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

Open Access 01.12.2016 | Research

Association between malaria immunity and pregnancy outcomes among Malawian pregnant women receiving nutrient supplementation

verfasst von: Upeksha P. Chandrasiri, Freya J. I. Fowkes, James G. Beeson, Jack S. Richards, Steve Kamiza, Kenneth Maleta, Per Ashorn, Stephen J. Rogerson

Erschienen in: Malaria Journal | Ausgabe 1/2016

Abstract

Background

Malaria antibody responses measured at delivery have been associated with protection from maternal anaemia and low birth weight deliveries. Whether malarial antibodies present in the first half of pregnancy may protect from these or other poor birth outcomes is unclear. To determine whether malaria antibodies in the first half of pregnancy predict pregnancy outcomes, antibodies were measured to a range of merozoite antigens and to antigens expressed on the surface of parasitized red blood cells (pRBCs) in plasma samples collected at 14–20 weeks of gestation from Malawian women. The latter antibodies were measured as total IgG to pRBCs, and antibodies promoting opsonic phagocytosis of pRBCs. Associations between antibodies and maternal haemoglobin in late pregnancy or newborn size were investigated, after adjusting for potential covariates.

Results

Antibodies to pRBC surface antigens were associated with higher haemoglobin concentration at 36 weeks. Total IgG to pRBCs was associated with 0.4 g/l [(95% confidence interval (0.04, 0.8)] increase in haemoglobin, and opsonizing antibody with 0.5 (0.05, 0.9) increase in haemoglobin for each 10% increase in antibody. These antibodies were not associated with birthweight, placental malaria, or newborn anthropometrics. Antibodies to merozoite antigens and non-placental-binding IEs were not associated with decreased risk of any of these outcomes. In some instances, they were negatively associated with outcomes of interest.

Conclusion

Antibodies to placental-binding infected erythrocytes may be associated with higher haemoglobin levels in pregnancy, whereas antibodies to other malaria antigens may instead be markers of malaria exposure.
Trial registration clinicaltrials.gov NCT01239693. Registered Nov 10, 2010.
Abkürzungen
BMI
body mass index
CSA
chondroitin sulphate A
EBA
erythrocyte binding antigen
gw
gestation weeks
HCZ
head circumference z score
HHAZ
household assets z score
HIV
human immunodeficiency
IFA
iron and folic acid
IPTp
intermittent preventive treatment in pregnancy
IQR
interquartile range
ITN
insecticide treated net
LAZ
length-for-age z score
LBW
low birth weight
LNS
lipid-rich nutrient supplement
MMN
multiple micronutrients
MSP
merozoite surface protein
OR
odds ratio
PCR
polymerase chain reaction
PfRh2
Plasmodium falciparum reticulocyte binding homologue 2
PM
placental malaria
pRBC
parasitized red blood cell
SES
socioeconomic status
SGA
small for gestational age
VSA
variant surface antigen

Background

Malaria in pregnancy accounts for an estimated 10,000 maternal and 200,000 infant deaths per year worldwide with over 25 million pregnancies at risk of infection in sub-Saharan Africa alone [1], and is predominantly caused by Plasmodium falciparum. Many of the associated deaths are due to severe maternal anaemia and low birth weight (LBW) [2]. Peripheral blood P. falciparum infections generally peak between 13 and 18 gestation weeks, which is when parasitized red blood cells (pRBCs) begin to sequester in the placenta [3, 4]. Placental adherence of pRBCs is mediated by the interaction between the pregnancy-related variant surface antigen (VSA) VAR2CSA expressed on the surface of the pRBC and chondroitin sulphate A (CSA) receptors expressed by the placental syncytiotrophoblast [5, 6]. With repeated pregnancies, women acquire antibodies to pRBCs that express VAR2CSA and develop protection against infection [7, 8], that is associated with improved pregnancy outcomes [9, 10]. By contrast, antibodies to non-pregnancy-related malaria antigens (merozoite antigens, schizonts and non-pregnancy-specific VSA) have shown little association with pregnancy outcomes [1113].
Women who are parasitaemic at first antenatal care visit may have an increased risk of adverse outcomes such as LBW, placental infection, preterm birth and anaemia [1417]. It is possible that measures of malaria immunity at this point could be used to identify women at relatively high risk of poor pregnancy outcomes. Most studies to date but not all [1820] have investigated the relationship between immunity measured at or close to delivery with pregnancy outcomes. While relevant to defining the potential protective role of malaria immunity in pregnancy outcomes, such studies would not allow health workers to identify women at high or low risk of such outcomes in a timely way, to allow them to offer tailored malaria prevention strategies. If only a proportion of pregnant women are at high risk, and these women could be identified at first antenatal attendance, malaria preventive strategies could be targeted to those lacking protection and at greatest need.
Acquisition of antibodies is dependent on a number of factors such as maternal age, gravidity and presence of other infections such as HIV [7, 21, 22]. In addition, malaria infection, HIV and gravidity have been shown to modify the effects of the association between malaria immunity and pregnancy outcomes [12, 23]. Malaria prevention strategies such as indoor residual spraying, insecticide-treated bed nets (ITNs) and intermittent preventive treatment in pregnancy (IPTp) are designed to reduce the exposure to malaria and improve pregnancy outcomes, but may impair acquisition of immunity [24]. These factors must be considered when determining the relationship between malaria immunity and pregnancy outcomes.
Pregnant women residing in malaria-endemic regions often suffer from undernutrition as a result of poverty, seasonal variation in food availability and poor intake of micronutrients, which significantly impact their health and pregnancy outcomes. It was hypothesized that among Malawian women enrolled in a nutrient supplementation study, one or more measures of naturally acquired malaria antibody, taken at first antenatal visit, would be associated with protection from adverse pregnancy outcomes such as maternal anaemia, evidence of malaria on placental histology, or measures of fetal growth in utero.

Methods

Study context

From February 2011 to August 2012 one thousand three hundred and ninety-one pregnant women were enrolled in the iLiNS-DYAD-M randomized controlled trial, which evaluated whether home fortification of pregnant women’s diets with nutrient supplements could increase birth size. Participants received either iron and folic acid supplements, a multiple micronutrient capsule, or 20 g of lipid-based nutrient supplementation each day from enrolment, from 14 to 20 completed weeks of gestation, until delivery. Dating ultrasounds were performed at enrolment to determine gestation at delivery. All participants received 2 doses of sulfadoxine-pyrimethamine (SP, Fansidar®). The trial design, details of the nutrient supplements, and main findings have been published elsewhere [25, 26]; the type of nutrient supplementation did not affect maternal anti-malarial antibodies, or susceptibility to malaria. Maternal haemoglobin at 36 gestation weeks (gw) and birth weight, gestational age and infant length at delivery were measured, and placental histology was examined.
Peripheral venous blood samples were collected at enrolment and at 36 gw. Only samples collected at enrolment with a paired 36 gw sample were selected for analysis. Samples were not available at 36 gw from women who did not attend this visit, women who had already delivered, and women from whom blood was not available for other logistical reasons. After exclusion of eight sets of twins, all available pairs of samples were assayed. Blood plasma was separated at each time point and stored frozen at −80 °C before shipping to Australia for measurements of anti-malarial immunity. Malaria parasitaemia was determined using blood film microscopy, rapid diagnostic test (RDT), polymerase chain reaction (PCR), and placental histology at delivery. Placental malaria (PM) was categorized as active infection (presence of pRBCs with or without haemozoin in monocytes and/or fibrin); past infection (haemozoin without pRBCs); or no infection, based on placental histology. Due to the low numbers of active placental infections (n = 31), only past placental infection and no infection were compared.

Ethics approval

Ethical approval for the trial and the laboratory work was granted by the College of Medicine Research and Ethics Committee of Malawi, and by Tampere University Hospital Ethics Committee, Finland. The Melbourne Health Human Research Ethics Committee approved the laboratory work performed for the study.

Parasite and cell culture

Plasmodium falciparum isolates CS2 and E8B were selected as representatives of placental-binding and non-placental-binding parasite isolates. CS2 binds to CSA expressed on the placenta while E8B binds to endothelial receptors CD36 and ICAM-1 [27, 28]. Parasite cultures were maintained as described in [29]. Pre-monocytic THP-1 cells were cultured for the opsonic phagocytosis assay and maintained as described in [30].

Measurement of IgG and opsonizing antibodies to VSA expressed by placental-binding and non-placental-binding parasite isolates

Plasma samples from women who had samples available from study enrolment and at 36 gw were used to measure antibody levels. Flow cytometry based immunoassays were performed as previously described [25] to determine relative levels of naturally-acquired IgG to VSA expressed by the parasite isolates described above, and to measure levels of antibodies that opsonize pRBC for phagocytosis by THP-1 cells. Plasma was assayed at 1:20 dilution (IgG to VSA) or 1:10 (opsonizing antibodies). For both types of assay, antibody levels were reported as a percentage of the positive controls where the positive controls were a pool of human plasma with known high levels of antibody to VSA antigens. Positivity for each antigen was calculated as mean plus three standard deviations of the fluorescence intensity of 30 different negative controls.

Measurement of IgG to merozoite antigens and schizont extract via fluorescent immunosorbent assay

Recombinant merozoite antigens MSP1 19kD, MSP2, MSP3, PfRh2 (construct PfRh2-2030) (6088–7584 bp) and EBA175 (region III-V) were prepared as detailed [25]. A fluorescent immunosorbent assay was designed to measure IgG to merozoite antigens as described in [25] and the IgG levels were reported as a percentage of the positive control, where the positive control was a pool of human plasma collected from individuals with immunity to malaria.

Statistical methods

The outcome variables of interest included maternal haemoglobin levels (g/l, continuous) at 36 gw; past placental malaria (PM) compared to no placental malaria; birthweight (in grams, continuous variable); small for gestational age (SGA: binary variable defined as birthweight below the 10th centile on the United States national reference scale for fetal growth [31]), stunting: length-for-age Z-score (LAZ) <−2 at birth (binary variable); and small head circumference (HCZ <−2).
The selected covariates included gravidity, maternal age, HIV, bed net use, body mass index (BMI) at enrolment, malaria microscopy at enrolment, socioeconomic status (SES), study site, season at enrolment and supplementation group, and were recorded as described [25]. These covariates were chosen based on their effects on malaria antibody levels and pregnancy outcomes in the cohort, or their previously-demonstrated biological relevance to the association between antibody levels and pregnancy outcomes. Gravidity groups were categorized as primi, secundi and multigravidae. Maternal age was categorized as <21, 21–25, 26–30 and >30 years. Maternal BMI at enrolment was categorized into low (<18.5 kg/m2), normal (18.5–25 kg/m2) and increased (>25 kg/m2). The SES was calculated based on a household assets adjusted z-score (HHAZ) system [32], where a participant with a HHAZ below the median (z-score = −0.387) were considered as having a low SES. HIV, malaria infection, seasons at enrolment (rainy and dry) and ITN use were incorporated as dichotomous variables. Study sites (Lungwena, Malindi, Namwera and Mangochi) and supplementation groups (IFA, MMN and LNS) were used as categorical variables.

Statistical analyses of the relationship between antibody levels and pregnancy outcomes after adjusting for covariates

Statistical analyses were performed using STATA 13.0 version (StataCorp LP, Texas, USA). Differences in the characteristics between the participants included and excluded in the study were determined by performing a Student’s t test for normally distributed variables or Mann–Whitney U test for non-normally distributed variables, or test of proportion for categorical variables.
Univariate and multivariate linear or logistic regression analysis were performed as appropriate to assess the relationship between antibody levels at enrolment and pregnancy outcomes adjusting for the covariates described above. Antibody levels were treated as continuous variables or as categorical variables following categorization into low, middle and high antibody tertiles.

Results

Study participants

Following exclusion of twins, antibody determination was performed for plasma samples collected from 1002 participants at enrolment who also had a sample at 36 gw. Among participants with samples at 36 gw, only 2.8% had preterm births.
Socio-demographic characteristics and birth outcome differences between iLiNS study participants who were included and excluded from the present analyses were determined (Table 1). Women included in the analyses were similar to those excluded in the analysis in age, HIV prevalence, SES and BMI. However women who were excluded had shorter pregnancies (because only women with 36 gestation week data available were included), more frequently delivered children with low birthweight, were more likely to be in their first pregnancy, and less likely to use ITNs compared to those who were included. Among included women, 10% had malaria infection detected by microscopy and 25% by PCR at enrolment (Table 1).
Table 1
Comparison of characteristics of the participants included and excluded in the analysis
Characteristic
Included (n = 1002)
Excluded (n = 377)
p value
Gestation weeks at delivery, mean (SD)
39.8 (1.5)
37.0 (4.9)
<0.0001*
Maternal age, years, mean (SD)
24.5 (5.8)
25.3 (6.1)
0.214
Gravidity, number (%)
 Primigravidae
199 (19.9%)
104 (27.6%)
0.002*
 Secundigravidae
201 (20.1%)
80 (21.2%)
0.630
 Multigravidae
600 (59.9%)
192 (50.9%)
0.003*
Low socio-economic status, number (%)
554 (55.7%)
162 (50.9%)
0.135
BMI at enrolment, median (IQR), kg/m2
21.6 (20.3, 23.5)
22.1 (20.4, 23.7)
0.063
HIV infected, number (%)
129 (13.0%)
52 (15.9%)
0.186
Maternal haemoglobin levels at 36 gw, median (IQR), g/l
110 (101, 120)
115 (106, 123)
0.052
Prevalence of anaemia at 36 gw (Hb <100 g/l), number (%)
200 (20.9%)
12 (14.5%)
0.165
Birthweight, mean (SD), g
3011 (412.8)
2791 (536.5)
<0.0001*
Prevalence of low birthweight, <2500 g
96 (10.3%)
51 (23.8%)
<0.0001*
Prevalence of SGA, number (%)
255 (30.6%)
54 (29.0%)
0.668
Microscopy at enrolment, number (%)
107 (10.7%)
39 (10.4%)
0.872
PCR positivity for malaria at enrolment, number (%)
245 (25.0%)
111 (30.2%)
0.054
PCR positivity for malaria at 36 gw, number (%)
92 (9.3%)
4 (5.1%)
0.212
Placental malaria
 Active infection
31 (3.8%)
11 (5.6%)
0.257
 Past infection
293 (36.0%)
55 (28.1%)
0.037*
 Uninfected
489 (60.1%)
130 (66.3%)
0.110
ITN use at enrolment, number (%)
399 (72.0%)
122 (59.5%)
0.001*
Data presented as mean (SD), median (IQR) or number (proportion of women as a percentage) for women included and excluded in the study, allowing for missing observations for each variable
SD, standard deviation; IQR, interquartile range; BMI, body mass index; HIV, human immunodeficiency virus; Hb, haemoglobin; SGA, small for gestational age; PCR, polymerase chain reaction

Associations between malaria antibody at enrolment and maternal haemoglobin concentration at 36 weeks and birth weight

Median haemoglobin levels at 36 gw were 110 g/l (IQR 101–120; Table 1). After adjustment for covariates including gravidity and malaria at enrolment, there was a statistically significant positive association between antibodies measured before 20 gestation weeks to placental-binding VSA and haemoglobin concentration; with every 10% increase in antibody levels the haemoglobin concentration at 36 gw increases by 0.5 g/l (95% CI 0.05, 0.9; p = 0.029) for IgG to placental-binding VSA and 0.4 g/l (0.04, 0.8; p = 0.030) for opsonic phagocytosis activity of antibodies to placental-binding VSA (Table 2). The median levels of antibodies to placental-binding VSA and of opsonic phagocytosis activity of antibodies were 15.0 and 32.7% of positive controls, respectively. In contrast, antibodies to MSP1 19kD, MSP2, EBA175 and schizont extract were negatively associated with haemoglobin levels at 36 weeks (coefficients −0.5 to −1.3 g/l, 0.003 < p<0.032; Table 2). There was no statistically significant association between levels of antibodies to the non-placental-binding line, or to recombinant MSP3 or Rh2A9, and maternal haemoglobin concentration.
Table 2
Association between the participants’ malaria antibody levels at enrolment and their blood haemoglobin concentration at 36 gestation weeks and their child’s birthweight
Antibody type
Haemoglobin concentration (g/l)
Birthweight (g)
Coefficient (95% CI)
p value
Coefficient (95% CI)
p value
IgG to placental-binding VSA
0.5 (0.05, 0.9)
0.029*
−9.7 (−21.5, 2.1)
0.106
Opsonising antibodies to placental-binding VSA
0.4 (0.04, 0.8)
0.030*
2.0 (−9.0, 13.0)
0.721
Opsonising antibodies to non-placental-binding VSA
0.2 (−0.3, 0.6)
0.476
−3.1 (−16.4, 10.2)
0.651
MSP1 19kD
−0.7 (−1.3, −0.1)
0.018*
9.4 (−7.8, 26.6)
0.283
MSP2
−0.7 (−1.2, −0.1)
0.017*
5.2 (−10.3, 20.8)
0.507
MSP3
−0.2 (−0.7, 0.4)
0.592
−13.2 (−29.7, 3.3)
0.115
EBA175
−1.1 (−1.7, −0.4)
0.003*
5.3 (−14.0, 24.5)
0.589
PfRh2
−0.2 (−0.8, 0.5)
0.604
−8.6 (−27.3, 10.0)
0.364
Schizont extract
−0.5 (−0.10, −0.05)
0.032*
3.1 (−10.1, 16.2)
0.648
Analysis for each individual antibody was adjusted for gravidity, maternal age, HIV, ITN use, body mass index at enrolment, malaria microscope positivity at enrolment, socioeconomic status, study site, season at enrolment and supplementation groups. Results show increase in haemoglobin or birth weight per 10% increase in antibody
95 % CI, 95% confidence interval; IgG, immunoglobulin G; VSA, variant surface antigens, MSP, merozoite surface protein; EBA175, erythrocyte binding homologue 175; PfRh2, Plasmodium falciparum reticulocyte binding homologue 2
In adjusted analyses, antibodies to none of the targets examined were significantly associated with birthweight; differences attributable to each 10% increase in antibody ranged up to 13 g (Table 2).

Association between malaria antibody at enrolment and placental malaria, small for gestational age and low length-for-age

To examine relationships between antibody immunity and categorical outcomes, antibody responses were divided into three equal-sized groups, high, medium and low.
Because few women (31, 4%) had active placental malaria, the analysis compared only past PM (present in 36%) to no PM. In adjusted analyses, pregnant women in the high antibody tertile for MSP2 were more likely to have past PM than those in the low antibody tertile, OR 1.83 (95% CI 1.01, 3.31; p = 0.046) (Table 3). There were odds ratios of similar magnitude for high compared to low levels of opsonizing antibodies to the placental-binding and non-placental-binding parasite lines and high compared to low levels of antibody to schizont extract at enrolment but these associations were not statistically significant (Table 3).
Table 3
Association between antibody tertiles at enrolment and odds of past placental malaria
Antibody type
Past placental malaria
Middle compared to low antibody tertile
High compared to low antibody tertile
OR (95% CI)
p value
OR (95% CI)
p value
IgG to placental-binding VSA
1.20 (0.65, 2.22)
0.558
1.30 (0.70, 2.40)
0.402
Opsonising antibodies to placental-binding VSA
1.64 (0.91, 2.98)
0.102
1.82 (0.99, 3.36)
0.054
Opsonising antibodies to non-placental-binding VSA
1.31 (0.73, 2.35)
0.363
1.74 (0.97, 3.13)
0.065
MSP1 19kD
1.23 (0.69, 2.18)
0.488
1.27 (0.72, 2.25)
0.407
MSP2
1.47 (0.82, 2.64)
0.198
1.83 (1.01, 3.31)
0.046*
MSP3
0.59 (0.33, 1.05)
0.071
1.12 (0.65, 1.92)
0.691
EBA175
0.97 (0.55, 1.71)
0.913
1.14 (0.66, 1.99)
0.636
PfRh2
0.94 (0.54, 1.63)
0.818
1.05 (0.59, 1.85)
0.879
Schizont extract
1.51 (0.78, 2.92)
0.223
1.70 (0.90, 3.24)
0.104
Data presented as odds ratio (95% confidence interval). Multivariate logistic regression analysis performed to determine the risk of past PM between pregnant women in the middle antibody tertile compared to women in the low antibody tertile. Analysis adjusted for gravidity, maternal age, HIV, ITN use, body mass index at enrolment, malaria microscopy at enrolment, socioeconomic status, study site and supplementation groups
OR, odds ratio, 95 % CI, 95% confidence interval; IgG, immunoglobulin G; VSA, variant surface antigens, MSP, merozoite surface protein; EBA175, erythrocyte binding homologue 175; PfRh2 Plasmodium falciparum reticulocyte binding homologue 2
In adjusted analyses, antibodies to none of the targets examined were significantly associated with being born SGA (Table 4). Women in the middle tertile for opsonizing antibody to placental-binding VSA had a significantly higher risk of LAZ <−2, RRR 2.25 (95% CI 1.14, 4.42) than women in the low tertile group, but this was not observed in comparisons of high versus low antibody comparisons (OR 1.14 95%CI 0.53, 2.47) (Table 4). No other significant associations were found, and the OR for other associations was small.
Table 4
Association between antibody tertiles at enrolment and small for gestational age and low length-for-age (LAZ <−2)
Antibody type
Small for gestational age
Low length-for-age (LAZ <−2)
Middle compared to low antibody tertile
High compared to low antibody tertile
Middle compared to low antibody tertile
High compared to low antibody tertile
OR (95% CI)
p value
OR (95% CI)
p value
OR (95% CI)
p value
OR (95% CI)
p value
IgG to placental-binding VSA
0.74 (0.42, 1.31)
0.301
0.91 (0.52,1.60)
0.736
1.27 (0.62, 2.62)
0.513
1.05 (0.50, 2.21)
0.894
Opsonising antibodies to placental-binding VSA
0.92 (0.54, 1.57)
0.757
0.78 (0.45, 1.35)
0.369
2.25 (1.14, 4.42)
0.019*
1.14 (0.53, 2.47)
0.734
Opsonising antibodies to non-placental-binding VSA
1.34 (0.79, 2.28)
0.282
1.17 (0.67, 2.02)
0.584
1.29 (0.65, 2.56)
0.465
1.13 (0.56, 2.28)
0.735
MSP1 19kD
1.21 (0.71, 2.05)
0.476
0.93 (0.55, 1.59)
0.795
1.05 (0.54, 2.02)
0.889
0.75 (0.38, 1.48)
0.408
MSP2
1.12 (0.66, 1.89)
0.671
0.82 (0.48, 1.42)
0.486
0.97 (0.50, 1.90)
0.930
0.96 (0.48, 1.93)
0.918
MSP3
1.28 (0.75, 2.18)
0.358
1.65 (0.98, 2.80)
0.060
0.87 (0.44, 1.72)
0.682
1.28 (0.67, 2.46)
0.459
EBA175
1.62 (0.94, 2.75)
0.081
1.45 (0.85, 2.45)
0.172
0.88 (0.45, 1.72)
0.711
0.84 (0.43, 1.62)
0.608
PfRh2
1.26 (0.75, 2.13)
0.378
1.01 (0.59, 1.72)
0.979
1.18 (0.61, 2.29)
0.620
0.92 (0.46, 1.84)
0.815
Schizont extract
1.25 (0.68, 2.28)
0.469
1.44 (0.80, 2.61)
0.226
1.15 (0.54, 2.44)
0.724
1.27 (0.61, 2.63)
0.518
Data presented as odds ratio (95% confidence interval). Multivariate logistic regression analysis performed to determine the risk of giving birth to a child with SGA and LAZ <−2 among pregnant women in middle and high antibody tertile compared to women in the low antibody tertile. Analysis adjusted for gravidity, maternal age, HIV, bed net use, body mass index at enrolment, malaria microscopy at enrolment, socioeconomic status, study site, season at enrolment and supplementation groups
LAZ, length-for-age z-score; OR, odds ratio; 95 % CI, 95% confidence interval; IgG, immunoglobulin G; VSA, variant surface antigens, MSP, merozoite surface protein; EBA175, erythrocyte binding homologue 175; PfRh2, Plasmodium falciparum reticulocyte binding homologue 2

Discussion

This study tested the hypothesis that measures of malaria antibody immunity at first antenatal visit could predict pregnancy outcomes in Malawian women enrolled in the iLiNS-DYAD-M clinical trial. In 1002 women, the study showed that IgG to VSA and antibodies promoting opsonic phagocytosis of pRBCs of placental-binding parasite isolates were associated with higher maternal haemoglobin levels at 36 gw. These measures of pregnancy-specific malaria immunity were not significantly associated with birthweight or SGA in adjusted analyses. Antibodies to other malaria antigens assessed either demonstrated no significant associations or negative associations with the pregnancy outcomes assessed. Antibodies to merozoite antigens MSP1 19kD, MSP2, EBA175 and schizont extract were associated with lower haemoglobin concentration at 36 weeks and antibodies to MSP2 were associated with an increased likelihood of past PM. The study was performed in women receiving recommended doses of IPTp and ITNs, and investigated potential additional benefits of antibody immunity on outcomes.
The positive associations between both IgG and opsonizing antibodies to placental-binding isolate VSA and haemoglobin levels at 36 gw are in agreement with previous observations in pregnant women from Blantyre, Malawi [33, 34] and Kilifi, Kenya [35], although two of these studies examined antibody responses only at term. By contrast, IgG to VSA of a placental-binding isolate, measured (like this study) in samples collected in first half of pregnancy, was not associated with haemoglobin concentrations in a previous clinical trial in Lungwena, Malawi [18]. In that study, antibody responses were measured in 549 women (about half the present study), and the study lacked statistical power to detect modest associations. Maternal anaemia may increase the risk of preterm births [36] and interferes with fetal growth causing LBW [2, 37]. Although the haemoglobin concentration was only 0.4–0.5 g/l higher with every 10% increase in antibody levels (so a woman with antibody level 100% of controls is expected to have a haemoglobin 5 g/l higher than a matched woman with no antibody) the positive association observed in the current cohort suggests a potential role of these antibodies in preventing malaria-induced anaemia.
The associations between levels of antibodies to some merozoite proteins or to schizont extract and risk of anaemia suggest that some of these antibodies may be acting as markers of previous malaria infection, rather than predictors of better pregnancy outcomes. Antibodies to merozoite antigens are well known to increase with exposure [38]. Repeated malaria episodes will lead to cumulative destruction of RBCs and lower haemoglobin levels. SES was strongly associated with malaria risk (likely to be correlated with exposure, e.g. through poor housing) and was adjusted for in the analysis, but there may be other factors that affected malaria and anaemia risk in this group which were not identified and so not adjusted for, such as location and characteristics of residence, which may affect malaria exposure, but might also be markers of poverty and/or nutritional insecurity.
There was a weak association between antibodies to MSP2 and past placental malaria, and similar trends for antibody to schizont antigen and opsonizing antibody to both placental-binding and non-placental-binding parasite lines. Past placental malaria is characterized by persistent haemozoin deposits in fibrin in the placenta with no pRBCs [39], a sign of infection that has been cleared prior to delivery. Women with high antibody levels at study entry likely live in environments where they are at great risk of malaria infection, and this higher risk may persist in pregnancy despite use of IPTp and ITNs, and so will increase the likelihood they have experienced placental malaria at some point in pregnancy. Placental blood circulation is established at around 9–12 weeks gestation [40]. Infections occurring before study enrolment may increase antibodies to MSP2, while infections present at any time after 9–12 weeks’ gestation may affect the placenta, but will be cleared by IPTp leaving residual pigment. It is likely that the same women are at higher risk of malaria over both periods.
Antibodies to placental-binding VSA were not significantly associated with improved newborn anthropometrics unlike previous reports [9, 35, 41]. In previous reports the mothers were secundigravidae [9], secundigravidae with placental malaria and HIV [41] or women with chronic placental infection [35] and antibody levels were measured at delivery rather than in early pregnancy. Rather than demonstrating population-wide associations between malaria antibodies and birthweight (as studied here), most published studies have been restricted to women of a specific gravidity or HIV and/or malaria infection status [12, 30, 33]. Further prospective studies of these high-risk groups for associations between immunity and outcome may be indicated.
There was an unexpected negative association between opsonizing antibody to the placental-binding parasite line and newborn length, seen when women in the middle and low antibody groups were compared. The cause of this is not clear, and there was no similar trend when women with high and low antibody response were compared, nor was there a similar association between levels of IgG antibody to VSA of the placental-binding line and newborn length.
Endpoints such as maternal haemoglobin, birth weight, SGA and newborn length are multifactorial in aetiology, and estimating the proportion of each attributable to malaria is challenging. Study participants received protection against malaria with two doses of IPTp with sulphadoxine pyrimethamine and ITNs, limiting their exposure to malaria after enrolment, decreasing their risk of anaemia and LBW, and improving birth weights [42]. Only 3% of women had active malaria infection on placental histology, and malaria prevalence by RDT was 8%, a decrease from 23% at first antenatal visit (M Nkhoma, in submission). Malaria in later pregnancy was associated with low birth weight and decreased fetal size (M Nkhoma, in submission), but in settings where malaria prevalence is low (such as when malaria transmission falls), the attributable fractions of low birth weight and other outcomes due to malaria may be low, and it may therefore become increasingly difficult to identify immune responses to malaria that protect against adverse pregnancy outcomes.
Strengths of the study included a large sample size, of over 1000 women; analysis of a range of antibody measures; and extensive clinical data. Linear and logistic regression analyses were used to adjust for factors associated with the input and/or dependent variables of interest. Study limitations included few women with preterm delivery (because only women with a 36 gw visit were included), preventing an investigation of whether antibody protects against preterm delivery; measurement of antibody responses to single parasite isolates of each phenotype, and single variants of each of a modest number of merozoite proteins; and measurement of a single functional role of antibodies (opsonization of pRBC for phagocytosis). Apart from preterm delivery and associated lower birth weights, other characteristics more common in excluded women included being primigravid, and higher rates of malaria infection by PCR at enrolment. This may have limited our power to find protective associations. Quantitating antibody to other placental-binding parasite isolates, [43] and understanding other antibody functions, such as merozoite opsonization [44] or complement fixation [45] could be beneficial in future studies. HIV was included as a co-factor, but helminth and bacterial infections which are prevalent among Malawian pregnant women [4648] were not evaluated, which may have influenced certain analyses such as association with haemoglobin concentration. To dissect the determinants of aetiologically-complex pregnancy outcomes such as maternal haemoglobin, birth size, or preterm delivery, a broad range of comorbid conditions may need to be addressed.
The study findings suggest that antibody to pregnancy-specific parasite isolates, measured using two different approaches, may be associated with protection from malarial anaemia, whereas antibodies to merozoite-specific antigens are not. Further studies that prospectively evaluate pregnancy-specific and more general measures of malaria antibody, under a range of transmission settings, will be required to determine whether one or more measures may identify women at risk with sufficient sensitivity and specificity to be considered as a screening tool for high-risk pregnancies in the future. The lack of evidence that naturally-acquired antibodies to one placental binding parasite line were associated with improved fetal growth may reflect the relatively modest impact of malaria on birth weight in this cohort. Demonstration that induction of antibodies by a VAR2CSA based vaccine improves pregnancy outcomes would require careful selection of target populations in which malaria is a major contributor to adverse pregnancy outcomes.

Authors’ contributions

SJR and PA conceived the study with KM. UPC performed antibody measurements with assistance from JSR, and SK interpreted placental histology. UPC, FJIF, JGB, PA and SJR interpreted data. UPC and SJR drafted the manuscript. All authors read and approved the final manuscript.

Acknowledgements

We acknowledge all the study participants, the local communities, the health services staff and our research personnel at the study sites as well as members of the trial’s data safety and monitoring board, and Prof Kay Dewey and the International Lipid-based Nutrient Supplementation (iLiNS)-Project Steering Committee (http://​www.​ilins.​org/​) for their positive attitude, support and help in all stages of the study. In addition, we would like to thank Steven Meshnick, Kyaw L Thwai, Wina Hasang and Philippe Boeuf for advice and technical support.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

The datasets during and/or analysed during the current study available from the corresponding author on reasonable request.

Ethics approval

Ethical approval for the trial and the laboratory work was granted by the College of Medicine Research and Ethics Committee of Malawi, and by Tampere University Hospital Ethics Committee, Finland. The Melbourne Health Human Research Ethics Committee approved the laboratory work performed for the study.

Funding

This publication is based on research funded in part by a grant from the Office of Health, Infectious Diseases, and Nutrition, Bureau for Global Health, U.S. Agency for International Development (USAID) under terms of Cooperative Agreement No. AID-OAA-A-12-00005, through the Food and Nutrition Technical Assistance III Project (FANTA), managed by FHI 360. Additional funding was obtained from a grant to the University of California, Davis from the Bill & Melinda Gates Foundation and the Academy of Finland. SJR is supported by the National Health and Medical Research Council (NHMRC) of Australia. JGB supported by a Senior Research Fellowship by the NHMRC of Australia. The Burnet Institute is supported by the NHMRC IRIIS Scheme and Victorian State Government Operational Infrastructure Support.
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.
Literatur
1.
Zurück zum Zitat Desai M, Ter Kuile FO, Nosten F, McGready R, Asamoa K, Brabin B, Newman RD. Epidemiology and burden in malaria in pregnancy. Lancet Infect Dis. 2007;7:93–104.CrossRefPubMed Desai M, Ter Kuile FO, Nosten F, McGready R, Asamoa K, Brabin B, Newman RD. Epidemiology and burden in malaria in pregnancy. Lancet Infect Dis. 2007;7:93–104.CrossRefPubMed
2.
Zurück zum Zitat Steketee RW, Nahlen BL, Parise ME, Menendez C. The burden of malaria in pregnancy in malaria-endemic areas. Am J Trop Med Hyg. 2001;64:28–35.PubMed Steketee RW, Nahlen BL, Parise ME, Menendez C. The burden of malaria in pregnancy in malaria-endemic areas. Am J Trop Med Hyg. 2001;64:28–35.PubMed
4.
Zurück zum Zitat Doritchamou J, Bertin G, Moussiliou A, Bigey P, Viwami F, Ezinmegnon S, et al. First-trimester Plasmodium falciparum infections display a typical “placental” phenotype. J Infect Dis. 2012;206:1911–9.CrossRefPubMed Doritchamou J, Bertin G, Moussiliou A, Bigey P, Viwami F, Ezinmegnon S, et al. First-trimester Plasmodium falciparum infections display a typical “placental” phenotype. J Infect Dis. 2012;206:1911–9.CrossRefPubMed
5.
Zurück zum Zitat Doritchamou J, Sossou-tchatcha S, Cottrell G, Moussiliou A, Hounton Houngbeme C, Massougbodji A, et al. Dynamics in the cytoadherence phenotypes of Plasmodium falciparum infected erythrocytes isolated during pregnancy. PLoS ONE. 2014;9:e98577.CrossRefPubMedPubMedCentral Doritchamou J, Sossou-tchatcha S, Cottrell G, Moussiliou A, Hounton Houngbeme C, Massougbodji A, et al. Dynamics in the cytoadherence phenotypes of Plasmodium falciparum infected erythrocytes isolated during pregnancy. PLoS ONE. 2014;9:e98577.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Srivastava A, Gangnard S, Round A, Dechavanne S, Juillerat A, Raynal B, et al. Full-length extracellular region of the var2CSA variant of PfEMP1 is required for specific, high-affinity binding to CSA. Proc Natl Acad Sci USA. 2010;107:4884–9.CrossRefPubMedPubMedCentral Srivastava A, Gangnard S, Round A, Dechavanne S, Juillerat A, Raynal B, et al. Full-length extracellular region of the var2CSA variant of PfEMP1 is required for specific, high-affinity binding to CSA. Proc Natl Acad Sci USA. 2010;107:4884–9.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat O’Neil-Dunne I, Achur RN, Agbor-Enoh ST, Valiyaveettil M, Naik RS, Ockenhouse CF, et al. Gravidity-dependent production of antibodies that inhibit binding of Plasmodium falciparum-infected erythrocytes to placental chondroitin sulfate proteoglycan during pregnancy. Infect Immun. 2001;69:7487–92.CrossRefPubMedPubMedCentral O’Neil-Dunne I, Achur RN, Agbor-Enoh ST, Valiyaveettil M, Naik RS, Ockenhouse CF, et al. Gravidity-dependent production of antibodies that inhibit binding of Plasmodium falciparum-infected erythrocytes to placental chondroitin sulfate proteoglycan during pregnancy. Infect Immun. 2001;69:7487–92.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Fried M, Nosten F, Brockman A, Brabin BJ, Duffy PE. Maternal antibodies block malaria. Nature. 1998;395:851–2.CrossRefPubMed Fried M, Nosten F, Brockman A, Brabin BJ, Duffy PE. Maternal antibodies block malaria. Nature. 1998;395:851–2.CrossRefPubMed
9.
Zurück zum Zitat Duffy PE, Fried M. Antibodies that inhibit Plasmodium falciparum adhesion to chondroitin sulfate A are associated with increased birth weight and the gestational age of newborns. Infect Immun. 2003;71:6620–3.CrossRefPubMedPubMedCentral Duffy PE, Fried M. Antibodies that inhibit Plasmodium falciparum adhesion to chondroitin sulfate A are associated with increased birth weight and the gestational age of newborns. Infect Immun. 2003;71:6620–3.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Ndam NT, Denoeud-Ndam L, Doritchamou J, Viwami F, Salanti A, Nielsen MA, et al. Protective antibodies against placental malaria and poor outcomes during pregnancy. Benin. Emerg Infect Dis. 2015;21:813–23.CrossRefPubMed Ndam NT, Denoeud-Ndam L, Doritchamou J, Viwami F, Salanti A, Nielsen MA, et al. Protective antibodies against placental malaria and poor outcomes during pregnancy. Benin. Emerg Infect Dis. 2015;21:813–23.CrossRefPubMed
11.
Zurück zum Zitat Serra-Casas E, Menendez C, Bardaji A, Quinto L, Dobano C, Sigauque B, et al. The effect of intermittent preventive treatment during pregnancy on malarial antibodies depends on HIV status and is not associated with poor delivery outcomes. J Infect Dis. 2010;201:123–31.CrossRefPubMed Serra-Casas E, Menendez C, Bardaji A, Quinto L, Dobano C, Sigauque B, et al. The effect of intermittent preventive treatment during pregnancy on malarial antibodies depends on HIV status and is not associated with poor delivery outcomes. J Infect Dis. 2010;201:123–31.CrossRefPubMed
12.
Zurück zum Zitat Mayor A, Kumar U, Bardaji A, Gupta P, Jimenez A, Hamad A, et al. Improved pregnancy outcomes in women exposed to malaria with high antibody levels against Plasmodium falciparum. J Infect Dis. 2013;207:1664–74.CrossRefPubMed Mayor A, Kumar U, Bardaji A, Gupta P, Jimenez A, Hamad A, et al. Improved pregnancy outcomes in women exposed to malaria with high antibody levels against Plasmodium falciparum. J Infect Dis. 2013;207:1664–74.CrossRefPubMed
13.
Zurück zum Zitat Owalla TJ, Palacpac NM, Shirai H, Horii T, Egwang TG. Association of naturally acquired IgG antibodies against Plasmodium falciparum serine repeat antigen-5 with reduced placental parasitemia and normal birth weight in pregnant Ugandan women: a pilot study. Parasitol Int. 2013;62:237–9.CrossRefPubMed Owalla TJ, Palacpac NM, Shirai H, Horii T, Egwang TG. Association of naturally acquired IgG antibodies against Plasmodium falciparum serine repeat antigen-5 with reduced placental parasitemia and normal birth weight in pregnant Ugandan women: a pilot study. Parasitol Int. 2013;62:237–9.CrossRefPubMed
14.
Zurück zum Zitat Kalilani L, Mofolo I, Chaponda M, Rogerson SJ, Meshnick SR. The effect of timing and frequency of Plasmodium falciparum infection during pregnancy on the risk of low birth weight and maternal anemia. Trans R Soc Trop Med Hyg. 2010;104:416–22.CrossRefPubMedPubMedCentral Kalilani L, Mofolo I, Chaponda M, Rogerson SJ, Meshnick SR. The effect of timing and frequency of Plasmodium falciparum infection during pregnancy on the risk of low birth weight and maternal anemia. Trans R Soc Trop Med Hyg. 2010;104:416–22.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Huynh BT, Fievet N, Gbaguidi G, Dechavanne S, Borgella S, Guezo-Mevo B, et al. Influence of the timing of malaria infection during pregnancy on birth weight and on maternal anemia in Benin. Am J Trop Med Hyg. 2011;85:214–20.CrossRefPubMedPubMedCentral Huynh BT, Fievet N, Gbaguidi G, Dechavanne S, Borgella S, Guezo-Mevo B, et al. Influence of the timing of malaria infection during pregnancy on birth weight and on maternal anemia in Benin. Am J Trop Med Hyg. 2011;85:214–20.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Kalilani-Phiri L, Thesing PC, Nyirenda OM, Mawindo P, Madanitsa M, Membe G, et al. Timing of malaria infection during pregnancy has characteristic maternal, infant and placental outcomes. PLoS ONE. 2013;8:e74643.CrossRefPubMedPubMedCentral Kalilani-Phiri L, Thesing PC, Nyirenda OM, Mawindo P, Madanitsa M, Membe G, et al. Timing of malaria infection during pregnancy has characteristic maternal, infant and placental outcomes. PLoS ONE. 2013;8:e74643.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Walker PG, ter Kuile FO, Garske T, Menendez C, Ghani AC. Estimated risk of placental infection and low birthweight attributable to Plasmodium falciparum malaria in Africa in 2010: a modelling study. Lancet Glob Health. 2014;2:e460–7.CrossRefPubMed Walker PG, ter Kuile FO, Garske T, Menendez C, Ghani AC. Estimated risk of placental infection and low birthweight attributable to Plasmodium falciparum malaria in Africa in 2010: a modelling study. Lancet Glob Health. 2014;2:e460–7.CrossRefPubMed
18.
Zurück zum Zitat Aitken EH, Mbewe B, Luntamo M, Maleta K, Kulmala T, Friso M, et al. Antibodies to Chondroitin sulfate a–binding infected erythrocytes: dynamics and protection during pregnancy in women receiving intermittent preventive treatment. J Infect Dis. 2010;201:1316–25.CrossRefPubMed Aitken EH, Mbewe B, Luntamo M, Maleta K, Kulmala T, Friso M, et al. Antibodies to Chondroitin sulfate a–binding infected erythrocytes: dynamics and protection during pregnancy in women receiving intermittent preventive treatment. J Infect Dis. 2010;201:1316–25.CrossRefPubMed
19.
Zurück zum Zitat Tutterrow YL, Salanti A, Avril M, Smith JD, Pagano IS, Ako S, et al. High avidity antibodies to full-length VAR2CSA correlate with absence of placental malaria. PLoS ONE. 2012;7:e40049.CrossRefPubMedPubMedCentral Tutterrow YL, Salanti A, Avril M, Smith JD, Pagano IS, Ako S, et al. High avidity antibodies to full-length VAR2CSA correlate with absence of placental malaria. PLoS ONE. 2012;7:e40049.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Tutterrow YL, Avril M, Singh K, Long CA, Leke RJ, Sama G, et al. High levels of antibodies to multiple domains and strains of VAR2CSA correlate with absence of placental malaria in Cameroonian women living in an area of high Plasmodium falciparum transmission. Infect Immun. 2012;80:1479–90.CrossRefPubMedPubMedCentral Tutterrow YL, Avril M, Singh K, Long CA, Leke RJ, Sama G, et al. High levels of antibodies to multiple domains and strains of VAR2CSA correlate with absence of placental malaria in Cameroonian women living in an area of high Plasmodium falciparum transmission. Infect Immun. 2012;80:1479–90.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Keen J, Serghides L, Ayi K, Patel SN, Ayisi J, van Eijk A, et al. HIV impairs opsonic phagocytic clearance of pregnancy-associated malaria parasites. PLoS Med. 2007;4:e181.CrossRefPubMedPubMedCentral Keen J, Serghides L, Ayi K, Patel SN, Ayisi J, van Eijk A, et al. HIV impairs opsonic phagocytic clearance of pregnancy-associated malaria parasites. PLoS Med. 2007;4:e181.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Mount AM, Mwapasa V, Elliott SR, Beeson JG, Tadesse E, Lema VM, et al. Impairment of humoral immunity to Plasmodium falciparum malaria in pregnancy by HIV infection. Lancet. 2004;363:1860–7.CrossRefPubMed Mount AM, Mwapasa V, Elliott SR, Beeson JG, Tadesse E, Lema VM, et al. Impairment of humoral immunity to Plasmodium falciparum malaria in pregnancy by HIV infection. Lancet. 2004;363:1860–7.CrossRefPubMed
23.
Zurück zum Zitat Ataide R, Mayor A, Rogerson SJ. Malaria, primigravidae, and antibodies: knowledge gained and future perspectives. Trends Parasitol. 2014;30:85–94.CrossRefPubMed Ataide R, Mayor A, Rogerson SJ. Malaria, primigravidae, and antibodies: knowledge gained and future perspectives. Trends Parasitol. 2014;30:85–94.CrossRefPubMed
24.
Zurück zum Zitat Aitken EH, Mbewe B, Luntamo M, Kulmala T, Beeson JG, Ashorn P, Rogerson SJ. Antibody to P. falciparum in pregnancy varies with intermittent preventive treatment regime and bed net use. PLoS ONE. 2012;7:e29874.CrossRefPubMedPubMedCentral Aitken EH, Mbewe B, Luntamo M, Kulmala T, Beeson JG, Ashorn P, Rogerson SJ. Antibody to P. falciparum in pregnancy varies with intermittent preventive treatment regime and bed net use. PLoS ONE. 2012;7:e29874.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Chandrasiri UP, Fowkes FJ, Richards JS, Langer C, Fan YM, Taylor SM, et al. The impact of lipid-based nutrient supplementation on anti-malarial antibodies in pregnant women in a randomized controlled trial. Malar J. 2015;14:193.CrossRefPubMedPubMedCentral Chandrasiri UP, Fowkes FJ, Richards JS, Langer C, Fan YM, Taylor SM, et al. The impact of lipid-based nutrient supplementation on anti-malarial antibodies in pregnant women in a randomized controlled trial. Malar J. 2015;14:193.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Ashorn P, Alho L, Ashorn U, Cheung YB, Dewey KG, Harjunmaa U, et al. The impact of lipid-based nutrient supplement provision to pregnant women on newborn size in rural Malawi: a randomized controlled trial. Am J Clin Nutr. 2015;101:387–97.CrossRefPubMed Ashorn P, Alho L, Ashorn U, Cheung YB, Dewey KG, Harjunmaa U, et al. The impact of lipid-based nutrient supplement provision to pregnant women on newborn size in rural Malawi: a randomized controlled trial. Am J Clin Nutr. 2015;101:387–97.CrossRefPubMed
27.
Zurück zum Zitat Duffy MF, Byrne TJ, Elliott SR, Wilson DW, Rogerson SJ, Beeson JG, et al. Broad analysis reveals a consistent pattern of var gene transcription in Plasmodium falciparum repeatedly selected for a defined adhesion phenotype. Mol Microbiol. 2005;56:774–88.CrossRefPubMed Duffy MF, Byrne TJ, Elliott SR, Wilson DW, Rogerson SJ, Beeson JG, et al. Broad analysis reveals a consistent pattern of var gene transcription in Plasmodium falciparum repeatedly selected for a defined adhesion phenotype. Mol Microbiol. 2005;56:774–88.CrossRefPubMed
28.
Zurück zum Zitat Beeson JG, Brown GV. Plasmodium falciparum-infected erythrocytes demonstrate dual specificity for adhesion to hyaluronic acid and chondroitin sulfate A and have distinct adhesive properties. J Infect Dis. 2004;189:169–79.CrossRefPubMed Beeson JG, Brown GV. Plasmodium falciparum-infected erythrocytes demonstrate dual specificity for adhesion to hyaluronic acid and chondroitin sulfate A and have distinct adhesive properties. J Infect Dis. 2004;189:169–79.CrossRefPubMed
29.
Zurück zum Zitat Chandrasiri UP, Randall LM, Saad AA, Bashir AM, Rogerson SJ, Adam I. Low antibody levels to pregnancy-specific malaria antigens and heightened cytokine responses associated with severe malaria in pregnancy. J Infect Dis. 2014;209:1408–17.CrossRefPubMed Chandrasiri UP, Randall LM, Saad AA, Bashir AM, Rogerson SJ, Adam I. Low antibody levels to pregnancy-specific malaria antigens and heightened cytokine responses associated with severe malaria in pregnancy. J Infect Dis. 2014;209:1408–17.CrossRefPubMed
30.
Zurück zum Zitat Ataide R, Hasang W, Wilson DW, Beeson JG, Mwapasa V, Molyneux ME. Using an improved phagocytosis assay to evaluate the effect of HIV on specific antibodies to pregnancy-associated malaria. PLoS ONE. 2010;5:e10807.CrossRefPubMedPubMedCentral Ataide R, Hasang W, Wilson DW, Beeson JG, Mwapasa V, Molyneux ME. Using an improved phagocytosis assay to evaluate the effect of HIV on specific antibodies to pregnancy-associated malaria. PLoS ONE. 2010;5:e10807.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Alexander GR, Himes JH, Kaufman RB, Mor J, Kogan M. A United States national reference for fetal growth. Obstet Gynecol. 1996;87:163–8.CrossRefPubMed Alexander GR, Himes JH, Kaufman RB, Mor J, Kogan M. A United States national reference for fetal growth. Obstet Gynecol. 1996;87:163–8.CrossRefPubMed
32.
Zurück zum Zitat Filmer D, Pritchett LH. Estimating wealth effects without expenditure data–or tears: an application to educational enrollments in states of India. Demography. 2001;38:115–32.PubMed Filmer D, Pritchett LH. Estimating wealth effects without expenditure data–or tears: an application to educational enrollments in states of India. Demography. 2001;38:115–32.PubMed
33.
Zurück zum Zitat Feng G, Aitken E, Yosaatmadja F, Kalilani L, Meshnick SR, Jaworowski A, et al. Antibodies to variant surface antigens of Plasmodium falciparum-infected erythrocytes are associated with protection from treatment failure and the development of anemia in pregnancy. J Infect Dis. 2009;200:299–306.CrossRefPubMedPubMedCentral Feng G, Aitken E, Yosaatmadja F, Kalilani L, Meshnick SR, Jaworowski A, et al. Antibodies to variant surface antigens of Plasmodium falciparum-infected erythrocytes are associated with protection from treatment failure and the development of anemia in pregnancy. J Infect Dis. 2009;200:299–306.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Jaworowski A, Fernandes LA, Yosaatmadja F, Feng G, Mwapasa V, Molyneux ME, et al. Relationship between human immunodeficiency virus type 1 coinfection, anemia, and levels and function of antibodies to variant surface antigens in pregnancy-associated malaria. Clin Vaccine Immunol. 2009;16:312–9.CrossRefPubMedPubMedCentral Jaworowski A, Fernandes LA, Yosaatmadja F, Feng G, Mwapasa V, Molyneux ME, et al. Relationship between human immunodeficiency virus type 1 coinfection, anemia, and levels and function of antibodies to variant surface antigens in pregnancy-associated malaria. Clin Vaccine Immunol. 2009;16:312–9.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Staalsoe T, Shulman CE, Bulmer JN, Kawuondo K, Marsh K, Hviid L. Variant surface antigen-specific IgG and protection against clinical consequences of pregnancy-associated Plasmodium falciparum malaria. Lancet. 2004;363:283–9.CrossRefPubMed Staalsoe T, Shulman CE, Bulmer JN, Kawuondo K, Marsh K, Hviid L. Variant surface antigen-specific IgG and protection against clinical consequences of pregnancy-associated Plasmodium falciparum malaria. Lancet. 2004;363:283–9.CrossRefPubMed
36.
Zurück zum Zitat van den Broek NR, Jean-Baptiste R, Neilson JP. Factors associated with preterm, early preterm and late preterm birth in Malawi. PLoS ONE. 2014;9:e90128.CrossRefPubMedPubMedCentral van den Broek NR, Jean-Baptiste R, Neilson JP. Factors associated with preterm, early preterm and late preterm birth in Malawi. PLoS ONE. 2014;9:e90128.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Christensen DL, Kapur A, Bygbjerg IC. Physiological adaption to maternal malaria and other adverse exposure: low birth weight, functional capacity, and possible metabolic disease in adult life. Int J Gynaecol Obstet. 2011;115(Suppl 1):S16–9.CrossRefPubMed Christensen DL, Kapur A, Bygbjerg IC. Physiological adaption to maternal malaria and other adverse exposure: low birth weight, functional capacity, and possible metabolic disease in adult life. Int J Gynaecol Obstet. 2011;115(Suppl 1):S16–9.CrossRefPubMed
38.
Zurück zum Zitat Beeson JG, Drew DR, Boyle MJ, Feng G, Fowkes FJ, Richards JS. Merozoite surface proteins in red blood cell invasion, immunity and vaccines against malaria. FEMS Microbiol Rev. 2016;40:347–72.CrossRef Beeson JG, Drew DR, Boyle MJ, Feng G, Fowkes FJ, Richards JS. Merozoite surface proteins in red blood cell invasion, immunity and vaccines against malaria. FEMS Microbiol Rev. 2016;40:347–72.CrossRef
39.
Zurück zum Zitat Ismail MR, Ordi J, Menendez C, Ventura PJ, Aponte JJ, Kahigwa E, et al. Placental pathology in malaria: a histological, immunohistochemical, and quantitative study. Hum Pathol. 2000;31:85–93.CrossRefPubMed Ismail MR, Ordi J, Menendez C, Ventura PJ, Aponte JJ, Kahigwa E, et al. Placental pathology in malaria: a histological, immunohistochemical, and quantitative study. Hum Pathol. 2000;31:85–93.CrossRefPubMed
40.
Zurück zum Zitat Carbillon L, Challier JC, Alouini S, Uzan M, Uzan S. Uteroplacental circulation development: doppler assessment and clinical importance. Placenta. 2001;22:795–9.CrossRefPubMed Carbillon L, Challier JC, Alouini S, Uzan M, Uzan S. Uteroplacental circulation development: doppler assessment and clinical importance. Placenta. 2001;22:795–9.CrossRefPubMed
41.
Zurück zum Zitat Ataide R, Mwapasa V, Molyneux ME, Meshnick SR, Rogerson SJ. Antibodies that induce phagocytosis of malaria infected erythrocytes: effect of HIV infection and correlation with clinical outcomes. PLoS ONE. 2011;6:e22491.CrossRefPubMedPubMedCentral Ataide R, Mwapasa V, Molyneux ME, Meshnick SR, Rogerson SJ. Antibodies that induce phagocytosis of malaria infected erythrocytes: effect of HIV infection and correlation with clinical outcomes. PLoS ONE. 2011;6:e22491.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Radeva-Petrova D, Kayentao K, ter Kuile FO, Sinclair D, Garner P. Drugs for preventing malaria in pregnant women in endemic areas: any drug regimen versus placebo or no treatment. Cochrane Database Syst Rev. 2014;10:CD000169. Radeva-Petrova D, Kayentao K, ter Kuile FO, Sinclair D, Garner P. Drugs for preventing malaria in pregnant women in endemic areas: any drug regimen versus placebo or no treatment. Cochrane Database Syst Rev. 2014;10:CD000169.
43.
Zurück zum Zitat Richards JS, Arumugam TU, Reiling L, Healer J, Hodder AN, Fowkes FJ, et al. Identification and prioritization of merozoite antigens as targets of protective human immunity to Plasmodium falciparum malaria for vaccine and biomarker development. J Immunol. 2013;191:795–809.CrossRefPubMedPubMedCentral Richards JS, Arumugam TU, Reiling L, Healer J, Hodder AN, Fowkes FJ, et al. Identification and prioritization of merozoite antigens as targets of protective human immunity to Plasmodium falciparum malaria for vaccine and biomarker development. J Immunol. 2013;191:795–809.CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat Osier FH, Feng G, Boyle MJ, Langer C, Zhou J, Richards JS, et al. Opsonic phagocytosis of Plasmodium falciparum merozoites: mechanism in human immunity and a correlate of protection against malaria. BMC Med. 2014;12:108.CrossRefPubMedPubMedCentral Osier FH, Feng G, Boyle MJ, Langer C, Zhou J, Richards JS, et al. Opsonic phagocytosis of Plasmodium falciparum merozoites: mechanism in human immunity and a correlate of protection against malaria. BMC Med. 2014;12:108.CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat Boyle MJ, Reiling L, Feng G, Langer C, Osier FH, Aspeling-Jones H, et al. Human antibodies fix complement to inhibit Plasmodium falciparum invasion of erythrocytes and are associated with protection against malaria. Immunity. 2015;42:580–90.CrossRefPubMedPubMedCentral Boyle MJ, Reiling L, Feng G, Langer C, Osier FH, Aspeling-Jones H, et al. Human antibodies fix complement to inhibit Plasmodium falciparum invasion of erythrocytes and are associated with protection against malaria. Immunity. 2015;42:580–90.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Aboud S, Msamanga G, Read JS, Mwatha A, Chen YQ, Potter D, et al. Genital tract infections among HIV-infected pregnant women in Malawi, Tanzania and Zambia. Int J STD AIDS. 2008;19:824–32.CrossRefPubMedPubMedCentral Aboud S, Msamanga G, Read JS, Mwatha A, Chen YQ, Potter D, et al. Genital tract infections among HIV-infected pregnant women in Malawi, Tanzania and Zambia. Int J STD AIDS. 2008;19:824–32.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Thigpen MC, Filler SJ, Kazembe PN, Parise ME, Macheso A, Campbell CH, et al. Associations between peripheral Plasmodium falciparum malaria parasitemia, human immunodeficiency virus, and concurrent helminthic infection among pregnant women in Malawi. Am J Trop Med Hyg. 2011;84:379–85.CrossRefPubMedPubMedCentral Thigpen MC, Filler SJ, Kazembe PN, Parise ME, Macheso A, Campbell CH, et al. Associations between peripheral Plasmodium falciparum malaria parasitemia, human immunodeficiency virus, and concurrent helminthic infection among pregnant women in Malawi. Am J Trop Med Hyg. 2011;84:379–85.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Yatich NJ, Jolly PE, Funkhouser E, Agbenyega T, Rayner JC, Ehiri JE, et al. The effect of malaria and intestinal helminth coinfection on birth outcomes in Kumasi. Ghana. Am J Trop Med Hyg. 2010;82:28–34.CrossRefPubMed Yatich NJ, Jolly PE, Funkhouser E, Agbenyega T, Rayner JC, Ehiri JE, et al. The effect of malaria and intestinal helminth coinfection on birth outcomes in Kumasi. Ghana. Am J Trop Med Hyg. 2010;82:28–34.CrossRefPubMed
Metadaten
Titel
Association between malaria immunity and pregnancy outcomes among Malawian pregnant women receiving nutrient supplementation
verfasst von
Upeksha P. Chandrasiri
Freya J. I. Fowkes
James G. Beeson
Jack S. Richards
Steve Kamiza
Kenneth Maleta
Per Ashorn
Stephen J. Rogerson
Publikationsdatum
01.12.2016
Verlag
BioMed Central
Erschienen in
Malaria Journal / Ausgabe 1/2016
Elektronische ISSN: 1475-2875
DOI
https://doi.org/10.1186/s12936-016-1597-7

Weitere Artikel der Ausgabe 1/2016

Malaria Journal 1/2016 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.