Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-31932376

RESUMO

Antimalarial drugs have long half-lives, so clinical trials to monitor their efficacy require long periods of follow-up to capture drug failure that may become patent only weeks after treatment. Reinfections often occur during follow-up, so robust methods of distinguishing drug failures (recrudescence) from emerging new infections are needed to produce accurate failure rate estimates. Molecular correction aims to achieve this by comparing the genotype of a patient's pretreatment (initial) blood sample with that of any infection that occurs during follow-up, with matching genotypes indicating drug failure. We use an in silico approach to show that the widely used match-counting method of molecular correction with microsatellite markers is likely to be highly unreliable and may lead to gross under- or overestimates of the true failure rates, depending on the choice of matching criterion. A Bayesian algorithm for molecular correction was previously developed and utilized for analysis of in vivo efficacy trials. We validated this algorithm using in silico data and showed it had high specificity and generated accurate failure rate estimates. This conclusion was robust for multiple drugs, different levels of drug failure rates, different levels of transmission intensity in the study sites, and microsatellite genetic diversity. The Bayesian algorithm was inherently unable to accurately identify low-density recrudescence that occurred in a small number of patients, but this did not appear to compromise its utility as a highly effective molecular correction method for analyzing microsatellite genotypes. Strong consideration should be given to using Bayesian methodology to obtain accurate failure rate estimates during routine monitoring trials of antimalarial efficacy that use microsatellite markers.


Assuntos
Antimaláricos/uso terapêutico , Biologia Computacional/métodos , Malária Falciparum/tratamento farmacológico , Repetições de Microssatélites/genética , Plasmodium falciparum/efeitos dos fármacos , Algoritmos , Combinação Arteméter e Lumefantrina/uso terapêutico , Artesunato/uso terapêutico , Simulação por Computador , Interpretação Estatística de Dados , Feminino , Humanos , Masculino , Mefloquina/uso terapêutico , Plasmodium falciparum/genética , Reinfecção/genética , Reinfecção/parasitologia , Falha de Tratamento
2.
J Infect Dis ; 219(8): 1243-1253, 2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30517708

RESUMO

BACKGROUND: Standard treatment for severe malaria is with artesunate; patient survival in the 24 hours immediately posttreatment is the key objective. Clinical trials use clearance rates of circulating parasites as their clinical outcome, but the pathology of severe malaria is attributed primarily to noncirculating, sequestered, parasites, so there is a disconnect between existing clinical metrics and objectives. METHODS: We extend existing pharmacokinetic/pharmacodynamic modeling methods to simulate the treatment of 10000 patients with severe malaria and track the pathology caused by sequestered parasites. RESULTS: Our model recovered the clinical outcomes of existing studies (based on circulating parasites) and showed a "simplified" artesunate regimen was noninferior to the existing World Health Organization regimen across the patient population but resulted in worse outcomes in a subgroup of patients with infections clustered in early stages of the parasite life cycle. This same group of patients were extremely vulnerable to resistance emerging in parasite early ring stages. CONCLUSIONS: We quantify patient outcomes in a manner appropriate for severe malaria with a flexible framework that allows future researchers to implement their beliefs about underlying pathology. We highlight with some urgency the threat posed to treatment of severe malaria by artemisinin resistance in parasite early ring stages.


Assuntos
Antimaláricos/uso terapêutico , Artemisininas/uso terapêutico , Malária Falciparum/tratamento farmacológico , Doença Aguda , Antimaláricos/administração & dosagem , Artesunato/administração & dosagem , Artesunato/uso terapêutico , Resistência a Medicamentos , Humanos , Malária Falciparum/parasitologia , Malária Falciparum/patologia , Modelos Biológicos , Parasitemia/tratamento farmacológico , Parasitemia/parasitologia , Plasmodium falciparum/efeitos dos fármacos , Resultado do Tratamento
4.
PLoS Comput Biol ; 13(1): e1005327, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28095406

RESUMO

We develop a flexible, two-locus model for the spread of insecticide resistance applicable to mosquito species that transmit human diseases such as malaria. The model allows differential exposure of males and females, allows them to encounter high or low concentrations of insecticide, and allows selection pressures and dominance values to differ depending on the concentration of insecticide encountered. We demonstrate its application by investigating the relative merits of sequential use of insecticides versus their deployment as a mixture to minimise the spread of resistance. We recover previously published results as subsets of this model and conduct a sensitivity analysis over an extensive parameter space to identify what circumstances favour mixtures over sequences. Both strategies lasted more than 500 mosquito generations (or about 40 years) in 24% of runs, while in those runs where resistance had spread to high levels by 500 generations, 56% favoured sequential use and 44% favoured mixtures. Mixtures are favoured when insecticide effectiveness (their ability to kill homozygous susceptible mosquitoes) is high and exposure (the proportion of mosquitoes that encounter the insecticide) is low. If insecticides do not reliably kill homozygous sensitive genotypes, it is likely that sequential deployment will be a more robust strategy. Resistance to an insecticide always spreads slower if that insecticide is used in a mixture although this may be insufficient to outperform sequential use: for example, a mixture may last 5 years while the two insecticides deployed individually may last 3 and 4 years giving an overall 'lifespan' of 7 years for sequential use. We emphasise that this paper is primarily about designing and implementing a flexible modelling strategy to investigate the spread of insecticide resistance in vector populations and demonstrate how our model can identify vector control strategies most likely to minimise the spread of insecticide resistance.


Assuntos
Evolução Biológica , Culex/efeitos dos fármacos , Culex/genética , Resistência a Inseticidas/genética , Inseticidas/administração & dosagem , Malária/prevenção & controle , Animais , Simulação por Computador , Política de Saúde , Humanos , Malária/parasitologia , Modelos Genéticos
5.
Malar J ; 17(1): 80, 2018 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-29448925

RESUMO

BACKGROUND: Insecticide resistance threatens effective vector control, especially for mosquitoes and malaria. To manage resistance, recommended insecticide use strategies include mixtures, sequences and rotations. New insecticides are being developed and there is an opportunity to develop use strategies that limit the evolution of further resistance in the short term. A 2013 review of modelling and empirical studies of resistance points to the advantages of mixtures. However, there is limited recent, accessible modelling work addressing the evolution of resistance under different operational strategies. There is an opportunity to improve the level of mechanistic understanding within the operational community of how insecticide resistance can be expected to evolve in response to different strategies. This paper provides a concise, accessible description of a flexible model of the evolution of insecticide resistance. The model is used to develop a mechanistic picture of the evolution of insecticide resistance and how it is likely to respond to potential insecticide use strategies. The aim is to reach an audience unlikely to read a more detailed modelling paper. The model itself, as described here, represents two independent genes coding for resistance to two insecticides. This allows the representation of the use of insecticides in isolation, sequence and mixtures. RESULTS: The model is used to demonstrate the evolution of resistance under different scenarios and how this fits with intuitive reasoning about selection pressure. Using an insecticide in a mixture, relative to alone, always prompts slower evolution of resistance to that insecticide. However, when resistance to both insecticides is considered, resistance thresholds may be reached later for a sequence relative to a mixture. Increasing the ability of insecticides to kill susceptible mosquitoes (effectiveness), has the most influence on favouring a mixture over a sequence because one highly effective insecticide provides more protection to another in a mixture. CONCLUSIONS: The model offers an accessible description of the process of insecticide resistance evolution and how it is likely to respond to insecticide use. A simple online user-interface allowing further exploration is also provided. These tools can contribute to an improved discussion about operational decisions in insecticide resistance management.


Assuntos
Anopheles/efeitos dos fármacos , Resistência a Inseticidas/genética , Inseticidas/farmacologia , Controle de Mosquitos/métodos , Mosquitos Vetores/efeitos dos fármacos , Animais , Evolução Molecular , Malária/prevenção & controle , Modelos Genéticos
6.
Antimicrob Agents Chemother ; 60(5): 2747-56, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26902760

RESUMO

Pharmacological modeling of antiparasitic treatment based on a drug's pharmacokinetic and pharmacodynamic properties plays an increasingly important role in identifying optimal drug dosing regimens and predicting their potential impact on control and elimination programs. Conventional modeling of treatment relies on methods that do not distinguish between parasites at different developmental stages. This is problematic for malaria parasites, as their sensitivity to drugs varies substantially during their 48-h developmental cycle. We investigated four drug types (short or long half-lives with or without stage-specific killing) to quantify the accuracy of the standard methodology. The treatment dynamics of three drug types were well characterized with standard modeling. The exception were short-half-life drugs with stage-specific killing (i.e., artemisinins) because, depending on time of treatment, parasites might be in highly drug-sensitive stages or in much less sensitive stages. We describe how to bring such drugs into pharmacological modeling by including additional variation into the drug's maximal killing rate. Finally, we show that artemisinin kill rates may have been substantially overestimated in previous modeling studies because (i) the parasite reduction ratio (PRR) (generally estimated to be 10(4)) is based on observed changes in circulating parasite numbers, which generally overestimate the "true" PRR, which should include both circulating and sequestered parasites, and (ii) the third dose of artemisinin at 48 h targets exactly those stages initially hit at time zero, so it is incorrect to extrapolate the PRR measured over 48 h to predict the impact of doses at 48 h and later.


Assuntos
Antimaláricos/farmacocinética , Antimaláricos/uso terapêutico , Relação Dose-Resposta a Droga , Malária/tratamento farmacológico , Modelos Teóricos
7.
Malar J ; 15(1): 430, 2016 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-27557806

RESUMO

BACKGROUND: Haplotypes are important in anti-malarial drug resistance because genes encoding drug resistance may accumulate mutations at several codons in the same gene, each mutation increasing the level of drug resistance and, possibly, reducing the metabolic costs of previous mutation. Patients often have two or more haplotypes in their blood sample which may make it impossible to identify exactly which haplotypes they carry, and hence to measure the type and frequency of resistant haplotypes in the malaria population. RESULTS: This study presents two novel statistical methods expectation-maximization (EM) and Markov chain Monte Carlo (MCMC) algorithms to investigate this issue. The performance of the algorithms is evaluated on simulated datasets consisting of patient blood characterized by their multiplicity of infection (MOI) and malaria genotype. The datasets are generated using different resistance allele frequencies (RAF) at each single nucleotide polymorphisms (SNPs) and different limit of detection (LoD) of the SNPs and the MOI. The EM and the MCMC algorithm are validated and appear more accurate, faster and slightly less affected by LoD of the SNPs and the MOI compared to previous related statistical approaches. CONCLUSIONS: The EM and the MCMC algorithms perform well when analysing malaria genetic data obtained from infected human blood samples. The results are robust to genotyping errors caused by LoDs and function well even in the absence of MOI data on individual patients.


Assuntos
Coinfecção/epidemiologia , Coinfecção/parasitologia , Haplótipos , Malária/epidemiologia , Malária/parasitologia , Plasmodium/genética , Plasmodium/isolamento & purificação , Algoritmos , Bioestatística , Humanos , Cadeias de Markov , Plasmodium/classificação
8.
Antimicrob Agents Chemother ; 59(10): 6428-36, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26239987

RESUMO

Artemisinin-based combination therapies (ACTs) are currently the first-line drugs for treating uncomplicated falciparum malaria, the most deadly of the human malarias. Malaria parasite clearance rates estimated from patients' blood following ACT treatment have been widely adopted as a measure of drug effectiveness and as surveillance tools for detecting the presence of potential artemisinin resistance. This metric has not been investigated in detail, nor have its properties or potential shortcomings been identified. Herein, the pharmacology of drug treatment, parasite biology, and human immunity are combined to investigate the dynamics of parasite clearance following ACT. This approach parsimoniously recovers the principal clinical features and dynamics of clearance. Human immunity is the primary determinant of clearance rates, unless or until artemisinin killing has fallen to near-ineffective levels. Clearance rates are therefore highly insensitive metrics for surveillance that may lead to overconfidence, as even quite substantial reductions in drug sensitivity may not be detected as lower clearance rates. Equally serious is the use of clearance rates to quantify the impact of ACT regimen changes, as this strategy will plausibly miss even very substantial increases in drug effectiveness. In particular, the malaria community may be missing the opportunity to dramatically increase ACT effectiveness through regimen changes, particularly through a switch to twice-daily regimens and/or increases in artemisinin dosing levels. The malaria community therefore appears overreliant on a single metric of drug effectiveness, the parasite clearance rate, that has significant and serious shortcomings.


Assuntos
Antimaláricos/farmacologia , Artemisininas/farmacologia , Malária Falciparum/tratamento farmacológico , Modelos Estatísticos , Parasitemia/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Adulto , Antimaláricos/farmacocinética , Artemisininas/farmacocinética , Artesunato , Criança , Relação Dose-Resposta a Droga , Esquema de Medicação , Combinação de Medicamentos , Cálculos da Dosagem de Medicamento , Resistência a Medicamentos , Humanos , Imunidade Inata , Malária Falciparum/diagnóstico , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Mefloquina/farmacocinética , Mefloquina/farmacologia , Parasitemia/diagnóstico , Parasitemia/imunologia , Testes de Sensibilidade Parasitária , Plasmodium falciparum/fisiologia , Quinolinas/farmacocinética , Quinolinas/farmacologia , Resultado do Tratamento
9.
Antimicrob Agents Chemother ; 59(10): 6419-27, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26239993

RESUMO

There is considerable concern that malaria parasites are starting to evolve resistance to the current generation of antimalarial drugs, the artemisinin-based combination therapies (ACTs). We use pharmacological modeling to investigate changes in ACT effectiveness likely to occur if current regimens are extended from 3 to 5 days or, alternatively, given twice daily over 3 days. We show that the pharmacology of artemisinins allows both regimen changes to substantially increase the artemisinin killing rate. Malaria patients rarely contain more than 10(12) parasites, while the standard dosing regimens allow approximately 1 in 10(10) parasites to survive artemisinin treatment. Parasite survival falls dramatically, to around 1 in 10(17) parasites if the dose is extended or split; theoretically, this increase in drug killing appears to be more than sufficient to restore failing ACT efficacy. One of the most widely used dosing regimens, artemether-lumefantrine, already successfully employs a twice-daily dosing regimen, and we argue that twice-daily dosing should be incorporated into all ACT regimen design considerations as a simple and effective way of ensuring the continued long-term effectiveness of ACTs.


Assuntos
Antimaláricos/farmacocinética , Artemisininas/farmacocinética , Etanolaminas/farmacocinética , Fluorenos/farmacocinética , Malária Falciparum/tratamento farmacológico , Modelos Estatísticos , Plasmodium falciparum/efeitos dos fármacos , Adulto , Antimaláricos/farmacologia , Combinação Arteméter e Lumefantrina , Artemisininas/farmacologia , Criança , Relação Dose-Resposta a Droga , Esquema de Medicação , Combinação de Medicamentos , Cálculos da Dosagem de Medicamento , Etanolaminas/farmacologia , Feminino , Fluorenos/farmacologia , Humanos , Malária Falciparum/parasitologia , Masculino , Testes de Sensibilidade Parasitária , Plasmodium falciparum/fisiologia , Gravidez , Resultado do Tratamento
10.
Malar J ; 14: 292, 2015 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-26228915

RESUMO

BACKGROUND: The long half-lives of malaria 'partner' drugs are a potent force selecting for drug resistance. Clinical trials can quantify this effect by estimating a window of selection (WoS), defined as the amount of time post-treatment when drug levels are sufficiently high that resistant parasites can re-establish an infection while preventing drug-sensitive parasites from establishing viable infections. METHODS: The ability of clinical data to accurately estimate the true WoS was investigated using standard pharmacokinetic-pharmacodynamic models for three widely used malaria drugs: artemether-lumefantrine (AR-LF), artesunate-mefloquine (AS-MQ) and dihydroartemisinin-piperaquine (DHA-PPQ). Estimates of the clinical WoS either (1) ignored all new infections occurring after the 63-day follow-up period, as is currently done in clinical trials, or, (2) recognized that all individuals would eventually be re-infected and arbitrarily assigned them a new infection day. RESULTS: The results suggest current methods of estimating the clinical WoS underestimate the true WoS by as much as 9 days for AR-LF, 33 days for AS-MQ and 7 days for DHA-PPQ. The new method of estimating clinical WoS (i.e., retaining all individuals in the analysis) was significantly better at estimating the true WoS for AR-LF and AS-MQ. CONCLUSIONS: Previous studies, based on clinically observed WoS, have probably underestimated the 'true' WoS and hence the role of drugs with long half-lives in driving resistance. This has important policy implications: high levels of drug use are inevitable in mass drug administration programmes and intermittent preventative treatment programmes and the analysis herein suggests these policies will be far more potent drivers of resistance than previously thought.


Assuntos
Antimaláricos , Malária Falciparum , Plasmodium falciparum/efeitos dos fármacos , Antimaláricos/administração & dosagem , Antimaláricos/farmacocinética , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Combinação Arteméter e Lumefantrina , Artemisininas/administração & dosagem , Artemisininas/farmacocinética , Artemisininas/farmacologia , Artemisininas/uso terapêutico , Combinação de Medicamentos , Etanolaminas/administração & dosagem , Etanolaminas/farmacocinética , Etanolaminas/farmacologia , Etanolaminas/uso terapêutico , Fluorenos/administração & dosagem , Fluorenos/farmacocinética , Fluorenos/farmacologia , Fluorenos/uso terapêutico , Humanos , Malária Falciparum/tratamento farmacológico , Malária Falciparum/parasitologia , Modelos Biológicos , Quinolinas/administração & dosagem , Quinolinas/farmacocinética , Quinolinas/farmacologia , Quinolinas/uso terapêutico
11.
Antimicrob Agents Chemother ; 58(10): 5643-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24982091

RESUMO

It is now World Health Organization (WHO) policy that drug concentrations on day 7 be measured as part of routine assessment in antimalarial drug efficacy trials. The rationale is that this single pharmacological measure serves as a simple and practical predictor of treatment outcome for antimalarial drugs with long half-lives. Herein we review theoretical data and field studies and conclude that the day 7 drug concentration (d7c) actually appears to be a poor predictor of therapeutic outcome. This poor predictive capability combined with the fact that many routine antimalarial trials will have few or no failures means that there appears to be little justification for this WHO recommendation. Pharmacological studies have a huge potential to improve antimalarial dosing, and we propose study designs that use more-focused, sophisticated, and cost-effective ways of generating these data than the mass collection of single d7c concentrations.


Assuntos
Antimaláricos/farmacocinética , Antimaláricos/uso terapêutico , Artemisininas/farmacocinética , Artemisininas/uso terapêutico , Ensaios Clínicos como Assunto , Avaliação de Medicamentos , Humanos , Quinolinas/farmacocinética , Quinolinas/uso terapêutico
12.
PLoS Comput Biol ; 9(7): e1003151, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23874190

RESUMO

Mechanism-based pharmacokinetic-pharmacodynamic (PK/PD) modelling is the standard computational technique for simulating drug treatment of infectious diseases with the potential to enhance our understanding of drug treatment outcomes, drug deployment strategies, and dosing regimens. Standard methodologies assume only a single drug is used, it acts only in its unconverted form, and that oral drugs are instantaneously absorbed across the gut wall to their site of action. For drugs with short half-lives, this absorption period accounts for a significant period of their time in the body. Treatment of infectious diseases often uses combination therapies, so we refined and substantially extended the PK/PD methodologies to incorporate (i) time lags and drug concentration profiles resulting from absorption across the gut wall and, if required, conversion to another active form; (ii) multiple drugs within a treatment combination; (iii) differing modes of action of drugs in the combination: additive, synergistic, antagonistic; (iv) drugs converted to an active metabolite with a similar mode of action. This methodology was applied to a case study of two first-line malaria treatments based on artemisinin combination therapies (ACTs, artemether-lumefantrine and artesunate-mefloquine) where the likelihood of increased artemisinin tolerance/resistance has led to speculation on their continued long-term effectiveness. We note previous estimates of artemisinin kill rate were underestimated by a factor of seven, both the unconverted and converted form of the artemisinins kill parasites and the extended PK/PD methodology produced results consistent with field observations. The simulations predict that a potentially rapid decline in ACT effectiveness is likely to occur as artemisinin resistance spreads, emphasising the importance of containing the spread of artemisinin resistance before it results in widespread drug failure. We found that PK/PD data is generally very poorly reported in the malaria literature, severely reducing its value for subsequent re-application, and we make specific recommendations to improve this situation.


Assuntos
Antimaláricos/farmacologia , Antimaláricos/farmacocinética , Modelos Biológicos , Animais , Meia-Vida , Humanos , Plasmodium/efeitos dos fármacos
13.
Malar J ; 13: 62, 2014 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-24552440

RESUMO

Anti-malarial drugs are now mainly deployed as combination therapy (CT), primarily as a mechanism to prevent or slow the spread of resistance. This strategy is justified by mathematical arguments that generally assume that drug 'resistance' is a binary all-or-nothing genetic trait. Herein, a pharmacological, rather than a purely genetic, approach is used to investigate resistance and it is argued that this provides additional insight into the design principles of anti-malarial CTs. It is usually suggested that half-lives of constituent drugs in a CT be matched: it appears more important that their post-treatment anti-malarial activity profiles be matched and strategies identified that may achieve this. In particular, the considerable variation in pharmacological parameters noted in both human and parasites populations may compromise this matching and it is, therefore, essential to accurately quantify the population pharmacokinetics of the drugs in the CTs. Increasing drug dosages will likely follow a law of diminishing returns in efficacy, i.e. a certain increase in dose will not necessarily lead to the same percent increase in efficacy. This may allow individual drug dosages to be lowered without proportional decrease in efficacy, reducing any potential toxicity, and allowing the other drug(s) in the CT to compensate for this reduced dosage; this is a dangerous strategy which is discussed further. Finally, pharmacokinetic and pharmacodynamic drug interactions and the role of resistance mechanisms are discussed. This approach generated an idealized target product profile (TPP) for anti-malarial CTs. There is a restricted pipeline of anti-malarial drugs but awareness of pharmacological design principles during the development stages could optimize CT design pre-deployment. This may help prevent changes in drug dosages and/or regimen that have previously occurred post-deployment in most current anti-malarial drugs.


Assuntos
Antimaláricos/administração & dosagem , Antimaláricos/farmacocinética , Malária/tratamento farmacológico , Antimaláricos/farmacologia , Combinação de Medicamentos , Interações Medicamentosas , Resistência a Medicamentos , Quimioterapia Combinada/métodos , Meia-Vida , Humanos , Plasmodium/efeitos dos fármacos
14.
Malar J ; 13: 138, 2014 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-24708571

RESUMO

BACKGROUND: Successful programmatic use of anti-malarials faces challenges that are not covered by standard drug development processes. The development of appropriate pragmatic dosing regimens for low-resource settings or community-based use is not formally regulated, even though these may alter factors which can substantially affect individual patient and population level outcome, such as drug exposure, patient adherence and the spread of drug resistance and can affect a drug's reputation and its eventual therapeutic lifespan. METHODS: An in silico pharmacological model of anti-malarial drug treatment with the pharmacokinetic/pharmacodynamic profiles of artemether-lumefantrine (AM-LF, Coartem®) and dihydroartemisinin-piperaquine (DHA-PPQ, Eurartesim®) was constructed to assess the potential impact of programmatic factors, including regionally optimized, age-based dosing regimens, poor patient adherence, food effects and drug resistance on treatment outcome at population level, and compared both drugs' susceptibility to these factors. RESULTS: Compared with DHA-PPQ, therapeutic effectiveness of AM-LF seems more robust to factors affecting drug exposure, such as age- instead of weight-based dosing or poor adherence. The model highlights the sub-optimally low ratio of DHA:PPQ which, in combination with the narrow therapeutic dose range of PPQ compared to DHA that drives the weight or age cut-offs, leaves DHA at a high risk of under-dosing. CONCLUSION: Pharmacological modelling of real-life scenarios can provide valuable supportive data and highlight modifiable determinants of therapeutic effectiveness that can help optimize the deployment of anti-malarials in control programmes.


Assuntos
Antimaláricos/administração & dosagem , Artemisininas/administração & dosagem , Etanolaminas/administração & dosagem , Fluorenos/administração & dosagem , Malária/tratamento farmacológico , Quinolinas/administração & dosagem , Adolescente , Adulto , Antimaláricos/farmacocinética , Combinação Arteméter e Lumefantrina , Artemisininas/farmacocinética , Criança , Pré-Escolar , Simulação por Computador , Combinação de Medicamentos , Etanolaminas/farmacocinética , Feminino , Fluorenos/farmacocinética , Humanos , Lactente , Masculino , Quinolinas/farmacocinética , Resultado do Tratamento , Adulto Jovem
15.
Lancet Microbe ; 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38996497

RESUMO

BACKGROUND: Seasonal malaria chemoprevention (SMC) with sulfadoxine-pyrimethamine plus amodiaquine prevents millions of clinical malaria cases in children younger than 5 years in Africa's Sahel region. However, Plasmodium falciparum parasites partially resistant to sulfadoxine-pyrimethamine (with quintuple mutations) potentially threaten the protective effectiveness of SMC. We evaluated the spread of quintuple-mutant parasites and the clinical consequences. METHODS: We used an individual-based malaria transmission model with explicit parasite dynamics and drug pharmacological models to identify and quantify the influence of factors driving quintuple-mutant spread and predict the time needed for the mutant to spread from 1% to 50% of inoculations for several SMC deployment strategies. We estimated the impact of this spread on SMC effectiveness against clinical malaria. FINDINGS: Higher transmission intensity, SMC coverage, and expanded age range of chemoprevention promoted mutant spread. When SMC was implemented in a high-transmission setting (40% parasite prevalence in children aged 2-10 years) with four monthly cycles to children aged 3 months to 5 years (with 95% initial coverage declining each cycle), the quintuple mutant required 53·1 years (95% CI 50·5-56·0) to spread from 1% to 50% of inoculations. This time increased in lower-transmission settings and reduced by half when SMC was extended to children aged 3 months to 10 years, or reduced by 10-13 years when an additional monthly cycle of SMC was deployed. For the same setting, the effective reduction in clinical cases in children receiving SMC was 79·0% (95% CI 77·8-80·8) and 60·4% (58·6-62·3) during the months of SMC implementation when the quintuple mutant was absent or fixed in the population, respectively. INTERPRETATION: SMC with sulfadoxine-pyrimethamine plus amodiaquine leads to a relatively slow spread of sulfadoxine-pyrimethamine-resistant quintuple mutants and remains effective at preventing clinical malaria despite the mutant spread. SMC with sulfadoxine-pyrimethamine plus amodiaquine should be considered in seasonal settings where this mutant is already prevalent. FUNDING: Swiss National Science Foundation and Marie Curie Individual Fellowship.

16.
G3 (Bethesda) ; 14(1)2023 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-37972212

RESUMO

Fisher's reproductive compensation (fRC) occurs when a species' demography means the death of an individual results in increased survival probability of his/her relatives, usually assumed to be full sibs. This likely occurs in many species, including humans. Several important recessive human genetic diseases cause early foetal/infant death allowing fRC to act on these mutations. The impact of fRC on these genetic conditions has been previously calculated and shown to be substantial as quantified by ω, the fold increase in equilibrium frequencies of the mutation under fRC compared with its absence, i.e. ω = 1.22 and ω = 1.33 for autosomal and sex-linked loci, respectively. However, the impact of fRC on the frequency of the much larger class of semidominant, nonlethal mutations is unknown. This is calculated here as ω = 2 - h*s for autosomal loci and ω up to 2 for sex-linked loci where h is dominance (varied between 0.05 and 0.95) and s is selection coefficient (varied between 0.05 and 0.9). These results show that the actions of fRC can almost double the equilibrium frequency of deleterious mutations with low values of h and/or s (noting that "low" is s∼0.05 to 0.1). It is noted that fRC may act differentially across the genome with genes expressed early in life being fully exposed to fRC while those expressed later in life may be unaffected; this could lead to systematic differences in deleterious allele frequency across the genome.


Assuntos
Reprodução , Seleção Genética , Humanos , Feminino , Masculino , Mutação , Frequência do Gene , Reprodução/genética , Genoma , Modelos Genéticos
17.
Bioinformatics ; 27(9): 1335-6, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21414984

RESUMO

MOTIVATION: The evolution of resistance in Plasmodium falciparum malaria against most available treatments is a major global health threat. Population genetics approaches are commonly used to model the spread of drug resistance. Due to uncommon features in malaria biology, existing forward-time population genetics simulators cannot suitably model Plasmodium falciparum malaria. RESULTS: Here we present ogaraK, a population genetics simulator for modelling the spread of drug-resistant malaria. OgaraK is designed to make malaria simulation computationally tractable as it models infections, not individual parasites. OgaraK is also able to model the life cycle of the parasite which includes both haploid and diploid phases and sexual and asexual reproduction. We also allow for the simulation of different inbreeding levels, an important difference between high and low transmission areas and a fundamental factor influencing the outcome of strategies to control or eliminate malaria. AVAILABILITY: OgaraK is available as free software (GPL) from the address http://popgen.eu/soft/ogaraK.


Assuntos
Simulação por Computador , Resistência a Medicamentos/genética , Genética Populacional/métodos , Plasmodium falciparum/genética , Software , Genes de Protozoários , Malária Falciparum/epidemiologia , Malária Falciparum/parasitologia , Plasmodium falciparum/efeitos dos fármacos
18.
Malar J ; 11: 258, 2012 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-22856525

RESUMO

BACKGROUND: Insecticides are an effective and practical tool for reducing malaria transmission but the development of resistance to the insecticides can potentially compromise controls efforts. In this study a mathematical model was developed to explore the effects on mosquito populations of spatial heterogeneous deployment of insecticides. This model was used to identify important parameters in the evolution of insecticide resistance and to examine the contribution of new generation long-lasting insecticidal bed nets, that incorporate a chemical synergist on the roof panel, in delaying insecticide resistance. METHODS: A genetic model was developed to predict changes in mosquito fitness and resistance allele frequency. Parameters describing insecticide selection, fitness cost and the additional use of synergist were incorporated. Uncertainty and sensitivity analysis were performed followed by investigation of the evolution of resistance under scenarios of fully effective or ineffective synergists. RESULTS: The spread of resistance was most sensitive to selection coefficients, fitness cost and dominance coefficients while mean fitness was most affected by baseline fitness levels. Using a synergist delayed the spread of resistance but could, in specific circumstances that were thoroughly investigated, actually increase the rate of spread. Different spread dynamics were observed, with simulations leading to fixation, loss and most interestingly, equilibrium (without explicit overdominance) of the resistance allele. CONCLUSIONS: This strategy has the potential to delay the spread of resistance but note that in an heterogeneous environment it can also lead to the opposite effect, i.e., increasing the rate of spread. This clearly emphasizes that selection pressure acting inside the house cannot be treated in isolation but must be placed in context of overall insecticide use in an heterogeneous environment.


Assuntos
Resistência a Inseticidas/genética , Malária/prevenção & controle , Mosquiteiros , Sinergistas de Praguicidas/administração & dosagem , Animais , Anopheles/efeitos dos fármacos , Anopheles/genética , Anopheles/parasitologia , Frequência do Gene , Genes de Insetos , Humanos , Inseticidas/administração & dosagem , Modelos Logísticos , Malária/parasitologia , Malária/transmissão , Modelos Genéticos , Butóxido de Piperonila/administração & dosagem
19.
Malar J ; 11: 380, 2012 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-23171123

RESUMO

BACKGROUND: Although malaria is highly prevalent throughout Malawi, little is known of its transmission dynamics. This paper describes the seasonal activity of the different vectors, human biting indices, sporozoite rates and the entomological inoculation rate in a low-lying rural area in southern Malawi. METHODS: Vectors were sampled over 52 weeks from January 2002 to January 2003, by pyrethrum knockdown catch in two villages in Chikhwawa district, in the Lower Shire Valley. RESULTS: In total, 7,717 anophelines were collected of which 55.1% were Anopheles gambiae sensu lato and 44.9% were Anopheles funestus. Three members of the An. gambiae complex were identified by PCR: Anopheles arabiensis (75%) was abundant throughout the year, An. gambiae s.s. (25%) was most common during the wet season and Anopheles quadriannulatus occurred at a very low frequency (n=16). An. funestus was found in all samples but was most common during the dry season.Anopheles gambiae s.s. and An. funestus were highly anthropophilic with human blood indices of 99.2% and 96.3%, respectively. Anopheles arabiensis had fed predominantly on humans (85.0%) and less commonly on cattle (10.9%; 1.2% of blood meals were of mixed origin). Plasmodium falciparum (192/3,984) and Plasmodium malariae (1/3,984) sporozoites were detected by PCR in An. arabiensis (3.2%) and An. funestus (4.5%), and in a significantly higher proportion of An. gambiae s.s. (10.6%)(p<0.01). All three vectors were present throughout the year and malaria transmission occurred in every month, although with greatest intensity during the rainy season (January to April). The combined human blood index exceeded 92% and the P. falciparum sporozoite rate was 4.8%, resulting in estimated inoculation rates of 183 infective bites/ person per annum, or an average rate of ~15 infective bites/person/month. CONCLUSIONS: The results demonstrate the importance of An. gambiae s.s., An. arabiensis and An. funestus in driving the high levels of malaria transmission in the south of Malawi. Sustained and high coverage or roll out of current approaches to malaria control (primarily insecticide-treated bed nets and indoor residual house spraying) in the area are likely to reduce the observed high malaria transmission rate and consequently the incidence of human infections, unless impeded by increasing resistance of vectors to insecticides.


Assuntos
Malária/transmissão , Animais , Anopheles/parasitologia , Bovinos , Feminino , Humanos , Insetos Vetores/parasitologia , Malária/epidemiologia , Malária/prevenção & controle , Malaui/epidemiologia , Controle de Mosquitos , Oocistos , Plasmodium/isolamento & purificação , População Rural , Estações do Ano , Esporozoítos
20.
Trends Parasitol ; 38(11): 933-941, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36068129

RESUMO

Estimating antimalarial drug efficacy requires differentiating treatment failures from new infections arising during the several-week follow-up period in drug trials. Genetic profiling of malaria infections can guide this decision but is notoriously difficult in practice. Previous World Health Organisation (WHO) guidelines were based on assumptions with an inherently high risk of underestimating failure rates. A recent update to WHO guidelines recognises a wider range of analyses to overcome these limitations. We discuss these new analyses and their underlying logic. Drug failure rate estimates in moderate to high transmissions areas will become more accurate but will likely rise twofold due to better detection of treatment failures, and the malaria community needs to anticipate and prepare for potentially large increases in estimated failure rates.


Assuntos
Antimaláricos , Malária Falciparum , Malária , Antimaláricos/uso terapêutico , Humanos , Malária/diagnóstico , Malária/tratamento farmacológico , Malária Falciparum/tratamento farmacológico , Organização Mundial da Saúde
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA