Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Int J Nanomedicine ; 13: 3689-3711, 2018.
Article in English | MEDLINE | ID: mdl-29983563

ABSTRACT

BACKGROUND: Adjuvants have the potential to increase the efficacy of protein-based vaccines but need to be maintained within specific temperature and storage conditions. Lyophilization can be used to increase the thermostability of protein pharmaceuticals; however, no marketed vaccine that contains an adjuvant is currently lyophilized, and lyophilization of oil-in-water nanoemulsion adjuvants presents a specific challenge. We have previously demonstrated the feasibility of lyophilizing a candidate adjuvanted protein vaccine against Mycobacterium tuberculosis (Mtb), ID93 + GLA-SE, and the subsequent improvement of thermostability; however, further development is required to prevent physicochemical changes and degradation of the TLR4 agonist glucopyranosyl lipid adjuvant formulated in an oil-in-water nanoemulsion (SE). MATERIALS AND METHODS: In this study, we took a systematic approach to the development of a thermostable product by first identifying compatible solution conditions and stabilizing excipients for both antigen and adjuvant. Next, we applied a design-of-experiments approach to identify stable lyophilized drug product formulations. RESULTS: We identified specific formulations that contain disaccharide or a combination of disaccharide and mannitol that can achieve substantially improved thermostability and maintain immunogenicity in a mouse model when tested in accelerated and real-time stability studies. CONCLUSION: These efforts will aid in the development of a platform formulation for use with other similar vaccines.


Subject(s)
Adjuvants, Immunologic/pharmacology , Emulsions/chemistry , Nanoparticles/chemistry , Temperature , Tuberculosis Vaccines/immunology , Animals , Antibody Formation , Chemistry, Pharmaceutical , Dynamic Light Scattering , Excipients , Female , Freeze Drying , Hydrogen-Ion Concentration , Immunity, Cellular , Lipids/chemistry , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Nephelometry and Turbidimetry , Particle Size , Tuberculosis/immunology , Tuberculosis/pathology
2.
Methods Mol Biol ; 1494: 313-320, 2017.
Article in English | MEDLINE | ID: mdl-27718205

ABSTRACT

Quantification of cytokine production by CD4 and CD8 T cells after in vitro recall stimulation with the immunizing antigen is a powerful approach to characterize the cellular immune responses to immunization. Here we describe three complementary methods for such quantification including flow cytometric analysis of cytokine production by intracellular staining, ELISpot determination of the numbers of cytokine-producing cells, and generation of secreted cytokines and chemokines in culture supernatants for analysis by ELISA and/or cytometric bead arrays.


Subject(s)
Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Enzyme-Linked Immunospot Assay/methods , Immunity, Cellular , Staining and Labeling/methods , Animals , Antigens/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Culture Media, Conditioned/chemistry , Mice
3.
J Control Release ; 244(Pt A): 98-107, 2016 12 28.
Article in English | MEDLINE | ID: mdl-27847326

ABSTRACT

For nearly a century, aluminum salts have been the most widely used vaccine adjuvant formulation, and have thus established a history of safety and efficacy. Nevertheless, for extremely challenging disease targets such as tuberculosis or HIV, the adjuvant activity of aluminum salts may not be potent enough to achieve protective efficacy. Adsorption of TLR ligands to aluminum salts facilitates enhanced adjuvant activity, such as in the human papilloma virus vaccine Cervarix®. However, some TLR ligands such as TLR7/8 agonist imidazoquinolines do not efficiently adsorb to aluminum salts. The present report describes a formulation approach to solving this challenge by developing a lipid-based nanosuspension of a synthetic TLR7/8 ligand (3M-052) that facilitates adsorption to aluminum oxyhydroxide via the structural properties of the helper lipid employed. In immunized mice, the aluminum oxyhydroxide-adsorbed formulation of 3M-052 enhanced antibody and TH1-type cellular immune responses to vaccine antigens for tuberculosis and HIV.


Subject(s)
Adjuvants, Immunologic/chemistry , Aluminum Hydroxide/chemistry , Aluminum Oxide/chemistry , Imidazoles/chemistry , Nanoparticles/chemistry , Quinolines/chemistry , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/metabolism , AIDS Vaccines/immunology , Adsorption , Animals , Drug Stability , Humans , Imidazoles/immunology , Immunity, Cellular , Immunity, Humoral , Ligands , Lipids/chemistry , Mice , Mice, Inbred C57BL , Particle Size , Quinolines/immunology , Surface Properties , Tuberculosis Vaccines/immunology
4.
Clin Vaccine Immunol ; 23(2): 137-47, 2016 02.
Article in English | MEDLINE | ID: mdl-26656121

ABSTRACT

Mycobacterium tuberculosis HN878 represents a virulent clinical strain from the W-Beijing family, which has been tested in small animal models in order to study its virulence and its induction of host immune responses following infection. This isolate causes death and extensive lung pathology in infected C57BL/6 mice, whereas lab-adapted strains, such as M. tuberculosis H37Rv, do not. The use of this clinically relevant isolate of M. tuberculosis increases the possibilities of assessing the long-lived efficacy of tuberculosis vaccines in a relatively inexpensive small animal model. This model will also allow for the use of knockout mouse strains to critically examine key immunological factors responsible for long-lived, vaccine-induced immunity in addition to vaccine-mediated prevention of pulmonary immunopathology. In this study, we show that the ID93/glucopyranosyl lipid adjuvant (GLA)-stable emulsion (SE) tuberculosis vaccine candidate, currently in human clinical trials, is able to elicit protection against M. tuberculosis HN878 by reducing the bacterial burden in the lung and spleen and by preventing the extensive lung pathology induced by this pathogen in C57BL/6 mice.


Subject(s)
Mycobacterium tuberculosis/immunology , Th1 Cells/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/immunology , Tuberculosis/immunology , Tuberculosis/prevention & control , Adjuvants, Immunologic , Animals , Bacterial Load , Clinical Trials as Topic , Disease Models, Animal , Humans , Lung/immunology , Lung/microbiology , Lung/pathology , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/isolation & purification , Spleen/microbiology , Tuberculosis/microbiology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
5.
Vaccine ; 33(48): 6570-8, 2015 Nov 27.
Article in English | MEDLINE | ID: mdl-26541135

ABSTRACT

Pulmonary tuberculosis (TB) remains one of the leading causes of infectious disease death despite widespread usage of the BCG vaccine. A number of new TB vaccines have moved into clinical evaluation to replace or boost the BCG vaccine including ID93+GLA-SE, an adjuvanted subunit vaccine. The vast majority of new TB vaccines in trials are delivered parenterally even though intranasal delivery can augment lung-resident immunity and protective efficacy in small animal models. Parenteral immunization with the adjuvanted subunit vaccine ID93+GLA-SE elicits robust TH1 immunity and protection against aerosolized Mycobacterium tuberculosis in mice and guinea pigs. Here we describe the immunogenicity and efficacy of this vaccine when delivered intranasally. Intranasal delivery switches the CD4 T cell response from a TH1 to a TH17 dominated tissue-resident response with increased frequencies of ID93-specific cells in both the lung tissue and at the lung surface. Surprisingly these changes do not affect the protective efficacy of ID93+GLA-SE. Unlike intramuscular immunization, ID93+GLA does not require the squalene-based oil-in-water emulsion SE to elicit protective CD4 T cells when delivered intranasally. Finally we demonstrate that TNF and the IL-17 receptor are dispensable for the efficacy of the intranasal vaccine suggesting an alternative mechanism of protection.


Subject(s)
Th1 Cells/immunology , Th17 Cells/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Adjuvants, Immunologic/administration & dosage , Administration, Mucosal , Animals , Antigens, Bacterial/immunology , BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Guinea Pigs , Humans , Mice , Mycobacterium tuberculosis/immunology , Peptide Fragments/immunology , Tumor Necrosis Factor-alpha/immunology
6.
J Immunol ; 195(7): 3190-7, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26297758

ABSTRACT

The discovery of new vaccines against infectious diseases and cancer requires the development of novel adjuvants with well-defined activities. The TLR4 agonist adjuvant GLA-SE elicits robust Th1 responses to a variety of vaccine Ags and is in clinical development for both infectious diseases and cancer. We demonstrate that immunization with a recombinant protein Ag and GLA-SE also induces granzyme A expression in CD4 T cells and produces cytolytic cells that can be detected in vivo. Surprisingly, these in vivo CTLs were CD4 T cells, not CD8 T cells, and this cytolytic activity was not dependent on granzyme A/B or perforin. Unlike previously reported CD4 CTLs, the transcription factors Tbet and Eomes were not necessary for their development. CTL activity was also independent of the Fas ligand-Fas, TRAIL-DR5, and canonical death pathways, indicating a novel mechanism of CTL activity. Rather, the in vivo CD4 CTL activity induced by vaccination required T cell expression of CD154 (CD40L) and target cell expression of CD40. Thus, vaccination with a TLR4 agonist adjuvant induces CD4 CTLs, which kill through a previously unknown CD154-dependent mechanism.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/immunology , CD40 Ligand/immunology , T-Lymphocytes, Cytotoxic/immunology , Adjuvants, Immunologic/pharmacology , Animals , CD40 Antigens/biosynthesis , CD40 Ligand/biosynthesis , Cytotoxins/immunology , Fas Ligand Protein/immunology , Granzymes/biosynthesis , Granzymes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , T-Box Domain Proteins/immunology , TNF-Related Apoptosis-Inducing Ligand/immunology , Th1 Cells/immunology , Vaccination
7.
J Infect Dis ; 212(3): 495-504, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25637347

ABSTRACT

BACKGROUND: Mycobacterium tuberculosis infects one third of the world's population and causes >8 million cases of tuberculosis annually. New vaccines are necessary to control the spread of tuberculosis. T cells, interferon γ (IFN-γ), and tumor necrosis factor (TNF) are necessary to control M. tuberculosis infection in both humans and unvaccinated experimental animal models. However, the immune responses necessary for vaccine efficacy against M. tuberculosis have not been defined. The multifunctional activity of T-helper type 1 (TH1) cells that simultaneously produce IFN-γ and TNF has been proposed as a candidate mechanism of vaccine efficacy. METHODS: We used a mouse model of T-cell transfer and aerosolized M. tuberculosis infection to assess the contributions of TNF, IFN-γ, and inducible nitric oxide synthase (iNOS) to vaccine efficacy. RESULTS: CD4(+) T cells were necessary and sufficient to transfer protection against aerosolized M. tuberculosis, but neither CD4(+) T cell-produced TNF nor host cell responsiveness to IFN-γ were necessary. Transfer of Tnf(-/-) CD4(+) T cells from vaccinated donors to Ifngr(-/-) recipients was also sufficient to confer protection. Activation of iNOS to produce reactive nitrogen species was not necessary for vaccine efficacy. CONCLUSIONS: Induction of TH1 cells that coexpress IFN-γ and TNF is not a requirement for vaccine efficacy against M. tuberculosis, despite these cytokines being essential for control of M. tuberculosis in nonvaccinated animals.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interferon-gamma/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/immunology , Tuberculosis/prevention & control , Tumor Necrosis Factor-alpha/immunology , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide Synthase Type II/immunology , Receptors, Interleukin-17/immunology , Tuberculosis Vaccines/pharmacology
8.
J Immunol ; 193(6): 2911-8, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25086172

ABSTRACT

Unlike most pathogens, many of the immunodominant epitopes from Mycobacterium tuberculosis are under purifying selection. This startling finding suggests that M. tuberculosis may gain an evolutionary advantage by focusing the human immune response against selected proteins. Although the implications of this to vaccine development are incompletely understood, it has been suggested that inducing strong Th1 responses against Ags that are only weakly recognized during natural infection may circumvent this evasion strategy and increase vaccine efficacy. To test the hypothesis that subdominant and/or weak M. tuberculosis Ags are viable vaccine candidates and to avoid complications because of differential immunodominance hierarchies in humans and experimental animals, we defined the immunodominance hierarchy of 84 recombinant M. tuberculosis proteins in experimentally infected mice. We then combined a subset of these dominant or subdominant Ags with a Th1 augmenting adjuvant, glucopyranosyl lipid adjuvant in stable emulsion, to assess their immunogenicity in M. tuberculosis-naive animals and protective efficacy as measured by a reduction in lung M. tuberculosis burden of infected animals after prophylactic vaccination. We observed little correlation between immunodominance during primary M. tuberculosis infection and vaccine efficacy, confirming the hypothesis that subdominant and weakly antigenic M. tuberculosis proteins are viable vaccine candidates. Finally, we developed two fusion proteins based on strongly protective subdominant fusion proteins. When paired with the glucopyranosyl lipid adjuvant in stable emulsion, these fusion proteins elicited robust Th1 responses and limited pulmonary M. tuberculosis for at least 6 wk postinfection with a single immunization. These findings expand the potential pool of M. tuberculosis proteins that can be considered as vaccine Ag candidates.


Subject(s)
Immunodominant Epitopes/immunology , Mycobacterium tuberculosis/immunology , Recombinant Fusion Proteins/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/immunology , Adjuvants, Immunologic , Animals , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Female , Mice , Th1 Cells/immunology , Tuberculosis, Pulmonary/prevention & control , Vaccination
9.
J Control Release ; 177: 20-6, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24382398

ABSTRACT

Next-generation rationally-designed vaccine adjuvants represent a significant breakthrough to enable development of vaccines against challenging diseases including tuberculosis, HIV, and malaria. New vaccine candidates often require maintenance of a cold-chain process to ensure long-term stability and separate vials to enable bedside mixing of antigen and adjuvant. This presents a significant financial and technological barrier to worldwide implementation of such vaccines. Herein we describe the development and characterization of a tuberculosis vaccine comprised of both antigen and adjuvant components that are stable in a single vial at sustained elevated temperatures. Further this vaccine retains the ability to elicit both antibody and TH1 responses against the vaccine antigen and protect against experimental challenge with Mycobacterium tuberculosis. These results represent a significant breakthrough in the development of vaccine candidates that can be implemented throughout the world without being hampered by the necessity of a continuous cold chain or separate adjuvant and antigen vials.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Bacterial/administration & dosage , Nanostructures/administration & dosage , Tuberculosis Vaccines/administration & dosage , Tuberculosis/prevention & control , Adjuvants, Immunologic/chemistry , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/chemistry , B-Lymphocytes/immunology , Bacterial Load , Emulsions , Female , Freeze Drying , Leukocyte Count , Lung/microbiology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/immunology , Nanostructures/chemistry , Spleen/microbiology , T-Lymphocytes/immunology , Temperature , Tuberculosis/immunology , Tuberculosis/microbiology , Tuberculosis Vaccines/chemistry
10.
PLoS One ; 9(1): e83884, 2014.
Article in English | MEDLINE | ID: mdl-24404140

ABSTRACT

With over eight million cases of tuberculosis each year there is a pressing need for the development of new vaccines against Mycobacterium tuberculosis. Subunit vaccines consisting of recombinant proteins are an attractive vaccine approach due to their inherent safety compared to attenuated live vaccines and the uniformity of manufacture. Addition of properly formulated TLR agonist-containing adjuvants to recombinant protein vaccines enhances the antigen-specific CD4(+) T cell response characterized by IFN-γ and TNF, both of which are critical for the control of TB. We have developed a clinical stage vaccine candidate consisting of a recombinant fusion protein ID93 adjuvanted with the TLR4 agonist GLA-SE. Here we examine whether ID93+GLA-SE can be improved by the addition of a second TLR agonist. Addition of CpG containing DNA to ID93+GLA-SE enhanced the magnitude of the multi-functional TH1 response against ID93 characterized by co-production of IFN-γ, TNF, and IL-2. Addition of CpG also improved the protective efficacy of ID93+GLA-SE. Finally we demonstrate that this adjuvant synergy between GLA and CpG is independent of TRIF signaling, whereas TRIF is necessary for the adjuvant activity of GLA-SE in the absence of CpG.


Subject(s)
Adjuvants, Immunologic , Mycobacterium tuberculosis/immunology , Toll-Like Receptors/agonists , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Antigens/immunology , Antimicrobial Cationic Peptides/immunology , Disease Models, Animal , Female , Mice , Mice, Knockout , Signal Transduction , Th1 Cells/immunology , Tuberculosis/immunology , Tuberculosis/metabolism , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...