Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Nat Med ; 29(12): 3203-3211, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37884028

ABSTRACT

Anopheles stephensi, an Asian malaria vector, continues to expand across Africa. The vector is now firmly established in urban settings in the Horn of Africa. Its presence in areas where malaria resurged suggested a possible role in causing malaria outbreaks. Here, using a prospective case-control design, we investigated the role of An. stephensi in transmission following a malaria outbreak in Dire Dawa, Ethiopia in April-July 2022. Screening contacts of patients with malaria and febrile controls revealed spatial clustering of Plasmodium falciparum infections around patients with malaria in strong association with the presence of An. stephensi in the household vicinity. Plasmodium sporozoites were detected in these mosquitoes. This outbreak involved clonal propagation of parasites with molecular signatures of artemisinin and diagnostic resistance. To our knowledge, this study provides the strongest evidence so far for a role of An. stephensi in driving an urban malaria outbreak in Africa, highlighting the major public health threat posed by this fast-spreading mosquito.


Subject(s)
Anopheles , Malaria, Falciparum , Malaria , Animals , Humans , Malaria/epidemiology , Malaria/parasitology , Anopheles/parasitology , Mosquito Vectors/parasitology , Malaria, Falciparum/diagnosis , Malaria, Falciparum/epidemiology , Ethiopia/epidemiology
2.
Malar J ; 22(1): 235, 2023 Aug 14.
Article in English | MEDLINE | ID: mdl-37580690

ABSTRACT

BACKGROUND: Urbanization generally improves health outcomes of residents and is one of the potential factors that might contribute to reducing malaria transmission. However, the expansion of Anopheles stephensi, an urban malaria vector, poses a threat for malaria control and elimination efforts in Africa. In this paper, malaria trends in urban settings in Ethiopia from 2014 to 2019 are reported with a focus on towns and cities where An. stephensi surveys were conducted. METHODS: A retrospective study was conducted to determine malaria trends in urban districts using passive surveillance data collected at health facilities from 2014 to 2019. Data from 25 towns surveyed for An. stephensi were used in malaria trend analysis. Robust linear models were used to identify outliers and impute missing and anomalous data. The seasonal Mann-Kendal test was used to test for monotonic increasing or decreasing trends. RESULTS: A total of 9,468,970 malaria cases were reported between 2014 and 2019 through the Public Health Emergency Management (PHEM) system. Of these, 1.45 million (15.3%) cases were reported from urban settings. The incidence of malaria declined by 62% between 2014 and 2018. In 2019, the incidence increased to 15 per 1000 population from 11 to 1000 in 2018. Both confirmed (microscopy or RDT) Plasmodium falciparum (67%) and Plasmodium vivax (28%) were reported with a higher proportion of P. vivax infections in urban areas. In 2019, An. stephensi was detected in 17 towns where more than 19,804 malaria cases were reported, with most of the cases (56%) being P. falciparum. Trend analysis revealed that malaria cases increased in five towns in Afar and Somali administrative regions, decreased in nine towns, and had no obvious trend in the remaining three towns. CONCLUSION: The contribution of malaria in urban settings is not negligible in Ethiopia. With the rapid expansion of An. stephensi in the country, the receptivity is likely to be higher for malaria. Although the evidence presented in this study does not demonstrate a direct linkage between An. stephensi detection and an increase in urban malaria throughout the country, An. stephensi might contribute to an increase in malaria unless control measures are implemented as soon as possible. Targeted surveillance and effective response are needed to assess the contribution of this vector to malaria transmission and curb potential outbreaks.


Subject(s)
Anopheles , Malaria, Falciparum , Malaria, Vivax , Malaria , Animals , Humans , Malaria/epidemiology , Malaria/prevention & control , Malaria/diagnosis , Ethiopia/epidemiology , Anopheles/physiology , Retrospective Studies , Mosquito Vectors , Malaria, Falciparum/epidemiology , Malaria, Vivax/epidemiology
4.
Malar J ; 21(1): 359, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36451216

ABSTRACT

BACKGROUND: Routine monitoring of anti-malarial drugs is recommended for early detection of drug resistance and to inform national malaria treatment guidelines. In Ethiopia, the national treatment guidelines employ a species-specific approach. Artemether-lumefantrine (AL) and chloroquine (CQ) are the first-line schizonticidal treatments for Plasmodium falciparum and Plasmodium vivax, respectively. The National Malaria Control and Elimination Programme in Ethiopia is considering dihydroartemisinin-piperaquine (DHA/PPQ) as an alternative regimen for P. falciparum and P. vivax. METHODS: The study assessed the clinical and parasitological efficacy of AL, CQ, and DHA/PPQ in four arms. Patients over 6 months and less than 18 years of age with uncomplicated malaria mono-infection were recruited and allocated to AL against P. falciparum and CQ against P. vivax. Patients 18 years or older with uncomplicated malaria mono-infection were recruited and randomized to AL or dihydroartemisinin-piperaquine (DHA/PPQ) against P. falciparum and CQ or DHA/PPQ for P. vivax. Patients were followed up for 28 (for CQ and AL) or 42 days (for DHA/PPQ) according to the WHO recommendations. Polymerase chain reaction (PCR)-corrected and uncorrected estimates were analysed by Kaplan Meier survival analysis and per protocol methods. RESULTS: A total of 379 patients were enroled in four arms (n = 106, AL-P. falciparum; n = 75, DHA/PPQ- P. falciparum; n = 142, CQ-P. vivax; n = 56, DHA/PPQ-P. vivax). High PCR-corrected adequate clinical and parasitological response (ACPR) rates were observed at the primary end points of 28 days for AL and CQ and 42 days for DHA/PPQ. ACPR rates were 100% in AL-Pf (95% CI: 96-100), 98% in CQ-P. vivax (95% CI: 95-100) at 28 days, and 100% in the DHA/PPQ arms for both P. falciparum and P. vivax at 42 days. For secondary endpoints, by day three 99% of AL-P. falciparum patients (n = 101) cleared parasites and 100% were afebrile. For all other arms, 100% of patients cleared parasites and were afebrile by day three. No serious adverse events were reported. CONCLUSION: This study demonstrated high therapeutic efficacy for the anti-malarial drugs currently used by the malaria control programme in Ethiopia and provides information on the efficacy of DHA/PPQ for the treatment of P. falciparum and P. vivax as an alternative option.


Subject(s)
Antimalarials , Artemisinins , Malaria, Falciparum , Malaria, Vivax , Humans , Artemether, Lumefantrine Drug Combination/therapeutic use , Chloroquine/therapeutic use , Plasmodium falciparum , Antimalarials/therapeutic use , Plasmodium vivax , Ethiopia , Artemether , Artemisinins/therapeutic use , Malaria, Vivax/drug therapy , Malaria, Falciparum/drug therapy
5.
Trials ; 23(1): 267, 2022 Apr 07.
Article in English | MEDLINE | ID: mdl-35392979

ABSTRACT

BACKGROUND: Reactive and proactive case detection measures are widely implemented by national malaria elimination programs globally. Ethiopia decided to include Reactive Case Detection (RCD) and targeted Mass Drug Administration (tMDA) approaches as part of their elimination strategy along with rigorous evaluation. The purpose of this study is to compare the impact of RCD and tMDA on malaria elimination over the 2-year study period, by looking at the annual parasite incidence before and after the intervention. METHODS: The study will be conducted in the East Hararghe zone of Ethiopia. Malaria transmission in the area is low to moderate. This study will deploy a community-based, three-arm, cluster-randomized control trial implemented over 2 years. Forty-eight clusters (16 clusters per arm) will be selected based on the annual number of confirmed malaria cases seen in the cluster. All clusters will receive the current standard of care in terms of malaria elimination interventions provided by the national malaria control program. In addition, following the identification of malaria parasite infection, individuals who reside within a 100-m radius of the index case will receive a diagnosis for malaria and treatment if positive in the RCD arm or presumptive treatment in the tMDA arm. The primary effectiveness endpoint will be measured at baseline and endline for each intervention arm and compared to the control arm using a difference in difference approach. DISCUSSION: This randomized controlled trial will provide evidence of the impact of the proposed intervention approaches for malaria elimination. TRIAL REGISTRATION: ClinicalTrials.gov NCT04241705 . Registration date: January 27, 2020.


Subject(s)
Antimalarials , Malaria , Antimalarials/adverse effects , Ethiopia/epidemiology , Humans , Incidence , Malaria/diagnosis , Malaria/drug therapy , Malaria/epidemiology , Mass Drug Administration
6.
BMC Public Health ; 21(1): 1996, 2021 11 03.
Article in English | MEDLINE | ID: mdl-34732150

ABSTRACT

BACKGROUND: Encouraged by the previous success in malaria control and prevention strategies, the Ethiopian ministry of health launched malaria elimination with a stepwise approach by primarily targeting the low-transmission Districts and their adjacent areas/zones in order to shrink the country's malaria map progressively. Hence, this community survey was conducted to establish baseline malaria information at the preliminary phase of elimination at targeted settings. METHODS: A community-based cross-sectional survey was conducted at 20 malaria-elimination targeted Districts selected from five Regional states and one city administration in Ethiopia. The GPS-enabled smartphones programmed with Open Data Kit were used to enumerate 9326 study households and collect data from 29,993 residents. CareStart™ Malaria PAN (pLDH) Rapid Diagnostic Tests (RDTs) were used for blood testing at the field level. Armpit digital thermometers were used to measure axillary temperature. RESULT: Overall malaria prevalence by RDTs was 1.17% (339/28973). The prevalence at District levels ranged from 0.0 to 4.7%. The proportion of symptomatic cases (axillary temperature > 37.5oc) in the survey was 9.2% (2760/29993). Among the 2510 symptomatic individuals tested with RDTs, only 3.35% (84/2510) were malaria positive. The 75.2% (255/339) of all malaria positives were asymptomatic. Of the total asymptomatic malaria cases, 10.2% (26/255) were under-five children and 89.8% (229/255) were above 5 years of age. CONCLUSION: The study shows a decrease in malaria prevalence compared to the reports of previous malaria indicator surveys in the country. The finding can be used as a baseline for measuring the achievement of ongoing malaria elimination efforts. Particularly, the high prevalence of asymptomatic individuals (0.88%) in these transmission settings indicates there may be sustaining hidden transmission. Therefore, active case detection with more sensitive diagnostic techniques is suggested to know more real magnitude of residual malaria in the elimination-targeted areas.


Subject(s)
Malaria, Falciparum , Malaria , Child , Cross-Sectional Studies , Diagnostic Tests, Routine , Ethiopia/epidemiology , Humans , Malaria/diagnosis , Malaria/epidemiology , Malaria/prevention & control , Prevalence
8.
Malar J ; 20(1): 263, 2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34107943

ABSTRACT

BACKGROUND: Anopheles stephensi, an invasive malaria vector, was first detected in Africa nearly 10 years ago. After the initial finding in Djibouti, it has subsequently been found in Ethiopia, Sudan and Somalia. To better inform policies and vector control decisions, it is important to understand the distribution, bionomics, insecticide susceptibility, and transmission potential of An. stephensi. These aspects were studied as part of routine entomological monitoring in Ethiopia between 2018 and 2020. METHODS: Adult mosquitoes were collected using human landing collections, pyrethrum spray catches, CDC light traps, animal-baited tent traps, resting boxes, and manual aspiration from animal shelters. Larvae were collected using hand-held dippers. The source of blood in blood-fed mosquitoes and the presence of sporozoites was assessed through enzyme-linked immunosorbent assays (ELISA). Insecticide susceptibility was assessed for pyrethroids, organophosphates and carbamates. RESULTS: Adult An. stephensi were collected with aspiration, black resting boxes, and animal-baited traps collecting the highest numbers of mosquitoes. Although sampling efforts were geographically widespread, An. stephensi larvae were collected in urban and rural sites in eastern Ethiopia, but An. stephensi larvae were not found in western Ethiopian sites. Blood-meal analysis revealed a high proportion of blood meals that were taken from goats, and only a small proportion from humans. Plasmodium vivax was detected in wild-collected An. stephensi. High levels of insecticide resistance were detected to pyrethroids, carbamates and organophosphates. Pre-exposure to piperonyl butoxide increased susceptibility to pyrethroids. Larvae were found to be susceptible to temephos. CONCLUSIONS: Understanding the bionomics, insecticide susceptibility and distribution of An. stephensi will improve the quality of a national response in Ethiopia and provide additional information on populations of this invasive species in Africa. Further work is needed to understand the role that An. stephensi will have in Plasmodium transmission and malaria case incidence. While additional data are being collected, national programmes can use the available data to formulate and operationalize national strategies against the threat of An. stephensi.


Subject(s)
Animal Distribution , Anopheles/physiology , Insecticide Resistance , Life History Traits , Animals , Anopheles/growth & development , Ethiopia , Insecticides/pharmacology , Larva/growth & development , Larva/physiology , Malaria/transmission
9.
Malar J ; 20(1): 224, 2021 May 19.
Article in English | MEDLINE | ID: mdl-34011347

ABSTRACT

BACKGROUND: Sixty percent of the Ethiopia population is at risk of malaria, with the highest prevalence reported in Gambella (6%) and Benishangul-Gumuz (3%) regions. Within these regions are large agricultural developments with high numbers of seasonal migrant workers. The migrant workers are believed to be at increased risk for malaria infection due to their poor living conditions and outdoor activities, but there is little information on their specific behaviours and health risks. This study was conducted to address this gap. METHODS: Quantitative observations were conducted from September to December 2017 in the Benishangul-Gumuz Region. The nightly routines of mobile migrant workers were observed every month for 4 consecutive months. The study team collected quantitative data including nocturnal behavioural observations of worker living conditions, malaria prevention efforts, and work activities and surveys of worker representatives. Qualitative data was collected from migrant workers, farm managers and local health providers using focus group discussions and semi-structured interviews. RESULTS: Migrant workers arrived in the study area during the peak malaria transmission season and the workers in focus groups reported repeated cases of malaria during their stay on the farms. Overall, less than a quarter of the migrant workers were sleeping under a mosquito net by midnight in all 4 observation months. Some work activities also took place outdoors at night. The study additionally found a lack of access to malaria prevention and treatment at the farms and challenges in utilizing local public health facilities. CONCLUSIONS: There is a need to better address malaria prevention and treatment needs among migrant workers in Ethiopia through outreach from existing healthcare infrastructure and within the farms themselves. This will help prevent malaria transmission both within this population and prevent transmission of malaria back to home communities in lower burden areas in Ethiopia.


Subject(s)
Farmers/statistics & numerical data , Malaria/prevention & control , Transients and Migrants/statistics & numerical data , Ethiopia , Humans
10.
Emerg Infect Dis ; 27(2): 603-607, 2021 02.
Article in English | MEDLINE | ID: mdl-33496217

ABSTRACT

Anopheles stephensi mosquitoes, efficient vectors in parts of Asia and Africa, were found in 75.3% of water sources surveyed and contributed to 80.9% of wild-caught Anopheles mosquitoes in Awash Sebat Kilo, Ethiopia. High susceptibility of these mosquitoes to Plasmodium falciparum and vivax infection presents a challenge for malaria control in the Horn of Africa.


Subject(s)
Anopheles , Plasmodium vivax , Animals , Asia , Ethiopia , Mosquito Vectors , Plasmodium falciparum
11.
Malar J ; 19(1): 108, 2020 Mar 04.
Article in English | MEDLINE | ID: mdl-32131841

ABSTRACT

BACKGROUND: Ethiopia has set a goal for malaria elimination by 2030. Low parasite density infections may go undetected by conventional diagnostic methods (microscopy and rapid diagnostic tests) and their contribution to malaria transmission varies by transmission settings. This study quantified the burden of subpatent infections from samples collected from three regions of northwest Ethiopia. METHODS: Sub-samples of dried blood spots from the Ethiopian Malaria Indicator Survey 2015 (EMIS-2015) were tested and compared using microscopy, rapid diagnostic tests (RDTs), and nested polymerase chain reaction (nPCR) to determine the prevalence of subpatent infection. Paired seroprevalence results previously reported along with gender, age, and elevation of residence were explored as risk factors for Plasmodium infection. RESULTS: Of the 2608 samples collected, the highest positive rate for Plasmodium infection was found with nPCR 3.3% (95% CI 2.7-4.1) compared with RDT 2.8% (95% CI 2.2-3.5) and microscopy 1.2% (95% CI 0.8-1.7). Of the nPCR positive cases, Plasmodium falciparum accounted for 3.1% (95% CI 2.5-3.8), Plasmodium vivax 0.4% (95% CI 0.2-0.7), mixed P. falciparum and P. vivax 0.1% (95% CI 0.0-0.4), and mixed P. falciparum and Plasmodium malariae 0.1% (95% CI 0.0-0.3). nPCR detected an additional 30 samples that had not been detected by conventional methods. The majority of the nPCR positive cases (61% (53/87)) were from the Benishangul-Gumuz Region. Malaria seropositivity had significant association with nPCR positivity [adjusted OR 10.0 (95% CI 3.2-29.4), P < 0.001]. CONCLUSION: Using nPCR the detection rate of malaria parasites increased by nearly threefold over rates based on microscopy in samples collected during a national cross-sectional survey in 2015 in Ethiopia. Such subpatent infections might contribute to malaria transmission. In addition to strengthening routine surveillance systems, malaria programmes may need to consider low-density, subpatent infections in order to accelerate malaria elimination efforts.


Subject(s)
Disease Eradication/methods , Malaria, Falciparum/epidemiology , Malaria, Vivax/epidemiology , Adolescent , Adult , Child , Child, Preschool , Cross-Sectional Studies , Dried Blood Spot Testing , Ethiopia/epidemiology , Female , Humans , Malaria, Falciparum/diagnosis , Malaria, Falciparum/prevention & control , Malaria, Vivax/diagnosis , Malaria, Vivax/prevention & control , Male , Middle Aged , Plasmodium falciparum , Plasmodium vivax , Prevalence , Seroepidemiologic Studies , Young Adult
12.
Malar J ; 18(1): 246, 2019 Jul 22.
Article in English | MEDLINE | ID: mdl-31331340

ABSTRACT

BACKGROUND: Measures of malaria burden using microscopy and rapid diagnostic tests (RDTs) in cross-sectional household surveys may incompletely describe the burden of malaria in low-transmission settings. This study describes the pattern of malaria transmission in Ethiopia using serological antibody estimates derived from a nationwide household survey completed in 2015. METHODS: Dried blood spot (DBS) samples were collected during the Ethiopian Malaria Indicator Survey in 2015 from malarious areas across Ethiopia. Samples were analysed using bead-based multiplex assays for IgG antibodies for six Plasmodium antigens: four human malaria species-specific merozoite surface protein-1 19kD antigens (MSP-1) and Apical Membrane Antigen-1 (AMA-1) for Plasmodium falciparum and Plasmodium vivax. Seroprevalence was estimated by age, elevation and region. The seroconversion rate was estimated using a reversible catalytic model fitted with maximum likelihood methods. RESULTS: Of the 10,278 DBS samples available, 93.6% (9622/10,278) had valid serological results. The mean age of participants was 15.8 years and 53.3% were female. National seroprevalence for antibodies to P. falciparum was 32.1% (95% confidence interval (CI) 29.8-34.4) and 25.0% (95% CI 22.7-27.3) to P. vivax. Estimated seroprevalences for Plasmodium malariae and Plasmodium ovale were 8.6% (95% CI 7.6-9.7) and 3.1% (95% CI 2.5-3.8), respectively. For P. falciparum seroprevalence estimates were significantly higher at lower elevations (< 2000 m) compared to higher (2000-2500 m) (aOR 4.4; p < 0.01). Among regions, P. falciparum seroprevalence ranged from 11.0% (95% CI 8.8-13.7) in Somali to 65.0% (95% CI 58.0-71.4) in Gambela Region and for P. vivax from 4.0% (95% CI 2.6-6.2) in Somali to 36.7% (95% CI 30.0-44.1) in Amhara Region. Models fitted to measure seroconversion rates showed variation nationally and by elevation, region, antigen type, and within species. CONCLUSION: Using multiplex serology assays, this study explored the cumulative malaria burden and regional dynamics of the four human malarias in Ethiopia. High malaria burden was observed in the northwest compared to the east. High transmission in the Gambela and Benishangul-Gumuz Regions and the neglected presence of P. malariae and P. ovale may require programmatic attention. The use of a multiplex assay for antibody detection in low transmission settings has the potential to act as a more sensitive biomarker.


Subject(s)
Malaria/epidemiology , Plasmodium/isolation & purification , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Protozoan , Child , Child, Preschool , Ethiopia/epidemiology , Female , Humans , Immunoglobulin G/analysis , Infant , Infant, Newborn , Malaria/classification , Male , Middle Aged , Plasmodium/classification , Prevalence , Seroepidemiologic Studies , Serologic Tests , Young Adult
14.
Malar J ; 17(1): 388, 2018 Oct 26.
Article in English | MEDLINE | ID: mdl-30367627

ABSTRACT

BACKGROUND: Building on the declining trend of malaria in Ethiopia, the Federal Ministry of Health aims to eliminate malaria by 2030. As Plasmodium falciparum and Plasmodium vivax are co-endemic in Ethiopia, the use of primaquine is indicated for both transmission interruption and radical cure, respectively. However, the limited knowledge of the local prevalence of glucose-6-phosphate dehydrogenase (G6PD) deficiency and its associated variants has hindered the use of primaquine. METHODS: Some 11,138 dried blood spot (DBS) samples were collected in 2011 as part of a national, household Malaria Indicator Survey, a multi-stage nationally representative survey of all malaria-endemic areas of Ethiopia. A randomly selected sub-set of 1414 DBS samples was successfully genotyped by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) technique. Considering the geographical position and ethnic mix of the country, three common variants: G6PD*A (A376G), G6PD*A- (G202A) and Mediterranean (C563T) were investigated. RESULTS: Of the 1998 randomly selected individuals, 1429 (71.5%) DBS samples were genotyped and merged to the database, of which 53.5% were from females. G6PD*A (A376G) was the only genotype detected. No sample was positive for either G6PD*A- (G202A) or Mediterranean (C563T) variants. The prevalence of G6PD*A (A376G) was 8.9% [95% confidence interval (CI) 6.7-11.2] ranging from 12.2% in the Southern Nations, Nationalities and Peoples' (95% CI 5.7-18.7) to none in Dire Dawa/Harari Region. CONCLUSION: The common G6PD*A- (G202A) or Mediterranean (C563T) variants were not observed in this nationwide study. The observed G6PD*A (A376G) mutation has little or no clinical significance. These findings supported the adoption of primaquine for P. falciparum transmission interruption and radical cure of P. vivax in Ethiopia. As the presence of other clinically important, less common variants cannot be ruled out, the implementation of radical cure will be accompanied by active haematological and adverse events monitoring in Ethiopia.


Subject(s)
Genotype , Glucosephosphate Dehydrogenase Deficiency/epidemiology , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Child , Child, Preschool , Endemic Diseases , Ethiopia/epidemiology , Female , Glucosephosphate Dehydrogenase Deficiency/genetics , Humans , Infant , Malaria, Falciparum/epidemiology , Malaria, Vivax/epidemiology , Male , Middle Aged , Prevalence , Young Adult
15.
PLoS Med ; 14(5): e1002299, 2017 05.
Article in English | MEDLINE | ID: mdl-28510573

ABSTRACT

BACKGROUND: Recent efforts in malaria control have resulted in great gains in reducing the burden of Plasmodium falciparum, but P. vivax has been more refractory. Its ability to form dormant liver stages confounds control and elimination efforts. To compare the efficacy and safety of primaquine regimens for radical cure, we undertook a randomized controlled trial in Ethiopia. METHODS AND FINDINGS: Patients with normal glucose-6-phosphate dehydrogenase status with symptomatic P. vivax mono-infection were enrolled and randomly assigned to receive either chloroquine (CQ) or artemether-lumefantrine (AL), alone or in combination with 14 d of semi-supervised primaquine (PQ) (3.5 mg/kg total). A total of 398 patients (n = 104 in the CQ arm, n = 100 in the AL arm, n = 102 in the CQ+PQ arm, and n = 92 in the AL+PQ arm) were followed for 1 y, and recurrent episodes were treated with the same treatment allocated at enrolment. The primary endpoints were the risk of P. vivax recurrence at day 28 and at day 42. The risk of recurrent P. vivax infection at day 28 was 4.0% (95% CI 1.5%-10.4%) after CQ treatment and 0% (95% CI 0%-4.0%) after CQ+PQ. The corresponding risks were 12.0% (95% CI 6.8%-20.6%) following AL alone and 2.3% (95% CI 0.6%-9.0%) following AL+PQ. On day 42, the risk was 18.7% (95% CI 12.2%-28.0%) after CQ, 1.2% (95% CI 0.2%-8.0%) after CQ+PQ, 29.9% (95% CI 21.6%-40.5%) after AL, and 5.9% (95% CI 2.4%-13.5%) after AL+PQ (overall p < 0.001). In those not prescribed PQ, the risk of recurrence by day 42 appeared greater following AL treatment than CQ treatment (HR = 1.8 [95% CI 1.0-3.2]; p = 0.059). At the end of follow-up, the incidence rate of P. vivax was 2.2 episodes/person-year for patients treated with CQ compared to 0.4 for patients treated with CQ+PQ (rate ratio: 5.1 [95% CI 2.9-9.1]; p < 0.001) and 2.3 episodes/person-year for AL compared to 0.5 for AL+PQ (rate ratio: 6.4 [95% CI 3.6-11.3]; p < 0.001). There was no difference in the occurrence of adverse events between treatment arms. The main limitations of the study were the early termination of the trial and the omission of haemoglobin measurement after day 42, resulting in an inability to estimate the cumulative risk of anaemia. CONCLUSIONS: Despite evidence of CQ-resistant P. vivax, the risk of recurrence in this study was greater following treatment with AL unless it was combined with a supervised course of PQ. PQ combined with either CQ or AL was well tolerated and reduced recurrence of vivax malaria by 5-fold at 1 y. TRIAL REGISTRATION: ClinicalTrials.gov NCT01680406.


Subject(s)
Artemisinins/therapeutic use , Chloroquine/therapeutic use , Ethanolamines/therapeutic use , Fluorenes/therapeutic use , Malaria, Vivax/prevention & control , Primaquine/therapeutic use , Adolescent , Adult , Antimalarials/pharmacology , Antimalarials/therapeutic use , Artemether, Lumefantrine Drug Combination , Artemisinins/pharmacology , Child , Child, Preschool , Chloroquine/pharmacology , Drug Combinations , Ethanolamines/pharmacology , Ethiopia , Female , Fluorenes/pharmacology , Humans , Infant , Male , Plasmodium vivax/drug effects , Primaquine/pharmacology , Young Adult
16.
Malar J ; 14: 501, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26671012

ABSTRACT

BACKGROUND: Malaria and HIV/AIDS constitute major public health problems in Ethiopia, but the burden associated with malaria-HIV co-infection has not been well documented. In this study, the burden of malaria among HIV positive and HIV negative adult outpatients attending health facilities in Oromia National Regional State, Ethiopia was investigated. METHODS: A comparative cross-sectional study among HIV-positive patients having routine follow-up visits at HIV care and treatment clinics and HIV-seronegative patients attending the general medical outpatient departments in 12 health facilities during the peak malaria transmission season was conducted from September to November, 2011. A total of 3638 patients (1819 from each group) were enrolled in the study. Provider initiated testing and counseling of HIV was performed for 1831 medical outpatients out of whom 1819 were negative and enrolled into the study. Malaria blood microscopy and hemoglobin testing were performed for all 3638 patients. Data was analyzed using descriptive statistics, Chi square test and multivariate logistic regression. RESULTS: Of the 3638 patients enrolled in the study, malaria parasitaemia was detected in 156 (4.3%); malaria parasitaemia prevalence was 0.7% (13/1819) among HIV-seropositive patients and 7.9% (143/1819) among HIV-seronegative patients. Among HIV-seropositive individuals 65.4% slept under a mosquito bed net the night before data collection, compared to 59.4% of HIV-seronegative individuals. A significantly higher proportion of HIV-seropositive malaria-negative patients were on co-trimoxazole (CTX) prophylaxis as compared to HIV-malaria co-infected patients: 82% (1481/1806) versus 46% (6/13) (P = 0.001). HIV and malaria co-infected patients were less likely to have the classical symptoms of malaria (fever, chills and headache) compared to the HIV-seronegative and malaria positive counterparts. Multivariate logistic regression showed that HIV-seropositive patients who come for routine follow up were less likely to be infected by malaria (OR = 0.23, 95% CI = 0.09-0.74). CONCLUSION: The study documented lower malaria prevalence among the HIV-seropositive attendants who come for routine follow up. Clinical symptoms of malaria were more pronounced among HIV-seronegative than HIV-seropositive patients. This study also re-affirmed the importance of co-trimoxazole in preventing malaria symptoms and parasitaemia among HIV-positive patients.


Subject(s)
Coinfection/epidemiology , HIV Infections/complications , Malaria/epidemiology , Outpatients , Adolescent , Adult , Aged , Aged, 80 and over , Anti-Infective Agents/therapeutic use , Chemoprevention , Cross-Sectional Studies , Ethiopia/epidemiology , Female , Humans , Malaria/prevention & control , Male , Middle Aged , Prevalence , Trimethoprim, Sulfamethoxazole Drug Combination/therapeutic use , Young Adult
17.
Am J Trop Med Hyg ; 93(1): 168-177, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25962770

ABSTRACT

Ethiopia has a diverse ecology and geography resulting in spatial and temporal variation in malaria transmission. Evidence-based strategies are thus needed to monitor transmission intensity and target interventions. A purposive selection of dried blood spots collected during cross-sectional school-based surveys in Oromia Regional State, Ethiopia, were tested for presence of antibodies against Plasmodium falciparum and P. vivax antigens. Spatially explicit binomial models of seroprevalence were created for each species using a Bayesian framework, and used to predict seroprevalence at 5 km resolution across Oromia. School seroprevalence showed a wider prevalence range than microscopy for both P. falciparum (0-50% versus 0-12.7%) and P. vivax (0-53.7% versus 0-4.5%), respectively. The P. falciparum model incorporated environmental predictors and spatial random effects, while P. vivax seroprevalence first-order trends were not adequately explained by environmental variables, and a spatial smoothing model was developed. This is the first demonstration of serological indicators being used to detect large-scale heterogeneity in malaria transmission using samples from cross-sectional school-based surveys. The findings support the incorporation of serological indicators into periodic large-scale surveillance such as Malaria Indicator Surveys, and with particular utility for low transmission and elimination settings.


Subject(s)
Antibodies, Protozoan/immunology , Endemic Diseases/statistics & numerical data , Malaria, Falciparum/epidemiology , Malaria, Vivax/epidemiology , Plasmodium falciparum/immunology , Plasmodium vivax/immunology , Adolescent , Antigens, Protozoan/immunology , Bayes Theorem , Child , Child, Preschool , Cross-Sectional Studies , Enzyme-Linked Immunosorbent Assay , Ethiopia/epidemiology , Humans , Malaria, Falciparum/immunology , Malaria, Vivax/immunology , Male , Membrane Proteins/immunology , Merozoite Surface Protein 1/immunology , Multivariate Analysis , Plasmodium falciparum/isolation & purification , Plasmodium vivax/isolation & purification , Protozoan Proteins/immunology , Schools , Seroepidemiologic Studies , Spatial Analysis
18.
Malar J ; 13: 292, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25073561

ABSTRACT

BACKGROUND: Accurate early diagnosis and prompt treatment is one of the key strategies to control and prevent malaria in Ethiopia where both Plasmodium falciparum and Plasmodium vivax are sympatric and require different treatment regimens. Microscopy is the standard for malaria diagnosis at the health centres and hospitals whereas rapid diagnostic tests are used at community-level health posts. The current study was designed to assess malaria microscopy capacity of health facilities in Oromia Regional State and Dire Dawa Administrative City, Ethiopia. METHODS: A descriptive cross-sectional study was conducted from February to April 2011 in 122 health facilities, where health professionals were interviewed using a pre-tested, standardized assessment tool and facilities' laboratory practices were assessed by direct observation. RESULTS: Of the 122 assessed facilities, 104 (85%) were health centres and 18 (15%) were hospitals. Out of 94 health facilities reportedly performing blood films, only 34 (36%) used both thin and thick smears for malaria diagnosis. The quality of stained slides was graded in 66 health facilities as excellent, good and poor quality in 11(17%), 31 (47%) and 24 (36%) respectively. Quality assurance guidelines and malaria microscopy standard operating procedures were found in only 13 (11%) facilities and 12 (10%) had involved in external quality assessment activities, and 32 (26%) had supportive supervision within six months of the survey. Only seven (6%) facilities reported at least one staff's participation in malaria microscopy refresher training during the previous 12 months. Although most facilities, 96 (79%), had binocular microscopes, only eight (7%) had the necessary reagents and supplies to perform malaria microscopy. Treatment guidelines for malaria were available in only 38 (31%) of the surveyed facilities. Febrile patients with negative malaria laboratory test results were managed with artemether-lumefantrine or chloroquine in 51% (53/104) of assessed health facilities. CONCLUSIONS: The current study indicated that most of the health facilities had basic infrastructure and equipment to perform malaria laboratory diagnosis but with significant gaps in continuous laboratory supplies and reagents, and lack of training and supportive supervision. Overcoming these gaps will be critical to ensure that malaria laboratory diagnosis is of high-quality for better patient management.


Subject(s)
Clinical Laboratory Techniques/statistics & numerical data , Community Health Centers/statistics & numerical data , Malaria, Falciparum/diagnosis , Malaria, Vivax/diagnosis , Parasitology/statistics & numerical data , Clinical Laboratory Techniques/instrumentation , Clinical Laboratory Techniques/standards , Cross-Sectional Studies , Ethiopia/epidemiology , Humans , Malaria, Falciparum/epidemiology , Malaria, Falciparum/prevention & control , Malaria, Vivax/epidemiology , Malaria, Vivax/prevention & control
19.
Trop Med Int Health ; 17(10): 1192-201, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22863170

ABSTRACT

To understand the drivers and consequences of malaria in epidemic-prone regions, it is important to know whether epidemics emerge independently in different areas as a consequence of local contingencies, or whether they are synchronised across larger regions as a result of climatic fluctuations and other broad-scale drivers. To address this question, we collected historical malaria surveillance data for the Amhara region of Ethiopia and analysed them to assess the consistency of various indicators of malaria risk and determine the dominant spatial and temporal patterns of malaria within the region. We collected data from a total of 49 districts from 1999-2010. Data availability was better for more recent years and more data were available for clinically diagnosed outpatient malaria cases than confirmed malaria cases. Temporal patterns of outpatient malaria case counts were correlated with the proportion of outpatients diagnosed with malaria and confirmed malaria case counts. The proportion of outpatients diagnosed with malaria was spatially clustered, and these cluster locations were generally consistent from year to year. Outpatient malaria cases exhibited spatial synchrony at distances up to 300 km, supporting the hypothesis that regional climatic variability is an important driver of epidemics. Our results suggest that decomposing malaria risk into separate spatial and temporal components may be an effective strategy for modelling and forecasting malaria risk across large areas. They also emphasise both the value and limitations of working with historical surveillance datasets and highlight the importance of enhancing existing surveillance efforts.


Subject(s)
Climate , Epidemics , Malaria/epidemiology , Ethiopia/epidemiology , Humans , Population Surveillance , Risk Factors
20.
Int Health ; 4(1): 70-3, 2012 Mar.
Article in English | MEDLINE | ID: mdl-24030883

ABSTRACT

Little is known about the contribution of the private health sector in managing malaria cases and contributing to malaria prevention and control efforts in Ethiopia. We assessed 102 private health facilities and 92 drug outlets in 20 districts of Oromia Regional State, Ethiopia, for their provision of malaria-specific services. Of the assessed health facilities 86% provided such services. Diagnosis was largely clinical, with only 31% and 15% of all health facilities seen using rapid diagnostic tests and microscopy, respectively. Facilities had chloroquine, artemether-lumefantrine, quinine and sulfadoxine-pyremethamine. Gaps were seen in provision of guidelines and other malaria-related materials, training of facility staff and supervision. Inclusion of the private health sector in malaria control program is crucial to expand current malaria prevention and control efforts in Ethiopia.

SELECTION OF CITATIONS
SEARCH DETAIL
...