Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.755
Filtrar
1.
J Med Chem ; 67(8): 6268-6291, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38619191

RESUMEN

Overactivation of cyclic GMP-AMP synthase (cGAS) is implicated in the occurrence of many inflammatory and autoimmune diseases, and inhibition of cGAS with a specific inhibitor has been proposed as a potential therapeutic strategy. However, only a few low-potency cGAS inhibitors have been reported, and few are suitable for clinical investigation. As a continuation of our structural optimization on the reported cGAS inhibitor 6 (G140), we developed a series of spiro[carbazole-3,3'-pyrrolidine] derivatives bearing a unique 2-azaspiro[4.5]decane structural motif, among which compound 30d-S was identified with high cellular effects against cGAS. This compound showed improved plasma exposure, lower clearance, and an oral bioavailability of 35% in rats. Moreover, in the LPS-induced acute lung injury (ALI) mice model, oral administration of compound 30d-S at 30 mg/kg markedly reduced lung inflammation and alleviated histopathological changes. These results confirm that 30d-S is a new efficacious cGAS inhibitor and is worthy of further investigation.


Asunto(s)
Lesión Pulmonar Aguda , Carbazoles , Diseño de Fármacos , Nucleotidiltransferasas , Pirrolidinas , Lesión Pulmonar Aguda/tratamiento farmacológico , Animales , Ratones , Masculino , Humanos , Ratas , Carbazoles/síntesis química , Carbazoles/farmacología , Carbazoles/química , Carbazoles/uso terapéutico , Carbazoles/farmacocinética , Pirrolidinas/farmacología , Pirrolidinas/síntesis química , Pirrolidinas/química , Pirrolidinas/uso terapéutico , Pirrolidinas/farmacocinética , Nucleotidiltransferasas/antagonistas & inhibidores , Nucleotidiltransferasas/metabolismo , Lipopolisacáridos , Ratas Sprague-Dawley , Compuestos de Espiro/síntesis química , Compuestos de Espiro/farmacología , Compuestos de Espiro/química , Compuestos de Espiro/uso terapéutico , Compuestos de Espiro/farmacocinética , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/química , Relación Estructura-Actividad , Simulación del Acoplamiento Molecular
2.
J Med Chem ; 67(8): 6064-6080, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38595098

RESUMEN

It has been shown that PRMT5 inhibition by small molecules can selectively kill cancer cells with homozygous deletion of the MTAP gene if the inhibitors can leverage the consequence of MTAP deletion, namely, accumulation of the MTAP substrate MTA. Herein, we describe the discovery of TNG908, a potent inhibitor that binds the PRMT5·MTA complex, leading to 15-fold-selective killing of MTAP-deleted (MTAP-null) cells compared to MTAPintact (MTAP WT) cells. TNG908 shows selective antitumor activity when dosed orally in mouse xenograft models, and its physicochemical properties are amenable for crossing the blood-brain barrier (BBB), supporting clinical study for the treatment of both CNS and non-CNS tumors with MTAP loss.


Asunto(s)
Antineoplásicos , Proteína-Arginina N-Metiltransferasas , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Proteína-Arginina N-Metiltransferasas/metabolismo , Humanos , Animales , Ratones , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Antineoplásicos/síntesis química , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacocinética , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias/tratamiento farmacológico , Encéfalo/metabolismo , Relación Estructura-Actividad
3.
J Pharm Biomed Anal ; 244: 116116, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38537542

RESUMEN

EC5026 is a novel soluble epoxide hydrolase inhibitor being developed clinically to treat neuropathic pain and inflammation. In the current study, we employed the LC-ESI-Q-TOF-MS/MS technique to identify four in-vivo phase-I metabolites of EC5026 in rat model, out of which three were found to be novel. The identified metabolites include aliphatic hydroxylation, di-hydroxylation, terminal desaturation, and carboxylation. No phase-II metabolites were found. The pharmacokinetic profile of identified metabolites was established after a single oral dose of EC5026 to Wistar rats. The Tmax of the drug and metabolites were found to be in the range of 1-2 hours and 4-12 hours, respectively. The major metabolites M1 and M2 were found to have more than 2-fold (263.87% AUC) and equivalent exposure (96.33% AUC) compared to the parent drug, respectively. Further, the docking study revealed that the mono-hydroxylated and terminally desaturated metabolites possess better binding affinity than the parent drug. Therefore, these metabolites may hold sEH inhibition potential and can be followed through future research.


Asunto(s)
Epóxido Hidrolasas , Ratas Wistar , Espectrometría de Masas en Tándem , Epóxido Hidrolasas/antagonistas & inhibidores , Epóxido Hidrolasas/metabolismo , Animales , Ratas , Espectrometría de Masas en Tándem/métodos , Masculino , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/farmacología , Cromatografía Liquida/métodos , Hidroxilación , Administración Oral , Espectrometría de Masa por Ionización de Electrospray/métodos
4.
J Med Chem ; 66(13): 9201-9222, 2023 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-37334504

RESUMEN

Acute pancreatitis (AP) is a potentially life-threatening illness characterized by an exacerbated inflammatory response with limited options for pharmacological treatment. Here, we describe the rational development of a library of soluble epoxide hydrolase (sEH) inhibitors for the treatment of AP. Synthesized compounds were screened in vitro for their sEH inhibitory potency and selectivity, and the results were rationalized by means of molecular modeling studies. The most potent compounds were studied in vitro for their pharmacokinetic profile, where compound 28 emerged as a promising lead. In fact, compound 28 demonstrated a remarkable in vivo efficacy in reducing the inflammatory damage in cerulein-induced AP in mice. Targeted metabololipidomic analysis further substantiated sEH inhibition as a molecular mechanism of the compound underlying anti-AP activity in vivo. Finally, pharmacokinetic assessment demonstrated a suitable profile of 28 in vivo. Collectively, compound 28 displays strong effectiveness as sEH inhibitor with potential for pharmacological AP treatment.


Asunto(s)
Pancreatitis , Ratones , Animales , Pancreatitis/inducido químicamente , Pancreatitis/tratamiento farmacológico , Epóxido Hidrolasas , Enfermedad Aguda , Inhibidores Enzimáticos/uso terapéutico , Inhibidores Enzimáticos/farmacocinética
5.
Clin Drug Investig ; 43(6): 413-420, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37269489

RESUMEN

BACKGROUND AND OBJECTIVE: Deficiencies of enzymes acting downstream of glucosylceramide synthase (GCS) can cause severe substrate accumulation. Venglustat is a small-molecule, brain-penetrant GCS inhibitor under investigation for multiple diseases involving pathogenic glycosphingolipid accumulation. Here, we evaluate the pharmacokinetics, safety, and tolerability of venglustat in healthy Chinese volunteers. METHODS: Study PKM16116 was a phase I, single-center, non-randomized, open-label study to investigate the pharmacokinetics, safety, and tolerability of a single 15 mg dose of orally administered venglustat in healthy Chinese volunteers aged 18 to 45 years. RESULTS: A total of 14 volunteers (7 male; 7 female) with a body mass index from 20.9 kg/m2 to 27.1 kg/m2 were enrolled. The median time to reach the venglustat maximum plasma concentration was 2.50 h post-dose. The mean terminal half-life of venglustat was 30.6 ± 7.40 h. The mean systemic exposures across all participants were 60.3 ± 17.3 ng/mL for the maximum plasma concentration, and 2280 ± 697 ng·h/mL for the area under the plasma concentration-time curve extrapolated to infinity. There were no relevant differences in venglustat pharmacokinetics between male and female volunteers. A post hoc cross-study comparison analysis showed comparable venglustat pharmacokinetics in Chinese and non-Chinese volunteers. Venglustat was safe and well tolerated in the current study (a total of five Grade 1 treatment-emergent adverse events were reported in three volunteers). CONCLUSION: Venglustat showed a favorable pharmacokinetic, safety, and tolerability profile in healthy Chinese volunteers following a single oral 15 mg dose. CLINICAL TRIAL REGISTRY NO: CTR20201012 ( http://www.chinadrugtrials.org.cn ) registered on 24 February 2021 and ChiCTR2200066559 ( http://www.chictr.org.cn ) retrospectively registered on 9 December 2022.


Asunto(s)
Pueblo Asiatico , Femenino , Humanos , Masculino , Administración Oral , Área Bajo la Curva , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Voluntarios Sanos , Adolescente , Adulto Joven , Adulto , Persona de Mediana Edad , China , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/uso terapéutico
6.
Xenobiotica ; 52(8): 904-915, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36149349

RESUMEN

In vitro incubation of the bacterial ß-glucuronidase inhibitor UNC10201652 (4-(8-(piperazin-1-yl)-1,2,3,4-tetrahydro-[1,2,3]triazino[4',5':4,5]thieno[2,3-c]isoquinolin-5-yl)morpholine) with mouse, rat, and human liver microsomes and hepatocytes generated metabolites at multiple sites via deethylations, oxidations and glucuronidation.Two UNC10201652 metabolites were detected in human, and four in mouse and rat liver microsomal incubations. Intrinsic clearances of UNC10201652 in human, mouse, and rat liver microsomes were 48.1, 115, and 194 µL/min/mg respectively.Intrinsic clearances for human, mouse, and rat hepatocytes were 20.9, 116, and 140 µL/min/106 cells respectively and 24 metabolites were characterised: 9 for human and 11 for both rodent species.Plasma clearance was 324.8 mL/min/kg with an elimination half-life of 0.66 h following IV administration of UNC10201652 to Swiss Albino mice (3 mg/kg). Pre-treatment with 1-aminobenzotriazole (ABT) decreased clearance to 127.43 mL/min/kg, increasing the t1/2 to 3.66 h.Comparison of profiles after oral administration of UNC10201652 to control and pre-treated mice demonstrated a large increase in Cmax (from 15.2 ng/mL to 184.0 ng/mL), a delay in Tmax from 0.25 to 1 h and increased AUC from 20.1 to 253 h ng/ml. ABT pre-treatment increased oral bioavailability from 15% to >100% suggesting that CYP450's contributed significantly to UNC10201652 clearance in mice.


Asunto(s)
Inhibidores Enzimáticos , Animales , Humanos , Ratones , Ratas , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Hepatocitos/metabolismo , Microsomas Hepáticos/metabolismo , Morfolinas/metabolismo , Morfolinas/farmacología , Piperazinas/metabolismo , Piperazinas/farmacocinética
7.
Invest New Drugs ; 40(5): 1042-1050, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35932388

RESUMEN

Pevonedistat (TAK-924/MLN4924) is an investigational small molecule inhibitor of the NEDD8-activating enzyme that has demonstrated clinical activity across solid tumors and hematological malignancies. Here we report the results of a phase 1 study evaluating the effect of rifampin, a strong CYP3A inducer, on the pharmacokinetics (PK) of pevonedistat in patients with advanced solid tumors (NCT03486314). Patients received a single 50 mg/m2 pevonedistat dose via a 1-h infusion on Days 1 (in the absence of rifampin) and 10 (in the presence of rifampin), and daily oral dosing of rifampin 600 mg on Days 3-11. Twenty patients were enrolled and were evaluable for PK and safety. Following a single dose of pevonedistat at 50 mg/m2, the mean terminal half-life of pevonedistat was 5.7 and 7.4 h in the presence and in the absence of rifampin, respectively. The geometric mean AUC0-inf of pevonedistat in the presence of rifampin was 79% of that without rifampin (90% CI: 69.2%-90.2%). The geometric mean Cmax of pevonedistat in the presence of rifampin was similar to that in the absence of rifampin (96.2%; 90% CI: 79.2%-117%). Coadministration of pevonedistat with rifampin, a strong metabolic enzyme inducer, did not result in clinically meaningful decreases in systemic exposures of pevonedistat. The study results support the recommendation that no pevonedistat dose adjustment is needed for patients receiving concomitant CYP3A inducers. CLINICALTRIALS.GOV IDENTIFIER: NCT03486314.


Asunto(s)
Neoplasias , Rifampin , Área Bajo la Curva , Ciclopentanos , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacocinética , Humanos , Proteína NEDD8 , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Pirimidinas/efectos adversos , Rifampin/farmacología , Rifampin/uso terapéutico
8.
Clin Exp Pharmacol Physiol ; 49(11): 1197-1208, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35877984

RESUMEN

Some studies have shown that the area under the concentration-time curve (AUC) of mycophenolic acid (MPA) should be higher for children with nephrotic syndrome (NS) than after renal transplantation. The pharmacodynamic aspect of MPA, the activity of inosine monophosphate dehydrogenase (IMPDH), has not been studied in children with NS. The study included 21 children (4-16 years old) with NS treated with mycophenolate mofetil. MPA and its glucuronide plasma concentrations were determined using validated high-performance liquid chromatography-ultraviolet (HPLC-UV). The separate HPLC-UV method was applied for IMPDH activity determination. The variability was expressed by the coefficient of variation (CV). IMPDH activity and MPA concentration (Ctrough ) before the morning dose amounted to 29.95 µmol s-1  mol-1 adenosine monophosphate (AMP) (range, 6.71-98.60 µmol s-1  mol-1 AMP) and 1.72 µg/mL (range, 0.39-4.34 µg/mL), respectively, whereas the area under the effect-time curve from 0 to 4 h and MPA AUC0-4 were 130.36 µmol s-1  mol-1 AMP × h (range, 23.58-306.57 µmol s-1  mol-1 AMP × h) and 24.63 µg h/mL (range, 12.21-67.48 µg h/mL), respectively. IMPDH activity decreased concomitantly with MPA concentration increase, however, the variability of the pharmacodynamic parameters was greater than of the pharmacokinetics. The median degree of maximum IMPDH inhibition was 61%. MPA Ctrough and predicted AUC were lower than in our previous study. Only a few MPA pharmacokinetic parameters correlated with the pharmacodynamics. IMPDH activity did not correlate with the children's age and did not differ between boys and girls. MPA clearance was the highest in younger children (median, 10.54 L/m2 /h) and cholesterol correlated negatively with the children's age (r = -0.659, P = 0.003). IMPDH minimum activity and the degree of maximum IMPDH inhibition were similar to those obtained in renal transplant recipients. IMPDH activity does not undergo developmental or gender-specific regulation in children with NS. MPA underexposure might be more frequent in younger children, especially with high cholesterol and triglycerides levels because of high MPA clearance.


Asunto(s)
Ácido Micofenólico , Síndrome Nefrótico , Adenosina Monofosfato , Adolescente , Niño , Preescolar , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/uso terapéutico , Femenino , Glucurónidos , Humanos , IMP Deshidrogenasa , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Inosina Monofosfato , Masculino , Ácido Micofenólico/uso terapéutico , Síndrome Nefrótico/tratamiento farmacológico , Triglicéridos
9.
J Med Chem ; 65(4): 3343-3358, 2022 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-35166541

RESUMEN

Cholesterol 24-hydroxylase (CH24H or CYP46A1) is a brain-specific cytochrome P450 enzyme that metabolizes cholesterol into 24S-hydroxycholesterol (24HC) for regulating brain cholesterol homeostasis. For the development of a novel and potent CH24H inhibitor, we designed and synthesized 3,4-disubstituted pyridine derivatives using a structure-based drug design approach starting from compounds 1 (soticlestat) and 2 (thioperamide). Optimization of this series by focusing on ligand-lipophilicity efficiency value resulted in the discovery of 4-(4-methyl-1-pyrazolyl)pyridine derivative 17 (IC50 = 8.5 nM) as a potent and highly selective CH24H inhibitor. The X-ray crystal structure of CH24H in complex with compound 17 revealed a unique binding mode. Both blood-brain barrier penetration and reduction of 24HC levels (26% reduction) in the mouse brain were confirmed by oral administration of 17 at 30 mg/kg, indicating that 17 is a promising tool for the novel and selective inhibition of CH24H.


Asunto(s)
Anticolesterolemiantes/síntesis química , Anticolesterolemiantes/farmacología , Colesterol 24-Hidroxilasa/antagonistas & inhibidores , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Piridinas/síntesis química , Piridinas/farmacología , Animales , Anticolesterolemiantes/farmacocinética , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Colesterol/metabolismo , Cristalografía por Rayos X , Diseño de Fármacos , Inhibidores Enzimáticos/farmacocinética , Femenino , Hidroxicolesteroles , Lípidos/química , Ratones , Ratones Endogámicos C57BL , Relación Estructura-Actividad
10.
Molecules ; 27(3)2022 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-35164277

RESUMEN

The pharmacokinetic profile of ZST316 and ZST152, arginine analogues with inhibitory activity towards human dimethylarginine dimethylaminohydrolase-1 (DDAH1), was investigated in mice using a newly developed HPLC-MS/MS method. The method proved to be reproducible, precise, and accurate for the measurement of the compounds in plasma and urine. Four-week-old female FVB mice received a single dose of ZST316 and ZST152 by intravenous bolus (30 mg/Kg) and oral gavage (60 mg/Kg). ZST316 Cmax was 67.4 µg/mL (intravenous) and 1.02 µg/mL (oral), with a half-life of 6 h and bioavailability of 4.7%. ZST152 Cmax was 24.9 µg/mL (intravenous) and 1.65 µg/mL (oral), with a half-life of 1.2 h and bioavailability of 33.3%. Urinary excretion of ZST152 and ZST316 was 12.5%-22.2% and 2.3%-7.5%, respectively. At least eight urinary metabolites were identified. After chronic intraperitoneal treatment with the more potent DDAH1 inhibitor, ZST316 (30 mg/Kg/day for three weeks), the bioavailability was 59% and no accumulation was observed. Treatment was well tolerated with no changes in body weight vs. untreated animals and no clinical signs of toxicity or distress. The results of this study show that ZST316 has a favorable pharmacokinetic profile, following intraperitoneal administration, to investigate the effects of DDAH1 inhibition in mice.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacocinética , Animales , Arginina/administración & dosificación , Arginina/análogos & derivados , Arginina/farmacocinética , Disponibilidad Biológica , Cromatografía Líquida de Alta Presión , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/química , Femenino , Humanos , Ratones , Espectrometría de Masas en Tándem
11.
J Med Chem ; 65(3): 2191-2207, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35089028

RESUMEN

Monoacylglycerol lipase (MAGL) is one of the key enzymes in the endocannabinoid system. Inhibition of MAGL has been proposed as an attractive approach for the treatment of various diseases. In this study, we designed and successfully synthesized two series of piperazinyl pyrrolidin-2-one derivatives as novel reversible MAGL inhibitors. (R)-[18F]13 was identified through the preliminary evaluation of two carbon-11-labeled racemic structures [11C]11 and [11C]16. In dynamic positron-emission tomography (PET) scans, (R)-[18F]13 showed a heterogeneous distribution and matched the MAGL expression pattern in the mouse brain. High brain uptake and brain-to-blood ratio were achieved by (R)-[18F]13 in comparison with previously reported reversible MAGL PET radiotracers. Target occupancy studies with a therapeutic MAGL inhibitor revealed a dose-dependent reduction of (R)-[18F]13 accumulation in the mouse brain. These findings indicate that (R)-[18F]13 ([18F]YH149) is a highly promising PET probe for visualizing MAGL non-invasively in vivo and holds great potential to support drug development.


Asunto(s)
Encéfalo/diagnóstico por imagen , Inhibidores Enzimáticos/química , Monoacilglicerol Lipasas/metabolismo , Neuroimagen/métodos , Radiofármacos/química , Animales , Encéfalo/metabolismo , Radioisótopos de Carbono/química , Cristalografía por Rayos X , Estabilidad de Medicamentos , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Ratones , Conformación Molecular , Monoacilglicerol Lipasas/química , Tomografía de Emisión de Positrones , Radiofármacos/metabolismo , Radiofármacos/farmacocinética , Ratas , Ratas Wistar , Relación Estructura-Actividad , Distribución Tisular
12.
Eur J Med Chem ; 229: 114082, 2022 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-34995925

RESUMEN

Snail and histone deacetylases (HDACs) have an important impact on cancer treatment, especially for their synergy. Therefore, the development of inhibitors targeting both Snail and HDAC might be a promising strategy for the treatment of cancers. In this work, we synthesized a series of Snail/HDAC dual inhibitors. Compound 9n displayed the most potent inhibitory activity against HDAC1 with an IC50 of 0.405 µM, potent inhibition against Snail with a Kd of 0.180 µM, and antiproliferative activity in HCT-116 cell lines with an IC50 of 0.0751 µM. Compound 9n showed a good inhibitory effect on NCI-H522 (GI50 = 0.0488 µM), MDA-MB-435 (GI50 = 0.0361 µM), and MCF7 (GI50 = 0.0518 µM). Docking studies showed that compound 9n can be well docked into the active binding sites of Snail and HDAC. Further studies showed that compound 9n increased histone H4 acetylation in HCT-116 cells and decreased the expression of Snail protein to induce cell apoptosis. These findings highlight the potential for the development of Snail/HDAC dual inhibitors as anti-solid tumour cancer drugs.


Asunto(s)
Aminopiridinas/química , Antineoplásicos/síntesis química , Benzamidas/química , Inhibidores Enzimáticos/síntesis química , Histona Desacetilasas/metabolismo , Factores de Transcripción de la Familia Snail/síntesis química , Aminopiridinas/farmacocinética , Animales , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Benzamidas/farmacocinética , Biomarcadores de Tumor , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/farmacocinética , Células HCT116 , Humanos , Simulación del Acoplamiento Molecular , Unión Proteica , Conformación Proteica , Ratas , Factores de Transcripción de la Familia Snail/farmacocinética , Relación Estructura-Actividad
13.
Clin Pharmacol Ther ; 111(2): 391-403, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33998672

RESUMEN

This study evaluated the safety, tolerability, pharmacokinetics, and pharmacodynamics of BIA 10-2474, a fatty acid amide hydrolase (FAAH) inhibitor, after first administration to healthy male and female participants. Participants (n = 116) were recruited into this phase I, double-blind, randomized, placebo-controlled, single ascending dose and multiple ascending dose (10-day) study. The primary outcome was the safety and tolerability of BIA 10-2474. Secondary outcomes were pharmacokinetics of BIA 10-2474 and pharmacodynamics, considering plasma concentrations of anandamide and three other fatty acid amides (FAAs) and leukocyte FAAH activity. Single oral doses of 0.25-100 mg and repeated oral doses of 2.5-50 mg were evaluated. BIA 10-2474 was well tolerated up to 100 mg as a single dose and up to 20 mg once daily for 10 days. In the cohort receiving repeated administrations of 50 mg, there were central nervous system adverse events in five of six participants, one with fatal outcome, which led to early termination of the study. BIA 10-2474 showed a linear relationship between dose and area under plasma concentration-time curve (AUC) across the entire dose range and reached steady state within 5-6 days of administration, with an accumulation ratio, based on AUC0-24h , of <2 on Day 10. BIA 10-2474 was rapidly absorbed with a mean terminal elimination half-life of 8-10 hours (Day 10). BIA 10-2474 caused reversible, dose-related increases in plasma FAAs. In conclusion, we propose that these data, as well as the additional data generated since the clinical trial was stopped, do not provide a complete mechanistic explanation for the tragic fatality.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Sistema Nervioso Central/efectos de los fármacos , Óxidos N-Cíclicos/efectos adversos , Inhibidores Enzimáticos/efectos adversos , Piridinas/efectos adversos , Administración Oral , Sistema Nervioso Central/fisiopatología , Óxidos N-Cíclicos/administración & dosificación , Óxidos N-Cíclicos/farmacocinética , Método Doble Ciego , Esquema de Medicación , Cálculo de Dosificación de Drogas , Terminación Anticipada de los Ensayos Clínicos , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacocinética , Femenino , Francia , Voluntarios Sanos , Humanos , Masculino , Seguridad del Paciente , Piridinas/administración & dosificación , Piridinas/farmacocinética , Medición de Riesgo , Factores de Riesgo
15.
CPT Pharmacometrics Syst Pharmacol ; 11(5): 604-615, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34951129

RESUMEN

TAS-114 is a dual deoxyuridine triphosphatase (dUTPase) and dihydropyrimidine dehydrogenase (DPD) inhibitor expected to widen the therapeutic index of capecitabine. Its maximum tolerated dose (MTD) was determined from a safety perspective in a combination study with capecitabine; however, its inhibitory effects on DPD activity were not assessed in the study. The dose justification to select its MTD as the recommended dose in terms of DPD inhibition has been required, but the autoinduction profile of TAS-114 made it difficult. To this end, an approach using a population pharmacokinetic (PPK)/pharmacodynamic (PD) model incorporating autoinduction was planned; however, the utility of this approach in the dose justification has not been reported. Thus, the aim of this study was to demonstrate the utility of a PPK/PD model incorporating autoinduction in the dose justification via a case study of TAS-114. Plasma concentrations of TAS-114 from 185 subjects and those of the endogenous DPD substrate uracil from 24 subjects were used. A two-compartment model with first-order absorption with lag time and an enzyme turnover model were selected for the pharmacokinetic (PK) model. Moreover, an indirect response model was selected for the PD model to capture the changes in plasma uracil concentrations. Model-based simulations provided the dose justification that DPD inhibition by TAS-114 reached a plateau level at the MTD, whereas exposures of TAS-114 increased dose dependently. Thus, the utility of a PPK/PD model incorporating autoinduction in the dose justification was demonstrated via this case study of TAS-114.


Asunto(s)
Pirimidinas , Sulfonamidas , Capecitabina , Inhibidores Enzimáticos/farmacocinética , Humanos , Pirimidinas/uso terapéutico , Uracilo/farmacocinética
16.
Chem Biol Drug Des ; 99(3): 496-503, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34951520

RESUMEN

Inhibition of extracellular secreted enzyme autotaxin (ATX) represents an attractive strategy for the development of new therapeutics to treat various diseases and a few inhibitors entered in clinical trials. We herein describe structure-based design, synthesis, and biological investigations revealing a potent and orally bioavailable ATX inhibitor 1. During the molecular docking and scoring studies within the ATX enzyme (PDB-ID: 4ZGA), the S-enantiomer (Gscore = -13.168 kcal/mol) of the bound ligand PAT-494 scored better than its R-enantiomer (Gscore = -9.562 kcal/mol) which corroborated with the reported observation and analysis of the results suggested the scope of manipulation of the hydantoin substructure in PAT-494. Accordingly, the docking-based screening of a focused library of 10 compounds resulted in compound 1 as a better candidate for pharmacological studies. Compound 1 was synthesized from L-tryptophan and evaluated against ATX enzymatic activities with an IC50 of 7.6 and 24.6 nM in biochemical and functional assays, respectively. Further, ADME-PK studies divulged compound 1 as non-cytotoxic (19.02% cell growth inhibition at 20 µM in human embryonic kidney cells), metabolically stable against human liver microsomes (CLint  = 15.6 µl/min/mg; T1/2  = 113.2 min) with solubility of 4.82 µM and orally bioavailable, demonstrating its potential to be used for in vivo experiments.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/química , Indoles/química , Hidrolasas Diéster Fosfóricas/química , Administración Oral , Animales , Sitios de Unión , Estabilidad de Medicamentos , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Semivida , Humanos , Imidazoles/química , Indoles/metabolismo , Indoles/farmacocinética , Microsomas Hepáticos/metabolismo , Simulación del Acoplamiento Molecular , Hidrolasas Diéster Fosfóricas/metabolismo , Piridinas/química , Ratas , Ratas Sprague-Dawley , Estereoisomerismo
17.
J Med Chem ; 65(1): 592-615, 2022 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-34957834

RESUMEN

Inflammatory bowel disease (IBD) is a multifactorial autoimmune disease, representing a major clinical challenge. Herein, a strategy of dual-targeting approach employing retinoic acid receptor-related orphan receptor γ-t (RORγt) and dihydroorotate dehydrogenase (DHODH) was proposed for the treatment of IBD. Dual RORγt/DHODH inhibitors are expected not only to reduce RORγt-driven Th17 cell differentiation but also to mitigate the expansion and activation of T cells, which may enhance anti-inflammatory effects. Starting from 2-aminobenzothiazole hit 1, a series of 2-aminotetrahydrobenzothiazoles were discovered as potent dual RORγt/DHODH inhibitors. Compound 14d stands out with IC50 values of 0.110 µM for RORγt and of 0.297 µM for DHODH. With acceptable mouse pharmacokinetic profiles, 14d exhibited remarkable in vivo anti-inflammatory activity and dose-dependently alleviated the severity of dextran sulfate sodium (DSS)-induced acute colitis in mice. Taken together, the present study provides a novel framework for the development of therapeutic agents for the treatment of IBD.


Asunto(s)
Dihidroorotato Deshidrogenasa/antagonistas & inhibidores , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/efectos de los fármacos , Receptores de Ácido Retinoico/efectos de los fármacos , Animales , Antiinflamatorios/síntesis química , Antiinflamatorios/farmacología , Diferenciación Celular/efectos de los fármacos , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Sulfato de Dextran , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Resistencia a Medicamentos , Inhibidores Enzimáticos/farmacocinética , Humanos , Enfermedades Inflamatorias del Intestino/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Modelos Moleculares , Relación Estructura-Actividad , Células Th17/efectos de los fármacos
18.
Eur J Med Chem ; 229: 114055, 2022 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-34971874

RESUMEN

The development of inhibitors targeting the PI3K-Akt-mTOR signaling pathway has been greatly hindered by the on-target AEs, such as hyperglycemia and hepatotoxicities. In this study, a series of diaryl urea derivatives has been designed and synthesized based on clinical candidate gedatolisib (6aa), and most of the newly synthesized derivatives showed kinase inhibitory and antiproliferative activities within nanomolar and submicromolar level, respectively. The terminal l-prolineamide substituted derivative 6 ab showed 8.6-fold more potent PI3Kα inhibitory activity (0.7 nM) and 4.6-fold more potent antiproliferative effect against HCT116 cell lines (0.11 µM) compared with control 6aa. The potential antitumor mechanism and efficacy of 6 ab in HCT116 xenograft models have also been evaluated, and found 6 ab showed comparable in vivo antitumor activity with 6aa. The safety investigations revealed that compound 6 ab exhibited more safer profiles in the selectivity of liver cells (selectivity index: >6.6 vs 1.85) and blood glucose regulation than 6aa. In addition, the in vitro stability assays also indicated our developed compound 6 ab possessed good metabolic stabilities.


Asunto(s)
Antineoplásicos/química , Neoplasias Colorrectales/tratamiento farmacológico , Inhibidores Enzimáticos/síntesis química , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Urea/síntesis química , Animales , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/farmacocinética , Femenino , Humanos , Ratones Endogámicos BALB C , Modelos Moleculares , Simulación del Acoplamiento Molecular , Morfolinas/farmacología , Neoplasias Experimentales , Unión Proteica , Conformación Proteica , Transducción de Señal , Relación Estructura-Actividad , Triazinas/farmacología , Urea/farmacocinética
19.
Biomed Pharmacother ; 145: 112471, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34852990

RESUMEN

NF-κB contributes to the biosynthesis of various chemokines, cytokines, and enzymes. It plays many crucial roles in the upstream neuroinflammatory pathways. Briefly, the inhibitory IkB subunit is cleaved and phosphorylated by the IKK-α/ß enzyme. It leads to the activation and translocation of the NF-κB (p50/p65) complex into the nucleus. Subsequently, the activated NF-κB interacts with the genomic DNA and contributes to expressing various proinflammatory cytokines. In the present study, we developed a novel NF-κB inhibitor encoded (D5) and investigated the efficacy of our druggable compound through several in silico, in vitro, and in situ analysis. The results demonstrated that D5 not only inhibited the mRNA expression of the IKK-α/ß enzyme (around 86-96% suppression rate for both cell lines at 12 and 24 h time frames) but also by interacting to the active site of the mentioned kinase (dock score -6.14 and binding energy -23.60 kcal/mol) reduced the level of phosphorylated IkB-α in the cytosol around 96-99% and p65 subunit in the nucleus around 73-90% (among all groups in 12 and 24 h time points). Additionally, the results indicated that D5 suppressed the NF-κB target mRNA levels of TNF-α and IL-6 in a total average of around 92%. Overall, The results demonstrated that D5 in a considerably lower concentration than Dis (0.71 µM vs. 52.73 µM) showed significantly higher inhibitory efficacy on NF-κB translocation approx. 200-300%. The results suggested D5 as a potent NF-κB silencer, but further investigations are required to validate our outcomes.


Asunto(s)
Quinasa I-kappa B , FN-kappa B/metabolismo , Enfermedades Neuroinflamatorias , Sistemas de Translocación de Proteínas , Alcaloides/farmacología , Benzodioxoles/farmacología , Línea Celular , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Desarrollo de Medicamentos/métodos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacocinética , Humanos , Quinasa I-kappa B/antagonistas & inhibidores , Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Fosforilación/efectos de los fármacos , Piperidinas/farmacología , Alcamidas Poliinsaturadas/farmacología , Sistemas de Translocación de Proteínas/efectos de los fármacos , Sistemas de Translocación de Proteínas/metabolismo , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo
20.
J Med Chem ; 64(24): 17777-17794, 2021 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-34871500

RESUMEN

In our efforts to identify novel small molecule inhibitors for the treatment of adrenoleukodystrophy (ALD), we conducted a high-throughput radiometric screen for inhibitors of elongation of very long chain fatty acid 1 (ELOVL1) enzyme. We developed a series of highly potent, central nervous system (CNS)-penetrant pyrimidine ether-based compounds with favorable pharmacokinetics culminating in compound 22. Compound 22 is a selective inhibitor of ELOVL1, reducing C26:0 VLCFA synthesis in ALD patient fibroblasts and lymphocytes in vitro. Compound 22 reduced C26:0 lysophosphatidyl choline (LPC), a subtype of VLCFA, in the blood of ATP binding cassette transporter D1 (ABCD1) KO mice, a murine model of ALD to near wild-type levels. Compound 22 is a low-molecular-weight, potent ELOVL1 inhibitor that may serve as a useful tool for exploring therapeutic approaches to the treatment of ALD.


Asunto(s)
Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Elongasas de Ácidos Grasos/antagonistas & inhibidores , Pirimidinas/farmacología , Administración Oral , Adrenoleucodistrofia/tratamiento farmacológico , Animales , Disponibilidad Biológica , Perros , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacocinética , Éteres/química , Células HEK293 , Humanos , Macaca fascicularis , Ratones , Pirimidinas/administración & dosificación , Pirimidinas/farmacocinética , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA