Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Nat Commun ; 11(1): 3799, 2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32732892

RESUMO

Plasmodium falciparum in pregnancy is a major cause of adverse pregnancy outcomes. We combine performance estimates of standard rapid diagnostic tests (RDT) from trials of intermittent screening and treatment in pregnancy (ISTp) with modelling to assess whether screening at antenatal visits improves upon current intermittent preventative therapy with sulphadoxine-pyrimethamine (IPTp-SP). We estimate that RDTs in primigravidae at first antenatal visit are substantially more sensitive than in non-pregnant adults (OR = 17.2, 95% Cr.I. 13.8-21.6), and that sensitivity declines in subsequent visits and with gravidity, likely driven by declining susceptibility to placental infection. Monthly ISTp with standard RDTs, even with highly effective drugs, is not superior to monthly IPTp-SP. However, a hybrid strategy, recently adopted in Tanzania, combining testing and treatment at first visit with IPTp-SP may offer benefit, especially in areas with high-grade SP resistance. Screening and treatment in the first trimester, when IPTp-SP is contraindicated, could substantially improve pregnancy outcomes.


Assuntos
Malária Falciparum/diagnóstico , Malária Falciparum/prevenção & controle , Programas de Rastreamento/métodos , Complicações Parasitárias na Gravidez/prevenção & controle , Cuidado Pré-Natal/métodos , Antimaláricos/uso terapêutico , Combinação de Medicamentos , Feminino , Política de Saúde , Humanos , Malária Falciparum/tratamento farmacológico , Testes de Sensibilidade Parasitária , Plasmodium falciparum/efeitos dos fármacos , Gravidez , Complicações Parasitárias na Gravidez/tratamento farmacológico , Primeiro Trimestre da Gravidez , Pirimetamina/uso terapêutico , Sulfadoxina/uso terapêutico , Tanzânia , Organização Mundial da Saúde
2.
Lancet Infect Dis ; 20(6): 669-677, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32240634

RESUMO

BACKGROUND: In the face of rapidly changing data, a range of case fatality ratio estimates for coronavirus disease 2019 (COVID-19) have been produced that differ substantially in magnitude. We aimed to provide robust estimates, accounting for censoring and ascertainment biases. METHODS: We collected individual-case data for patients who died from COVID-19 in Hubei, mainland China (reported by national and provincial health commissions to Feb 8, 2020), and for cases outside of mainland China (from government or ministry of health websites and media reports for 37 countries, as well as Hong Kong and Macau, until Feb 25, 2020). These individual-case data were used to estimate the time between onset of symptoms and outcome (death or discharge from hospital). We next obtained age-stratified estimates of the case fatality ratio by relating the aggregate distribution of cases to the observed cumulative deaths in China, assuming a constant attack rate by age and adjusting for demography and age-based and location-based under-ascertainment. We also estimated the case fatality ratio from individual line-list data on 1334 cases identified outside of mainland China. Using data on the prevalence of PCR-confirmed cases in international residents repatriated from China, we obtained age-stratified estimates of the infection fatality ratio. Furthermore, data on age-stratified severity in a subset of 3665 cases from China were used to estimate the proportion of infected individuals who are likely to require hospitalisation. FINDINGS: Using data on 24 deaths that occurred in mainland China and 165 recoveries outside of China, we estimated the mean duration from onset of symptoms to death to be 17·8 days (95% credible interval [CrI] 16·9-19·2) and to hospital discharge to be 24·7 days (22·9-28·1). In all laboratory confirmed and clinically diagnosed cases from mainland China (n=70 117), we estimated a crude case fatality ratio (adjusted for censoring) of 3·67% (95% CrI 3·56-3·80). However, after further adjusting for demography and under-ascertainment, we obtained a best estimate of the case fatality ratio in China of 1·38% (1·23-1·53), with substantially higher ratios in older age groups (0·32% [0·27-0·38] in those aged <60 years vs 6·4% [5·7-7·2] in those aged ≥60 years), up to 13·4% (11·2-15·9) in those aged 80 years or older. Estimates of case fatality ratio from international cases stratified by age were consistent with those from China (parametric estimate 1·4% [0·4-3·5] in those aged <60 years [n=360] and 4·5% [1·8-11·1] in those aged ≥60 years [n=151]). Our estimated overall infection fatality ratio for China was 0·66% (0·39-1·33), with an increasing profile with age. Similarly, estimates of the proportion of infected individuals likely to be hospitalised increased with age up to a maximum of 18·4% (11·0-37·6) in those aged 80 years or older. INTERPRETATION: These early estimates give an indication of the fatality ratio across the spectrum of COVID-19 disease and show a strong age gradient in risk of death. FUNDING: UK Medical Research Council.


Assuntos
Infecções por Coronavirus/mortalidade , Pandemias/estatística & dados numéricos , Pneumonia Viral/mortalidade , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Betacoronavirus , COVID-19 , Criança , Pré-Escolar , China/epidemiologia , Hospitalização/estatística & dados numéricos , Humanos , Incidência , Lactente , Recém-Nascido , Pessoa de Meia-Idade , Modelos Estatísticos , SARS-CoV-2 , Adulto Jovem
3.
Proc Natl Acad Sci U S A ; 116(30): 15086-15095, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31285346

RESUMO

The antimalarial efficacy of the most important vector control interventions-long-lasting insecticidal nets (LLINs) and indoor residual spraying (IRS)-primarily protect against mosquitoes' biting people when they are in bed and indoors. Mosquito bites taken outside of these times contribute to residual transmission which determines the maximum effectiveness of current malaria prevention. The likelihood mosquitoes feed outside the time of day when LLINs and IRS can protect people is poorly understood, and the proportion of bites received outdoors may be higher after prolonged vector control. A systematic review of mosquito and human behavior is used to quantify and estimate the public health impact of outdoor biting across Africa. On average 79% of bites by the major malaria vectors occur during the time when people are in bed. This estimate is substantially lower than previous predictions, with results suggesting a nearly 10% lower proportion of bites taken at the time when people are beneath LLINs since the year 2000. Across Africa, this higher outdoor transmission is predicted to result in an estimated 10.6 million additional malaria cases annually if universal LLIN and IRS coverage was achieved. Higher outdoor biting diminishes the cases of malaria averted by vector control. This reduction in LLIN effectiveness appears to be exacerbated in areas where mosquito populations are resistant to insecticides used in bed nets, but no association was found between physiological resistance and outdoor biting. Substantial spatial heterogeneity in mosquito biting behavior between communities could contribute to differences in effectiveness of malaria control across Africa.


Assuntos
Anopheles/fisiologia , Comportamento Alimentar/fisiologia , Mordeduras e Picadas de Insetos/epidemiologia , Malária Falciparum/epidemiologia , Modelos Estatísticos , África/epidemiologia , Animais , Feminino , Mordeduras e Picadas de Insetos/prevenção & controle , Inseticidas , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Masculino , Controle de Mosquitos/métodos , Mosquiteiros/provisão & distribuição , Fotoperíodo , Plasmodium falciparum/patogenicidade , Plasmodium falciparum/fisiologia , Risco , Análise Espaço-Temporal
4.
Nat Commun ; 9(1): 4982, 2018 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-30478327

RESUMO

Indoor residual spraying (IRS) is an important part of malaria control. There is a growing list of insecticide classes; pyrethroids remain the principal insecticide used in bednets but recently, novel non-pyrethroid IRS products, with contrasting impacts, have been introduced. There is an urgent need to better assess product efficacy to help decision makers choose effective and relevant tools for mosquito control. Here we use experimental hut trial data to characterise the entomological efficacy of widely-used, novel IRS insecticides. We quantify their impact against pyrethroid-resistant mosquitoes and use a Plasmodium falciparum transmission model to predict the public health impact of different IRS insecticides. We report that long-lasting IRS formulations substantially reduce malaria, though their benefit over cheaper, shorter-lived formulations depends on local factors including bednet use, seasonality, endemicity and pyrethroid resistance status of local mosquito populations. We provide a framework to help decision makers evaluate IRS product effectiveness.


Assuntos
Inseticidas/toxicidade , Plasmodium falciparum/efeitos dos fármacos , África , Animais , Culicidae/efeitos dos fármacos , Mosquiteiros Tratados com Inseticida , Malária/parasitologia , Saúde Pública , Piretrinas/toxicidade , Ensaios Clínicos Controlados Aleatórios como Assunto , Fatores de Tempo
5.
BMC Med ; 16(1): 109, 2018 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-30001708

RESUMO

BACKGROUND: The RTS,S/AS01 vaccine for Plasmodium falciparum malaria demonstrated moderate efficacy in 5-17-month-old children in phase 3 trials, and from 2018, the vaccine will be evaluated through a large-scale pilot implementation program. Work is ongoing to optimise this vaccine, with higher efficacy for a different schedule demonstrated in a phase 2a challenge study. The objective of our study was to investigate the population-level impact of a modified RTS,S/AS01 schedule and dose amount in order to inform the target product profile for a second-generation malaria vaccine. METHODS: We used a mathematical modelling approach as the basis for our study. We simulated the changing anti-circumsporozoite antibody titre following vaccination and related the titre to vaccine efficacy. We then implemented this efficacy profile within an individual-based model of malaria transmission. We compared initial efficacy, duration and dose timing, and evaluated the potential public health impact of a modified vaccine in children aged 5-17 months, measuring clinical cases averted in children younger than 5 years. RESULTS: In the first decade of delivery, initial efficacy was associated with a higher reduction in childhood clinical cases compared to vaccine duration. This effect was more pronounced in high transmission settings and was due to the efficacy benefit occurring in younger ages where disease burden is highest. However, the low initial efficacy and long duration schedule averted more cases across all age cohorts if a longer time horizon was considered. We observed an age-shifting effect due to the changing immunological profile in higher transmission settings, in scenarios where initial efficacy was higher, and the fourth dose administered earlier. CONCLUSIONS: Our findings indicate that, for an imperfect childhood malaria vaccine with suboptimal efficacy, it may be advantageous to prioritise initial efficacy over duration. We predict that a modified vaccine could outperform the current RTS,S/AS01, although fourth dose timing will affect the age group that derives the greatest benefit. Further, the outcome measure and timeframe over which a vaccine is assessed are important when prioritising vaccine elements. This study provides insight into the most important characteristics of a malaria vaccine for at-risk groups and shows how distinct vaccine properties translate to public health outcomes. These findings may be used to prioritise target product profile elements for second-generation childhood malaria vaccines.


Assuntos
Vacinas Antimaláricas/uso terapêutico , Malária/prevenção & controle , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Vacinas Antimaláricas/farmacologia , Fatores de Tempo
6.
PLoS Med ; 14(11): e1002448, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29161259

RESUMO

BACKGROUND: Although significant progress has been made in reducing malaria transmission globally in recent years, a large number of people remain at risk and hence the gains made are fragile. Funding lags well behind amounts needed to protect all those at risk and ongoing contributions from major donors, such as the President's Malaria Initiative (PMI), are vital to maintain progress and pursue further reductions in burden. We use a mathematical modelling approach to estimate the impact of PMI investments to date in reducing malaria burden and to explore the potential negative impact on malaria burden should a proposed 44% reduction in PMI funding occur. METHODS AND FINDINGS: We combined an established mathematical model of Plasmodium falciparum transmission dynamics with epidemiological, intervention, and PMI-financing data to estimate the contribution PMI has made to malaria control via funding for long-lasting insecticide treated nets (LLINs), indoor residual spraying (IRS), and artemisinin combination therapies (ACTs). We estimate that PMI has prevented 185 million (95% CrI: 138 million, 230 million) malaria cases and saved 940,049 (95% CrI: 545,228, 1.4 million) lives since 2005. If funding is maintained, PMI-funded interventions are estimated to avert a further 162 million (95% CrI: 116 million, 194 million) cases, saving a further 692,589 (95% CrI: 392,694, 955,653) lives between 2017 and 2020. With an estimate of US$94 (95% CrI: US$51, US$166) per Disability Adjusted Life Year (DALY) averted, PMI-funded interventions are highly cost-effective. We also demonstrate the further impact of this investment by reducing caseloads on health systems. If a 44% reduction in PMI funding were to occur, we predict that this loss of direct aid could result in an additional 67 million (95% CrI: 49 million, 82 million) cases and 290,649 (95% CrI: 167,208, 395,263) deaths between 2017 and 2020. We have not modelled indirect impacts of PMI funding (such as health systems strengthening) in this analysis. CONCLUSIONS: Our model estimates that PMI has played a significant role in reducing malaria cases and deaths since its inception. Reductions in funding to PMI could lead to large increases in the number of malaria cases and deaths, damaging global goals of malaria control and elimination.


Assuntos
Malária Falciparum/prevenção & controle , Plasmodium falciparum , Artemisininas/uso terapêutico , Feminino , Humanos , Inseticidas/uso terapêutico , Malária Falciparum/mortalidade , Modelos Biológicos , Mosquiteiros , Resultado do Tratamento
7.
Sci Rep ; 7(1): 7389, 2017 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-28785011

RESUMO

Intermittent preventive treatment (IPT) with sulphadoxine-pyrimethamine in vulnerable populations reduces malaria morbidity in Africa, but resistance mutations in the parasite dhps gene (combined with dhfr mutations) threaten its efficacy. We update a systematic review to map the prevalence of K540E and A581G mutations in 294 surveys of infected humans across Africa from 2004-present. Interpreting these data is complicated by multiclonal infections in humans, especially in high transmission areas. We extend statistical methods to estimate the frequency, i.e. the proportion of resistant clones in the parasite population at each location, and so standardise for varying transmission levels. Both K540E and A581G mutations increased in prevalence and frequency in 60% of areas after 2008, highlighting the need for ongoing surveillance. Resistance measures within countries were similar within 300 km, suggesting an appropriate spatial scale for surveillance. Spread of the mutations tended to accelerate once their prevalence exceeded 10% (prior to fixation). Frequencies of resistance in parasite populations are the same or lower than prevalence in humans, so more areas would be classified as likely to benefit from IPT if similar frequency thresholds were applied. We propose that the use of resistance frequencies as well as prevalence measures for policy decisions should be evaluated.


Assuntos
Di-Hidropteroato Sintase/genética , Resistência a Medicamentos , Malária Falciparum/epidemiologia , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Tetra-Hidrofolato Desidrogenase/genética , África/epidemiologia , Animais , Antimaláricos/farmacologia , Combinação de Medicamentos , Humanos , Malária Falciparum/parasitologia , Malária Falciparum/veterinária , Modelos Estatísticos , Mutação , Plasmodium falciparum/efeitos dos fármacos , Vigilância da População , Prevalência , Pirimetamina/farmacologia , Sulfadoxina/farmacologia
8.
Malar J ; 16(1): 341, 2017 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-28814310

RESUMO

BACKGROUND: Anti-malarial drugs are an important tool for malaria control and elimination. Alongside their direct benefit in the treatment of disease, drug use has a community-level effect, clearing the reservoir of infection and reducing onward transmission of the parasite. Different compounds potentially have different impacts on transmission-with some providing periods of prolonged chemoprophylaxis whilst others have greater transmission-blocking potential. The aim was to quantify the relative benefit of such properties for transmission reduction to inform target product profiles in the drug development process and choice of first-line anti-malarial treatment in different endemic settings. METHODS: A mathematical model of Plasmodium falciparum epidemiology was used to estimate the transmission reduction that can be achieved by using drugs of varying chemoprophylactic (protection for 3, 30 or 60 days) or transmission-blocking activity (blocking 79, 92 or 100% of total onward transmission). Simulations were conducted at low, medium or high transmission intensity (slide-prevalence in 2-10 year olds being 1, 10 or 40%, respectively), with drugs administered either via case management or mass drug administration (MDA). RESULTS: Transmission reductions depend strongly on deployment strategy, treatment coverage and endemicity level. Transmission-blocking was most effective at low endemicity, whereas chemoprophylaxis was most useful at high endemicity levels. Increasing the duration of protection as much as possible was beneficial. Increasing transmission-blocking activity from the level of ACT to a 100% transmission-blocking drug (close to the effect estimated for ACT combined with primaquine) produced moderate impact but was not as effective as increasing the duration of protection in medium-to-high transmission settings (slide prevalence 10-40%). Combining both good transmission-blocking activity (e.g. as achieved by ACT or ACT + primaquine) and a long duration of protection (30 days or more, such as provided by piperaquine or mefloquine) within a drug regimen can substantially increase impact compared with drug regimens with only one of these properties in medium to high transmission areas (slide-prevalence in 2-10 year olds ~10 to 40%). These results applied whether the anti-malarials were used for case management or for MDA. DISCUSSION: These results emphasise the importance of increasing access to treatment for routine case management, and the potential value of choosing first-line anti-malarial treatment policies according to local malaria epidemiology to maximise impact on transmission. There is no indication that the optimal drug choice should differ between delivery via case management or MDA.


Assuntos
Antimaláricos/uso terapêutico , Administração de Caso/estatística & dados numéricos , Quimioterapia Combinada/estatística & dados numéricos , Malária Falciparum/prevenção & controle , Administração Massiva de Medicamentos/estatística & dados numéricos , Plasmodium falciparum/efeitos dos fármacos , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Modelos Teóricos
9.
BMJ Glob Health ; 2(1): e000090, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28588994

RESUMO

OBJECTIVES: To evaluate the relative cost-effectiveness of introducing the RTS,S malaria vaccine in sub-Saharan Africa compared with further scale-up of existing interventions. DESIGN: A mathematical modelling and cost-effectiveness study. SETTING: Sub-Saharan Africa. PARTICIPANTS: People of all ages. INTERVENTIONS: The analysis considers the introduction and scale-up of the RTS,S malaria vaccine and the scale-up of long-lasting insecticide-treated bed nets (LLINs), indoor residual spraying (IRS) and seasonal malaria chemoprevention (SMC). MAIN OUTCOME MEASURE: The number of Plasmodium falciparum cases averted in all age groups over a 10-year period. RESULTS: Assuming access to treatment remains constant, increasing coverage of LLINs was consistently the most cost-effective intervention across a range of transmission settings and was found to occur early in the cost-effectiveness scale-up pathway. IRS, RTS,S and SMC entered the cost-effective pathway once LLIN coverage had been maximised. If non-linear production functions are included to capture the cost of reaching very high coverage, the resulting pathways become more complex and result in selection of multiple interventions. CONCLUSIONS: RTS,S was consistently implemented later in the cost-effectiveness pathway than the LLINs, IRS and SMC but was still of value as a fourth intervention in many settings to reduce burden to the levels set out in the international goals.

10.
PLoS Pathog ; 13(1): e1006108, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28081253

RESUMO

Over a century since Ronald Ross discovered that malaria is caused by the bite of an infectious mosquito it is still unclear how the number of parasites injected influences disease transmission. Currently it is assumed that all mosquitoes with salivary gland sporozoites are equally infectious irrespective of the number of parasites they harbour, though this has never been rigorously tested. Here we analyse >1000 experimental infections of humans and mice and demonstrate a dose-dependency for probability of infection and the length of the host pre-patent period. Mosquitoes with a higher numbers of sporozoites in their salivary glands following blood-feeding are more likely to have caused infection (and have done so quicker) than mosquitoes with fewer parasites. A similar dose response for the probability of infection was seen for humans given a pre-erythrocytic vaccine candidate targeting circumsporozoite protein (CSP), and in mice with and without transfusion of anti-CSP antibodies. These interventions prevented infection more efficiently from bites made by mosquitoes with fewer parasites. The importance of parasite number has widespread implications across malariology, ranging from our basic understanding of the parasite, how vaccines are evaluated and the way in which transmission should be measured in the field. It also provides direct evidence for why the only registered malaria vaccine RTS,S was partially effective in recent clinical trials.


Assuntos
Anopheles/parasitologia , Insetos Vetores/parasitologia , Vacinas Antimaláricas/administração & dosagem , Malária/prevenção & controle , Plasmodium/imunologia , Animais , Anticorpos Antiprotozoários , Modelos Animais de Doenças , Humanos , Malária/parasitologia , Malária/transmissão , Camundongos , Plasmodium/crescimento & desenvolvimento , Dinâmica Populacional , Proteínas de Protozoários/imunologia , Glândulas Salivares/parasitologia , Esporozoítos/imunologia , Vacinação
11.
Elife ; 52016 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-27547988

RESUMO

Long lasting pyrethroid treated bednets are the most important tool for preventing malaria. Pyrethroid resistant Anopheline mosquitoes are now ubiquitous in Africa, though the public health impact remains unclear, impeding the deployment of more expensive nets. Meta-analyses of bioassay studies and experimental hut trials are used to characterise how pyrethroid resistance changes the efficacy of standard bednets, and those containing the synergist piperonyl butoxide (PBO), and assess its impact on malaria control. New bednets provide substantial personal protection until high levels of resistance, though protection may wane faster against more resistant mosquito populations as nets age. Transmission dynamics models indicate that even low levels of resistance would increase the incidence of malaria due to reduced mosquito mortality and lower overall community protection over the life-time of the net. Switching to PBO bednets could avert up to 0.5 clinical cases per person per year in some resistance scenarios.


Assuntos
Resistência a Inseticidas/fisiologia , Mosquiteiros Tratados com Inseticida/estatística & dados numéricos , Inseticidas , Malária/prevenção & controle , Modelos Estatísticos , Controle de Mosquitos/métodos , África , Animais , Anopheles/fisiologia , Bioensaio/estatística & dados numéricos , Humanos , Mosquiteiros Tratados com Inseticida/provisão & distribuição , Malária/transmissão , Mosquitos Vetores/fisiologia , Nitrilas , Permetrina , Sinergistas de Praguicidas/provisão & distribuição , Butóxido de Piperonila/provisão & distribuição , Piretrinas
12.
Malar J ; 15(1): 389, 2016 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-27456218

RESUMO

BACKGROUND: The basic reproduction number (R 0) is an important summary of the dynamics of an infectious disease. It is a threshold parameter: an infection can only invade a population if R 0 is greater than 1. However, a number of studies using simple models have suggested that for malaria, it is in theory possible for infection to persist indefinitely even if an intervention has reduced R 0 below 1. Such behaviour is known as a bistable equilibrium. Using two published mathematical models which have both been fitted to detailed, age-stratified data on multiple outcomes, the article investigates whether these more complex models behave in such a way, and hence whether a bistable equilibrium might be a real feature of Plasmodium falciparum malaria in Africa. RESULTS: With the best-fitting parameter values, neither model has a bistable state, because immunity reduces onwards infectiousness. The results imply that there is a threshold such that if interventions can reduce transmission so that R 0 is below 1 for long enough, then malaria will be locally eliminated. CONCLUSIONS: This means that calculations of the reduction in R 0 that interventions can achieve (the effect size) have a useful and straightforward interpretation, whereas if the theoretical possibility of a bistable equilibrium were the real behaviour, then such effect size calculations would not have a clear interpretation.


Assuntos
Número Básico de Reprodução , Erradicação de Doenças , Transmissão de Doença Infecciosa , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , África/epidemiologia , Humanos , Malária Falciparum/epidemiologia , Modelos Estatísticos
13.
Lancet Glob Health ; 4(7): e474-84, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27269393

RESUMO

BACKGROUND: Reducing the burden of malaria is a global priority, but financial constraints mean that available resources must be allocated rationally to maximise their effect. We aimed to develop a model to estimate the most efficient (ie, minimum cost) ordering of interventions to reduce malaria burden and transmission. We also aimed to estimate the efficiency of different spatial scales of implementation. METHODS: We combined a dynamic model capturing heterogeneity in malaria transmission across Africa with financial unit cost data for key malaria interventions. We combined estimates of patterns of malaria endemicity, seasonality in rainfall, and mosquito composition to map optimum packages of these interventions across Africa. Using non-linear optimisation methods, we examined how these optimum packages vary when control measures are deployed and assessed at national, subnational first administrative (provincial), or fine-scale (5 km(2) pixel) spatial scales. FINDINGS: The most efficient package in a given setting varies depending on whether disease reduction or elimination is the target. Long-lasting insecticide-treated nets are generally the most cost-effective first intervention to achieve either goal, with seasonal malaria chemoprevention or indoor residual spraying added second depending on seasonality and vector species. These interventions are estimated to reduce malaria transmission to less than one case per 1000 people per year in 43·4% (95% CI 40·0-49·0) of the population at risk in Africa. Adding three rounds of mass drug administration per year is estimated to increase this proportion to 90·9% (95% CI 86·9-94·6). Further optimisation can be achieved by targeting policies at the provincial level, achieving an estimated 32·1% (95% CI 29·6-34·5) cost saving relative to adopting country-wide policies. Nevertheless, we predict that only 26 (95% CI 22-29) of 41 countries could reduce transmission to these levels with these approaches. INTERPRETATION: These results highlight the cost-benefits of carefully tailoring malaria interventions to the ecological landscape of different areas. However, novel interventions are necessary if malaria eradication is to be achieved. FUNDING: Bill & Melinda Gates Foundation, UK Medical Research Council.


Assuntos
Malária Falciparum/epidemiologia , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Modelos Estatísticos , África , Antimaláricos/administração & dosagem , Antimaláricos/economia , Humanos , Mosquiteiros Tratados com Inseticida/economia , Controle de Mosquitos/métodos , Plasmodium falciparum/isolamento & purificação
14.
Malar J ; 15: 200, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27068686

RESUMO

BACKGROUND: As malaria prevalence declines in many parts of the world due to widescale control efforts and as drug-resistant parasites begin to emerge, a quantitative understanding of human movement is becoming increasingly relevant to malaria control. However, despite its importance, significant knowledge gaps remain regarding human movement, particularly in sub-Saharan Africa. METHODS: A quantitative survey of human movement patterns was conducted in four countries in sub-Saharan Africa: Mali, Burkina Faso, Zambia, and Tanzania, with three to five survey locations chosen in each country. Questions were included on demographic and trip details, malaria risk behaviour, children accompanying travellers, and mobile phone usage to enable phone signal data to be better correlated with movement. A total of 4352 individuals were interviewed and 6411 trips recorded. RESULTS: A cluster analysis of trips highlighted two distinct traveller groups of relevance to malaria transmission: women travelling with children (in all four countries) and youth workers (in Mali). Women travelling with children were more likely to travel to areas of relatively high malaria prevalence in Mali (OR = 4.46, 95% CI = 3.42-5.83), Burkina Faso (OR = 1.58, 95% CI = 1.23-1.58), Zambia (OR = 1.50, 95% CI = 1.20-1.89), and Tanzania (OR = 2.28, 95% CI = 1.71-3.05) compared to other travellers. They were also more likely to own bed nets in Burkina Faso (OR = 1.77, 95% CI = 1.25-2.53) and Zambia (OR = 1.74, 95% CI = 1.34 2.27), and less likely to own a mobile phone in Mali (OR = 0.50, 95% CI = 0.39-0.65), Burkina Faso (OR = 0.39, 95% CI = 0.30-0.52), and Zambia (OR = 0.60, 95% CI = 0.47-0.76). Malian youth workers were more likely to travel to areas of relatively high malaria prevalence (OR = 23, 95% CI = 17-31) and for longer durations (mean of 70 days cf 21 days, p < 0.001) compared to other travellers. CONCLUSIONS: Women travelling with children were a remarkably consistent traveller group across all four countries surveyed. They are expected to contribute greatly towards spatial malaria transmission because the children they travel with tend to have high parasite prevalence. Youth workers were a significant traveller group in Mali and are expected to contribute greatly to spatial malaria transmission because their movements correlate with seasonal rains and hence peak mosquito densities. Interventions aimed at interrupting spatial transmission of parasites should consider these traveller groups.


Assuntos
Malária/epidemiologia , Malária/transmissão , Viagem , Adolescente , Adulto , África Subsaariana/epidemiologia , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Recém-Nascido , Entrevistas como Assunto , Masculino , Pessoa de Meia-Idade , Movimento , Adulto Jovem
15.
Malar J ; 15: 10, 2016 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-26739092

RESUMO

BACKGROUND: Artemisinin and partner drug resistant malaria parasites have emerged in Southeast Asia. If resistance were to emerge in Africa it could have a devastating impact on malaria-related morbidity and mortality. This study estimates the potential impact of artemisinin and partner drug resistance on disease burden in Africa if it were to emerge. METHODS: Using data from Asia and Africa, five possible artemisinin and partner drug resistance scenarios are characterized. An individual-based malaria transmission model is used to estimate the impact of each resistance scenario on clinical incidence and parasite prevalence across Africa. Artemisinin resistance is characterized by slow parasite clearance and partner drug resistance is associated with late clinical failure or late parasitological failure. RESULTS: Scenarios with high levels of recrudescent infections resulted in far greater increases in clinical incidence compared to scenarios with high levels of slow parasite clearance. Across Africa, it is estimated that artemisinin and partner drug resistance at levels similar to those observed in Oddar Meanchey province in Cambodia could result in an additional 78 million cases over a 5 year period, a 7% increase in cases compared to a scenario with no resistance. A scenario with high levels of slow clearance but no recrudescence resulted in an additional 10 million additional cases over the same period. CONCLUSION: Artemisinin resistance is potentially a more pressing concern than partner drug resistance due to the lack of viable alternatives. However, it is predicted that a failing partner drug will result in greater increases in malaria cases and morbidity than would be observed from artemisinin resistance only.


Assuntos
Resistência a Medicamentos , Malária/tratamento farmacológico , África , África Subsaariana , Antimaláricos/uso terapêutico , Artemisininas , Ásia , Humanos
16.
Lancet Infect Dis ; 16(4): 465-72, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26809816

RESUMO

BACKGROUND: Rapid declines in malaria prevalence, cases, and deaths have been achieved globally during the past 15 years because of improved access to first-line treatment and vector control. We aimed to assess the intervention coverage needed to achieve further gains over the next 15 years. METHODS: We used a mathematical model of the transmission of Plasmodium falciparum malaria to explore the potential effect on case incidence and malaria mortality rates from 2015 to 2030 of five different intervention scenarios: remaining at the intervention coverage levels of 2011-13 (Sustain), for which coverage comprises vector control and access to treatment; two scenarios of increased coverage to 80% (Accelerate 1) and 90% (Accelerate 2), with a switch from quinine to injectable artesunate for management of severe disease and seasonal malaria chemoprevention where recommended for both Accelerate scenarios, and rectal artesunate for pre-referral treatment at the community level added to Accelerate 2; a near-term innovation scenario (Innovate), which included longer-lasting insecticidal nets and expansion of seasonal malaria chemoprevention; and a reduction in coverage to 2006-08 levels (Reverse). We did the model simulations at the first administrative level (ie, state or province) for the 80 countries with sustained stable malaria transmission in 2010, accounting for variations in baseline endemicity, seasonality in transmission, vector species, and existing intervention coverage. To calculate the cases and deaths averted, we compared the total number of each under the five scenarios between 2015 and 2030 with the predicted number in 2015, accounting for population growth. FINDINGS: With an increase to 80% coverage, we predicted a reduction in case incidence of 21% (95% credible intervals [CrI] 19-29) and a reduction in mortality rates of 40% (27-61) by 2030 compared with 2015 levels. Acceleration to 90% coverage and expansion of treatment at the community level was predicted to reduce case incidence by 59% (Crl 56-64) and mortality rates by 74% (67-82); with additional near-term innovation, incidence was predicted to decline by 74% (70-77) and mortality rates by 81% (76-87). These scenarios were predicted to lead to local elimination in 13 countries under the Accelerate 1 scenario, 20 under Accelerate 2, and 22 under Innovate by 2030, reducing the proportion of the population living in at-risk areas by 36% if elimination is defined at the first administrative unit. However, failing to maintain coverage levels of 2011-13 is predicted to raise case incidence by 76% (Crl 71-80) and mortality rates by 46% (39-51) by 2020. INTERPRETATION: Our findings show that decreases in malaria transmission and burden can be accelerated over the next 15 years if the coverage of key interventions is increased. FUNDING: UK Medical Research Council, UK Department for International Development, the Bill & Melinda Gates Foundation, the Swiss Development Agency, and the US Agency for International Development.


Assuntos
Antimaláricos/administração & dosagem , Artemisininas/administração & dosagem , Culicidae/virologia , Insetos Vetores/virologia , Malária Falciparum/prevenção & controle , Modelos Teóricos , Animais , Artesunato , Feminino , Geografia , Humanos , Incidência , Mosquiteiros Tratados com Inseticida , Malária Falciparum/epidemiologia , Malária Falciparum/transmissão , Controle de Mosquitos , Prevalência
17.
Lancet ; 387(10016): 367-375, 2016 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-26549466

RESUMO

BACKGROUND: The phase 3 trial of the RTS,S/AS01 malaria vaccine candidate showed modest efficacy of the vaccine against Plasmodium falciparum malaria, but was not powered to assess mortality endpoints. Impact projections and cost-effectiveness estimates for longer timeframes than the trial follow-up and across a range of settings are needed to inform policy recommendations. We aimed to assess the public health impact and cost-effectiveness of routine use of the RTS,S/AS01 vaccine in African settings. METHODS: We compared four malaria transmission models and their predictions to assess vaccine cost-effectiveness and impact. We used trial data for follow-up of 32 months or longer to parameterise vaccine protection in the group aged 5-17 months. Estimates of cases, deaths, and disability-adjusted life-years (DALYs) averted were calculated over a 15 year time horizon for a range of levels of Plasmodium falciparum parasite prevalence in 2-10 year olds (PfPR2-10; range 3-65%). We considered two vaccine schedules: three doses at ages 6, 7·5, and 9 months (three-dose schedule, 90% coverage) and including a fourth dose at age 27 months (four-dose schedule, 72% coverage). We estimated cost-effectiveness in the presence of existing malaria interventions for vaccine prices of US$2-10 per dose. FINDINGS: In regions with a PfPR2-10 of 10-65%, RTS,S/AS01 is predicted to avert a median of 93,940 (range 20,490-126,540) clinical cases and 394 (127-708) deaths for the three-dose schedule, or 116,480 (31,450-160,410) clinical cases and 484 (189-859) deaths for the four-dose schedule, per 100,000 fully vaccinated children. A positive impact is also predicted at a PfPR2-10 of 5-10%, but there is little impact at a prevalence of lower than 3%. At $5 per dose and a PfPR2-10 of 10-65%, we estimated a median incremental cost-effectiveness ratio compared with current interventions of $30 (range 18-211) per clinical case averted and $80 (44-279) per DALY averted for the three-dose schedule, and of $25 (16-222) and $87 (48-244), respectively, for the four-dose schedule. Higher ICERs were estimated at low PfPR2-10 levels. INTERPRETATION: We predict a significant public health impact and high cost-effectiveness of the RTS,S/AS01 vaccine across a wide range of settings. Decisions about implementation will need to consider levels of malaria burden, the cost-effectiveness and coverage of other malaria interventions, health priorities, financing, and the capacity of the health system to deliver the vaccine. FUNDING: PATH Malaria Vaccine Initiative; Bill & Melinda Gates Foundation; Global Good Fund; Medical Research Council; UK Department for International Development; GAVI, the Vaccine Alliance; WHO.


Assuntos
Vacinas Antimaláricas/economia , Malária Falciparum/prevenção & controle , Modelos Teóricos , Saúde Pública , África/epidemiologia , Ensaios Clínicos Fase III como Assunto , Análise Custo-Benefício , Humanos , Esquemas de Imunização , Lactente , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/economia , Malária Falciparum/epidemiologia , Estudos Multicêntricos como Assunto
18.
Malar J ; 14: 416, 2015 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-26492873

RESUMO

BACKGROUND: Malaria morbidity and mortality has declined in recent years in a number of settings. The ability to describe changes in malaria transmission associated with these declines is important in terms of assessing the potential effects of control interventions, and for monitoring and evaluation purposes. METHODS: Data from five cross-sectional surveys conducted in Farafenni and surrounding villages on the north bank of River Gambia between 1988 and 2011 were compiled. Antibody responses to MSP-119 were measured in samples from all surveys, data were normalized and expressed as seroprevalence and seroconversion rates (SCR) using different mathematical models. RESULTS: Results showed declines in serological metrics with seroprevalence in children aged one to 5 years dropping from 19 % (95 % CI 15-23 %) in 1988 to 1 % (0-2 %) in 2011 (p value for trend in proportions < 0.001) and the SCR dropping from 0.069 year(-1) (0.059-0.080) to 0.022 year(-1) (0.017-0.028; p = 0.004). The serological data were consistent with previously described drops in both parasite prevalence in children aged 1-5 years (62 %, 57-66 %, in 1988 to 2 %, 0-4 %, in 2011; p < 0.001), and all-cause under five mortality rates (37 per 1000 person-years, 34-41, in 1990 to 17, 15-19, in 2006; p = 0.059). CONCLUSIONS: This analysis shows accurate reconstruction of historical malaria transmission patterns in the Farafenni area using anti-malarial antibody responses. Demonstrating congruence between serological measures, and conventional clinical and parasitological measures suggests broader utility for serology in monitoring and evaluation of malaria transmission.


Assuntos
Anticorpos Antiprotozoários/sangue , Malária/epidemiologia , Plasmodium/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Estudos Transversais , Feminino , Gâmbia/epidemiologia , Humanos , Lactente , Recém-Nascido , Masculino , Proteína 1 de Superfície de Merozoito/imunologia , Pessoa de Meia-Idade , Estudos Soroepidemiológicos , Adulto Jovem
19.
Nat Commun ; 6: 8170, 2015 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-26348689

RESUMO

In many countries health system data remain too weak to accurately enumerate Plasmodium falciparum malaria cases. In response, cartographic approaches have been developed that link maps of infection prevalence with mathematical relationships to predict the incidence rate of clinical malaria. Microsimulation (or 'agent-based') models represent a powerful new paradigm for defining such relationships; however, differences in model structure and calibration data mean that no consensus yet exists on the optimal form for use in disease-burden estimation. Here we develop a Bayesian statistical procedure combining functional regression-based model emulation with Markov Chain Monte Carlo sampling to calibrate three selected microsimulation models against a purpose-built data set of age-structured prevalence and incidence counts. This allows the generation of ensemble forecasts of the prevalence-incidence relationship stratified by age, transmission seasonality, treatment level and exposure history, from which we predict accelerating returns on investments in large-scale intervention campaigns as transmission and prevalence are progressively reduced.


Assuntos
Malária Falciparum/epidemiologia , Modelos Estatísticos , Adolescente , Adulto , África/epidemiologia , Idoso , Idoso de 80 Anos ou mais , Teorema de Bayes , Criança , Pré-Escolar , Simulação por Computador , Humanos , Incidência , Lactente , Recém-Nascido , Malária Falciparum/transmissão , Cadeias de Markov , Pessoa de Meia-Idade , Método de Monte Carlo , Prevalência , Adulto Jovem
20.
Lancet Infect Dis ; 15(12): 1450-8, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26342424

RESUMO

BACKGROUND: The RTS,S/AS01 malaria vaccine targets the circumsporozoite protein, inducing antibodies associated with the prevention of Plasmodium falciparum infection. We assessed the association between anti-circumsporozoite antibody titres and the magnitude and duration of vaccine efficacy using data from a phase 3 trial done between 2009 and 2014. METHODS: Using data from 8922 African children aged 5-17 months and 6537 African infants aged 6-12 weeks at first vaccination, we analysed the determinants of immunogenicity after RTS,S/AS01 vaccination with or without a booster dose. We assessed the association between the incidence of clinical malaria and anti-circumsporozoite antibody titres using a model of anti-circumsporozoite antibody dynamics and the natural acquisition of protective immunity over time. FINDINGS: RTS,S/AS01-induced anti-circumsporozoite antibody titres were greater in children aged 5-17 months than in those aged 6-12 weeks. Pre-vaccination anti-circumsporozoite titres were associated with lower immunogenicity in children aged 6-12 weeks and higher immunogenicity in those aged 5-17 months. The immunogenicity of the booster dose was strongly associated with immunogenicity after primary vaccination. Anti-circumsporozoite titres wane according to a biphasic exponential distribution. In participants aged 5-17 months, the half-life of the short-lived component of the antibody response was 45 days (95% credible interval 42-48) and that of the long-lived component was 591 days (557-632). After primary vaccination 12% (11-13) of the response was estimated to be long-lived, rising to 30% (28-32%) after a booster dose. An anti-circumsporozoite antibody titre of 121 EU/mL (98-153) was estimated to prevent 50% of infections. Waning anti-circumsporozoite antibody titres predict the duration of efficacy against clinical malaria across different age categories and transmission intensities, and efficacy wanes more rapidly at higher transmission intensity. INTERPRETATION: Anti-circumsporozoite antibody titres are a surrogate of protection for the magnitude and duration of RTS,S/AS01 efficacy, with or without a booster dose, providing a valuable surrogate of effectiveness for new RTS,S formulations in the age groups considered. FUNDING: UK Medical Research Council.


Assuntos
Anticorpos Antiprotozoários/sangue , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/prevenção & controle , Plasmodium falciparum/efeitos dos fármacos , Proteínas de Protozoários/antagonistas & inibidores , Vacinação , Feminino , Gana , Humanos , Imunização Secundária , Incidência , Lactente , Quênia , Vacinas Antimaláricas/química , Vacinas Antimaláricas/imunologia , Malária Falciparum/sangue , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Masculino , Plasmodium falciparum/química , Plasmodium falciparum/imunologia , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Tanzânia , Resultado do Tratamento , Vacinas de Subunidades Antigênicas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...