Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Int J Eat Disord ; 57(7): 1418-1432, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38456603

RESUMO

OBJECTIVE: Test the efficacy of the selective orexin 1 receptor (OX1R) antagonist (SO1RA) nivasorexant in an animal model of binge-eating disorder (BED) and study its dose-response relationship considering free brain concentrations and calculated OX1R occupancy. Compare nivasorexant's profile to that of other, structurally diverse SO1RAs. Gain understanding of potential changes in orexin-A (OXA) neuropeptide and deltaFosB (ΔFosB) protein expression possibly underlying the development of the binge-eating phenotype in the rat model used. METHOD: Binge-like eating of highly palatable food (HPF) in rats was induced through priming by intermittent, repeated periods of dieting and access to HPF, followed by an additional challenge with acute stress. Effects of nivasorexant were compared to the SO1RAs ACT-335827 and IDOR-1104-2408. OXA expression in neurons and neuronal fibers as well as ΔFosB and OXA-ΔFosB co-expression was studied in relevant brain regions using immuno- or immunofluorescent histochemistry. RESULTS: All SO1RAs dose-dependently reduced binge-like eating with effect sizes comparable to the positive control topiramate, at unbound drug concentrations selectively blocking brain OX1Rs. Nivasorexant's efficacy was maintained upon chronic dosing and under conditions involving more frequent stress exposure. Priming for binge-like eating or nivasorexant treatment resulted in only minor changes in OXA or ΔFosB expression in few brain areas. DISCUSSION: Selective OX1R blockade reduced binge-like eating in rats. Neither ΔFosB nor OXA expression proved to be a useful classifier for their binge-eating phenotype. The current results formed the basis for a clinical phase II trial in BED, in which nivasorexant was unfortunately not efficacious compared with placebo. PUBLIC SIGNIFICANCE: Nivasorexant is a new investigational drug for the treatment of binge-eating disorder (BED). It underwent clinical testing in a phase II proof of concept trial in humans but was not efficacious compared with placebo. The current manuscript investigated the drug's efficacy in reducing binge-like eating behavior of a highly palatable sweet and fat diet in a rat model of BED, which initially laid the foundation for the clinical trial.


Assuntos
Transtorno da Compulsão Alimentar , Modelos Animais de Doenças , Antagonistas dos Receptores de Orexina , Receptores de Orexina , Animais , Antagonistas dos Receptores de Orexina/farmacologia , Ratos , Masculino , Transtorno da Compulsão Alimentar/tratamento farmacológico , Receptores de Orexina/metabolismo , Ratos Sprague-Dawley , Orexinas/metabolismo
2.
Xenobiotica ; 54(4): 182-194, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38400854

RESUMO

1. Ponesimod is a selective modulator of the sphingosine 1-phosphate receptor 1 (S1P1) approved for the treatment of active relapsing forms of multiple sclerosis. The chemical structure of ponesimod contains a glycerol side chain which is the major target of drug metabolism in humans.2. The two major metabolic pathways give the acids M12 (-OCH2CH(OH)COOH) and M13 (-OCH2COOH). While the former results from oxidation of the terminal alcohol, the mechanism yielding the chain-shortened acid M13 is less obvious. A detailed mechanistic study with human liver microsomes and hepatocytes using ponesimod, M12 and some of the suspected intermediates revealed an unexpectedly complex pattern of enzyme-mediated and chemical reactions.3. Metabolic pathways for both acids were not independent and several of the transformations were reversible, depending on reaction conditions. Formation of M13 occurred either via initial oxidation of the secondary alcohol, or as a downstream process starting from M12.4. The phenol metabolite M32 was produced as part of several pathways. Control experiments at various pH values and in the absence of metabolising enzymes support the conclusion that its formation resulted from chemical degradation rather than from metabolic processes.


Assuntos
Microssomos Hepáticos , Oxirredução , Humanos , Microssomos Hepáticos/metabolismo , Glicerol/metabolismo , Tiazóis/metabolismo , Hepatócitos/metabolismo , Moduladores do Receptor de Esfingosina 1 Fosfato/metabolismo
3.
Xenobiotica ; 54(3): 124-137, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38358311

RESUMO

Nivasorexant was the first orexin-1 selective receptor antagonist entering clinical development. Despite encouraging preclinical evidence in animal models, a proof-of-concept trial in binge-eating patients recently failed to demonstrate its clinical utility in this population.Across species, nivasorexant clearance was driven by metabolism along seven distinct pathways, five of which were hydroxylation reactions in various locations of the molecule. The exact sites of metabolism were identified by means of mass spectrometry, the use of deuterated analogues, and finally confirmed by chemical references.CYP3A4 was the main cytochrome P450 enzyme involved in nivasorexant metabolism in vitro and accounting for about 90% of turnover in liver microsomes. Minor roles were taken by CYP2C9 and CYP2C19 but individually did not exceed 3-7%.In the rat, nivasorexant was mostly excreted via the bile after extensive metabolism, while urinary excretion was negligible. Only traces of the parent drug were detected in urine, bile, or faeces.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Humanos , Ratos , Animais , Hidrocarboneto de Aril Hidroxilases/metabolismo , Orexinas/metabolismo , Orexinas/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Hidroxilação , Citocromo P-450 CYP3A/metabolismo , Microssomos Hepáticos/metabolismo , Citocromo P-450 CYP2C19/metabolismo
4.
Xenobiotica ; 53(3): 173-183, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36809238

RESUMO

Daridorexant is a dual orexin receptor antagonist developed for the treatment of insomnia disorder and has shown improvement in sleep outcomes and daytime functioning. The present work describes its biotransformation pathways in vitro and in vivo and provides a cross-species comparison between the animal species used in preclinical safety assessments and man.Daridorexant clearance is driven by metabolism along seven distinct pathways. Metabolic profiles were characterised by downstream products while primary metabolic products were of minor importance. The metabolic pattern differed between rodent species, with the rat reflecting the human pattern better than the mouse.In rodents, daridorexant is mostly excreted via the bile after extensive metabolism while urinary excretion was negligible in the rat. Only traces of the parent drug were detected in urine, bile, or faeces.Daridorexant has three major metabolites which are well covered in these preclinical safety species. All of them retain some residual affinity towards orexin receptors. However, none of these is considered to contribute to the pharmacological effect of daridorexant as their active concentrations in the human brain are too low.


Assuntos
Antagonistas dos Receptores de Orexina , Distúrbios do Início e da Manutenção do Sono , Masculino , Ratos , Humanos , Camundongos , Animais , Imidazóis , Distúrbios do Início e da Manutenção do Sono/tratamento farmacológico , Pirrolidinas
5.
Xenobiotica ; 49(3): 284-301, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29468921

RESUMO

1. The metabolism of the prostacyclin receptor agonist selexipag (NS-304; ACT-293987) and its active metabolite MRE-269 (ACT-333679) has been investigated in liver microsomes and hepatocytes of rats, dogs, and monkeys. MRE-269 formation is the main pathway of selexipag metabolism, irrespective of species. Some interspecies differences were evident for both compounds in terms of both metabolic turnover and metabolic profiles. The metabolism of MRE-269 was slower than that of selexipag in all three species. 2. The metabolism of selexipag was also studied in bile-duct-cannulated rats and dogs after a single oral and intravenous dose of [14C]selexipag. MRE-269 acyl glucuronide was found in both rat and dog bile. Internal acyl migration reactions of MRE-269 glucuronide were identified in an experiment with the synthetic standard MRE-6001. 3. MRE-269 was the major component in the faeces of rats and dogs. In ex vivo study using rat and dog faeces, selexipag hydrolysis to MRE-269 by the intestinal microflora is considered to be a contributory factor in rats and dogs. 4. A taurine conjugate of MRE-269 was identified in rat bile sample. Overall, selexipag was eliminated via multiple routes in animals, including hydrolysis, oxidative metabolism, conjugation, intestinal deconjugation, and gut flora metabolism.


Assuntos
Acetamidas/farmacocinética , Pirazinas/farmacocinética , Acetamidas/química , Acetamidas/metabolismo , Acetatos/química , Acetatos/metabolismo , Animais , Bile/metabolismo , Líquidos Corporais/química , Cromatografia Líquida de Alta Pressão , Cães/metabolismo , Hepatócitos/metabolismo , Macaca fascicularis/metabolismo , Metaboloma , Microssomos Hepáticos/metabolismo , Pirazinas/química , Pirazinas/metabolismo , Ratos/metabolismo , Ratos Sprague-Dawley , Especificidade da Espécie
6.
Xenobiotica ; 48(2): 186-196, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28277164

RESUMO

1. This study examined the pharmacokinetics, distribution, metabolism and excretion of the selective prostacyclin receptor agonist selexipag (NS-304; ACT-293987) and its active metabolite MRE-269 (ACT-33679). The compounds were investigated following oral and/or intravenous administration to intact rats, dogs and monkeys, and bile-duct-cannulated rats and dogs. 2. After oral administration of [14C]selexipag, selexipag was well absorbed in rats and dogs with total recoveries of over 90% of the dose, mainly in the faeces. Biliary excretion was the major elimination pathway for [14C]MRE-269 as well as [14C]selexipag, while renal elimination was of little importance. [14C]Selexipag-related radioactivity was secreted into the milk in lactating rats. 3. Plasma was analysed for total radioactivity, selexipag and MRE-269 in rats and monkeys. Selexipag was negligible in rat plasma due to extensive metabolism, and MRE-269 was present in rat and monkey plasma. A species difference was clearly evident when selexipag was incubated in rat, dog and monkey plasma. 4. Total radioactivity was rapidly distributed to tissues. The highest concentrations were found in the bile duct and liver without significant accumulation or persistence, while there was limited melanin-associated binding, penetration of the blood-brain barrier and placental transfer of drug-related materials.


Assuntos
Acetamidas/farmacocinética , Anti-Hipertensivos/farmacocinética , Pirazinas/farmacocinética , Animais , Cães , Absorção Intestinal , Macaca fascicularis , Ratos , Receptores de Epoprostenol/agonistas , Especificidade da Espécie , Distribuição Tecidual
7.
Xenobiotica ; 48(7): 704-719, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28737453

RESUMO

1. The metabolism of selexipag has been studied in vivo in man and the main excreted metabolites were identified. Also, metabolites circulating in human plasma have been structurally identified and quantified. 2. The main metabolic pathway of selexipag in man is the formation of the active metabolite ACT-333679. Other metabolic pathways include oxidation and dealkylation reactions. All primary metabolites undergo subsequent hydrolysis of the sulphonamide moiety to their corresponding acids. ACT-333679 undergoes conjugation with glucuronic acid and aromatic hydroxylation to P10, the main metabolite detected in human faeces. 3. The formation of the active metabolite ACT-333679 is catalysed by carboxylesterases, while the oxidation and dealkylation reactions are metabolized by CYP2C8 and CYP3A4. CYP2C8 is the only P450 isoform catalysing the aromatic hydroxylation to P10. CYP2C8 together with CYP3A4 are also involved in the formation of several minor ACT-333679 metabolites. UGT1A3 and UGT2B7 catalyse the glucuronidation of ACT-333679. 4. The potential of selexipag to inhibit or induce cytochrome P450 enzymes or drug transport proteins was studied in vitro. Selexipag is an inhibitor of CYP2C8 and CYP2C9 and induces CYP3A4 and CYP2C9 in vitro. Also, selexipag inhibits the transporters OATP1B1, OATP1B3, OAT1, OAT3, and BCRP. However, due to its low dose and relatively low unbound exposure, selexipag has a low potential for causing drug-drug interactions.


Assuntos
Acetamidas/metabolismo , Acetamidas/farmacologia , Pirazinas/metabolismo , Pirazinas/farmacologia , Receptores de Epoprostenol/agonistas , Acetamidas/sangue , Acetamidas/química , Acetatos/farmacologia , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Esterases/antagonistas & inibidores , Esterases/metabolismo , Hepatócitos/metabolismo , Humanos , Proteínas de Membrana Transportadoras/metabolismo , Redes e Vias Metabólicas/efeitos dos fármacos , Metaboloma , Metabolômica , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , NADP/metabolismo , Pirazinas/sangue , Pirazinas/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Epoprostenol/metabolismo , Proteínas Recombinantes/metabolismo
8.
J Pharmacol Exp Ther ; 362(3): 489-503, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28663311

RESUMO

The identification of new sleep drugs poses particular challenges in drug discovery owing to disease-specific requirements such as rapid onset of action, sleep maintenance throughout major parts of the night, and absence of residual next-day effects. Robust tools to estimate drug levels in human brain are therefore key for a successful discovery program. Animal models constitute an appropriate choice for drugs without species differences in receptor pharmacology or pharmacokinetics. Translation to man becomes more challenging when interspecies differences are prominent. This report describes the discovery of the dual orexin receptor 1 and 2 (OX1 and OX2) antagonist ACT-541468 out of a class of structurally related compounds, by use of physiology-based pharmacokinetic and pharmacodynamic (PBPK-PD) modeling applied early in drug discovery. Although all drug candidates exhibited similar target receptor potencies and efficacy in a rat sleep model, they exhibited large interspecies differences in key factors determining their pharmacokinetic profile. Human PK models were built on the basis of in vitro metabolism and physicochemical data and were then used to predict the time course of OX2 receptor occupancy in brain. An active ACT-541468 dose of 25 mg was estimated on the basis of OX2 receptor occupancy thresholds of about 65% derived from clinical data for two other orexin antagonists, almorexant and suvorexant. Modeling predictions for ACT-541468 in man were largely confirmed in a single-ascending dose trial in healthy subjects. PBPK-PD modeling applied early in drug discovery, therefore, has great potential to assist in the identification of drug molecules when specific pharmacokinetic and pharmacodynamic requirements need to be met.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Descoberta de Drogas/métodos , Imidazóis/farmacocinética , Antagonistas dos Receptores de Orexina/farmacocinética , Pirrolidinas/farmacocinética , Animais , Células CHO , Cricetinae , Cricetulus , Cães , Relação Dose-Resposta a Droga , Método Duplo-Cego , Humanos , Masculino , Ratos , Ratos Wistar
9.
Xenobiotica ; 46(3): 253-67, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26337830

RESUMO

1. The metabolism of the endothelin receptor antagonist macitentan has been characterized in bile duct-cannulated rats and dogs. 2. In both species, macitentan was metabolized along five primary pathways, i.e. conjugation with glucose (M9), oxidative depropylation (M6), aliphatic hydroxylation (M7), oxidative cleavage of the ethylene glycol linker (M4) and hydrolysis of the sulfamide moiety (M3). Most of the primary metabolites underwent subsequent biotransformation including conjugation with glucuronic acid or glucose, hydrolysis of the sulfamide group or secondary oxidation of the ethylene glycol moiety. 3. Though there were species differences in their relative importance, all metabolic pathways were present in rat and dog. The depropylated M6 was the only metabolite present in plasma of both species. 4. Metabolism was a prerequisite for macitentan excretion as relevant amounts of parent drug were neither detected in bile nor urine. Biliary excretion was the major elimination pathway, while renal elimination was of little importance.


Assuntos
Antagonistas dos Receptores de Endotelina/farmacocinética , Pirimidinas/farmacocinética , Sulfonamidas/farmacocinética , Animais , Ductos Biliares/metabolismo , Biotransformação , Cromatografia Líquida de Alta Pressão , Cães , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Antagonistas dos Receptores de Endotelina/urina , Etilenoglicol/metabolismo , Feminino , Glucose/metabolismo , Hepatócitos/metabolismo , Hidroxilação , Masculino , Redes e Vias Metabólicas , Microssomos Hepáticos/metabolismo , Pirimidinas/urina , Ratos , Ratos Wistar , Sulfonamidas/urina
10.
Bioorg Med Chem Lett ; 25(9): 1884-91, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25838147

RESUMO

Starting from advanced pyrrolidin-2-one lead compounds, this novel series of small-molecule orexin receptor antagonists was further optimized by fine-tuning of the C-3 substitution at the γ-lactam ring. We discuss our design to align in vitro potency with metabolic stability and improved physicochemical/pharmacokinetic properties while avoiding P-glycoprotein-mediated efflux. These investigations led to the identification of the orally active 3-hydroxypyrrolidin-2-one 46, a potent and selective orexin-2 receptor antagonist, that achieved good brain exposure and promoted physiological sleep in rats.


Assuntos
Antagonistas dos Receptores de Orexina/farmacologia , Receptores de Orexina/metabolismo , Pirrolidinonas/farmacologia , Sono/efeitos dos fármacos , Administração Oral , Animais , Relação Dose-Resposta a Droga , Humanos , Lactamas/administração & dosagem , Lactamas/farmacologia , Estrutura Molecular , Antagonistas dos Receptores de Orexina/síntese química , Antagonistas dos Receptores de Orexina/química , Pirrolidinonas/síntese química , Pirrolidinonas/química , Ratos , Relação Estrutura-Atividade
11.
Eur J Clin Pharmacol ; 71(1): 15-23, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25323804

RESUMO

PURPOSE: Setipiprant, a selective oral CRTH2 antagonist, has been investigated for the treatment of allergic rhinitis and asthma. In vitro data showed that setipiprant has a weak induction potential on CYP3A4. An interaction at the hepatic level between setipiprant and CYP3A4 substrates was not expected even at the dosing regimen of 1,000 mg setipiprant b.i.d. due to the high plasma protein binding. However, at this dosing regimen, interactions at the gut level could not be excluded. METHODS: In this single-center, open-label study, 40 mg of simvastatin was administered orally on Day 1, and then concomitantly with setipiprant on Day 10 following 9 days of setipiprant 1,000 mg b.i.d. to 22 healthy male subjects. RESULTS: In the presence of setipiprant, the simvastatin concentration-time profile was similar to that of simvastatin alone. The concentrations of simvastatin were, however, slightly lower, resulting in a 9 % decrease in C max (geometric mean ratio (GMR) 0.91, 90 % confidence interval (CI) (0.73, 1.13)) and in a 16 % lower AUC0-∞ (GMR 0.84, 90 % CI (0.72, 0.99)). Exposure to simvastatin acid was similar when comparing simvastatin with or without setipiprant. The GMR and 90 % CI for AUC0-∞ were within the 0.8 to 1.25 limits, whereas those for C max were outside (GMR 2.73, 90 % CI (2.11, 3.53)). Moreover, the median t max of simvastatin acid occurred earlier (1.8 h) when combined compared to 3.0 h when administered alone. CONCLUSIONS: As setipiprant has little impact on simvastatin pharmacokinetics, it does not modulate CYP3A4 in a clinically relevant manner.


Assuntos
Indóis/farmacocinética , Naftalenos/farmacocinética , Sinvastatina/farmacocinética , Adulto , Citocromo P-450 CYP3A/metabolismo , Interações Medicamentosas , Humanos , Indóis/sangue , Masculino , Pessoa de Meia-Idade , Naftalenos/sangue , Receptores Imunológicos/antagonistas & inibidores , Receptores de Prostaglandina/antagonistas & inibidores , Sinvastatina/análogos & derivados , Sinvastatina/sangue , Adulto Jovem
12.
J Pharmacol Exp Ther ; 350(1): 130-43, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24769543

RESUMO

Treatment of pulmonary arterial hypertension with the endothelin receptor antagonist bosentan has been associated with transient increases in liver transaminases. Mechanistically, bosentan inhibits the bile salt export pump (BSEP) leading to an intrahepatic accumulation of cytotoxic bile salts, which eventually results in hepatocellular damage. BSEP inhibition by bosentan is amplified by its accumulation in the liver as bosentan is a substrate of organic anion-transporting polypeptide (OATP) transport proteins. The novel endothelin receptor antagonist macitentan shows a superior liver safety profile. Introduction of the less acidic sulfamide moiety and increased lipophilicity yield a hepatic disposition profile different from other endothelin receptor antagonists. Passive diffusion rather than OATP-mediated uptake is the driving force for macitentan uptake into the liver. Interaction with the sodium taurocholate cotransporting polypeptide and BSEP transport proteins involved in hepatic bile salt homeostasis is therefore limited due to the low intrahepatic drug concentrations. Evidence for this conclusion is provided by in vitro experiments in drug transporter-expressing cell lines, acute and long-term studies in rats and dogs, absence of plasma bile salt changes in healthy human volunteers after multiple dosing, and finally the liver safety profile of macitentan in the completed phase III morbidity/mortality SERAPHIN (Study with an Endothelin Receptor Antagonist in Pulmonary Arterial Hypertension to Improve Clinical Outcome) trial.


Assuntos
Transportadores de Cassetes de Ligação de ATP/efeitos dos fármacos , Ácidos e Sais Biliares/sangue , Fígado/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/efeitos dos fármacos , Pirimidinas/farmacocinética , Pirimidinas/toxicidade , Sulfonamidas/farmacocinética , Sulfonamidas/toxicidade , Simportadores/efeitos dos fármacos , Membro 11 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Animais , Bosentana , Linhagem Celular , Cricetinae , Cães , Relação Dose-Resposta a Droga , Antagonistas dos Receptores de Endotelina , Hepatócitos , Humanos , Masculino , Transportadores de Ânions Orgânicos/efeitos dos fármacos , Pirimidinas/efeitos adversos , Ratos , Sulfonamidas/efeitos adversos
13.
Bioorg Med Chem Lett ; 24(17): 4323-31, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-25113932

RESUMO

2-Phenyl-pyrimidine-4-carboxamide analogs were identified as P2Y12 antagonists. Optimization of the carbon-linked or nitrogen-linked substituent at the 6-position of the pyrimidine ring provided compounds with excellent ex vivo potency in the platelet aggregation assay in human plasma. Compound 23u met the objectives for activity, selectivity and ADMET properties.


Assuntos
Glutamatos/administração & dosagem , Glutamatos/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Antagonistas do Receptor Purinérgico P2Y/administração & dosagem , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Pirimidinas/administração & dosagem , Pirimidinas/farmacologia , Administração Oral , Disponibilidade Biológica , Relação Dose-Resposta a Droga , Glutamatos/química , Humanos , Estrutura Molecular , Pirimidinas/química , Relação Estrutura-Atividade
14.
Bioorg Med Chem Lett ; 24(4): 1201-8, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24447850

RESUMO

Starting from a thiazolidin-4-one HTS hit, a novel series of substituted lactams was identified and developed as dual orexin receptor antagonists. In this Letter, we describe our initial efforts towards the improvement of potency and metabolic stability. These investigations delivered optimized lead compounds with CNS drug-like properties suitable for further optimization.


Assuntos
Descoberta de Drogas , Lactamas/farmacologia , Antagonistas dos Receptores de Orexina , Animais , Relação Dose-Resposta a Droga , Humanos , Lactamas/química , Lactamas/metabolismo , Estrutura Molecular , Ratos , Ratos Wistar , Relação Estrutura-Atividade
15.
RSC Med Chem ; 15(1): 344-354, 2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38283232

RESUMO

Selective orexin 2 receptor antagonists (2-SORA) such as seltorexant (15) are in clinical development for the treatment of insomnia and other conditions such as depression. Herein, we report our structure-activity-relationship (SAR) optimization efforts starting from an HTS hit (1) (N-(1-((5-acetylfuran-2-yl)methyl)-1H-pyrazol-4-yl)-5-(m-tolyl)oxazole-4-carboxamide) that was derived from an unrelated in-house GPCR-agonist program. Medicinal chemistry efforts focused on the optimization of orexin 2 receptor (OX2R) antagonistic activity, stability in liver microsomes, time dependent CYP3A4 inhibition, and aqueous solubility. Compounds were assessed for their brain-penetrating potential in in vivo experiments to select the most promising compounds for our in vivo sleep model. Our lead optimization efforts led to the discovery of the potent, brain penetrating and orally active, 2-SORA (N-(1-(2-(5-methoxy-1H-pyrrolo[3,2-b]pyridin-3-yl)ethyl)-1H-pyrazol-4-yl)-5-(m-tolyl)oxazole-4-carboxamide) 43 with efficacy in a sleep model in rats comparable to 15.

16.
J Med Chem ; 67(4): 2337-2348, 2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38331429

RESUMO

The orexin system consists of two neuropeptides (orexins A and B) and two receptors (OX1 and OX2). Selective OX1 receptor antagonists (SO1RA) are gaining interest for their potential use in the treatment of CNS disorders, including substance abuse, eating, obsessive compulsive, or anxiety disorders. While blocking OX2 reduces wakefulness, the expected advantage of selectively antagonizing OX1 is the ability to achieve clinical efficacy without the promotion of sleep. Herein we report our discovery efforts starting from a dual orexin receptor antagonist and describe a serendipitous finding that triggered a medicinal chemistry program that culminated in the identification of the potent SO1RA ACT-539313. Efficacy in a rat model of schedule-induced polydipsia supported the decision to select the compound as a preclinical candidate. Nivasorexant (20) represents the first SO1RA to enter clinical development and completed a first proof of concept phase II clinical trial in binge eating disorder in 2022.


Assuntos
Neuropeptídeos , Ratos , Animais , Orexinas , Neuropeptídeos/farmacologia , Receptores de Orexina , Morfolinas , Antagonistas dos Receptores de Orexina/farmacologia , Antagonistas dos Receptores de Orexina/uso terapêutico
17.
Drug Metab Dispos ; 41(5): 1046-59, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23431113

RESUMO

Almorexant [(2R)-2-{(1S)-6, 7-dimethoxy-1-[2-(4-trifluoromethyl-phenyl)-ethyl]-3,4-dihydro-1H-isoquinolin-2-yl}-N-methyl-2-phenyl-acetamide], a tetrahydroisoquinoline derivative, is a dual orexin receptor antagonist with sleep-promoting properties in both animals and humans. This study investigated the disposition, metabolism, and elimination of almorexant in humans. After oral administration of a 200-mg dose of ¹4C-almorexant, almorexant was rapidly absorbed (Tmax = 0.8 hour), and the apparent terminal half-life (t(1/2)) was 17.8 hours. The radioactive dose was almost completely recovered with 78.0% of the administered radioactive dose found in feces and 13.5% in urine. Unchanged almorexant was not found in urine and represented 10% of the administered dose in feces. In total, 47 metabolites were identified of which 21 were shown to be present in plasma. There are four primary metabolites, the isomeric phenols M3 and M8, formed by demethylation, the aromatic isoquinolinium ion M5, formed by dehydrogenation, and M6, formed by oxidative dealkylation with loss of the phenylglycine moiety. Most of the subsequent products are formed by permutations of these primary metabolic reactions followed by conjugation of the intermediate phenols with glucuronic or sulfonic acid. The percentage of dose excreted in urine or feces for any of the metabolites did not exceed 10% of the administered radioactive dose, nor did any of the metabolites represent more than 10% of the total drug-related exposure. In conclusion, after rapid absorption, almorexant is extensively metabolized, and excretion of metabolites in feces is the predominant route of elimination in humans.


Assuntos
Acetamidas/farmacologia , Isoquinolinas/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores de Neuropeptídeos/antagonistas & inibidores , Acetamidas/administração & dosagem , Administração Oral , Adulto , Cromatografia Líquida , Meia-Vida , Humanos , Isoquinolinas/administração & dosagem , Masculino , Receptores de Orexina , Espectrometria de Massas em Tandem
18.
Pharmacol Res Perspect ; 11(5): e01143, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37800597

RESUMO

Nivasorexant, a selective orexin-1-receptor antagonist, has recently been assessed in the treatment of humans with binge-eating disorder. Herein, the inhibitory potential of nivasorexant on cytochromes P450 (CYPs) 2C9, 2C19, and 3A4 was evaluated. Human liver microsomes/recombinant CYP enzymes were evaluated in vitro. In vivo, a single-center, open-label, fixed-sequence study was performed in healthy adults to explore the effect of 100 mg nivasorexant administered twice daily (b.i.d.) on the pharmacokinetics (PK) of flurbiprofen (50 mg, CYP2C9), omeprazole (20 mg, CYP2C19), midazolam (2 mg, CYP3A4) making use of a cocktail approach. Plasma PK sampling was performed over 24 h on Day 1 (Cocktail alone), 8 (Cocktail + nivasorexant), and 15 (Cocktail + nivasorexant at steady state). Genotyping of subjects' CYPs was performed while safety and tolerability were also assessed. In vitro, nivasorexant inhibited CYP2C9, 2C19, and 3A4 in competitive inhibition assays with IC50 values of 8.6, 1.6, and 19-44 µM, respectively, while showing a significant time-dependent CYP2C19 inhibition. In 22 subjects, exposure to flurbiprofen, omeprazole, and midazolam was generally higher during concomitant single- (i.e., area under the plasma concentration-time curve [AUC] ratio increased by 1.04-, 2.05-, and 1.56-fold, respectively) and repeated-dose (i.e., AUC ratio increased by 1.47-, 6.84-, and 3.71-fold, respectively) nivasorexant administration compared with the cocktail substrates administered alone. The most frequently reported adverse event was somnolence. According to regulatory guidance, nivasorexant is classified as a moderate CYP2C19 and weak CYP3A4 inhibitor after 1 day and as a weak CYP2C9, strong CYP2C19, and moderate CYP3A4 inhibitor after 8 days of 100 mg b.i.d. administration. Clinicaltrials.gov ID: NCT05254548.


Assuntos
Flurbiprofeno , Midazolam , Adulto , Humanos , Citocromo P-450 CYP2C19/genética , Citocromo P-450 CYP2C9 , Orexinas , Inibidores do Citocromo P-450 CYP3A , Voluntários Saudáveis , Interações Medicamentosas , Sistema Enzimático do Citocromo P-450/genética , Omeprazol/farmacocinética
19.
ChemMedChem ; 18(10): e202300030, 2023 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-36892179

RESUMO

The dual orexin receptor antagonist daridorexant was approved in 2022 in the USA and EU for the treatment of insomnia. The purpose of this study was the identification of its metabolic pathways and the human cytochrome P450 (P450) enzymes involved in its biotransformation. With human liver microsomes, daridorexant underwent hydroxylation at the methyl group of the benzimidazole moiety, oxidative O-demethylation of the anisole to the corresponding phenol, and hydroxylation to a 4-hydroxy piperidinol derivative. While the chemical structures of the benzylic alcohol and the phenol proved to be products of standard P450 reactions, 1D and 2D NMR data of the latter hydroxylation product was incompatible with the initially postulated hydroxylation of the pyrrolidine ring and suggested the disappearance of the pyrrolidine ring and formation of a new 6-membered ring. Its formation is best explained by initial hydroxylation of the pyrrolidine ring in 5-position to yield a cyclic hemiaminal. Hydrolytic ring opening then results in an aldehyde that subsequently cyclizes onto one of the benzimidazole nitrogen atoms to yield the final 4-hydroxy piperidinol. The proposed mechanism was substantiated using an N-methylated analogue, which might hydrolyze to the open-chain aldehyde but cannot undergo the final cyclization step. CYP3A4 was the major P450 enzyme responsible for daridorexant metabolism, accounting for 89 % of metabolic turnover.


Assuntos
Citocromo P-450 CYP3A , Antagonistas dos Receptores de Orexina , Humanos , Citocromo P-450 CYP3A/metabolismo , Antagonistas dos Receptores de Orexina/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Pirrolidinas/farmacologia , Microssomos Hepáticos/metabolismo , Benzimidazóis/farmacologia , Fenóis/farmacologia
20.
Biochim Biophys Acta ; 1808(7): 1827-31, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21439263

RESUMO

The influence of P-glycoprotein (ABCB1) in drug resistance as well as drug absorption and disposition is an important factor to be considered during the development of new drugs. Thus, the early identification and exclusion of compounds showing a high affinity towards P-glycoprotein can help to select drug candidates. The aim of our study was to implement a label-free assay for the identification of P-glycoprotein substrates in living cells. For this approach, a multiparametric, chip-based sensor system was used to determine extracellular acidification, cell respiration and adhesion upon stimulation with P-glycoprotein substrates. Using L-MDR1 cells, a human P-glycoprotein overexpressing cell line, the influence of P-glycoprotein activity was determined for seven different compounds, demonstrating the applicability of the system for P-glycoprotein substrate identification. Effects were concentration dependent, as shown for the P-glycoprotein substrate verapamil, and were associated with cellular acidification and respiration. P-glycoprotein ATPase activation by verapamil could be described by a Michaelis-Menten type kinetic profile showing saturation at high substrate concentrations. The Michaelis-Menten constants K(M) were determined to be 0.92µM (calculated based on extracellular acidification) and 4.9µM (calculated based on cellular respiration). Control experiments using 100nM of the P-glycoprotein inhibitor elacridar indicated that the observed effects were related to P-glycoprotein ATPase activity. In contrast, wild-type LLC-PK1 cells not expressing P-glycoprotein were not responsive towards stimulation with different P-glycoprotein substrates. Summarizing these findings, the used microsensor system is a generic system suitable for the identification of P-glycoprotein substrates. In contrast to biochemical P-glycoprotein assays, activation of the drug efflux pump can be monitored on-line in living cells to identify P-glycoprotein substrates and to study the molecular mechanisms of adenosintriphosphate-dependent active transport.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Ácidos/metabolismo , Oxigênio/metabolismo , Animais , Adesão Celular , Linhagem Celular , Humanos , Concentração de Íons de Hidrogênio , Células LLC-PK1 , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA