Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 67(6): 4541-4559, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38466661

RESUMEN

The optimization of an allosteric fragment, discovered by differential scanning fluorimetry, to an in vivo MAT2a tool inhibitor is discussed. The structure-based drug discovery approach, aided by relative binding free energy calculations, resulted in AZ'9567 (21), a potent inhibitor in vitro with excellent preclinical pharmacokinetic properties. This tool showed a selective antiproliferative effect on methylthioadenosine phosphorylase (MTAP) KO cells, both in vitro and in vivo, providing further evidence to support the utility of MAT2a inhibitors as potential anticancer therapies for MTAP-deficient tumors.


Asunto(s)
Neoplasias , Humanos , Entropía , Metionina Adenosiltransferasa/metabolismo
2.
J Med Chem ; 62(21): 9918-9930, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31622099

RESUMEN

In this article, we report the discovery of a series of 5-azaquinazolines as selective IRAK4 inhibitors. From modestly potent quinazoline 4, we introduced a 5-aza substitution to mask the 4-NH hydrogen bond donor (HBD). This allowed us to substitute the core with a 2-aminopyrazole, which showed large gains in cellular potency despite the additional formal HBD. Further optimization led to 6-cyanomethyl-5-azaquinazoline 13, a selective IRAK4 inhibitor, which proved efficacious in combination with ibrutinib, while showing very little activity as a single agent up to 100 mg/kg. This contrasted to previously reported IRAK4 inhibitors that exhibited efficacy in the same model as single agents and was attributed to the enhanced specificity of 13 toward IRAK4.


Asunto(s)
Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Terapia Molecular Dirigida , Factor 88 de Diferenciación Mieloide/genética , Quinazolinas/química , Quinazolinas/farmacología , Administración Oral , Animales , Línea Celular Tumoral , Diseño de Fármacos , Femenino , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/química , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Ratones , Modelos Moleculares , Mutación , Conformación Proteica , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/administración & dosificación , Quinazolinas/farmacocinética , Ratas , Ratas Wistar , Relación Estructura-Actividad , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Bioorg Med Chem ; 26(4): 913-924, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29398441

RESUMEN

We have developed a series of orally efficacious IRAK4 inhibitors, based on a scaffold hopping strategy and using rational structure based design. Efforts to tackle low permeability and high efflux in our previously reported pyrrolopyrimidine series (Scott et al., 2017) led to the identification of pyrrolotriazines which contained one less formal hydrogen bond donor and were intrinsically more lipophilic. Further optimisation of substituents on this pyrrolotriazine core culminated with the discovery of 30 as a promising in vivo probe to assess the potential of IRAK4 inhibition for the treatment of MyD88 mutant DLBCL in combination with a BTK inhibitor. When tested in an ABC-DLBCL model with a dual MyD88/CD79 mutation (OCI-LY10), 30 demonstrated tumour regressions in combination with ibrutinib.


Asunto(s)
Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Pirroles/química , Tiazinas/química , Animales , Sitios de Unión , Células CACO-2 , Perros , Diseño de Fármacos , Semivida , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma de Células B Grandes Difuso/patología , Simulación de Dinámica Molecular , Mutación , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Permeabilidad/efectos de los fármacos , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Pirroles/farmacocinética , Pirroles/farmacología , Ratas , Relación Estructura-Actividad , Tiazinas/farmacocinética , Tiazinas/farmacología
4.
J Med Chem ; 60(24): 10071-10091, 2017 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-29172502

RESUMEN

Herein we report the optimization of a series of pyrrolopyrimidine inhibitors of interleukin-1 receptor associated kinase 4 (IRAK4) using X-ray crystal structures and structure based design to identify and optimize our scaffold. Compound 28 demonstrated a favorable physicochemical and kinase selectivity profile and was identified as a promising in vivo tool with which to explore the role of IRAK4 inhibition in the treatment of mutant MYD88L265P diffuse large B-cell lymphoma (DLBCL). Compound 28 was shown to be capable of demonstrating inhibition of NF-κB activation and growth of the ABC subtype of DLBCL cell lines in vitro at high concentrations but showed greater effects in combination with a BTK inhibitor at lower concentrations. In vivo, the combination of compound 28 and ibrutinib led to tumor regression in an ABC-DLBCL mouse model.


Asunto(s)
Antineoplásicos/farmacología , Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Administración Oral , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Cristalografía por Rayos X , Perros , Femenino , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/química , Linfoma de Células B Grandes Difuso/genética , Espectroscopía de Resonancia Magnética , Masculino , Ratones SCID , Mutación , Factor 88 de Diferenciación Mieloide/genética , Inhibidores de Proteínas Quinasas/administración & dosificación , Pirimidinas/química , Pirroles/química , Ratas Wistar , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Bioorg Med Chem Lett ; 24(14): 3026-33, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24881567

RESUMEN

Structural modifications of the left-hand side of compound 1 were identified which retained or improved potent binding to Bcl-2 and Bcl-xL in in vitro biochemical assays and had strong activity in an RS4;11 apoptotic cellular assay. For example, sulfoxide diastereomer 13 maintained good binding affinity and comparable cellular potency to 1 while improving aqueous solubility. The corresponding diastereomer (14) was significantly less potent in the cell, and docking studies suggest that this is due to a stereochemical preference for the RS versus SS sulfoxide. Appending a dimethylaminoethoxy side chain (27) adjacent to the benzylic position of the biphenyl moiety of 1 improved cellular activity by approximately three-fold, and this activity was corroborated in cell lines overexpressing Bcl-2 and Bcl-xL.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteína bcl-X/antagonistas & inhibidores , Compuestos de Anilina/química , Compuestos de Anilina/farmacología , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Solubilidad , Estereoisomerismo , Relación Estructura-Actividad , Sulfonamidas/química , Sulfonamidas/farmacología , Proteína bcl-X/metabolismo
7.
Blood ; 123(6): 905-13, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24363397

RESUMEN

Upregulation of Pim kinases is observed in several types of leukemias and lymphomas. Pim-1, -2, and -3 promote cell proliferation and survival downstream of cytokine and growth factor signaling pathways. AZD1208 is a potent, highly selective, and orally available Pim kinase inhibitor that effectively inhibits all three isoforms at <5 nM or <150 nM in enzyme and cell assays, respectively. AZD1208 inhibited the growth of 5 of 14 acute myeloid leukemia (AML) cell lines tested, and sensitivity correlates with Pim-1 expression and STAT5 activation. AZD1208 causes cell cycle arrest and apoptosis in MOLM-16 cells, accompanied by a dose-dependent reduction in phosphorylation of Bcl-2 antagonist of cell death, 4EBP1, p70S6K, and S6, as well as increases in cleaved caspase 3 and p27. Inhibition of p4EBP1 and p-p70S6K and suppression of translation are the most representative effects of Pim inhibition in sensitive AML cell lines. AZD1208 inhibits the growth of MOLM-16 and KG-1a xenograft tumors in vivo with a clear pharmacodynamic-pharmacokinetic relationship. AZD1208 also potently inhibits colony growth and Pim signaling substrates in primary AML cells from bone marrow that are Flt3 wild-type or Flt3 internal tandem duplication mutant. These results underscore the therapeutic potential of Pim kinase inhibition for the treatment of AML.


Asunto(s)
Apoptosis/efectos de los fármacos , Compuestos de Bifenilo/farmacología , Proliferación Celular/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Tiazolidinas/farmacología , Animales , Compuestos de Bifenilo/farmacocinética , Western Blotting , Ciclo Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/patología , Ratones , Ratones SCID , Inhibidores de Proteínas Quinasas/farmacocinética , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Tiazolidinas/farmacocinética , Distribución Tisular , Células Tumorales Cultivadas
8.
J Med Chem ; 55(2): 852-70, 2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-22243413

RESUMEN

Thymidylate kinase (TMK) is a potential chemotherapeutic target because it is directly involved in the synthesis of an essential component, thymidine triphosphate, in DNA replication. All reported TMK inhibitors are thymidine analogues, which might retard their development as potent therapeutics due to cell permeability and off-target activity against human TMK. A small molecule hit (1, IC(50) = 58 µM), which has reasonable inhibition potency against Pseudomonas aeruginosa TMK (PaTMK), was identified by the analysis of the binding mode of thymidine or TP(5)A in a PaTMK homology model. This hit (1) was cocrystallized with PaTMK, and several potent PaTMK inhibitors (leads, 46, 47, 48, and 56, IC(50) = 100-200 nM) were synthesized using computer-aided design approaches including virtual synthesis/screening, which was used to guide the design of inhibitors. The binding mode of the optimized leads in PaTMK overlaps with that of other bacterial TMKs but not with human TMK, which shares few common features with the bacterial enzymes. Therefore, the optimized TMK inhibitors described here should be useful for the development of antibacterial agents targeting TMK without undesired off-target effects. In addition, an inhibition mechanism associated with the LID loop, which mimics the process of phosphate transfer from ATP to dTMP, was proposed based on X-ray cocrystal structures, homology models, and structure-activity relationship results.


Asunto(s)
Antibacterianos/síntesis química , Imidazoles/síntesis química , Nucleósido-Fosfato Quinasa/antagonistas & inhibidores , Pseudomonas aeruginosa/enzimología , Piridinas/síntesis química , Pirimidinas/síntesis química , Timidina/química , Antibacterianos/química , Antibacterianos/farmacología , Cristalografía por Rayos X , Humanos , Imidazoles/química , Imidazoles/farmacología , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Conformación Molecular , Imitación Molecular , Nucleósido-Fosfato Quinasa/química , Unión Proteica , Pseudomonas aeruginosa/efectos de los fármacos , Piridinas/química , Piridinas/farmacología , Pirimidinas/química , Pirimidinas/farmacología , Alineación de Secuencia
9.
J Biomol Screen ; 15(8): 937-48, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20625181

RESUMEN

Compounds that interfere with the synthesis of either mitochondrial DNA or mtDNA-encoded proteins reduce the levels of 13 proteins essential for oxidative phosphorylation, leading to a decrease in mitochondrial adenosine triphosphate (ATP) production. Toxicity caused by these compounds is seldom identified in 24- to 72-h cytotoxicity assays due to the low turnover rates of both mtDNA and mtDNA-encoded proteins. To address this problem, the authors developed a 96-well format, high-content screening (HCS) assay that measures, in eukaryotic cells, the level of Complex IV-subunit 1, an mtDNA-encoded protein synthesized on mitochondrial ribosomes, and the level of Complex V-alpha subunit, a nuclear DNA-encoded protein synthesized on cytosolic ribosomes. The effect of several antibiotics and antiretrovirals on these 2 proteins was assessed, in transformed human liver epithelial cells, 6 days after compound treatment. The results confirmed effects of drugs known to reduce mtDNA-encoded protein levels and also revealed novel information showing that several fluoroquinolones and a macrolide, josamycin, impaired expression of mtDNA-encoded proteins. The HCS assay was robust with an average Z' factor of 0.62. The assay enables large-scale screening of compounds to identify those that potentially affect mtDNA-encoded protein levels and can be implemented within a screening paradigm to minimize compound attrition.


Asunto(s)
ADN Mitocondrial/genética , Células Eucariotas/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Proteínas Mitocondriales/metabolismo , Inhibidores de la Síntesis de la Proteína/aislamiento & purificación , Inhibidores de la Síntesis de la Proteína/farmacología , Antibacterianos/aislamiento & purificación , Antibacterianos/farmacología , Antirretrovirales/aislamiento & purificación , Antirretrovirales/farmacología , Línea Celular Transformada , ADN Mitocondrial/efectos de los fármacos , ADN Mitocondrial/metabolismo , Eficiencia , Complejo IV de Transporte de Electrones/efectos de los fármacos , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Eucariotas/metabolismo , Fluoroquinolonas/aislamiento & purificación , Fluoroquinolonas/farmacología , Humanos , Josamicina/aislamiento & purificación , Josamicina/farmacología , Hígado/efectos de los fármacos , Hígado/metabolismo , Proteínas Mitocondriales/efectos de los fármacos , Proteínas Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/efectos de los fármacos , ATPasas de Translocación de Protón Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/análisis
10.
Proc Natl Acad Sci U S A ; 106(43): 18225-30, 2009 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-19828435

RESUMEN

We report the X-ray crystal structure of a phosphodiesterase (PDE) that includes both catalytic and regulatory domains. PDE2A (215-900) crystallized as a dimer in which each subunit had an extended organization of regulatory GAF-A and GAF-B and catalytic domains connected by long alpha-helices. The subunits cross at the GAF-B/catalytic domain linker, and each side of the dimer contains in series the GAF-A and GAF-B of one subunit and the catalytic domain of the other subunit. A dimer interface extends over the entire length of the molecule. The substrate binding pocket of each catalytic domain is occluded by the H-loop. We deduced from comparisons with structures of isolated, ligand-bound catalytic subunits that the H-loop swings out to allow substrate access. However, in dimeric PDE2A (215-900), the H-loops of the two catalytic subunits pack against each other at the dimer interface, necessitating movement of the catalytic subunits to allow for H-loop movement. Comparison of the unliganded GAF-B of PDE2A (215-900) with previous structures of isolated, cGMP-bound GAF domains indicates that cGMP binding induces a significant shift in the GAF-B/catalytic domain linker. We propose that cGMP binding to GAF-B causes movement, through this linker region, of the catalytic domains, such that the H-loops no longer pack at the dimer interface and are, instead, free to swing out to allow substrate access. This increase in substrate access is proposed as the basis for PDE2A activation by cGMP and may be a general mechanism for regulation of all PDEs.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/química , Regulación Alostérica , Animales , Biocatálisis , Línea Celular , Cristalografía por Rayos X , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 2/metabolismo , Activación Enzimática , Humanos , Modelos Moleculares , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Homología Estructural de Proteína
11.
Proc Natl Acad Sci U S A ; 105(36): 13309-14, 2008 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-18757755

RESUMEN

The phosphodiesterases (PDEs) are metal ion-dependent enzymes that regulate cellular signaling by metabolic inactivation of the ubiquitous second messengers cAMP and cGMP. In this role, the PDEs are involved in many biological and metabolic processes and are proven targets of successful drugs for the treatments of a wide range of diseases. However, because of the rapidity of the hydrolysis reaction, an experimental knowledge of the enzymatic mechanisms of the PDEs at the atomic level is still lacking. Here, we report the structures of reaction intermediates accumulated at the reaction steady state in PDE9/crystal and preserved by freeze-trapping. These structures reveal the catalytic process of a PDE and explain the substrate specificity of PDE9 in an actual reaction and the cation requirements of PDEs in general.


Asunto(s)
Hidrolasas Diéster Fosfóricas/química , Hidrolasas Diéster Fosfóricas/metabolismo , Catálisis , Cristalografía por Rayos X , Nucleótidos de Guanina/química , Nucleótidos de Guanina/metabolismo , Humanos , Hidrólisis , Cinética , Modelos Moleculares , Mutación/genética , Hidrolasas Diéster Fosfóricas/genética , Estructura Terciaria de Proteína , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...