Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Biochem Pharmacol ; 226: 116390, 2024 08.
Artículo en Inglés | MEDLINE | ID: mdl-38914316

RESUMEN

Infigratinib, an oral FGFR inhibitor for advanced cholangiocarcinoma, yielded two active metabolites, BHS697 and CQM157, with similar receptor affinity. Our study characterized P450s that are responsible for the metabolism of infigratinib to its two major active metabolites, BHS697 and CQM157. In vitro inhibition of P450s and UGTs by infigratinib, BHS697 or CQM157 was further investigated. The unbound apparent Km values for metabolism of infigratinib to BHS697 by HLM, human recombinant CYP2C8, CYP2C19, CYP2D6 and CYP3A4 enzymes are 4.47, 0.65, 2.50, 30.6 and 2.08 µM, while Vmax values are 90.0 pmol/min/mg protein, 0.13, 0.027, 0.81, and 0.56 pmol/min/pmol protein, respectively. The unbound apparent Km value for metabolism of infigratinib to CQM157 by HLM is 0.049 µM, while the Vmax value is 0.32 pmol/min/mg protein respectively. In HLM, infigratinib displayed moderate inhibition of CYP3A4 and CYP2C19 and weak or negligible inhibition of other P450 isoforms. BHS697 exhibited weak inhibition of CYP2B6, CYP2C9, CYP2C19 and CYP3A4, and no inhibition of CYP2C8 and CYP2D6. CQM157 moderately inhibited CYP2C9 and CYP3A4, and weakly or negligibly inhibited other P450 isoforms. Regarding UGTs, infigratinib moderately inhibited UGT1A4 and weakly inhibited UGT1A1, respectively. BHS697 weakly inhibited UGT1A1. In contrast, CQM157 moderately inhibited both UGT1A1 and UGT1A4. Our findings provide novel insights into the metabolism of and potential DDIs implicating infigratinib.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450 , Glucuronosiltransferasa , Humanos , Sistema Enzimático del Citocromo P-450/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Glucuronosiltransferasa/metabolismo , Glucuronosiltransferasa/antagonistas & inhibidores , Microsomas Hepáticos/metabolismo , Microsomas Hepáticos/efectos de los fármacos , Pirimidinas/farmacología , Pirimidinas/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/metabolismo , Compuestos de Fenilurea
2.
Toxicol Appl Pharmacol ; 419: 115502, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33774063

RESUMEN

The toxicological manifestation of many pollutants relies upon their binding to the aryl hydrocarbon receptor (AHR), and it follows a cascade of reactions culminating in an elevated expression of cytochrome P450 (CYP) 1 enzymes. CYP1A1 and CYP1B1 are associated with enhanced carcinogenesis when chronically exposed to certain polyaromatic hydrocarbons, and their inhibition may lead to chemoprevention. We evaluated dibenzyl trisulfide (DTS), expressed in the ethnomedical plant, Petiveria alliacea, for such potential chemoprevention. Using recombinant human CYP1A1 and CYP1B1 bactosomes on a fluorogenic assay, we first demonstrated that DTS moderately inhibited both enzymes with half maximal inhibitory concentration (IC50) values of 1.3 ± 0.3 and 1.7 ± 0.3 µM, respectively. Against CYP1A1, DTS was a reversible, competitive inhibitor with an apparent inhibitory constant (Ki) of 4.55 ± 0.37 µM. In silico molecular modeling showed that DTS binds with an affinity of -39.8 kJ·mol-1, situated inside the binding pocket, approximately 4.3 Å away from the heme group, exhibiting interactions with phenylalanine residue 123 (Phe-123), Phe-224, and Phe-258. Lastly, zebrafish (Danio rerio) embryos were exposed to 0.08-0.8 µM DTS from 24 to 96 h post fertilization (hpf) with the in vivo ethoxyresorufin-O-deethylase (EROD) assay, and, at 96 hpf, DTS significantly suppressed EROD CYP1A activity in a dose-dependent manner, with up to 60% suppression in the highest 0.8 µM exposure group. DTS had no impact on gene transcription levels for cyp1a and aryl hydrocarbon receptor 2 (ahr2). In co-exposure experiments, DTS suppressed CYP1A activity induced by both B[a]P and PCB-126, although these reductions were not significant. Taken together, these results demonstrate that DTS is a direct, reversible, competitive inhibitor of the carcinogen-activating CYP1A enzyme, binding in the active site pocket close to the heme site, and shows potential in chemoprevention.


Asunto(s)
Compuestos de Bencilo/farmacología , Citocromo P-450 CYP1A1/antagonistas & inhibidores , Citocromo P-450 CYP1B1/antagonistas & inhibidores , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Receptores de Hidrocarburo de Aril/metabolismo , Sulfuros/farmacología , Proteínas de Pez Cebra/metabolismo , Activación Metabólica , Animales , Benzo(a)pireno/metabolismo , Benzo(a)pireno/toxicidad , Compuestos de Bencilo/metabolismo , Sitios de Unión , Unión Competitiva , Dominio Catalítico , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1B1/genética , Citocromo P-450 CYP1B1/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Regulación de la Expresión Génica , Humanos , Bifenilos Policlorados/metabolismo , Bifenilos Policlorados/toxicidad , Unión Proteica , Receptores de Hidrocarburo de Aril/genética , Sulfuros/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
3.
Eur J Med Chem ; 215: 113255, 2021 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-33611185

RESUMEN

Human cytochrome P450 enzyme CYP4Z1 represents a promising target for the treatment of a multitude of malignancies including breast cancer. The most active known non-covalent inhibitor (1-benzylimidazole) only shows low micromolar affinity to CYP4Z1. We report a new, highly active inhibitor for CYP4Z1 showing confirmed binding in an enzymatic assay and an IC50 value of 63 ± 19 nM in stably transfected MCF-7 cells overexpressing CYP4Z1. The new inhibitor was identified by a systematically developed virtual screening protocol. Binding was rationalized using a carefully elaborated 3D pharmacophore hypothesis and thoroughly characterized using extensive molecular dynamics simulations and dynamic 3D pharmacophore (dynophore) analyses. This novel inhibitor represents a valuable pharmacological tool to accelerate characterization of the still understudied CYP4Z1 and might pave the way for a new treatment strategy in CYP4Z1-associated malignancies. The presented in silico model for predicting CYP4Z1 interaction provides novel mechanistic insights and revealed that the drug ozagrel interacts with CYP4Z1.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/farmacología , Familia 4 del Citocromo P450/antagonistas & inhibidores , Imidazoles/farmacología , Animales , Inhibidores Enzimáticos del Citocromo P-450/química , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Familia 4 del Citocromo P450/metabolismo , Descubrimiento de Drogas , Humanos , Imidazoles/química , Imidazoles/metabolismo , Células MCF-7 , Metacrilatos/farmacología , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Unión Proteica , Conejos , Relación Estructura-Actividad
4.
Arch Toxicol ; 94(11): 3671-3722, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33111191

RESUMEN

The cytochrome P450 (CYP) enzyme family is the most important enzyme system catalyzing the phase 1 metabolism of pharmaceuticals and other xenobiotics such as herbal remedies and toxic compounds in the environment. The inhibition and induction of CYPs are major mechanisms causing pharmacokinetic drug-drug interactions. This review presents a comprehensive update on the inhibitors and inducers of the specific CYP enzymes in humans. The focus is on the more recent human in vitro and in vivo findings since the publication of our previous review on this topic in 2008. In addition to the general presentation of inhibitory drugs and inducers of human CYP enzymes by drugs, herbal remedies, and toxic compounds, an in-depth view on tyrosine-kinase inhibitors and antiretroviral HIV medications as victims and perpetrators of drug-drug interactions is provided as examples of the current trends in the field. Also, a concise overview of the mechanisms of CYP induction is presented to aid the understanding of the induction phenomena.


Asunto(s)
Antirretrovirales/farmacología , Inductores de las Enzimas del Citocromo P-450/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Xenobióticos/metabolismo , Animales , Antirretrovirales/antagonistas & inhibidores , Antineoplásicos/farmacología , Interacciones Farmacológicas , Humanos
5.
Molecules ; 25(19)2020 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-33050066

RESUMEN

AB-FUBINACA, a synthetic indazole carboxamide cannabinoid, has been used worldwide as a new psychoactive substance. Because drug abusers take various drugs concomitantly, it is necessary to explore potential AB-FUBINACA-induced drug-drug interactions caused by modulation of drug-metabolizing enzymes and transporters. In this study, the inhibitory effects of AB-FUBINACA on eight major human cytochrome P450s (CYPs) and six uridine 5'-diphospho-glucuronosyltransferases (UGTs) of human liver microsomes, and on eight clinically important transport activities including organic cation transporters (OCT)1 and OCT2, organic anion transporters (OAT)1 and OAT3, organic anion transporting polypeptide transporters (OATP)1B1 and OATP1B3, P-glycoprotein, and breast cancer resistance protein (BCRP) in transporter-overexpressing cells were investigated. AB-FUBINACA inhibited CYP2B6-mediated bupropion hydroxylation via mixed inhibition with Ki value of 15.0 µM and competitively inhibited CYP2C8-catalyzed amodiaquine N-de-ethylation, CYP2C9-catalyzed diclofenac 4'-hydroxylation, CYP2C19-catalyzed [S]-mephenytoin 4'-hydroxylation, and CYP2D6-catalyzed bufuralol 1'-hydroxylation with Ki values of 19.9, 13.1, 6.3, and 20.8 µM, respectively. AB-FUBINACA inhibited OCT2-mediated MPP+ uptake via mixed inhibition (Ki, 54.2 µM) and competitively inhibited OATP1B1-mediated estrone-3-sulfate uptake (Ki, 94.4 µM). However, AB-FUBINACA did not significantly inhibit CYP1A2, CYP2A6, CYP3A4, UGT1A1, UGT1A3, UGT1A4, UGT1A6, or UGT2B7 enzyme activities at concentrations up to 100 µM. AB-FUBINACA did not significantly inhibit the transport activities of OCT1, OAT1/3, OATP1B3, P-glycoprotein, or BCRP at concentrations up to 250 µM. As the pharmacokinetics of AB-FUBINACA in humans and animals remain unknown, it is necessary to clinically evaluate potential in vivo pharmacokinetic drug-drug interactions induced by AB-FUBINACA-mediated inhibition of CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, OCT2, and OATP1B1 activities.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Glucuronosiltransferasa/metabolismo , Indazoles/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Uridina Difosfato/metabolismo , Cannabinoides/metabolismo , Línea Celular , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Interacciones Farmacológicas/fisiología , Células HEK293 , Humanos , Microsomas Hepáticos/metabolismo
6.
J Agric Food Chem ; 68(34): 9235-9244, 2020 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-32786866

RESUMEN

The products of the cytochrome P450 monooxygenase (CYP)-catalyzed oxidation of arachidonic acid (AA), that is, epoxy- and hydroxy-fatty acids, play a crucial role in the homeostasis of several physiological processes. In a liver microsome-based multienzyme assay using AA as natural substrate, we investigated how polyphenols inhibit different oxylipin-forming CYP in parallel but independently from each other. The ω-hydroxylating CYP4F2 and CYP4A11 were investigated, as well as the epoxidizing CYP2C-subfamily and CYP3A4 along with the (ω-n)-hydroxylating CYP1A1 and CYP2E1. The oxylipin formation was inhibited by several polyphenols with a remarkable selectivity and a potency comparable to known CYP inhibitors. The flavone apigenin inhibited the epoxidation, ω-hydroxylation, and (ω-n)-hydroxylation of AA with IC50 values of 4.4-9.8, 2.9-10, and 10-25 µM, respectively. Other flavones such as wogonin selectively inhibited CYP1A1-catalyzed (ω-n)-hydroxylation with an IC50 value of 0.10-0.22 µM, while the isoflavone genistein was a selective ω-hydroxylase inhibitor (IC50: 5.5-46 µM). Of note, the flavanone naringenin and the anthocyanidin perlargonidin did not inhibit CYPs of the AA cascade. Moderate permeability of apigenin as tested in the Caco-2 model of intestinal absorption (Papp: 4.5 ± 1 × 10-6 cm/s) and confirmation of the inhibition of 20-HETE formation by apigenin in the colorectal cancer-derived cell line HCT 116 (IC50: 1.5-8.8 µM) underline the possible in vivo relevance of these effects. Further research is needed to better understand how polyphenols impact human health by this newly described molecular mode of action.


Asunto(s)
Ácido Araquidónico/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/química , Sistema Enzimático del Citocromo P-450/metabolismo , Polifenoles/química , Ácido Araquidónico/química , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Sistema Enzimático del Citocromo P-450/química , Humanos , Microsomas Hepáticos/química , Microsomas Hepáticos/enzimología , Microsomas Hepáticos/metabolismo , Oxilipinas/química , Oxilipinas/metabolismo , Polifenoles/metabolismo
7.
Xenobiotica ; 50(12): 1413-1422, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32628085

RESUMEN

Oral antidyslipidaemic drug pemafibrate is cleared from human plasma via hepatic uptake by organic anion transporting polypeptide (OATP) 1B1 and oxidation by cytochromes P450 (P450) 2C8, 2C9 and 3A4. The pharmacokinetic profiles of pemafibrate with virtual administrations of P450 inhibitors and/or disease interactions were generated using a physiologically based pharmacokinetic (PBPK) model previously established for co-administration of pemafibrate with OATP1B1 inhibitors. This PBPK model was validated in the current study using reported maximum pemafibrate plasma concentrations and areas under the curve from interaction studies in healthy subjects co-administered with clopidogrel (P450 2C8 inhibitor), fluconazole (P450 2C9/3A4 inhibitor) or clarithromycin (P450 3A4 inhibitor). Virtual co-administrations of pemafibrate with clopidogrel, fluconazole or clarithromycin increased the predicted plasma exposures of pemafibrate 1.4-1.7-fold, 1.2-1.4-fold and 2.9-11-fold, respectively, in subjects with or without moderate or severe renal impairment or Child-Pugh A or B liver cirrhosis. Some of the exposure-enhancing effects of clarithromycin may originate from its inhibitory potential toward OATP1B1, because the estimated effects of itraconazole (a P450 3A4 inhibitor) were only minor. Simulations using the current PBPK model in groups of virtual subjects with or without renal or hepatic impairment revealed modified pharmacokinetic profiles for pemafibrate following co-administration of typical P450 inhibitors.


Asunto(s)
Benzoxazoles/farmacocinética , Butiratos/farmacocinética , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Preparaciones Farmacéuticas/metabolismo , Claritromicina/farmacocinética , Clopidogrel/farmacocinética , Sistema Enzimático del Citocromo P-450/metabolismo , Fluconazol/farmacocinética , Humanos , Hígado/metabolismo
8.
J Med Chem ; 63(9): 4824-4836, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32302132

RESUMEN

Mammary-tissue-restricted cytochrome P450 4Z1 (CYP4Z1) has garnered interest for its potential role in breast cancer progression. CYP4Z1-dependent metabolism of arachidonic acid preferentially generates 14,15-epoxyeicosatrienoic acid (14,15-EET), a metabolite known to influence cellular proliferation, migration, and angiogenesis. In this study, we developed time-dependent inhibitors of CYP4Z1 designed as fatty acid mimetics linked to the bioactivatable pharmacophore, 1-aminobenzotriazole (ABT). The most potent analogue, 8-[(1H-benzotriazol-1-yl)amino]octanoic acid (7), showed a 60-fold lower shifted-half-maximal inhibitory concentration (IC50) for CYP4Z1 compared to ABT, efficient mechanism-based inactivation of the enzyme evidenced by a KI = 2.2 µM and a kinact = 0.15 min-1, and a partition ratio of 14. Furthermore, 7 exhibited low off-target inhibition of other CYP isozymes. Finally, low micromolar concentrations of 7 inhibited 14,15-EET production in T47D breast cancer cells transfected with CYP4Z1. This first-generation, selective mechanism-based inhibitor (MBI) will be a useful molecular tool to probe the biochemical role of CYP4Z1 and its association with breast cancer.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/farmacología , Familia 4 del Citocromo P450/antagonistas & inhibidores , Ácidos Grasos/farmacología , Triazoles/farmacología , Ácido Araquidónico/metabolismo , Línea Celular Tumoral , Inhibidores Enzimáticos del Citocromo P-450/síntesis química , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Diseño de Fármacos , Ácidos Grasos/síntesis química , Ácidos Grasos/metabolismo , Humanos , Microsomas Hepáticos/metabolismo , Oxidación-Reducción , Triazoles/síntesis química , Triazoles/metabolismo
9.
Drug Des Devel Ther ; 14: 429-434, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32099327

RESUMEN

BACKGROUND: Calycosin (CAL), a type of O-methylated isoflavone extracted from the herb Astralagusmembranaceus (AM), is a bioactive chemical with antioxidative, antiphlogistic and antineoplastic activities commonly used in traditional alternative Chinese medicine. AM has been shown to confer health benefits as an adjuvant in the treatment of a variety of diseases. AIM: The main objective of this study was to determine whether CAL influences the cytochrome P450 (CYP450) system involved in drug metabolism. METHODS: Midazolam, tolbutamide, omeprazole, metoprolol and phenacetin were selected as probe drugs. Rats were randomly divided into three groups, specifically, 5% Carboxymethyl cellulose (CMC) for 8 days (Control), 5% CMC for 7 days + CAL for 1 day (single CAL) and CAL for 8 days (conc CAL), and metabolism of the five probe drugs evaluated using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). RESULTS: No significant differences were observed for omeprazole and midazolam, compared to the control group. T max and t1/2 values of only one probe drug, phenacetin, in the conc CAL group were significantly different from those of the control group (T max h: 0.50±0.00 vs 0.23±0.15; control vs conc CAL). C max of tolbutamide was decreased about two-fold in the conc CAL treatment group (conc vs control: 219.48 vs 429.56, P<0.001). CONCLUSION: Calycosin inhibits the catalytic activities of CYP1A2, CYP2D6 and CYP2C9. Accordingly, we recommend caution, particularly when combining CAL as a modality therapy with drugs metabolized by CYP1A2, CYP2D6 and CYP2C9, to reduce the potential risks of drug accumulation or ineffective treatment.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Medicamentos Herbarios Chinos/metabolismo , Isoflavonas/metabolismo , Animales , Inhibidores Enzimáticos del Citocromo P-450/química , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/farmacología , Isoflavonas/química , Isoflavonas/farmacología , Medicina Tradicional China , Metoprolol/química , Metoprolol/metabolismo , Midazolam/química , Midazolam/metabolismo , Omeprazol/química , Omeprazol/metabolismo , Fenacetina/química , Fenacetina/metabolismo , Ratas , Tolbutamida/química , Tolbutamida/metabolismo
10.
Chem Biol Drug Des ; 95(5): 520-533, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32060993

RESUMEN

Enzymes in the cytochrome P450 family 1 (CYP1) catalyze metabolic activation of procarcinogens and deactivation of certain anticancer drugs. Inhibition of these enzymes is a potential approach for cancer chemoprevention and treatment of CYP1-mediated drug resistance. We characterized inhibition of human CYP1A1, CYP1A2, and CYP1B1 enzymes by the novel inhibitor N-(3,5-dichlorophenyl)cyclopropanecarboxamide (DCPCC) and α-naphthoflavone (ANF). Depending on substrate, IC50 values of DCPCC for CYP1A1 or CYP1B1 were 10-95 times higher than for CYP1A2. IC50 of DCPCC for CYP1A2 was 100-fold lower than for enzymes in CYP2 and CYP3 families. DCPCC IC50 values were 10-680 times higher than the ones of ANF. DCPCC was a mixed-type inhibitor of CYP1A2. ANF was a competitive tight-binding inhibitor of CYP1A1, CYP1A2, and CYP1B1. CYP1A1 oxidized DCPCC more rapidly than CYP1A2 or CYP1B1 to the same metabolite. Molecular dynamics simulations and binding free energy calculations explained the differences of binding of DCPCC and ANF to the active sites of all three CYP1 enzymes. We conclude that DCPCC is a more selective inhibitor for CYP1A2 than ANF. DCPCC is a candidate structure to modulate CYP1A2-mediated metabolism of procarcinogens and anticancer drugs.


Asunto(s)
Amidas/química , Benzoflavonas/química , Ciclopropanos/química , Inhibidores Enzimáticos del Citocromo P-450/química , Familia 1 del Citocromo P450/antagonistas & inhibidores , Amidas/metabolismo , Benzoflavonas/metabolismo , Sitios de Unión , Dominio Catalítico , Cumarinas/química , Cumarinas/metabolismo , Ciclopropanos/metabolismo , Citocromo P-450 CYP1A1/antagonistas & inhibidores , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/química , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP1B1/antagonistas & inhibidores , Citocromo P-450 CYP1B1/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Familia 1 del Citocromo P450/metabolismo , Humanos , Hígado/enzimología , Simulación de Dinámica Molecular , Oxidación-Reducción
11.
J Chem Theory Comput ; 16(2): 1300-1310, 2020 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-31894691

RESUMEN

Calculating free energies of binding (ΔGbind) between ligands and their target protein is of major interest to drug discovery and safety, yet it is still associated with several challenges and difficulties. Linear interaction energy (LIE) is an efficient in silico method for ΔGbind computation. LIE models can be trained and used to directly calculate binding affinities from interaction energies involving ligands in the bound and unbound states only, and LIE can be combined with statistical weighting to calculate ΔGbind for flexible proteins that may bind their ligands in multiple orientations. Here, we investigate if LIE predictions can be effectively improved by explicitly including the entropy of (de)solvation into our free-energy calculations. For that purpose, we combine LIE calculations for the protein-ligand-bound state with explicit free-energy perturbation to rigorously compute the unbound ligand's solvation free energy. We show that for 28 Cytochrome P450 2A6 (CYP2A6) ligands, coupling LIE with alchemical solvation free-energy calculation helps to improve obtained correlation between computed and reference (experimental) binding data.


Asunto(s)
Citocromo P-450 CYP2A6/química , Ligandos , Simulación de Dinámica Molecular , Citocromo P-450 CYP2A6/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/química , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Humanos , Unión Proteica , Termodinámica
12.
Chem Commun (Camb) ; 56(11): 1733-1736, 2020 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-31938799

RESUMEN

We report here that pregnenolonyl-α-glucoside (2), a steryl glycoside synthesized directly from pregnenolone and glucose via a consecutive multienzyme-catalyzed process, exhibits marked dose-dependent cytotoxic activity against HT29, AGS, and ES-2 cells with IC50 values of 23.5 to 50.9 µM. An in vitro CYP17A1 binding pattern assay and protein-ligand docking model support that 2, like abiraterone, binds in the active site heme iron pocket of CYP17A1.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Glucósidos/farmacología , Pregnenolona/análogos & derivados , Pregnenolona/farmacología , Androstenos/metabolismo , Androstenos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Bacterias/enzimología , Dominio Catalítico , Línea Celular Tumoral , Inhibidores Enzimáticos del Citocromo P-450/química , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Glucósidos/síntesis química , Glucósidos/metabolismo , Glicosilación , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , Pregnenolona/metabolismo , Unión Proteica
13.
Chem Res Toxicol ; 32(12): 2488-2498, 2019 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-31799839

RESUMEN

Cytochrome P450 4B1 (CYP4B1) has been explored as a candidate enzyme in suicide gene systems for its ability to bioactivate the natural product 4-ipomeanol (IPO) to a reactive species that causes cytotoxicity. However, metabolic limitations of IPO necessitate discovery of new "pro-toxicant" substrates for CYP4B1. In the present study, we examined a series of synthetically facile N-alkyl-3-furancarboxamides for cytotoxicity in HepG2 cells expressing CYP4B1. This compound series maintains the furan warhead of IPO while replacing its alcohol group with alkyl chains of varying length (C1-C8). Compounds with C3-C6 carbon chain lengths showed similar potency to IPO (LD50 ≈ 5 µM). Short chain analogs (<3 carbons) and long chain analogs (>6 carbons) exhibited reduced toxicity, resulting in a parabolic relationship between alkyl chain length and cytotoxicity. A similar parabolic relationship was observed between alkyl chain length and reactive intermediate formation upon trapping of the putative enedial as a stable pyrrole adduct in incubations with purified recombinant rabbit CYP4B1 and common physiological nucleophiles. These parabolic relationships reflect the lower affinity of shorter chain compounds for CYP4B1 and increased ω-hydroxylation of the longer chain compounds by the enzyme. Furthermore, modest time-dependent inhibition of CYP4B1 by N-pentyl-3-furancarboxamide was completely abolished when trapping agents were added, demonstrating escape of reactive intermediates from the enzyme after bioactivation. An insulated CYP4B1 active site may explain the rarely observed direct correlation between adduct formation and cell toxicity reported here.


Asunto(s)
Amidas/toxicidad , Hidrocarburo de Aril Hidroxilasas/metabolismo , Furanos/toxicidad , Activación Metabólica , Amidas/síntesis química , Amidas/metabolismo , Animales , Hidrocarburo de Aril Hidroxilasas/antagonistas & inhibidores , Hidrocarburo de Aril Hidroxilasas/química , Dominio Catalítico , Inhibidores Enzimáticos del Citocromo P-450/síntesis química , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/toxicidad , Furanos/síntesis química , Furanos/metabolismo , Células Hep G2 , Humanos , Hidroxilación , Cinética , Simulación del Acoplamiento Molecular , Estructura Molecular , Unión Proteica , Conejos , Relación Estructura-Actividad , Terpenos/química , Terpenos/toxicidad
14.
Bioorg Med Chem Lett ; 29(16): 2016-2024, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31257085

RESUMEN

Cytochromes P450 (CYPs) play an important role in the metabolism of endogenic and xenobiotic substances, especially drugs. In addition, many CYPs may serve as targets for disease treatment. However, due to the presence of a common heme, the hydrophobicity of the CYP binding cavity, and the high homology within the binding pocket, most CYP inhibitors lack selectivity, which often leads to drug-drug interactions. Therefore, it is meaningful to develop highly selective CYP inhibitors. In this review, we summarize some of the strategies that have been used to develop highly selective CYP inhibitors, such as the weakening of the heme-binding group interaction, reduction of molecular lipophilicity and introduction of small structural changes within compounds.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Animales , Sitios de Unión , Inhibidores Enzimáticos del Citocromo P-450/química , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Sistema Enzimático del Citocromo P-450/química , Desarrollo de Medicamentos , Hemo/química , Hemo/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Unión Proteica
15.
J Biol Chem ; 294(28): 10928-10941, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31147443

RESUMEN

Cytochrome P450 (P450) enzymes are major catalysts involved in the oxidations of most drugs, steroids, carcinogens, fat-soluble vitamins, and natural products. The binding of substrates to some of the 57 human P450s and other mammalian P450s is more complex than a two-state system and has been proposed to involve mechanisms such as multiple ligand occupancy, induced-fit, and conformational-selection. Here, we used kinetic analysis of binding with multiple concentrations of substrates and computational modeling of these data to discern possible binding modes of several human P450s. We observed that P450 2D6 binds its ligand rolapitant in a mechanism involving conformational-selection. P450 4A11 bound the substrate lauric acid via conformational-selection, as did P450 2C8 with palmitic acid. Binding of the steroid progesterone to P450 21A2 was also best described by a conformational-selection model. Hexyl isonicotinate binding to P450 2E1 could be described by either a conformational-selection or an induced-fit model. Simulation of the binding of the ligands midazolam, bromocriptine, testosterone, and ketoconazole to P450 3A4 was consistent with an induced-fit or a conformational-selection model, but the concentration dependence of binding rates for varying both P450 3A4 and midazolam concentrations revealed discordance in the parameters, indicative of conformational-selection. Binding of the P450s 2C8, 2D6, 3A4, 4A11, and 21A2 was best described by conformational-selection, and P450 2E1 appeared to fit either mode. These findings highlight the complexity of human P450-substrate interactions and that conformational-selection is a dominant feature of many of these interactions.


Asunto(s)
Citocromo P-450 CYP3A/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Conformación Proteica/efectos de los fármacos , Catálisis , Citocromo P-450 CYP2D6/metabolismo , Citocromo P-450 CYP2E1/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Sistema Enzimático del Citocromo P-450/fisiología , Humanos , Cinética , Ácidos Láuricos , Ligandos , Conformación Molecular , Oxidación-Reducción , Ácido Palmítico , Unión Proteica/fisiología , Compuestos de Espiro , Especificidad por Sustrato/fisiología
16.
Curr Top Med Chem ; 19(5): 319-336, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30674264

RESUMEN

Drug-drug interaction (DDI) is the phenomenon of alteration of the pharmacological activity of a drug(s) when another drug(s) is co-administered in cases of so-called polypharmacy. There are three types of DDIs: pharmacokinetic (PK), pharmacodynamic, and pharmaceutical. PK is the most frequent type of DDI, which often appears as a result of the inhibition or induction of drug-metabolising enzymes (DME). In this review, we summarise in silico methods that may be applied for the prediction of the inhibition or induction of DMEs and describe appropriate computational methods for DDI prediction, showing the current situation and perspectives of these approaches in medicinal and pharmaceutical chemistry. We review sources of information on DDI, which can be used in pharmaceutical investigations and medicinal practice and/or for the creation of computational models. The problem of the inaccuracy and redundancy of these data are discussed. We provide information on the state-of-the-art physiologically- based pharmacokinetic modelling (PBPK) approaches and DME-based in silico methods. In the section on ligand-based methods, we describe pharmacophore models, molecular field analysis, quantitative structure-activity relationships (QSAR), and similarity analysis applied to the prediction of DDI related to the inhibition or induction of DME. In conclusion, we discuss the problems of DDI severity assessment, mention factors that influence severity, and highlight the issues, perspectives and practical using of in silico methods.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Preparaciones Farmacéuticas/metabolismo , Inducción Enzimática , Humanos , Preparaciones Farmacéuticas/química , Relación Estructura-Actividad
17.
J Med Chem ; 61(20): 9229-9245, 2018 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-30216063

RESUMEN

Cytochrome P450 (CYP) 1B1 is involved in the bioactivation of procarcinogens and drug resistance. To obtain selective CYP1B1 inhibitors over CYP1A1, we synthesized four series of estrane derivatives: (1) 12 estrone (E1)- and 17ß-estradiol (E2)-derivatives bearing a 3- or a 4-pyridinyl core at C2, C3, or C4, (2) eight estrane derivatives with different sulfur groups at C3, (3) 19 E1- and E2-derivatives bearing distinct aryls at C2, and (4) five D-ring derivatives. E2-derivatives were more active than oxidized E1-analogues, thus highlighting the key role of 17ß-OH for interaction with CYP1B1. 2-(4-Fluorophenyl)-E2 was the best CYP1B1 inhibitor (IC50 = 0.24 µM), with a selectivity index (SI) of 20 over CYP1A1. Furthermore, the addition of a C17α-ethynyl group as D-ring modification improved the selectivity index to 25 with only a slight loss of activity (IC50 = 0.37 µM). Our docking results showed that these compounds fit better into the CYP1B1 binding site than that of CYP1A1.


Asunto(s)
Citocromo P-450 CYP1B1/antagonistas & inhibidores , Inhibidores Enzimáticos del Citocromo P-450/síntesis química , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Diseño de Fármacos , Estranos/síntesis química , Estranos/farmacología , Técnicas de Química Sintética , Citocromo P-450 CYP1B1/química , Citocromo P-450 CYP1B1/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/química , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Estranos/química , Estranos/metabolismo , Simulación del Acoplamiento Molecular , Conformación Proteica , Azufre/química
18.
Drug Metab Lett ; 12(2): 101-116, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30117405

RESUMEN

BACKGROUND: The use of polypharmacy in the present day clinical therapy has made the identification of clinical drug-drug interaction risk an important aspect of drug development process. Although many drugs can be metabolized to sulfoxide and/or sulfone metabolites, seldom is known on the CYP inhibition potential and/or the metabolic fate for such metabolites. OBJECTIVE: The key objectives were: a) to evaluate the in vitro CYP inhibition potential of selected parent drugs with sulfoxide/sulfone metabolites; b) to assess the in vitro metabolic fate of the same panel of parent drugs and metabolites. METHODS: In vitro drug-drug interaction potential of test compounds was investigated in two stages; 1) assessment of CYP450 inhibition potential of test compounds using human liver microsomes (HLM); and 2) assessment of test compounds as substrate of Phase I enzymes; including CYP450, FMO, AO and MAO using HLM, recombinant human CYP enzymes (rhCYP), Human Liver Cytosol (HLC) and Human Liver Mitochondrial (HLMit). All samples were analysed by LC-MS-MS method. RESULTS: CYP1A2 was inhibited by methiocarb, triclabendazole, triclabendazole sulfoxide, and ziprasidone sulfone with IC50 of 0.71 µM, 1.07 µM, 4.19 µM, and 17.14 µM, respectively. CYP2C8 was inhibited by montelukast, montelukast sulfoxide, montelukast sulfone, tribendazole, triclabendazole sulfoxide, and triclabendazole sulfone with IC50 of 0.08 µM, 0.05 µM, 0.02 µM, 3.31 µM, 8.95 µM, and 1.05 µM, respectively. CYP2C9 was inhibited by triclabendazole, triclabendazole sulfoxide, triclabendazole sulfone, montelukast, montelukast sulfoxide and montelukast sulfone with IC50 of 1.17 µM, 1.95 µM, 0.69 µM, 1.34 µM, 3.61 µM and 2.15 µM, respectively. CYP2C19 was inhibited by triclabendazole and triclabendazole sulfoxide with IC50 of 0.25 and 0.22, respectively. CYP3A4 was inhibited by montelukast sulfoxide and triclabendazole with IC50 of 9.33 and 15.11, respectively. Amongst the studied sulfoxide/sulfone substrates, the propensity of involvement of CY2C9 and CYP3A4 enzyme was high (approximately 56% of total) in the metabolic fate experiments. CONCLUSION: Based on the findings, a proper risk assessment strategy needs to be factored (i.e., perpetrator and/or victim drug) to overcome any imminent risk of potential clinical drug-drug interaction when sulfoxide/sulfone metabolite(s) generating drugs are coadministered in therapy.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Sulfonas/farmacología , Sulfóxidos/farmacología , Acetatos/metabolismo , Albendazol/análogos & derivados , Albendazol/metabolismo , Aldicarb/análogos & derivados , Aldicarb/metabolismo , Biotransformación , Ciclopropanos , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/toxicidad , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Humanos , Isoenzimas , Metiocarb/análogos & derivados , Metiocarb/metabolismo , Microsomas Hepáticos/enzimología , Piperazinas/metabolismo , Quinolinas/metabolismo , Medición de Riesgo , Sulfuros , Sulfonas/metabolismo , Sulfonas/toxicidad , Sulfóxidos/metabolismo , Sulfóxidos/toxicidad , Tiazoles/metabolismo , Triclabendazol/metabolismo
19.
J Med Chem ; 61(13): 5594-5608, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29878770

RESUMEN

It is necessary for aldosterone synthase (CYP11B2) inhibitors to have both high potency and high selectivity over 11ß-hydroxylase (CYP11B1), a critical enzyme for cortisol synthesis. Previous studies have reported a number of CYP11B2 inhibitors, most of which have an imidazole or pyridine ring to coordinate the heme-iron motif of CYP11B2; however, highly selective inhibitors of human CYP11B2 are still needed. To expand the selectivity in humans, we explored alternative templates and found that pyrazoles were suitable templates for CYP11B2 inhibitors. Investigation of pyrazoles, especially N-alkyl pyrazoles, as a new template to coordinate the heme-iron motif led to a potent and highly selective CYP11B2 inhibitor 28 with an aldosterone-lowering effect at 1 mg/kg dosing in cynomolgus monkeys.


Asunto(s)
Citocromo P-450 CYP11B2/antagonistas & inhibidores , Descubrimiento de Drogas , Hemo , Hierro , Pirazoles/química , Pirazoles/farmacología , Secuencias de Aminoácidos , Citocromo P-450 CYP11B2/química , Citocromo P-450 CYP11B2/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/química , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Humanos , Concentración 50 Inhibidora , Simulación del Acoplamiento Molecular , Pirazoles/metabolismo
20.
J Med Chem ; 61(11): 4946-4960, 2018 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-29792703

RESUMEN

Inhibition of androgen biosynthesis is clinically effective for treating androgen-responsive prostate cancer. Abiraterone is a clinical first-in-class inhibitor of cytochrome P450 17A1 (CYP17A1) required for androgen biosynthesis. However, abiraterone also causes hypertension, hypokalemia, and edema, likely due in part to off-target inhibition of another steroidogenic cytochrome P450, CYP21A2. Abiraterone analogs were designed based on structural evidence that B-ring substituents may favorably interact with polar residues in binding CYP17A1 and sterically clash with residues in the CYP21A2 active site. The best analogs increased selectivity of CYP17A1 inhibition up to 84-fold compared with 6.6-fold for abiraterone. Cocrystallization with CYP17A1 validated the intended new contacts with CYP17A1 active site residues. Docking these analogs into CYP21A2 identified steric clashes that likely underlie decreased binding and CYP21A2 inhibition. Overall, these analogs may offer a clinical advantage in the form of reduced side effects.


Asunto(s)
Androstenos/química , Androstenos/farmacología , Inhibidores Enzimáticos del Citocromo P-450/química , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Diseño de Fármacos , Esteroide 17-alfa-Hidroxilasa/antagonistas & inhibidores , Esteroide 21-Hidroxilasa/antagonistas & inhibidores , Androstenos/metabolismo , Dominio Catalítico , Inhibidores Enzimáticos del Citocromo P-450/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Esteroide 17-alfa-Hidroxilasa/química , Esteroide 17-alfa-Hidroxilasa/metabolismo , Esteroide 21-Hidroxilasa/química , Esteroide 21-Hidroxilasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA