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.
Clin Cancer Res ; 25(9): 2783-2794, 2019 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-30765391

RESUMEN

PURPOSE: BRAF and MEK inhibitors (BRAFi and MEKi) are actively used for the treatment of metastatic melanoma in patients with BRAFV600E mutation in their tumors. However, the development of resistance to BRAFi and MEKi remains a difficult clinical challenge with limited therapeutic options available to these patients. In this study, we investigated the mechanism and potential therapeutic utility of combination BRAFi and adoptive T-cell therapy (ACT) in melanoma resistant to BRAFi. EXPERIMENTAL DESIGN: Investigations were performed in vitro and in vivo with various human melanoma cell lines sensitive and resistant to BRAFi as well as patient-derived xenografts (PDX) derived from patients. In addition, samples were evaluated from patients on a clinical trial of BRAFi in combination with ACT. RESULTS: Herein we report that in human melanoma cell lines, senstitive and resistant to BRAFi and in PDX from patients who progressed on BRAFi and MEKi therapy, BRAFi caused transient upregulation of mannose-6-phosphate receptor (M6PR). This sensitized tumor cells to CTLs via uptake of granzyme B, a main component of the cytotoxic activity of CTLs. Treatment of mice bearing resistant tumors with BRAFi enhanced the antitumor effect of patients' TILs. A pilot clinical trial of 16 patients with metastatic melanoma who were treated with the BRAFi vemurafenib followed by therapy with TILs demonstrated a significant increase of M6PR expression on tumors during vemurafenib treatment. CONCLUSIONS: BRAF-targeted therapy sensitized resistant melanoma cells to CTLs, which opens new therapeutic opportunities for the treatment of patients with BRAF-resistant disease.See related commentary by Goff and Rosenberg, p. 2682.


Asunto(s)
Melanoma , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Animales , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Ratones , Inhibidores de Proteínas Quinasas , Linfocitos T
2.
Cancer Immunol Res ; 4(4): 345-53, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26873574

RESUMEN

The checkpoint inhibitor nivolumab is active in patients with metastatic melanoma who have failed ipilimumab. In this phase I/II study, we assessed nivolumab's safety in 92 ipilimumab-refractory patients with unresectable stage III or IV melanoma, including those who experienced grade 3-4 drug-related toxicity to ipilimumab. We report long-term survival, response duration, and biomarkers in these patients after nivolumab treatment (3 mg/kg) every 2 weeks for 24 weeks, then every 12 weeks for up to 2 years, with or without a multipeptide vaccine. The response rate for ipilimumab-refractory patients was 30% (95% CI, 21%-41%). The median duration of response was 14.6 months, median progression-free survival was 5.3 months, and median overall survival was 20.6 months, when patients were followed up for a median of 16 months. One- and 2-year survival rates were 68.4% and 31.2%, respectively. Ipilimumab-naïve and ipilimumab-refractory patients showed no significant difference in survival. The 21 patients with prior grade 3-4 toxicity to ipilimumab that was managed with steroids tolerated nivolumab well, with 62% (95% CI, 38%-82%) having complete or partial responses or stabilized disease at 24 weeks. High numbers of myeloid-derived suppressor cells (MDSC) were associated with poor survival. Thus, survival and long-term safety were excellent in ipilimumab-refractory patients treated with nivolumab. Prior grade 3-4 immune-related adverse effects from ipilimumab were not indicative of nivolumab toxicities, and patients had a high overall rate of remission or stability at 24 weeks. Prospectively evaluating MDSC numbers before treatment could help assess the expected benefit of nivolumab.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/patología , Adulto , Anciano , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Biomarcadores , Estudios de Cohortes , Terapia Combinada , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Femenino , Humanos , Inmunofenotipificación , Ipilimumab , Estimación de Kaplan-Meier , Masculino , Melanoma/inmunología , Melanoma/mortalidad , Persona de Mediana Edad , Células Mieloides/inmunología , Células Mieloides/metabolismo , Metástasis de la Neoplasia , Estadificación de Neoplasias , Nivolumab , Retratamiento , Resultado del Tratamiento
3.
Bioorg Med Chem ; 21(17): 5029-38, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23890524

RESUMEN

Probes for use in time-resolved fluorescence competitive binding assays at melanocortin receptors based on the parental ligands MSH(4), MSH(7), and NDP-α-MSH were prepared by solid phase synthesis methods, purified, and characterized. The saturation binding of these probes was studied using HEK-293 cells engineered to overexpress the human melanocortin 4 receptor (hMC4R) as well as the human cholecystokinin 2 receptor (hCCK2R). The ratios of non-specific binding to total binding approached unity at high concentrations for each probe. At low probe concentrations, receptor-mediated binding and uptake was discernable, and so probe concentrations were kept as low as possible in determining Kd values. The Eu-DTPA-PEGO-MSH(4) probe exhibited low specific binding relative to non-specific binding, even at low nanomolar concentrations, and was deemed unsuitable for use in competition binding assays. The Eu-DTPA-PEGO probes based on MSH(7) and NDP-α-MSH exhibited Kd values of 27±3.9nM and 4.2±0.48nM, respectively, for binding with hMC4R. These probes were employed in competitive binding assays to characterize the interactions of hMC4R with monovalent and divalent MSH(4), MSH(7), and NDP-α-MSH constructs derived from squalene. Results from assays with both probes reflected only statistical enhancements, suggesting improper ligand spacing on the squalene scaffold for the divalent constructs. The Ki values from competitive binding assays that employed the MSH(7)-based probe were generally lower than the Ki values obtained when the probe based on NDP-α-MSH was employed, which is consistent with the greater potency of the latter probe. The probe based on MSH(7) was also competed with monovalent, divalent, and trivalent MSH(4) constructs that previously demonstrated multivalent binding in competitive binding assays against a variant of the probe based on NDP-α-MSH. Results from these assays confirm multivalent binding, but suggest a more modest increase in avidity for these MSH(4) constructs than was previously reported.


Asunto(s)
Colorantes Fluorescentes/síntesis química , Receptores de Melanocortina/metabolismo , Unión Competitiva , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Células HEK293 , Humanos , Ácido Pentético/química , Péptidos/síntesis química , Péptidos/química , Péptidos/metabolismo , Unión Proteica , Receptor de Colecistoquinina B/química , Receptor de Colecistoquinina B/genética , Receptor de Colecistoquinina B/metabolismo , Receptor de Melanocortina Tipo 4/química , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Receptores de Melanocortina/química , Técnicas de Síntesis en Fase Sólida
4.
Bioorg Med Chem Lett ; 23(7): 2061-4, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23481651

RESUMEN

A scaffold bearing eight terminal alkyne groups was synthesized from sucrose, and copies of an azide-terminated Gd-DOTA complex were attached via copper(I)-catalyzed azide-alkyne cycloaddition. The resulting contrast agent (CA) was administered by gavage to C3H mice. Passage of the CA through the gastrointestinal (GI) tract was followed by T1-weighted magnetic resonance imaging (MRI) over a period of 47h, by which time the CA had exited the GI tract. No evidence for leakage of the CA from the GI tract was observed. Thus, a new, orally administered CA for MRI of the GI tract has been developed and successfully demonstrated.


Asunto(s)
Medios de Contraste , Tracto Gastrointestinal/metabolismo , Compuestos Heterocíclicos , Imagen por Resonancia Magnética , Compuestos Organometálicos , Sacarosa , Animales , Medios de Contraste/administración & dosificación , Medios de Contraste/química , Compuestos Heterocíclicos/administración & dosificación , Compuestos Heterocíclicos/química , Modelos Lineales , Ratones , Ratones Endogámicos C3H , Estructura Molecular , Compuestos Organometálicos/administración & dosificación , Compuestos Organometálicos/química , Sacarosa/administración & dosificación , Sacarosa/química
5.
Bioconjug Chem ; 23(12): 2451-9, 2012 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-23116461

RESUMEN

The incidence of malignant melanoma is rising more rapidly than that of any other cancer in the United States. The melanocortin 1 receptor (MC1R) is overexpressed in most human melanoma metastases, thus making it a promising target for imaging and therapy of melanomas. We have previously reported the development of a peptidomimetic ligand with high specificity and affinity for MC1R. Here, we have conjugated near-infrared fluorescent dyes to the C-terminus of this ligand via lysine-mercaptopropionic acid linkers to generate MC1R specific optical probes (MC1RL-800, 0.4 nM K(i); and MC1RL-Cy5, 0.3 nM K(i)). Internalization of the imaging probe was studied in vitro by fluorescence microscopy using engineered A375/MC1R cells and B16F10 cells with endogenous MC1R expression. The in vivo tumor targeting of MC1RL-800 was evaluated by intravenous injection of probe into nude mice bearing bilateral subcutaneous A375 xenograft tumors with low MC1R expression and engineered A375/MC1R tumors with high receptor expression. Melanotic B16F10 xenografts were also studied. Fluorescence imaging showed that the agent has higher uptake values in tumors with high expression compared to low (p < 0.05), demonstrating the effect of expression levels on image contrast-to-noise. In addition, tumor uptake was significantly blocked by coinjection of excess NDP-α-MSH peptide (p < 0.05). In conclusion, the MC1R-specific imaging probe developed in this study displays excellent potential for the intraoperative detection of regional node involvement and for margin detection during melanoma metastasis resection.


Asunto(s)
Colorantes Fluorescentes/síntesis química , Melanoma Experimental/patología , Proteínas de Neoplasias/análisis , Peptidomiméticos/síntesis química , Receptor de Melanocortina Tipo 1/análisis , Neoplasias Cutáneas/patología , Animales , Endocitosis/efectos de los fármacos , Colorantes Fluorescentes/metabolismo , Expresión Génica , Humanos , Inyecciones Intravenosas , Ligandos , Masculino , Melanoma Experimental/diagnóstico , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones , Ratones Desnudos , Microscopía Fluorescente , Imagen Molecular/métodos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Peptidomiméticos/metabolismo , Receptor de Melanocortina Tipo 1/genética , Receptor de Melanocortina Tipo 1/metabolismo , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , alfa-MSH/análogos & derivados , alfa-MSH/farmacología
6.
J Med Chem ; 55(22): 9751-62, 2012 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-23098072

RESUMEN

Toll-like receptor 2 (TLR2) is a target for immune system stimulation during cancer immunotherapy and a cell-surface marker for pancreatic cancer. To develop targeted agents for cancer imaging and therapy, we designed, synthesized, and characterized 13 novel, fully synthetic high affinity TLR2 agonists. Analogue 10 had the highest agonist activity (NF-κB functional assay, EC(50) = 20 nM) and binding affinity (competitive binding assay, K(i) = 25 nM). As an immune adjuvant, compound 10 stimulated the immune system in vivo by generation and persistence of antigen-specific CD8+ T cells indicating its potential use in cancer immunotherapy. After conjugation of near-infrared dye to 10, agonist activity (EC(50) = 34 nM) and binding affinity (K(i) = 11 nM) were retained in 13. Fluorescence signal was present in TLR2 expressing pancreatic tumor xenografts 24 h after injection of 13, while an excess of unlabeled ligand blocked 13 from binding to the tumor, resulting in significantly decreased signal (p < 0.001) demonstrating in vivo selectivity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Diagnóstico por Imagen , Colorantes Fluorescentes , Inmunoterapia , Neoplasias Pancreáticas/tratamiento farmacológico , Receptor Toll-Like 2/agonistas , Animales , Células Cultivadas , Femenino , Humanos , Riñón/citología , Riñón/efectos de los fármacos , Riñón/metabolismo , Ligandos , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , FN-kappa B/genética , FN-kappa B/metabolismo , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/inmunología , Transducción de Señal , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA