Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
ACS Med Chem Lett ; 10(11): 1554-1560, 2019 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-31749910

RESUMEN

A medicinal chemistry effort focused on identifying a structurally diverse candidate for phosphoinositide 3-kinase delta (PI3Kδ) led to the discovery of clinical candidate INCB050465 (20, parsaclisib). The unique structure of 20 contains a pyrazolopyrimidine hinge-binder in place of a purine motif that is present in other PI3Kδ inhibitors, such as idelalisib (1), duvelisib (2), and INCB040093 (3, dezapelisib). Parsaclisib (20) is a potent and highly selective inhibitor of PI3Kδ with drug-like ADME properties that exhibited an excellent in vivo profile as demonstrated through pharmacokinetic studies in rats, dogs, and monkeys and through pharmacodynamic and efficacy studies in a mouse Pfeiffer xenograft model.

2.
Clin Cancer Res ; 25(1): 300-311, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30206163

RESUMEN

PURPOSE: Bromodomain and extraterminal domain (BET) proteins regulate the expression of many cancer-associated genes and pathways; BET inhibitors have demonstrated activity in diverse models of hematologic and solid tumors. We report the preclinical characterization of INCB054329, a structurally distinct BET inhibitor that has been investigated in phase I clinical trials. EXPERIMENTAL DESIGN: We used multiple myeloma models to investigate vulnerabilities created by INCB054329 treatment that could inform rational combinations. RESULTS: In addition to c-MYC, INCB054329 decreased expression of oncogenes FGFR3 and NSD2/MMSET/WHSC1, which are deregulated in t(4;14)-rearranged cell lines. The profound suppression of FGFR3 sensitized the t(4;14)-positive cell line OPM-2 to combined treatment with a fibroblast growth factor receptor inhibitor in vivo. In addition, we show that BET inhibition across multiple myeloma cell lines resulted in suppressed interleukin (IL)-6 Janus kinase-signal transducers and activators of transcription (JAK-STAT) signaling. INCB054329 displaced binding of BRD4 to the promoter of IL6 receptor (IL6R) leading to reduced levels of IL6R and diminished signaling through STAT3. Combination with JAK inhibitors (ruxolitinib or itacitinib) further reduced JAK-STAT signaling and synergized to inhibit myeloma cell growth in vitro and in vivo. This combination potentiated tumor growth inhibition in vivo, even in the MM1.S model of myeloma that is not intrinsically sensitive to JAK inhibition alone. CONCLUSIONS: Preclinical data reveal insights into vulnerabilities created in myeloma cells by BET protein inhibition and potential strategies that can be leveraged in clinical studies to enhance the activity of INCB054329.


Asunto(s)
Proteínas de Ciclo Celular/genética , Mieloma Múltiple/tratamiento farmacológico , Compuestos Orgánicos/farmacología , Receptores de Interleucina-6/genética , Factor de Transcripción STAT3/genética , Factores de Transcripción/genética , Animales , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Xenoinjertos , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Quinasas Janus/genética , Ratones , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Unión Proteica/efectos de los fármacos , Proteínas/antagonistas & inhibidores , Proteínas/genética , Proteínas Proto-Oncogénicas c-myc/genética , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/genética , Proteínas Represoras/genética , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/antagonistas & inhibidores
3.
Drug Metab Dispos ; 38(8): 1277-85, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20421447

RESUMEN

An investigation was conducted to follow up on the apparent species-dependent toxicity reported for 6-(6-(1-methyl-1H-pyrazol-4-yl)-[1,2,4]triazolo[4,3-b]pyridazin-3-ylthio)quinoline (SGX523), a mesenchymal-epithelial transition factor (c-MET) inhibitor that entered clinical development for the treatment of solid tumors. Patients treated with SGX523 exhibited compromised renal function presumably resulting from crystal deposits in renal tubules. Our independent metabo'lite profiling of SGX523 indicates that a major NADPH-independent, late-eluting metabolite [6-(6-(1-methyl-1H-pyrazol-4-yl)-[1,2,4]triazolo[4,3-b]pyridazin-3-ylthio)quinolin-2(1H)-one (M11)] was generated by monkey and human liver S-9, and to a lesser extent by rat S-9, whereas M11 was absent in dog S-9 incubations. We confirmed the identity of M11 as 2-quinolinone-SGX523. Experiments with various molybdenum hydroxylase inhibitors showed that aldehyde oxidase (AO), and not xanthine oxidase, metabolized SGX523 to M11 in monkey and human liver cytosol. In addition, the oxygen incorporated into M11 was derived from water rather than atmospheric oxygen, corroborating M11 formation via AO. After oral dosing in monkeys, metabolite profiling of plasma and urine showed that SGX523 was indeed metabolized to M11 and its N-demethyl analog (M8). In urine, M11 levels were approximately 70-fold greater than that of SGX523, and the solubility of M11 in urine was only 3% of that of SGX523. In summary, SGX523 is metabolized by AO in a species-specific manner to a markedly less-soluble metabolite, M11. We propose that M11 is likely involved in the observed obstructive nephropathy reported in clinical studies. Moreover, this study illustrates the need to conduct thorough metabolic evaluations early in drug development to select the most relevant nonclinical species for toxicological evaluation.


Asunto(s)
Aldehído Oxidasa/metabolismo , Piridazinas/metabolismo , Triazoles/metabolismo , Animales , Citosol/metabolismo , Perros , Humanos , Hígado/metabolismo , Macaca fascicularis , Microsomas Hepáticos/metabolismo , Oxígeno , Piridazinas/farmacocinética , Piridazinas/toxicidad , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Triazoles/farmacocinética , Triazoles/toxicidad
4.
Drug Metab Dispos ; 36(6): 1102-10, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18347085

RESUMEN

The role of the intestine in the elimination of (2R)-2-{(3R)-3-amino-3-[4-(2-methylquinolin-4-ylmethoxy)phenyl]-2-oxopyrrolidin-1-yl}-N-hydroxy-4-methylpentanamide (DPC 333), a potent inhibitor of tissue necrosis factor alpha-converting enzyme, was investigated in mice and rats in vivo and in vitro. In Madine-Darby canine kidney cells stably transfected with P-glycoprotein (P-gp) and DPC 333, the transport from B-->A reservoirs exceeded the transport from A-->B by approximately 7-fold. In Caco-2 monolayers and isolated rat ileal mucosa, DPC 333 was transported from basolateral to apical reservoirs in a concentration-dependent, saturable manner, and transport was blocked by N-(4-[2-(1,2,3,4-tetrahydro-6,7-dimethoxy-2-isoquinolinyl)ethyl]-phenyl)-9,10-dihydro-5-methoxy-9-oxo-4-acridine carboxamide (GF120918), confirming the contribution of P-gp/breast cancer resistance protein in B-->A efflux of DPC 333. In quantitative whole body autoradiography studies with [(14)C]DPC 333 in mice and rats, radioactivity was distributed throughout the small intestine in both species. In GF120918-pretreated bile duct-cannulated rats, radioactivity in feces was reduced 60%. Using the in situ perfused rat intestine model, approximately 20% of an i.v. dose of [(14)C]DPC 333 was measured in the intestinal lumen within 3 h postdose, 12% as parent. Kinetic analysis of data suggested that excreted DPC 333 may be further metabolized in the gut. Intestinal clearance was 0.2 to 0.35 l/h/kg. The above data suggest that in the rodent the intestine serves as an organ of DPC 333 excretion, mediated in part by the transporter P-gp.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Mucosa Intestinal/metabolismo , Quinolinas/farmacocinética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Células CACO-2 , Línea Celular , Perros , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Quinolinas/sangre , Ratas , Ratas Sprague-Dawley , Transfección
5.
Drug Metab Dispos ; 35(10): 1916-25, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17656469

RESUMEN

DPC 333 ((2R)-2-((3R)-3-amino-3{4-[2-methyl-4-quinolinyl) methoxy] phenyl}-2-oxopyrrolidinyl)-N-hydroxy-4-methylpentanamide)) is a potent and selective inhibitor of tumor necrosis factor (TNF)-alpha-converting enzyme (TACE). It significantly inhibits lipopolysaccharide-induced soluble TNF-alpha production in blood from rodents, chimpanzee, and human, with IC(50) values ranging from 17 to 100 nM. In rodent models of endotoxemia, DPC 333 inhibited the production of TNF-alpha in a dose-dependent manner, with an oral ED(50) ranging from 1.1 to 6.1 mg/kg. Oral dosing of DPC 333 at 5.5 mg/kg daily for 2 weeks in a rat collagen antibody-induced arthritis model suppressed the maximal response by approximately 50%. DPC 333 was distributed widely to tissues including the synovium, the site of action for antiarthritic drugs. Pharmacokinetic and pharmacodynamic studies in chimpanzee revealed a systemic clearance of 0.4 l/h/kg, a V(ss) of 0.6 l/kg, an oral bioavailability of 17%, and an ex vivo IC(50) for the suppression of TNF-alpha production of 55 nM (n = 1). In a phase I clinical trial with male volunteers after single escalating doses of oral DPC 333, the terminal half-life was between 3 and 6 h and the ex vivo IC(50) for suppressing TNF-alpha production was 113 nM. Measurement of the suppression of TNF-alpha production ex vivo may serve as a good biomarker in evaluating the therapeutic efficacy of TACE inhibitors. Overall, the pharmacological profiles of DPC 333 support the notion that suppression of TNF-alpha with TACE inhibitors like DPC 333 may provide a novel approach in the treatment of various inflammatory diseases including rheumatoid arthritis, via control of excessive TNF-alpha production.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Antiinflamatorios/farmacocinética , Antiinflamatorios/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Endotoxemia/tratamiento farmacológico , Quinolinas/farmacocinética , Quinolinas/uso terapéutico , Proteína ADAM17 , Adulto , Animales , Antiinflamatorios/sangre , Artritis Experimental/sangre , Artritis Experimental/patología , Perros , Método Doble Ciego , Endotoxemia/sangre , Endotoxemia/inducido químicamente , Femenino , Humanos , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos BALB C , Pan troglodytes , Quinolinas/sangre , Ratas , Ratas Endogámicas , Líquido Sinovial/química , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/sangre
6.
Drug Metab Dispos ; 35(6): 835-40, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17332143

RESUMEN

DPC 333 [(2R)-2-{(3R)-3-amino-3-[4-(2-methylquinolin-4-ylmethoxy)phenyl]-2-oxopyrrolidin-1-yl}-N-hydroxy-4-methylpentanamide] is a potent human tumor necrosis factor alpha-converting enzyme inhibitor with potential therapeutic implications for rheumatoid arthritis. Methotrexate (MTX), a drug for the treatment of rheumatoid arthritis, is eliminated primarily unchanged via renal and biliary excretion in humans as well as in rats and dogs. The objective of the present study was to investigate the potential effect of DPC 333 on the disposition of MTX. In dogs, DPC 333 administered orally at 1.7 mg/kg 15 min before the intravenous administration of [14C]MTX (0.5 mg/kg) did not alter the plasma concentration-time profile of MTX; however, the total amount of radioactivity excreted in urine increased from 58.7% to 92.2% of the dose, and the renal clearance increased from 1.8 ml/min/kg to 2.9 ml/min/kg, suggesting a decrease in MTX disposition via biliary excretion. The biliary excretion of MTX was investigated in isolated perfused livers prepared from wild-type and TR(-) [multidrug resistance-associated protein 2 (Mrp2)-deficient] Wistar rats in the absence and presence of DPC 333. Mrp2-mediated biliary excretion of MTX was confirmed with 95.8% and 5.1% of MTX recovered in the bile of wild-type and TR(-) Wistar rats, respectively. DPC 333 at an initial perfusate concentration of 50 microM completely blocked the biliary excretion of MTX, but not the clearance from perfusate, in both wild-type and TR(-) rats. These results suggest that the enhanced renal elimination of MTX may be due to the potent inhibition of biliary excretion and active renal reabsorption by DPC 333 and/or its metabolites.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Antiinflamatorios/farmacología , Antirreumáticos/farmacocinética , Metotrexato/farmacocinética , Quinolinas/farmacología , Proteína ADAM17 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Animales Modificados Genéticamente , Antirreumáticos/sangre , Bilis/metabolismo , Perros , Interacciones Farmacológicas , Humanos , Técnicas In Vitro , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Metotrexato/sangre , Ratas , Ratas Wistar
7.
Bioorg Med Chem Lett ; 16(10): 2699-704, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16516466

RESUMEN

A novel series of achiral TNF-alpha converting enzyme (TACE) inhibitors has been discovered. These compounds exhibited activities from 0.35 to 11nM in a porcine TACE assay and inhibited TNF-alpha production in an LPS-stimulated whole blood assay with an IC(50) value of 23nM for the most potent one. They also have excellent selectivities over related metalloproteases including aggrecanases.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/farmacología , Proteína ADAM17 , Animales , Línea Celular , Ciclización , Humanos , Ratones , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacocinética , Estereoisomerismo , Relación Estructura-Actividad
8.
Bioorg Med Chem Lett ; 16(9): 2357-63, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16473009

RESUMEN

The synthesis, evaluation, and structure-activity relationships of a series of succinoyl lactam inhibitors of the Alzheimer's disease gamma-secretase are described. Beginning with a screening hit with broad proteinase activity, optimization provided compounds with both high selectivity for inhibition of gamma-secretase and high potency in cellular assays of A beta reduction. The SAR and early in vivo properties of this series of inhibitors will be presented.


Asunto(s)
Caprolactama/química , Endopeptidasas/efectos de los fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Succinatos/química , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/enzimología , Secretasas de la Proteína Precursora del Amiloide , Animales , Ácido Aspártico Endopeptidasas , Caprolactama/análogos & derivados , Línea Celular , Perros , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Endopeptidasas/química , Inhibidores Enzimáticos/química , Humanos , Conformación Molecular , Estereoisomerismo , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA