Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
BMC Immunol ; 16: 63, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26493839

ABSTRACT

BACKGROUND: Requisites for an efficacious tuberculosis (TB) vaccine are a minimal genomic diversity among infectious Mycobacterium tuberculosis strains for the selected antigen, and the capability to induce robust T-cell responses in the majority of human populations. A tool in the identification of putative T-cell epitopes is in silico prediction of major histocompatibility complex (MHC)-peptide binding. Candidate TB vaccine antigen Mtb72F and its successor M72 are recombinant fusion proteins derived from Mtb32A and Mtb39A (encoded by Rv0125 and Rv1196, respectively). Adjuvanted Mtb72F and M72 candidate vaccines were shown to induce CD4(+) T-cell responses in European, US, African and Asian populations. METHODS: Sequence conservation of Mtb32A, Mtb39A, Mtb72F and M72 among 46 strains (prevalent Mycobacterium strains causing human TB disease, and H37Ra) was assessed by multiple alignments using ClustalX. For Mtb32A, Mtb39A and Mtb72F, 15-mer human leukocyte antigen (HLA)-class II-binding peptides were predicted for 158 DRB1 alleles prevailing in populations with high TB burden, 6 DRB3/4/5, 8 DQ and 6 DP alleles, using NetMHCII-pan-3.0. Results for 3 DRB1 alleles were compared with previously published allele-matched in vitro binding data. Additional analyses were done for M72. Nonameric MHC class I-binding peptides in Mtb72F were predicted for three alleles representative of class I supertypes A02, A03 and B07, using seven prediction algorithms. RESULTS: Sequence identity among strains was ≥98 % for each protein. Residue changes in Mtb39A comprised primarily single residue or nucleotide insertions and/or deletions in repeat regions, and were observed in 67 % of strains. For Mtb72F, 156 DRB1, 6 DRB3/4/5, 7 DQ and 5 DP alleles were predicted to contain at least one MHC class II-binding peptide, and class I-binding peptides were predicted for each HLA-A/B allele. Comparison of predicted MHC-II-binding peptides with experimental data indicated that the algorithm's sensitivity and specificity were variable among alleles. CONCLUSIONS: The sequences from which Mtb72F and M72 are derived are highly conserved among representative Mycobacterium strains. Predicted putative T-cell epitopes in M72 and/or Mtb72F covered a wide array of HLA alleles. In silico binding predictions for class I- and II-binding putative epitopes can be complemented with biochemical verification of HLA binding capacity, processing and immunogenicity of the predicted peptides.


Subject(s)
Antigens, Bacterial/chemistry , Antigens, Bacterial/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , HLA Antigens/chemistry , HLA Antigens/immunology , Peptides/immunology , Tuberculosis Vaccines/immunology , Alleles , Amino Acid Sequence , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Conserved Sequence , Epitopes, T-Lymphocyte/metabolism , HLA Antigens/genetics , HLA Antigens/metabolism , HLA-DRB1 Chains/chemistry , HLA-DRB1 Chains/genetics , HLA-DRB1 Chains/immunology , HLA-DRB1 Chains/metabolism , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Mycobacterium/genetics , Mycobacterium/immunology , Peptides/chemistry , Protein Binding , Tuberculosis/immunology , Tuberculosis/prevention & control
2.
PLoS One ; 10(4): e0122835, 2015.
Article in English | MEDLINE | ID: mdl-25856308

ABSTRACT

HIV-1-specific CD4+ and CD8+ T lymphocytes are important for HIV-1 replication control. F4/AS01 consists of F4 recombinant fusion protein (containing clade B Gag/p24, Pol/RT, Nef and Gag/p17) formulated in AS01 Adjuvant System, and was shown to induce F4-specific polyfunctional CD4+ T-cell responses in humans. While replication-incompetent recombinant HIV-1/SIV antigen-expressing human adenoviral vectors can elicit high-frequency antigen-specific CD8+ T-cell responses, their use is hampered by widespread pre-existing immunity to human serotypes. Non-human adenovirus serotypes associated with lower prevalence may offer an alternative strategy. We evaluated the immunogenicity of AdC7-GRN ('A'), a recombinant chimpanzee adenovirus type 7 vector expressing clade B Gag, RT and Nef, and F4/AS01 ('P'), when delivered intramuscularly in homologous (PP or AA) and heterologous (AAPP or PPAA) prime-boost regimens, in macaques and mice. Vaccine-induced HIV-1-antigen-specific T cells in peripheral blood (macaques), liver, spleen, and intestinal and genital mucosa (mice) were characterized by intracellular cytokine staining. Vaccine-specific IgG antibodies (macaques) were detected using ELISA. In macaques, only the heterologous prime-boost regimens induced polyfunctional, persistent and balanced CD4+ and CD8+ T-cell responses specific to each HIV-1 vaccine antigen. AdC7-GRN priming increased the polyfunctionality of F4/AS01-induced CD4+ T cells. Approximately 50% of AdC7-GRN-induced memory CD8+ T cells exhibited an effector-memory phenotype. HIV-1-specific antibodies were detected with each regimen. In mice, antigen-specific CD4+ and CD8+ T-cell responses were detected in the mucosal and systemic anatomical compartments assessed. When administered in heterologous prime-boost regimens, AdC7-GRN and F4/AS01 candidate vaccines acted complementarily in inducing potent and persistent peripheral blood HIV-1-specific CD4+ and CD8+ T-cell responses and antibodies in macaques. Besides, adenoviral vector priming modulated the cytokine-expression profile of the protein-induced CD4+ T cells. Each regimen induced HIV-1-specific T-cell responses in systemic/local tissues in mice. This suggests that prime-boost regimens combining adjuvanted protein and low-seroprevalent chimpanzee adenoviral vectors represent an attractive vaccination strategy for clinical evaluation.


Subject(s)
Genetic Vectors/genetics , HIV-1/immunology , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Viral Vaccines/immunology , Adenoviridae , Animals , Antibodies, Viral/blood , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Genes, pol/genetics , HIV Antigens/genetics , HIV Core Protein p24/genetics , Injections, Intramuscular , Macaca , Mice , Pan troglodytes , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Viral Vaccines/administration & dosage , gag Gene Products, Human Immunodeficiency Virus/genetics , nef Gene Products, Human Immunodeficiency Virus/genetics
3.
AIDS ; 28(12): 1769-81, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-24911353

ABSTRACT

OBJECTIVE: Tuberculosis (TB) is highly prevalent among HIV-infected people, including those receiving combination antiretroviral therapy (cART), necessitating a well tolerated and efficacious TB vaccine for these populations. We evaluated the safety and immunogenicity of the candidate TB vaccine M72/AS01 in adults with well controlled HIV infection on cART. DESIGN: A randomized, observer-blind, controlled trial (NCT00707967). METHODS: HIV-infected adults on cART in Switzerland were randomized 3 : 1 : 1 to receive two doses, 1 month apart, of M72/AS01, AS01 or 0.9% physiological saline (N = 22, N = 8 and N = 7, respectively) and were followed up to 6 months postdose 2 (D210). Individuals with CD4⁺ cell counts below 200 cells/µl were excluded. Adverse events (AEs) including HIV-specific and laboratory safety parameters were recorded. Cell-mediated (ICS) and humoral (ELISA) responses were evaluated before vaccination, 1 month after each dose (D30, D60) and D210. RESULTS: Thirty-seven individuals [interquartile range (IQR) CD4⁺ cell counts at screening: 438-872 cells/µl; undetectable HIV-1 viremia] were enrolled; 73% of individuals reported previous BCG vaccination, 97.3% tested negative for the QuantiFERON-TB assay. For M72/AS01 recipients, no vaccine-related serious AEs or cART-regimen adjustments were recorded, and there were no clinically relevant effects on laboratory safety parameters, HIV-1 viral loads or CD4⁺ cell counts. M72/AS01 was immunogenic, inducing persistent and polyfunctional M72-specific CD4⁺ T-cell responses [medians 0.70% (IQR 0.37-1.07) at D60] and 0.42% (0.24-0.61) at D210, predominantly CD40L⁺IL-2⁺TNF-α⁺, CD40L⁺IL-2⁺ and CD40L⁺IL-2⁺TNF-α⁺IFN-γ⁺]. All M72/AS01 vaccines were seropositive for anti-M72 IgG after second vaccination until study end. CONCLUSION: M72/AS01 was clinically well tolerated and immunogenic in this population, supporting further clinical evaluation in HIV-infected individuals in TB-endemic settings.


Subject(s)
Anti-Retroviral Agents/therapeutic use , HIV Infections/drug therapy , HIV Infections/immunology , Lipid A/analogs & derivatives , Saponins/adverse effects , Tuberculosis Vaccines/adverse effects , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Adolescent , Adult , Antibodies, Bacterial/blood , Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes/immunology , Drug Combinations , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , HIV Infections/complications , Humans , Immunoglobulin G/blood , Lipid A/administration & dosage , Lipid A/adverse effects , Male , Middle Aged , Placebos/administration & dosage , Saponins/administration & dosage , Single-Blind Method , Switzerland , T-Lymphocyte Subsets/immunology , Treatment Outcome , Tuberculosis Vaccines/administration & dosage , Vaccination/adverse effects , Vaccination/methods , Young Adult
4.
J Clin Immunol ; 33(8): 1360-75, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24142232

ABSTRACT

PURPOSE: In this dose-finding Phase II study (NCT00621322), we evaluated the safety and immunogenicity of different formulations of the candidate tuberculosis vaccine containing the M72 antigen (10/20/40 µg doses) and the liposome-based AS01 Adjuvant System. We aimed to select the lowest-dose combination of M72 and AS01 that was clinically well tolerated with immunogenicity comparable to that of the previously tested M72/AS01B (40 µg) candidate vaccine. METHODS: Healthy PPD-positive (induration 3-10 mm) adults (18-45 years) in The Philippines were randomized (4:4:4:4:1:1) to receive 2 injections, 1 month apart, of M72/AS01B (40 µg), M72/AS01E (10 µg), M72/AS01E (20 µg), M72/AS02D (10 µg), M72/Saline (40 µg) or AS01B alone, and were followed up for 6 months. AS01E and AS02D contain half the quantities of the immunostimulants present in AS01B. AS02D is an oil-in-water emulsion. Vaccine selection was based on the CD4(+) T-cell responses at 1 month post vaccination. RESULTS: All formulations had a clinically acceptable safety profile with no vaccine-related serious adverse events reported. Two vaccinations of each adjuvanted M72 vaccine induced M72-specific CD4(+) T-cell and humoral responses persisting at 6 months post vaccination. No responses were observed with AS01B alone. One month post second vaccination, CD4(+) T-cell responses induced by each of the three M72/AS01 vaccine formulations were of comparable magnitudes, and all were significantly higher than those induced by M72/AS02D (10 µg) and M72/Saline. CONCLUSIONS: The formulation with the lowest antigen and adjuvant dose, M72/AS01E (10 µg), fulfilled our pre-defined selection criteria and has been selected for further clinical development.


Subject(s)
Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Adolescent , Adult , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Dose-Response Relationship, Immunologic , Drug Combinations , Female , Humans , Immunity, Cellular , Immunity, Humoral , Lymphocyte Count , Male , Middle Aged , Tuberculosis Vaccines/adverse effects , Young Adult
5.
Am J Respir Crit Care Med ; 188(4): 492-502, 2013 08 15.
Article in English | MEDLINE | ID: mdl-23306546

ABSTRACT

RATIONALE: Tuberculosis (TB) is a major cause of morbidity and mortality worldwide, thus there is an urgent need for novel TB vaccines. OBJECTIVES: We investigated a novel TB vaccine candidate, M72/AS01, in a phase IIa trial of bacille Calmette-Guérin-vaccinated, HIV-uninfected, and Mycobacterium tuberculosis (Mtb)-infected and -uninfected adults in South Africa. METHODS: Two doses of M72/AS01 were administered to healthy adults, with and without latent Mtb infection. Participants were monitored for 7 months after the first dose; cytokine production profiles, cell cycling, and regulatory phenotypes of vaccine-induced T cells were measured by flow cytometry. MEASUREMENTS AND MAIN RESULTS: The vaccine had a clinically acceptable safety profile, and induced robust, long-lived M72-specific T-cell and antibody responses. M72-specific CD4 T cells produced multiple combinations of Th1 cytokines. Analysis of T-cell Ki67 expression showed that most vaccination-induced T cells did not express Th1 cytokines or IL-17; these cytokine-negative Ki67(+) T cells included subsets of CD4 T cells with regulatory phenotypes. PD-1, a negative regulator of activated T cells, was transiently expressed on M72-specific CD4 T cells after vaccination. Specific T-cell subsets were present at significantly higher frequencies after vaccination of Mtb-infected versus -uninfected participants. CONCLUSIONS: M72/AS01 is clinically well tolerated in Mtb-infected and -uninfected adults, induces high frequencies of multifunctional T cells, and boosts distinct T-cell responses primed by natural Mtb infection. Moreover, these results provide important novel insights into how this immunity may be appropriately regulated after novel TB vaccination of Mtb-infected and -uninfected individuals.Clinical trial registered with www.clinicaltrials.gov (NCT 00600782).


Subject(s)
T-Lymphocytes/immunology , Tuberculosis Vaccines/immunology , Adult , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Female , Flow Cytometry , Humans , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Interleukin-17/metabolism , Male , South Africa , Tuberculosis Vaccines/administration & dosage , Young Adult
6.
Vaccine ; 31(17): 2196-206, 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-22643213

ABSTRACT

BACKGROUND: The Bacille Calmette-Guérin (BCG) tuberculosis (TB) vaccine provides incomplete protection, necessitating development of an effective vaccine against TB disease. The Mtb72F/AS02 candidate vaccine was previously shown to be clinically well tolerated and immunogenic in Purified Protein Derivative (PPD)-negative adults. To improve the stability of Mtb72F, a point mutation was introduced into a putative serine protease site to give the final M72 construct. AS01 is an Adjuvant System that can potentially improve both humoral and cellular immune responses compared to the AS02 Adjuvant System or unadjuvanted vaccine. This study evaluated the safety and immunogenicity in Mtb-naïve adults of vaccines containing 40 µg of the M72 antigen with AS02 or AS01 and compared the results with Mtb72F/AS02 vaccine (40 µg dose), M72 in saline (40 µg dose) and AS01 alone. METHODS: In this Phase I/II observer-blind controlled trial, 110 participants were randomized (4:4:1:1:1) to receive M72/AS01, M72/AS02, Mtb72F/AS02, M72/saline or AS01, following a 0, 1-month schedule. Subjects receiving the adjuvanted M72 vaccines were followed up until 3 years post vaccination. Evaluation of the immune response and safety/reactogenicity was performed. RESULTS: For all vaccines, solicited adverse events (AEs) were predominantly mild to moderate and transient. No vaccine-related serious AEs occurred and no subject withdrew due to an AE. Immune responses induced by Mtb72F and M72 antigens combined with AS02 were similar. M72/AS01 and M72/AS02 induced robust polyfunctional M72-specific CD4(+) T cell and antibody responses persisting at 3 years, with the highest CD4(+) T cell responses found with M72/AS01. CONCLUSION: This first clinical study with M72/AS01 and M72/AS02 showed that both vaccines were clinically well tolerated and induced high magnitude and persistent cell-mediated and humoral immune responses. The Mtb72F/AS02 and M72/AS02 vaccines were comparably immunogenic with significantly higher immune responses compared to the M72/saline control. Of the formulations tested, M72/AS01 demonstrated significantly higher vaccine specific Th1 CD4(+) T cell responses supporting its further clinical evaluation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Mycobacterium tuberculosis/immunology , Tuberculin , Tuberculosis Vaccines/classification , Tuberculosis Vaccines/immunology , Adjuvants, Immunologic , Adolescent , Adult , Antibodies, Bacterial/immunology , Antibody Formation/immunology , Female , Humans , Immunity, Cellular/immunology , Immunity, Humoral , Male , Middle Aged , Tuberculosis/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/adverse effects , Vaccination/adverse effects , Young Adult
7.
Tuberculosis (Edinb) ; 93(2): 179-88, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23219236

ABSTRACT

UNLABELLED: Prevention of tuberculosis (TB) through vaccination would substantially reduce the global TB burden. Mtb72F/AS02 is a candidate TB vaccine shown to be immunogenic and well tolerated in PPD-negative adults. We evaluated the safety and immunogenicity of Mtb72F/AS02 in Mycobacterium-primed adults (BCG-vaccinated, or infected adults who had received post-exposure chemoprophylaxis or treatment for pulmonary TB disease). In this observer-blind controlled trial, 20 BCG-vaccinated adults and 18 adults previously infected with Mycobacterium tuberculosis (Mtb), were randomized 3:1 to receive three doses of Mtb72F/AS02 or AS02 at one-month intervals, and followed for 6 months post third vaccination. Mtb72F/AS02 was well tolerated in BCG-vaccinated adults, and tended to be more reactogenic in Mtb-infected adults. Adverse events were mainly self-limiting, resolving without sequelae. No serious adverse events were reported. The adverse events in Mtb72F/AS02 vaccinees were not clearly associated with vaccine-induced responses (as assessed by proinflammatory cytokines, total IgE and C-reactive protein levels). No Th2 T-cell responses, or vaccine-induced T-cell responses to Mtb antigens (CFP-10/PPD/ESAT-6) were detected by ICS. In both cohorts, Mtb72F/AS02 induced persistent polyfunctional Mtb72F-specific CD4(+) T-cell responses and anti-Mtb72F humoral responses. IFN-γ was detectable in serum one day post each vaccination. Further evaluation of the candidate vaccine, Mtb72F/AS02, is warranted. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT00146744.


Subject(s)
Tuberculin/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/immunology , Adolescent , Adult , Antibodies, Bacterial/biosynthesis , BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , Dose-Response Relationship, Immunologic , Double-Blind Method , Female , Humans , Immunity, Cellular , Immunoglobulin G/biosynthesis , Interferon-gamma/biosynthesis , Male , Middle Aged , Mycobacterium tuberculosis/immunology , T-Lymphocyte Subsets/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/adverse effects , Vaccination/adverse effects , Young Adult
8.
Naunyn Schmiedebergs Arch Pharmacol ; 385(12): 1211-25, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22983013

ABSTRACT

As a new human immunodeficiency virus type 1 (HIV-1) vaccine approach, the live-attenuated measles virus (MV) Schwarz vaccine strain was genetically engineered to express the F4 antigen (MV1-F4). F4 is a fusion protein comprising HIV-1 antigens p17 and p24, reverse transcriptase and Nef. This study assessed the toxicity, biodistribution and shedding profiles of MV1-F4. Cynomolgus macaques were intramuscularly immunized one or three times with the highest dose of MV1-F4 intended for clinical use, the reference (Schwarz) measles vaccine or saline, and monitored clinically for 11 or 85 days. Toxicological parameters included local and systemic clinical signs, organ weights, haematology, clinical and gross pathology and histopathology. Both vaccines were well tolerated, with no morbidity, clinical signs or gross pathological findings observed. Mean spleen weights were increased after three doses of either vaccine, which corresponded with increased numbers and/or sizes of germinal centers. This was likely a result of the immune response to the vaccines. Either vaccine virus replicated preferentially in secondary lymphoid organs and to a lesser extent in epithelium-rich tissues (e.g., intestine, urinary bladder and trachea) and the liver. At the expected peak of viremia, viral RNA was detected in some biological fluid samples from few animals immunized with either vaccine, but none of these samples contained infectious virus. In conclusion, no shedding of infectious viral particles was identified in cynomolgus monkeys after injection of MV1-F4 or Schwarz measles vaccines. Furthermore, no toxic effect in relation to the MV vaccination was found with these vaccines in this study.


Subject(s)
AIDS Vaccines/immunology , HIV Antigens/immunology , Measles Vaccine/immunology , Measles virus/immunology , AIDS Vaccines/pharmacokinetics , AIDS Vaccines/toxicity , Animals , Female , Genetic Engineering/methods , HIV-1/immunology , Injections, Intramuscular , Macaca fascicularis , Male , Measles Vaccine/pharmacokinetics , Measles Vaccine/toxicity , Organ Size/immunology , RNA, Viral/metabolism , Time Factors , Tissue Distribution , Virus Replication , Virus Shedding
9.
FEMS Immunol Med Microbiol ; 55(2): 258-70, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19281568

ABSTRACT

Chlamydia trachomatis infection is the most common sexually transmitted bacterial infection worldwide, with over 91 million cases estimated annually. An effective subunit vaccine against Chlamydia may require a multivalent subunit cocktail of antigens in a single formulation for broad coverage of a heterogeneous major histocompatibility complex population. Herein, we describe the identification of novel C. trachomatis antigens by CD4+ and CD8+ T-cell expression cloning, serological expression cloning, and an in silico analysis of the C. trachomatis genome. These antigens elicited human CD4+ T-cell responses, and a subset proved to be immunogenic and protective when administered as immunoprophylactic vaccines against C. trachomatis challenge. Candidate vaccines consisting of the prioritized C. trachomatis antigens adjuvanted in a GlaxoSmithKline proprietary AS01B adjuvant were prioritized based on induction of solid protection against challenge in C57BL/6 and BALB/c mice with C. trachomatis. Some of the vaccines prevented bacterial shedding and colonization of the upper genital tract to varying degrees by mechanisms that may include CD4+ T cells.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Vaccines/immunology , Chlamydia Infections/prevention & control , Chlamydia trachomatis/immunology , Genital Diseases, Female/prevention & control , Recombinant Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Chlamydia Infections/immunology , Colony Count, Microbial , Female , Genital Diseases, Female/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Vaccines, Synthetic/immunology
10.
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
11.
Vaccine ; 26(8): 1072-82, 2008 Feb 20.
Article in English | MEDLINE | ID: mdl-18258343

ABSTRACT

Protection against Plasmodium falciparum sporozoite infection can be achieved by vaccination with the recombinant circumsporozoite protein-based vaccine RTS,S formulated with the AS02A Adjuvant System. Since this protection is only partial and wanes over time, we have developed a new RTS,S-based vaccine adjuvanted with AS01B. RTS,S/AS01B-induced high specific antibody titers and increased the frequency of mouse CD4(+) and CD8(+) T cells expressing IFN-gamma, and of monkey CD4(+) T cells expressing IL-2 and/or IFN-gamma and/or TNF-alpha upon stimulation with vaccine antigens. Our data provides clear evidence that combining RTS,S antigen with a potent adjuvant induces strong humoral and cellular responses in vivo.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Animals , Antibodies, Protozoan/blood , Female , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Macaca mulatta , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/biosynthesis
12.
Vaccine ; 25(42): 7354-62, 2007 Oct 16.
Article in English | MEDLINE | ID: mdl-17870213

ABSTRACT

Nicotine replacement therapies (NRT) have limited success in smoking cessation. The efficacy of nicotine may be compromised by its main metabolite, cotinine. An anti-cotinine vaccine to remove this antagonism could enhance the efficacy of NRT. We show that cotinine is a weak nicotinic agonist and decreases responses to nicotine, consistent with antagonism through receptor desensitisation. trans-4-Thiol cotinine was coupled to tetanus toxoid, and rats immunised repeatedly. Vaccination raised antibodies specific for cotinine that do not recognise other metabolites or nicotine. Increased serum cotinine concentrations following nicotine administration indicate sequestration of cotinine by antibodies, encouraging further evaluation of this vaccine in behavioural models of nicotine addiction and relapse.


Subject(s)
Cotinine/antagonists & inhibitors , Cotinine/immunology , Nicotine/administration & dosage , Smoking Cessation/methods , Vaccines, Conjugate/isolation & purification , Animals , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Cotinine/metabolism , Dopamine/metabolism , Humans , In Vitro Techniques , Male , Nicotine/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism
13.
Infect Immun ; 75(5): 2283-90, 2007 May.
Article in English | MEDLINE | ID: mdl-17307942

ABSTRACT

The RTS,S/AS02A protein-based vaccine consistently demonstrates significant protection against infection with Plasmodium falciparum malaria and also against clinical malaria and severe disease in children in areas of endemicity. Here we demonstrate with rhesus macaques that priming with a replication-defective human adenovirus serotype 35 (Ad35) vector encoding circumsporozoite protein (CS) (Ad35.CS), followed by boosting with RTS,S in an improved MPL- and QS21-based adjuvant formulation, AS01B, maintains antibody responses and dramatically increases levels of T cells producing gamma interferon and other Th1 cytokines in response to CS peptides. The increased T-cell responses induced by the combination of Ad35.CS and RTS,S/AS01B are sustained for at least 6 months postvaccination and may translate to improved and more durable protection against P. falciparum infection in humans.


Subject(s)
Adenoviridae/genetics , Immunization Schedule , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Protozoan Proteins/immunology , Adenoviridae/classification , Adjuvants, Immunologic , Animals , Antibodies, Protozoan/blood , Female , Humans , Immunization , Immunization, Secondary , Interferon-gamma/metabolism , Macaca mulatta , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Male , Plasmodium falciparum/immunology , Protozoan Proteins/administration & dosage , Protozoan Proteins/genetics , Receptors, Thrombopoietin/immunology , Saponins/immunology , T-Lymphocytes/immunology , Th1 Cells
14.
Infect Immun ; 75(2): 838-45, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17101665

ABSTRACT

Liver-stage antigen 1 (LSA1) is expressed by Plasmodium falciparum only during the intrahepatic cell stage of the parasite's development. Immunoepidemiological studies in regions where malaria is endemic suggested an association between the level of LSA1-specific humoral and cell-mediated immune responses and susceptibility to clinical malaria. A recombinant LSA1 protein, FMP011, has been manufactured as a preerythrocytic vaccine to induce an immune response that would have the effect of controlling parasitemia and disease in humans. To evaluate the immunogenicity of FMP011, we analyzed the immune response of three inbred strains of mice to antigen immunization using two different adjuvant formulations, AS01B and AS02A. We report here the ability of BALB/c and A/J mice, but not C57BL/6J mice, to mount FMP011-specific humoral (antibody titer) and cellular (gamma interferon [IFN-gamma] production) responses following immunization with FMP011 formulated in AS01B or AS02A. Immunization of BALB/c and A/J mice with FMP011/AS01B induced more antigen-specific IFN-gamma-producing splenocytes than immunization with FMP011/AS02A. A slightly higher titer of antibody was induced using AS02A than AS01B in both strains. C57BL/6J mice did not respond with any detectable FMP011-specific IFN-gamma splenocytes or antibody when immunized with FMP011 in AS01B or AS02A. Intracellular staining of cells isolated from FMP011/AS01B-immunized BALB/c mice indicated that CD4(+) cells, but not CD8(+) cells, were the main IFN-gamma-producing splenocyte. However, inclusion of blocking anti-CD4(+) antibody during the in vitro restimulation ELISpot analysis failed to completely abolish IFN-gamma production, indicating that while CD4(+) T cells were the major source of IFN-gamma, other cell types also were involved.


Subject(s)
Adjuvants, Immunologic , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Animals , Antibodies, Protozoan/blood , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Interferon-gamma/biosynthesis , Lymphocyte Subsets/immunology , Mice , Mice, Inbred A , Mice, Inbred BALB C , Mice, Inbred C57BL , Vaccines, Subunit/immunology , Vaccines, Synthetic/immunology
15.
Br Med Bull ; 62: 175-86, 2002.
Article in English | MEDLINE | ID: mdl-12176859

ABSTRACT

Multiple immunotherapy approaches are under development to treat various chronic diseases or conditions. Thanks to dramatic technological improvements, the field of vaccinology can now extend from prophylaxis to therapy, and from infectious diseases to dysimmune disorders like cancer or autoimmunity, or even to non-immune related conditions, including neurological or cardiovascular disorders. Life-style vaccines defined as vaccines to manage chronic conditions in healthy individuals can, therefore, also be envisaged. Three examples will be reviewed and discussed here, from both technical and ethical aspects: contraceptive vaccines, vaccines to treat addictions, and anti-dental caries vaccines.


Subject(s)
Dental Caries/prevention & control , Substance-Related Disorders/therapy , Vaccination/methods , Vaccines, Contraceptive , Female , Humans , Male , Streptococcal Infections/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...