Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2789: 171-184, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38507003

RESUMEN

The assessment of antigen presentation by dendritic cells and subsequent antigen-dependent activation of T lymphocytes is a critical step underlying the efficacy of nanoparticle-based therapeutic vaccines. Since nanoparticle physicochemical properties determine their interactions with the immune system, the early stages of nanotechnology-based vaccine development commonly involve optimizing the particles' properties to create a formulation with desired stability, antigen release, targeting of desired cell populations, and efficacy. To accelerate this process, in vitro models suitable for the rapid assessment of a novel vaccine candidate's efficacy are highly desirable. One such model is described in this protocol. Herein, nanoparticles are formulated to deliver a model antigen, SIINFEKL (OVA257-264), the immunodominant class I peptide derived from ovalbumin. These nanoparticles are added to the culture of murine bone marrow-derived dendritic cells, which are subsequently co-incubated with CD8+ T cells from OT-I transgenic mice. The efficient antigen presentation by dendritic cells results in the antigen-dependent proliferation of CD8+ T cells, which is detected by flow cytometry.


Asunto(s)
Linfocitos T CD8-positivos , Nanopartículas , Ratones , Animales , Médula Ósea , Antígenos , Presentación de Antígeno , Ovalbúmina , Nanopartículas/química , Células Dendríticas , Ratones Endogámicos C57BL
2.
Methods Mol Biol ; 2789: 161-169, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38507002

RESUMEN

Nanoparticles are frequently considered in vaccine applications due to their ability to co-deliver multiple antigens and adjuvants to antigen-presenting cells. Some nanoparticles also have intrinsic adjuvant properties that further enhance their ability to stimulate immune cells. The delivery of tumor-specific antigens to antigen-presenting cells (APCs) with subsequent antigenic peptide presentation in the context of class I major histocompatibility complex (MHC-I) molecules represents an essential effort in developing nanotechnology-based cancer vaccines. Experimental models are, therefore, needed to gauge the efficiency of nanotechnology carriers in achieving peptide antigen delivery to APCs and presentation in the context of MHC-I. The assay described herein utilizes a model antigen ovalbumin and model APCs, murine bone marrow-derived dendritic cells. The 25-D1.16 antibody, specific to the ovalbumin (OVA) MHC-I peptide SIINFEKL, recognizes this peptide presented in the context of the murine H2-Kb class I MHC molecule, allowing the presentation of this antigen on APCs to be detected by flow cytometry after nanoparticle delivery.


Asunto(s)
Presentación de Antígeno , Nanopartículas , Ratones , Animales , Células Dendríticas , Ovalbúmina , Médula Ósea , Antígenos de Histocompatibilidad Clase I , Nanopartículas/química , Péptidos , Antígenos de Neoplasias , Ratones Endogámicos C57BL
3.
Small ; 18(46): e2204941, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36216772

RESUMEN

Nucleic acid nanoparticles, or NANPs, rationally designed to communicate with the human immune system, can offer innovative therapeutic strategies to overcome the limitations of traditional nucleic acid therapies. Each set of NANPs is unique in their architectural parameters and physicochemical properties, which together with the type of delivery vehicles determine the kind and the magnitude of their immune response. Currently, there are no predictive tools that would reliably guide the design of NANPs to the desired immunological outcome, a step crucial for the success of personalized therapies. Through a systematic approach investigating physicochemical and immunological profiles of a comprehensive panel of various NANPs, the research team developes and experimentally validates a computational model based on the transformer architecture able to predict the immune activities of NANPs. It is anticipated that the freely accessible computational tool that is called an "artificial immune cell," or AI-cell, will aid in addressing the current critical public health challenges related to safety criteria of nucleic acid therapies in a timely manner and promote the development of novel biomedical tools.


Asunto(s)
Nanopartículas , Ácidos Nucleicos , Humanos , Ácidos Nucleicos/química , Monocitos , Nanopartículas/química , Interferones , Inteligencia Artificial
4.
Cancer Cell Int ; 22(1): 286, 2022 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-36123697

RESUMEN

BACKGROUND: Intratumoral (IT) delivery of toll-like receptor (TLR) agonists has shown encouraging anti-tumor benefit in preclinical and early clinical studies. However, IT delivery of TLR agonists may lead to rapid effusion from the tumor microenvironment (TME), potentially limiting the duration of local inflammation and increasing the risk of systemic adverse events. METHODS: To address these limitations, TransCon™ TLR7/8 Agonist-an investigational sustained-release prodrug of resiquimod that uses a TransCon linker and hydrogel technology to achieve sustained and predictable IT release of resiquimod-was developed. TransCon TLR7/8 Agonist was characterized for resiquimod release in vitro and in vivo, in mice and rats, and was assessed for anti-tumor efficacy and pharmacodynamic activity in mice. RESULTS: Following a single IT dose, TransCon TLR7/8 Agonist mediated potent tumor growth inhibition which was associated with sustained resiquimod release over several weeks with minimal induction of systemic cytokines. TransCon TLR7/8 Agonist monotherapy promoted activation of antigen-presenting cells in the TME and tumor-draining lymph nodes, with evidence of activation and expansion of CD8+ T cells in the tumor-draining lymph node and TME. Combination of TransCon TLR7/8 Agonist with systemic immunotherapy further promoted anti-tumor activity in TransCon TLR7/8 Agonist-treated tumors. In a bilateral tumor setting, combination of TransCon TLR7/8 Agonist with systemic IL-2 potentiated tumor growth inhibition in both injected and non-injected tumors and conferred protection against tumor rechallenge following complete regressions. CONCLUSIONS: Our findings show that a single dose of TransCon TLR7/8 Agonist can mediate sustained local release of resiquimod in the TME and promote potent anti-tumor effects as monotherapy and in combination with systemic immunotherapy, supporting TransCon TLR7/8 Agonist as a novel intratumoral TLR agonist for cancer therapy. A clinical trial to evaluate the safety and efficacy of TransCon TLR7/8 Agonist, as monotherapy and in combination with pembrolizumab, in cancer patients is currently ongoing (transcendIT-101; NCT04799054).

5.
J Immunother Cancer ; 10(7)2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35817480

RESUMEN

BACKGROUND: Recombinant interleukin-2 (IL-2, aldesleukin) is an approved cancer immunotherapy but causes severe toxicities including cytokine storm and vascular leak syndrome (VLS). IL-2 promotes antitumor function of IL-2Rß/γ+ natural killer (NK) cells and CD8+, CD4+ and gamma delta (γδ) T cells. However, IL-2 also potently activates immunosuppressive IL-2Rα+ regulatory T cells (Tregs) and IL-2Rα+ eosinophils and endothelial cells, which may promote VLS. Aldesleukin is rapidly cleared requiring frequent dosing, resulting in high Cmax likely potentiating toxicity. Thus, IL-2 cancer immunotherapy has two critical drawbacks: potent activation of undesired IL-2Rα+ cells and suboptimal pharmacokinetics with high Cmax and short half-life. METHODS: TransCon IL-2 ß/γ was designed to optimally address these drawbacks. To abolish IL-2Rα binding yet retain strong IL-2Rß/γ activity, IL-2 ß/γ was created by permanently attaching a small methoxy polyethylene glycol (mPEG) moiety in the IL-2Rα binding site. To improve pharmacokinetics, IL-2 ß/γ was transiently attached to a 40 kDa mPEG carrier via a TransCon (transient conjugation) linker creating a prodrug, TransCon IL-2 ß/γ, with sustained release of IL-2 ß/γ. IL-2 ß/γ was characterized in binding and primary cell assays while TransCon IL-2 ß/γ was studied in tumor-bearing mice and cynomolgus monkeys. RESULTS: IL-2 ß/γ demonstrated selective and potent human IL-2Rß/γ binding and activation without IL-2Rα interactions. TransCon IL-2 ß/γ showed slow-release pharmacokinetics with a low Cmax and a long (>30 hours) effective half-life for IL-2 ß/γ in monkeys. In mouse tumor models, TransCon IL-2 ß/γ promoted CD8+ T cell and NK cell activation and antitumor activity. In monkeys, TransCon IL-2 ß/γ induced robust activation and expansion of CD8+ T cells, NK cells and γδ T cells, relative to CD4+ T cells, Tregs and eosinophils, with no evidence of cytokine storm or VLS. Similarly, IL-2 ß/γ enhanced proliferation and cytotoxicity of primary human CD8+ T cells, NK cells and γδ T cells. SUMMARY: TransCon IL-2 ß/γ is a novel long-acting prodrug with sustained release of an IL-2Rß/γ-selective IL-2. It has remarkable and durable pharmacodynamic effects in monkeys and potential for improved clinical efficacy and tolerability compared with aldesleukin. TransCon IL-2 ß/γ is currently being evaluated in a Phase 1/2 clinical trial (NCT05081609).


Asunto(s)
Neoplasias , Profármacos , Animales , Linfocitos T CD8-positivos , Síndrome de Liberación de Citoquinas , Preparaciones de Acción Retardada/farmacología , Células Endoteliales , Humanos , Interleucina-2/farmacología , Subunidad alfa del Receptor de Interleucina-2 , Ratones , Neoplasias/tratamiento farmacológico , Profármacos/farmacología
6.
Cancer Immunol Immunother ; 70(9): 2701-2719, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34244816

RESUMEN

Recombinant agonists that activate co-stimulatory and cytokine receptors have shown limited clinical anticancer utility, potentially due to narrow therapeutic windows, the need for coordinated activation of co-stimulatory and cytokine pathways and the failure of agonistic antibodies to recapitulate signaling by endogenous ligands. RTX-240 is a genetically engineered red blood cell expressing 4-1BBL and IL-15/IL-15Rα fusion (IL-15TP). RTX-240 is designed to potently and simultaneously stimulate the 4-1BB and IL-15 pathways, thereby activating and expanding T cells and NK cells, while potentially offering an improved safety profile through restricted biodistribution. We assessed the ability of RTX-240 to expand and activate T cells and NK cells and evaluated the in vivo efficacy, pharmacodynamics and tolerability using murine models. Treatment of PBMCs with RTX-240 induced T cell and NK cell activation and proliferation. In vivo studies using mRBC-240, a mouse surrogate for RTX-240, revealed biodistribution predominantly to the red pulp of the spleen, leading to CD8 + T cell and NK cell expansion. mRBC-240 was efficacious in a B16-F10 melanoma model and led to increased NK cell infiltration into the lungs. mRBC-240 significantly inhibited CT26 tumor growth, in association with an increase in tumor-infiltrating proliferating and cytotoxic CD8 + T cells. mRBC-240 was tolerated and showed no evidence of hepatic injury at the highest feasible dose, compared with a 4-1BB agonistic antibody. RTX-240 promotes T cell and NK cell activity in preclinical models and shows efficacy and an improved safety profile. Based on these data, RTX-240 is now being evaluated in a clinical trial.


Asunto(s)
Ligando 4-1BB/genética , Tratamiento Basado en Trasplante de Células y Tejidos , Eritrocitos/metabolismo , Expresión Génica , Terapia Genética , Interleucina-15/genética , Ligando 4-1BB/metabolismo , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Precursoras Eritroides/metabolismo , Femenino , Citometría de Flujo , Genes Reporteros , Ingeniería Genética , Terapia Genética/métodos , Humanos , Interleucina-15/metabolismo , Ratones , Modelos Animales , Unión Proteica , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Molecules ; 24(6)2019 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-30897721

RESUMEN

Infusion reactions (IRs) create a translational hurdle for many novel therapeutics, including those utilizing nanotechnology. Nucleic acid nanoparticles (NANPs) are a novel class of therapeutics prepared by rational design of relatively short oligonucleotides to self-assemble into various programmable geometric shapes. While cytokine storm, a common type of IR, has halted clinical development of several therapeutic oligonucleotides, NANP technologies hold tremendous potential to bring these reactions under control by tuning the particle's physicochemical properties to the desired type and magnitude of the immune response. Recently, we reported the very first comprehensive study of the structure⁻activity relationship between NANPs' shape, size, composition, and their immunorecognition in human cells, and identified the phagolysosomal pathway as the major route for the NANPs' uptake and subsequent immunostimulation. Here, we explore the molecular mechanism of NANPs' recognition by primary immune cells, and particularly the contributing role of the Toll-like receptors. Our current study expands the understanding of the immune recognition of engineered nucleic acid-based therapeutics and contributes to the improvement of the nanomedicine safety profile.


Asunto(s)
Leucocitos Mononucleares/metabolismo , Nanopartículas/química , Ácidos Nucleicos/química , Células Cultivadas , Electroporación , Humanos , Nanotecnología/métodos , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 9/metabolismo
9.
Adv Drug Deliv Rev ; 141: 3-22, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29339144

RESUMEN

Cancer is a complex systemic disorder that affects many organs and tissues and arises from the altered function of multiple cellular and molecular mechanisms. One of the systems malfunctioning in cancer is the immune system. Restoring and improving the ability of the immune system to effectively recognize and eradicate cancer is the main focus of immunotherapy, a topic which has garnered recent and significant interest. The initial excitement about immunotherapy, however, has been challenged by its limited efficacy in certain patient populations and the development of adverse effects such as therapeutic resistance and autoimmunity. At the same time, a number of advances in the field of nanotechnology have sought to address the challenges faced by modern immunotherapeutics and allow these therapeutic strategies to realize their full potential. This endeavour requires an understanding of not only the immunological barriers in cancer but also the mechanisms by which modern technologies and immunotherapeutics modulate the function of the immune system. Herein, we summarize the major barriers relevant to cancer immunotherapy and review current progress in addressing these obstacles using various approaches and clinically approved therapies. We then discuss the remaining challenges and how they can be addressed by nanotechnology. We lay out translational considerations relevant to the therapies described and propose a framework for the development of next-generation nanotechnology-enabled immunotherapies.


Asunto(s)
Inmunoterapia , Nanopartículas/administración & dosificación , Neoplasias/terapia , Animales , Humanos , Nanotecnología , Neoplasias/inmunología
10.
Nucleic Acids Res ; 47(3): 1350-1361, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30517685

RESUMEN

Nucleic acid-based assemblies that interact with each other and further communicate with the cellular machinery in a controlled manner represent a new class of reconfigurable materials that can overcome limitations of traditional biochemical approaches and improve the potential therapeutic utility of nucleic acids. This notion enables the development of novel biocompatible 'smart' devices and biosensors with precisely controlled physicochemical and biological properties. We extend this novel concept by designing RNA-DNA fibers and polygons that are able to cooperate in different human cell lines and that have defined immunostimulatory properties confirmed by ex vivo experiments. The mutual intracellular interaction of constructs results in the release of a large number of different siRNAs while giving a fluorescent response and activating NF-κB decoy DNA oligonucleotides. This work expands the possibilities of nucleic acid technologies by (i) introducing very simple design principles and assembly protocols; (ii) potentially allowing for a simultaneous release of various siRNAs together with functional DNA sequences and (iii) providing controlled rates of reassociation, stabilities in human blood serum, and immunorecognition.


Asunto(s)
ADN/genética , FN-kappa B/genética , ARN/genética , Transcripción Genética , ADN/química , Transferencia Resonante de Energía de Fluorescencia , Regulación de la Expresión Génica/genética , Humanos , Oligodesoxirribonucleótidos/genética , Oligonucleótidos/química , Oligonucleótidos/genética , ARN/química , ARN Interferente Pequeño/genética
11.
Nano Lett ; 18(7): 4309-4321, 2018 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-29894623

RESUMEN

Nucleic acid nanoparticles (NANPs) have evolved as a new class of therapeutics with the potential to detect and treat diseases. Despite tremendous advancements in NANP development, their immunotoxicity, one of the major impediments in clinical translation of traditional therapeutic nucleic acids (TNAs), has never been fully characterized. Here, we describe the first systematically studied immunological recognition of 25 representative RNA and DNA NANPs selected to have different design principles and physicochemical properties. We discover that, unlike traditional TNAs, NANPs used without a delivery carrier are immunoquiescent. We show that interferons (IFNs) are the key cytokines triggered by NANPs after their internalization by phagocytic cells, which agrees with predictions based on the experiences with TNAs. However, in addition to type I IFNs, type III IFNs also serve as reliable biomarkers of NANPs, which is usually not characteristic of TNAs. We show that overall immunostimulation relies on NANP shapes, connectivities, and compositions. We demonstrate that, like with traditional TNAs, plasmacytoid dendritic cells serve as the primary interferon producers among all peripheral blood mononuclear cells treated with NANPs, and scavenger receptor-mediated uptake and endosomal Toll-like receptor signaling are essential for NANP immunorecognition. The TLR involvement, however, is different from that expected for traditional TNA recognition. Based on these results, we suggest that NANP technology may serve as a prototype of auxiliary molecular language for communication with the immune system and the modulation of immune responses.


Asunto(s)
Inmunidad Innata/efectos de los fármacos , Interferones/antagonistas & inhibidores , Nanopartículas/uso terapéutico , Ácidos Nucleicos/uso terapéutico , ADN/efectos adversos , ADN/inmunología , ADN/uso terapéutico , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Interferones/genética , Interferones/inmunología , Nanopartículas/efectos adversos , Nanopartículas/ultraestructura , Ácidos Nucleicos/efectos adversos , Ácidos Nucleicos/inmunología , Ácidos Nucleicos/ultraestructura , ARN/efectos adversos , ARN/inmunología , ARN/uso terapéutico
12.
Small ; 13(42)2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28922553

RESUMEN

In the past few years, the study of therapeutic RNA nanotechnology has expanded tremendously to encompass a large group of interdisciplinary sciences. It is now evident that rationally designed programmable RNA nanostructures offer unique advantages in addressing contemporary therapeutic challenges such as distinguishing target cell types and ameliorating disease. However, to maximize the therapeutic benefit of these nanostructures, it is essential to understand the immunostimulatory aptitude of such tools and identify potential complications. This paper presents a set of 16 nanoparticle platforms that are highly configurable. These novel nucleic acid based polygonal platforms are programmed for controllable self-assembly from RNA and/or DNA strands via canonical Watson-Crick interactions. It is demonstrated that the immunostimulatory properties of these particular designs can be tuned to elicit the desired immune response or lack thereof. To advance the current understanding of the nanoparticle properties that contribute to the observed immunomodulatory activity and establish corresponding designing principles, quantitative structure-activity relationship modeling is conducted. The results demonstrate that molecular weight, together with melting temperature and half-life, strongly predicts the observed immunomodulatory activity. This framework provides the fundamental guidelines necessary for the development of a new library of nanoparticles with predictable immunomodulatory activity.


Asunto(s)
Inmunomodulación , Microglía/citología , Ácidos Nucleicos/química , Relación Estructura-Actividad Cuantitativa , Línea Celular Tumoral , ADN/química , Humanos , ARN/química , Reproducibilidad de los Resultados
13.
Transfus Apher Sci ; 56(2): 179-189, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28007431

RESUMEN

Extracorporeal Photochemotherapy (ECP) is a widely applied anti-cancer immunotherapy for patients with cutaneous T cell lymphoma (CTCL). By using apoptotic malignant cells as a source of patient-specific tumor antigen, it enables clinically relevant and curative anti-CTCL immunity, with potential efficacy in other tumors. Currentmethods to track patient-specific responses are tedious, and new methods are needed to assess putative global immunity. We developed a clinically practical method to assess antigen-specific T cell activation that does not rely on knowledge of the particular antigen, thereby eliminating the requirement for patient-specific reagents. In the OT-I transgenic murine system, we quantified calcium flux to reveal early T cell engagement by antigen presenting cells constitutively displaying a model antigenic peptide, ovalbumin (OVA)-derived SIINFEKL. We detected calcium flux in OVA-specific T cells, triggered by specific T cell receptor engagement by SIINFEKL peptide-loaded DC. This approach led to sensitive detection of antigen-specific calcium flux (ACF) down to a peptide-loading concentration of ∼10-3uM and at a frequency of ∼0.1% OT-I cells among wild-type (WT), non-responding cells. Antigen-specific T cells were detected in spleen, lymph nodes, and peripheral blood after adoptive transfer into control recipient mice. Methods like this for assessing therapeutic response are lacking in patients currently on immune-based therapies, such as ECP, where assessment of clinical response is made by delayed measurement of the size of the malignant clone. These findings suggest an early, practical way to measure therapeutically-induced anti-tumor responses in ECP-treated patients that have been immunized against their malignant cells.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Antígenos de Neoplasias/inmunología , Linfoma Cutáneo de Células T/inmunología , Fotoféresis , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Animales , Células Presentadoras de Antígenos/patología , Antígenos de Neoplasias/genética , Linfoma Cutáneo de Células T/genética , Linfoma Cutáneo de Células T/patología , Linfoma Cutáneo de Células T/terapia , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/patología
14.
J Biol Chem ; 291(17): 8931-50, 2016 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-26719339

RESUMEN

Here we report a "configuration-dependent" mechanism of action for IL-15:IL-15Rα (heterodimeric IL-15 or hetIL-15) where the manner by which IL-15:IL-15Rα molecules are presented to target cells significantly affects its function as a vaccine adjuvant. Although the cellular mechanism of IL-15 trans-presentation via IL-15Rα and its importance for IL-15 function have been described, the full effect of the IL-15:IL-15Rα configuration on responding cells is not yet known. We found that trans-presenting IL-15:IL-15Rα in a multivalent fashion on the surface of antigen-encapsulating nanoparticles enhanced the ability of nanoparticle-treated dendritic cells (DCs) to stimulate antigen-specific CD8(+) T cell responses. Localization of multivalent IL-15:IL-15Rα and encapsulated antigen to the same DC led to maximal T cell responses. Strikingly, DCs incubated with IL-15:IL-15Rα-coated nanoparticles displayed higher levels of functional IL-15 on the cell surface, implicating a mechanism for nanoparticle-mediated transfer of IL-15 to the DC surface. Using artificial antigen-presenting cells to highlight the effect of IL-15 configuration on DCs, we showed that artificial antigen-presenting cells presenting IL-15:IL-15Rα increased the sensitivity and magnitude of the T cell response, whereas IL-2 enhanced the T cell response only when delivered in a paracrine fashion. Therefore, the mode of cytokine presentation (configuration) is important for optimal immune responses. We tested the effect of configuration dependence in an aggressive model of murine melanoma and demonstrated significantly delayed tumor progression induced by IL-15:IL-15Rα-coated nanoparticles in comparison with monovalent IL-15:IL-15Rα. The novel mechanism of IL-15 transfer to the surface of antigen-processing DCs may explain the enhanced potency of IL-15:IL-15Rα-coated nanoparticles for antigen delivery.


Asunto(s)
Presentación de Antígeno/efectos de los fármacos , Antígenos de Neoplasias , Linfocitos T CD8-positivos/inmunología , Materiales Biocompatibles Revestidos/farmacología , Células Dendríticas/inmunología , Inmunidad Celular/efectos de los fármacos , Interleucina-15 , Nanopartículas , Neoplasias Experimentales , Receptores de Interleucina-15/inmunología , Animales , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/farmacología , Humanos , Interleucina-15/inmunología , Interleucina-15/farmacología , Ratones , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología
15.
Int J Nanomedicine ; 9: 5231-46, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25419128

RESUMEN

Targeting antigen to dendritic cells (DCs) is a powerful and novel strategy for vaccination. Priming or loading DCs with antigen controls whether subsequent immunity will develop and hence whether effective vaccination can be achieved. The goal of our present work was to increase the potency of DC-based antitumor vaccines by overcoming inherent limitations associated with antigen stability and cross-presentation. Nanoparticles prepared from the biodegradable polymer poly(lactic-co-glycolic acid) have been extensively used in clinical settings for drug delivery and are currently the subject of intensive investigation as antigen delivery vehicles for vaccine applications. Here we describe a nanoparticulate delivery system with the ability to simultaneously carry a high density of protein-based antigen while displaying a DC targeting ligand on its surface. Utilizing a targeting motif specific for the DC-associated surface ligand DEC-205, we show that targeted nanoparticles encapsulating a MART-127-35 peptide are both internalized and cross-presented with significantly higher efficiency than isotype control-coated nanoparticles in human cells. In addition, the DEC-205-labeled nanoparticles rapidly escape from the DC endosomal compartment and do not colocalize with markers of early (EEA-1) or late endosome/lysosome (LAMP-1). This indicates that encapsulated antigens delivered by nanoparticles may have direct access to the class I cytoplasmic major histocompatibility complex loading machinery, overcoming the need for "classical" cross-presentation and facilitating heightened DC stimulation of anti-tumor CD8(+) T-cells. These results indicate that this delivery system provides a flexible and versatile methodology to deliver melanoma-associated antigen to DCs, with both high efficiency and heightened potency.


Asunto(s)
Antígenos CD/inmunología , Vacunas contra el Cáncer/administración & dosificación , Células Dendríticas/inmunología , Ácido Láctico/química , Lectinas Tipo C/inmunología , Antígeno MART-1/administración & dosificación , Melanoma/inmunología , Nanopartículas/química , Ácido Poliglicólico/química , Receptores de Superficie Celular/inmunología , Presentación de Antígeno/efectos de los fármacos , Vacunas contra el Cáncer/inmunología , Humanos , Ácido Láctico/inmunología , Antígeno MART-1/inmunología , Melanoma/terapia , Antígenos de Histocompatibilidad Menor , Terapia Molecular Dirigida , Nanopartículas/administración & dosificación , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
16.
Nat Nanotechnol ; 9(8): 639-47, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25086604

RESUMEN

Clinical translation of cell therapies requires strategies that can manufacture cells efficiently and economically. One promising way to reproducibly expand T cells for cancer therapy is by attaching the stimuli for T cells onto artificial substrates with high surface area. Here, we show that a carbon nanotube-polymer composite can act as an artificial antigen-presenting cell to efficiently expand the number of T cells isolated from mice. We attach antigens onto bundled carbon nanotubes and combined this complex with polymer nanoparticles containing magnetite and the T-cell growth factor interleukin-2 (IL-2). The number of T cells obtained was comparable to clinical standards using a thousand-fold less soluble IL-2. T cells obtained from this expansion were able to delay tumour growth in a murine model for melanoma. Our results show that this composite is a useful platform for generating large numbers of cytotoxic T cells for cancer immunotherapy.


Asunto(s)
Proteínas Inmovilizadas/química , Melanoma/terapia , Nanotubos de Carbono/química , Polímeros/química , Linfocitos T/citología , Linfocitos T/trasplante , Animales , Antígenos/química , Antígenos/inmunología , Técnicas de Cultivo de Célula/métodos , Proliferación Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Humanos , Proteínas Inmovilizadas/inmunología , Inmunoterapia Adoptiva , Interleucina-2/química , Interleucina-2/inmunología , Melanoma/inmunología , Ratones , Linfocitos T/inmunología
17.
Proc Natl Acad Sci U S A ; 108(50): 20095-100, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22109549

RESUMEN

Implantation of biomaterials and devices into soft tissues leads to the development of the foreign body response (FBR), which can interfere with implant function and eventually lead to failure. The FBR consists of overlapping acute and persistent inflammatory phases coupled with collagenous encapsulation and currently there are no therapeutic options. Initiation of the FBR involves macrophage activation, proceeding to giant cell formation, fibroblast activation, and collagen matrix deposition. Despite the recognition of this sequence of events, the molecular pathways required for the FBR have not been elucidated. We have identified that the acute inflammatory response to biomaterials requires nucleotide-binding domain and leucine-rich repeat-containing 3 (Nlrp3), apoptosis-associated speck-like protein containing CARD (Asc), and caspase-1, as well as plasma membrane cholesterol, and Syk signaling. Full development of the FBR is dependent on Asc and caspase-1, but not Nlrp3. The common antiinflammatory drug aspirin can reduce inflammasome activation and significantly reduce the FBR. Taken together, these findings expand the role of the inflammasome from one of sensing damage associated molecular patterns (DAMPs) to sensing all particulate matter irrespective of size. In addition, implication of the inflammasome in biomaterial recognition identifies key pathways, which can be targeted to limit the FBR.


Asunto(s)
Materiales Biocompatibles/efectos adversos , Caspasa 1/metabolismo , Proteínas del Citoesqueleto/metabolismo , Reacción a Cuerpo Extraño/patología , Inflamasomas/metabolismo , Inflamación/patología , Administración Oral , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Aspirina/administración & dosificación , Aspirina/efectos adversos , Proteínas Adaptadoras de Señalización CARD , Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/metabolismo , Análisis por Conglomerados , Reacción a Cuerpo Extraño/complicaciones , Reacción a Cuerpo Extraño/enzimología , Reacción a Cuerpo Extraño/inmunología , Células Gigantes/efectos de los fármacos , Células Gigantes/inmunología , Células Gigantes/patología , Inflamación/complicaciones , Inflamación/enzimología , Inflamación/inmunología , Interleucina-1beta/biosíntesis , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/patología , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Microesferas , Proteína con Dominio Pirina 3 de la Familia NLR , Polimetil Metacrilato/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...