Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Hum Vaccin Immunother ; 10(8): 2199-210, 2014.
Article in English | MEDLINE | ID: mdl-25424923

ABSTRACT

Bacille Calmette-Guérin (BCG), the only licensed vaccine for the prevention of tuberculosis (TB), provides only limited protection against certain forms of Mycobacterium tuberculosis (Mtb) infection. While infection with Mtb can be treated with antibiotics, the therapy is expensive, toxic, and requires several months for treatment. In addition, the emergence of drug resistant strains limits the impact of antibiotics and underlines the importance of developing a more effective vaccine to control this disease. Given that pulmonary TB is the most common form of the disease, a vaccine capable of inducing lung-resident immunity may be advantageous for combating this infection. New advances in pulmonary delivery make this route of vaccination feasible and affordable. Here, we evaluate the safety and immunogenicity of an aerosolized Ad35-based vaccine, AERAS-402, delivered to the lungs in nonhuman primates as part of a GLP acute and chronic toxicology and safety study. In this study, animals received three high doses (1 x 10(11) vp) of AERAS-402 by inhalation via a nebulizer at 1-week intervals. Aerosol delivery of AERAS-402 resulted in an increase in relative lung weights as well as microscopic findings in the lungs, mediastinal lymph nodes, bronchus-associated lymphatic tissue, and the naso-oropharynx that were consistent with the induction of an immune response during the acute phase. These findings resolved by the chronic phase and were considered to be non-adverse. Furthermore, we observed transient vaccine-specific immune responses in the peripheral blood as well as sustained high-level polyfunctional CD4(+) and CD8(+) T cell responses in the bronchoalveolar lavage fluid of vaccinated nonhuman primates. The data suggest that pulmonary delivery of Ad35-based vaccines can be safe and can induce potent lung-resident immunity.


Subject(s)
Aerosols/administration & dosage , Aerosols/adverse effects , Lung/immunology , T-Lymphocytes/immunology , Tuberculosis Vaccines/adverse effects , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Adenoviruses, Human/genetics , Administration, Inhalation , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Drug Carriers/administration & dosage , Female , Macaca mulatta , Male , Treatment Outcome , Tuberculosis/immunology , Tuberculosis Vaccines/administration & dosage , Vaccines, DNA
2.
Tuberculosis (Edinb) ; 94(6): 634-43, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25224753

ABSTRACT

Continuous high global tuberculosis (TB) mortality rates and variable vaccine efficacy of mycobacterium Bacille Calmette-Guérin (BCG) emphasize the need for improved vaccines and drugs against TB, which require clinically relevant animal models for evaluation. We infected a total of 24 Chinese rhesus macaques with varying doses (CFU of 25, 100 and 500) of Mycobacterium tuberculosis (M.tb) Erdman strain via bronchoscopy. Regardless of the M.tb doses, all animals were infected successfully with minor differences in clinical progression; as evidenced by clinical manifestations, laboratory analyses, bacterial burden in infected tissues and histopathology evaluations. Rhesus macaques of Chinese origin are highly susceptible to infection with M.tb Erdman strain and develop acute TB disease, which is similar to that in humans. Pathologically, Chinese rhesus macaques recapitulated the complete spectrum of granulomatous lesions seen in human TB disease. These data indicate that low-dose infection of rhesus macaques of Chinese origin is a suitable model for acute M.tb infection.


Subject(s)
Disease Models, Animal , Mycobacterium tuberculosis/pathogenicity , Tuberculosis, Pulmonary/microbiology , Acute Disease , Animals , Blood Sedimentation , Bronchoscopy , C-Reactive Protein/metabolism , Colony Count, Microbial , Disease Susceptibility , Female , Macaca mulatta , Male , Mycobacterium tuberculosis/classification , Radiography , Survival Analysis , Tuberculin Test , Tuberculosis, Pulmonary/diagnostic imaging , Tuberculosis, Pulmonary/pathology , Virulence
3.
J Immunol ; 193(4): 1799-811, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25024382

ABSTRACT

Development of a vaccine against pulmonary tuberculosis may require immunization strategies that induce a high frequency of Ag-specific CD4 and CD8 T cells in the lung. The nonhuman primate model is essential for testing such approaches because it has predictive value for how vaccines elicit responses in humans. In this study, we used an aerosol vaccination strategy to administer AERAS-402, a replication-defective recombinant adenovirus (rAd) type 35 expressing Mycobacterium tuberculosis Ags Ag85A, Ag85B, and TB10.4, in bacillus Calmette-Guérin (BCG)-primed or unprimed rhesus macaques. Immunization with BCG generated low purified protein derivative-specific CD4 T cell responses in blood and bronchoalveolar lavage. In contrast, aerosolized AERAS-402 alone or following BCG induced potent and stable Ag85A/b-specific CD4 and CD8 effector T cells in bronchoalveolar lavage that largely produced IFN-γ, as well as TNF and IL-2. Such responses induced by BCG, AERAS-402, or both failed to confer overall protection following challenge with 275 CFUs M. tuberculosis Erdman, although vaccine-induced responses associated with reduced pathology were observed in some animals. Anamnestic T cell responses to Ag85A/b were not detected in blood of immunized animals after challenge. Overall, our data suggest that a high M. tuberculosis challenge dose may be a critical factor in limiting vaccine efficacy in this model. However, the ability of aerosol rAd immunization to generate potent cellular immunity in the lung suggests that using different or more immunogens, alternative rAd serotypes with enhanced immunogenicity, and a physiological challenge dose may achieve protection against M. tuberculosis.


Subject(s)
Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Vaccination/methods , Vaccines, Synthetic/immunology , Acyltransferases/immunology , Administration, Inhalation , Animals , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Bronchoalveolar Lavage Fluid/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Lung/immunology , Lung/microbiology , Macaca mulatta , Male , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/virology , Tuberculosis Vaccines/administration & dosage , Tuberculosis, Pulmonary/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Vaccines, DNA , Vaccines, Synthetic/administration & dosage
4.
Mol Med ; 18: 647-58, 2012 May 09.
Article in English | MEDLINE | ID: mdl-22396020

ABSTRACT

To prevent the global spread of tuberculosis (TB) infection, a novel vaccine that triggers potent and long-lived immunity is urgently required. A plasmid-based vaccine has been developed to enhance activation of major histocompatibility complex (MHC) class I-restricted CD8⁺ cytolytic T cells using a recombinant Bacille Calmette-Guérin (rBCG) expressing a pore-forming toxin and the Mycobacterium tuberculosis (Mtb) antigens Ag85A, 85B and TB10.4 followed by a booster with a nonreplicating adenovirus 35 (rAd35) vaccine vector encoding the same Mtb antigens. Here, the capacity of the rBCG/rAd35 vaccine to induce protective and biologically relevant CD8⁺ T-cell responses in a nonhuman primate model of TB was investigated. After prime/boost immunizations and challenge with virulent Mtb in rhesus macaques, quantification of immune responses at the single-cell level in cryopreserved tissue specimen from infected organs was performed using in situ computerized image analysis as a technological platform. Significantly elevated levels of CD3⁺ and CD8⁺ T cells as well as cells expressing interleukin (IL)-7, perforin and granulysin were found in TB lung lesions and spleen from rBCG/rAd35-vaccinated animals compared with BCG/rAd35-vaccinated or unvaccinated animals. The local increase in CD8⁺ cytolytic T cells correlated with reduced expression of the Mtb antigen MPT64 and also with prolonged survival after the challenge. Our observations suggest that a protective immune response in rBCG/rAd35-vaccinated nonhuman primates was associated with enhanced MHC class I antigen presentation and activation of CD8⁺ effector T-cell responses at the local site of infection in Mtb-challenged animals.


Subject(s)
BCG Vaccine/immunology , Mycobacterium tuberculosis/immunology , T-Lymphocytes, Cytotoxic/immunology , Tuberculosis/immunology , Tuberculosis/prevention & control , Animals , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Collagen Type I/metabolism , Female , Immunization, Secondary , Interleukin-7/metabolism , Macaca mulatta , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Splenic/immunology , Tuberculosis, Splenic/metabolism , Vaccination
5.
Vaccine ; 30(12): 2098-108, 2012 Mar 09.
Article in English | MEDLINE | ID: mdl-22296955

ABSTRACT

BACKGROUND: Despite the availability of Bacille Calmette Guérin (BCG) vaccines, Mycobacterium tuberculosis currently infects billions of people and millions die annually from tuberculosis (TB) disease. New TB vaccines are urgently needed. METHODS: We studied the ability of AERAS-402, a recombinant, replication-deficient adenovirus type 35 expressing the protective M. tuberculosis antigens Ag85A, Ag85B, and TB10.4, to boost BCG immunity in an area of low TB endemicity. RESULTS: In volunteers primed with BCG 3 or 6 months prior to AERAS-402 boosting, significant CD4(+) and CD8(+) T cell responses were induced. Ag85-specific responses were more strongly boosted than TB10.4-specific responses. Frequencies of TB-specific CD8(+) T cells reached>50 fold higher than pre-AERAS boosting levels, remarkably higher than reported in any previous human TB vaccine trial. Multiparameter flow cytometric assays demonstrated that AERAS-402-boosted CD4(+) and CD8(+) T cells were multifunctional, producing multiple cytokines and other immune effector molecules. Furthermore, boosted T cells displayed lymphoproliferative capacity, and tetramer analyses confirmed that antigen-specific CD8(+) T cells were induced. BCG and AERAS-402 vaccinations given 3 and 6 months apart appeared equivalent. CONCLUSIONS: Our results indicate that AERAS-402 is a promising TB vaccine candidate that can significantly enhance both CD4(+) and CD8(+) TB-specific T cell responses after BCG priming. ClinicalTrials.gov Identifier: NCT01378312.


Subject(s)
Acyltransferases/immunology , Adenoviruses, Human/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Genetic Vectors , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Acyltransferases/genetics , Adult , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Cytokines/biosynthesis , Flow Cytometry , Human Experimentation , Humans , Mycobacterium tuberculosis/genetics , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/genetics
6.
Hum Vaccin Immunother ; 8(3): 371-83, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22327496

ABSTRACT

Preclinical studies evaluating plague vaccine candidates have demonstrated that the F1 and V protein antigens of Yersinia pestis confer protection against challenge from virulent strains. Live-attenuated ΔphoP/Q Salmonella typhimurium recombinants were constructed expressing either F1, V antigens, F1 and V antigens, or a F1-V fusion from Asd (+) balanced-lethal plasmids. To improve antigen delivery, genes encoding plague antigens were modified in order to localize antigens to specific bacterial cellular compartments which include cytoplasmic, outer membrane, or secreted. Candidate vaccine strains were evaluated for growth characteristics, full-length lipopolysaccharide (LPS), plasmid stability, and antigen expression in vitro. Plague vaccine candidate strains with favorable in vitro profiles were evaluated in murine or rabbit preclinical oral immunogenicity studies. Attenuated S. typhimurium strains expressing cytoplasmically localized F1-V and V antigen antigens were more immunogenic than strains that secreted or localized plague antigens to the outer membrane. In particular, S. typhimurium M020 and M023, which express Asd(+)-plasmid derived soluble F1-V and soluble V antigen, respectively, at high levels in the bacterial cell cytoplasm were found to induce the highest levels of plague-specific serum antibodies. To further evaluate balanced-lethal plasmid retention capacity, ΔphoP/Q S. typhimurium PurB(+) and GlnA(+) balanced-lethal plasmid systems harboring F1-V were compared with M020 in vitro and in BALB/c mice in a immunogenicity study. Although there was no detectable difference in plague antigen expression in vitro, S. typhimurium M020 was the most immunogenic plague antigen vector strain evaluated, inducing high-titer serum IgG antibodies specific against F1, V and F1-V.


Subject(s)
Bacterial Proteins/genetics , Gene Knockout Techniques , Plague Vaccine/immunology , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Male , Mice , Plague Vaccine/genetics , Pore Forming Cytotoxic Proteins/genetics , Pore Forming Cytotoxic Proteins/immunology , Pore Forming Cytotoxic Proteins/metabolism , Rabbits , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Virulence Factors/genetics , Virulence Factors/immunology , Virulence Factors/metabolism
7.
Clin Vaccine Immunol ; 18(1): 125-34, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21084459

ABSTRACT

High-tuberculosis (TB)-burden countries are located in sub-Saharan Africa. We examined the frequency of human leukocyte antigen (HLA) alleles, followed by recombinant expression of the most frequent HLA-A alleles, i.e., HLA-A*3001 and HLA-A*3002, to study differences in mycobacterial peptide presentation and CD8(+) T-cell recognition. We screened a peptide library (9-mer peptides with an 8-amino-acid overlap) for binding, affinity, and off-rate of the Mycobacterium tuberculosis-associated antigen TB10.4 and identified only three TB10.4 peptides with considerable binding to HLA-A*3001. In contrast, 22 peptides bound to HLA-A*3002. This reflects a marked difference in the binding preference between the two alleles, with A*3002 tolerating a more promiscuous peptide-binding pattern and A*3001 accommodating only a very selective peptide repertoire. Subsequent analysis of the affinity and off-rate of the binding peptides revealed a strong affinity (8 nM to 7 µM) and moderate off-rate (20 min to 3 h) for both alleles. Construction of HLA-A*3001 and HLA-A*3002 tetramers containing selected binding peptides from TB10.4, including a peptide which was shared among both alleles, QIMYNYPAM (TB10.4(3-11)), allowed us to enumerate epitope-specific T cells in HLA-A*3001- and HLA-A*3002-typed patients with active TB. HLA-A*3001 and HLA-A*3002 major histocompatibility complex-peptide complexes were recognized in individuals with active TB, irrespective of their homozygous HLA-A*3001 or HLA-A*3002 genetic background. The antigen-specific T cells exhibited the CD45RA(+) CCR7(+) precursor phenotype and the interleukin-7 receptor (CD127), which were different from the phenotype and receptor exhibited by the parental CD8(+) T-cell population.


Subject(s)
Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , HLA Antigens/immunology , HLA-A Antigens/immunology , Peptides/immunology , Amino Acid Sequence , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Binding Sites , CD8-Positive T-Lymphocytes/immunology , Epitopes/genetics , Epitopes/immunology , HLA Antigens/chemistry , HLA Antigens/genetics , HLA Antigens/metabolism , HLA-A Antigens/chemistry , HLA-A Antigens/genetics , HLA-A Antigens/metabolism , Humans , Molecular Sequence Data , Mycobacterium tuberculosis , Peptide Library , Peptides/chemistry , Peptides/metabolism , Protein Binding , South Africa , Tuberculosis, Pulmonary/immunology
8.
Immunology ; 131(1): 128-40, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20465573

ABSTRACT

A better understanding of similarities and differences in the composition of the cellular immune system in non-human primates (NHPs) compared with human subjects will improve the interpretation of preclinical studies. It will also aid in addressing the usefulness of NHPs as subjects for studying chronic diseases, vaccine development and immune reconstitution. We employed high content colour flow cytometry and analysed simultaneously the expression of CD3, CD4, CD8alpha, CD8beta, CD16/CD56, CD45RA, CCR7, CD27, CD28, CD107a and the interleukin-7 receptor alpha-chain (IL-7Ralpha) in peripheral blood mononuclear cells (PBMCs) of 27 rhesus macaques and 16 healthy human subjects. Regulatory T cells (Tregs) were identified using anti-CD3, -CD4, -CD25, -FoxP3, and -IL-7Ralpha monoclonal antibodies. Responsiveness to IL-7 was gauged in a signal transducer and activation of transcription 5 (STAT-5) phosphorylation assay. Human and NHP PBMCs showed a similar T-cell composition pattern with some remarkable differences. Similarities: human and NHP CD4(+) and CD8(+) cells showed a similar STAT-5 phosphorylation pattern in response to IL-7. Multicolour flow cytometric analysis identified a CD4(+) CD8alphaalpha(+) CD8alphabeta(+) T-cell population in NHPs as well as in human subjects that expressed the degranulation marker CD107a and may represent a unique CD4(+) T-cell subset endowed with cytotoxic capacity. Differences: we identified in PBMCs from NHPs a higher proportion (5.16% in CD3(+) T cells) of CD8alphaalpha(+) T cells when compared with human donors (1.22% in CD3(+) T cells). NHP CD8alphaalpha(+) T cells produced tumour necrosis factor-alpha / interferon-gamma (TNF-alpha/IFN-gamma) or TNF-alpha, whereas human CD8alphaalpha(+) T cells produced simultaneously TNF-alpha/IFN-gamma and IL-2. A minor percentage of human CD8(+) T cells expressed CD25(bright) and FoxP3 (0.01%). In contrast, 0.07% of NHP CD8(+) T cells exhibited the CD25(bright) FoxP3(+) phenotype. PBMCs from NHPs showed less IL-7Ralpha-positive events in all T-cell subsets including CD4(+) Tregs (median 5%) as compared with human (median 12%). The data visualize commonalities and differences in immune cell subsets in humans and NHPs, most of them in long-lived memory cells and cells with suppressive functions. This provides a matrix to assess future efforts to study diseases and vaccines in NHPs.


Subject(s)
Immunity, Cellular , Macaca mulatta/immunology , T-Lymphocyte Subsets/immunology , Adult , Animals , Cytokines/metabolism , Female , Flow Cytometry , Humans , Interleukin-7/immunology , Phosphorylation , STAT5 Transcription Factor/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/metabolism , Young Adult
9.
Am J Respir Crit Care Med ; 181(12): 1407-17, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20167847

ABSTRACT

RATIONALE: AERAS-402 is a novel tuberculosis vaccine designed to boost immunity primed by bacillus Calmette-Guérin (BCG), the only licensed vaccine. OBJECTIVES: We investigated the safety and immunogenicity of AERAS-402 in healthy Mycobacterium tuberculosis-uninfected BCG-vaccinated adults from a tuberculosis-endemic region of South Africa. METHODS: Escalating doses of AERAS-402 vaccine were administered intramuscularly to each of three groups of healthy South African BCG-vaccinated adults, and a fourth group received two injections of the maximal dose. Participants were monitored for 6 months, with all adverse effects documented. Vaccine-induced CD4(+) and CD8(+) T-cell immunity was characterized by an intracellular cytokine staining assay of whole blood and peripheral blood mononuclear cells. MEASUREMENTS AND MAIN RESULTS: AERAS-402 was well tolerated, and no vaccine-related serious adverse events were recorded. The vaccine induced a robust CD4(+) T-cell response dominated by cells coexpressing IFN-gamma, tumor necrosis factor-alpha, and IL-2 ("polyfunctional" cells). AERAS-402 also induced a potent CD8(+) T-cell response, characterized by cells expressing IFN-gamma and/or tumor necrosis factor-alpha, which persisted for the duration of the study. CONCLUSIONS: Vaccination with AERAS-402 is safe and immunogenic in healthy adults. The immunity induced by the vaccine appears promising: polyfunctional T cells are thought to be important for protection against intracellular pathogens such as Mycobacterium tuberculosis, and evidence is accumulating that CD8(+) T cells are also important. AERAS-402 induced a robust and durable CD8(+) T-cell response, which appears extremely promising. Clinical trial registered with www.sanctr.gov.za (NHREC no. 1381).


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Tuberculosis Vaccines/therapeutic use , Adult , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dose-Response Relationship, Drug , Double-Blind Method , Female , Follow-Up Studies , Humans , Lymphocyte Activation/immunology , Male , South Africa , Tuberculosis Vaccines/immunology , Vaccines, DNA , Young Adult
10.
Immunology ; 129(4): 496-505, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20002212

ABSTRACT

The molecular definition of major histocompatibility complex (MHC) class I-presented CD8(+) T-cell epitopes from clinically relevant Mycobacterium tuberculosis (Mtb) target proteins will aid in the rational design of T-cell-based diagnostics of tuberculosis (TB) and the measurement of TB vaccine-take. We used an epitope discovery system, based on recombinant MHC class I molecules that cover the most frequent Caucasian alleles [human leucocyte antigen (HLA)-A*0101, A*0201, A*0301, A*1101, A*2402, B*0702, B*0801 and B*1501], to identify MHC class I-binding peptides from overlapping 9-mer peptides representing the Mtb protein TB10.4. A total of 33 MHC class I-binding epitopes were identified, spread across the entire amino acid sequence, with some clustering at the N- and C-termini of the protein. Binding of individual peptides or closely related peptide species to different MHC class I alleles was frequently observed. For instance, the common motif of xIMYNYPAMx bound to six of eight alleles. Affinity (50% effective dose) and off-rate (half life) analysis of candidate Mtb peptides will help to define the conditions for CD8(+) T-cell interaction with their nominal MHC class I-peptide ligands. Subsequent construction of tetramers allowed us to confirm the recognition of some of the epitopes by CD8(+) T cells from patients with active pulmonary TB. HLA-B alleles served as the dominant MHC class I restricting molecules for anti-Mtb TB10.4-specific CD8(+) T-cell responses measured in CD8(+) T cells from patients with pulmonary TB.


Subject(s)
Alleles , CD8-Positive T-Lymphocytes/immunology , HLA-B Antigens/immunology , Histocompatibility Antigens Class I/immunology , Mycobacterium tuberculosis/immunology , Peptides/immunology , Binding Sites , HLA-B7 Antigen , HLA-B8 Antigen , Humans , Tuberculosis, Pulmonary/diagnosis , Tuberculosis, Pulmonary/immunology
11.
PLoS One ; 3(11): e3790, 2008.
Article in English | MEDLINE | ID: mdl-19023426

ABSTRACT

BACKGROUND: BCG vaccination, combined with adenoviral-delivered boosts, represents a reasonable strategy to augment, broaden and prolong immune protection against tuberculosis (TB). We tested BCG (SSI1331) (in 6 animals, delivered intradermally) and a recombinant (rBCG) AFRO-1 expressing perfringolysin (in 6 animals) followed by two boosts (delivered intramuscullary) with non-replicating adenovirus 35 (rAd35) expressing a fusion protein composed of Ag85A, Ag85B and TB10.4, for the capacity to induce antigen-specific cellular immune responses in rhesus macaques (Macaca mulatta). Control animals received diluent (3 animals). METHODS AND FINDINGS: Cellular immune responses were analyzed longitudinally (12 blood draws for each animal) using intracellular cytokine staining (TNF-alpha, IL-2 and IFN-gamma), T cell proliferation was measured in CD4(+), CD8alpha/beta(+), and CD8alpha/alpha(+) T cell subsets and IFN-gamma production was tested in 7 day PBMC cultures (whole blood cell assay, WBA) using Ag85A, Ag85B, TB10.4 recombinant proteins, PPD or BCG as stimuli. Animals primed with AFRO-1 showed i) increased Ag85B-specific IFN-gamma production in the WBA assay (median >400 pg/ml for 6 animals) one week after the first boost with adenoviral-delivered TB-antigens as compared to animals primed with BCG (<200 pg/ml), ii) stronger T cell proliferation in the CD8alpha/alpha(+) T cell subset (proliferative index 17%) as compared to BCG-primed animals (proliferative index 5% in CD8alpha/alpha(+) T cells). Polyfunctional T cells, defined by IFN-gamma, TNF-alpha and IL-2 production were detected in 2/6 animals primed with AFRO-1 directed against Ag85A/b and TB10.4; 4/6 animals primed with BCG showed a Ag85A/b responses, yet only a single animal exhibited Ag85A/b and TB10.4 reactivity. CONCLUSION: AFRO-1 induces qualitatively and quantitatively different cellular immune responses as compared with BCG in rhesus macaques. Increased IFN-gamma-responses and antigen-specific T cell proliferation in the CD8alpha/alpha+ T cell subset represents a valuable marker for vaccine-take in BCG-based TB vaccine trials.


Subject(s)
BCG Vaccine/administration & dosage , T-Lymphocytes/immunology , Adenoviridae/genetics , Animals , Antigens, Bacterial/genetics , BCG Vaccine/genetics , Bacterial Toxins/genetics , Female , Genetic Vectors , Hemolysin Proteins/genetics , Immunity, Cellular , Immunization, Secondary , Interferon-gamma/biosynthesis , Lymphocyte Activation , Macaca mulatta , Tuberculosis Vaccines/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics
12.
Vaccine ; 24(18): 3793-803, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16135393

ABSTRACT

We describe the evaluation of three live, attenuated deltaphoP/Q Salmonella enteric serovar Typhimurium strains expressing PEB1 minus its signal sequence (PEB1-ss) from three different plasmids: a pBR-based asd plasmid, an arabinose-based runaway plasmid, which each expressed PEB1-ss in the bacterial cytosol, and a PEB1::HlyA fusion plasmid that directs secretion of PEB1-ss into the extracellular milieu. Serum IgG responses specific for PEB1-ss were induced by pBR-derived and runaway plasmids, with 100 and 90% seroconversion, respectively, at a 1:500 dilution of anti-sera as measured by Western blot analysis, while the PEB1-ss::HlyA fusion plasmid induced serum IgG in only 20% of the mice. Although significant levels of anti-PEB serum IgG were induced, no protection against oral Campylobacter jejuni challenge was observed.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Campylobacter Infections/prevention & control , Campylobacter jejuni/immunology , Salmonella typhimurium/immunology , Administration, Oral , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Blotting, Western , Campylobacter Infections/pathology , Campylobacter Infections/physiopathology , Colony Count, Microbial , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Feces/microbiology , Female , Genetic Vectors , Hemolysin Proteins/genetics , Hemolysin Proteins/immunology , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Plasmids , Protein Sorting Signals/genetics , Salmonella typhimurium/genetics , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
13.
Avian Dis ; 48(3): 445-52, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15529966

ABSTRACT

The purpose of this work was to develop a live, attenuated vaccine strain to protect chickens against colonization by group C Salmonella. We constructed two candidate vaccines: a deltacya deltacrp derivative and a deltaphoP derivative of Salmonella hadar. White Leghorn chickens were vaccinated at day of age and at 2 wk with one of the two strains. A nonvaccinated group served as a control. At 4 wk of age, all birds were challenged with wild-type S. hadar and necropsied 6 days later. Numbers of S. hadar in the ceca were determined. Enzyme-linked immunosorbent assay-derived serum immunoglobulin G responses against S. hadar lipopolysaccharide indicated that both strains induced a serum antibody response. The average optical density450 for birds vaccinated with the deltaphoP or deltacya deltacrp derivatives was 0.456 and 0.881, respectively. Although the deltacya deltacrp derivative induced higher levels of serum antibody, it did not provide an immune response protective against colonization by S. hadar. Conversely, birds vaccinated with the deltaphoP strain showed significant protection against S. hadar challenge. Seventy percent of the nonvaccinates, 60% of the deltacya deltacrp vaccinates, and 15% of deltaphoP vaccinates were positive for S. hadar in tissues. In a second experiment, birds were vaccinated with either the deltaphoP strain or buffer and challenged with a 10-fold higher dose than in the first experiment. After challenge, all of the birds in both groups were colonized. The geometric mean number of cecal S. hadar isolated from the control group was 1.0 x 10(6) colony-forming units (CFU)/g, and from the vaccinated group, this value was 32 CFU/g, indicating a four to five log reduction in colonization by the challenge strain.


Subject(s)
Bacterial Vaccines , Chickens/microbiology , Poultry Diseases/microbiology , Salmonella Infections, Animal/prevention & control , Salmonella/immunology , Animals , Bacterial Proteins/genetics , Cecum/microbiology , Enzyme-Linked Immunosorbent Assay , Gene Transfer Techniques , Immunoglobulin G/immunology , Mutation/genetics , Plasmids/genetics , Poultry Diseases/immunology , Salmonella/genetics , Salmonella Infections, Animal/immunology , Vaccines, Attenuated
14.
Avian Dis ; 48(3): 595-605, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15529982

ABSTRACT

Avian pathogenic Escherichia coli strains are associated with a variety of extraintestinal poultry diseases, including airsacculitis, colisepticemia, and cellulitis. A number of E. coli serotypes are associated with these diseases, although the most prevalent serotype is O78. Fimbrial proteins expressed by these strains appear to be important virulence factors, including type 1 fimbriae, P fimbriae, and curli. We have been working to develop an effective vaccine to protect chickens against these diseases. We have previously shown that an attenuated Salmonella typhimurium strain expressing O78 lipopolysaccharide provides protection against challenge with an O78 avian pathogenic E. coli strain. In this work, we have constructed an attenuated S. typhimurium that expresses both the O78 lipopolysaccharide and E. coli-derived type 1 fimbriae. In these studies, chickens were vaccinated at day of hatch and again at 2 wk of age. Birds were challenged at 4 wk of age. We found that the vaccine candidate provided significant protection against airsacculitis as compared to untreated controls or birds vaccinated with an attenuated S. typhimurium that did not express any E. coli antigens. In a separate experiment, challenged vaccinates showed significant weight gain compared to challenged nonvaccinates. We were not able to demonstrate protection against E. coli O1 or O2 serotype challenge, nor against challenge with wild-type S. typhimurium.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines , Chickens/microbiology , Escherichia coli Infections/veterinary , Escherichia coli/metabolism , Poultry Diseases/prevention & control , Respiratory Tract Infections/veterinary , Salmonella typhimurium/immunology , Air Sacs/microbiology , Animals , Antigens, Bacterial/metabolism , Body Weight , DNA Primers , Enzyme-Linked Immunosorbent Assay , Escherichia coli Infections/prevention & control , Fimbriae, Bacterial/metabolism , Plasmids/genetics , Polymerase Chain Reaction , Poultry Diseases/microbiology , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/prevention & control , Salmonella typhimurium/metabolism , Statistics, Nonparametric
15.
Expert Rev Vaccines ; 3(5): 585-95, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15485338

ABSTRACT

Enterotoxigenic Escherichia coli (ETEC) is recognized as one of the major causes of infectious diarrhea in developing countries. Worldwide, the incidence of ETEC infections is estimated to result in 650 million cases of diarrhea and 380,000 deaths in children under 5 years of age. ETEC is also an important cause of travelers' diarrhea in people traveling to endemic regions of the world. Although ETEC is an uncommon cause of infections in the USA, there have been 14 reported outbreaks of ETEC in the USA and seven on cruise ships over the 20-year period between 1975 and 1995. ETEC strains are comprised of a large number of serotypes that produce a variety of colonization factors and enterotoxins. On infection, ETEC first establishes itself by adhering to the epithelium of the small intestine via one or more colonization factor antigens or coli surface proteins. Once established, ETEC expresses one or more enterotoxin(s), which results in the production of secretory diarrhea. While the need for an efficacious, easily administered vaccine is great, there are currently no licensed ETEC vaccines available for use in endemic countries or for US travelers.


Subject(s)
Escherichia coli Infections/prevention & control , Escherichia coli Vaccines , Escherichia coli/immunology , Vaccination/trends , Animals , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Child , Child, Preschool , Clinical Trials as Topic , Diarrhea/microbiology , Diarrhea/prevention & control , Drug Design , Enterotoxins/immunology , Enterotoxins/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/immunology , Escherichia coli Proteins/metabolism , Fimbriae Proteins/immunology , Fimbriae Proteins/metabolism , Humans , Infant
16.
Infect Immun ; 70(9): 5065-74, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12183554

ABSTRACT

Cruzipain, the major cysteinyl proteinase of Trypanosoma cruzi, is expressed by all developmental forms and strains of the parasite and stimulates potent humoral and cellular immune responses during infection in both humans and mice. This information suggested that cruzipain could be used to develop an effective T. cruzi vaccine. To study whether cruzipain-specific T cells could inhibit T. cruzi intracellular replication, we generated cruzipain-reactive CD4(+) Th1 cell lines. These T cells produced large amounts of gamma interferon when cocultured with infected macrophages, resulting in NO production and decreased intracellular parasite replication. To study the protective effects in vivo of cruzipain-specific Th1 responses against systemic T. cruzi challenges, we immunized mice with recombinant cruzipain plus interleukin 12 (IL-12) and a neutralizing anti-IL-4 MAb. These immunized mice developed potent cruzipain-specific memory Th1 cell responses and were significantly protected against normally lethal systemic T. cruzi challenges. Although cruzipain-specific Th1 responses were associated with T. cruzi protective immunity in vitro and in vivo, adoptive transfer of cruzipain-specific Th1 cells alone did not protect BALB/c histocompatible mice, indicating that additional immune mechanisms are important for cruzipain-specific immunity. To study whether cruzipain could induce mucosal immune responses relevant for vaccine development, we prepared recombinant attenuated Salmonella enterica serovar Typhimurium vaccines expressing cruzipain. BALB/c mice immunized with salmonella expressing cruzipain were significantly protected against T. cruzi mucosal infection. Overall, these data indicate that cruzipain is an important T. cruzi vaccine candidate and that protective T. cruzi vaccines will need to induce more than CD4(+) Th1 cells alone.


Subject(s)
Chagas Disease/immunology , Chagas Disease/prevention & control , Cysteine Endopeptidases/immunology , Trypanosoma cruzi/enzymology , Trypanosoma cruzi/immunology , Adoptive Transfer , Animals , Base Sequence , Cysteine Endopeptidases/genetics , DNA, Protozoan/genetics , Humans , Immunity, Mucosal , Immunization , In Vitro Techniques , Interferon-gamma/biosynthesis , Interleukin-12/administration & dosage , Mice , Mice, Inbred BALB C , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Protozoan Vaccines/genetics , Protozoan Vaccines/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Th1 Cells/immunology , Trypanosoma cruzi/genetics , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...