Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 162
Filtrar
1.
Chem Res Toxicol ; 36(6): 934-946, 2023 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-37148271

RESUMEN

We recently disclosed SAR studies on systemically acting, amide-based inhibitors of diacylglycerol acyltransferase 2 (DGAT2) that addressed metabolic liabilities with the liver-targeted DGAT2 inhibitor PF-06427878. Despite strategic placement of a nitrogen atom in the dialkoxyaromatic ring in PF-06427878 to evade oxidative O-dearylation, metabolic intrinsic clearance remained high due to extensive piperidine ring oxidation as exemplified with compound 1. Piperidine ring modifications through alternate N-linked heterocyclic ring/spacer combination led to azetidine 2 that demonstrated lower intrinsic clearance. However, 2 underwent a facile cytochrome P450 (CYP)-mediated α-carbon oxidation followed by azetidine ring scission, resulting in the formation of ketone (M2) and aldehyde (M6) as stable metabolites in NADPH-supplemented human liver microsomes. Inclusion of GSH or semicarbazide in microsomal incubations led to the formation of Cys-Gly-thiazolidine (M3), Cys-thiazolidine (M5), and semicarbazone (M7) conjugates, which were derived from reaction of the nucleophilic trapping agents with aldehyde M6. Metabolites M2 and M5 were biosynthesized from NADPH- and l-cysteine-fortified human liver microsomal incubations with 2, and proposed metabolite structures were verified using one- and two-dimensional NMR spectroscopy. Replacement of the azetidine substituent with a pyridine ring furnished 8, which mitigated the formation of the electrophilic aldehyde metabolite, and was a more potent DGAT2 inhibitor than 2. Further structural refinements in 8, specifically introducing amide bond substituents with greater metabolic stability, led to the discovery of PF-06865571 (ervogastat) that is currently in phase 2 clinical trials for the treatment of nonalcoholic steatohepatitis.


Asunto(s)
Azetidinas , Diacilglicerol O-Acetiltransferasa , Humanos , Diacilglicerol O-Acetiltransferasa/metabolismo , Tiazolidinas/metabolismo , NADP/metabolismo , Glutatión/metabolismo , Microsomas Hepáticos/metabolismo , Piperidinas/metabolismo , Azetidinas/farmacología , Azetidinas/metabolismo , Amidas/metabolismo
2.
Int J Mol Sci ; 23(21)2022 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-36362063

RESUMEN

Siponimod (Mayzent®), a sphingosine 1-phosphate receptor (S1PR) modulator which prevents lymphocyte egress from lymphoid tissues, is approved for the treatment of relapsing-remitting and active secondary progressive multiple sclerosis. It can cross the blood-brain barrier (BBB) and selectively binds to S1PR1 and S1PR5 expressed by several cell populations of the central nervous system (CNS) including microglia. In multiple sclerosis, microglia are a key CNS cell population moving back and forth in a continuum of beneficial and deleterious states. On the one hand, they can contribute to neurorepair by clearing myelin debris, which is a prerequisite for remyelination and neuroprotection. On the other hand, they also participate in autoimmune inflammation and axonal degeneration by producing pro-inflammatory cytokines and molecules. In this study, we demonstrate that siponimod can modulate the microglial reaction to lipopolysaccharide-induced pro-inflammatory activation.


Asunto(s)
Azetidinas , Esclerosis Múltiple , Humanos , Microglía/metabolismo , Compuestos de Bencilo/farmacología , Azetidinas/farmacología , Azetidinas/metabolismo , Esclerosis Múltiple/metabolismo
3.
Biochem Biophys Res Commun ; 595: 1-6, 2022 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-35091108

RESUMEN

Farnesoid X receptor (FXR) is a bile acid-related nuclear receptor and is considered a promising target to treat several liver disorders. Cilofexor is a selective FXR agonist and has already entered phase III trials in primary sclerosing cholangitis (PSC) patients. Pruritis caused by cilofexor treatment is dose dependent. The binding characteristics of cilofexor with FXR and its pruritogenic mechanism remain unclear. In our research, the affinity of cilofexor bound to FXR was detected using an isothermal titration calorimetry (ITC) assay. The binding mechanism between cilofexor and FXR-LBD is explained by the cocrystal structure of the FXR/cilofexor complex. Structural models indicate the possibility that cilofexor activates Mas-related G protein-coupled receptor X4 (MRGPRX4) or G protein-coupled bile acid receptor 1 (GPBAR1), leading to pruritus. In summary, our analyses provide a molecular mechanism of cilofexor binding to FXR and provide a possible explanation for the dose-dependent pruritis of cilofexor.


Asunto(s)
Azetidinas/química , Ácidos Isonicotínicos/química , Simulación del Acoplamiento Molecular , Dominios Proteicos , Receptores Citoplasmáticos y Nucleares/química , Azetidinas/metabolismo , Azetidinas/farmacología , Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/metabolismo , Sitios de Unión , Unión Competitiva , Calorimetría/métodos , Cristalización , Humanos , Enlace de Hidrógeno , Ácidos Isonicotínicos/metabolismo , Ácidos Isonicotínicos/farmacología , Isoxazoles/química , Isoxazoles/metabolismo , Isoxazoles/farmacología , Ligandos , Estructura Molecular , Receptores Citoplasmáticos y Nucleares/agonistas , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo
4.
Cell Res ; 31(12): 1263-1274, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34526663

RESUMEN

Sphingosine-1-phosphate (S1P) is an important bioactive lipid molecule in cell membrane metabolism and binds to G protein-coupled S1P receptors (S1PRs) to regulate embryonic development, physiological homeostasis, and pathogenic processes in various organs. S1PRs are lipid-sensing receptors and are therapeutic targets for drug development, including potential treatment of COVID-19. Herein, we present five cryo-electron microscopy structures of S1PRs bound to diverse drug agonists and the heterotrimeric Gi protein. Our structural and functional assays demonstrate the different binding modes of chemically distinct agonists of S1PRs, reveal the mechanical switch that activates these receptors, and provide a framework for understanding ligand selectivity and G protein coupling.


Asunto(s)
Receptores de Esfingosina-1-Fosfato/agonistas , Azetidinas/química , Azetidinas/metabolismo , Compuestos de Bencilo/química , Compuestos de Bencilo/metabolismo , Microscopía por Crioelectrón , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Estructura Cuaternaria de Proteína , Transducción de Señal , Receptores de Esfingosina-1-Fosfato/genética , Receptores de Esfingosina-1-Fosfato/metabolismo
5.
Eur J Med Chem ; 219: 113418, 2021 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-33862516

RESUMEN

The occurrence of resistances in Gram negative bacteria is steadily increasing to reach extremely worrying levels and one of the main causes of resistance is the massive spread of very efficient ß-lactamases which render most ß-lactam antibiotics useless. Herein, we report the development of a series of imino-analogues of ß-lactams (namely azetidinimines) as efficient non-covalent inhibitors of ß-lactamases. Despite the structural and mechanistic differences between serine-ß-lactamases KPC-2 and OXA-48 and metallo-ß-lactamase NDM-1, all three enzymes can be inhibited at a submicromolar level by compound 7dfm, which can also repotentiate imipenem against a resistant strain of Escherichia coli expressing NDM-1. We show that 7dfm can efficiently inhibit not only the three main clinically-relevant carbapenemases of Ambler classes A (KPC-2), B (NDM-1) and D (OXA-48) with Ki's below 0.3 µM, but also the cephalosporinase CMY-2 (class C, 86% inhibition at 10 µM). Our results pave the way for the development of a new structurally original family of non-covalent broad-spectrum inhibitors of ß-lactamases.


Asunto(s)
Antibacterianos/química , Azetidinas/química , Inhibidores de beta-Lactamasas/química , beta-Lactamasas/química , Antibacterianos/metabolismo , Antibacterianos/farmacología , Azetidinas/metabolismo , Sitios de Unión , Dominio Catalítico , Línea Celular , Proliferación Celular/efectos de los fármacos , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Bacterias Gramnegativas/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Pruebas de Sensibilidad Microbiana , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad , Inhibidores de beta-Lactamasas/metabolismo , Inhibidores de beta-Lactamasas/farmacología , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
6.
J Med Chem ; 64(1): 695-710, 2021 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-33352047

RESUMEN

We optimized our previously reported proline-based STAT3 inhibitors into an exciting new series of (R)-azetidine-2-carboxamide analogues that have sub-micromolar potencies. 5a, 5o, and 8i have STAT3-inhibitory potencies (IC50) of 0.55, 0.38, and 0.34 µM, respectively, compared to potencies greater than 18 µM against STAT1 or STAT5 activity. Further modifications derived analogues, including 7e, 7f, 7g, and 9k, that addressed cell membrane permeability and other physicochemical issues. Isothermal titration calorimetry analysis confirmed high-affinity binding to STAT3, with KD of 880 nM (7g) and 960 nM (9k). 7g and 9k inhibited constitutive STAT3 phosphorylation and DNA-binding activity in human breast cancer, MDA-MB-231 or MDA-MB-468 cells. Furthermore, treatment of breast cancer cells with 7e, 7f, 7g, or 9k inhibited viable cells, with an EC50 of 0.9-1.9 µM, cell growth, and colony survival, and induced apoptosis while having relatively weaker effects on normal breast epithelial, MCF-10A or breast cancer, MCF-7 cells that do not harbor constitutively active STAT3.


Asunto(s)
Azetidinas/química , Factor de Transcripción STAT3/antagonistas & inhibidores , Amidas/química , Apoptosis/efectos de los fármacos , Azetidinas/metabolismo , Azetidinas/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , ADN/química , ADN/metabolismo , Evaluación Preclínica de Medicamentos , Humanos , Concentración 50 Inhibidora , Fosforilación/efectos de los fármacos , Unión Proteica , Factor de Transcripción STAT3/metabolismo , Relación Estructura-Actividad
7.
Cells ; 9(8)2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32722245

RESUMEN

The modulation of the sphingosine 1-phosphate receptor is an approved treatment for relapsing multiple sclerosis because of its anti-inflammatory effect of retaining lymphocytes in lymph nodes. Different sphingosine 1-phosphate receptor subtypes are expressed in the brain and spinal cord, and their pharmacological effects may improve disease development and neuropathology. Siponimod (BAF312) is a novel sphingosine 1-phosphate receptor modulator that has recently been approved for the treatment of active secondary progressive multiple sclerosis (MS). In this review article, we summarize recent evidence suggesting that the active role of siponimod in patients with progressive MS may be due to direct interaction with central nervous system cells. Additionally, we tried to summarize our current understanding of the function of siponimod and discuss the effects observed in the case of MS.


Asunto(s)
Azetidinas/metabolismo , Compuestos de Bencilo/metabolismo , Sistema Nervioso Central/fisiopatología , Esclerosis Múltiple/tratamiento farmacológico , Moduladores de los Receptores de fosfatos y esfingosina 1/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones
9.
Eur J Med Chem ; 197: 112323, 2020 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-32339854

RESUMEN

Inhibition of tubulin polymerisation with small molecules has been clinically validated as a promising therapy for multiple solid tumours. Herein, a series of chiral azetidin-2-ones were asymmetrically synthesised and biologically evaluated for antitumour activities. Among them, a chiral fluorinated azetidin-2-one, 18, was found to exhibit the most potent activities against five cancer cell lines, including a drug-resistant cell line, with IC50 values ranging from 1.0 to 3.6 nM. Further mechanistic studies revealed that the compound 18 worked by disrupting tubulin polymerisation, blocking the cell cycle in the G2/M phase, inducing cellular apoptosis, and suppressing angiogenesis. Additionally, 18 exhibited higher human-microsomal metabolic stability and aqueous solubility compared to those of combretastatin A-4. Finally, 18 was also found to effectively inhibit tumour growth in a xenograft mice model with low toxicity and thus might be a promising lead for further clinical development.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Azetidinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Moduladores de Tubulina/uso terapéutico , Tubulina (Proteína)/metabolismo , Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/toxicidad , Animales , Apoptosis/efectos de los fármacos , Azetidinas/síntesis química , Azetidinas/metabolismo , Azetidinas/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Estabilidad de Medicamentos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Ratones , Microsomas Hepáticos/metabolismo , Simulación del Acoplamiento Molecular , Estructura Molecular , Relación Estructura-Actividad , Tubulina (Proteína)/química , Moduladores de Tubulina/síntesis química , Moduladores de Tubulina/metabolismo , Moduladores de Tubulina/toxicidad , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Org Biomol Chem ; 18(16): 3069-3081, 2020 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-32101243

RESUMEN

The use of selective covalent inhibitors with low binding affinity and high reactivity with the target enzyme is a promising way to solve a long-standing problem of the "undruggable" RAS-like proteins. Specifically, compounds of the ARS family that prevent the activation of the GDP-bound G12C mutant of Kirsten RAS (KRAS) are in the focus of recent experimental research. We report the first computational characterization of the entire reaction mechanism of the covalent binding of ARS-853 to the KRASG12C·GDP complex. The application of molecular dynamics, molecular docking and quantum mechanics/molecular mechanics approaches allowed us to model the inhibitor binding to the protein and the chemical reaction of ARS-853 with Cys12 in the enzyme binding site. We estimated a full set of kinetic constants and carried out numerical kinetic analysis of the process. Thus, we were able to compare directly the physicochemical parameters of the reaction obtained in silico and the macroscopic parameters observed in experimental studies. From our computational results, we explain the observed unusual dependence of the rate constant of covalent complex formation, kobs, on the ARS concentration. The latter depends both on the non-covalent binding step with the equilibrium constant, Ki, and on the rate constant of covalent adduct formation, kinact. The calculated ratio kinact/Ki = 213 M-1 s-1 reproduces the corresponding experimental value of 250 ± 30 M-1 s-1 for the interaction of ARS-853 with KRASG12C. Electron density analysis in the reactive region demonstrates that covalent bond formation occurs efficiently according to the Michael addition mechanism, which assumes the activation of the C[double bond, length as m-dash]C bond of ARS-853 by a water molecule and Lys16 in the binding site of KRASG12C. We also refine the kinact and Ki constants of the ARS-107 compound, which shares common features with ARS-853, and show that the decrease in the kinact/Ki ratio in the case of ARS-107 is explained by changes in both Ki and kinact constants.


Asunto(s)
Azetidinas/metabolismo , Piperazinas/metabolismo , Proteínas ras/antagonistas & inhibidores , Azetidinas/farmacología , Sitios de Unión , Guanosina Difosfato/metabolismo , Humanos , Cinética , Simulación del Acoplamiento Molecular , Mutación , Piperazinas/farmacología , Prueba de Estudio Conceptual , Unión Proteica , Proteínas ras/efectos de los fármacos , Proteínas ras/genética , Proteínas ras/metabolismo
11.
Mol Psychiatry ; 25(2): 461-475, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-29728704

RESUMEN

Rodent studies indicate that ghrelin receptor blockade reduces alcohol consumption. However, no ghrelin receptor blockers have been administered to heavy alcohol drinking individuals. Therefore, we evaluated the safety, tolerability, pharmacokinetic (PK), pharmacodynamic (PD) and behavioral effects of a novel ghrelin receptor inverse agonist, PF-5190457, when co-administered with alcohol. We tested the effects of PF-5190457 combined with alcohol on locomotor activity, loss-of-righting reflex (a measure of alcohol sedative actions), and on blood PF-5190457 concentrations in rats. Then, we performed a single-blind, placebo-controlled, within-subject human study with PF-5190457 (placebo/0 mg b.i.d., 50 mg b.i.d., 100 mg b.i.d.). Twelve heavy drinkers during three identical visits completed an alcohol administration session, subjective assessments, and an alcohol cue-reactivity procedure, and gave blood samples for PK/PD testing. In rats, PF-5190457 did not interact with the effects of alcohol on locomotor activity or loss-of-righting reflex. Alcohol did not affect blood PF-5190457 concentrations. In humans, all adverse events were mild or moderate and did not require discontinuation or dose reductions. Drug dose did not alter alcohol concentration or elimination, alcohol-induced stimulation or sedation, or mood during alcohol administration. Potential PD markers of PF-5190457 were acyl-to-total ghrelin ratio and insulin-like growth factor-1. PF-5190457 (100 mg b.i.d.) reduced alcohol craving during the cue-reactivity procedure. This study provides the first translational evidence of safety and tolerability of the ghrelin receptor inverse agonist PF-5190457 when co-administered with alcohol. PK/PD/behavioral findings support continued research of PF-5190457 as a potential pharmacological agent to treat alcohol use disorder.


Asunto(s)
Intoxicación Alcohólica/tratamiento farmacológico , Azetidinas/farmacología , Receptores de Ghrelina/agonistas , Compuestos de Espiro/farmacología , Adulto , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Consumo de Bebidas Alcohólicas/metabolismo , Alcoholismo/tratamiento farmacológico , Alcoholismo/metabolismo , Animales , Azetidinas/metabolismo , Azetidinas/farmacocinética , Etanol/química , Femenino , Ghrelina/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Ratas , Ratas Wistar , Receptores de Ghrelina/metabolismo , Proyectos de Investigación , Método Simple Ciego , Compuestos de Espiro/metabolismo , Compuestos de Espiro/farmacocinética
13.
J Chromatogr B Analyt Technol Biomed Life Sci ; 1130-1131: 121820, 2019 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-31670107

RESUMEN

PF-5190457 is a selective and potent ghrelin receptor inverse agonist presently undergoing clinical trials to treat alcohol use disorder (AUD). We describe the development and validation of a selective and sensitive liquid chromatography-tandem mass spectrometry-based method for quantification of PF-5190457 and its recently discovered hydroxy metabolite PF-6870961 in human plasma. Analytes were extracted after simple protein precipitation using methanol (2.5 ng mL-1 tacrine as an internal standard). A gradient liquid chromatography method was used to separate the analytes on an Acquity UPLC BEH C18 analytical column. The separation was achieved at a flow rate of 0.25 mL min-1 and the total chromatographic runtime was 11.30 min. Positive electrospray ionization and multiple reaction monitoring mode were used for the quantification of all the analytes. The calibration curves from six validation runs were linear with a correlation coefficient of ≥0.996 for the concentration range of 1-1000 ng mL-1 and 2-250 ng mL-1 for PF-5190457 and PF-6870961, respectively. The retention time for PF-5190457, PF-6870961 and tacrine were 4.4, 3.8, and 4.6 min, respectively. The lower limit of quantification for PF-5190457 and PF-6870961 was 1 and 2 ng mL-1, respectively. The inter-assay precision and accuracy results obtained were within the Food and Drug Administration recommended ±15% limit of nominal values. All the analytes were found to be stable under varied stability conditions. The recovery of PF-5190457 and PF-6870961 ranged from 95 to 103%. Further, the application of the method was demonstrated by measuring the concentration of PF-5190457 and its hydroxy metabolite in patient plasma samples from 100 mg dose.


Asunto(s)
Azetidinas/sangre , Cromatografía Líquida de Alta Presión/métodos , Compuestos de Espiro/sangre , Espectrometría de Masas en Tándem/métodos , Adulto , Azetidinas/química , Azetidinas/metabolismo , Azetidinas/farmacocinética , Femenino , Humanos , Límite de Detección , Modelos Lineales , Masculino , Receptores de Ghrelina/agonistas , Reproducibilidad de los Resultados , Compuestos de Espiro/química , Compuestos de Espiro/metabolismo , Compuestos de Espiro/farmacocinética
14.
Drug Metab Dispos ; 47(11): 1247-1256, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31492694

RESUMEN

AZD1979 [(3-(4-(2-oxa-6-azaspiro[3.3]heptan-6-ylmethyl)phenoxy)azetidin-1-yl)(5-(4-methoxyphenyl)-1,3,4-oxadiazol-2-yl)methanone] is a melanin-concentrating hormone receptor 1 antagonist designed for the treatment of obesity. In this study, metabolite profiles of AZD1979 in human hepatocytes revealed a series of glutathione-related metabolites, including the glutathionyl, cysteinyl, cysteinylglycinyl, and mercapturic acid conjugates. The formation of these metabolites was not inhibited by coincubation with the cytochrome P450 (P450) inhibitor 1-aminobenzotriazole. In efforts to identify the mechanistic features of this pathway, investigations were performed to characterize the structure of the glutathionyl conjugate M12 of AZD1979 and to identify the enzyme system catalyzing its formation. Studies with various human liver subcellular fractions established that the formation of M12 was NAD(P)H-independent and proceeded in cytosol and S9 fractions but not in microsomal or mitochondrial fractions. The formation of M12 was inhibited by ethacrynic acid, an inhibitor of glutathione S-transferases (GSTs). Several human recombinant GSTs, including GSTA1, A2-2, M1a, M2-2, T1-1, and GST from human placenta, were incubated with AZD1979. All GSTs tested catalyzed the formation of M12, with GSTA2-2 being the most efficient. Metabolite M12 was purified from rat liver S9 incubations and its structure elucidated by NMR. These results establish that M12 is the product of the GST-catalyzed glutathione attack on the carbon atom α to the nitrogen atom of the strained spiro-azetidinyl moiety to give, after ring opening, the corresponding amino-thioether conjugate product, a direct conjugation pathway that occurs without the prior substrate bioactivation by P450. SIGNIFICANCE STATEMENT: The investigated compound, AZD1979, contains a 6-substituted-2-oxa-6-azaspiro[3.3]heptanyl derivative that is an example of strained heterocycles, including spiro-fused ring systems, that are widely used in synthetic organic chemistry. An unusual azetidinyl ring-opening reaction involving a nucleophilic attack by glutathione, which does not involve prior cytochrome P450-catalyzed bioactivation of the substrate and which is catalyzed by glutathione transferases, is reported. We propose a mechanism involving the protonated cyclic aminyl intermediate that undergoes nucleophilic attack by glutathione thiolate anion in this reaction, catalyzed by glutathione transferases.


Asunto(s)
Azetidinas/metabolismo , Glutatión Transferasa/fisiología , Oxadiazoles/metabolismo , Activación Metabólica , Catálisis , Cromatografía Líquida de Alta Presión , Glutatión/metabolismo , Humanos , Hígado/metabolismo , Espectroscopía de Resonancia Magnética , Espectrometría de Masas en Tándem
15.
Bioorg Med Chem Lett ; 29(16): 2090-2093, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31311734

RESUMEN

Phenolic groups are responsible for the high clearance and low oral bioavailability of the estrogen receptor alpha (ERα) clinical candidate GDC-0927. An exhaustive search for a backup molecule with improved pharmacokinetic (PK) properties identified several metabolically stable analogs, although in general at the expense of the desired potency and degradation efficiency. C-8 hydroxychromene 30 is the first example of a phenol-containing chromene that not only maintained excellent potency but also exhibited 10-fold higher oral exposure in rats. The improved in vivo clearance in rat was hypothesized to be the result of C-8 hydroxy group being sterically protected from glucuronide conjugation. The excellent potency underscores the possibility of replacing the presumed indispensable phenolic group at C-6 or C-7 of the chromene core. Co-crystal structures were obtained to highlight the change in key interactions and rationalize the retained potency.


Asunto(s)
Azetidinas/farmacología , Receptor alfa de Estrógeno/metabolismo , Flavonoides/farmacología , Administración Oral , Animales , Azetidinas/administración & dosificación , Azetidinas/metabolismo , Azetidinas/farmacocinética , Cristalografía por Rayos X , Descubrimiento de Drogas , Estabilidad de Medicamentos , Flavonoides/administración & dosificación , Flavonoides/metabolismo , Flavonoides/farmacocinética , Humanos , Células MCF-7 , Microsomas Hepáticos/metabolismo , Ratas , Estereoisomerismo , Relación Estructura-Actividad
16.
Eur J Pharm Sci ; 128: 18-26, 2019 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-30465817

RESUMEN

Ezetimibe (EZE) is an extensively used antihyperlipidemic drug with an important cholesterol lowering activity. It undergoes extensive first-pass metabolism to form its active glucuronide metabolite (EZEG). Both drugs exhibit complex pharmacokinetic profiles attributed mainly to repetitive enterohepatic kinetics. The aim of the present study was the investigation of EZE and EZEG pharmacokinetics (PK), through the development of a joint population pharmacokinetic model able to characterize their kinetic processes and enterohepatic recirculation simultaneously. Concentration-time data derived from a bioequivalence study in 28 healthy subjects were used for the analysis. Population PK modeling was performed on the obtained data using nonlinear mixed effect modeling approach, where different methodologies were applied for the description of the complex metabolism and recirculation processes of the two compounds. EZE and EZEG concentrations were best described by a population PK model incorporating first-pass metabolism and an enterohepatic recirculation loop, accounting for the recycling process of the two moieties. This is the first joint population pharmacokinetic model describing the kinetics of both EZE and EZEG.


Asunto(s)
Azetidinas/farmacocinética , Ezetimiba/metabolismo , Ezetimiba/farmacocinética , Glucurónidos/farmacocinética , Adulto , Azetidinas/química , Azetidinas/metabolismo , Composición de Medicamentos , Ezetimiba/sangre , Ezetimiba/química , Glucurónidos/química , Glucurónidos/metabolismo , Humanos , Hipolipemiantes/química , Hipolipemiantes/metabolismo , Hipolipemiantes/farmacocinética , Modelos Biológicos
17.
Angew Chem Int Ed Engl ; 58(10): 3178-3182, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30548135

RESUMEN

Pseudomonas aeruginosa displays an impressive metabolic versatility, which ensures its survival in diverse environments. Reported herein is the identification of rare azetidine-containing alkaloids from P. aeruginosa PAO1, termed azetidomonamides, which are derived from a conserved, quorum-sensing regulated nonribosomal peptide synthetase (NRPS) pathway. Biosynthesis of the azetidine motif has been elucidated by gene inactivation, feeding experiments, and biochemical characterization in vitro, which involves a new S-adenosylmethionine-dependent enzyme to produce azetidine 2-carboxylic acid as an unusual building block of NRPS. The mutants of P. aeruginosa unable to produce azetidomonamides had an advantage in growth at high cell density in vitro and displayed rapid virulence in Galleria mellonella model, inferring functional roles of azetidomonamides in the host adaptation. This work opens the avenue to study the biological functions of azetidomonamides and related compounds in pathogenic and environmental bacteria.


Asunto(s)
Alcaloides/metabolismo , Azetidinas/metabolismo , Proteínas Bacterianas/metabolismo , Péptido Sintasas/metabolismo , Pseudomonas aeruginosa/fisiología , Alcaloides/química , Azetidinas/química , Vías Biosintéticas , Humanos , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/química , Pseudomonas aeruginosa/metabolismo , Percepción de Quorum
18.
Drug Metab Dispos ; 46(7): 1001-1013, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29735753

RESUMEN

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.


Asunto(s)
Azetidinas/metabolismo , Compuestos de Bencilo/metabolismo , Citocromo P-450 CYP2C9/metabolismo , Citocromo P-450 CYP3A/metabolismo , Receptores de Lisoesfingolípidos/agonistas , Adolescente , Adulto , Animales , Biotransformación/fisiología , Heces , Semivida , Voluntarios Sanos , Humanos , Masculino , Ratones , Microsomas Hepáticos/metabolismo , Persona de Mediana Edad , Oxidación-Reducción , Estrés Oxidativo/fisiología , Adulto Joven
19.
Eur J Clin Pharmacol ; 74(4): 455-464, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29273968

RESUMEN

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.


Asunto(s)
Azetidinas/farmacocinética , Compuestos de Bencilo/farmacocinética , Simulación por Computador , Inhibidores del Citocromo P-450 CYP2C9/farmacología , Citocromo P-450 CYP2C9/genética , Fluconazol/farmacología , Microsomas Hepáticos/enzimología , Modelos Biológicos , Variantes Farmacogenómicas , Polimorfismo Genético , Azetidinas/metabolismo , Compuestos de Bencilo/metabolismo , Citocromo P-450 CYP2C9/metabolismo , Interacciones Farmacológicas , Genotipo , Humanos , Farmacogenética , Fenotipo , Programas Informáticos
20.
Biochemistry ; 57(1): 117-135, 2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29039929

RESUMEN

Tabtoxinine-ß-lactam (TßL), also known as wildfire toxin, is a time- and ATP-dependent inhibitor of glutamine synthetase produced by plant pathogenic strains of Pseudomonas syringae. Here we demonstrate that recombinant glutamine synthetase from Escherichia coli phosphorylates the C3-hydroxyl group of the TßL 3-(S)-hydroxy-ß-lactam (3-HßL) warhead. Phosphorylation of TßL generates a stable, noncovalent enzyme-ADP-inhibitor complex that resembles the glutamine synthetase tetrahedral transition state. The TßL ß-lactam ring remains intact during enzyme inhibition, making TßL mechanistically distinct from traditional ß-lactam antibiotics such as penicillin. Our findings could enable the design of new 3-HßL transition state inhibitors targeting enzymes in the ATP-dependent carboxylate-amine ligase superfamily with broad therapeutic potential in many disease areas.


Asunto(s)
Adenosina Trifosfato/metabolismo , Azetidinas/farmacología , Toxinas Bacterianas/farmacología , Proteínas de Escherichia coli/antagonistas & inhibidores , Escherichia coli/enzimología , Glutamato-Amoníaco Ligasa/antagonistas & inhibidores , Azetidinas/aislamiento & purificación , Azetidinas/metabolismo , Toxinas Bacterianas/biosíntesis , Toxinas Bacterianas/aislamiento & purificación , Catálisis , Cromatografía Liquida , Escherichia coli/efectos de los fármacos , Escherichia coli/crecimiento & desarrollo , Espectrometría de Masas , Pruebas de Sensibilidad Microbiana , Resonancia Magnética Nuclear Biomolecular , Fosforilación , Pseudomonas syringae/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...