INTRODUCTION
Recent estimates suggest that in malaria-endemic sub-Saharan Africa, each year approximately 25 million women become pregnant and are at increased risk of infection with Plasmodium falciparum, particularly in their first two pregnancies.1 This results in maternal anemia and reduced neonatal birth weight due to preterm delivery and intrauterine growth retardation (IUGR).2,3 The vast majority of these infections are low-grade, frequently sub-patent,4,5 and in most women are asymptomatic and therefore undetected and untreated.6
The effects of human immunodeficiency virus (HIV) on maternal health have been superimposed on that of malaria in the malaria-endemic regions. In sub-Saharan Africa, 55% of the HIV-infected adults are reproductive age women,7 accounting for 80% of the world’s HIV-infected women.8 Studies in pregnant women suggest that in several parts of Africa the prevalence of HIV now exceeds 25%.7,8 Without intervention, it is estimated that ~25–45% of the HIV-infected women will transmit infection to their children. In ~15–30%, this occurs in the intrauterine and intrapartum period and the remainder is due to breastfeeding.7
There are several points in the immune system where malaria and HIV can interact.9,10 Although there is now accumulating evidence for an effect of HIV-1 infection in adults and children on malaria,11–15 initial studies conducted among children and adults failed to show a consistent pattern of a biologic or clinical interaction, with the exception of studies in pregnant women.16–19 The first clear indication of an effect of HIV on malaria was reported by Steketee and others who found in 1987–1989 that HIV in multigravid women appeared to impair a pregnant woman’s ability to control malaria parasitemia, resulting in more frequent and higher density parasitemia than in HIV-uninfected pregnant women.20 Because of the high prevalence of HIV and malaria in sub-Saharan Africa, co-infections are common. This has important implications, since both HIV and malaria are among the leading causes of morbidity in pregnancy in Africa, and even modest effects of one infection on the other could lead to a substantial negative impact on the health of pregnant women and their newborns.12,18 Here we review the available information collected in the 15-year period since the study by Steketee and others,20 discuss the effects of HIV and malaria on each other, and suggest programmatic implications.
We searched PubMed for English publications using the keywords HIV, malaria, pregnancy, or pregnant, and we obtained further information from abstracts of scientific meetings, the internet, and personal communications with scientists.
EFFECT OF MATERNAL HIV ON MALARIA DURING PREGNANCY
Parasitemia.
Figure 1 summarizes the data from 11 of 13 studies (12 published20–31 and one unpublished [Rogerson SJ, unpublished data]) identified that determined the prevalence of maternal and placental malaria in HIV-infected and uninfected women in sub-Saharan Africa. These studies show a consistent increased risk of malaria parasitemia in HIV-infected women during pregnancy (risk ratio [RR] = 1.58), at the time of delivery (RR = 1.65), and in the placenta (RR = 1.66). The HIV-infected pregnant women also had higher malaria parasite densities (Table 1).
In the study by Steketee and others, the effect of HIV was principally observed in multigravidae, but there was no significant increased risk of malaria prevalence in primigravidae, although parasite densities were significantly higher.20 Now, with the availability of data from three more studies that present details by gravidity, it is clear that HIV-infected primigravidae are also affected (Table 2). Nevertheless, the HIV-associated risk of malaria is consistently greater in multigravidae (Table 2), suggesting that HIV affects the immune memory mechanism responsible for the parity-dependent acquisition of antimalarial immunity in pregnancy.32 Alternatively, multigravid women have longer sexual experience and may be more immunosuppressed because they have been infected with HIV longer than younger primigravid women. Thus, HIV alters the typical gravidity specific pattern of malaria risk by shifting the burden from primarily primigravidae and secundigravidae to all pregnant women, placing HIV-infected multigravidae in western Kenya at similar risk of malaria as HIV-uninfected women in their first and second pregnancies after adjusting for maternal age.27
Maternal morbidity.
Clinical malaria.
The HIV-infected pregnant women were more likely to develop clinical malaria, defined as documented fever or a history of fever in the presence of microscopically detected malaria parasitemia.27 They were also more likely to have used antimalarials during pregnancy other than those provided as part of intermittent preventive therapy.27,33
Maternal anemia.
Both HIV (especially with advanced immunosuppression)34,35 and malaria36 are known causes of maternal anemia. Several studies describe a negative effect of the combined impact of HIV and malaria on maternal hemoglobin (Hb) concentrations.22,31,37,38 Table 3 shows prevalence of anemia for HIV-infected, malaria-infected, and dually infected women in three studies that stratified results by gravidity from western Kenya and Malawi. Women with single infections with HIV or malaria were more at risk than uninfected women. However, dually infected women were at considerably greater risk of having any anemia (Hb < 11 g/dL) or moderate-to-severe anemia (Hb < 8 g/dL) than those with single infections. There was evidence for a synergistic interaction between the effect of malaria and HIV in each study, suggesting that the degree of malaria-associated anemia is worse in HIV-infected women, possibly reflecting the higher parasite densities and longer duration of malaria infection.
Birth outcome.
Seven studies were identified that examined the effect of dual infection with malaria and HIV on birth outcome.25,31,33,37,39,40 Although differences in study design limit direct comparisons between the studies, they generally show an increased risk of poor birth outcome in terms of low birth weight (LBW <2,500 grams), preterm birth, and IUGR with both HIV and malaria, with the greatest risk in women with dual infection. Table 4 summarizes the effect of malaria alone, HIV alone, and dual infections in two studies that provided data by gravidity group. Although the risk of LBW was consistently greatest in women with dual infection, it was less clear than with the effect on anemia if the effect of HIV and malaria on birth weight were additive or synergistic. Studies that reported on gestational age suggest that the effect on birth weight reflects a combined effect of shortened gestational age and IUGR.31,37,39,40
The impact of HIV on the burden of maternal malaria.
The magnitude of the impact of the HIV epidemic on maternal malaria in sub-Saharan Africa can be expressed as the proportional increase in malaria during pregnancy in a population that is due to HIV (the population attributable fraction [PAF]). The PAF is a function of the HIV-associated increased risk of malaria during pregnancy and increases with increasing prevalence of HIV-1 in a population (Figure 2). The mean HIV prevalence among pregnant women visiting antenatal clinics in 39 sub-Saharan countries was 8.6% during 1985–2000.41 The PAF for malaria during pregnancy associated with this HIV prevalence is 4.8% (95% confidence interval [CI] = 3.9–5.8). In settings where HIV prevalence in pregnant women is 25% or 40%, 12.7% (95% CI = 10.5–15.1) and 18.8% (95% CI = 15.8–22.1) of the maternal malaria infections will be attributable to HIV-1, respectively (Figure 2).
The PAF can also be used to estimate the annual excess number of malaria cases during pregnancy that is due to HIV using a three-step process. First, the total number of pregnant women exposed each year to malaria in areas with stable transmission in sub-Saharan is estimated to be 24.6 million (in 2002).1 Second, previous estimates from 20 studies from 8 countries in sub-Saharan Africa conducted between 1985 and 2000 involving 24,451 pregnant women showed the weighted prevalence of maternal malaria during pregnancy to be 42.8% HIV.42 Combining these figures suggests that annually at least 10.5 million women have malaria in the second or third trimester. Third, with an average HIV prevalence of 8.6%41 (PAF = 4.8%), it can be estimated that the HIV-1 epidemic results in an additional 505,382 women per year who have malaria during pregnancy. This number would increase to 1.34 million pregnancies per year if the average HIV seroprevalence among pregnant women reached 25% (PAF = 12.7%). This reflects the minimum effect of HIV on malaria in pregnancy because the 42.8% estimate is based on single point prevalence data, and does not take into account women who may have had malaria before or after the time of the point estimate.
Why are HIV-infected pregnant women more susceptible to malaria?
The increased susceptibility of pregnant women to malaria is not well understood, but is presumed to be related to modifications in systemic and placental immunologic parameters. There have been a limited number of studies of the cellular and humoral immune responses to malaria that may allow further understanding of the impact of HIV on these malaria and pregnancy specific immune responses.
Cellular immune responses.
Intervillous blood mononuclear cells (IVBMCs) produce interferon-γ (IFN-γ), which has been implicated in protection against placental malaria.43 In HIV-infected pregnant women with P. falciparum malaria, this production is reduced in response to malarial antigen stimulation44 due to severe impairment of the interleukin-12 (IL-12), but not IL-18-mediated IFN-γ pathway.45 This impairment was more pronounced with low CD4 counts,44,45 and highlights the shift in the cytokine responses as infection with HIV progresses. Not all the cytokine responses were affected, indicating that there is no generalized suppression of immune response in these pregnant women.44–46 Given that P. falciparum is an intracellular parasite whose clearance requires an active cellular immune mechanism involving macrophages, it is understandable that impairment of the IL-12-mediated IFN-γ pathway due to HIV is a potential cause for increased malaria.
Humoral immune responses.
Two recent studies conducted in Kenya and Malawi also show partial impairment of the humoral immune response to malaria in HIV-infected pregnant women.32,47 Although the anti-malarial antibody responses were unaltered for most of the antigen or epitopes tested,32,47 responses were impaired to the pre-erythrocytic stage circumsporozoite protein (NANP-5),47 apical merozoite antigen 1 (AMA-1), and variant surface antigens (VSA) expressed on infected erythrocytes that bind to chondroitin sulfate A,32 a key receptor for placental sequestration.48 This impairment is greatest in women with more advanced HIV disease, and occurs across all gravidities and in women with and without current malaria infection.32 What is not presently clear is whether T cell, B cell, or macrophage functional impairments, or a combination of all three, best explain the predisposition to malaria described in infections with HIV.
EFFECT OF MATERNAL MALARIA ON HIV
HIV-1 viral load.
The role of immune-activation by coinfecting pathogens has long been postulated as a factor influencing the severity and rate of disease progression in HIV-infected individuals in developing countries.18,49 Two studies in non-pregnant adults with P. falciparum malaria showed that plasma HIV-1 RNA concentrations are transiently increased and this higher HIV viral load is in part reversible with successful antimalarial therapy.50,51 More recently, three of four studies confirmed that this also occurs in pregnant women (Table 5).29,52–54 The magnitude of the effect (approximately two-fold increase in viral load) appears smaller than the seven-fold increase seen in adults with symptomatic malaria,51 perhaps because few pregnant women are symptomatic. One study also assessed the relationship between placental malaria and placental viral load, and found a similar two-fold increase in placental HIV-1 RNA concentrations, with the greatest increase in women with highest placental parasite densities.53 Importantly, these observations were independent of the degree of immunosuppression as assessed by CD4 cell counts and can thus not be explained by an increased risk of malaria in subjects with more advanced immunosuppression and potentially greater HIV-1 viral load.53
As in non-pregnant adults, P. falciparum can increase HIV-1 replication by activating both the lymphocyte and macrophage cell pool55,56 through up-regulating pro-inflammatory cytokine production, particularly TNF-α. Similarly, rodent P. chabaudi malaria induces virus expression in HIV-infected transgenic mice.57 In addition, parasite sequestration in the placenta results in inflammatory increases in IVBMCs, predominantly monocytes and macrophages, the degree of which is correlated with the density of placental parasitemia, resulting in high levels of local proinflammatory cytokine responses such as TNF-α.46,58–63 Placental malaria is also associated with increased expression on placental macrophages and fetal Hofbauer cells of CC chemokine receptor 5 (CCR5),64,65 a major fusion co-receptor for HIV-1 cell-entry.64,66 The HIV-1 viruses that are transmitted from mother to child are predominantly CCR5 trophic.67 Thus, there are several pathways that combined could result in increased systemic and/or placental replication of HIV-1 and viral reservoir in placentas of dually infected women, and thus increased mother-to-child transmission (MTCT) of HIV-1.
Mother-to-child transmission of HIV-1.
The question of whether malaria during pregnancy enhances the risk of MTCT is pivotal for sub-Saharan Africa, where few women take anti-retroviral drugs and uptake of MTCT preventive services has been low.7 Although maternal HIV-1 viral load is the single most important risk factor for MTCT of HIV-1,68–70 the available evidence suggests that placental malaria may not necessarily increase the risk of MTCT (Table 6).28,29,54,71 Among three recently published studies that provided sufficient detail to allow comparison, significant heterogeneity (P = 0.01) was found in the risk of MTCT associated with placental malaria, ranging from an increased risk in Uganda (RR = 2.89, 95% CI = 1.12–7.52),28 to no effect in Mombasa, Kenya,29 and a significant protective effect in Kisumu in western Kenya (RR = 0.44, 95% CI = 0.27–0.72, P < 0.001).54
The apparent discrepancies may reflect a complex relationship between maternal immune responses to malaria that on the one hand may stimulate HIV viral replication in the placenta thereby increasing the local viral load, and on the other hand may potentially control the severity of malarial infection and HIV replication. The balance of these can tip either in the direction of enhanced risk of MTCT or a protective effect, depending on the degree of immune suppression, and on the severity of the malaria and thus the degree of placental monocyte infiltrates and proinflammatory cytokine and chemokine responses (Figure 3).
Although the precise mechanism needs to be established, it has been suggested that the family of CC-chemokines (β-chemokines) such as macrophage inflammatory protein (MIP)-1α, MIP-1β; and “regulated-upon activation normal T-cell expressed and secreted” (RANTES) protein may play a potential protective role in this relationship.54,72 The CC-chemokines have a broad spectrum of action, including lymphocyte and monocyte homing and migration. These three CC-chemokines are also ligands for the co-receptor CCR5 and competitively inhibit HIV-1 entry into macrophages. Individuals with high concentrations of these three chemokines remain uninfected despite repeated exposure to HIV.73 Furthermore, infants born to HIV-infected women who have high plasma concentrations of CC-chemokines and strong anti-HIV cytotoxic/suppressor T cell responses at birth have a reduced risk of HIV-1 infection.74 Although some differences exist between different studies, it has been generally found that the concentrations of one or more of these chemokines are significantly elevated in women with placental malaria, irrespective of their HIV status.63,72,75 Preliminary results from the MTCT study in western Kenya that found a protective effect of placental malaria on MTCT suggest that concentrations of MIP-1α and MIP-1β, but not RANTES, were lower in malaria-infected placentas from women who transmitted the virus to their infants than in malaria-infected placentas from non-transmitting women (Chaisavaneeyakorn S, Udhayakumar V, unpublished data). Based on these findings, one could speculate that these chemokines may play some protective role in MTCT of HIV-1. The half-lives of the CC-chemokines, however, are short and a causal role remains to be established. Alternatively, it is also possible that elevated levels of chemokines may be simply an immune marker for protection.
The immunocompetence of the host may be another important determinant in the MTCT of HIV outcome. The majority of women in the same study in western Kenya had CD4 counts > 400/μL and peripheral HIV-1 RNA concentrations were low (Table 5).54 No details were given of the viral load or CD4+ counts from the Ugandan study that found an increased risk of MTCT.28 Although individual studies adjusted for viral load and/or CD4 counts, large differences between the studies in immunocompetence of HIV-infected women cannot be excluded as an explanation for the differences in the effect of placental malaria on MTCT (Table 6).
Although further studies are needed to provide an explanation for the apparent heterogeneous effects of placental malaria on MTCT of HIV-1, we hypothesize that immune factors play an important balancing act in this relationship (Figure 3). Other potential explanations for the apparent difference between the three studies may include variation in the sensitivity of the techniques used for the determination of placental malaria (Table 6).
Future studies should also include assessment of the role of severity and the timing of malaria (acute versus chronic placental infection), viral phenotypes76 and potential host genetic factors that could provide a mechanism of resistance against transmission of HIV-1.77 Furthermore, children who develop T cell responses to HIV peptides in utero are less likely to acquire HIV-1,78 but no studies have systematically examined the effect of maternal or fetal malaria on the refractoriness or susceptibility of fetal leukocytes to HIV-1.
IMPLICATIONS OF MATERNAL CO-INFECTION WITH HIV AND MALARIA FOR INFANT HEALTH
Transfer of maternal antibodies to the fetus.
The maternofetal transfer of antibodies forms an important protection against infectious diseases in the first months of life. Maternal HIV-1 infection can cause placental pathologic changes that might interfere with transfer of antibodies to some malarial antigens, including against the pre-erythrocytic stage circumsporozoite protein NANP-5, the synthetic blood stage peptides merozoite surface protein 3 (MSP-3), AMA-1, and VSA.32,47 Immunoglobulin transfer across the placenta is an active process, with IgG being primarily taken up by FcRN receptors on the syncytiotrophoblast.79 How HIV-1 interferes with this process is presently unknown and it is also unclear whether this leads to an increased susceptibility to malaria in infants.
Malaria and anemia in infants.
Several published80–85 and unpublished studies (van Eijk AM and others, unpublished data) have shown that newborns of mothers who had malaria during pregnancy are at an increased risk of malaria and anemia during infancy, particularly in the first six months of life. Early immune priming/sensitization in utero and the development of immunologic tolerance could possibly explain the susceptibility of infants born the mothers with malaria.84,86–88 Very few studies have addressed the effect of maternal HIV-1 on cord blood parasitemia and infant malaria. The available evidence does not suggest that maternal HIV-1 has a consistent effect on congenital malaria20,89,90 or affects the relationship between maternal and infant malaria (Table 7)(van Eijk AM and others, unpublished data).91,92
Mother-to-child transmission of HIV-1 places their children at risk of severe anemia.93 Maternal HIV also has an indirect, albeit small, effect on infant anemia. The HIV-exposed but HIV-uninfected children born to Kenyan mothers with advanced HIV immunosuppression were at increased risk of severe anemia compared with HIV-uninfected children from HIV-uninfected mothers (RR = 1.14, 95% CI = 1.03–1.26),80 although in Malawi, no such increased risk of anemia was seen in infants of HIV-infected mothers.85
Infant mortality.
Previous data from Malawi showed that dual exposure to both placental malaria and maternal HIV-1 increased the risk of post-neonatal mortality 3–8 fold (depending on the birth weight) compared with infants born to mothers with HIV alone, suggesting an increased risk of MTCT or increased rate of HIV-1 disease progression in infants.94 This was not confirmed by two subsequent studies in southern Malawi and western Kenya where lower mortality rates were observed among infants born to HIV-infected women with placental malaria than those without placental malaria (Table 8).89,95 The protective effect in western Kenya is consistent with the reduced MTCT found in women with dual infection compared with women without placental malaria.54
TREATMENT ISSUES AND PROGRAMMATIC IMPLICATIONS
Response to treatment, chemoprophylaxis, and intermittent preventive treatment (IPT).
The World Health Organization (WHO) currently recommends a three-pronged approach for malaria control during pregnancy in sub-Saharan Africa: intermittent preventive treatment (IPT), insecticide-treated bed nets, and effective case management of malaria illness.96
The IPT involves the administration of full treatment doses of an effective antimalarial drug at predefined gestational intervals; currently at least two doses of sulfadoxine-pyrimethamine are recommended after quickening, given at routine antenatal clinic (ANC) visits.96–100 One clinical trial conducted in western Kenya demonstrated that HIV-infected primigravidae and secundigravidae had a suboptimal response to IPT, requiring at least three doses of sulfadoxine-pyrimethamine to achieve similar reductions in placental parasitemia as HIV-uninfected women who received two doses.99 Although both HIV-infected and uninfected women had an excellent parasitologic treatment response by day 14,99 HIV-infected women subsequently had a higher prevalence of placental parasitemia that was likely due to late recrudescence of parasitemia and reinfections in women with compromised immunity. Verhoeff and others noted a highly significant reduction in the frequency of LBW in both HIV-infected and HIV-uninfected Malawian women receiving the recommended two doses of IPT with sulfadoxine-pyrimethamine under program conditions (compared with women receiving just one dose), although HIV-infected women were twice as likely to give birth to LBW babies.90 This reduction in the frequency of LBW occurred despite high rates of placental parasitemia in women of either HIV serostatus.90 Further studies are in progress in Malawi and Zambia to address the issue of IPT frequency in HIV-infected women in more detail.
While the WHO recommends that women receive at least two doses of IPT, if doses are linked to the most recent WHO-recommended schedule of four ANC visits with three after quickening,96,101,102 this would potentially deliver three doses to each woman that, based on the limited data available, should be efficacious in areas with high HIV prevalence. Sixty-eight percent of women in sub-Saharan Africa make at least one ANC visit. Of those who attend an ANC, 95% make at least two visits and nearly 60% make at least four visits.103
In a clinical study comparing different chloroquine prophylaxis or IPT regimens in Malawi, HIV-infected women had higher rates of persistent and breakthrough parasitemia, and peripheral and placental parasitemia at delivery, indicating a poorer response to both prophylaxis and treatment.20
There are currently no published data on whether HIV-infected pregnant women exhibit an impaired response to the treatment of clinical malaria. Scarce data available from non-pregnant populations have shown variable results.104–108
Prevention of opportunistic infections and malaria.
UNAIDS recommends opportunistic infection (OI) prophylaxis with cotrimoxazole for certain groups of HIV-infected persons (or potentially infected infants), including all pregnant women after the first trimester.109 While WHO does not recommend a specific drug for IPT, the only available data are for sulfadoxine-pyrimethamine97–100 and chloroquine.40,110,111 Sulfadoxine-pyrimethamine is increasingly being used because chloroquine efficacy is limited by drug resistance.96,112,113 Thus, OI prophylaxis with cotrimoxazole and malaria prevention with sulfadoxine-pyrimethamine involve two similar sulfa drugs for HIV-infected pregnant women, which is inadvisable in view of the potential risk of increased adverse drug reactions (ADRs). Because sulfadoxine-pyrimethamine is not as effective against bacterial pathogens,114,115 cotrimoxazole might be used to prevent both bacterial infections and malaria. Cotrimoxazole has been shown to effectively treat malaria in children.116,117 Cotrimoxazole prophylaxis in HIV-infected persons has been shown to decrease malaria morbidity.118 Data are urgently needed on the efficacy of cotrimoxazole prophylaxis on reducing rates of placental malaria and anemia in HIV-infected pregnant women.
In addition, there is in vitro evidence for cross-resistance between trimethoprim and pyrimethamine.119 Widespread use of cotrimoxazole in prophylaxis programs for HIV-infected persons could potentially provide substantial drug pressure that could accelerate the development of P. falciparum resistance to sulfadoxine-pyrimethamine. Conversely, expanding use of sulfadoxine-pyrimethamine might further the development of resistance to cotrimoxazole by other pathogens, such as Streptococcus pneumoniae.114 The effectiveness of these drugs should be carefully monitored in HIV-1 and malaria control programs.
Interactions and commonalities between antimalarial and antiretroviral drugs.
As programs are put into place for the administration of antiretroviral therapy, potential interactions between antiretroviral drugs and antimalarial drugs must be considered. The use of quinidine (and thus, possibly quinine) is contraindicated because of potential cardiotoxicity among persons receiving a number of antiretroviral agents, including nelfinavir and ritonavir,120,121 and should be used only with caution and careful monitoring with amprenavir, delaviridine, and lopinavir/ritonavir in combination.122–124 The co-administration of cotrimoxazole and lamivudine results in decreased clearance of lamivudine;125 there are no reported studies on the interaction of sulfadoxine-pyrimethamine with lamivudine. There is also recent evidence that protease inhibitors may impair CD-36 mediated cytoadherence and non-opsonic phagocytosis of parasitized erythrocytes, which may contribute to altered malaria disease outcomes in co-infected patients.126–128
Chloroquine and hydroxy-chloroquine are antimalarial drugs that have been shown to have antiretroviral activity in vitro129–132 and in vivo,133,134 thought to be due to inhibition of post-translational modification of gp120.135–137 They have been proposed as potentially useful components of antiretroviral combination therapy in resource-poor settings,138,139 including for MTCT of HIV-1 because of their effect on reducing production of infectious virions129 and because they accumulate in breast milk cells.140 However, even if these drugs were used in antiretroviral programs (at much higher doses, i.e., 800 mg of hydroxy-chloroquine/day, than are used for malaria),133,134 their usefulness in preventing the adverse effects of malaria in HIV-infected women would likely be limited because of high levels of chloroquine resistance in sub-Saharan Africa.
Anemia prevention and HIV.
Iron deficiency, malaria, and HIV are among the major causes of anemia in pregnant women in sub-Saharan Africa. However, anemia prevention and treatment may be complicated by HIV. Friis and others have reported that relatively asymptomatic HIV-infected pregnant women have slightly reduced iron stores,141 while increased iron stores have been reported in those with more advanced disease.142 However, in a recent trial in Kenyan adults, iron supplementation was associated with reduced Hb, which was thought to be due to HIV progression.143 While iron is important for immune function, it is also required by enzymes involved in HIV replication and, because of its pro-oxidant properties, may even stimulate HIV replication.144,145 Epidemiologic studies have shown that supplemental iron,146,147 inadequate chelation of iron overload,148 and the size of iron stores149 are associated with the progression of HIV.141 Studies are urgently needed to clarify this further because a detrimental effect of iron supplementation on HIV infection would have major implications for antenatal care policies.143 Because pregnant women are one of the highest risk groups for anemia, prevention and treatment, especially of severe anemia, is paramount during pregnancy to minimize the potential need for transfusion of blood that may not be adequately screened for HIV.
Adverse drug reactions and pharmacovigilance.
Reports among non-pregnant persons indicate that the rate of ADRs to sulfonamides may be higher among HIV-infected than HIV-uninfected persons.150–152 Parise and others reported that 3.2% and 0.4% of HIV-infected and HIV- negative women in Kisumu, respectively, experienced ADRs after the first treatment dose (P = 0.08).106 Two (2%) of 94 HIV-infected and none of 230 HIV-uninfected women had sulfadoxine-pyrimethamine withheld because of ADRs.99
During one year of data collection in a district-wide surveillance study monitoring severe cutaneous reactions associated with sulfonamide use in pregnant and non-pregnant persons in Malawi (where HIV seroprevalence is approximately 25% among pregnant women in urban areas),153 rates of severe cutaneous adverse reactions were consistently greater among HIV-infected than among HIV-uninfected persons: 5.1/100,000 versus 1.4/100,000 (sulfadoxine-pyrimethamine only), 14.1/100,000 versus 1.5/100,000 (cotrimoxazole only), and 42.2/100,000 versus 0.4/100,000 (sulfadoxine-pyrimethamine plus cotrimoxazole). Estimated rates among pregnant women were 4.2/100,000 drug exposures; there were insufficient numbers to generate stable estimates comparing rates between HIV-infected and HIV-uninfected pregnant women (Gimnig J, unpublished data). The etiology of the increased rate of ADRs to sulfonamides among HIV-infected compared with HIV-uninfected persons is unclear and is likely multifactorial. Sulfonamide-induced ADRs have been linked to a slow acetylator phenotype154 and genotype.155 During acute periods of illness, patients with acquired immunodeficiency syndrome often express glutathione deficiency.156 The combination of these two acquired deficiencies could potentially lead to high levels of hydroxylamine metabolites, which have been associated with a higher rate of cutaneous ADRs.151,154,155 It has also been suggested that in some cases, the use of high doses of drugs may be a factor, given that there is some evidence for an abnormal accumulation of drug in toxic epidermal necrolysis.151,157
Programs for both non-pregnant and pregnant women must incorporate surveillance systems to monitor for severe ADRs. The apparent dramatic increase in the risk of severe cutaneous reactions among HIV-infected women receiving both cotrimoxazole and sulfadoxine-pyrimethamine also points to the need for both a greater understanding of the efficacy of cotrimoxazole alone in preventing malaria during pregnancy, and a greater integration of HIV prevention and ANC services.
CONCLUSIONS
Although much has been learned in the 15 years since the initial identification of interactions between malaria and HIV in pregnant women, many issues still require further information to improve our understanding. It is now clear that among pregnant women, the majority of co-infections with HIV and malaria occur in sub-Saharan Africa. The HIV-infected pregnant women experience more malaria and higher density malaria parasitemia, and dually infected pregnant women have more febrile illnesses, more anemia, and more adverse birth outcomes (LBW, prematurity, and IUGR) than women with single infections with malaria or HIV. There are caveats about each of these factors and although there is evidence for synergistic interactions exacerbating malarial anemia, present evidence does not allow us to exclude a simple additive effect on birth outcome.
Malaria also increases HIV viral replication and viral load and with inadequate antimalarial treatment this may worsen HIV disease progression, although the latter remains to be determined. The role of malaria as a contributing factor in MTCT of HIV is particularly important. Unfortunately, there are limited studies addressing this and existing findings are inconsistent. The sum of the current data suggests that malaria contributes little to MTCT, but more studies are needed, particularly in light of increasing efforts to prevent both MTCT and maternal malaria.
Similarly, there is limited information on the effects of HIV or malaria on the treatment and prevention interventions directed at the other infection/disease. As described here, there are drug-drug interactions that could be problematic and there is concern that each infection might alter the benefits achieved with drugs for treatment. Again, given the global push to extend the use of antiretroviral drugs and the advent of new and combination antimalarial treatments, further investigation into such interactions will be increasingly important.
While there has been some progress made in examining the human immune response to both HIV and malaria during pregnancy, there remain substantial gaps in our knowledge about how pregnant women respond to these two infections at a time when the immune responses required to maintain a healthy pregnancy are themselves complex. This is an area in which clearly reproductive immunologists and infectious disease immunologists could both make important contributions.
Finally, notwithstanding the present gaps in our knowledge, there is a clear need to strengthen the deployment of existing malaria and HIV prevention and intervention measures. For pregnant women who are HIV-infected, malaria prevention (with insecticide-treated nets and IPT) should be a priority to reduce the frequency of malaria associated maternal anemia and LBW. For women found to be dually infected, appropriate management of both infections is a priority both for the woman and for her developing fetus. Given that a high proportion of pregnant women do attend antenatal clinics for care, and that there are new resources available globally to tackle HIV and malaria,158 the global health community has a clear responsibility and opportunity to limit the scourge of these two diseases in this vulnerable population.
Parasite densities in human immunodeficiency virus (HIV)–infected and uninfected pregnant women*
Geometric mean parasite densities at first ANC visit | |||
---|---|---|---|
Study | HIV infected (N) | HIV uninfected (N) | P |
* ANC = antenatal clinic; AG = all gravidae; PG = primigravidae; MG = multigravidae. | |||
† Details of geometric mean parasite densities for multigravidae were not available (NA) from the published manuscript, but reported as “significantly higher in HIV-infected women than in seronegative women.”20 | |||
‡ Excluding women reporting recent antimalarial use. | |||
Steketee and others, 1996 Malawi20 | PG: 4,390 (45) | PG: 1,375 (678) | <0.001 |
MG: NA (104)† | MG: NA (1,867)† | <0.05† | |
AG: 1558 (149) | AG: 670 (2,545) | <0.001 | |
Verhoeff and others, 1999, Malawi33 and unpublished data | PG: 1,634 (18)‡ | PG: 1,048 (46)‡ | 0.17 |
MG: 822 (24)‡ | MG: 729 (31)‡ | 0.43 | |
AG: 1,143 (46) | AG: 908 (84) | 0.23 | |
van Eijk and others, 2003, Kenya27 | PG: 1,463 (81) | PG: 684 (201) | <0.001 |
MG: 757 (85) | MG: 323 (100) | <0.001 | |
AG: 1,031 (166) | AG: 499 (301) | <0.001 |
Prevalence, relative risk, and proportion of maternal and placental malaria attributable to human immunodeficiency virus (HIV) in primi,-, secundi,- and grand-multigravidae*
Prevalence (N) | ||||
---|---|---|---|---|
HIV-infected | HIV-uninfected | RR (95% CI) | AF (95% CI) | |
* Grand multigravidae is defined as ≥ third pregnancy. Published27,30,33 and unpublished (Rogerson SJ and others, unpublished data) data were used from four different studies conducted in Malawi and western Kenya that presented results by gravidity. Maternal malaria at delivery represents malaria detected using peripheral smears collected from the mother at the time of delivery or within 24 hours thereafter. Placental malaria was determined using malaria microscopy of the maternal side of the placenta20,27,33 or placental histopathology (Rogerson SJ and others, unpublished data). The attributable fraction (AF) was calculated using the weighted relative risk (RR) of malaria in HIV-infected women and uninfected women [AF = 100 × (RR − 1/RR)]. The RR and 95% confidence interval (CI) were obtained by the Mantel-Haenszel method for combining trials in fixed effect models.159 Proportions in the second and third column represent the weighted prevalence and numbers in parentheses represent the number of pregnant women. The effects of HIV on the risk of malaria differed significantly between the three gravidity groups (tests for heterogeneity between gravidity groups: maternal malaria at delivery: P = 0.03, placental malaria: P = 0.004). | ||||
Maternal malaria | ||||
Primigravidae | 34.3 (359) | 25.2 (1,590) | 1.36 (1.14–1.62) | 26.5 (12.3–38.3) |
Secundigravidae | 30.6 (241) | 16.0 (778) | 1.91 (1.47–2.49) | 47.6 (32.0–59.8) |
Grandmultigravidae | 21.2 (383) | 10.2 (1,691) | 2.09 (1.63–2.69) | 52.2 (38.7–62.8) |
Placental malaria | ||||
Primigravidae | 34.5 (359) | 27.2 (1,589) | 1.27 (1.06–1.51) | 21.3 (5.7–33.8) |
Secundigravidae | 30.4 (241) | 17.9 (774) | 1.70 (1.30–2.23) | 41.2 (23.1–55.2) |
Grandmultigravidae | 24.6 (382) | 10.3 (1,686) | 2.39 (1.87–3.07) | 58.2 (46.5–67.4) |
Effect of malaria and human immunodeficiency virus (HIV) on anemia in pregnant women
van Eijk and others38 (Hemoglobin < 8 g/dL) third trimester* | Ayisi and others37 (Hemoglobin < 8 g/dL) delivery† | Rogerson and others, unpublished data(Hemoglobin < 11 g/dL) delivery‡§ | ||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|
Primi gravidae | Multi gravidae§ | Primi gravidae | Multi gravidae§ | Primi gravidae§ | Multi gravidae¶ | |||||||
N | % | N | % | N | % | N | % | N | % | N | % | |
* Hemoglobin and maternal malaria assessed at ≥ 32 weeks gestation as part of antenatal care. | ||||||||||||
† Malaria defined as positive placental or peripheral blood smear (microscopy). Hemoglobin taken during or within 24 hours of delivery. Constitutes a sub-sample of the women seen in the third trimester by van Eijk and others.38 | ||||||||||||
‡ Malaria defined as positive placental blood smear (microscopy). Hemoglobin taken during or within 24 hours of delivery. | ||||||||||||
§ Statistically significant (P < 0.05) interaction between the effect of malaria and HIV on anemia. | ||||||||||||
¶ P = 0.09 for the interaction between the effect of malaria and HIV on anemia. | ||||||||||||
No infection | 1,233 | 11.8 | 1,889 | 13.6 | 607 | 10.7 | 708 | 12.1 | 89 | 20.3 | 175 | 13.1 |
HIV alone | 286 | 18.9 | 601 | 21.8 | 147 | 19.0 | 229 | 14.0 | 51 | 34.2 | 114 | 26.3 |
Malaria alone | 368 | 18.2 | 273 | 14.3 | 206 | 13.6 | 119 | 14.3 | 80 | 35.6 | 69 | 23.2 |
Dual infection | 172 | 23.8 | 194 | 34.0 | 81 | 23.5 | 76 | 32.9 | 19 | 72.7 | 28 | 35.7 |
Effects of malaria and human immunodeficiency syndrome (HIV) on the frequency of low birth weight*
Ayisi and others37† | Rogerson and others, unpublished data‡ | |||||||
---|---|---|---|---|---|---|---|---|
Primi gravidae | Multi gravidae | Primi gravidae | Multi gravidae§ | |||||
N | % | N | % | N | % | N | % | |
* Low birth weight (LBW) was defined as < 2,500 grams in singleton live births. | ||||||||
† Positive placental or peripheral blood smear (microscopy). | ||||||||
‡ Positive placental blood smear (microscopy). | ||||||||
§ Statistically significant (P < 0.05) interaction between the effect of malaria and HIV on LBW. | ||||||||
No infection | 676 | 4.1 | 809 | 2.7 | 30 | 21.0 | 79 | 5.1 |
HIV alone | 160 | 7.5 | 256 | 3.9 | 38 | 23.7 | 176 | 8.0 |
Malaria alone | 226 | 9.3 | 129 | 2.3 | 59 | 25.4 | 54 | 13.0 |
Dual infection | 91 | 14.3 | 84 | 4.8 | 14 | 35.7 | 24 | 16.7 |
Effect of maternal and placental malaria on human immunodeficiency virus 1 (HIV-1) viral load*
Peripheral viral load | Placental viral load | ||||
---|---|---|---|---|---|
Study | Design | Peripheral malaria at delivery | Placental malaria | Placental malaria | |
* NA = not assessed/available. Malaria was diagnosed by microscopy unless indicated otherwise. The average increase in peripheral HIV-1 RNA concentration was 1.9-fold for peripheral malaria and 2.08-fold for placental malaria (weighted by sample size). | |||||
† Values are the median (interquartile range). | |||||
‡ Ratio of median or geometric means. | |||||
§ Values are the geometric mean (95% confidence interval). | |||||
¶ Placental histology. | |||||
# Comparison of log10 viral load. | |||||
Kapiga and others, 200252 | Cohort | Malaria | 63,299 (13,461, 105,459)† n = 28 | NA | NA |
No malaria | 17,291 (3,336, 64,719)† n = 123 | NA | NA | ||
Difference‡ | Ratio 3.66, P = 0.01 | ||||
Mwapasa and others, 200453 | Cross-sectional | Malaria | 42,727 (25,745–70,892)§ n = 56 | 62,359 (41,706–93,218)§ n = 69¶ | 11,733 (7,091–19,413) n = 66§¶ |
No malaria | 29,833 (23,421–38,001)§ n = 258 | 24,814 (18,785–32,772)§ n = 200¶ | 4,919 (3,533–6,849) n = 196§¶ | ||
Difference‡ | Ratio 1.43, P = 0.21# | Ratio 2.51, P = 0.0007# | Ratio 2.39, P = 0.008# | ||
Ayisi and others, 200454 | Cross-sectional | Malaria | 2,979 (1,795–4,942)§ n = 85 | 2,399 (1,550–2,399)§ n = 107 | NA |
No malaria | 1,725 (1,393–2,137)§ n = 359 | 1,774 (1,426–2,208)§ n = 348 | NA | ||
Difference‡ | Ratio 1.73, P = 0.03# | Ratio 1.35, P = 0.20# | |||
Inion and others 200329 | Cross-sectional | Malaria¶ | NA | 13,029§ n = 34 | NA |
No malaria | NA | 12,419§ n = 338 | NA | ||
Difference‡ | Ratio 1.05, P = 0.921# |
Placental malaria and mother-to-child transmission of human immunodeficiency virus type 1 (HIV-1)*
Study | Method used | Results | |
---|---|---|---|
* The weighted summary relative risk (RR) (95% confidence intervals) for mother-to-child transmission (MTCT) of the three studies that provided numerator and denominator details was 0.79 (0.55–1.15) and was obtained with the Mantel-Haenzel method for combining trials in fixed effect models159 (HIV-infected women with [n = 170] and without [n = 759] placental malaria.28, 29 The effects of placental malaria on MTCT of HIV-1 differed significantly between the three recent studies (tests for heterogeneity: P = 0.01). | |||
† Hematoxylin and eosin stain was used to meet the original goal of the study (determining the impact of sexually transmitted diseases on HIV transmission). | |||
St Louis and others, 199371 | Placental histology | No data given | “No association between placental malaria and perinatal HIV-1 transmission” |
Brahmbhatt and others, 200328 | Placental histology(hemotoxylin and eosin stain)† | Placental malaria | 6/15 (40%) |
No malaria | 12/78 (15.4%) | ||
Crude RR | 2.60 (1.16–5.84), P = 0.03 | ||
Adjusted RR | 2.89 (1.12–7.52) | ||
Inion and others, 200329 | Placental histology | Placental malaria | 4/27 (14.8%) |
No malaria | 60/297 (20.2%) | ||
Crude RR | 0.73 (0.29–1.86), P = 0.501 | ||
Ayisi and others, 200454 | Microscopy | Malaria | 18/128 (14.1%) |
No malaria | 84/384 (21.9%) | ||
Crude RR | 0.64 (0.40–1.03), P = 0.055 | ||
Adjusted RR | 0.44 (0.27–0.72) P < 0.001 |
Association between placental malaria or maternal human immunodeficiency virus (HIV) infection and infant parasitemia during the first three months of life
Univariate odds ratio(95% confidence interval) | |
---|---|
* Placental malaria status was not known in this study. | |
† No evidence for interaction in multivariate model (P interaction term = 0.3). | |
Taha and others, 1994*92 | |
Maternal HIV infection | 0.78 (0.43–1.40) |
Slutsker and others, 199691 | |
Maternal HIV infection | 0.47 (0.21–1.02) |
Placental malaria | 0.91 (0.54–1.53) |
van Eijk and others, unpublished†97 | |
Maternal HIV infection | 0.94 (0.67–1.32) |
Placental malaria | 1.48 (1.08–2.01) |
Maternal human immunodeficiency status (HIV) status and placental malaria as risk factors for post-neonatal infant mortality in Malawi and Kenya*
HIV status | Placental malaria | RR (95% CI) Verhoeff and others, 2000†95 | RR (95% CI) van Eijk and others, unpublished‡89 | RR (95% CI) Bloland and others, 1995§94 |
---|---|---|---|---|
* RR = risk ratio; CI = confidence interval. Among infants of HIV-uninfected women, placental malaria was a consistent risk factor for post-neonatal infant mortality, and 56–71% of post-neonatal infant deaths may be attributed to placental malaria. | ||||
† Adjusted for low birth weight and fetal anemia. Adapted from Verhoeff and others95 after exclusion of twins. | ||||
‡ Adjusted for low birth weight. | ||||
§ Adjusted for low birth weight (Mantel-Haenszel summary risk ratio). | ||||
Negative | Negative | Reference | Reference | Reference |
Positive | Negative | 8.4 (3.3–21.6) | 7.1 (1.7–29.1) | 3.89 (1.17–12.91) |
Negative | Positive | 3.5 (1.1–10.8) | 3.1 (0.63–14.8) | 2.28 (0.94–5.53) |
Positive | Positive | 4.7 (1.2–19.1) | 4.5 (1.0–19.6) | 9.38 (3.85–22.84) |

Malaria during pregnancy, at delivery, and placental malaria by human immunodeficiency virus (HIV)–infected and uninfected women. A search was conducted in Pubmed for English publications using the keywords, HIV, malaria, pregnancy, or pregnant and we obtained further information from abstracts of scientific meetings, the internet, and personal communications with scientists. Only studies that provided data from all gravidity groups were included in this figure (Rogerson SJ and others, unpublished [UP*] data).20–29 We excluded the study by Parise and others99 because it included data only from primigravidae and secundigravidae, and that of Ticconi and others31 because it was not specified whether the malaria smears where taken during pregnancy or at the time of delivery, and from the mother or from the placenta. Placental malaria was determined by microscopy20,27,33 or placental histology (Rogerson SJ and others, unpublished data).29 One study used hematoxylin and eosin stain to meet the original goal of the study (determining the impact of sexually transmitted diseases on HIV transmission).28 pos. = positive; neg. = negative; RR = relative risk; CI = confidence interval; df = degrees of freedom.
Citation: The American Journal of Tropical Medicine and Hygiene Am J Trop Med Hyg 71, 2_suppl; 10.4269/ajtmh.2004.71.41

Malaria during pregnancy, at delivery, and placental malaria by human immunodeficiency virus (HIV)–infected and uninfected women. A search was conducted in Pubmed for English publications using the keywords, HIV, malaria, pregnancy, or pregnant and we obtained further information from abstracts of scientific meetings, the internet, and personal communications with scientists. Only studies that provided data from all gravidity groups were included in this figure (Rogerson SJ and others, unpublished [UP*] data).20–29 We excluded the study by Parise and others99 because it included data only from primigravidae and secundigravidae, and that of Ticconi and others31 because it was not specified whether the malaria smears where taken during pregnancy or at the time of delivery, and from the mother or from the placenta. Placental malaria was determined by microscopy20,27,33 or placental histology (Rogerson SJ and others, unpublished data).29 One study used hematoxylin and eosin stain to meet the original goal of the study (determining the impact of sexually transmitted diseases on HIV transmission).28 pos. = positive; neg. = negative; RR = relative risk; CI = confidence interval; df = degrees of freedom.
Citation: The American Journal of Tropical Medicine and Hygiene Am J Trop Med Hyg 71, 2_suppl; 10.4269/ajtmh.2004.71.41
Malaria during pregnancy, at delivery, and placental malaria by human immunodeficiency virus (HIV)–infected and uninfected women. A search was conducted in Pubmed for English publications using the keywords, HIV, malaria, pregnancy, or pregnant and we obtained further information from abstracts of scientific meetings, the internet, and personal communications with scientists. Only studies that provided data from all gravidity groups were included in this figure (Rogerson SJ and others, unpublished [UP*] data).20–29 We excluded the study by Parise and others99 because it included data only from primigravidae and secundigravidae, and that of Ticconi and others31 because it was not specified whether the malaria smears where taken during pregnancy or at the time of delivery, and from the mother or from the placenta. Placental malaria was determined by microscopy20,27,33 or placental histology (Rogerson SJ and others, unpublished data).29 One study used hematoxylin and eosin stain to meet the original goal of the study (determining the impact of sexually transmitted diseases on HIV transmission).28 pos. = positive; neg. = negative; RR = relative risk; CI = confidence interval; df = degrees of freedom.
Citation: The American Journal of Tropical Medicine and Hygiene Am J Trop Med Hyg 71, 2_suppl; 10.4269/ajtmh.2004.71.41

Proportion of overall malaria prevalence during pregnancy that can be attributed to human immunodeficiency virus type 1 (HIV-1) HIV-1 in the population as a function of HIV seroprevalence in pregnant women and by pregnancy order. G-All = all gravidae; G-1 = primigravidae; G-2= secundigravidae; G3+ = grandmultigravidae. The population attributable fraction (PAF) was calculated as 100 × [p(RR − 1)/(1 + p(RR − 1)] where p is the HIV seroprevalence and RR is the summary risk ratio for malaria parasitemia associated with HIV infection (1.58, see Figure 1). For example, 12.7% of the malaria during pregnancy can be attributed to HIV-1 alone in areas with an antenatal HIV seroprevalence of 25%.
Citation: The American Journal of Tropical Medicine and Hygiene Am J Trop Med Hyg 71, 2_suppl; 10.4269/ajtmh.2004.71.41

Proportion of overall malaria prevalence during pregnancy that can be attributed to human immunodeficiency virus type 1 (HIV-1) HIV-1 in the population as a function of HIV seroprevalence in pregnant women and by pregnancy order. G-All = all gravidae; G-1 = primigravidae; G-2= secundigravidae; G3+ = grandmultigravidae. The population attributable fraction (PAF) was calculated as 100 × [p(RR − 1)/(1 + p(RR − 1)] where p is the HIV seroprevalence and RR is the summary risk ratio for malaria parasitemia associated with HIV infection (1.58, see Figure 1). For example, 12.7% of the malaria during pregnancy can be attributed to HIV-1 alone in areas with an antenatal HIV seroprevalence of 25%.
Citation: The American Journal of Tropical Medicine and Hygiene Am J Trop Med Hyg 71, 2_suppl; 10.4269/ajtmh.2004.71.41
Proportion of overall malaria prevalence during pregnancy that can be attributed to human immunodeficiency virus type 1 (HIV-1) HIV-1 in the population as a function of HIV seroprevalence in pregnant women and by pregnancy order. G-All = all gravidae; G-1 = primigravidae; G-2= secundigravidae; G3+ = grandmultigravidae. The population attributable fraction (PAF) was calculated as 100 × [p(RR − 1)/(1 + p(RR − 1)] where p is the HIV seroprevalence and RR is the summary risk ratio for malaria parasitemia associated with HIV infection (1.58, see Figure 1). For example, 12.7% of the malaria during pregnancy can be attributed to HIV-1 alone in areas with an antenatal HIV seroprevalence of 25%.
Citation: The American Journal of Tropical Medicine and Hygiene Am J Trop Med Hyg 71, 2_suppl; 10.4269/ajtmh.2004.71.41

Model of immune interactions during placental malaria and human immunodeficiency virus type 1 (HIV-1) co-infection and their putative impact on mother-to-child transmission of HIV-1. Infection of the placenta with Plasmodium falciparum activates the immune system and results in up-regulation of placental proinflammatory cytokines, especially tumor necrosis factor-alpha (TNF-α), which in turn could increase HIV-1 replication. Placental malaria also results in the increased infiltration of mononuclear cells in the inter-villous spaces of the placenta (IVBMC) and increased expression of CC chemokine receptor 5 (CCR5) on placental macrophages and fetal Hofbauer cells. These changes may further increase the placental HIV-1 viral load. With a healthy immune status, tightly regulated proinflammatory response, and CC-chemokine responses to malaria (which block HIV-1 entry through the CCR5 receptor) may successfully control malaria parasite densities and favor protection against mother-to-child transmission (MTCT) by controlling viral load and promoting other as yet undefined protective factors. Conversely, these putative protective factors may be overwhelmed in women with high-density malaria infections and/or HIV-related immunosuppression associated with high placental HIV-1 viral load and sub-optimal immune response to malaria. This could result in a local environment that favors MTCT of HIV-1.
Citation: The American Journal of Tropical Medicine and Hygiene Am J Trop Med Hyg 71, 2_suppl; 10.4269/ajtmh.2004.71.41

Model of immune interactions during placental malaria and human immunodeficiency virus type 1 (HIV-1) co-infection and their putative impact on mother-to-child transmission of HIV-1. Infection of the placenta with Plasmodium falciparum activates the immune system and results in up-regulation of placental proinflammatory cytokines, especially tumor necrosis factor-alpha (TNF-α), which in turn could increase HIV-1 replication. Placental malaria also results in the increased infiltration of mononuclear cells in the inter-villous spaces of the placenta (IVBMC) and increased expression of CC chemokine receptor 5 (CCR5) on placental macrophages and fetal Hofbauer cells. These changes may further increase the placental HIV-1 viral load. With a healthy immune status, tightly regulated proinflammatory response, and CC-chemokine responses to malaria (which block HIV-1 entry through the CCR5 receptor) may successfully control malaria parasite densities and favor protection against mother-to-child transmission (MTCT) by controlling viral load and promoting other as yet undefined protective factors. Conversely, these putative protective factors may be overwhelmed in women with high-density malaria infections and/or HIV-related immunosuppression associated with high placental HIV-1 viral load and sub-optimal immune response to malaria. This could result in a local environment that favors MTCT of HIV-1.
Citation: The American Journal of Tropical Medicine and Hygiene Am J Trop Med Hyg 71, 2_suppl; 10.4269/ajtmh.2004.71.41
Model of immune interactions during placental malaria and human immunodeficiency virus type 1 (HIV-1) co-infection and their putative impact on mother-to-child transmission of HIV-1. Infection of the placenta with Plasmodium falciparum activates the immune system and results in up-regulation of placental proinflammatory cytokines, especially tumor necrosis factor-alpha (TNF-α), which in turn could increase HIV-1 replication. Placental malaria also results in the increased infiltration of mononuclear cells in the inter-villous spaces of the placenta (IVBMC) and increased expression of CC chemokine receptor 5 (CCR5) on placental macrophages and fetal Hofbauer cells. These changes may further increase the placental HIV-1 viral load. With a healthy immune status, tightly regulated proinflammatory response, and CC-chemokine responses to malaria (which block HIV-1 entry through the CCR5 receptor) may successfully control malaria parasite densities and favor protection against mother-to-child transmission (MTCT) by controlling viral load and promoting other as yet undefined protective factors. Conversely, these putative protective factors may be overwhelmed in women with high-density malaria infections and/or HIV-related immunosuppression associated with high placental HIV-1 viral load and sub-optimal immune response to malaria. This could result in a local environment that favors MTCT of HIV-1.
Citation: The American Journal of Tropical Medicine and Hygiene Am J Trop Med Hyg 71, 2_suppl; 10.4269/ajtmh.2004.71.41
Authors’ addresses: Feiko O. ter Kuile and Francine H. Verhoeff, Child and Reproductive Health Group, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, United Kingdom, E-mail: terkuile@liv.ac.uk. Monica E. Parise, Venkatachalam Udhayakumar, Robert D. Newman, and Richard W. Steketee, Malaria Branch, Division of Parasitic Diseases, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Mail-stop F-22, 4770 Buford Highway, Chamblee, Atlanta GA 30341. Anne M. van Eijk, Kenya Medical Research Institute, Vector Biology and Control Research Centre, PO Box 1578, Kisumu, Kenya. Stephen J. Rogerson, Department of Medicine (Royal Melbourne Hospital/Western Hospital), University of Melbourne, Post Office, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
Acknowledgments: We thank the many colleagues that contributed to these ideas, discussion, and field studies. We are particularly grateful to the following colleagues for providing details or unpublished results for this review: Dr. Steven Meshnick (University of North Carolina, Chapel Hill, NC); Dr. Victor Mwapasa (College of Medicine, Blantyre, Malawi and University of Michigan, Ann Arbor, MI); and Dr. Ingrid Inion and Professor Marleen Temmerman (University of Ghent, Ghent, Belgium).
Financial support: Feiko O. ter Kuile is grateful to Roll Back Malaria/World Health Organization and the Centers for Disease Control and Prevention for support as part of a collaboration between the Liver-pool School of Tropical Medicine and the Malaria Branch of the Division of Parasitic Diseases, National Center for Infectious Diseases, Centers for Disease Control and Prevention. Francine H. Verhoeff was supported by funds from PREgnancy Malaria and Anemia European Union (PREMA-EU), which received financial support from the European Commission Research Directorate Fifth Framework Program (contract no PREMA-EU-ICA4-CT4-2001-10012). Stephen J. Rogerson is supported by the Wellcome Trust, United Kingdom, through an Overseas Senior Research Fellowship. Anne M. van Eijk is supported by the Oak Ridge Institute for Science and Education, Tennessee.
Disclaimer: Use of trade names is for identification only and does not imply endorsement by U.S. Public Health Service or Department of Health and Human Services.
REFERENCES
- 1↑
Regional Office for Africa of the World Health Organization, 2004. A Policy Framework for Malaria Prevention and Control During Pregnancy in the African Region, Final Draft December 19, 2003. Harare: World Health Organization.
- 2↑
Steketee RW, Wirima JJ, Slutsker L, Heymann DL, Breman JG, 1996. The problem of malaria and malaria control in pregnancy in sub-Saharan Africa. Am J Trop Med Hyg 55 :2–7.
- 3↑
Brabin BJ, 1983. An analysis of malaria in pregnancy in Africa. Bull World Health Organ 61 :1005–1016.
- 4↑
Mockenhaupt FP, Ulmen U, von Gaertner C, Bedu-Addo G, Bienzle U, 2002. Diagnosis of placental malaria. J Clin Microbiol 40 :306–308.
- 5↑
Leke RF, Djokam RR, Mbu R, Leke RJ, Fogako J, Megnekou R, Metenou S, Sama G, Zhou Y, Cadigan T, Parra M, Taylor DW, 1999. Detection of the Plasmodium falciparum antigen histidine-rich protein 2 in blood of pregnant women: implications for diagnosing placental malaria. J Clin Microbiol 37 :2992–2996.
- 6↑
Shulman CE, 1999. Malaria in pregnancy: its relevance to safe-motherhood programmes. Ann Trop Med Parasitol 93 (Suppl 1):S59–S66.
- 7↑
Dabis F, Ekpini ER, 2002. HIV-1/AIDS and maternal and child health in Africa. Lancet 359 :2097–2104.
- 8↑
De Cock KM, Fowler MG, Mercier E, de Vincenzi I, Saba J, Hoff E, Alnwick DJ, Rogers M, Shaffer N, 2000. Prevention of mother-to-child HIV transmission in resource-poor countries: translating research into policy and practice. JAMA 283 :1175–1182.
- 10↑
Leke RG, 2001. The state of immunology in Africa: HIV/AIDS and malaria. Curr Opin Immunol 13 :523–527.
- 11↑
French N, Nakiyingi J, Lugada E, Watera C, Whitworth JA, Gilks CF, 2001. Increasing rates of malarial fever with deteriorating immune status in HIV-1-infected Ugandan adults. AIDS 15 :899–906.
- 12↑
Whitworth J, Morgan D, Quigley M, Smith A, Mayanja B, Eotu H, Omoding N, Okongo M, Malamba S, Ojwiya A, 2000. Effect of HIV-1 and increasing immunosuppression on malaria parasitaemia and clinical episodes in adults in rural Uganda: a cohort study. Lancet 356 :1051–1056.
- 13
Francesconi P, Fabiani M, Dente MG, Lukwiya M, Okwey R, Ouma J, Ochakachon R, Cian F, Declich S, 2001. HIV, malaria parasites, and acute febrile episodes in Ugandan adults: a case-control study. AIDS 15 :2445–2450.
- 14
Grimwade K, French N, Mbatha DD, Zunga DD, Dedicoat M, Gilks CF, 2004.HIV-infection as a co-factor for severe falciparum malaria in adults living in a region of unstable malaria transmission in South Africa. AIDS 18 :547–554.
- 15↑
Grimwade K, French N, Mbatha DD, Zungu DD, Dedicoat M, Gilks CF, 2003. Childhood malaria in a region of unstable transmission and high human immunodeficiency virus prevalence. Pediatr Infect Dis J 22 :1057–1063.
- 16↑
Chandramohan D, Greenwood BM, 1998. Is there an interaction between human immunodeficiency virus and Plasmodium falciparum? Int J Epidemiol 27 :296–301.
- 17
French N, Gilks CF, 2000. Royal Society of Tropical Medicine and Hygiene Meeting at Manson House, London, March 18, 1999. Fresh from the field: some controversies in tropical medicine and hygiene. HIV and malaria, do they interact? Trans R Soc Trop Med Hyg 94 :233–237.
- 18↑
Corbett EL, Steketee RW, ter Kuile FO, Latif AS, Kamali A, Hayes RJ, 2002. HIV-1/AIDS and the control of other infectious diseases in Africa. Lancet 359 :2177–2187.
- 19↑
Rowland-Jones SL, Lohman B, 2002. Interactions between malaria and HIV infection-an emerging public health problem? Microbes Infect 4 :1265–1270.
- 20↑
Steketee RW, Wirima JJ, Bloland PB, Chilima B, Mermin JH, Chitsulo L, Breman JG, 1996. Impairment of a pregnant woman’s acquired ability to limit Plasmodium falciparum by infection with human immunodeficiency virus type-1. Am J Trop Med Hyg 55 :42–49.
- 21
Shulman CE, Graham WJ, Jilo H, Lowe BS, New L, Obiero J, Snow RW, Marsh K, 1996. Malaria is an important cause of anaemia in primigravidae: evidence from a district hospital in coastal Kenya. Trans R Soc Trop Med Hyg 90 :535–539.
- 22↑
Verhoeff FH, 2003. HIV and Malaria in Pregnancy. Symposium presentation. PREMA-EU, ed. Malaria and Pregnancy, PREMA. 52nd Annual Meeting of the American Society of Tropical Medicine and Hygiene. Philadelphia, PA.
- 23
Steketee RW, Wirima JJ, Slutsker L, Breman JG, Heymann DL, 1996. Comparability of treatment groups and risk factors for parasitemia at the first antenatal clinic visit in a study of malaria treatment and prevention in pregnancy in rural Malawi. Am J Trop Med Hyg 55 :17–23.
- 24
Kalyesubula I, Musoke-Mudido P, Marum L, Bagenda D, Aceng E, Ndugwa C, Olness K, 1997. Effects of malaria infection in human immunodeficiency virus type 1-infected Ugandan children. Pediatr Infect Dis J 16 :876–881.
- 25↑
Weng S, Bulterys M, Chao A, Stidley CA, Dushimimana A, Mbarutso E, Saah A, 1998. Perinatal human immunodeficiency virus-1 transmission and intrauterine growth: a cohort study in Butare, Rwanda. Pediatrics 102 :e24.
- 26
Ladner J, Leroy V, Simonon A, Karita E, Bogaerts J, de Clercq A, van de Perre P, Dabis F, 2002. HIV infection, malaria, and pregnancy: a prospective cohort study in Kigali, Rwanda. Am J Trop Med Hyg 66 :56–60.
- 27↑
van Eijk AM, Ayisi JG, ter Kuile FO, Misore AO, Otieno JA, Rosen DH, Kager PA, Steketee RW, Nahlen BL, 2003. HIV increases the risk of malaria in women of all gravidities in Kisumu, Kenya. AIDS 17 :595–603.
- 28↑
Brahmbhatt H, Kigozi G, Wabwire-Mangen F, Serwadda D, Sewankambo N, Lutalo T, Wawer MJ, Abramowsky C, Sullivan D, Gray R, 2003. The effects of placental malaria on mother-to-child HIV transmission in Rakai, Uganda. AIDS 17 :2539–2541.
- 29↑
Inion I, Mwanyumba F, Gaillard P, Chohan V, Verhofstede C, Claeys P, Mandaliya K, van Marck E, Temmerman M, 2003. Placental malaria and perinatal transmission of human immunodeficiency virus type 1. J Infect Dis 188 :1675–1678.
- 30↑
Parise ME, Ayisi JG, Nahlen BL, Schultz LJ, Roberts JM, Misore A, Muga R, Oloo AJ, Steketee RW, 1998. Efficacy of sulfadoxine-pyrimethamine for prevention of placental malaria in an area of Kenya with a high prevalence of malaria and human immunodeficiency virus infection. Am J Trop Med Hyg 59 :813–822.
- 31↑
Ticconi C, Mapfumo M, Dorrucci M, Naha N, Tarira E, Pietropolli A, Rezza G, 2003. Effect of maternal HIV and malaria infection on pregnancy and perinatal outcome in Zimbabwe. J Acquir Immune Defic Syndr Hum Retrovirol 34 :289–294.
- 32↑
Mount AM, Mwapasa V, Elliott SR, Beeson JG, Tadesse E, Lema VM, Molyneux ME, Meshnick SR, Rogerson SJ, 2004. HIV infection impairs humoral immunity to Plasmodium falciparum malaria in pregnancy. Lancet: (in press).
- 33↑
Verhoeff FH, Brabin BJ, Hart CA, Chimsuku L, Kazembe P, Broadhead RL, 1999. Increased prevalence of malaria in HIV-infected pregnant women and its implications for malaria control. Trop Med Int Health 4 :5–12.
- 34↑
Mocroft A, Kirk O, Barton SE, Dietrich M, Proenca R, Colebunders R, Pradier C, dAarminio MA, Ledergerber B, Lundgren JD, 1999. Anaemia is an independent predictive marker for clinical prognosis in HIV-infected patients from across Europe. EuroSIDA study group. AIDS 13 :943–950.
- 35↑
Moses A, Nelson J, Bagby GC Jr, 1998. The influence of human immunodeficiency virus-1 on hematopoiesis. Blood 91 :1479–1495.
- 36↑
Guyatt HL, Snow RW, 2001. The epidemiology and burden of Plasmodium falciparum-related anemia among pregnant women in sub-Saharan Africa. Am J Trop Med Hyg 64 :36–44.
- 37↑
Ayisi JG, van Eijk AM, ter Kuile FO, Kolczak MS, Otieno JA, Misore AO, Kager PA, Steketee RW, Nahlen BL, 2003. The effect of dual infection with HIV and malaria on pregnancy outcome in western Kenya. AIDS 17 :585–594.
- 38↑
van Eijk AM, Ayisi JG, ter Kuile FO, Misore A, Otieno JA, Kolczak MS, Kager PA, Steketee RW, Nahlen BL, 2001. Human immunodeficiency virus seropositivity and malaria as risk factors for third-trimester anemia in asymptomatic pregnant women in western Kenya. Am J Trop Med Hyg 65 :623–630.
- 39↑
Leroy V, Ladner J, Nyiraziraje M, De Clercq A, Bazubagira A, van de Perre P, Karita E, Dabis F, 1998. Effect of HIV-1 infection on pregnancy outcome in women in Kigali, Rwanda, 1992–1994. Pregnancy and HIV Study Group. AIDS 12 :643–650.
- 40↑
Steketee RW, Wirima JJ, Hightower AW, Slutsker L, Heymann DL, Breman JG, 1996. The effect of malaria and malaria prevention in pregnancy on offspring birthweight, prematurity, and intrauterine growth retardation in rural Malawi. Am J Trop Med Hyg 55 :33–41.
- 41↑
Walker N, Schwartlander B, Bryce J, 2002. Meeting international goals in child survival and HIV/AIDS. Lancet 360 :284–289.
- 42↑
Steketee RW, Nahlen BL, Parise ME, Menendez C, 2001. The burden of malaria in pregnancy in malaria-endemic areas. Am J Trop Med Hyg 64 :28–35.
- 43↑
Moore JM, Nahlen BL, Misore A, Lal AA, Udhayakumar V, 1999. Immunity to placental malaria. I. Elevated production of interferon-gamma by placental blood mononuclear cells is associated with protection in an area with high transmission of malaria. J Infect Dis 179 :1218–1225.
- 44↑
Moore JM, Ayisi J, Nahlen BL, Misore A, Lal AA, Udhayakumar V, 2000. Immunity to placental malaria. II. Placental antigen-specific cytokine responses are impaired in human immunodeficiency virus-infected women. J Infect Dis 182 :960–964.
- 45↑
Chaisavaneeyakorn S, Moore JM, Otieno J, Chaiyaroj SC, Perkins DJ, Shi YP, Nahlen BL, Lal AA, Udhayakumar V, 2002. Immunity to placental malaria. III. Impairment of interleukin (IL)-12, not IL-18, and interferon-inducible protein-10 responses in the placental intervillous blood of human immunodeficiency virus/malaria-coinfected women. J Infect Dis 185 :127–131.
- 46↑
Chaisavaneeyakorn S, Moore JM, Othoro C, Otieno J, Chaiyaroj SC, Shi YP, Nahlen BL, Lal AA, Udhayakumar V, 2002. Immunity to placental malaria. IV. Placental malaria is associated with up-regulation of macrophage migration inhibitory factor in intervillous blood. J Infect Dis 186 :1371–1375.
- 47↑
Ayisi JG, Branch OH, Rafi-Janajreh A, van Eijk AM, ter Kuile FO, Rosen DH, Kager PA, Lanar DE, Barbosa A, Kaslow D, Nahlen BL, Lal AA, 2003. Does infection with human immunodeficiency virus affect the antibody responses to Plasmodium falciparum antigenic determinants in asymptomatic pregnant women? J Infect 46 :164–172.
- 48↑
Fried M, Duffy PE, 1996. Adherence of Plasmodium falciparum to chondroitin sulfate A in the human placenta. Science 272 :1502–1504.
- 49↑
Quinn TC, Piot P, McCormick JB, Feinsod FM, Taelman H, Kapita B, Stevens W, Fauci AS, 1987. Serologic and immunologic studies in patients with AIDS in North America and Africa. The potential role of infectious agents as cofactors in human immunodeficiency virus infection. JAMA 257 :2617–2621.
- 50↑
Hoffman IF, Jere CS, Taylor TE, Munthali P, Dyer JR, Wirima JJ, Rogerson SJ, Kumwenda N, Eron JJ, Fiscus SA, Chakraborty H, Taha TE, Cohen MS, Molyneux ME, 1999. The effect of Plasmodium falciparum malaria on HIV-1 RNA blood plasma concentration. AIDS 13 :487–494.
- 51↑
Kublin JG, Jere CS, Miller WC, Hoffman IF, Chimbiya N, Taylor TE, Molyneux ME, 2002. Malaria Infection Elevates HIV-1 Viral Load. AIDS 2002 The XIV International AIDS Conference. Barcelona, Spain.
- 52↑
Kapiga SH, Bang H, Spiegelman D, Msamanga GI, Coley J, Hunter DJ, Fawzi WW, 2002. Correlates of plasma HIV-1 RNA viral load among HIV-1-seropositive women in Dar es Salaam, Tanzania. J Acquir Immune Defic Syndr Hum Retrovirol 30 :316–323.
- 53↑
Mwapasa V, Rogerson SJ, Molyneux ME, Abrams ET, Kamwendo DD, Lema VM, Tadesse E, Chaluluka E, Wilson PE, Meshnick SR, 2004. The effect of Plasmodium falciparum malaria on peripheral and placental HIV-1 RNA concentrations in pregnant Malawian women. AIDS 18 :1051–1059.
- 54↑
Ayisi JG, van Eijk AM, Newman RD, ter Kuile FO, Shi YP, Yang C, Kolczak MS, Otieno JA, Misore AO, Kager PA, Lal RB, Steketee RW, Nahlen BL, 2004. Maternal malaria infection and perinatal HIV transmission in a malarious area of western Kenya. Emerg Infect Dis 10 :643–652.
- 55↑
Xiao L, Owen SM, Rudolph DL, Lal RB, Lal AA, 1998. Plasmodium falciparum antigen-induced human immunodeficiency virus type 1 replication is mediated through induction of tumor necrosis factor- alpha. J Infect Dis 177 :437–445.
- 56↑
Pisell TL, Hoffman IF, Jere CS, Ballard SB, Molyneux ME, Butera ST, Lawn SD, 2002. Immune activation and induction of HIV-1 replication within CD14 macrophages during acute Plasmodium falciparum malaria coinfection. AIDS 16 :1503–1509.
- 57↑
Freitag C, Chougnet C, Schito M, Near KA, Shearer GM, Li C, Langhorne J, Sher A, 2001. Malaria infection induces virus expression in human immunodeficiency virus transgenic mice by CD4 T cell-dependent immune activation. J Infect Dis 183 :1260–1268.
- 58↑
Ordi J, Ismail MR, Ventura PJ, Kahigwa E, Hirt R, Cardesa A, Alonso PL, Menendez C, 1998. Massive chronic intervillositis of the placenta associated with malaria infection. Am J Surg Pathol 22 :1006–1011.
- 59
Bulmer JN, Rasheed FN, Francis N, Morrison L, Greenwood BM, 1993. Placental malaria. I. Pathological classification. Histopathology 22 :211–218.
- 60
Ordi J, Menendez C, Ismail MR, Ventura PJ, Palacin A, Kahigwa E, Ferrer B, Cardesa A, Alonso PL, 2001. Placental malaria is associated with cell-mediated inflammatory responses with selective absence of natural killer cells. J Infect Dis 183 :1100–1107.
- 61
Ismail MR, Ordi J, Menendez C, Ventura PJ, Aponte JJ, Kahigwa E, Hirt R, Cardesa A, Alonso PL, 2000. Placental pathology in malaria: a histological, immunohistochemical, and quantitative study. Hum Pathol 31 :85–93.
- 62
Rogerson SJ, Pollina E, Getachew A, Tadesse E, Lema VM, Molyneux ME, 2003. Placental monocyte infiltrates in response to Plasmodium falciparum malaria infection and their association with adverse pregnancy outcomes. Am J Trop Med Hyg 68 :115–119.
- 63↑
Chaisavaneeyakorn S, Moore JM, Mirel L, Othoro C, Otieno J, Chaiyaroj SC, Shi YP, Nahlen BL, Lal AA, Udhayakumar V, 2003. Levels of macrophage inflammatory protein 1 alpha (MIP-1 alpha) and MIP-1 beta in intervillous blood plasma samples from women with placental malaria and human immunodeficiency virus infection. Clin Diagn Lab Immunol 10 :631–636.
- 64↑
Tkachuk AN, Moormann AM, Poore JA, Rochford RA, Chensue SW, Mwapasa V, Meshnick SR, 2001. Malaria enhances expression of CC chemokine receptor 5 on placental macrophages. J Infect Dis 183 :967–972.
- 65↑
Salvatori F, Scarlatti G, 2001. HIV type 1 chemokine receptor usage in mother-to-child transmission. AIDS Res Hum Retroviruses 17 :925–935.
- 66↑
Wu L, Paxton WA, Kassam N, Ruffing N, Rottman JB, Sullivan N, Choe H, Sodroski J, Newman W, Koup RA, Mackay CR, 1997. CCR5 levels and expression pattern correlate with infectability by macrophage-tropic HIV-1, in vitro.J Exp Med 185 :1681–1691.
- 67↑
Wolinsky SM, Wike CM, Korber BT, Hutto C, Parks WP, Rosenblum LL, Kunstman KJ, Furtado MR, Munoz JL, 1992. Selective transmission of human immunodeficiency virus type-1 variants from mothers to infants. Science 255 :1134–1137.
- 68↑
Leroy V, Montcho C, Manigart O, Van de Perre P, Dabis F, Msellati P, Meda N, You B, Simonon A, Rouzioux C, 2001. Maternal plasma viral load, zidovudine and mother-to-child transmission of HIV-1 in Africa: DITRAME ANRS 049a trial. AIDS 15 :517–522.
- 69
Mofenson LM, Lambert JS, Stiehm ER, Bethel J, Meyer WA III, Whitehouse J, Moye J Jr, Reichelderfer P, Harris DR, Fowler MG, Mathieson BJ, Nemo GJ, 1999. Risk factors for perinatal transmission of human immunodeficiency virus type 1 in women treated with zidovudine. Pediatric AIDS Clinical Trials Group Study 185 Team. N Engl J Med 341 :385–393.
- 70↑
Shaffer N, Roongpisuthipong A, Siriwasin W, Chotpitaya-sunondh T, Chearskul S, Young NL, Parekh B, Mock PA, Bhadrakom C, Chinayon P, Kalish ML, Phillips SK, Granade TC, Subbarao S, Weniger BG, Mastro TD, 1999. Maternal virus load and perinatal human immunodeficiency virus type 1 subtype E transmission, Thailand. Bangkok Collaborative Perinatal HIV Transmission Study Group. J Infect Dis 179 :590–599.
- 71↑
St Louis ME, Kamenga M, Brown C, Nelson AM, Manzila T, Batter V, Behets F, Kabagabo U, Ryder RW, Oxtoby M, Quinn TC, Heyward WL, 1993. Risk for perinatal HIV-1 transmission according to maternal immunologic, virologic, and placental factors. JAMA 269 :2853–2859.
- 72↑
Abrams ET, Brown H, Chensue SW, Turner GD, Tadesse E, Lema VM, Molyneux ME, Rochford R, Meshnick SR, Rogerson SJ, 2003. Host response to malaria during pregnancy: placental monocyte recruitment is associated with elevated beta chemokine expression. J Immunol 170 :2759–2764.
- 73↑
Alkhatib G, Combadiere C, Broder CC, Feng Y, Kennedy PE, Murphy PM, Berger EA, 1996. CC CKR5: a RANTES, MIP-1alpha, MIP-1beta receptor as a fusion cofactor for macrophage-tropic HIV-1. Science 272 :1955–1958.
- 74↑
Wasik TJ, Bratosiewicz J, Wierzbicki A, Whiteman VE, Rutstein RR, Starr SE, Douglas SD, Kaufman D, Sison AV, Polansky M, Lischner HW, Kozbor D, 1999. Protective role of beta-chemokines associated with HIV-specific Th responses against perinatal HIV transmission. J Immunol 162 :4355–4364.
- 75↑
Suguitan AL Jr, Leke RG, Fouda G, Zhou A, Thuita L, Metenou S, Fogako J, Megnekou R, Taylor DW, 2003. Changes in the levels of chemokines and cytokines in the placentas of women with Plasmodium falciparum malaria. J Infect Dis 188 :1074–1082.
- 76↑
Yang C, Li M, Newman RD, Shi YP, Ayisi J, van Eijk AM, Otieno J, Misore AO, Steketee RW, Nahlen BL, Lal RB, 2003. Genetic diversity of HIV-1 in western Kenya: subtype-specific differences in mother-to-child transmission. AIDS 17 :1667–1674.
- 77↑
Paxton WA, Koup RA, 1997. Mechanisms of resistance to HIV infection. Springer Semin Immunopathol 18 :323–340.
- 78↑
Kuhn L, Coutsoudis A, Moodley D, Trabattoni D, Mngqundaniso N, Shearer GM, Clerici M, Coovadia HM, Stein Z, 2001. T-helper cell responses to HIV envelope peptides in cord blood: protection against intrapartum and breast-feeding transmission. AIDS 15 :1–9.
- 80↑
van Eijk AM, Ayisi JG, ter Kuile FO, Misore AO, Otieno JA, Kolczak MS, Kager PA, Steketee RW, Nahlen BL, 2002. Malaria and human immunodeficiency virus infection as risk factors for anemia in infants in Kisumu, western Kenya. Am J Trop Med Hyg 67 :44–53.
- 81
Cornet M, Le Hesran JY, Fievet N, Cot M, Personne P, Gounoue R, Beyeme M, Deloron P, 1998. Prevalence of and risk factors for anemia in young children in southern Cameroon. Am J Trop Med Hyg 58 :606–611.
- 82
Reed SC, Wirima JJ, Steketee RW, 1994. Risk factors for anemia in young children in rural Malawi. Am J Trop Med Hyg 51 :170–174.
- 83
Le Hesran JY, Cot M, Personne P, Fievet N, Dubois B, Beyeme M, Boudin C, Deloron P, 1997. Maternal placental infection with Plasmodium falciparum and malaria morbidity during the first 2 years of life. Am J Epidemiol 146 :826–831.
- 84↑
Deloron P, Dubois B, Le Hesran JY, Riche D, Fievet N, Cornet M, Ringwald P, Cot M, 1997. Isotypic analysis of maternally transmitted Plasmodium falciparum-specific antibodies in Cameroon, and relationship with risk of P. falciparum infection. Clin Exp Immunol 110 :212–218.
- 85↑
le Cessie S, Verhoeff FH, Mengistie G, Kazembe P, Broadhead R, Brabin BJ, 2002. Changes in haemoglobin levels in infants in Malawi: effect of low birth weight and fetal anaemia. Arch Dis Child Fetal Neonatal Ed 86 :F182–F187.
- 86↑
Cot M, Le Hesran JY, Staalsoe T, Fievet N, Hviid L, Deloron P, 2003. Maternally transmitted antibodies to pregnancy-associated variant antigens on the surface of erythrocytes infected with Plasmodium falciparum: relation to child susceptibility to malaria. Am J Epidemiol 157 :203–209.
- 87
King CL, Malhotra I, Wamachi A, Kioko J, Mungai P, Wahab SA, Koech D, Zimmerman P, Ouma J, Kazura JW, 2002. Acquired immune responses to Plasmodium falciparum merozoite surface protein-1 in the human fetus. J Immunol 168 :356–364.
- 88↑
Xi G, Leke RG, Thuita LW, Zhou A, Leke RJ, Mbu R, Taylor DW, 2003. Congenital exposure to Plasmodium falciparum antigens: prevalence and antigenic specificity of in utero-produced antimalarial immunoglobulin M antibodies. Infect Immun 71 :1242–1246.
- 89↑
van Eijk AM, Ayisi JG, ter Kuile FO, Misore AO, Otieno JA, Kolczak MS, Kager PA, Steketee RW, Nahlen BL, 2002. Placental malaria and HIV infection as risk factors for post-neonatal infant mortality in Kisumu, Kenya. The XIV International AIDS Conference. Barcelona, Spain.
- 90↑
Verhoeff FH, Brabin BJ, Chimsuku L, Kazembe P, Russell WB, Broadhead RL, 1998. An evaluation of the effects of intermittent sulfadoxine-pyrimethamine treatment in pregnancy on parasite clearance and risk of low birthweight in rural Malawi. Ann Trop Med Parasitol 92 :141–150.
- 91↑
Slutsker L, Khoromana CO, Hightower AW, Macheso A, Wirima JJ, Breman JG, Heymann DL, Steketee RW, 1996. Malaria infection in infancy in rural Malawi. Am J Trop Med Hyg 55 :71–76.
- 92↑
Taha TE, Canner JK, Dallabetta GA, Chiphangwi JD, Liomba G, Wangel AM, Saah AJ, Miotti PG, 1994. Childhood malaria parasitaemia and human immunodeficiency virus infection in Malawi. Trans R Soc Trop Med Hyg 88 :164–165.
- 93↑
Mueller BU, 1994. Hematological problems and their management in children with HIV infection. Pizzo PA, Wilfert CM, eds. Pediatric AIDS: The Challenge of HIV Infection in Infants, Children and Adolescents. Baltimore: Williams & Wilkins, 591–602.
- 94↑
Bloland PB, Wirima JJ, Steketee RW, Chilima B, Hightower A, Breman JG, 1995. Maternal HIV infection and infant mortality in Malawi: evidence for increased mortality due to placental malaria infection. AIDS 9 :721–726.
- 95↑
Verhoeff FH, le Cessie S, Kalanda BF, Kazembe PN, Broad-head RL, Brabin BJ, 2004. Increase in post-neonatal infant mortality in Malawi: the importance of maternal. Ann Trop Paediatr: (in press).
- 96↑
WHO, 2003. A Policy Framework for Malaria Prevention and Control during Pregnancy in the African Region. Geneva: World Health Organization.
- 97↑
Shulman CE, Dorman EK, Cutts F, Kawuondo K, Bulmer JN, Peshu N, Marsh K, 1999. Intermittent sulphadoxine-pyrimethamine to prevent severe anaemia secondary to malaria in pregnancy: a randomised placebo-controlled trial. Lancet 353 :632–636.
- 98
Schultz LJ, Steketee RW, Macheso A, Kazembe P, Chitsulo L, Wirima JJ, 1994. The efficacy of antimalarial regimens containing sulfadoxine-pyrimethamine and/or chloroquine in preventing peripheral and placental Plasmodium falciparum infection among pregnant women in Malawi. Am J Trop Med Hyg 51 :515–522.
- 99↑
Parise ME, Ayisi JG, Nahlen BL, Schultz LJ, Roberts JM, Misore A, Muga R, Oloo AJ, Steketee RW, 1998. Efficacy of sulfadoxine-pyrimethamine for prevention of placental malaria in an area of Kenya with a high prevalence of malaria and human immunodeficiency virus infection. Am J Trop Med Hyg 59 :813–822.
- 100↑
Verhoeff FH, Brabin BJ, Chimsuku L, Kazembe P, Russell WB, Broadhead RL, 1998. An evaluation of the effects of intermittent sulfadoxine-pyrimethamine treatment in pregnancy on parasite clearance and risk of low birthweight in rural Malawi. Ann Trop Med Parasitol 92 :141–150.
- 101↑
WHO, 2002. WHO Antenatal Care Randomized Trial: Manual for the Implementation of the New Model. Geneva: World Health Organization.
- 102↑
WHO, 2004. Integrated Management of Pregnancy and Childbirth. Pregnancy, Childbirth and Postpartum Care. A Guide for Essential Practice. Geneva: World Health Organization.
- 103↑
Colebunders R, Ryder R, Francis H, Nekwei W, Bahwe Y, Lebughe I, Ndilu M, Vercauteren G, Nseka K, Perriens J, 1991. Seroconversion rate, mortality, and clinical manifestations associated with the receipt of a human immunodeficiency virus-infected blood transfusion in Kinshasa, Zaire. J Infect Dis 164 :450–456.
- 104↑
Kamya MR, Kigonya CN, McFarland W, 2001. HIV infection may adversely affect clinical response to chloroquine therapy for uncomplicated malaria in children. AIDS 15 :1187–1188.
- 105
Colebunders R, Bahwe Y, Nekwei W, Ryder R, Perriens J, Nsimba K, Turner A, Francis H, Lebughe I, van der Stuyft P, Piot P, 1990. Incidence of malaria and efficacy of oral quinine in patients recently infected with human immunodeficiency virus in Kinshasa, Zaire. J Infect 21 :167–173.
- 106↑
Greenberg A, Nsa W, Ryder R, Medi M, Nzeza M, Kitadi N, Baangi M, Malanda N, Davachi F, Hassig S, 1991. Plasmodium falciparum malaria and perinatally acquired human immunodeficiency virus type 1 infection in Kinshasa, Zaire. A prospective, longitudinal cohort study of 587 children. N Engl J Med 325 :105–109.
- 107
Muller O, Moser R, 1990. The clinical and parasitological presentation of Plasmodium falciparum malaria in Uganda is unaffected by HIV-1 infection. Trans R Soc Trop Med Hyg 84 :336–338.
- 108↑
Birku Y, Mekonnen E, Bjorkman A, Wolday D, 2002. Delayed clearance of Plasmodium falciparum in patients with human immunodeficiency virus co-infection treated with artemisinin. Ethiopian Med J 40 :17–26.
- 109↑
UNAIDS, 2000. Provisional WHO/UNAIDS Secretariat Recommendations on the Use of Cotrimoxazole Prophylaxis in Adults and Children Living with HIV/AIDS in Africa. Geneva: UNAIDS, World Health Organization.
- 110↑
Steketee RW, Wirima JJ, Slutsker L, Roberts JM, Khoromana CO, Heymann DL, Breman JG, 1996. Malaria parasite infection during pregnancy and at delivery in mother, placenta, and newborn: efficacy of chloroquine and mefloquine in rural Malawi. Am J Trop Med Hyg 55 :24–32.
- 111↑
Kayentao K, Kodio M, Mungai M, Parise ME, Coulibaly D, Issa C, Keita AS, Maiga B, Doumtabe D, Maiga H, Ongoiba A, Doumbo O, 2002. The Efficacy of Chloroquine- and Sulfadoxine-Pyrimethamine-Containing Regimens in the Prevention of Maternal Anemia, Placental Malaria, and Low Birthweight in Mali. The Third MIM Pan-African Malaria Conference. November 17–22, 2002. Arusha, Tanzania.
- 112↑
Steketee RW, Wirima JJ, Slutsker L, Heymann DL, Breman JG, 1996. The problem of malaria and malaria control in pregnancy in sub-Saharan Africa. Am J Trop Med Hyg 55 :2–7.
- 113↑
Newman RD, Parise ME, Slutsker L, Nahlen B, Steketee RW, 2003. Safety, efficacy and determinants of effectiveness of antimalarial drugs during pregnancy: implications for prevention programmes in Plasmodium falciparum-endemic sub-Saharan Africa. Trop Med Int Health 8 :488–506.
- 114↑
Feikin DR, Dowell SF, Nwanyanwu OC, Klugman KP, Kazembe PN, Barat LM, Graf C, Bloland PB, Ziba C, Huebner RE, Schwartz B, 2000. Increased carriage of trimethoprim/sulfamethoxazole-resistant Streptococcus pneumoniae in Malawian children after treatment for malaria with sulfadoxine/pyrimethamine. J Infect Dis 181 :1501–1505.
- 115↑
Mehaffey PC, Barrett MS, Putnam SD, Jones RN, 1995. Antigonococcal activity of 11 drugs used for therapy or prophylaxis of malaria. Diagn Microbiol Infect Dis 23 :11–13.
- 116↑
Bloland PB, Redd SC, Kazembe P, Tembenu R, Wirima JJ, Campbell CC, 1991. Co-trimoxazole for childhood febrile illness in malaria-endemic regions. Lancet 337 :518–520.
- 117↑
Omar SA, Bakari A, Owiti A, Adagu IS, Warhurst DC, 2001. Co-trimoxazole compared with sulfadoxine-pyrimethamine in the treatment of uncomplicated malaria in Kenyan children. Trans R Soc Trop Med Hyg 95 :657–660.
- 118↑
Anglaret X, Chene G, Attia A, Toure S, Lafont S, Combe P, Manlan K, N’Dri-Yoman T, Salamon R, 1999. Early chemoprophylaxis with trimethoprim-sulphamethoxazole for HIV-1-infected adults in Abidjan, Côte d’Ivoire: a randomised trial. Cotrimo-CI Study Group. Lancet 353 :1463–1468.
- 119↑
Iyer JK, Milhous WK, Cortese JF, Kublin JG, Plowe CV, 2001. Plasmodium falciparum cross-resistance between trimethoprim and pyrimethamine. Lancet 358 :1066–1067.
- 120↑
2002. Viracept, (Agouron). Monographs from the 2002 Physicians Desk Reference. Montvale, NJ: Medical Economics Company, Inc.
- 121↑
2002. Norvir, (Abbott). Monographs from the 2002 Physicians Desk Reference. Montvale, NJ: Medical Economics Company, Inc.
- 122↑
2002. Agenerase, (GlaxoSmithKline). Monographs from the 2002 Physicians Desk Reference. Montvale, NJ: Medical Economics Company, Inc.
- 123
2002. Rescriptor, (Agouron). Monographs from the 2002 Physicians Desk Reference. Montvale, NJ: Medical Economics Company, Inc.
- 124↑
2002. Kaletra, (Abbott). Monographs from the 2002 Physicians Desk Reference. Montvale, NJ: Medical Economics Company, Inc.
- 125↑
2002. Epivir, (GlaxoSmithKline). Monographs from the 2002 Physicians Desk Reference. Montvale, NJ: Medical Economics Company, Inc.
- 126↑
Nathoo S, Serghides L, Kain KC, 2003. Effect of HIV-1 antiret-roviral drugs on cytoadherence and phagocytic clearance of Plasmodium falciparum-parasitised erythrocytes. Lancet 362:1039–1041.
- 127
Serghides L, Nathoo S, Walmsley S, Kain KC, 2002. CD36 deficiency induced by antiretroviral therapy. AIDS 16 :353–358.
- 129↑
Sperber K, Kalb TH, Stecher VJ, Banerjee R, Mayer L, 1993. Inhibition of human immunodeficiency virus type 1 replication by hydroxychloroquine in T cells and monocytes. AIDS Res Hum Retroviruses 9 :91–98.
- 130
Chiang G, Sassaroli M, Louie M, Chen H, Stecher VJ, Sperber K, 1996. Inhibition of HIV-1 replication by hydroxychloroquine: mechanism of action and comparison with zidovudine. Clin Ther 18 :1080–1092.
- 131
Tsai WP, Nara PL, Kung HF, Oroszlan S, 1990. Inhibition of human immunodeficiency virus infectivity by chloroquine. AIDS Res Hum Retroviruses 6 :481–489.
- 132↑
Savarino A, Gennero L, Chen HC, Serrano D, Malavasi F, Boelaert JR, Sperber K, 2001. Anti-HIV effects of chloroquine: mechanisms of inhibition and spectrum of activity. AIDS 15 :2221–2229.
- 133↑
Sperber K, Louie M, Kraus T, Proner J, Sapira E, Lin S, Stecher V, Mayer L, 1995. Hydroxychloroquine treatment of patients with human immunodeficiency virus type 1. Clin Ther 17 :622–636.
- 134↑
Sperber K, Chiang G, Chen H, Ross W, Chusid E, Gonchar M, Chow R, Liriano O, 1997. Comparison of hydroxychloroquine with zidovudine in asymptomatic patients infected with human immunodeficiency virus type 1. Clin Ther 19 :913–923.
- 135↑
Savarino A, Gennero L, Sperber K, Boelaert JR, 2001. The anti-HIV-1 activity of chloroquine. J Clin Virol 20 :131–135.
- 136
Savarino A, Boelaert JR, Cassone A, Majori G, Cauda R, 2003. Effects of chloroquine on viral infections: an old drug against today’s diseases? Lancet Infect Dis 3 :722–727.
- 137↑
Boelaert JR, Sperber K, Piette J, 1999. Chloroquine exerts an additive in vitro anti-HIV type 1 effect when associated with didanosine and hydroxyurea. AIDS Res Hum Retroviruses 15 :1241–1247.
- 138↑
Boelaert JR, Piette J, Sperber K, 2001. The potential place of chloroquine in the treatment of HIV-1-infected patients. J Clin Virol 20 :137–140.
- 139↑
Paton NI, Aboulhab J, Karim F, 2002. Hydroxychloroquine, hydroxycarbamide, and didanosine as economic treatment for HIV-1. Lancet 359 :1667–1668.
- 140↑
Boelaert JR, Yaro S, Augustijns P, Meda N, Schneider YJ, Schols D, Mols R, De Laere EA, Van de Perre P, 2001. Chloroquine accumulates in breast-milk cells: potential impact in the prophylaxis of postnatal mother-to-child transmission of HIV-1. AIDS 15 :2205–2207.
- 141↑
Friis H, Gomo E, Koestel P, Ndhlovu P, Nyazema N, Krarup H, Michaelsen KF, 2001. HIV and other predictors of serum folate, serum ferritin, and hemoglobin in pregnancy: a cross-sectional study in Zimbabwe. Am J Clin Nutr 73 :1066–1073.
- 142↑
Boelaert JR, Weinberg GA, Weinberg ED, 1996. Altered iron metabolism in HIV infection: mechanisms, possible consequences, and proposals for management. Infect Agents Dis 5 :36–46.
- 143↑
Friis H, Gomo E, Nyazema N, Ndhlovu P, Krarup H, Madsen PH, Michaelsen KF, 2003. Iron, haptoglobin phenotype, and HIV-1 viral load: a cross-sectional study among pregnant Zimbabwean women. J Acquir Immune Defic Syndr Hum Retrovirol 33 :74–81.
- 145↑
Georgiou NA, van der Bruggen T, Oudshoorn M, Nottet HS, Marx JJ, van Asbeck BS, 2000. Inhibition of human immunodeficiency virus type 1 replication in human mononuclear blood cells by the iron chelators deferoxamine, deferiprone, and bleomycin. J Infect Dis 181 :484–490.
- 146↑
Jacobus DP, 1996. Randomization to iron supplementation of patients with advanced human immunodeficiency virus disease–an inadvertent but controlled study with results important for patient care. J Infect Dis 173 :1044–1045.
- 147↑
Salmon-Ceron D, Fontbonne A, Saba J, May T, Raffi F, Chidiac C, Patey O, Aboulker JP, Schwartz D, Vilde JL, 1995. Lower survival in AIDS patients receiving dapsone compared with aerosolized pentamidine for secondary prophylaxis of Pneumocystis carinii pneumonia. Study Group. J Infect Dis 172 :656–664.
- 148↑
Salhi Y, Costagliola D, Rebulla P, Dessi C, Karagiorga M, Lena-Russo D, de Montalembert M, Girot R, 1998. Serum ferritin, desferrioxamine, and evolution of HIV-1 infection in thalassemic patients. J Acquir Immune Defic Syndr Hum Retrovirol 18 :473–478.
- 149↑
de Monye C, Karcher DS, Boelaert JR, Gordeuk VR, 1999. Bone marrow macrophage iron grade and survival of HIV-seropositive patients. AIDS 13 :375–380.
- 150↑
Teira R, Virosta M, Munoz J, Zubero Z, Santamaria JM, 1997. The safety of pyrimethamine and sulfadoxine for the prevention of Pneumocystis carinii pneumonia. Scand J Infect Dis 29 :595–596.
- 151↑
Saiag P, Caumes E, Chosidow O, Revuz J, Roujeau JC, 1992. Drug-induced toxic epidermal necrolysis (Lyell syndrome) in patients infected with the human immunodeficiency virus. J Am Acad Dermatol 26 :567–574.
- 152↑
Nunn P, Kibuga D, Gathua S, Brindle R, Imalingat A, Wasunna K, Lucas S, Gilks C, Omwega M, Were J, McAdam K, 1991. Cutaneous hypersensitivity reactions due to thiacetazone in HIV-1 seropositive patients treated for tuberculosis. Lancet 337 :627–630.
- 154↑
Rieder MJ, Shear NH, Kanee A, Tang BK, Spielberg SP, 1991. Prominence of slow acetylator phenotype among patients with sulfonamide hypersensitivity reactions. Clin Pharmacol Ther 49 :13–17.
- 155↑
Wolkenstein P, Carriere V, Charue D, Bastuji-Garin S, Revuz J, Roujeau JC, Beaune P, Bagot M, 1995. A slow acetylator genotype is a risk factor for sulphonamide-induced toxic epidermal necrolysis and Stevens-Johnson syndrome. Pharmacogenetics 5 :255–258.
- 156↑
Lee BL, Wong D, Benowitz NL, Sullam PM, 1993. Altered patterns of drug metabolism in patients with acquired immunodeficiency syndrome. Clin Pharmacol Ther 53 :529–535.
- 157↑
Saiag P, Capesius C, Roujeau JC, 1988. Abnormal pharmacokinetics of the culprit drug in toxic epidermal necrolysis. J Invest Dermatol 91 :402.
- 159↑
The Cochrane Collaboration, 2003. Review Manager (RevMan) [Computer program]. Oxford, United Kingdom.