Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Pestic Biochem Physiol ; 191: 105363, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36963934

RESUMO

Spodoptera frugiperda is an economically important agricultural pest and poses a serious threat to food security globally. Its management is gravely challenged by its high polyphagous nature, strong migratory ability, and massive fecundity. Chlorantraniliprole (CHL) is widely utilized in controlling S. frugiperda, its intensive application and over-reliance pose adverse health risks, development of resistance, toxicity to beneficial insects, natural enemies, and environmental contamination. To address S. frugiperda resistance to CHL and its inherent challenges, this study explores the synergistic effects of camptothecin (CPT) with CHL in its management. The binary mixed adversely induced the larvae weight and mortality when compared to single-treated. CHL + CPT (1:20 mg/L) had the highest larvae mortality of (73.80 %) with a high antagonistic factor (0.90), while (1:10 mg/L) with (66.10%) mortality exhibited a high synergistic factor (1.43). Further, CHL + CPT (1:10 mg/L) considerably altered the midgut epithelial cell, peritrophic membrane, microvilli, basement membrane, and regenerative cells. For biochemical analysis, CHL + CPT (1:10 mg/L) significantly decreased glutathione-S-transferase (1-chloro-2,4-dinitrobenzene CDNB) and cytochrome P450 (7-ethoxycoumarin O-deethylation) activities in the midgut in a dose and time dependent manner. Based on RNA-Seq analysis, a total of 4,373 differentially expressed genes (DEGs) were identified from the three treatments. CPT vs CK (Control) had 1694 (968 up-, 726 down-regulated), CHL vs CK with 1771 (978 up-, 793 down-regulated), and CHL + CPT vs CK had 908 (394 up-, 514 down-regulated) DEGs. The enrichment analysis disclosed significant pathways such as metabolism of xenobiotics by cytochrome P450, glutathione metabolism, TOLL and IMD (Immune Deficiency) signaling pathway, longevity regulating pathway. This study provides basis to expatiate on the molecular toxicological mechanism of CHL + CPT in management of fall armyworm.


Assuntos
Camptotecina , Inseticidas , Larva , Spodoptera , Sinergismo Farmacológico , Spodoptera/anatomia & histologia , Spodoptera/efeitos dos fármacos , Spodoptera/crescimento & desenvolvimento , Spodoptera/fisiologia , Camptotecina/administração & dosagem , Camptotecina/farmacologia , Camptotecina/toxicidade , Larva/anatomia & histologia , Larva/efeitos dos fármacos , Segurança Alimentar , Inseticidas/administração & dosagem , Inseticidas/farmacologia , Inseticidas/toxicidade , Animais , Metabolismo Secundário , Peso Corporal/efeitos dos fármacos , Glutationa Transferase/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Inibidores das Enzimas do Citocromo P-450/farmacologia , Inibidores das Enzimas do Citocromo P-450/toxicidade , Perfilação da Expressão Gênica , Ontologia Genética , RNA-Seq , Reprodutibilidade dos Testes
2.
Toxicol Appl Pharmacol ; 434: 115797, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34780725

RESUMO

Dabrafenib is a BRAF inhibitor used in combination treatment of malignant melanoma and non-small cell lung carcinoma. In this study, we aimed to characterize its interactions with cytochrome P450 (CYP) isoenzymes and ATP-binding cassette (ABC) efflux transporters that have critical impact on the pharmacokinetics of drugs and play a role in drug resistance development. Using accumulation assays, we showed that dabrafenib inhibited ABCG2 and, less potently, ABCB1 transporter. We also confirmed dabrafenib as a CYP2C8, CYP2C9, CYP3A4, and CYP3A5 inhibitor. Importantly, inhibition of ABCG2 and CYP3A4 by dabrafenib led to the potentiation of cytotoxic effects of mitoxantrone and docetaxel toward respective resistant cell lines in drug combination studies. On the contrary, the synergistic effect was not consistently observed in ABCB1-expressing models. We further demonstrated that mRNA levels of ABCB1, ABCG2, ABCC1, and CYP3A4 were increased after 24 h and 48 h exposure to dabrafenib. Overall, our data confirm dabrafenib as a drug frequently and potently interacting with ABC transporters and CYP isoenzymes. This feature should be addressed with caution when administering dabrafenib to patients with polypharmacy but also could be utilized advantageously when designing new dabrafenib-containing drug combinations to improve the therapeutic outcome in drug-resistant cancer.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Antineoplásicos/farmacologia , Inibidores das Enzimas do Citocromo P-450/farmacologia , Daunorrubicina/farmacologia , Imidazóis/farmacocinética , Mitoxantrona/farmacologia , Oximas/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Daunorrubicina/administração & dosagem , Cães , Quimioterapia Combinada , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Imidazóis/administração & dosagem , Mitoxantrona/administração & dosagem , Oximas/administração & dosagem , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
3.
Chem Biol Interact ; 352: 109775, 2022 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-34910929

RESUMO

Vicagrel, an antiplatelet drug candidate targeting platelet P2Y12 receptor and has finished its phase II clinical trial. The inhibition of six major cytochrome P450 enzymes (P450) (CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP3A4) and six UDP-glucuronosyltransferases (UGT) (UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9, and UGT2B7) by vicagrel was evaluated using pooled human liver microsomes and specific probe substrates. Physiology-based pharmacokinetic (PBPK) simulation was further applied to predict the in vivo drug-drug interaction (DDI) potential between vicagrel and bupropion as well as S-mephenytoin. The results suggested that vicagrel inhibited CYP2B6 and CYP2C19 potently with apparent IC50 values of 1.6 and 2.0 µM, respectively. In terms of mode of reversible inhibition, vicagrel exhibited mixed-type inhibition of CYP2B6-catalyzed bupropion hydroxylation and noncompetitive inhibition of CYP2C19-mediated S-mephenytoin 4'-hydroxylation with Ki values of 0.19 µM and 1.2 µM, respectively. Vicagrel displayed profound time-dependent inhibition towards CYP2B6 with maximal rate constant of inactivation (kinact) and half-maximal inactivator concentration (KI) values of 0.062 min-1 and 1.52 µM, respectively. No time-dependent inhibition by vicagrel was noted for CYP2C19. For UGT, negligible to moderate inhibition by vicagrel was observed with IC50 values of >50.0, >50.0, 28.2, 8.7, >50.0 and 28.2 µM for UGT1A1, UGT1A3, UGT1A4, UGT1A6, UGT1A9 and UGT2B7, respectively. In terms of mode of reversible inhibition, vicagrel exhibited mixed-type inhibition of UGT1A6-catalyzed N-Acetylserotonin ß-D-glucuronidation with a Ki value of 5.6 µM. No time-dependent inhibition by vicagrel was noted for UGT1A6. PBPK simulation indicated that neither altered AUC nor Cmax of bupropion and S-mephenytoin was observed in the presence of vicagrel. Our study provides inhibitory constants for future DDI prediction between vicagrel and drug substrates of CYP2B6, CYP2C19 and UGT1A6. In addition, our simulation suggests the lack of clinically important DDI between vicagrel and bupropion or S-mephenytoin.


Assuntos
Inibidores das Enzimas do Citocromo P-450/farmacologia , Glucuronosiltransferase/antagonistas & inibidores , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Fenilacetatos/farmacologia , Tiofenos/farmacologia , Bupropiona/administração & dosagem , Bupropiona/farmacocinética , Simulação por Computador , Citocromo P-450 CYP2B6/metabolismo , Inibidores do Citocromo P-450 CYP2B6/administração & dosagem , Inibidores do Citocromo P-450 CYP2B6/farmacologia , Citocromo P-450 CYP2C19/metabolismo , Inibidores do Citocromo P-450 CYP2C19/administração & dosagem , Inibidores do Citocromo P-450 CYP2C19/farmacologia , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Sistema Enzimático do Citocromo P-450/metabolismo , Interações Medicamentosas , Glucuronosiltransferase/metabolismo , Humanos , Técnicas In Vitro , Cinética , Mefenitoína/administração & dosagem , Mefenitoína/farmacocinética , Fenilacetatos/administração & dosagem , Inibidores da Agregação Plaquetária/administração & dosagem , Inibidores da Agregação Plaquetária/farmacologia , Tiofenos/administração & dosagem
4.
Toxicol Appl Pharmacol ; 431: 115729, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34592323

RESUMO

Rosemary (Salvia Rosmarinus) is a rich source of dietary diterpenes with carnosol as one of the major polyphenols used to standardize rosemary extracts approved as a food preservative, however, at present there is not any information on the murine pharmacokinetic profile of carnosol or its potential for drug interactions. The present study utilizes cell-free, cell-based, and animal-based experiments to define the pharmacokinetic profile of the food based phytochemical carnosol. Mice were administered carnosol (100 mg/kg body weight) by oral gavage and plasma levels were analyzed by LC-MS/MS to establish a detailed pharmacokinetic profile. The maximum plasma concentration exceeded 1 µM after a single administration. The results are significant as they offer insights on the potential for food-drug interactions between carnosol from rosemary and active pharmaceutical ingredients. Carnosol was observed to inhibit selected CYP450 enzymes and modulate metabolic enzymes and transporters in in vitro assays.


Assuntos
Abietanos/farmacocinética , Inibidores das Enzimas do Citocromo P-450/farmacocinética , Sistema Enzimático do Citocromo P-450/metabolismo , Conservantes de Alimentos/farmacocinética , Abietanos/administração & dosagem , Abietanos/sangue , Abietanos/isolamento & purificação , Administração Oral , Animais , Disponibilidade Biológica , Óleo de Sementes de Algodão/química , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Inibidores das Enzimas do Citocromo P-450/sangue , Inibidores das Enzimas do Citocromo P-450/isolamento & purificação , Estabilidade de Medicamentos , Conservantes de Alimentos/administração & dosagem , Conservantes de Alimentos/isolamento & purificação , Células HT29 , Células Hep G2 , Humanos , Isoenzimas , Masculino , Proteínas de Membrana Transportadoras/efeitos dos fármacos , Proteínas de Membrana Transportadoras/metabolismo , Camundongos Endogâmicos C57BL , Rosmarinus/química , Temperatura
5.
Drug Des Devel Ther ; 15: 3661-3673, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34456561

RESUMO

PURPOSE: Avitinib is the first third-generation epithelial growth factor receptor (EGFR) inhibitor independently developed in China and is mainly used for treating non-small cell lung cancer. However, pharmacokinetic details are limited. This study explored the in vivo and in vitro effects of avitinib on cytochrome CYP450 enzymes metabolic activity. METHODS: A rapid and sensitive ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method was developed and validated for determining six probe substrates and their metabolites. Avitinib influence on activity levels of CYP isozymes was examined in vitro using human and rat liver microsomes (HLMs/RLMs). For in vivo studies, rats were pretreated with 30 mg/kg avitinib once daily for 7 days (avitinib multiple-doses group), 30 mg/kg avitinib on day 7 (avitinib single-dose group), or an equivalent amount of CMC-Na once daily for 7 days (control group), followed by intragastrical administration of the probe substrates (1 mg/kg tolbutamide and 10 mg/kg phenacetin, bupropion, chlorzoxazone, dextromethorphan, and midazolam). Plasma pharmacokinetics and IC50 values of the probe substrates were then compared. Pharmacokinetic parameters were determined using non-compartmental analysis implemented in a pharmacokinetic program. RESULTS: In vitro experiments revealed different inhibitory effects of avitinib on the six probe substrates with various IC50 values (bupropion, 6.39/22.64 µM; phenacetin, 15.79/48.36 µM; chlorzoxazone, 23.15/57.09 µM; midazolam, 27.64/59.6 µM; tolbutamide, 42.18/6.91 µM; dextromethorphan, 44.39/56.57 µM, in RLMs and HLMs respectively). In vivo analysis revealed significant differences (P <0.05) in distinct pharmacokinetic parameters (AUC(0-t), AUC (0-∞), Cmax, MRT(0-t), MRT (0-∞), and CLz/F) for the six probe substrates after avitinib pretreatment. CONCLUSION: A sensitive and reliable UPLC-MS/MS method was established to determine the concentration of six probe substrates in rat plasma. Avitinib had inhibitory effects on CYP450 enzymes, especially cyp2b1, cyp1a2 in RLMs, CYP2C9 in HLMs, and cyp1a2, cyp2b1, cyp2d1, and cyp2e1 in vivo. Our data recommend caution when avitinib was taken simultaneously with drugs metabolized by CYP450 enzymes.


Assuntos
Inibidores das Enzimas do Citocromo P-450/farmacologia , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Animais , Área Sob a Curva , Cromatografia Líquida de Alta Pressão/métodos , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Sistema Enzimático do Citocromo P-450/metabolismo , Interações Medicamentosas , Humanos , Concentração Inibidora 50 , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Preparações Farmacêuticas/metabolismo , Inibidores de Proteínas Quinases/administração & dosagem , Pirimidinas/administração & dosagem , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem/métodos
6.
Ann Hematol ; 99(9): 2193-2195, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32621180
7.
Drug Des Devel Ther ; 14: 1963-1970, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32546963

RESUMO

BACKGROUND AND PURPOSE: Apatinib is a small-molecule tyrosine kinase inhibitor for the treatment of recurrent or progressive advanced-stage gastric adenocarcinoma or gastroesophageal junction cancer. The in vitro inhibition studies suggested that apatinib exerted potent inhibition on CYP3A4 and CYP2C9. To evaluate the potential of apatinib as a perpetrator in CYP450-based drug-drug interactions in vivo, nifedipine and warfarin were, respectively, selected in the present study as the probe substrates of CYP3A4 and CYP2C9 for clinical drug-drug interaction studies. Since hypertension and thrombus are common adverse effects of vascular targeting anticancer agents, nifedipine and warfarin are usually coadministered with apatinib in clinical practice. METHODS: A single-center, open-label, single-arm, and self-controlled trial was conducted in patients with advanced solid tumors. The patients received a single dose of 30 mg nifedipine on Day 1/14 and a single dose of 3 mg warfarin on Day 3/16. On Day 9-21, the subjects received a daily dose of 750 mg apatinib, respectively. The pharmacokinetics of nifedipine and warfarin in the absence or presence of apatinib was, respectively, investigated. RESULTS: Compared with the single oral administration, coadministration with apatinib contributed to the significant increases of AUC0-48h and Cmax of nifedipine by 83% (90% confidence interval [CI] 1.46-2.31) and 64% (90% CI 1.34-2.01), respectively. Similarly, coadministration with apatinib contributed to the significant increases of AUC0-t and Cmax of S-warfarin by 92% (90% CI 1.68-2.18) and 24% (90% CI 1.10-1.39), respectively. CONCLUSION: Concomitant apatinib administration resulted in significant increases in systemic exposure to nifedipine and S-warfarin. Owing to the risk of pharmacokinetic drug-drug interactions based on CYP3A4/CYP2C9 inhibition by apatinib, caution is advised in the concurrent use of apatinib with either CYP2C9 or CYP3A4 substrates.


Assuntos
Citocromo P-450 CYP2C9/metabolismo , Citocromo P-450 CYP3A/metabolismo , Inibidores das Enzimas do Citocromo P-450/farmacocinética , Neoplasias/tratamento farmacológico , Nifedipino/farmacocinética , Piridinas/farmacocinética , Varfarina/farmacocinética , Administração Oral , Adolescente , Adulto , Idoso , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Interações Medicamentosas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/metabolismo , Nifedipino/administração & dosagem , Piridinas/administração & dosagem , Varfarina/administração & dosagem , Adulto Jovem
8.
Cancer Chemother Pharmacol ; 86(1): 87-95, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32537715

RESUMO

PURPOSE: Fedratinib, an oral selective kinase inhibitor with activity against both wild type and mutationally activated Janus kinase 2, has been approved for the treatment of adult patients with intermediate-2 or high-risk myelofibrosis by the US Food and Drug Administration. In vitro studies indicated that fedratinib was an inhibitor of several cytochrome P450 (CYP) enzymes. The primary objective of this study was to evaluate the effects of repeated doses of fedratinib on the activity of CYP2D6, CYP2C19, and CYP3A4 in patients with solid tumors using a CYP probe cocktail. METHODS: An open-label, one-sequence, two-period, two-treatment crossover study was conducted. Patients were administered a single oral dose cocktail of metoprolol (100 mg), omeprazole (20 mg), and midazolam (2 mg) used as probe substrates for CYP2D6, CYP2C19, and CYP3A4 enzyme activities, respectively, without fedratinib on Day -1 or with fedratinib on Day 15. RESULTS: Coadministration of 500 mg once-daily doses of fedratinib for 15 days increased the mean area under the plasma concentration-time curve from time zero to infinity following a single-dose cocktail containing metoprolol (CYP2D6 substrate), omeprazole (CYP2C19 substrate), and midazolam (CYP3A4 substrate) by 1.77-fold (90% confidence interval [CI] 1.27-2.47) for metoprolol, 2.82-fold (90% CI 2.26-3.53) for omeprazole, and 3.84-fold (90% CI 2.62-5.63) for midazolam, respectively. The mean plasma Day 14/Day 1 ratio of 4ß-hydroxycholesterol, an endogenous biomarker of CYP3A4 activity, was 0.59 (90% CI 0.54-0.66), suggesting a net inhibition of CYP3A4 by fedratinib. CONCLUSION: Fedratinib is a weak inhibitor of CYP2D6, and a moderate inhibitor of CYP2C19 and CYP3A4. These results serve as the basis for dose modifications of these CYP substrate drugs when co-administered with fedratinib.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Inibidores das Enzimas do Citocromo P-450/farmacocinética , Neoplasias/tratamento farmacológico , Pirrolidinas/administração & dosagem , Sulfonamidas/administração & dosagem , Administração Oral , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Estudos Cross-Over , Citocromo P-450 CYP2C19/sangue , Citocromo P-450 CYP2D6/sangue , Citocromo P-450 CYP3A/sangue , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Interações Medicamentosas , Feminino , Humanos , Hidroxicolesteróis/sangue , Masculino , Metoprolol/administração & dosagem , Metoprolol/sangue , Midazolam/administração & dosagem , Midazolam/sangue , Pessoa de Meia-Idade , Omeprazol/administração & dosagem , Omeprazol/sangue , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/farmacocinética , Pirrolidinas/efeitos adversos , Pirrolidinas/farmacocinética , Sulfonamidas/efeitos adversos , Sulfonamidas/farmacocinética
9.
Cancer Chemother Pharmacol ; 85(4): 805-816, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32185484

RESUMO

PURPOSE: Metarrestin is a first-in-class pyrrolo-pyrimidine-derived small molecule targeting a marker of genome organization associated with metastasis and is currently in preclinical development as an anti-cancer agent. Here, we report the in vitro ADME characteristics and in vivo pharmacokinetic behavior of metarrestin. METHODS: Solubility, permeability, and efflux ratio as well as in vitro metabolism of metarrestin in hepatocytes, liver microsomes and S9 fractions, recombinant cytochrome P450 (CYP) enzymes, and potential for CYP inhibition were evaluated. Single dose pharmacokinetic profiles after intravenous and oral administration in mice, rat, dog, monkey, and mini-pig were obtained. Simple allometric scaling was applied to predict human pharmacokinetics. RESULTS: Metarrestin had an aqueous solubility of 150 µM at pH 7.4, high permeability in PAMPA and moderate efflux ratio in Caco-2 assays. The compound was metabolically stable in liver microsomes, S9 fractions, and hepatocytes from six species, including human. Metarrestin is a CYP3A4 substrate and, in mini-pigs, is also directly glucuronidated. Metarrestin did not show cytochrome P450 inhibitory activity. Plasma concentration-time profiles showed low to moderate clearance, ranging from 0.6 mL/min/kg in monkeys to 48 mL/min/kg in mice and moderate to high volume of distribution, ranging from 1.5 L/kg in monkeys to 17 L/kg in mice. Metarrestin has greater than 80% oral bioavailability in all species tested. The excretion of unchanged parent drug in urine was < 5% in dogs and < 1% in monkeys over collection periods of ≥ 144 h; in bile-duct cannulated rats, the excretion of unchanged drug was < 1% in urine and < 2% in bile over a collection period of 48 h. CONCLUSIONS: Metarrestin is a low clearance compound which has good bioavailability and large biodistribution after oral administration. Biotransformation appears to be the major elimination process for the parent drug. In vitro data suggest a low drug-drug interaction potential on CYP-mediated metabolism. Overall favorable ADME and PK properties support metarrestin's progression to clinical investigation.


Assuntos
Sistema Enzimático do Citocromo P-450/química , Microssomos Hepáticos/metabolismo , Pirimidinas/administração & dosagem , Pirimidinas/farmacocinética , Pirróis/administração & dosagem , Pirróis/farmacocinética , Administração Oral , Animais , Biotransformação , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Inibidores das Enzimas do Citocromo P-450/farmacocinética , Sistema Enzimático do Citocromo P-450/metabolismo , Cães , Avaliação Pré-Clínica de Medicamentos , Interações Medicamentosas , Feminino , Haplorrinos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microssomos Hepáticos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie , Suínos , Porco Miniatura , Distribuição Tecidual
10.
Eur J Drug Metab Pharmacokinet ; 45(4): 433-444, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32125665

RESUMO

BACKGROUND AND OBJECTIVES: In vivo studies were performed with the novel, selective, non-steroidal mineralocorticoid receptor antagonist finerenone to assess the relevance of inductive and/or inhibitory effects on cytochrome P450 (CYP) enzymes observed in vitro. METHODS: CYP isoenzyme-specific substrates were incubated in vitro with finerenone or its metabolites to investigate reversible and irreversible inhibitory as well as inductive potential. Three crossover studies in healthy male volunteers investigated the effects of finerenone (20 mg orally) on the pharmacokinetics of the index substrates midazolam (CYP3A4, n = 30), repaglinide (CYP2C8, n = 28) and warfarin (CYP2C9, n = 24). RESULTS: Finerenone caused direct inhibitory effects on CYP activities in vitro in the rank order CYP2C8, CYP1A1 > CYP3A4 > CYP2C9 and CYP2C19, but not on other major CYP isoforms. Moreover, irreversible inhibition of CYP3A4 was observed. The major metabolites of finerenone demonstrated minor reversible inhibition of CYP1A1, CYP2C9 and CYP3A4 with no hint of time-dependent inhibition of any CYP isoform. Calculations from in vitro data according to regulatory guidelines suggested likely inhibition of CYP2C8 and CYP3A4 in vivo, whereas this was not the case for CYP1A1, CYP2C9 and CYP2C19. Furthermore, finerenone and three of its metabolites were inducers of CYP3A4 in vitro with predicted weak-to-moderate in vivo relevance. Studies in healthy volunteers, prompted by these results, demonstrated no effect of finerenone on CYP isoenzymes for which in vitro data had indicated potential inhibition or induction. CONCLUSION: Administration of finerenone 20 mg once daily confers no risk of clinically relevant drug-drug interactions with substrates of cytochrome P450 enzymes.


Assuntos
Carbamatos/farmacocinética , Indutores das Enzimas do Citocromo P-450/farmacologia , Inibidores das Enzimas do Citocromo P-450/farmacologia , Midazolam/farmacocinética , Antagonistas de Receptores de Mineralocorticoides/farmacologia , Naftiridinas/farmacologia , Piperidinas/farmacocinética , Varfarina/farmacocinética , Administração Oral , Adolescente , Adulto , Estudos Cross-Over , Citocromo P-450 CYP2C8/metabolismo , Citocromo P-450 CYP2C9/metabolismo , Citocromo P-450 CYP3A/metabolismo , Indutores das Enzimas do Citocromo P-450/administração & dosagem , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Método Duplo-Cego , Interações Medicamentosas , Alemanha , Humanos , Isoenzimas , Masculino , Pessoa de Meia-Idade , Antagonistas de Receptores de Mineralocorticoides/administração & dosagem , Modelos Biológicos , Naftiridinas/administração & dosagem , Especificidade por Substrato , Adulto Jovem
11.
Pharmacogenomics J ; 20(2): 192-201, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31616047

RESUMO

Clozapine is an atypical antipsychotic metabolized by CYP1A2, CYP2D6, and CYP2C19 enzymes. Among 66 adult schizophrenia patients treated with clozapine-based combination therapies, we explored the impact of genotype-predicted CYP1A2, CYP2D6, and CYP2C19 activity on dose-adjusted clozapine concentrations and symptom severity, with and without correction for inhibitors and inducers of these enzymes. Uncorrected activity scores were not associated with dose-adjusted clozapine concentrations or symptom severity. CYP1A2 and CYP2D6 activity scores corrected for known inducers (i.e., smoking) and inhibitors (e.g., concomitant medications) were associated with dose-adjusted clozapine levels and in the case of CYP1A2, symptom severity. However, smoking status and certain inhibitors of clozapine metabolism (i.e., esomeprazole) explained significantly more variance in dose-adjusted clozapine levels relative to corrected activity scores. These findings highlight the clinical importance of nongenetic factors (smoking, concomitant medications) and suggest that the added utility of CYP1A2, CYP2D6, and CYP2C19 activity scores to guide clozapine dosing is currently limited.


Assuntos
Antipsicóticos/sangue , Clozapina/sangue , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP2C19/metabolismo , Citocromo P-450 CYP2D6/metabolismo , Polimorfismo de Nucleotídeo Único , Esquizofrenia/tratamento farmacológico , Adolescente , Adulto , Idoso , Antipsicóticos/administração & dosagem , Antipsicóticos/uso terapêutico , Clozapina/administração & dosagem , Clozapina/uso terapêutico , Citocromo P-450 CYP1A2/genética , Citocromo P-450 CYP2C19/genética , Citocromo P-450 CYP2D6/genética , Indutores das Enzimas do Citocromo P-450/administração & dosagem , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Relação Dose-Resposta a Droga , Interações Medicamentosas , Genótipo , Humanos , Pessoa de Meia-Idade , Esquizofrenia/sangue , Esquizofrenia/genética , Índice de Gravidade de Doença , Fumar/metabolismo , Adulto Jovem
12.
J Clin Psychopharmacol ; 39(5): 462-471, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31433338

RESUMO

PURPOSE: Increased cannabis use and recent drug approvals pose new challenges for avoiding drug interactions between cannabis products and conventional medications. This review aims to identify drug-metabolizing enzymes and drug transporters that are affected by concurrent cannabis use and, conversely, those co-prescribed medications that may alter the exposure to one or more cannabinoids. METHODS: A systematic literature search was conducted utilizing the Google Scholar search engine and MEDLINE (PubMed) database through March 2019. All articles describing in vitro or clinical studies of cannabis drug interaction potential were retrieved for review. Additional articles of interest were obtained through cross-referencing of published bibliographies. FINDINGS: After comparing the in vitro inhibition parameters to physiologically achievable cannabinoid concentrations, it was concluded that CYP2C9, CYP1A1/2, and CYP1B1 are likely to be inhibited by all 3 major cannabinoids Δ-tetrahydrocannabinol (THC), cannabidiol (CBD), and cannabinol (CBN). The isoforms CYP2D6, CYP2C19, CYP2B6, and CYP2J2 are inhibited by THC and CBD. CYP3A4/5/7 is potentially inhibited by CBD. Δ-Tetrahydrocannabinol also activates CYP2C9 and induces CYP1A1. For non-CYP drug-metabolizing enzymes, UGT1A9 is inhibited by CBD and CBN, whereas UGT2B7 is inhibited by CBD but activated by CBN. Carboxylesterase 1 (CES1) is potentially inhibited by THC and CBD. Clinical studies suggest inhibition of CYP2C19 by CBD, inhibition of CYP2C9 by various cannabis products, and induction of CYP1A2 through cannabis smoking. Evidence of CBD inhibition of UGTs and CES1 has been shown in some studies, but the data are limited at present. We did not identify any clinical studies suggesting an influence of cannabinoids on drug transporters, and in vitro results suggest that a clinical interaction is unlikely. CONCLUSIONS: Medications that are prominent substrates for CYP2C19, CYP2C9, and CYP1A2 may be particularly at risk of altered disposition by concomitant use of cannabis or 1 or more of its constituents. Caution should also be given when coadministered drugs are metabolized by UGT or CES1, on which subject the information remains limited and further investigation is warranted. Conversely, conventional drugs with strong inhibitory or inductive effects on CYP3A4 are expected to affect CBD disposition.


Assuntos
Canabinoides/administração & dosagem , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Interações Medicamentosas , Canabinoides/farmacocinética , Canabinoides/farmacologia , Cannabis/química , Indutores das Enzimas do Citocromo P-450/administração & dosagem , Indutores das Enzimas do Citocromo P-450/farmacocinética , Indutores das Enzimas do Citocromo P-450/farmacologia , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Inibidores das Enzimas do Citocromo P-450/farmacocinética , Inibidores das Enzimas do Citocromo P-450/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Fumar Maconha/epidemiologia
13.
Phytomedicine ; 42: 199-206, 2018 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-29655687

RESUMO

BACKGROUND: Broussonetia papyrifera (L.) Ventenat, a traditional medicinal herb, has been applied as a folk medicine to treat various diseases. Broussochalcone A (BCA), a chalcone compound isolated from the cortex of Broussonetia papyrifera (L.) Ventenat, exhibits several biological activities including potent anti-oxidant, antiplatelet, and cytotoxic effects. PURPOSE: The purpose of this study is to elucidate the inhibitory effect of BCA against CYP2J2 enzyme which is predominantly expressed in human tumor tissues and carcinoma cell lines. STUDY DESIGN: The inhibitory effect of BCA on the activities of CYP2J2-mediated metabolism were investigated using human liver microsomes (HLMs), and its anti-cancer effect against human hepatoma HepG2 cells was also evaluated. METHODS: Two representative CYP2J2-specific probe substrates, astemizole and ebastine, were incubated in HLMs with BCA. After incubation, the samples were analyzed using liquid chromatography-tandem mass spectrometry. To investigate the binding model between BCA and CYP2J2, we carried out structure-based docking simulations by using software and scripts written in-house. RESULTS: BCA inhibited CYP2J2-mediated astemizole O-demethylation and ebastine hydroxylase activities in a concentration dependent manner with Ki values of 2.3 and 3.7 µM, respectively. It also showed cytotoxic effects against human hepatoma HepG2 cells in a dose-dependent manner with activation of apoptosis related proteins. CONCLUSION: Overall, this was the first report of the inhibitory effects of BCA on CYP2J2 in HLMs. The present data suggest that BCA is a potential candidate for further evaluation for its CYP2J2 targeting anti-cancer activities.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Chalconas/farmacologia , Inibidores das Enzimas do Citocromo P-450/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Proteína Forkhead Box O3/metabolismo , Resorcinóis/farmacologia , Antineoplásicos Fitogênicos/administração & dosagem , Astemizol/metabolismo , Butirofenonas/metabolismo , Proliferação de Células/efeitos dos fármacos , Chalconas/administração & dosagem , Chalconas/química , Cromatografia Líquida , Citocromo P-450 CYP2J2 , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Sistema Enzimático do Citocromo P-450/química , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Células Hep G2 , Humanos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Simulação de Acoplamento Molecular , Piperidinas/metabolismo , Resorcinóis/administração & dosagem , Resorcinóis/química , Espectrometria de Massas em Tandem
14.
Biopharm Drug Dispos ; 38(9): 553-556, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28866862

RESUMO

Acetylshikonin is a biologically active compound with anti-cancer and anti-inflammatory activity, which is isolated from the roots of Lithospermum erythrorhizoma. An inhibitory effect of acetylshikonin against CYP2J2 activity was discovered recently. Based on this result, this study was expanded to evaluate the inhibitory effects of acetylshikonin against nine different cytochrome P450 (P450) isoforms in human liver microsomes (HLMs) using substrate cocktails incubation assay. Acetylshikonin showed a strong inhibitory effect against all P450s tested with IC50 values of 1.4-4.0 µ m. Pre-incubation of acetylshikonin with HLMs and NADPH did not alter the inhibition potency, indicating that acetylshikonin is not a mechanism-based inhibitor. SKF-525A, a widely used non-specific P450 inhibitor, had no inhibitory activity against CYP1A2, 2A6, 2E1 and 2J2, while it showed an inhibitory effect against CYP2B6, CYP2C19 and 2D6 with IC50 values of 2.5, 3.6 and 0.5 µ m, respectively. Our findings indicate that acetylshikonin may be a novel general P450 inhibitor, which could replace SKF-525A.


Assuntos
Antraquinonas/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Inibidores das Enzimas do Citocromo P-450/farmacologia , Microssomos Hepáticos/efeitos dos fármacos , Antraquinonas/administração & dosagem , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/farmacologia , Antineoplásicos Fitogênicos/administração & dosagem , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Lithospermum/química , Microssomos Hepáticos/enzimologia , Proadifeno/farmacologia
15.
J Pharm Pharmacol ; 69(12): 1762-1772, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28872689

RESUMO

OBJECTIVES: Excessive exposure to acetaminophen (APAP, paracetamol) can cause liver injury through formation of a reactive metabolite that depletes hepatic glutathione and causes hepatocellular oxidative stress and damage. Generation of this metabolite is mediated by Cytochrome-P450 (CYP) isoforms, mainly CYP2E1. A number of naturally occurring flavonoids can mitigate APAP-induced hepatotoxicity in experimental animal models. Our objective was to determine the mechanism of these protective effects and to evaluate possible human applicability. METHODS: Two flavonoids, luteolin and quercetin, were evaluated as potential inhibitors of eight human CYP isoforms, of six UDP-glucuronosyltransferase (UGT) isoforms and of APAP glucuronidation and sulfation. The experimental model was based on in-vitro metabolism by human liver microsomes, using isoform-specific substrates. KEY FINDINGS: Luteolin and quercetin inhibited human CYP isoforms to varying degrees, with greatest potency towards CYP1A2 and CYP2C8. However, 50% inhibitory concentrations (IC50 values) were generally in the micromolar range. UGT isoforms were minimally inhibited. Both luteolin and quercetin inhibited APAP sulfation but not glucuronidation. CONCLUSIONS: Inhibition of human CYP activity by luteolin and quercetin occurred with IC50 values exceeding customary in-vivo human exposure with tolerable supplemental doses of these compounds. The findings indicate that luteolin and quercetin are not likely to be of clinical value for preventing or treating APAP-induced hepatotoxicity.


Assuntos
Acetaminofen/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Luteolina/farmacologia , Quercetina/farmacologia , Acetaminofen/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Citocromo P-450 CYP1A2/efeitos dos fármacos , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP2C8/efeitos dos fármacos , Citocromo P-450 CYP2C8/metabolismo , Citocromo P-450 CYP2E1/efeitos dos fármacos , Citocromo P-450 CYP2E1/metabolismo , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Inibidores das Enzimas do Citocromo P-450/farmacologia , Interações Medicamentosas , Humanos , Concentração Inibidora 50 , Luteolina/administração & dosagem , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Quercetina/administração & dosagem
16.
Nanomedicine ; 13(5): 1715-1723, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28343019

RESUMO

Nanomedicines are mainly used as drug delivery systems; here we evaluate a new application - to inhibit a drug's metabolism thereby enhancing its effective dose. Micelles containing the natural furanocoumarin 6',7'-dihydroxybergamottin (DHB), a known CYP450 inhibitor, were developed to transiently block hepatic CYP450-mediated drug metabolism and increase the bioavailability of the oncology drug docetaxel. Administered in mice 24h prior to the drug, DHB-micelles enhanced antitumor efficacy in the tumor xenograft models HT-29 and MDA-MB-231, when compared to the drug alone. These DHB-micelles have similar composition to marketed docetaxel-micelles for human use. Despite not being optimized in terms of targeting hepatocytes, they do represent the first injectable example of nanosized metabolism-blocking agents and open the way for further work on such nanomedicines in man.


Assuntos
Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Micelas , Nanomedicina/métodos , Animais , Antineoplásicos , Linhagem Celular Tumoral , Sistema Enzimático do Citocromo P-450 , Humanos , Masculino , Camundongos
17.
Sci Rep ; 7: 41809, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28139732

RESUMO

Glioblastoma (GBM) is a hypervascular primary brain tumor with poor prognosis. HET0016 is a selective CYP450 inhibitor, which has been shown to inhibit angiogenesis and tumor growth. Therefore, to explore novel treatments, we have generated an improved intravenous (IV) formulation of HET0016 with HPßCD and tested in animal models of human and syngeneic GBM. Administration of a single IV dose resulted in 7-fold higher levels of HET0016 in plasma and 3.6-fold higher levels in tumor at 60 min than that in IP route. IV treatment with HPßCD-HET0016 decreased tumor growth, and altered vascular kinetics in early and late treatment groups (p < 0.05). Similar growth inhibition was observed in syngeneic GL261 GBM (p < 0.05). Survival studies using patient derived xenografts of GBM811, showed prolonged survival to 26 weeks in animals treated with focal radiation, in combination with HET0016 and TMZ (p < 0.05). We observed reduced expression of markers of cell proliferation (Ki-67), decreased neovascularization (laminin and αSMA), in addition to inflammation and angiogenesis markers in the treatment group (p < 0.05). Our results indicate that HPßCD-HET0016 is effective in inhibiting tumor growth through decreasing proliferation, and neovascularization. Furthermore, HPßCD-HET0016 significantly prolonged survival in PDX GBM811 model.


Assuntos
Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Glioblastoma/metabolismo , Glioblastoma/patologia , Actinas/metabolismo , Administração Intravenosa , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Inibidores das Enzimas do Citocromo P-450/química , Inibidores das Enzimas do Citocromo P-450/farmacocinética , Modelos Animais de Doenças , Glioblastoma/tratamento farmacológico , Glioblastoma/mortalidade , Humanos , Neovascularização Patológica/tratamento farmacológico , Ratos , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Clin Pharmacol Drug Dev ; 5(3): 188-95, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27163497

RESUMO

This study aimed to determine the impact of food on the pharmacokinetics of orteronel, an investigational nonsteroidal, reversible selective inhibitor of 17,20-lyase. In this open-label, randomized crossover study, healthy subjects received single doses of orteronel 400 mg with a low-fat meal, a high-fat meal, and under fasting conditions in a randomized sequence. Plasma concentrations of orteronel and its primary M-I metabolite were determined by ultra-performance liquid chromatography, and pharmacokinetic parameters were evaluated using mixed-effects analysis of variance model. Compared with fasting conditions, the oral bioavailability of orteronel was increased under fed conditions. The least-squares mean ratio for area under the plasma concentration-time curve after a low-fat breakfast was 135% (90% confidence interval [CI], 126%-145%) compared with fasting conditions. Similarly, after a high-fat breakfast, the corresponding value was 142% (90%CI, 132%-152%). No unexpected safety concerns were raised with orteronel 400 mg administered in the fasted state or after either a high-fat or a low-fat meal; mild adverse events were experienced by 36% of the healthy male subjects.


Assuntos
Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Interações Alimento-Droga , Imidazóis/administração & dosagem , Naftalenos/administração & dosagem , Administração Oral , Adolescente , Adulto , Disponibilidade Biológica , Cromatografia Líquida de Alta Pressão , Estudos Cross-Over , Inibidores das Enzimas do Citocromo P-450/efeitos adversos , Inibidores das Enzimas do Citocromo P-450/farmacocinética , Gorduras na Dieta , Jejum , Humanos , Imidazóis/efeitos adversos , Imidazóis/farmacocinética , Análise dos Mínimos Quadrados , Masculino , Pessoa de Meia-Idade , Naftalenos/efeitos adversos , Naftalenos/farmacocinética , Esteroide 17-alfa-Hidroxilase/antagonistas & inibidores , Adulto Jovem
20.
Eur J Drug Metab Pharmacokinet ; 41(6): 689-703, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26254911

RESUMO

BACKGROUND AND OBJECTIVE: Amiodarone (AMD) is one of the most effective drugs for rhythm control of atrial fibrillation. The use of AMD is also associated with adverse effects in multiple tissues. Both the parent compound and its major metabolite desethylamiodarone (DEA) contribute to the drug's therapeutic and toxic action. The present study aimed to build a whole-body physiologically based pharmacokinetic (PBPK) model for AMD and DEA in rats. METHODS: Pharmacokinetic data from multiple studies were collected. Some of the data were pooled together to develop the PBPK model; others were used to evaluate the model. Development of the model also involved in vitro to in vivo extrapolation based on in vitro metabolism data. RESULTS: The final model consisted of 11 tissue compartments, including therapeutic target organs and those to which AMD and DEA may be harmful. Model simulations were in good agreement with the observed time courses of the drug-metabolite pair in tissues, under various dosing scenarios. The key pharmacokinetic properties of AMD, such as extensive tissue distribution, substantial storage in the fat tissue, and long half-lives in many tissues, were closely reflected. CONCLUSION: The developed PBPK model can be regarded as the first step towards a PBPK-pharmacodynamic model that can used to mechanistically evaluate and explain the high adverse event rate and potentially to determine which factors are the primary drives for experiencing an adverse event.


Assuntos
Amiodarona/análogos & derivados , Amiodarona/farmacocinética , Antiarrítmicos/farmacocinética , Inibidores das Enzimas do Citocromo P-450/farmacocinética , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Modelos Biológicos , Bloqueadores dos Canais de Potássio/farmacocinética , Algoritmos , Amiodarona/administração & dosagem , Amiodarona/efeitos adversos , Amiodarona/sangue , Amiodarona/metabolismo , Animais , Antiarrítmicos/administração & dosagem , Antiarrítmicos/efeitos adversos , Antiarrítmicos/metabolismo , Biotransformação , Barreira Hematoencefálica/metabolismo , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Inibidores das Enzimas do Citocromo P-450/efeitos adversos , Inibidores das Enzimas do Citocromo P-450/metabolismo , Relação Dose-Resposta a Droga , Canais de Potássio Éter-A-Go-Go/metabolismo , Meia-Vida , Infusões Intravenosas , Injeções Intravenosas , Masculino , Taxa de Depuração Metabólica , Microssomos Hepáticos/enzimologia , Microssomos Hepáticos/metabolismo , Bloqueadores dos Canais de Potássio/administração & dosagem , Bloqueadores dos Canais de Potássio/efeitos adversos , Bloqueadores dos Canais de Potássio/metabolismo , Ratos , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA