Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.482
Filter
1.
JCI Insight ; 9(9)2024 May 08.
Article in English | MEDLINE | ID: mdl-38716733

ABSTRACT

Vaccination of malaria-naive volunteers with a high dose of Plasmodium falciparum sporozoites chemoattenuated by chloroquine (CQ) (PfSPZ-CVac [CQ]) has previously demonstrated full protection against controlled human malaria infection (CHMI). However, lower doses of PfSPZ-CVac [CQ] resulted in incomplete protection. This provides the opportunity to understand the immune mechanisms needed for better vaccine-induced protection by comparing individuals who were protected with those not protected. Using mass cytometry, we characterized immune cell composition and responses of malaria-naive European volunteers who received either lower doses of PfSPZ-CVac [CQ], resulting in 50% protection irrespective of the dose, or a placebo vaccination, with everyone becoming infected following CHMI. Clusters of CD4+ and γδ T cells associated with protection were identified, consistent with their known role in malaria immunity. Additionally, EMRA CD8+ T cells and CD56+CD8+ T cell clusters were associated with protection. In a cohort from a malaria-endemic area in Gabon, these CD8+ T cell clusters were also associated with parasitemia control in individuals with lifelong exposure to malaria. Upon stimulation with P. falciparum-infected erythrocytes, CD4+, γδ, and EMRA CD8+ T cells produced IFN-γ and/or TNF, indicating their ability to mediate responses that eliminate malaria parasites.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Sporozoites , Humans , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , CD8-Positive T-Lymphocytes/immunology , Adult , Sporozoites/immunology , Male , CD4-Positive T-Lymphocytes/immunology , Chloroquine/therapeutic use , Chloroquine/pharmacology , Female , Young Adult , Gabon , Vaccination/methods , Antimalarials/therapeutic use , Antimalarials/administration & dosage , Europe , Parasitemia/immunology , Adolescent , Vaccines, Attenuated/immunology , Vaccines, Attenuated/administration & dosage , European People
2.
Front Immunol ; 15: 1372584, 2024.
Article in English | MEDLINE | ID: mdl-38745665

ABSTRACT

Among Plasmodium spp. responsible for human malaria, Plasmodium vivax ranks as the second most prevalent and has the widest geographical range; however, vaccine development has lagged behind that of Plasmodium falciparum, the deadliest Plasmodium species. Recently, we developed a multistage vaccine for P. falciparum based on a heterologous prime-boost immunization regimen utilizing the attenuated vaccinia virus strain LC16m8Δ (m8Δ)-prime and adeno-associated virus type 1 (AAV1)-boost, and demonstrated 100% protection and more than 95% transmission-blocking (TB) activity in the mouse model. In this study, we report the feasibility and versatility of this vaccine platform as a P. vivax multistage vaccine, which can provide 100% sterile protection against sporozoite challenge and >95% TB efficacy in the mouse model. Our vaccine comprises m8Δ and AAV1 viral vectors, both harboring the gene encoding two P. vivax circumsporozoite (PvCSP) protein alleles (VK210; PvCSP-Sal and VK247; -PNG) and P25 (Pvs25) expressed as a Pvs25-PvCSP fusion protein. For protective efficacy, the heterologous m8Δ-prime/AAV1-boost immunization regimen showed 100% (short-term; Day 28) and 60% (long-term; Day 242) protection against PvCSP VK210 transgenic Plasmodium berghei sporozoites. For TB efficacy, mouse sera immunized with the vaccine formulation showed >75% TB activity and >95% transmission reduction activity by a direct membrane feeding assay using P. vivax isolates in blood from an infected patient from the Brazilian Amazon region. These findings provide proof-of-concept that the m8Δ/AAV1 vaccine platform is sufficiently versatile for P. vivax vaccine development. Future studies are needed to evaluate the safety, immunogenicity, vaccine efficacy, and synergistic effects on protection and transmission blockade in a non-human primate model for Phase I trials.


Subject(s)
Dependovirus , Genetic Vectors , Malaria Vaccines , Malaria, Vivax , Plasmodium vivax , Animals , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Plasmodium vivax/immunology , Plasmodium vivax/genetics , Malaria, Vivax/prevention & control , Malaria, Vivax/transmission , Malaria, Vivax/immunology , Mice , Dependovirus/genetics , Dependovirus/immunology , Female , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Antibodies, Protozoan/immunology , Antibodies, Protozoan/blood , Disease Models, Animal , Vaccinia virus/genetics , Vaccinia virus/immunology , Humans , Mice, Inbred BALB C , Immunization, Secondary , Vaccine Efficacy
3.
Vaccine ; 42(16): 3621-3629, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38704253

ABSTRACT

Recent data indicate increasing disease burden and importance of Plasmodium vivax (Pv) malaria. A robust assay will be essential for blood-stage Pv vaccine development. Results of the in vitro growth inhibition assay (GIA) with transgenic P. knowlesi (Pk) parasites expressing the Pv Duffy-binding protein region II (PvDBPII) correlate with in vivo protection in the first PvDBPII controlled human malaria infection (CHMI) trials, making the PkGIA an ideal selection tool once the precision of the assay is defined. To determine the precision in percentage of inhibition in GIA (%GIA) and in GIA50 (antibody concentration that gave 50 %GIA), ten GIAs with transgenic Pk parasites were conducted with four different anti-PvDBPII human monoclonal antibodies (mAbs) at concentrations of 0.016 to 2 mg/mL, and three GIAs with eighty anti-PvDBPII human polyclonal antibodies (pAbs) at 10 mg/mL. A significant assay-to-assay variation was observed, and the analysis revealed a standard deviation (SD) of 13.1 in the mAb and 5.94 in the pAb dataset for %GIA, with a LogGIA50 SD of 0.299 (for mAbs). Moreover, the ninety-five percent confidence interval (95 %CI) for %GIA or GIA50 in repeat assays was calculated in this investigation. The error range determined in this study will help researchers to compare PkGIA results from different assays and studies appropriately, thus supporting the development of future blood-stage malaria vaccine candidates, specifically second-generation PvDBPII-based formulations.


Subject(s)
Antibodies, Protozoan , Antigens, Protozoan , Malaria Vaccines , Plasmodium knowlesi , Plasmodium vivax , Protozoan Proteins , Receptors, Cell Surface , Malaria Vaccines/immunology , Plasmodium knowlesi/immunology , Plasmodium knowlesi/genetics , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Plasmodium vivax/immunology , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Humans , Receptors, Cell Surface/immunology , Receptors, Cell Surface/genetics , Antibodies, Protozoan/immunology , Antibodies, Protozoan/blood , Malaria, Vivax/prevention & control , Malaria, Vivax/immunology , Antibodies, Monoclonal/immunology , Vaccine Development/methods , Animals
4.
Malar J ; 23(1): 136, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711053

ABSTRACT

Malaria vaccine introduction in endemic countries is a game-changing milestone in the fight against the disease. This article examines the inequity in the global pharmaceutical research, development, manufacturing, and trade landscape. The role of inequity in hindering progress towards malaria elimination is explored. The analysis finds that transformational changes are required to create an equity-enabling environment. Addressing the inequity is critical to maximizing the public health impact of vaccines and attaining sustainability. Avenues to catalyze progress by leveraging malaria vaccines and messenger ribonucleic acid (mRNA) technology are discussed.


Subject(s)
Malaria Vaccines , Malaria , Malaria Vaccines/immunology , Malaria Vaccines/genetics , Humans , Malaria/prevention & control , Disease Eradication/methods , RNA, Messenger/genetics , Global Health , Pharmaceutical Research
5.
Vaccine ; 42(15): 3379-3383, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38704250

ABSTRACT

The Immunization and Vaccine-related Implementation Research Advisory Committee (IVIR-AC) is the World Health Organization's key standing advisory body to conduct an independent review of research, particularly of transmission and economic modeling analyses that estimate the impact and value of vaccines. From 26th February-1st March 2024, at its first of two semi-annual meetings, IVIR-AC provided feedback and recommendations across four sessions; this report summarizes the proceedings and recommendations from that meeting. Session topics included modeling of the impact and cost-effectiveness of the R21/Matrix-M malaria vaccine, meta-analysis of economic evaluations of vaccines, a global analysis estimating the impact of vaccination over the last 50 years, and modeling the impact of different RTS,S malaria vaccine dose schedules in seasonal settings.


Subject(s)
Advisory Committees , Malaria Vaccines , World Health Organization , Humans , Malaria Vaccines/administration & dosage , Malaria Vaccines/immunology , Cost-Benefit Analysis , Vaccination/methods , Malaria/prevention & control , Immunization/methods
6.
NPJ Syst Biol Appl ; 10(1): 44, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678051

ABSTRACT

Malaria vaccine development is hampered by extensive antigenic variation and complex life stages of Plasmodium species. Vaccine development has focused on a small number of antigens, many of which were identified without utilizing systematic genome-level approaches. In this study, we implement a machine learning-based reverse vaccinology approach to predict potential new malaria vaccine candidate antigens. We assemble and analyze P. falciparum proteomic, structural, functional, immunological, genomic, and transcriptomic data, and use positive-unlabeled learning to predict potential antigens based on the properties of known antigens and remaining proteins. We prioritize candidate antigens based on model performance on reference antigens with different genetic diversity and quantify the protein properties that contribute most to identifying top candidates. Candidate antigens are characterized by gene essentiality, gene ontology, and gene expression in different life stages to inform future vaccine development. This approach provides a framework for identifying and prioritizing candidate vaccine antigens for a broad range of pathogens.


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/immunology , Malaria, Falciparum/prevention & control , Machine Learning , Humans , Proteomics/methods , Vaccine Development/methods , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Computational Biology/methods
7.
Front Immunol ; 15: 1331474, 2024.
Article in English | MEDLINE | ID: mdl-38650939

ABSTRACT

Malaria remains a global health challenge, necessitating the development of effective vaccines. The RTS,S vaccination prevents Plasmodium falciparum (Pf) malaria but is ineffective against Plasmodium vivax (Pv) disease. Herein, we evaluated the murine immunogenicity of a recombinant PvCSP incorporating prevalent polymorphisms, adjuvanted with Alhydrogel or Poly I:C. Both formulations induced prolonged IgG responses, with IgG1 dominance by the Alhydrogel group and high titers of all IgG isotypes by the Poly I:C counterpart. Poly I:C-adjuvanted vaccination increased splenic plasma cells, terminally-differentiated memory cells (MBCs), and precursors relative to the Alhydrogel-combined immunization. Splenic B-cells from Poly I:C-vaccinated mice revealed an antibody-secreting cell- and MBC-differentiating gene expression profile. Biological processes such as antibody folding and secretion were highlighted by the Poly I:C-adjuvanted vaccination. These findings underscore the potential of Poly I:C to strengthen immune responses against Pv malaria.


Subject(s)
Aluminum Hydroxide , Antibodies, Protozoan , Immunoglobulin G , Malaria Vaccines , Malaria, Vivax , Plasmodium vivax , Poly I-C , Protozoan Proteins , Animals , Malaria Vaccines/immunology , Protozoan Proteins/immunology , Protozoan Proteins/genetics , Mice , Plasmodium vivax/immunology , Antibodies, Protozoan/immunology , Poly I-C/immunology , Malaria, Vivax/immunology , Malaria, Vivax/prevention & control , Aluminum Hydroxide/immunology , Immunoglobulin G/immunology , Immunoglobulin G/blood , Female , Adjuvants, Immunologic , Immunity, Humoral , Immunity, Cellular , Mice, Inbred BALB C
9.
Int Immunopharmacol ; 132: 111982, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38569430

ABSTRACT

RTS,S is the first malaria vaccine recommended for implementation among young children at risk. However, vaccine efficacy is modest and short-lived. To mitigate the risk of cerebral malaria (CM) among children under the age of 5, it is imperative to develop new vaccines. EVs are potential vaccine candidates as they obtain the ability of brain-targeted delivery and transfer plasmodium antigens and immunomodulators during infections. This study extracted EVs from BALB/c mice infected with Plasmodium yoelii 17XNL (P.y17XNL). C57BL/6J mice were intravenously immunized with EVs (EV-I.V. + CM group) or subcutaneously vaccinated with the combination of EVs and CpG ODN-1826 (EV + CPG ODN-S.C. + CM group) on days 0 and 20, followed by infection with Plasmodium berghei ANKA (P.bANKA) on day 20 post-second immunization. We monitored Parasitemia and survival rate. The integrity of the Blood-brain barrier (BBB) was examined using Evans blue staining.The levels of cytokines and adhesion molecules were evaluated using Luminex, RT-qPCR, and WB. Brain pathology was evaluated by hematoxylin and eosin and immunohistochemical staining. The serum levels of IgG, IgG1, and IgG2a were analyzed by enzyme-linked immunosorbent assay. Compared with those in the P.bANKA-infected group, parasitemia increased slowly, death was delayed (day 10 post-infection), and the survival rate reached 75 %-83.3 % in the EV-I.V. + ECM and EV + CPG ODN-S.C. + ECM groups. Meanwhile, compared with the EV + CPG ODN-S.C. + ECM group, although parasitemia was almost the same, the survival rate increased in the EV-I.V. + ECM group.Additionally, EVs immunization markedly downregulated inflammatory responses in the spleen and brain and ameliorated brain pathological changes, including BBB disruption and infected red blood cell (iRBC) sequestration. Furthermore, the EVs immunization group exhibited enhanced antibody responses (upregulation of IgG1 and IgG2a production) compared to the normal control group. EV immunization exerted protective effects, improving the integrity of the BBB, downregulating inflammation response of brain tissue, result in reduces the incidence of CM. The protective effects were determined by immunological pathways and brain targets elicited by EVs. Intravenous immunization exhibited better performance than subcutaneous immunization, which perhaps correlated with EVs, which can naturally cross BBB to play a better role in brain protection.


Subject(s)
Blood-Brain Barrier , Erythrocytes , Extracellular Vesicles , Malaria, Cerebral , Mice, Inbred BALB C , Mice, Inbred C57BL , Oligodeoxyribonucleotides , Plasmodium berghei , Animals , Malaria, Cerebral/immunology , Malaria, Cerebral/parasitology , Malaria, Cerebral/prevention & control , Plasmodium berghei/immunology , Extracellular Vesicles/immunology , Erythrocytes/parasitology , Erythrocytes/immunology , Blood-Brain Barrier/immunology , Mice , Oligodeoxyribonucleotides/administration & dosage , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Female , Brain/parasitology , Brain/immunology , Brain/pathology , Cytokines/metabolism , Cytokines/blood , Plasmodium yoelii/immunology , Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Parasitemia/immunology , Disease Models, Animal , Immunoglobulin G/blood , Immunoglobulin G/immunology
10.
Acta Trop ; 255: 107231, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38685340

ABSTRACT

Malaria remains a public health challenge. Since many control strategies have proven ineffective in eradicating this disease, new strategies are required, among which the design of a multivalent vaccine stands out. However, the effectiveness of this strategy has been hindered, among other reasons, by the genetic diversity observed in parasite antigens. In Plasmodium vivax, the Erythrocyte Binding Protein (PvEBP, also known as DBP2) is an alternate ligand to Duffy Binding Protein (DBP); given its structural resemblance to DBP, EBP/DBP2 is proposed as a promising antigen for inclusion in vaccine design. However, the extent of genetic diversity within the locus encoding this protein has not been comprehensively assessed. Thus, this study aimed to characterize the genetic diversity of the locus encoding the P. vivax EBP/DBP2 protein and to determine the evolutionary mechanisms modulating this diversity. Several intrapopulation genetic variation parameters were estimated using 36 gene sequences of PvEBP/DBP2 from Colombian P. vivax clinical isolates and 186 sequences available in databases. The study then evaluated the worldwide genetic structure and the evolutionary forces that may influence the observed patterns of genetic variation. It was found that the PvEBP/DBP2 gene exhibits one of the lowest levels of genetic diversity compared to other vaccine-candidate antigens. Four major haplotypes were shared worldwide. Analysis of the protein's 3D structure and epitope prediction identified five regions with potential antigenic properties. The results suggest that the PvEBP/DBP2 protein possesses ideal characteristics to be considered when designing a multivalent effective antimalarial vaccine against P. vivax.


Subject(s)
Antigens, Protozoan , Genetic Variation , Malaria Vaccines , Malaria, Vivax , Plasmodium vivax , Protozoan Proteins , Plasmodium vivax/genetics , Plasmodium vivax/immunology , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Humans , Malaria Vaccines/immunology , Malaria Vaccines/genetics , Malaria, Vivax/prevention & control , Malaria, Vivax/parasitology , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Colombia , Phylogeny , Receptors, Cell Surface
11.
Trends Parasitol ; 40(5): 362-366, 2024 May.
Article in English | MEDLINE | ID: mdl-38582683

ABSTRACT

The Equatorial Guinea Malaria Vaccine Initiative (EGMVI) highlights how long-term African government and international energy industry investment, plus novel partnerships, can enable clinical development of vaccines in Africa, for Africa. We review achievements and challenges of this pioneering, award-winning, public-private partnership which offers a model for future Africa-centric clinical research and development (R&D).


Subject(s)
Malaria Vaccines , Vaccine Development , Equatorial Guinea , Malaria Vaccines/immunology , Humans , Malaria/prevention & control , Public-Private Sector Partnerships , Africa
12.
BMC Med ; 22(1): 170, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38649867

ABSTRACT

BACKGROUND: The stalling global progress in malaria control highlights the need for novel tools for malaria elimination, including transmission-blocking vaccines. Transmission-blocking vaccines aim to induce human antibodies that block parasite development in the mosquito and mosquitoes becoming infectious. The Pfs48/45 protein is a leading Plasmodium falciparum transmission-blocking vaccine candidate. The R0.6C fusion protein, consisting of Pfs48/45 domain 3 (6C) and the N-terminal region of P. falciparum glutamate-rich protein (R0), has previously been produced in Lactococcus lactis and elicited functional antibodies in rodents. Here, we assess the safety and transmission-reducing efficacy of R0.6C adsorbed to aluminium hydroxide with and without Matrix-M™ adjuvant in humans. METHODS: In this first-in-human, open-label clinical trial, malaria-naïve adults, aged 18-55 years, were recruited at the Radboudumc in Nijmegen, the Netherlands. Participants received four intramuscular vaccinations on days 0, 28, 56 and 168 with either 30 µg or 100 µg of R0.6C and were randomised for the allocation of one of the two different adjuvant combinations: aluminium hydroxide alone, or aluminium hydroxide combined with Matrix-M1™ adjuvant. Adverse events were recorded from inclusion until 84 days after the fourth vaccination. Anti-R0.6C and anti-6C IgG titres were measured by enzyme-linked immunosorbent assay. Transmission-reducing activity of participants' serum and purified vaccine-specific immunoglobulin G was assessed by standard membrane feeding assays using laboratory-reared Anopheles stephensi mosquitoes and cultured P. falciparum gametocytes. RESULTS: Thirty-one participants completed four vaccinations and were included in the analysis. Administration of all doses was safe and well-tolerated, with one related grade 3 adverse event (transient fever) and no serious adverse events occurring. Anti-R0.6C and anti-6C IgG titres were similar between the 30 and 100 µg R0.6C arms, but higher in Matrix-M1™ arms. Neat participant sera did not induce significant transmission-reducing activity in mosquito feeding experiments, but concentrated vaccine-specific IgGs purified from sera collected two weeks after the fourth vaccination achieved up to 99% transmission-reducing activity. CONCLUSIONS: R0.6C/aluminium hydroxide with or without Matrix-M1™ is safe, immunogenic and induces functional Pfs48/45-specific transmission-blocking antibodies, albeit at insufficient serum concentrations to result in transmission reduction by neat serum. Future work should focus on identifying alternative vaccine formulations or regimens that enhance functional antibody responses. TRIAL REGISTRATION: The trial is registered with ClinicalTrials.gov under identifier NCT04862416.


Subject(s)
Malaria Vaccines , Malaria, Falciparum , Membrane Glycoproteins , Plasmodium falciparum , Protozoan Proteins , Adolescent , Adult , Animals , Female , Humans , Male , Middle Aged , Young Adult , Adjuvants, Immunologic/administration & dosage , Aluminum Hydroxide/administration & dosage , Antibodies, Protozoan , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Malaria, Falciparum/transmission , Malaria, Falciparum/immunology , Netherlands , Plasmodium falciparum/immunology , Protozoan Proteins/immunology
13.
Trends Parasitol ; 40(5): 367-368, 2024 May.
Article in English | MEDLINE | ID: mdl-38604871

ABSTRACT

Recently, Day et al. identified a receptor-binding site on the malaria parasite protein PfCyRPA that binds the host sugar Neu5Ac, and they found that disrupting this interaction impedes parasite growth. A map of the receptor-binding site identifies an attractive target for antimalarial vaccines and therapeutics.


Subject(s)
Protozoan Proteins , Protozoan Proteins/metabolism , Binding Sites , Humans , Malaria/prevention & control , Malaria/parasitology , Plasmodium falciparum/metabolism , Malaria Vaccines/immunology , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use
14.
Am J Trop Med Hyg ; 110(5): 892-901, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38531102

ABSTRACT

Malaria eradication efforts prioritize safe and efficient vaccination strategies, although none with high-level efficacy against malaria infection are yet available. Among several vaccine candidates, Sanaria® PfSPZ Vaccine and Sanaria PfSPZ-CVac are, respectively, live radiation- and chemo-attenuated sporozoite vaccines designed to prevent infection with Plasmodium falciparum, the leading cause of malaria-related morbidity and mortality. We are conducting a randomized normal saline placebo-controlled trial called IDSPZV1 that will analyze the safety, tolerability, immunogenicity, and efficacy of PfSPZ Vaccine and PfSPZ-CVac administered pre-deployment to malaria-naive Indonesian soldiers assigned to temporary duties in a high malaria transmission area. We describe the manifold challenges of enrolling and immunizing 345 soldier participants at their home base in western Indonesia before their nearly 6,000-km voyage to eastern Indonesia, where they are being monitored for incident P. falciparum and Plasmodium vivax malaria cases during 9 months of exposure. The unique regulatory, ethical, and operational complexities of this trial demonstrate the importance of thorough planning, frequent communication, and close follow-up with stakeholders. Effective engagement with the military community and the ability to adapt to unanticipated events have proven key to the success of this trial.


Subject(s)
Malaria Vaccines , Malaria, Falciparum , Malaria, Vivax , Military Personnel , Plasmodium falciparum , Sporozoites , Vaccines, Attenuated , Humans , Malaria Vaccines/immunology , Malaria Vaccines/therapeutic use , Malaria Vaccines/administration & dosage , Indonesia/epidemiology , Malaria, Falciparum/prevention & control , Malaria, Falciparum/epidemiology , Sporozoites/immunology , Vaccines, Attenuated/immunology , Vaccines, Attenuated/therapeutic use , Plasmodium falciparum/immunology , Malaria, Vivax/prevention & control , Malaria, Vivax/epidemiology , Male , Adult , Young Adult , Plasmodium vivax/immunology , Female
16.
Lancet Infect Dis ; 24(5): 465-475, 2024 May.
Article in English | MEDLINE | ID: mdl-38342107

ABSTRACT

BACKGROUND: The R21/Matrix-M vaccine has demonstrated high efficacy against Plasmodium falciparum clinical malaria in children in sub-Saharan Africa. Using trial data, we aimed to estimate the public health impact and cost-effectiveness of vaccine introduction across sub-Saharan Africa. METHODS: We fitted a semi-mechanistic model of the relationship between anti-circumsporozoite protein antibody titres and vaccine efficacy to data from 3 years of follow-up in the phase 2b trial of R21/Matrix-M in Nanoro, Burkina Faso. We validated the model by comparing predicted vaccine efficacy to that observed over 12-18 months in the phase 3 trial. Integrating this framework within a mathematical transmission model, we estimated the cases, malaria deaths, and disability-adjusted life-years (DALYs) averted and cost-effectiveness over a 15-year time horizon across a range of transmission settings in sub-Saharan Africa. Cost-effectiveness was estimated incorporating the cost of vaccine introduction (dose, consumables, and delivery) relative to existing interventions at baseline. We report estimates at a median of 20% parasite prevalence in children aged 2-10 years (PfPR2-10) and ranges from 3% to 65% PfPR2-10. FINDINGS: Anti-circumsporozoite protein antibody titres were found to satisfy the criteria for a surrogate of protection for vaccine efficacy against clinical malaria. Age-based implementation of a four-dose regimen of R21/Matrix-M vaccine was estimated to avert 181 825 (range 38 815-333 491) clinical cases per 100 000 fully vaccinated children in perennial settings and 202 017 (29 868-405 702) clinical cases per 100 000 fully vaccinated children in seasonal settings. Similar estimates were obtained for seasonal or hybrid implementation. Under an assumed vaccine dose price of US$3, the incremental cost per clinical case averted was $7 (range 4-48) in perennial settings and $6 (3-63) in seasonal settings and the incremental cost per DALY averted was $34 (29-139) in perennial settings and $30 (22-172) in seasonal settings, with lower cost-effectiveness ratios in settings with higher PfPR2-10. INTERPRETATION: Introduction of the R21/Matrix-M malaria vaccine could have a substantial public health benefit across sub-Saharan Africa. FUNDING: The Wellcome Trust, the Bill & Melinda Gates Foundation, the UK Medical Research Council, the European and Developing Countries Clinical Trials Partnership 2 and 3, the NIHR Oxford Biomedical Research Centre, and the Serum Institute of India, Open Philanthropy.


Subject(s)
Cost-Benefit Analysis , Malaria Vaccines , Malaria, Falciparum , Models, Theoretical , Public Health , Humans , Malaria Vaccines/economics , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Malaria, Falciparum/prevention & control , Malaria, Falciparum/epidemiology , Malaria, Falciparum/economics , Burkina Faso/epidemiology , Child, Preschool , Public Health/economics , Plasmodium falciparum/immunology , Child , Protozoan Proteins/immunology , Antibodies, Protozoan/blood , Vaccine Efficacy , Infant , Male , Female
17.
J Clin Invest ; 134(6)2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38194272

ABSTRACT

BACKGROUNDSanaria PfSPZ Vaccine, composed of attenuated Plasmodium falciparum (Pf) sporozoites (SPZ), protects against malaria. We conducted this clinical trial to assess the safety and efficacy of PfSPZ Vaccine in HIV-positive (HIV+) individuals, since the HIV-infection status of participants in mass vaccination programs may be unknown.METHODSThis randomized, double-blind, placebo-controlled trial enrolled 18- to 45-year-old HIV-negative (HIV-) and well-controlled HIV+ Tanzanians (HIV viral load <40 copies/mL, CD4 counts >500 cells/µL). Participants received 5 doses of PfSPZ Vaccine or normal saline (NS) over 28 days, followed by controlled human malaria infection (CHMI) 3 weeks later.RESULTSThere were no solicited adverse events in the 9 HIV- and 12 HIV+ participants. After CHMI, 6 of 6 NS controls, 1 of 5 HIV- vaccinees, and 4 of 4 HIV+ vaccinees were Pf positive by quantitative PCR (qPCR). After immunization, anti-Pf circumsporozoite protein (anti-PfCSP) (isotype and IgG subclass) and anti-PfSPZ antibodies, anti-PfSPZ CD4+ T cell responses, and Vδ2+ γδ CD3+ T cells were nonsignificantly higher in HIV- than in HIV+ vaccinees. Sera from HIV- vaccinees had significantly higher inhibition of PfSPZ invasion of hepatocytes in vitro and antibody-dependent complement deposition (ADCD) and Fcγ3B binding by anti-PfCSP and ADCD by anti-cell-traversal protein for ookinetes and SPZ (anti-PfCelTOS) antibodies.CONCLUSIONSPfSPZ Vaccine was safe and well tolerated in HIV+ vaccinees, but not protective. Vaccine efficacy was 80% in HIV- vaccinees (P = 0.012), whose sera had significantly higher inhibition of PfSPZ invasion of hepatocytes and enrichment of multifunctional PfCSP antibodies. A more potent PfSPZ vaccine or regimen is needed to protect those living with HIV against Pf infection in Africa.TRIAL REGISTRATIONClinicalTrials.gov NCT03420053.FUNDINGEquatorial Guinea Malaria Vaccine Initiative (EGMVI), made up of the Government of Equatorial Guinea Ministries of Mines and Hydrocarbons, and Health and Social Welfare, Marathon Equatorial Guinea Production Limited, Noble Energy, Atlantic Methanol Production Company, and EG LNG; Swiss government, through ESKAS scholarship grant no. 2016.0056; Intramural Research Program of the National Institute of Allergy and Infectious Diseases, NIH; NIH grant 1U01AI155354-01.


Subject(s)
HIV Infections , Malaria Vaccines , Malaria, Falciparum , Adolescent , Adult , Humans , Middle Aged , Young Adult , Antibodies, Protozoan , East African People , HIV Infections/complications , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum , Tanzania , HIV Seronegativity , HIV Seropositivity , Vaccine Efficacy
18.
Nature ; 625(7995): 578-584, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38123677

ABSTRACT

The symptoms of malaria occur during the blood stage of infection, when parasites invade and replicate within human erythrocytes. The PfPCRCR complex1, containing PfRH5 (refs. 2,3), PfCyRPA, PfRIPR, PfCSS and PfPTRAMP, is essential for erythrocyte invasion by the deadliest human malaria parasite, Plasmodium falciparum. Invasion can be prevented by antibodies3-6 or nanobodies1 against each of these conserved proteins, making them the leading blood-stage malaria vaccine candidates. However, little is known about how PfPCRCR functions during invasion. Here we present the structure of the PfRCR complex7,8, containing PfRH5, PfCyRPA and PfRIPR, determined by cryogenic-electron microscopy. We test the hypothesis that PfRH5 opens to insert into the membrane9, instead showing that a rigid, disulfide-locked PfRH5 can mediate efficient erythrocyte invasion. We show, through modelling and an erythrocyte-binding assay, that PfCyRPA-binding antibodies5 neutralize invasion through a steric mechanism. We determine the structure of PfRIPR, showing that it consists of an ordered, multidomain core flexibly linked to an elongated tail. We also show that the elongated tail of PfRIPR, which is the target of growth-neutralizing antibodies6, binds to the PfCSS-PfPTRAMP complex on the parasite membrane. A modular PfRIPR is therefore linked to the merozoite membrane through an elongated tail, and its structured core presents PfCyRPA and PfRH5 to interact with erythrocyte receptors. This provides fresh insight into the molecular mechanism of erythrocyte invasion and opens the way to new approaches in rational vaccine design.


Subject(s)
Erythrocytes , Malaria, Falciparum , Multiprotein Complexes , Parasites , Plasmodium falciparum , Protozoan Proteins , Animals , Humans , Antibodies, Neutralizing/immunology , Antigens, Protozoan/chemistry , Antigens, Protozoan/immunology , Cryoelectron Microscopy , Disulfides/chemistry , Disulfides/metabolism , Erythrocytes/metabolism , Erythrocytes/parasitology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Merozoites/metabolism , Multiprotein Complexes/chemistry , Multiprotein Complexes/immunology , Multiprotein Complexes/metabolism , Multiprotein Complexes/ultrastructure , Parasites/metabolism , Parasites/pathogenicity , Plasmodium falciparum/metabolism , Plasmodium falciparum/pathogenicity , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Protozoan Proteins/ultrastructure
19.
Med ; 4(10): 668-686.e7, 2023 Oct 13.
Article in English | MEDLINE | ID: mdl-37572659

ABSTRACT

BACKGROUND: RH5 is a leading blood-stage candidate antigen for a Plasmodium falciparum vaccine; however, its safety and immunogenicity in malaria-endemic populations are unknown. METHODS: A phase 1b, single-center, dose-escalation, age-de-escalation, double-blind, randomized, controlled trial was conducted in Bagamoyo, Tanzania (NCT03435874). Between 12th April and 25th October 2018, 63 healthy adults (18-35 years), young children (1-6 years), and infants (6-11 months) received a priming dose of viral-vectored ChAd63 RH5 or rabies control vaccine. Sixty participants were boosted with modified vaccinia virus Ankara (MVA) RH5 or rabies control vaccine 8 weeks later and completed 6 months of follow-up post priming. Primary outcomes were the number of solicited and unsolicited adverse events post vaccination and the number of serious adverse events over the study period. Secondary outcomes included measures of the anti-RH5 immune response. FINDINGS: Vaccinations were well tolerated, with profiles comparable across groups. No serious adverse events were reported. Vaccination induced RH5-specific cellular and humoral responses. Higher anti-RH5 serum immunoglobulin G (IgG) responses were observed post boost in young children and infants compared to adults. Vaccine-induced antibodies showed growth inhibition activity (GIA) in vitro against P. falciparum blood-stage parasites; their highest levels were observed in infants. CONCLUSIONS: The ChAd63-MVA RH5 vaccine shows acceptable safety and reactogenicity and encouraging immunogenicity in children and infants residing in a malaria-endemic area. The levels of functional GIA observed in RH5-vaccinated infants are the highest reported to date following human vaccination. These data support onward clinical development of RH5-based blood-stage vaccines to protect against clinical malaria in young African infants. FUNDING: Medical Research Council, London, UK.


Subject(s)
Malaria Vaccines , Malaria, Falciparum , Adult , Child , Child, Preschool , Humans , Infant , Adenoviruses, Simian , Antibodies, Viral , Malaria Vaccines/adverse effects , Malaria Vaccines/immunology , Malaria, Falciparum/prevention & control , Rabies , Tanzania , Adolescent , Young Adult , Double-Blind Method
20.
Viruses ; 15(2)2023 01 23.
Article in English | MEDLINE | ID: mdl-36851527

ABSTRACT

Virus-like particles (VLPs), composed of the small hepatitis B virus surface antigen (HBsAgS), are the antigenic components of the hepatitis B virus (HBV) vaccine and represent the backbones for a chimeric anti-malaria vaccine and various vaccine candidates. Biological vectors have to face pre-existing anti-vector immune responses due to previous immune exposure. Vector recognition after natural infections or vaccinations can result in unwarranted outcomes, with compromising effects on clinical outcomes. In order to evaluate the impact of a pre-existing anti-HBsAgS immune response, we developed mutant VLPs composed of subunits with reduced HBsAgS-specific antigenicity. The insertion of a Plasmodium falciparum circumsporozoite protein (CSP)-derived epitope as a read-out allowed the assessment of wild type (wt) and mutant VLPs in the context of a pre-existing immune response. Mutant and wt VLP platforms with a CSP-epitope insert are immunogenic and have the ability to generate anti-CSP antibody responses in both naïve BALB/c mice and mice with a pre-existing anti-HBsAgS immune response, but with superior anti-CSP responses in mice with a pre-existing immunity. The data indicate that previous HBsAgS exposure facilitates enhanced antibody responses against foreign epitopes delivered by the HBsAgS platform, and, in this context, the state of immune sensitization alters the outcome of subsequent vaccinations.


Subject(s)
Hepatitis B Surface Antigens , Immunogenicity, Vaccine , Malaria Vaccines , Plasmodium falciparum , Vaccines, Virus-Like Particle , Animals , Mice , Epitopes/genetics , Epitopes/immunology , Hepatitis B Surface Antigens/genetics , Hepatitis B Surface Antigens/immunology , Immunogenicity, Vaccine/genetics , Immunogenicity, Vaccine/immunology , Malaria/prevention & control , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Mice, Inbred BALB C , Models, Animal , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Vaccination , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...