Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 521
Filter
1.
Malar J ; 23(1): 303, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39385192

ABSTRACT

BACKGROUND: Malaria prevention and control strategies have been hampered by urbanization and the spread of Anopheles stephensi. The spread of this vector into Africa further complicates the already complex malaria situation, that could put about 126 million Africans at risk of infection. Hence, this study aimed to assess the determinants of urban malaria, focusing on the role of urbanization and the distribution of An. stephensi in Eastern Ethiopia. METHODS: A matched case control study was conducted among febrile urban residents of Dire Dawa (malaria positive as cases and negative as a control). A capillary blood sample was collected for parasite identification using microscopic examination and an interviewer administered questionnaire was used to collect additional data. Centers for Disease Control and Prevention miniature light traps (CDC-LT) and Prokopack aspirator were used to collect adult mosquito vectors from the selected cases and control houses to identify the mosquito vector species. Then, the data were exported to STATA for analysis. Conditional logistic regression was done to identify determinants, and principal component Analysis (PCA) was done for some independent variables. RESULTS: This study enrolled 132 cases and 264 controls from urban setting only. Of the 132 cases, 90 cases were positive for Plasmodium falciparum, 34 were positive for Plasmodium vivax and 8 had mixed infections. All cases and controls were similar with regard to their respective age and sex. Travel history (AOR: 13.1, 95% CI 2.8-61.4), presence of eves and holes on walls (AOR: 2.84, 95% CI 1.5-5.5), history of malaria diagnosis (AOR: 2.4, 95% CI 1.1-5.3), owning any livestock (AOR: 7.5, 95% CI 2.4-22.8), presence of stagnant water in the area (AOR: 3.2, 95% CI 1.7-6.1), sleeping under bed net the previous night (AOR: 0.21, 95% CI 0.1-0.6) and knowledge on malaria and its prevention (AOR: 2.2, 95% CI 1.2-4.1) were determinants of urban malaria infection. About 34 adult Anopheles mosquitoes were collected and identified from those selected cases and control houses and 27 of them were identified as An. stephensi. CONCLUSION: Among the cases, the dominant species were P. falciparum. This study identified travel history, house condition, past infection, livestock ownership, stagnant water, bed net use, and malaria knowledge as determinants of infection. This study also found the dominance of the presence of An. stephensi among the collected mosquito vectors. This suggests that the spread of An. stephensi may be impacting malaria infection in the study area. Hence, strengthening urban-targeted malaria interventions should be enhanced to prevent and control further urban malaria infection and spread.


Subject(s)
Anopheles , Malaria, Falciparum , Mosquito Vectors , Urban Population , Urbanization , Animals , Ethiopia/epidemiology , Anopheles/physiology , Anopheles/parasitology , Female , Male , Humans , Adult , Adolescent , Young Adult , Mosquito Vectors/parasitology , Mosquito Vectors/physiology , Case-Control Studies , Urban Population/statistics & numerical data , Child , Middle Aged , Malaria, Falciparum/epidemiology , Malaria, Falciparum/prevention & control , Child, Preschool , Malaria, Vivax/epidemiology , Malaria, Vivax/parasitology , Plasmodium falciparum/isolation & purification , Plasmodium vivax/physiology , Infant
2.
Malar J ; 23(1): 271, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39256842

ABSTRACT

BACKGROUND: In most countries engaged on the last mile towards malaria elimination, residual transmission mainly persists among vulnerable populations represented by isolated and mobile (often cross-border) communities. These populations are sometimes involved in informal or even illegal activities. In regions with Plasmodium vivax transmission, the specific biology of this parasite poses additional difficulties related to the need for a radical treatment against hypnozoites to prevent relapses. Among hard-to-reach communities, case management, a pillar of elimination strategy, is deficient: acute malaria attacks often occur in remote areas, where there is limited access to care, and drugs acquired outside formal healthcare are often inadequately used for treatment, which typically does not include radical treatment against P. vivax. For these reasons, P. vivax circulation among these communities represents one of the main challenges for malaria elimination in many non-African countries. The objective of this article is to describe the protocol of the CUREMA study, which aims to meet the challenge of targeting malaria in hard-to-reach populations with a focus on P. vivax. RESULTS: CUREMA is a multi-centre, international public health intervention research project. The study population is represented by persons involved in artisanal and small-scale gold mining who are active and mobile in the Guiana Shield, deep inside the Amazon Forest. The CUREMA project includes a complex intervention composed of a package of actions: (1) health education activities; (2) targeted administration of treatment against P. vivax after screening against G6PD deficiency to asymptomatic persons considered at risk of silently carrying the parasite; (3) distribution of a self-testing and self-treatment kit (malakit) associated with user training for self-management of malaria symptoms occurring while in extreme isolation. These actions are offered by community health workers at settlements and neighbourhoods (often cross-border) that represent transit and logistic bases of gold miners. The study relies on hybrid design, aiming to evaluate both the effectiveness of the intervention on malaria transmission with a pre/post quasi-experimental design, and its implementation with a mixed methods approach. CONCLUSIONS: The purpose of this study is to experiment an intervention that addresses both Plasmodium falciparum and P. vivax malaria elimination in a mobile and isolated population and to produce results that can be transferred to many contexts facing the same challenges around the world.


Subject(s)
Disease Eradication , Malaria, Vivax , Humans , Malaria, Vivax/prevention & control , Disease Eradication/methods , Disease Eradication/statistics & numerical data , Male , Female , Antimalarials/therapeutic use , Adult , Middle Aged , Adolescent , Young Adult , Child , Plasmodium vivax/physiology
3.
Epidemics ; 48: 100789, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39255654

ABSTRACT

Plasmodium vivax is the most geographically widespread malaria parasite. P. vivax has the ability to remain dormant (as a hypnozoite) in the human liver and subsequently reactivate, which makes control efforts more difficult. Given the majority of P. vivax infections are due to hypnozoite reactivation, targeting the hypnozoite reservoir with a radical cure is crucial for achieving P. vivax elimination. Stochastic effects can strongly influence dynamics when disease prevalence is low or when the population size is small. Hence, it is important to account for this when modelling malaria elimination. We use a stochastic multiscale model of P. vivax transmission to study the impacts of multiple rounds of mass drug administration (MDA) with a radical cure, accounting for superinfection and hypnozoite dynamics. Our results indicate multiple rounds of MDA with a high-efficacy drug are needed to achieve a substantial probability of elimination. This work has the potential to help guide P. vivax elimination strategies by quantifying elimination probabilities for an MDA approach.


Subject(s)
Antimalarials , Disease Eradication , Malaria, Vivax , Mass Drug Administration , Plasmodium vivax , Humans , Malaria, Vivax/prevention & control , Malaria, Vivax/drug therapy , Malaria, Vivax/epidemiology , Mass Drug Administration/statistics & numerical data , Plasmodium vivax/drug effects , Plasmodium vivax/physiology , Disease Eradication/methods , Disease Eradication/statistics & numerical data , Antimalarials/therapeutic use , Antimalarials/administration & dosage , Stochastic Processes , Computer Simulation
4.
Nat Commun ; 15(1): 7625, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223117

ABSTRACT

Most Plasmodium vivax infections contain genetically distinct parasites, but the consequences of this polyclonality on the development of asexual parasites, their sexual differentiation, and their transmission remain unknown. We describe infections of Saimiri monkeys with two strains of P. vivax and the analyses of 80,024 parasites characterized by single cell RNA sequencing and individually genotyped. In our model, consecutive inoculations fail to establish polyclonal infections. By contrast, simultaneous inoculations of two strains lead to sustained polyclonal infections, although without detectable differences in parasite regulation or sexual commitment. Analyses of sporozoites dissected from mosquitoes fed on coinfected monkeys show that all genotypes are successfully transmitted to mosquitoes. However, after sporozoite inoculation, not all genotypes contribute to the subsequent blood infections, highlighting an important bottleneck during pre-erythrocytic development. Overall, these studies provide new insights on the mechanisms regulating the establishment of polyclonal P. vivax infections and their consequences for disease transmission.


Subject(s)
Genotype , Malaria, Vivax , Plasmodium vivax , Saimiri , Single-Cell Analysis , Sporozoites , Animals , Plasmodium vivax/genetics , Plasmodium vivax/physiology , Malaria, Vivax/transmission , Malaria, Vivax/parasitology , Female , Culicidae/parasitology , Humans , Male
5.
Malar J ; 23(1): 258, 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39182127

ABSTRACT

BACKGROUND: Despite the successful efforts in controlling malaria in Vietnam, the disease remains a significant health concern, particularly in Central Vietnam. This study aimed to assess correlations between environmental, climatic, and socio-economic factors in the district with malaria cases. METHODS: The study was conducted in 15 provinces in Central Vietnam from January 2018 to December 2022. Monthly malaria cases were obtained from the Institute of Malariology, Parasitology, and Entomology Quy Nhon, Vietnam. Environmental, climatic, and socio-economic data were retrieved using a Google Earth Engine script. A multivariable Zero-inflated Poisson regression was undertaken using a Bayesian framework with spatial and spatiotemporal random effects with a conditional autoregressive prior structure. The posterior random effects were estimated using Bayesian Markov Chain Monte Carlo simulation with Gibbs sampling. RESULTS: There was a total of 5,985 Plasmodium falciparum and 2,623 Plasmodium vivax cases during the study period. Plasmodium falciparum risk increased by five times (95% credible interval [CrI] 4.37, 6.74) for each 1-unit increase of normalized difference vegetation index (NDVI) without lag and by 8% (95% CrI 7%, 9%) for every 1ºC increase in maximum temperature (TMAX) at a 6-month lag. While a decrease in risk of 1% (95% CrI 0%, 1%) for a 1 mm increase in precipitation with a 6-month lag was observed. A 1-unit increase in NDVI at a 1-month lag was associated with a four-fold increase (95% CrI 2.95, 4.90) in risk of P. vivax. In addition, the risk increased by 6% (95% CrI 5%, 7%) and 3% (95% CrI 1%, 5%) for each 1ºC increase in land surface temperature during daytime with a 6-month lag and TMAX at a 4-month lag, respectively. Spatial analysis showed a higher mean malaria risk of both species in the Central Highlands and southeast parts of Central Vietnam and a lower risk in the northern and north-western areas. CONCLUSION: Identification of environmental, climatic, and socio-economic risk factors and spatial malaria clusters are crucial for designing adaptive strategies to maximize the impact of limited public health resources toward eliminating malaria in Vietnam.


Subject(s)
Bayes Theorem , Climate , Malaria, Falciparum , Malaria, Vivax , Socioeconomic Factors , Spatio-Temporal Analysis , Vietnam/epidemiology , Malaria, Falciparum/epidemiology , Malaria, Vivax/epidemiology , Humans , Environment , Plasmodium falciparum , Plasmodium vivax/physiology
6.
Malar J ; 23(1): 231, 2024 Aug 04.
Article in English | MEDLINE | ID: mdl-39098946

ABSTRACT

BACKGROUND: The government of Lao PDR has increased efforts to control malaria transmission in order to reach its national elimination goal by 2030. Weather can influence malaria transmission dynamics and should be considered when assessing the impact of elimination interventions but this relationship has not been well characterized in Lao PDR. This study examined the space-time association between climate variables and Plasmodium falciparum and Plasmodium vivax malaria incidence from 2010 to 2022. METHODS: Spatiotemporal Bayesian modelling was used to investigate the monthly relationship, and model selection criteria were used to evaluate the performance of the models and weather variable specifications. As the malaria control and elimination situation was spatially and temporally dynamic during the study period, the association was examined annually at the provincial level. RESULTS: Malaria incidence decreased from 2010 to 2022 and was concentrated in the southern regions for both P. falciparum and P. vivax. Rainfall and maximum humidity were identified as most strongly associated with malaria during the study period. Rainfall was associated with P. falciparum incidence in the north and central regions during 2010-2011, and with P. vivax incidence in the north and central regions during 2012-2015. Maximum humidity was persistently associated with P. falciparum and P. vivax incidence in the south. CONCLUSIONS: Malaria remains prevalent in Lao PDR, particularly in the south, and the relationship with weather varies between regions but was strongest for rainfall and maximum humidity for both species. During peak periods with suitable weather conditions, vector control activities and raising public health awareness on the proper usage of intervention measures, such as indoor residual spraying and personal protection, should be prioritized.


Subject(s)
Bayes Theorem , Climate , Malaria, Falciparum , Malaria, Vivax , Spatio-Temporal Analysis , Laos/epidemiology , Malaria, Vivax/epidemiology , Malaria, Vivax/prevention & control , Malaria, Falciparum/epidemiology , Malaria, Falciparum/prevention & control , Incidence , Humans , Plasmodium vivax/physiology , Weather , Disease Eradication/statistics & numerical data
7.
J Vis Exp ; (210)2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39185871

ABSTRACT

Visual Dynamics (VD) is a web tool that aims to facilitate the use and application of Molecular Dynamics (MD) executed in Gromacs, allowing users without computational familiarity to run short-time simulations for validation, demonstration, and teaching purposes. It is true that quantum methods are the most accurate. However, there is currently no computational feasibility to carry out the experiments that MD performs. The tool described here has continuously received improvements over the course of the last couple of years. This protocol will describe what is needed to run a simulation in VD with a protein-ligand complex previously prepared in ACPYPE and some general directions on the other simulation models available. For the detailed simulation, the FK506-binding protein from Plasmodium vivax complexed with the inhibitor D5 (PDB ID: 4mgv) will be used, and all files used will be provided. Note that this protocol will tell every option to be used to achieve the same results presented, but these options are not necessarily the only ones available.


Subject(s)
Molecular Dynamics Simulation , Plasmodium vivax/physiology , Software , Tacrolimus Binding Proteins/chemistry , Protozoan Proteins/chemistry
8.
Malar J ; 23(1): 212, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39020432

ABSTRACT

BACKGROUND: Thailand is approaching local elimination of malaria in the eastern provinces. It has successfully reduced the number of cases over the past decade, but there are persistent transmission hot spots in and around forests. This study aimed to use data from the malaria surveillance system to describe the spatiotemporal trends of malaria in Northeast Thailand and fine-scale patterns in locally transmitted cases between 2011 and 2021. METHODS: Case data was stratified based on likely location of infection and parasite species. Annual Parasite Index per 1000 population (API) was calculated for different categories. Time series decomposition was performed to identify trends and seasonal patterns. Statistically significant clusters of high (hot spots) and low (cold spots) API were identified using the Getis-Ord Gi* statistic. The stability of those hot spots and the absolute change in the proportion of API density from baseline were compared by case type. RESULTS: The total number of confirmed cases experienced a non-linear decline by 96.6%, from 1061 in 2011 to 36 in 2021. There has been a decline in both Plasmodium vivax and Plasmodium falciparum case numbers, with only four confirmed P. falciparum cases over the last two years-a 98.89% drop from 180 in 2011. API was generally higher in Si Sa Ket province, which had peaks every 2-3 years. There was a large outbreak in Ubon Ratchathani in 2014-2016 which had a high proportion of P. falciparum reported. The proportion of cases classified increased over the study period, and the proportion of cases classed as indigenous to the village of residence increased from 0.2% to 33.3%. There were stable hot spots of indigenous and imported cases in the south of Si Sa Ket and southeast of Ubon Ratchathani. Plasmodium vivax hot spots were observed into recent years, while those of P. falciparum decreased to zero in Ubon in 2020 and emerged in the eastern part in 2021, the same year that P. falciparum hot spots in Si Sa Ket reached zero. CONCLUSIONS: There has been a large, non-linear decline in the number of malaria cases reported and an increasing proportion of cases are classed as indigenous to the patient's village of residence. Stable hot spots of ongoing transmission in the forested border areas were identified, with transmission likely persisting because of remote location and high-risk forest-going behaviours. Future efforts should include cross-border collaboration and continued targeting of high-risk behaviours to reduce the risk of imported cases seeding local transmission.


Subject(s)
Malaria, Falciparum , Malaria, Vivax , Plasmodium falciparum , Thailand/epidemiology , Malaria, Vivax/epidemiology , Malaria, Vivax/transmission , Malaria, Vivax/prevention & control , Malaria, Falciparum/epidemiology , Malaria, Falciparum/transmission , Malaria, Falciparum/prevention & control , Humans , Plasmodium vivax/physiology , Spatio-Temporal Analysis , Disease Eradication/statistics & numerical data , Seasons , Disease Hotspot
9.
Malar J ; 23(1): 189, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38880891

ABSTRACT

BACKGROUND: Malaria, a prominent vector borne disease causing over a million annual cases worldwide, predominantly affects vulnerable populations in the least developed regions. Despite their preventable and treatable nature, malaria remains a global public health concern. In the last decade, India has faced a significant decline in malaria morbidity and mortality. As India pledged to eliminate malaria by 2030, this study examined a decade of surveillance data to uncover space-time clustering and seasonal trends of Plasmodium vivax and Plasmodium falciparum malaria cases in West Bengal. METHODS: Seasonal and trend decomposition using Loess (STL) was applied to detect seasonal trend and anomaly of the time series. Univariate and multivariate space-time cluster analysis of both malaria cases were performed at block level using Kulldorff's space-time scan statistics from April 2011 to March 2021 to detect statistically significant space-time clusters. RESULTS: From the time series decomposition, a clear seasonal pattern is visible for both malaria cases. Statistical analysis indicated considerable high-risk P. vivax clusters, particularly in the northern, central, and lower Gangetic areas. Whereas, P. falciparum was concentrated in the western region with a significant recent transmission towards the lower Gangetic plain. From the multivariate space-time scan statistics, the co-occurrence of both cases were detected with four significant clusters, which signifies the regions experiencing a greater burden of malaria cases. CONCLUSIONS: Seasonal trends from the time series decomposition analysis show a gradual decline for both P. vivax and P. falciparum cases in West Bengal. The space-time scan statistics identified high-risk blocks for P. vivax and P. falciparum malaria and its co-occurrence. Both malaria types exhibit significant spatiotemporal variations over the study area. Identifying emerging high-risk areas of P. falciparum malaria over the Gangetic belt indicates the need for more research for its spatial shifting. Addressing the drivers of malaria transmission in these diverse clusters demands regional cooperation and strategic strategies, crucial steps towards overcoming the final obstacles in malaria eradication.


Subject(s)
Malaria, Falciparum , Malaria, Vivax , Plasmodium vivax , Seasons , India/epidemiology , Malaria, Vivax/epidemiology , Malaria, Vivax/parasitology , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Humans , Plasmodium vivax/physiology , Space-Time Clustering , Plasmodium falciparum/physiology
10.
Sci Rep ; 14(1): 13669, 2024 06 13.
Article in English | MEDLINE | ID: mdl-38871839

ABSTRACT

Among the factors affecting the effectiveness of malaria control is poor knowledge of the entomologic drivers of the disease. We investigated anopheline populations as part of a baseline study to implement house screening of windows and doors as a supplementary malaria control tool towards elimination in Jabi Tehnan district, Amhara Regional State of Ethiopia. The samples were surveyed monthly using CDC light traps between June 2020 and May 2021. Mosquito trap density (< 3 mosquitoes/trap) was low, however, with a high overall Plasmodium sporozoite rate (9%; indoor = 4.3%, outdoor = 13.1%) comprising P. falciparum (88.9%) and P. vivax (11.1%). Anopheles gambiae s.l., mostly An. arabiensis, comprised > 80% of total anopheline captures and contributed ~ 42% of Plasmodium-infected mosquitoes. On the other hand, morphologically scored Anopheles funestus s.l., constituting about 6% of anopheline collections, accounted for 50% of sporozoite-infected mosquitoes. Most of the infected An. funestus s.l. specimens (86.7%) were grouped with previously unknown or undescribed Anopheles species previously implicated as a cryptic malaria vector in the western Kenyan highlands, confirming its wider geographic distribution in eastern Africa. Other species with Plasmodium infection included An. longipalpis C, An. theileri, An. demillioni, and An. nili. Cumulatively, 77.8% of the infected mosquitoes occurred outdoors. These results suggest efficient malaria parasite transmission despite the low vector densities, which has implications for effective endpoint indicators to monitor malaria control progress. Additionally, the largely outdoor infection and discovery of previously unknown and cryptic vectors suggest an increased risk of residual malaria transmission and, thus, a constraint on effective malaria prevention and control.


Subject(s)
Anopheles , Mosquito Vectors , Ethiopia/epidemiology , Animals , Anopheles/parasitology , Mosquito Vectors/parasitology , Humans , Malaria/transmission , Malaria/epidemiology , Plasmodium falciparum/isolation & purification , Plasmodium falciparum/pathogenicity , Plasmodium vivax/physiology , Sporozoites , Mosquito Control/methods , Malaria, Vivax/transmission , Malaria, Vivax/epidemiology , Malaria, Vivax/parasitology , Malaria, Falciparum/transmission , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Female
11.
Malar J ; 23(1): 143, 2024 May 12.
Article in English | MEDLINE | ID: mdl-38735957

ABSTRACT

BACKGROUND: Despite continuous prevention and control strategies in place, malaria remains a major public health problem in sub-Saharan Africa including Ethiopia. Moreover, prevalence of malaria differs in different geographical settings and epidemiological data were inadequate to assure disease status in the study area. This study was aimed to determine the prevalence of malaria and associated risk factors in selected rural kebeles in South Ethiopia. METHODS: A community-based cross-sectional study was conducted between February to June 2019 in eight malaria-endemic kebeles situated in four zones in South Ethiopia. Mult-stage sampling techniques were employed to select the study zones, districts, kebeles and households. Blood sample were collected from 1674 participants in 345 households by finger prick and smears were examined by microscopy. Sociodemographic data as well as risk factors for Plasmodium infection were collected using questionnaires. Bivariate and multivariate logistic regressions were used to analyse the data. RESULTS: The overall prevalence of malaria in the study localities was 4.5% (76/1674). The prevalence was varied among the study localities with high prevalence in Bashilo (14.6%; 33/226) followed by Mehal Korga (12.1%; 26/214). Plasmodium falciparum was the dominant parasite accounted for 65.8% (50/76), while Plasmodium vivax accounted 18.4% (14/76). Co-infection of P. falciparum and P. vivax was 15.8% (12/76). Among the three age groups prevalence was 7.8% (27/346) in age less than 5 years and 7.5% (40/531) in 5-14 years. The age groups > 14years were less likely infected with Plasmodium parasite (AOR = 0.14, 95% CI 0.02-0.82) than under five children. Non-febrile individuals 1638 (97.8%) were more likely to had Plasmodium infection (AOR = 28.4, 95% CI 011.4-70.6) than febrile 36 (2.2%). Individuals living proximity to mosquito breeding sites have higher Plasmodium infection (AOR = 6.17, 95% CI 2.66-14.3) than those at distant of breeding sites. CONCLUSIONS: Malaria remains a public health problem in the study localities. Thus, malaria prevention and control strategies targeting children, non-febrile cases and individuals living proximity to breeding sites are crucial to reduce malaria related morbidity and mortality.


Subject(s)
Family Characteristics , Malaria, Falciparum , Malaria, Vivax , Ethiopia/epidemiology , Cross-Sectional Studies , Prevalence , Humans , Risk Factors , Female , Male , Adolescent , Adult , Child, Preschool , Young Adult , Child , Middle Aged , Infant , Malaria, Vivax/epidemiology , Malaria, Vivax/parasitology , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Plasmodium vivax/physiology , Plasmodium falciparum/isolation & purification , Aged , Rural Population/statistics & numerical data , Malaria/epidemiology , Malaria/parasitology
12.
J Math Biol ; 89(1): 7, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38772937

ABSTRACT

Malaria is a vector-borne disease that exacts a grave toll in the Global South. The epidemiology of Plasmodium vivax, the most geographically expansive agent of human malaria, is characterised by the accrual of a reservoir of dormant parasites known as hypnozoites. Relapses, arising from hypnozoite activation events, comprise the majority of the blood-stage infection burden, with implications for the acquisition of immunity and the distribution of superinfection. Here, we construct a novel model for the transmission of P. vivax that concurrently accounts for the accrual of the hypnozoite reservoir, (blood-stage) superinfection and the acquisition of immunity. We begin by using an infinite-server queueing network model to characterise the within-host dynamics as a function of mosquito-to-human transmission intensity, extending our previous model to capture a discretised immunity level. To model transmission-blocking and antidisease immunity, we allow for geometric decay in the respective probabilities of successful human-to-mosquito transmission and symptomatic blood-stage infection as a function of this immunity level. Under a hybrid approximation-whereby probabilistic within-host distributions are cast as expected population-level proportions-we couple host and vector dynamics to recover a deterministic compartmental model in line with Ross-Macdonald theory. We then perform a steady-state analysis for this compartmental model, informed by the (analytic) distributions derived at the within-host level. To characterise transient dynamics, we derive a reduced system of integrodifferential equations, likewise informed by our within-host queueing network, allowing us to recover population-level distributions for various quantities of epidemiological interest. In capturing the interplay between hypnozoite accrual, superinfection and acquired immunity-and providing, to the best of our knowledge, the most complete population-level distributions for a range of epidemiological values-our model provides insights into important, but poorly understood, epidemiological features of P. vivax.


Subject(s)
Epidemiological Models , Malaria, Vivax , Mosquito Vectors , Plasmodium vivax , Humans , Animals , Plasmodium vivax/growth & development , Plasmodium vivax/physiology , Malaria, Vivax/immunology , Malaria, Vivax/parasitology , Malaria, Vivax/transmission , Mosquito Vectors/parasitology , Mosquito Vectors/physiology , Superinfection/immunology , Superinfection/parasitology , Liver/parasitology , Probability
13.
Malar J ; 23(1): 140, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38725027

ABSTRACT

BACKGROUND: Plasmodium vivax relapses due to dormant liver hypnozoites can be prevented with primaquine. However, the dose must be adjusted in individuals with glucose-6-phosphate-dehydrogenase (G6PD) deficiency. In French Guiana, assessment of G6PD activity is typically delayed until day (D)14 to avoid the risk if misclassification. This study assessed the kinetics of G6PD activity throughout P. vivax infection to inform the timing of treatment. METHODS: For this retrospective monocentric study, data on G6PD activity between D1 and D28 after treatment initiation with chloroquine or artemisinin-based combination therapy were collected for patients followed at Cayenne Hospital, French Guiana, between January 2018 and December 2020. Patients were divided into three groups based on the number of available G6PD activity assessments: (i) at least two measurements during the P. vivax malaria infection; (ii) two measurements: one during the current infection and one previously; (iii) only one measurement during the malaria infection. RESULTS: In total, 210 patients were included (80, 20 and 110 in groups 1, 2 and 3, respectively). Data from group 1 showed that G6PD activity remained stable in each patient over time (D1, D3, D7, D14, D21, D28). None of the patients with normal G6PD activity during the initial phase (D1-D3) of the malaria episode (n = 44) was categorized as G6PD-deficient at D14. Patients with G6PD activity < 80% at D1 or D3 showed normal activity at D14. Sex and reticulocyte count were statistically associated with G6PD activity variation. In the whole sample (n = 210), no patient had severe G6PD deficiency (< 10%) and only three between 10 and 30%, giving a G6PD deficiency prevalence of 1.4%. Among the 100 patients from group 1 and 2, 30 patients (26.5%) were lost to follow-up before primaquine initiation. CONCLUSIONS: In patients treated for P. vivax infection, G6PD activity did not vary over time. Therefore, G6PD activity on D1 instead of D14 could be used for primaquine dose-adjustment. This could allow earlier radical treatment with primaquine, that could have a public health impact by decreasing early recurrences and patients lost to follow-up before primaquine initiation. This hypothesis needs to be confirmed in larger prospective studies.


Subject(s)
Antimalarials , Glucosephosphate Dehydrogenase , Malaria, Vivax , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Antimalarials/therapeutic use , Artemisinins/therapeutic use , Chloroquine/therapeutic use , French Guiana/epidemiology , Glucosephosphate Dehydrogenase/metabolism , Glucosephosphate Dehydrogenase Deficiency/epidemiology , Glucosephosphate Dehydrogenase Deficiency/complications , Kinetics , Malaria, Vivax/drug therapy , Plasmodium vivax/drug effects , Plasmodium vivax/physiology , Primaquine/therapeutic use , Retrospective Studies , Aged, 80 and over
14.
Trends Parasitol ; 40(7): 573-590, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38749866

ABSTRACT

Plasmodium vivax contributes significantly to global malaria morbidity. Key advances include the discovery of pathways facilitating invasion by P. vivax merozoites of nascent reticulocytes, crucial for vaccine development. Humanized mouse models and hepatocyte culture systems have enhanced understanding of hypnozoite biology. The spleen has emerged as a major reservoir for asexual vivax parasites, replicating in an endosplenic life cycle, and contributing to recurrent and chronic infections, systemic inflammation, and anemia. Splenic accumulation of uninfected red cells is the predominant cause of anemia. Recurring and chronic infections cause progressive anemia, malnutrition, and death in young children in high-transmission regions. Endothelial activation likely contributes to vivax-associated organ dysfunction. The many recent advances in vivax pathobiology should help guide new approaches to prevention and management.


Subject(s)
Malaria, Vivax , Plasmodium vivax , Humans , Malaria, Vivax/parasitology , Malaria, Vivax/immunology , Malaria, Vivax/physiopathology , Animals , Plasmodium vivax/physiology , Plasmodium vivax/pathogenicity , Spleen/parasitology , Spleen/physiopathology , Spleen/immunology
15.
Trends Parasitol ; 40(7): 562-572, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38806300

ABSTRACT

The geographic origin of Plasmodium vivax, a leading cause of human malaria, has been the subject of much speculation. Here we review the evolutionary history of P. vivax and P. vivax-like parasites in humans and non-human primates on three continents, providing overwhelming evidence for an African origin. This conclusion is consistent with recent reports showing that Duffy-negative humans in Africa are, in fact, susceptible to P. vivax, with parasites invading Duffy-antigen-expressing erythroid precursors. Thus, the African origin of P. vivax not only explains the distribution of the Duffy-negative genotype but also provides new insight into the history and status of P. vivax malaria in Africa and efforts geared toward its eradication.


Subject(s)
Malaria, Vivax , Plasmodium vivax , Plasmodium vivax/physiology , Plasmodium vivax/genetics , Humans , Animals , Malaria, Vivax/parasitology , Africa , Duffy Blood-Group System/genetics , Primates/parasitology
16.
J Vector Borne Dis ; 61(3): 413-419, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38634456

ABSTRACT

BACKGROUND OBJECTIVES: Anopheles stephensi is responsible for the transmission of malaria in urban areas. Vector competence of An. stephensi from a non-malarious (Coimbatore) and highly malarious (Chennai) urban area were investigated to find out the reason for the non-transmission of malaria in Coimbatore. METHODS: Vector competence (Susceptibility/refractoriness) of An. stephensi mosquitoes from Chennai (Malarious) and Coimbatore (Non-malarious), Tamil Nadu, India to Plasmodium vivax (Chennai) were investigated. Bioassays were carried out concurrently in both these strains by artificial membrane feeding technique using the same malaria infected blood. An. stephensi were dissected to observe infection in the midgut and salivary glands. The parasite infection, oocyst and sporozoite positivity rate, the oocyst load, correlation between male-female gametocyte ratio and infection, and Survival Analysis of parasitic stages during sporogony were analyzed and compared. RESULTS: The overall infection rate was 45.8 and 41.2 per cent in Chennai and Coimbatore. Oocyst count ranged from 1-80 and 1-208 respectively and not statistically significant. Oocyst positivity was high from Day 8-21in both strains. The Mean Survival Day (MSD) for oocyst was Day 14 in both strains. Sporozoite was observed in four experiments in each of the strains and the MSD for sporozoites was Day 20 and Day 17 in Chennai and Coimbatore. INTERPRETATION CONCLUSION: An. stephensi of Chennai and Coimbatore are equally susceptible to P. vivax infection and the non-transmission of malaria in Coimbatore can be attributed to external factors such as the presence of preferential breeding habitat, vector density, vector survival, and weather. The only difference observed was the comparatively shortened oocyst maturation time in the Coimbatore strain which requires further investigation.


Subject(s)
Anopheles , Malaria, Vivax , Mosquito Vectors , Plasmodium vivax , Sporozoites , Anopheles/parasitology , Anopheles/physiology , Animals , Plasmodium vivax/physiology , India/epidemiology , Mosquito Vectors/parasitology , Mosquito Vectors/physiology , Female , Malaria, Vivax/parasitology , Malaria, Vivax/transmission , Malaria, Vivax/epidemiology , Male , Sporozoites/physiology , Humans , Oocysts , Salivary Glands/parasitology , Biological Assay
17.
Malar J ; 23(1): 116, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664687

ABSTRACT

BACKGROUND: Pregnancy Associated Malaria (PAM) include malaria in pregnancy (MiP), placental malaria (PM), and congenital malaria (CM). The evidence available in Colombia on PAM focuses on one of the presentations (MiP, PM or CM), and no study longitudinally analyses the infection from the pregnant woman, passing through the placenta, until culminating in the newborn. This study determined the frequency of MiP, PM, and CM caused by Plasmodium vivax, Plasmodium falciparum, or mixed infections, according to Thick Blood Smear (TBS) and quantitative Polymerase Chain Reaction (qPCR). Identifying associated factors of PAM and clinical-epidemiological outcomes in northwestern Colombia. METHODS: Prospective study of 431 pregnant women, their placenta, and newborns registered in the data bank of the research Group "Salud y Comunidad César Uribe Piedrahíta" which collected information between 2014 and 2020 in endemic municipalities of the departments of Córdoba and Antioquia. The frequency of infection was determined with 95% confidence intervals. Comparisons were made with the Chi-square test, Student t-test, prevalence ratios, and control for confounding variables by log-binomial regression. RESULTS: The frequency of MiP was 22.3% (4.6% using TBS), PM 24.8% (1.4% using TBS), and CM 11.8% (0% using TBS). Using TBS predominated P. vivax. Using qPCR the proportions of P. vivax and P. falciparum were similar for MiP and PM, but P. falciparum predominated in CM. The frequency was higher in nulliparous, and women with previous malaria. The main clinical effects of PAM were anaemia, low birth weight, and abnormal APGAR score. CONCLUSIONS: The magnitude of infections was not detected with TBS because most cases were submicroscopic (TBS-negative, qPCR-positive). This confirmed the importance of improving the molecular detection of cases. PAM continue being underestimated in the country due to that in Colombia the control programme is based on TBS, despite its outcomes on maternal, and congenital health.


Subject(s)
Malaria, Falciparum , Malaria, Vivax , Pregnancy Complications, Parasitic , Humans , Female , Pregnancy , Colombia/epidemiology , Prospective Studies , Adult , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Malaria, Vivax/epidemiology , Malaria, Vivax/parasitology , Young Adult , Infant, Newborn , Pregnancy Complications, Parasitic/epidemiology , Pregnancy Complications, Parasitic/parasitology , Adolescent , Plasmodium falciparum/isolation & purification , Prevalence , Plasmodium vivax/isolation & purification , Plasmodium vivax/physiology , Placenta/parasitology , Placenta Diseases/epidemiology , Placenta Diseases/parasitology
18.
Int J Infect Dis ; 143: 107010, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38490637

ABSTRACT

OBJECTIVE: A 15-month longitudinal study was conducted to determine the duration and infectivity of asymptomatic qPCR-detected Plasmodium falciparum and Plasmodium vivax infections in Ethiopia. METHOD: Total parasite and gametocyte kinetics were determined by molecular methods; infectivity to Anopheles arabiensis mosquitoes by repeated membrane feeding assays. Infectivity results were contrasted with passively recruited symptomatic malaria cases. RESULTS: For P. falciparum and P. vivax infections detected at enrolment, median durations of infection were 37 days (95% confidence interval [CI], 15-93) and 60 days (95% CI, 18-213), respectively. P. falciparum and P. vivax parasite densities declined over the course of infections. From 47 feeding assays on 22 asymptomatic P. falciparum infections, 6.4% (3/47) were infectious and these infected 1.8% (29/1579) of mosquitoes. No transmission was observed in feeding assays on asymptomatic P. vivax mono-infections (0/56); one mixed-species infection was highly infectious. Among the symptomatic cases, 4.3% (2/47) of P. falciparum and 73.3% (53/86) of P. vivax patients were infectious to mosquitoes. CONCLUSION: The majority of asymptomatic infections were of short duration and low parasite density. Only a minority of asymptomatic individuals were infectious to mosquitoes. This contrasts with earlier findings and is plausibly due to the low parasite densities in this population.


Subject(s)
Anopheles , Malaria, Falciparum , Malaria, Vivax , Plasmodium falciparum , Plasmodium vivax , Ethiopia/epidemiology , Malaria, Vivax/transmission , Malaria, Vivax/epidemiology , Malaria, Vivax/parasitology , Humans , Longitudinal Studies , Malaria, Falciparum/transmission , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Animals , Plasmodium vivax/isolation & purification , Plasmodium vivax/physiology , Plasmodium falciparum/isolation & purification , Anopheles/parasitology , Male , Female , Adult , Adolescent , Child , Young Adult , Child, Preschool , Asymptomatic Infections/epidemiology , Mosquito Vectors/parasitology , Middle Aged
19.
Eur J Immunol ; 53(8): e2350372, 2023 08.
Article in English | MEDLINE | ID: mdl-37160134

ABSTRACT

Regulatory and effector cell responses to Plasmodium vivax, the most common human malaria parasite outside Africa, remain understudied in naturally infected populations. Here, we describe peripheral CD4+ T- and B-cell populations during and shortly after an uncomplicated P. vivax infection in 38 continuously exposed adult Amazonians. Consistent with previous observations, we found an increased frequency in CD4+ CD45RA- CD25+ FoxP3+ T regulatory cells that express the inhibitory molecule CTLA-4 during the acute infection, with a sustained expansion of CD21- CD27- atypical memory cells within the CD19+ B-cell compartment. Both Th1- and Th2-type subsets of CXCR5+ ICOShi PD-1+ circulating T follicular helper (cTfh) cells, which are thought to contribute to antibody production, were induced during P. vivax infection, with a positive correlation between overall cTfh cell frequency and IgG antibody titers to the P. vivax blood-stage antigen MSP119 . We identified significant changes in cell populations that had not been described in human malaria, such as an increased frequency of CTLA-4+ T follicular regulatory cells that antagonize Tfh cells, and a decreased frequency of circulating CD24hi CD27+ B regulatory cells in response to acute infection. In conclusion, we disclose a complex immunoregulatory network that is critical to understand how naturally acquired immunity develops in P. vivax malaria.


Subject(s)
Malaria, Vivax , Plasmodium vivax , Adult , Humans , Plasmodium vivax/physiology , CTLA-4 Antigen , T-Lymphocytes, Helper-Inducer , CD4-Positive T-Lymphocytes
20.
PLoS Negl Trop Dis ; 16(12): e0010990, 2022 12.
Article in English | MEDLINE | ID: mdl-36534705

ABSTRACT

A key characteristic of Plasmodium vivax parasites is their ability to adopt a latent liver-stage form called hypnozoites, able to cause relapse of infection months or years after a primary infection. Relapses of infection through hypnozoite activation are a major contributor to blood-stage infections in P vivax endemic regions and are thought to be influenced by factors such as febrile infections which may cause temporary changes in hypnozoite activation leading to 'temporal heterogeneity' in reactivation risk. In addition, immunity and variation in exposure to infection may be longer-term characteristics of individuals that lead to 'population heterogeneity' in hypnozoite activation. We analyze data on risk of P vivax in two previously published data sets from Papua New Guinea and the Thailand-Myanmar border region. Modeling different mechanisms of reactivation risk, we find strong evidence for population heterogeneity, with 30% of patients having almost 70% of all P vivax infections. Model fitting and data analysis indicates that individual variation in relapse risk is a primary source of heterogeneity of P vivax risk of recurrences. Trial Registration: ClinicalTrials.gov NCT01640574, NCT01074905, NCT02143934.


Subject(s)
Malaria, Vivax , Parasites , Animals , Humans , Chronic Disease , Liver , Malaria, Vivax/epidemiology , Malaria, Vivax/parasitology , Plasmodium vivax/physiology , Recurrence
SELECTION OF CITATIONS
SEARCH DETAIL