Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 10: 983, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31139180

RESUMEN

Infiltration of immune cells into the tumor microenvironment (TME) can regulate growth and survival of neoplastic cells, impacting tumorigenesis and tumor progression. Correlations between the number of effector immune cells present in a tumor and clinical outcomes in many human tumors, including breast, have been widely described. Current immunotherapies utilizing checkpoint inhibitors or co-stimulatory molecule agonists aim to activate effector immune cells. However, tumors often lack adequate effector cell numbers within the TME, resulting in suboptimal responses to these agents. Chemerin (RARRES2) is a leukocyte chemoattractant widely expressed in many tissues and is known to recruit innate leukocytes. CMKLR1 is a chemotactic cellular receptor for chemerin and is expressed on subsets of dendritic cells, NK cells, and macrophages. We have previously shown that chemerin acts as a tumor suppressive cytokine in mouse melanoma models by recruiting innate immune defenses into the TME. Chemerin/RARRES2 is down-regulated in many tumors, including breast, compared to normal tissue counterparts. Here, using a syngeneic orthotopic EMT6 breast carcinoma model, we show that forced overexpression of chemerin by tumor cells results in significant recruitment of NK cells and T cells within the TME. While chemerin secretion by EMT6 cells did not alter their phenotypic behavior in vitro, it did significantly suppress tumor growth in vivo. To define the cellular effectors required for this anti-tumor phenotype, we depleted NK cells or CD8+ T cells and found that either cell type is required for chemerin-dependent suppression of EMT6 tumor growth. Finally, we show significantly reduced levels of RARRES2 mRNA in human breast cancer samples compared to matched normal tissues. Thus, for the first time we have shown that increasing chemerin expression within the breast carcinoma TME can suppress growth by recruitment of NK and T cells, thereby supporting this approach as a promising immunotherapeutic strategy.


Asunto(s)
Quimiocinas/inmunología , Inmunidad Celular , Péptidos y Proteínas de Señalización Intercelular/inmunología , Leucocitos/inmunología , Neoplasias Mamarias Animales/inmunología , Proteínas de Neoplasias/inmunología , Microambiente Tumoral/inmunología , Animales , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Leucocitos/patología , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados
2.
Eur J Med Chem ; 156: 790-799, 2018 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-30055464

RESUMEN

8-Hydroxyquinolines (HQ), including clioquinol, possess cytotoxic properties and are widely used as ligands for metal-based anticancer drug research. The number and identity of substituents on the HQ can have a profound effect on activity for a variety of inorganic compounds. Ruthenium complexes of HQ exhibit radically improved potencies, and operate by a new, currently unknown, mechanism of action. To define structure-activity relationships (SAR), a family of 22 Ru(II) coordination complexes containing mono-, di- and tri-substituted hydroxyquinoline ligands were synthesized and their biological activity evaluated. The complexes exhibited promising cytotoxic activity against a cancer cell line, and the SAR data revealed the 2- and 7-positions as key sites for the incorporation of halogens to improve potency. The Ru(II) complexes potently inhibited translation, as demonstrated by an in-cell translation assay. The effects were seen at 2-15-fold higher concentrations than those required to observe cytotoxicity, suggesting that prevention of protein synthesis may be a primary, but not the exclusive mechanism for the observed cytotoxic activity.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Hidroxiquinolinas/química , Hidroxiquinolinas/farmacología , Rutenio/química , Rutenio/farmacología , Línea Celular Tumoral , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Humanos , Ligandos , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Biosíntesis de Proteínas/efectos de los fármacos , Relación Estructura-Actividad , Transcripción Genética/efectos de los fármacos
3.
Mol Ther ; 26(5): 1354-1365, 2018 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-29606504

RESUMEN

Glioblastoma (GBM) is the least treatable type of brain tumor, afflicting over 15,000 people per year in the United States. Patients have a median survival of 16 months, and over 95% die within 5 years. The chemokine receptor ACKR3 is selectively expressed on both GBM cells and tumor-associated blood vessels. High tumor expression of ACKR3 correlates with poor prognosis and potential treatment resistance, making it an attractive therapeutic target. We engineered a single chain FV-human FC-immunoglobulin G1 (IgG1) antibody, X7Ab, to target ACKR3 in human and mouse GBM cells. We used hydrodynamic gene transfer to overexpress the antibody, with efficacy in vivo. X7Ab kills GBM tumor cells and ACKR3-expressing vascular endothelial cells by engaging the cytotoxic activity of natural killer (NK) cells and complement and the phagocytic activity of macrophages. Combining X7Ab with TMZ allows the TMZ dosage to be lowered, without compromising therapeutic efficacy. Mice treated with X7Ab and in combination with TMZ showed significant tumor reduction by MRI and longer survival overall. Brain-tumor-infiltrating leukocyte analysis revealed that X7Ab enhances the activation of M1 macrophages to support anti-tumor immune response in vivo. Targeting ACKR3 with immunotherapeutic monoclonal antibodies (mAbs) in combination with standard of care therapies may prove effective in treating GBM.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Glioblastoma/inmunología , Glioblastoma/metabolismo , Receptores CXCR/antagonistas & inhibidores , Temozolomida/farmacología , Animales , Anticuerpos Monoclonales/metabolismo , Afinidad de Anticuerpos/inmunología , Antineoplásicos Inmunológicos/farmacología , Línea Celular Tumoral , Citotoxicidad Inmunológica/efectos de los fármacos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Glioblastoma/diagnóstico , Glioblastoma/mortalidad , Humanos , Imagen por Resonancia Magnética , Ratones , Mortalidad , Unión Proteica/inmunología , Receptores CXCR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Eur J Inorg Chem ; 2017(12): 1687-1694, 2017 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-29200939

RESUMEN

Ruthenium complexes capable of light-triggered cytotoxicity are appealing potential prodrugs for photodynamic therapy (PDT) and photoactivated chemotherapy (PACT). Two groups of Ru(II) polypyridyl complexes with 2-(2-pyridyl)-benzazole ligands were synthesized and investigated for their photochemical properties and anticancer activity to compare strained and unstrained systems that are likely to have different biological mechanisms of action. The structure-activity relationship was focused on the benzazole core bioisosterism and replacement of coligands in Ru(II) complexes. Strained compounds rapidly ejected the 2-(2-pyridyl)-benzazole ligand after light irradiation, and possessed strong toxicity in the HL-60 cell line both under dark and light conditions. In contrast, unstrained Ru(II) complexes were non-toxic in the absence of light, induced cytotoxicity at nanomolar concentrations after light irradiation, and are capable of light-induced DNA damage. The 90-220-fold difference in light and dark IC50 values provides a large potential therapeutic window to allow for selective targeting of cells by exposure to light.

5.
Inorg Chem ; 56(20): 12214-12223, 2017 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-28949518

RESUMEN

Light-activated compounds are powerful tools and potential agents for medical applications, as biological effects can be controlled in space and time. Ruthenium polypyridyl complexes can induce cytotoxic effects through multiple mechanisms, including acting as photosensitizers for singlet oxygen (1O2) production, generating other reactive oxygen species (ROS), releasing biologically active ligands, and creating reactive intermediates that form covalent bonds to biological molecules. A structure-activity relationship (SAR) study was performed on a series of Ru(II) complexes containing isomeric tetramethyl-substituted bipyridyl-type ligands. Three of the ligand systems studied contained strain-inducing methyl groups and created photolabile metal complexes, which can form covalent bonds to biomolecules upon light activation, while the fourth was unstrained and resulted in photostable complexes, which can generate 1O2. The compounds studied included both bis-heteroleptic complexes containing two bipyridine ligands and a third, substituted ligand and tris-homoleptic complexes containing only the substituted ligand. The photophysics, electrochemistry, photochemistry, and photobiology were assessed. Strained heteroleptic complexes were found to be more photoactive and cytotoxic then tris-homoleptic complexes, and bipyridine ligands were superior to bipyrimidine. However, the homoleptic complexes exhibited an enhanced ability to inhibit protein production in live cells. Specific methylation patterns were associated with improved activation with red light, and photolabile complexes were generally more potent cytotoxic agents than the photostable 1O2-generating compounds.


Asunto(s)
2,2'-Dipiridil/análogos & derivados , 2,2'-Dipiridil/efectos de la radiación , Complejos de Coordinación/efectos de la radiación , Rutenio/química , 2,2'-Dipiridil/síntesis química , 2,2'-Dipiridil/farmacología , Quelantes/química , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Aductos de ADN/efectos de los fármacos , Roturas del ADN , Replicación del ADN/efectos de los fármacos , Células HL-60 , Humanos , Ligandos , Luz , Metilación , Biosíntesis de Proteínas , Pirimidinas/síntesis química , Pirimidinas/química , Pirimidinas/farmacología , Pirimidinas/efectos de la radiación , Oxígeno Singlete/química , Relación Estructura-Actividad
6.
Dalton Trans ; 46(7): 2165-2173, 2017 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-28121322

RESUMEN

Cytochrome P450s are key players in drug metabolism, and overexpression in tumors is associated with significant resistance to many medicinal agents. Consequently, inhibition of P450s could serve as a strategy to restore drug efficacy. However, the widespread expression of P450s throughout the human body and the critical roles they play in various biosynthetic pathways motivates the development of P450 inhibitors capable of controlled local administration. Ruthenium complexes containing P450 inhibitors as ligands were synthesized in order to develop pro-drugs that can be triggered to release the inhibitors in a spatially and temporally controlled fashion. Upon light activation the compounds release ligands that directly bind and inhibit P450 enzymes, while the ruthenium center is able to directly damage DNA.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/química , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Daño del ADN , Rutenio/química , Rutenio/farmacología , Benceno/química , Sistema Enzimático del Citocromo P-450/química , Imidazoles/química , Modelos Moleculares , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA