Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters

Publication year range
1.
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
2.
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
3.
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
4.
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
5.
Biochim Biophys Acta ; 1860(4): 694-707, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26774644

ABSTRACT

BACKGROUND: In the molybdenum cofactor biosynthesis pathway, MoaA and MoaC catalyze the first step of transformation of GTP to cPMP. In M. tuberculosis H37Rv, three different genes (Rv3111, Rv0864 and Rv3324c) encode for MoaC homologs. Out of these three only MoaC1 (Rv3111) is secretory in nature. METHODS: We have characterized MoaC1 protein through biophysical, in-silico, and immunological techniques. RESULTS: We have characterized the conformation and thermodynamic stability of MoaC1, and have established its secretory nature by demonstrating the presence of anti-MoaC1 antibodies in human tuberculosis patients' sera. Further, MoaC1 elicited a dominant Th1 immune response in mice characterized by increased induction of IL-2 and IFN-γ. CONCLUSION: Integrating these results, we conclude that MoaC1 is a structured secretory protein capable of binding with GTP and eliciting induced immune response. GENERAL SIGNIFICANCE: This study would be useful for the development of vaccines against tuberculosis and to improve methods used for diagnosis of tuberculosis.


Subject(s)
Bacterial Proteins , Interferon-gamma/immunology , Interleukin-2/immunology , Mycobacterium tuberculosis , Th1 Cells/immunology , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Female , Genes, Bacterial , Humans , Male , Mice , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/immunology , Protein Stability , Sequence Homology, Amino Acid , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/genetics , Tuberculosis Vaccines/immunology
6.
Nature ; 473(7348): 463-9, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21614073

ABSTRACT

Acquired immune deficiency syndrome (AIDS), malaria and tuberculosis collectively cause more than five million deaths per year, but have nonetheless eluded conventional vaccine development; for this reason they represent one of the major global public health challenges as we enter the second decade of the twenty-first century. Recent trials have provided evidence that it is possible to develop vaccines that can prevent infection by human immunodeficiency virus (HIV) and malaria. Furthermore, advances in vaccinology, including novel adjuvants, prime-boost regimes and strategies for intracellular antigen presentation, have led to progress in developing a vaccine against tuberculosis. Here we discuss these advances and suggest that new tools such as systems biology and structure-based antigen design will lead to a deeper understanding of mechanisms of protection which, in turn, will lead to rational vaccine development. We also argue that new and innovative approaches to clinical trials will accelerate the availability of these vaccines.


Subject(s)
AIDS Vaccines , Malaria Vaccines , Tuberculosis Vaccines , AIDS Vaccines/chemistry , AIDS Vaccines/immunology , Animals , Antigens/chemistry , Antigens/immunology , Clinical Trials as Topic , HIV Infections/epidemiology , HIV Infections/immunology , HIV Infections/prevention & control , Humans , Malaria/epidemiology , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/chemistry , Malaria Vaccines/immunology , Systems Biology/trends , Tuberculosis/epidemiology , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/prevention & control , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/immunology
7.
Biotechnol Lett ; 39(8): 1159-1166, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28470626

ABSTRACT

OBJECTIVES: To investigate the potential of interleukin (IL)-15 as a novel adjuvant for Mycobacterium tuberculosis (Mtb) antigen 85A (Ag85A) vaccine. RESULTS: C57BL/6 mice were intramuscularly immunized three times with a plasmid expressing the Ag85A-IL-15 fusion protein (pcDNA3.1-Ag85A-IL-15), with the empty pcDNA3.1 vector and the pcDNA3.1-Ag85A as control. Mice vaccinated with pcDNA3.1-Ag85A-IL-15 generated more secretory IgA (sIgA) into their lung (209 ± 21 µg/ml) and acquired an enhanced serum IgG response to Ag85A. IgG2a/IgG1 ratios were upregulated, natural killer cell activity was augmented and Ag85A-specific splenic T cell proliferation was enhanced in these mice as well. Vaccination with pcDNA3.1-Ag85A-IL-15 promoted the polarization of CD4+ T cells towards a Th1 type in the spleen, and significantly upregulated the serum level of interferon (IFN)-γ (458 ± 98 pg/ml), a typical Th1 cytokine. IFN-γ-expressing CD8+ cells were also increased in the spleen after pcDNA3.1-Ag85A-IL-15 immunization. CONCLUSIONS: A superior immune type I response in mice vaccinated with plasmid Ag85A-IL-15 has been achieved.


Subject(s)
Acyltransferases/immunology , Adjuvants, Immunologic , Antigens, Bacterial/immunology , Interferon-gamma/immunology , Recombinant Fusion Proteins/immunology , Tuberculosis Vaccines/immunology , Vaccines, DNA/immunology , Acyltransferases/chemistry , Acyltransferases/genetics , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/genetics , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bronchoalveolar Lavage Fluid/cytology , Cells, Cultured , Cloning, Molecular , HEK293 Cells , Humans , Interferon-gamma/chemistry , Interferon-gamma/genetics , Mice , Mice, Inbred C57BL , Plasmids , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Spleen/cytology , Tuberculosis , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/genetics , Vaccines, DNA/chemistry , Vaccines, DNA/genetics
8.
Anal Bioanal Chem ; 408(22): 6123-32, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27372716

ABSTRACT

A capillary electrophoresis-mass spectrometry (CE-MS) method was developed for the characterization and integrity assessment of the Mycobacterium tuberculosis (MTB) antigens TB10.4 and Ag85B and their chemically produced glycoconjugates, which are glycovaccine candidates against tuberculosis (TB). In order to prevent protein adsorption to the inner capillary wall and to achieve efficient separation of the antigen proteoforms, a polyionic multilayer coating of polybrene-dextran sulfate-polybrene (PB-DS-PB) was used in combination with 1.5 M acetic acid as background electrolyte (BGE). Coupling of CE to high-resolution time-of-flight MS was achieved by a coaxial interface employing a sheath liquid of isopropanol-water (50:50, v/v) containing 0.1 % formic acid. The MTB antigens were exposed to experimental conditions used for chemical glycosylation (but no activated saccharide was added) in order to investigate their stability during glycovaccine production. CE-MS analysis revealed the presence of several closely related degradation products, including truncated, oxidized and conformational variants, which were assigned by accurate mass. Analysis of synthesized mannose conjugates of TB10.4 and Ag85B allowed the determination of the glycoform composition of the neo-glycoproteins next to the characterization of degradation products which were shown to be partly glycoconjugated. Moreover, the selectivity of CE-MS allowed specific detection of deamidated species (protein mass change of 1.0 Da only), indicating that chemical glycosylation increased susceptibility to deamidation. Overall, the results show that CE-MS represents a useful analytical tool for the detailed characterization and optimization of neo-glycoconjugate products. Graphical Abstract Flowchart illustrating Mycobacterium tuberculosis (MTB) antigen glycosylation, glycoconjugate variant and degradation product separation by capillary electrophoresis (CE) and their characterization by intact mass spectrometry (MS).


Subject(s)
Acyltransferases/chemistry , Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Electrophoresis, Capillary/methods , Glycoconjugates/chemistry , Mycobacterium tuberculosis/chemistry , Tuberculosis Vaccines/chemistry , Acyltransferases/immunology , Adsorption , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Glycoconjugates/immunology , Glycosylation , Hexadimethrine Bromide/chemistry , Humans , Mass Spectrometry/methods , Models, Molecular , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis/prevention & control , Tuberculosis Vaccines/immunology
9.
Crit Rev Microbiol ; 40(2): 117-45, 2014 May.
Article in English | MEDLINE | ID: mdl-23445450

ABSTRACT

Tuberculosis (TB) is an air-born, transmissible disease, having an estimated 9.4 million new TB cases worldwide in 2009. Eventual control of this disease by developing a safe and efficient new vaccine able to detain its spread will have an enormous impact on public health policy. Selecting potential antigens to be included in a multi-epitope, minimal subunit-based, chemically-synthesized vaccine containing the minimum sequences needed for blocking mycobacterial interaction with host cells is a complex task due to the multiple mechanisms involved in M. tuberculosis infection and the mycobacterium's immune evasion mechanisms. Our methodology, described here takes into account a highly robust, specific, sensitive and functional approach to the search for potential epitopes to be included in an anti-TB vaccine; it has been based on identifying short mycobacterial protein fragments using synthetic peptides having high affinity interaction with alveolar epithelial cells (A549) and monocyte-derived macrophages (U937) which are able to block the microorganism's entry to target cells in in vitro assays. This manuscript presents a review of the results obtained with some of the MTB H37Rv proteins studied to date, aimed at using these high activity binding peptides (HABPs) as platforms to be included in a minimal subunit-based, multiepitope, chemically-synthesized, antituberculosis vaccine.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Adhesion/drug effects , Bacterial Proteins/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Antigens, Bacterial/chemistry , Antigens, Bacterial/isolation & purification , Antigens, Bacterial/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Cell Line , Epithelial Cells/microbiology , Humans , Macrophages/microbiology , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/physiology , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/isolation & purification
10.
Scand J Infect Dis ; 46(3): 161-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24450840

ABSTRACT

It is estimated that there are approximately eight million new cases of active tuberculosis (TB) worldwide annually. There is only 1 vaccine available for prevention: bacillus Calmette-Guérin (BCG). This has variable efficacy and is only protective for certain extrapulmonary TB cases in children, therefore new strategies for the creation of novel vaccines have emerged. One of the promising approaches is the DNA vaccine, used as a direct vaccination or as a prime-boost vaccine. This review describes the experimental data obtained during the design of DNA vaccines for TB.


Subject(s)
Tuberculosis Vaccines/administration & dosage , Tuberculosis/prevention & control , Vaccines, DNA/administration & dosage , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Humans , Tuberculosis/immunology , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/genetics , Tuberculosis Vaccines/immunology , Vaccines, DNA/chemistry , Vaccines, DNA/genetics , Vaccines, DNA/immunology
11.
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
12.
Mol Pharm ; 10(2): 586-97, 2013 Feb 04.
Article in English | MEDLINE | ID: mdl-23301613

ABSTRACT

This study investigates both the physicochemical properties and immunogenicity of a genetically engineered elastin-like block corecombinamer (ELbcR) containing a major membrane protein sequence from Mycobacterium tuberculosis. The recombinant production of this ELbcR allows the production of large quantities of safe, antigenic particle-based constructs that directly and reversibly self-assemble into highly biocompatible, multivalent, monodisperse, and stable nanovesicles with a diameter of 55 nm from the same gene product using a highly efficient and cost-effective inverse transition cycling (ITC) procedure. The compositional complexity of these vesicles is retained after secondary processes such as endotoxin removal, sterilization, and lyophilization. An initial pro-chemotactic cytokine response (IL-1ß) followed by a pro-Th2/IL-5 response was observed in mice plasma following subcutaneous administration of the antigen-loaded nanovesicles in mice. This biphasic model of cytokine production was coupled with humoral isotype switching from IgM- to IgG-specific antibodies against the antigen, which was only observed in the presence of both the antigen and the polymer in the same construct and in the absence of additional adjuvants.


Subject(s)
Elastin/immunology , Immunologic Factors/immunology , Mycobacterium tuberculosis/immunology , Nanoparticles , Tuberculosis Vaccines/immunology , Immunologic Factors/chemistry , Tuberculosis Vaccines/chemistry
13.
Proc Natl Acad Sci U S A ; 106(7): 2301-6, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19188599

ABSTRACT

The development of a vaccine for tuberculosis requires a combination of antigens and adjuvants capable of inducing appropriate and long-lasting T cell immunity. We evaluated Mtb72F formulated in AS02A in the cynomolgus monkey model. The vaccine was immunogenic and caused no adverse reactions. When monkeys were immunized with bacillus Calmette-Guérin (BCG) and then boosted with Mtb72F in AS02A, protection superior to that afforded by using BCG alone was achieved, as measured by clinical parameters, pathology, and survival. We observed long-term survival and evidence of reversal of disease progression in monkeys immunized with the prime-boost regimen. Antigen-specific responses from protected monkeys receiving BCG and Mtb72F/AS02A had a distinctive cytokine profile characterized by an increased ratio between 3 Th1 cytokines, IFN-gamma, TNF, and IL-2 and an innate cytokine, IL-6. To our knowledge, this is an initial report of a vaccine capable of inducing long-term protection against tuberculosis in a nonhuman primate model, as determined by protection against severe disease and death, and by other clinical and histopathological parameters.


Subject(s)
Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Adjuvants, Immunologic/chemistry , Animals , Cytokines/metabolism , Disease Progression , Haplorhini , Immune System , Interferon-gamma/metabolism , Interleukin-6/metabolism , Macaca fascicularis , Mycobacterium tuberculosis/metabolism , Time Factors , Treatment Outcome , Tuberculosis/microbiology , Tuberculosis Vaccines/chemistry
14.
Crit Rev Microbiol ; 37(4): 349-57, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21919701

ABSTRACT

Heat shock proteins (HSPs) are conserved and ubiquitous house keeping entities that act as molecular chaperones, which protect the cell from damage during stress. One such HSP, the 16 kDa antigen, from Mycobacterium tuberculosis (Mtb) has received considerable attention due to its importance in tuberculosis latency and immunodominant property. In this article, we discuss about the potential role of 16 kDa antigen of Mtb in latency, its expression, regulation, and implication in host immune response. We also highlight the scope of employing 16 kDa in early diagnosis, development of vaccine and as a potential drug target.


Subject(s)
Antigens, Bacterial/physiology , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/immunology , Animals , Antigens, Bacterial/immunology , Humans , Tuberculosis/diagnosis , Tuberculosis/therapy , Tuberculosis Vaccines/chemistry , Tuberculosis Vaccines/pharmacology
15.
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
16.
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
17.
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
18.
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
19.
J Proteome Res ; 9(9): 4812-22, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20690665

ABSTRACT

Tuberculosis (TB) remains to be a major infectious disease throughout the world. However, the current vaccine for TB has variable protective efficacy, and there is no commercially available serodiagnostic test for this disease with acceptable sensitivity and specificity for routine laboratory use. One of the potential strategies in developing a new diagnostic method and in improving the TB vaccine involves the identification of novel antigenic candidates. This paper aims to identify systematically the novel antigenic proteins with the greatest potential as protective or diagnostic antigens by using the differential response of Mycobacterium tuberculosis proteins to serum from TB patients and healthy individuals. Approximately 87% of the open reading frames of M. tuberculosis were successfully cloned into IPTG-inducible expression vectors. The clone sets were expressed in Escherichia coli, purified under denatured conditions, and tested for antigenicity using a mixture of sera from 15 TB patients. Out of the 3480 proteins screened, 249 proteins had significant reactions with the serum samples. Among the 249 proteins, 20 proteins were identified as most reactive. Compared with the commercial test kits, 3 novel antigens from the top 20 proteins, namely, Rv1987, Rv3807c, and Rv3887c, provided better sensitivity and accuracy. These newly identified antigenic proteins may be used as candidates for serodiagnostic application and vaccine development. Overall, this study's findings may serve as an essential reference for developing new TB diagnostic methods and more effective tuberculosis vaccines.


Subject(s)
Antigens, Bacterial/isolation & purification , Bacterial Proteins/isolation & purification , Mycobacterium tuberculosis/chemistry , Proteomics/methods , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Base Sequence , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Molecular Sequence Data , Mycobacterium tuberculosis/immunology , Proteome/chemistry , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Sensitivity and Specificity , Serologic Tests , Tuberculosis Vaccines/chemistry
20.
Hum Vaccin ; 6(12): 1007-15, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21178394

ABSTRACT

Tuberculosis (TB) remains a major killer worldwide. The only available TB-vaccine, the nearly century-old Mycobacterium bovis BCG, has had only a limited effect on TB incidence. Therefore, developing new TB vaccines is a key priority, and the first new generation TB vaccines are now being tested in clinical trials. Here we describe the development and first testing in humans of a novel, wholly synthetic TB subunit vaccine. This vaccine has proven safe and highly immunogenic in all species in which it was tested, including mice, guinea pigs, non-human primates and humans. Most encouragingly, following vaccination in humans, strong IFN-γ responses persisted through at least 2½ years of follow-up, indicating induction of a substantial memory response by this new TB vaccine. These findings encourage further preclinical and clinical studies with TB subunit vaccines and cellular immunity-stimulating new adjuvants.


Subject(s)
Mycobacterium tuberculosis/immunology , Th1 Cells/immunology , Tuberculosis Vaccines/adverse effects , Tuberculosis Vaccines/immunology , Animals , Guinea Pigs , Humans , Interferon-gamma/metabolism , Mice , Time Factors , Tuberculosis Vaccines/chemistry , Vaccines, Subunit/adverse effects , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL