Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 209
Filtrar
1.
Br J Clin Pharmacol ; 88(10): 4540-4551, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35508762

RESUMO

AIMS: The absorption, metabolism and excretion of opicapone (2,5-dichloro-3-(5-[3,4-dihydroxy-5-nitrophenyl]-1,2,4-oxadiazol-3-yl)-4,6-dimethylpyridine 1-oxide), a selective catechol-O-methyltransferase inhibitor, were investigated. METHODS: Plasma, urine and faeces were collected from healthy male subjects following a single oral dose of 100 mg [14 C]-opicapone. The mass balance of [14 C]-opicapone and metabolic profile were evaluated. RESULTS: The recovery of total administered radioactivity averaged >90% after 144 hours. Faeces were the major route of elimination, representing 70% of the administered dose; 5% and 20% were excreted in urine and expired air, respectively. The Cmax of total radioactivity matched that of unchanged opicapone, whereas the total radioactivity remained quantifiable for a longer period, attributed to the contribution of opicapone metabolites, involving primarily 3-O-sulfate conjugation (58.6% of total circulating radioactivity) at the nitrocatechol ring. Other circulating metabolites, accounting for <10% of the radioactivity exposure, were formed by glucuronidation, methylation, N-oxide reduction and gluthatione conjugation. Additionally, various other metabolites resulting from combinations with the opicapone N-oxide reduced form at the 2,5-dichloro-4,6-dimethylpyridine 1-oxide moiety, including nitro reduction and N-acetylation, reductive opening and cleavage of the 1,2,4-oxadiazole ring and the subsequent hydrolysis products were identified, but only in faeces, suggesting the involvement of gut bacteria. CONCLUSION: [14 C]-opicapone was fully excreted through multiple metabolic pathways. The main route of excretion was in faeces, where opicapone may be further metabolized via reductive metabolism involving the 1,2,4-oxadiazole ring-opening and subsequent hydrolysis.


Assuntos
Inibidores de Catecol O-Metiltransferase , Oxidiazóis , Administração Oral , Inibidores de Catecol O-Metiltransferase/farmacocinética , Fezes , Voluntários Saudáveis , Humanos , Masculino , Oxidiazóis/farmacocinética
2.
Org Biomol Chem ; 20(5): 1041-1052, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-35029272

RESUMO

Assessment of sphingosine-1-phosphate receptor 1 (S1PR1) expression could be a unique tool to determine the neuroinflammatory status for central nervous system (CNS) disorders. Our preclinical results indicate that PET imaging with [11C]CS1P1 radiotracer can quantitatively measure S1PR1 expression changes in different animal models of inflammatory diseases. Here we developed a multiple step F-18 labeling strategy to synthesize the radiotracer [18F]FS1P1, sharing the same structure with [11C]CS1P1. We explored a wide range of reaction conditions for the nucleophilic radiofluorination starting with the key ortho-nitrobenzaldehyde precursor 10. The tertiary amine additive TMEDA proved crucial to achieve high radiochemical yield of ortho-[18F]fluorobenzaldehyde [18F]12 starting with a small amount of precursor. Based on [18F]12, a further four-step modification was applied in one-pot to generate the target radiotracer [18F]FS1P1 with 30-50% radiochemical yield, >95% chemical and radiochemical purity, and a high molar activity (37-166.5 GBq µmol-1, decay corrected to end of synthesis, EOS). Subsequently, tissue distribution of [18F]FS1P1 in rats showed a high brain uptake (ID% g-1) of 0.48 ± 0.06 at 5 min, and bone uptake of 0.27 ± 0.03, 0.11 ± 0.02 at 5, and 120 min respectively, suggesting no in vivo defluorination. MicroPET studies showed [18F]FS1P1 has high macaque brain uptake with a standard uptake value (SUV) of ∼2.3 at 120 min. Radiometabolite analysis of macaque plasma samples indicated that [18F]FS1P1 has good metabolic stability, and no major radiometabolite confounded PET measurements of S1PR1 in nonhuman primate brain. Overall, [18F]FS1P1 is a promising F-18 S1PR1 radiotracer worthy of further clinical investigation for human use.


Assuntos
Oxidiazóis/química , Compostos Radiofarmacêuticos/química , Receptores de Esfingosina-1-Fosfato/metabolismo , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Feminino , Radioisótopos de Flúor/química , Humanos , Marcação por Isótopo , Macaca , Masculino , Oxidiazóis/síntese química , Oxidiazóis/farmacocinética , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/farmacocinética , Ratos Sprague-Dawley
3.
Pharmacol Res Perspect ; 10(1): e00891, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34939338

RESUMO

Opicapone (2,5-dichloro-3-(5-(3,4-dihydroxy-5-nitrophenyl)-1,2,4-oxadiazol-3-yl)-4,6-dimethylpyridine 1-oxide) is a selective catechol-O-methyltransferase inhibitor that has been granted marketing authorization in Europe, Japan, and United States. The present work describes the metabolism and disposition of opicapone in the rat obtained in support to its development and regulatory filling. Plasma levels and elimination of total radioactivity were determined after oral and intravenous administration of [14 C]-opicapone. The maximum plasma concentrations of opicapone-related radioactivity were reached at early time points followed by a gradual return to baseline with a biphasic elimination. Fecal excretion was the primary route of elimination of total radioactivity. Quantitative distribution of drug-related radioactivity demonstrated that opicapone and related metabolites did not distribute to the central nervous system. Opicapone was extensively metabolized in rats resulting in more than 20 phase I and phase II metabolites. Although O-glucuronidation, -sulfation, and -methylation of the nitrocatechol moiety were the principal metabolic pathways, small amount of the N-acetyl derivative was detected, as a result of reduction of the nitro group and subsequent conjugation. Other metabolic transformations included N-oxide reduction to the pyridine derivative and reductive cleavage of 1,2,4-oxadiazole ring followed by further conjugative reactions. Reaction phenotyping studies suggested that SULT 1A1*1 and *2 and UGT1A7, UGT1A8, UGT1A9, and UGT1A10 may be involved in opicapone sulfation and glucuronidation, respectively. However, the reductive metabolic pathways mediated by gut microflora cannot be excluded. Opicapone, in the rat, was found to be rapidly absorbed, widely distributed to peripheric tissues, metabolized mainly via conjugative pathways at the nitro catechol ring, and primarily excreted via feces.


Assuntos
Inibidores de Catecol O-Metiltransferase/farmacocinética , Oxidiazóis/farmacocinética , Administração Intravenosa , Administração Oral , Animais , Arilsulfotransferase/metabolismo , Inibidores de Catecol O-Metiltransferase/administração & dosagem , Glucuronosiltransferase/metabolismo , Masculino , Oxidiazóis/administração & dosagem , Fenótipo , Ratos , Ratos Wistar , Distribuição Tecidual
4.
J Med Chem ; 64(16): 12397-12413, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34387471

RESUMO

The orphan receptor GPR88 has been implicated in a number of striatal-associated disorders, yet its endogenous ligand has not been discovered. We have previously reported that the amine functionality in the 2-AMPP-derived GPR88 agonists can be replaced with an amide (e.g., 4) without losing activity. Later, we have found that the amide can be replaced with a bioisosteric 1,3,4-oxadiazole with improved potency. Here, we report a further study of amide bioisosteric replacement with a variety of azoles containing three heteroatoms, followed by a focused structure-activity relationship study, leading to the discovery of a series of novel 1,4-disubstituted 1H-1,2,3-triazoles as GPR88 agonists. Collectively, our medicinal chemistry efforts have resulted in a potent, efficacious, and brain-penetrant GPR88 agonist 53 (cAMP EC50 = 14 nM), which is a suitable probe to study GPR88 functions in the brain.


Assuntos
Benzenoacetamidas/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Triazóis/farmacologia , Animais , Benzenoacetamidas/síntese química , Benzenoacetamidas/farmacocinética , Barreira Hematoencefálica/metabolismo , Corpo Estriado/metabolismo , Desenho de Fármacos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estrutura Molecular , Oxidiazóis/síntese química , Oxidiazóis/farmacocinética , Oxidiazóis/farmacologia , Receptores Acoplados a Proteínas G/deficiência , Relação Estrutura-Atividade , Triazóis/síntese química , Triazóis/farmacocinética
5.
Drug Metab Dispos ; 49(8): 601-609, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34011531

RESUMO

Ozanimod, recently approved for treating relapsing multiple sclerosis, produced a disproportionate, active, MAO B-catalyzed metabolite (CC112273) that showed remarkable interspecies differences and led to challenges in safety testing. This study explored the kinetics of CC112273 formation from its precursor RP101075. Incubations with human liver mitochondrial fractions revealed K Mapp, V max, and intrinsic clearance (Clint) for CC112273 formation to be 4.8 µM, 50.3 pmol/min/mg protein, and 12 µl/min/mg, respectively, whereas Michaelis-Menten constant (K M) with human recombinant MAO B was 1.1 µM. Studies with liver mitochondrial fractions from preclinical species led to K Mapp, V max, and Clint estimates of 3.0, 35, and 33 µM, 80.6, 114, 37.3 pmol/min/mg, and 27.2, 3.25, and 1.14 µl/min/mg in monkey, rat, and mouse, respectively, and revealed marked differences between rodents and primates, primarily attributable to differences in the K M Comparison of Clint estimates revealed monkey to be ∼2-fold more efficient and the mouse and rat to be 11- and 4-fold less efficient than humans in CC112273 formation. The influence of stereochemistry on MAO B-mediated oxidation was also investigated using the R-isomer of RP101075 (RP101074). This showed marked selectivity toward catalysis of the S-isomer (RP101075) only. Docking into MAO B crystal structure suggested that although both the isomers occupied its active site, only the orientation of RP101075 presented the C-H on the α-carbon that was ideal for the C-H bond cleavage, which is a requisite for oxidative deamination. These studies explain the basis for the observed interspecies differences in the metabolism of ozanimod as well as the substrate stereospecificity for formation of CC112273. SIGNIFICANCE STATEMENT: This study evaluates the enzymology and the species differences of the major circulating metabolite of ozanimod, CC112273. Additionally, the study also explores the influence of stereochemistry on MAO B-catalyzed reactions. The study is of significance to the DMD readers given that this oxidation is catalyzed by a non-cytochrome P450 enzyme, and that marked species difference and notable stereospecificity was observed in MAO B-catalyzed biotransformation when the indaneamine enantiomers were used as substrates.


Assuntos
Indanos/farmacocinética , Monoaminoxidase/metabolismo , Oxidiazóis/farmacocinética , Animais , Biotransformação , Desaminação , Avaliação Pré-Clínica de Medicamentos , Haplorrinos , Humanos , Indanos/sangue , Taxa de Depuração Metabólica , Camundongos , Mitocôndrias Hepáticas/metabolismo , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Oxidiazóis/sangue , Oxirredução , Ratos , Especificidade da Espécie , Moduladores do Receptor de Esfingosina 1 Fosfato/sangue , Moduladores do Receptor de Esfingosina 1 Fosfato/farmacocinética , Estereoisomerismo
6.
Clin Pharmacol Drug Dev ; 10(11): 1316-1324, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33864709

RESUMO

Opicapone (OPC) is a third-generation catechol-O-methyltransferase inhibitor developed to treat Parkinson disease and motor fluctuations. This open-label, single-center, phase 1 study aimed to evaluate the pharmacokinetics (PK) of OPC and its metabolites when administered as single and multiple doses in healthy White and Chinese subjects. The study enrolled a total of 30 White and Chinese healthy subjects, equally balanced among groups. The first dose of OPC was administered orally as a single dose of 50 mg on day 1, followed by a 10-day once-daily treatment from day 5 to day 14. Plasma concentrations of OPC and its metabolites were measured at 0 to 72 and 0 to 144 hours after dosing for single dose and multiple dose, respectively. Moreover, urine concentrations of OPC and its metabolite were measured 0 to 24 hours after dosing. PK parameters were derived from noncompartmental analysis. Geometric mean ratios and 90% confidence intervals for the main PK parameters were conducted to evaluate the ethnic difference between White and Chinese subjects. The plasma and urine exposure of OPC and its metabolites in Chinese subjects were similar to those in White subjects. These results indicated that ethnicity had no significant impact on PK of OPC between White and Chinese subjects.


Assuntos
Povo Asiático , Inibidores de Catecol O-Metiltransferase/farmacocinética , Oxidiazóis/farmacocinética , População Branca , Adulto , Inibidores de Catecol O-Metiltransferase/sangue , Inibidores de Catecol O-Metiltransferase/urina , China , Feminino , Voluntários Saudáveis , Humanos , Masculino , Oxidiazóis/sangue , Oxidiazóis/urina
7.
CNS Drugs ; 35(3): 331-344, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33755924

RESUMO

BACKGROUND: Vafidemstat, an inhibitor of the histone lysine-specific demethylase KDM1A, corrects cognition deficits and behavior alterations in rodent models. Here, we report the results from the first-in-human trial of vafidemstat in healthy young and older adult volunteers. A total of 110 volunteers participated: 87 were treated with vafidemstat and 23 with placebo. OBJECTIVES: The study aimed to determine the safety and tolerability of vafidemstat, to characterize its pharmacokinetic and pharmacodynamic profiles, to assess its central nervous system (CNS) exposure, and to acquire the necessary data to select the appropriate doses for long-term treatment of patients with CNS disease in phase II trials. METHODS: This single-center, randomized, double-blind, placebo-controlled phase I trial included a single and 5-day repeated dose-escalation and open-label CNS penetration substudy. Primary outcomes were safety and tolerability; secondary outcomes included analysis of the pharmacokinetics and pharmacodynamics, including chemoprobe-based immune analysis of KDM1A target engagement (TE) in peripheral blood mononuclear cells (PBMCs) and platelet monoamine oxidase B (MAOB) inhibition. CNS and cognitive function were also evaluated. RESULTS: No severe adverse events (AEs) were reported in the dose-escalation stage. AEs were reported at all dose levels; none were dose dependent, and no significant differences were observed between active treatment and placebo. Biochemistry, urinalysis, vital signs, electrocardiogram, and hematology did not change significantly with dose escalation, with the exception of a transient reduction of platelet counts in an extra dose level incorporated for that purpose. Vafidemstat exhibits rapid oral absorption, approximate dose-proportional exposures, and moderate systemic accumulation after 5 days of treatment. The cerebrospinal fluid-to-plasma unbound ratio demonstrated CNS penetration. Vafidemstat bound KDM1A in PBMCs in a dose-dependent manner. No MAOB inhibition was detected. Vafidemstat did not affect the CNS or cognitive function. CONCLUSIONS: Vafidemstat displayed good safety and tolerability. This phase I trial confirmed KDM1A TE and CNS penetration and permitted characterization of platelet dynamics and selection of phase IIa doses. TRIAL REGISTRATION: EUDRACT No. 2015-003721-33, filed 30 October 2015.


Assuntos
Histona Desmetilases/antagonistas & inibidores , Oxidiazóis/farmacocinética , Oxidiazóis/uso terapêutico , Área Sob a Curva , Sistema Nervoso Central/efeitos dos fármacos , Método Duplo-Cego , Feminino , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Masculino , Inibidores da Monoaminoxidase/farmacocinética , Inibidores da Monoaminoxidase/farmacologia
8.
Eur J Pharmacol ; 899: 174011, 2021 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-33705803

RESUMO

Forkhead transcription factor forkhead box O1 (FoxO1) plays an important role in glucose and lipid metabolism, contributing to the pathogenesis of metabolic disorders. This study aimed to discover a novel FoxO1 inhibitor as a potential new anti-diabetic drug candidate, and describes the biological effects of JY-2, 5-(2,4-dichlorophenyl)-3-(pyridin-2-yl)-1,2,4-oxadiazole in vitro and in vivo. JY-2 inhibited FoxO1 transcriptional activity in a concentration-dependent manner, with an IC50 value of 22 µM. The inhibitory effects of JY-2 on FoxO3a and FoxO4 appeared to be weaker than that on FoxO1. Consistent with its inhibitory effect on FoxO1, JY-2 reduced the palmitic acid (PA)-stimulated mRNA expression of glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK), two key enzymes involved in gluconeogenesis in HepG2 cells. In association with the reduced expression of lipid metabolism genes, triglyceride accumulation was also reduced by JY-2, as determined by Oil Red O staining. In addition, JY-2 restored PA-impaired glucose-stimulated insulin secretion (GSIS), in conjunction with an increased mRNA expression of PDX1, MafA, and insulin in INS-1 cells. The in vivo efficacy of JY-2 was examined using C57BL/6J, db/db, and high fat-diet induced obese and diabetic (DIO) mice models, and showed that JY-2 improved glucose tolerance, in parallel with a reduced mRNA expression of gluconeogenic genes. Pharmacokinetic analysis revealed that JY-2 exhibited excellent oral bioavailability (98%), with little adverse effects. These results demonstrated that the novel FoxO1 inhibitor, JY-2, may exert beneficial anti-diabetic effects and that it warrants further investigation as a novel anti-diabetic drug candidate.


Assuntos
Diabetes Mellitus/tratamento farmacológico , Gluconeogênese/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Oxidiazóis/farmacologia , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Diabetes Mellitus/enzimologia , Diabetes Mellitus/patologia , Dieta Hiperlipídica , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica , Células Hep G2 , Hepatócitos/enzimologia , Hepatócitos/patologia , Humanos , Hipoglicemiantes/farmacocinética , Masculino , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Obesidade/complicações , Oxidiazóis/farmacocinética , Ácido Palmítico/toxicidade , Ratos , Transdução de Sinais
9.
J Clin Pharmacol ; 61(1): 82-89, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32656870

RESUMO

Half-life is a standard result reported with analysis of pharmacokinetic data. Different definitions such as noncompartmental half-life, terminal half-life, effective half-life, and context-sensitive half-life can yield substantially different estimates of the quantity "half-life." Time to attainment of steady-state conditions is generally derived from (terminal) half-life and therefore sensitive toward the definition of half-life. Thus, estimates of the time to attain steady state must be provided with a precise definition of steady state and the method used for estimation, particularly for drugs with long (terminal) half-life. For clinical purposes, terminal half-life can have limited relevance if drug concentrations in the terminal elimination phase are low. A general rule for which half-life to use is infeasible. While limited accumulation can be negligible if a plateau in pharmacokinetics/pharmacodynamics is reached or with a wide therapeutic window (ie, exposure range), small additional drug accumulation can be highly relevant for drugs with a narrow therapeutic window. Beyond the average, estimation of individual time to attainment of steady state can add highly relevant information about the variability between subjects. Simulations from population models and the use of different definitions of steady state provide an assessment of robustness of the results. The relevance of accurate estimation of time to attainment of steady state is illustrated with cenerimod, an sphingosine-1-phosphate 1 receptor modulator with long half-life currently in clinical development for which estimates of time to steady state ranged from 35 to 110 days with different calculations.


Assuntos
Meia-Vida , Farmacocinética , Humanos , Oxidiazóis/farmacocinética , Propilenoglicóis/farmacocinética , Receptores de Esfingosina-1-Fosfato/efeitos dos fármacos , Fatores de Tempo
10.
Clin Transl Sci ; 14(1): 143-147, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32860737

RESUMO

Cenerimod is a sphingosine-1-phosphate 1 receptor (S1P1 R) modulator in phase II development for treatment of systemic lupus erythematosus. Its pharmacokinetics (PKs), pharmacodynamics (PDs), as well as safety and tolerability were investigated in white and Asian subjects to allow for recruitment of Asian patients in future studies. A randomized, double-blind, placebo-controlled parallel-group study was performed in 20 healthy male subjects (n = 10 per ethnicity). A single, oral dose of 4 mg cenerimod or placebo (ratio 8:2) was administered under fasted conditions. The PKs of cenerimod were similar in white and Asian subjects indicated by geometric mean ratios (90% confidence interval) of 0.99 (0.80-1.21) for maximum plasma concentration, 0.96 (0.75-1.24) for area under the plasma concentration-time curve from 0 to infinity, and 1.04 (0.86-1.25) for terminal half-life. Accordingly, the extent and time course of reduction in lymphocyte count (as PD biomarker) were also similar in white and Asian subjects as compared with placebo. As observed for other S1PR modulators, a transient mean (SD) heart rate reduction in white (15.1 (14.8) bpm) and Asian (11.8 (6.16) bpm) subjects was observed following administration of cenerimod. The drug was safe and well-tolerated indicated by occurrence of a single adverse event of chemical conjunctivitis in a white subject that was not suspected as study drug related. In conclusion, the determined absence of any relevant PK or PD differences supports using the same doses of cenerimod in white and Asian patients in upcoming late-phase studies.


Assuntos
Oxidiazóis/farmacocinética , Propilenoglicóis/farmacocinética , Receptores de Esfingosina-1-Fosfato/metabolismo , Administração Oral , Adolescente , Adulto , Idoso , Área Sob a Curva , Asiático , Relação Dose-Resposta a Droga , Método Duplo-Cego , Meia-Vida , Voluntários Saudáveis , Frequência Cardíaca/efeitos dos fármacos , Humanos , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Oxidiazóis/administração & dosagem , Oxidiazóis/efeitos adversos , Seleção de Pacientes , Propilenoglicóis/administração & dosagem , Propilenoglicóis/efeitos adversos , População Branca , Adulto Jovem
11.
Clin Pharmacol Drug Dev ; 10(2): 173-179, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32459885

RESUMO

Opicapone, a peripheral, long-acting catechol-O-methyltransferase inhibitor has been shown to improve wearing-off phenomenon in randomized, double-blind studies. This study compared the pharmacokinetic characteristics of opicapone small-tablet and size 1 capsule formulations after single oral administration to healthy Japanese subjects. In this open-label, randomized, 2-way and 2-period crossover phase 1 study, 48 healthy male subjects (aged 20 to 45 years; body mass index, 18.5 to <30.0 kg/m2 ) were randomly assigned to 2 cohorts (n = 24 each), which were administered opicapone 25 or 50 mg in a tablet-capsule or capsule-tablet sequence under fasted conditions. Blood samples were collected for pharmacokinetic analysis before opicapone capsule/tablet administration and at regular intervals over 24 hours after administration. Compared with capsules, tablets were associated with higher Cmax and AUClast/0-∞ values. However, t1/2 and tmax values were similar with opicapone 25- and 50-mg capsules/tablets. Geometric mean ratios (tablets/capsules) of Cmax , AUClast , and AUC0-∞ were 1.24, 1.18, and 1.19, respectively, for the 25-mg dose and 1.42, 1.28, and 1.27, respectively, for the 50-mg dose. Opicapone was well tolerated, and no serious adverse events occurred. A small tablet formulation of opicapone proposed for use in Japanese clinical trials was associated with apparent greater exposure compared with the existing hard capsule formulation, which should be considered when developing opicapone for Japanese patients.


Assuntos
Inibidores de Catecol O-Metiltransferase/administração & dosagem , Oxidiazóis/administração & dosagem , Administração Oral , Adulto , Área Sob a Curva , Povo Asiático , Cápsulas , Inibidores de Catecol O-Metiltransferase/farmacocinética , Estudos Cross-Over , Relação Dose-Resposta a Droga , Meia-Vida , Humanos , Masculino , Pessoa de Meia-Idade , Oxidiazóis/farmacocinética , Comprimidos , Adulto Jovem
12.
J Labelled Comp Radiopharm ; 64(2): 65-72, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33326121

RESUMO

Understanding the metabolic transformations of a potential drug molecule is important to understanding the safety profile of a drug candidate. Liquid chromatography-mass spectrometry is a standard method for detecting metabolites in the drug discovery stage, but this can lead to an incomplete understanding of the molecule's metabolism. In this manuscript, we highlight the role radiolabeling played in determining the metabolism and in quantifying the metabolites of AZD8529, AZD7325, and AZD6280. A quantitative whole-body autoradiography study can detect covalent adducts in vivo as was the case with AZD5248 in which the compound was bound to the aorta. Ultimately another compound free of aortic binding was developed, AZD7986.


Assuntos
Desenvolvimento de Medicamentos/métodos , Cromatografia Gasosa-Espectrometria de Massas/métodos , Compostos Heterocíclicos com 2 Anéis/química , Indóis/química , Oxidiazóis/química , Animais , Radioisótopos de Carbono/química , Compostos Heterocíclicos com 2 Anéis/farmacocinética , Humanos , Indóis/farmacocinética , Oxidiazóis/farmacocinética
13.
Eur J Med Chem ; 210: 112952, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33139114

RESUMO

ALS is a rare type of progressive neurological disease with unknown etiology. It results in the gradual degeneration and death of motor neurons responsible for controlling the voluntary muscles. Identification of mutations in the superoxide dismutase (SOD) 1 gene has been the most significant finding in ALS research. SOD1 abnormalities have been associated with both familial as well as sporadic ALS cases. SOD2 is a highly inducible SOD that performs in concurrence with SOD1 to detoxify ROS. Induction of SOD2 can be obtained through activation of NF-Ò¡Bs. We previously reported that SRI-22819 increases NF-Ò¡B expression and activation in vitro, but it has poor ADME properties in general and has no oral bioavailability. Our initial studies were focused on direct modifications of SRI-22819. There were active compounds identified but no improvement in microsomal stability was observed. In this context, we focused on making more significant structural changes in the core of the molecule. Ataluren, an oxadiazole compound that promotes read-through and expression of dystrophin in patients with Duchenne muscular dystrophy, bears some structural similarity to SRI-22819. Thus, we synthesized a series of SRI-22819 and Ataluren (PTC124) hybrid compounds. Several compounds from this series exhibited improved activity, microsomal stability and lower calculated polar surface area (PSA). This manuscript describes the synthesis and biological evaluation of SRI-22819 analogs and its hybrid combination with Ataluren.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , NF-kappa B/agonistas , Esclerose Lateral Amiotrófica/metabolismo , Animais , Linhagem Celular , Humanos , Camundongos , Simulação de Acoplamento Molecular , NF-kappa B/metabolismo , Oxidiazóis/química , Oxidiazóis/farmacocinética , Oxidiazóis/farmacologia , Relação Estrutura-Atividade , Superóxido Dismutase/metabolismo
14.
CPT Pharmacometrics Syst Pharmacol ; 10(2): 119-126, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33314790

RESUMO

Ozanimod, approved by regulatory agencies in multiple countries for the treatment of adults with relapsing multiple sclerosis, is a sphingosine 1-phosphate (S1P) receptor modulator, which binds with high affinity selectively to S1P receptor subtypes 1 and 5. The relationships between plasma concentrations of ozanimod and its major active metabolites, CC112273 and CC1084037, and the QTc interval (C-QTc) from a phase I multiple-dose study in healthy subjects were analyzed using nonlinear mixed effects modeling. QTc was modeled linearly as the sum of a sex-related fixed effect, baseline, and concentration-related random effects that incorporated interindividual and residual variability. Common linear, power, and maximum effect (Emax ) functions were assessed for characterizing the relationship of QTc with concentrations. Model goodness-of-fit and performance were evaluated by standard diagnostic tools, including a visual predictive check. The placebo-corrected change from baseline in QTc (ΔΔQTc) was estimated based on the developed C-QTc model using a nonparametric bootstrapping approach. QTc was better derived using a study-specific population formula (QTcP). Among the investigated functions, an Emax function most adequately described the relationship of QTcP with concentrations. Separate models for individual analytes characterized the C-QTcP relationship better than combined analytes models. Attributing QT prolongation independently to CC1084037 or CC112273, the upper bound of the 95% confidence interval of the predicted ΔΔQTcP was ~ 4 msec at the plateau of the Emax curves. Therefore, ΔΔQTcP is predicted to remain below 10 msec at the supratherapeutic concentrations of the major active metabolites.


Assuntos
Indanos/farmacocinética , Esclerose Múltipla Recidivante-Remitente/tratamento farmacológico , Oxidiazóis/farmacocinética , Moduladores do Receptor de Esfingosina 1 Fosfato/farmacocinética , Receptores de Esfingosina-1-Fosfato/metabolismo , Administração Oral , Adulto , Estudos de Casos e Controles , Método Duplo-Cego , Eletrocardiografia/efeitos dos fármacos , Feminino , Voluntários Saudáveis , Frequência Cardíaca/efeitos dos fármacos , Humanos , Indanos/administração & dosagem , Indanos/efeitos adversos , Síndrome do QT Longo , Masculino , Oxidiazóis/administração & dosagem , Oxidiazóis/efeitos adversos , Placebos/administração & dosagem , Valor Preditivo dos Testes , Moduladores do Receptor de Esfingosina 1 Fosfato/administração & dosagem , Moduladores do Receptor de Esfingosina 1 Fosfato/efeitos adversos
15.
Bioorg Chem ; 105: 104439, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33161252

RESUMO

The development of NSAIDs/iNOS inhibitor hybrids is a new strategy for the treatment of inflammatory diseases by suppression of the overproduction of PGE2 and NO. A novel series of aryl carboximidamides 4a-g and their cyclized 3-aryl-1,2,4-oxadiazoles 5a-g counterparts derived from indomethacin 1 were synthesized. Most of the target compounds displayed lower LPS-induced NO production IC50 in RAW 264.7 cells and potent in vitro iNOS and PGE2 inhibitory activity than indomethacin. Moreover, in carrageenan-induced rat paw oedema method, most of them exhibited higher in vivo anti-inflammatory activity than the reference drug indomethacin. Notably, 4 hrs after carrageenan injection, compound 4a proved to be the most potent anti-inflammatory agent in this study, with almost two- and eight-fold more active than the reference drugs indomethacin (1) and celecoxib, respectively. Compound 4a proved to be inhibitor to LPS-induced NO production, iNOS activity and PGE2 with IC50 of 10.70 µM, 2.31 µM, and 29 nM; respectively. Compounds 4a and 5b possessed the lowest ulcerogenic liabilities (35% and 38%, respectively) compared to 1. Histopathological analysis revealed that compounds 4a and 5b demonstrated reduced degeneration and healing of ulcers. Molecular docking studies into the catalytic binding pocket of the iNOS protein receptor (PDB ID: 1r35) showed good correlation with the obtained biological results. Parameters of Lipinski's rule of five and ADMET analysis were calculated where compound 4a had reasonable drug-likeness with acceptable physicochemical properties so it could be used as promising orally absorbed anti-inflammatory therapy and entitled to be used as future template for further investigations.


Assuntos
Anti-Inflamatórios não Esteroides/síntese química , Dinoprostona/antagonistas & inibidores , Inibidores Enzimáticos/síntese química , Indometacina/química , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Oxidiazóis/síntese química , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/farmacocinética , Carragenina/química , Celecoxib/metabolismo , Relação Dose-Resposta a Droga , Edema/tratamento farmacológico , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacocinética , Humanos , Lipopolissacarídeos/química , Masculino , Camundongos , Simulação de Acoplamento Molecular , Estrutura Molecular , Óxido Nítrico/metabolismo , Oxidiazóis/administração & dosagem , Oxidiazóis/farmacocinética , Oximas/química , Células RAW 264.7 , Ratos
16.
J Med Chem ; 63(23): 14989-15012, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-33205975

RESUMO

Increasing evidence implicates the orphan G protein-coupled receptor 88 (GPR88) in a number of striatal-associated disorders. In this study, we report the design and synthesis of a series of novel (4-alkoxyphenyl)glycinamides (e.g., 31) and the corresponding 1,3,4-oxadiazole bioisosteres derived from the 2-AMPP scaffold (1) as GPR88 agonists. The 5-amino-1,3,4-oxadiazole derivatives (84, 88-90) had significantly improved potency and lower lipophilicity compared to 2-AMPP. Compound 84 had an EC50 of 59 nM in the GPR88 overexpressing cell-based cAMP assay. In addition, 84 had an EC50 of 942 nM in the [35S]GTPγS binding assay using mouse striatal membranes but was inactive in membranes from GPR88 knockout mice, even at a concentration of 100 µM. In vivo pharmacokinetic testing of 90 in rats revealed that the 5-amino-1,3,4-oxadiazole analogues may have limited brain permeability. Taken together, these results provide the basis for further optimization to develop a suitable agonist to probe GPR88 functions in the brain.


Assuntos
Glicina/análogos & derivados , Glicina/farmacologia , Oxidiazóis/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Animais , Desenho de Fármacos , Glicina/farmacocinética , Masculino , Camundongos Knockout , Estrutura Molecular , Oxidiazóis/síntese química , Oxidiazóis/farmacocinética , Ratos Long-Evans , Solubilidade , Relação Estrutura-Atividade
17.
J Med Chem ; 63(21): 12942-12956, 2020 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-33124429

RESUMO

Carboxylesterase Notum is a negative regulator of the Wnt signaling pathway. There is an emerging understanding of the role Notum plays in disease, supporting the need to discover new small-molecule inhibitors. A crystallographic X-ray fragment screen was performed, which identified fragment hit 1,2,3-triazole 7 as an attractive starting point for a structure-based drug design hit-to-lead program. Optimization of 7 identified oxadiazol-2-one 23dd as a preferred example with properties consistent with drug-like chemical space. Screening 23dd in a cell-based TCF/LEF reporter gene assay restored the activation of Wnt signaling in the presence of Notum. Mouse pharmacokinetic studies with oral administration of 23dd demonstrated good plasma exposure and partial blood-brain barrier penetration. Significant progress was made in developing fragment hit 7 into lead 23dd (>600-fold increase in activity), making it suitable as a new chemical tool for exploring the role of Notum-mediated regulation of Wnt signaling.


Assuntos
Inibidores Enzimáticos/química , Esterases/antagonistas & inibidores , Oxidiazóis/química , Administração Oral , Animais , Sítios de Ligação , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Cristalografia por Raios X , Desenho de Fármacos , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Esterases/metabolismo , Meia-Vida , Humanos , Concentração Inibidora 50 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microssomos Hepáticos/metabolismo , Simulação de Dinâmica Molecular , Oxidiazóis/farmacocinética , Oxidiazóis/farmacologia , Relação Estrutura-Atividade , Via de Sinalização Wnt/efeitos dos fármacos
18.
Adv Ther ; 37(12): 4944-4958, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33025342

RESUMO

INTRODUCTION: The aims of this study were to characterize the multiple-dose pharmacokinetics (PK) of ozanimod's major active metabolites (CC112273 and CC1084037) and to evaluate the pharmacodynamic and PK interactions with pseudoephedrine (PSE). METHODS: In this phase 1, single-center, randomized, double-blind, placebo-controlled study, 56 healthy adult subjects were randomized to receive either placebo or ozanimod once daily for 30 days (0.23 mg on days 1-4, 0.46 mg on days 5-7, 0.92 mg on days 8-10, and 1.84 mg on days 11-30). On day 30, a single oral dose of PSE 60 mg was co-administered with placebo or ozanimod. Maximum time-matched change in systolic blood pressure (SBP) from baseline (day 29) following PSE administration on day 30 was calculated. Plasma PK parameters for ozanimod, CC112273, CC1084037, and PSE were estimated using noncompartmental methods. RESULTS: Fifty-two subjects (92.9%) completed the study. Following multiple dosing, approximately 94% of circulating total active drug exposure was represented by ozanimod (6%), CC112273 (73%), and CC1084037 (15%). Exposures of CC112273 and CC1084037 were highly correlated. Mean maximum time-matched change from baseline for SBP was not significantly different between ozanimod + PSE and placebo + PSE. Ozanimod also had no effect on the PK of PSE. Co-administration of ozanimod with a single dose of PSE in healthy subjects was generally well tolerated. While CC112273 and CC1084037 selectively inhibited monoamine oxidase (MAO)-B in vitro, both active metabolites do not inhibit platelet MAO-B activity in vivo. CONCLUSION: Concomitant administration of ozanimod with PSE, a sympathomimetic agent, did not potentiate the effects on blood pressure. TRIAL REGISTRATION: NCT03644576.


Assuntos
Indanos/farmacocinética , Oxidiazóis/farmacocinética , Pseudoefedrina/metabolismo , Simpatomiméticos/metabolismo , Administração Oral , Adulto , Pressão Sanguínea/efeitos dos fármacos , Preparações de Ação Retardada , Método Duplo-Cego , Feminino , Voluntários Saudáveis , Humanos , Indanos/metabolismo , Masculino , Pessoa de Meia-Idade , Oxidiazóis/metabolismo , Equivalência Terapêutica , Adulto Jovem
19.
Neurochem Int ; 141: 104870, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33035603

RESUMO

Epilepsy is a common chronic neurological disease characterized by recurrent epileptic seizures. A seizure is an uncontrolled electrical activity in the brain that can cause different levels of behavior, emotion, and consciousness. One-third of patients fail to receive sufficient seizure control, even though more than fifty FDA-approved anti-seizure drugs (ASDs) are available. In this study, we attempted small molecule screening to identify potential therapeutic agents for the treatment of seizures using seizure-induced animal models. Through behavioral phenotype-based screening, 4-(2-chloro-4-fluorobenzyl)-3-(2-thienyl)-1,2,4-oxadiazol-5(4H)-one (GM-90432) was identified as a prototype. GM-90432 treatment effectively decreased seizure-like behaviors in zebrafish and mice with chemically induced seizures. These results were consistent with decreased neuronal activity through immunohistochemistry for pERK in zebrafish larvae. Additionally, electroencephalogram (EEG) analysis revealed that GM-90432 decreases seizure-specific EEG events in adult zebrafish. Moreover, we revealed the preferential binding of GM-90432 to voltage-gated Na+ channels using a whole-cell patch clamp technique. Through pharmacokinetic analysis, GM-90432 effectively penetrated the blood-brain barrier and was distributed into the brain. Taken together, we suggest that GM-90432 has the potential to be developed into a new ASD candidate.


Assuntos
Anticonvulsivantes/farmacocinética , Anticonvulsivantes/uso terapêutico , Oxidiazóis/farmacocinética , Oxidiazóis/uso terapêutico , Convulsões/tratamento farmacológico , Animais , Comportamento Animal , Barreira Hematoencefálica , Eletroencefalografia , Imuno-Histoquímica , Larva , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Programas de Rastreamento , Camundongos , Camundongos Endogâmicos ICR , Técnicas de Patch-Clamp , Convulsões/psicologia , Bibliotecas de Moléculas Pequenas , Canais de Sódio/metabolismo , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...