Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.462
Filter
1.
Trop Biomed ; 41(2): 190-195, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-39154272

ABSTRACT

The Plasmodium secreted protein with an altered thrombospondin repeat (SPATR) has been known to play an important role in the malaria parasite's invasion into host erythrocytes. This protein is immunogenic and has been considered as one of the potential vaccine candidates against malaria parasite infection. Thus far, only a handful immunological studies have been carried out on P. knowlesi SPATR (PkSPATR), and none of these studies investigated the immunoprotective properties of the protein. In the present study, the ability of anti-PkSPATR antibodies to inhibit invasion of human erythrocytes was assessed in an in vitro merozoite invasion inhibition assay. The antibodies were harvested from the serum of a rabbit which was immunised with recombinat PkSPATR. Results from the merozoite invasion inhibition assay revealed significant antibody invasion inhibitory activity in a concentration dependent manner (concentration range: 0.375 - 3.00 mg/ml) with inhibition rate ranging from 20% to 32%. Future studies, such as anti-PkSPATR antibodies inhibitory effect on sporozoite invasion of human liver cells, need to be carried out to assess the potential of PkSPATR as a knowlesi malaria vaccine candidate.


Subject(s)
Antibodies, Protozoan , Erythrocytes , Merozoites , Plasmodium knowlesi , Protozoan Proteins , Plasmodium knowlesi/immunology , Humans , Erythrocytes/parasitology , Rabbits , Animals , Antibodies, Protozoan/immunology , Protozoan Proteins/immunology , Merozoites/immunology , Thrombospondins/immunology , Malaria Vaccines/immunology
2.
Pan Afr Med J ; 47: 175, 2024.
Article in English | MEDLINE | ID: mdl-39036016

ABSTRACT

Introduction: in areas with intense perennial malaria transmission, limited data is available on the impact of environmental conditions especially rainfall on naturally acquired immunity against promising malaria vaccine candidates. For this reason, we have compared IgG antibody responses specific to Plasmodium spp. derived MSP3 and UB05 vaccine candidates, in plasma of children living in two areas of Cameroon differing in rainfall conditions. Methods: data about children less than 5 years old was collected during the years 2017 and 2018. Next malaria asymptomatic P. falciparum (Pf) infected children were selected following malaria test confirmation. MSP3 and UB05 specific IgG antibody responses were measured in participant´s plasma using enzyme-linked immunosorbent assay (ELISA). Results: interestingly, IgG antibody responses specific to UB05 were significantly higher (p<0.0001) in Pf-negative children when compared to their asymptomatic Pf-infected counterparts living in monomodal rainfall areas. In contrast, a significantly higher (p<0.0001) IgG response to MSP3 was observed instead in asymptomatic Pf-infected children in the same population. In addition, IgG responses specific to UB05 remained significantly higher in bimodal when compared to monomodal rainfall areas irrespective of children´s Pf infection status (p<0.0055 for Pf-positive and p<0.0001 for negative children). On the contrary, IgG antibody responses specific to MSP3 were significantly higher in bimodal relative to monomodal rainfall areas (P<0.0001) just for Pf-negative children. Conclusion: thus IgG antibody responses specific to UBO5 are a better correlate of naturally acquired immunity against malaria in Pf-negative Cameroonian children especially in monomodal rainfall areas.


Subject(s)
Antibodies, Protozoan , Antigens, Protozoan , Enzyme-Linked Immunosorbent Assay , Immunoglobulin G , Malaria, Falciparum , Plasmodium falciparum , Protozoan Proteins , Humans , Cameroon , Malaria, Falciparum/immunology , Malaria, Falciparum/epidemiology , Immunoglobulin G/blood , Child, Preschool , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Antigens, Protozoan/immunology , Antibodies, Protozoan/blood , Infant , Female , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Male , Rain , Recombinant Proteins/immunology
3.
Front Immunol ; 15: 1392043, 2024.
Article in English | MEDLINE | ID: mdl-38962015

ABSTRACT

In the Americas, P. vivax is the predominant causative species of malaria, a debilitating and economically significant disease. Due to the complexity of the malaria parasite life cycle, a vaccine formulation with multiple antigens expressed in various parasite stages may represent an effective approach. Based on this, we previously designed and constructed a chimeric recombinant protein, PvRMC-1, composed by PvCyRPA, PvCelTOS, and Pvs25 epitopes. This chimeric protein was strongly recognized by naturally acquired antibodies from exposed population in the Brazilian Amazon. However, there was no investigation about the induced immune response of PvRMC-1. Therefore, in this work, we evaluated the immunogenicity of this chimeric antigen formulated in three distinct adjuvants: Stimune, AddaVax or Aluminum hydroxide (Al(OH)3) in BALB/c mice. Our results suggested that the chimeric protein PvRMC-1 were capable to generate humoral and cellular responses across all three formulations. Antibodies recognized full-length PvRMC-1 and linear B-cell epitopes from PvCyRPA, PvCelTOS, and Pvs25 individually. Moreover, mice's splenocytes were activated, producing IFN-γ in response to PvCelTOS and PvCyRPA peptide epitopes, affirming T-cell epitopes in the antigen. While aluminum hydroxide showed notable cellular response, Stimune and Addavax induced a more comprehensive immune response, encompassing both cellular and humoral components. Thus, our findings indicate that PvRMC-1 would be a promising multistage vaccine candidate that could advance to further preclinical studies.


Subject(s)
Antibodies, Protozoan , Antigens, Protozoan , Malaria Vaccines , Malaria, Vivax , Mice, Inbred BALB C , Plasmodium vivax , Protozoan Proteins , Animals , Plasmodium vivax/immunology , Plasmodium vivax/genetics , Mice , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Malaria, Vivax/immunology , Malaria, Vivax/prevention & control , Antibodies, Protozoan/immunology , Malaria Vaccines/immunology , Female , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/genetics , Disease Models, Animal , Adjuvants, Immunologic , Immunogenicity, Vaccine , Antigens, Surface
4.
Gene ; 927: 148744, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38964492

ABSTRACT

Current understanding of genetic polymorphisms and natural selection in Plasmodium falciparum circumsporozoite (PfCSP), the leading malaria vaccine, is crucial for the development of next-generation vaccines, and such data is lacking in Africa. Blood samples were collected among Plasmodium-infected individuals living in four Cameroonian areas (Douala, Maroua, Mayo-Oulo, Pette). DNA samples were amplified using nested PCR protocols, sequenced, and BLASTed. Single nucleotide polymorphisms (SNPs) were analysed in each PfCSP region, and their impact on PfCSP function/structure was predicted in silico. The N-terminal region showed a limited polymorphism with four haplotypes, and three novel SNPs (N68Y, R87W, K93E) were found. Thirty-five haplotypes were identified in the central region, with several variants (e.g., NVNP and KANP). The C-terminal region was also highly diverse, with 25 haplotypes and eight novel SNPs (N290D, N308I, S312G, K317A, V344I, D356E, E357L, D359Y). Most polymorphic codon sites were mainly observed in the Th2R subregion in isolates from Douala and Pette. The codon site 321 was under episodic positive selection. One novel (E357L) and three known (K322I, G349D, D359Y) SNPs show an impact on function/structure. This study showed extensive genetic diversity with geographical patterns and evidence of the selection of Cameroonian PfCSP central and C-terminal regions.


Subject(s)
Haplotypes , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Polymorphism, Single Nucleotide , Protozoan Proteins , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Cameroon , Protozoan Proteins/genetics , Humans , Malaria, Falciparum/parasitology , Malaria, Falciparum/prevention & control , Malaria Vaccines/genetics , Malaria Vaccines/immunology
6.
Clin Obstet Gynecol ; 67(3): 620-632, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39061127

ABSTRACT

Malaria still presents a grave threat to the health of pregnancies worldwide with prevention currently stalling as traditional control and prevention strategies are limited by both insecticide and drug resistance. Furthermore, climate change is bringing malaria to locations where it was once eradicated and intensifying malaria in other areas. Even where malaria is not currently common, obstetricians will need to understand the pathogenesis of the disease, how it is transmitted, methods for prevention and treatment in pregnancy, and promising emerging strategies such as vaccines. A renewed global response is needed for this age-old disease in which pregnancy poses specific susceptibility.


Subject(s)
Climate Change , Malaria , Pregnancy Complications, Parasitic , Humans , Female , Pregnancy , Malaria/prevention & control , Pregnancy Complications, Parasitic/prevention & control , Antimalarials/therapeutic use , Malaria Vaccines/therapeutic use
7.
Cell Rep Med ; 5(7): 101654, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39019011

ABSTRACT

Plasmodium falciparum reticulocyte-binding protein homolog 5 (RH5) is a leading blood-stage malaria vaccine antigen target, currently in a phase 2b clinical trial as a full-length soluble protein/adjuvant vaccine candidate called RH5.1/Matrix-M. We identify that disordered regions of the full-length RH5 molecule induce non-growth inhibitory antibodies in human vaccinees and that a re-engineered and stabilized immunogen (including just the alpha-helical core of RH5) induces a qualitatively superior growth inhibitory antibody response in rats vaccinated with this protein formulated in Matrix-M adjuvant. In parallel, bioconjugation of this immunogen, termed "RH5.2," to hepatitis B surface antigen virus-like particles (VLPs) using the "plug-and-display" SpyTag-SpyCatcher platform technology also enables superior quantitative antibody immunogenicity over soluble protein/adjuvant in vaccinated mice and rats. These studies identify a blood-stage malaria vaccine candidate that may improve upon the current leading soluble protein vaccine candidate RH5.1/Matrix-M. The RH5.2-VLP/Matrix-M vaccine candidate is now under evaluation in phase 1a/b clinical trials.


Subject(s)
Antibodies, Protozoan , Malaria Vaccines , Plasmodium falciparum , Protozoan Proteins , Vaccines, Virus-Like Particle , Animals , Malaria Vaccines/immunology , Antibodies, Protozoan/immunology , Plasmodium falciparum/immunology , Vaccines, Virus-Like Particle/immunology , Humans , Mice , Protozoan Proteins/immunology , Rats , Malaria, Falciparum/prevention & control , Malaria, Falciparum/immunology , Antigens, Protozoan/immunology , Female , Carrier Proteins/immunology , Mice, Inbred BALB C
8.
Sci Transl Med ; 16(758): eadn6605, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39083589

ABSTRACT

Authorization of the Matrix-M (MM)-adjuvanted R21 vaccine by three countries and its subsequent endorsement by the World Health Organization for malaria prevention in children are a milestone in the fight against malaria. Yet, our understanding of the innate and adaptive immune responses elicited by this vaccine remains limited. Here, we compared three clinically relevant adjuvants [3M-052 + aluminum hydroxide (Alum) (3M), a TLR7/8 agonist formulated in Alum; GLA-LSQ, a TLR4 agonist formulated in liposomes with QS-21; and MM, the now-approved adjuvant for R21] for their capacity to induce durable immune responses to R21 in macaques. R21 adjuvanted with 3M on a 0, 8, and 23-week schedule elicited anti-circumsporozoite antibody responses comparable in magnitude to the R21/MM vaccine administered using a 0-4-8-week regimen and persisted up to 72 weeks with a half-life of 337 days. A booster dose at 72 weeks induced a recall response similar to the R21/MM vaccination. In contrast, R21/GLA-LSQ immunization induced a lower, short-lived response at the dose used. Consistent with the durable serum antibody responses, MM and 3M induced long-lived plasma cells in the bone marrow and other tissues, including the spleen. Furthermore, whereas 3M stimulated potent and persistent antiviral transcriptional and cytokine signatures after primary and booster immunizations, MM induced enhanced expression of interferon- and TH2-related signatures more highly after the booster vaccination. Collectively, these findings provide a resource on the immune responses of three clinically relevant adjuvants with R21 and highlight the promise of 3M as another adjuvant for malarial vaccines.


Subject(s)
Adjuvants, Immunologic , Malaria Vaccines , Animals , Malaria Vaccines/immunology , Adjuvants, Immunologic/pharmacology , Macaca mulatta , Adjuvants, Vaccine , Antibodies, Protozoan/immunology , Cytokines/metabolism
9.
Protein Sci ; 33(8): e5095, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38988315

ABSTRACT

The Duffy-binding protein (DBP) is a promising antigen for a malaria vaccine that would protect against clinical symptoms caused by Plasmodium vivax infection. Region II of DBP (DBP-II) contains the receptor-binding domain that engages host red blood cells, but DBP-II vaccines elicit many non-neutralizing antibodies that bind distal to the receptor-binding surface. Here, we engineered a truncated DBP-II immunogen that focuses the immune response to the receptor-binding surface. This immunogen contains the receptor-binding subdomain S1S2 and lacks the immunodominant subdomain S3. Structure-based computational design of S1S2 identified combinatorial amino acid changes that stabilized the isolated S1S2 without perturbing neutralizing epitopes. This immunogen elicited DBP-II-specific antibodies in immunized mice that were significantly enriched for blocking activity compared to the native DBP-II antigen. This generalizable design process successfully stabilized an integral core fragment of a protein and focused the immune response to desired epitopes to create a promising new antigen for malaria vaccine development.


Subject(s)
Antibodies, Protozoan , Antigens, Protozoan , Epitopes , Malaria Vaccines , Plasmodium vivax , Protozoan Proteins , Receptors, Cell Surface , Protozoan Proteins/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Antigens, Protozoan/immunology , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Plasmodium vivax/immunology , Animals , Malaria Vaccines/immunology , Malaria Vaccines/chemistry , Epitopes/immunology , Epitopes/chemistry , Mice , Antibodies, Protozoan/immunology , Receptors, Cell Surface/immunology , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Models, Molecular , Malaria, Vivax/immunology , Malaria, Vivax/prevention & control , Mice, Inbred BALB C
10.
PLoS One ; 19(7): e0306664, 2024.
Article in English | MEDLINE | ID: mdl-38968270

ABSTRACT

BACKGROUNDS: Malaria, a preventive and treatable disease, is still responsible for annual deaths reported in most tropical regions, principally in sub-Saharan Africa. Subunit recombinant transmission-blocking vaccines (TBVs) have been proposed as promising vaccines to succeed in malaria elimination and eradication. Here, a provisional study was designed to assess the immunogenicity and functional activity of alanyl aminopeptidase N (APN1) of Anopheles stephensi, as a TBV candidate, administered with MPL, CpG, and QS21 adjuvants in the murine model. METHODOLOGY/PRINCIPAL FINDINGS: The mouse groups were immunized with recombinant APN1 (rAPN1) alone or formulated with CpG, MPL, QS-21, or a combination of adjuvants (CMQ), and the elicited immune responses were evaluated after the third immunization. The standard membrane feeding assay (SMFA) measured the functional activity of antibodies against bacterial-expressed APN1 protein in adjuvanted vaccine groups on transmission of P. falciparum (NF54) to An. stephensi mosquitoes. Evaluation of mice vaccinated with rAPN1 formulated with distinct adjuvants manifested a significant increase in the high-avidity level of anti-APN1 IgG and IgG subclasses; however, rAPN1 induced the highest level of high-avidity anti-APN1 IgG1, IgG2a, and IgG2b antibodies in the immunized vaccine group 5 (APN1/CMQ). In addition, vaccine group 5 (receiving APN1/CMQ), had still the highest level of anti-APN1 IgG antibodies relative to other immunized groups after six months, on day 180. The SMFA data indicates a trend towards higher transmission-reducing activity in groups 2 and 5, which received the antigen formulated with CpG or a combination of three adjuvants. CONCLUSIONS/SIGNIFICANCE: The results have shown the capability of admixture to stimulate high-affinity and long-lasting antibodies against the target antigen to hinder Plasmodium parasite development in the mid-gut of An. stephensi. The attained results authenticated APN1/CMQ and APN1/CpG as a potent APN1-based TBV formulation which will be helpful in designing a vaccine in the future.


Subject(s)
Adjuvants, Immunologic , Anopheles , CD13 Antigens , Malaria Vaccines , Saponins , Animals , Anopheles/parasitology , Anopheles/immunology , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Mice , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Saponins/pharmacology , Saponins/administration & dosage , CD13 Antigens/immunology , CD13 Antigens/metabolism , Female , Plasmodium falciparum/immunology , Malaria/prevention & control , Malaria/transmission , Malaria/immunology , Malaria/parasitology , Oligodeoxyribonucleotides/pharmacology , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Mice, Inbred BALB C , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology
11.
Expert Rev Vaccines ; 23(1): 715-729, 2024.
Article in English | MEDLINE | ID: mdl-39042099

ABSTRACT

INTRODUCTION: The use of novel adjuvants in human vaccines continues to expand as their contribution to preventing disease in challenging populations and caused by complex pathogens is increasingly understood. AS01 is a family of liposome-based vaccine Adjuvant Systems containing two immunostimulants: 3-O-desacyl-4'-monophosphoryl lipid A and the saponin QS-21. AS01-containing vaccines have been approved and administered to millions of individuals worldwide. AREAS COVERED: Here, we report advances in our understanding of the mode of action of AS01 that contributed to the development of efficacious vaccines preventing disease due to malaria, herpes zoster, and respiratory syncytial virus. AS01 induces early innate immune activation that induces T cell-mediated and antibody-mediated responses with optimized functional characteristics and induction of immune memory. AS01-containing vaccines appear relatively impervious to baseline immune status translating into high efficacy across populations. Currently licensed AS01-containing vaccines have shown acceptable safety profiles in clinical trials and post-marketing settings. EXPERT OPINION: Initial expectations that adjuvantation with AS01 could support effective vaccine responses and contribute to disease control have been realized. Investigation of the utility of AS01 in vaccines to prevent other challenging diseases, such as tuberculosis, is ongoing, together with efforts to fully define its mechanisms of action in different vaccine settings.


Adjuvants are added to vaccines to increase the immune response produced after vaccination. Adjuvant Systems contain two or more molecules that stimulate the immune system. AS01 is an Adjuvant System that contains two components, MPL and QS-21, that stimulate the immune system. AS01 is included in three approved vaccines: a malaria vaccine for children, a herpes zoster vaccine for older adults, and a respiratory syncytial virus vaccine also for older adults. Vaccines containing AS01 have been extensively evaluated in clinical trials and administered to millions of individuals during market use. These vaccines are effective in preventing disease and have acceptable safety in different age groups. Experiments have been done to investigate how AS01 works in vaccines to produce an efficient immune response that helps to protect against the disease being targeted. A key effect of AS01 is to encourage specific immune cells to produce chemicals that stimulate the immune system. We now know that this effect is due to co-operation between MPL and QS-21. Experiments have shown that AS01 induces a sophisticated immune 'gene signature' in blood within 24 h after vaccination, and people who developed this 'gene signature' had a stronger response to vaccination. AS01 seems to be able to stimulate the immune system of most people ­ even if they are older or have a weakened immune system. This means that AS01 could be included in other vaccines against other challenging diseases, such as tuberculosis, or could be used in the treatment of some disease, such as chronic hepatitis B.


Subject(s)
Adjuvants, Immunologic , Adjuvants, Vaccine , Saponins , Humans , Saponins/immunology , Saponins/pharmacology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Lipid A/analogs & derivatives , Lipid A/immunology , Lipid A/pharmacology , Animals , Immunity, Innate/drug effects , Respiratory Syncytial Virus Vaccines/immunology , Liposomes , Malaria/prevention & control , Malaria/immunology , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Drug Combinations
12.
EBioMedicine ; 106: 105227, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39018754

ABSTRACT

BACKGROUND: A highly effective vaccine for malaria remains an elusive target, at least in part due to the under-appreciated natural parasite variation. This study aimed to investigate genetic and structural variation, and immune selection of leading malaria vaccine candidates across the Plasmodium falciparum's life cycle. METHODS: We analysed 325 P. falciparum whole genome sequences from Zambia, in addition to 791 genomes from five other African countries available in the MalariaGEN Pf3k Database. Ten vaccine antigens spanning three life-history stages were examined for genetic and structural variations, using population genetics measures, haplotype network analysis, and 3D structure selection analysis. FINDINGS: Among the ten antigens analysed, only three in the transmission-blocking vaccine category display P. falciparum 3D7 as the dominant haplotype. The antigens AMA1, CSP, MSP119 and CelTOS, are much more diverse than the other antigens, and their epitope regions are under moderate to strong balancing selection. In contrast, Rh5, a blood stage antigen, displays low diversity yet slightly stronger immune selection in the merozoite-blocking epitope region. Except for CelTOS, the transmission-blocking antigens Pfs25, Pfs48/45, Pfs230, Pfs47, and Pfs28 exhibit minimal diversity and no immune selection in epitopes that induce strain-transcending antibodies, suggesting potential effectiveness of 3D7-based vaccines in blocking transmission. INTERPRETATION: These findings offer valuable insights into the selection of optimal vaccine candidates against P. falciparum. Based on our results, we recommend prioritising conserved merozoite antigens and transmission-blocking antigens. Combining these antigens in multi-stage approaches may be particularly promising for malaria vaccine development initiatives. FUNDING: Purdue Department of Biological Sciences; Puskas Memorial Fellowship; National Institute of Allergy and Infectious Diseases (U19AI089680).


Subject(s)
Antigens, Protozoan , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Plasmodium falciparum/immunology , Plasmodium falciparum/genetics , Malaria Vaccines/immunology , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Malaria, Falciparum/parasitology , Malaria, Falciparum/immunology , Humans , Genetic Variation , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Haplotypes , Epitopes/immunology , Epitopes/genetics
13.
PLoS One ; 19(7): e0302243, 2024.
Article in English | MEDLINE | ID: mdl-39046960

ABSTRACT

The sequestration of Plasmodium falciparum-infected erythrocytes to the host endothelium is central to the pathogenesis of malaria. The sequestration is mediated by the parasite´s diverse Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) variants, which bind select human receptors on the endothelium. Severe malaria is associated with PfEMP1 binding human endothelial protein C receptor (EPCR) via their CIDRα1 domains. Antibodies binding and inhibiting across the sequence diverse CIDRα1 domains are likely important in acquired immunity against severe malaria. In this study, we explored if immunization with AP205 bacteriophage capsid-virus-like particles (cVLPs) presenting a mosaic of diverse CIDRα1 protein variants would stimulate broadly reactive and inhibitory antibody responses in mice. Three different mosaic cVLP vaccines each composed of five CIDRα1 protein variants with varying degrees of sequence conservation of residues at and near the EPCR binding site, were tested. All mosaic cVLP vaccines induced functional antibodies comparable to those induced by matched cocktails of cVLPs decorated with the single CIDRα1 variant. No broadly reactive responses were observed. However, the vaccines did induce some cross-reactivity and inhibition within the CIDRα1 subclasses included in the vaccines, demonstrating potential use of the cVLP vaccine platform for the design of multivalent vaccines.


Subject(s)
Endothelial Protein C Receptor , Protozoan Proteins , Vaccines, Virus-Like Particle , Animals , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Mice , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/administration & dosage , Humans , Endothelial Protein C Receptor/immunology , Endothelial Protein C Receptor/metabolism , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Plasmodium falciparum/immunology , Antibodies, Protozoan/immunology , Female , Protein Domains , Protein Binding , Mice, Inbred BALB C , Receptors, Cell Surface/immunology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology
14.
PLoS Comput Biol ; 20(6): e1012131, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38848436

ABSTRACT

Immunization through repeated direct venous inoculation of Plasmodium falciparum (Pf) sporozoites (PfSPZ) under chloroquine chemoprophylaxis, using the PfSPZ Chemoprophylaxis Vaccine (PfSPZ-CVac), induces high-level protection against controlled human malaria infection (CHMI). Humoral and cellular immunity contribute to vaccine efficacy but only limited information about the implicated Pf-specific antigens is available. Here, we examined Pf-specific antibody profiles, measured by protein arrays representing the full Pf proteome, of 40 placebo- and PfSPZ-immunized malaria-naïve volunteers from an earlier published PfSPZ-CVac dose-escalation trial. For this purpose, we both utilized and adapted supervised machine learning methods to identify predictive antibody profiles at two different time points: after immunization and before CHMI. We developed an adapted multitask support vector machine (SVM) approach and compared it to standard methods, i.e. single-task SVM, regularized logistic regression and random forests. Our results show, that the multitask SVM approach improved the classification performance to discriminate the protection status based on the underlying antibody-profiles while combining time- and dose-dependent data in the prediction model. Additionally, we developed the new fEature diStance exPlainabilitY (ESPY) method to quantify the impact of single antigens on the non-linear multitask SVM model and make it more interpretable. In conclusion, our multitask SVM model outperforms the studied standard approaches in regard of classification performance. Moreover, with our new explanation method ESPY, we were able to interpret the impact of Pf-specific antigen antibody responses that predict sterile protective immunity against CHMI after immunization. The identified Pf-specific antigens may contribute to a better understanding of immunity against human malaria and may foster vaccine development.


Subject(s)
Antibodies, Protozoan , Machine Learning , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Malaria Vaccines/immunology , Humans , Plasmodium falciparum/immunology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Antibodies, Protozoan/immunology , Antibodies, Protozoan/blood , Vaccine Efficacy , Support Vector Machine , Computational Biology/methods
15.
Expert Rev Vaccines ; 23(1): 645-654, 2024.
Article in English | MEDLINE | ID: mdl-38888098

ABSTRACT

INTRODUCTION: Malaria continues to remain a major global health problem with nearly a quarter of a billion clinical cases and more than 600,000 deaths in 2022. There has been significant progress toward vaccine development, however, poor efficacy of approved vaccines requiring multiple immunizing doses emphasizes the need for continued efforts toward improved vaccines. Progress to date, nonetheless, has provided impetus for malaria elimination. AREAS COVERED: In this review we will focus on diverse immune mechanisms targeting gametocytes in the human host and gametocyte-mediated malaria transmission via the mosquito vector. EXPERT OPINION: To march toward the goal of malaria elimination it will be critical to target the process of malaria transmission by mosquitoes, mediated exclusively by the sexual stages, i.e. male, and female gametocytes, ingested from infected vertebrate host. Studies over several decades have established antigens in the parasite sexual stages developing in the mosquito midgut as attractive targets for the development of transmission blocking vaccines (TBVs). Immune clearance of gametocytes in the vertebrate host can synergize with TBVs and directly aid in maintaining effective transmission reducing immune potential.


Subject(s)
Malaria Vaccines , Malaria , Mosquito Vectors , Vaccine Development , Humans , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Animals , Malaria/prevention & control , Malaria/transmission , Malaria/immunology , Malaria/parasitology , Mosquito Vectors/parasitology , Mosquito Vectors/immunology , Plasmodium/immunology
17.
Front Immunol ; 15: 1350560, 2024.
Article in English | MEDLINE | ID: mdl-38863702

ABSTRACT

Background: Despite decades of effort, Plasmodium falciparum malaria remains a leading killer of children. The absence of a highly effective vaccine and the emergence of parasites resistant to both diagnosis as well as treatment hamper effective public health interventions. Methods and results: To discover new vaccine candidates, we used our whole proteome differential screening method and identified PfGBP130 as a parasite protein uniquely recognized by antibodies from children who had developed resistance to P. falciparum infection but not from those who remained susceptible. We formulated PfGBP130 as lipid encapsulated mRNA, DNA plasmid, and recombinant protein-based immunogens and evaluated the efficacy of murine polyclonal anti-PfGBP130 antisera to inhibit parasite growth in vitro. Immunization of mice with PfGBP130-A (aa 111-374), the region identified in our differential screen, formulated as a DNA plasmid or lipid encapsulated mRNA, but not as a recombinant protein, induced antibodies that inhibited RBC invasion in vitro. mRNA encoding the full ectodomain of PfGBP130 (aa 89-824) also generated parasite growth-inhibitory antibodies. Conclusion: We are currently advancing PfGBP130-A formulated as a lipid-encapsulated mRNA for efficacy evaluation in non-human primates.


Subject(s)
Antibodies, Protozoan , Erythrocytes , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Protozoan Proteins , Animals , Plasmodium falciparum/immunology , Antibodies, Protozoan/immunology , Mice , Erythrocytes/parasitology , Erythrocytes/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/parasitology , Humans , Malaria Vaccines/immunology , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Antigens, Protozoan/immunology , Immunization , Female
18.
Medicine (Baltimore) ; 103(24): e38565, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38875411

ABSTRACT

Malaria remains an endemic public health concern in Africa, significantly contributing to morbidity and mortality rates. The inadequacies of traditional prevention measures, like integrated vector management and antimalarial drugs, have spurred efforts to strengthen the development and deployment of malaria vaccines. In addition to existing interventions like insecticide-treated bed nets and artemisinin-based combination therapies, malaria vaccine introduction and implementation in Africa could drastically reduce the disease burden and hasten steps toward malaria elimination. The malaria vaccine rollout is imminent as optimistic results from final clinical trials are anticipated. Thus, determining potential hurdles to malaria vaccine delivery and uptake in malaria-endemic regions of sub-Saharan Africa will enhance decisions and policymakers' preparedness to facilitate efficient and equitable vaccine delivery. A multisectoral approach is recommended to increase funding and resources, active community engagement and participation, and the involvement of healthcare providers.


Subject(s)
Malaria Vaccines , Malaria , Humans , Malaria Vaccines/administration & dosage , Malaria Vaccines/therapeutic use , Malaria/prevention & control , Africa/epidemiology , Vaccination , Africa South of the Sahara/epidemiology
19.
PLoS Negl Trop Dis ; 18(6): e0012231, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38865344

ABSTRACT

BACKGROUND: Malaria transmission-blocking vaccines (TBVs) aim to inhibit malaria parasite development in mosquitoes and prevent further transmission to the human host. The putative-secreted ookinete protein 25 (PSOP25), highly conserved in Plasmodium spp., is a promising TBV target. Here, we investigated PvPSOP25 from P. vivax as a TBV candidate using transgenic murine parasite P. berghei and clinical P. vivax isolates. METHODS AND FINDINGS: A transgenic P. berghei line expressing PvPSOP25 (TrPvPSOP25Pb) was generated. Full-length PvPSOP25 was expressed in the yeast Pichia pastoris and used to immunize mice to obtain anti-rPvPSOP25 sera. The transmission-blocking activity of the anti-rPvPSOP25 sera was evaluated through in vitro assays and mosquito-feeding experiments. The antisera generated by immunization with rPvPSOP25 specifically recognized the native PvPSOP25 antigen expressed in TrPvPSOP25Pb ookinetes. In vitro assays showed that the immune sera significantly inhibited exflagellation and ookinete formation of the TrPvPSOP25Pb parasite. Mosquitoes feeding on mice infected with the transgenic parasite and passively transferred with the anti-rPvPSOP25 sera showed a 70.7% reduction in oocyst density compared to the control group. In a direct membrane feeding assay conducted with five clinical P. vivax isolates, the mouse anti-rPvPSOP25 antibodies significantly reduced the oocyst density while showing a negligible influence on mosquito infection prevalence. CONCLUSIONS: This study supported the feasibility of transgenic murine malaria parasites expressing P. vivax antigens as a useful tool for evaluating P. vivax TBV candidates. Meanwhile, the moderate transmission-reducing activity of the generated anti-rPvPSOP25 sera necessitates further research to optimize its efficacy.


Subject(s)
Malaria Vaccines , Malaria, Vivax , Plasmodium berghei , Plasmodium vivax , Protozoan Proteins , Animals , Mice , Plasmodium vivax/genetics , Plasmodium vivax/immunology , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Humans , Malaria, Vivax/transmission , Malaria, Vivax/parasitology , Malaria, Vivax/prevention & control , Malaria, Vivax/immunology , Female , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Malaria/transmission , Malaria/prevention & control , Malaria/parasitology , Malaria/immunology , Mice, Inbred BALB C
20.
Parasites Hosts Dis ; 62(2): 193-204, 2024 May.
Article in English | MEDLINE | ID: mdl-38835260

ABSTRACT

Malaria is a global disease affecting a large portion of the world's population. Although vaccines have recently become available, their efficacies are suboptimal. We generated virus-like particles (VLPs) that expressed either apical membrane antigen 1 (AMA1) or microneme-associated antigen (MIC) of Plasmodium berghei and compared their efficacy in BALB/c mice. We found that immune sera acquired from AMA1 VLP- or MIC VLP-immunized mice specifically interacted with the antigen of choice and the whole P. berghei lysate antigen, indicating that the antibodies were highly parasite-specific. Both VLP vaccines significantly enhanced germinal center B cell frequencies in the inguinal lymph nodes of mice compared with the control, but only the mice that received MIC VLPs showed significantly enhanced CD4+ T cell responses in the blood following P. berghei challenge infection. AMA1 and MIC VLPs significantly suppressed TNF-α and interleukin-10 production but had a negligible effect on interferon-γ. Both VLPs prevented excessive parasitemia buildup in immunized mice, although parasite burden reduction induced by MIC VLPs was slightly more effective than that induced by AMA1. Both VLPs were equally effective at preventing body weight loss. Our findings demonstrated that the MIC VLP was an effective inducer of protection against murine experimental malaria and should be the focus of further development.


Subject(s)
Antigens, Protozoan , Malaria Vaccines , Membrane Proteins , Plasmodium berghei , Protozoan Proteins , Vaccines, Virus-Like Particle , Animals , Female , Mice , Antibodies, Protozoan/immunology , Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , Malaria/prevention & control , Malaria/immunology , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Membrane Proteins/immunology , Mice, Inbred BALB C , Parasitemia/immunology , Parasitemia/prevention & control , Plasmodium berghei/immunology , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL