Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Expert Opin Pharmacother ; 21(9): 997-1004, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32237914

RESUMEN

INTRODUCTION: In chronic obstructive pulmonary disease (COPD), inhaled long-acting antimuscarinic agents (LAMA) are effective maintenance therapies used across all severity stages of the disease. Most of them are administered via dry powder inhalers, but these devices require a potent inspiratory flow which cannot be effectively achieved by patients with advanced disease. In such patients, inhaled therapy via nebulization might be an option. AREAS COVERED: Revefenacin is a LAMA that was specifically formulated for once daily nebulization and which was authorized by the FDA as a maintenance therapy for COPD. In phase II and III clinical studies discussed in this review, revefenacin demonstrated its rapid onset of action and sustained effect on lung function on both a short- and long-term basis. EXPERT OPINION: Nebulized revefenacin with once daily use does not require any particular effort of administration and hence can be used by patients with severe airways obstruction or by those having milder cognitive deficits. Further studies are needed, however, to better document the long-term cardiovascular safety and its ability to reduce the exacerbation rate.


Asunto(s)
Benzamidas/uso terapéutico , Carbamatos/uso terapéutico , Antagonistas Muscarínicos/uso terapéutico , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Benzamidas/farmacocinética , Benzamidas/farmacología , Carbamatos/farmacocinética , Carbamatos/farmacología , Humanos , Antagonistas Muscarínicos/farmacocinética , Antagonistas Muscarínicos/farmacología
2.
Molecules ; 25(4)2020 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-32102361

RESUMEN

The aim of the present study was to evaluate the possible gut inhibitory role of the phosphodiesterase (PDE) inhibitor roflumilast. Increasing doses of roflumilast were tested against castor oil-induced diarrhea in mice, whereas the pharmacodynamics of the same effect was determined in isolated rabbit jejunum tissues. For in silico analysis, the identified PDE protein was docked with roflumilast and papaverine using the Autodock vina program from the PyRx virtual screening tool. Roflumilast protected against diarrhea significantly at 0.5 and 1.5 mg/kg doses, with 40% and 80% protection. Ex vivo findings from jejunum tissues show that roflumilast possesses an antispasmodic effect by inhibiting spontaneous contractions in a concentration-dependent manner. Roflumilast reversed carbachol (CCh, 1 µM)-mediated and potassium (K+, 80 mM)-mediated contractile responses with comparable efficacies but different potencies. The observed potency against K+ was significantly higher in comparison to CCh, similar to verapamil. Experiments were extended to further confirm the inhibitory effect on Ca++ channels. Interestingly, roflumilast deflected Ca++ concentration-response curves (CRCs) to the right with suppression of the maximum peak at both tested doses (0.001-0.003 mg/mL), similar to verapamil. The PDE-inhibitory effect was authenticated when pre-incubation of jejunum tissues with roflumilast (0.03-0.1 mg/mL) produced a leftward deflection of isoprenaline-mediated inhibitory CRCs and increased the tissue level of cAMP, similar to papaverine. This idea was further strengthened by molecular docking studies, where roflumilast exhibited a better binding affinity (-9.4 kcal/mol) with the PDE protein than the standard papaverine (-8.3 kcal/mol). In conclusion, inhibition of Ca++ channels and the PDE-4 enzyme explains the pharmacodynamics of the gut inhibitory effect of roflumilast.


Asunto(s)
Aminopiridinas/farmacología , Antidiarreicos/farmacología , Benzamidas/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Diarrea/prevención & control , Parasimpatolíticos/farmacología , Inhibidores de Fosfodiesterasa 4/farmacología , Aminopiridinas/química , Aminopiridinas/farmacocinética , Animales , Antidiarreicos/química , Antidiarreicos/farmacocinética , Benzamidas/química , Benzamidas/farmacocinética , Sitios de Unión , Bloqueadores de los Canales de Calcio/química , Bloqueadores de los Canales de Calcio/farmacocinética , Carbacol/farmacología , Aceite de Ricino/administración & dosificación , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/química , Ciclopropanos/química , Ciclopropanos/farmacocinética , Ciclopropanos/farmacología , Diarrea/inducido químicamente , Diarrea/metabolismo , Diarrea/fisiopatología , Isoproterenol/farmacología , Yeyuno/efectos de los fármacos , Yeyuno/metabolismo , Ratones , Simulación del Acoplamiento Molecular , Papaverina/farmacología , Parasimpatolíticos/química , Parasimpatolíticos/farmacocinética , Inhibidores de Fosfodiesterasa 4/química , Inhibidores de Fosfodiesterasa 4/farmacocinética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Conejos , Verapamilo/farmacología
3.
Bioorg Med Chem Lett ; 30(5): 126857, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31982234

RESUMEN

The discovery and optimization of a novel series of GPR142 agonists are described. These led to the identification of compound 21 (LY3325656), which demonstrated anti-diabetic benefits in pre-clinical studies and ADME/PK properties suitable for human dosing. Compound 21 is the first GPR142 agonist molecule advancing to phase 1 clinic trials for the treatment of Type 2 diabetes.


Asunto(s)
Benzamidas/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Receptores Acoplados a Proteínas G/agonistas , Triazoles/uso terapéutico , Animales , Benzamidas/síntesis química , Benzamidas/farmacocinética , Perros , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Técnicas de Inactivación de Genes , Humanos , Hipoglucemiantes/síntesis química , Hipoglucemiantes/farmacocinética , Ratones Noqueados , Estructura Molecular , Ratas , Receptores Acoplados a Proteínas G/genética , Relación Estructura-Actividad , Triazoles/síntesis química , Triazoles/farmacocinética
4.
AAPS PharmSciTech ; 20(4): 155, 2019 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-30924008

RESUMEN

In this paper, a novel formulation of dual-release dry suspension of mosapride citrate (DRDS-MC) was designed which can be quickly released in the stomach while having sustained-release effect. Co-grinding mixture of mosapride citrate (MC) together with L-HPC as hydrophilic excipient was prepared in order to improve the solubility of MC. The co-grinding mixture was characterized by solubility studies, DSC, X-RD, SEM, FTIR, and size distribution before the preparation of the DRDS-MC. Then, the co-grinding mixture was used to prepare DRDS-MC via wet granulation method. The evaluation of DRDS-MC was focused on physicochemical properties, intestinal absorption, and pharmacokinetics. The results of DSC, X-RD, SEM, FTIR, and size distribution indicated that MC resides in co-grinding mixture with no crystalline changes, hydrogen bonds made L-HPC greatly improving the solubility of MC. Then, the dissolution of DRDS-MC reached 70% in pH 1.2 within 2 h, and the 12-h dissolution of MC in pH 6.8 was nearly 80%. The sedimentation volume after 3 h was 0.94 and redispersibility was good. The linear regression equation between in vitro release of DRDS-MC and intestinal absorption fraction in rats was: Y = 29.215 + 47.535*X (r = 0.952). At last, pharmacokinetic studies in beagle dogs demonstrated that DRDS-MC has prolonged effect compared with commercial formulation Gasmotin as a reference. All results indicated that the DRDS-MC could be quickly released in the stomach while having sustained-release effect.


Asunto(s)
Benzamidas/síntesis química , Benzamidas/farmacocinética , Absorción Gastrointestinal/efectos de los fármacos , Fármacos Gastrointestinales/síntesis química , Fármacos Gastrointestinales/farmacocinética , Morfolinas/síntesis química , Morfolinas/farmacocinética , Animales , Estudios Cruzados , Preparaciones de Acción Retardada/síntesis química , Preparaciones de Acción Retardada/farmacocinética , Perros , Evaluación Preclínica de Medicamentos/métodos , Liberación de Fármacos/efectos de los fármacos , Liberación de Fármacos/fisiología , Excipientes/síntesis química , Excipientes/farmacocinética , Absorción Gastrointestinal/fisiología , Masculino , Distribución Aleatoria , Ratas , Solubilidad , Suspensiones
5.
J Appl Toxicol ; 39(3): 485-497, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30345541

RESUMEN

The benzoylurea chitin synthesis inhibitor teflubenzuron, widely used against sea lice in North Atlantic aquaculture, may pose an environmental threat to non-targeted crustaceans. In this experiment, laboratory acclimated pink shrimp (Pandalus montagui), a species found in fjords with Atlantic salmon farming, were exposed to dietary teflubenzuron for 46 days (control; low dose: 0.01 µg/g; high dose: 0.1 µg/g). The exposure doses represent 0.1% and 1% of a standard treatment dose for Atlantic salmon. Mortality and prevalence of deformities, pharmacokinetics, oxidative stress and transcriptomic and metabolomic profiling were used to assess the response to teflubenzuron exposure. Mortality in the high-dose group was 25% (five of 20 individuals). No control or low-dose group shrimps died. Phenotypic responses,i.e., leg deformities (0 control, 6 low, 8 high) and cloudy eyes (0 control, 3 low, 7 high), were observed in some surviving shrimps (control n = 15, low n = 17, high n = 15). Accumulated levels of teflubenzuron in shrimps from the high-dose group ranged from 4.7 to 369 ng/g wet weight. Transcriptomic profiling showed very few significantly altered genes in the exposed shrimps. Teflubenzuron-induced changes to the metabolome pointed to well-known effects of benzoylurea agents, with reduced levels of N-acetylglucosamine indicating an effect on chitin synthesis. The metabolomic profiling showed that teflubenzuron exposure was associated with reduced energy metabolism. Some metabolites pointed to increased necrosis and/or bacterial overgrowth in the teflubenzuron-exposed shrimps. In conclusion, this study shows that teflubenzuron causes phenotypic effects in P. montagui exposed to 0.1% of the treatment dose given to Atlantic salmon.


Asunto(s)
Benzamidas/toxicidad , Metaboloma/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Pandalidae/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Aminoácidos/análisis , Animales , Benzamidas/farmacocinética , Quitina/biosíntesis , Metabolismo de los Lípidos/efectos de los fármacos , Pandalidae/metabolismo
6.
Eur J Haematol ; 102(2): 163-173, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30347469

RESUMEN

OBJECTIVES: Domatinostat (4SC-202) is a selective class I histone deacetylase inhibitor (HDACi). This phase I study investigated safety, tolerability, pharmacokinetics (PK), pharmacodynamics, and antitumor activity in patients with advanced hematological malignancies. METHODS: Domatinostat was administered orally once (QD) or twice daily (BID) on days 1-14 with 7 days off or continuously days 1-21 in a 3 + 3 design at 7 dose levels from 25 to 400 mg total daily dose (TDD). Twenty-four patients were treated with domatinostat. RESULTS: No formal maximum tolerated dose (MTD) was determined. One dose-limiting toxicity (DLT, grade 4 hypercalcemia) occurred during 200 mg BID continuous treatment. Six patients were reported with ≥ grade 3 treatment-related adverse events (TRAE; grade 3 hematological in three patients, grade 3 and grade 4 liver enzyme increase in 2 patients, grade 4 pulmonary embolism, and grade 4 hypercalcemia in one patient each). Higher grade hepatic TRAE occurred in the 200 mg BID continuous treatment cohort. Out of 24 patients, 1 achieved a complete response, 1 achieved a partial response, and 18 had stable disease as best response. CONCLUSION: Administration of domatinostat was safe, well tolerated with signs of antitumor activity. Four hundred milligram TDD in a 200 mg BID schedule (14 + 7) is the recommended phase II dose for monotherapy.


Asunto(s)
Benzamidas/uso terapéutico , Neoplasias Hematológicas/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Benzamidas/administración & dosificación , Benzamidas/efectos adversos , Benzamidas/farmacocinética , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Femenino , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/mortalidad , Inhibidores de Histona Desacetilasas/administración & dosificación , Inhibidores de Histona Desacetilasas/efectos adversos , Inhibidores de Histona Desacetilasas/farmacocinética , Humanos , Masculino , Metástasis de la Neoplasia , Estadificación de Neoplasias , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
7.
Future Oncol ; 15(6): 579-589, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30381956

RESUMEN

Acalabrutinib received an accelerated US FDA approval for patients with relapsed/refractory mantle cell lymphoma in 2017 and is currently being evaluated in chronic lymphocytic leukemia (CLL). To date, ibrutinib is the only Bruton tyrosine kinase (BTK) inhibitor that's approved for treatment of CLL. Acalabrutinib is a second generation BTK inhibitor that binds covalently to the Cys481 residue on BTK and has half maximal inhibitory concentration (IC50) of 3 nM. In preclinical mouse models, acalabrutinib significantly reduced proliferation of CLL cells. Results of Phase I/II trials revealed overall response rates (ORR) of 96% in treatment-naive, 93% in relapsed/refractory and 76% in ibrutinib intolerant patients with CLL. The most common adverse effects (>20%) were grade 1-2 comprising constitutional symptoms, GI toxicity, rash and myelosuppression. There were limited grade 3 or 4 toxicities, involving syncope, pneumonia, hypertension, atrial fibrillation, neutropenia and thrombocytopenia.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzamidas/uso terapéutico , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazinas/uso terapéutico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Benzamidas/administración & dosificación , Benzamidas/efectos adversos , Benzamidas/farmacocinética , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Humanos , Leucemia Linfocítica Crónica de Células B/patología , Terapia Molecular Dirigida , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacocinética , Pirazinas/administración & dosificación , Pirazinas/efectos adversos , Pirazinas/farmacocinética , Resultado del Tratamiento
8.
Artículo en Inglés | MEDLINE | ID: mdl-30373799

RESUMEN

NVR 3-778 is the first capsid assembly modulator (CAM) that has demonstrated antiviral activity in hepatitis B virus (HBV)-infected patients. NVR 3-778 inhibited the generation of infectious HBV DNA-containing virus particles with a mean antiviral 50% effective concentration (EC50) of 0.40 µM in HepG2.2.15 cells. The antiviral profile of NVR 3-778 indicates pan-genotypic antiviral activity and a lack of cross-resistance with nucleos(t)ide inhibitors of HBV replication. The combination of NVR 3-778 with nucleos(t)ide analogs in vitro resulted in additive or synergistic antiviral activity. Mutations within the hydrophobic pocket at the dimer-dimer interface of the core protein could confer resistance to NVR 3-778, which is consistent with the ability of the compound to bind to core and to induce capsid assembly. By targeting core, NVR 3-778 inhibits pregenomic RNA encapsidation, viral replication, and the production of HBV DNA- and HBV RNA-containing particles. NVR 3-778 also inhibited de novo infection and viral replication in primary human hepatocytes with EC50 values of 0.81 µM against HBV DNA and between 3.7 and 4.8 µM against the production of HBV antigens and intracellular HBV RNA. NVR 3-778 showed favorable pharmacokinetics and safety in animal species, allowing serum levels in excess of 100 µM to be achieved in mice and, thus, enabling efficacy studies in vivo The overall preclinical profile of NVR 3-778 predicts antiviral activity in vivo and supports its further evaluation for safety, pharmacokinetics, and antiviral activity in HBV-infected patients.


Asunto(s)
Antivirales/farmacología , Benzamidas/farmacología , Cápside/efectos de los fármacos , ADN Viral/antagonistas & inhibidores , Virus de la Hepatitis B/efectos de los fármacos , Hepatitis B/tratamiento farmacológico , Piperidinas/farmacología , ARN Viral/antagonistas & inhibidores , Animales , Antígenos Virales/genética , Antígenos Virales/metabolismo , Antivirales/sangre , Antivirales/química , Antivirales/farmacocinética , Benzamidas/sangre , Benzamidas/química , Benzamidas/farmacocinética , Cápside/química , Cápside/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , Evaluación Preclínica de Medicamentos , Femenino , Células Hep G2 , Hepatitis B/virología , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Hepatocitos/virología , Humanos , Masculino , Ratones , Pruebas de Sensibilidad Microbiana , Piperidinas/sangre , Piperidinas/química , Piperidinas/farmacocinética , Cultivo Primario de Células , ARN Viral/genética , ARN Viral/metabolismo , Proteínas del Núcleo Viral/antagonistas & inhibidores , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/metabolismo , Replicación Viral/efectos de los fármacos
9.
PLoS One ; 13(10): e0204605, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30359371

RESUMEN

Obesity and insulin resistance are primary risk factors for Non-Alcoholic Fatty Liver Disease (NAFLD). NAFLD is generally exhibited by non-progressive simple steatosis. However, a significant subset of patient's progress to nonalcoholic steatohepatitis (NASH) that is defined by the presence of steatosis, inflammation and hepatocyte injury with fibrosis. Unfortunately, there are no approved therapies for NAFLD or NASH and therefore therapeutic approaches are urgently needed. Niclosamide is an U.S. Food and Drug Administration (FDA)-approved anthelmintic drug that mediates its effect by uncoupling oxidative phosphorylation. Niclosamide and its salt forms, Niclosamide Ethanolamine (NEN), and Niclosamide Piperazine (NPP) have shown efficacy in murine models of diet induced obesity characterized by attenuation of the prominent fatty liver disease phenotype and improved glucose metabolism. While the exact mechanism(s) underlying these changes remains unclear, the ability to uncouple oxidative phosphorylation leading to increased energy expenditure and lipid metabolism or attenuation of PKA mediated glucagon signaling in the liver have been proposed. Unfortunately, niclosamide has very poor water solubility, leading to low oral bioavailability. This, in addition to mitochondrial uncoupling activity and potential genotoxicity have reduced enthusiasm for its clinical use. More recently, salt forms of niclosamide, NEN and NPP, have demonstrated improved oral bioavailability while retaining activity. This suggests that development of safer more effective niclosamide derivatives for the treatment of NAFLD and Type 2 Diabetes may be possible. Herein we explored the ability of a series of N-substituted phenylbenzamide derivatives of the niclosamide salicylanilide chemotype to attenuate hepatic steatosis using a novel phenotypic in vitro model of fatty liver and the high fat diet-fed mouse model of diet induced obesity. These studies identified novel compounds with improved pre-clinical properties that attenuate hepatic steatosis in vitro and in vivo. These compounds with improved drug properties may be useful in alleviating symptoms and protection against disease progression in patients with metabolic syndrome and NAFLD.


Asunto(s)
Fármacos Antiobesidad/farmacología , Benzamidas/farmacología , Dieta Alta en Grasa/efectos adversos , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Animales , Fármacos Antiobesidad/química , Fármacos Antiobesidad/farmacocinética , Benzamidas/química , Benzamidas/farmacocinética , Respiración de la Célula/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Escherichia coli/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Lipogénesis/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Obesidad/metabolismo , Ratas Sprague-Dawley , Salmonella typhimurium/efectos de los fármacos
10.
ACS Chem Neurosci ; 8(11): 2374-2380, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28841278

RESUMEN

Adamantanyl benzamide 1 was identified as a potent P2X7R antagonist but failed to progress further due to poor metabolic stability. We describe the synthesis and SAR of a series of bioisosteres of benzamide 1 to explore improvements in the pharmacological properties of this lead. Initial efforts investigated a series of heteroaromatic bioisosteres, which demonstrated improved physicochemical properties but reduced P2X7R antagonism. Installation of bioisosteric fluorine on the adamantane bridgeheads was well tolerated and led to a series of bioisosteres with improved physicochemical properties and metabolic stability. Trifluorinated benzamide 34 demonstrated optimal physicochemical parameters, superior metabolic stability (ten times longer than lead benzamide 1), and an improved physicokinetic profile and proved effective in the presence of several known P2X7R polymorphisms.


Asunto(s)
Adamantano/análogos & derivados , Benzamidas/farmacología , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X7/efectos de los fármacos , Adamantano/farmacología , Animales , Benzamidas/síntesis química , Benzamidas/química , Benzamidas/farmacocinética , Biotransformación , Evaluación Preclínica de Medicamentos , Estabilidad de Medicamentos , Humanos , Microsomas Hepáticos/metabolismo , Estructura Molecular , Oxidación-Reducción , Polimorfismo de Nucleótido Simple , Antagonistas del Receptor Purinérgico P2X/síntesis química , Antagonistas del Receptor Purinérgico P2X/química , Antagonistas del Receptor Purinérgico P2X/farmacocinética , Ratas , Receptores Purinérgicos P2X7/genética , Relación Estructura-Actividad
11.
Br J Pharmacol ; 174(16): 2662-2681, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28677901

RESUMEN

BACKGROUND AND PURPOSE: Our initial aim was to generate cannabinoid agents that control spasticity, occurring as a consequence of multiple sclerosis (MS), whilst avoiding the sedative side effects associated with cannabis. VSN16R was synthesized as an anandamide (endocannabinoid) analogue in an anti-metabolite approach to identify drugs that target spasticity. EXPERIMENTAL APPROACH: Following the initial chemistry, a variety of biochemical, pharmacological and electrophysiological approaches, using isolated cells, tissue-based assays and in vivo animal models, were used to demonstrate the activity, efficacy, pharmacokinetics and mechanism of action of VSN16R. Toxicological and safety studies were performed in animals and humans. KEY RESULTS: VSN16R had nanomolar activity in tissue-based, functional assays and dose-dependently inhibited spasticity in a mouse experimental encephalomyelitis model of MS. This effect occurred with over 1000-fold therapeutic window, without affecting normal muscle tone. Efficacy was achieved at plasma levels that are feasible and safe in humans. VSN16R did not bind to known CB1 /CB2 /GPPR55 cannabinoid-related receptors in receptor-based assays but acted on a vascular cannabinoid target. This was identified as the major neuronal form of the big conductance, calcium-activated potassium (BKCa ) channel. Drug-induced opening of neuronal BKCa channels induced membrane hyperpolarization, limiting excessive neural-excitability and controlling spasticity. CONCLUSIONS AND IMPLICATIONS: We identified the neuronal form of the BKCa channel as the target for VSN16R and demonstrated that its activation alleviates neuronal excitability and spasticity in an experimental model of MS, revealing a novel mechanism to control spasticity. VSN16R is a potential, safe and selective ligand for controlling neural hyper-excitability in spasticity.


Asunto(s)
Benzamidas/uso terapéutico , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Espasticidad Muscular/tratamiento farmacológico , Animales , Benzamidas/química , Benzamidas/farmacocinética , Benzamidas/farmacología , Perros , Método Doble Ciego , Endocannabinoides/química , Endocannabinoides/farmacocinética , Endocannabinoides/farmacología , Endocannabinoides/uso terapéutico , Femenino , Hepatocitos/metabolismo , Isomerismo , Macaca , Masculino , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/fisiología , Ratones , Ratones Noqueados , Conejos , Ratas Sprague-Dawley , Ratas Wistar , Receptor Cannabinoide CB1/genética , Receptores de Cannabinoides/genética , Conducto Deferente/efectos de los fármacos , Conducto Deferente/fisiología
12.
Blood Rev ; 31(4): 205-211, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28185693

RESUMEN

Approximately 900,000 people are affected by some sort of venous thromboembolic (VTE) event every year in the United States. VTE diagnosis used to mean treatment with medications that required routine lab monitoring for safety and efficacy. Activated factor X (FXa) inhibition has emerged as a convenient pathway for management of VTE and currently three FXa inhibitors are available for anticoagulation management - rivaroxaban, apixaban, and edoxaban. Continued development of medications utilizing this pathway may offer advantages via novel pharmacokinetic or pharmacodynamic properties that may minimize the adverse effects associated with traditional anticoagulant therapy. This review summarizes the available information regarding pharmacokinetic, pharmacodynamic, and early safety and efficacy data for three factor Xa inhibitors being developed - darexaban, betrixaban and nokxaban. The studies reviewed in this article suggests that three newer agents possess the potential for promise based on early phase I and II trials.


Asunto(s)
Descubrimiento de Drogas , Inhibidores del Factor Xa/uso terapéutico , Tromboembolia Venosa/tratamiento farmacológico , Administración Oral , Animales , Azepinas/administración & dosificación , Azepinas/farmacocinética , Azepinas/farmacología , Azepinas/uso terapéutico , Benzamidas/administración & dosificación , Benzamidas/farmacocinética , Benzamidas/farmacología , Benzamidas/uso terapéutico , Coagulación Sanguínea/efectos de los fármacos , Inhibidores del Factor Xa/administración & dosificación , Inhibidores del Factor Xa/farmacocinética , Inhibidores del Factor Xa/farmacología , Humanos , Pirazoles/administración & dosificación , Pirazoles/farmacocinética , Pirazoles/farmacología , Pirazoles/uso terapéutico , Piridinas/administración & dosificación , Piridinas/farmacocinética , Piridinas/farmacología , Piridinas/uso terapéutico , Piridonas/administración & dosificación , Piridonas/farmacocinética , Piridonas/farmacología , Piridonas/uso terapéutico , Rivaroxabán/administración & dosificación , Rivaroxabán/farmacocinética , Rivaroxabán/farmacología , Rivaroxabán/uso terapéutico , Tiazoles/administración & dosificación , Tiazoles/farmacocinética , Tiazoles/farmacología , Tiazoles/uso terapéutico
13.
Synapse ; 70(3): 112-20, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26671330

RESUMEN

Glycine transporter type-1 (GlyT1) has been proposed as a target for drug development for schizophrenia. PET imaging with a GlyT1 specific radiotracer will allow for the measurement of target occupancy of GlyT1 inhibitors, and for in vivo investigation of GlyT1 alterations in schizophrenia. We conducted a comparative evaluation of two GlyT1 radiotracers, [(11) C]GSK931145, and [(18) F]MK-6577, in baboons. Two baboons were imaged with [(11) C]GSK931145 and [(18) F]MK-6577. Blocking studies with GSK931145 (0.3 or 0.2 mg/kg) were conducted to determine the level of tracer specific binding. [(11) C]GSK931145 and [(18) F]MK-6577 were synthesized in good yield and high specific activity. Moderately fast metabolism was observed for both tracers, with ∼ 30% of parent at 30 min post-injection. In the brain, both radiotracers showed good uptake and distribution profiles consistent with regional GlyT1 densities. [(18) F]MK-6577 displayed higher uptake and faster kinetics than [(11) C]GSK931145. Time activity curves were well described by the two-tissue compartment model. Regional volume of distribution (VT ) values were higher for [(18) F]MK-6577 than [(11) C]GSK931145. Pretreatment with GSK931145 reduced tracer uptake to a homogeneous level throughout the brain, indicating in vivo binding specificity and lack of a reference region for both radiotracers. Linear regression analysis of VT estimates between tracers indicated higher specific binding for [(18) F]MK-6577 than [(11) C]GSK931145, consistent with higher regional binding potential (BPND ) values of [(18) F]MK-6577 calculated using VT from the baseline scans and non-displaceable distribution volume (VND ) derived from blocking studies. [(18) F]MK-6577 appears to be a superior radiotracer with higher brain uptake, faster kinetics, and higher specific binding signals than [(11) C]GSK931145.


Asunto(s)
Benzamidas , Radioisótopos de Carbono , Glicinérgicos , Proteínas de Transporte de Glicina en la Membrana Plasmática/metabolismo , Radiofármacos , Sulfonamidas , Animales , Benzamidas/síntesis química , Benzamidas/química , Benzamidas/farmacocinética , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Mapeo Encefálico , Radioisótopos de Carbono/farmacocinética , Cromatografía Líquida de Alta Presión , Evaluación Preclínica de Medicamentos , Femenino , Glicinérgicos/síntesis química , Glicinérgicos/química , Glicinérgicos/farmacocinética , Cinética , Modelos Lineales , Imagen por Resonancia Magnética , Estructura Molecular , Papio , Tomografía de Emisión de Positrones , Radiofármacos/síntesis química , Radiofármacos/química , Radiofármacos/farmacocinética , Sulfonamidas/síntesis química , Sulfonamidas/química , Sulfonamidas/farmacocinética
14.
Nucl Med Biol ; 42(12): 967-74, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26320813

RESUMEN

INTRODUCTION: Metabotropic glutamate subtype receptor 1 (mGluR1) is implicated in several neuropsychiatric disorders and is a target for drug development. [(18)F]FIMX ([(18)F]4-fluoro--N-methyl-N--(4-(6-(methylamino)pyrimidin-4-yl)thiazol-2-yl)benzamide) is an effective radioligand for imaging brain mGluR1 with PET. A similarly effective radioligand with a shorter half-life would usefully allow PET studies of mGluR1 at baseline and after pharmacological or other challenge on the same day. Here we describe the preparation of [(11)C]FIMX for evaluation in monkey with PET. METHODS: [(11)C]FIMX was prepared via Pd-promoted carbonylation of 1-fluoro-4-iodobenzene with [(11)C]carbon monoxide, aminolysis of the [(11)C]acyl-palladium complex with the requisite Boc-protected amine, and deprotection with HCl in THF. PET scans of [(11)C]FIMX injected into a monkey were performed at baseline and after preblock of mGluR1 with measurement of the arterial input function. RESULTS: The radiosynthesis required 42 min and gave [(11)C]FIMX in about 5% overall decay-corrected radiochemical yield and with a specific activity of about 100 GBq/µmol. PET in rhesus monkey at baseline showed that radioactivity peaked high in receptor-rich cerebellum and much lower in receptor-poor occipital cortex. Radioactivity in cerebellum declined to 32% of peak at 85 min. VT at baseline appeared stable in all brain regions after 60 min. Under mGluR1 pre-blocked condition, radioactivity uptake in all regions declined more rapidly to a low level. Receptor pre-block reduced VT from 13.0 to 1.5 in cerebellum and from 2.9 to 1.4 in occipital cortex. CONCLUSION: [(11)C]FIMX is an effective radioligand for imaging mGluR1 in monkey with PET.


Asunto(s)
Benzamidas/farmacocinética , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Radioisótopos de Carbono/farmacocinética , Tomografía de Emisión de Positrones/métodos , Radiofármacos/farmacocinética , Receptores de Glutamato Metabotrópico/metabolismo , Tiazoles/farmacocinética , Animales , Evaluación Preclínica de Medicamentos , Procesamiento de Imagen Asistido por Computador , Macaca mulatta , Radioquímica
15.
Neuropharmacology ; 99: 337-46, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26239818

RESUMEN

The α7nAChR agonist, PNU-282987, has previously been shown to have a neuroprotective effect against loss of retinal ganglion cells (RGCs) in an in vivo glaucoma model when the agent was injected into the vitreous chamber of adult Long Evans rat eyes. Here, we characterized the neuroprotective effect of PNU-282987 at the nerve fiber and retinal ganglion cell layer, determined that neuroprotection occurred when the agonist was applied as eye drops and verified detection of the agonist in the retina, using LC/MS/MS. To induce glaucoma-like conditions in adult Long Evans rats, hypertonic saline was injected into the episcleral veins to induce scar tissue and increase intraocular pressure. Within one month, this procedure produced significant loss of RGCs compared to untreated conditions. RGCs were quantified after immunostaining with an antibody against Thy 1.1 and imaged using a confocal microscope. In dose-response studies, concentrations of PNU-282987 were applied to the animal's right eye two times each day, while the left eye acted as an internal control. Eye drops of PNU-282987 resulted in neuroprotection against RGC loss in a dose-dependent manner using concentrations between 100 µM and 2 mM PNU-282987. LC/MS/MS results demonstrated that PNU-282987 was detected in the retina when applied as eye drops, relatively small amounts of PNU-282987 were measured in blood plasma and no PNU-282987 was detected in cardiac tissue. These results support the hypothesis that eye drop application of PNU-282987 can prevent loss of RGCs associated with glaucoma, which can lead to neuroprotective treatments for diseases that involve α7nAChRs.


Asunto(s)
Benzamidas/farmacología , Compuestos Bicíclicos con Puentes/farmacología , Glaucoma/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Células Ganglionares de la Retina/efectos de los fármacos , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Animales , Benzamidas/farmacocinética , Compuestos Bicíclicos con Puentes/farmacocinética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Glaucoma/metabolismo , Glaucoma/patología , Corazón/efectos de los fármacos , Masculino , Microscopía Confocal , Miocardio/metabolismo , Fármacos Neuroprotectores/farmacocinética , Soluciones Oftálmicas/farmacocinética , Soluciones Oftálmicas/farmacología , Ratas Long-Evans , Células Ganglionares de la Retina/metabolismo , Células Ganglionares de la Retina/patología , Solución Salina Hipertónica , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
16.
Nucl Med Biol ; 42(6): 570-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25858513

RESUMEN

INTRODUCTION: The aims of the present study were to develop an optimized microfluidic method for the production of the selective nicotinic acetylcholine α4ß2 receptor radiotracer [(18)F]-(-)-NCFHEB ([(18)F]-Flubatine) and to investigate its receptor binding profile and pharmacokinetic properties in rhesus monkeys in vivo. METHODS: [(18)F]-(-)-NCFHEB was prepared in two steps, a nucleophilic fluorination followed by N-Boc deprotection. PET measurements were performed in rhesus monkeys including baseline and preblocking experiments with nicotine (0.24 mg/kg). Radiometabolites in plasma were measured using HPLC. RESULTS: [(18)F]-(-)-NCFHEB was prepared in a total synthesis time of 140 min. The radiochemical purity in its final formulation was >98% and the mean specific radioactivity was 97.3 ± 16.1 GBq/µmol (n = 6) at end of synthesis (EOS). In the monkey brain, radioactivity concentration was high in the thalamus, moderate in the putamen, hippocampus, frontal cortex, and lower in the cerebellum. Nicotine blocked 98-100% of [(18)F]-(-)-NCFHEB specific binding, and the non-displaceable distribution volume (VND) was estimated at 5.9 ± 1.0 mL/cm(3) (n = 2), or 6.6 ± 1.1 mL/cm(3) after normalization by the plasma free fraction fP. Imaging data are amenable to kinetic modeling analysis using the multilinear analysis (MA1) method, and model-derived binding parameters display good test-retest reproducibility. In rhesus monkeys, [(18)F]-(-)-NCFHEB can yield robust regional binding potential (BPND) values (thalamus = 4.1 ± 1.5, frontal cortex = 1.2 ± 0.2, putamen = 0.96 ± 0.45, and cerebellum = 0.10 ± 0.29). CONCLUSION: An efficient microfluidic synthetic method was developed for preparation of [(18)F]-(-)-NCFHEB. PET examination in rhesus monkeys showed that [(18)F]-(-)-NCFHEB entered the brain readily and its regional radioactivity uptake pattern was in accordance with the known distribution of α4ß2 receptors. Estimated non-displaceable binding potential (BPND) values in brain regions were better than those of [(18)F]2-FA and comparable to [(18)F]AZAN. These results confirm previous findings and support further examination of [(18)F]-(-)-NCFHEB in humans.


Asunto(s)
Benzamidas/farmacocinética , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacocinética , Radioisótopos de Flúor/farmacocinética , Tomografía de Emisión de Positrones/métodos , Radiofármacos/farmacocinética , Receptores Nicotínicos/metabolismo , Animales , Evaluación Preclínica de Medicamentos , Humanos , Macaca mulatta , Radioquímica , Distribución Tisular
17.
Int J Cancer ; 137(8): 2007-18, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25868794

RESUMEN

Enhancer of Zeste Homolog 2 (EZH2) has emerged as a promising therapeutic target for treatment of a broad spectrum of tumors including gliomas. We explored the interactions of five novel, structurally similar EZH2 inhibitors (EPZ005687, EPZ-6438, UNC1999, GSK343 and GSK126) with P-glycoprotein (P-gp/ABCB1) and breast cancer resistance protein (BCRP/ABCG2). The compounds were screened by in vitro transwell assays and EPZ005687, EPZ-6438 and GSK126 were further tested in vivo using wild-type (WT), Abcb1 and/or Abcg2 knockout mice. All EZH2 inhibitors are transported by P-gp and BCRP, although in vitro the transporter affinity of GSK126 was obscured by very low membrane permeability. Both P-gp and Bcrp1 restrict the brain penetration of EPZ005687 and GSK126, whereas the brain accumulation of EPZ-6438 is limited by P-gp only and efflux of EPZ-6438 was completely abrogated by elacridar. Intriguingly, an unknown factor present in all knockout mouse strains causes EPZ005687 and EPZ-6438 retention in plasma relative to WT mice, a phenomenon not seen with GSK126. In WT mice, the GSK126 tissue-to-plasma ratio for all tissues is lower than for EPZ005687 or EPZ-6438. Moreover, the oral bioavailability of GSK126 is only 0.2% in WT mice, which increases to 14.4% in Abcb1;Abcg2 knockout mice. These results are likely due to poor membrane permeability and question the clinical usefulness of GSK126. Although all tested EZH2 inhibitors are substrates of P-gp and BCRP, restricting the brain penetration and potential utility for treatment of glioma, EPZ-6438 would be the most suitable candidate of this series.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Inhibidores Enzimáticos/farmacocinética , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Administración Oral , Animales , Benzamidas/administración & dosificación , Benzamidas/farmacocinética , Disponibilidad Biológica , Compuestos de Bifenilo , Línea Celular , Perros , Evaluación Preclínica de Medicamentos , Proteína Potenciadora del Homólogo Zeste 2 , Inhibidores Enzimáticos/administración & dosificación , Humanos , Indazoles/administración & dosificación , Indazoles/farmacocinética , Indoles/administración & dosificación , Indoles/farmacocinética , Células de Riñón Canino Madin Darby , Ratones , Ratones Noqueados , Morfolinas , Piridonas/administración & dosificación , Piridonas/farmacocinética
18.
Molecules ; 20(2): 2081-99, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25633335

RESUMEN

Research towards the non-invasive imaging of atherosclerotic plaques is of high clinical priority as early recognition of vulnerable plaques may reduce the incidence of cardiovascular events. The fibroblast activation protein alpha (FAP) was recently proposed as inflammation-induced protease involved in the process of plaque vulnerability. In this study, FAP mRNA and protein levels were investigated by quantitative polymerase chain reaction and immunohistochemistry, respectively, in human endarterectomized carotid plaques. A published boronic-acid based FAP inhibitor, MIP-1232, was synthetized and radiolabeled with iodine-125. The potential of this radiotracer to image plaques was evaluated by in vitro autoradiography with human carotid plaques. Specificity was assessed with a xenograft with high and one with low FAP level, grown in mice. Target expression analyses revealed a moderately higher protein level in atherosclerotic plaques than normal arteries correlating with plaque vulnerability. No difference in expression was determined on mRNA level. The radiotracer was successfully produced and accumulated strongly in the FAP-positive SK-Mel-187 melanoma xenograft in vitro while accumulation was negligible in an NCI-H69 xenograft with low FAP levels. Binding of the tracer to endarterectomized tissue was similar in plaques and normal arteries, hampering its use for atherosclerosis imaging.


Asunto(s)
Benzamidas , Compuestos de Boro , Enfermedades de las Arterias Carótidas/diagnóstico por imagen , Placa Aterosclerótica/diagnóstico por imagen , Radiofármacos , Actinas/genética , Actinas/metabolismo , Anciano , Animales , Benzamidas/farmacocinética , Compuestos de Boro/farmacocinética , Enfermedades de las Arterias Carótidas/metabolismo , Evaluación Preclínica de Medicamentos , Endopeptidasas , Femenino , Gelatinasas/antagonistas & inhibidores , Gelatinasas/genética , Gelatinasas/metabolismo , Expresión Génica , Humanos , Radioisótopos de Yodo , Masculino , Melanoma Experimental/diagnóstico por imagen , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Trasplante de Neoplasias , Placa Aterosclerótica/metabolismo , Cintigrafía , Radiofármacos/farmacocinética , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo
19.
ChemMedChem ; 10(1): 57-61, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25209672

RESUMEN

Previous studies have shown that the activation of mouse MrgC11, a G-protein-coupled receptor, by its peptide ligand BAM8-22 can inhibit chronic pain. A large-scale screen has been carried out to isolate small-molecule allosteric agonists of MrgX1, the human homologue of MrgC11. The goal of this study is to improve the efficacy and potency of positive allosteric modulators (PAMs) with therapeutic implications in combating chronic pain. Herein we report an iterative parallel synthesis effort and a structure-activity relationship study of a series of arylsulfonamides which led to the discovery of the first PAM of MrgX1, ML382.


Asunto(s)
Benzamidas/química , Receptores Acoplados a Proteínas G/metabolismo , Sulfonamidas/química , Regulación Alostérica , Animales , Benzamidas/metabolismo , Benzamidas/farmacocinética , Evaluación Preclínica de Medicamentos , Células HEK293 , Semivida , Humanos , Ratones , Unión Proteica , Ratas , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/metabolismo , Sulfonamidas/farmacocinética
20.
Antimicrob Agents Chemother ; 58(8): 4703-12, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24890599

RESUMEN

Amixicile shows efficacy in the treatment of Clostridium difficile infections (CDI) in a mouse model, with no recurrence of CDI. Since amixicile selectively inhibits the action of a B vitamin (thiamine pyrophosphate) cofactor of pyruvate:ferredoxin oxidoreductase (PFOR), it may both escape mutation-based drug resistance and spare beneficial probiotic gut bacteria that do not express this enzyme. Amixicile is a water-soluble derivative of nitazoxanide (NTZ), an antiparasitic therapeutic that also shows efficacy against CDI in humans. In comparative studies, amixicile showed no toxicity to hepatocytes at 200 µM (NTZ was toxic above 10 µM); was not metabolized by human, dog, or rat liver microsomes; showed equivalence or superiority to NTZ in cytochrome P450 assays; and did not activate efflux pumps (breast cancer resistance protein, P glycoprotein). A maximum dose (300 mg/kg) of amixicile given by the oral or intraperitoneal route was well tolerated by mice and rats. Plasma exposure (rats) based on the area under the plasma concentration-time curve was 79.3 h · µg/ml (30 mg/kg dose) to 328 h · µg/ml (100 mg/kg dose), the maximum concentration of the drug in serum was 20 µg/ml, the time to the maximum concentration of the drug in serum was 0.5 to 1 h, and the half-life was 5.6 h. Amixicile did not concentrate in mouse feces or adversely affect gut populations of Bacteroides species, Firmicutes, segmented filamentous bacteria, or Lactobacillus species. Systemic bioavailability was demonstrated through eradication of Helicobacter pylori in a mouse infection model. In summary, the efficacy of amixicile in treating CDI and other infections, together with low toxicity, an absence of mutation-based drug resistance, and excellent drug metabolism and pharmacokinetic metrics, suggests a potential for broad application in the treatment of infections caused by PFOR-expressing microbial pathogens in addition to CDI.


Asunto(s)
Antibacterianos/farmacocinética , Benzamidas/farmacocinética , Infecciones por Helicobacter/tratamiento farmacológico , Helicobacter pylori/efectos de los fármacos , Tiazoles/farmacocinética , Animales , Antibacterianos/sangre , Antibacterianos/farmacología , Área Bajo la Curva , Benzamidas/sangre , Benzamidas/farmacología , Disponibilidad Biológica , Línea Celular , Supervivencia Celular/efectos de los fármacos , Perros , Evaluación Preclínica de Medicamentos , Semivida , Infecciones por Helicobacter/sangre , Infecciones por Helicobacter/microbiología , Helicobacter pylori/crecimiento & desarrollo , Helicobacter pylori/metabolismo , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Masculino , Pruebas de Sensibilidad Microbiana , Microbiota/efectos de los fármacos , Microbiota/fisiología , Microsomas Hepáticos/efectos de los fármacos , Piruvato-Sintasa/metabolismo , Ratas , Tiamina Pirofosfato/metabolismo , Tiazoles/sangre , Tiazoles/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA