Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Pharmacol ; 13: 829063, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35795558

RESUMEN

Pharmacokinetic/pharmacodynamic (PK/PD) modeling was performed to quantitatively integrate preclinical pharmacology and toxicology data for determining the therapeutic index (TI) of an interleukin-10 (IL-10) fragment crystallizable (Fc) fusion protein. Mouse Fc fused with mouse IL-10 (mFc-mIL-10) was studied in mice for antitumor efficacy, and the elevation of interleukin-18 (IL-18) was examined as a PD biomarker. The in vivo mFc-mIL-10 EC50 for the IL-18 induction was estimated to be 2.4 nM, similar to the in vitro receptor binding affinity (Kd) of 3.2 nM. The IL-18 induction was further evaluated in cynomolgus monkeys, where the in vivo induction EC50 by a human IL-10 human Fc-fusion protein (hFc-hIL-10) was 0.08 nM vs. 0.3 nM measured as the in vitro Kd. The extent of the IL-18 induction correlated with mouse antitumor efficacy and was used to connect mouse efficacy to that in monkeys. The PD-based efficacious dose projected in monkeys was comparable to the results obtained using a PK-based method in which mouse efficacious exposure was targeted and corrected for affinity differences between the species. Furthermore, PK/PD relationships were developed for anemia and thrombocytopenia in monkeys treated with hFc-hIL-10, with thrombocytopenia predicted to be dose-limiting toxicity. Using quantitative pharmacology and toxicology information obtained through modeling work in the same species, the TI of hFc-hIL-10 in monkeys was determined to be 2.4 (vs. PD-based efficacy) and 1.2-3 (vs. PK-based efficacy), indicating a narrow safety margin. The model-based approaches were proven valuable to the developability assessment of the IL-10 Fc-fusion protein.

2.
Cancer Cell ; 33(5): 843-852.e4, 2018 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-29657128

RESUMEN

Combination immune checkpoint blockade has demonstrated promising benefit in lung cancer, but predictors of response to combination therapy are unknown. Using whole-exome sequencing to examine non-small-cell lung cancer (NSCLC) treated with PD-1 plus CTLA-4 blockade, we found that high tumor mutation burden (TMB) predicted improved objective response, durable benefit, and progression-free survival. TMB was independent of PD-L1 expression and the strongest feature associated with efficacy in multivariable analysis. The low response rate in TMB low NSCLCs demonstrates that combination immunotherapy does not overcome the negative predictive impact of low TMB. This study demonstrates the association between TMB and benefit to combination immunotherapy in NSCLC. TMB should be incorporated in future trials examining PD-(L)1 with CTLA-4 blockade in NSCLC.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Secuenciación del Exoma/métodos , Ipilimumab/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Nivolumab/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/genética , Femenino , Humanos , Inmunoterapia , Neoplasias Pulmonares/genética , Masculino , Persona de Mediana Edad , Mutación , Supervivencia sin Progresión
3.
Bioorg Med Chem Lett ; 28(4): 732-736, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29398543

RESUMEN

A novel series of o-phenylenediamine-based inhibitors of indoleamine 2,3-dioxygenase (IDO) has been identified. IDO is a heme-containing enzyme, overexpressed in the tumor microenvironment of many cancers, which can contribute to the suppression of the host immune system. Synthetic modifications to a previously described diarylether series resulted in an additional degree of molecular diversity which was exploited to afford compounds that demonstrated significant potency in the HeLa human cervical cancer IDO1 assay. .


Asunto(s)
Inhibidores Enzimáticos/farmacología , Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Fenilendiaminas/farmacología , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Células HeLa , Humanos , Microsomas Hepáticos/metabolismo , Fenilendiaminas/síntesis química , Fenilendiaminas/química , Fenilendiaminas/metabolismo , Relación Estructura-Actividad
5.
Cancer Res ; 69(7): 3213-20, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19293180

RESUMEN

Hypoxia inducible factors (HIF) are critical mediators of the cellular response to decreased oxygen tension and are overexpressed in a number of tumors. Although HIF1alpha and HIF2alpha share a high degree of sequence homology, recent work has shown that the two alpha subunits can have contrasting and tissue-specific effects on tumor growth. To directly compare the role of each HIFalpha subunit in spontaneous tumorigenesis, we bred a mouse model of expanded HIF2alpha expression and Hif1alpha(+/-) mice to homozygotes for the R270H mutation in p53. Here, we report that p53(R270H/R270H) mice, which have not been previously described, develop a unique tumor spectrum relative to p53(R270H/-) mice, including a high incidence of thymic lymphomas. Heterozygosity for Hif1alpha significantly reduced the incidence of thymic lymphomas observed in this model. Moreover, reduced Hif1alpha levels correlated with decreased stabilization of activated Notch1 and expression of the Notch target genes, Dtx1 and Nrarp. These observations uncover a novel role for HIF1alpha in Notch pathway activation during T-cell lymphomagenesis.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Linfoma/genética , Neoplasias del Timo/genética , Factores de Edad , Alelos , Animales , Femenino , Genes p53 , Heterocigoto , Linfoma/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mutación , Receptor Notch1/biosíntesis , Receptor Notch1/genética , Neoplasias del Timo/metabolismo
6.
Clin Cancer Res ; 14(24): 8123-31, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19088027

RESUMEN

PURPOSE: Angiogenesis is a critical step in the establishment, growth, and metastasis of solid tumors, and combination of antiangiogenic agents with chemotherapy is an attractive therapeutic option. We investigated the potential of ixabepilone, the first in a new class of antineoplastic agents known as epothilones, to synergize with antiangiogenic agents to inhibit tumor growth. EXPERIMENTAL DESIGN: In vitro and in vivo cytotoxicity of ixabepilone as single agent and in combination with two targeted antiangiogenic agents, bevacizumab or sunitinib, were examined in preclinical tumor models. Direct effects of the agents against endothelial cells was also examined and compared with the effects of paclitaxel as single agent and in combination with bevacizumab. RESULTS: Ixabepilone showed robust synergistic antitumor activity in combination with bevacizumab and sunitinib in preclinical in vivo models derived from breast, colon, lung, and kidney cancers. The synergistic antitumor effect was greater with ixabepilone compared with paclitaxel. Furthermore, ixabepilone was more effective than paclitaxel at killing endothelial cells expressing P-glycoprotein in vitro and inhibiting endothelial cell proliferation and tumor angiogenesis in vivo. CONCLUSIONS: Ixabepilone may enhance the antitumor effects of antiangiogenic therapy by direct cytotoxicity and also indirectly via the killing of tumor-associated endothelial cells. Given that ixabepilone has reduced susceptibility to drug efflux pumps compared with taxanes, these data may explain the increased antiangiogenic and antitumor activity of ixabepilone in combination with antiangiogenic agents. Phase II studies to assess the efficacy and safety of ixabepilone plus bevacizumab in locally recurrent or metastatic breast cancer are planned.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Antineoplásicos/administración & dosificación , Epotilonas/administración & dosificación , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/análisis , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Bevacizumab , Sinergismo Farmacológico , Células Endoteliales/efectos de los fármacos , Epotilonas/farmacología , Femenino , Humanos , Indoles/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Pirroles/administración & dosificación , Sunitinib , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Genes Dev ; 21(9): 1037-49, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17437992

RESUMEN

Mammalian cells are believed to have a cell-intrinsic ability to increase glucose metabolism in response to hypoxia. Here we show that the ability of hematopoietic cells to up-regulate anaerobic glycolysis in response to hypoxia is dependent on receptor-mediated signal transduction. In the absence of growth factor signaling, hematopoietic cells fail to express hypoxia-inducible transcription factor (Hif-1alpha) mRNA. Growth factor-deprived hematopoietic cells do not engage in glucose-dependent anabolic synthesis and neither express Hif-1alpha mRNA nor require HIF-1alpha protein to regulate cell survival in response to hypoxia. However, HIF-1alpha is adaptive for the survival of growth factor-stimulated cells, as suppression of HIF-1alpha results in death when growing cells are exposed to hypoxia. Growth factor-dependent HIF-1alpha expression reprograms the intracellular fate of glucose, resulting in decreased glucose-dependent anabolic synthesis and increased lactate production, an effect that is enhanced when HIF-1alpha protein is stabilized by hypoxia. Together, these data suggest that HIF-1alpha contributes to the regulation of growth factor-stimulated glucose metabolism even in the absence of hypoxia.


Asunto(s)
Glucólisis/genética , Glucólisis/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Aerobiosis , Anaerobiosis , Animales , Secuencia de Bases , Hipoxia de la Célula , Línea Celular , Proliferación Celular , Supervivencia Celular , Eliminación de Gen , Glucosa/metabolismo , Sustancias de Crecimiento/metabolismo , Sistema Hematopoyético/citología , Sistema Hematopoyético/metabolismo , Interleucina-3/metabolismo , Ratones , Ratones Mutantes , Modelos Biológicos , Células 3T3 NIH , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal
8.
Mol Cell Biol ; 26(9): 3514-26, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16611993

RESUMEN

Transcriptional responses to hypoxia are primarily mediated by hypoxia-inducible factors (HIFs), HIF-1alpha and HIF-2alpha. The HIF-1alpha and HIF-2alpha subunits are structurally similar in their DNA binding and dimerization domains but differ in their transactivation domains, implying they may have unique target genes and require distinct transcriptional cofactors. Our previous results demonstrated that HIF-1alpha and HIF-2alpha regulate distinct target genes. Here, we report that HIF-2alpha is not transcriptionally active in embryonic stem (ES) cells, as well as possible inhibition by a HIF-2alpha-specific transcriptional repressor. Using DNA microarray analysis of hypoxia-inducible genes in wild-type (WT), Hif-1alpha(-)(/)(-), and Hif-2alpha(-)(/)(-) ES cells, we show that HIF-1alpha induces a large number of both confirmed and novel hypoxia-inducible genes, while HIF-2alpha does not activate any of its previously described targets. We further demonstrate that inhibition of HIF-2alpha function occurs at the level of transcription cofactor recruitment to endogenous target gene promoters. Overexpression of WT and, notably, a DNA-binding-defective HIF-2alpha mutant restores endogenous HIF-2alpha protein activity, suggesting that ES cells express a HIF-2alpha-specific corepressor that can be titrated by overexpressed HIF-2alpha protein. HIF-2alpha repression may explain why patients with mutations in the VHL tumor suppressor gene display cancerous lesions in specific tissue types.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/genética , Células Madre/metabolismo , Factores de Transcripción/fisiología , Animales , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células Cultivadas , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Activación Transcripcional , Regulación hacia Arriba/genética
9.
Genes Dev ; 20(5): 557-70, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16510872

RESUMEN

The division, differentiation, and function of stem cells and multipotent progenitors are influenced by complex signals in the microenvironment, including oxygen availability. Using a genetic "knock-in" strategy, we demonstrate that targeted replacement of the oxygen-regulated transcription factor HIF-1alpha with HIF-2alpha results in expanded expression of HIF-2alpha-specific target genes including Oct-4, a transcription factor essential for maintaining stem cell pluripotency. We show that HIF-2alpha, but not HIF-1alpha, binds to the Oct-4 promoter and induces Oct-4 expression and transcriptional activity, thereby contributing to impaired development in homozygous Hif-2alpha KI/KI embryos, defective hematopoietic stem cell differentiation in embryoid bodies, and large embryonic stem cell (ES)-derived tumors characterized by altered cellular differentiation. Furthermore, loss of HIF-2alpha severely reduces the number of embryonic primordial germ cells, which require Oct-4 expression for survival and/or maintenance. These results identify Oct-4 as a HIF-2alpha-specific target gene and indicate that HIF-2alpha can regulate stem cell function and/or differentiation through activation of Oct-4, which in turn contributes to HIF-2alpha's tumor promoting activity.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Hipoxia de la Célula/fisiología , Desarrollo Embrionario/fisiología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre/fisiología , Teratoma/metabolismo , Alelos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Hipoxia de la Célula/genética , Transformación Celular Neoplásica , Regulación hacia Abajo , Desarrollo Embrionario/genética , Femenino , Inmunohistoquímica , Ratones , Ratones Desnudos , Modelos Genéticos , Factor 3 de Transcripción de Unión a Octámeros/genética , Embarazo , ARN Mensajero/metabolismo , Células Madre/citología , Células Madre/metabolismo , Teratoma/genética , Teratoma/patología , Factor de Crecimiento Transformador alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
Cancer Res ; 65(6): 2277-86, 2005 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15781641

RESUMEN

Hypoxia-inducible factors (HIF) are essential transcriptional regulators that mediate adaptation to hypoxic stress in rapidly growing tissues such as tumors. HIF activity is regulated by hypoxic stabilization of the related HIF-1alpha and HIF-2alpha subunits, which are frequently overexpressed in cancer cells. To assess the relative tumor-promoting functions of HIF-1alpha and HIF-2alpha directly, we replaced HIF-1alpha expression with HIF-2alpha by creating a novel "knock-in" allele at the Hif-1alpha locus through homologous recombination in primary murine embryonic stem cells. Compared with controls, s.c. teratomas derived from knock-in embryonic stem cells were larger and more proliferative, had increased microvessel density, and exhibited increased expression of vascular endothelial growth factor, transforming growth factor-alpha, and cyclin D1. These and other data indicate that HIF-2alpha promotes tumor growth more effectively than HIF-1alpha in multiple contexts.


Asunto(s)
Alelos , Teratoma/genética , Teratoma/patología , Transactivadores/genética , Factores de Transcripción/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular/genética , Procesos de Crecimiento Celular/genética , Transformación Celular Neoplásica/genética , Embrión de Mamíferos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones , Ratones Desnudos , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Células Madre/citología , Células Madre/metabolismo , Células Madre/fisiología , Teratoma/irrigación sanguínea , Teratoma/metabolismo , Transactivadores/biosíntesis , Factores de Transcripción/biosíntesis , Factor de Crecimiento Transformador alfa/biosíntesis , Factor A de Crecimiento Endotelial Vascular/biosíntesis
11.
BMC Evol Biol ; 5: 19, 2005 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-15725349

RESUMEN

BACKGROUND: Yellowfin and skipjack tuna are globally distributed in the world's tropical and sub-tropical oceans. Since little, if any, migration of these fishes occurs between the Atlantic and Indo-Pacific Oceans, one might expect to see genetic differences between sub-populations in these ocean basins. However, yellowfin and skipjack tuna have extremely large population sizes. Thus, the rate of genetic drift should be slower than that observed for other tunas. RESULTS: Low levels of genetic differentiation were observed between Atlantic and Pacific samples of yellowfin tuna. In contrast, no genetic differentiation was observed between Atlantic and Pacific samples of skipjack tuna. CONCLUSION: Much lower levels of genetic differentiation were found among sub-populations of yellowfin tuna compared to those observed for other large tunas, probably due to the large population size of yellowfin tuna. Since skipjack tuna appear to have even larger population sizes, it is not surprising that no genetic differentiation was detected between Atlantic and Pacific samples of these fish.


Asunto(s)
Evolución Molecular , Atún/genética , Alelos , Análisis de Varianza , Animales , Océano Atlántico , Disparidad de Par Base , Citocromos b/genética , ADN Mitocondrial/genética , Frecuencia de los Genes , Variación Genética , Genética de Población , Modelos Genéticos , Océano Pacífico , Polimorfismo de Longitud del Fragmento de Restricción , Densidad de Población , Especificidad de la Especie
12.
Curr Top Dev Biol ; 62: 37-54, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15522738

RESUMEN

Cellular oxygen (O2) concentrations are tightly regulated to maintain ATP levels required for metabolic reactions in the human body. Responses to changes in O2 concentrations are primarily regulated by the transcription factor hypoxia inducible factor (HIF). HIF activates transcription of genes that increase systemic O2 delivery or provide cellular metabolic adaptation under conditions of hypoxia. HIF activity is essential for embryogenesis and various processes in postnatal life, and therefore, HIF levels need to be precisely controlled. Abnormal HIF expression is related to numerous diseases of the vascular system, including heart disease, cancer, and chronic obstructive pulmonary disease.


Asunto(s)
Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/metabolismo , Hipoxia de la Célula/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Animales , Vasos Sanguíneos/embriología , Vasos Sanguíneos/patología , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Homeostasis , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neoplasias/metabolismo , Proteínas Nucleares/genética , Oxígeno/metabolismo , Factores de Transcripción/genética
13.
Genesis ; 39(2): 115-21, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15170697

RESUMEN

Mutations in the notch ligand delta-like 3 have been identified in both the pudgy mouse (Dll3(pu); Kusumi et al.: Nat Genet 19:274-278, 1998) and the human disorder spondylocostal dysostosis (SCD; Bulman et al.: Nat Genet 24:438-441, 2000), and a targeted mutation has been generated (Dll3(neo); Dunwoodie et al.: Development 129:1795-1806, 2002). Vertebral and rib malformations deriving from defects in somitic patterning are key features of these disorders. In the mouse, notch pathway genes such as Lfng, Hes1, Hes7, and Hey2 display dynamic patterns of expression in paraxial mesoderm, cycling in synchrony with somite formation (Aulehla and Johnson: Dev Biol 207:49-61, 1999; Forsberg et al.: Curr Biol 8:1027-1030, 1998; Jouve et al.: Development 127:1421-1429, 2000; McGrew et al.: Curr Biol 8:979-982, 1998; Nakagawa et al.: Dev Biol 216:72-84, 1999). We report here that the Dll3(pu) mutation has different effects on the expression of cycling (Lfng and Hes7) and stage-specific genes (Hey3 and Mesp2). This suggests a more complex situation than a single oscillatory mechanism in somitogenesis and provides an explanation for the unique radiological features of the human DLL3-type of SCD.


Asunto(s)
Disostosis/embriología , Regulación del Desarrollo de la Expresión Génica , Proteínas de la Membrana/genética , Mutación , Somitos/metabolismo , Columna Vertebral/anomalías , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Glicosiltransferasas/metabolismo , Secuencias Hélice-Asa-Hélice , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana/metabolismo , Mesodermo/citología , Mesodermo/patología , Ratones , Ratones Noqueados , Proteínas Represoras/metabolismo , Transducción de Señal , Somitos/patología , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...