Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.519
Filtrar
1.
Front Cell Infect Microbiol ; 14: 1451063, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39258252

RESUMO

Background: Transmission-blocking vaccines (TBVs) can effectively prevent the community's spread of malaria by targeting the antigens of mosquito sexual stage parasites. At present, only a few candidate antigens have demonstrated transmission-blocking activity (TBA) potential in P. vivax. Quiescin-sulfhydryl oxidase (QSOX) is a sexual stage protein in the rodent malaria parasite Plasmodium berghei and is associated with a critical role in protein folding by introducing disulfides into unfolded reduced proteins. Here, we reported the immunogenicity and transmission-blocking potency of the PvQSOX in P. vivax. Methods and findings: The full-length recombinant PvQSOX protein (rPvQSOX) was expressed in the Escherichia coli expression system. The anti-rPvQSOX antibodies were generated following immunization with the rPvQSOX in rabbits. A parasite integration of the pvqsox gene into the P. berghei pbqsox gene knockout genome was developed to express full-length PvQSOX protein in P. berghei (Pv-Tr-PbQSOX). In western blot, the anti-rPvQSOX antibodies recognized the native PvQSOX protein expressed in transgenic P. berghei gametocyte and ookinete. In indirect immunofluorescence assays, the fluorescence signal was detected in the sexual stages, including gametocyte, gamete, zygote, and ookinete. Anti-rPvQSOX IgGs obviously inhibited the ookinetes and oocysts development both in vivo and in vitro using transgenic parasites. Direct membrane feeding assays of anti-rPvQSOX antibodies were conducted using four field P. vivax isolates (named isolates #1-4) in Thailand. Oocyst density in mosquitoes was significantly reduced by 32.00, 85.96, 43.52, and 66.03% with rabbit anti-rPvQSOX antibodies, respectively. The anti-rPvQSOX antibodies also showed a modest reduction of infection prevalence by 15, 15, 20, and 22.22%, respectively, as compared to the control, while the effect was insignificant. The variation in the DMFA results may be unrelated to the genetic polymorphisms. Compared to the P.vivax Salvador (Sal) I strain sequences, the pvqsox in isolate #1 showed no amino acid substitution, whereas isolates #2, #3, and #4 all had the M361I substitution. Conclusions: Our results suggest that PvQSOX could serve as a potential P. vivax TBVs candidate, which warrants further evaluation and optimization.


Assuntos
Anticorpos Antiprotozoários , Vacinas Antimaláricas , Malária Vivax , Plasmodium berghei , Plasmodium vivax , Proteínas Recombinantes , Plasmodium vivax/imunologia , Plasmodium vivax/genética , Plasmodium vivax/enzimologia , Animais , Coelhos , Vacinas Antimaláricas/imunologia , Anticorpos Antiprotozoários/imunologia , Malária Vivax/prevenção & controle , Malária Vivax/transmissão , Malária Vivax/imunologia , Plasmodium berghei/imunologia , Plasmodium berghei/genética , Plasmodium berghei/enzimologia , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Camundongos , Escherichia coli/genética , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/genética , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Feminino , Humanos , Imunogenicidade da Vacina , Camundongos Endogâmicos BALB C
2.
Nat Commun ; 15(1): 7487, 2024 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-39209843

RESUMO

CelTOS is a malaria vaccine antigen that is conserved in Plasmodium and other apicomplexan parasites and plays a role in cell-traversal. The structural basis and mechanisms of CelTOS-induced protective immunity to parasites are unknown. Here, CelTOS-specific monoclonal antibodies (mAbs) 7g7 and 4h12 demonstrated multistage activity, protecting against liver infection and preventing parasite transmission to mosquitoes. Both mAbs demonstrated cross-species activity with sterile protection against in vivo challenge with transgenic parasites containing either P. falciparum or P. vivax CelTOS, and with transmission reducing activity against P. falciparum. The mAbs prevented CelTOS-mediated pore formation providing insight into the protective mechanisms. X-ray crystallography and mutant-library epitope mapping revealed two distinct broadly conserved neutralizing epitopes. 7g7 bound to a parallel dimer of CelTOS, while 4h12 bound to a novel antiparallel dimer architecture. These findings inform the design of antibody therapies and vaccines and raise the prospect of a single intervention to simultaneously combat P. falciparum and P. vivax malaria.


Assuntos
Anticorpos Monoclonais , Anticorpos Antiprotozoários , Vacinas Antimaláricas , Plasmodium falciparum , Plasmodium vivax , Anticorpos Monoclonais/imunologia , Animais , Plasmodium falciparum/imunologia , Plasmodium vivax/imunologia , Vacinas Antimaláricas/imunologia , Anticorpos Antiprotozoários/imunologia , Camundongos , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Malária Falciparum/parasitologia , Cristalografia por Raios X , Epitopos/imunologia , Malária Vivax/prevenção & controle , Malária Vivax/imunologia , Malária Vivax/parasitologia , Antígenos de Protozoários/imunologia , Humanos , Feminino , Mapeamento de Epitopos , Malária/imunologia , Malária/prevenção & controle , Malária/parasitologia , Camundongos Endogâmicos BALB C , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/química
3.
EMBO Mol Med ; 16(9): 2060-2079, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39103697

RESUMO

Malaria vaccination approaches using live Plasmodium parasites are currently explored, with either attenuated mosquito-derived sporozoites or attenuated blood-stage parasites. Both approaches would profit from the availability of attenuated and avirulent parasites with a reduced blood-stage multiplication rate. Here we screened gene-deletion mutants of the rodent parasite P. berghei and the human parasite P. falciparum for slow growth. Furthermore, we tested the P. berghei mutants for avirulence and resolving blood-stage infections, while preserving sporozoite formation and liver infection. Targeting 51 genes yielded 18 P. berghei gene-deletion mutants with several mutants causing mild infections. Infections with the two most attenuated mutants either by blood stages or by sporozoites were cleared by the immune response. Immunization of mice led to protection from disease after challenge with wild-type sporozoites. Two of six generated P. falciparum gene-deletion mutants showed a slow growth rate. Slow-growing, avirulent P. falciparum mutants will constitute valuable tools to inform on the induction of immune responses and will aid in developing new as well as safeguarding existing attenuated parasite vaccines.


Assuntos
Vacinas Antimaláricas , Malária , Plasmodium berghei , Plasmodium falciparum , Esporozoítos , Vacinas Atenuadas , Animais , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/administração & dosagem , Esporozoítos/imunologia , Plasmodium berghei/imunologia , Plasmodium berghei/genética , Plasmodium falciparum/imunologia , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Malária/prevenção & controle , Malária/parasitologia , Malária/imunologia , Camundongos , Vacinação/métodos , Humanos , Deleção de Genes , Feminino
4.
Nat Immunol ; 25(9): 1530-1545, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39198635

RESUMO

Despite various public health strategies, malaria caused by Plasmodium falciparum parasites remains a major global health challenge that requires development of new interventions. Extended half-life human monoclonal antibodies targeting the P. falciparum circumsporozoite protein on sporozoites, the infective form of malaria parasites, prevent malaria in rodents and humans and have been advanced into clinical development. The protective epitopes on the circumsporozoite protein targeted by monoclonal antibodies have been defined. Cryogenic electron and multiphoton microscopy have enabled mechanistic structural and functional investigations of how antibodies bind to the circumsporozoite protein and neutralize sporozoites. Moreover, innovations in bioinformatics and antibody engineering have facilitated enhancement of antibody potency and durability. Here, we summarize the latest scientific advances in understanding how monoclonal antibodies to the circumsporozoite protein prevent malaria and highlight existing clinical data and future plans for how this emerging intervention can be used alone or alongside existing antimalarial interventions to control malaria across at-risk populations.


Assuntos
Anticorpos Monoclonais , Anticorpos Antiprotozoários , Malária Falciparum , Plasmodium falciparum , Proteínas de Protozoários , Proteínas de Protozoários/imunologia , Humanos , Anticorpos Monoclonais/imunologia , Plasmodium falciparum/imunologia , Animais , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Malária Falciparum/parasitologia , Anticorpos Antiprotozoários/imunologia , Esporozoítos/imunologia , Vacinas Antimaláricas/imunologia , Epitopos/imunologia
5.
Nat Commun ; 15(1): 7206, 2024 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-39174515

RESUMO

Apical membrane antigen-1 (AMA1) is a conserved malarial vaccine candidate essential for the formation of tight junctions with the rhoptry neck protein (RON) complex, enabling Plasmodium parasites to invade human erythrocytes, hepatocytes, and mosquito salivary glands. Despite its critical role, extensive surface polymorphisms in AMA1 have led to strain-specific protection, limiting the success of AMA1-based interventions beyond initial clinical trials. Here, we identify an i-body, a humanised single-domain antibody-like molecule that recognises a conserved pan-species conformational epitope in AMA1 with low nanomolar affinity and inhibits the binding of the RON2 ligand to AMA1. Structural characterisation indicates that the WD34 i-body epitope spans the centre of the conserved hydrophobic cleft in AMA1, where interacting residues are highly conserved among all Plasmodium species. Furthermore, we show that WD34 inhibits merozoite invasion of erythrocytes by multiple Plasmodium species and hepatocyte invasion by P. falciparum sporozoites. Despite a short half-life in mouse serum, we demonstrate that WD34 transiently suppressed P. berghei infections in female BALB/c mice. Our work describes the first pan-species AMA1 biologic with inhibitory activity against multiple life-cycle stages of Plasmodium. With improved pharmacokinetic characteristics, WD34 could be a potential immunotherapy against multiple species of Plasmodium.


Assuntos
Antígenos de Protozoários , Eritrócitos , Fígado , Proteínas de Membrana , Camundongos Endogâmicos BALB C , Proteínas de Protozoários , Animais , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/genética , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/metabolismo , Feminino , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Humanos , Eritrócitos/parasitologia , Eritrócitos/imunologia , Fígado/parasitologia , Fígado/imunologia , Fígado/metabolismo , Vacinas Antimaláricas/imunologia , Malária/imunologia , Malária/parasitologia , Malária/prevenção & controle , Reações Cruzadas/imunologia , Plasmodium falciparum/imunologia , Plasmodium berghei/imunologia , Epitopos/imunologia , Hepatócitos/parasitologia , Hepatócitos/imunologia , Hepatócitos/metabolismo , Plasmodium/imunologia , Merozoítos/imunologia , Merozoítos/metabolismo
6.
Diagn Microbiol Infect Dis ; 110(3): 116480, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39163788

RESUMO

The apical membrane antigen-1 (AMA-1) is a crucial target for malaria management and prevention strategies. While the immunogenicity of AMA-1 has been extensively studied for Plasmodium falciparum and Plasmodium vivax, there is a notable scarcity of information for Plasmodium malariae. In this study, recombinant PmAMA-1 was expressed in Escherichia coli, and its integrity was confirmed via western blotting and indirect immunofluorescence assays. Immunization of BALB/c mice with rPmAMA-1 emulsified in Freund's adjuvant resulted in significantly elevated specific IgG antibodies, predominantly IgG1. The immune response exhibited Th1, Th2, and Th17 phenotypes, with a notable Th1 bias. Antisera from immunized mice effectively recognized native PmAMA-1 on P. malariae. These results suggest that PmAMA-1 is a promising target for both vaccine development and diagnostic applications for P. malariae infections, offering dual preventive and diagnostic benefits in malaria control.


Assuntos
Anticorpos Antiprotozoários , Antígenos de Protozoários , Malária , Proteínas de Membrana , Plasmodium malariae , Proteínas de Protozoários , Animais , Feminino , Camundongos , Anticorpos Antiprotozoários/sangue , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/genética , Escherichia coli/genética , Imunoglobulina G/sangue , Malária/diagnóstico , Malária/prevenção & controle , Malária/imunologia , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/administração & dosagem , Proteínas de Membrana/imunologia , Proteínas de Membrana/genética , Camundongos Endogâmicos BALB C , Plasmodium malariae/imunologia , Plasmodium malariae/genética , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/genética
7.
Trop Biomed ; 41(2): 190-195, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-39154272

RESUMO

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.


Assuntos
Anticorpos Antiprotozoários , Eritrócitos , Merozoítos , Plasmodium knowlesi , Proteínas de Protozoários , Plasmodium knowlesi/imunologia , Humanos , Eritrócitos/parasitologia , Coelhos , Animais , Anticorpos Antiprotozoários/imunologia , Proteínas de Protozoários/imunologia , Merozoítos/imunologia , Trombospondinas/imunologia , Vacinas Antimaláricas/imunologia
8.
Vaccine ; 42(21): 126140, 2024 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-39033079

RESUMO

Transmission-blocking vaccines interrupting malaria transmission within mosquitoes represent an ideal public health tool to eliminate malaria at the population level. Plasmodium falciparum and P. vivax account for more than 90% of the global malaria burden, co-endemic in many regions of the world. P25 and P48/45 are two leading candidates for both species and have shown promising transmission-blocking activity in preclinical and clinical studies. However, neither of these target antigens as individual vaccines has induced complete transmission inhibition in mosquitoes. In this study, we assessed immunogenicity of combination vaccines based on P25 and P48/45 using a DNA vaccine platform to broaden vaccine specificity against P. falciparum and P. vivax. Individual DNA vaccines encoding Pvs25, Pfs25, Pvs48/45 and Pfs48/45, as well as various combinations including (Pvs25 + Pvs48/45), (Pfs25 + Pfs48/45), (Pvs25 + Pfs25), and (Pvs48/45 + Pfs48/45), were evaluated in mice using in vivo electroporation. Potent antibody responses were induced in mice immunized with individual and combination DNA vaccines, and specific antibody responses were not compromised when combinations of DNA vaccines were evaluated against individual DNA vaccines. The anti-Pvs25 IgG from individual and combination groups revealed concentration-dependent transmission-reducing activity (TRA) in direct membrane feeding assays (DMFA) using blood from P. vivax-infected donors in Brazil and independently in ex vivo MFA using Pvs25-transgenic P. berghei. Similarly, anti-Pfs25 and anti-Pfs48/45 IgGs from mice immunized with Pfs25 and Pfs48/45 DNA vaccines individually and in various combinations revealed antibody dose-dependent TRA in standard membrane feeding assays (SMFA) using culture-derived P. falciparum gametocytes. However, antibodies induced by immunization with Pvs48/45 DNA vaccines were ineffective in DMFA and require further vaccine construct optimization, considering the possibility of induction of both transmission-blocking and transmission-enhancing antibodies revealed by competition ELISA. These studies provide a rationale for combining multiple antigens to simultaneously target transmission of malaria caused by P. falciparum and P. vivax.


Assuntos
Anticorpos Antiprotozoários , Vacinas Antimaláricas , Malária Falciparum , Malária Vivax , Plasmodium falciparum , Plasmodium vivax , Vacinas de DNA , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/administração & dosagem , Animais , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Malária Falciparum/imunologia , Plasmodium falciparum/imunologia , Plasmodium falciparum/genética , Plasmodium vivax/imunologia , Plasmodium vivax/genética , Malária Vivax/prevenção & controle , Malária Vivax/transmissão , Malária Vivax/imunologia , Camundongos , Vacinas de DNA/imunologia , Vacinas de DNA/administração & dosagem , Anticorpos Antiprotozoários/imunologia , Anticorpos Antiprotozoários/sangue , Feminino , Vacinas Combinadas/imunologia , Vacinas Combinadas/administração & dosagem , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/genética , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/genética , Camundongos Endogâmicos BALB C , Humanos
9.
Cell ; 187(18): 4964-4980.e21, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39059380

RESUMO

The highly conserved and essential Plasmodium falciparum reticulocyte-binding protein homolog 5 (PfRH5) has emerged as the leading target for vaccines against the disease-causing blood stage of malaria. However, the features of the human vaccine-induced antibody response that confer highly potent inhibition of malaria parasite invasion into red blood cells are not well defined. Here, we characterize 236 human IgG monoclonal antibodies, derived from 15 donors, induced by the most advanced PfRH5 vaccine. We define the antigenic landscape of this molecule and establish that epitope specificity, antibody association rate, and intra-PfRH5 antibody interactions are key determinants of functional anti-parasitic potency. In addition, we identify a germline IgG gene combination that results in an exceptionally potent class of antibody and demonstrate its prophylactic potential to protect against P. falciparum parasite challenge in vivo. This comprehensive dataset provides a framework to guide rational design of next-generation vaccines and prophylactic antibodies to protect against blood-stage malaria.


Assuntos
Anticorpos Monoclonais , Anticorpos Antiprotozoários , Antígenos de Protozoários , Imunoglobulina G , Vacinas Antimaláricas , Malária Falciparum , Plasmodium falciparum , Proteínas de Protozoários , Animais , Humanos , Camundongos , Anticorpos Monoclonais/imunologia , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Proteínas de Transporte/imunologia , Epitopos/imunologia , Eritrócitos/parasitologia , Eritrócitos/imunologia , Imunoglobulina G/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Malária Falciparum/parasitologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia
10.
Front Immunol ; 15: 1392043, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38962015

RESUMO

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.


Assuntos
Anticorpos Antiprotozoários , Antígenos de Protozoários , Vacinas Antimaláricas , Malária Vivax , Camundongos Endogâmicos BALB C , Plasmodium vivax , Proteínas de Protozoários , Animais , Plasmodium vivax/imunologia , Plasmodium vivax/genética , Camundongos , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/genética , Malária Vivax/imunologia , Malária Vivax/prevenção & controle , Anticorpos Antiprotozoários/imunologia , Vacinas Antimaláricas/imunologia , Feminino , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/genética , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito B/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/genética , Modelos Animais de Doenças , Adjuvantes Imunológicos , Imunogenicidade da Vacina , Antígenos de Superfície
11.
Sci Transl Med ; 16(758): eadn6605, 2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39083589

RESUMO

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.


Assuntos
Adjuvantes Imunológicos , Vacinas Antimaláricas , Animais , Vacinas Antimaláricas/imunologia , Adjuvantes Imunológicos/farmacologia , Macaca mulatta , Adjuvantes de Vacinas , Anticorpos Antiprotozoários/imunologia , Citocinas/metabolismo
12.
PLoS One ; 19(7): e0306664, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38968270

RESUMO

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.


Assuntos
Adjuvantes Imunológicos , Anopheles , Antígenos CD13 , Vacinas Antimaláricas , Saponinas , Animais , Anopheles/parasitologia , Anopheles/imunologia , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Camundongos , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/administração & dosagem , Saponinas/farmacologia , Saponinas/administração & dosagem , Antígenos CD13/imunologia , Antígenos CD13/metabolismo , Feminino , Plasmodium falciparum/imunologia , Malária/prevenção & controle , Malária/transmissão , Malária/imunologia , Malária/parasitologia , Oligodesoxirribonucleotídeos/farmacologia , Oligodesoxirribonucleotídeos/administração & dosagem , Oligodesoxirribonucleotídeos/imunologia , Camundongos Endogâmicos BALB C , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Malária Falciparum/imunologia , Malária Falciparum/parasitologia
13.
Expert Rev Vaccines ; 23(1): 715-729, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39042099

RESUMO

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.


Assuntos
Adjuvantes Imunológicos , Adjuvantes de Vacinas , Saponinas , Humanos , Saponinas/imunologia , Saponinas/farmacologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/farmacologia , Lipídeo A/análogos & derivados , Lipídeo A/imunologia , Lipídeo A/farmacologia , Animais , Imunidade Inata/efeitos dos fármacos , Vacinas contra Vírus Sincicial Respiratório/imunologia , Lipossomos , Malária/prevenção & controle , Malária/imunologia , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/administração & dosagem , Combinação de Medicamentos
14.
Cell Rep Med ; 5(7): 101654, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39019011

RESUMO

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.


Assuntos
Anticorpos Antiprotozoários , Vacinas Antimaláricas , Plasmodium falciparum , Proteínas de Protozoários , Vacinas de Partículas Semelhantes a Vírus , Animais , Vacinas Antimaláricas/imunologia , Anticorpos Antiprotozoários/imunologia , Plasmodium falciparum/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia , Humanos , Camundongos , Proteínas de Protozoários/imunologia , Ratos , Malária Falciparum/prevenção & controle , Malária Falciparum/imunologia , Antígenos de Protozoários/imunologia , Feminino , Proteínas de Transporte/imunologia , Camundongos Endogâmicos BALB C
15.
Pan Afr Med J ; 47: 175, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39036016

RESUMO

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.


Assuntos
Anticorpos Antiprotozoários , Antígenos de Protozoários , Ensaio de Imunoadsorção Enzimática , Imunoglobulina G , Malária Falciparum , Plasmodium falciparum , Proteínas de Protozoários , Humanos , Camarões , Malária Falciparum/imunologia , Malária Falciparum/epidemiologia , Imunoglobulina G/sangue , Pré-Escolar , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Antígenos de Protozoários/imunologia , Anticorpos Antiprotozoários/sangue , Lactente , Feminino , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/imunologia , Masculino , Chuva , Proteínas Recombinantes/imunologia
16.
Gene ; 927: 148744, 2024 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-38964492

RESUMO

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.


Assuntos
Haplótipos , Vacinas Antimaláricas , Malária Falciparum , Plasmodium falciparum , Polimorfismo de Nucleotídeo Único , Proteínas de Protozoários , Plasmodium falciparum/genética , Plasmodium falciparum/imunologia , Camarões , Proteínas de Protozoários/genética , Humanos , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/imunologia
17.
Protein Sci ; 33(8): e5095, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38988315

RESUMO

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.


Assuntos
Anticorpos Antiprotozoários , Antígenos de Protozoários , Epitopos , Vacinas Antimaláricas , Plasmodium vivax , Proteínas de Protozoários , Receptores de Superfície Celular , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Plasmodium vivax/imunologia , Animais , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/química , Epitopos/imunologia , Epitopos/química , Camundongos , Anticorpos Antiprotozoários/imunologia , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/química , Receptores de Superfície Celular/genética , Modelos Moleculares , Malária Vivax/imunologia , Malária Vivax/prevenção & controle , Camundongos Endogâmicos BALB C
18.
EBioMedicine ; 106: 105227, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39018754

RESUMO

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).


Assuntos
Antígenos de Protozoários , Vacinas Antimaláricas , Malária Falciparum , Plasmodium falciparum , Plasmodium falciparum/imunologia , Plasmodium falciparum/genética , Vacinas Antimaláricas/imunologia , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/genética , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Malária Falciparum/parasitologia , Malária Falciparum/imunologia , Humanos , Variação Genética , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/genética , Haplótipos , Epitopos/imunologia , Epitopos/genética
19.
PLoS One ; 19(7): e0302243, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39046960

RESUMO

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.


Assuntos
Receptor de Proteína C Endotelial , Proteínas de Protozoários , Vacinas de Partículas Semelhantes a Vírus , Animais , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/genética , Camundongos , Vacinas de Partículas Semelhantes a Vírus/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Humanos , Receptor de Proteína C Endotelial/imunologia , Receptor de Proteína C Endotelial/metabolismo , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/administração & dosagem , Plasmodium falciparum/imunologia , Anticorpos Antiprotozoários/imunologia , Feminino , Domínios Proteicos , Ligação Proteica , Camundongos Endogâmicos BALB C , Receptores de Superfície Celular/imunologia , Malária Falciparum/prevenção & controle , Malária Falciparum/imunologia , Malária Falciparum/parasitologia
20.
Cell ; 187(18): 4981-4995.e14, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39059381

RESUMO

Plasmodium falciparum reticulocyte-binding protein homolog 5 (RH5) is the most advanced blood-stage malaria vaccine candidate and is being evaluated for efficacy in endemic regions, emphasizing the need to study the underlying antibody response to RH5 during natural infection, which could augment or counteract responses to vaccination. Here, we found that RH5-reactive B cells were rare, and circulating immunoglobulin G (IgG) responses to RH5 were short-lived in malaria-exposed Malian individuals, despite repeated infections over multiple years. RH5-specific monoclonal antibodies isolated from eight malaria-exposed individuals mostly targeted non-neutralizing epitopes, in contrast to antibodies isolated from five RH5-vaccinated, malaria-naive UK individuals. However, MAD8-151 and MAD8-502, isolated from two malaria-exposed Malian individuals, were among the most potent neutralizers out of 186 antibodies from both cohorts and targeted the same epitopes as the most potent vaccine-induced antibodies. These results suggest that natural malaria infection may boost RH5-vaccine-induced responses and provide a clear strategy for the development of next-generation RH5 vaccines.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antiprotozoários , Antígenos de Protozoários , Vacinas Antimaláricas , Malária Falciparum , Plasmodium falciparum , Humanos , Anticorpos Neutralizantes/imunologia , Plasmodium falciparum/imunologia , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Malária Falciparum/parasitologia , Vacinas Antimaláricas/imunologia , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Imunoglobulina G/imunologia , Imunoglobulina G/sangue , Proteínas de Protozoários/imunologia , Anticorpos Monoclonais/imunologia , Adulto , Linfócitos B/imunologia , Epitopos/imunologia , Feminino , Mali , Proteínas de Transporte/imunologia , Masculino , Adolescente
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...