Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Malar J ; 13: 215, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24893777

ABSTRACT

BACKGROUND: When rhesus monkeys (Macaca mulatta) are used to test malaria vaccines, animals are often challenged by the intravenous injection of sporozoites. However, natural exposure to malaria comes via mosquito bite, and antibodies can neutralize sporozoites as they traverse the skin. Thus, intravenous injection may not fairly assess humoral immunity from anti-sporozoite malaria vaccines. To better assess malaria vaccines in rhesus, a method to challenge large numbers of monkeys by mosquito bite was developed. METHODS: Several species and strains of mosquitoes were tested for their ability to produce Plasmodium knowlesi sporozoites. Donor monkey parasitaemia effects on oocyst and sporozoite numbers and mosquito mortality were documented. Methylparaben added to mosquito feed was tested to improve mosquito survival. To determine the number of bites needed to infect a monkey, animals were exposed to various numbers of P. knowlesi-infected mosquitoes. Finally, P. knowlesi-infected mosquitoes were used to challenge 17 monkeys in a malaria vaccine trial, and the effect of number of infectious bites on monkey parasitaemia was documented. RESULTS: Anopheles dirus, Anopheles crascens, and Anopheles dirus X (a cross between the two species) produced large numbers of P. knowlesi sporozoites. Mosquito survival to day 14, when sporozoites fill the salivary glands, averaged only 32% when donor monkeys had a parasitaemia above 2%. However, when donor monkey parasitaemia was below 2%, mosquitoes survived twice as well and contained ample sporozoites in their salivary glands. Adding methylparaben to sugar solutions did not improve survival of infected mosquitoes. Plasmodium knowlesi was very infectious, with all monkeys developing blood stage infections if one or more infected mosquitoes successfully fed. There was also a dose-response, with monkeys that received higher numbers of infected mosquito bites developing malaria sooner. CONCLUSIONS: Anopheles dirus, An. crascens and a cross between these two species all were excellent vectors for P. knowlesi. High donor monkey parasitaemia was associated with poor mosquito survival. A single infected mosquito bite is likely sufficient to infect a monkey with P. knowlesi. It is possible to efficiently challenge large groups of monkeys by mosquito bite, which will be useful for P. knowlesi vaccine studies.


Subject(s)
Anopheles/physiology , Anopheles/parasitology , Malaria/transmission , Plasmodium knowlesi/growth & development , Animals , Female , Macaca mulatta , Malaria Vaccines/administration & dosage , Male , Survival Analysis
2.
PLoS One ; 7(2): e31247, 2012.
Article in English | MEDLINE | ID: mdl-22355349

ABSTRACT

Live attenuated malaria vaccines are more potent than the recombinant protein, bacterial or viral platform vaccines that have been tested, and an attenuated sporozoite vaccine against falciparum malaria is being developed for humans. In mice, attenuated malaria sporozoite vaccines induce CD8(+) T cells that kill parasites developing in the liver. We were curious to know if CD8(+) T cells were also important in protecting primates against malaria. We immunized 9 rhesus monkeys with radiation attenuated Plasmodium knowlesi sporozoites, and found that 5 did not develop blood stage infections after challenge with live sporozoites. We then injected 4 of these protected monkeys with cM-T807, a monoclonal antibody to the CD8 molecule which depletes T cells. The fifth monkey received equivalent doses of normal IgG. In 3 of the 4 monkeys receiving cM-T807 circulating CD8(+) T cells were profoundly depleted. When re-challenged with live sporozoites all 3 of these depleted animals developed blood stage malaria. The fourth monkey receiving cM-T807 retained many circulating CD8(+) T cells. This monkey, and the vaccinated monkey receiving normal IgG, did not develop blood stage malaria at re-challenge with live sporozoites. Animals were treated with antimalarial drugs and rested for 4 months. During this interval CD8(+) T cells re-appeared in the circulation of the depleted monkeys. When all vaccinated animals received a third challenge with live sporozoites, all 5 monkeys were once again protected and did not develop blood stage malaria infections. These data indicate that CD8(+) T cells are important effector cells protecting monkeys against malaria sporozoite infection. We believe that malaria vaccines which induce effector CD8+ T cells in humans will have the best chance of protecting against malaria.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Malaria Vaccines/therapeutic use , Malaria/immunology , Malaria/prevention & control , Plasmodium knowlesi/immunology , Sporozoites/immunology , Vaccines, Attenuated/immunology , Animals , Case-Control Studies , Flow Cytometry , Immunization , Immunologic Memory , Interferon-gamma/blood , Liver/immunology , Liver/parasitology , Macaca mulatta , Malaria/parasitology , Mice , Vaccination
3.
Malar J ; 10: 29, 2011 Feb 08.
Article in English | MEDLINE | ID: mdl-21303498

ABSTRACT

BACKGROUND: A DNA prime, poxvirus (COPAK) boost vaccination regime with four antigens, i.e. a combination of two Plasmodium knowlesi sporozoite (csp/ssp2) and two blood stage (ama1/msp142) genes, leads to self-limited parasitaemia in 60% of rhesus monkeys and survival from an otherwise lethal infection with P. knowlesi. In the present study, the role of the blood stage antigens in protection was studied in depth, focusing on antibody formation against the blood stage antigens and the functionality thereof. METHODS: Rhesus macaques were immunized with the four-component vaccine and subsequently challenged i.v. with 100 P. knowlesi sporozoites. During immunization and challenge, antibody titres against the two blood stage antigens were determined, as well as the in vitro growth inhibition capacity of those antibodies. Antigen reversal experiments were performed to determine the relative contribution of antibodies against each of the two blood stage antigens to the inhibition. RESULTS: After vaccination, PkAMA1 and PkMSP119 antibody titres in vaccinated animals were low, which was reflected in low levels of inhibition by these antibodies as determined by in vitro inhibition assays. Interestingly, after sporozoite challenge antibody titres against blood stage antigens were boosted over 30-fold in both protected and not protected animals. The in vitro inhibition levels increased to high levels (median inhibitions of 59% and 56% at 6 mg/mL total IgG, respectively). As growth inhibition levels were not significantly different between protected and not protected animals, the ability to control infection appeared cannot be explained by GIA levels. Judged by in vitro antigen reversal growth inhibition assays, over 85% of the inhibitory activity of these antibodies was directed against PkAMA1. CONCLUSIONS: This is the first report that demonstrates that a DNA prime/poxvirus boost vaccination regimen induces low levels of malaria parasite growth inhibitory antibodies, which are boosted to high levels upon challenge. No association could, however, be established between the levels of inhibitory capacity in vitro and protection, either after vaccination or after challenge.


Subject(s)
Macaca mulatta/immunology , Malaria Vaccines/immunology , Malaria/immunology , Plasmodium knowlesi/genetics , Poxviridae/genetics , Animals , Antibodies, Protozoan , Antigens, Protozoan/blood , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Enzyme-Linked Immunosorbent Assay , Immunization, Secondary , Macaca mulatta/blood , Malaria/blood , Malaria/prevention & control , Malaria Vaccines/chemistry , Plasmids/metabolism , Plasmodium knowlesi/immunology , Protozoan Proteins/blood , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Sporozoites/immunology , Treatment Outcome
4.
PLoS One ; 4(8): e6559, 2009 Aug 10.
Article in English | MEDLINE | ID: mdl-19668343

ABSTRACT

Using newer vaccine platforms which have been effective against malaria in rodent models, we tested five immunization regimens against Plasmodium knowlesi in rhesus monkeys. All vaccines included the same four P. knowlesi antigens: the pre-erythrocytic antigens CSP, SSP2, and erythrocytic antigens AMA1, MSP1. We used four vaccine platforms for prime or boost vaccinations: plasmids (DNA), alphavirus replicons (VRP), attenuated adenovirus serotype 5 (Ad), or attenuated poxvirus (Pox). These four platforms combined to produce five different prime/boost vaccine regimens: Pox alone, VRP/Pox, VRP/Ad, Ad/Pox, and DNA/Pox. Five rhesus monkeys were immunized with each regimen, and five Control monkeys received a mock vaccination. The time to complete vaccinations was 420 days. All monkeys were challenged twice with 100 P. knowlesi sporozoites given IV. The first challenge was given 12 days after the last vaccination, and the monkeys receiving the DNA/Pox vaccine were the best protected, with 3/5 monkeys sterilely protected and 1/5 monkeys that self-cured its parasitemia. There was no protection in monkeys that received Pox malaria vaccine alone without previous priming. The second sporozoite challenge was given 4 months after the first. All 4 monkeys that were protected in the first challenge developed malaria in the second challenge. DNA, VRP and Ad5 vaccines all primed monkeys for strong immune responses after the Pox boost. We discuss the high level but short duration of protection in this experiment and the possible benefits of the long interval between prime and boost.


Subject(s)
Macaca mulatta/immunology , Malaria Vaccines/immunology , Malaria/veterinary , Plasmodium knowlesi/immunology , Animals , Antibodies, Protozoan/biosynthesis , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Genetic Vectors , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , T-Lymphocytes/immunology , Viruses/genetics
5.
PLoS One ; 2(10): e1063, 2007 Oct 24.
Article in English | MEDLINE | ID: mdl-17957247

ABSTRACT

BACKGROUND: We have previously described a four antigen malaria vaccine consisting of DNA plasmids boosted by recombinant poxviruses which protects a high percentage of rhesus monkeys against Plasmodium knowlesi (Pk) malaria. This is a multi-stage vaccine that includes two pre-erythrocytic antigens, PkCSP and PkSSP2(TRAP), and two erythrocytic antigens, PkAMA-1 and PkMSP-1(42kD). The present study reports three further experiments where we investigate the effects of DNA dose, timing, and formulation. We also compare vaccines utilizing only the pre-erythrocytic antigens with the four antigen vaccine. METHODOLOGY: In three experiments, rhesus monkeys were immunized with malaria vaccines using DNA plasmid injections followed by boosting with poxvirus vaccine. A variety of parameters were tested, including formulation of DNA on poly-lactic co-glycolide (PLG) particles, varying the number of DNA injections and the amount of DNA, varying the interval between the last DNA injection to the poxvirus boost from 7 to 21 weeks, and using vaccines with from one to four malaria antigens. Monkeys were challenged with Pk sporozoites given i.v. 2 to 4 weeks after the poxvirus injection, and parasitemia was measured by daily Giemsa stained blood films. Immune responses in venous blood samples taken after each vaccine injection were measured by ELIspot production of interferon-gamma, and by ELISA. CONCLUSIONS: 1) the number of DNA injections, the formulation of the DNA plasmids, and the interval between the last DNA injection and the poxvirus injection are critical to vaccine efficacy. However, the total dose used for DNA priming is not as important; 2) the blood stage antigens PkAMA-1 and PkMSP-1 were able to protect against high parasitemias as part of a genetic vaccine where antigen folding is not well defined; 3) immunization with PkSSP2 DNA inhibited immune responses to PkCSP DNA even when vaccinations were given into separate legs; and 4) in a counter-intuitive result, higher interferon-gamma ELIspot responses to the PkCSP antigen correlated with earlier appearance of parasites in the blood, despite the fact that PkCSP vaccines had a protective effect.


Subject(s)
Malaria Vaccines/chemistry , Malaria/metabolism , Malaria/prevention & control , Poxviridae/genetics , Animals , Antibodies, Protozoan , Antigens, Protozoan/blood , Antigens, Protozoan/immunology , Enzyme-Linked Immunosorbent Assay , Erythrocytes/virology , Immune System , Immunization, Secondary , Macaca mulatta , Malaria/immunology , Plasmids/metabolism , Plasmodium knowlesi , T-Lymphocytes/metabolism , Treatment Outcome
6.
Malar J ; 6: 135, 2007 Oct 09.
Article in English | MEDLINE | ID: mdl-17925026

ABSTRACT

The present study has evaluated the immunogenicity of single or multiple Plasmodium falciparum (Pf) antigens administered in a DNA prime/poxvirus boost regimen with or without the poloxamer CRL1005 in rhesus monkeys. Animals were primed with PfCSP plasmid DNA or a mixture of PfCSP, PfSSP2/TRAP, PfLSA1, PfAMA1 and PfMSP1-42 (CSLAM) DNA vaccines in PBS or formulated with CRL1005, and subsequently boosted with ALVAC-Pf7, a canarypox virus expressing the CSLAM antigens. Cell-mediated immune responses were evaluated by IFN-gamma ELIspot and intracellular cytokine staining, using recombinant proteins and overlapping synthetic peptides. Antigen-specific and parasite-specific antibody responses were evaluated by ELISA and IFAT, respectively. Immune responses to all components of the multi-antigen mixture were demonstrated following immunization with either DNA/PBS or DNA/CRL1005, and no antigen interference was observed in animals receiving CSLAM as compared to PfCSP alone. These data support the down-selection of the CSLAM antigen combination. CRL1005 formulation had no apparent effect on vaccine-induced T cell or antibody responses, either before or after viral boost. In high responder monkeys, CD4+IL-2+ responses were more predominant than CD8+ T cell responses. Furthermore, CD8+ IFN-gamma responses were detected only in the presence of detectable CD4+ T cell responses. Overall, this study demonstrates the potential for multivalent Pf vaccines based on rational antigen selection and combination, and suggests that further formulation development to increase the immunogenicity of DNA encoded antigens is warranted.


Subject(s)
Antigens, Protozoan/immunology , Immunization, Secondary/methods , Malaria Vaccines/administration & dosage , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Poxviridae/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Protozoan/biosynthesis , Antigens, Protozoan/administration & dosage , Antigens, Protozoan/genetics , Immunization , Macaca mulatta , Malaria Vaccines/immunology , Plasmids , Vaccines, DNA/administration & dosage
7.
J Exp Med ; 203(5): 1249-58, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16636134

ABSTRACT

There is a remarkable heterogeneity in the functional profile (quality) of T cell responses. Importantly, the magnitude and/or quality of a response required for protection may be different depending on the infection. Here, we assessed the capacity of different Toll like receptor (TLR)-binding compounds to influence T helper cell (Th)1 and CD8+ T cell responses when used as adjuvants in nonhuman primates (NHP) with HIV Gag as a model antigen. NHP were immunized with HIV Gag protein emulsified in Montanide ISA 51, an oil-based adjuvant, with or without a TLR7/8 agonist, a TLR8 agonist, or the TLR9 ligand cytosine phosphate guanosine oligodeoxynucleotides (CpG ODN), and boosted 12 wk later with a replication-defective adenovirus-expressing HIV-Gag (rAD-Gag). Animals vaccinated with HIV Gag protein/Montanide and CpG ODN or the TLR7/8 agonist had higher frequencies of Th1 responses after primary immunization compared to all other vaccine groups. Although the rAD-Gag boost did not elevate the frequency of Th1 memory cytokine responses, there was a striking increase in HIV Gag-specific CD8+ T cell responses after the boost in all animals that had received a primary immunization with any of the TLR adjuvants. Importantly, the presence and type of TLR adjuvant used during primary immunization conferred stability and dramatically influenced the magnitude and quality of the Th1 and CD8+ T cell responses after the rAD-Gag boost. These data provide insights for designing prime-boost immunization regimens to optimize Th1 and CD8+ T cell responses.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Immunologic Memory/drug effects , Mannitol/analogs & derivatives , Oleic Acids/administration & dosage , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Animals , CD8-Positive T-Lymphocytes/immunology , CpG Islands/immunology , Cytokines/immunology , Gene Products, gag/administration & dosage , Gene Products, gag/immunology , Immunization , Macaca mulatta , Mannitol/administration & dosage , Mannitol/immunology , Oleic Acids/immunology , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Th1 Cells/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 8/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
8.
Proc Natl Acad Sci U S A ; 102(42): 15190-4, 2005 Oct 18.
Article in English | MEDLINE | ID: mdl-16219698

ABSTRACT

Induction and maintenance of antibody and T cell responses will be critical for developing a successful vaccine against HIV. A rational approach for generating such responses is to design vaccines or adjuvants that have the capacity to activate specific antigen-presenting cells. In this regard, dendritic cells (DCs) are the most potent antigen-presenting cells for generating primary T cell responses. Here, we report that Toll-like receptor (TLR) agonists and ligands that activate DCs in vitro influence the magnitude and quality of the cellular immune response in nonhuman primates (NHPs) when administered with HIV Gag protein. NHPs immunized with HIV Gag protein and a TLR7/8 agonist or a TLR9 ligand [CpG oligodeoxynucleotides (CpG ODN)] had significantly increased Gag-specific T helper 1 and antibody responses, compared with animals immunized with HIV Gag protein alone. Importantly, conjugating the HIV Gag protein to the TLR7/8 agonist (Gag-TLR7/8 conjugate) dramatically enhanced the magnitude and altered the quality of the T helper 1 response, compared with animals immunized with HIV Gag protein and the TLR7/8 agonist or CpG ODN. Furthermore, immunization with the Gag-TLR7/8 conjugate vaccine elicited Gag-specific CD8+ T responses. Collectively, our results show that conjugating HIV Gag protein to a TLR7/8 agonist is an effective way to elicit broad-based adaptive immunity in NHPs. This type of vaccine formulation should have utility in preventive or therapeutic vaccines in which humoral and cellular immunity is required.


Subject(s)
AIDS Vaccines/immunology , CD8-Positive T-Lymphocytes/immunology , Gene Products, gag/immunology , HIV Antigens/immunology , Th1 Cells/immunology , Animals , Dendritic Cells/cytology , Dendritic Cells/immunology , Gene Products, gag/genetics , HIV Antigens/genetics , Humans , Interferon-gamma/immunology , Interleukin-2/immunology , Ligands , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/immunology , Primates , T-Lymphocyte Subsets/immunology
9.
Infect Immun ; 72(3): 1594-602, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14977966

ABSTRACT

We describe a novel approach for identifying target antigens for preerythrocytic malaria vaccines. Our strategy is to rapidly test hundreds of DNA vaccines encoding exons from the Plasmodium yoelii yoelii genomic sequence. In this antigen identification method, we measure reduction in parasite burden in the liver after sporozoite challenge in mice. Orthologs of protective P. y. yoelii genes can then be identified in the genomic databases of Plasmodium falciparum and Plasmodium vivax and investigated as candidate antigens for a human vaccine. A pilot study to develop the antigen identification method approach used 192 P. y. yoelii exons from genes expressed during the sporozoite stage of the life cycle. A total of 182 (94%) exons were successfully cloned into a DNA immunization vector with the Gateway cloning technology. To assess immunization strategies, mice were vaccinated with 19 of the new DNA plasmids in addition to the well-characterized protective plasmid encoding P. y. yoelii circumsporozoite protein. Single plasmid immunization by gene gun identified a novel vaccine target antigen which decreased liver parasite burden by 95% and which has orthologs in P. vivax and P. knowlesi but not P. falciparum. Intramuscular injection of DNA plasmids produced a different pattern of protective responses from those seen with gene gun immunization. Intramuscular immunization with plasmid pools could reduce liver parasite burden in mice despite the fact that none of the plasmids was protective when given individually. We conclude that high-throughput cloning of exons into DNA vaccines and their screening is feasible and can rapidly identify new malaria vaccine candidate antigens.


Subject(s)
Antigens, Protozoan/genetics , Malaria Vaccines/genetics , Plasmodium yoelii/genetics , Plasmodium yoelii/immunology , Animals , Antibodies, Protozoan/blood , Base Sequence , Biolistics , Cloning, Molecular , DNA Primers , DNA, Protozoan/genetics , Exons , Female , Fluorescent Antibody Technique, Indirect , Genome, Protozoan , Humans , Injections, Intramuscular , Liver/parasitology , Malaria/immunology , Malaria/parasitology , Malaria/prevention & control , Malaria Vaccines/administration & dosage , Malaria Vaccines/pharmacology , Mice , Mice, Inbred BALB C , Plasmids/genetics , Plasmodium yoelii/growth & development , Polymerase Chain Reaction , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/pharmacology
10.
J Immunol ; 171(6): 3148-53, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12960342

ABSTRACT

In some parts of Africa, 50% of deaths attributed to malaria occur in infants less than 8 mo. Thus, immunization against malaria may have to begin in the neonatal period, when neonates have maternally acquired Abs against malaria parasite proteins. Many malaria vaccines in development rely upon CD8 cells as immune effectors. Some studies indicate that neonates do not mount optimal CD8 cell responses. We report that BALB/c mice first immunized as neonates (7 days) with a Plasmodium yoelii circumsporozoite protein (PyCSP) DNA vaccine mixed with a plasmid expressing murine GM-CSF (DG) and boosted at 28 days with poxvirus expressing PyCSP were protected (93%) as well as mice immunized entirely as adults (70%). Protection was dependent on CD8 cells, and mice had excellent anti-PyCSP IFN-gamma and cytotoxic T lymphocyte responses. Mice born of mothers previously exposed to P. yoelii parasites or immunized with the vaccine were protected and had excellent T cell responses. These data support assessment of this immunization strategy in neonates/young infants in areas in which malaria exacts its greatest toll.


Subject(s)
Animals, Newborn/immunology , CD8-Positive T-Lymphocytes/immunology , Immunity, Maternally-Acquired , Lymphocyte Activation , Malaria Vaccines/immunology , Malaria/prevention & control , Poxviridae/immunology , Vaccines, DNA/administration & dosage , Animals , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/blood , Female , Immunity, Maternally-Acquired/genetics , Immunization Schedule , Immunization, Secondary/methods , Injections, Intramuscular , Kinetics , Lymphocyte Activation/genetics , Malaria/immunology , Malaria/parasitology , Malaria Vaccines/administration & dosage , Malaria Vaccines/genetics , Male , Maternal-Fetal Exchange/immunology , Mice , Mice, Inbred BALB C , Poxviridae/genetics , Pregnancy , Sporozoites/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology
11.
Infect Immun ; 70(8): 4329-35, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12117942

ABSTRACT

We tested a cytokine-enhanced, multiantigen, DNA priming and poxvirus boosting vaccine regimen for prevention of malaria in the Plasmodium knowlesi-rhesus macaque model system. Animals were primed with a mixture of DNA plasmids encoding two preerythrocytic-stage proteins and two erythrocytic-stage proteins from P. knowlesi and combinations of the cytokines granulocyte-macrophage colony-stimulating factor, interleukin-4, and tumor necrosis factor alpha and were boosted with a mixture of four recombinant, attenuated vaccinia virus strains encoding the four P. knowlesi antigens. Two weeks after boosting, the geometric mean immunofluorescence titers in the immunized groups against sporozoites and infected erythrocytes ranged from 160 to 8,096 and from 1,810 to 5,120, respectively. The geometric mean anti-P. knowlesi circumsporozoite protein (PkCSP) titers ranged from 1,761 to 24,242. Peripheral blood mononuclear cells (PBMC) from the immunized monkeys produced gamma interferon (IFN-gamma) in response to incubation with pooled peptides from the PkCSP at frequencies of 10 to 571 spot-forming cells/10(6) PBMC. Following challenge with 100 infectious P. knowlesi sporozoites, 2 of 11 immunized monkeys were sterilely protected, and 7 of the 9 infected monkeys resolved their parasitemias spontaneously. In contrast, all four controls became infected and required treatment for overwhelming parasitemia. Early protection was strongly associated with IFN-gamma responses against a pool of peptides from the preerythrocytic-stage antigen, PkCSP. These findings demonstrate that a multistage, multiantigen, DNA priming and poxvirus boosting vaccine regimen can protect nonhuman primates from an otherwise lethal malaria sporozoite challenge.


Subject(s)
Antigens, Protozoan/genetics , DNA, Protozoan/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interleukin-4/immunology , Malaria Vaccines/immunology , Malaria/prevention & control , Plasmodium knowlesi/immunology , Tumor Necrosis Factor-alpha/immunology , Vaccines, DNA/immunology , Amino Acid Sequence , Animals , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Disease Models, Animal , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunization, Secondary , Interleukin-4/genetics , Macaca mulatta , Malaria Vaccines/genetics , Molecular Sequence Data , Plasmodium knowlesi/genetics , Protozoan Proteins/genetics , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/genetics , Vaccines, DNA/genetics , Vaccinia virus
SELECTION OF CITATIONS
SEARCH DETAIL
...