Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Transl Autoimmun ; 4: 100103, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34041473

RESUMEN

Impaired interleukin-2 (IL-2) production and regulatory T-cell dysfunction have been implicated as immunological mechanisms central to the pathogenesis of multiple autoimmune and inflammatory diseases. NKTR-358, a novel regulatory T-cell stimulator, is an investigational therapeutic that selectively restores regulatory T-cell homeostasis in these diseases. We investigated NKTR-358's selectivity for regulatory T-cells, receptor-binding properties, ex vivo and in vivo pharmacodynamics, ability to suppress conventional T-cell proliferation in mice and non-human primates, and functional activity in a murine model of systemic lupus erythematosus. In vitro, NKTR-358 demonstrated decreased affinity for IL-2Rα, IL-2Rß, and IL-2Rαß compared with recombinant human IL-2 (rhIL-2). A single dose of NKTR-358 in cynomolgus monkeys produced a greater than 15-fold increase in regulatory T-cells, and the increase lasted until day 14, while daily rhIL-2 administration for 5 days only elicited a 3-fold increase, which lasted until day 7. Repeated dosing of NKTR-358 over 6 months in cynomolgus monkeys elicited cyclical, robust increases in regulatory T-cells with no loss in drug activity over the course of treatment. Regulatory T-cells isolated from NKTR-358-treated mice displayed a sustained, higher suppression of conventional T-cell proliferation than regulatory T-cells isolated from vehicle-treated mice. NKTR-358 treatment in a mouse model (MRL/MpJ-Faslpr) of systemic lupus erythematosus for 12 weeks maintained elevated regulatory T-cells for the treatment duration and ameliorated disease progression. Together, these results suggest that NKTR-358 has the ability to elicit sustained and preferential proliferation and activation of regulatory T-cells without corresponding effects on conventional T-cells, with improved pharmacokinetics compared with rhIL-2.

2.
J Med Chem ; 63(23): 14885-14904, 2020 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-33258605

RESUMEN

Overexpression of PIM 1, 2, and 3 kinases is frequently observed in many malignancies. Previously, we discovered a potent and selective pan-PIM kinase inhibitor, compound 2, currently in phase I clinical trials. In this work, we were interested in replacing the amino group on the cyclohexane ring in compound 2 with a hydroxyl group. Structure-based drug design led to cellularly potent but metabolically unstable tetra-substituted cyclohexyl diols. Efforts on the reduction of Log D by introducing polar heterocycles improved metabolic stability. Incorporating fluorine to the tetra-substituted cyclohexyl diol moiety further reduced Log D, resulting in compound 14, a cellularly potent tetra-substituted cyclohexyl diol inhibitor with moderate metabolic stability and good permeability. We also describe the development of efficient and scalable synthetic routes toward synthetically challenging tetra-substituted cyclohexyl diol compounds. In particular, intermediate 36 was identified as a versatile intermediate, enabling a large-scale synthesis of highly substituted cyclohexane derivatives.


Asunto(s)
Ciclohexanoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Línea Celular Tumoral , Ciclohexanoles/síntesis química , Ciclohexanoles/metabolismo , Humanos , Microsomas Hepáticos/metabolismo , Simulación del Acoplamiento Molecular , Estructura Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/metabolismo , Relación Estructura-Actividad
3.
Clin Cancer Res ; 22(3): 680-90, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26832745

RESUMEN

PURPOSE: Aldesleukin, recombinant human IL2, is an effective immunotherapy for metastatic melanoma and renal cancer, with durable responses in approximately 10% of patients; however, severe side effects limit maximal dosing and thus the number of patients able to receive treatment and potential cure. NKTR-214 is a prodrug of conjugated IL2, retaining the same amino acid sequence as aldesleukin. The IL2 core is conjugated to 6 releasable polyethylene glycol (PEG) chains. In vivo, the PEG chains slowly release to generate active IL2 conjugates. EXPERIMENTAL DESIGN: We evaluated the bioactivity and receptor binding of NKTR-214 and its active IL2 conjugates in vitro; the tumor immunology, tumor pharmacokinetics, and efficacy of NKTR-214 as a single agent and in combination with anti-CTLA-4 antibody in murine tumor models. Tolerability was evaluated in non-human primates. RESULTS: In a murine melanoma tumor model, the ratio of tumor-killing CD8(+) T cells to Foxp3(+) regulatory T cells was greater than 400 for NKTR-214 compared with 18 for aldesleukin, supporting preferential activation of the IL2 receptor beta over IL2 receptor alpha, due to the location of PEG molecules. NKTR-214 provides a 500-fold greater exposure of the tumor to conjugated IL2 compared with aldesleukin. NKTR-214 showed efficacy as a single agent and provided durable immunity that was resistant to tumor rechallenge in combination with anti-CTLA-4 antibody. NKTR-214 was well tolerated in non-human primates. CONCLUSIONS: These data support further evaluation of NKTR-214 in humans for a variety of tumor types, adding to the repertoire of potent and potentially curative cancer immunotherapies.


Asunto(s)
Antineoplásicos/farmacología , Interleucina-2/análogos & derivados , Neoplasias/metabolismo , Neoplasias/patología , Polietilenglicoles/farmacología , Profármacos , Receptores de Interleucina-2/metabolismo , Proteínas Recombinantes de Fusión/farmacología , Animales , Antineoplásicos/química , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Antígeno CTLA-4/antagonistas & inhibidores , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Humanos , Memoria Inmunológica , Interleucina-2/química , Interleucina-2/farmacología , Linfocitos Infiltrantes de Tumor , Masculino , Melanoma Experimental , Ratones , Modelos Moleculares , Conformación Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Polietilenglicoles/química , Unión Proteica , Receptores de Interleucina-2/química , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Carga Tumoral/efectos de los fármacos
4.
J Med Chem ; 58(21): 8373-86, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26505898

RESUMEN

Pan proviral insertion site of Moloney murine leukemia (PIM) 1, 2, and 3 kinase inhibitors have recently begun to be tested in humans to assess whether pan PIM kinase inhibition may provide benefit to cancer patients. Herein, the synthesis, in vitro activity, in vivo activity in an acute myeloid leukemia xenograft model, and preclinical profile of the potent and selective pan PIM kinase inhibitor compound 8 (PIM447) are described. Starting from the reported aminopiperidyl pan PIM kinase inhibitor compound 3, a strategy to improve the microsomal stability was pursued resulting in the identification of potent aminocyclohexyl pan PIM inhibitors with high metabolic stability. From this aminocyclohexyl series, compound 8 entered the clinic in 2012 in multiple myeloma patients and is currently in several phase 1 trials of cancer patients with hematological malignancies.


Asunto(s)
Leucemia Mieloide Aguda/tratamiento farmacológico , Ácidos Picolínicos/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Amidas/síntesis química , Amidas/química , Amidas/uso terapéutico , Animales , Línea Celular Tumoral , Halogenación , Humanos , Leucemia Mieloide Aguda/metabolismo , Ratones , Modelos Moleculares , Ácidos Picolínicos/síntesis química , Ácidos Picolínicos/química , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-pim-1/metabolismo
5.
Clin Cancer Res ; 20(7): 1834-45, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24474669

RESUMEN

PURPOSE: PIM kinases have been shown to act as oncogenes in mice, with each family member being able to drive progression of hematologic cancers. Consistent with this, we found that PIMs are highly expressed in human hematologic cancers and show that each isoform has a distinct expression pattern among disease subtypes. This suggests that inhibitors of all three PIMs would be effective in treating multiple hematologic malignancies. EXPERIMENTAL DESIGN: Pan-PIM inhibitors have proven difficult to develop because PIM2 has a low Km for ATP and, thus, requires a very potent inhibitor to effectively block the kinase activity at the ATP levels in cells. We developed a potent and specific pan-PIM inhibitor, LGB321, which is active on PIM2 in the cellular context. RESULTS: LGB321 is active on PIM2-dependent multiple myeloma cell lines, where it inhibits proliferation, mTOR-C1 signaling and phosphorylation of BAD. Broad cancer cell line profiling of LGB321 demonstrates limited activity in cell lines derived from solid tumors. In contrast, significant activity in cell lines derived from diverse hematological lineages was observed, including acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), multiple myeloma and non-Hodgkin lymphoma (NHL). Furthermore, we demonstrate LGB321 activity in the KG-1 AML xenograft model, in which modulation of pharmacodynamics markers is predictive of efficacy. Finally, we demonstrate that LGB321 synergizes with cytarabine in this model. CONCLUSIONS: We have developed a potent and selective pan-PIM inhibitor with single-agent antiproliferative activity and show that it synergizes with cytarabine in an AML xenograft model. Our results strongly support the development of Pan-PIM inhibitors to treat hematologic malignancies.


Asunto(s)
Neoplasias Hematológicas/terapia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-pim-1/genética , Proteínas Proto-Oncogénicas/genética , Animales , Línea Celular Tumoral , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patología , Humanos , Ratones , Fosforilación , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-pim-1/antagonistas & inhibidores , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Blood ; 122(9): 1610-20, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23818547

RESUMEN

Multiple myeloma (MM) is the second most common hematologic malignancy. Despite recent treatment advances, it remains incurable. Here, we report that Pim2 kinase expression is highly elevated in MM cells and demonstrate that it is required for MM cell proliferation. Functional interference of Pim2 activity either by short hairpin RNAs or by a potent and selective small-molecule inhibitor leads to significant inhibition of MM cell proliferation. Pim inhibition results in a significant decrease of mammalian target of rapamycin C1 (mTOR-C1) activity, which is critical for cell proliferation. We identify TSC2, a negative regulator of mTOR-C1, as a novel Pim2 substrate and show that Pim2 directly phosphorylates TSC2 on Ser-1798 and relieves the suppression of TSC2 on mTOR-C1. These findings support Pim2 as a promising therapeutic target for MM and define a novel Pim2-TSC2-mTOR-C1 pathway that drives MM proliferation.


Asunto(s)
Proliferación Celular , Mieloma Múltiple/patología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Supresoras de Tumor/metabolismo , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Modelos Biológicos , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Complejos Multiproteicos/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/genética , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Piridinas/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cell Endocrinol ; 280(1-2): 6-12, 2008 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-17945411

RESUMEN

Epidermal gamma delta T cells (gammadeltaT) and Langerhans cells (LC) are immune cells altered by exposure to ultraviolet radiation (UVB), a powerful stressor resulting in immune suppression. Prolactin (PRL) has been characterized as an immunomodulator, particularly during stress. In this study, we have asked whether separate administration of the 2 major forms of prolactin, unmodified and phosphorylated, to groups of 15 mice (3 experiments, each with 5 mice per treatment group) affected the number and morphology of these epidermal immune cells under control conditions, and following UV-irradiation. Under control conditions, both PRLs reduced the number of gammadeltaT, but a molecular mimic of phosphorylated PRL (S179D PRL) was more effective, resulting in a 30% reduction. In the irradiated group, however, S179D PRL was protective against a UV-induced reduction in gammadeltaT number and change in morphology (halved the reduction and normalized the morphology). In addition, S179D PRL, but not unmodified (U-PRL), maintained a normal LC:gammadeltaT ratio and sustained the dendritic morphology of LC after UV exposure. These findings suggest that S179D PRL may have an overall protective effect, countering UV-induced cellular alterations in the epidermis.


Asunto(s)
Epidermis/efectos de los fármacos , Factores Inmunológicos/farmacología , Células de Langerhans/efectos de los fármacos , Prolactina/farmacología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/efectos de los fármacos , Rayos Ultravioleta , Animales , Epidermis/metabolismo , Epidermis/efectos de la radiación , Factores Inmunológicos/administración & dosificación , Factores Inmunológicos/metabolismo , Bombas de Infusión Implantables , Células de Langerhans/metabolismo , Células de Langerhans/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C3H , Fosforilación , Prolactina/administración & dosificación , Prolactina/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , Linfocitos T/metabolismo , Linfocitos T/efectos de la radiación
8.
Trends Immunol ; 28(5): 207-12, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17395538

RESUMEN

During tumorigenesis, selective pressure drives tumor cells to develop several strategies that enable growth and propagation. Transformed cells produce or elicit factors that provide growth signals, nutrients and a favorable tumor microenvironment. In addition, tumor cells can evade elimination by the immune system by several mechanisms, including developing resistance to T cell-induced apoptosis or the local expression of immune-modulatory molecules and cytokines. Recently, we described a role for the cytokine interleukin (IL)-23 in promoting tumor incidence and growth. In addition, IL-23 not only stimulates neutrophil and macrophage infiltration, but also promotes angiogenesis and inflammatory mediators in the tumor microenvironment. IL-23 antagonizes IL-12 and interferon gamma, both of which are essential cytokines for cytotoxic immune responses, and controls the influx and activity of anti-tumor effector lymphocytes. We suggest that IL-23 inflicts a repurposing of the adaptive cytotoxic effector response away from anti-tumor immunity ('sword') and towards proinflammatory and proangiogenic effector pathways that nourish the tumor ('plowshare'). Consequently, IL-23 enables the persistence of the recognized tumor cells, accompanied by tumor-associated inflammation. This concept can explain tumor growth in the presence of large quantities of tumor-specific T cells.


Asunto(s)
Vigilancia Inmunológica , Inflamación/patología , Interleucina-23/inmunología , Neoplasias/inmunología , Neoplasias/patología , Animales , Linfocitos T CD8-positivos/inmunología , Humanos , Interleucina-17/metabolismo , Interleucina-23/metabolismo , Ratones , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neovascularización Patológica/inmunología
9.
Nature ; 442(7101): 461-5, 2006 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-16688182

RESUMEN

Chronic inflammation has long been associated with increased incidence of malignancy and similarities in the regulatory mechanisms have been suggested for more than a century. Infiltration of innate immune cells, elevated activities of matrix metalloproteases and increased angiogenesis and vasculature density are a few examples of the similarities between chronic and tumour-associated inflammation. Conversely, the elimination of early malignant lesions by immune surveillance, which relies on the cytotoxic activity of tumour-infiltrating T cells or intra-epithelial lymphocytes, is thought to be rate-limiting for the risk to develop cancer. Here we show a molecular connection between the rise in tumour-associated inflammation and a lack of tumour immune surveillance. Expression of the heterodimeric cytokine interleukin (IL)-23, but not of its close relative IL-12, is increased in human tumours. Expression of these cytokines antagonistically regulates local inflammatory responses in the tumour microenvironment and infiltration of intra-epithelial lymphocytes. Whereas IL-12 promotes infiltration of cytotoxic T cells, IL-23 promotes inflammatory responses such as upregulation of the matrix metalloprotease MMP9, and increases angiogenesis but reduces CD8 T-cell infiltration. Genetic deletion or antibody-mediated elimination of IL-23 leads to increased infiltration of cytotoxic T cells into the transformed tissue, rendering a protective effect against chemically induced carcinogenesis. Finally, transplanted tumours are growth-restricted in hosts depleted for IL-23 or in IL-23-receptor-deficient mice. Although many strategies for immune therapy of cancer attempt to stimulate an immune response against solid tumours, infiltration of effector cells into the tumour tissue often appears to be a critical hurdle. We show that IL-23 is an important molecular link between tumour-promoting pro-inflammatory processes and the failure of the adaptive immune surveillance to infiltrate tumours.


Asunto(s)
Interleucinas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Animales , Linfocitos T CD8-positivos/inmunología , División Celular , Predisposición Genética a la Enfermedad , Humanos , Inflamación/genética , Inflamación/patología , Interleucina-12/biosíntesis , Interleucina-12/inmunología , Interleucina-23 , Subunidad p19 de la Interleucina-23 , Interleucinas/biosíntesis , Interleucinas/deficiencia , Interleucinas/genética , Ratones , Ratones Endogámicos C57BL , Neoplasias/genética , Neoplasias/inmunología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...