Skip to main content

Intermittent preventive treatment for malaria in pregnancy in Africa: What's new, what's needed?

Abstract

Falciparum malaria is an important cause of maternal, perinatal and neonatal morbidity in high transmission settings in Sub-Saharan Africa. Intermittent preventive treatment with sulphadoxine-pyrimethamine (SP-IPT) has proven efficacious in reducing the burden of pregnancy-associated malaria but increasing levels of parasite resistance mean that the benefits of national SP-IPT programmes may soon be seriously undermined in much of the region. Hence, there is an urgent need to develop alternative drug regimens for IPT in pregnancy. This paper reviews published safety and efficacy data on various antimalarials and proposes several candidate combination regimens for assessment in phase II/III clinical trials.

Background

Falciparum malaria is an important cause of maternal anaemia, intra-uterine growth retardation, intrauterine death, stillbirth, premature delivery, low birth weight (LBW), perinatal and neonatal morbidity and mortality [1–5] and postpartum morbidity [6–8]. In sub-Saharan Africa, poor nutrition, micronutrient imbalances (particularly vitamin A, zinc, iron and folate)[1], HIV co-infection [9–12], poverty and limited access to effective primary health care and emergency obstetric services [13–15] exacerbate the impact of pregnancy-associated malaria.

In areas of high or moderate transmission, most malaria infections in pregnant women are asymptomatic and infected women do not present for treatment. In such areas, the World Health Organization recommends a combination of interventions to prevent malaria in pregnancy including insecticide-treated bednets (ITNs), intermittent preventive treatment in pregnancy (IPTp) and effective case management and treatment[16, 17]. A small number of randomized controlled trials and prospective studies conducted in Kenya[18, 19] and Malawi[20, 21] in the 1990s demonstrated the efficacy, safety and cost-effectiveness[22] of sulphadoxine-pyrimethamine (SP) IPTp in preventing maternal anaemia and LBW. The results of these studies led to a recommendation by the World Health Organization that, in areas of medium or high malaria transmission, IPTp with SP should be given on at least two occasions following quickening[17]. Many countries in sub-Saharan Africa have subsequently introduced SP-IPTp into national malaria control programmes [23–25], but levels of coverage are still only modest in most. Although strenuous efforts are being made to increase coverage levels with SP IPTp, its effectiveness is being threatened by increasing levels of resistance to SP across Africa[23, 26–32] and in SE Asia[33, 34]. Thus, some authors have suggested that SP should be combined with artemisinins or with cheaper and more readily available alternatives, such as chloroquine or amodiaquine[35] to maintain the effectiveness of SP-IPTp or that other drug regimens should be used[36]. Several antimalarials, notably chloroquine, proguanil, mefloquine and proguanil-atovaquone, have been evaluated for malaria chemoprophylaxis in pregnancy[4, 37–43], but few clinical trials have attempted to evaluate alternatives to SP in IPTp regimens. Three IPTp clinical trials are currently underway in Benin, Malawi and Tanzania [44–46] and will evaluate SP vs mefloquine[44]; SP alone vs SP plus artesunate[45]; and SP alone vs SP plus azithromycin[46] respectively. A phase III clinical trial among 900 pregnant women in Ghana concluded that amodiaquine alone or in combination with SP was effective in treating uncomplicated falciparum malaria[47], but concerns about the safety and tolerability of amodiaquine in pregnancy[48, 49] and widespread resistance, particularly in East Africa[23], are likely to hinder development of amodiaquine-containing combinations for IPTp.

The future of SP for intermittent preventive treatment of malaria in pregnancy is tenuous, particularly in East and Southern Africa, and there are insufficient, reliable data on the safety and efficacy of alternative antimalarials for the prevention and treatment of malaria in pregnancy[50]. Hence, there is an urgent need to evaluate new drug combinations, particularly in areas where multi-drug resistant falciparum malaria is common[17]. This paper summarizes the current literature on this topic and suggests which antimalarial combinations should be considered for future phase II/III clinical trials among pregnant women in Africa. Key factors reviewed include evidence of safety, acceptability and favourable pharmacokinetic profile in pregnancy; the feasibility of producing fixed-dose co-formulations at costs affordable to most malaria-endemic countries; and the possibility of simple dosing regimens [51–54]. The value of combining drugs with differing molecular targets, those likely to show synergy in combination and those having comparable or disparate elimination half-lives are also discussed[55].

Methods

Search strategy

A two-stage, systematic search was carried out in PubMed and EMBASE using combinations of the keywords: pregnancy, malaria, antenatal, prophylaxis, intermittent, prevention, clinical trial. Alternative spellings and truncated forms were also used[56]. Articles published between 1985 and 2006 were included. Relevant references cited in review articles were also obtained. Full text articles were accessed through the WHO HINARI system designed for research institutions in developing counties[57]. Following a review of the initial search results, several antimalarial drugs were excluded from further review due to their established toxicity during pregnancy (e.g. tetracycline, doxycycline), theoretical contra-indications (e.g. primaquine, halofantrine)[54, 58] or widespread resistance in the region (chloroquine, amodiaquine)[23]. The following drugs were included in a second literature search (conducted in combination with the keywords above): mefloquine; azithromycin; proguanil; chlorproguanil; dapsone; atovaquone; artesunate; artemether; dihydroartemisinin; lumefantrine; piperaquine. Trade names (e.g. Artekin©, Malorone©, Coartem©, Lapdap©, truncated forms and associated terms (e.g. macrolide, artemisinin, arte*) were used where it was felt appropriate. Manufacturers' information sheets, guidelines and reports from the World Health Organization (WHO), Medicines for Malaria Venture (MMV), Gates Malaria Partnership (GMP) and other leading organizations in the field were also scrutinized.

Results

Mefloquine

Mefloquine hydrochloride is a 4-quinolinemethanol derivative with a chemical structure related to that of quinine. In healthy, non-pregnant human volunteers, peak plasma concentrations of around 250 μg/L are observed 6–24 h following a single 250 mg oral administration[59]. Mefloquine has a mean elimination half-life of 2–4 weeks in healthy adults and is excreted in the bile following hepatic clearance from plasma. It can be detected in low concentrations in human breast milk following a single 250 mg dose[59]. In animal toxicity studies, mefloquine was teratogenic in mice and rats at a dose of 100 mg/kg/day and in rabbits at 80 mg/kg/day. At high doses (160 mg/kg/day), mefloquine was also embryotoxic in rabbits[59].

Mefloquine has been used alone for prophylaxis in the pre-conception period and in all trimesters of pregnancy[4, 37–39, 41, 60] and used for the treatment of chloroquine and multidrug-resistant falciparum malaria in pregnancy, both as monotherapy[4, 61–63] and in combination with other antimalarials, particularly artemisinins [64–66].

Mefloquine, at standard prophylactic doses of 250 mg/week appears safe and effective when taken before conception and during all trimesters of pregnancy[37, 39, 41]. In a prospective cohort of 236 pregnant travellers who took a variety of prophylactic regimens, the risk of adverse maternal and foetal outcomes among those exposed to mefloquine in the first trimester was no greater than that observed in women who took other antimalarials (chloroquine plus proguanil or sulphadoxine-pyrimethamine) or background population estimates[37]. Post-marketing surveillance by Roche Pharmaceuticals based on 1,627 reports received between 1985 and 1996 found that the risk of congenital abnormalities among women who took mefloquine for prophylaxis either before conception or at any point during pregnancy was around 4%, which was no higher than reported background population rates of around 2.5%[39]. Risk of miscarriage was unrelated to time of first exposure. Mefloquine prophylaxis 250 mg/week in the second and third trimester was also found to be safe and effective in a randomized placebo-controlled trial conducted among 339 women in Thailand[41]. Transient dizziness followed an initial 10 mg/kg loading dose, but no serious maternal adverse events, spontaneous abortions, stillbirths or neonatal deaths were reported.

However, an observational, non-comparative study among seventy-two, female US soldiers stationed in Somalia reported a risk of spontaneous abortion of 16.7% (12/72) among women who took a standard dose mefloquine prophylaxis in the first trimester[60]. One molar pregnancy was reported and 17 elective terminations carried out. No developmental abnormalities were seen among the 13 infants who were followed up for 1 year. Given the context in which the study was conducted, the small sample size and non-comparative study design, the high rate of miscarriage reported should be interpreted with caution.

There is good evidence that mefloquine is safe and effective in the treatment of falciparum malaria in pregnancy. A low dose mefloquine treatment regimen (12.5 mg/kg as single oral dose) given to 33 women in the second or third trimester in Zaria, Nigeria was well tolerated, safe and effective with no adverse maternal or foetal outcomes reported[61]. In a large prospective study in Malawi, 4,187 women who presented to an antenatal clinic with parasitaemia in the second or third trimester were randomly assigned to 1 of 4 treatment and/or prophylaxis regimens containing chloroquine or mefloquine[4]. Around 60% of women in all groups experienced minor transient symptoms such as itching or dizziness but no serious maternal adverse events were reported. The risk of spontaneous abortion was 1.2% and that of stillbirth 3.9%. No neonatal deaths or congenital abnormalities were reported and no group difference in adverse foetal outcome was observed. Mefloquine in a total treatment dose of 25 mg/kg was also safe and effective for the treatment of chloroquine-resistant falciparum malaria in 40 women enrolled in the second or third trimester in a prospective, non-comparative study conducted in Eastern Sudan[63]. Minor adverse events only were reported (e.g. 18% experienced itching and 35% nausea).

In contrast, a retrospective study in Thailand reported a borderline statistical association between mefloquine treatment during the second and third trimester of pregnancy and excess odds of stillbirth compared to women treated either with quinine or other antimalarials[62]. Among 208 women treated with 25 mg/kg mefloquine as a single treatment dose during the second and third trimester there was an excess of stillbirth (4.5%) compared to 909 women treated either with quinine (1.6%) or other antimalarials (1.4%). The stillbirth rate among women who did not take antimalarials during their pregnancy (n = 2470) was similar to that of the quinine group (1.8%). There was no group difference in the odds of spontaneous abortion or congenital abnormalities. Of the 61 stillbirths that occurred, 38 could be verified in detail including all nine cases associated with mefloquine exposure. Of these, four stillbirths resulted from intrapartum complications (prolonged labour, dystocia, eclampsia, cord prolapse), three were associated with placenta praevia and two cases were unclear ('accidental'; 'fever'). Among women exposed to all other antimalarials, 3/5 cases were of unknown cause as were 2/21 cases that occurred in women who were not exposed to antimalarials during pregnancy. After adjustment for confounders such as maternal age, gravidity, malaria attack rates and trimester of exposure, mefloquine was not significantly associated with increased odds of stillbirth if taken in the three-month prior to conception. In addition, the odds of stillbirth following mefloquine exposure in any trimester of pregnancy was not significantly different to that associated with quinine. The retrospective design, ability to trace only 72% (3,587/5,012) of women attending antenatal clinic in the observation period and biological implausibility of the associations observed suggest that these findings should be viewed within the context of other reported research, particularly clinical trials of mefloquine chemoprophylaxis, which indicate that mefloquine is safe in pregnancy.

Several studies were identified which looked at the safety and efficacy of mefloquine in combination with other antimalarials. All were conducted among women in the second or third trimester. A randomized, controlled trial with 40 subjects in Nigeria compared artemether (3.2 mg/kg intramuscularly once at entry) plus low dose mefloquine (7.5 mg/kg orally at 24, 48 h) versus artemether alone (3.2 mg/kg at entry followed by 1.6 mg/kg/day for 4 days) for the treatment of multidrug-resistant falciparum malaria[64]. No serious adverse maternal or foetal outcomes were reported. The artemether-mefloquine combination was acceptable, safe and efficacious. In another small study in Thailand, 60 women were enrolled during the second or third trimester and treated with artesunate (2 mg/kg once then 1 mg/kg twice daily for 5 days) plus mefloquine (15 mg/kg once then 10 mg/kg 6 h later) or quinine alone (10 mg/kg/day for 7 days)[67]. Fever and parasite clearance times were significantly shorter in the artesunate-mefloquine group, and fewer minor adverse events reported. No serious adverse events were observed.

A randomized, open-label clinical trial among 108 second and third trimester pregnant women in northwest Thailand compared a three-day mefloquine-artesunate regimen (MAS3; mefloquine 15 mg/kg on day 1; 10 mg/kg on day 2 plus artesunate 4 mg/kg/day on days 0, 1, 2) versus seven days treatment with quinine (Q7; 10 mg/kg three times daily for 7 days) for multidrug-resistant falciparum malaria[66]. A number of serious adverse events were reported: one maternal death, unrelated to malaria (it is not stated in which arm); two mid-trimester abortions (both in the MAS3 arm vs. none in the Q7 arm); and five neonatal deaths (three in the MAS3 arm vs. two in the Q7 arm). There were no stillbirths and no significant differences between arms in mean birthweight, proportion of LBW infants or developmental milestones at 12 months. Minor adverse events such as dizziness (87% vs. 45%; RR 1.93; 95% CI 1.14, 3.25) and tinnitus (66% vs. 17%; RR 3.93; 95% CI 1.98, 7.80) were more common in the Q7 than MAS3 group. MAS3 was more effective and better tolerated than Q7, and was associated with significantly reduced gametocyte carriage time (2.3 vs. 46.9/1000 person weeks respectively; p < 0.001). PCR adjusted recrudescence rates at 63 days were 33% in the Q7 group compared to 2% in the MAS3 group (p = 0.001).

A phase III equivalence trial comparing intermittent SP and mefloquine alone for the prevention of malaria-related morbidity in pregnancy is currently underway in Benin[44].

Azithromycin

Azithromycin is a macrolide antibiotic derived from erythromycin which has an elimination half-life of 68 h in healthy human volunteers and is excreted largely unchanged in the bile following hepatic clearance[68]. In pregnancy, the placenta appears to act as an effective barrier to foetal exposure with limited placental transfer of macrolide antibiotics observed following maternal administration[69]. In a prospective study among 20 third trimester pregnant women, undertaken to assess maternal and transplacental pharmacokinetics of azithromycin, peak maternal serum azithromycin levels occurred within 6 h of a single 1 g oral dose and declined rapidly over the next 24 h[70]. Myometrial, adipose and placental azithromycin levels were sustained for up to 72 h post administration.

Azithromycin has been used extensively to treat sexually-transmitted and other bacterial infections during pregnancy in total treatment doses of up to 1–2 g and is considered a safe alternative for the treatment of chlamydia, mycoplasma and group B streptococci in pregnant women who are allergic to beta-lactam antibiotics [71–73].

Azithromycin is not yet licensed for use as an antimalarial agent but has shown promising activity against P. falciparum in vitro[74, 75], in the murine malaria model[76] and in randomized controlled clinical trials [77–79]. A phase III randomized placebo-controlled clinical trial of azithromycin prophylaxis (750 mg loading dose followed by 250 mg/day for 20 weeks) versus doxycycline (100 mg/day for 20 weeks) conducted among 300 non-pregnant Indonesian adults (286/300 male) found that azithromycin was safe and well tolerated when used alone for the prevention of falciparum malaria in Northeast Papua[77]. No serious adverse events were reported. One subject developed an erythematous maculopapular rash that was probably related to azithromycin exposure and which necessitated withdrawal from the study. Prophylactic efficacy was 72% which suggests that azithromycin should be used in combination with other antimalarials rather than used alone as a first-line agent for prevention. A clinical trial among non-pregnant adults in Kenya reported similar levels of tolerability, safety and efficacy[78]. The efficacy of azithromycin in combination with other antimalarials for the treatment of uncomplicated falciparum malaria was assessed in a randomized controlled trial among 202 non-pregnant adults in Thailand[79]. A combination of artesunate 200 mg once followed by azithromycin 500 mg daily for 3 days was more effective than artesunate alone given daily for 3 days but was less effective than an artesunate plus mefloquine regimen given over 3 days. Cure rates at 28 days in the three groups were 56%, 44% and 98% respectively. No serious adverse events were reported in any of the treatment groups.

To date, no clinical trials have been conducted among pregnant women but a phase III clinical trial of intermittent SP plus azithromycin in pregnancy versus intermittent SP alone is currently underway in Malawi[46].

Proguanil

The elimination half-life of proguanil is 12 – 21 h in healthy adult volunteers and around 40–60% of the drug is eliminated by urinary excretion. Proguanil is metabolized by a cytochrome P450 isoenzyme to an active metabolite (cycloguanil), responsible for antimalarial activity[80]. Late pregnancy and use of oral contraceptives impair formation of the active metabolite, probably due to inhibition of the metabolizing isoenzyme as a result of elevated oestrogen levels[80]. The pharmacokinetics of proguanil and cycloguanil are significantly altered in pregnant women with malaria compared to healthy volunteers and children with uncomplicated falciparum malaria[81]

Proguanil has been used extensively for malaria chemoprophylaxis in travellers, including pregnant women and children and is considered one of the safest antimalarials currently available[37, 42, 58, 82]. A randomized controlled clinical trial in 200 antenatal clinic attenders carried out in Nigeria concluded that proguanil given in a daily dose of 100 mg daily was safe, well tolerated and effective in all trimesters of pregnancy[42]. Taken alone or in combination with chloroquine, proguanil chemoprophylaxis was similarly well tolerated and effective in a randomized prospective study among 327 antenatal clinic attenders in Muheza, Tanzania[82] and in a prospective cohort of 236 first trimester pregnant travellers[37].

Proguanil-atovaquone

Proguanil has also been used in fixed combination with atovaquone (100 mg/250 mg respectively) in the commercially available preparation Malarone© (GlaxoSmithKline). Atovaquone has an elimination half life of 2–3 days in adult volunteers and around 95% is excreted unchanged in the bile[83]. A randomized double-blind study compared mefloquine alone and proguanil-atovaquone for chemoprophylaxis in 976 non-pregnant travelers and found that both regimens were similarly effective but that the proguanil-atovaquone combination was better tolerated[84]. In an open-label randomized controlled treatment trial among 182 non-pregnant adults with multidrug-resistant falciparum malaria in Thailand, proguanil-atovaquone was more effective and better tolerated than mefloquine[85]. In a small safety and efficacy study among 26 third trimester pregnant women conducted in Thailand and Zambia, subjects presenting with acute uncomplicated falciparum malaria were given proguanil-atovaquone 100 mg/250 mg daily for 3 days[81]. Median fever clearance time was 56 h in Thailand and 52 h in Zambia; median parasite clearance times were 51 h and 24 h respectively. The 28-day cure rate was 100% at both sites. No serious maternal or foetal adverse outcomes were reported. Minor adverse events included headache, nausea and vomiting but occurred in less than 10% and were probably due to the underlying infection.

A randomized open-label clinical trial among 81 second and third trimester pregnant women in Thailand compared quinine and artesunate-atovaquone-proguanil (AAP) for the treatment of uncomplicated falciparum malaria and found that women treated with AAP had shorter fever and parasite clearance times and lower treatment failure rates (5% vs. 37%) compared to those in the quinine group[86]. The AAP combination also appeared safe and was well tolerated compared to quinine.

Chlorproguanil-dapsone

Chlorproguanil (Lapudrine®) has a similar mode of action and pharmacokinetic profile to proguanil but has a slightly longer elimination half-life (around 32 h)[80]. The drug has primarily been used in fixed dose co-formulation with dapsone (diaminodiphenylsulfone), available commercially as Lapdap© (GlaxoSmithKline). Despite evidence from a number of clinical trials of the safety and efficacy of this combination in the treatment of uncomplicated malaria in children[29, 87, 88] and pregnant women[89], a recent WHO report[80] concluded that the combination should continue to be used with caution, particularly in areas where glucose-6-phosphate dehydrogenase (G6PD) deficiency is common. Treatment trials of chlorproguanil-dapsone in pregnancy are currently underway in Tanzania[90] and in Mali[91]. A combination of chlorproguanil-dapsone plus artesunate (CDA) is also under development[92] and a multicentre phase III clinical trial comparing the efficacy and safety of CDA and artemether-lumefantrine in the treatment of uncomplicated falciparum malaria in children and adolescents is about to start in four countries in East and West Africa[93].

Artemisinins

Artemisinin compounds (artemether, artesunate, dihydroartemisinin) are derived from sweet wormwood (Artemisia annua), a herb which has been used in traditional Chinese medicine for over 2000 years[55, 94]. All are potent rapid-acting blood schizonticides with elimination half-lives from around 40 minutes to several hours[51].

All artemisinin analogues are associated with embryotoxicity over a narrow dose range in the rat and rabbit where embryo-lethality, late resorption and morphological abnormalities (such as abnormal development of the cardiovascular system, axial skeleton and limbs) have been reported[94, 95]. There is also some evidence from animal models that artemisinins given later in pregnancy have adverse effects on foetal body weight and survival[95]. The mechanism of developmental toxicity in animals is unclear. In rats, the yolk sac is highly susceptible to artemisinin compounds[96], but recent studies in monkeys, which showed high rates of foetal loss, indicate that other mechanisms must be involved in higher mammals[97]. Published data from clinical trials in humans is very limited. Data from a total of 864 women exposed to artemisinin compounds during the second and third trimester of pregnancy have been reported to date with no evidence of drug-related adverse maternal or foetal outcome[64, 66, 67, 86, 98–100]. Published data on safety in the first trimester is limited to 50 women in The Gambia[99] and 108 in Thailand[49, 100] with similar findings. None of the studies conducted to date have been specifically designed to capture excess early pregnancy loss, the key adverse pregnancy outcome that might be expected based on animal toxicity data.

Following two expert consultations in 2002, WHO recommended that artemisinins should not be used in the first trimester except in life-threatening situations where other antimalarials were not suitable, should be used with caution in the second and third trimesters and that further research be conducted to establish the pharmacokinetics of artemisinins during pregnancy, mechanism of toxicity and the relevance of animal toxicity data to humans[95]. Pharmacovigilance including post-exposure surveillance and pregnancy registers were also recommended. The pharmacokinetics of dihydroartemisinin following oral artesunate treatment was recently described in 24 second and third trimester pregnant women with uncomplicated falciparum malaria in Thailand[101].

Artemether-lumefantrine

The only fixed-dose artemisinin combination therapy (ACT) currently available is artemether-lumefantrine (Coartem©/Riamet©), which is available at subsidized cost to malaria-endemic countries following a special pricing agreement established in 2001 between WHO and the manufacturer, Novartis[52]. Artemether-lumefantrine is now first-line treatment for uncomplicated falciparum malaria in adults and children in 19 countries in sub-Saharan Africa and 56 countries worldwide[52]. No treatment or prevention trials in pregnancy have to date been published.

Dihydroartemisinin-piperaquine

Dihydroartemisinin is a derivative of artemisinin and the principal active metabolite of artesunate and artemether with an elimination half-life of 40–60 minutes[51]. Piperaquine is a 4-aminoquinoline structurally related to chloroquine, originally developed in China in the 1960's[102] and has an elimination half-life of 3 to 4 weeks[51]. Following the widespread introduction of piperaquine as monotherapy in China during the 1970's (including mass prophylaxis programmes in some provinces), P. falciparum piperaquine-resistance emerged in the mid-1990's, when efforts were then made to formulate the drug in various combinations e.g. with trimethoprim, primaquine and others[103]. Dihydroartemisinin was selected as the best partner and a fixed dose co-formulation dihyroartemisinin-piperaquine (DP; Artekin©/Eurartekin©) was developed through an international public-private partnership between Holleykin Pharmaceutical, China; Sigma-Tau Industries, Italy; the University of Oxford, UK; and the Medicines for Malaria Venture (MMV)[104]. DP has been used extensively in China, Viet Nam, Cambodia and other parts of SE Asia and shown to be safe, effective and well tolerated in the treatment of uncomplicated, multidrug-resistant falciparum malaria in children and adults[103, 105–107]. Phase II/III clinical trials of DP for intermittent preventive treatment in infants (IPTi) are already underway in SE Asia and Africa[92] and the combination may soon become more widely available once international drug licensing and registration procedures have been completed. To date however, no clinical trials have been conducted among pregnant women.

Other artemisinin combination therapies

A variety of new ACTs are currently being evaluated in phase II and III clinical trials in children and non-pregnant adults. Amodiaquine plus artesunate is being developed in fixed dose co-formulation as Coarsucam© (Sanofi-Aventis). Clinical trials in Ghana[108] and Tanzania[109] have reported encouraging results with this combination in children under 5 years and a multicentre treatment trial in adults and children is about to start in Cameroon, Madagascar, Mali and Senegal.[110]. Given uncertainty about the safety of amodiaquine in pregnancy[48, 49], it is unlikely that this combination will enter clinical IPTp trials in the near future. The combination may, however, be a promising candidate for evaluation in clinical trials for IPTi, particularly in West Africa where amodiaquine-SP has shown encouraging results [111–113].

The efficacy of dihydroartemisinin-mefloquine (DHA-MQ) compared to DHA alone and chloroquine alone for the treatment of uncomplicated falciparum malaria was recently described in a small study among 75 children aged 2–13 years conducted in Nigeria[114] but no published data on the use of this combination in pregnancy, evidence of clinical trials in progress or plans to develop a fixed dose formulation were identified during this review.

Pyronaridine, a Mannich-base schizontocide structurally related to the aminoacridine drug quinacrine, which exerts its antimalarial action by forming complexes with haematin[115], is being developed in fixed dose co-formulation with artesunate (PANDA)[92]. A phase II clinical trial is currently ongoing in children and adolescents in Gabon[116]. Pyronaridine has also shown promising activity in combination with dihydroartemisinin[117].

Chlorproguanil-dapsone-artesunate (CDA), artesunate-atovaquone-proguanil (AAP) and artesunate-SP are other promising ACTs under development, as described above.

Discussion

The pharmacokinetics, dosing regimen, efficacy and safety profile of antimalarials currently used to treat and prevent falciparum malaria in adults and children are well-described. This is not the case in pregnancy where the limited data available in each of these areas makes it difficult to predict which drugs are likely to be the best candidates for treating and preventing infection. Many antimalarials are contra-indicated in pregnancy (e.g. primaquine, doxycycline, halofantrine, tafenoquine), whilst the safety of several promising candidates remains unclear[58]. In malaria-endemic countries, where antenatal HIV prevalence is high, the increased risk of serious adverse events associated with some antimalarials (e.g. severe skin reactions with SP in women with co-infection[54]) and drug interactions with anti-retrovirals used for the prevention of mother to child HIV transmission (e.g. SP interacts with nevirapine and zidovudine[118]) are also important factors to consider. Malaria in pregnancy is unique and characterized by placental sequestration of P. falciparum-infected erythrocytes following their adherence to placental chondroitin sulphate A (CSA)[119] and hyaluronic acid[120], which essentially allows the placenta to act as a 'privileged site' for parasite replication[36]. Pregnant women in all transmission areas have an increased susceptibility to malaria and other infections due to changes in cellular mediated immune responses [121–123], which persist into the postpartum period[6, 8]. Physiological changes such as increased intravascular volume, delayed gastric emptying time, elevated oestrogen and cortisol levels and increased body fat content alter the absorption, distribution and elimination of many antimalarials during pregnancy[81]. For example the pharmacokinetics of atovaquone, proguanil, cycloguanil and dihydroartemisnin are significantly altered in pregnant women with malaria compared to healthy volunteers or adults and children with malaria[81, 101]. For candidate IPTp combinations there is the added complication that there is still some debate as to how IPTp actually works: repeated short courses of antimalarials may act mainly by intermittent suppressive chemotherapy ('prophylactic effect'); by intermittent treatment of repeated infections ('treatment effect'); or by some combination of these effects[36, 55]. Similar issues are also being debated about IPTi [124]. Given the early success of SP-IPTp in high transmission areas, the slow parasite clearance times and long elimination half-lives of each of the component drugs[125], it seems likely that the 'prophylactic effect' is the predominant mode of action, at least in this combination.

How then to decide which antimalarials are likely to be effective for IPT in pregnancy and which combinations should be prioritized for investigation in clinical trials (Table 1)? If the effect of IPTp is mainly prophylactic, then short-acting drugs such as quinine and the artemesinins would be expected to provide little direct benefit in asymptomatic pregnant women living in high-transmission areas since rapid parasite elimination is unnecessary. Drugs with long half-lives such as mefloquine, piperaquine or lumefantrine would, therefore, appear better choices, but monotherapy with any of these agents would be inappropriate given global patterns of drug resistance and the consensus on the need for combination therapies [51–54]. There is some evidence that combining short-acting artemisinins with longer-acting agents is likely to delay the emergence of resistance to the slowly-eliminated component[51, 53, 55]. Conversely, some authors advise combining drugs with similar elimination half-lives due to the risk of resistance emerging to the longer-acting drug because of it's persistence alone at sub-therapeutic levels once the rapidly eliminated drug has been excreted[54]. Combining drugs that have different molecular targets is also preferable to reduce the risk of resistance[51, 54]. Acceptability, cost, possibility of a fixed-dose formulation, and a simple dosing regimen are other important considerations[53].

Table 1 Pharmacology of selected candidates for IPTp

Based on these criteria, dihydroartemisinin-piperaquine appears an appropriate candidate for IPTp and should be considered for phase II/III clinical trials in second and third trimester pregnant women. The combination is relatively inexpensive (around US$ 1.00 for an adult treatment course, which is comparable to Coartem©)[126]; combines a very short acting artemisinin with a longer acting partner with a different mode of action; and is available in fixed-dose co-formulation. Dihydroartemisinin is the active metabolite of many artemisinin compounds, which appear to be effective, well tolerated and safe after the first trimester; the evidence for the safety of piperaquine is, however, less clear, indicating the need for preliminary phase II studies.

Artemether-lumefantrine may also be a suitable candidate: a relatively inexpensive, fixed co-formulation is available and the partner drugs have different molecular targets. The 'prophylactic effect' conferred by lumefantrine may not be sufficient however, given its relatively short half-life compared to sulphadoxine, pyrimethamine, mefloquine or piperaquine.

Mefloquine in combination with artemether or artesunate has the advantage of a similar elimination profile to dihydroartemisnin-piperaquine and the safety of such combinations in the treatment of pregnancy-associated malaria is reasonably well-established. Indeed, the extensive data available on mefloquine use in pregnancy compared to piperaquine suggest that dihydroartemisinin-mefloquine could enter phase III trials ahead of dihydroartemisinin-piperaquine. Cost and the lack of fixed dose co-formulations (although these are currently under development) are disadvantages. Combining mefloquine with the relatively long acting azithromycin is another option and likely to have additional health benefits by reducing maternal and perinatal infections, which may offset cost concerns. Sexually-transmitted infections (STIs) are common in women of childbearing age in Africa and may go undiagnosed and untreated during pregnancy[127]. Monthly azithromycin reduces STI incidence significantly in high-risk groups[128] and is safe and effective for treating gonorrhoea as well as chlamydia infection during pregnancy[71], which is associated with increased risk of stillbirth, prematurity and LBW[129]. Mefloquine-azithromycin may, therefore, represent a promising, safe and effective combination for IPTp and should be considered urgently for evaluation in phase III clinical trials.

Patterns of SP resistance vary across sub-Saharan Africa so that new artemisinin-containing combinations with SP may be appropriate in West Africa, given that these would fulfil many of the criteria above. Such combinations are, however, unlikely to succeed in East and Southern Africa, given prevailing levels of resistance.

Are different types of drug combination needed in different areas of malaria transmission? Evidence for the effectiveness of IPTp comes primarily from studies conducted in areas of intense perennial transmission (stable malaria), but there may be a case for evaluating IPTp in areas of low transmission (unstable malaria). The World Health Organization recommends that IPTp be an integral part of national malaria control programmes in high/medium perennial or seasonal transmission areas but caution that there is insufficient evidence for introducing IPTp in low transmission settings where the principal focus should be on effective case management and insecticide-treated bednets[17]. In areas of highly seasonal transmission there is some evidence to suggest that IPT given only during periods of high transmission may be effective, at least in infants[112], but no studies to date have been reported from low transmission settings such as South America or Asia, where the burden of pregnancy-associated malaria is significant, but quite different from that observed in areas of stable malaria and where the contribution of Plasmodium vivax infection also needs to be taken into consideration[36]. In addition, combinations found to be efficacious in sub-Saharan Africa may be of limited benefit in Asia due to their inability to prevent relapse in P. vivax infection[130, 131].

Conflict of interests

The author(s) declare that they have no competing interests.

References

  1. Breman JG, Alilio MS, Mills A: Conquering the intolerable burden of malaria: what's new, what's needed: a summary. Am J Trop Med Hyg. 2004, 7(Suppl 2): 1-15.

    Google Scholar 

  2. Schellenberg D, Armstrong-Schellenberg JRM, Mushi A, de Savigny D, Mgalula L, Mbuya C, Victora CG: The silent burden of anaemia in Tanzanian children: a community based study. Bull World Health Organ. 2003, 81 (8): 581-590.

    PubMed Central  CAS  PubMed  Google Scholar 

  3. Shulman CE, Graham WJ, Jilo H, Lowe BS, New L, Obiero J, Snow RW, Marsh K: Malaria as an important cause of anemia in primigravidae: evidence from a district hospital in coastal Kenya. Trans R Soc Trop Med Hyg. 1996, 90: 535-539. 10.1016/S0035-9203(96)90312-0.

    CAS  PubMed  Google Scholar 

  4. Steketee RW, Wirima JJ, Slutsker L, Khoromana CO, Heymann DL, Breman JG: Malaria treatment and prevention in pregnancy: indications for use and adverse events associated with use of chloroquine or mefloquine . Am J Trop Med Hyg. 1996 , 55 (1 Suppl): 50-56.

    CAS  PubMed  Google Scholar 

  5. Marchant T, Armstrong-Schellenberg J, Nathan R, Abdulla S, Mukasa O, Mshinda H, Lengeler C: Anaemia in pregnancy and infant mortality in Tanzania. Trop Med Int Health. 2004, 9 (2): 262-266. 10.1046/j.1365-3156.2003.01178.x.

    PubMed  Google Scholar 

  6. Diagne N, Rogier C, Sokhna CS, Tall A, Fontenille D, Roussilhon C, Spiegel A, Trape JF: Increased susceptibility to malaria during the early post-partum period. N Eng J Med. 2000 , 343 (9): 598-603. 10.1056/NEJM200008313430901.

    CAS  Google Scholar 

  7. Vallely L, Ahmed Y, Murray SF: Postpartum maternal morbidity requiring hospital admission in Lusaka, Zambia-a descriptive study. ,. BMC Pregnancy Childbirth. 2005, 5 (1):

  8. Ramharter M, Grobusch MP, Kiessling G, Adegnika AA, Moller U, Agnandji ST, Kramer M, Schwarz N, Kun JF, Oyakhirome S, Issifou S, Borrmann S, Lell B, Mordmuller B, Kremsner PG: Clinical and parasitological characteristics of puerperal malaria. J Inf Dis. 2005, 191 (6): 1005-1009. 10.1086/427781.

    Google Scholar 

  9. Ayisi JG, van Eijk AM, ter Kuile FO, Kolczak MS, Otieno JA, Misore AO, Kager PA, Steketee RW, Nahlen BL: The effect of dual infection with HIV and malaria on pregnancy outcome in western Kenya. AIDS. 2003, 17 (4): 585-594. 10.1097/00002030-200303070-00014.

    PubMed  Google Scholar 

  10. Ladner J, Leroy V, Simonon A, Karita E, Bogearts J, De Clercq A, Van De Perre P, Dabis F, and the Pregnancy and HIV Study Group: HIV infection, malaria and pregnancy: a prospective cohort study in Kigali, Rwanda. Am J Trop Med Hyg. 2002, 66 (1): 56-60.

    PubMed  Google Scholar 

  11. Steketee RW, Wirima JJ, Bloland PB, Chilima B, Mermin JH, Chitsulo L, Breman JG: 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. 1996, 55 (1 Suppl): 42-49.

    CAS  PubMed  Google Scholar 

  12. Brahmbhatta H, Kigozib G, Wabwire-Mangenc F, Serwadda D, Sewankamboc N, Lutalob T, Wawerd MJ, Abramowskye C, Sullivana D, Graya R: The effects of placental malaria on mother-to-child HIV transmission in Rakai, Uganda. AIDS. 2003, 17: 2539–2541-

    Google Scholar 

  13. Adam T, Lim SS, Mehta S, Buhta ZA, Fogstad H, Mathai M, Zupan J, Darmstadt GL: Cost effectiveness analysis of strategies for maternal and neonatal health in developing countries. BMJ. 2005, 331: 1107-1110. 10.1136/bmj.331.7525.1107.

    PubMed Central  PubMed  Google Scholar 

  14. Evans DB, Adam T, Edejer T, Lim SS, Cassels A, Evans TG: Time to reassess strategies for improving health in developing countries. BMJ. 2005, 331: 1133-1136. 10.1136/bmj.331.7525.1133.

    PubMed Central  PubMed  Google Scholar 

  15. Sanders DM, Todd C, Chopra M: Confronting Africa's health crisis: more of the samewill not be enough. BMJ. 2005, 331: 755-758. 10.1136/bmj.331.7519.755.

    PubMed Central  CAS  PubMed  Google Scholar 

  16. Nahlen BL: Rolling Back Malaria. N Eng J Med. 2000, 343: 651-652. 10.1056/NEJM200008313430909.

    CAS  Google Scholar 

  17. WHO: A Strategic Framework for Malaria Prevention and Control during Pregnancy in the Africa Region. 2004, World Health Organization Regional Office for Africa

    Google Scholar 

  18. Parise ME, Ayisi JG, Nahlen BL, Schultz LJ, Roberts J, Misore A, Muga R, Oloo AJ, Steketee RW: 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. 1998, 59 (5): 813-822.

    CAS  PubMed  Google Scholar 

  19. Shulman CE, Dorman EK, Cutts F, Kawuondo K, Bulmer JN, Peshu N, Marsh K: Intermittent sulphadoxine-pyrimethamine to prevent severe anaemia secondary to malaria in pregnancy: a randomised placebo-controlled trial. Lancet. 1999, 353: 632-636. 10.1016/S0140-6736(98)07318-8.

    CAS  PubMed  Google Scholar 

  20. Schultz LJ, Steketee RW, Macheso A, Kazembe P, Chitsulo L, Wirima JJ: 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. 1994, 51 (5): 515-522.

    CAS  PubMed  Google Scholar 

  21. Verhoeff FH, Brabin BJ, Chimsuku L, Kazembe P, Russel WB, Broadhead RL: An evaluation of intermittent sulfadoxine-pyrimethamine treatment in pregnancy on parasite clearance and risk of low birth weight in rural Malawi. Ann Trop Med Parasitol. 1998, 92: 141-150. 10.1080/00034989859979.

    CAS  PubMed  Google Scholar 

  22. Schultz LJ, Steketee RW, Chitsulo L, Wirima JJ: Antimalarials during pregnancy: a cost-effectiveness analysis. Bull WHO. 1995, 73 (2): 207-214.

    PubMed Central  CAS  PubMed  Google Scholar 

  23. EANMAT: The efficacy of antimalarial monotherapies, sulphadoxine-pyrimethamine and amodiaquine in East Africa: implications for sub-regional policy. Trop Med Int Health. 2003, 8 (10): 860-867. 10.1046/j.1360-2276.2003.01114.x.

    Google Scholar 

  24. Newman RD, Moran AC, Kayentao K, Benga-De E, Yameogo M, Gaye O, Faye O, Lo Y, Moreira PM, Duombo O, Parise ME, Steketee RW: Prevention of malaria during pregnancy in West Africa: policy change and the power of subregional action. Trop Med Int Health. 2006, 11 (4): 462-469. 10.1111/j.1365-3156.2006.01593.x.

    PubMed  Google Scholar 

  25. Hill J, Kazembe P: Reaching the Abuja target for intermittent preventive treatment of malaria in pregnancy in African women: a review of progress and operational challenges. Trop Med Int Health. 2006, 11 (4): 409-418. 10.1111/j.1365-3156.2006.01585.x.

    PubMed  Google Scholar 

  26. Plowe CV, Kublin JG, Dzinjalamala FK, Kamwendo DS, Mukadam RA, Chimpeni P, Molyneux ME, Taylor TE: Sustained clinical efficacy of sulfadoxine-pyrimethamine for uncomplicated falciparum malaria in Malawi after 10 years as first line treatment: five year prospective study. BMJ. 2004, 328 (6): 545-548. 10.1136/bmj.37977.653750.EE.

    PubMed Central  CAS  PubMed  Google Scholar 

  27. White N: Sulfadoxine-pyrimethamine is not working in Malawi. BMJ. 2004, 328: 1259-10.1136/bmj.328.7450.1259.

    PubMed Central  PubMed  Google Scholar 

  28. Ringwald P: Treatment failure and resistance in Malawi remain subject for debate. BMJ. 2004, 328: 1259-10.1136/bmj.328.7450.1259-a.

    PubMed Central  CAS  PubMed  Google Scholar 

  29. Alloueche A, Bailey W, Barton S, Bwika J, Chimpeni P, Falade COF, Horton J, Jaffar S, Kanyok T, Kremsner PG, Kublin JGL, Missinou MA, Mkandala C, Oduola AMJ, Premji Z, Robertson L, Sowumni A, Ward SA, Winstanley PA: Comparison of chlorproguanil-dapsone with sulfadoxine-pyrimethamine for the treatment of uncomplicated falciparum malaria in young African children: double blind randomized controlled trial. Lancet. 2004, 363: 1843-1848. 10.1016/S0140-6736(04)16350-2.

    CAS  PubMed  Google Scholar 

  30. Jima D, Tesfaye G, Medhin A, Kebede A, Argaw D, Babaniyi O: Efficacy of sulfadoxine-pyrimethamine for the treatment of uncomplicated falciparum malaria in Ethiopia. East Afr Med J. 2005, 82 (8): 391-395.

    CAS  PubMed  Google Scholar 

  31. Zurovac D, Ndhlovu M, Rowe AK, Hamer DH, Thea DM, Snow RW: Treatment of paediatric malaria during a period of drug transition to artemether-lumefantrine in Zambia: cross-sectional study. BMJ. 2005, 331: 734-737. 10.1136/bmj.331.7519.734.

    PubMed Central  CAS  PubMed  Google Scholar 

  32. WHO: Recommendations on the use of Sulfadoxine-Pyrimethamine (SP) for Intermittent Preventive Treatment during Pregnancy (IPT) in areas of moderate to high resistance to SP in the African Region. Edited by: WHO . [http://www.afro.who.int/malaria/publications/who_sp_statement.pdf#search=%22WHO%20AFRO%20Harare%20meeting%20IPT%22]

  33. Campbell P, Baruah S, Narauin K, Rogers CC: A randomised trial comparing the efficacy of four treatment regimens for uncomplicated falciparum malaria in Assam state, India. Trans R Soc Trop Med Hyg. 2006, 100 (2): 108-118. 10.1016/j.trstmh.2005.06.016.

    CAS  PubMed  Google Scholar 

  34. van den Broek IV, Maung UA, Peters A, Liem L, Kamal M, Rahman M, Rahman MR, Bangali AM, Das S, Barends M, Faiz AM: Efficacy of chloroquine + sulfadoxine--pyrimethamine, mefloquine + artesunate and artemether + lumefantrine combination therapies to treat Plasmodium falciparum malaria in the Chittagong Hill Tracts, Bangladesh. Trans R Soc Trop Med Hyg. 2005, 99 (10): 727-735. 10.1016/j.trstmh.2005.02.007.

    CAS  PubMed  Google Scholar 

  35. McIntosh HM, Jones KL: Chloroquine or amodiaquine combined with sulfadoxine-pyrimethamine for treating uncomplicated malaria. Cochrane Database Syst Rev. 2005, 4: CD000386-

    PubMed  Google Scholar 

  36. White NJ: Intermittent Presumptive Treatment for Malaria. PLoS Med. 2005, 2 (1): e3 -10.1371/journal.pmed.0020003.

    PubMed Central  PubMed  Google Scholar 

  37. Phillips-Howard PA, Stefen R, Kerr L, Vanhauwere B, Fuchs E, Edwards R: Safety of mefloquine and other antimalarial agents in the first trimester of pregnancy. J Travel Med. 1998, 5: 121-126. 10.1111/j.1708-8305.1998.tb00484.x.

    CAS  PubMed  Google Scholar 

  38. Schlagenhauf P: Mefloquine for malaria chemoprophylaxis 1992-1998: A review. J Travel Med. 1999, 6 (2): 122-133.

    CAS  PubMed  Google Scholar 

  39. Vanhauwere B, Maradit H, Kerr L: Post-marketing surveillance of prophylactic mefloquine (Larium) use in pregnancy. Am J Trop Med Hyg. 1998, 58 (1): 17-21.

    CAS  PubMed  Google Scholar 

  40. Garner P, Brabin B: A review of randomized controlled trials of routine antimalarial drug prophylaxis during pregnancy in endemic malarious areas. Bull World Health Organ. 1994, 72 (1): 89-99.

    PubMed Central  CAS  PubMed  Google Scholar 

  41. Nosten F, ter Kuile F, Maelankiri L, Chongsuphajaisiddhi T: Mefloquine prophylaxis prevents malaria during pregnancy: a double-blind, placebo-controlled study , . J Infect Dis. 1994, 169: 595-603.

    CAS  PubMed  Google Scholar 

  42. Fleming AF, Ghatoura GB, Harrison KA, Briggs ND, Dunn DT: The prevention of anaemia in pregnancy in primigravidae in the guinea savanna of Nigeria. Ann Trop Med Parasitol. 1986, 80 (2): 211-233.

    CAS  PubMed  Google Scholar 

  43. Mutabingwa TK, Malle LN, de Geus A, Oosting J: Malaria chemosuppression in pregnancy. I. The effect of chemosuppressive drugs on maternal parasitaemia.Trop Geogr Med. 1993, 45 (1): 6-14.

    CAS  PubMed  Google Scholar 

  44. National Institutes of Health USA: Intermittent preventive treatment during pregnancy in Benin: a randomized, open, and equivalent trial comparing sulfadoxine-pyrimethamine with mefloquine. 2006, [http://www.clinicaltrials.gov/ct/show/NCT00274235?order=59]

    Google Scholar 

  45. National Institutes of Health USA: Efficacy of intermittent sulfadoxine-pyrimethamine and sulfadoxine-pyrimethamine + artesunate treatment in the prevention of malaria in pregnancy in an area with chloroquine-resistant Plasmodium falciparum. 2006, [http://www.clinicaltrials.gov/ct/show/NCT00164255?order=38]

    Google Scholar 

  46. National Institutes of Health USA: Lungwena Antenatal Intervention Study. A single-centre intervention trial in rural Malawi, testing maternal and infant health effects of presumptive intermittent treatment of pregnant women with sulfadoxine-pyrimethamine and azithromycin. 2006, [http://www.clinicaltrials.gov/ct/show/NCT00131235?order=72]

    Google Scholar 

  47. Tagbor H, Bruce J, Browne E, Randal A, Greenwood B, Chandramohan D: Efficacy, safety, and tolerability of amodiaquine plus sulphadoxine-pyrimethamine used alone or in combination for malaria treatment in pregnancy: a randomised trial. Lancet. 2006, 368 (1349-56):

  48. Thomas F, Erhart A, D'Alessandro U: Can amodiaquine be used safely during pregnancy?. Lancet Inf Dis. 2004, 4 (4): 235-239. 10.1016/S1473-3099(04)00974-0.

    CAS  Google Scholar 

  49. Nosten F, McGready R, d'Alessandro U, Bonell A, Verhoeff F, Menendez C, Mutabingwa T, Brabin B: Antimalarial Drugs in Pregnancy: A Review. Curr Drug Safety. 2006, 1: 1-15.

    CAS  Google Scholar 

  50. Orton L, Garner P: Drugs for treating uncomplicated malaria in pregnant women. Cochrane Database Syst Rev. 2005, 3: CD004912-

    PubMed  Google Scholar 

  51. Davis TME, Karunajeewa HA, Ilett KF: Artemisinin-combination therapies for uncomplicated malaria. Med J Aust. 2005, 184 (4): 181-185.

    Google Scholar 

  52. WHO: Facts on ACTs (Artemisinin-Based Combination Therapies): January 2006 Update. 2006, [http://www.rbm.who.int/cmc_upload/0/000/015/364/RBMInfosheet_9.pdf]

    Google Scholar 

  53. Yeung S, Pongtavornpinyo W, Hastings IM, Mills AJ, White NJ: Antimalarial drug resistance, Artemisinin-based Combination Therapy and the contribution of modeling to elucidating policy choices. Am J Trop Med Hyg. 2004, 71(Suppl 2) : 179-186.

    Google Scholar 

  54. Kremsner PG, Krishna S: Antimalarial combinations. Lancet. 2004, 364: 285-294. 10.1016/S0140-6736(04)16680-4.

    CAS  PubMed  Google Scholar 

  55. Winstanley P, Ward S, Snow R, Breckenridge A: Therapy of falciparum malaria in Sub-Saharan Africa: from molecule to policy. Clin Microbiol Rev. 2004, 17 (3): 612-637. 10.1128/CMR.17.3.612-637.2004.

    PubMed Central  CAS  PubMed  Google Scholar 

  56. Eyers JE: Searching bibliographic databases effectively. Health Policy Plan. 1998, 13 (3): 339-342. 10.1093/heapol/13.3.339.

    CAS  PubMed  Google Scholar 

  57. WHO: Health Information Network . 2005, [http://www.who.int/hinari]

    Google Scholar 

  58. Newman RD, Parise ME, Slutsker L, Nahlen B, Steketee RW: 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. 2003, 8: 488-506. 10.1046/j.1365-3156.2003.01066.x.

    CAS  PubMed  Google Scholar 

  59. Roche: LARIAM (mefloquine hydrochloride): Complete product information. 2004, Nutley, New Jersey, USA , Roche Laboratories, Inc., 1-18.

    Google Scholar 

  60. Smoak BL, Writer JV, Keep LW, Cowan J, Chantelois JL: The effects of inadvertent exposure of mefloquine chemoprophylaxis on pregnancy outcomes and infants of US Army servicewomen . J Inf Dis. 1997, 176: 831-833.

    CAS  Google Scholar 

  61. Okeyeh JN, Lege-Oguntove L, Emembolu JO, Agbo M: Malaria in pregnancy: efficacy of a low dose of mefloquine in an area holoendemic for multi-resistant Plasmodium falciparum. Ann Trop Med Parasitol. 1996 , 90 (3): 265-268.

    CAS  PubMed  Google Scholar 

  62. Nosten F, Vincenti M, Simpson J, Yei P, Thwai KL, de Vries A, Chongsuphajaisiddhi T, White NJ: The effects of mefloquine treatment in pregnancy. Clin Inf Dis. 1999, 28: 808-815.

    CAS  Google Scholar 

  63. Adam I, Ali DA, Alwaseila A, Kheir MM, Elbashir MI: Mefloquine in the treatment of falciparum malaria during pregnancy in Eastern Sudan. Saudi Med J. 2004, 25 (10): 1400-1402.

    PubMed  Google Scholar 

  64. Sowunmi A, Oduola AMJ, Ogundahunsi OAT, Fehintola FA, Ilesanmi OA, Akinyinka OO, Arowojolu AO: Randomised trial of artemether versus artemether and mefloquine for the treatment of chloroquine/sulfadoxine-pyrimethamine-resistant falciparum malaria during pregnancy. J Obstet Gynaecol. 1998, 18 (4): 322-327. 10.1080/01443619867038.

    CAS  PubMed  Google Scholar 

  65. Bounyasong S: Randomized trial of trial of artesunate and mefloquine in comparison with quinine sulphate to treat P.falciparum malaria in pregnant women. J Med Assoc Thai. 2000, 84 (9): 1289-1299.

    Google Scholar 

  66. McGready R, Brockman A, Cho T, Cho D, van Vugt M, Luxemburger C, Chongsuphajaisiddhi T, White NJ, Nosten F: Randomized comparison of mefloquine-artesunate versus quinine in the treatment of multidrug-resistant falciparum malaria in pregnancy. Trans R Soc Trop Med Hyg. 2000, 94: 689-693. 10.1016/S0035-9203(00)90235-9.

    CAS  PubMed  Google Scholar 

  67. Bounyasong S: Randomized trial of trial of artesunate and mefloquine in comparison with quinine sulphate to treat P. falciparum malaria in pregnant women. J Med Assoc Thai. 2001, 84 (9): 1289-1299.

    CAS  PubMed  Google Scholar 

  68. Pfizer: Zithromax. 2004, New York, USA , Pfizer Inc, 1-32.

    Google Scholar 

  69. Heikkinen T, Laine K, Neuvonen PJ, Ekblad U: The transplacental transfer of the macrolide antibiotics erythromycin, roxithromycin and azithromycin. Br J Obstet Gynaecol. 2000, 107 (6): 770-775.

    CAS  Google Scholar 

  70. Ramsey PS, Vaules MB, Vasdev GM, Andrews WW, Ramin KD: Maternal and transplacental pharmacokinetics of azithromycin. Am J Obstet Gynecol. 2003, 188 (3): 714-718. 10.1067/mob.2003.141.

    CAS  PubMed  Google Scholar 

  71. Donders GG: Treatment of sexually transmitted bacterial diseases in pregnant women. Drugs. 2000, 59 (3): 477-485. 10.2165/00003495-200059030-00005.

    CAS  PubMed  Google Scholar 

  72. Duff P: Antibiotic selection in obstetric patients. Infect Dis Clin North Am. 1997, 11 (1): 1-12. 10.1016/S0891-5520(05)70338-X.

    CAS  PubMed  Google Scholar 

  73. Ogasawara KK, Godwin TM: Efficacy of azithromycin in reducing lower genital Ureaplasma urealyticum colonization in women at risk for preterm delivery. J Matern Fetal Med. 1999 , 8 (1): 12-16. 10.1002/(SICI)1520-6661(199901/02)8:1<12::AID-MFM3>3.0.CO;2-8.

    CAS  PubMed  Google Scholar 

  74. Ohrt C, Willingmyre GD, Lee P, Knirsch C, Milhous W: Assessment of azithromycin in combination with other antimalarial drugs against Plasmodium falciparum in vitro. Antimicrob Agents Chemother. 2002, 46 (8): 2518-2524. 10.1128/AAC.46.8.2518-2524.2002.

    PubMed Central  CAS  PubMed  Google Scholar 

  75. Pradines B, Rogier C, Fusai T, Mosnier J, Daries W, Barret E, Parzy D: In vitro activities of antibiotics against Plasmodium falciparum are inhibited by iron. Antimicrob Agents Chemother. 2001, 45 (6): 1746-1750. 10.1128/AAC.45.6.1746-1750.2001.

    PubMed Central  CAS  PubMed  Google Scholar 

  76. Andersen SL, Ager AL, McGreevy P, Schuster BG, Ellis W, Berman J: Efficacy of azithromycin as a causal prophylactic agent against murine malaria. Antimicrob Agents Chemother. 1994, 38 (8): 1862-1863.

    PubMed Central  CAS  PubMed  Google Scholar 

  77. Taylor WR, Richie TL, Fryauff DJ, Ohrt C, Picarima H, Tang D, Murphy GS, Widjaja H, Braitman D, Tjitra E, Ganjar A, Jones TR, Basri H, Berman J: Tolerability of azithromycin as malaria prophylaxis in adults in North East Papua, Indonesia. Antimicrob Agents Chemother. 2003, 47 (7): 2199-2203. 10.1128/AAC.47.7.2199-2203.2003.

    PubMed Central  CAS  PubMed  Google Scholar 

  78. Andersen SL, Oloo AJ, Gordon DM, Ragama OB, Aleman GM, Berman JD, Tang DB, Dunne MW, Shanks GD: Successful double-blinded, randomized, placebo-controlled field trial of azithromycin and doxycycline as prophylaxis for malaria in western Kenya. Clin Inf Dis. 1998, 26: 146-150.

    CAS  Google Scholar 

  79. Krudsood S, Silchamroon U, Wilairatana P, Singhasivanon P, Phumratanaprapin W, Chalermrut K, Phophak N, Popa C: A randomised clinical trial of combination artesunate and azithromycin for treatment of uncomplicated Plasmodium falciparum malaria in Thailand. Southeast Asian J Trop Med Public Health. 2000, 31 (4): 801-807.

    CAS  PubMed  Google Scholar 

  80. WHO: Review of the safety of chlorproguanil-dapsone in the treatment of uncomplicated falciparum malaria in Africa. Report of a technical consultation convened by WHO, Geneva, 1-2 July 2004. 2005, Geneva , World Health Organization

    Google Scholar 

  81. Na-Bachang K, Manyando C, Ruengweerayut R, Kioy D, Mulenga M, Miller GB, Konsil J: The pharmacokinetics and pharmacodynamics of atovoquone and proguanil for the treatment of uncomplicated facliparum malaria in third-trimester pregnant women. Eur J Clin Pharmacol. 2005, 61: 573-582. 10.1007/s00228-005-0969-7.

    Google Scholar 

  82. Mutabingwa TK, Malle LN, de Geus A, Osting J: Malaria chemosuppression in pregnancy. II. Its effect on maternal haemoglobin levels, placental malaria and birth weight. Trop Geogr Med. 1993, 45 (2): 49-55.

    CAS  PubMed  Google Scholar 

  83. GlaxoSmithKline: MALARONE (atovaquone and proguanil hydrochloride): Prescribing Information. 2005, Research Triangle Park, NC 27709 , GlaxoSmithKline , 1-19.

    Google Scholar 

  84. Overbosch D, Schilthuis H, Bienzle U, Behrens RH, Kain KC, Clarke PD, Toovey S, Knobloch J, Nothdurft HD, Shaw D, Roskell NS, Chulay JD, and the Malarone International Study Team: Atovaquone-proguanil versus mefloquine for malaria prophylaxis in non-immune travelers: results from a randomized, double-blind study. Clin Inf Dis. 2001, 33: 1015-1021. 10.1086/322694.

    CAS  Google Scholar 

  85. Looareesuwan S, Wilairatana P, Chalermarut K, Rattanapong Y, Canfield CJ, Hutchinson DBA: Efficacy and safety of atovaquone/proguanil compared with mefloquine for treatment of acute Plasmodium falciparum malaria in Thailand. Am J Trop Med Hyg. 1999, 60 (4): 526-532.

    CAS  PubMed  Google Scholar 

  86. McGready R, Ashley EA, Moo E, Cho T, Barends M, Hutagalung R, Looareesuwan S, White NJ, Nosten F: A randomised comparison of artesunate-atovaquone-proguanil versus quinine in treatment for uncomplicated falciparum malaria during pregnancy. J Inf Dis. 2005, 192: 846-853. 10.1086/432551.

    CAS  Google Scholar 

  87. Mutabingwa TK, Nzila A, Mberu E, Nduati E, Winstanley P, Hills E, Watkins W: Chlorproguanil-dapsone for treatment of drug-resistant falciparum malaria in Tanzania. Lancet. 2001, 358: 1218-1223. 10.1016/S0140-6736(01)06344-9.

    CAS  PubMed  Google Scholar 

  88. Sulo J, Chimpeni P, Hatcher J, Kublin JG, Plowe CV, Molyneux ME, Marsh K, Taylor TE, Watkins WM, Winstanley PA: Comparison of chlorproguanil-dapsone versus sulfadoxine-pyrimethamine for sequential episodes of uncomplicated falciparum malaria in Kenya and Malawi: a randomized clinical trial. Lancet. 2002, 360: 1136-1143. 10.1016/S0140-6736(02)11198-6.

    CAS  PubMed  Google Scholar 

  89. Keuter M, van Eijk A, Hoogstrate M, Raasveld M, van de Ree B, Ngwawe WA, Watkins WM, Were JB, Brandling-Bennett AD: Comparison of chloroquine, pyrimethamine and sulfadoxine, and chlorproguanil and dapsone as treatment for falciparum malaria in pregnant and non-pregnant women, Kakamega District, Kenya. BMJ. 1990, 301 (6750): 466-470.

    PubMed Central  CAS  PubMed  Google Scholar 

  90. National Institutes of Health USA: Treating malaria during pregnancy: a randomized trial of potential options for treatment in an area of high drug resistance in Tanzania. 2006, [http://www.clinicaltrials.gov/ct/show/NCT00146731?order=34]

    Google Scholar 

  91. National Institutes of Health USA: Pharmacokinetics of chlorproguanil-dapsone in pregnant women with Plasmodium falciparum infection, and re-infection with P. falciparum during pregnancy following treatment. 2006, [http://www.clinicaltrials.gov/ct/show/NCT00126971?order=8]

    Google Scholar 

  92. Mohrle JJ: MMV Portfolio: Curing malaria together. 2005, Medicines for Malaria Venture , 26-[http://www.mmv.org]

    Google Scholar 

  93. National Institutes of Health USA: A multi-centre, randomized, double-blind, double dummy study comparing the efficacy and safety of chlorproguanil-dapsone-artesunate versus artemether-lumefantrine in the treatment of acute uncomplicated Plasmodium falciparum malaria in children and adolescents in Africa. 2006, [http://www.clinicaltrials.gov/ct/show/NCT00344006?order=1]

    Google Scholar 

  94. Novartis: Coartem/Riamet. Product monograph: A novel antimalarial combination: one product, two concepts. 2005, Novartis Pharma AG

    Google Scholar 

  95. WHO: Assessment of the safety of artemisinin compounds in pregnancy. Report of two informal consultations convened by WHO in 2002 (Roll Back Malaria and the UNDP/World Bank/WHO Special Programme for Research and Training in Tropical Diseases). 2003, World Health Organization. WHO/CDS/MAL/2003.1094. WHO/RBM/TDR/Artemisinin/03.1 , [http://www.who.int/malaria/cmc_upload/0/000/016/323/artemisinin_pregnancy.pdf]

    Google Scholar 

  96. Longo M, Zanoncelli S, Manera D, Brughera M, Colombo P, Lansen J, Mazue G, Gomes M, Taylor WR, Olliaro P: Effects of the antimalarial drug dihydroartemisinin (DHA) on rat embryos in vitro. Reprod Toxicol. 2006, 21 (1): 83-93. 10.1016/j.reprotox.2005.05.005.

    CAS  PubMed  Google Scholar 

  97. Clark RL: 2006, Wiley InterScience. Epub DOI: 10.1002/bdra.20261, [http://www.interscience.wiley.com] Artesunate: Developmental Toxicity in Monkeys., (Abstract No. 30)., Platform Session Abstracts.

  98. Adam I, Ali DM, Abdalla MA: Artesunate plus sulfadoxine-pyrimethamine in the treatment of uncomplicated Plasmodium falciparum malaria during pregnancy in eastern Sudan. Trans R Soc Trop Med Hyg. 2006, 100: 632-635. 10.1016/j.trstmh.2005.09.019.

    CAS  PubMed  Google Scholar 

  99. Deen JL, von Seidlein L, Pinder M, Walraven GE, Greenwood BM: The safety of the combination artesunate and pyrimethamine-sulfadoxine given during pregnancy. Trans R Soc Trop Med Hyg. 2001, 95 (4): 424-428. 10.1016/S0035-9203(01)90204-4.

    CAS  PubMed  Google Scholar 

  100. McGready R, Cho T, Keo NK, Thwai KL, Villegas L, Looareesuwan S, White NJ, Nosten FR: Artemisinin antimalarials in pregnancy: a prospective treatment study of 539 episodes of multidrug-resistant Plasmodium falciparum. Clin Inf Dis. 2001, 33: 2009-2016. 10.1086/324349.

    CAS  Google Scholar 

  101. McGready R, Stepniewska K, Ward SA, Cho T, Gilveray G, Looareesuwan S, White NJ, Nosten F: Pharmacokinetics of dihydroartemisinin following oral artesunate treatment of pregnant women with acute uncomplicated falciparum malaria. Eur J Clin Pharmacol. 2006, 62 (5): 367-371. 10.1007/s00228-006-0118-y.

    CAS  PubMed  Google Scholar 

  102. Basco LK, Ringwald P: In vitro activities of piperaquine and other 4-aminoquinilones against clinical isolates of Plasmodium falciparum in Cameroon. Antimicrob Agents Chemother. 2003, 47 (4): 1391-1394. 10.1128/AAC.47.4.1391-1394.2003.

    PubMed Central  CAS  PubMed  Google Scholar 

  103. Ashley EA, McGready R, Hutagalung R, Phaiphun L, Slight T, Proux S, Thwai KL, Barends M, Looareesuwan S, White NJ, Nosten F: A randomized, controlled study of a simple, once daily regimen of dihydroartemsinin-piperaquine for the treatment of uncomplicated multi-resistant falciparum malaria. Clin Inf Dis. 2005, 41: 425-432. 10.1086/432011.

    CAS  Google Scholar 

  104. MMV: Chongqing Holley, Sigma-Tau, Medicines for Malaria Venture and University of Oxford sign historic Partnership Agreement to develop antimalarial drug. 2004, [http://www.mmv.org/IMG/pdf/ArtekinPR304.pdf]

    Google Scholar 

  105. Denis MB, Savis TME, Hewitt S, Incardona S, Nimol K, Fandeur T, Poravuth Y, Lim C, Socheat D: Efficacy and safety of dihyroartemesinin-piperaquine (Artekin) in Cambodian children and adults with uncomplicated falciparum malaria. Clin Inf Dis. 2002 , 35: 1469-1476. 10.1086/344647.

    CAS  Google Scholar 

  106. Ashley EA, Krudsoos S, Phaiphun L, Srivilairit S, McGready R, Leowattana W, Hutagalung R, Wilairatana P, Brockman A, Looareesuwan S, Nosten F, White NJ: Randomized, controlled dose-optimization studies of dihydroartemsinin-piperaquine for the treatment of uncomplicated multi-resistant falciparum malaria in Thailand. ,. J Infect Dis. 2004, 190: 1773-1782. 10.1086/425015.

    CAS  PubMed  Google Scholar 

  107. Hien TT, Dolecek C, Mai PP, Dung NT, Truong N, Thai LH, An DTH, Thanh TT, Stepniewska K, White NJ, Farrar J: Dihydroartemsinin-piperaquine against multi-resistant Plasmodium falciparum malaria in Vietnam: randomized clinical trial. Lancet. 2004, 363: 18-22. 10.1016/S0140-6736(03)15163-X.

    CAS  Google Scholar 

  108. Koram KA, Abuaku B, Duah N, Quashie N: Comparative efficacy of antimalarial drugs including ACTs in the treatment of uncomplicated malaria among children under 5 years in Ghana. Acta Trop. 2005, 95 (3): 194-203. 10.1016/j.actatropica.2005.06.018.

    CAS  PubMed  Google Scholar 

  109. Mutabingwa TK, Anthony D, Heler A, Hallett R, Ahmed J, Drakeley C, Greenwood BM, Whitty CJM: Amodiaquine alone, amodiaquine + sulfadoxine-pyrimethamine, amodiaquine + artesunate and artemether-lumefantrine for out patient treatment of malaria in Tanzanian children: a four arm randomised effectiveness trial. Lancet. 2005, 365: 1474-1480. 10.1016/S0140-6736(05)66417-3.

    CAS  PubMed  Google Scholar 

  110. National Institutes of Health USA: Multinational, randomized, comparative study of the efficacy and safety of three therapeutic regimens: Coarsucam (artesunate + amodiaquine fixed-dose combination) administered in 1 or 2 intakes per day versus Coartem (artemether + lumefantrine) in the treatment of uncomplicated Plasmodium falciparum malaria. 2006, [http://www.clinicaltrials.gov/ct/show/NCT00316329?order=9]

    Google Scholar 

  111. Chandramohan D, Owusu-Agyei S, Carneiro I, Awine T, Amponsa-Achiano K, Mensah N, Jaffar S, Baiden R, Hodgson A, Binka F, Greenwood B: Cluster randomised trial of intermittent preventive treatment for malaria in infants in area of high, seasonal transmission in Ghana. BMJ. 2005, 331: 727-733. 10.1136/bmj.331.7519.727.

    PubMed Central  PubMed  Google Scholar 

  112. Cisse B, Sokhna C, Boulanger D, Milet J, Ba EH, Richardson K, Hallett R, Sutherland C, Simondon K, Simondon F, Alexander N, Gaye O, Targett G, Lines J, Greenwood B, Trape JF: Seasonal intermittent preventive treatment with artesunate and sulfadoxine-pyrimethamine for prevention of malaria in Senegalese children: a randomized, placebo-controlled, double-blind trial. Lancet. 2006, 367 (9511): 659-667. 10.1016/S0140-6736(06)68264-0.

    CAS  PubMed  Google Scholar 

  113. Zongo I, Dorsey G, Rouamba N, Dokomajilar C, Lankoande M, Ouedraogo JB, Rosenthal PJ: Amodiaquine, sulfadoxine-pyrimethamine, and combination therapy for uncomplicated falciparum malaria: a randomized controlled trial from Burkina Faso. Am J Trop Med Hyg. 2005, 73 (5): 826-832.

    CAS  PubMed  Google Scholar 

  114. Aina O, Emeka P, Agomo P, Akintonwa A: Comparative efficacy study of CQ, DHA and DHA plus mefloquine combination in children with acute uncomplicated falciparum malaria. The 4th MIM Pan-African Malaria Conference,. 2005, Yaoundé, Cameroon

    Google Scholar 

  115. Auparakkitanon S, Chapoomram S, Kuaha K, Chirachariyavej T, Wilairat P: Targeting of hematin by the antimalarial pyronaridine. Antimicrob Agents Chemother. 2006, 50 (6): 2197-2200. 10.1128/AAC.00119-06.

    PubMed Central  CAS  PubMed  Google Scholar 

  116. National Institutes of Health USA: An open-label, phase II, dose-escalation clinical study to assess the pharmacokinetics, safety, tolerability and pharmacodynamics of fixed dose combination tablet of pyronaridine and artesunate (3:1) in children with acute uncomplicated Plasmodium falciparum malaria . 2006, [http://www.clinicaltrials.gov/ct/show/NCT00331136?order=39]

    Google Scholar 

  117. Liu DQ, Lin SG, Feng XP, Chen WJ, Chen PL, Wu HM, Chen C, Liu J: Study on treatment of multi-drug resistant falciparum malaria by using a combination of dihydroartemisinin and pyronaridine [Article in Chinese]. Zhongguo Ji Sheng Chong Xue Yu Ji Sheng Chong Bing Za Zhi [=Chinese Journal of Parasitology and Parasitic Diseases]. 2002, 20 (4): 193-196.

    Google Scholar 

  118. Brentlinger PE, Behrens CB, Micek MA: Challenges in the concurrent management of malaria and HIV in pregnancy in sub-Saharan Africa. Lancet Inf Dis. 2006, 6 (2): 100-111. 10.1016/S1473-3099(06)70383-8.

    Google Scholar 

  119. Fried M, Duffy PE: Adherence of Plasmodium falciparum to chondroitin sulfate A in the human placenta. Science. 1996, 272: 1502-1504. 10.1126/science.272.5267.1502.

    CAS  PubMed  Google Scholar 

  120. Beeson JG, Rogerson SJ, Cooke BM, Reeder JC, Chai W, Lawson AM, Molyneux ME, Brown GV: Adhesion of Plasodium falciparm-infected erythrocytes to hyaluronic acid in placental malaria. Nat Med. 2000, 6 (86-90):

  121. Riley EM, Schnieder G, Sambou I, Greenwood BM: Suppression of cell-mediated immune responses to malaria antigens in pregnant Gambian women. Am J Trop Med Hyg. 1989, 40 (2): 141-144.

    CAS  PubMed  Google Scholar 

  122. Fievet N, Cot M, Chougnet C, Maubert B, Bickii J, Dubois B, Lehesran JY, Frobert Y, Migot F: Malaria and pregnancy in Cameroonian primigravidae-humoral and cellular immune responses to Plasmodium falciparum blood-stage antigens. Am J Trop Med Hyg. 1995, 53: 612-617.

    CAS  PubMed  Google Scholar 

  123. Fievet N, Cot M, Ringwald P, Bickii J, Dubois B, Le Hesran JY, Migot F, Deloron P: Immune response to Plasmodium falciparum antigens in Cameroonian primigravidae: evolution after delivery and during second trimester. Clin Exp Immunol. 1997, 107 : 462-467. 10.1046/j.1365-2249.1997.d01-966.x.

    PubMed Central  CAS  PubMed  Google Scholar 

  124. O'Meara WP, Breman JG, McKenzie FE: The promise and potential challenges of intermittent preventive treatment for malaria in infants (IPTi). Malar J. 2005, 4 (33):

  125. Roche: Fanisdar brand of sulfadoxine and pyrimethamine tablets: Complete product information. 2004, Nutley, New Jersey, USA , Roche Laboratories, Inc.

    Google Scholar 

  126. Mutabingwa TK: Artemisinin-based combination therapies (ACTs): Best hope for malaria treatment but inaccessible to the needy!. Acta Trop. 2005, 95 (3): 305-315. 10.1016/j.actatropica.2005.06.009.

    CAS  PubMed  Google Scholar 

  127. Wilkinson D, Abdool Karim SS, Harrison A, Lurie M, Colvin M, Connolly C, Sturm AW: Unrecognized sexually transmitted infections in rural South African women: A hidden epidemic. Bull World Health Organ. 1999, 77 (1): 22-28.

    PubMed Central  CAS  PubMed  Google Scholar 

  128. Kaul R, Kimani J, Nagelkerke NJ, Fonck K, Ngugi EN, Keli F, MacDonald KS, Maclean IW, Bwayo JJ, Temmerman M, Ronald AR, Moses SKHIVSG: Monthly antibiotic chemoprophylaxis and incidence of sexually transmitted infections and HIV-1 infection in Kenyan sex workers: a randomized controlled trial. JAMA. 2004, 291 (21): 2555-2562. 10.1001/jama.291.21.2555.

    CAS  PubMed  Google Scholar 

  129. Rastogi S, Kapur S, Salhan S, Mittal A: Chlamydia trachomatis infection in pregnancy: risk factor for an adverse outcome. Br J Biomed Sci. 1999, 56 (2): 94-98.

    CAS  PubMed  Google Scholar 

  130. Pukrittayakamee S, Chantra A, Simpson JA, Vanijanonta S, Clemens R, Looareesuwan S, White NJ: Therapeutic responses to different antimalarial drugs in vivax malaria. Antimicrob Agents Chemother. 2000, 44 (6): 1680-1685. 10.1128/AAC.44.6.1680-1685.2000.

    PubMed Central  CAS  PubMed  Google Scholar 

  131. Ranque S, Badiaga S, Delmont J, Brouqui P: Triangular test applied to the clinical trial of azithromycin against relapses in Plasmodium vivax infections. Malar J. 2002, 1: 13-10.1186/1475-2875-1-13.

    PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

We thank our colleagues for discussions and their support during the writing of this review, particularly Dr Alex Mwita, Director, National Malaria Control Programme, Dar Es Salaam, Tanzania and Dr Roly Gosling, London School of Hygiene and Tropical Medicine, UK/Kili IPTi Programme, Tanga, Tanzania.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Andrew Vallely.

Additional information

Authors' contributions

AV participated in the conception and design of the review, coordinated the literature review and drafted the manuscript.

LV participated in the conception and design of the review, conducted the literature search, obtained full text articles and helped draft the manuscript.

JC participated in the design of the review and helped draft the manuscript.

BG participated in the conception and design of the review, provided additional information e.g. conference proceedings and helped draft the manuscript.

DC conceived the study, participated in its design and coordination and helped draft the manuscript.

All authors have read and approved the final manuscript.

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Vallely, A., Vallely, L., Changalucha, J. et al. Intermittent preventive treatment for malaria in pregnancy in Africa: What's new, what's needed?. Malar J 6, 16 (2007). https://doi.org/10.1186/1475-2875-6-16

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1475-2875-6-16

Keywords