Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Toxicol In Vitro ; 87: 105533, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36473578

RESUMO

Bile acid (BA) homeostasis is a complex and precisely regulated process to prevent impaired BA flow and the development of cholestasis. Several reactions, namely hydroxylation, glucuronidation and sulfation are involved in BA detoxification. In the present study, we employed a comprehensive approach to identify the key enzymes involved in BA metabolism using human recombinant enzymes, human liver microsomes (HLM) and human liver cytosol (HLC). We showed that CYP3A4 was a crucial step for the metabolism of several BAs and their taurine and glycine conjugated forms and quantitatively described their metabolites. Glucuronidation and sulfation were also identified as important drivers of the BA detoxification process in humans. Moreover, lithocholic acid (LCA), the most hydrophobic BA with the highest toxicity potential, was a substrate for all investigated processes, demonstrating the importance of hepatic metabolism for its clearance. Collectively, this study identified CYP3A4, UGT1A3, UGT2B7 and SULT2A1 as the major contributing (metabolic) processes in the BA detoxification network. Inhibition of these enzymes by drug candidates is therefore considered as a critical mechanism in the manifestation of drug-induced cholestasis in humans and should be addressed during the pre-clinical development.


Assuntos
Ácidos e Sais Biliares , Colestase , Humanos , Ácidos e Sais Biliares/metabolismo , Citocromo P-450 CYP3A/metabolismo , Colestase/induzido quimicamente , Colestase/metabolismo , Microssomos Hepáticos/metabolismo , Homeostase , Fígado/metabolismo , Glucuronosiltransferase/metabolismo
2.
Drug Metab Pharmacokinet ; 39: 100400, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34146821

RESUMO

Although Accelerator Mass Spectrometry (AMS) offers unparalleled sensitivity by investigating the fate of 14C-labeled compounds within the organism, its widespread use in ADME (absorption, distribution, metabolism, excretion) studies is limited. Conventional approaches based on Liquid Scintillation Counting (LSC) are still preferred, in particular because of complexity and costs associated with AMS measurements. Progress made over the last decade towards more compact AMS systems increased the interest in a combustion-based AMS approach allowing the analysis of samples in gaseous form. Thus, a novel gas Double Trap Interface (DTI) was designed, providing high sample throughput for the analysis of biomedical samples. DTI allows the coupling of an Elemental Analyzer (EA) for sample combustion to the hybrid ion source of a MICADAS (MIni CArbon DAting System) AMS system. The performance was evaluated in two studies through the analysis of more than 1000 samples from 14C-labeled biomatrices and fractions collected after liquid chromatography (LC). The covered activity ranged from 1 to 1000 mBq/g for labeled biomatrices and from 1 to 10000 mBq/g(C) for LC fractions. The implemented routine allows automated measurements requiring less than 5 min per sample (12-13 analyses per hour) without the need for sample conversion to graphite.


Assuntos
Preparações Farmacêuticas , Farmacocinética , Radioisótopos de Carbono , Cromatografia Gasosa-Espectrometria de Massas/métodos , Ensaios de Triagem em Larga Escala/instrumentação , Ensaios de Triagem em Larga Escala/métodos , Humanos , Marcação por Isótopo/métodos , Preparações Farmacêuticas/análise , Preparações Farmacêuticas/metabolismo , Contagem de Cintilação/métodos
3.
Drug Metab Dispos ; 49(7): 548-562, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33952610

RESUMO

Tropifexor (NVP-LJN452) is a highly potent, selective, nonsteroidal, non-bile acid farnesoid X receptor agonist for the treatment of nonalcoholic steatohepatitis. Its absorption, metabolism, and excretion were studied after a 1-mg oral dose of [14C]tropifexor was given to four healthy male subjects. Mass balance was achieved with ∼94% of the administered dose recovered in excreta through a 312-hour collection period. Fecal excretion of tropifexor-related radioactivity played a major role (∼65% of the total dose). Tropifexor reached a maximum blood concentration (Cmax) of 33.5 ng/ml with a median time to reach Cmax of 4 hours and was eliminated with a plasma elimination half-life of 13.5 hours. Unchanged tropifexor was the principal drug-related component found in plasma (∼92% of total radioactivity). Two minor oxidative metabolites, M11.6 and M22.4, were observed in circulation. Tropifexor was eliminated predominantly via metabolism with >68% of the dose recovered as metabolites in excreta. Oxidative metabolism appeared to be the major clearance pathway of tropifexor. Metabolites containing multiple oxidative modifications and combined oxidation and glucuronidation were also observed in human excreta. The involvement of direct glucuronidation could not be ruled out based on previous in vitro and nonclinical in vivo studies indicating its contribution to tropifexor clearance. The relative contribution of the oxidation and glucuronidation pathways appeared to be dose-dependent upon further in vitro investigation. Because of these complexities and the instability of glucuronide metabolites in the gastrointestinal tract, the contribution of glucuronidation remained undefined in this study. SIGNIFICANCE STATEMENT: Tropifexor was found to be primarily cleared from the human body via oxidative metabolism. In vitro metabolism experiments revealed that the relative contribution of oxidation and glucuronidation was concentration-dependent, with glucuronidation as the predominant pathway at higher concentrations and the oxidative process becoming more important at lower concentrations near clinical exposure range. The body of work demonstrated the importance of carefully designed in vivo and in vitro experiments for better understanding of disposition processes during drug development.


Assuntos
Benzotiazóis/farmacocinética , Isoxazóis/farmacocinética , Administração Oral , Adolescente , Adulto , Benzotiazóis/administração & dosagem , Absorção Gastrointestinal , Voluntários Saudáveis , Humanos , Isoxazóis/administração & dosagem , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Adulto Jovem
4.
Drug Metab Dispos ; 48(10): 873-885, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32665418

RESUMO

Capmatinib (INC280), a highly selective and potent inhibitor of the MET receptor tyrosine kinase, has demonstrated clinically meaningful efficacy and a manageable safety profile in patients with advanced non-small-cell lung cancer harboring MET exon 14-skipping mutations. We investigated the absorption, distribution, metabolism, and excretion of capmatinib in six healthy male volunteers after a single peroral dose of 600 mg 14C-labeled capmatinib. The mass balance, blood and plasma radioactivity, and plasma capmatinib concentrations were determined along with metabolite profiles in plasma, urine, and feces. The metabolite structures were elucidated using mass spectrometry and comparing with reference compounds. The parent compound accounted for most of the radioactivity in plasma (42.9% ± 2.9%). The extent of oral absorption was estimated to be 49.6%; the Cmax of capmatinib in plasma was reached at 2 hours (median time to reach Cmax). The apparent mean elimination half-life of capmatinib in plasma was 7.84 hours. Apparent distribution volume of capmatinib during the terminal phase was moderate-to-high (geometric mean 473 l). Metabolic reactions involved lactam formation, hydroxylation, N-dealkylation, formation of a carboxylic acid, hydrogenation, N-oxygenation, glucuronidation, and combinations thereof. M16, the most abundant metabolite in plasma, urine, and feces was formed by lactam formation. Absorbed capmatinib was eliminated mainly by metabolism and subsequent biliary/fecal and renal excretion. Excretion of radioactivity was complete after 7 days. CYP phenotyping demonstrated that CYP3A was the major cytochrome P450 enzyme subfamily involved in hepatic microsomal metabolism, and in vitro studies in hepatic cytosol indicated that M16 formation was mainly catalyzed by aldehyde oxidase. SIGNIFICANCE STATEMENT: The absorption, distribution, metabolism, and excretion of capmatinib revealed that capmatinib had substantial systemic availability after oral administration. It was also extensively metabolized and largely distributed to the peripheral tissue. Mean elimination half-life was 7.84 hours. The most abundant metabolite, M16, was formed by imidazo-triazinone formation catalyzed by cytosolic aldehyde oxidase. Correlation analysis, specific inhibition, and recombinant enzymes phenotyping demonstrated that CYP3A is the major enzyme subfamily involved in the hepatic microsomal metabolism of [14C]capmatinib.


Assuntos
Aldeído Oxidase/metabolismo , Benzamidas/farmacocinética , Citocromo P-450 CYP3A/metabolismo , Imidazóis/farmacocinética , Inibidores de Proteínas Quinases/farmacocinética , Triazinas/farmacocinética , Administração Oral , Benzamidas/administração & dosagem , Benzamidas/efeitos adversos , Biotransformação , Citosol/metabolismo , Voluntários Saudáveis , Hepatócitos/citologia , Hepatócitos/metabolismo , Humanos , Imidazóis/administração & dosagem , Imidazóis/efeitos adversos , Absorção Intestinal , Masculino , Microssomos Hepáticos , Pessoa de Meia-Idade , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/efeitos adversos , Distribuição Tecidual , Triazinas/administração & dosagem , Triazinas/efeitos adversos
5.
Phys Rev Lett ; 124(21): 213001, 2020 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-32530687

RESUMO

Molecular helium represents a benchmark system for testing ab initio calculations on few-electron molecules. We report on the determination of the adiabatic ionization energy of the a ^{3}Σ_{u}^{+} state of He_{2}, corresponding to the energy interval between the a ^{3}Σ_{u}^{+} (v^{''}=0, N^{''}=1) state of He_{2} and the X^{+} ^{2}Σ_{u}^{+} (v^{+}=0, N^{+}=1) state of He_{2}^{+}, and of the lowest rotational interval of He_{2}^{+}. These measurements rely on the excitation of metastable He_{2} molecules to high Rydberg states using frequency-comb-calibrated continuous-wave UV radiation in a counterpropagating laser-beam setup. The observed Rydberg states were extrapolated to their series limit using multichannel quantum-defect theory. The ionization energy of He_{2} (a ^{3}Σ_{u}^{+}) and the lowest rotational interval of He_{2}^{+} (X^{+} ^{2}Σ_{u}^{+}) are 34 301.207 002(23)±0.000 037_{syst} cm^{-1} and 70.937 589(23)±0.000 060_{syst} cm^{-1}, respectively.

6.
Pharmacol Res Perspect ; 8(3): e00599, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32524755

RESUMO

Ribociclib (LEE011, Kisqali ®) is a highly selective small molecule inhibitor of cyclin-dependent kinases 4 and 6 (CDK4/6), which has been approved for the treatment of advanced or metastatic breast cancer. A human ADME study was conducted in healthy male volunteers following a single oral dose of 600 mg [14 C]-ribociclib. Mass balance, blood and plasma radioactivity, and plasma ribociclib concentrations were measured. Metabolite profiling and identification was conducted in plasma, urine, and feces. An assessment integrating the human ADME results with relevant in vitro and in vivo non-clinical data was conducted to provide an estimate of the relative contributions of various clearance pathways of the compound. Ribociclib is moderately to highly absorbed across species (approx. 59% in human), and is extensively metabolized in vivo, predominantly by oxidative pathways mediated by CYP3A4 (ultimately forming N-demethylated metabolite M4) and, to a lesser extent, by FMO3 (N-hydroxylated metabolite M13). It is extensively distributed in rats, based on QWBA data, and is eliminated rapidly from most tissues with the exception of melanin-containing structures. Ribociclib passed the placental barrier in rats and rabbits and into milk of lactating rats. In human, 69.1% and 22.6% of the radiolabeled dose were excreted in feces and urine, respectively, with 17.3% and 6.75% of the 14 C dose attributable to ribociclib, respectively. The remainder was attributed to numerous metabolites. Taking into account all available data, ribociclib is estimated to be eliminated by hepatic metabolism (approx. 84% of total), renal excretion (7%), intestinal excretion (8%), and biliary elimination (1%).


Assuntos
Aminopiridinas/farmacocinética , Antineoplásicos/farmacocinética , Inibidores de Proteínas Quinases/farmacocinética , Purinas/farmacocinética , Administração Oral , Aminopiridinas/administração & dosagem , Animais , Antineoplásicos/administração & dosagem , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Cães , Feminino , Humanos , Lactação , Masculino , Placenta/metabolismo , Gravidez , Inibidores de Proteínas Quinases/administração & dosagem , Purinas/administração & dosagem , Coelhos , Ratos , Especificidade da Espécie , Distribuição Tecidual
7.
Phys Chem Chem Phys ; 21(35): 18850-18865, 2019 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-31432055

RESUMO

Proton detected solid-state NMR under fast magic-angle-spinning (MAS) conditions is currently redefining the applications of solid-state NMR, in particular in structural biology. Understanding the contributions to the spectral linewidth is thereby of paramount importance. When disregarding the sample-dependent inhomogeneous contributions, the NMR proton linewidth is defined by homogeneous broadening, which has incoherent and coherent contributions. Understanding and disentangling these different contributions in multi-spin systems like proteins is still an open issue. The coherent contribution is mainly caused by the dipolar interaction under MAS and is determined by the molecular structure and the proton chemical shifts. Numerical simulation approaches based on numerically exact direct integration of the Liouville-von Neumann equation can give valuable information about the lineshape, but are limited to small spin systems (<12 spins). We present an alternative simulation method for the coherent contributions based on the rapid and partially analytic calculation of the second moments of large spin systems. We first validate the method on a simple system by predicting the 19F linewidth in CaF2 under MAS. We compare simulation results to experimental data for microcrystalline ubiquitin (deuterated 100% back-exchanged at 110 kHz and fully-protonated at 125 kHz). Our results quantitatively explain the observed linewidth per-residue basis for the vast majority of residues.


Assuntos
Simulação por Computador , Modelos Químicos , Proteínas/química , Ressonância Magnética Nuclear Biomolecular , Prótons
8.
Pulm Pharmacol Ther ; 57: 101809, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31195091

RESUMO

Fevipiprant, a prostaglandin D2 receptor 2 antagonist, is in clinical development as a treatment for asthma. The goal of this study was to assess the potential of fevipiprant to cause drug-drug interactions (DDI) as a perpetrator, that is, by altering the pharmacokinetics (PK) of co-medications. In vitro drug interaction studies of clinically relevant drug metabolizing enzymes and transporters were conducted for fevipiprant and its acyl glucuronide (AG) metabolite. Comparison of Ki values with unbound systemic or portal vein steady-state plasma exposure of fevipiprant and its AG metabolite revealed the potential for inhibition of organic anion transporting polypeptide 1B1 (OATP1B1) transporters (R-value of 5.99), while other targets including cytochrome P450 enzymes were not, or only marginally, inhibited. Consequently, an open-label, two-part, two-period, single-sequence clinical study assessed the effect of fevipiprant 450 mg QD on the pharmacokinetics of simvastatin 20 mg and rosuvastatin 20 mg, two statins with different dependency in OATP1B1-mediated hepatic uptake, in healthy adult volunteers. The study also assessed the pharmacogenetics of the SLCO1B1 gene, which encodes OATP1B1. Clinically, fevipiprant 450 mg QD showed a low potential for interaction and increased the peak concentrations of simvastatin acid and rosuvastatin by 2.23- and 1.87-fold, respectively, with little or no impact on total exposure. Genotype analysis confirmed that SLCO1B1 genotype influences statin pharmacokinetics to a similar extent either with or without fevipiprant co-administration. In summary, fevipiprant at 450 mg QD has only minor liabilities as a perpetrator for DDI.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/farmacocinética , Ácidos Indolacéticos/farmacologia , Transportador 1 de Ânion Orgânico Específico do Fígado/genética , Piridinas/farmacologia , Rosuvastatina Cálcica/farmacocinética , Sinvastatina/farmacocinética , Adulto , Interações Medicamentosas , Feminino , Genótipo , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Transportadores de Ânions Orgânicos , Farmacogenética , Receptores Imunológicos/antagonistas & inibidores , Receptores de Prostaglandina/antagonistas & inibidores
9.
Eur J Pharm Sci ; 132: 132-141, 2019 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-30857914

RESUMO

The generation of reliable kinetic parameters to describe P-glycoprotein (P-gp) activity is essential for predicting the impact of efflux transport on gastrointestinal drug absorption. The compound-specific selection of in vitro assay designs and ensuing data analysis methods is explored in this manuscript. We measured transcellular permeability and cellular uptake of five P-gp substrates in Caco-2 and LLC-PK1 MDR1 cells. Kinetic parameters of P-gp-mediated efflux transport (Km, Vmax) were derived from conventional and mechanistic compartmental models. The estimated apparent Km values based on medium concentrations in the conventional permeability model indicated significant differences between the cell lines. The respective intrinsic Km values based on unbound intracellular concentrations in the mechanistic compartmental models were significantly lower and comparable between cell lines and assay formats. Non-specific binding or lysosomal trapping were shown to cause discrepancies in the kinetic parameters obtained from different assay formats. A guidance for the selection of in vitro assays and kinetic assessment methods is proposed in line with the Biopharmaceutics Drug Disposition Classification System (BDDCS). The recommendations are expected to aid the acquisition of robust and reproducible kinetic parameters of P-gp-mediated efflux transport.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Biofarmácia/métodos , Modelos Biológicos , Preparações Farmacêuticas/metabolismo , Animais , Células CACO-2 , Técnicas de Cultura de Células , Permeabilidade da Membrana Celular , Relação Dose-Resposta a Droga , Guias como Assunto , Humanos , Cinética , Células LLC-PK1 , Preparações Farmacêuticas/administração & dosagem , Especificidade por Substrato , Suínos
10.
Xenobiotica ; 49(5): 503-512, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-29694257

RESUMO

The expression of flavin-containing monooxygenase (FMO) varies extensively between human and commonly used preclinical species such as rat and mouse. The aim of this study was to investigate the pulmonary FMO activity in rat using benzydamine. Furthermore, the contribution of rat lung to the clearance of benzydamine was investigated using an in vivo pulmonary extraction model. Benzydamine N-oxygenation was observed in lung microsomes and lung slices. Thermal inactivation of FMO and CYP inhibition suggested that rat pulmonary N-oxygenation is predominantly FMO mediated while any contribution from CYPs is negligible. The predicted lung clearance (CLlung) estimated from microsomes and slices was 16 ± 0.6 and 2.1 ± 0.3 mL/min/kg, respectively. The results from in vivo pulmonary extraction indicated no pulmonary extraction following intravenous and intra-arterial dosing to rats. Interestingly, the predicted CLlung using rat lung microsomes corresponded to approximately 35% of rat CLliver suggesting that the lung makes a smaller contribution to the whole body clearance of benzydamine. Although benzydamine clearance in rat appears to be predominantly mediated by hepatic metabolism, the data suggest that the lung may also make a smaller contribution to its whole body clearance.


Assuntos
Benzidamina/farmacocinética , Pulmão/enzimologia , Microssomos/enzimologia , Oxigenases de Função Mista/metabolismo , Animais , Benzidamina/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley
11.
Drug Metab Lett ; 13(1): 53-63, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30345935

RESUMO

BACKGROUND: Although the liver is the primary organ of drug metabolism, the lungs also contain drug-metabolizing enzymes and may, therefore, contribute to the elimination of drugs. In this investigation, the Precision-cut Lung Slice (PCLS) technique was standardized with the aims of characterizing and comparing rat and human pulmonary drug metabolizing activity. METHOD: Due to the limited availability of human lung tissue, standardization of the PCLS method was performed with rat lung tissue. Pulmonary enzymatic activity was found to vary significantly with rat age and rat strain. The Dynamic Organ Culture (DOC) system was superior to well-plates for tissue incubations, while oxygen supply appeared to have a limited impact within the 4h incubation period used here. RESULTS: The metabolism of a range of phase I and phase II probe substrates was assessed in rat and human lung preparations. Cytochrome P450 (CYP) activity was relatively low in both species, whereas phase II activity appeared to be more significant. CONCLUSION: PCLS is a promising tool for the investigation of pulmonary drug metabolism. The data indicates that pulmonary CYP activity is relatively low and that there are significant differences in enzyme activity between rat and human lung.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Técnicas de Preparação Histocitológica/métodos , Pulmão/enzimologia , Farmacologia Clínica/métodos , Animais , Feminino , Humanos , Masculino , Modelos Animais , Técnicas de Cultura de Órgãos , Ratos , Especificidade da Espécie
12.
Drug Metab Dispos ; 46(11): 1670-1683, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30111625

RESUMO

The eye is a complex organ with a series of anatomic barriers that provide protection from physical and chemical injury while maintaining homeostasis and function. The physiology of the eye is multifaceted, with dynamic flows and clearance mechanisms. This review highlights that in vitro ocular transport and metabolism models are confined by the availability of clinically relevant absorption, distribution, metabolism, and excretion (ADME) data. In vitro ocular transport models used for pharmacology and toxicity poorly predict ocular exposure. Although ocular cell lines cannot replicate in vivo conditions, these models can help rank-order new chemical entities in discovery. Historic ocular metabolism of small molecules was assumed to be inconsequential or assessed using authentic standards. While various in vitro models have been cited, no single system is perfect, and many must be used in combination. Several studies document the use of laboratory animals for the prediction of ocular pharmacokinetics in humans. This review focuses on the use of human-relevant and human-derived models which can be utilized in discovery and development to understand ocular disposition of new chemical entities. The benefits and caveats of each model are discussed. Furthermore, ADME case studies are summarized retrospectively and capture the ADME data collected for health authorities in the absence of definitive guidelines. Finally, we discuss the novel technologies and a hypothesis-driven ocular drug classification system to provide a holistic perspective on the ADME properties of drugs administered by the ocular route.


Assuntos
Olho/efeitos dos fármacos , Olho/metabolismo , Preparações Farmacêuticas/administração & dosagem , Preparações Farmacêuticas/metabolismo , Bibliotecas de Moléculas Pequenas/administração & dosagem , Bibliotecas de Moléculas Pequenas/metabolismo , Administração Oftálmica , Animais , Descoberta de Drogas/métodos , Humanos , Bibliotecas de Moléculas Pequenas/efeitos adversos
13.
Drug Metab Dispos ; 46(7): 1001-1013, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29735753

RESUMO

Siponimod, a next-generation selective sphingosine-1-phosphate receptor modulator, is currently being investigated for the treatment of secondary progressive multiple sclerosis. We investigated the absorption, distribution, metabolism, and excretion (ADME) of a single 10-mg oral dose of [14C]siponimod in four healthy men. Mass balance, blood and plasma radioactivity, and plasma siponimod concentrations were measured. Metabolite profiles were determined in plasma, urine, and feces. Metabolite structures were elucidated using mass spectrometry and comparison with reference compounds. Unchanged siponimod accounted for 57% of the total plasma radioactivity (area under the concentration-time curve), indicating substantial exposure to metabolites. Siponimod showed medium to slow absorption (median Tmax: 4 hours) and moderate distribution (Vz/F: 291 l). Siponimod was mainly cleared through biotransformation, predominantly by oxidative metabolism. The mean apparent elimination half-life of siponimod in plasma was 56.6 hours. Siponimod was excreted mostly in feces in the form of oxidative metabolites. The excretion of radioactivity was close to complete after 13 days. Based on the metabolite patterns, a phase II metabolite (M3) formed by glucuronidation of hydroxylated siponimod was the main circulating metabolite in plasma. However, in subsequent mouse ADME and clinical pharmacokinetic studies, a long-lived nonpolar metabolite (M17, cholesterol ester of siponimod) was identified as the most prominent systemic metabolite. We further conducted in vitro experiments to investigate the enzymes responsible for the oxidative metabolism of siponimod. The selective inhibitor and recombinant enzyme results identified cytochrome P450 2C9 (CYP2C9) as the predominant contributor to the human liver microsomal biotransformation of siponimod, with minor contributions from CYP3A4 and other cytochrome P450 enzymes.


Assuntos
Azetidinas/metabolismo , Compostos de Benzil/metabolismo , Citocromo P-450 CYP2C9/metabolismo , Citocromo P-450 CYP3A/metabolismo , Receptores de Lisoesfingolipídeo/agonistas , Adolescente , Adulto , Animais , Biotransformação/fisiologia , Fezes , Meia-Vida , Voluntários Saudáveis , Humanos , Masculino , Camundongos , Microssomos Hepáticos/metabolismo , Pessoa de Meia-Idade , Oxirredução , Estresse Oxidativo/fisiologia , Adulto Jovem
14.
Bioanalysis ; 10(5): 321-339, 2018 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-29451392

RESUMO

AIM: Although regulatory guidances require human metabolism information of drug candidates early in the development process, the human mass balance study (or hADME study), is performed relatively late. hADME studies typically involve the administration of a 14C-radiolabelled drug where biological samples are measured by conventional scintillation counting analysis. Another approach is the administration of therapeutic doses containing a 14C-microtracer followed by accelerator mass spectrometry (AMS) analysis, enabling hADME studies completion much earlier. Consequently, there is an opportunity to change the current drug development paradigm. MATERIALS & METHODS: To evaluate the applicability of the MICADAS-cAMS method, we successfully performed: the validation of MICADAS-cAMS for radioactivity quantification in biomatrices and, a rat ADME study, where the conventional methodology was assessed against a microtracer MICADAS-cAMS approach. RESULTS & DISCUSSION: Combustion AMS (cAMS) technology is applicable to microtracer studies. A favorable opinion from EMA to complete the hADME in a Phase I setting was received, opening the possibilities to change drug development.


Assuntos
Radioisótopos de Carbono/sangue , Radioisótopos de Carbono/farmacocinética , Radioisótopos de Carbono/urina , Piridinas/sangue , Piridinas/farmacocinética , Piridinas/urina , Pirimidinas/sangue , Pirimidinas/farmacocinética , Pirimidinas/urina , Animais , Radioisótopos de Carbono/administração & dosagem , Descoberta de Drogas , Fezes/química , Humanos , Masculino , Espectrometria de Massas , Metaboloma , Piridinas/administração & dosagem , Pirimidinas/administração & dosagem , Traçadores Radioativos , Ratos , Ratos Wistar , Contagem de Cintilação , Sensibilidade e Especificidade
15.
Xenobiotica ; 48(8): 793-803, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28879796

RESUMO

1. AFQ056 phenotyping results indicate that CYP1A1 is responsible for the formation of the oxidative metabolite, M3. In line with the predominant assumption that CYP1A1 is mainly expressed in extrahepatic tissues, only traces of M3 were detected in hepatic systems. The aim of this study was to investigate the pulmonary CYP1A1 mediated metabolism of AFQ056 in rat. 2. Western blot analysis confirmed that CYP1A1 is expressed in rat lung albeit at low levels. M3 formation was clearly observed in recombinant rat CYP1A1, lung microsomes and lung tissue slices and was strongly inhibited by ketoconazole in the incubations. As CYP3A4 and CYP2C9 metabolites were only observed at trace levels, we concluded that the reduced M3 formation was due to CYP1A1 inhibition. 3. AFQ056 lung clearance (CLlung) as estimated from in vitro data was predicted to be negligible (<1% pulmonary blood flow). This was confirmed by in vivo experiments where intravenous and intra-arterial dosing to rats failed to show significant pulmonary extraction. 4. While rat lung may make a contribution to the formation of M3, it is unlikely to be the only organ involved in this process and further experiments are required to investigate the potential metabolic elimination routes for AFQ056.


Assuntos
Citocromo P-450 CYP1A1/metabolismo , Indóis/farmacocinética , Pulmão/enzimologia , Animais , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Indóis/farmacologia , Pulmão/irrigação sanguínea , Masculino , Ratos , Ratos Sprague-Dawley
16.
Eur J Clin Pharmacol ; 74(4): 455-464, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29273968

RESUMO

PURPOSE: The purpose of the study is to investigate the enzyme(s) responsible for siponimod metabolism and to predict the inhibitory effects of fluconazole as well as the impact of cytochrome P450 (CYP) 2C9 genetic polymorphism on siponimod pharmacokinetics (PK) and metabolism. METHODS: In vitro metabolism studies were conducted using human liver microsomes (HLM), and enzyme phenotyping was assessed using a correlation analysis method. SimCYP, a physiologically based PK model, was developed and used to predict the effects of fluconazole and CYP2C9 genetic polymorphism on siponimod metabolism. Primary PK parameters were generated using the SimCYP and WinNonlin software. RESULTS: Correlation analysis suggested that CYP2C9 is the main enzyme responsible for siponimod metabolism in humans. Compared with the CYP2C9*1/*1 genotype, HLM incubations from CYP2C9*3/*3 and CYP2C9*2/*2 donors showed ~ 10- and 3-fold decrease in siponimod metabolism, respectively. Simulations of enzyme contribution predicted that in the CYP2C9*1/*1 genotype, CYP2C9 is predominantly responsible for siponimod metabolism (~ 81%), whereas in the CYP2C9*3/*3 genotype, its contribution is reduced to 11%. The predicted exposure increase of siponimod with fluconazole 200 mg was 2.0-2.4-fold for CYP2C9*1/*1 genotype. In context of single dosing, the predicted mean area under the curve (AUC) is 2.7-, 3.0- and 4.5-fold higher in the CYP2C9*2/*2, CYP2C9*2/*3 and CYP2C9*3/*3 genotypes, respectively, compared with the CYP2C9*1/*1 genotype. CONCLUSION: .Enzyme phenotyping with correlation analysis confirmed the predominant role of CYP2C9 in the biotransformation of siponimod and demonstrated the functional consequence of CYP2C9 genetic polymorphism on siponimod metabolism. Simulation of fluconazole inhibition closely predicted a 2-fold AUC change (ratio within ~ 20% deviation) to the observed value. In silico simulation predicted a significant reduction in siponimod clearance in the CYP2C9*2/*2 and CYP2C9*3/*3 genotypes based on the in vitro metabolism data; the predicted exposure was close (within 30%) to the observed results for the CYP2C9*2/*3 and CYP2C9*3/*3 genotypes.


Assuntos
Azetidinas/farmacocinética , Compostos de Benzil/farmacocinética , Simulação por Computador , Inibidores do Citocromo P-450 CYP2C9/farmacologia , Citocromo P-450 CYP2C9/genética , Fluconazol/farmacologia , Microssomos Hepáticos/enzimologia , Modelos Biológicos , Variantes Farmacogenômicos , Polimorfismo Genético , Azetidinas/metabolismo , Compostos de Benzil/metabolismo , Citocromo P-450 CYP2C9/metabolismo , Interações Medicamentosas , Genótipo , Humanos , Farmacogenética , Fenótipo , Software
17.
Drug Metab Dispos ; 45(8): 900-907, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28566285

RESUMO

The human mass balance study is the definitive study for the assessment of absorption, distribution, metabolism, and excretion (ADME) properties of a new chemical entity in humans. Traditionally this has been carried out by the administration of radiolabeled drug substances, typically 14C or occasionally 3H, as detection methods for these isotopes allow the absolute quantification of drug-related material (DRM) in blood, plasma, and excreta. Coupled with the use of analytical techniques such as liquid chromatography-mass spectrometry, a picture of the metabolic fate of a compound can be elucidated. In this study, we demonstrate the capabilities of 19F nuclear magnetic resonance (NMR) spectroscopy, applied as an alternative to radiolabeling, for the determination of mass balance and for metabolite profiling of an orally administered fluorinated drug. To demonstrate the capabilities of NMR, the study was conducted on remaining samples from a 14C human mass balance study conducted on Alpelisib (BYL719), a compound in late stage development at Novartis for the treatment of solid tumors. Quantitative 14C data were used to cross-validate the data obtained by NMR. The data show that, using 19F NMR, comparable data can be obtained for key human ADME endpoints including mass balance, total DRM determination in plasma and metabolite profiling and identification in plasma and excreta. Potential scenarios where NMR could be employed as an alternative to radiolabeling for the conduct of an early human ADME study are discussed.


Assuntos
Radioisótopos de Carbono/química , Flúor/química , Imageamento por Ressonância Magnética/métodos , Espectroscopia de Ressonância Magnética/métodos , Tiazóis/química , Tiazóis/metabolismo , Cromatografia Líquida/métodos , Humanos , Masculino , Espectrometria de Massas/métodos
18.
Drug Metab Dispos ; 45(7): 817-825, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28442499

RESUMO

Fevipiprant is a novel oral prostaglandin D2 receptor 2 (DP2; also known as CRTh2) antagonist, which is currently in development for the treatment of severe asthma and atopic dermatitis. We investigated the absorption, distribution, metabolism, and excretion properties of fevipiprant in healthy subjects after a single 200-mg oral dose of [14C]-radiolabeled fevipiprant. Fevipiprant and metabolites were analyzed by liquid chromatography coupled to tandem mass spectrometry and radioactivity measurements, and mechanistic in vitro studies were performed to investigate clearance pathways and covalent plasma protein binding. Biotransformation of fevipiprant involved predominantly an inactive acyl glucuronide (AG) metabolite, which was detected in plasma and excreta, representing 28% of excreted drug-related material. The AG metabolite was found to covalently bind to human plasma proteins, likely albumin; however, in vitro covalent binding to liver protein was negligible. Excretion was predominantly as unchanged fevipiprant in urine and feces, indicating clearance by renal and possibly biliary excretion. Fevipiprant was found to be a substrate of transporters organic anion transporter 3 (OAT3; renal uptake), multidrug resistance gene 1 (MDR1; possible biliary excretion), and organic anion-transporting polypeptide 1B3 (OATP1B3; hepatic uptake). Elimination of fevipiprant occurs via glucuronidation by several uridine 5'-diphospho glucuronosyltransferase (UGT) enzymes as well as direct excretion. These parallel elimination pathways result in a low risk of major drug-drug interactions or pharmacogenetic/ethnic variability for this compound.


Assuntos
Hepatócitos/metabolismo , Ácidos Indolacéticos/farmacocinética , Microssomos Hepáticos/metabolismo , Piridinas/farmacocinética , Receptores Imunológicos/antagonistas & inibidores , Receptores de Prostaglandina/antagonistas & inibidores , Administração Oral , Adolescente , Adulto , Biotransformação , Fezes/química , Voluntários Saudáveis , Humanos , Técnicas In Vitro , Ácidos Indolacéticos/sangue , Ácidos Indolacéticos/urina , Masculino , Taxa de Depuração Metabólica , Metaboloma , Pessoa de Meia-Idade , Ligação Proteica , Piridinas/sangue , Piridinas/urina , Eliminação Renal , Distribuição Tecidual , Adulto Jovem
19.
J Pharm Sci ; 106(9): 2805-2814, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28385545

RESUMO

Unbound intrahepatic drug concentrations determine the interaction potential with intracellular targets related to toxicity, pharmacokinetics, or pharmacodynamics. Recently, the unbound liver-to-blood partition coefficient (Kpuu) based on the Extended Clearance Model (ECM) has been developed providing indirect estimates of unbound intrahepatic drug concentrations. This study aimed to determine Kpuu for 18 diverse drug compounds by 3 alternative in vitro methods and to compare the outcome with the ECM approach. Kpuu was calculated from independent measurements of hepatocellular drug accumulation (Kp) and unbound fraction in hepatocytes (fuhep) either assessed from steady-state accumulation at 4°C (temperature method), using equilibrium dialysis (homogenization method), or empirically from logD7.4 (logD7.4 method). Deviations to ECM-based Kpuu data were closely linked to the absence of intrinsic clearance processes (metabolism, biliary secretion) in the investigated methods. Differences in fuhep additionally contributed to deviations in Kpuu. The homogenization method generally provided lowest fuhep values, especially for compounds with high molecular weight or low logD7.4. Kpuu values of compounds with low intrinsic clearance correlated well between the ECM and temperature methods independent of physicochemical properties. Therefore, only the ECM provides an integrated quantitative determination of hepatic Kpuu. Temperature and homogenization methods, however, represent useful alternatives if compound properties are appropriately considered.


Assuntos
Hepatócitos/metabolismo , Preparações Farmacêuticas/sangue , Preparações Farmacêuticas/metabolismo , Células Cultivadas , Humanos , Fígado/metabolismo , Lisossomos/metabolismo , Taxa de Depuração Metabólica , Modelos Biológicos , Temperatura
20.
Drug Metab Dispos ; 45(5): 523-531, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28254950

RESUMO

Inhibition of the bile salt export pump (BSEP) has been recognized as a key factor in the development of drug-induced cholestasis (DIC). The risk of DIC in humans has been previously assessed using in vitro BSEP inhibition data (IC50) and unbound systemic drug exposure under assumption of the "free drug hypothesis." This concept, however, is unlikely valid, as unbound intrahepatic drug concentrations are affected by active transport and metabolism. To investigate this hypothesis, we experimentally determined the in vitro liver-to-blood partition coefficients (Kpuu) for 18 drug compounds using the hepatic extended clearance model (ECM). In vitro-in vivo translatability of Kpuu values was verified for a subset of compounds in rat. Consequently, unbound intrahepatic concentrations were calculated from clinical exposure (systemic and hepatic inlet) and measured Kpuu data. Using these values, corresponding safety margins against BSEP IC50 values were determined and compared with the clinical incidence of DIC. Depending on the ECM class of a drug, in vitro Kpuu values deviated up to 14-fold from unity, and unbound intrahepatic concentrations were affected accordingly. The use of in vitro Kpuu-based safety margins allowed separation of clinical cholestasis frequency into three classes (no cholestasis, cholestasis in ≤2%, and cholestasis in >2% of subjects) for 17 out of 18 compounds. This assessment was significantly superior compared with using unbound extracellular concentrations as a surrogate for intrahepatic concentrations. Furthermore, the assessment of Kpuu according to ECM provides useful guidance for the quantitative evaluation of genetic and physiologic risk factors for the development of cholestasis.


Assuntos
Colestase/induzido quimicamente , Colestase/metabolismo , Fígado/metabolismo , Modelos Biológicos , Preparações Farmacêuticas/metabolismo , Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Colestase/sangue , Humanos , Preparações Farmacêuticas/sangue , Farmacocinética , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...