Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
Proc Natl Acad Sci U S A ; 121(24): e2320898121, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38833464

ABSTRACT

The World Health Organization identifies a strong surveillance system for malaria and its mosquito vector as an essential pillar of the malaria elimination agenda. Anopheles salivary antibodies are emerging biomarkers of exposure to mosquito bites that potentially overcome sensitivity and logistical constraints of traditional entomological surveys. Using samples collected by a village health volunteer network in 104 villages in Southeast Myanmar during routine surveillance, the present study employs a Bayesian geostatistical modeling framework, incorporating climatic and environmental variables together with Anopheles salivary antigen serology, to generate spatially continuous predictive maps of Anopheles biting exposure. Our maps quantify fine-scale spatial and temporal heterogeneity in Anopheles salivary antibody seroprevalence (ranging from 9 to 99%) that serves as a proxy of exposure to Anopheles bites and advances current static maps of only Anopheles occurrence. We also developed an innovative framework to perform surveillance of malaria transmission. By incorporating antibodies against the vector and the transmissible form of malaria (sporozoite) in a joint Bayesian geostatistical model, we predict several foci of ongoing transmission. In our study, we demonstrate that antibodies specific for Anopheles salivary and sporozoite antigens are a logistically feasible metric with which to quantify and characterize heterogeneity in exposure to vector bites and malaria transmission. These approaches could readily be scaled up into existing village health volunteer surveillance networks to identify foci of residual malaria transmission, which could be targeted with supplementary interventions to accelerate progress toward elimination.


Subject(s)
Anopheles , Bayes Theorem , Malaria , Mosquito Vectors , Animals , Anopheles/parasitology , Mosquito Vectors/parasitology , Humans , Malaria/transmission , Malaria/epidemiology , Malaria/immunology , Malaria/parasitology , Seroepidemiologic Studies , Insect Bites and Stings/epidemiology , Insect Bites and Stings/immunology , Insect Bites and Stings/parasitology , Sporozoites/immunology
2.
PLOS Glob Public Health ; 3(8): e0002134, 2023.
Article in English | MEDLINE | ID: mdl-37611001

ABSTRACT

Access to medical treatment for fever is essential to prevent morbidity and mortality in individuals and to prevent transmission of communicable febrile illness in communities. Quantification of the rates at which treatment is accessed is critical for health system planning and a prerequisite for disease burden estimates. In this study, national data on the proportion of children under five years old with fever who were taken for medical treatment were collected from all available countries in Africa, Latin America, and Asia (n = 91). We used generalised additive mixed models to estimate 30-year trends in the treatment-seeking rates across the majority of countries in these regions (n = 151). Our results show that the proportions of febrile children brought for medical treatment increased steadily over the last 30 years, with the greatest increases occurring in areas where rates had originally been lowest, which includes Latin America and Caribbean, North Africa and the Middle East (51 and 50% increase, respectively), and Sub-Saharan Africa (23% increase). Overall, the aggregated and population-weighted estimate of children with fever taken for treatment at any type of facility rose from 61% (59-64 95% CI) in 1990 to 71% (69-72 95% CI) in 2020. The overall population-weighted average for fraction of treatment in the public sector was largely unchanged during the study period: 49% (42-58 95% CI) sought care at public facilities in 1990 and 47% (44-52 95% CI) in 2020. Overall, the findings indicate that improvements in access to care have been made where they were most needed, but that despite rapid initial gains, progress can plateau without substantial investment. In 2020 there remained significant gaps in care utilisation that must be factored in when developing control strategies and deriving disease burden estimates.

3.
Trop Med Infect Dis ; 8(7)2023 Jul 14.
Article in English | MEDLINE | ID: mdl-37505659

ABSTRACT

No studies have yet examined high-resolution shifts in the spatial patterns of human movement in Australia throughout 2020 and 2021, a period coincident with the repeated enactment and removal of varied governmental restrictions aimed at reducing community transmission of SARS-CoV-2. We compared overlapping timeseries of COVID-19 pandemic-related restrictions, epidemiological data on cases and vaccination rates, and high-resolution human movement data to characterize population-level responses to the pandemic in Australian cities. We found that restrictions on human movement and/or mandatory business closures reduced the average population-level weekly movement volumes in cities, as measured by aggregated travel time, by almost half. Of the movements that continued to occur, long movements reduced more dramatically than short movements, likely indicating that people stayed closer to home. We also found that the repeated lockdowns did not reduce their impact on human movement, but the effect of the restrictions on human movement waned as the duration of restrictions increased. Lastly, we found that after restrictions ceased, the subsequent surge in SARS-CoV-2 transmission coincided with a substantial, non-mandated drop in human movement volume. These findings have implications for public health policy makers when faced with anticipating responses to restrictions during future emergency situations.

4.
Int J Epidemiol ; 52(4): 1124-1136, 2023 08 02.
Article in English | MEDLINE | ID: mdl-37164625

ABSTRACT

BACKGROUND: Reliable and detailed data on the prevalence of tuberculosis (TB) with sub-national estimates are scarce in Ethiopia. We address this knowledge gap by spatially predicting the national, sub-national and local prevalence of TB, and identifying drivers of TB prevalence across the country. METHODS: TB prevalence data were obtained from the Ethiopia national TB prevalence survey and from a comprehensive review of published reports. Geospatial covariates were obtained from publicly available sources. A random effects meta-analysis was used to estimate a pooled prevalence of TB at the national level, and model-based geostatistics were used to estimate the spatial variation of TB prevalence at sub-national and local levels. Within the MBG Plugin Framework, a logistic regression model was fitted to TB prevalence data using both fixed covariate effects and spatial random effects to identify drivers of TB and to predict the prevalence of TB. RESULTS: The overall pooled prevalence of TB in Ethiopia was 0.19% [95% confidence intervals (CI): 0.12%-0.28%]. There was a high degree of heterogeneity in the prevalence of TB (I2 96.4%, P <0.001), which varied by geographical locations, data collection periods and diagnostic methods. The highest prevalence of TB was observed in Dire Dawa (0.96%), Gambela (0.88%), Somali (0.42%), Addis Ababa (0.28%) and Afar (0.24%) regions. Nationally, there was a decline in TB prevalence from 0.18% in 2001 to 0.04% in 2009. However, prevalence increased back to 0.29% in 2014. Substantial spatial variation of TB prevalence was observed at a regional level, with a higher prevalence observed in the border regions, and at a local level within regions. The spatial distribution of TB prevalence was positively associated with population density. CONCLUSION: The results of this study showed that TB prevalence varied substantially at sub-national and local levels in Ethiopia. Spatial patterns were associated with population density. These results suggest that targeted interventions in high-risk areas may reduce the burden of TB in Ethiopia and additional data collection would be required to make further inferences on TB prevalence in areas that lack data.


Subject(s)
Tuberculosis , Humans , Ethiopia/epidemiology , Prevalence , Tuberculosis/epidemiology , Logistic Models , Population Density
5.
Malar J ; 22(1): 138, 2023 Apr 26.
Article in English | MEDLINE | ID: mdl-37101269

ABSTRACT

BACKGROUND: As both mechanistic and geospatial malaria modeling methods become more integrated into malaria policy decisions, there is increasing demand for strategies that combine these two methods. This paper introduces a novel archetypes-based methodology for generating high-resolution intervention impact maps based on mechanistic model simulations. An example configuration of the framework is described and explored. METHODS: First, dimensionality reduction and clustering techniques were applied to rasterized geospatial environmental and mosquito covariates to find archetypal malaria transmission patterns. Next, mechanistic models were run on a representative site from each archetype to assess intervention impact. Finally, these mechanistic results were reprojected onto each pixel to generate full maps of intervention impact. The example configuration used ERA5 and Malaria Atlas Project covariates, singular value decomposition, k-means clustering, and the Institute for Disease Modeling's EMOD model to explore a range of three-year malaria interventions primarily focused on vector control and case management. RESULTS: Rainfall, temperature, and mosquito abundance layers were clustered into ten transmission archetypes with distinct properties. Example intervention impact curves and maps highlighted archetype-specific variation in efficacy of vector control interventions. A sensitivity analysis showed that the procedure for selecting representative sites to simulate worked well in all but one archetype. CONCLUSION: This paper introduces a novel methodology which combines the richness of spatiotemporal mapping with the rigor of mechanistic modeling to create a multi-purpose infrastructure for answering a broad range of important questions in the malaria policy space. It is flexible and adaptable to a range of input covariates, mechanistic models, and mapping strategies and can be adapted to the modelers' setting of choice.


Subject(s)
Malaria , Animals , Humans , Malaria/prevention & control , Mosquito Control/methods
6.
Trop Med Infect Dis ; 8(4)2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37104342

ABSTRACT

The COVID-19 pandemic has led to far-reaching disruptions to health systems, including preventative and curative services for malaria. The aim of this study was to estimate the magnitude of disruptions in malaria case management in sub-Saharan Africa and their impact on malaria burden during the COVID-19 pandemic. We used survey data collected by the World Health Organization, in which individual country stakeholders reported on the extent of disruptions to malaria diagnosis and treatment. The relative disruption values were then applied to estimates of antimalarial treatment rates and used as inputs to an established spatiotemporal Bayesian geostatistical framework to generate annual malaria burden estimates with case management disruptions. This enabled an estimation of the additional malaria burden attributable to pandemic-related impacts on treatment rates in 2020 and 2021. Our analysis found that disruptions in access to antimalarial treatment in sub-Saharan Africa likely resulted in approximately 5.9 (4.4-7.2 95% CI) million more malaria cases and 76 (20-132) thousand additional deaths in the 2020-2021 period within the study region, equivalent to approximately 1.2% (0.3-2.1 95% CI) greater clinical incidence of malaria and 8.1% (2.1-14.1 95% CI) greater malaria mortality than expected in the absence of the disruptions to malaria case management. The available evidence suggests that access to antimalarials was disrupted to a significant degree and should be considered an area of focus to avoid further escalations in malaria morbidity and mortality. The results from this analysis were used to estimate cases and deaths in the World Malaria Report 2022 during the pandemic years.

7.
Spat Spatiotemporal Epidemiol ; 41: 100357, 2022 06.
Article in English | MEDLINE | ID: mdl-35691633

ABSTRACT

Maps of disease burden are a core tool needed for the control and elimination of malaria. Reliable routine surveillance data of malaria incidence, typically aggregated to administrative units, is becoming more widely available. Disaggregation regression is an important model framework for estimating high resolution risk maps from aggregated data. However, the aggregation of incidence over large, heterogeneous areas means that these data are underpowered for estimating complex, non-linear models. In contrast, prevalence point-surveys are directly linked to local environmental conditions but are not common in many areas of the world. Here, we train multiple non-linear, machine learning models on Plasmodium falciparum prevalence point-surveys. We then ensemble the predictions from these machine learning models with a disaggregation regression model that uses aggregated malaria incidences as response data. We find that using a disaggregation regression model to combine predictions from machine learning models improves model accuracy relative to a baseline model.


Subject(s)
Malaria, Falciparum , Malaria , Humans , Incidence , Malaria/epidemiology , Malaria, Falciparum/epidemiology , Nonlinear Dynamics , Prevalence
8.
Proc Biol Sci ; 289(1972): 20220089, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35414241

ABSTRACT

Understanding the temporal dynamics of mosquito populations underlying vector-borne disease transmission is key to optimizing control strategies. Many questions remain surrounding the drivers of these dynamics and how they vary between species-questions rarely answerable from individual entomological studies (that typically focus on a single location or species). We develop a novel statistical framework enabling identification and classification of time series with similar temporal properties, and use this framework to systematically explore variation in population dynamics and seasonality in anopheline mosquito time series catch data spanning seven species, 40 years and 117 locations across mainland India. Our analyses reveal pronounced variation in dynamics across locations and between species in the extent of seasonality and timing of seasonal peaks. However, we show that these diverse dynamics can be clustered into four 'dynamical archetypes', each characterized by distinct temporal properties and associated with a largely unique set of environmental factors. Our results highlight that a range of environmental factors including rainfall, temperature, proximity to static water bodies and patterns of land use (particularly urbanicity) shape the dynamics and seasonality of mosquito populations, and provide a generically applicable framework to better identify and understand patterns of seasonal variation in vectors relevant to public health.


Subject(s)
Anopheles , Animals , Climate , Mosquito Control/methods , Mosquito Vectors , Population Dynamics , Seasons
9.
BMJ Glob Health ; 7(2)2022 02.
Article in English | MEDLINE | ID: mdl-35217531

ABSTRACT

BACKGROUND: HIV, tuberculosis (TB) and malaria are the three most important infectious diseases in Ethiopia, and sub-Saharan Africa. Understanding the spatial codistribution of these diseases is critical for designing geographically targeted and integrated disease control programmes. This study investigated the spatial overlap and drivers of HIV, TB and malaria prevalence in Ethiopia. METHODS: HIV, TB and malaria data were obtained from different nationwide prevalence surveys, and geospatial covariates were obtained from publicly available sources. A Bayesian model-based geostatistical framework was applied to each survey leveraging the strength of high-resolution spatial covariates to predict continuous disease-specific prevalence surfaces and their codistribution. RESULTS: The national prevalence was 1.54% (95% CI 1.40 to 1.70) for HIV, 0.39% (95% CI 0.34 to 0.45) for TB and 1.1% (95%CI 0.95 to 1.32) for malaria. Substantial subnational variation was predicted with the highest HIV prevalence estimated in Gambela (4.52%), Addis Ababa (3.52%) and Dire Dawa (2.67%) regions. TB prevalence was highest in Dire Dawa (0.96%) and Gambela (0.88%), while malaria was highest in Gambela (6.1%) and Benishangul-Gumuz (3.8%). Spatial overlap of their prevalence was observed in some parts of the country, mainly Gambela region. Spatial distribution of the diseases was significantly associated with healthcare access, demographic, and climatic factors. CONCLUSIONS: The national distribution of HIV, TB and malaria was highly focal in Ethiopia, with substantial variation at subnational and local levels. Spatial distribution of the diseases was significantly associated with healthcare access, demographic and climatic factors. Spatial overlap of HIV, TB and malaria prevalence was observed in some parts of the country. Integrated control programmes for these diseases should be targeted to these areas with high levels of co-endemicity.


Subject(s)
HIV Infections , Malaria , Tuberculosis , Bayes Theorem , Ethiopia/epidemiology , HIV Infections/epidemiology , Humans , Malaria/epidemiology , Tuberculosis/epidemiology
10.
Stat Med ; 41(1): 1-16, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34658042

ABSTRACT

Disaggregation regression has become an important tool in spatial disease mapping for making fine-scale predictions of disease risk from aggregated response data. By including high resolution covariate information and modeling the data generating process on a fine scale, it is hoped that these models can accurately learn the relationships between covariates and response at a fine spatial scale. However, validating these high resolution predictions can be a challenge, as often there is no data observed at this spatial scale. In this study, disaggregation regression was performed on simulated data in various settings and the resulting fine-scale predictions are compared to the simulated ground truth. Performance was investigated with varying numbers of data points, sizes of aggregated areas and levels of model misspecification. The effectiveness of cross validation on the aggregate level as a measure of fine-scale predictive performance was also investigated. Predictive performance improved as the number of observations increased and as the size of the aggregated areas decreased. When the model was well-specified, fine-scale predictions were accurate even with small numbers of observations and large aggregated areas. Under model misspecification predictive performance was significantly worse for large aggregated areas but remained high when response data was aggregated over smaller regions. Cross-validation correlation on the aggregate level was a moderately good predictor of fine-scale predictive performance. While these simulations are unlikely to capture the nuances of real-life response data, this study gives insight into the effectiveness of disaggregation regression in different contexts.


Subject(s)
Computer Simulation , Humans
11.
PLoS Med ; 18(6): e1003614, 2021 06.
Article in English | MEDLINE | ID: mdl-34061843

ABSTRACT

BACKGROUND: In 2017, an estimated 14 million cases of Plasmodium vivax malaria were reported from Asia, Central and South America, and the Horn of Africa. The clinical burden of vivax malaria is largely driven by its ability to form dormant liver stages (hypnozoites) that can reactivate to cause recurrent episodes of malaria. Elimination of both the blood and liver stages of the parasites ("radical cure") is required to achieve a sustained clinical response and prevent ongoing transmission of the parasite. Novel treatment options and point-of-care diagnostics are now available to ensure that radical cure can be administered safely and effectively. We quantified the global economic cost of vivax malaria and estimated the potential cost benefit of a policy of radical cure after testing patients for glucose-6-phosphate dehydrogenase (G6PD) deficiency. METHODS AND FINDINGS: Estimates of the healthcare provider and household costs due to vivax malaria were collated and combined with national case estimates for 44 endemic countries in 2017. These provider and household costs were compared with those that would be incurred under 2 scenarios for radical cure following G6PD screening: (1) complete adherence following daily supervised primaquine therapy and (2) unsupervised treatment with an assumed 40% effectiveness. A probabilistic sensitivity analysis generated credible intervals (CrIs) for the estimates. Globally, the annual cost of vivax malaria was US$359 million (95% CrI: US$222 to 563 million), attributable to 14.2 million cases of vivax malaria in 2017. From a societal perspective, adopting a policy of G6PD deficiency screening and supervision of primaquine to all eligible patients would prevent 6.1 million cases and reduce the global cost of vivax malaria to US$266 million (95% CrI: US$161 to 415 million), although healthcare provider costs would increase by US$39 million. If perfect adherence could be achieved with a single visit, then the global cost would fall further to US$225 million, equivalent to $135 million in cost savings from the baseline global costs. A policy of unsupervised primaquine reduced the cost to US$342 million (95% CrI: US$209 to 532 million) while preventing 2.1 million cases. Limitations of the study include partial availability of country-level cost data and parameter uncertainty for the proportion of patients prescribed primaquine, patient adherence to a full course of primaquine, and effectiveness of primaquine when unsupervised. CONCLUSIONS: Our modelling study highlights a substantial global economic burden of vivax malaria that could be reduced through investment in safe and effective radical cure achieved by routine screening for G6PD deficiency and supervision of treatment. Novel, low-cost interventions for improving adherence to primaquine to ensure effective radical cure and widespread access to screening for G6PD deficiency will be critical to achieving the timely global elimination of P. vivax.


Subject(s)
Antimalarials/economics , Antimalarials/therapeutic use , Drug Costs , Global Health/economics , Malaria, Vivax/drug therapy , Malaria, Vivax/economics , Primaquine/economics , Primaquine/therapeutic use , Adolescent , Adult , Antimalarials/adverse effects , Child , Child, Preschool , Clinical Decision-Making , Cost Savings , Cost-Benefit Analysis , Directly Observed Therapy , Female , Genetic Testing/economics , Glucosephosphate Dehydrogenase Deficiency/blood , Glucosephosphate Dehydrogenase Deficiency/diagnosis , Glucosephosphate Dehydrogenase Deficiency/economics , Glucosephosphate Dehydrogenase Deficiency/genetics , Health Expenditures , Hemolysis/drug effects , Humans , Incidence , Infant , Infant, Newborn , Malaria, Vivax/epidemiology , Male , Medication Adherence , Models, Economic , Patient Selection , Primaquine/adverse effects , Remission Induction , Treatment Outcome , Young Adult
12.
Article in English | MEDLINE | ID: mdl-34067393

ABSTRACT

Malaria in Bhutan has fallen significantly over the last decade. As Bhutan attempts to eliminate malaria in 2022, this study aimed to characterize the space-time clustering of malaria from 2010 to 2019. Malaria data were obtained from the Bhutan Vector-Borne Disease Control Program data repository. Spatial and space-time cluster analyses of Plasmodium falciparum and Plasmodium vivax cases were conducted at the sub-district level from 2010 to 2019 using Kulldorff's space-time scan statistic. A total of 768 confirmed malaria cases, including 454 (59%) P. vivax cases, were reported in Bhutan during the study period. Significant temporal clusters of cases caused by both species were identified between April and September. The most likely spatial clusters were detected in the central part of Bhutan throughout the study period. The most likely space-time cluster was in Sarpang District and neighboring districts between January 2010 to June 2012 for cases of infection with both species. The most likely cluster for P. falciparum infection had a radius of 50.4 km and included 26 sub-districts with a relative risk (RR) of 32.7. The most likely cluster for P. vivax infection had a radius of 33.6 km with 11 sub-districts and RR of 27.7. Three secondary space-time clusters were detected in other parts of Bhutan. Spatial and space-time cluster analysis identified high-risk areas and periods for both P. vivax and P. falciparum malaria. Both malaria types showed significant spatial and spatiotemporal variations. Operational research to understand the drivers of residual transmission in hotspot sub-districts will help to overcome the final challenges of malaria elimination in Bhutan.


Subject(s)
Malaria, Falciparum , Malaria, Vivax , Malaria , Bhutan/epidemiology , Humans , Incidence , Malaria/epidemiology , Malaria, Falciparum/epidemiology , Malaria, Vivax/epidemiology , Space-Time Clustering
13.
Nat Commun ; 12(1): 3589, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34117240

ABSTRACT

Insecticide-treated nets (ITNs) are one of the most widespread and impactful malaria interventions in Africa, yet a spatially-resolved time series of ITN coverage has never been published. Using data from multiple sources, we generate high-resolution maps of ITN access, use, and nets-per-capita annually from 2000 to 2020 across the 40 highest-burden African countries. Our findings support several existing hypotheses: that use is high among those with access, that nets are discarded more quickly than official policy presumes, and that effectively distributing nets grows more difficult as coverage increases. The primary driving factors behind these findings are most likely strong cultural and social messaging around the importance of net use, low physical net durability, and a mixture of inherent commodity distribution challenges and less-than-optimal net allocation policies, respectively. These results can inform both policy decisions and downstream malaria analyses.


Subject(s)
Benchmarking/methods , Insecticide-Treated Bednets , Insecticides , Malaria/prevention & control , Africa , Communicable Disease Control/methods , Computational Biology , Humans , Life Style , Malaria/epidemiology , Mosquito Control/methods
14.
Elife ; 102021 06 01.
Article in English | MEDLINE | ID: mdl-34058123

ABSTRACT

Towards the goal of malaria elimination on Hispaniola, the National Malaria Control Program of Haiti and its international partner organisations are conducting a campaign of interventions targeted to high-risk communities prioritised through evidence-based planning. Here we present a key piece of this planning: an up-to-date, fine-scale endemicity map and seasonality profile for Haiti informed by monthly case counts from 771 health facilities reporting from across the country throughout the 6-year period from January 2014 to December 2019. To this end, a novel hierarchical Bayesian modelling framework was developed in which a latent, pixel-level incidence surface with spatio-temporal innovations is linked to the observed case data via a flexible catchment sub-model designed to account for the absence of data on case household locations. These maps have focussed the delivery of indoor residual spraying and focal mass drug administration in the Grand'Anse Department in South-Western Haiti.


Subject(s)
Endemic Diseases , Malaria/epidemiology , Seasons , Antimalarials/therapeutic use , Bayes Theorem , Catchment Area, Health , Endemic Diseases/prevention & control , Haiti/epidemiology , Humans , Incidence , Malaria/diagnosis , Malaria/prevention & control , Models, Statistical , Mosquito Control , Spatio-Temporal Analysis , Time Factors
15.
Lancet Infect Dis ; 21(1): 59-69, 2021 01.
Article in English | MEDLINE | ID: mdl-32971006

ABSTRACT

BACKGROUND: Substantial progress has been made in reducing the burden of malaria in Africa since 2000, but those gains could be jeopardised if the COVID-19 pandemic affects the availability of key malaria control interventions. The aim of this study was to evaluate plausible effects on malaria incidence and mortality under different levels of disruption to malaria control. METHODS: Using an established set of spatiotemporal Bayesian geostatistical models, we generated geospatial estimates across malaria-endemic African countries of the clinical case incidence and mortality of malaria, incorporating an updated database of parasite rate surveys, insecticide-treated net (ITN) coverage, and effective treatment rates. We established a baseline estimate for the anticipated malaria burden in Africa in the absence of COVID-19-related disruptions, and repeated the analysis for nine hypothetical scenarios in which effective treatment with an antimalarial drug and distribution of ITNs (both through routine channels and mass campaigns) were reduced to varying extents. FINDINGS: We estimated 215·2 (95% uncertainty interval 143·7-311·6) million cases and 386·4 (307·8-497·8) thousand deaths across malaria-endemic African countries in 2020 in our baseline scenario of undisrupted intervention coverage. With greater reductions in access to effective antimalarial drug treatment, our model predicted increasing numbers of cases and deaths: 224·1 (148·7-326·8) million cases and 487·9 (385·3-634·6) thousand deaths with a 25% reduction in antimalarial drug coverage; 233·1 (153·7-342·5) million cases and 597·4 (468·0-784·4) thousand deaths with a 50% reduction; and 242·3 (158·7-358·8) million cases and 715·2 (556·4-947·9) thousand deaths with a 75% reduction. Halting planned 2020 ITN mass distribution campaigns and reducing routine ITN distributions by 25%-75% also increased malaria burden to a total of 230·5 (151·6-343·3) million cases and 411·7 (322·8-545·5) thousand deaths with a 25% reduction; 232·8 (152·3-345·9) million cases and 415·5 (324·3-549·4) thousand deaths with a 50% reduction; and 234·0 (152·9-348·4) million cases and 417·6 (325·5-553·1) thousand deaths with a 75% reduction. When ITN coverage and antimalarial drug coverage were synchronously reduced, malaria burden increased to 240·5 (156·5-358·2) million cases and 520·9 (404·1-691·9) thousand deaths with a 25% reduction; 251·0 (162·2-377·0) million cases and 640·2 (492·0-856·7) thousand deaths with a 50% reduction; and 261·6 (167·7-396·8) million cases and 768·6 (586·1-1038·7) thousand deaths with a 75% reduction. INTERPRETATION: Under pessimistic scenarios, COVID-19-related disruption to malaria control in Africa could almost double malaria mortality in 2020, and potentially lead to even greater increases in subsequent years. To avoid a reversal of two decades of progress against malaria, averting this public health disaster must remain an integrated priority alongside the response to COVID-19. FUNDING: Bill and Melinda Gates Foundation; Channel 7 Telethon Trust, Western Australia.


Subject(s)
COVID-19/epidemiology , Malaria/epidemiology , Malaria/mortality , SARS-CoV-2 , Africa/epidemiology , Antimalarials/therapeutic use , Bayes Theorem , Humans , Incidence , Insecticide-Treated Bednets , Malaria/drug therapy , Malaria/prevention & control , Models, Statistical , Morbidity
16.
Malar J ; 19(1): 374, 2020 Oct 20.
Article in English | MEDLINE | ID: mdl-33081784

ABSTRACT

BACKGROUND: Anti-malarial drugs play a critical role in reducing malaria morbidity and mortality, but their role is mediated by their effectiveness. Effectiveness is defined as the probability that an anti-malarial drug will successfully treat an individual infected with malaria parasites under routine health care delivery system. Anti-malarial drug effectiveness (AmE) is influenced by drug resistance, drug quality, health system quality, and patient adherence to drug use; its influence on malaria burden varies through space and time. METHODS: This study uses data from 232 efficacy trials comprised of 86,776 infected individuals to estimate the artemisinin-based and non-artemisinin-based AmE for treating falciparum malaria between 1991 and 2019. Bayesian spatiotemporal models were fitted and used to predict effectiveness at the pixel-level (5 km × 5 km). The median and interquartile ranges (IQR) of AmE are presented for all malaria-endemic countries. RESULTS: The global effectiveness of artemisinin-based drugs was 67.4% (IQR: 33.3-75.8), 70.1% (43.6-76.0) and 71.8% (46.9-76.4) for the 1991-2000, 2006-2010, and 2016-2019 periods, respectively. Countries in central Africa, a few in South America, and in the Asian region faced the challenge of lower effectiveness of artemisinin-based anti-malarials. However, improvements were seen after 2016, leaving only a few hotspots in Southeast Asia where resistance to artemisinin and partner drugs is currently problematic and in the central Africa where socio-demographic challenges limit effectiveness. The use of artemisinin-based combination therapy (ACT) with a competent partner drug and having multiple ACT as first-line treatment choice sustained high levels of effectiveness. High levels of access to healthcare, human resource capacity, education, and proximity to cities were associated with increased effectiveness. Effectiveness of non-artemisinin-based drugs was much lower than that of artemisinin-based with no improvement over time: 52.3% (17.9-74.9) for 1991-2000 and 55.5% (27.1-73.4) for 2011-2015. Overall, AmE for artemisinin-based and non-artemisinin-based drugs were, respectively, 29.6 and 36% below clinical efficacy as measured in anti-malarial drug trials. CONCLUSIONS: This study provides evidence that health system performance, drug quality and patient adherence influence the effectiveness of anti-malarials used in treating uncomplicated falciparum malaria. These results provide guidance to countries' treatment practises and are critical inputs for malaria prevalence and incidence models used to estimate national level malaria burden.


Subject(s)
Antimalarials/therapeutic use , Artemisinins/therapeutic use , Drug Resistance , Malaria, Falciparum/prevention & control , Plasmodium falciparum/drug effects , Humans
17.
Sci Rep ; 10(1): 18129, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33093622

ABSTRACT

Malaria transmission in Madagascar is highly heterogeneous, exhibiting spatial, seasonal and long-term trends. Previous efforts to map malaria risk in Madagascar used prevalence data from Malaria Indicator Surveys. These cross-sectional surveys, conducted during the high transmission season most recently in 2013 and 2016, provide nationally representative prevalence data but cover relatively short time frames. Conversely, monthly case data are collected at health facilities but suffer from biases, including incomplete reporting and low rates of treatment seeking. We combined survey and case data to make monthly maps of prevalence between 2013 and 2016. Health facility catchment populations were estimated to produce incidence rates from the case data. Smoothed incidence surfaces, environmental and socioeconomic covariates, and survey data informed a Bayesian prevalence model, in which a flexible incidence-to-prevalence relationship was learned. Modelled spatial trends were consistent over time, with highest prevalence in the coastal regions and low prevalence in the highlands and desert south. Prevalence was lowest in 2014 and peaked in 2015 and seasonality was widely observed, including in some lower transmission regions. These trends highlight the utility of monthly prevalence estimates over the four year period. By combining survey and case data using this two-step modelling approach, we were able to take advantage of the relative strengths of each metric while accounting for potential bias in the case data. Similar modelling approaches combining large datasets of different malaria metrics may be applicable across sub-Saharan Africa.


Subject(s)
Malaria, Falciparum/diagnosis , Malaria, Falciparum/epidemiology , Plasmodium falciparum/isolation & purification , Population Surveillance , Spatio-Temporal Analysis , Bayes Theorem , Cross-Sectional Studies , Health Surveys , Humans , Madagascar/epidemiology , Malaria, Falciparum/parasitology , Prevalence
18.
PLoS Biol ; 18(6): e3000633, 2020 06.
Article in English | MEDLINE | ID: mdl-32584814

ABSTRACT

Mitigating the threat of insecticide resistance in African malaria vector populations requires comprehensive information about where resistance occurs, to what degree, and how this has changed over time. Estimating these trends is complicated by the sparse, heterogeneous distribution of observations of resistance phenotypes in field populations. We use 6,423 observations of the prevalence of resistance to the most important vector control insecticides to inform a Bayesian geostatistical ensemble modelling approach, generating fine-scale predictive maps of resistance phenotypes in mosquitoes from the Anopheles gambiae complex across Africa. Our models are informed by a suite of 111 predictor variables describing potential drivers of selection for resistance. Our maps show alarming increases in the prevalence of resistance to pyrethroids and DDT across sub-Saharan Africa from 2005 to 2017, with mean mortality following insecticide exposure declining from almost 100% to less than 30% in some areas, as well as substantial spatial variation in resistance trends.


Subject(s)
Insecticide Resistance , Malaria/parasitology , Mosquito Vectors/parasitology , Africa , DDT/toxicity , Insecticide Resistance/drug effects , Machine Learning , Mosquito Vectors/drug effects , Nitriles/toxicity , Phenotype , Prevalence , Pyrethrins/toxicity , Spatio-Temporal Analysis
19.
PLoS Med ; 17(3): e1003055, 2020 03.
Article in English | MEDLINE | ID: mdl-32203504

ABSTRACT

BACKGROUND: Housing is essential to human well-being but neglected in global health. Today, housing in Africa is rapidly improving alongside economic development, creating an urgent need to understand how these changes can benefit health. We hypothesised that improved housing is associated with better health in children living in sub-Saharan Africa (SSA). We conducted a cross-sectional analysis of housing conditions relative to a range of child health outcomes in SSA. METHODS AND FINDINGS: Cross-sectional data were analysed for 824,694 children surveyed in 54 Demographic and Health Surveys, 21 Malaria Indicator Surveys, and two AIDS Indicator Surveys conducted in 33 countries between 2001 and 2017 that measured malaria infection by microscopy or rapid diagnostic test (RDT), diarrhoea, acute respiratory infections (ARIs), stunting, wasting, underweight, or anaemia in children aged 0-5 years. The mean age of children was 2.5 years, and 49.7% were female. Housing was categorised into a binary variable based on a United Nations definition comparing improved housing (with improved drinking water, improved sanitation, sufficient living area, and finished building materials) versus unimproved housing (all other houses). Associations between house type and child health outcomes were determined using conditional logistic regression within surveys, adjusting for prespecified covariables including age, sex, household wealth, insecticide-treated bed net use, and vaccination status. Individual survey odds ratios (ORs) were pooled using random-effects meta-analysis. Across surveys, improved housing was associated with 8%-18% lower odds of all outcomes except ARI (malaria infection by microscopy: adjusted OR [aOR] 0.88, 95% confidence intervals [CIs] 0.80-0.97, p = 0.01; malaria infection by RDT: aOR 0.82, 95% CI 0.77-0.88, p < 0.001; diarrhoea: aOR 0.92, 95% CI 0.88-0.97, p = 0.001; ARI: aOR 0.96, 95% CI 0.87-1.07, p = 0.49; stunting: aOR 0.83, 95% CI 0.77-0.88, p < 0.001; wasting: aOR 0.90, 95% CI 0.83-0.99, p = 0.03; underweight: aOR 0.85, 95% CI 0.80-0.90, p < 0.001; any anaemia: aOR 0.87, 95% CI 0.82-0.92, p < 0.001; severe anaemia: aOR 0.89, 95% CI 0.84-0.95, p < 0.001). In comparison, insecticide-treated net use was associated with 16%-17% lower odds of malaria infection (microscopy: aOR 0.83, 95% CI 0.78-0.88, p < 0.001; RDT: aOR 0.84, 95% CI 0.79-0.88, p < 0.001). Drinking water source and sanitation facility alone were not associated with diarrhoea. The main study limitations are the use of self-reported diarrhoea and ARI, as well as potential residual confounding by socioeconomic position, despite adjustments for household wealth and education. CONCLUSIONS: In this study, we observed that poor housing, which includes inadequate drinking water and sanitation facility, is associated with health outcomes known to increase child mortality in SSA. Improvements to housing may be protective against a number of important childhood infectious diseases as well as poor growth outcomes, with major potential to improve children's health and survival across SSA.


Subject(s)
Anemia/epidemiology , Child Health , Child Nutrition Disorders/epidemiology , Diarrhea/epidemiology , Housing , Malaria/epidemiology , Social Determinants of Health , Africa South of the Sahara/epidemiology , Age Factors , Anemia/diagnosis , Anemia/mortality , Anemia/prevention & control , Child Nutrition Disorders/diagnosis , Child Nutrition Disorders/mortality , Child Nutrition Disorders/prevention & control , Child, Preschool , Cross-Sectional Studies , Diarrhea/diagnosis , Diarrhea/mortality , Diarrhea/prevention & control , Drinking Water , Female , Health Status , Health Surveys , Humans , Infant , Infant, Newborn , Insecticide-Treated Bednets , Malaria/diagnosis , Malaria/mortality , Malaria/prevention & control , Male , Prospective Studies , Protective Factors , Risk Assessment , Risk Factors , Sanitation
20.
BMC Med ; 18(1): 26, 2020 02 10.
Article in English | MEDLINE | ID: mdl-32036785

ABSTRACT

BACKGROUND: Many malaria-endemic areas experience seasonal fluctuations in case incidence as Anopheles mosquito and Plasmodium parasite life cycles respond to changing environmental conditions. Identifying location-specific seasonality characteristics is useful for planning interventions. While most existing maps of malaria seasonality use fixed thresholds of rainfall, temperature, and/or vegetation indices to identify suitable transmission months, we construct a statistical modelling framework for characterising the seasonal patterns derived directly from monthly health facility data. METHODS: With data from 2669 of the 3247 health facilities in Madagascar, a spatiotemporal regression model was used to estimate seasonal patterns across the island. In the absence of catchment population estimates or the ability to aggregate to the district level, this focused on the monthly proportions of total annual cases by health facility level. The model was informed by dynamic environmental covariates known to directly influence seasonal malaria trends. To identify operationally relevant characteristics such as the transmission start months and associated uncertainty measures, an algorithm was developed and applied to model realisations. A seasonality index was used to incorporate burden information from household prevalence surveys and summarise 'how seasonal' locations are relative to their surroundings. RESULTS: Positive associations were detected between monthly case proportions and temporally lagged covariates of rainfall and temperature suitability. Consistent with the existing literature, model estimates indicate that while most parts of Madagascar experience peaks in malaria transmission near March-April, the eastern coast experiences an earlier peak around February. Transmission was estimated to start in southeast districts before southwest districts, suggesting that indoor residual spraying should be completed in the same order. In regions where the data suggested conflicting seasonal signals or two transmission seasons, estimates of seasonal features had larger deviations and therefore less certainty. CONCLUSIONS: Monthly health facility data can be used to establish seasonal patterns in malaria burden and augment the information provided by household prevalence surveys. The proposed modelling framework allows for evidence-based and cohesive inferences on location-specific seasonal characteristics. As health surveillance systems continue to improve, it is hoped that more of such data will be available to improve our understanding and planning of intervention strategies.


Subject(s)
Health Facilities/statistics & numerical data , Malaria/epidemiology , Data Analysis , Humans , Incidence , Madagascar , Seasons
SELECTION OF CITATIONS
SEARCH DETAIL
...