Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Nano Lett ; 20(4): 2246-2256, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32160474

RESUMEN

Many favorable anticancer treatments owe their success to the induction immunogenic cell death (ICD) in cancer cells, which results in the release of endogenous danger signals along with tumor antigens for effective priming of anticancer immunity. We describe a strategy to artificially induce ICD by delivering the agonist of stimulator of interferon genes (STING) into tumor cells using hollow polymeric nanoshells. Following intracellular delivery of exogenous adjuvant, subsequent cytotoxic treatment creates immunogenic cellular debris that spatiotemporally coordinate tumor antigens and STING agonist in a process herein termed synthetic immunogenic cell death (sICD). sICD is indiscriminate to the type of chemotherapeutics and enables colocalization of exogenously administered immunologic adjuvants and tumor antigens for enhanced antigen presentation and anticancer adaptive response. In three mouse tumor models, sICD enhances therapeutic efficacy and restrains tumor progression. The study highlights the benefit of delivering STING agonists to cancer cells, paving ways to new chemo-immunotherapeutic designs.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Muerte Celular Inmunogénica/efectos de los fármacos , Proteínas de la Membrana/agonistas , Nanocáscaras/uso terapéutico , Neoplasias/terapia , Animales , Antineoplásicos Inmunológicos/administración & dosificación , Línea Celular Tumoral , Progresión de la Enfermedad , Humanos , Inmunoterapia , Ratones Endogámicos BALB C , Nanocáscaras/administración & dosificación , Neoplasias/inmunología
2.
Adv Funct Mater ; 29(28): 1807616, 2019 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-32313544

RESUMEN

The continued threat of emerging, highly lethal infectious pathogens such as Middle East respiratory syndrome coronavirus (MERS-CoV) calls for the development of novel vaccine technology that offers safe and effective prophylactic measures. Here, a novel nanoparticle vaccine is developed to deliver subunit viral antigens and STING agonists in a virus-like fashion. STING agonists are first encapsulated into capsid-like hollow polymeric nanoparticles, which show multiple favorable attributes, including a pH-responsive release profile, prominent local immune activation, and reduced systemic reactogenicity. Upon subsequent antigen conjugation, the nanoparticles carry morphological semblance to native virions and facilitate codelivery of antigens and STING agonists to draining lymph nodes and immune cells for immune potentiation. Nanoparticle vaccine effectiveness is supported by the elicitation of potent neutralization antibody and antigen-specific T cell responses in mice immunized with a MERS-CoV nanoparticle vaccine candidate. Using a MERS-CoV-permissive transgenic mouse model, it is shown that mice immunized with this nanoparticle-based MERS-CoV vaccine are protected against a lethal challenge of MERS-CoV without triggering undesirable eosinophilic immunopathology. Together, the biocompatible hollow nanoparticle described herein provides an excellent strategy for delivering both subunit vaccine candidates and novel adjuvants, enabling accelerated development of effective and safe vaccines against emerging viral pathogens.

3.
BMC Biotechnol ; 17(1): 2, 2017 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-28061848

RESUMEN

BACKGROUND: Human infection with avian influenza A virus (H7N9) was first reported in China in March 2013. Since then, hundreds of cases have been confirmed showing severe symptoms with a high mortality rate. The virus was transmitted from avian species to humans and has spread to many neighboring areas, raising serious concerns over its pandemic potential. Towards containing the disease, the goal of this study is to prepare a virus-like particle (VLP) that consists of hemagglutinin (HA), neuraminidase (NA) and matrix protein 1 (M1) derived from the human isolate A/Taiwan/S02076/2013(H7N9) for potential vaccine development. RESULTS: Full length HA, NA, and M1 protein genes were cloned and expressed using a baculoviral expression system, and the VLPs were generated by co-infecting insect cells with three respective recombinant baculoviruses. Nanoparticle tracking analysis and transmission electron microscopy were applied to verify the VLPs' structure and antigenicity, and the multiplicity of infection of the recombinant baculoviruses was adjusted to achieve the highest hemagglutination activity. In animal experiments, BALB/c mice and specific-pathogen-free chickens receiving the VLP immunization showed elevated hemagglutination inhibition serum titer and antibodies against NA and M1 proteins. In addition, examination of cellular immunity showed the VLP-immunized mice and chickens exhibited an increased splenic antigen-specific cytokines production. CONCLUSIONS: The H7N9 VLPs possess desirable immunogenicity in vivo and may serve as a candidate for vaccine development against avian influenza A (H7N9) infection.


Asunto(s)
Antígenos Virales/inmunología , Pollos/inmunología , Virus de la Influenza A/inmunología , Ratones/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Animales , Antígenos Virales/genética , Femenino , Virus de la Influenza A/genética , Ratones Endogámicos BALB C , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Especificidad de la Especie , Vacunas de Partículas Similares a Virus/genética
4.
Trends Biotechnol ; 42(2): 241-252, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37743158

RESUMEN

An emerging cellular engineering method creates synthetic polymer matrices inside cells. By contrast with classical genetic, enzymatic, or radioactive techniques, this materials-based approach introduces non-natural polymers inside cells, thus modifying cellular states and functionalities. Here, we cover various materials and chemistries that have been exploited to create intracellular polymer matrices. In addition, we discuss emergent cellular properties due to the intracellular polymerization, including nonreplicating but active metabolism, maintenance of membrane integrity, and resistance to environmental stressors. We also discuss past work and future opportunities for developing and applying synthetic cells that contain intracellular polymers. The materials-based approach will usher in new applications of synthetic cells for broad biotechnological applications.


Asunto(s)
Biotecnología , Polímeros , Polimerizacion , Ingeniería Celular , Materiales Biocompatibles
5.
EMBO Mol Med ; 16(2): 416-428, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38225455

RESUMEN

The tumor microenvironment (TME) presents differential selective pressure (DSP) that favors the growth of cancer cells, and monovalent therapy is often inadequate in reversing the cancer cell dominance in the TME. In this work, we introduce bacteria as a foreign species to the TME and explore combinatorial treatment strategies to alter DSP for tumor eradication. We show that cancer-selective chemotherapeutic agents and fasting can provide a strong selection pressure against tumor growth in the presence of bacteria. Moreover, we show that an immunogenic drug (oxaliplatin), but not a non-immunogenic one (5-FU), synergizes with the bacteria to activate both the innate and adaptive immunity in the TME, resulting in complete tumor remission and a sustained anti-tumor immunological memory in mice. The combination of oxaliplatin and bacteria greatly enhances the co-stimulatory and antigen-presenting molecules on antigen-presenting cells, which in turn bridge the cytotoxic T cells for cancer-cell killing. Our findings indicate that rational combination of bacterial therapy and immunogenic chemotherapy can promote anticancer immunity against the immunosuppressive TME.


Asunto(s)
Antineoplásicos , Neoplasias , Animales , Ratones , Oxaliplatino/uso terapéutico , Microambiente Tumoral , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Linfocitos T Citotóxicos , Inmunoterapia/métodos , Línea Celular Tumoral
6.
Elife ; 122024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38747577

RESUMEN

Certain bacteria demonstrate the ability to target and colonize the tumor microenvironment, a characteristic that positions them as innovative carriers for delivering various therapeutic agents in cancer therapy. Nevertheless, our understanding of how bacteria adapt their physiological condition to the tumor microenvironment remains elusive. In this work, we employed liquid chromatography-tandem mass spectrometry to examine the proteome of E. coli colonized in murine tumors. Compared to E. coli cultivated in the rich medium, we found that E. coli colonized in tumors notably upregulated the processes related to ferric ions, including the enterobactin biosynthesis and iron homeostasis. This finding indicated that the tumor is an iron-deficient environment to E. coli. We also found that the colonization of E. coli in the tumor led to an increased expression of lipocalin 2 (LCN2), a host protein that can sequester the enterobactin. We therefore engineered E. coli in order to evade the nutritional immunity provided by LCN2. By introducing the IroA cluster, the E. coli synthesizes the glycosylated enterobactin, which creates steric hindrance to avoid the LCN2 sequestration. The IroA-E. coli showed enhanced resistance to LCN2 and significantly improved the anti-tumor activity in mice. Moreover, the mice cured by the IroA-E. coli treatment became resistant to the tumor re-challenge, indicating the establishment of immunological memory. Overall, our study underscores the crucial role of bacteria's ability to acquire ferric ions within the tumor microenvironment for effective cancer therapy.


Asunto(s)
Escherichia coli , Hierro , Lipocalina 2 , Animales , Escherichia coli/genética , Escherichia coli/metabolismo , Lipocalina 2/metabolismo , Lipocalina 2/genética , Ratones , Hierro/metabolismo , Neoplasias/terapia , Neoplasias/inmunología , Enterobactina/metabolismo , Microambiente Tumoral , Línea Celular Tumoral
7.
Adv Sci (Weinh) ; : e2310204, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937984

RESUMEN

The development of immune cell engagers (ICEs) can be limited by logistical and functional restrictions associated with fusion protein designs, thus limiting immune cell recruitment to solid tumors. Herein, a high affinity superantigen-based multivalent ICE is developed for simultaneous activation and recruitment of NK and T cells for tumor treatment. Yeast library-based directed evolution is adopted to identify superantigen variants possessing enhanced binding affinity to immunoreceptors expressed on human T cells and NK cells. High-affinity superantigens exhibiting improved immune-stimulatory activities are then incorporated into a superantigen-based tri-functional yeast-display-enhanced multivalent immune cell engager (STYMIE), which is functionalized with a nanobody, a Neo-2/15 cytokine, and an Fc domain for tumor targeting, immune stimulation, and prolonged circulation, respectively. Intravenous administration of STYMIE enhances NK and T cell recruitment into solid tumors, leading to enhanced inhibition in multiple tumor models. The study offers design principles for multifunctional ICEs.

8.
Adv Healthc Mater ; 12(8): e2201708, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36455286

RESUMEN

The intricate functionalities of cellular membranes have inspired strategies for deriving and anchoring cell-surface components onto solid substrates for biological studies, biosensor applications, and tissue engineering. However, introducing conformal and right-side-out cell membrane coverage onto planar substrates requires cumbersome protocols susceptible to significant device-to-device variability. Here, a facile approach for biomembrane functionalization of planar substrates is demonstrated by subjecting confluent cellular monolayer to intracellular hydrogel polymerization. The resulting cell-gel hybrid, herein termed GELL (gelated cell), exhibits extraordinary stability and retains the structural integrity, membrane fluidity, membrane protein mobility, and topology of living cells. In assessing the utility of GELL layers as a tissue engineering feeder substrate for stem cell maintenance, GELL feeder prepared from primary mouse embryonic fibroblasts not only preserves the stemness of murine stem cells but also exhibits advantages over live feeder cells owing to the GELL's inanimate, non-metabolizing nature. The preparation of a xeno-free feeder substrate devoid of non-human components is further shown with HeLa cells, and the resulting  HeLa GELL feeder effectively sustains the growth and stemness of both murine and human induced pluripotent stem cells. The study highlights a novel bio-functionalization strategy that introduces new opportunities for tissue engineering and other biomedical applications.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Humanos , Animales , Ratones , Fibroblastos , Células HeLa , Células Nutrientes/metabolismo , Diferenciación Celular
9.
Adv Sci (Weinh) ; 9(13): e2105506, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35246961

RESUMEN

Membrane-lytic peptides offer broad synthetic flexibilities and design potential to the arsenal of anticancer therapeutics, which can be limited by cytotoxicity to noncancerous cells and induction of drug resistance via stress-induced mutagenesis. Despite continued research efforts on membrane-perforating peptides for antimicrobial applications, success in anticancer peptide therapeutics remains elusive given the muted distinction between cancerous and normal cell membranes and the challenge of peptide degradation and neutralization upon intravenous delivery. Using triple-negative breast cancer as a model, the authors report the development of a new class of anticancer peptides. Through function-conserving mutations, the authors achieved cancer cell selective membrane perforation, with leads exhibiting a 200-fold selectivity over non-cancerogenic cells and superior cytotoxicity over doxorubicin against breast cancer tumorspheres. Upon continuous exposure to the anticancer peptides at growth-arresting concentrations, cancer cells do not exhibit resistance phenotype, frequently observed under chemotherapeutic treatment. The authors further demonstrate efficient encapsulation of the anticancer peptides in 20 nm polymeric nanocarriers, which possess high tolerability and lead to effective tumor growth inhibition in a mouse model of MDA-MB-231 triple-negative breast cancer. This work demonstrates a multidisciplinary approach for enabling translationally relevant membrane-lytic peptides in oncology, opening up a vast chemical repertoire to the arms race against cancer.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama Triple Negativas , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Humanos , Ratones , Péptidos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo
10.
Mol Pharm ; 8(4): 1401-7, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21696189

RESUMEN

We report a novel approach for nanoparticle-based combination chemotherapy by concurrently incorporating two different types of drugs into a single polymeric nanoparticle with ratiometric control over the loading of the two drugs. By adapting metal alkoxide chemistry, we synthesize highly hydrophobic drug-poly-L-lactide (drug-PLA) conjugates, of which the polymer has the same chain length while the drug may differ. These drug-polymer conjugates are then encapsulated into lipid-coated polymeric nanoparticles through a single-step nanoprecipitation method. Using doxorubicin (DOX) and camptothecin (CPT) as two model chemotherapy drugs, various ratios of DOX-PLA and CPT-PLA conjugates are loaded into the nanoparticles with over 90% loading efficiency. The resulting nanoparticles are uniform in size, size distribution and surface charge. The loading yield of DOX and CPT in the particles can be precisely controlled by simply adjusting the DOX-PLA:CPT-PLA molar ratio. Cellular cytotoxicity results show that the dual-drug loaded nanoparticles are superior to the corresponding cocktail mixtures of single-drug loaded nanoparticles. This dual-drug delivery approach offers a solution to the long-standing challenge in ratiometric control over the loading of different types of drugs onto the same drug delivery vehicle. We expect that this approach can be exploited for many types of chemotherapeutic agents containing hydroxyl groups and thus enable codelivery of various drug combinations for combinatorial treatments of diseases.


Asunto(s)
Nanopartículas/química , Polímeros/química , Camptotecina/química , Doxorrubicina/química , Portadores de Fármacos/química , Poliésteres/química
11.
Drug Deliv Transl Res ; 11(4): 1420-1437, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33748879

RESUMEN

The COVID-19 pandemic's high mortality rate and severe socioeconomic impact serve as a reminder of the urgent need for effective countermeasures against viral pandemic threats. In particular, effective antiviral therapeutics capable of stopping infections in its tracks is critical to reducing infection fatality rate and healthcare burden. With the field of drug delivery witnessing tremendous advancement in the last two decades owing to a panoply of nanotechnology advances, the present review summarizes and expounds on the research and development of therapeutic nanoformulations against various infectious viral pathogens, including HIV, influenza, and coronaviruses. Specifically, nanotechnology advances towards improving pathogen- and host-targeted antiviral drug delivery are reviewed, and the prospect of achieving effective viral eradication, broad-spectrum antiviral effect, and resisting viral mutations are discussed. As several COVID-19 antiviral clinical trials are met with lackluster treatment efficacy, nanocarrier strategies aimed at improving drug pharmacokinetics, biodistributions, and synergism are expected to not only contribute to the current disease treatment efforts but also expand the antiviral arsenal against other emerging viral diseases.


Asunto(s)
Antivirales/administración & dosificación , COVID-19/prevención & control , Sistemas de Liberación de Medicamentos/métodos , Interacciones Huésped-Patógeno/efectos de los fármacos , Nanopartículas/administración & dosificación , Nanotecnología/métodos , Animales , Antivirales/inmunología , COVID-19/epidemiología , COVID-19/inmunología , Sistemas de Liberación de Medicamentos/tendencias , Interacciones Huésped-Patógeno/inmunología , Humanos , Nanotecnología/tendencias , Pandemias/prevención & control , Virosis/epidemiología , Virosis/inmunología , Virosis/prevención & control , Replicación Viral/efectos de los fármacos , Replicación Viral/fisiología
12.
Hum Vaccin Immunother ; 17(3): 654-655, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-32991231

RESUMEN

A safe and effective vaccine candidate is urgently needed for the ongoing COVID-19 pandemic, caused by SARS-CoV-2. Here we report that recombinant SARS-CoV-2 RBD protein immunization in mice is able to elicit a strong antibody response and potent neutralizing capability as measured using live or pseudotyped SARS-CoV-2 neutralization assays.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/inmunología , Unión Proteica/inmunología , Dominios Proteicos/inmunología , SARS-CoV-2/inmunología , Animales , Línea Celular , Células HEK293 , Humanos , Ratones , Pandemias/prevención & control , Proteínas Recombinantes/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología
13.
Mol Ther Methods Clin Dev ; 21: 299-314, 2021 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-33898629

RESUMEN

Antigen-specific lung-resident memory T cells (TRMs) constitute the first line of defense that mediates rapid protection against respiratory pathogens and inspires novel vaccine designs against infectious pandemic threats, yet effective means of inducing TRMs, particularly via non-viral vectors, remain challenging. Here, we demonstrate safe and potent induction of lung-resident TRMs using a biodegradable polymeric nanoshell that co-encapsulates antigenic peptides and TLR9 agonist CpG-oligodeoxynucleotide (CpG-ODN) in a virus-mimicking structure. Through subcutaneous priming and intranasal boosting, the combinatorial nanoshell vaccine elicits prominent lung-resident CD4+ and CD8+ T cells that surprisingly show better durability than live viral infections. In particular, nanoshells containing CpG-ODN and a pair of conserved class I and II major histocompatibility complex-restricted influenza nucleoprotein-derived antigenic peptides are demonstrated to induce near-sterilizing immunity against lethal infections with influenza A viruses of different strains and subtypes in mice, resulting in rapid elimination of replicating viruses. We further examine the pulmonary transport dynamic and optimal composition of the nanoshell vaccine conducive for robust TRM induction as well as the benefit of subcutaneous priming on TRM replenishment. The study presents a practical vaccination strategy for inducing protective TRM-mediated immunity, offering a compelling platform and critical insights in the ongoing quest toward a broadly protective vaccine against universal influenza as well as other respiratory pathogens.

14.
Small ; 6(13): 1442-8, 2010 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-20564488

RESUMEN

A new approach to loading multiple drugs onto the same drug-delivery nanocarrier in a precisely controllable manner, by covalently preconjugating multiple therapeutic agents through hydrolyzable linkers to form drug conjugates, is reported. In contrast to loading individual types of drugs separately, this drug-conjugates strategy enables the loading of multiple drugs onto the same carrier with a predefined stoichiometric ratio. The cleavable linkers allow the therapeutic activity of the individual drugs to be resumed after the drug conjugates are delivered into the target cells and unloaded from the delivery vehicle. As a proof of concept, the synthesis and characterization of paclitaxel-gemcitabine conjugates are demonstrated. The time-dependent hydrolysis kinetics and cytotoxicity of the combinatorial drug conjugates against human pancreatic cancer cells are examined. It is shown that the synthesized drug conjugates can be readily encapsulated into a lipid-coated polymeric drug-delivery nanoparticle, which significantly improves the cytotoxicity of the drug conjugates as compared to the free drug conjugates.


Asunto(s)
Desoxicitidina/análogos & derivados , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/química , Paclitaxel/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Desoxicitidina/síntesis química , Desoxicitidina/química , Desoxicitidina/farmacología , Humanos , Hidrólisis/efectos de los fármacos , Lípidos/química , Espectroscopía de Resonancia Magnética , Paclitaxel/síntesis química , Paclitaxel/química , Gemcitabina
15.
Mol Pharm ; 7(3): 914-20, 2010 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-20394436

RESUMEN

Current chemotherapy regimens against pancreatic cancer are met with little success as poor tumor vascularization significantly limits the delivery of oncological drugs. High-dose targeted drug delivery, through which a drug delivery vehicle releases a large payload upon tumor localization, is thus a promising alternative strategy against this lethal disease. Herein, we synthesize anti-carcinoembryonic antigen (CEA) half-antibody conjugated lipid-polymer hybrid nanoparticles and characterize their ligand conjugation yields, physicochemical properties, and targeting ability against pancreatic cancer cells. Under the same drug loading, the half-antibody targeted nanoparticles show enhanced cancer killing effect compared to the corresponding nontargeted nanoparticles.


Asunto(s)
Antígeno Carcinoembrionario/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Lípidos/química , Nanopartículas/química , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Polímeros/química , Línea Celular , Cromatografía Líquida de Alta Presión , Humanos , Paclitaxel/administración & dosificación , Paclitaxel/química , Paclitaxel/uso terapéutico
16.
Langmuir ; 26(22): 16958-62, 2010 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-20961057

RESUMEN

Lipid-polymer hybrid nanoparticle, consisting of a hydrophobic polymeric core and a lipid monolayer shell, represents a new and promising drug delivery platform that has shown controllable particle size and surface functionality, high drug loading yield, sustained drug release profile, and excellent in vitro and in vivo stability. These lipid monolayer-coated polymeric nanoparticles are typically fabricated through a modified nanoprecipitation method, which involves sample heating, vortexing, and solvent evaporation. Herein we report a new and fast method to synthesize lipid-polymer hybrid nanoparticles with controllable and nearly uniform particle size. Using a bath sonication approach, we demonstrate that the whole hybrid nanoparticle synthesis process can be completed in about 5 min compared with a few hours for previous synthesis approaches. The size and polydispersity of the resulting nanoparticles can be readily controlled by tuning the relative concentrations of individual building components. Colloidal stability tests of the synthesized hybrid nanoparticles in PBS buffer and serum show no signs of aggregation over a period of 5 days. The present method improves the production rate of the hybrid nanoparticles by near 20-fold while not compromising the physicochemical properties of the particles. This work may facilitate the bench-to-bedside translation of lipid-polymer hybrid nanoparticles as a robust drug nanocarrier by allowing for fabricating a large amount of these nanoparticles at high production rate.


Asunto(s)
Lípidos/química , Lípidos/síntesis química , Nanopartículas/química , Polímeros/química , Sonicación/métodos , Animales , Tampones (Química) , Bovinos , Calor , Cinética , Lípidos/sangre , Tamaño de la Partícula , Polietilenglicoles/química , Solventes/química , Volatilización
17.
Nanomedicine (Lond) ; 15(29): 2883-2894, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33252301

RESUMEN

The discovery of stimulator of interferon genes (STING) and their agonists as primary components that link antiviral innate and adaptive immunity has motivated growing research on STING agonist-mediated immunotherapy and vaccine development. To overcome the delivery challenge in shuttling highly polar STING agonists, typically in the form of cyclic dinucleotides, to target cells and to STING proteins in cellular cytosol, numerous nanoformulation strategies have been implemented for effective STING activation. While many STING-activating nanoparticles are developed to enhance anticancer immunotherapy, their adoption as vaccine adjuvant has vastly propelled antiviral vaccination efforts against challenging public health threats, including HIV, influenza and coronaviruses. In light of the COVID-19 pandemic that has thrusted vaccine development into the public spotlight, this review highlights advances in nanomedicinal STING agonist delivery with an emphasis on their applications in antiviral vaccination.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Vacunas contra la COVID-19/uso terapéutico , Inmunidad Innata/efectos de los fármacos , Pandemias , Antivirales/uso terapéutico , COVID-19/patología , COVID-19/virología , Vacunas contra la COVID-19/inmunología , Humanos , Inmunoterapia/tendencias , Nanopartículas/química , Nanopartículas/uso terapéutico , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Transducción de Señal/efectos de los fármacos
18.
Int J Nanomedicine ; 15: 3303-3318, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32494131

RESUMEN

BACKGROUND: Poultry vaccine has limited choices of adjuvants and is facing severe threat of infectious diseases due to ineffective of widely used commercial vaccines. Thus, development of novel adjuvant that offers safe and effective immunity is of urgent need. MATERIALS AND METHODS: The present research engineers a novel chicken adjuvant with potent immune-potentiating capability by incorporating avian toll-like receptor 21 (TLR21) agonist CpG ODN 2007 with a poly(lactic-co-glycolic acid) (PLGA)-based hollow nanoparticle platform (CpG-NP), which subsequently assessed ex vivo and in vivo. RESULTS: CpG-NPs with an average diameter of 164 nm capable of sustained release of CpG for up to 96 hours were successfully prepared. With the ex vivo model of chicken bone marrow-derived dendritic cells (chBMDCs), CpG-NP was engulfed effectively and found to induce DC maturation, promoting dendrite formation and upregulation of CD40, CD80 and CCR7. In addition to enhanced expression of IL-1ß, IL-6, IL-12 and IFN-γ, 53/84 immune-related genes were found to be stimulated in CpG-NP-treated chBMDCs, whereas only 39 of such genes were stimulated in free CpG-treated cells. These upregulated genes suggest immune skewing toward T helper cell 1 bias and evidence of improved mucosal immunity upon vaccination with the CpG-NP. The CpG-NP-treated chBMDCs showed protective effects to DF-1 cells against avian influenza virus H6N1 infection. Upon subsequent coupling with infectious bronchitis virus subunit antigen administration, chickens were immunostimulated to acquire higher humoral immune response and protective response against viral challenge. CONCLUSTION: This work presents a novel hollow CpG-NP formulation, demonstrating effective and long-lasting immunostimulatory ability ex vivo and in vivo for chickens, as systemically compared to free CpG. This enhanced immune stimulation benefits from high stability and controlled release of internal component of nanoparticles that improve cellular delivery, lymphoid organ targeting and sustainable DC activation. CpG-NP has broad application potential in antiviral and vaccine development.


Asunto(s)
Antivirales/farmacología , Pollos/inmunología , Inmunidad/efectos de los fármacos , Nanopartículas/química , Oligodesoxirribonucleótidos/farmacología , Polímeros/química , Vacunas/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Animales , Supervivencia Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Perros , Inmunidad Humoral/efectos de los fármacos , Inmunización , Virus de la Bronquitis Infecciosa/efectos de los fármacos , Células de Riñón Canino Madin Darby , Nanopartículas/administración & dosificación , Nanopartículas/ultraestructura , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química
19.
Adv Healthc Mater ; 7(13): e1701395, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29508547

RESUMEN

As the dawn of the postantibiotic era we approach, antibacterial vaccines are becoming increasingly important for managing bacterial infection and reducing the need for antibiotics. Despite the success of vaccination, vaccines remain unavailable for many pressing microbial diseases, including tuberculosis, chlamydia, and staphylococcus infections. Amid continuing research efforts in antibacterial vaccine development, the advancement of nanomaterial engineering has brought forth new opportunities in vaccine designs. With increasing knowledge in antibacterial immunity and immunologic adjuvants, innovative nanoparticles are designed to elicit the appropriate immune responses for effective antimicrobial defense. Rationally designed nanoparticles are demonstrated to overcome delivery barriers to shape the adaptive immunity. This article reviews the advances in nanoparticle- and nanomaterial-based antibacterial vaccines and summarizes the development of nanoparticulate adjuvants for immune potentiation against microbial pathogens. In addition, challenges and progress in ongoing antibacterial vaccine development are discussed to highlight the opportunities for future vaccine designs.


Asunto(s)
Infecciones Bacterianas/prevención & control , Vacunas Bacterianas/farmacología , Nanoestructuras/química , Receptores de Reconocimiento de Patrones/agonistas , Vacunas Bacterianas/inmunología , Humanos , Nanopartículas/química , Receptores de Reconocimiento de Patrones/inmunología
20.
Int J Nanomedicine ; 13: 8579-8593, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30587980

RESUMEN

BACKGROUND: Influenza virus infections are a major public health concern worldwide. Conventional treatments against the disease are designed to target viral proteins. However, the emergence of viral variants carrying drug-resistant mutations can outpace the development of pathogen-targeting antivirals. Diphyllin and bafilomycin are potent vacuolar ATPase (V-ATPase) inhibitors previously shown to have broad-spectrum antiviral activity. However, their poor water solubility and potential off-target effect limit their clinical application. METHODS: In this study, we report that nanoparticle encapsulation of diphyllin and bafilomycin improves the drugs' anti-influenza applicability. RESULTS: Using PEG-PLGA diblock copolymers, sub-200 nm diphyllin and bafilomycin nanoparticles were prepared, with encapsulation efficiency of 42% and 100%, respectively. The drug-loaded nanoparticles have sustained drug release kinetics beyond 72 hours and facilitate intracellular drug delivery to two different influenza virus-permissive cell lines. As compared to free drugs, the nanoparticulate V-ATPase inhibitors exhibited lower cytotoxicity and greater in vitro antiviral activity, improving the therapeutic index of diphyllin and bafilomycin by approximately 3 and 5-fold, respectively. In a mouse model of sublethal influenza challenge, treatment with diphyllin nanoparticles resulted in reduced body weight loss and viral titer in the lungs. In addition, following a lethal influenza viral challenge, diphyllin nanoparticle treatment conferred a survival advantage of 33%. CONCLUSIONS: These results demonstrate the potential of the nanoparticulate V-ATPase inhibitors for host-targeted treatment against influenza.


Asunto(s)
Antivirales/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Gripe Humana/tratamiento farmacológico , Nanopartículas/química , ATPasas de Translocación de Protón Vacuolares/antagonistas & inhibidores , Animales , Antivirales/química , Antivirales/farmacología , Benzodioxoles/química , Benzodioxoles/farmacología , Benzodioxoles/uso terapéutico , Línea Celular , Perros , Liberación de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Gripe Humana/virología , Concentración 50 Inhibidora , Cinética , Lignanos/química , Lignanos/farmacología , Lignanos/uso terapéutico , Macrólidos/química , Macrólidos/farmacología , Macrólidos/uso terapéutico , Ratones , Nanopartículas/ultraestructura , Orthomyxoviridae/efectos de los fármacos , Orthomyxoviridae/fisiología , Infecciones por Orthomyxoviridae/tratamiento farmacológico , Infecciones por Orthomyxoviridae/virología , ATPasas de Translocación de Protón Vacuolares/metabolismo , Proteínas Virales/metabolismo , Replicación Viral/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA