preparations, Al(OH)3 and LaCO3 , and polycationic polymer preparations, bixalomer (Bxl) and sevelamer (Svl). METHODS: Immediately before the oral administration of 40 mg/kg RAL, the rats were administered orally with the vehicle, Al(OH)3 , LaCO3 , Bxl, or Svl, and the time course of RAL serum concentration was followed. The in vitro binding affinity of RAL with multivalent metal cation and polycationic polymer was also evaluated using isothermal titration calorimetry (ITC). RESULTS: When Al(OH)3 , LaCO3 , Bxl, or Svl was concomitantly administered with RAL, the maximum concentration and area under the curve were significantly lower than those when RAL was administered alone. ITC showed the interaction of RAL with Al(OH)3 as an enthalpy-driven reaction and its interactions with LaCO3 and Bxl as entropy-enthalpy mixed reactions. CONCLUSIONS: The interaction of RAL with Al(OH)3 , LaCO3, Bxl, or Svl can inhibit RAL absorption into the gastrointestinal tract, and thus, the multivalent metal cation and polycationic polymer are the modifying factors that can affect RAL pharmacokinetics.


Assuntos
Quelantes/administração & dosagem , Absorção Gastrointestinal/fisiologia , Polímeros/administração & dosagem , Raltegravir Potássico/antagonistas & inibidores , Raltegravir Potássico/metabolismo , Administração Oral , Animais , Fármacos Anti-HIV/metabolismo , Cátions , Quelantes/farmacocinética , Combinação de Medicamentos , Absorção Gastrointestinal/efeitos dos fármacos , Masculino , Polímeros/farmacocinética , Ratos , Ratos Wistar
5.
PLoS One ; 15(5): e0223464, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32379830

RESUMO

Resistance associated mutations (RAMs) threaten the long-term success of combination antiretroviral therapy (cART) outcomes for HIV-1 treatment. HIV-1 Integrase (IN) strand transfer inhibitors (INSTIs) have proven to be a viable option for highly specific HIV-1 therapy. The INSTI, Dolutegravir is recommended by the World Health Organization for use as first-line cART. This study aims to understand how RAMs affect the stability of IN, as well as the binding of the drug Dolutegravir to the catalytic pocket of the protein. A homology model of HIV-1 subtype C IN was successfully constructed and validated. The site directed mutator webserver was used to predict destabilizing and/or stabilizing effects of known RAMs while FoldX confirmed any changes in protein energy upon introduction of mutation. Also, interaction analysis was performed between neighbouring residues. Three mutations known to be associated with Raltegravir, Elvitegravir and Dolutegravir resistance were selected; E92Q, G140S and Y143R, for molecular dynamics simulations. The structural quality assessment indicated high reliability of the HIV-1C IN tetrameric structure, with more than 90% confidence in modelled regions. Change in free energy for the three mutants indicated different effects, while simulation analysis showed G140S to have the largest affect on protein stability and flexibility. This was further supported by weaker non-bonded pairwise interaction energy and binding free energy values between the drug DTG and E92Q, Y143R and G140S mutants suggesting reduced binding affinity, as indicated by interaction analysis in comparison to the WT. Our findings suggest the G140S mutant has the strongest effect on the HIV-1C IN protein structure and Dolutegravir binding. To the best of our knowledge, this is the first study that uses the consensus wild type HIV-1C IN sequence to build an accurate 3D model to understand the effect of three known mutations on DTG drug binding in a South Africa context.


Assuntos
Farmacorresistência Viral/genética , Infecções por HIV/metabolismo , Inibidores de Integrase de HIV/metabolismo , Integrase de HIV/genética , Integrase de HIV/metabolismo , HIV-1/enzimologia , Compostos Heterocíclicos com 3 Anéis/metabolismo , Mutação , Sequência de Aminoácidos , Domínio Catalítico/genética , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , Integrase de HIV/química , Inibidores de Integrase de HIV/uso terapêutico , Compostos Heterocíclicos com 3 Anéis/uso terapêutico , Humanos , Simulação de Dinâmica Molecular , Oxazinas , Piperazinas , Ligação Proteica/genética , Estabilidade Proteica , Piridonas , Quinolonas/metabolismo , Quinolonas/uso terapêutico , Raltegravir Potássico/metabolismo , Raltegravir Potássico/uso terapêutico , África do Sul , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
6.
PLoS One ; 14(5): e0216712, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31120908

RESUMO

CONTEXT: Classical antiretroviral agents may acutely impact on metabolic, mitochondrial, renal and hepatic function in HIV-infected and uninfected persons. Fusion and integrase inhibitors are supposed to be safer, but have been scarcely investigated. To avoid any interference with HIV or other antiretrovirals, we assessed markers of these toxicities in healthy adult volunteers treated with Enfuvirtide (T20) or Raltegravir (RAL). METHODS: Twenty-six healthy participants were randomized to T20/90mg vs. placebo (n = 12) or RAL/400mg vs. placebo (n = 14) every 12h in two 7-day periods separated by a 4-week washout period. Major end-points were changes in lipid profile (total cholesterol, high-density-lipoprotein (HDL)-cholesterol, low-density-lipoprotein (LDL)-cholesterol, triglycerides), insulin resistance (glucose) and mitochondrial toxicity (mitochondrial DNA content-mtDNA-in peripheral blood mononuclear cells). Renal and hepatic toxicity (creatinine, alanine transaminase (AST), alanine aminotransferase (ALT), bilirubin and total plasma proteins) and overall safety were also analysed. Effect of period, treatment, and basal measures were evaluated for each end-point. RESULTS: Neither T20-administration nor RAL-administration yielded to any statistic significant change in the markers of metabolic, mitochondrial, renal or hepatic toxicity assessed. No symptoms indicative of drug toxicity were neither found in any subject. CONCLUSIONS: In absence of HIV infection, or concomitant treatment, short-term exposure to T20 or RAL in healthy adult volunteers did not lead to any indicative changes in toxicity markers thus presuming the safe profile of both drugs.


Assuntos
Enfuvirtida/farmacologia , Raltegravir Potássico/farmacologia , Adulto , Alanina Transaminase/análise , Alanina Transaminase/sangue , Antirretrovirais/uso terapêutico , Creatina/análise , Creatina/sangue , Enfuvirtida/metabolismo , Enfuvirtida/toxicidade , Infecções por HIV/tratamento farmacológico , Voluntários Saudáveis , Humanos , Resistência à Insulina , Rim/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Lipídeos/análise , Fígado/efeitos dos fármacos , Masculino , Metabolismo/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Raltegravir Potássico/metabolismo , Raltegravir Potássico/toxicidade
7.
Drug Metab Dispos ; 47(5): 535-544, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30804050

RESUMO

Integrase strand transfer inhibitor (INSTI)-based regimens dominate initial human immunodeficiency virus treatment. Most INSTIs are metabolized predominantly via UDP-glucuronosyltransferases (UGTs). For drugs predominantly metabolized by UGTs, including INSTIs, in vitro data recovered from human liver microsomes (HLMs) alone often underpredict human oral clearance. While several factors may contribute, extrahepatic glucuronidation may contribute to this underprediction. Thus, we comprehensively characterized the kinetics for the glucuronidation of INSTIs (cabotegravir, dolutegravir, and raltegravir) using pooled human microsomal preparations from liver (HLMs), intestine (HIMs), and kidney (HKMs) tissues; human embryonic kidney 293 cells expressing individual UGTs; and recombinant UGTs. In vitro glucuronidation of cabotegravir (HLMs≈HKMs>>>HIMs), dolutegravir (HLMs>HIMs>>HKMs), and raltegravir (HLMs>HKMs>> HIMs) occurred in hepatic and extrahepatic tissues. The kinetic data from expression systems suggested the major enzymes in each tissue: hepatic UGT1A9 > UGT1A1 (dolutegravir and raltegravir) and UGT1A1 (cabotegravir), intestinal UGT1A3 > UGT1A8 > UGT1A1 (dolutegravir) and UGT1A8 > UGT1A1 (raltegravir), and renal UGT1A9 (dolutegravir and raltegravir). Enzymes catalyzing cabotegravir glucuronidation in the kidney and intestine could not be identified unequivocally. Using data from dolutegravir glucuronidation as a prototype, a "bottom-up" physiologically based pharmacokinetic model was developed in a stepwise approach and predicted dolutegravir oral clearance within 4.5-fold (hepatic data only), 2-fold (hepatic and intestinal data), and 32% (hepatic, intestinal, and renal data). These results suggest clinically meaningful glucuronidation of dolutegravir in tissues other than the liver. Incorporation of additional novel mechanistic and physiologic underpinnings of dolutegravir metabolism along with in silico approaches appears to be a powerful tool to accurately predict the clearance of dolutegravir from in vitro data.


Assuntos
Glucuronosiltransferase/metabolismo , Integrases/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular , Criança , Pré-Escolar , Feminino , Células HEK293 , Compostos Heterocíclicos com 3 Anéis/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Rim/metabolismo , Cinética , Fígado/metabolismo , Masculino , Microssomos Hepáticos/metabolismo , Pessoa de Meia-Idade , Oxazinas , Piperazinas , Piridonas/metabolismo , Raltegravir Potássico/metabolismo , Adulto Jovem
8.
PLoS One ; 13(5): e0196395, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29746485

RESUMO

BACKGROUND: People with HIV are at for metabolic syndrome (MetS) and fatty liver disease, but the role of Antiretroviral therapy (ART) is poorly understood. MetS and fatty liver disease been associated with changes in adiponectin, soluble ST2 (sST2), chitinase 3-like 1 (Chi3L1), hyaluronic acid (HA), tissue inhibitor of metalloproteinase-1 (TIMP-1), lysyl oxidase-like-2 (LOXL2) and transforming growth factor ß (TGF-ß) concentrations in HIV-uninfected populations. Protease (PI) and non-nucleoside reverse transcriptase inhibitors (NNRTI) may contribute to these comorbidities, but the effects of switching from PI- or NNRTI to raltegravir (RAL) on these biomarkers is unknown. METHODS: Cryopreserved plasma was obtained from a completed, prospective trial of HIV-infected women with central adiposity on NNRTI- or PI-based ART during which they were randomized to remain on their current ART or switch to a RAL based regimen. Biomarker concentrations were quantified using ELISA and Multiplex assays at baseline and 24 weeks after randomization. Wilcoxon-signed rank test evaluated within-group changes, Spearman and linear regression models evaluated correlations between biomarkers and clinical covariates. RESULTS: Participants had a median age of 43 years, CD4+ T lymphocyte count 558 cells/mm3 and BMI 32 kg/m2; 35% met criteria for MetS. At baseline, higher adiponectin levels correlated with higher Chi3L1 levels (r = 0.42, p = 0.02), as did declines after 24 weeks (r = 0.40, p = 0.03). Changes in sST2 correlated with changes in Chi3L1 (r = 0.43, p = 0.02) and adiponectin (r = 0.40, p = 0.03). Adiponectin and Chi3L1 levels decreased significantly in women switched to RAL vs continue PI/NNRTI. CONCLUSION: In women with HIV and central obesity, the hepatic steatosis/fibrosis marker Chi3L1 and adiponectin decrease in conjunction with sST2 decreases following switch to RAL. Whether switching from NNRTI/PI-based regimens to RAL can improve hepatic steatosis and dysmetabolism requires further study. TRIAL REGISTRATION: Clinicaltrials.gov NCT00656175.


Assuntos
Adiponectina/uso terapêutico , Proteína 1 Semelhante à Quitinase-3/efeitos dos fármacos , Raltegravir Potássico/farmacologia , Adiponectina/sangue , Adiponectina/metabolismo , Adiponectina/farmacologia , Adulto , Aminoácido Oxirredutases/análise , Aminoácido Oxirredutases/sangue , Fármacos Anti-HIV/uso terapêutico , Biomarcadores/sangue , Contagem de Linfócito CD4 , Proteína 1 Semelhante à Quitinase-3/metabolismo , Fígado Gorduroso/tratamento farmacológico , Fígado Gorduroso/metabolismo , Feminino , Infecções por HIV/tratamento farmacológico , Inibidores da Protease de HIV/uso terapêutico , Humanos , Síndrome Metabólica/metabolismo , Pessoa de Meia-Idade , Obesidade/fisiopatologia , Estudos Prospectivos , Raltegravir Potássico/metabolismo , Raltegravir Potássico/uso terapêutico , Inibidores da Transcriptase Reversa/farmacologia , Inibidores da Transcriptase Reversa/uso terapêutico , Inibidor Tecidual de Metaloproteinase-1/análise , Inibidor Tecidual de Metaloproteinase-1/sangue , Carga Viral
9.
Sci Rep ; 8(1): 7359, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29743555

RESUMO

The aim of this study was to evaluate the effect of UGT1A1 polymorphisms on Raltegravir (RAL) and its metabolite RAL-glucuronide trough plasma concentrations ([RAL]plasma and [RAL-glu]plasma) and on the metabolic ratio (MR): [RAL-glu]plasma/[RAL]plasma. UGT1A1 genotyping was performed on 96 patients. 44% (n = 42) were homozygous UGT1A1*1/*1 while 50% (n = 48) and 6% (n = 6) were UGT1A1*28 and UGT1A1*36 carriers, respectively. The median concentration and interquartile range (IQR) of [RAL]plasma were 88.5 ng/ml (41.0-236), 168 ng/ml (85.8-318) and 92.5 ng/ml (36.4-316) for UGT1A1*1/*1, UGT1A1*28 and UGT1A1*36 carriers, respectively. Only the difference between UGT1A1*1/*1 and *28 carriers was statistically significant (p = 0.022). The median MR (IQR) were 5.8 (3-10), 2.9 (1.6-5.3) and 3.2 (1.7-5.9) for UGT1A1*1/*1, UGT1A1*28 and UGT1A1*36 carriers, respectively. Only the difference between UGT1A1*1/*1 and *28 carriers was statistically significant (p = 0.004) with an allele-dependent effect: UGT1A1*28 homozygous having lower MR than heterozygous carriers who show lower MR compared to *1/*1. Except for the sensation of fatigue, this PK effect did not correlate with clinical adverse events or biological abnormalities. In Conclusion, we demonstrate that UGT1A1*28 polymorphism has a significant impact on RAL metabolism: UGT1A1*28 carriers being characterized by higher [RAL]plasma and lower MR.


Assuntos
Glucuronídeos/sangue , Glucuronosiltransferase/genética , Infecções por HIV/sangue , Infecções por HIV/genética , HIV-1/fisiologia , Polimorfismo Genético , Raltegravir Potássico/metabolismo , Alelos , Estudos de Coortes , Feminino , Genótipo , Glucuronídeos/metabolismo , Infecções por HIV/tratamento farmacológico , Infecções por HIV/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Raltegravir Potássico/uso terapêutico
10.
Clin Infect Dis ; 64(9): 1198-1203, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28369189

RESUMO

Background: Drug reaction with eosinophilia and systemic symptoms (DRESS) syndrome is a rare, severe adverse event during treatment with raltegravir. The occurrence of DRESS syndrome during treatment with other drugs is strongly associated with particular HLA alleles. Methods: We performed HLA testing in 3 of the 5 patients previously reported to have developed raltegravir-induced DRESS syndrome and in 1 previously unreported patient. We then used virtual modeling to visualize interactions between raltegravir and the imputed HLA molecule. Results: Five of the 6 patients who developed raltegravir-induced DRESS syndrome were African, and 1 was Hispanic. HLA typing was performed in 4 patients, all of whom carried both the HLA-B*53 allele and the HLA-C*04 allele to which it is commonly haplotypic. No other HLA alleles were shared by all of the tested patients. Given the approximate prevalence of HLA-B*53 carriage in African (20%) and Hispanic (6%) populations, the probability of all 4 patients being HLA-B*53 carriers, and 2 of 3 African patients being homozygous for HLA-B*53:01, is approximately 0.00002. Conclusions: These data implicate the prevalent African allele HLA-B*53:01 in the immunopathogenesis of raltegravir-induced DRESS syndrome. Although the immunopathogenic mechanisms are currently unknown, virtual modeling suggests that raltegravir may bind within the antigen binding cleft of the HLA-B*53:01 molecule, but not within the closely related HLA-B*35:01 molecule. Further studies are necessary to confirm the strength of the association between carriage of the HLA-B*53:01 allele and raltegravir-induced DRESS syndrome, and the potential utility of HLA screening before raltegravir treatment.


Assuntos
Fármacos Anti-HIV/efeitos adversos , Síndrome de Hipersensibilidade a Medicamentos/genética , Predisposição Genética para Doença , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Antígenos HLA/genética , Raltegravir Potássico/efeitos adversos , Adolescente , Adulto , Alelos , Fármacos Anti-HIV/metabolismo , Fármacos Anti-HIV/uso terapêutico , Feminino , Antígenos HLA/química , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Raltegravir Potássico/metabolismo , Raltegravir Potássico/uso terapêutico
11.
J Pharm Biomed Anal ; 131: 333-344, 2016 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-27632783

RESUMO

BACKGROUND: Analytical methodologies for antiretroviral (ARV) quantification are important in determining both systemic and localized drug concentrations. The CCR5-antagonist maraviroc (MVC), the non-nucleoside reverse transcriptase inhibitors (NNRTIs) etravirine (ETV) and rilpivirine (RPV), as well as the integrase strand transfer inhibitor (INSTI) raltegravir (RAL), have all been evaluated using both oral and non-oral dosing regimens, demonstrating a need for dynamic and sensitive bioanalytical tools for drug quantification in plasma and tissue. METHODS: K2EDTA plasma or blank luminal tissue lysate were spiked with ETV, MVC, RAL, and RPV. Following the addition of isotopically-labeled internal standards and sample extraction via protein precipitation or solid phase extraction, respectively, samples were subjected to liquid chromatographic-tandem mass spectrometric (LC-MS/MS) analysis. Chromatographic separation was performed using a Waters BEH C8, 50×2.1mm, 1.7µm particle size column, and detected on an API 5000 mass analyzer operated in selective reaction monitoring mode. The method was validated according to FDA Bioanalytical Method Validation guidelines. RESULTS: Analytical methods were optimized for the multiplexed monitoring of ETV, MVC, RAL, and RPV in plasma and homogenized tissue lysate. The lower limits of quantification (LLOQs) for ETV, RAL, and RPV were 1ng/mL and the LLOQ for MVC was 0.1ng/mL in plasma; the LLOQs for all ARVs in homogenized tissue lysate was 0.05ng/sample. Standard curves were generated via weighted quadratic (plasma) or linear (tissue) regression of calibrators. Intra- and inter-assay precision and accuracy studies demonstrated %CVs≤15.93% and %DEVs ≤±13.52%, respectively. Stability and matrix effects studies, as well as external proficiency testing assessment, were also performed. All results were acceptable and in accordance with the guidelines recommended by the FDA, Guidance for Industry: Bioanalytical Method Validation document. CONCLUSIONS: LC-MS/MS assays that are sensitive, specific, and dynamic have been developed and validated for the multiplexed quantification of ETV, MVC, RAL, and RPV in plasma and homogenized tissue lysate. The described methods meet sufficient throughput criteria to support large research trials.


Assuntos
Cicloexanos/metabolismo , Piridazinas/metabolismo , Raltegravir Potássico/metabolismo , Rilpivirina/metabolismo , Espectrometria de Massas em Tandem/normas , Triazóis/metabolismo , Cromatografia Líquida/métodos , Cromatografia Líquida/normas , Cicloexanos/análise , Humanos , Maraviroc , Nitrilas , Piridazinas/análise , Pirimidinas , Raltegravir Potássico/análise , Reprodutibilidade dos Testes , Rilpivirina/análise , Espectrometria de Massas em Tandem/métodos , Triazóis/análise
12.
J Antimicrob Chemother ; 71(10): 2782-92, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27334660

RESUMO

OBJECTIVES: To evaluate the role of P-glycoprotein (P-gp) and multidrug-resistant-protein 1 (MRP1) on raltegravir intracellular drug disposition in CD4+ T cells, investigate the effect of HIV-1 infection on P-gp expression and correlate HIV-1 viraemia with P-gp activity in primary CD4+ T cell subsets. METHODS: The cellular accumulation ratio of [(3)H]raltegravir was quantified in CD4+ T cell lines overexpressing either P-gp (CEM-P-gp) or MRP1 (CEM-MRP1) and in primary CD3+CD4+ T cells with high (P-gp(high)) and low P-gp activity (P-gp(low)); inhibition of efflux transporters was confirmed by the intracellular retention of calcein-AM. The correlation of P-gp activity with HIV-1 viraemia was assessed in naive and memory T cell subsets from 21 HIV-1-infected treatment-naive subjects. RESULTS: [(3)H]Raltegravir cellular accumulation ratio decreased in CEM-P-gp cells (P < 0.0001). XR9051 (a P-gp inhibitor) and HIV-1 PIs reversed this phenomenon. Primary CD4+P-gp(high) cells accumulated less raltegravir (38.4% ±â€Š9.6%) than P-gp(low) cells, whereas XR9051 also reversed this effect. In vitro HIV-1 infection of PBMCs and stimulation of CD4+ T cells increased P-gp mRNA and P-gp activity, respectively, while primary CD4+P-gp(high) T cells sustained a higher HIV-1 replication than P-gp(low) cells. A significant correlation between HIV-1 viraemia and P-gp activity was found in different CD4+ T cell subsets, particularly memory CD4+ T cells (r = 0.792, P < 0.0001). CONCLUSIONS: Raltegravir is a substrate of P-gp in CD4+ T cells. Primary CD4+P-gp(high) T cells eliminate intracellular raltegravir more readily than P-gp(low) cells and HIV-1 viraemia correlates with P-gp overall activity. Specific CD4+P-gp(high) T cell subsets could facilitate the persistence of viral replication in vivo and ultimately promote the appearance of drug resistance.


Assuntos
Fármacos Anti-HIV/metabolismo , Linfócitos T CD4-Positivos/efeitos dos fármacos , HIV-1/fisiologia , Raltegravir Potássico/metabolismo , Carga Viral/efeitos dos fármacos , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Compostos de Benzilideno/farmacologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Células Cultivadas , Fluoresceínas/metabolismo , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , Voluntários Saudáveis , Humanos , Memória Imunológica , Ritonavir , Tetra-Hidroisoquinolinas/farmacologia , Viremia/tratamento farmacológico , Replicação Viral/efeitos dos fármacos
13.
Antimicrob Agents Chemother ; 60(5): 3112-4, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26833154

RESUMO

Placental transfer of the HIV integrase inhibitor raltegravir (RLT) was investigated in term human cotyledons in the maternal-to-fetal (n = 3) and fetal-to-maternal (n = 6) directions. In the maternal-to-fetal direction, the mean ± standard deviation (SD) fetal transfer rate (FTR) was 9.1% ± 1.4%, and the mean ± SD clearance index (IC), i.e., RLT FTR/antipyrine FTR, was 0.28 ± 0.05. In the fetal-to-maternal direction, the mean ± SD CI was 0.31 ± 0.09. Placental transfer of RLT was high in both directions.


Assuntos
Feto/metabolismo , Inibidores de Integrase de HIV/metabolismo , Placenta/metabolismo , Raltegravir Potássico/metabolismo , Feminino , Idade Gestacional , Humanos , Troca Materno-Fetal/fisiologia , Gravidez
14.
Antimicrob Agents Chemother ; 59(5): 2572-82, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25691630

RESUMO

The objectives of this study were to investigate raltegravir transport across several blood-tissue barrier models and the potential interactions with drug efflux transporters. Raltegravir uptake, accumulation, and permeability were evaluated in vitro in (i) P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), multidrug resistance-associated protein 1 (MRP1), or MRP4-overexpressing MDA-MDR1 (P-gp), HEK-ABCG2, HeLa-MRP1, or HEK-MRP4 cells, respectively; (ii) cell culture systems of the human blood-brain (hCMEC/D3), mouse blood-testicular (TM4), and human blood-intestinal (Caco-2) barriers; and (iii) rat jejunum and ileum segments using an in situ single-pass intestinal perfusion model. [(3)H]Raltegravir accumulation by MDA-MDR1 (P-gp) and HEK-ABCG2-overexpressing cells was significantly enhanced in the presence of PSC833 {6-[(2S,4R,6E)-4-methyl-2-(methylamino)-3-oxo-6-octenoic acid]-7-L-valine-cyclosporine}, a P-gp inhibitor, or Ko143 [(3S,6S,12aS)-1,2,3,4,6,7,12,12a-octahydro-9-methoxy-6-(2-methylpropyl)-1,4-dioxopyrazino[1',2':1,6]pyrido[3,4-b]indole-3-propanoic acid 1,1-dimethylethyl ester], a BCRP inhibitor, suggesting the inhibition of a P-gp- or BCRP-mediated efflux process, respectively. Furthermore, [(3)H]raltegravir accumulation by human cerebral microvessel endothelial hCMEC/D3 and mouse Sertoli TM4 cells was significantly increased by PSC833 and Ko143. In human intestinal Caco-2 cells grown on Transwell filters, PSC833, but not Ko143, significantly decreased the [(3)H]raltegravir efflux ratios. In rat intestinal segments, [(3)H]raltegravir in situ permeability was significantly enhanced by the concurrent administration of PSC833 and Ko143. In contrast, in the transporter inhibition assays, raltegravir (10 to 500 µM) did not increase the accumulation of substrate for P-gp (rhodamine-6G), BCRP ([(3)H]mitoxantrone), or MRP1 [2',7'-bis(2-carboxyethyl)-5(6)-carboxyfluorescein (BCECF)] by MDA-MDR1 (P-gp)-, HEK-ABCG2-, or HeLa-MRP1-overexpressing cells, respectively. Our data suggest that raltegravir is a substrate but not an inhibitor of the drug efflux transporters P-gp and BCRP. These transporters might play a role in the restriction of raltegravir permeability across the blood-brain, blood-testicular, and blood-intestinal barriers, potentially contributing to its low tissue concentrations and/or low oral bioavailability observed in the clinic setting.


Assuntos
Barreira Hematoencefálica/metabolismo , Raltegravir Potássico/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Fármacos Anti-HIV/metabolismo , Células CACO-2 , Linhagem Celular , Humanos , Íleo/metabolismo , Mucosa Intestinal/metabolismo , Jejuno/metabolismo , Camundongos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA