Skip to main content
Erschienen in: Malaria Journal 1/2016

Open Access 01.12.2016 | Research

Serological markers to measure recent changes in malaria at population level in Cambodia

verfasst von: Karen Kerkhof, Vincent Sluydts, Laura Willen, Saorin Kim, Lydie Canier, Somony Heng, Takafumi Tsuboi, Tho Sochantha, Siv Sovannaroth, Didier Ménard, Marc Coosemans, Lies Durnez

Erschienen in: Malaria Journal | Ausgabe 1/2016

Abstract

Background

Serological markers for exposure to different Plasmodium species have recently been used in multiplex immunoassays based on the Luminex technology. However, interpretation of the assay results requires consideration of the half-life of specific antibodies against these markers. Therefore, the aim of the present study was to document the half-life of malaria specific serological makers, as well as assessing the sensitivity of these markers to pick up recent changes in malaria exposure.

Methods

A recently developed multiplex immunoassay was used to measure the intensity of antibody (Ab) responses against 19 different Plasmodium specific antigens, covering different human malaria parasites and two vector saliva antigens. Therefore, 8439 blood samples from five cross-sectional surveys in Ratanakiri, Cambodia, were analysed. These involve a random selection from two selected surveys, and an additional set of blood samples of individuals that were randomly re-sampled three, four or five times. A generalized estimating equation model and linear regression models were fitted on log transformed antibody intensity data.

Results

Results showed that most (17/21) Ab-responses are higher in PCR positive than PCR negative individuals. Furthermore, these antibody-responses follow the same upward trend within each age group. Estimation of the half-lives showed differences between serological markers that reflect short- (seasonal) and long-term (year round) transmission trends. Ab levels declined significantly together with a decrease of PCR prevalence in a group of malaria endemic villages.

Conclusion

For Plasmodium falciparum, antibodies against LSA3.RE, GLURP and Pf.GLURP.R2 are most likely to be a reflexion of recent (range from 6 to 8 months) exposure in the Mekong Subregion. PvEBP is the only Plasmodium vivax Ag responding reasonably well, in spite of an estimated Ab half-life of more than 1 year. The use of Ab intensity data rather dichotomizing the continuous Ab-titre data (positive vs negative) will lead to an improved approach for serological surveillance.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12936-016-1576-z) contains supplementary material, which is available to authorized users.

Background

Despite a considerable decline, the malaria burden in the Greater Mekong Subregion is still high and has a major impact on public health, especially in some specific regions within countries [1]. With 68,000 malaria cases in 2013 and an annual parasite incidence rate of 4.6 per 1000 persons, malaria is the main single-cause infectious disease in Cambodia [2]. From 2000 to 2015, Cambodia has achieved a reduction of >75% in malaria case incidence [3], resulting in very low and heterogeneous malaria transmission clustered in hotspots and hotpops [2, 4, 5]. When aiming for malaria elimination, these focused areas of low malaria transmission pose considerable challenges for epidemiological surveillance and evaluation of control and elimination measures [6].
In malaria elimination areas, detection and surveillance of persisting malaria transmission is key for focussing interventions [7]. Traditional techniques for determining malaria transmission intensity include entomological inoculation rates (EIR) and parasite prevalence (PP) estimates [8]. However, in low transmission areas, the EIR and PP lack sensitivity, due to very low numbers of parasite-positive samples, both in mosquitoes and humans [9, 10].
Alternatively, serology is believed to be more informative in obtaining epidemiological information in malaria control programmes. In particular, serological markers can be used as a proxy for Plasmodium transmission intensity in low endemic areas [11] where parasite carriage is reduced and vector populations persevere [12]. More specifically this corresponds to the measurement of the force of infection (FOI), reflecting a rate at which susceptible individuals acquire an infection per year in a given area [9]. Where the EIR and PP mainly focus on the presence or absence of the infection [11] serology focuses on antibodies (Abs) that remain in the blood longer than the parasite. This is profitable in measuring host-parasite contact, and may provide information on recent or past malaria exposure, but only in case the half-lives of the Abs are known [11, 13, 14].
From an immunological point of view, partial protective immunity to malaria is built up after recurrent infections, typically over a period of several years of exposure. Hence, different types of memory are expected based on the frequency of short- (several months) and long-lived plasma cells (years) [15]. In areas of seasonal malaria transmission, it is assumed that short-lived plasma cells appear early in life and long-lived plasma cells later on [16], which can be used to understand the measurements of Ab half-lives [16]. It is important to take into account the different parasite life stages (skin, liver and early blood stages) as well [17]. Producing Abs against sporozoites or merozoites is challenging, due to the short time that sporozoites need to reach and penetrate liver cells, and for merozoites to reinvade erythrocytes. Strong Ab-responses are expressed against early blood stage Ags, which can be more useful in estimating cumulative exposure [12, 17]. Even though it is difficult to determine the half-life of Abs, some studies have already studied the Ab persistence [1820].
The aim of the current study is to document the half-life of IgG-Abs against 21 Ags from specific malaria parasites (Plasmodium falciparum, Plasmodium vivax, Plasmodium malariae) and saliva of vectors as well as assessing the sensitivity of the markers to pick up recent changes in malaria exposure. Methods to estimate the half-life of serological markers are still in its infancy, therefore, to reach this goal, three different approaches [13, 21] were applied directly on continuous Ab intensity values (Fig. 1) instead of using (arbitrary) thresholds to distinguish positives from negatives [22]. First, a comparison of the Ab-intensity between PCR-positive and PCR-negative samples was performed. Secondly, the half-life of the serological markers was documented by analysing sequential samples. Thirdly, the Ab-responses were examined in relation to the PCR endemicity. As there is no golden standard, the use of multiple approaches will improve the robustness of the results. As such, serological markers that perform best for all three approaches are considered to be most promising in reflecting recent exposure.

Methods

Malaria transmission patterns in the study area

Ratanakiri is the highest malaria endemic province of Cambodia. Malaria transmission occurs during the rainy season, between April/May and October/November. Over the years a major decline in malaria transmission had been observed in this province: In 2000, the recorded annual parasite incidence in Ratanakiri was 11/1000 inhabitants, whereas [23, 24] in 2012 an average incidence of 3.1/1000 inhabitants were reported [4]. The observed decline is attributed to the performance of the National Malaria Control Programme (high long-lasting insecticidal net coverage and improved case management) but also to environmental changes (deforestation) impacting the main vector (Anopheles dirus) [2].

Samples

Serum samples used for this study were collected during the cluster randomized trial organized within the MalaResT project, NCT01663831, that aimed to evaluate the use of topical repellents as added control measure to long-lasting insecticidal nets for prevention of malaria [4, 5] in Ratanakiri province, Cambodia. The project took place in the 98 most endemic clusters in Ratanakiri (88 single villages and 25 neighbouring villages grouped into 10 clusters) [25]. These 98 clusters were split into two arms, a control (long-lasting insecticidal nets) and an intervention arm (long-lasting insecticidal nets and the topical repellent picaridin). During 2 consecutive years (2012/2013) individuals were sampled by collecting two drops of blood on filter paper through a finger prick. To obtain baseline information on malaria prevalence at cluster level, the first survey (a pre-trial study) was conducted February 2012 (PCR prevalence 6.34%) [4, 25]. Four additional cross-sectional surveys were performed at the start of the rainy season in April/May (surveys 1, 2012 and 3, 2013) and 6 month later in October/November (surveys 2, 2012 and 4, 2013) with a PCR prevalence between 3 and 4% [25]. No differences in PCR prevalence were observed between the two study arms. PCR positive persons received a 3-day treatment with dihydroartemisinin plus piperaquine immediately following the national treatment guidelines [5, 25]. The immunoassay was performed on a total of 8438 out of 26,929 blood spot samples. Of this total, 6502 samples were randomly selected from survey 2 (3264 out of 4996 samples) and survey 4 (3238 out of 5431 samples). In addition, a total of 452 samples from the pre-trial and all four trial surveys were analysed as they were PCR positive for Plasmodium parasites. Furthermore, all samples of individuals that were randomly re-sampled three (430 individuals), four (36 individuals) or five times (10 individuals) during the entire MalaResT project were also included (Fig. 1). For the immunoassay blood spot filter papers were prepared by punching one disc of 6 mm (diameter), and eluted overnight in 160 μL of PBS-TBN [PBS-1% BSA-0.15% Tween, pH 7.4, Sigma-Aldrich (dilution 1:40)]. Just before use in the immunoassay, the eluted samples were diluted to 1:200 in PBS-CR, as previously described [26].

Antigens

The selection of Ags was based on their availability and tested in a previous study that focused on the implementation of the multiplex bead-based immunoassay [26]. The current assay includes eleven Plasmodium specific peptides, and eight Plasmodium specific recombinant proteins. These 19 Ags cover most stages of the life cycle of the parasite in the host (Table 1; Fig. 1). In addition, two peptides specific for the Anopheles gambiae saliva protein gSG6 (salivary gland 6) were included. All peptides were chemically synthesized with an added N-terminal cysteine residue and BSA (bovine serum albumin, Sigma-Aldrich, St. Louis, USA) [27] by GeneCust Europe (Dudelange, Luxembourg). The recombinant proteins were produced as described in Table 1.
Table 1
Overview of the antigens (peptides and recombinant proteins) used in this study
Antigens
Sequence (N-terminal to C-terminal)
g/mol
Life-cycle stages
Plasmodium species
Peptide or recombinant protein
Ref.
CSP
NANPNANPNANPNANPNVDPNVDPC
2257.67
Sporozoite
P. falciparum
Peptide
[27]
Pf13
C-terminal His-tag produced in E. coli
 
Sporozoite
P. falciparum
Recombinant protein
[69, 70]
STARP.R
STDNNNTKTISTDNNNTKTIC
2299.42
Sporozoite and liver stage
P. falciparum
Peptide
[27, 71]
SALSA2
NGKDDVKEEKKTNEKKDDGKTDKVQEKVLEKSPKC
4019.52
Sporozoite and liver stage
P. falciparum
Peptide
[27, 71]
SR11.1
EEVVEELIEEVIPEELVLC
2213.50
Sporozoite and liver stage
P. falciparum
Peptide
[27, 71]
LSA1.41
LAKEKLQEQQSDLEQERLAKEKLQEQQSDLEQERLAKEKE
KLQCERRAKEKLQEQQSDLEQRKADTKKC
5297.97
Liver stage
P. falciparum
Peptide
[27, 72, 73]
LSA1.J
ERRAKEKLQEQQSDLEQRKADTKKC
3046.43
Liver stage
P. falciparum
Peptide
[27, 71, 73]
LSA3.RE
VESVAPSVEESVAPSVEESVAENVEESVC
2991.20
Liver stage
P. falciparum
Peptide
[27, 71]
Pf.MSP1.19
Glutathione S-transferase (GST) fusion protein. C-terminal expressed in E. coli
 
Merozoite
P. falciparum
Recombinant protein
[38, 40]
GLURP
EDKNEKGQHEIVEVEEILC
2241.47
Trophozoite
P. falciparum
Peptide
[27, 71, 74]
Pf.GLURP.R2
C-terminal produced in E.coli
 
Trophozoite
P. falciparum
Recombinant protein
[27, 71, 75]
PvVK210.CSP
DGQPAGDRAAGQPAGDRADGQPAGDRADGQPAGC
3206.30
Sporozoite
P. vivax
Peptide
[71, 76, 77]
PvVK247.CSP
ANGAGNQPGANGAGNQPGANGAGNQPGANG AGNC
2905.95
Sporozoite
P. vivax
Peptide
[71, 76, 77]
PvCSP (chimera)
Soluble His-tag protein expressed in a wheat-germ cell free expression system
 
Sporozoite
P. vivax
Recombinant protein
[76, 77]
PvAMA1
  
Merozoite
P. vivax
Recombinant protein
[78, 79]
PvEBP
  
Merozoite
P. vivax
Recombinant protein
[80, 81]
PvDBP
  
Merozoite
P. vivax
Recombinant protein
[81]
Pv.MSP1.19
C-terminal produced in the baculovirus expression system
 
Merozoite
P. vivax
Recombinant protein
[38, 40, 48]
PmCSP
GNAAGNAAGNDAGNAAGNAAGNAAGNAAGNAAC
2358.37
Sporozoite
P. malariae
Peptide
[71]
SALIV1
EKVWVDRDNVYCGHLDCTRVATFC
2830.22
Salivary gland proteins
An gambiae
Peptide
[27, 82]
SALIV2
ATFKGERFCTLCDTRHFCECKETREPLC
3324.84
Salivary gland proteins
An gambiae
Peptide
[27, 82]
Ags are organized according to the Plasmodium species and the life-cycle stages in the human host

Covalent coupling of antigens to the beads/microspheres

Each Ag was covalently coupled at a concentration of 4 μg Ag/106 beads to 35 × 106 paramagnetic beads/beadset (MagPlex microspheres, Luminex Corp., Austin, TX, USA) [26]. BSA (Sigma-Aldrich, St Louis, USA) was coupled to an additional set of beads to serve as a background control. All beads and the BSA were mixed to prepare a microsphere working mixture. This mixture was then aliquoted and stored at 4 °C in portions of 500 μl per tube [26].

Bead-based immunoassay

The immunoassay was conducted as previously described [26]. First, a 500 μl aliquot of the microsphere working mixture was diluted to a final volume of 5000 μl (1:10) with a concentration of 1000 beads/Ag/well. From this microsphere working mixture 25 μl was added in each well of a 96-well plate followed by 50 μl of diluted serum sample (1:200) [26]. As a control for the immunoassay, a pool of negative sera (non-exposed European sera) was added to each plate in duplicate at a dilution of 1:100 in PBS-CR. A pool of positive sera containing sera from four individuals tested positive for P. falciparum and two for P. vivax [26] was added to each plate in duplicate at dilutions 1:100, 1:400 and 1:1600. For the washing steps PBS-TBN was used, and 100 μl/well of secondary Ab (R-phycoerythrin+-conjugated AffiniPure F(ab’)2 fragment of goat anti-human IgG, Jackson Immuno Research Laboratories) at a dilution of 1:500 was added [26]. All plates were analysed in a random order to minimize a bias. In a final step, beads were resuspended in 100 μl of 5% PBS-BSA, pH 7.4. In total, 216 plates were analysed and read by the MAGPIX® system with a minimum amount of 400 beads per spectral address. Results were represented as the median fluorescent intensity (MFI—Additional file 1) [26].

Statistical analysis

A scheme of the data analysis can be found in Fig. 1. Raw data were processed and analysed in R version 3.1.0. [28]. To assure the validity of the results of each plate a quality control was performed. Results were corrected for background signal by substracting the signal obtained with BSA-coupled beads (MFIBSA) from the median value of the Ag-coupled beads (MFIAg), defined by ∆MFI = MFIAg−MFIBSA [26]. The MFI-values of the high positive control pool samples and the percentage positivity (\(\frac{\Delta MFI Low \, positive \, control (Ag1)}{\Delta MFI High\, positive\, control (Ag1)}\) × 100%) from the low positive control (50% value of the high positive control) pool samples were plotted in Levey Jenning Charts. The high positive (100% value) is determined per Ag and based on the 1:100, 1:1400 and 1:1600 serum dilutions. The plates with samples outside the range of −2SD and +2SD were rejected and repeated (Additional file 2) [26].
Further differences between duplicate samples were explored using a quantile regression on an MA-plot using R package ‘OutlierD’ [29]. M is the difference between the duplicate samples and A is the average of the duplicate samples [30, 31]. Samples that fell outside the lower 25% and upper 25% quantiles of this MA-plot were rejected (Additional file 4). Hereafter, the means of the duplicates were calculated per sample of each Ag. All analyses were carried out on the natural logarithm (ln) of the raw MFI in accordance with Helb et al. [13].
As age influences Ab-responses [11, 32], age categories were created based on the work of Kusi et al. [8]. To explore whether the age categories defined were al biologically relevant in the study region, the (ln) MFI values were plotted against age (Additional file 5). Moreover, to account for multiple comparisons Bonferroni corrections were applied to each of the following approaches [33, 34].
Towards selecting markers that pick up contact with the parasite (recent of not recent), ln(MFI) values of PCR positive and PCR negative samples were compared per Plasmodium species. Generalized estimating equation (GEE) models were used (geeglm function in the R package ‘geepack’ [3537]), taking into account the within-cluster correlations that might exist due to the sampling design (village level clustering) [38]. The model also takes into account PCR positivity and age as factors allowing estimation of “population-averaged” effects [38, 39]. Different GEE-models were compared through ANOVA (a P value < 0.05 was considered significant) tests to select the most appropriate model per Ag: (1) a null model (intercept-only model), (2) a model with only PCR prevalence as a dependent variable, (3) a model with only age groups as a dependent variable, (4) a model with PCR prevalence and age groups as dependent variables (without interaction and (5) with interaction).
To estimate the half-life of the Abs after effective treatment of malaria, the presence of Abs in the same individuals (642 persons) followed up over time (±600 days) was examined [40]. Therefore, only individuals that were PCR positive in at least one survey were selected and the time of PCR positivity (and effective treatment) was taken as time point zero. This allows assessing the dynamics of the Abs against different Ags. Three linear models were compared per Ag through ANOVA tests: (1) a model with time since infection as dependent variable, (2) a model with time since infection and age groups as dependent variable (without interaction and (3) with interaction). Based on the slope (=λ) of the selected model, the half-life was estimated per serological marker (\({\text{t}}_{1/2} = \frac{{ - { \ln }(2)}}{\lambda }\)) [8, 41]. The 95% confidence intervals (95% CI) were estimated using the ‘confint’ function in R [28]. This analysis was performed on the systematically parasite negative individuals as well.
For determining the association between Ab-levels at population level and PCR-prevalence of asymptomatic Plasmodium infections, the ln(MFI) values were plotted against PCR prevalence at cluster level. Therefore, forest plots (multiplot function in the R package ‘grid’ [28] and forest plot function in R package ‘ggplot2’ [42]) were created on the cluster prevalence data from survey 2 and 4 per Plasmodium species. For P. falciparum and P. vivax this was performed on mono infections and for the An. gambiae saliva proteins on all infections (regardless of Plasmodium species). Each cluster was examined twice, once for survey 2 and once for survey 4. Then, the prevalence data were ordered from low to high prevalence (Additional file 7) and split into different groups representing low, medium and high prevalence (cut2 function in R package ‘Hmisc’ [43]). For each species, groups of clusters that showed an overlap between medium and high-level PCR prevalence were excluded, allowing a clear distinction between three groups (high-, medium- and low-level PCR prevalence). Thereafter, ln(MFI) values were age-adjusted as differences in Ab-levels can be found between different age categories. Finally, linear models taking into account cluster within survey as random effect (lmer function in R package ‘lme4’ [44]) were compared through ANOVA tests: (1) a null model (intercept-only model), (2) a model with PCR prevalence as a dependent variable, (3) a model with PCR prevalence and age as dependent variables (without interaction and (4) with interaction). The Incidence Rate Ratio (IRR) is corresponding to a ratio of Ab-levels estimated directly with the R-software.

Results

Although PmCSP was included in the battery of Ags, the results for PmCSP are not shown as the MFI signal (mean MFI < 600) did not surpass the background signal for the BSA.

Quality control

A total of 8654 field blood samples were analysed distributed over 108 96-well plates. Each plate was analysed in duplicate. After quality control, out of the 216 plates 30 plates were repeated (Additional file 2). The control samples fell within the linear range of the assay (Additional file 3). Samples were checked for uniformity across the measuring range of the assay by looking at the between plate variation of the samples (>96%). Thereafter, based on the quantile regression (Additional file 4), between 10 and 17% of the duplicate samples depending on the Ag were not consistent with each other and were excluded from the dataset. When determining the age categories, samples from individuals above 50 years were excluded, due to large variations in MFI values and a limited number of available samples (Additional file 5).

Comparison of MFI values between PCR positive and PCR negative samples

For 17 out of 20 Ags, the Ab levels were higher in PCR positive as compared to PCR negative blood samples (P < 0.0025, Bonferroni correction for 20 different tests) (Fig. 2). However, Ags PvVK210.CSP (P = 0.842), PvVK247.CSP (P = 0.557) and PvCSP (P = 0.009) did not show significant differences between the Ab levels within the age groups. As such, these three Ags were omitted from the figure. For 12 out of 20 Ags (CSP, Pf13, SALSA2, SR11.1, LSA1.J, LSA3.RE, Pf.MSP1.19, PvAMA1, PvDBP, Pv.MSP1.19, SALIV1 and SALIV2) there w0as no interaction between PCR status and age group, meaning that PCR-positive persons have significantly higher Ab levels for 12 Ags than PCR-negative persons (Fig. 2), within all age groups (model 4, P < 0.0038).
For five other Ags the model that includes the interaction between PCR-positivity and the age groups was selected as the best model, showing that there is a significant difference in MFI values between PCR positive and PCR negative samples (P < 0.01, Bonferroni correction for five tests), but that the magnitude of the difference depends on the age group. These Ags can be divided into two different groups. First PvEBP shows a significant difference (P < 0.01) between MFI values of PCR positive and PCR negative persons only in the younger age groups (2–5 and 6–15 years old), whereas for STARP.R, LSA1.41, GLURP and Pf.GLURP.R2 the differences in MFI values between PCR-positive and PCR-negative persons are significant (P < 0.01) in the older age groups (6–15 and 16–50 years old) only.

Ab-responses decline with days since infection, depending on age and serological marker

There is a lot of variation between the estimated half-life per serological marker, suggesting that it is possible to distinguish short (seasonal) and long-term (year round) transmission trends. The model with time since infection and age groups as dependent variables (without interaction) was selected as the best model for all 20 Ags, suggesting that age does not have an effect on the slope of the Ab decay, although the Ab titres at time point zero differ between age groups (Figs. 3, 4). For nine antigens (CSP, STARP.R, SALSA2, SR11.1, LSA1.J, LSA3.RE, GLURP, Pf.GLURP.R2 and PvEBP) a pronounced decay of the Ab titres was observed (Fig. 3). When comparing these graphs with the group of individuals being systematically negative, Ags LSA3.RE, GLURP and Pf.GLURP.R2 showed a much steeper decay in the group of individuals being PCR positive one time. Besides, the graphs of the PCR positive seems to flow into the negative groups, showing a difference between the intercept of >2. This outcome suggests that these Ags are most promising for reflecting short-term patterns in malaria transmission. Estimated half-lives, ranging between 6 months (176 [CI 119–338] days) for Pf.GLURP.R2 and more than 2 years for some P. vivax Ags, clearly depend on the serological marker used (Fig. 5; Additional file 6). Abs against five Ags (Pf.GLURP.R2, SR11.1, LSA3.RE, GLURP and Pf.MSP1.19) show estimated half-life values shorter than approximately 7.5 months, with estimated half-lives ranging from 6 months (176 [CI 119–338] days) until ~7.5 months (225 [CI 171–329] days). Estimated half-lives of Abs against STARP.R, Pf13, CSP, SALSA2 and SALIV.2 range between ~8.5 months (263 [CI 159–768] days) until ~1 year (373 [CI 284–546] days) and of Abs against LSA1.J, SALIV.1, LSA1.41, PvAMA1 and PvMSP1.19 from ~1.1 year (402 [CI 203–16,035] days) until ~1.6 year (597 [CI 350–2020] days). Remaining Abs show an estimated half-life of more than 2 years.
For this analysis for some Ags (CSP, STARP.R, SALSA2, SR11.1, LSA1.41 and Pf.MSP1.19) samples from young children (2–5 years) were not included, due to a limited amount of samples available. For the same reason the age group of 16–50 years old was excluded for LSA3.RE.

Intensity of the Ab-responses decreased together with the malaria exposure

To test whether Abs pick up recent malaria exposure in a population, the relation between Ab-titres (ln(MFI)) and the PCR prevalence of malaria in the province of Ratanakiri was explored. Therefore, the cluster PCR prevalence data from survey 2 and 4 was divided into different groups (forest plots: Additional file 7). In the forest plot, clusters with PCR prevalence between 3.5 and 7% were situated in the overlap between the medium- and high-level PCR prevalence and were not considered for present analysis, reflecting only those exhibiting high (7– >10%), medium (1–3.5%), and low (0– <1%) PCR-prevalence levels (Table 2). For Ags SALIV.1 and SALIV.2 all Plasmodium data (mono and mixed infections) were used, whereas the P. falciparum and P. vivax Ags were analysed species specific (only mono infections).
Table 2
Amount of cluster communities selected per species and per PCR prevalence level
PCR prevalence
P. falciparum
P. vivax
Plasmodium
High
S2
13
14
19
S4
3
9
10
Medium
S2
18
26
24
S4
21
23
24
Low
S2
52
39
26
S4
61
44
31
Cluster communities were ordered per survey exhibiting high-, medium- and low-levels of PCR prevalence. This was performed on PCR data from all Plasmodium species (mono and mixed infections), P. falciparum (mono infections) and P. vivax (mono infections)
For all 20 Ags, the model with PCR prevalence levels and age groups as dependent variables (without interaction) was selected. This model shows that when the PCR prevalence in groups of villages becomes lower, MFI levels also decrease (P < 0.0025; Additional file 8), within all age groups. Moreover, with decreasing endemicity Ab responses to the Ags presented in Fig. 6 show a more marked decline (drop in IRR between PCR prevalence level in respect to the reference group), as well as an increase in IRR with age. Biggest decline in IRR between PCR prevalence level was seen for Ags LSA3.RE, GLURP, Pf.GLURP.R2 and PvEBP, whereas the highest increase in IRR with respect to age was seen for Ags CSP, LSA1.41, LSA3.RE, GLURP, Pf.GLURP.R2 and PvEBP compared to the other Ags (Fig. 6).

Selection of informative antigens for recent infection are based on three criteria

To determine which Ags best reflect recent malaria exposure (seasonal) the following three criteria were used: (1) the outcome of the estimated Ab-responses based on the differences in ln(MFI) between PCR positive and PCR negative individuals, (2) the assessed half-lives and (3) the estimated Ab-responses in relation to high-, medium- and low levels of Plasmodium exposure (Table 3). Based on these criteria the Ags that are most likely to reflect recent malaria exposure to P. falciparum are LSA3.RE, GLURP and Pf.GLURP.R2. Among the seven Ags tested for P. vivax, PvEBP is the most prominent choice.
Table 3
Three criteria to select the most promising antigens
 
Antigens
Ags selection based on the three criteria
PCR+ vs PCR valuesa
Half-lives (PCR+)b
Differences in interceptc
Sensitivity by endemicityd
P. falciparum
CSP
+
+
+
+
Pf13
+
+
+
STARP.R
++
+
+
+
SALSA2
+
++
+
+
SR11.1
+
+
+
+
LSA1.41
++
+
++
LSA1.J
+
+
+
±
LSA3.RE
++
++
++
++
Pf.MSP1.19
+
GLURP
++
++
++
++
Pf.GLURP.R2
++
++
++
++
P. vivax
PvVK210.CSP
±
PvVK247.CSP
±
PvCSP
PvAMA1
±
PvEBP
++
++
++
PvDBP
Pv.MSP1.19
Vectors
SALIV1
SALIV2
+
Three criteria are used to select the most promising Ags for recent malaria infection. These criteria are based on: (1) the outcome of the estimated Ab-responses based on the differences in ln(MFI) between PCR positive and PCR negative individuals, (2) the assessed half-lives with the best Ab-responses of <7.5 months followed by Ab-responses between 8.5 months until 1 year and finally (3) the sensitivity to the level of malaria endemicity in communities
aDifference in ln(MFI) (ln-MFI PCR+ −ln-MFI PCR−); ++ (value > 1), + (value > 0.5),−(value < 0.5)
bHalf-lives estimated via linear regression models; ++ (half-life < 7.5 months), + (half-life between 8.5 months–1 year),− (half-life > 1 year)
cDifference in intercept between PCR+ and PCR− for half-life estimation; ++ (intercept difference > 2), + (intercept difference between 1 and 2), − (intercept difference < 1)
dSensitivity by endemicity estimated via linear regression models; ++ (IRR of PCR prev. decline with > 0.2, IRR increase by age with >4), + (IRR of PCR prev. decline with >0.1, IRR increase by age with >1)

Discussion

Several research groups recently developed multiplex serological assays for the detection of Abs against Plasmodium Ags [27, 38, 4548]. Previous studies combining short- and long-lived markers for Plasmodium have generated knowledge about past and recent changes in malaria transmission within communities [13, 40, 49]. This has led to an increasing interest in the use of serology in acquiring epidemiological information that can be implemented in malaria control programmes [9, 13, 40, 4955]. At this point, the majority of the studies and elimination programmes have focused in first instance on P. falciparum [13, 27, 38, 45, 46, 52, 56], which is easier to eliminate than P. vivax due to possible relapses in the latter one [57]. However, in contrast to other studies the current study was carried out in Southeast Asia (Cambodia) where all human malaria parasites are co-occurring [26, 58] and in which occupational and behavioural factors define the risk for malaria exposure [5961]. As such, adolescents and mainly adults performing plantation work and forest activities are the main risk groups for malaria infection [5962]. In this setting, insights into malaria transmission will be greatly enhanced by analysing age-adjusted Ab-responses directed against Plasmodium parasites including P. vivax. However, in this study age did not show any interactions on the outcomes.
The present study is to our knowledge the first study in Southeast Asia that uses multiplex serological measurements to document the half-life of IgG-Abs. Previous studies declare that Ab-levels follow a predictable pattern, in which every few months the Ab-level drops by half in the absence of re-infection [18, 41, 63]. To estimate this drop and apply those values in assessing the force of infection (FOI) in a study population, most researchers use a reversible catalytic conversion model [6, 40, 49, 51] on cross-sectional data. Such model is a simplification of a complex immunological process in which serological responses are converted to binary outcomes (seropositive or seronegative) through a threshold model [52]. Hence, these binary outcomes may lead to a loss of information due to the high range of MFI values within the seropositive group, and misclassification might occur because of subtle changes in the Ab-responses over time due to other infections or small laboratory variations [9]. Another limitation is that this model assumes a fixed sero-reversion rate, independent of age and transmission rate [51]. Therefore, some studies elaborated this model by comparing longitudinal- and cross-sectional data, which has led to large discrepancies in seroconversion and seroreversion rates [52]. Alternatively, linear regression models are used to provide information about the antibody acquisition [13, 21]. Helb et al. [13] and Yman et al. [12] estimated recent P. falciparum exposure at individual (longitudinal data) and population level (cross-sectional data). Studies based on longitudinal data mainly use approaches such as linear regression models [13, 20], generalized linear models (GLM) [64], or Ab-intensity models [12] to analyse Ab profiles.
A first criteria for selecting serological markers indicative for recent infection, is that the marker can at least pick up current infection. Therefore, the Ab intensity in Plasmodium infected people was compared to non-infected people. In agreement with results of previous studies, the present study shows that most Ab-responses (17/20) within each age group rise similarly by an infection. In contrast, in infected individuals Ab-responses to STARP.R, LSA1.41, GLURP, Pf.GLURP.R2 (among persons between 6 and 50 years) and PvEBP (among children from 2 to 15 years) seem to be influenced by age. [65]. The differences among children can be explained based on the role of immunological maturity-status [6], as children that acquire a malaria infection have the ability to boost their IgG titres, after which these Ab-titres decay again [66]. In this analysis it is important to take into consideration that the group of PCR-negatives is very large, and that PCR negative persons might have been infected in the six months between sampling periods. Moreover, for the PCR positive individuals, it is unknown when the infection was acquired.
In a next step, antibody decay rates (or half-lives) were estimated by using linear regression models that were fit on Ab-responses obtained from a cohort at different time points. In summary, the obtained results showed that the used serological markers could be divided in four groups with a wide variety of half-life estimates (Fig. 1; Table 3). Even though, limited information on the Abs persistence is available, some studies have previously estimated Ab half-lives [18, 20]. A study performed by Drakeley et al. [40] found a very long half-life of 49.8 years for Abs against Pv.MSP1.19, and Ondigo et al. [67] found half-lives from <1 year (3 Ags) to moderate (5–20 years for 3 Ags) and very long (>40 years for 5 Ags) by means of the threshold approach. Wipasa et al. [18] and Fowkes et al. [20] found a clearance of approximately 7.6 years [18] and 0.8–7.6 years [20], respectively, through linear regression models for the same Abs. The estimated Ab-clearance against merozoite Ags in the present study are best comparable with the latter results. However, these studies [18, 20] showed large ranges in 95% confidence intervals, which was also seen in the present study. The half-life estimates are in contrast with the underlying immunology showing the short period that merozoites need to reinvade erythrocytes. This is the short timespan in which the Abs have direct contact with the merozoite surface proteins, as this is the moment they are actually visible in the blood circulation [68]. A short clearance of Abs against merozoite Ags was also shown by Kinyanjui et al. [41] who estimated a clearance of 6–10 days and White et al. [66] that found a half-life of 7–72 days in children and 3–9 years for IgG Abs against AMA1, MSP1, MSP2 and CSP in infants, with a linear regression models. All these different outcomes suggest that using different model approaches (reversible catalytic conversion model and linear regression model) provide different outcomes, whereas with the linear regression models shorter half-lives were obtained. Moreover, large discrepancies in 95% confidence intervals were seen for some sporozoite related Ags as well. For these serological markers lower MFI values were observed [data not shown], resulting in larger 95% confidence intervals of Ab half-lives. This is not surprising as small numbers of sporozoites are only briefly present in the blood circulation to stimulate a sufficient production of sporozoite Abs, particularly, when situated in a region with low malaria transmission as the Mekong Subregion [17, 19]. Therefore, it has been previously suggested that the use of serological markers based on the sporozoite stage of malaria parasites might lead to an underestimation of the malaria endemicity in low transmission areas, while they possibly are more sensitive in hyper- and holo-endemic settings [19].
Finally, the cross-sectional data were examined to investigate whether the serological markers were capable of picking up current differences in malaria prevalence. Overall, for a lower PCR prevalence in a group of villages, the observed MFI levels were also lower within all age groups. This trend was similar as what was observed by Ambrosino et al. [27], who observed a rise in malaria specific Ab-responses with an increasing malaria exposure per village [27].
Based on the three different criteria i.e. (1) sensitive to infection in individuals, (2) sensitive to the level of endemicity in communities, and (3) a short half-life of IgG-Abs, the best serological markers (LSA3.RE, GLURP and Pf.GLURP.R2) that reflect recent P. falciparum exposure were identified. Moreover, Ags, CSP, Pf13, STARP.R, SALSA2, SR11.1, LSA1.41 and LSA1.J also seemed to respond well to a lesser extent. Therefore, these Ags should not be ruled out for future research in different settings. PvEBP was selected as the better candidate for P. vivax surveillance.

Conclusion

Given the broad utility of serology, identifying serological markers for recent exposure seems a worthwhile investment [13]. In summary, it appears that for P. falciparum LSA3.RE, GLURP and Pf.GLURP.R2 are most likely to be reflective for recent exposure, whereas PvEBP is the only Ag from P. vivax that responds reasonably well, in spite of a half-life of more than 1 year. Therefore, it is essential to explore other Ags from P. vivax. The only available P. malariae antigen did not provide a good response. It is remarkable that the best reflective Ags are mainly liver- and blood stage Ags, whereas in contrast to high endemic areas sporozoite level Ags are not useful in this setting (low-endemic). It is worth noting that all 20 Ags exhibit a direct link with the endemicity. In short, these Ags should certainly not be excluded for further in-depth analyses related to malaria control programmes. Another important aspect is that the use of Ab intensity data rather than dichotomizing the continuous Ab-titre data (positive vs negative) will lead to an improved approach for serological surveillance.

Authors’ contributions

Sample collection of the survey was performed by the CNM team (National Centre for Parasitology, Entomology and Malaria Control), the ITM Antwerp team and the Institut Pasteur du Cambodge team in Ratanakiri Province in Cambodia. The study design and literature research has been performed at the ITM by KK, LD and MC. Protocols were written by KK, based on protocols from Institut Pasteur de Madagascar (IVW), verified by LC and validated by DM. Screening of the blood spot samples was performed by KK, LW and SB. Selection of the pool of control sera and the high positive control dilutions was done by KK, LD and LC. Data entry and quality control was performed at the ITM by KK, LD, VS and LW. Analysis of the results was performed at the ITM by KK and VS and verified by LD and MC. The first draft of the manuscript was written by KK. All authors read and approved the final manuscript.

Acknowledgements

We would like to thank all colleagues at the ITM and IPC for all their support and contributions to fulfil this manuscript. We are extremely grateful that Odile Mercereau-Puijalon provided us recombinant proteins Pf13, PvDBP and PvEBP. We would like to thank Chris Drakeley for providing recombinant proteins PvMSP1-19 and PfMSP1-19 and Takafumi Tsuboi for providing recombinant proteins PvAMA1 and PvCSP-chimera. Furthermore, we would like to thank Prof Alfredo Mayor for the valuable discussions about this topic. Lastly, we are grateful to Myrthe Pareyn for her good comments and corrections during writing the manuscript.

Competing interests

The authors declare that they have no competing interests.

Availability of data and material

The datasets supporting the conclusions of this paper are included within the paper. Raw data may be obtained from the corresponding author on request.
The study protocol was reviewed and approved by the Cambodian National Ethics Committee on Health Research (Approval 265 NECHR), the Institutional Review Board of the Institute of Tropical Medicine Antwerp (Approval IRB/AB/ac/154) and the Ethics Committee of the University of Antwerp (Approval B300201112714). Gatekeepers provided informed written consent for the participation of their village. The survey participant or his/her parents or guardian provided informed written consent for individual participation.

Funding

This work is part of a larger project “MalaResT” funded by the Bill & Melinda Gates Foundation under the Global Health Grant number OPP1032354. This project aims to evaluate the effectiveness of topical repellents, in addition to long-lasting insecticidal nets, on malaria prevalence and incidence. Next, this work fits within the large-scale research project for the SOFI research program “MalaSpot” entitled ‘Eco-epidemiology of asymptomatic malaria hotspots, a prerequisite for malaria elimination?’. Furthermore, KK was supported by the association of Les Amis des Instituts Pasteur à Bruxelles.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Anhänge

Additional files

Literatur
1.
Zurück zum Zitat WHO. Strategy for malaria elimination in the Greater Mekong Subregion (2015–2030). Geneva: World Health Organization; 2015. WHO. Strategy for malaria elimination in the Greater Mekong Subregion (2015–2030). Geneva: World Health Organization; 2015.
2.
Zurück zum Zitat Maude RJ, Nguon C, Ly P, Bunkea T, Ngor P, de la Torre SEC, et al. Spatial and temporal epidemiology of clinical malaria in Cambodia 2004–2013. Malar J. 2014;13:385.CrossRefPubMedPubMedCentral Maude RJ, Nguon C, Ly P, Bunkea T, Ngor P, de la Torre SEC, et al. Spatial and temporal epidemiology of clinical malaria in Cambodia 2004–2013. Malar J. 2014;13:385.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat WHO. World Malaria Report 2015. Geneva: World Health Organization; 2015. WHO. World Malaria Report 2015. Geneva: World Health Organization; 2015.
4.
Zurück zum Zitat Sluydts V, Heng S, Coosemans M, Van Roey K, Gryseels C, Canier L, et al. Spatial clustering and risk factors of malaria infections in Ratanakiri Province, Cambodia. Malar J. 2014;13:387.CrossRefPubMedPubMedCentral Sluydts V, Heng S, Coosemans M, Van Roey K, Gryseels C, Canier L, et al. Spatial clustering and risk factors of malaria infections in Ratanakiri Province, Cambodia. Malar J. 2014;13:387.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Canier L, Khim N, Kim S, Sluydts V, Heng S, Dourng D, et al. An innovative tool for moving malaria PCR detection of parasite reservoir into the field. Malar J. 2013;12:405.CrossRefPubMedPubMedCentral Canier L, Khim N, Kim S, Sluydts V, Heng S, Dourng D, et al. An innovative tool for moving malaria PCR detection of parasite reservoir into the field. Malar J. 2013;12:405.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Cook J, Speybroeck N, Sochanta T, Somony H, Sokny M, Claes F, et al. Sero-epidemiological evaluation of changes in Plasmodium falciparum and Plasmodium vivax transmission patterns over the rainy season in Cambodia. Malar J. 2012;11:86.CrossRefPubMedPubMedCentral Cook J, Speybroeck N, Sochanta T, Somony H, Sokny M, Claes F, et al. Sero-epidemiological evaluation of changes in Plasmodium falciparum and Plasmodium vivax transmission patterns over the rainy season in Cambodia. Malar J. 2012;11:86.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Elliott SR, Fowkes FJI, Richards JS, Reiling L, Drew DR, Beeson JG. Research priorities for the development and implementation of serological tools for malaria surveillance. F1000Prime Rep. 2014;6:100.CrossRefPubMedPubMedCentral Elliott SR, Fowkes FJI, Richards JS, Reiling L, Drew DR, Beeson JG. Research priorities for the development and implementation of serological tools for malaria surveillance. F1000Prime Rep. 2014;6:100.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Kusi KA, Bosomprah S, Dodoo D, Kyei-Baafour E, Dickson EK, Mensah D, et al. Anti-sporozoite antibodies as alternative markers for malaria transmission intensity estimation. Malar J. 2014;13:103.CrossRefPubMedPubMedCentral Kusi KA, Bosomprah S, Dodoo D, Kyei-Baafour E, Dickson EK, Mensah D, et al. Anti-sporozoite antibodies as alternative markers for malaria transmission intensity estimation. Malar J. 2014;13:103.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Bretscher MT, Supargiyono S, Wijayanti MA, Nugraheni D, Widyastuti AN, Lobo NF, et al. Measurement of Plasmodium falciparum transmission intensity using serological cohort data from Indonesian schoolchildren. Malar J. 2013;12:21.CrossRefPubMedPubMedCentral Bretscher MT, Supargiyono S, Wijayanti MA, Nugraheni D, Widyastuti AN, Lobo NF, et al. Measurement of Plasmodium falciparum transmission intensity using serological cohort data from Indonesian schoolchildren. Malar J. 2013;12:21.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Cook J, Reid H, Iavro J, Kuwahata M, Taleo G, Clements A, et al. Using serological measures to monitor changes in malaria transmission in Vanuatu. Malar J. 2010;9:169.CrossRefPubMedPubMedCentral Cook J, Reid H, Iavro J, Kuwahata M, Taleo G, Clements A, et al. Using serological measures to monitor changes in malaria transmission in Vanuatu. Malar J. 2010;9:169.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Rogier E, Wiegand R, Moss D, Priest J, Angov E, Dutta S, et al. Multiple comparisons analysis of serological data from an area of low Plasmodium falciparum transmission. Malar J. 2015;14:436.CrossRefPubMedPubMedCentral Rogier E, Wiegand R, Moss D, Priest J, Angov E, Dutta S, et al. Multiple comparisons analysis of serological data from an area of low Plasmodium falciparum transmission. Malar J. 2015;14:436.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Yman V, White MT, Rono J, Arcà B, Osier FH, Troye-Blomberg M, et al. Antibody acquisition models: a new tool for serological surveillance of malaria transmission intensity. Sci Rep. 2016;6:19472.CrossRefPubMedPubMedCentral Yman V, White MT, Rono J, Arcà B, Osier FH, Troye-Blomberg M, et al. Antibody acquisition models: a new tool for serological surveillance of malaria transmission intensity. Sci Rep. 2016;6:19472.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Helb DA, Tetteh KKA, Felgner PL, Skinner J, Hubbard A, Arinaitwe E, et al. Novel serologic biomarkers provide accurate estimates of recent Plasmodium falciparum exposure for individuals and communities. Proc Natl Acad Sci USA. 2015;112:E4438–47.CrossRefPubMedPubMedCentral Helb DA, Tetteh KKA, Felgner PL, Skinner J, Hubbard A, Arinaitwe E, et al. Novel serologic biomarkers provide accurate estimates of recent Plasmodium falciparum exposure for individuals and communities. Proc Natl Acad Sci USA. 2015;112:E4438–47.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat van den Hoogen LL, Griffin JT, Cook J, Sepúlveda N, Corran P, Conway DJ, et al. Serology describes a profile of declining malaria transmission in Farafenni, The Gambia. Malar J. 2015;14:416.CrossRefPubMedPubMedCentral van den Hoogen LL, Griffin JT, Cook J, Sepúlveda N, Corran P, Conway DJ, et al. Serology describes a profile of declining malaria transmission in Farafenni, The Gambia. Malar J. 2015;14:416.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Andraud M, Lejeune O, Musoro JZ, Ogunjimi B, Beutels P. Living on three time scales: the dynamics of plasma cell and antibody populations illustrated for Hepatitis A virus. PLoS Comp Biol. 2012;8:e1002418.CrossRef Andraud M, Lejeune O, Musoro JZ, Ogunjimi B, Beutels P. Living on three time scales: the dynamics of plasma cell and antibody populations illustrated for Hepatitis A virus. PLoS Comp Biol. 2012;8:e1002418.CrossRef
16.
Zurück zum Zitat Hviid L, Barfod L, Fowkes FJI. Trying to remember: immunological B cell memory to malaria. Trends Parasitol. 2015;31:89–94.CrossRefPubMed Hviid L, Barfod L, Fowkes FJI. Trying to remember: immunological B cell memory to malaria. Trends Parasitol. 2015;31:89–94.CrossRefPubMed
17.
Zurück zum Zitat Crompton PD, Moebius J, Portugal S, Waisberg M, Hart G, Garver LS, et al. Malaria immunity in man and mosquito: insights into unsolved mysteries of a deadly infectious disease. Annu Rev Immunol. 2014;32:157.CrossRefPubMedPubMedCentral Crompton PD, Moebius J, Portugal S, Waisberg M, Hart G, Garver LS, et al. Malaria immunity in man and mosquito: insights into unsolved mysteries of a deadly infectious disease. Annu Rev Immunol. 2014;32:157.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Wipasa J, Suphavilai C, Okell LC, Cook J, Corran PH, Thaikla K, et al. Long-lived antibody and B Cell memory responses to the human malaria parasites, Plasmodium falciparum and Plasmodium vivax. PLoS Pathog. 2010;6:e1000770.CrossRefPubMedPubMedCentral Wipasa J, Suphavilai C, Okell LC, Cook J, Corran PH, Thaikla K, et al. Long-lived antibody and B Cell memory responses to the human malaria parasites, Plasmodium falciparum and Plasmodium vivax. PLoS Pathog. 2010;6:e1000770.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Wong J, Hamel MJ, Drakeley CJ, Kariuki S, Shi YP, Lal AA, et al. Serological markers for monitoring historical changes in malaria transmission intensity in a highly endemic region of Western Kenya, 1994–2009. Malar J. 2014;13:451.CrossRefPubMedPubMedCentral Wong J, Hamel MJ, Drakeley CJ, Kariuki S, Shi YP, Lal AA, et al. Serological markers for monitoring historical changes in malaria transmission intensity in a highly endemic region of Western Kenya, 1994–2009. Malar J. 2014;13:451.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Fowkes FJ, McGready R, Cross NJ, Hommel M, Simpson JA, Elliott SR, et al. New insights into acquisition, boosting, and longevity of immunity to malaria in pregnant women. J Infect Dis. 2012;206:1612.CrossRefPubMedPubMedCentral Fowkes FJ, McGready R, Cross NJ, Hommel M, Simpson JA, Elliott SR, et al. New insights into acquisition, boosting, and longevity of immunity to malaria in pregnant women. J Infect Dis. 2012;206:1612.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Stanisic DI, Fowkes FJI, Koinari M, Javati S, Lin E, Kiniboro B, et al. Acquisition of antibodies against Plasmodium falciparum merozoites and malaria immunity in young children and the influence of age, force of infection, and magnitude of response. Infect Immun. 2015;83:646.CrossRefPubMed Stanisic DI, Fowkes FJI, Koinari M, Javati S, Lin E, Kiniboro B, et al. Acquisition of antibodies against Plasmodium falciparum merozoites and malaria immunity in young children and the influence of age, force of infection, and magnitude of response. Infect Immun. 2015;83:646.CrossRefPubMed
22.
Zurück zum Zitat Hens N, Shkedy Z, Aerts M, Faes C, Van Damme P, Beutels P. Modeling infectious disease parameters based on serological and social contact data. Stat Biol Health Springer, New York. 2012;63:316. Hens N, Shkedy Z, Aerts M, Faes C, Van Damme P, Beutels P. Modeling infectious disease parameters based on serological and social contact data. Stat Biol Health Springer, New York. 2012;63:316.
23.
Zurück zum Zitat Trung HD, Van Bortel W, Sochanta T, Keokenchanh K, Quang NT, Cong LD, et al. Malaria transmission and major malaria vectors in different geographical areas of Southeast Asia. Trop Med Int Health. 2004;9:230.CrossRefPubMed Trung HD, Van Bortel W, Sochanta T, Keokenchanh K, Quang NT, Cong LD, et al. Malaria transmission and major malaria vectors in different geographical areas of Southeast Asia. Trop Med Int Health. 2004;9:230.CrossRefPubMed
24.
Zurück zum Zitat Ministry of Health. Ministry of Health strategic master plan. Cambodia: Phnom Penh; 2010. Ministry of Health. Ministry of Health strategic master plan. Cambodia: Phnom Penh; 2010.
25.
Zurück zum Zitat Sluydts V, Durnez L, Somony H, Gryseels C, Canier L, Kim S, et al. Efficacy of topical mosquito repellent (Picaridin) plus long-lasting insecticidal nets versus long-lasting insecticidal nets alone for control of malaria: a cluster randomised controlled trial. Lancet Infect Dis. 2016;16:1169–77.CrossRefPubMed Sluydts V, Durnez L, Somony H, Gryseels C, Canier L, Kim S, et al. Efficacy of topical mosquito repellent (Picaridin) plus long-lasting insecticidal nets versus long-lasting insecticidal nets alone for control of malaria: a cluster randomised controlled trial. Lancet Infect Dis. 2016;16:1169–77.CrossRefPubMed
26.
Zurück zum Zitat Kerkhof K, Canier L, Kim S, Heng S, Sochantha T, Sovannaroth S, et al. Implementation and application of a multiplex assay to detect malaria-specific antibodies: a promising tool for assessing malaria transmission in Southeast Asian pre-elimination areas. Malar J. 2015;14:338.CrossRefPubMedPubMedCentral Kerkhof K, Canier L, Kim S, Heng S, Sochantha T, Sovannaroth S, et al. Implementation and application of a multiplex assay to detect malaria-specific antibodies: a promising tool for assessing malaria transmission in Southeast Asian pre-elimination areas. Malar J. 2015;14:338.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Ambrosino E, Dumoulin C, Orlandi-Pradines E, Remoue F, Toure-Baldé A, Tall A, et al. A multiplex assay for the simultaneous detection of antibodies against 15 Plasmodium falciparum and Anopheles gambiae saliva antigens. Malar J. 2010;9:317.CrossRefPubMedPubMedCentral Ambrosino E, Dumoulin C, Orlandi-Pradines E, Remoue F, Toure-Baldé A, Tall A, et al. A multiplex assay for the simultaneous detection of antibodies against 15 Plasmodium falciparum and Anopheles gambiae saliva antigens. Malar J. 2010;9:317.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Cho H, Kim YJ, Jung HJ, Lee SW, Lee JW. OutlierD: an R package for outlier detection using quantile regression on mass spectrometry data. Bioinformatics. 2008;24:882.CrossRefPubMed Cho H, Kim YJ, Jung HJ, Lee SW, Lee JW. OutlierD: an R package for outlier detection using quantile regression on mass spectrometry data. Bioinformatics. 2008;24:882.CrossRefPubMed
31.
Zurück zum Zitat Eo SH, Pak D, Choi J, Cho H. Outlier detection using projection quantile regression for mass spectrometry data with low replication. BMC Res Notes. 2012;5:236.CrossRefPubMedPubMedCentral Eo SH, Pak D, Choi J, Cho H. Outlier detection using projection quantile regression for mass spectrometry data with low replication. BMC Res Notes. 2012;5:236.CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Scholzen A, Sauerwein RW. How malaria modulates memory: activation and dysregulation of B cells in Plasmodium infection. Trends Parasitol. 2013;29:252.CrossRefPubMed Scholzen A, Sauerwein RW. How malaria modulates memory: activation and dysregulation of B cells in Plasmodium infection. Trends Parasitol. 2013;29:252.CrossRefPubMed
34.
Zurück zum Zitat Orlandi-Pradines E, Penhoat K, Durand C, Pons C, Bay C, Pradines B, et al. Antibody responses to several malaria pre-erythrocytic antigens as a marker of malaria exposure among travelers. Am J Trop Med Hyg. 2006;74:979.PubMed Orlandi-Pradines E, Penhoat K, Durand C, Pons C, Bay C, Pradines B, et al. Antibody responses to several malaria pre-erythrocytic antigens as a marker of malaria exposure among travelers. Am J Trop Med Hyg. 2006;74:979.PubMed
35.
Zurück zum Zitat Højsgaard S, Halekoh U, Yan J. The R Package geepack for generalized estimating equations. J Stat Softw. 2006;15:1. Højsgaard S, Halekoh U, Yan J. The R Package geepack for generalized estimating equations. J Stat Softw. 2006;15:1.
36.
37.
Zurück zum Zitat Yan J. Geepack: yet another package for generalized estimating equations. R-News. 2002;2:12. Yan J. Geepack: yet another package for generalized estimating equations. R-News. 2002;2:12.
38.
Zurück zum Zitat Khaireh BA, Briolant S, Pascual A, Mokrane M, Machault V, Travaillé C, et al. Plasmodium vivax and Plasmodium falciparum infections in the Republic of Djibouti: evaluation of their prevalence and potential determinants. Malar J. 2012;11:395.CrossRefPubMedPubMedCentral Khaireh BA, Briolant S, Pascual A, Mokrane M, Machault V, Travaillé C, et al. Plasmodium vivax and Plasmodium falciparum infections in the Republic of Djibouti: evaluation of their prevalence and potential determinants. Malar J. 2012;11:395.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Hu FB, Goldberg J, Hedeker D, Flay BR, Pentz MA. Comparison of population-averaged and subject-specific approaches for analyzing repeated binary outcomes. Am J Epidemiol. 1998;147:694.CrossRefPubMed Hu FB, Goldberg J, Hedeker D, Flay BR, Pentz MA. Comparison of population-averaged and subject-specific approaches for analyzing repeated binary outcomes. Am J Epidemiol. 1998;147:694.CrossRefPubMed
40.
Zurück zum Zitat Drakeley CJ, Corran PH, Coleman PG, Tongren JE, McDonald SLR, Carneiro I, 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.CrossRefPubMedPubMedCentral Drakeley CJ, Corran PH, Coleman PG, Tongren JE, McDonald SLR, Carneiro I, 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.CrossRefPubMedPubMedCentral
41.
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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Frank E, Harrell JR, Dupont C, et al. Hmisc: Harrell Miscellaneous. 2015. R Package version 316-0. Accessed 2015. Frank E, Harrell JR, Dupont C, et al. Hmisc: Harrell Miscellaneous. 2015. R Package version 316-0. Accessed 2015.
44.
Zurück zum Zitat Bates D, Martin M. Fitting linear mixed-effects models using lme4. J Stat Softw. 2015;67:1.CrossRef Bates D, Martin M. Fitting linear mixed-effects models using lme4. J Stat Softw. 2015;67:1.CrossRef
45.
Zurück zum Zitat Sarr JB, Orlandi-Pradines E, Fortin S, Sow C, Cornelie S, Rogerie F, et al. Assessment of exposure to Plasmodium falciparum transmission in a low endemicity area by using multiplex fluorescent microsphere-based serological assays. Parasit Vectors. 2011;4:212.CrossRefPubMedPubMedCentral Sarr JB, Orlandi-Pradines E, Fortin S, Sow C, Cornelie S, Rogerie F, et al. Assessment of exposure to Plasmodium falciparum transmission in a low endemicity area by using multiplex fluorescent microsphere-based serological assays. Parasit Vectors. 2011;4:212.CrossRefPubMedPubMedCentral
46.
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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Fouda GG, Leke RFG, Long C, Druilhe P, Zhou A, Taylor DW, et al. Multiplex assay for simultaneous measurement of antibodies to multiple Plasmodium falciparum antigens. Clin Vaccine Immunol. 2006;13:1307.CrossRefPubMedPubMedCentral Fouda GG, Leke RFG, Long C, Druilhe P, Zhou A, Taylor DW, et al. Multiplex assay for simultaneous measurement of antibodies to multiple Plasmodium falciparum antigens. Clin Vaccine Immunol. 2006;13:1307.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Fernandez-Becerra C, Sanz S, Brucet M, Stanisic DI, Alves FP, Camargo EP. Naturally-acquired humoral immune responses against the N- and C-termini of the Plasmodium vivax MSP1 protein in endemic regions of Brazil and Papua New Guinea using a multiplex assay. Malar J. 2010;9:29.CrossRefPubMedPubMedCentral Fernandez-Becerra C, Sanz S, Brucet M, Stanisic DI, Alves FP, Camargo EP. Naturally-acquired humoral immune responses against the N- and C-termini of the Plasmodium vivax MSP1 protein in endemic regions of Brazil and Papua New Guinea using a multiplex assay. Malar J. 2010;9:29.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Corran P, Coleman P, Riley E, Drakeley C. Serology: a robust indicator of malaria transmission intensity? Trends Parasitol. 2007;23:575.CrossRefPubMed Corran P, Coleman P, Riley E, Drakeley C. Serology: a robust indicator of malaria transmission intensity? Trends Parasitol. 2007;23:575.CrossRefPubMed
50.
Zurück zum Zitat Drakeley C, Cook J. Potential contribution of sero-epidemiological analysis for monitoring malaria control and elimination: historical and current perspectives. Adv Parasitol. 2009;69:299.CrossRefPubMed Drakeley C, Cook J. Potential contribution of sero-epidemiological analysis for monitoring malaria control and elimination: historical and current perspectives. Adv Parasitol. 2009;69:299.CrossRefPubMed
51.
Zurück zum Zitat Stewart L, Gosling R, Griffin J, Gesase S, Campo J, Hashim R, et al. Rapid assessment of malaria transmission using age-specific sero-conversion rates. PLoS ONE. 2009;4:e6083.CrossRefPubMedPubMedCentral Stewart L, Gosling R, Griffin J, Gesase S, Campo J, Hashim R, et al. Rapid assessment of malaria transmission using age-specific sero-conversion rates. PLoS ONE. 2009;4:e6083.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Arnold BF, Priest JW, Hamlin KL, Moss DM, Colford JM, Lammie PJ. Serological measures of malaria transmission in Haiti: comparison of longitudinal and cross-sectional methods. PLoS ONE. 2014;9:e93684.CrossRefPubMedPubMedCentral Arnold BF, Priest JW, Hamlin KL, Moss DM, Colford JM, Lammie PJ. Serological measures of malaria transmission in Haiti: comparison of longitudinal and cross-sectional methods. PLoS ONE. 2014;9:e93684.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Cook J, Kleinschmidt I, Schwabe C, Nseng G, Bousema T, Corran PH, et al. Serological markers suggest heterogeneity of effectiveness of malaria control interventions on Bioko Island, equatorial Guinea. PLoS ONE. 2011;6:e25137.CrossRefPubMedPubMedCentral Cook J, Kleinschmidt I, Schwabe C, Nseng G, Bousema T, Corran PH, et al. Serological markers suggest heterogeneity of effectiveness of malaria control interventions on Bioko Island, equatorial Guinea. PLoS ONE. 2011;6:e25137.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Badu K, Afrane YA, Larbi J, Stewart VA, Waitumbi J, Angov E, et al. Marked variation in MSP-119 antibody responses to malaria in western Kenyan highlands. BMC Infect Dis. 2012;12:50.CrossRefPubMedPubMedCentral Badu K, Afrane YA, Larbi J, Stewart VA, Waitumbi J, Angov E, et al. Marked variation in MSP-119 antibody responses to malaria in western Kenyan highlands. BMC Infect Dis. 2012;12:50.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat von Fricken ME, Weppelmann TA, Lam B, Eaton WT, Schick L, Masse R, et al. Age-specific malaria seroprevalence rates: a cross-sectional analysis of malaria transmission in the Ouest and Sud-Est departments of Haiti. Malar J. 2014;13:361.CrossRef von Fricken ME, Weppelmann TA, Lam B, Eaton WT, Schick L, Masse R, et al. Age-specific malaria seroprevalence rates: a cross-sectional analysis of malaria transmission in the Ouest and Sud-Est departments of Haiti. Malar J. 2014;13:361.CrossRef
56.
Zurück zum Zitat Cham GKK, Kurtis J, Lusingu J, Theander TG, Jensen ATR, Turner L. A semi-automated multiplex high-throughput assay for measuring IgG antibodies against Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) domains in small volumes of plasma. Malar J. 2008;7:108.CrossRefPubMedPubMedCentral Cham GKK, Kurtis J, Lusingu J, Theander TG, Jensen ATR, Turner L. A semi-automated multiplex high-throughput assay for measuring IgG antibodies against Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) domains in small volumes of plasma. Malar J. 2008;7:108.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat Gething PW, Elyazar IRF, Moyes CL, Smith DL, Battle KE, et al. A long neglected world malaria map: Plasmodium vivax endemicity in 2010. PLoS Negl Trop Dis. 2012;6:e1814.CrossRefPubMedPubMedCentral Gething PW, Elyazar IRF, Moyes CL, Smith DL, Battle KE, et al. A long neglected world malaria map: Plasmodium vivax endemicity in 2010. PLoS Negl Trop Dis. 2012;6:e1814.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Khim N, Siv S, Kim S, Mueller T, Fleischmann E, Singh B, et al. Plasmodium knowlesi infection in humans, Cambodia, 2007–2010. Emerg Infect Dis. 2011;17:1900.CrossRefPubMedPubMedCentral Khim N, Siv S, Kim S, Mueller T, Fleischmann E, Singh B, et al. Plasmodium knowlesi infection in humans, Cambodia, 2007–2010. Emerg Infect Dis. 2011;17:1900.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Sturrock HJW, Hsiang MS, Cohen JM, Smith DL, Greenhouse B, Bousema T, et al. Targeting asymptomatic malaria infections: active surveillance in control and elimination. PLoS Med. 2013;10:1.CrossRef Sturrock HJW, Hsiang MS, Cohen JM, Smith DL, Greenhouse B, Bousema T, et al. Targeting asymptomatic malaria infections: active surveillance in control and elimination. PLoS Med. 2013;10:1.CrossRef
60.
Zurück zum Zitat Smith C, Whittaker M. Malaria elimination without stigmatization: a note of caution about the use of terminology in elimination settings. Malar J. 2014;13:377.CrossRefPubMedPubMedCentral Smith C, Whittaker M. Malaria elimination without stigmatization: a note of caution about the use of terminology in elimination settings. Malar J. 2014;13:377.CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat Cotter C, Sturrock HJ, Hsiang MS, Liu J, Phillips AA, Hwang J, et al. The changing epidemiology of malaria elimination: new strategies for new challenges. Lancet. 2013;382:900.CrossRefPubMed Cotter C, Sturrock HJ, Hsiang MS, Liu J, Phillips AA, Hwang J, et al. The changing epidemiology of malaria elimination: new strategies for new challenges. Lancet. 2013;382:900.CrossRefPubMed
62.
Zurück zum Zitat Incardona S, Vong S, Chiv L, Lim P, Nhem S, Sem R, et al. Large-scale malaria survey in Cambodia: novel insights on species distribution and risk factors. Malar J. 2007;6:37.CrossRefPubMedPubMedCentral Incardona S, Vong S, Chiv L, Lim P, Nhem S, Sem R, et al. Large-scale malaria survey in Cambodia: novel insights on species distribution and risk factors. Malar J. 2007;6:37.CrossRefPubMedPubMedCentral
63.
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.CrossRefPubMedPubMedCentral 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.CrossRefPubMedPubMedCentral
64.
Zurück zum Zitat Koffi D, Touré AO, Varela ML, Vigan-Womas I, Béourou S, Brou S, et al. Analysis of antibody profiles in symptomatic malaria in three sentinel sites of Ivory Coast by using multiplex, fluorescent, magnetic, bead-based serological assay (MAGPIX™). Malar J. 2015;14:509.CrossRefPubMedPubMedCentral Koffi D, Touré AO, Varela ML, Vigan-Womas I, Béourou S, Brou S, et al. Analysis of antibody profiles in symptomatic malaria in three sentinel sites of Ivory Coast by using multiplex, fluorescent, magnetic, bead-based serological assay (MAGPIX™). Malar J. 2015;14:509.CrossRefPubMedPubMedCentral
65.
Zurück zum Zitat De Souza JB, Todd J, Krishegowda G, Gowda DC, Kwiatkowski D, Riley EM. Prevalence and boosting of antibodies to Plasmodium falciparum glycosylphosphatidylinositols and evaluation of their association with protection from mild and severe clinical malaria. Infect Immun. 2002;70:5045.CrossRefPubMedPubMedCentral De Souza JB, Todd J, Krishegowda G, Gowda DC, Kwiatkowski D, Riley EM. Prevalence and boosting of antibodies to Plasmodium falciparum glycosylphosphatidylinositols and evaluation of their association with protection from mild and severe clinical malaria. Infect Immun. 2002;70:5045.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat White MT, Griffin JT, Akpogheneta O, Conway DJ, Koram KA, Riley EM, et al. Dynamics of the antibody response to Plasmodium falciparum infection in African children. J Infect Dis. 2014;210:1115.CrossRefPubMed White MT, Griffin JT, Akpogheneta O, Conway DJ, Koram KA, Riley EM, et al. Dynamics of the antibody response to Plasmodium falciparum infection in African children. J Infect Dis. 2014;210:1115.CrossRefPubMed
67.
Zurück zum Zitat Ondigo BN, Hodges JS, Ireland KF, Magak NG, Lanar DE, Dutta S, et al. Estimation of recent and long-term malaria transmission in a population by antibody testing to multiple Plasmodium falciparum antigens. J Infect Dis. 2014;7:1123–32.CrossRef Ondigo BN, Hodges JS, Ireland KF, Magak NG, Lanar DE, Dutta S, et al. Estimation of recent and long-term malaria transmission in a population by antibody testing to multiple Plasmodium falciparum antigens. J Infect Dis. 2014;7:1123–32.CrossRef
68.
Zurück zum Zitat Zenonos ZA, Rayner JC, Wright GJ. Towards a comprehensive Plasmodium falciparum merozoite cell surface and secreted recombinant protein library. Malar J. 2014;13:93.CrossRefPubMedPubMedCentral Zenonos ZA, Rayner JC, Wright GJ. Towards a comprehensive Plasmodium falciparum merozoite cell surface and secreted recombinant protein library. Malar J. 2014;13:93.CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat Sutherland CJ. Surface antigens of Plasmodium falciparum gametocytes—a new class of transmission-blocking vaccine targets? Mol Biochem Parasitol. 2009;166:93.CrossRefPubMed Sutherland CJ. Surface antigens of Plasmodium falciparum gametocytes—a new class of transmission-blocking vaccine targets? Mol Biochem Parasitol. 2009;166:93.CrossRefPubMed
70.
Zurück zum Zitat Vigan-Womas I, Guillotte M, Juillerat A, Vallieres C, Lewit-Bentley A, Tall A, et al. Allelic diversity of the Plasmodium falciparum erythrocyte membrane protein 1 entails variant-specific red cell surface epitopes. PLoS ONE. 2011;6:e16544.CrossRefPubMedPubMedCentral Vigan-Womas I, Guillotte M, Juillerat A, Vallieres C, Lewit-Bentley A, Tall A, et al. Allelic diversity of the Plasmodium falciparum erythrocyte membrane protein 1 entails variant-specific red cell surface epitopes. PLoS ONE. 2011;6:e16544.CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat Fidock A, Gras-Masse H, Lepers J, Brahimi K, Benmohamed L, Mellouk S, et al. Plasmodium falciparum liver stage antigen-1 is well conserved and contains potent B and T cell determinants. J Immunol. 1994;153:190.PubMed Fidock A, Gras-Masse H, Lepers J, Brahimi K, Benmohamed L, Mellouk S, et al. Plasmodium falciparum liver stage antigen-1 is well conserved and contains potent B and T cell determinants. J Immunol. 1994;153:190.PubMed
74.
Zurück zum Zitat Theisen M, Vuust J, Gottschau A, Jepsen S, Høgh B. Antigenicity and immunogenicity of recombinant glutamate-rich protein of Plasmodium falciparum expressed in Escherichia coli. Clin Diagn Lab Immunol. 1995;2:30.PubMedPubMedCentral Theisen M, Vuust J, Gottschau A, Jepsen S, Høgh B. Antigenicity and immunogenicity of recombinant glutamate-rich protein of Plasmodium falciparum expressed in Escherichia coli. Clin Diagn Lab Immunol. 1995;2:30.PubMedPubMedCentral
75.
Zurück zum Zitat Theisen M, Soe S, Jessing SG, Meng L, Oeuvray C, Druilhe P, et al. Identification of a major B-cell epitope of the Plasmodium falciparum glutamate-rich protein (GLURP), targeted by human antibodies mediating parasite killing. Vaccine. 2001;19:204.CrossRef Theisen M, Soe S, Jessing SG, Meng L, Oeuvray C, Druilhe P, et al. Identification of a major B-cell epitope of the Plasmodium falciparum glutamate-rich protein (GLURP), targeted by human antibodies mediating parasite killing. Vaccine. 2001;19:204.CrossRef
76.
Zurück zum Zitat Henry-Halldin CN, Sepe D, Susapu M, McNamara DT, Bockarie M, King CL, et al. High-throughput molecular diagnosis of circumsporozoite variants VK210 and VK247 detects complex Plasmodium vivax infections in malaria endemic populations in Papua New Guinea. Infect Genet Evol. 2011;11:391.CrossRefPubMed Henry-Halldin CN, Sepe D, Susapu M, McNamara DT, Bockarie M, King CL, et al. High-throughput molecular diagnosis of circumsporozoite variants VK210 and VK247 detects complex Plasmodium vivax infections in malaria endemic populations in Papua New Guinea. Infect Genet Evol. 2011;11:391.CrossRefPubMed
77.
Zurück zum Zitat Cheng Y, Ito D, Sattabongkot J, Lim CS, Kong DH, Ha KS, et al. Serological responses to a soluble recombinant chimeric Plasmodium vivax circumsporozoite protein in VK210 and VK247 population. Malar J. 2013;12:323.CrossRefPubMedPubMedCentral Cheng Y, Ito D, Sattabongkot J, Lim CS, Kong DH, Ha KS, et al. Serological responses to a soluble recombinant chimeric Plasmodium vivax circumsporozoite protein in VK210 and VK247 population. Malar J. 2013;12:323.CrossRefPubMedPubMedCentral
78.
Zurück zum Zitat Triglia T, Healer J, Caruana SR, Hodder AN, Anders RF, Crabb BS, et al. Apical membrane antigen 1 plays a central role in erythrocyte invasion by Plasmodium species. Mol Microbiol. 2000;38:706.CrossRefPubMed Triglia T, Healer J, Caruana SR, Hodder AN, Anders RF, Crabb BS, et al. Apical membrane antigen 1 plays a central role in erythrocyte invasion by Plasmodium species. Mol Microbiol. 2000;38:706.CrossRefPubMed
79.
Zurück zum Zitat Yildiz Zeyrek F, Palacpac N, Yuksel F, Yagi M, Honjo K, Fujita Y, et al. Serologic markers in relation to parasite exposure history help to estimate transmission dynamics of Plasmodium vivax. PLoS ONE. 2011;6:e28126.CrossRefPubMedPubMedCentral Yildiz Zeyrek F, Palacpac N, Yuksel F, Yagi M, Honjo K, Fujita Y, et al. Serologic markers in relation to parasite exposure history help to estimate transmission dynamics of Plasmodium vivax. PLoS ONE. 2011;6:e28126.CrossRefPubMedPubMedCentral
80.
Zurück zum Zitat Hester J, Chan ER, Menard D, Mercereau-Puijalon O, Barnwell J, Zimmerman PA, et al. De novo assembly of a field isolate genome reveals novel Plasmodium vivax erythrocyte invasion genes. PLoS Negl Trop Dis. 2013;7:e2569.CrossRefPubMedPubMedCentral Hester J, Chan ER, Menard D, Mercereau-Puijalon O, Barnwell J, Zimmerman PA, et al. De novo assembly of a field isolate genome reveals novel Plasmodium vivax erythrocyte invasion genes. PLoS Negl Trop Dis. 2013;7:e2569.CrossRefPubMedPubMedCentral
81.
Zurück zum Zitat Menard D, Chan ER, Benedet C, Ratsimbasoa A, Kim S, Chim P, et al. Whole genome sequencing of field isolates reveals a common duplication of the duffy binding protein gene in Malagasy Plasmodium vivax strains. PLoS Negl Trop Dis. 2013;7:e2489.CrossRefPubMedPubMedCentral Menard D, Chan ER, Benedet C, Ratsimbasoa A, Kim S, Chim P, et al. Whole genome sequencing of field isolates reveals a common duplication of the duffy binding protein gene in Malagasy Plasmodium vivax strains. PLoS Negl Trop Dis. 2013;7:e2489.CrossRefPubMedPubMedCentral
82.
Zurück zum Zitat Poinsignon A, Cornelie S, Mestres-Simon M, Lanfrancotti A, Rossignol M, Boulanger D, et al. Novel peptide marker corresponding to salivary protein gSG6 potentially identifies exposure to Anopheles bites. PLoS ONE. 2008;3:e247.CrossRef Poinsignon A, Cornelie S, Mestres-Simon M, Lanfrancotti A, Rossignol M, Boulanger D, et al. Novel peptide marker corresponding to salivary protein gSG6 potentially identifies exposure to Anopheles bites. PLoS ONE. 2008;3:e247.CrossRef
Metadaten
Titel
Serological markers to measure recent changes in malaria at population level in Cambodia
verfasst von
Karen Kerkhof
Vincent Sluydts
Laura Willen
Saorin Kim
Lydie Canier
Somony Heng
Takafumi Tsuboi
Tho Sochantha
Siv Sovannaroth
Didier Ménard
Marc Coosemans
Lies Durnez
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Malaria Journal / Ausgabe 1/2016
Elektronische ISSN: 1475-2875
DOI
https://doi.org/10.1186/s12936-016-1576-z

Weitere Artikel der Ausgabe 1/2016

Malaria Journal 1/2016 Zur Ausgabe

Reviewer Acknowledgement

Reviewer acknowledgement 2015

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.