Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Annu Rev Immunol ; 41: 153-179, 2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-36696570

RESUMEN

Modulation of the immune system is an important therapeutic strategy in a wide range of diseases, and is fundamental to the development of vaccines. However, optimally safe and effective immunotherapy requires precision in the delivery of stimulatory cues to the right cells at the right place and time, to avoid toxic overstimulation in healthy tissues or incorrect programming of the immune response. To this end, biomaterials are being developed to control the location, dose, and timing of vaccines and immunotherapies. Here we discuss fundamental concepts of how biomaterials are used to enhance immune modulation, and evidence from preclinical and clinical studies of how biomaterials-mediated immune engineering can impact the development of new therapeutics. We focus on immunological mechanisms of action and in vivo modulation of the immune system, and we also discuss challenges to be overcome to speed translation of these technologies to the clinic.


Asunto(s)
Neoplasias , Vacunas , Humanos , Animales , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/uso terapéutico , Inmunoterapia , Sistema Inmunológico , Inmunidad
2.
Nanomedicine ; 22: 102099, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31648039

RESUMEN

Lipid particles for drug delivery can be modified to create multilayer vesicles with higher stability and improved cargo interaction. Here, we used lipids capable of forming hydrogen bonds instead of covalent bonds and designed stable vesicles-inside-vesicles with a high capacity of entrapping antimalarial drugs such as chloroquine (hydrophilic) and Artemisinin (lipophilic). In vitro treatment of the drug-sensitive P. falciparum strain NF54 showed that encapsulated drugs resulted in 72% and 60% lower IC50 values for each drug, respectively. Fluorochrome-labeling of a cargo-peptide or of membrane-resident lipids indicated that vesicles interacted more specifically with parasite-infected erythrocytes than with normal red blood cells. Accordingly, vesicle-confined chloroquine was able to elicit a stronger antiparasitic effect than free chloroquine in a lethal murine model of infection. Being permissive not only to small molecules but also to larger peptides, hydrogen-bond linked multilamellar liposomes are a very promising approach for enhanced drug delivery.


Asunto(s)
Antimaláricos/farmacología , Nanopartículas/química , Animales , Antimaláricos/administración & dosificación , Antimaláricos/uso terapéutico , Artemisininas/farmacología , Cloroquina/farmacología , Reactivos de Enlaces Cruzados/química , Sistemas de Liberación de Medicamentos , Enlace de Hidrógeno , Liposomas , Malaria Falciparum/tratamiento farmacológico , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nanopartículas/ultraestructura , Tamaño de la Partícula , Plasmodium falciparum/efectos de los fármacos , Resultado del Tratamiento
3.
Bioconjug Chem ; 29(4): 1131-1140, 2018 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-29465986

RESUMEN

The development of synthetic nanomaterials that could embed within, penetrate, or induce fusion between membranes without permanent disruption would have great significance for biomedical applications. Here we describe structure-function relationships of highly water-soluble gold nanoparticles comprised of an ∼1.5-5 nm diameter metal core coated by an amphiphilic organic ligand shell, which exhibit membrane embedding and fusion activity mediated by the surface ligands. Using an environment-sensitive dye anchored within the ligand shell as a sensor of membrane embedding, we demonstrate that particles with core sizes of ∼2-3 nm are capable of embedding within and penetrating fluid bilayers. At the nanoscale, these particles also promote spontaneous fusion of liposomes or spontaneously embed within intact liposomal vesicles. These studies provide nanoparticle design and selection principles that could be used in drug delivery applications, as membrane stains, or for the creation of novel organic/inorganic nanomaterial self-assemblies.


Asunto(s)
Membrana Dobles de Lípidos , Fusión de Membrana , Nanopartículas/química , Permeabilidad , Compuestos de Boro/química , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Liposomas , Tamaño de la Partícula , Electricidad Estática , Relación Estructura-Actividad
4.
Annu Rev Biomed Eng ; 17: 317-49, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26421896

RESUMEN

Strategies to enhance, suppress, or qualitatively shape the immune response are of importance for diverse biomedical applications, such as the development of new vaccines, treatments for autoimmune diseases and allergies, strategies for regenerative medicine, and immunotherapies for cancer. However, the intricate cellular and molecular signals regulating the immune system are major hurdles to predictably manipulating the immune response and developing safe and effective therapies. To meet this challenge, biomaterials are being developed that control how, where, and when immune cells are stimulated in vivo, and that can finely control their differentiation in vitro. We review recent advances in the field of biomaterials for immunomodulation, focusing particularly on designing biomaterials to provide controlled immunostimulation, targeting drugs and vaccines to lymphoid organs, and serving as scaffolds to organize immune cells and emulate lymphoid tissues. These ongoing efforts highlight the many ways in which biomaterials can be brought to bear to engineer the immune system.


Asunto(s)
Materiales Biocompatibles , Inmunomodulación , Animales , Células Presentadoras de Antígenos/inmunología , Células Artificiales/inmunología , Ingeniería Biomédica , Células Dendríticas/inmunología , Humanos , Inmunización , Factores Inmunológicos/administración & dosificación , Inmunoterapia , Tejido Linfoide/inmunología , Nanopartículas , Nanotecnología , Proteínas/inmunología , Biología de Sistemas , Ingeniería de Tejidos
5.
Nat Mater ; 12(4): 367-76, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23353628

RESUMEN

DNA vaccines have many potential benefits but have failed to generate robust immune responses in humans. Recently, methods such as in vivo electroporation have demonstrated improved performance, but an optimal strategy for safe, reproducible, and pain-free DNA vaccination remains elusive. Here we report an approach for rapid implantation of vaccine-loaded polymer films carrying DNA, immune-stimulatory RNA, and biodegradable polycations into the immune-cell-rich epidermis, using microneedles coated with releasable polyelectrolyte multilayers. Films transferred into the skin following brief microneedle application promoted local transfection and controlled the persistence of DNA and adjuvants in the skin from days to weeks, with kinetics determined by the film composition. These 'multilayer tattoo' DNA vaccines induced immune responses against a model HIV antigen comparable to electroporation in mice, enhanced memory T-cell generation, and elicited 140-fold higher gene expression in non-human primate skin than intradermal DNA injection, indicating the potential of this strategy for enhancing DNA vaccination.


Asunto(s)
Adyuvantes Inmunológicos/química , Polímeros/química , Vacunas de ADN/química , Animales , Electroporación/métodos , Humanos , Inyecciones Intradérmicas , Ratones , Piel/inmunología , Vacunación/métodos , Vacunas de ADN/inmunología
6.
Biomacromolecules ; 15(7): 2475-81, 2014 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-24894061

RESUMEN

Lipid-coated poly(lactide-co-glycolide) microparticles (LCMPs) consist of a solid polymer core wrapped by a surface lipid bilayer. Previous studies demonstrated that immunization with LCMPs surface-decorated with nanograms of antigen elicit potent humoral immune responses in mice. However, the mechanism of action for these vaccines remained unclear, as LCMPs are too large to drain efficiently to lymph nodes from the vaccination site. Here, we characterized the stability of the lipid envelope of LCMPs and discovered that in the presence of serum the lipid coating of the particles spontaneously delaminates, shedding antigen-displaying vesicles. Lipid delamination generated 180 nm liposomes in a temperature- and lipid/serum-dependent manner. Vesicle shedding was restricted by inclusion of high-TM lipids or cholesterol in the LCMP coating. Administration of LCMPs bearing stabilized lipid envelopes generated weaker antibody responses than those of shedding-competent LCMPs, suggesting that in situ release of antigen-loaded vesicles plays a key role in the remarkable potency of LCMPs as vaccine adjuvants.


Asunto(s)
Adyuvantes Inmunológicos/química , Ácido Láctico/química , Ácido Poliglicólico/química , Vacunas/química , Adyuvantes Inmunológicos/farmacología , Animales , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Femenino , Ácido Láctico/farmacología , Liposomas , Ratones , Ratones Endogámicos BALB C , Nanopartículas/química , Ácido Poliglicólico/farmacología , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Polímeros/química
7.
Proc Natl Acad Sci U S A ; 108(38): 15745-50, 2011 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-21896725

RESUMEN

Recent studies have demonstrated a simple, potentially universal strategy to enhance vaccine potency, via intralymph node (i.LN) injection. To date, intranodal immunization studies have focused on the delivery of unadjuvanted vaccines (e.g., naked DNA, peptide, or protein). We hypothesized that combining i.LN vaccination with controlled release biomaterials permitting sustained dosing of molecular adjuvants to the local tissue microenvironment would further enhance this promising vaccination strategy. To test this idea, we encapsulated the Toll-like receptor-3 ligand poly(inosinic:cytidylic acid) (polyIC) in biodegradable poly(lactide-co-glycolide) microparticles (MPs) designed to remain extracellular and release polyIC in the LN over several days. Intranodal injection of MPs increased persistence of polyIC in LNs compared to the same dose of soluble polyIC or polyIC formulated in nanoparticles, leading to increased accumulation of Toll-like receptor agonist in LN-resident antigen presenting cells and more enduring dendritic cell activation. Intralymph node injection of ovalbumin mixed with polyIC-releasing MPs enhanced the humoral response and expanded ovalbumin-specific T cells to frequencies as high as 18% among all CD8(+) cells following a single injection (8.2-fold greater than the same vaccine given i.m.), a response that could not be matched by antigen mixed with polyIC-loaded nanoparticles or a 10-fold greater dose of soluble polyIC. Thus, i.LN immunization with slow release-formulated adjuvants may be a broadly applicable strategy to enhance therapeutic or prophylactic vaccines.


Asunto(s)
Microambiente Celular/inmunología , Ganglios Linfáticos/inmunología , Nanopartículas/química , Poli I-C/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Proliferación Celular/efectos de los fármacos , Células Dendríticas/inmunología , Femenino , Citometría de Flujo , Colorantes Fluorescentes/química , Inmunización/métodos , Mediciones Luminiscentes/métodos , Ganglios Linfáticos/citología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Tamaño de la Partícula , Poli I-C/administración & dosificación , Poli I-C/química , Poliglactina 910/química , Linfocitos T/inmunología
9.
Science ; 381(6657): 508-514, 2023 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-37535724

RESUMEN

Proton leakage from organelles is a common signal for noncanonical light chain 3B (LC3B) lipidation and inflammasome activation, processes induced upon stimulator of interferon genes (STING) activation. On the basis of structural analysis, we hypothesized that human STING is a proton channel. Indeed, we found that STING activation induced a pH increase in the Golgi and that STING reconstituted in liposomes enabled transmembrane proton transport. Compound 53 (C53), a STING agonist that binds the putative channel interface, blocked STING-induced proton flux in the Golgi and in liposomes. STING-induced LC3B lipidation and inflammasome activation were also inhibited by C53, suggesting that STING's channel activity is critical for these two processes. Thus, STING's interferon-induction function can be decoupled from its roles in LC3B lipidation and inflammasome activation.


Asunto(s)
Canales Iónicos , Proteínas de la Membrana , Protones , Humanos , Aparato de Golgi/metabolismo , Concentración de Iones de Hidrógeno , Inflamasomas/metabolismo , Canales Iónicos/agonistas , Canales Iónicos/química , Canales Iónicos/metabolismo , Liposomas , Proteínas de la Membrana/agonistas , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Dominios Proteicos , Células HEK293
10.
Nat Mater ; 10(3): 243-51, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21336265

RESUMEN

Vaccines based on recombinant proteins avoid the toxicity and antivector immunity associated with live vaccine (for example, viral) vectors, but their immunogenicity is poor, particularly for CD8(+) T-cell responses. Synthetic particles carrying antigens and adjuvant molecules have been developed to enhance subunit vaccines, but in general these materials have failed to elicit CD8(+) T-cell responses comparable to those for live vectors in preclinical animal models. Here, we describe interbilayer-crosslinked multilamellar vesicles formed by crosslinking headgroups of adjacent lipid bilayers within multilamellar vesicles. Interbilayer-crosslinked vesicles stably entrapped protein antigens in the vesicle core and lipid-based immunostimulatory molecules in the vesicle walls under extracellular conditions, but exhibited rapid release in the presence of endolysosomal lipases. We found that these antigen/adjuvant-carrying vesicles form an extremely potent whole-protein vaccine, eliciting endogenous T-cell and antibody responses comparable to those for the strongest vaccine vectors. These materials should enable a range of subunit vaccines and provide new possibilities for therapeutic protein delivery.


Asunto(s)
Inmunidad Celular , Inmunidad Humoral , Liposomas/química , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Adyuvantes Inmunológicos/química , Animales , Portadores de Fármacos , Diseño de Fármacos , Memoria Inmunológica , Membrana Dobles de Lípidos/química , Liposomas/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Vacunas Sintéticas/química , Vacunas Virales/química , Vacunas Virales/inmunología
11.
Mol Pharm ; 8(3): 774-87, 2011 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-21417235

RESUMEN

Biodegradable core--shell structured nanoparticles with a poly(ß-amino ester) (PBAE) core enveloped by a phospholipid bilayer shell were developed for in vivo mRNA delivery with a view toward delivery of mRNA-based vaccines. The pH-responsive PBAE component was chosen to promote endosome disruption, while the lipid surface layer was selected to minimize toxicity of the polycation core. Messenger RNA was efficiently adsorbed via electrostatic interactions onto the surface of these net positively charged nanoparticles. In vitro, mRNA-loaded particle uptake by dendritic cells led to mRNA delivery into the cytosol with low cytotoxicity, followed by translation of the encoded protein in these difficult-to-transfect cells at a frequency of ~30%. Particles loaded with mRNA administered intranasally (i.n.) in mice led to the expression of the reporter protein luciferase in vivo as soon as 6 h after administration, a time point when naked mRNA given i.n. showed no expression. At later time points, luciferase expression was detected in naked mRNA-treated mice, but this group showed a wide variation in levels of transfection, compared to particle-treated mice. This system may thus be promising for noninvasive delivery of mRNA-based vaccines.


Asunto(s)
Portadores de Fármacos/química , Lípidos/química , Nanopartículas/química , Polímeros/química , ARN Mensajero/administración & dosificación , ARN Mensajero/química , Animales , Línea Celular , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación
12.
Adv Mater ; 33(7): e2006057, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33448062

RESUMEN

Short-wave infrared (SWIR, 900-1700 nm) enables in vivo imaging with high spatiotemporal resolution and penetration depth due to the reduced tissue autofluorescence and decreased photon scattering at long wavelengths. Although small organic SWIR dye molecules have excellent biocompatibility, they have been rarely exploited as compared to their inorganic counterparts, mainly due to their low quantum yield. To increase their brightness, in this work, the SWIR dye molecules are placed in close proximity to gold nanorods (AuNRs) for surface plasmon-enhanced emission. The fluorescence enhancement is optimized by controlling the dye-to-AuNR number ratio and up to ≈45-fold enhancement factor is achieved. In addition, the results indicate that the highest dye-to-AuNR number ratio gives the highest emission intensity per weight and this is used for synthesizing SWIR imaging probes using layer-by-layer (LbL) technique with polymer coating protection. Then, the SWIR imaging probes are applied for in vivo imaging of ovarian cancer and the surface coating effect on intratumor distribution of the imaging probes is investigated in two orthotopic ovarian cancer models. Lastly, it is demonstrated that the plasmon-enhanced SWIR imaging probe has great potential for fluorescence imaging-guided surgery by showing its capability to detect sub-millimeter-sized tumors.


Asunto(s)
Materiales Biocompatibles/química , Colorantes Fluorescentes/química , Oro/química , Nanotubos/química , Imagen Óptica/métodos , Neoplasias Ováricas/diagnóstico por imagen , Animales , Refuerzo Biomédico , Línea Celular Tumoral , Femenino , Humanos , Rayos Infrarrojos , Luciferasas/química , Luciferasas/genética , Ratones Desnudos , Polímeros/química , Ondas de Radio , Propiedades de Superficie , Distribución Tisular
13.
Adv Drug Deliv Rev ; 158: 91-115, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32598970

RESUMEN

Vaccines are one of the most powerful technologies supporting public health. The adaptive immune response induced by immunization arises following appropriate activation and differentiation of T and B cells in lymph nodes. Among many parameters impacting the resulting immune response, the presence of antigen and inflammatory cues for an appropriate temporal duration within the lymph nodes, and further within appropriate subcompartments of the lymph nodes- the right timing and location- play a critical role in shaping cellular and humoral immunity. Here we review recent advances in our understanding of how vaccine kinetics and biodistribution impact adaptive immunity, and the underlying immunological mechanisms that govern these responses. We discuss emerging approaches to engineer these properties for future vaccines, with a focus on subunit vaccines.


Asunto(s)
Vacunas/inmunología , Vacunas/farmacocinética , Adyuvantes Inmunológicos/farmacología , Linfocitos B/inmunología , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Humanos , Inmunidad Humoral/inmunología , Mediadores de Inflamación/metabolismo , Liposomas/metabolismo , Ganglios Linfáticos/inmunología , Nanopartículas/metabolismo , Plásmidos/farmacocinética , ARN Mensajero/farmacocinética , Linfocitos T/inmunología , Distribución Tisular
14.
Nat Cancer ; 1(9): 882-893, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-34447945

RESUMEN

Therapies that synergistically stimulate immunogenic cancer cell death (ICD), inflammation, and immune priming are of great interest for cancer immunotherapy. However, even multi-agent therapies often fail to trigger all of the steps necessary for self-sustaining anti-tumor immunity. Here we describe self-replicating RNAs encapsulated in lipid nanoparticles (LNP-replicons), which combine three key elements: (1) an LNP composition that potently promotes ICD, (2) RNA that stimulates danger sensors in transfected cells, and (3) RNA-encoded IL-12 for modulation of immune cells. Intratumoral administration of LNP-replicons led to high-level expression of IL-12, stimulation of a type I interferon response, and cancer cell ICD, resulting in a highly inflamed tumor microenvironment and priming of systemic anti-tumor immunity. In several mouse models of cancer, a single intratumoral injection of replicon-LNPs eradicated large established tumors, induced protective immune memory, and enabled regression of distal uninjected tumors. LNP-replicons are thus a promising multifunctional single-agent immunotherapeutic.


Asunto(s)
Nanopartículas , Neoplasias , Animales , Inmunoterapia/métodos , Interleucina-12/genética , Liposomas , Ratones , Neoplasias/genética , ARN , Microambiente Tumoral
15.
Sci Transl Med ; 12(569)2020 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-33177180

RESUMEN

Adoptive cell transfer of ex vivo expanded regulatory T cells (Tregs) has shown immense potential in animal models of auto- and alloimmunity. However, the effective translation of such Treg therapies to the clinic has been slow. Because Treg homeostasis is known to require continuous T cell receptor (TCR) ligation and exogenous interleukin-2 (IL-2), some investigators have explored the use of low-dose IL-2 injections to increase endogenous Treg responses. Systemic IL-2 immunotherapy, however, can also lead to the activation of cytotoxic T lymphocytes and natural killer cells, causing adverse therapeutic outcomes. Here, we describe a drug delivery platform, which can be engineered to autostimulate Tregs with IL-2 in response to TCR-dependent activation, and thus activate these cells in sites of antigen encounter. To this end, protein nanogels (NGs) were synthesized with cleavable bis(N-hydroxysuccinimide) cross-linkers and IL-2/Fc fusion (IL-2) proteins to form particles that release IL-2 under reducing conditions, as found at the surface of T cells receiving stimulation through the TCR. Tregs surface-conjugated with IL-2 NGs were found to have preferential, allograft-protective effects relative to unmodified Tregs or Tregs stimulated with systemic IL-2. We demonstrate that murine and human NG-modified Tregs carrying an IL-2 cargo perform better than conventional Tregs in suppressing alloimmunity in murine and humanized mouse allotransplantation models. In all, the technology presented in this study has the potential to improve Treg transfer therapy by enabling the regulated spatiotemporal provision of IL-2 to antigen-primed Tregs.


Asunto(s)
Interleucina-2 , Linfocitos T Reguladores , Animales , Ratones , Nanogeles , Receptores de Antígenos de Linfocitos T , Transducción de Señal
16.
Nat Mater ; 7(7): 588-95, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18500347

RESUMEN

Nanoscale objects are typically internalized by cells into membrane-bounded endosomes and fail to access the cytosolic cell machinery. Whereas some biomacromolecules may penetrate or fuse with cell membranes without overt membrane disruption, no synthetic material of comparable size has shown this property yet. Cationic nano-objects pass through cell membranes by generating transient holes, a process associated with cytotoxicity. Studies aimed at generating cell-penetrating nanomaterials have focused on the effect of size, shape and composition. Here, we compare membrane penetration by two nanoparticle 'isomers' with similar composition (same hydrophobic content), one coated with subnanometre striations of alternating anionic and hydrophobic groups, and the other coated with the same moieties but in a random distribution. We show that the former particles penetrate the plasma membrane without bilayer disruption, whereas the latter are mostly trapped in endosomes. Our results offer a paradigm for analysing the fundamental problem of cell-membrane-penetrating bio- and macro-molecules.


Asunto(s)
Membrana Celular/metabolismo , Nanopartículas del Metal , Animales , Transporte Biológico Activo , Línea Celular , Permeabilidad de la Membrana Celular , Materiales Biocompatibles Revestidos/química , Células Dendríticas/metabolismo , Células Dendríticas/ultraestructura , Endocitosis , Oro/química , Interacciones Hidrofóbicas e Hidrofílicas , Nanopartículas del Metal/química , Ratones , Microscopía Electrónica de Transmisión , Nanotecnología , Tamaño de la Partícula , Propiedades de Superficie
17.
Biomacromolecules ; 10(4): 732-41, 2009 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-19235932

RESUMEN

Biocompatible oils are used in a variety of medical applications ranging from vaccine adjuvants to vehicles for oral drug delivery. To enable such nonpolar organic phases to serve as reservoirs for delivery of hydrophilic compounds, we explored the ability of block copolymer micelles in organic solvents to sequester proteins for sustained release across an oil-water interface. Self-assembly of the block copolymer, poly(-caprolactone)-block-poly(2-vinyl pyridine) (PCL-b-P2VP), was investigated in toluene and oleic acid, a biocompatible naturally occurring fatty acid. Micelle formation in toluene was characterized by dynamic light scattering (DLS) and atomic force microscopy (AFM) imaging of micelles cast onto silicon substrates. Cryogenic transmission electron microscopy confirmed a spherical morphology in oleic acid. Studies of homopolymer solubility implied that micelles in oleic acid consist of a P2VP corona and a PCL core, while P2VP formed the core of micelles assembled in toluene. The loading of two model proteins (ovalbumin (ova) and bovine serum albumin (BSA)) into micelles was demonstrated with loadings as high as 7.8% wt of protein per wt of P2VP in oleic acid. Characterization of block copolymer morphology in the two solvents after protein loading revealed spherical particles with similar size distributions to the as-assembled micelles. Release of ova from micelles in oleic acid was sustained for 12-30 h upon placing the oil phase in contact with an aqueous bath. Unique to the situation of micelle assembly in an oily phase, the data suggest protein is sequestered in the P2VP corona block of PCL-b-P2VP micelles in oleic acid. More conventionally, protein loading occurs in the P2VP core of micelles assembled in toluene.


Asunto(s)
Micelas , Ácido Oléico/química , Ovalbúmina/administración & dosificación , Poliésteres/química , Polímeros/química , Polivinilos/química , Albúmina Sérica Bovina/administración & dosificación , Animales , Materiales Biocompatibles/farmacología , Bovinos , Materiales Biocompatibles Revestidos , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Microscopía de Fuerza Atómica , Microscopía Electrónica de Transmisión , Polímeros/síntesis química , Polímeros/farmacología , Dispersión de Radiación , Tolueno/química
18.
Biomater Sci ; 7(4): 1345-1357, 2019 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-30698174

RESUMEN

Interleukin-2 (IL-2) is a potent T-cell mitogen that can adjuvant anti-cancer adoptive T-cell transfer (ACT) immunotherapy by promoting T-cell engraftment. However, the clinical applications of IL-2 in combination with ACT are greatly hindered by the severe adverse effects such as vascular leak syndrome (VLS). Here, we developed a synthetic delivery strategy for IL-2 via backpacking redox-responsive IL-2/Fc nanogels (NGs) to the plasma membrane of adoptively transferred T-cells. The NGs prepared by traceless chemical cross-linking of cytokine proteins selectively released the cargos in response to T-cell receptor activation upon antigen recognition in tumors. We found that IL-2/Fc delivered by T-cell surface-bound NGs expanded transferred tumor-reactive T-cells 80-fold more than the free IL-2/Fc of an equivalent dose administered systemically and showed no effects on tumor-infiltrating regulatory T-cell expansion. Intriguingly, IL-2/Fc NG backpacks that facilitated a sustained and slow release of IL-2/Fc also promoted the CD8+ memory precursor differentiation and induced less T-cell exhaustion in vitro compared to free IL-2/Fc. The controlled responsive delivery of IL-2/Fc enabled the safe administration of repeated doses of the stimulant cytokine with no overt toxicity and improved efficacy against melanoma metastases in a mice model.


Asunto(s)
Interleucina-2/farmacología , Melanoma/patología , Polietilenglicoles/farmacología , Polietileneimina/farmacología , Linfocitos T/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Interleucina-2/síntesis química , Interleucina-2/química , Melanoma/inmunología , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Nanogeles , Oxidación-Reducción , Polietilenglicoles/síntesis química , Polietilenglicoles/química , Polietileneimina/síntesis química , Polietileneimina/química , Linfocitos T/inmunología , Linfocitos T/patología
19.
Science ; 363(6427): 649-654, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30573546

RESUMEN

In vaccine design, antigens are often arrayed in a multivalent nanoparticle form, but in vivo mechanisms underlying the enhanced immunity elicited by such vaccines remain poorly understood. We compared the fates of two different heavily glycosylated HIV antigens, a gp120-derived mini-protein and a large, stabilized envelope trimer, in protein nanoparticle or "free" forms after primary immunization. Unlike monomeric antigens, nanoparticles were rapidly shuttled to the follicular dendritic cell (FDC) network and then concentrated in germinal centers in a complement-, mannose-binding lectin (MBL)-, and immunogen glycan-dependent manner. Loss of FDC localization in MBL-deficient mice or via immunogen deglycosylation significantly affected antibody responses. These findings identify an innate immune-mediated recognition pathway promoting antibody responses to particulate antigens, with broad implications for humoral immunity and vaccine design.


Asunto(s)
Vacunas contra el SIDA/inmunología , Formación de Anticuerpos , Centro Germinal/inmunología , Proteína gp120 de Envoltorio del VIH/inmunología , Infecciones por VIH/inmunología , Inmunidad Innata , Polisacáridos/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Pruebas de Fijación del Complemento , Proteínas del Sistema Complemento/inmunología , Células Dendríticas/inmunología , Femenino , Anticuerpos Anti-VIH/inmunología , Infecciones por VIH/prevención & control , Liposomas , Lectina de Unión a Manosa/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Complejos Multiproteicos , Nanopartículas , Receptores de Complemento/inmunología
20.
Nat Commun ; 9(1): 6, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29295974

RESUMEN

Immunostimulatory agents such as agonistic anti-CD137 and interleukin (IL)-2 generate effective anti-tumor immunity but also elicit serious toxicities, hampering their clinical application. Here we show that combination therapy with anti-CD137 and an IL-2-Fc fusion achieves significant initial anti-tumor activity, but also lethal immunotoxicity deriving from stimulation of circulating leukocytes. To overcome this toxicity, we demonstrate that anchoring IL-2 and anti-CD137 on the surface of liposomes allows these immune agonists to rapidly accumulate in tumors while lowering systemic exposure. In multiple tumor models, immunoliposome delivery achieves anti-tumor activity equivalent to free IL-2/anti-CD137 but with the complete absence of systemic toxicity. Immunoliposomes stimulated tumor infiltration by cytotoxic lymphocytes, cytokine production, and granzyme expression, demonstrating equivalent immunostimulatory effects to the free drugs in the local tumor microenvironment. Thus, surface-anchored particle delivery may provide a general approach to exploit the potent stimulatory activity of immune agonists without debilitating systemic toxicities.


Asunto(s)
Antineoplásicos/farmacología , Interleucina-2/farmacología , Liposomas , Melanoma Experimental , Nanopartículas , Neoplasias Cutáneas , Microambiente Tumoral/efectos de los fármacos , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/agonistas , Animales , Línea Celular Tumoral , Citocinas/efectos de los fármacos , Citocinas/inmunología , Sistemas de Liberación de Medicamentos , Granzimas/efectos de los fármacos , Granzimas/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Microambiente Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA