Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Future Oncol ; 19(8): 559-573, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37070653

RESUMEN

ESR1 mutation (ESR1m) is a frequent cause of acquired resistance to aromatase inhibitor (AI) plus cyclin-dependent kinase 4 and 6 inhibitors (CDK4/6i), which is a first-line therapy for hormone-receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) advanced breast cancer (ABC). Camizestrant is a next-generation oral selective estrogen receptor degrader (SERD) that in a phase II study significantly improved progression-free survival (PFS) over fulvestrant (also a SERD) in ER+/HER2- ABC. SERENA-6 (NCT04964934) is a randomized, double-blind, phase III study evaluating the efficacy and safety of switching from an AI to camizestrant, while maintaining the same CDK4/6i, upon detection of ESR1m in circulating tumor DNA before clinical disease progression on first-line therapy for HR+/HER2- ABC. The aim is to treat ESR1m clones and extend the duration of control of ER-driven tumor growth, delaying the need for chemotherapy. The primary end point is PFS; secondary end points include chemotherapy-free survival, time to second progression event (PFS2), overall survival, patient-reported outcomes and safety.


Why will we perform this study? Patients with advanced breast cancer in which the cancer cells have the receptor for the hormone estrogen and/or progesterone are typically treated with an aromatase inhibitor, a hormone therapy that decreases estrogen being made in the body, together with an inhibitor of cyclin-dependent kinases 4 and 6 (CDK4/6), a drug that blocks the growth of cancer cells. Although cancers usually respond to treatment initially, the cancer cells eventually change, so the drug combination no longer works. For example, mutation of the estrogen receptor (referred to as ESR1m) can stop aromatase inhibitors from working. Camizestrant is an investigational drug that blocks estrogen receptors, including mutated receptors, reducing the growth and spread of cancer. Here we describe the SERENA-6 clinical trial, which is testing camizestrant as a treatment for patients with breast cancer with ESR1m. How will we perform this research? The phase III SERENA-6 trial will use blood tests to monitor if patients with breast cancer develop ESR1m while being treated with an aromatase inhibitor and a CDK4/6 inhibitor. If ESR1m is detected, yet the disease is stable, participants will be randomly assigned to either continue with the same aromatase inhibitor or switch to camizestrant while continuing with the same CDK4/6 inhibitor. The study will assess whether switching to camizestrant prolongs the time before the cancer grows, spreads or worsens. It will also assess the length of time that participants live for versus those who continue with an aromatase inhibitor. Clinical Trial Registration: NCT04964934 (ClinicalTrials.gov).


Asunto(s)
Neoplasias de la Mama , Femenino , Humanos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Inhibidores de la Aromatasa/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Ensayos Clínicos Fase III como Asunto , Fulvestrant/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo
2.
J Clin Pharmacol ; 61(3): 394-405, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32989795

RESUMEN

Doravirine, a novel nonnucleoside reverse transcriptase inhibitor for the treatment of human immunodeficiency virus 1 (HIV-1), is predominantly cleared by cytochrome P450 (CYP) 3A4 and metabolized to an oxidative metabolite (M9). Coadministration with rifabutin, a moderate CYP3A4 inducer, decreased doravirine exposure. Based on nonparametric superposition modeling, a doravirine dose adjustment from 100 mg once daily to 100 mg twice daily during rifabutin coadministration was proposed. However, M9 exposure may also be impacted by induction, in addition to the dose adjustment. As M9 concentrations have not been quantified in previous clinical studies, a physiologically based pharmacokinetic model was developed to investigate the change in M9 exposure when doravirine is coadministered with CYP3A inducers. Simulations demonstrated that although CYP3A induction increases doravirine clearance by up to 4.4-fold, M9 exposure is increased by only 1.2-fold relative to exposures for doravirine 100 mg once daily in the absence of CYP3A induction. Thus, a 2.4-fold increase in M9 exposure relative to the clinical dose of doravirine is anticipated when doravirine 100 mg twice daily is coadministered with rifabutin. In a subsequent clinical trial, doravirine and M9 exposures, when doravirine 100 mg twice daily was coadministered with rifabutin, were found to be consistent with model predictions using rifampin and efavirenz as representative inducers. These findings support the dose adjustment to doravirine 100 mg twice daily when coadministered with rifabutin.


Asunto(s)
Modelos Biológicos , Piridonas/farmacocinética , Inhibidores de la Transcriptasa Inversa/farmacocinética , Rifabutina/farmacología , Triazoles/farmacocinética , Adulto , Alquinos/farmacología , Benzoxazinas/farmacología , Simulación por Computador , Ciclopropanos/farmacología , Citocromo P-450 CYP3A/efectos de los fármacos , Citocromo P-450 CYP3A/metabolismo , Inductores del Citocromo P-450 CYP3A/farmacología , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Femenino , Humanos , Masculino , Persona de Mediana Edad , Piridonas/administración & dosificación , Inhibidores de la Transcriptasa Inversa/administración & dosificación , Rifabutina/administración & dosificación , Rifampin/farmacología , Triazoles/administración & dosificación , Adulto Joven
3.
J Virol ; 83(14): 7049-61, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19403671

RESUMEN

Proteasomes are the major source of proteases responsible for the generation of peptides bound to major histocompatibility complex class I molecules. Antigens, adjuvants, and cytokines can modulate the composition and enzymatic activity of proteasomes and thus alter the epitopes generated. In the present study, we examined the effect of human immunodeficiency virus type 1 (HIV-1) p24 on proteasomes from a dendritic cell line (JAWS II), from a macrophage cell line (C2.3), and from murine primary bone marrow-derived macrophages and dendritic cells. HIV-1 p24 downregulated PA28beta and the beta2i subunit of the immunoproteasome complex in JAWS II cells but did not decrease the immunoproteasome subunits in macrophages, whereas in primary dendritic cells, PA28alpha, beta2i, and beta5i were downregulated. Exposure of JAWS II cells and primary dendritic cells to HIV-1 p24 for 90 min significantly decreased the presentation of ovalbumin to a SIINFEKL-specific CD8(+) T-cell hybridoma. The decrease in antigen presentation and the downmodulation of the immunoproteasome subunits in JAWS II cells and primary dendritic cells could be overcome by pretreating the cells with gamma interferon for 6 h or by exposing the cells to HIV-1 p24 encapsulated in liposomes containing lipid A. These results suggest that early antigen processing kinetics could influence the immunogenicity of CD8(+) T-cell epitopes generated.


Asunto(s)
Presentación de Antígeno , Proteína p24 del Núcleo del VIH/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Complejo de la Endopetidasa Proteasomal/inmunología , Animales , Línea Celular , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/virología , Regulación hacia Abajo , Femenino , Infecciones por VIH/virología , Humanos , Macrófagos/inmunología , Macrófagos/virología , Ratones , Ratones Endogámicos C57BL
4.
Vaccine ; 27(49): 6939-49, 2009 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-19748578

RESUMEN

Liposomal lipid A is an effective adjuvant for the delivery of antigens and for the induction of both cellular and humoral immunity. In this study, we demonstrate that following the third immunization with HIV-1 Gag p24 encapsulated in liposomes containing lipid A [L(p24+LA)], central memory CD8+ T-cells were localized in the spleen and lymph nodes of mice while effector memory CD8+ T-cells and effector CD4+ T-cells were found in the PBMC. Effector CD4+ T-cells were also detected in the spleen and lymph nodes. The predominant cytokine secreted from splenic lymphocytes and lymph nodes was IFN-gamma. In contrast, IL-6 and IL-10 were the major cytokines produced from PBMC. The peptide stimulation indicated that the cytokine responses observed were T-cell specific. The results demonstrate the importance of the adjuvant liposomal lipid A for the induction of HIV-1 Gag p24 -specific CD8+ T-cells, effector CD4+ T-cells, and cytokines with a Th-1 type profile after immunization with L(p24+LA).


Asunto(s)
Vacunas contra el SIDA/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteína p24 del Núcleo del VIH/inmunología , Lípido A/inmunología , Liposomas/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Femenino , VIH-1/inmunología , Memoria Inmunológica/inmunología , Interferón gamma , Interleucina-10/inmunología , Interleucina-6/inmunología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Bazo/inmunología , Subgrupos de Linfocitos T/inmunología
5.
Virus Res ; 145(1): 31-8, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19540887

RESUMEN

Eighteen dengue viruses (DENVs) representing all four serotypes, isolated from pediatric patients at children's hospital, Queen Sirikit National Institute of Child Health, Bangkok, Thailand exhibiting a diverse spectrum of disease ranging from uncomplicated dengue fever (DF) to severe dengue hemorrhagic fever (DHF), were tested for their ability to attach to host cells, replicate and interfere with the IFNalpha signaling pathway by interfering with signal transducer and activator of transcription 1 (STAT-1) function. Although most isolates suppressed IFNalpha-induced STAT-1 phosphorylation, our results showed no difference between DENV strains associated with DF and those associated with DHF. However, the DHF isolates tended replicate to higher titers in dendritic cells (DCs) than the DF isolates, but this ability was independent of their cell-binding capability. Our results suggest that the emergence early in infection of viruses with a high degree of replication fitness may play an important role in DENV pathogenesis.


Asunto(s)
Virus del Dengue , Interferón-alfa/metabolismo , Dengue Grave/virología , Acoplamiento Viral , Replicación Viral , Adolescente , Línea Celular , Niño , Preescolar , Virus del Dengue/aislamiento & purificación , Virus del Dengue/fisiología , Femenino , Interacciones Huésped-Patógeno , Humanos , Lactante , Masculino , Factor de Transcripción STAT1/metabolismo , Dengue Grave/metabolismo , Transducción de Señal , Tailandia
6.
Infect Immun ; 75(3): 1359-63, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17220311

RESUMEN

The Escherichia coli maltose-binding protein (MBP) is used to increase the stability and solubility of proteins in bacterial protein expression systems and is increasingly being used to facilitate the production and delivery of subunit vaccines against various pathogenic bacteria and viruses. The MBP tag is presumed inert, with minimum effects on the bioactivity of the tagged protein or its biodistribution. However, few studies have characterized the immunological attributes of MBP. Here, we analyze the phenotypic and functional outcomes of MBP-treated dendritic cells (DCs) and show that MBP induces DC activation and production of proinflammatory cytokines (interleukin-1beta [IL-1beta], IL-6, IL-8, tumor necrosis factor alpha, and IL-12p70) within 24 h and strongly increases Ikappabeta phosphorylation in treated cells. Interestingly, phosphorylation of Ikappabeta was largely abrogated by the addition of anti-human Toll-like receptor 4 (TLR4) antibodies, indicating that MBP activates signaling for DC maturation via TLR4. Consistent with this hypothesis, MBP activated the TLR4-expressing cell line 293-hTLR4A but not control cultures to secrete IL-8. The observed data were independent of lipopolysaccharide contamination and support a role for TLR4 in mediating the effects of MBP. These results provide insight into a mechanism by which MBP might enhance immune responses to vaccine fusion proteins.


Asunto(s)
Proteínas Portadoras/fisiología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Escherichia coli/inmunología , Receptor Toll-Like 4/fisiología , Adyuvantes Inmunológicos/metabolismo , Adyuvantes Inmunológicos/fisiología , Proteínas Portadoras/metabolismo , Línea Celular , Células Cultivadas , Células Clonales , Citocinas/metabolismo , Escherichia coli/fisiología , Humanos , Proteínas de Unión a Maltosa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA