Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Toxicol Appl Pharmacol ; 438: 115905, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35122773

RESUMEN

Systemic therapies targeting transforming growth factor beta (TGFß) or TGFßR1 kinase (ALK5) have been plagued by toxicities including cardiac valvulopathy and bone physeal dysplasia in animals, posing a significant challenge for clinical development in pulmonary indications. The current work aims to demonstrate that systemic ALK5-associated toxicities can be mitigated through localized lung delivery. Lung-selective (THRX-144644) and systemically bioavailable (galunisertib) ALK5 inhibitors were compared to determine whether lung selectivity is sufficient to maintain local tissue concentrations while mitigating systemic exposure and consequent pathway-related findings. Both molecules demonstrated potent ALK5 activity in rat precision cut lung slices (PCLS; p-SMAD3 half-maximal inhibitory concentration [IC50], 141 nM and 1070 nM for THRX-144644 and galunisertib, respectively). In 14-day repeat-dose studies in rats, dose-related cardiac valvulopathy was recapitulated with oral galunisertib at doses ≥150 mg/kg/day. In contrast, inhaled nebulized THRX-144644 did not cause similar systemic findings up to the maximally tolerated doses in rats or dogs (10 and 1.5 mg/kg/day, respectively). THRX-144644 lung-to-plasma ratios ranged from 100- to 1200-fold in rats and dogs across dose levels. THRX-144644 lung trough (24 h) concentrations in rats and dogs ranged from 3- to 17-fold above the PCLS IC50 across tolerated doses. At a dose level exceeding tolerability (60 mg/kg/day; 76-fold above PCLS IC50) minimal heart and bone changes were observed when systemic drug concentrations reached pharmacologic levels. In conclusion, the current preclinical work demonstrates that localized pulmonary delivery of an ALK5 inhibitor leads to favorable TGFß pathway pharmacodynamic inhibition in lung while minimizing key systemic toxicities.


Asunto(s)
Pulmón/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Administración Oral , Animales , Perros , Femenino , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Pirazoles/toxicidad , Quinolinas/toxicidad , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo
2.
Bioorg Med Chem ; 28(2): 115227, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31862310

RESUMEN

Aberrant hedgehog (Hh) pathway signaling is implicated in multiple cancer types and targeting the Smoothened (SMO) receptor, a key protein of the Hh pathway, has proven effective in treating metastasized basal cell carcinoma. Our lead optimization effort focused on a series of heteroarylamides. We observed that a methyl substitution ortho to the heteroaryl groups on an aniline core significantly improved the potency of this series of compounds. These findings predated the availability of SMO crystal structure in 2013. Here we retrospectively applied quantum mechanics calculations to demonstrate the o-Me substitution favors the bioactive conformation by inducing a dihedral twist between the heteroaryl rings and the core aniline. The o-Me also makes favorable hydrophobic interactions with key residue side chains in the binding pocket. From this effort, two compounds (AZD8542 and AZD7254) showed excellent pharmacokinetics across multiple preclinical species and demonstrated in vivo activity in abrogating the Hh paracrine pathway as well as anti- tumor effects.


Asunto(s)
Antineoplásicos/farmacología , Benzamidas/farmacología , Descubrimiento de Drogas , Imidazoles/farmacología , Receptor Smoothened/antagonistas & inhibidores , Proteína con Dedos de Zinc GLI1/antagonistas & inhibidores , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Benzamidas/síntesis química , Benzamidas/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Imidazoles/síntesis química , Imidazoles/química , Ratones , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/metabolismo , Receptor Smoothened/metabolismo , Relación Estructura-Actividad , Proteína con Dedos de Zinc GLI1/metabolismo
3.
Drug Metab Rev ; 46(3): 379-419, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24909234

RESUMEN

Aliphatic nitrogen heterocycles such as piperazine, piperidine, pyrrolidine, morpholine, aziridine, azetidine, and azepane are well known building blocks in drug design and important core structures in approved drug therapies. These core units have been targets for metabolic attack by P450s and other drug metabolizing enzymes such as aldehyde oxidase and monoamine oxidase (MAOs). The electron rich nitrogen and/or α-carbons are often major sites of metabolism of alicyclic amines. The most common biotransformations include N-oxidation, N-conjugation, oxidative N-dealkylation, ring oxidation, and ring opening. In some instances, the metabolic pathways generate electrophilic reactive intermediates and cause bioactivation. However, potential bioactivation related adverse events can be attenuated by structural modifications. Hence it is important to understand the biotransformation pathways to design stable drug candidates that are devoid of metabolic liabilities early in the discovery stage. The current review provides a comprehensive summary of biotransformation and bioactivation pathways of aliphatic nitrogen containing heterocycles and strategies to mitigate metabolic liabilities.


Asunto(s)
Aminas/metabolismo , Biotransformación/fisiología , Preparaciones Farmacéuticas/metabolismo , Animales , Sistema Enzimático del Citocromo P-450/metabolismo , Humanos , Inactivación Metabólica/fisiología
4.
Bioorg Med Chem ; 22(5): 1782-90, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24495863

RESUMEN

Comprehensive structure activity relationship (SAR) studies were conducted on a focused screening hit, 2-(methylthio)-3-(phenylsulfonyl)-4H-pyrido[1,2-a]pyrimidin-4-imine (1, IC50: 4.0 nM), as 5-HT6 selective antagonists. Activity was improved some 2-4 fold when small, electron-donating groups were added to the central core as observed in 19, 20 and 26. Molecular docking of key compounds in a homology model of the human 5-HT6 receptor was used to rationalize our structure-activity relationship (SAR) findings. In pharmacokinetic experiments, compound 1 displayed good brain uptake in rats following intra-peritoneal administration, but limited oral bioavailability.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Iminas/farmacocinética , Receptores de Serotonina/uso terapéutico , Animales , Humanos , Iminas/farmacología , Simulación del Acoplamiento Molecular , Ratas , Relación Estructura-Actividad
5.
Drug Metab Rev ; 44(3): 224-52, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22697420

RESUMEN

The high rate of attrition during drug development and its associated high research and development (R&D) cost have put pressure on pharmaceutical companies to ensure that candidate drugs going to clinical testing have the appropriate quality such that the biological hypothesis could be evaluated. To help achieve this ambition, drug metabolism and pharmacokinetic (DMPK) science and increasing investment have been deployed earlier in the R&D process. To gain maximum return on investment, it is essential that DMPK concepts are both appropriately integrated into the compound design process and that compound selection is focused on accurate prediction of likely outcomes in patients. This article describes key principles that underpin the contribution of DMPK science for small-molecule research based on 15 years of discovery support in a major pharmaceutical company. It does not aim to describe the breadth and depth of DMPK science, but more the practical application for decision making in real-world situations.


Asunto(s)
Industria Farmacéutica/métodos , Drogas en Investigación/farmacocinética , Animales , Toma de Decisiones en la Organización , Evaluación Preclínica de Medicamentos/métodos , Interacciones Farmacológicas , Drogas en Investigación/efectos adversos , Drogas en Investigación/metabolismo , Drogas en Investigación/farmacología , Humanos , Inactivación Metabólica , Tasa de Depuración Metabólica
6.
Bioorg Med Chem Lett ; 22(14): 4907-11, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22704236

RESUMEN

Cell-based subset screening of compounds using a Gli transcription factor reporter cell assay and shh stimulated cell differentiation assay identified a series of bisamide compounds as hedgehog pathway inhibitors with good potency. Using a ligand-based optimization strategy, heteroaryl groups were utilized as conformationally restricted amide isosteres replacing one of the amides which significantly increased their potency against SMO and the hedgehog pathway while decreasing activity against p38α kinase. We report herein the identification of advanced lead compounds such as imidazole 11c and 11f encompassing good p38α selectivity, low nanomolar potency in both cell assays, excellent physiochemical properties and in vivo pharmacokinetics.


Asunto(s)
Amidas/química , Proteínas Hedgehog/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Amidas/farmacología , Animales , Evaluación Preclínica de Medicamentos , Ratones , Estructura Molecular , Inhibidores de Proteínas Quinasas/farmacología , Relación Estructura-Actividad
7.
Bioorg Med Chem Lett ; 22(6): 2330-7, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22342147

RESUMEN

Checkpoint kinase 1 (Chk1, CHEK1) is a Ser/Thr protein kinase that plays a key role in mediating the cellular response to DNA-damage. Synthesis and evaluation of a previously described class of Chk1 inhibitors, triazoloquinolones/triazolones (TZs) is further described herein. Our investigation of structure-activity relationships led to the identification of potent inhibitors 14c, 14h and 16e. Key challenges included modulation of physicochemical properties and pharmacokinetic (PK) parameters to enable compound testing in a Chk1 specific hollow fiber pharmacodynamic model. In this model, 16e was shown to abrogate topotecan-induced cell cycle arrest in a dose dependent manner. The demonstrated activity of TZs in this model in combination with a chemotherapeutic agent as well as radiotherapy validates this series of Chk1 inhibitors. X-ray crystal structures (PDB code: 2YEX and 2YER) for an initial lead and an optimized analog are also presented.


Asunto(s)
Antineoplásicos/síntesis química , Neoplasias del Colon/terapia , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Quinasas/metabolismo , Triazoles/síntesis química , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Neoplasias del Colon/enzimología , Terapia Combinada , Cristalografía por Rayos X , Daño del ADN , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Conformación Proteica , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/uso terapéutico , Relación Estructura-Actividad , Topotecan/farmacología , Triazoles/farmacocinética , Triazoles/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Toxicol Sci ; 186(2): 323-337, 2022 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-35134999

RESUMEN

Izencitinib (TD-1473), an oral, gut-selective pan-Janus kinase (JAK) inhibitor under investigation for treatment of inflammatory bowel diseases, was designed for optimal efficacy in the gastrointestinal tract while minimizing systemic exposures and JAK-related safety findings. The nonclinical safety of izencitinib was evaluated in rat and dog repeat-dose and rat and rabbit reproductive and developmental toxicity studies. Systemic exposures were compared with JAK inhibitory potency to determine effects at or above pharmacologic plasma concentrations (≥1× plasma average plasma concentration [Cave]:JAK 50% inhibitory concentration [IC50] ratio). In rats and dogs, 1000 and 30 mg/kg/day izencitinib, respectively, produced minimal systemic findings (ie, red/white cell changes) and low systemic concentrations (approximately 1× plasma Cave:JAK IC50 ratio) with an 8× nonclinical:clinical systemic area under the curve (AUC) margin compared with exposures at the highest clinically tested dose (300 mg, quaque die, once daily, phase 1 study in healthy volunteers). In dogs, it was possible to attain sufficient systemic exposures to result in immunosuppression characteristic of systemic JAK inhibition, but at high AUC margins (43×) compared with systemic exposures observed at the highest tested dose in humans. No adverse findings were observed in the gastrointestinal tract or systemic tissues. Izencitinib did not affect male or female fertility. Izencitinib did not affect embryonic development in rats and rabbits as commonly reported with systemic JAK inhibition, consistent with low maternal systemic concentrations (2-6× plasma Cave:JAK IC50 ratio, 10-33× nonclinical:clinical AUC margin) and negligible fetal exposures. In conclusion, the izencitinib gut-selective approach resulted in minimal systemic findings in nonclinical species at pharmacologic, clinically relevant systemic exposures, highlighting the impact of organ-selectivity in reducing systemic safety findings.


Asunto(s)
Quinasas Janus , Naftiridinas , Nitrilos , Administración Oral , Animales , Perros , Desarrollo Embrionario/efectos de los fármacos , Femenino , Voluntarios Sanos , Humanos , Enfermedades Inflamatorias del Intestino , Quinasas Janus/antagonistas & inhibidores , Masculino , Naftiridinas/farmacología , Naftiridinas/toxicidad , Nitrilos/farmacología , Nitrilos/toxicidad , Embarazo , Conejos , Ratas , Reproducción/efectos de los fármacos , Pruebas de Toxicidad
10.
J Crohns Colitis ; 14(9): 1202-1213, 2020 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-32161949

RESUMEN

BACKGROUND AND AIMS: Oral systemic pan-Janus kinase [JAK] inhibition is effective for ulcerative colitis [UC] but is limited by toxicities. We describe preclinical to clinical translation of TD-1473-an oral gut-selective pan-JAK inhibitor-from in vitro characterization through a Phase 1b study in patients with UC. METHODS: TD-1473 JAK inhibition potency was evaluated in vitro; plasma pharmacokinetics, safety and efficacy were assessed in mice. In a first-time-in-human study, plasma pharmacokinetics and safety were assessed after single and multiple [14 days] ascending doses administered orally to healthy subjects. The Phase 1b study randomized patients with moderately to severely active UC to receive once-daily oral TD-1473 20, 80 or 270 mg, or placebo for 28 days. Plasma and colonic tissue concentrations were measured; safety was assessed; and efficacy was evaluated by UC clinical parameters, disease-surrogate biomarkers, endoscopy, histology and colonic tissue JAK signalling. RESULTS: TD-1473 exhibited potent pan-JAK inhibitory activity in vitro. Oral TD-1473 administration to mice achieved high, biologically active colonic tissue concentrations with low plasma exposure and decreased oxazolone-induced colitis activity without reducing blood cell counts vs placebo. TD-1473 administration in healthy human subjects and patients with UC yielded low plasma exposure and was generally well tolerated; treatment in patients with UC resulted in biologically active colonic tissue concentrations and descriptive trends toward reduced clinical, endoscopic and histological disease activity vs placebo. CONCLUSION: Gut-selective pan-JAK inhibition with TD-1473 administration resulted in high intestinal vs plasma drug exposure, local target engagement, and trends toward reduced UC disease activity. [Clinicaltrials.gov NCT02657122, NCT02818686].


Asunto(s)
Colitis Ulcerosa , Mucosa Intestinal , Inhibidores de las Cinasas Janus , Administración Oral , Adulto , Animales , Biomarcadores Farmacológicos/análisis , Recuento de Células Sanguíneas/métodos , Colitis Ulcerosa/diagnóstico , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inmunología , Relación Dosis-Respuesta Inmunológica , Voluntarios Sanos , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Inhibidores de las Cinasas Janus/inmunología , Inhibidores de las Cinasas Janus/farmacocinética , Masculino , Ratones , Índice de Severidad de la Enfermedad , Distribución Tisular/inmunología , Investigación Biomédica Traslacional/métodos , Resultado del Tratamiento
11.
Drug Metab Dispos ; 37(6): 1259-68, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19307295

RESUMEN

Prototypic CYP3A4 inducers were tested in a pregnane X receptor (PXR) reporter gene assay, Fa2N-4 cells, HepaRG cells, and primary human hepatocytes, along with negative controls, using CYP3A4 mRNA and activity endpoints, where appropriate. Over half of the compounds tested (14 of 24) were identified as time-dependent inhibitors of CYP3A4 and high mRNA/activity ratios (>10) were consistent with CYP3A4 time-dependent inhibition for compounds such as troleandomycin, ritonavir, and verapamil. Induction response was compared between two human donors; there was an excellent correlation in the EC(50) estimates (r(2) = 0.89, p < 0.001), and a weak but statistically significant correlation was noted for maximum observed induction at an optimum concentration (E(max)) (r(2) = 0.38, p = 0.001). E(max) and EC(50) estimates determined from the PXR reporter gene assay and Fa2N-4 and HepaRG cells were compared with those from hepatocytes. Overall, EC(50) values generated using hepatocytes agreed with those generated in the PXR reporter gene assay (r(2) = 0.85, p < 0.001) and Fa2N-4 (r(2) = 0.65, p < 0.001) and HepaRG (r(2) = 0.99, p < 0.001) cells. However, E(max) values generated in hepatocytes were only significantly correlated to those determined in Fa2N-4 (r(2) = 0.33, p = 0.005) and HepaRG cells (r(2) = 0.79, p < 0.001). "Gold standard" cytochrome P450 induction data can be generated using primary human hepatocytes, but a restricted, erratic supply and interdonor variability somewhat restrict routine application within a drug discovery setting. HepaRG cells are a valuable recent addition to the armory of in vitro tools for assessing CYP3A4 induction and seem to be an excellent surrogate of primary cells.


Asunto(s)
Citocromo P-450 CYP3A/biosíntesis , Evaluación Preclínica de Medicamentos , Interacciones Farmacológicas , Células Cultivadas , Diseño de Fármacos , Inducción Enzimática/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Preparaciones Farmacéuticas , Receptor X de Pregnano , Receptores de Esteroides/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
J Clin Pharmacol ; 48(10): 1226-36, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18559487

RESUMEN

A multiexponential allometry (MA) method was developed to predict human drug clearance from preclinical data. Separate data sets containing clearances from human and preclinical species were chosen for the study. Human clearance was estimated using the MA technique according to the equation: CL = aBWb + cBWd, where CL is clearance in milliliters/minute, and a, b, c, and d are constants derived from preclinical pharmacokinetic data. Simple allometry (SA) gave the poorest prediction using any data set, and the percentage outliers remained larger than MA or monkey liver blood flow within 1.5-, 2-, and 3-fold error. Analysis of compounds common to both data sets suggested that MA could accurately predict human clearances within approximately 10% of 3-fold error. The analysis also showed that monkey is an important species for scaling, and MA is a better predictor of human clearance when the slope of SA is >0.7.


Asunto(s)
Modelos Biológicos , Preparaciones Farmacéuticas/metabolismo , Farmacocinética , Animales , Peso Corporal , Interpretación Estadística de Datos , Evaluación Preclínica de Medicamentos/métodos , Haplorrinos , Humanos , Circulación Hepática/fisiología , Especificidad de la Especie
14.
J Med Chem ; 61(3): 1061-1073, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29301085

RESUMEN

Checkpoint kinase 1 (CHK1) inhibitors are potential cancer therapeutics that can be utilized for enhancing the efficacy of DNA damaging agents. Multiple small molecule CHK1 inhibitors from different chemical scaffolds have been developed and evaluated in clinical trials in combination with chemotherapeutics and radiation treatment. Scaffold morphing of thiophene carboxamide ureas (TCUs), such as AZD7762 (1) and a related series of triazoloquinolines (TZQs), led to the identification of fused-ring bicyclic CHK1 inhibitors, 7-carboxamide thienopyridines (7-CTPs), and 7-carboxamide indoles. X-ray crystal structures reveal a key intramolecular noncovalent sulfur-oxygen interaction in aligning the hinge-binding carboxamide group to the thienopyridine core in a coplanar fashion. An intramolecular hydrogen bond to an indole NH was also effective in locking the carboxamide in the preferred bound conformation to CHK1. Optimization on the 7-CTP series resulted in the identification of lead compound 44, which displayed respectable drug-like properties and good in vitro and in vivo potency.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Descubrimiento de Drogas , Compuestos Heterocíclicos/química , Compuestos Heterocíclicos/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/química , Daño del ADN , Humanos , Indoles/química , Modelos Moleculares , Dominios Proteicos , Piridinas/química
15.
J Inflamm (Lond) ; 14: 28, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29225517

RESUMEN

BACKGROUND: An unmet need remains for safe and effective treatments to induce and maintain remission in inflammatory bowel disease (IBD) patients. The Janus kinase (JAK) inhibitor, tofacitinib, has demonstrated robust efficacy in ulcerative colitis patients although, like other systemic immunosuppressants, there may be safety concerns associated with its use. This preclinical study evaluated whether modulating intestinal inflammation via local JAK inhibition can provide efficacy without systemic immunosuppression. METHODS: The influence of tofacitinib, dosed orally or intracecally, on oxazolone-induced colitis, oxazolone or interferon-γ (IFNγ)-induced elevation of colonic phosphorylated signal transducer and activator of transcription1 (pSTAT1) levels, and basal splenic natural killer (NK) cell counts was investigated in mice. RESULTS: Tofacitinib, dosed orally or intracecally, inhibited, with similar efficacy, oxazolone-induced colitis, represented by improvements in the disease activity index and its sub-scores (body weight, stool consistency and blood content). Intracecal dosing of tofacitinib resulted in a higher colon:plasma drug exposure ratio compared to oral dosing. At equieffective oral and intracecal doses, colonic levels of tofacitinib were similar, while the plasma levels for the latter were markedly lower, consistent with a lack of effect on splenic NK cell counts. Tofacitinib, dosed orally, intracecally, or applied to the colonic lumen in vitro, produced dose-dependent, and maximal inhibition of oxazolone or IFNγ-induced STAT1 phosphorylation in the colon. CONCLUSIONS: Localized colonic JAK inhibition, by intracecal delivery of tofacitinib, provides colonic target engagement and efficacy in a mouse colitis model at doses which do not impact splenic NK cell counts. Intestinal targeting of JAK may permit separation of local anti-inflammatory activity from systemic immunosuppression, and thus provide a larger therapeutic index compared to systemic JAK inhibitors.

16.
J Med Chem ; 48(18): 5639-43, 2005 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-16134929
17.
Pharmacol Biochem Behav ; 80(3): 521-8, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15740795

RESUMEN

BMS-505130 is a potent and selective serotonin transport inhibitor; K(i) for binding to the serotonin transporter = 0.18 nM (K(i) values for binding to the norepinephrine and dopamine transporters = 4.6 and 2.1 microM, respectively). In platelet serotonin uptake studies BMS-505130 (5 mg/kg, p.o.) produced a robust inhibition of serotonin uptake. In microdialysis studies oral dosing with BMS-505130 produced a dose-dependent increase in cortical serotonin levels that reached a maximal effect of 200% above baseline at a dose of 1 mg/kg, p.o.; the peak serotonin response was transient in nature. Following oral administration, peak plasma concentrations of BMS-505130 reached Tmax at 1.6 +/- 0.7 h and then declined to concentrations <10% of Cmax within the following 6 h; plasma half-life following i.v. dosing was 0.46 +/- 0.02 h. Parallel microdialysis and pharmacokinetic studies revealed that changes in serotonin levels in the cortex mirrored changes in the brain concentration of BMS-505130. In a behavioral assay known to be sensitive to selective serotonin reuptake inhibitors (SSRIs), mouse tail suspension, BMS-505130 produced a robust response after either oral or intraperitoneal dosing. BMS-505130 exhibits a pharmacological, neurochemical and behavioral profile consistent with a potent SSRI. Moreover, BMS-505130's short half-life may be advantageous for the treatment of premature ejaculation where an acute effect to delay ejaculation followed by a relatively rapid fall in SSRI plasma concentrations might be desirable.


Asunto(s)
Suspensión Trasera/métodos , Indoles/farmacocinética , Glicoproteínas de Membrana/antagonistas & inhibidores , Moduladores del Transporte de Membrana , Proteínas de Transporte de Membrana/antagonistas & inhibidores , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Inhibidores Selectivos de la Recaptación de Serotonina/farmacocinética , Administración Oral , Animales , Células CHO , Línea Celular , Cricetinae , Ciclopropanos , Perros , Relación Dosis-Respuesta a Droga , Humanos , Indoles/administración & dosificación , Indoles/farmacología , Masculino , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley , Proteínas de Transporte de Serotonina en la Membrana Plasmática , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
18.
J Med Chem ; 57(21): 9078-95, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25286019

RESUMEN

The compounds described herein with a spirocyclic architecture fused to a benzisoxazole ring represent a new class of antibacterial agents that operate by inhibition of DNA gyrase as corroborated in an enzyme assay and by the inhibition of precursor thymidine into DNA during cell growth. Activity resided in the configurationally lowest energy (2S,4R,4aR) diastereomer. Highly active compounds against Staphylococcus aureus had sufficiently high solubility, high plasma protein free fraction, and favorable pharmacokinetics to suggest that in vivo efficacy could be demonstrated, which was realized with compound (-)-1 in S. aureus mouse infection models. A high drug exposure NOEL on oral dosing in the rat suggested that a high therapeutic margin could be achieved. Importantly, (-)-1 was not cross-resistant with other DNA gyrase inhibitors such as fluoroquinolone and aminocoumarin antibacterials. Hence, this class shows considerable promise for the treatment of infections caused by multidrug resistant bacteria, including S. aureus.


Asunto(s)
Antibacterianos/síntesis química , Barbitúricos/síntesis química , Compuestos Heterocíclicos de 4 o más Anillos/síntesis química , Isoxazoles/síntesis química , Inhibidores de Topoisomerasa II/síntesis química , Animales , Antibacterianos/farmacocinética , Antibacterianos/uso terapéutico , Barbitúricos/farmacocinética , Barbitúricos/uso terapéutico , Femenino , Fluoroquinolonas/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/farmacocinética , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Humanos , Concentración 50 Inhibidora , Isoxazoles/farmacocinética , Isoxazoles/uso terapéutico , Masculino , Ratones , Piridonas/síntesis química , Piridonas/farmacocinética , Piridonas/uso terapéutico , Ratas Wistar , Compuestos de Espiro/síntesis química , Compuestos de Espiro/farmacocinética , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Estereoisomerismo , Relación Estructura-Actividad , Inhibidores de Topoisomerasa II/farmacocinética , Inhibidores de Topoisomerasa II/uso terapéutico
19.
J Chromatogr B Analyt Technol Biomed Life Sci ; 942-943: 107-12, 2013 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-24239935

RESUMEN

Tropomyosin-related kinases (Trk) are tyrosine kinase receptors implicated in tumor proliferation, invasion, and survival signaling across a number of tumors, making them potentially attractive targets for the treatment of cancer. AZD7451 is a potent and selective inhibitor of Trk kinases currently undergoing a Phase I dose escalation in glioblastoma multiforme at the National Cancer Institute. A key part of early clinical testing for AZD7451 involves demonstrating that pharmacokinetic half-life and clinical exposures of AZD7451 are sufficient to inhibit Trk receptors in preclinical models. To address this need, an ultra sensitive analytical method was developed to measure the AZD7451 profile in human plasma. A liquid-liquid extraction recovered >80% of AZD7451 before quantitative analysis by ultra HPLC-MS/MS. A Varian Polaris(®) C18-A column and a mass transition of m/z 383.5→340.5 (m/z 389.6→342.0 for the internal standard [(2)H6]-AZD7451) was used, and a dynamic calibration range of 0.5-1000ng/mL was established, which provided a sensitive (<8.5% deviation), and precise (<6%) quantitative assay for AZD7451. AZD7451 demonstrated stability in human plasma at room temperature for 24h (<7% change) and after extraction at 4°C for 24h (<8% change), and was stable through 4 freeze/thaw cycles (<8% change). This method was used to measure AZD7451 plasma levels in clinical samples to confirm the sensitivity at several time points following AZD7451 treatment in subjects with glioblastoma.


Asunto(s)
2-Aminopurina/sangre , Antineoplásicos/sangre , Cromatografía Líquida de Alta Presión/métodos , Inhibidores de Proteínas Quinasas/sangre , Pirazoles/sangre , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Espectrometría de Masas en Tándem/métodos , 2-Aminopurina/química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Estabilidad de Medicamentos , Humanos , Modelos Lineales , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Pirazoles/química , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
20.
J Med Chem ; 55(15): 6916-33, 2012 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-22779424

RESUMEN

Novel non-fluoroquinolone inhibitors of bacterial type II topoisomerases (DNA gyrase and topoisomerase IV) are of interest for the development of new antibacterial agents that are not impacted by target-mediated cross-resistance with fluoroquinolones. N-Linked amino piperidines, such as 7a, generally show potent antibacterial activity, including against quinolone-resistant isolates, but suffer from hERG inhibition (IC(50) = 44 µM for 7a) and QT prolongation in vivo. We now disclose the finding that new analogues of 7a with reduced pK(a) due to substitution with an electron-withdrawing substituent in the piperidine moiety, such as R,S-7c, retained the Gram-positive activity of 7a but showed significantly less hERG inhibition (IC(50) = 233 µM for R,S-7c). This compound exhibited moderate clearance in dog, promising efficacy against a MRSA strain in a mouse infection model, and an improved in vivo QT profile as measured in a guinea pig in vivo model. As a result of its promising activity, R,S-7c was advanced into phase I clinical studies.


Asunto(s)
Antibacterianos/síntesis química , Dioxanos/síntesis química , Piperidinas/síntesis química , Quinolonas/síntesis química , Inhibidores de Topoisomerasa II/síntesis química , Administración Oral , Animales , Antibacterianos/farmacología , Antibacterianos/toxicidad , Disponibilidad Biológica , Topoisomerasa de ADN IV/antagonistas & inhibidores , Dioxanos/farmacología , Dioxanos/toxicidad , Perros , Farmacorresistencia Bacteriana , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Cobayas , Staphylococcus aureus Resistente a Meticilina , Ratones , Pruebas de Sensibilidad Microbiana , Piperidinas/farmacología , Piperidinas/toxicidad , Quinolonas/farmacología , Quinolonas/toxicidad , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Estereoisomerismo , Relación Estructura-Actividad , Inhibidores de Topoisomerasa II/farmacología , Inhibidores de Topoisomerasa II/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA