Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Nanomedicine ; 58: 102749, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38719107

ABSTRACT

New adjuvant strategies are needed to improve protein-based subunit vaccine immunogenicity. We examined the potential to use nanostructure of 6-O-ascorbyl palmitate to formulate ovalbumin (OVA) protein and an oligodeoxynucleotide (CpG-ODN) (OCC). In mice immunized with a single dose, OCC elicited an OVA-specific immune response superior to OVA/CpG-ODN solution (OC). Rheological studies demonstrated OCC's self-assembling viscoelastic properties. Biodistribution studies indicated that OCC prolonged OVA and CpG-ODN retention at injection site and lymph nodes, reducing systemic spread. Flow-cytometry assays demonstrated that OCC promoted OVA and CpG-ODN co-uptake by Ly6ChiCD11bhiCD11c+ monocytes. OCC and OC induced early IFN-γ in lymph nodes, but OCC led to higher concentration. Conversely, mice immunized with OC showed higher serum IFN-γ concentration compared to those immunized with OCC. In mice immunized with OCC, NK1.1+ cells were the IFN-γ major producers, and IFN-γ was essential for OVA-specific IgG2c switching. These findings illustrate how this nanostructure improves vaccine's response.


Subject(s)
Nanostructures , Oligodeoxyribonucleotides , Ovalbumin , Vaccines, Subunit , Animals , Nanostructures/chemistry , Vaccines, Subunit/immunology , Vaccines, Subunit/chemistry , Vaccines, Subunit/pharmacokinetics , Mice , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/pharmacokinetics , Ovalbumin/immunology , Ovalbumin/chemistry , Female , Mice, Inbred C57BL , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacokinetics , Interferon-gamma/metabolism , Tissue Distribution , Ascorbic Acid/analogs & derivatives
2.
Mol Pharm ; 17(10): 3845-3856, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32902989

ABSTRACT

Ionic liquids (ILs) attract significant attention as novel solvents for drug delivery systems because of their ability to solubilize poorly soluble drugs and tune the physiological properties of active pharmaceutical ingredients. For the next generation of IL-based drug delivery systems, biocompatibility is a high priority. In the current study, choline-fatty acids ([Cho][FA]) were used as a biocompatible IL to mediate the dissolution of a water-soluble antigen peptide in an oil-based skin penetration enhancer. Among the candidate fatty acids (C8, C10, C12, C14, C16, C18:0, and C18:1), C18:1 was selected because of its low cytotoxicity and mediation of skin permeability for an antigen peptide. Using IL[Cho][C18:1] and an oil-based penetration enhancer, the flux of transdermal delivery of the peptide increased 28-fold compared with delivery using an aqueous vehicle. Furthermore, the IL-mediated transcutaneous vaccination succeeded in suppressing tumor growth in vivo compared to injection. The skin irritation produced by this formulation was tested using an in vitro 3D constructed skin tissue model and an in vivo histological study, which concluded that the formulation did not cause skin irritation. The results suggest that biocompatible IL-mediated dissolution in an oil-based skin penetration enhancer is a promising strategy for transdermal drug delivery.


Subject(s)
Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Drug Carriers/chemistry , Ionic Liquids/chemistry , Neoplasms/prevention & control , Administration, Cutaneous , Animals , Cancer Vaccines/chemistry , Cancer Vaccines/pharmacokinetics , Cell Line, Tumor/transplantation , Choline/chemistry , Disease Models, Animal , Fatty Acids/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Neoplasms/immunology , Permeability , Skin , Skin Absorption , Solvents/chemistry , Vaccination/methods , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/chemistry , Vaccines, Subunit/pharmacokinetics
3.
Nat Biomed Eng ; 4(6): 636-648, 2020 06.
Article in English | MEDLINE | ID: mdl-32483299

ABSTRACT

The formulations of peptide-based antitumour vaccines being tested in clinical studies are generally associated with weak potency. Here, we show that pharmacokinetically tuning the responses of peptide vaccines by fusing the peptide epitopes to carrier proteins optimizes vaccine immunogenicity in mice. In particular, we show in immunized mice that the carrier protein transthyretin simultaneously optimizes three factors: efficient antigen uptake in draining lymphatics from the site of injection, protection of antigen payloads from proteolytic degradation and reduction of antigen presentation in uninflamed distal lymphoid organs. Optimizing these factors increases vaccine immunogenicity by up to 90-fold and maximizes the responses to viral antigens, tumour-associated antigens, oncofetal antigens and shared neoantigens. Protein-peptide epitope fusions represent a facile and generalizable strategy for enhancing the T-cell responses elicited by subunit vaccines.


Subject(s)
Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Immunogenicity, Vaccine/immunology , T-Lymphocytes/immunology , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacokinetics , Albumins/immunology , Animals , Antigens, Neoplasm , Basic-Leucine Zipper Transcription Factors , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Epitopes , Immunity, Cellular , Immunotherapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/genetics
4.
Int J Pharm ; 573: 118868, 2020 Jan 05.
Article in English | MEDLINE | ID: mdl-31765785

ABSTRACT

Vaccines are the front line in the fight against diseases. However, setbacks with existing cholera vaccines have ignited a considerable effort to develop more suitable vaccine formulations. In this study, we aim to investigate the effect of antigen stability and controlled release in inducing an immune response. Therefore, two types of silica and carbon mesoporous nanoparticles of the same size and shape but different pore architectures were synthesized and loaded with recombinant cholera toxin subunit B to serve as a model for antigen stability and controlled release of antigenic CTB. In order to evaluate immune response efficacy for these model formulations, IgG and IgA responses and fluid accumulation (FA) index were measured in immunized rabbits, which were challenged with wild-type Vibrio cholerae. Our result suggests that mesoporous silica nanoparticles have greater efficacy in inducing mucosal immune responses, and it proved more proficiency in overall immune responses in challenge experiments and FA index (p < 0.05). These findings indicate that mesoporous nanoparticles and, in particular, mesoporous silica nanoparticles, could be used in oral vaccine formulation against cholera.


Subject(s)
Cholera Toxin/immunology , Cholera Vaccines/immunology , Cholera/prevention & control , Drug Carriers/chemistry , Administration, Oral , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Cholera/blood , Cholera/microbiology , Cholera Toxin/genetics , Cholera Toxin/pharmacokinetics , Cholera Vaccines/administration & dosage , Cholera Vaccines/pharmacokinetics , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Disease Models, Animal , Dose-Response Relationship, Immunologic , Female , Humans , Immunity, Mucosal , Immunogenicity, Vaccine , Nanoparticles/chemistry , Porosity , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacokinetics , Silicon Dioxide/chemistry , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacokinetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vibrio cholerae/immunology
5.
Drug Dev Ind Pharm ; 45(6): 882-894, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30767591

ABSTRACT

Pulmonary infections are the most common cause of death globally. However, the development of mucosal vaccines that provide protective immunity against respiratory pathogens are limited. In contrast to needle-based vaccines, efficient vaccines that are delivered via noninvasive mucosal routes (such as via the lungs and nasal passage) produce both antigen-specific local mucosal IgA and systemic IgG protective antibodies. One major challenge in the development of pulmonary vaccines using subunit antigens however, is the production of optimal immune responses. Subunit vaccines therefore rely upon use of adjuvants to potentiate immune responses. While the lack of suitable mucosal adjuvants has hindered progress in the development of efficient pulmonary vaccines, particle-based systems can provide an alternative approach for the safe and efficient delivery of subunit vaccines. In particular, the rational engineering of particulate vaccines with optimal physicochemical characteristics can produce long-term protective immunity. These protect antigens against enzymatic degradation, target antigen presenting cells and initiate optimal humoral and cellular immunity. This review will discuss our current understanding of pulmonary immunology and developments in fabricating particle characteristics that may evoke potent and durable pulmonary immunity.


Subject(s)
Drug Delivery Systems/methods , Lung/metabolism , Respiratory Mucosa/metabolism , Respiratory Tract Infections/prevention & control , Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacokinetics , Administration, Inhalation , Administration, Intranasal/instrumentation , Animals , Dry Powder Inhalers , Feasibility Studies , Humans , Immunogenicity, Vaccine , Lung/immunology , Nanoparticles/chemistry , Particle Size , Permeability , Respiratory Mucosa/immunology , Respiratory Tract Infections/microbiology , Vaccines/immunology , Vaccines/pharmacokinetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacokinetics
6.
Front Immunol ; 9: 2825, 2018.
Article in English | MEDLINE | ID: mdl-30555488

ABSTRACT

Pulmonary tuberculosis (TB), which is caused by Mycobacterium tuberculosis (Mtb), remains a global pandemic, despite the widespread use of the parenteral live attenuated Bacillus Calmette-Guérin (BCG) vaccine during the past decades. Mucosal administration of next generation TB vaccines has great potential, but developing a safe and efficacious mucosal vaccine is challenging. Hence, understanding the in vivo biodistribution and pharmacokinetics of mucosal vaccines is essential for shaping the desired immune response and for optimal spatiotemporal targeting of the appropriate effector cells in the lungs. A subunit vaccine consisting of the fusion antigen H56 (Ag85B-ESAT-6-Rv2660) and the liposome-based cationic adjuvant formulation (CAF01) confers efficient protection in preclinical animal models. In this study, we devise a novel immunization strategy for the H56/CAF01 vaccine, which comply with the intrapulmonary (i.pulmon.) route of immunization. We also describe a novel dual-isotope (111In/67Ga) radiolabeling approach, which enables simultaneous non-invasive and longitudinal SPECT/CT imaging and quantification of H56 and CAF01 upon parenteral prime and/or i.pulmon. boost immunization. Our results demonstrate that the vaccine is distributed evenly in the lungs, and there are pronounced differences in the pharmacokinetics of H56 and CAF01. We provide convincing evidence that the H56/CAF01 vaccine is not only well-tolerated when administered to the respiratory tract, but it also induces strong lung mucosal and systemic IgA and polyfunctional Th1 and Th17 responses after parenteral prime and i.pulmon. boost immunization. The study furthermore evaluate the application of SPECT/CT imaging for the investigation of vaccine biodistribution after parenteral and i.pulmon. immunization of mice.


Subject(s)
Antibodies, Bacterial/immunology , Immunity, Mucosal/drug effects , Immunization, Secondary , Immunoglobulin A/immunology , Lung/immunology , Mycobacterium tuberculosis/immunology , T-Lymphocytes/immunology , Tuberculosis Vaccines , Animals , Female , Mice , Single Photon Emission Computed Tomography Computed Tomography , T-Lymphocytes/pathology , Tuberculosis Vaccines/pharmacokinetics , Tuberculosis Vaccines/pharmacology , Vaccines, Subunit/pharmacokinetics , Vaccines, Subunit/pharmacology
7.
Cell Rep ; 23(5): 1435-1447, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29719256

ABSTRACT

There are a limited number of adjuvants that elicit effective cell-based immunity required for protection against intracellular bacterial pathogens. Here, we report that STING-activating cyclic dinucleotides (CDNs) formulated in a protein subunit vaccine elicit long-lasting protective immunity to Mycobacterium tuberculosis in the mouse model. Subcutaneous administration of this vaccine provides equivalent protection to that of the live attenuated vaccine strain Bacille Calmette-Guérin (BCG). Protection is STING dependent but type I IFN independent and correlates with an increased frequency of a recently described subset of CXCR3-expressing T cells that localize to the lung parenchyma. Intranasal delivery results in superior protection compared with BCG, significantly boosts BCG-based immunity, and elicits both Th1 and Th17 immune responses, the latter of which correlates with enhanced protection. Thus, a CDN-adjuvanted protein subunit vaccine has the capability of eliciting a multi-faceted immune response that results in protection from infection by an intracellular pathogen.


Subject(s)
Adjuvants, Immunologic/pharmacology , BCG Vaccine/pharmacology , Membrane Proteins/immunology , Mycobacterium tuberculosis/immunology , Th17 Cells/immunology , Tuberculosis, Pulmonary/prevention & control , Animals , BCG Vaccine/immunology , Disease Models, Animal , Immunity, Cellular/drug effects , Mice , Mice, Knockout , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/pathology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacokinetics
8.
Bioconjug Chem ; 29(3): 776-785, 2018 03 21.
Article in English | MEDLINE | ID: mdl-29436221

ABSTRACT

Peptide subunit vaccines are desirable because they increase control over the immune response and safety of the vaccine by reducing the risk of off-target responses to molecules other than the target antigen. The immunogenicity of most peptides, however, is low. Peptide nanoclusters (PNC) are proposed as a subunit peptide vaccine delivery system made completely of cross-linked peptide antigen that could improve the immunogenicity of a peptide vaccine. Proof of concept is demonstrated with oncofetal antigen (OFA), an immature laminin receptor protein expressed by many hematologic cancer cells but not by healthy cells. Peptide epitopes from this protein, called OFA 1, 2, and 3, were synthesized into PNC as a potential cancer peptide vaccine delivery system. PNC were formed by desolvation and stabilized with disulfide bonds using a trithiol cross-linker. Cysteines were added to the C-terminus of each peptide to assist in this cross-linking step, denoted OFA 1C, 2C, and 3C PNC. OFA 2C was found to form the smallest PNC, 148 ± 15 nm in diameter and stable in solution. This size is in the range where particles are readily internalized by dendritic cells (DCs) and may also passively diffuse to regional lymph nodes. OFA 2C PNC and soluble OFA 2C were internalized similarly by DCs in vitro, but only PNC resulted in significant peptide presentation by DCs. This indicates the potential for PNC to improve immune activation against this antigen. Additionally, PNC displayed higher retention at the intradermal injection site in vivo than soluble peptide, allowing more time to interact with DCs in an area of increased DC activity. While offering traditional nanoparticle benefits such as increased DC recognition, slower diffusion, and potential for multivalent cellular interactions, PNC also maximize antigen delivered per particle while minimizing off-target material delivery because the antigens are the main building blocks of the particle. With these properties, PNC are a delivery system with potential to increase peptide subunit vaccine immunogenicity for OFA and other peptide antigens.


Subject(s)
Antigens, Neoplasm/administration & dosage , Cancer Vaccines/administration & dosage , Peptides/administration & dosage , Animals , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Cancer Vaccines/pharmacokinetics , Cell Line , Cross-Linking Reagents/chemistry , Dendritic Cells/immunology , Humans , Mice , Neoplasms/immunology , Neoplasms/prevention & control , Peptides/chemistry , Peptides/immunology , Peptides/pharmacokinetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacokinetics
9.
Mol Pharm ; 15(3): 975-984, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29359945

ABSTRACT

In immunotherapy, induction of potent cellular immunity by vaccination is essential to treat intracellular infectious diseases and tumors. In this work, we designed a new synthetic peptide carrier, Cys-Trp-Trp-Arg8-Cys-Arg8-Cys-Arg8-Cys, for vaccine delivery by integrating a redox-responsive disulfide bond cross-linking and cell-penetrating peptide arginine octamer. The carrier peptide bound to the antigen protein ovalbumin (OVA) via electrostatic self-assembly to form peptide/OVA nanocomposites. Then, the spontaneous oxidization of the thiols of the cysteine residues induced interpeptide disulfide bond cross-linking to construct denser peptide/OVA condensates. The cell-penetrating peptides incorporated in the carrier peptide could increase antigen uptake by antigen presenting cells. After being internalized by antigen presenting cells, the antigen could be rapidly released in cytoplasm along with degradation of the disulfide bonds by intracellular glutathione, which could promote potent CD8+ T cell immunity. The cross-linked peptide/OVA condensates were used for subcutaneous vaccination. The results showed that the peptide carrier mediated potent antigen-specific immune response by significantly increasing IgG titer; splenocyte proliferation; the secretion level of cytokines INF-γ, IL-12, IL-4, and IL-10; immune memory function, and the activation and maturation of dendritic cells. From the results, the low-molecular weight vaccine-condensing peptide with definite chemical composition could be developed as a novel class of vaccine delivery systems.


Subject(s)
Cell-Penetrating Peptides/chemistry , Drug Delivery Systems/methods , Ovalbumin/administration & dosage , Vaccination/methods , Vaccines, Subunit/administration & dosage , Animals , Antigens/administration & dosage , Antigens/immunology , Cell Line , Cell Membrane Permeability/drug effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Female , Immunity, Cellular/drug effects , Immunologic Memory/drug effects , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Nanoparticles/chemistry , Ovalbumin/immunology , Ovalbumin/pharmacokinetics , Oxidation-Reduction , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacokinetics
10.
Curr Pharm Des ; 22(17): 2561-76, 2016.
Article in English | MEDLINE | ID: mdl-26831645

ABSTRACT

In the past century, vaccines have contributed to a significant improvement in global public health by preventing a number of infectious diseases. Despite this, the vaccine field is still facing challenges related to incomplete vaccine coverage and persistent difficult vaccine targets, such as influenza, tuberculosis, and Ebola, for which no good universal vaccines exist. At least two pharmaceutical improvements are expected to help filling this gap: i) The development of thermostable vaccine dosage forms, and ii) the full exploitation of the adjuvant technology for subunit vaccines to potentiate strong immune responses. This review highlights the status and recent advances in formulation and pulmonary delivery of thermostable human subunit vaccines. Such vaccines are very appealing from compliance, distribution and immunological point of view: Being non-invasive, inhalable vaccines are self-administrable, can be distributed independently of functioning freezers and refrigerators, and can be designed to induce mucosal and/or cell-mediated immunity, which is attractive for a number of diseases requiring stimulation of local mucosal immunity for protection. However, the design and delivery of thermostable dry powder-based vaccines represents a technological challenge: It calls for careful formulation and dosage form design, combined with cheap and efficient delivery devices, which must be engineered via a thorough understanding of the physiological barrier and the requirements for induction of mucosal immunity. Here, I review state of the art and perspectives in formulation design and processing methods for powder-based subunit vaccines intended for pulmonary administration, and present dry powder inhaler technologies suitable for translating these vaccines into clinical trials.


Subject(s)
Drug Delivery Systems , Dry Powder Inhalers , Lung/metabolism , Temperature , Vaccines, Subunit/chemistry , Vaccines, Subunit/pharmacokinetics , Administration, Inhalation , Humans , Vaccines, Subunit/administration & dosage
11.
Vaccine ; 33(36): 4630-8, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26141014

ABSTRACT

Transdermal immunization results in poor immunogenicity, which can be attributed to poor permeability of antigens through the skin. Therefore, elastic liposome, ultradeformable lipid vesicles, may overcome the challenges faced during transdermal immunization. This versatile carrier proves better vehicle for transcutaneous delivery of protein, peptide and nucleic acid antigens. The present results are suggestive of improved immunogenicity of carboxyl-terminal 19 kDa fragment of merozoite surface protein-1 (PfMSP-119) of Plasmodium falciparum when administered subcutaneously through elastic liposomes. The prepared elastic liposomes were characterized with respect to vesicles shape and surface morphology, size and size distribution, entrapment efficiency, elasticity, stability and in vitro release. Humoral and cell-mediated immune (CMI) response elicited by topically applied PfMSP-119-loaded elastic liposomes, intramuscularly administered alum-adsorbed PfMSP-119 solution, and topically applied PfMSP-119-loaded conventional liposomes were compared and normalized with vehicle control. Results suggest greater transcutaneous immunization via elastic liposomes, and induced robust and perdurable IgG-specific antibody and cytophilic isotype responses. We report to have achieved sizeable CMI activating factor (IFNγ), a crucial player in conferring resistance to asexual blood stage malaria, responses with elastic liposomes when compared with other formulations. The fluorescence microscopy and histopathology results are suggestive of prominent skin permeation and biodistribution, and demonstrate efficient delivery of malaria antigen via elastic liposomes to immunocompetent Langerhans cells (LC) and lymphatics. In conclusion, elastic liposomal formulation provided greater entrapment efficiency, enhanced penetration and heightened and long-lasting immune response. Moreover, effective immunoadjuvant property of this carrier justifies its potential for improved vaccine delivery, and opens new avenues to explore further on the development of malaria vaccine.


Subject(s)
Antigens, Protozoan/immunology , Drug Carriers/administration & dosage , Liposomes/administration & dosage , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Merozoite Surface Protein 1/immunology , Plasmodium falciparum/immunology , Administration, Cutaneous , Animals , Antibodies, Protozoan/blood , Drug Carriers/pharmacokinetics , Female , Humans , Injections, Intramuscular , Liposomes/pharmacokinetics , Malaria Vaccines/pharmacokinetics , Mice, Inbred BALB C , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacokinetics
12.
Cancer Sci ; 105(7): 924-31, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25040224

ABSTRACT

Recent progress in fundamental understanding of tumor immunology has opened a new avenue of cancer vaccines. Currently, the development of new cancer vaccines is a global topic and has attracted attention as one of the most important issues in Japan. There is an urgent need for the development of guidance for cancer vaccine clinical studies in order to lead to drug development. Peptide vaccines characteristically have the effect of indirectly acting against cancer through the immune system - a mechanism of action that clearly differs from anticancer drugs that exert a direct effect. Thus, the clinical development of cancer peptide vaccines should be planned and implemented based on the mechanism of action, which differs significantly from conventional anticancer drug research. The Japanese Society for Biological Therapy has created and published Guidance for peptide vaccines for the treatment of cancer as part of its mission and responsibilities towards cancer peptide vaccine development, which is now pursued globally. We welcome comments from regulators and business people as well as researchers in this area.


Subject(s)
Cancer Vaccines/pharmacology , Neoplasms/immunology , Vaccines, Subunit/pharmacology , Cancer Vaccines/therapeutic use , Clinical Trials as Topic , Humans , Neoplasms/therapy , Quality Control , Vaccines, Subunit/pharmacokinetics
13.
Int J Nanomedicine ; 8: 2085-99, 2013.
Article in English | MEDLINE | ID: mdl-23785233

ABSTRACT

We recently demonstrated by in vitro experiments that PLGA (poly D, L-lactide-co-glycolide) potentiates T helper 1 (Th1) immune responses induced by a peptide derived from the recombinant major outer membrane protein (rMOMP) of Chlamydia trachomatis, and may be a promising vaccine delivery system. Herein we evaluated the immune-potentiating potential of PLGA by encapsulating the full-length rMOMP (PLGA-rMOMP), characterizing it in vitro, and investigating its immunogenicity in vivo. Our hypothesis was that PLGA-rMOMP triggers Th1 immune responses in mice, which are desirable prerequisites for a C. trachomatis candidate nanovaccine. Physical-structural characterizations of PLGA-rMOMP revealed its size (approximately 272 nm), zeta potential (-14.30 mV), apparent spherical smooth morphology, and continuous slow release pattern. PLGA potentiated the ability of encapsulated rMOMP to trigger production of cytokines and chemokines by mouse J774 macrophages. Flow cytometric analyses revealed that spleen cells from BALB/c mice immunized with PLGA-rMOMP had elevated numbers of CD4+ and CD8+ T cell subsets, and secreted more rMOMP-specific interferon-gamma (Th1) and interleukin (IL)-12p40 (Th1/Th17) than IL-4 and IL-10 (Th2) cytokines. PLGA-rMOMP-immunized mice produced higher serum immunoglobulin (Ig)G and IgG2a (Th1) than IgG1 (Th2) rMOMP-specific antibodies. Notably, sera from PLGA-rMOMP-immunized mice had a 64-fold higher Th1 than Th2 antibody titer, whereas mice immunized with rMOMP in Freund's adjuvant had only a four-fold higher Th1 than Th2 antibody titer, suggesting primarily induction of a Th1 antibody response in PLGA-rMOMP-immunized mice. Our data underscore PLGA as an effective delivery system for a C. trachomatis vaccine. The capacity of PLGA-rMOMP to trigger primarily Th1 immune responses in mice promotes it as a highly desirable candidate nanovaccine against C. trachomatis.


Subject(s)
Bacterial Vaccines/immunology , Chlamydia trachomatis/immunology , Nanoparticles/chemistry , Porins/immunology , Vaccines, Subunit/immunology , Animals , Antibodies, Bacterial/blood , Bacterial Vaccines/chemistry , Bacterial Vaccines/pharmacokinetics , Cell Line , Chemokines/analysis , Chemokines/metabolism , Cytokines/analysis , Cytokines/metabolism , Female , Flow Cytometry , Lactic Acid/chemistry , Macrophages , Mice , Mice, Inbred BALB C , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Porins/chemistry , Porins/pharmacokinetics , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/pharmacokinetics , Th1 Cells , Vaccines, Subunit/chemistry , Vaccines, Subunit/pharmacokinetics
14.
Article in English | MEDLINE | ID: mdl-23532930

ABSTRACT

Vaccination has had a major impact on the control of infectious diseases. However, there are still many infectious diseases for which the development of an effective vaccine has been elusive. In many cases the failure to devise vaccines is a consequence of the inability of vaccine candidates to evoke appropriate immune responses. This is especially true where cellular immunity is required for protective immunity and this problem is compounded by the move toward devising sub-unit vaccines. Over the past decade nanoscale size (<1000 nm) materials such as virus-like particles, liposomes, ISCOMs, polymeric, and non-degradable nanospheres have received attention as potential delivery vehicles for vaccine antigens which can both stabilize vaccine antigens and act as adjuvants. Importantly, some of these nanoparticles (NPs) are able to enter antigen-presenting cells by different pathways, thereby modulating the immune response to the antigen. This may be critical for the induction of protective Th1-type immune responses to intracellular pathogens. Their properties also make them suitable for the delivery of antigens at mucosal surfaces and for intradermal administration. In this review we compare the utilities of different NP systems for the delivery of sub-unit vaccines and evaluate the potential of these delivery systems for the development of new vaccines against a range of pathogens.


Subject(s)
Drug Carriers/administration & dosage , Nanoparticles/administration & dosage , Vaccines, Subunit/administration & dosage , Antigen-Presenting Cells/immunology , Drug Carriers/pharmacokinetics , Immunity, Cellular , Th1 Cells/immunology , Vaccines, Subunit/pharmacokinetics
15.
J Pharm Pharmacol ; 64(11): 1560-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23058043

ABSTRACT

OBJECTIVES: Lipid colloidal vaccines, including liposomes, transfersomes, ethosomes and cubosomes, were formulated, characterised and investigated for their ability to enhance penetration of a peptide vaccine through stillborn piglet skin in vitro. METHODS: Liposomes and transfersomes were formulated using a film-hydration method, ethosomes using a modified reverse phase method and cubosomes using a lipid precursor method. The size, zeta potential, peptide loading and interfacial behaviour of the formulations were characterised. Skin penetration studies were performed using Franz diffusion cells with piglet skin as the membrane. The localization of peptide in the skin was examined using confocal laser scanning microscopy. KEY FINDING: The various formulations contained negatively charged particles of similar size (range: 134-200 nm). Addition of the saponin adjuvant Quil A to the formulations destabilised the monolayers and reduced peptide loading. Cubosomes and ethosomes showed superior skin retention compared with the other systems. Confocal laser scanning microscopy showed greater peptide penetration and accumulation in the skin treated with cubosomes and ethosomes. With the other systems peptide was only located in the vicinity of the hair follicles and within the hair shaft. CONCLUSIONS: We conclude from the in-vitro studies that cubosomes and ethosomes are promising lipid carriers for transcutaneous immunisation.


Subject(s)
Drug Delivery Systems , Saponins/chemistry , Skin Absorption , Vaccines, Subunit/pharmacokinetics , Adjuvants, Immunologic/chemistry , Animals , Colloids , Lipids/chemistry , Liposomes , Microscopy, Confocal , Particle Size , Peptides/administration & dosage , Peptides/immunology , Peptides/pharmacokinetics , Quillaja Saponins , Swine , Vaccines, Subunit/administration & dosage
16.
Nat Med ; 18(8): 1291-6, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22797811

ABSTRACT

Both rectal and vaginal mucosal surfaces serve as transmission routes for pathogenic microorganisms. Vaccination through large intestinal mucosa, previously proven protective for both of these mucosal sites in animal studies, can be achieved successfully by direct intracolorectal (i.c.r.) administration, but this route is clinically impractical. Oral vaccine delivery seems preferable but runs the risk of the vaccine's destruction in the upper gastrointestinal tract. Therefore, we designed a large intestine-targeted oral delivery with pH-dependent microparticles containing vaccine nanoparticles, which induced colorectal immunity in mice comparably to colorectal vaccination and protected against rectal and vaginal viral challenge. Conversely, vaccine targeted to the small intestine induced only small intestinal immunity and provided no rectal or vaginal protection, demonstrating functional compartmentalization within the gut mucosal immune system. Therefore, using this oral vaccine delivery system to target the large intestine, but not the small intestine, may represent a feasible new strategy for immune protection of rectal and vaginal mucosa.


Subject(s)
Drug Delivery Systems/methods , Intestine, Large , Rectum/immunology , Vaccinia virus/immunology , Vaccinia/prevention & control , Vagina/immunology , Viral Vaccines/administration & dosage , Adjuvants, Immunologic , Administration, Oral , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Immunity, Mucosal , Intestine, Large/virology , Lactic Acid , Lipopeptides , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Nanoparticles , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Organ Specificity , Ovary/virology , Poly I-C , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Polymethacrylic Acids , Specific Pathogen-Free Organisms , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacokinetics , Vaccinia/immunology , Vaccinia virus/isolation & purification , Viral Load , Viral Vaccines/immunology , Viral Vaccines/pharmacokinetics
17.
J Immunother ; 27(1): 60-72, 2004.
Article in English | MEDLINE | ID: mdl-14676634

ABSTRACT

Two different trials of peptide vaccination were conducted for patients with recurrent gynecologic cancers. In the first regimen, four HLA-A24+ patients (two with cervical cancer and two with ovarian cancer) were vaccinated with peptides that were predesignated before vaccination. Three patients exhibited with a grade 1 adverse effect, and no clinical response was observed in any patients. In the second regimen, six HLA-A24+ and four HLA-A2+ patients (five with cervical cancer, one with endometrial cancer, one with uterine sarcoma, and three with ovarian cancer) were vaccinated with peptides (maximum four) to which preexisting cytotoxic T lymphocyte precursors in the periphery were confirmed before vaccination. With this regimen, grade 1 adverse effects were observed in eight patients, a grade 2 adverse effect in one patient, and a grade 3 adverse effect (ie, rectal bleeding) in one patient. However, this regimen was able to enhance peptide-specific cytotoxic T lymphocytes in seven of ten patients, and three of five cervical cancer patients showed objective tumor regression. Analysis of immunoglobulin G -reactive to administered peptides suggested that the induction of peptide-specific immunoglobulin G was correlated with clinical responses. Overall, these results suggest that peptide vaccination of patients showing evidence of preexisting peptide-specific cytotoxic T lymphocyte precursors could be superior to vaccination with predesignated peptides, and that the evidence-based regimen is applicable for clinical trials in treatment of patients with recurrent gynecologic cancers.


Subject(s)
Cancer Vaccines/therapeutic use , Vaccines, Subunit/therapeutic use , Adult , Aged , Carcinosarcoma/therapy , Endometrial Neoplasms/therapy , Female , HLA-A Antigens/immunology , HLA-A2 Antigen/immunology , HLA-A24 Antigen , Humans , Immunity, Cellular , Immunoglobulin G/analysis , Immunoglobulin G/blood , Middle Aged , Ovarian Neoplasms/therapy , Skin Tests , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Time Factors , Uterine Cervical Neoplasms/therapy , Vaccination , Vaccines, Subunit/pharmacokinetics
18.
Biosci Rep ; 22(2): 355-69, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12428910

ABSTRACT

Oral vaccination requires an antigen delivery vehicle to protect the antigen and to enhance translocation of the antigen to the mucosa-associated lymphoid tissue. A variety of antigen delivery vehicles including liposomes have been studied for mucosal immunization. The advantages of liposome formulations are their particulate form and the ability to accommodate immunomodulators and targeting molecules in the same package. Many conventional liposomes are variably unstable in acids, pancreatic juice and bile. Nevertheless, carefully designed liposomes have demonstrated an impressive efficacy in inducing mucosal IgA responses, compared to free antigens and other delivery vehicles. However, liposomes as an oral vaccine vehicle are not yet optimized. To design liposomes that are stable in the harsh intestinal environment and are efficiently taken up by the M cells remains a challenge. This review summarizes recent research efforts using liposomes as an antigen carrier for oral vaccines with practical attention to liposome designs and interaction with the M cells.


Subject(s)
Antigens/administration & dosage , Immunity, Mucosal , Liposomes , Vaccination/methods , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacokinetics , Administration, Oral , Administration, Rectal , Animals , Antigen-Presenting Cells/metabolism , Antigens/immunology , Biological Transport , Drug Stability , Female , Gastric Mucosa/cytology , Gastric Mucosa/immunology , Glycocalyx/metabolism , Haplorhini , Humans , Immunoglobulin A, Secretory/biosynthesis , Intestinal Absorption , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Male , Membrane Lipids/pharmacokinetics , Phospholipids/pharmacokinetics , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...