Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Drug Metab Dispos ; 51(12): 1591-1606, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37751998

RESUMEN

Underestimation of aldehyde oxidase (AO)-mediated clearance by current in vitro assays leads to uncertainty in human dose projections, thereby reducing the likelihood of success in drug development. In the present study we first evaluated the current drug development practices for AO substrates. Next, the overall predictive performance of in vitro-in vivo extrapolation of unbound hepatic intrinsic clearance (CLint,u) and unbound hepatic intrinsic clearance by AO (CLint,u,AO) was assessed using a comprehensive literature database of in vitro (human cytosol/S9/hepatocytes) and in vivo (intravenous/oral) data collated for 22 AO substrates (total of 100 datapoints from multiple studies). Correction for unbound fraction in the incubation was done by experimental data or in silico predictions. The fraction metabolized by AO (fmAO) determined via in vitro/in vivo approaches was found to be highly variable. The geometric mean fold errors (gmfe) for scaled CLint,u (mL/min/kg) were 10.4 for human hepatocytes, 5.6 for human liver cytosols, and 5.0 for human liver S9, respectively. Application of these gmfe's as empirical scaling factors improved predictions (45%-57% within twofold of observed) compared with no correction (11%-27% within twofold), with the scaling factors qualified by leave-one-out cross-validation. A road map for quantitative translation was then proposed following a critical evaluation on the in vitro and clinical methodology to estimate in vivo fmAO In conclusion, the study provides the most robust system-specific empirical scaling factors to date as a pragmatic approach for the prediction of in vivo CLint,u,AO in the early stages of drug development. SIGNIFICANCE STATEMENT: Confidence remains low when predicting in vivo clearance of AO substrates using in vitro systems, leading to de-prioritization of AO substrates from the drug development pipeline to mitigate risk of unexpected and costly in vivo impact. The current study establishes a set of empirical scaling factors as a pragmatic tool to improve predictability of in vivo AO clearance. Developing clinical pharmacology strategies for AO substrates by utilizing mass balance/clinical drug-drug interaction data will help build confidence in fmAO.


Asunto(s)
Aldehído Oxidasa , Hígado , Humanos , Aldehído Oxidasa/metabolismo , Tasa de Depuración Metabólica , Hígado/metabolismo , Hepatocitos/metabolismo , Microsomas Hepáticos/metabolismo
2.
Proc Natl Acad Sci U S A ; 112(3): 755-60, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25564664

RESUMEN

Cytochrome bc1 is a proven drug target in the prevention and treatment of malaria. The rise in drug-resistant strains of Plasmodium falciparum, the organism responsible for malaria, has generated a global effort in designing new classes of drugs. Much of the design/redesign work on overcoming this resistance has been focused on compounds that are presumed to bind the Q(o) site (one of two potential binding sites within cytochrome bc1 using the known crystal structure of this large membrane-bound macromolecular complex via in silico modeling. Cocrystallization of the cytochrome bc1 complex with the 4(1H)-pyridone class of inhibitors, GSK932121 and GW844520, that have been shown to be potent antimalarial agents in vivo, revealed that these inhibitors do not bind at the Q(o) site but bind at the Q(i )site. The discovery that these compounds bind at the Q(i) site may provide a molecular explanation for the cardiotoxicity and eventual failure of GSK932121 in phase-1 clinical trial and highlight the need for direct experimental observation of a compound bound to a target site before chemical optimization and development for clinical trials. The binding of the 4(1H)-pyridone class of inhibitors to Q(i) also explains the ability of this class to overcome parasite Q(o)-based atovaquone resistance and provides critical structural information for future design of new selective compounds with improved safety profiles.


Asunto(s)
Antimaláricos/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Piridonas/metabolismo , Sitios de Unión , Complejo III de Transporte de Electrones/química , Simulación del Acoplamiento Molecular
3.
Artículo en Inglés | MEDLINE | ID: mdl-27799216

RESUMEN

Adequate concentrations of efavirenz in the central nervous system (CNS) are necessary to suppress viral replication, but high concentrations may increase the likelihood of CNS adverse drug reactions. The aim of this investigation was to evaluate the efavirenz distribution in the cerebrospinal fluid (CSF) and the brain by using a physiologically based pharmacokinetic (PBPK) simulation for comparison with rodent and human data. The efavirenz CNS distribution was calculated using a permeability-limited model on a virtual cohort of 100 patients receiving efavirenz (600 mg once daily). Simulation data were then compared with human data from the literature and with rodent data. Wistar rats were administered efavirenz (10 mg kg of body weight-1) once daily over 5 weeks. Plasma and brain tissue were collected for analysis via liquid chromatography-tandem mass spectrometry (LC-MS/MS). The median maximum concentrations of drug (Cmax) were predicted to be 3,184 ng ml-1 (interquartile range [IQR], 2,219 to 4,851 ng ml-1), 49.9 ng ml-1 (IQR, 36.6 to 69.7 ng ml-1), and 50,343 ng ml-1 (IQR, 38,351 to 65,799 ng ml-1) in plasma, CSF, and brain tissue, respectively, giving a tissue-to-plasma ratio of 15.8. Following 5 weeks of oral dosing of efavirenz (10 mg kg-1), the median plasma and brain tissue concentrations in rats were 69.7 ng ml-1 (IQR, 44.9 to 130.6 ng ml-1) and 702.9 ng ml-1 (IQR, 475.5 to 1,018.0 ng ml-1), respectively, and the median tissue-to-plasma ratio was 9.5 (IQR, 7.0 to 10.9). Although it is useful, measurement of CSF concentrations may give an underestimation of the penetration of antiretrovirals into the brain. The limitations associated with obtaining tissue biopsy specimens and paired plasma and CSF samples from patients make PBPK modeling an attractive tool for probing drug distribution.


Asunto(s)
Fármacos Anti-VIH/farmacocinética , Benzoxazinas/farmacocinética , Encéfalo/metabolismo , Modelos Estadísticos , Administración Oral , Alquinos , Animales , Fármacos Anti-VIH/sangre , Fármacos Anti-VIH/líquido cefalorraquídeo , Benzoxazinas/sangre , Benzoxazinas/líquido cefalorraquídeo , Simulación por Computador , Ciclopropanos , Esquema de Medicación , Cálculo de Dosificación de Drogas , Humanos , Masculino , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica , Ratas , Ratas Wistar , Distribución Tisular
4.
Artículo en Inglés | MEDLINE | ID: mdl-28416547

RESUMEN

Tenofovir disoproxil fumarate (TDF), a prodrug of tenofovir, has oral bioavailability (25%) limited by intestinal transport (P-glycoprotein), and intestinal degradation (carboxylesterase). However, the influence of luminal pancreatic enzymes is not fully understood. Physiologically based pharmacokinetic (PBPK) modeling has utility for estimating drug exposure from in vitro data. This study aimed to develop a PBPK model that included luminal enzyme activity to inform dose reduction strategies. TDF and tenofovir stability in porcine pancrelipase concentrations was assessed (0, 0.48, 4.8, 48, and 480 U/ml of lipase; 1 mM TDF; 37°C; 0 to 30 min). Samples were analyzed using mass spectrometry. TDF stability and permeation data allowed calculation of absorption rates within a human PBPK model to predict plasma exposure following 6 days of once-daily dosing with 300 mg of TDF. Regional absorption of drug was simulated across gut segments. TDF was degraded by pancrelipase (half-lives of 0.07 and 0.62 h using 480 and 48 U/ml, respectively). Previously reported maximum concentration (Cmax; 335 ng/ml), time to Cmax (Tmax; 2.4 h), area under the concentration-time curve from 0 to 24 h (AUC0-24; 3,045 ng · h/ml), and concentration at 24 h (C24; 48.3 ng/ml) were all within a 0.5-fold difference from the simulated Cmax (238 ng/ml), Tmax (3 h), AUC0-24 (3,036 ng · h/ml), and C24 (42.7 ng/ml). Simulated TDF absorption was higher in duodenum and jejunum than in ileum (p<0.05). These data support that TDF absorption is limited by the action of intestinal lipases. Our results suggest that bioavailability may be improved by protection of drug from intestinal transporters and enzymes, for example, by coadministration of enzyme-inhibiting agents or nanoformulation strategies.


Asunto(s)
Fármacos Anti-VIH/farmacología , Fármacos Anti-VIH/farmacocinética , Tenofovir/farmacología , Tenofovir/farmacocinética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Adolescente , Adulto , Carboxilesterasa/metabolismo , Infecciones por VIH/metabolismo , Humanos , Lipasa/metabolismo , Masculino , Persona de Mediana Edad , Pancrelipasa/metabolismo , Adulto Joven
5.
J Antimicrob Chemother ; 69(12): 3275-81, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25114168

RESUMEN

OBJECTIVES: Recent clinical data have suggested high raltegravir concentrations in gut tissue after oral administration, with implications for treatment and prevention. We have used in silico, in vitro, ex vivo and in vivo models to further investigate the accumulation of raltegravir in gut tissue. METHODS: Affinity of raltegravir for gut tissue was assessed in silico (Poulin-Theil method), in vitro (Caco-2 accumulation) and ex vivo (rat intestine) and compared with the lipophilic drug lopinavir. Finally, raltegravir concentrations in plasma, gut contents, small intestine and large intestine were determined after oral dosing to Wistar rats 1 and 4 h post-dose. Samples were analysed using LC-MS/MS and scintillation counting. RESULTS: Gut tissue accumulation of raltegravir was less than for lopinavir in silico, in vitro and ex vivo (P < 0.05). After oral administration to rats, raltegravir concentrations 4 h post-dose were lower in plasma (0.05 µM) compared with small intestine (0.47 µM, P = 0.06) and large intestine (1.36 µM, P < 0.05). However, raltegravir concentrations in the contents of both small intestine (4.0 µM) and large intestine (40.6 µM) were also high. CONCLUSIONS: In silico, in vitro and ex vivo data suggest low raltegravir accumulation in intestinal tissue. In contrast, in vivo animal data suggest raltegravir concentrates in intestinal tissue even when plasma concentrations are minimal. However, high raltegravir concentrations in gut contents are the likely driving factor behind this observation, rather than blood-to-tissue drug distribution. The methods described can be combined with clinical investigations to provide a complete strategy for selection of drugs with high gut accumulation.


Asunto(s)
Fármacos Anti-VIH/farmacocinética , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/prevención & control , Intestinos/química , Profilaxis Pre-Exposición , Pirrolidinonas/farmacocinética , Administración Oral , Animales , Fármacos Anti-VIH/administración & dosificación , Cromatografía Liquida , Masculino , Pirrolidinonas/administración & dosificación , Raltegravir Potásico , Ratas Wistar , Espectrometría de Masas en Tándem
6.
Antimicrob Agents Chemother ; 57(11): 5612-8, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24002095

RESUMEN

Rilpivirine is a nonnucleoside reverse transcriptase inhibitor approved for treatment of HIV-1 infection in antiretroviral-naive adult patients. Potential interactions with drug transporters have not been fully investigated. Transport by and inhibition of drug transporters by rilpivirine were analyzed to further understand the mechanisms governing rilpivirine exposure and determine the potential for transporter-mediated drug-drug interactions. The ability of rilpivirine to inhibit or be transported by ABCB1 was determined using ABCB1-overexpressing CEMVBL100 cells and Caco-2 cell monolayers. The Xenopus laevis oocyte heterologous protein expression system was used to clarify if rilpivirine was either transported by or inhibited the function of influx transporters SLCO1A2, SLCO1B1, SLCO1B3, SLC22A2, SLC22A6, and SLC22A8. The ability of rilpivirine to inhibit or be transported by SLC22A1 was determined using SLC22A1-expressing KCL22 cells. Rilpivirine showed higher accumulation in SLC22A1-overexpressing KCL22 cells than control cells (27% increase, P = 0.03) and inhibited the functionality of SLC22A1 and SLC22A2 transport with 50% inhibitory concentrations (IC50s) of 28.5 µM and 5.13 µM, respectively. Inhibition of ABCB1-mediated digoxin transport was determined for rilpivirine, which inhibited digoxin transport in the B-to-A direction with an IC50 of 4.48 µM. The maximum rilpivirine concentration in plasma in patients following a standard 25-mg dosing regimen is around 0.43 µM, lower than that necessary to substantially inhibit ABCB1, SLC22A1, or SLC22A2 in vitro. However, these data indicate that SLC22A1 may contribute to variability in rilpivirine exposure and that interactions of rilpivirine with substrates of SLC22A1, SLC22A2, or ABCB1 may be possible.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Fármacos Anti-VIH/farmacología , Nitrilos/farmacología , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Transportador 1 de Catión Orgánico/antagonistas & inhibidores , Pirimidinas/farmacología , Inhibidores de la Transcriptasa Inversa/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Línea Celular Tumoral , Digoxina/metabolismo , Relación Dosis-Respuesta a Droga , Expresión Génica , VIH-1/química , VIH-1/enzimología , Humanos , Cinética , Linfocitos/citología , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Oocitos/citología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 1 de Catión Orgánico/genética , Transportador 1 de Catión Orgánico/metabolismo , Transportador 2 de Cátion Orgánico , Rilpivirina , Transfección , Xenopus laevis
7.
J Antimicrob Chemother ; 68(7): 1627-34, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23515248

RESUMEN

OBJECTIVES: Raltegravir pharmacokinetics (PK) show high intra- and inter-patient variability and are also influenced by co-administered substances that alter the gastrointestinal tract environment, such as pH-altering or metal-containing agents. The aim of this investigation was to develop a population-based absorption, distribution, metabolism and excretion (ADME) model to investigate the effects of gastrointestinal pH and ingested magnesium on raltegravir PK. METHODS: In vitro data describing the disposition of raltegravir were obtained from literature sources or generated by standard methods. Raltegravir (400 mg single dose) PK were simulated in healthy volunteers (50 subjects per group, 20-50 years old, 0.5 proportion female subjects) over a 12 h period. RESULTS: Simulated raltegravir PK correlated well with data from clinical trials, with a mean deviation in C(max), AUC(0-12) and C(trough) of <20%. Solubility of raltegravir in the gastrointestinal tract was increased at higher luminal pH. Increased intestinal pH and transit time both correlated with higher raltegravir absorption (P<0.001). Magnesium ingestion reduced raltegravir exposure in simulated subjects, with mean C(trough) reduced by 32% (P<0.001). CONCLUSIONS: The in vitro-in vivo extrapolation model developed in this study predicted raltegravir PK in virtual individuals with different gastrointestinal pH profiles. The main PK variables were predicted with good accuracy compared with reference data, and both luminal pH and magnesium were able to influence drug absorption. This modelling system provides a tool for investigating the absorption of other drugs, including HIV integrase inhibitors currently in development, which have also shown interactions with food and metal-containing products.


Asunto(s)
Fármacos Anti-VIH/farmacocinética , Absorción Intestinal , Pirrolidinonas/farmacocinética , Adulto , Fármacos Anti-VIH/administración & dosificación , Femenino , Voluntarios Sanos , Humanos , Concentración de Iones de Hidrógeno , Intestinos/química , Magnesio/metabolismo , Masculino , Persona de Mediana Edad , Modelos Estadísticos , Pirrolidinonas/administración & dosificación , Raltegravir Potásico , Adulto Joven
8.
J Antimicrob Chemother ; 68(5): 977-85, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23292347

RESUMEN

Atovaquone is used as a fixed-dose combination with proguanil (Malarone) for treating children and adults with uncomplicated malaria or as chemoprophylaxis for preventing malaria in travellers. Indeed, in the USA, between 2009 and 2011, Malarone prescriptions accounted for 70% of all antimalarial pre-travel prescriptions. In 2013 the patent for Malarone will expire, potentially resulting in a wave of low-cost generics. Furthermore, the malaria scientific community has a number of antimalarial quinolones with a related pharmacophore to atovaquone at various stages of pre-clinical development. With this in mind, it is timely here to review the current knowledge of atovaquone, with the purpose of aiding the decision making of clinicians and drug developers involved in the future use of atovaquone generics or atovaquone derivatives.


Asunto(s)
Antimaláricos/uso terapéutico , Atovacuona/uso terapéutico , Malaria/tratamiento farmacológico , Antimaláricos/farmacología , Atovacuona/farmacología , Quimioprevención/métodos , Combinación de Medicamentos , Humanos , Malaria/prevención & control , Proguanil/farmacología , Proguanil/uso terapéutico , Estados Unidos
9.
J Antimicrob Chemother ; 68(4): 869-80, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23228936

RESUMEN

OBJECTIVES: Phenothiazines have been shown to exhibit in vitro and in vivo activity against Mycobacterium tuberculosis (Mtb) and multidrug-resistant Mtb. They are predicted to target the genetically validated respiratory chain component type II NADH:quinone oxidoreductase (Ndh). Using a set of compounds containing the phenothiazine pharmacophore, we have (i) investigated whether chemical validation data support the molecular target and (ii) evaluated pharmacophore tractability for further drug development. METHODS: Recombinant Mtb Ndh was generated and its functionality confirmed by steady-state kinetics. Pharmacodynamic profiling of the phenothiazines, including antitubercular efficacy in aerobic and O2-limited conditions, time-kill assays and isobole analyses against first-line antituberculars, was performed. Potential mitochondrial toxicity was assessed in a modified HepG2 cell-line assay and against bovine cytochrome bc1. RESULTS: Steady-state kinetic analyses revealed a substrate preference for coenzyme Q2 and an inability to utilize NADPH. A positive correlation between recombinant Ndh inhibition and kill of aerobically cultured Mtb was observed, whilst enhanced potency was demonstrated in a hypoxic model. Time-kill studies revealed the phenothiazines to be bactericidal whilst isobolograms exposed antagonism with isoniazid, indicative of intracellular NADH/NAD(+) couple perturbation. At therapeutic levels, phenothiazine-mediated toxicity was appreciable; however, specific mitochondrial targeting was excluded. CONCLUSIONS: Data generated support the hypothesis that Ndh is the molecular target of phenothiazines. The favourable pharmacodynamic properties of the phenothiazines are consistent with a target product profile that includes activity against dormant/persistent bacilli, rapid bactericidal activity and activity against drug-resistant Mtb by a previously unexploited mode of action. These properties warrant further medicinal chemistry to improve potency and safety.


Asunto(s)
Antituberculosos/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Fenotiazinas/farmacología , Antituberculosos/química , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Viabilidad Microbiana/efectos de los fármacos , Mycobacterium tuberculosis/fisiología , Fenotiazinas/química
10.
Antimicrob Agents Chemother ; 56(6): 3020-6, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22450971

RESUMEN

Raltegravir shows marked pharmacokinetic variability in patients, with gastrointestinal pH and divalent-metal binding being potential factors. We investigated raltegravir solubility, lipophilicity, pK(a), and permeativity in vitro to elucidate known interactions with omeprazole, antacids, and food, all of which increase gastric pH. Solubility of raltegravir was determined at pH 1 to 8. Lipophilicity of raltegravir was determined using octanol-water partition. Raltegravir pK(a) was determined using UV spectroscopy. The effects of pH, metal salts, and omeprazole on the cellular permeativity of raltegravir were determined using Caco-2 monolayers. Cellular accumulation studies were used to determine the effect of interplay between pH and ABCB1 transport on raltegravir accumulation. Samples were analyzed using liquid chromatography-tandem mass spectroscopy (LC-MS/MS) or scintillation counting. Raltegravir at 10 mM was partly insoluble at pH 6.6 and below. Raltegravir lipophilicity was pH dependent and was reduced as pH was increased from 5 to 9. The pK(a) of raltegravir was 6.7. Raltegravir cellular permeativity was heavily influenced by changes in extracellular pH, where apical-to-basolateral permeativity was reduced 9-fold (P < 0.05) when apical pH was increased from 5 to 8.5. Raltegravir cellular permeativity was also reduced in the presence of magnesium and calcium. Omeprazole did not alter raltegravir cellular permeativity. Cellular accumulation of raltegravir was increased independently by inhibiting ABCB1 and by lowering extracellular pH from pH 8 to 5. Gastrointestinal pH and polyvalent metals can potentially alter the pharmacokinetic properties of raltegravir, and these data provide an explanation for the variability in raltegravir exposure in patients. The evaluation of how divalent-metal-containing products, such as multivitamins, that do not affect gastric pH alter raltegravir pharmacokinetics in patients is now justified.


Asunto(s)
Cationes Bivalentes/metabolismo , Pirrolidinonas/metabolismo , Pirrolidinonas/farmacología , Células CACO-2 , Calcio/metabolismo , Membrana Celular/efectos de los fármacos , Cromatografía Liquida , Interacciones Farmacológicas , Humanos , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Magnesio/metabolismo , Omeprazol/metabolismo , Omeprazol/farmacología , Raltegravir Potásico , Solubilidad , Espectrometría de Masas en Tándem
11.
Ther Drug Monit ; 34(2): 232-5, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22406652

RESUMEN

INTRODUCTION: Raltegravir (RAL) is the first in class integrase inhibitor and is licensed for administration at 400 mg twice daily. RAL pharmacokinetics are characterized by high interpatient variability and recently RAL plasma exposure has been correlated with efficacy. RAL is primarily metabolized by glucuronidation via uridine diphosphate glucuronosyltransferase 1A1 (UGT1A1) and UGT1A1*28 considered to be the main genetic variant associated with decreased UGT1A1 expression. This study investigated variability in RAL trough plasma concentrations (Ctrough) in the clinical setting, the effect of UGT1A1*28 and concomitant antiretrovirals. METHODS: A total of 86 patients, from Turin, Italy, and Madrid, Spain, were included in the analysis. Blood samples were obtained 10-14 hours postdose. Genotyping for UGT1A1*28 was conducted by sequencing. RESULTS: High interpatient and intrapatient variabilities were observed; 13 patients had ≥3 samples available, and the median coefficient of variation was 128 (64-265). Coadministration of RAL with atazanavir (ATV, n = 9) resulted in higher raltegravir Ctrough, 517 (307-2706) ng/mL when compared with patients not receiving ATV (n = 77) 223 (95-552; P = 0.02). UGT1A1*28 did not influence RAL plasma exposure. DISCUSSION: We have documented large intersubject and intrasubject variabilities in RAL plasma concentrations and confirmed the interaction with ATV. Further studies are required to better understand the mechanisms that influence the pharmacokinetics of RAL.


Asunto(s)
Glucuronosiltransferasa/genética , Inhibidores de Integrasa VIH/farmacocinética , Pirrolidinonas/farmacocinética , Adulto , Fármacos Anti-VIH/administración & dosificación , Fármacos Anti-VIH/farmacocinética , Fármacos Anti-VIH/farmacología , Sulfato de Atazanavir , Interacciones Farmacológicas , Monitoreo de Drogas , Quimioterapia Combinada , Femenino , Regulación Enzimológica de la Expresión Génica , Variación Genética , Genotipo , Humanos , Italia , Masculino , Persona de Mediana Edad , Oligopéptidos/farmacología , Piridinas/farmacología , Raltegravir Potásico , España
12.
Antimicrob Agents Chemother ; 55(2): 879-87, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21078936

RESUMEN

The identification of transporters of the HIV integrase inhibitor raltegravir could be a factor in an understanding of the pharmacokinetic-pharmacodynamic relationship and reported drug interactions of raltegravir. Here we determined whether raltegravir was a substrate for ABCB1 or the influx transporters SLCO1A2, SLCO1B1, SLCO1B3, SLC22A1, SLC22A6, SLC10A1, SLC15A1, and SLC15A2. Raltegravir transport by ABCB1 was studied with CEM, CEM(VBL100), and Caco-2 cells. Transport by uptake transporters was assessed by using a Xenopus laevis oocyte expression system, peripheral blood mononuclear cells, and primary renal cells. The kinetics of raltegravir transport and competition between raltegravir and tenofovir were also investigated using SLC22A6-expressing oocytes. Raltegravir was confirmed to be an ABCB1 substrate in CEM, CEM(VBL100), and Caco-2 cells. Raltegravir was also transported by SLC22A6 and SLC15A1 in oocyte expression systems but not by other transporters studied. The K(m) and V(max) for SLC22A6 transport were 150 µM and 36 pmol/oocyte/h, respectively. Tenofovir and raltegravir competed for SLC22A6 transport in a concentration-dependent manner. Raltegravir inhibited 1 µM tenofovir with a 50% inhibitory concentration (IC(50)) of 14.0 µM, and tenofovir inhibited 1 µM raltegravir with an IC(50) of 27.3 µM. Raltegravir concentrations were not altered by transporter inhibitors in peripheral blood mononuclear cells or primary renal cells. Raltegravir is a substrate for SLC22A6 and SLC15A1 in the oocyte expression system. However, transport was limited compared to endogenous controls, and these transporters are unlikely to have a great impact on raltegravir pharmacokinetics.


Asunto(s)
Adenina/análogos & derivados , Fármacos Anti-VIH/metabolismo , Inhibidores de Integrasa VIH/metabolismo , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Organofosfonatos/metabolismo , Pirrolidinonas/metabolismo , Inhibidores de la Transcriptasa Inversa/metabolismo , Adenina/metabolismo , Animales , Transporte Biológico , Células CACO-2 , Línea Celular , Interacciones Farmacológicas , Humanos , Riñón/citología , Riñón/metabolismo , Leucocitos Mononucleares/metabolismo , Oocitos/metabolismo , Raltegravir Potásico , Especificidad por Sustrato , Tenofovir , Xenopus laevis/metabolismo
13.
Pharmacogenet Genomics ; 20(12): 759-65, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21217360

RESUMEN

BACKGROUND: Organic anion transporting polypeptides (OATPs) are emerging as major determinants of pharmacokinetics for numerous drugs, with the 1B1 isoform-mediating hepatic uptake. The 521 T>C polymorphism has been correlated earlier with higher plasma concentrations of several drugs and the aim of this study was to determine whether this polymorphism influences trough concentrations of maraviroc. METHODS: The uptake of maraviroc by OATP1B1 was assessed using a heterologous Xenopus laevis oocyte expression system and quantified using a novel liquid chromatography-mass spectrometry method. Regression analyses were conducted to identify factors associated with maraviroc Ctrough in 59 patients treated with maraviroc at 150, 300, or 600 mg twice daily. RESULTS: Maraviroc was identified as a substrate for OATP1B1 with a Km of 33.9 µmol/l. A dose of 600 mg of etravirine or efavirenz [odds ratio (OR) = 0.22, 95% confidence interval (95% CI): 0.06-0.76; P = 0.016] and SLCO1B1 521 heterozygosity were both associated with maraviroc Ctrough, above the suggested target concentration of 50 ng/ml (OR = 20.3, 95% CI: 2.2-182; P = 0.007). CONCLUSION: These findings show the importance of OATP1B1 for variability in maraviroc pharmacokinetics. Furthermore, the SLCO1B1 521 T>C polymorphism maybe useful in predicting higher plasma concentrations but these data should be confirmed before prospective clinical studies to define the clinical usefulness.


Asunto(s)
Ciclohexanos/sangre , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Polimorfismo de Nucleótido Simple/genética , Triazoles/sangre , Adulto , Animales , Transporte Biológico/efectos de los fármacos , Cromatografía Liquida , Ciclohexanos/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Estudios de Asociación Genética , Genotipo , Humanos , Transportador 1 de Anión Orgánico Específico del Hígado , Masculino , Maraviroc , Espectrometría de Masas , Persona de Mediana Edad , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Reproducibilidad de los Resultados , Especificidad por Sustrato/efectos de los fármacos , Triazoles/farmacología , Xenopus laevis
14.
Expert Rev Gastroenterol Hepatol ; 12(3): 223-236, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29088978

RESUMEN

INTRODUCTION: Nano-scale formulations are being developed to improve the delivery of orally administered medicines, and the interactions between nanoformulations and the gastrointestinal luminal, mucosal and epithelial environment is currently being investigated. The mucosal surface of the gastrointestinal tract is capable of trapping and eliminating large particles and pathogens as part of the natural defences of the body, it is becoming clearer that nanoformulation properties such as particle size, charge, and shape, as well as mucous properties such as viscoelasticity, thickness, density, and turn-over time are all relevant to these interactions. However, progress has been slow to utilise this information to produce effective mucous-penetrating particles. Areas covered: This review focuses on delivery method of nanomedicines both into and across the gastrointestinal mucosal surface, and aims to summarise the biological barriers that exist to successful oral nanomedicine delivery and how these barriers may be investigated and overcome. Expert commentary: Despite successes in the laboratory, no nanotechnology-enabled products are currently in clinical use which either specifically target the intestinal mucous surface or cross the epithelial barrier intact. New nanomedicine-based treatments of local diseases (intestinal cancer, inflammation, infection) and systemic diseases are advancing towards clinical use, and offer genuine opportunities to improve therapy.


Asunto(s)
Membrana Celular/metabolismo , Mucosa Intestinal/metabolismo , Nanopartículas/administración & dosificación , Nanopartículas/metabolismo , Uniones Estrechas/metabolismo , Administración Oral , Animales , Bilis , Sistemas de Liberación de Medicamentos , Endocitosis , Exocitosis , Jugo Gástrico , Humanos , Absorción Intestinal , Moco , Permeabilidad , Saliva , Transcitosis
15.
Pharmaceutics ; 9(4)2017 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-28946666

RESUMEN

Pancreatic cancer has been classified as a cancer of unmet need. After diagnosis the patient prognosis is dismal with few surviving over 5 years. Treatment regimes are highly patient variable and often the patients are too sick to undergo surgical resection or chemotherapy. These chemotherapies are not effective often because patients are diagnosed at late stages and tumour metastasis has occurred. Nanotechnology can be used in order to formulate potent anticancer agents to improve their physicochemical properties such as poor aqueous solubility or prolong circulation times after administration resulting in improved efficacy. Studies have reported the use of nanotechnologies to improve the efficacy of gemcitabine (the current first line treatment) as well as investigating the potential of using other drug molecules which have previously shown promise but were unable to be utilised due to the inability to administer through appropriate routes-often related to solubility. Of the nanotechnologies reported, many can offer site specific targeting to the site of action as well as a plethora of other multifunctional properties such as image guidance and controlled release. This review focuses on the use of the major nanotechnologies both under pre-clinical development and those which have recently been approved for use in pancreatic cancer therapy.

16.
Sci Rep ; 7(1): 5410, 2017 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-28710496

RESUMEN

Pre-clinical and retrospective studies of patients using statins to reduce plasma cholesterol have suggested that statins may be useful to treat cancer. However, prospective clinical trials have yet to demonstrate significant efficacy. We have previously shown that this is in part because a hydrophobic statin with a long half-life is necessary. Pitavastatin, the only statin with this profile, has not undergone clinical evaluation in oncology. The target of pitavastatin, hydroxymethylglutarate coenzyme-A reductase (HMGCR), was found to be over-expressed in all ovarian cancer cell lines examined and upregulated by mutated TP53, a gene commonly altered in ovarian cancer. Pitavastatin-induced apoptosis was blocked by geranylgeraniol and mevalonate, products of the HMGCR pathway, confirming that pitavastatin causes cell death through inhibition of HMGCR. Solvent extracts of human and mouse food were also able to block pitavastatin-induced apoptosis, suggesting diet might influence the outcome of clinical trials. When nude mice were maintained on a diet lacking geranylgeraniol, oral pitavastatin caused regression of Ovcar-4 tumour xenografts. However, when the animal diet was supplemented with geranylgeraniol, pitavastatin failed to prevent tumour growth. This suggests that a diet containing geranylgeraniol can limit the anti-tumour activity of pitavastatin and diet should be controlled in clinical trials of statins.


Asunto(s)
Dieta , Diterpenos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Quinolinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Diterpenos/administración & dosificación , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Hidroximetilglutaril-CoA Reductasas/genética , Hidroximetilglutaril-CoA Reductasas/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Ratones Desnudos , Ratones SCID , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Quinolinas/administración & dosificación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
17.
J Med Chem ; 60(9): 3703-3726, 2017 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-28304162

RESUMEN

A high-throughput screen (HTS) was undertaken against the respiratory chain dehydrogenase component, NADH:menaquinone oxidoreductase (Ndh) of Mycobacterium tuberculosis (Mtb). The 11000 compounds were selected for the HTS based on the known phenothiazine Ndh inhibitors, trifluoperazine and thioridazine. Combined HTS (11000 compounds) and in-house screening of a limited number of quinolones (50 compounds) identified ∼100 hits and four distinct chemotypes, the most promising of which contained the quinolone core. Subsequent Mtb screening of the complete in-house quinolone library (350 compounds) identified a further ∼90 hits across three quinolone subtemplates. Quinolones containing the amine-based side chain were selected as the pharmacophore for further modification, resulting in metabolically stable quinolones effective against multi drug resistant (MDR) Mtb. The lead compound, 42a (MTC420), displays acceptable antituberculosis activity (Mtb IC50 = 525 nM, Mtb Wayne IC50 = 76 nM, and MDR Mtb patient isolates IC50 = 140 nM) and favorable pharmacokinetic and toxicological profiles.


Asunto(s)
Mycobacterium tuberculosis/efectos de los fármacos , Quinolonas/síntesis química , Quinolonas/farmacología , Animales , Células CACO-2 , Espectroscopía de Resonancia Magnética con Carbono-13 , Diseño de Fármacos , Transporte de Electrón/efectos de los fármacos , Células Hep G2 , Ensayos Analíticos de Alto Rendimiento , Humanos , Pruebas de Sensibilidad Microbiana , Mycobacterium tuberculosis/metabolismo , Espectroscopía de Protones por Resonancia Magnética , Quinolonas/química , Quinolonas/farmacocinética , Ratas , Espectrometría de Masa por Ionización de Electrospray , Relación Estructura-Actividad , Pruebas de Toxicidad
18.
Bioanalysis ; 8(20): 2125-34, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27611731

RESUMEN

AIM: The non-nucleoside reverse transcriptase inhibitor efavirenz is one of the most prescribed antiretroviral therapeutics. Efavirenz-containing therapy has become associated with the occurrence of CNS side effects, including sleep disturbances, depression and even psychosis. RESULTS: The investigation of efavirenz distribution required the development of a versatile and sensitive method. In addition to plasma, quantification was required in brain tissue and phosphate-buffered saline. The assay presented here was linear from 1.9 to 500 ng/ml. Accuracy and precision ranged between 93.7 and 99.5%, and 1.5 and 5.6%, respectively. DISCUSSION: The method developed here represents a versatile, sensitive and easy-to-use assay. The assay has been applied to in vitro and in vivo samples demonstrating reliable efavirenz quantification in multiple matrices.


Asunto(s)
Benzoxazinas/análisis , Cromatografía Líquida de Alta Presión , Pruebas de Química Clínica/métodos , Inhibidores de la Transcriptasa Inversa/análisis , Espectrometría de Masas en Tándem , Alquinos , Animales , Benzoxazinas/sangre , Benzoxazinas/normas , Encéfalo/metabolismo , Cromatografía Líquida de Alta Presión/normas , Ciclopropanos , Masculino , Control de Calidad , Ratas , Ratas Wistar , Inhibidores de la Transcriptasa Inversa/sangre , Inhibidores de la Transcriptasa Inversa/normas , Espectrometría de Masas en Tándem/normas
19.
Nat Commun ; 7: 13184, 2016 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-27767027

RESUMEN

Considerable scope exists to vary the physical and chemical properties of nanoparticles, with subsequent impact on biological interactions; however, no accelerated process to access large nanoparticle material space is currently available, hampering the development of new nanomedicines. In particular, no clinically available nanotherapies exist for HIV populations and conventional paediatric HIV medicines are poorly available; one current paediatric formulation utilizes high ethanol concentrations to solubilize lopinavir, a poorly soluble antiretroviral. Here we apply accelerated nanomedicine discovery to generate a potential aqueous paediatric HIV nanotherapy, with clinical translation and regulatory approval for human evaluation. Our rapid small-scale screening approach yields large libraries of solid drug nanoparticles (160 individual components) targeting oral dose. Screening uses 1 mg of drug compound per library member and iterative pharmacological and chemical evaluation establishes potential candidates for progression through to clinical manufacture. The wide applicability of our strategy has implications for multiple therapy development programmes.


Asunto(s)
Fármacos Anti-VIH/farmacología , Sistemas de Liberación de Medicamentos , Infecciones por VIH/tratamiento farmacológico , Lopinavir/farmacología , Nanopartículas/química , Ritonavir/farmacología , Administración Oral , Animales , Fármacos Anti-VIH/sangre , Fármacos Anti-VIH/farmacocinética , Células CACO-2 , Permeabilidad de la Membrana Celular , Niño , Infecciones por VIH/sangre , Infecciones por VIH/virología , Ensayos Analíticos de Alto Rendimiento , Humanos , Lopinavir/sangre , Lopinavir/farmacocinética , Masculino , Nanomedicina/métodos , Nanopartículas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Ritonavir/sangre , Ritonavir/farmacocinética , Bibliotecas de Moléculas Pequeñas/análisis , Investigación Biomédica Traslacional/métodos
20.
Front Pharmacol ; 6: 18, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25708098

RESUMEN

[This corrects the article on p. 248 in vol. 5, PMID: 25426075.].

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA