Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
1.
Mol Ther ; 30(9): 3078-3094, 2022 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-35821637

RESUMEN

mRNA vaccines have recently proved to be highly effective against SARS-CoV-2. Key to their success is the lipid-based nanoparticle (LNP), which enables efficient mRNA expression and endows the vaccine with adjuvant properties that drive potent antibody responses. Effective cancer vaccines require long-lived, qualitative CD8 T cell responses instead of antibody responses. Systemic vaccination appears to be the most effective route, but necessitates adaptation of LNP composition to deliver mRNA to antigen-presenting cells. Using a design-of-experiments methodology, we tailored mRNA-LNP compositions to achieve high-magnitude tumor-specific CD8 T cell responses within a single round of optimization. Optimized LNP compositions resulted in enhanced mRNA uptake by multiple splenic immune cell populations. Type I interferon and phagocytes were found to be essential for the T cell response. Surprisingly, we also discovered a yet unidentified role of B cells in stimulating the vaccine-elicited CD8 T cell response. Optimized LNPs displayed a similar, spleen-centered biodistribution profile in non-human primates and did not trigger histopathological changes in liver and spleen, warranting their further assessment in clinical studies. Taken together, our study clarifies the relationship between nanoparticle composition and their T cell stimulatory capacity and provides novel insights into the underlying mechanisms of effective mRNA-LNP-based antitumor immunotherapy.


Asunto(s)
COVID-19 , Vacunas contra el Cáncer , Nanopartículas , Animales , Inmunización/métodos , Inmunoterapia , ARN Mensajero/metabolismo , SARS-CoV-2/genética , Bazo , Distribución Tisular , Vacunación/métodos
2.
Hum Mutat ; 36(11): 1039-42, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26224408

RESUMEN

Congenital secondary erythrocytosis is a rare disorder characterized by increased red blood cell production. An important cause involves defects in the oxygen sensing pathway, in particular the PHD2-VHL-HIF axis. Mutations in VHL are also associated with the von Hippel-Lindau tumor predisposition syndrome. The differences in phenotypic expression of VHL mutations are poorly understood. We report on three patients with erythrocytosis, from two unrelated families. All patients show exceptionally high erythropoietin (EPO) levels, and are homozygous for a novel missense mutation in VHL: c.162G>C p.(Met54Ile). The c.162G>C mutation is the most upstream homozygous VHL mutation described so far in patients with erythrocytosis. It abolishes the internal translational start codon, which directs expression of VHLp19, resulting in the production of only VHLp30. The exceptionally high EPO levels and the absence of VHL-associated tumors in the patients suggest that VHLp19 has a role for regulating EPO levels that VHLp30 does not have, whereas VHLp30 is really the tumor suppressor isoform.


Asunto(s)
Codón Iniciador , Homocigoto , Mutación , Iniciación de la Cadena Peptídica Traduccional/genética , Policitemia/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Adolescente , Sustitución de Aminoácidos , Preescolar , Índices de Eritrocitos , Eritropoyetina/sangre , Femenino , Orden Génico , Sitios Genéticos , Humanos , Masculino , Policitemia/sangre , Policitemia/diagnóstico , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Adulto Joven
3.
Nanoscale ; 10(5): 2413-2426, 2018 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-29334397

RESUMEN

Extracellular vesicles (EVs) are increasingly being recognized as candidate drug delivery systems due to their ability to functionally transfer biological cargo between cells. However, manipulation of targeting properties of EVs through engineering of the producer cells can be challenging and time-consuming. As a novel approach to confer tumor targeting properties to isolated EVs, we generated recombinant fusion proteins of nanobodies against the epidermal growth factor receptor (EGFR) fused to phosphatidylserine (PS)-binding domains of lactadherin (C1C2). C1C2-nanobody fusion proteins were expressed in HEK293 cells and isolated from culture medium with near-complete purity as determined by SDS-PAGE. Fusion proteins specifically bound PS and showed no affinity for other common EV membrane lipids. Furthermore, C1C2 fused to anti-EGFR nanobodies (EGa1-C1C2) bound EGFR with high affinity and competed with binding of its natural ligand EGF, as opposed to C1C2 fused to non-targeting control nanobodies (R2-C1C2). Both proteins readily self-associated onto membranes of EVs derived from erythrocytes and Neuro2A cells without affecting EV size and integrity. EV-bound R2-C1C2 did not influence EV-cell interactions, whereas EV-bound EGa1-C1C2 dose-dependently enhanced specific binding and uptake of EVs by EGFR-overexpressing tumor cells. In conclusion, we developed a novel strategy to efficiently and universally confer tumor targeting properties to PS-exposing EVs after their isolation, without affecting EV characteristics, circumventing the need to modify EV-secreting cells. This strategy may also be employed to decorate EVs with other moieties, including imaging probes or therapeutic proteins.


Asunto(s)
Sistemas de Liberación de Medicamentos , Vesículas Extracelulares , Fosfatidilserinas/química , Anticuerpos de Dominio Único/química , Antígenos de Superficie/metabolismo , Receptores ErbB/metabolismo , Células HEK293 , Humanos , Proteínas de la Leche/metabolismo , Proteínas Recombinantes de Fusión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA