Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Pharm Res ; 41(2): 305-319, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38332390

RESUMEN

PURPOSE: A dissolving microneedle array (dMNA) is a vaccine delivery device with several advantages over conventional needles. By incorporating particulate adjuvants in the form of poly(D,L-lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) into the dMNA, the immune response against the antigen might be enhanced. This study aimed to prepare PLGA-NP-loaded dMNA and to compare T-cell responses induced by either intradermally injected aqueous-PLGA-NP formulation or PLGA-NP-loaded dMNA in mice. METHODS: PLGA NPs were prepared with microfluidics, and their physicochemical characteristics with regard to encapsulation efficiencies of ovalbumin (OVA) and CpG oligonucleotide (CpG), zeta potentials, polydispersity indexes, and sizes were analysed. PLGA NPs incorporated dMNA was produced with three different dMNA formulations by using the centrifugation method, and the integrity of PLGA NPs in dMNAs was evaluated. The immunogenicity was evaluated in mice by comparing the T-cell responses induced by dMNA and aqueous formulations containing ovalbumin and CpG (OVA/CpG) with and without PLGA NP. RESULTS: Prepared PLGA NPs had a size of around 100 nm. The dMNA formulations affected the particle integrity, and the dMNA with poly(vinyl alcohol) (PVA) showed almost no aggregation of PLGA NPs. The PLGA:PVA weight ratio of 1:9 resulted in 100% of penetration efficiency and the fastest dissolution in ex-vivo human skin (< 30 min). The aqueous formulation with soluble OVA/CpG and the aqueous-PLGA-NP formulation with OVA/CpG induced the highest CD4 + T-cell responses in blood and spleen cells. CONCLUSIONS: PLGA NPs incorporated dMNA was successfully fabricated and the aqueous formulation containing PLGA NPs induce superior CD4+ and CD8+ T-cell responses.


Asunto(s)
Nanopartículas , Vacunación , Ratones , Humanos , Animales , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Ovalbúmina , Vacunación/métodos , Antígenos , Ácido Láctico
2.
J Immunother Cancer ; 11(11)2023 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-38030302

RESUMEN

BACKGROUND: The survival of patients with cervical cancer who are treated with cisplatin in conjunction with the topoisomerase I inhibitor topotecan is enhanced when compared with patients treated with only one of these chemotherapeutics. Moreover, cisplatin-based and T cell-based immunotherapy have been shown to synergize, resulting in stronger antitumor responses. Here, we interrogated whether topotecan could further enhance the synergy of cisplatin with T cell-based cancer immunotherapy. METHODS: Mice bearing human papilloma virus 16 (HPV16) E6/E7-expressing TC-1 tumors were vaccinated with HPV16 E7 long peptides and additionally received chemotherapy consisting of cisplatin and topotecan. We performed an in-depth study of this combinatorial chemoimmunotherapy on the effector function and expansion/contraction kinetics of vaccine-induced CD8+ T cells in the peripheral blood and tumor microenvironment (TME). In addition, we interrogated the particular role of chemotherapy-induced upregulation of costimulatory ligands by tumor-infiltrated myeloid cells on T cell proliferation and survival. RESULTS: We show that E7 long peptide vaccination combined with cisplatin and topotecan, results in CD8+ T cell-dependent durable rejection of established tumors and 94% long-term survival. Although topotecan initially repressed the expansion of vaccine-induced CD8+ T cells, these cells eventually expanded vigorously, which was followed by delayed contraction. These effects associated with the induction of the proliferation marker Ki-67 and the antiapoptosis molecule Bcl-2 by intratumoral tumor-specific CD8+ T cells, which was regulated by topotecan-mediated upregulation of the costimulatory ligand CD70 on myeloid cells in the TME. CONCLUSIONS: Taken together, our data show that although treatment with cisplatin, topotecan and vaccination initially delays T cell expansion, this combinatorial therapy results eventually in a more robust T cell-mediated tumor eradication due to enhancement of costimulatory molecules in the TME.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias del Cuello Uterino , Femenino , Humanos , Animales , Ratones , Cisplatino/farmacología , Cisplatino/uso terapéutico , Linfocitos T CD8-positivos , Vacunas contra el Cáncer/uso terapéutico , Topotecan/farmacología , Topotecan/uso terapéutico , ADN-Topoisomerasas de Tipo I , Proteínas E7 de Papillomavirus , Vacunas de Subunidad , Neoplasias del Cuello Uterino/tratamiento farmacológico , Proliferación Celular , Microambiente Tumoral , Ligando CD27
3.
Biomater Sci ; 11(20): 6790-6800, 2023 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-37622228

RESUMEN

Dissolving microneedle arrays (dMNAs) can be used to deliver vaccines via the intradermal route. Fabrication of dMNAs using centrifugation is the most common preparation method of dMNAs, but it results in a substantial loss of antigens. In order to solve the issue of antigen waste, we engineered an automatic dispensing system for dMNA preparation. Here, we report on the fabrication of influenza whole inactivated virus (WIV) vaccine-loaded dMNAs (WIV dMNAs) by using the automatic dispensing system. Prior to the dispensing process, polydimethylsiloxane (PDMS) moulds were treated with oxygen plasma to increase surface hydrophilicity. WIV dMNAs were prepared with 1% (w/v) trehalose and pullulan (50 : 50 weight ratio). During the dispensing process, reduced pressure was applied to the PDMS mould via a vacuum chamber to make microneedle cavities airless. After producing dMNAs, WIV was quantified and 1.9 µg of WIV was loaded per dMNA, of which 1.3 µg was in the microneedle tips. Compared to the centrifugation method, this automatic dispensing system resulted in a 95% reduction of antigen waste. A hemagglutination assay confirmed that WIV dMNA maintained the stability of the antigen for at least four weeks of storage, even at room temperature or at 37 °C. The WIV dMNAs displayed 100% penetration efficiency in human skin, and 83% of the microneedle volume was dissolved in the skin within 10 minutes. In a vaccination study, mice immunised with WIV dMNAs showed similar IgG levels to those that received WIV intramuscularly. In conclusion, using the automatic dispensing system for dMNA production strongly reduced antigen waste and yielded dMNAs with excellent physical, mechanical, and immunological properties.

4.
J Pharm Sci ; 111(4): 1040-1049, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35101553

RESUMEN

Antigenic peptide-loaded cationic liposomes have shown promise as cancer vaccines. Quantification of both peptides and lipids is critical for quality control of such vaccines for clinical translation. In this work we describe a reversed phase ultra-performance liquid chromatography (RP-UPLC) method that separates lipids (DOTAP, DOPC and their degradation products) and two physicochemically different peptides within 12 min. Samples were prepared by dilution in a 1:1 (v/v) mixture of methanol and water. Peptide quantification was done via UV detection and lipids were quantified by an evaporative light scattering detector (ELSD), both coupled to the RP-UPLC system, with high precision (RSD < 3.5%). We showed that the presence of lipids and peptides did not mutually influence their quantification. Limit of detection (LOD) and limit of quantification (LOQ), as determined in the ICH guidelines, were 6 and 20 ng for DOTAP, 12 ng and 40 ng for DOPC, 3.0 ng and 8.0 ng for peptide A and 2.4 ng and 7.2 ng for the more hydrophobic peptide B. Finally, lipid degradation of DOTAP and DOPC was monitored in peptide loaded DOTAP:DOPC liposomes upon storage at 4 °C and 40 °C.


Asunto(s)
Cromatografía de Fase Inversa , Liposomas , Cationes , Cromatografía Líquida de Alta Presión/métodos , Luz , Lipólisis , Liposomas/química , Péptidos , Dispersión de Radiación
5.
Pharmaceutics ; 13(5)2021 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-33919378

RESUMEN

Cationic nanoparticles have been shown to be surprisingly effective as cancer vaccine vehicles in preclinical and clinical studies. Cationic nanoparticles deliver tumor-associated antigens to dendritic cells and induce immune activation, resulting in strong antigen-specific cellular immune responses, as shown for a wide variety of vaccine candidates. In this review, we discuss the relation between the cationic nature of nanoparticles and the efficacy of cancer immunotherapy. Multiple types of lipid- and polymer-based cationic nanoparticulate cancer vaccines with various antigen types (e.g., mRNA, DNA, peptides and proteins) and adjuvants are described. Furthermore, we focus on the types of cationic nanoparticles used for T-cell induction, especially in the context of therapeutic cancer vaccination. We discuss different cationic nanoparticulate vaccines, molecular mechanisms of adjuvanticity and biodistribution profiles upon administration via different routes. Finally, we discuss the perspectives of cationic nanoparticulate vaccines for improving immunotherapy of cancer.

6.
Int J Pharm ; 600: 120473, 2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-33737094

RESUMEN

Dissolving microneedle arrays (dMNAs) are promising devices for intradermal vaccine delivery. The aim of this study was to develop a reproducible fabrication method for dMNAs based on an automated nano-droplet dispensing system that minimizes antigen waste. First, a polymer formulation was selected to dispense sufficiently small droplets (<18 nL) that can enter the microneedle cavities (base diameter 330 µm). Besides, three linear stages were assembled to align the dispenser with the cavities, and a vacuum chamber was designed to fill the cavities with dispensed droplets without entrapped air. Lastly, the dispenser and stages were incorporated to build a fully automated system. To examine the function of dMNAs as a vaccine carrier, ovalbumin was loaded in dMNAs by dispensing a mixture of ovalbumin and polymer formulation, followed by determining the ovalbumin loading and release into the skin. The results demonstrate that functional dMNAs which can deliver antigen into the skin were successfully fabricated via the automatic fabrication system, and hardly any antigen waste was encountered. Compared to the method that centrifuges the mould, it resulted in a 98.5% volume reduction of antigen/polymer solution and a day shorter production time. This system has potential for scale-up of manufacturing to an industrial scale.


Asunto(s)
Sistemas de Liberación de Medicamentos , Vacunas , Administración Cutánea , Antígenos , Microinyecciones , Agujas
7.
Br J Clin Pharmacol ; 87(8): 3162-3176, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33403697

RESUMEN

AIMS: To evaluate feasibility of intradermal (i.d.) adalimumab administration using hollow microneedles, and to compare a single i.d. dose of adalimumab using a hollow microneedle with a single subcutaneous (s.c.) dose using a conventional needle. METHODS: In this single-centre double-blind, placebo-controlled, double-dummy clinical trial in 24 healthy adults we compared 40 mg adalimumab (0.4 mL) administered i.d. using a hollow microneedle with a s.c. dose using a conventional needle. Primary parameters were pain, acceptability and local tolerability; secondary parameters safety, pharmacokinetics and immunogenicity. We explored usability of optical coherence tomography, clinical photography, thermal imaging, and laser speckle contrast imaging to evaluate skin reaction after i.d. injections. In vitro protein analysis was performed to assess compatibility of adalimumab with the hollow microneedle device. RESULTS: While feasible and safe, injection pain of i.d. adalimumab was higher compared to s.c. adalimumab (35.4 vs. 7.9 on a 100-point visual analogue scale). Initial absorption rate and relative bioavailability were higher after i.d. adalimumab (time to maximum plasma concentration = 95 h [47-120]; Frel = 129% [6.46%]) compared to s.c. adalimumab (time to maximum plasma concentration = 120 h [96-221]). Anti-adalimumab antibodies were detected in 50% and 83% of the subjects after i.d. and s.c. adalimumab, respectively. We observed statistically significantly more erythema and skin perfusion after i.d. adalimumab, compared to s.c. adalimumab and placebo injections (P < .0001). Cytokine secretion after whole blood lipopolysaccharide challenge was comparable between administration routes. CONCLUSIONS: Intradermal injection of adalimumab using hollowing microneedles was perceived as more painful and less accepted than s.c. administration, but yields a higher relative bioavailability with similar safety and pharmacodynamic effects.


Asunto(s)
Agujas , Piel , Adalimumab , Adulto , Humanos , Inyecciones Intradérmicas , Inyecciones Subcutáneas , Dimensión del Dolor
8.
Front Allergy ; 2: 642788, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35386985

RESUMEN

The skin is an attractive alternative administration route for allergy vaccination, as the skin is rich in dendritic cells (DCs) and is easily accessible. In the skin multiple subsets of DCs with distinct roles reside at different depths. In this study antigen (=allergen for allergy) formulations were injected in ex vivo human skin in a depth-controlled manner by using a hollow microneedle injection system. Biopsies were harvested at the injection site, which were then cultured for 72 h. Subsequently, the crawled-out cells were collected from the medium and analyzed with flow cytometry. Intradermal administration of ovalbumin (OVA, model antigen) solution at various depths in the skin did not affect the migration and maturation of DCs. OVA was taken up efficiently by the DCs, and this was not affected by the injection depth. In contrast, Bet v 1, the major allergen in birch pollen allergy, was barely taken up by dermal DCs (dDCs). Antigens were more efficiently taken up by CD14+ dDCs than CD1a+ dDCs, which in turn were more efficient at taken up antigen than Langerhans cells. Subsequently, both OVA and Bet v 1 were formulated in cationic and anionic liposomes, which altered antigen uptake drastically following intradermal microinjection. While OVA uptake was reduced by formulation in liposomes, Bet v 1 uptake in dDCs was increased by encapsulation in both cationic and anionic liposomes. This highlights the potential use of liposomes as adjuvant in intradermal allergy vaccine delivery. In conclusion, we observed that antigen uptake after intradermal injection was not affected by injection depth, but varied between different antigens and formulation.

9.
Pharmaceutics ; 12(11)2020 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-33158166

RESUMEN

Cancer immunotherapy has shown remarkable progress in recent years. Nanocarriers, such as liposomes, have favorable advantages with the potential to further improve cancer immunotherapy and even stronger immune responses by improving cell type-specific delivery and enhancing drug efficacy. Liposomes can offer solutions to common problems faced by several cancer immunotherapies, including the following: (1) Vaccination: Liposomes can improve the delivery of antigens and other stimulatory molecules to antigen-presenting cells or T cells; (2) Tumor normalization: Liposomes can deliver drugs selectively to the tumor microenvironment to overcome the immune-suppressive state; (3) Rewiring of tumor signaling: Liposomes can be used for the delivery of specific drugs to specific cell types to correct or modulate pathways to facilitate better anti-tumor immune responses; (4) Combinational therapy: Liposomes are ideal vehicles for the simultaneous delivery of drugs to be combined with other therapies, including chemotherapy, radiotherapy, and phototherapy. In this review, different liposomal systems specifically developed for immunomodulation in cancer are summarized and discussed.

10.
Eur J Pharm Sci ; 150: 105331, 2020 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-32470845

RESUMEN

Conventional transdermal drug patches have been on the market since 1997 but their applicability for drug delivery is limited: currently only nearly two dozen of molecules have been approved by the regulatory authorities for transdermal administration and have reached the market. The possibilities for drug delivery via the skin can be improved and expanded by using microneedle patch technologies. However, most microneedle patches focus on the delivery of low amounts of drugs that are generally very potent due to the small dimensions of the microneedle systems. In this study nanoporous microneedle arrays (npMNAs) were combined with a liquid drug reservoir. The parameters that influence the diffusion of memantine from the drug reservoir through the npMNAs in an acceptor solution were investigated. Based on these results a model was developed to predict the diffusion of low-molecular-weight drugs as a function of npMNA properties (i.e., backplate thickness and surface area) and reservoir properties (i.e., volume and drug concentration). This generated an in silico model to predict the release of low-molecular-weight drug from a drug reservoir through a microneedle array into receptor solution, showed a good correlation with the delivery of memantine in a preclinical minipig study. The drug release rates by the npMNAs can be tuned and allow for both zero and first order release kinetics. Summarizing, this work shows that the npMNA technology is a versatile drug delivery system. The npMNAs can be combined with a (seamlessly connected) external drug reservoir and this integrated drug delivery system can be used to deliver at least 9 mg of memantine over 72 h in a preclinical minipig study.


Asunto(s)
Sistemas de Liberación de Medicamentos , Memantina/administración & dosificación , Microinyecciones , Agujas , Fármacos Neuroprotectores/administración & dosificación , Administración Cutánea , Animales , Memantina/sangre , Memantina/farmacocinética , Nanoporos , Fármacos Neuroprotectores/sangre , Fármacos Neuroprotectores/farmacocinética , Porosidad , Porcinos , Porcinos Enanos
11.
J Control Release ; 323: 1-11, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32247805

RESUMEN

Thermosensitive poloxamer 407 (P407) hydrogels were evaluated as slow release system for optimizing CTLA-4 therapy. Slow release reduces systemic antibody levels and potentially mitigates the side effects of CTLA-4 therapy. The 25% P407 hydrogel is injectable at room temperature and depots are established quickly after subcutaneous injection. Scanning electron microscopy revealed the porous structure of the hydrogel, average pore surface was 1335 µm2. Release studies were optimized using the human IgG antibody. IgG was easily incorporated in the hydrogel by simple mixing and no antibodies were lost during preparation. In vitro, hydrogels showed low burst release within the first 24 h. Total IgG load was gradually released within 120 h. In vitro cytotoxicity assays showed that P407 is not cytotoxic and induces no immune activation by itself. In vivo, P407 hydrogels significantly reduced serum IgG levels, were biocompatible and were broken down 1 week after injection. Finally, local hydrogel delivery of anti-CTLA-4 antibodies near established tumors effectively slowed down tumor growth, whilst significantly reduced serum anti-CTLA-4 levels. Altogether, P407 hydrogels represent promising delivery systems for the optimization of CTLA-4 blocking therapy.


Asunto(s)
Portadores de Fármacos , Hidrogeles , Anticuerpos Bloqueadores , Antígeno CTLA-4 , Sistemas de Liberación de Medicamentos , Humanos , Poloxámero , Temperatura
12.
Eur J Pharm Biopharm ; 136: 259-266, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30731115

RESUMEN

Microneedle arrays (MNAs) are a promising mean to administer vaccines. Without the need of highly trained personnel, MNAs can be applied to deliver vaccines into the dermis, which is well equipped to initiate potent immune responses. While vaccination using dissolving microneedle arrays has been extensively investigated, the use of solid nanoporous MNAs (npMNAs) to deliver vaccines remained largely unexplored. In this report we investigated whether npMNAs with an average pore size of 80 nm, can be used for influenza vaccination based on recombinant hemagglutinin (HA) protein of the 2009 pandemic H1N1 (pH1N1) virus. Fluorescently labeled HA loaded in the npMNAs was effectively delivered into the skin of mouse ears, as a result of a diffusion-based process. Compared to intramuscular immunization, intradermal HA vaccination of mice using npMNAs elicited high levels of HA antigen specific antibodies, with pH1N1 hemagglutination inhibition and neutralization activity. Moreover, mice vaccinated with pH1N1 HA loaded npMNAs were completely protected against a potentially lethal challenge with mouse adapted pH1N1 virus. These results illustrate that intradermal subunit vaccine immunization using npMNAs is a promising approach to facilitate effective vaccination.


Asunto(s)
Hemaglutininas/administración & dosificación , Vacunas contra la Influenza/administración & dosificación , Gripe Humana/prevención & control , Microinyecciones/métodos , Nanoporos , Vacunación/métodos , Animales , Cerámica/química , Cerámica/farmacocinética , Perros , Hemaglutininas/química , Hemaglutininas/metabolismo , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/fisiología , Vacunas contra la Influenza/química , Vacunas contra la Influenza/farmacocinética , Gripe Humana/inmunología , Gripe Humana/metabolismo , Células de Riñón Canino Madin Darby , Ratones , Microinyecciones/instrumentación , Agujas , Vacunación/instrumentación
13.
Pharmaceutics ; 10(4)2018 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388786

RESUMEN

Microneedle technologies have been developed for dermal drug and vaccine delivery, including hollow-, solid-, coated-, and dissolving microneedles. Microneedles have been made in many different geometries and of many different materials, all of which may influence their skin-penetrating ability. To ensure reproducible and effective drug and vaccine delivery via microneedles, the optimal insertion parameters should be known. Therefore, a digitally-controlled microneedle applicator was developed to insert microneedles into the skin via impact insertion (velocity) or via pressing force insertion. Six microneedle arrays with different geometries and/or materials were applied onto ex vivo human skin with varying velocities or pressing forces. Penetration efficiency and delivered antigen dose into the skin after application of microneedles were determined. In general, microneedles pierced the skin more efficiently when applied by impact application as compared to application via pressing force. However, the angle of application of the applicator on the skin can affect the velocity of the impact, influencing the penetration efficiency of microneedles. Regarding the antigen delivery into the skin, the delivered dose was increasing by increasing the velocity or pressure, and thus, increasing the penetration efficiency. These data demonstrate that an applicator is an important tool to determine optimal application conditions with ex vivo human skin.

14.
Pharm Res ; 35(11): 207, 2018 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-30209623

RESUMEN

PURPOSE: Personalized peptide-based cancer vaccines will be composed of multiple patient specific synthetic long peptides (SLPs) which may have various physicochemical properties. To formulate such SLPs, a flexible vaccine delivery system is required. We studied whether cationic liposomes are suitable for this purpose. METHODS: Fifteen SIINFEKL T cell epitope-containing SLPs, widely differing in hydrophobicity and isoelectric point, were separately loaded in cationic liposomes via the dehydration-rehydration method. Particle size and polydispersity index (PDI) were measured via dynamic light scattering (DLS), and zeta potential with laser Doppler electrophoresis. Peptide loading was fluorescently determined and the immunogenicity of the formulated peptides was assessed in co-cultures of dendritic cells (DCs) and CD8+ T-cells in vitro. RESULTS: All SLPs were loaded in cationic liposomes by using three different loading method variants, depending on the SLP characteristics. The fifteen liposomal formulations had a comparable size (< 200 nm), PDI (< 0.3) and zeta potential (22-30 mV). Cationic liposomes efficiently delivered the SLPs to DCs that subsequently activated SIINFEKL-specific CD8+ T-cells, indicating improved immunological activity of the SLPs. CONCLUSION: Cationic liposomes can accommodate a wide range of different SLPs and are therefore a potential delivery platform for personalized cancer vaccines.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Portadores de Fármacos/química , Epítopos de Linfocito T , Liposomas/química , Oligopéptidos/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Cationes , Composición de Medicamentos , Liberación de Fármacos , Colorantes Fluorescentes/química , Humanos , Activación de Linfocitos , Oligopéptidos/química , Oligopéptidos/inmunología , Ovalbúmina/química , Tamaño de la Partícula , Fragmentos de Péptidos/química , Biblioteca de Péptidos , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/química , Vacunas de Subunidad/inmunología
15.
Pharm Res ; 35(10): 189, 2018 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-30105542

RESUMEN

PURPOSE: To examine the immunogenicity of diphtheria toxoid (DT) loaded mesoporous silica nanoparticles (MSNs) after coated and hollow microneedle-mediated intradermal immunization in mice. METHODS: DT was loaded into MSNs and the nanoparticle surface was coated with a lipid bilayer (LB-MSN-DT). To prepare coated microneedles, alternating layers of negatively charged LB-MSN-DT and positively charged N-trimethyl chitosan (TMC) were coated onto pH-sensitive microneedle arrays via a layer-by-layer approach. Microneedle arrays coated with 5 or 3 layers of LB-MSN-DT were used to immunize mice and the elicited antibody responses were compared with those induced by hollow microneedle-injected liquid formulation of LB-MSN-DT. Liquid DT formulation with and without TMC (DT/TMC) injected by a hollow microneedle were used as controls. RESULTS: LB-MSN-DT had an average size of about 670 nm and a zeta potential of -35 mV. The encapsulation efficiency of DT in the nanoparticles was 77%. The amount of nano-encapsulated DT coated onto the microneedle array increased linearly with increasing number of the coating layers. Nano-encapsulated DT induced stronger immune responses than DT solution when delivered intradermally via hollow microneedles, but not when delivered via coated microneedles. CONCLUSION: Both the nano-encapsulation of DT and the type of microneedles affect the immunogenicity of the antigen.


Asunto(s)
Toxoide Diftérico/administración & dosificación , Nanopartículas/química , Dióxido de Silicio/química , Animales , Toxoide Diftérico/química , Toxoide Diftérico/inmunología , Composición de Medicamentos , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Inmunización , Inmunogenicidad Vacunal , Inyecciones Intradérmicas , Ratones , Ratones Endogámicos BALB C , Tamaño de la Partícula , Porosidad , Propiedades de Superficie
16.
J Control Release ; 269: 347-354, 2018 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-29174441

RESUMEN

Recent studies have shown that intradermal vaccination has great potential for T cell-mediated cancer immunotherapy. However, classical intradermal immunization with a hypodermic needle and syringe has several drawbacks. Therefore, in the present study a digitally controlled hollow microneedle injection system (DC-hMN-iSystem) with an ultra-low dead volume was developed to perform micro-injections (0.25-10µL) into skin in an automated manner. A synthetic long peptide derived from human papilloma virus formulated in cationic liposomes, which was used as a therapeutic cancer vaccine, was administered intradermally by using the DC-hMN-iSystem. Fused silica hollow microneedles with an inner diameter of 50µm and a bevel length of 66±26µm were successfully fabricated via hydrofluoric acid etching. Upon piercing these microneedles into the skin using a protrusion length of 400µm, microneedles were inserted at a depth of 350±55µm. Micro-injections of 1-10µL had an accuracy between 97 and 113% with a relative standard deviation (RSD) of 9%, and lower volumes (0.25 and 0.5µL) had an accuracy of 86-103% with a RSD of 29% in ex vivo human skin. Intradermal administration of the therapeutic cancer vaccine via micro-injections induced strong functional cytotoxic and T-helper responses in mice, while requiring much lower volumes as compared to classical intradermal immunization. In conclusion, by using the newly developed DC-hMN-iSystem, very low vaccine volumes can be precisely injected into skin in an automated manner. Thereby, this system shows potential for minimally-invasive and potentially pain-free therapeutic cancer vaccination.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Microinyecciones , Agujas , Proteínas E7 de Papillomavirus/administración & dosificación , Vacunas de Subunidad/administración & dosificación , Animales , Femenino , Humanos , Inyecciones Intradérmicas , Liposomas , Ratones Endogámicos C57BL , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología
17.
J Control Release ; 262: 28-36, 2017 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-28710002

RESUMEN

Dermal immunization using antigen-coated microneedle arrays is a promising vaccination strategy. However, reduction of microneedle sharpness and the available surface area for antigen coating is a limiting factor. To overcome these obstacles, a layer-by-layer coating approach can be applied onto pH-sensitive microneedles. Following this approach, pH-sensitive microneedle arrays (positively charged at coating pH5.8 and nearly uncharged at pH7.4) were alternatingly coated with negatively charged diphtheria toxoid (DT) and N-trimethyl chitosan (TMC), a cationic adjuvant. First, the optimal DT dose for intradermal immunization was determined in a dose-response study, which revealed that low-dose intradermal immunization was more efficient than subcutaneous immunization and that the EC50 dose of DT upon intradermal immunization is 3-fold lower, as compared to subcutaneous immunization. In a subsequent immunization study, microneedle arrays coated with an increasing number (2, 5, and 10) of DT/TMC bilayers resulted in step-wise increasing DT-specific immune responses. Dermal immunization with microneedle arrays coated with 10 bilayers of DT/TMC (corresponding with ±0.6µg DT delivered intradermally) resulted in similar DT-specific immune responses as subcutaneous immunization with 5µg of DT adjuvanted with aluminum phosphate (8-fold dose reduction). Summarizing, the layer-by-layer coating approach onto pH-sensitive microneedles is a versatile method to precisely control the amount of coated and dermally-delivered antigen that is highly suitable for dermal immunization.


Asunto(s)
Quitosano/administración & dosificación , Toxoide Diftérico/administración & dosificación , Microinyecciones , Agujas , Vacunación/instrumentación , Animales , Quitosano/química , Toxoide Diftérico/química , Relación Dosis-Respuesta Inmunológica , Liberación de Fármacos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Inmunoglobulina G/sangre , Inyecciones Subcutáneas , Ratones Endogámicos BALB C , Piel/metabolismo , Vacunación/métodos
18.
Pharm Res ; 34(8): 1693-1706, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28536970

RESUMEN

PURPOSE: To develop a new intradermal antigen delivery system by coating microneedle arrays with lipid bilayer-coated, antigen-loaded mesoporous silica nanoparticles (LB-MSN-OVA). METHODS: Synthesis of MSNs with 10-nm pores was performed and the nanoparticles were loaded with the model antigen ovalbumin (OVA), and coated with a lipid bilayer (LB-MSN-OVA). The uptake of LB-MSN-OVA by bone marrow-derived dendritic cells (BDMCs) was studied by flow cytometry. The designed LB-MSN-OVA were coated onto pH-sensitive pyridine-modified microneedle arrays and the delivery of LB-MSN-OVA into ex vivo human skin was studied. RESULTS: The synthesized MSNs demonstrated efficient loading of OVA with a maximum loading capacity of about 34% and the lipid bilayer enhanced the colloidal stability of the MSNs. Uptake of OVA loaded in LB-MSN-OVA by BMDCs was higher than that of free OVA, suggesting effective targeting of LB-MSN-OVA to antigen-presenting cells. Microneedles were readily coated with LB-MSN-OVA at pH 5.8, yielding 1.5 µg of encapsulated OVA per microneedle array. Finally, as a result of the pyridine modification, LB-MSN-OVA were effectively released from the microneedles upon piercing the skin. CONCLUSION: Microneedle arrays coated with LB-MSN-OVA were successfully developed and shown to be suitable for intradermal delivery of the encapsulated protein antigen.


Asunto(s)
Antígenos/administración & dosificación , Nanopartículas/química , Agujas , Ovalbúmina/administración & dosificación , Dióxido de Silicio/química , Células Presentadoras de Antígenos/metabolismo , Portadores de Fármacos , Liberación de Fármacos , Humanos , Concentración de Iones de Hidrógeno , Inyecciones Intradérmicas , Membrana Dobles de Lípidos , Macrófagos/metabolismo , Tamaño de la Partícula , Porosidad , Piel , Propiedades de Superficie
19.
Front Immunol ; 8: 1789, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29375544

RESUMEN

The skin is immunologically very potent because of the high number of antigen-presenting cells in the dermis and epidermis, and is therefore considered to be very suitable for vaccination. However, the skin's physical barrier, the stratum corneum, prevents foreign substances, including vaccines, from entering the skin. Microneedles, which are needle-like structures with dimensions in the micrometer range, form a relatively new approach to circumvent the stratum corneum, allowing for minimally invasive and pain-free vaccination. In this study, we tested ceramic nanoporous microneedle arrays (npMNAs), representing a novel microneedle-based drug delivery technology, for their ability to deliver the subunit vaccines diphtheria toxoid (DT) and tetanus toxoid (TT) intradermally. First, the piercing ability of the ceramic (alumina) npMNAs, which contained over 100 microneedles per array, a length of 475 µm, and an average pore size of 80 nm, was evaluated in mouse skin. Then, the hydrodynamic diameters of DT and TT and the loading of DT, TT, and imiquimod into, and subsequent release from the npMNAs were assessed in vitro. It was shown that DT and TT were successfully loaded into the tips of the ceramic nanoporous microneedles, and by using near-infrared fluorescently labeled antigens, we found that DT and TT were released following piercing of the antigen-loaded npMNAs into ex vivo murine skin. Finally, the application of DT- and TT-loaded npMNAs onto mouse skin in vivo led to the induction of antigen-specific antibodies, with titers similar to those obtained upon subcutaneous immunization with a similar dose. In conclusion, we show for the first time, the potential of npMNAs for intradermal (ID) immunization with subunit vaccines, which opens possibilities for future ID vaccination designs.

20.
J Control Release ; 242: 141-147, 2016 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-27496634

RESUMEN

The purpose of this study was to investigate the effect of various repeated fractional intradermal dosing schedules of inactivated polio vaccine serotype 1 (IPV1) on IPV1-specific IgG responses in rats. By utilizing an applicator that allowed for precisely controlled intradermal microinjections by using a single hollow microneedle, rats were immunized intradermally with 5 D-antigen units (DU) of IPV1 at 150µm skin depth. This dose was administered as a bolus, or in a repeated fractional dosing schedule: 4 doses of 1.25 DU (1/4th of total dose) were administered on four consecutive days or every other day; 8 doses of 0.625 DU (1/8th of total dose) were administered on eight consecutive days; or 4 exponentially increasing doses (0.04, 0.16, 0.8 and 4 DU), either with or without an exponentially increasing CpG oligodeoxynucleotide 1826 (CpG) dose, were administered on four consecutive days. All of these fractional dosing schedules resulted in up to ca. 10-fold higher IPV1-specific IgG responses than intradermal and intramuscular bolus dosing. IPV1 combined with adjuvant CpG in exponential dosing did not significantly increase the IPV1-specific IgG responses further, which demonstrated that maximal responses were achieved by fractional dosing. In conclusion, repeated fractional intradermal IPV1 dosing leads to superior IPV1-specific IgG responses without the use of adjuvants. These results indicate that a controlled release delivery system for intradermal IPV1 delivery can potentiate IPV1-specific IgG responses.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Inmunoglobulina G/inmunología , Oligodesoxirribonucleótidos/administración & dosificación , Vacuna Antipolio de Virus Inactivados/administración & dosificación , Animales , Preparaciones de Acción Retardada , Esquema de Medicación , Sistemas de Liberación de Medicamentos , Femenino , Inyecciones Intradérmicas , Microinyecciones , Agujas , Vacuna Antipolio de Virus Inactivados/inmunología , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...