Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Small ; 20(11): e2304378, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38072809

RESUMEN

With six therapies approved by the Food and Drug Association, chimeric antigen receptor (CAR) T cells have reshaped cancer immunotherapy. However, these therapies rely on ex vivo viral transduction to induce permanent CAR expression in T cells, which contributes to high production costs and long-term side effects. Thus, this work aims to develop an in vivo CAR T cell engineering platform to streamline production while using mRNA to induce transient, tunable CAR expression. Specifically, an ionizable lipid nanoparticle (LNP) is utilized as these platforms have demonstrated clinical success in nucleic acid delivery. Though LNPs often accumulate in the liver, the LNP platform used here achieves extrahepatic transfection with enhanced delivery to the spleen, and it is further modified via antibody conjugation (Ab-LNPs) to target pan-T cell markers. The in vivo evaluation of these Ab-LNPs confirms that targeting is necessary for potent T cell transfection. When using these Ab-LNPs for the delivery of CAR mRNA, antibody and dose-dependent CAR expression and cytokine release are observed along with B cell depletion of up to 90%. In all, this work conjugates antibodies to LNPs with extrahepatic tropism, evaluates pan-T cell markers, and develops Ab-LNPs capable of generating functional CAR T cells in vivo.


Asunto(s)
Nanopartículas , Receptores Quiméricos de Antígenos , Receptores Quiméricos de Antígenos/genética , Liposomas , Transfección , Anticuerpos , Ingeniería Celular , ARN Interferente Pequeño
2.
Nat Mater ; 22(12): 1571-1580, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37696939

RESUMEN

Chimeric antigen receptor T (CAR T) cell immunotherapy is successful at treating many cancers. However, it often induces life-threatening cytokine release syndrome (CRS) and neurotoxicity. Here, we show that in situ conjugation of polyethylene glycol (PEG) to the surface of CAR T cells ('PEGylation') creates a polymeric spacer that blocks cell-to-cell interactions between CAR T cells, tumour cells and monocytes. Such blockage hinders intensive tumour lysing and monocyte activation by CAR T cells and, consequently, decreases the secretion of toxic cytokines and alleviates CRS-related symptoms. Over time, the slow expansion of CAR T cells decreases PEG surface density and restores CAR T cell-tumour-cell interactions to induce potent tumour killing. This occurs before the restoration of CAR T cell-monocyte interactions, opening a therapeutic window for tumour killing by CAR T cells before monocyte overactivation. Lethal neurotoxicity is also lower when compared with treatment with the therapeutic antibody tocilizumab, demonstrating that in situ PEGylation of CAR T cells provides a materials-based strategy for safer cellular immunotherapy.


Asunto(s)
Neoplasias , Síndromes de Neurotoxicidad , Receptores Quiméricos de Antígenos , Humanos , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Síndrome de Liberación de Citoquinas/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Receptores Quiméricos de Antígenos/uso terapéutico , Inmunoterapia Adoptiva , Síndromes de Neurotoxicidad/tratamiento farmacológico , Síndromes de Neurotoxicidad/metabolismo , Linfocitos T
3.
ACS Nano ; 18(25): 16151-16165, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38861479

RESUMEN

Immune modulation through the intracellular delivery of nucleoside-modified mRNA to immune cells is an attractive approach for in vivo immunoengineering, with applications in infectious disease, cancer immunotherapy, and beyond. Lipid nanoparticles (LNPs) have come to the fore as a promising nucleic acid delivery platform, but LNP design criteria remain poorly defined, making the rate-limiting step for LNP discovery the screening process. In this study, we employed high-throughput in vivo LNP screening based on molecular barcoding to investigate the influence of LNP composition on immune tropism with applications in vaccines and systemic immunotherapies. Screening a large LNP library under both intramuscular (i.m.) and intravenous (i.v.) injection, we observed differential influences on LNP uptake by immune populations across the two administration routes, gleaning insight into LNP design criteria for in vivo immunoengineering. In validation studies, the lead LNP formulation for i.m. administration demonstrated substantial mRNA translation in the spleen and draining lymph nodes with a more favorable biodistribution profile than LNPs formulated with the clinical standard ionizable lipid DLin-MC3-DMA (MC3). The lead LNP formulations for i.v. administration displayed potent immune transfection in the spleen and peripheral blood, with one lead LNP demonstrating substantial transfection of splenic dendritic cells and another inducing substantial transfection of circulating monocytes. Altogether, the immunotropic LNPs identified by high-throughput in vivo screening demonstrated significant promise for both locally- and systemically-delivered mRNA and confirmed the value of the LNP design criteria gleaned from our screening process, which could potentially inform future endeavors in mRNA vaccine and immunotherapy applications.


Asunto(s)
Lípidos , Ratones Endogámicos C57BL , Nanopartículas , ARN Mensajero , Animales , Nanopartículas/química , ARN Mensajero/genética , Ratones , Lípidos/química , Ensayos Analíticos de Alto Rendimiento , Femenino , Inyecciones Intramusculares , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Inyecciones Intravenosas , Inmunoterapia , Liposomas
4.
Adv Mater ; 36(26): e2313226, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38419362

RESUMEN

Chimeric antigen receptor (CAR) T cell therapy has achieved remarkable clinical success in the treatment of hematological malignancies. However, producing these bespoke cancer-killing cells is a complicated ex vivo process involving leukapheresis, artificial T cell activation, and CAR construct introduction. The activation step requires the engagement of CD3/TCR and CD28 and is vital for T cell transfection and differentiation. Though antigen-presenting cells (APCs) facilitate activation in vivo, ex vivo activation relies on antibodies against CD3 and CD28 conjugated to magnetic beads. While effective, this artificial activation adds to the complexity of CAR T cell production as the beads must be removed prior to clinical implementation. To overcome this challenge, this work develops activating lipid nanoparticles (aLNPs) that mimic APCs to combine the activation of magnetic beads and the transfection capabilities of LNPs. It is shown that aLNPs enable one-step activation and transfection of primary human T cells with the resulting mRNA CAR T cells reducing tumor burden in a murine xenograft model, validating aLNPs as a promising platform for the rapid production of mRNA CAR T cells.


Asunto(s)
Células Presentadoras de Antígenos , Inmunoterapia Adoptiva , Nanopartículas , ARN Mensajero , Receptores Quiméricos de Antígenos , Linfocitos T , Humanos , Nanopartículas/química , Animales , Ratones , Células Presentadoras de Antígenos/inmunología , Inmunoterapia Adoptiva/métodos , Linfocitos T/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Neoplasias/terapia , Neoplasias/inmunología , Inmunoterapia/métodos , Línea Celular Tumoral , Lípidos/química , Transfección/métodos , Liposomas
5.
Sci Adv ; 8(8): eabo2423, 2022 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-35196095

RESUMEN

Nanoparticles, mRNA, and ultraviolet light combine to reprogram specific immune cells directly in the body.

6.
ACS Sens ; 5(6): 1555-1566, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32337979

RESUMEN

Precise pH measurements in the immediate environment of receptors is essential for elucidating the mechanisms through which local pH changes associated with diseased phenotypes manifest into aberrant receptor function. However, current pH sensors lack the ability to localize and target specific receptor molecules required to make these measurements. Herein we present the Litmus-body, our recombinant protein-based pH sensor, which through fusion to an anti-IgG nanobody is capable of piggybacking on IgG antibodies for molecular targeting to specific proteins on the cell surface. By normalizing a pH-dependent green fluorescent protein to a long Stokes shift red fluorophore or fluorescent protein, we readily report pH independent of sensor concentration using a single 488 nm excitation. Our Litmus-body showed excellent responsiveness in solution, with a greater than 50-fold change across the regime of physiological pH. The sensor was further validated for use on live cells and shown to be specific to the protein of interest. In complex with our Litmus-body, cetuximab therapeutic antibody retained its functionality in binding and inhibiting ligand interaction of its target epidermal growth factor receptor (EGFR), triggering receptor-mediated endocytosis that allowed tracking of local pH from the cell surface through the endocytic pathway.


Asunto(s)
Endocitosis , Colorantes Fluorescentes , Cetuximab , Concentración de Iones de Hidrógeno , Ligandos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA