Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Vaccine ; 35(24): 3232-3238, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28479180

ABSTRACT

Pfs25, a Plasmodium falciparum surface protein expressed during zygote and ookinete stages in infected mosquitoes, is a lead transmission-blocking vaccine candidate against falciparum malaria. To enhance immunogenicity, recombinant Pfs25 was chemically conjugated to recombinant nontoxic Pseudomonas aeruginosa ExoProtein A (rEPA) in conformance with current good manufacturing practices (cGMP), and formulated with the alum adjuvant Alhydrogel. In order to meet the regulatory requirements for a phase 1 human clinical trial, the vaccine product was extensively evaluated for stability at an initial time point and through the clinical trial period annually. Because basic quality control methods to characterize alum-based vaccines remain unavailable, a thermal forced degradation study was performed prior to the initial evaluation to identify the methods suitable to detect the quality of vaccine formulations. Our results show that the vaccine product Pfs25-EPA formulated on Alhydrogel is in conformance with regulatory guidelines and suitable for human trials.


Subject(s)
Aluminum Hydroxide/immunology , Malaria Vaccines/immunology , Protozoan Proteins/immunology , Vaccine Potency , Adjuvants, Immunologic , Aluminum/analysis , Aluminum Hydroxide/chemistry , Animals , Clinical Trials, Phase I as Topic , Drug Compounding , Malaria Vaccines/administration & dosage , Malaria Vaccines/chemistry , Malaria, Falciparum/prevention & control , Mice , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/genetics , Recombinant Proteins/immunology , Temperature
2.
PLoS One ; 11(10): e0163144, 2016.
Article in English | MEDLINE | ID: mdl-27749907

ABSTRACT

Transmission-blocking vaccines (TBVs) that target sexual stage parasite development could be an integral part of measures for malaria elimination. Pfs25 is a leading TBV candidate, and previous studies conducted in animals demonstrated an improvement of its functional immunogenicity after conjugation to EPA, a recombinant, detoxified ExoProtein A from Pseudomonas aeruginosa. In this report, we describe results of an open-label, dose-escalating Phase 1 trial to assess the safety and immunogenicity of Pfs25-EPA conjugates formulated with Alhydrogel®. Thirty malaria-naïve healthy adults received up to four doses of the conjugate vaccine, with 8, 16, or 47 µg of conjugated Pfs25 mass, at 0, 2, 4, and 10 months. Vaccinations were generally well tolerated. The majority of solicited adverse events were mild in severity with pain at the injection site the most common complaint. Anemia was the most common laboratory abnormality, but was considered possibly related to the study in only a minority of cases. No vaccine-related serious adverse events occurred. The peak geometric mean anti-Pfs25 antibody level in the highest dose group was 88 (95% CI 53, 147) µg/mL two weeks after the 4th vaccination, and declined to near baseline one year later. Antibody avidity increased over successive vaccinations. Transmission blocking activity demonstrated in a standard membrane feeding assay (SMFA) also increased from the second to the third dose, and correlated with antibody titer and, after the final dose, with antibody avidity. These results support the further evaluation of Pfs25-EPA/Alhydrogel® in a malaria-endemic population.


Subject(s)
Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Recombinant Proteins/immunology , Adolescent , Adult , Antibodies, Protozoan/blood , Antibody Affinity/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Malaria Vaccines/adverse effects , Malaria Vaccines/chemistry , Malaria, Falciparum/immunology , Malaria, Falciparum/transmission , Microscopy, Fluorescence , Middle Aged , Pain/etiology , Protozoan Proteins/adverse effects , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Recombinant Proteins/adverse effects , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Vaccines, Conjugate/immunology , Young Adult
3.
J Biol Chem ; 291(38): 19913-22, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27432885

ABSTRACT

Development of a Plasmodium falciparum (Pf) transmission blocking vaccine (TBV) has the potential to significantly impact malaria control. Antibodies elicited against sexual stage proteins in the human bloodstream are taken up with the blood meal of the mosquitoes and inactivate parasite development in the mosquito. In a phase 1 trial, a leading TBV identified as Pfs25-EPA/Alhydrogel® appeared safe and immunogenic, however, the level of Pfs25-specific antibodies were likely too low for an effective vaccine. Pfs230, a 230-kDa sexual stage protein expressed in gametocytes is an alternative vaccine candidate. A unique 6-cysteine-rich domain structure within Pfs230 have thwarted its recombinant expression and characterization for clinical evaluation for nearly a quarter of a century. Here, we report on the identification, biochemical, biophysical, and immunological characterization of recombinant Pfs230 domains. Rabbit antibodies generated against recombinant Pfs230 domains blocked mosquito transmission of a laboratory strain and two field isolates using an ex vivo assay. A planned clinical trial of the Pfs230 vaccine is a significant step toward the potential development of a transmission blocking vaccine to eliminate malaria.


Subject(s)
Antigens, Protozoan/chemistry , Antigens, Protozoan/immunology , Malaria Vaccines/chemistry , Malaria Vaccines/immunology , Plasmodium falciparum/chemistry , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/pharmacology , Humans , Malaria Vaccines/genetics , Malaria Vaccines/pharmacology , Malaria, Falciparum/genetics , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Plasmodium falciparum/genetics , Protein Domains , Protozoan Proteins/genetics , Protozoan Proteins/pharmacology , Rabbits
4.
J Infect Dis ; 214(4): 577-86, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27190180

ABSTRACT

BACKGROUND: Placental malaria is caused by Plasmodium falciparum-infected erythrocytes (IEs) that surface-express VAR2CSA and bind chondroitin sulfate A. The inflammatory response to placenta-sequestered parasites is associated with poor pregnancy outcomes, and protection may be mediated in part by VAR2CSA antibodies that block placental IE adhesion. METHODS: In this study, we used a new approach to assess VAR2CSA domains for functional epitopes recognized by naturally acquired antibodies. Antigen-specific immunoglobulin (Ig) G targeting Duffy binding-like (DBL) domains from different alleles were sequentially purified from plasma pooled from multigravid women and then characterized using enzyme-linked immunosorbent assay, flow cytometry, and antiadhesion assays. RESULTS: Different DBL domain-specific IgGs could react to homologous as well as heterologous antigens and parasites, suggesting that conserved epitopes are shared between allelic variants. Homologous blocking of IE binding was observed with ID1-DBL2-ID2a-, DBL4-, and DBL5-specific IgG (range, 42%-75%), whereas partial cross-inhibition activity was observed with purified IgG specific to ID1-DBL2-ID2a and DBL4 antigens. Plasma retained broadly neutralizing activity after complete depletion of these VAR2CSA specificities. CONCLUSIONS: Broadly neutralizing antibodies of multigravidae are not depleted on VAR2CSA recombinant antigens, and hence development of VAR2CSA vaccines based on a single construct and variant might induce antibodies with limited broadly neutralizing activity.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Malaria, Falciparum/immunology , Placenta Diseases/immunology , Plasmodium falciparum/immunology , Cell Adhesion , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes, B-Lymphocyte , Female , Flow Cytometry , Humans , Malaria Vaccines/immunology , Pregnancy
5.
Infect Immun ; 83(10): 3771-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26169272

ABSTRACT

The extended rod-like Plasmodium falciparum circumsporozoite protein (CSP) is comprised of three primary domains: a charged N terminus that binds heparan sulfate proteoglycans, a central NANP repeat domain, and a C terminus containing a thrombospondin-like type I repeat (TSR) domain. Only the last two domains are incorporated in RTS,S, the leading malaria vaccine in phase 3 trials that, to date, protects about 50% of vaccinated children against clinical disease. A seroepidemiological study indicated that the N-terminal domain might improve the efficacy of a new CSP vaccine. Using a panel of CSP-specific monoclonal antibodies, well-characterized recombinant CSPs, label-free quantitative proteomics, and in vitro inhibition of sporozoite invasion, we show that native CSP is N-terminally processed in the mosquito host and undergoes a reversible conformational change to mask some epitopes in the N- and C-terminal domains until the sporozoite interacts with the liver hepatocyte. Our findings show the importance of understanding processing and the biophysical change in conformation, possibly due to a mechanical or molecular signal, and may aid in the development of a new CSP vaccine.


Subject(s)
Malaria, Falciparum/parasitology , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Sporozoites/immunology , Animals , Anopheles/parasitology , Antibodies, Protozoan/immunology , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Hepatocytes/immunology , Hepatocytes/parasitology , Humans , Malaria, Falciparum/immunology , Plasmodium falciparum/chemistry , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protein Conformation , Protein Structure, Tertiary , Protozoan Proteins/genetics , Sporozoites/chemistry , Sporozoites/growth & development
6.
Vaccine ; 31(28): 2954-62, 2013 Jun 19.
Article in English | MEDLINE | ID: mdl-23623858

ABSTRACT

Successful efforts to control infectious diseases have often required the use of effective vaccines. The current global strategy for control of malaria, including elimination and eradication will also benefit from the development of an effective vaccine that interrupts malaria transmission. To this end, a vaccine that disrupts malaria transmission within the mosquito host has been investigated for several decades targeting a 25 kDa ookinete specific surface protein, identified as Pfs25. Phase 1 human trial results using a recombinant Pfs25H/Montanide ISA51 formulation demonstrated that human Pfs25 specific antibodies block parasite infectivity to mosquitoes; however, the extent of blocking was likely insufficient for an effective transmission blocking vaccine. To overcome the poor immunogenicity, processes to produce and characterize recombinant Pfs25H conjugated to a detoxified form of Pseudomonas aeruginosa exoprotein A (EPA) have been developed and used to manufacture a cGMP pilot lot for use in human clinical trials. The Pfs25-EPA conjugate appears as a nanoparticle with an average molar mass in solution of approximately 600 kDa by static light scattering with an average diameter 20 nm (range 10-40 nm) by dynamic light scattering. The molar ratio of Pfs25H to EPA is about 3 to 1 by amino acid analysis, respectively. Outbred mice immunized with the Pfs25-EPA conjugated nanoparticle formulated on Alhydrogel(®) had a 75-110 fold increase in Pfs25H specific antibodies when compared to an unconjugated Pfs25H/Alhydrogel(®) formulation. A phase 1 human trial using the Pfs25-EPA/Alhydrogel(®) formulation is ongoing in the United States.


Subject(s)
Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Recombinant Proteins/immunology , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/immunology , Aluminum Hydroxide , Animals , Bacterial Toxins/chemistry , Bacterial Toxins/immunology , Exotoxins/chemistry , Exotoxins/immunology , Female , Humans , Malaria Vaccines/chemistry , Malaria, Falciparum/immunology , Malaria, Falciparum/transmission , Mice , Nanoparticles , Protein Conformation , Protozoan Proteins/chemistry , Pseudomonas aeruginosa , Recombinant Proteins/chemistry , Vaccination , Vaccines, Conjugate/chemistry , Vaccines, Conjugate/immunology , Virulence Factors/chemistry , Virulence Factors/immunology , Pseudomonas aeruginosa Exotoxin A
7.
PLoS One ; 8(2): e56326, 2013.
Article in English | MEDLINE | ID: mdl-23457550

ABSTRACT

The malaria parasite, Plasmodium falciparum, and related parasites use a variety of proteins with Duffy-Binding Like (DBL) domains to bind glycoproteins on the surface of host cells. Among these proteins, the 175 kDa erythrocyte binding antigen, EBA-175, specifically binds to glycophorin A on the surface of human erythrocytes during the process of merozoite invasion. The domain responsible for glycophorin A binding was identified as region II (RII) which contains two DBL domains, F1 and F2. The crystal structure of this region revealed a dimer that is presumed to represent the glycophorin A binding conformation as sialic acid binding sites and large cavities are observed at the dimer interface. The dimer interface is largely composed of two loops from within each monomer, identified as the F1 and F2 ß-fingers that contact depressions in the opposing monomers in a similar manner. Previous studies have identified a panel of five monoclonal antibodies (mAbs) termed R215 to R218 and R256 that bind to RII and inhibit invasion of erythrocytes to varying extents. In this study, we predict the F2 ß-finger region as the conformational epitope for mAbs, R215, R217, and R256, and confirm binding for the most effective blocking mAb R217 and R215 to a synthetic peptide mimic of the F2 ß-finger. Localization of the epitope to the dimerization and glycan binding sites of EBA-175 RII and site-directed mutagenesis within the predicted epitope are consistent with R215 and R217 blocking erythrocyte invasion by Plasmodium falciparum by preventing formation of the EBA-175- glycophorin A complex.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Protozoan/immunology , Epitope Mapping , Erythrocytes/microbiology , Glycophorins/metabolism , Plasmodium falciparum/physiology , Protein Multimerization , Protozoan Proteins/immunology , Antibody Specificity , Antigens, Protozoan/chemistry , Antigens, Protozoan/metabolism , Binding Sites , Computational Biology , Glycophorins/chemistry , Humans , Models, Molecular , Peptide Fragments/immunology , Peptide Library , Plasmodium falciparum/immunology , Plasmodium falciparum/metabolism , Polysaccharides/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism
8.
PLoS One ; 7(10): e46094, 2012.
Article in English | MEDLINE | ID: mdl-23056238

ABSTRACT

UNLABELLED: A Phase 1 dose escalating study was conducted in malaria naïve adults to assess the safety, reactogenicity, and immunogenicity of the blood stage malaria vaccine BSAM2/Alhydrogel®+ CPG 7909. BSAM2 is a combination of the FVO and 3D7 alleles of recombinant AMA1 and MSP1(42), with equal amounts by weight of each of the four proteins mixed, bound to Alhydrogel®, and administered with the adjuvant CPG 7909. Thirty (30) volunteers were enrolled in two dose groups, with 15 volunteers receiving up to three doses of 40 µg total protein at Days 0, 56, and 180, and 15 volunteers receiving up to three doses of 160 µg protein on the same schedule. Most related adverse events were mild or moderate, but 4 volunteers experienced severe systemic reactions and two were withdrawn from vaccinations due to adverse events. Geometric mean antibody levels after two vaccinations with the high dose formulation were 136 µg/ml for AMA1 and 78 µg/ml for MSP1(42). Antibody responses were not significantly different in the high dose versus low dose groups and did not further increase after third vaccination. In vitro growth inhibition was demonstrated and was closely correlated with anti-AMA1 antibody responses. A Phase 1b trial in malaria-exposed adults is being conducted. TRIAL REGISTRATION: Clinicaltrials.gov NCT00889616.


Subject(s)
Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Vaccination/methods , Adolescent , Adult , Aluminum Hydroxide/immunology , Antibodies, Protozoan/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Dose-Response Relationship, Drug , Drug Administration Schedule , Erythrocytes/parasitology , Female , Follow-Up Studies , Headache/chemically induced , Humans , Malaria Vaccines/therapeutic use , Malaria, Falciparum/parasitology , Malaria, Falciparum/prevention & control , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Merozoite Surface Protein 1/genetics , Merozoite Surface Protein 1/immunology , Middle Aged , Oligodeoxyribonucleotides/immunology , Plasmodium falciparum/growth & development , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Treatment Outcome , Vaccination/adverse effects , Young Adult
9.
PLoS One ; 7(6): e36996, 2012.
Article in English | MEDLINE | ID: mdl-22675476

ABSTRACT

Self-associated protein aggregates or cross-linked protein conjugates are, in general, more immunogenic than oligomeric or monomeric forms. In particular, the immunogenicity in mice of a recombinant malaria transmission blocking vaccine candidate, the ookinete specific Plasmodium falciparum 25 kDa protein (Pfs25), was increased more than 1000-fold when evaluated as a chemical cross-linked protein-protein conjugate as compared to a formulated monomer. Whether alternative approaches using protein complexes improve the immunogenicity of other recombinant malaria vaccine candidates is worth assessing. In this work, the immunogenicity of the recombinant 42 kDa processed form of the P. falciparum merozoite surface protein 1 (MSP1(42)) was evaluated as a self-associated, non-covalent aggregate and as a chemical cross-linked protein-protein conjugate to ExoProtein A, which is a recombinant detoxified form of Pseudomonas aeruginosa exotoxin A. MSP1(42) conjugates were prepared and characterized biochemically and biophysically to determine their molar mass in solution and stoichiometry, when relevant. The immunogenicity of the MSP1(42) self-associated aggregates, cross-linked chemical conjugates and monomers were compared in BALB/c mice after adsorption to aluminum hydroxide adjuvant, and in one instance in association with the TLR9 agonist CPG7909 with an aluminum hydroxide formulation. Antibody titers were assessed by ELISA. Unlike observations made for Pfs25, no significant enhancement in MSP1(42) specific antibody titers was observed for any conjugate as compared to the formulated monomer or dimer, except for the addition of the TLR9 agonist CPG7909. Clearly, enhancing the immunogenicity of a recombinant protein vaccine candidate by the formation of protein complexes must be established on an empirical basis.


Subject(s)
Cross-Linking Reagents/metabolism , Merozoite Surface Protein 1/chemistry , Merozoite Surface Protein 1/immunology , Plasmodium falciparum/immunology , Animals , Antibodies, Protozoan/immunology , Bacterial Proteins/immunology , Chromatography, Gel , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Immunoglobulin G/immunology , Light , Mice , Mice, Inbred BALB C , Molecular Weight , Protein Isoforms/chemistry , Protein Isoforms/immunology , Protein Multimerization , Protein Structure, Quaternary , Recombinant Proteins , Scattering, Radiation , Titrimetry
10.
Am J Trop Med Hyg ; 86(4): 573-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22492138

ABSTRACT

Laboratory capacity in the developing world frequently lacks quality management systems (QMS) such as good clinical laboratory practices, proper safety precautions, and adequate facilities; impacting the ability to conduct biomedical research where it is needed most. As the regulatory climate changes globally, higher quality laboratory support is needed to protect study volunteers and to accurately assess biological parameters. The University of Bamako and its partners have undertaken a comprehensive QMS plan to improve quality and productivity using the Clinical and Laboratory Standards Institute standards and guidelines. The clinical laboratory passed the College of American Pathologists inspection in April 2010, and received full accreditation in June 2010. Our efforts to implement high-quality standards have been valuable for evaluating safety and immunogenicity of malaria vaccine candidates in Mali. Other disease-specific research groups in resource-limited settings may benefit by incorporating similar training initiatives, QMS methods, and continual improvement practices to ensure best practices.


Subject(s)
Accreditation , Clinical Laboratory Techniques/standards , Laboratories/standards , Africa South of the Sahara , Biomedical Research , Clinical Trials as Topic , Enzyme-Linked Immunosorbent Assay/methods , Humans , Laboratories/organization & administration , Mali , Reproducibility of Results , Total Quality Management/methods
11.
PLoS One ; 6(7): e22271, 2011.
Article in English | MEDLINE | ID: mdl-21799809

ABSTRACT

BACKGROUND: Inhibition of parasite growth is a major objective of blood-stage malaria vaccines. The in vitro assay of parasite growth inhibitory activity (GIA) is widely used as a surrogate marker for malaria vaccine efficacy in the down-selection of candidate blood-stage vaccines. Here we report the first study to examine the relationship between in vivo Plasmodium falciparum growth rates and in vitro GIA in humans experimentally infected with blood-stage malaria. METHODS: In this phase I/IIa open-label clinical trial five healthy malaria-naive volunteers were immunised with AMA1/C1-Alhydrogel+CPG 7909, and together with three unvaccinated controls were challenged by intravenous inoculation of P. falciparum infected erythrocytes. RESULTS: A significant correlation was observed between parasite multiplication rate in 48 hours (PMR) and both vaccine-induced growth-inhibitory activity (Pearson r = -0.93 [95% CI: -1.0, -0.27] P = 0.02) and AMA1 antibody titres in the vaccine group (Pearson r = -0.93 [95% CI: -0.99, -0.25] P = 0.02). However immunisation failed to reduce overall mean PMR in the vaccine group in comparison to the controls (vaccinee 16 fold [95% CI: 12, 22], control 17 fold [CI: 0, 65] P = 0.70). Therefore no impact on pre-patent period was observed (vaccine group median 8.5 days [range 7.5-9], control group median 9 days [range 7-9]). CONCLUSIONS: Despite the first observation in human experimental malaria infection of a significant association between vaccine-induced in vitro growth inhibitory activity and in vivo parasite multiplication rate, this did not translate into any observable clinically relevant vaccine effect in this small group of volunteers. TRIAL REGISTRATION: ClinicalTrials.gov [NCT00984763].


Subject(s)
Adjuvants, Immunologic , Malaria Vaccines/immunology , Malaria/prevention & control , Malaria/parasitology , Plasmodium falciparum/growth & development , Plasmodium falciparum/immunology , Vaccination/methods , Adjuvants, Immunologic/adverse effects , Adolescent , Adult , Aluminum Hydroxide/immunology , Antibodies/immunology , Antigens, Protozoan/immunology , Female , Humans , Malaria Vaccines/adverse effects , Male , Membrane Proteins/immunology , Middle Aged , Oligodeoxyribonucleotides/immunology , Protozoan Proteins/immunology , Vaccination/adverse effects , Young Adult
12.
PLoS One ; 6(6): e20947, 2011.
Article in English | MEDLINE | ID: mdl-21695140

ABSTRACT

BACKGROUND: Apical membrane antigen 1 (AMA1) is one of the best-studied blood-stage malaria vaccine candidates. When an AMA1 vaccine was tested in a malaria naïve population, it induced functionally active antibodies judged by Growth Inhibition Assay (GIA). However, the same vaccine failed to induce higher growth-inhibitory activity in adults living in a malaria endemic area. Vaccination did induce functionally active antibodies in malaria-exposed children with less than 20% inhibition in GIA at baseline, but not in children with more than that level of baseline inhibition. METHODS: Total IgGs were purified from plasmas collected from the pediatric trial before and after immunization and pools of total IgGs were made. Another set of total IgGs was purified from U.S. adults immunized with AMA1 (US-total IgG). From these total IgGs, AMA1-specific and non-AMA1 IgGs were affinity purified and the functional activity of these IgGs was evaluated by GIA. Competition ELISA was performed with the U.S.-total IgG and non-AMA1 IgGs from malaria-exposed children. RESULTS: AMA1-specific IgGs from malaria-exposed children and U.S. vaccinees showed similar growth-inhibitory activity at the same concentrations. When mixed with U.S.-total IgG, non-AMA1 IgGs from children showed an interference effect in GIA. Interestingly, the interference effect was higher with non-AMA1 IgGs from higher titer pools. The non-AMA1 IgGs did not compete with anti-AMA1 antibody in U.S.-total IgG in the competition ELISA. CONCLUSION: Children living in a malaria endemic area have a fraction of IgGs that interferes with the biological activity of anti-AMA1 antibody as judged by GIA. While the mechanism of interference is not resolved in this study, these results suggest it is not caused by direct competition between non-AMA1 IgG and AMA1 protein. This study indicates that anti-malaria IgGs induced by natural exposure may interfere with the biological effect of antibody induced by an AMA1-based vaccine in the target population.


Subject(s)
Antigens, Protozoan/immunology , Immunoglobulin G/immunology , Membrane Proteins/immunology , Plasmodium falciparum/growth & development , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Adolescent , Adult , Antibody Specificity , Child , Humans , Immunization , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Mali , Middle Aged , Young Adult
13.
Vaccine ; 29(20): 3640-5, 2011 May 09.
Article in English | MEDLINE | ID: mdl-21440641

ABSTRACT

Plasmodium falciparum apical membrane antigen 1 (AMA1) is an asexual blood-stage vaccine candidate against the malaria parasite. AMA1-C1/ISA 720 refers to a mixture of recombinant AMA1 proteins representing the FVO and 3D7 alleles in 1:1 mass ratio, formulated with Montanide(®) ISA 720 as a water-in oil emulsion. In order to develop the AMA1-C1/ISA 720 vaccine for human use, it was important to determine the shelf life of this formulation. Previously it was found 267 mM glycine stabilized the proteins in Montanide(®) ISA 720 formulations for a short period of time at 2-8°C [25]. We now test the long term stability of AMA1-C1 at 10 and 40 µg/mL formulated with Montanide(®) ISA 720 with 50mM glycine as a stabilizer. Stability of AMA1-C1/ISA 720 at different time points following formulation (0, 5, 12 or 18 months) was evaluated by determining the mean particle size (diameter of the mean droplet volume), total protein content by a Modified Lowry assay, identity and integrity using western blot and SDS-PAGE. Our results showed that the mean particle size of these emulsions increased over time, whereas protein content, as determined by an ELISA method using a monoclonal antibody against penta-his, decreased over time. For the 10 µg/mL AMA1-C1/ISA 720 vaccine, the protein content was 6.5±2.2 µg/mL, and for the 40 µg/mL AMA1-C1/ISA 720 vaccine, the protein content was only 8.2±2.3 µg/mL after 18 months of storage at 2-8°C. These results suggest that the integrity of the protein was affected by long-term storage. The results of the present study indicate that the AMA1-C1/ISA 720 emulsion was unstable after 12 months of storage, after which AMA1-C1 proteins were partially degraded.


Subject(s)
Antigens, Protozoan/immunology , Glycine/chemistry , Malaria Vaccines/chemistry , Mannitol/analogs & derivatives , Membrane Proteins/immunology , Oleic Acids/chemistry , Protozoan Proteins/immunology , Adjuvants, Immunologic/chemistry , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/administration & dosage , Drug Stability , Drug Storage , Female , Glycine/immunology , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Mannitol/chemistry , Mannitol/immunology , Membrane Proteins/administration & dosage , Mice , Mice, Inbred BALB C , Oleic Acids/immunology , Particle Size , Plasmodium falciparum/immunology , Protozoan Proteins/administration & dosage , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/immunology
14.
Vaccine ; 29(12): 2255-61, 2011 Mar 09.
Article in English | MEDLINE | ID: mdl-21277408

ABSTRACT

Clinical development of malaria vaccines progresses from trials in malaria naïve adults to malaria exposed adults followed by malaria exposed children. It is not well known whether immune responses in non-target populations are predictive of those in target populations, particularly in African children. Therefore humoral responses in three different populations (U.S. adults, Malian adults and Malian children) were compared in this study. They were immunized with 80 µg of Apical Membrane Antigen 1 (AMA1)/alhydrogel on days 0 and 28. Sera were collected on days 0 and 42; antibody levels were determined by ELISA and the functionality of antibodies was evaluated by Growth Inhibition Assay. After immunization, there was no significant difference in antibody levels between the Malian children and the Malian adults, but U.S. adults showed lower antibody levels. Vaccination did not significantly change growth-inhibitory activity in Malian adults, but inhibition increased significantly in both U.S. adults and Malian children. Vaccine-induced inhibitory activity was reversed by pre-incubation with AMA1 protein, but pre-existing infection-induced inhibition was not. This study shows that humoral responses elicited by the AMA1 vaccine varied depending on the population, most likely reflecting different levels of previous malaria exposure. Thus predicting immune responses from non-target populations is not desirable.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Membrane Proteins/immunology , Protozoan Proteins/immunology , Adolescent , Adult , Antibodies, Protozoan/blood , Child, Preschool , Clinical Trials as Topic , Enzyme-Linked Immunosorbent Assay , Humans , Immunity, Humoral , Infant , Malaria, Falciparum/immunology , Mali , Middle Aged , Plasmodium falciparum/immunology , United States , Young Adult
15.
Microbes Infect ; 11(3): 408-12, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19146977

ABSTRACT

In this paper we report our efforts to enhance the immunogenicity of Pfs28, a transmission blocking vaccine candidate of Plasmodium falciparum, using a strategy of chemical conjugation. With an improved procedure, Pfs28 was covalently coupled to the mutant and non-toxic ExoProtein A of Pseudomonas aeruginosa by the reaction between thiolated antigen and maleimide modified carrier protein. The optimized process resulted in a higher antigen-carrier conjugation ratio, and the conjugation product could be purified using single-step size-exclusion chromatography. A significant increase in immunogenicity measured by ELISA was observed in mice immunized with conjugated Pfs28 as compared to unconjugated Pfs28.


Subject(s)
Antigens, Protozoan/immunology , Bacterial Proteins/immunology , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Pseudomonas aeruginosa/immunology , Animals , Antibodies, Protozoan/blood , Enzyme-Linked Immunosorbent Assay , Mice , Vaccines, Conjugate/immunology
16.
PLoS One ; 3(7): e2636, 2008 Jul 09.
Article in English | MEDLINE | ID: mdl-18612426

ABSTRACT

BACKGROUND: Pfs25 and Pvs25, surface proteins of mosquito stage of the malaria parasites P. falciparum and P. vivax, respectively, are leading candidates for vaccines preventing malaria transmission by mosquitoes. This single blinded, dose escalating, controlled Phase 1 study assessed the safety and immunogenicity of recombinant Pfs25 and Pvs25 formulated with Montanide ISA 51, a water-in-oil emulsion. METHODOLOGY/PRINCIPAL FINDINGS: The trial was conducted at The Johns Hopkins Center for Immunization Research, Washington DC, USA, between May 16, 2005-April 30, 2007. The trial was designed to enroll 72 healthy male and non-pregnant female volunteers into 1 group to receive adjuvant control and 6 groups to receive escalating doses of the vaccines. Due to unexpected reactogenicity, the vaccination was halted and only 36 volunteers were enrolled into 4 groups: 3 groups of 10 volunteers each were immunized with 5 microg of Pfs25/ISA 51, 5 microg of Pvs25/ISA 51, or 20 microg of Pvs25/ISA 51, respectively. A fourth group of 6 volunteers received adjuvant control (PBS/ISA 51). Frequent local reactogenicity was observed. Systemic adverse events included two cases of erythema nodosum considered to be probably related to the combination of the antigen and the adjuvant. Significant antibody responses were detected in volunteers who completed the lowest scheduled doses of Pfs25/ISA 51. Serum anti-Pfs25 levels correlated with transmission blocking activity. CONCLUSION/SIGNIFICANCE: It is feasible to induce transmission blocking immunity in humans using the Pfs25/ISA 51 vaccine, but these vaccines are unexpectedly reactogenic for further development. This is the first report that the formulation is associated with systemic adverse events including erythema nodosum. TRIAL REGISTRATION: ClinicalTrials.gov NCT00295581.


Subject(s)
Antigens, Protozoan/immunology , Antigens, Surface/immunology , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Mannitol/analogs & derivatives , Oleic Acids/administration & dosage , Protozoan Proteins/adverse effects , Protozoan Proteins/immunology , Recombinant Proteins/adverse effects , Recombinant Proteins/immunology , Adolescent , Adult , Animals , Antigens, Protozoan/chemistry , Antigens, Surface/chemistry , Disease Transmission, Infectious , Female , Humans , Malaria Vaccines/chemistry , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Malaria, Vivax/prevention & control , Malaria, Vivax/transmission , Male , Mannitol/administration & dosage , Mannitol/chemistry , Middle Aged , Oleic Acids/chemistry , Plasmodium falciparum/immunology , Plasmodium vivax/immunology , Protozoan Proteins/chemistry , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/chemistry
17.
Vaccine ; 25(29): 5343-7, 2007 Jul 20.
Article in English | MEDLINE | ID: mdl-17566616

ABSTRACT

CpG oligodeoxynucleotides are potent immunostimulants. In this study, CPG 7909 was formulated with the recombinant Plasmodium falciparum protein AMA1-C1 adsorbed to Alhydrogel (aluminum hydroxide) and used to immunize mice. Mice receiving free CPG 7909 in a separate same site injection to the AMA1-C1/Alhydrogel had the same antibody responses as mice receiving AMA1-C1/Alhydrogel alone. For mice immunized with CPG 7909 bound to the AMA1-C1/Alhydrogel formulation, there was a bell shaped CPG 7909 dose-response curve with the highest antibody response co-incident with the concentration of CPG 7909 that saturated binding to the Alhydrogel. At a higher CPG 7909 dose where 74% was unbound, there was no enhancement of response over AMA1-C1/Alhydrogel alone. Our results suggest that the adjuvant effects of CpGs are optimal when adsorbed to Alhydrogel and highlight the need for careful characterization of the vaccine formulation.


Subject(s)
Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Malaria/immunology , Oligodeoxyribonucleotides/immunology , Plasmodium falciparum/immunology , Animals , Dose-Response Relationship, Immunologic , Enzyme-Linked Immunosorbent Assay , Female , Immunoglobulin G/blood , Mice
18.
J Immunol Methods ; 323(2): 139-46, 2007 Jun 30.
Article in English | MEDLINE | ID: mdl-17512533

ABSTRACT

CpG oligodeoxynucleotides are potent immunostimulants. For parenterally delivered alum-based vaccines, the immunostimulatory effect of CpG depends on the association of the CpG and antigen to the alum. We describe effects of buffer components on the binding of CPG 7909 to aluminum hydroxide (Alhydrogel), assays for measuring binding of CPG 7909 to alum and CPG 7909 induced dissociation of antigen from the alum. Free CPG 7909 is a potent inducer of IP-10 in mice. However the lack of IP-10 production from formulations containing bound CPG 7909 suggested that CPG 7909 does not rapidly dissociate from the alum after injection. It also suggests that IP-10 assays are not a good basis for potency assays for alum-based vaccines containing CPG 7909.


Subject(s)
Aluminum Hydroxide/chemistry , Densitometry/methods , Mass Spectrometry/methods , Oligodeoxyribonucleotides/analysis , Vaccines/chemistry , Alum Compounds/chemistry , Animals , Antigens/chemistry , Buffers , Chemokine CXCL10 , Chemokines, CXC/blood , Mice , Oligodeoxyribonucleotides/chemistry , Phosphates/chemistry
19.
Vaccine ; 24(14): 2497-505, 2006 Mar 24.
Article in English | MEDLINE | ID: mdl-16434128

ABSTRACT

Apical membrane antigen 1 (AMA1) has been shown to be a promising malaria vaccine candidate. The multiallelic AMA1-C1 vaccine currently in Phase 1 trials in the US and Mali contains an equal mixture of the ectodomain portion of recombinant AMA1 from the FVO and 3D7 clones of Plasmodium falciparum, formulated on Alhydrogel. It is hoped that inclusion of a human-optimized CpG oligodeoxynucleotide (ODN) (CPG 7909) with our existing AMA1-C1/Alhydrogel vaccine will lead to a higher concentration of functional AMA1-C1 antibodies. Preclinical studies were performed in mice, rats and guinea pigs to assess the safety, immunogenicity and functionality of the immune response to AMA1-C1 with Alhydrogel + CPG 7909 compared to antigen with Alhydrogel alone. Day 42 mean anti-AMA1 ELISA titer values derived from individual animals were compared between Alhydrogel and Alhydrogel + CPG 7909 groups at each antigen dose for each species. Sera from Alhydrogel + CPG 7909 groups displayed significantly higher antibody titers (P < 0.025) than their comparable Alhydrogel alone group. Mouse IgG isotype analysis showed that AMA1-C1/Alhydrogel induced a predominately Th2 type response while AMA1-C1/Alhydrogel + CPG 7909 gave a mixed Th1/Th2 type response. When tested for functional activity by in vitro inhibition of parasite invasion, IgG isolated from serum pools of AMA1-C1/Alhydrogel + CPG 7909 animals was more effective against both FVO and 3D7 parasites than an equal concentration of IgG from animals receiving vaccines adjuvanted with Alhydrogel alone. These promising preclinical results have recently led to the start of a Phase 1 trial in the US.


Subject(s)
Aluminum Hydroxide/pharmacology , Antibodies, Protozoan/immunology , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Plasmodium falciparum/drug effects , Animals , Antibody Formation , Antibody Specificity , Guinea Pigs , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Mice , Oligodeoxyribonucleotides/metabolism , Rats
20.
J Virol Methods ; 123(1): 89-94, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15582703

ABSTRACT

A new method of inoculation of gladiolus with cucumber mosaic virus (CMV) was developed using the Bio-Rad Helios Gene Gun System. This method circumvents the traditional use of aphids to transmit CMV, a virus that is mechanically transmissible to many plant species but only with difficulty to gladiolus. Cartridges containing virus-coated gold microcarriers were prepared and the virus shot into Nicotianabenthamiana leaves and gladiolus corms and cormels. The biolistic procedure successfully transmitted three CMV isolates, two from serogroup I and one from serogroup II. Survival rates of two cultivars of gladiolus cormels and corms in sterile and non-sterile environments were compared. Infection rates of 100% were obtained when as little as 2 microg of virus was used in cartridge preparation. CMV remained viable after the cartridges were stored for many months at 4 degrees C.


Subject(s)
Biolistics/methods , Cucumovirus/physiology , Iridaceae/virology , Plant Diseases/virology , Iridaceae/genetics , Particle Size , Nicotiana/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...