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.
Mol Cancer Ther ; 20(9): 1723-1734, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34224361

RESUMEN

A recombinant Newcastle Disease Virus (NDV), encoding either a human (NDVhuGM-CSF, MEDI5395) or murine (NDVmuGM-CSF) GM-CSF transgene, combined broad oncolytic activity with the ability to significantly modulate genes related to immune functionality in human tumor cells. Replication in murine tumor lines was significantly diminished relative to human tumor cells. Nonetheless, intratumoral injection of NDVmuGM-CSF conferred antitumor effects in three syngeneic models in vivo; with efficacy further augmented by concomitant treatment with anti-PD-1/PD-L1 or T-cell agonists. Ex vivo immune profiling, including T-cell receptor sequencing, revealed profound immune-contexture changes consistent with priming and potentiation of adaptive immunity and tumor microenvironment (TME) reprogramming toward an immune-permissive state. CRISPR modifications rendered CT26 tumors significantly more permissive to NDV replication, and in this setting, NDVmuGM-CSF confers immune-mediated effects in the noninjected tumor in vivo Taken together, the data support the thesis that MEDI5395 primes and augments cell-mediated antitumor immunity and has significant utility as a combination partner with other immunomodulatory cancer treatments.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Inmunomodulación , Inmunoterapia/métodos , Virus de la Enfermedad de Newcastle/genética , Viroterapia Oncolítica/instrumentación , Microambiente Tumoral , Animales , Apoptosis , Proliferación Celular , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Mol Cancer Ther ; 20(3): 541-552, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33653945

RESUMEN

Resistance to antibody-drug conjugates (ADCs) has been observed in both preclinical models and clinical studies. However, mechanisms of resistance to pyrrolobenzodiazepine (PBD)-conjugated ADCs have not been well characterized and thus, this study was designed to investigate development of resistance to PBD dimer warheads and PBD-conjugated ADCs. We established a PBD-resistant cell line, 361-PBDr, by treating human breast cancer MDA-MB-361 cells with gradually increasing concentrations of SG3199, the PBD dimer released from the PBD drug-linker tesirine. 361-PBDr cells were over 20-fold less sensitive to SG3199 compared with parental cells and were cross-resistant to other PBD warhead and ADCs conjugated with PBDs. Proteomic profiling revealed that downregulation of Schlafen family member 11 (SLFN11), a putative DNA/RNA helicase, sensitizing cancer cells to DNA-damaging agents, was associated with PBD resistance. Confirmatory studies demonstrated that siRNA knockdown of SLFN11 in multiple tumor cell lines conferred reduced sensitivity to SG3199 and PBD-conjugated ADCs. Treatment with EPZ011989, an EZH2 inhibitor, derepressed SLFN11 expression in 361-PBDr and other SLFN11-deficient tumor cells, and increased sensitivity to PBD and PBD-conjugated ADCs, indicating that the suppression of SLFN11 expression is associated with histone methylation as reported. Moreover, we demonstrated that combining an ataxia telangiectasia and Rad3-related protein (ATR) inhibitor, AZD6738, with SG3199 or PBD-based ADCs led to synergistic cytotoxicity in either resistant 361-PBDr cells or cells that SLFN11 was knocked down via siRNA. Collectively, these data provide insights into potential development of resistance to PBDs and PBD-conjugated ADCs, and more importantly, inform strategy development to overcome such resistance.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Benzodiazepinas/metabolismo , Proteínas Nucleares/metabolismo , Pirroles/metabolismo , Regulación hacia Abajo , Resistencia a Antineoplásicos , Femenino , Humanos , Transfección
3.
J Immunother Cancer ; 5(1): 63, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28807001

RESUMEN

BACKGROUND: T-cell checkpoint blockade and MEK inhibitor combinations are under clinical investigation. Despite progress elucidating the immuno-modulatory effects of MEK inhibitors as standalone therapies, the impact of MEK inhibition on the activity of T-cell checkpoint inhibitors remains incompletely understood. Here we sought to characterize the combined effects of MEK inhibition and anti-CTLA-4 mAb (anti-CTLA-4) therapy, examining effects on both T-cells and tumor microenvironment (TME). METHODS: In mice, the effects of MEK inhibition, via selumetinib, and anti-CTLA-4 on immune responses to keyhole limpet haemocyanin (KLH) immunization were monitored using ex vivo functional assays with splenocytes. In a KRAS-mutant CT26 mouse colorectal cancer model, the impact on the tumor microenvironment (TME) and the spleen were evaluated by flow cytometry. The TME was further examined by gene expression and immunohistochemical analyses. The combination and sequencing of selumetinib and anti-CTLA-4 were also evaluated in efficacy studies using the CT26 mouse syngeneic model. RESULTS: Anti-CTLA-4 enhanced the generation of KLH specific immunity following KLH immunization in vivo; selumetinib was found to reduce, but did not prevent, this enhancement of immune response by anti-CTLA-4 in vivo. In the CT26 mouse model, anti-CTLA-4 treatment led to higher expression levels of the immunosuppressive mediators, Cox-2 and Arg1 in the TME. Combination of anti-CTLA-4 with selumetinib negated this up-regulation of Cox-2 and Arg1, reduced the frequency of CD11+ Ly6G+ myeloid cells, and led to the accumulation of differentiating monocytes at the Ly6C+ MHC+ intermediate state in the tumor. We also report that MEK inhibition had limited impact on anti-CTLA-4-mediated increases in T-cell infiltration and T-cell activation in CT26 tumors. Finally, we show that pre-treatment, but not concurrent treatment, with selumetinib enhanced the anti-tumor activity of anti-CTLA-4 in the CT26 model. CONCLUSION: These data provide evidence that MEK inhibition can lead to changes in myeloid cells and immunosuppressive factors in the tumor, thus potentially conditioning the TME to facilitate improved response to anti-CTLA-4 treatment. In summary, the use of MEK inhibitors to alter the TME as an approach to enhance the activities of immune checkpoint inhibitors warrants further investigation in clinical trials.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Bencimidazoles/administración & dosificación , Neoplasias Colorrectales/tratamiento farmacológico , Microambiente Tumoral/efectos de los fármacos , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales Humanizados , Bencimidazoles/farmacología , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Reprogramación Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Immunol Res ; 5(1): 29-41, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27923825

RESUMEN

Murine syngeneic tumor models are critical to novel immuno-based therapy development, but the molecular and immunologic features of these models are still not clearly defined. The translational relevance of differences between the models is not fully understood, impeding appropriate preclinical model selection for target validation, and ultimately hindering drug development. Across a panel of commonly used murine syngeneic tumor models, we showed variable responsiveness to immunotherapies. We used array comparative genomic hybridization, whole-exome sequencing, exon microarray analysis, and flow cytometry to extensively characterize these models, which revealed striking differences that may underlie these contrasting response profiles. We identified strong differential gene expression in immune-related pathways and changes in immune cell-specific genes that suggested differences in tumor immune infiltrates between models. Further investigation using flow cytometry showed differences in both the composition and magnitude of the tumor immune infiltrates, identifying models that harbor "inflamed" and "non-inflamed" tumor immune infiltrate phenotypes. We also found that immunosuppressive cell types predominated in syngeneic mouse tumor models that did not respond to immune-checkpoint blockade, whereas cytotoxic effector immune cells were enriched in responsive models. A cytotoxic cell-rich tumor immune infiltrate has been correlated with increased efficacy of immunotherapies in the clinic, and these differences could underlie the varying response profiles to immunotherapy between the syngeneic models. This characterization highlighted the importance of extensive profiling and will enable investigators to select appropriate models to interrogate the activity of immunotherapies as well as combinations with targeted therapies in vivo Cancer Immunol Res; 5(1); 29-41. ©2016 AACR.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Animales , Antígeno B7-H1/antagonistas & inhibidores , Antígeno CTLA-4/antagonistas & inhibidores , Hibridación Genómica Comparativa , Variaciones en el Número de Copia de ADN , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Exoma , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunomodulación/efectos de los fármacos , Inmunomodulación/genética , Ratones , Terapia Molecular Dirigida , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Transducción de Señal/efectos de los fármacos , Transcriptoma , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
5.
Mol Immunol ; 40(10): 733-41, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14644099

RESUMEN

MHC class I heavy chains (HC) that fail to acquire a mature conformation in the endoplasmic reticulum (ER) as a result of defective folding or assembly with beta2-microglobulin, or lack of appropriate peptide cargo are retrotranslocated through the Sec61 channel to the cytosol for degradation by proteasomes. The mechanisms involved in ER retrotranslocation of HC are as yet incompletely understood. Using a microsomal system, we characterized the molecular requirements for the release of HC into the soluble fraction. Extraction of ubiquitinated HC was facilitated by cytosol, or by addition of proteins that stabilized the membrane association of the cytoplasmic ATPase p97. Functional proteasomes were not needed for HC mobilization. ATP supply to the ER lumen was found to be an essential factor since an inhibitor of the ATP importing pump in the ER membrane blocked HC release. Also non-hydrolyzable ATP analogs delivered to the ER lumen facilitated HC export suggesting that ATP binding by ER chaperones rather than ATP hydrolysis is involved.


Asunto(s)
Adenosina Trifosfato/metabolismo , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Adenosina Trifosfatasas/metabolismo , Transporte Biológico Activo , Cisteína Endopeptidasas/metabolismo , Citosol/inmunología , Citosol/metabolismo , Estabilidad de Enzimas , Glicosilación , Antígenos HLA/química , Antígenos HLA/metabolismo , Antígenos de Histocompatibilidad Clase I/química , Humanos , Técnicas In Vitro , Microsomas/inmunología , Microsomas/metabolismo , Complejos Multienzimáticos/metabolismo , Proteínas Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal
6.
Rapid Commun Mass Spectrom ; 17(5): 455-62, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12590394

RESUMEN

A surface for the capture of biotin-tagged proteins on matrix-assisted laser desorption/ionisation (MALDI) targets has been investigated. Binding of a poly-L-lysine poly(ethylene glycol)-biotin polymer to glass and gold surfaces has been demonstrated using dual wavelength interferometry. Biotinylated proteins were captured onto this surface using tetrameric neutravidin as a multivalent bridging molecule. Biotin tagging of proteins was achieved by chemical biotinylation or by expressing a protein with a biotinylation consensus sequence in E. coli. The specificity of the surface for biotin-tagged proteins allowed the purification of biotin-tagged glutathione-S-transferase from a bacterial lysate directly onto a MALDI target. Subsequently, the protein was digested on the MALDI target and a protein fingerprint analysis confirmed its presence directly, but no E. coli proteins were detected. Therefore, we conclude that this surface is highly specific for the capture of biotin-labelled proteins and has low non-specific binding properties for non-biotinylated proteins. Furthermore, protein-protein interactions using biotinylated lectins were investigated, and the selective capture of the glycoprotein fetuin with wheat germ agglutinin was demonstrated. Also, immobilised Arachis hypogea agglutinin recognised a minor asialo component of this glycoprotein on the array. The high affinity immobilisation of proteins onto this surface allowed effective desalting procedures to be used which improved the desorption of high molecular weight proteins. Another aspect of this surface is that a highly ordered coupling of the analyte can be achieved which eliminates the search for the sweet spot and allows the creation of densely packed protein microarrays for use in mass spectrometry.


Asunto(s)
Polilisina/análogos & derivados , Análisis por Matrices de Proteínas/métodos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/instrumentación , Arachis/química , Biotina/metabolismo , Mapeo Peptídico , Polietilenglicoles/química , Polilisina/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Aglutininas del Germen de Trigo/química , alfa-Fetoproteínas/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...