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.
iScience ; 27(5): 109801, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38726365

RESUMEN

Combining an immune checkpoint inhibitor with batiraxcept (AVB-S6-500), an AXL inhibitor that acts via selective binding to growth arrest-specific protein 6 (GAS6), may improve anti-tumor immunity in platinum-resistant ovarian cancer (PROC). This phase 1b trial of durvalumab in combination with escalating doses of batiraxcept enrolled patients with recurrent PROC (NCT04019288). The primary objective was to determine the toxicity profile of the combination. Eleven patients were enrolled on the trial. No dose-limiting toxicities were observed, and no objective responses were noted. Median progression free survival (PFS) was 1.81 months (95% confidence interval (CI) 1.71-2.40), and median overall survival (OS) was 4.53 months (95% CI 2.10-24.74). Batiraxcept effectively reduced serum GAS6 levels at 1-h post-treatment, resulting in trough levels below the limit of detection in all cases but one. In conclusion, the combination of batiraxcept and durvalumab was safe and tolerable but did not demonstrate anti-tumor activity in a heterogenous population of patients with recurrent PROC.

2.
J Med Chem ; 67(3): 1734-1746, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38267212

RESUMEN

Fibroblast growth factor receptor (FGFR) alterations are present as oncogenic drivers and bypass mechanisms in many forms of cancer. These alterations can include fusions, amplifications, rearrangements, and mutations. Acquired drug resistance to current FGFR inhibitors often results in disease progression and unfavorable outcomes for patients. Genomic profiling of tumors refractory to current FGFR inhibitors in the clinic has revealed several acquired driver alterations that could be the target of next generation therapeutics. Herein, we describe how structure-based drug design (SBDD) was used to enable the discovery of the potent and kinome selective pan-FGFR inhibitor KIN-3248, which is active against many acquired resistance mutations. KIN-3248 is currently in phase I clinical development for the treatment of advanced tumors harboring FGFR2 and/or FGFR3 gene alterations.


Asunto(s)
Neoplasias , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Humanos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Mutación , Progresión de la Enfermedad , Inhibidores de Proteínas Quinasas/efectos adversos , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos
3.
J Med Chem ; 67(3): 1747-1757, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38230963

RESUMEN

RAF, a core signaling component of the MAPK kinase cascade, is often mutated in various cancers, including melanoma, lung, and colorectal cancers. The approved inhibitors were focused on targeting the BRAFV600E mutation that results in constitutive activation of kinase signaling through the monomeric protein (Class I). However, these inhibitors also paradoxically activate kinase signaling of RAF dimers, resulting in increased MAPK signaling in normal tissues. Recently, significant attention has turned to targeting RAF alterations that activate dimeric signaling (class II and III BRAF and NRAS). However, the discovery of a potent and selective inhibitor with biopharmaceutical properties suitable to sustain robust target inhibition in the clinical setting has proven challenging. Herein, we report the discovery of exarafenib (15), a highly potent and selective inhibitor that intercepts the RAF protein in the dimer compatible αC-helix-IN conformation and demonstrates anti-tumor efficacy in preclinical models with BRAF class I, II, and III and NRAS alterations.


Asunto(s)
Melanoma , Proteínas Proto-Oncogénicas B-raf , Humanos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Línea Celular Tumoral , Melanoma/patología , Sistema de Señalización de MAP Quinasas , Mutación
4.
Mol Cell ; 56(2): 323-332, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25284223

RESUMEN

Tyrosyl-tRNA synthetase (TyrRS) is known for its essential aminoacylation function in protein synthesis. Here we report a function for TyrRS in DNA damage protection. We found that oxidative stress, which often downregulates protein synthesis, induces TyrRS to rapidly translocate from the cytosol to the nucleus. We also found that angiogenin mediates or potentiates this stress-induced translocalization. The nuclear-localized TyrRS activates transcription factor E2F1 to upregulate the expression of DNA damage repair genes such as BRCA1 and RAD51. The activation is achieved through direct interaction of TyrRS with TRIM28 to sequester this vertebrate-specific epigenetic repressor and its associated HDAC1 from deacetylating and suppressing E2F1. Remarkably, overexpression of TyrRS strongly protects against UV-induced DNA double-strand breaks in zebrafish, whereas restricting TyrRS nuclear entry completely abolishes the protection. Therefore, oxidative stress triggers an essential cytoplasmic enzyme used for protein synthesis to translocate to the nucleus to protect against DNA damage.


Asunto(s)
Núcleo Celular/metabolismo , Daño del ADN/genética , Reparación del ADN/genética , Estrés Oxidativo/genética , Tirosina-ARNt Ligasa/metabolismo , Transporte Activo de Núcleo Celular/genética , Animales , Proteína BRCA1/biosíntesis , Línea Celular Tumoral , Núcleo Celular/genética , Roturas del ADN de Doble Cadena , Factor de Transcripción E2F1/metabolismo , Activación Enzimática , Células HEK293 , Células HeLa , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Morfolinos/genética , Estructura Terciaria de Proteína , Recombinasa Rad51/biosíntesis , Proteínas Represoras/metabolismo , Ribonucleasa Pancreática/metabolismo , Proteína 28 que Contiene Motivos Tripartito , Tirosina-ARNt Ligasa/biosíntesis , Tirosina-ARNt Ligasa/genética , Regulación hacia Arriba , Pez Cebra
5.
Science ; 345(6194): 328-32, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-25035493

RESUMEN

Genetic efficiency in higher organisms depends on mechanisms to create multiple functions from single genes. To investigate this question for an enzyme family, we chose aminoacyl tRNA synthetases (AARSs). They are exceptional in their progressive and accretive proliferation of noncatalytic domains as the Tree of Life is ascended. Here we report discovery of a large number of natural catalytic nulls (CNs) for each human AARS. Splicing events retain noncatalytic domains while ablating the catalytic domain to create CNs with diverse functions. Each synthetase is converted into several new signaling proteins with biological activities "orthogonal" to that of the catalytic parent. We suggest that splice variants with nonenzymatic functions may be more general, as evidenced by recent findings of other catalytically inactive splice-variant enzymes.


Asunto(s)
Aminoacil-ARNt Sintetasas/metabolismo , Dominio Catalítico , Empalme Alternativo , Aminoacil-ARNt Sintetasas/química , Aminoacil-ARNt Sintetasas/genética , Catálisis , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Especificidad de Órganos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
6.
Bioorg Med Chem Lett ; 20(11): 3361-6, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20434334

RESUMEN

We identified a new benzothiophene containing Rho kinase inhibitor scaffold in an ultra high-throughput enzymatic activity screen. SAR studies, driven by a novel label-free cellular impedance assay, were used to derive 39, which substantially reduced intraocular pressure in a monkey model of glaucoma-associated ocular hypertension.


Asunto(s)
Modelos Animales de Enfermedad , Glaucoma/enzimología , Hipertensión Ocular/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Tiofenos/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Animales , Glaucoma/fisiopatología , Haplorrinos , Células HeLa , Humanos , Presión Intraocular/efectos de los fármacos , Hipertensión Ocular/fisiopatología
7.
Blood ; 108(8): 2814-20, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16809619

RESUMEN

Chemotactic responsiveness is crucial to neutrophil recruitment to sites of infection. During chemotaxis, highly divergent cytoskeletal programs are executed at the leading and trailing edge of motile neutrophils. The Rho family of small GTPases plays a critical role in cell migration, and recent work has focused on elucidating the specific roles played by Rac1, Rac2, Cdc42, and Rho during cellular chemotaxis. Rac GTPases regulate actin polymerization and extension of the leading edge, whereas Rho GTPases control myosin-based contraction of the trailing edge. Rac and Rho signaling are thought to crosstalk with one another, and previous research has focused on mutual inhibition of Rac and Rho signaling during chemotaxis. Indeed, polarization of neutrophils has been proposed to involve the activity of a negative feedback system where Rac activation at the front of the cell inhibits local Rho activation, and vice versa. Using primary human neutrophils and neutrophils derived from a Rac1/Rac2-null transgenic mouse model, we demonstrate here that Rac1 (and not Rac2) is essential for Rho and myosin activation at the trailing edge to regulate uropod function. We conclude that Rac plays both positive and negative roles in the organization of the Rhomyosin "backness" program, thereby promoting stable polarity in chemotaxing neutrophils.


Asunto(s)
Quimiotaxis de Leucocito/fisiología , Miosinas/metabolismo , Neuropéptidos/metabolismo , Neutrófilos/fisiología , Proteínas de Unión al GTP rac/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Polaridad Celular , Humanos , Técnicas In Vitro , Ratones , Ratones Noqueados , Ratones Transgénicos , Cadenas Ligeras de Miosina/metabolismo , Neuropéptidos/deficiencia , Neuropéptidos/genética , Neutrófilos/metabolismo , Neutrófilos/ultraestructura , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción Genética , Proteínas de Unión al GTP rac/deficiencia , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1 , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo , Proteína RCA2 de Unión a GTP
8.
Mol Biol Cell ; 15(6): 2965-77, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15047871

RESUMEN

Cytoskeletal remodeling is critical for cell adhesion, spreading, and motility. p21-activated kinase (PAK), an effector molecule of the Rho GTPases Rac and Cdc42, has been implicated in cytoskeletal remodeling and cell motility. PAK kinase activity and subcellular distribution are tightly regulated by rapid and transient localized Rac and Cdc42 activation, and by interactions mediated by adapter proteins. Here, we show that endogenous PAK is constitutively activated in certain breast cancer cell lines and that this active PAK is mislocalized to atypical focal adhesions in the absence of high levels of activated Rho GTPases. PAK localization to focal adhesions in these cells is independent of PAK kinase activity, NCK binding, or GTPase binding, but requires the association of PAK with PIX. Disruption of the PAK-PIX interaction with competitive peptides displaces PAK from focal adhesions and results in a substantial reduction in PAK hyperactivity. Moreover, disruption of the PAK-PIX interaction is associated with a dramatic decrease of PIX and paxillin in focal adhesions, indicating that PAK localization to these structures via PIX is required for the maintenance of paxillin- and PIX-containing focal adhesions. Abnormal regulation of PAK localization and activity may contribute to the tumorigenic properties of certain breast cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Adhesiones Focales/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Neoplasias de la Mama/enzimología , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas del Citoesqueleto/metabolismo , Activación Enzimática , Adhesiones Focales/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Mutación , Proteínas Oncogénicas/metabolismo , Paxillin , Fosfoproteínas/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas , Factores de Intercambio de Guanina Nucleótido Rho , Quinasas p21 Activadas , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
9.
Curr Biol ; 12(23): 2029-34, 2002 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-12477392

RESUMEN

The ability of cells to recognize and respond with directed motility to chemoattractant agents is critical to normal physiological function. Neutrophils represent the prototypic chemotactic cell in that they respond to signals initiated through the binding of bacterial peptides and other chemokines to G protein-coupled receptors with speeds of up to 30 microm/min. It has been hypothesized that localized regulation of cytoskeletal dynamics by Rho GTPases is critical to orchestrating cell movement. Using a FRET-based biosensor approach, we investigated the dynamics of Rac GTPase activation during chemotaxis of live primary human neutrophils. Rac has been implicated in establishing and maintaining the leading edge of motile cells, and we show that Rac is dynamically activated at specific locations in the extending leading edge. However, we also demonstrate activated Rac in the retracting tail of motile neutrophils. Rac activation is both stimulus and adhesion dependent. Expression of a dominant-negative Rac mutant confirms that Rac is functionally required both for tail retraction and for formation of the leading edge during chemotaxis. These data establish that Rac GTPase is spatially and temporally regulated to coordinate leading-edge extension and tail retraction during a complex motile response, the chemotaxis of human neutrophils.


Asunto(s)
Quimiotaxis de Leucocito , Neutrófilos/fisiología , Proteínas de Unión al GTP rac/sangre , Activación Enzimática , Humanos , Técnicas In Vitro , Microscopía Confocal , Neutrófilos/citología , Neutrófilos/enzimología , Proteínas de Unión al GTP rac/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA