Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cancer Immunol Res ; 12(9): 1221-1235, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-38990554

RESUMEN

Innate inflammation promotes tumor development, although the role of innate inflammatory cytokines in established human tumors is unclear. Herein, we report clinical and translational results from a phase Ib trial testing whether IL1ß blockade in human pancreatic cancer would alleviate myeloid immunosuppression and reveal antitumor T-cell responses to PD1 blockade. Patients with treatment-naïve advanced pancreatic ductal adenocarcinoma (n = 10) were treated with canakinumab, a high-affinity monoclonal human antiinterleukin-1ß (IL1ß), the PD1 blocking antibody spartalizumab, and gemcitabine/n(ab)paclitaxel. Analysis of paired peripheral blood from patients in the trial versus patients receiving multiagent chemotherapy showed a modest increase in HLA-DR+CD38+ activated CD8+ T cells and a decrease in circulating monocytic myeloid-derived suppressor cells (MDSC) by flow cytometry for patients in the trial but not in controls. Similarly, we used patient serum to differentiate monocytic MDSCs in vitro and showed that functional inhibition of T-cell proliferation was reduced when using on-treatment serum samples from patients in the trial but not when using serum from patients treated with chemotherapy alone. Within the tumor, we observed few changes in suppressive myeloid-cell populations or activated T cells as assessed by single-cell transcriptional profiling or multiplex immunofluorescence, although increases in CD8+ T cells suggest that improvements in the tumor immune microenvironment might be revealed by a larger study. Overall, the data indicate that exposure to PD1 and IL1ß blockade induced a modest reactivation of peripheral CD8+ T cells and decreased circulating monocytic MDSCs; however, these changes did not lead to similarly uniform alterations in the tumor microenvironment.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Interleucina-1beta , Células Supresoras de Origen Mieloide , Neoplasias Pancreáticas , Receptor de Muerte Celular Programada 1 , Humanos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/inmunología , Interleucina-1beta/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Masculino , Femenino , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología , Anciano , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Desoxicitidina/administración & dosificación , Persona de Mediana Edad , Gemcitabina , Microambiente Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Paclitaxel/uso terapéutico , Paclitaxel/administración & dosificación , Paclitaxel/farmacología , Metástasis de la Neoplasia , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/patología
2.
Cancer Immunol Res ; 11(6): 777-791, 2023 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-37040466

RESUMEN

High levels of IL1ß can result in chronic inflammation, which in turn can promote tumor growth and metastasis. Inhibition of IL1ß could therefore be a promising therapeutic option in the treatment of cancer. Here, the effects of IL1ß blockade induced by the mAbs canakinumab and gevokizumab were evaluated alone or in combination with docetaxel, anti-programmed cell death protein 1 (anti-PD-1), anti-VEGFα, and anti-TGFß treatment in syngeneic and humanized mouse models of cancers of different origin. Canakinumab and gevokizumab did not show notable efficacy as single-agent therapies; however, IL1ß blockade enhanced the effectiveness of docetaxel and anti-PD-1. Accompanying these effects, blockade of IL1ß alone or in combination induced significant remodeling of the tumor microenvironment (TME), with decreased numbers of immune suppressive cells and increased tumor infiltration by dendritic cells (DC) and effector T cells. Further investigation revealed that cancer-associated fibroblasts (CAF) were the cell type most affected by treatment with canakinumab or gevokizumab in terms of change in gene expression. IL1ß inhibition drove phenotypic changes in CAF populations, particularly those with the ability to influence immune cell recruitment. These results suggest that the observed remodeling of the TME following IL1ß blockade may stem from changes in CAF populations. Overall, the results presented here support the potential use of IL1ß inhibition in cancer treatment. Further exploration in ongoing clinical studies will help identify the best combination partners for different cancer types, cancer stages, and lines of treatment.


Asunto(s)
Interleucina-1beta , Neoplasias , Microambiente Tumoral , Animales , Ratones , Línea Celular Tumoral , Docetaxel/farmacología , Inmunidad , Inmunoterapia , Neoplasias/tratamiento farmacológico , Interleucina-1beta/antagonistas & inhibidores
3.
Cancer Immunol Res ; 10(12): 1441-1461, 2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36129967

RESUMEN

Immune-stimulator antibody conjugates (ISAC) combining tumor-targeting monoclonal antibodies with immunostimulatory agents allow targeted delivery of immune activators into tumors. NJH395 is a novel, first-in-class ISAC comprising a Toll-like receptor 7 (TLR7) agonist conjugated to an anti-HER2 antibody via a noncleavable linker payload. Preclinical characterization showed ISAC-mediated activation of myeloid cells in the presence of antigen-expressing cancer cells, with antigen targeting and TLR7 agonism contributing to antitumor activity. Safety, efficacy, immunogenicity, pharmacokinetics, and pharmacodynamics were investigated in a phase I, multicenter, open-label study in patients with HER2+ non-breast advanced malignancies (NCT03696771). Data from 18 patients enrolled in single ascending dose escalation demonstrated delivery of the TLR7-agonist payload in HER2+ tumor cells and induction of type I IFN responses, which correlated with immune modulation in the tumor microenvironment. Cytokine release syndrome was a common, but manageable, drug-related adverse event. Antidrug antibodies and neuroinflammation at high doses represented significant clinical challenges. Data provide proof-of-mechanism and critical insights for novel immunotherapies.


Asunto(s)
Antineoplásicos Inmunológicos , Antineoplásicos , Inmunoconjugados , Neoplasias , Humanos , Receptor Toll-Like 7/agonistas , Inmunoconjugados/efectos adversos , Neoplasias/tratamiento farmacológico , Antineoplásicos Inmunológicos/uso terapéutico , Receptor ErbB-2 , Microambiente Tumoral
4.
Adv Exp Med Biol ; 497: 209-24, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11993734

RESUMEN

Dr. Carlen reviews the evidence that seizures may cause cell death and discusses possible strategies for preventing seizure-induced brain damage.


Asunto(s)
Epilepsia/tratamiento farmacológico , Epilepsia/patología , Fármacos Neuroprotectores/uso terapéutico , Animales , Encéfalo/patología , Epilepsia/fisiopatología , Humanos
5.
J Forensic Sci ; 58(5): 1238-1250, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23879433

RESUMEN

The operation of a motor vehicle requires the integrity of sensory, motor, and intellectual faculties. Impairment of these faculties following the consumption of alcohol has been studied extensively through laboratory, closed-course and on-road driving, and epidemiological studies. The scientific literature was reviewed critically, with a focus on low-to-moderate blood alcohol concentrations (BAC ≤ 0.100%), to identify the most reliable determinants of alcohol-impaired driving. Variables such as age, gender, driving skill, and tolerance were shown to have limited impact on impairment. It was concluded the most relevant variables are BAC and complexity of the driving task. The scientific literature provides a high degree of confidence to support the conclusion that a BAC of 0.050% impairs faculties required in the operation of a motor vehicle. Whether impairment is apparent depends upon the complexity of the driving task, which applies to both study design and actual driving.


Asunto(s)
Conducción de Automóvil , Depresores del Sistema Nervioso Central/sangre , Etanol/sangre , Análisis y Desempeño de Tareas , Accidentes de Tránsito , Factores de Edad , Consumo de Bebidas Alcohólicas/fisiopatología , Atención/fisiología , Tolerancia a Medicamentos , Movimientos Oculares/fisiología , Toxicología Forense , Humanos , Puntaje de Gravedad del Traumatismo , Destreza Motora/fisiología , Tiempo de Reacción/fisiología , Factores Sexuales , Visión Ocular/fisiología
6.
Cancer Discov ; 3(7): 742-50, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23614898

RESUMEN

The high frequency of activating RAS or BRAF mutations in cancer provides strong rationale for targeting the mitogen-activated protein kinase (MAPK) pathway. Selective BRAF and MAP-ERK kinase (MEK) inhibitors have shown clinical efficacy in patients with melanoma. However, the majority of responses are transient, and resistance is often associated with pathway reactivation of the extracellular signal-regulated kinase (ERK) signaling pathway. Here, we describe the identification and characterization of SCH772984, a novel and selective inhibitor of ERK1/2 that displays behaviors of both type I and type II kinase inhibitors. SCH772984 has nanomolar cellular potency in tumor cells with mutations in BRAF, NRAS, or KRAS and induces tumor regressions in xenograft models at tolerated doses. Importantly, SCH772984 effectively inhibited MAPK signaling and cell proliferation in BRAF or MEK inhibitor-resistant models as well as in tumor cells resistant to concurrent treatment with BRAF and MEK inhibitors. These data support the clinical development of ERK inhibitors for tumors refractory to MAPK inhibitors.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas Quinasa Quinasa PAM/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Mutación , Neoplasias/tratamiento farmacológico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
7.
Mol Cell Neurosci ; 19(2): 138-51, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11860268

RESUMEN

Lack of expression of the fragile X mental retardation protein (FMRP), due to silencing of the FMR1 gene, causes the Fragile X syndrome. Although FMRP was characterized previously to be an RNA binding protein, little is known about its function or the mechanisms underlying the Fragile X syndrome. Here we report that the alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate receptor subunit, GluR1, was decreased in the cortical synapses, but not in the hippocampus or cerebellum, of FMR1 gene knockout mice. Reduced long-term potentiation (LTP) was also found in the cortex but not in the hippocampus. Another RNA binding protein, FXR; the N-methyl-D-aspartate receptor subunit, NR2; and other learning-related proteins including c-fos, synapsin, myelin proteolipid protein, and cAMP response element binding protein were not different between FMR1 gene knockout and wild-type mice. These findings suggest that the depressed cortical GluR1 expression and LTP associated with FMRP deficiency could contribute to the Fragile X phenotype.


Asunto(s)
Corteza Cerebral/metabolismo , Regulación hacia Abajo/genética , Síndrome del Cromosoma X Frágil/metabolismo , Potenciación a Largo Plazo/genética , Proteínas del Tejido Nervioso/deficiencia , Receptores AMPA/metabolismo , Sinapsis/metabolismo , Transmisión Sináptica/genética , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/fisiopatología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Antagonistas de Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/patología , Regulación de la Expresión Génica/genética , Discapacidades para el Aprendizaje/genética , Discapacidades para el Aprendizaje/metabolismo , Discapacidades para el Aprendizaje/fisiopatología , Hígado/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Proteína Proteolipídica de la Mielina/metabolismo , Miocardio/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/metabolismo , Receptores AMPA/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/efectos de los fármacos , Sinapsinas/metabolismo , Membranas Sinápticas/metabolismo , Transmisión Sináptica/efectos de los fármacos , Tetrodotoxina/farmacología
8.
J Neurochem ; 86(3): 687-99, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12859682

RESUMEN

Chronic (18 h) exposure of cultured hippocampal slices to the type-A GABA receptor blocker, bicuculline methiodide (BMI) 10 micro m increased the levels of connexin 43 (Cx43) and connexin 32 (Cx32) mRNAs, but not connexin 26 and connexin 36, as demonstrated by RNase protection assays. The levels of Cx43 and Cx32 proteins in membrane fractions detected by western blotting were also significantly increased. Immunoblotting indicated that BMI also promoted a significant expression of the transcription protein c-fos. The rate of fluorescence recovery after photobleaching, an index of gap junctional coupling, was also significantly increased, whereas it was blocked by the gap junctional blocker, carbenoxolone (100 micro m). Extracellular recordings in CA1 stratum pyramidale, performed in BMI-free solution, demonstrated that BMI-exposed cultures possessed synaptic responses characteristic of epileptiform discharges: (i) significantly greater frequency of spontaneous epileptiform discharges, (ii) post-synaptic potentials with multiple population spikes, and (iii) significantly longer duration of primary afterdischarges. Carbenoxolone (100 micro m), but not its inactive analog, oleanolic acid (100 micro m), reversibly inhibited spontaneous and evoked epileptiform discharges. The findings of BMI-induced parallel increases in levels of gap junction expression and function, and the increase in epileptiform discharges, which were sensitive to gap junctional blockers, are consistent with the hypothesis that increased gap junctional communication plays an intrinsic role in the epileptogenic process.


Asunto(s)
Bicuculina/análogos & derivados , Epilepsia/fisiopatología , Antagonistas del GABA , Uniones Comunicantes/fisiología , Hipocampo/fisiopatología , Animales , Carbenoxolona/farmacología , Conexina 26 , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/genética , Conexinas/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Estimulación Eléctrica , Epilepsia/inducido químicamente , Recuperación de Fluorescencia tras Fotoblanqueo , Uniones Comunicantes/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Técnicas In Vitro , Masculino , Proteínas del Tejido Nervioso/metabolismo , Octanoles/farmacología , Técnicas de Placa-Clamp , Proteínas Proto-Oncogénicas c-fos/biosíntesis , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Factores de Tiempo , Proteína beta1 de Unión Comunicante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA