Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Eur J Pharm Sci ; 195: 106730, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38382622

ABSTRACT

Tuberculosis (TB) is still among the deadliest infectious diseases, hence there is a pressing need for more effective TB vaccines. Cationic liposome subunit vaccines are excellent vaccine candidates offering effective protection with a better safety profile than live vaccines. In this study, we aim to explore intrinsic adjuvant properties of cationic liposomes to maximize immune activation while minimizing aspecific cytotoxicity. To achieve this, we developed a rational strategy to select liposomal formulation compositions and assessed their physicochemical and immunological properties in vitro models using human monocyte-derived dendritic cells (MDDCs). A broad selection of commercially available cationic compounds was tested to prepare liposomes containing Ag85B-ESAT6-Rv2034 (AER) fusion protein antigen. 1,2-Dioleoyl-sn­glycero-3-ethylphosphocholine (EPC)-based liposomes exhibited the most advantageous activation profile in MDDCs as assessed by cell surface activation markers, cellular uptake, antigen-specific T-cell activation, cytokine production, and cellular viability. The addition of cholesterol to 20 mol% improved the performance of the tested formulations compared to those without it; however, when its concentration was doubled there was no further benefit, resulting in reduced cell viability. This study provides new insights into the role of cationic lipids and cholesterol in liposomal subunit vaccines.


Subject(s)
Tuberculosis Vaccines , Vaccines , Humans , Animals , Mice , Tuberculosis Vaccines/chemistry , Liposomes/chemistry , Adjuvants, Immunologic/chemistry , Vaccines, Subunit , Lipids/chemistry , Cholesterol/chemistry , Mice, Inbred C57BL
2.
Life Sci ; 282: 119806, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34252419

ABSTRACT

PURPOSE: Tuberculosis, a cost and life threatening disease, was being subjected for improving vaccine strategies beyond BCG. Thus, a novel particulate delivery system using alginate-coated chitosan nanoparticles including PPE17 protein and CpG were administered through intranasal (IN) and subcutaneous (SC) routes. METHODS: The encapsulated nanoparticles were first characterized for size, surface charge, encapsulation efficiency and in vitro release of PPE17 antigen. The nanoparticles were then administered intranasal and subcutaneously to evaluate the induction of systemic and/or mucosal immune responses in mice. RESULTS: According to our result, the mean size of nanoparticles was measured about 427 nm, and exhibited a negative zeta potential of -37 mV. Following subcutaneous and intranasal administration, the results from cytokines assay showed that an increasing in the level of IFN-γ, and adversely a decrease in the level of IL-4 (presumptive Th1 biased immune response) was happened and also a notable elicitation in IL-17 cytokine was observed. CONCLUSION: In conclusion, our study demonstrated that alginate-coated chitosan nanoparticles showed to be an effective way to improve BCG efficiency as booster strategy for subcutaneous vaccine, and also can induce strong immune responses as prime strategy through intranasal vaccination.


Subject(s)
Antigens, Bacterial , Drug Carriers , Nanoparticles , Th1 Cells/immunology , Tuberculosis Vaccines , Tuberculosis/immunology , Administration, Intranasal , Alginates/chemistry , Alginates/pharmacology , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/pharmacology , Chitosan/chemistry , Chitosan/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacology , Injections, Subcutaneous , Male , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Th1 Cells/pathology , Tuberculosis/prevention & control , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/pharmacology
3.
Front Immunol ; 12: 666290, 2021.
Article in English | MEDLINE | ID: mdl-33981313

ABSTRACT

Background: Tuberculosis (TB) is still a global infectious disease that seriously threatens human beings. The only licensed TB vaccine Bacille Calmette-Guérin (BCG)'s protective efficacy varies significantly among populations and regions. It is very urgent to develop more effective vaccines. Methods: In this study, eleven candidate proteins of Mycobacterium tuberculosis were selected to predict peptides with high-affinity binding capacity for the HLA-DRB1*01:01 molecule. The immunodominant peptides were identified with the enzyme-linked immunospot assay (ELISPOT) and linked in silico to result in a novel polypeptide vaccine in Escherichia coli cells. The vaccine's protective efficacy was evaluated in humanized and wild-type C57BL/6 mice. The potential immune protective mechanisms were explored with Enzyme-linked Immunosorbent Assay (ELISA), flow cytometry, and ELISPOT. Results: Six immunodominant peptides screened from 50 predicted peptides were used to construct a new polypeptide vaccine named MP3RT. After challenge with M. tuberculosis, the colony-forming units (CFUs), lung lesion area, and the number of inflammatory cells in humanized mice rather than wild-type mice vaccinated with MP3RT were significantly lower than these in mice immunized with PBS. The humanized mice vaccinated with MP3RT revealed significant increases in IFN-γ cytokine production, IFN-γ+ T lymphocytes, CD3+IFN-γ+ T lymphocytes, and the MP3RT-specific IgG antibody. Conclusions: Taken together, MP3RT is a promising peptides-based TB vaccine characterized by inducing high levels of IFN-γ and CD3+IFN-γ+ T lymphocytes in humanized mice. These new findings will lay a foundation for the development of peptides-based vaccines against TB.


Subject(s)
Mycobacterium tuberculosis/immunology , Peptides/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Disease Models, Animal , Humans , Immunodominant Epitopes/chemistry , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Interferon-gamma/immunology , Lymphocytes/immunology , Mice , Mice, Transgenic , Peptides/administration & dosage , Peptides/chemistry , Peptides/genetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/genetics , Vaccination , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/chemistry , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
4.
Eur J Pharm Biopharm ; 163: 23-37, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33753213

ABSTRACT

Spray drying is a technique that can be used to stabilize biopharmaceuticals, such as vaccines, within dry particles. Compared to liquid pharmaceutical products, dry powder has the potential to reduce costs associated with refrigerated storage and transportation. In this study, spray drying was investigated for processing an adjuvanted tuberculosis subunit vaccine, formulated as an oil-in-water nanoemulsion, into a dry powder composed of microparticles. Applying in-silico approaches to the development of formulation and processing conditions, successful encapsulation of the adjuvanted vaccine within amorphous microparticles was achieved in only one iteration, with high retention (>90%) of both the antigen and adjuvant system. Moisture-controlled stability studies on the powder were conducted over 26 months at temperatures up to 40 °C. Results showed that the powder was physically stable after 26 months of storage for all tested temperatures. Adjuvant system integrity was maintained at temperatures up to 25 °C after 26 months and after one month of storage at 40 °C. The spray-dried product demonstrated improved antigen thermostability when stored above refrigerated temperatures as compared to the liquid product. These results demonstrate the feasibility of spray drying as a method of encapsulating and stabilizing an adjuvanted vaccine.


Subject(s)
Adjuvants, Immunologic/chemistry , Drug Compounding/methods , Spray Drying , Tuberculosis Vaccines/chemistry , Tuberculosis/prevention & control , Adjuvants, Immunologic/administration & dosage , Chemistry, Pharmaceutical , Drug Stability , Drug Storage , Emulsions , Excipients , Humans , Nanoparticles/chemistry , Particle Size , Powders , Tuberculosis Vaccines/administration & dosage , Vaccines, Subunit/administration & dosage
5.
Sci Rep ; 11(1): 1249, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441913

ABSTRACT

Tuberculosis (TB) kills more individuals in the world than any other disease, and a threat made direr by the coverage of drug-resistant strains of Mycobacterium tuberculosis (Mtb). Bacillus Calmette-Guérin (BCG) is the single TB vaccine licensed for use in human beings and effectively protects infants and children against severe military and meningeal TB. We applied advanced computational techniques to develop a universal TB vaccine. In the current study, we select the very conserved, experimentally confirmed Mtb antigens, including Rv2608, Rv2684, Rv3804c (Ag85A), and Rv0125 (Mtb32A) to design a novel multi-epitope subunit vaccine. By using the Immune Epitopes Database (IEDB), we predicted different B-cell and T-cell epitopes. An adjuvant (Griselimycin) was also added to vaccine construct to improve its immunogenicity. Bioinformatics tools were used to predict, refined, and validate the 3D structure and then docked with toll-like-receptor (TLR-3) using different servers. The constructed vaccine was used for further processing based on allergenicity, antigenicity, solubility, different physiochemical properties, and molecular docking scores. The in silico immune simulation results showed significant response for immune cells. For successful expression of the vaccine in E. coli, in-silico cloning and codon optimization were performed. This research also sets out a good signal for the design of a peptide-based tuberculosis vaccine. In conclusion, our findings show that the known multi-epitope vaccine may activate humoral and cellular immune responses and maybe a possible tuberculosis vaccine candidate. Therefore, more experimental validations should be exposed to it.


Subject(s)
Epitopes, T-Lymphocyte , Molecular Docking Simulation , Mycobacterium tuberculosis , Tuberculosis Vaccines , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Humans , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Tuberculosis/prevention & control , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/genetics , Tuberculosis Vaccines/immunology , Vaccines, Subunit/chemistry , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccinology
6.
Front Immunol ; 11: 575504, 2020.
Article in English | MEDLINE | ID: mdl-33117374

ABSTRACT

Bacillus Calmette-Guerin (BCG) is the only licensed vaccine to prevent children from tuberculosis (TB), whereas it cannot provide effective protection for adults. Our previous work showed a novel vaccine candidate, liposomal adjuvant DMT emulsified with a multistage antigen CMFO, could protect mice against primary progressive TB, latency, and reactivation. To develop a more effective vaccine against adult TB, we aimed to further understand the role of pattern recognition receptor (PRR) agonists monophosphoryl lipid A (MPLA) and trehalose-6,6'-dibehenate (TDB) of the liposomal adjuvant DMT in the CMFO subunit vaccine-induced protection. Using C57BL/6 mouse models, the current study prepared different dimethyldioctadecylammonium (DDA)-based liposomal adjuvants with MPLA, TDB, or both (DMT), and then compared the immunogenicity and the protective efficacy among different liposomal adjuvanted CMFO subunit vaccines. Our study demonstrated that CMFO/DMT provided stronger and longer-lasting protective efficacy than the CMFO emulsified with adjuvants DDA or DDA/TDB. In addition, DDA/MPLA adjuvanted CMFO conferred a comparable protection in the lung as CMFO/DMT did. Higher levels of IFN-γ, IL-2, TNF-α, and IL-17A secreted by splenocytes were related with a more powerful and durable protection induced by CMFO/DMT through a putative synergistic effect of both MPLA and TDB via binding to TLR4 and Mincle. IL-2+ CD4+ T cells, especially IL-2+ CD4+ TCM cells, in the lung after infection were significantly associated with the vaccine-induced protection, whereas stronger IL-10 response and lower IL-2+ CD4+ T cells also contributed to the inferior protection of the DDA/TDB adjuvanted CMFO subunit vaccine. Given their crucial roles in vaccine-induced protection, combinational different PRR agonists in adjuvant formulation represent a promising strategy for the development of next-generation TB vaccine.


Subject(s)
Adjuvants, Immunologic/pharmacology , Glycolipids/pharmacology , Immunogenicity, Vaccine , Lipid A/analogs & derivatives , Lung/drug effects , Mycobacterium tuberculosis/pathogenicity , Quaternary Ammonium Compounds/pharmacology , Receptors, Pattern Recognition/agonists , Tuberculosis Vaccines/pharmacology , Tuberculosis, Pulmonary/prevention & control , Adjuvants, Immunologic/chemistry , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/microbiology , Cytokines/metabolism , Disease Models, Animal , Drug Compounding , Female , Glycolipids/chemistry , Host-Pathogen Interactions , Lipid A/chemistry , Lipid A/pharmacology , Liposomes , Lung/immunology , Lung/metabolism , Lung/microbiology , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Quaternary Ammonium Compounds/chemistry , Receptors, Pattern Recognition/metabolism , Time Factors , Tuberculosis Vaccines/chemistry , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Pulmonary/microbiology , Vaccination , Vaccines, Subunit/chemistry , Vaccines, Subunit/pharmacology , Virulence
7.
Med Microbiol Immunol ; 209(2): 163-176, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32020284

ABSTRACT

A major roadblock in the development of novel vaccines is the formulation and delivery of the antigen. Liposomes composed of a dimethyldioctadecylammonium (DDA) backbone and the adjuvant trehalose-6-6-dibehenate (TDB, termed "cationic adjuvant formulation (CAF01)", promote immunogenicity and protective efficacy of vaccines, most notably against infection with Mycobacterium tuberculosis. Specifically, the multicomponent antigen H56 delivered by CAF01 protects against tuberculosis in mice. Here we investigated whether the inclusion of immune-modulatory adjuvants into CAF01 modulates the immunogenicity of H56/CAF01 in vitro and in vivo. Based on our recent findings we selected the active sequence of the mycobacterial 19 kDa lipoprotein, Pam3Cys, which interacts with Toll like receptor 2 to induce an antimicrobial pathway. H56/CAF01-Pam3Cys liposomes were characterized for Pam3Cys incorporation, size, toxicity and activation of primary human macrophages. Macrophages efficiently take up H56/CAF01-Pam3Cys and trigger the release of significantly higher levels of TNF, IL-12 and IL-10 than H56/CAF01 alone. To evaluate the immunogenicity in vivo, we immunized mice with H56/CAF01-Pam3Cys and measured the release of IFN-γ and IL-17A by lymph node cells and spleen cells. While the antigen-specific production of IFN-γ was reduced by inclusion of Pam3Cys into H56/CAF01, the levels of IL-17A remained unchanged. In agreement with this finding, the concentration of the IFN-γ-associated IgG2a antibodies in the serum was lower than in H56/CAF01 immunized animals. These results provide proof of concept that Toll like-receptor agonist can be included into liposomes to modulate immune responses. The discordant results between the in vitro studies with human macrophages and in vivo studies in mice highlight the relevance and complexity of comparing immune responses in different species.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antigens, Bacterial/immunology , Lipoproteins/immunology , Toll-Like Receptors/agonists , Tuberculosis Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Bacterial/administration & dosage , Cells, Cultured , Cytokines/metabolism , Female , Humans , Immunomodulation , Liposomes/administration & dosage , Liposomes/chemistry , Liposomes/immunology , Liposomes/toxicity , Macrophages/immunology , Mice , Mycobacterium tuberculosis/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/chemistry , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology
8.
Mol Pharm ; 16(11): 4725-4737, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31539263

ABSTRACT

Designing effective and safe tuberculosis (TB) subunit vaccines for inhalation requires identification of appropriate antigens and adjuvants and definition of the specific areas to target in the lungs. Magnetic resonance imaging (MRI) enables high spatial resolution, but real-time anatomical and functional MRI of lungs is challenging. Here, we describe the design of a novel gadoteridol-loaded cationic adjuvant formulation 01 (CAF01) for MRI-guided vaccine delivery of the clinically tested TB subunit vaccine candidate H56/CAF01. Gadoteridol-loaded CAF01 liposomes were engineered by using a quality-by-design approach to (i) increase the mechanistic understanding of formulation factors governing the loading of gadoteridol and (ii) maximize the loading of gadoteridol in CAF01, which was confirmed by cryotransmission electron microscopy. The encapsulation efficiency and loading of gadoteridol were highly dependent on the buffer pH due to strong attractive electrostatic interactions between gadoteridol and the cationic lipid component. Optimal gadoteridol loading of CAF01 liposomes showed good in vivo stability and safety upon intrapulmonary administration into mice while generating 1.5-fold MRI signal enhancement associated with approximately 30% T1 relaxation change. This formulation principle and imaging approach can potentially be used for other mucosal nanoparticle-based formulations, species, and lung pathologies, which can readily be translated for clinical use.


Subject(s)
Cations/chemistry , Heterocyclic Compounds/administration & dosage , Heterocyclic Compounds/chemistry , Liposomes/chemistry , Lung/drug effects , Organometallic Compounds/administration & dosage , Organometallic Compounds/chemistry , Adjuvants, Immunologic/chemistry , Adjuvants, Pharmaceutic , Animals , Chemistry, Pharmaceutical/methods , Female , Gadolinium/administration & dosage , Gadolinium/chemistry , Lipids/chemistry , Magnetic Resonance Imaging/methods , Mice , Mice, Inbred BALB C , Nanoparticles/chemistry , Tuberculosis/drug therapy , Tuberculosis Vaccines/chemistry , Vaccines, Subunit/chemistry
9.
Sci Rep ; 9(1): 11409, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31391509

ABSTRACT

There is an urgent need for the development of vaccine thermostabilisation methodologies as the maintenance of a continuous and reliable cold chain remains a major hurdle to the global distribution of safe and effective vaccines. Ensilication, a method that encases proteins in a resistant silica cage has been shown to physically prevent the thermal denaturation of a number of model proteins. In this study we investigate the utility of this promising approach in improving the thermal stability of antigens and vaccine conjugates highly relevant to the development of candidate tuberculosis vaccines, including antigen 85b conjugated with the Staphylococcus aureus-protein based adjuvant Sbi. Here we analyse the sensitivity of these constructs to thermal denaturation and demonstrate for the first time the benefits of ensilication in conferring these vaccine-relevant proteins with protection against temperature-induced loss of structure and function without the need for refrigeration. Our results reveal the potential of ensilication in facilitating the storage and transport of vaccines at ambient temperatures in the future and therefore in delivering life-saving vaccines globally, and in particular to remote areas of developing countries where disease rates are often highest.


Subject(s)
Acyltransferases/chemistry , Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Carrier Proteins/chemistry , Silicon Dioxide/chemistry , Temperature , Tuberculosis Vaccines/chemistry , Vaccines, Conjugate/chemistry , Drug Stability , Escherichia coli , Humans , Proteolysis , Serum/chemistry
10.
Biochem J ; 476(14): 1995-2016, 2019 07 18.
Article in English | MEDLINE | ID: mdl-31320388

ABSTRACT

Bacterial capsules have evolved to be at the forefront of the cell envelope, making them an essential element of bacterial biology. Efforts to understand the Mycobacterium tuberculosis (Mtb) capsule began more than 60 years ago, but the relatively recent development of mycobacterial genetics combined with improved chemical and immunological tools have revealed a more refined view of capsule molecular composition. A glycogen-like α-glucan is the major constituent of the capsule, with lower amounts of arabinomannan and mannan, proteins and lipids. The major Mtb capsular components mediate interactions with phagocytes that favor bacterial survival. Vaccination approaches targeting the mycobacterial capsule have proven successful in controlling bacterial replication. Although the Mtb capsule is composed of polysaccharides of relatively low complexity, the concept of antigenic variability associated with this structure has been suggested by some studies. Understanding how Mtb shapes its envelope during its life cycle is key to developing anti-infective strategies targeting this structure at the host-pathogen interface.


Subject(s)
Bacterial Capsules , Lipids , Mycobacterium tuberculosis , Polysaccharides, Bacterial , Tuberculosis Vaccines , Bacterial Capsules/chemistry , Bacterial Capsules/immunology , Bacterial Capsules/metabolism , Humans , Lipids/chemistry , Lipids/immunology , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/immunology , Polysaccharides, Bacterial/metabolism , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/immunology
11.
Semin Immunol ; 39: 4-13, 2018 10.
Article in English | MEDLINE | ID: mdl-30396811

ABSTRACT

The development of the CAF family adjuvant was initiated around 20 years ago when Statens Serum Institut was preparing its first generation protein based recombinant subunit vaccine against tuberculosis for clinical testing, but realized that there were no clinically relevant adjuvants available that would support the strong CMI response needed. Since then the aim for the adjuvant research at Statens Serum Institut has been to provide adjuvants with distinct immunogenicity profiles correlating with protection for any given infectious disease. Two of the adjuvants CAF01 and CAF09 are currently being evaluated in human clinical trials. The purpose of this review is to give an overview of the immunocorrelates of those CAF adjuvants furthest in development. We further aim at giving an overview of the mechanism of action of the CAF adjuvants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Glycolipids/pharmacology , Immunity, Cellular/drug effects , Immunogenicity, Vaccine , Lipid A/analogs & derivatives , Quaternary Ammonium Compounds/pharmacology , Tuberculosis, Pulmonary/prevention & control , Adjuvants, Immunologic/chemistry , Animals , Glycolipids/chemistry , Humans , Immunity, Humoral/drug effects , Lipid A/chemistry , Lipid A/pharmacology , Liposomes/administration & dosage , Liposomes/chemistry , Liposomes/immunology , Mice , Quaternary Ammonium Compounds/chemistry , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/microbiology , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/microbiology , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/microbiology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology
12.
PLoS One ; 13(7): e0201253, 2018.
Article in English | MEDLINE | ID: mdl-30063728

ABSTRACT

There is a need for the rational design of safe and effective vaccines to protect against chronic bacterial pathogens such as Mycobacterium tuberculosis and Mycobacterium avium subsp. paratuberculosis in a number of species. One of the main challenges for vaccine development is the lack of safe adjuvants that induce protective immune responses. Cationic Adjuvant Formulation 01 (CAF01)-an adjuvant based on trehalose dibehenate (TDB) and targeting the Mincle receptor-has entered human trials based on promising pre-clinical results in a number of species. However, in cattle CAF01 only induces weak systemic immune responses. In this study, we tested the ability of three pattern recognition receptors, either alone or in combination, to activate bovine monocytes and macrophages. We found that addition of the TLR3 agonist, polyinosinic:polycytidylic acid (Poly(I:C)) to either one of the Mincle receptor agonists, TDB or monomycoloyl glycerol (MMG), enhanced monocyte activation, and calves vaccinated with CAF09 containing MMG and Poly(I:C) had increased cell-mediated and humoral immune response compared to CAF01 vaccinated animals. In contrast to the highly reactogenic Montanide ISA 61 VG, CAF09-primed T cells maintained a higher frequency of polyfunctional CD4+ T cells (IFN-γ+ TNF-α+ IL-2+). In conclusion, CAF09 supports the development of antibodies along with a high-quality cell-mediated immune response and is a promising alternative to oil-in-water adjuvant in cattle and other ruminants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunologic Memory/drug effects , Lectins, C-Type/immunology , Mycobacterium avium subsp. paratuberculosis/immunology , Mycobacterium tuberculosis/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 3/immunology , Tuberculosis Vaccines/pharmacology , Adjuvants, Immunologic/chemistry , Animals , Cattle , Male , Paratuberculosis/immunology , Paratuberculosis/pathology , Paratuberculosis/prevention & control , T-Lymphocytes/pathology , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/immunology , Tuberculosis, Bovine/immunology , Tuberculosis, Bovine/pathology , Tuberculosis, Bovine/prevention & control
13.
J Pharm Biomed Anal ; 157: 235-243, 2018 Aug 05.
Article in English | MEDLINE | ID: mdl-29866391

ABSTRACT

Tuberculosis (TB) is one of the leading causes of death worldwide, making the development of effective TB vaccines a global priority. A TB vaccine consisting of a recombinant fusion protein, H4, combined with a novel synthetic cationic adjuvant, IC31®, is currently being developed. The H4 fusion protein consists of two immunogenic mycobacterial antigens, Ag85 B and TB10.4, and the IC31® adjuvant is a mixture of KLK, a leucine-rich peptide (KLKL5KLK), and the oligodeoxynucleotide ODN1a, a TLR9 ligand. However, efficient and robust methods for assessing these formulated components are lacking. Here, we developed and optimized phase analysis light scattering (PALS), electrical sensing zone (ESZ), and Raman, FTIR, and CD spectroscopy methods to characterize the H4-IC31 vaccine formulation. PALS-measured conductivity and zeta potential values could differentiate between the similarly sized particles of IC31® adjuvant and the H4-IC31 vaccine candidate and could thereby serve as a control during vaccine formulation. In addition, zeta potential is indicative of the adjuvant to antigen ratio which is the key in the immunomodulatory response of the vaccine. ESZ was used as an orthogonal method to measure IC31® and H4-IC31 particle sizes. Raman, FTIR, and CD spectroscopy revealed structural changes in H4 protein and IC31® adjuvant, inducing an increase in both the ß-sheet and random coil content as a result of adsorption. Furthermore, nanoDSF showed changes in the tertiary structure of H4 protein as a result of adjuvantation to IC31®. Our findings demonstrate the applicability of biophysical methods to characterize vaccine components in the final H4-IC31 drug product without the requirement for desorption.


Subject(s)
Tuberculosis Vaccines/chemistry , Adjuvants, Immunologic/chemistry , Chemistry, Pharmaceutical/methods , Oligodeoxyribonucleotides/chemistry , Particle Size , Recombinant Fusion Proteins/chemistry , Spectrum Analysis/methods , Tuberculosis/immunology , Tuberculosis Vaccines/immunology
14.
Vaccine ; 36(23): 3331-3339, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29699790

ABSTRACT

Liquid vaccine dosage forms have limited stability and require refrigeration during their manufacture, distribution and storage. In contrast, solid vaccine dosage forms, produced by for example spray drying, offer improved storage stability and reduced dependence on cold-chain facilities. This is advantageous for mass immunization campaigns for global public health threats, e.g., tuberculosis (TB), and offers cheaper vaccine distribution. The multistage subunit vaccine antigen H56, which is a fusion protein of the Mycobacterium tuberculosis (Mtb) antigens Ag85B, ESAT-6, and Rv2660, has been shown to confer protective efficacy against active TB before and after Mtb exposure in preclinical models, and it is currently undergoing clinical phase 2a testing. In several studies, including a recent study comparing multiple clinically relevant vaccine adjuvants, the T helper type 1 (Th1)/Th17-inducing adjuvant CAF01 was the most efficacious adjuvant for H56 to stimulate protective immunity against Mtb. With the long-term goal of designing a thermostable and self-administrable dry powder vaccine based on H56 and CAF01 for inhalation, we compared H56 spray-dried with CAF01 with the non-spray-dried H56/CAF01 vaccine with respect to their ability to induce systemic Th1, Th17 and humoral responses after subcutaneous immunization. Here we show that spray drying of the H56/CAF01 vaccine results in preserved antigenic epitope recognition and adjuvant activity of CAF01, and the spray-dried, reconstituted vaccine induces antigen-specific Th1, Th17 and humoral immune responses, which are comparable to those stimulated by the non-spray-dried H56/CAF01 vaccine. In addition, the spray-dried and reconstituted H56/CAF01 vaccine promotes similar polyfunctional CD4+ T-cell responses as the non-spray-dried vaccine. Thus, our study provides proof-of-concept that spray drying of the subunit vaccine H56/CAF01 preserves vaccine-induced humoral and cell-mediated immune responses. These results support our ongoing efforts to develop a thermostable, dry powder-based TB vaccine.


Subject(s)
Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/immunology , Vaccines, Subunit/immunology , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , CD4-Positive T-Lymphocytes/immunology , Dry Powder Inhalers , Female , Immunity, Humoral/drug effects , Immunologic Memory , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice, Inbred Strains , Powders , Th1 Cells/immunology , Th17 Cells/immunology , Tuberculosis Vaccines/chemistry , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/chemistry
15.
Front Immunol ; 9: 310, 2018.
Article in English | MEDLINE | ID: mdl-29535714

ABSTRACT

Despite the vaccine Mycobacterium bovis Bacillus Calmette-Guérin is used worldwide, tuberculosis (TB) remains the first killer among infectious diseases. An effective vaccine is urgently required. DNA vaccine has shown prophylactic as well as therapeutic effects against TB, while its weak immunogenicity hinders the application. As a strong inducer of Th1-biased immune response, DMT, consisting of dimethyldioctadecylammonium (DDA) and two pattern recognition receptor agonists monophosphoryl lipid A and trehalose 6,6'-dibehenate (TDB), was a newly developed liposomal adjuvant. To elucidate the action mechanism of DMT and improve immunological effects induced by DNA vaccine, a new recombinant eukaryotic expression plasmid pCMFO that secretes the fusion of four multistage antigens (Rv2875, Rv3044, Rv2073c, and Rv0577) of Mycobacterium tuberculosis was constructed. pCMFO/DDA and pCMFO/DMT complexes were then prepared and their physicochemical properties were analyzed. The immunogenicity and protection against M. tuberculosis infection in vaccinated C57BL/6 mice were compared. Formulation of DNA and two agonists into the DDA liposome decreased zeta potential but increased the stability of storage, which resulted in a slower and longer-lasting release of DNA from the DNA-DMT complex than the DNA-DDA liposome. Besides Th1-biased responses, pCMFO/DMT vaccinated mice elicited more significantly CFMO-specific IL2+ TCM cell responses in the spleen and provided an enhanced and persistent protection against M. tuberculosis aerosol infection, compared to pCMFO/DDA and pCMFO groups. Therefore, the adjuvant DMT can release DNA and agonists slowly, which might attribute to the improved protection of DMT adjuvanted vaccines. pCMFO/DMT, a very promising TB vaccine, warrants for further preclinical and clinical trials.


Subject(s)
Immunogenicity, Vaccine , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/prevention & control , Vaccines, DNA , Animals , Female , Liposomes , Mice , Mycobacterium tuberculosis/genetics , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/pharmacology , Tuberculosis, Pulmonary/genetics , Tuberculosis, Pulmonary/pathology , Vaccines, DNA/chemistry , Vaccines, DNA/immunology , Vaccines, DNA/pharmacology
16.
Mol Immunol ; 97: 56-62, 2018 05.
Article in English | MEDLINE | ID: mdl-29567319

ABSTRACT

Effective control of Mycobacterium tuberculosis is a global necessity. In 2015, tuberculosis (TB) caused more deaths than HIV. Considering the increasing prevalence of multi-drug resistant forms of M. tuberculosis, the need for effective TB vaccines becomes imperative. Currently, the only licensed TB vaccine is Bacillus Calmette-Guérin (BCG). Yet, BCG has many drawbacks limiting its efficacy and applicability. We applied advanced computational procedures to derive a universal TB vaccine and one targeting East Africa. Our approach selects an optimal set of highly conserved, experimentally validated epitopes, with high projected population coverage (PPC). Through rigorous data analysis, five different potential vaccine combinations were selected each with PPC above 80% for East Africa and above 90% for the World. Two potential vaccines only contained CD8+ epitopes, while the others included both CD4+ and CD8+ epitopes. Our prime vaccine candidate was a putative seven-epitope ensemble comprising: SRGWSLIKSVRLGNA, KPRIITLTMNPALDI, AAHKGLMNIALAISA, FPAGGSTGSL, MLLAVTVSL, QSSFYSDW and KMRCGAPRY, with a 97.4% global PPC and a 92.7% East African PPC.


Subject(s)
Drug Design , Epitope Mapping , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/chemical synthesis , Tuberculosis/prevention & control , Amino Acid Sequence , BCG Vaccine/chemistry , BCG Vaccine/immunology , Computational Biology , Computer Simulation , Epitope Mapping/methods , Epitopes , Humans , Mycobacterium tuberculosis/chemistry , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/therapeutic use
17.
PLoS One ; 13(3): e0194620, 2018.
Article in English | MEDLINE | ID: mdl-29554138

ABSTRACT

Tuberculosis places a staggering burden on human health globally. The new World Health Organisation End-TB Strategy has highlighted the urgent need for more effective TB vaccines to improve control of the disease. Protein-based subunit vaccines offer potential as safe and effective generators of protective immunity, and the use of particulate vaccine formulation and delivery by the pulmonary route may enhance local immunogenicity. In this study, novel particulate subunit vaccines were developed utilising biodegradable poly(lactic-co-glycolic acid) (PLGA) slow-release particles as carriers for the Mycobacterium tuberculosis lipoprotein MPT83, together with the adjuvants trehalose-dibehenate (TDB) or Monophosphoryl lipid A (MPL). Following delivery by the pulmonary or subcutaneous routes, the immunogenicity and protective efficacy of these vaccines were assessed in a murine model of M. tuberculosis infection. When delivered peripherally, these vaccines induced modest, antigen-specific Th1 and Th17 responses, but strong anti-MPT83 antibody responses. Mucosal delivery of the PLGA(MPT83) vaccine, with or without TDB, increased antigen-specific Th17 responses in the lungs, however, PLGA-encapsulated vaccines did not provide protection against M. tuberculosis challenge. By contrast, peripheral delivery of DDA liposomes containing MPT83 and TDB or MPL, stimulated both Th1 and Th17 responses and generated protection against M. tuberculosis challenge. Therefore, PLGA-formulated vaccines primarily stimulate strong humoral immunity, or Th17 responses if used mucosally, and may be a suitable carrier for vaccines against extracellular pathogens. This study emphasises the critical nature of the vaccine carrier, adjuvant and route of delivery for optimising vaccine efficacy against TB.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunity, Humoral/drug effects , Lactic Acid/pharmacology , Mycobacterium tuberculosis/immunology , Polyglycolic Acid/pharmacology , Th17 Cells/drug effects , Tuberculosis Vaccines/immunology , Tuberculosis/immunology , Animals , Female , Lymphocyte Count , Mice , Mice, Inbred C57BL , Polylactic Acid-Polyglycolic Acid Copolymer , Th17 Cells/immunology , Tuberculosis/prevention & control , Tuberculosis Vaccines/chemistry , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacology
18.
Article in English | MEDLINE | ID: mdl-29085809

ABSTRACT

Induction of specific humoral and cellular immunity in the lung airways is proposed to be critical for vaccine protection against Mycobacterium tuberculosis (M. tb). To facilitate airway delivery and antigen targeting to the antigen presenting cells in the alveoli, we employed mannosylated chitosan (MCS) to formulate a multi-T-epitope DNA vaccine, pPES, as an intranasal TB vaccine. MCS-DNA nanoparticles appeared spherical with the average particle sizes as 400 nm. HSP65-specific bronchoalveolar lavage fluid SIgA level was significantly elevated by 4 doses of MCS-pPES intranasal immunization as compared to chitosan (CS)-DNA and BCG vaccine. I.n. immunization with MCS-DNA induced a modest peptide-specific Th1(IFN-γ, TNF-α, and IL-2) response in the spleen, while a potent poly-functional CD4+ T response that largely produced TNF-α and IFN-γ, as well as IL-2 in the lung, qualitatively better than that induced by CS-DNA and BCG vaccination. Such response by i.n. immunization with MCS-DNA provided improved protection in the lung against airway Mycobacterial bovis BCG challenge over i.n. CS-DNA and DNA, that is comparable to protection achieved by s.c. BCG vaccination. This enhanced protection was correlated with much greater accessibility of DNA particles to the alveolar macrophages in the lung mediated by man-chitosan. Thus, man-chitosan TB vaccine represents a promising vaccine platform capable of eliciting robust multi-functional T response in the lung mucus and achieving enhanced mucosal immune protection against pulmonary TB.


Subject(s)
Immunity, Cellular/immunology , Immunoglobulin A/immunology , Mycobacterium tuberculosis/immunology , Pulmonary Alveoli/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis, Pulmonary/immunology , Administration, Intranasal , Animals , BCG Vaccine/immunology , Bronchoalveolar Lavage Fluid/immunology , Chitosan/chemistry , Cytokines/immunology , Disease Models, Animal , Epitopes, T-Lymphocyte/chemistry , Female , Immunoglobulin A/analysis , Mannose/chemistry , Mice , Mice, Inbred C57BL , Mycobacterium bovis/immunology , Nanoparticles/chemistry , Tuberculosis Vaccines/chemistry , Tuberculosis, Pulmonary/therapy , Vaccination/methods , Vaccines, DNA/administration & dosage , Vaccines, DNA/chemistry
19.
Vaccine ; 35(38): 5123-5130, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28818565

ABSTRACT

The main challenge for vaccine development or improvement is the lack of safe adjuvants or immunostimulants that induce protective immune responses and can be used for mucosal immunization, which is a highly desirable strategy for vaccination against infectious diseases acquired by oral or intranasal routes. One promising alternative is the use of biodegradable and biocompatible polymeric microparticles. Recently, we developed an immobilization and delivery system with starch microparticles (SMPs) and a starch-binding domain (SBDtag) suitable for the mucosal administration of antigens and the induction of antigen-specific immune responses. Here, we explore the immunostimulant and reinforcing potential of the system using BALB/c mice with progressive pulmonary tuberculosis (PPT). The heat shock protein alpha-crystallin from Mycobacterium tuberculosis immobilized on SMPs (µAcr-SBDtag) or SMPs alone were administered nasally as boosters to BCG-vaccinated mice without any extra adjuvant. The mice were challenged intratracheally with either moderately virulent or highly virulent M. tuberculosis strains. Our results showed that the administration of either the immobilized antigen or SMPs asa booster for the BCG vaccination induced a significant reduction of bacterial loads in the lungs of mice, even more than in mice that received the BCG vaccination alone. Since no difference was observed in pulmonary bacillary burdens between the two reinforced groups, the obtained effect was most likely primarily caused by the starch. As determined by histological study, the administration of boosters did not contribute to the progress of pneumonia, which diminishes the safety concerns related to the administration of SMPs intranasally. Taken together, our findings suggest that this system may be considered asa new carbohydrate-based adjuvant suitable for mucosal vaccines against tuberculosis and other infectious diseases, and more generally, they highlight the potential of particulate α-glucans as immune response modifiers.


Subject(s)
Adjuvants, Immunologic/therapeutic use , BCG Vaccine/therapeutic use , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Starch/chemistry , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , BCG Vaccine/chemistry , BCG Vaccine/immunology , Immunity, Mucosal/immunology , Male , Mice , Mice, Inbred BALB C , Mycobacterium bovis/immunology , Mycobacterium bovis/pathogenicity , Tuberculosis/immunology , Tuberculosis/prevention & control , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/therapeutic use
20.
Talanta ; 175: 273-279, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28841990

ABSTRACT

Analysis of proteinogenic vaccine antigens in a quality control environment requires an accurate, precise, and reliable method for protein separation and quantitation. While having multiple advantages over the classical SDS-PAGE, capillary gel electrophoresis (CGE) has not yet become a standard tool in vaccine antigen analysis. Here we report on development of a CGE-based method for quantitative analysis of a tuberculosis vaccine fusion antigen protein, H4, currently in clinical trials. We demonstrate that our method can monitor antigen purity and relative quantity with greater precision and accuracy versus SDS-PAGE. In addition, due to use of direct light-absorbance detection, the CGE method is suitable for absolute quantitation, an application for which SDS-PAGE is limited due to the need for staining and limited dynamic range of detection. To further improve the performance of our quantitation method, we introduced Bovine Serum Albumin (BSA) as an injection standard to correct for signal variance associated with the injected sample volume. We found that, for our specific application, BSA was more appropriate as an injection standard versus one provided in a commercial kit, in terms of precision and accuracy for quantitation of H4. In addition to providing better method performance versus SDS-PAGE, CGE is also faster and less resource-intensive. We conclude that CGE should be considered as a replacement for traditional SDS-PAGE methods for vaccine antigen quantitation in a quality-control environment.


Subject(s)
Antigens, Bacterial/chemistry , Electrophoresis, Capillary/methods , Mycobacterium tuberculosis/chemistry , Tuberculosis Vaccines/chemistry , Antigens, Bacterial/immunology , Electrophoresis, Polyacrylamide Gel , Humans , Mycobacterium tuberculosis/immunology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/prevention & control , Tuberculosis Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...