Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Arthritis Rheumatol ; 73(12): 2206-2218, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34423919

RESUMEN

OBJECTIVE: To investigate the role of PF-06650833, a highly potent and selective small-molecule inhibitor of interleukin-1-associated kinase 4 (IRAK4), in autoimmune pathophysiology in vitro, in vivo, and in the clinical setting. METHODS: Rheumatoid arthritis (RA) inflammatory pathophysiology was modeled in vitro through 1) stimulation of primary human macrophages with anti-citrullinated protein antibody immune complexes (ICs), 2) RA fibroblast-like synoviocyte (FLS) cultures stimulated with Toll-like receptor (TLR) ligands, as well as 3) additional human primary cell cocultures exposed to inflammatory stimuli. Systemic lupus erythematosus (SLE) pathophysiology was simulated in human neutrophils, dendritic cells, B cells, and peripheral blood mononuclear cells stimulated with TLR ligands and SLE patient ICs. PF-06650833 was evaluated in vivo in the rat collagen-induced arthritis (CIA) model and the mouse pristane-induced and MRL/lpr models of lupus. Finally, RNA sequencing data generated with whole blood samples from a phase I multiple-ascending-dose clinical trial of PF-06650833 were used to test in vivo human pharmacology. RESULTS: In vitro, PF-06650833 inhibited human primary cell inflammatory responses to physiologically relevant stimuli generated with RA and SLE patient plasma. In vivo, PF-06650833 reduced circulating autoantibody levels in the pristane-induced and MRL/lpr murine models of lupus and protected against CIA in rats. In a phase I clinical trial (NCT02485769), PF-06650833 demonstrated in vivo pharmacologic action pertinent to SLE by reducing whole blood interferon gene signature expression in healthy volunteers. CONCLUSION: These data demonstrate that inhibition of IRAK4 kinase activity can reduce levels of inflammation markers in humans and provide confidence in the rationale for clinical development of IRAK4 inhibitors for rheumatologic indications.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Isoquinolinas/uso terapéutico , Lactamas/uso terapéutico , Macrófagos/efectos de los fármacos , Enfermedades Reumáticas/tratamiento farmacológico , Sinoviocitos/efectos de los fármacos , Animales , Artritis Experimental/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Humanos , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Isoquinolinas/farmacología , Lactamas/farmacología , Leucocitos Mononucleares/inmunología , Macrófagos/inmunología , Ratones , Ratas , Enfermedades Reumáticas/inmunología , Sinoviocitos/inmunología
2.
J Med Chem ; 61(19): 8597-8612, 2018 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-30113844

RESUMEN

Cytokine signaling is an important characteristic of autoimmune diseases. Many pro-inflammatory cytokines signal through the Janus kinase (JAK)/Signal transducer and activator of transcription (STAT) pathway. JAK1 is important for the γ-common chain cytokines, interleukin (IL)-6, and type-I interferon (IFN) family, while TYK2 in addition to type-I IFN signaling also plays a role in IL-23 and IL-12 signaling. Intervention with monoclonal antibodies (mAbs) or JAK1 inhibitors has demonstrated efficacy in Phase III psoriasis, psoriatic arthritis, inflammatory bowel disease, and rheumatoid arthritis studies, leading to multiple drug approvals. We hypothesized that a dual JAK1/TYK2 inhibitor will provide additional efficacy, while managing risk by optimizing selectivity against JAK2 driven hematopoietic changes. Our program began with a conformationally constrained piperazinyl-pyrimidine Type 1 ATP site inhibitor, subsequent work led to the discovery of PF-06700841 (compound 23), which is in Phase II clinical development (NCT02969018, NCT02958865, NCT03395184, and NCT02974868).


Asunto(s)
Antituberculosos/farmacología , Artritis Experimental/prevención & control , Janus Quinasa 1/antagonistas & inhibidores , Mycobacterium tuberculosis/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , TYK2 Quinasa/antagonistas & inhibidores , Tuberculosis/complicaciones , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/microbiología , Femenino , Estructura Molecular , Ratas , Ratas Endogámicas Lew , Tuberculosis/microbiología
3.
J Med Chem ; 61(3): 1130-1152, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29298069

RESUMEN

Janus kinases (JAKs) are intracellular tyrosine kinases that mediate the signaling of numerous cytokines and growth factors involved in the regulation of immunity, inflammation, and hematopoiesis. As JAK1 pairs with JAK2, JAK3, and TYK2, a JAK1-selective inhibitor would be expected to inhibit many cytokines involved in inflammation and immune function while avoiding inhibition of the JAK2 homodimer regulating erythropoietin and thrombopoietin signaling. Our efforts began with tofacitinib, an oral JAK inhibitor approved for the treatment of rheumatoid arthritis. Through modification of the 3-aminopiperidine linker in tofacitinib, we discovered highly selective JAK1 inhibitors with nanomolar potency in a human whole blood assay. Improvements in JAK1 potency and selectivity were achieved via structural modifications suggested by X-ray crystallographic analysis. After demonstrating efficacy in a rat adjuvant-induced arthritis (rAIA) model, PF-04965842 (25) was nominated as a clinical candidate for the treatment of JAK1-mediated autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/tratamiento farmacológico , Ciclobutanos/farmacología , Janus Quinasa 1/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Sulfonamidas/farmacología , Animales , Artritis Experimental/tratamiento farmacológico , Ciclobutanos/química , Ciclobutanos/farmacocinética , Ciclobutanos/uso terapéutico , Perros , Evaluación Preclínica de Medicamentos , Humanos , Concentración 50 Inhibidora , Janus Quinasa 1/química , Janus Quinasa 2/antagonistas & inhibidores , Modelos Moleculares , Conformación Proteica , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacocinética , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/química , Pirimidinas/farmacocinética , Pirimidinas/uso terapéutico , Pirroles/química , Pirroles/farmacocinética , Pirroles/uso terapéutico , Ratas , Especificidad por Sustrato , Sulfonamidas/química , Sulfonamidas/farmacocinética , Sulfonamidas/uso terapéutico , Distribución Tisular
4.
J Med Chem ; 60(13): 5521-5542, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28498658

RESUMEN

Through fragment-based drug design focused on engaging the active site of IRAK4 and leveraging three-dimensional topology in a ligand-efficient manner, a micromolar hit identified from a screen of a Pfizer fragment library was optimized to afford IRAK4 inhibitors with nanomolar potency in cellular assays. The medicinal chemistry effort featured the judicious placement of lipophilicity, informed by co-crystal structures with IRAK4 and optimization of ADME properties to deliver clinical candidate PF-06650833 (compound 40). This compound displays a 5-unit increase in lipophilic efficiency from the fragment hit, excellent kinase selectivity, and pharmacokinetic properties suitable for oral administration.


Asunto(s)
Descubrimiento de Drogas , Quinasas Asociadas a Receptores de Interleucina-1/antagonistas & inhibidores , Isoquinolinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Administración Oral , Relación Dosis-Respuesta a Droga , Humanos , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Isoquinolinas/administración & dosificación , Isoquinolinas/química , Lactamas , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
5.
Arthritis Rheum ; 64(11): 3531-42, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22899318

RESUMEN

OBJECTIVE: The mechanistic link between Janus kinase (JAK) signaling and structural damage to arthritic joints in rheumatoid arthritis (RA) is poorly understood. This study was undertaken to investigate how selective inhibition of JAK with tofacitinib (CP-690,550) affects osteoclast-mediated bone resorption in a rat adjuvant-induced arthritis (AIA) model, as well as human T lymphocyte RANKL production and human osteoclast differentiation and function. METHODS: Hind paw edema, inflammatory cell infiltration, and osteoclast-mediated bone resorption in rat AIA were assessed using plethysmography, histopathologic analysis, and immunohistochemistry; plasma and hind paw tissue levels of cytokines and chemokines (including RANKL) were also assessed. In vitro RANKL production by activated human T lymphocytes was evaluated by immunoassay, while human osteoclast differentiation and function were assessed via quantitative tartrate-resistant acid phosphatase staining and degradation of human bone collagen, respectively. RESULTS: Edema, inflammation, and osteoclast-mediated bone resorption in rats with AIA were dramatically reduced after 7 days of treatment with the JAK inhibitor, which correlated with reduced numbers of CD68/ED-1+, CD3+, and RANKL+ cells in the paws; interleukin-6 (transcript and protein) levels were rapidly reduced in paw tissue within 4 hours of the first dose, whereas it took 4-7 days of therapy for RANKL levels to decrease. Tofacitinib did not impact human osteoclast differentiation or function, but did decrease human T lymphocyte RANKL production in a concentration-dependent manner. CONCLUSION: These results suggest that the JAK inhibitor tofacitinib suppresses osteoclast-mediated structural damage to arthritic joints, and this effect is secondary to decreased RANKL production.


Asunto(s)
Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo , Quinasas Janus/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Ligando RANK/metabolismo , Animales , Artritis Experimental/inmunología , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/inmunología , Resorción Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Quinasas Janus/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Monocitos/citología , Monocitos/efectos de los fármacos , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/enzimología , Piperidinas , Ratas , Ratas Endogámicas Lew , Transducción de Señal/efectos de los fármacos , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/enzimología
6.
Neurosci Lett ; 362(2): 150-3, 2004 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-15193774

RESUMEN

Sensory irritants initiate respiratory reflexes by stimulating trigeminal sensory nerves. The vanilloid receptor (TRPV1) is expressed on sensory C fibers. The current experiments were aimed at examining the role of this receptor in mediating responses to several airborne irritants including an acidic (acetic acid), electrophilic (acrolein), and lipophilic solvent (styrene) vapor. Wild-type (C57Bl/6J) and VR1 knockout [B6.129S4-VR1(tm1jul)] mice were exposed to these irritants and breathing pattern responses were assessed by plethysmographic techniques; both wild-type and knockout animals responded similarly to the irritants. The TRPV1 antagonist iodoresiniferatoxin was also without effect on the responses to the irritants. Thus, in the C57Bl/6J mouse the TRPV1 receptor does not appear to play a major role in the stimulation of nasal trigeminal central reflex responses by these irritant air pollutants.


Asunto(s)
Contaminantes Atmosféricos/farmacología , Inhalación/efectos de los fármacos , Receptores de Droga/fisiología , Ácido Acético/farmacología , Acroleína/farmacología , Animales , Femenino , Inhalación/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Droga/deficiencia , Receptores de Droga/genética , Estireno/farmacología
7.
J Appl Physiol (1985) ; 94(4): 1563-71, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12626476

RESUMEN

The immediate responses of the upper respiratory tract (URT) to the irritants acrolein and acetic acid were examined in healthy and allergic airway-diseased C57Bl/6J mice. Acrolein (1.1 ppm) and acetic acid (330 ppm) vapors induced an immediate increase in flow resistance, as measured in the surgically isolated URT of urethane-anesthetized healthy animals. Acrolein, but not acetic acid, induced a small URT vasodilatory response. In awake spontaneously breathing mice, both vapors induced a prolonged pause at the start of expiration (a response mediated via stimulation of nasal trigeminal nerves) and an increase in total respiratory specific airway flow resistance, the magnitude of which was similar to that observed in the isolated URT. Both responses were significantly reduced in animals pretreated with large doses of capsaicin to defunctionalize sensory nerves, strongly suggesting a role for sensory nerves in development of these responses. The breathing pattern and/or obstructive responses were enhanced in mice with ovalbumin-induced allergic airway disease. These results suggest that the primary responses to acrolein and acetic acid vapors are altered breathing patterns and airway obstruction, that sensory nerves play an important role in these responses, and that these responses are enhanced in animals with allergic airway disease.


Asunto(s)
Hipersensibilidad/fisiopatología , Irritantes/farmacología , Neuronas Aferentes/fisiología , Respiración/efectos de los fármacos , Fenómenos Fisiológicos Respiratorios , Enfermedades Respiratorias/fisiopatología , Ácido Acético/administración & dosificación , Acroleína/administración & dosificación , Administración por Inhalación , Obstrucción de las Vías Aéreas/inducido químicamente , Resistencia de las Vías Respiratorias/efectos de los fármacos , Animales , Femenino , Hipersensibilidad/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Ventilación Pulmonar/efectos de los fármacos , Mecánica Respiratoria/efectos de los fármacos , Sistema Respiratorio/irrigación sanguínea , Sistema Respiratorio/efectos de los fármacos , Enfermedades Respiratorias/inmunología , Vasodilatación
8.
J Toxicol Environ Health A ; 65(23): 1999-2005, 2002 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-12490044

RESUMEN

The effect of exposure to irritant air pollutants on the development of allergic airway disease is poorly understood. This study examines the effects of the lower respiratory tract irritant, NO(2), on the outcome of ovalbumin (OVA)-induced allergic airway disease. Male and female C57Bl/6 mice were sensitized by weekly intraperitoneal (ip) OVA injections for 3 wk followed by daily 1-h OVA aerosol inhalation challenge for 3 or 10 d. Initially, mice were exposed daily for 3 d to air or 0.7 or 5 ppm NO(2) for 2 h following each OVA aerosol challenge. OVA exposure resulted in pronounced lower airway inflammation, as evidenced by a significant increase in bronchoalveolar lavage (BAL) total cellularity and eosinophil levels. BAL eosinophil levels were significantly lower in OVA-NO(2) compared to OVA-air animals. The reduction was similar at both NO(2) exposure concentrations. In a subsequent study, sensitized animals were exposed for 3 or 10 d to aerosolized OVA followed by air or 0.7 ppm NO(2). BAL eosinophils were again reduced at 3 d by OVA-NO(2) exposure compared to OVA-air mice. At 10 d the eosinophilia was virtually abolished. This reduction in OVA-induced cellular inflammation by NO(2) was confirmed by histopathological analysis. Contrary to expectations, exposure to NO(2) during the aerosol challenge to OVA dramatically diminished the outcome of allergic disease in lungs as measured by airway cellular inflammation.


Asunto(s)
Hipersensibilidad/fisiopatología , Exposición por Inhalación , Enfermedades Pulmonares/inducido químicamente , Enfermedades Pulmonares/inmunología , Dióxido de Nitrógeno/efectos adversos , Oxidantes Fotoquímicos/efectos adversos , Aerosoles , Animales , Modelos Animales de Enfermedad , Humanos , Inflamación , Enfermedades Pulmonares/veterinaria , Ratones , Ratones Endogámicos C57BL , Dióxido de Nitrógeno/administración & dosificación , Oxidantes Fotoquímicos/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...