Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 467
Filter
1.
Proc Natl Acad Sci U S A ; 121(24): e2320898121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38833464

ABSTRACT

The World Health Organization identifies a strong surveillance system for malaria and its mosquito vector as an essential pillar of the malaria elimination agenda. Anopheles salivary antibodies are emerging biomarkers of exposure to mosquito bites that potentially overcome sensitivity and logistical constraints of traditional entomological surveys. Using samples collected by a village health volunteer network in 104 villages in Southeast Myanmar during routine surveillance, the present study employs a Bayesian geostatistical modeling framework, incorporating climatic and environmental variables together with Anopheles salivary antigen serology, to generate spatially continuous predictive maps of Anopheles biting exposure. Our maps quantify fine-scale spatial and temporal heterogeneity in Anopheles salivary antibody seroprevalence (ranging from 9 to 99%) that serves as a proxy of exposure to Anopheles bites and advances current static maps of only Anopheles occurrence. We also developed an innovative framework to perform surveillance of malaria transmission. By incorporating antibodies against the vector and the transmissible form of malaria (sporozoite) in a joint Bayesian geostatistical model, we predict several foci of ongoing transmission. In our study, we demonstrate that antibodies specific for Anopheles salivary and sporozoite antigens are a logistically feasible metric with which to quantify and characterize heterogeneity in exposure to vector bites and malaria transmission. These approaches could readily be scaled up into existing village health volunteer surveillance networks to identify foci of residual malaria transmission, which could be targeted with supplementary interventions to accelerate progress toward elimination.


Subject(s)
Anopheles , Bayes Theorem , Malaria , Mosquito Vectors , Animals , Anopheles/parasitology , Mosquito Vectors/parasitology , Humans , Malaria/transmission , Malaria/epidemiology , Malaria/immunology , Malaria/parasitology , Seroepidemiologic Studies , Insect Bites and Stings/epidemiology , Insect Bites and Stings/immunology , Insect Bites and Stings/parasitology , Sporozoites/immunology
2.
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 , Adolescent , Adult , Female , Humans , Male , Young Adult , Antimalarials/therapeutic use , Antimalarials/administration & dosage , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chloroquine/therapeutic use , Chloroquine/pharmacology , Europe , European People , Gabon , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Malaria, Falciparum/immunology , Malaria, Falciparum/prevention & control , Parasitemia/immunology , Plasmodium falciparum/immunology , Sporozoites/immunology , Vaccination/methods , Vaccines, Attenuated/immunology , Vaccines, Attenuated/administration & dosage , Central African People
3.
JCI Insight ; 9(11)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38687615

ABSTRACT

A systems analysis was conducted to determine the potential molecular mechanisms underlying differential immunogenicity and protective efficacy results of a clinical trial of the radiation-attenuated whole-sporozoite PfSPZ vaccine in African infants. Innate immune activation and myeloid signatures at prevaccination baseline correlated with protection from P. falciparum parasitemia in placebo controls. These same signatures were associated with susceptibility to parasitemia among infants who received the highest and most protective PfSPZ vaccine dose. Machine learning identified spliceosome, proteosome, and resting DC signatures as prevaccination features predictive of protection after highest-dose PfSPZ vaccination, whereas baseline circumsporozoite protein-specific (CSP-specific) IgG predicted nonprotection. Prevaccination innate inflammatory and myeloid signatures were associated with higher sporozoite-specific IgG Ab response but undetectable PfSPZ-specific CD8+ T cell responses after vaccination. Consistent with these human data, innate stimulation in vivo conferred protection against infection by sporozoite injection in malaria-naive mice while diminishing the CD8+ T cell response to radiation-attenuated sporozoites. These data suggest a dichotomous role of innate stimulation for malaria protection and induction of protective immunity by whole-sporozoite malaria vaccines. The uncoupling of vaccine-induced protective immunity achieved by Abs from more protective CD8+ T cell responses suggests that PfSPZ vaccine efficacy in malaria-endemic settings may be constrained by opposing antigen presentation pathways.


Subject(s)
Immunity, Innate , Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Sporozoites , Vaccines, Attenuated , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Immunity, Innate/immunology , Humans , Animals , Malaria, Falciparum/prevention & control , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Mice , Vaccines, Attenuated/immunology , Vaccines, Attenuated/administration & dosage , Sporozoites/immunology , Sporozoites/radiation effects , CD8-Positive T-Lymphocytes/immunology , Infant , Protozoan Proteins/immunology , Antibodies, Protozoan/immunology , Female , Parasitemia/immunology , Parasitemia/prevention & control , Immunoglobulin G/immunology , Immunoglobulin G/blood , Vaccine Efficacy
4.
Microbes Infect ; 26(5-6): 105343, 2024.
Article in English | MEDLINE | ID: mdl-38670216

ABSTRACT

Hemozoin is a crystal synthesized by Plasmodium parasites during hemoglobin digestion in the erythrocytic stage. The hemozoin released when the parasites egress from the red blood cell, which is complexed with parasite DNA, is cleared from the circulation by circulating and tissue-resident monocytes and macrophages, respectively. Recently, we reported that intravenous administration of purified hemozoin complexed with Plasmodium berghei DNA (HzPbDNA) resulted in an innate immune response that blocked liver stage development of sporozoites that was dose-dependent and time-limited. Here, we further characterize the organismal, cellular, and molecular events associated with this protective innate response in the liver and report that a large proportion of the IV administered HzPbDNA localized to F4/80+ cells in the liver and that the rapid and strong protection against liver-stage development waned quickly such that by 1 week post-HzPbDNA treatment animals were fully susceptible to infection. RNAseq of the liver after IV administration of HzPbDNA demonstrated that the rapid and robust induction of genes associated with the acute phase response, innate immune activation, cellular recruitment, and IFN-γ signaling observed at day 1 was largely absent at day 7. RNAseq analysis implicated NK cells as the major cellular source of IFN-γ. In vivo cell depletion and IFN-γ neutralization experiments supported the hypothesis that tissue-resident macrophages and NK cells are major contributors to the protective response and the NK cell-derived IFN-γ is key to induction of the mechanisms that block sporozoite development in the liver. These findings advance our understanding of the innate immune responses that prevent liver stage malaria infection.


Subject(s)
Hemeproteins , Immunity, Innate , Interferon-gamma , Liver , Malaria , Plasmodium berghei , Sporozoites , Animals , Plasmodium berghei/immunology , Sporozoites/immunology , Malaria/immunology , Malaria/prevention & control , Malaria/parasitology , Hemeproteins/immunology , Mice , Liver/parasitology , Liver/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice, Inbred C57BL , Macrophages/immunology , Macrophages/parasitology , DNA, Protozoan/genetics , Female
5.
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
6.
Infection ; 52(3): 707-722, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38319556

ABSTRACT

BACKGROUND AND OBJECTIVE: Despite the significant burden of Plasmodium falciparum (Pf) malaria and the licensure of two vaccines for use in infants and young children that are partially effective in preventing clinical malaria caused by Pf, a highly effective vaccine against Pf infection is still lacking. Live attenuated vaccines using Pf sporozoites as the immunogen (PfSPZ Vaccines) hold promise for addressing this gap. Here we review the safety and efficacy of two of the most promising PfSPZ approaches: PfSPZ Vaccine (radiation attenuated PfSPZ) and PfSPZ-CVac (chemo-attenuated PfSPZ). METHODS: We conducted a systematic review and meta-analysis by searching PubMed, EMBASE, SCOPUS, CENTRAL, and WOS until 22nd December 2021. We included randomized controlled trials (RCTs) of these two vaccine approaches that measured protection against parasitaemia following controlled human malaria infection (CHMI) in malaria-naive and malaria-exposed adults or following exposure to naturally transmitted Pf malaria in African adults and children (primary outcome) and that also measured the incidence of solicited and unsolicited adverse events as indicators of safety and tolerability after vaccination (secondary outcome). We included randomized controlled trials (RCTs) that measured the detected parasitaemia after vaccination (primary outcome) and the incidence of various solicited and unsolicited adverse events (secondary outcome). The quality of the included RCTs using the Cochrane ROB 1 tool and the quality of evidence using the GRADE system were evaluated. We pooled dichotomous data using the risk ratio (RR) for development of parasitemia in vaccinees relative to controls as a measure of vaccine efficacy (VE), including the corresponding confidence interval (CI). This study was registered with PROSPERO (CRD42022308057). RESULTS: We included 19 RCTs. Pooled RR favoured PfSPZ Vaccine (RR: 0.65 with 95% CI [0.53, 0.79], P = 0.0001) and PfSPZ-table (RR: 0.42 with 95% CI [0.27, 0.67], P = 0.0002) for preventing parasitaemia, relative to normal saline placebo. Pooled RR showed no difference between PfSPZ Vaccine and the control in the occurrence of any solicited adverse event (RR: 1.00 with 95% CI [0.82, 1.23], P = 0.98), any local solicited adverse events (RR: 0.73 with 95% CI [0.49, 1.08], P = 0.11), any systemic solicited adverse events (RR: 0.94 with 95% CI [0.75, 1.17], P = 0.58), and any unsolicited adverse event (RR: 0.93 with 95% CI [0.78, 1.10], P = 0.37). CONCLUSION: PfSPZ and PfSPZ-CVacs showed comparable efficacy. Therefore, they can introduce a promising strategy for malaria prophylaxis, but more large-scale field trials are required to sustain efficacy and yield clinically applicable findings.


Subject(s)
Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Randomized Controlled Trials as Topic , Sporozoites , Vaccines, Attenuated , Humans , Malaria Vaccines/immunology , Malaria Vaccines/adverse effects , Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Malaria, Falciparum/immunology , Parasitemia/prevention & control , Plasmodium falciparum/immunology , Sporozoites/immunology , Vaccines, Attenuated/immunology
7.
Proc Natl Acad Sci U S A ; 119(35): e2209729119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994647

ABSTRACT

Glutaminyl cyclase (QC) modifies N-terminal glutamine or glutamic acid residues of target proteins into cyclic pyroglutamic acid (pGlu). Here, we report the biochemical and functional analysis of Plasmodium QC. We show that sporozoites of QC-null mutants of rodent and human malaria parasites are recognized by the mosquito immune system and melanized when they reach the hemocoel. Detailed analyses of rodent malaria QC-null mutants showed that sporozoite numbers in salivary glands are reduced in mosquitoes infected with QC-null or QC catalytically dead mutants. This phenotype can be rescued by genetic complementation or by disrupting mosquito melanization or phagocytosis by hemocytes. Mutation of a single QC-target glutamine of the major sporozoite surface protein (circumsporozoite protein; CSP) of the rodent parasite Plasmodium berghei also results in melanization of sporozoites. These findings indicate that QC-mediated posttranslational modification of surface proteins underlies evasion of killing of sporozoites by the mosquito immune system.


Subject(s)
Aminoacyltransferases , Culicidae , Malaria , Protein Processing, Post-Translational , Sporozoites , Aminoacyltransferases/immunology , Animals , Culicidae/immunology , Glutamic Acid/metabolism , Glutamine/metabolism , Humans , Malaria/genetics , Malaria/immunology , Malaria/parasitology , Plasmodium berghei/genetics , Plasmodium berghei/immunology , Protein Processing, Post-Translational/immunology , Protozoan Proteins/immunology , Sporozoites/immunology
8.
J Biol Chem ; 298(9): 102241, 2022 09.
Article in English | MEDLINE | ID: mdl-35809642

ABSTRACT

Malaria and other apicomplexan-caused diseases affect millions of humans, agricultural animals, and pets. Cell traversal is a common feature used by multiple apicomplexan parasites to migrate through host cells and can be exploited to develop therapeutics against these deadly parasites. Here, we provide insights into the mechanism of the Cell-traversal protein for ookinetes and sporozoites (CelTOS), a conserved cell-traversal protein in apicomplexan parasites and malaria vaccine candidate. CelTOS has previously been shown to form pores in cell membranes to enable traversal of parasites through cells. We establish roles for the distinct protein regions of Plasmodium vivax CelTOS and examine the mechanism of pore formation. We further demonstrate that CelTOS dimer dissociation is required for pore formation, as disulfide bridging between monomers inhibits pore formation, and this inhibition is rescued by disulfide-bridge reduction. We also show that a helix-destabilizing amino acid, Pro127, allows CelTOS to undergo significant conformational changes to assemble into pores. The flexible C terminus of CelTOS is a negative regulator that limits pore formation. Finally, we highlight that lipid binding is a prerequisite for pore assembly as mutation of a phospholipids-binding site in CelTOS resulted in loss of lipid binding and abrogated pore formation. These findings identify critical regions in CelTOS and will aid in understanding the egress mechanism of malaria and other apicomplexan parasites as well as have implications for studying the function of other essential pore-forming proteins.


Subject(s)
Malaria Vaccines , Malaria, Vivax , Plasmodium vivax , Protozoan Proteins , Binding Sites , Disulfides/chemistry , Humans , Malaria Vaccines/chemistry , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Malaria, Vivax/prevention & control , Phospholipids/immunology , Plasmodium vivax/genetics , Plasmodium vivax/immunology , Proline/chemistry , Proline/genetics , Protein Conformation, alpha-Helical , Protein Multimerization , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Sporozoites/genetics , Sporozoites/immunology
9.
J Immunol ; 208(5): 1292-1304, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35131868

ABSTRACT

Pathogen-specific CD8 T cells face the problem of finding rare cells that present their cognate Ag either in the lymph node or in infected tissue. Although quantitative details of T cell movement strategies in some tissues such as lymph nodes or skin have been relatively well characterized, we still lack quantitative understanding of T cell movement in many other important tissues, such as the spleen, lung, liver, and gut. We developed a protocol to generate stable numbers of liver-located CD8 T cells, used intravital microscopy to record movement patterns of CD8 T cells in livers of live mice, and analyzed these and previously published data using well-established statistical and computational methods. We show that, in most of our experiments, Plasmodium-specific liver-localized CD8 T cells perform correlated random walks characterized by transiently superdiffusive displacement with persistence times of 10-15 min that exceed those observed for T cells in lymph nodes. Liver-localized CD8 T cells typically crawl on the luminal side of liver sinusoids (i.e., are in the blood); simulating T cell movement in digital structures derived from the liver sinusoids illustrates that liver structure alone is sufficient to explain the relatively long superdiffusive displacement of T cells. In experiments when CD8 T cells in the liver poorly attach to the sinusoids (e.g., 1 wk after immunization with radiation-attenuated Plasmodium sporozoites), T cells also undergo Lévy flights: large displacements occurring due to cells detaching from the endothelium, floating with the blood flow, and reattaching at another location. Our analysis thus provides quantitative details of movement patterns of liver-localized CD8 T cells and illustrates how structural and physiological details of the tissue may impact T cell movement patterns.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Movement/physiology , Liver/immunology , Malaria/prevention & control , Plasmodium berghei/immunology , Animals , Capillaries/cytology , Cellular Microenvironment/physiology , Liver/blood supply , Malaria/pathology , Mice , Plasmodium berghei/growth & development , Sporozoites/growth & development , Sporozoites/immunology , Vaccination
10.
PLoS Pathog ; 18(2): e1010282, 2022 02.
Article in English | MEDLINE | ID: mdl-35108339

ABSTRACT

Immunization with radiation-attenuated sporozoites (RAS) can confer sterilizing protection against malaria, although the mechanisms behind this protection are incompletely understood. We performed a systems biology analysis of samples from the Immunization by Mosquito with Radiation Attenuated Sporozoites (IMRAS) trial, which comprised P. falciparum RAS-immunized (PfRAS), malaria-naive participants whose protection from malaria infection was subsequently assessed by controlled human malaria infection (CHMI). Blood samples collected after initial PfRAS immunization were analyzed to compare immune responses between protected and non-protected volunteers leveraging integrative analysis of whole blood RNA-seq, high parameter flow cytometry, and single cell CITEseq of PBMCs. This analysis revealed differences in early innate immune responses indicating divergent paths associated with protection. In particular, elevated levels of inflammatory responses early after the initial immunization were detrimental for the development of protective adaptive immunity. Specifically, non-classical monocytes and early type I interferon responses induced within 1 day of PfRAS vaccination correlated with impaired immunity. Non-protected individuals also showed an increase in Th2 polarized T cell responses whereas we observed a trend towards increased Th1 and T-bet+ CD8 T cell responses in protected individuals. Temporal differences in genes associated with natural killer cells suggest an important role in immune regulation by these cells. These findings give insight into the immune responses that confer protection against malaria and may guide further malaria vaccine development. Trial registration: ClinicalTrials.gov NCT01994525.


Subject(s)
Immunity , Inflammation , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Sporozoites/immunology , Adult , Animals , Anopheles/parasitology , Female , Humans , Immunization/methods , Insect Bites and Stings/immunology , Malaria, Falciparum/parasitology , Male , Mosquito Vectors/parasitology , T-Lymphocytes/immunology , Vaccination/methods , Vaccines, Attenuated/immunology
11.
Front Immunol ; 13: 795463, 2022.
Article in English | MEDLINE | ID: mdl-35197971

ABSTRACT

Protection from liver-stage malaria requires high numbers of CD8+ T cells to find and kill Plasmodium-infected cells. A new malaria vaccine strategy, prime-target vaccination, involves sequential viral-vectored vaccination by intramuscular and intravenous routes to target cellular immunity to the liver. Liver tissue-resident memory (TRM) CD8+ T cells have been shown to be necessary and sufficient for protection against rodent malaria by this vaccine regimen. Ultimately, to most faithfully assess immunotherapeutic responses by these local, specialised, hepatic T cells, periodic liver sampling is necessary, however this is not feasible at large scales in human trials. Here, as part of a phase I/II P. falciparum challenge study of prime-target vaccination, we performed deep immune phenotyping, single-cell RNA-sequencing and kinetics of hepatic fine needle aspirates and peripheral blood samples to study liver CD8+ TRM cells and circulating counterparts. We found that while these peripheral 'TRM-like' cells differed to TRM cells in terms of previously described characteristics, they are similar phenotypically and indistinguishable in terms of key T cell residency transcriptional signatures. By exploring the heterogeneity among liver CD8+ TRM cells at single cell resolution we found two main subpopulations that each share expression profiles with blood T cells. Lastly, our work points towards the potential for using TRM-like cells as a correlate of protection by liver-stage malaria vaccines and, in particular, those adopting a prime-target approach. A simple and reproducible correlate of protection would be particularly valuable in trials of liver-stage malaria vaccines as they progress to phase III, large-scale testing in African infants. We provide a blueprint for understanding and monitoring liver TRM cells induced by a prime-target malaria vaccine approach.


Subject(s)
Malaria Vaccines/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors , Hepatocytes/immunology , Humans , Immunity, Cellular , Immunologic Memory/immunology , Liver/immunology , Malaria/immunology , Plasmodium/immunology , Sporozoites/immunology , Transcriptome , Vaccination
12.
Methods Mol Biol ; 2410: 555-566, 2022.
Article in English | MEDLINE | ID: mdl-34914067

ABSTRACT

Live attenuated vaccines (LAVs) are among the most critical interventions in modern medicine and have already proven their potential to save millions of lives. LAVs are always explored as potential vaccine candidates since they induce an immune response, which is as good as the wild type pathogen. For parasitic diseases, the efficacy of LAVs is still under investigation and needs extensive research to mark their presence in the field. In malaria, live attenuated sporozoites have been evaluated for a vaccine against the liver stage. This vaccine approach is limited due to the highly cumbersome technique of sporozoite isolation and related relapse issues. We have developed a novel vaccine against malaria by expressing Plasmodium falciparum antigens in Leishmania donovani promastigotes. These hybrid, recombinant L. donovani parasites mimicking P. falciparum parasite antigens were analyzed for their anti-malarial efficacy in preclinical studies. We demonstrate the potential of Leishmania spp. parasites in developing an important live vector vaccine against malaria for the induction of protective immune responses. Herein, we describe a method to express malaria parasite antigens in L. donovani promastigotes and analyze its potential for a vaccine against malaria. This methodology can be extended to live, attenuated Leishmania promastigotes parasites to develop LAV against malaria.


Subject(s)
Malaria Vaccines , Malaria, Falciparum , Plasmodium falciparum , Animals , Antigens, Protozoan , Leishmania donovani , Malaria, Falciparum/prevention & control , Parasites , Plasmodium falciparum/immunology , Sporozoites/immunology , Vaccine Development , Vaccines, Attenuated
13.
PLoS Pathog ; 17(12): e1010133, 2021 12.
Article in English | MEDLINE | ID: mdl-34871332

ABSTRACT

Combinations of monoclonal antibodies (mAbs) against different epitopes on the same antigen synergistically neutralize many viruses. However, there are limited studies assessing whether combining human mAbs against distinct regions of the Plasmodium falciparum (Pf) circumsporozoite protein (CSP) enhances in vivo protection against malaria compared to each mAb alone or whether passive transfer of PfCSP mAbs would improve protection following vaccination against PfCSP. Here, we isolated a panel of human mAbs against the subdominant C-terminal domain of PfCSP (C-CSP) from a volunteer immunized with radiation-attenuated Pf sporozoites. These C-CSP-specific mAbs had limited binding to sporozoites in vitro that was increased by combination with neutralizing human "repeat" mAbs against the NPDP/NVDP/NANP tetrapeptides in the central repeat region of PfCSP. Nevertheless, passive transfer of repeat- and C-CSP-specific mAb combinations did not provide enhanced protection against in vivo sporozoite challenge compared to repeat mAbs alone. Furthermore, combining potent repeat-specific mAbs (CIS43, L9, and 317) that respectively target the three tetrapeptides (NPDP/NVDP/NANP) did not provide additional protection against in vivo sporozoite challenge. However, administration of either CIS43, L9, or 317 (but not C-CSP-specific mAbs) to mice that had been immunized with R21, a PfCSP-based virus-like particle vaccine that induces polyclonal antibodies against the repeat region and C-CSP, provided enhanced protection against sporozoite challenge when compared to vaccine or mAbs alone. Collectively, this study shows that while combining mAbs against the repeat and C-terminal regions of PfCSP provide no additional protection in vivo, repeat mAbs do provide increased protection when combined with vaccine-induced polyclonal antibodies. These data should inform the implementation of PfCSP human mAbs alone or following vaccination to prevent malaria infection.


Subject(s)
Antibodies, Monoclonal/immunology , Immunization, Passive/methods , Malaria Vaccines/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/immunology , Humans , Malaria, Falciparum/prevention & control , Mice , Sporozoites/immunology
14.
PLoS Pathog ; 17(11): e1010042, 2021 11.
Article in English | MEDLINE | ID: mdl-34748617

ABSTRACT

Rare and potent monoclonal antibodies (mAbs) against the Plasmodium falciparum (Pf) circumsporozoite protein (CSP) on infective sporozoites (SPZ) preferentially bind the PfCSP junctional tetrapeptide NPDP or NVDP minor repeats while cross-reacting with NANP central repeats in vitro. The extent to which each of these epitopes is required for protection in vivo is unknown. Here, we assessed whether junction-, minor repeat- and central repeat-preferring human mAbs (CIS43, L9 and 317 respectively) bound and protected against in vivo challenge with transgenic P. berghei (Pb) SPZ expressing either PfCSP with the junction and minor repeats knocked out (KO), or PbCSP with the junction and minor repeats knocked in (KI). In vivo protection studies showed that the junction and minor repeats are necessary and sufficient for CIS43 and L9 to neutralize KO and KI SPZ, respectively. In contrast, 317 required major repeats for in vivo protection. These data establish that human mAbs can prevent malaria infection by targeting three different protective epitopes (NPDP, NVDP, NANP) in the PfCSP repeat region. This report will inform vaccine development and the use of mAbs to passively prevent malaria.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Protozoan/immunology , Epitopes/immunology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Sporozoites/immunology , Animals , Female , Liver/immunology , Liver/metabolism , Liver/parasitology , Liver/pathology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mice , Mice, Inbred C57BL , Sporozoites/growth & development
15.
Nat Commun ; 12(1): 6773, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799567

ABSTRACT

After inoculation by the bite of an infected mosquito, Plasmodium sporozoites enter the blood stream and infect the liver, where each infected cell produces thousands of merozoites. These in turn, infect red blood cells and cause malaria symptoms. To initiate a productive infection, sporozoites must exit the circulation by traversing the blood lining of the liver vessels after which they infect hepatocytes with unique specificity. We screened a phage display library for peptides that structurally mimic (mimotope) a sporozoite ligand for hepatocyte recognition. We identified HP1 (hepatocyte-binding peptide 1) that mimics a ~50 kDa sporozoite ligand (identified as phospholipid scramblase). Further, we show that HP1 interacts with a ~160 kDa hepatocyte membrane putative receptor (identified as carbamoyl-phosphate synthetase 1). Importantly, immunization of mice with the HP1 peptide partially protects them from infection by the rodent parasite P. berghei. Moreover, an antibody to the HP1 mimotope inhibits human parasite P. falciparum infection of human hepatocytes in culture. The sporozoite ligand for hepatocyte invasion is a potential novel pre-erythrocytic vaccine candidate.


Subject(s)
Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Phospholipid Transfer Proteins/immunology , Protozoan Proteins/immunology , Sporozoites/immunology , Animals , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Disease Models, Animal , Epitopes/immunology , Female , Hep G2 Cells , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/parasitology , Humans , Liver/enzymology , Liver/parasitology , Malaria Vaccines/immunology , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Male , Mice , Peptide Library , Phospholipid Transfer Proteins/isolation & purification , Phospholipid Transfer Proteins/metabolism , Plasmodium berghei/immunology , Plasmodium berghei/metabolism , Plasmodium falciparum/immunology , Plasmodium falciparum/metabolism , Primary Cell Culture , Protozoan Proteins/isolation & purification , Protozoan Proteins/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sporozoites/metabolism , Vaccines, Subunit/immunology , Vaccines, Subunit/therapeutic use
16.
J Immunol ; 207(11): 2631-2635, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34716185

ABSTRACT

Radiation-attenuated sporozoite (RAS) vaccination offers hope for global malaria control through induction of protective liver-stage-specific memory CD8 T cells. Effective RAS vaccination regimens exist; however, widespread implementation remains unfeasible. A key difficulty resides in the need to administer three or more doses i.v. to achieve sufficient immunity. Strategies to reduce the number of RAS doses are therefore desirable. Here we used mice to model human immune responses to a single, suboptimal weight-normalized RAS dose administered i.v. followed by subunit vaccination to amplify liver-stage-specific memory CD8 T cells. RAS+subunit prime-boost regimens increased the numbers of liver-stage-specific memory CD8 T cells to a level greater than is present after one RAS vaccination. Both i.v. and i.m. subunit vaccine delivery induced immunity in mice, and many vaccinated mice completely cleared liver infection. These findings are particularly relevant to human vaccine development because RAS+subunit prime-boost vaccination would reduce the logistical challenges of multiple RAS-only immunizations.


Subject(s)
Liver Diseases/immunology , Malaria Vaccines/immunology , Malaria/immunology , Sporozoites/immunology , Vaccines, Attenuated/immunology , Vaccines, Subunit/immunology , Animals , Immunization , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Vaccination
17.
Parasit Vectors ; 14(1): 473, 2021 Sep 15.
Article in English | MEDLINE | ID: mdl-34526109

ABSTRACT

BACKGROUND: In characterizing malaria epidemiology, measuring mosquito infectiousness informs the entomological inoculation rate, an important metric of malaria transmission. PCR-based methods have been touted as more sensitive than the current "gold-standard" circumsporozoite (CSP) ELISA. Wider application of PCR-based methods has been limited by lack of specificity for the infectious sporozoite stage. We compared a PCR method for detecting the parasite's mitochondrial (mt) cytochrome oxidase I (COX-I) gene with ELISA for detecting circumsporozoite protein for identification of different life stages of the parasite during development within a mosquito. METHODS: A PCR-based method targeting the Plasmodium mt COX-I gene was compared with the CSP ELISA method to assess infectivity in Anopheles arabiensis colony mosquitoes fed on blood from patients infected with Plasmodium vivax. Mosquitoes were tested at six post-infection time points (days 0.5, 1, 6, 9, 12, 15). The head and thorax and the abdomen for each specimen were tested separately with each method. Agreement between methods at each infection stage was measured using Cohen's kappa measure of test association. RESULTS: Infection status of mosquitoes was assessed in approximately 90 head/thorax and 90 abdomen segments at each time point; in total, 538 head/thorax and 534 abdomen segments were tested. In mosquitoes bisected after 0.5, 1, and 6 days post-infection (dpi), the mt COX-I PCR detected Plasmodium DNA in both the abdomen (88, 78, and 67%, respectively) and head/thorax segments (69, 60, and 44%, respectively), whilst CSP ELISA detected sporozoites in only one abdomen on day 6 post-infection. PCR was also more sensitive than ELISA for detection of Plasmodium in mosquitoes bisected after 9, 12, and 15 dpi in both the head and thorax and abdomen. There was fair agreement between methods for time points 9-15 dpi (κ = 0.312, 95% CI: 0.230-0.394). CONCLUSIONS: The mt COX-I PCR is a highly sensitive, robust method for detecting Plasmodium DNA in mosquitoes, but its limited Plasmodium life-stage specificity cannot be overcome by bisection of the head and thorax from the abdomen prior to PCR. Thus, the mt COX-I PCR is a poor candidate for identifying infectious mosquitoes.


Subject(s)
Anopheles/parasitology , Enzyme-Linked Immunosorbent Assay/standards , Life Cycle Stages/genetics , Plasmodium vivax/genetics , Polymerase Chain Reaction/standards , Sporozoites/genetics , Animals , Enzyme-Linked Immunosorbent Assay/methods , Female , Plasmodium vivax/immunology , Polymerase Chain Reaction/methods , Sporozoites/immunology
18.
Cell Rep ; 36(5): 109489, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34348141

ABSTRACT

Both subunit and attenuated whole-sporozoite vaccination strategies against Plasmodium infection have shown promising initial results in malaria-naive westerners but less efficacy in malaria-exposed individuals in endemic areas. Here, we demonstrate proof of concept by using a rodent malaria model in which non-neutralizing antibodies (nNAbs) can directly interfere with protective anti-circumsporozoite protein (CSP) humoral responses. We characterize a monoclonal antibody, RAM1, against Plasmodium yoelii sporozoite major surface antigen CSP. Unlike the canonical PyCSP repeat domain binding and neutralizing antibody (NAb) 2F6, RAM1 does not inhibit sporozoite traversal or entry of hepatocytes in vitro or infection in vivo. Although 2F6 and RAM1 bind non-overlapping regions of the CSP-repeat domain, pre-treatment with RAM1 abrogates the capacity of NAb to block sporozoite traversal and invasion in vitro. Importantly, RAM1 reduces the efficacy of the polyclonal humoral response against PyCSP in vivo. Collectively, our data provide a proof of concept that nNAbs can alter the efficacy of malaria vaccination.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Protozoan/immunology , Immunity, Humoral , Life Cycle Stages , Liver/parasitology , Plasmodium yoelii/growth & development , Plasmodium yoelii/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Cell Line , Epitopes/immunology , Female , Kinetics , Malaria Vaccines/immunology , Mice, Inbred BALB C , Models, Biological , Protein Binding , Protein Domains , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Sporozoites/immunology , Vaccines, Synthetic/immunology
19.
Malar J ; 20(1): 308, 2021 Jul 09.
Article in English | MEDLINE | ID: mdl-34243763

ABSTRACT

BACKGROUND: Vaccination with radiation-attenuated Plasmodium falciparum sporozoites is known to induce protective immunity. However, the mechanisms underlying this protection remain unclear. In this work, two recent radiation-attenuated sporozoite vaccination studies were used to identify potential transcriptional correlates of vaccination-induced protection. METHODS: Longitudinal whole blood RNAseq transcriptome responses to immunization with radiation-attenuated P. falciparum sporozoites were analysed and compared across malaria-naïve adult participants (IMRAS) and malaria-experienced adult participants (BSPZV1). Parasite dose and method of delivery differed between trials, and immunization regimens were designed to achieve incomplete protective efficacy. Observed protective efficacy was 55% in IMRAS and 20% in BSPZV1. Study vaccine dosings were chosen to elicit both protected and non-protected subjects, so that protection-associated responses could be identified. RESULTS: Analysis of comparable time points up to 1 week after the first vaccination revealed a shared cross-study transcriptional response programme, despite large differences in number and magnitude of differentially expressed genes between trials. A time-dependent regulatory programme of coherent blood transcriptional modular responses was observed, involving induction of inflammatory responses 1-3 days post-vaccination, with cell cycle responses apparent by day 7 in protected individuals from both trials. Additionally, strongly increased induction of inflammation and interferon-associated responses was seen in non-protected IMRAS participants. All individuals, except for non-protected BSPZV1 participants, showed robust upregulation of cell-cycle associated transcriptional responses post vaccination. CONCLUSIONS: In summary, despite stark differences between the two studies, including route of vaccination and status of malaria exposure, responses were identified that were associated with protection after PfRAS vaccination. These comprised a moderate early interferon response peaking 2 days post vaccination, followed by a later proliferative cell cycle response steadily increasing over the first 7 days post vaccination. Non-protection is associated with deviations from this model, observed in this study with over-induction of early interferon responses in IMRAS and failure to mount a cell cycle response in BSPZV1.


Subject(s)
Malaria Vaccines/therapeutic use , Malaria, Falciparum/prevention & control , Antibodies, Protozoan/blood , Clinical Trials as Topic , Humans , Malaria Vaccines/administration & dosage , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Protozoan Proteins/genetics , Sporozoites/genetics , Sporozoites/immunology , Transcription, Genetic , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/therapeutic use
20.
PLoS Pathog ; 17(6): e1009445, 2021 06.
Article in English | MEDLINE | ID: mdl-34181697

ABSTRACT

We conducted a longitudinal study of cryptosporidiosis from birth to three years of age in an urban slum of Dhaka Bangladesh. Fecal DNA was extracted from monthly surveillance samples and diarrheal stool samples collected from 392 infants from birth to three years. A pan-Cryptosporidium qPCR assay was used to identify sub-clinical and symptomatic cryptosporidiosis. Anthropometric measurements were collected quarterly to assess child nutritional status. 31% (121/392) of children experienced a single and 57% (222/392) multiple infections with Cryptosporidium. Repeat infections had a lower burden of parasites in the stool (Cq slope = -1.85; p<0.0001) and were more likely to be sub-clinical (Chi square test for trend; p = 0.01). Repeat infections were associated with the development of growth faltering (Pearson correlation = -0.18; p = 0.0004). High levels of fecal IgA antibodies against the Cryptosporidium Cp23 sporozoite protein at one year of life were associated with a delay in reinfection and amelioration of growth faltering through three years of life (HAZ IgA high responders -1.323 ± 0.932 versus HAZ -1.731 ± 0.984 p = 0.0001). We concluded that nonsterile immunity to cryptosporidiosis in young children was associated with high levels of mucosal IgA anti-Cp23 and protection from diarrhea and growth faltering. Trial Registration: NCT02764918.


Subject(s)
Child Nutrition Disorders/immunology , Child Nutrition Disorders/parasitology , Cryptosporidiosis/immunology , Immunity, Mucosal/immunology , Immunoglobulin A/immunology , Bangladesh , Child, Preschool , Cryptosporidiosis/complications , Diarrhea/parasitology , Female , Humans , Infant , Infant, Newborn , Longitudinal Studies , Male , Protozoan Proteins/immunology , Sporozoites/immunology
SELECTION OF CITATIONS
SEARCH DETAIL