Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2024 Sep 28.
Published in final edited form as: Immunity. 2024 May 23;57(6):1215–1224.e6. doi: 10.1016/j.immuni.2024.05.001

Breadth of Fc-mediated effector function correlates with clinical immunity following human malaria challenge

Irene N Nkumama 1,2,6,#, Rodney Ogwang 2,#, Dennis Odera 1,2, Fauzia Musasia 1, Kennedy Mwai 2,3, Lydia Nyamako 2, Linda Murungi 2, James Tuju 2,4, Kristin Fürle 1, Micha Rosenkranz 1, Rinter Kimathi 2, Patricia Njuguna 2, Mainga Hamaluba 2, Melissa C Kapulu 2, Roland Frank 1; CHMI-SIKA study team , Faith HA Osier 1,2,5,8,*
PMCID: PMC7616646  EMSID: EMS198791  PMID: 38788711

Abstract

Malaria is a life-threatening disease of global health importance, particularly in sub-Saharan Africa. The growth inhibition assay (GIA) is routinely used to evaluate, prioritize, and quantify the efficacy of malaria blood-stage vaccine candidates but does not reliably predict either naturally acquired or vaccine-induced protection. Controlled human malaria challenge studies in semi-immune volunteers provide an unparalleled opportunity to robustly identify mechanistic correlates of protection. We leveraged this platform to undertake a head-to-head comparison of seven functional antibody assays that are relevant to immunity against the erythrocytic merozoite stage of Plasmodium falciparum. Fc-mediated effector functions were strongly associated with protection from clinical symptoms of malaria and exponential parasite multiplication, while the gold standard GIA was not. The breadth of Fc-mediated effector function discriminated clinical immunity following the challenge. These findings present a shift in the understanding of the mechanisms that underpin immunity to malaria and have important implications for vaccine development.

Graphical abstract

graphic file with name EMS198791-f004.jpg

Introduction

Malaria is caused by protozoan parasites of the phylum Apicomplexa and the genus Plasmodium.1 Multiple species of Plasmodia can infect humans, but Plasmodium falciparum (P. falciparum) is the deadliest and a global health priority.2,3 In 2022, it accounted for ~249 million clinical cases and ~608,000 deaths, mostly in sub-Saharan Africa4. The burden of malaria has remained high despite control efforts targeting the mosquito vector or improving parasite detection and clinical treatment.5 Vaccines have been challenging to develop because of the multistage life cycle of the parasite that involves different invasive forms and a complex genome encoding over 5,000 proteins, many of which are polymorphic and variably expressed.2,6 These difficulties are compounded by the lack of reliable immune correlates of protection.

Following decades of effort, two subunit vaccines against P. falciparum malaria have recently been licensed.79 Although a major achievement, challenges remain, including sub-optimal efficacy and durability of protection across diverse parasite strains. Furthermore, both vaccines target a small portion of a single protein expressed in the pre-erythrocytic sporozoite stage of the parasite life cycle.7,9,10 More consideration is now being given to multistage vaccines, encompassing combinations of proteins from the pre-erythrocytic, erythrocytic, and gametocyte stages. This report focuses on the erythrocytic merozoite stage, where progress in vaccine development has been largely unsuccessful, despite the fact that passive transfer studies in humans demonstrated that antibodies against the proteins expressed here were protective.6,11 An understanding of the mechanisms that underpin antibody-dependent protection could guide the prioritization of vaccine candidates and provide immune correlates to accelerate vaccine development.

Studies designed to understand antibody-dependent mechanisms typically focus on Fab-mediated neutralization that blocks the interaction between pathogens and host cell receptors, thereby preventing infection.12,13 For erythrocytic stage vaccines, this is exemplified in the growth inhibition assay (GIA), which measures the ability of antibodies to block merozoite invasion of red blood cells in culture, thus limiting parasite multiplication and growth.14 Although routinely used to evaluate, prioritize, and quantify the efficacy of blood-stage vaccine candidates, the GIA does not reliably predict either naturally acquired or vaccine-induced protection.15 Malaria vaccines inducing high levels of GIA have had limited success in clinical trials in humans.1519

Antibody Fc-dependent effector functions are less well studied but appear to be increasingly important in mediating protection against infectious diseases such as HIV,20 Ebola,21 COVID-19,22 and malaria.23,24 Antibodies inducing Fc-effector function against merozoite-stage parasites are acquired with age and correlated with protection in independent cohort studies.2529 However, single cellular or soluble effectors such as neutrophils,25,30 monocytes,26,27 natural killer (NK) cells,31 or complement28,32 have been analyzed in disparate studies. Neither the breadth nor the relative importance of individual Fc-effector functions have been assessed concurrently in a well-characterized experimental study of naturally acquired immunity (NAI).

We utilized the Controlled Human Malaria Infection in Semi-Immune Kenyan Adults (CHMI-SIKA) study to address these gaps, leveraging its’ distinct advantages over traditional cohort studies. First, the precise timing, strain, and dose of the inoculum are known, hence minimizing misclassification bias.33 Second, the close monitoring of participants in residential facilities enabled early and accurate capture of clinical outcomes, essential for safety,34,35 and further minimizing misclassification bias due to misdiagnoses. Third, time-dependent analyses are more robust since the timing of exposure is known, and the rapidity with which an endpoint is met is directly reflected in in vivo parasite growth. Consequently, the precision around the estimates of the correlates of protection is higher than that observed in cohort studies.36

We analyzed a panel of Fc-mediated functional assays targeting P. falciparum merozoites concurrently in the same study: neutrophil antibody-dependent respiratory burst (ADRB),25 antibody-dependent complement fixation (AbC’),28 antibody-mediated NK cell activation (Ab-NK),31 and opsonic phagocytosis of merozoites (M_OPA)27 and ring-stage parasites (R_OPA).37 We also tested the GIA38 and asked which assay best predicted the primary clinical outcome, defined as the need for anti-malarial treatment post-challenge.35 We found that Fc-mediated effector functions were correlated with protection from clinical symptoms of malaria and exponential parasite multiplication, while the gold standard GIA was not. The breadth of Fc-mediated effector function discriminated clinical immunity following challenge.

Results

Controlled human challenge provides clear endpoints for clinical immunity

One hundred and forty-two volunteers were closely monitored in residential facilities for 21 days post-challenge and eligible for analysis (Figure 1A; Table S1). However, a retrospective analysis of the blood samples collected a day before challenge (C-1) revealed the presence of the anti-malarial drugs lumefantrine or sulfadoxine (N = 78).35 Although these were below the standard minimum inhibitory concentrations,39 we excluded all such participants from further analysis to minimize potential confounding. Of the remaining volunteers (N = 64), 28 met the treatment criteria defined as parasitaemia > 500/μL or the presence of fever (>37.5°C) and any parasitaemia. These volunteers are hereafter referred to as non-immune (NI, Figure 1B). The remaining 36 volunteers did not meet the treatment criteria (parasitaemia < 500/μL and no fever) and are hereafter referred to as clinically immune (CI, Figure 1C). This reduced sample size precluded additional subgroup analyses.35

Figure 1. Controlled human challenge provides clear endpoints for clinical immunity.

Figure 1

(A) Study design. Kenyan adult volunteers (N = 64) were infected with 3,200 live P. falciparum sporozoites via direct venous injection. Parasitaemia was quantified by qPCR from day 7 to 21.

(B) NI volunteers; parasitaemia > 500/μL of blood and/or developed fever (≥37.5°C) with any parasitaemia, n = 28.

(C) CI volunteers; remained afebrile with parasitaemia < 500/μL, n = 36. The dotted line indicates the treatment threshold. See also Table S1.

Clinical immunity is associated with cytophilic anti-merozoite antibodies

We measured immunoglobulin (Ig)G, IgM, and IgG subclasses against merozoites by enzyme-linked immunosorbent assays (ELISA) in plasma samples collected 1 day before challenge (C-1). Samples were considered positive if the optical density (OD) was greater than the mean plus 3 standard deviations of malaria-naive sera.40,41 Antibody prevalence was defined as the proportion of all test samples that were positive. ELISAs were conducted in duplicate at a single dilution.42,43

Total IgG and IgM antibodies against merozoites were equally abundant in all test samples, with a prevalence of 60.9% and 70.3%, respectively (Figure 2A). Consistent with previous studies, anti-merozoite IgG subclass antibodies were predominantly cytophilic IgG1 (70.3%) and IgG3 (54.7%) (Figure 2A). Across all the isotypes measured, the antibody prevalence against merozoites was lower in the NI compared with the CI (Figure 2B). Similarly, the quantity of anti-merozoite antibodies was lower in the NI compared with the CI (p < 0.0001, Figure 2C). Neither the prevalence nor the quantity of total IgG to tetanus toxoid differed between groups (p = 0.21).

Figure 2. Cytophilic antibodies and Fc functions targeting merozoites are correlated with clinical immunity.

Figure 2

(A) The overall prevalence of IgG, IgM, and IgG1−4 antibodies against merozoites measured by ELISA in plasma samples collected 1 day before the challenge (C-1).

(B) The prevalence of IgG, IgM, and IgG1−4 against merozoites and total IgG against tetanus toxoid extract measured by ELISA in C-1 plasma samples.

(C) Quantities of IgG, IgM, and IgG1−4 antibodies against merozoites and total IgG against tetanus toxoid measured by ELISA in C-1 plasma samples. Error bars: median and 95% confidence intervals. Analysis using the Mann-Whitney U test. Seropositivity cutoff: dotted black horizontal line.

(D) A comparison of plasma GIA (neutralization) and Fc-mediated effector functions in NI and CI. GIA and Fc-mediated activity measured using functional assays in C-1 plasma samples. Error bars: median and 95% confidence intervals. Analysis using the Mann-Whitney U test. Seropositivity cutoff: dotted black horizontal line. RLU, relative light units; RPI, relative phagocytosis index.

(E) ROC curves for effector functions and area under the curve (AUC).

(F) Forest plot showing the adjusted hazard ratios for each effector function using the Cox regression hazard model, comparing the time to treatment between volunteers with high versus low Fc-mediated function. Error bars: 95% confidence intervals. The red line indicates no protection (hazard ratio = 1.0).

(G) Heatmap showing Spearman’s correlation matrix for antibody effector functions targeting merozoites and IgG, IgG subtypes (1−4), and IgM binding to merozoites (M). The color intensity indicates the strength of the correlation, while numbers indicate pairwise Spearman’s rho.

See also Table S2.

Fc-mediated effector function is superior to neutralization in discriminating clinical immunity

We compared the ability of the GIA versus a panel of discrete Fcmediated assays to distinguish between NI and CI volunteers in C-1 samples. For the GIA, we cultured parasites of the NF54 strain used in the CHMI-SIKA study for 96 h (two cycles of parasite replication) in the presence of dialyzed and heat-inactivated plasma.38 For Fc-effector function, we measured ADRB, AbC’, Ab-NK, M_OPA, and R_OPA. The Ab-NK assay had two readouts: degranulation (the proportion of NK cells that were cluster of differentiation (CD) 107a+, AB-NK_CD107a) and interferon gamma (IFNγ) production (the proportion of NK cells that were IFNγ+, AB-NK_ IFNγ).31

There was no difference in GIA between the NI and CI, but all Fc-mediated effector functions were lower in the NI compared with CI group (p < 0.0001, Figure 2D). We tested this further and compared the accuracy of each effector function in predicting the clinical outcome using a receiver operating characteristic (ROC) curves analysis. Fc-mediated functions were superior to the GIA in discriminating clinical immunity, with the area under curve (AUC) between 0.74−0.94 versus 0.62, respectively (Figure 2E).

We validated our results in a Cox proportional hazards survival regression model. We dichotomized each functional assay measurement into a high versus low category using statistically derived thresholds.40,44 For each assay, we compared the time to treatment between volunteers with high versus low antibodies. Each Fc-mediated effector function was associated with a longer time to treatment (hazard ratios [HRs] between 0.05 to 0.19, p < 0.001, Figure 2F, lower panel). By contrast, the GIA was not associated with a longer time to treatment (HR 0.53, 95% CI [0.25−1.12], p = 0.095). The effect size for individual Fc-mediated effector functions was comparable, with an overlap between the respective confidence intervals. Gender did not modify the effect of Fc function on outcome (Table S2).

Correlation analyses support Fc-mediated effector function over antibody binding as the key correlate of clinical immunity

Total IgG, IgM, and IgG subclass antibodies against merozoites correlated with clinical immunity (Figure 2F, upper panel). We reasoned that this was likely due to correlation between these antibodies. We found high and statistically significant pairwise correlations between total IgG, IgG1, and IgG3 (Spearman’s rho 0.93 and 0.91, respectively, p < 0.0001, Figure 2G). These correlations remained significant but were lower for IgG2 and IgG4 (Spearman’s rho 0.70 and 0.62, respectively, p < 0.0001).

Unlike the non-cytophilic IgG isotypes, cytophilic IgG1 and IgG3 isotypes are known to promote binding to Fcγ receptors.45 We compared the pairwise correlation coefficients between IgG1-4 and Fc-mediated functional assays. We found these correlations to be lower for the non-cytophilic isotypes (Spearman’s rho = 0.4−0.6) and higher for the cytophilic isotypes (Spearman’s rho = 0.6−0.8, Figure 2F). These data support the notion that it is Fc function and not merely antibody binding that is responsible for the clinical outcome. Furthermore, for GIA, immune effector cells are excluded, complement is deactivated through heat treatment, and plasma is dialyzed to exclude residual anti-malarial drugs using established protocols.15 GIA therefore only tests antibody binding and its effect on parasite growth. As shown in Figures 2G and 2F, GIA was neither associated with clinical immunity nor correlated with Fc function (Spearman’s rho < 0.25, p > 0.05), supporting the conclusion that Fc function rather than antibody binding per se was responsible for clinical immunity.

Next, we asked whether the antibody effector functions were correlated and found this to be true for ADRB, M_OPA, R_OPA, and AbC’ (Spearman’s rho > 0.70, p < 0.0001, Figure 2F). Ab-NK_CD107a and Ab-NK_IFNγ production were positively correlated (Spearman’s rho = 0.88, p < 0.0001). However, Ab-NK activity correlated least with other Fc-mediated effector functions (Spearman’s rho 0.28−0.57, p < 0.0001), suggesting the engagement of different Fcγ receptors.45 Overall, these data suggest that antibodies inducing Fc-mediated functions are co-acquired and may be in part polyfunctional.

Low parasite densities in vivo are a marker of clinical immunity, particularly in endemic areas with a high intensity of malaria transmission.46 We therefore tested whether Fc-mediated effector functions were more negatively correlated with parasitaemia than neutralization post-challenge. We found a weak correlation between GIA and either the mean or maximum parasite densities (Table 1). By contrast, there was a strong negative correlation between Fc-mediated effector functions with the mean and maximum parasitaemia.

Table 1. Correlations between parasitaemia and Fc-mediated effector functions.

Secondary outcome variable Mean parasitaemia Maximum parasitaemia
Rho p 95% confidence interval Rho p 95% confidence interval
GIA −0.19 0.128 −0.42, 0.06 −0.27 0.031 −0.49, −0.42
ADRB −0.73 <0.0001 −0.82, −0.58 −0.68 <0.0001 −0.79, −0.50
AbC’ −0.62 <0.0001 −0.75, −0.43 −0.59 <0.0001 −0.73, −0.40
M_OPA −0.63 <0.0001 −0.76, −0.44 −0.61 <0.0001 −0.74, −0.42
R_OPA −0.52 <0.0001 −0.68, −0.31 −0.50 <0.0001 −0.67, −0.28
Ab-NK_CD107a −0.41 0.0008 −0.59, −0.17 −0.28 0.023 −0.50, −0.03
Ab-NK_IFNγ −0.30 0.016 −0.51, −0.05 −0.16 0.199 −0.39, 0.09

The breadth of Fc-mediated effector function discriminates clinically immune from non-immune volunteers and identifies an intermediate category

We undertook a principal-component analysis (PCA) of the six Fc-mediated effector functions to visualize how well they discriminated clinical immunity. This initial analysis did not completely discriminate NI from CI volunteers (Figures 3A and S1). The breadth of the antibody response has previously been shown to be an important correlate of protection.27,41,4749 We hypothesized that this failure of the PCA to discriminate CI was due to variation in the breadth of Fc function. To test this, we created a breadth score defined as the sum of responses to the six Fc-mediated functions and compared this between NIs and CIs. The full range of each immunological measure was categorized into tertiles, with corresponding scores of 1 to 4 from the lowest to the highest. The maximum score across the six Fc functions was thus 24. In keeping with our hypothesis, most of the NI volunteers (19/28, 67.8%) had a breadth score of <11, compared with CI volunteers in whom a majority (25/36, 69.4%) had a breadth score of ≥ 18 (Figure 3B). Four of thirty-six (4/36, 11.1%) CI volunteers that had breadth scores in the lowest category of between 6 and 11, while a further 7/36 (19.4%) had intermediate scores between 12 and 17. By contrast, none of the twenty-eight NI volunteers had a breadth score of ≥ 18, while 9/28 (32.1%) had intermediate scores (Figure 3B). This intermediate category was more easily visualized in a repeat PCA using the breadth score and suggests a category of individuals that at least for the NIs, are actively in the process of acquiring immunity (Figures 3C and S1). A high area under the curve of 0.94 in an ROC analysis supported the accuracy of the breadth score in predicting clinical immunity (Figure 3D).

Figure 3. The breadth of Fc-mediated function is correlated with clinical immunity.

Figure 3

(A) PCA for the six Fc-mediated effector functions with colors representing clinical outcome.

(B) The distribution of breadth scores within CI versus NI.

(C) PCA for the six Fc-mediated effector functions with colors representing breadth scores.

(D) ROC curve for the breadth score with AUC.

(E) Survival analysis using the breadth score, showing the percentage of volunteers who were CI over time post-challenge. Each line represents the breadth of Fc function. Analysis using the log-rank (Mantel-Cox) test.

(F) The quantity of anti-merozoite IgG measured in C-1 plasma samples is compared between volunteers with varying Fc breadth scores. Each dot represents an individual. Error bars: median and 95% confidence intervals. Analysis using the Kruskal-Wallis test. Seropositivity cutoff: dotted black horizontal line.

(G) Heatmap of the six Fc-mediated effector functions in NI and CI volunteers. The quantity of Fc function increases from 0 to 1 and is demonstrated by darker shades of red as shown in the scale. Columns represent specific Fc-mediated functions, while rows represent individual volunteers.

See also Figure S1.

The breadth of Fc-mediated effector function predicts the time to clinical malaria

We tested the breadth score categories in Kaplan-Meier survival curve analysis. None of the volunteers with a breadth score of ≥ 18 developed clinical malaria during the 21 days of follow up (n = 25, 100% protection, Figure 3E). By marked contrast, only a proportion of volunteers with a breadth score of either 12−17 (7/16, 44%) or 6−11 (4/23, 17%) (Figure 3E) did not develop malaria. Thus, there was a significant stepwise increase in the proportion of people protected against clinical malaria, with increasing breadth from 17% to 44% to 100% (Figure 3E). Similarly, the time to clinical malaria was shorter in the group with a breadth score of 6−11, compared with 12−17 (p < 0.0001), and the breadth of Fc function increased with rising quantities of anti-merozoite IgG (Figure 3F), further supporting the idea that antibody binding to the merozoite was directly related to both the intensity (captured in breadth scores) and diversity (different types of Fc function, also captured in the breadth score) of Fc-effector function induced.

We explored these data further by generating a heatmap to visualize the breadth of Fc function across NI and CI individuals. It was immediately obvious that most NI individuals had large gaps in their Fc-function repertoire and consequently low breadth scores. Notably, a proportion had intermediate breadth scores, typically developed malaria toward the tail end of the study, and potentially could represent individuals actively in the process of acquiring immunity (Figures 3E and 3G). By contrast, although most of the CI volunteers had a breadth score of ≥ 18, a proportion did not, implying that additional factors contribute to immunity.

Discussion

Passive transfer studies demonstrated the importance of antibodies in mediating protection against malaria, but the precise mechanisms that underpin immunity remain unclear. We found that individual Fc mechanisms were strongly associated with protection and superior to the well-established GIA in discriminating CI versus NI volunteers. A high breadth score of ≥ 18 that was comprised only of Fc mechanisms was completely predictive of protection from clinical malaria. Our ability to control the exposure (challenge agent) and to accurately discern who was protected versus susceptible in the CHMI-SIKA study was far higher than is typically possible in cohort studies of natural malaria infections. We consider this to be a driving factor for the compelling point estimates and narrow confidence intervals we observed in analyses of correlates of protection.

Our data compare well with those from human challenge trials following vaccination with the RTS,S malaria vaccine. Although these were conducted in malaria-naive volunteers and the analysis focused the repeat region of circumsporozoite protein (CSP) (NANP6), univariate analyses from multi-functional antibody profiling identified shared signatures of protection across independent studies, two of which were measures of Fc-mediated function: binding to FcγR3A and antibody-dependent cellular phagocytosis (ADCP).50 In separate studies, Fc-mediated mechanisms of immunity targeting sporozoites and involving both neutrophils and NK cells were shown to be important, particularly through FcγR3A.51 Other studies targeting additional stages of the malaria parasite have also found an important role for Fc-mediated mechanisms of immunity.52 Notably, measures of Fc function are often classified as “non-neutralizing,” even though neutralizing antibodies could also mediate Fc functions, and activity across both the Fc and Fab domains could well be synergistic.53 For example, invasion inhibition by anti-merozoite antibodies was increased in the presence of NK cells31 and complement.28 Similarly, neutralizing antibodies against Ebola21 influenza virus,54 HIV,55,56 and Hanta virus57 were more potent in vivo when they could also induce Fc-activity.

We studied a selection of Fc-mediated effector functions across three major immune cell types and C1q. A recent study found that the predominant cell types mediating phagocytosis varied depending on whether the abundance of antibody against merozoites was low or high.58 Notably, we used either NK cells and neutrophils from malaria-naive donors or the THP1 monocyte cell line and not the autologous cells. In Uganda, high frequencies of atypical CD56 NK cells were expanded following repeated exposure to malaria and associated with clinical immunity.59 In addition, the expression of FcγRs on autologous leukocytes and consequently their responsiveness to IgG-mediated signaling can be modulated by inflammatory cues and differentiation status,60 adding a layer of complexity to data interpretation. In a similar vein, although our selection of immune cells captured the main activating and inhibitory FcγRs (Figures 1, 2A−2C, 3A, and 3B), polymorphisms in their autologous counterparts may also contribute to protection.61 Polymorphisms in FcγR2A (H131R) and FcγR3A (V158F) are thought to have the highest functional significance,62 followed by FcγR2B (I232T) and FcγR2C (Q77X). Our PCA distinguished Ab_NK (FcγR3A) from that mediated by neutrophils and monocyte responses (FcγRs Figures 1, 2A, 2B, 3B, and S1), highlighting the importance of these signaling pathways. Nevertheless, in the work presented here, our priority was to test our hypotheses by keeping the source of the immune cells constant and asking whether the variability in the quantity and quality of antibodies in the test samples was correlated with protection.

We used the NF54 parasite strain that was isolated from one of the CHMI volunteers to minimize the possibility that differences between in vitro GIA activity and in vivo parasite growth were due to the parasite strain.6,10,63 However, GIA was only very weakly correlated with parasitemia and was not associated with protection post-challenge. In vitro GIA activity was correlated with in vivo parasite growth in Aotus monkeys immunized with PfRH5.16 In a recent phase 1 study, vaccination with PfRH5 induced antibodies with GIA activity that suggest they may be associated with clinical protection in young infants and children.64 Efficacy studies are eagerly awaited.

Given that our CHMI setup involved the administration of sporozoites, it remains possible that pre-erythrocytic immune mechanisms contributed to the clinical outcomes. Three distinct pieces of evidence lead us to believe that the primary mechanism of immunity is predominantly erythrocytic. First, although complete pre-erythrocytic immunity would result in sterile immunity, patent parasitaemia was observed in most volunteers.

Additional investigations in independent laboratories to ascertain the PCR-negative status of the remaining participants yielded equivocal results, suggesting these were potentially positive at the margin of detection.35 Thus, even in those that were apparently PCR-negative, parasites were most likely being suppressed at the blood stage. Second, alanine transferase was elevated as expected at approximately 10 days post-challenge and not different in PCR-positive versus PCR-negative volunteers, suggesting that the latter had very low parasitemia.35 Lastly, we considered the fact that sporozoite immunity could lower the blood-stage inoculum and thus contribute to our results. If indeed blood-stage immunity did not have a strong role, parasitaemias would have increased toward the tail end of the study.

Limitations of the study

For safety reasons, study volunteers were treated when the parasitemia reached a predefined threshold, whether they became ill or not, and some may have been misclassified as susceptible. Notably, their breadth scores were <18, suggesting that they were not yet CI. We excluded volunteers with sickle cell trait as this is known to confer protection against malaria, but other undetermined genetic factors may be important. However, most genetic variants have a stronger impact on severe compared with mild malaria or parasitemia.65 Finally, antibody responses to antigens expressed on the surface of infected red blood cells66 or sporozoites, as well as T cell responses, Fc-receptor polymorphisms,6769 and donor-specific differences in effector cell phenotypes and FcγR expression13,70 may have confounded our analyses and need further study.

Star ⋆ Methods

Detailed methods are provided in the online version of this paper and include the following:

  • KEY RESOURCES TABLE

  • RESOURCE AVAILABILITY

    • Lead contact

    • Materials availability

    • Data and code availability

  • EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS

    • CHMI-SIKA study

    • Cell lines

  • METHOD DETAILS

    • Quantitative Polymerase Chain Reaction (qPCR)

    • Merozoite isolation

    • Anti-Merozoite ELISAs

    • Tetanus toxoid ELISA

    • Total IgG sandwich ELISAs

    • Total IgM sandwich ELISAs

    • Growth inhibition activity (GIA)

    • Antibody dependent complement fixation (AbC’)

    • Opsonic phagocytosis activity of merozoites

    • Phagocytosis of ring stage parasites

    • Antibody dependent respiratory burst (ADRB)

    • Antibody-mediated natural killer cells (Ab-NK) assay

  • QUANTIFICATION AND STATISTICAL ANALYSIS

Star ⋆ Methods

Key Resources Table

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
Donkey anti-human IgG-Fcγ fragment specific Alexafluor647 antibody Jackson Immuno Research Cat# 709-605-098
RRID: AB_2340577
Goat anti-human IgM − Alkaline phosphatase conjugated Southern Biotech Cat#2020-04
RRID: AB_2795602
Goat anti-human IgM HRP conjugate antibody Thermo Fischer Scientific Cat#31415
AB_228282
Mouse anti-human CD107a-PE antibody BD Biosciences Cat# 555801
RRID: AB_396135
Mouse anti-human CD16-APC-Cy7 antibody BD Biosciences Cat#561726
RRID AB_396864
Mouse anti-human CD3-PE-Cy5 antibody BD Biosciences Cat#555334
RRID: AB_395741
Mouse anti-human CD56-APC antibody BD Biosciences Cat# 341025
Mouse anti-human IFNγ-PE-Cy7 antibody BD Biosciences, Cat#557643
RRID:AB_396760
Peroxidase-conjugated anti-human IgG1 antibody The Binding Site GmbH Cat#NK006.OPT
Peroxidase-conjugated anti-human IgG2 antibody The Binding Site GmbH Cat#NK007.OPT
Peroxidase-conjugated anti-human IgG3 antibody The Binding Site GmbH Cat#LK008.OPT
Peroxidase-conjugated anti-human IgG4 antibody The Binding Site GmbH Cat#LK009.OPT
Purified human IgG Sigma-Aldrich Cat#I4506
Purified human IgM Sigma-Aldrich Cat#I8260
Rabbit anti-human IgG HRP antibody Agilent Cat#P021402-5
EAN 05700573006786
Sheep anti-human C1q, HRP conjugate antibody Abcam Cat#ab46191
Tetanus toxoid hyperimmune serum pool from Kenyan adults KEMRI-Wellcome Trust Research Programme, Nduati et al.71 N/A
Goat anti-human IgM Southern Biotech Cat#2020-05
RRID: AB_2795603
Rabbit anti-human IgG Southern Biotech Cat#6145-01
RRID:AB_2796200
Biological samples
Human plasma from German adults Bloodbank Heidelberg N/A
Human plasma from Kenyan Adults KEMRI-Wellcome Trust Research Programme, Nduati et al.71 N/A
Purified human IgG from Malawian adults (MIG) KEMRI-Wellcome Trust Research Programme, Taylor et al.72 N/A
Chemicals, peptides and recombinant proteins
AlbuMAXTM I Thermo Fischer Scientific Cat# 11021029
Bovine Serum Albumin (BSA) Sigma-Aldrich Cat#A7030
Brefeldin A (BFA) Sigma-Aldrich Cat # B7651
Carbonate-bicarbonate capsules Sigma-Aldrich Cat# C3041
Casein in PBS (1% w/v) Thermo Fischer Scientific Cat# 37532
Dihydroethidium (DHE) dye Abcam Limited, Cambridge Cat#ab145360
Ethidium bromide (EtBr) dye Thermo Fischer Scientific Cat#15585011
SYBR green nucleic acid dye Thermo Fischer Scientific Cat# S7563
Trypan blue Carl Roth, Karlsruhe Cat# 1680.1
Circumsporozoite protein (CSP) peptide (NANP)6C Pepscan, Germany N/A
Complement C1q, Human Calbiochem Cat#204876-1MG
CountBrightTM Absolute Counting Beads Thermo Fisher Scientific Cat# C36950
D-Sorbitol Sigma-Aldrich Cat#S1876
Fetal Bovine Serum, heat inactivated Thermo Fisher Scientific Cat# A3840001
Paraformaldehyde (PFA) AppliChem Cat# 211511
Isoluminol Santa Cruz Biotechnology sc-485868
RPMI 1640-Medium (Gibco) Thermo Fischer Scientific Cat#11875093
SIGMA FAST o-Phenylenediamine dihydrochloride (OPD) tablets Sigma-Aldrich Cat# P9187
Tetanus toxoid TE-3 NIBSC, UK Cat#02/232
Trans-epoxysuccinyl-L-leucylamido
(4-guanidino) butane (E64)
Sigma Aldrich Cat#E3132
Experimental models: Cells and Organisms
Human O+ erythrocytes Blood bank for Heidelberg University hospital N/A
Plasmodium falciparum 3D7 strain KEMRI-Wellcome Trust
Research Programme
N/A
Plasmodium falciparum NF54 strain KEMRI-Wellcome Trust
Research Programme
N/A
THP1 cell line ATCC, Osier et al.27 RRID: CVCL_0006
Software and Algorithms
GraphPad Prism 9 GraphPad Software, Inc. RRID: SCR_002798
STATA16 StataCorp LLC. 2019 https://www.stata.com/
R Studio v 4.2.3 R Project for Statistical Computing https://www.R-project.org/
Flow Jo V10.9.0 BD Bioscience RRID: SCR_008520

Resource Availability

Lead contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by lead contact, Prof. Faith Osier (f.osier@imperial.ac.uk)

Materials availability

This study did not generate new unique reagents.

Experimental Model And Study Participant Details

Chmi-Sika Study

All study volunteers were challenged under identical circumstances, in the same physical facilities, with a single identical strain, at a standardized dose, and using standardized procedures as detailed in the published study protocol.34 The study was open, non-blind and non-randomized. Notably, although the study subjects were all Kenyan and of similar ethnicity, they were deliberately recruited from different areas of the country to provide a diversity of previous exposures to malaria. We excluded participants with sickle cell trait as this is known to influence the outcome of malaria infections. The study was conducted in 3 phases between 2016 and 2018. One hundred and forty-two (142) participants were infected with Sanaria® 3,200 aseptic, purified, cryopreserved NF54 sporozoites by direct venous injection (DVI). Blood stage parasitemia was monitored by quantitative polymerase chain reaction (qPCR) twice a day from day 7 to day 14, and then once daily from day 15 to day 21 post challenge.35 The study endpoint was met when parasitaemia exceeded 500 parasites/μl of blood, or signs and symptoms of malaria developed in the presence of any parasitaemia, or at 21 days post-challenge when the study ended.35 Thus, our primary outcome was the need for treatment before day 21 and volunteers meeting this criterion are referred to as non-immune (NI), while those that did not are considered clinically immune (CI). We also analysed our immunological outputs against a secondary outcome of parasite density. The mean parasite density was calculated as a geometric mean of the parasite density observed between days 8.5 and day 22 post-challenge and excludes any time points after treatment. The maximum parasite density was the highest parasite density observed between the same period and similarly excluding any time points after treatment. All immunological assays presented here were conducted in plasma samples collected the day before challenge (C-1).

The CHMI-SIKA study was conducted at the Kenya Medical Research Institute (KEMRI)-Wellcome Trust Research Programme in Kilifi, Kenya, with ethical approval from the KEMRI Scientific and Ethics Review Unit (KEMRI//SERU/CGMR-C/029/3190) and by the University of Oxford Tropical Research Ethics Committee (OxTREC 2-16). All participants gave written informed consent. The study was registered on ClinicalTrials.gov (NCT02739763), conducted based on good clinical practice (GCP), and under the principles of the Declaration of Helsinki.

Cell lines

The human monocyte cell line, (THP-1, sourced from ATCC), was cultured in RPMI 1640 medium supplemented with 2mM L-glutamine, 10 mM HEPES, 1% pen strep (10,000 units/ml penicillin and 10,000 μg/ml streptomycin) and 10% foetal bovine serum (FBS) in a humidified incubator with 5% CO2 at 37°C as previously described.27 The cell density was monitored closely and maintained between 1 ×105 and 1 ×106 cells/ml. Cells were passaged when the cell density approached 1 ×106 cells/ml.

Method Details

Quantitative Polymerase Chain Reaction (qPCR)

The qPCR assay is published.35 Briefly, DNA was extracted from 500μl of whole blood and eluted in 100μl. An aliquot of 13.5μl of the DNA was used for qPCR using a TaqMan® probe (5′-FAM-AACAATTGGAGGGCAAG-NFQ-MGB-3′) that targets the multicopy 18S ribosomal RNA gene. Six serial dilutions of DNA extracted from an in vitro parasite culture of known parasitemia were included as standards and used to determine the parasitemia of the samples.

Merozoite isolation

Plasmodium falciparum merozoites of the NF54 or 3D7 strains were isolated as previously described.73 Parasites were thawed and cultured to obtain trophozoites at high parasitemia (8-12%). The trophozoites were isolated by magnetic purification and cultured in fresh complete medium to allow development to early schizont stage. A protease inhibitor, trans-epoxysuccinyl-L-leucylamido (4-guanidino) butane (E64, Sigma Aldrich), was added to allow maturation of the schizonts without rapture.73 Merozoites were harvested by filtration of the mature schizonts through 1.2μm filters (Pall). The merozoites were stained using either 1μg/ml ethidium bromide (EtBr, Thermo Fischer) or with 1X SYBR green dye (Thermo Fischer) for 30min and counted against CountBright™ Absolute Counting Beads (Thermo Fischer) using BD FACS Canto II flow cytometer.

Anti-Merozoite ELISAs

Enzyme Linked Immunosorbent Assays (ELISAs) for merozoite antigens are well-established using a standardized protocol.27,28,40,41 Briefly, 96-well plates (Thermo Fischer) were coated with 100μl/well of NF54 merozoites at 5x106 merozoites/ml at 4°C overnight. The plates were washed and blocked with 200μl/well of blocking buffer, 1% casein (Thermo Fischer), for 2 hours at 37°C. Samples were diluted at a single dilution (1:500) with blocking buffer, added at 200μl/well and incubated for 1 hour at 37°C. Plates were washed before the addition of 100μl/well of the respective Horseradish peroxidase (HRP)-conjugated secondary antibodies: rabbit anti-human IgG (Dako), goat anti-human IgM (Southern Biotech) and rabbit anti-human IgG1, 2, 3 or 4 (The Binding Site GmbH), and incubation for 1 hour at 37°C. The plates were then washed four times and 100μl/well of substrate (o-phenylenediamine dihydrochloride (OPD), Sigma-Aldrich) was added before incubation for 20 min in the dark at room temperature (RT). The reaction was stopped with 30μl of 1M Hydrochloric acid (HCl, Sigma-Aldrich) and the absorbance measured at 492nM using a BioTek Cytation 3 cell imaging multi-mode reader. Each ELISA plate contained two sets of positive and negative controls. The positive controls were a pool of hyperimmune serum from Kenyan adults40 (PHIS) and a pool of purified IgG from malaria exposed adults from Malawi72 (MIG). The negative controls were plasma samples from malaria naïve German adults and blank wells. All assays were conducted in duplicate and the final data presented for each individual represents the mean of duplicates. Samples were re-tested afresh and in duplicate if the coefficient of variation between duplicate results exceeded 20%. The cut-off for seropositivity was defined as the mean plus 3 standard deviations of the malaria-naïve negative controls and used to estimate the antibody prevalence.

Tetanus toxoid ELISA

The protocol for tetanus toxoid ELISA was like the merozoite ELISA protocol above with a few variations. Briefly, plates were coated with 2μg/ml of tetanus toxoid (NIBSC), diluted in carbonate-bicarbonate buffer. Plasma samples were tested in duplicate at a 1:1000 dilution. Antibodies that bound were detected with rabbit anti-human IgG conjugated HRP (Dako). The substrate (OPD) was added and incubated for 15 min in the dark at room temperature. The reaction was stopped with 30μl of 1M Hydrochloric acid (HCL) and the absorbance measured at 492nM. Positive controls on each plate included Tetanus Immunoglublin TE-3 (National Institute for Biological Standards and Controls, UK Official Medicines Control Laboratory) and a tetanus hyperimmune serum pool from recently immunized adults from Kilifi, Kenya.71 Phosphate buffered saline (PBS) and blank wells were used as negative controls on each plate. The final data presented for everyone represents the mean of duplicates. Samples were re-tested afresh and in duplicate if the coefficient of variation between duplicate results exceeded 20%.

Total IgG sandwich ELISAs

Sandwich ELISAs to detect IgG were conducted as previously published.71,74 Briefly, ELISA plates (Thermo Fischer) were coated overnight at 4°C with 10μg/ml of unlabelled rabbit anti-human IgG (Southern Biotech) diluted in bicarbonate buffer. The plates were blocked with 200μl of blocking buffer (3% skimmed milk) for 1 hour at RT. Plasma samples were diluted to 1:500,000 for total IgG and incubated for 2 hours in a humidified chamber at room temperature. Antibodies that bound were detected with an HRP-conjugated rabbit anti-human IgG (Dako). The OPD substrate was added and incubated for 5-15 min in the dark at room temperature before stopping the reaction using 30μl of 1M Hydrochloric acid (HCl). The absorbance measured at 492nM. All assays were conducted in duplicate. The positive control was purified human IgG (Sigma-Aldrich), while the negative control was purified human IgM (Sigma-Aldrich). The final data presented for each individual represents the mean of duplicates. Samples were re-tested afresh and in duplicate if the coefficient of variation between duplicate results exceeded 20%.

Total IgM sandwich ELISAs

Sandwich ELISAs to detect IgM were similar in principle to those for IgG with minor variations.71 Briefly, ELISA plates (Thermo Fischer) were coated overnight at 4°C with 2.5μg/ml of unlabelled goat anti-human IgM (Southern Biotech) diluted in bicarbonate buffer. The plates were blocked with 200μl of blocking buffer (3% skimmed milk) for 1 hour at RT. Plasma samples were diluted at 1:5000. Antibodies that bound were detected using an alkaline phosphatase-conjugated goat anti-human IgM (Southern Biotech). The substrate p-Nitrophenyl Phosphate and disodium Salt (PNPP) was added and incubated for 5-15 min in the dark at room temperature. The reaction was stopped with 50 μl of 3 N NaOH and absorbance measured at 405nm. All assays were conducted in duplicate. The positive control was purified human IgM (Sigma-Aldrich), while the negative control was purified human IgG (SigmaAldrich). The final data presented for everyone represents the mean of duplicates. Samples were re-tested afresh and in duplicate if the coefficient of variation between duplicate results exceeded 20%.

Growth inhibition activity (GIA)

Plasma samples were dialysed using 10kDa columns (Merck Millipore GmbH) to remove any residual antimalarial drugs, and subsequently heat-treated at 56°C for 30min to inactivate complement proteins. The samples were transferred into U-bottomed 96-well plates (Thermo Fischer) at 5μl/well. Control wells contained medium only and were used as the reference to estimate inhibition as described below. Tightly synchronized trophozoite stage parasites at 0.5% and 1% haematocrit were added into each well at a volume of 45μl/well. The parasites were incubated for 96 hours to capture two cycles of parasite growth. Ten μl of fresh medium was added to each well at 48 hours. After 96 hours, the parasites were stained with 1x SYBR Green, washed and fixed in 200μl of 2% paraformaldehyde (PFA, AppliChem). The parasitemia in each well was then measured by flow cytometry using the high throughput sampler (HTS) of the BD FACS Canto II™ flow cytometer. The FACs data was analysed in FlowJo version 10 software. Percentage parasitemia was determined as the percentage of SYBR green positive erythrocytes. Percentage inhibition was calculated as: 100 (parasitemia of test sample / parasitemia of reference x 100). The positive controls were a pool of hyperimmune serum from Kenyan adults40 (PHIS) and a pool of purified IgG from malaria exposed adults from Malawi72 (MIG)) while the negative controls were plasma samples from malaria naïve German adults.

Antibody dependent complement fixation (AbC’)

Complement fixation was measured as previously published.28,32 Briefly, 96-well ELISA plates (Thermo Fischer) were coated with 100μl of freshly isolated merozoites at 5×106 merozoites/ml for 2 hours at 37°C. Plates were washed and blocked with 200μl of 1% Casein in PBS for 2 hours at 37°C. Plasma samples were diluted 1:100 in 0.1% casein in PBS, added to the plates 50μl/well and incubated for 2 hours at 37°C. The plates were then washed and 40μl of human C1q (Calbiochem) was added at 10μg/ml and incubated for 30 min at RT. The bound C1q was detected using HRP-conjugated sheep polyclonal anti-C1q antibody (Abcam) diluted 1:100 for 1 hour at 37°C. The plates were washed and 100μl of OPD substrate was added and incubated for 15 min at RT before the reaction was stopped by the addition of 25μl of 2M H2SO4. Absorbance read at 492nm. Positive and negative controls were included as described for the anti-merozoite ELISAs above.

Opsonic phagocytosis activity of merozoites

The OPA assay is published.27 Briefly, THP-1 cells were cultured and diluted to 6.7×105 cells/ml. 96-well U-bottom plates were precoated with 1% casein in PBS for 20 min. THP-1 cells were transferred into the plates at 150μl/well and placed in an incubator at 37°C. Separately, plasma samples were diluted 1:100 in RPMI and 10μl each was co- incubated with 100μl of ethidium bromide stained merozoites (8×106merozoites/ml) for 40min at room temperature to allow opsonization to occur. For each sample, 50μl of opsonized merozoites were transferred in duplicates into the plates containing THP-1 cells and incubated at 37°C for 10 minutes to allow phagocytosis to occur.

Phagocytosis was stopped by centrifugation at 350 x g for 5 min at 4°C. Cells were washed once with 200μl of ice-cold FACS buffer (PBS + 0.5% BSA + 2mM EDTA) before resuspension in 200μl of ice-cold 2% PFA. Phagocytosis was measured using the high throughput sampler (HTS) on BD FACS Canto II™ flow cytometer. The data was analysed in FlowJo version 10 software. Phagocytosis was determined as the percentage of THP1-cells that were positive for ethidium bromide staining. A pool of hyperimmune serum from Kenyan adults (PHIS) was used as the reference sample. The relative phagocytosis index (RPI) of each sample was calculated as: phagocytosis of test sample / phagocytosis of reference sample (PHIS) X 100.

Phagocytosis of ring stage parasites

The OPA for ring stage parasites is published.37 Briefly, tightly synchronised ring stage parasites (0 10hours post-infection) at 10-15% parasitemia were stained with 5μg/ml dihydroethidium (DHE, Abcam Limited) and μM Cell Trace™ violet dye (Thermo Fischer) in PBS for 30 min at 37°C. The stained ring culture was washed twice using RPMI 1640 medium and transferred into U-bottomed 96-well plates at 0.5μl pellet/well. The ring culture was opsonised with heat-inactivated plasma diluted 1:12.5 for 30 minutes at room temperature. The opsonised cells were washed twice with RPMI 1640 and transferred into plates containing 150μl of THP1 cells (2.0 x 104 cells/well) and incubated for 4 hrs at 37°C. Unphagocytosed erythrocytes were lysed using a red cell lysis buffer. The cells were then fixed with 2% PFA and phagocytosis was measured using the high throughput sampler (HTS) on the BD FACS Canto II™ flow cytometer. Phagocytosis was determined as the percentage of THP1-cells that were positive for ethidium bromide staining. A pool of hyperimmune serum from Kenyan adults (PHIS) was used as the reference sample. Previous studies demonstrated that antibody-mediated OPA was specific to the IgG fraction of malaria-exposed sera in a dose-dependent fashion.27 The relative phagocytosis index (RPI) of each sample was calculated as: phagocytosis of test sample / phagocytosis of reference sample (PHIS) X 100.

Antibody dependent respiratory burst (ADRB)

The ADRB assay is published and involved three major steps; i) parasite preparation, ii) neutrophil isolation, and iii) a chemiluminescence detection method.75

Parasite preparation: Plasmodium falciparum trophozoites of the 3D7 strain were isolated by magnetic purification and cultured in fresh complete medium to allow development to the early schizont stage. A protease inhibitor E64 (Sigma Aldrich) was added to allow the maturation of the trophozoites for 8-12 hours whilst preventing schizont rupture.73 The schizonts were then quantified and stored at -80°C until use. At the time of the assay, schizonts were thawed, diluted to 10 x 105 schizonts/ml in PBS, and coated into white opaque 96-well plates (Greiner) at 100μl/well and incubated overnight at room temperature. The plates were then washed and blocked with 200μl/well casein in PBS for 1 hour at room temperature. The plates were washed before the addition of 50μl of diluted plasma (1:50 in 1X PBS) to each well, followed by a 1-hour incubation at 37°C.

Neutrophil isolation

Polymorphonuclear leucocytes (PMN) were prepared from freshly collected whole blood for each assay using a standard procedure. Forty mls of whole blood from healthy donors was mixed at a ratio of 1:1 with Hank’s Balanced Salt Solution (HBSS, Thermo Fischer). The diluted blood was carefully layered on 7.5ml aliquots of Ficoll (GE healthcare). This was then centrifuged at 600 x g for 15 min. The supernatant was carefully removed without disturbing the red cell pellet. The pellet was resuspended in 5ml of HBSS then mixed with 3% dextran at a ratio of 1:2 and incubated at room temperature in the dark for 30min. The supernatant was then carefully collected and centrifuged at 500 x g for 7min at 4°C. The supernatant was discarded, and red cell contaminants were lysed. The cells were then centrifuged at 500 x g for 7min at 4°C and the pellet resuspended in 1ml of ice cold PMN buffer (HBSS with 0.1% bovine serum albumin (BSA, Sigma-Aldrich), 1% D -glucose). The PMN count was determined using a haemocytometer and the concentration adjusted using the PMN buffer to 1.0 X 107 / ml. The cells were kept on ice.

Chemiluminescence detection

The schizont-coated plates were washed before the addition 50μl of PMNs (1 x 107 PMNs/ml) and 50μl of 0.04mg/ml isoluminol (Santa Cruz Biotechnology) per well. The plates were immediately loaded onto a plate reader and ADRB activity quantified via chemiluminescence at 450nM every two minutes for 1.5 hours, captured as the maximal relative light units (RLU). The positive controls were PHIS and MIG, while the negative controls were malaria-naïve sera from German donors as described under merozoite ELISAs above. To account for the variability of ADRB activity from PMNs from different individuals, each sample tested in duplicate using PMNs from two independent donors.47,76 To further minimize variability between donors, the RLU values were indexed based on the PHIS positive controls in each plate.47,76 Thus, the mean RLU for the PHIS control was used as the reference sample with an indexed RLU of 1.0. The indexed RLU value for each test sample was calculated as: RLU of sample / RLU of PHIS. The mean indexed RLU values using PMNs from two donors was then calculated.

Antibody-mediated natural killer cells (Ab-NK) assay

The antibody-mediated natural killer cells (Ab-NK) assay is published.31

Opsonization of merozoites

Briefly, 96 well culture-plates were coated with 100μl of merozoites in PBS overnight at 4°C. Plates were then washed three times with 200μl of sterile PBS and blocked with casein in PBS for 2 hours at 37°C. Plates were washed three times with sterile PBS after blocking and before the addition of 50μl/well of plasma samples diluted 1:20 in blocking buffer. Incubation proceeded for 4 hours at 37°C and plates were subsequently washed five times with sterile PBS to remove unbound plasma components.

Natural Killer Cell Preparation

Peripheral blood mononuclear cells (PBMCs) were harvested by density gradient centrifugation from fresh blood collected from healthy donors, resuspended in 2 ml of culture medium and the cell density quantified using a hemocytometer. NK cells were enriched from the PBMCs using a NK negative isolation kit (Miltenyi) and magnetic cell sorting using LS columns (Miltenyi) as per the manufacturer’s instructions, and stored at 1X105 cells per ml.

Co-incubation of NK cells and opsonized merozoites

Freshly isolated NK cells (20,000 NK cells/well), 2μl/well of brefeldin A (BFA, Sigma-Aldrich), 2μl/well of monensin (Sigma) and 2.5μ/well of PE conjugated anti-human CD107a antibody (BD Biosciences) were added to the opsonised merozoites and incubated for 18 hours at 4°C. The stimulated NK cells were then transferred into V bottom 96 well plates, centrifuged at 800 x g for 5 min at 4°C and washed with FACS buffer (0.1% Sodium Azide + 1% BSA + 1 x PBS).

Cell staining

The viability of NK cells was assessed using eFluor 520 (ThermoFischer Scientific). NK cell surface receptors were stained using a cocktail of anti-human mAbs comprising anti- CD56 allophycocyanin (APC), anti-CD3 phycoerythrin (PE)−Cy5, and anti-CD16 APC-Cy7 (BD Biosciences) for 30 min at 4°C. The plates were then washed twice with FACS buffer, fixed with Cell Fix (BD Biosciences) for 10 mins, and permeabilized using Perm/Wash (BD Biosciences) for 10 min at 4°C. The permeabilized cells were then stained intracellularly with anti-human IFNɣ-PE-Cy7 antibody (BD biosciences) for one hour in the dark. Finally, the cells were washed with Perm/Wash, resuspended in FACS buffer, and stored at 4°C prior to data acquisition.

Flow cytometry

Data was acquired using a BD Biosciences FACS Calibur II high throughput instrument in a 96-well format using FACSDiva. The flow data gating strategy and analysis are published.31,76The assay positive controls were PHIS and MIG, while the negative controls were malaria-naïve sera from German donors as described under merozoite ELISAs above. The proportion of NK cells expressing IFNɣ and or CD107a (degranulating NK cells) were determined using FlowJo version 10.

Quantification And Statistical Analysis

Statistical tests are indicated in the corresponding figure legends. A significance threshold of p < 0.05 was used for all tests. Continuous variables were compared using the nonparametric Mann-Whitney U test and the Kruskal-Wallis H test with Dunn’s multiple comparisons test (GraphPad Prism) as appropriate. The accuracy of antibody function in predicting clinical outcome was analysed using the area under the curve (AUC) in receiver operating curves (ROC) analyses. The relationships between variables were examined using Spearman’s rank correlations. Time to event analyses were conducted using the Cox proportional hazards regression model in STATA16. Data were visualized using principal component analysis (PCA) tools in Rv4.2.3.

Supplementary Material

Supplementary Information

Highlights.

  • Used a human malaria infection study to investigate functional correlates of immunity

  • IgG Fc-dependent mechanisms are correlated with acquired immunity, but GIA is not

  • The breadth of IgG Fc-effector function is associated with protective immunity

Acknowledgments

We are grateful to all volunteers in the CHMI-SIKA study, the teams in Kilifi, Ahero, Sanaria, Wellcome (grant number 107499), and Oxford University for Sponsorship. Immunology was supported by a Discovery Award (226669/Z/22/Z) from Wellcome, a Sofja Kovalevskaja Award from the Alexander von Humboldt Foundation (3.2—1184811—KEN—SKP), and an EDCTP Senior Fellowship (TMA 2015 SF1001), which is part of the EDCTP2 programme supported by the European Union all awarded to F.H.A.O. F.K.M. was supported by a scholarship from the German Academic Exchange Service (DAAD), Funding Programme 57214224, ST-32-PKZ 91608705. K.M. was supported by an NIHR Global Health Research Unit grant number 16/136/33 and Tackling Infections to Benefit Africa (TIBA). K.M. and R.O. were also supported through the DELTAS Africa Initiative grant no. 107754/Z/15/Z-DELTAS Africa SSACAB and from DELTAS Africa Initiative (DEL-15-003). The DELTAS Africa Initiative is an independent funding scheme of the African Academy of Sciences (AAS)’s Alliance for Accelerating Excellence in Science in Africa (AESA) and supported by the New Partnership for Africa’s Development Planning and Coordinating Agency (NEPAD Agency) with funding from Wellcome (107769/Z/10/Z) and the UK Government. The views expressed in this publication are those of the author(s) and not necessarily those of AAS, NEPAD Agency, Wellcome Trust, or the UK Government.

Footnotes

Author Contributions

Conceptualization, F.H.A.O. and I.N.N.; methodology, I.N.N., D.O., F.M., L.N., L.M., J.T., R.K., K.F., and M.R.; investigation, I.N.N., D.O., F.M., L.N., L.M., J.T., R.K., K.F., M.R., and R.O.; writing − original draft, I.N.N., R.O., and F.H.A.O.; writing − review and editing, R.O., I.N.N., R.F., D.O, F.M., P.N., M.H., M.C.K., and F.H.A.O.; funding acquisition, F.H.A.O.; resources, C.S.S.T.; supervision, F.H.A.O.

Declaration of Interests

In the CHMI-SIKA team, Y.A., P.F.B., S.L.H., E.R.J., B.K.L.S., and T.L.R. are salaried, full-time employees of Sanaria Inc., the manufacturer of Sanaria PfSPZ Challenge. Thus, all authors associated with Sanaria Inc. have potential conflicts of interest.

Contributor Information

CHMI-SIKA study team:

Abdirahman I. Abdi, Yonas Abebe, Philip Bejon, Peter F. Billingsley, Peter C Bull, Zaydah de Laurent, Mainga Hamaluba, Stephen L. Hoffman, Eric R. James, Melissa C. Kapulu, Silvia Kariuki, Domitila Kimani, Rinter Kimathi, Sam Kinyanjui, Cheryl Kivisi, Johnstone Makale, Kevin Marsh, Khadija Said Mohammed, Moses Mosobo, Janet Musembi, Jennifer Musyoki, Michelle Muthui, Jedidah Mwacharo, Kennedy Mwai, Francis Ndungu, Joyce M. Ngoi, Patricia Njuguna, Irene N. Nkumama, Omar Ngoto, Dennis O. Odera, Bernhards Ogutu, Fredrick Olewe, Donwilliams Omuoyo, John Ong’echa, Faith H.A. Osier, Edward Otieno, Jimmy Shangala, Betty Kim Lee Sim, Thomas L. Richie, James Tuju, Juliana Wambua, and Thomas N Williams

Data and code availability

  • All data reported in this paper will be shared by the lead contact upon request.

  • This paper does not report original code.

  • Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

References

  • 1.Phillips MA, Burrows JN, Manyando C, van Huijsduijnen RH, Van Voorhis WC, Wells TNC. Malaria. Nat Rev Dis Primers. 2017;3:17050. doi: 10.1038/nrdp.2017.50. [DOI] [PubMed] [Google Scholar]
  • 2.Cowman AF, Healer J, Marapana D, Marsh K. Malaria: Biology and Disease. Cell. 2016;167:610–624. doi: 10.1016/j.cell.2016.07.055. [DOI] [PubMed] [Google Scholar]
  • 3.Paton RS, Kamau A, Akech S, Agweyu A, Ogero M, Mwandawiro C, Mturi N, Mohammed S, Mpimbaza A, Kariuki S, et al. Malaria infection and severe disease risks in Africa. Science. 2021;373:926–931. doi: 10.1126/science.abj0089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.WHO. World Malaria Report. World Health Organization; 2023. [Google Scholar]
  • 5.Bhatt S, Weiss DJ, Cameron E, Bisanzio D, Mappin B, Dalrymple U, Battle KE, Moyes CL, Henry A, Eckhoff PA, et al. The effect of malaria control on Plasmodium falciparum in Africa between 2000 and 2015. Nature. 2015;526:207–211. doi: 10.1038/nature15535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Draper SJ, Sack BK, King CR, Nielsen CM, Rayner JC, Higgins MK, Long CA, Seder RA. Malaria Vaccines: Recent Advances and New Horizons. Cell Host Microbe. 2018;24:43–56. doi: 10.1016/j.chom.2018.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Moorthy V, Hamel MJ, Smith PG. Malaria vaccines for children: and now there are two. Lancet. 2024;403:504–505. doi: 10.1016/S0140-6736(23)02743-5. [DOI] [PubMed] [Google Scholar]
  • 8.RTS,S Clinical Trials Partnership. Efficacy and safety of RTS,S/AS01 malaria vaccine with or without a booster dose in infants and children in Africa: final results of a phase 3, individually randomised, controlled trial. Lancet. 2015;386:31–45. doi: 10.1016/S0140-6736(15)60721-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Datoo MS, Dicko A, Tinto H, Ouédraogo J-B, Hamaluba M, Olotu A, Beaumont E, Ramos Lopez F, Natama HM, Weston S, et al. Safety and efficacy of malaria vaccine candidate R21/Matrix-M in African children: a multicentre, double-blind, randomised, phase 3 trial. Lancet. 2024;403:533–544. doi: 10.1016/S0140-6736(23)02511-4. [DOI] [PubMed] [Google Scholar]
  • 10.Laurens MB. The Promise of a Malaria Vaccine-Are We Closer? Annu Rev Microbiol. 2018;72:273–292. doi: 10.1146/annurev-micro-090817-062427. [DOI] [PubMed] [Google Scholar]
  • 11.Cohen S, McGregor IA, Carrington S. Gamma-Globulin and Acquired Immunity to Human Malaria. Nature. 1961;192:733–737. doi: 10.1038/192733a0. [DOI] [PubMed] [Google Scholar]
  • 12.Forthal DN. Functions of Antibodies. Microbiol Spectr. 2014;2:1–17. [PMC free article] [PubMed] [Google Scholar]
  • 13.Lu LL, Suscovich TJ, Fortune SM, Alter G. Beyond binding: antibody effector functions in infectious diseases. Nat Rev Immunol. 2018;18:46–61. doi: 10.1038/nri.2017.106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Miura K. Progress and prospects for blood-stage malaria vaccines. Expert Rev Vaccines. 2016;15:765–781. doi: 10.1586/14760584.2016.1141680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Duncan CJA, Hill AVS, Ellis RD. Can growth inhibition assays (GIA) predict blood-stage malaria vaccine efficacy? Hum Vaccin Immunother. 2012;8:706–714. doi: 10.4161/hv.19712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Douglas AD, Baldeviano GC, Lucas CM, Lugo-Roman LA, Crosnier C, Bartholdson SJ, Diouf A, Miura K, Lambert LE, Ventocilla JA, et al. A PfRH5-based vaccine is efficacious against heterologous strain blood-stage Plasmodium falciparum infection in aotus monkeys. Cell Host Microbe. 2015;17:130–139. doi: 10.1016/j.chom.2014.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Douglas AD, Baldeviano GC, Jin J, Miura K, Diouf A, Zenonos ZA, Ventocilla JA, Silk SE, Marshall JM, Alanine DGW, et al. A defined mechanistic correlate of protection against Plasmodium falciparum malaria in non-human primates. Nat Commun. 2019;10:1953. doi: 10.1038/s41467-019-09894-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Payne RO, Milne KH, Elias SC, Edwards NJ, Douglas AD, Brown RE, Silk SE, Biswas S, Miura K, Roberts R, et al. Demonstration of the Blood-Stage Plasmodium falciparum Controlled Human Malaria Infection Model to Assess Efficacy of the P. falciparum Apical Membrane Antigen 1 Vaccine, FMP2.1/AS01. J Infect Dis. 2016;213:1743–1751. doi: 10.1093/infdis/jiw039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Minassian AM, Silk SE, Barrett JR, Nielsen CM, Miura K, Diouf A, Loos C, Fallon JK, Michell AR, White MT, et al. Reduced blood-stage malaria growth and immune correlates in humans following RH5 vaccination. Med. 2021;2:701–719.:e19. doi: 10.1016/j.medj.2021.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Su B, Dispinseri S, Iannone V, Zhang T, Wu H, Carapito R, Bahram S, Scarlatti G, Moog C. Update on Fc-Mediated Antibody Functions Against HIV-1 Beyond Neutralization. Front Immunol. 2019;10:2968. doi: 10.3389/fimmu.2019.02968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Gunn BM, Yu WH, Karim MM, Brannan JM, Herbert AS, Wec AZ, Halfmann PJ, Fusco ML, Schendel SL, Gangavarapu K, et al. A Role for Fc Function in Therapeutic Monoclonal Antibody-Mediated Protection against Ebola Virus. Cell Host Microbe. 2018;24:221–233.:e5. doi: 10.1016/j.chom.2018.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Winkler ES, Gilchuk P, Yu J, Bailey AL, Chen RE, Chong Z, Zost SJ, Jang H, Huang Y, Allen JD, et al. Human neutralizing antibodies against SARS-CoV-2 require intact Fc effector functions for optimal therapeutic protection. Cell. 2021;184:1804–1820.:e16. doi: 10.1016/j.cell.2021.02.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Teo A, Feng G, Brown GV, Beeson JG, Rogerson SJ. Functional Antibodies and Protection against Blood-stage Malaria. Trends Parasitol. 2016;32:887–898. doi: 10.1016/j.pt.2016.07.003. [DOI] [PubMed] [Google Scholar]
  • 24.Moormann AM, Nixon CE, Forconi CS. Immune effector mechanisms in malaria: An update focusing on human immunity. Parasite Immunol. 2019;41:e12628. doi: 10.1111/pim.12628. [DOI] [PubMed] [Google Scholar]
  • 25.Joos C, Marrama L, Polson HEJ, Corre S, Diatta AM, Diouf B, Trape JF, Tall A, Longacre S, Perraut R. Clinical protection from falciparum malaria correlates with neutrophil respiratory bursts induced by merozoites opsonized with human serum antibodies. PLoS One. 2010;5:e9871. doi: 10.1371/journal.pone.0009871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hill DL, Eriksson EM, Li Wai Suen CSN, Chiu CY, Ryg-Cornejo V, Robinson LJ, Siba PM, Mueller I, Hansen DS, Schofield L. Opsonising antibodies to P. falciparum merozoites associated with immunity to clinical malaria. PLoS One. 2013;8:e74627. doi: 10.1371/journal.pone.0074627. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Osier FH, Feng G, Boyle MJ, Langer C, Zhou J, Richards JS, McCallum FJ, Reiling L, Jaworowski A, Anders RF, et al. Opsonic phagocytosis of Plasmodium falciparum merozoites: mechanism in human immunity and a correlate of protection against malaria. BMC Med. 2014;12:108. doi: 10.1186/1741-7015-12-108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Boyle MJ, Reiling L, Feng G, Langer C, Osier FH, Aspeling-Jones H, Cheng YS, Stubbs J, Tetteh KKA, Conway DJ, et al. Human antibodies fix complement to inhibit Plasmodium falciparum invasion of erythrocytes and are associated with protection against malaria. Immunity. 2015;42:580–590. doi: 10.1016/j.immuni.2015.02.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Tiendrebeogo RW, Adu B, Singh SK, Dziegiel MH, Nébié I, Sirima SB, Christiansen M, Dodoo D, Theisen M. Antibody-Dependent Cellular Inhibition Is Associated With Reduced Risk Against Febrile Malaria in a Longitudinal Cohort Study Involving Ghanaian Children. Open Forum Infect Dis. 2015;2:ofv044. doi: 10.1093/ofid/ofv044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Feng G, Wines BD, Kurtovic L, Chan JA, Boeuf P, Mollard V, Cozijnsen A, Drew DR, Center RJ, Marshall DL, et al. Mechanisms and targets of Fcgamma-receptor mediated immunity to malaria sporozoites. Nat Commun. 2021;12:1742. doi: 10.1038/s41467-021-21998-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Odera DO, Tuju J, Mwai K, Nkumama IN, Fürle K, Chege T, Kimathi R, Diehl S, Musasia FK, Rosenkranz M, et al. Anti-merozoite antibodies induce natural killer cell effector function and are associated with immunity against malaria. Sci Transl Med. 2023;15:eabn5993. doi: 10.1126/scitranslmed.abn5993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Reiling L, Boyle MJ, White MT, Wilson DW, Feng G, Weaver R, Opi DH, Persson KEM, Richards JS, Siba PM, et al. Targets of complement-fixing antibodies in protective immunity against malaria in children. Nat Commun. 2019;10:610. doi: 10.1038/s41467-019-08528-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kinyanjui SM, Bejon P, Osier FH, Bull PC, Marsh K. What you see is not what you get: implications of the brevity of antibody responses to malaria antigens and transmission heterogeneity in longitudinal studies of malaria immunity. Malar J. 2009;8:242. doi: 10.1186/1475-2875-8-242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Kapulu MC, Njuguna P, Hamaluba MM, CHMI-SIKA Study Team Controlled Human Malaria Infection in Semi-Immune Kenyan Adults (CHMI-SIKA): a study protocol to investigate in vivo Plasmodium falciparum malaria parasite growth in the context of pre-existing immunity. Wellcome Open Res. 2018;3:155. doi: 10.12688/wellcomeopenres.14909.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kapulu MC, Njuguna P, Hamaluba M, Kimani D, Ngoi JM, Musembi J, Ngoto O, Otieno E, Billingsley PF, Controlled Human Malaria Infection in Semi-Immune Kenyan Adults (CHMI-SIKA) Study Team Safety and PCR monitoring in 161 semi-immune Kenyan adults following controlled human malaria infection. JCI Insight. 2021;6:e146443. doi: 10.1172/jci.insight.146443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kapulu MC, Kimani D, Njuguna P, Hamaluba M, Otieno E, Kimathi R, Tuju J, Sim BKL, CHMI-SIKA Study Team Controlled human malaria infection (CHMI) outcomes in Kenyan adults is associated with prior history of malaria exposure and anti-schizont antibody response. BMC Infect Dis. 2022;22:86. doi: 10.1186/s12879-022-07044-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Musasia FK, Nkumama IN, Frank R, Kipkemboi V, Schneider M, Mwai K, Odera DO, Rosenkranz M, Fürle K, Kimani D, et al. Phagocytosis of Plasmodium falciparum ring-stage parasites predicts protection against malaria. Nat Commun. 2022;13:4098. doi: 10.1038/s41467-022-31640-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Persson KEM, Lee CT, Marsh K, Beeson JG. Development and optimization of high-throughput methods to measure Plasmodium falciparum-specific growth inhibitory antibodies. J Clin Microbiol. 2006;44:1665–1673. doi: 10.1128/JCM.44.5.1665-1673.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.WorldWide Antimalarial Resistance Network (WWARN) Lumefantrine PK/PD Study Group. Artemether-lumefantrine treatment of uncomplicated Plasmodium falciparum malaria: a systematic review and meta-analysis of day 7 lumefantrine concentrations and therapeutic response using individual patient data. BMC Med. 2015;13:227. doi: 10.1186/s12916-015-0456-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Murungi LM, Kamuyu G, Lowe B, Bejon P, Theisen M, Kinyanjui SM, Marsh K, Osier FHA. A threshold concentration of anti-merozoite antibodies is required for protection from clinical episodes of malaria. Vaccine. 2013;31:3936–3942. doi: 10.1016/j.vaccine.2013.06.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Osier FHA, Fegan G, Polley SD, Murungi L, Verra F, Tetteh KKA, Lowe B, Mwangi T, Bull PC, Thomas AW, et al. Breadth and magnitude of antibody responses to multiple Plasmodium falciparum merozoite antigens are associated with protection from clinical malaria. Infect Immun. 2008;76:2240–2248. doi: 10.1128/IAI.01585-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Drakeley CJ, Corran PH, Coleman PG, Tongren JE, McDonald SL, Carneiro I, Malima R, Lusingu J, Manjurano A, Nkya WM, et al. Estimating medium- and long-term trends in malaria transmission by using serological markers of malaria exposure. Proc Natl Acad Sci USA. 2005;102:5108–5113. doi: 10.1073/pnas.0408725102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Tongren JE, Drakeley CJ, McDonald SLR, Reyburn HG, Manjurano A, Nkya WMM, Lemnge MM, Gowda CD, Todd JE, Corran PH, Riley EM. Target antigen, age, and duration of antigen exposure independently regulate immunoglobulin G subclass switching in malaria. Infect Immun. 2006;74:257–264. doi: 10.1128/IAI.74.1.257-264.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Rono J, Osier FHA, Olsson D, Montgomery S, Mhoja L, Rooth I, Marsh K, Färnert A. Breadth of anti-merozoite antibody responses is associated with the genetic diversity of asymptomatic Plasmodium falciparum infections and protection against clinical malaria. Clin Infect Dis. 2013;57:1409–1416. doi: 10.1093/cid/cit556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Vidarsson G, Dekkers G, Rispens T. IgG subclasses and allotypes: from structure to effector functions. Front Immunol. 2014;5:520. doi: 10.3389/fimmu.2014.00520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Mwangi TW, Mohammed M, Dayo H, Snow RW, Marsh K. Clinical algorithms for malaria diagnosis lack utility among people of different age groups. Trop Med Int Health. 2005;10:530–536. doi: 10.1111/j.1365-3156.2005.01439.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Murungi LM, Sondén K, Llewellyn D, Rono J, Guleid F, Williams AR, Ogada E, Thairu A, Färnert A, Marsh K, et al. Targets and Mechanisms Associated with Protection from Severe Plasmodium falciparum Malaria in Kenyan Children. Infect Immun. 2016;84:950–963. doi: 10.1128/IAI.01120-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Obiero JM, Campo JJ, Scholzen A, Randall A, Bijker EM, Roestenberg M, Hermsen CC, Teng A, Jain A, Davies DH, et al. Antibody Biomarkers Associated with Sterile Protection Induced by Controlled Human Malaria Infection under Chloroquine Prophylaxis. mSphere. 2019;4:e00027–19. doi: 10.1128/mSphereDirect.00027-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Proietti C, Krause L, Trieu A, Dodoo D, Gyan B, Koram KA, Rogers WO, Richie TL, Crompton PD, Felgner PL, Doolan DL. Immune Signature Against Plasmodium falciparum Antigens Predicts Clinical Immunity in Distinct Malaria Endemic Communities. Mol Cell Proteomics. 2020;19:101–113. doi: 10.1074/mcp.RA118.001256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Suscovich TJ, Fallon JK, Das J, Demas AR, Crain J, Linde CH, Michell A, Natarajan H, Arevalo C, Broge T, et al. Mapping functional humoral correlates of protection against malaria challenge following RTS,S/AS01 vaccination. Sci Transl Med. 2020;12:eabb4757. doi: 10.1126/scitranslmed.abb4757. [DOI] [PubMed] [Google Scholar]
  • 51.Feng S, Phillips DJ, White T, Sayal H, Aley PK, Bibi S, Dold C, Fuskova M, Gilbert SC, Hirsch I, et al. Correlates of protection against symptomatic and asymptomatic SARS-CoV-2 infection. Nat Med. 2021;27:2032–2040. doi: 10.1038/s41591-021-01540-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Opi DH, Kurtovic L, Chan JA, Horton JL, Feng G, Beeson JG. Multi-functional antibody profiling for malaria vaccine development and evaluation. Expert Rev Vaccines. 2021;20:1257–1272. doi: 10.1080/14760584.2021.1981864. [DOI] [PubMed] [Google Scholar]
  • 53.Richardson SI, Moore PL. Targeting Fc effector function in vaccine design. Expert Opin Ther Targets. 2021;25:467–477. doi: 10.1080/14728222.2021.1907343. [DOI] [PubMed] [Google Scholar]
  • 54.DiLillo DJ, Tan GS, Palese P, Ravetch JV. Broadly neutralizing hemagglutinin stalk-specific antibodies require FcgammaR interactions for protection against influenza virus in vivo. Nat Med. 2014;20:143–151. doi: 10.1038/nm.3443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Hessell AJ, Hangartner L, Hunter M, Havenith CEG, Beurskens FJ, Bakker JM, Lanigan CMS, Landucci G, Forthal DN, Parren PWHI, et al. Fc receptor but not complement binding is important in antibody protection against HIV. Nature. 2007;449:101–104. doi: 10.1038/nature06106. [DOI] [PubMed] [Google Scholar]
  • 56.Bournazos S, Klein F, Pietzsch J, Seaman MS, Nussenzweig MC, Ravetch JV. Broadly neutralizing anti-HIV-1 antibodies require Fc effector functions for in vivo activity. Cell. 2014;158:1243–1253. doi: 10.1016/j.cell.2014.08.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Garrido JL, Prescott J, Calvo M, Bravo F, Alvarez R, Salas A, Riquelme R, Rioseco ML, Williamson BN, Haddock E, et al. Two recombinant human monoclonal antibodies that protect against lethal Andes hantavirus infection in vivo. Sci Transl Med. 2018;10 doi: 10.1126/scitranslmed.aat6420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Garcia-Senosiain A, Kana IH, Singh S, Das MK, Dziegiel MH, Hertegonne S, Adu B, Theisen M. Neutrophils dominate in opsonic phagocytosis of P. falciparum blood-stage merozoites and protect against febrile malaria. Commun Biol. 2021;4:984. doi: 10.1038/s42003-021-02511-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Ty M, Sun S, Callaway PC, Rek J, Press KD, van der Ploeg K, Nideffer J, Hu Z, Klemm S, Greenleaf W, et al. Malaria-driven expansion of adaptive-like functional CD56-negative NK cells correlates with clinical immunity to malaria. Sci Transl Med. 2023;15:eadd9012. doi: 10.1126/scitranslmed.add9012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bournazos S, Corti D, Virgin HW, Ravetch JV. Fc-optimized antibodies elicit CD8 immunity to viral respiratory infection. Nature. 2020;588:485–490. doi: 10.1038/s41586-020-2838-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Amiah MA, Ouattara A, Okou DT, N’Guetta SA, Yavo W. Polymorphisms in Fc Gamma Receptors and Susceptibility to Malaria in an Endemic Population. Front Immunol. 2020;11:561142. doi: 10.3389/fimmu.2020.561142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bournazos S, Woof JM, Hart SP, Dransfield I. Functional and clinical consequences of Fc receptor polymorphic and copy number variants. Clin Exp Immunol. 2009;157:244–254. doi: 10.1111/j.1365-2249.2009.03980.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kennedy MC, Wang J, Zhang Y, Miles AP, Chitsaz F, Saul A, Long CA, Miller LH, Stowers AW. In vitro studies with recombinant Plasmodium falciparum apical membrane antigen 1 (AMA1): production and activity of an AMA1 vaccine and generation of a multiallelic response. Infect Immun. 2002;70:6948–6960. doi: 10.1128/IAI.70.12.6948-6960.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Silk SE, Kalinga WF, Mtaka IM, Lilolime NS, Mpina M, Milando F, Ahmed S, Diouf A, Mkwepu F, Simon B, et al. Superior antibody immunogenicity of a viral-vectored RH5 blood-stage malaria vaccine in Tanzanian infants as compared to adults. Med. 2023;4:668–686.:e667. doi: 10.1016/j.medj.2023.07.003. [DOI] [PubMed] [Google Scholar]
  • 65.Kariuki SN, Williams TN. Human genetics and malaria resistance. Hum Genet. 2020;139:801–811. doi: 10.1007/s00439-020-02142-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kimingi HW, Kinyua AW, Achieng NA, Wambui KM, Mwangi S, Nguti R, Kivisi CA, Jensen ATR, Bejon P, Kapulu MC, et al. Breadth of Antibodies to Plasmodium falciparum Variant Surface Antigens Is Associated With Immunity in a Controlled Human Malaria Infection Study. Front Immunol. 2022;13:894770. doi: 10.3389/fimmu.2022.894770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Adu B, Dodoo D, Adukpo S, Hedley PL, Arthur FKN, Gerds TA, Larsen SO, Christiansen M, Theisen M. Fc gamma receptor IIIB (FcgammaRIIIB) polymorphisms are associated with clinical malaria in Ghanaian children. PLoS One. 2012;7:e46197. doi: 10.1371/journal.pone.0046197. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Rohatgi S, Gohil S, Kuniholm MH, Schultz H, Dufaud C, Armour KL, Badri S, Mailliard RB, Pirofski LA. Fc gamma receptor 3A polymorphism and risk for HIV-associated cryptococcal disease. mBio. 2013;4:e00573–13. doi: 10.1128/mBio.00573-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Munde EO, Okeyo WA, Raballah E, Anyona SB, Were T, Ong’echa JM, Perkins DJ, Ouma C. Association between Fcgamma receptor IIA, IIIA and IIIB genetic polymorphisms and susceptibility to severe malaria anemia in children in western Kenya. BMC Infect Dis. 2017;17:289. doi: 10.1186/s12879-017-2390-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Damelang T, Rogerson SJ, Kent SJ, Chung AW. Role of IgG3 in Infectious Diseases. Trends Immunol. 2019;40:197–211. doi: 10.1016/j.it.2019.01.005. [DOI] [PubMed] [Google Scholar]
  • 71.Nduati EW, Nkumama IN, Gambo FK, Muema DM, Knight MG, Hassan AS, Jahangir MN, Etyang TJ, Berkley JA, Urban BC. HIV-Exposed Uninfected Infants Show Robust Memory B-Cell Responses in Spite of a Delayed Accumulation of Memory B Cells: an Observational Study in the First 2 Years of Life. Clin Vaccine Immunol. 2016;23:576–585. doi: 10.1128/CVI.00149-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Taylor TE, Molyneux ME, Wirima JJ, Borgstein A, Goldring JD, Hommel M. Intravenous immunoglobulin in the treatment of paediatric cerebral malaria. Clin Exp Immunol. 1992;90:357–362. doi: 10.1111/j.1365-2249.1992.tb05851.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Boyle MJ, Wilson DW, Richards JS, Riglar DT, Tetteh KKA, Conway DJ, Ralph SA, Baum J, Beeson JG. Isolation of viable Plasmodium falciparum merozoites to define erythrocyte invasion events and advance vaccine and drug development. Proc Natl Acad Sci USA. 2010;107:14378–14383. doi: 10.1073/pnas.1009198107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Paweska JT, Jansen van Vuren P, Fenton KA, Graves K, Grobbelaar AA, Moolla N, Leman P, Weyer J, Storm N, McCulloch SD, et al. Lack of Marburg Virus Transmission From Experimentally Infected to Susceptible In-Contact Egyptian Fruit Bats. J Infect Dis. 2015;212:S109–S118. doi: 10.1093/infdis/jiv132. [DOI] [PubMed] [Google Scholar]
  • 75.Murungi LM, Sondén K, Odera D, Oduor LB, Guleid F, Nkumama IN, Otiende M, Kangoye DT, Fegan G, Färnert A, et al. Cord blood IgG and the risk of severe Plasmodium falciparum malaria in the first year of life. Int J Parasitol. 2017;47:153–162. doi: 10.1016/j.ijpara.2016.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Llewellyn D, Miura K, Fay MP, Williams AR, Murungi LM, Shi J, Hodgson SH, Douglas AD, Osier FH, Fairhurst RM, et al. Standardization of the antibody-dependent respiratory burst assay with human neutrophils and Plasmodium falciparum malaria. Sci Rep. 2015;5:14081. doi: 10.1038/srep14081. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Information

Data Availability Statement

  • All data reported in this paper will be shared by the lead contact upon request.

  • This paper does not report original code.

  • Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.

RESOURCES