Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Int J Infect Dis ; 134: 114-122, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37269941

RESUMO

OBJECTIVES: To assess the pharmacokinetics, safety, and tolerability of two high-dose, short-course primaquine (PQ) regimens compared with standard care in children with Plasmodium vivax infections. METHODS: We performed an open-label pediatric dose-escalation study in Madang, Papua New Guinea (Clinicaltrials.gov NCT02364583). Children aged 5-10 years with confirmed blood-stage vivax malaria and normal glucose-6-phosphate dehydrogenase activity were allocated to one of three PQ treatment regimens in a stepwise design (group A: 0.5 mg/kg once daily for 14 days, group B: 1 mg/kg once daily for 7 days, and group C: 1 mg/kg twice daily for 3.5-days). The study assessments were completed at each treatment time point and fortnightly for 2 months after PQ administration. RESULTS: Between August 2013 and May 2018, 707 children were screened and 73 met the eligibility criteria (15, 40, and 16 allocated to groups A, B, and C, respectively). All children completed the study procedures. The three regimens were safe and generally well tolerated. The pharmacokinetic analysis indicated that an additional weight adjustment of the conventionally recommended milligram per kilogram PQ doses is not necessary to ensure the therapeutic plasma concentrations in pediatric patients. CONCLUSIONS: A novel, ultra-short 3.5-day PQ regimen has potential benefits for improving the treatment outcomes in children with vivax malaria that warrants further investigation in a large-scale clinical trial.


Assuntos
Antimaláricos , Malária Vivax , Humanos , Criança , Primaquina/efeitos adversos , Malária Vivax/tratamento farmacológico , Antimaláricos/efeitos adversos , Resultado do Tratamento , Fígado , Plasmodium vivax
2.
Sci Rep ; 9(1): 3333, 2019 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-30833657

RESUMO

Longitudinal tracking of individual Plasmodium falciparum strains in multi-clonal infections is essential for investigating infection dynamics of malaria. The traditional genotyping techniques did not permit tracking changes in individual clone density during persistent natural infections. Amplicon deep sequencing (Amp-Seq) offers a tool to address this knowledge gap. The sensitivity of Amp-Seq for relative quantification of clones was investigated using three molecular markers, ama1-D2, ama1-D3, and cpmp. Amp-Seq and length-polymorphism based genotyping were compared for their performance in following minority clones in longitudinal samples from Papua New Guinea. Amp-Seq markers were superior to length-polymorphic marker msp2 in detecting minority clones (sensitivity Amp-Seq: 95%, msp2: 85%). Multiplicity of infection (MOI) by Amp-Seq was 2.32 versus 1.73 for msp2. The higher sensitivity had no effect on estimates of force of infection because missed minority clones were detected in preceding or succeeding bleeds. Individual clone densities were tracked longitudinally by Amp-Seq despite MOI > 1, thus providing an additional parameter for investigating malaria infection dynamics. Amp-Seq based genotyping of longitudinal samples improves detection of minority clones and estimates of MOI. Amp-Seq permits tracking of clone density over time to study clone competition or the dynamics of specific, i.e. resistance-associated genotypes.


Assuntos
Malária Falciparum/parasitologia , Plasmodium falciparum/genética , Animais , Pré-Escolar , Estudos de Coortes , Genótipo , Humanos , Lactente , Estudos Longitudinais
3.
BMC Genomics ; 18(1): 864, 2017 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-29132317

RESUMO

BACKGROUND: Amplicon deep sequencing permits sensitive detection of minority clones and improves discriminatory power for genotyping multi-clone Plasmodium falciparum infections. New amplicon sequencing and data analysis protocols are needed for genotyping in epidemiological studies and drug efficacy trials of P. falciparum. METHODS: Targeted sequencing of molecular marker csp and novel marker cpmp was conducted in duplicate on mixtures of parasite culture strains and 37 field samples. A protocol allowing to multiplex up to 384 samples in a single sequencing run was applied. Software "HaplotypR" was developed for data analysis. RESULTS: Cpmp was highly diverse (He = 0.96) in contrast to csp (He = 0.57). Minority clones were robustly detected if their frequency was >1%. False haplotype calls owing to sequencing errors were observed below that threshold. CONCLUSIONS: To reliably detect haplotypes at very low frequencies, experiments are best performed in duplicate and should aim for coverage of >10'000 reads/amplicon. When compared to length polymorphic marker msp2, highly multiplexed amplicon sequencing displayed greater sensitivity in detecting minority clones.


Assuntos
Marcadores Genéticos/genética , Técnicas de Genotipagem/métodos , Sequenciamento de Nucleotídeos em Larga Escala , Malária Falciparum/parasitologia , Plasmodium falciparum/genética , Plasmodium falciparum/fisiologia , Polimorfismo de Nucleotídeo Único
4.
Elife ; 62017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28862132

RESUMO

The molecular force of blood-stage infection (molFOB) is a quantitative surrogate metric for malaria transmission at population level and for exposure at individual level. Relationships between molFOB, parasite prevalence and clinical incidence were assessed in a treatment-to-reinfection cohort, where P.vivax (Pv) hypnozoites were eliminated in half the children by primaquine (PQ). Discounting relapses, children acquired equal numbers of new P. falciparum (Pf) and Pv blood-stage infections/year (Pf-molFOB = 0-18, Pv-molFOB = 0-23) resulting in comparable spatial and temporal patterns in incidence and prevalence of infections. Including relapses, Pv-molFOB increased >3 fold (relative to PQ-treated children) showing greater heterogeneity at individual (Pv-molFOB = 0-36) and village levels. Pf- and Pv-molFOB were strongly associated with clinical episode risk. Yearly Pf clinical incidence rate (IR = 0.28) was higher than for Pv (IR = 0.12) despite lower Pf-molFOB. These relationships between molFOB, clinical incidence and parasite prevalence reveal a comparable decline in Pf and Pv transmission that is normally hidden by the high burden of Pv relapses. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov NCT02143934.


Assuntos
Exposição Ambiental , Malária Falciparum/epidemiologia , Malária Vivax/epidemiologia , Criança , Feminino , Humanos , Incidência , Masculino , Papua Nova Guiné/epidemiologia , Prevalência , Recidiva , Análise Espaço-Temporal
5.
PLoS Negl Trop Dis ; 11(9): e0005888, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28892517

RESUMO

Plasmodium vivax remains an important cause of malaria in South America and the Asia-Pacific. Naturally acquired antibody responses against multiple P. vivax proteins have been described in numerous countries, however, direct comparison of these responses has been difficult with different methodologies employed. We measured antibody responses against 307 P. vivax proteins at the time of P. vivax infection, and at 2-3 later time-points in three countries. We observed that seropositivity rates at the time of infection were highest in Thailand, followed by Brazil then PNG, reflecting the level of antigenic input. The majority of sero-reactive antigens in all sites induced short-lived antibody responses with estimated half-lives of less than 6 months, although there was a trend towards longer-lived responses in PNG children. Despite these differences, IgG seropositivity rates, magnitude and longevity were highly and significantly rank-correlated between the different regions, suggesting such features are reflective of the individual protein.


Assuntos
Formação de Anticorpos , Antígenos de Protozoários/imunologia , Malária Vivax/imunologia , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Adolescente , Adulto , Idoso , Anticorpos Antiprotozoários/sangue , Brasil , Criança , Pré-Escolar , Feminino , Meia-Vida , Humanos , Imunoglobulina G/sangue , Malária Vivax/parasitologia , Masculino , Pessoa de Meia-Idade , Papua Nova Guiné , Tailândia , Adulto Jovem
6.
PLoS Negl Trop Dis ; 11(7): e0005753, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28732068

RESUMO

BACKGROUND: Primaquine (PQ) is the only currently licensed antimalarial that prevents Plasmodium vivax (Pv) relapses. It also clears mature P. falciparum (Pf) gametocytes, thereby reducing post-treatment transmission. Randomized PQ treatment in a treatment-to-reinfection cohort in Papua New Guinean children permitted the study of Pv and Pf gametocyte carriage after radical cure and to investigate the contribution of Pv relapses. METHODS: Children received radical cure with Chloroquine, Artemether-Lumefantrine plus either PQ or placebo. Blood samples were subsequently collected in 2-to 4-weekly intervals over 8 months. Gametocytes were detected by quantitative reverse transcription-PCR targeting pvs25 and pfs25. RESULTS: PQ treatment reduced the incidence of Pv gametocytes by 73%, which was comparable to the effect of PQ on incidence of blood-stage infections. 92% of Pv and 79% of Pf gametocyte-positive infections were asymptomatic. Pv and to a lesser extent Pf gametocyte positivity and density were associated with high blood-stage parasite densities. Multivariate analysis revealed that the odds of gametocytes were significantly reduced in mixed-species infections compared to single-species infections for both species (ORPv = 0.39 [95% CI 0.25-0.62], ORPf = 0.33 [95% CI 0.18-0.60], p<0.001). No difference between the PQ and placebo treatment arms was observed in density of Pv gametocytes or in the proportion of Pv infections that carried gametocytes. First infections after blood-stage and placebo treatment, likely caused by a relapsing hypnozoite, were equally likely to carry gametocytes than first infections after PQ treatment, likely caused by an infective mosquito bite. CONCLUSION: Pv relapses and new infections are associated with similar levels of gametocytaemia. Relapses thus contribute considerably to the Pv reservoir highlighting the importance of effective anti-hypnozoite treatment for efficient control of Pv. TRIAL REGISTRATION: ClinicalTrials.gov NCT02143934.


Assuntos
Antimaláricos/uso terapêutico , Malária Falciparum/tratamento farmacológico , Malária Vivax/tratamento farmacológico , Plasmodium falciparum/efeitos dos fármacos , Plasmodium vivax/efeitos dos fármacos , Primaquina/uso terapêutico , Combinação Arteméter e Lumefantrina , Artemisininas/uso terapêutico , Sangue/parasitologia , Criança , Pré-Escolar , Cloroquina/uso terapêutico , Método Duplo-Cego , Combinação de Medicamentos , Etanolaminas/uso terapêutico , Feminino , Fluorenos/uso terapêutico , Humanos , Fígado/efeitos dos fármacos , Fígado/parasitologia , Masculino , Análise Multivariada , Papua Nova Guiné , Recidiva , Análise de Regressão , Fatores de Risco
7.
PLoS Negl Trop Dis ; 11(6): e0005606, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28604825

RESUMO

BACKGROUND: Despite that over 90 million pregnancies are at risk of Plasmodium vivax infection annually, little is known about the epidemiology and impact of the infection in pregnancy. METHODOLOGY AND PRINCIPAL FINDINGS: We undertook a health facility-based prospective observational study in pregnant women from Guatemala (GT), Colombia (CO), Brazil (BR), India (IN) and Papua New Guinea PNG). Malaria and anemia were determined during pregnancy and fetal outcomes assessed at delivery. A total of 9388 women were enrolled at antennal care (ANC), of whom 53% (4957) were followed until delivery. Prevalence of P. vivax monoinfection in maternal blood at delivery was 0.4% (20/4461) by microscopy [GT 0.1%, CO 0.5%, BR 0.1%, IN 0.2%, PNG 1.2%] and 7% (104/1488) by PCR. P. falciparum monoinfection was found in 0.5% (22/4463) of women by microscopy [GT 0%, CO 0.5%, BR 0%, IN 0%, PNG 2%]. P. vivax infection was observed in 0.4% (14/3725) of placentas examined by microscopy and in 3.7% (19/508) by PCR. P. vivax in newborn blood was detected in 0.02% (1/4302) of samples examined by microscopy [in cord blood; 0.05% (2/4040) by microscopy, and 2.6% (13/497) by PCR]. Clinical P. vivax infection was associated with increased risk of maternal anemia (Odds Ratio-OR, 5.48, [95% CI 1.83-16.41]; p = 0.009), while submicroscopic vivax infection was not associated with increased risk of moderate-severe anemia (Hb<8g/dL) (OR, 1.16, [95% CI 0.52-2.59]; p = 0.717), or low birth weight (<2500g) (OR, 0.52, [95% CI, 0.23-1.16]; p = 0.110). CONCLUSIONS: In this multicenter study, the prevalence of P. vivax infection in pregnancy by microscopy was overall low across all endemic study sites; however, molecular methods revealed a significant number of submicroscopic infections. Clinical vivax infection in pregnancy was associated with maternal anemia, which may be deleterious for infant's health. These results may help to guide maternal health programs in settings where vivax malaria is endemic; they also highlight the need of addressing a vulnerable population such as pregnant women while embracing malaria elimination in endemic countries.


Assuntos
Malária Vivax/complicações , Plasmodium vivax , Complicações Parasitárias na Gravidez/patologia , Adolescente , Adulto , Brasil/epidemiologia , Colômbia/epidemiologia , Feminino , Sangue Fetal , Guatemala/epidemiologia , Humanos , Índia/epidemiologia , Recém-Nascido , Transmissão Vertical de Doenças Infecciosas , Malária Vivax/epidemiologia , Papua Nova Guiné/epidemiologia , Gravidez , Complicações Parasitárias na Gravidez/epidemiologia , Adulto Jovem
8.
BMC Med ; 15(1): 114, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28615061

RESUMO

BACKGROUND: γδ T cells are important for both protective immunity and immunopathogenesis during malaria infection. However, the immunological processes determining beneficial or detrimental effects on disease outcome remain elusive. The aim of this study was to examine expression and regulatory effect of the inhibitory receptor T-cell immunoglobulin domain and mucin domain 3 (TIM3) on γδ T cells. While TIM3 expression and function on conventional αß T cells have been clearly defined, the equivalent characterization on γδ T cells and associations with disease outcomes is limited. This study investigated the functional capacity of TIM3+ γδ T cells and the underlying mechanisms contributing to TIM3 upregulation and established an association with malaria disease outcomes. METHODS: We analyzed TIM3 expression on γδ T cells in 132 children aged 5-10 years living in malaria endemic areas of Papua New Guinea. TIM3 upregulation and effector functions of TIM3+ γδ T cells were assessed following in vitro stimulation with parasite-infected erythrocytes, phosphoantigen and/or cytokines. Associations between the proportion of TIM3-expressing cells and the molecular force of infection were tested using negative binomial regression and in a Cox proportional hazards model for time to first clinical episode. Multivariable analyses to determine the association of TIM3 and IL-18 levels were conducted using general linear models. Malaria infection mouse models were utilized to experimentally investigate the relationship between repeated exposure and TIM3 upregulation. RESULTS: This study demonstrates that even in the absence of an active malaria infection, children of malaria endemic areas have an atypical population of TIM3-expressing γδ T cells (mean frequency TIM3+ of total γδ T cells 15.2% ± 12). Crucial factors required for γδ T cell TIM3 upregulation include IL-12/IL-18, and plasma IL-18 was associated with TIM3 expression (P = 0.002). Additionally, we show a relationship between TIM3 expression and infection with distinct parasite clones during repeated exposure. TIM3+ γδ T cells were functionally impaired and were associated with asymptomatic malaria infection (hazard ratio 0.54, P = 0.032). CONCLUSIONS: Collectively our data demonstrate a novel role for IL-12/IL-18 in shaping the innate immune response and provide fundamental insight into aspects of γδ T cell immunoregulation. Furthermore, we show that TIM3 represents an important γδ T cell regulatory component involved in minimizing malaria symptoms.


Assuntos
Receptor Celular 2 do Vírus da Hepatite A/fisiologia , Interleucina-12/fisiologia , Interleucina-18/fisiologia , Malária/imunologia , Linfócitos T/imunologia , Animais , Criança , Pré-Escolar , Citocinas , Eritrócitos , Humanos , Interleucina-12/sangue , Interleucina-18/sangue , Camundongos , Papua Nova Guiné , Receptores de Antígenos de Linfócitos T gama-delta , Risco
9.
PLoS One ; 11(3): e0152447, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27011010

RESUMO

Plasmodium vivax is the most widely distributed human parasite and the main cause of human malaria outside the African continent. However, the knowledge about the genetic variability of P. vivax is limited when compared to the information available for P. falciparum. We present the results of a study aimed at characterizing the genetic structure of P. vivax populations obtained from pregnant women from different malaria endemic settings. Between June 2008 and October 2011 nearly 2000 pregnant women were recruited during routine antenatal care at each site and followed up until delivery. A capillary blood sample from the study participants was collected for genotyping at different time points. Seven P. vivax microsatellite markers were used for genotypic characterization on a total of 229 P. vivax isolates obtained from Brazil, Colombia, India and Papua New Guinea. In each population, the number of alleles per locus, the expected heterozygosity and the levels of multilocus linkage disequilibrium were assessed. The extent of genetic differentiation among populations was also estimated. Six microsatellite loci on 137 P. falciparum isolates from three countries were screened for comparison. The mean value of expected heterozygosity per country ranged from 0.839 to 0.874 for P. vivax and from 0.578 to 0.758 for P. falciparum. P. vivax populations were more diverse than those of P. falciparum. In some of the studied countries, the diversity of P. vivax population was very high compared to the respective level of endemicity. The level of inter-population differentiation was moderate to high in all P. vivax and P. falciparum populations studied.


Assuntos
Genótipo , Malária/parasitologia , Repetições de Microssatélites , Plasmodium vivax/genética , Alelos , Brasil , Colômbia , Feminino , Marcadores Genéticos , Variação Genética , Técnicas de Genotipagem , Geografia , Haplótipos , Heterozigoto , Humanos , Índia , Desequilíbrio de Ligação , Papua Nova Guiné , Plasmodium falciparum/genética , Reação em Cadeia da Polimerase , Gravidez , Complicações Infecciosas na Gravidez/parasitologia
10.
PLoS Med ; 12(10): e1001891, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26505753

RESUMO

BACKGROUND: The undetectable hypnozoite reservoir for relapsing Plasmodium vivax and P. ovale malarias presents a major challenge for malaria control and elimination in endemic countries. This study aims to directly determine the contribution of relapses to the burden of P. vivax and P. ovale infection, illness, and transmission in Papua New Guinean children. METHODS AND FINDINGS: From 17 August 2009 to 20 May 2010, 524 children aged 5-10 y from East Sepik Province in Papua New Guinea (PNG) participated in a randomised double-blind placebo-controlled trial of blood- plus liver-stage drugs (chloroquine [CQ], 3 d; artemether-lumefantrine [AL], 3 d; and primaquine [PQ], 20 d, 10 mg/kg total dose) (261 children) or blood-stage drugs only (CQ, 3 d; AL, 3 d; and placebo [PL], 20 d) (263 children). Participants, study staff, and investigators were blinded to the treatment allocation. Twenty children were excluded during the treatment phase (PQ arm: 14, PL arm: 6), and 504 were followed actively for 9 mo. During the follow-up time, 18 children (PQ arm: 7, PL arm: 11) were lost to follow-up. Main primary and secondary outcome measures were time to first P. vivax infection (by qPCR), time to first clinical episode, force of infection, gametocyte positivity, and time to first P. ovale infection (by PCR). A basic stochastic transmission model was developed to estimate the potential effect of mass drug administration (MDA) for the prevention of recurrent P. vivax infections. Targeting hypnozoites through PQ treatment reduced the risk of having at least one qPCR-detectable P. vivax or P. ovale infection during 8 mo of follow-up (P. vivax: PQ arm 0.63/y versus PL arm 2.62/y, HR = 0.18 [95% CI 0.14, 0.25], p < 0.001; P. ovale: 0.06 versus 0.14, HR = 0.31 [95% CI 0.13, 0.77], p = 0.011) and the risk of having at least one clinical P. vivax episode (HR = 0.25 [95% CI 0.11, 0.61], p = 0.002). PQ also reduced the molecular force of P. vivax blood-stage infection in the first 3 mo of follow-up (PQ arm 1.90/y versus PL arm 7.75/y, incidence rate ratio [IRR] = 0.21 [95% CI 0.15, 0.28], p < 0.001). Children who received PQ were less likely to carry P. vivax gametocytes (IRR = 0.27 [95% CI 0.19, 0.38], p < 0.001). PQ had a comparable effect irrespective of the presence of P. vivax blood-stage infection at the time of treatment (p = 0.14). Modelling revealed that mass screening and treatment with highly sensitive quantitative real-time PCR, or MDA with blood-stage treatment alone, would have only a transient effect on P. vivax transmission levels, while MDA that includes liver-stage treatment is predicted to be a highly effective strategy for P. vivax elimination. The inclusion of a directly observed 20-d treatment regime maximises the efficiency of hypnozoite clearance but limits the generalisability of results to real-world MDA programmes. CONCLUSIONS: These results suggest that relapses cause approximately four of every five P. vivax infections and at least three of every five P. ovale infections in PNG children and are important in sustaining transmission. MDA campaigns combining blood- and liver-stage treatment are predicted to be a highly efficacious intervention for reducing P. vivax and P. ovale transmission. TRIAL REGISTRATION: ClinicalTrials.gov NCT02143934.


Assuntos
Antimaláricos/uso terapêutico , Malária/tratamento farmacológico , Malária/transmissão , Modelos Estatísticos , Plasmodium ovale/efeitos dos fármacos , Plasmodium vivax/efeitos dos fármacos , Esporozoítos/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Criança , Pré-Escolar , Erradicação de Doenças/tendências , Método Duplo-Cego , Feminino , Humanos , Masculino , Papua Nova Guiné/epidemiologia , Placebos , Reação em Cadeia da Polimerase em Tempo Real , Recidiva , Resultado do Tratamento
11.
PLoS One ; 10(5): e0126747, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25996916

RESUMO

A better understanding of human-to-mosquito transmission is crucial to control malaria. In order to assess factors associated with gametocyte carriage, 2083 samples were collected in a cross-sectional survey in Papua New Guinea. Plasmodium species were detected by light microscopy and qPCR and gametocytes by detection of pfs25 and pvs25 mRNA transcripts by reverse-transcriptase PCR (qRT-PCR). The parasite prevalence by PCR was 18.5% for Plasmodium falciparum and 13.0% for P. vivax. 52.5% of all infections were submicroscopic. Gametocytes were detected in 60% of P. falciparum-positive and 51% of P. vivax-positive samples. Each 10-fold increase in parasite density led to a 1.8-fold and 3.3-fold increase in the odds of carrying P. falciparum and P. vivax gametocytes. Thus the proportion of gametocyte positive and gametocyte densities was highest in young children carrying high asexual parasite densities and in symptomatic individuals. Dilution series of gametocytes allowed absolute quantification of gametocyte densities by qRT-PCR and showed that pvs25 expression is 10-20 fold lower than pfs25 expression. Between 2006 and 2010 parasite prevalence in the study site has decreased by half. 90% of the remaining infections were asymptomatic and likely constitute an important reservoir of transmission. However, mean gametocyte densities were low (approx. 1-2 gametocyte/µL) and it remains to be determined to what extent low-density gametocyte positive individuals are infective to mosquitos.


Assuntos
Malária Falciparum/epidemiologia , Malária Falciparum/parasitologia , Malária Vivax/epidemiologia , Malária Vivax/parasitologia , Parasitemia , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium vivax/crescimento & desenvolvimento , Adolescente , Adulto , Criança , Pré-Escolar , Eritrócitos/parasitologia , Feminino , Genes de Protozoários , Humanos , Lactente , Recém-Nascido , Estágios do Ciclo de Vida , Masculino , Papua Nova Guiné/epidemiologia , Carga Parasitária , Plasmodium falciparum/genética , Plasmodium vivax/genética , Prevalência , Adulto Jovem
12.
Malar J ; 14: 37, 2015 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-25626445

RESUMO

BACKGROUND: In northern Papua New Guinea (PNG), most Plasmodium falciparum isolates proved resistant to chloroquine (CQ) in vitro between 2005 and 2007, and there was near-fixation of pfcrt K76T, pfdhfr C59R/S108N and pfmdr1 N86Y. To determine whether the subsequent introduction of artemisinin combination therapy (ACT) and reduced CQ-sulphadoxine-pyrimethamine pressure had attenuated parasite drug susceptibility and resistance-associated mutations, these parameters were re-assessed between 2011 and 2013. METHODS: A validated fluorescence-based assay was used to assess growth inhibition of 52 P. falciparum isolates from children in a clinical trial in Madang Province. Responses to CQ, lumefantrine, piperaquine, naphthoquine, pyronaridine, artesunate, dihydroartemisinin, artemether were assessed. Molecular resistance markers were detected using a multiplex PCR ligase detection reaction fluorescent microsphere assay. RESULTS: CQ resistance (in vitro concentration required for 50% parasite growth inhibition (IC50) >100 nM) was present in 19% of isolates. All piperaquine and naphthoquine IC50s were <100 nM and those for lumefantrine, pyronaridine and the artemisinin derivatives were in low nM ranges. Factor analysis of IC50s showed three groupings (lumefantrine; CQ, piperaquine, naphthoquine; pyronaridine, dihydroartemisinin, artemether, artesunate). Most isolates (96%) were monoclonal pfcrt K76T (SVMNT) mutants and most (86%) contained pfmdr1 N86Y (YYSND). No wild-type pfdhfr was found but most isolates contained wild-type (SAKAA) pfdhps. Compared with 2005-2007, the geometric mean (95% CI) CQ IC50 was lower (87 (71-107) vs 167 (141-197) nM) and there had been no change in the prevalence of pfcrt K76T or pfmdr1 mutations. There were fewer isolates of the pfdhps (SAKAA) wild-type (60 vs 100%) and pfdhfr mutations persisted. CONCLUSIONS: Reflecting less drug pressure, in vitro CQ sensitivity appears to be improving in Madang Province despite continued near-fixation of pfcrt K76T and pfmdr1 mutations. Temporal changes in IC50s for other anti-malarial drugs were inconsistent but susceptibility was preserved. Retention or increases in pfdhfr and pfdhps mutations reflect continued use of sulphadoxine-pyrimethamine in the study area including through paediatric intermittent preventive treatment. The susceptibility of local isolates to lumefantrine may be unrelated to those of other ACT partner drugs. TRIAL REGISTRATION: Australian New Zealand Clinical Trials Registry ACTRN12610000913077 .


Assuntos
Antimaláricos/farmacologia , Resistência a Medicamentos/genética , Malária Falciparum/parasitologia , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/genética , Pré-Escolar , Humanos , Lactente , Malária Falciparum/epidemiologia , Mutação/genética , Papua Nova Guiné/epidemiologia
13.
BMC Med ; 13: 9, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25591391

RESUMO

BACKGROUND: Intermittent preventive treatment in pregnancy has not been evaluated outside of Africa. Low birthweight (LBW, <2,500 g) is common in Papua New Guinea (PNG) and contributing factors include malaria and reproductive tract infections. METHODS: From November 2009 to February 2013, we conducted a parallel group, randomised controlled trial in pregnant women (≤ 26 gestational weeks) in PNG. Sulphadoxine-pyrimethamine (1,500/75 mg) plus azithromycin (1 g twice daily for 2 days) (SPAZ) monthly from second trimester (intervention) was compared against sulphadoxine-pyrimethamine and chloroquine (450 to 600 mg, daily for three days) (SPCQ) given once, followed by SPCQ placebo (control). Women were assigned to treatment (1:1) using a randomisation sequence with block sizes of 32. Participants were blinded to assignments. The primary outcome was LBW. Analysis was by intention-to-treat. RESULTS: Of 2,793 women randomised, 2,021 (72.4%) were included in the primary outcome analysis (SPCQ: 1,008; SPAZ: 1,013). The prevalence of LBW was 15.1% (305/2,021). SPAZ reduced LBW (risk ratio [RR]: 0.74, 95% CI: 0.60-0.91, P = 0.005; absolute risk reduction (ARR): 4.5%, 95% CI: 1.4-7.6; number needed to treat: 22), and preterm delivery (0.62, 95% CI: 0.43-0.89, P = 0.010), and increased mean birthweight (41.9 g, 95% CI: 0.2-83.6, P = 0.049). SPAZ reduced maternal parasitaemia (RR: 0.57, 95% CI: 0.35-0.95, P = 0.029) and active placental malaria (0.68, 95% CI: 0.47-0.98, P = 0.037), and reduced carriage of gonorrhoea (0.66, 95% CI: 0.44-0.99, P = 0.041) at second visit. There were no treatment-related serious adverse events (SAEs), and the number of SAEs (intervention 13.1% [181/1,378], control 12.7% [174/1,374], P = 0.712) and AEs (intervention 10.5% [144/1,378], control 10.8% [149/1,374], P = 0.737) was similar. A major limitation of the study was the high loss to follow-up for birthweight. CONCLUSIONS: SPAZ was efficacious and safe in reducing LBW, possibly acting through multiple mechanisms including the effect on malaria and on sexually transmitted infections. The efficacy of SPAZ in the presence of resistant parasites and the contribution of AZ to bacterial antibiotic resistance require further study. The ability of SPAZ to improve pregnancy outcomes warrants further evaluation. TRIAL REGISTRATION: ClinicalTrials.gov NCT01136850 (06 April 2010).


Assuntos
Antimaláricos/administração & dosagem , Azitromicina/administração & dosagem , Recém-Nascido de Baixo Peso , Malária/prevenção & controle , Complicações Parasitárias na Gravidez/prevenção & controle , Pirimetamina/administração & dosagem , Sulfadoxina/administração & dosagem , Adulto , Cloroquina/administração & dosagem , Combinação de Medicamentos , Feminino , Humanos , Recém-Nascido , Malária/complicações , Papua Nova Guiné , Gravidez , Método Simples-Cego , Adulto Jovem
14.
PLoS Med ; 11(12): e1001773, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25549086

RESUMO

BACKGROUND: Artemisinin combination therapies (ACTs) with broad efficacy are needed where multiple Plasmodium species are transmitted, especially in children, who bear the brunt of infection in endemic areas. In Papua New Guinea (PNG), artemether-lumefantrine is the first-line treatment for uncomplicated malaria, but it has limited efficacy against P. vivax. Artemisinin-naphthoquine should have greater activity in vivax malaria because the elimination of naphthoquine is slower than that of lumefantrine. In this study, the efficacy, tolerability, and safety of these ACTs were assessed in PNG children aged 0.5-5 y. METHODS AND FINDINGS: An open-label, randomized, parallel-group trial of artemether-lumefantrine (six doses over 3 d) and artemisinin-naphthoquine (three daily doses) was conducted between 28 March 2011 and 22 April 2013. Parasitologic outcomes were assessed without knowledge of treatment allocation. Primary endpoints were the 42-d P. falciparum PCR-corrected adequate clinical and parasitologic response (ACPR) and the P. vivax PCR-uncorrected 42-d ACPR. Non-inferiority and superiority designs were used for falciparum and vivax malaria, respectively. Because the artemisinin-naphthoquine regimen involved three doses rather than the manufacturer-specified single dose, the first 188 children underwent detailed safety monitoring. Of 2,542 febrile children screened, 267 were randomized, and 186 with falciparum and 47 with vivax malaria completed the 42-d follow-up. Both ACTs were safe and well tolerated. P. falciparum ACPRs were 97.8% and 100.0% in artemether-lumefantrine and artemisinin-naphthoquine-treated patients, respectively (difference 2.2% [95% CI -3.0% to 8.4%] versus -5.0% non-inferiority margin, p = 0.24), and P. vivax ACPRs were 30.0% and 100.0%, respectively (difference 70.0% [95% CI 40.9%-87.2%], p<0.001). Limitations included the exclusion of 11% of randomized patients with sub-threshold parasitemias on confirmatory microscopy and direct observation of only morning artemether-lumefantrine dosing. CONCLUSIONS: Artemisinin-naphthoquine is non-inferior to artemether-lumefantrine in PNG children with falciparum malaria but has greater efficacy against vivax malaria, findings with implications in similar geo-epidemiologic settings within and beyond Oceania. TRIAL REGISTRATION: Australian New Zealand Clinical Trials Registry ACTRN12610000913077. Please see later in the article for the Editors' Summary.


Assuntos
Antimaláricos/uso terapêutico , Artemisininas/uso terapêutico , Etanolaminas/uso terapêutico , Fluorenos/uso terapêutico , Malária Falciparum/tratamento farmacológico , Adulto , Artemeter , Feminino , Humanos , Lumefantrina , Masculino , Pessoa de Meia-Idade
15.
BMC Infect Dis ; 14: 630, 2014 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-25423900

RESUMO

BACKGROUND: Viral central nervous system (CNS) infections are common in countries where malaria is endemic but, due to limited laboratory facilities, few studies have systematically examined the prevalence and clinical consequences of the presence of viruses in cerebrospinal fluid (CSF) from children with suspected CNS infection. METHODS: We performed a prospective study of Papua New Guinean children hospitalized with signs and symptoms of CNS infection. CSF samples from 300 children without proven bacterial/fungal meningitis were analyzed for human herpes viruses (HHV), picornaviruses, influenza, adenoviruses, flaviviruses and bacteria. RESULTS: Fifty-five children (18%) had viral (42), bacterial (20) or both viral and bacterial (7) nucleic acids (NA) identified in their CSF. Human herpes viruses accounted for 91% of all viruses found. The identification of viral or bacterial NA was not associated with any characteristic clinical features. By contrast, malaria was associated with increased identification of viral and bacterial NA and with impaired consciousness, multiple convulsions and age. Malaria was also inversely associated with an adverse outcome. Amongst children with HHV infection, those with HHV-6 and -7 were younger, were more likely have impaired consciousness and had a higher proportion of adverse outcomes than children with CMV. Dengue and enteroviral infections were infrequent. Adenoviral and influenza infections were not identified. CONCLUSION: Infections with HHV-6, HHV-7, dengue and enterovirus have the potential to cause serious CNS disease in young PNG children. However most HHVs in this malaria-endemic setting should be considered to be the result of reactivation from a latent reservoir without clinical sequelae.


Assuntos
Viroses do Sistema Nervoso Central/epidemiologia , Viroses do Sistema Nervoso Central/líquido cefalorraquidiano , Viroses do Sistema Nervoso Central/virologia , Criança , Criança Hospitalizada/estatística & dados numéricos , Pré-Escolar , Enterovirus/isolamento & purificação , Feminino , Herpesvirus Humano 6/isolamento & purificação , Humanos , Lactente , Malária/complicações , Malária/epidemiologia , Masculino , Papua Nova Guiné/epidemiologia , Picornaviridae/isolamento & purificação , Prevalência , Estudos Prospectivos , Estudos Retrospectivos
16.
Malar J ; 13: 319, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-25123055

RESUMO

BACKGROUND: Gametocytes are the transmission stages of Plasmodium parasites, the causative agents of malaria. As their density in the human host is typically low, they are often undetected by conventional light microscopy. Furthermore, application of RNA-based molecular detection methods for gametocyte detection remains challenging in remote field settings. In the present study, a detailed comparison of three methods, namely light microscopy, magnetic fractionation and reverse transcriptase polymerase chain reaction for detection of Plasmodium falciparum and Plasmodium vivax gametocytes was conducted. METHODS: Peripheral blood samples from 70 children aged 0.5 to five years with uncomplicated malaria who were treated with either artemether-lumefantrine or artemisinin-naphthoquine were collected from two health facilities on the north coast of Papua New Guinea. The samples were taken prior to treatment (day 0) and at pre-specified intervals during follow-up. Gametocytes were measured in each sample by three methods: i) light microscopy (LM), ii) quantitative magnetic fractionation (MF) and, iii) reverse transcriptase PCR (RTPCR). Data were analysed using censored linear regression and Bland and Altman techniques. RESULTS: MF and RTPCR were similarly sensitive and specific, and both were superior to LM. Overall, there were approximately 20% gametocyte-positive samples by LM, whereas gametocyte positivity by MF and RTPCR were both more than two-fold this level. In the subset of samples collected prior to treatment, 29% of children were positive by LM, and 85% were gametocyte positive by MF and RTPCR, respectively. CONCLUSIONS: The present study represents the first direct comparison of standard LM, MF and RTPCR for gametocyte detection in field isolates. It provides strong evidence that MF is superior to LM and can be used to detect gametocytaemic patients under field conditions with similar sensitivity and specificity as RTPCR.


Assuntos
Separação Imunomagnética/métodos , Malária/parasitologia , Microscopia/métodos , Parasitologia/métodos , Plasmodium falciparum/isolamento & purificação , Plasmodium vivax/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Antimaláricos/uso terapêutico , Combinação Arteméter e Lumefantrina , Artemisininas/uso terapêutico , Pré-Escolar , Técnicas de Laboratório Clínico/métodos , Combinação de Medicamentos , Etanolaminas/uso terapêutico , Feminino , Fluorenos/uso terapêutico , Humanos , Lactente , Malária/tratamento farmacológico , Masculino , Naftoquinonas/uso terapêutico , Papua Nova Guiné , Sensibilidade e Especificidade
17.
PLoS One ; 9(5): e96810, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24828338

RESUMO

BACKGROUND: Poor-quality life-saving medicines are a major public health threat, particularly in settings with a weak regulatory environment. Insufficient amounts of active pharmaceutical ingredients (API) endanger patient safety and may contribute to the development of drug resistance. In the case of malaria, concerns relate to implications for the efficacy of artemisinin-based combination therapies (ACT). In Papua New Guinea (PNG), Plasmodium falciparum and P. vivax are both endemic and health facilities are the main source of treatment. ACT has been introduced as first-line treatment but other drugs, such as primaquine for the treatment of P. vivax hypnozoites, are widely available. This study investigated the quality of antimalarial drugs and selected antibiotics at all levels of the health facility supply chain in PNG. METHODS AND FINDINGS: Medicines were obtained from randomly sampled health facilities and selected warehouses and hospitals across PNG and analysed for API content using validated high performance liquid chromatography (HPLC). Of 360 tablet/capsule samples from 60 providers, 9.7% (95% CI 6.9, 13.3) contained less, and 0.6% more, API than pharmacopoeial reference ranges, including 29/37 (78.4%) primaquine, 3/70 (4.3%) amodiaquine, and one sample each of quinine, artemether, sulphadoxine-pyrimethamine and amoxicillin. According to the package label, 86.5% of poor-quality samples originated from India. Poor-quality medicines were found in 48.3% of providers at all levels of the supply chain. Drug quality was unrelated to storage conditions. CONCLUSIONS: This study documents the presence of poor-quality medicines, particularly primaquine, throughout PNG. Primaquine is the only available transmission-blocking antimalarial, likely to become important to prevent the spread of artemisinin-resistant P. falciparum and eliminating P. vivax hypnozoites. The availability of poor-quality medicines reflects the lack of adequate quality control and regulatory mechanisms. Measures to stop the availability of poor-quality medicines should include limiting procurement to WHO prequalified products and implementing routine quality testing.


Assuntos
Antimaláricos/análise , Medicamentos Biossimilares/análise , Malária Falciparum/tratamento farmacológico , Malária Vivax/tratamento farmacológico , Amodiaquina/análise , Amodiaquina/normas , Amodiaquina/uso terapêutico , Amoxicilina/análise , Amoxicilina/normas , Amoxicilina/uso terapêutico , Antimaláricos/normas , Antimaláricos/uso terapêutico , Artemeter , Artemisininas/análise , Artemisininas/normas , Artemisininas/uso terapêutico , Medicamentos Biossimilares/normas , Medicamentos Biossimilares/uso terapêutico , Cloroquina/análise , Cloroquina/normas , Cloroquina/uso terapêutico , Resistência a Medicamentos , Instalações de Saúde , Humanos , Papua Nova Guiné , Primaquina/análise , Primaquina/normas , Primaquina/uso terapêutico , Controle de Qualidade , Quinina/análise , Quinina/normas , Quinina/uso terapêutico
18.
Malar J ; 13: 145, 2014 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-24739250

RESUMO

BACKGROUND: The accuracy of the World Health Organization method of estimating malaria parasite density from thick blood smears by assuming a white blood cell (WBC) count of 8,000/µL has been questioned in several studies. Since epidemiological investigations, anti-malarial efficacy trials and routine laboratory reporting in Papua New Guinea (PNG) have all relied on this approach, its validity was assessed as part of a trial of artemisinin-based combination therapy, which included blood smear microscopy and automated measurement of leucocyte densities on Days 0, 3 and 7. RESULTS: 168 children with uncomplicated malaria (median (inter-quartile range) age 44 (39-47) months) were enrolled, 80.3% with Plasmodium falciparum monoinfection, 14.9% with Plasmodium vivax monoinfection, and 4.8% with mixed P. falciparum/P. vivax infection. All responded to allocated therapy and none had a malaria-positive slide on Day 3. Consistent with a median baseline WBC density of 7.3 (6.5-7.8) × 10(9)/L, there was no significant difference in baseline parasite density between the two methods regardless of Plasmodium species. Bland Altman plots showed that, for both species, the mean difference between paired parasite densities calculated from assumed and measured WBC densities was close to zero. At parasite densities <10,000/µL by measured WBC, almost all between-method differences were within the 95% limits of agreement. Above this range, there was increasing scatter but no systematic bias. CONCLUSIONS: Diagnostic thresholds and parasite clearance assessment in most PNG children with uncomplicated malaria are relatively robust, but accurate estimates of a higher parasitaemia, as a prognostic index, requires formal WBC measurement.


Assuntos
Contagem de Leucócitos/métodos , Malária Falciparum/parasitologia , Malária Vivax/parasitologia , Microscopia/métodos , Carga Parasitária/métodos , Antimaláricos/uso terapêutico , Artemisininas/uso terapêutico , Pré-Escolar , Combinação de Medicamentos , Feminino , Humanos , Masculino , Papua Nova Guiné , Plasmodium falciparum/fisiologia , Plasmodium vivax/fisiologia
19.
Antimicrob Agents Chemother ; 58(1): 432-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24189254

RESUMO

Since conventional 14-day primaquine (PMQ) radical cure of vivax malaria is associated with poor compliance, and as total dose, not therapy duration, determines efficacy, a preliminary pharmacokinetic study of two doses (0.5 and 1.0 mg/kg of body weight) was conducted in 28 healthy glucose-6-phosphate dehydrogenase-normal Papua New Guinean children, aged 5 to 12 years, to facilitate development of abbreviated high-dose regimens. Dosing was with food and was directly observed, and venous blood samples were drawn during a 168-h postdose period. Detailed safety monitoring was performed for hepatorenal function and hemoglobin and methemoglobin concentrations. Plasma concentrations of PMQ and its metabolite carboxyprimaquine (CPMQ) were determined by liquid chromatography-mass spectrometry and analyzed using population pharmacokinetic methods. The derived models were used in simulations. Both single-dose regimens were well tolerated with no changes in safety parameters. The mean PMQ central volume of distribution and clearance relative to bioavailability (200 liters/70 kg and 24.6 liters/h/70 kg) were within published ranges for adults. The median predicted maximal concentrations (Cmax) for both PMQ and CPMQ after the last dose of a 1.0 mg/kg 7-day PMQ regimen were approximately double those at the end of 14 days of 0.5 mg/kg daily, while a regimen of 1.0 mg/kg twice daily resulted in a 2.38 and 3.33 times higher Cmax for PMQ and CPMQ, respectively. All predicted median Cmax concentrations were within ranges for adult high-dose studies that also showed acceptable safety and tolerability. The present pharmacokinetic data, the first for PMQ in children, show that further studies of abbreviated high-dose regimens are feasible in this age group.


Assuntos
Antimaláricos/farmacocinética , Antimaláricos/uso terapêutico , Malária Vivax/sangue , Malária Vivax/tratamento farmacológico , Primaquina/farmacocinética , Primaquina/uso terapêutico , Antimaláricos/administração & dosagem , Criança , Esquema de Medicação , Feminino , Humanos , Masculino , Primaquina/administração & dosagem
20.
P N G Med J ; 57(1-4): 68-74, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-26930890

RESUMO

Plasmodium vivax is a major cause of malarial infection and disease in young children in Papua New Guinea. Recent increase in funding for malaria control has improved accessibility to preventive measures, diagnosis and artemisinin combination therapies. Yet the current treatment and control measures are more effective against P. falciparum than against P. vivax and P. ovale due to the biological differences in the liver stage life-cycle of these parasites. P. vivax and P. ovale have a dormant liver stage called a hypnozoite. The artemisinin combination therapies, while highly effective against the blood stages of all plasmodium species causing human malarial illness, have no effect upon the hypnozoites in the liver and the stage V gametocytes of P. falciparum. Currently, primaquine is the only licensed drug shown to be effective against both the hypnozoites of P. vivax and P. ovale in the liver and the stage V gametocytes of P. falciparum. Primaquine has a high associated risk of life-threatening haemolytic anaemia when administered to glucose-6-phosphate dehydrogenase (G6PD)-deficient persons. The lack of cheap, reliable point-of-care testing for the diagnosis of G6PD deficiency remains a major obstacle to the widespread use of primaquine in clinical and public health practice. Furthermore, there is a paucity of primaquine safety and tolerability data, especially in young children with the highest P. vivax disease burden. For malaria control and elimination efforts to be effective, interventions such as mass drug administration must include primaquine. This opinion paper highlights the need to eradicate hypnozoites in the liver of the human host with primaquine treatment for radical cure of malarial illness and discusses the challenges in the use of primaquine as a public health tool for malaria control and elimination programs in countries such as Papua New Guinea.


Assuntos
Antimaláricos/uso terapêutico , Malária Vivax/tratamento farmacológico , Plasmodium vivax , Primaquina/uso terapêutico , Humanos , Papua Nova Guiné
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA