Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Malar J ; 14: 247, 2015 Jun 18.
Article in English | MEDLINE | ID: mdl-26082036

ABSTRACT

BACKGROUND: Measurement of densities of host-seeking malaria vectors is important for estimating levels of disease transmission, for appropriately allocating interventions, and for quantifying their impact. The gold standard for estimating mosquito-human contact rates is the human landing catch (HLC), where human volunteers catch mosquitoes that land on their exposed body parts. This approach necessitates exposure to potentially infectious mosquitoes, and is very labour intensive. There are several safer and less labour-intensive methods, with Centers for Disease Control light traps (LT) placed indoors near occupied bed nets being the most widely used. METHODS: This paper presents analyses of 13 studies with paired mosquito collections of LT and HLC to evaluate these methods for their consistency in sampling indoor-feeding mosquitoes belonging to the two major taxa of malaria vectors across Africa, the Anopheles gambiae sensu lato complex and the Anopheles funestus s.l. group. Both overall and study-specific sampling efficiencies of LT compared with HLC were computed, and regression methods that allow for the substantial variations in mosquito counts made by either method were used to test whether the sampling efficacy varies with mosquito density. RESULTS: Generally, LT were able to collect similar numbers of mosquitoes to the HLC indoors, although the relative sampling efficacy, measured by the ratio of LT:HLC varied considerably between studies. The overall best estimate for An. gambiae s.l. was 1.06 (95% credible interval: 0.68-1.64) and for An. funestus s.l. was 1.37 (0.70-2.68). Local calibration exercises are not reproducible, since only in a few studies did LT sample proportionally to HLC, and there was no geographical pattern or consistent trend with average density in the tendency for LT to either under- or over-sample. CONCLUSIONS: LT are a crude tool at best, but are relatively easy to deploy on a large scale. Spatial and temporal variation in mosquito densities and human malaria transmission exposure span several orders of magnitude, compared to which the inconsistencies of LT are relatively small. LT, therefore, remain an invaluable and safe alternative to HLC for measuring indoor malaria transmission exposure in Africa.


Subject(s)
Anopheles/physiology , Insect Vectors/physiology , Mosquito Control/methods , Africa, Eastern , Africa, Western , Animals , Centers for Disease Control and Prevention, U.S. , Feeding Behavior , Humans , Malaria/prevention & control , Malaria/transmission , Mozambique , United States
2.
Lancet ; 380(9857): 1916-26, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23122217

ABSTRACT

BACKGROUND: Malaria is one of the greatest causes of mortality worldwide. Use of the most effective treatments for malaria remains inadequate for those in need, and there is concern over the emergence of resistance to these treatments. In 2010, the Global Fund launched the Affordable Medicines Facility--malaria (AMFm), a series of national-scale pilot programmes designed to increase the access and use of quality-assured artemisinin based combination therapies (QAACTs) and reduce that of artemisinin monotherapies for treatment of malaria. AMFm involves manufacturer price negotiations, subsidies on the manufacturer price of each treatment purchased, and supporting interventions such as communications campaigns. We present findings on the effect of AMFm on QAACT price, availability, and market share, 6-15 months after the delivery of subsidised ACTs in Ghana, Kenya, Madagascar, Niger, Nigeria, Uganda, and Tanzania (including Zanzibar). METHODS: We did nationally representative baseline and endpoint surveys of public and private sector outlets that stock antimalarial treatments. QAACTs were identified on the basis of the Global Fund's quality assurance policy. Changes in availability, price, and market share were assessed against specified success benchmarks for 1 year of AMFm implementation. Key informant interviews and document reviews recorded contextual factors and the implementation process. FINDINGS: In all pilots except Niger and Madagascar, there were large increases in QAACT availability (25·8-51·9 percentage points), and market share (15·9-40·3 percentage points), driven mainly by changes in the private for-profit sector. Large falls in median price for QAACTs per adult equivalent dose were seen in the private for-profit sector in six pilots, ranging from US$1·28 to $4·82. The market share of oral artemisinin monotherapies decreased in Nigeria and Zanzibar, the two pilots where it was more than 5% at baseline. INTERPRETATION: Subsidies combined with supporting interventions can be effective in rapidly improving availability, price, and market share of QAACTs, particularly in the private for-profit sector. Decisions about the future of AMFm should also consider the effect on use in vulnerable populations, access to malaria diagnostics, and cost-effectiveness. FUNDING: The Global Fund to Fight AIDS, Tuberculosis and Malaria, and the Bill & Melinda Gates Foundation.


Subject(s)
Antimalarials/economics , Artemisinins/economics , Lactones/economics , Malaria/drug therapy , Africa , Antimalarials/standards , Antimalarials/supply & distribution , Artemisinins/standards , Artemisinins/supply & distribution , Drug Costs , Humans , Lactones/standards , Lactones/supply & distribution , Malaria/economics , Marketing of Health Services , Pharmacies/economics , Pharmacies/statistics & numerical data , Pilot Projects , Private Sector/economics , Public Sector/economics
3.
PLoS One ; 7(3): e32900, 2012.
Article in English | MEDLINE | ID: mdl-22412946

ABSTRACT

BACKGROUND: Intermittent preventive treatment of malaria in children (IPTc) is a highly efficacious method of malaria control where malaria transmission is highly seasonal. However, no studies published to date have examined community perceptions of IPTc. METHODS: A qualitative study was undertaken in parallel with a double-blind, placebo-controlled, randomized trial of IPTc conducted in Mali and Burkina Faso in 2008-2009 to assess community perceptions of and recommendations for IPTc. Caregivers and community health workers (CHWs) were purposively sampled. Seventy-two in-depth individual interviews and 23 focus group discussions were conducted. FINDINGS: Widespread perceptions of health benefits for children led to enthusiasm for the trial and for IPTc specifically. Trust in and respect for those providing the tablets and a sense of obligation to the community to participate in sanctioned activities favoured initial adoption. IPTc fits in well with existing understandings of childhood illness. Participants did not express concerns about the specific drugs used for IPTc or about providing tablets to children without symptoms of malaria. There was no evidence that IPTc was perceived as a substitute for bed net usage, nor did it inhibit care seeking. Participants recommended that distribution be "closer to the population", but expressed concern over caregivers' ability to administer tablets at home. CONCLUSIONS: The trial context mediated perceptions of IPTc. Nonetheless, the results indicate that community perceptions of IPTc in the settings studied were largely favourable and that the delivery strategy rather than the tablets themselves presented the main areas of concern for caregivers and CHWs. The study identifies a number of key questions to consider in planning an IPTc distribution strategy. Single-dose formulations could increase the success of IPTc implementation, as could integration of IPTc within a package of activities, such as bed net distribution and free curative care, for which demand is already high.


Subject(s)
Early Medical Intervention , Malaria/prevention & control , Antimalarials/therapeutic use , Burkina Faso , Child, Preschool , Female , Health Knowledge, Attitudes, Practice , Humans , Infant , Malaria/drug therapy , Male , Mali , Qualitative Research , Residence Characteristics
4.
PLoS One ; 6(8): e23391, 2011.
Article in English | MEDLINE | ID: mdl-21858097

ABSTRACT

BACKGROUND: Interventions that reduce exposure to malaria infection may lead to delayed malaria morbidity and mortality. We investigated whether intermittent preventive treatment of malaria in children (IPTc) was associated with an increase in the incidence of malaria after cessation of the intervention. METHODS: An individually randomised, trial of IPTc, comparing three courses of sulphadoxine pyrimethamine (SP) plus amodiaquine (AQ) with placebos was implemented in children aged 3-59 months during the 2008 malaria transmission season in Burkina Faso. All children in the trial were given a long lasting insecticide treated net; 1509 children received SP+AQ and 1505 received placebos. Passive surveillance for malaria was maintained until the end of the subsequent malaria transmission season in 2009, and active surveillance for malaria infection, anaemia and malnutrition was conducted. RESULTS: On thousand, four hundred and sixteen children (93.8%) and 1399 children (93.0%) initially enrolled in the intervention and control arms of the trial respectively were followed during the 2009 malaria transmission season. During the period July 2009 to November 2009, incidence rates of clinical malaria were 3.84 (95%CI; 3.67-4.02) and 3.45 (95%CI; 3.29-3.62) episodes per child during the follow up period in children who had previously received IPT or placebos, indicating a small increase in risk for children in the former intervention arm (IRR = 1.12; 95%CI 1.04-1.20) (P = 0.003). Children who had received SP+AQ had a lower prevalence of malaria infection (adjusted PR: 0.88 95%CI: 0.79-0.98) (P = 0.04) but they had a higher parasite density (P = 0.001) if they were infected. There was no evidence that the risks of moderately severe anaemia (Hb<8 g/dL), wasting, stunting, or of being underweight in children differed between treatment arms. CONCLUSION: IPT with SP+AQ was associated with a small increase in the incidence of clinical malaria in the subsequent malaria transmission season. TRIAL REGISTRATION: ClinicalTrials.gov NCT00738946.


Subject(s)
Amodiaquine/therapeutic use , Malaria, Falciparum/prevention & control , Pyrimethamine/therapeutic use , Sulfadoxine/therapeutic use , Anemia/epidemiology , Antimalarials/therapeutic use , Body Weight , Burkina Faso/epidemiology , Child, Preschool , Double-Blind Method , Drug Administration Schedule , Drug Combinations , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Incidence , Infant , Kaplan-Meier Estimate , Malaria, Falciparum/epidemiology , Malaria, Falciparum/transmission , Male , Morbidity , Mosquito Nets , Plasmodium falciparum/drug effects , Prevalence , Seasons , Treatment Outcome
5.
PLoS One ; 6(8): e23390, 2011.
Article in English | MEDLINE | ID: mdl-21858096

ABSTRACT

BACKGROUND: Intermittent preventive treatment of malaria in children (IPTc) is a promising strategy for malaria control. A study conducted in Mali in 2008 showed that administration of three courses of IPTc with sulphadoxine-pyrimethamine (SP) and amodiaquine (AQ) at monthly intervals reduced clinical malaria, severe malaria and malaria infection by >80% in children under 5 years of age. Here we report the results of a follow-on study undertaken to establish whether children who had received IPTc would be at increased risk of malaria during the subsequent malaria transmission season. METHODS: Morbidity from malaria and the prevalence of malaria parasitaemia and anaemia were measured in children who had previously received IPTc with SP and AQ using similar surveillance methods to those employed during the previous intervention period. RESULTS: 1396 of 1508 children (93%) who had previously received IPTc and 1406 of 1508 children (93%) who had previously received placebos were followed up during the high malaria transmission season of the year following the intervention. Incidence rates of clinical malaria during the post-intervention transmission season (July-November 2009) were 1.87 (95% CI 1.76-1.99) and 1.73 (95% CI; 1.62-1.85) episodes per child year in the previous intervention and placebo groups respectively; incidence rate ratio (IRR) 1.09 (95% CI 0.99-1.21) (P = 0.08). The prevalence of malaria infection was similar in the two groups, 7.4% versus 7.5%, prevalence ratio (PR) of 0.99 (95% CI 0.73-1.33) (P = 0.95). At the end of post-intervention malaria transmission season, the prevalence of anaemia, defined as a haemoglobin concentration<11g/dL, was similar in the two groups (56.2% versus 55.6%; PR = 1.01 [95% CI 0.91-1.12]) (P = 0.84). CONCLUSION: IPTc with SP+AQ was not associated with an increase in incidence of malaria episodes, prevalence of malaria infection or anaemia in the subsequent malaria transmission season. TRIAL REGISTRATION: ClinicalTrials.gov NCT00738946.


Subject(s)
Amodiaquine/therapeutic use , Malaria, Falciparum/prevention & control , Pyrimethamine/therapeutic use , Sulfadoxine/therapeutic use , Anemia/epidemiology , Antimalarials/therapeutic use , Body Weight , Child, Preschool , Drug Administration Schedule , Drug Combinations , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Incidence , Infant , Kaplan-Meier Estimate , Malaria, Falciparum/epidemiology , Malaria, Falciparum/transmission , Male , Mali/epidemiology , Morbidity , Mosquito Nets , Parasitemia/epidemiology , Plasmodium falciparum/drug effects , Prevalence , Seasons , Treatment Outcome
6.
PLoS Med ; 8(2): e1000407, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21304923

ABSTRACT

BACKGROUND: Previous studies have shown that in areas of seasonal malaria transmission, intermittent preventive treatment of malaria in children (IPTc), targeting the transmission season, reduces the incidence of clinical malaria. However, these studies were conducted in communities with low coverage with insecticide-treated nets (ITNs). Whether IPTc provides additional protection to children sleeping under an ITN has not been established. METHODS AND FINDINGS: To assess whether IPTc provides additional protection to children sleeping under an ITN, we conducted a randomised, double-blind, placebo-controlled trial of IPTc with sulphadoxine pyrimethamine (SP) plus amodiaquine (AQ) in three localities in Kati, Mali. After screening, eligible children aged 3-59 mo were given a long-lasting insecticide-treated net (LLIN) and randomised to receive three rounds of active drugs or placebos. Treatments were administered under observation at monthly intervals during the high malaria transmission season in August, September, and October 2008. Adverse events were monitored immediately after the administration of each course of IPTc and throughout the follow-up period. The primary endpoint was clinical episodes of malaria recorded through passive surveillance by study clinicians available at all times during the follow-up. Cross-sectional surveys were conducted in 150 randomly selected children weekly and in all children at the end of the malaria transmission season to assess usage of ITNs and the impact of IPTc on the prevalence of malaria, anaemia, and malnutrition. Cox regression was used to compare incidence rates between intervention and control arms. The effects of IPTc on the prevalence of malaria infection and anaemia were estimated using logistic regression. 3,065 children were screened and 3,017 (1,508 in the control and 1,509 in the intervention arm) were enrolled in the study. 1,485 children (98.5%) in the control arm and 1,481 (98.1%) in the intervention arm completed follow-up. During the intervention period, the proportion of children reported to have slept under an ITN was 99.7% in the control and 99.3% in intervention arm (p = 0.45). A total of 672 episodes of clinical malaria defined as fever or a history of fever and the presence of at least 5,000 asexual forms of Plasmodium falciparum per microlitre (incidence rate of 1.90; 95% confidence interval [CI] 1.76-2.05 episodes per person year) were observed in the control arm versus 126 (incidence rate of 0.34; 95% CI 0.29-0.41 episodes per person year) in the intervention arm, indicating a protective effect (PE) of 82% (95% CI 78%-85%) (p<0.001) on the primary endpoint. There were 15 episodes of severe malaria in children in the control arm compared to two in children in the intervention group giving a PE of 87% (95% CI 42%-99%) (p = 0.001). IPTc reduced the prevalence of malaria infection by 85% (95% CI 73%-92%) (p<0.001) during the intervention period and by 46% (95% CI 31%-68%) (p<0.001) at the end of the intervention period. The prevalence of moderate anaemia (haemoglobin [Hb] <8 g/dl) was reduced by 47% (95% CI 15%-67%) (p<0.007) at the end of intervention period. The frequencies of adverse events were similar between the two arms. There was no drug-related serious adverse event. CONCLUSIONS: IPTc given during the malaria transmission season provided substantial protection against clinical episodes of malaria, malaria infection, and anaemia in children using an LLIN. SP+AQ was safe and well tolerated. These findings indicate that IPTc could make a valuable contribution to malaria control in areas of seasonal malaria transmission alongside other interventions. TRIAL REGISTRATION: ClinicalTrials.gov NCT00738946. Please see later in the article for the Editors' Summary.


Subject(s)
Insecticide-Treated Bednets , Malaria/prevention & control , Child, Preschool , Double-Blind Method , Female , Humans , Infant , Malaria/transmission , Male , Mali , Placebos
7.
PLoS Med ; 8(2): e1000408, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21304925

ABSTRACT

BACKGROUND: Intermittent preventive treatment of malaria in children (IPTc) is a promising new approach to the control of malaria in areas of seasonal malaria transmission but it is not known if IPTc adds to the protection provided by an insecticide-treated net (ITN). METHODS AND FINDINGS: An individually randomised, double-blind, placebo-controlled trial of seasonal IPTc was conducted in Burkina Faso in children aged 3 to 59 months who were provided with a long-lasting insecticide-treated bednet (LLIN). Three rounds of treatment with sulphadoxine pyrimethamine plus amodiaquine or placebos were given at monthly intervals during the malaria transmission season. Passive surveillance for malaria episodes was established, a cross-sectional survey was conducted at the end of the malaria transmission season, and use of ITNs was monitored during the intervention period. Incidence rates of malaria were compared using a Cox regression model and generalized linear models were fitted to examine the effect of IPTc on the prevalence of malaria infection, anaemia, and on anthropometric indicators. 3,052 children were screened and 3,014 were enrolled in the trial; 1,505 in the control arm and 1,509 in the intervention arm. Similar proportions of children in the two treatment arms were reported to sleep under an LLIN during the intervention period (93%). The incidence of malaria, defined as fever or history of fever with parasitaemia ≥ 5,000/µl, was 2.88 (95% confidence interval [CI] 2.70-3.06) per child during the intervention period in the control arm versus 0.87 (95% CI 0.78-0.97) in the intervention arm, a protective efficacy (PE) of 70% (95% CI 66%-74%) (p<0.001). There was a 69% (95% CI 6%-90%) reduction in incidence of severe malaria (p = 0.04) and a 46% (95% CI 7%-69%) (p = 0.03) reduction in the incidence of all-cause hospital admissions. IPTc reduced the prevalence of malaria infection at the end of the malaria transmission season by 73% (95% CI 68%-77%) (p<0.001) and that of moderately severe anaemia by 56% (95% CI 36%-70%) (p<0.001). IPTc reduced the risks of wasting (risk ratio [RR] = 0.79; 95% CI 0.65-1.00) (p = 0.05) and of being underweight (RR = 0.84; 95% CI 0.72-0.99) (p = 0.03). Children who received IPTc were 2.8 (95% CI 2.3-3.5) (p<0.001) times more likely to vomit than children who received placebo but no drug-related serious adverse event was recorded. CONCLUSIONS: IPT of malaria provides substantial protection against malaria in children who sleep under an ITN. There is now strong evidence to support the integration of IPTc into malaria control strategies in areas of seasonal malaria transmission. TRIAL REGISTRATION: ClinicalTrials.govNCT00738946. Please see later in the article for the Editors' Summary.


Subject(s)
Insecticide-Treated Bednets , Malaria/prevention & control , Amodiaquine/therapeutic use , Antimalarials/therapeutic use , Burkina Faso , Child, Preschool , Double-Blind Method , Drug Combinations , Humans , Infant , Pyrimethamine/therapeutic use , Sulfadoxine/therapeutic use
8.
PLoS Med ; 6(4): e1000055, 2009 Apr 14.
Article in English | MEDLINE | ID: mdl-19365539

ABSTRACT

BACKGROUND: Although the molecular basis of resistance to a number of common antimalarial drugs is well known, a geographic description of the emergence and dispersal of resistance mutations across Africa has not been attempted. To that end we have characterised the evolutionary origins of antifolate resistance mutations in the dihydropteroate synthase (dhps) gene and mapped their contemporary distribution. METHODS AND FINDINGS: We used microsatellite polymorphism flanking the dhps gene to determine which resistance alleles shared common ancestry and found five major lineages each of which had a unique geographical distribution. The extent to which allelic lineages were shared among 20 African Plasmodium falciparum populations revealed five major geographical groupings. Resistance lineages were common to all sites within these regions. The most marked differentiation was between east and west African P. falciparum, in which resistance alleles were not only of different ancestry but also carried different resistance mutations. CONCLUSIONS: Resistant dhps has emerged independently in multiple sites in Africa during the past 10-20 years. Our data show the molecular basis of resistance differs between east and west Africa, which is likely to translate into differing antifolate sensitivity. We have also demonstrated that the dispersal patterns of resistance lineages give unique insights into recent parasite migration patterns.


Subject(s)
Antimalarials/pharmacology , Dihydropteroate Synthase/genetics , Drug Resistance/genetics , Malaria, Falciparum/drug therapy , Membrane Transport Proteins/genetics , Plasmodium falciparum/drug effects , Protozoan Proteins/genetics , Africa/epidemiology , Alleles , Animals , Antimalarials/therapeutic use , Chloroquine/pharmacology , Chloroquine/therapeutic use , DNA, Protozoan/genetics , Drug Combinations , Humans , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Malaria, Falciparum/prevention & control , Microsatellite Repeats , Phylogeny , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , Plasmodium falciparum/isolation & purification , Polymorphism, Single Nucleotide , Population Surveillance , Pyrimethamine/pharmacology , Pyrimethamine/therapeutic use , Selection, Genetic , Sulfadoxine/pharmacology , Sulfadoxine/therapeutic use
9.
J Infect Dis ; 196(1): 138-44, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17538894

ABSTRACT

BACKGROUND: It has been suggested that reducing exposure to malaria by vector control might impair the development of naturally acquired immunity to malaria. It is also thought that an individual's ability to clear drug-resistant malarial parasites after treatment is enhanced by acquired immunity. METHODS: To investigate the hypothesis that insecticide-treated materials may affect the acquisition of immunity to malaria, we compared the ability of children living in villages in which insecticide-treated curtains (ITCs) had been used for 6-8 years to clear resistant parasites after treatment with chloroquine (CQ) with that of children living in unprotected villages. RESULTS: A total of 1035 children aged 6-59 months with falciparum malaria were treated with CQ; 409 were subsequently identified as carrying parasites with the pfcrt-76T allele. More children from ITC villages cleared parasites harboring this allele than did children from non-ITC villages (34.1% vs. 24.0%; adjusted odds ratio [OR], 1.80 [95% confidence interval {CI}, 1.15-2.80]; P=.01). The difference in the clearance of parasites with the pfcrt-76T allele was seen in children aged 6-35 months (32.3% vs. 19.3%; adjusted OR, 2.34 [95% CI, 1.18-4.66]; P=.02) but not in older children (37.3% vs. 37.0%; adjusted OR, 1.09 [95% CI, 0.56-2.10]; P=.97). Rates of adequate clinical response among children carrying parasites with the pfcrt-76T allele were similar in ITC and non-ITC villages (75.1% vs. 68.6%; adjusted OR, 1.21 [95% CI, 0.61-2.39]; P=.58). CONCLUSION: Our data suggest that the children who were protected from malaria by ITCs acquired functional immunity more rapidly than did the control children.


Subject(s)
Antimalarials/therapeutic use , Bedding and Linens , Chloroquine/therapeutic use , Insecticides , Malaria, Falciparum/drug therapy , Mosquito Control , Plasmodium falciparum/isolation & purification , Animals , Burkina Faso , Child, Preschool , Drug Resistance, Microbial , Female , Humans , Infant , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Male , Membrane Transport Proteins/genetics , Parasitemia , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Rural Population
10.
Am J Trop Med Hyg ; 76(2): 237-44, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17297030

ABSTRACT

The impact of vector control measures on the evolution of antimalarial drug resistance is an important issue for malaria control programs. We investigated whether the in vivo efficacy of chloroquine (CQ) in children aged 6-59 months with uncomplicated malaria differed in 9 villages that had benefited from long-term use of insecticide-treated curtains (ITCs) and in 9 nearby non-ITC villages. We also compared the prevalence of genetic markers of resistance to CQ and sulfadoxine-pyrimethamine (SP) between the two groups of villages. The study enrolled 1,035 children with uncomplicated malaria and 231 infected but asymptomatic children. After taking account of re-infections, the proportions of children who experienced clinical failure after treatment with CQ were 14% and 19% in ITC and non-ITC villages, respectively (OR = 0.68; 95% CI: 0.39, 1.18). Parasitologic failure was observed in 49% of children in ITC villages and 58% of children in non-ITC villages (OR = 0.71 95%CI: 0.44, 1.13). The proportion of symptomatic children who harbored parasites carrying the pfcrt-76T allele was 43% in ITC villages and 40% in non-ITC villages (OR = 1.09; 95%CI: 0.80, 1.50). The pfmdr1-86Y allele was detected in 31% and 29% of children in the two groups of villages (OR = 1.14; 95%CI: 0.75, 1.72). Triple mutations in the dhfr gene were observed in 12% of children in both groups. No double mutations in the dhps gene were observed. Similar results were observed in asymptomatic children. In this setting, ITC use was not associated with increased circulation of parasites resistant to standard antimalarial drugs, or with a greater risk of treatment failure among children less than 5 years of age.


Subject(s)
Antimalarials/therapeutic use , Bedding and Linens , Chloroquine/therapeutic use , Insecticides , Malaria, Falciparum/prevention & control , Mosquito Control/methods , Plasmodium falciparum/growth & development , Animals , Burkina Faso/epidemiology , Child, Preschool , Cross-Sectional Studies , DNA, Protozoan/chemistry , DNA, Protozoan/genetics , Drug Combinations , Drug Resistance , Humans , Infant , Insect Vectors/parasitology , Malaria, Falciparum/drug therapy , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/genetics , Plasmodium falciparum/genetics , Point Mutation , Polymerase Chain Reaction , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Pyrimethamine/therapeutic use , Rural Population , Sulfadoxine/therapeutic use , Tetrahydrofolate Dehydrogenase/chemistry , Tetrahydrofolate Dehydrogenase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...