Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
J Thromb Haemost ; 16(3): 555-564, 2018 03.
Article in English | MEDLINE | ID: mdl-29274194

ABSTRACT

Essentials AFSTYLA exhibits ≈50% underestimation in activity when the one-stage (OS) assay is utilized. A field study compared the performance of AFSTYLA with Advate in factor VIII activity assays. AFSTYLA activity can be monitored with both the chromogenic substrate and the OS assay. The consistent OS underestimation allows for a conversion factor to be applied to OS results. SUMMARY: Introduction AFSTYLA (antihemophilic factor [recombinant] single chain) is a novel B-domain truncated recombinant factor VIII (rFVIII). For AFSTYLA, an approximate 50% discrepancy was observed between results of the one-stage (OS) and chromogenic substrate (ChS) FVIII activity assays. An investigation was undertaken to test whether there is a linear relationship between ChS and OS assay results that would allow reliable clinical interpretation of results independent of the assay method used. Aims To provide confidence in future clinical monitoring, this field study investigated the performance of AFSTYLA and a full-length rFVIII (Advate® ) in FVIII activity assays routinely performed in clinical laboratories. Methods The comparison of AFSTYLA and Advate was performed in an international, multicenter and blinded field study of simulated post-infusion samples. The study documented the extent of variability between methods and laboratories and characterized the relationship between the ChS and OS assays. Results Results from 23 laboratories demonstrate that intra and interlaboratory variability in OS assays were similar for both products. When comparing within the OS assay format, there was a similar and reagent-correlated variability in response to different activators for both AFSTYLA and Advate. The OS underestimation was highly predictable and consistent across the complete range of FVIII plasma concentrations. Conclusion Post-infusion plasma AFSTYLA levels can be monitored in patients by the OS and ChS assays. The consistent and predictable difference between the two assay formats provides clinicians with adequate guidance on how to interpret the results of the OS assay using a single conversion factor.


Subject(s)
Blood Coagulation Tests/standards , Blood Coagulation , Clinical Laboratory Services/standards , Factor VIII/analysis , Hemostasis , Plasma/chemistry , Chromogenic Compounds/chemistry , Hemophilia A/blood , Humans , Indicators and Reagents , International Cooperation , Recombinant Proteins/chemistry , Reproducibility of Results
2.
Gene Ther ; 11(5): 448-56, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14973538

ABSTRACT

We measured the ability of nine DNA vaccine plasmids encoding candidate malaria vaccine antigens to induce antibodies and interferon-gamma responses when delivered alone or in a mixture containing all nine plasmids. We further examined the possible immunosuppressive effect of individual plasmids, by assessing a series of mixtures in which each of the nine vaccine plasmids was replaced with a control plasmid. Given alone, each of the vaccine plasmids induced significant antibody titers and, in the four cases for which appropriate assays were available, IFN-gamma responses. Significant suppression or complete abrogation of responses were seen when the plasmids were pooled in a nine-plasmid cocktail and injected in a single site. Removal of single genes from the mixture frequently reduced the observed suppression. Boosting with recombinant poxvirus increased the antibody response in animals primed with either a single gene or the mixture, but, even after boosting, responses were higher in animals primed with single plasmids than in those primed with the nine-plasmid mixture. Boosting did not overcome the suppressive effect of mixing for IFN-gamma responses. Interactions between components in a multiplasmid DNA vaccine may limit the ability to use plasmid pools alone to induce responses against multiple targets simultaneously.


Subject(s)
Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Protozoan/biosynthesis , Antigens, Protozoan/administration & dosage , Antigens, Protozoan/immunology , DNA, Protozoan/administration & dosage , Dose-Response Relationship, Immunologic , Female , Immune Tolerance , Immunization/methods , Interferon-gamma/biosynthesis , Malaria Vaccines/administration & dosage , Mice , Mice, Inbred BALB C , Plasmids/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Combined/immunology
4.
Expert Opin Biol Ther ; 1(4): 619-28, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11727498

ABSTRACT

Malaria is still a leading cause of morbidity and mortality in human populations. Problems, including drug-resistant parasites and insecticide resistant mosquitoes, ensure the continued hold of malaria in the tropics and sub-tropics. Each year around 100 million cases of malaria result in at least 50,000 deaths outside of sub-Saharan Africa; within sub-Saharan Africa itself, malaria causes around one million child deaths per year. New approaches for malaria control are badly needed and much effort has gone to develop malaria vaccines. In addition to giving personal protection, most such vaccines would also tend to reduce the transmission of malaria. One class of vaccine is being developed specifically for this purpose--the malaria transmission-blocking vaccines (TBV). TBVs are based upon antigens expressed on the surface of the sexual and mosquito mid-gut stages of malaria parasites. These antigens are the targets of antibodies induced by vaccination of the host and ingested with the parasites in a mosquito blood meal. The antibodies act by inhibiting the parasite's development within the mosquito itself and they thereby prevent the onward transmission of the parasites. TBVs could contribute to the total interruption of malaria transmission in many locations with relatively low transmission rates, mostly outside sub-Saharan Africa. Under almost all transmission rates, however, TBVs would help reduce malaria incidence and malaria-related morbidity and mortality. Promising recombinant TBV candidate antigens for the two main human malaria parasite species, Plasmodium falciparum and Plasmodium vivax, have been produced and tested in the laboratory; one has undergone early clinical trials.


Subject(s)
Malaria Vaccines/immunology , Malaria/drug therapy , Plasmodium falciparum/immunology , Plasmodium vivax/immunology , Animals , Antigens, Protozoan/immunology , Antigens, Surface/immunology , Child , Culicidae/parasitology , Culicidae/physiology , Humans , Malaria/immunology , Malaria/physiopathology , Malaria/transmission , Malaria Vaccines/chemistry , Plasmodium falciparum/physiology , Plasmodium vivax/physiology , Protozoan Proteins/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology
5.
Vaccine ; 20(5-6): 763-70, 2001 Dec 12.
Article in English | MEDLINE | ID: mdl-11738740

ABSTRACT

Transmission-blocking vaccines against malaria parasites target molecules expressed by sexual stage parasites to elicit antibodies that prevent the infection of the mosquito vector. Pvs25 and Pvs28, expressed on the surface of ookinetes, are potential candidates for such a vaccine and induce antibodies that block the infectivity of Plasmodium vivax in immunized animals. To improve the ability to induce transmission-blocking antibodies, Pvs25 and Pvs28 were produced as a single fusion protein by the yeast Saccharomyces cerevisiae. Mice immunized with a low dose of the chimeric molecule (Pvs25-28) developed higher antibody responses compared with mice immunized with either Pvs25 or Pvs28. In membrane feeding assays, both anti-Pvs25-28 and anti-Pvs25 antisera had similarly potent transmission-blocking activities (and both were much greater than anti-Pvs28). Furthermore, serum from mice simultaneously immunized with both Pvs25 and Pvs28, or serum mixtures of anti-Pvs25 alone and anti-Pvs28 alone did not enhance the efficacy over anti-Pvs25 serum alone, demonstrating that there is no synergism in the ability to block transmission of P. vivax between anti-Pvs25 and anti-Pvs28 antibodies.


Subject(s)
Antibodies, Protozoan/biosynthesis , Antigens, Protozoan/administration & dosage , Malaria Vaccines/administration & dosage , Plasmodium vivax/immunology , Animals , Antigens, Protozoan/genetics , Base Sequence , Culicidae/parasitology , DNA, Protozoan/genetics , Drug Synergism , Female , Immunization , Insect Vectors/parasitology , Malaria Vaccines/genetics , Malaria, Vivax/immunology , Malaria, Vivax/prevention & control , Malaria, Vivax/transmission , Mice , Plasmodium vivax/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics
6.
Protein Expr Purif ; 23(3): 468-75, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11722185

ABSTRACT

P30P2MSP1(19) is a recombinant subunit vaccine derived from merozoite surface protein 1 (MSP1) of Plasmodium falciparum, the causative agent of malaria. P30P2MSP1(19) consists of two universal T-cell epitopes fused to the most C-terminal 19-kDa portion of MSP1, and this protein has previously shown promising potential as a vaccine for malaria. However, previous attempts at producing this molecule in Saccharomyces cerevisiae resulted in the production of a truncated form of the molecule missing most of the universal T-cell epitopes. Here, we report the production of full-length P30P2MSP1(19) in Pichia pastoris. As salt precipitation is a common problem during P. pastoris high-density fermentation, we utilized an alternative low-salt, fully defined medium that did not reduce growth rates or biomass yields to avoid precipitation. A total of 500 mg/L of secreted purified protein was produced in high cell density fermentation and the protein was purified in one step utilizing nickel-chelate chromatography. P30P2MSP1(19) produced in Pichia was reactive with monoclonal antibodies that recognize only conformational epitopes on correctly folded MSP1. Rabbits immunized with this molecule generated higher and more uniform antibody titers than rabbits immunized with the protein produced in Saccharomyces. P30P2MSP1(19) produced in Pichia may prove to be a more efficacious vaccine than that produced in Saccharomyces and Pichia would provide a system for the cost-effective production of such a vaccine.


Subject(s)
Malaria Vaccines/immunology , Merozoite Surface Protein 1/genetics , Merozoite Surface Protein 1/isolation & purification , Pichia/genetics , Plasmodium falciparum/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibodies, Protozoan/immunology , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Fermentation , Gene Expression , Genetic Vectors , Malaria, Falciparum/prevention & control , Merozoite Surface Protein 1/biosynthesis , Merozoite Surface Protein 1/immunology , Pichia/metabolism , Protein Conformation , Protein Folding , Rabbits , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/isolation & purification , Transformation, Genetic
7.
Trends Parasitol ; 17(9): 415-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11530352

ABSTRACT

The development of malaria blood-stage vaccines is gathering momentum: there are several new funding initiatives, one multiantigen formulation is currently being tested and at least one other blood-stage vaccine is expected to begin trials in 2001. However, there is no consensus over the best way to select which form of an antigen to take into clinical testing. There is thus a danger that less-effective vaccines might be tested in the field in the order of their availability, rather than merit. Here, we argue that first proving efficacy in the New World monkey challenge model would accelerate development.


Subject(s)
Aotus trivirgatus , Clinical Trials as Topic , Erythrocytes/parasitology , Malaria Vaccines , Malaria/prevention & control , Plasmodium falciparum/immunology , Animals , Antigens, Protozoan/immunology , Disease Models, Animal , Humans , Life Cycle Stages
8.
J Immunol ; 167(7): 3903-9, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11564808

ABSTRACT

Immunity induced by the 19-kDa fragment of merozoite surface protein 1 is dependent on CD4+ Th cells. However, we found that adoptively transferred CFSE-labeled Th cells specific for an epitope on Plasmodium yoelii 19-kDa fragment of merozoite surface protein 1 (peptide (p)24), but not OVA-specific T cells, were deleted as a result of P. yoelii infection. As a result of infection, spleen cells recovered from infected p24-specific T cell-transfused mice demonstrated reduced response to specific Ag. A higher percentage of CFSE-labeled p24-specific T cells stained positive with annexin and anti-active caspase-3 in infected compared with uninfected mice, suggesting that apoptosis contributed to deletion of p24-specific T cells during infection. Apoptosis correlated with increased percentages of p24-specific T cells that stained positive for Fas from infected mice, suggesting that P. yoelii-induced apoptosis is, at least in part, mediated by Fas. However, bystander cells of other specificities also showed increased Fas expression during infection, suggesting that Fas expression alone is not sufficient for apoptosis. These data have implications for the development of immunity in the face of endemic parasite exposure.


Subject(s)
Apoptosis , Malaria/immunology , Merozoite Surface Protein 1/immunology , Plasmodium yoelii/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adoptive Transfer , Animals , Antibodies, Protozoan/biosynthesis , Cell Line , Cells, Cultured , Epitopes/immunology , Female , Fluoresceins/chemistry , Fluorescent Dyes/chemistry , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Nude , Succinimides/chemistry , T-Lymphocytes, Helper-Inducer/transplantation , fas Receptor/metabolism
9.
Vaccine ; 19(32): 4661-8, 2001 Sep 14.
Article in English | MEDLINE | ID: mdl-11535314

ABSTRACT

The gene encoding the Plasmodium yoelii homologue of P. falciparum merozoite surface proteins 4 (MSP4) and 5 (MSP5) has been expressed in Escherichia coli and Saccharomyces cerevisiae. The protein contains a single epidermal growth factor (EGF)-like domain and is expressed in a form lacking the predicted N-terminal signal and glycosyl phosphatidylinositol (GPI) attachment sequences. The recombinant protein derived from E. coli (EcMSP4/5) was highly effective at protecting mice against lethal challenge with 10(5) parasites of the P. yoelii YM strain. In contrast, the protective efficacy of yeast-derived MSP4/5 (yMSP4/5) was considerably less. The antibody titres in both groups were significantly different with mice immunised with yeast-derived protein showing significantly lower pre-challenge antibody responses. There was a significant inverse correlation between antibody levels as measured by ELISA and peak parasitaemia. Mice immunised with EcMSP4/5 produced anti-PyMSP4/5 antibodies predominantly of the IgG2a and IgG2b isotypes, whereas, mice immunised with yMSP4/5 mainly produced antibodies of the IgG1 isotype. The differences in antibody titres and subtype distribution may account for the observed differences in protective efficacy of these protein preparations. Levels of protective efficacy of MSP4/5 were compared with that obtained using P. yoelii MSP1 produced in S. cerevisiae. Levels of protection induced by E. coli derived MSP4/5 were superior to those induced by MSP1 which in turn were better than those induced by yeast-derived MSP4/5.


Subject(s)
Antigens, Protozoan/immunology , Escherichia coli/metabolism , Malaria Vaccines/immunology , Malaria/prevention & control , Membrane Proteins/immunology , Plasmodium yoelii/immunology , Protozoan Proteins/immunology , Saccharomyces cerevisiae/metabolism , Animals , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/immunology , Antigens, Protozoan/biosynthesis , Antigens, Protozoan/genetics , Antigens, Protozoan/isolation & purification , Female , Immunoglobulin G/biosynthesis , Immunoglobulin G/immunology , Immunoglobulin Isotypes/biosynthesis , Immunoglobulin Isotypes/immunology , Malaria/immunology , Malaria Vaccines/biosynthesis , Malaria Vaccines/genetics , Malaria Vaccines/isolation & purification , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/isolation & purification , Mice , Mice, Inbred BALB C , Parasitemia/immunology , Plasmodium yoelii/genetics , Protein Structure, Tertiary , Protozoan Proteins/biosynthesis , Protozoan Proteins/genetics , Protozoan Proteins/isolation & purification , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/isolation & purification , Species Specificity , Structure-Activity Relationship , Vaccination
10.
Infect Immun ; 69(7): 4390-7, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11401978

ABSTRACT

Merozoite surface protein 4 (MSP4) of Plasmodium falciparum is a glycosylphosphatidylinositol-anchored integral membrane protein that is being developed as a component of a subunit vaccine against malaria. We report here the measurement of naturally acquired antibodies to MSP4 in a population of individuals living in the Khanh-Hoa region of Vietnam, an area where malaria is highly endemic. Antibodies to MSP4 were detected in 94% of the study population at titers of 1:5,000 or greater. Two forms of recombinant MSP4 produced in either Escherichia coli or Saccharomyces cerevisiae were compared as substrates in the enzyme-linked immunosorbent assay. There was an excellent correlation between reactivity measured to either, although the yeast substrate was recognized by a higher percentage of sera. Four different regions of MSP4 were recognized by human antibodies, demonstrating that there are at least four distinct epitopes in this protein. In the carboxyl terminus, where the single epidermal growth factor-like domain is located, the reactive epitope(s) was shown to be conformation dependent, as disruption of the disulfide bonds almost completely abolished reactivity with human antibodies. The anti-MSP4 antibodies were mainly of the immunoglobulin G1 (IgG1) and IgG3 subclasses, suggesting that such antibodies may play a role in opsonization and complement-mediated lysis of free merozoites. Individuals in the study population were drug-cured and followed up for 6 months; no significant correlation was observed between the anti-MSP4 antibodies and the absence of parasitemia during the surveillance period. As a comparison, antibodies to MSP1(19), a leading vaccine candidate, were measured, and no correlation with protection was observed in these individuals. The anti-MSP1(19) antibodies were predominantly of the IgG1 isotype, in contrast to the IgG3 predominance noted for MSP4.


Subject(s)
Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Endemic Diseases , Malaria, Falciparum/immunology , Protozoan Proteins/immunology , Adolescent , Adult , Animals , Antibodies, Protozoan/immunology , Antibody Specificity , Child , Epitopes, B-Lymphocyte/immunology , Humans , Immunoglobulin Isotypes , Malaria, Falciparum/blood , Malaria, Falciparum/epidemiology , Malaria, Falciparum/prevention & control , Merozoite Surface Protein 1/immunology , Middle Aged , Plasmodium falciparum/immunology , Prevalence , Time Factors , Vietnam/epidemiology
11.
Infect Immun ; 69(3): 1536-46, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11179324

ABSTRACT

In an attempt to produce a more defined, clinical-grade version of a vaccine based on Plasmodium falciparum merozoite surface protein 1 (MSP1), we evaluated the efficacy of two recombinant forms of MSP1 in an Aotus nancymai challenge model system. One recombinant vaccine, bvMSP1(42), based on the 42-kDa C-terminal portion of MSP1, was expressed as a secreted protein in baculovirus-infected insect cells. A highly pure baculovirus product could be reproducibly expressed and purified at yields in excess of 8 mg of pure protein per liter of culture. This protein, when tested for efficacy in the Aotus challenge model, gave significant protection, with only one of seven monkeys requiring treatment for uncontrolled parasitemia after challenge with P. falciparum. The second recombinant protein, P30P2MSP1(19), has been used in previous studies and is based on the smaller, C-terminal 19-kDa portion of MSP1 expressed in Saccharomyces cerevisiae. Substantial changes were made in its production process to optimize expression. The optimum form of this vaccine antigen (as judged by in vitro and in vivo indicators) was then evaluated, along with bvMSP1(42), for efficacy in the A. nancymai system. The new formulation of P30P3MSP1(19) performed significantly worse than bvMSP1(42) and appeared to be less efficacious than we have found in the past, with four of seven monkeys in the vaccinated group requiring treatment for uncontrolled parasitemia. With both antigens, protection was seen only when high antibody levels were obtained by formulation of the vaccines in Freund's adjuvant. Vaccine formulation in an alternate adjuvant, MF59, resulted in significantly lower antibody titers and no protection.


Subject(s)
Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Merozoite Surface Protein 1/therapeutic use , Plasmodium falciparum/immunology , Vaccination , Animals , Antibodies, Protozoan/blood , Aotidae , Baculoviridae/genetics , Genetic Variation , Merozoite Surface Protein 1/genetics , Parasitemia , Rabbits , Recombinant Fusion Proteins/therapeutic use , Technology, Pharmaceutical/methods , Tetanus Toxin/therapeutic use , Vaccines, Synthetic/therapeutic use
12.
Yeast ; 18(2): 137-50, 2001 Jan 30.
Article in English | MEDLINE | ID: mdl-11169756

ABSTRACT

A recombinant protein expression system based on Saccharomyces cerevisiae has been used to express malarial vaccine candidate antigens. The antigens so produced have been used in three Phase 1 clinical trials and numerous preclinical non-human primate trials. Further Phase I trials are planned using these candidate vaccine antigens. These molecules were identified as attractive candidates for antimalarial vaccines, as they are all surface-exposed at some stage in the parasite's life cycle. They all share an unusual structural feature: epidermal growth factor (EGF)-like motifs. When these proteins are expressed in our S. cerevisiae expression system, they are produced as a series of stable structural conformers, each with a different disulphide bonding pattern. This leads to both biochemical and, more importantly, antigenic differences between the conformers (e.g. presence or absence of an antibody B cell epitope). These findings have important ramifications for other EGF-domain-containing proteins expressed in S. cerevisiae, or for proteins which contain other cysteine-folding motifs not normally expressed by this organism, both for vaccine production or for research/reagent purposes.


Subject(s)
Antigens, Protozoan/chemistry , Malaria Vaccines/chemistry , Vaccines, Synthetic/chemistry , Animals , Antigens, Protozoan/immunology , Antigens, Protozoan/isolation & purification , Clinical Trials, Phase I as Topic , Disulfides/chemistry , Epidermal Growth Factor/chemistry , Humans , Malaria Vaccines/immunology , Malaria Vaccines/isolation & purification , Plasmodium falciparum/immunology , Protein Processing, Post-Translational , Protein Structure, Secondary , Saccharomyces cerevisiae , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification
13.
Vaccine ; 19(7-8): 816-24, 2000 Nov 22.
Article in English | MEDLINE | ID: mdl-11115704

ABSTRACT

Plasmodium falciparum merozoite surface protein 4 (MSP4) is being developed as a component of a subunit vaccine against asexual stages of malaria. Three DNA constructs were produced that induced expression of MSP4 either in the cytoplasm of transfected cells or secreted from cells under the control of the human tissue plasminogen activator (TPA) signal or the native P. falciparum MSP4 signal. Only the construct containing the TPA signal induced detectable antibodies in mice, although gene expression was demonstrated in all constructs and MSP4 was shown to be secreted using either signal by in vitro transient transfection of COS cells. Two recombinant MSP4 proteins that encoded the same sequence as the plasmid DNA were produced in E. coli (EcMSP4-His) and S. cerevisiae (yMSP4-His) and used to raise antibodies in mice. Comparison of the antibodies elicited by these various antigen formulations showed differences in titer, isotype and epitope recognition. The titer of antibodies induced by DNA vaccination was lower than that induced by yMSP4-His, which in turn was lower than that induced by EcMSP4-His. The isotype profiles of the antibodies were also different, the plasmid DNA induced predominantly IgG(2a) responses whereas the two proteins induced predominantly IgG(1) responses. The antibodies induced by DNA and yMSP4-His recognized predominantly the C-terminal epidermal growth factor (EGF)-like domain of the protein, whereas EcMSP4-His induced antibodies recognizing all domains of the protein equally. The antibodies induced by DNA vaccination were directed almost extensively to conformational epitopes so that reactivity with native MSP4 was abolished after disulfide bonds in the protein were disrupted. Antibodies induced by recombinant proteins recognized linear epitopes as well and reactivity to native MSP4 was preserved after reduction and alkylation of parasite proteins.


Subject(s)
Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Protozoan/biosynthesis , COS Cells , Epitopes/genetics , Female , Gene Expression , Genes, Protozoan , Humans , Malaria Vaccines/genetics , Mice , Mice, Inbred C3H , Transfection , Vaccines, DNA/genetics , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
14.
Infect Immun ; 68(12): 6618-23, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11083773

ABSTRACT

Transmission-blocking vaccines are one strategy for controlling malaria, whereby sexual-stage parasites are inhibited from infecting mosquitoes by human antibodies. To evaluate whether the recently cloned Plasmodium vivax proteins Pvs25 and Pvs28 are candidates for a transmission-blocking vaccine, the molecules were expressed in yeast as secreted recombinant proteins. Mice vaccinated with these proteins adsorbed to aluminum hydroxide developed strong antibody responses against the immunogens, although for Pvs28, this response was genetically restricted. Antisera against both recombinant Pvs25 and Pvs28 recognized the corresponding molecules expressed by cultured sexual-stage parasites isolated from patients with P. vivax malaria. The development of malaria parasites in mosquitoes was completely inhibited when these antisera were ingested with the infected blood meal. Pvs25 and Pvs28, expressed in Saccharomyces cerevisiae, are as yet the only fully characterized transmission-blocking vaccine candidates against P. vivax that induce such a potent antiparasite response.


Subject(s)
Antibodies, Protozoan/therapeutic use , Antigens, Protozoan/immunology , Antigens, Surface/immunology , Culicidae/parasitology , Malaria Vaccines/immunology , Plasmodium vivax/immunology , Amino Acid Sequence , Animals , Antigens, Protozoan/genetics , Antigens, Surface/genetics , Cloning, Molecular , Immune Sera/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Pan troglodytes , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification
15.
Infect Immun ; 68(10): 5530-8, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10992450

ABSTRACT

Each of the four epidermal growth factor (EGF)-like domains of the Plasmodium falciparum sexual-stage antigen Pfs25 has been individually expressed as a yeast-secreted recombinant protein (yEGF1 through yEGF4). All four are recognized by the immune sera of animals and humans vaccinated with TBV25H (the corresponding yeast-secreted full-length recombinant form of Pfs25), with antibody titers to yEGF1 and yEGF2 weakly correlating with the ability of the sera to block the transmission of parasites to the mosquito host. All four proteins are poorly immunogenic in mice vaccinated with aluminum hydroxide-absorbed formulations. However, all four successfully primed the mice to mount an effective secondary antibody response after a single boost with TBV25H. Sera from mice vaccinated with yEGF2-TBV25H completely block the development of oocysts in mosquito midguts in membrane-feeding assays. Further, of the four proteins, only the depletion of antibodies to yEGF2 from the sera of rabbits vaccinated with TBV25H consistently abolished the ability of those sera to block oocyst development. Thus, antibodies to the second EGF-like domain of Pfs25 appear to mediate a very potent blocking activity, even at low titers. Vaccination strategies that target antibody response towards this domain may improve the efficacy of future transmission-blocking vaccines.


Subject(s)
Antibodies, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/transmission , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/blood , Culicidae/parasitology , Epidermal Growth Factor/chemistry , Epidermal Growth Factor/genetics , Epidermal Growth Factor/immunology , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/prevention & control , Mice , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Rabbits , Recombinant Proteins/immunology , Vaccination , Vaccines, Synthetic/immunology
17.
Parasite Immunol ; 22(5): 211-21, 2000 May.
Article in English | MEDLINE | ID: mdl-10792760

ABSTRACT

The merozoite surface protein 2 of P. falciparum is highly polymorphic in nature, but has regions of almost complete conservation at its N- and C-termini. We produced a chimeric recombinant protein comprising these regions only (hereafter termed NC). Mice immunized with the NC antigen produce antibodies at levels comparable to those immunized with 1624, a full-length recombinant protein representing MSP2 from P. falciparum. Antisera raised against NC recognized P. falciparum schizonts by IFA and a P. falciparum protein of Mr 45 kDa by Western blot. However, antibody specificities were observed to differ between anti-NC and anti-1624 sera, and this resulted in differences in parasite recognition, despite similar levels of antibodies having been produced. The response to the NC antigen was also shown to be restricted in some mice (H2-d), but this was overcome by including appropriate T-cell help, which was accomplished by creating recombinant protein chimeras that contained NC and T-helper epitopes from Tetanus toxoid, or MSP119 from P. berghei.


Subject(s)
Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Recombinant Fusion Proteins/immunology , Adolescent , Adult , Amino Acid Sequence , Animals , Antibody Specificity , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Child , Child, Preschool , Epitope Mapping , Humans , Immunization , Infant , Malaria, Falciparum/immunology , Mice , Mice, Inbred BALB C , Middle Aged , Molecular Sequence Data , Peptides/chemistry , Peptides/immunology , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics
18.
Vaccine ; 18(18): 1925-31, 2000 Mar 17.
Article in English | MEDLINE | ID: mdl-10699342

ABSTRACT

A placebo controlled, randomised, double blind trial was conducted in human volunteers to test a mixture of three recombinant Plasmodium falciparum blood stage antigens for its ability to reduce the initial growth rates of parasites. The vaccine contained recombinant MSP2 (3D7 allele), a portion of MSP1 (190LCS.T3) and part of the RESA antigen (C terminal 771 amino acids) in the Montanide ISA 720 adjuvant (SEPPIC). Twelve volunteers received two doses of the vaccine, 6 weeks apart. The five participants in the placebo group received an equivalent volume of the adjuvant emulsion using the same schedule. Antibody responses were low, as has been reported in earlier studies with this combination, while T cell responses were stronger. All the volunteers were challenged with approximately 140 ring infected red cells of the 3D7 cloned line, 4 weeks after the second dose. Parasitaemia was determined once daily from day 4 using a sensitive and quantitative PCR assay. All the volunteers were infected and were treated on day 8, before any developed symptoms. There was no significant difference in initial parasite growth rates between the verum and placebo groups, nor was there any significant correlation between parasite growth rates and any of the measured immunological responses. These results suggest that the formulation tested in this trial did not generate immune responses that were strong enough to reduce parasite growth in naive volunteers.


Subject(s)
Malaria Vaccines/immunology , Malaria/immunology , Plasmodium falciparum/growth & development , Plasmodium falciparum/immunology , Vaccines, DNA/immunology , Adult , Animals , Antibodies, Protozoan/biosynthesis , Double-Blind Method , Female , Humans , Lymphocyte Activation/immunology , Malaria Vaccines/adverse effects , Male , Middle Aged , Placebo Effect , T-Lymphocytes/immunology
19.
Infect Immun ; 68(4): 2231-6, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10722624

ABSTRACT

The Plasmodium falciparum rhoptry-associated proteins 1 and 2 (RAP1 and RAP2) are candidate antigens for a subunit malaria vaccine. The design of the study, which looks at the acquisition of immunity to malaria from childhood to old age, has allowed us to document the interaction of HLA and age on levels of antibody to specific malarial antigens. Antibodies reach maximum levels to RAP1 after the age of 15 but to RAP2 only after the age of 30. The effect of HLA-DRB1 and -DQB1 and age on levels of antibody to rRAP1 and rRAP2 was analyzed with a multiple regression model in which all HLA alleles and age were independent variables. DQB1*0301 and -*03032 showed an age-dependent association with levels of antibody to rRAP1, being significant in children 5 to 15 years (P < 0.001) but not in individuals over 15 years of age. DRB1*03011 showed an age-dependent association with antibody levels to rRAP2; however, this association was in adults over the age of 30 years (P < 0.01) but not in individuals under the age of 30 years. No associations were detected between DRB1 alleles and RAP1 antibody levels or between DQB1 alleles and RAP2 antibody levels. Thus, not only the HLA allele but also the age at which an interaction is manifested varies for different malarial antigens. The interaction may influence either the rate of acquisition of antibody or the final level of antibody acquired by adults.


Subject(s)
Antibodies, Protozoan/immunology , HLA-DQ Antigens/metabolism , HLA-DR Antigens/metabolism , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Adolescent , Adult , Age Factors , Aged , Alleles , Animals , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , HLA-DRB1 Chains , Hemoglobin, Sickle/genetics , Hemoglobin, Sickle/immunology , Humans , Immunity , Malaria, Falciparum/genetics , Malaria, Falciparum/immunology , Middle Aged , Polymorphism, Genetic , Protozoan Proteins/blood , Recombinant Proteins/immunology
20.
Am J Trop Med Hyg ; 62(4): 466-79, 2000 Apr.
Article in English | MEDLINE | ID: mdl-11220762

ABSTRACT

A vaccine trial was conducted with rhoptry-associated proteins 1 and 2 (RAP1 and RAP2) of Plasmodium falciparum in Saimiri boliviensis monkeys to compare the ability of parasite-derived (PfRAP1 and 2) and recombinant proteins (rRAP1 and 2) to induce protective immune responses and to find adjuvants suitable for use in humans. Eight groups of 6 monkeys each were immunized with parasite-derived or recombinant RAP1 and 2 with Freund's complete adjuvant (FCA) followed by Freund's incomplete adjuvant (FIA), Montanide ISA720 adjuvant, or CRL1005 adjuvant. Recombinant RAP1 and RAP2 were also administered separately, with Montanide ISA720. After 3 immunizations, monkeys were challenged by iv inoculation of 50,000 parasites of the Uganda Palo Alto strain of P. falciparum. Of the animals vaccinated using FCA/FIA, 1 of 6 control monkeys, 3 of 6 immunized with PfRAP1 and 2, and 2 of 6 with rRAP1 and 2 did not require drug treatment. Of the monkeys vaccinated with Montanide ISA720 adjuvant, 0 of the 6 control monkeys, 2 of 6 immunized with RAP1 and 2, 1 of 6 immunized with rRAP1, and 4 of 6 immunized with RAP2 did not require drug treatment. Two of 6 monkeys immunized with PfRAP1 and 2 with CRL1005 did not require treatment. All groups receiving RAP1, RAP2, or both had a significant decrease in initial parasite multiplication rates and there was a significant negative correlation between anti-RAP2 antibody and multiplication rates. Animals were rechallenged with the homologous parasite 126 days after the first challenge. Of the monkeys that did not require drug treatment after the first challenge, none developed detectable parasitemia following rechallenge.


Subject(s)
Malaria Vaccines , Malaria, Falciparum/prevention & control , Parasitemia/prevention & control , Plasmodium falciparum/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Protozoan/biosynthesis , Antibodies, Protozoan/blood , Female , Malaria Vaccines/administration & dosage , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Male , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Recombinant Proteins/immunology , Saimiri
SELECTION OF CITATIONS
SEARCH DETAIL
...