15 research outputs found

    Comparison of an assumed versus measured leucocyte count in parasite density calculations in Papua New Guinean children with uncomplicated malaria

    Get PDF
    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. © 2014 Laman et al.; licensee BioMed Central Ltd

    Strategies for Understanding and Reducing the Plasmodium vivax and Plasmodium ovale Hypnozoite Reservoir in Papua New Guinean Children: A Randomised Placebo-Controlled Trial and Mathematical Model

    Get PDF
    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.; 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.; 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.; ClinicalTrials.gov NCT02143934

    Mass drug administration trial to eliminate lymphatic filariasis in Papua New Guinea: changes in microfilaremia, filarial antigen, and Bm14 antibody after cessation

    No full text
    Laboratory tools to monitor infection burden are important to evaluate progress and determine endpoints in programs to eliminate lymphatic filariasis. We evaluated changes in Wuchereria bancrofti microfilaria, filarial antigen and Bm14 antibody in individuals who participated in a five-year mass drug administration trial in Papua New Guinea. Comparing values before treatment and one year after four annual treatments, the proportion of microfilaria positive individuals declined to the greatest degree, with less marked change in antibody and antigen rates. Considering children as sentinel groups who reflect recent transmission intensity, children surveyed before the trial were more frequently microfilaria and antibody positive than those examined one year after the trial stopped. In contrast, antigen positive rates were similar in the two groups. All infection indicators continued to decline five years after cessation of mass drug administration; Bm14 antibody persisted in the greatest proportion of individuals. These data suggest that Bm14 antibody may be a sensitive test to monitor continuing transmission during and after mass drug administration aimed at eliminating transmission of lymphatic filariasis

    Data from: Strategies for understanding and reducing the Plasmodium vivax and Plasmodium ovale hypnozoite reservoir in Papua New Guinean children: a randomised placebo-controlled trial and mathematical model

    No full text
    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

    Strategies for Understanding and Reducing the Plasmodium vivax and Plasmodium ovale Hypnozoite Reservoir in Papua New Guinean Children: A Randomised Placebo-Controlled Trial and Mathematical Model

    No full text
    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

    Data repository analyses presented in Robinson et al. 2015 PLoS Medince

    No full text
    1) Albinama_Table1.dta: Baseline characteristics of cohort; 2) Albinama_Table2_Po: Survival analyses of risk of P. ovale infections; 3) Albinama_Table2_Pv_clin: Survival analyses of incidence risk of P. vivax clinical episodes; 4) Albinama_Table2_Pv_LM: survival analyses of incidence risk of light-microscopically positive P. vivax infections; 5) Albinama_Table2_Pv_PCR: survival analyses of incidence risk of PCR positive P. vivax infections; 6)Albinama_Table2_Pvg: survival analyses of incidence risk of gametocyte positive P. vivax infections; 7) Albinama_Table3_0-3mo: incidence of malaria endpoints for first 3 months of follow-up; 8) Albinama_Table3_4-8mo: incidence of malaria endpoints for months 4-8 of follow-up; 9) Albinama_Table3_0-8mo: incidence of malaria endpoints for entire 8 months follow-up; 10) Albinama_Table4_Pf_clin: Survival analyses of incidence risk of P. falciparum clinical episodes; 11) Albinama_Table4_Pf_LM: survival analyses of incidence risk of light-microscopically positive P. falciparum infections; 12) Albinama_Table4_Pf_PCR: survival analyses of incidence risk of PCR positive P. falciparum infections; 13)Albinama_Table4_Pm_PCR: survival analyses of incidence risk of PCR positive P. malariae infections

    Mathematical-model-based predictions of impact of MDA and MSAT with either blood-stage drugs only or blood- plus liver-stage drugs on the population prevalence of <i>P</i>. <i>vivax</i> and <i>P</i>. <i>falciparum</i> infections.

    No full text
    <p>The effect of two rounds (6 mo apart) of MDA (A and B) or MSAT (C and D) with anti-malarial drugs at 80% coverage on <i>P</i>. <i>vivax</i> (A and C) and <i>P</i>. <i>falciparum</i> (B and D) blood-stage parasite prevalence, as predicted by a stochastic model in a human population of size 5,000. The lines represent the mean of 1,000 repeat simulations, and the shaded areas represent the envelopes containing 95% of stochastic simulations. The grey and green shaded bars denote the duration of prophylactic protection for DHA-PIP/CQ and tafenoquine, respectively, after each treatment round. DHA-PIP and CQ were assumed to be administered as part of a 3-d regimen, providing prophylaxis for 1 mo. PQ was assumed to be administered as part of a 14-d regimen, providing prophylaxis for 15 d. Tafenoquine was assumed to be administered via a single dose, providing prophylaxis for 2 mo.</p

    Time to first <i>Plasmodium</i> spp. infection and <i>P</i>. <i>vivax</i> clinical episode.

    No full text
    <p>Kaplan-Meier plots showing the time to first (or only) (A) <i>P</i>. <i>vivax</i> infection by qPCR, (B) <i>P</i>. <i>vivax</i> infection by LM, (C) <i>P</i>. <i>vivax</i> clinical episode, (D) <i>P</i>. <i>falciparum</i> infection by qPCR, (E) <i>P</i>. <i>malariae</i> infection by qPCR, and (F) <i>P</i>. <i>ovale</i> infection by qPCR, in the two treatment arms. Dashed lines represent the respective 95% confidence intervals.</p
    corecore