Skip to main content
  • Research article
  • Open access
  • Published:

Factors associated with sub-microscopic placental malaria and its association with adverse pregnancy outcomes among HIV-negative women in Dar es Salaam, Tanzania: a cohort study

Abstract

Background

Malaria infection during pregnancy has negative health consequences for both mothers and offspring. Sub-microscopic malaria infection during pregnancy is common in most African countries. We sought to identify factors associated with sub-microscopic placental malaria, and its association with adverse pregnancy outcomes among HIV-negative pregnant women in Dar es Salaam, Tanzania.

Methods

We recruited a cohort of pregnant women during their first trimester and assessed for the occurrence of placental malaria and pregnancy outcomes. The follow-up was done monthly from recruitment until delivery. Histopathology placental malaria positive results were defined as the presence of malaria pigment or parasitized erythrocytes on the slide (histology-positive (HP)), and the sub-microscopic placental infection was defined as positive Plasmodium falciparum DNA by polymerase chain reaction (DNA PCR) amplification in a negative histopathology test. Adverse pregnancy outcomes investigated included low birth weight (birth weight below 2.5 kg), prematurity (live birth below 37 weeks), and small-for-gestational-age (SGA) (live born with a birth weight below 10th percentile for gestational age and sex). Weighted baseline category logit, log-binomial, and log-Poisson models were used to assess factors associated with placental malaria, and its association with adverse pregnancy outcomes.

Results

Among 1115 women who had histopathology and DNA PCR performed, 93 (8%) had HP placental infection, and 136 (12%) had the sub-microscopic placental infection. The risk of sub-microscopic placental malaria was greater in women who did not use mosquito prevention methods such as bed nets, fumigation, or mosquito coils (odds ratio (OR) = 1.75; 95% confidence interval (CI): 1.05–2.92; P = 0.03) and in women who were anemic (OR = 1.59; 95% CI: 1.20–2.11; P = 0.001). Women who were underweight had reduced odds of sub-microscopic placental malaria infection (OR = 0.33; 95% CI: 0.17–0.62; P = 0.001). Women who were overweight/obese had 1.48 times higher the odds of HP placental malaria compared to normal weight (OR = 1.48; 95% CI: 1.03–2.11; P = 0.03). HP placental malaria infection was associated with an increased risk of SGA births (RR = 1.30, 95% CI: 0.98–1.72, P = 0.07). In contrast, the sub-microscopic infection was associated with a reduced risk of SGA births (RR = 0.61, 95% CI: 0.43–0.88, P = 0.01). Placental malaria was not associated with low birth weight or prematurity.

Conclusion

Malaria prevention methods and maternal nutrition status during early pregnancy were important predictors of sub-microscopic placental malaria. More research is needed to understand sub-microscopic placental malaria and the possible mechanisms mediating the association between placental malaria and SGA.

Peer Review reports

Background

Worldwide, 75,000 to 200,000 infant deaths every year are attributable to malaria infection during pregnancy (MiP) [1]. Approximately 25 million pregnant women are at risk of Plasmodium falciparum (P. falciparum) infections each year in sub-Saharan Africa [2]. Twenty-six percent of severe anemia during pregnancy is attributable to MiP, and the proportion of maternal deaths linked to malaria infections ranges from 0.5 to 23% in hospital-based studies and 3 to 18% in community-based studies [1, 2]. MiP also increases the risk of various adverse pregnancy outcomes such as low birth weight (LBW), prematurity, intrauterine growth retardation (IUGR), and stillbirth [1,2,3]. Further, the offspring of mothers diagnosed with placental malaria also have an increased risk of all-cause anemia and malaria infection during infancy [4, 5].

Histopathology examination of stained placental biopsies is considered as the gold standard for diagnosis of placental malaria (hereafter referred to as histology-positive (HP) placental malaria), but this test is impractical or not available in some settings [6, 7]. Previous studies have pointed out that pregnant women often have low parasite densities, and the use of highly sensitive diagnostic methods such a DNA polymerase chain reaction (DNA PCR) (referred to as sub-microscopic placental malaria) has been recommended to enhance detection of placental malaria [6]. Sub-microscopic MiP remains common in most African countries including Tanzania [8,9,10,11], even though there was a 41% reduction of the malaria incidence rate in sub-Saharan Africa for the period of 2000–2015 [12]. The decreasing proportion of individuals presenting with clinical malaria infection over time has been linked with the scale-up of effective malaria interventions including insecticide-treated nets, effective case management, and indoor residual spraying [13].

Peripheral sub-microscopic malaria is strongly associated with placental malaria and low maternal hemoglobin levels [14, 15], but there are contradictory findings about the association between placental malaria and adverse pregnancy outcomes [11, 16, 17]. Cottrell et al. [16] found that sub-microscopic malaria infection during early pregnancy was associated with adverse pregnancy outcomes, whereas infection detected at delivery in the placenta and peripheral blood sample was not. In contrast, Cohee et al. [14] did not find an association between adverse pregnancy outcomes and peripheral sub-microscopic infection detected during pregnancy, and at delivery, and sub-microscopic placental malaria. As a result, the WHO has recommended more studies to investigate the association between sub-microscopic infection and adverse pregnancy outcomes [18].

Limited evidence is available on the association between adverse pregnancy outcomes and placental malaria infections (particularly sub-microscopic variant) in tropical African cities such as Dar-es-Salaam, Tanzania, where the malaria burden has been significantly reduced, but low-level transmission remains. We, therefore designed a study with the objectives of identifying factors associated with sub-microscopic placental malaria and examined its association with LBW, prematurity, and small-for-gestational-age (SGA) births among HIV-negative pregnant women in their first and second pregnancy in Dar Es Salaam, Tanzania.

Methods

Study design and participants

We used data from a trial that examined the effect of prenatal vitamin A and zinc supplements on malaria in pregnancy among women in their first or second pregnancy [19]. In brief, women were randomized to placebo (625 women), vitamin A alone (625 women), zinc alone (625 women), and vitamin A and zinc (625 women). Women were enrolled in their first trimester (below 13 weeks of gestation) and followed up monthly until delivery for placental malaria and adverse pregnancy outcomes. Gestational age was estimated using the last menstrual period. Recruitment occurred between December 2010 and September 2013, and follow-up ended in June 2014. Trial recruitment sites were in Ilala and Temeke districts, Dar-es-Salaam, Tanzania. All women included in the trial received iron (60 mg daily) and folic acid (5 mg daily) supplements, a voucher for insecticide-treated bed nets, and sulphadoxine-pyrimethamine (SP) (1500 mg sulfadoxine and 75 mg pyrimethamine) as per Tanzanian standard of care.

Socio-demographic and clinical variables were assessed at recruitment (baseline); these include age, parity, twin pregnancy status, years of education, marital status, gestational age, employment status, and use of fumigation, mosquito coil or bed nets at night. Selected possessions were asked about, from which a Filmer-Pritchet wealth index was computed [20]. Body mass index (BMI) (weight in kg divided by height (in meters) squared) and hemoglobin (in g/dL) were assessed. Age was classified into ≤24, 25–34 and ≥ 35 years, and years of education were categorized into 0–7 (no education to primary school education), 8–11 (secondary school education), and ≥ 12 years (high school and above education). The wealth index was grouped into quartiles, BMI was classified as < 18.5 (underweight), 18.5–24.9 (normal weight) or ≥ 25 kg/m2 (overweight/obese) [21], and hemoglobin was classified as ≥11.0 g/dL (not anemic) or < 11.0 g/dL (anemic) [22].

Outcomes assessed at delivery

Placental malaria was evaluated using both histopathology and DNA PCR. HP placental malaria was defined as the presence of malaria pigment or parasitized erythrocytes on the slide, and sub-microscopic placental malaria infection was defined as positive P. falciparum DNA by PCR amplification in samples negative for histopathology test. Details on the two methods have been previously described [19]. Briefly, tissue from fresh sampled placentas was collected during delivery and the collected sample was divided for both uses. Histopathologic infections were defined as the presence of malaria pigment or parasitized erythrocytes on tissue from a central parenchymal section of the placenta. Placenta samples were stained and then examined using polarized light microscopy. A single placental histopathologist classified all slides, and quality control of diagnoses were done externally in a subset of 100 slides.

For the nucleic acid studies, tissue was stabilized in RNA later (Qiagen, Hilden, Germany) and homogenized. Genomic DNA was extracted using DNeasy (Qiagen). Taqman® qRT-PCR (Thermo Fisher Scientific, Waltham, MA) was used for amplification using published primer and probe sets (P. falciparum-specific [23] and general Plasmodium 18S rRNA genes [24]). The tissue was then tested for PCR inhibitors. Positive and negative controls were included on each plate for quality, and the minimum parasite density detection level was 0.000362 parasites/μL [24]. At recruitment and during follow-up peripheral malaria infection was diagnosed using microscopy of thin and thick Giemsa staining blood smears.

Adverse pregnancy outcomes investigated in this study were LBW (birth weight below 2.5 kg), SGA births (live birth weight below 10th percentile for the exact day of gestation and sex based on INTERGROWTH-21st standards [25, 26]) as a proxy outcome of IUGR, and prematurity (live birth before 37 weeks of gestation).

Statistical analysis

Factors associated with placental malaria infection at delivery

Baseline continuous variables were presented as medians and their interquartile ranges (IQR) and categorical variables as numbers and their percentage. To assess factors associated with placental malaria, baseline category logit regression models were used. Since we were unable to obtain placental samples for over 40% of women recruited for the original study due to changes in delivery plans, we weighted observations to account for non-response bias by the inverse of the probability of women having placental samples (for both tests) as determined by a multivariable logistic regression model [27]. In building the multivariable models, variables were selected based on univariable analysis. Variables with p-values less than 0.2 were considered potential variables for the multivariable models (these included employment, gestational age at baseline, and BMI). Other covariates were included based on prior knowledge of the topic (years of education, wealth index, gravidity, mosquito prevention methods, anemia status). Due to the positive correlation between maternal age and parity in this population (ρ = 0.3 with P < 0.0001) maternal age was not included in any multivariable models. The trial regimens were forced into the multivariable models. Lastly, results were presented as odds ratio (OR) and their 95% confidence intervals.

The association between placental malaria and adverse pregnancy outcomes

In each outcome, the distributions of outcomes by exposure were presented in numbers and their percentage. To investigate the relationship of placental malaria with LBW, premature birth, and SGA birth, weighted log-binomial models, and log-Poisson models (with weights obtained from logistic regression models as described above) were used, [27, 28]. For HP placental malaria, the logistic regression model used to estimate the probabilities for weights was not stable due to a few cases in one of the age categories. In this case, we used the Firth penalized maximum likelihood estimation method [29]. All models were adjusted for employment status, years of education, wealth quartiles, trial regimen, gravidity, use of malaria interventions, twin pregnancy status, BMI, and anemia status at baseline. Results were presented using relative risk (RR) and their 95% confidence intervals.

Analyses were performed among women with both tests done. In this case, placental diagnoses were classified as HP placental malaria, sub-microscopic positive placental malaria when histopathology was negative, and negative for both diagnoses. The level of significance was 0.05, and all data analyses were conducted in SAS version 9.4 (SAS Institute Inc., Cary, NC, USA).

Results

The original trial recruited 2500 pregnant women in their first trimester (Fig. 1). Of these 2434 had known birth outcomes and among those with known birth outcomes, we excluded 1030 women due to missing placentas. Among women with placenta samples, 1115 (79%) women had samples for both histology and PCR, 246 had samples only for histology and 43 had samples only for DNA PCR. Baseline characteristics in the groups of women with and without placenta samples were similar (Table 1). Among women included in the analysis, at baseline, the median age was 22 years with IQR [20,21,22,23,24,25] and the median gestational age was 10 weeks with IQR [8,9,10,11,12] (Table 2). Further, the majority were unemployed (53%), married or cohabiting (90%), with 0–7 years of education level (72%). Almost all women had singleton pregnancies (98%) and 93% used either fumigation or mosquito coils or bed nets at night. Underweight and peripheral malaria was less frequent (11 and 2% respectively), while anemia was very common (29%).

Fig. 1
figure 1

Participants flow chart

Table 1 The distribution of baseline characteristics among pregnant women with and without placenta sample for placental malaria diagnosis, 2010–2014 Dar-es-Salaam, Tanzania
Table 2 Distribution of baseline characteristics of pregnant women by placental malaria outcome, 2010–2014, Dar-es-Salaam, Tanzania

Among 1404 women, there were 1115 (79%) pregnant women with sufficient placenta samples for both diagnostic methods, 886 (80%) tested negative by both methods, 93 (8%) had HP placental malaria, and 136 (12%) had sub-microscopic placental malaria.

Factors associated with placental malaria infection at delivery

The distribution of baseline characteristics was presented in Table 2. According to both diagnostic methods, participants with placental malaria were more frequently unemployed (59% for DNA PCR and 65% for histopathology versus 51% for negative results), less educated (78% for DNA PCR and 76% for histopathology versus 71% for negative results), and anemic (33% for DNA PCR and 36% for histopathology versus 28% for negative results). Placental malaria was also somewhat more common among women not using mosquito prevention methods such as bed nets, fumigation, and coils (10% for DNA PCR and 8% for histopathology versus 6% for negative results).

Results of the multinomial model for factors associated with placental malaria are summarized in Table 3. Being employed and other forms of employment were associated with reduced odds of HP placental malaria compared to unemployed women (OR = 0.69; 95% CI: 0.47–1.00; P = 0.05 and OR = 0.49; 95% CI: 0.28–0.86; P = 0.01 respectively). Compared to those with a higher education level, pregnant women with the least education (0–7 years of education) had an increased odd of sub-microscopic placental malaria (OR = 2.21; 95% CI: 0.99–4.97; P = 0.05). Similar findings for HP placental malaria infection were observed but estimates were not stable due to the low number of outcomes. Pregnant women who did not use mosquito prevention methods such as fumigation, mosquito coil, or a bed net at night had a 75% increased risk of sub-microscopic placental malaria (OR = 1.75; 95% CI: 1.05–2.92; P = 0.03).

Table 3 Factors associated with placental malaria among women with sample for both diagnosis, 2010–2014, Dar es Salaam, Tanzania (n = 941)a, b

Being underweight was associated with reduced odds of sub-microscopic placental malaria compared to normal BMI (OR = 0.33; 95% CI: 0.17–0.62; P = 0.001). In contrast, being overweight/obese was associated with increased odds of HP placental malaria (OR = 1.48; 95% CI: 1.03–2.11; P = 0.03). Compared to pregnant women who were not anemic in the first trimester, anemic pregnant women had increased odds of sub-microscopic (OR = 1.59; 95% CI: 1.20–2.11; P = 0.001) and HP placental malaria (OR = 1.70; 95% CI: 1.22–2.38; P = 0.002).

The association between placental malaria and adverse pregnancy outcomes

The prevalence of LBW was 8% among HP positive women and 5% among sub-microscopic positive women (Table 4). LBW was not associated with placental malaria by both diagnostic method. Premature births were common among women with placental malaria by both diagnostic methods (28 and 25% for HP and sub-microscopic placental malaria respectively). Compared to women negative for placental malaria, women with placental malaria infection had a slightly elevated risk of prematurity, although no statistical evidence for the association (RR = 1.16; CI: 0.89–1.52; P = 0.26 for HP placental malaria and RR = 1.14; CI: 0.90–1.45; P = 0.27 for sub-microscopic placental malaria).

Table 4 Relationship between low birth weight, premature, and small for gestational age births and placental malaria, 2010–2014, Dar es Salaam, Tanzaniaa,b

A high prevalence of SGA births was found in women with HP placental malaria (23%), while women with sub-microscopic placental malaria had a low prevalence of SGA births (12%). Compared to women with negative results for both tests, HP placental malaria infection was associated with increased risk of SGA births (RR = 1.30; 95% CI: 0.98–1.72; P = 0.07), while sub-microscopic infection was associated with reduced risk of SGA births (RR = 0.61; 95% CI: 0.43–0.88; P = 0.01).

Discussion

The overall prevalence of placental malaria was 8, and 12% for HP and sub-microscopic placental malaria respectively. Being underweight was associated with a reduced risk of sub-microscopic placental malaria while being overweight/obese was associated with a higher risk of HP placental malaria infection. Not using anti-malarial preventive measures and being anemic were associated with increased risk of sub-microscopic placental malaria. We noted an increased risk of SGA births in women with HP placental malaria and a decreased risk of SGA among women with sub-microscopic placental malaria. There was no statistically significant increased risk of premature births in women with either HP or sub-microscopic placental malaria.

Increased premature births among women infected with placental malaria have been observed in Uganda [30] and Papua New Guinea [31]. The Papua New Guinea [31] study further showed that chronic placental malaria was associated with an increased risk of prematurity. A potential mechanism for the association being the stimulation of early labor by the active immune triggered in malaria-infected placentas [32]. However, in the current study, we found a non-significant increased risk of premature births among women with placental malaria compared to women without the infection. The difference could be partly explained by differences in malaria transmission rates. In general urban areas receive fewer infectious bites per year compared to rural settings [33], and in 2009 the entomological inoculation rate in Dar-es-Salaam was 1.3 infectious bites per person per year [34].

The occurrence of IUGR in pregnant women infected with malaria is related to nutrient supply to the fetus. A study of the histopathology of infected placentas showed that the thickening of the cytotrophoblastic membranes may interfere with the nutrient supply to the fetus, resulting in growth retardation [32]. The association we observed between HP placental malaria and SGA was consistent with studies done in Uganda [30], and Gambia [35], although the latter did not account for potential confounders. A study conducted in the Democratic Republic of Congo showed that IUGR was highly associated with three or more cumulative infections, and there was a 2–8-fold increased risk of IUGR among women with evidence of under-nutrition [36]. However, we could not investigate a similar interaction effect between nutritional status and infection in the current study because few women were classified as underweight.

Our findings that sub-microscopic placental malaria was associated with a reduced risk of SGA birth was similar to results from a study done in Benin [37] despite differences in diagnostic methods and timing. The potential mechanism for this finding is unknown, but we speculate that it may involve the acquisition of antibodies in the placenta that inhibited the binding of P. falciparum-infected erythrocytes to the placental receptor chondroitin sulfate A (CSA) [38]. High levels of immunoglobulin G (IgG) against one of the placental isolates were previously found to be associated with increased birth weight among women who experienced at least one episode of malaria during pregnancy [39]. Further, Duffy and Fried [38] found that antibodies to placental parasites were related to decreased placental parasitemia and increased birth weight. Although our study participants included women in their first pregnancy who presumably lack immunity to CSA-binding parasite [40], the relationship may be plausible in the context of chronic or repeated infections in the same pregnancy. Findings from a study done in Kenya, that assessed pregnancy-specific P. falciparum immune responses among primiparous women showed low-to-medium pregnancy-associated malaria IgG concentration among women with acute or chronic MiP, which was an indication of the development of the immune response to infection [41]. On other hand, malaria infection during early pregnancy has been linked to SGA births [42]. Sub-microscopic placental malaria infection being interpreted to be a recent infection [16], we also suspect that the infections would have been those that occurred in the weeks preceding birth. Therefore, the association observed could have happened by chance.

Being underweight weakens antibody-mediated anti-malarial immunity [3]. This study, however, found that underweight was associated with reduced odds of sub-microscopic placental malaria. A possible explanation of this finding could be that pregnant women with macronutrient deficiencies (i.e. energy, fat, and protein) are at increased risk of micronutrient deficiency especially iron deficiency [3]. This was found in a study of Indonesian and Ghanaian pregnant women, in which the risk of anemia during early pregnancy was higher in women with lower levels of BMI during early pregnancy [43]. Being underweight may be associated with a reduced risk of placental malaria because of concurrent iron deficiency during pregnancy, which has been associated with reduced risk of malaria infection even during pregnancy [3, 44, 45]. Others have noted that pregnant women who are underweight during early pregnancy may already have been exposed to malaria infection and acquired pregnancy-specific immunity that protects them from further infection later in the same pregnancy [46].

We also noted the increased odds of HP placental malaria in women classified as overweight/obese. This finding was similar to that of a previous study done in Sweden [47], which found an increased risk of severe malaria among obese participants compared to normal weight participants, even though the study focused on the general adult population. Given the increased burden of overweight and obesity among women of reproductive age in urban Africa [48], our findings suggest that malaria control programs should also consider the nutrition status of the targeted population.

Being anemic during the first trimester was associated with an increased risk of sub-microscopic placental malaria, consistent with a study done in Malawi [15], but inconsistent with a study done in Cameroon [49]. Since anemia was detected at recruitment there is a possibility that low hemoglobin levels observed at recruitment resulted from prior malaria infections. Malaria infection detected in the placenta typically first occurred in the periphery many months earlier (possibly in the first trimester) resulting in anemia. Therefore, establishing a temporal pathway for the association observed in the current study is not possible given the nature of the study.

Intermittent preventive treatment in pregnancy (IPTp) with SP is among the strategies used for preventing adverse pregnancy outcomes associated with MiP in Tanzania [50], and it is available as standard of care for women attending antenatal care clinics. It has been shown that SP-IPTp can treat even sub-microscopic infection, but does not prevent new infections [14]. Hence, other malaria vector control strategies such as mosquito coils [51], fumigation [52], and treated bed nets [52, 53] are also used for malaria control. We found that not using these preventive methods was associated with an increased risk of sub-microscopic placental malaria. These findings support the protective role of alternative malaria control measures [52, 54]. However, others have suggested that mosquito coils have low mosquito repellency ability [51], and their effect on malaria prevention is not well established [55].

To our knowledge, our study is one of the first large studies to investigate the associations of placental malaria and adverse pregnancy outcomes among women in their first and second pregnancy in urban Tanzania. However, the study has some limitations first, placental malaria was diagnosed at delivery. Establishing the temporal relationship between malaria and baseline characteristics such as maternal anemia and underweight is therefore challenging, especially if malaria infection may have happened before enrolment. However, past infections diagnosed at delivery are informative with respect to the presence of MiP [6]. We also had very few women diagnosed with peripheral malaria at baseline (n = 41). DNA PCR diagnosis for placental malaria, though highly sensitive compared to histopathology, may detect both viable, and non-viable or gametocytes [6], which may complicate the interpretation of our results. However, sub-microscopic gametocytes have an important role in malaria transmission and pregnant women have been found to be reservoirs of sub-microscopic gametocytes [12]. It has been suggested that the majority of sub-microscopic malaria infection may reflect recent infection based on genotyping findings [16]. Therefore, learning about the relevance of this infection on clinical outcomes is crucial. Also, gestational age was estimated using the last menstrual period. This method is known to be vulnerable to misclassification of prematurity compared to ultrasound [56, 57]. However, in Tanzania ultrasound is not routinely available and where available it is not affordable for the majority. Lastly, approximately half of the study participants did not have a placental sample for placental malaria infection. To address potential selection bias due to missing data, we applied a weighted analysis.

In summary, social determinants such as low educational level may influence the understanding and interpretation of anti-malaria messages delivered at antenatal clinics. It is crucial that these programs tailor their messages and campaigns such that even those with low or no education are well informed. Further, even though over 90% reported the use of at least one malaria preventive measure, more effort should be directed to pregnant women who do not use recommended preventive measures. The integration of nutrition services that target both extremes of the malnutrition spectrum is warranted. Lastly, further research is also needed to understand sub-microscopic placental malaria and the underlying malaria immune response mechanisms that might be linked with the protective effect of SGA births among those infected.

Availability of data and materials

The datasets generated and/or analyzed are not publicly available because they contain sensitive information of participants but are available from the corresponding author on reasonable request.

Abbreviations

HP:

Histology-positive

PCR:

Polymerase Chain Reaction

SGA:

Small-for-gestational age

IQR:

Interquartile range

RR:

Relative risk

CI:

Confidence interval

P.falciparum :

Plasmodium falciparum

MiP:

Malaria infection during pregnancy

HIV:

Human immunodeficiency virus

SP:

Sulphadoxine-pyrimethamine

LBW:

Low birth weight

BMI:

Body mass index

RNA:

Ribonucleic acid

qRT-PCR:

Quantitative real-time polymerase chain reaction

IUGR:

Intrauterine growth retardation

IgG:

Immunoglobulin G

CSA:

Chondroitin sulfate A

IPTp:

Intermittent preventive treatment in pregnancy

References

  1. Steketee RW, Nahlen BL, Parise ME, Menendez C. The Burden of Malaria in Pregnancy in Malaria-Endemic Areas. American Society of Tropical Medicine and Hygiene; 2001 [cited 2019 Aug 16]. Available from: https://www.ncbi.nlm.nih.gov/books/NBK2623/.

    Google Scholar 

  2. Desai M, ter Kuile FO, Nosten F, McGready R, Asamoa K, Brabin B, et al. Epidemiology and burden of malaria in pregnancy. Lancet Infect Dis. 2007;7(2):93–104 [cited 2019 Aug 16]. Available from: https://linkinghub.elsevier.com/retrieve/pii/S147330990770021X.

    Article  PubMed  Google Scholar 

  3. Unger HW, Ashorn P, Cates JE, Dewey KG, Rogerson SJ. Undernutrition and malaria in pregnancy – a dangerous dyad? BMC Med. 2016;14(1):142 [cited 2019 Aug 16]. Available from: doi: https://doi.org/10.1186/s12916-016-0695-2.

  4. Lohoue J, Leke RFG, Leke R, Tako EA, Zhou A, Taylor DW. RISK FACTORS FOR PLACENTAL MALARIA AND ITS EFFECT ON PREGNANCY OUTCOME IN YAOUNDE, CAMEROON. Am J Trop Med Hygiene . 2005;72(3):236–242 [cited 2019 Aug 16]. Available from: http://www.ajtmh.org/content/journals/10.4269/ajtmh.2005.72.236.

  5. Mutabingwa TK, Bolla MC, Li J-L, Domingo GJ, Li X, Fried M, et al. Maternal Malaria and Gravidity Interact to Modify Infant Susceptibility to Malaria. PLoS Medicine. 2005;2(12):e407 [cited 2019 Aug 16]. Available from: doi: https://doi.org/10.1371/journal.pmed.0020407.

  6. Kattenberg JH, Ochodo EA, Boer KR, Schallig HD, Mens PF, Leeflang MM. Systematic review and meta-analysis: rapid diagnostic tests versus placental histology, microscopy and PCR for malaria in pregnant women. Malaria J. 2011;10(1):321 [cited 2019 Aug 16]. Available from. https://doi.org/10.1186/1475-2875-10-321.

    Article  Google Scholar 

  7. Zhou Z, Mitchell RM, Gutman J, Wiegand RE, Mwandama DA, Mathanga DP, et al. Pooled PCR testing strategy and prevalence estimation of submicroscopic infections using Bayesian latent class models in pregnant women receiving intermittent preventive treatment at Machinga District Hospital, Malawi, 2010. Malaria J. 2014;13(1):509 [cited 2019 Aug 16]. Available from. https://doi.org/10.1186/1475-2875-13-509.

    Article  Google Scholar 

  8. Minja DT, Schmiegelow C, Oesterholt M, Magistrado PA, Boström S, John D, et al. Reliability of rapid diagnostic tests in diagnosing pregnancy-associated malaria in north-eastern Tanzania. Malaria J. 2012;11(1):211 [cited 2019 Aug 16]. Available from. https://doi.org/10.1186/1475-2875-11-211.

    Article  Google Scholar 

  9. Valente B, Campos PA, do Rosário VE, Varandas L, Silveira H. Prevalence and risk factors of Plasmodium falciparum infections in pregnant women of Luanda, Angola: P. falciparum infections in pregnant women in Luanda. Trop Med Int Health. 2011;16(10):1206–14 [cited 2019 Aug 16]. Available from: http://doi.wiley.com/10.1111/j.1365-3156.2011.02830.x.

    Article  PubMed  Google Scholar 

  10. Mayor A, Serra-Casas E, Bardají A, Sanz S, Puyol L, Cisteró P, et al. Sub-microscopic infections and long-term recrudescence of Plasmodium falciparum in Mozambican pregnant women. Malaria J. 2009;8(1):9 [cited 2019 Aug 16]. Available from: doi: https://doi.org/10.1186/1475-2875-8-9.

  11. Omer SA, Noureldein AN, Eisa H, Abdelrahim M, Idress HE, Abdelrazig AM, et al. Impact of Submicroscopic Plasmodium falciparum Parasitaemia on Maternal Anaemia and Low Birth Weight in Blue Nile State, Sudan. J Trop Med. 2019;2019:1–7 [cited 2019 Aug 16]. Available from: https://www.hindawi.com/journals/jtm/2019/3162378/.

    Article  Google Scholar 

  12. Lamptey H, Ofori MF, Kusi KA, Adu B, Owusu-Yeboa E, Kyei-Baafour E, et al. The prevalence of submicroscopic Plasmodium falciparum gametocyte carriage and multiplicity of infection in children, pregnant women and adults in a low malaria transmission area in Southern Ghana. Malaria J. 2018;17(1):331 [cited 2019 Aug 16]. Available from. https://doi.org/10.1186/s12936-018-2479-y.

    Article  Google Scholar 

  13. Bhatt S, Weiss DJ, Cameron E, Bisanzio D, Mappin B, Dalrymple U, et al. The effect of malaria control on Plasmodium falciparum in Africa between 2000 and 2015. Nature. 2015 Oct 8;526(7572):207–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Cohee LM, Kalilani-Phiri L, Boudova S, Joshi S, Mukadam R, Seydel KB, et al. Submicroscopic malaria infection during pregnancy and the impact of intermittent preventive treatment. Malaria J. 2014;13(1):274 [cited 2019 Aug 16]. Available from. https://doi.org/10.1186/1475-2875-13-274.

    Article  Google Scholar 

  15. 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(9):e74643 [cited 2019 Aug 16]. Available from: https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0074643.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Cottrell G, Moussiliou A, Luty AJF, Cot M, Fievet N, Massougbodji A, et al. Submicroscopic Plasmodium falciparum Infections Are Associated With Maternal Anemia, Premature Births, and Low Birth Weight. Clin Infect Dis. 2015;60(10):1481–1488 [cited 2019 Aug 16]. Available from: https://academic.oup.com/cid/article/60/10/1481/338982.

  17. Adegnika AA, Verweij JJ, Agnandji ST, Chai SK, Breitling LP, Ramharter M, et al. Microscopic and sub-microscopic Plasmodium falciparum infection, but not inflammation caused by infection, is associated with low birth weight. Am J Trop Med Hyg. 2006;75(5):798–803.

    Article  CAS  PubMed  Google Scholar 

  18. mpac-sept2015-erg-mip-report.pdf [Internet]. [cited 2020 May 3]. Available from: https://www.who.int/malaria/mpac/mpac-sept2015-erg-mip-report.pdf.

  19. Darling AM, Mugusi FM, Etheredge AJ, Gunaratna NS, Abioye AI, Aboud S, et al. Vitamin A and Zinc Supplementation among Pregnant Women to Prevent Placental Malaria: A Randomized, Double-Blind, Placebo-Controlled Trial in Tanzania. Am J Trop Med Hygiene. 2017:16–0599 [cited 2019 Aug 16]. Available from: http://www.ajtmh.org/lookup/doi/10.4269/ajtmh.16-0599.

  20. Filmer D, Pritchett LH. Estimating Wealth Effects without Expenditure Data-or Tears: An Application to Educational Enrollments in States of India. Demography. 2001;38(1):115–32 [cited 2019 Aug 16]. Available from: https://www.jstor.org/stable/3088292.

    CAS  PubMed  Google Scholar 

  21. Body mass index - BMI [Internet]. 2019 [cited 2019 Dec 27]. Available from: http://www.euro.who.int/en/health-topics/disease-prevention/nutrition/a-healthy-lifestyle/body-mass-index-bmi.

  22. WHO_NMH_NHD_MNM_11.1_eng.pdf [Internet]. [cited 2019 Dec 27]. Available from: https://apps.who.int/iris/bitstream/handle/10665/85839/WHO_NMH_NHD_MNM_11.1_eng.pdf?sequence=22&isAllowed=y.

  23. Rosanas-Urgell A, Mueller D, Betuela I, Barnadas C, Iga J, Zimmerman PA, et al. Comparison of diagnostic methods for the detection and quantification of the four sympatric Plasmodium species in field samples from Papua New Guinea. Malaria J. 2010;9(1):361 [cited 2019 Aug 16]. Available from. https://doi.org/10.1186/1475-2875-9-361.

    Article  CAS  Google Scholar 

  24. Kamau E, Tolbert LS, Kortepeter L, Pratt M, Nyakoe N, Muringo L, et al. Development of a Highly Sensitive Genus-Specific Quantitative Reverse Transcriptase Real-Time PCR Assay for Detection and Quantitation of Plasmodium by Amplifying RNA and DNA of the 18S rRNA Genes. J Clin Microbiol. 2011;49(8):2946–53 [cited 2019 Aug 16]. Available from: https://jcm.asm.org/content/49/8/2946.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Villar J, Cheikh Ismail L, Victora CG, Ohuma EO, Bertino E, Altman DG, et al. International standards for newborn weight, length, and head circumference by gestational age and sex: the newborn cross-sectional study of the INTERGROWTH-21st project. Lancet. 2014;384(9946):857–68.

    Article  PubMed  Google Scholar 

  26. Villar J, Giuliani F, Fenton TR, Ohuma EO, Ismail LC, Kennedy SH, et al. INTERGROWTH-21st very preterm size at birth reference charts. Lancet. 2016;387(10021):844–5.

    Article  PubMed  Google Scholar 

  27. Seaman SR, White IR. Review of inverse probability weighting for dealing with missing data. Stat Methods Med Res. 2013;22(3):278–95 [cited 2019 Aug 16]. Available from. https://doi.org/10.1177/0962280210395740.

    Article  PubMed  Google Scholar 

  28. Spiegelman D, Hertzmark E. Easy SAS Calculations for Risk or Prevalence Ratios and Differences. Am J Epidemiol. 2005;162(3):199–200 [cited 2019 Aug 16]. Available from: https://academic.oup.com/aje/article/162/3/199/171116.

    Article  PubMed  Google Scholar 

  29. Firth D. Bias Reduction of Maximum Likelihood Estimates. Biometrika. 1993;80(1):27–38 [cited 2019 May 6]. Available from: https://www.jstor.org/stable/2336755.

    Article  Google Scholar 

  30. Kapisi J, Kakuru A, Jagannathan P, Muhindo MK, Natureeba P, Awori P, et al. Relationships between infection with Plasmodium falciparum during pregnancy, measures of placental malaria, and adverse birth outcomes. Malar J. 2017;16(1):400.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lufele E, Umbers A, Ordi J, Ome-Kaius M, Wangnapi R, Unger H, et al. Risk factors and pregnancy outcomes associated with placental malaria in a prospective cohort of Papua New Guinean women. Malar J. 2017;16(1):427.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Uneke CJ. Impact of placental Plasmodium falciparum malaria on pregnancy and perinatal outcome in sub-Saharan Africa: II: effects of placental malaria on perinatal outcome; malaria and HIV. Yale J Biol Med. 2007;80(3):95–103.

    PubMed  Google Scholar 

  33. Robert V, Macintyre K, Keating J, Trape J-F, Duchemin J-B, Warren M, et al. MALARIA TRANSMISSION IN URBAN SUB-SAHARAN AFRICA. Am J Trop Med Hygiene. 2003;68(2):169–76.

    Article  Google Scholar 

  34. Geissbühler Y, Kannady K, Chaki PP, Emidi B, Govella NJ, Mayagaya V, et al. Microbial larvicide application by a large-scale, community-based program reduces malaria infection prevalence in urban Dar Es Salaam, Tanzania. PLoS ONE. 2009;4(3):e5107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Okoko BJ, Ota MO, Yamuah LK, Idiong D, Mkpanam SN, Avieka A, et al. Influence of placental malaria infection on foetal outcome in the Gambia: twenty years after Ian Mcgregor. J Health Popul Nutr. 2002;20(1):4–11.

    PubMed  Google Scholar 

  36. Landis SH, Lokomba V, Ananth CV, Atibu J, Ryder RW, Hartmann KE, et al. Impact of maternal malaria and under-nutrition on intrauterine growth restriction: a prospective ultrasound study in Democratic Republic of Congo. Epidemiol Infect. 2009 Feb;137(2):294–304.

    Article  CAS  PubMed  Google Scholar 

  37. Accrombessi M, Yovo E, Fievet N, Cottrell G, Agbota G, Gartner A, et al. Effects of Malaria in the First Trimester of Pregnancy on Poor Maternal and Birth Outcomes in Benin. Clin Infect Dis. 2018.

  38. 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(11):6620–3 [cited 2019 Aug 16]. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC219546/.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Mayor A, Kumar U, Bardají A, Gupta P, Jiménez 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(11):1664–74.

    Article  CAS  PubMed  Google Scholar 

  40. Fried M, Duffy PE. Malaria during Pregnancy. Cold Spring Harb Perspect Med. 2017;7(6) [cited 2019 Aug 16]. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5453384/.

  41. 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(9405) [cited 2019 Aug 16]. Available from: https://ora.ox.ac.uk/objects/uuid:172544d7-2e73-4c9f-8d5d-213e91e5d04a.

  42. Umbers AJ, Aitken EH, Rogerson SJ. Malaria in pregnancy: small babies, big problem. Trends Parasitol. 2011 Apr;27(4):168–75.

    Article  PubMed  Google Scholar 

  43. Mocking M, Savitri AI, Uiterwaal CSPM, Amelia D, Antwi E, Baharuddin M, et al. Does body mass index early in pregnancy influence the risk of maternal anaemia? An observational study in Indonesian and Ghanaian women. BMC Public Health. 2018;18(1):873 [cited 2019 Aug 16]. Available from. https://doi.org/10.1186/s12889-018-5704-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Moya-Alvarez V, Cottrell G, Ouédraogo S, Accrombessi M, Massougbodgi A, Cot M. Does Iron Increase the Risk of Malaria in Pregnancy? Open Forum Infect Dis. 2015;2(2) [cited 2019 Aug 16]. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4567087/.

  45. Kabyemela ER, Fried M, Kurtis JD, Mutabingwa TK, Duffy PE. Decreased susceptibility to Plasmodium falciparum infection in pregnant women with iron deficiency. J Infect Dis. 2008 Jul 15;198(2):163–6.

    Article  PubMed  Google Scholar 

  46. Chandrasiri UP, Fowkes FJ, Richards JS, Langer C, Fan Y-M, Taylor SM, et al. The impact of lipid-based nutrient supplementation on anti-malarial antibodies in pregnant women in a randomized controlled trial. Malaria J. 2015;14(1):193 [cited 2019 Aug 16]. Available from. https://doi.org/10.1186/s12936-015-0707-2.

    Article  CAS  Google Scholar 

  47. Wyss K, Wångdahl A, Vesterlund M, Hammar U, Dashti S, Naucler P, et al. Obesity and diabetes as risk factors for severe Plasmodium falciparum Malaria: results from a Swedish Nationwide study. Clin Infect Dis. 2017 Sep 15;65(6):949–58.

    Article  PubMed  PubMed Central  Google Scholar 

  48. Amugsi DA, Dimbuene ZT, Mberu B, Muthuri S, Ezeh AC. Prevalence and time trends in overweight and obesity among urban women: an analysis of demographic and health surveys data from 24 African countries, 1991-2014. BMJ Open. 2017 Oct 27;7(10):e017344.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Walker-Abbey A, Djokam RRT, Eno A, Leke RFG, Titanji VPK, Fogako J, et al. Malaria in pregnant Cameroonian women: the effect of age and gravidity on submicroscopic and mixed-species infections and multiple parasite genotypes. Am J Trop Med Hyg. 2005;72(3):229–35.

    Article  PubMed  Google Scholar 

  50. Mubyazi G, Bloch P, Kamugisha M, Kitua A, Ijumba J. Intermittent preventive treatment of malaria during pregnancy: a qualitative study of knowledge, attitudes and practices of district health managers, antenatal care staff and pregnant women in Korogwe District, North-Eastern Tanzania. Malaria J. 2005;4(1):31 [cited 2019 Aug 16]. Available from. https://doi.org/10.1186/1475-2875-4-31.

    Article  Google Scholar 

  51. Hogarh JN, Agyekum TP, Bempah CK, Owusu-Ansah EDJ, Avicor SW, Awandare GA, et al. Environmental health risks and benefits of the use of mosquito coils as malaria prevention and control strategy. Malaria J. 2018;17(1):265 [cited 2019 Aug 16]. Available from: doi: https://doi.org/10.1186/s12936-018-2412-4.

  52. Choi L, Richardson M, Malone D, Pryce J. Insecticide space spraying for preventing malaria transmission. Cochrane Database Syst Rev. 2018;2018(11) [cited 2019 Aug 16]. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6516806/.

  53. Escamilla V, Alker A, Dandalo L, Juliano JJ, Miller WC, Kamthuza P, et al. Effects of community-level bed net coverage on malaria morbidity in Lilongwe, Malawi. Malaria J [Internet]. 2017;16(1):142 [cited 2019 Aug 16]. Available from: doi: https://doi.org/10.1186/s12936-017-1767-2.

  54. Atieli HE, Zhou G, Afrane Y, Lee M-C, Mwanzo I, Githeko AK, et al. Insecticide-treated net (ITN) ownership, usage, and malaria transmission in the highlands of western Kenya. Parasit Vectors. 2011 Jun 18;4:113.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Lawrance CE, Croft AM. Do Mosquito Coils Prevent Malaria? A Systematic Review of Trials. J Travel Med. 2004;11(2):92–96 [cited 2019 Aug 16]. Available from: https://academic.oup.com/jtm/article/11/2/92/1803449.

  56. Weinstein JR, Thompson LM, Díaz Artiga A, Bryan JP, Arriaga WE, Omer SB, et al. Determining gestational age and preterm birth in rural Guatemala: a comparison of methods. PLoS One. 2018;13(3):e0193666.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Katz J, Wu LA, Mullany LC, Coles CL, Lee ACC, Kozuki N, et al. Prevalence of small-for-gestational-age and its mortality risk varies by choice of birth-weight-for-gestation reference population. PLoS One. 2014;9(3):e92074.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

We would like to thank the study participants and the field teams, including study coordinators, doctors, nurses, midwives, supervisors, and the laboratory, administrative, and data staff at MUHAS and the clinic sites for their contributions to the study. Particular appreciation to the dedication offered by the field team including Vera Juma (deceased), Jeremy Kane, Juliana Mghamba, Fee Msafiri, Mwanaidi Said, and Kristina Lugangira. We also thank Paul Ng’walali and Amos Mwakigonja at MUHAS for their technical assistance in placental histopathology; Jaume Ordi and Atis Muehlenbachs for external review of the placental histopathology; and Michael Waisberg for malarial PCR on representative cases, as well as Manoj Duraisingh and Katy Shaw-Saliba at the Harvard TH Chan School of Public Health, and Ambroise Ahouidi at Le Dantec Hospital, Senegal, for their expertise in tissue homogenization, DNA extraction, and qPCR methods for diagnosing placental malaria. Finally, we thank DSMB members Jeffrey Griffiths, Mohamed Bakari, Henrik Friis, David Wypij, and Hassan Mshinda.

Disclaimer

“Research reported in this publication was supported by the Fogarty International Center, of the National Institutes of Health under Award Number D43 TW 007886. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.”

Funding

The current study was possible through the Global Infectious Diseases (GID) training program sponsored by the U.S. National Institute of Health (NIH) Fogarty International Center, grant number D43 TW 007886. The parent trial was supported by a grant from the National Institute of Child Health and Human Development (NICHD RO1 HD057941–01). Both funding institutions had no role in study design, data collection, analysis and interpretation, manuscript writing, and the choice of journal for submission of the manuscript for publication.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, W.W.F., A.V.K., E.H., A.M.D.; formal analysis, A.V.K.; investigation, A.V.K., A.M.D., F.M.M., A.I.A., S.A., F.O.O., H.M., D.H.H. and W.W.F.; writing—original draft preparation, A.V.K.; writing—review and editing, A.V.K., A.M.D., E.H., F.O.O., H.M., D.H.H., A.I.A., S.A. and W.W.F.; supervision, E.H. and W.W.F.; funding acquisition, W.W.F. All authors of this manuscript provided technical input and approved the final manuscript submitted to the journal.

Corresponding author

Correspondence to Aneth Vedastus Kalinjuma.

Ethics declarations

Ethics approval and consent to participate

The parent trial obtained ethical approval from the Harvard T.H. Chan School of Public Health Institutional Review Board (the IRB approval code is 18,573), the Muhimbili University of Health and Allied Sciences (MUHAS) Senate of Research and Publication Committee (the MUHAS approval reference is Ref.No.2015-06-26/AEC/Vol.IX/119), and Tanzania’s National Institute of Medical Research (NIMRI approval code is NIMR/HQ/R.8a/Vol. IX/966). All study participants were informed about the study and written consent was obtained from participants who accepted to participate in the study before data collection was initiated.

Consent for publication

Not applicable.

Competing interests

The authors declared that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. 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 in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kalinjuma, A.V., Darling, A.M., Mugusi, F.M. et al. Factors associated with sub-microscopic placental malaria and its association with adverse pregnancy outcomes among HIV-negative women in Dar es Salaam, Tanzania: a cohort study. BMC Infect Dis 20, 796 (2020). https://doi.org/10.1186/s12879-020-05521-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12879-020-05521-6

Keywords