Skip to main content
Erschienen in: Malaria Journal 1/2015

Open Access 01.12.2015 | Research

Protection of Malian children from clinical malaria is associated with recognition of multiple antigens

verfasst von: Modibo Daou, Bourèma Kouriba, Nicolas Ouédraogo, Issa Diarra, Charles Arama, Yamoussa Keita, Sibiri Sissoko, Boucary Ouologuem, Seydou Arama, Teun Bousema, Ogobara K Doumbo, Robert W Sauerwein, Anja Scholzen

Erschienen in: Malaria Journal | Ausgabe 1/2015

Abstract

Background

Naturally acquired immunity to clinical malaria is thought to be mainly antibody-mediated, but reports on antigen targets are contradictory. Recognition of multiple antigens may be crucial for protection. In this study, the magnitude of antibody responses and their temporal stability was assessed for a panel of malaria antigens in relation to protection against clinical Plasmodium falciparum malaria.

Methods

Malian children aged two to 14 years were enrolled in a longitudinal study and followed up by passive and active case detection for seven months. Plasma was collected at enrolment and at the beginning, in the middle and after the end of the transmission season. Antibody titres to the P. falciparum-antigens apical membrane protein (AMA)-1, merozoite surface protein (MSP)-119, MSP-3, glutamine-rich protein (GLURP-R0) and circumsporozoite antigen (CSP) were assessed by enzyme-linked immunosorbent assay (ELISA) for 99 children with plasma available at all time points. Parasite carriage was determined by microscopy and nested PCR.

Results

Antibody titres to all antigens, except MSP-119, and the number of antigens recognized increased with age. After malaria exposure, antibody titres increased in children that had low titres at baseline, but decreased in those with high baseline responses. No significant differences were found between antibody titers for individual antigens between children remaining symptomatic or asymptomatic after exposure, after adjustment for age. Instead, children remaining asymptomatic following parasite exposure had a broader repertoire of antigen recognition.

Conclusions

The present study provides immune-epidemiological evidence from a limited cohort of Malian children that strong recognition of multiple antigens, rather than antibody titres for individual antigens, is associated with protection from clinical malaria.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12936-015-0567-9) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

MD and NO conducted the experiments; MD and AS planned the experiments; MD, TB and AS analysed the data; MD, BK, OKD, and RS designed and supervised the field study; MD, BK, ID, CA, SS, SA, BO, and YK performed the field studies and collected samples and clinical data; MD, BK, OKD, TB, RWS, and AS interpreted the data and wrote the manuscript. All authors read and approved the final manuscript.
Abkürzungen
AMA
Apical membrane protein
AU
Arbitrary unit
CSP
Circumsporozoite antigen
ELISA
Enzyme-linked immunosorbent assay
GLURP
Glutamine-rich protein
IQR
Interquartile range
MSP
Merozoite surface protein
OR
Odds ratio

Background

Despite the success of major public health control efforts in recent years [1], malaria remains one of the most important causes of morbidity and mortality in the world with an estimated 207 million cases and 627,000 deaths in 2012 [2]. Antibody-mediated immune responses to malaria antigens help to control blood-stage parasitaemia and have a protective effect on clinical disease, as shown in passive transfer experiments [3,4]. Identification of antigens that are the target(s) of these protective antibodies and their induction by natural exposure or immunization are a long-standing subject of fundamental and epidemiological studies as well as clinical vaccine trials [5].
The most commonly studied antigens in this respect are merozoite antigens, which are expressed both by mature liver schizonts as well as during the late schizont and merozoite-stage of blood-stage Plasmodium parasites [6]. Studies in different cohorts or transmission settings sometimes yield contradictory results [6]. One hypothesis is that a certain threshold of antibody responses has to be reached for them to be protective [7]. Other important considerations in immuno-epidemiological studies when assessing associations between immunological readouts, such as humoral responses and clinical protection, include the type of follow-up, the time point of assessment of humoral responses, the definition of ‘protection’ and the number of time points at which immunological readouts are assessed [6]. Additionally, recognition of multiple antigens in combination rather than just a single antigen are likely required for protection [8-10], which is already taken into account in several vaccine development studies and clinical trials [5].
Using a set of samples collected at four time points from a cohort of children followed up longitudinally for seven months, it was investigated whether a high concentration of antibodies against different pre-erythrocytic, blood-stage and cross-stage antigens, namely circumsporozoite antigen (CSP), merozoite surface protein (MSP)-3, apical membrane protein (AMA)-1, MSP-119 and glutamine-rich protein (GLURP-R0), alone or in combination, may be associated with protection from clinical malaria in a hyper-endemic area. It was further assessed whether this association was temporally stable or dependent on the time point of sampling.

Methods

Study area

This study was conducted in Malian children from Samako, a village of about 3,000 people located in the Sudanese savannah zone of the Upper Niger valley (district of Kati) about 70 km southwest of Bamako, the capital of Mali. Samako is 5 km from Bancoumana where the Malaria Research and Training Centre has established a malaria vaccine site since 2000. Plasmodium falciparum is the predominant Plasmodium species in this region and accounts for more than 95% of malaria cases [11]. Transmission is mainly seasonal from June to December [11]. The study area has previously been described as a malaria hyper-endemic area [12]. From July to December of 2011, the overall incidence rate of clinical malaria during the transmission season period was 1.0 (244/243) episodes of malaria per person per season with 0.56 (28/50), 1.56 (78/50), 1.46 (102/70), 0.48 (35/73) episodes per person per season respectively in the age categories 3–11 months, 1–4 years, 5–14 years, and 15–50 years (Kone et al., unpublished data).

Ethical issues

Written informed consent was obtained from parents or legal guardians who consented on behalf of their children. All laboratory procedures were carried out within the guidelines of good laboratory practice. Ethical clearance to conduct the study was sought from the ethical committee of the Faculty of Medicine, Pharmacy and Odonto-Stomatology at the University of Science, Techniques and Technologies of Bamako (approval number 2011-58/FMPOS).

Study subjects, design and conduct

One physician and one biologist were based in the village to follow up a cohort of children aged two to 14 years during one malaria transmission season. In December 2011, 171 children were enrolled in the study and subsequently attended three additional cross-sectional visits during the transmission season in July 2012 (n = 134) and September 2012 (n = 137) and after the end of the transmission season in February 2013 (n = 111). Ninety-nine children attended all three cross-sectional visits and provided samples during and after the 2012 transmission season and were selected for longitudinal immunological analysis in this study.
From July until December clinical malaria infection was monitored by active and passive case detection. The active case detection was carried out by house visits and cross-sectional survey. On a 14-day basis, field workers conducted active house visits to all the children to assess malaria infection. Participants were instructed to register any possible malaria symptoms in a diary. Passive case detection of clinical malaria episodes was carried out at the village health clinic. Children who visited the health clinic were identified using an individual identification card and were screened by a doctor. When children reported any of the malaria symptoms (fever, headache, diarrhoea, vomiting), a rapid diagnostic test (OptiMAL, Flow Inc, Portland, OR, USA) and thick smear were performed and axillary temperature was checked. A clinical malaria episode was defined by presence of any symptom of malaria including fever of 38°C or more, associated with a parasitaemia of at least 5,000 trophozoites/mm3, taking into account that low parasite densities are not always the causative factor of clinical symptoms [13]. Children with asymptomatic parasitaemia were not drug treated, neither at enrolment nor during follow-up.
Out of the 99 children providing plasma samples at all cross-section visits, 74 developed thick-smear detectable and 17 developed PCR-detectable infections with P. falciparum during follow-up (Figure 1). For eight children, no parasitaemia was detected at any time point during follow-up, so they were deemed unexposed. Children with co-infection of P. falciparum with other Plasmodium species, such as Plasmodium malariae, Plasmodium ovale or Plasmodium vivax were excluded from the analysis in the present study.

Sample collection and sample processing

At each cross-sectional visit, 2–5 ml of blood was collected by venipuncture. About 20 μl of the blood was used to prepare a thick and thin smear for the detection of malaria parasites. Slides were stained with Giemsa at pH 7.2. A drop of whole blood was placed on Whatman filter paper strips number 3 (Whatman, Clifton, NJ, USA) and stored at room temperature for retrospective PCR analysis to detect sub-microscopic infections. EDTA anti-coagulated plasma was collected and cryopreserved at −20°C.

Indirect enzyme-linked immunosorbent assay (ELISA)

The following antigens were used to assess humoral responses: (i) the ectodomain of apical membrane antigen (AMA)-1 of P. falciparum FVO, comprising amino acids 25–545, expressed in the yeast Pichia pastoris [14,15]; (ii) the conserved non-repeat N-terminal region R0 of glutamate rich protein (GLURP-R0; amino acids 85–213) [16]; (iii) full-length merozoite surface protein (MSP)-3 (K1) [17]; (iv) the 19-kilodalton carboxy-terminal fragment of MSP-1, expressed in Escherichia coli (MSP-119) [18]; and, (v) full-length circumsporozoite protein (CSP) obtained from Genova Biotechniques Pvt. Ltd, Hyderabad, India.
While CSP is a pure pre-erythrocytic antigen expressed during the sporozoite and liver-stage of the parasite [19-21], and MSP-3 can only be detected on blood-stage parasites [22], AMA-1 [23-26], MSP-1 [27-29] and GLURP [30] are cross-stage antigens expressed during both the pre-erythrocytic and blood-stage of the parasite.
Microtitre plates (Maxisorb; NUNC) were coated with 50 μl of antigen diluted in PBS at a concentration of 1 μg/ml (AMA-1), 0.5 μg/ml (GLURP-R0), 0.18 μg/ml (MSP-119), 0.5 μg/ml (MSP-3) or 0.25 μg/ml (CSP). Plates were incubated overnight at 4°C, washed and blocked with 200 μl of 5% milk powder in PBS for one hour. Due to the expected high inter-individual variation in antibody titers, plasma samples were assayed at two different dilutions (1:500 and 1:50) to ensure that at least one of the two would fit into the range of the standard curve. Fifty μl of diluted plasma samples were added in duplicate and incubated at room temperature for three hours. Plates were washed four times between steps. Polyclonal goat anti-human IgG-HRP (Pierce, Thermo Scientific) diluted 1:40,000 was added to antigen-coated plates. Bound secondary antibodies for total IgG were quantified with ready-to-use TMB (tetramethylbenzidine; TebuBio Laboratories) substrate for 30 min. Fifty μl of 0.2 M H2SO4 were used to stop the reaction. The plates were read out at the spectrophotometrical absorbance of 450 nm.
A plasma pool of hyperimmune Tanzanian adults living in a highly malaria-endemic area was used as serum standard, defined to contain 100 arbitrary units (AU) [31]. Optical density values for the test samples were converted into antibody units with the standard reference curves generated for each ELISA plate using a four-parameter, curve-fit, Microsoft Excel-based application ADAMSEL-v1.1 [32].

Parasite detection by PCR

DNA was extracted from filter papers using the Chelex protocol as described by Walsh et al. [33]. The 18S PCR protocol developed by Snounou et al. [34] targeting the small ribosomal subunit of P. falciparum was used. PCR was performed according to the original protocol except that the quantity of template used in the N1 reaction was increased from 1 to 5 μl. In every set of PCR conditions 5 μL template was used in the N1 reaction and 1.5 μl of product in the N2 reaction. For a more detailed overview of primer sequences and product sizes, and PCR cycling conditions, see Additional file 1 and Additional file 2, respectively. Pooled DNA extracts from P. falciparum NF54 cultured in Nijmegen, The Netherlands were run on every PCR plate as a positive control, alongside a negative water control. The positive control was diluted to the extent that both N1 and N2 fragments were sufficiently amplified so that both amplicons could be visualized on agarose gel. N1 and N2 products were mixed and 10 μl was visualized on 0.8% agarose gel by electrophoresis in 0.5× Tris-acetate-EDTA buffer (0.04 M Tris-acetate and 1 mM EDTA, pH 8.0).

Statistical analysis

Statistical analysis was performed using GraphPad Prism v5 software and STATA version 12.0 (Statacorp, College Station, Texas, USA). Differences in responses among different subject groups (different age groups; asymptomatic and symptomatic individuals; low, intermediate and high responders) were analysed by non-parametric measures. Mann–Whitney U test was used for two separate groups, Kruskal-Wallis with Dunn’s multiple comparison post-hoc test for more than two groups and Wilcoxon matched-pairs signed rank test for paired analysis of two time points. Associations between age and humoral responses were assessed by Spearman correlation; associations between the number of antigens recognized (AU > 10) and age in categories (2–5, 6–9 and 10–15 years), parasite status at enrolment (parasite-free, submicroscopic and microscopic parasite carriage) and ‘protection’ (asymptomatic parasite carriage or more than one clinical malaria episode) was determined by Poisson regression where the number of antigens recognized was analysed as count variable. Trends in the proportion of individuals recognizing ≥1 or ≥3 antigens and age in categories or parasite status were determined by logistic regression. The association between experiencing clinical malaria episodes or asymptomatic malaria and the density of antibody responses was further assessed by logistic regression models with antibody densities as categorical variables (<1 AU, 1–10 AU and ≥10 AU). For analyses where ELISA outcomes were analysed on a continuous scale, AU were log-transformed (log10) and analysed by linear regression. All associations were adjusted for age where appropriate. A p-value of < 0.05 was considered statistically significant.

Results

Association of antibody titres and breadth of the humoral response with age and parasitaemia

A longitudinal study was performed in 99 children during one malaria transmission season in Samako, a village in a hyperendemic area of Mali. The frequency of P. falciparum infections was highest during the July cross-sectional visit at the beginning of transmission season, with a frequency of slide positive children of 27.6% (27/98), and a proportion of parasite-positive children by PCR of 78.6% (77/98) (Table 1). Both the demographic and parasitological parameters of this longitudinal cohort were highly similar to the original total cohort of 171 children, from which this longitudinal cohort was selected based on attendance of all four cross-sectional visits (Additional file 3). The number of clinical malaria episodes recorded during longitudinal follow-up of this cohort from July to Dec 2012 peaked in October (July n = 1, Aug n = 8, Sept n = 9, Oct n = 19, Nov n = 17, Dec n = 4).
Table 1
Demographic and parasitological parameters
 
Dec 2011
Jul 2012
Sept 2012
Feb 2013
Number of individuals (N)
n = 99
   
Gender,% male (n/total)
55.6% (55/99)
   
Age in year, median (range)
7 (2–14)
   
Haemoglobin (g/dL), median (range)
11.6 (7.3-14.5)
11 (7.4-14.3)
11.6 (6.5-14.6)
12.2 (5.7-16.3)
Anaemia,% Hb <11 g/dL (n/total)
26.3% (26/99)
47.5% (47/99)
27.3% (27/99)
13.3% (13/98)a
Thick smear positive (%, n/total)
18.2% (18/99)
27.6% (27/98)a
20.2% (20/99)
10.8% (10/93)a
PCR positive (%, n/total)
n.d.
78.6% (77/98)a
43.6% (41/94)a
n.d.
Parasitaemia in thick-smear positive individuals (n = 35), median (range)
12,900 (100–51,300)
3,150 (100–132,275)
1,262 (100–58,675)
1,325 (100–7775)
n.d. not done.
aFor some children not all data were collected at the time of visit.
Antibody responses against five P. falciparum malaria antigens were assessed in the cohort at enrolment, the beginning of the transmission season (baseline), during and after the transmission season. At all time points there was a positive correlation between age and antibody titres for AMA-1, MSP-3, CSP and GLURP-R0, while responses to MSP-119 were not associated with age (Additional file 4). Stratification of children into three age categories of two to five years, six to nine years and ten to 15 years revealed that the main increase in AMA-1 titres occurred before the age of six, while antibody levels for MSP-3, CSP and GLURP-R0 rose in a more continuous manner, which was evident both at baseline (Figure 2) and at the peak and end of the transmission season (Additional file 5 and Additional file 6).
The next question was whether the breadth of the response also increased with age. Indeed, at all time points during follow-up there was a significant correlation between age and the number of antigens to which children showed an antibody level that reached at least 10% (>10 AU) of the hyperimmune reference serum (Additional file 4). Stratification into age groups showed that this increase occurred again before the age of six, with no difference between the older age groups (Figure 3A). Early in the transmission season (July 2012) 47.2% (17/36) of the children in the youngest age group had no high reactivity to any of the antigens, which decreased to 29.6% (8/27) in six to nine years old and 16.7% (6/36) in ten to 14 years old (p for trend = 0.007). While only 13.9% (5/36) of the two to five years old had high levels of antibodies against three or more antigens, this proportion was more than twice as high in six to nine year old children (29.6%, 8/27) and ten to 14 years old children (30.6%, 11/36); Figure 3B) (p for trend = 0.10). The breadth of the humoral response was significantly associated with parasitaemia at time of sampling (Figure 3C): 57.1% (12/21) of children with no detectable parasites recognized not a single antigen strongly, while this was only true for 28.0 (14/50) and 14.8% (4/27) of children with sub-microscopic or microscopic parasitaemia, respectively (p for trend = 0.007), after adjustment for age. While there was no significant difference in microscopically detectable parasitaemia between the different age categories (p = 0.79), PCR detectable parasitaemia, regardless of thick smear positivity, however, increased with age (p = 0.04; Figure 3D).

Dependency of boosting of humoral responses on early season antibody levels

The breadth of the antibody response, i.e., the number of antigens strongly recognized (>10 AU) per individual, was stable over the time of follow-up, as evidenced by the strong correlation between this readout at enrolment (December 2011) and the beginning (July 2012; Spearman r = 0.59, p < 0.0001), peak (September 2012; r = 0.70, p < 0.0001) and end of the transmission season (February 2013; r = 0.67, p < 0.0001). Next it was investigated whether exposure during the transmission season altered antibody levels for the individual antigens. During the seven months of follow-up, 91/99 children became parasite-positive by either PCR or thick smear during one or more visits. Of these, 47 children remain asymptomatic while 44 experienced one or more episodes of clinical malaria (Figure 1).
Comparing antibody responses in all n = 91 malaria exposed children, there were no statistically significant difference for any of the antigens were observed between the beginning and end of the transmission season, (median AU [July 2012, February 2013]: AMA-1 [7.8, 7.2] p = 0.058; MSP-119 [2.37, 2.51] p = 0.46; MSP-3 [11.6, 12.0] p = 0.27; CSP [6.14, 4.66] p = 0.18; GLURP-R0 [3.40, 3.09] p = 0.80). Since boosting of antibody responses may depend on the strength of the pre-existing response, all exposed children (n = 75) were stratified into low (<1 AU), intermediate (1–10 AU) and high responders (>10 AU) for each antigen. Low early season-responders for AMA-1, MSP-119 and GLURP-R0 showed higher antibody levels after the transmission season, while these titers remained largely unchanged in intermediate early season-responders (Table 2). For MSP-3, there was only a single low early season-responder. For this antigen, intermediate early season-responders showed boosted antibody levels after the transmission season. The opposite was observed for children showing high early season antibody responses: for these children, post-transmission season antibody levels for MSP-119, MSP-3 and CSP were significantly lower than their titers at the beginning of the season (Table 2). In contrast, boosting or waning of antibody titers was not depend on age, since without stratification by early season antibody titers, there was no significant difference between early and post-season antibody titers in any of the different age categories (Additional file 7).
Table 2
Changes of antibody titers after parasite exposure during the transmission season dependent on baseline reactivity
 
AMA-1
MSP-1 19
MSP-3
CSP
GLURP-R0
<1 AU
n = 21a
n = 28
n = 1
n = 6
n = 15
July 2012 median [IQR]
0.094 [0.01-0.25]
0.22 [0.01-0.6]
 
0.73 [0.50-0.86]
0.48 [0.2-0.62]
Feb 2013 median [IQR]
0.34 [0.09-2.0]
0.95 [0.05-2.0]
 
1.99 [1.39-2.24]
1.30 [0.88-2.16]
p-value b
0.002
<0.0001
n.d.
0.03
0.002
1-10 AU
n = 28
n = 39
n = 43
n =59
n = 56
July 2012 median [IQR]
3.0 [1.62-6.89]
2.5 [1.35-4.72]
5.05 [3.75-7.23]
4.16 [1.88-6.98]
3.15 [2.12-4.90]
Feb 2013 median [IQR]
4.4 [1.91-10.05]
2.46 [1.08-4.21]
7.21 [4.87-12.73]
4.66 [2.19-8.12]
3.06 [2.08-6.05]
p-value b
0.067
0.96
<0.0001
0.11
0.33
>10 AU
n = 42
n = 24
n = 47
n = 26
n = 20
July 2012 median [IQR]
37.1 [15.1-75.7]
23.9 [16.1-68.9]
29.8 [16.1-82.2]
14.7 [11.7-22.4]
22.4 [13.3-57.6]
Feb 2013 median [IQR]
36.1 [11.1-108.8]
15.3 [6.9-46.2]
19.9 [9.7-51.2]
8.1 [3.96-14.1]
20.2 [3.5-89.9]
p-value b
0.45
0.047
0.002
<0.0001
0.59
a91 children that experiences parasitaemia detected by qPCR or thick smear where included in this analysis.
bAntibody reactivity was compared between July and February for each group and antigen by Wilcoxon matched-pairs signed rank test.
Preferential boosting in the group of children with low antibodies at the beginning of the season could be due to the particularly high frequency of clinical malaria episodes during the transmission season in this group, compared to the intermediate or high early season-responders. This was indeed observed regardless of which antigen was used for stratification (Additional file 8). One possible explanation for lower antibody levels after the transmission season compared to baseline, as found in the high early season-responder group, might be temporarily elevated antibody levels at baseline due to an ongoing malaria infection. Indeed, there was a trend that the frequency of children with microscopic parasitaemia was highest in the early season high responder group and lowest in the early season low responder group for GLURP (p = 0.02), AMA-1 (p = 0.06) and MSP-1 (p = 0.11) (Additional file 9). For AMA-1 (p = 0.04) and MSP-3 (p = 0.03), this distinction was even found for the total proportion of children with any (either sub-microscopic or microscopic) parasitaemia, despite the fact that the majority of children (77/90) included in this longitudinal analysis was PCR positive at baseline.

Association of antibody responses with clinical protection

Because the youngest children showed overall weaker antibody responses and recognized a smaller number of antigens strongly than older children, it was next verified whether age might be a determinant for protection from clinical malaria during the transmission season. Overall, children remaining asymptomatic were older than those experiencing clinical malaria episodes, although this difference was not statistically significant (Figure 4A; p = 0.09). When age was dichotomized, children two to five years of age had a higher proportion of clinical episodes than the two older age groups (p = 0.049; Figure 4B). Therefore, all the following statistical analyses were adjusted for age.
Compared to children developing clinical malaria episodes, asymptomatic children showed significantly higher levels of antibodies against AMA-1, and non-significantly higher antibody responses for MSP-3 and GLURP-R0, at the start of the season (Figures 5A, C and E). These differences in antibody levels found early in the transmission season were also observed at enrolment and during longitudinal follow-up for AMA-1, MSP-3 and GLURP-R0 (Additional file 10A, C and E; Additional file 11A and C; Additional file 12A and E). However, none of these differences were statistically significant when adjusted for age. The levels of anti-MSP-119 and anti-CSP antibodies were comparable between children remaining asymptomatic and symptomatic at enrollment (Additional file 10) and during the entire follow-up (Additional files 11B and 12B). When antibody responses were dichotomized as high responders (>10 AU) and low responders, only high antibody responses to AMA-1 (AU > 10) in July 2012 were associated with a lower risk of clinical malaria episodes in the subsequent season (OR 0.37, 95% CI 0.15-0.91, p = 0.03). No such association was observed MSP-1 (OR 0.92, 95% CI 0.35-2.37, p = 0.86), MSP-3 (OR 0.60, 95% CI 0.25-1.40, p = 0.24), GLURP (OR 0.62, 95% CI 0.21-1.80, p = 0.38) or CSP (OR 1.00, 95% CI 0.39-2.57, p = 0.99).
While recognition of individual antigen was therefore no good predictor of protection from clinical disease, children remaining asymptomatic recognized a broader repertoire of antigens compared to those that developed clinical disease (Figure 6A (December 2011, p = 0.03); B (July 2012, p = 0.12); C (February 2013, p = 0.008)). Although not always statistically significant, across all time points analysed, a greater proportion of children that became symptomatic during the season lacked high reactivity to any of the antigens compared to asymptomatic children, while strong responses to three or more antigens were found in a greater proportion of asymptomatic children compared to those that were clinically unprotected (Figure 6). Amongst those children with high reactivity for three to five antigens, 95.4% had strong responses for MSP-3 (median across all time points), 81.0% for AMA-1, 72.3% for CSP, 66.3% for GLURP-R0 and 53.8% for MSP-119. Children strongly recognizing three or more antigens has a reduced risk of developing clinical malaria, both when antibody titers were assessed prior to (prospective analysis, December 2011 and July 2012) or after (retrospective analysis, February 2013) the 2012 transmission season, in which clinical malaria was detected (Figure 6D). Again, these data did not all reach significance, but showed a clear trend across time points.

Discussion

Immuno-epidemiological studies in multiple settings are needed to further an understanding of the relationship between naturally acquired antibody responses to the parasite and protection from clinical malaria. The key finding of the present study is that in a highly endemic area in Mali, the risk of developing clinical malaria upon exposure was reduced when children showed strong recognition of three or more malaria antigens at any time point during the study, regardless whether those responses were measured before or after the malaria risk was assessed.
Studies into antibody correlates of protection can be affected by a number of confounders. A meta-analysis of other studies assessing the association between antibodies and protection from clinical disease found that most studies do not exclude individuals without detected parasitaemia during follow-up from the ‘protected’ group, although there is no evidence that these individuals were even exposed [6]. Such practice may decrease the magnitude of the observed effect between high and low responders [35]. In the present study, individuals without evidence of malaria exposure during the study, as assessed by PCR and microscopy, were excluded from the analysis on clinical protection. In this context, a limitation of any immune-epidemiological study including the present one, in assessing exposure to the parasite is the frequency of monitoring for parasitaemia. Ideally, parasitaemia would be determined at weekly intervals by PCR. In the present study, parasitaemia was only assessed microscopically at the four cross-sectional visits, or when children reported malaria symptoms during the bi-weekly active case detection visits or at the local health clinic. Furthermore, PCR data were only available for two of the cross-sectional visits. This sampling approach detected many asymptomatic infections that allowed us to exclude potentially unexposed individuals [35], but may not have captured all infections and therefore may have resulted in the exclusion of some individuals with asymptomatic infections of short duration [36]. Of note, in the present study setting, a considerable proportion of infections were detectable by PCR only [37,38]. Parasite prevalence was almost threefold higher by PCR than by microscopy, which has been reported before but is higher than typically observed in areas with this level of transmission intensity [37]. The findings suggest that a considerable proportion of children are capable of controlling infections to densities below the microscopic threshold density for detection.
Age, reflecting cumulative malaria exposure, is an important determinant of protection against clinical malaria. In this study, children ranging from age two to 14 years were enrolled. A decreasing risk for clinical malaria [39], severe disease [40], and an increase of antibody responses [41,42] with age are commonly reported. Accordingly, in the present study, an increase in humoral responses and clinical protection particularly up to the age of six years was also observed. As a result, apparently stronger antibody responses to AMA-1, MSP-3 and GLURP-R0 at the beginning of the transmission season in children protected from clinical disease were found to be largely explained by their higher age. Nevertheless, even when adjusting for age, high responders to AMA-1 and high responders to a combination of three or more antigens had a lower risk for clinical disease. Therefore, it is no age per se, but the acquisition of specific and strong responses with the degree of cumulative exposure that determines humoral protection.
An important consideration when interpreting the findings from the current study is the size of the study cohort. In the current study, the number of children from which samples could be analysed was reduced by a number of factors, including commitment to longitudinal assessment and attendance of all cross-sectional visits. This approach was chosen to allow for a single consistent data set to be used for all analysis, as opposed to doing analysis for each time point with a different, only partially overlapping set of participants. While this did not lead to a skewing in the demographic or parasitological parameters compared to the original cohort, the two groups of children remaining asymptomatic or becoming symptomatic upon parasite exposure were too small for several of the results to reach statistical significance. Nevertheless, clear trends were observed throughout time points, justifying future studies in larger cohorts.
It was previously highlighted that studies measuring responses at multiple time points are needed to better determine the serological status of an individual and whether the time point of observation may have an impact on the association of the humoral responses with protection [6,43]. Changes in antibody titres after exposure were, therefore, also examined. Overall, breadth and magnitude of the antibody response in this cohort were stable over the period of observation that comprised one complete transmission season. This stability of antibody responses may explain why associations of clinical protection with recognition of multiple antigens could be observed at any time point examined. This may, however be different in other transmission settings, where antibody levels are less stable. With the exception of AMA-1, boosting of antibody titres during the transmission season could only be observed when children were stratified by their baseline antibody levels, rather than when analysed as a whole group or stratified by age. Children with low baseline antibodies level showed boosting of blood- and cross-stage antigens and a similar trend for the pre-erythrocytic antigen CSP. Surprisingly, children with high baseline antibodies level (>10 AU) showed decreased rather than increased titres after the transmission season, which coincided with a lower frequency of clinical malaria episodes that these children experienced during the transmission season. The risk of symptomatic malaria and disease severity is often associated with higher parasitaemia [39,44]. Whether the lower frequency of clinical malaria episodes in this group also reflects lower parasitaemia and hence less antigen exposure during asymptomatic episodes in the transmission season could not be determined, since parasitaemia measurements were available from too few time points. Lower antigen exposure would, however, at least partially explain the less efficient boosting of antibody responses in this group of children.
The apparent decrease in antibody titres over the transmission season in these children might be an artifact caused by asymptomatic parasitaemia at time of plasma sampling: At the beginning of the transmission season the group of children with the highest antibody titres for individual antigens also had the highest frequency of parasitaemia, detected either by thick smear or PCR.
That concurrent, even asymptomatic, low-density parasitaemia is associated with elevated antibody titres or prevalence, is in line with previous data [35,41,45-48]. Similarly, the number of strongly recognized antigens was also higher when children carried parasites at the time of sampling, again in line with other studies [10]. Such elevated antibody levels during or shortly after acute infection mostly stem from short-lived plasma blasts, only some of which will eventually establish themselves in survival niches as long-lived plasma cells [49]. Contraction of this short-lived response soon after parasite clearance results in a drop of antibody levels that is well reported for malaria antigens [47,50] and might be relatively greater than the small increase in stable antibody levels (in the absence of parasites or exposure) derived from long-lived plasma cells induced during the transmission season [42].
Amongst the four blood- and cross-stage antigens examined, MSP-119 was the only one for which reactivity did not increase with age, nor did titers for it differ at all clinical protection in this study. Although antibodies to MSP-119 are generally found to confer protection [6,51], this effect is weakest for the Wellcome allele of MSP-119 [6] and also was not found by all studies [52], the reason for which could be the exact source and sequence of antigen, but also actual difference between cohorts. The result of the present study is in line with a previous study finding no protection-association when using the same MSP-119 antigen [10]. Antibody titers for the other blood- and cross-stage antigens AMA-1, MSP-3 and GLURP showed either no significant difference or only trends for slightly higher responses in children remaining asymptomatic; any apparent differences here were clearly age-confounded. Further, there was no difference in CSP antibody titers between children protected or not from clinical disease. Although occasionally reported [53], CSP responses are not commonly found to be associated with clinical protection [54-57], but can rather confer protection from infection at high titres [58].
When analysed as single antigens, only antibody responses to AMA-1 were found to be associated with a reduced risk of clinical malaria. In contrast, high responses for a combination of at least 3/5 malaria antigens showed a relatively consistent protective effect across all time points analysed. These findings are in line with previous studies showing that protection against malaria may not be associated with the humoral responses to one antigen in isolation, but instead with recognition of a panel of antigens [8-10,59,60]. Moreover, MSP-3 and AMA-1 were more prevalently recognized than other antigens amongst children showing responses to multiple antigens, confirming a previous study showing that recognition of these two antigens in combination was more strongly associated with protection than other combinations [10]. In the present study, assessment of antibody responses was limited to five classical vaccine candidate antigens by conventional ELISA. Recently, it was reported that Kenyan children have antibody responses against novel and little-studied merozoites proteins, which had high protective efficacy, particularly when five out of the ten leading proteins were recognized [59]. Application of protein microarrays probed with a large set of P. falciparum antigens to samples from the present and similar studies with carefully defined, protected and non-protected cohorts will be a useful tool to extend these findings and add to the first few existing studies to identify novel antigens and panels thereof associated with protection from clinical disease [9,54]. Based on this, screening for a panel of antigens in field studies could be implemented using bead-based multiplex ELISAs [61-63], which are more cost-efficient than full protein microarrays.

Conclusions

The present study provides immune-epidemiological evidence from a limited cohort of Malian children that protection from clinical malaria is associated with high antibody titres for three or more malaria antigens. Future studies should confirm these findings for other settings and study periods with study populations that are adequately powered to detect more subtle associations between antibody recognition, age and clinical malaria.

Acknowledgements

The field study and MD were funded by the APRIORI (African Poverty Related Infection Oriented Research Initiative, project number W.07.05.203.00) programme, subsidized by WOTRO Science for Global Development, a subdivision of The Netherlands Organization of Scientific Research (NOW). BK has received an EDCTP (European an Developing Countries Clinical Trial Partnership) senior fellowship. EDCTP project TA.2010.40200.007 enabled the establishment of the study site. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
We thank Kevin Tetteh for providing recombinant MSP-119 and MSP-3 antigens, and Amrish Baidjoe for assistance with DNA extraction and PCR analysis.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

MD and NO conducted the experiments; MD and AS planned the experiments; MD, TB and AS analysed the data; MD, BK, OKD, and RS designed and supervised the field study; MD, BK, ID, CA, SS, SA, BO, and YK performed the field studies and collected samples and clinical data; MD, BK, OKD, TB, RWS, and AS interpreted the data and wrote the manuscript. All authors read and approved the final manuscript.
Anhänge

Additional files

Literatur
1.
Zurück zum Zitat Feachem RG, Phillips AA, Hwang J, Cotter C, Wielgosz B, Greenwood BM, et al. Shrinking the malaria map: progress and prospects. Lancet. 2010;376:1566–78.CrossRefPubMedCentralPubMed Feachem RG, Phillips AA, Hwang J, Cotter C, Wielgosz B, Greenwood BM, et al. Shrinking the malaria map: progress and prospects. Lancet. 2010;376:1566–78.CrossRefPubMedCentralPubMed
2.
Zurück zum Zitat WHO. World malaria report. Geneva: World Health Organization; 2013. WHO. World malaria report. Geneva: World Health Organization; 2013.
3.
Zurück zum Zitat Cohen S, Mc GI, Carrington S. Gamma-globulin and acquired immunity to human malaria. Nature. 1961;192:733–7.CrossRefPubMed Cohen S, Mc GI, Carrington S. Gamma-globulin and acquired immunity to human malaria. Nature. 1961;192:733–7.CrossRefPubMed
4.
Zurück zum Zitat Sabchareon A, Burnouf T, Ouattara D, Attanath P, Bouharoun-Tayoun H, Chantavanich P, et al. Parasitologic and clinical human response to immunoglobulin administration in falciparum malaria. Am J Trop Med Hyg. 1991;45:297–308.PubMed Sabchareon A, Burnouf T, Ouattara D, Attanath P, Bouharoun-Tayoun H, Chantavanich P, et al. Parasitologic and clinical human response to immunoglobulin administration in falciparum malaria. Am J Trop Med Hyg. 1991;45:297–308.PubMed
5.
6.
Zurück zum Zitat Fowkes FJ, Richards JS, Simpson JA, Beeson JG. The relationship between anti-merozoite antibodies and incidence of Plasmodium falciparum malaria: a systematic review and meta-analysis. PLoS Med. 2010;7:e1000218.CrossRefPubMedCentralPubMed Fowkes FJ, Richards JS, Simpson JA, Beeson JG. The relationship between anti-merozoite antibodies and incidence of Plasmodium falciparum malaria: a systematic review and meta-analysis. PLoS Med. 2010;7:e1000218.CrossRefPubMedCentralPubMed
7.
Zurück zum Zitat Murungi LM, Kamuyu G, Lowe B, Bejon P, Theisen M, Kinyanjui SM, et al. A threshold concentration of anti-merozoite antibodies is required for protection from clinical episodes of malaria. Vaccine. 2013;31:3936–42.CrossRefPubMedCentralPubMed Murungi LM, Kamuyu G, Lowe B, Bejon P, Theisen M, Kinyanjui SM, et al. A threshold concentration of anti-merozoite antibodies is required for protection from clinical episodes of malaria. Vaccine. 2013;31:3936–42.CrossRefPubMedCentralPubMed
8.
Zurück zum Zitat John CC, Moormann AM, Pregibon DC, Sumba PO, McHugh MM, Narum DL, et al. Correlation of high levels of antibodies to multiple pre-erythrocytic Plasmodium falciparum antigens and protection from infection. Am J Trop Med Hyg. 2005;73:222–8.PubMed John CC, Moormann AM, Pregibon DC, Sumba PO, McHugh MM, Narum DL, et al. Correlation of high levels of antibodies to multiple pre-erythrocytic Plasmodium falciparum antigens and protection from infection. Am J Trop Med Hyg. 2005;73:222–8.PubMed
9.
Zurück zum Zitat Trieu A, Kayala MA, Burk C, Molina DM, Freilich DA, Richie TL, et al. Sterile protective immunity to malaria is associated with a panel of novel P. falciparum antigens. Mol Cell Proteomics. 2011;10:M111.007948.CrossRefPubMedCentralPubMed Trieu A, Kayala MA, Burk C, Molina DM, Freilich DA, Richie TL, et al. Sterile protective immunity to malaria is associated with a panel of novel P. falciparum antigens. Mol Cell Proteomics. 2011;10:M111.007948.CrossRefPubMedCentralPubMed
10.
Zurück zum Zitat Osier FH, Fegan G, Polley SD, Murungi L, Verra F, Tetteh KK, et al. Breadth and magnitude of antibody responses to multiple Plasmodium falciparum merozoite antigens are associated with protection from clinical malaria. Infect Immun. 2008;76:2240–8.CrossRefPubMedCentralPubMed Osier FH, Fegan G, Polley SD, Murungi L, Verra F, Tetteh KK, et al. Breadth and magnitude of antibody responses to multiple Plasmodium falciparum merozoite antigens are associated with protection from clinical malaria. Infect Immun. 2008;76:2240–8.CrossRefPubMedCentralPubMed
11.
Zurück zum Zitat Sagara I, Rulisa S, Mbacham W, Adam I, Sissoko K, Maiga H, et al. Efficacy and safety of a fixed dose artesunate-sulphamethoxypyrazine-pyrimethamine compared to artemether-lumefantrine for the treatment of uncomplicated falciparum malaria across Africa: a randomized multi-centre trial. Malar J. 2009;8:63.CrossRefPubMedCentralPubMed Sagara I, Rulisa S, Mbacham W, Adam I, Sissoko K, Maiga H, et al. Efficacy and safety of a fixed dose artesunate-sulphamethoxypyrazine-pyrimethamine compared to artemether-lumefantrine for the treatment of uncomplicated falciparum malaria across Africa: a randomized multi-centre trial. Malar J. 2009;8:63.CrossRefPubMedCentralPubMed
12.
Zurück zum Zitat Dolo A, Camara F, Poudiougo B, Toure A, Kouriba B, Bagayogo M, et al. Epidemiology of malaria in a village of Sudanese savannah area in Mali (Bancoumana). 2. Entomo-parasitological and clinical study. Bull Soc Pathol Exot. 2003;96:308–12.PubMed Dolo A, Camara F, Poudiougo B, Toure A, Kouriba B, Bagayogo M, et al. Epidemiology of malaria in a village of Sudanese savannah area in Mali (Bancoumana). 2. Entomo-parasitological and clinical study. Bull Soc Pathol Exot. 2003;96:308–12.PubMed
13.
Zurück zum Zitat Schellenberg JR, Smith T, Alonso PL, Hayes RJ. What is clinical malaria? finding case definitions for field research in highly endemic areas. Parasitol Today. 1994;10:439–42.CrossRefPubMed Schellenberg JR, Smith T, Alonso PL, Hayes RJ. What is clinical malaria? finding case definitions for field research in highly endemic areas. Parasitol Today. 1994;10:439–42.CrossRefPubMed
14.
Zurück zum Zitat Kocken CH, Withers-Martinez C, Dubbeld MA, van der Wel A, Hackett F, Valderrama A, et al. High-level expression of the malaria blood-stage vaccine candidate Plasmodium falciparum apical membrane antigen 1 and induction of antibodies that inhibit erythrocyte invasion. Infect Immun. 2002;70(8):4471–6.CrossRefPubMedCentralPubMed Kocken CH, Withers-Martinez C, Dubbeld MA, van der Wel A, Hackett F, Valderrama A, et al. High-level expression of the malaria blood-stage vaccine candidate Plasmodium falciparum apical membrane antigen 1 and induction of antibodies that inhibit erythrocyte invasion. Infect Immun. 2002;70(8):4471–6.CrossRefPubMedCentralPubMed
15.
Zurück zum Zitat Faber BW, Remarque EJ, Kocken CH, Cheront P, Cingolani D, Xhonneux F, et al. Production, quality control, stability and pharmacotoxicity of cGMP-produced Plasmodium falciparum AMA1 FVO strain ectodomain expressed in Pichia pastoris. Vaccine. 2008;26(48):6143–50.CrossRefPubMed Faber BW, Remarque EJ, Kocken CH, Cheront P, Cingolani D, Xhonneux F, et al. Production, quality control, stability and pharmacotoxicity of cGMP-produced Plasmodium falciparum AMA1 FVO strain ectodomain expressed in Pichia pastoris. Vaccine. 2008;26(48):6143–50.CrossRefPubMed
16.
Zurück zum Zitat Hermsen CC, Verhage DF, Telgt DS, Teelen K, Bousema JT, Roestenberg M, et al. Glutamate-rich protein (GLURP) induces antibodies that inhibit in vitro growth of Plasmodium falciparum in a phase 1 malaria vaccine trial. Vaccine. 2007;25:2930–40.CrossRefPubMed Hermsen CC, Verhage DF, Telgt DS, Teelen K, Bousema JT, Roestenberg M, et al. Glutamate-rich protein (GLURP) induces antibodies that inhibit in vitro growth of Plasmodium falciparum in a phase 1 malaria vaccine trial. Vaccine. 2007;25:2930–40.CrossRefPubMed
17.
Zurück zum Zitat Polley SD, Tetteh KK, Lloyd JM, Akpogheneta OJ, Greenwood BM, Bojang KA, et al. Plasmodium falciparum merozoite surface protein 3 is a target of allele-specific immunity and alleles are maintained by natural selection. J Infect Dis. 2007;195:279–87.CrossRefPubMed Polley SD, Tetteh KK, Lloyd JM, Akpogheneta OJ, Greenwood BM, Bojang KA, et al. Plasmodium falciparum merozoite surface protein 3 is a target of allele-specific immunity and alleles are maintained by natural selection. J Infect Dis. 2007;195:279–87.CrossRefPubMed
18.
Zurück zum Zitat Burghaus PA, Holder AA. Expression of the 19-kilodalton carboxy-terminal fragment of the Plasmodium falciparum merozoite surface protein-1 in Escherichia coli as a correctly folded protein. Mol Biochem Parasitol. 1994;64:165–9.CrossRefPubMed Burghaus PA, Holder AA. Expression of the 19-kilodalton carboxy-terminal fragment of the Plasmodium falciparum merozoite surface protein-1 in Escherichia coli as a correctly folded protein. Mol Biochem Parasitol. 1994;64:165–9.CrossRefPubMed
19.
Zurück zum Zitat Singh AP, Buscaglia CA, Wang Q, Levay A, Nussenzweig DR, Walker JR, et al. Plasmodium circumsporozoite protein promotes the development of the liver stages of the parasite. Cell. 2007;131:492–504.CrossRefPubMed Singh AP, Buscaglia CA, Wang Q, Levay A, Nussenzweig DR, Walker JR, et al. Plasmodium circumsporozoite protein promotes the development of the liver stages of the parasite. Cell. 2007;131:492–504.CrossRefPubMed
20.
Zurück zum Zitat Hollingdale MR, Leland P, Leef JL, Leef MF, Beaudoin RL. Serological reactivity of in vitro cultured exoerythrocytic stages of Plasmodium berghei in indirect immunofluorescent or immunoperoxidase antibody tests. Am J Trop Med Hyg. 1983;32:24–30.PubMed Hollingdale MR, Leland P, Leef JL, Leef MF, Beaudoin RL. Serological reactivity of in vitro cultured exoerythrocytic stages of Plasmodium berghei in indirect immunofluorescent or immunoperoxidase antibody tests. Am J Trop Med Hyg. 1983;32:24–30.PubMed
21.
Zurück zum Zitat Vaughan AM, Mikolajczak SA, Wilson EM, Grompe M, Kaushansky A, Camargo N, et al. Complete Plasmodium falciparum liver-stage development in liver-chimeric mice. J Clin Invest. 2012;122:3618–28.CrossRefPubMedCentralPubMed Vaughan AM, Mikolajczak SA, Wilson EM, Grompe M, Kaushansky A, Camargo N, et al. Complete Plasmodium falciparum liver-stage development in liver-chimeric mice. J Clin Invest. 2012;122:3618–28.CrossRefPubMedCentralPubMed
22.
Zurück zum Zitat Oeuvray C, Bouharoun-Tayoun H, Gras-Masse H, Bottius E, Kaidoh T, Aikawa M, et al. Merozoite surface protein-3: a malaria protein inducing antibodies that promote Plasmodium falciparum killing by cooperation with blood monocytes. Blood. 1994;84:1594–602.PubMed Oeuvray C, Bouharoun-Tayoun H, Gras-Masse H, Bottius E, Kaidoh T, Aikawa M, et al. Merozoite surface protein-3: a malaria protein inducing antibodies that promote Plasmodium falciparum killing by cooperation with blood monocytes. Blood. 1994;84:1594–602.PubMed
23.
Zurück zum Zitat Zhou Y, Ramachandran V, Kumar KA, Westenberger S, Refour P, Zhou B, et al. Evidence-based annotation of the malaria parasite’s genome using comparative expression profiling. PLoS One. 2008;3:e1570.CrossRefPubMedCentralPubMed Zhou Y, Ramachandran V, Kumar KA, Westenberger S, Refour P, Zhou B, et al. Evidence-based annotation of the malaria parasite’s genome using comparative expression profiling. PLoS One. 2008;3:e1570.CrossRefPubMedCentralPubMed
24.
Zurück zum Zitat Tarun AS, Peng X, Dumpit RF, Ogata Y, Silva-Rivera H, Camargo N, et al. A combined transcriptome and proteome survey of malaria parasite liver stages. Proc Natl Acad Sci U S A. 2008;105:305–10.CrossRefPubMedCentralPubMed Tarun AS, Peng X, Dumpit RF, Ogata Y, Silva-Rivera H, Camargo N, et al. A combined transcriptome and proteome survey of malaria parasite liver stages. Proc Natl Acad Sci U S A. 2008;105:305–10.CrossRefPubMedCentralPubMed
25.
Zurück zum Zitat Silvie O, Franetich JF, Charrin S, Mueller MS, Siau A, Bodescot M, et al. A role for apical membrane antigen 1 during invasion of hepatocytes by Plasmodium falciparum sporozoites. J Biol Chem. 2004;279:9490–6.CrossRefPubMed Silvie O, Franetich JF, Charrin S, Mueller MS, Siau A, Bodescot M, et al. A role for apical membrane antigen 1 during invasion of hepatocytes by Plasmodium falciparum sporozoites. J Biol Chem. 2004;279:9490–6.CrossRefPubMed
26.
Zurück zum Zitat Schussek S, Trieu A, Apte SH, Sidney J, Sette A, Doolan DL. Immunization with apical membrane antigen 1 confers sterile infection-blocking immunity against Plasmodium sporozoite challenge in a rodent model. Infect Immun. 2013;81:3586–99.CrossRefPubMedCentralPubMed Schussek S, Trieu A, Apte SH, Sidney J, Sette A, Doolan DL. Immunization with apical membrane antigen 1 confers sterile infection-blocking immunity against Plasmodium sporozoite challenge in a rodent model. Infect Immun. 2013;81:3586–99.CrossRefPubMedCentralPubMed
27.
Zurück zum Zitat Kawabata Y, Udono H, Honma K, Ueda M, Mukae H, Kadota J, et al. Merozoite surface protein 1-specific immune response is protective against exoerythrocytic forms of Plasmodium yoelii. Infect Immun. 2002;70:6075–82.CrossRefPubMedCentralPubMed Kawabata Y, Udono H, Honma K, Ueda M, Mukae H, Kadota J, et al. Merozoite surface protein 1-specific immune response is protective against exoerythrocytic forms of Plasmodium yoelii. Infect Immun. 2002;70:6075–82.CrossRefPubMedCentralPubMed
28.
Zurück zum Zitat Szarfman A, Walliker D, McBride JS, Lyon JA, Quakyi IA, Carter R. Allelic forms of gp195, a major blood-stage antigen of Plasmodium falciparum, are expressed in liver stages. J Exp Med. 1988;167:231–6.CrossRefPubMed Szarfman A, Walliker D, McBride JS, Lyon JA, Quakyi IA, Carter R. Allelic forms of gp195, a major blood-stage antigen of Plasmodium falciparum, are expressed in liver stages. J Exp Med. 1988;167:231–6.CrossRefPubMed
29.
Zurück zum Zitat Suhrbier A, Holder AA, Wiser MF, Nicholas J, Sinden RE. Expression of the precursor of the major merozoite surface antigens during the hepatic stage of malaria. Am J Trop Med Hyg. 1989;40:351–5.PubMed Suhrbier A, Holder AA, Wiser MF, Nicholas J, Sinden RE. Expression of the precursor of the major merozoite surface antigens during the hepatic stage of malaria. Am J Trop Med Hyg. 1989;40:351–5.PubMed
30.
Zurück zum Zitat Borre MB, Dziegiel M, Hogh B, Petersen E, Rieneck K, Riley E, et al. Primary structure and localization of a conserved immunogenic Plasmodium falciparum glutamate rich protein (GLURP) expressed in both the preerythrocytic and erythrocytic stages of the vertebrate life cycle. Mol Biochem Parasitol. 1991;49:119–31.CrossRefPubMed Borre MB, Dziegiel M, Hogh B, Petersen E, Rieneck K, Riley E, et al. Primary structure and localization of a conserved immunogenic Plasmodium falciparum glutamate rich protein (GLURP) expressed in both the preerythrocytic and erythrocytic stages of the vertebrate life cycle. Mol Biochem Parasitol. 1991;49:119–31.CrossRefPubMed
31.
Zurück zum Zitat Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJ, van Gemert GJ, et al. Protection against a malaria challenge by sporozoite inoculation. N Engl J Med. 2009;361:468–77.CrossRefPubMed Roestenberg M, McCall M, Hopman J, Wiersma J, Luty AJ, van Gemert GJ, et al. Protection against a malaria challenge by sporozoite inoculation. N Engl J Med. 2009;361:468–77.CrossRefPubMed
33.
Zurück zum Zitat Walsh PS, Metzger DA, Higuchi R. Chelex 100 as a medium for simple extraction of DNA for PCR-based typing from forensic material. Biotechniques. 1991;10(4):506–13.PubMed Walsh PS, Metzger DA, Higuchi R. Chelex 100 as a medium for simple extraction of DNA for PCR-based typing from forensic material. Biotechniques. 1991;10(4):506–13.PubMed
34.
Zurück zum Zitat Snounou G, Pinheiro L, Goncalves A, Fonseca L, Dias F, Brown KN, et al. The importance of sensitive detection of malaria parasites in the human and insect hosts in epidemiological studies, as shown by the analysis of field samples from Guinea Bissau. Trans R Soc Trop Med Hyg. 1993;87:649–53.CrossRefPubMed Snounou G, Pinheiro L, Goncalves A, Fonseca L, Dias F, Brown KN, et al. The importance of sensitive detection of malaria parasites in the human and insect hosts in epidemiological studies, as shown by the analysis of field samples from Guinea Bissau. Trans R Soc Trop Med Hyg. 1993;87:649–53.CrossRefPubMed
35.
Zurück zum Zitat Bejon P, Warimwe G, Mackintosh CL, Mackinnon MJ, Kinyanjui SM, Musyoki JN, et al. Analysis of immunity to febrile malaria in children that distinguishes immunity from lack of exposure. Infect Immun. 2009;77:1917–23.CrossRefPubMedCentralPubMed Bejon P, Warimwe G, Mackintosh CL, Mackinnon MJ, Kinyanjui SM, Musyoki JN, et al. Analysis of immunity to febrile malaria in children that distinguishes immunity from lack of exposure. Infect Immun. 2009;77:1917–23.CrossRefPubMedCentralPubMed
36.
Zurück zum Zitat Bretscher MT, Maire N, Chitnis N, Felger I, Owusu-Agyei S, Smith T. The distribution of Plasmodium falciparum infection durations. Epidemics. 2011;3:109–18.CrossRefPubMed Bretscher MT, Maire N, Chitnis N, Felger I, Owusu-Agyei S, Smith T. The distribution of Plasmodium falciparum infection durations. Epidemics. 2011;3:109–18.CrossRefPubMed
37.
Zurück zum Zitat Okell LC, Bousema T, Griffin JT, Ouedraogo AL, Ghani AC, Drakeley CJ. Factors determining the occurrence of submicroscopic malaria infections and their relevance for control. Nat Commun. 2012;3:1237.CrossRefPubMedCentralPubMed Okell LC, Bousema T, Griffin JT, Ouedraogo AL, Ghani AC, Drakeley CJ. Factors determining the occurrence of submicroscopic malaria infections and their relevance for control. Nat Commun. 2012;3:1237.CrossRefPubMedCentralPubMed
38.
Zurück zum Zitat Bousema T, Okell L, Felger I, Drakeley C. Asymptomatic malaria infections: detectability, transmissibility and public health relevance. Nat Rev Microbiol. 2014;12:833–40.CrossRefPubMed Bousema T, Okell L, Felger I, Drakeley C. Asymptomatic malaria infections: detectability, transmissibility and public health relevance. Nat Rev Microbiol. 2014;12:833–40.CrossRefPubMed
39.
Zurück zum Zitat Laishram DD, Sutton PL, Nanda N, Sharma VL, Sobti RC, Carlton JM, et al. The complexities of malaria disease manifestations with a focus on asymptomatic malaria. Malar J. 2012;11:29.CrossRefPubMedCentralPubMed Laishram DD, Sutton PL, Nanda N, Sharma VL, Sobti RC, Carlton JM, et al. The complexities of malaria disease manifestations with a focus on asymptomatic malaria. Malar J. 2012;11:29.CrossRefPubMedCentralPubMed
40.
Zurück zum Zitat Goncalves BP, Huang CY, Morrison R, Holte S, Kabyemela E, Prevots DR, et al. Parasite burden and severity of malaria in Tanzanian children. N Engl J Med. 2014;370:1799–808.CrossRefPubMedCentralPubMed Goncalves BP, Huang CY, Morrison R, Holte S, Kabyemela E, Prevots DR, et al. Parasite burden and severity of malaria in Tanzanian children. N Engl J Med. 2014;370:1799–808.CrossRefPubMedCentralPubMed
41.
Zurück zum Zitat Proietti C, Verra F, Bretscher MT, Stone W, Kanoi BN, Balikagala B, et al. Influence of infection on malaria-specific antibody dynamics in a cohort exposed to intense malaria transmission in northern Uganda. Parasite Immunol. 2013;35:164–73.CrossRefPubMed Proietti C, Verra F, Bretscher MT, Stone W, Kanoi BN, Balikagala B, et al. Influence of infection on malaria-specific antibody dynamics in a cohort exposed to intense malaria transmission in northern Uganda. Parasite Immunol. 2013;35:164–73.CrossRefPubMed
42.
Zurück zum Zitat Weiss GE, Traore B, Kayentao K, Ongoiba A, Doumbo S, Doumtabe D, et al. The Plasmodium falciparum-specific human memory B cell compartment expands gradually with repeated malaria infections. PLoS Pathog. 2010;6:e1000912.CrossRefPubMedCentralPubMed Weiss GE, Traore B, Kayentao K, Ongoiba A, Doumbo S, Doumtabe D, et al. The Plasmodium falciparum-specific human memory B cell compartment expands gradually with repeated malaria infections. PLoS Pathog. 2010;6:e1000912.CrossRefPubMedCentralPubMed
43.
Zurück zum Zitat 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.CrossRefPubMedCentralPubMed 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.CrossRefPubMedCentralPubMed
44.
Zurück zum Zitat Njama-Meya D, Kamya MR, Dorsey G. Asymptomatic parasitaemia as a risk factor for symptomatic malaria in a cohort of Ugandan children. Trop Med Int Health. 2004;9:862–8.CrossRefPubMed Njama-Meya D, Kamya MR, Dorsey G. Asymptomatic parasitaemia as a risk factor for symptomatic malaria in a cohort of Ugandan children. Trop Med Int Health. 2004;9:862–8.CrossRefPubMed
45.
Zurück zum Zitat Ibison F, Olotu A, Muema DM, Mwacharo J, Ohuma E, Kimani D, et al. Lack of avidity maturation of merozoite antigen-specific antibodies with increasing exposure to Plasmodium falciparum amongst children and adults exposed to endemic malaria in Kenya. PLoS One. 2012;7:e52939.CrossRefPubMedCentralPubMed Ibison F, Olotu A, Muema DM, Mwacharo J, Ohuma E, Kimani D, et al. Lack of avidity maturation of merozoite antigen-specific antibodies with increasing exposure to Plasmodium falciparum amongst children and adults exposed to endemic malaria in Kenya. PLoS One. 2012;7:e52939.CrossRefPubMedCentralPubMed
46.
Zurück zum Zitat Kinyanjui SM, Mwangi T, Bull PC, Newbold CI, Marsh K. Protection against clinical malaria by heterologous immunoglobulin G antibodies against malaria-infected erythrocyte variant surface antigens requires interaction with asymptomatic infections. J Infect Dis. 2004;190:1527–33.CrossRefPubMed Kinyanjui SM, Mwangi T, Bull PC, Newbold CI, Marsh K. Protection against clinical malaria by heterologous immunoglobulin G antibodies against malaria-infected erythrocyte variant surface antigens requires interaction with asymptomatic infections. J Infect Dis. 2004;190:1527–33.CrossRefPubMed
47.
Zurück zum Zitat Soares IS, da Cunha MG, Silva MN, Souza JM, Del Portillo HA, Rodrigues MM. Longevity of naturally acquired antibody responses to the N- and C-terminal regions of Plasmodium vivax merozoite surface protein 1. Am J Trop Med Hyg. 1999;60:357–63.PubMed Soares IS, da Cunha MG, Silva MN, Souza JM, Del Portillo HA, Rodrigues MM. Longevity of naturally acquired antibody responses to the N- and C-terminal regions of Plasmodium vivax merozoite surface protein 1. Am J Trop Med Hyg. 1999;60:357–63.PubMed
48.
Zurück zum Zitat Ndungu FM, Olotu A, Mwacharo J, Nyonda M, Apfeld J, Mramba LK, et al. Memory B cells are a more reliable archive for historical antimalarial responses than plasma antibodies in no-longer exposed children. Proc Natl Acad Sci U S A. 2012;109:8247–52.CrossRefPubMedCentralPubMed Ndungu FM, Olotu A, Mwacharo J, Nyonda M, Apfeld J, Mramba LK, et al. Memory B cells are a more reliable archive for historical antimalarial responses than plasma antibodies in no-longer exposed children. Proc Natl Acad Sci U S A. 2012;109:8247–52.CrossRefPubMedCentralPubMed
49.
Zurück zum Zitat Manz RA, Hauser AE, Hiepe F, Radbruch A. Maintenance of serum antibody levels. Annu Rev Immunol. 2005;23:367–86.CrossRefPubMed Manz RA, Hauser AE, Hiepe F, Radbruch A. Maintenance of serum antibody levels. Annu Rev Immunol. 2005;23:367–86.CrossRefPubMed
50.
Zurück zum Zitat Kinyanjui SM, Conway DJ, Lanar DE, Marsh K. IgG antibody responses to Plasmodium falciparum merozoite antigens in Kenyan children have a short half-life. Malar J. 2007;6:82.CrossRefPubMedCentralPubMed Kinyanjui SM, Conway DJ, Lanar DE, Marsh K. IgG antibody responses to Plasmodium falciparum merozoite antigens in Kenyan children have a short half-life. Malar J. 2007;6:82.CrossRefPubMedCentralPubMed
51.
Zurück zum Zitat Egan AF, Morris J, Barnish G, Allen S, Greenwood BM, Kaslow DC, et al. Clinical immunity to Plasmodium falciparum malaria is associated with serum antibodies to the 19-kDa C-terminal fragment of the merozoite surface antigen, PfMSP-1. J Infect Dis. 1996;173:765–9.CrossRefPubMed Egan AF, Morris J, Barnish G, Allen S, Greenwood BM, Kaslow DC, et al. Clinical immunity to Plasmodium falciparum malaria is associated with serum antibodies to the 19-kDa C-terminal fragment of the merozoite surface antigen, PfMSP-1. J Infect Dis. 1996;173:765–9.CrossRefPubMed
52.
Zurück zum Zitat Dodoo D, Aikins A, Kusi KA, Lamptey H, Remarque E, Milligan P, et al. Cohort study of the association of antibody levels to AMA1, MSP119, MSP3 and GLURP with protection from clinical malaria in Ghanaian children. Malar J. 2008;7:142.CrossRefPubMedCentralPubMed Dodoo D, Aikins A, Kusi KA, Lamptey H, Remarque E, Milligan P, et al. Cohort study of the association of antibody levels to AMA1, MSP119, MSP3 and GLURP with protection from clinical malaria in Ghanaian children. Malar J. 2008;7:142.CrossRefPubMedCentralPubMed
53.
Zurück zum Zitat Olotu A, Lusingu J, Leach A, Lievens M, Vekemans J, Msham S, et al. Efficacy of RTS, S/AS01E malaria vaccine and exploratory analysis on anti-circumsporozoite antibody titres and protection in children aged 5–17 months in Kenya and Tanzania: a randomised controlled trial. Lancet Infect Dis. 2011;11:102–9.CrossRefPubMedCentralPubMed Olotu A, Lusingu J, Leach A, Lievens M, Vekemans J, Msham S, et al. Efficacy of RTS, S/AS01E malaria vaccine and exploratory analysis on anti-circumsporozoite antibody titres and protection in children aged 5–17 months in Kenya and Tanzania: a randomised controlled trial. Lancet Infect Dis. 2011;11:102–9.CrossRefPubMedCentralPubMed
54.
Zurück zum Zitat Crompton PD, Kayala MA, Traore B, Kayentao K, Ongoiba A, Weiss GE, et al. A prospective analysis of the Ab response to Plasmodium falciparum before and after a malaria season by protein microarray. Proc Natl Acad Sci U S A. 2010;107:6958–63.CrossRefPubMedCentralPubMed Crompton PD, Kayala MA, Traore B, Kayentao K, Ongoiba A, Weiss GE, et al. A prospective analysis of the Ab response to Plasmodium falciparum before and after a malaria season by protein microarray. Proc Natl Acad Sci U S A. 2010;107:6958–63.CrossRefPubMedCentralPubMed
55.
Zurück zum Zitat Offeddu V, Thathy V, Marsh K, Matuschewski K. Naturally acquired immune responses against Plasmodium falciparum sporozoites and liver infection. Int J Parasitol. 2012;42:535–48.CrossRefPubMed Offeddu V, Thathy V, Marsh K, Matuschewski K. Naturally acquired immune responses against Plasmodium falciparum sporozoites and liver infection. Int J Parasitol. 2012;42:535–48.CrossRefPubMed
56.
Zurück zum Zitat Hoffman SL, Oster CN, Plowe CV, Woollett GR, Beier JC, Chulay JD, et al. Naturally acquired antibodies to sporozoites do not prevent malaria: vaccine development implications. Science. 1987;237:639–42.CrossRefPubMed Hoffman SL, Oster CN, Plowe CV, Woollett GR, Beier JC, Chulay JD, et al. Naturally acquired antibodies to sporozoites do not prevent malaria: vaccine development implications. Science. 1987;237:639–42.CrossRefPubMed
57.
Zurück zum Zitat Bejon P, Lusingu J, Olotu A, Leach A, Lievens M, Vekemans J, et al. Efficacy of RTS, S/AS01E vaccine against malaria in children 5 to 17 months of age. N Engl J Med. 2008;359:2521–32.CrossRefPubMedCentralPubMed Bejon P, Lusingu J, Olotu A, Leach A, Lievens M, Vekemans J, et al. Efficacy of RTS, S/AS01E vaccine against malaria in children 5 to 17 months of age. N Engl J Med. 2008;359:2521–32.CrossRefPubMedCentralPubMed
58.
Zurück zum Zitat Abdulla S, Oberholzer R, Juma O, Kubhoja S, Machera F, Membi C, et al. Safety and immunogenicity of RTS, S/AS02D malaria vaccine in infants. N Engl J Med. 2008;359:2533–44.CrossRefPubMed Abdulla S, Oberholzer R, Juma O, Kubhoja S, Machera F, Membi C, et al. Safety and immunogenicity of RTS, S/AS02D malaria vaccine in infants. N Engl J Med. 2008;359:2533–44.CrossRefPubMed
59.
Zurück zum Zitat Osier FH, Mackinnon MJ, Crosnier C, Fegan G, Kamuyu G, Wanaguru M, et al. New antigens for a multicomponent blood-stage malaria vaccine. Sci Transl Med. 2014;6:247ra102.CrossRefPubMed Osier FH, Mackinnon MJ, Crosnier C, Fegan G, Kamuyu G, Wanaguru M, et al. New antigens for a multicomponent blood-stage malaria vaccine. Sci Transl Med. 2014;6:247ra102.CrossRefPubMed
60.
Zurück zum Zitat Meraldi V, Nebie I, Tiono AB, Diallo D, Sanogo E, Theisen M, et al. Natural antibody response to Plasmodium falciparum Exp-1, MSP-3 and GLURP long synthetic peptides and association with protection. Parasite Immunol. 2004;26:265–72.CrossRefPubMed Meraldi V, Nebie I, Tiono AB, Diallo D, Sanogo E, Theisen M, et al. Natural antibody response to Plasmodium falciparum Exp-1, MSP-3 and GLURP long synthetic peptides and association with protection. Parasite Immunol. 2004;26:265–72.CrossRefPubMed
61.
Zurück zum Zitat Ondigo BN, Park GS, Gose SO, Ho BM, Ochola LA, Ayodo GO, et al. Standardization and validation of a cytometric bead assay to assess antibodies to multiple Plasmodium falciparum recombinant antigens. Malar J. 2012;11:427.CrossRefPubMedCentralPubMed Ondigo BN, Park GS, Gose SO, Ho BM, Ochola LA, Ayodo GO, et al. Standardization and validation of a cytometric bead assay to assess antibodies to multiple Plasmodium falciparum recombinant antigens. Malar J. 2012;11:427.CrossRefPubMedCentralPubMed
62.
Zurück zum Zitat Jepsen MP, Roser D, Christiansen M, Olesen Larsen S, Cavanagh DR, Dhanasarnsombut K, et al. Development and evaluation of a multiplex screening assay for Plasmodium falciparum exposure. J Immunol Methods. 2012;384:62–70.CrossRefPubMed Jepsen MP, Roser D, Christiansen M, Olesen Larsen S, Cavanagh DR, Dhanasarnsombut K, et al. Development and evaluation of a multiplex screening assay for Plasmodium falciparum exposure. J Immunol Methods. 2012;384:62–70.CrossRefPubMed
63.
Zurück zum Zitat van der Heyde HC, Burns JM, Weidanz WP, Horn J, Gramaglia I, Nolan JP. Analysis of antigen-specific antibodies and their isotypes in experimental malaria. Cytometry A. 2007;71:242–50.CrossRefPubMed van der Heyde HC, Burns JM, Weidanz WP, Horn J, Gramaglia I, Nolan JP. Analysis of antigen-specific antibodies and their isotypes in experimental malaria. Cytometry A. 2007;71:242–50.CrossRefPubMed
Metadaten
Titel
Protection of Malian children from clinical malaria is associated with recognition of multiple antigens
verfasst von
Modibo Daou
Bourèma Kouriba
Nicolas Ouédraogo
Issa Diarra
Charles Arama
Yamoussa Keita
Sibiri Sissoko
Boucary Ouologuem
Seydou Arama
Teun Bousema
Ogobara K Doumbo
Robert W Sauerwein
Anja Scholzen
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Malaria Journal / Ausgabe 1/2015
Elektronische ISSN: 1475-2875
DOI
https://doi.org/10.1186/s12936-015-0567-9

Weitere Artikel der Ausgabe 1/2015

Malaria Journal 1/2015 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.