Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
PLoS Med ; 21(5): e1004376, 2024 May.
Article in English | MEDLINE | ID: mdl-38723040

ABSTRACT

BACKGROUND: Recently revised WHO guidelines on malaria chemoprevention have opened the door to more tailored implementation. Countries face choices on whether to replace old drugs, target additional age groups, and adapt delivery schedules according to local drug resistance levels and malaria transmission patterns. Regular routine assessment of protective efficacy of chemoprevention is key. Here, we apply a novel modelling approach to aid the design and analysis of chemoprevention trials and generate measures of protection that can be applied across a range of transmission settings. METHODS AND FINDINGS: We developed a model of genotype-specific drug protection, which accounts for underlying risk of infection and circulating genotypes. Using a Bayesian framework, we fitted the model to multiple simulated scenarios to explore variations in study design, setting, and participant characteristics. We find that a placebo or control group with no drug protection is valuable but not always feasible. An alternative approach is a single-arm trial with an extended follow-up (>42 days), which allows measurement of the underlying infection risk after drug protection wanes, as long as transmission is relatively constant. We show that the currently recommended 28-day follow-up in a single-arm trial results in low precision of estimated 30-day chemoprevention efficacy and low power in determining genotype differences of 12 days in the duration of protection (power = 1.4%). Extending follow-up to 42 days increased precision and power (71.5%) in settings with constant transmission over this time period. However, in settings of unstable transmission, protective efficacy in a single-arm trial was overestimated by 24.3% if recruitment occurred during increasing transmission and underestimated by 15.8% when recruitment occurred during declining transmission. Protective efficacy was estimated with greater precision in high transmission settings, and power to detect differences by resistance genotype was lower in scenarios where the resistant genotype was either rare or too common. CONCLUSIONS: These findings have important implications for the current guidelines on chemoprevention efficacy studies and will be valuable for informing where these studies should be optimally placed. The results underscore the need for a comparator group in seasonal settings and provide evidence that the extension of follow-up in single-arm trials improves the accuracy of measures of protective efficacy in settings with more stable transmission. Extension of follow-up may pose logistical challenges to trial feasibility and associated costs. However, these studies may not need to be repeated multiple times, as the estimates of drug protection against different genotypes can be applied to different settings by adjusting for transmission intensity and frequency of resistance.


Subject(s)
Antimalarials , Chemoprevention , Drug Resistance , Malaria , Humans , Antimalarials/therapeutic use , Drug Resistance/genetics , Malaria/prevention & control , Malaria/transmission , Malaria/epidemiology , Chemoprevention/methods , Bayes Theorem , Genotype , Research Design
2.
Am J Trop Med Hyg ; 109(2): 248-257, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37364860

ABSTRACT

Zambia's National Malaria Elimination Program transitioned to Fludora Fusion in 2019 for annual indoor residual spraying (IRS) in Nchelenge District, an area with holoendemic malaria transmission. Previously, IRS was associated with reductions in parasite prevalence during the rainy season only, presumably because of insufficient residual insecticide longevity. This study assessed the impact of transitioning from Actellic 300CS to long-acting Fludora Fusion using active surveillance data from 2014 through 2021. A difference-in-differences analysis estimated changes in rainy season parasite prevalence associated with living in a sprayed house, comparing insecticides. The change in the 2020 to 2021 dry season parasite prevalence associated with living in a house sprayed with Fludora Fusion was also estimated. Indoor residual spraying with Fludora Fusion was not associated with decreased rainy season parasite prevalence compared with IRS with Actellic 300CS (ratio of prevalence ratios [PRs], 1.09; 95% CI, 0.89-1.33). Moreover, living in a house sprayed with either insecticide was not associated with decreased malaria risk (Actellic 300CS: PR, 0.97; 95% CI, 0.86-1.10; Fludora Fusion: rainy season PR, 1.06; 95% CI, 0.89-1.25; dry season PR, 1.21; 95% CI, 0.99-1.48). In contrast, each 10% increase in community IRS coverage was associated with a 4% to 5% reduction in parasite prevalence (rainy season: PR, 0.95; 95% CI, 0.92-0.97; dry season: PR, 0.96; 95% CI, 0.94-0.99), suggesting a community-level protective effect, and corroborating the importance of high-intervention coverage.


Subject(s)
Insecticides , Malaria , Humans , Zambia/epidemiology , Mosquito Control , Malaria/epidemiology , Malaria/prevention & control , Malaria/parasitology
3.
Science ; 378(6623): eadd8737, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36454863

ABSTRACT

The geographic and evolutionary origins of the SARS-CoV-2 Omicron variant (BA.1), which was first detected mid-November 2021 in Southern Africa, remain unknown. We tested 13,097 COVID-19 patients sampled between mid-2021 to early 2022 from 22 African countries for BA.1 by real-time RT-PCR. By November-December 2021, BA.1 had replaced the Delta variant in all African sub-regions following a South-North gradient, with a peak Rt of 4.1. Polymerase chain reaction and near-full genome sequencing data revealed genetically diverse Omicron ancestors already existed across Africa by August 2021. Mutations, altering viral tropism, replication and immune escape, gradually accumulated in the spike gene. Omicron ancestors were therefore present in several African countries months before Omicron dominated transmission. These data also indicate that travel bans are ineffective in the face of undetected and widespread infection.

5.
Am J Trop Med Hyg ; 107(4_Suppl): 55-67, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36228903

ABSTRACT

For a decade, the Southern and Central Africa International Center of Excellence for Malaria Research has operated with local partners across study sites in Zambia and Zimbabwe that range from hypo- to holoendemic and vary ecologically and entomologically. The burden of malaria and the impact of control measures were assessed in longitudinal cohorts, cross-sectional surveys, passive and reactive case detection, and other observational designs that incorporated multidisciplinary scientific approaches: classical epidemiology, geospatial science, serosurveillance, parasite and mosquito genetics, and vector bionomics. Findings to date have helped elaborate the patterns and possible causes of sustained low-to-moderate transmission in southern Zambia and eastern Zimbabwe and recalcitrant high transmission and fatality in northern Zambia. Cryptic and novel mosquito vectors, asymptomatic parasite reservoirs in older children, residual parasitemia and gametocytemia after treatment, indoor residual spraying timed dyssynchronously to vector abundance, and stockouts of essential malaria commodities, all in the context of intractable rural poverty, appear to explain the persistent malaria burden despite current interventions. Ongoing studies of high-resolution transmission chains, parasite population structures, long-term malaria periodicity, and molecular entomology are further helping to lay new avenues for malaria control in southern and central Africa and similar settings.


Subject(s)
Insecticides , Malaria , Parasites , Africa, Central , Animals , Child , Cross-Sectional Studies , Humans , Malaria/epidemiology , Malaria/prevention & control , Mosquito Control , Zambia/epidemiology , Zimbabwe/epidemiology
6.
Am J Trop Med Hyg ; 107(4_Suppl): 68-74, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36228913

ABSTRACT

The International Centers of Excellence for Malaria Research (ICEMR) were established by the National Institute of Allergy and Infectious Diseases more than a decade ago to provide multidisciplinary research support to malaria control programs worldwide, operating in endemic areas and contributing technology, expertise, and ultimately policy guidance for malaria control and elimination. The Southern and Central Africa ICEMR has conducted research across three main sites in Zambia and Zimbabwe that differ in ecology, entomology, transmission intensity, and control strategies. Scientific findings led to new policies and action by the national malaria control programs and their partners in the selection of methods, materials, timing, and locations of case management and vector control. Malaria risk maps and predictive models of case detection furnished by the ICEMR informed malaria elimination programming in southern Zambia, and time series analyses of entomological and parasitological data motivated several major changes to indoor residual spray campaigns in northern Zambia. Along the Zimbabwe-Mozambique border, temporal and geospatial data are currently informing investigations into a recent resurgence of malaria. Other ICEMR findings pertaining to parasite and mosquito genetics, human behavior, and clinical epidemiology have similarly yielded immediate and long-term policy implications at each of the sites, often with generalizable conclusions. The ICEMR programs thereby provide rigorous scientific investigations and analyses to national control and elimination programs, without which the impediments to malaria control and their potential solutions would remain understudied.


Subject(s)
Malaria , Mosquito Vectors , Africa, Central , Animals , Humans , Malaria/epidemiology , Malaria/prevention & control , Mosquito Control/methods , Policy , Zambia/epidemiology , Zimbabwe/epidemiology
7.
Am J Trop Med Hyg ; 107(5): 1145-1153, 2022 11 14.
Article in English | MEDLINE | ID: mdl-36252797

ABSTRACT

Human movement drives spatial transmission patterns of infectious diseases. Population-level mobility patterns are often quantified using aggregated data sets, such as census migration surveys or mobile phone data. These data are often unable to quantify individual-level travel patterns and lack the information needed to discern how mobility varies by demographic groups. Individual-level datasets can capture additional, more precise, aspects of mobility that may impact disease risk or transmission patterns and determine how mobility differs across cohorts; however, these data are rare, particularly in locations such as sub-Saharan Africa. Using detailed GPS logger data collected from three sites in southern Africa, we explore metrics of mobility such as percent time spent outside home, number of locations visited, distance of locations, and time spent at locations to determine whether they vary by demographic, geographic, or temporal factors. We further create a composite mobility score to identify how well aggregated summary measures would capture the full extent of mobility patterns. Although sites had significant differences in all mobility metrics, no site had the highest mobility for every metric, a distinction that was not captured by the composite mobility score. Further, the effects of sex, age, and season on mobility were all dependent on site. No factor significantly influenced the number of trips to locations, a common way to aggregate datasets. When collecting and analyzing human mobility data, it is difficult to account for all the nuances; however, these analyses can help determine which metrics are most helpful and what underlying differences may be present.


Subject(s)
Cell Phone , Communicable Diseases , Humans , Travel , Surveys and Questionnaires
8.
Malar J ; 20(1): 361, 2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34488784

ABSTRACT

Malaria control has stalled in a number of African countries and novel approaches to malaria control are needed for these areas. The encouraging results of a recent trial conducted in young children in Burkina Faso and Mali in which a combination of the RTS,S/AS01E malaria vaccine and seasonal malaria chemoprevention led to a substantial reduction in clinical cases of malaria, severe malaria, and malaria deaths compared with the administration of either intervention given alone suggests that there may be other epidemiological/clinical situations in which a combination of malaria vaccination and chemoprevention could be beneficial. Some of these potential opportunities are considered in this paper. These include combining vaccination with intermittent preventive treatment of malaria in infants, with intermittent preventive treatment of malaria in pregnancy (through vaccination of women of child-bearing age before or during pregnancy), or with post-discharge malaria chemoprevention in the management of children recently admitted to hospital with severe anaemia. Other potential uses of the combination are prevention of malaria in children at particular risk from the adverse effects of clinical malaria, such as those with sickle cell disease, and during the final stages of a malaria elimination programme when vaccination could be combined with repeated rounds of mass drug administration. The combination of a pre-erythrocytic stage malaria vaccine with an effective chemopreventive regimen could make a valuable contribution to malaria control and elimination in a variety of clinical or epidemiological situations, and the potential of this approach to malaria control needs to be explored.


Subject(s)
Chemoprevention/statistics & numerical data , Communicable Disease Control/methods , Malaria Vaccines/therapeutic use , Malaria/prevention & control , Vaccination/statistics & numerical data , Communicable Disease Control/statistics & numerical data , Humans
9.
Am J Trop Med Hyg ; 104(6): 2131-2138, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33844650

ABSTRACT

Since the late nineteenth century, the importance of house structure as a determinant of malaria risk has been recognized. Few studies to date have examined the association of housing and malaria in clinical populations. We conducted a cross-sectional study of febrile patients (n = 282) at two rural health clinics in a high malaria-transmission area of northern Zambia. Participants underwent testing for Plasmodium falciparum infection by PCR. Demographic and other risk factors including house structure, indoor residual spraying (IRS), bed net use, education level, and household income were collected. Data were fitted to logistic regression models for relational and mediation analyses. Residing in a house with a thatch roof was associated with higher odds of malaria than residing in a house with corrugated metal (odds ratio: 2.6; 95% CI: 1.0-6.3, P = 0.04). Lower income and educational attainment were also associated with greater odds of malaria. Living under a thatch roof accounted for 24% (95% CI: 14-82) of the effect of household income on malaria risk, and income accounted for 11% (95% CI: 8-19) of the effect of education. Neither IRS nor bed net use was associated with malaria risk despite large, local investments in these vector control interventions. The findings testify to malaria as a disease of rural poverty and contribute further evidence to the utility of housing improvements in vector control programs.


Subject(s)
Fever/epidemiology , Fever/parasitology , Housing/standards , Malaria, Falciparum/transmission , Adolescent , Adult , Child , Child, Preschool , Cross-Sectional Studies , Female , Humans , Malaria, Falciparum/epidemiology , Malaria, Falciparum/prevention & control , Male , Middle Aged , Odds Ratio , Plasmodium falciparum/physiology , Prevalence , Risk Factors , Rural Population , Young Adult , Zambia/epidemiology
11.
Am J Trop Med Hyg ; 104(2): 683-694, 2020 12 21.
Article in English | MEDLINE | ID: mdl-33350376

ABSTRACT

The global malaria burden has decreased substantially, but gains have been uneven both within and between countries. In Zambia, the malaria burden remains high in northern and eastern regions of the country. To effectively reduce malaria transmission in these areas, evidence-based intervention strategies are needed. Zambia's National Malaria Control Centre conducted targeted indoor residual spraying (IRS) in 40 high-burden districts from 2014 to 2016 using the novel organophosphate insecticide pirimiphos-methyl. The Southern and Central Africa International Centers of Excellence for Malaria Research conducted an evaluation of the impact of the IRS campaign on household vector abundance in Nchelenge District, Luapula Province. From April 2012 to July 2017, field teams conducted indoor overnight vector collections from 25 to 30 households per month using Centers for Disease Control light traps. Changes in indoor anopheline counts before versus after IRS were assessed by species using negative binomial regression models with robust standard errors, controlling for geographic and climatological covariates. Counts of Anopheles funestus declined by approximately 50% in the study area and within areas targeted for IRS, and counts of Anopheles gambiae declined by approximately 40%. Within targeted areas, An. funestus counts declined more in sprayed households than in unsprayed households; however, this relationship was not observed for An. gambiae. The moderate decrease in indoor vector abundance indicates that IRS with pirimiphos-methyl is an effective vector control measure, but a more comprehensive package of interventions is needed with sufficient coverage to effectively reduce the malaria burden in this setting.


Subject(s)
Anopheles/drug effects , Insecticides/pharmacology , Malaria/prevention & control , Malaria/transmission , Mosquito Control/methods , Mosquito Vectors/drug effects , Organothiophosphorus Compounds/pharmacology , Animals , Family Characteristics , Female , Malaria/epidemiology , Mosquito Control/standards , Time Factors , Zambia/epidemiology
12.
Malar J ; 18(1): 400, 2019 Dec 04.
Article in English | MEDLINE | ID: mdl-31801548

ABSTRACT

BACKGROUND: While the utility of parasite genotyping for malaria elimination has been extensively documented in low to moderate transmission settings, it has been less well-characterized in holoendemic regions. High malaria burden settings have received renewed attention acknowledging their critical role in malaria elimination. Defining the role for parasite genomics in driving these high burden settings towards elimination will enhance future control programme planning. METHODS: Amplicon deep sequencing was used to characterize parasite population genetic diversity at polymorphic Plasmodium falciparum loci, Pfama1 and Pfcsp, at two timepoints in June-July 2016 and January-March 2017 in a high transmission region along the international border between Luapula Province, Zambia and Haut-Katanga Province, the Democratic Republic of the Congo (DRC). RESULTS: High genetic diversity was observed across both seasons and in both countries. No evidence of population structure was observed between parasite populations on either side of the border, suggesting that this region may be one contiguous transmission zone. Despite a decline in parasite prevalence at the sampling locations in Haut-Katanga Province, no genetic signatures of a population bottleneck were detected, suggesting that larger declines in transmission may be required to reduce parasite genetic diversity. Analysing rare variants may be a suitable alternative approach for detecting epidemiologically important genetic signatures in highly diverse populations; however, the challenge is distinguishing true signals from potential artifacts introduced by small sample sizes. CONCLUSIONS: Continuing to explore and document the utility of various parasite genotyping approaches for understanding malaria transmission in holoendemic settings will be valuable to future control and elimination programmes, empowering evidence-based selection of tools and methods to address pertinent questions, thus enabling more efficient resource allocation.


Subject(s)
Genetic Variation , Malaria, Falciparum/prevention & control , Plasmodium falciparum/genetics , Democratic Republic of the Congo/epidemiology , Malaria, Falciparum/epidemiology , Seasons , Zambia/epidemiology
13.
Int J Health Geogr ; 18(1): 19, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31426819

ABSTRACT

BACKGROUND: Human movement is a driver of malaria transmission and has implications for sustainable malaria control. However, little research has been done on the impact of fine-scale movement on malaria transmission and control in high-transmission settings. As interest in targeted malaria control increases, evaluations are needed to determine the appropriateness of these strategies in the context of human mobility across a variety of transmission settings. METHODS: A human mobility study was conducted in Nchelenge District, a high-transmission setting in northern Zambia. Over 1 year, 84 participants were recruited from active malaria surveillance cohorts to wear a global positioning system data logger for 1 month during all daily activity. Participants completed a survey questionnaire and underwent malaria testing and treatment at the time of logger distribution and at collection 1 month later. Incident malaria infections were identified using polymerase chain reaction. Participant movement was characterized throughout the study area and across areas targeted for an indoor residual spraying (IRS) intervention. Participant movement patterns were compared using movement intensity maps, activity space plots, and statistical analyses. Malaria risk was characterized across participants using spatial risk maps and time spent away from home during peak vector biting hours. RESULTS: Movement data were collected from 82 participants, and 63 completed a second study visit. Participants exhibited diverse mobility patterns across the study area, including movement into and out of areas targeted for IRS, potentially mitigating the impact of IRS on parasite prevalence. Movement patterns did not differ significantly by season or age, but male participants traveled longer distances and spent more time away from home. Monthly malaria incidence was 22%, and malaria risk was characterized as high across participants. Participants with incident parasitemia traveled a shorter distance and spent more time away from home during peak biting hours; however, these relationships were not statistically significant, and malaria risk score did not differ by incident parasitemia. CONCLUSIONS: Individual movement patterns in Nchelenge District, Zambia have implications for malaria control, particularly the effectiveness of targeted IRS strategies. Large and fine-scale population mobility patterns should be considered when planning intervention strategies across transmission settings.


Subject(s)
Geographic Information Systems , Malaria/epidemiology , Malaria/transmission , Mosquito Control/methods , Movement , Spatial Behavior , Adolescent , Adult , Aged , Cross-Sectional Studies , Data Interpretation, Statistical , Female , Follow-Up Studies , Humans , Longitudinal Studies , Male , Middle Aged , Movement/physiology , Spatial Behavior/physiology , Time Factors , Young Adult , Zambia/epidemiology
14.
Am J Epidemiol ; 188(12): 2120-2130, 2019 12 31.
Article in English | MEDLINE | ID: mdl-31062839

ABSTRACT

Malaria transmission in northern Zambia has increased in the past decade, despite malaria control activities. Evidence-based intervention strategies are needed to effectively reduce malaria transmission. Zambia's National Malaria Control Centre conducted targeted indoor residual spraying (IRS) in Nchelenge District, Luapula Province, from 2014 to 2016 using the organophosphate insecticide pirimiphos-methyl. An evaluation of the IRS campaign was conducted by the Southern Africa International Centers of Excellence for Malaria Research using actively detected malaria cases in bimonthly household surveys carried out from April 2012 to July 2017. Changes in malaria parasite prevalence after IRS were assessed by season using Poisson regression models with robust standard errors, controlling for clustering of participants in households and demographic, geographical, and climatological covariates. In targeted areas, parasite prevalence declined approximately 25% during the rainy season following IRS with pirimiphos-methyl but did not decline during the dry season or in the overall study area. Within targeted areas, parasite prevalence declined in unsprayed households, suggesting both direct and indirect effects of IRS. The moderate decrease in parasite prevalence within sprayed areas indicates that IRS with pirimiphos-methyl is an effective malaria control measure, but a more comprehensive package of interventions is needed to effectively reduce the malaria burden in this setting.


Subject(s)
Insecticides , Malaria/epidemiology , Mosquito Control/methods , Mosquito Vectors , Organothiophosphorus Compounds , Adolescent , Child , Child, Preschool , Female , Humans , Malaria/prevention & control , Malaria/transmission , Male , Zambia/epidemiology
15.
Am J Trop Med Hyg ; 101(1): 126-136, 2019 07.
Article in English | MEDLINE | ID: mdl-31074411

ABSTRACT

Malaria transmission is dependent on the density and distribution of mosquito vectors, but drivers of vector abundance have not been adequately studied across a range of transmission settings. To inform intervention strategies for high-burden areas, further investigation is needed to identify predictors of vector abundance. Active household (HH) surveillance was conducted in Nchelenge district, Luapula Province, northern Zambia, a high-transmission setting with limited impact of malaria control. Between April 2012 and July 2017, mosquitoes were collected indoors during HH visits using CDC light traps. Demographic, environmental, and climatological correlates of vector abundance were identified using log-binomial regression models with robust standard errors. The primary malaria vectors in this setting were Anopheles funestus sensu stricto (s.s.) and Anopheles gambiae s.s. Anopheles funestus predominated in both seasons, with a peak in the dry season. Anopheles gambiae peaked at lower numbers in the rainy season. Environmental, climatic, and demographic factors were correlated with HH vector abundance. Higher vector counts were found in rural areas with low population density and among HHs close to roads and small streams. Vector counts were lower with increasing elevation and slope. Anopheles funestus was negatively associated with rainfall at lags of 2-6 weeks, and An. gambiae was positively associated with rainfall at lags of 3-10 weeks. Both vectors had varying relationships with temperature. These results suggest that malaria vector control in Nchelenge district should occur throughout the year, with an increased focus on dry-season transmission and rural areas.


Subject(s)
Family Characteristics , Housing , Light , Malaria/transmission , Mosquito Control , Mosquito Vectors , Centers for Disease Control and Prevention, U.S. , Humans , Malaria/epidemiology , Population Density , Risk Factors , United States , Zambia/epidemiology
16.
Malar J ; 18(1): 180, 2019 May 24.
Article in English | MEDLINE | ID: mdl-31126288

ABSTRACT

BACKGROUND: HIV-infected individuals on antiretroviral therapy (ART) require treatment with artemisinin-based combination therapy (ACT) when infected with malaria. Artemether-lumefantrine (AL) is the most commonly used ACT for treatment of falciparum malaria in Africa but there is limited evidence on the safety and efficacy of AL in HIV-infected individuals on ART, among whom drug-drug interactions are expected. Day-42 adequate clinical and parasitological response (ACPR) and incidence of adverse events was assessed in HIV-infected individuals on efavirenz-based ART with uncomplicated falciparum malaria treated with AL. METHODS: A prospective, open label, non-randomized, interventional clinical trial was conducted at St Paul's Hospital in northern Zambia, involving 152 patients aged 15-65 years with uncomplicated falciparum malaria, who were on efavirenz-based ART. They received a 3-day directly observed standard treatment of AL and were followed up until day 63. Day-42 polymerase chain reaction (PCR)-corrected ACPRs (95% confidence interval [CI]) were calculated for the intention-to-treat population. RESULTS: Enrolled patients had a baseline geometric mean (95% CI) parasite density of 1108 (841-1463) parasites/µL; 16.4% (25/152) of the participants had a recurrent malaria episode by day 42. However, PCR data was available for 17 out of the 25 patients who had malaria recurrence. Among all the 17 patients, PCR findings demonstrated malaria re-infection, making the PCR-adjusted day-42 ACPR 100% in the 144 patients who could be evaluated. Even when eight patients with missing PCR data were considered very conservatively as failures, the day-42 ACPR was over 94%. None of the participants, disease or treatment characteristics, including day-7 lumefantrine concentrations, predicted the risk of malaria recurrence by day 42. AL was well tolerated following administration. There were only two cases of grade 3 neutropaenia and one serious adverse event of lobar pneumonia, none of which was judged as probably related to intake of AL. CONCLUSIONS: AL was well tolerated and efficacious in treating uncomplicated falciparum malaria in HIV co-infected adults on efavirenz-based ART. However, a higher than anticipated proportion of participants experienced malaria re-infection, which highlights the need for additional malaria prevention measures in this sub-population after treatment with AL. Trial registration Pan African Clinical Trials Registry (PACTR): PACTR201311000659400. Registered on 4 October 2013. https://pactr.samrc.ac.za/Search.aspx.


Subject(s)
Antimalarials/therapeutic use , Artemether, Lumefantrine Drug Combination/therapeutic use , Benzoxazines/therapeutic use , Malaria, Falciparum/drug therapy , Reverse Transcriptase Inhibitors/therapeutic use , Adolescent , Adult , Aged , Alkynes , Antimalarials/adverse effects , Artemether, Lumefantrine Drug Combination/adverse effects , Cyclopropanes , Female , HIV Infections/drug therapy , Humans , Male , Middle Aged , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Polymerase Chain Reaction , Prospective Studies , Young Adult , Zambia
17.
mSphere ; 4(2)2019 03 27.
Article in English | MEDLINE | ID: mdl-30918058

ABSTRACT

Antibodies to Plasmodium falciparum are specific biomarkers that can be used to monitor parasite exposure over broader time frames than microscopy, rapid diagnostic tests, or molecular assays. Consequently, seroprevalence surveys can assist with monitoring the impact of malaria control interventions, particularly in the final stages of elimination, when parasite incidence is low. The protein array format to measure antibodies to diverse P. falciparum antigens requires only small sample volumes and is high throughput, permitting the monitoring of malaria transmission on large spatial and temporal scales. We expanded the use of a protein microarray to assess malaria transmission in settings beyond those with a low malaria incidence. Antibody responses in children and adults were profiled, using a P. falciparum protein microarray, through community-based surveys in three areas in Zambia and Zimbabwe at different stages of malaria control and elimination. These three epidemiological settings had distinct serological profiles reflective of their malaria transmission histories. While there was little correlation between transmission intensity and antibody signals (magnitude or breadth) in adults, there was a clear correlation in children younger than 5 years of age. Antibodies in adults appeared to be durable even in the absence of significant recent transmission, whereas antibodies in children provided a more accurate picture of recent levels of transmission intensity. Seroprevalence studies in children could provide a valuable marker of progress toward malaria elimination.IMPORTANCE As malaria approaches elimination in many areas of the world, monitoring the effect of control measures becomes more important but challenging. Low-level infections may go undetected by conventional tests that depend on parasitemia, particularly in immune individuals, who typically show no symptoms of malaria. In contrast, antibodies persist after parasitemia and may provide a more accurate picture of recent exposure. Only a few parasite antigens-mainly vaccine candidates-have been evaluated in seroepidemiological studies. We examined antibody responses to 500 different malaria proteins in blood samples collected through community-based surveillance from areas with low, medium, and high malaria transmission intensities. The breadth of the antibody responses in adults was broad in all three settings and was a poor correlate of recent exposure. In contrast, children represented a better sentinel population for monitoring recent malaria transmission. These data will help inform the use of multiplex serology for malaria surveillance.


Subject(s)
Antibodies, Protozoan/blood , Antibodies, Protozoan/immunology , Antibody Formation , Malaria/immunology , Malaria/transmission , Plasmodium falciparum/immunology , Adolescent , Adult , Age Factors , Aged , Antigens, Protozoan/immunology , Biomarkers/blood , Child , Child, Preschool , Community Participation , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Malaria/epidemiology , Male , Middle Aged , Protein Array Analysis , Seroepidemiologic Studies , Young Adult , Zambia/epidemiology , Zimbabwe/epidemiology
18.
Am J Trop Med Hyg ; 100(4): 842-845, 2019 04.
Article in English | MEDLINE | ID: mdl-30719965

ABSTRACT

We attempted to identify Plasmodium falciparum histidine-rich protein 2/3 (pfhrp2/3) deletions among rapid diagnostic test (RDT)-negative but PCR- or microscopy-positive P. falciparum-infected individuals in areas of low transmission (Choma District, 2009-2011) and high transmission (Nchelenge District, 2015-2017) in Zambia. Through community-based surveys, 5,167 participants were screened at 1,147 households by P. falciparum histidine-rich protein 2 (PfHRP2)-based RDTs. Slides were made and dried blood spots were obtained for molecular analysis. Of 28 samples with detectable P. falciparum DNA, none from Nchelenge District were pfhrp2/3 negative. All eight samples from Choma District had detectable pfhrp3 genes, but pfhrp2 was undetectable in three. DNA concentrations of pfhrp2-negative samples were low (< 0.001 ng/µL). These findings suggest that PfHRP2-based RDTs remain effective tools for malaria diagnosis in Nchelenge District, but further study is warranted to understand the potential for pfhrp2/3 deletions in southern Zambia where malaria transmission declined over the past decade.


Subject(s)
Antigens, Protozoan/genetics , Gene Deletion , Malaria, Falciparum/diagnosis , Molecular Diagnostic Techniques , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , Cross-Sectional Studies , DNA, Protozoan/genetics , Dried Blood Spot Testing , Humans , Zambia
19.
Sci Transl Med ; 11(473)2019 01 02.
Article in English | MEDLINE | ID: mdl-30602535

ABSTRACT

A large proportion of ongoing malaria parasite transmission is attributed to low-density subclinical infections not readily detected by available rapid diagnostic tests (RDTs) or microscopy. Plasmodium falciparum gametocyte carriage is subclinical, but gametocytemic individuals comprise the parasite reservoir that leads to infection of mosquitoes and local transmission. Effective detection and quantification of these carriers can help advance malaria elimination strategies. However, no point-of-need (PON) RDTs for gametocyte detection exist, much less one that can perform noninvasive sampling of saliva outside a clinical setting. Here, we report on the discovery of 35 parasite markers from which we selected a single candidate for use in a PON RDT. We performed a cross-sectional, multi-omics study of saliva from 364 children with subclinical infection in Cameroon and Zambia and produced a prototype saliva-based PON lateral flow immunoassay test for P. falciparum gametocyte carriers. The test is capable of identifying submicroscopic carriage in both clinical and nonclinical settings and is compatible with archived saliva samples.


Subject(s)
Asymptomatic Infections , Diagnostic Tests, Routine/methods , Disease Reservoirs/parasitology , Malaria, Falciparum/diagnosis , Malaria, Falciparum/parasitology , Parasites/physiology , Saliva/parasitology , Adolescent , Animals , Biomarkers/metabolism , Cameroon , Child , Cross-Sectional Studies , Female , Humans , Limit of Detection , Parasitemia/diagnosis , Parasitemia/parasitology , Protozoan Proteins/metabolism , Zambia
20.
Am J Trop Med Hyg ; 98(6): 1699-1704, 2018 06.
Article in English | MEDLINE | ID: mdl-29692306

ABSTRACT

Malaria remains a public health crisis in areas where it has resisted control efforts. In Nchelenge District, a high-transmission area in northern Zambia, malaria accounts for more than one-third of pediatric hospitalizations and nearly one-half of hospital deaths in children. To identify risk factors for death due to malaria, we conducted a retrospective, time-matched case-control study of 126 children hospitalized with malaria who died (cases) and 126 children who survived (controls). There were no differences in age, gender, hemoglobin concentration, or prevalence of severe anemia between cases and controls. Children who died were more likely to come from villages located at greater distances from the hospital than children who survived (median 13.5 versus 3.2 km). Each additional kilometer of distance from the hospital increased the odds of death by 4% (odds ratio 1.04, 95% confidence interval 1.01-1.07, P < 0.01). Extent of anemia and admission during periods when blood was unavailable for transfusion were associated with early death (P ≤ 0.03). Delays in initiation of treatment of severe malaria contribute to the increased odds of death in children referred from more distant health centers, and might be mitigated by transportation improvements, capacity at rural health posts to administer treatment before transfer, hospital triage systems that minimize time to treatment, and reliable blood product stores at referral hospitals.


Subject(s)
Anemia/epidemiology , Malaria/epidemiology , Anemia/mortality , Anemia/parasitology , Case-Control Studies , Child, Preschool , Cross-Sectional Studies , Female , Hospitalization , Hospitals , Humans , Infant , Malaria/mortality , Malaria/parasitology , Male , Outpatients , Prevalence , Retrospective Studies , Risk Factors , Rural Health , Zambia/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL