Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
Lancet Microbe ; 5(9): 100892, 2024 Sep.
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.


Assuntos
Amodiaquina , Antimaláricos , Quimioprevenção , Combinação de Medicamentos , Resistência a Medicamentos , Malária Falciparum , Plasmodium falciparum , Pirimetamina , Estações do Ano , Sulfadoxina , Sulfadoxina/uso terapêutico , Sulfadoxina/farmacologia , Pirimetamina/farmacologia , Pirimetamina/uso terapêutico , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Humanos , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/genética , Pré-Escolar , Malária Falciparum/prevenção & controle , Malária Falciparum/epidemiologia , Malária Falciparum/transmissão , Malária Falciparum/parasitologia , Resistência a Medicamentos/genética , Criança , Lactente , Amodiaquina/uso terapêutico , Amodiaquina/farmacologia , Mutação , Feminino
2.
Lancet Microbe ; 5(9): 100891, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39068937

RESUMO

BACKGROUND: The majority of Plasmodium spp infections in endemic countries are asymptomatic and a source of onward transmission to mosquitoes. We aimed to examine whether Plasmodium falciparum transmission and malaria burden could be reduced by improving early detection and treatment of infections with active screening approaches. METHODS: In this 18-month cluster randomised study in Sapone, Burkina Faso, households were enrolled and randomly assigned (1:1:1) to one of three groups: group 1 (control) received standard of care only, group 2 received active weekly, at home, fever screening by a community health worker regardless of symptoms, participants with a fever received a rapid diagnostic test (RDT) and treatment if RDT positive, and group 3 received active weekly fever screening (as in group 2) plus a monthly RDT regardless of symptoms, and treatment if RDT positive. Eligible households had a minimum of three eligible residents, one in each age group (<5 years, 5-15 years, and >15 years). The primary outcome was parasite prevalence by quantitative PCR (qPCR) in the end-of-study cross-sectional survey. Secondary outcomes included parasite and gametocyte prevalence and density in all three end-of-season cross-sectional surveys, incidence of infection, and the transmissibility of infections to mosquitoes. This trial was registered at ClinicalTrials.gov (NCT03705624) and is completed. FINDINGS: A total of 906 individuals from 181 households were enrolled during two phases, and participated in the study. 412 individuals were enrolled between Aug 9 and 17, 2018, and participated in phase 1 and 494 individuals were enrolled between Jan 10 and 31, 2019, in phase 2. In the end-of-study cross-sectional survey (conducted between Jan 13 and 21, 2020), Pfalciparum prevalence by qPCR was significantly lower in group 3 (29·26%; 79 of 270), but not in group 2 (45·66%; 121 of 265), when compared with group 1 (48·72%; 133 of 273; risk ratio 0·65 [95% CI 0·52-0·81]; p=0·0001). Total parasite and gametocyte prevalence and density were also significantly lower in group 3 in all surveys. The largest differences were seen at the end of the dry season, with gametocyte prevalence 78·4% and predicted transmission potential 98·2% lower in group 3 than in group 1. INTERPRETATION: Active monthly RDT testing and treatment can reduce parasite carriage and the infectious reservoir of P falciparum to less than 2% when used during the dry season. This insight might inform approaches for malaria control and elimination. FUNDING: Bill & Melinda Gates Foundation, European Research Council, and The Netherlands Organization for Scientific Research.


Assuntos
Febre , Malária Falciparum , Plasmodium falciparum , Humanos , Burkina Faso/epidemiologia , Masculino , Criança , Pré-Escolar , Adolescente , Feminino , Malária Falciparum/epidemiologia , Malária Falciparum/diagnóstico , Malária Falciparum/transmissão , Plasmodium falciparum/isolamento & purificação , Febre/epidemiologia , Estudos Transversais , Adulto , Prevalência , Testes Diagnósticos de Rotina , Antimaláricos/uso terapêutico , Lactente , Programas de Rastreamento/métodos , Adulto Jovem , Animais , Pessoa de Meia-Idade , Reservatórios de Doenças/parasitologia
3.
Lancet Microbe ; 5(7): 633-644, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38705163

RESUMO

BACKGROUND: Artemether-lumefantrine is widely used for uncomplicated Plasmodium falciparum malaria; sulfadoxine-pyrimethamine plus amodiaquine is used for seasonal malaria chemoprevention. We aimed to determine the efficacy of artemether-lumefantrine with and without primaquine and sulfadoxine-pyrimethamine plus amodiaquine with and without tafenoquine for reducing gametocyte carriage and transmission to mosquitoes. METHODS: In this phase 2, single-blind, randomised clinical trial conducted in Ouelessebougou, Mali, asymptomatic individuals aged 10-50 years with P falciparum gametocytaemia were recruited from the community and randomly assigned (1:1:1:1) to receive either artemether-lumefantrine, artemether-lumefantrine with a single dose of 0·25 mg/kg primaquine, sulfadoxine-pyrimethamine plus amodiaquine, or sulfadoxine-pyrimethamine plus amodiaquine with a single dose of 1·66 mg/kg tafenoquine. All trial staff other than the pharmacist were masked to group allocation. Participants were not masked to group allocation. Randomisation was done with a computer-generated randomisation list and concealed with sealed, opaque envelopes. The primary outcome was the median within-person percent change in mosquito infection rate in infectious individuals from baseline to day 2 (artemether-lumefantrine groups) or day 7 (sulfadoxine-pyrimethamine plus amodiaquine groups) after treatment, assessed by direct membrane feeding assay. All participants who received any trial drug were included in the safety analysis. This study is registered with ClinicalTrials.gov, NCT05081089. FINDINGS: Between Oct 13 and Dec 16, 2021, 1290 individuals were screened and 80 were enrolled and randomly assigned to one of the four treatment groups (20 per group). The median age of participants was 13 (IQR 11-20); 37 (46%) of 80 participants were female and 43 (54%) were male. In individuals who were infectious before treatment, the median percentage reduction in mosquito infection rate 2 days after treatment was 100·0% (IQR 100·0-100·0; n=19; p=0·0011) with artemether-lumefantrine and 100·0% (100·0-100·0; n=19; p=0·0001) with artemether-lumefantrine with primaquine. Only two individuals who were infectious at baseline infected mosquitoes on day 2 after artemether-lumefantrine and none at day 5. By contrast, the median percentage reduction in mosquito infection rate 7 days after treatment was 63·6% (IQR 0·0-100·0; n=20; p=0·013) with sulfadoxine-pyrimethamine plus amodiaquine and 100% (100·0-100·0; n=19; p<0·0001) with sulfadoxine-pyrimethamine plus amodiaquine with tafenoquine. No grade 3-4 or serious adverse events occurred. INTERPRETATION: These data support the effectiveness of artemether-lumefantrine alone for preventing nearly all mosquito infections. By contrast, there was considerable post-treatment transmission after sulfadoxine-pyrimethamine plus amodiaquine; therefore, the addition of a transmission-blocking drug might be beneficial in maximising its community impact. FUNDING: Bill & Melinda Gates Foundation.


Assuntos
Amodiaquina , Antimaláricos , Combinação Arteméter e Lumefantrina , Combinação de Medicamentos , Fluorenos , Malária Falciparum , Plasmodium falciparum , Primaquina , Pirimetamina , Sulfadoxina , Humanos , Antimaláricos/uso terapêutico , Antimaláricos/administração & dosagem , Pirimetamina/uso terapêutico , Pirimetamina/administração & dosagem , Amodiaquina/uso terapêutico , Amodiaquina/administração & dosagem , Sulfadoxina/uso terapêutico , Sulfadoxina/administração & dosagem , Masculino , Adulto , Feminino , Adolescente , Criança , Malária Falciparum/transmissão , Malária Falciparum/prevenção & controle , Malária Falciparum/tratamento farmacológico , Malária Falciparum/epidemiologia , Método Simples-Cego , Pessoa de Meia-Idade , Primaquina/uso terapêutico , Primaquina/administração & dosagem , Combinação Arteméter e Lumefantrina/uso terapêutico , Combinação Arteméter e Lumefantrina/administração & dosagem , Adulto Jovem , Fluorenos/administração & dosagem , Fluorenos/uso terapêutico , Mali/epidemiologia , Plasmodium falciparum/efeitos dos fármacos , Artemisininas/administração & dosagem , Artemisininas/uso terapêutico , Aminoquinolinas/administração & dosagem , Aminoquinolinas/uso terapêutico , Aminoquinolinas/efeitos adversos , Etanolaminas/administração & dosagem , Etanolaminas/uso terapêutico , Animais , Quimioterapia Combinada
4.
Front Immunol ; 12: 732667, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34659219

RESUMO

Plasmodium falciparum transmission-blocking vaccines (TBVs) targeting the Pfs25 antigen have shown promise in mice but the same efficacy has never been achieved in humans. We have previously published pre-clinical data related to a TBV candidate Pfs25-IMX313 encoded in viral vectors which was very promising and hence progressed to human clinical trials. The results from the clinical trial of this vaccine were very modest. Here we unravel why, contrary to mice, this vaccine has failed to induce robust antibody (Ab) titres in humans to elicit transmission-blocking activity. We examined Pfs25-specific B cell and T follicular helper (Tfh) cell responses in mice and humans after vaccination with Pfs25-IMX313 encoded by replication-deficient chimpanzee adenovirus serotype 63 (ChAd63) and the attenuated orthopoxvirus modified vaccinia virus Ankara (MVA) delivered in the heterologous prime-boost regimen via intramuscular route. We found that after vaccination, the Pfs25-IMX313 was immunologically suboptimal in humans compared to mice in terms of serum Ab production and antigen-specific B, CD4+ and Tfh cell responses. We identified that the key determinant for the poor anti-Pfs25 Ab formation in humans was the lack of CD4+ T cell recognition of Pfs25-IMX313 derived peptide epitopes. This is supported by correlations established between the ratio of proliferated antigen-specific CD4+/Tfh-like T cells, CXCL13 sera levels, and the corresponding numbers of circulating Pfs25-specific memory B cells, that consequently reflected on antigen-specific IgG sera levels. These correlations can inform the design of next-generation Pfs25-based vaccines for robust and durable blocking of malaria transmission.


Assuntos
Anticorpos Antiprotozoários/sangue , Linfócitos T CD4-Positivos/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Imunogenicidade da Vacina , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Proteínas de Protozoários/administração & dosagem , Proteínas Recombinantes/administração & dosagem , Adolescente , Adulto , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/parasitologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/parasitologia , Células Cultivadas , Modelos Animais de Doenças , Epitopos , Feminino , Humanos , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/imunologia , Proteínas Recombinantes/imunologia , Especificidade da Espécie , Vacinação , Adulto Jovem
5.
Front Immunol ; 12: 694759, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34335606

RESUMO

Background: Transmission blocking vaccines targeting the sexual-stages of the malaria parasite could play a major role to achieve elimination and eradication of malaria. The Plasmodium falciparum Pfs25 protein (Pfs25) is the most clinically advanced candidate sexual-stage antigen. IMX313, a complement inhibitor C4b-binding protein that forms heptamers with the antigen fused to it, improve antibody responses. This is the first time that viral vectors have been used to induce antibodies in humans against an antigen that is expressed only in the mosquito vector. Methods: Clinical trial looking at safety and immunogenicity of two recombinant viral vectored vaccines encoding Pfs25-IMX313 in healthy malaria-naive adults. Replication-deficient chimpanzee adenovirus serotype 63 (ChAd63) and the attenuated orthopoxvirus modified vaccinia virus Ankara (MVA), encoding Pfs25-IMX313, were delivered by the intramuscular route in a heterologous prime-boost regimen using an 8-week interval. Safety data and samples for immunogenicity assays were taken at various time-points. Results: The reactogenicity of the vaccines was similar to that seen in previous trials using the same viral vectors encoding other antigens. The vaccines were immunogenic and induced both antibody and T cell responses against Pfs25, but significant transmission reducing activity (TRA) was not observed in most volunteers by standard membrane feeding assay. Conclusion: Both vaccines were well tolerated and demonstrated a favorable safety profile in malaria-naive adults. However, the transmission reducing activity of the antibodies generated were weak, suggesting the need for an alternative vaccine formulation. Trial Registration: Clinicaltrials.gov NCT02532049.


Assuntos
Imunogenicidade da Vacina , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Vacinas Sintéticas/administração & dosagem , Anticorpos Antiprotozoários/sangue , Células Cultivadas , Inglaterra , Voluntários Saudáveis , Humanos , Imunização , Vacinas Antimaláricas/efeitos adversos , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/parasitologia , Fatores de Tempo , Vacinas Sintéticas/efeitos adversos , Vacinas Sintéticas/imunologia
6.
Malar J ; 20(1): 326, 2021 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-34315475

RESUMO

BACKGROUND: Seasonal malaria chemoprevention (SMC) consists of administration of sulfadoxine-pyrimethamine (SP) + amodiaquine (AQ) at monthly intervals to children during the malaria transmission period. Whether the addition of azithromycin (AZ) to SMC could potentiate the benefit of the intervention was tested through a double-blind, randomized, placebo-controlled trial. The effect of SMC and the addition of AZ, on malaria transmission and on the life history traits of Anopheles gambiae mosquitoes have been investigated. METHODS: The study included 438 children randomly selected from among participants in the SMC + AZ trial and 198 children from the same area who did not receive chemoprevention. For each participant in the SMC + AZ trial, blood was collected 14 to 21 days post treatment, examined for the presence of malaria sexual and asexual stages and provided as a blood meal to An. gambiae females using a direct membrane-feeding assay. RESULTS: The SMC treatment, with or without AZ, significantly reduced the prevalence of asexual Plasmodium falciparum (LRT X22 = 69, P < 0.0001) and the gametocyte prevalence (LRT X22 = 54, P < 0.0001). In addition, the proportion of infectious feeds (LRT X22 = 61, P < 0.0001) and the prevalence of oocysts among exposed mosquitoes (LRT X22 = 22.8, P < 0.001) was reduced when mosquitoes were fed on blood from treated children compared to untreated controls. The addition of AZ to SPAQ was associated with an increased proportion of infectious feeds (LRT X21 = 5.2, P = 0.02), suggesting a significant effect of AZ on gametocyte infectivity. There was a slight negative effect of SPAQ and SPAQ + AZ on mosquito survival compared to mosquitoes fed with blood from control children (LRTX22 = 330, P < 0.0001). CONCLUSION: This study demonstrates that SMC may contribute to a reduction in human to mosquito transmission of P. falciparum, and the reduced mosquito longevity observed for females fed on treated blood may increase the benefit of this intervention in control of malaria. The addition of AZ to SPAQ in SMC appeared to enhance the infectivity of gametocytes providing further evidence that this combination is not an appropriate intervention.


Assuntos
Amodiaquina/administração & dosagem , Antimaláricos/administração & dosagem , Azitromicina/administração & dosagem , Culicidae/fisiologia , Aptidão Genética , Malária Falciparum , Plasmodium falciparum/fisiologia , Pirimetamina/administração & dosagem , Sulfadoxina/administração & dosagem , Animais , Quimioprevenção , Pré-Escolar , Combinação de Medicamentos , Humanos , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Estações do Ano
7.
Sci Rep ; 11(1): 6905, 2021 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-33767384

RESUMO

In areas where malaria remains entrenched, novel transmission-reducing interventions are essential for malaria elimination. We report the impact screening-and-treatment of asymptomatic Malawian schoolchildren (n = 364 in the rainy season and 341 in the dry season) had on gametocyte-the parasite stage responsible for human-to-mosquito transmission-carriage. We used concomitant household-based surveys to predict the potential reduction in transmission in the surrounding community. Among 253 students with P. falciparum infections at screening, 179 (71%) had infections containing gametocytes detected by Pfs25 qRT-PCR. 84% of gametocyte-containing infections were detected by malaria rapid diagnostic test. While the gametocyte prevalence remained constant in untreated children, treatment with artemether-lumefantrine reduced the gametocyte prevalence (p < 0.0001) from 51.8 to 9.7% and geometric mean gametocyte density (p = 0.008) from 0.52 to 0.05 gametocytes/microliter. In community surveys, 46% of all gametocyte-containing infections were in school-age children, who comprised only 35% of the population. Based on these estimates six weeks after the intervention, the gametocyte burden in the community could be reduced by 25-55% depending on the season and the measure used to characterize gametocyte carriage. Thus, school-based interventions to treat asymptomatic infections may be a high-yield approach to not only improve the health of schoolchildren, but also decrease malaria transmission.


Assuntos
Malária Falciparum/diagnóstico , Malária Falciparum/prevenção & controle , Programas de Rastreamento/estatística & dados numéricos , Plasmodium falciparum/isolamento & purificação , Antimaláricos/uso terapêutico , Combinação Arteméter e Lumefantrina/uso terapêutico , Criança , Estudos de Coortes , Feminino , Humanos , Malária Falciparum/tratamento farmacológico , Malária Falciparum/transmissão , Malaui , Masculino , Serviços de Saúde Escolar/estatística & dados numéricos
8.
Nat Commun ; 12(1): 1750, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33741942

RESUMO

Malaria elimination requires tools that interrupt parasite transmission. Here, we characterize B cell receptor responses among Malian adults vaccinated against the first domain of the cysteine-rich 230 kDa gamete surface protein Pfs230, a key protein in sexual stage development of P. falciparum parasites. Among nine Pfs230 human monoclonal antibodies (mAbs) that we generated, one potently blocks transmission to mosquitoes in a complement-dependent manner and reacts to the gamete surface; the other eight show only low or no blocking activity. The structure of the transmission-blocking mAb in complex with vaccine antigen reveals a large discontinuous conformational epitope, specific to domain 1 of Pfs230 and comprising six structural elements in the protein. The epitope is conserved, suggesting the transmission-blocking mAb is broadly functional. This study provides a rational basis to improve malaria vaccines and develop therapeutic antibodies for malaria elimination.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Antiprotozoários/farmacologia , Epitopos/imunologia , Células Germinativas/imunologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/efeitos dos fármacos , Adulto , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/química , Antígenos de Protozoários/imunologia , Sítios de Ligação , Células Cultivadas , Epitopos/química , Interações Hospedeiro-Parasita/efeitos dos fármacos , Interações Hospedeiro-Parasita/imunologia , Humanos , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Mosquitos Vetores/parasitologia , Plasmodium falciparum/imunologia , Plasmodium falciparum/fisiologia , Conformação Proteica , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia
9.
Am J Trop Med Hyg ; 104(1): 240-242, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33236701

RESUMO

Postexposure prophylaxis using artemisinin-based combination therapy (ACT) was prescribed to a malaria-naive nurse who experienced an injury with a hollow needle previously used on a patient admitted for severe imported Plasmodium falciparum malaria (blood parasitemia 10.8%). Artemether-lumefantrine, four tablets twice daily for 3 days, was started 12 hours after exposure, and no side effects were reported. During the six following months, she only developed one episode of fever that was associated with pyelonephritis. Biological follow-up, based on blood smears, molecular biology, and serology, did not evidence P. falciparum malaria. This case suggests that ACT can prevent occupational P. falciparum malaria following needle-stick injury. We found evidence of only one other unpublished similar case where a Turkish resident doctor did not develop malaria after postexposure prophylaxis using ACT. Such a prophylaxis could be prescribed not only in case of occupational exposure to Plasmodium spp. in nonvector-borne laboratory-acquired infections but also following blood exposure in healthcare setting.


Assuntos
Combinação Arteméter e Lumefantrina/administração & dosagem , Combinação Arteméter e Lumefantrina/uso terapêutico , Malária Falciparum/prevenção & controle , Malária Falciparum/transmissão , Profilaxia Pós-Exposição , Adulto , Antimaláricos/administração & dosagem , Antimaláricos/uso terapêutico , Feminino , Humanos , Ferimentos Penetrantes Produzidos por Agulha/complicações
10.
Am J Trop Med Hyg ; 104(2): 671-679, 2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-33236715

RESUMO

Malaria elimination strategies are designed to more effectively identify and treat infected individuals to interrupt transmission. One strategy, reactive screen-and-treat, starts with passive detection of symptomatic cases at health facilities. Individuals residing within the index case and neighboring households are screened with a malaria rapid diagnostic test (RDT) and treated if positive. However, it is unclear to what extent this strategy is effective in reducing transmission. Reactive screen-and-treat was implemented in Choma district, Southern Province, Zambia, in 2013, in which residents of the index case and neighboring households within 140 m were screened with an RDT. From March 2016 to July 2018, the screening radius was extended to 250-m, and additional follow-up visits at 30 and 90 days were added to evaluate the strategy. Plasmodium falciparum parasite prevalence was measured using an RDT and by quantitative PCR (qPCR). A 24-single nucleotide polymorphism molecular bar-code assay was used to genotype parasites. Eighty-four index case households with 676 residents were enrolled between March 2016 and March 2018. Within each season, parasite prevalence declined significantly in index households at the 30-day visit and remained low at the 90-day visit. However, parasite prevalence was not reduced to zero. Infections identified by qPCR persisted between study visits and were not identified by RDT. Parasites identified within the same household were most genetically related; however, overall parasite relatedness was low and similar across time and space. Thus, despite implementation of a reactive screen-and-treat program, parasitemia was not eliminated, and persisted in targeted households for at least 3 months.


Assuntos
Antimaláricos/uso terapêutico , Malária Falciparum/diagnóstico , Malária Falciparum/transmissão , Programas de Rastreamento/normas , Plasmodium falciparum/genética , Adolescente , Adulto , Animais , Anopheles/parasitologia , Criança , Pré-Escolar , Estudos Transversais , Características da Família , Feminino , Genótipo , Humanos , Malária Falciparum/tratamento farmacológico , Malária Falciparum/epidemiologia , Masculino , Programas de Rastreamento/estatística & dados numéricos , Parasitemia , Prevalência , Adulto Jovem , Zâmbia/epidemiologia
11.
Rev. chil. pediatr ; 91(5): 749-753, oct. 2020. graf
Artigo em Espanhol | LILACS | ID: biblio-1144274

RESUMO

INTRODUCCIÓN: La malaria congénita (MC) es la infección por Plasmodium spp adquirida in útero o durante el parto y sus manifestaciones clínicas son inespecíficas. Puede causar enfermedad grave en la embaraza da y en el recién nacido. OBJETIVO: describir dos casos de MC causados por Plasmodium falciparum, diagnóstico diferencial de sepsis en recién nacidos de gestantes que hayan visitado o residan en áreas endémicas para malaria. CASOS CLÍNICOS: Neonatos de sexo femenino, nacidos en área no endémica para malaria, diagnosticados con sepsis neonatal y tratados con antibióticos sin respuesta clínica. Después de la primera semana de vida la gota gruesa identificó trofozoítos de Plasmodium falciparum y los neonatos recibieron tratamiento con quinina intravenosa con mejoría. Las madres de las recién nacidas tuvieron malaria en el embarazo, una de ellas recibió tratamiento y estaba asintomática y otra tenía malaria complicada al momento del parto. CONCLUSIONES: La MC puede causar enfermedad neonatal grave con manifestaciones clínicas inespecíficas y similares a la sepsis, el tratamiento oportuno disminuye el riesgo de malaria complicada. Es un diagnóstico diferencial en recién nacidos de mujeres con malaria durante el embarazo o gestantes que visiten o residan en áreas endémicas.


INTRODUCTION: Congenital malaria (CM) is a Plasmodium spp infection acquired in utero or during delivery with nonspecific clinical manifestations. Plasmodium falciparum can cause severe illness in pregnant wo men and newborns. OBJECTIVE: to describe two cases of CM caused by Plasmodium falciparum, di fferential diagnosis of sepsis in newborns of pregnant women who live in or have visited endemic malaria zones. CLINICAL CASES: Female neonates born in a non-endemic malaria area, diagnosed with neonatal sepsis and treated with antibiotics without clinical response. After the first week of life, the peripheral blood smear identified trophozoites of Plasmodium falciparum thus the newborns were treated with intravenous quinine, improving their condition. The mothers of the two newborns who had malaria in pregnancy, one of them received treatment and she was asymptomatic, and the other one had severe malaria at the time of delivery. CONCLUSIONS: CM can cause severe neonatal disease with non-specific, sepsis-like clinical manifestations in which early treatment decreases the risk of complicated malaria. It is a differential diagnosis in newborns of women with a history of malaria during pregnancy or pregnant women visiting or living in endemic malaria areas.


Assuntos
Humanos , Feminino , Gravidez , Recém-Nascido , Adolescente , Adulto Jovem , Malária Falciparum/congênito , Malária Falciparum/diagnóstico , Complicações Parasitárias na Gravidez/diagnóstico , Complicações Parasitárias na Gravidez/tratamento farmacológico , Sepse Neonatal/diagnóstico , Malária Falciparum/transmissão , Transmissão Vertical de Doenças Infecciosas , Diagnóstico Diferencial , Sepse Neonatal/parasitologia , Antimaláricos/uso terapêutico
12.
Commun Biol ; 3(1): 395, 2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32709983

RESUMO

Proteins Pfs230 and Pfs48/45 are Plasmodium falciparum transmission-blocking (TB) vaccine candidates that form a membrane-bound protein complex on gametes. The biological role of Pfs230 or the Pfs230-Pfs48/45 complex remains poorly understood. Here, we present the crystal structure of recombinant Pfs230 domain 1 (Pfs230D1M), a 6-cysteine domain, in complex with the Fab fragment of a TB monoclonal antibody (mAb) 4F12. We observed the arrangement of Pfs230 on the surface of macrogametes differed from that on microgametes, and that Pfs230, with no known membrane anchor, may exist on the membrane surface in the absence of Pfs48/45. 4F12 appears to sterically interfere with Pfs230 function. Combining mAbs against different epitopes of Pfs230D1 or of Pfs230D1 and Pfs48/45, significantly increased TB activity. These studies elucidate a mechanism of action of the Pfs230D1 vaccine, model the functional activity induced by a polyclonal antibody response and support the development of TB vaccines targeting Pfs230D1 and Pfs230D1-Pfs48/45.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/farmacologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/patogenicidade , Animais , Antígenos de Protozoários/genética , Humanos , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Glicoproteínas de Membrana/antagonistas & inibidores , Glicoproteínas de Membrana/imunologia , Plasmodium falciparum/imunologia , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/imunologia
13.
Front Immunol ; 11: 505, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32318061

RESUMO

Understanding the functional role of proteins expressed by Plasmodium falciparum is an important step toward unlocking potential targets for the development of therapeutic or diagnostic interventions. The armadillo (ARM) repeat protein superfamily is associated with varied functions across the eukaryotes. Therefore, it is important to understand the role of members of this protein family in Plasmodium biology. The Plasmodium falciparum armadillo repeats only (PfARO; Pf3D7_0414900) and P. falciparum merozoite organizing proteins (PfMOP; Pf3D7_0917000) are armadillo-repeat containing proteins previously characterized in P. falciparum. Here, we describe the characterization of another ARM repeat-containing protein in P. falciparum, which we have named the P. falciparum Merozoites-Associated Armadillo repeats protein (PfMAAP). Antibodies raised to three different synthetic peptides of PfMAAP show apical staining of free merozoites and those within the mature infected schizont. We also demonstrate that the antibodies raised to the PfMAAP peptides inhibited invasion of erythrocytes by merozoites from different parasite isolates. In addition, naturally acquired human antibodies to the N- and C- termini of PfMAAP are associated with a reduced risk of malaria in a prospective cohort analysis.


Assuntos
Proteínas do Domínio Armadillo/metabolismo , Eritrócitos/imunologia , Malária Falciparum/metabolismo , Peptídeos/metabolismo , Plasmodium falciparum/imunologia , Proteínas de Protozoários/metabolismo , Animais , Anticorpos Antiprotozoários/sangue , Proteínas do Domínio Armadillo/genética , Estudos de Coortes , Eritrócitos/parasitologia , Humanos , Imunidade Humoral , Malária Falciparum/transmissão , Merozoítos , Peptídeos/genética , Estudos Prospectivos , Transporte Proteico , Proteínas de Protozoários/genética , Esquizontes
14.
Am J Trop Med Hyg ; 103(1): 369-377, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32342846

RESUMO

Progress with malaria control in western Kenya has stagnated since 2007. Additional interventions to reduce the high burden of malaria in this region are urgently needed. We conducted a two-arm, community-based, cluster-randomized, controlled trial of active case detection and treatment of malaria infections in all residents mass testing and treatment (MTaT) of 10 village clusters (intervention clusters) for two consecutive years to measure differences in the incidence of clinical malaria disease and malaria infections compared with 20 control clusters where MTaT was not implemented. All residents of intervention clusters, irrespective of history of fever or other malaria-related symptoms, were tested three times per year before the peak malaria season using malaria rapid diagnostic tests. All positive cases were treated with dihydroartemisinin-piperaquine. The incidence of clinical malaria was measured through passive surveillance, whereas the cumulative incidence of malaria infection was measured using active surveillance in a cohort comprising randomly selected residents. The incidence of clinical malaria was 0.19 cases/person-year (p-y, 95% CI: 0.13-0.28) in the intervention arm and 0.24 cases/p-y (95% CI: 0.15-0.39) in the control arm (incidence rate ratio [IRR] 0.79, 95% CI: 0.61-1.02). The cumulative incidence of malaria infections was similar between the intervention (2.08 infections/p-y, 95% CI: 1.93-2.26) and control arms (2.19 infections/p-y, 95% CI: 2.02-2.37) with a crude IRR of 0.95 (95% CI: 0.87-1.04). Six rounds of MTaT over 2 years did not have a significant impact on the incidence of clinical malaria or the cumulative incidence of malaria infection in this area of high malaria transmission.


Assuntos
Antimaláricos/uso terapêutico , Malária Falciparum/diagnóstico , Adolescente , Artemisininas/uso terapêutico , Criança , Pré-Escolar , Feminino , Humanos , Incidência , Quênia/epidemiologia , Malária Falciparum/tratamento farmacológico , Malária Falciparum/epidemiologia , Malária Falciparum/transmissão , Masculino , Programas de Rastreamento/métodos , Quinolinas/uso terapêutico
15.
Transfusion ; 60(4): 799-805, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32129497

RESUMO

BACKGROUND: Risk of transfusion-transmitted (TT) malaria is mainly associated with whole blood (WB) or red blood cell (RBC) transfusion. Risk mitigation relies mostly on donor deferral while a limited number of countries perform blood testing, both negatively impacting blood availability. This study investigated the efficacy of the pathogen reduction system using amustaline and glutathione (GSH) to inactivate Plasmodium falciparum in WB. STUDY DESIGN AND METHODS: WB units were spiked with ring stage P. falciparum infected RBCs. Parasite loads were measured in samples at time of infection, after 24 hours at room temperature (RT), and after a 24-hour incubation at RT post-treatment with 0.2 mM amustaline and 2 mM GSH. Serial 10-fold dilutions of the samples were inoculated to RBC cultures and maintained up to 4 weeks. Parasitemia was quantified by cytometry. RESULTS: The P. falciparum viability assay has a limit of detection of a single live parasite per sample. Input parasite titer was >5.7 log10 TCID50 per mL. A 24-hour incubation at RT paused parasite development in controls, but they retained viability and infectivity when tested in culture. In contrast, no infectious parasites were detected in the amustaline/GSH-treated sample after 4 weeks of culture. CONCLUSION: A robust level of P. falciparum inactivation was achieved in WB using amustaline/GSH treatment. Parasite log reduction was >5.7 log10 TCID50 per mL. Development of such a pathogen reduction system may provide an opportunity to reduce the risk of TT malaria and improve blood availability.


Assuntos
Acridinas/farmacologia , Glutationa/farmacologia , Malária Falciparum/prevenção & controle , Viabilidade Microbiana/efeitos dos fármacos , Compostos de Mostarda Nitrogenada/farmacologia , Segurança do Sangue/métodos , Eritrócitos/microbiologia , Eritrócitos/parasitologia , Humanos , Malária Falciparum/sangue , Malária Falciparum/transmissão , Carga Parasitária , Parasitemia/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/crescimento & desenvolvimento
16.
Cell Host Microbe ; 27(1): 93-103.e4, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31901523

RESUMO

In high-transmission regions, we expect parasite lineages within complex malaria infections to be unrelated due to parasite inoculations from different mosquitoes. This project was designed to test this prediction. We generated 485 single-cell genome sequences from fifteen P. falciparum malaria patients from Chikhwawa, Malawi-an area of intense transmission. Patients harbored up to seventeen unique parasite lineages. Surprisingly, parasite lineages within infections tend to be closely related, suggesting that superinfection by repeated mosquito bites is rarer than co-transmission of parasites from a single mosquito. Both closely and distantly related parasites comprise an infection, suggesting sequential transmission of complex infections between multiple hosts. We identified tetrads and reconstructed parental haplotypes, which revealed the inbred ancestry of infections and non-Mendelian inheritance. Our analysis suggests strong barriers to secondary infection and outbreeding amongst malaria parasites from a high transmission setting, providing unexpected insights into the biology and transmission of malaria.


Assuntos
Malária Falciparum/transmissão , Plasmodium falciparum/genética , Animais , Biodiversidade , Evolução Clonal , Coinfecção/parasitologia , Culicidae/parasitologia , Variação Genética , Genômica , Haplótipos , Humanos , Plasmodium falciparum/isolamento & purificação
17.
Sci Rep ; 10(1): 395, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31942034

RESUMO

The Plasmodium falciparum gametocyte surface protein, Pfs48/45, is a potential target for malaria transmission-blocking vaccines. However, due to its size and complexity, expression of the full-length protein has been difficult, leading to focus on the C-terminal six cysteine domain (6C) with the use of fusion proteins to facilitate expression and folding. In this study, we utilized the baculovirus system to evaluate the expression of three Pfs48/45 proteins including the full-length protein, the 6C domain fragment and the 6C domain mutant to prevent glycosylation. Expression of the recombinant Pfs48/45 proteins was conducted in super Sf9 cells combined with the use of tunicamycin to prevent N-glycosylation. The proteins were then evaluated as immunogens in mice to demonstrate the induction of functionally active polyclonal antibody responses as measured in the standard membrane feeding assay (SMFA). Only the 6C protein was found to exhibit significant transmission-reducing activity. Further characterization of the biologically active 6C protein demonstrated it was homogeneous in terms of size, charge, conformation, absence of glycosylation, and containing proper disulfide bond pairings. This study presents an alternative expression system, without the need of a fusion protein partner, for the Pfs48/45 6C protein fragment including further evaluation as a potential transmission-blocking vaccine candidate.


Assuntos
Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/biossíntese , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Proteínas Recombinantes/biossíntese , Animais , Baculoviridae/genética , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Proteínas Recombinantes/imunologia
18.
Front Immunol ; 11: 606266, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33505395

RESUMO

The cysteine-rich Pfs48/45 protein, a Plasmodium falciparum sexual stage surface protein, has been advancing as a candidate antigen for a transmission-blocking vaccine (TBV) for malaria. However, Pfs48/45 contains multiple disulfide bonds, that are critical for proper folding and induction of transmission-blocking (TB) antibodies. We have previously shown that R0.6C, a fusion of the 6C domain of Pfs48/45 and a fragment of PfGLURP (R0), expressed in Lactococcus lactis, was properly folded and induced transmission-blocking antibodies. Here we describe the process development and technology transfer of a scalable and reproducible process suitable for R0.6C manufacturing under current Good Manufacturing Practices (cGMP). This process resulted in a final purified yield of 25 mg/L, sufficient for clinical evaluation. A panel of analytical assays for release and stability assessment of R0.6C were developed including HPLC, SDS-PAGE, and immunoblotting with the conformation-dependent TB mAb45.1. Intact mass analysis of R0.6C confirmed the identity of the product including the three disulfide bonds and the absence of post-translational modifications. Multi-Angle Light Scattering (MALS) coupled to size exclusion chromatography (SEC-MALS), further confirmed that R0.6C was monomeric (~70 kDa) in solution. Lastly, preclinical studies demonstrated that the R0.6C Drug Product (adsorbed to Alhydrogel®) elicited functional antibodies in small rodents and that adding Matrix-M™ adjuvant further increased the functional response. Here, building upon our past work, we filled the gap between laboratory and manufacturing to ready R0.6C for production under cGMP and eventual clinical evaluation as a malaria TB vaccine.


Assuntos
Biotecnologia , Microbiologia Industrial , Lactobacillus/metabolismo , Vacinas Antimaláricas/biossíntese , Malária Falciparum/prevenção & controle , Glicoproteínas de Membrana/biossíntese , Proteínas de Protozoários/biossíntese , Adjuvantes Imunológicos/farmacologia , Animais , Anticorpos Antiprotozoários/imunologia , Composição de Medicamentos , Imunização , Imunogenicidade da Vacina , Lactobacillus/genética , Vacinas Antimaláricas/química , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/farmacologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Malária Falciparum/transmissão , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/farmacologia , Camundongos , Nanopartículas , Conformação Proteica , Dobramento de Proteína , Estabilidade Proteica , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas de Protozoários/farmacologia , Saponinas/farmacologia , Relação Estrutura-Atividade , Vacinas Sintéticas/biossíntese , Vacinas Sintéticas/farmacologia
19.
Vaccine ; 37(13): 1799-1806, 2019 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-30824357

RESUMO

A transmission-blocking vaccine (TBV) against Plasmodium falciparum is likely to be a valuable tool in a malaria eradication program. Pfs230 is one of the major TBV candidates, and multiple Pfs230-based vaccines induced antibodies, which prevented oocyst formation in mosquitoes as determined by a standard membrane-feeding assay (SMFA). Pfs230 is a >300 kDa protein consisting of 14 cysteine motif (CM) domains, and the size and cysteine-rich nature of the molecule have hampered its production as an intact protein. Except for one early study with maltose-binding protein fusion Pfs230 constructs expressed in Esherichia coli, all other studies have focused on only the first four CM domains in the Pfs230 molecule. To identify all possible TBV candidate domains, we systematically produced either single-CM-domain (a total of 14), 2-CM-domain (7), or 4-CM-domain (6) recombinant protein fragments using a eukaryotic wheat germ cell-free expression system (WGCFS). In addition, two more constructs which covered previously published regions, and an N-terminal prodomain construct spanning the natural cleavage site of Pfs230 were produced. Antisera against each fragment were generated in mice and we evaluated the reactivity to native Pfs230 protein by Western blots and immunofluorescence assay (IFA), and functionality by SMFA. All 30 WGCFS-produced Pfs230 constructs were immunogenic in mice. Approximately half of the mouse antibodies specifically recognized native Pfs230 by Western blots with variable band intensities. Among them, seven antibodies showed higher reactivities against native Pfs230 determined by IFA. Interestingly, antibodies against all protein fragments containing CM domain 1 displayed strong inhibitions in SMFA, while antibodies generated using constructs without CM domain 1 showed no inhibition. The results strongly support the concept that future Pfs230-based vaccine development should focus on the Pfs230 CM domain 1.


Assuntos
Anticorpos Bloqueadores/imunologia , Anticorpos Antiprotozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Animais , Antígenos de Protozoários/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Humanos , Imunogenicidade da Vacina , Imunoglobulina G/imunologia , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/transmissão , Camundongos , Proteínas de Protozoários/imunologia , Proteínas Recombinantes/imunologia
20.
Rev. Soc. Bras. Med. Trop ; Rev. Soc. Bras. Med. Trop;52: e20180537, 2019. tab, graf
Artigo em Inglês | LILACS | ID: biblio-990433

RESUMO

Abstract INTRODUCTION: Elimination of malaria in areas of interrupted transmission warrants careful case assessment to avoid the reintroduction of this disease. Occasional malaria cases are reported among visitors of the Atlantic Forest area of Brazil, while data on residents of this area are scarce. METHODS: A sectional study was carried out to examine 324 individuals living in a municipality where autochthonous cases were detected. RESULTS: Asymptomatic Plasmodium infections were detected in 2.8% of the individuals by polymerase chain reaction (PCR), with one case of P. falciparum (0.3%), two cases of P. vivax (0.6%), and six cases of P. malariae (1.9%). The thick blood smears were negative in all individuals. Serological tests performed in 314 subjects were reactive in 11.1%, with 3.5% for P. falciparum and 7.7% for P. vivax. A subsample of 42 reactive individuals for any Plasmodium species showed P. malariae in 30.9% of specimens. Individuals who entered the Atlantic Forest region were 2.7 times more likely to exhibit reactive serology for P. vivax compared with individuals who did not enter this region (p<0.05). Children <15 years had a higher chance of reactive serology for P. falciparum and P. vivax than individuals ≥15 years of age (p<0.05). Individuals living in the Paraiso district had a higher chance of reactive serology for P. vivax compared to other districts (p<0.05). No associations were found between sex, past exposure to malaria, or serological response to antibodies of any Plasmodium species. CONCLUSIONS: The implications of these results for the elimination of malaria were discussed.


Assuntos
Humanos , Masculino , Feminino , Adulto , Idoso , Idoso de 80 Anos ou mais , Malária Vivax/diagnóstico , Malária Vivax/transmissão , Malária Falciparum/diagnóstico , Malária Falciparum/transmissão , Brasil/epidemiologia , Ensaio de Imunoadsorção Enzimática , Reação em Cadeia da Polimerase , Estudos Transversais , DNA de Protozoário/análise , Malária Vivax/epidemiologia , Malária Falciparum/epidemiologia , Infecções Assintomáticas/epidemiologia , Antígenos de Protozoários/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA