Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Eur J Clin Pharmacol ; 79(9): 1219-1228, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37436495

RESUMEN

PURPOSE: We evaluated the safety, pharmacokinetics (PK), pharmacodynamics (PD), and immunogenicity of intravenous (IV) inclacumab, a fully human IgG4 anti-P-selectin monoclonal antibody in development for the treatment of sickle cell disease, at doses up to and exceeding those previously tested in healthy individuals. METHODS: In this phase 1, open-label, single-ascending-dose study, 15 healthy participants were enrolled into cohorts receiving 20 mg/kg (n = 6) or 40 mg/kg (n = 9) IV inclacumab and observed for up to 29 weeks post-dose. Safety, PK parameters, thrombin receptor-activating peptide (TRAP)-activated platelet-leukocyte aggregate (PLA) formation, P-selectin inhibition, plasma soluble P-selectin, and anti-drug antibodies were characterized. RESULTS: Two inclacumab-related treatment-emergent adverse events were reported in 1 participant; no dose-limiting toxicities were observed. Plasma PK parameters were generally dose-proportional, with a terminal half-life of 13 to 17 days. Mean TRAP-activated PLA formation decreased within 3 h from the start of infusion, and inhibition was sustained for ~ 23 weeks. Mean P-selectin inhibition > 90% was observed up to 12 weeks post-dose. The mean ratio of free to total soluble P-selectin decreased rapidly from pre-dose to end of infusion, then increased gradually to 78% of the baseline ratio by week 29. Treatment-emergent anti-drug antibodies were observed in 2 of 15 participants (13%), without apparent impact on safety, PK, or PD. CONCLUSIONS: Inclacumab was well tolerated, with PK as expected for a monoclonal antibody against a membrane-bound target and a long duration of PD effects after both single IV doses, supporting a prolonged dosing interval. TRIAL REGISTRATION: ACTRN12620001156976; registered November 4, 2020.


Asunto(s)
Anemia de Células Falciformes , Anticuerpos Monoclonales , Humanos , Voluntarios Sanos , Anticuerpos Monoclonales/efectos adversos , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/inducido químicamente , Selectinas , Poliésteres , Método Doble Ciego , Relación Dosis-Respuesta a Droga
2.
AAPS J ; 23(5): 96, 2021 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-34324079

RESUMEN

Monoclonal antibodies that block the interaction between programmed cell death 1 (PD-1) and its ligand (PD-L1) have revolutionized cancer immunotherapy. However, immunogenic responses to these new therapies-such as the development of antidrug antibodies (ADAs) and neutralizing antibodies (NAbs)-may represent a significant challenge to both efficacy and safety in some patients. Dostarlimab (TSR-042) is an approved, humanized, anti-PD-1 monoclonal antibody that has shown efficacy in multiple solid tumor types. Here, we report the results of an immunogenicity analysis of dostarlimab monotherapy in patients enrolled in the GARNET trial, a multicenter, open-label, single-arm phase 1 study. Overall, 477 of 478 patients (99.8%) were included in the analysis of dostarlimab antibody prevalence, and 349 out of 478 enrolled patients (73.0%) were evaluable for treatment-emergent antibodies to dostarlimab. The incidence of treatment-emergent ADAs was 2.5% at the recommended therapeutic dose (500 mg Q3W for the first 4 doses, 1000 mg Q6W until discontinuation), which is comparable to other anti-PD-(L)1 drugs. NAbs were detected in only 1.3% of patients. In the small percentage of patients who developed ADAs, there was no evidence of altered efficacy or safety of dostarlimab at the recommended dosing regimen. These findings demonstrated that treatment with dostarlimab was associated with a low risk of eliciting clinically meaningful ADAs over the course of this study, and dostarlimab is already approved by health authorities.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Neutralizantes/sangre , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Inhibidores de Puntos de Control Inmunológico/inmunología , Neoplasias/tratamiento farmacológico , Adulto , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Neutralizantes/inmunología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/sangre , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/inmunología , Femenino , Estudios de Seguimiento , Humanos , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Incidencia , Masculino , Persona de Mediana Edad , Neoplasias/sangre , Neoplasias/inmunología , Criterios de Evaluación de Respuesta en Tumores Sólidos
3.
J Biomed Mater Res B Appl Biomater ; 100(1): 41-50, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22114061

RESUMEN

Magnesium-based bioabsorbable cardiovascular stents have been developed to overcome limitations of permanent metallic stents, such as late stent thrombosis. During stent degradation, endothelial and smooth muscle cells will be exposed to locally high magnesium concentrations with yet unknown physiological consequences. Here, we investigated the effects of elevated magnesium concentrations on human coronary artery endothelial and smooth muscle cell (HCAEC, HCASMC) growth and gene expression. In the course of 24 h after incubation with magnesium chloride solutions (1 or 10 mM) intracellular magnesium level in HCASMC raised from 0.55 ± 0.25 mM (1 mM) to 1.38 ± 0.95 mM (10 mM), while no increase was detected in HCAEC. Accordingly, a DNA microarray-based study identified 69 magnesium regulated transcripts in HCAEC, but 2172 magnesium regulated transcripts in HCASMC. Notably, a significant regulation of various growth factors and extracellular matrix components was observed. In contrast, viability and proliferation of HCAEC were increased at concentrations of up to 25 mM magnesium chloride, while in HCASMC viability and proliferation appeared to be unaffected. Taken together, our data indicate that magnesium halts smooth muscle cell proliferation and stimulates endothelial cell proliferation, which might translate into a beneficial effect in the setting of stent associated vascular injury.


Asunto(s)
Implantes Absorbibles , Proliferación Celular , Vasos Coronarios/metabolismo , Células Endoteliales/metabolismo , Magnesio , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Stents , Supervivencia Celular , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/lesiones , Células Endoteliales/citología , Regulación de la Expresión Génica , Humanos , Ensayo de Materiales/métodos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...